+ All Categories
Home > Documents > Dietary immune-modulation – carbohydrate specific effects ...

Dietary immune-modulation – carbohydrate specific effects ...

Date post: 28-Feb-2022
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
98
Dietary immune-modulation – carbohydrate specific effects of cereal and yeast -glucans Philosophiae Doctor (PhD) Thesis Anne Rieder 2013 Department of Nutrition Institute of Basic Medical Sciences Faculty of Medicine University of Oslo Nofima, Norwegian Institute of Food, Fisheries and Aquaculture Research
Transcript
Page 1: Dietary immune-modulation – carbohydrate specific effects ...

Dietary immune-modulation – carbohydrate specific effects of cereal and yeast �-glucans

Philosophiae Doctor (PhD) Thesis

Anne Rieder

2013

Department of Nutrition

Institute of Basic Medical Sciences

Faculty of Medicine

University of Oslo

Nofima, Norwegian Institute of

Food, Fisheries and Aquaculture

Research

Page 2: Dietary immune-modulation – carbohydrate specific effects ...

© Anne Rieder, 2013 Series of dissertations submitted to the Faculty of Medicine, University of Oslo No. 1521 ISBN 978-82-8264-525-6 All rights reserved. No part of this publication may be reproduced or transmitted, in any form or by any means, without permission. Cover: Inger Sandved Anfinsen. Printed in Norway: AIT Oslo AS. Produced in co-operation with Akademika publishing. The thesis is produced by Akademika publishing merely in connection with the thesis defence. Kindly direct all inquiries regarding the thesis to the copyright holder or the unit which grants the doctorate.

Page 3: Dietary immune-modulation – carbohydrate specific effects ...

1

Acknowledgements

The work presented in this thesis was carried out at Nofima, the Norwegian Institute of Food, Fisheries and Aquaculture Research in Ås. The financial support was provided by the Fund for the Research Levy on Agricultural Products.

I would like to thank my supervisors at Nofima, Svein Halvor Knutsen and Stine Grimmer for their great support. You have been fantastic and I have learned so much from you in the last years! I could fill pages (you know I like looong paragraphs and loooong sentences) with praise, but will restrict myself to saying thanks for making me a part of the research team at Nofima.

I would also like to thank my supervisor at the University of Oslo, Svein Olav Kolset, for great scientific discussions, good ideas, the opportunities to present our work for his research group and of course also for the charming end-of-term celebrations at his house. Even before I started my PhD studies, I was trying to sing Norwegian Christmas songs at your house!

I want to thank all my co-authors for their contribution and expertise that greatly improved the quality of the presented scientific papers. A special thanks to Anne Berit Samuelsen at the Department of Pharmaceutical Chemistry at the University of Oslo for our joint project of immerging deeply into the world of cereal �-glucans. I really enjoyed working with you! And of course I would like to thank Diego for all the Saturdays we spent with the Fluorescence instrument until our method finally worked. I learned a lot form you and I miss our bike trips to Drøbak and your addiction to the day light lamp.

Merete, Signe, Elin, Linda, Helle, Hanne, Marte, Janne and Stine, I would like to thank you for sharing your knowledge of cell line cultures and all cell related test systems at Nofima with me. It was good to have someone to share the trouble with the “wee” cells with.

I want to thank all my colleagues at the dietary fiber research group for fruitful scientific discussions, a great working environment and all the cakes from the bakery. A special thanks to Ann Katrin for always being there for me at work and after work!

Thanks to my fellow PhD students and friends at Nofima and FODOS for all our social events and sharing our PhD frustration with each other. I would like to thank my office made Anastasia for nice conversations and friendly company. I thank Ulrike for her friendship and sharing the complete language confusion with me. Thanks also to Jib, Elena, Silje, Gunna, Natasa, Trygve, Johannes, Nebojsa, Håkon and Sarin for their company and friendship, all ski trips, cabin trips and day light lamp parties.

I want to thank my family and friends in Germany for their support and their frequent visits to Norway. What would I have done without all the phone calls complaining about the weather? �

I thank Nils for all our scientific discussions at the dinner table and especially for repeating “det kommer til å gå så bra” so many times that even I finally believed it. Let`s hope it`s true….min løk �

Ås, January 2013 Anne Rieder

Page 4: Dietary immune-modulation – carbohydrate specific effects ...

2

Page 5: Dietary immune-modulation – carbohydrate specific effects ...

3

Table of Contents Abbreviations ............................................................................................................................. 5

Explanation of key expressions .................................................................................................. 7

1. Introduction ............................................................................................................................ 9

2. The immune system ............................................................................................................. 10

2.1 Innate and adaptive immune responses .......................................................................... 10

2.2 Cytokines ........................................................................................................................ 13

2.2.1 IL-8 (CXCL-8) ......................................................................................................... 13

2.2.2 TNF-� ....................................................................................................................... 14

2.2.3 IL-1� ........................................................................................................................ 15

2.3 The gut immune system .................................................................................................. 16

3. �-glucan ................................................................................................................................ 18

3.1 �-glucan structure and structure-function relationships ................................................. 18

3.2 �-glucan receptors ........................................................................................................... 23

3.3 Possible mechanisms behind immune modulation by dietary �-glucans ....................... 24

4. LPS ....................................................................................................................................... 27

5. Aims of the study ................................................................................................................. 31

6. List of papers and manuscripts ............................................................................................. 32

7. Results .................................................................................................................................. 33

7.1 Review of cereal �-glucans immune-modulating properties (Paper I) ........................... 33

7.2 Effect of high MW �-glucan fractions and arabinoxylan fractions isolated from barley on IL-8 secretion by the intestinal epithelial cell lines Caco-2 and HT-29, NF-�B activity in monocytic U937 cells and complement fixation (Paper II) .................................................. 33

7.3 Effect of cereal �-glucan fractions with different weight average MW (Mw) on the cytokine secretion by intestinal epithelial cell lines Caco-2 and HT-29 (Paper III) ............ 34

7.4 Development of a new method to measure low concentrations of cereal �-glucans in cell culture supernatants and application to analysis of cereal �-glucan transport over intact Caco-2 cell monolayers (Paper IV) ...................................................................................... 35

7.5 Tools to study the carbohydrate specific effect of cereal and yeast �-glucan preparations (Paper V) ............................................................................................................................... 37

7.6 Effect of cereal and yeast �-glucan preparations on THP-1 derived human macrophages, differentiated Caco-2 cells and Caco-2 macrophage co-cultures (Paper VI) ....................... 38

8. Discussion ............................................................................................................................ 39

8.1 Interaction with IEC ....................................................................................................... 39

8.2 Uptake ............................................................................................................................. 41

Page 6: Dietary immune-modulation – carbohydrate specific effects ...

4

8.3 Macrophages ................................................................................................................... 45

8.4 Carbohydrate specific effects ......................................................................................... 48

8.5 Beta-glucans structure-function relationship .................................................................. 52

8.6 Current knowledge of dietary �-glucans mechanisms of action ..................................... 53

9. Concluding Remarks ............................................................................................................ 56

10. Reference List .................................................................................................................... 58

Page 7: Dietary immune-modulation – carbohydrate specific effects ...

5

Abbreviations

AP-1 activator protein 1

CR3 complement receptor 3

DB degree of branching

DC dendritic cell

DP degree of polymerization

ELK-1 Eph-related tyrosine kinase

GI-tract gastrointestinal tract

IEC intestinal epithelial cell

IEL intraepithelial lymphocyte

Ig immunoglobulin

IL interleukin

LAL limulus amoebocyte lysate

LBP lipopolysaccharide binding protein

LPS lipopolysaccharide

MBL mannose binding lectin

M cell microfold cell

MW molecular weight

Mw weight average molecular weight

MyD88 myeloid differentiation primary response gene 88

NF-�B nuclear factor kappa-light-chain-enhancer of activated B cells

NK cells natural killer cells

NMR Nuclear magnetic resonance

PAMP pathogen associated molecular pattern

PBMC peripheral blood mononuclear cells

PRR pattern recognition receptor

Page 8: Dietary immune-modulation – carbohydrate specific effects ...

6

ROS reactive oxygen species

SCFA short chain fatty acids

TEER trans-epithelial electrical resistance

TNF-� tumor necrosis factor alpha

TLR toll like receptor

Page 9: Dietary immune-modulation – carbohydrate specific effects ...

7

Explanation of key expressions

Dietary �-glucans

The expression dietary �-glucans is used in this thesis for all �-glucans that are consumed as a

part of the diet. This can be �-glucans that naturally occur in the human diet like cereal �-

glucans or �-glucans from baker's yeast or it can be �-glucans that are added to foods during

manufacturing as a functional food ingredient. In animal studies dietary �-glucans is used for

orally applied �-glucans.

Targeted enzymatic degradation

Targeted enzymatic degradation describes a technique that was developed as a part of this

thesis and can be used to determine the carbohydrate specific effect of e.g. a �-glucan

preparation. The technique is based on the degradation of the component of interest (here �-

glucans) by specific enzymes. Comparison of the activity of the degraded preparation with the

activity elicited by the un-degraded preparation makes it possible to draw conclusion about

the specific activity of the components of interest. In the case of �-glucans the complete

degradation of �-glucans in the test samples should result in the complete abrogation of

observed effects if the activity is actually mediated by the �-glucan molecules.

Immune competent cells

In this thesis the term immune competent cell is used for any cell type that may play a role in

immune responses. This includes also cells that are not part of the immune system like

intestinal epithelial cells since they play an important role in orchestrating gut immune

responses.

Page 10: Dietary immune-modulation – carbohydrate specific effects ...

8

Page 11: Dietary immune-modulation – carbohydrate specific effects ...

9

1. Introduction

The consumption of dietary fiber is generally regarded as important for the maintenance of

human health. Dietary recommendations promote an increase in fiber consumption. A high

intake of dietary fiber is regarded as an important part of primary prevention of major diseases

such as type II diabetes, cardiovascular disease and several forms of cancer [1]. Since the

concept of dietary fiber was introduced in the 1970s, it`s definition has been constantly under

debate. In 2009, the Codex Alimentarius Commission adopted an internationally accepted

regulatory definition for dietary fiber: Carbohydrate polymers that are resistant to digestive

enzymes in the small intestine of humans [2]. These can be either naturally occurring in food

as consumed, obtained from food raw material or be synthetic carbohydrate polymers [2]. The

latter two groups need to have demonstrated physiological benefits to human health in order

to be defined as dietary fiber [2]. The classic documented physiological benefits of dietary

fiber consumption include increased laxation, decreased risk of cardiovascular disease and

type II diabetes and positive effects on weight management [3, 4]. Furthermore, a high fiber

intake, especially cereal fiber and whole grain products, has been linked to a reduced risk of

colorectal cancer [5]. This protective effect may be due to decreased intestinal transit time,

decreasing the exposure time to environmental or food carcinogens, and colonic fermentation

of fiber leading to butyrate production, which has a beneficial effect on colonocyte cell cycle

regulation [6, 7]. In recent years, dietary fiber has also been linked to immune-modulation [8].

Either as a part of the prebiotic effects or due to structure mediated direct interaction with

immune competent cells [9-11].

�eta-glucans are major cell wall components in some cereal grains and fungi, and form a

heterogeneous group of indigestible carbohydrates with possible immune-modulating

properties. Furthermore, cereal �-glucans are known for their ability to decrease blood

cholesterol levels, a risk factor for cardiovascular disease, mainly by increasing the viscosity

Page 12: Dietary immune-modulation – carbohydrate specific effects ...

10

in the gastrointestinal (GI) tract [12, 13], and the European Food Safety Authority has

recently approved a health claim on oat �-glucan's ability to reduce blood cholesterol [14].

�eta-glucans from different sources have shown potential for direct interaction with immune-

competent cells [15]. Potential benefits of dietary immune stimulation with �-glucans include

increased resistance to infection [9, 16], therapeutic effects in treatment of various cancers

[17-21] and dampening of allergic disease [22]. Research on the immune-modulating

properties of �-glucans has been focused on �-glucans from fungi. However, also cereal

derived �-glucans have shown beneficial effects both in vitro and in animal studies [23].

Despite the identification of several �-glucan receptors, the precise mechanisms of action of

the immune-modulating properties of dietary �-glucans (cereal or fungal) are still far from

understood. The aim of this thesis was therefore to increase the current understanding of how

dietary �-glucans may modulate the human immune system by focusing on the first step from

the gut lumen to the activation of effector cells. Since cereal �-glucans are a natural part of the

human diet they received the main focus, but also �-glucans from yeast were investigated for

comparison purposes.

2. The immune system

The immune system is very complex, and thus this paragraph gives a brief overview of the

most important modes of action relevant for studies on �-glucans.

2.1 Innate and adaptive immune responses

Beta-glucans have been shown to modulate both innate and adaptive immune responses [24,

25]. The innate immune system is the evolutionary older part of the immune system that can

react fast to a broad range of pathogens. The adaptive immune system on the other hand

Page 13: Dietary immune-modulation – carbohydrate specific effects ...

11

creates highly specific, very powerful responses to particular pathogens that often need some

days to fully develop.

Immune responses in the innate immune system are initiated by binding of so called pathogen

associated molecular patterns (PAMP) on pathogens to pattern recognition receptors (PRR)

such as toll like receptors (TLR). PAMPs are molecular structures that are shared by different

pathogen groups such as lipopolysaccharides (LPS) from gram negative bacteria (interacting

with TLR-4), or peptidoglycan from gram positive bacteria (binding to TLR-2). Also �-

glucans are PAMPs, even though only a small part of this group derives from pathogens such

as the opportunistic pathogenic yeast Candida albicans. PRR signaling may activate

phagocytosis or lead to the secretion of cytokines that recruit or activate other effector cells.

Figure 1 illustrates the effector cells of the innate and adaptive immune system. Besides

effector cells such as neutrophil, macrophages and NK cells, the innate immune system also

comprises a series of proteins that activate one another, called the complement system.

Opsonization, recruitment of phagocytes and assembly of the membrane attack complex are

the main effector functions of the complement system. The complement cascade can be

activated by three different pathways. The classical pathway is activated by antigen:antibody

complexes. The lectin pathway is initiated by the binding of carbohydrate binding proteins

such as mannose binding lectin (MBL) to specific carbohydrates on pathogen surfaces. The

alternative pathway is activated by the direct binding of activated complement component 3 to

the pathogen surface.

Page 14: Dietary immune-modulation – carbohydrate specific effects ...

12

Figure 1: Cells of the innate and adaptive immune system from [26]

The effector cells of the adaptive immune system are highly specific towards one specific

antigen and have to be activated by an antigen presenting cell that has encountered their

specific antigen in order to be able to exert their effector functions. The effector cells of the

adaptive immune system can be divided into T-cells, exerting cell mediated immunity, and B-

cells, producing antibody for humoral immunity. Beta-glucans have been shown to play a role

in T-cell activation and T-cell differentiation into different effector cell subsets [21]. T-cells

can be divided into CD8+ T-cells, which have cytotoxic effector functions and CD4+ T-cells,

whose effector function is to activate other cells like macrophages or B-cells. CD4+ T-cells

are therefore also called helper T-cells (TH). TH cells can develop into different subsets with

distinct functions depending on the cytokine environment. TH1 cells for example secret

macrophage activating molecules, while TH2 cells activate B-cells. TH17 cells are mainly

known for their ability to recruit neutrophils and are often found in the gut. Regulatory T-cells

(Treg) secrete suppressive cytokines such as TGF-� and IL-10 and play an important role in

Page 15: Dietary immune-modulation – carbohydrate specific effects ...

13

maintaining homeostasis e.g. in the gut. B-cells need T-cell help for their activation and the

cytokine environment created by T-cells also determines the immunoglobulin (Ig) class of the

antibody produced. The specific effector functions of the antibody (neutralization,

opsonization, complement and cell activation) depend on it's Ig class.

2.2 Cytokines

Cytokines are small, soluble secreted proteins or peptides that effect the growth or function of

cells. Mostly cytokines act in an autocrine (on the same cell that produces them) or paracrine

(cells in the near proximity) way [27]. However, TNF-�, IL-1� and IL-6 may also exhibit

systemic effects [28]. Even though cytokines may be secreted by somatic cells and somatic

cells may be affected by cytokines, their action usually comprises immune cells and they are

functionally involved in orchestrating most aspects of the immune system.

2.2.1 IL-8 (CXCL-8)

Cytokines that exert a chemotactic function towards leukocytes are called chemokines [29].

The most prominent of them is IL-8 or CXCL-8 which primarily attracts phagocytes such as

neutrophils and macrophages. Chemokines are divided into groups depending on their

structure. IL-8 belongs to the chemokine group with one amino acid residue (X) between the

first two cysteins (C) and has therefore got the new name CXCL-8. IL-8 is secreted by

leukocytic cells such as monocytes, macrophages, neutrophils, T-cells and NK-cells; and

somatic cells like endothelial cells, fibroblasts and epithelial cells [29]. Secretion of IL-8 is

activated by pro-inflammatory cytokines (IL-1, TNF-�), bacterial (LPS) or viral products via

the transcription factors NF-�B and AP-1 [29]. The main effect of IL-8 is the recruitment of

neutrophils and monocytes to the site of infection. IL-8 also plays an important role in

neutrophil activation by increasing degranulation, oxidative burst and intracellular calcium

concentrations as well as enhancing the killing of intracellular pathogens such as Candida

Page 16: Dietary immune-modulation – carbohydrate specific effects ...

14

albicans [29]. Basophils, eosinophils, T-cells and B-cells have also been reported to follow

IL-8 gradients [29]. IL-8 can selectively inhibit IL-4 induced IgE production, and mice

deficient in CXCR2 (the receptor for IL-8) showed increased IgE production in allergen-

induced lung inflammation [29]. In the gut, IL-8 has been shown to play an important role in

mucosal healing [30]. IL-8 enhances the migration of intestinal epithelial cells (IEC) in vitro

[31]. Furthermore, the secretion of IL-8 by IEC was shown to be sufficient to recruit

neutrophils into the subepithelial matrix, but neutrophil activation, transepithelial transversion

and resulting tissue damage were not observed [32, 33]. The role of IL-8 in tumor biology is

at least two-sided. On the one hand, IL-8 recruits neutrophils, which can directly kill tumor

cells [29]. On the other hand, the potent angiogenic activity of IL-8 may induce

neovascularization of the tumor thereby promoting tumor survival. IL-8 has also been shown

to induce the expression of matrix metallo-proteinases, which are related to metastasis [29].

Interestingly, oxygen deprivation has been reported to be an activating factor for IL-8

secretion by tumor cells [29]. Malignant transformations in some cell lines have been linked

to constitutive activation of the transcription factors NF-�B and AP-1, which amongst other

things results in increased IL-8 production [29]. Most malignant colonic epithelial cells have

been reported to overexpress IL-8 [34].

2.2.2 TNF-�

TNF-� is a pro-inflammatory cytokine with diverse biological effects, which was originally

identified as an anti-tumor agent [35]. Local production of TNF-� plays an important role in

containment and elimination of local infections [28]. However, systemically released TNF-�

is responsible for substantial pathology in connection with septic shock [36]. The expression

of TNF-� is therefore tightly regulated on the transcriptional, translational and post-

translational level [35]. Monocytes and macrophages release TNF-� in response to stimuli

such as LPS [35]. TNF-� secretion by T-cells is initiated by T-cell receptor activation. Also

Page 17: Dietary immune-modulation – carbohydrate specific effects ...

15

B-cells, NK cells and some non-immune cells can produce TNF-� [35]. The diverse effects of

TNF-� are mediated by two receptors TNFR1 and TNFR2, which are present on basically all

cell types [35]. The effect of TNF-� on macrophages includes the increased production of

cytokines, enhanced phagocytosis and anti-microbial response [36]. Endothelial cells

upregulate leucocyte adhesion molecules in response to TNF-� and contribute thereby to

leucocyte recruitment [36]. Furthermore, TNF-� is also involved in cell proliferation,

differentiation and death through apoptosis [36]. The anti-tumor effect of TNF-� has been

suggested to be mediated by direct cytotoxicity against tumor cells, activation of immune

responses against the tumor and damage of tumor blood vessels [35]. TNF-� is also involved

in chronic inflammatory diseases such as rheumatoid arthritis and Crohn`s disease [36].

2.2.3 IL-1�

The cytokines IL-1� and IL-1� display the same biological activities and bind to the same

receptor complex. Both cytokines are synthesized as pre-cursor proteins that have to be

cleaved into their active forms in order to be secreted [36]. However, most cells are unable to

process pro-IL-1�, which therefore only shows local activity as a membrane bound cytokine

[36]. IL-1� is one of the major cytokines released by monocytes and macrophages in response

to LPS [28]. Locally, IL-1� contributes to pathogen clearance by activation of macrophages

and T-cells. IL-1� increases the recruitment of inflammatory cells by activation of the

vascular endothelium and matrix metallo-proteinases that lead to local tissue destruction

thereby increasing the influx of effector cells [28, 36]. Systemically, IL-1� induces fever by

binding to cells of the hypothalamus and mobilizes neutrophils from the bone marrow [28].

Furthermore, IL-1� stimulates the production of IL-6, which in turn activates liver cells to

secrete acute phase proteins, thereby contributing to pathogen clearance [28].

Page 18: Dietary immune-modulation – carbohydrate specific effects ...

16

2.3 The gut immune system

The mucosal immune system constitutes the largest immune tissue of the body due to its

extensive exposure to antigens [28]. The gut immune system has to accomplish the task of

protecting the enormous surface area against the invasion by pathogens and maintaining

tolerance towards commensal bacteria and food antigens to avoid tissue damage. Peyer's

patches in the small intestine, solitary lymphoid follicles over the whole intestine and

mesenteric lymph nodes form the organized lymphoid tissue of the mucosal immune system

in the gut. Figure 2 illustrates the organization of a Peyer's patch. The epithelium covering

Peyer`s patches and isolated lymphoid follicels contains specialized epithelial cells called

microfold cells (M cells). Unlike normal enterocytes M cells are not covered by mucus and

lack a thick surface glycocalix [37]. M cells are specialized for the uptake and transcellular

transport of antigens and macromolecules from the gut lumen.

