new characterizing effects of prebiotics and human milk … · 2017. 12. 19. · [ii]...

263
Characterizing effects of prebiotics and human milk oligosaccharides on the intestinal epithelial barrier by You (Richard) Wu A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Laboratory Medicine and Pathobiology University of Toronto © Copyright by You Wu 2017

Upload: others

Post on 23-Oct-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

  • Characterizing effects of prebiotics and human milk oligosaccharides on the intestinal epithelial barrier

    by

    You (Richard) Wu

    A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

    Laboratory Medicine and Pathobiology University of Toronto

    © Copyright by You Wu 2017

  • [ii]

    Characterizing effects of prebiotics and human milk oligosaccharides on the intestinal epithelial barrier

    You (Richard) Wu

    Doctor of Philosophy

    Laboratory Medicine and Pathobiology

    University of Toronto

    2017

    Abstract

    Prebiotics are non-digestible compounds that enhance the growth of certain microbes within the

    gut microbiota and confer benefits on the host, but whether they can also elicit direct immune-

    regulatory effects is unknown. This thesis addresses this gap through three specific aims: (1) to

    characterize the effects of prebiotics on the intestinal epithelial barrier, (2) to delineate the

    underlying signaling mechanisms underlying barrier- and immune-regulation, and (3) to evaluate

    the effects of human milk oligosaccharides using relevant models of human disease.

    In the first part of this thesis, two prebiotics, inulin and short-chain fructooligosaccharides

    (scFOS), were tested in an in vitro bacterial challenge system with enterohemorrhagic

    Escherichia coli serotype O157:H7. Inulin and scFOS increased the resistance of Caco-2Bbe1

    monolayers, reduced permeability and increased the expression of select tight junction proteins.

    These changes were associated with the activation of protein kinase C isoform δ - a host kinase

    enzyme that controls multiple cell functions.

    The second part of this thesis expands on PKCδ signaling and characterizes the global signaling

    of inulin and scFOS using Caco-2Bbe1 cells. This was performed using a kinome peptide array

    to profile kinase phosphorylation levels, which revealed several immune pathways and

  • [iii]

    functional networks that were regulated by prebiotics. These findings were then validated in vivo

    using a neonatal endotoxemia model, which showed that prebiotics dampened intestinal

    inflammation without altering the composition of gut microbiota.

    In the third part of this thesis, I demonstrate a unique mechanism whereby HMOs promote

    barrier function by inducing the production of mucus. This finding was confirmed using a

    neonatal mouse model of necrotizing enterocolitis (NEC), where HMOs prevented the

    development of NEC by enhancing intestinal goblet cell production.

    Taken together, these data demonstrate that prebiotics are not biologically inert. In fact,

    prebiotics directly trigger host cell signaling to alter epithelial barrier integrity and gut

    inflammation – indicating previously unrecognized mechanisms of prebiotics and HMOs.

  • [iv]

    Acknowledgements

    Foremost, I would like to thank my family for their loving support throughout this incredible

    journey. My mother and father, Susan Jiang and Zhiping Wu, whom despite knowing absolutely

    nothing about cell biology, have always given me their utmost attention to be the most tuned-in

    listeners of my work. To Gary and Tracy, my “left and right arms”, I appreciate you simply

    being there when I needed you.

    Second, I thank both the past and present members of the Sherman Lab. I must thank Kathene

    Johnson-Henry for her encouragements, wisdom and willingness to listen, allowing me to

    confide to her my various concerns as a graduate student. I thank all the valuable lab mates:

    Kathryn, Linda and Lee, for being so patient in my early transition from medicine to graduate

    training; Thomas, for helping me draw the connection to clinical relevance; Ana and Will, for the

    endless entertainment and wisdom both inside and outside of work; Pekka and Ebi, for the

    wonderful conversations and debates about meat versus vegetable.

    Thirdly, I owe my deepest gratitude to my supervisor, Philip Sherman for being an outstanding

    mentor. The excitement and enthusiasm that he shares for science have been absolutely

    infectious. He helped me see the positives of science and reminded me of its ultimate purpose

    and where this pipeline leads to - people. His nurturing helped me cultivate the skill of “thick-

    skin” - where no rejections or reviewer comments are bad enough to put me down. “Get up, do

    something else, then come back and try again” is now my life motto.

    Fourth, I thank my committee members, David Hwang and Nicola Jones for providing the

    guidance for my PhD; to Agostino, Peter, Yuhki and rest of the Pierro lab, for the incredible

    collaborations and friendships; to the Friday journal club, for all the fun and wonderful

    discussions; to the lab neighbors of the 21st floor, for all the coffees, cookies, “pick-me-ups” in

    the hallway that made my training enjoyable. I thank Monique, for teaching me the mastery of

    the ancient relic “rotovap” and importantly, for being so strong of a person.

    Lastly, I thank my grandmother Jie, whose battle with cancer took a toll on her life, but whose

    story forever fills a social consciousness to my scientific and clinical career, and forever reminds

    me about the needs of patients and the endless questions of human disease.

  • [v]

    Table of Contents

    Acknowledgements ......................................................................................................... iv

    Table of Contents ............................................................................................................. v

    List of Abbreviations ...................................................................................................... viii

    List of Figures ............................................................................................................... xiii

    List of Tables ................................................................................................................ xvi

    1 Introduction .................................................................................................................. 1

    1.1 Host-microbe interactions in health and disease ................................................... 3

    1.2 Dietary impacts on microbiota ............................................................................... 7

    1.2.1 Infant microbiota ......................................................................................... 7

    1.2.2 Adult microbiota ........................................................................................ 13

    1.3 Prebiotics as microbiota-targeted therapeutics ................................................... 16

    1.4 Prebiotic metabolites ........................................................................................... 21

    1.4.1 Prebiotic utilization.................................................................................... 21

    1.4.2 Polysaccharide utilization loci ................................................................... 22

    1.4.3 Microbiome population shifts .................................................................... 25

    1.5 Prebiotic-derived metabolites: short-chain fatty acids ......................................... 27

    1.6 Direct innate immune response to prebiotics ...................................................... 32

    1.6.1 Intestinal epithelial barrier ......................................................................... 33

    1.6.2 Pattern recognition receptors ................................................................... 45

  • [vi]

    1.6.3 Innate immunity in health and disease ..................................................... 53

    1.7 Direct innate immune modulation by prebiotics ................................................... 59

    2 Hypothesis and Objectives ........................................................................................ 63

    3 Protein Kinase C δ Signaling is Required for Dietary Prebiotic-Induced

    Strengthening of Intestinal Epithelial Barrier Function ............................................... 64

    3.1 Abstract ............................................................................................................... 65

    3.2 Introduction ......................................................................................................... 67

    3.3 Materials and Methods ........................................................................................ 69

    3.4 Results ................................................................................................................ 76

    3.5 Discussion ........................................................................................................... 96

    4 Prebiotics Modulate Pathogen-Induced Inflammatory Processes Through

    Regulation of Host Kinase Activities ........................................................................ 101

    4.1 Abstract ............................................................................................................. 102

    4.2 Introduction ....................................................................................................... 104

    4.3 Materials and Methods ...................................................................................... 106

    4.4 Results .............................................................................................................. 113

    4.5 Discussion ......................................................................................................... 148

    5 Dietary Human Milk Oligosaccharides Protect Experimental Necrotizing

    Enterocolitis by Inducing Mucin Production ............................................................. 153

    5.1 Abstract ............................................................................................................. 154

    5.2 Introduction ....................................................................................................... 156

  • [vii]

    5.3 Materials and Methods ...................................................................................... 159

    5.4 Results .............................................................................................................. 167

    5.5 Discussion ......................................................................................................... 195

    6 Discussion and Future Directions ............................................................................ 199

    6.1 Re-definition of “prebiotics” ............................................................................... 201

    6.2 Limitations and Future Directions ...................................................................... 205

    6.3 Significance of the research undertaken ........................................................... 210

    References .................................................................................................................. 211

  • [viii]

    List of Abbreviations

    Abbreviations Terms

    A/E Attaching and effacing

    AMPs Antimicrobial peptides

    BF Breastfed

    BPs Binding proteins

    CBP Creb-binding protein

    CC3 Cleaved caspase 3

    CCL C-C motif chemokine ligand

    CD Crohn’s Disease

    CDC Cell division control protein

    CE Carbohydrate esterase

    CFU Colony-forming unit

    CXCL C-X-C motif chemokine ligand

    DAPI 4',6-diamidino-2-phenylindole

    DCs Dendritic cells

    DMEM Dulbecco's modified Eagle medium

    DMSO Dimethyl sulfoxide

    DP Degrees of polymerization

    DPPs Differentially phosphorylated peptides

    DSS Dextran sodium sulfate

    EDTA Ethylenediaminetetraacetic acid

    EGF Epidermal growth factor

  • [ix]

    EGFR Epidermal growth factor receptor

    EHEC Enterohemorrhagic Escherichia coli O157:H7

    ERK1/2 P44/42 extracellular-regulated kinases

    FBS Fetal bovine serum

    FDR False discovery rate

    FF Formula-fed

    FITC Fluorescein-labeled isothiocyanate

    FOS Fructooligosaccharides

    GCs Goblet cells

    G-CSF Granulocyte colony stimulating factor

    GH Glycoside hydrolases

    GI Gastrointestinal

    GLP Glucagon-like peptide

    GO Gene ontologies

    GOS Galactooligosaccharides

    GPR G-coupled protein receptor

    GTP Guanosine-5'-triphosphate

    GUK Guanylate kinase domain

    HMOs Human milk oligosaccharides

    HPAEC-PAD High-performance anion-exchange chromatography with

    pulsed amperometric detection

    IBD Inflammatory bowel disease

    IECs Intestinal epithelial cells

  • [x]

