Hostname: page-component-848d4c4894-xfwgj Total loading time: 0 Render date: 2024-06-16T20:39:45.026Z Has data issue: false hasContentIssue false

A rat model of mild intestinal inflammation induced by Staphylococcus aureus enterotoxin B

Published online by Cambridge University Press:  25 June 2010

Anna Pérez-Bosque*
Affiliation:
Grup de Fisiologia i Nutrició Experimental, Departament de Fisiologia, Facultat de Farmàcia, Institut de Recerca en Nutrició i Seguretat Alimentària, Universitat de Barcelona, Barcelona, Spain APC Europe, Granollers, Spain
Miquel Moretó
Affiliation:
Grup de Fisiologia i Nutrició Experimental, Departament de Fisiologia, Facultat de Farmàcia, Institut de Recerca en Nutrició i Seguretat Alimentària, Universitat de Barcelona, Barcelona, Spain
*
*Corresponding author: Dr Anna Pérez-Bosque, fax +34 934035901, email anna.perez@ub.edu
Rights & Permissions [Opens in a new window]

Abstract

The epithelial barrier of the intestine and the gut-associated lymphoid tissue (GALT) protects the host against luminal pathogenic micro-organisms. This is important at weaning, when animals are exposed to infectious agents and stresses. We have developed a rat model of intestinal inflammation post weaning, based on the systemic administration of Staphylococcus aureus enterotoxin B (SEB). Since the inflammatory response obtained is mild, the food intake pattern is not affected, which makes this model useful for studies of nutritional therapies for intestinal inflammatory disease. SEB increased T-lymphocytes in Peyer's patches and the number of activated T-lymphocytes in mesenteric lymph nodes (organized GALT). In the lamina propria, SEB increased activated T-lymphocytes as well as cytotoxic and natural killer-cell populations of the diffuse GALT. It also increased pro-inflammatory cytokines and inflammatory mediators in both Peyer's patches and mucosa. Rats given SEB had higher paracellular permeability to macromolecules, which was associated with a reduction in epithelial tightness. This model was used to examine whether dietary supplementation with spray-dried animal plasma proteins affects intestinal inflammation. Results showed that dietary plasma proteins can attenuate the mucosal immune response in both organized and diffuse GALT and that these effects are mediated by a reduction in the production of pro-inflammatory cytokines.

Type
3rd International Immunonutrition Workshop
Copyright
Copyright © The Authors 2010

Abbreviations:
GALT

gut-associated lymphoid tissue

IC

Ig fraction concentrate

PP

Peyer's patches

SDP

spray dried plasma

SEB

Staphylococcus aureus enterotoxin B

Intestinal physiology

The intestinal epithelium has two functions. First, it acts as a barrier to the passage of harmful intraluminal agents including foreign antigens, micro-organisms and their toxins. Second, it acts as a selective filter, allowing the absorption of essential dietary nutrients, electrolytes and water from the intestinal lumen to the circulation. The contact between neighbouring intestinal epithelial cells involves desmosomes, adherent junctions and tight junctions which regulate paracellular permeability. This is accomplished by interlocking proteins called claudins that bind scaffolding proteins, such as ZO-1, which in turn link them to the cellular cytoskeleton(Reference Turner1). An alteration of the intestinal barrier function contributes to disease, especially when the intestine is challenged by luminal antigens.

The intestine is a large surface in contact with the external environment. In a healthy intestine, mucosal immune cells must distinguish between beneficial antigens such as those present in food, innocuous antigens present in the commensal flora and potentially harmful antigens mainly involving pathogenic bacteria. The gut constitutes the largest lymphoid organ in the body. It contains a broad network of secondary lymphoid organs and a large number of lymphocytes including several intestine-specific subpopulations(Reference Mayer2). Upon activation, the intestinal immune system coordinates a strong inflammatory response against invasive pathogenic bacteria, while providing inhibitory mechanisms to prevent an excessive response against commensal bacteria. However, if the immune system is stimulated and the response is not controlled, the tissue may be damaged; there may be extracellular matrix destruction due to metalloproteinase release and increased pass of neutrophils and other immune cells across the epithelium due to increased epithelial permeability(Reference Mumy and McCormick3). In addition, stimulation of the immune system diverts energy and nutrients from growth and other productive functions(Reference Colditz4).

Gut-associated lymphoid tissue (GALT) accounts for up to 80% of the mucosal immune system and is distributed along the intestine in two forms: as organized GALT, which includes Peyer's patches (PP), isolated follicles and mesenteric lymph nodes, and as diffuse GALT, consisting of lymphocytes scattered in the epithelium and the lamina propria(Reference Granger, Kevil, Grisham, Johnson, Barrett, Merchant, Ghishan, Said and Wood5). Both compartments are part of a regulatory system with specific roles; organized GALT is the inductor site of the immune response and diffuse GALT is the effector site.

