Next Article in Journal
Insights into the Composition and Antibacterial Activity of Amomum tsao-ko Essential Oils from Different Regions Based on GC-MS and GC-IMS
Next Article in Special Issue
The Impact of Dietary Fiber as a Prebiotic on Inflammation in Children with Obesity
Previous Article in Journal
Effects of Defatting Methods on the Physicochemical Properties of Proteins Extracted from Hermetia illucens Larvae
Previous Article in Special Issue
The Concept of Postbiotics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gastrointestinal Microbiota and Their Manipulation for Improved Growth and Performance in Chickens

by
Shahna Fathima
1,
Revathi Shanmugasundaram
2,*,
Daniel Adams
1 and
Ramesh K. Selvaraj
1
1
Department of Poultry Science, The University of Georgia, Athens, GA 30605, USA
2
Toxicology and Mycotoxin Research Unit, US National Poultry Research Center, Athens, GA 30605, USA
*
Author to whom correspondence should be addressed.
Submission received: 26 March 2022 / Revised: 1 May 2022 / Accepted: 9 May 2022 / Published: 12 May 2022
(This article belongs to the Special Issue Postbiotics: Emerging Applications in Food Field)

Abstract

:
The gut of warm-blooded animals is colonized by microbes possibly constituting at least 100 times more genetic material of microbial cells than that of the somatic cells of the host. These microbes have a profound effect on several physiological functions ranging from energy metabolism to the immune response of the host, particularly those associated with the gut immune system. The gut of a newly hatched chick is typically sterile but is rapidly colonized by microbes in the environment, undergoing cycles of development. Several factors such as diet, region of the gastrointestinal tract, housing, environment, and genetics can influence the microbial composition of an individual bird and can confer a distinctive microbiome signature to the individual bird. The microbial composition can be modified by the supplementation of probiotics, prebiotics, or synbiotics. Supplementing these additives can prevent dysbiosis caused by stress factors such as infection, heat stress, and toxins that cause dysbiosis. The mechanism of action and beneficial effects of probiotics vary depending on the strains used. However, it is difficult to establish a relationship between the gut microbiome and host health and productivity due to high variability between flocks due to environmental, nutritional, and host factors. This review compiles information on the gut microbiota, dysbiosis, and additives such as probiotics, postbiotics, prebiotics, and synbiotics, which are capable of modifying gut microbiota and elaborates on the interaction of these additives with chicken gut commensals, immune system, and their consequent effects on health and productivity. Factors to be considered and the unexplored potential of genetic engineering of poultry probiotics in addressing public health concerns and zoonosis associated with the poultry industry are discussed.

1. Introduction

Over the last three decades, the poultry industry has expanded tremendously with an annual growth rate of over 5% when compared to the bovine and swine industries, which grew at 1.5% and 3%, respectively [1]. Such intense growth demands effective disease prevention and control strategies. Economic loss because of loss in production associated with disease outbreaks accounts for 20% of the value of poultry production [2]. The regulation on the use of antibiotics necessitated the need to find potential alternatives to replace antibiotic growth promoters and reduce the prevalence of bacterial diseases in organic livestock production. Some of the alternatives which have gained research interest are probiotics, prebiotics, postbiotics, organic acids, and plant extracts [3]. Probiotics emerged as a potential alternative to antibiotics because probiotics are primarily of host origin and are considered Generally Recognized As Safe (GRAS) [3]. Probiotics are commensal bacteria and are typically isolated from the gastrointestinal tract of healthy chickens, screened for probiotic characteristics in vitro first, followed by in vivo characterization.
The microbes in the gastrointestinal tract play a crucial role in promoting the health and production performances of chickens. The diversity of gut microbiota varies by region, with cecum having the highest microbial diversity [4]. The gut microbiota functions to improve the utilization of nutrients, prevent pathogen colonization, improve growth performance and metabolize mycotoxins in the feed [5,6,7]. It is important to monitor the gut microbiota in chickens as some of the gut microbiota such as Salmonella and Campylobacter cause zoonotic diseases in humans [8]. The early colonization of the gut by microbes is crucial as it can affect the morphology and physiology of the intestine and susceptibility to infections. In ovo inoculation of probiotics and prebiotics favors the early colonization of the gut by ‘beneficial’ microbiota post-hatch [9,10,11].
The gut barrier is comprised of intestinal microbiota and their metabolites, mucins secreted by goblet cells, host-derived antimicrobial peptides such as defensins, and cathelicidins, IgA, intestinal epithelium, microfold cells (M cells), Paneth cells, tuft cells and lymphoid tissues in the sub-epithelium and lamina propria [12,13]. The gut barrier serves to contain the gut microbiota within the lumen while permitting the absorption of nutrients [12,14]. Intestinal health, tolerance to food and microbial antigens, and homeostasis are achieved through complex interactions between the multiple components in the gut [13]. This review will provide an in-depth analysis of the current knowledge on prebiotics, probiotics, postbiotics, and synbiotics supplementation in poultry. The effects of supplementing probiotics, postbiotics, prebiotics, and synbiotics and the mechanisms through which these additives exert beneficial effects to modulate the gut microbiota, and the interaction of the gut microbiota with the growth, performance, and immunity in chickens are included. This review also addresses the potential disadvantages of probiotic application in poultry. This review highlights some of the challenges in probiotic supplementation in poultry, identifies gaps in our knowledge, and identifies new avenues to develop the next generation of effective probiotic products.

2. Development of Gut Microbiota

Intestinal epithelial cells, microbial community, and the immune system are components of the gut ecosystem [14]. Prior to large-scale incubation of eggs in incubators, the eggs were in contact with the nest or hen during the incubation period and thereby ensuring the vertical transmission of the maternal microbiota to chicks [15]. However, in commercial hatchery settings, chicks are hatched in a clean environment with no contact with the hen. Hence, the gut microbiota of newly hatched chicks is entirely dependent on environmental sources which can lead to a decrease in microbial diversity [16] and an increase in foodborne pathogen colonization of the gut. Transferring the gut microbiota of healthy adult chickens to day-old chicks will be a future approach to controlling foodborne pathogens in poultry.
Microorganisms colonize the gut mucosal epithelium, gut lumen, and caeca of chickens [17]. The microbial composition and diversity are influenced by age of the bird, region of the gastrointestinal tract, genotype of the bird, housing conditions, and feed composition [18,19]. Conventional microbiological methods limited the research and identification of the individual components of the chicken gut microbiota. The development of 16S rRNA-based next-generation sequencing tools has made it possible to characterize the gut microbiota of chickens [20,21].
Chicks can acquire microbiota at the embryonic stage during egg formation in the oviduct and during transport through the reproductive tract [22]. Post-hatch microbial acquisition is dependent on various factors such as production practices, diet, and environment. With the modernization of chicken production in large-scale hatcheries, the natural vertical transfer of microbiota from the hen is considerably diminished. Nevertheless, the passage of eggs through the reproductive tract and cloaca will deposit microbiota on the eggshell in the form of a complex biofilm [23]. The obligates anaerobes in the eggshell biofilm can survive the incubation period and can shape the microbial population of the chick. Hatchery sanitation protocols such as washing, fumigation, and chemical disinfection of the egg, though, will reduce the vertical transfer of microbiota. In a study conducted by Olsen et al. [24], it was demonstrated that the bacterial load on eggs decreased from 1 × 104 to 1 × 105 CFU to less than 10 CFU per egg. Although disinfection practices are inevitable to ensure optimal hatchability in commercial hatcheries, disinfection impedes the vertical transfer of microbiota. Hence chicks acquire a considerable proportion of gut microbiota from the environment post-hatch.
The first species to colonize the chicken gastrointestinal tract are coliforms and fecal Streptococci, which are abundant by day 3 post-hatch [17,25]. The small intestinal microbiota is established by approximately 2 weeks of age. By day 40, Lactobacillus dominates the small intestinal microflora. Cecal microbiota is established by 6–7 weeks and is dominated by facultative and obligate anaerobic microbes, consisting of Clostridia, enterobacteria, fecal Streptococci, Pediococci, and Pseudomonas aeruginosa [17]. Microbial composition and complexity increase in the distal digestive tract leading to the fluctuation in fecal microbial composition [21]. The microbial density and the predominant species in the different regions of the gut are summarized in Figure 1.

3. Microbiota in the Crop

The crop harbors a large bacterial community consisting of bacterial cells in the order of 1 × 108 to 1 × 109 CFU g−1. The crop is colonized predominantly by gram-positive bacteria such as Lactobacillus spp. [26]. Other bacterial species isolated from the crop include Bifidobacterium, Klebsiella pneumoniae, K. ozaenae, Escherichia coli, E. fergusonii, Enterobacter aerogenes, Eubacterium spp., Pseudomonas aeruginosa, Micrococcus luteus, Staphylococcus lentus, and Sarcina spp. [27,28]. Crop microbiota ferments dietary fiber to short-chain fatty acids (SCFA). Acetate is the major SCFA in the crop [29]. SCFA decreases the pH of the crop to inhibit the growth of those pathogens that colonize and proliferate at neutral or slightly alkaline pH [30].

4. Microbiota in the Proventriculus and Gizzard

The proventriculus and gizzard have acidic pH which is not ideal for microbial colonization [31]. Gastric acid can penetrate the cell membrane of microbes resulting in decreased intracellular pH and disruption of the transmembrane proton motive force [32]. Similarly, lactic acid and acetic acid prevent the colonization of pathogens that are sensitive to acidic pH [33]. Lactobacilli is the dominant species in proventriculus and gizzard. Lactose-negative enterobacteria, enterococci, and coliform bacteria are also prevalent in the proventriculus and gizzard [20,25,34]. The bacterial concentration in the gizzard is similar to that of the crop, however, the bacterial fermentation is hindered by the acidic pH resulting in decreased acetate and lactate concentrations in the gizzard [27].

5. Microbiota in the Small Intestine

The concentration of bacteria in the small intestine is approximately 1 × 105 CFU g−1. The microbial density is low in the duodenum due to the low pH, presence of digestive enzymes, high concentrations of bile salts, high oxygen pressure, and pancreatic secretions [31,35]. The conditions are favorable for microbial growth in the distal small intestine. The small intestine is colonized by Lactobacillus, Turicibacter, Enterococcus, Clostridia, and Streptococci [25,27,35]. The predominant SCFA in the small intestine is acetate, although the concentration is considerably lower than that in the caeca [27].

6. Microbiota in the Caeca

Caeca has the highest microbial density of 1 × 1011 CFU g−1 and is comprised mainly of obligate anaerobes of the phyla Firmicutes and Bacteroidetes [36]. Phyla Actinobacteria and Proteobacteria contribute to 2–3% of the cecal microbiota [25]. The cecal microbiota is comprised of gram-positive cocci, Bifidobacterium spp., Clostridium spp., E. coli, Lactobacillus spp., Streptococcus spp., and Bacteroides spp. [37]. The colonic microbiota is almost identical to that of the cecal microbiota and might contain ileal microbiota [25]. The SCFA composition of caeca can vary significantly based on the diet [27]. A study conducted by Walugembe, et al. [38] reported that increasing the insoluble dietary fiber increased the relative concentration of butyric acid and decreased the relative concentration of acetic acid. A pea-containing diet can increase the SCFA concentrations, particularly the acetate and butyrate concentration in the ceca [39]. In a study conducted by Wielen et al. [40], a negative correlation was observed between the concentrations of undissociated forms of acetate, propionate, and butyrate, and Enterobacteriaceae in caeca.

7. Dysbiosis

Dysbacteriosis or dysbiosis is defined as an undesirable alteration of the microbiota resulting in an imbalance between protective and harmful bacteria. The host harbors opportunistic microbes which under certain circumstances will overgrow and become pathogenic to the host. It is important to maintain homeostasis in the gut by preventing the entry of pathogenic microbes while preserving the balance between the existing commensal gut microbiota. The first line of defense against the entry of microbes into the submucosa is the mucin, a glycoprotein polymer secreted by the goblet cells. The outer loose layer of mucin acts as a substrate for the attachment and colonization of the bacteria while the inner compact layer does not permit attachment and colonization of most bacteria [41,42,43]. Gut microbiota regulates the secretion of mucus [44,45] and hence, dysbiosis can induce disruption of the mucin layer [46]. The microbes are separated from the sterile submucosa by a layer of selectively permeable intestinal epithelial cells (IECs) and adjoining tight junctions. The IECs secrete antimicrobial peptides such as defensins, cathelicidins, and C- type lectins to protect the host from the pathogens in the gut. Loss of submucosal integrity or loss of tight junction proteins such as claudins, occludins, and zona occludens can disrupt the barrier leading to the entry of microorganisms from the lumen into the submucosa and ultimately into the systemic circulation [47]. IECs express pattern recognition receptors (PRRs) such as toll-like receptors (TLRs) and nod-like receptors (NLRs) which are regulated by compartmentalization and a higher activation threshold. These receptors are activated by the antigens that breach the intestinal epithelial barrier [48]. The microbial-associated molecular patterns such as lipopolysaccharide, flagellin, and peptidoglycan stimulate the immune system, induce pro-inflammatory signaling, and induce inflammation which will act to eliminate the pathogens [48,49]. To summarize, dysbiosis is prevented by limiting the contact of pathogens with the intestinal epithelium and rapid detection and elimination of pathogens that breach the epithelial barrier.
The gut microbiota lies at the barrier of the internal and external environment of the gut and plays an important role in preventing gut dysbiosis [50]. The gut microbiota ferments complex carbohydrates to SCFA, which is the major energy source for the intestinal epithelial cells. Gut microbiota produces antimicrobial peptides to protect the host against pathogen colonization [51,52]. Gut microbiota stimulates the immune system by releasing ligands such as microbial-associated molecular patterns which bind to the host cell receptors. In a study conducted by Brisbin et al. [53], it was demonstrated that treatment of macrophages with L. acidophilus increased the expression of IFN-α. IFN-α activates the STAT2 pathway to increase MHC class I antigen processing and presentation and T-cell activation leading to the elimination of pathogens and prevention of dysbiosis.
In an ideal state, the beneficial bacteria compete with and prevent the colonization of pathogenic bacteria. An imbalance between beneficial and harmful intestinal microbiota results in dysbiosis [54]. Dysbiosis can be caused by non-infectious factors such as dietary changes, non-starch polysaccharides (NSPs), and mycotoxins or infectious agents such as Clostridium perfringens and Coccidia [55,56] which are summarized in Figure 2. Dysbiosis in broilers is characterized by intestinal inflammation and villus atrophy of the small intestine [57]. For many years, the use of antibiotic growth promoters (AGP) prevented dysbiosis in chickens. However, with the restrictions on the use of AGPs dysbiosis emerged as a challenge [58,59]. The most common factors that can cause dysbiosis are discussed below.
Stress can be of dietary, environmental, or managemental origin and can have detrimental effects on the health and production of poultry. During stress, the body redirects the nutrients towards inflammation which will decrease production [60,61]. Feed ingredients directly or indirectly influence the composition of gut microbiota, gut physiology, and gut-associated mucosal immune responses and can contribute to dysbiosis [62].
Non-starch polysaccharides (NSP) are naturally present in grains such as wheat, rye, and barley. NSP includes cellulose, glucans, inulin, arabinoxylans, chitin, gums, and mucilages and constitutes a major portion of dietary fiber [63,64]. Insoluble NSPs are resistant to digestion by mammalian enzymes and are fermented by the gut microbiome into SCFA [65]. However, soluble NSPs have negative effects on villus height, width and surface area, and gut immune response in poultry. Soluble NSPs increase the viscosity of gut digesta, increase the transit time and create an anaerobic environment in the gut [66,67] leading to colonization by pathogenic anaerobic microbes such as C. perfringens and Escherichia coli and acute inflammation [68,69].
Dietary proteins play a significant role in modulating the immune response. Amino acid composition of dietary protein modulates the immune system by altering the gut microbial composition [70]. Poultry feeds include plant protein such as soybean meal, and cottonseed meal, or animal protein such as fish meal [71]. A high proportion of animal proteins, which are rich in glycine and methionine, favors colonization by pathogenic C. perfringens and high levels of fish meal in poultry diets can lead to C. perfringens overgrowth and induce necrotic enteritis in chicken [72]. Undigested low-quality protein in the caeca will be fermented by caecal microbiota, in a process termed putrefaction, to produce toxic metabolites such as indole and cresol leading to inflammation and intestinal damage [73].
Heat stress is an environmental factor that negatively impacts chicken production performance and health. Heat stress decreases the expression of tight junction proteins resulting in leaky gut syndrome [74]. Heat stress can also cause immunosuppression, lipid peroxidation, and endocrine imbalances [75] leading to dysbiosis in chickens. In a study conducted by Park et al. [76], it was identified that heat stress increased the proliferation and colonization of enteric pathogens such as Escherichia, Salmonella, and total aerobic bacteria.
Mycotoxins are a diverse class of toxic secondary metabolites produced by fungi [77] and molds belonging to the genera Aspergillus, Penicillium, and Fusarium [78]. Because mycotoxins are present in the feed, the gut is the primary organ affected by mycotoxins [79]. The gut microbiome can metabolize mycotoxins, thereby affecting toxicity. Microbial enzymes are capable of chemical transformation of mycotoxins [80]. The toxicity of aflatoxin, a common mycotoxin, is dependent on the transformation by gut microbes. Mycotoxins can cause dysbiosis by their direct antimicrobial properties or by an indirect effect on intestinal cells. Mycotoxins cause apoptosis of the intestinal epithelial cells, increase intestinal permeability, increase mucus production, damage the intestinal villi, and disrupt tight junction proteins [81]. Mycotoxins increase the production of proinflammatory cytokines, immunoglobulin A, and antimicrobial peptides to alter the gut microbial composition [77,80].
Antibiotic growth promoters (AGPs) improve the performance and health of chickens by altering the gut microbiome. AGPs inhibit the gram-positive bacteria and pathogenic bacteria which compete with the host for nutrients [82] or produce toxins [83]. AGP reduces the total number and diversity of the gut bacteria [84], though the loss in gut microbial diversity due to the use of AGPs can also lead to dysbiosis in poultry. Gut infections can alter the balance in gut microbiota. In a study conducted on the effect of Eimeria tenella infection on the cecal microbiome diversity, it was identified that the diversity of cecal microbiota remained stable, but there were significant changes in the abundance of some microbial taxa. In birds with severe cecal pathology, there was a decrease in taxa belonging to the order Bacillales and Lactobacillales and an increase in Enterobacteriaceae. In asymptomatic birds, there was an increase in Lactobacillus and a decrease in Bacteroides. The variations in gut microbiota correlated with the E. tenella lesion severity [85].