    IESCs Intestinal epithelial stem cells

    IFN-γ Interferon-gamma

    IgA/G/M Immunoglobulin A/G/M

    IGF Insulin-like growth factor

    IκBα Inhibitor of kappa B alpha

    Iκκ IκB kinase

    Iκκα IκB kinase alpha

    IL Interleukin

    IRAK Interleukin-1 receptor-associated kinase

    IRFs Interferon regulatory factors

    KDa Kilodalton

    KEGG Kyoto encyclopedia of genes and genomes

    LPL Lipoprotein lipases

    LPS Lipopolysaccharide

    MAPKs Mitogen-activated protein kinases

    MDCK Madin-Darby canine kidney cells

    MIF Migration inhibiting factor

    MOI Multiplicity of infection

    MUC Mucin

    MW Molecular weight

    MyD88 Myeloid differentiation primary response gene 88

    NEC Necrotizing enterocolitis

    NF-κB Nuclear factor kappa B

  • [xi]

    NGF Nerve growth factor

    OTU Operational taxonomic unit

    PAMPs Pathogen-associated molecular patterns

    PCoA Principle coordinates analysis

    PCs Paneth cells

    PDI Protein disulfide isomerase

    PFA Paraformaldehyde

    PI3K Phosphatidylinositol-3-kinase

    PKC Protein kinase C

    PL Polysaccharide lyases

    PRRs Pattern recognition receptors

    PTMs Post-translational modifications

    PUL Polysaccharide utilization loci

    RACK Receptor for activated C kinase

    RALDH Retinaldehyde dehydrogenase

    RhoA Ras homolog gene family member A

    RIPK Receptor for receptor-interacting serine/threonine-protein

    kinase

    ROCK Rho-associated protein kinase

    rRNA Ribosomal RNA

    SAM Severe acute malnutrition

    SCFAs Short-chain fatty acids

    scFOS Short-chain fructooligosaccharides

  • [xii]

    SDS-PAGE Sodium dodecyl sulfate polyacrylamide gel electrophoresis

    SH3 Src-homology 3

    siRNA Silencer RNA

    SPDEF SAM pointed domain containing ETS transcription factor

    Sus Starch utilization system

    TAB TAK-1-binding protein

    TAK TGFβ-activated kinase

    TER Transepithelial electrical resistance

    TFF3 Trefoil factor 3

    TGFβ Transforming growth factor beta

    TIR Toll/IL-1R homology domain

    TJ Tight junctions

    TLRs Toll-like receptors

    TMAO Trimethylamine oxide

    TNF-α Tumor necrosis factor alpha

    TRAF-6 TNF receptor associated factor-6

    VEGF Vascular endothelial growth factor

    ZO Zona occludens

  • [xiii]

    List of Figures

    Chapter 1

    Figure 1.1: Environmental factors affecting the development of gut microbiota. ........... 10

    Figure 1.2: Structures of the three main categories of prebiotics .................................. 19

    Figure 1.3: Starch utilization system employed by B. thetaiotathemicron ..................... 23

    Figure 1.4: Local and systemic impacts of bacterial-derived metabolites SCFAs. ........ 30

    Figure 1.5: The cell morphology of small intestine villus-crypt unit. .............................. 35

    Figure 1.6: Intestinal TJ complexes .............................................................................. 41

    Figure 1.7: Zonula occludens binding domains ............................................................. 43

    Figure 1.8: MyD88-dependent and MyD88-independent pathways of TLR pathways .. 48

    Figure 1.9: Transwell system for the measurement of TER. ......................................... 56

    Figure 1.10: Direct immune-modulatory effects of prebiotics ........................................ 61

    Chapter 3

    Figure 3.1: Inulin and scFOS reduce EHEC O157:H7-induced barrier disruption in

    Caco-2Bbe1 monolayers. ............................................................................................. 78

    Figure 3.2: Characterization of 2D-grown intestinal organoids. .................................... 80

    Figure 3.3: Inulin and scFOS regulate TJ protein expression ....................................... 82

    Figure 3.4: ScFOS alters host kinase activities across multiple immune regulatory

    pathways ....................................................................................................................... 86

    Figure 3.5: Inulin and scFOS induce activation of host PKC signaling in a time- and

    dose-dependent manner ............................................................................................... 89

  • [xiv]

    Figure 3.6: Host signaling response to prebiotic inulin and scFOS ............................... 91

    Figure 3.7: Inhibition of PKC phosphorylation abolishes prebiotic-mediated protection of

    epithelial barrier function ............................................................................................... 94

    Chapter 4

    Figure 4.1: Kinome response of prebiotic-treated IECs to EHEC O157:H7 challenge. 115

    Figure 4.2: Kinome differences between inulin and scFOS. ....................................... 117

    Figure 4.3: Biological functions of prebiotic-induced kinome responses. .................... 121

    Figure 4.4: Comparison of the effect inulin and scFOS have on canonical pathways. 124

    Figure 4.5: Functional validation of NF-κB pathway in IECs. ...................................... 128

    Figure 4.6: Functional validation of MAPK pathway in IECs. ...................................... 130

    Figure 4.7: Effect of inulin and scFOS on LPS-induced murine endotoxemia. ............ 133

    Figure 4.8: Effects of inulin and scFOS on colonic microbiota of LPS-treated mouse

    pups. ........................................................................................................................... 137

    Figure 4.9: Effects of inulin on colonic microbiota of LPS-treated mouse pups. ......... 139

    Figure 4.10: Effects of scFOS on colonic microbiota of LPS-treated mouse pups. ..... 141

    Figure 4.11: Effects of scFOS on colonic microbiota of LPS-treated mouse pups. ..... 143

    Figure 4.12: Effects of inulin and scFOS on metagenome functional content of LPS-

    treated mouse pups. ................................................................................................... 145

    Chapter 5

    Figure 5.1: The composition of HMOs in the pooled human milk. ............................... 168

    Figure 5.2: HMO prevent the NEC in neonatal mice. .................................................. 170

  • [xv]

    Figure 5.3: HMO increase cell proliferation and the number of muc-2 producing cells.

    .................................................................................................................................... 173

    Figure 5.4: HMO induce mucin production in LS174T and Caco-2Bbe1 cells. ........... 176

    Figure 5.5: HMO protect Caco-2Bbe1 monolayers from EHEC-induced disruption. ... 179

    Figure 5.6: HMO rescue Caco-2Bbe1 TJ-mediated barrier function. .......................... 181

    Figure 5.7: HMO-mediated protection of the epithelial barrier is size and dose-

    dependent. .................................................................................................................. 183

    Figure 5.8: Inhibition of PDI prevents HMO-mediated protection of the intestinal

    epithelial barrier. ......................................................................................................... 186

    Figure 5.9: HMO increase the expression of PDI. ....................................................... 188

    Figure 5.10: Inhibition of PDI abolishes the protective effect of HMO on NEC

    development. .............................................................................................................. 191

    Figure 5.11: Rutin on healthy mouse pups. ................................................................ 193

  • [xvi]

    List of Tables

    Table 1.1: Dysbiosis in human disease ........................................................................... 6

    Table 1.2: Major bioactive components present in mature breast milk. ........................ 12

    Table 1.3: List of current prebiotic candidates and their structures. .............................. 17

    Table 1.4: TLR localization and ligands. ....................................................................... 46

    Table 1.5: Innate immune cytokines and chemokines .................................................. 51

    Table 1.6: TJs linked to hereditary diseases and infectious agents .............................. 55

    Table 4.1: Quality statistics for metagenome functional content prediction from 16S

    rDNA sequences using PICRUSt. ............................................................................... 147

  • [1]

    Chapter

    1 Introduction

    This chapter was published, in part, as:

    Abrahamsson TR, Wu RY, Sherman PM. Microbiota in functional gastrointestinal disorders

    in infancy: implications for management. Nestlé Nutrition Institute Workshop Series, 2017;

    88: 107-115.

    Wu RY, Jeffrey M, Johnson-Henry KC, Green-Johnson J, Sherman PM. Impact of

    Prebiotics, Probiotics and Gut Derived Metabolites on Host Immunity. LymphoSign Journal,

    2016; 4(1): 1-24.

    Johnson-Henry KC, Abrahamsson TR, Wu RY, Sherman PM. Probiotics, prebiotics, and

    synbiotics for the prevention of necrotizing enterocolitis. Advances in Nutrition, 2016; 7(5):

    928-937.

    Wu RY, Sherman P. Host-intestinal microbe interactions in human health and disease.

    University of Toronto Medical Journal. 2015; 92(3): 30-34.

    Abrahamsson TR, Wu RY, Jenmalm MC. Gut microbiota and allergy: the importance of the

    pregnancy period. Pediatric Research, 2015; 77(1-2): 214-219.

  • [2]

    Foreword

    The intestinal microbiota plays a crucial role in host well-being and disruption in the gut

    microbiota (referred to as dysbiosis) is linked to a variety of human diseases. Therapeutic efforts

    aimed at restoring microbial imbalances include the use of dietary prebiotics, which are non-

    digestible food substances metabolized by specific members of the intestinal microbiota to alter

    the composition and/or functions of the intestinal microbiome. Bacterial breakdown of prebiotics

    is both glycan- and bacteria-dependent, and through this process, metabolites including short-

    chain fatty acids are produced which can elicit numerous effects both locally and systemically.

