The Role of Bacterial Products and Intestinal Microbiota in Non … · 2015-11-24 · NAFLD...

133
The Role of Bacterial Products and Intestinal Microbiota in Non-Alcoholic Fatty Liver Disease by Hannah Elizabeth Da Silva A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Nutritional Sciences University of Toronto © Copyright by Hannah E Da Silva, 2015

Transcript of The Role of Bacterial Products and Intestinal Microbiota in Non … · 2015-11-24 · NAFLD...

The Role of Bacterial Products and Intestinal Microbiota in Non-Alcoholic Fatty Liver Disease

by

Hannah Elizabeth Da Silva

A thesis submitted in conformity with the requirements for the degree of Master of Science

Department of Nutritional Sciences University of Toronto

© Copyright by Hannah E Da Silva, 2015

ii

The Role of Bacterial Products and Intestinal Microbiota in

Non-Alcoholic Fatty Liver Disease

Hannah Elizabeth Da Silva

Master of Science

Department of Nutritional Sciences

University of Toronto

2015

Abstract

Non-alcoholic fatty liver disease (NAFLD) includes simple steatosis (SS) and non-alcoholic

steatohepatitis (NASH), which can progress to cirrhosis. NAFLD pathogenesis is complex.

Recent research suggests a role for the intestinal microbiota (IM) with various potential

mechanisms involving bacterial metabolism and products. This cross-sectional study compared

bacterial products and metabolites in the blood and feces, and fecal levels of total bacteria,

Bacteroidetes, C. coccoides, C. leptum, Bifidobacteria, Lactobacilli, E. coli and Archaea in

biopsy confirmed SS, NASH, and healthy controls (HC). NAFLD patients (SS+NASH) had

higher levels of bacterial fecal metabolites, including choline, trimethylamine, total short-chain

fatty acids, propionate, isobutyric acid, and higher serum 2-hydroxybutyrate than HC. NASH

patients had lower proportions of fecal C. leptum and higher amounts of E. coli than HC. Several

bacterial metabolites correlated with histological findings, and with specific bacteria and diet.

These findings suggest a potential role for bacteria and bacterial metabolism in NAFLD

pathogenesis.

iii

Acknowledgments

I would like to thank the many individuals who have contributed to this study and assisted me

during my graduate studies.

First, I must thank our study participants who were willing to sacrifice their time to be part of our

study during what was often a very stressful period in their lives.

Thank you to Dr. Allard who has been a constant source of motivation, support, and inspiration.

You are and will continue to be a great role model for my career.

Thank you as well to the members of my advisory committee: Dr. Elena Comelli, Dr. Thomas

Wolever, and Dr. Eberhard Renner. Your expertise and support has been influential throughout

this process. It was an honour to work with each of you.

A very special thank you is owed to Dr. Bianca Arendt who was involved in planning and

conducting this research long before me. Without your help in patient recruitment, grant writing,

presentation edits, and guidance in the lab, this thesis would not have been possible.

I am very grateful for the expertise and support of our wonderful statistician, Anastasia Teterina.

Thank you for your assistance with everything from course work to abstract writing to my final

analyses and your veterinary expertise. Our research group is very lucky to have you.

Thank you to my other officemates past and present, especially Katherine Schwenger and Sultan

Alenezi who have been with me since day one. A special thanks goes to Dr. Paulina Pettinelli

who assisted with stool homogenization.

Thank you to Dr. Amel Taibi who completed our DNA extractions and the qPCR analyses. I am

also very grateful for your willingness to answer all of my other laboratory associated questions.

Thank you to our many collaborators. Thank you to Dr. Scott Fung and Dr. David Wong who

assisted with the recruitment of NAFLD patients. Thank you to Dr. Ian McGilvray and the entire

Toronto General Hospital Multi-Organ Transplant Program team who assisted with recruiting

iv

healthy controls. I am very appreciative of the excellent work of Dr. Sandra Fischer who

assessed all liver biopsies.

Thank you to my colleagues and manager, Karen Smith, at Sunnybrook Health Science Centre

for continuing to keep me on as a Registered Dietitian while also giving me the time to complete

my graduate studies in a timely manner.

Lastly, but most importantly, I would like to thank my friends and family who have been a

source of encouragement and support and who have accepted my absence in the last several

months with understanding. Thank you to my brilliant husband Michael who believes that I can

do amazing things. Your patience and encouragement have not gone unnoticed.

v

Table of Contents

Acknowledgments.......................................................................................................................... iii

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

List of Tables ................................................................................................................................. ix

List of Figures ................................................................................................................................ xi

List of Appendices ....................................................................................................................... xiii

List of Abbreviations ................................................................................................................... xiv

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

2 Review of the Literature..............................................................................................................3

2.1 An Introduction to Non-Alcoholic Fatty Liver Disease Pathogenesis.................................3

2.1.1 Insulin Resistance ....................................................................................................3

2.1.2 Lipid Peroxidation and Lipotoxicity ........................................................................3

2.1.3 Mitochondrial and Endoplasmic Reticulum Stress ..................................................4

2.1.4 Genetic Factors ........................................................................................................4

2.1.5 Diet and Physical Activity .......................................................................................5

2.1.5.1 Carbohydrates ............................................................................................5

2.1.5.2 Fat ..............................................................................................................6

2.1.5.3 Vitamins ....................................................................................................7

2.2 Role of intestinal microbiota in non-alcoholic fatty liver disease .......................................7

2.2.1 Increase energy uptake .............................................................................................8

2.2.2 Increase Chronic Inflammation..............................................................................10

2.2.2.1 Toll-Like Receptor 4 and Inflammatory Cytokines ................................10

2.2.2.2 Endotoxin ................................................................................................10

2.2.2.3 Intestinal Permeability .............................................................................11

vi

2.2.3 The Role of Products of Bacterial Metabolism in NAFLD ...................................12

2.2.3.1 Short-Chain Fatty Acids ..........................................................................12

2.2.3.1.1 Butyrate .................................................................................................. 12

2.2.3.1.2 Propionate ............................................................................................... 13

2.2.3.1.3 Acetate .................................................................................................... 14

2.2.3.2 Ethanol .....................................................................................................14

2.2.3.3 Choline metabolism .................................................................................14

2.2.4 NAFLD and the Intestinal Microbiota: Clinical Studies .......................................15

2.2.5 Probiotic and Prebiotic Trials ................................................................................18

2.2.6 Proposed Mechanism .............................................................................................20

3 Research Aim and Hypotheses .................................................................................................21

4 Materials and Methods ..............................................................................................................22

4.1 Study Design ......................................................................................................................22

4.2 Clinical Data, Environmental Questionnaire, and Anthropometric Measurements ..........23

4.3 Nutrition and Activity Assessment ....................................................................................23

4.4 Clinical Biochemistry ........................................................................................................25

4.5 Liver Histology ..................................................................................................................26

4.6 Plasma Endotoxin ..............................................................................................................28

4.7 Serum Metabolites .............................................................................................................29

4.8 Stool Sample Collection and Analysis ...............................................................................31

4.8.1 Stool Homogenization ...........................................................................................31

4.8.2 Fecal Metabolite Analysis......................................................................................32

4.8.3 DNA Extraction .....................................................................................................32

4.8.4 Quantitative Real-time PCR ..................................................................................32

vii

4.9 Statistical Analysis .............................................................................................................33

4.9.1 Data Analysis .........................................................................................................33

4.9.2 Power Calculations ................................................................................................34

5 Results .......................................................................................................................................35

5.1 Subjects ..............................................................................................................................35

5.2 Dietary Intake and Physical Activity .................................................................................40

5.3 Bacterial Products ..............................................................................................................45

5.3.1 Plasma Endotoxin ..................................................................................................45

5.3.2 Short Chain Fatty Acids and Other Organic Acids ................................................46

5.3.3 Metabolites Related to the Bacterial Metabolism of Choline ................................49

5.3.4 Ethanol ...................................................................................................................52

5.4 Intestinal Microbiota ..........................................................................................................52

5.5 Relationships between Bacterial Metabolites, Bacteria of Interest, Disease Status, and

Diet .....................................................................................................................................55

6 Discussion .................................................................................................................................66

6.1 Endotoxin ...........................................................................................................................66

6.2 SCFA and Related Metabolites ..........................................................................................68

6.3 Metabolites Related to the Bacterial Metabolism of Choline ............................................71

6.4 Ethanol ...............................................................................................................................73

6.5 Intestinal Microbiota ..........................................................................................................73

6.6 Diet and Physical Activity .................................................................................................75

6.7 Potential Confounders ........................................................................................................76

7 Strengths and Limitations .........................................................................................................79

8 Conclusions ...............................................................................................................................81

9 Future Directions .......................................................................................................................82

References ......................................................................................................................................84

viii

Appendices ...................................................................................................................................104

ix

List of Tables

Page

Table 1: Summary of human studies on intestinal microbiota and non-alcoholic fatty

liver disease

17

Table 2: Bacteria of interest in non-alcoholic fatty liver disease 19

Table 3: Physical activity coefficients for estimated energy expenditure calculation 25

Table 4: Assessment of liver histology: Brunt System 27

Table 5: Metabolites measured in stool and serum samples 31

Table 6: Standard curves used in the interpretation of the qPCR results 33

Table 7: Subject demographics and health history 37

Table 8: Biochemistry by disease group 38

Table 9: Liver histology by disease group 39

Table 10: Reported dietary intake and estimated requirements: Macronutrients 41

Table 11: Reported dietary intake and estimated requirements: Micronutrients 43

Table 12: Fecal and serum SCFA and organic acids in healthy controls and NAFLD

patients

49

Table 13: Bacterial taxa of interest from qPCR by disease group 53

Table 14: Correlation between fecal and blood metabolites and NAFLD related

parameters

57

Table 15: Correlation between intestinal microbiota by qPCR and NAFLD related 60

x

parameters

Table 16: Correlation between intestinal microbiota and metabolites in all subjects 62

Table 17: Correlations between metabolites and diet in all subjects 63

Table 18: Correlations between intestinal microbiota by qPCR and diet in all subjects 64

xi

List of Figures

Page

Figure 1: Reported proportion of estimated energy requirement consumed 42

Figure 2: Reported proportion of basal metabolic rate consumed 42

Figure 3: Reported total daily physical activity completed 44

Figure 4: Reported daily minutes of strenuous and very strenuous physical activity 44

Figure 5: Comparison of plasma endotoxin concentration between groups 45

Figure 6: Comparison of fecal butyric acid between groups 47

Figure 7: Comparison of fecal propionate between groups 47

Figure 8: Comparison of fecal acetic acid between groups 48

Figure 9: Comparison of serum acetic acid between groups 48

Figure 10: Fecal choline in healthy controls and NAFLD patients 50

Figure 11: Fecal trimethylamine in healthy controls and NAFLD patients 50

Figure 12: Serum choline in healthy controls and NAFLD patients 51

Figure 13: Serum trimethylamine N-oxide in healthy controls and NAFLD patients 51

Figure 14: Serum ethanol in healthy controls and NAFLD patients 52

Figure 15: C. leptum relative abundance in stool samples from three study groups 54

Figure 16: E. coli count in stool samples from three study groups 55

Figure 17: Relationship between fecal formic acid and total inflammation in NAFLD 58

xii

patients

Figure 18: Relationship between fecal choline and body mass index in all subjects 58

Figure 19: Relationship between fecal trimethylamine and body mass index in all

subjects

59

Figure 20: Relationship between serum acetic acid and hepatocyte ballooning intensity

in NAFLD patients

59

Figure 21: Relationship between serum choline and liver steatosis in NAFLD patients 60

Figure 22: Relationship between Firmicutes count and liver steatosis in NAFLD

patients

61

Figure 23: Relationship between C. leptum count and fecal butyric acid in all subjects 61

Figure 24: Relationship between relative abundance of C. leptum and reported dietary

fibre intake in all subjects

65

Figure 25: Relationship between plasma endotoxin and reported carbohydrate intake in

all subjects

65

xiii

List of Appendices

Page

Appendix 1: Consent Forms 104

Appendix 2: Instructions for stool sample collection and transportation 111

Appendix 3: Environmental Questionnaire 112

Appendix 4: 7-day Food and Activity Log 115

xiv

List of Abbreviations

ALP Alkaline Phosphatase

ALT Alanine Transaminase

AST Aspartate Transaminase

BMR Basal Metabolic Rate

CV Coefficient of Variation

EDTA Ethylenediaminetetraacetic Acid

EER Estimated Energy Requirement

ES Effect Size

FFA Free Fatty Acid

FMO3 Flavin Monooxygenase 3

GLP-1 Glucagon-like Peptide

GLUT4 Glucose Transporter Type 4

GPR43 G-protein Coupled Receptor 43

HbA1c Hemoglobin A1c

HC Healthy Controls

HDL High Density Lipoprotein

HOMA-IR Homeostasis Model Assessment Estimated Insulin Resistance

IL-6 Interleukin 6

xv

IM Intestinal Microbiota

kcal Kilocalorie

LAL Limulus Amebocyte Lysate

LDL Low Density Lipoprotein

MM Master Mix

MRI Magnetic Resonance Imaging

n-3 Omega-3

NAFLD Non-Alcoholic Fatty Liver Disease

NAS NAFLD Activity Score

NASH Non-Alcoholic Steatohepatitis

NMR Nuclear Magnetic Resonance

NSAID Nonsteroidal Anti-inflammatory Drug

PA Physical Activity Coefficient

PAI-1 Plasminogen Activator Inhibiter

PEMT Phosphatidylethanolamine N-methyltransferase

PNLPA3 Patatin-like Phospholipase 3

PUFA Polyunsaturated Fatty Acid

qPCR Quantitative Polymerase Chain Reactions

ROS Reactive Oxygen Species

xvi

SCFA Short-Chain Fatty Acid

SD Standard Deviation

SS Simple Steatosis

TG Triglycerides

TLR-4 Toll-like Receptor 4

TMAO Trimethylamine N-oxide

TNF-α Tumor Necrosis Factor Alpha

VLDL Very Low Density Lipoprotein

1

1 Introduction

Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in the

Western World [1]. NAFLD describes a spectrum of disease severity from simple steatosis (SS)

to non-alcoholic steatohepatitis (NASH) in the absence of significant alcohol consumption

(<20g/day) [2]. SS is defined by the accumulation of fat (triglycerides) within the hepatocytes

without hepatocyte injury and is considered a benign condition [2]. NASH involves steatosis and

inflammation as well as hepatocyte injury which presents as ballooning and may also include

spotty necrosis and/or fibrosis [2]. The gold standard for the diagnosis of NASH is liver biopsy

[2] where histology is assessed using the Brunt scale to classify SS versus NASH and to grade

disease severity using the NAFLD Activity Score (NAS) [3].