Figure 2: Schematic illustration of the organization of a Peyer`s patch follicle from [28]

Scattered immune cells can be found in the epithelial lining or in the lamina propria below. In

the human gut there are approximately 10-20 intraepithelial lymphocytes (IEL) per 100

enterocytes [38]. Over 80% of the IEL are CD8+ T-cells with a high proportion of innate like

T-cells [28]. Throughout the entire intestinal tract DC can protrude their dendrites through the

Page 19: Dietary immune-modulation – carbohydrate specific effects ...

17

epithelial lining to sample the gut lumen for antigens [38]. Intestinal macrophages are located

in the subepithelial area where they can phagocytose microorganisms that have crossed the

epithelial barrier [38]. The lamina propria also contains CD4+ T-cells, CD8+ T-cells, plasma

cells, DC, mast cells and eosinophils [28]. The organization of the mucosal immune system is

illustrated in figure 3.

Figure 3: Illustration of the organization of the mucosal immune system, modified from [28]

The mucosa contains a high number of immune cells in the absence of disease or

inflammation. This is also reflected in the high number of IgA producing B-cells in the lamina

propria, an antibody type that is non-inflammatory and can be translocated to the gut lumen

via a special secretion system of IEC [28, 38]. Maintaining intestinal immune homeostasis is

an active process that involves commensal bacteria and IEC [39-41]. IEC do not only provide

a physical barrier to the gut lumen, recent research findings suggest that they play a primary

role in initiating innate immune responses and maintaining homeostasis [39, 40]. One

important role of IEC in this respect is the “conditioning” of DCs to drive non-inflammatory

TH2 type responses [42]. This process is counteracted by basolateral exposure of IEC to

Page 20: Dietary immune-modulation – carbohydrate specific effects ...

18

bacteria, which restores the ability of DC to drive protective TH1 responses to bacterial

pathogens [42]. IEC express TLR-3 (recognizing double stranded RNA) and TLR-5

(recognizing bacterial flagellin) and low levels of TLR-2 and TLR-4 [40]. TLR-3 is located

intracellular at endosomal membranes, while TLR-2, 4 and 5 are expressed on the cell surface

[40]. It has been reported that the apical activation of TLR-5 on IEC lead to expression of

anti-apoptotic genes, while basolateral TLR-5 ligation induces pro-inflammatory responses

[40]. Besides compartmentalization, different responses can also be evoked by the length of

exposure. The stimulation of IEC with TLR-4 ligands e.g. induces proliferation, NF-�B

activation and inflammatory cytokine secretion while persistent exposure has an inhibitory

effect [40]. TLR signaling may also be important for epithelial cell function as TLR-2

signaling was shown to decrease permeability of the epithelial layer and maintain its integrity

[40]. It has been suggested that the composition of the diet influences IEC gene expression

and signaling to immune cells [43]. In particular the SCFA butyrate was shown to upregulate

IL-8 secretion by IEC [43]. Targeted expression of the mouse equivalent to IL-8 (MIP-2) in

epithelial cells of the small intestine and proximal colon resulted in targeted neutrophil and

lymphocyte infiltration [43]. One may therefore hypothesize, that immune-modulating dietary

components such as �-glucans could influence the complex network of IEC and underlying

immune-cells.

3. �-glucan

3.1 �-glucan structure and structure-function relationships

�eta-glucans form a very heterogeneous group of carbohydrate polymers found in the cell

walls of yeast, fungi, brown algae, and cereal grains or as extracellular polysaccharides

produced by certain bacteria. Beta-glucans from different sources share the common structure

Page 21: Dietary immune-modulation – carbohydrate specific effects ...

19

of �-linked glucose monomers, but differ in linkage type and side chains. Differences in �-

glucan structure for �-glucans from different sources are schematically illustrated in figure 4.

Figure 4: Schematic illustration of the �-glucan structure of �-glucans from different sources

adapted from [44].

Cereal �-glucans are linear mixed linked polymers consisting of blocks of �-(1,3)-linked

cellotriosyl and cellotetraosyl units [45]. They are soluble in hot water, have high molecular

weight (MW) in their native form in the grain cell walls and primarily exhibit random coil

conformation in solution [23, 46-48]. The properties of cereal �-glucans are discussed in more

detail in Paper I (review paper). Curdlan, a linear �-(1,3) linked polymer from Alcaligenes

faecalis, is the best known bacterial �-glucan, while the small, soluble laminarin is the most

common algae derived �-glucan. The structure of laminarin consists of a �-(1,3)-linked

backbone with a low degree of branching (DB) of single �-(1,6) linked residues [49]. Fungal

�-glucans are branched polysaccharides with a backbone of �-(1,3)-linked glucose monomers

and �-(1,6)-linked side chains of varying length and distribution [50]. Among the fungal �-

glucans, �-glucans from yeasts usually contain longer side chains than mushroom derived �-

glucans which may have single �-glucopyranosyl residues as side chains giving them a comb

Page 22: Dietary immune-modulation – carbohydrate specific effects ...

20

like structure [50-52]. The detailed structure of cereal and yeast �-glucans is shown in

figure 5.

Figure 5: Structural properties of cereal and yeast �-glucans adapted from [44]

Molecular weight, molecular structure, solubility and conformation in solution (triple helix,

single helix or random coil), which may actually depend on each other, and co-extracted

compounds are all supposed to be important for �-glucans biological effects. In a review from

1995 Bohn and BeMiller summarized the structure-function relationships of fungal �-glucans

with a comb like structure on anti-tumor activity and concluded that DB of 0.2-0.33, high

molecular weight and a triple helical conformation were important [51]. Also later studies

found a correlation between triple helical structure and anti-tumor activity or cytokine

secretion [52, 53]. However, the use of alkali treatment to disrupt the triple helical structures

may also have affected the activity of contaminants like LPS. This is further strengthened by

the fact that re-naturation of the triple helical structure after alkali treatment was unable to

recover the previously observed immune modulating effect [52]. Accordingly, the extent to

which the degree of branching, triple or single helix conformation and molecular weight

influence the immune modulating properties of �-glucans is still debated. The influence of

these parameters likely depends on the immune activation pathway studied, including effector

Page 23: Dietary immune-modulation – carbohydrate specific effects ...

21

cells, specific �-glucan receptors, way of administration (oral or parenteral), animal model

(genetic background), tumor or infection type. So far, only differences in the ability to

activate the �-glucan receptor dectin-1 for particulate versus soluble �-glucans have been

clearly demonstrated and linked to their respective mechanisms in tumor therapy [21, 54].

Table 1 gives an overview over the most commonly used �-glucan preparations in immune

modulation studies.

Page 24: Dietary immune-modulation – carbohydrate specific effects ...

22

Tabl

e 1:

Com

mon

ly u

sed

�-gl

ucan

pre

para

tions

in im

mun

e m

odul

atio

n. H

igh

MW

> 5

00kD

a; lo

w M

W <

100

kDa.

Ref

eren

ces g

ive

exam

ples

of s

tudi

es u

sing

the

part

icul

ar p

repa

ratio

n. Y

east

�-g

luca

n pr

epar

atio

ns c

an b

e fo

und

unde

r diff

eren

t tra

de n

ames

.

Nam

e So

urce

Sp

ecia

l str

uctu

re/p

urity

M

W

Solu

bilit

y an

d co

nfor

mat

ion

in

wat

er

Refe

renc

es

Yeas

t; �-

(1,3

)link

ed b

ackb

one

with

long

�-(1

,6)-l

inke

d sid

e ch

ain

bran

ches

Zym

osan

Sa

ccha

rom

yces

cer

evisi

ae

Crud

e ce

ll w

all p

repa

ratio

n,

cont

aini

ng m

anno

se e

tc.

Pa

rtic

ular

, uns

olub

le

[55-

59]

WGP

(who

le g

luca

n pa

rtic

le)

Sacc

haro

myc

es c

erev

isiae

Pu

re �

-glu

can

prep

arat

ion

High

Pa

rtic

ular

, uns

olub

le

[19,

21,

54]

PGG

Sa

ccha

rom

yces

cer

evisi

ae

150

kDa

Solu

ble

[21,

60]

Gl

ucan

pho

spat

e Sa

ccha

rom

yces

cer

evisi

ae

Chem

ical

ly m

odifi

ed

Solu

ble

[61,

62]

M

ushr

oom

/Fun

gus;

�-(1

,3)-l

inke

d ba

ckbo

ne w

ith sh

ort �

-(1,6

)-lin

ked

side

chai

n br

anch

es

Lent

inan

Le

ntin

us e

dode

s (Sh

iitak

e m

ushr

oom

) Si

ngle

�-(1

,6)-l

inke

d gl

ucos

e re

sidue

s

May

form

trip

le

helic

es

[63-

65]

SSG

(scl

erot

inia

sc

lero

tioru

m g

luca

n)

Scle

rotin

ia sc

lero

tioru

m

Sing

le �

-(1,6

)-lin

ked

gluc

ose

resid

ues,

DB

0.33

N

ativ

e hi

gh

Solu

ble,

form

s trip

le

helix

in so

lutio

n [6

1, 6

2, 6

6]

SPG

(Son

ifila

n/Sc

hizo

phyl

lan)

Sc

hizo

phyl

lum

com

mun

e Si

ngle

�-(1

,6)-l

inke

d gl

ucos

e re

sidue

s, D

B 0.

33

So

lubl

e, tr

iple

hel

ix in

so

lutio

n [6

7, 6

8]

Bact

eria

l; lin

ear �

-(1,3

)-lin

ked

glu

can

Curd

lan

Alca

ligen

es fa

ecal

is Hi

gh

Uns

olub

le

[59,

69-

71]

Seaw

eed;

�-(1

,3)-l

inke

d gl

ucan

with

som

e �-

(1,6

)-lin

ked

bran

ches

Lam

inar

in

Lam

inar

ia d

igita

ta

Sing

le �

-(1,6

)-lin

ked

gluc

ose

resid

ues,

DB

<0.1

5 Lo

w

Solu

ble

[54,

61,

62,

72]

Cere

als;

line

ar m

ixed

link

age

�-(1

,4)-�

-(1,3

) glu

cans

Barle

y an

d oa

t Ho

rdeu

m v

ulga

re L

./Av

ena

sativ

a

MW

in th

e pl

ant >

106

Da; i

n pr

epar

atio

ns

depe

ndin

g on

isol

atio

n pr

oced

ure

Larg

ely

solu

ble

[19,

63,

73-

78]

Page 25: Dietary immune-modulation – carbohydrate specific effects ...

23

3.2 �-glucan receptors

Dectin-1

Dectin-1 is a non TLR pattern-recognition receptor with a single extracellular C-type lectin

like domain [79]. Dectin-1 is expressed on human macrophages, neutrophils and DC [80].

Recent investigation has revealed that dectin-1 activation requires the clustering of the

receptor in a synapse like fashion [54]. This can only be achieved by particulate �-glucan,

while soluble �-glucans, regardless of their molecular weight, remained unable to activate

dectin-1 [54]. Dectin-1 activation increases phagocytosis, triggers the release of reactive

oxygen species (ROS) and activates the transcription of cytokines [24, 54]. Dectin-1 is also

able to induce adaptive immunity as the activation of dectin-1 by particulate yeast �-glucan

has been shown to activate DC [21, 69]. �eta-glucan activated DC in turn preferentially

primed TH1 or TH17 differentiation of naïve CD4 T-cells and were found to activate CD8+ T-

cells to become cytotoxic effector cells [21, 69]. The effectiveness of particulate �-glucan

induced TH1 and cytotoxic T-cell responses to mediate tumor regression was further

demonstrated in mouse tumor models [21].

Complement receptor 3

Complement receptor 3 (CR3) is expressed on most myeloid cells like monocytes,

macrophages, DC, neutrophils and NK cells [24]. Beta-glucan binding to the lectin side of the

CR 3 receptor induces a primed state of the receptor that enables neutrophil or NK cell

mediated killing of iC3b opsonized target cells [72, 81]. In recent years, �-glucans (both from

yeast and barley) have been demonstrated to significantly increase the therapeutic effect of

anti-tumor monoclonal antibodies by priming the CR 3 receptor for complement mediated

killing of opsonized tumor cells [17-21].

Page 26: Dietary immune-modulation – carbohydrate specific effects ...

24

Lactosylceramide

Lactosylceramide is a glycosphingolipid PRR found in lipid rafts of the plasma membranes of

many cell types [24, 79]. The receptor has been implicated in soluble yeast �-glucan induced

respiratory burst activity and NF-�B activation in human polymorphonuclear leucocytes [82].

Lactosylceramide has also been demonstrated to mediate NF-�B activation and cytokine

production in response to fungal �-glucan preparations in murine alveolar epithelial cells [83,

84]. Lactosylceramide is supposed to play a role in �-glucan induced immune-modulation

especially in non-immune cells [79].

Scavenger Receptors

By definition, scavenger receptors are all able to recognize modified low density lipoproteins.

Apart from that, their structures may vary greatly [79]. Scavenger receptors on human

monocytes have been shown to bind soluble yeast �-glucan, however, no specific receptor

was identified [85]. The scavenger receptor family member CD5 was later shown to bind

fungal cell wall components and facilitate Zymosan induced IL-8 secretion by CD5

transfected HEK293 cells [86].

3.3 Possible mechanisms behind immune modulation by dietary �-glucans

The immune-modulating effect of orally applied �-glucans has been demonstrated in several

animal models [18, 19, 21, 74, 77, 78, 87-92]. However, the mechanisms by which they exert

their effects are still poorly understood. Even though several receptors and signaling pathways

have been identified, the multiple steps from uptake in the intestine to the activation of

effector cells are still unknown. Figure 6 gives an illustration of dietary �-glucans possible

pathways from the gut to immune-modulation.

Page 27: Dietary immune-modulation – carbohydrate specific effects ...

25

Figure 6: �-glucans possible steps from the intestine to immune-modulation

Dietary �-glucans are indigestible by mammalian enzymes and are supposed to reach the

large intestine more or less intact. Here they may be fermented by the indigenous microbiota,

which can lead to changes in bacterial composition and released metabolic compounds

(Figure 6, part A). Gastrointestinal microorganisms and their metabolites may affect the

immune system as immune-modulation is one of the specific health benefits that can be

mediated by probiotics, which by definition are live microorganism that survive

gastrointestinal passage and exert positive health effects on the host [10]. Most probiotic

microorganisms belong to the genera of Lactobacilli and Bifidobacterium [10]. Cereal �-

glucan is readily fermented and in vitro fermentation studies have shown increased production

of short chain fatty acids (SCFA) especially propionate but without selective stimulation of

the growth of Bifidobacteria or Lactobacilli [93, 94]. Even though �-glucans may not be

classic prebiotics (selectively promoting the growth of Bifidobacteria or Lactobacilli [95]),

they could still promote immune-modulation by the intestinal flora through e.g. changes in

Page 28: Dietary immune-modulation – carbohydrate specific effects ...

26

SCFA production or microbiota composition other than Bifidobacteria or Lactobacilli.

However, the exploration of possible microbiota based mechanisms for �-glucans was not

part of this thesis and emphasis is instead laid on the direct interaction of �-glucans with

intestinal epithelial cells.

Direct interaction of �-glucans with immune competent cells (Figure 6, part B) can take place

locally in the gut by interaction with intestinal epithelial cells, a cell type that has been shown

to play a major role in orchestrating the gut immune system [39]. Also intra-epithelial

lymphocytes and DC that protrude their dendrites through the epithelial lining are in direct

contact with the gut lumen and therefore potential targets for direct interactions with luminal

�-glucans (illustrated in the right panel of figure 6) [38]. DC have been suggested to

contribute to �-glucan uptake by sampling of the gut lumen and transporting �-glucans to

mesenteric lymph nodes [96]. Recent studies have shown the potential of particulate yeast �-

glucan and curdlan to activate DC [21, 70]. However, the contribution of DC to �-glucan

uptake has still to be shown experimentally. How and to what extent �-glucans may enter the

blood stream is still debated [15]. Fluorescently marked barley and yeast �-glucans have been

detected in macrophages isolated from spleen, lymph nodes and bone marrow of mice

following oral administration [19]. The authors hypothesized that the �-glucans were taken up

and transported by gastrointestinal macrophages. However, since gastrointestinal

macrophages are located in the subepithelial area uptake over the intestinal cell layer must

have occurred. The presence of different fluorescently labeled fungal �-glucans in the plasma

of rats after oral administration has also been demonstrated [62]. On the other hand,

fluorescence labeling may change the properties of the polymers, especially if the fluorescent

molecules are bound to the main chain and not only to the reducing end [97]. This may alter

the mechanism of uptake. Labeling of the reducing end may influence the polymer structure

to a lesser degree. However, it increases the chance of dissociation of the fluorescent

Page 29: Dietary immune-modulation – carbohydrate specific effects ...

27

molecule. Hence, fluorescent measurements may not reflect the distribution of the polymer

but the detached label or detached labeled fragment of the original polymer. Specific and

highly sensitive detection methods that do not require �-glucan derivatization may therefore

be helpful tools in further investigations on �-glucan uptake.

Figure 6, part C, illustrates the possible uptake mechanisms for �-glucans from the gut lumen.

Specialized epithelial cells (M-cells) in the follicle associated epithelium of Peyer`s patches

have repeatedly been suggested to mediate �-glucan uptake [44, 61, 66]. The possible role of

M-cells in �-glucan uptake is discussed in more detail in Paper I (review paper). However,

even though uptake of �-glucans via M-cells is a highly plausible mechanism, experimental

verification is still lacking. Besides M-cells, normal intestinal enterocytes may also play a role

in �-glucan uptake. Even though they are not specialized for the uptake of macromolecules

and antigens, enterocytes have been shown to transcytose nanoparticles [98] and protein

antigens [99], and they are highly abundant in the GI tract.

4. LPS

LPS is a part of the membrane of gram negative bacteria. Humans have evolved to react to

low levels of LPS to initiate innate immunity [100]. Especially monocytes and macrophages

respond to LPS by changing the expression of thousands of genes often by several orders of

magnitude [36]. The intestinal tissue, however, does not strongly respond to LPS [40]. LPS

induces the production of cytokines such as IL-1�, TNF-�, IL-12 and IL-8. The systemic

secretion of TNF-� by e.g. liver macrophages during sepsis is the main cause of fatal septic

shock [28]. LPS is also known to upregulate the expression of adhesion molecules on

endothelial cells, which increases leucocyte migration and to increase the expression of the

co-stimulatory molecules B7.1 and B7.2 on macrophages and DCs thereby helping to initiate

adaptive immune responses [28].

Page 30: Dietary immune-modulation – carbohydrate specific effects ...

28

The LPS molecule consists of three different parts: the hydrophobic Lipid A, a core

oligosaccharide and the O-antigen or O-chain. The molecular structure of LPS is illustrated in

figure 7. While Lipid A anchors the molecule in the bacterial membrane; the O-antigen forms

the outer most part of LPS. The O-antigen consists of repeating sugar sequences of 3-5 sugar

moieties [101]. Differences in the composition of the O-antigen between bacterial species and

strains are responsible for their different antigenicity and determine the bacterial serotype

[36]. The core region is composed of unusual monosaccharides such as 2-keto-3-

deoxyoctonoic acid, which is characteristic for LPS and therefore sometimes used for LPS

quantification [102]. The Lipid A part contains several long chain fatty acid residues and is

highly conserved among gram negative bacteria [101]. Lipid A is responsible for the

induction of innate immunity by LPS [36, 101].

Figure 7: Illustration of the molecular structure of LPS and its localization in the cell wall of

gram negative bacteria, from [103]

Page 31: Dietary immune-modulation – carbohydrate specific effects ...

29

The toll like receptor 4 (TLR-4) was first identified as the LPS signaling receptor through

genetic studies of mutant mice, which were completely unresponsive to LPS and had a single

mutation in the TLR-4 gene [104]. LPS signaling is now known to be mediated by the LPS

receptor complex consisting of LPS-binding protein (LBP), CD14, the lipid binding accessory

protein MD-2 and TLR-4. The circulating acute phase protein LBP is able to extract LPS

form the membrane of gram negative bacteria [36]. CD14 is a cell surface marker of myeloid

cells but can also be present in a soluble, secreted form [104]. LPS binding to TLR-4 results

in the initiation of two signaling pathways the MyD88 dependent and the MyD88 independent

pathway. In the MyD88 dependent pathway the MyD88 adaptor protein is directly recruited to

the cytoplasmic tail of TLR-4. A sequence of several adaptor proteins and kinases leads to the

activation of transcription factors such as NF-�B, AP1 and ELK-1 [36]. In myeloid cells an

additional MyD88 independent pathway exists, which starts from endocytosed TLR-4. Signal

transduction via the MyD88 independent pathway may lead to NF-�B activation or the

activation of interferon regulatory factor 3, a transcription factor promoting the expression of

type I interferons [28].

Due to its strong effects on innate immunity at very low doses (pg to ng level) LPS is a usual

confounder in in vitro immune-modulation assays. Several methods to assess the LPS content

of different samples or the contribution of LPS to the observed immune-modulation have

therefore been developed. Besides the above mentioned method making use of the unusual

sugar 2-keto-3-deoxyoctonoic acid [102], GC-MS detection of 3-hydroxy fatty acid from the

Lipid A part of LPS as methyl esters has also been used [105]. However, due to their high

sensitivity, assays based on the specific reaction of limulus amoebocyte lysate (LAL), an

extract from the amoebocytes of the horsehoe crab Limulus polyphemus, with LPS are

preferentially used [106]. LAL also contains a protein, called factor G, which reacts with �-

glucans and therefore has to be removed if the test is to be used for �-glucan preparations

Page 32: Dietary immune-modulation – carbohydrate specific effects ...

30

[106]. Alternatively, the LPS inhibitor Polymyxin B is often used for indirect evaluation of

LPS contamination in cell culture tests [73, 106-108].

Page 33: Dietary immune-modulation – carbohydrate specific effects ...

