Hostname: page-component-8448b6f56d-cfpbc Total loading time: 0 Render date: 2024-04-23T07:37:41.804Z Has data issue: false hasContentIssue false

The role of dietary nucleotides in single-stomached animals

Published online by Cambridge University Press:  13 January 2011

Nadja Sauer
Affiliation:
Institute of Animal Nutrition, University of Hohenheim, 70593Stuttgart, Germany
Rainer Mosenthin*
Affiliation:
Institute of Animal Nutrition, University of Hohenheim, 70593Stuttgart, Germany
Eva Bauer
Affiliation:
Institute of Animal Nutrition, University of Hohenheim, 70593Stuttgart, Germany
*
*Corresponding author: Professor Rainer Mosenthin, fax +49 711 459 22421, email rainer.mosenthin@uni-hohenheim.de
Rights & Permissions [Opens in a new window]

Abstract

The transition from liquid to solid feed during weaning results in morphological, histological and microbial changes in the young animal's intestinal tract and often is associated with diarrhoea. The ban of in-feed antibiotics in pig production in the European Union has led to increasing interest in alternatives to overcome weaning-associated problems. Among others, nucleotides may have the potential to alleviate health impairments due to weaning. Nucleotides are natural components of the non-protein fraction of milk and have important effects on the maintenance of health in young animals. Nucleotides and their related metabolic products play key roles in many biological processes and become essential dietary components when endogenous supply is insufficient for normal function. The present review summarises nucleotide composition of milk from different species, the biology of nucleotides and possible effects of dietary nucleotides on intestinal morphology and function, intestinal microbiota, immune function, nutrient metabolism, hepatic morphology and function as well as growth performance. Special attention is given to data available for pigs, and suggestions are made for inclusion of nucleotides in the diet to benefit piglets' health and reduce the consequences accompanying early weaning.

Type
Review Article
Copyright
Copyright © The Authors 2011

Introduction

Until 2006, in-feed antibiotics were used to overcome weaning-associated problems in pig production. Nowadays, an interest in antibiotic-free animal production has been established worldwide due to the risk of antibiotic resistance in humans(Reference Bager, Aarestrup and Wegener1). As weaning is accompanied by morphological, histological and microbial changes in the gastrointestinal tract of young mammals(Reference Cummins, Steele and LaBrooy2, Reference Pluske, Hampson and Williams3), nutritional strategies are being developed to aid immature animals to overcome weaning-associated problems and withstand infectious diseases. Among others, dietary nucleotides are one group of bioactive agents which may have the potential to diminish challenges related to weaning(Reference Andrés-Elias, Pujols and Badiola4). Nucleotides are naturally present in all foods of animal and vegetable origin(Reference Clifford and Story5) but in lower concentrations than in mammalian milk(Reference Mateo, Peters and Stein6). Soluble nucleotides have been determined in milk from various mammals, contributing to as much as 20 % of its non-protein fraction(Reference Uauy and Lebenthal7). The nucleotide pattern in milk is species-specific, and concentrations vary with stage of lactation. Nucleotides and their metabolic products play key roles in many biological processes, for example, as monomeric units of nucleic acids, in transferring chemical energy, in biosynthetic pathways, as biological regulators and as coenzyme components(Reference Cosgrove8). Although nucleotides are synthesised endogenously, there is also evidence that dietary supplementation with nucleotides may exert beneficial effects both in humans and animals (for reviews, see Carver & Walker(Reference Carver and Walker9) and Taylor-Pickard(Reference Taylor-Pickard10)). In animals, including pigs and rodents, positive effects on small-intestinal growth and development(Reference Domeneghini, Di Giancamillo and Savoini11), on intestinal hyperaemia(Reference Bustamante12), on stimulation of systemic immunity(Reference Nagafuchi, Katayanagi and Nakagawa13) and on hepatic composition(Reference Novak, Carver and Barness14) have been described. In addition, a protective role in the prevention of diarrhoea in pigs has been reported by Martinez-Puig et al. (Reference Martinez-Puig, Manzanilla and Morales15). Accordingly, Gil et al. (Reference Gil, Corral and Martínez16) described in human subjects beneficial effects of supplemental soluble nucleotides on the composition of the faecal microbiota, as measured by an increase in bifidobacteria and lactobacilli. Inconsistent results, however, have been found for small-intestinal disaccharidase activity as influenced by nucleotide supplementation(Reference Uauy, Stringel and Thomas17, Reference Carver18). However, there are few data on the animals' requirement for exogenous nucleotides, and most of the studies have investigated effects of dietary nucleotides on the immune function in rodents, whereas only limited information exists on the possible role of nucleotides in promoting intestinal health or in stimulating the development of the immune system in other species. The present review summarises data published on nucleotide composition of various food and feed ingredients with special emphasis on milk originating from different species. Furthermore, possible effects of dietary nucleotides on intestinal morphology and function, intestinal microflora, immune function as well as on growth performance are described with special focus on pigs, but, where applicable, data from human subjects and rodents are also included. Special attention is given to data describing the potential role of dietary nucleotides to improve piglets' health and development.

Biochemical and functional characteristics of nucleotides

Nucleotides are low-molecular-weight intracellular compounds that participate in numerous biochemical processes. They are composed of a nitrogenous base (pyrimidine or purine) linked to a pentose (ribose or deoxyribose) sugar ( = nucleoside) to which one, two or three phosphate groups are attached. The pyrimidines comprise cytosine (C), uridine (U) or thymine (T), as well as orotic acid, which is present in the milk of ruminants(Reference Larson and Hegarty19), and which is produced as an intermediate product during the synthesis of uridine 5′-monophosphate (UMP). The purines comprise adenine (A), guanine (G) and hypoxanthine (I). The prefix d is added when the sugar of the nucleotide consists of deoxyribose. The capital letters MP, DP and TP indicate mono-, di- and tri-phosphate esters, respectively(Reference Cosgrove8). The purine and pyrimidine bases are hydrophobic and relatively insoluble in water at the near-neutral pH of the cell. At acid or alkaline pH, the bases become charged and their solubility in water increases(Reference Nelson, Cox, Nelson and Cox20). Nucleotides and nucleic acids are constantly being formed and degraded in all tissues, especially in tissues with a rapid turnover rate such as cells in the immune system, intestinal mucosa, skin, and progenitors of leucocytes or erythrocytes(Reference Uauy and Lebenthal7). For example, according to an in vitro study on intestinal explants from suckling piglets, the jejunum of piglets at weaning is able to hydrolyse RNA and free nucleotides to nucleosides, which, except for cytidine, are then efficiently taken up by the small intestine(Reference Gil, Gomez-Leon and Rueda21). Generally, there are three potential sources of nucleotides: de novo synthesis, salvage pathways and the diet(Reference Cosgrove8, Reference Boza22). The de novo synthesis of nucleotides is a metabolically costly process requiring substantial amounts of energy in the form of ATP(Reference Carver and Walker9). Within the de novo pathways in mammalian cells, the pyrimidine ring can be synthesised from aspartate, glutamine and CO2 whereas the purine ring can be formed from glycine, aspartate, glutamine, tetrahydrofolate derivates and CO2(Reference Chu23). Another mechanism for maintaining the cellular nucleotide pools is the salvage pathway. Under fed conditions, this pathway recycles 90 % or more of the purine bases. Thus, it is suggested that the salvage pathway is dependent on the availability of free purine and pyrimidine bases(Reference Uauy and Lebenthal7). This pathway requires less energy than the reactions needed for the de novo synthesis of nucleotides and is characterised by linkage of a ribose phosphate moiety to free bases formed by hydrolytic degradation of nucleic acid and nucleotides(Reference Cosgrove8). Also, some tissues have limited capacity for the de novo synthesis of nucleotides, thus requiring exogenously supplied bases that can be utilised by the salvage pathway(Reference Savaiano and Clifford24Reference Gil, Uauy and Jensen26). For example, the intestinal mucosa, the haematopoietic cells of the bone marrow, leucocytes, erythrocytes and lymphocytes are incapable of de novo synthesis(Reference Sanderson and He27), and thus utilise the salvage pathway, suggesting that an exogenous supply of nucleotides via the diet might be important for these cells(Reference Cosgrove8).

The absorption, transport and use of exogenous nucleotides and nucleosides are complex processes, which have mainly been investigated in vitro by using mucosal exposures of the ileum and ex vivo by everted gut sacs of rabbits and rats(Reference Harms and Stirling28, Reference Bronk and Hastewell29) or in cultured cell lines(Reference Plagemann, Marz and Wohlhueter30, Reference Marz, Wohlhueter and Plagemann31). Under physiological conditions, nucleotides have a limited capacity to pass through cell membranes(Reference Sanderson and He27). This may be due to the absence of a nucleotide transport system. Nucleotides also have a high negatively charged phosphate group that prevents them from being absorbed(Reference Sanderson and He27). Except for small amounts, dietary nucleotides need to be enzymically hydrolysed before absorption, because only nucleosides and bases can be absorbed in the small intestine(Reference Quan and Uauy32). Endonucleases, phosphodiesterases, and nucleoside phosphorylases are the major enzymes involved in this transport process (Fig. 1). They originate from the brush-border epithelium(Reference Markiewicz, Kaminski and Chocilowski33, Reference Morley, Hawley and Ulbright34), pancreas(Reference Weickmann, Elson and Glitz35) and bile(Reference Holdsworth and Coleman36). The duodenum has the most developed absorptive capacity for nucleosides(Reference Bronk and Hastewell37). Differences in the efficiency of uptake among nucleosides have been reported, with guanosine being taken up most rapidly(Reference Sanderson and He27). More than 90 % of dietary and endogenous nucleosides and bases are transported into the enterocytes(Reference Uauy and Lebenthal7, Reference Salati, Gross and Henderson38). Nucleoside transport into the enterocyte occurs by facilitated diffusion and by specific Na+-dependent carrier-mediated mechanisms(Reference Bronk and Hastewell37). Several enterocyte degradation products of dietary and endogenous nucleotides and nucleosides, mainly nitrogenous bases, such as adenine, cytosine, guanine, hypoxanthine, uracil or thymine, enter the hepatic portal vein. These molecules are carried to the hepatocytes for further metabolism; thereafter, they are released from the liver into systemic circulation and enter muscle tissue. If these products are not re-utilised for nucleotide production or remain unabsorbed, the purine bases are catabolised into uric acid, whereas the pyrimidine bases are catabolised into β-alanine or β-aminoisobutyrate(Reference Chu23). In mammals, except for primates, the catabolic product of purine, uric acid, is further catabolised into allantoin via the enzyme uricase to be excreted into the urine, whereas in avian species and primates, uric acid is excreted via the urine. The catabolic products of pyrimidine bases (β-alanine or β-aminoisobutyrate) are further metabolised into ammonia, carbon dioxide and acetyl coenzyme A.

Fig. 1 Digestion and absorption of nucleoproteins and their related products (adapted from Quan & Uauy(Reference Quan and Uauy32)).

Nucleotides play a key role in many biochemical processes, predominantly in cellular functions. They are precursors of nucleic acids, namely RNA and DNA, as they provide their monomeric units. The concentration of ribonucleotides is relatively constant in all cells, whereas the concentration of deoxyribonucleotides varies during the cell cycle(Reference Barness39). Furthermore, nucleotides may influence the cell cycle, for example, their withdrawal leads to the arrest of T-cells in the G-phase, preventing a response to various immunological signals that occur by transition to the S-phase(Reference Kulkarni, Rudolph and van Buren40). Additionally, Paubert-Braquet et al. (Reference Paubert-Braquet, Dupont, Hedef, Paubert-Braquet, Dupont and Paoletti41) reported a decrease in phagocygotic activity, lymphokine production and/or inhibited lymphocyte maturation due to nucleotide removal. Furthermore, nucleotides, mainly the adenine nucleotides (ATP, ADP, AMP), are the most important molecules for the transfer of chemical energy from energy-yielding reactions to energy-requiring reactions(Reference Uauy and Lebenthal7, Reference Cosgrove8). Special forms of nucleotides activate as co-enzymes such as NAD, NADP, FAD and CoA, and play key roles as activated intermediates in the synthesis of lipids, carbohydrates and protein(Reference Uauy and Lebenthal7). Nucleotides participate in biological regulation, particularly as cyclic AMP which is a second messenger used for intracellular signal transduction, such as transferring the action of hormones including glucagon and adrenaline, which cannot pass the cell membrane(Reference Zivkovic42). Further properties of cyclic AMP include the activation of protein kinase and regulating the effects of adrenaline and glucagon(Reference Poggioli, Mauger and Claret43). It also serves as a regulator of the passage of CaReference Cummins, Steele and LaBrooy2+ through ion channels(Reference Zivkovic42).

Besides their role in biochemical processes, nucleotides are eventually involved in biological reactions as well. Several in vivo studies suggest that dietary nucleotides may promote the development of the gastrointestinal ultrastructure(Reference Uauy, Stringel and Thomas17), modulate the intestinal microbiota(Reference Gil, Corral and Martínez16) and have influences on immune function(Reference Lee, Liu and Chen44). Although nucleotides are synthesised endogenously, it has been suggested that dietary supplementation with nucleotides may exert beneficial effects on small-intestinal growth and development, lipid metabolism and hepatic function (for a review, see Carver & Walker(Reference Carver and Walker9)). Conditions under which nucleotides may become essential include certain disease states, periods of limited nutrient intake or rapid growth, and the presence of regulatory or developmental factors which may interfere with full expression of the endogenous synthetic capacity(Reference Uauy and Lebenthal7). Under such conditions, dietary nucleotides may spare the cost of de novo synthesis and/or the salvage pathway, and thus may help to optimise the function of tissues requiring high concentrations of nucleotides (for example, intestinal mucosa). Furthermore, there is evidence that some cells, for example, erythrocytes, possess no or limited capacity for the de novo synthesis of nucleotides(Reference Carver and Walker9). Additionally, the intestinal mucosa, as well as the haematopoietic cells of the bone marrow, for example, leucocyte proliferation, preferentially utilise preformed pyrimidine and purine bases(Reference Gil, Uauy and Jensen26). These cells require exogenously supplied nucleotides that can be utilised via the salvage pathway rather than forming bases by de novo synthesis. For these cells, an exogenous supply of nucleotides may be essential for optimal function. There is sufficient de novo synthesis of purines and pyrimidines by the liver to meet its own requirements and the liver delivers bases to other tissues, for example, the intestine with high demands due to rapid turnover rates(Reference Gil, Uauy and Jensen26). The allocation of purines and pyrimidines via de novo synthesis in the liver is linked to high energy expenditure. In the absence of dietary nucleotides, the de novo pathway for purine and pyrimidine synthesis is activated(Reference Gil, Uauy and Jensen26). However, supplementing nucleotides may suppress the metabolic costly de novo synthesis, thereby activating the salvage pathway(Reference Savaiano and Clifford24, Reference LeLeiko, Bronstein and Baliga25, Reference Rudolph, Kulkarni and Schandel45). Thus, the term ‘semi-essential nutrient’ has been introduced to describe the role of dietary nucleotides in nutrition of newborn, adult or ill mammals when endogenous supply is insufficient for normal body function, even though their absence from the diet does not necessarily lead to classical clinical aetiopathology(Reference Carver46).

Contents of nucleotides in milk and other food and feed ingredients

Colostrum and milk are the only nutrient supply in early life, and provide young animals with a source of nourishment which is ideally adapted to species-specific digestive and metabolic requirements at every stage of early life. Protection of the young animal against microbes is ensured via the intake of maternal immunoglobulins, lactoferrin, lysozymes, lactoperoxidase and oligosaccharides as well as leucocytes present in colostrum and milk (for a review, see Xu(Reference Xu, Xu and Cranwell47)). Furthermore, milk lipids have the potential to suppress inflammatory reactions in the piglet's gastrointestinal tract due to their antiviral, antibacterial and antiprotozoal activity(Reference Isaacs and Thormar48). Colostrum and milk assure sufficient supply with enzymes needed for digestion due to low production rates of endogenous enzymes in early life(Reference Hartmann, Holmes, Barnett and Hennessy49). Furthermore, gastrointestinal functions may be promoted by the presence of bioactive components, for example, polyamines(Reference Bardocz50), hormones and growth factors(Reference Xu, Xu and Cranwell47), as well as nucleotides(Reference Schlimme, Martin and Meisel51) that have the potential to stimulate cell division and differentiation in the small intestine, by, for example, increased mitosis rates and lower apoptosis rates(Reference Domeneghini, Di Giancamillo and Savoini11). A higher production rate of digestive enzymes in the epithelial cells may occur, thereby leading to an increased digestive and absorptive capacity of the intestine. Furthermore, according to Hartmann & Holmes(Reference Hartmann, Holmes, Barnett and Hennessy49), nucleotides present in sows' colostrum and milk belong to biologically active compounds that have the potential to influence the behaviour of piglets such as stimulating voluntary feed intake(Reference Mathew, Chattin and Robbins52Reference Van der Peet-Schwering, Jansman and Smidt55).

The nucleotide concentration in milk of mammals is species-specific and the concentration of most nucleotides changes during the lactation period (Table 1). When comparing colostrum and milk of different species, colostrum shows generally higher amounts of nucleotides, but there are species-specific differences in composition. For example, orotic acid, an intermediate product of UMP synthesis, is present in the colostrum and milk of ruminants but not of single-stomached animals (Table 1)(Reference Larson and Hegarty19).

Table 1 Comparison of the nucleotide composition (μmol/100 ml) of colostrum and milk from different species (adapted from Gil & Sanchez-Medina(Reference Gil and Sanchez-Medina122))

AMP, adenosine 5′-monophosphate; CMP, cytosine 5′-monophosphate; GMP, guanosine 5′-monophosphate; Nd, not detected; IMP, inosine 5′-monophosphate; –, not analysed; UMP, uridine 5′-monophosphate.

* Analysed with HPLC.

Analysed with enzymic methods.

The composition of sows' milk, in terms of total milk solids, crude protein (N × 6·38) and 5′-monophosphate nucleotide contents, during lactation is shown in Table 2. High concentrations of total milk solids, crude protein and UMP can be found at day 0 of lactation, followed by declining levels during the first week of lactation, while contents of AMP and GMP increase. From day 14 of lactation, the content of total milk solids, crude protein and all nucleotides decreases compared with the first 7 d of lactation. After day 21 of lactation, UMP (122·8 μmol/100 ml) dominates in sows' milk, whereas concentrations of GMP, AMP, cytosine 5′-monophosphate (CMP) and inosine 5′-monophosphate (IMP) average 6·0, 4·3, 2·3 and < 1·0 μmol/100 ml, respectively.

Table 2 Concentration of total milk solids, crude protein (nitrogen×6·38) and 5′-monophosphate nucleotides in colostrum and milk from sows at different lactation stages (adapted from Mateo et al. (Reference Mateo, Peters and Stein6))

(Mean values with their pooled standard errors)

AMP, adenosine 5′-monophosphate; CMP, cytosine 5′-monophosphate; GMP, guanosine 5′-monophosphate; IMP, inosine 5′-monophosphate; UMP, uridine 5′-monophosphate.

Nucleotides, particularly IMP, are mainly found in protein-rich food ingredients(Reference Clifford and Story5), such as organ meats, poultry and seafood(Reference Clifford and Story5, Reference Barness39, Reference Kojima56). Yeast protein sources, such as baker's or brewer's yeast and yeast extract, are ingredients that also show a relatively high concentration of nucleotides(Reference Maloney, Kroschwitz and Howe-Grant57Reference Tibbetts, Lyons and Jacques59). However, feed ingredients commonly used in animal nutrition are not routinely analysed for nucleotide content, resulting in limited information for many feedstuffs. The few data that are available show that, in comparison with colostrum and milk, there are rather low concentrations of individual nucleotides in feedstuffs such as barley, soya protein concentrate and soyabean meal (Table 3)(Reference Mateo, Stein, Lyons and Jacques60). For example, the concentration of UMP in milk amounts to 122·8 μmol/100 ml on lactation day 21 (Table 2), whereas barley contains no UMP and soyabean meal only 0·9 mg/100 g (Table 3). Furthermore, AMP, CMP, GMP and IMP concentrations vary in feedstuffs between non-detectable levels (for example, GMP content of casein) and 1·9 mg/100 g (AMP content of dried whey). Higher concentrations can be found for AMP in spray-dried erythrocytes (4·4 mg/100 g), for CMP in dried whey (27·0 mg/100 g) and in fish meal (2·6 mg/100 g) as well as for IMP in fish meal (3·5 mg/100 g).

Table 3 Nucleotide concentration (mg/100 g, as is) in commonly used feed ingredients (adapted from Mateo & Stein(Reference Mateo, Stein, Lyons and Jacques60))

5′-CMP, cytosine 5′-monophosphate; 5′-AMP, adenosine 5′-monophosphate; 5′-GMP, guanosine 5′-monophosphate; 5′-UMP, uridine 5′-monophosphate; 5′-IMP, inosine 5′-monophosphate.

Biological effects of dietary nucleotides

Physiological, morphological and metabolic effects

Hepatic morphology and function

Several studies with rodents and cats are available focusing on potential effects of supplemental dietary nucleotides and/or nucleosides on blood parameters(Reference Novak, Carver and Barness14), on hepatic blood flow(Reference Lautt, Legare and D'Almeida61) and on liver morphology(Reference Ogoshi, Iwasa and Kitagawa62). Novak et al. (Reference Novak, Carver and Barness14) reported increased hepatic cholesterol and lipid P concentration as well as decreased liver weight and glycogen content when feeding nucleotide-free diets to weanling mice, in comparison with mice receiving nucleotide-enriched diets. They concluded that dietary nucleotide supplementation in general may affect hepatic growth and composition in weanling mice. However, these differences were more pronounced if the diet was supplemented with AMP alone rather than with a mixture of different nucleotides including AMP, CMP, GMP, IMP and UMP. Furthermore, the effects of added extracellular nucleotides or nucleosides, and their mixture, on primary monolayer cultures of normal hepatocytes and cultures of hepatoma cells (AH130) on DNA and RNA syntheses was studied by Ohyanagi et al. (Reference Ohyanagi, Nishimatsu and Kanbara63), who found that a mixture of nucleotides and nucleosides at a concentration of 335 mg/dl (3350 mg/l) inhibited the syntheses of DNA and RNA by hepatocytes and hepatoma cells. Also, they found that excessive concentrations of nucleotide and nucleoside mixtures reduce the proliferation of both hepatocytes and hepatoma cells. However, Ohyanagi et al. (Reference Ohyanagi, Nishimatsu and Kanbara63) assumed that the addition of mixtures of nucleosides and nucleotides at lower levels (33·5 mg/dl; 335 mg/l) would enhance the growth of hepatocytes, particularly during liver regeneration, but on the other hand they may also enhance the proliferation of tumour cells in the liver. These findings were confirmed in studies with rats(Reference Ogoshi, Iwasa and Kitagawa62) and rabbits(Reference Yamaguchi, Takada and Shimahara64) that were diagnosed with liver damage after hepatectomy. For example, rats receiving total parenteral nutrition with a nucleotide and nucleoside mixture after 70 % hepatectomy showed earlier restoration of their N balance when compared with animals that were not supplied with this mixture(Reference Ogoshi, Iwasa and Yonezawa65). Further studies by Ogoshi et al. (Reference Ogoshi, Iwasa and Kitagawa62) showed that parenteral nutrition based on a balanced mixture of nucleosides and nucleotides at a physiological level may improve hepatic function in rats and promote earlier restoration of N balance following liver injury. These findings were confirmed in experiments with hepatectomised rabbits which showed an enhanced mitochondrial phosphorylative activity in the remnant liver, when fed a nucleoside–nucleotide mixture(Reference Yamaguchi, Takada and Shimahara64). Similarly, in hepatectomised rabbits, a continuous intravenous infusion of a nucleoside–nucleotide mixture led to an increased mitochondrial phosphorylative activity and elevated DNA content of the remnant liver(Reference Jin, Yamaguchi and Shimahara66).

Lipid metabolism

A study with human infants revealed no differences in HDL-cholesterol levels when a group receiving a nucleotide-enriched diet was compared with a breast-fed and a control group (milk formula-fed without nucleotide supplementation)(Reference Sanchez-Pozo, Pita and Martinez67). However, the authors found lower VLDL-cholesterol concentrations and increased HDL concentrations in the nucleotide-fed and breast-fed groups than in the control group. These findings suggest that nucleotides may play a role in lipoprotein synthesis(Reference Gil68). Moreover, nucleotides eventually interfere with the formation of LDL-cholesterol by affecting apoprotein synthesis(Reference Uauy and Lebenthal7). Similarly, orotic acid, an intermediate product of UMP synthesis, may induce fatty liver in rats by blocking VLDL secretion due to an inhibition of hepatic apo B synthesis(Reference Von Euler, Rubin and Handschumacher69Reference Pottenger and Getz71).

Feeding a nucleotide-supplemented formula to rats has been associated with higher plasma levels of long-chain PUFA(Reference Ramírez, Hortelano and Boza72, Reference Boza, Jimenez and Faus73) and, according to the results of a study by Jimenez et al. (Reference Jimenez, Boza and Suarez74), supplementation of nucleotides (AMP, CMP, GMP, IMP, UMP) to diets of rats enhanced the concentrations (mainly of the n-6 series) of long-chain PUFA of erythrocyte membranes compared with the control group. The authors, therefore, suggest that dietary nucleotides may have the potential to modify the conversion of essential fatty acids into their long-chain derivates. Ramírez et al. (Reference Ramírez, Hortelano and Boza72) obtained an increase in the prostaglandin thromboxane (TXA2), and a trend towards higher prostaglandin 6-keto-PGF levels when a semi-purified standard diet for weanling rats was supplemented with 50 mg/100 g each of the nucleotides AMP, CMP, GMP, IMP and UMP. These results coincided with an increased plasma concentration of arachidonic acid, a precursor for the production of the eicosanoids TXA2 and PGI2. The authors concluded that dietary purines and pyrimidines positively affect PUFA composition and therefore eicosanoid synthesis.

Dietary nucleotides may promote faster recoveries from nodular cirrhosis of the liver artificially induced by thioacetamide. For example, the addition of dietary nucleotides (AMP, CMP, GMP, IMP, UMP) following thioacetamide administration induced a decrease in the percentage area of fibrous septae in the liver of rats(Reference Torres, Fernandez and Gil75). Furthermore, the authors found a reduced total area of fibrosis in animals with liver cirrhosis after feeding the nucleotide-enriched diet for 2 weeks. In a study by Fontana et al. (Reference Fontana, Moreira and Torres76), rats with experimental cirrhosis induced by carbon tetrachloride (CCl4) showed lower saturated (mainly stearic acid), monounsaturated and n-6 long-chain PUFA (mainly arachidonic acid) concentrations as well as a reduced unsaturation index in plasma. Administration of a diet supplemented with nucleotides (AMP, GMP, CMP, IMP, UMP) to CCl4-treated rats led to a microsomal fatty acid profile closer to the group that received the semi-purified diet without CCl4 treatment. Furthermore, a high level of (n − 6) PUFA, particularly arachidonic acid, was observed in liver microsomal phospholipids of the rats fed the nucleotide-supplemented diet. The authors concluded that nucleotides are important for the recovery of liver function following hepatic damage.

Intestinal morphology and function

In the intestine, exogenous nucleotides are important for rapidly dividing mucosal cells, due to absent(Reference Savaiano and Clifford24) or limited(Reference LeLeiko, Bronstein and Baliga25)de novo nucleotide synthesis. Some studies in single-stomached animals have focused on the effect of supplemental commercial products containing pure nucleotides or nucleotide-enriched diets on the intestinal ultrastructure(Reference Martinez-Puig, Manzanilla and Morales15), on intestinal cell proliferation(Reference Sato, Nakano and Kawakami77), but also on intestinal enzyme activities(Reference Ortega, Gil and Sánchez-Pozo78), and on recovery from diarrhoea(Reference Arnaud, Lopez-Pedrosa and Torres79). For example, intestinal cell proliferation could be increased by adding a nucleoside–nucleotide mixture to a diet for rats(Reference Tsujinaka, Iijima and Kido80) or by supplementing pigs' diets with a commercial product containing nucleotides(Reference Domeneghini, Di Giancamillo and Savoini11). Under in vitro conditions, exogenous nucleotides (AMP, CMP, GMP, IMP, UMP) did not affect human colon tumour cell line (Caco-2 cell) proliferation, although significantly higher maltase and sucrase activities could be found(Reference Sato, Nakano and Kawakami77). In the same study, however, an enhancement of cell proliferation and maltase activity could be observed in a normal small-intestinal crypt cell line (intestinal epithelial cell-6) of rats. Under in vivo conditions with early-weaned rats, these authors reported higher small-intestinal sucrase activities, and they concluded that dietary nucleotides may enhance enterocyte proliferation and/or maturation both under in vitro and in vivo conditions. However, N Sauer, M Eklund, S Roth and R Mosethin (unpublished results) could not confirm these findings in a study with piglets fed a commercial product containing nucleotides, since α-amylase, maltase, lactase and leucine aminopeptidase activities in ileal digesta on days 13 and 20 after weaning were not affected. Furthermore, Lee et al. (Reference Lee, Liu and Chen44) found an increase in gastric pepsin and jejunal alkaline phosphatase activity, associated with a decrease in ileal aminopeptidase, sucrase and maltase activity, following the supplementation of the diet for weaned pigs with 0·1 % of a commercially available nucleotide product. Moreover, a study with rats revealed that withdrawal of dietary nucleotides led to a decrease in the content and specific activity of alkaline phosphatase, leucine aminopeptidase, maltase, sucrase and lactase in the villous tip(Reference Ortega, Gil and Sánchez-Pozo78). These authors hypothesised that dietary nucleotides may affect the maturation status of the small-intestinal epithelium, since the aforementioned enzymes are maturation markers of intestinal cells.

The addition of a commercial nucleotide-containing product to the diet of weanling piglets may influence intestinal morphology, as indicated by a higher mitosis and lower apoptosis rate(Reference Domeneghini, Di Giancamillo and Savoini11) and a decrease in villous atrophy after weaning(Reference Martinez-Puig, Manzanilla and Morales15). For example, Shen et al. (Reference Shen, Piao and Kim81) found a higher villous height and villous height:crypt depth ratio in a yeast-supplemented group compared with an unsupplemented group. Furthermore, Godlewski et al. (Reference Godlewski, Bierła and Strzałkowski82) showed an increase in the rate of apoptosis as well as lower DNA damage and lower mitosis apoptosis index in pigs fed a diet supplemented with a commercial product containing nucleotides in comparison with the unsupplemented control treatment. Similarly, adding nucleosides, the degradation products of nucleotides, to the diets of rats enhanced mucosal protein and intestinal DNA content(Reference Uauy, Stringel and Thomas17). On the other hand, studies by van der Peet-Schwering et al. (Reference Van der Peet-Schwering, Jansman and Smidt55) did not indicate an increased villous length and crypt depth when adding a commercial product based on yeast culture to diets for piglets.

There are further contradictory results with regard to possible effects of dietary nucleotides on nutrient digestibilities. It can be speculated if these differences can be attributed to differences in origin and dietary level of nucleotides, to variations in the adaptation period of the administered nucleotides before the start of the experiment and/or to differences in the age of the experimental animals. For example, in a study by Kornegay et al. (Reference Kornegay, Rhein-Welker and Lindemann83) with piglets (aged 25–29 d) there was no effect on apparent digestibilities of DM, N, neutral-detergent fibre and acid-detergent fibre, as well as on P, when 0·75 % of a commercial product containing nucleotides was fed. Similarly, N Sauer, M Eklund, S Roth and R Mosethin (unpublished results) found no effects on ileal digestibilities of DM, crude protein, crude ash and crude fibre in piglets (aged 24–38 d) fed a diet supplemented with 1000 parts per million of a commercial product containing nucleotides. However, Shen et al. (Reference Shen, Piao and Kim81) reported higher apparent digestibilities of DM, crude protein and net energy in piglets (aged 21–42 d) fed a diet supplemented with 5 g/kg of a yeast culture product compared with the control group. On the other hand, van Heugten et al. (Reference Van Heugten, Funderburke and Dorton53) observed lower digestibilities of DM, fat and net energy in the pre-starter phase, and also a decrease in DM, fat, P and net energy digestibility in the starter phase of piglets (aged 17–45 d) which were fed a live yeast product (1·6 × 10Reference Uauy and Lebenthal7 colony-forming units Saccharomyces cerevisiae/g feed) in comparison with the control treatment.

After recovery from diarrhoea, rats fed a nucleotide-enriched diet showed an intestinal histology and ultrastructure close to those of the normal uninfected control group(Reference Bueno, Torres and Almendros84). The authors suggested that dietary nucleotides may be important nutrients for intestinal repair following infection. Accordingly, Nunez et al. (Reference Nunez, Ayudarte and Morales85) reported in weanling rats faster intestinal recovery after chronic diarrhoea, as indicated by higher intestinal DNA contents and disaccharidase activities when adding nucleotides (AMP, CMP, GMP, IMP, UMP) to a basal diet with lactose as the main carbohydrate compared with a basal diet containing glucose polymers as the main dietary carbohydrate source. Additionally, Arnaud et al. (Reference Arnaud, Lopez-Pedrosa and Torres79) reported that nucleotide supplementation (AMP, CMP, GMP, IMP, UMP) of diets of rats with lactose-induced diarrhoea resulted in faster recoveries for the nucleotide-supplemented group than for the control group. The authors concluded that dietary nucleotides are able to promote earlier restoration of the ileal mitochondrial function after chronic diarrhoea. With regard to weaning-associated diseases in pigs, few studies have been performed investigating the effect of supplemental nucleotides on the disposition to diarrhoea. Martinez-Puig et al. (Reference Martinez-Puig, Manzanilla and Morales15) observed faster recovery from diarrhoea when a commercial product containing nucleotides was included in a diet of weanling piglets. On the other hand, according to Bekaert et al. (Reference Bekaert, Moermans and Eeckhout86), there was no effect on the incidence of diarrhoea in pigs fed a diet supplemented with a yeast culture (Saccharomyces cerevisiae).

With regard to the use of dietary nucleotides in weaned pigs, possible effects on Fe absorption might be of importance, as Fe is a mineral which frequently becomes deficient in swine diets (for a review, see National Research Council(87)). The hepatic Fe stores of the newborn piglet combined with the low Fe concentration in sows' milk are not sufficient to meet the pig's Fe need for rapid growth and increase in blood volume(Reference Rincker, Hill and Link88). Consequently, the use of exogenous sources of Fe to prevent Fe deficiency in neonatal pigs has been well documented(Reference Ullrey, Miller and West89, Reference Kernkamp, Clawson and Ferneyhough90). However, the bioavailability of Fe from different sources may vary considerably(Reference Kornegay91, Reference Deming and Czarnecki-Maulden92), and, according to Rincker et al. (Reference Rincker, Hill and Link88), is not only dependent on the Fe status of the animal but also on dietary Fe as well as on various nutritional and non-nutritional substances within the diet. Dietary nucleotides, particularly the degradation product of IMP, inosine, might play a role in facilitating intestinal Fe absorption(Reference Cheney and Finch93). Cheney & Finch(Reference Cheney and Finch93) found an improved Fe absorption in rats following intravenous inosine administration. Furthermore, Faelli & Esposito(Reference Faelli and Esposito94) investigated the effect of inosine supplementation on Fe absorption in the everted gut of rats. The authors confirmed an increase in Fe absorption and suggested that nucleotides in milk may affect the gut microflora indirectly by increasing Fe absorption in the small intestine.

Immunological effects

Various studies have focused on the effect of supplemental dietary nucleotides on the immune function in human subjects(Reference Carver, Pimentel and Cox95) and pigs(Reference Cameron, Wong, Hinch, Lindberg and Ogle96); however, most studies have been performed in rodents(Reference Nagafuchi, Katayanagi and Nakagawa13). For example, feeding nucleotide-supplemented diets to rodents has been associated with an increase in graft v. host disease mortality(Reference Kulkarni, Bhateley and Zander97), a rejection of allogenic grafts(Reference Van Buren, Kulkarni and Rudolph98, Reference Van Buren, Kulkarni and Schandle99) and a delayed cutaneous hypersensitivity(Reference Kulkarni, Fanslow and Rudolph100). However, other studies with rodents have revealed faster recovery of malnutrition- and starvation-induced immunosuppression(Reference Pizzini, Kumar and Kulkarni101, Reference Van Buren, Rudolph and Kulkarni102), an increase in antigen-induced lymphoproliferation(Reference Van Buren, Rudolph and Kulkarni102) as well as higher natural killer cell activity and IL-2 production(Reference Carver, Cox and Barness103). Additionally, resistance to a challenge with Candida albicans (Reference Fanslow, Kulkarni and van Buren104) and Staphylococcus aureus (Reference Kulkarni, Fanslow and Rudolph105) by activating humoral and cell-mediated immunity has been observed in mice fed diets enriched with nucleotides originating from yeast and RNA, respectively. The results of several studies pertaining to possible effects of dietary nucleotides on immune function in mice are summarised in Table 4.

Table 4 Effect of dietary nucleotides on immune functions in mice

 ↑ , Increase; ↓ , decrease; Cp, commercial product containing nucleotides; IFN, interferon; CMP, cytosine 5′-monophosphate; GMP, guanosine 5′-monophosphate; IMP, inosine 5′-monophosphate; UMP, uridine 5′-monophosphate; TCR, T-cell receptor; TGF, transforming growth factor; IEL, intra-epithelial lymphocytes; Nr, not reported; AMP, adenosine 5′-monophosphate.

Dietary nucleotides may also affect immune function in pigs. For example, Godlewski et al. (Reference Godlewski, Bierła and Strzałkowski82) observed higher autophagy capacity in the mid-jejunum of pigs fed a diet supplemented with a commercial nucleotide product compared with the control group. Additionally, Domeneghini et al. (Reference Domeneghini, Di Giancamillo and Savoini11) reported differences in the growth and maturation of the intestinal mucosa by adding 0·05 % nucleotides to a weanling piglet diet, especially as indicated by higher percentages of macrophages in the intestine and higher intra-epithelial lymphocytes. Due to close and intimate contact with the epithelial cells and the environment, higher numbers of intra-epithelial lymphocytes, components of the intra-epithelial defensive system, play an important role in mucosal immunity. They possess a number of pathogen-specific and non-specific functions enabling them to play a critical role in host defences at mucosal surfaces. For example, higher intra-epithelial lymphocytes have been shown to possess pathogen-specific cytotoxic T-cell activity(Reference Couedel-Courteille, Le Grand and Tulliez106), and to be capable of mediating delayed-type hypersensitivity responses(Reference Shields and Parrott107). Moreover, they also secrete various cytokines such as interferon-γ, TNF-α and IL-2(Reference Dillon, Dalton and MacDonald108, Reference Lundqvist, Melgar and Yeung109), which may play an important role in immune modulation at the mucosal surfaces. Furthermore, Lee et al. (Reference Lee, Liu and Chen44) demonstrated an immunomodulating effect as indicated by increased levels of plasma immunoglobulins (IgA, IgM) in weaned pigs when fed a commercial product containing nucleotides at 0·1 %. A study by Cameron et al. (Reference Cameron, Wong, Hinch, Lindberg and Ogle96) revealed that feeding nucleotides to early-weaned piglets may enhance T-cell functions in delayed hypersensitivity to antigenic challenge in the intestine and in vitro lymphocyte proliferation response to concanavalin A, a mitogen which stimulates T-lymphocytes. The duration of the nucleotide supplementation seems to be critical for exhibiting effects on cell functions and cell proliferation, as the immuno-enhancing effects described above require at least 2 weeks of administration to the pigs(Reference Cameron, Wong, Hinch, Lindberg and Ogle96). However, a higher mitotic activity in the ileum could be shown when feeding a combination of 0·05 % nucleotides and 0·5 % glutamine. Thus, the inclusion of nucleotides and glutamine to a weanling piglet diet appears to have a positive effect on the maturation of the ileal mucosa by supporting the development of lymphocytes and thereby providing an immune-reactive intestine. Furthermore, Salobir et al. (Reference Salobir, Rezar and Pajk110) found, upon feeding a diet supplemented with different levels of PUFA to growing pigs, that a commercial product of nucleotides may effectively eliminate the genotoxic effect of high PUFA intakes on blood lymphocytes, thus demonstrating new evidence for the immuno-nutritive potential of dietary nucleotides.

Microbial effects

Few in vitro studies have been performed pertaining to possible effects of supplemental nucleotides or nucleosides on the growth of single bacterial strains; however, results are rather controversial. For example, an in vitro study of Tanaka & Mutai(Reference Tanaka and Mutai111) showed that nucleosides (adenine, guanine, xanthine, uracil) in combination with pyruvic acid and riboflavin in a minimal culture medium promote the growth of bifidobacteria. Accordingly, Uauy(Reference Uauy and Lebenthal7) summarised that Gil(Reference Gil112) obtained a slight but significant stimulation of bifidobacterial in vitro growth upon the addition of single nucleotides (AMP, CMP, GMP, IMP, UMP) to a minimal medium. This stimulatory effect was even more pronounced when a mixture of these monophosphate nucleotides was included in the assay. On the other hand, Sauer et al. (Reference Sauer, Bauer and Vahjen113) obtained no differences in the growth responses of Lactobacillus reuteri (strain DSM 20 016), L. amylovorus (DSM 20 513), Enterococcus faecium (DSM 2146) and E. faecalis (DSM 6134) when adding single nucleotides (AMP, CMP, GMP, IMP, UMP) to a minimal medium. However, the authors found diverse growth responses for different Escherichia coli strains (E. coli DSM 2840, E. coli PS 37, E. coli PS 79) upon the addition of single nucleotides to a minimal medium, depending on the type of nucleotide as well as on the concentration used, but also on the specific bacterial strain tested. These results suggest that bacteria might be able to use nucleosides and/or nucleotides for their proliferation, although this has to be confirmed by further in vitro studies by expanding the range of bacterial strains under investigation.

Roselli et al. (Reference Roselli, Britti and Le Huërou-Luron114) evaluated the potential of several natural substances (for example, yeast extract) to protect against enterotoxigenic Escherichia coli (ETEC) K88-induced membrane damage by using intestinal pig epithelial cells-1. The authors showed that a commercial product containing nucleotides could protect the cells against an increase in membrane permeability caused by enterotoxigenic Escherichia coli. Additionally, results from an in vivo study with human subjects suggest that dietary nucleotides may modify the composition of the intestinal microflora by promoting beneficial bacteria including bifidobacteria and lactobacilli(Reference Gil, Corral and Martínez16). Young infants fed a formula containing dietary nucleotides (AMP, CMP, GMP, IMP, UMP) had higher percentages of faecal bifidobacteria and lactobacilli and lower percentages of Gram-negative enterobacteria than formula-fed infants without supplementation(Reference Gil, Corral and Martínez16). High numbers of intestinal bifidobacteria and lactobacilli are required, in particular around the time of weaning during the transition from liquid to solid feeding. For example, bifidobacteria may lower the pH of the colonic content due to their capacity to hydrolyse sugars to lactic acid which, in turn, may suppress the proliferation of acid-susceptible pathogenic bacteria. Bifidobacteria also may inhibit the growth of enterobacteria responsible for diarrhoeal diseases (for a review, see Braun(Reference Braun and Lebenthal115)). However, only few studies are available pertaining to the effect of dietary nucleotides on the composition and activity of the intestinal microflora of young pigs. For example, Andrés-Elias et al. (Reference Andrés-Elias, Pujols and Badiola4) concluded that nucleotides appear to modulate the intestinal ecology in weanling piglets by stabilising the microbiota in the ileum, when testing nucleotides (commercially available product) as possible alternatives to antimicrobial growth promoters. However, Mathew et al. (Reference Mathew, Chattin and Robbins52) could not show any differences in total ileal numbers of Escherichia coli, lactobacilli and streptococci as well as in microbial metabolites (SCFA composition), when feeding yeast cultures (Saccharomyces cerevisiae) to weanling piglets. Similarly, N Sauer, M Eklund, S Roth and R Mosethin (unpublished results) failed to observe a response in ileal counts of lactobacilli, bifidobacteria, enterococci and eubacteria as well as in the level of microbial metabolites, including SCFA and lactic acids, when feeding a diet to weaning piglets supplemented with a commercial product containing nucleotides. Also, no changes in the PCR-denaturing gradient gel electrophoresis fingerprint analysis of total bacterial community profile could be shown when adding nucleotides (yeast culture) to diets of pigs(Reference Van der Peet-Schwering, Jansman and Smidt55). Results of studies pertaining to the effect of nucleotides, given either as single nucleotides or originating from yeast products, on the composition and activity of the intestinal microbiota of humans and pigs are still equivocal.

Zootechnical effects

Several studies have been conducted focusing on the growth-promoting effect of dietary nucleotides, supplemented as yeast products to pig diets (Table 5). In several studies dietary nucleotides as a yeast product were added to the diets of pigs of different ages but failed to improve growth performance(Reference Andrés-Elias, Pujols and Badiola4, Reference Domeneghini, Di Giancamillo and Savoini11, Reference Van Heugten, Funderburke and Dorton53, Reference Van der Peet-Schwering, Jansman and Smidt55, Reference Kornegay, Rhein-Welker and Lindemann83, Reference Bekaert, Moermans and Eeckhout86, Reference Cameron, Wong, Hinch, Lindberg and Ogle96, Reference Bowman and Veum116, Reference Veum and Bowman117). Similarly, a study by Sijben et al. (Reference Sijben, van Vugt and Swinkels118) showed that dietary yeast nucleotides (Saccharomyces cerevisiae) did not affect energy intake, heat production and energy retention of barrows. On the other hand, supplementation of yeast products to diets of piglets resulted in similar improvements in growth performance as compared with antimicrobial growth promoters(Reference Van der Peet-Schwering, Jansman and Smidt55, Reference Shen, Piao and Kim81). Similarly, Mathew et al. (Reference Mathew, Chattin and Robbins52) observed a trend towards an increased growth rate in pigs fed a diet supplemented with live yeast (Saccharomyces cerevisiae) from weaning until age 41 d. Furthermore, the addition of nucleotide products alone or in combination with glutamine to diets of weaning piglets has revealed contradictory results. Domeneghini et al. (Reference Domeneghini, Di Giancamillo and Savoini11) observed no difference in body weight and feed intake, whereas in a study by Yi et al. (Reference Yi, Allee and Toride119) a combination of nucleotides (RNA) and glutamine improved average daily gain and feed intake. Further studies investigated sows' and piglets' performance upon supplementation of sows' diets with yeast products. Jurgens et al. (Reference Jurgens, Rikabi and Zimmerman120) and Moore et al. (Reference Moore, D'Souza, Mullan, Paterson and Barker121) found higher growth performance in piglets when adding live yeast (Saccharomyces cerevisiae) and a commercial product containing Saccharomyces cerevisiae, respectively, to sows' diets.

Table 5 Zootechnical effects of nucleotide supplementation to diets of pigs compared with the control diet

–, No effect; S.c., Saccharomyces cerevisiae; ↓ , decrease; Cp, commercial product containing nucleotides; ↑ , increase; cfu, colony-forming units; ppm, parts per million.

Conclusions

There is increasing evidence that dietary nucleotides may affect different aspects of body function and health in single-stomached animals, including intestinal morphology and function, immune response, the composition of the intestinal microbiota, liver function and morphology as well as growth performance. Although nucleotides can be synthesised de novo, several tissues, including those with a rapid turnover rate such as the intestine, may benefit from an exogenous supply to decrease energy expenditures. In times of disease states, periods of limited nutrient intake or rapid growth, such as the weaning period, nucleotides can become semi-essential nutrients, as in general nucleotides are present at very low levels in animal diets. Furthermore, due to the ban of in-feed antibiotics, efficient and feasible alternatives have gained increasing attention to improve the health of animals, to increase the potential to withstand infectious diseases and to secure growth performance. For dietary nucleotides, beneficial effects have been proven for a faster intestinal recovery after challenge with, for example, diarrhoea, and also for a faster immune response in several disease situations, for example, malnutrition. A decreased permeability of the lamina propria and an increased stabilisation of the intestinal microbial ecosystem by promoting beneficial bacteria may further improve animal health. Additionally, there is evidence that IMP may facilitate Fe absorption, which is of special interest in pigs as Fe has been acknowledged as a mineral which frequently becomes deficient in pig nutrition.

However, results of studies pertaining to biological effects of nucleotides in single-stomached animals are equivocal. In most studies, commercially available yeast products have been assayed both under in vitro and in vivo conditions. Since these yeast culture products contain variable amounts of nucleotides in addition to viable cells, cell wall components, and parts of the medium on which the yeast cells were grown, it is difficult to assign the observed biological effects exclusively to specific nucleotides present in these products. Studies in which the biological effects of purified sources of nucleotides, alone or in combination, were assayed are scarce. Therefore, further research should be directed to the evaluation of the efficiency of purified nucleotides, including the effects of varying concentrations and different combinations of nucleotides. In addition, future research should also focus on the potential associated effects of nucleotides if supplied in combination with other bioactive substances, for example, glutamine.

Finally, advanced knowledge about the contents of nucleotides present in commonly used feed components would facilitate the development of feeding strategies to meet animals' requirements for nucleotides, in particular in young animals suffering from various kinds of stress.

Acknowledgements

The present review received no specific grant from any funding agency in the public, commercial or not-for-profit sectors.

We thank Ingrid Neff for assisting in preparing the manuscript.

Conceptualisation and implementation of the manuscript was performed by R. M. and E. B. The manuscript was written by N. S. All authors reviewed the manuscript and approved submission. There are no conflicts of interest.

References

1Bager, F, Aarestrup, FM & Wegener, HC (2000) Dealing with antimicrobial resistance – the Danish experience. Can J Anim Sci 80, 223228.CrossRefGoogle Scholar
2Cummins, AG, Steele, TW, LaBrooy, JT, et al. . (1988) Maturation of the rat small intestine at weaning: changes in epithelial cell kinetics, bacterial flora, and mucosal immune activity. Gut 29, 16721679.Google Scholar
3Pluske, JR, Hampson, DJ & Williams, IH (1997) Factors influencing the structure and function of the small intestine in the weaned pig: a review. Livest Prod Sci 51, 215236.CrossRefGoogle Scholar
4Andrés-Elias, N, Pujols, J, Badiola, I, et al. . (2007) Effect of nucleotides and carob pulp on gut health and performance of weanling piglets. Livest Sci 108, 280283.CrossRefGoogle Scholar
5Clifford, AJ & Story, DL (1976) Levels of purines in foods and their metabolic effects in rats. J Nutr 106, 435442.CrossRefGoogle Scholar
6Mateo, CD, Peters, DN & Stein, HH (2004) Nucleotides in sow colostrum and milk at different stages of lactation. J Anim Sci 82, 13391342.CrossRefGoogle ScholarPubMed
7Uauy, R (1989) Dietary nucleotides and requirements in early life. In Textbook of Gastroenterology and Nutrition in Infancy, 2nd ed., pp. 265280 [Lebenthal, E, editor]. New York: Raven Press, Ltd.Google Scholar
8Cosgrove, M (1998) Nucleotides. Nutrition 14, 748751.Google Scholar
9Carver, JD & Walker, WA (1995) The role of nucleotides in human nutrition. J Nutr Biochem 6, 5872.CrossRefGoogle Scholar
10Taylor-Pickard, J (2008) Dietary nucleotides – the foundation for the future. Int Pig Top 23, 79.Google Scholar
11Domeneghini, C, Di Giancamillo, A, Savoini, G, et al. . (2004) Structural patterns of swine ileal mucosa following l-glutamine and nucleotide administration during the weaning period. An histochemical and histometrical study. Histol Histopathol 19, 4958.Google ScholarPubMed
12Bustamante, SA (1990) Intestinal hyperemia evoked by nucleotides in newborn swine. Pediatr Res 27, 102(A).Google Scholar
13Nagafuchi, S, Katayanagi, T, Nakagawa, E, et al. . (1997) Effects of dietary nucleotides on serum antibody and splenic cytokine production in mice. Nutr Res 17, 11631174.CrossRefGoogle Scholar
14Novak, DA, Carver, JD & Barness, LA (1994) Dietary nucleotides affect hepatic growth and composition in the weanling mouse. JPEN J Parenter Enteral Nutr 18, 6266.CrossRefGoogle ScholarPubMed
15Martinez-Puig, D, Manzanilla, EG, Morales, J, et al. . (2007) Dietary nucleotide supplementation reduces occurrence of diarrhoea in early weaned pigs. Livest Sci 108, 276279.Google Scholar
16Gil, A, Corral, E, Martínez, A, et al. . (1986) Effects of the addition of nucleotides to an adapted milk formula on the microbial pattern of faeces in at term newborn infants. J Clin Nutr Gastroenterol 1, 127132.Google Scholar
17Uauy, R, Stringel, G, Thomas, R, et al. . (1990) Effect of dietary nucleosides on growth and maturation of the developing gut in the rat. J Pediatr Gastroenterol Nutr 10, 497503.Google Scholar
18Carver, JD (1994) Dietary nucleotides: cellular immune, intestinal and hepatic system effects. J Nutr 124, 144S148S.Google Scholar
19Larson, BL & Hegarty, HM (1979) Orotic acid in milks of various species and commercial dairy products. J Dairy Sci 62, 16411644.CrossRefGoogle Scholar
20Nelson, DL & Cox, MM (2008) Nucleotides and nucleic acids. In Lehninger Principles of Biochemistry, 5th ed., pp. 271–298 [Nelson, DL and Cox, MM, editors]. New York: WH Freeman and Company.Google Scholar
21Gil, A, Gomez-Leon, C & Rueda, R (2007) Exogenous nucleic acids and nucleotides are efficiently hydrolysed and taken up as nucleosides by intestinal explants from suckling piglets. Br J Nutr 98, 285291.Google Scholar
22Boza, J (1998) Nucleotides in infant nutrition. Monatsschr Kinderheilkd 146, Suppl. 1, S39S48.Google Scholar
23Chu, SW (1991) Nucleotides: biochemistry and metabolism. Sem Pediat Gastr Nutr 2, 611.Google Scholar
24Savaiano, DA & Clifford, AJ (1981) Adenine, the precursor of nucleic acids in intestinal cells unable to synthesize purines de novo. J Nutr 111, 18161822.CrossRefGoogle ScholarPubMed
25LeLeiko, NS, Bronstein, AD, Baliga, BS, et al. . (1983) De novo purine nucleotide synthesis in the rat small and large intestine: effect of dietary protein and purines. J Pediatr Gastroenterol Nutr 2, 313319.Google Scholar
26Gil, A & Uauy, R (1995) Nucleotides and related compounds in human and bovine milks. In Handbook of Milk Composition, pp. 436464 [Jensen, RG, editor]. San Diego: Academic Press, Inc.CrossRefGoogle Scholar
27Sanderson, IR & He, Y (1994) Nucleotide uptake and metabolism by intestinal epithelial cells. J Nutr 124, 131S137S.CrossRefGoogle ScholarPubMed
28Harms, V & Stirling, CE (1977) Transport of purine nucleotides and nucleosides by in vitro rabbit ileum. Am J Physiol Gastrointest Liver Physiol 233, G47G55.Google Scholar
29Bronk, JR & Hastewell, JG (1988) The transport and metabolism of naturally occurring pyrimidine nucleosides by isolated rat jejunum. J Physiol 395, 349361.CrossRefGoogle ScholarPubMed
30Plagemann, PGW, Marz, R & Wohlhueter, RM (1978) Uridine transport in Novikoff rat hepatoma cells and other cell lines and its relationship to uridine phosphorylation and phosphorolysis. J Cell Physiol 97, 4972.CrossRefGoogle ScholarPubMed
31Marz, R, Wohlhueter, RM & Plagemann, PGW (1979) Purine and pyrimidine transport and phosphoribosylation and their interaction in overall uptake by cultured mammalian cells. J Biol Chem 254, 23292338.CrossRefGoogle ScholarPubMed
32Quan, R & Uauy, R (1991) Nucleotides and gastrointestinal development. Sem Pediat Gastr Nutr 2, 36.Google Scholar
33Markiewicz, A, Kaminski, M, Chocilowski, W, et al. . (1983) Circadian rhythms of four marker enzymes activity of the jejunal villi in man. Acta Histochem 72, 9199.Google Scholar
34Morley, DJ, Hawley, DM, Ulbright, TM, et al. . (1987) Distribution of phosphodiesterase I in normal human tissues. J Histochem Cytochem 35, 7582.Google Scholar
35Weickmann, JL, Elson, M & Glitz, DG (1981) Purification and characterization of human pancreatic ribonuclease. Biochemistry 20, 12721278.Google Scholar
36Holdsworth, G & Coleman, R (1975) Enzyme profiles of mammalian bile. Biochim Biophys Acta 389, 4750.CrossRefGoogle ScholarPubMed
37Bronk, JR & Hastewell, JG (1987) The transport of pyrimidines into tissue rings cut from rat small intestine. J Physiol 382, 475488.CrossRefGoogle ScholarPubMed
38Salati, LM, Gross, CJ, Henderson, LM, et al. . (1984) Absorption and metabolism of adenine, adenosine-5′-monophosphate, adenosine and hypoxanthine by the isolated vascularly perfused rat small intestine. J Nutr 114, 753760.CrossRefGoogle ScholarPubMed
39Barness, LA (1994) Dietary sources of nucleotides – from breast milk to weaning. J Nutr 124, 128S130S.CrossRefGoogle ScholarPubMed
40Kulkarni, AD, Rudolph, FB & van Buren, CT (1994) The role of dietary sources of nucleotides in immune function: a review. J Nutr 124, 1442S1446S.CrossRefGoogle ScholarPubMed
41Paubert-Braquet, M, Dupont, C, Hedef, N, et al. . (1992) Quantification of nucleotides in human milk and their effects on cytokine production by murine fibroblasts, J774A1 macrophages and human monocytes. In Foods, Nutrition and Immunity, 1st ed., pp. 2234 [Paubert-Braquet, M, Dupont, C and Paoletti, R, editors]. Basel: Karger.Google Scholar
42Zivkovic, D (2007) Investigations on 2,7-diamino-9-fluorenol photochemistry. PhD Thesis, University of Basel.Google Scholar
43Poggioli, J, Mauger, J-P & Claret, M (1986) Effect of cyclic AMP-dependent hormones and Ca2+-mobilizing hormones on the Ca2+ influx and polyphosphoinositide metabolism in isolated rat hepatocytes. Biochem J 235, 663669.Google Scholar
44Lee, DN, Liu, SR, Chen, YT, et al. . (2007) Effects of diets supplemented with organic acids and nucleotides on growth, immune responses and digestive tract development in weaned pigs. J Anim Physiol Anim Nutr (Berl) 91, 508518.CrossRefGoogle ScholarPubMed
45Rudolph, FB, Kulkarni, AD, Schandel, VB, et al. . (1984) Involvement of dietary nucleotides in T-lymphocyte function. Adv Exp Med Biol 165B, 175178.CrossRefGoogle Scholar
46Carver, JD (1999) Dietary nucleotides: effects on the immune and gastrointestinal systems. Acta Paediatr Suppl 88, 8388.CrossRefGoogle ScholarPubMed
47Xu, RJ (2003) Composition of porcine milk. In The Neonatal Pig – Gastrointestinal Physiology and Nutrition, pp. 213246 [Xu, RJ and Cranwell, P, editors]. Nottingham: Nottingham University Press.Google Scholar
48Isaacs, CE & Thormar, H (1991) The role of milk-derived antimicrobial lipids as antiviral and antibacterial agents. Adv Exp Med Biol 310, 159165.Google Scholar
49Hartmann, PE & Holmes, MA (1989) Sow lactation. In Manipulating Pig Production II, pp. 7279 [Barnett, JL and Hennessy, DP, editors]. Melbourne: Australasian Pig Science Association Publication.Google Scholar
50Bardocz, S (1993) The role of dietary polyamines. Eur J Clin Nutr 47, 683690.Google Scholar
51Schlimme, E, Martin, D & Meisel, H (2000) Nucleosides and nucleotides: natural bioactive substances in milk and colostrum. Br J Nutr 84, S59S68.CrossRefGoogle ScholarPubMed
52Mathew, AG, Chattin, SE, Robbins, CM, et al. . (1998) Effects of a direct-fed yeast culture on enteric microbial populations, fermentation acids, and performance of weanling pigs. J Anim Sci 76, 21382145.CrossRefGoogle ScholarPubMed
53Van Heugten, E, Funderburke, DW & Dorton, KL (2003) Growth performance, nutrient digestibility, and fecal microflora in weanling pigs fed live yeast. J Anim Sci 81, 10041012.CrossRefGoogle ScholarPubMed
54Carlson, MS, Veum, TL & Turk, JR (2005) Effects of yeast extract versus animal plasma in weanling pig diets on growth performance and intestinal morphology. J Swine Health Prod 13, 204209.Google Scholar
55Van der Peet-Schwering, CMC, Jansman, AJM, Smidt, H, et al. . (2007) Effects of yeast culture on performance, gut integrity, and blood cell composition of weanling pigs. J Anim Sci 85, 30993109.CrossRefGoogle ScholarPubMed
56Kojima, K (1974) Safety evaluation of disodium 5′-inosinate, disodium 5′-guanylate and disodium 5′-ribonucleotide. Toxicology 2, 185206.CrossRefGoogle Scholar
57Maloney, D (1998) Yeasts. In Kirk-Othmer Encyclopedia of Chemical Technology, 4th ed., pp. 761–788 [Kroschwitz, JI and Howe-Grant, M, editors]. New York: John Wiley and Sons, Inc.Google Scholar
58Ingledew, WM (1999) Yeast – could you base a business on this bug? In Biotechnology in the Feed Industry. Proceedings of Alltech's 15th Annual Symposium, pp. 2747 [Lyons, TP and Jacques, KA, editors]. Nottingham: Nottingham University Press.Google Scholar
59Tibbetts, GW (2002) Nucleotides from yeast extract: potential to replace animal protein sources in food animal diets. In Nutritional Biotechnology in the Feed and Food Industries. Proceedings of Alltech's 18th Annual Symposium, pp. 435443 [Lyons, TP and Jacques, KA, editors]. Nottingham: Nottingham University Press.Google Scholar
60Mateo, CD & Stein, HH (2004) Nucleotides and young animal health: can we enhance intestinal tract development and immune function? In Nutritional Biotechnology in the Feed and Food Industries. Proceedings of Alltech's 20th Annual Symposium, pp. 159168 [Lyons, TP and Jacques, KA, editors]. Nottingham: Nottingham University Press.Google Scholar
61Lautt, WW, Legare, DJ & D'Almeida, MS (1985) Adenosine as putative regulator of hepatic arterial flow (the buffer response). Am J Physiol Heart Circ Physiol 248, H331H338.CrossRefGoogle ScholarPubMed
62Ogoshi, S, Iwasa, M, Kitagawa, S, et al. . (1988) Effects of total parenteral nutrition with nucleoside and nucleotide mixture on d-galactosamine-induced liver injury in rats. JPEN J Parenter Enteral Nutr 12, 5357.Google Scholar
63Ohyanagi, H, Nishimatsu, S, Kanbara, Y, et al. . (1989) Effects of nucleosides and a nucleotide on DNA and RNA syntheses by the salvage and de novo pathway in primary monolayer cultures of hepatocytes and hepatoma cells. JPEN J Parenter Enteral Nutr 13, 5158.Google Scholar
64Yamaguchi, T, Takada, Y, Shimahara, Y, et al. . (1992) A mixture of nucleosides and a nucleotide alters hepatic energy metabolism 24 hours after hepatectomy in rabbits. J Nutr 122, 340344.CrossRefGoogle Scholar
65Ogoshi, S, Iwasa, M, Yonezawa, T, et al. . (1985) Effect of nucleotide and nucleoside mixture on rats given total parenteral nutrition after 70 % hepatectomy. JPEN J Parenter Enteral Nutr 9, 339342.Google Scholar
66Jin, MB, Yamaguchi, T, Shimahara, Y, et al. . (1996) Significance of nucleosides and a nucleotide mixture infusion on hepatic energy metabolism of 70 % hepatectomized rabbits in postoperative phase. JPEN J Parenter Enteral Nutr 20, 211214.Google Scholar
67Sanchez-Pozo, A, Pita, ML, Martinez, A, et al. . (1986) Effects of dietary nucleotides upon lipoprotein pattern of newborn infants. Nutr Res 6, 763771.Google Scholar
68Gil, A (1983) Human milk nucleotides: influences on the newborn metabolism. In Abstract of Symposia XVI International Congress of Pediatrics, Symposium Human milk: Recent knowledge, pp. 82. Manila: Philippines.Google Scholar
69Von Euler, LH, Rubin, RJ & Handschumacher, RE (1963) Fatty livers induced by orotic acid. II. Changes in nucleotide metabolism. J Biol Chem 238, 24642469.Google Scholar
70Windmueller, HG & Levy, RI (1967) Total inhibition of hepatic β-lipoprotein production in the rat by orotic acid. J Biol Chem 242, 22462254.Google Scholar
71Pottenger, LA & Getz, GS (1971) Serum lipoprotein accumulation in the livers of orotic acid-fed rats. J Lipid Res 12, 450459.CrossRefGoogle ScholarPubMed
72Ramírez, M, Hortelano, P, Boza, J Jr, et al. . (1991) Effect of dietary nucleotides and orotate on the blood levels of prostacyclin (PGI2) and thromboxane (TXA2) in the weanling rat. Prostaglandins Leukot Essent Fatty Acids 43, 4954.Google Scholar
73Boza, J, Jimenez, J, Faus, MJ, et al. . (1992) Influences of postnatal age and dietary nucleotides on plasma fatty acids in the weanling rat. JPEN J Parenter Enteral Nutr 16, 322326.CrossRefGoogle ScholarPubMed
74Jimenez, J Jr, Boza, J, Suarez, MD, et al. . (1992) Changes in fatty acid profiles of red blood cell membranes mediated by dietary nucleotides in weanling rats. J Pediatr Gastroenterol Nutr 14, 293299.Google Scholar
75Torres, MI, Fernandez, MI, Gil, A, et al. . (1997) Effect of dietary nucleotides on degree of fibrosis and steatosis induced by oral intake of thioacetamide. Dig Dis Sci 42, 13221328.Google Scholar
76Fontana, L, Moreira, E, Torres, MI, et al. . (1999) Effects of dietary polyunsaturated fatty acids and nucleotides on tissue fatty acid profiles of rats with carbon tetrachloride-induced liver damage. Clin Nutr 18, 93101.Google Scholar
77Sato, N, Nakano, T, Kawakami, H, et al. . (1999) In vitro and in vivo effects of exogenous nucleotides on the proliferation and maturation of intestinal epithelial cells. J Nutr Sci Vitaminol 45, 107118.Google Scholar
78Ortega, MA, Gil, A & Sánchez-Pozo, A (1995) Maturation status of small intestine epithelium in rats deprived of dietary nucleotides. Life Sci 56, 16231630.CrossRefGoogle ScholarPubMed
79Arnaud, A, Lopez-Pedrosa, JM, Torres, MI, et al. . (2003) Dietary nucleotides modulate mitochondrial function of intestinal mucosa in weanling rats with chronic diarrhea. J Pediatr Gastroenterol Nutr 37, 124131.Google ScholarPubMed
80Tsujinaka, T, Iijima, S, Kido, Y, et al. . (1993) Role of nucleosides and nucleotides mixture in intestinal mucosal growth under total parenteral nutrition. Nutrition 9, 532535.Google Scholar
81Shen, YB, Piao, XS, Kim, SW, et al. . (2009) Effects of yeast culture supplementation on growth performance, intestinal health, and immune response of nursery pigs. J Anim Sci 87, 26142624.CrossRefGoogle ScholarPubMed
82Godlewski, MM, Bierła, JB, Strzałkowski, A, et al. . (2009) A novel cytometric approach to study intestinal mucosa rebuilding in weaned pigs fed with dietary nucleotides. Livest Sci 123, 215220.Google Scholar
83Kornegay, ET, Rhein-Welker, D, Lindemann, MD, et al. . (1995) Performance and nutrient digestibility in weanling pigs as influenced by yeast culture additions to starter diets containing dried whey or one of two fiber sources. J Anim Sci 73, 13811389.Google Scholar
84Bueno, J, Torres, M, Almendros, A, et al. . (1994) Effect of dietary nucleotides on small intestinal repair after diarrhoea. Histological and ultrastructural changes. Gut 35, 926933.CrossRefGoogle ScholarPubMed
85Nunez, MC, Ayudarte, MV, Morales, D, et al. . (1990) Effect of dietary nucleotides on intestinal repair in rats with experimental chronic diarrhea. JPEN J Parenter Enteral Nutr 14, 598604.CrossRefGoogle ScholarPubMed
86Bekaert, H, Moermans, R & Eeckhout, W (1996) Effect of a live yeast culture in diets of weanling piglets on performances and frequency of diarrhoea. Ann Zootech 45, 369376.Google Scholar
87National Research Council (1998) Nutrient Requirements of Swine, 10th rev ed.. Washington, DC: National Academy Press.Google Scholar
88Rincker, MJ, Hill, GM, Link, JE, et al. . (2004) Effects of dietary iron supplementation on growth performance, hematological status, and whole-body mineral concentrations of nursery pigs. J Anim Sci 82, 31893197.Google Scholar
89Ullrey, DE, Miller, ER, West, DR, et al. . (1959) Oral and parenteral administration of iron in the prevention and treatment of baby pig anemia. J Anim Sci 18, 256263.CrossRefGoogle Scholar
90Kernkamp, HCH, Clawson, AJ & Ferneyhough, RH (1962) Preventing iron-deficiency anemia in baby pigs. J Anim Sci 21, 527532.Google Scholar
91Kornegay, ET (1972) Availability of iron contained in defluorinated phosphate. J Anim Sci 34, 569572.Google Scholar
92Deming, JG & Czarnecki-Maulden, GL (1989) Iron bioavailability in calcium and phosphorus sources. J Anim Sci 67, Suppl. 1, 253A.Google Scholar
93Cheney, B & Finch, CA (1960) Effect of inosine on iron absorption in rats. Proc Soc Exp Biol Med 103, 3738.Google Scholar
