Skip to main content

Maintenance of gut microbiome stability for optimum intestinal health in pigs – a review

Abstract

Pigs are exposed to various challenges such as weaning, environmental stressors, unhealthy diet, diseases and infections during their lifetime which adversely affects the gut microbiome. The inability of the pig microbiome to return to the pre-challenge baseline may lead to dysbiosis resulting in the outbreak of diseases. Therefore, the maintenance of gut microbiome diversity, robustness and stability has been influential for optimum intestinal health after perturbations. Nowadays human and animal researches have focused on more holistic approaches to obtain a robust gut microbiota that provides protection against pathogens and improves the digestive physiology and the immune system. In this review, we present an overview of the swine gut microbiota, factors affecting the gut microbiome and the importance of microbial stability in promoting optimal intestinal health. Additionally, we discussed the current understanding of nutritional interventions using fibers and pre/probiotics supplementation as non-antibiotic alternatives to maintain microbiota resilience to replace diminished species.

Introduction

The swine gut microbiome is a complex and dynamic ecosystem harboring immensely diverse microbiota including bacteria, viruses, archaea, and fungi that ideally reside symbiotically in the gut of host animals. Among the microorganisms, the number of bacteria outnumbers other microorganisms [1, 2]. There is a co-existence of several hundred anaerobic bacterial species in the caecum and colon of swine [3, 4] ranging between 1011 and 1012 CFU/g digesta [5], whereas in the stomach and small intestine, a relatively lower numbers of bacteria (107–109 CFU/g fresh matter) are found [6]. These bacteria which inhabit the gastro intestinal tract (GIT) from birth significantly impact animal health, since they protect against the pathogens forming a key barrier, provide essential nutrients to the host from fermentation processes [7, 8] and play a pivotal role in maintaining the host physiological homeostasis, in promoting immune system development, and in regulating host metabolism [9,10,11]. Although the bacterial component of the microbiome is the major component affecting gut microbiome, the ability of the fungal microbiome, mycobiome has also been found to alter gut microbial community structure and cause disease [12,13,14,15]. Commensal fungi and viruses may also cause the alteration in the severity of disease and modification of immunological responses [16,17,18,19].

Gut microbiota composition varies among individuals and throughout the growth state, and is dependent on host genotype and environmental factors. Early microbial exposure, diet, age, breed, and antibiotics have all been implicated to the onset and maintenance of microbial diversity in the human gut [20]. The disruption of the gut ecosystem by perturbations would cause significant decreases in functional richness and microbial diversity as well impairment in metabolic functions. The loss of diversity due to the altered composition of microbes is called “dysbiosis” and can impact the immune system resulting in the emergence and outbreak of diseases and growth deprivation in pigs [21, 22]. The fundamental paradigm shift in our understanding of microorganisms in the GIT has become evident. For instance, all eukaryotes are meta-organisms and it is now accepted that they must be considered together as an inseparable functional unit [23]. This concept also takes into consideration that a tiny fraction of microorganisms consists of pathogens in the microbiome.

The microbiome robustness, the maintenance of diverse and functional microbiota in GIT is crucial for effective swine production. The microbiome robustness depends on the diversity of the microbiome, so it is not enough just to have the presence of a few different beneficial microbes. Accordingly, new strategies are required to manipulate the gut microbiome to prevent or revert unhealthy states caused by perturbations. This paper highlights the importance of microbial stability and nutritional intervention to maintain the stability of microbiota that inhabit within the gut microbiome. Dietary manipulation through the alterations of diet composition [24, 25], nutritional concentrations [26], energy resources [27], and diet types [28] had been widely proven to shape the gut microbiota communities. For conducting this review, literature search was done using the web of science database and data collection was based on more than 140 peer reviewed articles. The search words for this review article were ‘dietary intervention, microbial diversity, perturbation, resilience, stability, and swine gut microbiome’.

Overview of swine gut microbiome

The intestinal microbiota in swine contributes in maintaining its good health of host and producing meat for human consumption [29,30,31]. Among all the microbes, bacteria accounts for the major share of microbiota (> 98% of the entire microbiota) in pig’s microbiome [32] and are influential in the digestion and metabolism of nutrients, immune homeostasis as well as energy supply [33,34,35].

The diverse microbial community originated from the sow and/or the environment rapidly colonizes the sterile GIT of newly born piglets [5, 36]. The initial microbial exposure primarily occurs at birth via urogenital and environmental exposure and at ingestion of colostrum and milk throughout lactation [37]. Immediately after birth, the GIT of piglets are mainly colonized by facultative aerobes or anaerobes. For instance, Swords et al. [38] reported that the sterile colon at birth was initially dominated by facultative anaerobes that stabilized at 109–1010 bacteria/g colonic contents within 12 h from birth subsequently followed by the domination of obligate anaerobes within 48 h after birth making up more than 90% of the microflora thereafter. Some other studies showed that GIT of piglets within 6 h after birth was colonized by bacteria belonging to Streptococcaceae family which became most numerous during 1 to 3 days of birth and were gradually replaced by Lactobacillaceae and Clostridiaceae because of secondary colonization [39]. The dominant bacterial genera found during the suckling period are reported to be Clostridium, Bacteroides and Bifidobacterium [38].

Before weaning, the microbiota remains quite stable in piglet GIT after the initial colonization [40, 41]. Very early and sudden weaning stage of piglets aggravates the qualitative and quantitative alterations of gut microbiota, which may increase pathogenic microorganisms [42]. Besides the diet change from milk to solid food, separation from the dam and co-mingling with other littermates induce a lot of stress to the piglets and it provokes changes in the gut microbiota [42] as well as deprivation in feed intake and growth [43, 44]. More specifically, weaning-associated starvation results in shifts in microbial communities in the GIT that become unstable and less diverse due to the reduction in fermentable substrates [45]. The weaning transition is characterized by a loss of microbial diversity, a decrease in the abundance of bacteria belonging to the Lactobacillus group and an increase in the abundance of facultative anaerobes, including bacteria belonging to the Enterobacteriaceae, Proteobacteriaceae, Clostridiaceae and Prevotellaceae families [46, 47]. Moreover, the phylogenetic composition of fecal microbial community was dominated by Bacteroidetes, Firmicutes, Proteobacteria, and Spirochaetes at the phylum level and, at the genus level, Prevotella, Lactobacillus, and Treponema were the three most abundant genera [48, 49].

The meta-analysis conducted by Holman et al. [50] using 20 publicly available data sets from high-throughput 16S rRNA gene sequence studies revealed that the core genera Prevotella, Clostridium, Alloprevotella, Ruminococcus, and the RC9 were detected in 99% of the faecal samples obtained from commercial swine worldwide. The symbiosis of these core microbiome plays an important role in regulating nutrient metabolism and immunity of the host, ultimately contributing to the health and production of pigs [51, 52]. In a recent study, Li et al. [53] identified a “core” microbiome of 69 bacterial features that were present in all the physiological stages of pigs (lactation, nursery, growing, and finishing). In agreement with the findings of Holman et al. [50] most of these bacterial features were associated with the order Clostridiales, Bacteroidiales, and Lactobacillales and the top three families were Prevotellaceae, Ruminococcaceae, and Lactobacillaceae. Although Megasphaera and Prevotella spp. were present during all the growth stages, others such as Clostridiaceae and Bacteroidetes were not noticeable at lactation and nursery stages but emerged rapidly and became the dominant taxa at the growing and finishing stages [53].

Impact of various factors on the gut microbiome in swine

The distribution and composition of gut microbiota may be influenced by various factors as follows:

Birth weight

The gut microbiota and metabolic status in the piglets are affected by their birth weights indicating that suckling period might be critical for modulating the gut microbiota in low-birth-weight piglets [54].

Physiological stage

Among several factors, physiological stage is considered as one of the determinant factors affecting the colonization and stabilization of gut microbiota in neonatal piglets [55] and the abundance of bacterial diversity is also influenced by age [56]. During the weaning at 21–28 days of age, the change in diet, as well as other environmental factors induce several stressors leading to significant alterations in the composition of swine gut microbiota and the pathways associated with nutrient metabolism [57].