The World Gastroenterology Organisation estimates the prevalence of NALFD in the Western

World to be approximately 27-34% in the general population, upwards of 75% in those with

diabetes or obesity, and 75-92% in the morbidly obese [1]. The prevalence of NASH is

significantly lower, estimated at 2-3% in the general population [4], but upwards of 18.5-30% in

obese individuals [4, 5], and 55% in diabetics [6]. Approximately 3-15% of individuals with

NASH will advance to cirrhosis, while 38-45% of those with cirrhosis will advance to liver

failure within 7-10 years [1]. NASH cirrhosis is the third most common indication for liver

transplant in the United States and has increased as the primary indication for liver transplant

from 1.2% to 9.7% of all transplants performed from 2001 to 2009 [7].

Currently there are limited treatment options for NAFLD. The American College of

Gastroenterology guidelines (2012) recommends weight loss via diet and exercise or bariatric

surgery. Vitamin E supplementation is also recommended in non-diabetics [2]. However, due to

difficulties achieving adequate weight loss, eligibility criteria limiting the number of candidates

for, and the risk and invasiveness of bariatric procedures [8], few viable options exist for treating

NAFLD and preventing its progression.

A significant barrier in developing these treatments is that the pathogenesis of NAFLD is

complex and it is still not completely understood why some remain with only SS while others

progress to the more severe form, NASH, with risks of developing cirrhosis and liver failure [9].

Lately, there has been emerging literature on the role of the intestinal microbiota in the

2

pathogenesis of NAFLD [10-15]. The aim of this research project was to characterize the

intestinal microbiota of patients with NASH or SS versus healthy controls (HC) and determine if

there were any differences in bacterial products or metabolites that could play a role in the

pathogenesis of this disease.

3

2 Review of the Literature

2.1 An Introduction to Non-Alcoholic Fatty Liver Disease Pathogenesis

2.1.1 Insulin Resistance

Individuals with NAFLD are universally insulin resistant [16] and this is considered to be a

driving factor behind much of NAFLD pathogenesis. Traditionally, the first stage of NAFLD

pathogenesis is the accumulation of lipids within the hepatocytes, called steatosis. This occurs

when the supply of free fatty acids (FFA) to the liver exceeds the needs for mitochondrial

oxidation, phospholipid synthesis, and cholesterol ester synthesis [17-19]. Insulin resistance is

associated with lipolysis from adipose tissue leading to increased circulating FFA and uptake by

the liver [20]. FFA may also be supplied to the liver through dietary intake and absorption as

chylomicrons. FFA uptake by the liver is unregulated [21] and therefore thought to be

responsible for about two thirds of the lipid accumulation in the liver [22]. Elevated insulin

levels resulting from insulin resistance also inhibit mitochondrial β-oxidation [23] and increase

de novo lipogenesis in the liver via glycolysis [24]. High insulin levels can also inhibit the

hepatic release of fatty acids as very low density lipoproteins (VLDL) by decreasing the

production of apolipoprotein B100 [24].

The majority of lipids in steatosis are stored as triglycerides [25] which are benign [26] and have

recently even been considered as protective against the oxidative stress and cellular damage that

is seen in the progression from SS to NASH [27, 28]. It is therefore the FFA accumulation which

causes the deleterious effects of steatohepatitis.

2.1.2 Lipid Peroxidation and Lipotoxicity

An increased presence of FFA in the liver leads to lipid peroxidation causing the release of

inflammatory cytokines such as tumor-necrosis factor alpha (TNF-α) and interleukin six (IL-6)

and reactive oxygen species (ROS) [29-31]. The resulting inflammation and oxidative stress

from the release of these molecules leads to disease progression from the benign SS to the

cellular damage and inflammation seen in NASH [32, 33]. Oxidative stress has also been shown

to impair the proliferation of hepatocyte progenitors leading to impaired tissue recovery, fibrosis,

and cirrhosis [34, 35].

4

The presence of FFA in hepatocytes may also have a direct toxic effect [36]. FFA is thought to

be responsible for the significantly higher presence of apoptosis in NASH patients compared to

healthy controls [37-39]. The type of fatty acid also seems to have an effect with saturated fatty

acids causing more apoptosis [40] than monounsaturated [36] or polyunsaturated fatty acids [41].

2.1.3 Mitochondrial and Endoplasmic Reticulum Stress

Another important component of NAFLD pathogenesis is the stress that the influx of fatty acids

causes to the cellular organelles, particularly the mitochondria and endoplasmic reticulum.

Mitochondrial stress and resulting dysfunction further promotes cellular damage through TNF-α

and over-production of ROS [30, 42]. An accumulation of FFA can also lead to endoplasmic

reticulum stress [43] which triggers the unfolded protein response. The unfolded protein response

is an adaptive response initiated by the endoplasmic reticulum when its main function, protein

folding, is disrupted by a variety of causes including the accumulation of FFA, a lack of energy

from glucose, or an insufficient presence of calcium [44]. This response may start an

inflammatory cascade which has been associated with apoptosis and exacerbation of metabolic

diseases including obesity and insulin resistance, contributing to NAFLD pathogenesis [45].

2.1.4 Genetic Factors

Research has shown that genetic factors may play some role in NAFLD development and

progression. The most consistent finding has been with polymorphisms in patatin-like

phospholipase three (PNLPA3) [46-48]. Polymorphisms in PNLPA3 have been associated with

an increased percentage of steatosis in NAFLD [49]. The homozygous 148M polymorphism for

PNLPA3 has also been associated with NAFLD severity, including inflammation and fibrosis

[50-52]. The Genetics of Obesity-Related Liver Disease Consortium found four additional gene

polymorphisms that were associated with NAFLD. These were found in or near the genes:

lysophospholipase like 1, protein phosphatase 1 regulatory subunit 3b, neurocan, and

glucokinase regulatory protein [53]. Peterson et al. additionally found that the polymorphisms C-

482T and T-455C in the gene for apolipoprotein 3C were associated with NAFLD and insulin

resistance [54]. The role of genetics in NAFLD pathogenesis is still emerging.

5

2.1.5 Diet and Physical Activity

As obesity is a main risk factor for the development of SS and NASH it follows that energy

balance, the difference between energy intake through diet and energy expenditure through the

basal metabolic rate, thermic effect of food, and physical activity, could play an important role in

NAFLD pathogenesis. Observational studies have consistently found that individuals with

NAFLD have significantly lower fitness level and/or participation rates in physical activity [55-

60]. The association between NAFLD and excessive energy intake, however, has only been

shown in a few studies [61-63]. This relatively weak association in the literature may be due to

underreporting by study participants, which is consistently encountered in studies using food

frequency questionnaires and self-reported diet records, especially among obese individuals [64,

65]. This led to a more significant focus on the composition of the food consumed rather than the

caloric content. A study from our research group, for instance, found that energy intake and

macronutrient composition did not differ between healthy controls and biopsy confirmed SS and

NASH patients, but that NASH patients were significantly more likely to have inadequate

consumptions of micronutrients such as vitamin B6, folate, and zinc [60]. It is difficult at this

point to think of a role for these micronutrients in the pathogenesis of NASH as other reports on

low micronutrient intakes were primarily focused on antioxidants [66-68]. There is also

additional literature on macronutrients, particularly fat and carbohydrates. Further details

regarding dietary composition are discussed below.

2.1.5.1 Carbohydrates

Two observational studies have found an association between the percent of energy consumed

from carbohydrates and the risk of advanced NAFLD [69, 70]. Solga et al. investigated dietary

intake using a 24 hour recall in obese patients undergoing bariatric surgery. Liver biopsies taken

at the time of surgery were evaluated for markers of inflammation and fibrosis. Higher percent of

energy from carbohydrates was associated with higher odds of having inflammation on liver

histology [69]. A similar study using a non-validated dietary questionnaire found that higher

percent of energy from carbohydrates was associated with a greater risk of fibrosis [70].

Carbohydrate quality also appears to be important. The consumption of sugar sweetened

beverages [71], soft drinks [57, 72], and fructose [73, 74] have been associated with a higher risk

of NAFLD. Fructose has been of particular interest in NAFLD pathogenesis, upregulating de

6

novo lipogenesis, promoting lipogenesis, inhibiting β-oxidation, and contributing to the

inflammatory cascade [75]. Fructose may also promote hepatic insulin resistance, further

contributing to NAFLD development and progression [76, 77].

Diets low in beneficial carbohydrates such as fibre have also been observed in NAFLD patients

[62, 78]. There are two main mechanisms through which fibre intake can influence NAFLD

pathogenesis. Some fibres, such as cellulose and pectins can be fermented by the intestinal

microbiota (IM) to produce short-chain fatty acids (SCFA) [79] which can exert many potentially

beneficial roles in preventing NAFLD. There is more discussion on this in section 2.2.3.1 as this

is relevant to this project. Fibre can also aid in increasing satiety [80-82] and improving glycemic

control [83], including insulin sensitivity in NAFLD patients [84].

2.1.5.2 Fat

Several observational studies have found a higher dietary fat intake in individuals with NAFLD

[62, 85-87] and NASH [88, 89] compared to healthy controls. Of these, saturated fats may

increase hepatic inflammation, fibrosis and apoptosis [90, 91]. Several studies have found that

there was a greater consumption of saturated fats in NAFLD patients [62, 73, 78, 85]. On the

other hand, mono- and polyunsaturated fatty acids (PUFA), particularly omega-3 fatty acids, may

be of benefit because of their anti-inflammatory effect [92]. Inadequate intake of these fatty acids

in NAFLD patients was reported in several studies [62, 73, 78, 93], particularly for omega-3 (n-

3) fatty acids [73, 89, 94].

As discussed in section 2.1.2, dietary fat is a main source of FFA in the liver and those with

several double-bonds, like PUFA, are susceptible to lipid peroxidation, triggering oxidative

stress and inflammation [29-31]. PUFA are fatty acids with multiple double bonds which can be

classified at n-3, n-6, or n-9 depending on the placement of the first double bond. PUFA are

particularly susceptible to lipid peroxidation due to their many double bonds. However,

elongation of n-3 causes the production of anti-inflammatory eicosanoids but elongation of n-6

results in the production of pro-inflammatory prostaglandins and leukotrienes [92]. Therefore,

the ratio of n-3:n-6 fatty acids can be important when considering disease progression through

hepatic inflammation. Several studies found differences in fatty composition with reduction in n-

3 or n-3:n-6 ratio in NASH [95-98]. Of additional note are findings that low levels of hepatic

7

PUFA, especially n-3 PUFA, are associated with a predisposition to lipogenesis over β-oxidation

and export as VLDL [95, 99, 100].

2.1.5.3 Vitamins

Due to the driving force of oxidative stress in NAFLD pathogenesis it follows that diets deficient

in antioxidant nutrients, including vitamin C and E, could promote NAFLD development and

progression. Cross-sectional studies have found that dietary vitamin C [62, 78] and vitamin E

[78] intake are lower in NAFLD patients compared to healthy controls. Vos et al. found that

patients with lower vitamin E consumption had a higher grade of steatosis [66]. Plasma vitamin

C and E levels have also been associated with disease status. Cankutaran et al. found that

NAFLD patients with vitamin E deficiency had worse steatosis [67] while another study found

that plasma vitamin E and C were lower in patients with NASH than with SS [68]. Given these

findings and positive results from clinical trials [101] the current treatment guidelines suggest

treating all non-diabetic, biopsy-proven NASH patients with 800 IU of vitamin E daily [2].

Choline is another nutrient found to play a role in NAFLD pathogenesis. It is discussed in the

context of intestinal microbiota in section 2.2.3.3.

Therefore, there are several factors playing a role in the pathogenesis of NAFLD and lack of a

healthy diet and sedentary lifestyle are important contributors. However, there is emerging

literature suggesting that the intestinal microbiota may also play a role, possibly influenced by

environmental factors such as diet and lifestyle. A role for intestinal microbiota is supported by a

recent study from our group [13], showing a lower percentage of Bacteroidetes in NASH patients

compared to healthy controls, independent of diet and body mass index. Therefore the present

project was developed to further assess this relationship and determine if certain bacterial

products or metabolites were associated with NAFLD.