GALT has a role in both innate and acquired immune responses. Innate immunity is non-specific and acts as the first line of defence by preventing the entry of infectious agents or eliminating invading pathogens. It comprises physical barriers, such as skin or mucous membranes; cells in blood and tissues, such as phagocytes, neutrophils and monocytes; and soluble mediators, such as complement proteins, cytokines and defensins, which are antimicrobial peptides secreted by Paneth cells. While protection by innate immunity is effective, some pathogens escape detection or clearance by this system, which leads to the activation of the acquired immune system. This system consists of two major cell types, the T- and B-lymphocytes, which enable the specific recognition of and response to invaders. T-lymphocytes mediate mainly cellular acquired immunity while B-lymphocytes are involved in humoral acquired immunity(Reference Schenk and Mueller6).

Another component of the mucosal immune system consists of inducible regulatory T-cells, which maintain immunological unresponsiveness to self-antigens and suppress excessive immune responses that are deleterious to the host(Reference Sakaguchi, Yamaguchi and Nomura7). Regulatory T-cells mediate peripheral T-cell tolerance to antigens derived from the dietary origin or from the commensal flora.

Physiology of the gastrointestinal tract at weaning

In mammals, weaning is associated with intestinal maturation and changes in diet. Many changes in the intestinal physiology occur in the first 2 weeks after weaning(Reference Boundry, Péron and Le Huéron-Luron8), affecting villous and crypt development as well as the activity of many brush-border digestive enzymes(Reference Pluske, Hampson and Williams9). Weaning increases antigenic exposure, resulting in physiological inflammation mediated by the mucosal immune system. The number of mucosal mast cells and intraepithelial lymphocytes in the jejunum increases(Reference Bailey, Haverson and Miller10). In rodents, weaning is associated with changes in the activation status and numbers of leucocytes in the intestinal mucosa(Reference Manzano, Abadía-Molina and García-Olivares11).

The permeability of the immature intestine is higher than in adults and in weaning permeability depends on the integrity and stability of the tight junctions, as they control the diffusion of ions, water and macromolecules as well as luminal antigens. At weaning, dietary manipulation can affect the properties of the intestinal mucosa, such as permeability and nutrient absorption(Reference Pácha12). For example, young pigs are highly susceptible to enteric challenge, particularly as passive immunity declines. At weaning, the digestive system of pigs adapts to a dry diet. Consequently, weanling pigs often acquire enteric infections that may cause intestinal inflammation, villous atrophy and malabsorption, which contribute to high rates of mortality(Reference Van Dijk, Enthoven and Van den Hoven13).

Models of intestinal inflammation to study the role of drugs and diets

There are several models to study intestinal inflammation, most of them based on the administration of micro-organisms such as enteropathogenic Escherichia coli, Clostridium difficile or Cryptosporidium parvum. E. coli is the most abundant facultative anaerobe in the normal microflora of the mammalian colon(Reference Kaper, Nataro and Mobley14). In physiological conditions, the relationship between the luminal bacteria and the host is mutually beneficial; however, certain strains of E. coli (e.g. enteropathogenic E. coli) have acquired virulence and may contribute to the development of acute gastroenteritis(Reference Lapointe, O'Connor and Buret15). C. difficile has been implicated as the main cause of antibiotic-associated diarrhoea in adult human subjects and similar clinical conditions in a variety of other mammals(Reference Hurley and Nguyen16). C. parvum is a protozoan parasite that causes diarrhoea and gastroenteritis in human subjects and animals, which may become a chronic, life-threatening disease in immunocompromised patients(Reference Farthing17).

A common feature of the intestinal inflammation induced by these micro-organisms is the increased permeability of the intestinal epithelium, which is associated with secretory diarrhoea(Reference Toivola, Krishnan and Binder18). Toxins from Clostridium change the localization of several tight-junction proteins(Reference Chen, Pothoulakis and LaMont19) and enteropathogenic E. coli infection induces redistribution of occludin which decreases the barrier function(Reference Shifflett, Clayburgh and Koutsouris20). Moreover, enterotoxins can also induce the release of pro-inflammatory cytokines, such as interferon-γ and TNFα. Both cytokines increase the epithelial permeability by reducing the expression of zonula occludens-1 and occludin(Reference Bruewer, Luegering and Kucharzik21, Reference Tsukita, Furuse, Itoh and Berkeley22).

