Open access peer-reviewed chapter

Dietary Intervention to Reduce E. coli Infectious Diarrhea in Young Pigs

Written By

Peng Ji, Xunde Li and Yanhong Liu

Submitted: 04 October 2019 Reviewed: 16 January 2020 Published: 10 February 2020

DOI: 10.5772/intechopen.91219

From the Edited Volume

E. Coli Infections - Importance of Early Diagnosis and Efficient Treatment

Edited by Luis Rodrigo

Chapter metrics overview

998 Chapter Downloads

View Full Metrics

Abstract

Postweaning piglets are immediately imposed to remarkable environmental and psychosocial stressors, which adversely affect their intestinal development and health and predispose them to diarrhea. The ratio of postweaning mortality is 6–10% and may rise up to 20% with poor management strategies. Diarrhea per se accounts for 20–30% of cases of mortality in weanling pigs. E. coli postweaning diarrhea is one of the most important causes of postweaning diarrhea in pigs. This diarrhea is responsible for huge economic losses due to high mortality and morbidity, weight loss, and cost of medication. Burgeoning evidence suggested feed-based intervention are one of the promising measures to prevent postweaning diarrhea and to enhance overall health of weaned pigs. Although the exact protective mechanisms may vary and are still not completely understood, a number of feed ingredients or feed additives are marketed to assist in boosting intestinal immunity and regulating gut microbiota. The promising results have been demonstrated in several nutrients (i.e., functional amino acids, organic acids, micro minerals, nondigestible carbohydrates, and antimicrobial peptides), non-nutrients (i.e., phytochemicals and probiotics), and many other feed additives. The efficiencies of each candidate may differ based on their exact modes of action, the basal diet formulation, and the health status of pigs.

Keywords

  • dietary intervention
  • E. coli infectious diarrhea
  • ETEC
  • young pigs

1. Introduction

Escherichia coli (E. coli), a Gram-negative rod-shaped bacterium, was first discovered in 1885 by Theodor Escherich, who noted that E. coli are highly prevalent in the intestinal microflora of healthy individuals and have potential to cause disease when directly inoculated into extraintestinal sites. Diarrheagenic E. coli can be further divided into six groups: enterotoxigenic E. coli (ETEC), enteropathogenic E. coli, enterohemorrhagic E. coli, enteroinvasive E. coli, diffusely adhering E. coli, and enteroaggregative E. coli [1]. Different groups of diarrheagenic E. coli express different virulence genes, exhibit different adhesion characteristics, and therefore have different mechanisms of pathogenicity. This book chapter only covers the infection caused by ETEC.

ETEC is the major etiological agent causing acute watery diarrhea in postweaning piglets. The duration of diarrheal symptom may be shortened by antibiotic treatment, but ETEC is relative refractory to common antibiotics. A growing evidence suggested some nutritional components (e.g., functional amino acids, nondigestible carbohydrates, etc.) and non-nutrients (e.g., phytochemicals, probiotics, etc.) may provide preventive benefits to control ETEC infection. In general, the compounds listed above are supplemented into animal feed with small amount, also named as feed additives. The exact protective mechanisms are largely unknown and may differ for each compound. However, based on the literature review, these feed additives may alleviate ETEC infection by targeting at least one of the following mechanisms: (1) modification of intestinal microbiota by directly killing pathogens or competitively inhibiting the binding of pathogens and toxins to gut epithelium and (2) regulation or stimulation of host immunity that may include intestinal mucosal immunity and systemic immune defense.

Advertisement

2. E. coli infectious diarrhea

E. coli postweaning diarrhea is an important cause of death in weaned pigs. This diarrhea is responsible for economic losses due to mortality, morbidity, decreased growth performance, and cost of medication [2, 3]. ETEC are the most predominant types of pathogenic E. coli that cause diarrhea in both preweaning and postweaning piglets [4, 5].

2.1 Clinical signs

Clinical signs of ETEC infection in pigs include reduced appetite, depression, weakness, rapid dehydration, watery diarrhea (light orange-colored feces), anorexia, and shock due to hypovolemia and electrolyte imbalance [6, 7]. Cyanotic discoloration may appear on the tip of the nose, the ears, and the abdomen. The rectal temperature of infected pigs is generally normal. Pigs may spontaneously recover within 1 week if the infection is mild. However, severe infection may cause death within 12 hours, even without the symptoms of diarrhea. Dehydration of the carcass and distension of the small intestine by colorless mucoid fluid are most common necropsy characteristics for ETEC-infected pigs. During ETEC infection, bacteria normally line the epithelial cells of the intestine rather than invade the mucosa; gross and histological lesions, therefore, may not be directly caused by the bacteria. However, physiological changes in the intestine may be caused by the toxins released by ETEC [8, 9].

2.2 The pathogenesis of E. coli infection

There are two major virulence factors involved in the pathogenesis of ETEC infection, including the expression of fimbriae that enable the attachment of bacteria to the small intestinal epithelial cells, and the production of toxins by the colonized ETEC [10, 11, 12]. In addition, other structural components from E. coli, such as capsular polysaccharides, cell wall lipopolysaccharides (LPS), and iron-binding proteins, may also be involved in the pathogenesis of ETEC [13]. The endotoxins produced by ETEC could induce intestinal physiological changes, which are leading to the disrupted water and fluid absorption and ion secretion, finally causing dehydration and acidosis. The bacterial structural components could also initial a cascade of immune stimulation, resulting in intestinal inflammation and systemic inflammation [14, 15].

2.2.1 Fimbriae of E. coli

Fimbriae are proteinaceous appendages located at the outer membrane of the bacterial cells. They are straight or kinky shapes. The major role of fimbriae is to facilitate the adhesion and colonization of ETEC at the small intestinal mucosa [16, 17, 18]. The adhesion of bacteria is extremely important for ETEC infection. It will stabilize the location of bacteria in the intestinal lumen, which allow the pathogens with better access to luminal nutrition, facilitate the secretion and delivery of endotoxins through epithelium, and help the bacteria penetrate into the tissue if needed [16, 17, 18]. Diarrheagenic ETEC may express many kinds of fimbria, including F4 (K88), F5 (K99), F6 (987p), F18, etc.; F4 (K88) and F18 ETEC are the most common pathogenic ETEC in young pigs.

F4 fimbriae are typically identified in ETEC isolated from pre- and postweaning pigs. F4 ETEC tend to colonize throughout the whole segments of the small intestine in pigs [19]. F4 fimbriae are encoded by the fae operon, which comprises genes coding for several regulatory proteins, distal tip protein, minor subunits, and a major subunit, FaeG, that enable F4+ ETEC binding to specific receptors on intestinal brush border cells [2, 3, 20]. There are three naturally occurring serological variants of F4 fimbriae, including F4ab, F4ac, and F4ad. They are interchangeable by changing a residue stretch in the FaeG protein. However, F4ac variant is the most common F4 fimbriae variant expressed in porcine pathogenic ETEC in the United States [2, 3]. The adhesion receptors of F4 fimbriae appear to be glycoconjugates, including glycoproteins and glycolipids, which have been identified from the brush borders of epithelial cells, intestinal membranes, and mucosa [21, 22]. It is interesting to note that F4ad adhesin appears to preferentially bind to glycolipids, whereas F4ab and F4ac adhesins preferentially bind to glycoproteins [22, 23, 24].

F18 fimbriae are associated with E. coli strains isolated from postweaning diarrhea and edema disease in pigs. These fimbriae are long flexible appendages that show a characteristic zigzag pattern [16]. Based on morphological, serological, functional, and genetic characteristics, two antigenic variants of F18 fimbriae were determined and designated: F18ab and F18ac [25]. F18ab-positive strains are usually isolated from cases of edema disease, whereas F18ac-positive strains are associated with cases of postweaning diarrhea [16, 26]. F18 fimbriae are composed of protein subunits (FedA) with molecular weights of approximately 15.1 kDa [27]. Five structural genes (fedA, fedB, fedC, fedE, and fedF) encoded on a plasmid have been identified [28]. Among these genes, fedE and fedF genes are essential for F18 adhesion and fimbrial length [29]. However, receptors for F18 fimbriae actually increase with age and have not been detected in newborn pigs [30]. This may in part explain the reason why ETEC strains carrying F18 are more prevalent in weaned pigs.

2.2.2 Toxin effects

After adhering to the small intestinal surface, ETEC induce enteric infectious disease and diarrhea through release of enterotoxins, which stimulate copious secretion by the small intestinal mucosa. The enterotoxins include heat-labile toxin, heat-stable toxin, LPS, and Shiga toxins.

Heat-labile toxins. Heat-labile toxin mainly accumulates in the periplasmic space, with limited amount appears on the surface of the bacteria. Heat-labile toxin consists of a single A subunit and five B subunits. The binding of B subunits to the monosialotetrahexosylganglioside (GM1) ganglioside on the cell surface facilitates the translocation of a fragment of A domain into the cell, which then activates the adenylate cyclase system and increases the expression of cyclic adenosine monophosphate (cAMP) [12]. Several physiological changes are mediated by the increased cAMP. First, cAMP stimulates the phosphorylation of the cystic fibrosis transmembrane conductance regulator (CFTR), inducing chloride secretion from the apical region of enterocytes [31]. Second, cAMP stimulates the activation of an apical chloride channel and a basolateral Na/K/2Cl cotransporter, which results in the release of prostaglandin E2 and vasoactive intestinal peptide and loosening of tight junctions [32, 33]. These activities all contribute to increased chloride secretion, reduced sodium absorption, and a concomitant massive loss of water into the intestinal lumen. The effect of heat-labile toxin is irreversible [12].

2.2.2.1 Heat-stable toxins

Heat-stable toxins in porcine isolates are further classified as STa and STb [32]. STa is a small, non-immunogenic protein with a molecular weight of approximately 2 kDa [34]. The major receptor for STa is a particular transmembrane form of guanylate cyclase (GC-C) [35]. Therefore, STa can stimulate the GC-C system, leading to excessive levels of cyclic guanosine monophosphate (cGMP) in enterocytes. Signals resulting from cGMP accumulation induce the activation of CFTR, elevated secretion of Cl- and water in crypt cells, but reduced Na+ and Cl absorption from cells at the tips of villi [11]. Heat-stable toxin a mainly induces small intestinal fluid secretion in newborn but not in weaned pigs [4].

Heat-stable toxin b is a 48-amino acid protein with a molecular weight of approximately 5.1 kDa [36]. STb is antigenically and genetically unrelated to STa and is poorly immunogenic [37]. Production of STb is restricted to porcine ETEC [13]. The mechanisms of action and molecular characteristics of STb are still less known than heat-labile toxins and STa. STb does not stimulate an increase in intracellular levels of either cAMP or cGMP, either Na+ or Cl [38], but stimulates the secretion of HCO3− from intestinal epithelial cells [10, 39]. Heat-stable toxin b causes mild histological damage in the intestinal epithelium, including loss of villous epithelial cells and villous atrophy. This damage may be responsible for impaired absorption of fluids [40]. Another proposed mechanism of action is that STb could increase the level of prostaglandins by opening a G-protein-linked receptor-operated calcium channel in the plasma membrane and elevating intracellular Ca++ [41, 42]. STb can induce small intestinal fluid secretion in newborn and weaned pigs [4].