8. Probiotics in Broiler Production

With the ban on the use of antibiotics by the European Union (EU) and the limited use of antibiotics in the United States (US) in chicken production, probiotics are emerging as a potential alternative. Probiotics or direct-fed microbials (DFM) are defined as “live microorganisms which when administered in adequate amounts confer a health benefit on the host”, by improving the gastrointestinal microbial balance and consequent enhancement in nutrient absorption, growth rate, feed efficiency, and immune competence against potential pathogens [84,86,87]. An ideal probiotic should be of host origin, have demonstrated a beneficial effect on the host, should be able to alter the gut microflora advantageous to the host, should be resistant to gastric acid and bile salts, should adhere to the gut mucosal epithelium, and should exhibit beneficial effects. Probiotics should also be non-pathogenic, free of adverse side effects, sensitive to antibiotics, exhibit antimicrobial properties against common pathogens, modulate intestinal functions, and immune responses of the host [88,89]. The probiotic should also be able to withstand the processing and storage conditions [90].
Ideally, probiotics should be host-derived because 1. The microbes have evolved and adapted to the ecology of the host gastrointestinal tract and hence, can readily proliferate and establish a stable population 2. Express beneficial effects better than probiotic strains obtained from other species [91]. It is therefore essential to develop host-specific probiotics for achieving optimal health and production. Probiotic bacteria should be typically isolated from the gut of healthy animals. Multiple species and strains of gut microbes must be evaluated for their probiotic potential and the one conferring maximum benefit to the host must be selected [92]. For example, only 6 out of 57 lactic acid bacteria (LAB) strains, isolated from the crop and intestine of healthy broilers, possess probiotic properties that included the ability to adhere to ileal epithelial cells, hydrophobicity, tolerance and survivability in gastric juice, bile salts, and phenol, susceptibility to antibiotics, ability to auto-aggregate and co-aggregate [93].
Poultry is a major source of foodborne pathogens of zoonotic significance [94] and foodborne pathogen infection cause economic loss to the poultry industry. Probiotics possess antimicrobial activity and can be applied to decrease the incidence of foodborne pathogens in poultry [95]. Probiotics secrete bacteriocins, hydrogen peroxide, alcohol, and organic acids and exhibit broad spectrum antimicrobial activity [96]. In a study conducted to evaluate the probiotic efficiency of LAB isolated from poultry, it was demonstrated that 77 strains out of the 296 strains screened inhibited the proliferation of E. coli and Salmonella enteritidis. Of the 77 strains, eight strains of their greater spectrum of pathogen antagonism and other desirable properties of probiotics. The strain with maximum probiotic potential was identified as L. salivarius [97].
The probiotic strains should be able to survive the acidic pH in the proventriculus and gizzard for the strains to successfully colonize the intestine [98]. The transit time of feed through the proventriculus and gizzard is approximately 2 h. L. acidophilus survives at pH 4 for 2 h [99]. pH tolerance is strain specific. For example, L. plantarum VJI21 exhibit 80% survivability whereas the isolate VJC1 exhibit only 64% survivability at low pH [100].
Potential probiotic strains should be able to withstand bile salts in the duodenum and cecum [101]. Probiotics of host origin will be able to tolerate the bile salt concentration of the host from which the probiotic strain was isolated. L. acidophilus can survive for 15 h in 2% bile. Lactobacillus strains possess bile salt hydrolase activity and therefore neutralize the antimicrobial activity of bile salts to survive in the gut [100].
The ability of probiotics to adhere to the intestinal epithelial cells and mucosal surfaces is essential for forming biofilms, which prevents the detachment of bacteria during gut contraction and peristaltic movement. This property of biofilm formation is termed auto-aggregation [102]. Co-aggregation enables the probiotic species to displace pathogenic bacteria [101]. Hydrophobicity enables probiotics to adhere to the gut epithelium [97]. Auto-aggregation, co-aggregation, and microbial hydrophobicity enables the probiotics to competitively exclude pathogenic species from the host gut [100,102,103]. L. acidophilus decreases the adhesion of E. coli and S. typhimurium to Caco-2 cells [99].
Antibiotic susceptibility of potential probiotics should be evaluated to ensure no antibiotic resistant genes are transferred from the probiotic species to the gut microbiota [100]. In a study evaluating antibiotic susceptibility of Lactobacillus strains isolated from chicken, 90% of the 88 isolates were resistant to tiamulin, 74% to tetracyclines, and 70% to lincomycin. Multidrug resistance was detected in 79.5% of isolates [104]. These results indicate that caution should be applied before choosing a particular probiotic species as the probiotic species might carry antibiotic resistant genes.
Probiotic organisms should be consumed at adequate doses to confer a desired health benefit. A minimum viable cell concentration of 1 × 106 CFU g−1 is essential for successful colonization of the gut by probiotics [105]. Probiotic supplementation at 1 × 108 to 1 × 109 CFU g−1 is effective and can increase the number of beneficial bacteria [90,106]. In a study conducted in broiler chickens by Mountzouris et al., 2010 [106], it was demonstrated that there is a significant increase in the concentration of Lactobacillus and Bifidobacterium and a decrease in coliform counts in the caeca of birds fed probiotics. Microbiological factors such as the strain of the probiotic, water activity, pH, presence of salt, and molecular oxygen determine the survivability of probiotics during storage. Feed processing parameters such as heat treatment, cooling rate, storage length, and packaging materials also determine the survivability of probiotics [107,108,109]. The survivability of probiotic bacteria during the processing and storage of the product is dependent on the species and strain of probiotics [110]. Thermotolerance of Lactobacilli can be increased by subjecting the bacterium to heat shock at sublethal temperatures, which is 10 °C above the optimum temperature for growth, before exposure to lethal temperatures [111]. Bacillus spores can survive at high temperatures and are stable during feed processing steps such as pelleting [105].
Probiotics can be a single strain or a combination of different microorganisms such as bacteria of the species Lactobacillus, Bifidobacterium, Bacteroides, Bacillus, or Enterococcus. Saccharomyces spp., a yeast, is used as a probiotic either alone or in combination with other bacterial strains [112]. Probiotic preparations can also contain multiple strains of the same species such as Lactobacillus casei, L. acidophilus, L. bulgaricus, L. reuteri, L. salivarius, and L. animalis [90,113,114]. Probiotics can also be categorized as ‘colonizing’ species which consists of Lactobacillus, Streptococcus, and Enterococcus spp., and ‘non-colonizing’ species which consists of Bacillus spp. spores and the yeast Saccharomyces cerevisiae [115]. Bacteria of the colonizing species compete for potential binding sites on the intestinal epithelium or mucosa while non-colonizing species though viable in the intestinal content, will not colonize the intestinal epithelium [116]. Colonizing species resist the growth of pathogens by competitive exclusion. In contrast to the requirement for continuous administration of non-colonizing species, colonizing species requires to be administered once, though a supplementary dosage at a later age can be beneficial [117]. Gut microbiota is also classified as luminal and mucosal associated microbiota. The expression of adhesion molecules on the intestinal epithelial cells, rate of mucus production, and secretory immunoglobulins determine the composition of mucosal microbiota [118]. The composition of luminal microbiota is influenced by the presence of antimicrobial substances, feed passage rate, nutrient availability, and the interaction between the microbiota and the host immune system [94,110].
Among the different probiotics that are available, three major microorganisms, namely Lactic acid bacteria, Bifidobacteria, and Saccharomyces, dominate the probiotic market [119]. Other probiotics that are available for the poultry industry include Enterococcus, Pediococcus [120], Bacillus, E. coli, Aspergillus, Candida, and several other microbial species [121]. Although the probiotics industry is rapidly growing with several commercially available probiotics flooding the market, at any given time it is customary to supplement birds with a combination of a limited number of four or five microbial species [122]. It is not clear how the supplemented four to five different strains can contribute to the bacterial diversity in healthy chickens or even restore diversity in antibiotic treated birds. In humans treated with antibiotics, gut mucosal microbiome reconstitution is impaired by probiotics suggesting that probiotics might prevent the original gut population from recovering to the pre-antibiotic phase [123].
One alternative to applying limited numbers of probiotic species would be to transfer gut microbiota from healthy adult chickens to young chicks. The poultry industry will benefit from transferring mixed cultures of gut microbiota from healthy chickens rather than supplementing a limited number of probiotic species. Such an attempt has been extensively applied to control Clostridium difficile infection in humans. Fecal microbiota transplantation is the transfer of fecal matter from healthy donors into the gut of recipients to alter the gut microbiome of the recipient [124]. Fecal microbiota transplantation increases gut microbiota diversity and restores the gut microbiome post-antibiotic therapy. Post-transplantation, the fecal microbiota of fecal transplant recipients was more diverse and similar to that of the healthy donor [125]. Such an approach was attempted in early 1973, wherein transferring an undefined mixed culture from healthy chickens, but not a single Lactobacillus probiotic species, successfully inhibited Salmonella colonization in chicks [126]. This procedure was termed the Nurmi competitive exclusion concept [121] and has since been applied to control salmonellosis and campylobacteriosis in poultry [127].
It is necessary to understand the symbiotic relationship between the host and the microbiota. A nutrient-rich ambient environment of the host facilitates the colonization and proliferation of microbiota while the microbiota stimulates the digestive and immune systems of the host to improve the production performance [89,128]. In a meta-analysis study conducted by Blajman et al. (2014) [129], it was identified that probiotic supplementation improves BWG by 661 g and improves FCR with 281 g less feed consumed/kg of weight gain. The meta-analysis study also concluded that administration of probiotics in drinking water is more efficacious compared to supplementation in feed. The major limitations impeding the use of probiotics are the inconsistent effects and the incomplete understanding of the mode of action of probiotics.

9. Routes of Administration

Probiotics should be administered as early in life as possible to achieve desired beneficial effects [130]. Supplementation in feed or water on the day of hatch is the most common and convenient method to apply probiotics in poultry production [86]. Supplementation in feed or water is dependent on the individual consumption and thus the dose of probiotic consumed varies depending on the quantity of feed or water consumed. Supplementing probiotics in feed and water provide a mean for continuous supplementation of the probiotics to the bird, though the presence of antimicrobials in food or water can affect the viability of the probiotic bacteria [131,132].
Several studies have reported higher efficiency of probiotics when supplemented in drinking water [129,133]. This can be due to the shorter transit time of water, compared to the feed, in the upper gastrointestinal tract. A shorter transit time will reduce the duration for which probiotics are exposed to lower pH and bile salts in the upper gut. Water dilutes the acid, enzymes, and other digestive secretions and thereby reduces their negative effects on probiotics [132]. Bacteria require a period of acclimatization to the new environment. This period of time, termed the lag phase, will be longer when the conditions are less optimal. Supplementing probiotics in water mimics the reconstitution of lyophilized products and reduces the lag phase, thereby enabling the bacteria to adapt better and proliferate in the gut [132,134,135].
Probiotics can be injected into the incubating egg to achieve early colonization of the beneficial bacteria [136]. Delivery of probiotics to chicken embryos in ovo establishes a healthy gut microbiome [137,138]. In ovo probiotic inoculation can be applied either as in ovo stimulation [139] or in ovo feeding [140] which are inoculation on day 12 and day 17–18 of egg incubation respectively. In in ovo stimulation, prebiotic species are deposited on the air cell on day 12 to stimulate the development of innate gut microflora, which is ingested when the chicks start piping [141]. In in ovo feeding, probiotic species are injected into the amnion or embryo [142,143,144]. In a study conducted by Oliveira et al. [136], it was demonstrated that in ovo inoculation of probiotics significantly reduced mortality when challenged with Salmonella on day-4 post-hatch compared to the control group. In ovo inoculation of probiotics facilitates early colonization of beneficial bacteria and competitively excludes Salmonella. However, in ovo injection of one or a mixture containing only a few beneficial bacteria will likely not be effective in establishing a diverse intestinal microbiome comparable to that of a healthy adult chicken. On the other hand, inoculation of a mixed culture can introduce unknown species of bacteria that could be detrimental to the embryonated egg [145].
Spraying is another method of probiotic administration wherein the undiluted culture is sprayed on chicks when they are 50–70% hatched [146]. Cultures can also be sprayed in the environment or the litter material of the bird. Spraying eliminates the water quality concerns and other variables associated with probiotic administration in feed or water. Spraying is a cost-effective and suitable for mass applications [147,148]. Probiotics can also be delivered by oral gavage, but the high labor requirement makes the route of delivery unfeasible [143].

10. Postulated Mode of Action and Effects of Probiotics

10.1. Competitive Exclusion and Antagonism

Competitive exclusion refers to the phenomenon in which a strain of bacteria competes and prevents the colonization of enteric epithelium by other bacteria [149]. Probiotics inhibit the growth of pathogenic bacteria and selectively enhance the growth of beneficial microorganisms through several mechanisms [150]. Probiotics produce organic acids and SCFAs such as lactate, propionate, acetate, and isovalerate to lower the pH of the gut and thus inhibit the colonization and growth of pathogenic microbes [151]. Lactobacillus and Bacillus spp. secrete bacteriocins to inhibit E. coli, Vibrio spp., Salmonella, Proteus, Campylobacter, C. perfringens, Staphylococcus aureus, and Shigella [152]. Our group earlier identified that the cell-free supernatant of P. acidilacti can inhibit the proliferation C. jejuni in vitro at 1:1 supernatant: pathogen dilution [153]. Probiotics also compete with pathogenic bacteria for nutrients and adherence to the gastrointestinal epithelium. We earlier found that probiotic supplementation during Salmonella challenge significantly reduced the cecal Salmonella load [154]. Lactobacillus plantarum hinders the adherence and translocation of C. jejuni to the enteric epithelium by inducing mucin secretion [155]. Probiotics can act as an adjuvant to stimulate the immune system, thus inhibiting pathogen colonization and reducing mortality [149,156].

10.2. Host Intestinal Health and Integrity

The three principal regions of probiotic colonization in the gut are the enterocytes, colonic and cecal epithelium [157]. Probiotics enhance the epithelial barrier integrity by increasing the production of mucus [158]. Probiotic bacteria enhance mucin secretion by the upregulation of MUC2 in goblet cells [159]. Probiotic bacteria regulate tight junction permeability through upregulating zonulin, a protein that regulates tight junction permeability [160]. In a study conducted in our laboratory, we demonstrated that supplementing B. subtilis to the chicken during necrotic enteritis increased the mRNA expression of claudin proteins 2.1-fold compared to the challenged control group [161]. B. longum, Pediococcus pentosaceus, L. plantarum, and L. acidophilus scavenge free radicals and thereby reduce host DNA damage and thereby improve gut integrity. Probiotics also reduce lipid peroxidation of the gut epithelial cells and maintain gut integrity [162].
The host immune system can distinguish pathogenic bacteria from probiotic bacteria [163]. It was demonstrated that the host produces IL-8 in response to pathogenic bacteria infection. Probiotic bacteria did not induce the production of IL-8 and reversed the pathogen-induced increase in IL-8 [164]. Probiotic bacteria induce the production of antimicrobial peptides and protective cytokines such as IL-12, IL-10, TNF-α, and IFN-γ through their interaction with the Toll-like receptors (TLR) on epithelial cells. This enhances the regeneration of intestinal epithelial cells and decreases apoptosis in intestinal cells by inhibiting the pro-apoptotic pathway (MAPK/p38) and activating the anti-apoptotic pathway [160,165]. In a study conducted by Gharib et al. [166], it was demonstrated that Bacillus amyloliquefaciens supplementation downregulates IFN-γ expression and upregulates IgG and IgM. Bacillus amyloliquefaciens supplementation decreased caspase-3 and enhanced occludin expression to maintain gut integrity [166].

10.3. Digestion and Absorption

Probiotics increase the nutritional value of the feed by fermenting undigestible carbohydrates. The fermentation of undigested carbohydrates and proteins in the ileum and colon by probiotics produces SCFAs, which provide energy to the host [167]. DFM supplementation increased the epithelial flux of D-glucose, DL-methionine, and L-lysine and increased trans-epithelial resistance, which is an indicator of the tight junction integrity [168]. Probiotic supplementation increases the ratio of villus height to crypt depth resulting in increased intestinal absorption of nutrients [169]. We earlier identified that probiotic supplementation during Salmonella challenge in broilers reversed the loss in villus height and crypt depth [154]. The effect of probiotics on villus height and crypt depth can be correlated with the increased FCR and BWG in the probiotic supplemented birds. Some probiotic bacteria such as Bifidobacterium and Lactobacillus spp. synthesize vitamins such as vitamin A, vitamin K, folate, nicotinic acid, pyridoxine, riboflavin, thiamine, cobalamin, pantothenic acid, biotin, and increase the activity of antioxidant enzymes such as glutathione peroxidase, catalase, superoxide dismutase, and glutathione S-transferase which adds to the nutritional value of the probiotic [170,171,172]. The exopolysaccharides (EPS) produced by probiotic bacteria aid in biofilm formation and modulate the immune system [173]. Probiotics secrete extracellular enzymes such as amylases, phytases, proteases, and lipases to increase feed digestibility [174].