94Faelli, A & Esposito, G (1970) Effect of inosine and its metabolites on intestinal iron absorption in the rat. Biochem Pharmacol 19, 25512554.CrossRefGoogle ScholarPubMed
95Carver, JD, Pimentel, B, Cox, WI, et al. . (1991) Dietary nucleotide effects upon immune function in infants. Pediatrics 88, 359363.Google Scholar
96Cameron, BF, Wong, CW, Hinch, GN, et al. . (2001) Effects of nucleotides on the immune function of early-weaned piglets. In Digestive Physiology of Pigs. Proceedings of the 8th Symposium, pp. 6668 [Lindberg, JE and Ogle, B, editors]. Wallingford: CABI Publishing.Google Scholar
97Kulkarni, SS, Bhateley, DC, Zander, AR, et al. . (1984) Functional impairment of T-lymphocytes in mouse radiation chimeras by nucleotide-free diet. Exp Hematol 12, 694699.Google Scholar
98Van Buren, CT, Kulkarni, AD & Rudolph, FB (1983) Synergistic effect of a nucleotide-free diet and cyclosporine on allograft survival. Transplant Proc 15, Suppl. 1, 29672968.Google Scholar
99Van Buren, CT, Kulkarni, AD, Schandle, VB, et al. . (1983) The influence of dietary nucleotides on cell-mediated immunity. Transplantation 36, 350351.Google Scholar
100Kulkarni, AD, Fanslow, WC, Rudolph, FB, et al. . (1987) Modulation of delayed hypersensitivity in mice by dietary nucleotide restriction. Transplantation 44, 847849.Google ScholarPubMed
101Pizzini, RP, Kumar, S, Kulkarni, AD, et al. . (1990) Dietary nucleotides reverse malnutrition and starvation-induced immunosuppression. Arch Surg 125, 8689.CrossRefGoogle ScholarPubMed
102Van Buren, CT, Rudolph, FB, Kulkarni, AD, et al. . (1990) Reversal of immunosuppression induced by a protein-free diet: comparison of nucleotides, fish oil and arginine. Crit Care Med 18, Suppl. 2, S114S117.Google Scholar
103Carver, JD, Cox, WI & Barness, LA (1990) Dietary nucleotide effects upon murine natural killer cell activity and macrophage activation. JPEN J Parenter Enteral Nutr 14, 1822.Google Scholar
104Fanslow, WC, Kulkarni, AD, van Buren, CT, et al. . (1988) Effect of nucleotide restriction and supplementation on resistance to experimental murine candidiasis. JPEN J Parenter Enteral Nutr 12, 4952.Google Scholar
105Kulkarni, AD, Fanslow, WC, Rudolph, FB, et al. . (1986) Effect of dietary nucleotides on response to bacterial infections. JPEN J Parenter Enteral Nutr 10, 169171.Google Scholar
106Couedel-Courteille, A, Le Grand, R, Tulliez, M, et al. . (1997) Direct ex vivo simian immunodeficiency virus (SIV)-specific cytotoxic activity detected from small intestine intraepithelial lymphocytes of SIV-infected macaques at an advanced stage of infection. J Virol 71, 10521057.Google Scholar
107Shields, JG & Parrott, DM (1985) Appearance of delayed-type hypersensitivity effector cells in murine gut mucosa. Immunology 54, 771776.Google Scholar
108Dillon, SB, Dalton, BJ & MacDonald, TT (1986) Lymphokine production by mitogen and antigen activated mouse intraepithelial lymphocytes. Cell Immunol 103, 326338.Google Scholar
109Lundqvist, C, Melgar, S, Yeung, MM, et al. . (1996) Intraepithelial lymphocytes in human gut have lytic potential and a cytokine profile that suggest T helper 1 and cytotoxic functions. J Immunol 157, 19261934.Google Scholar
110Salobir, J, Rezar, V, Pajk, T, et al. . (2005) Effect of nucleotide supplementation on lymphocyte DNA damage induced by dietary oxidative stress in pigs. Anim Sci 81, 135140.Google Scholar
111Tanaka, R & Mutai, M (1980) Improved medium for selective isolation and enumeration of Bifidobacterium. Appl Environ Microbiol 40, 866869.Google Scholar
112Gil, A (1984) Nucleotidos en la leche humana. Fundamento para su empleo en leches infantiles (Nucleotides in Human Milk. Rationale for Use in Infant Formula). Granada: Graficas del Sur.Google Scholar
113Sauer, N, Bauer, E, Vahjen, W, et al. . (2010) Nucleotides modify growth of selected intestinal bacteria in vitro. Livest Sci 133, 161163.Google Scholar
114Roselli, M, Britti, MS, Le Huërou-Luron, I, et al. . (2007) Effect of different plant extracts and natural substances (PENS) against membrane damage induced by enterotoxigenic Escherichia coli K88 in pig intestinal cells. Toxicol In Vitro 21, 224229.Google Scholar
115Braun, OH (1981) Effect of consumption of human milk and other formulas on intestinal bacterial flora in infants. In Textbook of Gastroenterology and Nutrition in Infancy, vol. 1. Gastrointestinal Development and Perinatal Nutrition, 1st ed., pp. 247253 [Lebenthal, E, editor]. New York: Raven Press.Google Scholar
116Bowman, GL & Veum, TL (1973) Saccharomyces cerevisiae yeast culture in growing-finishing swine diets. J Anim Sci 37, 7274.Google Scholar
117Veum, TL & Bowman, GL (1973) Saccharomyces cerevisiae yeast culture in diets for mechanically-fed neonatal piglets and early growing self-fed pigs. J Anim Sci 37, 6771.Google Scholar
118Sijben, JWC, van Vugt, PNA, Swinkels, JWGM, et al. . (1998) Energy metabolism of immunized weanling piglets is not affected by dietary yeast. J Anim Physiol Anim Nutr 79, 153161.Google Scholar
119Yi, GF, Allee, GL, Toride, Y, et al. . (2002) Impact of glutamine, glutamate, and nucleotide on the growth performance and intestinal morphology of weaned piglets. J Anim Sci 80, Suppl. 1, 198.Google Scholar
120Jurgens, MH, Rikabi, RA & Zimmerman, DR (1997) The effect of dietary active dry yeast supplement on performance of sows during gestation–lactation and their pigs. J Anim Sci 75, 593597.CrossRefGoogle ScholarPubMed
121Moore, KL, D'Souza, DN & Mullan, BP (2007) Piglet growth after weaning is improved when a yeast extract is included in the diet of the lactating sow. In Manipulating Pig Production XI, pp. 61 [Paterson, JE and Barker, JA, editors]. Warribee: Australasian Pig Science Association, Inc.Google Scholar
122Gil, A & Sanchez-Medina, F (1982) Acid soluble nucleotides of human milk at different stages of lactation. J Dairy Res 49, 301307.Google Scholar
123Matsumoto, Y, Adjei, AA, Yamauchi, K, et al. . (1995) A mixture of nucleosides and nucleotides increases bone marrow cell and peripheral neutrophil number in mice infected with methicillin-resistant Staphylococcus aureus. J Nutr 125, 817822.Google Scholar
124Yamauchi, K, Adjei, AA, Ameho, CK, et al. . (1996) A nucleoside–nucleotide mixture and its components increase lymphoproliferative and delayed hypersensitivity responses in mice. J Nutr 126, 15711577.CrossRefGoogle ScholarPubMed
125Nagafuchi, S, Totsuka, M, Hachimura, S, et al. . (2000) Dietary nucleotides increase the proportion of a TCRγδ+ subset of intraepithelial lymphocytes (IEL) and IL-7 production by intestinal epithelial cells (IEC); implications for modification of cellular and molecular cross-talk between IEL and IEC by dietary nucleotides. Biosci Biotechnol Biochem 64, 14591465.Google Scholar
126Nagafuchi, S, Hachimura, S, Totsuka, M, et al. . (2000) Dietary nucleotides can up-regulate antigen-specific Th 1 immune responses and suppress antigen-specific IgE responses in mice. Int Arch Allergy Immunol 122, 3341.CrossRefGoogle Scholar
127Nagafuchi, S, Totsuka, M, Hachimura, S, et al. . (2002) Dietary nucleotides increase the mucosal IgA response and the secretion of transforming growth factor β from intestinal epithelial cells in mice. Cytotechnology 40, 4958.CrossRefGoogle ScholarPubMed
128Manzano, M, Gil, A & Rueda, R (2004) New insights in the potential mechanism of action of nucleotides to modulate immunity. Abstract: oral presentation. J Pediatr Gastroenterol Nutr 39, S32.Google Scholar
129Wang, L-F, Gong, X, Le, G-W, et al. . (2008) Dietary nucleotides protect thymocyte DNA from damage induced by cyclophosphamide in mice. J Anim Physiol Anim Nutr 92, 211218.Google Scholar
130Bontempo, V, Di Giancamillo, A, Savoini, G, et al. . (2006) Live yeast dietary supplementation acts upon intestinal morpho-functional aspects and growth in weanling piglets. Anim Feed Sci Technol 129, 224236.Google Scholar
131Martinez-Puig, D, Morales, J, Borda, E, et al. . (2007) Dietary nucleotides supplementation improves growth performance of early weaned piglets. J Anim Sci 85, Suppl. 1, 73.Google Scholar
Figure 0

Fig. 1 Digestion and absorption of nucleoproteins and their related products (adapted from Quan & Uauy(32)).

Figure 1

Table 1 Comparison of the nucleotide composition (μmol/100 ml) of colostrum and milk from different species (adapted from Gil & Sanchez-Medina(122))

Figure 2

Table 2 Concentration of total milk solids, crude protein (nitrogen×6·38) and 5′-monophosphate nucleotides in colostrum and milk from sows at different lactation stages (adapted from Mateo et al.(6))(Mean values with their pooled standard errors)

Figure 3

Table 3 Nucleotide concentration (mg/100 g, as is) in commonly used feed ingredients (adapted from Mateo & Stein(60))

Figure 4

Table 4 Effect of dietary nucleotides on immune functions in mice

Figure 5

Table 5 Zootechnical effects of nucleotide supplementation to diets of pigs compared with the control diet