Sex

The other determinant factor is sex. For instance, a higher abundance of Veillonellaceae, Roseburia, Bulleidia and Escherichia was seen in boars whereas the relative abundance of Treponema and Bacteroides was observed in gilts suggesting the influence of sex hormone, specifically androgen, in gut microbial composition [58]. A negative correlation was found between Treponema and androgen metabolites which is consistent with the reports that demonstrated the inhibition of Treponema growth due to higher level of testosterone [59]. However, the gut microbial structure of castrated boars was of higher similarity to gilts indicating this shift in microbiota composition of the boars towards that of gilts might be linked with inadequate secretion of androgen hormone due to castration in boars [58].

Breeds

A distinct gut microbiota composition is found in different breeds of pigs [60]. For instance, Landrace displayed a higher abundance of cellulolytic bacteria, indicating this breed has a better ability in fiber digestion [61] and Yorkshire showed reduced Firmicutes and greater Bacteroidetes concentrations, whereas Tibetian pigs had greater concentrations of bacteria from Elusimicrobia, Fibrobacteres and Spirochaetes [62] suggesting that microbiome composition may be affected depending on where the breed is originated or raised. The reduced Firmicutes/Bacteroidetes ratio in the Yorkshire pigs exhibited apparent differences compared to Rongchang and Tibetain pigs [62]. The various compositions of the intestinal microbiome can influence the usage of the host energy and nutrients [63]. Therefore, difference of microbial community could contribute to the quality and quantity of production in different breeds.

Different intestinal segments/contents

The intestinal tract of pigs is segmented into different compartments based on the differences in anatomical structures, physiological functions, and microbiota communities. The small intestine which is again divided into duodenum, ileum and jejunum mainly host microorganisms that are involved mainly in the digestion and absorption of the proteins, lipids, amino acids, monosaccharides, and some oligosaccharides. On the contrary, the large intestine (colon and cecum) is the habitat for microorganisms which play the role of the degradation of nutrients such as insoluble cellulose that are not digestible in the small intestine [64]. Furthermore, the number of microorganisms is higher in large intestine compared with the small intestine [5], and there is significant variation in microbial composition of the ileum as compared to that of the cecum and colon. For instance, in the ileum, the genera Escherichia-Shigella (23.1%), Terrisporobacter (17.9%), Romboutsia (13.7%) and Clostridium sensustricto (12.9%) are more abundant than in the cecum and colon. In cecum the three most prevalent genera are Alloprevotella (7.2%), Lactobacillus (5.0%), and the Prevotellaceae NK3B31 group (4.4%) whereas in the colon, the 3 most prevalent genera are Streptococcus (10.4%), Lactobacillus (8.8%), and Clostridium (8.0%) [65]. The intestinal microbiota can vary in animals based on biogeographic and geographic distributions [66, 67]. The geographical differences significantly affected the distribution of the phyla Actinobacteria, Verrucomicrobia, Firmicutes, and Fibrobacteres [68]. However, despite the same biogeographical area, the composition and abundance of gut microorganisms attached to the digesta or intestinal mucosa were different [69].

Stability and diversity of gut microbiome

Stability is one of the essential ecological characteristics of the gut microbiome. The gut microbiome shows dynamic equilibrium and remains in its stable ecological state unless it is perturbed [70]. Nevertheless, the relative abundance of each microbe fluctuates over time and varies between and within individuals over the course of their lives [20, 71]. The microbial stability is influenced by several factors as shown in Fig. 1.

Fig. 1
figure 1

The increase in the risk of losing gut microbiome stability is influenced by several factors including therapeutic and sub-therapeutic antibiotics administration, feed types, physical activity, farm management, barn environment, age, breed, microbial infection and stressors

Generally, the gut microbiota is resilient when perturbed and allows the host to keep key species for a long period of time [72]. Thus, resilience is the property of an ecosystem to resist changes under stress or to quickly and fully recover from the perturbations [73]. However, the duration and severity of the perturbations can determine their impact on microbial community. The severe and intense external perturbations would alter the stable microbiota to unstable or transient state leading to an alternative stable state or unhealthy state associated with diseases. If the host acquires unhealthy microbiota having high self-regeneration or resilience potential, it may contribute to chronic microbial associated diseases [74]. Moreover, previous studies have suggested that very low diversity in a microbiome is a good predictor of poor health status [63, 75]. Thus, a healthy functional microbiome should comprise not a single static state but rather a dynamic ecosystem having the ability to recover to an equilibrium state after stress and perturbation [76].

The development of an unhealthy state of the gut is reported to be due to the drastic changes in dietary patterns, microbial infections and the extensive use of antibiotics [77] resulting in significant variation in compositional and/or functional microbiome, with marked decrease in diversity [78]. Diversity of the gut microbiota is likely very important to animal health [79]. The decrease in diversity consequently leads to the reduction of beneficial microorganisms and expansion of pathogenic microbes [80, 81]. The unhealthy states of the gut microbiome due to perturbation can either be temporary or it may develop into a permanent unhealthy state with negative implications. The most drastic perturbations to the gut microbiome are induced by prolonged antibiotic therapy that affects not only the targeted pathogens but also other members of the microbiota [82]. Furthermore, in the last few decades, antibiotics were not only used for therapeutic purpose but also as a growth promoter. The imprudent use of antibiotics as a therapeutic agent or a growth promoter over time has shifted the gut microbial population affecting its stability and diversity and has increased microbial resistance [83, 84] thereby affecting gut health consequently leading to adverse effects on the overall health of the animals as well as humans. For instance, administration of lincomycin (0.1%) through feed to the finishing pigs daily for 1–2 weeks resulted in the relative abundance of pathogenic microbes such as species of Escherichia-Shigella, Clostridium, and Corynebacterium but reduction of fiber degrading bacterial species such as Treponema, Succinivibrio, Fibrobacter, and Cellulosilyticum [85]. With the change in microbial community, lincomycin-administered swine microbiota showed deficiency in polysaccharide degradation and an increase in metabolic pathways related to peptidoglycan maturation and CMP-legionaminate biosynthesis and this pathway is linked with the adherence of pathogenic bacteria to mammalian cell surfaces [86] Thus, by impacting the composition of the microbial community, antibiotics alter microbiota functionality and the metabolites produced [87]. The detrimental impacts of prolonged use of antimicrobials on GI microbiota and host health are presented in Fig. 2.

Fig. 2
figure 2

The overuse of antibiotics on swine gut microbiota has detrimental effects on the host health resulting in the loss of microbial stability and diversity, increased microbial resistance, compromised immunity, and metabolic disruption

In addition, the impact of antibiotic therapy is greatly influenced by ages of the studied populations, the chemical nature, pharmacokinetic and pharmacodynamics properties of the drug, target spectrum, route of administration and excretion, dose and duration, as well as the residing gut microbiota [88]. In a recent study, Gao et al. [89] demonstrated that in-feed administration of 200 mg/kg ampicillin, 5 mg/kg gentamicin, 40 mg/kg metronidazole modified GI microbial population structure and function in 42-day-old ileal-cannulated pigs. In addition, the reduction in Lactobacillus and Bifidobacterium abundance and increase in the abundance of Shigella species by 256-fold compared to the control pigs were also noted with the administration of these antibiotics [89]. Similarly, Li et al. [90] noted that the oral feeding of amoxicillin (30 mg/kg/d) twice daily to neonatal piglets during the first 14 days of age impacted developing gut microbiota and reduced the genes associated with short-chain fatty acid signaling and pancreatic development.

A mixture of olaquindox, kitasamycin, and oxytetracycline calcium (50 mg/kg each) administration as in-feed antibiotics to neonatal piglets has been reported to reduce the relative abundance of beneficial Lactobacillus species and increase the relative abundance of potentially pathogenic Streptococcus suis in both the small intestine and stomach lumen resulting in dysbiosis [91]. In growing piglets, antimicrobial administration induced alterations in microbiota composition in both abundant and less abundant GI microbiota. For instance, a higher relative abundance of Lactobacillus, Eggerthella, Acetanaerobacterium, and Sporacetigenium genera was observed in piglets receiving feed supplemented with tylosin (40 g/t feed) compared to control piglets [92].