The S. aureus Enterotoxin B model

Rodent models are frequently used to analyse the immune mediated pathways of intestinal inflammation, and most of them are currently applied in studies of the pathophysiology of colitis(Reference Xavier and Podolsky23, Reference Uhlig and Powrie24). However, few of these models reproduce mild intestinal inflammation. Mild or transient inflammation can be induced by low doses of chemical agents used to induce colitis (e.g.: dextran sodium sulfate;Reference Vicario, Crespí and Franch25), parasite infections, such as the C. parvum model(Reference Laurent, McCole and Eckmann26) and administration of bacterial superantigens like Staphylococcus aureus enterotoxins(Reference Mumy and McCormick3).

Exposure to staphylococcal enterotoxins induces a range of clinical abnormalities from gastrointestinal upset to lethal toxic shock syndrome(Reference Van Gessel, Mani and Bi27). They can induce nausea, abdominal pain and diarrhoea in human subjects(Reference McKay28) and cause severe pathologies in farm animals(Reference Wilhelm, Rajic and Waddell29). In mice, intraperitoneal administration of S. aureus enterotoxin B (SEB) evokes a self-limiting enteropathy characterized by various degrees of histopathology, increased MHCII expression and increase in CD3+T cells(Reference Benjamin, Lu and Donnelly30).

Our objective is to study the functional properties of dietary supplements for use in nutritional therapy of intestinal inflammatory diseases, especially after weaning. At this stage, the gastrointestinal tract, still in transition to its adult characteristics, is exposed to a wide range of new bacterial populations and food antigens. We hypothesized that SEB would be a good candidate to reproduce a mild inflammatory condition in the laboratory, in young rats. The rats received an intraperitoneal injection of SEB (0·5 mg/kg) on days 30 and 33 after birth. This resulted in an intestinal inflammation syndrome, characterized by the activation of T helper lymphocytes and an increase in several of the cell populations involved in inflammation(Reference Pérez-Bosque, Pelegrí and Vicario31, Reference Pérez-Bosque, Miró and Polo32) (Fig. 1). SEB induced the recruitment of immune cells belonging to the innate immune system like neutrophils (31) and eosinophils(Reference Moretó and Pérez-Bosque33). It also increased the γδ-T lymphocyte population in PP, in lamina propria and in intraepithelial lymphocytes(Reference Pérez-Bosque, Miró and Polo32). The γδ-T cell subset modulates the inflammatory response by promoting the influx of lymphocytes and monocytes to mucosal surfaces(Reference Soltys and Quinn34).

Fig. 1. Characteristics of the Staphylococcus aureus enterotoxin B (SEB) model. SEB is a superantigen that binds to the T-cell receptor expressed by T-cells and to the MHC complex II, expressed by antigen presenting cells (APC) in an unrestricted fashion (outside of the binding groove). In the organized gut-associated lymphoid tissue (GALT), SEB promotes CD4 activation and recruitment of cytotoxic populations, such as γδ-T lymphocytes, Natural Killer (NK) cells and activated CD8 lymphocytes, in Peyer's patches (PP) and in mesenteric lymph nodes (MLN). SEB also activates the diffuse GALT. The components of the diffuse GALT are the lamina propria lymphocytes (LPL) and the intraepithelial lymphocytes (IEL). The enterotoxin increased the numbers of activated CD4 lymphocytes and stimulated the recruitment of cytotoxic populations and neutrophils. This activation of GALT provoked an increase in the release of pro-inflammatory mediators such as leukotriene B4 (LTB4) and cytokines like TNFα, interferon (IFN)γ and IL-6. The expression of inducible nitric oxide synthase (iNOS) was also increased. As a result of this immune activation there is an alteration in the expression of different proteins at the epithelial level. The expression of zonula occludens-1 (ZO-1) (tight junction) and β-catenin (adherent junction) is reduced, consistent with the observed increased mucosal permeability to dextran (4 kDa) and horseradish peroxidase (HRP, 40 kDa) and with an increased luminal water content. SEB also reduced the expression of the sodium-glucose transporter 1 (SGLT1) present in the apical membrane and the expression of mucosal defensins like cryptdin 4 (secreted by Paneth cells) and β-defensin 1 (secreted by enterocytes).

Administration of SEB also increased the transmural permeability of the small intestine, tested with tracers like dextran, with relatively low molecular weight (4 kDa) and horseradish peroxidase with a molecular weight similar to that of food antigens(Reference Pérez-Bosque, Amat and Polo35). These results indicate that the paracellular route can increase the protein flux across the intestine in animals challenged with the enterotoxin and they are consistent with the facilitated flux of antigen across the paracellular space observed by Toivola et al.(Reference Toivola, Krishnan and Binder18) SEB also reduced the expression of junctional proteins, such as ZO-1 and β-catenin, indicating a relation between the reduced epithelial tightness and the higher mucosal permeability(Reference Pérez-Bosque, Amat and Polo35) (Fig. 2). Furthermore, the increased luminal water content observed after SEB administration indicates that the effects of SEB on intestinal permeability can alter the absorptive/secretory water balance(Reference Pérez-Bosque, Pelegrí and Vicario31). SEB also reduced the expression of the apical sodium-glucose transporter 1 glucose and galactose transporter, especially at the villous apex where its expression is maximal, indicating that the inflammatory syndrome also impairs nutrient uptake(Reference Garriga, Pérez-Bosque and Amat36).