2.2.2.2 LPS

Lipopolysaccharide is the major surface component of the outer membrane of most Gram-negative bacteria, including ETEC [43]. LPS consists of three distinct regions, lipid A, core oligosaccharides, and the O-antigen polysaccharide with the structural variability from low to high. Lipid A is the primary immunostimulatory component of LPS and is highly recognized by numerous cellular signaling pathways in the innate immunity. The receptors that respond to LPS are mainly located on the cells in the innate immune system, such as macrophages and endothelial cells [44]. Therefore, LPS not only contributes to the physiological membrane functions but also plays an important role in the pathogenesis of Gram-negative bacterial infection [44]. The pathogenic impacts of LPS are mainly through stimulating immune cells of the host, resulting in the release of large amounts of cytokines. The CD14, expressed on monocytes or macrophages, are highly involved in this process. Briefly, the soluble CD14 (sCD14) and LPS-binding protein facilitate the interaction of LPS and the membrane CD14 (mCD14). After binding to mCD14 in the cell surface, LPS is recognized by TLR4-MD-2 complex, which transduces intracellular LPS signals via several signal pathways [45], ultimately resulting in the activation of NFκB and subsequently the release of inflammatory cytokines [15].

2.2.2.3 Shiga toxin

Some ETEC strains that cause postweaning diarrhea possess additional genes that encode Shiga toxin, allowing them to cause edema disease as well [4]. Similar to heat-labile toxin, Shiga toxin is also a protein toxin, consisting of one A subunit and five B subunits. However, Shiga toxin has completely different mechanisms infecting cells, in comparison with heat-labile toxins. Briefly, Shiga toxin first binds to the cells that possess the glycolipid receptors, globotriaosylceramide (Gb)3 or Gb4 [46, 47]. After binding, Shiga toxin is transported to the Golgi apparatus through endocytosis. The Golgi apparatus further transports Shiga toxin to the endoplasmic reticulum, where the subunit A is cleaved by trypsin and is separated into A1 and A2 subunits. The A1 subunit is released into the cytosol and subsequently impacts ribosomes [14]. Shiga toxin can inhibit protein synthesis and induce synthesis of cytokines, including IL-1, IL-6, IL-8, and TNF-α [47, 48]. In addition, Shiga toxin also induces DNA degradation and release of the cellular contents that facilitate proteolytic attack on neighboring cells, contribute to cell apoptosis, and have a toxic effect in the whole organism [14].

Advertisement

3. Dietary intervention on E. coli infectious diarrhea

Nutrients are compounds in feed ingredients that are essential to animal maintenance and production, by providing animals with energy, the building components for repair and growth, and the substances to regulate biological processes. Nutrients are generally grouped into six major classes: water, carbohydrates, proteins, lipids, minerals, and vitamins. With the exception of carbohydrates, all five classes of nutrients are indispensable and have to be provided through animal feed. In addition, a group of specific nutrients, such as functional amino acids, nondigestible carbohydrates, short-chain fatty acids (SCFA), and several micro minerals, has beneficial effects on animal health and performance beyond their nutritional contributions. Recently, a novel concept, non-nutrients, is illuminated to describe a group of dietary compounds, which has no nutrient contribution to animals, but have physiological activities beyond the nutritional pyramid, formulation practices, and feeding methods that similarly alter physiological condition. Emerging evidence suggested that several non-nutrient feed additives (i.e., plant extracts, probiotics, enzymes, etc.) improved animal health through modulating microbial ecology in the digestive tract and/or enhancing immune responses of animals to enteric infections.

3.1 Functional amino acids

Amino acids are defined as organic substances that contain both amino and carboxyl groups. Amino acids are classified according to their molecular weights, chemical structures, the composition of nitrogen and sulfur, and physiological functions. The 20 common proteinogenic amino acids shared by all animal species are further categorized into indispensable, semi-dispensable (conditional essential), and dispensable amino acids, dependent on their dietary essentiality. Functional amino acids are defined with a group of amino acids that are traditionally classified as dispensable amino acids, but with extra biological functions [49]. The well-investigated functional amino acids are the arginine family, which includes arginine, glutamine, glutamate, aspartate, proline, etc. The basic functions of these amino acids have been well summarized by Wu et al. [49, 50], which include but not limited to (1) providing substrates for the synthesis of tissue protein; (2) impacting hormone synthesis and secretion; (3) regulating endothelial function, vasodilation, and blood flow; (4) affecting nutrient metabolism; and (5) maintaining acid-base balance and whole-body homeostasis.

Large amounts of literature have reported the impacts of dietary supplementation of functional amino acids on health and performance of newly weaned pigs. For example, supplementation of 0.2 to 1% L-arginine could enhance growth performance and alleviate the negative effects of different insults or challenges in young pigs [51, 52, 53, 54]. Administration of proline was shown to improve mucosal proliferation, intestinal morphology, as well as intestinal tight junction of weaned pigs [55]. Dietary supplementation of glutamine or dipeptides that are composed of glutamine has shown positive impacts on intestinal integrity, enzyme activities, and growth performance of weaned pigs [56, 57, 58]. Several mechanisms are highly involved in the benefits of the arginine family on intestinal health of weaned pigs, which could prevent the intestinal dysfunction caused by E. coli infectious diarrhea in weaned pigs. First, these amino acids could provide major fuel for small intestinal epithelial cell proliferation and provide energy required for intestinal ATP-dependent metabolic processes [59]. Second, catabolism of these amino acids provides precursors or substrates for the synthesis of nitric oxide, polyamines, and creatine, which are important regulators in blood flow, intestinal integrity and secretion, and epithelial cell repair and migration [60, 61, 62]. Third, glutamine is also a major substrate for glutathione synthesis, which is an important endogenous antioxidant in cells regulating the homeostasis of free radicals [63]. Fourth, these amino acids could enhance intestinal secretory IgA production via regulating the intestinal microbiota and immunity [53, 64].

3.2 Fatty acids

Dietary fat and lipids are extremely important for animal health and production. They have three major fundamental roles in swine nutrition by providing energy, compound lipids, and steroids to animals. Triglycerides and free fatty acids are the primary forms of metabolic energy storage and transport in the animal body. Short-chain fatty acids and medium-chain fatty acids (MCFA) have recently attracted increased research attention as potential candidates to reduce enteric infectious disease in animal production due to their potential antimicrobial activities [65, 66, 67].

SCFA are fatty acids with a chain of less than six carbon atoms, which are primarily produced by hindgut fermentation of dietary fiber. The most abundant SCFA in the gastrointestinal tract are acetic (C2), propionic (C3), and butyric acid (C4). They are the major fuel source for colonocytes and are essential for maintaining the normal metabolism of colon mucosa, including colonocyte growth and proliferation [68, 69]. Butyric acid has received particular attention and has been widely investigated to enhance disease resistance of weaned pigs. Addition of this acid directly to a swine diet may be limited because of its pungent odor and unpalatable flavor. Thus, the salt form (sodium or calcium) or glyceryl form (monobutyrin or tributyrin) of butyric acid has been adopted in animal feed industry. One major advantage of glyceryl forms in comparison with salt forms is that they stay intact in the stomach and are slowly released as butyrate and/or monobutyrin in the small intestine where pancreatic lipase appears [70]. Many research have confirmed the positive protective effects of sodium butyrate or tributyrin on intestinal health of weaned pigs, as they reduced diarrhea, enhanced gut integrity, and improved overall immunity of newly weaned pigs [71, 72, 73, 74]. The mechanisms resulting in improved disease resistance of weaned pigs are highly associated with the antimicrobial activities of SCFA; however, other mechanisms may be also involved. Butyric acid could penetrate into epithelial cells either by simple diffusion or monocarboxylate transporter [75]. Butyric acid could also bind to G-protein-coupled receptor expressed in epithelial cells or immune cells. The binding will mediate a cascade of immune regulation and regulate large amount of gene expression [76, 77, 78].

MCFA are saturated fatty acids containing 6 to 12 carbon atoms, including caproic (C6), caprylic (C8), capric (C10), and lauric acids (C12). MCFA are naturally occurred lipids that are enriched in animal milk fat and in the oil fraction of various plants, such as coconuts, palm kernels, and Cuphea seeds. Similar to SCFA, MCFA have unpleasant smell; therefore, they are commonly used in their glyceryl forms in animal feed. MCFA may have particular nutritional and metabolic effects on young animals due to their rapid digestion, passive absorption, and obligatory oxidation [79, 80]. Although the evidence for the favorable energetic attributes of MCFA is strong, the results of in vivo studies using weaned pigs have been inconsistent [81]. However, the beneficial effects of MCFA on gut health of weaned pigs have been suggested, as they could influence intestinal morphology and physiology, gut microbiome, and intestinal immunity [80]. More research is necessary in the future to explore the influences of MCFA on disease resistance of weaned pigs.

3.3 Micro minerals

Minerals required in smaller quantities are called micro minerals or trace minerals, which include Zn, Cu, Mn, Fe, Se, and others. Micro minerals have confirmed physiological roles and are needed for normal bodily functions of pigs. However, unlike most other minerals, Cu and Zn have antimicrobial properties, and they are, therefore, often added to animal feed in quantities greater than the amount of nutritional requirements.

Zinc serves as a component or an activator of several metalloenzymes and is involved in many intracellular and intercellular signaling pathways. Zinc also plays important roles in skin and wound healing and in regulating immune system [82]. Zinc deficiency in weaned pigs leads to growth retardation, loss of appetite, skeletal abnormalities, and parakeratosis if Zn concentration in feed is much lower than the requirement of 80–100 mg/kg for nursery pigs [83, 84, 85]. However, pharmacological levels (2000–3000 mg/kg) of inorganic Zn in the form of ZnO have been widely adopted to control postweaning diarrhea and enhance feed intake and overall growth performance [86, 87, 88, 89]. The benefits of pharmacological Zn on disease resistance of weaned pigs are likely related to several mechanisms: enhancement of intestinal integrity [90], regeneration of injured intestinal mucosa, stability of intestinal microbiota diversity [91], reduction of intestinal permeability [92], and modulation of intestinal immunity [93]. However, more recent research indicate that feeding pharmacological ZnO could reduce the digestibility of Ca and P, reduce the effectiveness of microbial phytase in pig diet, and increase the abundance of multiresistant bacteria in weaned pigs [94, 95, 96]. Inclusion of pharmacological levels of ZnO has recently been banned in the European Union due to increased Zn pollution from pigs fed with high Zn diets. Meantime, animal feed industry and nutritionists are actively working together to search alternatives that could replace pharmacological ZnO. For instance, low dose of organic Zn sources (i.e., 125 mg/kg of Zn-methionine) has been confirmed to have beneficial effects that are equivalent to addition of pharmacological ZnO due to their greater bioavailability [97, 98].