2.2 Role of intestinal microbiota in non-alcoholic fatty liver disease

The human intestinal microbiota (IM) is composed of all microorganisms present in the gut. The

IM is estimated to contain more than 1013 microorganisms [102] with between 15 000 and 35

000 species of bacteria [103]. The dominant phyla, Bacteroidetes and Firmicutes, make up

approximately 90% of the bacteria in the human digestive system [104-107]. Until recently, very

8

little was known about the human IM composition because most IM bacteria are obligate

anaerobes making them difficult to culture. Recent technological advances now allow

researchers to identify IM species and groups and their relative abundance through the

sequencing of the 16s rRNA gene in bacterial DNA [108]. Since the advent of this technology,

researchers have discovered the many roles that the IM plays in human health and disease,

including its potential role in NAFLD pathogenesis.

2.2.1 Increase energy uptake

One of the primary mechanisms through which the IM is thought to influence NAFLD

development is by increasing energy absorption from the intestinal tract. In 2004, Bäckhed et al.

colonized germ-free mice with the IM of conventionally raised mice [109]. Within 14 days, the

colonized mice had a 60% increase in body fat and development of insulin resistance despite

lower oral intake [109]. The proposed mechanism for this adiposity was an increased absorption

of monosaccharides resulting from alterations in the IM which then lead to an increase in de

novo lipogenesis [109]. The increased absorption was proposed to be due to an increase in the

vascularization of the small intestinal villus epithelium [110]. An alternative mechanism was

suggested by Turnbaugh et al. when metagenomic analyses from the microbiota of ob/ob and

lean mice found that the ob/ob microbiome was enriched with genes coding for proteins involved

in the breakdown of “indigestible” polysaccharides and import of fermentation products. The

feces of these ob/ob mice also showed lower caloric content when measured by bomb

calorimetry in comparison to lean mice [111]. A subsequent study from Bäckhed et al. found that

germ-free mice were protected from diet induced obesity, gaining less weight and maintaining

lower blood insulin, glucose, and FFA levels than conventionally raised mice when consuming a

high-fat, high-sugar Western diet [112].

This relationship has also been studied by treating obese mice with antibiotics when exposed to

an obesogenic (high fat, high sugar) diet. Changes in IM related to antibiotic treatment resulted

in decreased body weight, fat mass, and glucose intolerance compared to controls [113] and a

significant decrease in hepatic lipids after steatosis development on a fructose rich diet [114]. Le

Roy et al. ran an interesting study that determined that NAFLD risk may be related to the IM,

independent of obesity. Mice were fed a high fat diet leading to weight gain. Even though all

mice gained weight, some mice also developed hyperglycemia and increased circulating

9

inflammatory cytokines [115]. These mice were called “responders”. Other mice remained

normoglycemic with low cytokine levels and were called “nonresponders”. A donor mouse was

selected from each group and IM was transplanted into recipient mice who were also placed on a

high fat diet. All recipient mice became obese. Mice who received IM from a “responder”

developed hepatic steatosis and insulin resistance with higher HOMA-IR, blood glucose, and

insulin, and increased expression of genes involved in de novo lipogenesis than mice who

received IM from the “non-responder”. Responder and non-responder recipients had distinct IM,

suggesting that a certain IM composition lead to increased risk of developing NAFLD in

response to high dietary fat intake [115]. Therefore, animal studies support the role of IM in

obesity. Since obesity is associated with NAFLD, it is possible that IM will also contribute to

NAFLD pathogenesis.

Human studies on obesity have also reported differences in the bacterial communities of lean

versus obese adults [116-121] which are similar to animal studies. The first landmark study that

suggested an association between obesity and the IM was published by Ley et al. in 2006 [116].

This simple study measured the IM of 12 obese patients for one year as they undertook a calorie-

restricted diet that was either low-fat or low-carbohydrate. Before starting the diet obese patients

had fewer Bacteroidetes and proportionally more Firmicutes. Through the course of their weight

loss, Bacteroidetes increased and Firmicutes decreased with a significant positive correlation

between proportion of Bacteroidetes and percentage of body weight lost. This was the start of a

long debate on whether obesity in humans is associated with a lower abundance of Bacteroidetes

and a proportionally increased abundance of Firmicutes. Most studies agree that there is a

relationship [116, 117, 120, 121].

In studies that have also investigated bacterial function via metagenomics this difference in

bacterial community was accompanied by an increase in genes associated with macronutrient

metabolism [118, 119, 121]. The first study of this kind looked at the IM and microbiome

(bacterial genes) of monozygotic and dizygotic twins and their mothers [119]. Obesity was

associated with a lower overall bacterial diversity. At the metagenomics level they found that

383 genes were significantly different between obese and lean subjects and that 75% of the

obesity enriched genes were from Actinobacteria (versus 0% of the lean enriched genes) and

42% of the lean enriched genes were from Bacteroidetes (versus 0% of the obese enriched

genes). Functionally, these genes were associated with carbohydrate, lipid, and amino acid

10

metabolism [119]. This, again, suggests a role of bacterial energy harvest in obesity. Jumpertz et

al. found that a 20% increase in Firmicutes was associated with a 150 kilocalorie (kcal) increase

in energy harvest [121], a value that, if it occurred daily, would amount to about 1.3lb weight

gain in one month or a nearly 16lb weight gain over the course of a year. It is therefore clear that

alterations in the IM can be associated with increased energy absorption, leading to increased

adiposity, which could in turn result in a greater risk of NAFLD.

2.2.2 Increase Chronic Inflammation

IM can also contribute to chronic systemic inflammation through several mechanisms.

Considering that chronic systemic inflammation plays a role in insulin resistance and obesity

[122], this certainly has relevance to NAFLD.

2.2.2.1 Toll-Like Receptor 4 and Inflammatory Cytokines

There are several mechanisms leading to the increased production of inflammatory cytokines in

NAFLD. One of them is the accumulation of hepatic triglycerides and particularly FFA

promoting inflammation through the activation of toll-like receptor 4 (TLR-4). This mechanism

is supported by animal studies. Cai et al. used a transgenic mouse model to show that hepatic

triglyceride accumulation leads to the activation of NF-kB by TLR-4 which causes the release of

inflammatory cytokines such as IL-6 and TNF-α [123]. TLR-4 activation has also been

associated with insulin resistance [124], the activation of Kupffer cells (involved in

fibrogenesis), and worsening liver histology [125]. The IM can also play a role in this as another

mechanism activating TLR-4 is through exposure to lipopolysaccharide or endotoxin, which is a

constituent of the outer membrane of Gram negative bacteria, which can cross the intestinal

epithelium [126]. Several studies support a role for endotoxin, which is discussed in the next

section.

2.2.2.2 Endotoxin

Increased endotoxin levels have been associated with NAFLD pathogenesis in both animal [124,

127, 128] and human studies [129-132]. Cani et al. found that after four weeks on a high-fat diet

mice developed a chronic increase in plasma endotoxin with an increased proportion of

endotoxin producing IM [127]. To further examine the effect of endotoxemia on mice the

investigators provided a constant infusion of endotoxin which resulted in increased fasting

11

glucose, insulin, and weight, including increased liver size and adipose tissue [127]. Brun et al.

found that obese, leptin deficient mice had higher levels of endotoxin leading to inflammatory

and fibrogenic hepatic responses [128]. Similar findings were published by Song et al. from

mouse and tissue culture experiments [124] and Spruss et al. in studies on mice fed a fructose

rich diet [133].

The first study that examined the role of endotoxin in NAFLD pathogenesis in humans was by

Thuy et al. [129]. This cross-sectional study found that NAFLD patients had elevated hepatic

expression of plasminogen activator inhibitor (PAI-1), which is used as a marker for hepatic

damage, compared to healthy controls. PAI-1 was associated with an increased endotoxin

concentration and hepatic expression of TLR-4. Harte et al. found that NAFLD patients had a

significantly higher plasma endotoxin level than healthy controls and that endotoxin

concentration was positively correlated with insulin resistance [130]. Another study measured

IgG levels against endotoxin as a marker of chronic low level endotoxin exposure over time in

severely obese NASH patients and severely obese healthy controls [131]. They found that

despite both groups being obese, there was a significantly higher IgG level in NASH patients,

which increased with NASH grade (severity) [131]. Volynets et al. similarly found that NAFLD

patients had higher endotoxin and PAI-1, and additionally higher blood ethanol levels, and

increased intestinal permeability than healthy controls [132].

2.2.2.3 Intestinal Permeability

Endotoxemia is present when there is an increase in intestinal permeability and this may also be

influenced by IM. The finding of increased intestinal permeability is further supported by Miele

et al. who found that NAFLD patients had higher gut permeability and prevalence of small

intestinal bacterial overgrowth, and lower expression of tight junction protein ZO-1 than healthy

controls, however, the barrier deficiency was not as severe as in untreated celiac disease patients

[134]. A recent study by Jiang et al. found significant differences in the IM community between

NAFLD patients and healthy controls. They also saw increased tight junction gaps, decreased

numbers of vital immune factors (CD4+ and CD8+ T lymphocytes), and increased expression of

inflammatory markers (TNF-α, IL-6) in the duodenal mucosa of NAFLD patients compared to

healthy controls [15]. These studies suggest that NAFLD patients have increased blood

endotoxin levels from an increase in endotoxin producing gram negative bacteria and/or

12

increased intestinal permeability which may lead to hepatic inflammation through the activation

of TLR-4. However, these studies do not differentiate between patients with the generally benign

SS and the more detrimental NASH. In addition, other bacterial products or metabolites may also

play a role.

2.2.3 The Role of Products of Bacterial Metabolism in NAFLD

2.2.3.1 Short-Chain Fatty Acids

One potential cause of the increased intestinal permeability observed in NAFLD patients is a

decrease in the colonic production of butyrate. About 10-30% of consumed energy reaches the

colon without being absorbed [135, 136]. These substrates, primarily starches and components of

the plant cell-wall including cellulose, pectins, xylans, arabinogalactans, gums and mucilage, are

fermented by the IM to form SCFA [79] and gases including CO2, CH4, and H2 [137]. The main

SCFA produced in the human colon are acetate, propionate, and butyrate in a molar ratio of

about 60:20:20 which remains in the same proportions in the feces [138-140]. SCFA are a source

of caloric intake and account for up to 10% of required daily energy intake in humans [135].

SCFA can also have other significant functions in NAFLD pathogenesis pathways. Lin et al.

conducted a study on mice to look at the effects of SCFA on obesity and related measures [141].

They found that the administration of any of the three main SCFA protected against the

development of obesity and insulin resistance on a high fat diet, but that only butyrate and

propionate regulated food intake through the stimulation of gut hormones [141]. SCFA have also

been shown to upregulate fatty acid oxidation [142]. Other important functions of butyrate,

propionate, and acetate are discussed below.

2.2.3.1.1 Butyrate

Butyrate’s main functional role is as an energy source for colonic mucosal cells, however it also

strengthens this important immunological barrier through involvement in cell proliferation and

differentiation [137, 143, 144]. Butyrate decreases intestinal permeability through effects on tight

junction proteins [145-147] and may have anti-carcinogenic effects in the colon [148, 149].

Butyrate has also been demonstrated to have a significant anti-inflammatory role through the

inhibition of NF-κB [150, 151] and may exhibit beneficial antioxidant functions, reducing the

production of reactive oxygen species [152]. All of these functions could prove useful in

preventing the progression of SS to NASH.

13

Butyrate producers are strict anaerobes and more than 90% belong to the Eubacterium or

Roseburia genus from clostridium cluster XIVa or are Faecalibacterium prausnitzii from

clostridium cluster IV [153]. A study by Wong et al. found that patients with NASH had a lower

abundance of Faecalibacterium and therefore perhaps produced less butyrate, however,

unfortunately SCFA were not measured in this study [14]. A probiotic supplementation study in

rats on a NAFLD inducing choline deficient diet found that providing a probiotic that includes

butyrate producing bacteria (Clostridium butyricum, MIYAIRI 588) led to lower blood

endotoxin, insulin resistance, hepatic markers of inflammation, and hepatic lipid deposition

[154]. The treated rats also had increased expression of tight junction proteins and decreased

hepatic fibrosis and hepatocarcinogenesis [154]. A similar study in a mice model found that

supplementation with the VSL#3 probiotic led to alterations in the IM, reduction in insulin

resistance, weight, inflammatory markers (TNF-α, IL-6), FFA, and steatosis [155]. These

beneficial effects were associated with increased levels of butyrate in the feces and plasma [155].

These findings suggest that butyrate and butyrate producing bacteria may have a beneficial effect

in NAFLD by reducing intestinal permeability and translocation of bacterial products, thereby

reducing chronic inflammation and insulin resistance. However, no human studies have assessed

both butyrate and IM in stools.

2.2.3.1.2 Propionate

Propionate is primarily produced by colonic fermentation by Bacteroides, Prevotella, and

Clostridium [156]. Thirty to fifty percent of propionate is taken up by the liver and the rest is

taken up by the peripheral tissues [157, 158]. Propionate inhibits lipolysis in adipocytes,

reducing plasma FFA, through the activation of free fatty acid receptor 2 [159, 160] and inhibits

the production of new fatty acids in the liver [161, 162]. Propionate also has several anti-

inflammatory properties, reducing the production of pro-inflammatory eicosanoids [163] and

resistin, a pro-inflammatory cytokine [164]. Propionate has also been shown to inhibit the release

of TNF-α by neutrophils in response to bacterial endotoxin, a proposed significant contributor to

the inflammation seen in NASH [165]. As briefly mentioned above, propionate may also play a

role in increasing satiety and has been shown to promote the release of leptin [159, 164, 166] and

improve glucose uptake by peripheral tissues though increasing glucose transporter type four

(GLUT4) [167]. Therefore, propionate may have beneficial effects in NAFLD.

