bioactive components from lactobacillus acidophilus and

97
Bioactive Components from Lactobacillus acidophilus and Lactobacillus helveticus Fermented Milk Enhance Epithelial Membrane Integrity against Salmonella enterica serovars Typhimurium Infection by Jingya Peng A Thesis presented to The University of Guelph In partial fulfilment of requirements for the degree of Master of Science in Food Science Guelph, Ontario, Canada © Jingya Peng, May, 2014

Upload: others

Post on 08-Dec-2021

13 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Bioactive Components from Lactobacillus acidophilus and

Bioactive Components from Lactobacillus acidophilus and Lactobacillus

helveticus Fermented Milk Enhance Epithelial Membrane Integrity against

Salmonella enterica serovars Typhimurium Infection

by

Jingya Peng

A Thesis

presented to

The University of Guelph

In partial fulfilment of requirements

for the degree of

Master of Science

in

Food Science

Guelph, Ontario, Canada

© Jingya Peng, May, 2014

Page 2: Bioactive Components from Lactobacillus acidophilus and

ABSTRACT

BIOACTIVE COMPONENTS FROM LACTOBACILLUS ACIDOPHILUS AND

LACTOBACILLUS HELVETICUS FERMENTED MILK ENHANCE EPITHELIAL

MEMBRANE INTEGRITY AGAINST SALMONELLA ENTERICA SEROVARS

TYPHIMURIUM INFECTION

Jingya Peng Advisor:

University of Guelph, 2014 Professor Mansel W. Griffiths

Previous in vitro and in vivo studies showed that bioactive components produced by Lactobacillus

acidophilus (La-5) and Lactobacillus helveticus (LH-2) exerted protective effects against a variety of

enteric pathogens infection, including Salmonella. The purpose of this study is to determine the

mechanism of protective effect from La-5 and LH-2 against Salmonella Typhimurium infection on

epithelial cells.

Cell free spent medium (CFSM) were obtained after 48 h fermentation in whey protein-based medium

or milk for La-5 and LH-2, respectively. Human colonic carcinoma HT-29 cells were grown in

Transwell inserts for 40 days until they were polarized (TEER ~120 ohm×cm2). Cells were

pre-incubated with CFSMs 24 h prior to Salmonella infection and co-incubated during infection.

Lactate dehydrogenase (LDH) activity and TUNEL apoptotic assay were tested. Finally, an invasion

assay was carried out using chicken hepatoma LMH cells as an in vitro model for Salmonella

presence in poultry.

Results shown that Salmonella induced TEER loss was attenuated when pre-incubated and

co-incubated with non-toxic dose of CFSMs at 8th h post-infection (P <0.05). LDH was reduced up to

Page 3: Bioactive Components from Lactobacillus acidophilus and

49.1% (La-5) and 46.8% (LH-2) (P <0.01). In terms of apoptosis, 75.8% (La-5) and 62.5% (LH-2)

less apoptotic cells were observed (P <0.01). The effectiveness of Salmonella invasion on LMH cells

was strain dependent. Less than one log10 cycle reduction was obtained when La-5 or LH-2 CFSM

was applied.

These data suggest that LH-2 and La-5 have antagonized function against Salmonella infection in

epithelial cells by enhancing epithelial membrane integrity and would bring further beneficial

evidence of consuming bioactives as functional supplements.

Page 4: Bioactive Components from Lactobacillus acidophilus and

iii

ACKNOWLEDGEMENT

First and foremost, I would like to thank my supervisor, Prof. Mansel W. Griffiths, for his insightful

guidance and financial support. I thank him for giving me this chance and becoming someone whom I

can always rely on whenever I needed help throughout the course of my research. My sincerest

gratitude goes to my charming and intelligent committee members, Prof. Milena Corredig and Dr.

Angela Tellez, whose encouragements and suggestions always contributed to improving my work and

inspiring me in not only my research but also my life.

My appreciation extends to my fellow friends in the Canadian Research Institute for Food Safety and

Food Science department, especially to Rocio Morales, Sapana Sharma, Ruiqin Wu and Sandy Smith.

Thank you for all the encouragements, support and trust they provided for me. I would not arrive at

this stage of my life without all their help.

Above all, my greatest gratitude goes to my parents. I thank them for giving birth to me and being my

role models. I deeply appreciate their encouragement on challenging myself and chasing my dreams. I

thank my aunt, uncle and cousin in Toronto for their care throughout my study in Canada. Finally, I

thank my boyfriend for his unconditional love and support. I would not finish the Master degree on

time without the support from all of them.

Lastly, I thank the University of Guelph for endowing me with this precious journey. I will cherish

everything that I have learnt in Guelph. This moment is not a happy ending, but the beginning to a

new adventure, as my motto says, “live and learn”.

Page 5: Bioactive Components from Lactobacillus acidophilus and

iv

TABLE OF CONTENTS

Abstract

Acknowledgements………………………………………………………...…………………………..iii

Table of contents…………………………………………………………………………………….....iv

List of tables……………………………………………………………………………………………vi

List of figures………………………………………………………………………………………….vii

List of abbreviations………………………………………………………………………………….viii

CHAPTER 1 .......................................................................................................................................... 1

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

Literature Review .................................................................................................................................... 2

1. Salmonella spp. ............................................................................................................................... 2

2. Probiotics and Their Fermented Products ....................................................................................... 3

3. Bioactive Peptides ........................................................................................................................... 8

Definition and Characteristics ......................................................................................................... 8

Bioactive Peptides Derived from Milk ........................................................................................... 8

4. Intestinal Epithelial Cells .............................................................................................................. 12

Epithelial Cells Polarization.......................................................................................................... 12

Trans-epitheliall Electrical Resistance .......................................................................................... 13

Epithelial Cells Immune Response to Salmonella Invasion ......................................................... 14

5. Apoptosis....................................................................................................................................... 15

Characteristics ............................................................................................................................... 15

Molecular Basics of Apoptosis ..................................................................................................... 18

Assays for Apoptosis ..................................................................................................................... 18

Flow Cytometry ............................................................................................................................ 19

Probiotics Protection against Pathogen-induced Apoptosis .......................................................... 20

Objectives ............................................................................................................................................. 23

CHAPTER 2: METHODOLOGY ..................................................................................................... 24

La-5 Cell Free Spent Medium (CFSM) Preparation ............................................................................. 24

LH-2 Cell Free Spent Medium (CFSM) Preparation ............................................................................ 25

Salmonella Strain and Growth Conditions ............................................................................................ 26

Cell Culture and Maintenance............................................................................................................... 27

Trypan Blue Exclusion Assay ............................................................................................................... 28

Confocal Scanning Laser Microscopy (CSLM) .................................................................................... 28

Trans-epithelial Electrical Resistance (TEER) ..................................................................................... 30

Epithelial monolayer Integrity Challenge Study ................................................................................... 31

Lactate Dehydrogenase (LDH) Cytotoxicity Assay .............................................................................. 32

Apoptosis TUNEL Assay ...................................................................................................................... 33

Sulforhodamine B (SRB) Assay ........................................................................................................... 34

Invasion Assay ...................................................................................................................................... 35

Statistical Analysis ................................................................................................................................ 37

CHAPTER 3: RESULT AND DISCUSSION ................................................................................... 38

Trypan Blue Exclusion Assay ............................................................................................................... 38

Confocal Scanning Laser Microscopy .................................................................................................. 40

Page 6: Bioactive Components from Lactobacillus acidophilus and

v

Trans-epithelial Electrical Resistance (TEER) ..................................................................................... 43

Epithelial Barrier Integrity Study .......................................................................................................... 47

LDH Cytotoxicity Assay ....................................................................................................................... 54

Apoptosis Assay Using Flow Cytometry .............................................................................................. 58

SRB Assay ............................................................................................................................................ 66

Invasion Assay ...................................................................................................................................... 69

Conclusions ........................................................................................................................................... 71

CHAPTER 4 ........................................................................................................................................ 73

General Conclusions ............................................................................................................................. 73

Future Research .................................................................................................................................... 75

REFERENCE ...................................................................................................................................... 76

Page 7: Bioactive Components from Lactobacillus acidophilus and

vi

LIST OF TABLES

TABLE PAGE

CHAPTER 1

Table 1.1 In vitro studies focus on pathogenic bacteria inhibitory function of probiotics………………….5

Table 1.2 In vivo studies focus on pathogenic bacteria inhibitory function of probiotics……………….....6

Table 1.3 Bioactive peptides identification within the fraction of cell-free supernatant from L. helveticus

LH-2 fermented milk using mass spectrometry and NCBI databas. ………………………………………11

Table 1.4 Bioactive peptides identification within the fraction of cell-free spent modified MRS medium

fermented with L. acidophilus La-5 using mass spectrometry and BLASTp………………………………11

CHAPTER 2

Table 2.1 Bacterial strains used in these studies …………………………………………………………..27

Page 8: Bioactive Components from Lactobacillus acidophilus and

vii

LIST OF FIGURES

FIGURE PAGE

CHAPTER 1

Figure 1.1 Milk protein-derived bioactive peptides and their health promoting targets……………………9

Figure 1.2 Schematic representations of three forms of cell death. See context for detail description……17

Figure 1.3 Schematic representation of a flow cytometry….……………………………………………..22

CHAPTER 2

Figure 2.1 Catalytic function of the Lactate Dehydrogenase (LDH) enzyme……………………………..32

CHAPTER 3

Figure 3.1 La-5 and LH-2 toxic dose test on HT-29 cells proliferation, estimated by trypan blue exclusive

assay………………………………………………………………………………………………………..39

Figure 3.2 Confocal image of a single S. Typhimurium GFP cell .……………………………………….41

Figure 3.3 Merged confocal images of S. Typhimurium GFP invasion of HT-29 cells…………………...42

Figure 3.4 Determination of optimal HT-29 cell density in Transwell permeable inserts………………..45

Figure 3.5 Growth of HT-29 cells in Transwell permeable inserts………………………………………..46

Figure 3.6 Ratio of TEER of polarized HT-29 cell monolayers after exposure to Salmonella Typhimurium

to initial values.…………………………………………………………………………………………….50

Figure 3.7 Temperature dependent of TEER on polarized MDCK (Madin-Darby Canine Kidney) cells

grown on permeable inserts for 5 days ……………………………………………………………………54

Figure 3.8 Cytotoxicity (%) of HT-29 cells produced by S. Typhimurium at different times of exposure and

different MOI ……………………………………………………………………………………………..55

Figure 3.9 Evaluation of CFSMs incubation conditions………………………………………………….56

Figure 3.10 LDH % cytotoxicity of epithelial cells induced by Salmonella Typhimurium in the presence or

absence of CFSM treatment ………………………………………………………………………………57

Figure 3.11 Effect of CFSM on Salmonella Typhimurium induced apoptosis of polarized HT-29 cells...59

Figure 3.12 Apoptotic rate of polarized HT-29 cells and the protective effect of CFSM pre-incubation (24 h)

and co-incubation during exposure to Salmonella (1 h) (Flow cytometric TUNEL analysis)……………62

Figure 3.13 Cell viability by comparing efficiency of FITC (Y-axis)/PI (X-axis) dual stain signals……63

Figure 3.14 La-5 and LH-2 CFSM toxic dose test on LMH cells proliferation, estimated by SRB

colorimetric assay…………………………………………………………………………………………68

Figure 3.15 Invasion assay on LMH cell using Salmonella Typhimurium DT104 (SA2001-4368 &

SA2000-0406) isolated from chicken for 1 h infection with MOI of 10 …………………………………70

Figure 3.16 Invasion assay using LMH cells exposed to different infection conditions…………………70

Page 9: Bioactive Components from Lactobacillus acidophilus and

viii

LIST OF ABBREVIATIONS

ANOVA

ATCC

CDC

CFSM

CFU

CSLM

EHEC

FACS

FAO

FITC

GFP

GI Tract

IEC

IFN-γ

IgG

IL-8

La-5

LAB

LDH

LH-2

NLR

MDCK

MOI

OD

PBS

PE

PI

S-IgA

SPI

SRB

T3SS

TCA

TEER

TJ

TLR

TNF-α

TSA

TSB

TUNEL

WHO

Analysis-of-Variance

American Type Culture Collection

Center for Disease Control

Cell Free Spent Medium

Colony-Forming Unit

Confocal Scanning Laser Microscopy

Enterohemorrhagic Escherichia coli

Fluorescence-Activated Cell Sorting

Food and Agriculture Organization

Fluorescein Isothiocyanate

Green Fluorescent Protein

Gastrointestinal Tract

Intestinal Epithelial Cell

Interferon Gamma

Immunoglobulin G

Interleukin 8

Lactobacillus acidophilus La-5

Lactic Acid Bacteria

Lactate Dehydrogenase

Lactobacillus helveticus LH-2

Nod-like Receptor

Madin-Darby Canine Kidney

Multiplicity of Infection

Optical Density

Phosphate-Buffered Saline

Phycoerythrin

Propidium Iodide

Secretory IgA

Salmonella Pathogenicity Island

Sulforhodamine B

Type Three Secretion System

Trichloroacetic Acid

Trans-epithelial Electrical Resistance

Tight Junction

Toll-like Receptors

Tumor necrosis Factor Alpha

Trypticase Soy Agar

Trypticase Soy Broth

Terminal eoxynucleotidyl transferase dUTP nick end labeling

World Health Organization

Page 10: Bioactive Components from Lactobacillus acidophilus and

1

CHAPTER 1

Introduction

The public health effects and financial costs incurred following contamination of food with Salmonella

can be severe. In addition, the increase in antibiotic resistance of Salmonella has stressed the need to

find therapeutic alternatives. One such alternative may rest with bioactive peptides, especially those

produced following fermentation of milk by lactic acid bacteria (LAB). A variety of studies have

revealed the health promoting benefits of these peptides, including their ability to interfere with

virulence mechanisms of pathogens. Based on previous in vitro and in vivo observations made in our

laboratory, bioactive fractions from cell-free spent medium (CFSM) of L. acidophilus and L.

helveticus showed potent immunomodulating effects on the host and suppression of infectivity of

pathogens (Chin, 2002; Ding, Wang, & Griffiths, 2005; M. J. Medellin-Peña, 2007; Ng, 2000; Tellez

Garay, 2009). However, the mechanisms whereby CFSMs exert these protective functions on

epithelial cells are still not fully understood. We hypothesize that CFSMs enhance resistance of

epithelial cells against Salmonella infections by maintaining cell membrane integrity and constrain

Salmonella induced apoptosis. Herein, we propose to have a better comprehension of the mechanisms

behind these effects, which could support the use of bioactive components from LAB fermented milk

as supplements in functional food.

Page 11: Bioactive Components from Lactobacillus acidophilus and

2

Literature Review

1. Salmonella spp.

Salmonella are one of the main causes of human food-borne illnesses. Digestion of Salmonella

contaminated food leads to two major disease patterns in humans: a systemic disease (typhoid fever)

and a self-limiting gastrointestinal illness, salmonellosis (Caron et al., 2006). Non-typhoidal

Salmonella are the strains mainly associated with gastrointestinal infections. Approximately 1.2 million

illnesses, 23,000 hospitalizations, and 450 deaths result from non-typhoidal Salmonella infections

every year in the United States, resulting in an estimated $365 million dollars spent in direct medical

costs annually (CDC, 2013). No significant decline of Salmonella infection has been found in more than

a decade (CDC, 2011), whereas the increasing resistance of Salmonella to clinical-used antibiotics

continues to be noticed since 1996. In 2013, CDC showed that around 5% non-typhoidal Salmonella

were resistant to five or more types of antibiotic (CDC, 2013).

The taxonomy and nomenclature of the genus Salmonella used to be a prevalent topic (Tindall,

Grimont, Garrity, & Euzeby, 2005). Over 2500 serotypes are found in the Salmonella genus, and

possibly all of them are pathogenic (Burkholder & Bhunia, 2009). In 2007, the World Health

Organization (WHO) defined that Salmonella consisted of only two species according to molecular

detection, S. enterica and S. bongori. There are 6 subspecies in S. enterica, namely S. enterica subsp.

enterica; S. enterica subsp. salamae; S. enterica subsp. arizonae; S. enterica subsp. diarizonae; S.

enterica subsp. houtenae and S. enterica subsp. indica. Specifically, Salmonella enterica serovar

Typhimurium (S. Typhimurium) is one of the most common species involved in human food-borne

illnesses, and often contaminates poultry, pork, beef and dairy products (Burkholder & Bhunia, 2009;

Gaggìa, Mattarelli, & Biavati, 2010). As a highly antibiotic-resistant strain, S. Typhimurium DT104

Page 12: Bioactive Components from Lactobacillus acidophilus and

3

has attracted a great deal of public health attention (Burkholder & Bhunia, 2009; Wu, Carlson, &

Meyerholz, 2002). In addition to its clinical value, Salmonella is an interesting bacterial model to

examine for host-pathogen interaction since it can manipulate the functions of the host cells in order

to prolong its own survival. Its virulence functions are connected to Salmonella pathogenicity islands

(SPI1 and SPI2) and it possesses a needle-like Type Three Secretion System (T3SS) (Bayoumi &

Griffiths, 2010). Therefore, S. Typhimurium DT104 strains were selected in this study to test for in

vitro interference of epithelial cell functions.

2. Probiotics and Their Fermented Products

The Food and Agriculture Organization of the United Nations (FAO) and the World Health

Organization (WHO) defined probiotics as live microorganisms which, when administered in adequate

amounts, confer a health benefit on the host (FAO & WHO, 2001; Khani, Hosseini, Taheri, Nourani, &

Fooladi, 2012). Probiotics include bacteria, molds, and yeasts, among which lactic acid bacteria (LAB)

are one of the most intriguing groups due to their presence in yogurt, fermented milks and other

fermented foods. After Grigoroff isolated the first LAB in 1905 (Grigoroff, 1905), numerous studies

of LAB have been done to ensure LAB fermented foods have health benefits along with a GRAS

status (generally recognized as safe) (Divya, Varsha, Nampoothiri, Ismail, & Pandey, 2012). It is

reported in many reviews that benefits of consuming LAB and their bioactive components include: (1)

aiding in digestion and nutrient assimilation; (2) stimulating the immune system; (3) competing with

unfavorable pathogens and producing antimicrobial bioactive molecules; (4) preventing the risk of

certain cancers; (5) reducing the prevalence of allergy in susceptible individuals; (6) alleviating

symptoms of lactose intolerance and diarrhea; (7) lowering serum cholesterol concentrations and (h)

reducing blood pressure in hypertensive individuals (Divya et al., 2012; Khani et al., 2012; Masood,

Page 13: Bioactive Components from Lactobacillus acidophilus and

4

Qadir, Shirazi, & Khan, 2011; Parvez, Malik, Kang, & Kim, 2006; Schrezenmeir & De Vrese, 2001).

Although it is well known that consuming LAB fermented foods exert positive effects on human and

animal health, possible mechanisms behind these effects still remain largely unknown. The ingestion of

LAB is considered as a promising alternative for antibiotic in order to deal with pathogens‟ increased

antibiotic resistance. Many in vivo and in vitro studies have focused on using specific LAB strains, with

or without their culture medium, which exert antagonistic effects against various types of pathogens.

Tables 1.1 and 1.2 summarize information on protection afforded to the host against specific pathogens

by probiotics for both in vivo and in vitro studies, respectively.

Page 14: Bioactive Components from Lactobacillus acidophilus and

5

Table 1.1 In vitro studies focus on pathogenic bacteria inhibitory function of probiotics.

Probiotic Sample format Targeted

pathogen Findings Reference

Lactobacillus

plantarum 299v &

Lactobacillus

rhamnosus GG

Live microorganisms Escherichi coli Inhibited enteropathogenic E. coli adherence in vitro by

inducing intestinal mucin gene expression.

(Mack, Michail, Wei,

McDougall, & Hollingsworth,

1999)

Lactobacillus spp. Live microorganisms

Yersinia

enterocolitica

DSM4780

Affected the likelihood of Y. enterocolitica survival by

compromising urease functionality and cell viability.

(Lavermicocca, Valerio,

Lonigro, Di Leo, & Visconti,

2008)

Lactobacillus casei

strain Shirota

Live microorganism &

cell-free culture

supernatant

Helicobacter

pylori SS1

Inhibited H. pylori growth on solid agar as well as in liquid

medium with the presence of living L. casei strain Shirota. (Sgouras et al., 2004)

Lactobacillus casei

Rhamnosus Live microorganism

Escherichia coli

C25

Inhibited bacterial translocation of E. coli C25 in a

dose-dependent manner.

(Mattar, Drongowski, Coran,

& Harmon, 2001)

Lactobacillus casei

& Lactobacillus

acidophilus

Live microorganism Shigella sonnei Lactobacillus affected shigella growth rate. (Apella, Gonzalez, Demacias,

Romero, & Oliver, 1992)

Lactobacillus

acidophilus La5 cell-free spent medium

Escherichia coli

O157:H7

L. acidophilus La-5 secreted a molecule(s) that could block or

interfere with EHEC‟s virulence genes involved in

colonization.

(M. Medellin-Peña, Wang,

Johnson, Anand, & Griffiths,

2007; M. Medellin-Peña &

Griffiths, 2009)

Bifidobacterium

bifidum Cell-free spent medium

Salmonella

Typhimurium

Down-regulated S. Typhimurium reporter gene expression

driven by both hilA and ssrB at a dose dependent manner. (Bayoumi & Griffiths, 2010)

Bifidobacterium

bifidum

fraction from Cell-free

culture medium

Salmonella

Typhimurium &

Fraction #67 and #68 down-regulated S. Typhimurium and

E. coli reporter gene expression driven by hilA and ssrB, (Bayoumi & Griffiths, 2012)

Page 15: Bioactive Components from Lactobacillus acidophilus and

6

Escherichia coli LEE1, respectively; reduced pathogens colonization on

eukaryotic cells; diminished survival and multiply capacities

of Salmonella within macrophages.

Lactobacilli spp.,

Bifidobacteria spp.,

Lactococcus lactis

& Stococcus

thermophilus

Cell-free extracts from

fermented milk

Campylobacter

jejuni

Cell-free extracts from ten probiotic bacteria inhibited

expression of the C. jejuni flaA s28 promoter, which was

independent of pH and lactic acid concentration. Two

non-probiotic lactic acid bacterial strains, Lactococcus lactis

and Stococcus thermophilus, were less inhibitory.

(Ding et al., 2005)

Lactobacillus

crispatus K313 and

K243

S-proteins

Salmonella

Braenderup

H9812

Inhibited S. Braenderup H9812 adherence to HT-29 cells;

attenuated the response of HT-29 infected with S.

braenderup H9812.

(Sun et al., 2012)

Lactobacillus

reuteri & Bacillus

licheniformis

Live microorganism

Salmonella

Typhimurium &

Salmonella

Choleraesuis

S. Typhimurium stimulated secretion of IL-8 was inhibited

basolaterally in the presence of B. licheniformis.

(Skjolaas, Burkey, Dritz, &

Minton, 2007)

Table 1.2 In vivo studies focus on pathogenic bacteria inhibitory function of probiotics.

Probiotic Sample format Targeted pathogen Findings Reference

Lactobacillus casei

strain Shirota

Live microorganism

& cell-free culture

supernatant

Helicobacter pylori SS1

Administration of L. casei strain Shirota reduced colonizing H.

pylori viable counts and the associated inflammation of the

gastric mucosa in the H. pylori SS1 murine infection model.

(Sgouras et al., 2004)

Lactobacillus

salivarius UCC118 Live microorganism Listeria monocytogenes

Produced bacteriocin Abp118 that could significantly protect

mice against infection with the invasive foodborne pathogen L.

monocytogenes.

(Corr et al., 2007)

Page 16: Bioactive Components from Lactobacillus acidophilus and

7

Lactobacillus casei

GG Live microorganism Escherichia coli K1

Decreased the frequency of E. coli K1A translocation in a

neonatal rabbit model.

(Lee, Drongowski,

Coran, & Harmon, 2000)

Lactobacillus casei

& Latobacillus

acidophilus

Fermented milk

Listeria monocytogenes

& enteroinvasive

Escherichia coli

Higher survival rate, higher levels of anti-pathogen sera &

intestinal, less pathogen colonization of liver & spleen were

observed among LAB treated mice when challenged with

pathogens.

(Demacias, Romero,

Apella, Gonzalez, &

Oliver, 1993)

Lactobacillus casei Live microorganism Salmonella

Typhimurium

Oral administration of LAB increased the mucosal intestinal

immunity.

(Perdigon, Alvarez,

Demacias, Roux, &

Holgado, 1990)

Lactobacillus casei

& Lactobacillus

acidophilus

Ferment milk Salmonella

Typhimurium

Anti-salmonella protective immunity mainly mediated by the

mucosal tissue using L. casei + L. acidophilus mixture

fermented milk.

(Perdigon, Demacias,

Alvarez, Oliver, &

Holgado, 1990)

Lactobacillus

helveticus

A peptidic fraction

from fermented milk

Salmonella

Typhimurium

A peptide fraction composed of α-lactalbumin and β-casein

derived peptides played a dose-dependent role in protecting

mice against Salmonella translocation; mechanism involved

cell-mediated immune response and inference with virulence

gene expression.

(Tellez, Corredig,

Turner, Morales, &

Griffiths, 2011)

Lactobacillus

helveticus

cell-free fractions

from fermented milk

Bioluminescent

Salmonella Enteritidis

(lux CDABE)

Bioluminescence emitted by mice and the physical condition

of the mice indicated that animals fed with fermented milk or

fermented milk components prior to infection were less

susceptible to bacterial colonization and, subsequently,

bacteremia.

(Brovko et al., 2003)

Lactobacillus

acidophilus La5

cell-free spent

medium

enterohemorrhagic

Escherichia coli

(EHEC).

Cell-free spent medium fractions were able to down-regulate

several virulence genes of EHEC, including stxB2, qseA, luxS,

tir, ler, eaeA, and hlyB.

(Zeinhom et al., 2012)

Page 17: Bioactive Components from Lactobacillus acidophilus and

8

3. Bioactive Peptides

Definition and Characteristics

Bioactive peptides are defined as specific protein fragments that have a positive impact on body

functions and conditions, ultimately benefiting health (Kitts & Weiler, 2003). Most bioactive peptides

are obtained from proteins of animal origin, such as milk, egg, gelatin, fish, as well as plant proteins

such as wheat gluten and soy (Elawadli, 2012). During digestion in the gastrointestinal tract, bioactive

peptides can be generated from inactive parental protein sequences through three ways of proteolysis:

(1) enzymatic hydrolysis by digestive enzymes, (2) fermentation with proteolytic starter cultures and

(3) proteolysis by enzyme derived from microorganisms or plants (Korhonen, 2009; Muro Urista,

Alvarez Fernandez, Riera Rodriguez, Arana Cuenca, & Tellez Jurado, 2011). A combination of the

above methods could also be used in the effective generation of short functional peptides (Korhonen,

2009). The size of the active peptides ranges from two to twenty amino acid residues. Their

bioactivities rely on the parental protein source and the composition of amino acid sequences they

have inherited. Many peptides are known to exhibit multi-functional properties (Meisel & FitzGerald,

2003).

Bioactive Peptides Derived from Milk

Bioactive peptides derived from milk protein have received increasing attention due to their positive

health-promoting effects on digestive, endocrine, cardiovascular, immune and nervous sytems

(Korhonen, 2009). Research done on peptide sequences have expanded our horizons allowing us to

have a better understanding of antimicrobial, antioxidative, antithrombotic, antihypertensive,

immunomodulatory and opioid activities that bioactive peptides possess (Muro Urista et al., 2011;

Page 18: Bioactive Components from Lactobacillus acidophilus and

9

Silva & Malcata, 2005). Figure 1.1 exhibits the potential health benefits of various milk protein

derived bioactive peptides. Moreover, it is revealed that LAB have an ability to degrade milk protein

in order to fulfill their nutritional requirements for essential amino acids (Benkerroum, 2010).

Because of the many different sources of proteinases and their modes of action, bioactive peptides

from milk hydrolysed by lactic acid bacteria or their proteases represent a fundamental difference

from those generated by digestive proteinases (Benkerroum, 2010). As a result, there has been

growing scientific and commercial interest on the evaluation of milk fermentation with microbial

proteolysis on human health; specifically on reducing the risk of chronic diseases or enhancing natural

immune protection (Hartmann & Meisel, 2007).

Figure 1.1 Milk protein-derived bioactive peptides and their health promoting targets, adapted from Korhonen,

2009.

Page 19: Bioactive Components from Lactobacillus acidophilus and

10

The wide range of nutritional, functional and biological benefits of milk proteins place them as the

most valuable source of bioactive peptides at present (Korhonen, 2009). The protein concentration of

bovine milk is about 32 g/L, 80% of which are caseins and 20% of which are whey proteins. LAB

prefer the substrate casein due to its porous structure and poor solubility but whey protein also

undergoes limited degradation (Griffiths & Tellez, 2013). Caseins can be divided into α-, β- and

κ-caseins (Muro Urista et al., 2011). The whey fraction contains α-lactalbumin, β-lactoglobulin and

other proteins, e.g., immunoglobulins, lactoferrin and serum albumin (Ebringer, Ferencik, &

Krajcovic, 2008; Haug, Hostmark, & Harstad, 2007).

Since the functionality of peptides is closely related to parental protein in milk, a great number of

peptide sequences with specific functionality in caseins and whey milk proteins have been identified

(Muro Urista et al., 2011). Previous studies on peptide isolation and purification have been conducted

in our laboratory. Tellez Garay (2009) found five putative bioactive peptides from a cell-free

supernatant of L. hevleticus LH-2 fermented milk. The fraction, which included these five peptides,

showed an immunomodulatory effect and antagonistic effect against Salmonella infection in both in

vitro and in vivo studies (Tellez Garay, 2009). Table 1.3 shows the five putative bioactive peptide

sequences within this fraction. Other related research carried out by Medellin-Peña (2007) found three

bioactive peptides in cell-free spent modified MRS medium (CFSM) fermented with L. acidophilus

La-5. The bioactive molecule(s) from the CFSM not only down-regulated different virulence genes in

enterohemorrhagic E. coli (EHEC) O157:H7 but also interfered with E. coli quorum sensing. Both in

vitro and in vivo studies indicated increased resistance against infection and colonization with EHEC

O157:H7 when biologically active La-5 CFSM fractions were applied (M. J. Medellin-Peña, 2007).

Table 1.4 shows the three bioactive peptide sequences from biological active La-5 CFSM fractions.

Page 20: Bioactive Components from Lactobacillus acidophilus and

11

Furthermore, databases (BIOPEPE etc.) and programs (BLAST etc.) are available to simulate and

demonstrate proteolysis processes targeted at obtaining expected bioactive peptides from precursor

protein (Minkiewicz, Dziuba, Iwaniak, Dziuba, & Darewicz, 2008).

Table 1.3 Bioactive peptides identification within the fraction of cell-free supernatant from L. helveticus LH-2

fermented milk using mass spectrometry and NCBI database, adapted from Tellez Garay, 2009.

Peptide Protein Sequence assignment

HQPHQPLPPTVMFPPQ β –Casein 145–160

HQPHQPLPPT β –Casein 145–154

WMHQPHQPLPPT β –Casein 143–154

LYQEPVLGPVR β –Casein 192–202

LDQWLCEK α-Lactalbumin 115–122

Table 1.4 Bioactive peptides identification within the fraction of cell-free spent modified MRS medium

fermented with L. acidophilus La-5 using mass spectrometry and BLASTp, adapted from M. J. Medellin-Peña,

2007.

Peak sequence/sequence aligned BLASTp protein

YPVEPF/YPVEPF YP 194702 ncopullulanase

[L. acidophilus NCFM]

YPPGGP/YPPG YP 193877 ornithine decarboxylase chain A

[L. acidophilus NCFM]

NQPY/NQPY YP 193484 glutamine ABC transporter

[L. acidophilus NCFM]

Page 21: Bioactive Components from Lactobacillus acidophilus and

12

4. Intestinal Epithelial Cells

Enterocytes in the gastrointestinal (GI) tract consist of a single monolayer of intestinal epithelial cells

(IECs) and this monolayer is the frontline in host defense. The IEC is the most important physical

barrier that separates the vast number of microbes in the intestinal lumen from the host tissue. IEC also

plays a role in innate immune defense since the GI tract has the greatest number of lymphoid organs in

the human body, which can interface with a myriad of internal and external stimuli (Ouwehand,

Salminen, & Isolauri, 2002). IEC helps monitor the luminal bacterial signals in the GI tract, interpreting

and transmitting this information to mucosal innate and adaptive immune cells in the lamina propria,

and collaborates with lymphoid tissue to initiate an immune response to certain stimuli (Goto & Ivaylo,

2013; Pinto et al., 2009).

Epithelial Cells Polarization

In vitro models using isolated epithelia are invaluable for observing intracellular activities in response

to external stimuli. The most common epithelial cell lines used in studies are small intestine isolated

carcinoma Caco-2 cells and large intestine isolated carcinoma HT-29 cells for their ability to better

mimic the natural intestinal mucosal barrier when they are polarized (Backert, Boehm, Wessler, &

Tegtmeyer, 2013). Numerous in vitro studies revealed that epithelial cell polarization improves

relevance of real human intestinal epithelia. Human IEC, together with in vitro polarized epithelial

cells are both endowed with physiological, and biochemical characteristics as well as structural

markers. One of the most distinct structural features is the tight junction (TJ). TJs form between

adjacent cells leading to apical and basolateral cell membrane formation. Additionally, a brush border

composed of microvilli can be observed on the apical cell surface. Each microvillus possesses a

bundle of actin filaments, which are cross-linked by numerous actin-bundling proteins like villin,

Page 22: Bioactive Components from Lactobacillus acidophilus and

13

fimbrin and espin to form the microvilli structural core (Chalghoumi et al., 2009; Cohen, Ophir, & Ben

Shaul, 1999; Le Bivic, Hirn, & Reggio, 1988; Rodriguez-Boulan & Nelson, 1989).

Transwell permeable inserts with membrane filters in cell culture well plates have long been adopted

for cross-epithelial transport assessment (Hubatsch, Ragnarsson, & Artursson, 2007; Zemans et al.,

2011). The permeable insert is reported to be beneficial for epithelial cell polarization and

differentiation. Due to the similar procedure for polarization of HT-29 cells in the permeable inserts to

that observed during intestine embryonic development, HT-29 cells are utilized in epithelial

differentiation studies (Le Bivic et al., 1988). When cultured in glucose for a certain period of time,

HT-29 cells form very tight junctions and microvillus brush border at the apical surface, indicating cell

polarization (Fitzgerald, Omary, & Triadafilopoulos, 1997; Höner zu Bentrup et al., 2006).

Consequently, their resemblance to native IEC makes this model more representative than cells grown

in mono-culture. Even though mono-cultures of epithelial cells are still useful and continue to be used,

their restrictions, such as the inability to transport agents (from small molecules to immune cells)

across the epithelial monolayer, are concerned (Höner zu Bentrup et al., 2006).

Trans-epitheliall Electrical Resistance

The increase in Trans-epithelial Electrical Resistance (TEER) indicates the polarized status of

epithelial cells grown in Transwell permeable inserts. The measurement of TEER is a well-developed

approach to quantitatively monitor tight junction integrity and qualitatively observe monolayer health

(Grajek & Olejnik, 2004). The Millicell ERS (EMD Millipore, Billerica, MA, USA) is a device

designed to facilitate measurements of TEER to determine cultured epithelial monolayer integrity

directly in tissue culture wells. The Millicell ERS-2 uses alternating currents to eliminate adverse

effects on the cell membrane. It contains a silver electrode with a fixed pair of probes. These “chopstick”

Page 23: Bioactive Components from Lactobacillus acidophilus and

14

like probes can measure the voltage deflection at 37°C in tissue culture medium (Balda et al., 1996).

TEER evaluates tight junction formation under a wide variety of experimental conditions (Ferrell et al.,

2010; Höner zu Bentrup et al., 2006). This model has provided invaluable insights into the intracellular

events that occur in response to pathogenic bacterial stimuli. Solano et al. (2001) used TEER

measurement as one of the methods to compare the virulence of different Salmonella Enteritidis

strains, since the more virulent strains produce a greater disruption of the epithelial cell monolayer

(Solano et al., 2001). In addition, probiotic bacteria have been reported to strengthen the intestinal

barrier function as well as limit pathogen induced TEER loss. L. plantarum MB452 (Anderson et al.,

2010), St. thermophilus, L. acidophilus (Resta-Lenert & Barrett, 2003) and L. amylophilus D14 (Yu,

Wang, & Yang, 2012) inhibited pathogen-induced cell junction loss and mucosal barrier damage.

Epithelial Cells Immune Response to Salmonella Invasion

The intestine contains a great deal of immunoglobulin producing cells, e.g., IgA, comprising the

largest immunological organ in the body (Tellez Garay, 2009). The epithelial cells located in the

gastrointestinal tract collaborate with intraepithelial lymphocytes by possessing Toll-like receptors

(TLRs) and Nod-like receptors (NLRs) on their surface to sense the existence of pathogens or

pathogen-associated molecular patterns (Magrone & Jirillo, 2013; Tlaskalova-Hogenova et al., 2002).

Furthermore, cytokines secretion is also involved in the immune responses to pathogen invasion.

Cytokines are secreted soluble polypeptide or glycoprotein which play diverse biological roles in

normal and pathological events, such as cell growth, differentiation, and immune response activation

(Steinke & Borish, 2006; Zanabria Eyzaguirre, 2013). They may affect other cytokines production and

action in order to achieve regulated immune response by balancing between provoking and

suppressive influences (Zanabria Eyzaguirre, 2013).

Page 24: Bioactive Components from Lactobacillus acidophilus and

15

Cytokines play essential roles in apoptosis and inflammation on epithelial cells. Accordingly,

Salmonella infection in all organs stimulates IL-1 and TNF-α expression (Eckmann & Kagnoff, 2001).

In response to bacterial endotoxin, the first released cytokine is tumor necrosis factor TNF (Zanabria

Eyzaguirre, 2013). Moreover, TNF often triggers the initiation and development of apoptotic and

inflammatory processes (Benderska et al., 2012). Even though the detail of the cytokine‟ function still

remain to be elucidated, Eckmann & Kagnoff (2001) assumed that they may have indirect functions,

like overall endothelial adhesion molecules up-regulation in order to initiate macrophage functions.

IFN-γ is another most studied cytokine, which plays an important role in the host defense against

Salmonella, since production of IFN-γ is readily detectable in Salmonella infected mice (Eckmann &

Kagnoff, 2001). Kagaya et al., (1989) hypothesized that the most likely mechanism of operation of

IFN-γ is the ability to activate macrophages to kill Salmonella (Kagaya, Watanabe, & Fukazawa,

1989).

5. Apoptosis

Characteristics

Programmed cell death is essential in the hosts‟ normal development. Not only does it help with organs

differentiation, but it also eliminates useless and dangerous cells. It serves as a balance with cell

division to maintain the normal cell number over time (Raff, 1992). Salmonella manipulates the cell

death pathway of mammalian cells by inducing their apoptosis (Kim et al., 1998). It is hypothesized

that their ability to trigger apoptosis is a crucial step in the pathogenesis of Salmonella (Ashida et al.,

2011; Gobbato, Galdeano, & Perdigon, 2008; Knodler, Finlay, & Steele-Mortimer, 2005). By inducing

cell death, previously replicated Salmonella inside the host cells are released in the late apoptosis phase

by evading the immune response, hence more effectively re-infecting other cells, resulting in systemic

Page 25: Bioactive Components from Lactobacillus acidophilus and

16

infection (Ashida et al., 2011; De Moreno De Leblanc et al., 2010; Knodler & Finlay, 2001). Yet this

mechanism is not fully proven whether apoptosis is a direct outcome of pathogen infection or is the

result of inflammatory mediators released by the host (Torchinsky, Garaude, & Blander, 2010). Indeed,

Kim et al. (1998) proposed that human colon epithelial cells generated inflammatory mediators in

response to bacterial invasion. Some of these mediators also could induce apoptosis (Kim et al.,

1998).

The term apoptosis was first introduced in 1972 by Kerr et al., who differentiated a naturally

programmed cell death from severe tissue injury necrosis (Kerr, Wyllie, & Currie, 1972). There are

three types of cell death involved in mammalian cells categorized by morphological criteria, namely

apoptosis, autophagy, and necrosis. Apoptotic cells exhibit alterations to their nuclear morphology,

including DNA fragmentation, chromatin condensation, membrane blebbing, overall cell shrinkage,

and formation of apoptotic bodies that contain nuclear or cytoplasmic material (Elmore, 2007;

Hans-Jurgen, 2008). Autophagy is characterized by an extensive accumulation of autophagosomes

(double-membrane vacuoles) followed by fusion with lysosomes, leading to the degeneration of

contents of autophagosomes. Cytoplasmic swelling is the characteristic of Necrosis, and the swelling

occurs until the intracellular contents are released through the ruptured plasma membrane resulting in

pro-inflammatory leakage (Hans-Jurgen, 2008). Figure 1.2 shows the different morphological

alterations in the three forms of cell death. Among the three types, apoptosis has been studied

extensively since it is a major form of removing unwanted and harmful cells with little inflammatory

response and little disturbance of tissue homeostasis in the host (Ashida et al., 2011; de LeBlanc,

Castillo, & Perdigon, 2010; Knodler & Finlay, 2001).

Page 26: Bioactive Components from Lactobacillus acidophilus and

17

Figure 1.2 Schematic representations of three forms of cell death. See context for detail description, adapted

from Hans-Jurgen, 2008.

Page 27: Bioactive Components from Lactobacillus acidophilus and

18

Molecular Basics of Apoptosis

The molecular mechanism of apoptosis is dependent on a family of cysteine proteases called caspases.

They mediate apoptosis with irreversible activation of proteins involved in DNA repair, DNA

replication, and RNA splicing (Santini, Rainaldi, & Indovina, 2000). Even though all members of the

caspase family have some overlapping amino acid sequences and possess similar structures,

individual caspases play different physiological roles in apoptosis or inflammatory responses (Fink &

Cookson, 2005). There are two major signaling routes to initiate caspase activation: the extrinsic death

receptor pathway and the intrinsic mitochondrial pathway (Ashida et al., 2011; Elmore, 2007;

Hans-Jurgen, 2008). Extracellular signals such as toxins (Popov et al., 2002), hormones, growth

factors, nitric oxide (Brune, 2003) and cytokines (Benderska et al., 2012), either penetrate through the

plasma membrane or transduce through membrane receptors to activate the executioners caspase-3

and -7 (Ashida et al., 2011). These signals may trigger (positively affect) or inhibit (negatively affect)

apoptotic activity in order to maintain tissue homeostasis, especially in the immune system (Ashida et

al., 2011; Hans-Jurgen, 2008; Levi et al., 2014). Intrinsic signaling pathways are mitochondrial

initiated events involved with a diverse array of non-receptor mediated stimuli, which can activate

caspase-9 production and directly aim at targets within the cells (Ashida et al., 2011). The “cross-talk”

between the extrinsic (death receptor) pathway and the intrinsic (mitochondrial) pathway leads to the

final execution phase of apoptosis (Elmore, 2007). In this phase, morphological and biochemical

alterations such as DNA fragmentation and membrane blebbing appear (Slee, Adrain, & Martin,

2001).

Assays for Apoptosis

Due to the distinct features of apoptotic cells, there are a variety of assays to determine apoptotic

Page 28: Bioactive Components from Lactobacillus acidophilus and

19

activity activators, effectors and regulators and count the functional consequences of their actions.

These assays can be classified into five categories: (1) morphological alterations detected by

microscopy (Elmore, 2007; Fink & Cookson, 2005); (2) DNA fragmentation detected by DNA

laddering (Compton, 1992) and TUNEL (Fink & Cookson, 2005; Hans-Jurgen, 2008); (3) membrane

alterations detected by Annexin-V binding (van Engeland, Nieland, Ramaekers, Schutte, &

Reutelingsperger, 1998), LDH and impermeable DNA dyes (Elmore, 2007; Fink & Cookson, 2005;

Hans-Jurgen, 2008); (4) caspase activation detected by western blotting, PCR microarray (Elmore,

2007), colorimetric or fluorometric assays (Fink & Cookson, 2005; Hans-Jurgen, 2008); (5)

mitochondrial damage detected by ATP production and mitochondrial dyes (Elmore, 2007;

Hans-Jurgen, 2008)

Flow Cytometry

Amongst the assays previously described, the detection of DNA fragmentation is currently the most

frequently used method when studying apoptosis (Fink & Cookson, 2005) together with flow

cytometry, which can facilitate apoptosis detection. Flow cytometry, also referred to as

fluorescence-activated cell sorting (FACS), is laser-based biophysical method, which allows

multi-parameter measurements. It requires samples prepared in single cell format, to be measured in a

fluid stream, which will be spectrophotometrically measured by the laser light. The properties of the

designated single cell are acquired, including cell size (shown on forward-scattered light graph),

conformation of inner structure (shown on side-scattered light graph), and relative fluorescence

intensity (fluorescent intensity graph).

Fluidics, optics and electronics are the three main systems that make up the flow cytometer. The fluidics

Page 29: Bioactive Components from Lactobacillus acidophilus and

20

system transports cells in single file through a narrow nozzle with arranged sheath fluid. A fluorescent

photon resonance vibrating mechanism helps separate the stream of cells into individual droplets. The

optics system consists of lasers at different wavelengths to illuminate cells in the same stream and

optical filters to channel designated light to the appropriate detection system. Last, but not least, the

electronics system converts detected light signals into a measurement that can be analyzed by computer

software. As a result, electronic signals can be interpreted after adjusting the sorting settings of the

software in order to obtain valuable information. Dong et al. (2008) concluded that flow cytometry was

a versatile method of detecting the apoptotic process, including morphological alteration and chromatin

shrinkage, activation of caspases, DNA condensation, etc. (Dong, Kleinberg, Davidson, & Risberg,

2008). Figure 1.3 provides a schematic representation of a flow cytometer (Jahan-Tigh, Ryan,

Obermoser, & Schwarzenberger, 2012).

Apoptotic cells share noticeable characteristics, which can be captured by flow cytometry with

appropriate fluorescent labels or dyes. From the data, it is able to differentiate the different phases of

cell death (Vermes, Haanen, & Reutelingsperger, 2000). In addition, flow cytometry allows several

measurements to be obtained on each cell simultaneously by applying different stains such as

fluorescein isothiocyanate (FITC), propidium iodide (PI), or phycoerythrin (PE) (Corver, Cornelisse,

& Fleuren, 1994). The stains‟ fluorescent intensity can be quantified using CellQuest™ software (BD

Bioscience, Mississauga, ON, Canada). In this study, the Apo-Direct™ Kit (BD Bioscience,

Mississauga, ON, Canada) was employed to assess DNA strand breakage using FITC/PI dual stains.

Probiotics Protection against Pathogen-induced Apoptosis

Although the detailed mechanisms of pathogen-induced apoptosis are still under evaluation, the

Page 30: Bioactive Components from Lactobacillus acidophilus and

21

hypothesis that probiotic strains or bioactive components from LAB culture medium may interfere

with pathogen-induced apoptosis was investigated. Myllyluoma et al. (2008), performed a 1 h

pretreatment of H. pylori infected polarized Caco-2 cells in the presence of L. rhamnosus GG, L.

rhamnosus Lc705 and B. breve Bb99 cells and found that they reduced caspase-3 production

significantly at 24 h post-infection (Myllyluoma, Ahonen, Korpela, Vapaatalo, & Kankuri, 2008). An in

vitro study undertaken by Valdez et al. (2001) showed that the probiotic strains L. delbrueckii subsp.

bulgaricus and St. thermophilus down-regulated Salmonella induced apoptosis in macrophages. In this

case, the inhibitory effect did not seem to be related to an increase in the uptake of LAB, but more to

secretory IgA (S-IgA) production. The latter is an immunoglobulin known to be associated with

inhibition of pathogen internalization and toxin neutralization (Mantis & Forbes, 2010). In addition to

pro-inflammatory cytokine production, such as TNF-α, oxidant radicals (superoxide, hydrogen

peroxide and nitric oxide) released from macrophages stimulated by LAB may also partially explain the

protective capacity of LAB (Valdez, Rachid, Gobbato, & Perdigon, 2001). Further, an in vivo study by

Gobbato et al. (2008) found administration of L. delbrueckii subsp. bulgaricus and St. thermophilus

strains improved microbiocidal activity and down-regulated apoptosis produced by S. Typhimurium

infection, possibly through increasing cytokine IFN- γ, immunoglobulin IgA, and apoptosis related

protein Bcl-2 release (Gobbato et al., 2008).

There are a limited number of publications describing the putative mechanisms of the anti-apoptotic

effect on epithelial cells induced by cell free preparations of probiotics against pathogens. Li et al.

(2001) showed that Lactobacillus acidophilus S-layer protein inhibited caspase-3 production by

Caco-2 cells following Salmonella infection. In addition, the extracellular signal-regulated kinase 1 and

2 (ERK 1/2) signaling pathway plays a fundamental role in epithelial recovery after pathogen infection,

Page 31: Bioactive Components from Lactobacillus acidophilus and

22

and the L. acidophilus S-layer protein activated the signaling pathway, reducing further apoptotic cell

damage (Li, Yin, Yu, & Yang, 2011). Yan et al. (2007), found Lactobacillus rhamnosus GG (LGG)

soluble factors p75 and p40 could regulate signaling pathways in order to prevent cytokine-induced

apoptosis in human and mouse intestinal epithelial cells. Soluble factors p75 and p40 not only

suppressed cytokine-induced apoptosis, but also promoted cell survival by stimulating Akt related

proapoptotic pathways (Yan et al., 2007).

Figure 1.3 Schematic representation of a flow cytometry (adapted from Jahan-Tigh et al., 2012). Samples are

prepared in single cell format and are labeled by fluorochrome-linked antibodies or stained with fluorescent

nuclear, cytoplasmic, or membrane dyes. The labeled/stained cells go through a beam of laser light with known

speed and position. A set of optical system is used to detect forward-scatter, side-scatter and fluorescence. Lastly,

an electronic system converts these signals into electronic signals on computer-based software.

Page 32: Bioactive Components from Lactobacillus acidophilus and

23

Objectives

Bioactive peptides from LAB fermentations have shown potential as a therapeutic alternative of

clinic-dependent antibiotic, therefore, attracted attention both from academic research and related

industries. Based on previous in vitro and in vivo studies in our laboratory, cell-free spent medium

(CFSM) of L. acidophilus and L. helveticus showed potent immunomodulatory function on the host

and interference of pathogens virulence (Chin, 2002; Ding et al., 2005; M. J. Medellin-Peña, 2007; Ng,

2000; Tellez Garay, 2009). Our hypothesis that CFSMs reinforce epithelial cell membrane integrity

when invaded by Salmonella is tested in four individual steps.

1. Using Trypan Blue Exclusion, LDH Assay and SRB Assay to determine the non-toxic dose of

bioactive compounds from L. acidophilus or L. helveticus fermented milk (La-5 or LH-2) when

applied on epithelial cells;

2. Examine the protective effect on epithelial cell membrane integrity present in the bioactive

components against Salmonella infection through TEER Measurement and LDH Assay;

3. Employing TUNEL Assay to investigate down-regulation of Salmonella-induced apoptosis in

epithelial cells when La-5 and LH-2 are applied;

4. Evaluate Salmonella internalization interference of La-5 and LH-2 in an in vitro chicken

model by counting amount of invaded Salmonella.

Page 33: Bioactive Components from Lactobacillus acidophilus and

24

CHAPTER 2: Methodology

La-5 Cell Free Spent Medium (CFSM) Preparation

Lactobacillus acidophilus La-5 strain was obtained from the culture collection of the Canadian

Research Institute for Food Safety (University of Guelph, Guelph, Canada), streaked on Lactobacillis

MRS Agar (BD Difco™, Mississauga, ON, Canada) and incubated under anaerobic conditions with a

BBL GasPak system (BD Bioscience, Mississauga, ON, Canada) at 37 °C for 48 h. Chemically defined

medium (CDM) was composed of 28 g whey protein isolate (Ergogenics Nutrition, Vancouver, BC,

Canada), and 10 ml of 0.25 g/ml sterile sucrose solution (Sigma, Markham, ON, Canada) in 500 ml

sterile distilled water. The CDM medium (500 ml) was inoculated with L. acidophilus La-5 (50 colonies

removed from an MRS agar plate cultured as described above) and incubated anaerobically with a BBL

GasPak system (BD Bioscience, Mississauga, ON, Canada) at 37°C for 48 h. Following growth,

bacterial cells were removed by centrifugation at 12,000 × g for 30 min at 4 °C (Avanti J-20 XPI,

Beckman Coulter, Canada). The supernatant was collected and sequentially filtered through a 0.7 μm

pore-size filter (EMD Millipore, Billerica, MA, USA), and then through a 0.45 μm pore-size filter

(Fisher Scientific, Mississauga, ON, Canada) in order to remove any bacterial cells present. The

cell-free preparation was frozen at -80°C (Thermo Fisher Scientific, Mississauga, ON, Canada),

followed by freeze drying (Unitop 600 SL, VirTis Co., Inc. Gardiner, NY, USA). The freeze dried

samples were stored at -80 °C (Thermo Fisher Scientific, Canada). The freeze-dried supernatant was

reconstituted with cell culture grade water (Water for Injection, WFI, Life technologies, Burlington, ON,

Canada) to 1/10 of its original volume before use. Portions of the reconstituted samples were also kept

at -20°C until analyzed. The protein concentration was measured spectrophotometrically using the

Page 34: Bioactive Components from Lactobacillus acidophilus and

25

Protein A280 program in Nanodrop according to manufacturer‟s instruction (ND-1000, Thermo Fisher

Scientific, Mississauga, ON, Canada). The protein concentration of La-5 CFSM was 99.5 ± 0.4 mg/ml.

LH-2 Cell Free Spent Medium (CFSM) Preparation

Lactobacillus helveticus LH-2 strain was obtained from the culture collection of the Canadian Research

Institute for Food Safety (University of Guelph, Guelph, Canada), streaked on Lactobacillis MRS Agar

(BD Difco™, Mississauga, ON, Canada) and incubated under anaerobic conditions with a BBL GasPak

system (BD Bioscience, Mississauga, ON, Canada) at 37 °C for 48 h. Skim milk powder (Smucker's,

Markham, ON, Canada) was reconstituted 10% (w/w) with distilled water. Reconstituted skim milk was

heat treated at 95°C for 30 min and then cooled in the biosafety cabinet until it reached room

temperature. Isolated colonies of the L. helveticus strain were used to inoculate four replicates of 5 ml

sterilized reconstituted milk, which were then incubated anaerobically with a BBL GasPak system (BD

Bioscience, Mississauga, ON, Canada) at 37°C for 24 h, followed by aerobic incubation at the same

temperature for 26 h. The fermented milk samples were centrifuged at 16,000 × g for 10 min at 15 °C

(Avanti J-20 XPI, Beckman Coulter, Canada). The supernatant was filtered through a 0.2 μm pore-size

filter (EMD Millipore, Billerica, MA, USA) in order to remove any bacterial cells present. From 800 ml

of fermented milk, 500 ml of supernatant were obtained. The cell-free supernatant (10 ml) was

dispensed in 50 ml tubes and was frozen at -80°C (Thermo Fisher Scientific, Mississauga, ON, Canada),

followed by freeze drying (Unitop 600 SL, VirTis Co., Inc. Gardiner, NY, USA). The freeze dried

samples were stored at -80 °C (Thermo Fisher Scientific, Canada). The freeze-dried supernatant was

reconstituted with cell culture grade water (Water for Injection, WFI, Life technologies, Burlington, ON,

Canada) to 1/10 of its original volume before use. Portions of the reconstituted samples were also kept

Page 35: Bioactive Components from Lactobacillus acidophilus and

26

at -20°C until analyzed. The protein concentration was measured spectrophotometrically using

Nanodrop (ND-1000, Thermo Fisher Scientific, Mississauga, ON, Canada) as described above. The

protein concentration of LH-2 CFSM was 37.1 ± 0.1 mg/ml.

Salmonella Strain and Growth Conditions

A human isolate (SA1997-0934) and two chicken isolates (SA2000-0406, SA2001-4368) of

Salmonella Typhimurium DT104 along with a GFP expressing mutant of Salmonella Typhimurium

were obtained from the culture collection of the Canadian Research Institute of Food Safety (University

of Guelph, Guelph, Canada). All strains were stored in 25% glycerol (v/v) (Fisher Scientific, Canada) at

-80°C, and transferred once to Trypticase Soy Agar (TSA, BD Bioscience, Mississauga, ON, Canada).

Following overnight growth at 37°C, a colony was removed aseptically from the plate and transferred

into 5 ml Trypticase Soy Broth (TSB, BD Bioscience, Mississauga, ON, Canada), which was then

incubated at 37°C with shaking (200 rpm; orbitary shaker incubator, New Brunswick Scientific, USA)

for 18~19 h. An aliquot (50 μl) of this culture was added into 5 ml of fresh TSB, and incubated at 37°C

with shaking for 4 h. By the end of 4 h incubation period, all S. Typhimurium strains were in the late

exponential phase and inoculum absorbance at OD600 was adjusted to 1.00. The bacteria count was

approximately 1 × 109 CFU (colony forming unit)/ml (= 9 log10 CFU/ml).

All the strains used in these experiments are listed in Table 2.1.

Page 36: Bioactive Components from Lactobacillus acidophilus and

27

Table 2.1 Bacterial strains used in these studies

Species Strain No Source Gram stain Growth conditions

Salmonella Typhimurium

SA 1997-0934 CRIFS* Negative

TSB/TSA, 37°C SA 2000-0406 CRIFS

* Negative

SA 2001-4368 CRIFS* Negative

GFP CRIFS* Negative

Lactobacillus acidophilus La-5 CRIFS* Positive MRS, 37 °C

Lactobacillus helveticus LH-2 CRIFS* Positive MRS, 37 °C

* CRIFS: Canadian Research Institute for Food Safety, University of Guelph, ON, Canada

Cell Culture and Maintenance

The human colonic carcinoma cell line HT-29 was obtained from the culture collection of the Canadian

Research Institute of Food Safety (University of Guelph, Guelph, Canada). Cells were cultured in

Dulbecco‟s modified Eagle medium (DMEM, high glucose, Life technologies, Burlington, ON, Canada)

supplemented with 1% (v/v) 10, 000 U/mL penicillin–streptomycin (Life technologies, Burlington, ON,

Canada) and 10% heat-inactivated fetal bovine serum (HI FBS, Life technologies, Burlington, ON,

Canada). The HT-29 cells (20–40 passages) were kept in a humid atmosphere of 5% CO2 at 37°C in an

incubator (Forma™ Series II 3110 Water-Jacketed CO2 Incubators, Thermo Fisher Scientific,

Mississauga, ON, Canada), and grown as a monolayer in 25 or 75 cm2 flasks (Corning, NY, USA). The

medium was changed every two days and passed (80-90% confluence) using 0.25% trypsin-EDTA

reagent (Life technologies, Burlington, ON, Canada).

The chicken hepatoma cell line, LMH (ATCC CRL-2117) was purchased from American Type Culture

Collection (ATCC, Manassas, VA, USA). Cells were cultured in Waymouth‟s MB 752/1 medium (Life

technologies, Burlington, ON, Canada) supplemented with 1% (v/v) 10, 000 U/mL penicillin–

streptomycin (Life technologies, Burlington, ON, Canada) and 10% fetal bovine serum (FBS, Life

technologies, Burlington, ON, Canada). LMH cells (6-23 passages) were kept in a humid atmosphere of

Page 37: Bioactive Components from Lactobacillus acidophilus and

28

5% CO2 at 37°C, and grown as a monolayer in 0.1% gelatin (PCS-999-027, Cedar Lane Laboratories,

Burlington, Canada) coated 25 or 75 cm2 flasks (Corning, NY, USA). The medium was changed every

two days and was passed (80-90% confluence) using 0.25% trypsin-EDTA reagent (Life technologies,

Burlington, ON, Canada).

Trypan Blue Exclusion Assay

This assay based on the principle that viable cells have intact cell membranes which can resist trypan

blue dye penetration while dead cells do not. Hence live cells can be easily differentiated and counted

with a light microscope using trypan blue (Strober, 2001). Viable cells are clear, unstained, small, and

round under light microscopy, while dead cells are blue, stained, and swollen (Louis & Siegel, 2011) .

For this test, approximately 1 × 106 HT-29 cells/well were seeded in a 24-well plate (Fisher Scientific,

Canada) and left for twenty-four h at 37°C and 5% CO2. Cells were then stimulated with different

concentrations (v/v) of La-5 (0~12.5%) and LH-2 (0~10%) CFSM adjusted in cell culture medium and

left for twenty-four h at 37°C, in a CO2 (5 %) incubator. The cells were trypsinized using 250 μl of 0.25%

trypsin-EDTA reagent (Life technologies, Burlington, ON, Canada), and agitated pipetting with 750 μl

cell culture medium to make total volume of 1 ml cell suspension. The viable cells were counted by

adding 50 μl of the cell suspension in 450 μl of trypan blue dye (0.4% v/v, Life Technologies,

Burlington, ON, Canada). 50 μl of above cell suspension in trypan blue dye was loaded on

haemocytometer. The exclusion of trypan blue from cells were observed under 10× magnification light

microscopy (AE2000, Opti-Tech Scientific Inc., Scarborough, ON, Canada).

Confocal Scanning Laser Microscopy (CSLM)

The green fluorescent protein (GFP) from the jellyfish Aequorea victori can be used as an endogenous

Page 38: Bioactive Components from Lactobacillus acidophilus and

29

fluorescent tag, which allows visualization of bacterial cells even when present intracellularly in host

cells (Ling, Wang, Xie, Lim, & Leung, 2000). The most outstanding advantage of using CSLM to study

Salmonella invasion is that bacterial internalization on epithelial cells can be observed in three

dimensions without physically disturbing the specimens (Takeuchi & Frank, 2001). The GFP protein

encoded on the plasmid inserted into Salmonella shares a common excitation and emission profile with

the commercially available GFP, but is optimized for an excitation wavelength of 488 nm (Auty et al.,

2005; Burnett, Chen, & Beuchat, 2000). For Leica CSLM, emitted light was collected through a 480 nm

dichroic mirror, a 520 nm long-pass filter, and a 680 nm short-pass filter (Takeuchi & Frank, 2001).

Leica confocal scanning laser microscopy (DMIRB Inverted Fluorescence Microscope, Leica

Microsystem, Concord, ON, Canada) was used to observe the intracellular populations of transformed S.

Typhimurium into the epithelial cell monolayer. In this study, 24 h prior to Salmonella infection, 5 × 106

HT-29 cells were seeded in 35mm glass bottom dishes (MatTek Corporation, Ashland, MA, USA). 2 ml

of a non-toxic concentration of CFSMs (1.5% of La-5 or 1% of LH-2) adjusted with antibiotics free cell

culture medium was added to designated dishes. The GFP labeled S. Typhimurium inoculum containing

5 × 107 CFU/ml (bacteria: cell MOI of 10) was added to dishes and incubated for 2 h at 37°C and 5%

CO2. A negative control was included, comprising HT-29 cells without exposure to bacteria and CFSM.

The positive control consisted of non-stimulated cells (not exposed to CFSM) infected with Salmonella

for 2 h. Test groups consisted of HT-29 cells stimulated with either La-5 or LH-2 CFSM and exposed to

Salmonella with the presence of CFSM for 2 h. After 2 h of infection, extracellular bacteria were

removed by extensive washing with PBS together with 1 ml of 350 μg/ml gentamicin sulfate (Sigma,

Markham, ON, Canada) treatment for 1 h at 37°C, in a CO2 (5 %) incubator. Medium was subsequently

changed to 1 ml of 200 μg/ml gentamicin for an additional 30 min. Following 2 washing steps with PBS,

Page 39: Bioactive Components from Lactobacillus acidophilus and

30

1 ml of antibiotic-free cell culture medium was added in each dish. Then all the dishes were visualized

under CSLM (DMIRB Inverted Fluorescence Microscope, Leica Microsystem, Concord, ON, Canada)

according to manufacturer‟s instruction. Random fields were examined for bacterial invasion.

Trans-epithelial Electrical Resistance (TEER)

Millicell-ERS is designed to facilitate measurements of TEER of cultured epithelial monolayer

integrity directly in tissue culture wells. Millicell ERS-2 (EMD Millipore, Billerica, MA, USA) is a

device which uses alternating current to eliminate adverse effects on the cell membrane. It contains a

silver electrode with a fixed pair of probes, which can measure the voltage deflection at 37°C in tissue

culture medium (Balda et al., 1996). The change in TEER value is an indication of cell monolayer

confluence quantitatively and cell monolayer health qualitatively.

Transwell inserts (BD FalconTM

, Mississauga, ON, Canada) are permeable supports which create an

apical and a basolateral chamber in each well to allow epithelial and other cell types to be grown and

studied in a polarized state.

For TEER measurements, HT-29 cells at a concentration of 1 × 105 cells/ml were pipetted on top of

Transwell inserts, resulting in a seeding density of approximately 5×104 cells/insert. The medium was

changed every two days in both apical (500 μl) and basolateral (700 μl) chambers in each well. After 40

days, cells formed a fully polarized monolayer and reached the plateau TEER value of ~ 120 ohm × cm2.

TEER was measured with a Millicell ERS-2 apparatus (EMD Millipore, Billerica, MA, USA) by

immersing the shorter tip of electrode in the insert and the longer tip in the outer well at a 90° angle to

the plate insert. The shorter tip should not contact cells growing on the insert membrane and the longer

tip should just touch the bottom of the outer well. TEER values were obtained from blank inserts

Page 40: Bioactive Components from Lactobacillus acidophilus and

31

(without cells) and test inserts (with cells) and was calculated to ohm × cm2. The final values were

obtained from triplicate inserts by subtracting average value of blank inserts from all samples and

multiplying by the area of the monolayer (Hasegawa et al., 1999).

Epithelial monolayer Integrity Challenge Study

In this study, we used polarized HT-29 cells which had been grown in Transwell inserts for 40 days with

~120 ohm × cm2

TEER value to investigate whether La-5 or LH-2 CFSM could prevent S.

Typhimurium from interfering with the HT-29 monolayer integrity. In each experiment, the monolayer

in all inserts was from the same passage number and stage of maturation. 24 h before Salmonella

infection, medium in both apical and basolateral chambers created by the Transwell insert in each well

was replaced with antibiotic-free cell culture medium or stimulated with 1.5% of La-5 or 1% of LH-2

CFSM adjusted in antibiotic-free cell culture medium. Subsequently, 1 × 107 CFU/ml S. Typhimurium

DT104 (SA1997-0934) inoculum (Multiplicity of Infection bacteria: cell = 10) was added to the apical

chamber of the inserts and left at 37°C, in a 5% CO2 incubator for 2 h. Gentamicin sulfate (Sigma,

Markham, ON, Canada) was used to eradicate extracellular Salmonella as described above. After

calibrating the Millicell ERS-2 using culture medium, the sterile probes were vertically immersed into

the apical or basolateral chamber at a 90° angle to the plate insert. The first measurement after the

addition of gentamicin was regarded as time t0. TEER readings were recorded at various time intervals

and expressed as the ratio of TEER at time t to the initial value at t0 for each series. This approach has

been utilized in the study of evaluating probiotic activity (Klingberg, Pedersen, Cencic, & Budde,

2005).

Page 41: Bioactive Components from Lactobacillus acidophilus and

32

Lactate Dehydrogenase (LDH) Cytotoxicity Assay

LDH is a stable and soluble enzyme present in the cytoplasm of all mammalian cells. LDH is

impermeable in normal cells, but if the cell‟s plasma membrane is damaged then LDH is rapidly

released into the surrounding medium. In this research, LDH release caused by pathogenic bacteria

damage to the plasma membrane can be quantified as an accurate index of cell death or cytotoxicity.

The LDH levels in cell culture medium were determined using the LDH-Cytotoxicity Assay Kit II

(Abcam, Toronto, ON, Canada) according to the manufacturer‟s instructions. The assay utilizes an

enzymatic coupling reaction: LDH oxidized lactate to generate NADH, which then reacts with

cell-impermeable Tetrazolium Salt WST to generate yellow color (see Figure 2.1). The yellow color

formed can be detected spectrophotometrically at 450 nm and the intensity of the generated color is

proportional to the degree of cell lysis.

Figure 2.1 Catalytic function of the Lactate Dehydrogenase (LDH) enzyme.

Preliminary studies showed that the optimal cell density for LDH quantification was 1 × 105

cells/ml/well in 96-well plate. 24 h before Salmonella infection, 100 μl of HT-29 cells at a concentration

of 1 × 105 cells/ml were seeded into a 96-well plate, resulting in a seeding density of approximately 1 ×

104 cells/well. Simultaneously, 100 μl of non-toxic concentration of La-5 (1.5%) or LH-2 (1%) CFSM

(v/v) were adjusted using antibiotic-free cell culture medium and added in designated wells. Two wells

with cell culture medium only were included as background controls. The background value has to be

Page 42: Bioactive Components from Lactobacillus acidophilus and

33

subtracted from all other values. The negative control consisted of supernatant from cells without

exposure to bacteria or CFSM. The positive control consisted of HT-29 cells infected with S.

Typhimurium DT104 (SA1997-0934; MOI of 10) in the presence of the appropriate CFSM for 2 h. All

supernatants were centrifuged (AllegraTM

21R Centrifuge, Beckman CoulterTM

, Mississauga, ON,

Canada) at 600 × g for 10 min to remove bacterial and eukaryotic cells. A 10 μl aliquot from each well

was dispensed into a new 96-well plate and reacted with 100 μl of WST substrate mix (within the kit).

The absorbance was measured at 450 nm using a Multilabel Counter (Wallac 1420 VictorTM

3V,

Perkin-Elmer Life Sciences, Woodbridge, ON, Canada). LDH cytotoxicity % was then determined as

shown.

Cytotoxicity (%) =(Test sample − Low Control)

(High Control − Low Control)× 100

Low Control: untreated cells

High Control: cells treated with 10 μl Cell Lysis Solution (within the kit)

Values of test groups were expressed as % cytotoxicity relative to the positive controls.

Apoptosis TUNEL Assay

Briefly, polarized HT-29 cells were seeded at 1 × 106

cells/well of a 24-well plate 2 days prior to S.

Typhimurium infection. 24 h later, designated wells were pre-incubated with 1.5% La-5 or 1% LH-2

CFSM in antibiotic-free cell culture medium. 1 × 107 CFU/ml S. Typhimurium DT104 (SA1997-0934;

MOI of 10) was added in designated well with or without the presence of appropriate concentration of

La-5 or LH-2. After 1 h, extracellular bacteria were removed by extensive washing with PBS and

addition of gentamicin incubation as described above. The cells were trypsinized using 250 μl of 0.25%

trypsin-EDTA reagent (Life technologies, Burlington, ON, Canada), and agitated pipetting with 750 μl

Page 43: Bioactive Components from Lactobacillus acidophilus and

34

cell culture medium to make total volume of 1 ml cell suspension. After washing with PBS and

centrifuged at 500g, 5 min (AllegraTM

21R Centrifuge, Beckman CoulterTM

, Mississauga, ON, Canada)

several times, cells were fixed in 1 ml 1% paraformaldehyde (Sigma, Markham, ON, Canada) for 1 h on

ice, followed by PBS washing and suspending in cold 70% (v/v) ethanol and left at 4°C for at least 15 h

for cell membrane permeabilization. The Apo-Direct ™ Kit (BD Biosciences, Mississauga, ON,

Canada) was used in this research according to the manufacturer‟s instruction. Positive and negative

controls in this kit were run in parallel to the samples in order to define M1/M2 markers. 1 ml of BD

positive and negative control cells and all of 15 h frozen cells were both washed with 1 ml of BD wash

buffer (within the kit) and centrifuged at 500 g, 5 min (AllegraTM

21R Centrifuge, Beckman CoulterTM

,

Mississauga, ON, Canada) two times. Subsequently, cells were incubated with 50 μl DNA labeling

solution for 2 h. Then cells were washed with 1 ml of BD Rinse Buffer (within the kit) and centrifuged

at 500g, 5 min (AllegraTM

21R Centrifuge, Beckman CoulterTM

, Mississauga, ON, Canada) two times,

followed by adding 0.3 ml PI/RNase staining buffer. Based on DNA doublet discrimination and

exclusion of cell debris, cells were counted adjusting the sorting threshold. Afterwards, cells were

analyzed by flow cytometry (FACScan, BD, Mississauga, ON, Canada) with excitation at 488 nm.

Fluorescence emission by FITC was measured using a 530/30 band-pass filter while that of PI was

measured using a 585/42 band-pass filter. A total number of 5,000 cells were counted for each sample

and the percentage of FITC positive (apoptotic) cells was determined.

Sulforhodamine B (SRB) Assay

SRB is a bright-pink aminoxanthene dye. Under mild acidic conditions, it electrostatically binds to

basic amino-acid residues, while it dissociates under weakly basic conditions (Skehan et al., 1990). The

Page 44: Bioactive Components from Lactobacillus acidophilus and

35

cell mass is correlated with the amount of dye extracted from cells. The SRB colorimetric assay is an

accurate and reproducible assay based on the sensitive linearity of quantitative staining of cellular

proteins with optical density (OD) between 560 and 580 nm wavelengths. This assay has been widely

used as an efficient and highly cost-effective method for screening drug toxicity on different types of

cell lines (Vichai & Kirtikara, 2006).

The SRB assay was tested on LMH cells in order to determine non-toxic doses of La-5 and LH-2 CFSM.

After seeding 3 × 103 cells/well in a 96-well plate and incubating for 24 h at 37°C in a 5% CO2 incubator,

cells were stimulated with different concentrations of La-5 (0~3%) or LH-2 (0~3%) CFSM, followed

by cells fixation with 50 μl of 50% (w/v) Trichloroacetic Acid (TCA, Sigma, Markham, ON, Canada).

After removing the TCA, cells were stained with 0.4% (w/v) SRB (Sigma, Markham, ON, Canada) in 1%

acetic acid (Glacial Acetic Acid, Fisher Scientific, Canada) for 30 min. Unbound dye was removed with

1% acetic acid, and the protein-bound dye was dissolved when 10 mM Tris-base

(Tris-hydroxymethyl-aminomethane, Sigma, Markham, ON, Canada) was added. The developed color

was measured spectrophotometrically at 570 nm using a microplate reader (Synergy H5, BioTek,

Winooski, VT, USA) and results expressed as percentage with respect to control wells (set as 100%)

grown under regular conditions.

Invasion Assay

The invasion assay was carried out on LMH cells in order to determine the protective ability of La-5 and

LH-2 CFSM to limit the invasion of S. Typhimurium in in vitro chicken model.

Invasion assay was assessed as described elsewhere (Bishop et al., 2008) with specific minor

modifications as follows. LMH cells were seeded at 2 × 106 cells/well in 0.1% gelatin pre-coated

Page 45: Bioactive Components from Lactobacillus acidophilus and

36

12-well plate for 24 h at 37°C in a 5% CO2 incubator. Then 2 ml of a non-toxic concentration 1 % of

La-5 or 1% of LH-2 adjusted with antibiotics free cell culture medium was added to designated dishes

24 h prior to infection. Two chicken isolates of S. Typhimurium (SA2000-0406 or SA2001-4368)

containing 2 × 107 CFU/ml (bacteria: cell MOI of 10) were added to appropriate wells and incubated for

designated time in 37°C and 5% CO2 incubator. Negative controls were included, comprising LMH

cells without exposure to bacteria or CFSM. The positive control consisted of non-stimulated cells (not

exposed to CFSM) infected with Salmonella. Test groups consisted of LMH cells stimulated with either

La-5 or LH-2 CFSM and exposed to Salmonella with the presence of CFSM. At each time point,

extracellular bacteria were removed by extensive washing with PBS, followed by exposure to 350

μg/ml gentamicin sulfate (Sigma, Markham, ON, Canada) for 1 h, and subsequently 200 μg/ml

gentamicin for 30 min. Following 2 times of PBS washing, cells were lysed with 500 μl of 1% Triton

X-100 solution (Sigma, Markham, ON, Canada) in high-pure water (WFI water, Life Technologies,

Burlington, ON, Canada) for 2 min and suspended in 500 μl of PBS (Life Technologies, Burlington, ON,

Canada). Eckmann et al. (1993) stated that within 1 h after cell lysis, Triton would not affect bacterial

viability (Eckmann, Kagnoff, & Fierer, 1993). The number of released viable bacteria was determined

by serial dilution in PBS (Life technologies, Burlington, ON, Canada) and plating on TSA agar (BD

Bioscience, Mississauga, ON, Canada). After overnight incubation at 37°C, the numbers of bacterial

colony were enumerated. CFU calculation was shown in followed formula. Values of test groups were

expressed as % invasion relative to the positive controls.

CFU/ml =Number of Colonies

Dilution Factor × Volume (0.1 ml)

Page 46: Bioactive Components from Lactobacillus acidophilus and

37

Statistical Analysis

Statistical analyses were performed using the Microsoft Excel 2010 analysis tool package. Unless

otherwise stated, all results are the average ± SD of two independent experiments with at least replicates.

Data from each experiment were analyzed using analysis of variance (ANOVA). Differences at P < 0.05

were considered to be statistically significant. Differences at P < 0.01 were considered to be statistically

very significant.

Page 47: Bioactive Components from Lactobacillus acidophilus and

38

CHAPTER 3: Result and Discussion

Trypan Blue Exclusion Assay

In order to use tissue culture to determine the effect of CFSM on the infectivity of Salmonella, a

tolerable dose of CFSM for eukaryotic cells used in these studies needs to be established, since negative

effects (e.g. cell death) may be triggered when applying doses beyond the cells‟ threshold. Based on

previous in vivo studies using mice fed with a partially purified LH-2 CFSM fraction (F5), a high dose

of this fraction (0.08 μg/day) caused intolerance and the mice exhibited a lower survival rate after

Salmonella infection than the non-stimulated mice. However, a low dose of F5 (0.02 μg/day) protected

mice against Salmonella infection. One possible reason for negative effects in mice exposed to the high

dose of F5 might be perturbation of the immune system (Tellez Garay, 2009).

The trypan blue exclusion assay is based on the principle that viable cells have intact cell membranes

which can resist trypan blue dye penetration while dead cells do not. Hence live cells can be easily

differentiated and counted with a light microscope (Strober, 2001). This assay may show a relation

between the CFSM concentration and HT-29 cell proliferation. In the case of La-5, the CFSM showed

no significant effect (P ≥0.05) on cells at all concentrations (see Fig.3.1.A). Interestingly, HT-29 cells

showed highest viability in the presence of 1.6% La-5 CFSM. In the case of LH-2, only 1% showed no

effect (P ≥0.05) on cell viability (see Fig.3.1.B). Thus, 1.5% La-5 (protein concentration 1.5 mg/ml)

or 1% LH-2 (protein concentration 370 μg/ml) were chosen as a non-toxic dose on HT-29 cells and

used in subsequent experiments.

Page 48: Bioactive Components from Lactobacillus acidophilus and

39

Figure 3.1 La-5 and LH-2 toxic dose test on HT-29 cells, estimated by trypan blue exclusive assay. A: La-5

treatment; B: LH-2 treatment. HT-29 cells were seeded and after 24 h the medium was replaced with new cell

culture medium containing different concentrations of La-5 CFSM or LH-2 CFSM and incubated for 24 h. Results

represent the means ± SD of two independent experiments performed in triplicate. Bars identified with ** are

significantly different from untreated cells (P < 0.01). NS means there is no significant difference compared to

untreated cells (P ≥0.05)

0.0E+00

1.0E+05

2.0E+05

3.0E+05

4.0E+05

5.0E+05

6.0E+05

Via

ble

Ce

ll A

mo

ut/

ml

A NS

NS

NS NS NS NS

0.0E+00

1.0E+05

2.0E+05

3.0E+05

4.0E+05

5.0E+05

6.0E+05

7.0E+05

10% LH-2 5% LH-2 2.5% LH-2 1% LH-2 untreatedcells

Via

ble

Ce

lls A

mo

un

t/m

l

B

**

**

**

NS

Page 49: Bioactive Components from Lactobacillus acidophilus and

40

Confocal Scanning Laser Microscopy

In our preliminary studies, the effectiveness of gentamicin treatment in removing extracellular

Salmonella was assessed using confocal scanning laser microscopy (CSLM) and optical density

monitoring; we also evaluated effectiveness of Salmonella internalization through different infection

time using CSLM.

It is important to know whether the gentamicin treatment effectively removes all extracellular

Salmonella since S. Typhimurium, phage-type DT104 used in our experiments were reported to be

resistant to multiple antibiotics. In addition, some strains possess an enhanced virulence phenotype (Wu

et al., 2002). The inhibitory effect of different concentrations of gentamicin (from 50 μg/ml to 350

μg/ml) was tested on the growth of Salmonella Typhimurium DT104 (SA 1997-0934, SA 2000-0406 &

SA 2001-4368) by monitoring optical density (OD) at 600 nm in TSB for up to 40 h. The results showed

that the lowest tested concentration of gentamicin (50 μg/ml) inhibited the growth of all Salmonella

strains (data not shown). It was concluded that 1 h incubation in the presence of 350 μg/ml gentamicin

followed by 30 min incubation with 200 μg/ml gentamicin could effectively remove extracellular

bacteria. To confirm this, confocal scanning laser microscopy (CSLM) was used to visualize the

internalized and externalized Salmonella GFP strain.

By using CSLM it was possible to visualize Salmonella invasion status in three dimensions with

minimum disruption of the specimen. In Figure 3.2, since Salmonella cells are visible, and the cell

morphology is close to what has been reported in the literature (Collins & Kennedy, 1999). The cells

appeared rod-shaped and 2.0- 5.0 µm in size. Furthermore, the use of CSLM allowed the detection of

minimum infection time for S. Typhimurium to invade HT-29 cells. Different Salmonella infection time

Page 50: Bioactive Components from Lactobacillus acidophilus and

41

courses (1 h, 2 h and 3 h) with MOI of 10 were investigated in our preliminary study. All tested infection

times showed internalized Salmonella. Figure 3.3 exhibits confocal images for 2 h infection with or

without La-5 or LH-2 CFSM treatment. In addition, by adjusting the Z-stacks of the scan field and

performing a 3D reconstruction using the Leica confocal software, intracellularly located bacterial cells

were distinguished from those remaining outside of the host cells. Rarely motile Salmonella cells were

observed in the medium. However, a very small portion of bacteria were attached to the external surface

of the host cell membrane. These attached but not internalized bacteria to some extent affect the

accuracy of internalized Salmonella count. However, 1 h incubation with 350 μg/ml gentamicin and 30

min incubation with 200 μg/ml gentamicin removed the majority of extracellular bacteria. Therefore,

this gentamicin treatment was used in the further studies.

Figure 3.2 Confocal image of a single S. Typhimurium GFP cell. Light channel + argon channel (set at GFP

specific parameters 488 nm) merged confocal image of invaded S. Typhimurium GFP in HT-29 cells after 2 h of

infection (MOI 10). Image was taken after remove extracellular Salmonella using gentamicin. A white arrow

indicates one single S. Typhimurium GFP cell. Scale bar: 2 µm.

Page 51: Bioactive Components from Lactobacillus acidophilus and

42

Figure 3.3 Merged confocal images of S. Typhimurium GFP invasion of HT-29 cells. HT-29 cells were seeded and incubated for 24 h. Then the medium was replaced with new

medium only (A) or new medium containing 1.5% of La-5 CFSM (B) or 1% LH-2 CFSM (C) and incubated for 24 h. S.Typhimurium GFP (MOI 10) was added without any

CFSM (A) or with the presence of 1.5% La-5 (B) or with the presence of 1% LH-2 CFSM (C) and incubated for 2 h. Images were taken after using gentamicin to remove

extracellular Salmonella. Scale bar: 20 µm.

Page 52: Bioactive Components from Lactobacillus acidophilus and

43

Trans-epithelial Electrical Resistance (TEER)

The permeable insert used in this study is reported to benefit epithelial cell polarization and

differentiation (Ferruzza, Rossi, Scarino, & Sambuy, 2012). Polarized cells improve physiological

relevance of cell culture studies as they possess tight junctions and microvilli composed brush border,

which are similar to those of human intestinal epithelial cells of the gastrointestinal tract (Chalghoumi

et al., 2009; Cohen et al., 1999; Le Bivic et al., 1988; Rodriguez-Boulan & Nelson, 1989). Therefore,

cell culture using permeable inserts provides a useful model that can mimic specific properties of the

human intestinal epithelium. When cultured in glucose for a certain period of time, HT-29 cells form

very tight junctions and microvillus brush border at the apical surface, indicating cell polarization

(Fitzgerald et al., 1997; Höner zu Bentrup et al., 2006). Taking all these benefits into consideration, the

following experiments were performed using polarized HT-29 cells.

Initial tests were carried out to determine the optimal HT-29 cell density necessary to seed in Transwell

permeable inserts. A 500 μl of four different cell concentrations (1×104 cells/ml, 6×10

4 cells/ml, 1×10

5

cells/ml and 2×105 cells/ml) was seeded in apical chamber of Transwell inserts. Medium in both apical

and basolateral chambers in each well was changed every two days. TEER values for each well were

measured by Millicell ERS-2 apparatus as described in Chapter 2. Consequently, recorded TEER value

was calculated to ohm × cm2. The final values were obtained from triplicate inserts of each cell density

by subtracting average value of blank inserts (without cell) and multiplying by the area of the

monolayer (0.33 cm2) (Hasegawa et al., 1999). As shown in Figure 3.4, it was found that 1×10

5 cells/ml

gave a more linear and stable response in TEER values with less fluctuation. Hence, a 1×105 cells/ml

level was used as optimal cell density for inoculation of Transwell inserts in subsequent experiments.

Page 53: Bioactive Components from Lactobacillus acidophilus and

44

In order to know how long it took for cells to become polarized, the growth of HT-29 cells in the

Transwell permable inserts was determined. Similar with previous TEER measurement, we grew 1×105

cells/ml HT-29 cells and recorded the growth in TEER values. From the Figure 3.5, it can be seen that

before the 12th

day, the TEER value was lower than 20 ohm × cm2. Cells showed steady increase in

TEER values between the 12th day to the 40

th day. After the 40

th day, the TEER value was stable in the

range of 120~140 ohm × cm2. Since the increase in TEER value was an index of cell confluence, tight

junctions formation, and cell polarization establishment (Ghadimi, de Vrese, Heller, & Schrezenmeir,

2010), 120 ohm×cm2 TEER value on day 40 was considered as a mature status of cell polarization. Cells

with TEER value ≥ 120 ohm×cm2 were used in the following experiments.

Page 54: Bioactive Components from Lactobacillus acidophilus and

45

Figure 3.4 Determination of optimal HT-29 cell density in Transwell permeable inserts. A 500 μl of four different cell concentrations (1×104 cells/ml, 6×10

4 cells/ml, 1×10

5

cells/ml and 2×105 cells/ml) was seeded in apical chamber of Transwell inserts. Medium in both apical and basolateral chambers in each well was changed every two days.

TEER values for each well were measured by Millicell ERS-2. Consequently, recorded TEER value was calculated to ohm × cm2. Results represent the means ± SD of two

independent experiments performed in triplicate.

-20

0

20

40

60

80

100

0 5 10 15 20 25 30 35

TEER

(o

hm

× c

m2)

Time (Day)

1×10^4

6×10^4

1×10^5

2×10^5

Page 55: Bioactive Components from Lactobacillus acidophilus and

46

Figure 3.5 Growth of HT-29 cells in Transwell permeable inserts. 1×10

5 cells/ml HT-29 cells was seeded in the apical chambers of the inserts. Medium in both apical and

basolateral chambers in each well was changed every two days. TEER values for each well were measured by Millicell ERS-2. Consequently, recorded TEER value was

calculated to ohm × cm2. Results represent the means ± SD of two independent experiments performed in triplicate.

0

20

40

60

80

100

120

140

160

180

200

0 10 20 30 40 50 60 70

TEER

(o

hm

× c

m2)

Time (day)

Page 56: Bioactive Components from Lactobacillus acidophilus and

47

Polarization of HT-29 cells occurs after a prolonged period of post-confluence culture. Instead of

waiting for 40 days to obtain polarized cells, other researchers have investigated using substances to

induce cell differentiation, including drugs, chemicals or by changing the energy source from glucose to

galactose in standard culture medium (Mitchell & Ball, 2004). Cohen et al. (1999) determined that 20 h

of exposure to the drug forskolin could induce up to 80% cell polarization among 21-day old HT-29

cells. Mocodazole and taxol, functioning as microtubule disrupters, were also discovered to affect brush

border formation in cells grown for 21 days. While another microtubule disrupting agent, colchicine,

could induce apical polarization of most HT-29 cells in only 7 days (Cohen et al., 1999). Fitzgerald et al.

(1997) revealed that HT-29 cells became polarized in 21 days when the extracellular environment was

adjusted to pH 5 (Fitzgerald et al., 1997). In addition, molecular engineering of HT-29 accelerates the

cell polarization period, as well as expands choices in cell line selection. Cloned HT-29 cell lines

exhibiting various cell forms in order to meet different research needs have been produced (Huet,

Sahuquillomerino, Coudrier, & Louvard, 1987; Mitchell & Ball, 2004). After treating cells with sodium

butyrate, the HT-29cl.19A cell line became differentiated and polarized, whereas HT-29cl.16E was

goblet-like and secretory (Mitchell & Ball, 2004). After replacing glucose as an energy source with

galactose, H29-18-C1 became absorptive, while HT29-18-N2 became mucus secreting (Huet et al.,

1987). In conclusion, the prolonged cell culture times used in this study could be replaced by other

methods to create polarized cells, which will shorten the experimental time.

Epithelial Barrier Integrity Study

Many investigators have demonstrated that pathogenic microorganisms have the ability to disrupt

epithelial cell integrity, resulting in abnormal effects on the host, e.g., diarrhea (Canil et al., 1993;

Page 57: Bioactive Components from Lactobacillus acidophilus and

48

Guttman & Finlay, 2008; Solano et al., 2001). A polarized monolayer of epithelial cells form tight

junctions, while S. Typhimurium infection cause a rapid loss in monolayer integrity which can be

detected by TEER (Chalghoumi et al., 2009). TEER loss is correlated with Salmonella invasiveness,

since non-invasive Salmonella does not affect TEER values (Finlay & Falkow, 1990). In addition to

TEER loss, Salmonella can cause apical and basolateral cell depolarization, which is presumable the

reason for tight junction disruption (Finlay et al., 1988; Finlay & Falkow, 1990). When Salmonella

attach to the host cell surface, they use a Type Three Secretion System (T3SS) to inject signals which

assist in their internalization. The host cell membrane becomes ruffled to allow uptake of associated

bacteria (Guttman & Finlay, 2009; Solano et al., 2001).

Probiotics are reported to enhance epithelial barrier integrity and modify the host cell cytoskeleton

(Johnson-Henry, Hagen, Gordonpour, Tompkins, & Sherman, 2007; Resta-Lenert & Barrett, 2003).

Klingberg et al. (2005) concluded that measurement of TEER, where intestinal epithelial cells

monolayer grown in permeable inserts, could be used to evaluate probiotic activity. Hence, in this

epithelial barrier integrity study, the protection afforded by La-5 or LH-2 CFSM on cell integrity

following S. Tyhpimurium infection in polarized HT-29 cells was evaluated.

A preliminary study found that 24 h incubation with 1.5% La-5 CFSM or 1% LH-2 CFSM produced a

small but insignificant increase in TEER value of polarized HT-29 in the inserts (data not shown). In

accordance with previous findings, HT-29 inserts were used when TEER values were above 120 ohm ×

cm2. The initial TEER value in this study was 132 ± 10 ohm × cm

2. The first measurement at time t0

was conducted right after the gentamicin step used to reduce extracellular populations of Salmonella.

Following Salmonella infection, the TEER value at t0 fell broadly (60.4 ± 0.8%) compared to the

initial value. However, cells to which no Salmonella were added also showed a reduction in TEER of

Page 58: Bioactive Components from Lactobacillus acidophilus and

49

43 ± 3%. Figure 3.6 shows the ratio of TEER values obtained at the time of analysis to that obtained

initially for all experimental groups. La-5 infected or LH-2 infected groups were pre-incubated with

1.5% La-5 CFSM or 1% LH-2 CFSM, respectively, and 107 CFU/ml S. Typhimurium (bacteria: cell

MOI of 10) were added. The positive control group comprised HT-29 cells, which were exposed to 107

CFU/ml S. Typhimurium (MOI of 10) only. The negative control group consisted of HT-29 cells to

which neither CFSM, nor Salmonella were added. There was an increase in TEER values for all

groups at t1 and t2 (the first 2 h post-infection). In Fig. 3.6, the negative control group maintained

stable TEER values after time t2. On the contrary, there was a gradual decline in TEER value for all

cells to which Salmonella was added. However, the reduction in TEER value was significantly

alleviated at time t8 (8 h post-infection) when La-5 (P <0.05) or LH-2 (P <0.01) CFSM was presented.

At t22 there was attenuation but insignificantly in TEER loss among La-5 or LH-2 infected groups

than positive control.

Page 59: Bioactive Components from Lactobacillus acidophilus and

50

Figure 3.6 Ratio of TEER of polarized HT-29 cell monolayers after exposure to Salmonella Typhimurium to initial values. HT-29 cells were exposed to 107 CFU/ml S.

Typhimurium (MOI of 10) for 2 h. After 1, 2, 3, 4, 8 and 22 h of recovery in antibiotic-free cell culture medium, the changes in the TEER were measured using a Millicell ERS-2

apparatus. Result was expressed as the ratio of TEER at time t in relation to the time zero t0 (the first TEER measurement after gentamicin step) for each series. Results represent

the means ± SD of two independent experiments performed in triplicate. The dot identified with ** is significantly different (P < 0.01) compared to positive control group, and

of which identified with * is significant difference (P <0.05).

0.6

0.7

0.8

0.9

1

1.1

1.2

1.3

1.4

1.5

1.6

1.7

1.8

0 5 10 15 20 25

Rat

io o

f TE

ER (t/t 0

)

Time (h)

positive ctrl

La-5 infected

LH-2 infected

negative ctrl

**

*

Page 60: Bioactive Components from Lactobacillus acidophilus and

51

The most vulnerable point to pathogen penetration is the paracellular space between cells. The normal

network of tight junction associated proteins is able to seal this pathway for microbial translocation

while allowing the transportation of electrolytes and other nutrients (Moorthy, Murali, & Devaraj,

2009). Probiotics have an advantageous impact on tight junction related proteins by increasing

expression of protein zonula occludens ZO-1 and preventing adverse alteration of protein occluding,

both of which benefit epithelial monolayer integrity (Yu et al., 2012). Klingberg et al. (2005) found a

time and dose dependent enhancement of monolayer integrity for polarized Caco-2 cells when probiotic

L. plantarum MF1298 and L. salivarius DC5 were presented. They concluded that the increase in

TEER value was correlated to the increase in the expression of ZO-1. Furthermore, they demonstrated

that L. monocytogenes-induced epithelial barrier malfunction was attenuated when cells were

pre-incubated with L. plantarum MF129 (Klingberg et al., 2005). Our finding that Lactobacillus CFSM

improved epithelial barrier integrity 8 h post-infection is in contrast with the Klingberg et al. work,

whereby they proposed that only live probiotics could exert the beneficial effects within 8 h since

heat-inactivated L. plantarum cells and L. plantarum cell-free supernatants from culture medium did

not increase TEER values (Klingberg et al., 2005). The reason for this difference could be the virulence

of the pathogen (Solano et al., 2001) or the probiotic strains applied. Yu et al. (2012) found that S.

Typhimurium induced more severe damage to cell integrity than E. coli (Yu et al., 2012). Interestingly,

even though Resta-Lenert & Barrett (2003) used Salmonella in their study, they still concluded that only

live probiotics improved epithelial barrier integrity since antibiotic killed, heat inactivated and spent

medium of Streptococcus thermophilus and Lactobacillus acidophilus failed to enhance TEER values.

This may be due to the amount of spent medium applied since no information was provided on the

concentrations (Resta-Lenert & Barrett, 2003). In another probiotic study, Yu et al. (2012) tested

Page 61: Bioactive Components from Lactobacillus acidophilus and

52

expression of tight junction proteins, including ZO-1, claudin-1, and E-cadherin using a fluorescent dye.

The probiotic strain L. amylophilus D14 did not change the distribution of tight junction proteins, but it

was able to improve the expression of tight junction proteins and moderate their abnormal distribution

triggered by the presence of pathogenic E. coli or S. Typhimurium (Yu et al., 2012).

Probiotics are speculated to modulate Salmonella-induced cytokine-mediated alteration in paracellular

permeability, including interferon gamma (IFN-γ) and tumor necrosis factor alpha (TNF-α) (Capaldo

& Nusrat, 2009). Epithelial cells respond to IFN-γ by restructuring actin, reducing tight junction protein

expression or displacing scaffolding protein ZO-1 (Blum et al., 1997; Youakim & Ahdieh, 1999). Even

though conflicting reports exist, TNF-α has been shown in several studies to directly impair tight

junction function associated with NF-κB pathway interruption (Ma et al., 2004; Ye, Ma, & Ma, 2006).

Furthermore, IFN-γ and TNF-α co-treatment exerts a synergistic effect, which increases barrier

sensitization and malfunction (Fish, Proujansky, & Reenstra, 1999; Wang et al., 2005). The CFSMs

used in this study were expected to perform cytokine adjustment, since Tellez Garay (2009) revealed

a dose dependent effect for IFN-γ and TNF-α modulation by a peptidic fraction of LH-2 CFSM (F5)

in vivo. She found that when the fraction was applied at a rate of 0.02 μg/mouse/day, the IFN-γ level

was significantly lower than in mice not given the fraction, but the fraction also induced TNF-α

production (Tellez Garay, 2009). Hence, the modulation of cytokines when epithelial cells are

stimulated with CFSMs may contribute to the epithelial barrier integrity.

Salmonella entry into the host cell induces membrane ruffling and rearrangement of actin filaments,

resulted in a reduction in TEER values. On the contrary, increase of TEER values were observed at

the first two h after Salmonella infection (see t1 & t2 in Fig. 3.6). Similar TEER increase trend was

also observed by Klingberg et al. (2005) when 107 CFU/cm

2 of Listeria monocytogenes was added in

Page 62: Bioactive Components from Lactobacillus acidophilus and

53

polarized Caco-2 monolayer cells. However, the authors failed to give an explanation for this

phenomenon. The reason for the increase in TEER values at time t1 and t2 could be due to temperature

and homogeneity of culture medium. Matter and Balda (2003) discovered that the TEER value was

dependent on temperature using polarized Madin-Darby Canine Kidney (MDCK) cells. The first

measurement of TEER was carried out at 37°C. After 15 min, a second measurement was performed at

24°C and a final measurement after a further15 min was carried out at 4°C. They found that the higher

temperature the lower TEER value was. In order to get stable and representative values, TEER

measurements require a highly controlled ambient temperature (Matter & Balda, 2003). Figure 3.7

shows the effect of temperature on TEER value. In our experiment, the TEER value at t0 was obtained

after several rinses with PBS at room-temperature (24°C); whereas later TEER values were obtained at

37°C. Different temperature causes TEER value fluctuation. The other factor that may influence TEER

value is whether the cell culture medium is in homeostasis. Stable TEER result would be achieved if

cell culture medium are regulated and homogenized (Matter & Balda, 2003).

In conclusion, La-5 or LH-2 CFSM enhances the epithelial barrier integrity by protecting cells‟ tight

junctions. The measurement of TEER showed that CFSMs started to alleviate the TEER loss caused by

S. Typhimurium late after its infection (8 h post-infection).

Page 63: Bioactive Components from Lactobacillus acidophilus and

54

Figure 3.7 Temperature dependent of TEER on polarized MDCK (Madin-Darby Canine Kidney) cells grown on

permeable inserts for 5 days. Data were obtained from four separate culture with 15 min time interval under

37°C, 24 °C and 4 °C conditions, adapted from Matter & Balda, 2003.

LDH Cytotoxicity Assay

The extracellular stimuli on plasma membrane damage can be quantified by measuring LDH

production. It is an accurate index of cell death or cytotoxicity, as well as a convenient method for the

evaluation of protective effect of CFSM on cell membrane integrity. First of all, we confirmed that 1.5%

La-5 or 1% LH-2 CFSM were not toxic for cells since the LDH production of cell incubated with CFSM

for 24 h was not significant different compared to untreated cells (data not shown). The optimal HT-29

cell seeding concentration was 1 × 105 cells/ml due to its rapid and reproducible detection of LDH.

Salmonella concentrations were evaluated to provide a Multiplicity of Infection (MOI, number of

bacteria: number of cells) of 10, 20, or 50 with 1, 2, or 3 h infection periods. The LDH percentage

cytotoxicity was calculated for each group, as shown in Figure 3.8. 1 h infection time was not adequate

for all MOI (data not shown). However, after 2 h of infection and with MOI of 10 the LDH cytotoxicity %

(22.3 ± 8.7%) was twice that observed for all the other test conditions. After 3 h exposure to

Salmonella (MOI of 50), destruction of HT-29 cells was nearly completed (98.6 ± 3.4%). Taking

previous confocal microscopy results into consideration that 2 h infection period allowed S.

Page 64: Bioactive Components from Lactobacillus acidophilus and

55

Typhimurium to enter cells, the Salmonella Typhimurium infection condition with MOI of 10 for 2 h

was chosen for the LDH study.

Figure 3.8 Cytotoxicity (%) of HT-29 cells produced by S. Typhimurium at different times of exposure and

different MOI. 1 × 105 cells/ml HT-29 cells were seeded in a 96-well plate. LDH production was measured after

adding different MOI of S. Typhimurium and incubated for 2 or 3 h. Results represent the means ± SD of two

independent experiments performed in triplicate.

In addition, the effect of CFSM on Salmonella infection of HT-29 cells was investigated. Pre-exposure

to probiotic strains exerted an inhibitory effect on pathogen activities like adhesion, cytotoxicity and

invasion (Burkholder & Bhunia, 2009; Jankowska, Laubitz, Antushevich, Zabielski, & Grzesiuk, 2008;

Myllyluoma et al., 2008). Therefore, we compared the release of LDH from infected HT-29 cells when

they were pre-incubated with CFSM for 24 h prior to Salmonella infection or co-incubated with CFSM

during 2 h of Salmonella infection. From Figure 3.9, it is shown that co-incubation with CFSM had

almost triple LDH release than with which were co-incubated. The pre-incubation with La-5 CFSM

significantly reduced LDH production caused by Salmonella invasion than positive control. Therefore,

pre-incubation with CFSM for 24 h improved cell monolayer integrity. To strengthen the protective

22.3%

74.7%

12.7%

78.0%

12.5%

98.6%

0%

20%

40%

60%

80%

100%

120%

2 h 3 h

Cyt

oto

xici

ty %

S. Typhimurium infection time (h)

MOI10

MOI20

MOI50

Page 65: Bioactive Components from Lactobacillus acidophilus and

56

effect of CFSM, in addition to pre-incubation, we also co-incubated non-toxic doses of CFSM with

HT-29 cells during exposure to Salmonella. Both pre-incubation and co-incubation with Salmonella

were applied in further studies.

Figure 3.9 Evaluation of CFSMs incubation conditions. 1 × 105 cells/ml polarized HT-29 cells were seeded in a

96-well plate. LDH production was measured after adding S. Typhimurium (SA1997-0934, MOI of 10) and

incubated for 2 h. Positive control represents S. Typhmurium infected cells. Negative control represents untreated

cells. Test groups were either pre-incubated with CFSM or co-incubated with CFSM. All the data were

normalized to positive control (positive control = 100% cytotoxicity). Results represent the means ± SD of two

independent experiments performed in triplicate. Bars identified with ** are very significantly different (P < 0.01)

compared to positive control group, and of which identified with * is significant different (P < 0.05). NS means

no significant difference compared to positive control (P ≥0.05)

As for the preliminary study, 100 μl of 1 × 105 HT-29 cells/ml cells was seeded in a 96-well plate. The

cells were pre-incubated with La-5 or LH-2 CFSM for 24 h and co-incubated with the CFSM when

exposed to Salmonella (MOI 10) for 2 h. Figure 3.10 shows the effect of CFSM on Salmonella infection

of epithelial cells. Compared with the positive control, very significant differences (P <0.01) were

found for both La-5 treated and LH-2 treated groups on normal HT-29 cells and polarized HT-29 cells.

100.0% 75.1%

62.9% 64.4%

192.8% 185.1%

0%

50%

100%

150%

200%

250%

La-5 LH-2 La-5 LH-2

positivecontrol

negativecontrol

pre-incubation co-incubation

Cyt

oto

cxic

ity

%

on polarized HT-29

** **

* NS

Page 66: Bioactive Components from Lactobacillus acidophilus and

57

In the normal HT-29 cell line, cytotoxicity % (as measured by LDH release) in the La-5 treated group

and LH-2 treated group declined by 57 ± 1% and 58 ± 2%, respectively; whereas for the polarized

HT-29 cell line cytotoxicity % in the La-5 treated group and LH-2 treated group was reduced by 46 ± 3%

and 51 ± 2%, respectively. Interestingly, we found on both normal and polarized HT-29 cells, La-5

treated and LH-2 treated groups exhibited very significant reduction (P <0.01) in cytotoxicity

compared to the negative control. Besides protecting cells against S. Typhmurium infection, it is

speculated that CFSM could also benefit epithelial cells under normal growth conditions by reducing

the amount of necrotic cells, which would result in improvement of epithelial membrane integrity.

Figure 3.10 LDH cytotoxicity of epithelial cells induced by Salmonella Typhimurium in the presence or absence

of CFSM treatment. Positive control represents S. Typhmurium infected cells. Negative control represents

untreated cells. La-5 represents La-5 pre-incubation and co-incubation during S. Typhimurium infection. LH-2

represents LH-2 pre-incubation and co-incubation during S. Typhimurium infection. All the data were normalized

to positive control (positive control = 100% cytotoxicity). Results represent the means ± SD of two independent

experiments performed in four replications. Bars identified with ** are very significantly different (P < 0.01)

compared to positive control group.

LDH cytotoxicity evoked by pathogen invasion was alleviated by bioactive components from probiotic

strains. This result was in agreement with previous studies where the cytoprotective effect of

probiotics against a variety of pathogens or physiological stress have been reported. LDH liberation is

100.0% 100.0%

43.2% 53.2%

41.8% 50.9%

75.2% 68.8%

0%

20%

40%

60%

80%

100%

120%

on HT-29 on Polarized HT-29

Rel

ativ

e C

yto

tocx

icit

y %

positive control La-5 LH-2 negative control

** ** ** **

*

*

Page 67: Bioactive Components from Lactobacillus acidophilus and

58

an indicator of cell membrane damage and cell viability. Hence, a reduction in the amount of LDH

released could demonstrate an advantagous impact of probiotics on the host. Burkholder & Bhunia

(2009) found that 1 h pre-incubation with L. rhamnosus GG (LGG) strain significantly attenuated S.

Typhimurium-induced cytotoxicity for both normal and thermally stressed (41°C, 1 h) Caco-2 cells.

Therefore, LGG effectively improved epithelial cell health and mucosal integrity during infection or

exposure to toxins (Burkholder & Bhunia, 2009). H. pylori-evoked cell membrane damage was very

significantly alleviated (P <0.001) by three probiotic strains, namely L. rhamnosus GG, L. rhamnosus

Lc705, P. freudenreichii subsp. shermanii Js (Myllyluoma et al., 2008). In conclusion, La-5 and LH-2

CFSM have potent ability to mitigate pathogen-induced cell damage.

Apoptosis Assay Using Flow Cytometry

To evaluate whether the beneficial effects of CFSM indicated by the LDH assay were reflected in a

reduction of Salmonella-induced apoptosis of HT-29 cells, flow cytometric TUNEL analysis of

apoptotic cells with FITC/PI dual stains was counted out.

The optimum Salmonella exposure time to cause cell apoptosis was determined. HT-29 cells were

pre-incubated with non-toxic doses of CFSM for 24 h, and this was followed by co-incubation in the

presence of CFSM and Salmonella (MOI 10). The selection of Salmonella infection period was based

on the results obtained using the LDH assay, as LDH release indicates the phase of late apoptosis/early

necrosis (Milovic et al., 2001). Infection periods of 1 h, 2 h or 3 h were then investigated for the

apoptosis assay. 1 h infection period caused up to 95% apoptosis on polarized HT-29 cells (data now

shown). Therefore, 1h infection time was chosen for the apoptosis assay.

DNA fragmentation is one of the most distinguishing features of apoptotic cells. When a DNA strand

Page 68: Bioactive Components from Lactobacillus acidophilus and

59

breaks, the FITC stain labels the 3‟-hydroxyl (OH) end of double- and single-stranded DNA catalyzed

by the enzyme, terminal transferase (TdT; a template-independent addition). Apo-Direct™ Kit (BD

Bioscience, Mississauga, ON, Canada) was used to assess DNA strand breakage using FITC/PI dual

stains. Cell viability: vital cells, apoptosis and necrosis were discriminated by different staining. Cells

with the staining patterns FITC(-) and PI(-) were designated as vital cells, FITC(+) and PI(-) as

apoptotic cells, and FITC(+) and PI(+) as late apoptotic or necrotic cells (Punj et al., 2004). Figure 3.11

shows the effect of CFSM on Salmonella Typhimurium induced apoptosis of polarized HT-29 cells.

Cell diameter is reflected by forward-angle light scatter (FSC) and the conformation of inner cellular

structure is reflected by side-angle light scatter (SSC) (Vermes et al., 2000). Comparing light scattered

figures with different exogenous stimuli (A- D in figure 3.11), positive control (B), La-5 treatment

group (C) and LH-2 treatment group (D) resulted in a slight decrease of both FSC and SSC compared to

the negative control (A). Vermes et al. (2000) stated that during the initial phases of apoptosis, FSC

decreased while SSC increased or remained unchanged. After several h both FSC and SSC diminished;

indicating that cells underwent apoptosis (Vermes et al., 2000). Therefore, it was an evident that 1h

Salmonella infection (MOI 10) induced apoptosis of polarized HT-29 cells. To check the effects of

CFSM, FITC signals (Fig 3.11 E-H) and cell viability figures (Fig. 3.11 I-L) were examined.

Figure 3.11 E-H shows the intensity of FITC stain. The M1 and M2 regions in FL1-H reflected negative

FITC stain and positive FITC stain, respectively. The positive control (F) had its FITC signal peak in

the M2 region, indicating the greatest amount of apoptotic cells in this group compared to others. Even

though the peak of fluorescence intensity in graphs of the La-5 treatment group (G) and LH-2 treatment

group (H) was shifted more to the right side (higher FITC fluorescence intensity) than the negative

control (E), the major proportion of FITC signals in these groups were still in the M1 region.

Page 69: Bioactive Components from Lactobacillus acidophilus and

60

A B C D

E F G H

Figure 3.11 Effect of CFSM on Salmonella Typhimurium induced apoptosis of polarized HT-29 cells. A-D showed Forward (FSC-H) and Side (SSC-H) light scattering graphs.

E-H showed the dot plot of FITC signals intensity. The first lane (A & E) represents negative control (untreated cells); the second lane (B & F) represents positive control (S.

Typhimurium infected cells); the third lane (C & G) represents La-5 treatment (La-5 pre-incubation and co-incubation during infection); the fourth lane (D & H) represents

LH-2 treatment (LH-2 pre-incubation and co-incubation during infection). Light scattering signals indicate morphological change of cells. FL1-H displays the number of FITC

positive stain cells. For statistical analysis see Figure 3.12.

Page 70: Bioactive Components from Lactobacillus acidophilus and

61

The data shown in Figure 3.12 were obtained from the cell number ratio of M2/M1, indicating the

percentage of apoptotic cells. This analysis provided a better view of the effect of CFSM on Salmonella

induced apoptosis in polarized HT-29 cells. Compared with the positive control, very significant

differences (P <0.01) were found in cells treated with La-5 and LH-2. Apoptosis of La-5 treated or

LH-2 treated groups compared with positive control were dramatically reduced by 76 ± 2% and 62 ±

7%, respectively. Interestingly, we found La-5 stimulated or LH-2 stimulated groups exhibited a

slight reduction in apoptosis compared to the negative control. These findings further supported the

results obtained using the LDH assay and demonstrate that CFSM enhanced the health of epithelial

cells under normal growing conditions and protected them from Salmonella infection.

To further confirm the anti-apoptotic effect of CFSM, cell viability was determined by comparing

levels of FITC (FL1-H)/PI (FL2-A) dual stain signals (as shown in Figure 3.13).

As shown in Figure 3.13 FITC and PI dual stains differentiated cell viability. Cell population in the

lower left quadrant [FITC(-) and PI(-)] were designated as vital cells; cell populations in the upper left

quadrant [FITC(+) and PI(-)] were designated as apoptotic cells. The upper right quadrant represented

late apoptotic or necrotic cells, which was merely observed in all the tested groups. For the figures, the

positive control (B) consisted of cell population distributed mainly in the upper left quadrant, indicating

the presence of more apoptotic cells than other groups. Even though cells treated with La-5 or LH-2 was

distributed in the upper left quadrant than negative control, it was obvious that they were significantly

lower than the positive control. Cell viability, determined by comparing efficiency of the two stains,

suggested that CFSM treatment had a significant anti-apoptotic effect on Salmonella-induced apoptosis

of polarized HT-29 cells.

Page 71: Bioactive Components from Lactobacillus acidophilus and

62

Figure 3.12 Apoptotic rate of polarized HT-29 cells and the protective effect of CFSM pre-incubation (24 h) and

co-incubation during exposure to Salmonella (1 h) (Flow cytometric TUNEL analysis). La-5 and LH-2 stimulated

cells represented cells only pre-incubated with La-5 and LH-2, respectively. La-5 and LH-2 groups represented

cells pre-incubated with CFSM and co-incubated with Salmonella during 1h infection. Negative control

represented untreated cells. Positive control represented cells infected with Salmonella for 1 h. Results were

obtained after analyzing M2/M1 populations in FL1-H graph (As shown in Fig. 3.10 E-H), displaying in means ±

SD of two independent experiments performed in replicate. Bars identified with ** are very significantly different

(P < 0.01) compared to positive control group.

0.8% 1.0% 1.7% 8.0%

21.3%

83.8%

0%

20%

40%

60%

80%

100%

120%

La-5stimulated

cells

LH-2stimulated

cells

negativecontrol

La-5infected

LH-2infected

positivecontrol

Ap

op

tosi

s R

ate

%

** **

Page 72: Bioactive Components from Lactobacillus acidophilus and

63

A B C D E

Figure 3.13 Cell viability by comparing efficiency of FITC (Y-axis)/PI (X-axis) dual stain signals. A: negative control (untreated cells); B: positive control (S. Typhimurium

infected cells); C: La-5 treatment (La-5 pre-incubation and co-incubation during Salmonella infection); D: LH-2 treatment (LH-2 pre-incubation and co-incubation during

Salmonella infection). E: cartoon detailing elaboration of quadrant gates of figures A-D, adapted from Jahan-Tigh et al., 2012.

Page 73: Bioactive Components from Lactobacillus acidophilus and

64

Many apoptotic studies revealed that pathogen induced apoptosis of epithelial cells was a prolonged

process. Conversely, our experiment found that S. Typhimurium with MOI of 10 caused up to 84 ± 15%

apoptosis of polarized HT-29 cells within 1 h. The differences could be due to using different strains,

different MOI, different treatment before fluorescence staining and, most likely, different cell models.

It is speculated that apoptosis is not easily induced in carcinoma derived cells. Kim et al. (1998) claimed

that it took 12-18 h for bacterial adhesion, invasion and replication, as well as expression of apoptotic

signals to initialize the appearance of apoptosis in HT-29 cells (Kim et al., 1998). Lundberge et al.

(1999) revealed that HeLa, Caco-2 and MDCK cells did not show apoptosis after Salmonella infection

(MOI 25, 30 min), while 80% of macrophages were not viable under the same conditions, elaborating

apoptosis induction from Salmonella infection was more rapid in macrophages than in epithelial cells

(Lundberg, Vinatzer, Berdnik, von Gabain, & Baccarini, 1999). A Salmonella-effector associated with

phosphoinositide phosphatase activity SigD, also named SopB, may account for the late appearance of

apoptosis in epithelial cells compare with macrophages (Knodler et al., 2005). Furthermore, Knodler

and Finlay (2001) compared the time for the occurrence of apoptosis among carcinoma HeLa cells and

a non-transformed rat intestinal epithelial cell line, IEC-6. They found much faster kinetics of

Salmonella induced apoptosis on IEC-6 cells than immortalized carcinoma HeLa cells (Knodler &

Finlay, 2001). This suggests that it is harder to show apoptosis in a carcinoma cell line. In our apoptosis

assay, the cell line we used was a polarized human colon carcinoma HT-29 cell line, which was grown

in Transwell inserts for up to 40 days. The biophysical features of these cells were closer to naïve

human intestine epithelial cells. It is speculated that cell polarization could exert a major role on the rate

of appearance of apoptosis.

Besides the diverse kinetics of using different strains, which may lead to variations in time to induce

Page 74: Bioactive Components from Lactobacillus acidophilus and

65

apoptosis, another factor could be the amount of bacteria that were added. It is reported that the

apoptotic rate of epithelial cells increased with bacterial inoculum size (Kim et al., 1998). Cerquetti et al.

(2008) found more than 80% apoptosis caused by S. Enteritidis with MOI 10 in macrophage RAW

264.7 cells, whereas around 25% apoptosis and less than 10% apoptosis were produced by S. Enteritidis

with MOIs of 1 and 0.1, respectively (Cristina Cerquetti, Hovsepian, Sarnacki, & Goren, 2008).

Therefore, the amount of bacteria added to cells was a crucial factor to determine time to induce

apoptosis. Furthermore, how cells are treated before the fixation step of the assay, together with the

procedures for fluorescence staining may also affect the apoptosis timeline. After 1 h infection of S.

Dublin, Kim et al. (1998) left cells in the presence of gentamicin for a designated time before fixation.

As a result, they found apoptotic cells (cell nuclei stained with Hoechst dye 33258) did not always

appear in the presence of Salmonella (GFP expressing). Some apoptotic cells did not contain

GFP-labeled Salmonella inside. They hypothesized during apoptosis intracellular Salmonella cells were

either killed possibly by entry of gentamicin from culture medium and/or they were released from

apoptotic cells. Moreover, they found prostaglandin H synthase-2, which should inhibit apoptosis in

both bacteria-infected intestinal epithelial cells and non-infected cells, contractedly increased the

numbers of apoptotic cells in both infected and non-infected neighboring epithelial cells after more than

24 h post-infection (Kim et al., 1998). In our study, extracellular Salmonella cells were treated with

gentamicin for 1.5 h in total, followed by an instant fixation step. The exact mechanism of transmission

of apoptotic signals is yet to be elucidated. From the host perspective, an extended gentamicin step

would affect the amount of apoptotic cells to some extent. Instant fixation is crucial to determine the

total amount of cells undergoing Salmonella-induced apoptosis.

In recent years, there have been numerous publications indicating that probiotics suppress cellular

Page 75: Bioactive Components from Lactobacillus acidophilus and

66

apoptosis following infection by a pathogen. Putative mechanisms involved in this suppression effect

have been investigated (as described in Chapter 1). Some of the cytokines, such as Interferon INF-γ

(Gobbato et al., 2008), Tumor Necrosis Factor TNF-α and Interleukin IL-8 (Hausmann, 2010) are

considered to play a central role for triggering apoptosis. Based on previous in vivo and in vitro studies,

La-5 and LH-2 CFSM showed an ability to modulate cytokines. Tellez (2009) revealed a dose

dependent modulation on cytokines such as IFN-γ and TNF-α in mice fed with a peptidic fraction of

LH-2 CFSM (Tellez Garay, 2009). Further study on dendritic cells revealed that levels of the

proinflammatory cytokine TNF-α increased significantly (P <0.0001) in the presence of LH-2 and

La-5 CFSM (1:10 dilution) compared to a negative control (PBS treated). However a 1:100 dilution of

LH-2 and La-5 CFSM, which is similar to the concentration used in the present study, resulted in a

small but insignificant increase in the production of TNF-α by dendritic cells (Elawadli, 2012). All

these results could lead to the conclusion that the modulation of cytokines when epithelial cells are

stimulated with CFSM may contribute to their anti-apoptotic capacity during Salmonella infection.

La-5 or LH-2 CFSMs showed a significant reduction of Salmonella-induced apoptosis on polarized

HT-29 cells line.

SRB Assay

As one of the most consumed meats, much attention has been focused on broiler chickens. Chicken

meat provides valuable nutrition with relative reasonable price. Unfortunately, it also offers the largest

and most vital reservoirs for Salmonella colonization. S. Typhimurium and S. Enteritidis are two main

pathogenic strains responsible for human Salmonellosis in the past few years. These two serovars have

the ability to asymptomatically colonize chickens (Chalghoumi et al., 2009). With the previous

Page 76: Bioactive Components from Lactobacillus acidophilus and

67

beneficial results in human epithelial cells, we created an in vitro chicken model using LMH chicken

hepatoma cells to evaluate the protective efficiency of La-5 and LH-2 CFSM against S. Typhimurium

isolated from chicken.

SRB viability results (see Figure 3.14) show a correlation between the CFSM concentration and LMH

cell proliferation. For La-5 CFSM, cells treated with 1% (v/v) exhibited higher viability than untreated

cells. In terms of LH-2, cells treated with 0.5%, 1%, 1.5% or 3% of LH-2 CFSM showed no statistical

significant difference with untreated cells. Further LDH test confirmed that 1% of La-5 (protein

concentration 1.0 mg/ml) or 1% of LH-2 (370 μg/ml) was not toxic for LMH cell line (data not shown)

and was used in the following experiments.

Page 77: Bioactive Components from Lactobacillus acidophilus and

68

Figure 3.14 La-5 and LH-2 CFSM toxic dose test on LMH cells proliferation, estimated by SRB colorimetric

assay. Cells were plated and after 24 h the medium was replaced with new medium containing different

concentrations of La-5 CFSM (A) or LH-2 CFSM (B) and incubated for 24 h. Results are normalized to untreated

cells (viability of untreated cells = 100%) and display as means ± SD of two independent experiments performed

in triplicate. Bars identified with ** are very significantly different from untreated cells (P < 0.01). NS means

there is no significant difference compared to untreated cells (P ≥0.05).

73.9%

110.9% 100.8% 100.0%

87.3% 100.0%

0%

20%

40%

60%

80%

100%

120%

0.5% La-5 1% La-5 1.5% La-5 2% La-5 3% La-5 untreatedcells

Via

bil

ity

%

A

**

NS NS NS

NS

99.1% 102.4% 102.8%

67.8%

101.9% 100.0%

0%

20%

40%

60%

80%

100%

120%

0.5% LH-2 1% LH-2 1.5% LH-2 2% LH-2 3% LH-2 untreatedcells

Via

bili

ty %

B

**

NS NS NS NS

Page 78: Bioactive Components from Lactobacillus acidophilus and

69

Invasion Assay

Invasion by Salmonella results in bacteria internalization and systematic spread (Chalghoumi et al.,

2009). Bacterial pathogenicity is determined by the organism‟s propensity to colonize and invade the

host cells. Therefore, an assay was conducted to evaluate CFSMs regulation of Salmonella invasion in

the chicken model.

Salmonella infection depends on factors such as bacterial strain, duration of exposure to the pathogen,

and MOI. Initially, same incubation condition as apoptosis assay (MOI of 10, 1 h infection) was

assessed on LMH cell line infected with chicken isolated Salmonella Typhimurium DT104

(SA2001-4368 & SA2000-0406). The results are shown in Figure 3.15. Only LMH cells treated with

LH-2 CFSM showed a significant reduction and this was limited to the SA2000-0406 isolate. Hence, it

may be concluded that Salmonella internalization depends on the bacterial strain. Lin et al. (2008) also

found that levels of inhibition due to L. acidophilus LAP5 differed between strains S. Choleraesuis 2a

and 26a, and this was probably due to differences in their ability to adhere to and invade cells (Lin, Tsai,

Lin, Tsen, & Tsai, 2008).

Afterward, we assessed the regulation of LH-2 CFSM under different infection conditions (shown in

Fig. 3.16), e.g., 2 h infection with MOI of 20 using S. Typhimurium SA 2000-0406, or 1 h infection with

MOIs of 1 or 0.1 using S. Typhimurium SA2001-4368. Under these conditions, the ability of

Salmonella to invade cells was reduced, albeit not significantly, in the presence of LH-2 CFSM. This

led to the conclusion that a reduction of Salmonella internalization in host cells may be one of the

mechanisms enabling CFSM to execute an antagonistic effect against Salmonella infection, but it is not

the most important factor.

Page 79: Bioactive Components from Lactobacillus acidophilus and

70

Figure 3.15 Invasion assay on LMH cell using Salmonella Typhimurium DT104 (SA2001-4368 & SA2000-0406)

isolated from chicken for 1 h infection with MOI of 10. Positive control: infected cells. La-5: cells pre-incubated

with La-5 CFSM and co-incubated Salmonella in the presence of La-5 CFSM. LH-2: cells pre-incubated with

LH-2 CFSM and co-incubated Salmonella in the presence of LH-2 CFSM. Results are normalized to positive

control (positive control = 100%) and display as means ± SD of two independent experiments performed in

triplicate. The bar identified with * is significantly different (P < 0.05) compared to positive control. NS means

there is no significant difference compared to positive control (P ≥0.05).

Figure 3.16 Invasion assay using LMH cells exposed to different infection conditions. Positive control: infected

cells. LH-2: cells pre-incubated with LH-2 CFSM and co-incubated Salmonella with the presence of LH-2 CFSM.

Results are normalized to positive control (positive control = 100%) and display as means ± SD of two

independent experiments performed in triplicate. NS means there is no significant difference compared to

positive control (P ≥0.05).

100.0% 100.0% 98.6% 98.9% 95.2% 94.6%

0%

20%

40%

60%

80%

100%

120%

SA2001-4368 SA2000-0406

Rel

ativ

e In

vasi

on

Rat

e %

positive control La-5 LH-2

* NS NS NS

100.0% 100.0% 100.0% 98.9% 97.9% 95.5%

0%

20%

40%

60%

80%

100%

120%

140%

MOI 20, 2 h MOI 1, 1 h MOI 0.1, 1 h

SA2000-0406 SA2001-4368

Rel

ativ

e In

vasi

on

Rat

e %

positive control LH-2

NS NS NS

Page 80: Bioactive Components from Lactobacillus acidophilus and

71

The ability to prevent pathogen invasion depends on the specific probiotics and tested pathogens,

indicating a high variability. Lin et al. (2008) found both a culture of L. acidophilus LAP5 strain and its

spent culture supernatant had a strong antagonistic effect on S. Choleraesuis infection of Caco-2 cells,

of which live probiotic strain showed a stronger antagonistic effect than its spent culture supernatant

(Lin et al., 2008). Ingrassia et al. (2005) determined that E. coli invasion of differentiated and

undifferentiated intestinal epithelial cells was inhibited by pre-incubation (75% and 84%) or

co-incubation (43% and 62%) with L. casei. A better inhibitory effect on E. coli invasion was observed

when L. casei culture supernatant was added to the incubation medium together with its live cells, and

this effect was thought to be associated with the probiotic adhesion improvement to the cells when the

supernatant was present (Ingrassia, Leplingard, & Darfeuille-Michaud, 2005). Interestingly, mucus

pre-treated with probiotic strains such as Lactobacillus (Gueimonde, Jalonen, He, Hiramatsu, &

Salminen, 2006; Tuomola, Ouwehand, & Salminen, 1999) or Bifidobacterium (Collado, Gueimonde,

Hernandez, Sanz, & Salminen, 2005) have been reported to increase the adhesion of pathogenic E. coli,

L. monocytogenes and S. Typhimurium. The biological significance of these increases is unknown, but

Collado et al. (2007) thought, instead of decrease the pathogens‟ adhesive ability, the protective

function of these probiotic strains against pathogens might compete for the specific adhesion site(s) and

receptors or other factors (Collado, Meriluoto, & Salminen, 2007).

Conclusions

In summary, results demonstrate that bioactive components from L. acidophilus (La-5) and L.

helveticus (LH-2) fermented milk enhanced the epithelial membrane integrity and exerted antagonized

effect against Salmonella Typhimurium infection. First of all, non-toxic doses of CFSMs were tested.

Page 81: Bioactive Components from Lactobacillus acidophilus and

72

Trypan blue exclusion and LDH results indicated that 1.5% La-5 or 1% LH-2 CFSM (v/v) was safe on

HT-29 cell line; whereas SRB and LDH results showed that 1% La-5 or 1% LH-2 CFSM (v/v) was safe

on LMH cell line. Afterward, when HT-29 cells were pre-incubated with non-toxic dose of CFSM and

co-incubated during Salmonella infection with the presence of CFSM, Salmonella triggered TEER loss

[8 h post-infection: La-5 (P < 0.05); LH-2 (P < 0.01)], LDH production [La-5 or LH-2 (P < 0.01)] and

apoptosis [La-5 or LH-2 (P < 0.01)] were significantly attenuated. Since Salmonella often colonizes

and contaminates chickens asymptomatically, chicken hepatoma LMH cell line together with

Salmonella isolates from chicken was utilized as in vitro poultry model. Then the evaluation of CFSMs

on Salmonella invasion was carried out under various infection conditions. Results showed CFSMs

disruption of Salmonella invasion was highly strain dependent. Therefore, a reduction of Salmonella

internalization in host cells may be served as one of the mechanisms, but not the most important factor,

endorsing CFSM to perform an antagonistic effect against Salmonella infection.

Page 82: Bioactive Components from Lactobacillus acidophilus and

73

CHAPTER 4

General Conclusions

The main purpose of this study is to determine the mechanism whereby bioactive components from L.

acidophilus or L. helveticus fermented milk (La-5 or LH-2) protect epithelial cells from Salmonella

Typhimurium infection. Based on previous in vitro and in vivo studies related to milk fermented by

probiotic bacteria performed in our laboratory, it was found that the bioactive components derived

from the probiotic bacteria fermentation stimulate the immune system and exert an antagonistic effect

against various enteric infections (Chin, 2002; Elawadli, 2012; M. J. Medellin-Peña, 2007; Ng, 2000;

Tellez Garay, 2009). It was further hypothesized that the bioactive components from milk fermented

with two probiotic bacterial strains (L. acidophilus La-5 and L. helveticus LH-2) could inhibit

Salmonella infectivity through modulation of cell membrane integrity and inhibition of

Salmonella-induced apoptosis in epithelial cells. To test this hypothesis, the non-toxic dose of La-5 and

LH-2 cell free spent medium (CFSM) was determined for application to epithelial cells using the

Trypan Blue Exclusion assay, LDH and SRB colorimetric assays. The integrity of the epithelial

monolayer was assessed by measuring trans-epithelial electrical resistance (TEER), as well as release

of intracellular LDH by a colorimetric assay. Afterward, the inhibitory effect of CFSM on Salmonella

induced apoptosis in epithelial cells using the TUNEL assay was determined. Finally, the evaluation of

CFSMs on Salmonella invasion was carried out under various infection conditions.

The research was divided in three sections: (1) the preparation of CFSM, (2) an in vitro study using

human colon carcinoma HT-29 cells, as a human model, and (3) an in vitro study using chicken

hepatoma LMH cells, as a poultry model. L. acidophilus La-5 or L. helveticus LH-2 cell-free spent

Page 83: Bioactive Components from Lactobacillus acidophilus and

74

medium (CFSM) were prepared from a 48-h fermentation either with whey protein-based medium for

La-5 or skim milk for LH-2. The CFSMs were filtered, lyophilized and concentrated (10×). Human

colon carcinoma HT-29 cells and chicken hepatoma LMH cells were used as the in vitro human

intestinal model and the chicken model, respectively. In the human model, HT-29 cells were grown in

Transwell permeable inserts for 40 days to allow the cells to become polarized, as polarized cells are

more physiologically and biochemically related to native human intestinal cells. Pre-incubation and

co-incubation with CFSM during Salmonella infection sensibilized and enhanced cell membrane

integrity. In TEER measurement, the presence of La-5 (P <0.05) or LH-2 (P <0.01) significantly

attenuated the S. Typhimurium-induced disruption of the epithelial barrier function at 8 h post-infection.

In addition, release of intracellular LDH, as an index of cell membrane damage induced by Salmonella,

was also attenuated (P <0.01) in the presence of both La-5 or LH-2. To further elaborate the mechanism

of the protective action on cells, apoptosis induced by Salmonella was investigated. When cells were

exposed to CFSM from La-5 or LH-2, significant (P <0.01) anti-apoptotic effects were observed.

Interestingly, without Salmonella infection, cell membrane integrity was also improved when cells

were exposed to La-5 or LH-2 CFSM.

The effects were less pronounced when chicken hepatoma LMH cells were used. Exposure of the LMH

cells to CFSMs of La-5 or LH-2 resulted in less than a log10 cycle reduction in the number of

internalized Salmonella cells. The impact varied depending on the Salmonella strain tested. Further

work using the chicken cell line needs to be done in order to elucidate whether CFSMs exert similar

protective mechanisms against Salmonella and whether this offers a promising approach to reduce

Salmonella carriage in poultry.

Overall, it can be concluded that the inhibitory function of CFSMs on Salmonella internalization may

Page 84: Bioactive Components from Lactobacillus acidophilus and

75

not play a pivotal role in preventing salmonellosis, but the enhancement of cell membrane integrity and

anti-apoptotic effect on Salmonella-infected cells suggest that bioactive molecules produced by

probiotics hold promise as therapeutic and prophylactic agents to combat enteric infections.

Future Research

Encouraged by such in vitro results, future work could focus on the anti-microbial effects of CFSM

from various LAB strains and how medium composition affects the activity of the bioactives.

Moreover, the synergistic effects of diverse bioactive components from different LAB should be

investigated to determine if their combinatorial effects offer better protection to the intestine.

Furthermore, the stability of the bioactive components present in the CFSMs needs to be investigated.

Improvement could be achieved by using protective agents and other formulations e.g., encapsulation

material (Jankovic, Sybesma, Phothirath, Ananta, & Mercenier, 2010). Probiotic bacterial genes could

be modified to improve proteolytic activity during fermentation in order to acquire a greater array of

bioactive components. Along with the previous in vivo studies that revealed CFSMs had an

antagonistic effect against various enteric pathogens, in vivo anti-apoptotic activity of La-5 or LH-2

against S. Typhimurium infection as well as their active doses in laboratory animals or in clinical

trials need to be assessed. These findings will contribute to providing solid evidence to related

government departments, such as Health Canada, for the approval of using these CFSMs as

therapeutic supplements in functional foods.

Page 85: Bioactive Components from Lactobacillus acidophilus and

76

REFERENCE

Anderson, R. C., Cookson, A. L., McNabb, W. C., Park, Z., McCann, M. J., Kelly, W. J., & Roy, N.

C. (2010). Lactobacillus plantarum MB452 enhances the function of the intestinal barrier by

increasing the expression levels of genes involved in tight junction formation. Bmc Microbiology,

10, 316.

Apella, M. C., Gonzalez, S. N., Demacias, M. E. N., Romero, N., & Oliver, G. (1992). Invitro studies

on the inhibition of the growth of shigella-sonnei by lactobacillus-casei and lact-acidophilus.

Journal of Applied Bacteriology, 73(6), 480-483.

Ashida, H., Mimuro, H., Ogawa, M., Kobayashi, T., Sanada, T., Kim, M., & Sasakawa, C. (2011).

Host-pathogen interactions cell death and infection: A double-edged sword for host and

pathogen survival. Journal of Cell Biology, 195(6), 931-942.

Auty, M., Duffy, G., O'Beirne, D., McGovern, A., Gleeson, E., & Jordan, K. (2005). In situ

localization of escherichia coli O157 : H7 in food by confocal scanning laser microscopy.

Journal of Food Protection, 68(3), 482-486.

Backert, S., Boehm, M., Wessler, S., & Tegtmeyer, N. (2013). Transmigration route of campylobacter

jejuni across polarized intestinal epithelial cells: Paracellular, transcellular or both? Cell

Communication and Signaling, 11, 72.

Balda, M. S., Whitney, J. A., Flores, C., Gonzalez, S., Cereijido, M., & Matter, K. (1996). Functional

dissociation of paracellular permeability and transepithelial electrical resistance and disruption of

the apical-basolateral intramembrane diffusion barrier by expression of a mutant tight junction

membrane protein. Journal of Cell Biology, 134(4), 1031-1049.

Bayoumi, M. A., & Griffiths, M. W. (2012). In vitro inhibition of expression of virulence genes

responsible for colonization and systemic spread of enteric pathogens using bifidobacterium

bifidum secreted molecules. International Journal of Food Microbiology, 156(3), 255-263.

Bayoumi, M. A., & Griffiths, M. W. (2010). Probiotics down-regulate genes in salmonella enterica

serovar typhimurium pathogenicity islands 1 and 2. Journal of Food Protection, 73(3), 452-460.

Benderska, N., Chakilam, S., Hugle, M., Ivanovska, J., Gandesiri, M., Schulze-Luehrmann, J., . . .

Schneider-Stock, R. (2012). Apoptosis signalling activated by TNF in the lower gastrointestinal

tract-review. Current Pharmaceutical Biotechnology, 13(11), 2248-2258.

Benkerroum, N. (2010). Antimicrobial peptides generated from milk proteins: A survey and prospects

for application in the food industry. A review. International Journal of Dairy Technology, 63(3),

320-338.

Page 86: Bioactive Components from Lactobacillus acidophilus and

77

Bishop, A., House, D., Perkins, T., Baker, S., Kingsley, R. A., & Dougan, G. (2008). Interaction of

salmonella enterica serovar typhi with cultured epithelial cells: Roles of surface structures in

adhesion and invasion. Microbiology-(UK), 154(7), 1914-1926.

Blum, M. S., Toninelli, E., Anderson, J. M., Balda, M. S., Zhou, J. Y., O'Donnell, L., . . . Bender, J. R.

(1997). Cytoskeletal rearrangement mediates human microvascular endothelial tight junction

modulation by cytokines. American Journal of Physiology-Heart and Circulatory Physiology,

273(1), H286-H294.

Brovko, L. Y., Vandenende, C., Chu, B., Ng, K. Y., Brooks, A., & Griffiths, M. W. (2003). In vivo

assessment of effect of fermented milk diet on course of infection in mice with bioluminescent

salmonella. Journal of Food Protection, 66(11), 2160-2163.

Brune, B. (2003). Nitric oxide: NO apoptosis or turning it ON? Cell Death and Differentiation, 10(8),

864-869.

Burkholder, K. M., & Bhunia, A. K. (2009). Salmonella enterica serovar typhimurium adhesion and

cytotoxicity during epithelial cell stress is reduced by lactobacillus rhamnosus GG. Gut

Pathogens, 1, 14.

Burnett, S. L., Chen, J., & Beuchat, L. R. (2000). Attachment of escherichia coli O157: H7 to the

surfaces and internal structures of apples as detected by confocal scanning laser microscopy.

Applied and Environmental Microbiology, 66(11), 4679-4687.

Canil, C., Rosenshine, I., Ruschkowski, S., Donnenberg, M. S., Kaper, J. B., & Finlay, B. B. (1993).

Enteropathogenic escherichia-coli decreases the transepithelial electrical-resistance of polarized

epithelial monolayers. Infection and Immunity, 61(7), 2755-2762.

Capaldo, C. T., & Nusrat, A. (2009). Cytokine regulation of tight junctions. Biochimica Et Biophysica

Acta-Biomembranes, 1788(4), 864-871.

Caron, J., Lariviere, L., Nacache, M., Tam, M., Stevenson, M. M., McKerly, C., . . . Malo, D. (2006).

Influence of Slc11a1 on the outcome of salmonella enterica serovar enteritidis infection in mice

is associated with th polarization. Infection and Immunity, 74(5), 2787-2802.

CDC. (2011). Vital signs: Incidence and trends of infection with pathogens transmitted commonly

through food --- foodborne diseases active surveillance network, 10 U.S. sites, 1996—2010.

Atlanta, Georgia: U.S. Department of Health and Human Services, CDC.

CDC. (2013). Antibiotic resistance threats in the united states, 2013. Atlanta, Georgia: U.S.

Department of Health and Human Services, CDC.

Chalghoumi, R., Thewis, A., Beckers, Y., Marcq, C., Portetelle, D., & Schneider, Y. J. (2009).

Adhesion and growth inhibitory effect of chicken egg yolk antibody (IgY) on salmonella

Page 87: Bioactive Components from Lactobacillus acidophilus and

78

enterica serovars enteritidis and typhimurium in vitro. Foodborne Pathogens and Disease, 6(5),

593-604.

Chin, N. M. L. (2002). In Griffiths M. W. (Ed.), Isolation and characterization of the

immunomodulating properties of milk peptides fermented by LB. helveticus (M.Sc. Thesis ed.).

Guelph, Ont.: University of Guelph.

Cohen, E., Ophir, I., & Ben Shaul, Y. (1999). Induced differentiation in HT29, a human colon

adenocarcinoma cell line. Journal of Cell Science, 112(16), 2657-2666.

Collado, M. C., Gueimonde, M., Hernandez, M., Sanz, Y., & Salminen, S. (2005). Adhesion of

selected bifidobacterium strains to human intestinal mucus and the role of adhesion in

enteropathogen exclusion. Journal of Food Protection, 68(12), 2672-2678.

Collado, M. C., Meriluoto, J., & Salminen, S. (2007). Role of commercial probiotic strains against

human pathogen adhesion to intestinal mucus. Letters in Applied Microbiology, 45(4), 454-460.

Collins, C. H., & Kennedy, D. A. (1999). Laboratory-acquired infections : History, incidence, causes,

and prevention (4th ed. ed.). Oxford ; Boston: Butterworth-Heinemann.

Compton, M. M. (1992). A biochemical hallmark of apoptosis - internucleosomal degradation of the

genome. Cancer and Metastasis Reviews, 11(2), 105-119.

Corr, S. C., Li, Y., Riedel, C. U., O'Toole, P., Hill, C., & Gahan, C. (2007). Bacteriocin production as

a mechanism for the antfinfective activity of lactobacillus salivarius UCC118. Proceedings of

the National Academy of Sciences of the United Statesof America, 104(18), 7617-7621.

Corver, W. E., Cornelisse, C. J., & Fleuren, G. J. (1994). Simultaneous measurement of 2 cellular

antigens and dna using fluorescein-isothiocyanate, R-phycoerythrin, and propidium iodide on a

standard facscan. Cytometry, 15(2), 117-128.

Cristina Cerquetti, M., Hovsepian, E., Sarnacki, S. H., & Goren, N. B. (2008). Salmonella enterica

serovar enteritidis dam mutant induces low NOS-2 and COX-2 expression in macrophages via

attenuation of MAPK and NF-kappa B pathways. Microbes and Infection, 10(14-15),

1431-1439.

de LeBlanc, A. M., Castillo, N. A., & Perdigon, G. (2010). Anti-infective mechanisms induced by a

probiotic lactobacillus strain against salmonella enterica serovar typhimurium infection.

International Journal of Food Microbiology, 138(3), 223-231.

De Moreno De Leblanc, A., Maldonado Galdeano, C., Dogi, C. A., Carmuega, E., Weill, R., &

Perdigon, G. (2010). Adjuvant effect of a probiotic fermented milk in the protection against

salmonella enteritidis serovar typhimurium infection: Mechanisms involved. International

Journal of Immunopathology and Pharmacology, 23(4), 1235-44.

Page 88: Bioactive Components from Lactobacillus acidophilus and

79

Demacias, M. E. N., Romero, N. C., Apella, M. C., Gonzalez, S. N., & Oliver, G. (1993). Prevention

of infections produced by escherichia-coli and listeria-monocytogenes by feeding milk

fermented with lactobacilli. Journal of Food Protection, 56(5), 401-405.

Ding, W., Wang, H., & Griffiths, M. W. (2005). Probiotics down-regulate flaA sigma28 promoter in

campylobacter jejuni. Journal of Food Protection®, 68(11), 2295-2300.

Divya, J. B., Varsha, K. K., Nampoothiri, K. M., Ismail, B., & Pandey, A. (2012). Probiotic fermented

foods for health benefits. Engineering in Life Sciences, 12(4), 377-390.

Dong, H. P., Kleinberg, L., Davidson, B., & Risberg, B. (2008). Methods for simultaneous

measurement of apoptosis and cell surface phenotype of epithelial cells in effusions by flow

cytometry. Nature Protocols, 3(6), 955-964.

Ebringer, L., Ferencik, M., & Krajcovic, J. (2008). Beneficial health effects of milk and fermented

dairy products--review. Folia Microbiologica, 53(5), 378-394.

Eckmann, L., Kagnoff, M. F., & Fierer, J. (1993). Epithelial cells secrete the chemokine interleukin-8

in response to bacterial entry. Infection and Immunity, 61(11), 4569-4574.

Eckmann, L., & Kagnoff, M. F. (2001). Cytokines in host defense against salmonella. Microbes and

Infection, 3(14-15), 1191-1200.

Elawadli, I. (2012). In Sharif S. (Ed.), Investigations into the effects of lactobacilli on murine

dendritic cells (M.Sc. Thesis ed.). Guelph, Ont: University of Guelph.

Elmore, S. (2007). Apoptosis: A review of programmed cell death. Toxicologic Pathology, 35(4),

495-516.

FAO, & WHO. (2001). Health and nutritional properties of probiotics in food including powder milk

with live lactic acid bacteria. Report of a joint FAO/ WHO expert consultation on evaluation of

health and nutritional properties of probiotics in food including powder milk with live lactic acid

bacteria. Retrieved March/30, 2014, from

http://www.who.int/foodsafety/publications/fs_management/en/probiotics.pdf

Ferrell, N., Desai, R. R., Fleischman, A. J., Roy, S., Humes, H. D., & Fissell, W. H. (2010). A

microfluidic bioreactor with integrated transepithelial electrical resistance ( TEER) measurement

electrodes for evaluation of renal epithelial cells. Biotechnology and Bioengineering, 107(4),

707-716.

Ferruzza, S., Rossi, C., Scarino, M. L., & Sambuy, Y. (2012). A protocol for differentiation of human

intestinal caco-2 cells in asymmetric serum-containing medium. Toxicology in Vitro, 26(8),

1252-1255.

Fink, S. L., & Cookson, B. T. (2005). Apoptosis, pyroptosis, and necrosis: Mechanistic description of

dead and dying eukaryotic cells. Infection and Immunity, 73(4), 1907-1916.

Page 89: Bioactive Components from Lactobacillus acidophilus and

80

Finlay, B. B., & Falkow, S. (1990). Salmonella interactions with polarized human intestinal caco-2

epithelial-cells. Journal of Infectious Diseases, 162(5), 1096-1106.

Finlay, B. B., Starnbach, M. N., Francis, C. L., Stocker, B. A. D., Chatfield, S., Dougan, G., &

Falkow, S. (1988). Identification and characterization of tnphoa mutants of salmonella that are

unable to pass through a polarized mdck epithelial-cell monolayer. Molecular Microbiology,

2(6), 757-766.

Fish, S. M., Proujansky, R., & Reenstra, W. W. (1999). Synergistic effects of interferon gamma and

tumour necrosis factor alpha on T84 cell function. Gut, 45(2), 191-198.

Fitzgerald, R. C., Omary, M. B., & Triadafilopoulos, G. (1997). Acid modulation of HT29 cell growth

and differentiation - an in vitro model for barrett's esophagus. Journal of Cell Science, 110,

663-671.

Gaggìa, F., Mattarelli, P., & Biavati, B. (2010). Probiotics and prebiotics in animal feeding for safe

food production. International Journal of Food Microbiology, 141, 515-528.

Ghadimi, D., de Vrese, M., Heller, K. J., & Schrezenmeir, J. (2010). Effect of natural

commensal-origin DNA on toll-like receptor 9 (TLR9) signaling cascade, chemokine IL-8

expression, and barrier integritiy of polarized intestinal epithelial cells. Inflammatory Bowel

Diseases, 16(3), 410-427.

Gobbato, N., Galdeano, C. M., & Perdigon, G. (2008). Study of some of the mechanisms involved in

the prevention against salmonella enteritidis serovar typhimurium infection by lactic acid

bacteria. Food and Agricultural Immunology, 19(1), 11-23.

Goto, Y., & Ivaylo, I. (2013). Intestinal epithelial cells as mediators of the commensal–host immune

crosstalk. Immunology and Cell Biology, 91(3), 204-214.

Grajek, W., & Olejnik, A. (2004). Epithelial cell cultures in vitro as a model to study functional

properties of food. Polish Journal of Food and Nutrition Sciences, 13(SI 1), 5-24.

Griffiths, M. W., & Tellez, A. M. (2013). Lactobacillus helveticus: The proteolytic system. Frontiers

in Microbiology, 4, 30.

Grigoroff, S. (1905). E´tude sur un lait fermente comestible. le „„Kisselo-mleko‟‟ de bulgarie. Revue

Medical De La Suisse Romande, 25, 714-720.

Gueimonde, M., Jalonen, L., He, F., Hiramatsu, M., & Salminen, S. (2006). Adhesion and

competitive inhibition and displacement of human enteropathogens by selected lactobacilli.

Food Research International, 39(4), 467-471.

Guttman, J. A., & Finlay, B. B. (2008). Subcellular alterations that lead to diarrhea during bacterial

pathogenesis. Trends in Microbiology, 16(11), 535-542.

Page 90: Bioactive Components from Lactobacillus acidophilus and

81

Guttman, J. A., & Finlay, B. B. (2009). Tight junctions as targets of infectious agents. Biochimica Et

Biophysica Acta-Biomembranes, 1788(4), 832-841.

Hans-Jurgen, R. (Ed.). (2008). Apoptosis, cytotoxicity and cell proliferation<br /> (Fourth Edition

ed.). Germany: Roche Diagnostics.

Hartmann, R., & Meisel, H. (2007). Food-derived peptides with biological activity: From research to

food applications. Current Opinion in Biotechnology, 18(2), 163-169.

Hasegawa, H., Fujita, H., Katoh, H., Aoki, J., Nakamura, K., Ichikawa, A., & Negishi, M. (1999).

Opposite regulation of transepithelial electrical resistance and paracellular permeability by rho in

madin-darby canine kidney cells. Journal of Biological Chemistry, 274(30), 20982-20988.

Haug, A., Hostmark, A. T., & Harstad, O. M. (2007). Bovine milk in human nutrition - a review.

Lipids in Health and Disease, 6, 25.

Hausmann, M. (2010). How bacteria-induced apoptosis of intestinal epithelial cells contributes to

mucosal inflammation. International Journal of Inflammation, 2010, 574568-574568.

Höner zu Bentrup, K., Ramamurthy, R., Ott, C. M., Emami, K., Nelman-Gonzalez, M., Wilson, J.

W., . . . Nickerson, C. A. (2006). Three-dimensional organotypic models of human colonic

epithelium to study the early stages of enteric salmonellosis. Microbes and Infection, 8(7),

1813-1825.

Hubatsch, I., Ragnarsson, E., & Artursson, P. (2007). Determination of drug permeability and

prediction of drug absorption in caco-2 monolayers. NATURE PROTOCOLS, 2(9), 2111-2119.

Huet, C., Sahuquillomerino, C., Coudrier, E., & Louvard, D. (1987). Absorptive and mucus-secreting

subclones isolated from a multipotent intestinal-cell line (ht-29) provide new models for cell

polarity and terminal differentiation. Journal of Cell Biology, 105(1), 345-357.

Ingrassia, I., Leplingard, A., & Darfeuille-Michaud, A. (2005). Lactobacillus casei DN-114 001

inhibits the ability of adherent-invasive escherichia coli isolated from crohn's disease patients to

adhere to and to invade intestinal epithelial cells. Applied and Environmental Microbiology,

71(6), 2880-2887.

Jahan-Tigh, R. R., Ryan, C., Obermoser, G., & Schwarzenberger, K. (2012). Flow cytometry. Journal

of Investigative Dermatology, 132(10), 1-6.

Jankovic, I., Sybesma, W., Phothirath, P., Ananta, E., & Mercenier, A. (2010). Application of

probiotics in food products-challenges and new approaches. Current Opinion in Biotechnology,

21(2), 175-181.

Jankowska, A., Laubitz, D., Antushevich, H., Zabielski, R., & Grzesiuk, E. (2008). Competition of

lactobacillus paracasei with salmonella enterica for adhesion to caco-2 cells. Journal of

Biomedicine and Biotechnology, , 357964.

Page 91: Bioactive Components from Lactobacillus acidophilus and

82

Johnson-Henry, K. C., Hagen, K. E., Gordonpour, M., Tompkins, T. A., & Sherman, P. M. (2007).

Surface-layer protein extracts from lactobacillus helveticus inhibit enterohaemorrhagic

escherichia coli O157 : H7 adhesion to epithelial cells. Cellular Microbiology, 9(2), 356-367.

Kagaya, K., Watanabe, K., & Fukazawa, Y. (1989). Capacity of recombinant gamma interferon to

activate macrophages for salmonella-killing activity. Infection and Immunity, 57(2), 609-615.

Kerr, J. F. R., Wyllie, A. H., & Currie, A. R. (1972). Apoptosis - basic biological phenomenon with

wide-ranging implications in tissue kinetics. British Journal of Cancer, 26(4), 239-257.

Khani, S., Hosseini, H. M., Taheri, M., Nourani, M. R., & Fooladi, A. A. I. (2012). Probiotics as an

alternative strategy for prevention and treatment of human diseases: A review. Inflammation &

Allergy Drug Targets, 11(2), 79-89.

Kim, J. M., Eckmann, L., Savidge, T. C., Lowe, D. C., Witthoft, T., & Kagnoff, M. F. (1998).

Apoptosis of human intestinal epithelial cells after bacterial invasion. Journal of Clinical

Investigation, 102(10), 1815-1823.

Kitts, D. D., & Weiler, K. (2003). Bioactive proteins and peptides from food sources. applications of

bioprocesses used in isolation and recovery. Current Pharmaceutical Design, 9(16), 1309-1323.

Klingberg, T. D., Pedersen, M. H., Cencic, A., & Budde, B. B. (2005). Application of measurements

of transepithelial electrical resistance of intestinal epithelial cell monolayers to evaluate probiotic

activity. Applied and Environmental Microbiology, 71(11), 7528-7530.

Knodler, L. A., & Finlay, B. B. (2001). Salmonella and apoptosis: To live or let die? Microbes and

Infection, 3(14-15), 1321-1326.

Knodler, L. A., Finlay, B. B., & Steele-Mortimer, O. (2005). The salmonella effector protein SopB

protects epithelial cells from apoptosis by sustained activation of akt. Journal of Biological

Chemistry, 280(10), 9058-9064.

Korhonen, H. (2009). Milk-derived bioactive peptides: From science to applications. Journal of

Functional Foods, 1(2), 177-187.

Lavermicocca, P., Valerio, F., Lonigro, S. L., Di Leo, A., & Visconti, A. (2008). Antagonistic activity

of potential probiotic lactobacilli against the ureolytic pathogen yersinia enterocolitica. Current

Microbiology, 56(2), 175-181.

Le Bivic, A., Hirn, M., & Reggio, H. (1988). HT-29 cells are an in vitro model for the generation of

cell polarity in epithelia during embryonic differentiation. Proceedings of the National Academy

of Sciences of the United States of America, 85(1), 136-140.

Lee, D. J., Drongowski, R. A., Coran, A. G., & Harmon, C. M. (2000). Evaluation of probiotic

treatment in a neonatal animal model. Pediatric Surgery International, 16(4), 237-242.

Page 92: Bioactive Components from Lactobacillus acidophilus and

83

Levi, C. A., Ejere, V. C., Asogwa, C. N., Iweh, P., Nwatu, K. U., & Levi, U. E. (2014). Apoptosis: Its

physiological implication and therapeutic possibilities . Journal of Pharmacy and Biological

Sciences, 9(1), 38-45.

Li, P., Yin, Y., Yu, Q., & Yang, Q. (2011). Lactobacillus acidophilus S-layer protein-mediated

inhibition of salmonella-induced apoptosis in caco-2 cells. Biochemical and Biophysical

Research Communications, 409(1), 142-147.

Lin, C., Tsai, H., Lin, P., Tsen, H., & Tsai, C. (2008). Lactobacillus acidophilus LAP5 able to inhibit

the salmonella choleraesuis invasion to the human caco-2 epithelial cell. Anaerobe, 14(5),

251-255.

Ling, S., Wang, X. H., Xie, L., Lim, T. M., & Leung, K. Y. (2000). Use of green fluorescent protein

( GFP) to study the invasion pathways of edwardsiella tarda in in vivo and in vitro fish models.

MICROBIOLOGY-SGM; Microbiology-(UK), 146(1), 7-19.

Louis, K. S., & Siegel, A. C. (2011). Cell viability analysis using trypan blue: Manual and automated

methods. Mammalian Cell Viability: Methods and Protocols, 740, 7-12.

Lundberg, U., Vinatzer, U., Berdnik, D., von Gabain, A., & Baccarini, M. (1999). Growth

phase-regulated induction of salmonella-induced macrophage apoptosis correlates with transient

expression of SPI-1 genes. Journal of Bacteriology, 181(11), 3433-3437.

Ma, T. Y., Iwamoto, G. K., Hoa, N. T., Akotia, V., Pedram, A., Boivin, M. A., & Said, H. M. (2004).

TNF-alpha-induced increase in intestinal epithelial tight junction permeability requires

NF-kappa B activation. American Journal of Physiology-Gastrointestinal and Liver Physiology,

286(3), G367-G376.

Mack, D. R., Michail, S., Wei, S., McDougall, L., & Hollingsworth, M. A. (1999). Probiotics inhibit

enteropathogenic E- coli adherence in vitro by inducing intestinal mucin gene expression.

American Journal of Physiology-Gastrointestinal and Liver Physiology, 276(4), G941-G950.

Magrone, T., & Jirillo, E. (2013). The interplay between the gut immune system and microbiota in

health and disease: Nutraceutical intervention for restoring intestinal homeostasis. Current

Pharmaceutical Design, 19(7), 1329-1342.

Mantis, N. J., & Forbes, S. J. (2010). Secretory IgA: Arresting microbial pathogens at epithelial

borders. Immunological Investigations, 39(4-5), 383-406.

Masood, M. I., Qadir, M. I., Shirazi, J. H., & Khan, I. U. (2011). Beneficial effects of lactic acid

bacteria on human beings. Critical Reviews in Microbiology, 37(1), 91-98.

Mattar, A. F., Drongowski, R. A., Coran, A. G., & Harmon, C. M. (2001). Effect of probiotics on

enterocyte bacterial translocation in vitro. Pediatric Surgery International, 17(4), 265-268.

Matter, K., & Balda, M. S. (2003). Functional analysis of tight junctions. Methods, 30(3), 228-234.

Page 93: Bioactive Components from Lactobacillus acidophilus and

84

Medellin-Peña, M., & Griffiths, M. W. (2009). Effect of molecules secreted by lactobacillus

acidophilus strain la-5 on escherichia coli O157: H7 colonization. Applied and Environmental

Microbiology, 75(4), 1165-1172.

Medellin-Peña, M., Wang, H., Johnson, R., Anand, S., & Griffiths, M. W. (2007). Probiotics affect

virulence-related gene expression in escherichia coli O157: H7. Applied and Environmental

Microbiology, 73(13), 4259-4267.

Medellin-Peña, M. J. (2007). In Griffiths M. W. (Ed.), Interference of virulence in escherichia coli

O157:H7 by molecule(s) secreted by probiotic lactic acid bacteria (Ph.D Thesis ed.). Guelph,

Ont.: University of Guelph.

Meisel, H., & FitzGerald, R. J. (2003). Biofunctional peptides from milk proteins: Mineral binding

and cytomodulatory effects. Current Pharmaceutical Design, 9(16), 1289-1295.

Milovic, V., Turchanowa, L., Khomutov, A. R., Khomutov, R. M., Caspary, W. F., & Stein, J. (2001).

Hydroxylamine-containing inhibitors of polyamine biosynthesis and impairment of colon cancer

cell growth. Biochemical Pharmacology, 61(2), 199-206.

Minkiewicz, P., Dziuba, J., Iwaniak, A., Dziuba, M., & Darewicz, M. (2008). BIOPEP database and

other programs for processing bioactive peptide sequences. Journal of AOAC International,

91(4), 965-980.

Mitchell, D. M., & Ball, J. M. (2004). Craracterization of a spontaneously polarizing HT-29 cell line,

HT-29/cl.f8. In Vitro Cellular & Developmental Biology-Animal, 40(10), 297-302.

Moorthy, G., Murali, M. R., & Devaraj, S. N. (2009). Lactobacilli facilitate maintenance of intestinal

membrane integrity during shigella dysenteriae 1 infection in rats. Nutrition, 25(3), 350-358.

Muro Urista, C., Alvarez Fernandez, R., Riera Rodriguez, F., Arana Cuenca, A., & Tellez Jurado, A.

(2011). Review: Production and functionality of active peptides from milk. Food Science and

Technology International, 17(4), 293-317.

Myllyluoma, E., Ahonen, A. -., Korpela, R., Vapaatalo, H., & Kankuri, E. (2008). Effects of

multispecies probiotic combination on helicobacter pylori infection in vitro. Clinical and

Vaccine Immunology, 15(9), 1472-1482.

Ng, K. (2000). In Griffiths M. W. (Ed.), Enhancement of macrophage cytokine production by cell-free

fractions of fermented milk (M.Sc. Thesis ed.). Guelph, Ont.: University of Guelph.

Ouwehand, A. C., Salminen, S., & Isolauri, E. (2002). Probiotics: An overview of beneficial effects.

Antonie Van Leeuwenhoek, 82(1-4), 279-289.

Parvez, S., Malik, K., Kang, S., & Kim, H. (2006). Probiotics and their fermented food products are

beneficial for health. Journal of Applied Microbiology, 100(6), 1171-1185.

Page 94: Bioactive Components from Lactobacillus acidophilus and

85

Perdigon, G., Alvarez, S., Demacias, M. E. N., Roux, M. E., & Holgado, A. P. D. (1990). The

oral-administration of lactic-acid bacteria increase the mucosal intestinal immunity in response

to enteropathogens. Journal of Food Protection, 53(5), 404-410.

Perdigon, G., Demacias, M. E. N., Alvarez, S., Oliver, G., & Holgado, A. A. P. D. (1990). Prevention

of gastrointestinal infection using immunobiological methods with milk fermented with

lactobacillus-casei and lactobacillus-acidophilus. Journal of Dairy Research, 57(2), 255-264.

Pinto, M., Gomez, M. R., Seifert, S., Watzl, B., Holzapfel, W. H., & Franz, C. (2009). Lactobacilli

stimulate the innate immune response and modulate the TLR expression of HT29 intestinal

epithelial cells in vitro. International Journal of Food Microbiology, 133(1-2), 86-93.

Popov, S. G., Villasmil, R., Bernardi, J., Grene, E., Cardwell, J., Wu, A., . . . Alibek, K. (2002).

Lethal toxin of bacillus anthracis causes apoptosis of macrophages. Biochemical and Biophysical

Research Communications, 293(1), 349-355.

Punj, V., Bhattacharyya, S., Saint-Dic, D., Vasu, C., Cunningham, E. A., Graves, J., . . . Das Gupta, T.

(2004). Bacterial cupredoxin azurin as an inducer of apoptosis and regression in human breast

cancer. Oncogene, 23(13), 2367-2378.

Raff, M. (1992). Social controls on cell-survival and cell-death. Nature, 356(6368), 397-400.

Resta-Lenert, S., & Barrett, K. E. (2003). Live probiotics protect intestinal epithelial cells from the

effects of infection with enteroinvasive escherichia coli (EIEC). Gut, 52(7), 988-997.

Rodriguez-Boulan, E., & Nelson, W. J. (1989). Morphogenesis of the polarized epithelial cell

phenotype. Science (New York, N.Y.), 245(4919), 718-725.

Santini, M. T., Rainaldi, G., & Indovina, P. L. (2000). Apoptosis, cell adhesion and the extracellular

matrix in the three-dimensional growth of multicellular tumor spheroids. Critical Reviews in

Oncology and Hematology, 36(2), 75-87.

Schrezenmeir, J., & De Vrese, M. (2001). Probiotics, prebiotics, and synbiotics--approaching a

definition. American Journal of Clinical Nutrition, 73(2), 361S-364S.

Sgouras, D., M., Sgouras, D., Maragkoudakis, P., Petraki, K., Martinez-Gonzalez, B., . . . Mentis, A.

(2004). In vitro and in vivo inhibition of helicobacter pylori by lactobacillus casei strain shirota.

Applied and Environmental Microbiology, 70(1), 518-526.

Silva, S. V., & Malcata, F. X. (2005). Caseins as source of bioactive peptides. International Dairy

Journal, 15(1), 1-15.

Skehan, P., Storeng, R., Scudiero, D., Monks, A., McMahon, J., Vistica, D., . . . Boyd, M. R. (1990).

New colorimetric cytotoxicity assay for anticancer-drug screening. Journal of the National

Cancer Institute, 82(13), 1107-1112.

Page 95: Bioactive Components from Lactobacillus acidophilus and

86

Skjolaas, K. A., Burkey, T. E., Dritz, S. S., & Minton, J. E. (2007). Effects of salmonella enterica

serovar typhimurium, or serovar choleraesuis, lactobacillus reuteri and bacillus licheniformis on

chemokine and cytokine expression in the swine jejunal epithelial cell line, IPEC-J2. Veterinary

Immunology and Immunopathology, 115(3), 299-308.

Slee, E. A., Adrain, C., & Martin, S. J. (2001). Executioner caspase-3,-6, and-7 perform distinct,

non-redundant roles during the demolition phase of apoptosis. Journal of Biological Chemistry,

276(10), 7320-7326.

Solano, C., Sesma, B., Alvarez, M., Urdaneta, E., Garcia-Ros, D., Calvo, A., & Gamazo, C. (2001).

Virulent strains of salmonella enteritidis disrupt the epithelial barrier of caco-2 and HEp-2 cells.

Archives of Microbiology, 175(1), 46-51.

Steinke, J. W., & Borish, L. (2006). Cytokines and chemokines. Journal of Allergy and Clinical

Immunology, 117(2), S441-S445.

Strober, W. (2001). Trypan blue exclusion test of cell viability. Current Protocols in Immunology /

Edited by John E.Coligan ...[Et Al.], Appendix 3, 3B-Appendix 3B.

Sun, Z. L., Huang, L. H., Kong, J., Hu, S. M., Zhang, X. W., & Kong, W. T. (2012). In vitro

evaluation of lactobacillus crispatus K313 and K243: High-adhesion activity and

anti-inflammatory effect on salmonella braenderup infected intestinal epithelial cell. Veterinary

Microbiology, 159(1-2), 212-220.

Takeuchi, K., & Frank, J. F. (2001). Confocal microscopy and microbial viability detection for food

research. Journal of Food Protection, 64(12), 2088-2102.

Tellez Garay, A. M. (2009). In Griffiths M. W. (Ed.), Characterization of immuno active peptides

present in cell free preparations obtained from milk fermented by lactobacillus helveticus (Ph.D.

Thesis ed.). Guelph, Ont.: University of Guelph.

Tellez, A., Corredig, M., Turner, P. V., Morales, R., & Griffiths, M. W. (2011). A peptidic fraction

from milk fermented with Lactobacillus helveticus protects mice against Salmonella infection.

International Dairy Journal, 21(9), 607-614.

Tindall, B. J., Grimont, P. A. D., Garrity, G. M., & Euzeby, J. P. (2005). Nomenclature and taxonomy

of the genus salmonella. International Journal of Systematic and Evolutionary Microbiology, 55,

521-524.

Tlaskalova-Hogenova, H., Tuckova, L., Lodinova-Zadnikova, R., Stepankova, R., Cukrowska, B.,

Funda, D. P., . . . Sanchez, D. (2002). Mucosal immunity: Its role in defense and allergy.

International Archives of Allergy and Immunology, 128(2), 77-89.

Torchinsky, M. B., Garaude, J., & Blander, J. M. (2010). Infection and apoptosis as a combined

inflammatory trigger. Current Opinion in Immunology, 22(1), 55-62.

Page 96: Bioactive Components from Lactobacillus acidophilus and

87

Tuomola, E. M., Ouwehand, A. C., & Salminen, S. J. (1999). The effect of probiotic bacteria on the

adhesion of pathogens to human intestinal mucus. FEMS Immunology and Medical

Microbiology, 26(2), 137-142.

Valdez, J. C., Rachid, M., Gobbato, N., & Perdigon, G. (2001). Lactic acid bacteria induce apoptosis

inhibition in salmonella typhimurium infected macrophages. Food and Agricultural Immunology,

13(3), 189-197.

van Engeland, M., Nieland, L. J. W., Ramaekers, F. C. S., Schutte, B., & Reutelingsperger, C. P. M.

(1998). Annexin V-affinity assay: A review on an apoptosis detection system based on

phosphatidylserine exposure. Cytometry, 31(1), 1-9.

Vermes, I., Haanen, C., & Reutelingsperger, C. (2000). Flow cytometry of apoptotic cell death.

Journal of Immunological Methods, 243(1-2), 167-190.

Wang, F. J., Graham, W. V., Wang, Y. M., Witkowski, E. D., Schwarz, B. T., & Turner, J. R. (2005).

Interferon-gamma and tumor necrosis factor-alpha synergize to induce intestinal epithelial

barrier dysfunction by up-regulating myosin light chain kinase expression. American Journal of

Pathology, 166(2), 409-419.

Wu, M. T., Carlson, S. A., & Meyerholz, D. K. (2002). Cytopathic effects observed upon expression

of a repressed collagenase gene present in salmonella and related pathogens: Mimicry of a

cytotoxin from multiple antibiotic-resistant salmonella enterica serotype typhimurium phagetype

DT104. Microbial Pathogenesis, 33(6), 279-287.

Yan, F., Cao, H., Cover, T. L., Whitehead, R., Washington, M. K., & Polk, D. B. (2007). Soluble

proteins produced by probiotic bacteria regulate intestinal epithelial cell survival and growth.

Gastroenterology, 132(2), 562-575.

Ye, D. M., Ma, I., & Ma, T. Y. (2006). Molecular mechanism of tumor necrosis factor-alpha

modulation of intestinal epithelial tight junction barrier. American Journal of

Physiology-Gastrointestinal and Liver Physiology, 290(3), G496-G504.

Youakim, A., & Ahdieh, M. (1999). Interferon-gamma decreases barrier function in T84 cells by

reducing ZO-1 levels and disrupting apical actin. American Journal of

Physiology-Gastrointestinal and Liver Physiology, 276(5), G1279-G1288.

Yu, Q., Wang, Z., & Yang, Q. (2012). Lactobacillus amylophilus D14 protects tight junction from

enteropathogenic bacteria damage in caco-2 cells. Journal of Dairy Science, 95(10), 5580-5587.

Zanabria Eyzaguirre, R. (2013). In Corredig M. (Ed.), Anticarcinogenic and immunomodulatory

properties of the milk fat globule membrane (Ph.D Thesis ed.). Guelph, Ont.: University of

Guelph.

Page 97: Bioactive Components from Lactobacillus acidophilus and

88

Zeinhom, M., T., Zeinhom, M., Tellez, A. M., Delcenserie, V., El-Kholy, A., . . . Griffiths, M. W.

(2012). Yogurt containing bioactive molecules produced by lactobacillus acidophilus la-5 exerts

a protective effect against enterohemorrhagic escherichia coli in mice. Journal of Food

Protection, 75(10), 1796-1805.

Zemans, R. L., Briones, N., Campbell, M., McClendon, J., Young, S. K., Suzuki, T., . . . Downey, G.

P. (2011). Neutrophil transmigration triggers repair of the lung epithelium via beta-catenin

signaling. Proceedings of the National Academy of Sciences of the United States of America,

108(38), 15990-15995.