    Although it is traditionally believed that prebiotics benefit the host by manipulating the

    microbiome and facilitating the production of short-chain fatty acids, more recent evidence

    suggests that prebiotics may directly stimulate cytokine and chemokine production in the host

    intestinal epithelia. However, the signaling mechanism of specific prebiotics on the host

    intestinal epithelia has not been investigated and thus forms the subject of the studies presented

    in this thesis. My overarching hypothesis is that prebiotics, besides altering bacterial functions,

    directly trigger host cell signaling to provide intestinal homeostasis to regulate inflammation and

    barrier protection. To approach this, this thesis will first introduce the prior studies and theories

    that helped formulate my questions, and then detail my experiments, results and interpretations

    in Sections 3-5.

  • [3]

    1.1 Host-microbe interactions in health and disease

    Historical context

    During the 19th

    century, when studying the anthrax epidemic, Louis Pasteur noted that the

    growth of Bacillus anthracis was reduced by the presence of other microorganisms. He described

    this phenomenon as “la lutte pour la vie” or “battle for life” (Sams et al., 2014). Pasteur predicted

    that certain microbes are beneficial for health, and this was supported in 1907 when Eli

    Mechnikov, a Nobel Prize laureate, made the seminal observation that peasants who regularly

    consumed lactic acid-producing bacteria in fermented dairy products lived longer and healthier.

    Mechnikov reasoned “the dependence of the intestinal microbes on the food makes it possible to

    adopt measures to modify the flora in our bodies and to replace the harmful microbes with useful

    microbes” (Wallace et al., 2011). This concept of balancing “harmful versus beneficial” or “good

    versus bad” microorganisms forms the basis of the current understanding of host-microbe

    interactions.

    Human microbiota

    Every one of us harbours a complex collection of microorganisms in our body referred to as the

    microbiota. The most complex microbiota lies in the gastrointestinal (GI) tract, which consists of

    over 1014

    microbial cells (i.e. 1.3 times that of human somatic and germ cells) that collectively

    express 150 times more genes than human cells combined (Ley et al., 2006). Although the

    majority of studies have focused on the bacterial populations, the GI microbiota encompasses

    other microbial domains including Archaea, viruses, fungi and protozoa (Lloyd-Price et al.,

    2016). Methanobrevibacter smithii, for example, is an Archaea species documented in healthy

  • [4]

    individuals that plays an important role in the carbohydrate metabolization of select bacterial

    communities (Samuel et al., 2007).

    The GI microbiota also harbours extensive populations of viruses, with more than 109

    viral

    particles recoverable per gram of human feces (Minot et al., 2013). Even though the majority of

    these viruses are bacteriophages (viruses that infect and replicate within bacteria), virome

    composition is highly unique amongst individuals (Virgin, 2014). Unlike viruses and Archaea,

    protozoa and fungi are eukaryotic microorganisms that are also present in the GI microbiota. For

    example, the fungus Saccharomyces boulardii is present in healthy individuals and may protect

    against cholera infection (Hatoum et al., 2012). Despite the growing evidence for Archaea,

    viruses, fungi and protozoa within the gut microbiota, knowledge surrounding their precise

    composition and functionality remains limited.

    The community most intensively characterized within the GI microbiota is the bacterial

    population. To characterize diverse bacterial compositions, substantial efforts have been made to

    catalogue the plethora of bacteria found in the GI tract. Previously, this was performed with

    classic culture-dependent techniques using isolated bacterial strains. These traditional methods of

    identification are now replaced by high-throughput 16S rRNA genome sequencing - a technique

    that allows investigators to rapidly profile the microbiome in detail and overcome challenges

    with culturing bacterial species. From this large collection of data, over 1,000 bacterial species

    have now been identified (Rajilić-Stojanović and de Vos, 2014), at least 160 of which are readily

    found among healthy individuals (Qin et al., 2010). It is also evident that the bacterial taxa

    belong to the two predominant phyla: Bacteroidetes and Firmicutes (Huttenhower et al., 2012).

  • [5]

    Dysbiosis in disease

    Impairment in the microbial balance, or dysbiosis, can trigger disease pathogenesis through

    changes in microbial diversity. For instance, the replacement of existing microbiota through co-

    housing or transplantation from colitogenic donors (Blanton et al., 2016; Wlodarska et al., 2014)

    causes colitis in otherwise healthy mice, indicating that exposure to pathogenic bacteria is

    sufficient to initiate mucosal inflammation. On the other hand, germ-free animals devoid of

    microbial communities develop poorly with severe disruption in the development of lymphoid

    follicles, T-cell function and an increased susceptibility to infection (Olszak et al., 2012).

    Therefore, the gut microbiota is important not only in the progression of disease, but is also

    crucial for normal physiological development. Using next generation sequencing, a wide

    spectrum of human disease has now been linked with intestinal dysbiosis (Table 1.1). However,

    the next challenge is how to harness this wealth of information to improve human health using

    microbial manipulations.

  • [6]

    Table 1.1: Dysbiosis in human disease

    Diseases Dysbiotic microbiota

    Irritable bowel

    syndrome

    Increased Firmicutes (Ruminococcus, Clostridium and Dorea species)

    Decreased Bifidobacterium and Faecalibacterium species

    Colorectal

    cancer

    Increased Fusobacterium members

    Altered Coprococcus, Eubacterium rectale, Roseburia and

    Faecalibacterium prausnitzii

    Obesity

    Increased Lactobacillus species, Methanobrevibacter smithii,

    Faecalibacterium prausnitzii

    Decreased Bacteroides, Akkermansia mucinophila

    Type 2 diabetes Increased Clostridium, Akkermansia mucinophila, Bacteroides and

    Desulfovibrio

    Ulcerative

    colitis

    Increased Proteobacteria, Fusobacteria and Spirochaetes

    Decreased Firmicutes, Lentisphaerae and Verrucombicroa

    Necrotizing

    enterocolitis

    Increased Proteobacteria, emergence of Cronobacter sakazakii and

    enteropathogenic Escherichia coli

    Cardiovascular

    disease Increased Proteobacteria

    Severe acute

    malnutrition

    Increased Proteobacteria (genera Klebsiella, Escherchia)

    Decreased Bacteroidetes (genera Bifidobacteria)

    *Table adapted from: Magno da Costa Maranduba C et al. Intestinal microbiota as modulators of the immune

    system and neuroimmune system: impact on the host health and homeostasis. Journal of Immunology Research.

    2015; 2015: 1-15.

  • [7]

    1.2 Dietary impacts on microbiota

    1.2.1 Infant microbiota

    Studying how microbial communities are initially established in infants provides clues to the

    factors that impact the microbiome. The newborn, believed to be largely sterile at birth,

    immediately begins a chaotic progression towards acquiring a functional microbiota. Factors that

    influence initial microbial colonization in the GI tract include mode of delivery (i.e. Caesarean

    section versus vaginal birth) (Hansen et al., 2014), antibiotic use (Alm et al., 2008), maternal

    microbiome (Matsumiya et al., 2002) and environmental exposures (Hanski et al., 2012) (Figure

    1.1). For example, vaginally-delivered babies are predominantly colonized by vagina-residing

    microbes including Lactobacillus and Prevotella, whereas Caesarean-delivered babies have more

    potentially pathogenic skin-derived bacteria like staphlyococci and actinobacteria. In addition,

    perinatal use of antibiotics has been shown to disrupt bacterial diversity in newborn babies

    (Walker, 2010).

    Dietary influences on the infant microbiome

    Amongst the factors impacting gut microbiota, diet instills long-lasting impacts on the

    microbiome (Albenberg and Wu, 2014). Studies characterizing the microbiome during the first 3

    years of age reveal considerable differences in bacterial taxa that are specifically linked to infant

    dietary intakes (Koenig et al., 2011). By 18 months of age, breastfed infants have almost double

    the proportion of Bifidobacteria with more diverse Bifidobacterium species than formula-fed

    infants (Roger et al., 2010). However, despite this direct correlation, recent studies revealed large

    variations in Bifidobacteria of breastfed infants and in some locations, Bifidobacteria may

  • [8]

    actually be completely absent in the infant stools (Lewis and Mills, 2017). This suggests that

    while breastfeeding provides a propensity for microbial shifts, differences in non-diet factors

    such as glycan composition, genetics, and rate of feeding may place selective pressure on the

    intestinal colonization. For instance, babies of secretor mothers (those that express

    fucosyltransferase 2) were shown to have greater relative abundance of Bifidobacteria than non-

    secretor mothers (Lews et al., 2015), suggesting that maternal genetics may also significantly

    influence the bifidogenic effects of breastfeeding. The dessimination of these environmental

    effects across geography is an ongoing endeavor in the field of infant microbiome.

    Breast milk as the gold standard

    Exclusive breast milk feeding for the first 6 months of life is recognized as the gold standard for

    infant feeding (Ballard and Morrow, 2013). This is because, in addition to being a complete

    nutritional source supporting the infant growth, breast milk also contains a plethora of bioactive

    components including proteins, enzymes, growth factors, immunoglobulins, and

    oligosaccharides that each could protect the developing baby (Table 1.2). In addition, live

    bacteria such as staphylococci, streptococci, bifidobacteria, and lactic acid-producing bacteria

    are also identified in breast milk (Walker, 2010). Microbes from maternal skin can be routinely

    inoculated into mammary ducts during breastfeeding. Alternatively, it has also been proposed

    that milk microbes could originate from maternal gut via the entero-mammary pathway (Gomez-

    Gallego et al., 2016). Currently, the origin of breast milk microbiota and its biological function

    on the infant are both unclear (Fernández et al., 2013; Gomez-Gallego et al., 2016).