14

2.2.3.1.3 Acetate

Acetate is produced by many bacteria, particularly Bacteroidetes and Bifidobacterium [156, 168].

It is primarily used as an energy source in the liver and for the production of cholesterol [158].

Acetate may also have an anti-inflammatory function through interaction with G-protein coupled

receptor 43 (GPR43) [169]. Lastly, a study by Fukuda et al. suggested that the main source of

Bifidobacterium’s prevention against intestinal infection by pathogens (E. coli) was its

production of acetate [170]. The various functions of butyrate, propionate, and acetate are closely

linked with various steps in NAFLD pathogenesis and warrant further investigation in NAFLD

populations. Human studies in this area are lacking.

2.2.3.2 Ethanol

One of the bacterial products of particular interest to NAFLD pathogenesis is ethanol. Ethanol is

formed during the fermentation of carbohydrates by the IM and absorbed into the portal vein for

delivery to the liver where it is metabolized [171, 172]. Ethanol is a known hepatotoxic agent

which has also been associated with increasing intestinal permeability [173]. Two studies have

looked at blood ethanol levels in NAFLD patients. Volynets et al. found that NAFLD patients

had a significantly higher blood ethanol concentration than healthy controls. Unfortunately this

could not be tied back to the microbiota as IM characterization was not performed [132]. Zhu et

al., however, measured both blood ethanol and IM composition in 22 children with NASH

compared to 25 obese children and 16 healthy controls [11]. They found that blood ethanol

concentrations were higher in NASH compared to healthy controls and obese subjects who did

not differ significantly. When examining IM composition, NASH patients were found to have a

higher abundance of Proteobacteria than the other groups, particularly Enterobacteriaceae which

include Escherichia which are ethanol producers [11]. The role of bacterial ethanol production in

NAFLD development warrants further investigation.

2.2.3.3 Choline metabolism

Several studies have used choline deficient diets to induce NAFLD in test subjects [10, 125,

154]. Choline deficiency is thought to induce NAFLD through the inhibition of VLDL

formation and consequent accumulation of triglycerides within the liver [174], through

mitochondrial dysfunction causing increased levels of reactive oxygen species [175-177] and

through endoplasmic reticulum stress [178]. Choline is converted to trimethylamines by the IM

15

[179]. This could lead to reduced choline absorption from the colon despite adequate dietary

intake. The resulting choline deficiency may promote NAFLD development via the mechanisms

described above.

In a well-controlled inpatient study by Spencer et al. 15 women were placed on a choline

deficient diet and were monitored for steatosis via magnetic resonance imaging (MRI) [10]. IM

composition at baseline was associated with the development of steatosis during choline

deficient diet. Specifically, Gammaproteobacteria abundance at baseline was negatively

correlated with steatosis and Erysipelotrichi abundance at baseline was positively associated with

steatosis development on MRI. This study also found that phosphatidylethanolamine N-

methyltransferase (PEMT) genotype was an effective predictor of steatosis accumulation. The

mechanism behind the relationship between choline, PEMT, and NAFLD pathogenesis is quite

clear. The active form of choline, phosphatidylcholine, which is needed in the liver for VLDL

formation, is lower in NAFLD patients [180]. The formation of this active form is catalyzed by

PEMT; a polymorphism in PEMT has also been associated with NAFLD [181]. The role of the

IM metabolism of choline in this process, however, remains unclear.

2.2.4 NAFLD and the Intestinal Microbiota: Clinical Studies

There have been six human studies that have identified significant differences between the IM in

NAFLD patients and healthy controls. Table 1 summarizes the design and results of these

studies. The first study, discussed above in section 2.2.3.3, found that a particular IM

composition put participants at a greater risk of developing steatosis while on a choline deficient

diet [10]. This study did not, however, look at liver histology and therefore no conclusion can be

made regarding differences between NASH and SS. Additionally, this study was conducted

using healthy women where steatosis was artificially induced therefore results cannot necessarily

be extrapolated to the NAFLD population. A cross-sectional study from our research group

investigated the intestinal microbiota of biopsy confirmed SS and NASH patients compared to

healthy controls [13]. We found that patients with NASH had significantly lower fecal

Bacteroidetes compared to both SS and healthy controls even when controlling for BMI and

dietary intake [13]. This study, however, was conducted using quantitative polymerase chain

reactions (qPCR) rather than pyrosequencing, therefore only selected bacterial groups were

investigated and measurements of bacterial products were not conducted [13]. Raman et al.

16

compared the IM community and fecal volatile organic compounds (VOC) of obese patients with

clinically suspected NAFLD diagnosis via ultrasound and liver enzymes with lean healthy

controls recruited from a colon cancer screening program [12]. They found that patients with

NAFLD had a significantly different IM community than lean controls, including a higher

abundance of Lactobacilli and certain Firmicutes of the Lachnospiraceae family including

Dorea, Robinsoniella, and Roseburia genus. NAFLD patients had a lower relative abundance of

Oscillibacter and higher levels of VOC, which were primarily fatty acid esters [12].

Unfortunately, as liver biopsies were not conducted differences between SS and NASH could not

be evaluated. In addition, dietary intake was not controlled prior to sample collection and not

accounted for in data analysis.

A pediatric study by Zhu et al., discussed in section 2.2.3.2, investigated the IM of children with

biopsy confirmed NASH compared to obese and lean controls with normal liver enzymes [11].

Obese and NASH patients had increased Bacteroidetes and decreased Firmicutes which is

contradictory to the majority of findings between obese and lean adults. Additionally, NASH

patients had elevated blood ethanol levels and a higher proportion of ethanol producing

Enterobacteriaceae compared to obese and lean controls [11]. Due to minimal diagnostic testing,

however, we cannot confirm the liver status of obese controls who may have had SS. Wong et al.

ran a small proof-of-concept trial of the probiotic and prebiotic formula, Lepicol, in adults with

NASH [14]. At baseline NASH patients had a lower abundance of Facecalibacterium and

Anaerosporobacter and increased Parabacteroides and Allisonella when compared to healthy

controls. Probiotic supplementation led to improvements in steatosis which were associated with

a decrease in the abundance of Firmicutes and an increase in Bacteroidetes [14]. Further

probiotic and prebiotic trials are discussed in section 2.2.5. Finally, a recent study discussed in

section 2.2.2.3, found that NAFLD patients (diagnosed by biopsy or imaging) had an increased

abundance of Escherichia, Anaerobacter, Lactobacilli, and Streptococcus compared to healthy

controls which was associated with impaired gut associated immunity and increased

inflammation and permeability of the duodenal mucosa [15]. NASH and SS patients, however,

were not distinguished. The body of literature on the role of the IM in NAFLD pathogenesis

remains limited. Most studies did not use liver biopsies to differentiate NASH from SS which is

important as NASH may progress to cirrhosis and hepatic failure while SS has a benign course.

Additionally, the role of bacterial products in NAFLD has not been well described.

17

Table 1: Summary of human studies on intestinal microbiota and non-alcoholic fatty liver disease

Reference Study Design Study Population Liver Status Methodology IM Methodology Results

Spencer et al. (2011)

Inpatient Intervention

Adult Women n=15

Magnetic Resonance Imaging

454-pyrosequencing Women who started with a lower abundance of Gammaproteobacteria or a higher abundance of Erysipelotrichi were more vulnerable to the development of fatty liver during choline depletion

Mouzaki et al. (2013)

Cross-Sectional

Adults n=11 SS n=22 NASH n=17 Healthy Controls

Liver Biopsy for all groups qPCR NASH patients had lower fecal Bacteroidetes compared to both patients with SS and controls. Differences remained regardless of BMI and fat intake.

Raman et al. (2013)

Observational Case-Control

Adults n=30 Lean Controls n=30 NAFLD

Ultrasound and elevated liver enzymes (>1.5 normal limit) for NAFLD Normal liver enzymes for six months prior to study for controls

Multitag pyrosequencing NAFLD patients had a higher abundance of Lactobacilli and certain Firmicutes of the Lachnospiraceae family (Dorea, Robinsoniella, Roseburia). NAFLD patients had a lower Oscillibacter and higher levels of

fecal VOC, especially fatty acid esters.

Zhu et al. (2013)

Cross-Sectional

Children n=25 Obese n=22 NASH n=16 Lean Controls

Liver Biopsy for NASH Obese and lean controls had normal liver function tests

454-pyrosequencing Obese and NASH patients had increased Bacteroidetes and decreased Firmicutes. Ethanol was higher in NASH vs Obese and Healthy Controls. Proteobacteria was higher in NASH versus Obese and Healthy Controls, primarily Enterobacteriaceae and Escherichia (an ethanol producer)

Wong et al. (2013)

Randomized Controlled Trial

Adults n=16 NASH (9 usual care, 7 probiotic) n=22 Healthy Controls

Liver Biopsy for NASH Normal liver function tests for healthy controls

454-pyrosequencing NASH patients had lower Facecalibacterium and Anaerosporobacter and increased Parabacteroides and Allisonella abundance

Jiang et al. (2015)

Cross-Sectional

Adults n=53 NAFLD n=32 Healthy Controls

Liver Biopsy or imaging for NAFLD No diagnosis of liver disease for healthy controls

Illumina next-generation sequencing

NAFLD patients had higher Escherichia, Anaerobacter, Lactobacilli, Streptococcus, lower CD4+ and CD8+ T-lymphocytes and higher TNF-α, IL-6 in mucosa, microvilli

abnormalities, and wider tight jxn gaps

18

2.2.5 Probiotic and Prebiotic Trials

Given the potential relationship between the IM and NAFLD pathogenesis, several studies have

attempted to prevent or improve NAFLD with the provision of pre- or probiotic supplements. In

animal studies, probiotic treatment led to improved steatosis, insulin resistance, and

inflammation in mice on a high fat diet [182] and in rats on a choline deficient diet [154].

Probiotic supplementation in rats led to decreased hepatic expression of genes involved in

lipogenesis and increased excretion of cholesterol transporters, reducing steatosis while on a high

fat diet [183]. VSL#3, a probiotic used in many trials, reversed insulin resistance, inflammation,

and NASH caused by increased intestinal permeability in mice [184]. Yadev et al. recently

explored the function of VSL#3 in mice, showing that it prevented and treated genetic and diet

induced obesity, reducing weight (fat mass), insulin resistance, steatosis, and inflammatory

markers [155]. The beneficial effects of VSL#3 were associated with increased fecal and plasma

butyrate which triggered increased expression of free fatty acid receptor 3, upregulating the

release of glucagon-like peptide 1 (GLP-1) which is involved in appetite regulation, and

therefore decreasing food intake [155]. VSL#3 also significantly altered the IM, decreasing

Firmicutes and increasing Bacteriodetes and Bifidobacteria. Similarly, supplementary

oligofructose (a prebiotic) in obese (fa/fa) rats led to slower weight gain and lower hepatic

triglyceride concentrations when compared to controls [185].

Results have been equally promising in human studies; however, many studies are not able to

sufficiently explain the mechanism behind the benefits of probiotic supplementation. Two

studies have tested VSL#3 supplementation. Alisi et al. provided children with biopsy proven

NAFLD either VSL#3 or a placebo for 4 months [186]. Children who received the probiotic had

a significantly decreased probability of having severe NAFLD after four months than children

receiving the placebo; 21% had no NAFLD after 4 months. VSL#3 supplementation was also

associated with a higher plasma GLP-1 level and lower body mass index [186]. Loguercio et al.

previously looked at VSL#3 supplementation in patients with NAFLD and other chronic liver

disease, finding lower markers of lipid peroxidation and oxidative stress post-supplementation

[187]. Aller et al. and Vajro et al. ran trials with other probiotic formulations, resulting in

decreased liver enzymes, but no change in inflammatory measures, insulin resistance, or fatty

liver [188, 189]. Trials with synbiotics (pre- and probiotics delivered together) appear more

19

promising, including the small trial discussed in section 2.2.4 [14]. Malaguarnera et al. found

that synbiotic supplementation for 24 weeks resulted in a greater reduction in AST, insulin

resistance, inflammatory markers, endotoxin levels, steatosis, and NASH activity score than diet

and exercise alone [190]. Taken together these studies seem to suggest a beneficial role of

probiotics, particularly VSL#3 or synbiotic combinations, in treating NAFLD. The mechanism of

this improvement is unclear, but likely multifactorial. IM that may be of particular interest in

NAFLD pathogenesis, considering the findings of previous studies, are included in Table 2.

Table 2: Bacteria of interest in non-alcoholic fatty liver disease

Bacteria of Interest Expected Finding Notes on Mechanism

Reference

Bacteroidetes Lower in NASH vs SS & HC Increased energy

absorption

[13]

Proteobacteria Higher in NAFLD

[11, 12]

Erysipelotrichi Higher abundance increased

susceptibility to NAFLD

[10]

Gammaproteobacteria Mixed results with lower or higher

abundance in NAFLD

[10, 12]

Enterobacteriaceae,

especially Escherichia

Higher in NAFLD/NASH vs HC Ethanol producer

[11, 15]

Ruminococcaceae Higher in HC Butyrate producer

[15]

Faecalibacterium Lower in NASH vs HC Butyrate producer

[14]

Lactobacilli Higher in NAFLD (SS &NASH)

[45, 78]

Prevotella Higher in HC Butyrate producer

[15]

Streptococcus Higher in NAFLD vs HC

[15]

Clostridium Clusters

XIVa and IV

Lower in NASH vs HC & SS Butyrate producers Not

extensively

studied in

NAFLD

population

20

2.2.6 Proposed Mechanism

Given the background information provided, the intestinal microbiota may contribute to NAFLD

pathogenesis via the following mechanisms:

• Increased energy harvest by IM may increase total energy absorbed, contributing to obesity, a

known risk factor for NAFLD

• Metabolism of choline by IM may promote steatosis by decreasing lipid excretion from the

liver as VLDL

• Decreased production of butyrate by IM may impair the intestinal epithelium and increase

intestinal permeability

• Bacterial endotoxin may cross the permeable intestinal mucosa causing the activation of TLR-

4, triggering systemic inflammation and insulin resistance

• Alterations in IM composition may lead to increased production of ethanol while increased

permeability may lead to increased absorption of ethanol entering the blood stream and causing

increased metabolic stress and known risk to liver health.

21

3 Research Aim and Hypotheses

Aim: The aim of this research project is to compare bacterial products between biopsy confirmed

SS, NASH and healthy controls (HC) as well as to further assess the IM between groups.

Main Hypothesis: Patients with NASH have higher plasma endotoxin compared to those with

SS and HC.

Secondary Hypothesis: Patients with NASH have lower fecal butyrate compared to SS and HC.

Additional Outcomes: We will also measure whether there is a difference in serum choline,

ethanol and other serum and fecal metabolites between the three groups and assess if there is any

association with differences in IM and diet.

22

4 Materials and Methods

4.1 Study Design

This is a cross-sectional study comparing patients with liver biopsy proven simple steatosis (SS),

non-alcoholic steatohepatitis (NASH) and healthy controls (HC).

Adult subjects seen by hepatologists of the University Health Network, Toronto, were

approached for this study. Patients with a clinical suspicion of NAFLD, typically due to

persistently elevated liver enzymes and obesity, were assessed by a hepatologist using standard

medical practice. Patients were given general advice to lose weight and increase physical

activity; no specific instructions for lifestyle change were provided. Patients who returned for

their follow-up appointment approximately six months later without success in lifestyle

improvement or weight loss were booked for a liver biopsy to assess the severity of NAFLD.

During this visit (study visit 1) the study was explained in detail and informed consent was

obtained from interested participants. See Appendix 1 for consent forms. Following consent,

subjects were provided with detailed instructions for the completion of a 7-day food record, 7-

day activity record, and environmental questionnaire (see below for further details). Patients

were also provided with instructions for the collection and transport of their stool sample

(Appendix 2), which they were asked to return along with their food/activity record on the day

of their liver biopsy (study visit 2). On study visit 2, subjects underwent anthropometric

measurements and provided a fasting blood sample for clinical (section 4.4) and study specific

(section 4.6 and 4.7) bloodwork.

For healthy controls, individuals undergoing candidacy assessment as potential healthy living

liver donors at Toronto General Hospital were approached during their first screening

appointment. The study details were explained and informed consent was provided by interested

participants. At this visit (study visit 1) the same instructions for the food/activity records,

environmental questionnaire, and stool sample collection were provided. Approximately one

week prior to liver donation, healthy controls returned for study visit 2 where they underwent

anthropometric measurements, provided a fasting blood sample, and returned their stool sample

and food/activity record. On study visit 3, during their donation surgery, a wedge liver biopsy

was collected for histological analysis.

23

Inclusion criteria for this study were age greater than or equal to18 years and confirmation of

NAFLD or healthy liver status via liver biopsy. Exclusion criteria were diagnosis of liver disease

other than NAFLD on liver biopsy, liver transplant expected to be required within one year,

significant liver complications (e.g. variceal bleeding, jaundice, etc.) or any other

contraindications for liver biopsy, alcohol intake greater than 20g per day, pregnancy or

lactation, presence of gastrointestinal disease, use of medications known to cause steatohepatitis,

insulin, NSAIDS (other than low dose acetylsalicylic acid), antibiotics, prebiotics, probiotics, or

experimental drugs within the last three months.

4.2 Clinical Data, Environmental Questionnaire, and Anthropometric Measurements

Clinical data were collected on study visit one. Study participant’s smoking and alcohol

consumption history and medication and supplement use were reviewed, including medications

taken in the last three months. Study participants were also asked to answer a number of

questions regarding their personal and family history of disease. Age, ethnicity, and menstrual

history were also recorded.

An environmental questionnaire (Appendix 3) was completed and returned with the stool

sample. This questionnaire collected information that may affect an individual’s IM composition

and included questions such as country of origin, method of birth (vaginal versus caesarian

section), breastfeeding, and pets at home.

Anthropometric measurements including height (ht), weight (wt), and waist circumference (WC)

were measured by a trained research professional. Weight was measured using a calibrated

hospital-grade chair scale; height was measured using a stadiometer. Waist circumference was

measured at the umbilicus level. All measurements were taken in triplicate and the average value

was used.

4.3 Nutrition and Activity Assessment

Each participant was given a food record and activity log (Appendix 4) to complete in the week

prior to returning their stool sample. Detailed instructions were provided for the completion of

both of these tools. The food log included all food and beverages consumed over each 24 hours

for seven days. A three-day food record including one weekend day was also accepted.

24

Participants used the 2D Food Portion Visual Chart (Nutrition Consulting Enterprises,

Framingham, MA) to estimate portion sizes. This is a validated tool which has been used in our

previous studies [60, 191, 192]. Food records were reviewed by an experienced registered

dietitian and were analyzed using Food Processor Diet and Nutrition Analysis Software (Version

7, ESHA Research, Salem, OR).

Physical activity logs were recorded for 7 days concurrent with the food records. Participants

were asked to record any activity, including household chores, the duration of the activity, and

the intensity level. Detailed instructions were provided including examples for each intensity

level (mild, moderate, strenuous, and very strenuous). For details see Appendix 4. This

information was used to calculate daily physical activity units: 1 unit = 30 minutes mild, 20

minutes moderate, 10 minutes strenuous, or 5 minutes very strenuous activity. This is a validated

method for measuring physical activity level [193]. Basal metabolic rate (BMR) was calculated

using the Harris-Benedict equation: BMR for men = 66.5 + [13.75 × wt(kg)] + [5.003 × ht(cm)]

– [6.755 × age(y)], BMR for women = 655.1 [9.563 × wt(kg)] + [1.850 × ht(cm)] – [4.676 ×

age(y)]. Estimated energy requirement (EER) was calculated using Health Canada Guidelines:

EER for men = 662 – [9.53 x age(y)] + PA x {[15.91 x wt(kg])] + [539.6 x ht(m)]}, and EER for

women = 354 – [6.91 x age(y)] + PA x {[9.36 x wt(kg)] + (726 x ht(m)]} where PA is the

physical activity coefficients which are included in Table 3 [194].

25

Table 3: Physical activity coefficients for estimated energy expenditure calculation [194]

Activity Level Sedentary Low Active Active Very Active

Description Typical daily

living activities

(e.g., household

tasks, walking

to the bus)

Typical daily

living activities

PLUS 30 - 60

minutes of

daily moderate

activity (ex.

walking at 5-7

km/h)

Typical daily

living activities

PLUS At least

60 minutes of

daily moderate

activity

Typical daily

living activities

PLUS At least

60 minutes of

daily moderate

activity PLUS

An additional

60 minutes of

vigorous

activity or 120

minutes of

moderate

activity

Men 1.00 1.11 1.25 1.48

Women 1.00 1.12 1.27 1.45

4.4 Clinical Biochemistry

Blood was drawn after a 12 hour overnight fast on study visit 2. Liver markers were drawn as a

routine clinical measure and included: aspartate transaminase (AST), alanine transaminase

(ALT), alkaline phosphatase (ALP), and bilirubin. Measures of glucose metabolism included

plasma glucose, insulin, hemoglobin A1c (HbA1c), and homeostasis model assessment estimated

insulin resistance (HOMA-IR) which was calculated as fasting glucose (mmol/L) × fasting

insulin (mU/L)/22.5 [195]. A lipid profile was also conducted, including total cholesterol, low

density lipoprotein (LDL), high density lipoprotein (HDL), and triglycerides (TG). These

analyses were conducted by the UHN Laboratory Medicine Program. Liver enzymes and lipid

profile were measured using the Architect c8000 system (Abbott Laboratories). LDL was

calculated from total cholesterol – HDL. Fasting plasma glucose and plasma insulin were

measured by the enzymatic hexokinase method and radioimmunoassay, respectively.

26

4.5 Liver Histology

Liver biopsies were taken percutaneously (needle biopsy) for NAFLD patients and

intraoperatively (wedge biopsy) for HC and preserved immediately in formalin. Liver biopsies

were assessed by the same pathologist using standard stains for the diagnosis of NAFLD,

morphologic evaluation, and to rule out any iron overload. The evaluation of NAFLD related

measures of steatosis, inflammation, and fibrosis were conducted using the validated and

reproducible Brunt system (Table 4) [196]. Disease severity was also evaluated using the NAS

which accounts for degree of steatosis, lobular inflammation, and hepatocellular ballooning for a

final score of 0-8 [197].

27

Table 4: Assessment of liver histology: Brunt System [196]

Steatosis % hepatocytes involved

Grade 0 <5%

Grade 1 5-33%

Grade 2 >33-66%

Grade 3 >66%

NASH (Inflammation + Steatosis) Description

Grade 1: Mild

Steatosis Predominantly macrovesicular, involves < 33% -

66% of the lobules

Ballooning Occasionally observed; zone 3 hepatocytes

Lobular Inflammation Scattered and mild acute (polymorphs) inflammation

and occasional chronic inflammation (mononuclear

cells)

Portal Inflammation None or mild

Grade 2: Moderate

Steatosis Any degree and usually mixed macrovesicular and

microvesicular

Ballooning Obvious and present in zone 3

Lobular Inflammation Polymorphs may be noted associated with ballooned

hepatocytes, pericellular fibrosis; mild chronic

inflammation may be seen

Portal Inflammation Mild to moderate

Grade 3: Severe

Steatosis Typically > 66% (panacinar); commonly mixed

steatosis

Ballooning Predominantly zone 3; marked

Lobular Inflammation Scattered acute and chronic inflammation;

polymorphs may appear concentrated in zone 3 areas

of ballooning and perisinusoidal fibrosis

Portal Inflammation Mild or moderate

Fibrosis

Stage 1 Zone 3 perivenular perisinusoidal/pericellular

fibrosis, focal or extensive

Stage 2 As above with focal or extensive periportal fibrosis

Stage 3 Bridging fibrosis, focal or extensive

Stage 4 Cirrhosis

28

4.6 Plasma Endotoxin

Blood was drawn in a fasting state using an ethylenediaminetetraacetic acid (EDTA) coated

vacutainer and separated by centrifugation at 2400 rpm for 10 minutes at 20oC. Plasma was then

stored at -80°C until the time of measurement. Endotoxin quantification was conducted using the

QCL-1000 chromogenic limulus amebocyte lysate (LAL) endpoint assay (Lonza, Walkersville,

MD) in our own laboratory. This assay has been effectively used in the literature [130, 198-201].

The assay is based on the principle that endotoxin catalyzes the activation of a proenzyme that is

found in LAL. The rate of activation is determined by the amount of endotoxin present. Once

activated, the enzyme catalyzes the splitting of pNA from the colourless substrate. Once the

reaction is stopped, the separated pNA can be measured via photometry at 405-410nm. The

correlation between absorbance and endotoxin concentration is linear from 0.1 to 1.0 EU/mL.

Endotoxin concentration is determined using a standard curve.

The LAL assay was optimized for use with our samples prior to study measurements. The ideal

dilution and processing of samples was determined using a positive product control or “spiked”

sample. Each sample dilution was spiked with 0.4 EU/mL. The spiked and unspiked sample

dilution were then measured as per the study protocol (see below). Optimization occurred when

spike recovery was in the 75-125% range, indicating that significant inhibition or enhancement

has not occurred.

Following this process the optimal LAL assay protocol was developed. Plasma samples were

thawed at room temperature. An initial 1:5 dilution was heat treated at 70°C for 15 minutes to

deactivate interference from plasma proteins. Samples were then cooled. Meanwhile a standard

curve was produced, using the provided endotoxin standard and endotoxin free water. The

standard curve including five points: 0.4 EU/mL, 0.3 EU/mL, 0.2 EU/mL, 0.1 EU/mL, and 0.05

EU/mL.

Once the 1:5 plasma dilution was cooled, a 1:60 dilution was produced by adding 0.1 mL of the

1:5 solution to 1.1 mL of 10mM MgCl2. MgCl2 was used to overcome the inhibitory effect of

EDTA. An endotoxin-free 96 well plate was filled with 50 µL of each 1:60 sample dilution, the

standard curve, and a water blank in triplicate. A sample blank was also plated in duplicate to

control of any absorbance due to colour present in the sample prior to assay. The plate was then

placed on a block heater and preheated to 37°C. Meanwhile the LAL reagent and substrate were

29

reconstituted with endotoxin-free water and the substrate was heated to 37°C in a water bath. All

pipetting was done using endotoxin-free pipette tips and dilutions were made in endotoxin-free

test tubes.

The timed test protocol then occurred as follows: at T=0, 50 µL of LAL reagent was added to

each standard, blank, and test well using a repeater pipette. The plate was then tapped to mix and

left to incubate for 10 minutes. At T=10 minutes, 100 µL of substrate (heated to 37°C) was

added to each of these wells and the plate was tapped to mix. At T=16 minutes, 50 µL of a stop

solution (10g/100mL sodium dodecylsulfate) was added to all wells, including the sample blanks

and the plate was tapped to mix. Any bubbles present in the wells were then popped using a

sterile syringe. Absorbance was measured at 405nm using the PHERAstar FS plate reader (BMG

Labtech, Offenburg, Germany) and endotoxin concentrations were extrapolated from the

standard curve using the MARS Data Analysis software (Version 3.01 R2, BMG Labtech, 2013).

4.7 Serum Metabolites

Serum metabolites, including choline, ethanol, and TMAO, were measured to evaluate potential

implications of bacterial metabolism at a systemic level. For the list of metabolites measured see

Table 5. Serum was drawn in a fasting state using a gel serum separation vacutainer and was

immediately placed in an insulated container with cooling elements. Blood was separated by

centrifuge at 4°C at 2800 x g for 20 minutes. Serum was then aliquoted and stored at -80°C until

all study samples were collected. Serum samples were shipped to the Metabolomic Innovation

Centre (Edmonton, AB) where metabolites were analyzed using nuclear magnetic resonance

(NMR) spectrometry [202]. The following method was used by the centre [202]: serum samples

were deproteinized using ultrafiltration. Filtrates were checked visually for a red tint, which

indicates that the membrane was compromised. For those “membrane compromised” samples,

the filtration process was repeated with a different filter and filtrates were inspected again. Then,

each filtrate (300 μL) was combined with 35 μL of D2O and 15 μL of a standard buffer solution.

The serum sample (350 μL) was then transferred to a standard Shigemi microcell NMR tube for

subsequent spectral analysis.

All 1H-NMR spectra were collected on a 500 MHz Inova (Varian Inc., Palo Alto, CA)

spectrometer.1H-NMR spectra were acquired at 25°C using the first transient of the tnnoesy-

presaturation pulse sequence, which was chosen for its high degree of quantitative accuracy

30

[203]. Spectra were collected with 128 transients and 8 steady-state scans using a 4 second

acquisition time and a 1 second recycle delay.

All free induction decay NMR signals were zero-filled to 64k data points and subjected to line

broadening of 0.5 Hz. The singlet produced by a known quantity of DSS methyl groups was used

as an internal standard (set to 0 ppm). All 1H-NMR spectra were processed and analyzed using

the Chenomx NMR Suite Professional software package version 6.0 (Chenomx Inc., Edmonton,

AB), as previously described [204]. Each spectrum was processed and analyzed by at least two

experienced NMR spectroscopists to minimize compound mis-identification and mis-

quantification.

Serum trimethylamine N-oxide (TMAO) was not detectable using NMR therefore TMAO was

measured using a targeted quantitative metabolomics approach by Liquid Chromatography Mass

Spectrometry (LCMS). Isotopically-labeled internal standards were added to the serum to

facilitate metabolite quantification. Sample extraction was performed on a 96 well plate with a

0.2 µm solvent filter. 10 µL of serum was spiked with the internal standard (TMAO D9) and then

150 µL of methanol with 10 mM ammonium acetate was added for extraction. Each sample was

diluted with 150 μL of water. Seven calibrant solutions with known concentrations went through

the same extraction steps. LCMS analysis was performed on AB SCIEX 4000 QTrap mass

spectrometer with Agilent 1100 HPLC. 10 µL of the extracted samples were injected onto the

Kinetex C18 (2.6 µm, 3.0x100mm, 100A) Column with guard column. Isobaric elution was

performed with 90% A (10 mM ammonium formate in water, PH3) and 10% B (10 mM

ammonium formate in 90:10 acetonitrile:water, PH3). Total LC method run time was 3 min with

flow rate of 500 µl/min. A seven-point calibration curve was generated to quantify the

concentration of TMAO in samples.

31

Table 5: Metabolites measured in stool and serum samples

Stool Metabolites Serum Metabolites

Acetic Acid* Acetic Acid*

Butyric Acid Choline*

Choline* Ethanol

Formic Acid* Formic Acid*

Isobutyric Acid 2-Hydroxybutyrate

Isovalerate L-Lactic Acid*

L-Lactic Acid* Trimethylamine N-Oxide*

Propionate

Succinate

Trimethylamine*

* Metabolite measured in both stool and serum

4.8 Stool Sample Collection and Analysis

Stool collection kits included a plastic collection/storage container with a tightly closing lid, an

insulated bag, and cooling elements. Within 24 hours of study visit 2, subjects collected one stool

sample, which was frozen immediately after defecation in their home freezer (-20°C). Subjects

brought the frozen sample in the insulated bag with cooling elements to the hospital, where it

was stored at -80C until homogenization. This method of stool collection was used in a

landmark study to establish a human gut microbial gene catalogue [205].

4.8.1 Stool Homogenization

Stool samples were homogenized prior to DNA extraction and metabolite measurements. The

entire sample was first transferred into a sterile masticator bag. The sample was allowed to thaw

until a smooth consistency was reached, typically 2-3 hours depending on sample size. Once

32

thawed, excess air was released from the bag and the sample was homogenized for two minutes

using a masticator blender (IUL, S.A., Barcelona, Spain). This was followed by one minute of

hand mastication of any areas that were missed. The corner of the masticator bag was then cut

and the sample was aliquoted: 1-2 g samples were stored for metabolite analysis and 0.1-0.2 g

aliquots were stored for DNA extraction. Samples were immediately placed on dry ice and then

transferred to -80°C for storage until analysis. Weight was recorded for each aliquot and pH was

measured for each sample.

4.8.2 Fecal Metabolite Analysis

Homogenized fecal samples were transferred to the Metabolomic Innovation Centre on dry ice

and were analyzed by NMR spectroscopy to identify 10 metabolites of interest (Table 5). Fecal

samples were prepared by mixing 20 mg of frozen fecal material with 1 mL of saline phosphate

buffer in deuterium oxide, followed by centrifugation (18 000 × g, 1 minute). Fecal supernatants

were removed and filtered through 0.2 μm membrane filters as described by Le Gall and

colleagues [206]. The measurement in fecal supernatant was then conducted as described above

in section 4.7 for serum metabolites.

4.8.3 DNA Extraction

DNA was extracted using the E.Z.N.A. Stool DNA Kit (Omega Bio-Tek, Norcross, GA) and a

modified manufacturer’s protocol with the addition of a lysozyme and proteinase digestion step

(incubation at 37°C for 30 minutes) as used by Mouzaki et al. [13]. An elution volume of 100 µL

was used. DNA was analyzed for purity and concentration using the Nanodrop 1000

Spectrophotometer (ThermoScientific, Rockford, IL). DNA samples were stored at -80°C.

4.8.4 Quantitative Real-time PCR

Total bacteria and selected microbial groups of interest: Bacteroidetes, Bifidobacteria,

Clostridium leptum, Clostridium coccoides, Lactobacilli, E. coli, and Archaea were analyzed by

qPCR. Assays were run in triplicate in a 384 well optical plate using a 7900HT thermocycler

(Applied Biosystems, Foster City, CA) under default thermocycling conditions. Fifty nanograms

of DNA with TaqMan Gene Expression Master Mix (MM) and specific TaqMan primers

(Applied Biosystems) were used. Custom-made TaqMan primers for total bacteria, Lactobacilli,

C. coccoides, C. leptum, Bacteroidetes, Bifidobacteria, and Archaea were used [207-209]. Real-

33

time PCR for E. coli was done using SYBR Green Gene Expression MM (Applied Biosystems)

and the specific forward and reverse primer [207]. The ramping profile for SYBR Green

amplifications was similar and a dissociation stage was added in the end. The number of cells of

each microorganism in fecal samples was calculated by interpolation from pre-constructed

standard curves (Table 6) and expressed as CFU/g of wet feces.

Table 6: Standard curves used in the interpretation of the qPCR results

Microorganism Standard curve Efficiency (%)

Total bacteria y=-3.325x + 37.670 99.87

Bacteroidetes y=-3.289x + 33.635 101.39

C. leptum y=-3.315x + 38.932 100.29

C. coccoides y=-3.3236x + 37.013 99.93

Lactobacilli y=-3.371x + 37.660 97.99

Bifidobacteria y=-3.357x + 37.826 98.56

E. coli y=-3.2928x + 34.939 101.23

Archaea y=-3.351x + 39.680 98.80

4.9 Statistical Analysis

4.9.1 Data Analysis

Descriptive summaries of all measurements were calculated: means and standard deviations (SD)

for normal variables, medians and first quartile and third quartile for skewed variables, and

proportions for categorical variables. Plots were used as appropriate to examine the data.

Bivariate relationships were assessed using Pearson’s or Spearman’s correlation coefficients,

Chi-square and Fisher exact tests where appropriate. Comparisons across diagnosis groups were

performed for normally distributed data using one-way ANOVA followed by the t-test (if

necessary, data were log-transformed to normalize distribution), or for non-normally distributed

data using the Kruskal-Wallis test followed by Wilcoxon rank-sums test. All tests were two-

sided and performed at the 5% significance (alpha) level. Bonferroni’s correction method was

34

used to account for multiple comparisons. Outliers were identified by visual inspection and

sensitivity analyses were run. If the exclusion of outliers altered the result of the statistical test

this was noted in the results section. All statistical analyses were conducted using SAS version

9.4 (SAS Institute Inc., Cary, NC).

4.9.2 Power Calculations

Calculations were performed using standard statistical software packages, primarily SAS version

9.3 (SAS Institute Inc., Cary, NC). Calculations for the main variable (endotoxin) was based on a

sample size of n=30 per group and estimates from Harte et al. obtained from a similar patient

population of HC, SS, and NASH [130]. SD and effect size (ES), where ES=mean difference,

were estimated from Harte et al. (SD=5.22, ES=3.84 EU/mL). The significance level was set at

5%. Power calculations were performed for one-way ANOVA. We estimated the power to detect

differences between our three groups in ANOVA as >0.999.

35

5 Results

5.1 Subjects

A total of 30 HC, 15 SS, and 24 NASH patients were found eligible and provided consent to

participate in this study. Demographics, health history, and anthropometric data for these

subjects are described in Table 7. Both SS and NASH groups were significantly older than HC,

but did not differ by sex. BMI in NASH patients was significantly higher than HC, but BMI did

not differ between HC and SS or SS and NASH groups. Waist circumference also followed this

pattern. There appeared to be a difference in diabetes prevalence between groups, but when

Bonferroni correction was used for multiple comparisons this significance was lost. Prevalence

of hypertension was greater in both patient groups than in HC and corresponded with a higher

use of cardiovascular drugs which included antihypertensive medications. The use of lipid

lowering drugs such as statins was higher in NASH patients and the use of mild non-steroidal

anti-inflammatory drugs (low dose acetylsalicylic acid only) was higher in SS than in HC.

Biochemistry data is described in Table 8. ALT was significantly higher in SS patients versus

HC and in NASH patients versus both SS and HC. AST was significantly higher in NASH

patients than in both HC and SS patients. Fasting glucose appeared to be higher in NASH

patients but this did not remain significant in post-hoc pairwise comparisons. Fasting insulin was

significantly higher in NASH patients than in HC and SS patients, however, HOMA-IR and

HbA1c where only significantly different between HC and NASH patients with NASH patients

having higher levels. Lipids did not differ significantly between the groups with the exception of

triglycerides which were significantly higher in NASH patients versus HC.

Liver biopsies were available for 17 HC, 15 SS, and 23 NASH patients. Healthy controls who

did not have a liver biopsy had liver health confirmed via several diagnostic tests including blood

measurements of liver enzymes (ALT, AST, ALP), computerized tomography and MRI scans

which were reviewed by a panel of surgeons and hepatologists and confirmed to be healthy and

without fat deposits. One NASH patient was diagnosed via liver biopsy at an outside facility and

therefore histological analysis by our study pathologist was not possible.

Histological analysis is displayed in Table 9. Occasionally biopsies were not of sufficient size or

quality for all analyses and the total inflammation score was not measured consistently for all

36

patients. The sample size available for each histological feature is therefore noted. NASH

patients appear to have relatively mild disease with a median NAS of 5 out of a possible 8.

Environmental questionnaires were only received from 53% of study participants and were often

incomplete therefore statistical analysis was limited. Patient’s country of birth was also collected

as part of demography in 22 HC, 9 SS, and 16 NASH patients. There was a significant difference

in the proportion of patients born in Canada between groups with more HC (86%) being born in

Canada than SS (0%) and NASH (31%) patients (p<0.0001). Ethnicity, however, did not differ

by group as shown in Table 7.

37

Table 7: Subject demographics and health history

Variable n HC n SS n NASH p-value

Age (years) 30 37 (10)ab 15 47 (8)a 24 48 (9)b 0.0001

Sex (% female) 30 50% (15) 15 40% (6) 24 54 (13) 0.7

Ethnicity (%

Caucasian)

19 84% (16) 9 44% (4) 12 58% (7) 0.09

BMI (kg/m2) 30 26.2 (23.6,

28.8)a

15 27.4 (24.9,

32.5)

24 32.1 (29.7,

34.3)a

<0.0001

Waist Circumference

(cm)

28 89.9 (84.4,

94.8)a

13 97.1 (89.6,

106)

22 103 (97.8,

109)a

0.0002

Diabetes 30 0% (0) 15 7% (1) 24 17% (4) 0.049

Cardiovascular

Disease

30 0% (0) 15 7% (1) 24 7% (1) 0.2

Hypertension 30 0% (0)ab 15 27% (4)a 24 38% (9)b 0.0003

Renal Disease 30 0% (0) 15 0% (0) 24 8% (2) 0.16

Cardiovascular Drugs 30 0% (0)ab 15 33% (5)a 24 25% (6)b 0.001

Diabetes Drugs 30 0% (0) 15 7% (1) 24 13% (4) 0.049

Lipid Lowering Drugs 30 3% (1)a 15 27% (4) 24 33% (8)a 0.008

Anti-Depressants 30 7% (2) 15 7% (1) 24 0% (0) 0.4

NSAID† 30 0% (0)a 15 27% (4)a 24 13% (3) 0.011

Continuous variables are expressed as mean (standard deviation) for normal variables and

median (1st quartile, 3

rd quartile) for non-normally distributed variables.