The impacts of in-feed sub-therapeutic concentrations of two common antimicrobials such as tylosin (44–11 mg/kg feed) and chlortetracycline (5.5 mg/kg feed) during weaning, starter and growing-finishing periods on swine GIT microbiota composition have shown variable results. For example, tylosin administration resulted in a major shift in the relative abundance of several taxa, while chlortetracycline administration resulted only in minor alterations [93]. Similarly, administration of vancomycin and metronidazole in mice by oral gavage have different effects on Clostridium difficile, where only vancomycin had an obvious impact on microbial community structure [94].

To overcome the adverse effects of therapeutic and sub-therapeutic antibiotic administration in pigs gut microbiome, several possible alternatives have been mentioned [95,96,97]. In the following section, we will focus on the nutritional intervention (mainly dietary fiber), and feed additives, e.g., pre/probiotics supplementation to increase the microbiota diversity in the intestine of pigs thereby increasing resilience.

Impact of nutritional intervention on gut microbiome

Dietary fiber

Dietary fiber mainly constitutes non-starch polysaccharides (NSPs) such as (cellulose, arabinoxylans, chitins, pectins and beta-glucans), lignin and resistant starch [98]. These NSPs are naturally occurring compounds and are present in all plant-based feedstuffs including cereals, tubers, and agro-industrial byproducts [99]. The inclusion of dietary fiber in monogastric animal diets has gained considerable attention due to its potential beneficial effects on gut health and welfare, even though some adverse effects on nutrient and energy digestibility are exhibited [100]. Since pigs cannot degrade non-starch polysaccharides due to the lack of endogenous enzymes, the NSP and resistant starch escapes degradation in the small intestine and reach the lower part of the GIT being available for microbial fermentation [101]. Jha et al. [99], noted that fibers impact the composition and function of the microbiota, in monogastric animals especially the production of short-chain fatty acids. By increasing the proportion of defatted rice bran in the basal diet, the relative abundance of specific microbiota was found to be enhanced resulting in β-diversity variation in growing-finishing pigs [102]. The microbial process of fiber fermentation is variable due to the range of fiber sources and the physicochemical properties of that fiber. It has been reported that fibers fermented at a slow rate results in increased stool output, diluted colonic contents and production of distal colonic short chain fatty acid (SCFA), which is a major energy source for colonocytes [103,104,105]. A complex mix of dietary fibers providing a wide range of structures and monosaccharide units increase the microbiota diversity [106]. A recent study in Durco × Bamei crossbred pigs fed the basal diet supplemented with 10%, 17% and 24% dietary fiber (fermented silage broad bean) significantly altered the diversity of the bacterial community. The abundance of Bacteroidetes and Turicibacter increased with high dietary fiber in cecum and jejunum respectively resulting in alteration of concentration of their metabolites such as bile acids, fatty acids, carbohydrates and carbohydrate conjugate, and organic acids which may potentially influence nutrition absorption [107]. Tang et al. [108] suggested that adding fibers (Broad bean straw silage) to the basal diet significantly increased the α-diversity of the bacterial community in the jejunum and cecum, while the β-diversity decreased of Durco × Bamei crossbred pigs. Consequently, among the most abundant bacterial genera in the cecum, the relative abundance of unidentified Prevotellaceae and Oscillibacter increased with the increase in dietary fiber, while the richness of Romboutsia, Intestinibacter, and Faecalibacterium, decreased with the increase in dietary fiber. An earlier study had demonstrated that the gut microbiota of mice challenged with antibiotic returned to pre-challenge state by feeding fiber-enriched diet whereas the antibiotic challenged mice fed a low fiber diet lost their microbial diversity [109] suggesting that fibers have a direct effect on improving microbiota resilience.

Feed additives (prebiotics and probiotics)

The use of feed additives such as pre/probiotics specifically at post-weaning, have been implemented to minimize the weaning-induced stress and improve microbiome status. Prebiotics are special non-digestible fibers that influence the composition and/or activity of the gastrointestinal microbiota and induce positive effects on host well-being and health [110, 111]. The prebiotics provides a substrate to be fermented by the gut beneficial microbiota. The inclusion of prebiotics in swine diets stimulates the proliferation and metabolic activity of beneficial microbes, contributing to a stable microbial ecosystem [112]. The most widely accepted prebiotics are lactulose, inulin, fructo-oligosaccharides (FOS) and galacto-oligosaccharides (GOS). Oligosaccharides which are short chain prebiotics have also been shown to be a potential alternative to in-feed antibiotics in young piglets due to their effect on the gut microbiota by providing a substrate for beneficial microorganisms [113]. Several studies in swine investigated the effect of prebiotics on the GIT microbiota in piglets around weaning period during previous years. For instance, Konstantinov and co-workers [114, 115] demonstrated that weaning piglets fed diet supplemented with a mix of sugar beet pulp, inulin, lactulose and wheat starch affected the composition of microbiota in the gut. Moreover, the fermentable carbohydrates having the ability to enhance colonic microbial stability and diversity simultaneously enhanced the growth of Lactobacillus sobrius [115]. Lactobacillus sorbius is found to colonize abundantly in the ileum of pigs where it exerts probiotic activity resulting in the prevention of epithelial damage by enteropathogenic E. coli as well as improvement in the daily weight gain of piglets [116,117,118]. In another study, Jiao et al. [119] demonstrated that increasing dose of cello-oligosaccharide supplementation resulted in increased Lactobacillus proportions and a reduction in potential pathogenic groups such as Clostridium in the weaning pigs suggesting the use of prebiotics as a promising approach to alleviate the post-weaning intestinal tract disorders.

Probiotics have been suggested and used as alternatives to antibiotic as a remedy to post-weaning diarrhea and as growth promoters [120,121,122]. Probiotics are direct-fed microbial which when administered in sufficient amounts confer health benefits to the host [123] and consist of organisms such as bacteria cells, yeast cells, or a blend of the two which modulate the gastrointestinal microbiota so as to improve the health of the host. The mechanism of action by probiotic has been suggested to be due to the suppression of pathogens, intestinal microbial communities’ manipulation, and immunomodulation, stimulation of epithelial cell proliferation and differentiation and fortification of the intestinal barrier [124]. Lactobacillus species, Bacillus species, Bifidobacterium, Enterococcus faecium, and E. coli have been developed as probiotics to promote the growth performance, mucosal immunity and epithelial function as well as to inhibit growth of pathogenic bacteria in swine [125,126,127]. The probiotic L. sobrius was found to be effective in the reduction of the E. coli F4 colonization and weight gain improvement of infected piglets [118]. The exopolysaccharide (EPS) secreted by lactic acid bacteria had exclusive properties in modifying the gut microbiota [128]. EPS have shown the potential to act as prebiotics to promote the increase of probiotics, providing support for the adhesion of probiotics in the GIT and their long-term survival, necessary for their effective propagation. It also acts as a source of carbon, helping the growth and colonization of gut bacteria by feeding them nutrients [129]. The supplementation of L. rhamnosus LB1 has been reported to alleviate ETEC’s adverse effects in pigs by improving host immune response, and restoring intestinal integrity [130]. However, in previous study, the dietary supplementation with Lactobacillus rhamnosus GG (originally used for human subjects) reduced the growth performance and impaired the health of Escherichia coli F4-challenged piglets [131]. Thus, the probiotic effects rely on the specific bacterial isolates suggesting the need of host target-specific probiotic strain [132]. Walsh et al. [133] reported that Salmonella-challenged pigs fed probiotics complex consisting of Enterococcus faecium, Bacillus subtilis, and Bacillus licheniformis in drinking water (109 cfu/L for each strain of bacteria) showed no Salmonella in feces at 5 d post challenge. Lu et al. [134] indicated that probiotic complex supplementation including Enterococcus faecium DSM 7134, Bacillus subtilis plus Lactobacillus paracasei regulated the composition of the intestinal microbiota. Naqid et al. [135] demonstrated that Lactobacillus plantarum (B2984) strain supplementation into the feed of weaned piglets orally challenged with S. Typhimurium resulted in significant increase in immunoglobulins concentrations compared to their control counterparts. In our previous study, it was found that by supplementing the diet of Salmonella-challenged weaning pigs with Bacillus-based probiotics (B. subtilis RX7 and B. methylotrophicus C14 strains) boosted the immune system by improving RBC, lymphocyte, IgG, and IgM concentrations in the blood [136]. Beyond the restoration of the microbiota composition, due consideration must be given on how to minimize the effects of perturbed microbiota on the host. Dysbiosis often results in the emergence and outbreak of diseases [137] and increased gut permeability [138], consequently impacting the gut microbiota negatively. To overcome these adverse effects, interventions with probiotics complex with proven anti-inflammatory properties or having the ability to strengthen the gut barrier functions may be a good complementary strategy to improving the microbiota by acting on the host physiology [139,140,141,142]. Based on the reports from different studies, our recently published review work [120] summarized that the impact of pre/probiotics in reducing the stress associated with weaning is due to the antimicrobial effects of these feed additives against the harmful microbes and favoring the growth of beneficial microorganisms, enhancement of mucin expression thereby blocking E. coli invasion or due to the modulatory effect in the intestinal tight junction proteins thereby enhancing intestinal barrier functions as well as immune functions.