Fig. 2. Effects of Staphylococcus aureus enterotoxin B (SEB) on the epithelial barrier. (A,B): Immunolocalization of β-catenin, present at the adherens junctions (A) and ZO-1, present at the tight junctions (B) in control rats and in rats administered with SEB. The expression of both proteins was decreased by SEB, consistent with a reduction in epithelial tightness. (C) Shows the distribution of horseradish peroxidase in the intercellular space in Control and SEB treated rats. The mucosa of the jejunum was mounted in using chambers and incubated with horseradish peroxidase added to the mucosal side(Reference Pérez-Bosque, Amat and Polo35). The results are consistent with an increase in paracellular permeability in the rats challenged with SEB.

The obvious candidates to mediate the SEB effects are the mucosal pro-inflammatory cytokines. SEB stimulates lymphocytes to secrete interferon-γ and TNFα(Reference Huang and Koller37) and this is correlated with reduced tight-junction protein expression(Reference Bruewer, Luegering and Kucharzik21) and increased permeability(Reference McKay28). In our model, SEB administration also increased the release of TNFα and interferon-γ, IL-6 and leukotriene B4 in both mucosa and PP(Reference Pérez-Bosque, Miró and Polo38). In addition, the stimulation of mucosal GALT by SEB led the mucosal immune response to a T-helper 1 response, since IFN-γ and TNFα enhanced activation and proliferation of T-helper 1 cells(Reference Arad, Hillman and Levy39).

In human explants, SEB modulates the expression of both constitutive and inducible defensins(Reference Dhaliwal, Kelly and Bajaj-Elliott40). In the rat model, SEB reduced the expression of α-defensin cryptdin 4 and β-defensin 1(Reference Pérez-Bosque, Miró and Polo41). Since cryptdin 4 can block IL-1β release from lipopolysaccharide-activated monocytes(Reference Shi, Aono and Lu42), a decrease in its expression may increase intestinal IL-1β production. This would make the intestine more susceptible to SEB-induced damage and contribute to inflammatory bowel diseases(Reference Pérez-Bosque, Miró and Polo41).

The use of the S. aureus enterotoxin B model to study the properties of plasma supplements in the prevention of intestinal inflammation

Spray dried plasma (SDP) and its Ig fraction concentrate (IC) from either porcine or bovine origin are widely used to feed farm animals(Reference Quigley, Campbell and Polo43). They have also been used in human trials to reduce the clinical effects of cryptosporidiosis associated with AIDS(Reference Greenberg and Cello44). The protein composition of plasma is: approximately 50% albumin, 25% globulin (including α-, β- and γ-globulin), 5% fibrin and 20% other proteins, such as haptoglobulin, transferrin, growth factors and other proteins and peptides(Reference Anderson and Anderson45). There is evidence that Ig and other functional components present in SDP and IC are well conserved and are highly functional, providing immunological benefits(Reference Arthington, Cattell and Quigley46). For example, they reduce the intestinal inflammation produced by pathogenic bacteria and viruses(Reference Arthington, Jaynes and Tyler47) or by protozoa(Reference Hunt, Fu and Armstrong48).

The results of our studies on the effects of animal plasma preparations on intestinal inflammation indicate that these supplements can reduce, and in some cases prevent, the inflammatory syndrome induced by SEB. In summary, rats challenged with SEB fed dietary SDP and IC supplements show reduced activation of CD4 cells (i.e. T-helper lymphocytes) in the intestine (in PP, lamina propria and intraepithelial compartments) and a lower increase in the γδ-T-lymphocytes population in PP and in lamina propria. These data support the hypothesis that diets supplemented with animal plasma can attenuate the immune response(Reference Pérez-Bosque, Pelegrí and Vicario31, Reference Pérez-Bosque, Miró and Polo32). In pigs challenged with E. coli K88, Bosi et al.(Reference Bosi, Casini and Finamore49) observed that the expression of pro-inflammatory cytokines was lower in animals fed SDP. Results from our laboratory indicate that plasma supplements reduce the expression of mucosal pro-inflammatory cytokines(Reference Moretó, Miró and Polo50, Reference Pérez-Bosque, Miró and Polo38) and the activation of T-helper subsets in the lamina propria and in the epithelium(Reference Pérez-Bosque, Miró and Polo32). These effects on mucosal cytokine profile can explain the reduction of mucosal inflammation by plasma supplements and the preventive effects of SDP and IC on changes in the mucosal permeability(Reference Pérez-Bosque, Pelegrí and Vicario31), tight-junctional protein expression(Reference Pérez-Bosque, Amat and Polo35) and nutrient absorption(Reference Garriga, Pérez-Bosque and Amat36) following SEB administration. This reduction of the toxin-induced increase in mucosal permeability may prevent the passage of microbial and food antigens to the interstitial space, thereby blocking local inflammation(Reference Santos, Yang and Soderholm51). Recent results, summarized in Fig. 3, show that both SDP and IC reduce the SEB-induced release of pro-inflammatory cytokines, while increasing IL-10 concentration in GALT as well as at the systemic level. Interestingly, SDP can increase mucosal IL-10 concentration either in the presence or absence of an SEB challenge(Reference Pérez-Bosque, Miró and Polo38).