Copper is also an essential component of several metalloenzymes including cytochrome oxidase and lysyl oxidase. Copper is highly involved in oxidation-reduction reactions, transport of oxygen and electrons, antioxidant system, and many other metabolic functions, including cellular respiration, tissue pigmentation, hemoglobin formation, and connective tissue development [82]. In general, neonatal pigs only require 5–6 mg/kg of Cu for normal metabolism [85, 99], and Cu requirement decreases as animal gets older. Cu deficiency may lead to critical dysfunctions and hypocuprosis in pigs [100]. Pigs may also suffer from microcytic anemia and bone abnormalities [101, 102]. Addition of pharmacological levels of Cu (125–500 mg/kg) in pig diets has been a common practice to reduce postweaning diarrhea and improve growth performance [103, 104, 105, 106]. The beneficial effects of pharmacological Cu have been attributed to its bacteriostatic and bactericidal properties [107, 108]. Similar to zinc, many Cu forms could be used in animal feed, including copper sulfate (CuSO4), copper chloride (CuCl2), tribasic copper chloride (Cu2(OH)3Cl), and copper citrate. Copper sulfate and copper chloride are the most common supplementing forms. Tribasic copper chloride has been suggested to have similar bioavailability but less negative impacts on phosphorus digestibility and intestinal microbiota than copper sulfate [109, 110, 111]. Chelated Cu, such as Cu citrate, has greater availability than inorganic Cu sources, which may be used in animal feed as low dose, resulting in reduced Cu excretion [112].

3.4 Prebiotics and probiotics

Prebiotics are a category of nutritional compounds that may not share similar structures but have the ability to improve the growth of beneficial microorganism in the gastrointestinal tract. Gibson et al. [113] offered a definition of prebiotics, which contains three key aspects: resistance to digestion, fermentation by the large intestinal microbiota, and a selective effect on the microbiota associated with health-promoting effects. Most well-studied prebiotics are nondigestible oligosaccharides or polysaccharides [114]. For instance, inulin-type prebiotics are a group of nondigestible carbohydrates that mainly comprise fructose, including inulin, oligofructose, and fructo-oligosaccharides. They are commonly used in the pig industry and human foods [115]. Galactooligosaccharides that exist in human milk have been reported to have prebiotic effects by enhancing colonic health of breast-fed infants [116]. Many other naturally occurring prebiotics have been reported as well, including polydextrose, trans-galactooligosaccharides, xylo-oligosaccharides, lactulose, pyrodextrins, and isomalto-oligosaccharides. However, a few other nondigestible carbohydrates are not categorized as prebiotics (e.g., mannan-oligosaccharides, β-glucan etc.), but manifest health-promoting functions [117]. For example, a growing evidence demonstrates that β-glucans, either produced by bacteria or extracted from different sources (i.e., cereal, algae, and fungi), could boost host immunity, therefore enhancing disease resistance of human and animals [118, 119, 120].

Probiotics, also known as direct-fed microbials, are live microorganisms and, when administered in adequate amounts, confer a health benefit on the host [121]. Probiotics are categorized into three main groups, including Bacillus, lactic acid-producing bacteria, and yeast [122]. Based on the Food and Drug Administration instruction, the term probiotics is used for human microbial products, whereas the term direct-fed microbials is used for the US feed industry. However, “probiotics” are interchangeably used with human and animal feed worldwide. Bacillus-based probiotics are spore-forming, which makes them thermostable and able to survive at low pH. Bacillus-based probiotics have been identified as potent producers of extracellular fiber-degrading enzymes, which may aid nutrient digestion and utilization [123]. Lactic acid-producing bacteria are not spore-forming; therefore, their survival during feed processing is of concern [124]. Lactic acid-producing bacteria dominate the gastrointestinal tract of the nursing pig [125], which helps reduce the pH in the gut by producing lactic acid through fermentation, inhibiting enteric pathogens [126], and improving host immunity [124, 127]. However, after weaning of pigs, the concentration of lactic acid-producing bacteria diminishes; therefore, supplementation of weaned pig diets with lactic acid-producing probiotics may be beneficial [122]. Yeast include a broad range of products that may be available in pig feed, including whole live yeast cells, heat-treated yeast cells, ground yeast cells, purified yeast cell cultures, and yeast extracts. The efficacy of yeast-based products varies depending on their forms. Yeast or yeast-based product supplementation may boost feed intake and overall growth performance, augment mucosal immunity, promote intestinal development, adsorb mycotoxins, reduce postweaning diarrhea, and modulate gut microbiota in weaned pigs [128, 129, 130, 131].

The most notable effect of prebiotics and probiotics is their modification of intestinal microbiota. They may control or prevent pathogenic bacterial infection by specifically stimulating the growth of beneficial microorganisms in the intestine. The beneficial microorganisms may include but not limited to Bifidobacteria and Lactobacilli, which have confirmed benefit to suppress the growth of pathogenic microorganisms, such as E. coli, through the potential mechanisms described below. For example, the desired bacteria produce SCFA and lactic acid, which may indirectly and specifically kill or inhibit the growth of pathogens [132]. The production of acids may reduce the pH of the intestinal environment, which is unsupportive of the growth of several pathogens [133]. The desired bacteria may produce antimicrobial compounds such as bacteriocins or antibiotics [134]. The desired bacteria compete the available nutrients against pathogens [135].

Many research articles have been published on the impacts of prebiotics and probiotics on infectious diseases in young pigs. For instance, supplementation of 8% inulin reduce the incidence and severity of postweaning diarrhea, probably by increasing SCFA production in the cecum and proximal colon [136]. The addition of fructo-oligosaccharide prevented the mortality and morbidity of weaned pigs infected with K88 ETEC [137]. Supplementation of β-glucan originated from different sources (yeast or algae) could enhance the resistance of pigs against K88 or F18 ETEC infection [120, 138]. The α-ᴅ-mannans from yeast could bind to mannose-specific receptors that are present on many bacteria such as E. coli and Salmonella spp., which prevents adhesion of these pathogens to the mannose-rich glycoproteins lining the intestinal lumen [128]. Indeed, pigs supplemented with live yeast or a yeast fermentation product had reduced disease-related stress, diarrhea scores, duration of diarrhea, and shedding of E. coli and enhanced intestinal integrity in pigs challenged with ETEC [139, 140, 141]. Supplementation of Bacillus subtilis also enhanced disease resistance and growth performance and reduced diarrhea of weaned pigs infected with F18 ETEC [142].

3.5 Phytochemicals

Phytochemicals are secondary plant metabolites that are either naturally obtained from plant materials or directly synthetized. Phytochemicals are used in solid powder form, as crude extracts, or as concentrated extracts. The extracts are further classified as essential oils or oleoresins based on the extraction methods. Essential oils are volatile lipophilic substances obtained by cold extraction or distillation, whereas oleoresins are derived by nonaqueous solvents [143]. A few examples of well-known phytochemicals are curcumin, flavonoids, phenolic acids, isoflavones, carotenoids, etc. The major bioactive compounds in phytochemicals are polyphenols, terpenoids, alkaloids, or sulfur-containing compounds. However, the composition and concentration of bioactive compounds in different phytochemicals may vary a lot, completely depending on the types of plant, the parts of plant, geographical origins, growing conditions, harvesting seasons, processing techniques, and storage conditions [144]. The in vitro biological properties of many phytochemicals have been well investigated, including antimicrobial, antioxidant, anti-inflammatory, and antiviral effects [145, 146, 147, 148]. Therefore, phytochemicals have been largely applied in food processing, cosmetics, and other areas related to human nutrition and health.

Various phytochemicals have been reported to exhibit a broad spectrum of antimicrobial activities against Gram-negative and Gram-positive bacteria [149, 150, 151]. The potential mechanisms of action of antimicrobial activities of phytochemicals are described below. Many phytochemicals are lipophilic, which could damage bacterial membrane, eventually causing the leakage of intracellular materials and cell death [152, 153, 154, 155, 156]. In addition, the phenolic compounds possess strong antibacterial properties by inhibiting virulence factors, such as enzymes and toxins [157, 158, 159]. Lastly, certain bioactive components may also prevent the development of virulent structure (i.e., flagella) in bacteria, therefore inhibiting ETEC adhesion and toxin binding [160, 161].

Our previously published research reported that dietary supplementation of 10 mg/kg of capsicum oleoresin, garlic botanical, or turmeric oleoresin reduced the frequency of diarrhea and enhanced disease resistance of pigs infected with F18 ETEC [162]. The active components in these phytochemicals are capsaicin, propyl thiosulfonates, and curcuminoides, respectively. The results of gene expression profiles in ileal mucosa indicated that supplementation of these phytochemicals modified the expression of genes related to mucin production, cell membrane integrity, and antigen processing and presentations in ETEC-infected pigs [163]. In addition to the enhanced intestinal mucosal health, pigs fed with those phytochemicals had less recruitment of macrophages and neutrophils in the ileum [162]. These observations also suggest that the weaned pigs supplemented with those phytochemicals actually had less gut inflammation than infected control. The gene expression profile analysis by microarray also confirmed the reduced gut inflammation by feeding those phytochemicals to weaned pigs [163]. The phytochemicals discussed above can be naturally obtained from seasonings that are commonly used in kitchen. Many other phytochemicals have been thoroughly investigated to against ETEC infection as well. For example, the anti-diarrheal activity of back or green tea extract has been revealed, because the reduced net fluid and electrolyte losses were observed when F4 ETEC-infected jejunal segments were perfused with black or green tea extract [164]. The administration of cranberry extract (1 g/L) in drinking water also reduced the diarrhea of F18 ETEC-infected piglets [165].

3.6 Antimicrobial peptides

Antimicrobial peptides, also called host defense peptides, are polypeptides that are naturally occurring molecules in various organisms from prokaryotes to mammals. Antimicrobial peptides can be synthesized as recombinant molecules, such as recombinant lactoferrin, or can be isolated from bacteria, insects, vertebrates, or plants, such as bovine lactoferrin and plant defensins [166, 167]. Most of antimicrobial peptides are cationic (positively charged) and amphiphilic (hydrophobic and hydrophilic). Antimicrobial peptides were firstly discovered in the 1980s. They can be classified into different groups based on the different amino acid components, structures, and biological function. The antimicrobial peptides derived from mammals are mainly classified into two families, defensins or cathelicidins. Defensins are further subgrouped into α-, β-, and θ-defensins according to the spacing patterns of their cysteine residues [168]. Defensins are more abundant in epithelial cells and phagocytic cells, whereas cathelicidins are highly expressed in mammalian neutrophils [169].