10.4. Performance Parameters

Probiotics enhance the health of birds and consequently improve production performance. Probiotics improve feed intake by decreasing gastric emptying time [175]. The increased weight gain can be attributed to better feed digestibility associated with the secretion of the enzymes amylase, lipase, and protease to improve nutrient availability [176]. Probiotic supplementation improves the live weight gain, carcass yield, and cut-up parameters in broilers [177]. Probiotic supplementation improves the microbiological quality of meat by reversing the pathogen-induced loss in gut permeability [121]. Probiotics are capable of modulating bone health by regulating the absorption of calcium and phosphorus in the intestine and secretion of neuroendocrine molecules (incretins and serotonin). Gut dysbiosis leads to decreased bone density and strength [178,179]. Yan et al., 2019 [180] demonstrated that probiotic supplementation increased mineralization of the femur and tibia resulting in significantly higher bone strength and gait score. Nevertheless, some studies have reported decreased growth and performance parameters associated with the supplementation of probiotics and symbiotics [181,182]. Several factors such as the probiotic strain, dosage, general health of the bird, sex of the bird, duration of poultry house downtime, and environmental contamination levels determine the success of probiotic application in poultry [183].
Probiotic supplementation mitigates heat stress by restoring the intestinal microbial balance and preventing dysbiosis associated with heat stress [180]. Heat stress decreases ileal and caecal Bifidobacterium and Lactobacillus loads and increases coliforms and Clostridium loads, which was reversed by probiotic supplementation [184]. Probiotics decrease the expression of heat shock proteins and decrease the adhesion sites for pathogenic bacteria and thereby decreasing pathogenic bacteria colonization [185].

10.5. Immunomodulation

The gastrointestinal system is considered to be the largest immune organ. The gut-associated lymphoid tissue (GALT) is constantly being exposed to environmental and dietary antigens [158]. The innate immune system is usually the first line of defense against pathogenic intestinal microbiota [13]. Mucins and antimicrobial peptides such as defensins and lysozyme are secreted by epithelial cells in response to cues from the intestinal microbiota. Mucins cover the epithelium and regulate the diffusion of macromolecules, nutrients, gases, and toxins while serving as an adhesion site for resident microbiota [186,187]. Probiotic bacteria produce SCFAs which stimulate the expression of mucin glycoprotein. Butyrate is utilized as an energy source by the colon epithelial cells, improves gastrointestinal function, and exerts anti-inflammatory effects by inhibiting NFκB [188]. In a study conducted by Zhou et al. [189], it was demonstrated that butyrate supplementation in poultry significantly decrease nitric oxide production by macrophages in a concentration dependent manner by possibly inhibiting the activation of NFκB. Butyrate also inhibits the expression of pro-inflammatory cytokines IFN-γ, IL-6, and IL-1β in LPS-activated macrophages.
Intestinal epithelial cells and intraepithelial lymphocytes express TLRs which are PRRs and can be classified as the ‘gate keepers’ of the innate immune system. The binding of ligand to the PRRs initiates a cascade of signaling pathways that include interferon response factors, mitogen activated protein kinases, NFκB, and activator protein 1 leading to the secretion of proinflammatory cytokines [190]. TLR signaling plays an important role in maintaining the epithelial barrier function, intestinal epithelial homeostasis, and epithelial cell proliferation. In the enterocytes, TLRs are expressed on the basolateral surface to prevent the interaction of TLRs with commensal microbiota. However, TLRs are also expressed on the luminal surface to a lesser extent which plays an important role in gut homeostasis [191]. Stimulation of the TLRs by the normal gut microbiota induces the production of IL-6 and KC-1 which are tissue protective factors [192]. Pathogens equipped with virulence factors traverse the epithelium, resulting in the activation of TLRs located in IEC, macrophages, and dendritic cells resulting in inflammation [193]. Probiotic bacteria preferentially activate the TLR9 pathway which attenuates NFκB activation and IL-8 expression. Probiotic bacteria activate the TLR9 signaling pathway and upregulated the anti-inflammatory cytokines in mice [194]. This study suggests the significance of TLR signaling by probiotic bacteria in maintaining gut homeostasis.
Probiotics increase the expression of TLRs, increase antibody production, and increase the number of intraepithelial lymphocytes in the gut [195]. Probiotics induce the secretion of IgA in mucins to inhibit pathogen colonization [196]. Probiotics can enhance macrophage activity, induce specific and non-specific humoral and cellular immune responses, enhance cytokine production and secretion of antibodies (IgA, IgG, and IgM) [170]. In a study conducted by Bai et al. [197] probiotic supplementation significantly increases the CD4+, CD8+, CD3+, and intraepithelial lymphocyte populations. Probiotics supplementation increases the expression of TLR 2 and TLR 4. Probiotics maintain the balance between the pro-inflammatory and anti-inflammatory cytokines [198,199]. Probiotics shift T-cell maturation towards the Th1 pathway through the modulation of cytokines [156]. In contrast to pathogenic bacteria, which activate the proinflammatory transcription factor NFκB, probiotic bacteria activate the regulatory factor IκB thus exerting anti-inflammatory effects. Inhibition of the NFκB-pathway and the increased production of cytoprotective heat shock proteins by probiotics confer an advantage during heat stress [200].

11. Factors to Be Considered during Probiotics Supplementation

The survivability of the probiotic organisms in the upper gastrointestinal tract is of paramount importance for the probiotic species to colonize the gut. Microencapsulation preserves the viability of probiotics in a feed matrix. However, in vitro tests with simulated gastric juices are typically applied to study the efficiency of microencapsulation in maintaining the viability of probiotics [201]. The high complexity of the gastrointestinal tract of live animals with variable pH, gastric and intestinal juices, peristalsis, and presence of gut microbiota makes in vitro studies to be of low predictive value for in vivo success. L. casei failed to colonize mice’s gut when supplemented with sodium caseinate capsules [202]. More studies have to be conducted using in vivo chicken models to assess the protective effect of microencapsulation on probiotic survival.
Even if the commercial probiotics remain viable on reaching the poultry gut, direct evidence to show the colonization of probiotic species post supplementation is lacking. The survival and colonization of a probiotic species are a function of several factors including bile salts and pH. Considering that the pH of the chicken intestine ranges from 3.1 to 6.6 [203], it is possible that the supplemented probiotics may even fail to colonize the gut. The classic approach to identifying the survival and colonization of the in-feed probiotic species is to isolate them by cultivation in species-specific agar plates [204]. But this technique has limitations in that the supplemented probiotic species have no antibiotic resistance genes and species-specific media is not available for most of the probiotic species. Advanced genome-based techniques though have been extensively used to determine the diversity and structure of the microbial population, they cannot quantify individual bacterial species. Because of these constraints, very rarely attempts have been made to quantify the survival and colonization of the supplemented probiotics. A recent article from our group identified that out of the five supplemented probiotics, only four species successfully colonized the gut post supplementation and were identified at levels ranging between 0.03 and 1.2% depending on the region of the intestine and the probiotic species studied [205]. Among the sparse articles that measured the survival of the supplemented probiotics in the poultry gut, it is not clear whether it can be stated with certainty that the probiotics, in fact, colonized the gut considering the fact that humans supplemented with probiotics only transiently increased the probiotic species concentration in their feces [206].
One of the major factors that can prevent the colonization of the gut by the supplemented probiotics is “Colonization resistance”. Colonization resistance is loosely defined as a mechanism by which the resident microbiota prevents colonization of exogenous microorganisms [207] where the existing microorganism prevents the probiotics from colonizing the gut. In mice colonization of the gut was limited by the colonization resistance of the existing microbiome [206]. Humans can be classified as “permissive” or “resistant” to the colonization of a particular strain of probiotics based on their genetic makeup [208] and the probiotic colonization pattern of individual humans can be predicted using the microbiome features of the individual [206]. The poultry industry might benefit from probiotics that are designed for specific breeds of birds so that the bacterial strains in the probiotics have a greater chance of colonization.
Another extension of these individualized breed-specific probiotics would be developing probiotics specific for age. Although humans [209], mice [210], and poultry [211] show age-specific differences in their gut microbiota composition, it is common to find that the same combination of four to five probiotic species is being fed to birds of all ages to counter all disease conditions. Developing age-based probiotic supplements should be one of the future approaches for developing new probiotics for the poultry industry.
Most probiotics are a normal inhabitants of the host gastrointestinal tract. The presence of antimicrobial resistance genes in bacteria such as Bifidobacteria and Lactobacilli by itself is not a matter of concern because of their lack of infectivity. Antibiotic resistance in probiotic bacteria is desirable to some extent as it can restore the gut microbiota after antibiotic therapy [212]. But the abundance of these bacteria in the gut presents the risk of transfer of antibiotic resistance genes from these probiotic species to pathogenic bacteria [213]. Transfer of these genes from animals or meat to humans through feces, soil, water, or food can result in the development of antibiotic resistance in human pathogens and treatment failure during infection [214]. Consumption of raw or undercooked meat can also result in the transfer of antibiotic resistance genes harboring zoonotic pathogens such as Salmonella and Campylobacter from poultry [215,216].
Generally, probiotic supplementation is associated with improved performance parameters including enhanced weight gain, superior carcass quality, increased carcass yield, improved meat sensory characteristics, and increased egg size, shell strength, shell thickness, and weight [217,218,219]. A commonly used bacterial species in probiotics is Lactobacillus spp., which is known to reduce gastrointestinal pathogen load, stimulate the immune system, and improve the growth and performance of the bird. However, direct exposure of rooster semen to Lactobacillus acidophilus was observed to be detrimental to semen quality. In a study conducted by Haines et al. [220], it was observed that exposure of rooster semen to Lactobacillus and Bifidobacterium virtually rendered the sperm immotile. The pH of semen was also found to be significantly reduced on exposure to these bacteria. This could be due to the direct attachment of the bacteria to the sperm, obstructing the movement of sperm by the non-motile bacteria, or reduction in pH due to the production of lactic acid. Therefore, long-term supplementation of Lactobacillus and Bifidobacterium containing probiotics to breeder stock should be done with caution.

12. Postbiotics and Paraprobiotics

The efficacy of probiotics is associated with the viability of the microorganisms used. However, there is evidence that the viability of the microbes is not a requisite for conferring the beneficial effects of probiotics to the host [221]. The emergence of terms such as ‘postbiotics’, ‘paraprobiotics’, ‘metabiotics’, ‘inactivated probiotics’, and ‘ghost probiotics’ indicates that supplementing non-viable microbes or microbial products can also confer health benefits to the host. Postbiotics are low molecular weight soluble factors that are either secreted by live bacteria or released after bacterial cell lysis and when administered in sufficient amounts confer health benefits to the host [221]. Postbiotics are obtained by disrupting the microbial cell structure through heat treatment [222], enzymatic treatment [223], solvent extraction [224], or sonication [225]. Postbiotics are further purified through centrifugation, column purification, freeze-drying, microfiltration, and dialysis [226,227].
Paraprobiotics also termed as “inactivated probiotics” or “ghost probiotics”, are non-viable probiotic or non-probiotic microbial cells or cell fractions which when administered in sufficient amounts confer beneficial effects to the host [228,229]. Paraprobiotics are essentially non-culturable, but immunologically active microbial cells that benefit the host. Thermal treatment, high pressure, irradiation, and sonication, which induce cell death without membrane degradation, are applied in the production of paraprobiotics [228,229,230,231,232]. Flow cytometry using fluorescent dyes is used to assess the functional state of inactivated cells [233].
Postbiotics (Nonbiotics) are low molecular weight non-viable factors such as metabolic products, byproducts, or cell wall components (metabiotics, cell-free supernatants, secretions, cell lysates, or biogenic metabolites) derived from probiotic microorganisms which when administered in sufficient amounts confer health benefits to the consumer [221,229]. Postbiotics can be soluble substances secreted by the live bacteria or the products of bacterial cell lysis, as summarized in Figure 3. This soluble fraction contains SCFAs, bacteriocins, vitamins, peptides, organic acids, hydrogen peroxide, enzymes, cell surface proteins, plasmalogens, peptidoglycan-derived muropeptides, teichoic acids, exopolysaccharides and endopolysaccharides [229,234,235]. The exact mechanism of action of postbiotics is not completely elucidated. In a study conducted by Yan et al. [236], it was demonstrated that Lactobacillus rhamnosus derived protein p40 activates EGFR and prevents the cytokine-induced apoptosis of intestinal epithelial cells in inflammatory conditions such as inflammatory bowel disease (IBD). In a study conducted by Humam et al. [237], it was demonstrated that the birds fed postbiotics derived from different strains of L. plantarum exhibited significantly improved FCR values compared to the control group. Postbiotic supplementation increased the expression of IGF-1 and growth hormone receptors during heat stress [237].
Although the mechanism of postbiotic action is not completely explained, scientific data provide evidence for the role of postbiotics in several physiological functions. Postbiotics can be classified based on their structural composition or their physiological functions. Based on their role in host physiology, postbiotics can be 1. antimicrobials such as cell-free supernatants of L. plantarum [238], 2. immunomodulatory substances such as cell wall components of Bifidobacterium and Lactobacillus, lipoteichoic acid of L. plantarum [224], 3. anti-inflammatory factors such as cell free supernatant of L. paracasei and L. rhamnosus [236,239], 4. anti-proliferative substances such as sonicated cell suspension of L. casei [240], 5. anti-obesogenic compounds such as fragmented cells of L. amylovorus [241], 6. Hypocholesterolemic substances [231], 7. anti-hypertensives such as polysaccharide glycopeptide complexes of L. casei [227], and 8. antioxidants such as intracellular contents of Streptococcus salivarius, L. acidophilus, and L. casei [236,240]. Supplementation of bacteriocin from L. plantarum improves growth rate, increases the fecal LAB population, and reduces the abundance of Enterobacteriaceae in broilers [242]. Thus, the concept of feeding live microbials is being replaced by postbiotics which have been demonstrated to confer similar health benefits to the host.
Based on the composition, postbiotics can be 1. carbohydrates such as galactose-rich polysaccharides and teichoic acids 2. lipids such as propionate and butyrate 3. proteins such as p40 molecule and lactocepins 4. organic acids such as propionic acid and 3-phenyl lactic acid 5. vitamins such as B-complex vitamins 6. complex molecules such as lipoteichoic acids and peptidoglycan derived muropeptides [234,243,244]. In a study conducted by Abd El-Ghany et al. [245], it was reported that supplementation of inactivated Lactobacillus to chickens improved the immune response to Newcastle disease virus vaccines. The improved immune response of the birds is due to the lipopolysaccharides, lipoteichoic acid, and teichoic acid present in the bacterial cell wall which acts as adjuvants to stimulate the host immune response. Lysis of the inactivated bacteria in the host gut releases nuclear antigens which might stimulate the host humoral and cell-mediated immune responses.
One of the advantages of postbiotics and paraprobiotics over probiotics is the possibility of postbiotics and paraprobiotics applications in immunocompromised patients. Administration of live microbes presents a risk to immunocompromised patients because of the possibility of occurrence of opportunistic infections and metastasis of probiotic microbes. For instance, translocation of the Streptococcus gallolyticus from the gut to the bloodstream has been implicated in colorectal cancer in humans [246]. Paraprobiotics or postbiotics such as SCFA have antimutagenic activities and selectively target cancer cells and can be applied to treat cancer patients [244]. Postbiotics have a clear chemical structure, longer shelf life, and safety dose parameters. In a study conducted by Shigwedha et al. [247], it was observed that probiotic cell fragments, the structural components of probiotic cell lysates, exert a broad spectrum of immunomodulatory functions. The shelf stability of probiotic cell fragments can be up to 5 years. The use of postbiotics can also eliminate the risk of prevalence and transfer of antibiotic resistance genes from probiotic species to the consumer [248].

13. Future Prospects: Designer Probiotics and Postbiotics

The concept of designer probiotics, where probiotic strains are genetically modified to improve gut survival and resistance to stress to enhance therapeutic effects has been attempted in humans [249]. For example, Bifidobacterium breve that was modified to increase the expression of BetL, a betaine uptake system, had a considerably improved osmotolerance, survival in simulated gastric juice, and growth rate [250]. Several such modifications have been attempted to improve the quality of human life. Genetically engineered Lactobacillus reuteri modified to express phenylalanine ammonia lyase enzyme have been applied to treat phenylketonuria [251].
Although designing probiotics to treat chicken genetic diseases can be an overstretch, the development of designer probiotics to treat or prevent microbial diseases or control foodborne pathogens is a real possibility in near future. Recombinant Lactococcus lactis that secrete antimicrobial peptides such as A3APO and Alyteserin inhibited the growth of pathogenic E. coli and Salmonella by 20-fold [252]. Supplementing Lactococcus lactis, modified to express the VP8 spike protein of the human rotavirus, provided 100% protection against rotavirus infection in a murine model [253]. Similarly, probiotics have been successfully designed to sequester toxins produced by Shiga toxigenic E. coli, Vibrio cholerae, Clostridium difficile, and Listeria in human and mice models of infection [254,255]. Considering the economic burden caused by Salmonella and E. coli to the poultry industry and since no single solution exists to control any foodborne pathogen of poultry origin including Salmonella and E. coli, a logical future step would be to develop genetically modified probiotics to control foodborne pathogens. Also, supplementing probiotics capable of producing antimicrobial peptides can overcome the limitations such as short half-life, production costs, and purification costs associated with the production of antimicrobial peptides.
Probiotics can be modified to deliver immunogenic molecules and antigens to the mucosal surface and hence can act as vaccines. Oral vaccination of mice with L. casei, modified to express the enterotoxigenic Escherichia coli fimbrial antigens protected more than 80% of mice challenged with a lethal dose of enterotoxigenic E. coli [254]. L. lactis engineered to express rotavirus spike protein VP8 induces anti-VP8 IgA in mice [253]. There are several advantages of utilizing recombinant probiotics as a vaccine vector. Antigens delivered orally through recombinant probiotics will be resistant to degradation by proteases in the gut. Further, recombinant probiotics are easy to administer and mimic the route of infection [256]. Hence, recombinant vaccine vectors using probiotic species is a future approach with the potential to control foodborne pathogen infections in poultry.
With a more thorough understanding of the mechanism of action of postbiotics, designer postbiotics can be used for targeted prevention and control of enteric diseases in poultry. In humans, supplementation of SCFAs and tryptophan was found to have a therapeutic effect on IBD [257]. In a study conducted by Tsilingiri et al. [239], it was found that potent postbiotics can prevent the colonization of Salmonella on healthy intestinal epithelium and downregulate pro-inflammatory pathways in IBD on a polarized ex-vivo organ culture model. Although extensive research on postbiotics is undertaken for various human disease models, only a few studies have been conducted on poultry disease models. In a study conducted by Chaney et al. [258], it was demonstrated that supplementing postbiotics (Saccharomyces cerevisiae fermentation product) in broiler diet significantly reduced the cecal load and prevalence of Salmonella enterica compared to the group fed a standard diet. The large-scale production of postbiotics and associated cost, considering the low yield of metabolites from cells, is limiting their widespread commercial application in the poultry industry.