31

5. Aims of the study

The main aim of the study was to increase the current understanding of how dietary �-glucans

may modulate the human immune system by focusing on the first step from the gut lumen to

the activation of effector cells. Since cereal derived �-glucans are a part of our everyday diet

they received the main focus, but also �-glucans from yeast were investigated for comparison

purposes. The study comprises work on possible mechanisms of action and structure

functional relationships. The development of new methods to study �-glucan uptake and

carbohydrate specific effects in cell cultures were a major part of the project. Specifically the

aims were to:

� Investigate possible interactions of �-glucans with intestinal epithelial cells by

studying cytokine secretion from intestinal epithelial cell lines Caco-2 and HT-29

alone (mono-culture) or in co-culture with THP-1 derived human macrophages

� Develop a new method for the detection of low concentrations of cereal �-glucans in

cell culture supernatants and apply this method to study the transport of cereal �-

glucan across differentiated Caco-2 cell monolayers

� Investigate possible structure-function relationships of cereal �-glucans by using

different highly purified �-glucan samples (commercial and extracted from Norwegian

barley varieties) over a wide molecular weight range and comparing the effect with

yeast �-glucan preparations by studying cytokine secretion from different intestinal

epithelial cell lines.

� Develop tools to study the carbohydrate specific effects of yeast and cereal �-glucan

preparations in vitro taking potential contamination by e.g. LPS into account

Page 34: Dietary immune-modulation – carbohydrate specific effects ...

32

6. List of papers and manuscripts

I Rieder, A., Samuelsen, A. B., Do cereal mixed-linked beta-glucans possess immune-modulating activities? Mol. Nutr. Food Res. 2012, 56, 536-547.

II Samuelsen, A. B., Rieder, A., Grimmer, S., Michaelsen, T. E., Knutsen, S. H.,

Immunomodulatory Activity of Dietary Fiber: Arabinoxylan and Mixed-Linked Beta-Glucan Isolated from Barley Show Modest Activities in Vitro. Int. J. Mol. Sci. 2011, 12, 570-587.

III Rieder, A., Grimmer, S., Kolset, S. O., Michaelsen, T. E., Knutsen, S. H., Cereal

beta-glucan preparations of different weight average molecular weights induce variable cytokine secretion in human intestinal epithelial cell lines. Food Chem. 2011, 128, 1037-1043.

IV Rieder, A., Knutsen, S. H., Ballance, S., Grimmer, S., Airado-Rodriguez, D.,

Cereal beta-glucan quantification with calcofluor-application to cell culture supernatants. Carbohydrate Polymers 2012, 90, 1564-1572.

V Rieder, A., Grimmer, S., Aachmann,F.L., Westereng, B., Kolset, S.O., Knusten, S.H., Generic tools to assess genuine carbohydrate specific effects on in vitro immune-modulation exemplified by �-glucans. Carbohydrate Polymers 2013, 92,2075-2083.

VI Rieder, A., Knutsen, S.H., Berget, I., Kolset, S.O., Grimmer, S., Cereal and yeast

�-glucan preparations modulate cytokine secretion by human macrophages, differentiated Caco-2 cells and Caco-2 macrophage co-cultures. Manuscript

Page 35: Dietary immune-modulation – carbohydrate specific effects ...

33

7. Results

7.1 Review of cereal �-glucans immune-modulating properties (Paper I)

Beta-glucans are a heterogeneous group of glucose polymers with distinct structures

depending on their respective sources. However, this has not always been fully accounted for

in reviews on the immune-modulating properties of �-glucans. We have therefore chosen to

focus on cereal �-glucans in this review. Studies on cereal �-glucans have shown effects in

vitro on cytokine secretion, phagocytic activity and cytotoxicity of isolated immune cells/cell

lines, and activation of the complement system. However, not all conducted studies take the

potential contamination of test samples with e.g. LPS or other immunologically active

substances into account and therefore need to be interpreted with care. Cereal �-glucans have

been shown to increase antibody dependent cellular cytotoxicity in murine cancer models.

Animal studies further suggest a protective effect against infections caused by intestinal

parasites, bacteria or virus. However, uptake is still debated even though activity in animal

studies has been demonstrated for orally applied cereal �-glucan. The use of completely

different model systems in the currently available studies made it impossible to conclude on

clear structure-function relationships and more research is needed to clarify mechanisms of

action, uptake and possible activity of cereal �-glucans in humans.

7.2 Effect of high MW �-glucan fractions and arabinoxylan fractions isolated from

barley on IL-8 secretion by the intestinal epithelial cell lines Caco-2 and HT-29, NF-�B

activity in monocytic U937 cells and complement fixation (Paper II)

In this study we have investigated the potential immune-modulatory effects of fiber fractions

isolated from barley in different test systems. Two arabinoxylan and �-glucan rich fractions

with different purity were isolated from the Norwegian barley variety Tyra. The four fractions

Page 36: Dietary immune-modulation – carbohydrate specific effects ...

34

showed no significant effect on IL-8 secretion and cell proliferation of the two intestinal

epithelial cell lines HT-29 and Caco-2. Furthermore, the fractions had no effect on basal or

LPS induced NF-�B activity in the stably transfected monocytic NF-�B reporter cell line

U937-3kB-LUC. Arabinoxylan and �-glucan rich fractions were also extracted from three

other barley varieties and their ability to fix complement via the classical pathway was

investigated. Some �-glucan fractions showed activity on the same level as the positive

control (a pectin fraction from Plantago major with known complement activating abilities

[109, 110]) and �-glucan fractions generally exhibited higher activity than arabinoxylan

fractions. Differences between the �-glucan fractions could not be ascribed to differences in

�(1,3)/�(1,4) linkage ratios or MW. The estimated MW of the arabinoxylan fractions was

significantly lower than for the �-glucan fractions and when both fibre types were included in

the statistical test a significant correlation between complement fixing activity and MW was

found.

7.3 Effect of cereal �-glucan fractions with different weight average MW (Mw) on the

cytokine secretion by intestinal epithelial cell lines Caco-2 and HT-29 (Paper III)

In order to investigate the possible relationship between immune-modulatory activity and

MW, cereal �-glucan preparations with weight average MW (Mw) of 40, 123, 245 and 359

kDa were tested for their ability to induce cytokine secretion in intestinal epithelial cells. The

yeast �-glucan preparation Zymosan was used as a positive control. Screening of the secretion

of 18 different cytokines (IL-1�, IL-1�, IL-2, IL-4, IL-5, IL-7, IL-8, IL-10, IL-12, IL-13, IL-

17, G-CSF, GM-CSF, MCP-1, MIP-1�, INF-�, TNF-�) in response to Zymosan and cereal �-

glucans of 40 and 359 kDa Mw revealed that HT-29 cells mainly secreted IL-8 and minor

amounts of IL-2, GM-CSF and INF-�. The secretion of IL-8 was increased by exposure of the

cells to all three �-gucans, while the other three cytokines were not influenced. Unpublished

Page 37: Dietary immune-modulation – carbohydrate specific effects ...

35

results from Caco-2 cells incubated with Zymosan or control medium also showed that IL-8

was the main secreted cytokine, and the only tested cytokine whose secretion was influenced

by the presence of �-glucan (data not shown in paper III). Incubation of HT-29 cells with all

four cereal �-glucans and Zymosan resulted in significantly increased IL-8 secretion for

Zymosan and 40 kDa sample, slightly increased secretion for 359 kDa and no effect for 123

and 245 kDa samples. Caco-2 cells significantly increased IL-8 secretion in response to

Zymosan. None of the tested �-glucan preparation distinctively modulated IL-1� (Caco-2) or

TNF-� (HT-29) induced IL-8 secretion. IL-8 secretion by HT-29 cells in response to the 40

kDa cereal �-glucan was dose and time dependent and not significantly influenced by the

presence of the known LPS inhibitor Polymyxin B. Chemical characterization of the 40 kDa

sample showed glucose as the only sugar constituent and low or no protein contamination.

The increased IL-8 secretion in response to the 40 kDa sample was consequently attributed to

stimulation from the �-glucan molecules.

7.4 Development of a new method to measure low concentrations of cereal �-glucans in

cell culture supernatants and application to analysis of cereal �-glucan transport over

intact Caco-2 cell monolayers (Paper IV)

Fluorescence labeling of �-glucans to enable the study of �-glucan uptake may change the

chemical and biological properties of the polymer. Furthermore, a possible detachment of the

label during the study may confound the obtained results. We have therefore developed a

method to measure low concentrations of cereal �-glucans without the requirement of

derivatization. The method is based on the specific binding of the fluorescent dye calcofluor

to cereal �-glucans resulting in increased fluorescence intensity of the formed complex and

was optimized for high sensitivity. A concentration dependent spectroscopic response value

was calculated based on the use of the derivative signal to emphasize spectral information of

Page 38: Dietary immune-modulation – carbohydrate specific effects ...

36

the calcofluor/�-glucan complex. This strategy combined with working in batch mode gave

rise to a lower detection limit of 0.045μg/mL. The method can be easily applied to measure

the transport of cereal �-glucans over differentiated Caco-2 cell monolayers. Hereby, the

transport experiment is carried out with underivatized cereal �-glucan, whose presence in the

basolateral media is detected with calcofluor after the end of the cell culture experiment.

Basolateral �-glucan concentrations above the detection limit could be measured for

approximately 50% of the Caco-2 cell monolayers after 9 h incubation with 1 mg/mL cereal

�-glucans of different Mw on the apical side. Basolateral �-glucan concentrations were

unrelated to punctures in the Caco-2 cell monolayers as no significant correlation between

transepithelial electrical resistance (TEER) and basolateral �-glucan could be found. In a

follow up experiment (unpublished data) the incubation time was increased to 22 h in order to

obtain basolateral �-glucan concentrations above the detection limit for more than 50% of the

filters. However, no increase in the number of filters with basolateral �-glucan concentrations

above the detection limit could be observed (data not shown in paper IV). Further

investigation revealed that the measured �-glucan in the basolateral compartment originated

from tiny contaminations introduced by the electrode, which was used to measure the TEER

at beginning and end of the experiment (data not shown in paper IV). In an experiment

conducted without the measurement of TEER none of the analyzed filters showed basolateral

�-glucan concentrations above the detection limit (data not shown in paper IV). In spite of the

absence of evidence for �-glucan transport, the data clearly demonstrate the high sensitivity of

the method.

Page 39: Dietary immune-modulation – carbohydrate specific effects ...

37

7.5 Tools to study the carbohydrate specific effect of cereal and yeast �-glucan

preparations (Paper V)

Even if carbohydrate preparations from plant/fungal sources have been prepared to a high

degree of purity, observed immune-stimulation may be caused by minute sample

contaminations. Contamination with LPS is often ruled out by the use of Polymyxin B as a

LPS inhibitor and we have used this technique in paper 3 for testing the effect of the 40 kDa

cereal �-glucan sample. However, the use of targeted enzymatic degradation and careful

evaluation of the degradation products revealed that the effect of the 40 kDa sample on IL-8

secretion by HT-29 cells was unrelated to its �-glucan content. Instead low levels of LPS

contamination were detected by the limulus amoebocyte lysate test and the effect of the 40

kDa sample could be completely described by this contamination. Furthermore, the active

component was removed from the 40 kDa sample by preparative size exclusion

chromatography and subsequently identified as LPS like component by NMR spectroscopy.

Interestingly, the addition of Polymyxin B to the enzymatically degraded 40 kDa sample

suppressed its effect, and demonstrated clearly that Polymyxin B can not be regarded as a

general tool to suppress LPS effects in relevant polysaccharide samples. Antibodies against

TLR-2 and TLR-4 were unable to block the effect of E.coli LPS or active �-glucan samples.

Targeted enzymatic degradation of different yeast �-glucan preparations significantly

decreased the effect of one preparation (MacroGard), indicating an effect of the �-glucan

component of this preparation. Compared to other methods to study carbohydrate specific

effects, targeted enzymatic degradation has the advantage of directly relating an observed

effect to the carbohydrate of interest instead of aiming to exclude the effects of possible

contaminations.

Page 40: Dietary immune-modulation – carbohydrate specific effects ...

38

7.6 Effect of cereal and yeast �-glucan preparations on THP-1 derived human

macrophages, differentiated Caco-2 cells and Caco-2 macrophage co-cultures (Paper VI)

We have previously found undifferentiated Caco-2 and HT-29 cells relatively unresponsive to

�-glucan preparations with very low levels of LPS contamination. However, it has been

indicated that Caco-2 cells only respond to apical stimuli in the presence of basolateral

leukocytes [111]. We therefore used differentiated Caco-2 cells alone (mono-culture) and in

co-culture with THP-1 derived macrophages to further investigate the possible interaction of

�-glucan preparations with intestinal epithelial cells. Caco-2 cells in mono- and co-culture

remained unresponsive towards apical addition of the cereal �-glucans tested. In contrast, two

particulate yeast �-glucans increased basolateral (mono-cultures) and apical (mono- and co-

culture) IL-8 secretion. Both preparations contain low levels of LPS. However, LPS was not

found to influence basolateral IL-8 secretion in monocultures and had a lower effect on apical

secretion, especially in co-culture, than the yeast �-glucans. Thus, the observed effects of the

two particulate yeast �-glucan preparations can not be solely explained by their LPS content,

leaving the possibility of a �-glucan related effect open. Incubation of macrophages with

different �-glucan preparations resulted in increased secretion of IL-8, TNF-� and IL-1�.

Beta-glucan preparations with higher LPS contamination generally resulted in higher cytokine

secretions. However, regression analyses of cytokine secretion data obtained with E.coli LPS

revealed that several �-glucan preparations resulted in considerably lower cytokine secretions

than expected based on their LPS content. This indicates that �-glucans may have a

dampening effect on LPS induced cytokine secretion by macrophages. However, more data

are needed to verify this hypothesis.

Page 41: Dietary immune-modulation – carbohydrate specific effects ...

39

8. Discussion

8.1 Interaction with IEC

Beta-glucans are resistant to degradation by the enzymes of the mammalian digestive tract

and thus reach the large intestine intact. Enterocytes are not only the most abundant cell type

�-glucans can encounter during their passage through the GI tract, they also play an important

role in regulating intestinal immune responses [38, 39]. However, studies on the effect of �-

glucans on IEC are limited. Oral administration of oat �-glucan has been shown to increase

NF-�B activation in both leukocytes and enterocytes of the proximal small intestine but not in

the colon of NF-�B reporter mice [77]. Furthermore, oat �-glucan (Mw 60 kDa) enriched fecal

water from ileostomy patients consuming a �-glucan rich diet has been reported to increase

IL-8 secretion and ICAM-1 expression in selected intestinal epithelial cell lines [112]. The

effect was, however, only observed when the cells were co-stimulated with a mixture of pro-

inflammatory cytokines (TNF-�, IL-1�, INF-�). Due to the experimental design it was not

possible to conclude wether the observed effect was mediated directly by the �-glucan

contained in the fecal water (0.12 – 0.18 mg/mL) or indirectly by �-glucan induced changes in

the composition of the in fecal water.

In order to study the direct interaction of �-glucan with intestinal epithelial cell lines in vitro,

we developed a protocol to solubilize cereal �-glucans in cell culture media (Papers II, III, V

and VI). Commercial �-glucan samples of different Mw (40-359 kDa) and �-glucan isolated

from different barley varieties (Mw about 600-900 kDa) were tested. The cereal �-glucan

sample with the lowest Mw (40 kDa) significantly increased IL-8 secretion by HT-29 cells

(Paper III). However, the activity was subsequently attributed to contamination of the sample

with LPS (Paper V). None of the other tested cereal �-glucan samples significantly increased

the cytokine secretion by HT-29 or Caco-2 cells (Paper II, III and V). Stimulation of HT-29

cells with TNF-� and Caco-2 cells with IL-1� did not show any effect of the tested �-glucan

Page 42: Dietary immune-modulation – carbohydrate specific effects ...

40

samples on cytokine induced IL-8 secretion (Paper III). Furthermore, apical addition of cereal

�-glucans to differentiated Caco-2 cells in mono- or co-culture with THP-1 derived

macrophages did not show any effect on cytokine secretion (Paper VI). However, our

experiments with yeast �-glucans revealed a potential of the particulate yeast �-glucan

preparations MacroGard and Zymosan to stimulate cytokine secretion by IEC. Both samples

increased IL-8 secretion by HT-29 cells, and targeted enzymatic degradation revealed that at

least the observed activity of MacroGard could be related to the �-glucan part of the sample

(Paper V). Apical addition of MacroGard or Zymosan to differentiated Caco-2 cells in mono-

culture or in co-culture with THP-1 derived macrophages resulted in increased basolateral

(mono-culture) and apical (mono- and co-culture) IL-8 secretion (Paper VI). Interestingly,

differentiated Caco-2 cells, especially when in co-culture, did not react strongly to the

presence of LPS. Thus, even though both �-glucan samples contained low levels of LPS, their

observed effects on IL-8 secretion could not be solely explained by their LPS content, leaving

the possibility of a �-glucan related effect open. On the other hand, a highly purified soluble

yeast �-glucan (Wellmune) did not elicit any effect on cytokine secretion by HT-29 cells or

differentiated Caco-2 cells in mono- or co-cultures (Paper V and VI).

Our experiments with cereal �-glucans as well as other reported studies [77, 112] suggest that

cereal �-glucans are unable to directly activate enterocytes to secret cytokines. However, even

though we did not find any effect of cereal �-glucans on IL-8 secretion by IEC lines, cereal �-

glucans may have a direct effect on other aspects of enterocyte biology. It has for instance

been shown that the consumption of oat �-glucan decreased the levels of antimicrobial

peptides in the fecal water of ileostomy patients [113]. Cereal �-glucans may activate

intestinal leukocytes, which then in turn may activate enterocytes as suggested by Volman et

al., 2010 [77]. More research is needed to increase the current understanding of the interaction

of cereal �-glucans with IEC. Our results with yeast �-glucans on the other hand, suggest that

Page 43: Dietary immune-modulation – carbohydrate specific effects ...

41

particulate but not soluble yeast �-glucan preparations may increase IL-8 secretion by IEC.

Increased IL-8 secretion by IEC may be beneficial as IL-8 has been shown to play an

important role in mucosal healing [30, 31]. Furthermore, IL-8 secretion by IEC has been

shown to lead to the recruitment of neutrophils and lymphocytes [32]. One may speculate that

the increased number of immune cells in the epithelial lining may contribute to the potential

benefit of �-glucans on resistance to infection. Interestingly, oral administration of yeast �-

glucan to mice has also been reported to increase the amount of intraepithelial lymphocytes

[90].

8.2 Uptake

Uptake of �-glucans from the gut is an important aspect in the understanding of the potential

immune-modulating properties of dietary �-glucans. However, only limited information on �-

glucan uptake is available. Uptake has been indicated by the detection of fluorescein

dichlorotriazine labeled yeast �-glucan particles (WGP) and barley �-glucan in the bone

marrow, peritoneal lymph nodes and spleen of mice following oral administration [19].

However, the label fluorescein dichlorotriazine is able to covalently react with hydroxyl

groups of the �-glucan chains and may due to it`s two reactive groups also lead to cross-

linking of the polymers. The resulting �-glucan complexes may have dramatically changed

properties compared to the parent molecules, including a possible altered uptake mechanism

or receptor affinity. Interestingly, the fluorescein labeled �-glucans were also detected in

intracellular compartments in macrophages [19]. In another study Alexa Fluor 488 labeled

laminarin, scleroglucan and glucan phosphate were detected in the serum of rats after oral

administration of 1 mg/kg [61]. Labeling of only the reducing end ensured identical properties

of labeled and unlabeled polymers. Maximum detected serum concentrations of the different

compounds occurred between 3 and 4h after administration and were approximately 40, 115

Page 44: Dietary immune-modulation – carbohydrate specific effects ...

42

and 355 ng/mL for glucan phosphate, laminarin and scleroglucan, respectively. The

bioavailability ranged from 0.5 to 4.9%. Alexa Fluor 488 labeled particulate yeast �-glucan

were included in the same study but could not be detected in the serum of rats. Thus, the

authors hypothesized that particulate �-glucans may be taken up and transported by

macrophages as described by Hong et al. [19]. Another group has used the reactivity of the

limulus amoebocyte lysate (LAL) factor G with �-glucan for the detection of unlabeled

soluble yeast �-glucan in rat serum after oral administration of 20 mg/kg daily over 14 days

[96]. Even though �-glucan concentrations were reported to be significantly different from the

control group, levels of only 5 ng/mL were reached. Together these studies indicate that only

low amounts of �-glucans may be taken up from the gut.

The uptake mechanisms for �-glucans are still unclear, but Rice et al. have found that glucan

phosphate is internalized by a subset of IEC through a dectin-1 independent mechanism [61].

It has been repeatedly suggested that �-glucans are taken up by M cells [44, 61, 89]. M cells

are epithelial cells specialized for the uptake and transport of macromolecules and antigens

over follicle associated epithelium throughout the intestine [37, 114]. However, even though

this is a plausible mechanism, it has never been shown experimentally. Normal IEC are highly

abundant in the gut and have been shown to transcytose soluble protein antigens and

nanoparticles [98, 99], which makes them an interesting candidate for �-glucan uptake. In

order to study the uptake of cereal �-glucans across differentiated Caco-2 cell monolayers, we

developed a new method to detect low concentrations of cereal �-glucans in cell culture

supernatants (Paper IV). The new detection method is based on the specific interaction of the

fluorescent dye calcofluor with cereal �-glucans, and the interaction is carried out after the

cellular studies. Thus, the test does not require �-glucan derivatization and consequently

avoids structural changes that may affect cellular uptake and transport. Detection of low

levels of cereal �-glucan in the basolateral compartments in the first experiments were

Page 45: Dietary immune-modulation – carbohydrate specific effects ...