    Unlike formula milk, which is commercially standardized to contain nutrient and micronutrient

    contents within a small range, breast milk composition is dynamic with considerable

  • [9]

    heterogeneity present both within mothers, as well as between mothers and populations (Walker,

    2010). Colostrum, the breast milk produced during the first few days post-partum, is higher in

    lipids (Khan et al., 2013), carbohydrates (Bode, 2012), growth factors, secretory IgA and

    lactoferrin than mature milk 2-6 months post-partum (Ballard and Morrow, 2013).

    There are substantial interpersonal variations in macronutrient compositions between mothers

    due to blood type, secretor status, maternal weight, as well as environmental factors such as

    nursing frequency and dietary intakes (Nommsen et al., 1991; Wacklin et al., 2011). Recently,

    several groups have also identified inter-personal differences in genes expressed by cells present

    in breast milk, which further highlights the high heterogeneity in milk contents (Twigger et al.,

    2015).

  • [10]

    Figure 1.1: Environmental factors affecting the development of gut microbiota.

    At birth, the neonatal intestine is largely sterile, where factors including mode of delivery, diet

    environment (i.e. maternal microbes, diet, and exposure to antibiotics) influence the progression

    towards a diverse and stable adult gut microbiota.

  • [11]

  • [12]

    Table 1.2: Major bioactive components present in mature breast milk.

    Component Function

    Macromolecules (g/dL)

    Protein (1.2) Provision of nutrients and energy

    Fat (3.2) Provision of nutrients and energy

    Lactose (7.8)

    Oligosaccharides (5-10)

    Provision of nutrients and energy

    Microbiota substrates, anti-adhesives, immune effects

    Cells

    Macrophages Protection against infections

    Stem cells Regeneration

    Immunoglobulins

    IgA/sIgA Pathogen inhibition

    IgG Anti-microbial, activates phagocytosis, anti-inflammatory

    IgM Agglutination, complement activation

    Cytokines & chemokines

    IL-6 Acute phase response, B-cell activation, pro-inflammatory

    IL-7 Increased thymic size and output

    IL-8 (CXCL8) Recruitment of neutrophils, pro-inflammatory

    IL-10 Repressing inflammation, antibody production, tolerance

    IFNγ Pro-inflammatory, stimulates Th1 response

    TGFβ Anti-inflammatory, stimulation of T cell, phenotype switch

    TNFα Stimulates inflammatory immune activation

    C-CSF Trophic factor

    MIF Prevents macrophage movement, increase activity

    Growth factors

    EGF Stimulate proliferation and maturation

    VEGF Promote angiogenesis and tissue repair

    NGF Promote neuron growth and maturation

    IGF Stimulate growth and development

    Erythropoietin Erythropoesis, intestinal development

    Lactoferrin Acute phase protein, chelates iron, anti-bacterial

    *Table adapted from: Ballard O, Morron A. Human Milk Composition: Nutrients and Bioactive Factors. Pediatr

    Clin North Am. 2013; 60(1): 49-74.

  • [13]

    1.2.2 Adult microbiota

    Diet alters the gut microbiome

    With weaning and the introduction of solid foods (~4-6 months of age is the current norm for

    breastfed infants in North America), the infant microbiota eventually transitions to an adult

    composition by 2-3 years of age with increased abundance of Bacteroides, streptococci and

    clostridia (Palmer et al., 2007). Unlike infants, the individual gut microbiota of adults is largely

    stable (Faith et al., 2013), and this long-term composition is classified into several broad

    “enterotypes” that are intimately associated with food consumption (Wu et al., 2011). For

    instance, children from Burkina Faso, whose diets consist mainly of carbohydrates, fiber and

    non-animal proteins, have higher Prevotella and lower Bacteroides compared to children living

    in Europe who consume a Western diet consisting of high animal protein, fats, sugar and low

    fiber content (De Filippo et al., 2010). Similar fluctuations in the microbiota can also be

    triggered by short-term dietary changes, such as the extreme deprivation of carbohydrates (David

    et al., 2014). Besides altering microbial abundance, dietary exposures also influence bacterial

    function. For example, in a comparison between healthy human vegans and omnivores, diet had

    a surprisingly small impact on microbial composition. Nevertheless, the production of bacterial

    metabolites was divergent between the two groups (Wu et al. 2016), indicating that dietary

    factors not only regulate the growth of select microbial communities, but can also alter bacterial

    gene expression.

  • [14]

    Diet-induced dysbiosis influences health and disease

    The diet-microbiota relationship is closely connected with human disease. The contrast that

    exists in the microbiome between Agrarian and Western diets is mirrored by the varying

    incidence of chronic diseases amongst the two populations. The microbiome associated with the

    Western diet is correlated with higher rates of obesity and chronic inflammatory diseases.

    Conversely, diets high in vegetables and fibers are linked with lower levels of disease states and

    frailty (Wu et al., 2011). For example, obese individuals have decreased bacterial diversity and

    altered levels of Firmicutes and Bacteroidetes compared to lean counterparts (Turnbaugh et al.,

    2008, 2009). In fact, transferring the gut microbiota of obese humans to germ-free mice can

    induce higher energy balance and fat accumulation despite constant food intake (Turnbaugh et

    al., 2006), thereby suggesting that obesity is associated with an “obesogenic” microbiota.

    However, due to differences in technique and study design, the specific microbial changes in

    obesity have not always been replicated (Sze and Schloss, 2016). Therefore, the link between

    obesity and microbiome remains an active area of ongoing investigation.

    Dysbiotic disease mechanisms

    These diet-driven chronic diseases are likely due to the functional activity of microbiome-

    derived metabolites. For instance, the high consumption of lipids and red meats increases the

    levels of choline and L-carnitine, respectively. Both are microbial substrates that are readily

    metabolized by the intestinal microbiota to form the atherosclerosis-promoting molecule

    trimethylamine oxide (TMAO) (Koeth et al., 2013; Wang et al., 2011). This is also supported by

    studies showing that meat-eaters produce higher levels of TMAO after L-carnitine consumption

    due to the different composition of the gut microbiota (Koeth et al., 2013). Therefore,

  • [15]

    modulation of diet plays a role in the maintenance of well-being by modulating the composition

    and the function of the host gut microbiota.

  • [16]

    1.3 Prebiotics as microbiota-targeted therapeutics

    Prebiotic definition

    The relationship between diet, microbial communities and human health and disease has

    generated tremendous interest in microbiota-targeted strategies to improve human health

    (Bindels et al., 2015; Jain and Walker, 2014). Amongst these proposed strategies is the idea of

    dietary prebiotics initially described by Gibson and Roberfroid (Gibson and Roberfroid, 1995).

    Prebiotics are defined as “non-digestible food ingredients that resist intestinal digestion and

    absorption, and through their use as substrate by the gut microbiota, modulate the composition

    and activity of gut microbiota, thus conferring a benefit on the host” (Bindels et al., 2015).

    Sources of prebiotics

    Common examples of prebiotic compounds are plant-derived fructans, such as inulin and

    fructooligosaccharides (FOS), commercially-derived galactooligosaccharides (GOS) and human

    milk oligosaccharides (HMOs). Inulin and FOS/oligofructose are energy-storage carbohydrate

    polymers commonly found in plants and vegetables, including asparagus, leafy green vegetables,

    garlic, leek and onions, with the highest abundance in chicory root (Cichorium intybus). On the

    other hand, GOS are linear galactose chains commercially synthesized by the enzymatic trans-

    galactosylation and hydrolysis of lactose sugars (Bindels et al., 2015). HMOs can be extracted

    from human breast milk through the serial removal of macronutrients (i.e. fats and proteins).

    Besides these 3 main categories, numerous alternative candidates for prebiotics have been

    introduced with ongoing research underway (Table 1.3). However, such candidates are not the

    core focus of the current thesis.

  • [17]

    Table 1.3: List of current prebiotic candidates and their structures.

    Prebiotic Linkages Monosaccharides

    Inulin β-(2,1) Fructose

    Glucose

    Fructooligosaccharides β-(2,1) Fructose

    Glucose

    Galactooligosaccharides β-(1,4) Galactose

    Glucose

    Human milk

    oligosaccharides

    β-(1,3)

    β-(1,6)

    α-(1,2)

    α-(1,3)

    α-(1,4)

    α-(2,3)

    α-(2,6)

    Glucose

    Fructose

    Galactose

    Sialic acid

    N-acetylglucosamine

    Resistant starch* α-(1,4)

    α-(1,6) Glucose

    Pectin* α-(1,4)

    D-galacturonic acid

    Galactose

    Aplose

    Keto-deoxyoctulosonic acid

    Hydroxycinnamic acid

    Rhamnose

    Arabinose

    Fucose

    Arabinoxylan* β-(1,4) Xylopyranose

    *Indicates candidate prebiotics that are currently under evaluation.

  • [18]

    Prebiotics versus dietary fibers

    The non-digestible aspect of prebiotics overlaps with the concept of dietary fiber. Dietary fibers,

    such as non-digestible starch, are the complex carbohydrates that form part of the plant cell wall

    that resists hydrolysis and digestion and therefore, can travel the GI tract undigested (DeVries,

    2003). Dietary fibers are classified into soluble and insoluble fiber based on their physical

    properties. Soluble fibers (i.e. pectins, gums and FOS) dissolve in water and are fermented by

    colonic microbiota, whereas insoluble fibers (i.e. lignins and cellulose) do not solubilize in

    water, have limited fermentation capacities and mainly serve as a colonic bulking agent (Wong

    and Jenkins, 2007). Although most prebiotic compounds can be classified as soluble dietary

    fibers, not all dietary fibers are considered prebiotic agents.