Antimicrobial peptides possess a strong and wide-spectrum activity against Gram-negative and Gram-positive bacteria, as well as against parasites, fungi, and viruses [170]. One potential advantage of antimicrobial peptides is that they may kill pathogenic bacteria that are resistant to specific medically important antibiotics [168, 171]. Most antimicrobial peptides are small, positively charged, and amphipathic molecules that allow them to actively interact with bacterial membranes through different models, such as barrel-stave model, carpet model, or toroidal pore model [172, 173]. These properties will also allow them to disrupt cell membrane structure, penetrate into cells, regulate intracellular signaling pathways, and ultimately cause bacterial cell death. Many research findings have demonstrated that antimicrobial peptide treatment could inhibit protein and nucleic acid synthesis, suppress bacterial cell wall synthesis, as well as inhibit enzyme activities in bacteria [174]. In addition to their antibacterial properties, antimicrobial peptides may also act as epithelial “preservatives” or immunomodulators to protect host against enteric infectious agents [166, 175, 176].

The protective effects of antimicrobial peptides on infectious diarrhea and intestinal integrity have been reported in weaned pigs [177, 178]. For example, feeding 0.4% of a mixture of bovine lactoferrin, plant defensins, and active yeast increased intestinal integrity and reduced gut permeability of weaned pigs [178]. Addition of cecropin AD reduced incidence of diarrhea and enhanced intestinal Lactobacilli counts in E. coli-challenged piglets [177]. The regulation of gut microbiota may also attribute to the potential benefits of antimicrobial peptides. Supplementation of recombinant lactoferrin or lactoferramoin-lactoferricin reduced the total viable counts of E. coli and Salmonella but enriched the abundance of Lactobacillus and Bifidobacterium in the colon of weaned pigs [179, 180].

3.7 Lysozyme

Lysozyme, also known as muramidase or N-acetylmuramide glycanhydrolase, is an antimicrobial enzyme that is naturally present in body fluids of all mammalian species [181, 182, 183]. Lysozyme could catalyze the hydrolysis of its natural substrate peptidoglycan that is the major component of bacterial cell wall. The hydrolysis of peptidoglycan eventually results in cell lysis. Gram-positive bacteria have cell walls composed of thick layers of peptidoglycan; they are, therefore, more sensitive to the enzymatic degradation of lysozyme [184]. However, a growing evidence supports that lysozyme also displays bactericidal activity against a variety of Gram-negative strains through non-enzymatic mechanisms [184, 185, 186]. For instance, lysozyme has been found to act synergistically with antimicrobial peptides, lactoferrin, in killing Gram-negative bacteria, such as E. coli [181, 187]. In addition to its antimicrobial activity, lysozyme also exhibited anti-inflammatory property that was mediated through inhibiting neutrophil migration and shown the ability to modulate intestinal microbiota [188].

Consumption of lysozyme-rich milk significantly enhanced the relative abundance of Bifidobacteriaceae and Lactobacillaceae in feces of weaned pigs [189]. Those bacterial families are known for their health-promoting functions in lower gastrointestinal tract of human and pigs. Interestingly, consumption of lysozyme-rich milk reduced the relative abundance of bacteria (Mycobacteriaceae, Streptococcaceae, Campylobacterales) that are associated with diseases in pig feces [189]. In another trial from the same research group, feeding of lysozyme milk reduced the incidence of diarrhea and reduced total bacteria translocation into the mesenteric lymph nodes by 83% in ETEC (O149:F4 strain)-infected pigs [190]. Feeding lysozyme-rich milk also tended to reduce fecal Enterobacteriaceae family, in which many prevalent enteric pathogens such as E. coli and Salmonella belong to [190]. Similar results were also reported in one animal trial focusing on human lysozyme-rich milk [191]. In this experiment, neonatal pigs were used and infected with F4 ETEC. Consumption of human lysozyme-rich milk (approximately 1300 mg/L lysozyme) increased survival rate, reduced diarrhea, and facilitated the recovery of infected pigs [191]. Lysozyme treatment also increased the relative abundance of Lactobacillus in feces and enhanced intestinal integrity and mucosa immunity of these neonatal pigs [191].

Advertisement

4. Conclusions

Accumulating evidence has confirmed the importance of nutritional interventions, including modified feeding strategies and nutrient supplements, in the control of diarrheal diseases, and preventing enteric infection as the use of antibiotics will be progressively restricted in many countries. Interest is particularly growing in the use of probiotics and/or prebiotics to increase the populations of target microbes in the digestive tract, thereby improving gut health and performance of animals. Phytochemicals can be an additional tool that producers use to keep pigs healthy and reduce the negative impacts of disease. Dietary supplementation of certain phytochemicals may enhance disease resistance of pigs by improving gut mucosal integrity and optimizing immune response. There are much more candidates of feed additives/nutritional interventions, which may be effective in regulating intestinal environments and immunity and alleviating postweaning enteric infection (Table 1) [192]. It is very important to keep in mind that the efficiencies of each candidate may differ on the basis of their modes of action, the basal diet formulation, and the health status of pigs.

Strain1Dietary supplementsOutcomeReference
K88Milk from human lysozyme transgenic goatsReduced diarrhea, reduced bacterial translocation in mesenteric lymph nodesBrundige et al. [193]; Cooper et al. [194]; Garas et al. [190]
K88Chito-oligosaccharideReduced diarrheaLiu et al. [195]
K88Combination of raw potato starch and probiotic E. coli strainsReduced diarrhea, enhanced gut microbial diversityKrause et al. [196]
K88, F18Probiotics: Pediococcus acidilactici, Saccharomyces cerevisiae boulardii, Bacillus subtilisReduced ETEC attachment to ileal mucosa, upregulated inflammatory responses in the gutKim et al. [142]; Daudelin et al. [197]
K88Saccharomyces cerevisiae fermented productsEnhanced appetite and ileal digesta bacteria richness, reduced ETEC adhering to the mucosa and colonic ammoniaKiarie et al. [139, 140]
K88Probiotics: Lactobacillus plantarum CJLP243Enhanced growth performance, reduced diarrhea, reduced gut inflammation, enhanced gut barrier functionLee et al. [198]; Yang et al. [199]
K88PhytogenicsEnhanced growth performanceDevi et al. [200]
K88NucleotidesEnhanced growth performance and nutrient digestibility, reduced diarrheaLi et al. [201]
F18Clays (smectite, zeolite, kaolinite)Reduced diarrhea, enhanced gut integritySong et al. [202]; Almeida et al. [203]
F18Phytochemicals (capsicum oleoresin, garlic botanical, turmeric oleoresin)Reduced diarrhea, enhanced gut morphology, decreased systemic and gut mucosal inflammationLiu et al. [162, 163]
K88, F18β-glucanEnhanced gut barrier function, reduced systemic inflammationStuyven et al. [138], Kim et al. [120]

Table 1.

Dietary interventions on enterotoxigenic Escherichia coli infection of weaned pigs.

Modified from Liu and Ji [192].


Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Torres AG, Zhou X, Kaper JB. Adherence of diarrheagenic Escherichia coli strains to epithelial cells. Infection and Immunity. 2005;73(1):18-29
  2. 2. Fairbrother JM, Nadeau E, Gyles CL. Escherichia coli in postweaning diarrhea in pigs: An update on bacterial types, pathogenesis, and prevention strategies. Animal Health Research Reviews. 2005;6(1):17-39
  3. 3. Nagy B, Fekete PZ. Enterotoxigenic Escherichia coli in veterinary medicine. International Journal of Medical Microbiology. 2005;295(6-7):443-454
  4. 4. Nagy B, Fekete PZ. Enterotoxigenic Escherichia coli (ETEC) in farm animals. Veterinary Research. 1999;30(2-3):259-284
  5. 5. Hampson DJ. Postweaning Escherichia coli diarrhea in pigs. In: Gyles CL, editor. Escherichia coli in Domestic Animals and Humans. Guildford: CAB International; 1994. pp. 171-191
  6. 6. Sarmiento JI, Casey TA, Moon HW. Postweaning diarrhea in swine: Experimental model of enterotoxigenic Escherichia coli infection. American Journal of Veterinary Research. 1988;49(7):1154-1159
  7. 7. Nollet H, Deprez P, Van Driessche E, Muylle E. Protection of just weaned pigs against infection with F18+ Escherichia coli by non-immune plasma powder. Veterinary Microbiology. 1999;65(1):37-45
  8. 8. Faubert C, Drolet R. Hemorrhagic gastroenteritis caused by Escherichia coli in piglets: Clinical, pathological and microbiological findings. The Canadian Veterinary Journal = La revue veterinaire canadienne. 1992;33(4):251-256
  9. 9. Berberov EM, Zhou Y, Francis DH, Scott MA, Kachman SD, Moxley RA. Relative importance of heat-labile enterotoxin in the causation of severe diarrheal disease in the gnotobiotic piglet model by a strain of enterotoxigenic Escherichia coli that produces multiple enterotoxins. Infection and Immunity. 2004;72(7):3914-3924
  10. 10. Weikel CS, Guerrant RL. STb enterotoxin of Escherichia coli: Cyclic nucleotide-independent secretion. Ciba Foundation Symposium. 1985;112:94-115
  11. 11. Forte LR, Thorne PK, Eber SL, Krause WJ, Freeman RH, Francis SH, et al. Stimulation of intestinal Cl− transport by heat-stable enterotoxin: Activation of cAMP-dependent protein kinase by cGMP. The American Journal of Physiology. 1992;263(3 Pt 1):C607-C615
  12. 12. O’Brien ADRKH. Protein toxins of Escherichia coli and Salmonella. In: Neidhart FC, Curtiss R, Ingraham JL, Lin ECC, Low KB, Magasanik B, Reznikoff WS, Riley M, et al., editors. Escherichia coli and Salmonella: Cellular and Molecular Biology. Washington, DC: ASM Press; 1996. pp. 2788-2802
  13. 13. Gyles CL. Escherichia coli. In: Gyles CL, Charles O. Thoen editor. Pathogenesis of Bacterial Infections in Animals. Ames, IA: Iowa State University Press; 1993. p. 164-187
  14. 14. Sandvig K. Shiga toxins. Toxicon : Official Journal of the International Society on Toxinology. 2001;39(11):1629-1635
  15. 15. Bannerman DD, Goldblum SE. Mechanisms of bacterial lipopolysaccharide-induced endothelial apoptosis. American Journal of Physiology Lung Cellular and Molecular Physiology. 2003;284(6):L899-L914
  16. 16. Nagy B, Whipp SC, Imberechts H, Bertschinger HU, Dean-Nystrom EA, Casey TA, et al. Biological relationship between F18ab and F18ac fimbriae of enterotoxigenic and verotoxigenic Escherichia coli from weaned pigs with oedema disease or diarrhoea. Microbial Pathogenesis. 1997;22(1):1-11
  17. 17. Isaacson RE, Fusco PC, Brinton CC, Moon HW. In vitro adhesion of Escherichia coli to porcine small intestinal epithelial cells: Pili as adhesive factors. Infection and Immunity. 1978;21(2):392-397
  18. 18. Morris JA, Thorns C, Scott AC, Sojka WJ, Wells GA. Adhesion in vitro and in vivo associated with an adhesive antigen (F41) produced by a K99 mutant of the reference strain Escherichia coli B41. Infection and Immunity. 1982;36(3):1146-1153
  19. 19. Nagy B, Moon HW, Isaacson RE. Colonization of porcine small intestine by Escherichia coli: Ileal colonization and adhesion by pig enteropathogens that lack K88 antigen and by some acapsular mutants. Infection and Immunity. 1976;13(4):1214-1220
  20. 20. Van den Broeck W, Cox E, Oudega B, Goddeeris BM. The F4 fimbrial antigen of Escherichia coli and its receptors. Veterinary Microbiology. 2000;71(3-4):223-244
  21. 21. Blomberg L, Krivan HC, Cohen PS, Conway PL. Piglet ileal mucus contains protein and glycolipid (galactosylceramide) receptors specific for Escherichia coli K88 fimbriae. Infection and Immunity. 1993;61(6):2526-2531
  22. 22. Grange PA, Erickson AK, Levery SB, Francis DH. Identification of an intestinal neutral glycosphingolipid as a phenotype-specific receptor for the K88ad fimbrial adhesin of Escherichia coli. Infection and Immunity. 1999;67(1):165-172
  23. 23. Erickson AK, Baker DR, Bosworth BT, Casey TA, Benfield DA, Francis DH. Characterization of porcine intestinal receptors for the K88ac fimbrial adhesin of Escherichia coli as mucin-type sialoglycoproteins. Infection and Immunity. 1994;62(12):5404-5410
  24. 24. Jin LZ, Zhao X. Intestinal receptors for adhesive fimbriae of enterotoxigenic Escherichia coli (ETEC) K88 in swine—A review. Applied Microbiology and Biotechnology. 2000;54(3):311-318
  25. 25. Rippinger P, Bertschinger HU, Imberechts H, Nagy B, Sorg I, Stamm M, et al. Designations F18ab and F18ac for the related fimbrial types F107, 2134P and 8813 of Escherichia coli isolated from porcine postweaning diarrhoea and from oedema disease. Veterinary Microbiology. 1995;45(4):281-295
  26. 26. Wittig W, Klie H, Gallien P, Lehmann S, Timm M, Tschape H. Prevalence of the fimbrial antigens F18 and K88 and of enterotoxins and verotoxins among Escherichia coli isolated from weaned pigs. Zentralblatt fur Bakteriologie: International Journal of Medical Microbiology. 1995;283(1):95-104
  27. 27. Imberechts H, De Greve H, Lintermans P. The pathogenesis of edema disease in pigs. A review. Veterinary Microbiology. 1992;31(2-3):221-233
  28. 28. Smeds A, Hemmann K, Jakava-Viljanen M, Pelkonen S, Imberechts H, Palva A. Characterization of the adhesin of Escherichia coli F18 fimbriae. Infection and Immunity. 2001;69(12):7941-7945
  29. 29. Imberechts H, Wild P, Charlier G, De Greve H, Lintermans P, Pohl P. Characterization of F18 fimbrial genes fedE and fedF involved in adhesion and length of enterotoxemic Escherichia coli strain 107/86. Microbial Pathogenesis. 1996;21(3):183-192
  30. 30. Nagy B, Casey TA, Whipp SC, Moon HW. Susceptibility of porcine intestine to pilus-mediated adhesion by some isolates of piliated enterotoxigenic Escherichia coli increases with age. Infection and Immunity. 1992;60(4):1285-1294
  31. 31. Thiagarajah JR, Verkman AS. CFTR pharmacology and its role in intestinal fluid secretion. Current Opinion in Pharmacology. 2003;3(6):594-599
  32. 32. Nataro JP, Kaper JB. Diarrheagenic Escherichia coli. Clinical Microbiology Reviews. 1998;11(1):142-201
  33. 33. De Haan L, Hirst TR. Cholera toxin: A paradigm for multi-functional engagement of cellular mechanisms (review). Molecular Membrane Biology. 2004;21(2):77-92
  34. 34. Lallier R, Bernard F, Gendreau M, Lazure C, Seidah NG, Chretien M, et al. Isolation and purification of Escherichia coli heat-stable enterotoxin of porcine origin. Analytical Biochemistry. 1982;127(2):267-275
  35. 35. Schulz S, Green CK, Yuen PS, Garbers DL. Guanylyl cyclase is a heat-stable enterotoxin receptor. Cell. 1990;63(5):941-948
  36. 36. Arriaga YL, Harville BA, Dreyfus LA. Contribution of individual disulfide bonds to biological action of Escherichia coli heat-stable enterotoxin B. Infection and Immunity. 1995;63(12):4715-4720
  37. 37. Dubreuil JD. Escherichia coli STb enterotoxin. Microbiology (Reading, England). 1997;143(Pt 6):1783-1795
  38. 38. Weikel CS, Nellans HN, Guerrant RL. In vivo and in vitro effects of a novel enterotoxin, STb, produced by Escherichia coli. The Journal of Infectious Diseases. 1986;153(5):893-901
  39. 39. Argenzio RA, Liacos J, Berschneider HM, Whipp SC, Robertson DC. Effect of heat-stable enterotoxin of Escherichia coli and theophylline on ion transport in porcine small intestine. Canadian Journal of Comparative Medicine: Revue Canadienne de Medecine Comparee. 1984;48(1):14-22
  40. 40. Whipp SC, Kokue E, Morgan RW, Rose R, Moon HW. Functional significance of histologic alterations induced by Escherichia coli pig-specific, mouse-negative, heat-stable enterotoxin (STb). Veterinary Research Communications. 1987;11(1):41-55
  41. 41. Dreyfus LA, Harville B, Howard DE, Shaban R, Beatty DM, Morris SJ. Calcium influx mediated by the Escherichia coli heat-stable enterotoxin B (STB). Proceedings of the National Academy of Sciences of the United States of America. 1993;90(8):3202-3206
  42. 42. Harville BA, Dreyfus LA. Involvement of 5-hydroxytryptamine and prostaglandin E2 in the intestinal secretory action of Escherichia coli heat-stable enterotoxin B. Infection and Immunity. 1995;63(3):745-750
  43. 43. Alexander C, Rietschel ET. Bacterial lipopolysaccharides and innate immunity. Journal of Endotoxin Research. 2001;7(3):167-202
  44. 44. Raetz CR, Whitfield C. Lipopolysaccharide endotoxins. Annual Review of Biochemistry. 2002;71:635-700
  45. 45. Wiese A, Brandenburg K, Ulmer AJ, Seydel U, Muller-Loennies S. The dual role of lipopolysaccharide as effector and target molecule. Biological Chemistry. 1999;380(7-8):767-784
  46. 46. Sandvig K, van Deurs B. Endocytosis, intracellular transport, and cytotoxic action of Shiga toxin and ricin. Physiological Reviews. 1996;76(4):949-966
  47. 47. Paton JC, Paton AW. Pathogenesis and diagnosis of Shiga toxin-producing Escherichia coli infections. Clinical Microbiology Reviews. 1998;11(3):450-479
  48. 48. Hughes AK, Stricklett PK, Kohan DE. Shiga toxin-1 regulation of cytokine production by human glomerular epithelial cells. Nephron. 2001;88(1):14-23
  49. 49. Wu G. Amino acids: Metabolism, functions, and nutrition. Amino Acids. 2009;37(1):1-17
  50. 50. Wu G, Bazer FW, Davis TA, Jaeger LA, Johnson GA, Kim SW, et al. Important roles for the arginine family of amino acids in swine nutrition and production. Livestock Science. 2007;112(1):8-22
  51. 51. Kim SW, Wu G. Dietary arginine supplementation enhances the growth of milk-fed young pigs. The Journal of Nutrition. 2004;134(3):625-630
  52. 52. Hernandez A, Hansen CF, Mullan BP, Pluske JR. L-arginine supplementation of milk liquid or dry diets fed to pigs after weaning has a positive effect on production in the first three weeks after weaning at 21 days of age. Animal Feed Science and Technology. 2009;154(1):102-111
  53. 53. Wu X, Ruan Z, Gao Y, Yin Y, Zhou X, Wang L, et al. Dietary supplementation with L-arginine or N-carbamylglutamate enhances intestinal growth and heat shock protein-70 expression in weanling pigs fed a corn- and soybean meal-based diet. Amino Acids. 2010;39(3):831-839