Fig. 3. Cytokine release in the Staphylococcus aureus enterotoxin B (SEB) model. Release of pro-inflammatory TNFα and anti-inflammatory IL-10 in intestinal mucosa (A), peyer's patches (B) and serum (C). Experimental groups were: Control group; rats administered with SEB (SEB group) and rats challenged with SEB and supplemented with spray-dried plasma (SEB-SDP group) or with Ig concentrate (SEB-IC group). The statistical analysis (one-way ANOVA followed by Bonferroni post-hoc test) showed that the SEB group has a higher TNFα concentration in the three tissues than controls and that both dietary supplements can reduce the expression of TNFα and stimulate the release of IL-10(Reference Pérez-Bosque, Miró and Polo38).

There are several mechanisms by which SDP and IC can modulate the intestinal immune response. Luminal effects were suggested by van Dijk et al.(Reference van Dijk, Niewold and Margry52) who reported a reduction in mucosal binding of luminal antigens by plasma glycoproteins. Moreover, Ig present in SDP may bind to potential antigens in the lumen of the small intestine and prevent their attachment to the mucosa(Reference Bosi, Casini and Finamore49). These Ig may also cause changes in intestinal microbiota, increasing the richness of the ecosystem, as suggested by Martin-Orúe et al.(Reference Martin-Orúe, Pérez-Bosque and Gómez de Segura53). The full plasma supplement is a complex mixture of growth factors, cytokines and biologically active compounds. Therefore, a role for these proteins interacting with immune cells present in the mucosa, thus changing the mucosal cytokine profile, should also be considered.

In conclusion, the rat model based on the systemic administration of SEB induces a mild inflammatory syndrome, which does not affect the animal's welfare or feeding behaviour. These characteristics make this model appropriate for studies on the use of nutrition therapy in inflammatory diseases. It has contributed to the discovery of the anti-inflammatory properties of SDP supplements.

Acknowledgements

We thank J. Campbell, J. Crenshaw, J. Polo, L. Russell and E. Weaver for their comments and suggestions. This study was supported by the Eureka Program Euroagri (E!2452) and by funds from APC Inc., IA and APC Europe. Data presented within and figures of this paper have been recreated with permission from the primary authors. M.M. declares no conflicts of interest; A.P.-B. is employed part-time by APC-Europe supported by a Beatriu de Pinós grant (Generalitat de Catalunya, Spain). A.P.-B. and M.M. analysed the data and wrote the paper. Both authors have read and approved the final manuscript.