14. Prebiotics

Prebiotics are defined as “selectively fermented ingredients that result in specific changes in the composition and/or activity of the gastrointestinal microbiota, thus conferring benefits upon host health” [259]. Compounds of carbohydrates considered prebiotics include fructo-oligosaccharides (FOS), galactooligosaccharides (GOS), Mannanoligosaccharide, oligochitosan, inulin, pyrodextrins, stachyose, maltooligosaccharides, isomaltooligosaccharide (IOS), xylooligosaccharide, glucooligosaccharides, soya-oligosaccharide, lactitol, lactulose, pectin, and arabinoxylan. Non-carbohydrate compounds such as polyphenols, certain lipids, peptides, and proteins are also considered candidate prebiotics. These macromolecules are either synthesized by microorganisms or obtained from plants [90,260,261,262,263]. However, the beneficial effects of prebiotic supplementation depend on the individual components. The chemical structure (short-chain or long-chain), duration of ingestion, and dose of the prebiotic determine the effects of prebiotics on gut microbial composition [262,264]. The desirable characteristics of a prebiotic are 1. Resistance to gastric pH, hydrolysis by host enzymes, and absorption in the gut 2. Fermentable by the intestinal microflora 3. Selective stimulation of intestinal bacteria that positively contribute to host health and wellbeing 4. Enhance local and systemic immunity against pathogen invasion [82,90].
Some of the advantages of prebiotic supplementation include the reduced occurrence of enteric diseases, improved gut health, improved performance, decreased odor, and enhanced nutrient utilization [263,265]. The mechanism of action of probiotics is 1. Positive modulation of intestinal microbiota for host benefit 2. Improved gut epithelial health 3. Immune system stimulation [266]. Prebiotics prevent the colonization of the intestinal epithelium by pathogenic bacteria directly by adsorption or by competitive exclusion wherein prebiotics preferentially promote the growth of beneficial microorganisms [267]. Prebiotic supplementation significantly reduces Campylobacter spp. loads by 1.0 log10 CFU g−1 in the caeca by inhibiting the adhesion of Campylobacter mannose-binding lectins to the host enterocytes [268,269]. The metabolization of prebiotics by the gut microorganisms results in the production of lactic acid, SCFAs, or antibacterial peptides such as bacteriocins which can modulate the composition and/or activity of the gut microbiota, conferring health benefits to the host as summarized in Figure 4. The SCFA produced by the fermentation of prebiotics by bacteria acts as an energy source for the intestinal epithelial cells, thus promoting gut health and integrity [270]. Prebiotics are non-pathogenic antigens capable of stimulating the host immune system beneficially [266]. In a study conducted by Janardhana et al. [271], it was demonstrated that prebiotics affects the systemic antibody levels and proliferation of caecal tonsil immune cells. These effects are mediated by the modulation of gut microbiota by prebiotics. Prebiotics selectively enrich the microorganisms such as Lactobacillus spp., and Bifidobacteria spp. which are able to utilize the non-digestible, but fermentable substrates [262]. Further, prebiotics inhibits the attachment of pathogens such as Salmonella [272], E. coli [273], and Campylobacter [274] to the receptors on host intestinal cells by acting as decoy receptors [272], and thus in addition to contributing to better growth and performance, it also improves the microbiological quality and safety of poultry products.

15. Synbiotics

A product containing a combination of prebiotic and probiotic is termed symbiotic. The prebiotic compound in the combination selectively promotes the growth of the probiotic by providing the specific substrate for fermentation [275,276]. A summary of the concept is illustrated in Figure 5. In a study conducted by Mookiah et al. [277] on the effects of probiotics, prebiotics, and synbiotics in broiler performance parameters, significant improvement in weight gain and feed conversion rate were observed. In addition, there was a shift in caecal microbiota with an increase in the population of lactobacilli and bifidobacteria and a significant decrease in E. coli at day 21 of age. Increased caecal VFA production was also noticed in the groups fed probiotics prebiotics and synbiotics. However, the findings did not suggest the synergistic effect of probiotic and prebiotic as the synbiotic group performance parameters were not significantly different from the groups fed probiotic or prebiotic alone [277]. The results of the study conducted by our group are in agreement with the above study where we identified that the FCR and BWG were not different between the birds supplemented with Bacillus subtilis probiotic and the mannooligosaccharide symbiotic group [161]. Further, In a study conducted by Chen et al. [278], it was reported that the effects of synbiotic supplementation were comparable to that of AGPs. Synbiotic supplementation improved the feed conversion efficiency, decreased abdominal fat yield, and decreased drip loss in breast muscle in ducks. These effects are due to the modulation of intestinal microbiota, maintenance of intestinal barrier integrity, positive modulation of the immune system, and improved lipid metabolism by the components of synbiotics. There is also evidence of increased antibody production and immune response when the symbiotic fed groups are vaccinated. The positive effects of synbiotics are dose-dependent. Adverse effects such as decreased villus surface area in the jejunum and ileum were observed when the dose was doubled from 0.1% to 0.2% synbiotic supplementation [279].

16. Conclusions

The gastrointestinal microbial community plays an important role in many physiological and immunological systems. In poultry, the gastrointestinal tract is sterile at hatch, providing the poultry industry with a unique opportunity to have significant control over microbial colonization in young birds. However, due to increased variability in gut microbiota between birds based on breed, housing, environment, diet, and immune status, a clear relationship between host and microbiota is not established. Prebiotics, probiotics, postbiotics, and synbiotics have been studied in depth over recent years in response to increased limitations on the use of antimicrobials in poultry. These additives can alter chicken gut microbiota positively and thereby improving growth, performance, and immune parameters. Designer probiotics customized for specific breeds, age groups, and diseases have the potential to completely replace antibiotics in poultry. With the development of appropriate technology, gut microbiota can be routinely monitored and modulated for host benefit in near future.