Conclusion

This study highlights the importance of microbial stability and reviews the nutritional intervention to maintain the stability of microbiota in GIT. There is immense diversity in swine gut microbiota that varies between individuals and the gut microbiota can fluctuate over time, especially during early development, and under diseased conditions. The gut microbiota and their stability are influenced by host genetics, age, breed, physical activity, microbial infection, stressors, diet quality and types, antibiotics etc. Short-term perturbation resulting from dietary changes may restore microbiota to its original state, but long-term disturbances, such as antibiotic administration, could cause alterations in microbial diversity. Furthermore, the disturbance of the gut microbiota equilibrium through long-term perturbations, such as inflammation, poor feed or antibiotic, could lead to dysbiosis resulting in compromised immunity and consequently causing diseases. Thus, the landscape of stable states for the microbiota and its implications for resilience is an important research direction. To overcome the adverse effect of the perturbations especially due to long term antibiotic use, the nutritional intervention with feed additives could be one of the possible solutions among others. The selected feed additives including dietary fiber, prebiotics, and probiotics were focused in this review. The reported positive impact of these feed additives indicate that these feed additives can be effectively used in maintaining gut microbiome robustness and stability for optimum intestinal health in pigs although some inconsistent effects of probiotics are reported suggesting to select probiotics or probiotic complex that are host target-specific probiotic strain, safe and have proven anti-inflammatory and gut strengthening properties. Furthermore, due consideration must be given to the dose, efficacy as well as safety on the usage of these feed additives.

Availability of data and materials

Not applicable.

Abbreviations

EPA:

Exopolysaccharide

FOS:

Fructo-oligosaccharides

GIT:

Gastrointestinal tract

GOS:

Galacto-oligosaccharides

IgG:

Immunoglobulin G

IgM:

Immunoglobulin M

NSP:

Non-starch polysaccharides

RBC:

Red blood cell

SCFA:

Short chain fatty acid

References

  1. Hill JE, Seipp RP, Betts M, Hawkins L, Van Kessel AG, Crosby WL, et al. Extensive profiling of a complex microbial community by high-throughput sequencing. Appl Environ Microbiol. 2002;68:3055–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Leser TD, Amenuvor JZ, Jensen TK, Lindecrona RH, Boye M, Moller K. Culture-independent analysis of gut bacteria: the pig gastrointestinal tract microbiota revisited. Appl Environ Microbiol. 2002;68:673–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Pryde SE, Richardson AJ, Stewart CS, Flint HJ. Molecular analysis of the microbial diversity present in the colonic wall, colonic lumen, and cecal lumen of a pig. Appl Environ Microbiol. 1999;65:5372–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Gaskins HR. Intestinal bacteria and their influence on swine growth. In: Lewis AJ, Southern LL, editors. Swine nutrition. Boca Raton: CRC Press; 2001. p. 585–608.

    Google Scholar 

  5. Ewing W, Cole D. The living gut: an introduction to micro-organisms in nutrition. Dungannon: Context; 1994. p. 220.

    Google Scholar 

  6. Jensen BB, Jorgensen H. Effect of dietary fiber on microbial activity and microbial gas production in various regions of the gastrointestinal tract of pigs. Appl Environ Microbiol. 1994;60:1897–904.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Berg RD. The indigenous gastrointestinal microflora. Trends Microbiol. 1996;4:430–35.

    Article  CAS  PubMed  Google Scholar 

  8. Isolauri E, Sutas Y, Kankaanpaa P, Arvilommi H, Salminen S. Probiotics: effects on immunity. Am J Clin Nutr. 2001;73:444S-50S.

    Article  Google Scholar 

  9. Maynard CL, Elson CO, Hatton RD, Weaver CT. Reciprocal interactions of the intestinal microbiota and immune system. Nature. 2012;489:231–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Sommer F, Bäckhed F. The gut microbiota-masters of host development and physiology. Nat Rev Microbiol. 2013;11:227–38.

    Article  CAS  PubMed  Google Scholar 

  11. Tremaroli V, Backhed F. Functional interactions between the gut microbiota and host metabolism. Nature. 2012;489:242–49.

    Article  CAS  PubMed  Google Scholar 

  12. Erb Downward JR, Falkowski NR, Mason KL, Muraglia R, Huffnagle GB. Modulation of post-antibiotic bacterial community reassembly and host response by Candida albicans. Sci Rep. 2013;3:2191.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Ott SJ, Kühbacher T, Musfeldt M, Rosenstiel P, Hellmig S, Rehman A, et al. Fungi and inflammatory bowel diseases: Alterations of composition and diversity. Scand J Gastroenterol. 2008;43(7):831–41.

    Article  CAS  PubMed  Google Scholar 

  14. Iliev ID, Leonardi I. Fungal dysbiosis: immunity and interactions at mucosal barriers. Nat Rev Immunol. 2017;17(10):635–46.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Li XV, Leonardi I, Iliev ID. Gut mycobiota in immunity and inflammatory disease. Immunity. 2019;50(6):1365–79. https://doi.org/10.1016/j.immuni.2019.05.023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Mukherjee PK, Sendid B, Hoarau G, Colombel JF, Poulain D, Ghannoum MA. Mycobiota in gastrointestinal diseases. Nat Rev Gastroenterol Hepatol. 2015;12:77–87.

    Article  PubMed  Google Scholar 

  17. Weissenbacher-Lang C, Kureljusic B, Nedorost N, Matula B, Schiessl W, Stixenberger D, et al. Retrospective analysis of bacterial and viral co-infections in pneumocystis spp. positive lung samples of Austrian pigs with pneumonia. PLoS ONE. 2016;11:e0158479.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Iliev ID, Cadwell K. Effects of Intestinal fungi and viruses on immune responses and inflammatory bowel diseases. Gastroenterol. 2021;160(4):1050–66. https://doi.org/10.1053/j.gastro.2020.06.100.

    Article  CAS  Google Scholar 

  19. Blasi B, Sipos W, Knecht C, Durlinger S, Ma L, Cisse OH, et al. Pneumocystis spp. in pigs: a longitudinal quantitative study and co-infection assessment in Austrian farms. J Fungi. 2022;8:43. https://doi.org/10.3390/jof8010043.

    Article  Google Scholar 

  20. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489(7415):220–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hooks KB, O’Malley MA. Dysbiosis and its discontents. mBio. 2017;8:e01492–17.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Bello MGD, Knight R, Gilbert JA, Blaser MJ. Preserving microbial diversity. Science. 2018;362:33–4.

    Article  PubMed  Google Scholar 

  23. Jones S. Trends in microbiome research. Nat Biotechnol. 2013;31:277.

    Article  CAS  Google Scholar 

  24. Liu P, Zhao J, Wang W, Guo P, Lu W, Wang C. Dietary corn bran altered the diversity of microbial communities and cytokine production in weaned pigs. Front Microbiol. 2018;9:2090.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Yu D, Zhu W, Hang S. Effects of long-term dietary protein restriction on intestinal morphology, digestive enzymes, gut hormones, and colonic microbiota in pigs. Animals. 2019;9:180.