  54. 54. Wu L, Liao P, He L, Feng Z, Ren W, Yin J, et al. Dietary L-arginine supplementation protects weanling pigs from deoxynivalenol-induced toxicity. Toxins. 2015;7(4):1341-1354
  55. 55. Wang J, Li GR, Tan BE, Xiong X, Kong XF, Xiao DF, et al. Oral administration of putrescine and proline during the suckling period improves epithelial restitution after early weaning in piglets. Journal of Animal Science. 2015;93(4):1679-1688
  56. 56. Wu G, Meier SA, Knabe DA. Dietary glutamine supplementation prevents jejunal atrophy in weaned pigs. The Journal of Nutrition. 1996;126(10):2578-2584
  57. 57. Li Y, Li J, Jiang J, Li N, Wang X, Wang Z, et al. Glycyl-glutamine-supplemented long-term total parenteral nutrition selectively improves structure and function in heterotopic small-bowel autotransplantation in the pig. Transplant International: Official Journal of the European Society for Organ Transplantation. 2003;16(12):866-871
  58. 58. Jiang ZY, Sun LH, Lin YC, Ma XY, Zheng CT, Zhou GL, et al. Effects of dietary glycyl-glutamine on growth performance, small intestinal integrity, and immune responses of weaning piglets challenged with lipopolysaccharide. Journal of Animal Science. 2009;87(12):4050-4056
  59. 59. Burrin DG, Reeds PJ. Alternative fuels in the gastrointestinal tract. Current Opinion in Gastroenterology. 1997;13(2):165-170
  60. 60. Zhan Z, Ou D, Piao X, Kim SW, Liu Y, Wang J. Dietary arginine supplementation affects microvascular development in the small intestine of early-weaned pigs. The Journal of Nutrition. 2008;138(7):1304-1309
  61. 61. Eklou-Lawson M, Bernard F, Neveux N, Chaumontet C, Bos C, Davila-Gay AM, et al. Colonic luminal ammonia and portal blood L-glutamine and L-arginine concentrations: A possible link between colon mucosa and liver ureagenesis. Amino Acids. 2009;37(4):751-760
  62. 62. Tan B, Xiao H, Xiong X, Wang J, Li G, Yin Y, et al. L-arginine improves DNA synthesis in LPS-challenged enterocytes. Frontiers in Bioscience (Landmark Edition). 2015;20:989-1003
  63. 63. Wang J, Chen L, Li P, Li X, Zhou H, Wang F, et al. Gene expression is altered in piglet small intestine by weaning and dietary glutamine supplementation. The Journal of Nutrition. 2008;138(6):1025-1032
  64. 64. Wu M, Xiao H, Liu G, Chen S, Tan B, Ren W, et al. Glutamine promotes intestinal SIgA secretion through intestinal microbiota and IL-13. Molecular Nutrition & Food Research. 2016;60(7):1637-1648
  65. 65. Ruzin A, Novick RP. Equivalence of lauric acid and glycerol monolaurate as inhibitors of signal transduction in Staphylococcus aureus. Journal of Bacteriology. 2000;182(9):2668-2671
  66. 66. Mallo JJ, Balfagon A, Gracia MI, Honrubia P, Puyalto M. Evaluation of different protections of butyric acid aiming for release in the last part of the gastrointestinal tract of piglets. Journal of Animal Science. 2012;90(Suppl 4):227-229
  67. 67. Kovanda L, Zhang W, Wei X, Luo J, Wu X, Atwill ER, et al. In vitro antimicrobial activities of organic acids and their derivatives on several species of Gram-negative and Gram-positive bacteria. Molecules (Basel, Switzerland). 2019;24(20):3770-3783
  68. 68. Rossi R, Pastorelli G, Cannata S, Corino C. Recent advances in the use of fatty acids as supplements in pig diets: A review. Animal Feed Science and Technology. 2010;162(1):1-11
  69. 69. Bedford A, Gong J. Implications of butyrate and its derivatives for gut health and animal production. Animal Nutrition (Zhongguo Xu Mu Shou Yi Xue Hui). 2018;4(2):151-159
  70. 70. Xiong X, Tan B, Song M, Ji P, Kim K, Yin Y, et al. Nutritional intervention for the intestinal development and health of weaned pigs. Frontiers in Veterinary Science. 2019;6:46
  71. 71. Lu JJ, Zou XT, Wang YM. Effects of sodium butyrate on the growth performance, intestinal microflora and morphology of weanling pigs. Journal of Animal and Feed Sciences. 2008;17(4):568-578
  72. 72. Fang CL, Sun H, Wu J, Niu HH, Feng J. Effects of sodium butyrate on growth performance, haematological and immunological characteristics of weanling piglets. Journal of animal physiology and animal nutrition. 2014;98(4):680-685
  73. 73. Hou Y, Wang L, Yi D, Ding B, Chen X, Wang Q , et al. Dietary supplementation with tributyrin alleviates intestinal injury in piglets challenged with intrarectal administration of acetic acid. The British Journal of Nutrition. 2014;111(10):1748-1758
  74. 74. Huang C, Song P, Fan P, Hou C, Thacker P, Ma X. Dietary sodium butyrate decreases postweaning diarrhea by modulating intestinal permeability and changing the bacterial communities in weaned piglets. The Journal of Nutrition. 2015;145(12):2774-2780
  75. 75. Cuff M, Dyer J, Jones M, Shirazi-Beechey S. The human colonic monocarboxylate transporter isoform 1: Its potential importance to colonic tissue homeostasis. Gastroenterology. 2005;128(3):676-686
  76. 76. Thangaraju M, Cresci GA, Liu K, Ananth S, Gnanaprakasam JP, Browning DD, et al. GPR109A is a G-protein-coupled receptor for the bacterial fermentation product butyrate and functions as a tumor suppressor in colon. Cancer Research. 2009;69(7):2826-2832
  77. 77. Fontenelle B, Gilbert KM. n-butyrate anergized effector CD4+ T cells independent of regulatory T cell generation or activity. Scandinavian Journal of Immunology. 2012;76(5):457-463
  78. 78. Zeng X, Sunkara LT, Jiang W, Bible M, Carter S, Ma X, et al. Induction of porcine host defense peptide gene expression by short-chain fatty acids and their analogs. PLoS One. 2013;8(8):e72922
  79. 79. Odle J. New insights into the utilization of medium-chain triglycerides by the neonate: Observations from a piglet model. The Journal of Nutrition. 1997;127(6):1061-1067
  80. 80. Zentek J, Buchheit-Renko S, Ferrara F, Vahjen W, Van Kessel AG, Pieper R. Nutritional and physiological role of medium-chain triglycerides and medium-chain fatty acids in piglets. Animal Health Research Reviews. 2011;12(1):83-93
  81. 81. Hanczakowska E. The use of medium-chain fatty acids in piglet feeding—A review. Annals of Animal Science. 2017;17(4):967
  82. 82. McDowell LR. Minerals in Animal and Human Nutrition. New York, NY: Academic Press; 1992
  83. 83. Ku PK, Ullery DE, Miller ER. Zinc deficiency and tissue nucleic acid and protein concentration. In: Mills CF, editor. Trace Element Metabolism in Animals. Edinburgh, UK: E. & S. Livingstone; 1970. pp. 158-164
  84. 84. Fernandez-Madrid F, Prasad AS, Oberleas D. Effect of zinc deficiency on collagen metabolism. The Journal of Laboratory and Clinical Medicine. 1971;78(5):853
  85. 85. NRC. Nutrient Requirements of Swine. 11th revised ed. Washington, DC: Natl. Acad. Press; 2012
  86. 86. Hahn JD, Baker DH. Growth and plasma zinc responses of young pigs fed pharmacologic levels of zinc. Journal of Animal Science. 1993;71(11):3020-3024
  87. 87. Hill GM, Mahan DC, Carter SD, Cromwell GL, Ewan RC, Harrold RL, et al. Effect of pharmacological concentrations of zinc oxide with or without the inclusion of an antibacterial agent on nursery pig performance. Journal of Animal Science. 2001;79(4):934-941
  88. 88. Case CL, Carlson MS. Effect of feeding organic and inorganic sources of additional zinc on growth performance and zinc balance in nursery pigs. Journal of Animal Science. 2002;80(7):1917-1924
  89. 89. Starke IC, 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 Microbiology Ecology. 2014;87(2):416-427
  90. 90. Pearce SC, Sanz Fernandez MV, Torrison J, Wilson ME, Baumgard LH, Gabler NK. Dietary organic zinc attenuates heat stress-induced changes in pig intestinal integrity and metabolism. Journal of Animal Science. 2015;93(10):4702-4713
  91. 91. Katouli M, Melin L, Jensen-Waern M, Wallgren P, Mollby R. The effect of zinc oxide supplementation on the stability of the intestinal flora with special reference to composition of coliforms in weaned pigs. Journal of Applied Microbiology. 1999;87(4):564-573
  92. 92. Zhang B, Guo Y. Supplemental zinc reduced intestinal permeability by enhancing occludin and zonula occludens protein-1 (ZO-1) expression in weaning piglets. The British Journal of Nutrition. 2009;102(5):687-693
  93. 93. van Heugten E, Spears JW, Kegley EB, Ward JD, Qureshi MA. Effects of organic forms of zinc on growth performance, tissue zinc distribution, and immune response of weanling pigs. Journal of Animal Science. 2003;81(8):2063-2071
  94. 94. Walk CL, Srinongkote S, Wilcock P. Influence of a microbial phytase and zinc oxide on young pig growth performance and serum minerals. Journal of Animal Science. 2013;91(1):286-291
  95. 95. Walk CL, Wilcock P, Magowan E. Evaluation of the effects of pharmacological zinc oxide and phosphorus source on weaned piglet growth performance, plasma minerals and mineral digestibility. Animal: An International Journal of Animal Bioscience. 2015;9(7):1145-1152
  96. 96. Ciesinski L, Guenther S, Pieper R, Kalisch M, Bednorz C, Wieler LH. High dietary zinc feeding promotes persistence of multi-resistant E. coli in the swine gut. PloS One. 2018;13(1):e0191660
  97. 97. Ward TLAG, Louis GF, Pollman DS. Zinc-methionine improves growth performance of starter pigs. Jornal of Animal Science. 1996;74(Suppl 1):182
  98. 98. Mavromichalis I, Webel DM, Parr EN, Baker DH. Growth-promoting efficacy of pharmacological doses of tetrabasic zinc chloride in diets for nursery pigs. Canadian Journal of Animal Science. 2001;81(3):387-391
  99. 99. Underwood EJ. Trace Elements in Human and Animal Nutrition. 4th ed. New York: Academic Press; 1977
  100. 100. Suttle NF. Mineral Nutrition of Livestock. 4th ed. Oxon, UK: CABI Publishing; 2010
  101. 101. Baxter JH, Van Wyk JJ, Follis RH Jr. A bone disorder associated with copper deficiency. II. Histological and chemical studies on the bones. Bulletin of the Johns Hopkins Hospital. 1953;93(1):25-39
  102. 102. Hart EB, Steenbock H, Waddell J, Elvehjem CA. Iron in nutrition. VII. Copper as a supplement to iron for hemoglobin building in the rat. 1928. The Journal of Biological Chemistry. 2002;277(34):e22
  103. 103. Zhao J, Harper AF, Estienne MJ, Webb KE Jr, McElroy AP, Denbow DM. Growth performance and intestinal morphology responses in early weaned pigs to supplementation of antibiotic-free diets with an organic copper complex and spray-dried plasma protein in sanitary and nonsanitary environments. Journal of Animal Science. 2007;85(5):1302-1310
  104. 104. Perez VG, Waguespack AM, Bidner TD, Southern LL, Fakler TM, Ward TL, et al. Additivity of effects from dietary copper and zinc on growth performance and fecal microbiota of pigs after weaning. Journal of Animal Science. 2011;89(2):414-425