Author Contributions

S.F.: Writing—original draft preparation; R.S.: writing—review, editing, and funding acquisition, D.A.: editing; R.K.S.: editing and funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by USDA-ARS grant number 6040-42000-046-000D to RS and 58-6040-8-034, awarded to RKS.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. FAO, R. Prospects for food, nutrition, agriculture and major commodity groups. World Agric. Towards 2030–2050 2006, 2030, 2050. [Google Scholar]
  2. Biggs, P.M. The world of poultry disease. Avian Pathol. 1982, 11, 281–300. [Google Scholar] [CrossRef] [PubMed]
  3. Griggs, J.P.; Jacob, J.P. Alternatives to antibiotics for organic poultry production. J. Appl. Poult. Res. 2005, 14, 750–756. [Google Scholar] [CrossRef]
  4. Yan, W.; Sun, C.; Yuan, J.; Yang, N. Gut metagenomic analysis reveals prominent roles of Lactobacillus and cecal microbiota in chicken feed efficiency. Sci. Rep. 2017, 7, 45308. [Google Scholar] [CrossRef]
  5. Barnes, E.M.; Mead, G.C.; Barnuml, D.A.; Harry, E.G. The intestinal flora of the chicken in the period 2 to 6 weeks of age, with particular reference to the anaerobic bacteria. Br. Poult. Sci. 1972, 13, 311–326. [Google Scholar] [CrossRef]
  6. Barnes, E.M. The avian intestinal flora with particular reference to the possible ecological significance of the cecal anaerobic bacteria. Am. J. Clin. Nutr. 1972, 25, 1475–1479. [Google Scholar] [CrossRef]
  7. Slizewska, K.; Piotrowska, M. Reduction of ochratoxin A in chicken feed using probiotic. Ann. Agric. Environ. Med. 2014, 21, 676–680. [Google Scholar] [CrossRef]
  8. Davies, R.H.; Wray, C. Determination of an effective sampling regime to detect Salmonella enteritidis in the environment of poultry units. Vet. Microbiol. 1996, 50, 117–127. [Google Scholar] [CrossRef]
  9. Rubio, L.A. Possibilities of early life programming in broiler chickens via intestinal microbiota modulation. Poult. Sci. 2019, 98, 695–706. [Google Scholar] [CrossRef]
  10. Baldwin, S.; Hughes, R.J.; Hao Van, T.T.; Moore, R.J.; Stanley, D. At-hatch administration of probiotic to chickens can introduce beneficial changes in gut microbiota. PLoS ONE 2018, 13, e0194825. [Google Scholar] [CrossRef]
  11. Kers, J.G.; Velkers, F.C.; Fischer, E.A.; Hermes, G.D.; Stegeman, J.A.; Smidt, H. Host and environmental factors affecting the intestinal microbiota in chickens. Front. Microbiol. 2018, 9, 235. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Broom, L.J. Gut barrier function: Effects of (antibiotic) growth promoters on key barrier components and associations with growth performance. Poult. Sci. 2018, 97, 1572–1578. [Google Scholar] [CrossRef] [PubMed]
  13. Broom, L.J.; Kogut, M.H. The role of the gut microbiome in shaping the immune system of chickens. Vet. Immunol. Immunopathol. 2018, 204, 44–51. [Google Scholar] [CrossRef] [PubMed]
  14. Befus, A.D.; Johnston, N.; Leslie, G.A.; Bienenstock, J. Gut-associated lymphoid tissue in the chicken. I. Morphology, ontogeny, and some functional characteristics of Peyer’s patches. J. Immunol. 1980, 125, 2626–2632. [Google Scholar]
  15. Lavelle, E.C.; Murphy, C.; O’Neill, L.; Creagh, E.M. The role of TLRs, NLRs, and RLRs in mucosal innate immunity and homeostasis. Mucosal Immunol. 2010, 3, 17–28. [Google Scholar] [CrossRef] [Green Version]
  16. Maki, J.J.; Bobeck, E.A.; Sylte, M.J.; Looft, T. Eggshell and environmental bacteria contribute to the intestinal microbiota of growing chickens. J. Anim. Sci. Biotechnol. 2020, 11, 1–17. [Google Scholar] [CrossRef]
  17. Donaldson, E.E.; Stanley, D.; Hughes, R.J.; Moore, R.J. The time-course of broiler intestinal microbiota development after administration of cecal contents to incubating eggs. PeerJ 2017, 5, e3587. [Google Scholar] [CrossRef] [Green Version]
  18. Lan, Y.; Verstegen, M.; Tamminga, S.; Williams, B.A. The role of the commensal gut microbial community in broiler chickens. Worlds Poult. Sci. J. 2005, 61, 95–104. [Google Scholar] [CrossRef] [Green Version]
  19. Ding, J.; Dai, R.; Yang, L.; He, C.; Xu, K.; Liu, S.; Zhao, W.; Xiao, L.; Luo, L.; Zhang, Y. Inheritance and establishment of gut microbiota in chickens. Front. Microbiol. 2017, 8, 1967. [Google Scholar] [CrossRef]
  20. Ngunjiri, J.M.; Taylor, K.J.; Abundo, M.C.; Jang, H.; Elaish, M.; Kc, M.; Ghorbani, A.; Wijeratne, S.; Weber, B.P.; Johnson, T.J. Farm stage, bird age, and body site dominantly affect the quantity, taxonomic composition, and dynamics of respiratory and gut microbiota of commercial layer chickens. Appl. Environ. Microbiol. 2019, 85, 3137. [Google Scholar] [CrossRef] [Green Version]
  21. Shang, Y.; Kumar, S.; Oakley, B.; Kim, W.K. Chicken gut microbiota: Importance and detection technology. Front. Vet. Sci. 2018, 5, 254. [Google Scholar] [CrossRef] [PubMed]
  22. Pourabedin, M.; Zhao, X. Prebiotics and gut microbiota in chickens. FEMS Microbiol. Lett. 2015, 362, fnv122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Romero, R.; Hassan, S.S.; Gajer, P.; Tarca, A.L.; Fadrosh, D.W.; Nikita, L.; Galuppi, M.; Lamont, R.F.; Chaemsaithong, P.; Miranda, J. The composition and stability of the vaginal microbiota of normal pregnant women is different from that of non-pregnant women. Microbiome 2014, 2, 1–19. [Google Scholar]
  24. Olsen, R.; Kudirkiene, E.; Thøfner, I.; Pors, S.; Karlskov-Mortensen, P.; Li, L.; Papasolomontos, S.; Angastiniotou, C.; Christensen, J. Impact of egg disinfection of hatching eggs on the eggshell microbiome and bacterial load. Poult. Sci. 2017, 96, 3901–3911. [Google Scholar] [CrossRef]
  25. Rychlik, I. Composition and function of chicken gut microbiota. Animals 2020, 10, 103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Gong, J.; Si, W.; Forster, R.J.; Huang, R.; Yu, H.; Yin, Y.; Yang, C.; Han, Y. 16S rRNA gene-based analysis of mucosa-associated bacterial community and phylogeny in the chicken gastrointestinal tracts: From crops to ceca. FEMS Microbiol. Ecol. 2007, 59, 147–157. [Google Scholar] [CrossRef] [Green Version]
  27. Rehman, H.U.; Vahjen, W.; Awad, W.A.; Zentek, J. Indigenous bacteria and bacterial metabolic products in the gastrointestinal tract of broiler chickens. Arch. Anim. Nutr. 2007, 61, 319–335. [Google Scholar] [CrossRef]
  28. Hinton, A., Jr.; Buhr, R.J.; Ingram, K.D. Physical, chemical, and microbiological changes in the crop of broiler chickens subjected to incremental feed withdrawal. Poult. Sci. 2000, 79, 212–218. [Google Scholar] [CrossRef]
  29. Rehman, H.; Böhm, J.; Zentek, J. Effects of diets with inulin and sucrose on the microbial fermentation in the gastrointestinal tract of broilers. Proc. Soc. Nutr. Physiol. 2006, 15, 155–158. [Google Scholar]
  30. Christl, S.U.; Bartram, P.; Paul, A.; Kelber, E.; Scheppach, W.; Kasper, H. Bile acid metabolism by colonic bacteria in continuous culture: Effects of starch and pH. Ann. Nutr. Metab. 1997, 41, 45–51. [Google Scholar] [CrossRef]
  31. Wang, J.; Fan, H.; Han, Y.; Wei, J.; Zhao, J.; Zhou, Z. Pyrosequencing of the broiler chicken gastrointestinal tract reveals the regional similarity and dissimilarity of microbial community. Can. J. Anim. Sci. 2016, 97, 302–313. [Google Scholar] [CrossRef] [Green Version]
  32. Alakomi, H.; Skytta, E.; Saarela, M.; Mattila-Sandholm, T.; Latva-Kala, K.; Helander, I.M. Lactic acid permeabilizes gram-negative bacteria by disrupting the outer membrane. Appl. Environ. Microbiol. 2000, 66, 2001–2005. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Narendranath, N.V.; Thomas, K.C.; Ingledew, W.M. Acetic acid and lactic acid inhibition of growth of Saccharomyces cerevisiae by different mechanisms. J. Am. Soc. Brew. Chem. 2001, 59, 187–194. [Google Scholar]
  34. Zhang, W.F.; Li, D.F.; Lu, W.Q.; Yi, G.F. Effects of isomalto-oligosaccharides on broiler performance and intestinal microflora. Poult. Sci. 2003, 82, 657–663. [Google Scholar] [CrossRef]
  35. Gabriel, I.; Lessire, M.; Mallet, S.; Guillot, J.F. Microflora of the digestive tract: Critical factors and consequences for poultry. Worlds Poult. Sci. J. 2006, 62, 499–511. [Google Scholar]
  36. Flickinger, E.A.; Loo, J.V.; Fahey, G.C. Nutritional responses to the presence of inulin and oligofructose in the diets of domesticated animals: A review. Crit. Rev. Food Sci. Nutr. 2003, 43, 19–60. [Google Scholar] [CrossRef]
  37. Barnes, E.M.; Impey, C.S.; Stevens, B. Factors affecting the incidence and anti-salmonella activity of the anaerobic caecal flora of the young chick. Epidemiol. Infect. 1979, 82, 263–283. [Google Scholar] [CrossRef] [Green Version]
  38. Walugembe, M.; Hsieh, J.C.; Koszewski, N.J.; Lamont, S.J.; Persia, M.E.; Rothschild, M.F. Effects of dietary fiber on cecal short-chain fatty acid and cecal microbiota of broiler and laying-hen chicks. Poult. Sci. 2015, 94, 2351–2359. [Google Scholar] [CrossRef]
  39. Czerwiński, J.; Højberg, O.; Smulikowska, S.; Engberg, R.M.; Mieczkowska, A. Influence of dietary peas and organic acids and probiotic supplementation on performance and caecal microbial ecology of broiler chickens. Br. Poult. Sci. 2010, 51, 258–269. [Google Scholar] [CrossRef]
  40. Van der Wielen Paul, W.J.J.; Biesterveld, S.; Notermans, S.; Hofstra, H.; Urlings, B.A.; van Knapen, F. Role of volatile fatty acids in development of the cecal microflora in broiler chickens during growth. Appl. Environ. Microbiol. 2000, 66, 2536–2540. [Google Scholar] [CrossRef] [Green Version]
  41. Yurong, Y.; Ruiping, S.; ShiMin, Z.; Yibao, J. Effect of probiotics on intestinal mucosal immunity and ultrastructure of cecal tonsils of chickens. Arch. Anim. Nutr. 2005, 59, 237–246. [Google Scholar] [CrossRef] [PubMed]
  42. Kogut, M.H.; Arsenault, R.J. Immunometabolic phenotype alterations associated with the induction of disease tolerance and persistent asymptomatic infection of Salmonella in the chicken intestine. Front. Immunol. 2017, 8, 372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Mukherjee, S.; Vaishnava, S.; Hooper, L.V. Multi-layered regulation of intestinal antimicrobial defense. Cell. Mol. Life Sci. 2008, 65, 3019–3027. [Google Scholar] [CrossRef]
  44. Zhang, M.; Wu, C. The relationship between intestinal goblet cells and the immune response. Biosci. Rep. 2020, 40, BSR20201471. [Google Scholar] [CrossRef] [PubMed]
  45. Becker, S.; Oelschlaeger, T.A.; Wullaert, A.; Pasparakis, M.; Wehkamp, J.; Stange, E.F.; Gersemann, M. Bacteria regulate intestinal epithelial cell differentiation factors both in vitro and in vivo. PLoS ONE 2013, 8, e55620. [Google Scholar] [CrossRef]
  46. Tellez, G.; Higgins, S.E.; Donoghue, A.M.; Hargis, B.M. Digestive physiology and the role of microorganisms. J. Appl. Poult. Res. 2006, 15, 136–144. [Google Scholar] [CrossRef]
  47. Chan, Y.K.; Estaki, M.; Gibson, D.L. Clinical consequences of diet-induced dysbiosis. Ann. Nutr. Metab. 2013, 63, 28–40. [Google Scholar] [CrossRef]
  48. Wells, J.M.; Loonen, L.M.; Karczewski, J.M. The role of innate signaling in the homeostasis of tolerance and immunity in the intestine. Int. J. Med. Microbiol. 2010, 300, 41–48. [Google Scholar] [CrossRef]
  49. Kawai, T.; Akira, S. TLR signaling. Cell Death Differ. 2006, 13, 816–825. [Google Scholar] [CrossRef] [Green Version]
  50. Turner, J.R. Intestinal mucosal barrier function in health and disease. Nat. Rev. Immunol. 2009, 9, 799–809. [Google Scholar] [CrossRef]
  51. Mancabelli, L.; Ferrario, C.; Milani, C.; Mangifesta, M.; Turroni, F.; Duranti, S.; Lugli, G.A.; Viappiani, A.; Ossiprandi, M.C.; van Sinderen, D. Insights into the biodiversity of the gut microbiota of broiler chickens. Environ. Microbiol. 2016, 18, 4727–4738. [Google Scholar] [CrossRef] [PubMed]
  52. Chassard, C.; Lacroix, C. Carbohydrates and the human gut microbiota. Curr. Opin. Clin. Nutr. Metab. Care 2013, 16, 453–460. [Google Scholar] [CrossRef] [PubMed]
  53. Brisbin, J.T.; Zhou, H.; Gong, J.; Sabour, P.; Akbari, M.R.; Haghighi, H.R.; Yu, H.; Clarke, A.; Sarson, A.J.; Sharif, S. Gene expression profiling of chicken lymphoid cells after treatment with Lactobacillus acidophilus cellular components. Dev. Comp. Immunol. 2008, 32, 563–574. [Google Scholar] [CrossRef] [PubMed]
  54. Hooper, L.V.; Macpherson, A.J. Immune adaptations that maintain homeostasis with the intestinal microbiota. Nat. Rev. Immunol. 2010, 10, 159–169. [Google Scholar] [CrossRef] [PubMed]
  55. Langhout, D.J.; Schutte, J.B.; Van Leeuwen, P.; Wiebenga, J.; Tamminga, S. Effect of dietary high-and low-methylated citrus pectin on the activity of the ileal microflora and morphology of the small intestinal wall of broiler chicks. Br. Poult. Sci. 1999, 40, 340–347. [Google Scholar] [CrossRef] [PubMed]
  56. Teirlynck, E.; Bjerrum, L.; Eeckhaut, V.; Huygebaert, G.; Pasmans, F.; Haesebrouck, F.; Dewulf, J.; Ducatelle, R.; Van Immerseel, F. The cereal type in feed influences gut wall morphology and intestinal immune cell infiltration in broiler chickens. Br. J. Nutr. 2009, 102, 1453–1461. [Google Scholar] [CrossRef]
  57. Teirlynck, E.; Gussem, M.; Dewulf, J.; Haesebrouck, F.; Ducatelle, R.; Van Immerseel, F. Morphometric evaluation of “dysbacteriosis” in broilers. Avian Pathol. 2011, 40, 139–144. [Google Scholar] [CrossRef] [Green Version]
  58. Teirlynck, E.; De Gussem, M.; Marien, M.; Vancraeynest, D.; Haesebrouck, F.; Ducatelle, R.; Van Immerseel, F. Intestinal morphometry in ‘dysbacteriosis’ in broilers. Avian Pathol. 2011, 40, 139–144. [Google Scholar]
  59. Teirlynck, E.; Haesebrouck, F.; Pasmans, F.; Dewulf, J.; Ducatelle, R.; Van Immerseel, F. The cereal type in feed influences Salmonella Enteritidis colonization in broilers. Poult. Sci. 2009, 88, 2108–2112. [Google Scholar] [CrossRef]
  60. Tsiouris, V.; Georgopoulou, I.; Batzios, C.; Pappaioannou, N.; Ducatelle, R.; Fortomaris, P. High stocking density as a predisposing factor for necrotic enteritis in broiler chicks. Avian Pathol. 2015, 44, 59–66. [Google Scholar] [CrossRef] [Green Version]
  61. Estevez, I. Density allowances for broilers: Where to set the limits? Poult. Sci. 2007, 86, 1265–1272. [Google Scholar] [CrossRef] [PubMed]
  62. Stanley, D.; Geier, M.S.; Denman, S.E.; Haring, V.R.; Crowley, T.M.; Hughes, R.J.; Moore, R.J. Identification of chicken intestinal microbiota correlated with the efficiency of energy extraction from feed. Vet. Microbiol. 2013, 164, 85–92. [Google Scholar] [CrossRef] [PubMed]
  63. Adhikari, P.; Kiess, A.; Adhikari, R.; Jha, R. An approach to alternative strategies to control avian coccidiosis and necrotic enteritis. J. Appl. Poult. Res. 2020, 29, 515–534. [Google Scholar] [CrossRef]
  64. Branton, S.L.; Lott, B.D.; Deaton, J.W.; Maslin, W.R.; Austin, F.W.; Pote, L.M.; Keirs, R.W.; Latour, M.A.; Day, E.J. The effect of added complex carbohydrates or added dietary fiber on necrotic enteritis lesions in broiler chickens. Poult. Sci. 1997, 76, 24–28. [Google Scholar] [CrossRef]
  65. Józefiak, D.; Rutkowski, A.; Fratczak, M.; Boros, D. The effect of dietary fibre fractions from different cereals and microbial enzymes supplementation on performance, ileal viscosity and short-chain fatty acids concentration in caeca of broiler chickens. J. Anim. Feed. Sci. 2004, 13, 487–496. [Google Scholar] [CrossRef] [Green Version]
  66. Yaghobfar, A.; Kalantar, M. Effect of non-starch polysaccharide (NSP) of wheat and barley supplemented with exogenous enzyme blend on growth performance, gut microbial, pancreatic enzyme activities, expression of glucose transporter (SGLT1) and mucin producer (MUC2) genes of broiler chickens. Braz. J. Poult. Sci. 2017, 19, 629–638. [Google Scholar]
  67. Choct, M.; Hughes, R.J.; Wang, J.; Bedford, M.R.; Morgan, A.J.; Annison, G. Increased small intestinal fermentation is partly responsible for the anti-nutritive activity of non-starch polysaccharides in chickens. Br. Poult. Sci. 1996, 37, 609–621. [Google Scholar] [CrossRef]
  68. Hashemipour, H.; Khaksar, V.; Rubio, L.A.; Veldkamp, T.; Van Krimpen, M.M. Effect of feed supplementation with a thymol plus carvacrol mixture, in combination or not with an NSP-degrading enzyme, on productive and physiological parameters of broilers fed on wheat-based diets. Anim. Feed Sci. Technol. 2016, 211, 117–131. [Google Scholar] [CrossRef]
  69. Collier, C.T.; Van der Klis, J.D.; Deplancke, B.; Anderson, D.B.; Gaskins, H.R. Effects of tylosin on bacterial mucolysis, Clostridium perfringens colonization, and intestinal barrier function in a chick model of necrotic enteritis. Antimicrob. Agents Chemother. 2003, 47, 3311–3317. [Google Scholar] [CrossRef] [Green Version]
  70. Ma, N.; Tian, Y.; Wu, Y.; Ma, X. Contributions of the interaction between dietary protein and gut microbiota to intestinal health. Curr. Protein Pept. Sci. 2017, 18, 795–808. [Google Scholar] [CrossRef]
  71. Liu, N.; Wang, J.Q.; Gu, K.T.; Deng, Q.Q.; Wang, J.P. Effects of dietary protein levels and multienzyme supplementation on growth performance and markers of gut health of broilers fed a miscellaneous meal based diet. Anim. Feed Sci. Technol. 2017, 234, 110–117. [Google Scholar] [CrossRef]
  72. Shojadoost, B.; Vince, A.R.; Prescott, J.F. The successful experimental induction of necrotic enteritis in chickens by Clostridium perfringens: A critical review. Vet. Res. 2012, 43, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Chimerel, C.; Murray, A.J.; Oldewurtel, E.R.; Summers, D.K.; Keyser, U.F. The effect of bacterial signal indole on the electrical properties of lipid membranes. ChemPhysChem 2013, 14, 417–423. [Google Scholar] [CrossRef] [Green Version]
  74. Hoover, J. Mechanistic Understanding of Leaky Gut Syndrome in Heat Stressed Broiler Chickens. Bachelor’s Thesis, University of Arkansas, Fayetteville, AR, USA, 2020. [Google Scholar]