References

1.Turner, JR (2009) Intestinal mucosal barrier function in health and disease. Nat Rev Immunol 9, 799809.Google Scholar
2.Mayer, L (2005) Mucosal immunity. Immunol Rev 206, 5.CrossRefGoogle ScholarPubMed
3.Mumy, KL & McCormick, BA (2009) The role of neutrophils in the event of intestinal inflammation. Curr Opin Pharmacol 9, 697701.CrossRefGoogle ScholarPubMed
4.Colditz, IG (2008) Six costs of immunity to gastrointestinal nematode infections. Parasite Immunol 30, 6370.CrossRefGoogle ScholarPubMed
5.Granger, N, Kevil, CG & Grisham, MB (2006) Recruitment of inflammatory and immune cells in the gut: physiology and pathophysiology. In Physiology of the gastrointestinal tract, pp. 11371162 [Johnson, LR, Barrett, KE, Merchant, JL, Ghishan, FK, Said, HM and Wood, JD, editors]. Amsterdam: Elsevier Academic Press.Google Scholar
6.Schenk, M & Mueller, C (2008) The mucosal immune system at the gastrointestinal barrier. Best Pract Res Clin Gastroenterol 22, 391409.Google Scholar
7.Sakaguchi, S, Yamaguchi, T, Nomura, T et al. (2008) Regulatory T cells and immune tolerance. Cell 133, 775787.CrossRefGoogle ScholarPubMed
8.Boundry, G, Péron, V, Le Huéron-Luron, I et al. (2004) Weaning induces both transient and long-lasting modifications of absorptive, secretory, and barrier properties of piglet intestine. J Nutr 134, 22562262.CrossRefGoogle Scholar
9.Pluske, JR, Hampson, DJ & Williams, IH (1997) Factors influencing the structure and function of the small intestine in the weaned pigs: a review. Livest Prod Sci 51, 215236.CrossRefGoogle Scholar
10.Bailey, M, Haverson, K, Miller, B et al. (2004) Effects of infection with transmissible gastroenteritis virus on concomitant immune responses to dietary and injected antigens. Clin Diagn Lab Immunol 11, 337343.Google ScholarPubMed
11.Manzano, M, Abadía-Molina, AC, García-Olivares, E et al. (2002) Absolute counts and distribution of lymphocyte subsets in small intestine of BALB/c mice change during weaning. J Nutr 132, 27572762.CrossRefGoogle ScholarPubMed
12.Pácha, J (2000) Development of intestinal transport functions in mammals. Physiol Rev 80, 16331667.Google Scholar
13.Van Dijk, AJ, Enthoven, PM, Van den Hoven, SG et al. (2002) The effect of dietary spray-dried porcine plasma on clinical response in weaned piglets challenged with a pathogenic Escherichia coli. Vet Microbiol 84, 207218.CrossRefGoogle ScholarPubMed
14.Kaper, JB, Nataro, JP & Mobley, HL (2004) Pathogenic Escherichia coli. Nat Rev Microbiol 2, 123140.CrossRefGoogle ScholarPubMed
15.Lapointe, TK, O'Connor, PM & Buret, AG (2009) The role of epithelial malfunction in the pathogenesis of enteropathogenic E. coli-induced diarrhea. Lab Invest 89, 964970.CrossRefGoogle ScholarPubMed
16.Hurley, BW & Nguyen, CC (2002) The spectrum of pseudomembranous enterocolitis and antibiotic-associated diarrhea. Arch Intern Med 162, 21772184.Google Scholar
17.Farthing, MJ (2000) Clinical aspects of human cryptosporidiosis. Contrib Microbiol 6, 5074.Google Scholar
18.Toivola, DM, Krishnan, S, Binder, HJ et al. (2004) Keratins modulate colonocyte electrolyte transport via protein mistargeting. J Cell Biol 164, 911921.Google Scholar
19.Chen, ML, Pothoulakis, C & LaMont, JT (2002) Protein kinase C signaling regulates ZO-1 translocation and increased paracellular flux of T84 colonocytes exposed to Clostridium difficile toxin A. J Biol Chem 277, 42474254.CrossRefGoogle ScholarPubMed
20.Shifflett, DE, Clayburgh, DR, Koutsouris, A et al. (2005) Enteropathogenic E. coli disrupts tight junction barrier function and structure in vivo. Lab Invest 85, 13081324.CrossRefGoogle ScholarPubMed
21.Bruewer, M, Luegering, A, Kucharzik, T et al. (2003) Pro-inflammatory cytokines disrupt epithelial barrier function by apoptosis-independent mechanisms. J Immunol 171, 61646172.CrossRefGoogle Scholar
22.Tsukita, S, Furuse, M & Itoh, M (2002) The tight junction. In Cell adhesion, pp. 369395 [Berkeley, MC editor]. New York: Oxford University Press.Google Scholar
23.Xavier, RJ & Podolsky, DK (2007) Unravelling the pathogenesis of inflammatory bowel disease. Nature 448, 427434.Google Scholar
24.Uhlig, HH & Powrie, F (2009) Mouse models of intestinal inflammation as tools to understand the pathogenesis of inflammatory bowel disease. Eur J Immunol 39, 20212026.Google Scholar
25.Vicario, M, Crespí, M, Franch, A et al. (2005) Induction of colitis in young rats by dextran sulfate sodium. Dig Dis Sci 50, 143150.Google Scholar
26.Laurent, F, McCole, D, Eckmann, L et al. (1999) Pathogenesis of Cryptosporidium parvum infection. Microbes Infect 1, 141148.Google Scholar
27.Van Gessel, YA, Mani, S, Bi, S et al. (2004) Functional piglet model for the clinical syndrome and post-mortem findings induced by sathylococcal enterotoxin B. Exp Biol Med 229, 10611071.CrossRefGoogle Scholar
28.McKay, DM (2001) Bacterial superantigens: provocateurs of gut dysfunction and inflammation? Trends Immunol 22, 497501.CrossRefGoogle ScholarPubMed