    Article  PubMed Central  Google Scholar 

  26. Lin S, Yang X, Yuan P, Yang J, Wang P, Zhong H. Undernutrition shapes the gut microbiota and bile acid profile in association with altered gut-liver FXR signaling in weaning pigs. J Agric Food Chem. 2019;67:3691–701.

    Article  CAS  PubMed  Google Scholar 

  27. Papadomichelakis G, Zoidis E, Mountzouris KC, Lippas T, Fegeros K. Glycerine kinase gene expression, nutrient digestibility and gut microbiota composition in post-weaned pigs fed diets with increasing crude glycerine levels. Anim Feed Sci Technol. 2012;177:247–52.

    Article  CAS  Google Scholar 

  28. Huang L, Ren P, Ouyang Z, Wei T, Kong X, Li T. Effect of fermented feed on growth performance, holistic metabolism and fecal microbiota in weanling piglets. Anim Feed Sci Technol. 2020;266:114505. https://doi.org/10.1016/j.anifeedsci.2020.114505.

    Article  CAS  Google Scholar 

  29. Ramayo-Caldas Y, Mach N, Lepage P, Levenez F, Denis C, Lemonnier G, et al. Phylogenetic network analysis applied to pig gut microbiota identifies an ecosystem structure linked with growth traits. ISME J. 2016;10:2973–77.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Yan HL, Diao H, Xiao Y, Li WX, Yu B, He J, et al. Gut microbiota can transfer fiber characteristics and lipid metabolic profiles of skeletal muscle from pigs to germ-free mice. Sci Rep. 2016;6:31786.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Knecht D, Cholewinska P, Jankowska-Makosa A, Czyz K. Development of swine’s digestive tract microbiota and its relation to production indices‐a review. Animals. 2020;10:527.

    Article  PubMed Central  Google Scholar 

  32. Xiao L, Estellé J, Kiilerich P, Ramayo-Caldas Y, Xia ZK, Feng Q, et al. A reference gene catalogue of the pig gut microbiome. Nat Microbiol. 2016;1:16161.

    Article  CAS  PubMed  Google Scholar 

  33. Salonen A, Salojarvi J, Lahti L, de Vos WM. The adult intestinal core microbiota is determined by analysis depth and health status. Clin Microbiol Infec. 2012;18:16–20.

    Article  CAS  Google Scholar 

  34. Lindberg JE. Fiber effects in nutrition and gut health in pigs. J Anim Sci Biotechnol. 2014;5:15.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Huang SM, Wu ZH, Li TT, Liu C, Han DD, Tao SY, et al. Perturbation of the lipid metabolism and intestinal inflammation in growing pigs with low birth weight is associated with the alterations of gut microbiota. Sci Total Environ. 2020;719:137382.

    Article  CAS  PubMed  Google Scholar 

  36. Katouli M, Lund A, Wallgren P, Kuhn I, Soderlind O, Mollby R. Metabolic fingerprinting and fermentative capacity of the intestinal flora of pigs during pre- and post-weaning periods. J Appl Microbiol. 1997;83:147–54.

    Article  CAS  PubMed  Google Scholar 

  37. Chen W, Mi J, Lv N, Gao J, Cheng J, Wu R, et al. Lactation stage-dependency of the sow milk microbiota. Front Microbiol. 2018;9:945.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Swords WE, Wu CC, Champlin FR, Buddington RK. Postnatal changes in selected bacterial groups of the pig colonic microflora. Biol Neonate. 1993;63(3):191–200.

    Article  CAS  PubMed  Google Scholar 

  39. Petri D, Hill JE, van Kessel AG. Microbial succession in the gastrointestinal tract (GIT) of the pre-weaned pig. Livest Sci. 2010;133:107–9.

    Article  Google Scholar 

  40. Drasar BS. The bacterial flora of the stomach and small intestine. Gastroenterol Clin Biol. 1989;13:18B–20B.

    CAS  PubMed  Google Scholar 

  41. Inoue R, Tsukahara T, Nakanishi N, Ushida K. Development of the intestinal microbiota in the piglet. J Gen Appl Microbiol. 2005;51:257–65.

    Article  CAS  PubMed  Google Scholar 

  42. Konstantinov SR, Awati AA, Williams BA, Miller BG. Jones P, Stokes CR, et al. Post-natal development of the porcine microbiota composition and activities. Environ Microbiol. 2006;8:1191–99.

    Article  CAS  PubMed  Google Scholar 

  43. Lallès JP, Boudry G, Favier C, Le Floc’h N, Lurona I, Montagne L, et al. Gut function and dysfunction in young pigs: physiology. Anim Res. 2004;53:301–16.

    Article  Google Scholar 

  44. Montagne L, Boudry G, Favier C, Le Huerou-Luron I, Lalles JP, Seve B. Main intestinal markers associated with the changes in gut architecture and function in piglets after weaning. Br J Nutr. 2007;97:45–57.

    Article  CAS  PubMed  Google Scholar 

  45. Wallgren P, Melin L. Weaning systems in relation to disease. In: Varley MA, Wiseman J, editors. The weaner pig: nutrition and management. Wallington, Oxon: CABI Publishing; 2001. p. 309–16.

    Google Scholar 

  46. Chen L, Xu Y, Chen X, Fang C, Zhao L, Chen F. The maturing development of gut microbiota in commercial piglets during the weaning transition. Front Microbiol. 2017;8:1688.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Gresse R, Chaucheyras-Durand F, Fleury MA, de Wiele TV, Forano E, Blanquet-Diot S. Gut Microbiota dysbiosis in post-weaning piglets: Understanding the keys to health. Trends Microbiol. 2017;25:851–73.

    Article  CAS  PubMed  Google Scholar 

  48. Isaacson R, Kim HB. The intestinal microbiome of the pig. Anim Health Res Rev. 2012;13(1):100–9.

    Article  PubMed  Google Scholar 

  49. Looft T, Allen HK, Cantarel BL, Levine UY, Bayles DO, Alt DP, et al. Bacteria, phages and pigs: the effects of in-feed antibiotics on the microbiome at different gut locations. ISME J. 2014;8(8):1566–76. https://doi.org/10.1038/ismej.2014.12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Holman DB, Brunelle BW, Trachsel J, Allen HK. Meta-analysis to define a core microbiota in the swine gut. mSystems. 2017;2:e00004e17.

    Article  Google Scholar 

  51. Geng S, Cheng S, Li Y, Wen Z, Ma X, Jiang X, et al. Faecal microbiota transplantation reduces susceptibility to epithelial injury and modulates tryptophan metabolism of the microbial community in a piglet model. J Crohns Colitis. 2018;12:1359e74.

    Google Scholar 

  52. Kang C, Wang B, Kaliannan K, Wang X, Lang H, Hui S, et al. Gut microbiota mediates the protective effects of dietary capsaicin against chronic low-grade inflammation and associated obesity induced by high-fat diet. mBio. 2017;8:e00470-17.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Li Y, Wang X, Wang X, Wang J, Zhao J. Life-long dynamics of the swine gut microbiome and their implications in probiotics development and food safety. Gut Microbes. 2020;11:1824–32.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Li N, Huang S, Jiang L, Wang W, Li T, Zuo B, et al. Differences in the gut microbiota establishment and metabolome characteristics between low-and normal-birth-weight piglets during early-life. Front Microbiol. 2018;9:1798.

    Article  PubMed  PubMed Central  Google Scholar 

  55. Bian G, Ma S, Zhu Z, Su Y, Zoetendal EG, Mackie R, et al. Age, introduction of solid feed and weaning are more important determinants of gut bacterial succession in piglets than breed and nursing mother as revealed by a reciprocal cross-fostering model. Environ Microbiol. 2016;18:1566e77.