43

subsequently shown to be related to contamination by the electrode used for TEER

measurement, but demonstrated the applicability of the developed method. Later results

indicated the absence of cereal �-glucan transport over differentiated Caco-2 cell monolayers

or transport below the detection limit of the assay of 45 ng/mL. As outlined above the

published data on �-glucan serum concentrations after oral administration to rats vary

considerably with the �-glucan type (laminarin, scleroglucan, yeast �-glucan, glucan

phosphate) and detection method (fluorescence vs LAL test) from 5 to 355 ng/mL. It is

therefore difficult to decide if the detection limit of 45 ng/mL is too high to measure cereal �-

glucan transport across the cell layer or if the results simply indicate the absence of transport.

In the last experiment, a total of 1.5 mg cereal �-glucan added to the apical side resulted in

less than 60 ng �-glucan on the basolateral side (the addition of calcofluor and buffer solution

to the cell culture supernatant during the �-glucan assay introduced a 3:4 dilution

corresponding to an increase of the detection limit from 45 ng/mL to 60 ng/mL). This

corresponds to an uptake rate of under 0.004% in 22 h. Thus, one might speculate that the

uptake rate is too low to give physiologically relevant concentrations, and furthermore that

IEC do not contribute to a major extent to the uptake of cereal �-glucans from the gut.

Caco-2 cells can be differentiated into cells with phenotypes resembling those of small

intestinal enterocytes. The development of microvilli during the differentiation process can be

shown by scanning electron microscopy (Figure 8).

Page 46: Dietary immune-modulation – carbohydrate specific effects ...

44

Figure 8: Scanning electron micrograph of differentiated Caco-2 cells showing the

development of microvilli (Rieder et al., unpublished results).

Even though differentiated Caco-2 cells are a commonly used in vitro model system for the

study of drug absorption [115-117], the reported TEER values for differentiated Caco-2 cell

layers (average 300-600 �cm2) are considerably higher than for the human intestine (average

30 �cm2) [118]. Consequently Caco-2 cell monolayers have a lower permeability towards

substances transported primarily by the paracellular route [115] and may therefore not be the

ideal model system for transport studies with hydrophilic molecules such as �-glucans, even

though paracellular transport of �-glucans might be limited by their size.

It has been shown that the presence of macrophages on the basolateral side increased the

uptake of microparticles by Caco-2 cell monolayers [119]. Furthermore, the addition of

supernatants from PBMC stimulated with wheat germ agglutinin to the basolateral side of

differentiated Caco-2 cells has been shown to alter the integrity of the Caco-2 cell monolayers

[120]. It may therefore be worthwhile to investigate the uptake of �-glucan over Caco-2 cell

Page 47: Dietary immune-modulation – carbohydrate specific effects ...

45

monolayers in the presence of leukocytes. Furthermore, the use of an in vitro M cell model,

which has been established by the co-culture of differentiated Caco-2 cells with Raji B-cells

[121, 122], to study �-glucan uptake may also prove to be a useful experimental system. The

new calcofluor based method for cereal �-glucan detection may also be a helpful tool in this

undertaking.

Attempts to apply the calcofluor method for the detection of cereal �-glucan in blood have so

far been hindered by fluorescence interference of serum. However, the use of appropriate

sample preparation techniques in order to remove serum fluorescence may render the

calcofluor method useful for quantification of cereal �-glucan also in serum samples.

Compared to the LAL test for �-glucans, the calcofluor method has a higher detection limit

and can only be applied for cereal �-glucans. On the other hand, the main advantage of the

calcofluor method is the specificity of the changes induced in the emission spectra of

calcofluor by cereal �-glucan binding. Activation of the limulus coagulation factor G depends

on �-glucan structure (lower activity for cereal �-glucans than yeast �-glucan and curdlan),

molecular weight and conformation in solution [123]. Higher MW and single helix

conformation of �-glucans in solution are associated with higher reactivity with LAL [124].

Due to this dependency of the LAL test specific standard curves for each tested �-glucan are

required. Furthermore, MW and conformation in solution of the tested �-glucans have to

remain unchanged under study conditions in order to obtain exact concentrations with the

LAL test.

8.3 Macrophages

Macrophages play an important role in tissue homeostasis by clearance of apoptotic cells,

tissue remodeling and repair [125]. Activation of macrophages during infection/inflammation

plays a crucial role for pathogen clearance both by the innate and adaptive immune systems

Page 48: Dietary immune-modulation – carbohydrate specific effects ...

46

[28]. Therefore the ability of �-glucans to activate macrophages has been preferentially

studied. Reports on the activation of macrophages by �-glucans include the activation of

phagocytosis [78, 126], increased secretion of cytokines [73, 126-128] and increased

production of ROS [54, 129]. The activation of cytokine secretion and ROS production have

been found to be dependent on the macrophage origin and micro-environment as well as on

the nature of the �-glucan (particulate vs. soluble) [24, 54, 126, 128]. However, even though

dectin-1 mediated cytokine secretion by macrophages has been shown to require crosslinking

of the receptor by particulate �-glucan [54], soluble oat �-glucan has been reported to increase

the secretion of IL-1 by murine peritoneal macrophages and the murine macrophage cell line

P338D1 [73]. This indicates that other mechanisms for cytokine secretion by �-glucans

besides dectin-1 signaling may exist.

We have studied the secretion of the cytokines IL-8, TNF-� and IL-1� by THP-1 derived

macrophages in response to different cereal and yeast �-glucan preparations (Paper VI). The

�-glucan samples that significantly increased cytokine secretion also contained the highest

levels of LPS (3.5-7 ng/mL). The high cytokine secretion levels induced by the LPS

containing samples also showed a relatively high variability, which may be the reason why

none of the �-glucan samples with minimal LPS contamination (0.01 to 0.04 ng/mL) had a

statistically significant effect on the cytokine secretion (Paper VI). The effect of oat and

barley �-glucan on cytokine gene expression by THP-1 derived macrophages has recently

been demonstrated with samples containing less than 1 pg LPS/mL [63]. Both extracted and

commercial cereal �-glucan samples increased the expression of the cytokines IL-1� and IL-8

and the expression of transcription factor NF-�B after 3-6 h of incubation, while the

expression of IL-10 was only upregulated after 24 h of incubation [63]. Statistical significance

testing of the observed increases compared to the control was unfortunately not performed in

this study. However, oat and barley �-glucans were shown to significantly decrease the LPS

Page 49: Dietary immune-modulation – carbohydrate specific effects ...

47

induced IL-1� expression by THP-1 derived macrophages [63]. LPS induced IL-8 expression

was also reduced, however this effect was not significant [63]. In order to investigate if the

increased cytokine secretion in response to the �-glucan preparations in our own study (Paper

VI) was due to contamination with LPS, results obtained with E.coli derived LPS were used

to build models for the prediction of the LPS-induced secretion of IL-8, TNF-� and IL-1�.

These regression analyses revealed that several �-glucan preparations gave a considerably

lower cytokine secretion than expected based on their LPS content. This indicates that �-

glucan may have a dampening effect on LPS-induced cytokine secretion from macrophages,

which is in accordance with the data obtained by Chanput et al. [63]. Furthermore, two studies

have suggested that �-glucans may protect from shock and organ injury during sepsis [96,

130]. Both studies used yeast �-glucan preparations and showed an attenuated cytokine

release in the �-glucan treated group of mice, which may have contributed to the observed

protection from lung, renal and hepatic injury [96, 130]. Also oral administration of yeast �-

glucan has been shown to attenuate the increase of plasma IL-6 and TNF-� in pigs challenged

with parenteral LPS [131]. However, more research is needed to understand the effect of

different �-glucans on the cytokine secretion from macrophages in the presence or absence of

inflammation.

The majority of our work with �-glucans and cell cultures has been conducted with different

intestinal epithelial cell lines and cereal �-glucan preparations. Hence, the applied �-glucan

dose of 1 mg/mL has been chosen based on the recommended intake for cereal �-glucan of 3

g/day and 0.75 g/serving [14]. A cereal �-glucan intake of 0.75 g/serving will with an

estimated stomach volume of 750 mL result in a cereal �-glucan concentration of 1 mg/mL in

the intestine. This concentration is therefore physiologically relevant for the interaction of

cereal �-glucan with IEC. Macrophages, however, are located in the subepithelial space and

will only be exposed to the �-glucan fraction that might be taken up across the epithelial

Page 50: Dietary immune-modulation – carbohydrate specific effects ...

48

layer. As outlined above, the uptake of �-glucans from the gut is still debated. However, the

few available studies on �-glucan uptake all show a very low bioavailability of �-glucans.

Hence, the concentration of 1 mg/mL which we have used for the experiments with

macrophages is probably too high in a physiological context. A dramatic reduction in �-

glucan dosage down to concentrations around 10 μg/mL would better reflect the probable in

vivo situation.

8.4 Carbohydrate specific effects

The assessment of immune-modulating properties of food or medicinal plant components

requires extremely pure preparations as co-extracted compounds or contaminants may have a

profound effect on the tested parameters, especially in in vitro test systems. One of the most

dreaded contaminants of test samples for biological activity testing is LPS, a membrane

molecule of gram negative bacteria. LPS binding to TLR-4 initiates a range of signaling

events that can lead to the activation of several transcription factors such as NF-�B and AP-1,

which may further lead to the secretion of pro-inflammatory cytokines [36]. In addition, LPS

is known to increase phagocytosis and nitric oxide production by e.g macrophages [28, 132].

Thus, the cellular responses towards LPS and immune-modulating food/plant components

may be difficult to distinguish from each other at first glance.

The addition of the antibiotic Polymyxin B to cell cultures is commonly used to eliminate the

effect of possible LPS contaminations [73, 106-108]. Polymyxin B binds to the Lipid A part

of the LPS molecule [133] thereby inhibiting its biological activity [134]. However, we have

shown that Polymyxin B was unable to suppress the activity of LPS found in a cereal �-

glucan preparation (Paper V). The activity of LPS in this sample could, however, be

suppressed by the addition of Polymyxin B after extensive �-glucan depolymerization into

oligosaccharides with lichenase. This indicates that polymeric �-glucan samples may complex

Page 51: Dietary immune-modulation – carbohydrate specific effects ...

49

LPS and make it unavailable for binding by Polymyxin B (Paper V). Therefore, an important

conclusion from our studies is that the sole use of Polymyxin B is not sufficient to rule out

LPS contamination of polymeric samples.

In Paper V we showed that targeted enzymatic degradation can be a good strategy to assess

the carbohydrate specific effect of polymeric samples. This approach has the main advantage

of attempting to detect a direct effect of the polymer of interest and can therefore both address

the possible effect of a potential LPS contamination and also exclude the potential

contribution of other sample components like for instance co-extracted compounds (Paper V).

The application of this approach to samples intended for cell culture testing, however, is not

without challenge as some enzyme preparations have been shown to influence cell viability

(Paper V). Membrane filtration of the enzyme preparations that removed their low molecular

compounds with negative effects on the cells but retained enzymatic activity was successfully

applied to overcome this challenge (Paper V).

In a different approach we tested the ability of a TLR-4 blocking antibody to suppress LPS

activity, although without success (Paper V). Even though the same antibody clone has been

previously described to reduce the effect of LPS on cytokine secretion by various cell types

[135-137], we did not observe any effect on LPS-induced IL-8 secretion by HT-29 cells

(Paper V, unpublished data). HT-29 cells have been reported to contain considerable amounts

of intracellular TLR-4 protein while cell surface expression is low [138], which may explain

the failure of the TLR-4 antibody to block LPS-induced cytokine secretion from HT-29 cells.

However, none of the other three studies that showed a suppression of LPS activity following

addition of the TLR-4 antibody could report a complete abrogation of the LPS effect [135-

137]. The combined use of different TLR-4 antibodies, binding to different epitopes of the

receptor, may increase the blocking potential. Alternatively, the use of specific cell lines that

do not express TLR-4 may be a possibility.

Page 52: Dietary immune-modulation – carbohydrate specific effects ...

50

In a third approach, we used E.coli derived LPS in concentrations corresponding to the range

of the contamination levels of the �-glucan samples to compare LPS and �-glucan effects

(Paper V and VI). Simple comparison of the effects obtained with LPS and �-glucan can give

a good first indication if observed effects may be due to LPS contamination in the sample. In

Paper VI we used regression analysis to further evaluate if the observed increase in cytokine

secretion by macrophages in response to �-glucan preparations was due to their LPS content.

We found that several �-glucan preparations gave a considerably lower cytokine secretion

than expected based on their LPS content. This indicates a potential dampening effect of �-

glucan on LPS induced cytokine secretion from macrophages. Even though more data is

needed to verify this hypothesis, regression analysis of cytokine secretion evoked by LPS

standards may become an interesting tool to investigate the effect of test samples in spite of

LPS contamination.

Our results clearly show that LPS contamination of �-glucan samples is a real problem for in

vitro test systems and has to be carefully evaluated in order to exclude false positive results.

However, we did find that the effect of particulate yeast �-glucan (MacroGard) on cytokine

secretion by different IEC systems was related to the �-glucan component of this preparation.

Others have also reported immune-modulating properties of �-glucan preparations with very

low LPS contamination (10 pg/mg or lower) [54, 63, 73, 78, 91, 92]. It is therefore unlikely

that the immune-modulating properties of �-glucan preparations shown in animal studies are

only caused by LPS contaminations.

A huge array of different LPS detection methods have been developed [102, 105, 139-142].

However, assays based on the ability of LPS to activate clotting enzymes of the haemolymph

of the horseshoe crab Limulus amoebocyte [141, 143] are preferentially used due to their high

sensitivity and easy application as several commercial assay kits based on the Limulus

amoebocyte lysate (LAL) are available. LAL also contains a factor, factor G, that reacts with

Page 53: Dietary immune-modulation – carbohydrate specific effects ...

51

�-glucans (from yeasts, mushrooms and cereals) in a clotting reaction similar to the one

induced by LPS [123, 124]. Commercial test kits where factor G has been removed are

available and can be used to quantify LPS in �-glucan samples. However, the solubility of �-

glucan preparations is sometimes limited and aggregate formation between polymeric �-

glucan and LPS may occur. To what extent this may influence the LAL test can only be

speculated upon. In our experiments we could detect considerable amounts of LPS (7 ng/mg)

in the 40 kDa cereal �-glucan samples, even though Polymyxin B was unable to suppress the

effect of 40 kDa on cytokine secretion (Paper VI). One may therefore speculate that complex

formation between �-glucan and LPS has less influence on the LAL test than on Polymyxin B

binding. Repeated use of the LAL test kit and a critical evaluation of the results revealed a

high overall standard deviation of 36%, which may be partly related to solubility/availability

problems of LPS in polymeric �-glucan samples or due to the fact that small amounts of

factor G, which reacts with �-glucan were not completely removed by the manufacturer.

However, the fact that some of the analyzed �-glucan samples (Wellmune and 123 kDa)

showed very low LPS contents (0.01 ng/mg) makes it unlikely that traces of factor G in the

assay may have caused considerable false positive results in the other �-glucan preparations.

An interesting hypothesis that exposure to LPS can have beneficial effects for human health

has recently been put forward [103, 144, 145]. LPS is supposed to play a role in homeostasis

of the immune system of the intestine [145]. Furthermore, Inagawa et al. have summarized

several animal studies that show a beneficial effect of oral LPS administration on various

infectious diseases [103]. Oral administration of 20 ng/mL LPS in the drinking water, for

example, was shown to protect mice from Toxoplasma gondii infection [146]. Interestingly,

LPS exposure or oral administration has also been shown to have some anti-carcinogenic

potential [144]. On the basis of this potential health effect of LPS it may be speculated that �-

glucan preparations act as LPS carriers in vivo and thereby mediated some of their reported

Page 54: Dietary immune-modulation – carbohydrate specific effects ...

52

beneficial effects. Furthermore, the LPS doses that can be achieved by LPS contaminated �-

glucan preparations (based on our results for LPS content in different �-glucan preparations)

are in the same range as the LPS doses (20 ng/mL) that have been reported to be effective in

animal studies [146].

8.5 Beta-glucans structure-function relationship

The anti-tumor activity of fungal �-glucans has been shown to depend on their DB, MW and

helical conformation [51]. Furthermore, fungal �-glucans with a higher DB, higher MW and a

single rather than triple helical conformation in solution have been shown to be more potent

activators of LAL [124]. Cereal �-glucans behave mostly like random coil polysaccharides in

solution [48]. However, they can vary considerably in MW and fine structure. The ratio of �-

(1,3)-linked cellotriosyl to cellotetraosyl units for example varies for �-glucans from different

cereals [46]. The cereal �-glucan preparations we used did not only vary in Mw but also

showed differences in fine structure as revealed by the different ratio of oligosaccharides with

degree of polymerization (DP) of 3 and 4 released by lichenase (Paper V and Table 2).

Table 2: Molar ratio of oligosaccharides released by lichenase treatment of different �-glucan preparations as determined by high performance anion exchange chromatography with pulsed amperometic detection (described in Paper V).

�eta-glucan standard DP3/DP4 molar ratio 40 kDa 1.78 123 kDa 1.92 245 kDa 2.80 359 kDa 3.19

As described before, none of the tested cereal �-glucan preparations, regardless of their Mw or

fine structure, showed any effect on cytokine secretion in the different IEC line test systems

that could be related to the �-glucan component of the sample (Paper III; V and VI).

Furthermore, �-glucan fractions isolated from barley showed no effect on basal or LPS

induced NF-�B activation in monocytic U937 cells stably transfected with a NF-�B reporter

Page 55: Dietary immune-modulation – carbohydrate specific effects ...

53

gene (Paper II). Nevertheless, two of the tested cereal �-glucan preparations, 359 kDa and

BG_STS (extracted from barley with an approximate Mw of 600 kDa), showed a potential to

dampen LPS induced cytokine secretion by macrophages (Paper VI). However, more data is

needed to verify this hypothesis and investigate a possible structure-function relationship of

cereal �-glucans in this respect. Interestingly, �-glucan fractions extracted from different

barley varieties were found to activate complement (Paper II), but differences in complement

fixing ability could not be ascribed to MW or fine structure (Paper II).

Interestingly, two particulate yeast �-glucans (MacroGard and Zymosan) significantly

increased IL-8 secretion by HT-29 and Caco-2 cells in mono- and co-culture, while a soluble

yeast �-glucan preparation (Wellmune) elicited no effect in the same test systems (Paper V

and VI). Since cereal �-glucans are largely soluble in water and have been used in a soluble

form in this study, it might be speculated that the physical properties (soluble vs. particulate)

more than the chemical structure (DB, MW, type of linkages) of different �-glucans

determine their specific activity. The recent finding that the known �-glucan receptor dectin-1

can only be activated by particulate �-glucans may strengthen this hypothesis [54]. However,

structure-function relationships of �-glucans are likely to depend on the studied immune

activation pathway, cellular or animal model system, involved �-glucan receptors, and way of

administration (oral or parenteral). Thus the complex relationships between �-glucans

chemical and physical properties and their immune-modulating potential are still far from

understood. A more general discussion of cereal �-glucans structure-function relationship in

immune modulation involving more results from other studies can be found in Paper I.

8.6 Current knowledge of dietary �-glucans mechanisms of action

Cereal as well as baker`s yeast (Saccharomyces cerevisiae) �-glucans are a part of the human

diet. Both types of �-glucans have been shown to be effective against different types of

Page 56: Dietary immune-modulation – carbohydrate specific effects ...

54

infection [74, 91, 92, 147-151] and they have been shown to enhance the activity of anti-

tumor antibodies in different animal models [17-21, 152]. The mechanisms of action behind

this synergistic effect in animal models of tumor therapy have been partially elucidated. Beta-

glucan binding to the lectin-side of CR3 on neutrophils and NK cells induces a primed state of

the receptor, which enhances the killing of iC3b opsonized target cells [19, 21, 81]. This

increases antibody dependent cellular cytotoxicity, which in combination with the activation

of complement deposition on tumor cells initiated by anti-tumor antibodies facilitates tumor

killing [19, 21].

It is possible that the anti-infective properties of cereal and yeast �-glucans partially rely on a

similar mechanism, which increases killing of complement coated pathogens. However, this

has never been shown experimentally. Apart from a potential microbiota based mechanism,

which has not been addressed in this thesis, dietary �-glucans can initiate immune-modulation

either by interacting with specific cells of the GI tract or by uptake across the intestinal

epithelium. We have demonstrated the potential of particulate yeast �-glucans to increase IL-8

secretion by IEC lines, which may also be an important mechanism for dietary yeast �-

glucans immune-modulating effect. On the other hand, our data do not indicate any effect of

cereal �-glucans on cytokine secretion by IEC nor uptake of cereal �-glucans across IEC

(Paper II, III, V, VI and additional data presented together with Paper IV). However,

interaction of dietary cereal �-glucans with IEL or uptake over the intestinal epithelium via M

cells and subsequent activation of macrophages could be other possible explanations for the

protective effect of cereal �-glucans demonstrated in animal models and should be further

investigated. Another possibility is the activation of DC that protrude their dendrites through

the epithelial lining in the gut by cereal or yeast �-glucans, as both types of �-glucans have

been shown to activate DC to express the co-stimulatory molecule CD86 in vitro [15].

Page 57: Dietary immune-modulation – carbohydrate specific effects ...

55

We also found indications for a dampening effect of some cereal and yeast �-glucan

preparations on LPS induced cytokine secretion by macrophages (Paper VI). Increased

phagocytic activity and cytokine secretion by macrophages in response to cereal and yeast �-

glucans have been repeatedly demonstrated in vitro [63, 73, 78, 153]. It seems therefore, that

macrophages may play an important role in mediating �-glucans immune-modulating effects.

However, more research is needed to understand the protective effects that cereal and yeast �-

glucans have shown against various diseases in animal models. A more detailed knowledge of

the potential mechanism behind the activity of these two types of �-glucans deducted from

animal and in vitro models will undoubtedly be helpful for the subsequent conduction of

meaningful human studies. So far, dietary yeast �-glucans have been reported to increase

salivary IgA concentrations in healthy volunteers and protected against exercise induced

depletion of monocytes in healthy subjects [154, 155]. Oral application of cereal �-glucan on

the other hand did not show any effect on C-reactive protein level or LPS induced cytokine

secretion by PBMC in hyper-cholesterolemic subjects or exercise induced immune-changes of

trained cyclists [156-158].