  105. 105. Ma YL, Zanton GI, Zhao J, Wedekind K, Escobar J, Vazquez-Anon M. Multitrial analysis of the effects of copper level and source on performance in nursery pigs. Journal of Animal Science. 2015;93(2):606-614
  106. 106. Kloubert V, Blaabjerg K, Dalgaard TS, Poulsen HD, Rink L, Wessels I. Influence of zinc supplementation on immune parameters in weaned pigs. Journal of Trace Elements in Medicine and Biology: Organ of the Society for Minerals and Trace Elements (GMS). 2018;49:231-240
  107. 107. Stahly TS, Cromwell GL, Monegue HJ. Effects of the dietary inclusion of copper and(or) antibiotics on the performance of weanling pigs. Journal of Animal Science. 1980;51(6):1347-1351
  108. 108. Hojberg O, Canibe N, Poulsen HD, Hedemann MS, Jensen BB. Influence of dietary zinc oxide and copper sulfate on the gastrointestinal ecosystem in newly weaned piglets. Applied and Environmental Microbiology. 2005;71(5):2267-2277
  109. 109. Cromwell GL, Lindemann MD, Monegue HJ, Hall DD, Orr DE Jr. Tribasic copper chloride and copper sulfate as copper sources for weanling pigs. Journal of Animal Science. 1998;76(1):118-123
  110. 110. Banks KM, Thompson KL, Rush JK, Applegate TJ. Effects of copper source on phosphorus retention in broiler chicks and laying hens. Poultry Science. 2004;83(6):990-996
  111. 111. Pang Y, Applegate TJ. Effects of copper source and concentration on in vitro phytate phosphorus hydrolysis by phytase. Journal of Agricultural and Food Chemistry. 2006;54(5):1792-1796
  112. 112. Armstrong TA, Cook DR, Ward MM, Williams CM, Spears JW. Effect of dietary copper source (cupric citrate and cupric sulfate) and concentration on growth performance and fecal copper excretion in weanling pigs. Journal of Animal Science. 2004;82(4):1234-1240
  113. 113. Gibson GR, Probert HM, Loo JV, Rastall RA, Roberfroid MB. Dietary modulation of the human colonic microbiota: Updating the concept of prebiotics. Nutrition Research Reviews. 2004;17(2):259-275
  114. 114. Macfarlane S, Macfarlane GT, Cummings JH. Review article: Prebiotics in the gastrointestinal tract. Alimentary Pharmacology & Therapeutics. 2006;24(5):701-714
  115. 115. Kelly G. Inulin-type prebiotics—A review: Part 1. Alternative Medicine Review: A Journal of Clinical Therapeutic. 2008;13(4):315-329
  116. 116. Boehm G, Jelinek J, Stahl B, van Laere K, Knol J, Fanaro S, et al. Prebiotics in infant formulas. Journal of Clinical Gastroenterology. 2004;38(6 Suppl):S76-S79
  117. 117. Zeković DB, Kwiatkowski S, Vrvić MM, Jakovljević D, Moran CA. Natural and modified (1→3)-β-D-glucans in health promotion and disease alleviation. Critical Reviews in Biotechnology. 2005;25(4):205-230
  118. 118. Samuelsen AB, Schrezenmeir J, Knutsen SH. Effects of orally administered yeast-derived beta-glucans: A review. Molecular Nutrition & Food Research. 2014;58(1):183-193
  119. 119. Soltanian S, Stuyven E, Cox E, Sorgeloos P, Bossier P. Beta-glucans as immunostimulant in vertebrates and invertebrates. Critical Reviews in Microbiology. 2009;35(2):109-138
  120. 120. Kim K, Ehrlich A, Perng V, Chase JA, Raybould H, Li X, et al. Algae-derived β-glucan enhanced gut health and immune responses of weaned pigs experimentally infected with a pathogenic E. coli. Animal Feed Science and Technology. 2019;248:114-125
  121. 121. World Heath Organization, Food and Agriculture Organization of the United Nations, United Nations University. Protein and amino acid requirements in human nutrition. 2007. Report of a joint FAO/WHO/UNU expert consultation (WHO Technical Report Series 935)
  122. 122. Stein HH, Kil DY. Reduced use of antibiotic growth promoters in diets fed to weanling pigs: Dietary tools, part 2. Animal Biotechnology. 2006;17(2):217-231
  123. 123. Ferrari E, Jarnagin A, Schmidt BF. Commercial production of extracellular enzymes. In: Sonenshein AL, Hoch JA, Losick R, editors. Bacillus subtilis and Other Gram-Positive Bacteria. Washington, DC: American Society for Microbiology; 1993. pp. 917-937
  124. 124. 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. Livestock Science. 2010;134(1):124-134
  125. 125. Richards JD, Gong J, de Lange CFM. The gastrointestinal microbiota and its role in monogastric nutrition and health with an emphasis on pigs: Current understanding, possible modulations, and new technologies for ecological studies. Canadian Journal of Animal Science. 2005;85(4):421-435
  126. 126. Vandenbergh PA. Lactic acid bacteria, their metabolic products and interference with microbial growth. FEMS Microbiology Reviews. 1993;12(1):221-237
  127. 127. Niers LE, Timmerman HM, Rijkers GT, van Bleek GM, van Uden NO, Knol EF, et al. Identification of strong interleukin-10 inducing lactic acid bacteria which down-regulate T helper type 2 cytokines. Clinical and Experimental Allergy: Journal of the British Society for Allergy and Clinical Immunology. 2005;35(11):1481-1489
  128. 128. Kogan G, Kocher A. Role of yeast cell wall polysaccharides in pig nutrition and health protection. Livestock Science. 2007;109(1):161-165
  129. 129. Sauer N, Eklund M, Roth S, Rink F, Jezierny D, Bauer E, et al. Short-term effect of dietary yeast nucleotide supplementation on small intestinal enzyme activities, bacterial populations and metabolites and ileal nutrient digestibilities in newly weaned pigs. Journal of Animal Physiology and Animal Nutrition. 2012;96(4):700-708
  130. 130. Shen YB, Piao XS, Kim SW, Wang L, Liu P, Yoon I, et al. Effects of yeast culture supplementation on growth performance, intestinal health, and immune response of nursery pigs. Journal of Animal Science. 2009;87(8):2614-2624
  131. 131. Jiang Z, Wei S, Wang Z, Zhu C, Hu S, Zheng C, et al. Effects of different forms of yeast Saccharomyces cerevisiae on growth performance, intestinal development, and systemic immunity in early-weaned piglets. Journal of Animal Science and Biotechnology. 2015;6:47
  132. 132. Toure R, Kheadr E, Lacroix C, Moroni O, Fliss I. Production of antibacterial substances by bifidobacterial isolates from infant stool active against listeria monocytogenes. Journal of Applied Microbiology. 2003;95(5):1058-1069
  133. 133. Gibson GR, Wang X. Regulatory effects of bifidobacteria on the growth of other colonic bacteria. The Journal of Applied Bacteriology. 1994;77(4):412-420
  134. 134. Corr SC, Li Y, Riedel CU, O’Toole PW, Hill C, Gahan CG. Bacteriocin production as a mechanism for the antiinfective activity of Lactobacillus salivarius UCC118. Proceedings of the National Academy of Sciences of the United States of America. 2007;104(18):7617-7621
  135. 135. Gibson GR, McCartney AL, Rastall RA. Prebiotics and resistance to gastrointestinal infections. The British Journal of Nutrition. 2005;93(Suppl 1):S31-S34
  136. 136. Halas D, Hansen CF, Hampson DJ, Mullan BP, Wilson RH, Pluske JR. Effect of dietary supplementation with inulin and/or benzoic acid on the incidence and severity of post-weaning diarrhoea in weaner pigs after experimental challenge with enterotoxigenic Escherichia coli. Archives of Animal Nutrition. 2009;63(4):267-280
  137. 137. Bunce TJ, Howard MD, Kerley MS, Alee GL, Pace LW. Protective effect of fructooligosaccharide (FOS) in prevention of mortality and morbidity from infectious E. coli K:88 challenge. Journal of Animal Science. 1995;63(Suppl 1):69
  138. 138. Stuyven E, Cox E, Vancaeneghem S, Arnouts S, Deprez P, Goddeeris BM. Effect of beta-glucans on an ETEC infection in piglets. Veterinary Immunology and Immunopathology. 2009;128(1-3):60-66
  139. 139. Kiarie E, Bhandari S, Scott M, Krause DO, Nyachoti CM. Growth performance and gastrointestinal microbial ecology responses of piglets receiving Saccharomyces cerevisiae fermentation products after an oral challenge with Escherichia coli (K88). Journal of Animal Science. 2011;89(4):1062-1078
  140. 140. Kiarie E, Scott M, Krause DO, Khazanehei H, Khafipour E, Nyachoti CM. Interactions of Saccharomyces cerevisiae fermentation product and in-feed antibiotic on gastrointestinal and immunological responses in piglets challenged with Escherichia coli K88+. Journal of Animal Science. 2012;90(Suppl 4):1-3
  141. 141. Trckova M, Faldyna M, Alexa P, Sramkova Zajacova Z, Gopfert E, Kumprechtova D, et al. The effects of live yeast Saccharomyces cerevisiae on postweaning diarrhea, immune response, and growth performance in weaned piglets. Journal of Animal Science. 2014;92(2):767-774
  142. 142. Kim K, He Y, Xiong X, Ehrlich A, Li X, Raybould H, et al. Dietary supplementation of Bacillus subtilis influenced intestinal health of weaned pigs experimentally infected with a pathogenic E. coli. Journal of Animal Science and Biotechnology. 2019;10(1):52
  143. 143. Gadde U, Kim WH, Oh ST, Lillehoj HS. Alternatives to antibiotics for maximizing growth performance and feed efficiency in poultry: A review. Animal Health Research Reviews. 2017;18(1):26-45
  144. 144. Windisch W, Schedle K, Plitzner C, Kroismayr A. Use of phytogenic products as feed additives for swine and poultry. Journal of Animal Science. 2008;86(14 Suppl):E140-E148
  145. 145. Baydar NG, Özkan G, Sağdiç O. Total phenolic contents and antibacterial activities of grape (Vitis vinifera L.) extracts. Food Control. 2004;15(5):335-339
  146. 146. Sokmen M, Serkedjieva J, Daferera D, Gulluce M, Polissiou M, Tepe B, et al. In vitro antioxidant, antimicrobial, and antiviral activities of the essential oil and various extracts from herbal parts and callus cultures of Origanum acutidens. Journal of Agricultural and Food Chemistry. 2004;52(11):3309-3312
  147. 147. Dundar E, Olgun EG, Isiksoy S, Kurkcuoglu M, Baser KH, Bal C. The effects of intra-rectal and intra-peritoneal application of Origanum onites L. essential oil on 2,4,6-trinitrobenzenesulfonic acid-induced colitis in the rat. Experimental and Toxicologic Pathology: Official Journal of the Gesellschaft fur Toxikologische Pathologie. 2008;59(6):399-408
  148. 148. Liu Y, Song M, Che TM, Bravo D, Pettigrew JE. Anti-inflammatory effects of several plant extracts on porcine alveolar macrophages in vitro. Journal of Animal Science. 2012;90(8):2774-2783
  149. 149. Hammer KA, Carson CF, Riley TV. Antimicrobial activity of essential oils and other plant extracts. Journal of Applied Microbiology. 1999;86(6):985-990
  150. 150. Dorman HJ, Deans SG. Antimicrobial agents from plants: Antibacterial activity of plant volatile oils. Journal of Applied Microbiology. 2000;88(2):308-316
  151. 151. Wong SY, Grant IR, Friedman M, Elliott CT, Situ C. Antibacterial activities of naturally occurring compounds against Mycobacterium avium subsp. paratuberculosis. Applied and Environmental Microbiology. 2008;74(19):5986-5990