  75. Ruff, J.; Barros, T.L.; Tellez Jr, G.; Blankenship, J.; Lester, H.; Graham, B.D.; Selby, C.A.; Vuong, C.N.; Dridi, S.; Greene, E.S. Research Note: Evaluation of a heat stress model to induce gastrointestinal leakage in broiler chickens. Poult. Sci. 2020, 99, 1687–1692. [Google Scholar] [CrossRef] [PubMed]
  76. Park, S.; Hwangbo, J.; Ryu, C.; Park, B.; Chae, H.; Choi, H.; Kang, H.; Seo, O.; Choi, Y. Effects of Extreme Heat Stress on Growth Performance, Lymphoid Organ, IgG and Cecum Microflora of Broiler Chickens. Int. J. Agric. Biol. 2013, 15, 1204–1208. [Google Scholar]
  77. Jewers, K. Mycotoxins and their effect on poultry production. Options Méditerr. 1990, 7, 195–202. [Google Scholar]
  78. Sweeney, M.J.; Dobson, A.D. Mycotoxin production by Aspergillus, Fusarium and Penicillium species. Int. J. Food Microbiol. 1998, 43, 141–158. [Google Scholar] [CrossRef]
  79. Alassane-Kpembi, I.; Pinton, P.; Oswald, I.P. Effects of Mycotoxins on the Intestine. Toxins 2019, 11, 159. [Google Scholar] [CrossRef] [Green Version]
  80. Guerre, P. Mycotoxin and gut microbiota interactions. Toxins 2020, 12, 769. [Google Scholar] [CrossRef]
  81. Ren, Z.; Guo, C.; Yu, S.; Zhu, L.; Wang, Y.; Hu, H.; Deng, J. Progress in mycotoxins affecting intestinal mucosal barrier function. Int. J. Mol. Sci. 2019, 20, 2777. [Google Scholar] [CrossRef] [Green Version]
  82. Gibson, G.R.; Probert, H.M.; Van Loo, J.; Rastall, R.A.; Roberfroid, M.B. Dietary modulation of the human colonic microbiota: Updating the concept of prebiotics. Nutr. Res. Rev. 2004, 17, 259–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Elwinger, K.; Berndtson, E.; Engström, B.; Fossum, O.; Waldenstedt, L. Effect of antibiotic growth promoters and anticoccidials on growth of Clostridium perfringens in the caeca and on performance of broiler chickens. Acta Vet. Scand. 1998, 39, 433–441. [Google Scholar] [CrossRef] [PubMed]
  84. Alagawany, M.; Abd El-Hack, M.E.; Farag, M.R.; Sachan, S.; Karthik, K.; Dhama, K. The use of probiotics as eco-friendly alternatives for antibiotics in poultry nutrition. Environ. Sci. Pollut. Res. 2018, 25, 10611–10618. [Google Scholar] [CrossRef] [PubMed]
  85. Macdonald, S.E.; Nolan, M.J.; Harman, K.; Boulton, K.; Hume, D.A.; Tomley, F.M.; Stabler, R.A.; Blake, D.P. Effects of Eimeria tenella infection on chicken caecal microbiome diversity, exploring variation associated with severity of pathology. PLoS ONE 2017, 12, e0184890. [Google Scholar] [CrossRef] [PubMed]
  86. Vila, B.; Esteve-Garcia, E.; Brufau, J. Probiotic micro-organisms: 100 years of innovation and efficacy; modes of action. Worlds Poult. Sci. J. 2010, 66, 369–380. [Google Scholar] [CrossRef] [Green Version]
  87. Joint FAO/WHO Working Group. Guidelines for the Evaluation of Probiotics in Food. In Food and Agriculture Organization; World Health Organization: London, UK, 2002. [Google Scholar]
  88. Fidanza, M.; Panigrahi, P.; Kollmann, T.R. Lactiplantibacillus plantarum–Nomad and Ideal Probiotic. Front. Microbiol. 2021, 12, 2911. [Google Scholar] [CrossRef] [PubMed]
  89. Ma, T.; Suzuki, Y. Dissect the mode of action of probiotics in affecting host-microbial interactions and immunity in food producing animals. Vet. Immunol. Immunopathol. 2018, 205, 35–48. [Google Scholar] [CrossRef]
  90. Patterson, J.A.; Burkholder, K.M. Application of prebiotics and probiotics in poultry production. Poult. Sci. 2003, 82, 627–631. [Google Scholar] [CrossRef]
  91. Dowarah, R.; Verma, A.K.; Agarwal, N.; Singh, P.; Singh, B.R. Selection and characterization of probiotic lactic acid bacteria and its impact on growth, nutrient digestibility, health and antioxidant status in weaned piglets. PLoS ONE 2018, 13, e0192978. [Google Scholar] [CrossRef]
  92. Ehrmann, M.A.; Kurzak, P.; Bauer, J.; Vogel, R.F. Characterization of lactobacilli towards their use as probiotic adjuncts in poultry. J. Appl. Microbiol. 2002, 92, 966–975. [Google Scholar] [CrossRef]
  93. Reuben, R.C.; Roy, P.C.; Sarkar, S.L.; Alam, R.; Jahid, I.K. Isolation, characterization, and assessment of lactic acid bacteria toward their selection as poultry probiotics. BMC Microbiol. 2019, 19, 253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. European Food Safety Authority and European Centre for Disease Prevention and Control (EFSA and ECDC). The European Union summary report on trends and sources of zoonoses, zoonotic agents and food-borne outbreaks in 2017. EFSa J. 2018, 16, e05077. [Google Scholar]
  95. Hesari, M.R.; Darsanaki, R.K.; Salehzadeh, A. Antagonistic activity of probiotic bacteria isolated from traditional dairy products against E. coli O157: H7. J. Med. Bacteriol. 2017, 6, 23–30. [Google Scholar]
  96. Liao, S.F.; Nyachoti, M. Using probiotics to improve swine gut health and nutrient utilization. Anim. Nutr. 2017, 3, 331–343. [Google Scholar] [CrossRef] [PubMed]
  97. Garriga, M.; Pascual, M.; Monfort, J.M.; Hugas, M. Selection of lactobacilli for chicken probiotic adjuncts. J. Appl. Microbiol. 1998, 84, 125–132. [Google Scholar] [CrossRef]
  98. Farner, D.S. The hydrogen ion concentration in avian digestive tracts. Poult. Sci. 1942, 21, 445–450. [Google Scholar] [CrossRef]
  99. Pan, X.; Chen, F.; Wu, T.; Tang, H.; Zhao, Z. The acid, bile tolerance and antimicrobial property of Lactobacillus acidophilus NIT. Food Control 2009, 20, 598–602. [Google Scholar] [CrossRef]
  100. Nallala, V.; Sadishkumar, V.; Jeevaratnam, K. Molecular characterization of antimicrobial Lactobacillus isolates and evaluation of their probiotic characteristics in vitro for use in poultry. Food Biotechnol. 2017, 31, 20–41. [Google Scholar] [CrossRef]
  101. Lin, J.; Sahin, O.; Michel, L.O.; Zhang, Q. Critical role of multidrug efflux pump CmeABC in bile resistance and in vivo colonization of Campylobacter jejuni. Infect. Immun. 2003, 71, 4250–4259. [Google Scholar] [CrossRef] [Green Version]
  102. Taheri, H.R.; Moravej, H.; Tabandeh, F.; Zaghari, M.; Shivazad, M. Screening of lactic acid bacteria toward their selection as a source of chicken probiotic. Poult. Sci. 2009, 88, 1586–1593. [Google Scholar] [CrossRef]
  103. Collado, M.C.; Meriluoto, J.; Salminen, S. Adhesion and aggregation properties of probiotic and pathogen strains. Eur. Food Res. Technol. 2008, 226, 1065–1073. [Google Scholar] [CrossRef]
  104. Dec, M.; Urban-Chmiel, R.; Stępień-Pyśniak, D.; Wernicki, A. Assessment of antibiotic susceptibility in Lactobacillus isolates from chickens. Gut Pathog. 2017, 9, 1–16. [Google Scholar] [CrossRef] [PubMed]
  105. Ramlucken, U.; Ramchuran, S.O.; Moonsamy, G.; van Rensburg, C.J.; Thantsha, M.S.; Lalloo, R. Production and stability of a multi-strain Bacillus based probiotic product for commercial use in poultry. Biotechnol. Rep. 2021, 29, e00575. [Google Scholar] [CrossRef] [PubMed]
  106. Mountzouris, K.C.; Tsitrsikos, P.; Palamidi, I.; Arvaniti, A.; Mohnl, M.; Schatzmayr, G.; Fegeros, K. Effects of probiotic inclusion levels in broiler nutrition on growth performance, nutrient digestibility, plasma immunoglobulins, and cecal microflora composition. Poult. Sci. 2010, 89, 58–67. [Google Scholar] [CrossRef]
  107. Tripathi, M.K.; Giri, S.K. Probiotic functional foods: Survival of probiotics during processing and storage. J. Funct. Foods 2014, 9, 225–241. [Google Scholar] [CrossRef]
  108. Lee, Y.K.; Salminen, S. Handbook of Probiotics and Prebiotics; John Wiley & Sons: Hoboken, NJ, USA, 2009. [Google Scholar]
  109. Teixeira, P.C.; Castro, M.H.; Malcata, F.X.; Kirby, R.M. Survival of Lactobacillus delbrueckii ssp. bulgaricus following spray-drying. J. Dairy Sci. 1995, 78, 1025–1031. [Google Scholar] [CrossRef]
  110. Tamime, A.Y.; Saarela, M.; Sondergaard, A.K.; Mistry, V.V.; Shah, N.P. Production and maintenance of viability of probiotic microorganisms in dairy products. Probiotic Dairy Prod. 2005, 3, 39–63. [Google Scholar]
  111. Teixeira, P.; Castro, H.; Kirby, R. Inducible thermotolerance in Lactobacillus bulgaricus. Lett. Appl. Microbiol. 1994, 18, 218–221. [Google Scholar] [CrossRef]
  112. Park, Y.H.; Hamidon, F.; Rajangan, C.; Soh, K.P.; Gan, C.Y.; Lim, T.S.; Abdullah, W.N.W.; Liong, M.T. Application of probiotics for the production of safe and high-quality poultry meat. Korean J. Food Sci. Anim. Resour. 2016, 36, 567. [Google Scholar] [CrossRef] [Green Version]
  113. Thomke, S.; Elwinger, K. Growth Promotants in Feeding Pigs and Poultry. III. Alternatives to Antibiotic Growth Promotants. Ann. Zootech. INRA/EDP Sci. 1998, 47, 245–271. Available online: https://hal.archives-ouvertes.fr/hal-00889716 (accessed on 1 March 2022).
  114. Gadde, U.; Kim, W.H.; Oh, S.T.; Lillehoj, H.S. Alternatives to antibiotics for maximizing growth performance and feed efficiency in poultry: A review. Anim. Health Res. Rev. 2017, 18, 26–45. [Google Scholar] [CrossRef]
  115. Huyghebaert, G.; Ducatelle, R.; Van Immerseel, F. An update on alternatives to antimicrobial growth promoters for broilers. Vet. J. 2011, 187, 182–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Gauthier, R. Defining the Alternatives. Canadian Poultry, 10 January 2008. [Google Scholar]
  117. Huyghebaert, G. Alternatives for Antibiotic in Poultry; CONFERENCE SPONSORS: Timonium, MD, USA, 2005; p. 38. [Google Scholar]
  118. Jeurissen, S.H.; Lewis, F.; van der Klis, J.D.; Mroz, Z.; Rebel, J.M.; Ter Huurne, A.A. Parameters and techniques to determine intestinal health of poultry as constituted by immunity, integrity, and functionality. Curr. Issues Intest. Microbiol. 2002, 3, 1–14. [Google Scholar] [PubMed]
  119. Figueroa-González, I.; Quijano, G.; Ramirez, G.; Cruz-Guerrero, A. Probiotics and prebiotics—Perspectives and challenges. J. Sci. Food Agric. 2011, 91, 1341–1348. [Google Scholar] [CrossRef] [PubMed]
  120. Markazi, A.; Luoma, A.; Shanmugasundaram, R.; Mohnl, M.; Murugesan, G.R.; Selvaraj, R. Effects of drinking water synbiotic supplementation in laying hens challenged with Salmonella. Poult. Sci. 2018, 97, 3510–3518. [Google Scholar] [CrossRef] [PubMed]
  121. Kabir, S.M. The role of probiotics in the poultry industry. Int. J. Mol. Sci. 2009, 10, 3531–3546. [Google Scholar] [CrossRef]
  122. Wei, S.; Morrison, M.; Yu, Z. Bacterial census of poultry intestinal microbiome. Poult. Sci. 2013, 92, 671–683. [Google Scholar] [CrossRef]
  123. Suez, J.; Zmora, N.; Zilberman-Schapira, G.; Mor, U.; Dori-Bachash, M.; Bashiardes, S.; Zur, M.; Regev-Lehavi, D.; Brik, R.B.; Federici, S. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell 2018, 174, 1406–1423.e16. [Google Scholar] [CrossRef] [Green Version]
  124. Gupta, S.; Allen-Vercoe, E.; Petrof, E.O. Fecal Microbiota Transplantation: In perspective. Ther. Adv. Gastroenterol. 2016, 9, 229–239. [Google Scholar] [CrossRef] [Green Version]
  125. Seekatz, A.M.; Aas, J.; Gessert, C.E.; Rubin, T.A.; Saman, D.M.; Bakken, J.S.; Young, V.B. Recovery of the gut microbiome following fecal microbiota transplantation. mBio 2014, 5, 893. [Google Scholar] [CrossRef] [Green Version]
  126. Nurmi, E.; Rantala, M. New aspects of Salmonella infection in broiler production. Nature 1973, 241, 210–211. [Google Scholar] [CrossRef] [PubMed]
  127. Stern, N.J.; Cox, N.A.; Bailey, J.S.; Berrang, M.E.; Musgrove, M.T. Comparison of mucosal competitive exclusion and competitive exclusion treatment to reduce Salmonella and Campylobacter spp. colonization in broiler chickens. Poult. Sci. 2001, 80, 156–160. [Google Scholar] [CrossRef] [PubMed]
  128. Lee, Y.K.; Mazmanian, S.K. Has the microbiota played a critical role in the evolution of the adaptive immune system? Science 2010, 330, 1768–1773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Blajman, J.E.; Frizzo, L.S.; Zbrun, M.V.; Astesana, D.M.; Fusari, M.L.; Soto, L.P.; Rosmini, M.R.; Signorini, M.L. Probiotics and broiler growth performance: A meta-analysis of randomised controlled trials. Br. Poult. Sci. 2014, 55, 483–494. [Google Scholar] [CrossRef] [PubMed]
  130. Edens, F.W. An alternative for antibiotic se in poultry: Probiotics. Braz. J. Poult. Sci. 2003, 5, 75–97. [Google Scholar] [CrossRef]
  131. Olnood, C.G.; Beski, S.S.; Iji, P.A.; Choct, M. Delivery routes for probiotics: Effects on broiler performance, intestinal morphology and gut microflora. Anim. Nutr. 2015, 1, 192–202. [Google Scholar] [CrossRef]
  132. Karimi Torshizi, M.A.; Moghaddam, A.R.; Rahimi, S.H.; Mojgani, N. Assessing the effect of administering probiotics in water or as a feed supplement on broiler performance and immune response. Br. Poult. Sci. 2010, 51, 178–184. [Google Scholar] [CrossRef]
  133. Eckert, N.H.; Lee, J.T.; Hyatt, D.; Stevens, S.M.; Anderson, S.; Anderson, P.N.; Beltran, R.; Schatzmayr, G.; Mohnl, M.; Caldwell, D.J. Influence of probiotic administration by feed or water on growth parameters of broilers reared on medicated and nonmedicated diets. J. Appl. Poult. Res. 2010, 19, 59–67. [Google Scholar] [CrossRef]
  134. Hogg, S. Essential Microbiology; John Wiley & Sons: West Sussex, UK; Hoboken, NJ, USA, 2013. [Google Scholar]
  135. Higgins, J.P.; Higgins, S.E.; Vicente, J.L.; Wolfenden, A.D.; Tellez, G.; Hargis, B. Temporal effects of lactic acid bacteria probiotic culture on Salmonella in neonatal broilers. Poult. Sci. 2007, 86, 1662–1666. [Google Scholar] [CrossRef]
  136. De Oliveira, J.E.; Van der Hoeven-Hangoor, E.; Van de Linde, I.B.; Montijn, R.C.; Van der Vossen, J. In ovo inoculation of chicken embryos with probiotic bacteria and its effect on posthatch Salmonella susceptibility. Poult. Sci. 2014, 93, 818–829. [Google Scholar] [CrossRef]
  137. Roto, S.M.; Kwon, Y.M.; Ricke, S.C. Applications of in ovo technique for the optimal development of the gastrointestinal tract and the potential influence on the establishment of its microbiome in poultry. Front. Vet. Sci. 2016, 3, 63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Ferket, P.R. Embryo Epigenomic Response to Breeder Management and Nutrition; World’s Poultry Congress (Abstr.); Semantic Scholar: Seattle, WA, USA, 2012. [Google Scholar]
  139. Gulewicz, K.; Bednarczyk, M. Method for Stimulating Favourable Bacteria Profile in Hatched Chicks. Sposób Stymulacji Korzystnego Profilu Bakteryjnego Wylężonych Piskląt. Poland Application PL20030364037, 12 December 2003. [Google Scholar]
  140. Uni, Z.; Ferket, P. Enhancement of Development of Oviparous Species by in OVO Feeding. U.S. Patent 6,592,878 B2, 15 July 2003. [Google Scholar]
  141. Sławińska, A.; Siwek, M.; Żylińska, J.; Bardowski, J.; Brzezińska, J.; Gulewicz, K.A.; Nowak, M.; Urbanowski, M.; Płowiec, A.; Bednarczyk, M. Influence of synbiotics delivered in ovo on immune organs development and structure. Folia Biol. 2014, 62, 277–285. [Google Scholar] [CrossRef] [PubMed]
  142. Bednarczyk, M.; Stadnicka, K.; Kozłowska, I.; Abiuso, C.; Tavaniello, S.; Dankowiakowska, A.; Sławińska, A.; Maiorano, G. Influence of different prebiotics and mode of their administration on broiler chicken performance. Animal 2016, 10, 1271–1279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Li, T.; Castañeda, C.D.; Miotto, J.; McDaniel, C.; Kiess, A.S.; Zhang, L. Effects of in ovo probiotic administration on the incidence of avian pathogenic Escherichia coli in broilers and an evaluation on its virulence and antimicrobial resistance properties. Poult. Sci. 2021, 100, 100903. [Google Scholar] [CrossRef]
  144. Siwek, M.; Slawinska, A.; Stadnicka, K.; Bogucka, J.; Dunislawska, A.; Bednarczyk, M. Prebiotics and synbiotics–in ovo delivery for improved lifespan condition in chicken. BMC Vet. Res. 2018, 14, 1–17. [Google Scholar] [CrossRef]
  145. Villaluenga, C.M.; Wardeńska, M.; Pilarski, R.; Bednarczyk, M.; Gulewicz, K. Utilization of the chicken embryo model for assessment of biological activity of different oligosaccharides. Folia Biol. 2004, 52, 135–142. [Google Scholar] [CrossRef] [Green Version]
  146. Blankenship, L.C.; Bailey, J.S.; Cox, N.A.; Stern, N.J.; Brewer, R.; Williams, O. Two-step mucosal competitive exclusion flora treatment to diminish salmonellae in commercial broiler chickens. Poult. Sci. 1993, 72, 1667–1672. [Google Scholar] [CrossRef]
  147. Wolfenden, A.D.; Pixley, C.M.; Higgins, J.P.; Higgins, S.E.; Vicente, J.; Torres-Rodriguez, A.; Hargis, B.M.; Tellez, G. Evaluation of spray application of a Lactobacillus-based probiotic on Salmonella enteritidis colonization in broiler chickens. Int. J. Poult. Sci. 2007, 6, 493–496. [Google Scholar] [CrossRef] [Green Version]
  148. Cox, N.A.; Bailey, J.S.; Blankenship, L.C. Alternative administration of competitive exclusion treatment. In Colonization Control of Human Bacterial Enteropathologens in Poultry; Academic Press: Cambridge, MA, USA, 1991; pp. 105–118. [Google Scholar]
  149. Dankowiakowska, A.; Kozłowska, I.; Bednarczyk, M. Probiotics, prebiotics and snybiotics in Poultry–mode of action, limitation, and achievements. J. Cent. Eur. Agric. 2013, 14, 467–478. [Google Scholar] [CrossRef]
  150. Vandenbergh, P.A. Lactic acid bacteria, their metabolic products and interference with microbial growth. FEMS Microbiol. Rev. 1993, 12, 221–237. [Google Scholar] [CrossRef]
  151. Immerseel, F.V.; Buck, J.D.; Smet, I.D.; Pasmans, F.; Haesebrouck, F.; Ducatelle, R. Interactions of butyric acid–and acetic acid–treated Salmonella with chicken primary Cecal epithelial cells in vitro. Avian Dis. 2004, 48, 384–391. [Google Scholar] [CrossRef] [PubMed]
  152. AL-Allaf, M.A.; Al-Rawi, A.M.; Al-Mola, A.T. Inhibitory effect of lactic acid bacteria isolated from minced beef meat on some pathogenic bacteria. Tikrit J. Pure Sci. 2011, 16, 17–20. [Google Scholar]
  153. Mortada, M. Characterizing the Immune Response of Broilers to Campylobacter jejuni and Evaluating the Efficacy of Probiotics and Organic Acids for Reducing Campylobacter coli Colonization. Ph.D. Dissertation, University of Georgia, Athens, GA, USA, 2020. [Google Scholar]