Page 58: Dietary immune-modulation – carbohydrate specific effects ...

56

9. Concluding Remarks

We have shown that cereal �-glucans of different origin, fine structure and molecular weight

are unable to modulate cytokine secretion by several IEC line systems including a co-culture

system of differentiated Caco-2 cells with THP-1 derived human macrophages. Furthermore,

the absence of cereal �-glucan uptake across differentiated Caco-2 cell monolayers indicated

that IEC may not play a major role in cereal �-glucan uptake from the intestine in vivo. The

developed method for cereal �-glucan quantification in cell cultures, based on the interaction

of cereal �-glucans with the fluorescent dye calcofluor, may be a useful tool to study cereal �-

glucan uptake in other cellular systems e.g. in vitro M cell models. Despite the

unresponsiveness of IEC line systems towards cereal �-glucans, these polysaccharides were

found to activate complement via the classical pathway and showed a potential to reduce LPS

induced cytokine secretion by THP-1 derived macrophages.

We have shown that particulate, but not soluble yeast �-glucans increase IL-8 secretion by

IEC lines, including basolateral secretion of IL-8 by differentiated Caco-2 cell mono-cultures.

Increased IL-8 secretion by IEC in vivo may be an important mechanism for the immune-

modulating effect of dietary yeast �-glucans.

We clearly showed the necessity to validate observed immune-modulating properties of

carbohydrate preparations from plants/fungi by investigating their carbohydrate specific

effects. To avoid false positive results a relationship between the carbohydrate

polymer/structure and the observed effect has to be established. We have demonstrated that

the use of Polymyxin B is insufficient to rule out LPS contamination of polymeric samples

like �-glucans. Instead targeted enzymatic degradation of the component of interest is

recommended since this method has been proven to be a powerful tool to assess possible

Page 59: Dietary immune-modulation – carbohydrate specific effects ...

57

carbohydrate specific effects and hence to validate results obtained with polysaccharide

preparations.

Page 60: Dietary immune-modulation – carbohydrate specific effects ...

58

10. Reference List

[1] Nordic Nutrition Recommendations 2004. Nordic Council of Ministers, Copenhagen 2004. [2] Phillips, G. O., Cui, S. W., An introduction: Evolution and finalisation of the regulatory definition of dietary fibre. Food Hydrocolloids 2011, 25, 139-143. [3] Collins, H. M., Burton, R. A., Topping, D. L., Liao, M. L., et al., Variability in Fine Structures of Noncellulosic Cell Wall Polysaccharides from Cereal Grains: Potential Importance in Human Health and Nutrition. Cereal Chem. 2010, 87, 272-282. [4] Lupton, J. R., From Basic Science to Dietary Guidance: Dietary Fiber as an Example. J. Food Drug Anal. 2012, 20, 346-349. [5] Aune, D., Chan, D. S. M., Lau, R., Vieira, R., et al., Dietary fibre, whole grains, and risk of colorectal cancer: systematic review and dose-response meta-analysis of prospective studies. Br. Med. J. 2011, 343. [6] Lim, C. C., Ferguson, L. R., Tannock, G. W., Dietary fibres as "prebiotics": Implications for colorectal cancer. Mol. Nutr. Food Res. 2005, 49, 609-619. [7] Young, G. P., Hu, Y., Le Leu, R. K., Nyskohus, L., Dietary fibre and colorectal cancer: A model for environment - gene interactions. Mol. Nutr. Food Res. 2005, 49, 571-584. [8] Kaczmarczyk, M. M., Miller, M. J., Freund, G. G., The health benefits of dietary fiber: Beyond the usual suspects of type 2 diabetes mellitus, cardiovascular disease and colon cancer. Metab.-Clin. Exp. 2012, 61, 1058-1066. [9] Wismar, R., Brix, S., Frokiaer, H., Laerke, H. N., in: Gershwin, M. E., Greenwood, M. R. C. (Eds.), Foods for Health in the 21st Century: A Road Map for the Future, Blackwell Publishing, Oxford 2010, pp. 70-85. [10] de Vrese, M., Schrezenmeir, J., in: Stahl, U., Donalies, U. E. B., Nevoigt, E. (Eds.), Food Biotechnology, Springer-Verlag Berlin, Berlin 2008, pp. 1-66. [11] Vos, A. P., M'Rabet, L., Stahl, B., Boehm, G., Garssen, J., Immune-modulatory effects and potential working mechanisms of orally applied nondigestible carbohydrates. Critical Reviews in Immunology 2007, 27, 97-140. [12] AbuMweis, S. S., Jew, S., Ames, N. P., beta-glucan from barley and its lipid-lowering capacity: a meta-analysis of randomized, controlled trials. Eur. J. Clin. Nutr. 2010, 64, 1472-1480. [13] Wolever, T. M. S., Tosh, S. M., Gibbs, A. L., Brand-Miller, J., et al., Physicochemical properties of oat beta-glucan influence its ability to reduce serum LDL cholesterol in humans: a randomized clinical trial. Am. J. Clin. Nutr. 2010, 92, 723-732. [14] EFSA, Scientific Opinion on the substantiation of a health claim related to oat beta-glucan and lowering blood cholesterol and reduced risk of (coronary) heart disease pursuant to Article 14 of Regulation (EC) No 1924/2006. EFSA Journal 2010, 8, 15. [15] Wismar, R., Brix, S., Laerke, H. N., Frokiaer, H., Comparative analysis of a large panel of non-starch polysaccharides reveals structures with selective regulatory properties in dendritic cells. Mol. Nutr. Food Res. 2011, 55, 443-454. [16] Thompson, I. J., Oyston, P. C. F., Williamson, D. E., Potential of the beta-glucans to enhance innate resistance to biological agents. Expert Rev. Anti-Infect. Ther. 2010, 8, 339-352. [17] Cheung, N. K. V., Modak, S., Oral (1 -> 3),(1 -> 4)-beta-D-glucan synergizes with antiganglioside GD2 monoclonal antibody 3F8 in the therapy of neuroblastoma. Clin. Cancer Res. 2002, 8, 1217-1223. [18] Cheung, N. K. V., Modak, S., Vickers, A., Knuckles, B., Orally administered beta-glucans enhance anti-tumor effects of monoclonal antibodies. Cancer Immunology Immunotherapy 2002, 51, 557-564. [19] Hong, F., Yan, J., Baran, J. T., Allendorf, D. J., et al., Mechanism by which orally administered beta-1,3-glucans enhance the tumoricidal activity of antitumor monoclonal antibodies in murine tumor models. Journal of Immunology 2004, 173, 797-806. [20] Modak, S., Koehne, G., Vickers, A., O'Reilly, R. J., Cheung, N. K. V., Rituximab therapy of lymphoma is enhanced by orally administered (1 -> 3),(l -> 4)-D-beta-glucan. Leuk. Res. 2005, 29, 679-683.

Page 61: Dietary immune-modulation – carbohydrate specific effects ...

59

[21] Qi, C. J., Cai, Y. H., Gunn, L., Ding, C. L., et al., Differential pathways regulating innate and adaptive antitumor immune responses by particulate and soluble yeast-derived beta-glucans. Blood 2011, 117, 6825-6836. [22] Wichers, H., Immunomodulation by food: promising concept for mitigating allergic disease? Anal. Bioanal. Chem. 2009, 395, 37-45. [23] Rieder, A., Samuelsen, A. B., Do cereal mixed-linked ss-glucans possess immune-modulating activities? Mol. Nutr. Food Res. 2012, 56, 536-547. [24] Goodridge, H. S., Wolf, A. J., Underhill, D. M., beta-glucan recognition by the innate immune system. Immunol. Rev. 2009, 230, 38-50. [25] Reid, D. M., Gow, N. A. R., Brown, G. D., Pattern recognition: recent insights from Dectin-1. Curr. Opin. Immunol. 2009, 21, 30-37. [26] Dranoff, G., Cytokines in cancer pathogenesis and cancer therapy. Nat. Rev. Cancer 2004, 4, 11-22. [27] Vilcek, J., The cytokines: an overview. In: Thomson, A. W., Lotze, M.T. (Ed.), The Cytokine Handbook, Elsevier, London 2003, 3-18. [28] Murphy, K., Travers, P., Walter, P., Janeway`s Immunobiology, Garland Sciences, Taylor & Francis Group, LLC, New York and London 2008. [29] Mukaida, N., Ketlinsky, S.A., Matsushima, K.,Interleukin-8 and other CXC chemokines. In: Thomson, A. W., Lotze, M.T. (Ed.), The Cytokine Handbook, Elsevier, London 2003, 1049-1070. [30] Sonnier, D. I., Bailey, S. R., Schuster, R. M., Lentsch, A. B., Pritts, T. A., TNF-alpha Induces Vectorial Secretion of IL-8 in Caco-2 Cells. J. Gastrointest. Surg. 2010, 14, 1592-1598. [31] Sturm, A., Baumgart, D. C., d'Heureuse, J. H., Hotz, A., et al., CXCL8 modulates human intestinal epithelial cells through a CXCR1 dependent pathway. Cytokine 2005, 29, 42-48. [32] Kucharzik, T., Hudson, J. T., Lugering, A., Abbas, J. A., et al., Acute induction of human IL-8 production by intestinal epithelium triggers neutrophil infiltration without mucosal injury. Gut 2005, 54, 1565-1572. [33] Kucharzik, T., Williams, I. R., Neutrophil migration across the intestinal epithelial barrier - Summary of in vitro data and description of a new transgenic mouse model with doxycycline-inducible interleukin-8 expression in intestinal epithelial cells. Pathobiology 2002, 70, 143-149. [34] Heidemann, J., Ogawa, H., Dwinell, M. B., Rafiee, P., et al., Angiogenic effects of interleukin 8 (CXCL8) in human intestinal microvascular endothelial cells are mediated by CXCR2. Journal of Biological Chemistry 2003, 278, 8508-8515. [35] Wang, H., Czura, C.J., Tracey, K.J., Tumor necrosis factor. In: Thomson, A. W., Lotze, M.T. (Ed.), The Cytokine Handbook, Elsevier, London 2003, 837-853. [36] Rosso, M., Heine, H., Meusch, U., Quandt, D., et al., LPS-Induced Cytokine Production in Human Monocytes and Macrophages. Critical Reviews in Immunology 2011, 31, 379-446. [37] Gebert, A., Rothkotter, H. J., Pabst, R., M cells in Peyer's patches of the intestine. Int.Rev.Cytol. 1996, 167, 91-159. [38] Mueller, C., Macpherson, A. J., Layers of mutualism with commensal bacteria protect us from intestinal inflammation. Gut 2006, 55, 276-284. [39] Artis, D., Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nat. Rev. Immunol. 2008, 8, 411-420. [40] Lavelle, E. C., Murphy, C., O'Neill, L. A. J., Creagh, E. M., The role of TLRs, NLRs, and RLRs in mucosal innate immunity and homeostasis. Mucosal Immunol. 2010, 3, 17-28. [41] Lotz, M., Gutle, D., Walther, S., Menard, S., et al., Postnatal acquisition of endotoxin tolerance in intestinal epithelial cells. J. Exp. Med. 2006, 203, 973-984. [42] Rimoldi, M., Chieppa, M., Salucci, V., Avogadri, F., et al., Intestinal immune homeostasis is regulated by the crosstalk between epithelial cells and dendritic cells. Nat. Immunol. 2005, 6, 507-514. [43] Sanderson, I. R., Dietary modulation of GALT. J. Nutr. 2007, 137, 2557S-2562S. [44] Volman, J. J., Ramakers, J. D., Plat, J., Dietary modulation of immune function by beta-glucans. Physiol. Behav. 2008, 94, 276-284.

Page 62: Dietary immune-modulation – carbohydrate specific effects ...

60

[45] Wood, P. J., Weisz, J., Blackwell, B. A., Structural studies of (1-]3),(1-]4)-beta-D-glucans by C (13)-nuclear magnetic-resonance spectroscopy and by rapid analysis of cellulose-like regions using high-performance anion-exchange chromatorgraphy of oligosaccharides release by lichenase. Cereal Chem. 1994, 71, 301-307. [46] Lazaridou, A., Biliaderis, C. G., Izydorczyk, M. S., Cereal beta-glucans: Structures, Physical Properties, and Physiological Functions. In: Biliaderis, C. G., Izydorczyk, M. S. (Eds.), Functional Food Carbohydrates, CRC Press, Boca Raton 2007, 1-72. [47] Ulmius, M., Onning, G., Nilsson, L., Solution behavior of barley beta-glucan as studied with asymmetrical flow field-flow fractionation. Food Hydrocolloids 2012, 26, 175-180. [48] Wood, P. J., Relationships between solution properties of cereal beta-glucans and physiological effects - a review. Trends Food Sci. Technol. 2004, 15, 313-320. [49] Pang, Z. C., Otaka, K., Maoka, T., Hidaka, K., et al., Structure of ss-glucan oligomer from laminarin and its effect on human monocytes to inhibit the proliferation of U937 cells. Biosci. Biotechnol. Biochem. 2005, 69, 553-558. [50] Ohno, N.,Yeast and Fungal Polysaccharides. In: Kamerling, J. P. (Ed.), Comprehensive Glycoscience - From Chemistry to Systems Biology, Elsevier, Oxford 2007, 559-577. [51] Bohn, J. A., BeMiller, J. N., (1->3)-beta-D-glucans as biological response modifiers: A review of structure-functional activity relationships. Carbohydrate Polymers 1995, 28, 3-14. [52] Falch, B. H., Espevik, T., Ryan, L., Stokke, B. T., The cytokine stimulating activity of (1 -> 3)-beta-D-glucans is dependent on the triple helix conformation. Carbohydr. Res. 2000, 329, 587-596. [53] Zhang, L., Li, X. L., Xu, X. J., Zeng, F. B., Correlation between antitumor activity, molecular weight, and conformation of lentinan. Carbohydr. Res. 2005, 340, 1515-1521. [54] Goodridge, H. S., Reyes, C. N., Becker, C. A., Katsumoto, T. R., et al., Activation of the innate immune receptor Dectin-1 upon formation of a 'phagocytic synapse'. Nature 2011, 472, 471-U541. [55] Brown, G. D., Herre, J., Williams, D. L., Willment, J. A., et al., Dectin-1 mediates the biological effects of beta-glucans. J. Exp. Med. 2003, 197, 1119-1124. [56] Dillon, S., Agrawal, S., Banerjee, K., Letterio, J., et al., Yeast zymosan, a stimulus for TLR2 and dectin-1, induces regulatory antigen-presenting cells and immunological tolerance. J. Clin. Invest. 2006, 116, 916-928. [57] Gantner, B. N., Simmons, R. M., Canavera, S. J., Akira, S., Underhill, D. M., Collaborative induction of inflammatory responses by dectin-1 and toll-like receptor 2. J. Exp. Med. 2003, 197, 1107-1117. [58] Ikeda, Y., Adachi, Y., Ishii, T., Miura, N., et al., Dissociation of toll-like receptor 2-mediated innate immune response to zymosan by organic solvent-treatment without loss of dectin-1 reactivity. Biol. Pharmacol. Bull. 2008, 31, 13-18. [59] Juul-Madsen, H. R., Norup, L., Laerke, H. N., Modulation of the immune response of porcine neutrophils by different beta-glucan preparations. Livest. Sci. 2010, 133, 249-252. [60] Li, B., Allendorf, D. J., Hansen, R., Marroquin, J., et al., Yeast beta-glucan amplifies phagocyte killing of iC3b-opsonized tumor cells via complement receptor 3-Syk-phosphatidylinositol 3-kinase pathway. Journal of Immunology 2006, 177, 1661-1669. [61] Rice, P. J., Adams, E. L., Ozment-Skelton, T., Gonzalez, A. J., et al., Oral delivery and gastrointestinal absorption of soluble glucans stimulate increased resistance to infectious challenge. J. Pharmacol. Exp. Ther. 2005, 314, 1079-1086. [62] Rice, P. J., Lockhart, B. E., Barker, L. A., Adams, E. L., et al., Phannacokinetics of fungal (1-3)-beta-D-glucans following intravenous administration in rats. International Immunopharmacology 2004, 4, 1209-1215. [63] Chanput, W., Reitsma, M., Kleinjans, L., Mes, J. J., et al., Beta-glucans are involved in immune-modulation of THP-1 macrophages. Mol. Nutr. Food Res. 2012, 56, 822-833. [64] Kupfahl, C., Geginat, G., Hof, H., Lentinan has a stimulatory effect on innate and adaptive immunity against murine Listeria monocytogenes infection. International Immunopharmacology 2006, 6, 686-696.

Page 63: Dietary immune-modulation – carbohydrate specific effects ...

61

[65] Yamada, J., Hamuro, J., Hatanaka, H., Hamabata, K., Kinoshita, S., Alleviation of seasonal allergic symptoms with superfine beta-1-glucan: A randomized study. J. Allergy Clin. Immunol. 2007, 119, 1119-1126. [66] Hashimoto, K., Suzuki, I., Yadomae, T., Oral administration of SSG, a beta-glucan obtained from Sclerotinia-Sclerotiorum, affects the function of Peyers patch cells. Int. J. Immunopharmacol. 1991, 13, 437-442. [67] Inomata, T., Goodman, G. B., Fryer, C. J. H., Chaplin, D. J., et al., Immune reaction induced by X-rays and pions and its stimulation by schizophyllan (SPG). Br. J. Cancer 1996, 74, S122-S125. [68] Kakumu, S., Ishikawa, T., Wakita, T., Yoshioka, K., et al., Effect of Sizofiran, a polysaccharide, on interferon gamma, antiblody-production and lymphocyte-proliferation specific for hepatitis-B virus antigen in patients with chronic hepatitisB. Int. J. Immunopharmacol. 1991, 13, 969-975. [69] LeibundGut-Landmann, S., Gross, O., Robinson, M. J., Osorio, F., et al., Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17. Nat. Immunol. 2007, 8, 630-638. [70] LeibundGut-Landmann, S., Osorio, F., Brown, G. D., Sousa, C. R. E., Stimulation of dendritic cells via the dectin-1/Syk pathway allows priming of cytotoxic T-cell responses. Blood 2008, 112, 4971-4980. [71] Osorio, F., LeibundGut-Landmann, S., Lochner, M., Lahl, K., et al., DC activated via dectin-1 convert Treg into IL-17 producers. Eur. J. Immunol. 2008, 38, 3274-3281. [72] Direnzo, L., Yefenof, E., Klein, E., The function of human NK cells is enhanced by beta-glucan a ligang of CR3 (CD11B/CD18). Eur. J. Immunol. 1991, 21, 1755-1758. [73] Estrada, A., Yun, C. H., Van Kessel, A., Li, B., et al., Immunomodulatory activities of oat beta-glucan in vitro and in vivo. Microbiol. Immunol. 1997, 41, 991-998. [74] Murphy, E. A., Davis, J. M., Brown, A. S., Carmichael, M. D., et al., Benefits of oat beta-glucan on respiratory infection following exercise stress: role of lung macrophages. Am. J. Physiol.-Regul. Integr. Comp. Physiol. 2008, 294, R1593-R1599. [75] Rieder, A., Grimmer, S., Kolset, S. O., Michaelsen, T. E., Knutsen, S. H., Cereal beta-glucan preparations of different weight average molecular weights induce variable cytokine secretion in human intestinal epithelial cell lines. Food Chem. 2011, 128, 1037-1043. [76] Samuelsen, A. B., Rieder, A., Grimmer, S., Michaelsen, T. E., Knutsen, S. H., Immunomodulatory Activity of Dietary Fiber: Arabinoxylan and Mixed-Linked Beta-Glucan Isolated from Barley Show Modest Activities in Vitro. Int. J. Mol. Sci. 2011, 12, 570-587. [77] Volman, J. J., Mensink, R. P., Ramakers, J. D., de Winther, M. P., et al., Dietary (1 -> 3), (1 -> 4)-beta-D-glucans from oat activate nuclear factor-kappa B in intestinal leukocytes and enterocytes from mice. Nutr. Res. 2010, 30, 40-48. [78] Yun, C. H., Estrada, A., Van Kessel, A., Park, B. C., Laarveld, B., Beta-glucan, extracted from oat, enhances disease resistance against bacterial and parasitic infections. FEMS Immunol. Med. Microbiol. 2003, 35, 67-75. [79] Brown, G. D., Dectin-1: a signalling non-TLR pattern-recognition receptor. Nat. Rev. Immunol. 2006, 6, 33-43. [80] Brown, G. D., Gordon, S., Fungal beta-glucans and mammalian immunity. Immunity 2003, 19, 311-315. [81] Vetvicka, V., Thornton, B. P., Ross, G. D., Soluble beta-glucan polysaccharide binding to the lectin site of neutrophil or natural killer cell complement receptor type 3 (CD11b/CD18) generates a primed state of the receptor capable of mediating cytotoxicity of iC3b-opsonized target cells. J. Clin. Invest. 1996, 98, 50-61. [82] Wakshull, E., Brunke-Reese, D., Lindermuth, J., Fisette, L., et al., PGG-Glucan, a soluble beta-(1,3)-glucan, enhances the oxidative burst response, microbicidal activity, and activates an NF-kappa B-like factor in human PMN: Evidence for a glycosphingolipid beta-(1,3)-glucan receptor. Immunopharmacology 1999, 41, 89-107.

Page 64: Dietary immune-modulation – carbohydrate specific effects ...