    Structure of Prebiotics

    The structures of prebiotics vary depending on the sources. Inulin and FOS are comprised of

    repeating monomers of fructose connected to a terminal glucose via β-(2,1) glycosidic linkages

    (Vogt et al., 2015). Inulin and FOS differ in length, with a degree of polymerization of more than

    and less than 10 for inulin and FOS, respectively. Short-chain FOS or scFOS have a chain length

    of 2-5. GOS are galactose polymers that are structurally linked by β-(1,4) glyosidic linkages. By

    contrast, HMOs are heavily branched oligosaccharides that contain up to five sugars: glucose

    (Glc), galactose (Gal), fucose (Fuc), sialic acid (Sia) and N-acetyl-glucosamine (GlcNac) (Bode,

    2012). HMO side chains are built on lactose at the reducing end and elongated by the addition of

    lacto-N-biose or N-acetyllactosamine in β-(1,3) or β-(1,6) linkages. Elongated side chains then

    terminate with sialic acid and fucose moieties: Fuc in α-(1,2), α-(1,3) or α-(1,4) linkages, and Sia

    residues in α-(2,3) or α-(2,6) linkages.

  • [19]

    Figure 1.2: Structures of the three main categories of prebiotics

    FOS and GOS are linear non-branching polymers of fructose and galactose sugar units organized

    in β-(2,1) and β-(1,4) glycosidic linkages with a glucose terminal end. HMOs are

    oligosaccharides that are made up of five different sugar units. Figure adapted from Bode et al.,

    2016.

  • [20]

  • [21]

    1.4 Prebiotic metabolites

    1.4.1 Prebiotic utilization

    Due to the complexity of prebiotic structures, the breakdown of prebiotic requires a cadre of

    degradation enzymes to hydrolyze glycosidic linkages. Given that humans express only a few of

    these enzymes, most of the prebiotic consumed travels along the GI tract undigested to reach the

    colon intact. In fact, with the exception of starch, lactose and sucrose, the majority of

    polysaccharides ingested are metabolized by colonic microbes whose genomes encode the

    required glycoside hydrolases, polysaccharide lyases and carbohydrate esterases (Flint et al.,

    2012; Sela et al., 2008). These enzymes are responsible for the bacterial sensing, importing and

    degradation of oligosaccharide structures (Comstock, 2009).

    One of the best described examples of oligosaccharide utilization is the polysaccharide

    degradation process employed by Bacteroides species. Bacteroides members such as B.

    thetaiotamicron express a large collection of oligosaccharide degrading enzymes. For instance,

    in order to breakdown the α-(1,4) and α-(1,6) glycosidic linkages in resistant starch, a dietary

    fiber that is currently evaluated as a potential prebiotic, B. thetaiotamicron employs a 4-enzyme

    starch utilization system (Sus) as shown in Figure 1.3. Not surprisingly, as the number of unique

    polysaccharide linkages increases, the more enzymes will be required in its degradation. Xylan,

    which has a total of 11 unique linkages, requires approximately 21 bacterial enzymes for

    degradation in a Sus-like system (Koropatkin et al., 2012). The concerted activities of Sus

    enzymes in Bacteroides provide the current paradigm of prebiotic degradation.

  • [22]

    1.4.2 Polysaccharide utilization loci

    The entire repertoire of degradation enzymes is expressed within bacterial gene clusters known

    as polysaccharide utilization loci (PUL). Although many PUL are common between bacterial

    species, many PUL are highly specific and render select species as primary metabolizers. For

    example, when closely related members from the genus Bacteroides are grown on a prebiotic

    inulin agar base, most members were excellent inulin metabolizers and increased bacterial

    growth with the exception of Bacteroidales vulgatus, which is an inulin “non-metabolizer”

    (Rakoff-Nahoum et al., 2014). Interestingly, when the agar substrate is switched from inulin to

    xylan, the inulin metabolizer B. thetaiotamicron failed to grow while inulin non-metabolizers

    like B. vulgatus demonstrated enhanced proliferation (Rakoff-Nahoum et al., 2014). This

    difference in response is due to differences in the expression of glycan degradation enzymes. For

    instance, B. thetaiotamicron is an excellent metabolizer of β-(2,6)-fructans because it specifically

    encodes the β-(2,6) endo-fructanase BT1760 (Sonnenburg et al., 2010). Therefore, these

    experiments show that bacterial fermentation of prebiotics is dependent on both the bacterial

    species as well as the type of prebiotic the microorganism is exposed to. Consequently,

    microbiota changes introduced by prebiotics are both substrate- and microbiota-specific.

  • [23]

    Figure 1.3: Starch utilization system employed by B. thetaiotathemicron

    Non-digestible starches are bound to bacterial membranes by the starch utilization system (Sus)

    E and F proteins, and starch fragments are imported within bacteria by SusC. Enzymatic

    breakdown is carried out by SusA and SusB glycoside hydrolases (GH). Figure adapted from

    Koropatkin et al., 2012.

  • [24]

  • [25]

    1.4.3 Microbiome population shifts

    The substrate- and species-specific nature of prebiotic utilization establishes ecological niches

    that can have a profound impact on the gut microbiome. One such impact is Bifidogenesis, or the

    expansion of Bifidobacterium, a genus of Gram-positive saccharolytic bacteria containing

    numerous members with reported health effects in the host. Although the characterization of

    Bifidobacterium polysaccharide utilization enzymes is not fully revealed, in vitro fermentation

    studies have identified the involvement of selective transporters (i.e. ABC transporters) and

    several degradation enzymes (i.e. β-(2-1)-galactosidases, α-sialidase and α-fucosidases). A well-

    documented example is B. longum subsp. infantis, which expresses numerous enzymes that

    together break down the plethora of prebiotic structures. B. infantis expresses β-

    fructofuranosidases for the digestion of inulin and FOS and lacS carriers and β-galactosidases for

    GOS. To tackle the complexity of HMO structures, the microorganism also expresses various

    glycosidases (i.e. α-sialidases and α-fucosidases) (Sela et al., 2008), specific binding proteins

    (BPs), as well as ABC transporters for the breakdown of HMOs (Smilowitz et al., 2014).

    Bifidogenic effects in humans

    The bifidogenic effect of prebiotics is well-supported by human studies. Infants receiving daily

    formula containing a 1.25 to 4 g mixture of inulin and FOS have a higher abundance of

    bifidobacteria in stools (Boehm, 2002; Boehm and Moro, 2008; Gonzalez et al., 2008). The

    youngest segment of the population tested were babies at 6 days of age, who showed a

    significant increase in both lactobacilli and bifidobacteria when taking a mixture of GOS/FOS at

    8 g/L for 28 days (Moro et al., 2002). The same GOS/FOS mixture induces a microbial

    composition that closely resembles the Lactobacillus and Bifidobacterium populations of

  • [26]

    breastfed infants (Haarman and Knol, 2006; Knol et al., 2005). Similar findings are seen in

    healthy adults where inulin at doses from 5-34 g/day or FOS at a range of 5-20 g/day

    consistently demonstrate bifidogenic shifts in the colonic microbiota (Meyer and Stasse-

    Wolthuis, 2009).

    Structure-specific effects on the gut microbiome

    Although the consumption of prebiotics induces bifidogenic effects in most cases, the extent of

    expansion varies between prebiotics – a finding similar to the in vitro setting. FOS is a preferable

    fermentation substrate, whereas inulin and maltodextrin are relatively poor for Bifidobacterium

    metabolization. Even at a high dose of 10 g/day, a few studies found that long-chain inulin could

    not produce Bifidogenic effects in the gut microbiome (Bouhnik et al., 2004). This structure-

    specific manipulation of the intestinal microbiota also applies to bacterial function. As

    demonstrated in the context of severe acute malnutrition, mothers whose children exhibit

    stunting have lower levels of sialyated HMOs. When both sialyated and non-sialyated prebiotics

    were fed to the malnourished mice, although there were no large differences in the bacterial

    communities, numerous bacterial strains responded transcriptionally by regulating genes

    involved in metabolism (Charbonneau et al. 2016). Interestingly, such changes were marked by

    improvements in host lean-body mass and bone growth in HMO-fed animals, but not in animals

    receiving either inulin or FOS. Similarly, sialyated HMOs, but not GOS, protect against

    intestinal pathology in a neonatal rat model of NEC (Autran et al. 2016). Given that more than

    200 unique HMO structures are present (Bode, 2012), further characterization of the structure-

    specific functions of these HMOs is warranted.

  • [27]

    1.5 Prebiotic-derived metabolites: short-chain fatty acids

    The bifidogenic effects of prebiotics are associated with numerous health benefits, which are

    believed to be partly mediated by the metabolites produced during prebiotic fermentation such as

    short-chain fatty acids (SCFAs) (Vogt et al., 2015). Structurally, SCFAs are 1-6 carbons in

    length (Figure 1.4), with the majority (>95%) of SCFAs being butyrate (4 carbons), propionate

    (3 carbons) and acetate (2 carbons) at a ratio of 1:1:3 in the colon (Brestoff and Artis, 2013;

    Macfarlane and Macfarlane, 2003). The levels of SCFAs fluctuate depending on the composition

    of the resident gut microbiota as well as the prebiotic metabolized. For instance, many

    Bacteroides members generate predominantly butyrate along with hydrogen and carbon dioxide

    gases, whereas for some of the Bifidobacterial members, the main metabolites produced are

    lactate and acetate (Pokusaeva et al., 2011). In addition, while xylan breakdown generates mostly

    acetate, the majority of SCFAs produced in inulin breakdown is butyrate. Germ-free animals

    devoid of a functional metabolizing microbiota are completely deficient in intestinal SCFAs

    (Maslowski et al., 2009).

    Levels of SCFAs have important implications on host health through impacts on host immunity

    and energy metabolism, which are not only demonstrated in local tissues, but also systemically

    (Arpaia et al., 2013; Brestoff and Artis, 2013). Animals low in SCFAs, such as germ-free mice

    or mice deprived of fiber intake, have defective immune responses and mount more severe

    inflammation in response to dextran sodium sulfate- (DSS) or Citrobacter rodentium-induced

    colitis (Kim et al., 2013; Maslowski et al., 2009). Despite the numerous supports for their

    benefits, it was recently shown that butyrate may also generate deleterious effects on crypt cell

    homeostasis and inhibit wound healing (Kaiko et al., 2016).

  • [28]

    One potential mechanism for how SCFAs regulate immunity is the binding of host cell receptors

    GPR-41 (G-coupled Protein Receptor 41) and GPR-43 – a small family of G protein coupled

    receptors expressed by various cell types including colonocytes, monocytes, neutrophils and

    macrophages (Le Poul et al., 2003). SCFAs directly activate GPR-41 and -43, which result in the

    decreased expression of pro-inflammatory cytokines and increased expression of the anti-

    inflammatory cytokines IL-10 and IL-23 in dendritic cells (Liu et al., 2012). Grp43-/-

    mice

    develop severe DSS colitis despite normalized tissue levels of SCFAs (Maslowski et al., 2009).

    Similarly, one recent study using a mouse model of peanut allergy described a GRP-43-mediated

    mechanism of SCFAs-induced induction of oral tolerance, whereby SCFAs activation of GRP-43

    enhanced retinaldehyde dehydrogenase (RALDH) activity in tolerogenic CD103+ dendritic cells

    (DC) and stimulated Treg production (Tan et al., 2016).

    The majority of intestinal SCFAs (>90%) are readily absorbed by colonocytes and used as an

    energy source. However, a small portion of SCFAs is taken up into systemic circulation via the

    superior and inferior mesenteric veins to mediate effects on distant organs (Brestoff and Artis,

    2013). Both GRP-41 and -43 receptors are expressed in the liver, adipose tissues and skeletal

    muscle (Canfora et al., 2015). In fact, several mouse studies demonstrate that SCFAs enhance

    weight loss and reduce adiposity (Frost et al., 2014; Gao et al., 2009). These effects are likely to

    be multifactorial in nature, since numerous reports have documented SCFAs-mediated effects on

    satiety, energy expenditure, adiposity and liver function. For instance, in both humans and

    rodents, SCFAs exposure stimulates the release of glucagon-like peptide-1 (GLP-1) – a gut

    hormone produced by enteroendocrine L-cells that is involved in satiety (Tolhurst et al., 2012).

    Oral feeding of butyrate in obese mice also accelerates fat loss via increased energy expenditure

    and fat oxidation in skeletal muscles due to the higher abundance of type 1 muscle fibers (Gao et

  • [29]

    al., 2009). Likewise, ex vivo studies using human adipose tissues also demonstrate higher

    expression of lipoprotein lypases (LPL) and enhanced glycerol release with SCFAs exposure –

    indicating reduced adiposity via lipolysis (Canfora et al., 2015). Whether these mechanisms are

    potential ways to combat human obesity warrants further investigation.

  • [30]

    Figure 1.4: Local and systemic impacts of bacterial-derived metabolites SCFAs.

    Main intestinal SCFAs (acetate, propionate and butyrate) directly bind GPR-41/43 receptors on

    colonocytes and immune cells to affect immune function. SCFAs are also taken up into the

    systemic circulation via superior and inferior mesenteric veins to elicit distant effects on liver,

    muscle and adipose tissues.

  • [31]

  • [32]

    1.6 Direct innate immune response to prebiotics

    Although health benefits related to prebiotic consumption have been largely attributed to the

    bifidogenic effects and induction of SCFAs (Vogt et al., 2015), a recent concept is that prebiotic

    oligosaccharides may also elicit direct effects via contact with host intestinal tissues (Roberfroid

    et al., 2010). In other words, prebiotics could elicit direct effects in the GI tract independent of

    microbial fermentation.

    The component of the host immune system responsible for initial recognition of foreign

    substances is the innate immune system (Takeuchi and Akira, 2010). As an early response to

    noxious stimuli, the innate immune system incorporates several mechanisms to provide a first

    line of defense, discrimination between self- versus non-self and an ability to mount appropriate

    immune responses through the induction of both cytokines and chemokines. In Section 1.6, I

    will discuss the innate immune mechanisms within the GI tract specifically relevant to the

    breadth of this thesis. In Section 1.7, I will consider how prebiotics have the potential to directly

    regulate these immune mechanisms.

  • [33]

    1.6.1 Intestinal epithelial barrier

    The first line of the innate immune system is the anatomical and chemical barrier that separates

    the host from luminal foreign substances. This includes anatomical barriers such as the skin, as

    well as a chemical barrier including secreted molecules such as lysozyme and antimicrobial

    peptides. The following sections will discuss individual components of the intestinal epithelial

    barrier.

    Intestinal cell lineages

    Within the GI tract, the first line of defense against antigens present in luminal contents is the

    intestinal barrier, made up of a single layer of polarized epithelial cells organized spatially into

    intestinal crypts and villi. Besides providing a physical separation, the intestinal barrier facilitates

    nutrient digestion and absorption and mounts innate immune responses to various insults present

    in the lumen of the GI tract (Kagnoff, 2014). To perform these diverse functions, the intestinal

    barrier contains several specialized cell lineages that are spatially distributed. The four main cell

    types include enterocytes, enteroendocrine cells, goblet cells (GCs) and Paneth cells (PCs)

    (Figure 1.5).

    Enterocytes are polarized epithelial cells found in the intestinal barrier and are specialized in

    nutrient absorption via fluid-phase endocytosis. Enteroendocrine cells release peptide hormones

    that regulate GI functions such as food consumption and motility and enable the GI tract to

    improve bacterial clearance. The intestinal epithelium also contains specialized secretory PCs

    and GCs, situated within the crypts and villi, respectively. Both cell types secrete soluble factors

    to either destroy or reduce the ability of enteric pathogens to adhere to the surface of the

    intestinal epithelium (Kagnoff, 2014).

  • [34]

    PCs secrete antimicrobial peptides (AMPs) including defensins, cathelicidins, and C-type lectins

    into crypts which disrupt bacterial membranes to destroy pathogens (Peterson and Artis, 2014).

    GCs cells secrete abundant amounts of mucins, which are large, O-glycosylated glycoproteins

    that coat the entire mucosal surface to entrap luminal microbes and prevent bacterial attachment

    to the apical surface of intestinal epithelial cells (IECs) (Johansson and Hansson, 2016). Mice

    deficient in Muc2, the main isotype of mucin secreted in the GI tract, develop spontaneous colitis

    and are more susceptible to colorectal cancers (Van der Sluis et al., 2006).

    Each of the specialized cell types in the gut undergoes continuous turnover and is replenished by

    the intestinal epithelial stem cells (IESCs) located at the base of the crypt, which generate a

    steady supply of committed cell-specific progenitors. Collectively, this forms a dynamic mucosal

    barrier that continuously self-regenerates to cover approximately 400 m2

    of surface area.

  • [35]

    Figure 1.5: The cell morphology of small intestine villus-crypt unit.

    The intestinal epithelium is comprised of diverse cell types, including: enterocytes, goblet cells

    (GC), Paneth cells (PC), enteroendocrine cells, and intestinal epithelial stem cells (IESC)

    organized into crypts and villus. DC denotes dendritic cells.

  • [36]

  • [37]

    Structural integrity of the villus-crypt unit

    The individual cells types in the GI tract are anchored together seamlessly into a single-layered

    sheet of epithelium. The key to maintaining this continuous structure is the intercalating network

    of intercellular TJ proteins that seal the intestinal lining. The primary role of the TJs is to

    function as a diffusion barrier to control the paracellular passage of molecules and ions (Peterson

    and Artis, 2014). TJs also create a molecular fence that juxtaposes the neighbouring membranes

    to prevent lateral diffusion of membrane components into adjacent cells. This creates cell

    polarity, which allows subcellular organelles to organize into apical and basolateral

    compartments. Furthermore, TJ structures facilitate host cell signaling by engaging with adaptor

    and signaling proteins to affect cellular responses to various external stimuli.

    Studies using electron microscopy revealed TJ complexes as a collection of transmembrane

    strands anchored to plaque proteins and the cytoskeleton of adjacent cells (Zihni et al., 2016).

    The main types of transmembrane proteins observed are the tetraspanning claudin and occludin

    family of proteins (Figure 1.6).

    Claudins

    The claudin family consists of 26 members that range in molecular size between 20-34

    kilodaltons (kDa) and in number of amino acids between 207 and 305. Claudins are spatially

    located in the intercellular space between adjacent cells and are structurally composed of

    intracellular N-terminal and C-terminal domains and two extracellular loop domains that thread

    back-and-forth across the membrane to form four transmembrane domains. Claudins of adjacent

    cells are thought to dimerize via the edges of the extracellular domains (Zihni et al., 2016). In

    Latin, “claudin” refers to “to close”, but considerable work reveals that claudins act as both para-

  • [38]

    cellular barriers (“barrier claudins”) and para-cellular pores (“leaky claudins”), and are the main

    determinants of epithelial permeability (Gunzel and Yu, 2013). A total of 19 claudin members

    are expressed throughout the GI tract in both humans and mice, but with considerable

    heterogeneity with respect to their ion permeability. For instance, claudin-2 is permissive to

    cations and considered a pore-forming claudin (Amasheh et al., 2002), whereas claudin-17 is an

    anion pore, and referred as a pore-closing claudin (Krug et al., 2012). The relative distribution

    and expression of individual claudins together regulates the electrical conductance of the cell

    monolayer, which can be readily measured in filter-grown polarized epithelial cell monolayers

    using chopstick voltage electrodes.

    Occludins

    The second main class of transmembrane proteins are occludins, which are tetraspanning

    proteins approximately 65 kDa in size and containing 522 amino acids (Cummins, 2012). Like

    claudins, occludins have intracellular N- and C-terminal domains, four transmembrane domains,

    and two overhanging extracellular loop domains that reside in the intercellular spaces (Figure

    1.6) (Feldman et al., 2005). In Latin, “occludin” refers “to occlude”, but its role in barrier

    function is much more complex than barrier sealing. Madin-Darby canine kidney (MDCK) cells

    overexpressing occludin have higher trans-epithelial electrical resistance, but also exhibit higher

    paracellular fluxes (Feldman et al., 2005; McCarthy et al., 1996). On the other hand, knockdown

    of occludin expression in MDCK cells does not affect the TJ ultrastructure of cell monolayers at

    steady state (Yu, 2005). However, these cells have lower protein levels of TJ claudin-1 and -7

    and a reduced transepithelial electrical resistance (TER), indicating that occludins mediate the

    expressions and assembly of other TJ proteins.

  • [39]

    Similarly, although occludin-knockout mice are completely viable and exhibit intact expression

    of TJ proteins in the gut, the animals display severe pathologies including infertility,

    inflammation, growth retardation, infertility, and exhibit TJ disruptions in other epithelial tissues

    (Saitou et al., 2000). These results suggest that the core function of occludin is likely involved in

    the host stability of TJ complexes.

    Junctional plaque proteins

    To provide cytosolic anchorage, the C-terminal domains of claudins and occludins are tethered to

    a family of intracellular scaffolding proteins (Zihni et al., 2016). One predominant example is the

    zonula occludens family which contains three known members that are 220 kDa in size with

    1,748 amino acids: ZO-1, ZO-2 and ZO-3 (Dörfel and Huber, 2012). Structurally, ZO proteins

    consist of an N-terminal PDZ domain, a Src-homology 3 (SH3) domain and a guanylate kinase

    (GUK) domain. The N-terminus domain provides three binding sites for the various

    transmembrane TJ proteins: PDZ1 bound by claudins, PDZ2 bound by ZO-2 and ZO-3, PDZ3

    bound by junctional adhesion molecules (JAMs) and GUK domain bound by occludins (Figure

    1.7). The C-terminus domain is bound to the actin cytoskeleton inside of the epithelial cell.

    There is substantial evidence that both the expression and localization of ZO proteins are critical

    to cell viability and epithelial barrier function (Dörfel and Huber, 2012). Knockout animals of

    ZO-1 and ZO-2 are embryonically lethal (Katsuno et al., 2008). MDCK cells under ZO-1

    knockout or RNA interference have altered intercellular junctions and disruptions in TJ protein

    localization (Tokuda et al., 2014; Van Itallie et al., 2009). Moreover, changes in ZO-1

    localization also affect the barrier integrity of epithelial monolayers. Studies by Cario and

    colleagues (2004) demonstrated that apical delocalization of ZO-1 produces a barrier-sealing

  • [40]

    effect and is correlated with a higher TER value in monolayers. Apart from maintaining an

    epithelial barrier, ZO proteins are also bound by kinases and phosphatases that can alter the

    phosphorylation status of various TJ proteins to regulate functional activities.

    TJ-mediated cell signaling

    In addition to the role of TJs in maintaining the physical barrier as a structural complex, TJ also

    engage in extensive cross-talk with a vast number of second messengers (Zihni et al., 2016).

    These interactions are imparted via direct intracellular signaling pathways given that numerous

    TJ proteins are bound or can readily recruit signaling mediators including kinases, phosphatases,

    GTP-binding proteins and transcription regulators. One example that illustrates TJ signaling is

    the mechanism by which TJs directly alter cytoskeleton architecture: loss of ZO-1 induces the

    formation of actin stress fibers (Terry et al., 2011). One proposed mechanism is that ZO-1

    recruits the junction-regulating proteins JACOP and p114RhoGEF, whose concerted activity

    triggers a RhoA-ROCK2 signaling pathway that modulates cell tension (Tornavaca et al., 2015).

    Similarly, ZO-1 also facilitates the recruitment of cell division control protein 42 (CDC42) to

    trigger formation of the actin cytoskeleton (Oda et al., 2014) as well as inducing cellular

    differentiation (Zihni et al., 2016).

  • [41]

    Figure 1.6: Intestinal TJ complexes

    The intestinal epithelium is held together by intercellular TJ that consist of transmembrane

    glycoproteins (claudins and occludins) tethered onto cytosolic plaque proteins (ZOs). Figure

    adapted from Zihni et al., 2016.

  • [42]

  • [43]

    Figure 1.7: Zonula occludens binding domains

    Zonula occludens acts as a scaffolding protein to provide anchorage for other TJ proteins

    including claudins (PDZ-1), ZOs (PDZ2), JAMs (PDZ3), Occludins (GUK domain) and C

    terminal domains attached to actin cytoskeleton. Figure adapted from Thevenin et al., 2013.

  • [44]

  • [45]

    1.6.2 Pattern recognition receptors

    In addition to imposing a physical barrier, the intestinal epithelium fulfills an important role in

    the host immune response by discriminating self versus non-self within luminal contents. To do

    this, IECs express pattern recognition receptors that recognize pathogen-associated and damage-

    associated molecular patterns. The four families of pattern recognition receptors include the

    transmembrane proteins Toll-like receptors (TLRs) and C-type lectin receptors, as well as the

    cytosolic proteins retinoic acid-inducible gene-I-like receptors and NOD-like receptors. My PhD

    thesis focuses on the regulation of TLRs by prebiotics using complementary in vitro and in vivo

    models of intestinal injury, and therefore will be discussed further.

    Toll-like receptors

    TLRs are the best-characterized pattern recognition receptors, and are expressed not only in

    immune cells, such as macrophages and dendritic cells, but are also present in other cell types

    including epithelial cells, goblet cells and Paneth cells (Barton and Kagan, 2009). The structure

    of TLRs consists of an N-terminal ligand domain, a transmembrane domain and a C-terminal

    cytoplasmic Toll/IL-1R (TIR) domain for signal transduction (Botos et al., 2011). So far, there

    are 10 known TLRs in humans and 12 in mice, each of which is involved in the recognition of

    different sets of ligands (Table 1.4). During binding, a ligand links the extracellular domains to

    form an M shape allowing for the dimerization of the two intracellular TIR domains of the TLRs.

    This re-arrangement initiates downstream signaling to recruit adapter proteins, such as myeloid

    differentiation factor 88 (MyD88) and TIR-domain-containing adapter-inducing interferon-β

    (TRIF) (Manavalan et al., 2011).

  • [46]

    Table 1.4: TLR localization and ligands.

    TLR Localization Ligand Origin of ligands

    TLR1 Plasma

    membrane Triacyl lipoprotein Bacteria

    TLR2 Plasma

    membrane Lipoprotein

    Bacteria, viruses, parasites,

    host

    TLR3 Endolysosome dsRNA Viruses

    TLR4 Plasma

    membrane LPS Bacteria, viruses, host

    TLR5 Plasma

    membrane Flagellin Bacteria

    TLR6 Plasma

    membrane Diacyl lipoprotein Bacteria, viruses

    TLR7 (human

    TLR8) Endolysosome ssRNA Virsues, bacteria, host

    TLR9 Endolysosome CpG-DNA Viruses, bacteria, protozoa,

    self

    TLR10 Endolysosome Unknown Unknown

    TLR11 Plasma

    membrane

    Profilin-like

    molecule Protozoa

    *Table adapted from: Takeuchi O, Akira S. Pattern recognition receptors and inflammation. Cell. 2010; 140(6): 805-

    820.

  • [47]

    MyD88-dependent pathway

    Following dimerization of TIR-TIR domains, one of the first adaptor proteins recruited to the

    TIR domain, with the exception of TLR3, is MyD88 (Akira and Takeda, 2004). In the inactive

    state, MyD88 is localized in the cytosol in a repressed state (Figure 1.8). Once active, MyD88

    forms a complex with IL-1 receptor associated kinase family members (IRAK) through death

    domain interactions. This binding phosphorylates IRAK-4 and the subsequent phosphorylation

    of IRAK-1, which leads to the dissociation from MyD88 and association with TNF receptor

    associated factor-6 (TRAF-6) (Gay et al., 2014). Next, the IRAK-1-TRAF-6 complex interacts

    with the TAK1 protein complex (consisting of TAB1, TAB2 or TAB3), which activates the IκB

    kinase (Iκκ) complex. The Iκκ complex is the central regulator of the NF-κB inflammation

    cascade: its activation leads to downstream phosphorylation of NF-κB and the mitogen-activated

    protein kinases (MAPKs) JNK and p38 (Akira and Takeda, 2004).

    MyD88-independent pathway

    In the Myd88-independent pathway, otherwise known as the TRIF-dependent pathway, the

    adaptor protein recruited following TIR dimerization is the TIR-domain-containing adaptor

    protein TRIF (Figure 1.8) (Akira and Takeda, 2004). TRIF recruits the Iκκ-TBK1 complex to

    initiate the activation of interferon-regulatory factor 3 (IRF-3). IRFs are transcription factors

    expressed within the cytosol. Once activated, IRF-3 translocates to the nucleus and recruits the

    transcriptional co-activators p300 and Creb-binding protein (CBP) to mediate gene regulation

    (Kawasaki and Kawai, 2014).

  • [48]

    Figure 1.8: MyD88-dependent and MyD88-independent pathways of TLR pathways

    Within the MyD88-dependent pathway, MyD88 facilitates the activation of the IRAK-TRAF6

    complex and subsequent activation of the Iκκ complex. In the MyD88-independent pathway, the

    adaptor protein TRIF facilitates the recruitment of signaling mediators. Figure adapted from

    Akira and Takeda, 2004; Takeuchi and Akira, 2010.

  • [49]

  • [50]

    Inflammatory responses

    The activation of NF-κB and IRF-3 initiate pro-inflammatory responses by upregulating genes

    that encode pro-inflammatory cytokines, chemokines, type 1 interferons, antimicrobial proteins

    and other inflammatory mediators, some of which are listed in Table 1.5 along with their effects

    on the host (Takeuchi and Akira, 2010).

    Cytokines and chemokines

    Cytokines are secreted extracellular proteins that trigger local inflammation by promoting

    vascular permeability, recruiting immune cells, inducing acute-phase response proteins and

    regulating cell death in inflamed tissues (Takeuchi and Akira, 2010). Within the plethora of

    cytokines expressed are a small group of molecules known as chemokines, which are specialized

    chemo-attractant cytokines that facilitate the recruitment of immune cells. Common examples of

    cytokines include TNF-α and the interleukin-1 (IL-1) superfamily of cytokines such as IL-1α, IL-

    1β and IL-18 (Sims and Smith, 2010), while examples of chemokines includes the C-X-C

    chemokine ligand family such as CXCL8 (IL-8) in humans (MIP-2 in mice), and the C-C

    chemokine ligand (CCL) family such as CCL2, CCL5 and CCL8. Cytokines and chemokines can

    be secreted by a number of non-professional immune cells such as epithelial cells. To attain their

    desired functions, secreted cytokines and chemokines bind to their designated receptors (listed in

    Table 1.5).

  • [51]

    Table 1.5: Innate immune cytokines and chemokines

    Immune functions Cytokine families Cytokine members

    Adaptive immunity

    (proliferation and

    activation of

    macrophages, T cells, B

    cells and NK cells)

    Common γ chain receptor

    ligands IL-2, IL-4, IL-7, IL-9, IL-15, IL-21

    Common β chain (CD131)

    receptor ligands IL-3, IL-5, GM-CSF

    Shared IL-2β chain (CD122) IL-2, IL-15

    Shared receptors IL-13 (IL-13R–IL-4R complex)

    TSLP (TSLPR–IL-7R complex)

    Pro-inflammatory

    signaling

    (activation of

    phagocytes, chemotaxis,

    and phagocytosis;

    induction of cytokines)

    (IFN: anti-viral

    capacities, macrophage

    activation)

    IL-1

    IL-1α, IL-1β, IL-1ra, IL-18, IL-33,

    IL-36α, IL-36β, IL-36γ, IL-36Ra,

    IL-37 and IL-1Hy2

    IL-6 IL-6, IL-11, IL-31, CNTF, CT-1,

    LIF, OPN, OSM

    TNFα TNFα, TNFβ, BAFF, APRIL

    IL-17 IL-17A-F, IL-25 (IL-17E)

    Type I IFN IFNα, IFNβ, IFNω, IFNκ, Limitin

    Type II IFN IFNγ

    Type III IFN IFNλ1 (IL-29), IFNλ2 (IL-28A),

    IFNλ3 (IL-28B)

    Anti-inflammatory

    signaling

    (inhibition of cytokine

    production; anti-

    inflammatory)

    IL-12 IL-12, IL-23, IL-27, IL-35

    IL-10 IL-10, IL-19, IL-20, IL-22, IL-24,

    IL-26, IL-28, IL-29

    Immune functions Chemokine families Chemokine members

    Recruitment of immune

    cells, T-cells,

    macrophages,

    neutrophils

    CC chemokine/receptor family CCL1-28

    C chemokine/receptor family XCL1-2

    CXC chemokine/receptor

    family CXCL1-17

    CX3C chemokine/receptor

    family CX3CL1

    *Table adapted from: Turner D. Turner et al. Cytokines and chemokines: at the crossroads of cell signaling and

    inflammatory disease. Biochim Biophys Acta. 2014; 1843(11): 2563-2582.

  • [52]

    Other TLR-regulatory molecules

    In addition to TLR signaling, expression of cytokines and chemokines is also regulated by other

    mediators such as phosphatidylinositol-3-kinase (PI3K), protein kinase B (AKT) and protein

    kinase C (PKC). My PhD thesis incorporates the activity of PKC in TJ regulation whose role will

    be discussed further below.

    PKC refers to a family of serine or threonine kinases with at least 15 isoforms (Mochly-Rosen et

    al., 2012). Depending on activation conditions, the PKC isoforms can be subdivided into 3

    families. The conventional isoforms PKC-α, -βI, -βII and γ are activated by calcium,

    diacylglycerol and phosphatidylserine; the novel isoforms, PKC-δ, -ε, -η and -θ are calcium-

    independent but require diacylglycerol and phosphatidylserine; and the atypical isoforms, PKC-

    ζ and λ/ι require only phosphatidylserine (Newton, 2003).

    Two isoforms that play a significant role in TLR signaling and are highly expressed in intestinal

    tissues are the PKC isoforms α and δ (Loegering and Lennartz, 2011). Early studies using

    chemical inhibitors demonstrated their direct roles in TLR-induced cytokine secretion.

    Macrophages and neutrophils exposed to the PKC inhibitor Gö6976, which specifically blocks α

    and β isoforms, have reduced NF-κB activation and decreased secretion of TNF-α and IL-1β

    following TLR stimulation (Asehnoune et al., 2005; Foey and Brennan, 2004). Likewise,

    immune cells derived from PKC-α-/-

    mice have reduced activation of MAPK and NF-κB

    signaling pathways and their associated cytokines following TLR activation (Langlet et al.,

    2010). Similarly, down-regulation of PKCδ or inhibition of its activity using the PKCδ-specific

    inhibitor Rottlerin significantly dampens the activation of NF-κB and MAPK pathways by TLR

    ligands (Bhatt et al., 2010).

  • [53]

    1.6.3 Innate immunity in health and disease

    The host innate immune system provides multiple mechanisms to prtect the host. Defects in any

    facet of these mechanisms can render the host susceptible to infection and disease pathogenesis.

    Disruption in TJ assembly and function results in intestinal barrier breakdown, which is a key

    process in the pathogenesis of several human diseases including inflammatory bowel diseases

    (IBD), celiac disease (CD) and necrotizing enterocolitis (NEC) (Odenwald and Turner, 2016). In

    IBD, for instance, increased intestinal permeability due in part to the higher expression of pore-

    forming TJ claudin-2 (Zeissig et al., 2007) and lower expression of occludin (Heller et al., 2005)

    correlates with lower rates of clinical remission (Wyatt et al., 1993). Mutations in genes

    encoding TJs are also linked to a wide range of hereditary human diseases (Table 1.6).

    Select bacterial pathogens, such as Clostridium perfringens, hijack specific claudin members to

    breakdown the integrity of the epithelial barrier (Saitoh et al., 2015). This breakdown of claudins

    opens the unrestricted pathway for luminal contents so that intact large proteins and bacterial

    cells and metabolites can translocate into the submucosa to initiate further damage.

    This sequence of events in epithelial barrier breakdown can be modeled using in vitro systems by

    co-culturing an enteric pathogen with intestinal epithelial cell lines on Transwell® inserts (Figure

    1.9). These inserts are made of a semi-permeable polystyrene membrane that forms an inner

    well, which is completely submersed into an outer well. Confluent epithelial cell monolayers can

    be grown on Transwell® inserts and juxtaposed between the inner and outer wells to form apical

    and basolateral environments. The human intestinal epithelial cells I used in experiments

    described in this thesis are the Caco-2Bbe1 colonic adenocarcinoma cells - a subclone of the

    Caco-2 cells with brush border expression on the apical surface of epithelia. To induce barrier

  • [54]

    defects in vitro, enteric pathogens, pro-inflammatory mediators and chemical reagents can be

    administered into the apical environment to simulate the luminal exposure of bacteria and their

    toxins on the intestinal lining. Within my thesis, the enteric pathogen selected for use was the

    non-invasive enterohemorrhagic Escherichia coli (EHEC) serotype O157:H7. EHEC O157:H7 is

    a Shiga-toxin-producing E. coli (STEC) responsible for sporadic cases and outbreaks of bloody

    diarrhea and, in the most severely affected cases, causes hemolytic-uremic syndrome.

    EHEC O157:H7 expresses a number of virulence factors that directly alter the F-actin

    cytoskeleton and disrupt TJ barrier function, including Map, EspG and EspF (Ugalde-Silva et al.,

    2016). EspG, for instance, contains structural motifs that allow tethering onto the host F-actin

    cytoskeleton. This interaction disrupts the microtubule network and alters the integrity of TJ

    proteins claudin-1, ZO-1/ZO-2 and occludins (Glotfelty et al., 2014). These effects results in a

    decrease in the transepithelial electrical resistance (TER), which is measured by inserting

    chopstick electrodes into the inner and outer wells of Transwells®.

  • [55]

    Table 1.6: TJs linked to hereditary diseases and infectious agents

    TJ

    her

    edit

    ary d

    isea

    se

    TJ proteins Human diseases

    Claudin 1 Neonatal icthyosis, sclerosing cholangitis

    Claudin 5 Velo-cardial-facial syndrome

    Claudin 14 Non-syndromic deafness

    Claudin 16 Familial hypomagnesemia, hypercalciuria, nephrocalcinosis

    Claudin 19 Familial hypomagnesemia, hypercalciuria, nephrocalcinosis,

    visual impairments

    ZO-2 Familial hyper