Categorical variables are expressed as prevalence in percentage (count).

Values with the same superscript are significantly different.

† NSAID = nonsteroidal anti-inflammatory drug

38

Table 8: Biochemistry by disease group

Variable n HC n SS n NASH p-value

ALT (U/L) 30 18 (14, 29)ab

15 45 (32, 54)ac

24 70.5 (45,

104)bc

<0.0001

AST (U/L) 30 20 (17, 25)a 15 26 (21, 33)

b 24 45 (31, 67)

ab <0.0001

ALP (U/L) 30 71 (21) 15 70 (18) 24 72 (19) 0.9

Bilirubin (µmol/L) 28 10 (7, 13) 14 8 (7, 13) 21 10 (8, 11) 0.9

Glucose (mmol/L) 29 4.9 (4.6, 5.1) 12 5.1 (4.7, 5.7) 23 5.7 (4.8, 6.2) 0.035†

Insulin (pmol/L) 24 36 (22, 56)a 12 31 (23, 82)

b 22 102 (56,

191)ab

0.0001

HOMA-IR‡ 24 1.18 (0.80,

1.93)a

11 1.33 (0.77,

4.65)

22 4.26 (2.03,

8.63)a

0.0001

HbA1c 29 0.052 (0.049,

0.054)a

12 0.056 (0.053,

0.059)

13 0.058 (0.053,

0.065)a

0.001

Total Cholesterol

(mmol/L)

23 4.97 (1.00) 13 5.28 (1.07) 24 4.81 (1.55) 0.5

LDL (mmol/L) 23 3.16 (2.67,

3.53)

12 3.37 (2.65,

4.27)

23 2.67 (2.09,

3.17)

0.06

HDL (mmol/L) 23 1.30 (0.33) 12 1.26 (0.24) 24 1.10 (0.29) 0.07

TG (mmol/L) 23 0.94 (0.77,

1.23)a

13 1.10 (0.86,

1.37)

24 1.63 (1.17,

2.62)a

0.002

Values are expressed as mean (standard deviation) for normal variables and median (1st quartile,

3rd

quartile) for non-normally distributed variables.

Values with the same superscript are significantly different.

† Despite significant comparison with Krusal-Wallis test, pairwise comparisons did not reach

significance.

‡ HOMA-IR = fasting glucose (mmol/L) × fasting insulin (mU/L)/22.5.

39

Table 9: Liver histology by disease group

Steatosis (% hepatocytes) Median (min, max)

HC (n=17) SS (n=15) NASH (n=23)

0 (0, 2.5) 15 (5, 70) 40 (10, 90)

Fibrosis Stage Proportion of subjects (count)

HC (n=14) SS (n=15) NASH (n=23)

0 79% (11) 67% (10) 13% (3)

1 21% (3) 20% (3) 26% (6)

2 0% (0) 13% (2) 35% (8)

3 0% (0) 0% (0) 13% (3)

4 0% (0) 0% (0) 13% (3)

Lobular Inflammation Intensity Proportion of subjects (count)

HC (n=15) SS (n=15) NASH (n=23)

0 100% (15) 87% (13) 0% (0)

1 0% (0) 13% (2) 69.5% (16)

2 0% (0) 0% (0) 22% (5)

3 0% (0) 0% (0) 8.5% (2)

Total Inflammation†

Proportion of subjects (count) HC (n=11) SS (n=5) NASH (n=11)

None 100% (11) 60% (3) 0% (0)

Mild 0% (0) 40% (2) 27% (3)

Moderate 0% (0) 0% (0) 73% (8)

Severe 0% (0) 0% (0) 0% (0)

Ballooning Intensity

Proportion of subjects (count) HC (n=16) SS (n=15) NASH (n=23)

None 100% (16) 100% (15) 4% (1)

Mild 0% (0) 0% (0) 61% (14)

Obvious 0% (0) 0% (0) 35% (8)

NAFLD Activity Score

Median (min, max) HC (n=15) SS (n=15) NASH (n=23)

0-8 0 (0, 0) 1 (1, 4) 5 (3, 8)

† This measure was not consistently recorded in all patients.

40

5.2 Dietary Intake and Physical Activity

Food records were completed for at least three days by 25 HC, 10 SS, and 20 NASH patients.

Analysis of energy and macronutrients is detailed in Table 10. Reported energy intake differed

by group with NASH patients reporting significantly fewer kilocalories than HC. To investigate

for possible underreporting, reported energy intake was compared with EER and BMR. Reported

proportion of EER consumed did not differ significantly between groups (p=0.14) (Figure 1).

Reported proportion of BMR consumed, however, was significantly lower in NASH patients

than HC (p=0.011) with the median reported proportion consumed at less than 100% for NASH

patients (Figure 2). As BMR is the estimated energy required for basic physiological functions

and none of these patients was losing weight we suspected that underreporting of energy intake

may have occurred, particularly in the NASH patients. For this reason macro and micronutrient

intake was expressed as a proportion of energy consumed to describe dietary composition.

Protein derived energy did differ slightly between groups but did not remain significant in

pairwise comparisons after Bonferroni correction.

Reported micronutrient intake is displayed in Table 11. Micronutrient intake did not differ

between groups. Data regarding omega 6:3 ratio and choline intake may not be reliable due to

incomplete data in the Food Processor database, but it is important to note that relative intake did

not differ between the three groups. The proportion of all subjects with acceptable intakes for

each nutrient (where requirements are available) are also noted in Table 10 and Table 11. These

proportions were also calculated and compared by disease group but did not differ significantly

(Fisher p>0.05). Notably, reported dietary intake was quite poor for many nutrients with less

than 50% of subjects having acceptable intakes for saturated fat, fibre, calcium, and sodium.

Physical activity records were completed by 24 HC, 11 SS, and 19 NASH patients. Total daily

activity (physical activity units per day) did not differ between groups (p=0.8) (Figure 3). As

strenuous physical activity may have a more significant effect on health as well as the IM, the

combined time spent participating in strenuous and very strenuous activity was also compared

between groups (Figure 4). Although there is a trend for higher strenuous activity in HC

compared to other subjects this did not reach significance (p=0.1).

41

Table 10: Reported dietary intake and estimated requirements: Macronutrients

Variable

HC (n=25) SS (n=10) NASH

(n=20) p-value

Requirement

(% total subjects

with acceptable

intake)*

Energy Intake

(kcal/kg/day) 27 (23, 33)a

24 (20, 30) 18 (13, 21)a

0.001

25-35 kcal/kg (22%) Energy Intake

(kcal/day) 2122 (569)a

1798 (525) 1639 (612)a

0.024

EER (kcal/day) 2582 (2141,

3316)

2399 (2185,

2639)

2565 (2117,

3052) 0.6 N/A

BMR (kcal/day) 1544 (1436,

1760)

1655 (1316,

1834)

1705 (1469,

1953) 0.4 N/A

Fat-derived Energy (%) 35 (7) 29 (7) 32 (10) 0.17

20-35% kcal

(53%)

Protein-derived Energy

(%) 17 (15, 19) 21 (18, 22) 19 (17, 21) 0.031†

10-35% kcal

(98%)

Carbohydrate-derived

Energy (%) 48 (44, 54) 51 (44, 56) 48 (39, 51) 0.5 45-35% kcal

(58%)

Saturated Fat-derived

Energy (%) 11 (10, 13) 8 (7, 11) 11 (10, 12) 0.15 <10% kcal

(40%)

Cholesterol (mg/day) 270 (174,

336)

239 (119,

354)

207 (146,

356) <300 mg

(64%) Cholesterol (mg/1000

kcal) 129 (88,

156)

134 (99,

155)

143 (99,

208) 0.7

Fibre (g/day) 24 (19, 27) 18 (14, 27) 17 (13, 21)

21-38 g

(22%) Fibre (g/1000 kcal) 11 (9, 13) 10 (8, 15) 10 (8, 12) 0.5

Sugar-derived Energy

(%) 19 (8) 15 (6) 16 (8) 0.17 N/A

Omega 6:3 6 (5, 8) 6 (4, 8) 7 (5, 8) 0.9 N/A

Values are expressed as mean (standard deviation) for normal variables and median (1st quartile, 3rd

quartile) for non-normally distributed variables.

Values with the same superscript are significantly different.

* Intake was compared to the Dietary Reference Intakes [210] and American Heart Association guidelines

[211].

† Despite significant comparison with Krusal-Wallis test, pairwise comparisons did not reach

significance.

42

Figure 1: Reported proportion of estimated energy requirement consumed

There was no statistically significant difference in proportion of estimated energy requirement (EER)

consumed between healthy control (HC), simple steatosis (SS), and non-alcoholic steatohepatis (NASH)

groups (HC: n=25; SS: n=10; NASH: n=20).

Figure 2: Reported proportion of basal metabolic rate consumed

There was no statistically significant difference in proportion of basal metabolic rate (BMR) consumed

between healthy control (HC), simple steatosis (SS), and non-alcoholic steatohepatis (NASH) groups

(HC: n=25; SS: n=10; NASH: n=20).

43

Table 11: Reported dietary intake and estimated requirements: Micronutrients

Variable

HC (n=25) SS (n=10) NASH

(n=20) p-value

Requirement

(% total subjects

with acceptable

intake)*

Vitamin A (IU/day) 11117 (5478,

16688)

6785 (5039,

12014)

8514 (6711,

13635) 2333-3000 IU

(89%) Vitamin A (IU/1000

kcal)

4892 (2687,

8683)

4935 (2921,

7409)

6465 (2418,

12198) 0.5

Vitamin C (mg/day) 161 (91, 227)

108 (61,

132)

128 (69,

218) 75-90mg

(71%) Vitamin C

(mg/1000 kcal) 79 (52, 112) 62 (42, 82) 76 (41, 177) 0.6

Choline (mg/day) 234 (183,

284)

212 (165,

303)

189 (119,

258) 425-550 mg

(2%) Choline (mg/1000

kcal) 117 (86, 147)

122 (95,

155)

120 (102,

155) 0.8

Calcium (mg/day) 752 (666,

1269)

657 (437,

871)

509 (354,

1031) 1000-1200 mg

(35%) Calcium (mg/1000

kcal)

443 (313,

624)

325 (271,

604)

322 (205,

704) 0.3

Iron (mg/day) 15 (11, 21) 15 (13, 20) 11 (9, 16)

8-18 mg

(74.5%) Iron (mg/1000 kcal) 7 (6, 9) 9 (8, 13) 7 (6, 10) 0.2

Sodium (mg/day) 2619 (2233,

3400)

1971 (1620,

3290)

2199 (1529,

2709) <2300 mg

(45%) Sodium (mg/1000

kcal)

1340 (1047,

1791)

1381 (917,

1722)

1365 (1095,

1658) 0.9

Values are expressed as mean (standard deviation) for normal variables and median (1st quartile, 3rd

quartile) for non-normally distributed variables.

Values with the same superscript are significantly different.

* Intake was compared to the Dietary Reference Intakes [212-216].

44

Figure 3: Reported total daily physical activity completed

There was no statistically significant difference in average daily physical activity units completed

between healthy control (HC), simple steatosis (SS), and non-alcoholic steatohepatis (NASH) groups

(HC: n=24; SS: n=11; NASH: n=19).

Figure 4: Reported daily minutes of strenuous and very strenuous physical activity

There was no statistically significant difference in average daily physical activity units completed

between healthy control (HC), simple steatosis (SS), and non-alcoholic steatohepatis (NASH) groups

(HC: n=24; SS: n=11; NASH: n=19).

45

5.3 Bacterial Products

5.3.1 Plasma Endotoxin

Plasma endotoxin was not significantly different between groups (p=0.9) with a slightly

narrower range of values for NASH compared to HC and SS (Figure 5). Intra-assay coefficient

of variation (CV) was consistently below 10% and inter-assay CV was acceptable at 13.5%.

Figure 5: Comparison of plasma endotoxin concentration between groups

There was no statistically significant difference in plasma endotoxin concentration (EU/mL) between

healthy control (HC), simple steatosis (SS), and non-alcoholic steatohepatis (NASH) groups (HC: n=25;

SS: n=14; NASH: n=25).

4

4.5

5

5.5

6

6.5

7

7.5

Pla

sm

a E

nd

oto

xin

(E

U/m

L)

HC SS NASH Median

46

5.3.2 Short Chain Fatty Acids and Other Organic Acids

Our secondary outcome, fecal butyric acid, was not significantly different between groups

(p=0.4) (Figure 6). Along with butyric acid the other main SCFA, propionate and acetate

(measured as acetic acid), were also examined. Fecal propionate was significantly higher in SS

patients than in HC (Figure 7). Fecal acetic acid and serum acetic acid did not differ between

groups, p=0.19 and p=0.09 respectively (Figure 8 and 9).

The small number of subjects per group, particularly for SS, and the absence of significant

differences between the three disease groups despite a tendency for SS and NASH values to be

similar and HC to be different from SS and NASH led us to suspect that there might have not

been sufficient power for 3-way comparisons. We therefore chose to combine the SS and NASH

group into one NAFLD group for further metabolite analyses. Comparisons between HC and

NAFLD for the above SCFA and other related organic acids are displayed below in Table 12.