  152. 152. Carson CF, Mee BJ, Riley TV. Mechanism of action of Melaleuca alternifolia (tea tree) oil on Staphylococcus aureus determined by time-kill, lysis, leakage, and salt tolerance assays and electron microscopy. Antimicrobial Agents and Chemotherapy. 2002;46(6):1914-1920
  153. 153. Burt S. Essential oils: Their antibacterial properties and potential applications in foods—A review. International Journal of Food Microbiology. 2004;94(3):223-253
  154. 154. Xu J, Zhou F, Ji BP, Pei RS, Xu N. The antibacterial mechanism of carvacrol and thymol against Escherichia coli. Letters in Applied Microbiology. 2008;47(3):174-179
  155. 155. Trombetta D, Castelli F, Sarpietro MG, Venuti V, Cristani M, Daniele C, et al. Mechanisms of Antibacterial Action of Three Monoterpenes. 2005;49(6):2474-2478
  156. 156. Sikkema J, de Bont JA, Poolman B. Mechanisms of membrane toxicity of hydrocarbons. Microbiological Reviews. 1995;59(2):201-222
  157. 157. Farag RS, Daw ZY, Hewedi FM, El-Baroty GSA. Antimicrobial activity of some Egyptian spice essential oils. Journal of Food Protection. 1989;52(9):665-667
  158. 158. Lambert RJ, Skandamis PN, Coote PJ, Nychas GJ. A study of the minimum inhibitory concentration and mode of action of oregano essential oil, thymol and carvacrol. Journal of Applied Microbiology. 2001;91(3):453-462
  159. 159. Ankri S, Mirelman D. Antimicrobial properties of allicin from garlic. Microbes and Infection. 1999;1(2):125-129
  160. 160. Burt SA, van der Zee R, Koets AP, de Graaff AM, van Knapen F, Gaastra W, et al. Carvacrol induces heat shock protein 60 and inhibits synthesis of flagellin in Escherichia coli O157:H7. Applied and Environmental Microbiology. 2007;73(14):4484-4490
  161. 161. Verhelst R, Schroyen M, Buys N, Niewold T. The effects of plant polyphenols on enterotoxigenic Escherichia coli adhesion and toxin binding. Livestock Science. 2010;133(1):101-103
  162. 162. Liu Y, Song M, Che TM, Almeida JA, Lee JJ, Bravo D, et al. Dietary plant extracts alleviate diarrhea and alter immune responses of weaned pigs experimentally infected with a pathogenic Escherichia coli. Journal of Animal Science. 2013;91(11):5294-5306
  163. 163. Liu Y, Song M, Che TM, Lee JJ, Bravo D, Maddox CW, et al. Dietary plant extracts modulate gene expression profiles in ileal mucosa of weaned pigs after an Escherichia coli infection. Journal of Animal Science. 2014;92(5):2050-2062
  164. 164. Bruins MJ, Cermak R, Kiers JL, van der Meulen J, van Amelsvoort JM, van Klinken BJ. In vivo and in vitro effects of tea extracts on enterotoxigenic Escherichia coli-induced intestinal fluid loss in animal models. Journal of Pediatric Gastroenterology and Nutrition. 2006;43(4):459-469
  165. 165. Coddens A, Loos M, Vanrompay D, Remon JP, Cox E. Cranberry extract inhibits in vitro adhesion of F4 and F18(+)Escherichia coli to pig intestinal epithelium and reduces in vivo excretion of pigs orally challenged with F18(+) verotoxigenic E. coli. Veterinary Microbiology. 2017;202:64-71
  166. 166. Zasloff M. Antimicrobial peptides of multicellular organisms. Nature. 2002;415(6870):389-395
  167. 167. Xiao H, Shao F, Wu M, Ren W, Xiong X, Tan B, et al. The application of antimicrobial peptides as growth and health promoters for swine. Journal of Animal Science and Biotechnology. 2015;6(1):19
  168. 168. Wang S, Zeng X, Yang Q , Qiao S. Antimicrobial peptides as potential alternatives to antibiotics in food animal industry. International Journal of Molecular Sciences. 2016;17(5):603-614
  169. 169. Selsted ME, Ouellette AJ. Mammalian defensins in the antimicrobial immune response. Nature Immunology. 2005;6(6):551-557
  170. 170. Hancock RE. Cationic peptides: Effectors in innate immunity and novel antimicrobials. The Lancet Infectious Diseases. 2001;1(3):156-164
  171. 171. Zhang L, Parente J, Harris SM, Woods DE, Hancock RE, Falla TJ. Antimicrobial peptide therapeutics for cystic fibrosis. Antimicrobial Agents and Chemotherapy. 2005;49(7):2921-2927
  172. 172. Shai Y. Mechanism of the binding, insertion and destabilization of phospholipid bilayer membranes by alpha-helical antimicrobial and cell non-selective membrane-lytic peptides. Biochimica et Biophysica Acta. 1999;1462(1-2):55-70
  173. 173. Yang L, Harroun TA, Weiss TM, Ding L, Huang HW. Barrel-stave model or toroidal model? A case study on melittin pores. Biophysical Journal. 2001;81(3):1475-1485
  174. 174. Brogden KA. Antimicrobial peptides: Pore formers or metabolic inhibitors in bacteria? Nature Reviews Microbiology. 2005;3(3):238-250
  175. 175. Mansour SC, Pena OM, Hancock RE. Host defense peptides: front-line immunomodulators. Trends in Immunology. 2014;35(9):443-450
  176. 176. Ren ZH, Yuan W, Deng HD, Deng JL, Dan QX, Jin HT, et al. Effects of antibacterial peptide on cellular immunity in weaned piglets. Journal of Animal Science. 2015;93(1):127-134
  177. 177. Wu S, Zhang F, Huang Z, Liu H, Xie C, Zhang J, et al. Effects of the antimicrobial peptide cecropin AD on performance and intestinal health in weaned piglets challenged with Escherichia coli. Peptides. 2012;35(2):225-230
  178. 178. Xiao H, Tan BE, Wu MM, Yin YL, Li TJ, Yuan DX, et al. Effects of composite antimicrobial peptides in weanling piglets challenged with deoxynivalenol: II. Intestinal morphology and function. Journal of Animal Science. 2013;91(10):4750-4756
  179. 179. 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. Animal Feed Science and Technology. 2007;135(3):263-272
  180. 180. Tang Z, Yin Y, Zhang Y, Huang R, Sun Z, Li T, et al. 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. The British Journal of Nutrition. 2009;101(7):998-1005
  181. 181. Lonnerdal B. Biochemistry and physiological function of human milk proteins. The American journal of clinical nutrition. 1985;42(6):1299-1317
  182. 182. Wiesner J, Vilcinskas A. Antimicrobial peptides: The ancient arm of the human immune system. Virulence. 2010;1(5):440-464
  183. 183. Masschalck B, Michiels CW. Antimicrobial properties of lysozyme in relation to foodborne vegetative bacteria. Critical Reviews in Microbiology. 2003;29(3):191-214
  184. 184. Ibrahim HR, Thomas U, Pellegrini A. A helix-loop-helix peptide at the upper lip of the active site cleft of lysozyme confers potent antimicrobial activity with membrane permeabilization action. The Journal of Biological Chemistry. 2001;276(47):43767-43774
  185. 185. Laible NJ, Germaine GR. Bactericidal activity of human lysozyme, muramidase-inactive lysozyme, and cationic polypeptides against Streptococcus sanguis and Streptococcus faecalis: Inhibition by chitin oligosaccharides. Infection and Immunity. 1985;48(3):720-728
  186. 186. Ibrahim HR, Matsuzaki T, Aoki T. Genetic evidence that antibacterial activity of lysozyme is independent of its catalytic function. FEBS letters. 2001;506(1):27-32
  187. 187. Ellison RT, Giehl TJ. Killing of gram-negative bacteria by lactoferrin and lysozyme. Journal of Clinical Investigation. 1991;88(4):1080-1091
  188. 188. Gordon LI, Douglas SD, Kay NE, Yamada O, Osserman EF, Jacob HS. Modulation of neutrophil function by lysozyme. potential negative feedback system of inflammation. Journal of Clinical Investigation. 1979;64(1):226-232
  189. 189. Maga EA, Desai PT, Weimer BC, Dao N, Kültz D, Murray JD. Consumption of lysozyme-rich milk can alter microbial fecal populations. Applied and Environmental Microbiology. 2012;78(17):6153-6160
  190. 190. Garas LC, Cooper CA, Dawson MW, Wang JL, Murray JD, Maga EA. Young pigs consuming lysozyme transgenic goat milk are protected from clinical symptoms of enterotoxigenic Escherichia coli infection. The Journal of Nutrition. 2017;147(11):2050-2059
  191. 191. Huang G, Li X, Lu D, Liu S, Suo X, Li Q , et al. Lysozyme improves gut performance and protects against enterotoxigenic Escherichia coli infection in neonatal piglets. Veterinary Research. 2018;49(1):20
  192. 192. Liu Y, Ji P. Dietary factors in prevention of pediatric Escherichia coli infection: A model using domestic piglets. ILAR Journal. 2018;59(3):338-351
  193. 193. Brundige DR, Maga EA, Klasing KC, Murray JD. Lysozyme transgenic goats’ milk influences gastrointestinal morphology in young pigs. The Journal of Nutrition. 2008;138(5):921-926
  194. 194. Cooper CA, Garas Klobas LC, Maga EA, Murray JD. Consuming transgenic goats’ milk containing the antimicrobial protein lysozyme helps resolve diarrhea in young pigs. PLoS One. 2013;8(3):e58409
  195. 195. Liu P, Piao XS, Thacker PA, Zeng ZK, Li PF, Wang D, et al. Chito-oligosaccharide reduces diarrhea incidence and attenuates the immune response of weaned pigs challenged with Escherichia coli K88. Journal of Animal Science. 2010;88(12):3871-3879
  196. 196. Krause DO, Bhandari SK, House JD, Nyachoti CM. Response of nursery pigs to a synbiotic preparation of starch and an anti-Escherichia coli K88 probiotic. Applied and Environmental Microbiology. 2010;76(24):8192-8200
  197. 197. Daudelin JF, Lessard M, Beaudoin F, Nadeau E, Bissonnette N, Boutin Y, et al. Administration of probiotics influences F4 (K88)-positive enterotoxigenic Escherichia coli attachment and intestinal cytokine expression in weaned pigs. Veterinary Research. 2011;42:69
  198. 198. Lee JS, Awji EG, Lee SJ, Tassew DD, Park YB, Park KS, et al. Effect of lactobacillus plantarum CJLP243 on the growth performance and cytokine response of weaning pigs challenged with enterotoxigenic Escherichia coli. Journal of Animal Science. 2012;90(11):3709-3717
  199. 199. Yang KM, Jiang ZY, Zheng CT, Wang L, Yang XF. Effect of lactobacillus plantarum on diarrhea and intestinal barrier function of young piglets challenged with enterotoxigenic Escherichia coli K88. Journal of Animal Science. 2014;92(4):1496-1503
  200. 200. Devi SM, Lee SI, Kim IH. Effect of phytogenics on growth performance, fecal score, blood profiles, fecal noxious gas emission, digestibility, and intestinal morphology of weanling pigs challenged with Escherichia coli K88. Polish Journal of Veterinary Sciences. 2015;18(3):557-564
  201. 201. Li H, Zhao P, Lei Y, Li T, Kim I. Response to an Escherichia coli K88 oral challenge and productivity of weanling pigs receiving a dietary nucleotides supplement. Journal of Animal Science and Biotechnology. 2015;6:49
  202. 202. Song M, Liu Y, Soares JA, Che TM, Osuna O, Maddox CW, et al. Dietary clays alleviate diarrhea of weaned pigs. Journal of Animal Science. 2012;90(1):345-360
  203. 203. Almeida JA, Liu Y, Song M, Lee JJ, Gaskins HR, Maddox CW, et al. Escherichia coli challenge and one type of smectite alter intestinal barrier of pigs. Journal of Animal Science and Biotechnology. 2013;4(1):52

Written By

Peng Ji, Xunde Li and Yanhong Liu

Submitted: 04 October 2019 Reviewed: 16 January 2020 Published: 10 February 2020