62

[83] Evans, S. E., Hahn, P. Y., McCann, F., Kottom, T. J., et al., Pneumocystis cell wall beta-glucans stimulate alveolar epithelial cell chemokine generation through nuclear factor-kappa B-dependent mechanisms. Am. J. Respir. Cell Mol. Biol. 2005, 32, 490-497. [84] Hahn, P. Y., Evans, S. E., Kottom, T. J., Standing, J. E., et al., Pneumocystis carinii cell wall beta-glucan induces release of macrophage inflammatory protein-2 from alveolar epithelial cells via a lactosylceramide-mediated mechanism. Journal of Biological Chemistry 2003, 278, 2043-2050. [85] Rice, P. J., Kelley, J. L., Kogan, G., Ensley, H. E., et al., Human monocyte scavenger receptors are pattern recognition receptors for (1 -> 3)-beta-D-glucans. J. Leukoc. Biol. 2002, 72, 140-146. [86] Vera, J., Fenutria, R., Canadas, O., Figueras, M., et al., The CD5 ectodomain interacts with conserved fungal cell wall components and protects from zymosan-induced septic shock-like syndrome. Proc. Natl. Acad. Sci. U. S. A. 2009, 106, 1506-1511. [87] Murphy, E. A., Davis, J. M., Brown, A. S., Carmichael, M. D., et al., Oat beta-glucan effects on neutrophil respiratory burst activity following exercise. Med. Sci. Sports Exerc. 2007, 39, 639-644. [88] Suzuki, I., Hashimoto, K., Ohno, N., Tanaka, H., Yadomae, T., immunomodulation by orally-administred beta-glucan in mice. Int. J. Immunopharmacol. 1989, 11, 761-769. [89] Suzuki, I., Tanaka, H., Kinoshita, A., Oikawa, S., et al., Effect of orally-administered beta-glucan on macrophage function in mice. Int. J. Immunopharmacol. 1990, 12, 675-684. [90] Tsukada, C., Yokoyama, H., Miyaji, C., Ishimoto, Y., et al., Immunopotentiation of intraepithelial lymphocytes in the intestine by oral administrations of beta-glucan. Cell. Immunol. 2003, 221, 1-5. [91] Yun, C. H., Estrada, A., Van Kessel, A., Gajadhar, A., et al., Immunomodulatory effects of oat beta-glucan administered intragastrically or parenterally on mice infected with Eimeria vermiformis. Microbiol. Immunol. 1998, 42, 457-465. [92] Yun, C. H., Estrada, A., VanKessel, A., Gajadhar, A. A., et al., beta-(1->3, 1->4) oat glucan enhances resistance to Eimeria vermiformis infection in immunosuppressed mice. Int. J. Parasit. 1997, 27, 329-337. [93] Hughes, S. A., Shewry, P. R., Gibson, G. R., McCleary, B. V., Rastall, R. A., In vitro fermentation of oat and barley derived beta-glucans by human faecal microbiota. FEMS Microbiol. Ecol. 2008, 64, 482-493. [94] Jonathan, M. C., van den Borne, J., van Wiechen, P., da Silva, C. S., et al., In vitro fermentation of 12 dietary fibres by faecal inoculum from pigs and humans. Food Chem. 2012, 133, 889-897. [95] Gibson, G. R., Roberfroid, M. B., Dietary Modulation of the Human Colonic Microbiota - Introducing the Concept of Prebiotics. J. Nutr. 1995, 125, 1401-1412. [96] Sandvik, A., Wang, Y. Y., Morton, H. C., Aasen, A. O., et al., Oral and systemic administration of beta-glucan protects against lipopolysaccharide-induced shock and organ injury in rats. Clinical and Experimental Immunology 2007, 148, 168-177. [97] Zhang, M., Kim, J. A., Effect of molecular size and modification pattern on the internalization of water soluble beta-(1 -> 3)-(1 -> 4)-glucan by primary murine macrophages. Int. J. Biochem. Cell Biol. 2012, 44, 914-927. [98] Florence, A. T., The oral absorption of micro- and nanoparticulates: Neither exceptional nor unusual. Pharm. Res. 1997, 14, 259-266. [99] Mayer, L., Blumberg, R. S.,Role of Epithelial Cells in Mucosal Antigen Presentation. In: Mestecky, J., Lamm, M. E., McGhee, J. R., Bienenstock, J., et al. (Eds.), Mucosal Immunology, Elsevier 2005, 435-450. [100] Guha, M., Mackman, N., LPS induction of gene expression in human monocytes. Cell. Signal. 2001, 13, 85-94. [101] Lerouge, I., Vanderleyden, J., O-antigen structural variation: mechanisms and possible roles in animal/plant-microbe interactions. Fems Microbiol. Rev. 2002, 26, 17-47. [102] Lee, C. H., Tsai, C. M., Quantification of bacterial lipopolysaccharides by the purpald assay: Measuring formaldehyde generated from 2-keto-3-deoxyoctonate and heptose at the inner core by periodate oxidation. Anal. Biochem. 1999, 267, 161-168. [103] Inagawa, H., Kohchi, C., Soma, G. I., Oral Administration of Lipopolysaccharides for the Prevention of Various Diseases: Benefit and Usefulness. Anticancer Res. 2011, 31, 2431-2436.

Page 65: Dietary immune-modulation – carbohydrate specific effects ...

63

[104] Beutler, B., Toll-like receptors. In: Thomson, A. W., Lotze, M.T. (Ed.), The Cytokine Handbook, Elsevier, London 2003, 809-819 [105] de Santana, A. P., Noleto, G. R., Gorin, P. A. J., de Souza, L. M., et al., GC-MS detection and quantification of lipopolysaccharides in polysaccharides through 3-O-acetyl fatty acid methyl esters. Carbohydrate Polymers 2012, 87, 2730-2734. [106] Schepetkin, I. A., Quinn, M. T., Botanical polysaccharides: Macrophage immunomodulation and therapeutic potential. International Immunopharmacology 2006, 6, 317-333. [107] Bhattacharyya, S., Gill, R., Chen, M. L., Zhang, F., et al., Toll-like receptor 4 mediates induction of the Bcl10-NF kappa B-interleukin-8 inflammatory pathway by carrageenan in human intestinal epithelial cells. Journal of Biological Chemistry 2008, 283, 10550-10558. [108] Han, S. B., Yoon, Y. D., Ahn, H. J., Lee, H. S., et al., Toll-like receptor-mediated activation of B cells and macrophages by polysaccharide isolated from cell culture of Acanthopanax senticosus. International Immunopharmacology 2003, 3, 1301-1312. [109] Michaelsen, T. E., Gilje, A., Samuelsen, A. B., Hogasen, K., Paulsen, B. S., Interaction between human complement and a pectin type polysaccharide fraction, PMII, from the leaves of Plantago major L. Scand. J. Immunol. 2000, 52, 483-490. [110] Samuelsen, A. B., Paulsen, B. S., Wold, J. K., Otsuka, H., et al., Characterization of a biologically active pectin from Plantago major L. Carbohydrate Polymers 1996, 30, 37-44. [111] Haller, D., Bode, C., Hammes, W. P., Pfeifer, A. M. A., et al., Non-pathogenic bacteria elicit a differential cytokine response by intestinal epithelial cell/leucocyte co-cultures. Gut 2000, 47, 79-87. [112] Ramakers, J. D., Volman, J. J., Biorklund, M., Onning, G., et al., Fecal water from ileostomic patients consuming oat beta-glucan enhances immune responses in enterocytes. Mol. Nutr. Food Res. 2007, 51, 211-220. [113] Volman, J. J., Mensink, R. P., Buurman, W. A., Plat, J., In vivo effects of dietary (1 -> 3), (1 -> 4)-beta-d-glucans from oat on mucosal immune responses in man and mice. Scand. J. Gastroenterol. 2011, 46, 603-610. [114] Neutra, M. R., Kraehenbuhl, J. P., Cellular and Molecular Basis for Antigen Transport Across Epithelial Barriers. In: Mestecky, J., Lamm, M. E., McGhee, J. R., Bienenstock, J., et al. (Eds.), Mucosal Immunology, Elsevier 2005, 111-130. [115] Artursson, P., Palm, K., Luthman, K., Caco-2 monolayers in experimental and theoretical predictions of drug transport. Adv. Drug Deliv. Rev. 2001, 46, 27-43. [116] Hidalgo, I. J., Raub, T. J., Borchardt, R. T., Characterization of the human-colon carcinoma cell-line (Caco-2) as a model system for intestinal epithelial permeability. Gastroenterology 1989, 96, 736-749. [117] Quaroni, A., Hochman, J., Development of intestinal cell culture models for drug transport and metabolism studies. Adv. Drug Deliv. Rev. 1996, 22, 3-52. [118] Linnankoski, J., Makela, J., Palmgren, J., Mauriala, T., et al., Paracellular Porosity and Pore Size of the Human Intestinal Epithelium in Tissue and Cell Culture Models. J. Pharm. Sci. 2010, 99, 2166-2175. [119] Moyes, S. M., Morris, J. F., Carr, K. E., Macrophages increase microparticle uptake by enterocyte-like Caco-2 cell monolayers. J. Anat. 2010, 217, 740-754. [120] Pellegrina, C. D., Perbellini, O., Scupoli, M. T., Tomelleri, C., et al., Effects of wheat germ agglutinin on human gastrointestinal epithelium: Insights from an experimental model of immune/epithelial cell interaction. Toxicol. Appl. Pharmacol. 2009, 237, 146-153. [121] Lyu, S. Y., Park, W. B., Transport of mistletoe lectin by M cells in human intestinal follicle-associated epithelium (FAE) In vitro. Arch. Pharm. Res. 2008, 31, 1613-1621. [122] Roberts, C. L., Keita, A. V., Duncan, S. H., O'Kennedy, N., et al., Translocation of Crohn's disease Escherichia coli across M-cells: contrasting effects of soluble plant fibres and emulsifiers. Gut 2010, 59, 1331-1339. [123] Tanaka, S., Aketagawa, J., Takahashi, S., Shibata, Y., et al., Activation of a limulus coagulation factor-G by (1-3)-beta-D-glucans. Carbohydr. Res. 1991, 218, 167-174.

Page 66: Dietary immune-modulation – carbohydrate specific effects ...

64

[124] Ohno, N., Emori, Y., Yadomae, T., Saito, K., et al., Reactivity of limulus amoebocyte lysate towards (1,3)-beta-D-glucans. Carbohydr. Res. 1990, 207, 311-318. [125] Gordon, S., Taylor, P. R., Monocyte and macrophage heterogeneity. Nat. Rev. Immunol. 2005, 5, 953-964. [126] Berner, M. D., Sura, M. E., Alves, B. N., Hunter, K. W., IFN-gamma primes macrophages for enhanced TNF-alpha expression in response to stimulatory and non-stimulatory amounts of microparticulate beta-glucan. Immunol. Lett. 2005, 98, 115-122. [127] Kataoka, K., Muta, T., Yamazaki, S., Takeshige, K., Activation of macrophages by linear (1 -> 3)-beta-D-glucans - Implications for the recognition of fungi by innate immunity. Journal of Biological Chemistry 2002, 277, 36825-36831. [128] Li, B., Cramer, D., Wagner, S., Hansen, R., et al., Yeast glucan particles activate murine resident macrophages to secrete proinflammatory cytokines via MyD88- and Syk kinase-dependent pathways. Clin. Immunol. 2007, 124, 170-181. [129] Underhill, D. M., Rossnagle, E., Lowell, C. A., Simmons, R. M., Dectin-1 activates Syk tyrosine kinase in a dynamic subset of macrophages for reactive oxygen production. Blood 2005, 106, 2543-2550. [130] Bedirli, A., Kerem, M., Pasaoglu, H., Akyurek, N., et al., Beta-glucan attenuates inflammatory cytokine release and prevents acute lung injury in an experimental model of sepsis. Shock 2007, 27, 397-401. [131] Li, J., Xing, J. J., Li, D. F., Wang, X., et al., Effects of beta-glucan extracted from Saccharomyces cerevisiae on humoral and cellular immunity in weaned piglets. Arch. Anim. Nutr. 2005, 59, 303-312. [132] Sun, X., Lv, Z., Peng, H., Fung, M., et al., Effects of a recombinant schistosomal-derived anti-inflammatory molecular (rSj16) on the lipopolysaccharide (LPS)-induced activated RAW264.7. Parasitol. Res. 2012, 110, 2429-2437. [133] Morrison, D. C., Jacobs, D. M., Binding of Polymyxin B to Lipid-A portion of bacterial Lipopolysaccharides. Immunochemistry 1976, 13, 813-818. [134] Tsuzuki, H., Tani, T., Ueyama, H., Kodama, M., Lipopolysaccharide: Neutralization by polymyxin B shuts down the signaling pathway of nuclear factor kappa B in peripheral blood mononuclear cells, even during activation. J. Surg. Res. 2001, 100, 127-134. [135] Akashi, S., Ogata, H., Kirikae, F., Kirikae, T., et al., Regulatory roles for CD14 and phosphatidylinositol in the signaling via toll-like receptor 4-MD-2. Biochemical and Biophysical Research Communications 2000, 268, 172-177. [136] Shimazu, R., Akashi, S., Ogata, H., Nagai, Y., et al., MD-2, a molecule that confers lipopolysaccharide responsiveness on Toll-like receptor 4. J. Exp. Med. 1999, 189, 1777-1782. [137] Yonekawa, K., Neidhart, M., Altwegg, L. A., Wyss, C. A., et al., Myeloid related proteins activate Toll-like receptor 4 in human acute coronary syndromes. Atherosclerosis 2011, 218, 486-492. [138] Suzuki, M., Hisamatsu, T., Podolsky, D. K., Gamma interferon augments the intracellular pathway for lipopolysaccharide (LPS) recognition in human intestinal epithelial cells through coordinated up-regulation of LPS uptake and expression of the intracellular toll-like receptor 4-MD-2 complex. Infection and Immunity 2003, 71, 3503-3511. [139] Hsu, Y. H., Fu, S. L., Detection of endotoxin contamination in chinese herbs by NF-kappa B activity-based reporter assays. J. Food Drug Anal. 2004, 12, 34-39. [140] Lin, H. H., Huang, S. P., Hsieh, H. C., Chen, C. S., Chen, Y. L., Performance characteristics of the limulus amebocyte lysate assay and gas chromatography-mass spectrum analysis of lipopolysaccharides relative to nitric oxide production by peritoneal exudates of cells. J. Hazard. Mater. 2007, 145, 431-436. [141] Park, C. Y., Jung, S. H., Bak, J. P., Lee, S. S., Rhee, D. K., Comparison of the rabbit pyrogen test and Limulus amoebocyte lysate (LAL) assay for endotoxin in hepatitis B vaccines and the effect of aluminum hydroxide. Biologicals 2005, 33, 145-151. [142] Peters, M., Fritz, P., Bufe, A., A bioassay for determination of lipopolysaccharide in environmental samples. Innate Immun. 2012, 18, 694-699.

Page 67: Dietary immune-modulation – carbohydrate specific effects ...

65

[143] Gutsmann, T., Howe, J., Zahringer, U., Garidel, P., et al., Structural prerequisites for endotoxic activity in the Limulus test as compared to cytokine production in mononuclear cells. Innate Immun. 2010, 16, 39-47. [144] Lundin, J. I., Checkoway, H., Endotoxin and Cancer. Environ. Health Perspect. 2009, 117, 1344-1350. [145] Taniguchi, Y., Yoshioka, N., Nakata, K., Nishizawa, T., et al., Mechanism for Maintaining Homeostasis in the Immune System of the Intestine. Anticancer Res. 2009, 29, 4855-4860. [146] Suzuki, Y., Kobayashi, A., Nishizawa, T., Inagawa, H., et al., Homeostasis as regulated by activated macrophage.6. Protective effect of LPSW (a lipopolysaccharide from wehat-flour) against acute infection by Toxoplasma gondii in mice. Chem. Pharm. Bull. 1992, 40, 1266-1267. [147] Breivik, T., Opstad, P. K., Engstad, R., Gundersen, G., et al., Soluble beta-1,3/1,6-glucan from yeast inhibits experimental periodontal disease in Wistar rats. J. Clin. Periodontol. 2005, 32, 347-352. [148] Davis, J. M., Murphy, E. A., Brown, A. S., Carmichael, M. D., et al., Effects of oat beta-glucan on innate immunity and infection after exercise stress. Med. Sci. Sports Exerc. 2004, 36, 1321-1327. [149] Jung, K., Ha, Y., Ha, S. K., Han, D. U., et al., Antiviral effect of Saccharomyces cerevisiae beta-glucan to swine influenza virus by increased production of interferon-gamma and nitric oxide. J. Vet. Med. Ser. B-Infect. Dis. Vet. Public Health 2004, 51, 72-76. [150] Stuyven, E., Cox, E., Vancaeneghem, S., Arnouts, S., et al., Effect of beta-glucans on an ETEC infection in piglets. Vet. Immunol. Immunopathol. 2009, 128, 60-66. [151] Vetvicka, V., Terayama, K., Mandeville, R., Brousseau, P., et al., Pilot Study: Orally-Administred Yeast beta-1,3-glucan Prophylactically Protects Against Anthrax Infection and Cancer in Mice. Journal of the American Neutraceutical Association 2002, 5, 5-9. [152] Li, B., Cai, Y. H., Qi, C. J., Hansen, R., et al., Orally Administered Particulate beta-Glucan Modulates Tumor-Capturing Dendritic Cells and Improves Antitumor T-Cell Responses in Cancer. Clin. Cancer Res. 2010, 16, 5153-5164. [153] Vetvicka, V., Vetvickova, J., Effects of yeast-derived beta-glucans on blood cholesterol and macrophage functionality. J. Immunotoxicol. 2009, 6, 30-35. [154] Carpenter, K. C., Breslin, W. L., Davidson, T., Adams, A., McFarlin, B. K., Baker`s yeast beta-glucan supplementation increases monocytes and cytokines post-exercise: implications for infection risk? Br. J. Nutr. 2012. [155] Lehne, G., Haneberg, B., Gaustad, P., Johansen, P. W., et al., Oral administration of a new soluble branched beta-1,3-D-glucan is well tolerated and can lead to increased salivary concentrations of immunoglobulin A in healthy volunteers. Clinical and Experimental Immunology 2006, 143, 65-69. [156] Nieman, D. C., Henson, D. A., McMahon, M., Wrieden, J. L., et al., Beta-glucan, immune function, and upper respiratory tract infections in athletes. Med. Sci. Sports Exerc. 2008, 40, 1463-1471. [157] Queenan, K. M., Stewart, M.L., Smith, K.N., Thomas, W., Fulcher, R.G., Slavin, J.L., Concentrated oat beta-glucan, a fermentable fibre, lowers serum cholesterol in hypercholesterolemic adults in a randomized controlled trial. Nutr. J. 2007, 6. [158] Theuwissen, E., Plat, J., Mensink, R. P., Consumption of oat beta-glucan with or without plant stanols did not influence inflammatory markers in hypercholesterolemic subjects. Mol. Nutr. Food Res. 2009, 53, 370-376.

Page 68: Dietary immune-modulation – carbohydrate specific effects ...
Page 69: Dietary immune-modulation – carbohydrate specific effects ...

I

Page 70: Dietary immune-modulation – carbohydrate specific effects ...
Page 71: Dietary immune-modulation – carbohydrate specific effects ...

II

Page 72: Dietary immune-modulation – carbohydrate specific effects ...
Page 73: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12, 570-587; doi:10.3390/ijms12010570�

International Journal of

Molecular Sciences ISSN 1422-0067

www.mdpi.org/ijms Article

Immunomodulatory Activity of Dietary Fiber: Arabinoxylan and Mixed-Linked Beta-Glucan Isolated from Barley Show Modest Activities in Vitro

Anne Berit Samuelsen 1,*, Anne Rieder 2, Stine Grimmer 2, Terje E. Michaelsen 1,3 and Svein H. Knutsen 2

1 Pharmacognosy, Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, Oslo N-0316, Norway

2 Nofima Mat, Norwegian Institute of Food, Fisheries and Aquaculture Research, Aas N-1430, Norway; E-Mails: [email protected] (A.R.); [email protected] (S.G.); [email protected] (S.H.K.)

3 Norwegian Institute of Public Health, Oslo N-0403, Norway; E-Mail: [email protected]

* Author to whom correspondence should be addressed; E-Mail: [email protected]; Tel.: +47-22-856-568; Fax: +47-22-854-402.

Received: 26 November 2010; in revised form: 20 December 2010 / Accepted: 4 January 2011 / Published: 18 January 2011

Abstract: High intake of dietary fiber is claimed to protect against development of colorectal cancer. Barley is a rich source of dietary fiber, and possible immunomodulatory effects of barley polysaccharides might explain a potential protective effect. Dietary fiber was isolated by extraction and enzyme treatment. A mixed-linked �-glucan (WSM-TPX, 96.5% �-glucan, Mw 886 kDa), an arabinoxylan (WUM-BS-LA, 96.4% arabinoxylan, Mw 156 kDa), a mixed-linked �-glucan rich fraction containing 10% arabinoxylan (WSM-TP) and an arabinoxylan rich fraction containing 30% mixed-linked �-glucan (WUM-BS) showed no significant effect on IL-8 secretion and proliferation of two intestinal epithelial cell lines, Caco-2 and HT-29, and had no significant effect on the NF-�B activity in the monocytic cell line U937-3�B-LUC. Further enriched arabinoxylan fractions (WUM-BS-LA) from different barley varieties (Tyra, NK96300, SB94897 and CDCGainer) were less active than the mixed-linked �-glucan rich fractions (WSM-TP and WSM-TPX) in the complement-fixing test. The mixed-linked �-glucan rich fraction from NK96300 and CDCGainer showed similar activities as the positive control while mixed-linked �-glucan rich fractions from Tyra and SB94897 were less active. From these

OPEN ACCESS

Page 74: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

571

results it is concluded that the isolated high molecular weight mixed-linked �-glucans and arabinoxylans from barley show low immunological responses in selected in vitro test systems and thus possible anti-colon cancer effects of barley dietary fiber cannot be explained by our observations.

Keywords: arabinoxylan; mixed-linked �-glucan; barley; Caco-2; complement-fixing test; dietary fiber; HT-29; IL-8; U937; NF-kappaB

1. Introduction

Dietary fiber have been claimed to protect against the development of colorectal cancer (CRC) [1], but according to several reviews, evidence of such a relationship is scarce [2–4]. CRC is one of the most common types of cancer world-wide, and also in Norway, the incidence of CRC has increased over the past 50 years. The reason for this is largely unknown, but lifestyle and diet probably contribute [5,6].