    Article  Google Scholar 

  56. Wang X, Tsai T, Deng F, Wei X, Chai J, Knapp J, et al. Longitudinal investigation of the swine gut microbiome from birth to market reveals stage and growth performance associated bacteria. Microbiome. 2019;7:109.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Meng Q, Luo Z, Cao C, Sun S, Ma Q, Li Z, et al. Weaning alters intestinal gene expression involved in nutrient metabolism by shaping gut microbiota in pigs. Front Microbiol. 2020;11:694.

    Article  PubMed  PubMed Central  Google Scholar 

  58. He M, Gao J, Wu J, Zhou Y, Fu H, Ke S, et al. Host gender and androgen levels regulate gut bacterial taxa in pigs leading to sex-biased serum metabolite profiles. Front Microbiol. 2019;10:1359.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Garcia-Gomez E, Gonzalez-Pedrajo B, Camacho-Arroyo I. Role of sex steroid hormones in bacterial-host interactions. Biomed Res Int. 2013;2013:928290.

    Article  PubMed  Google Scholar 

  60. Cheng PH, Liang JB, Wu YB, Wang Y, Tufarelli V, Laudadio V, et al. In vitro fermentative capacity of swine large intestine: comparison between native Lantang and commercial Duroc breeds. Anim Sci J. 2017;88(8):1141–48.

    Article  CAS  PubMed  Google Scholar 

  61. Guevarra RB, Kim J, Nguyen SG, Unno T. Comparison of fecal microbial communities between white and black pigs. J Appl Biol Chem. 2015;58:369–75.

    Article  Google Scholar 

  62. Diao H, Yan HL, Xiao Y, Yu B, Yu J, He J, et al. Intestinal microbiota could transfer host gut characteristics from pigs to mice. BMC Microbiol. 2016;16:238. https://doi.org/10.1186/s12866-016-0851-z.

    Google Scholar 

  63. Bergamaschi M, Maltecca C, Schillebeeckx C, McNulty NP, Schwab C, Shull C, et al. Heritability and genome wide association of swine gut microbiome features with growth and fatness parameters. Sci Rep. 2020;10:10134.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wang H, Xu R, Zhang H, Su Y, Zhu W. Swine gut microbiota and its interaction with host nutrient metabolism. Anim Nutr. 2020;6:410–20.

    Article  PubMed  PubMed Central  Google Scholar 

  65. Quan J, Cai G, Ye J, Yang M, Ding R, Wang X, et al. A global comparison of the microbiome compositions of three gut locations in commercial pigs with extreme feed conversion ratios. Sci Rep. 2018;8:4536. https://doi.org/10.1038/s41598-018-22692-0.

    Article  Google Scholar 

  66. Maradiaga N, Aldridge B, Zeineldin M, Lowe J. Gastrointestinal microbiota and mucosal immune gene expression in neonatal pigs reared in a cross-fostering model. Microb Pathog. 2018;121:27–39.

    Article  CAS  PubMed  Google Scholar 

  67. Yeoman CJ, Ishaq SL, Bichi SL, Olivo SK, Lowe J, Aldridge BM. Biogeographical differences in the influence of maternal microbial sources on the early successional development of the bovine neonatal gastrointestinal tract. Sci Rep. 2018;8:3197. https://doi.org/10.1038/s41598-018-21440-8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Kim J, Nguyen SG, Guevarra RB, Lee I, Unno T. Analysis of swine fecal microbiota at various growth stages. Arch Microbiol. 2015;197:753–59.

    Article  CAS  PubMed  Google Scholar 

  69. Adhikari B, Kim SW, Kwon YM. Characterization of microbiota associated with digesta and mucosa in different regions of gastrointestinal tract of nursery pigs. Intl J Mol Sci. 2019;20(7):1630.

    Article  CAS  Google Scholar 

  70. Relman DA. The human microbiome: ecosystem resilience and health. Nutr Rev. 2012;70:2–9.

    Article  Google Scholar 

  71. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature. 2014;505:559–63.

    Article  CAS  PubMed  Google Scholar 

  72. Faith JJ, Guruge JL, Charbonneau M, Subramanian S, Seedorf H, Goodman AL, et al. The long-term stability of the human gut microbiota. Science. 2013;341:1237439.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Ingrisch J, Bahn M. Towards a comparable quantification of resilience. Trends Ecol Evol. 2018;33:251–59.

    Article  PubMed  Google Scholar 

  74. Duvallet C, Gibbons SM, Gurry T, Irrizary RA, Alm EJ. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat Commun. 2017;8:1784.

    Article  PubMed  PubMed Central  Google Scholar 

  75. Heiman ML, Greenway FL. A healthy gastrointestinal microbiome is dependent on dietary diversity. Mol Metabol. 2016;5:317–20.

    Article  CAS  Google Scholar 

  76. Lloyd-Price J, Abu-Ali G, Huttenhower C. The healthy human microbiome. Genome Med. 2016;8:51.

    Article  PubMed  PubMed Central  Google Scholar 

  77. Tiffany CR, Bäumler AJ. Dysbiosis: from fiction to function. Am J Physiol Gastrointest Liver Physiol. 2019;317:G602–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Kriss M, Hazleton KZ, Nusbacher NM, Martin CG, Lozupone CA. Low diversity gut microbiota dysbiosis: drivers, functional implications and recovery. Curr Opin Microbiol. 2018;44:34–40.

    Article  PubMed  PubMed Central  Google Scholar 

  79. Chen C, Zhou Y, Fu H, Xiong X, Fang S, Jiang H, et al. Expanded catalog of microbial genes and metagenome-assembled genomes from the pig gut microbiome. Nat Commun. 2021;12:1106. https://doi.org/10.1038/s41467-021-21295-0.

    Google Scholar 

  80. Sassone-Corsi M, Nuccio SP, Liu H, Hernandez D, Vu CT, Takahasi AA, et al. Microcins mediate competition among. Enterobacteriaceae in the inflamed gut. Nature. 2016;540:280–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Litvak Y, Byndloss MX, Tsolis RM, Baumler AJ. Dysbiotic Proteobacteria expansion: a microbial signature of epithelial dysfunction. Curr Opin Microbiol. 2017;39:1–6. https://doi.org/10.1016/j.mib.2017.07.003.

    Article  CAS  PubMed  Google Scholar 

  82. Bhalodi AA, van Engelen TSR, Virk HS, Wiersinga WJ. Impact of antimicrobial therapy on the gut microbiome. J Antimicrob Chemother. 2019;74:i6–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Zeineldin M, Aldridge B, Lowe J. Antimicrobial effects on swine gastrointestinal microbiota and their accompanying antibiotic resistome. Front Microbiol. 2019;10:1035.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Gresse R, Chaucheyras-Durand F, Fleury MA, Van De Wiele T, Forano E, Blanquet-Diot S. Gut microbiota dysbiosis in post-weaning piglets: understanding the keys to health. Trends Microbiol. 2017;25:851–73. https://doi.org/10.1016/j.tim.2017.05.004.

    Article  CAS  PubMed  Google Scholar 

  85. Jo HE, Kwon MS, Whon TW, Kim DW, Yun M, Lee J, et al. Alteration of gut microbiota after antibiotic exposure in finishing swine. Front Microbiol. 2021;12:596002. https://doi.org/10.3389/fmicb.2021.596002.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Schoenhofen IC, Vinogradov E, Whitfield DM, Brisson JR, Logan SM. The CMP-legionaminic acid pathway in Campylobacter: Biosynthesis involving novel GDP-linked precursors. Glycobiology. 2009;19:715–25. https://doi.org/10.1093/glycob/cwp039.

    Article  CAS  PubMed  Google Scholar 

  87. Ferrer M, Méndez-García C, Rojo D, Barbas C, Moya A. Antibiotic use and microbiome function. Biochem Pharmacol. 2017;134:114–26. https://doi.org/10.1016/j.bcp.2016.09.007.

    Article  CAS  PubMed  Google Scholar 

  88. Neuman H, Forsythe P, Uzan A, Avni O, Koren O. Antibiotics in early life: dysbiosis and the damage done. FEMS Microbiol Rev. 2018;42:489–99. https://doi.org/10.1093/femsre/fuy018.