  154. Shanmugasundaram, R.; Applegate, T.J.; Selvaraj, R.K. Effect of Bacillus subtilis and Bacillus licheniformis probiotic supplementation on cecal Salmonella load in broilers challenged with salmonella. J. Appl. Poult. Res. 2020, 29, 808–816. [Google Scholar] [CrossRef]
  155. Šikić Pogačar, M.; Langerholc, T.; Mičetić-Turk, D.; Možina, S.S.; Klančnik, A. Effect of Lactobacillus spp. on adhesion, invasion, and translocation of Campylobacter jejuni in chicken and pig small-intestinal epithelial cell lines. BMC Vet. Res. 2020, 16, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Ouwehand, A.C.; Kirjavainen, P.V.; Shortt, C.; Salminen, S. Probiotics: Mechanisms and established effects. Int. Dairy J. 1999, 9, 43–52. [Google Scholar] [CrossRef]
  157. Yamauchi, K.; Snel, J. Transmission electron microscopic demonstration of phagocytosis and intracellular processing of segmented filamentous bacteria by intestinal epithelial cells of the chick ileum. Infect. Immun. 2000, 68, 6496–6504. [Google Scholar] [CrossRef]
  158. Chichlowski, M.; Croom, J.; McBride, B.W.; Havenstein, G.B.; Koci, M.D. Metabolic and physiological impact of probiotics or direct-fed-microbials on poultry: A brief review of current knowledge. Int. J. Poult. Sci. 2007, 6, 694–704. [Google Scholar] [CrossRef]
  159. Perdigon, G.; Alvarez, S.; de Macias, M.N.E.; Roux, M.E.; de Ruiz, A.P.H. The oral administration of lactic acid bacteria increase the mucosal intestinal immunity in response to enteropathogens. J. Food Prot. 1990, 53, 404–410. [Google Scholar] [CrossRef]
  160. Brown, M. Modes of action of probiotics: Recent developments. J. Anim. Vet. Adv. 2011, 10, 1895–1900. [Google Scholar] [CrossRef]
  161. Reddyvari, R. Evaluating the Immune Response and Performance Parameters of Broilers to Bacillus subtilis and Mannan Oligosaccharides and Assessing Their Efficacy in Reducing Necrotic Enteritis in Broilers. Ph.D. Thesis, University of Georgia, Athens, GA, USA, 2020. [Google Scholar]
  162. Kumar, M.; Kumar, A.; Nagpal, R.; Mohania, D.; Behare, P.; Verma, V.; Kumar, P.; Poddar, D.; Aggarwal, P.K.; Henry, C. Cancer-preventing attributes of probiotics: An update. Int. J. Food Sci. Nutr. 2010, 61, 473–496. [Google Scholar] [CrossRef]
  163. Oakley, B.B.; Kogut, M.H. Spatial and temporal changes in the broiler chicken cecal and fecal microbiomes and correlations of bacterial taxa with cytokine gene expression. Front. Vet. Sci. 2016, 3, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Otte, J.; Podolsky, D.K. Functional modulation of enterocytes by gram-positive and gram-negative microorganisms. Am. J. Physiol.-Gastrointest. Liver Physiol. 2004, 286, G613–G626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Yan, F.; Cao, H.; Cover, T.L.; Whitehead, R.; Washington, M.K.; Polk, D.B. Soluble proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth. Gastroenterology 2007, 132, 562–575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Gharib-Naseri, K.; de Paula, J.C.D.; Doranalli, K.; Kheravii, S.; Swick, R.A.; Choct, M.; Wu, S. Modulations of genes related to gut integrity, apoptosis, and immunity underlie the beneficial effects of Bacillus amyloliquefaciens CECT 5940 in broilers fed diets with different protein levels in a necrotic enteritis challenge model. J. Anim. Sci. Biotechnol. 2020, 11, 1–13. [Google Scholar] [CrossRef]
  167. Flint, H.J.; Scott, K.P.; Duncan, S.H.; Louis, P.; Forano, E. Microbial degradation of complex carbohydrates in the gut. Gut Microb. 2012, 3, 289–306. [Google Scholar] [CrossRef] [Green Version]
  168. Murugesan, G.R.; Gabler, N.K.; Persia, M.E. Effects of direct-fed microbial supplementation on broiler performance, intestinal nutrient transport and integrity under experimental conditions with increased microbial challenge. Br. Poult. Sci. 2014, 55, 89–97. [Google Scholar] [CrossRef]
  169. Awad, W.A.; Ghareeb, K.; Nitsch, S.; Pasteiner, S.; Abdel-Raheem, S.; Böhm, J. Effects of dietary inclusion of prebiotic, probiotic and synbiotic on the intestinal glucose absorption of broiler chickens. Int. J. Poult. Sci. 2008, 7, 686–691. [Google Scholar] [CrossRef] [Green Version]
  170. Ahmad, I. Effect of probiotics on broilers performance. Int. J. Poult. Sci. 2006, 5, 593–597. [Google Scholar]
  171. Neveling, D.P.; Dicks, L.M. Probiotics: An antibiotic replacement strategy for healthy broilers and productive rearing. Probiotics Antimicrob. Proteins 2021, 13, 1–11. [Google Scholar] [CrossRef]
  172. LeBlanc, J.G.; Milani, C.; De Giori, G.S.; Sesma, F.; Van Sinderen, D.; Ventura, M. Bacteria as vitamin suppliers to their host: A gut microbiota perspective. Curr. Opin. Biotechnol. 2013, 24, 160–168. [Google Scholar] [CrossRef]
  173. Dertli, E.; Mayer, M.J.; Narbad, A. Impact of the exopolysaccharide layer on biofilms, adhesion and resistance to stress in Lactobacillus johnsonii FI9785. BMC Microbiol. 2015, 15, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Jha, R.; Das, R.; Oak, S.; Mishra, P. Probiotics (direct-fed microbials) in poultry nutrition and their effects on nutrient utilization, growth and laying performance, and gut health: A systematic review. Animals 2020, 10, 1863. [Google Scholar] [CrossRef] [PubMed]
  175. Abdel-Raheem, S.M.; Abd-Allah, S.M. The effect of single or combined dietary supplementation of mannan oligosacharide and probiotics on performance and slaughter characteristics of broilers. Int. J. Poult. Sci. 2011, 10, 854–862. [Google Scholar]
  176. Bedford, M. Removal of antibiotic growth promoters from poultry diets: Implications and strategies to minimise subsequent problems. Worlds Poult. Sci. J. 2000, 56, 347–365. [Google Scholar] [CrossRef]
  177. Kabir, S.L.; Rahman, M.M.; Rahman, M.B.; Rahman, M.M.; Ahmed, S.U. The dynamics of probiotics on growth performance and immune response in broilers. Int. J. Poult. Sci. 2004, 3, 361–364. [Google Scholar]
  178. Lavoie, B.; Lian, J.B.; Mawe, G.M. Regulation of Bone Metabolism by Serotonin; Understanding the Gut-Bone Signaling Axis; Springer: Cham, Switzerland, 2017; pp. 35–46. [Google Scholar]
  179. Ramsey, W.; Isales, C.M. Intestinal incretins and the regulation of bone physiology. In Understanding the Gut-Bone Signaling Axis; Springer: Cham, Switzerland, 2017; Volume 1033, pp. 13–33. [Google Scholar]
  180. Jiang, S.; Yan, F.; Hu, J.; Mohammed, A.; Cheng, H. Bacillus subtilis-based probiotic improves skeletal health and immunity in broiler chickens exposed to heat stress. Animals 2021, 11, 1494. [Google Scholar] [CrossRef]
  181. Rehman, A.; Arif, M.; Sajjad, N.; Al-Ghadi, M.Q.; Alagawany, M.; Abd El-Hack, M.E.; Alhimaidi, A.R.; Elnesr, S.S.; Almutairi, B.O.; Amran, R.A. Dietary effect of probiotics and prebiotics on broiler performance, carcass, and immunity. Poult. Sci. 2020, 99, 6946–6953. [Google Scholar] [CrossRef]
  182. Nyamagonda, H.; Swamy, M.N.; Veena, T.; Jayakumar, K.; Swamy, H.D. Effect of prebiotic and probiotics on growth performance in broiler chickens. Indian J. Anim. Res. 2011, 45, 271–275. [Google Scholar]
  183. Ferreira, F.; Kussakawa, K. Probiotics-Microorganisms in Favor of Life; Biotecnologia, Ciência & Desenvolvimento; State University of Paraíba: Campina Grande, Brazil, 1999; Volume 8, pp. 40–43. [Google Scholar]
  184. Song, J.; Xiao, K.; Ke, Y.L.; Jiao, L.F.; Hu, C.H.; Diao, Q.Y.; Shi, B.; Zou, X.T. Effect of a probiotic mixture on intestinal microflora, morphology, and barrier integrity of broilers subjected to heat stress. Poult. Sci. 2014, 93, 581–588. [Google Scholar] [CrossRef]
  185. Wampler, J.L.; Kim, K.; Jaradat, Z.; Bhunia, A.K. Heat shock protein 60 acts as a receptor for the Listeria adhesion protein in Caco-2 cells. Infect. Immun. 2004, 72, 931–936. [Google Scholar] [CrossRef] [Green Version]
  186. Faderl, M.; Noti, M.; Corazza, N.; Mueller, C. Keeping bugs in check: The mucus layer as a critical component in maintaining intestinal homeostasis. IUBMB Life 2015, 67, 275–285. [Google Scholar] [CrossRef] [PubMed]
  187. McGuckin, M.A.; Lindén, S.K.; Sutton, P.; Florin, T.H. Mucin dynamics and enteric pathogens. Nat. Rev. Microbiol. 2011, 9, 265–278. [Google Scholar] [CrossRef] [PubMed]
  188. Inan, M.S.; Rasoulpour, R.J.; Yin, L.; Hubbard, A.K.; Rosenberg, D.W.; Giardina, C. The luminal short-chain fatty acid butyrate modulates NF-κB activity in a human colonic epithelial cell line. Gastroenterology 2000, 118, 724–734. [Google Scholar] [CrossRef]
  189. Zhou, Z.Y.; Packialakshmi, B.; Makkar, S.K.; Dridi, S.; Rath, N.C. Effect of butyrate on immune response of a chicken macrophage cell line. Vet. Immunol. Immunopathol. 2014, 162, 24–32. [Google Scholar] [CrossRef] [PubMed]
  190. Warren, H.S. Toll-like receptors. Crit. Care Med. 2005, 33, S457–S459. [Google Scholar] [CrossRef] [PubMed]
  191. Medzhitov, R. Toll-like receptors and innate immunity. Nat. Rev. Immunol. 2001, 1, 135–145. [Google Scholar] [CrossRef]
  192. Rakoff-Nahoum, S.; Paglino, J.; Eslami-Varzaneh, F.; Edberg, S.; Medzhitov, R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 2004, 118, 229–241. [Google Scholar] [CrossRef] [Green Version]
  193. Gewirtz, A.T.; Navas, T.A.; Lyons, S.; Godowski, P.J.; Madara, J.L. Cutting edge: Bacterial flagellin activates basolaterally expressed TLR5 to induce epithelial proinflammatory gene expression. J. Immunol. 2001, 167, 1882–1885. [Google Scholar] [CrossRef] [Green Version]
  194. Rachmilewitz, D.; Katakura, K.; Karmeli, F.; Hayashi, T.; Reinus, C.; Rudensky, B.; Akira, S.; Takeda, K.; Lee, J.; Takabayashi, K. Toll-like receptor 9 signaling mediates the anti-inflammatory effects of probiotics in murine experimental colitis. Gastroenterology 2004, 126, 520–528. [Google Scholar] [CrossRef]
  195. Dhama, K.; Verma, V.; Sawant, P.M.; Tiwari, R.; Vaid, R.K.; Chauhan, R.S. Applications of probiotics in poultry: Enhancing immunity and beneficial effects on production performances and health-A review. J. Immunol. Immunopathol. 2011, 13, 1–19. [Google Scholar]
  196. Dharmani, P.; Srivastava, V.; Kissoon-Singh, V.; Chadee, K. Role of intestinal mucins in innate host defense mechanisms against pathogens. J. Innate Immun. 2009, 1, 123–135. [Google Scholar] [CrossRef] [PubMed]
  197. Bai, S.P.; Wu, A.M.; Ding, X.M.; Lei, Y.; Bai, J.; Zhang, K.Y.; Chio, J.S. Effects of probiotic-supplemented diets on growth performance and intestinal immune characteristics of broiler chickens. Poult. Sci. 2013, 92, 663–670. [Google Scholar] [CrossRef] [PubMed]
  198. Menconi, A.; Bielke, L.R.; Hargis, B.M.; Tellez, G. Immuno-modulation and anti-inflammatory effects of antibiotic growth promoters versus probiotics in the intestinal tract. J. Microbiol. Res. Rev. 2014, 2, 62–67. [Google Scholar]
  199. Borchers, A.T.; Selmi, C.; Meyers, F.J.; Keen, C.L.; Gershwin, M.E. Probiotics and immunity. J. Gastroenterol. 2009, 44, 26–46. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  200. Jijon, H.; Backer, J.; Diaz, H.; Yeung, H.; Thiel, D.; McKaigney, C.; De Simone, C.; Madsen, K. DNA from probiotic bacteria modulates murine and human epithelial and immune function. Gastroenterology 2004, 126, 1358–1373. [Google Scholar] [CrossRef]
  201. Nicetic, M.; Kailasapathy, K.; Tarasoff, L. Mechanical Stability of Food Gum Gels for Immobilization of Probiotic Bacteria. In Proceedings of the 8th International Workshop on Bioencapsulation, Recent Progress in Research and Technology, Trondheim, Norway, 13–15 September 1999; Abstract, P11. pp. 13–15. [Google Scholar]
  202. Würth, R.; Hörmannsperger, G.; Wilke, J.; Foerst, P.; Haller, D.; Kulozik, U. Protective effect of milk protein based microencapsulation on bacterial survival in simulated gastric juice versus the murine gastrointestinal system. J. Funct. Foods 2015, 15, 116–125. [Google Scholar] [CrossRef]
  203. Cottyn, B.; Heylen, K.; Heyrman, J.; Vanhouteghem, K.; Pauwelyn, E.; Bleyaert, P.; Van Vaerenbergh, J.; Höfte, M.; De Vos, P.; Maes, M. Pseudomonas cichorii as the causal agent of midrib rot, an emerging disease of greenhouse-grown butterhead lettuce in Flanders. Syst. Appl. Microbiol. 2009, 32, 211–225. [Google Scholar] [CrossRef] [Green Version]
  204. Nkukwana, T.T.; Muchenje, V.; Masika, P.J.; Mushonga, B. Intestinal morphology, digestive organ size and digesta pH of broiler chickens fed diets supplemented with or without Moringa oleifera leaf meal. S. Afr. J. Anim. Sci. 2015, 45, 362–370. [Google Scholar] [CrossRef] [Green Version]
  205. Shanmugasundaram, R.; Mortada, M.; Murugesan, G.R.; Selvaraj, R.K. In vitro characterization and analysis of probiotic species in the chicken intestine by real-time polymerase chain reaction. Poult. Sci. 2019, 98, 5840–5846. [Google Scholar] [CrossRef]
  206. Zmora, N.; Zilberman-Schapira, G.; Suez, J.; Mor, U.; Dori-Bachash, M.; Bashiardes, S.; Kotler, E.; Zur, M.; Regev-Lehavi, D.; Brik, R.B. Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell 2018, 174, 1388–1405.e21. [Google Scholar] [CrossRef] [Green Version]
  207. Mullineaux-Sanders, C.; Suez, J.; Elinav, E.; Frankel, G. Sieving through gut models of colonization resistance. Nat. Microbiol. 2018, 3, 132–140. [Google Scholar] [CrossRef] [PubMed]
  208. Ferreiro, A.; Dantas, G.; Ciorba, M.A. Insights into how probiotics colonize the healthy human gut. Gastroenterology 2019, 156, 820–822. [Google Scholar] [CrossRef] [PubMed]
  209. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef] [PubMed]
  210. Murphy, E.F.; Cotter, P.D.; Healy, S.; Marques, T.M.; O’sullivan, O.; Fouhy, F.; Clarke, S.F.; O’toole, P.W.; Quigley, E.M.; Stanton, C. Composition and energy harvesting capacity of the gut microbiota: Relationship to diet, obesity and time in mouse models. Gut 2010, 59, 1635–1642. [Google Scholar] [CrossRef] [PubMed]
  211. Oakley, B.B.; Lillehoj, H.S.; Kogut, M.H.; Kim, W.K.; Maurer, J.J.; Pedroso, A.; Lee, M.D.; Collett, S.R.; Johnson, T.J.; Cox, N.A. The chicken gastrointestinal microbiome. FEMS Microbiol. Lett. 2014, 360, 100–112. [Google Scholar] [CrossRef] [PubMed]
  212. Bozdogan, B.; Galopin, S.; Leclercq, R. Characterization of a new erm-related macrolide resistance gene present in probiotic strains of Bacillus clausii. Appl. Environ. Microbiol. 2004, 70, 280–284. [Google Scholar] [CrossRef] [Green Version]
  213. Gueimonde, M.; Sánchez, B.; de Los Reyes-Gavilán Clara, G.; Margolles, A. Antibiotic resistance in probiotic bacteria. Front. Microbiol. 2013, 4, 202. [Google Scholar] [CrossRef] [Green Version]
  214. Feld, L.; Schjørring, S.; Hammer, K.; Licht, T.R.; Danielsen, M.; Krogfelt, K.; Wilcks, A. Selective pressure affects transfer and establishment of a Lactobacillus plantarum resistance plasmid in the gastrointestinal environment. J. Antimicrob. Chemother. 2008, 61, 845–852. [Google Scholar] [CrossRef] [Green Version]
  215. Witte, W. Ecological impact of antibiotic use in animals on different complex microflora: Environment. Int. J. Antimicrob. Agents 2000, 14, 321–325. [Google Scholar] [CrossRef]
  216. Schjørring, S.; Krogfelt, K.A. Assessment of bacterial antibiotic resistance transfer in the gut. Int. J. Microbiol. 2011, 2011, 312956. [Google Scholar] [CrossRef] [Green Version]
  217. Hidayat, M.N.; Malaka, R.; Agustina, L.; Pakiding, W. Abdominal fat percentage and carcass quality of broiler given probiotics Bacillus spp. Metab. Clin. Exp. 2016, 22, 3–60. [Google Scholar]
  218. Haines, M.D.; Parker, H.M.; McDaniel, C.D.; Kiess, A.S. When rooster semen is exposed to Lactobacillus fertility is reduced. Int. J. Poult. Sci. 2015, 14, 541–547. [Google Scholar] [CrossRef] [Green Version]
  219. Peralta-Sánchez, J.M.; Martín-Platero, A.M.; Ariza-Romero, J.J.; Rabelo-Ruiz, M.; Zurita-González, M.J.; Baños, A.; Rodríguez-Ruano, S.M.; Maqueda, M.; Valdivia, E.; Martínez-Bueno, M. Egg production in poultry farming is improved by probiotic bacteria. Front. Microbiol. 2019, 10, 1042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  220. Haines, M.D.; Parker, H.M.; McDaniel, C.D.; Kiess, A.S. Impact of 6 different intestinal bacteria on broiler breeder sperm motility in vitro. Poult. Sci. 2013, 92, 2174–2181. [Google Scholar] [CrossRef] [PubMed]
  221. Aguilar-Toalá, J.E.; Garcia-Varela, R.; Garcia, H.S.; Mata-Haro, V.; González-Córdova, A.F.; Vallejo-Cordoba, B.; Hernández-Mendoza, A. Postbiotics: An evolving term within the functional foods field. Trends Food Sci. Technol. 2018, 75, 105–114. [Google Scholar] [CrossRef]
  222. Lee, M.; Zang, Z.; Choi, E.; Shin, H.; Ji, G. Cytoskeleton reorganization and cytokine production of macrophages by bifidobacterial cells and cell-free extracts. J. Microbiol. Biotechnol. 2002, 12, 398–405. [Google Scholar]
  223. Li, S.; Zhao, Y.; Zhang, L.; Zhang, X.; Huang, L.; Li, D.; Niu, C.; Yang, Z.; Wang, Q. Antioxidant activity of Lactobacillus plantarum strains isolated from traditional Chinese fermented foods. Food Chem. 2012, 135, 1914–1919. [Google Scholar] [CrossRef]
  224. Kim, H.G.; Lee, S.Y.; Kim, N.R.; Lee, H.Y.; Ko, M.Y.; Jung, B.J.; Kim, C.M.; Lee, J.M.; Park, J.H.; Han, S.H. Lactobacillus plantarum lipoteichoic acid down-regulated Shigella flexneri peptidoglycan-induced inflammation. Mol. Immunol. 2011, 48, 382–391. [Google Scholar] [CrossRef]
  225. Tiptiri-Kourpeti, A.; Spyridopoulou, K.; Santarmaki, V.; Aindelis, G.; Tompoulidou, E.; Lamprianidou, E.E.; Saxami, G.; Ypsilantis, P.; Lampri, E.S.; Simopoulos, C. Lactobacillus casei exerts anti-proliferative effects accompanied by apoptotic cell death and up-regulation of TRAIL in colon carcinoma cells. PLoS ONE 2016, 11, e0147960. [Google Scholar]
  226. Matsuguchi, T.; Takagi, A.; Matsuzaki, T.; Nagaoka, M.; Ishikawa, K.; Yokokura, T.; Yoshikai, Y. Lipoteichoic acids from Lactobacillus strains elicit strong tumor necrosis factor alpha-inducing activities in macrophages through Toll-like receptor 2. Clin. Vaccine Immunol. 2003, 10, 259–266. [Google Scholar] [CrossRef] [Green Version]