29.Wilhelm, B, Rajic, A, Waddell, L et al. (2009) Prevalence of zoonotic or potentially zoonotic bacteria, antimicrobial resistance, and somatic cell counts in organic dairy production: current knowledge and research gaps. Foodborne Pathog Dis 6, 525539.CrossRefGoogle ScholarPubMed
30.Benjamin, MA, Lu, J, Donnelly, G et al. (1998) Changes in murine jejunal morphology evoked by the bacterial superantigen Staphylococcus aureus enterotoxin B are mediated by CD4+T cells. Infect Immun 66, 21932199.Google Scholar
31.Pérez-Bosque, A, Pelegrí, C, Vicario, M et al. (2004) Dietary plasma protein affects the immune response of weaned rats challenged with S. aureus superantigen B. J Nutr 134, 26672672.Google Scholar
32.Pérez-Bosque, A, Miró, L, Polo, J et al. . (2008) Dietary plasma proteins modulate the immune response of diffuse gut-associated lymphoid tissue in rats challenged with Staphylococcus aureus enterotoxin B. J Nutr 138, 533537.Google Scholar
33.Moretó, M & Pérez-Bosque, A (2009) Dietary plasma proteins, the intestinal immune system, and the barrier functions of the intestinal mucosa. J Anim Sci 87, E92–E100.CrossRefGoogle ScholarPubMed
34.Soltys, J & Quinn, MT (1999) Selective recruitment of T cell subsets to the udder during staphylococcal and streptococcal mastitis: analysis of lymphocyte subsets and adhesion molecule expression. Infect Immun 67, 62936302.Google Scholar
35.Pérez-Bosque, A, Amat, C, Polo, J et al. (2006) Spray-dried animal plasma prevents the effects of Staphylococcus aureus Enterotoxin B on intestinal barrier function in weaned rats. J Nutr 136, 28382843.CrossRefGoogle ScholarPubMed
36.Garriga, C, Pérez-Bosque, A, Amat, C et al. . (2005) Spray-dried porcine plasma reduces the effects of staphylococcal enterotoxin B on glucose transport in rat intestine. J Nutr 135, 16531658.Google Scholar
37.Huang, W & Koller, LD (1998) Superantigen activation and kinetics of cytokines in the Long-Evans rat. Immunology 95, 331338.CrossRefGoogle ScholarPubMed
38.Pérez-Bosque, A, Miró, LL, Polo, J et al. (2010) Dietary plasma protein supplements prevent the release of mucosal pro-inflammatory mediators in intestinal inflammation in rats. J Nutr 140, 2530.Google Scholar
39.Arad, G, Hillman, D, Levy, R et al. (2001) Superantigen antagonist blocks Th1 cytokine gene induction and lethal shock. J Leukoc Biol 69, 921927.Google Scholar
40.Dhaliwal, W, Kelly, P & Bajaj-Elliott, M (2009) Differential effects of Staphylococcal enterotoxin B-mediated immune activation on intestinal defensins. Clin Exp Immunol 156, 263270.CrossRefGoogle ScholarPubMed
41.Pérez-Bosque, , Miró, LL, Polo, J et al. (2010) Oral plasma proteins attenuate gut inflammatory effects induced by S. aureus enterotoxin B challenge in rats. Livest Sci, In the Press.Google Scholar
42.Shi, J, Aono, S, Lu, W et al. (2007) A novel role for defensins in intestinal homeostasis: regulation of IL-1β secretion. J Immunol 179, 12451253.Google Scholar
43.Quigley, JD 3rd, Campbell, JM, Polo, J et al. (2004) Effects of spray-dried animal plasma on intake and apparent digestibility in dogs. J Anim Sci 82, 16851692.Google Scholar
44.Greenberg, PD & Cello, JP (1996) Treatment of severe diarrhea caused by Cryptosporidium parvum with oral bovine immunoglobulin concentrate in patients with AIDS. J Acquir Immune Defic Syndr Hum Retrovirol 13, 348354.Google Scholar
45.Anderson, NL & Anderson, NG (2002) The human plasma proteome. History, character, and diagnostic prospects. Mol Cell Proteomics 1, 845867.Google Scholar
46.Arthington, JD, Cattell, MB, Quigley, JD 3rd et al. (2000) Passive immunoglobin transfer in newborn calves fed colostrum or spray-dried serum protein alone or as a supplement to colostrum of varying quality. J Dairy Sci 83, 28342838.Google Scholar
47.Arthington, JD, Jaynes, CA, Tyler, HD et al. (2002) The use of bovine serum protein as an oral support therapy following coronavirus challenge in calves. J Dairy Sci 85, 12491254.CrossRefGoogle ScholarPubMed
48.Hunt, E, Fu, Q, Armstrong, MU et al. (2002) Oral bovine serum concentrate improves cryptosporidial enteritis in calves. Pediatr Res 51, 370376.Google Scholar
49.Bosi, P, Casini, L, Finamore, A et al. (2004) Spray-dried plasma improves growth performance and reduces inflammatory status of weaned pigs challenged with enterotoxigenic Escherichia coli K88. J Anim Sci 82, 17641772.Google Scholar
50.Moretó, M, Miró, LL, Polo, J et al. (2008) Oral porcine plasma proteins prevent the release of mucosal pro-inflammatory cytokines in rats challenged with S. aureus enterotoxin B. Gastroenterology 134, A-524 [abs.]Google Scholar
51.Santos, J, Yang, PC, Soderholm, JD et al. (2001). Role of mast cells in chronic stress induced colonic epithelial barrier dysfunction in the rat. Gut 48, 630636.CrossRefGoogle ScholarPubMed
52.van Dijk, AJ, Niewold, TA, Margry, RJ et al. (2001) Small intestinal morphology in weaned piglets fed a diet containing spray-dried porcine plasma. Res Vet Sci 71, 1722.CrossRefGoogle ScholarPubMed
53.Martin-Orúe, SM, Pérez-Bosque, A, Gómez de Segura, A, et al. (2008) Feed added sprayed dried porcine plasma (SDPP) modifies cecal microbiota in rats. Gut Microbiome Meeting, Clermont-Ferrand, France, 2008.Google Scholar
Figure 0

Fig. 1. Characteristics of the Staphylococcus aureus enterotoxin B (SEB) model. SEB is a superantigen that binds to the T-cell receptor expressed by T-cells and to the MHC complex II, expressed by antigen presenting cells (APC) in an unrestricted fashion (outside of the binding groove). In the organized gut-associated lymphoid tissue (GALT), SEB promotes CD4 activation and recruitment of cytotoxic populations, such as γδ-T lymphocytes, Natural Killer (NK) cells and activated CD8 lymphocytes, in Peyer's patches (PP) and in mesenteric lymph nodes (MLN). SEB also activates the diffuse GALT. The components of the diffuse GALT are the lamina propria lymphocytes (LPL) and the intraepithelial lymphocytes (IEL). The enterotoxin increased the numbers of activated CD4 lymphocytes and stimulated the recruitment of cytotoxic populations and neutrophils. This activation of GALT provoked an increase in the release of pro-inflammatory mediators such as leukotriene B4 (LTB4) and cytokines like TNFα, interferon (IFN)γ and IL-6. The expression of inducible nitric oxide synthase (iNOS) was also increased. As a result of this immune activation there is an alteration in the expression of different proteins at the epithelial level. The expression of zonula occludens-1 (ZO-1) (tight junction) and β-catenin (adherent junction) is reduced, consistent with the observed increased mucosal permeability to dextran (4 kDa) and horseradish peroxidase (HRP, 40 kDa) and with an increased luminal water content. SEB also reduced the expression of the sodium-glucose transporter 1 (SGLT1) present in the apical membrane and the expression of mucosal defensins like cryptdin 4 (secreted by Paneth cells) and β-defensin 1 (secreted by enterocytes).

Figure 1

Fig. 2. Effects of Staphylococcus aureus enterotoxin B (SEB) on the epithelial barrier. (A,B): Immunolocalization of β-catenin, present at the adherens junctions (A) and ZO-1, present at the tight junctions (B) in control rats and in rats administered with SEB. The expression of both proteins was decreased by SEB, consistent with a reduction in epithelial tightness. (C) Shows the distribution of horseradish peroxidase in the intercellular space in Control and SEB treated rats. The mucosa of the jejunum was mounted in using chambers and incubated with horseradish peroxidase added to the mucosal side(35). The results are consistent with an increase in paracellular permeability in the rats challenged with SEB.

Figure 2

Fig. 3. Cytokine release in the Staphylococcus aureus enterotoxin B (SEB) model. Release of pro-inflammatory TNFα and anti-inflammatory IL-10 in intestinal mucosa (A), peyer's patches (B) and serum (C). Experimental groups were: Control group; rats administered with SEB (SEB group) and rats challenged with SEB and supplemented with spray-dried plasma (SEB-SDP group) or with Ig concentrate (SEB-IC group). The statistical analysis (one-way ANOVA followed by Bonferroni post-hoc test) showed that the SEB group has a higher TNFα concentration in the three tissues than controls and that both dietary supplements can reduce the expression of TNFα and stimulate the release of IL-10(38).