    Article  CAS  PubMed  Google Scholar 

  89. Gao K, Pi Y, Peng Y, Mu CL, Zhu WY. Time-course responses of ileal and fecal microbiota and metabolite profiles to antibiotics in cannulated pigs. Appl Microbiol Biotechnol. 2018;102:2289–99. https://doi.org/10.1007/s00253-018-8774-2.

    Article  CAS  PubMed  Google Scholar 

  90. Li J, Yang K, Ju T, Ho T, McKay CA, Gao Y, et al. Early life antibiotic exposure affects pancreatic islet development and metabolic regulation. Sci Rep. 2017;7:41778. https://doi.org/10.1038/srep41778.

    Article  CAS  Google Scholar 

  91. Mu C, Yang Y, Su Y, Zoetendal EG, Zhu W. Differences in microbiota membership along the gastrointestinal tract of piglets and their differential alterations following an early-life antibiotic intervention. Front Microbiol. 2017;8:797.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Kim HB, Borewicz K, White BA, Singer RS, Sreevatsan S, Tu ZJ, et al. Microbial shifts in the swine distal gut in response to the treatment with antimicrobial growth promoter, tylosin. Proc Natl Acad Sci. 2012;109:15485–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Holman DB, Chénier MR. Temporal changes and the effect of subtherapeutic concentrations of antibiotics in the gut microbiota of swine. FEMS Microbiol Ecol. 2014;90:599–608. https://doi.org/10.1111/1574-6941.12419.

    Article  CAS  PubMed  Google Scholar 

  94. Lewis BB, Buffie CG, Carter RA, Leiner I, Toussaint NC, Miller LC, et al. Loss of microbiota-mediated colonization resistance to Clostridium difficile infection with oral vancomycin compared with metronidazole. J Infect Dis. 2015;212:1656–65. https://doi.org/10.1093/infdis/jiv256.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Allen HK, Levine UY, Looft T, Bandrick M, Casey TA. Treatment, promotion, commotion: antibiotic alternatives in food-producing animals. Trends Microbiol. 2013;21:114–19. https://doi.org/10.1016/j.tim.2012.11.001.

    Article  CAS  PubMed  Google Scholar 

  96. Allen HK, Trachsel J, Looft T, Casey TA. Finding alternatives to antibiotics. Ann NY Acad Sci. 2014;1323:91–100. https://doi.org/10.1111/nyas.12468.

    Article  PubMed  Google Scholar 

  97. Czaplewski L, Bax R, Clokie M, Dawson M, Fairhead H, Fischetti VA, et al. Alternatives to antibiotics-a pipeline portfolio review. Lancet Infect Dis. 2016;16:239–51. https://doi.org/10.1016/S1473-3099(15)00466-1.

    Article  CAS  PubMed  Google Scholar 

  98. Conway PL. Function and regulation of the gastrointestinal microbiota of the pig. In: Souffrant WB, Hagemeister H, editors. Proceedings of the VIth international symposium on digestive physiology in Pigs. EAAP, Publication, Dummerstof; 1994. p. 231–40.

  99. Jha R, Fouhse JM, Tiwari UP, Li L, Willing BP. Dietary fiber and intestinal health of monogastric animals. Front Vet Sci. 2019;6:48. https://doi.org/10.3389/fvets.2019.00048.

    Article  PubMed  PubMed Central  Google Scholar 

  100. Jha R, Berrocoso JFD. Dietary fiber and protein fermentation in the intestine of swine and their interactive effects on gut health and on the environment: a review. Anim Feed Sci Technol. 2016;212:18–26. https://doi.org/10.1016/j.anifeedsci.2015.12.002.

    Article  CAS  Google Scholar 

  101. Montagne L, Pluske JR, Hampson DJ. A review of interactions between dietary fibre and the intestinal mucosa, and their consequences on digestive health in young non-ruminant animals. Anim Feed Sci Tech. 2003;108:95–117.

    Article  Google Scholar 

  102. Pu G, Li P, Du T, Niu Q, Fan L, Wang H, et al. Adding appropriate fiber in diet increases diversity and metabolic capacity of distal gut microbiota without altering fiber digestibility and growth rate of finishing pig. Front Microbiol. 2020;11:533. https://doi.org/10.3389/fmicb.2020.00533.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Cummings JH. In: Gibson GR, Macfarlanne GT, editors. Short chain fatty acids, in human colonic bacteria. Role in nutrition, physiology and pathology. Boca Raton: CRC Press; 1995. p. 101–30.

    Google Scholar 

  104. Edwards CA. Dietary fibre, fermentation and the colon. In: Cherbut C, Barry JL, Lairon D, Durand M, editors. Dietary fibre: mechanisms of action in human physiology and metabolism. Paris: John Libbey Eurotex; 1995. p. 51–60.

    Google Scholar 

  105. Topping DL, Clifton PM. Short-chain fatty acids and human colonic function: Roles of resistant starch and nonstarch polysaccharides. Physiol Rev. 2001;81:1031–64.

    Article  CAS  PubMed  Google Scholar 

  106. Cheng W, Lu J, Li B, Lin W, Zhang Z, Wei X, et al. Effect of functional oligosaccharides and ordinary dietary fiber on intestinal microbiota diversity. Front Microbiol. 2017;8:1750.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Wu G, Tang X, Fan C, Wang L, Shen W, Ren S, et al. Gastrointestinal tract and dietary fiber driven alterations of gut microbiota and metabolites in Durco × Bamei crossbred pigs. Front Nutr. 2022;8:806646. https://doi.org/10.3389/fnut.2021.806646.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  108. Tang X, Zhang L, Fan C, Wang L, Fu H, Ren S, et al. Dietary fiber influences bacterial community assembly processes in the gut microbiota of Durco × Bamei crossbred pig. Front Microbiol. 2021;12:688554. https://doi.org/10.3389/fmicb.2021.688554.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Hryckowian AJ, Van Treuren W, Smits SA, Davis NM, Gardner JO, Bouley DM, et al. Microbiota-accessible carbohydrates suppress Clostridium difficile infection in a murine model. Nat Microbiol. 2018;3:662–69. https://doi.org/10.1038/s41564-018-0150-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Bindels LB, Delzenne NM, Cani PD, Walter J. Opinion. Towards a more comprehensive concept for prebiotics. Nat Rev Gastroenterol. 2015;12:303–10.

    Article  CAS  Google Scholar 

  111. Gibson GR, Probert HM, Loo JV, Rastall RA, Roberfroid MB. Dietary modulation of the human colonic microbiota: Updating the concept of prebiotics. Nutr Res Rev. 2004;17(2):259–75.

    Article  CAS  PubMed  Google Scholar 

  112. Metzler B, Bauer E, Mosenthin R. Microflora management in the gastrointestinal tract of piglets. Asian Australas J Anim. 2005;18:1353–62. https://doi.org/10.5713/ajas.2005.1353.

    Article  Google Scholar 

  113. Van Laere KMJ, Bosveld M, Schols HA, Beldmen G, Voragen AGJ. Fermentative degradation of plant cell wall derived oligosaccharides by intestinal bacteria. In: Hartemink R, editor. Proceedings of symposium on non- digestible oligosaccharides: Healthy food for the colon? Wageningen: Wageningen Graduate School VLAG; 1997. p. 37–46.

  114. Konstantinov SR, Zhu WY, Williams BA, Tamminga S, de Vos WM, Akkermans ADL. Effect of fermentable carbohydrates on faecal bacterial communities as revealed by DGGE analysis of 16S rDNA. FEMS Microbiol Ecol. 2003;43:225–35.

    Article  CAS  PubMed  Google Scholar 

  115. Konstantinov SR, Awati A, Smidt H, Williams BA, Akkermans ADL, de Vos WM. Specific response of a novel and abundant Lactobacillus amylovorus-like phylotype to dietary prebiotics in the ileum and colon of weaning piglets. Appl Environ Microbiol. 2004;70:3821–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Roselli M, Finamore A, Britti MS, Konstantinov SR, Smidt H, de Vos WM, et al. The novel porcine Lactobacillus sobrius strain protects intestinal cells from enterotoxigenic Escherichia coli K88 infection and prevents membrane barrier damage. J Nutr. 2007;137:2709–16.

    Article  CAS  PubMed  Google Scholar 

  117. Konstantinov SR, Poznanski E, Fuentes S, Akkermans ADL, Smidt H, de Vos WM. Lactobacillus sobrius sp. nov., a novel isolate abundant in the intestine of weaning piglets. Intl J Syst Evol Microbial. 2006;56:29–32.

    Article  CAS  Google Scholar 

  118. Konstantinov SR, Smidt H, Akkermans ADL, Casini L, Trevisi P, Mazzoni M, et al. Feeding of Lactobacillus sobrius reduces Escherichia coli F4 levels in the gut and promotes growth of infected piglets. FEMS Microbiol Ecol. 2008;66:599–607.

    Article  CAS  PubMed  Google Scholar 

  119. Jiao LF, Song ZH, Ke YL, Xiao K, Hu CH, Shi B. Cello-oligosaccharide influences intestinal microflora, mucosal architecture and nutrient transport in weaned pigs. Anim Feed Sci Technol. 2014;195:85–91.

    Article  CAS  Google Scholar 

  120. Upadhaya SD, Kim IH. The impact of weaning stress on gut health and the mechanistic aspects of several feed additives contributing to improved gut health function in weanling piglets–A Review. Animals. 2021;11(8):2418. https://doi.org/10.3390/ani11082418.

    Article  Google Scholar 

  121. Pettigrew JE. Reduced use of antibiotic growth promoters in diets fed to weanling pigs: dietary tools, part 1. Anim Biotechnol. 2006;17:207–15. https://doi.org/10.1080/10495390600956946.

    Article  CAS  PubMed  Google Scholar 

  122. Stein HH, Kil DY. Reduced use of antibiotic growth promoters in diets fed to weanling pigs: dietary tools, part 2. Anim Biotechnol. 2006;17:217–31. https://doi.org/10.1080/10495390600957191.

    Article  CAS  PubMed  Google Scholar 

  123. FAO/WHO. Food and Agriculture Organization of the United Nations/World Health Organization working group joint report on drafting guidelines for the evaluation of probiotics in food. London Ontario, Canada; 2002. https://www.fao.org/3/a0512e/a0512e.pdf. (Accessed 24 Mar 2022).

  124. Thomas C, Versalovic J. Probiotics-host communication: modulation of signaling pathways in the intestine. Gut Microbes. 2010;1:148–63.

    Article  PubMed  PubMed Central  Google Scholar 

  125. Setia A, Bhandari SK, House JD, Nyachoti CM, Krause DO. Development and in vitro evaluation of an Escherichia coli probiotic able to inhibit the growth of pathogenic Escherichia coli K88. J Anim Sci. 2009;87:2005–12.

    Article  CAS  PubMed  Google Scholar 

  126. Lähteinen T, Malinen E, Koort JM, Mertaniemi-Hannus U, Hankimo T, Karikoski N, et al. Probiotic properties of Lactobacillus isolates originating from porcine intestine and feces. Anaerobe. 2010;16:293–300.

    Article  PubMed  Google Scholar 

  127. Klingspor S, Martens H, Caushi D, Twardziok S, Aschenbach JR, Lodemann U. Characterization of the effects of Enterococcus faecium on intestinal epithelial transport properties in piglets. J Anim Sci. 2013;91:1707–18.

    Article  CAS  PubMed  Google Scholar 

  128. Angelin J, Kavitha M. Exopolysaccharides from probiotic bacteria and their health potential. Int J Biol Macromol. 2020;162:853–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Kumar AS, Mody K, Jha B. Bacterial exopolysaccharides—A perception. J Basic Microbiol. 2007;47:103–17.

    Article  CAS  PubMed  Google Scholar 

  130. Wu T, Shi Y, Zhang Y, Zhang M, Zhang L, Ma Z, et al. Lactobacillus rhamnosus LB1 alleviates enterotoxigenic Escherichia coli-induced adverse effects in piglets by improving host immune response and anti-oxidation stress and restoring intestinal integrity. Front Cell Infect Microbiol. 2021;11:724401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Trevisi P, Casini L, Coloretti F, Mazzoni M, Merialdi G, Bosi P. Dietary addition of Lactobacillus rhamnosus GG impairs the health of Escherichia coli F4-challenged piglets. Animal. 2011;5:1354–60.

    Article  CAS  PubMed  Google Scholar 

  132. de Lange CFM, Pluske J, Gong J, Nyachoti CM. Strategic use of feed ingredients and feed additives to stimulate gut health and development in young pigs. Livest Sci. 2010;134:124–34.

    Article  Google Scholar 

  133. Walsh MC, Rostagno MH, Gardiner GE, Sutton AL, Richert BT, Radcliffe JS. Controlling infection in weanling pigs through water delivery of direct-fed microbials or organic acids. Part I: effects on growth performance, microbial populations and immune status. J Anim Sci. 2012;90:261–71.

    Article  CAS  PubMed  Google Scholar 

  134. Lu X, Zhang M, Zhao L, Ge K, Wang Z, Jun L, et al. Growth performance and post-weaning diarrhea in piglets fed a diet supplemented with probiotic complexes. J Microbiol Biotechnol. 2018;28:1791–99.

    Article  CAS  PubMed  Google Scholar 

  135. Naqid IA, Owen JP, Maddison BC, Gardner DS, Foster N, Tchorzewska MA, et al. Prebiotic and probiotic agents enhance antibody-based immune responses to Salmonella Typhimurium infection in pigs. Anim Feed Sci Technol. 2015;201:57–65.

    Article  CAS  Google Scholar 

  136. Upadhaya SD, Shanmugam SK, Kang DK, Kim IH. Preliminary assessment on potentials of probiotic B. subtilis RX7 and B. methylotrophicus C14 strains as an immune modulator in Salmonella-challenged weaned pigs. Trop Anim Health Prod. 2017;49(5):1065–70.

    Article  PubMed  Google Scholar 

  137. Blander JM, Longman RS, Iliev ID, Sonnenberg GF, Artis D. Regulation of inflammation by microbiota interactions with the host. Nat Immunol. 2017;18:851–60. https://doi.org/10.1038/ni.3780.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Jandhyala SM, Talukdar R, Subramanyam C, Vuyyuru H, Sasikala M, Nageshwar Reddy D. Role of the normal gut microbiota. World J Gastroenterol. 2015;21:8787–803.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. Karczewski J, Troost FJ, Konings I, Dekker J, Kleerebezem M, Brummer RJ, et al. Regulation of human epithelial tight junction proteins by Lactobacillus plantarum in vivo and protective effects on the epithelial barrier. Am J Physiol Gastrointest Liver Physiol. 2010;298:G851-59. https://doi.org/10.1152/ajpgi.00327.2009.

    Article  CAS  PubMed  Google Scholar 

  140. Vieira AT, Fukumori C, Ferreira CM. New insights into therapeutic strategies for gut microbiota modulation in inflammatory diseases. Clin Transl Immunol. 2016;5:e87.

    Article  Google Scholar 

  141. Mendes MCS, Paulino DS, Brambilla SR, Camargo JA, Persinoti GF, Carvalheira JBC. Microbiota modification by probiotic supplementation reduces colitis associated colon cancer in mice. World J Gastroenterol. 2018;24:1995–2008.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Capurso L. Thirty years of Lactobacillus rhamnosus GG: a review. J Clin Gastroenterol. 2019;53 Suppl 1:1–41.

    Article  Google Scholar 

Download references

Funding

This study was supported by the Department of Animal Resources & Science through the Research Focused Department Promotion Project as a part of the University Innovation Support Program for Dankook University in 2022.

Author information

Authors and Affiliations

Authors

Contributions

SDU conceptualized and designed the review research, searched literature and wrote the original manuscript. IHK conceptualized, reviewed and edited the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to In Ho Kim.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that no competing interests exist. The manuscript has not been published previously

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Upadhaya, S.D., Kim, I.H. Maintenance of gut microbiome stability for optimum intestinal health in pigs – a review. J Animal Sci Biotechnol 13, 140 (2022). https://doi.org/10.1186/s40104-022-00790-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40104-022-00790-4

Keywords