Chronic inflammation is associated with increased risk of cancer development [7], and patients with inflammatory bowel diseases, such as ulcerative colitis and Crohn’s disease, have increased risk of developing CRCs [8]. Plasma levels of the acute phase protein C-Reactive Protein (CRP) which is a marker of inflammation, are elevated in persons who subsequently develop CRC [9]. Increased intake of dietary fiber reduces CRP levels [10–12] as well as the levels of the proinflammatory cytokines IL-6 and TNF� [13]. Strengthening the immune system’s ability to detect and eliminate cancer cells, a process called cancer immunosurveillance [14], on the other hand may have a protective effect. The potential of dietary fiber to promote cancer immunosurveillance is currently unknown. However barley beta-glucan has been shown to increase the effect of anti-tumor antibodies in mice [15,16]. In general, dietary fiber may affect inflammatory processes and immune responses by several mechanisms. Amongst the most studied are the mechanisms exerted by butyrate, a short chain fatty acid produced in the colon following fermentation of dietary fiber. Butyrate has anti-inflammatory [17], apoptotic, and anti-proliferative activities on cancer cells [18,19]. Dietary fiber, depending on their structures, can affect the intestinal immune system by being taken up by M-cells in the Peyer’s patches and transported to underlying immune cells and other cells. This may result in a local cytokine production which can influence T-cells, B-cells, antigen presenting cells and other immune cells. Fiber may also be taken up by intestinal macrophages or dendritic cells (i.e., antigen presenting cells) and transported to lymph nodes, spleen and bone marrow [20,21]. In addition, direct interaction of fiber with colonic epithelial cells or leukocytes may induce changes in immune reactions relevant for inflammation and the development of cancer.

Barley (Hordeum vulgare) is an interesting source of dietary fiber and was previously the preferred grain for food in the Nordic region; mainly due to its short growing season due to the climate. In Norway, barley is still the major cereal crop, but only a part is used for human consumption, the majority is used as animal feed. Barley as well as oats are rich in dietary fiber, mainly mixed-linked �-glucans and arabinoxylans [22,23]. In these cereals, �-glucans are linear �-(1�3)/(1�4)-D-glucopyranosyl polymers referred to as mixed- linked or cereal �-glucans [24].

Page 75: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

572

Most of the previous studies on immunomodulatory activities of barley dietary fiber have focused on the mixed-linked �-glucans since they are structurally related to fungal and yeast �-glucans that are �-(1�3)-D-glucopyranosyl polymers with �-(1�6) linked side chains. In vitro and in vivo experiments on �-glucan preparations from yeast and fungi have shown immunomodulating properties and a potential to increase host resistance against infections [20]. Mixed-linked �-glucans from barley might have similar effects, although knowledge on immunomodulatory effects of barley polysaccharides is quite limited. Some activities have been reported on commercially available barley �-glucan; Intra peritoneal injections of barley �-glucan into fish enhanced the leukocyte count, phagocytic activity, lysozyme activity, complement activity via the alternative pathway and serum bactericidal activity [25]. Czop and Austen [26] found that turbid preparations of barley �-glucan activate the alternative pathway of the complement system in vitro. In addition, pre-treatment of human monocytes with barley �-glucan inhibited phagocytosis of zymosan particles [26]. �-glucans enhance cytotoxicity of phagocytes or NK cells towards iC3b-opsonized cells by binding to the lectin site on complement receptor 3 (CR3 or CD11b/CD18, Mac-1, �M�2 integrin) and thereby initiate cytotoxic degranulation of NK cells and phagocytosis by other cells [27–29]. Oral administered barley �-glucan increased the efficacy of photodynamic therapy of Lewis lung carcinoma in mice through binding to CR3 [27], but barley �-glucan binds to CR3 with lower affinity than yeast �-glucan [29,30]. Barley �-glucans also enhance the anti-tumor effect of monoclonal antibodies in mice when administered orally [16,31] by being taken up by gastrointestinal macrophages, transported to the spleen, lymph nodes and bone marrow where smaller fragments of glucan are bound to CR3 on granulocytes which in turn kill iC3b-opsonized tumor cells [15]. Barley �-glucan can also bind to Dectin-1[32] and activate NF-�B when Dectin-1, Syk, CARD9 and Bcl10 are co-expressed in the cells [33]. The transcription factor NF-�B plays a critical role in immune, cellular stress and inflammatory responses [34].

Instead of using commercially available dietary fiber from barley in the present study we isolated fiber fraction from barley, both mixed-linked �-glucan and arabinoxylan and tested for immunomodulatory activities related to inflammation. This involved extraction and the use of specific hydrolytic enzymes to isolate pure polysaccharide fractions and determination of biological activities by stimulation of the human colon epithelial cell lines Caco-2 and HT-29 followed by measurement of cell proliferation and cytokine secretion. In addition, we investigated the fiber`s ability to modulate NF-�B activity in monocytes and their influence on the complement system using the complement-fixing test [35], all systems involving factors with relevance to inflammatory processes.

2. Results and Discussion

2.1. Barley Dietary Fiber Fractions

�-glucan and arabinoxylan samples isolated and purified from the common Norwegian barley variety Tyra were the main basis for our investigations. As shown in Table 1, the constituent sugar analysis combined with 1H-NMR [23] (spectra not shown) revealed that WUM-BS contained 70% arabinoxylan and 30% mixed-linked �-glucan. Treatment with lichenase (L) and amyloglucosidase (A) efficiently removed most of the remaining mixed-linked �-glucan from this fraction; the enzyme treated fraction WUM-BS-LA contained 96.4% arabinoxylan. Trace amounts of mannose were

Page 76: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

573

attributed to the glycoprotein part of the enzyme preparation used. In this fraction, ferulic acid is not present due to alkali treatment during the extraction procedure.

Table 1. Characterization of fiber fractions from Hordeum vulgare var. Tyra: monosaccharide composition (mol %) and estimated molecular weight calculated on the basis of pullulan standards by GPC-SEC and refractive index detection, as weight average (Mw) and number average (Mn) of barley fiber fractions .

Monosaccharide Composition (mol%) Molecular Weight (kDa) Fraction Glc Man Xyl Ara Mw Mn WSM-TP 91.0 0.0 5.8 3.3 1090 599

WSM-TPX 96.5 0.0 1.9 1.6 886 501 WUM-BS 30.0 0.0 52.7 17.3 412 98

WUM-BS-LA 2.0 1.6 66.0 30.4 156 42 WSM-TP was composed of 90% mixed-linked �-glucan in addition to 10% co-extracted

arabinoxylan. Most of the arabinoxylan was removed by enzymatic treatment with xylanase (X). The enzyme treated fraction WSM-TPX was composed of 96.5% glucose and only 3.5% arabinoxylan.

The estimated relative weight average molecular weights (Mw) based on the pullulan series were about 886 and 156 kDa for WSM-TPX and WUM-BS-LA, respectively (Table 1). Molecular weight decreased during the enzymatic treatment of WSM-TP and WUM-BS from about 1090 and 412 kDa, respectively, giving samples with less polydispersities (Mw/Mn).

All previous studies on immunomodulatory activity of mixed-linked �-glucans from barley have been performed on commercially available samples. It should be noticed [20] that choice of isolation method may influence polysaccharide characteristics, such as molecular weight and solubility, and thereby their biological activities. In addition, co-extracted substances or contaminants of an endotoxin nature that may occur during isolation may contribute to significant activities in immunological test systems.

Potential degradation of dietary fiber during food processing has not been taken into account in this study. In addition, the fact that dietary fiber very seldom is eaten alone without subsequent intake of several other food constituents makes the picture quite complex and complicated to explore. In the present study, all fiber fractions had relatively high molecular weight after isolation, and no attempts were made to alter the chain length in either of the samples. This was because we primarily wanted to investigate intact carbohydrate dietary fibers with the presumption that dietary fiber remains undegraded until reaching the microflora in the colon.

2.2. Effect on IL-8 Secretion and Cell Proliferation in Caco-2 and HT-29

To test the inflammatory response of the fiber fractions on gut epithelial cells the modulation of IL-8 (CXCL8) secretion from the human intestinal epithelial cell lines Caco-2 and HT-29 cells was determined. The concentration used of 1 mg/mL is physiologically relevant as a concentration of 1 mg/mL barley fiber in the intestine corresponds to the consumption of approximately 20 g barley, an amount found, for example, in two slices of barley bread (40% barley flour). In addition, the potential toxic effect of the fiber fractions on the Caco-2 and HT-9 cells was determined by measuring the effect of the fiber fractions at different concentrations on cell proliferation using the MTT assay.

Page 77: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

574

We found that the fiber fractions had no significant effect on the cell proliferation of the human intestinal epithelial cell lines Caco-2 and HT-29 cells (Figure 1A, B). It was observed that the HT-29 cell line in general secretes considerably higher levels of IL-8 than Caco-2, but the barley fiber fractions had no significant effect on this secretion either from Caco-2 (Figure 2A) or HT-29 cells (Figure 2B). Only the positive controls, PMII and zymosan increased secretion of IL-8 from both cell lines significantly (p = 0.001) compared to the respective controls. Zymosan is a crude extract from yeast (Saccharomyces cerevisiae) and contains mainly �-glucan but also some mannan [20], protein, fat and chitin [36]. Immunomodulatory activities of �-glucan from yeast and from other sources have been studied extensively, for review on this topic see [37]. PMII is a pectic polysaccharide fraction isolated from Plantago major L. leaves, a plant used in traditional medicine to aid the healing of wounds. PMII has shown immunomodulatory activities both in vitro and in vivo: Increased resistance against bacterial infection in mice, activation of human monocytes and activation of the complement system [35,38,39]. PMII is therefore considered useful as positive control in immunological test systems.

Even though we did not find any direct effect of the barley fiber fractions on the intestinal epithelial cell model system, barley fiber may affect inflammatory processes and immune response by other mechanisms. As outlined in the introduction, barley fiber may be taken up by intestinal macrophages or M-cells and delivered to underlying immune cells where binding of barley �-glucan to the lectin site of CR3 on effector cells has been shown to enhance cytotoxic activity [15,27].

Figure 1. The effect of fiber fractions extracted from the barley variety Tyra on cell proliferation of (A) Caco-2 cells and (B) HT-29 cells. Cells were incubated with three different concentrations of the respective fiber fraction in cell culture medium for 24 hours before cell proliferation was measured. Each bar represents the mean of at least three experiments performed in triplicate (as % of medium control) ± SD.

Page 78: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

575

Figure 1. Cont.

Figure 2. IL-8 secretion from Caco-2 (A) and HT-29 (B) cells in response to treatment with fiber fractions extracted from the barley variety Tyra, zymosan and PMII (all 1 mg/mL). Cells were incubated with fiber of the respective fiber fractions in cell culture medium for 24 hours before IL-8 secretion was measured. Each bar represents the average ± SD of one representative experiment from a total of three independent experiments. * p < 0.05.

Page 79: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

576

Figure 2. Cont.

2.3. The Ability of the Fiber Fractions to Modulate NF-�B Activity in Monocytes

The nuclear transcription factor kappa B (NF-�B) plays a central role in inflammatory response [40]. Thus, to further study the effect of the fiber fractions on the immune response, the ability of the fiber fractions to modulate basal and LPS-induced NF-�B activity was tested using the U937-3�B-LUC monocytic cell line stably transfected with a luciferase reporter containing three NF-�B binding sites. It has been shown that this model system correlates well with in vivo NF-�B activity [41,42] Due to the limitations of the test system, lower concentrations of the samples (0.1, 0.2 and 0.4 mg/mL) were used compared to experiments with the Caco-2 and HT-29 cell lines. However, as the U937-3�B-LUC cell line is quite sensitive, the response is still considered relevant. The activities of the different fiber fractions were compared to the positive control, PMII [39]. Of the different fiber fractions only the highest concentration of WUM-BS had a significant effect on basal NF-�B activity (p = 0.004) (Figure 3A), giving an increase of the activity to 270% compared to control. The apparent dose response from 0.1 mg/mL to 0.4 mg/mL of all fractions of the basal NF-�B activity was statistically not significant compared to the control. However, all concentrations of PMII significantly increased basal NF-�B activity (p < 0.001) in the test system compared to the control. None of the fiber fractions had significant effect on the LPS-induced NF-�B activity, only the highest concentration of PMII increased the LPS-induced NF-�B activity significantly (p = 0.013) (Figure 3B).

Page 80: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

577

Figure 3. The effect of the fiber fractions extracted from the barley variety Tyra on basal (A) and LPS-induced (B) NF-�B activity. U937-3x�B-LUC cells were incubated with 0.1, 0.2 or 0.4 mg/mL as indicated of the respective fiber fraction in cell culture medium for 6.5 hours before luciferase activity was measured. For LPS-induction, 1 �g/mL LPS was added after 30 min, and the cells incubated further for six hours before the luciferase activity was measured. Each bar represents the mean of at least three experiments performed in triplicate ± SD. * p < 0.05.

A

B

Page 81: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

578

Only high concentrations of a fraction containing 70% arabinoxylan and 30% mixed-linked �-glucan (WUM-BS) increased the activity of the pro-inflammatory transcription factor NF-�B in monocytes. This fraction was obtained after alkaline extraction of a water insoluble residue. Neither pure arabinoxylan nor pure mixed-linked �-glucan was active in this test system. Some biological effects of �-glucans are initiated by binding to Dectin-1 on macrophages and dendritic cells. Barley �-glucan has previously been found to activate NF-�B when Dectin-1, Syk, SARD9 and Bcl10 were co-expressed in the cells [33], and it was concluded that Dectin-1 was involved in these activities. However, binding to Dectin-1 requires �-glucans with a minimum of 10- or 11- mer 1,3-linked glucose oligomers [43] which are structural elements not found in barley. Barley �-glucans only contain single 1,3-linked glucose units separating two or three 1,4-linked glucose oligomers [44]. Transcription factor NF-�B can be activated via many different pathways including proinflammatory cytokines, TLR activation, for example, by LPS and by T-cell activation [40]. As shown in Figure 3B, LPS induced NF-�B activity was not significantly altered by any of the barley fractions indicating that the basal activity observed after stimulation of 0.4 mg/mL WUM-BS may be due to contamination by LPS. In any case, the activity found in Figure 3A cannot be due to either arabinoxylan or mixed-linked �-glucan since other fractions containing higher levels of mixed-linked �-glucan (WSM-TP and WSM-TPX) and arabinoxylan (WUM-BS-LA) were inactive in the test system. The positive control PMII increased LPS induced activity but to a lesser extent than measured with PMII alone (Figure 3A). This shows that PMII is active per se, but confirms presence of LPS. Previously, it has been shown that PMII can activate monocytes and induce secretion of TNF� [39]. One might speculate that secreted TNF� in turn activate NF-�B [40], alternatively PMII may bind to NF-�B activating receptors directly.

2.4. Complement Fixing Test

Purified �-glucan (WSM-TPX) and arabinoxylan (WUM-BS-LA) from Tyra were tested for activity in the complement-fixing test. Both showed lower activities than the positive control, PMII. At 1 mg/mL WSM-TPX was significantly more active than WUM-BS-LA (p = 0.009) (Figure 4).

�-glucans and purified arabinoxylans from other barley varieties [45] were also subjected to this test. As shown in Figure 4, all arabinoxylan fractions (WUM-BS-LA) had relatively low activity compared to the positive control. Arabinoxylan isolated from Tyra, NK96300 and SB94897 had very similar activities; the one from CDC Gainer was almost inactive.

Starch-free mixed-linked �-glucans that had not been subjected to a xylanase treatment (WSM-TP) containing additional small amounts of arabinoxylan, had the highest activity in this test system. Such WSM-TP samples originating from CDC Gainer and NK96300 showed activity at the same level as the positive control, while similar fractions from Tyra and SB94897 were less active (p < 0.035). Figure 4 furthermore shows that a xylanase purification step of WSM-TP into purified mixed-linked �-glucan (WSM-TPX) did not alter the complement-fixing activity significantly.

In general, all mixed-linked �-glucan rich fractions had a significantly higher complement-fixing activity than the arabinoxylan-rich fractions (p < 0.027).

According to Figure 4 the fractions can be listed as follows with regard to decreasing activity in the complement fixing test: PMII = WSM-TP CDC Gainer = WSM-TP NK96300 > WSM-TP Tyra =

Page 82: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

579

WSM TPX Tyra = WSM TP SB94897 > WUM BS-LA Tyra = WUM-BS-LA NK96300 = WUM-BS-LA SB94897 > WUM-BS-LA CDC Gainer.

Figure 4. Complement fixing test of fiber fractions isolated from barley varieties. Each bar represents % activity (mean values of triplicates ± SD) of the positive control PMII measured at 1 mg/mL. The fractions are arabinoxylan (WUM-BS-LA) and mixed-linked �-glucan rich fractions (WSM-TP and WSM-TPX) from the barley varieties Tyra, NK96300, SB94897 and CDC Gainer. Activity bars denoted with the same letter (a, b or c) are not significantly different, p < 0.05.

Mixed-linked �-glucans from barley have previously shown to activate the complement system via the alternative pathway [26], the present study demonstrates an effect also on the classical pathway. The complement system provides a first line of protection against potential harmful invaders and is part of the innate immune system. It consists of a group of serum proteins that are activated in a cascade mechanism. Many of these proteins are pro-enzymes that are activated by proteolytical cleavage which in turn activate the next step in the cascade. Activation can be initiated by three pathways; the classical pathway, the alternative pathway or the lectin pathway, and is important for initiating inflammation, activation of leucocytes, lysis of target cells and opsonisation [46,47]. The test system employed has some limitations since it does not distinguish between activation and inhibition of the complement cascade, only a “consumption” of complement activity is registered. From previous studies however, it is established that PMII, the positive control, is an activator of the complement system [35], and it has also shown to protect against bacterial infection in vivo [38].

The mixed-linked �-glucan fractions tested were more active than the arabinoxylan fractions. The reason for the differences in activity of the different �-glucans might be due to differences in their primary structure. The ratio of (1�4)/(1�3) linkages present varies between the different barley varieties tested. NK96300 has the highest ratio (2.76) followed by CDC Gainer (2.59), Tyra (2.48) and SB94897 (2.30) [23]. The varieties with the highest (1�4)/(1�3) ratio have the highest activity in the complement fixing test, but statistical analysis shows no significant correlation between linkage ratio and activity or between molecular weight of the WSM-TP fractions and activity. Mw of WSM-TP

Page 83: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

580

fractions from NK96300, CDC Gainer and SB94897 were estimated to 1040, 1130 and 1040 kDa, respectively. The estimated molecular weights of the WSM-TP fractions were significantly higher than the corresponding arabinoxylan (WUM-BS-LA) fractions (p < 0.001). Mw of the WUM-BS-LA fractions from NK96300, CDC Gainer and SB94897 were estimated to 214, 203 and 190 kDa, respectively. The WUM-BS-LA fractions from different barley varieties tested had in general low activities in the complement fixing test. However, when both these arabinoxylan fractions and the more active �-glucan rich fractions were included in the statistical test a significant positive correlation was found between molecular weight and activity (p = 0.002). On the other hand, when these two classes of dietary fiber are evaluated separately there is no correlation between activity and their estimated molecular weights. Contamination of LPS does not affect this test system [48], so activity is not attributed to presence of endotoxin.

The arabinoxylans from different barley varieties tested had in general low activities in the complement fixing test leaving this hemicellulose type of dietary fiber non-responsive in all the test systems in the present study. To our knowledge, high molecular weight arabinoxylans have not been ascribed immunomodulatory activities. On the other hand arabinoxylan oligosaccharides have been studied for prebiotic properties [49] and have been shown to reduce preneoplastic lesions in the colon of rats treated with a carcinogen [50].

3. Experimental Section

3.1. Isolation of Fiber Fractions

Fiber fractions were extracted from four barley varieties; NK96300, Tyra, CDC Gainer and SB94897 basically as previously described [51]. Briefly; milled (0.5 mm) barley samples (48 g) were extracted and washed with boiling ethanol. This removed low molecular weight constituents and is promoting the denaturation of endogenous hydrolytic enzymes such as �-glucanase. Following defatting with hexane, extraction with boiling water gave a water soluble material (WSM) and a residue of water insoluble material (WUM). WSM was furthermore treated with 7 mL amylase (Termamyl 120 L, Type L, Novozymes ) and 75 mg protease (Porcine Pancreatine, SIGMA) filtered and recovered with alcohol precipitation resulting in the starch free fraction designated WSM-TP.

In an attempt to remove small amounts of co-extracted arabinoxylan, WSM-TP was treated with a xylanase. WSM-TP Tyra (1 g) was dissolved in sodium acetate buffer pH 4.5, and 10 μL (21 U) endo �-xylanase (�-xylanase M6, Megazyme) was added at 40 °C and left for 3 h with gentle stirring. Polysaccharide material was precipitated with isopropanol and centrifugated at 1000 × g for 10 min. The pellet was redissolved in water, dialyzed against distilled water using a dialyzing tube with cut off 12,000–14,000 (Medicell Int. Ltd); freeze dried and designated WSM-TPX.

Base soluble material (WUM-BS) was then extracted from the previous water insoluble residue (WUM) with 1 M NaOH added 1% NaBH4. Co-extracted mixed-linked �-glucans and starch were removed by adding 50 U lichenase (Lichenase EC 3.2.1.73, Megazyme) and 400 μL amyloglucosidase (Amyloglucosidase for Total Dietary Fiber Assay EC 3.2.1.3, SIGMA) as described elsewhere [51] giving fractions designated WUM-BS-LA.

Page 84: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

581

3.2. Monosaccharide Composition

Methanolysis combined with TMS-derivatisation and GC were performed according to the method of Chambers & Clamp [52] with modifications as previously described [53] using 4 M HCl in anhydrous methanol for 24 h at 80 °C.

3.3. 1H-NMR

1H-NMR spectra of selected samples were obtained on a Varian Mercury 300 system. Approximately 4 mg of freeze dried material was solubilized in 0.7 mL D2O, transferred to NMR glass tubes and acquired at 80 °C with typically 64 scans. Further details of the method are described in Knutsen & Holtekjolen [23] .