  227. Sawada, H.; Furushiro, M.; Hirai, K.; Motoike, M.; Watanabe, T.; Yokokura, T. Purification and Characterization of an Antihypertensive Compound from Lactohacillus casei. Agric. Biol. Chem. 1990, 54, 3211–3219. [Google Scholar] [PubMed] [Green Version]
  228. De Almada, C.N.; Almada, C.N.; Martinez, R.C.; Sant’Ana, A.S. Paraprobiotics: Evidences on their ability to modify biological responses, inactivation methods and perspectives on their application in foods. Trends Food Sci. Technol. 2016, 58, 96–114. [Google Scholar] [CrossRef]
  229. Taverniti, V.; Guglielmetti, S. The immunomodulatory properties of probiotic microorganisms beyond their viability (ghost probiotics: Proposal of paraprobiotic concept). Genes Nutr. 2011, 6, 261–274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Ou, C.; Lin, S.; Tsai, J.; Lin, M. Heat-killed lactic acid bacteria enhance immunomodulatory potential by skewing the immune response toward Th1 polarization. J. Food Sci. 2011, 76, M260–M267. [Google Scholar] [CrossRef] [PubMed]
  231. Shin, H.S.; Park, S.Y.; Lee, D.K.; Kim, S.; An, H.M.; Kim, J.R.; Kim, M.J.; Cha, M.G.; Lee, S.W.; Kim, K.J. Hypocholesterolemic effect of sonication-killed Bifidobacterium longum isolated from healthy adult Koreans in high cholesterol fed rats. Arch. Pharm. Res. 2010, 33, 1425–1431. [Google Scholar] [CrossRef]
  232. Ananta, E.; Knorr, D. Comparison of inactivation pathways of thermal or high pressure inactivated Lactobacillus rhamnosus ATCC 53103 by flow cytometry analysis. Food Microbiol. 2009, 26, 542–546. [Google Scholar] [CrossRef]
  233. Gandhi, A.; Shah, N.P. Effect of salt on cell viability and membrane integrity of Lactobacillus acidophilus, Lactobacillus casei and Bifidobacterium longum as observed by flow cytometry. Food Microbiol. 2015, 49, 197–202. [Google Scholar] [CrossRef]
  234. Tsilingiri, K.; Rescigno, M. Postbiotics: What else? Benef. Microb. 2013, 4, 101–107. [Google Scholar] [CrossRef]
  235. Konstantinov, S.R.; Kuipers, E.J.; Peppelenbosch, M.P. Functional genomic analyses of the gut microbiota for CRC screening. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 741–745. [Google Scholar] [CrossRef]
  236. Yan, F.; Cao, H.; Cover, T.L.; Washington, M.K.; Shi, Y.; Liu, L.; Chaturvedi, R.; Peek, R.M.; Wilson, K.T.; Polk, D.B. Colon-specific delivery of a probiotic-derived soluble protein ameliorates intestinal inflammation in mice through an EGFR-dependent mechanism. J. Clin. Investig. 2011, 121, 2242–2253. [Google Scholar] [CrossRef] [Green Version]
  237. Humam, A.M.; Loh, T.C.; Foo, H.L.; Samsudin, A.A.; Mustapha, N.M.; Zulkifli, I.; Izuddin, W.I. Effects of feeding different postbiotics produced by Lactobacillus plantarum on growth performance, carcass yield, intestinal morphology, gut microbiota composition, immune status, and growth gene expression in broilers under heat stress. Animals 2019, 9, 644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  238. Kareem, K.Y.; Hooi Ling, F.; Teck Chwen, L.; May Foong, O.; Anjas Asmara, S. Inhibitory activity of postbiotic produced by strains of Lactobacillus plantarum using reconstituted media supplemented with inulin. Gut Pathog. 2014, 6, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Tsilingiri, K.; Barbosa, T.; Penna, G.; Caprioli, F.; Sonzogni, A.; Viale, G.; Rescigno, M. Probiotic and postbiotic activity in health and disease: Comparison on a novel polarised ex-vivo organ culture model. Gut 2012, 61, 1007–1015. [Google Scholar] [CrossRef] [Green Version]
  240. Choi, S.S.; Kim, Y.; Han, K.S.; You, S.; Oh, S.; Kim, S.H. Effects of Lactobacillus strains on cancer cell proliferation and oxidative stress in vitro. Lett. Appl. Microbiol. 2006, 42, 452–458. [Google Scholar] [CrossRef]
  241. Nakamura, F.; Ishida, Y.; Sawada, D.; Ashida, N.; Sugawara, T.; Sakai, M.; Goto, T.; Kawada, T.; Fujiwara, S. Fragmented lactic acid bacterial cells activate peroxisome proliferator-activated receptors and ameliorate dyslipidemia in obese mice. J. Agric. Food Chem. 2016, 64, 2549–2559. [Google Scholar] [CrossRef] [PubMed]
  242. Thanh, N.T.; Loh, T.C.; Foo, H.L.; Hair-Bejo, M.; Azhar, B.K. Effects of feeding metabolite combinations produced by Lactobacillus plantarum on growth performance, faecal microbial population, small intestine villus height and faecal volatile fatty acids in broilers. Br. Poult. Sci. 2009, 50, 298–306. [Google Scholar] [CrossRef] [PubMed]
  243. Compare, D.; Rocco, A.; Coccoli, P.; Angrisani, D.; Sgamato, C.; Iovine, B.; Salvatore, U.; Nardone, G. Lactobacillus casei DG and its postbiotic reduce the inflammatory mucosal response: An ex-vivo organ culture model of post-infectious irritable bowel syndrome. BMC Gastroenterol. 2017, 17, 53. [Google Scholar] [CrossRef] [Green Version]
  244. Verma, A.; Shukla, G. Synbiotic (Lactobacillus rhamnosus Lactobacillus acidophilus inulin) attenuates oxidative stress and colonic damage in 1, 2 dimethylhydrazine dihydrochloride-induced colon carcinogenesis in Sprague–Dawley rats. Eur. J. Cancer Prev. 2014, 23, 550–559. [Google Scholar] [CrossRef]
  245. Abd El-Ghany, W.; Hosny, F.; Quesnell, R.; Sakai, L. The Effect of a Postbiotic Produced by Stabilized Non-Viable Lactobacilli on the Health, Growth Performance, Immunity, and Gut Status of Colisepticaemic Broiler Chickens; Research Square: Durham, NC, USA, 2022. [Google Scholar] [CrossRef]
  246. Abdulamir, A.S.; Hafidh, R.R.; Bakar, F.A. Molecular detection, quantification, and isolation of Streptococcus gallolyticus bacteria colonizing colorectal tumors: Inflammation-driven potential of carcinogenesis via IL-1, COX-2, and IL-8. Mol. Cancer 2010, 9, 249. [Google Scholar] [CrossRef] [Green Version]
  247. Shigwedha, N. Probiotical cell fragments (PCFs) as “novel nutraceutical ingredients”. J. Biosci. Med. 2014, 2, 43–55. [Google Scholar] [CrossRef] [Green Version]
  248. Lin, C.; Fung, Z.; Wu, C.; Chung, T. Molecular Characterization of a Plasmid-Borne (pTC82) Chloramphenicol Resistance Determinant (cat-TC) fromLactobacillus reuteriG4. Plasmid 1996, 36, 116–124. [Google Scholar] [CrossRef] [PubMed]
  249. Sleator, R.D. Designer probiotics: Development and applications in gastrointestinal health. World J. Gastrointest. Pathophysiol. 2015, 6, 73. [Google Scholar] [CrossRef] [PubMed]
  250. Sheehan, V.M.; Sleator, R.D.; Hill, C.; Fitzgerald, G.F. Improving gastric transit, gastrointestinal persistence and therapeutic efficacy of the probiotic strain Bifidobacterium breve UCC2003. Microbiology 2007, 153, 3563–3571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  251. Durrer, K.E.; Allen, M.S.; Hunt von Herbing, I. Genetically engineered probiotic for the treatment of phenylketonuria (PKU); assessment of a novel treatment in vitro and in the PAHenu2 mouse model of PKU. PLoS ONE 2017, 12, e0176286. [Google Scholar] [CrossRef] [Green Version]
  252. Volzing, K.; Borrero, J.; Sadowsky, M.J.; Kaznessis, Y.N. Antimicrobial peptides targeting Gram-negative pathogens, produced and delivered by lactic acid bacteria. ACS Synth. Biol. 2013, 2, 643–650. [Google Scholar] [CrossRef] [Green Version]
  253. Marelli, B.; Perez, A.R.; Banchio, C.; de Mendoza, D.; Magni, C. Oral immunization with live Lactococcus lactis expressing rotavirus VP8* subunit induces specific immune response in mice. J. Virol. Methods 2011, 175, 28–37. [Google Scholar] [CrossRef]
  254. Kitov, P.I.; Sadowska, J.M.; Mulvey, G.; Armstrong, G.D.; Ling, H.; Pannu, N.S.; Read, R.J.; Bundle, D.R. Shiga-like toxins are neutralized by tailored multivalent carbohydrate ligands. Nature 2000, 403, 669–672. [Google Scholar] [CrossRef]
  255. Mathipa, M.G.; Thantsha, M.S. Probiotic engineering: Towards development of robust probiotic strains with enhanced functional properties and for targeted control of enteric pathogens. Gut Pathog. 2017, 9, 28. [Google Scholar] [CrossRef]
  256. Muheem, A.; Shakeel, F.; Jahangir, M.A.; Anwar, M.; Mallick, N.; Jain, G.K.; Warsi, M.H.; Ahmad, F.J. A review on the strategies for oral delivery of proteins and peptides and their clinical perspectives. Saudi Pharm. J. 2016, 24, 413–428. [Google Scholar] [CrossRef] [Green Version]
  257. Russo, E.; Giudici, F.; Fiorindi, C.; Ficari, F.; Scaringi, S.; Amedei, A. Immunomodulating activity and therapeutic effects of short chain fatty acids and tryptophan post-biotics in inflammatory bowel disease. Front. Immunol. 2019, 10, 2754. [Google Scholar] [CrossRef] [Green Version]
  258. Chaney, W.E.; Naqvi, S.A.; Gutierrez, M.; Gernat, A.; Johnson, T.J.; Petry, D. Dietary Inclusion of a Saccharomyces cerevisiae-Derived Postbiotic Is Associated with Lower Salmonella enterica Burden in Broiler Chickens on a Commercial Farm in Honduras. Microorganisms 2022, 10, 544. [Google Scholar] [CrossRef] [PubMed]
  259. Gibson, G.R.; Scott, K.P.; Rastall, R.A.; Tuohy, K.M.; Hotchkiss, A.; Dubert-Ferrandon, A.; Gareau, M.; Murphy, E.F.; Saulnier, D.; Loh, G. Dietary prebiotics: Current status and new definition. Food Sci. Technol. Bull. Funct. Foods 2010, 7, 1–19. [Google Scholar] [CrossRef] [Green Version]
  260. Jiang, H.Q.; Gong, L.M.; Ma, Y.X.; He, Y.H.; Li, D.F.; Zhai, H.X. Effect of stachyose supplementation on growth performance, nutrient digestibility and caecal fermentation characteristics in broilers. Br. Poult. Sci. 2006, 47, 516–522. [Google Scholar] [CrossRef]
  261. Bindels, L.B.; Delzenne, N.M.; Cani, P.D.; Walter, J. Towards a more comprehensive concept for prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2015, 12, 303–310. [Google Scholar] [CrossRef] [PubMed]
  262. Valcheva, R.; Dieleman, L.A. Prebiotics: Definition and protective mechanisms. Best Pract. Res. Clin. Gastroenterol. 2016, 30, 27–37. [Google Scholar] [CrossRef] [PubMed]
  263. Hajati, H.; Rezaei, M. The application of prebiotics in poultry production. Int. J. Poult. Sci. 2010, 9, 298–304. [Google Scholar] [CrossRef] [Green Version]
  264. Everard, A.; Lazarevic, V.; Gaïa, N.; Johansson, M.; Ståhlman, M.; Backhed, F.; Delzenne, N.M.; Schrenzel, J.; François, P.; Cani, P.D. Microbiome of prebiotic-treated mice reveals novel targets involved in host response during obesity. ISME J. 2014, 8, 2116–2130. [Google Scholar] [CrossRef]
  265. Khaksar, V.; Golian, A.; Kermanshahi, H.; Movasseghi, A.R.; Jamshidi, A. Effect of prebiotic fermacto on gut development and performance of broiler chickens fed diet low in digestible amino acids. J. Anim. Vet. Adv. 2008, 7, 257–263. [Google Scholar]
  266. Teng, P.; Kim, W.K. Roles of prebiotics in intestinal ecosystem of broilers. Front. Vet. Sci. 2018, 5, 245. [Google Scholar] [CrossRef]
  267. Spring, P.; Wenk, C.; Dawson, K.A.; Newman, K.E. The effects of dietary mannaoligosaccharides on cecal parameters and the concentrations of enteric bacteria in the ceca of salmonella-challenged broiler chicks. Poult. Sci. 2000, 79, 205–211. [Google Scholar] [CrossRef]
  268. Froebel, L.K.; Jalukar, S.; Lavergne, T.A.; Lee, J.T.; Duong, T. Administration of dietary prebiotics improves growth performance and reduces pathogen colonization in broiler chickens. Poult. Sci. 2019, 98, 6668–6676. [Google Scholar] [CrossRef] [PubMed]
  269. Day, C.J.; Tiralongo, J.; Hartnell, R.D.; Logue, C.; Wilson, J.C.; von Itzstein, M.; Korolik, V. Differential carbohydrate recognition by Campylobacter jejuni strain 11168: Influences of temperature and growth conditions. PLoS ONE 2009, 4, e4927. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  270. Ferket, P.R.; Santos, A.A.; Oviedo-Rondón, E.O. Dietary Factors that Affect Gut Health and Pathogen Colonization. In Proceedings of the 32nd Annual Carolina Poultry Nutrition Conference, Raleigh, NC, USA, 27 October 2005. [Google Scholar]
  271. Janardhana, V.; Broadway, M.M.; Bruce, M.P.; Lowenthal, J.W.; Geier, M.S.; Hughes, R.J.; Bean, A.G. Prebiotics modulate immune responses in the gut-associated lymphoid tissue of chickens. J. Nutr. 2009, 139, 1404–1409. [Google Scholar] [CrossRef] [PubMed]
  272. Broderick, T.J.; Duong, T. Mechanisms of Lactobacillus persistence and colonization in the gastrointestinal tract of poultry, a review. Int. J. Probiotics Prebiotics 2016, 11, 15. [Google Scholar]
  273. Yang, Y.; Iji, P.A.; Kocher, A.; Mikkelsen, L.L.; Choct, M. Effects of mannanoligosaccharide and fructooligosaccharide on the response of broilers to pathogenic Escherichia coli challenge. Br. Poult. Sci. 2008, 49, 550–559. [Google Scholar] [CrossRef]
  274. Sharma, R.; Hinton, M.; Bedford, M.R. Diet influences the colonisation of Campylobacter jejuni and distribution of mucin carbohydrates in the chick intestinal tract. Cell. Mol. Life Sci. CMLS 2000, 57, 1793–1801. [Google Scholar]
  275. Schrezenmeir, J.; de Vrese, M. Probiotics, prebiotics, and synbiotics—Approaching a definition. Am. J. Clin. Nutr. 2001, 73, 361s–364s. [Google Scholar] [CrossRef] [Green Version]
  276. Farnworth, E.R. Probiotics and Prebiotics. In Handbook of Nutraceuticals and Functional Foods; CRC Press: Boca Raton, FL, USA, 2002; pp. 429–444. [Google Scholar]
  277. Mookiah, S.; Sieo, C.C.; Ramasamy, K.; Abdullah, N.; Ho, Y.W. Effects of dietary prebiotics, probiotic and synbiotics on performance, caecal bacterial populations and caecal fermentation concentrations of broiler chickens. J. Sci. Food Agric. 2014, 94, 341–348. [Google Scholar] [CrossRef]
  278. Chen, Y.; Cheng, Y.; Wen, C.; Kang, Y.; Wang, A.; Zhou, Y. Effects of dietary synbiotic supplementation as an alternative to antibiotic supplementation on the growth performance, carcass characteristics, meat quality, immunity and oxidative status of cherry valley ducks. J. Poult. Sci. 2018, 55, 0170128. [Google Scholar] [CrossRef] [Green Version]
  279. Hassanpour, H.; Moghaddam, A.Z.; Khosravi, M.; Mayahi, M. Effects of synbiotic on the intestinal morphology and humoral immune response in broiler chickens. Livest. Sci. 2013, 153, 116–122. [Google Scholar] [CrossRef]
Figure 1. Regional abundance and diversity of gastrointestinal microbiota of chicken. Created with BioRender.com (26 March 2022).
Figure 1. Regional abundance and diversity of gastrointestinal microbiota of chicken. Created with BioRender.com (26 March 2022).
Foods 11 01401 g001
Figure 2. Dysbiosis induced by different factors alters the gastrointestinal homeostasis causing impaired epithelial barrier function and systemic inflammation. Created with BioRender.com (26 March 2022).
Figure 2. Dysbiosis induced by different factors alters the gastrointestinal homeostasis causing impaired epithelial barrier function and systemic inflammation. Created with BioRender.com (26 March 2022).
Foods 11 01401 g002
Figure 3. Postbiotics are soluble low molecular weight metabolites or cell lysis products derived from live or inactivated probiotic bacteria which when administered in adequate quantities demonstrate beneficial effects on host health. Created with Biorender.com (4 April 2022).
Figure 3. Postbiotics are soluble low molecular weight metabolites or cell lysis products derived from live or inactivated probiotic bacteria which when administered in adequate quantities demonstrate beneficial effects on host health. Created with Biorender.com (4 April 2022).
Foods 11 01401 g003
Figure 4. Prebiotics are polysaccharides or oligosaccharides capable of resisting digestion and absorption in the proximal intestine and is selectively fermented by caecal and colonic bacteria such as Lactobacillus and Bifidobacterium, thus increasing their abundance in host gut. Prebiotics act as decoy receptors for the binding of pathogens, thus preventing their attachment to the host intestinal cells. Prebiotics also serve as a substrate for the production of SCFAs which serve as energy source for the intestinal epithelial cells. Created with Biorender.com (26 April 2022).
Figure 4. Prebiotics are polysaccharides or oligosaccharides capable of resisting digestion and absorption in the proximal intestine and is selectively fermented by caecal and colonic bacteria such as Lactobacillus and Bifidobacterium, thus increasing their abundance in host gut. Prebiotics act as decoy receptors for the binding of pathogens, thus preventing their attachment to the host intestinal cells. Prebiotics also serve as a substrate for the production of SCFAs which serve as energy source for the intestinal epithelial cells. Created with Biorender.com (26 April 2022).
Foods 11 01401 g004
Figure 5. Synbiotics are essentially a combination of probiotics and prebiotics where the probiotic component is specifically fermentable by the prebiotic component and thus helps in establishing a stable population in the host gut. Created with Biorender.com (28 April 2022).
Figure 5. Synbiotics are essentially a combination of probiotics and prebiotics where the probiotic component is specifically fermentable by the prebiotic component and thus helps in establishing a stable population in the host gut. Created with Biorender.com (28 April 2022).
Foods 11 01401 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fathima, S.; Shanmugasundaram, R.; Adams, D.; Selvaraj, R.K. Gastrointestinal Microbiota and Their Manipulation for Improved Growth and Performance in Chickens. Foods 2022, 11, 1401. https://0-doi-org.brum.beds.ac.uk/10.3390/foods11101401

AMA Style

Fathima S, Shanmugasundaram R, Adams D, Selvaraj RK. Gastrointestinal Microbiota and Their Manipulation for Improved Growth and Performance in Chickens. Foods. 2022; 11(10):1401. https://0-doi-org.brum.beds.ac.uk/10.3390/foods11101401

Chicago/Turabian Style

Fathima, Shahna, Revathi Shanmugasundaram, Daniel Adams, and Ramesh K. Selvaraj. 2022. "Gastrointestinal Microbiota and Their Manipulation for Improved Growth and Performance in Chickens" Foods 11, no. 10: 1401. https://0-doi-org.brum.beds.ac.uk/10.3390/foods11101401

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop