Next Article in Journal
Constant Rate Infusion of Lidocaine, Tumescent Anesthesia and Their Combination in Dogs Undergoing Unilateral Mastectomy
Next Article in Special Issue
Supplemental Effects of Functional Oils on the Modulation of Mucosa-Associated Microbiota, Intestinal Health, and Growth Performance of Nursery Pigs
Previous Article in Journal
Sequential Modulation of the Equine Fecal Microbiota and Fibrolytic Capacity Following Two Consecutive Abrupt Dietary Changes and Bacterial Supplementation
Previous Article in Special Issue
Impact of Supplementary Microbial Additives Producing Antimicrobial Substances and Digestive Enzymes on Growth Performance, Blood Metabolites, and Fecal Microflora of Weaning Pigs
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Weaning Induced Gut Dysfunction and Nutritional Interventions in Nursery Pigs: A Partial Review

Department of Animal Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72701, USA
*
Author to whom correspondence should be addressed.
Animals 2021, 11(5), 1279; https://doi.org/10.3390/ani11051279
Submission received: 19 March 2021 / Revised: 27 April 2021 / Accepted: 27 April 2021 / Published: 29 April 2021
(This article belongs to the Collection The Weaned Pig: Nutrition and Management)

Abstract

:

Simple Summary

The beneficial and functional roles of the gut microbiota in maintaining host health are becoming clearer. In general, the swine intestinal bacterial population is predominated by two phyla, Firmicutes and Bacteroidetes, followed by Proteobacteria, Actinobacteria, and Spirochaetes, among others. This general profile is relatively stable, although variations exhibit below the phylum level when the gut microbiota experiences dysbiosis. At weaning, dietary and environmental changes often result in gut microbiota dysbiosis, which is frequently associated with post-weaning diarrhea and enteric infections. A healthy gut microbiota possesses specific capacities involving nutrient metabolism, immunomodulation, controlling inflammation, and maintaining intestinal barrier function and structure. Manipulating the gut microbiota through diet is a useful strategy to attenuate weaning stress and restore microbiota homeostasis. Understanding the interaction between feed additives and gut microbiota would facilitate the development of effective feeding strategies to improve the gut health, growth performance, and well-being of nursery pigs. This review summarizes certain feed additives, which are used to modulate gut microbiota with some success.

Abstract

Weaning is one of the most stressful events in the life of a pig. Unsuccessful weaning often leads to intestinal and immune system dysfunctions, resulting in poor growth performance as well as increased morbidity and mortality. The gut microbiota community is a complex ecosystem and is considered an “organ,” producing various metabolites with many beneficial functions. In this review, we briefly introduce weaning-associated gut microbiota dysbiosis. Then, we explain the importance of maintaining a balanced gut microbiota. Finally, we discuss dietary supplements and their abilities to restore intestinal balance and improve the growth performance of weaning pigs.

1. Introduction

Weaning is a stressful period for pigs due to the challenge of adapting to solid feed, maternal separation, transportation, and establishing a social hierarchy [1]. Weaning stress could disrupt intestinal barrier integrity and disturb the ecological balance of the gut microbial community [2,3,4,5]. Disrupting the balance of the gastrointestinal (GI) tract microbiota, known as dysbiosis, favors pathogen colonization on epithelial cells, leading to local inflammation [6]. Moreover, the immature immune system of newly weaned pigs can exacerbate their inflammatory response and cause them to be more susceptible to cytokine storms and sepsis [7,8,9].
The gut microbial community is largely impacted by diet [10]. The digestive system of newly weaned pigs is inefficient at utilizing nutrients from plant-based feed ingredients [11,12]. At weaning, the diet changes from a highly digestible liquid milk to a solid, less digestible nursery diet, which facilitates structural remodeling of the GI tract morphology and microbiota [13]. During this transition period, feed efficiency and nutrient digestibility are typically low. Due to the ban on antibiotics as growth promoters in animal feed, nutritional strategies have been applied to minimize the adverse effects of weaning stress. Accumulating evidence suggests that the benefits of certain feed additives on animal growth performance are partially through gut microbiota modulation [14,15,16]. Nowadays, the advent of next-generation sequencing (NGS) technology provides insight into the diverse and complex gut microbiota, which remarkably expanded our knowledge regarding how the entire gut microbiota evolves under the influence of feed additives.
In this review, we introduce the relationship between gut microbiota dysbiosis and weaning. Then, we briefly discuss the importance of maintaining a healthy gut. Finally, we summarize available information on the efficacy of nutritional interventions such as zinc, peptides, organic acids, and probiotics on the gut ecosystem integrity of weaned pigs. This review can assist future researchers and industries in developing novel strategies to modulate the swine gut microbiota through diet with the ultimate goal of improving growth performance and gut health.

2. Weaning Stress Disturbs the Gut Microbiota and the Importance of a Healthy Gut

2.1. Intestinal Dysfunction Induced by Weaning Stress

During the weaning process, adverse changes occur in the GI tract, such as villous atrophy, crypt hyperplasia, intestinal inflammation, and increased gut permeability caused by the disruption of tight junctions [17,18,19,20]. A healthy gut harbors a highly diverse community of commensal bacteria and possesses a barrier function that helps to prevent pathogenic microbes from entering the body. However, a decreased microbiota diversity and a leaky gut, caused by weaning stress, would allow pathogenic bacteria, toxins, and harmful antigens to enter the host and cause diseases (Figure 1). These main intestinal characteristics in weaned piglets have recently been reviewed [1,21,22,23] and will not be reiterated here.
Nowadays, it is widely recognized that dietary and environmental changes at weaning contribute to the modification of the gut microbiota, which may be associated with post-weaning diarrhea and enteric infections [13]. The gut microbiota community is a complex ecosystem and is considered an “organ”, producing various metabolites with many potential functions [23,24]. Microorganisms colonize the sterile gastrointestinal tract immediately after birth and take several weeks to fully develop [25,26]. Sow’s milk is the primary nutrient source for both newborn piglets and their microbiota. Thus, it imposes a selection on the gut microbial community, allowing some microbes to dominate the gut ecosystem [27,28,29,30]. The initial predominant colonizers, such as Escherichia coli (E. coli) and Streptococcus spp., could facilitate the colonization of later species [10,31]. For example, E.coli depletes oxygen in the gut, creating an anaerobic environment for late-arriving Bacteroides spp. [32]. Milk also provides antimicrobial factors, including secretory immunoglobulin A (IgA), lysozyme, and lactoferrin, which promote intestinal homeostasis [33,34].
Weaning stress, however, disrupts gut microbiota homeostasis, providing pathogenic bacteria an opportunity to multiply, resulting in increased morbidity and mortality [35,36]. At weaning, piglets are suddenly shifted to a solid diet and transferred to a new environment, which causes the gut microbiota to reassemble. Within a short period of time, the gut microbiota must develop and learn to adapt to the new nutrient source. A study by Wei et al. found that the relative abundance of Prevotella increased dramatically and rapidly after introducing solid plant-based feed: from 8.9% on the day of weaning to 17.6% on day 7 post-weaning [15]. The relative abundance of Prevotella is closely related to the consumption of plant polysaccharides and is enriched by plant polysaccharides [37]. Similarly, Megasphaera and Blautia, which are involved in carbohydrate digestion, also increased during the post-weaning period [15]. These bacterial changes will impact others and affect the entire gut ecosystem. For example, Prevotella supplants members of Bacteroides. Many studies indicated that subjects with a high relative abundance of Prevotella usually harbor a lower prevalence of Bacteroides, indicating that gut niche competition occurs between these two genera [38]. Bacteroides benefits their host by conducting immunomodulatory, metabolic, and trophic functions [39,40]. The beneficial effects of Bacteroides fragilis on the immune system are mediated by capsular polysaccharides, which positively impact the development of CD4+ T cells, affect Th1/Th2 balance, and induce regulatory T cells to secrete interleukin 10 (IL-10, a potent anti-inflammatory cytokine that protects the gut against pathogenic inflammation) [41]. Thus, the relative lower abundance of Bacteroides post-weaning may put the piglets at increased risk of intestinal disease [42].
In addition, Li et al. reported that the weaned pigs’ gut microbiota composition was characterized by the decreased proportion of Alloprevotella and Oscillospira and increased Campylobacterales [43]. Alloprevotella is an acetic and succinic acid producer, which plays a key role in maintaining the intestinal epithelial barrier and enhancing intestinal immune function [44]. Li et al. found that succinate upregulated the expression of tight-junction proteins (claudin-1, ZO-1, and ZO-2) and positively modulated inflammatory cytokine (IL-6 and IL-18) expression in the growing pig jejunum [45]. Oscillospira, a core microbiota member related to health, is a butyrate producer [46,47,48,49]. Butyrate not only acts as the dominant energy source for colonocytes but also prevents intestinal inflammation as well as systemic infection [50]. A decrease in Oscillospira levels has always been associated with inflammatory disease [48,51]. Taken together, decreased relative abundances of Alloprevotella and Oscillospira demonstrated that the gut lost certain anti-inflammatory properties after weaning. Furthermore, Campylobacter was one of the opportunistic pathogens causing acute bacterial diarrhea [52]. Hence, an increase in Campylobacter relative abundance may be one of the factors triggering post-weaning diarrhea.

2.2. The Importance of Maintaining a Healthy Gut

The animal GI tract is colonized by dynamic and complex microorganisms, known as the gut microbiota [53]. With the advent of next-generation sequencing, the characteristics of the swine gut microbiota have become clear. In general, the swine intestinal microbiota is predominated by two phyla, Firmicutes and Bacteroidetes, followed by Proteobacteria, Actinobacteria, and Spirochaetes, among others [15,16]. This general profile is relatively stable, although variations exhibit below the phylum level when the gut microbiota is disturbed.
Nowadays, the beneficial and functional roles of the gut microbiota in maintaining host health have been better identified by germ-free and fecal transplant studies [54,55]. Studies conducted by Tsai et al. and Cheng et al. found that fecal microbial transplantation was capable of improving the growth performance of both suckling and weaning pigs by modulating the gut microbiota [56,57]. In addition, antibiotics lack growth-promoting functions in germ-free animals [58]. In this section, we will briefly summarize the main functions of the gut microbiota.

2.2.1. Nutrient Metabolism

The gut microbiota possesses the ability to produce various digestive enzymes, which facilitate energy extraction from complex carbohydrates [59]. The major end products of the fermentation of non-digestible carbohydrates by microbes are short-chain fatty acids (SCFAs) such as butyrate, propionate, and acetate [60]. SCFAs serve as energy sources for both host cells and intestinal bacteria and modulate host metabolic responses. They are involved in various host-signaling mechanisms and play a central role in inhibiting histone deacetylases and activating G-coupled-protein receptors [61].
Intestinal bacteria possess numerous enzyme genes that are associated with carbohydrate-degrading activities [59]. For example, Bacteroides thetaiotaomicron produces starch-degrading enzymes [62]. At the same time, enzymes from Bacteroides ovatus are characterized by hydrolyzing galactomannans, xylans, and mannans [63,64]. In addition, Bifidobacterium spp. are able to degrade high amylose starches [65].
The gut microbiota also has the ability to synthesize certain vitamins, such as vitamins K and B, which are important components in maintaining host health [66,67]. Examples of vitamin-producing bacteria are Bifidobacterium, Bacteroides, and Enterococcus [68].

2.2.2. Immunomodulatory and Anti-Inflammatory Effects

The crosstalk between the gut microbiota and the immune system has been investigated tremendously. Microbial components and metabolites impact the immune system through neutrophil function modulation, neutrophil recruitment and activation, and T cell differentiation [69,70]. Extensive mouse-model studies have demonstrated that microbial reduction or absence greatly decreases the number of neutrophils and their progenitors, making the host more vulnerable to infection. As we discussed earlier, gut microbiota-released metabolites such as SCFAs could act as histone deacetylase (HDAC) inhibitors and therefore attenuate inflammatory responses by blocking nuclear factor-κB (NF-κB) signaling [71,72,73,74]. In addition, microbiota-derived aryl hydrocarbon receptor (AHR) ligands enhance regulatory T cells’ (Tregs) immunosuppressive activities during inflammation, which indirectly influences neutrophil recruitment and activation [75,76]. Moreover, bioactive amines (e.g., histamine), generated by commensal bacteria from amino acids, play a crucial role in regulating immune responses [77].

2.2.3. Impacts of Gut Microbiota on Intestinal Barrier Function and Gut Structure

The intestinal barrier is the host’s first line of defense against potentially pathogenic bacteria and toxic molecules [78]. Mounting evidence indicates that commensal bacteria contribute to the structural and functional development of the gastrointestinal tract. For example, Lactobacillus rhamnosus GG-derived protein p40 inhibits cytokine-induced apoptosis of intestinal epithelial cells [79]. Bacteroides thetaiotaomicron is involved in desmosome maintenance at the epithelial villus by inducing the expression of SPRR2A (small proline-rich protein 2A) [80]. Another example of an intestinal bacteria that influences barrier function is Akkermansia muciniphilia, which regulates epithelial barrier permeability by increasing endocannabinoid levels [81].
The benefits of the microbiota in supporting gut structure and function can be easily appreciated from germ-free mice studies. Microbiota-lacking animals exhibit thin villi, weak peristalsis, decreased intestinal surface area, reduced villus capillary network, and prolonged cell cycle time [82].

3. The Effects of Nutritional Interventions on Swine Gut Health around Weaning

In order to overcome weaning stress, feed additives are commonly used in the swine industry to improve intestinal function, increase feed utilization, enhance immune response, prevent diseases, and improve growth rate [83]. Examples of additives used in swine diets include zinc, peptides, organic and inorganic acids, prebiotics, probiotics, plant extracts, essential oils, and yeast, as reviewed by Liu et al. [83]. Compared to other published reviews, this review includes more information about how feed additives impact the swine gut microbial ecology. This section will focus on several feed additives known to modulate the swine intestinal microbiota with some success; putative mechanisms and the growth performance results for each additive are also included.

3.1. Zinc

3.1.1. Pharmacological Role of Zinc

Nursery pigs usually require 80 to 100 ppm of zinc (Zn) to meet their requirement for growth [84]. Feeding pharmacological levels of inorganic Zn to nursery pigs is usually recommended to combat postweaning diarrhea and improve growth performance [85,86,87,88]. Hahn and Baker evaluated the effects of the additional of high levels (3000 ppm and 5000 ppm) of Zn from different sources on the growth performance of weanling pigs. They found that ZnO addition, regardless of level, significantly increased both daily gain and daily feed intake, whereas inconsistent growth performance response was observed when using ZnSO4. At the same time, they conducted a trial to determine whether 3000 ppm of dietary Zn from ZnO or ZnSO4 had positive effects on weaning pigs fed a high nutrient density diet (basal starting diet fortified with both plasma protein and fish meal and containing more dried whey). The results showed that only pigs fed 3000 ppm ZnO had significantly increased weight gain and voluntary feed intake. ZnSO4 addition did not elicit a performance response [89]. These results indicated that feed composition and different chemical forms of Zn might affect the response outcome. In addition, the duration of pigs on high Zn diets greatly impacts the growth performance. Carlson et al. determined that supplementing 3000 ppm of Zn as ZnO immediately after weaning for two weeks could accelerate the growth of both early (<14 d) and traditionally (>21 d) weaned pigs; however, the addition of Zn could not enhance growth if fed only during the first week after weaning [90]. It is possible that newly weaned pigs do not absorb enough Zn due to the low feed intake during the first week after weaning.
Compared with traditional ZnO, nano-size ZnO is smaller in size and has a larger surface area and higher bioavailability [91]. Pei et al. found that 450 ppm Zn from nano-ZnO could enhance the growth performance of weaned piglets as efficiently as high concentrations (3000 ppm) of conventional ZnO [92]. In the same study, they also suggested that 150 ppm nano-ZnO had positive influences on growth performance. However, a study from Li et al. reported that nano-ZnO at 120 ppm failed to improve the growth performance of weaned pigs [93]. The inconsistency of the results might be due to the different concentrations and methods of synthesis of nano-ZnO.
There are many possible mechanisms to explain why high levels of Zn have beneficial effects on the growth performance of weaned pigs. Studies have shown that Zn plays an important role in maintaining intestinal epithelial barrier function and even tightens “leaky gut” [94]. Hu et al. found that the benefits of Zn in the form of ZnO on intestinal integrity are related to its ability to decrease inflammatory gene expression by inhibiting the TLR4–MyD88 signaling pathways, which reduces the level of pro-inflammatory cytokines and chemokines [95]. The barrier protective role of Zn was also supported by a report that Zn deprivation could disassemble tight junction proteins and cause epithelial barrier dysfunction [96,97].
Weaning-induced villous atrophy in the small intestine could be reversed by Zn supplements [95]. Furthermore, high dietary levels of Zn have a positive impact on gut microbiota composition and stability. The weaning process dramatically affects gut microbiome composition [43]. Any disturbances in the stability of the gut microbiome could lead to the overgrowth of indigenous as well as exogenous pathogenic bacteria, which may lead to diarrhea. Katouli et al. suggested that 2500 ppm of Zn in pig diets could maintain the stability of the intestinal microbiota and high diversity of coliforms, which may compete with diarrheagenic strains for colonizing receptor sites [98]. More interestingly, high ZnO and antibiotics had similar effects on the gut microbiota of weaning piglets [99]. Both significantly increased the abundance of Verrucomicrobia, TM7, Spirochaetes, Tenericutes, and Euryarchaeota and reduced Chlamydiae in ileal digesta. Nevertheless, the impacts of high concentrations of ZnO on swine commensal microbiota remain controversial. Starke et al. reported that the supplementation of the piglet diet with high concentrations of ZnO was associated with an increase in Megasphaera and a decrease in E. coli and Enterobacteriaceae, whereas other researchers such as Højberg et al., Vahjen et al., and Wei et al. noticed opposite trends [16,100,101,102]. These inconsistent results might be related to differences between the studies, such as various weaning days, sampling timepoints, fecal collection from various intestinal segments and rectal swabs, and separate nursing facility environments. Although pharmacological levels of Zn are effective at controlling postweaning diarrhea, the environmental burden from excessive levels of Zn excretion from manure has led many countries to limit Zn usage as a feed supplement. For example, the European Union limits ZnO usage at 150 ppm in swine diet [103] and China restricts the use of ZnO to 1600 ppm [104].
Apart from ZnO, other organic forms of Zn such as Zn-methionine and Zn-lysine are available for inclusion in diets at lower levels due to their greater bioavailability compared to ZnO [105]. For example, Ward et al. reported that the inclusion of 250 ppm Zn-methionine in nursery starter diets had beneficial effects equivalent to 2000 ppm of Zn from ZnO when 160 ppm of Zn from ZnSO4 was added in both diets [106]. However, this finding is in contrast to those of Hollis et al., who reported that supplementing 500 ppm Zn from several organic sources (Zinc polysaccharide complex, Zinc proteinate, Zinc amino acid complex, Zinc amino acid chelate, and Zinc methionine) was not as effective at stimulating growth rate and feed efficiency as high levels of ZnO (2000 ppm) [107]. Taken together, these findings suggest that further research is needed to investigate and determine the most effective condition and dose for the inclusion of organic Zn in piglet diets.

3.1.2. Antimicrobial Activity of Zinc

A bacterial infection is one of the most serious animal health issues. ZnO is widely known for its antibacterial properties, which have been investigated largely with both Gram-positive and Gram-negative bacteria. The main conclusions of these investigations can be summarized as follows: (1) ZnO shows a size-dependent toxic effect on bacteria and smaller ZnO particle size exhibits better antimicrobial activity [108,109,110]; (2) the antibacterial activity is related to the particle surface area and concentration, and the higher concentration and larger surface area have better antibacterial activity [111]; (3) the antibacterial activity of ZnO decreased with increasing heating temperature [112]; and (4) several proposed mechanisms that explained ZnO antibacterial activity are illustrated in Figure 2 and described below.
(i)
Reactive oxygen species (ROS) generation.
Studies by Sawai et al. and Lipovsky et al. highlighted that ROS was the main mechanism for ZnO antibacterial activity [113,114]. The reactive species are hydrogen peroxide (H2O2), hydroxide (OH), and superoxide anion (O2). The toxicity of these species involves the destruction of cellular components, such as lipids, DNA, and proteins.
(ii)
Zinc ion (Zn2+) release.
The release of zinc ions in a medium containing ZnO and bacteria plays an important role in the toxic effect of ZnO particles [115]. Metal ions enter bacterial cells, followed by a reduction in ATP levels and disruption to DNA replication.
(iii)
Changes in bacterial membrane permeability.
Some researchers proposed that there are electrostatic forces that serve as a powerful bond between metal oxide nanoparticles and bacteria. As a result, the cell membrane is damaged. Stoimenov et al. demonstrated that the opposite charge brings the bacteria and metal oxide nanoparticles together [116]. Zhang et al. also referred to an electrostatic phenomenon arising when E. coli was treated with ZnO due to the negative charge of the bacterial cell wall and positive charge of ZnO in a water suspension [117]. Such direct interaction may be attributed to bacterial membrane disorganization. As the ZnO nanoparticles precipitated on the bacteria exterior, the cell internalized these nanoparticles, which caused the cellular contents to leak out [118].

3.2. Peptides

3.2.1. Functional Properties of Bioactive Peptides

Bioactive peptides, which possess multiple biological functions, have many potential beneficial effects on health [119,120]. A wide variety of natural sources, including animals, plants, and microorganisms, could produce antimicrobial peptides (AMP). These peptides show antimicrobial activities against pathogens, including bacteria, fungi, and viruses [121,122]. The direct killing mechanisms of AMP can be divided into membrane targeting and non-membrane targeting. The membrane targeting AMPs can be further divided into receptor-mediated and non-receptor-mediated interactions [123]. Three models have been proposed to describe how AMPs penetrate microbial membranes: the barrel-stave model [124], the carpet model [125], and the toroidal-pore model [126]. For non-membrane targeting mechanisms, AMPs can either target the bacterial cell wall to inhibit cell wall synthesis or have intracellular targets to inhibit protein and nucleic acid synthesis and to disrupt enzymatic and protein activity [127,128,129,130]. In addition to killing bacteria directly, bioactive peptides exert immunomodulatory functions to enhance microbial killing and reduce and control inflammation. In an infection, peptides activate and recruit immune cells to the site of infection [131,132,133]. Moreover, bioactive peptides could decrease pro-inflammatory chemokine expression and consequently suppress inflammation [132,134].

3.2.2. Peptide Absorption and Utilization in Animal Nutrition

Protein is an essential macronutrient for animal growth and reproduction. It needs to be degraded into amino acids or small peptides to be absorbed by the intestine and transported in the blood [135,136]. Evidence suggests that amino acids in peptide form are absorbed more readily and rapidly than free amino acids [137,138]. Thus, peptides derived from animals, plants, and microorganisms with well-defined physiological roles are incorporated into commercially available products. Scientific data suggest that peptides have promising applications in animal nutrition. Pigs fed a diet containing spray-dried porcine intestine hydrolysate for two weeks after weaning had better growth performance than those fed control diets containing spray-dried plasma or dried whey [139,140]. A positive carry-over effect was detected [140], which was likely associated with increased intestinal villus area as well as an improved digestibility and absorption rate of dietary nutrients [141]. Moreover, Opheim et al. reported that adding hydrolyzed Atlantic salmon viscera in a broiler diet, at inclusion levels of 5% and 10%, resulted in better growth performance compared with either a plant protein-based diet or a fishmeal control diet [142].
Tang et al. found that feeding AMPs (Lactoferrampin and Lactoferricin) derived from bovine milk improved growth performance and decreased diarrhea rate of weaning pigs infected by E. coli [143,144]. Xiong et al. observed that dietary AMP mixture (2.0 or 3.0 g/kg) supplementation (lactoferrin, cecropin, defensin, and plectasin) improved the average daily gain of weaned pigs reared on five different commercial farms and pigs fed with 2.0 g/kg AMP had greater daily gain than those fed with 3.0 g/kg AMP [145]. However, Yoon et al. reported that the average daily gain increased with increasing levels of AMP-A3 (60 and 90 mg/kg) or AMP-P5 (40 and 60 mg/kg) [146,147]. These findings were in contrast to a study by Shan et al., who did not observe improvement in daily gain when pigs were fed with 1.0 g/kg of lactoferrin [148]. The differences between the results may be related to the types and doses of AMPs applied in the experiments.
The beneficial effects of AMPs on intestinal development have also been reported in weaned pigs. A study from Xiao et al. found that AMP supplements were able to alleviate intestinal injury induced by mycotoxin deoxynivalenol in weaning pigs [149,150]. Wang et al. observed that lactoferrin increased villus height and decreased crypt depth of the intestine, directly affecting nutrient absorption and contributing to improved growth performance [151]. Studies by Wang et al. and Tang et al. revealed that adding the peptides lactoferrin or lactoferramoin-lactoferricin in nursery diets increased some beneficial bacteria such as Lactobacillus and Bifidobacterium, and decreased the pathogenic bacteria E. coli and Salmonella in the small intestine [151,152]. Wu et al. also reported that cecropin AD enriched Lactobacillus and reduced diarrhea incidence in weaned piglets challenged with E. coli [153]. In addition, Poudel et al. found that a peptide-based additive could accelerate the maturation of swine gut microbiota [154].

3.3. Organic Acids

The exact mode of action of dietary organic acids remains unknown, but several mechanisms have been proposed. Their benefits on animal growth performance are believed to be associated with reducing gastrointestinal pH, inhibiting pathogenic bacteria, serving as an energy source, and facilitating mineral utilization [155].

3.3.1. Lowering Gastrointestinal pH

Protein digestion begins in the stomach with the help of pepsin, which is a potent enzyme utilized for breaking down proteins into smaller peptides [156]. Pepsin activity depends on acidic environments. Piper and Fenton reported that pepsin exhibits maximum activity in the pH range of 1.5–2.5. A pH outside this range could reduce or even inhibit enzyme activity [157].
Newly weaned pigs generate insufficient levels of gastric acid, which results in high stomach pH and consequently reduces protein digestion [158]. Many experiments have been conducted to examine the effects of various acidifiers on the reduction of the stomach and gastrointestinal tract pH of nursery pigs. However, the results were inconsistent. For example, Radcliffe et al. found stomach digesta pH was significantly decreased when pigs were fed citric acid [159]. Scipioni et al. reported a nonsignificant decrease in the stomach and jejunal pH by feeding citric acid in a complex diet [160]. Similar results were found in later work conducted by Burnell et al. [161]. Further research is needed to clarify these inconsistent results.

3.3.2. Inhibition of Pathogenic Bacteria

Dietary organic acids lower stomach and intestinal pH, thus providing a less hospitable environment for pathogenic microbes. The pH in the stomach has two main functions: (1) stimulates digestive enzyme secretion, and (2) provides a barrier against the entry of foreign bacteria into the small intestine. As previously mentioned, early weaned piglets have a limited capacity to digest protein. These undigested proteins could promote microbial fermentation, leading to the proliferation of harmful bacteria in the gastrointestinal tract [162]. Thus, lowering the pH in the gastrointestinal tract is important for weanling pig health. A study by Thomlinson and Lawrence revealed that dietary lactic acid decreased stomach pH and delayed the multiplication of enterotoxigenic E. coli in weaned pigs [163]. In addition, adding butyric acid to chicken diets protected broilers from Salmonella infection [164,165]. Furthermore, these acids had no antimicrobial effects on pH-insensitive beneficial bacteria such as Lactobacilli and Bifidobacterium spp. [155]. A recent study by Wei et al. conducted in weaning pigs indicated that pigs fed an organic acid mixture had more diverse microbiota and produced more potential beneficial bacteria such as Turicibacter, Blautia, and Oscillospira and decreased proportions of Sarcina and Veillonella, which are linked to various inflammatory diseases [15].

3.3.3. Energy Source

Organic acids are commonly used in pig nutrition, where they serve as a direct energy source, and some are important players in metabolic pathways and cycles. It has been suggested that pigs can utilize fumaric acid as an energy source as efficiently as glucose [166]. Blank et al. pointed out that there is a possibility that fumaric acid, as a readily available energy source, may have a local trophic effect on the small intestine mucosa and lead to an increase in the absorptive surface area and capacity of the small intestine due to the faster recovery of the gastrointestinal epithelial cells after weaning [167].

3.3.4. Beneficial Effects of Organic Acids on Swine

The benefits of organic acids in enhancing nutrient digestibility and modulating intestinal microbiota should improve growth performance in weanling pigs. Falkowski and Aherne demonstrated that dietary fumaric acid or citric acid improved the average daily gain and feed conversion ratio of weanling pigs [168]. Likewise, Giesting and Easter found that there was a linear increase in feed efficiency and daily gain when weanling pigs were supplied with increasing levels of fumaric acids [169]. Various other acidifiers such as benzoic acid and butyric acid also showed positive effects on growth performance [170,171,172,173]. In contrast, Ahmed observed that the acidifier blend (formic acid, propionic acid, lactic acid, phosphoric acid, and SiO2) negatively impacted growth performance parameters of weaned pigs [174]. In addition, the effects of organic acid on nutrient digestibility remained controversial. For instance, fumaric acid had no or even adverse effects on the ileal digestibility of amino acids [175,176]. However, a positive effect on the apparent total tract digestibility of protein was reported [177]. Besides, it has been reported that dietary benzoic acid improved the apparent ileal digestibility of total nitrogen in weanling pigs [170,178]. Factors such as diet buffering capacity as well as acidifier types and concentrations seem to contribute to these inconsistent results. Further investigations are thus needed to clarify the modes of action of acidifiers and to outline the specific conditions under which they are most effective for various available acidifiers applied in weaning piglets.

3.4. Probiotics

3.4.1. Probiotics in Swine Nutrition

Probiotics are defined as live microorganisms, which, when administered in adequate amounts, confer a health benefit on the host [179]. Azad et al. thoroughly reviewed probiotics and their biological effects [180]. Members of the genera Lactobacillus, Bifidobacterium, and Bacillus with probiotic properties are the most common microorganisms applied in the swine industry, but not exclusively, as a growing number of probiotic microorganisms have been discovered and are available for swine production. The beneficial effects of probiotics such as Lactobacillus reuteri and Bacillus licheniformis on the growth performance of weanling pigs have been reported [181,182,183]. Giang et al. observed that the inclusion of 0.06% probiotic complex (Enterococcus faecium 6H2, Lactobacillus acidophilus C3, Pediococcus pentosaceus D7, L. plantarum 1K8, and L. plantarum 3K2) in diets had positive effects on average daily gain, average daily feed intake, and feed/gain ratio during the first two weeks post weaning, whereas no effects were found in the following period [184]. Likewise, Huang et al. demonstrated that dietary 0.1% Lactobacillus complex (L. gasseri, L. reuteri, L. acidophilus, and L. fermentum) supplementation improved the average feed intake of piglets during the first two weeks after weaning [185]. It is possible that gastrointestinal bacteria in newly weaned piglets were vulnerable and could be easily changed by external factors such as probiotics. Therefore, the positive effects of probiotics on growth performance were only observed in the first two weeks after weaning. However, different Lactobacillus complex and inclusion levels may cause different results. For example, Zhao et al. reported that the inclusion of L. reuteri and L. plantarum complex (0.1% or 0.2%) in diets had no effects on average daily gain, average daily feed intake, or feed/gain in weaning pigs [186]. In fact, a reduction in feed efficiency was reported by Frantz et al. as a result of a 0.1% Lactobacillus diet [187].

3.4.2. Probiotics as Modulators of Swine Gut Microbiota

In several recent studies, the supplementation of piglet diet with probiotics has been associated with an increase in Lactobacillus or Bifidobacterium spp. and a decrease in E. coli [188,189,190,191]. These changes possibly benefit intestinal health, which can lead to improved growth performance. Zhang et al. found that oral administration of the Bacillus licheniformis-B. subtilis mixture modulated the gut microbiota of weaned pigs by enriching certain members of Clostridium, Lactobacillus, and Turicibacter [192]. In addition, dietary Bacillus subtilis could enrich bacteria with high fiber fermentation efficacy [193]. Besides, Shin et al. indicated that gut microbial diversity and richness (Simpson, Shannon, ACE, and Chao 1) were higher in pigs fed the probiotic microbe L. plantarum JDFM LP1 than those fed the basal diet [194]. However, Wang et al. found that dietary L. plantarum PFM105 had no influences on Shannon, Chao 1, observed species, and ACE, although the Simpson index was increased. In addition, overconsumption of probiotics may disturb the gut microbial ecosystem and increase the risk of enteric disease [192,195,196]. Overall, these results suggest that the benefits of probiotics on the gut are strain- and dosage-dependent.

4. Conclusions

To mitigate the negative effects of weaning, nutrition intervention strategies are selected to increase nutrient digestibility, enhance intestinal health, and prevent or mitigate disease. In response to intervention, many notable shifts in the swine gut structure and microbial communities were detected. These changes benefit gut health and probably contribute to improved growth performance.

Author Contributions

Literature review and draft preparation, X.W.; discussion, reviewing, and revision, T.T. and S.H.; conception and editing, J.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Acknowledgments

Part of the content included in this publication first appeared in an author’s thesis [197].

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Campbell, J.M.; Crenshaw, J.D.; Polo, J. The biological stress of early weaned piglets. J. Anim. Sci. Biotechnol. 2013, 4, 1–4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Boudry, G.; Péron, V.; Le Huërou-Luron, I.; Lallès, J.P.; Sève, B. Weaning Induces Both Transient and Long-Lasting Modifications of Absorptive, Secretory, and Barrier Properties of Piglet Intestine. J. Nutr. 2004, 134, 2256–2262. [Google Scholar] [CrossRef] [PubMed]
  3. Spreeuwenberg, M.A.M.; Verdonk, J.M.A.J.; Gaskins, H.R.; Verstegen, M.W.A. Small Intestine Epithelial Barrier Function Is Compromised in Pigs with Low Feed Intake at Weaning. J. Nutr. 2001, 131, 1520–1527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Modesto, M.; D’Aimmo, M.R.; Stefanini, I.; Trevisi, P.; De Filippi, S.; Casini, L.; Biavati, B. A novel strategy to select Bifidobacterium strains and prebiotics as natural growth promoters in newly weaned pigs. Livest. Sci. 2009, 122, 248–258. [Google Scholar] [CrossRef]
  5. Konstantinov, S.R.; Awati, A.A.; Williams, B.A.; Miller, B.G.; Jones, P.; Stokes, C.R.; De Vos, W.M. Post-natal development of the porcine microbiota composition and activities. Environ. Microbiol. 2006, 8, 1191–1199. [Google Scholar] [CrossRef]
  6. Berkes, J.; Viswanathan, V.K.; Savkovic, S.D.; Hecht, G. Intestinal epithelial responses to enteric pathogens: Effects on the tight junction barrier, ion transport, and inflammation. Gut 2003, 52, 439–451. [Google Scholar] [CrossRef] [Green Version]
  7. Blecha, F.; Pollman, D.S.; Nichols, D.A. Weaning Pigs at an Early Age Decreases Cellular Immunity. J. Anim. Sci. 1983, 56, 396–400. [Google Scholar] [CrossRef]
  8. Pié, S.; Lallès, J.P.; Blazy, F.; Laffitte, J.; Sève, B.; Oswald, I.P. Weaning is Associated with an Upregulation of Expression of Inflammatory Cytokines in the Intestine of Piglets. J. Nutr. 2004, 134, 641–647. [Google Scholar] [CrossRef] [Green Version]
  9. Hu, C.H.; Xiao, K.; Luan, Z.S.; Song, J. Early weaning increases intestinal permeability, alters expression of cytokine and tight junction proteins, and activates mitogen-activated protein kinases in pigs1. J. Anim. Sci. 2013, 91, 1094–1101. [Google Scholar] [CrossRef] [Green Version]
  10. Wang, X.; Tsai, T.; Deng, F.; Wei, X.; Chai, J.; Knapp, J.; Zhao, J. Longitudinal investigation of the swine gut microbiome from birth to market reveals stage and growth performance associated bacteria. Microbiome 2019, 7, 1–18. [Google Scholar] [CrossRef] [Green Version]
  11. Cranwell, P.D. The development of acid and pepsin (EC 3. 4. 23. 1) secretory capacity in the pig; the effects of age and weaning: 1. Studies in anaesthetized pigs. Br. J. Nutr. 1985, 54, 305–320. [Google Scholar] [CrossRef] [Green Version]
  12. Manners, M.J. The development of digestive function in the pig. Proc. Nutr. Soc. 1976, 35, 49–55. [Google Scholar] [CrossRef] [Green Version]
  13. Lallès, J.P.; Bosi, P.; Smidt, H.; Stokes, C.R. Weaning—A challenge to gut physiologists. Livest. Sci. 2007, 108, 82–93. [Google Scholar] [CrossRef]
  14. Kogut, M.H. The effect of microbiome modulation on the intestinal health of poultry. Anim. Feed. Sci. Technol. 2019, 250, 32–40. [Google Scholar] [CrossRef]
  15. Wei, X.; Bottoms, K.; Stein, H.; Blavi, L.; Bradley, C.; Bergstrom, J.; Knapp, J.; Story, R.; Maxwell, C.; Tsai, T.; et al. Dietary Organic Acids Modulate Gut Microbiota and Improve Growth Performance of Nursery Pigs. Microorganisms 2021, 9, 110. [Google Scholar] [CrossRef]
  16. Wei, X.; Tsai, T.; Knapp, J.; Bottoms, K.; Deng, F.; Story, R.; Zhao, J. ZnO modulates swine gut microbiota and improves growth performance of nursery pigs when combined with peptide cocktail. Microorganisms 2020, 8, 146. [Google Scholar] [CrossRef] [Green Version]
  17. Brown, D.; Maxwell, C.; Erf, G.; Davis, M.; Singh, S.; Johnson, Z. The influence of different management systems and age on intestinal morphology, immune cell numbers and mucin production from goblet cells in post-weaning pigs. Veter. Immunol. Immunopathol. 2006, 111, 187–198. [Google Scholar] [CrossRef]
  18. Bomba, L.; Minuti, A.; Moisa, S.J.; Trevisi, E.; Eufemi, E.; Lizier, M.; Ajmone-Marsan, P. Gut response induced by weaning in piglet features marked changes in immune and inflammatory response. Funct. Integr. Gen. 2014, 14, 657–671. [Google Scholar] [CrossRef]
  19. Smith, F.; Clark, J.E.; Overman, B.L.; Tozel, C.C.; Huang, J.H.; Rivier, J.E.; Moeser, A.J. Early weaning stress impairs development of mucosal barrier function in the porcine intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 298, G352–G363. [Google Scholar] [CrossRef] [Green Version]
  20. Wan, J.; Zhang, J.; Chen, D.; Yu, B.; Mao, X.; Zheng, P.; He, J. Alginate oligosaccharide-induced intestinal morphology, barrier function and epithelium apoptosis modifications have beneficial effects on the growth performance of weaned pigs. J. Anim. Sci. Biotechnol. 2018, 9, 1–12. [Google Scholar] [CrossRef] [Green Version]
  21. Pluske, J.R.; Hampson, D.J.; Williams, I.H. Factors influencing the structure and function of the small intestine in the weaned pig: A review. Livest. Prod. Sci. 1997, 51, 215–236. [Google Scholar] [CrossRef]
  22. Xiong, X.; Tan, B.; Song, M.; Ji, P.; Kim, K.; Yin, Y.; Liu, Y. Nutritional Intervention for the Intestinal Development and Health of Weaned Pigs. Front. Veter. Sci. 2019, 6, 46. [Google Scholar] [CrossRef] [Green Version]
  23. Bocci, V. The Neglected Organ: Bacterial Flora Has a Crucial Immunostimulatory Role. Perspect. Biol. Med. 1992, 35, 251–260. [Google Scholar] [CrossRef]
  24. Baquero, F.; Nombela, C. The microbiome as a human organ. Clin. Microbiol. Infect. 2012, 18, 2–4. [Google Scholar] [CrossRef] [Green Version]
  25. Houghteling, P.D.; Walker, W.A. Why is initial bacterial colonization of the intestine important to the infant’s and child’s health? J. Pediatr. Gastroenterol. Nutr. 2015, 60, 294. [Google Scholar] [CrossRef] [Green Version]
  26. Mackie, R.I.; Sghir, A.; Gaskins, H.R. Developmental microbial ecology of the neonatal gastrointestinal tract. Am. J. Clin. Nutr. 1999, 69, 1035s–1045s. [Google Scholar] [CrossRef]
  27. Martín, V.; Maldonado-Barragán, A.; Moles, L.; Rodriguez-Baños, M.; del Campo, R.; Fernández, L.; Jiménez, E. Sharing of bacterial strains between breast milk and infant feces. J. Hum. Lact. 2012, 28, 36–44. [Google Scholar] [CrossRef]
  28. Marcobal, A.; Barboza, M.; Froehlich, J.W.; Block, D.E.; German, J.B.; Lebrilla, C.B.; Mills, D.A. Consumption of Human Milk Oligosaccharides by Gut-Related Microbes. J. Agric. Food Chem. 2010, 58, 5334–5340. [Google Scholar] [CrossRef] [Green Version]
  29. Ward, R.E.; Niñonuevo, M.; Mills, D.A.; Lebrilla, C.B.; German, J.B. In vitro fermentability of human milk oligosaccharides by several strains of bifidobacteria. Mol. Nutr. Food Res. 2007, 51, 1398–1405. [Google Scholar] [CrossRef]
  30. Sela, D.A.; Mills, D.A. Nursing our microbiota: Molecular linkages between bifidobacteria and milk oligosaccharides. Trends Microbiol. 2010, 18, 298–307. [Google Scholar] [CrossRef] [Green Version]
  31. Petri, D.; Hill, J.; Van Kessel, A. Microbial succession in the gastrointestinal tract (GIT) of the preweaned pig. Livest. Sci. 2010, 133, 107–109. [Google Scholar] [CrossRef]
  32. Bokulich, N.A.; Chung, J.; Battaglia, T.; Henderson, N.; Jay, M.; Li, H.; Blaser, M.J. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci. Transl. Med. 2016, 8, 343ra382. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Rogier, E.W.; Frantz, A.L.; Bruno, M.E.C.; Wedlund, L.; Cohen, D.A.; Stromberg, A.J.; Kaetzel, C.S. Secretory antibodies in breast milk promote long-term intestinal homeostasis by regulating the gut microbiota and host gene expression. Proc. Natl. Acad. Sci. USA 2014, 111, 3074–3079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Planer, J.D.; Peng, Y.; Kau, A.L.; Blanton, L.V.; Ndao, I.M.; Tarr, P.I.; Gordon, J.I. Development of the gut microbiota and mucosal IgA responses in twins and gnotobiotic mice. Nature 2016, 534, 263–266. [Google Scholar] [CrossRef] [Green Version]
  35. Guevarra, R.B.; Hong, S.H.; Cho, J.H.; Kim, B.R.; Shin, J.; Lee, J.H.; Kim, H.B. The dynamics of the piglet gut microbiome during the weaning transition in association with health and nutrition. J. Anim. Sci. Biotechnol. 2018, 9, 1–9. [Google Scholar] [CrossRef] [Green Version]
  36. Gresse, R.; Chaucheyras-Durand, F.; Fleury, M.A.; Van de Wiele, T.; Forano, E.; Blanquet-Diot, S. Gut Microbiota Dysbiosis in Postweaning Piglets: Understanding the Keys to Health. Trends Microbiol. 2017, 25, 851–873. [Google Scholar] [CrossRef]
  37. Ivarsson, E.; Roos, S.; Liu, H.Y.; Lindberg, J.E. Fermentable non-starch polysaccharides increases the abundance of Bacteroides–Prevotella–Porphyromonas in ileal microbial community of growing pigs. Animal 2014, 8, 1777–1787. [Google Scholar] [CrossRef]
  38. Kovatcheva-Datchary, P.; Nilsson, A.; Akrami, R.; Lee, Y.S.; De Vadder, F.; Arora, T.; Bäckhed, F. Dietary fiber-induced improvement in glucose metabolism is associated with increased abundance of Prevotella. Cell Metab. 2015, 22, 971–982. [Google Scholar] [CrossRef] [Green Version]
  39. Hooper, L.V.; Wong, M.H.; Thelin, A.; Hansson, L.; Falk, P.G.; Gordon, J.I. Molecular analysis of commensal host-microbial relationships in the intestine. Science 2001, 291, 881–884. [Google Scholar] [CrossRef] [Green Version]
  40. Xu, J.; Gordon, J.I. Honor thy symbionts. Proc. Natl. Acad. Sci. USA 2003, 100, 10452–10459. [Google Scholar] [CrossRef] [Green Version]
  41. Cohen-Poradosu, R.; McLoughlin, R.M.; Lee, J.C.; Kasper, D.L. Bacteroides fragilis–Stimulated Interleukin-10 Contains Expanding Disease. J. Infect. Dis. 2011, 204, 363–371. [Google Scholar] [CrossRef]
  42. Meng, Q.; Luo, Z.; Cao, C.; Sun, S.; Ma, Q.; Li, Z.; Shan, A. Weaning alters intestinal gene expression involved in nutrient metabolism by shaping gut microbiota in pigs. Front. Microbiol. 2020, 11, 694. [Google Scholar] [CrossRef]
  43. Li, Y.; Guo, Y.; Wen, Z.; Jiang, X.; Ma, X.; Han, X. Weaning Stress Perturbs Gut Microbiome and Its Metabolic Profile in Piglets. Sci. Rep. 2018, 8, 1–12. [Google Scholar] [CrossRef] [Green Version]
  44. Downes, J.; Dewhirst, F.E.; Tanner, A.C.R.; Wade, W.G. Description of Alloprevotella rava gen. nov., sp. nov., isolated from the human oral cavity, and reclassification of Prevotella tannerae Moore et al. 1994 as Alloprevotella tannerae gen. nov., comb. nov. Int. J. Syst. Evol. Microbiol. 2013, 63, 1214–1218. [Google Scholar] [CrossRef]
  45. Li, X.; Mao, M.; Zhang, Y.; Yu, K.; Zhu, W. Succinate Modulates Intestinal Barrier Function and Inflammation Response in Pigs. Biomolecules 2019, 9, 486. [Google Scholar] [CrossRef] [Green Version]
  46. Konikoff, T.; Gophna, U. Oscillospira: A Central, Enigmatic Component of the Human Gut Microbiota. Trends Microbiol. 2016, 24, 523–524. [Google Scholar] [CrossRef]
  47. Lee, G.-H.; Rhee, M.-S.; Chang, D.-H.; Lee, J.; Kim, S.; Yoon, M.H.; Kim, B.-C. Oscillibacter ruminantium sp. nov., isolated from the rumen of Korean native cattle. Int. J. Syst. Evol. Microbiol. 2013, 63, 1942–1946. [Google Scholar] [CrossRef] [Green Version]
  48. Gophna, U.; Konikoff, T.; Nielsen, H.B. Oscillospira and related bacteria–From metagenomic species to metabolic features. Environ. Microbiol. 2017, 19, 835–841. [Google Scholar] [CrossRef] [Green Version]
  49. Shetty, S.A.; Hugenholtz, F.; Lahti, L.; Smidt, H.; De Vos, W.M. Intestinal microbiome landscaping: Insight in community assemblage and implications for microbial modulation strategies. FEMS Microbiol. Rev. 2017, 41, 182–199. [Google Scholar] [CrossRef]
  50. Schulthess, J.; Pandey, S.; Capitani, M.; Rue-Albrecht, K.C.; Arnold, I.; Franchini, F.; Powrie, F. The short chain fatty acid butyrate imprints an antimicrobial program in macrophages. Immunity 2019, 50, 432–445.e437. [Google Scholar] [CrossRef] [Green Version]
  51. Walters, W.A.; Xu, Z.; Knight, R. Meta-analyses of human gut microbes associated with obesity and IBD. FEBS Lett. 2014, 588, 4223–4233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Janssen, R.; Krogfelt, K.A.; Cawthraw, S.A.; Van Pelt, W.; Wagenaar, J.A.; Owen, R.J. Host-Pathogen Interactions in Campylobacter Infections: The Host Perspective. Clin. Microbiol. Rev. 2008, 21, 505–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Sekirov, I.; Russell, S.L.; Antunes, L.C.M.; Finlay, B.B. Gut Microbiota in Health and Disease. Physiol. Rev. 2010, 90, 859–904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kennedy, E.A.; King, K.Y.; Baldridge, M.T. Mouse Microbiota Models: Comparing Germ-Free Mice and Antibiotics Treatment as Tools for Modifying Gut Bacteria. Front. Physiol. 2018, 9, 1534. [Google Scholar] [CrossRef] [Green Version]
  55. Jorup-Rönström, C.; Håkanson, A.; Sandell, S.; Edvinsson, O.; Midtvedt, T.; Persson, A.-K.; Norin, E. Fecal transplant against relapsingClostridium difficile-associated diarrhea in 32 patients. Scand. J. Gastroenterol. 2012, 47, 548–552. [Google Scholar] [CrossRef]
  56. Tsai, T.; Maxwell, C.V.; Zhao, J. 171 Fecal microbiota transplant at weaning improves growth performance and alters fecal microbiome in pigs. J. Anim. Sci. 2019, 97, 175. [Google Scholar] [CrossRef]
  57. Cheng, C.S.; Wei, H.K.; Wang, P.; Yu, H.C.; Zhang, X.M.; Jiang, S.W.; Peng, J. Early intervention with faecal microbiota transplantation: An effective means to improve growth performance and the intestinal development of suckling piglets. Animal 2019, 13, 533–541. [Google Scholar] [CrossRef] [Green Version]
  58. Coates, M.E.; Fuller, R.; Harrison, G.F.; Lev, M.; Suffolk, S.F. A comparision of the growth of chicks in the Gustafsson germ-free apparatus and in a conventional environment, with and without dietary supplements of penicillin. Br. J. Nutr. 1963, 17, 141–150. [Google Scholar] [CrossRef]
  59. 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]
  60. Macfarlane, S.; Macfarlane, G.T. Regulation of short-chain fatty acid production. Proc. Nutr. Soc. 2003, 62, 67–72. [Google Scholar] [CrossRef]
  61. Tungland, B. Chapter 2-Short-Chain Fatty Acid Production and Functional Aspects on Host Metabolism. In Human Microbiota in Health and Disease; Tungland, B., Ed.; Academic Press: Cambridge, UK, 2018; pp. 37–106. [Google Scholar]
  62. Salyers, A.; Vercellotti, J.R.; West, S.E.; Wilkins, T.D. Fermentation of mucin and plant polysaccharides by strains of Bacteroides from the human colon. Appl. Environ. Microbiol. 1977, 33, 319–322. [Google Scholar] [CrossRef] [Green Version]
  63. Gherardini, F.C.; Salyers, A.A. Characterization of an outer membrane mannanase from Bacteroides ovatus. J. Bacteriol. 1987, 169, 2031–2037. [Google Scholar] [CrossRef] [Green Version]
  64. Weaver, J.; Whitehead, T.R.; Cotta, M.A.; Valentine, P.C.; Salyers, A.A. Genetic analysis of a locus on the Bacteroides ovatus chromosome which contains xylan utilization genes. Appl. Environ. Microbiol. 1992, 58, 2764–2770. [Google Scholar] [CrossRef] [Green Version]
  65. Macfarlane, G.T.; Englyst, H.N. Starch utilization by the human large intestinal microflora. J. Appl. Bacteriol. 1986, 60, 195–201. [Google Scholar] [CrossRef]
  66. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Gordon, J.I. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef]
  67. 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]
  68. Morowitz, M.J.; Carlisle, E.M.; Alverdy, J.C. Contributions of Intestinal Bacteria to Nutrition and Metabolism in the Critically Ill. Surg. Clin. N. Am. 2011, 91, 771–785. [Google Scholar] [CrossRef] [Green Version]
  69. Zhang, D.; Frenette, P.S. Cross talk between neutrophils and the microbiota. Blood 2019, 133, 2168–2177. [Google Scholar] [CrossRef]
  70. Owaga, E.; Hsieh, R.-H.; Mugendi, B.; Masuku, S.; Shih, C.-K.; Chang, J.-S. Th17 Cells as Potential Probiotic Therapeutic Targets in Inflammatory Bowel Diseases. Int. J. Mol. Sci. 2015, 16, 20841–20858. [Google Scholar] [CrossRef] [Green Version]
  71. Usami, M.; Kishimoto, K.; Ohata, A.; Miyoshi, M.; Aoyama, M.; Fueda, Y.; Kotani, J. Butyrate and trichostatin A attenuate nuclear factor κB activation and tumor necrosis factor α secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr. Res. 2008, 28, 321–328. [Google Scholar] [CrossRef]
  72. Vinolo, M.A.; Rodrigues, H.G.; Hatanaka, E.; Sato, F.T.; Sampaio, S.C.; Curi, R. Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J. Nutr. Biochem. 2011, 22, 849–855. [Google Scholar] [CrossRef]
  73. Tedelind, S.; Westberg, F.; Kjerrulf, M.; Vidal, A. Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: A study with relevance to inflammatory bowel disease. World J. Gastroenterol. 2007, 13, 2826–2832. [Google Scholar] [CrossRef]
  74. Zhang, L.; Jin, S.; Wang, C.; Jiang, R.; Wan, J. Histone Deacetylase Inhibitors Attenuate Acute Lung Injury During Cecal Ligation and Puncture-Induced Polymicrobial Sepsis. World J. Surg. 2010, 34, 1676–1683. [Google Scholar] [CrossRef]
  75. Ye, J.; Qiu, J.; Bostick, J.W.; Ueda, A.; Schjerven, H.; Li, S.; Zhou, L. The aryl hydrocarbon receptor preferentially marks and promotes gut regulatory T cells. Cell Rep. 2017, 21, 2277–2290. [Google Scholar] [CrossRef] [Green Version]
  76. Zelante, T.; Iannitti, R.G.; Cunha, C.; De Luca, A.; Giovannini, G.; Pieraccini, G.; Romani, L. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 2013, 39, 372–385. [Google Scholar] [CrossRef] [Green Version]
  77. Barcik, W.; Wawrzyniak, M.; Akdis, C.A.; O’Mahony, L. Immune regulation by histamine and histamine-secreting bacteria. Curr. Opin. Immunol. 2017, 48, 108–113. [Google Scholar] [CrossRef]
  78. Podolsky, D.K.V. Innate mechanisms of mucosal defense and repair: The best offense is a good defense. Am. J. Physiol. Liver Physiol. 1999, 277, G495–G499. [Google Scholar] [CrossRef]
  79. Yan, F.; Cao, H.; Cover, T.L.; Washington, M.K.; Shi, Y.; Liu, L.; 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]
  80. Lutgendorff, F.; Akkermans, L.M.A.; Soderholm, J.D. The Role of Microbiota and Probiotics in Stress-Induced Gastrointestinal Damage. Curr. Mol. Med. 2008, 8, 282–298. [Google Scholar] [CrossRef]
  81. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Cani, P.D. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef] [Green Version]
  82. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787. [Google Scholar] [CrossRef] [PubMed]
  83. Liu, Y.; Espinosa, C.D.; Abelilla, J.J.; Casas, G.A.; Lagos, L.V.; Lee, S.A.; Stein, H.H. Non-antibiotic feed additives in diets for pigs: A review. Anim. Nutr. 2018, 4, 113–125. [Google Scholar] [CrossRef] [PubMed]
  84. NRC. Nutrient Requirements of Swine, 11th ed.; Natl. Acad. Press: Washington, DC, USA, 2012. [Google Scholar]
  85. Poulsen, H.D. Zinc and copper as feed additives, growth factors or unwanted environmental factors. J. Anim. Feed. Sci. 1998, 7 (Suppl. S1), 135–142. [Google Scholar] [CrossRef] [Green Version]
  86. Smith, J.W.; Tokach, M.D.; Goodband, R.D.; Nelssen, J.L.; Richert, B.T. Effects of the interrelationship between zinc oxide and copper sulfate on growth performance of early-weaned pigs. J. Anim. Sci. 1997, 75, 1861–1866. [Google Scholar] [CrossRef] [PubMed]
  87. Hill, G.M.; Cromwell, G.L.; Crenshaw, T.D.; Dove, C.R.; Ewan, R.C.; Knabe, D.A.; Veum, T.L. Growth promotion effects and plasma changes from feeding high dietary concentrations of zinc and copper to weanling pigs (regional study). J. Anim. Sci. 2000, 78, 1010–1016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Hu, C.; Song, J.; You, Z.; Luan, Z.; Li, W. Zinc Oxide–Montmorillonite Hybrid Influences Diarrhea, Intestinal Mucosal Integrity, and Digestive Enzyme Activity in Weaned Pigs. Biol. Trace Element Res. 2012, 149, 190–196. [Google Scholar] [CrossRef] [PubMed]
  89. Hahn, J.D.; Baker, D.H. Growth and plasma zinc responses of young pigs fed pharmacologic levels of zinc. J. Anim. Sci. 1993, 71, 3020–3024. [Google Scholar] [CrossRef] [PubMed]
  90. Carlson, M.S.; Hill, G.M.; Link, J.E. Early- and traditionally weaned nursery pigs benefit from phase-feeding pharmacological concentrations of zinc oxide: Effect on metallothionein and mineral concentrations. J. Anim. Sci. 1999, 77, 1199–1207. [Google Scholar] [CrossRef]
  91. Rajendran, D. Application of nano minerals in animal production system. Res. J. Biotechnol. 2013, 8, 1–3. [Google Scholar]
  92. Pei, X.; Xiao, Z.; Liu, L.; Wang, G.; Tao, W.; Wang, M.; Leng, D. Effects of dietary zinc oxide nanoparticles supplementation on growth performance, zinc status, intestinal morphology, microflora population, and immune response in weaned pigs. J. Sci. Food Agric. 2019, 99, 1366–1374. [Google Scholar] [CrossRef]
  93. Li, M.-Z.; Huang, J.-T.; Tsai, Y.-H.; Mao, S.-Y.; Fu, C.-M.; Lien, T.-F. Nanosize of zinc oxide and the effects on zinc digestibility, growth performances, immune response and serum parameters of weanling piglets. Anim. Sci. J. 2016, 87, 1379–1385. [Google Scholar] [CrossRef]
  94. Sturniolo, G.C.; Di Leo, V.; Ferronato, A.; D’Odorico, A.; D’Incà, R. Zinc supplementation tightens “leaky gut” in Crohn’s disease. Inflamm. Bowel Dis. 2001, 7, 94–98. [Google Scholar] [CrossRef]
  95. Hu, C.H.; Song, Z.H.; Xiao, K.; Song, J.; Jiao, L.F.; Ke, Y.L. Zinc oxide influences intestinal integrity, the expressions of genes associated with inflammation and TLR4-myeloid differentiation factor 88 signaling pathways in weanling pigs. Innate Immun. 2013, 20, 478–486. [Google Scholar] [CrossRef] [Green Version]
  96. Zhong, W.; McClain, C.J.; Cave, M.; Kang, Y.J.; Zhou, Z. The role of zinc deficiency in alcohol-induced intestinal barrier dysfunction. Am. J. Physiol. Liver Physiol. 2010, 298, G625–G633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Finamore, A.; Massimi, M.; Devirgiliis, L.C.; Mengheri, E. Zinc Deficiency Induces Membrane Barrier Damage and Increases Neutrophil Transmigration in Caco-2 Cells. J. Nutr. 2008, 138, 1664–1670. [Google Scholar] [CrossRef] [Green Version]
  98. Katouli, M.; Melin, L.; Jensen-Waern, M.; Wallgren, P.; Möllby, R. The effect of zinc oxide supplementation on the stability of the intestinal flora with special reference to composition of coliforms in weaned pigs. J. Appl. Microbiol. 1999, 87, 564–573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Yu, T.; Zhu, C.; Chen, S.; Gao, L.; Lv, H.; Feng, R.; Jiang, Z. Dietary high zinc oxide modulates the microbiome of ileum and colon in weaned piglets. Front. Microbiol. 2017, 8, 825. [Google Scholar] [CrossRef] [Green Version]
  100. Vahjen, W.; Pieper, R.; Zentek, J. Increased dietary zinc oxide changes the bacterial core and enterobacterial composition in the ileum of piglets1. J. Anim. Sci. 2011, 89, 2430–2439. [Google Scholar] [CrossRef] [Green Version]
  101. Højberg, O.; Canibe, N.; Poulsen, H.D.; Hedemann, M.S.; Jensen, B.B. Influence of Dietary Zinc Oxide and Copper Sulfate on the Gastrointestinal Ecosystem in Newly Weaned Piglets. Appl. Environ. Microbiol. 2005, 71, 2267–2277. [Google Scholar] [CrossRef] [Green Version]
  102. Starke, I.; Pieper, R.; Vahjen, W.; Zentek, J. The impact of dietary Zinc Oxide on the bacterial diversity of the small intestinal microbiota of weaned piglets. J. Vet. Sci. Technol. 2014, 5, 42424. [Google Scholar]
  103. Starke, I.C.; Pieper, R.; Neumann, K.; Zentek, J.; Vahjen, W. The impact of high dietary zinc oxide on the development of the intestinal microbiota in weaned piglets. FEMS Microbiol. Ecol. 2014, 87, 416–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Ministry of Agriculture of the People’s Republic of China. Regulations on the Safe. Use of Feed Additives Issued by Ministry of Agriculture of the People’s Republic of China (No. 2625); Ministry of Agriculture of the People’s Republic of China: Beijing, China, 2018.
  105. Nitrayova, S.; Windisch, W.; Von Heimendahl, E.; Müller, A.; Bartelt, J. Bioavailability of zinc from different sources in pigs. J. Anim. Sci. 2012, 90, 185–187. [Google Scholar] [CrossRef] [PubMed]
  106. Ward, T.L.; Asche, G.L.; Louis, G.F.; Pollmann, D.S. Zinc-methionine improves growth performance of starter pigs. J. Anim. Sci. 1996, 74, 182. [Google Scholar]
  107. Hollis, G.R.; Carter, S.D.; Cline, T.R.; Crenshaw, T.D.; Cromwell, G.L.; Hill, G.M.; Veum, T.L. Effects of replacing pharmacological levels of dietary zinc oxide with lower dietary levels of various organic zinc sources for weanling pigs. J. Anim. Sci. 2005, 83, 2123–2129. [Google Scholar] [CrossRef]
  108. Raghupathi, K.R.; Koodali, R.T.; Manna, A.C. Size-Dependent Bacterial Growth Inhibition and Mechanism of Antibacterial Activity of Zinc Oxide Nanoparticles. Langmuir 2011, 27, 4020–4028. [Google Scholar] [CrossRef]
  109. Yamamoto, O. Influence of particle size on the antibacterial activity of zinc oxide. Int. J. Inorg. Mater. 2001, 3, 643–646. [Google Scholar] [CrossRef]
  110. Zhang, L.; Jiang, Y.; Ding, Y.; Povey, M.; York, D.W. Investigation into the antibacterial behaviour of suspensions of ZnO nanoparticles (ZnO nanofluids). J. Nanoparticle Res. 2007, 9, 479–489. [Google Scholar] [CrossRef]
  111. Yamamoto, O.; Hotta, M.; Sawai, J.; Sasamoto, T.; Kojima, H. Influence of Powder Characteristic of ZnO on Antibacterial Activity. J. Ceram. Soc. Jpn. 1998, 106, 1007–1011. [Google Scholar] [CrossRef] [Green Version]
  112. Sawai, J.; Igarashi, H.; Hashimoto, A.; Kokugan, T.; Shimizu, M. Effect of particle size and heating temperature of ceramic powders on antibacterial activity of their slurries. J. Chem. Eng. Jpn. 1996, 29, 251–256. [Google Scholar] [CrossRef] [Green Version]
  113. Sawai, J.; Shoji, S.; Igarashi, H.; Hashimoto, A.; Kokugan, T.; Shimizu, M.; Kojima, H. Hydrogen peroxide as an antibacterial factor in zinc oxide powder slurry. J. Ferment. Bioeng. 1998, 86, 521–522. [Google Scholar] [CrossRef]
  114. Lipovsky, A.; Nitzan, Y.; Gedanken, A.; Lubart, R. Antifungal activity of ZnO nanoparticles—The role of ROS mediated cell injury. Nanotechnology 2011, 22, 105101. [Google Scholar] [CrossRef]
  115. Song, W.; Zhang, J.; Guo, J.; Zhang, J.; Ding, F.; Li, L.; Sun, Z. Role of the dissolved zinc ion and reactive oxygen species in cytotoxicity of ZnO nanoparticles. Toxicol. Lett. 2010, 199, 389–397. [Google Scholar] [CrossRef]
  116. Stoimenov, P.K.; Klinger, R.L.; Marchin, G.L.; Klabunde, K.J. Metal Oxide Nanoparticles as Bactericidal Agents. Langmuir 2002, 18, 6679–6686. [Google Scholar] [CrossRef]
  117. Zhang, L.; Ding, Y.; Povey, M.; York, D. ZnO nanofluids–A potential antibacterial agent. Prog. Nat. Sci. 2008, 18, 939–944. [Google Scholar] [CrossRef]
  118. Brayner, R.; Ferrari-Iliou, R.; Brivois, N.; Djediat, S.; Benedetti, A.M.F.; Fiévet, F. Toxicological Impact Studies Based on Escherichia coli Bacteria in Ultrafine ZnO Nanoparticles Colloidal Medium. Nano Lett. 2006, 6, 866–870. [Google Scholar] [CrossRef]
  119. Kitts, D.D.; Weiler, K. Bioactive proteins and peptides from food sources. Applications of bioprocesses used in isolation and recovery. Curr. Pharm. Des. 2003, 9, 1309–1323. [Google Scholar] [CrossRef]
  120. Korhonen, H.; Pihlanto, A. Bioactive peptides: Production and functionality. Int. Dairy J. 2006, 16, 945–960. [Google Scholar] [CrossRef]
  121. Lee, S.-H.; Kim, S.-J.; Lee, Y.-S.; Song, M.-D.; Kim, I.-H.; Won, H.-S. De novo generation of short antimicrobial peptides with simple amino acid composition. Regul. Pept. 2011, 166, 36–41. [Google Scholar] [CrossRef] [PubMed]
  122. Zasloff, M. Antimicrobial peptides of multicellular organisms. Nat. Cell Biol. 2002, 415, 389–395. [Google Scholar] [CrossRef] [PubMed]
  123. Kumar, P.; Kizhakkedathu, J.N.; Straus, S.K. Antimicrobial Peptides: Diversity, Mechanism of Action and Strategies to Improve the Activity and Biocompatibility In Vivo. Biomolecules 2018, 8, 4. [Google Scholar] [CrossRef] [Green Version]
  124. Shai, Y. Mechanism of the binding, insertion and destabilization of phospholipid bilayer membranes by α-helical antimicrobial and cell non-selective membrane-lytic peptides. Biochim. Biophys. Acta (BBA)-Biomembr. 1999, 1462, 55–70. [Google Scholar] [CrossRef] [Green Version]
  125. Pouny, Y.; Rapaport, D.; Mor, A.; Nicolas, P.; Shai, Y. Interaction of antimicrobial dermaseptin and its fluorescently labeled analogs with phospholipid membranes. Biochemistry 1992, 31, 12416–12423. [Google Scholar] [CrossRef]
  126. Matsuzaki, K.; Murase, O.; Fujii, N.; Miyajima, K. An Antimicrobial Peptide, Magainin 2, Induced Rapid Flip-Flop of Phospholipids Coupled with Pore Formation and Peptide Translocation. Biochemistry 1996, 35, 11361–11368. [Google Scholar] [CrossRef]
  127. Brogden, K.A. Antimicrobial peptides: Pore formers or metabolic inhibitors in bacteria? Nat. Rev. Genet. 2005, 3, 238–250. [Google Scholar] [CrossRef]
  128. Park, C.B.; Kim, H.S.; Kim, S.C. Mechanism of Action of the Antimicrobial Peptide Buforin II: Buforin II Kills Microorganisms by Penetrating the Cell Membrane and Inhibiting Cellular Functions. Biochem. Biophys. Res. Commun. 1998, 244, 253–257. [Google Scholar] [CrossRef] [Green Version]
  129. Malanovic, N.; Lohner, K. Antimicrobial Peptides Targeting Gram-Positive Bacteria. Pharmaceuticals 2016, 9, 59. [Google Scholar] [CrossRef] [Green Version]
  130. Münch, D.; Sahl, H.-G. Structural variations of the cell wall precursor lipid II in Gram-positive bacteria—Impact on binding and efficacy of antimicrobial peptides. Biochim. Biophys. Acta (BBA)-Biomembr. 2015, 1848, 3062–3071. [Google Scholar] [CrossRef] [Green Version]
  131. Hilchie, A.L.; Wuerth, K.; Hancock, R.E.W. Immune modulation by multifaceted cationic host defense (antimicrobial) peptides. Nat. Chem. Biol. 2013, 9, 761–768. [Google Scholar] [CrossRef]
  132. Afacan, N.J.; Yeung, A.T.; Pena, O.M.; Hancock, R.E. Therapeutic Potential of Host Defense Peptides in Antibiotic-resistant Infections. Curr. Pharm. Des. 2012, 18, 807–819. [Google Scholar] [CrossRef] [Green Version]
  133. Mader, J.S.; Hoskin, D.W. Cationic antimicrobial peptides as novel cytotoxic agents for cancer treatment. Expert Opin. Investig. Drugs 2006, 15, 933–946. [Google Scholar] [CrossRef]
  134. Payoungkiattikun, W.; Joompang, A.; Thongchot, S.; Nowichai, B.; Jangpromma, N.; Klaynongsruang, S. Evidence of multi-functional peptide activity: Potential role of KT2 and RT2 for anti-inflammatory, anti-oxidative stress, and anti-apoptosis properties. Appl. Biol. Chem. 2020, 63, 1–13. [Google Scholar] [CrossRef]
  135. Gitler, C. Protein Digestion and Absorption in Nonruminants. Mamm. Protein Metab. 1964, 1, 35–69. [Google Scholar] [CrossRef]
  136. Chung, Y.C.; Kim, Y.S.; Shadchehr, A.; Garrido, A.; Macgregor, I.L.; Sleisenger, M.H. Protein digestion and absorption in human small intestine. Gastroenterology 1979, 76, 1415–1421. [Google Scholar] [CrossRef]
  137. Silk, D.B.; Chung, Y.C.; Berger, K.L.; Conley, K.; Beigler, M.; Sleisenger, M.H.; Kim, Y.S. Comparison of oral feeding of peptide and amino acid meals to normal human subjects. Gut 1979, 20, 291–299. [Google Scholar] [CrossRef]
  138. Webb, K., Jr. Amino acid and peptide absorption from the gastrointestinal tract. Fed. Proc. 1986, 45, 2268–2271. [Google Scholar]
  139. Zimmerman, D. Interaction of Intestinal Hydrolysate and Spray-Dried Plasma Fed to Weanling Pigs; Experiment; Iowa State University: Ames, IA, USA, 1996; p. 9615. [Google Scholar]
  140. Zimmerman, D. The Duration of Carry-Over Growth Response to Intestinal Hydrolysate Fed to Weanling Pigs; Experiment; Iowa State University: Ames, IA, USA, 1996; p. 9612. [Google Scholar]
  141. Kim, J.H.; Chae, B.J.; Kim, Y.G. Effects of Replacing Spray Dried Plasma Protein with Spray Dried Porcine Intestine Hydrolysate on Ileal Digestibility of Amino Acids and Growth Performance in Early-Weaned Pigs. Asian-Australas. J. Anim. Sci. 2000, 13, 1738–1742. [Google Scholar] [CrossRef]
  142. Opheim, M.; Sterten, H.; Øverland, M.; Kjos, N. Atlantic salmon (Salmo salar) protein hydrolysate—Effect on growth performance and intestinal morphometry in broiler chickens. Livest. Sci. 2016, 187, 138–145. [Google Scholar] [CrossRef]
  143. Tang, X.-S.; Tang, Z.-R.; Wang, S.-P.; Feng, Z.-M.; Zhou, N.; Li, T.-J.; Yin, Y.-L. Expression, Purification, and Antibacterial Activity of Bovine Lactoferrampin–Lactoferricin in Pichia pastoris. Appl. Biochem. Biotechnol. 2011, 166, 640–651. [Google Scholar] [CrossRef]
  144. Yin, Y.; Tang, X.; Fatufe, A.A.; Tang, Z.; Wang, S.; Liu, Z.; Li, T.-J. Dietary Supplementation with Recombinant Lactoferrampin-Lactoferricin Improves Growth Performance and Affects Serum Parameters in Piglets. J. Anim. Veter-Adv. 2012, 11, 2548–2555. [Google Scholar] [CrossRef] [Green Version]
  145. Xiong, X.; Yang, H.S.; Li, L.; Wang, Y.F.; Huang, R.L.; Li, F.N.; Qiu, W. Effects of antimicrobial peptides in nursery diets on growth performance of pigs reared on five different farms. Livest. Sci. 2014, 167, 206–210. [Google Scholar] [CrossRef]
  146. Yoon, J.H.; Ingale, S.L.; Kim, J.S.; Kim, K.H.; Lee, S.H.; Park, Y.K.; Chae, B.J. Effects of dietary supplementation of antimicrobial peptide-A3 on growth performance, nutrient digestibility, intestinal and fecal microflora and intestinal morphology in weanling pigs. Anim. Feed Sci. Technol. 2012, 177, 98–107. [Google Scholar] [CrossRef]
  147. Yoon, J.H.; Ingale, S.L.; Kim, J.S.; Kim, K.H.; Lohakare, J.; Park, Y.K.; Chae, B.J. Effects of dietary supplementation with antimicrobial peptide-P5 on growth performance, apparent total tract digestibility, faecal and intestinal microflora and intestinal morphology of weanling pigs. J. Sci. Food Agric. 2013, 93, 587–592. [Google Scholar] [CrossRef]
  148. Shan, T.; Wang, Y.; Liu, J.; Xu, Z. Effect of dietary lactoferrin on the immune functions and serum iron level of weanling piglets1. J. Anim. Sci. 2007, 85, 2140–2146. [Google Scholar] [CrossRef]
  149. Xiao, H.; Wu, M.M.; Tan, B.E.; Yin, Y.L.; Li, T.J.; Xiao, D.F.; Li, L. Effects of composite antimicrobial peptides in weanling piglets challenged with deoxynivalenol: I. Growth performance, immune function, and antioxidation capacity1. J. Anim. Sci. 2013, 91, 4772–4780. [Google Scholar] [CrossRef]
  150. Xiao, H.; Tan, B.E.; Wu, M.M.; Yin, Y.L.; Li, T.J.; Yuan, D.X.; Li, L. Effects of composite antimicrobial peptides in weanling piglets challenged with deoxynivalenol: II. Intestinal morphology and function1. J. Anim. Sci. 2013, 91, 4750–4756. [Google Scholar] [CrossRef]
  151. Wang, Y.-Z.; Shan, T.-Z.; Xu, Z.-R.; Feng, J.; Wang, Z.-Q. Effects of the lactoferrin (LF) on the growth performance, intestinal microflora and morphology of weanling pigs. Anim. Feed. Sci. Technol. 2007, 135, 263–272. [Google Scholar] [CrossRef]
  152. Tang, Z.; Yin, Y.; Zhang, Y.; Huang, R.; Sun, Z.; Li, T.; Tu, Q. Effects of dietary supplementation with an expressed fusion peptide bovine lactoferricin–lactoferrampin on performance, immune function and intestinal mucosal morphology in piglets weaned at age 21 d. Br. J. Nutr. 2008, 101, 998–1005. [Google Scholar] [CrossRef] [Green Version]
  153. Wu, S.; Zhang, F.; Huang, Z.; Liu, H.; Xie, C.; Zhang, J.; Qiao, S. Effects of the antimicrobial peptide cecropin AD on performance and intestinal health in weaned piglets challenged with Escherichia coli. Peptides 2012, 35, 225–230. [Google Scholar] [CrossRef]
  154. Poudel, P.; Levesque, C.L.; Samuel, R.; St-Pierre, B. Dietary inclusion of Peptiva, a peptide-based feed additive, can accelerate the maturation of the fecal bacterial microbiome in weaned pigs. BMC Veter-Res. 2020, 16, 1–13. [Google Scholar] [CrossRef] [Green Version]
  155. Suiryanrayna, M.V.A.N.; Ramana, J.V. A review of the effects of dietary organic acids fed to swine. J. Anim. Sci. Biotechnol. 2015, 6, 1–11. [Google Scholar] [CrossRef] [Green Version]
  156. Erickson, R.H.; Kim, Y.S. Digestion and absorption of dietary protein. Ann. Rev. Med. 1990, 41, 133–139. [Google Scholar] [CrossRef] [PubMed]
  157. Piper, D.W.; Fenton, B.H. pH stability and activity curves of pepsin with special reference to their clinical importance. Gut 1965, 6, 506–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Barrow, P.A.; Fuller, R.; Newport, M.J. Changes in the Microflora and Physiology of the Anterior Intestinal Tract of Pigs Weaned at 2 Days, with Special Reference to the Pathogenesis of Diarrhea. Infect. Immun. 1977, 18, 586–595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Radcliffe, J.S.; Zhang, Z.; Kornegay, E.T. The effects of microbial phytase, citric acid, and their interaction in a corn-soybean meal-based diet for weanling pigs. J. Anim. Sci. 1998, 76, 1880–1886. [Google Scholar] [CrossRef] [Green Version]
  160. Scipioni, R.; Zaghini, G.; Biavati, B. Researches on the use of acidified diets for early weaning of piglets. Zootec. Nutr. Anim. 1978, 4, 201–218. [Google Scholar]
  161. Burnell, T.W.; Cromwell, G.L.; Stahly, T.S. Effects of Dried Whey and Copper Sulfate on the Growth Responses to Organic Acid in Diets for Weanling Pigs. J. Anim. Sci. 1988, 66, 1100–1108. [Google Scholar] [CrossRef]
  162. Heo, J.M.; Kim, J.C.; Yoo, J.; Pluske, J.R. A between-experiment analysis of relationships linking dietary protein intake and post-weaning diarrhea in weanling pigs under conditions of experimental infection with an enterotoxigenic strain of E scherichia coli. Anim. Sci. J. 2015, 86, 286–293. [Google Scholar] [CrossRef]
  163. Thomlinson, J.R.; Lawrence, T.L. Dietary manipulation of gastric pH in the prophylaxis of enteric disease in weaned pigs: Some field observations. Veter-Rec. 1981, 109, 120–122. [Google Scholar] [CrossRef]
  164. Van Immerseel, F.; Boyen, F.; Gantois, I.; Timbermont, L.; Bohez, L.; Pasmans, F.; Ducatelle, R. Supplementation of coated butyric acid in the feed reduces colonization and shedding of Salmonella in poultry. Poult. Sci. 2005, 84, 1851–1856. [Google Scholar] [CrossRef]
  165. Fernández-Rubio, C.; Ordóñez, C.; Abad-González, J.; Garcia-Gallego, A.; Honrubia, M.P.; Mallo, J.J.; Balaña-Fouce, R. Butyric acid-based feed additives help protect broiler chickens from Salmonella Enteritidis infection. Poult. Sci. 2009, 88, 943–948. [Google Scholar] [CrossRef]
  166. Kirchgessner, M. Fumaric acid as a feed additive in pig nutrition. Pig News Inform. 1982, 3, 259. [Google Scholar]
  167. Blank, R.; Mosenthin, R.; Sauer, W.C.; Huang, S. Effect of fumaric acid and dietary buffering capacity on ileal and fecal amino acid digestibilities in early-weaned pigs. J. Anim. Sci. 1999, 77, 2974–2984. [Google Scholar] [CrossRef] [Green Version]
  168. Falkowski, J.F.; Aherne, F.X. Fumaric and Citric Acid as Feed Additives in Starter Pig Nutrition. J. Anim. Sci. 1984, 58, 935–938. [Google Scholar] [CrossRef]
  169. Giesting, D.W.; Easter, R.A. Response of Starter Pigs to Supplementation of Corn-Soybean Meal Diets with Organic Acids. J. Anim. Sci. 1985, 60, 1288–1294. [Google Scholar] [CrossRef] [Green Version]
  170. Halas, D.; Hansen, C.; Hampson, D.; Mullan, B.; Kim, J.; Wilson, R.; Pluske, J. Dietary supplementation with benzoic acid improves apparent ileal digestibility of total nitrogen and increases villous height and caecal microbial diversity in weaner pigs. Anim. Feed. Sci. Technol. 2010, 160, 137–147. [Google Scholar] [CrossRef]
  171. Diao, H.; Zheng, P.; Yu, B.; He, J.; Mao, X.; Yu, J.; Chen, D. Effects of benzoic acid on growth performance, serum biochemical parameters, nutrient digestibility and digestive enzyme activities of jejunal digesta in weaner piglets. Chin. J. Anim. Nutr. 2013, 25, 768–777. [Google Scholar]
  172. Piva, A.; Morlacchini, M.; Casadei, G.; Gatta, P.P.; Biagi, G.; Prandini, A. Sodium butyrate improves growth performance of weaned piglets during the first period after weaning. Ital. J. Anim. Sci. 2002, 1, 35–41. [Google Scholar] [CrossRef]
  173. Fang, C.L.; Sun, H.; Wu, J.; Niu, H.H.; Feng, J. Effects of sodium butyrate on growth performance, haematological and immunological characteristics of weanling piglets. J. Anim. Physiol. Anim. Nutr. 2013, 98, 680–685. [Google Scholar] [CrossRef]
  174. Ahmed, S.T.; Hwang, J.A.; Hoon, J.; Mun, H.S.; Yang, C.J. Comparison of Single and Blend Acidifiers as Alternative to Antibiotics on Growth Performance, Fecal Microflora, and Humoral Immunity in Weaned Piglets. Asian-Australas. J. Anim. Sci. 2014, 27, 93–100. [Google Scholar] [CrossRef]
  175. Gabert, V.; Sauer, W. The effect of fumaric acid and sodium fumarate supplementation to diets for weanling pigs on amino acid digestibility and volatile fatty acid concentrations in ileal digesta. Anim. Feed. Sci. Technol. 1995, 53, 243–254. [Google Scholar] [CrossRef]
  176. Gabert, V.M.; Sauer, W.C.; Schmitz, M.; Ahrens, F.; Mosenthin, R. The effect of formic acid and buffering capacity on the ileal digestibilities of amino acids and bacterial populations and metabolites in the small intestine of weanling pigs fed semipurified fish meal diets. Can. J. Anim. Sci. 1995, 75, 615–623. [Google Scholar] [CrossRef]
  177. Eckel, B.; Kirchgessner, M.; Roth, F.X. Influence of formic acid on daily weight gain, feed intake, feed conversion rate and digestibility, 1: Investigations about the nutritive efficacy of organic acids in the rearing of piglets. J. Anim. Physiol. Anim. Nutr. 1992, 67, 93–100. [Google Scholar] [CrossRef]
  178. Guggenbuhl, P.; Séon, A.; Quintana, A.P.; Nunes, C.S. Effects of dietary supplementation with benzoic acid (VevoVitall®) on the zootechnical performance, the gastrointestinal microflora and the ileal digestibility of the young pig. Livest. Sci. 2007, 108, 218–221. [Google Scholar] [CrossRef]
  179. Food and Agriculture Organization; World Health Organization. Health and Nutritional Properties of Probiotics in Food Including Powder Milk with Live Lactic acid Bacteria. Report of a Joint FAO/WHO Expert Consultation on Evaluation of Health and Nutritional Properties of Probiotics in Food Including Powder Milk with Live Lactic Acid Bacteria; WHO: Geneva, Switzerland, 2001. [Google Scholar]
  180. Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic Species in the Modulation of Gut Microbiota: An Overview. BioMed Res. Int. 2018, 2018, 1–8. [Google Scholar] [CrossRef] [Green Version]
  181. Kyriakis, S.; Tsiloyiannis, V.; Vlemmas, J.; Sarris, K.; Tsinas, A.; Alexopoulos, C.; Jansegers, L. The effect of probiotic LSP 122 on the control of post-weaning diarrhoea syndrome of piglets. Res. Veter-Sci. 1999, 67, 223–228. [Google Scholar] [CrossRef]
  182. Chang, Y.H.; Kim, J.K.; Kim, H.J.; Kim, W.Y.; Kim, Y.B.; Park, Y.H. Probiotic effects of Lactobacillus reuteri BSA-131 on piglets. Korean J. Appl. Microb. Biotechnol. 2000, 28, 8–13. [Google Scholar]
  183. Xuan, Z.N.; Kim, J.D.; Heo, K.N.; Jung, H.J.; Lee, J.H.; Han, Y.K.; Han, I.K. Study on the development of a probiotics complex for weaned pigs. Asian-Aust. J. Anim. Sci. 2001, 14, 1425–1428. [Google Scholar]
  184. Giang, H.H.; Viet, T.Q.; Ogle, B.; Lindberg, J.E. Growth performance, digestibility, gut environment and health status in weaned piglets fed a diet supplemented with potentially probiotic complexes of lactic acid bacteria. Livest. Sci. 2010, 129, 95–103. [Google Scholar] [CrossRef]
  185. Huang, C.; Qiao, S.; Li, D.; Piao, X.; Ren, J. Effects of Lactobacilli on the Performance, Diarrhea Incidence, VFA Concentration and Gastrointestinal Microbial Flora of Weaning Pigs. Asian-Australas. J. Anim. Sci. 2004, 17, 401–409. [Google Scholar] [CrossRef]
  186. Zhao, P.; Kim, I. Effect of direct-fed microbial on growth performance, nutrient digestibility, fecal noxious gas emission, fecal microbial flora and diarrhea score in weanling pigs. Anim. Feed. Sci. Technol. 2015, 200, 86–92. [Google Scholar] [CrossRef]
  187. Frantz, N.Z.; Nelssen, J.L.; DeRouchey, J.M.; Goodband, R.D.; Tokach, M.D.; Dritz, S.S. Effects of a prebiotic, Inulin, and a direct fed microbial on growth performance of weanling pigs. Kans. Agric. Exp. Stn. Res. Rep. 2003, 123–127. [Google Scholar] [CrossRef] [Green Version]
  188. Hu, Y.; Dun, Y.; Li, S.; Zhang, D.; Peng, N.; Zhao, S.; Liang, Y. Dietary Enterococcus faecalis LAB31 Improves Growth Performance, Reduces Diarrhea, and Increases Fecal Lactobacillus Number of Weaned Piglets. PLoS ONE 2015, 10, e0116635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  189. Chiang, M.-L.; Chen, H.-C.; Chen, K.-N.; Lin, Y.-C.; Lin, Y.-T.; Chen, M.-J. Optimizing Production of Two Potential Probiotic Lactobacilli Strains Isolated from Piglet Feces as Feed Additives for Weaned Piglets. Asian-Australas. J. Anim. Sci. 2015, 28, 1163–1170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  190. Dowarah, R.; Verma, A.; Agarwal, N.; Patel, B.; Singh, P. Effect of swine based probiotic on performance, diarrhoea scores, intestinal microbiota and gut health of grower-finisher crossbred pigs. Livest. Sci. 2017, 195, 74–79. [Google Scholar] [CrossRef]
  191. Barszcz, M.; Taciak, M.; Skomiał, J. The effects of inulin, dried Jerusalem artichoke tuber and a multispecies probiotic preparation on microbiota ecology and immune status of the large intestine in young pigs. Arch. Anim. Nutr. 2016, 70, 278–292. [Google Scholar] [CrossRef]
  192. Zhang, W.; Zhu, Y.-H.; Zhou, D.; Wu, Q.; Song, D.; Dicksved, J.; Wang, J.-F. Oral Administration of a Select Mixture of Bacillus Probiotics Affects the Gut Microbiota and Goblet Cell Function following Escherichia coli Challenge in Newly Weaned Pigs of Genotype MUC4 That Are Supposed To Be Enterotoxigenic E. coli F4ab/ac Receptor Negative. Appl. Environ. Microbiol. 2016, 83, e02747-16. [Google Scholar] [CrossRef] [Green Version]
  193. Wang, X.; Tsai, T.; Wei, X.; Zuo, B.; Davis, E.; Rehberger, T.; Zhao, J. Effect of Lactylate and Bacillus subtilis on growth performance, peripheral blood cell profile, and gut microbiota of nursery pigs. Microorganisms 2021, 9, 803. [Google Scholar] [CrossRef]
  194. Shin, D.; Chang, S.Y.; Bogere, P.; Won, K.; Choi, J.Y.; Choi, Y.J.; Heo, J. Beneficial roles of probiotics on the modulation of gut microbiota and immune response in pigs. PLoS ONE 2019, 14, e0220843. [Google Scholar] [CrossRef] [Green Version]
  195. Li, X.Q.; Zhu, Y.H.; Zhang, H.F.; Yue, Y.; Cai, Z.X.; Lu, Q.P.; Wang, J.F. Risks associated with high-dose Lactobacillus rhamnosus in an Escherichia coli model of piglet diarrhoea: Intestinal microbiota and immune imbalances. PLoS ONE 2012, 7, e40666. [Google Scholar] [CrossRef]
  196. Zhu, Y.-H.; Li, X.-Q.; Zhang, W.; Zhou, N.; Liu, H.-Y.; Wang, J.-F. Dose-dependent effects of Lactobacillus rhamnosus on serum interleukin-17 production and intestinal T-cell responses in pigs challenged with Escherichia coli. Appl. Environ. Microbiol. 2014, 80, 1787–1798. [Google Scholar] [CrossRef] [Green Version]
  197. Wei, X. Effects of Commercial Feed Additives on Growth Performance and Gut Microbiota of Nursery Pigs. Ph.D. Thesis, University of Arkansas, Fayetteville, AR, USA, 2020. [Google Scholar]
Figure 1. Negative impacts of weaning stress on swine gut health.
Figure 1. Negative impacts of weaning stress on swine gut health.
Animals 11 01279 g001
Figure 2. Schematic representation of probable mechanisms involved in ZnO antimicrobial activity. (i) The generation of reactive oxygen species (ROS), (ii) release of Zn2+ ions, and (iii) alteration of bacterial membrane permeability.
Figure 2. Schematic representation of probable mechanisms involved in ZnO antimicrobial activity. (i) The generation of reactive oxygen species (ROS), (ii) release of Zn2+ ions, and (iii) alteration of bacterial membrane permeability.
Animals 11 01279 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wei, X.; Tsai, T.; Howe, S.; Zhao, J. Weaning Induced Gut Dysfunction and Nutritional Interventions in Nursery Pigs: A Partial Review. Animals 2021, 11, 1279. https://doi.org/10.3390/ani11051279

AMA Style

Wei X, Tsai T, Howe S, Zhao J. Weaning Induced Gut Dysfunction and Nutritional Interventions in Nursery Pigs: A Partial Review. Animals. 2021; 11(5):1279. https://doi.org/10.3390/ani11051279

Chicago/Turabian Style

Wei, Xiaoyuan, Tsungcheng Tsai, Samantha Howe, and Jiangchao Zhao. 2021. "Weaning Induced Gut Dysfunction and Nutritional Interventions in Nursery Pigs: A Partial Review" Animals 11, no. 5: 1279. https://doi.org/10.3390/ani11051279

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