3.4. HPLC

GPC-SEC was performed using a DIONEX P680 pump with a Spectraphysics AS3500 auto injector and a Shimadzu RID6A refractive index detector controlled with Chromeleon 6.80 software. Serially connected Shodex OHPack SB-806-HQ and SB 804-HQ columns were connected to a Shodex OHPack SB-LG precolumn and eluted at 40 °C with 50 mM Na2SO4 (0.5 mL/min), and samples (1 mg/mL) were injected using a 100 μL loop. Relative molecular weight averages (Mw and Mn) were estimated offline by the software WINGPC �6.2 using pullulan molecular weight standards ranging from Mp 342 to 1,520,000 Da for calibration. Software and standards were obtained from PSS (Polymer Standards Service GmbH, Mainz, Germany).

3.5. Cell Cultures

The Caco-2 cell line (obtained from the American Type Culture Collection (ATCC), and a generous gift from Professor Kirsten Sandvig, Norwegian Radium Hospital) and HT-29 cell line (obtained from ATCC, and a generous gift from Professor Tor Lea, Norwegian University of Life Sciences) were grown in DMEM medium containing 10% fetal calf serum, 1% non-essential amino acids, 100 U/mL penicillin, and 100 mg/mL streptomycin. The U937-3xkB-LUC cell line (a generous gift from Professor Rune Blomhoff, University of Oslo) was grown in RPMI-1640 medium supplemented with 10% fetal bovine serum, 2 mM L-glutamine, 50 U/mL penicillin, 50 mg/mL streptomycin and 75 μg/mL hygromycin (Sigma-Aldrich, St. Louis, MO). The cells were maintained at 37 °C and 5% CO2 in a humidified incubator. If not otherwise stated, all solutions were obtained from Invitrogen (Carlstad, CA).

3.6. Measurement of Cell Proliferation and IL-8 Secretion

For IL-8 secretion, cells were plated in 12-well plates. For cell proliferation, cells were plated in 96-well plates. Cells were plated at a concentration of 1.0 × 105 cells/mL (Caco-2) and 1.5 × 105 cells/mL (HT-29) and incubated until they reached 80 % confluency (48 h). The fiber fractions were solubilized in water by boiling for 20 min, aliquoted, freeze dried and re-solubilized in growth medium to treatment concentrations of 0.5–3 mg/mL. A yeast derived beta-glucan (Zymosan A

Page 85: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

582

Z4250 Sigma) and PMII, a plant polysaccharide fraction from Plantago major L with known immune stimulating activity [35,38,39] were used as positive controls for IL-8 secretion. Cells were incubated 24 h with 1.5 mL (12-well plates) or 100 μL (96-well plates) growth medium or solubilized fiber fractions in duplicate (12-well plates) or triplicate (96-well plates). At the end of the incubation, the plates were processed for measurement of either cell proliferation or IL-8 secretion.

Cell proliferation was determined using the colorimetric MTT assay (Roche Diagnostics GmbH, Mannheim, Germany) that measures the ability of metabolic active cells to cleave tetrazolium sodium salt to purple formazan crystals [54]. The resulting purple precipitate in each cell was dissolved in 100 �L isopropanol containing 0.04 M HCl, and the absorbance measured at 562 nm using Titertek Multiscan plus MK II plate reader (Labsystems, Finland). IL-8 concentrations in the cell culture supernatants were determined using an enzyme linked immunosorbent assay (ELISA). Monoclonal mouse anti-human IL-8 antibody (BD Bioscience Pharmingen, San Diego, CA) suspended in coating buffer (0.1 M Carbonate/Bicarbonate buffer pH 9.6) was added to MaxiSorpTM ELISA plates (Nunc, Roskilde, Denmark) and incubated over night at 4 °C. Plates were washed three times with PBS containing 0.01% Tween-20 and unspecific binding-sites were blocked by incubating with 5% BSA in PBS for 1 h at room temperature. After washing five times with PBS-Tween, samples and human recombinant IL-8 standards (BD Bioscience Pharmingen,) diluted in working strength high performance ELISA (HPE) buffer from Sanquin (Amsterdam, Netherlands) were added to the plates, which were then incubated for 1.5 h at room temperature followed by washing five times with PBS-Tween. Plates were then incubated for 1 h with biotinylated mouse anti-human IL-8 monoclonal antibody (BD Bioscience Pharmingen) in HPE buffer. After another washing step streptavidin-horseradishperoxidase conjugate (BD Bioscience Pharmingen) in HPE buffer was added and incubated at room temperature for 30 min. Plates were then washed five times with 30 sec between each wash. Color developed after addition of 3,3’,5,5’-tetramethylbenzidine (Sigma-Aldrich) in 0.05 M Phosphate-Citrate-Buffer containing H2O2. After 10 min the reaction was stopped by addition of 1 M H2SO4, and absorbance was measured at 450 nm using the Titertek Multiscan plus MK II plate reader (Labsystems, Finland). The detection limit of the IL-8 ELISA was 2 pg/mL.

3.7. NF-kB Activity Assay

In order to measure NF-�B activity the U937-3x�B-LUC cell line were transferred to RPMI medium with 2 % fetal bovine serum and seeded out in 96 well plates. The fiber fractions extracted from the barley variety Tyra were mixed with highly purified water (Milli-Q, 18.2 M� to a final concentration of 1 mg/mL in Precellys CK14 homogenization tubes (Bertin Technologies, Montigny le Bretonneux, France), solubilized using the Precellys 24 homogenizer (Bertin Technologies) followed by boiling the samples for 5 min, and then freeze dried. The resulting freeze dried fiber fractions were dissolved in medium with 2% serum to a concentration of 4 mg/mL. PMII from Plantago major L. leaves was used as positive control [35,38,39]. This final solution of the fiber fractions was diluted directly in the wells giving the final concentrations of 0.1, 0.2 and 0.4 mg/mL. To measure basal NF-�B activity, cells were incubated with fiber fractions or vehicle control for 6.5 hours. To measure lipopolysaccharide (LPS)-induced NF-�B activity, cells were pre-incubated with fiber fractions or vehicle control for 30 min, then 1 μg/mL lipopolysaccharide isolated from E. coli 0111:B4

Page 86: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

583

(Sigma-Aldrich, St. Louis, MO, U.S.) was added to the cells and the incubation continued for 6 hours. Cell viability for these cells was determined with the use of the CellTiter-Glo Luminiscent Cell Viability Assay (Promega, Madison, WI, U.S.) with cut-off value of 10% non-viable cells. The NF-�B activity was determined by measuring the luciferase activity after addition of Bright-GloTM Reagent (Pomega, Madison, WI, U.S.) in accordance to the manufacturer’s instructions. Luminescence was detected for 1 sec using the Glomax96 Microplate Luminometer (Promega, Madison, WI, U.S.).

3.8. Complement Fixing Test

Human complement proteins were incubated with fiber fractions that might either activate or inhibit activation of the complement proteins. In both situations complement activity is depleted with a negative influence on a balanced hemolysis system involving antibody-sensitized sheep red blood cells and a human serum diluted to give 50% hemolysis. The degree of hemolysis was measured as absorbency at 405 nm. Fiber fractions were tested in triplicates using PMII, a polysaccharide fraction from Plantago major L. as positive control [35,39].

3.9. Statistics

Analysis of significant differences was tested by one-way analysis of variance (ANOVA) with Dunnett’s comparisons with a control and Pearson correlation analysis using Minitab Version 16. Differences were considered significant when p < 0.05.

4. Conclusions

From the experiments presented, it is concluded that purified high molecular weight mixed-linked �-glucans from barley have quite low immunological responses and do not affect proliferation and secretion of IL-8 of the colon epithelial cell lines Caco-2 and HT-29, or NF-kappaB activity in the monocytic cell line U937-3�B-LUC but are active in the complement-fixing test. High molecular weight barley arabinoxylans have neglectible activities in all test systems mentioned. Taken together the results do not support that barley dietary fiber protect against the development of CRC through the immune responses or inflammatory responses tested. Still, one cannot overrule that such effects may occur through other mechanisms that may be shown in other test systems.

Acknowledgements

The project was financed by funding from Infigut (NFR 185125). Novozymes Nordic is thanked for the supply of Termamyl. Ann-Katrin Holtekjølen is thanked for providing the fiber fractions from different barley varieties. The authors acknowledge Merete Rusås Jensen, Nofima Mat, Norwegian Institute of Food Fisheries and Aquaculture Research, for technical assistance.

References

1. Burkitt, D.P. Some diseases characteristic of modern westernised civilisation. Br. Med. J. 1973, 1, 274–278.

Page 87: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

584

2. Asano, T.K.; McLeod, R.S. Dietary fibre for the prevention of colorectal adenomas and carcinomas. Cochrane Database Syst. Rev. 2002, 1, CD003430.

3. WHO; FAO. Diet, Nutrition and the Prevention of Chronic Diseases. WHO Technical Report Series 916; WHO: Geneva, Switzerland, 2002.

4. Marshall, J.R. Nutrition and colon cancer prevention. Curr. Opin. Clin. Nutri. Metab. Care 2009, 12, 539–343.

5. Bray, F.; Wibe, A.; Dorum, L.M.R.; Møller, B. Tykktarms-og endetarmskreft i Norge-epidemiologi. J. Nor. Med. Assoc. 2007, 127, 2682–2687.

6. Kirkegaard, H.; Johnsen, N.F.; Christensen, J.; Frederiksen, K.; Overvad, K.; Tjønneland, A. Association of adherence to lifestyle recommendations and risk of colorectal cancer: a prospective Danish cohort study. BMJ 2010, 341, c5504.

7. Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899.

8. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. 9. Erlinger, T.P.; Platz, E.A.; Rifai, N.; Helzlsouer, K.J. C-Reactive Protein and the risk of incident

colorectal cancer. JAMA 2004, 291, 585–590. 10. King, D.E.; Egan, B.M.; Woolson, R.F.; Mainous, A.G.; A-Solaiman, Y.; Jesri, A. Effect of a

high-fiber diet vs. a fiber-supplemented diet on D-reactive protein level. Achives Intern. Med. 2007, 167, 502–506.

11. Ajani, U.A.; Ford, E.S.; Mokdad, A.H. Dietary fiber and C-reactive protein: findings from national health and nutrition examination survey data. J. Nutri. 2004, 134, 1181–1184.

12. Ma, Y.; Griffith, J.A.; Chasan-Taber, L.; Olendzki, B.C.; Jackson, E.; Stanek, E.J.; Li, W.; Pagoto, S.L.; Hafner, A.R.; Ockene, I.S. Association between dietary fiber and serum C-reactive protein. Am. J. Clin. Nutri. 2006, 83, 760–766.

13. Ma, Y.; Hebert, J.R.; Li, W.; Bertone-Johnson, E.R.; Olendzki, B.; Pagoto, S.L.; Tinker, L.; Rosal, M.C.; Ockene, I.S.; Ockene, J.K.; Griffith, J.A.; Liu, S. Association between dietary fiber and markers of systemic inflammation in the Women`s Health Initative Observational Study. Nutrition 2008, 24, 941–949.

14. Dunn, G.P.; Old, L.J.; Schreiber, R.D. The immunobiology of cancer immunosurveillance and immunoediting. Immunity 2004, 21, 137–148.

15. Hong, F.; Yan, J.; Baran, J.T.; Allendorf, D.J.; Hansen, R.D.; Ostroff, G.R.; Xing, P.X.; Cheung, N.-K.V.; Ross, G.D. Mechanism by which orally administered beta-1,3-glucans enhance tumoricidal activity of antitumor monoclonal antibodies in murine tumor models. J. Immunol. 2004, 173, 797–806.

16. Cheung, N.-K.; Modak, S.; Vickers, A.; Knuckles, B. Orally administered �-glucans enhance anti-tumor effects of monoclonal antibodies. Cancer Immunol. Immunother. 2002, 51, 557–564.

17. Andoh, A.; Fujiyama, Y. Anti-inflammatory roles of dietary fiber and short-chain fatty acids as regards inflammatory bowel diseases. Agro Food Ind. Hi-Tech 2004, 1, 42–43.

18. Lupton, J.R. Microbial Degradation Products Influence Colon Cancer Risk: the Butyrate Controversy. J. Nutri. 2004, 134, 479.

19. Bordonaro, M.; Lazarova, D.L.; Sartorellil, A.C. Butyraate and Wnt signaling. Cell Cycle 2008, 7, 1178–1183.

Page 88: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

585

20. Volman, J.J.; Rmakers, J.D.; Plat, J. Dietary modulation of immune function by �-glucans. Physiol. Behav. 2008, 94, 276–284.

21. Yamada, H.; Kiyohara, H.; Cell Glycobiology and Development Health and Disease in Glycomedicine; Boons, G.-J., Lee, Y.C., Suzuki, A., Taniguchi, N., Voragen, A.G.J., Eds.; Elsevier: Oxford, UK, 2007; pp. 664–693.

22. Holtekjølen, A.K.; Uhlen, A.K.; Bråthen, E.; Sahlstrøm, S.; Knutsen, S.H. Contents of starch and non-starch polysaccharides in barley varieties of different origin. Food Chem. 2006, 94, 348–358.

23. Knutsen, S.H.; Holtekjolen, A.K. Preparation and analysis of dietary fibre constituents in whole grain from hulled and hull-less barley. Food Chem. 2007, 102, 707–715.

24. Westerlund, E.; Andersson, R.; Åman, P. Isolation and chemical characterization of water soluble mixed-linked beta-glucans and arabinoxylans in oat milling fractions. Carbohydr. Polym. 1993, 20, 115–123.

25. Misra, C.K.; Das, B.K.; Mukherjee, S.C.; Pattnaik, P. Effect of multiple injections of �-glucan on non-specific immune response and disease resistance in Labeo rohita fingerlings. Fish Shellfish Immunol. 2006, 20, 305–319.

26. Czop, J.K.; Austen, K.F. Properties of glycans that activate the human alternative complement pathway and interact with the human monocyte �-glucan receptor. J. Immunol. 1985; 135, 3388–3393.

27. Akramiene, D.; Grazeliene, G.; Didziapetrine, J.; Kevelaitis, E. Treatment of Lewis lung carcinoma by photodynamic therapy and glucan from barley. Medicina (Kaunas) 2009, 45, 480–485.

28. Ross, G.D.; Cain, J.A.; Lachmann, P.J. Membrane complement receptor type three (CR3) has lectin-like properties analogous to bovine conglutinin and functions as a receptor for zymosan and rabbit erythrocytes as well as a receptor for iC3b. J. Immunol. 1985, 134, 3307–3315.

29. Xia, Y.; Vetvicka, V.; Yan, J.; Hanikyrová, M.; Mayadas, T.; Ross, G.D. The �-glucan-binding lectin site of mouse CR3 (CD11b/CD18) and its function in generationg a primed state of the receptor that mediates cytotoxic activation in response to iC3b-opsonized target cells. J. Immunol. 1999, 162, 2281–2290.

30. Thornton, B.P.; Vetvicka, V.; Pitman, M.; Goldman, R.C.; Ross, G.D. Analysis of the sugar specificity and molecular location of the �-glucan-binding lectin site of complement receptor Type 3 (CD11b/CD18). J. Immunol.1996, 156, 1235–1246.

31. Cheung, N.-K.V.; Modak, S. Oral (1�3),(1�4)-�-D-glucan synergizes with antiganglioside GD2 monoclonal antibody 3F8 in the therapy of neuroblastoma. Clin. Cancer Res. 2002, 8, 1217–1223.

32. Tada, R.; Adachi, Y.; Ishibashi, K.-I.; Tsubaki, K.; Ohno, N. Binding capacity of a barley �-glucan to the �-glucan recognition molecule Dectin-1. J. Agric. Food Chem. 2008, 56, 1442–1450.

33. Tada, R.; Ikeda, F.; Aoki, K.; Yoshikawa, M.; Kato, Y.; Adachi, Y.; Tanioka, A.; Ishibashi, K.; Tsubaki, K.; Ohno, N. Barley-derived �-D-glucan induces immunostimulation via a dectin-1-mediated pathway. Immunol. Lett. 2009, 123, 144–148.

34. Karin, M.; Yamamoto, Y.; Wang, Q.M. The IKK NF-kappaB system: a treasure trove for drug development. Nat. Rev. Drug Discovery 2004, 3, 17–26.

Page 89: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

586

35. Michaelsen, T.E.; Gilje, A.; Samuelsen, A.B.; Høgåsen, K.; Paulsen, B.S. Interaction between human complement and a pectin type polysaccharide fraction, PMII, from the leaves of Plantago major L. Scand. J. Immunol. 2000, 52, 483–490.

36. Di Carlo, F.J.; Fiore, J.V. On the composition of Zymosan. Science 1958, 127, 756–757. 37. Soltanian, S.; Stuyven, E.; Cox, E.; Sorgeloos, P.; Bossier, P. Beta-glucans as immunostimulants

in vertebrates and invertebrates. Crit. Rev. Microbiol. 2009, 35, 109–138. 38. Hetland, G.; Samuelsen, A.B.; Lovik, M.; Paulsen, B.S.; Aaberge, I.S.; Groeng, E.C.;

Michaelsen, T.E. Protective effect of Plantago major L. pectin polysaccharide against systemic Streptococcus pneumoniae infection in mice. Scand. J. Immunol. 2000, 52, 348–355.

39. Samuelsen, A.B.; Paulsen, B.S.; Wold, J.K.; Otsuka, H.; Kiyohara, H.; Yamada, H.; Knutsen, S.H. Characterization of a biologically active pectin from Plantago major L. Carbohydr Polym. 1996, 30, 37–44.

40. Li, Q.; Verma, I.M. NF-B regulation in the immune system. Nat. Rev. 2002, 2, 725–734. 41. Carlsen, H.; Moskaug, J.Ø.; Fromm, S.H.; Blomhoff, R. In vivo imaging of NF-B activity. J.

Immunol. 2002, 168, 1441–1446. 42. Austenaa, L.M.; Carlsen, H.; Hollung, K.; Blomhoff, H.K.; Blomhoff, R. Retionoic acid dampens

LPS-induced NF-�B activity: results from human monoblasts and in vivo imaging of NF-�B reporter mice. J. Nutri. Biochem. 2009, 20, 726–734.

43. Palma, A.S.; Feizi, T.; Zhang, Y.; Stoll, M.S.; Lawson, A.M.; Díaz-Rodríguez, E.; Campanero-Rhodes, M.A.; Costa, J.; Gordon, S.; Brown, G.D.; Chai, W. Ligands for the �-glucan receptor, Dectin-1, assigned using “designer” microarrays of oligosaccharide probes (neoglycolipids) generated from glucan polysaccharides. J. Biol. Chem. 2006, 281, 5771–5779.

44. Izydorczyk, M.S.; Dextre, J.E. Barley �-glucans and arabinoglucans: Molecular structure, physiochemical properties, and uses in food products—a review. Food Res. Int. 2008, 41, 850–868.

45. Holtekjølen, A.K.; Uhlen, A.K.; Bråthen, E.; Sahlstrøm, S.; Knutsen, S.H. Contents of starch and son-starch polysaccharides in barley varieties of different origin. Food Chem. 2006, 94, 348–358.

46. Janeway, C.A.; Travers, P. Immunbiology the immune system in health and disease; Current Biology. Ltd.: London, UK, 1996; pp. 31–50.

47. Roitt, I.; Brostoff, J.; Male, D. Immunology; Mandarin Offset Ltd.: Hong Kong, China, 1993. 48. Samuelsen, A.B. Polysaccharides in Plantago major L. Studies of structure and biological

activity. Ph. D. Thesis, University of Oslo, Oslo, Norway, 1998. 49. Moure, A.; Gullon, P.; Dominguez, H.; Parajo, J.C. Advances in the manufacture, purification and

applications of xylo-oligosaccharides as food additives and nutraceuticals. Process Biochem. 2006, 41, 1913–1923.

50. Femia, A.P.; Salvadori, M.; Broekaert, W.F.; Francois, I.E.J.A.; Delcour, J.A.; Courtin, C.M.; Caderni, G. Arabinoxylan-oligosaccharides (AXOS) reduce preneoplastic lesions in the colon of rats treated with 1,2-dimethylhydrazine (DMH). Eur. J. Nutri. 2010, 49, 127–132.

51. Knutsen, S.H.; Holtekjølen, A.K. Preparation and analysis of dietary fibre constituents in whole grain from hulled and hull-less barley. Food Chem. 2007, 102, 707–715.

52. Chambers, R.E.; Clamp, J.R. An assessment of methanolysis and other factors used in the analysis of carbohydrate-containing materials. J. Biochem. 1971, 125, 1009–1018.

Page 90: Dietary immune-modulation – carbohydrate specific effects ...

Int. J. Mol. Sci. 2011, 12

587

53. Samuelsen, A.B.; Paulsen, B.S.; Wold, J.K.; Otsuka, H.; Yamada, H.; Espevik, T. Isolation and partial characterization of biologically active polysaccharides from Plantago major L. Phytother. Res. 1995, 9, 211–218.

54. Vistica, D.T.; Skehan, P.; Scudiero, D.; Monks, A.; Pittman, A.; Boyd, M.R. Tetrazolium-based Assays for Cellular Viability: A Critical Examination of Selected Parameters Affecting Formazan Production. Cancer Res. 1991, 51, 2515–2520.

© 2011 by the authors; licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution license (http://creativecommons.org/licenses/by/3.0/).

Page 91: Dietary immune-modulation – carbohydrate specific effects ...

III

Page 92: Dietary immune-modulation – carbohydrate specific effects ...
Page 93: Dietary immune-modulation – carbohydrate specific effects ...

IV

Page 94: Dietary immune-modulation – carbohydrate specific effects ...
Page 95: Dietary immune-modulation – carbohydrate specific effects ...

V

Page 96: Dietary immune-modulation – carbohydrate specific effects ...
Page 97: Dietary immune-modulation – carbohydrate specific effects ...

VI

Page 98: Dietary immune-modulation – carbohydrate specific effects ...

Recommended