When comparing these organic acids between HC and NAFLD subjects, total fecal SCFA, fecal

propionate and isobutyric acid and serum 2-hydroxybutyrate were all significantly higher in

NAFLD patients versus HC. The ratio of median acetate:propionate:butyrate in fecal samples for

HC was 62.4:22.6:15.0 and was similar for NAFLD patients with a ratio of 61.5:22.6:15.9.

47

Figure 6: Comparison of fecal butyric acid between groups

There was no statistically significant difference in fecal butyric acid concentration (µM) between healthy

control (HC), simple steatosis (SS), and non-alcoholic steatohepatis (NASH) groups (HC: n=28; SS:

n=14; NASH: n=24).

Figure 7: Comparison of fecal propionate between groups

Patients with simple steatosis (SS) had a significantly higher fecal propionate concentration (µM) than

healthy controls (HC). HC and SS patients did not have a significantly different fecal propionate

concentration than non-alcoholic steatohepatitis (NASH) patients (HC: n=28; SS: n=14; NASH: n=24).

0

1000

2000

3000

4000

5000

6000

7000

8000F

ecal B

uty

ric A

cid

M)

HC SS NASH Median

48

Figure 8: Comparison of fecal acetic acid between groups

There was no statistically significant difference in fecal acetic acid concentration (µM) between healthy

control (HC), simple steatosis (SS), and non-alcoholic steatohepatis (NASH) groups (HC: n=28; SS:

n=14; NASH: n=24).

Figure 9: Comparison of serum acetic acid between groups

There was no statistically significant difference in serum acetic acid concentration (µM) between healthy

control (HC), simple steatosis (SS), and non-alcoholic steatohepatis (NASH) groups (HC: n=28; SS:

n=15; NASH: n=18).

0

2000

4000

6000

8000

10000

12000

14000

16000F

ecal A

ceti

c A

cid

M)

HC SS NASH Median

0

20

40

60

80

100

120

140

Seru

m A

ceti

c A

cid

M)

HC SS NASH Median

49

Table 12: Fecal and serum SCFA and organic acids in healthy controls and NAFLD patients

Fecal Metabolite (μM) HC (n=28) NAFLD (n=38) p-value

Butyric Acid 1366 (993, 2072) 1753 (1130, 2266) 0.2

Propionate 2052 (1598, 2419) 2485 (1985, 3649) 0.019

Acetic Acid 5665 (4025, 7061) 6781 (4619, 8384) 0.09

Formic Acid 60 (50, 73) 56 (42, 68) 0.2

Total SCFA (sum of above) 9251 (3286) 11633 (4832) 0.020

Isobutyric Acid 249 (199, 285) 293 (239, 372) 0.017

Isovalerate 181 (115, 223) 172 (136, 208) 0.9

Succinate 121 (67, 175) 94 (58, 187) 0.5

Serum Metabolite (μM) HC (n=28) NAFLD (n=33) p-value

Acetic Acid 51 (37, 69) 37 (33, 59) 0.06

Formic Acid 55 (48, 64) 58 (51, 61) 0.6

2-Hydroxybutyrate 51 (40, 66) 69 (48, 78) 0.011

Values are expressed as mean (standard deviation) for normal variables and median (1st quartile, 3rd

quartile) for non-normally distributed variables.

5.3.3 Metabolites Related to the Bacterial Metabolism of Choline

Fecal choline and trimethylamine as well as serum choline and TMAO were measured. Both

fecal choline (Figure 10) and fecal trimethylamine (Figure 11) were significantly higher in

NAFLD patients than in HC. Despite this difference in fecal values, serum choline (Figure 12)

and TMAO (Figure 13) did not differ between HC and NAFLD patients, p=0.9 and p=0.5

respectively.

50

Figure 10: Fecal choline in healthy controls and NAFLD patients

There was a significantly higher level of fecal choline (µM) in the non-alcoholic fatty liver disease group

(NAFLD) than in healthy control (HC) group (HC: n=28; NAFLD: n=38).

Figure 11: Fecal trimethylamine in healthy controls and NAFLD patients

There was a significantly higher level of fecal trimethylamine (µM) in the non-alcoholic fatty liver

disease group (NAFLD) than in healthy control (HC) group (HC: n=28; NAFLD: n=38).

51

Figure 12: Serum choline in healthy controls and NAFLD patients

There was no statistically significant difference in serum choline concentration (µM) between the healthy

control (HC) and non-alcoholic fatty liver disease (NAFLD) groups (HC: n=28; NAFLD: n=33).

Figure 13: Serum trimethylamine N-oxide in healthy controls and NAFLD patients

There was no statistically significant difference in serum trimethylamine N-oxide (TMAO) concentration

(µM) between the healthy control (HC) and non-alcoholic fatty liver disease (NAFLD) groups (HC:

n=28; NAFLD: n=33).

0

5

10

15

20

25

Seru

m T

rim

eth

yla

min

e N

-ox

ide (

µM

)

HC NAFLD Median

52

5.3.4 Ethanol

Serum ethanol was compared in the three disease groups, but levels did not differ significantly

(p=0.5). There was also no significance when comparing HC with the merged NAFLD group

(p=0.8) (Figure 14). Average daily reported alcohol intake was similar in HC vs SS vs NASH: 0

(0, 1.9) g vs 0.39 (0, 3.0) g vs 0 (0, 0.36) g (p=0.4).

Figure 14: Serum ethanol in healthy controls and NAFLD patients

There was no statistically significant difference in serum ethanol concentration (µM) between the healthy

control (HC) and non-alcoholic fatty liver disease (NAFLD) groups (HC: n=28; NAFLD: n=33).

5.4 Intestinal Microbiota

Intestinal microbiota was compared between the three disease groups for total bacteria, Archaea,

and presence of bacterial taxa of interest as a percentage of total bacteria and as bacterial counts

(log cells/g wet feces). Firmicutes was defined as the sum of Lactobacilli, C. coccoides, and C.

leptum. Full statistical analyses for all taxa are included in Table 13. There was no statistically

significant difference in total bacteria or any other taxa of interest with the exception of C.

leptum as a percent of total bacteria which was significantly lower in NASH versus HC (Figure

15) and E. coli count which was significantly higher in NASH versus HC (Figure 16).

53

Proportion of individuals with Archaea present was not significantly different between HC, SS,

and NASH groups: 36%, 40%, and 50% respectively (p=0.6).

Table 13: Bacterial taxa of interest from qPCR by disease group

Microbiota

(log cells/g wet feces)

HC (n=28) SS (n=15) † NASH (n=24) p-value

Total Bacteria 24.4 (0.62) 24.1 (0.43) 24.3 (0.75) 0.4

Bacteroidetes 8.8 (0.66) 8.6 (0.83) 8.3 (1.2) 0.14

Firmicutes 22.8 (0.97) 22.4 (0.93) 22.6 (0.97) 0.5

Firmicutes:Bacteroidetes 1.1 (0.65) 1.1 (0.73) 1.5 (1.2) 0.2

Bifidobacteria 9.4 (8.4, 9.7) 9.6 (9.2, 10.2) 9.2 (7.5, 9.8) 0.4

Lactobacilli 6.4 (0.92) 6.4 (0.90) 6.5 (0.83) 0.8

C. coccoides 9.4 (0.42) 9.4 (0.37) 9.5 (0.32) 0.7

C. leptum 9.8 (9.5, 10.0) 9.5 (9.2, 9.8) 9.4 (8.5, 9.9) 0.10

E. coli 6.9 (1.2) 7.8 (0.99) 7.8 (1.4) 0.016

Archaea‡ 6.9 (2.3) 5.8 (2.1) 6.9 (2.3) 0.6

Microbiota

(% total bacteria)

HC (n=28) SS (n=15) † NASH (n=24) p-value

Bacteroidetes 2.4 (0.92, 3.8) 3.5 (0.29, 7.4) 0.78 (0.30, 2.8) 0.19

Firmicutes 23 (9.0) 21.4 (10.5) 21.7 (10.1) 0.9

Bifidobacteria 5.1 (0.73, 12.3) 17.2 (4.5, 27.6) 2.7 (0.14, 14.6)

Lactobacilli 0.005 (0.002,

0.020)

0.008 (0.002,

0.064)

0.011 (0.003,

0.023)

0.7

C. coccoides 6.5 (4.3, 9.5) 8.6 (4.9, 11.3) 8.7 (5.5, 14.6) 0.3

C. leptum 15.9 (8.8, 17.9) 10.2 (5.5, 14.2) 7.4 (1.5, 14.8) 0.030

E. coli 0.14 (0.02,0.63) 0.08 (0.01, 0.43) 0.06 (0.003, 0.86) 0.6 Values are expressed as mean (standard deviation) for normal variables and median (1st quartile, 3rd

quartile) for non-normally distributed variables.

Values with the same superscript are significantly different.

† Analysis of Bifidobacteria was done in n=14 patients.

‡ Archaea was only present in 10 HC, 6 SS, 12 NASH patients.

54

Figure 15: C. leptum relative abundance in stool samples from three study groups

C. leptum bacteria as a percentage of total bacteria was significantly lower in the non-alcoholic

steatohepatitis (NASH) group versus the healthy control (HC) group. There was no difference in

C. leptum relative abundance between the HC and simple steatosis (SS) groups or between the

NASH and SS groups (HC: n=28; SS: n=15; NASH: n=24).

55

Figure 16: E. coli count in stool samples from three study groups

Log E. coli bacteria cell count per gram wet feces was significantly higher in the non-alcoholic

steatohepatitis (NASH) group versus the healthy control (HC) group. There was no difference in

E. coli count between the HC and simple steatosis (SS) groups or between the NASH and SS

groups (HC: n=28; SS: n=15; NASH: n=24).

5.5 Relationships between Bacterial Metabolites, Bacteria of Interest, Disease Status, and Diet

Several correlations were studied to investigate potential mechanisms for bacteria and their

metabolites on disease status as well as the possible influence of diet. Correlations between

metabolites and NAFLD related parameters were investigated first (Table 14). These

correlations were run only in NAFLD patients with the exception of BMI which was examined in

all patients. Correlations that were considered moderately strong and relevant to potential disease

mechanisms are featured in Figures 17 to 21. In fecal metabolites there was a significant positive

correlation between fecal formic acid and total inflammation on liver histology, and between

56

both fecal choline and fecal trimethylamine and BMI. In serum metabolites there was a

significant negative correlation between serum acetic acid and ballooning intensity and between

serum choline and liver steatosis. A negative correlation also appeared to be significant between

serum choline and ballooning intensity (rs=-0.40, p=0.022) but is believed to be superfluous

following visual inspection.

Relationships between bacterial taxa of interest and NAFLD related parameters were also

examined in the same method as mentioned above. Results are summarized in Table 15.

Bacterial count and relative abundance were examined in all correlations, however, only

measures where there was some significant relationships are shown. The measure displayed is

noted in a footnote below the table. A significant positive correlation was seen between liver

steatosis and Firmicutes count (Figure 22).

Relationships between bacterial taxa of interest and metabolites were then investigated using

Spearman’s correlations in all patients (Table 16). There were several positive correlations

between bacterial taxa and metabolites, particularly between fermenters such as Firmicutes, C.

leptum, and C. coccoides, and fermentation products including butyric acid, formic acid, and

acetic acid. One of the strongest correlations was between C. leptum and fecal butyric acid

(Figure 23). E. coli count and relative abundance were not correlated to serum ethanol levels

(p<0.05).

Dietary composition was also compared with metabolites (Table 17) and bacterial taxa (Table

18) in all patients. Dietary variables examined included macronutrient composition (protein, fat,

and carbohydrates as a proportion of total energy), fibre (g/1000 kcal), and micronutrients

(vitamin C, E, A, and calcium in units per 1000 kcal). Some products of protein metabolism by

bacteria including succinate and isovalerate were positively correlated with protein intake while

some products of carbohydrate fermentation including butyric and acetic acid were correlated

with fibre and vitamin consumption. Although relatively weak, a relationship of particular

interest was seen between C. leptum and fibre intake (Figure 24). A positive correlation was also

observed between carbohydrate intake and plasma endotoxin (Figure 25).

Correlations between those metabolites that were measured in both stool and serum, for example

fecal choline and serum choline, were not significant (p>0.05). Correlations discussed above

57

were also done for HC and NAFLD groups separately in an attempt to investigate differences in

bacterial metabolism. Relationships were generally similar between disease groups.

Table 14: Correlation between fecal and blood metabolites and NAFLD related parameters

Fecal Metabolite

Steatosis

(%) Fibrosis

Total

Inflammation

Lobular

Inflammation

Ballooning

Intensity NAS

HOMA-

IR BMI

Butyric Acid NS NS NS NS NS NS NS NS

Propionate NS NS NS NS NS NS NS NS

Acetic Acid NS NS NS NS NS NS NS NS

Formic Acid NS NS

rs=0.61

p=0.015

n=15

NS NS NS NS NS

Isobutyric Acid NS NS NS NS NS NS NS NS

Isovalerate NS NS NS NS NS NS

rs=0.36

p=0.040

n=32

NS

Succinate NS NS NS NS NS NS NS NS

Choline NS NS NS NS NS NS NS

rs=0.43

p=0.0003

N=66

Trimethylamine NS NS NS NS NS NS NS

rs=0.32

p=0.010

N=66

Blood Metabolite Steatosis

(%) Fibrosis

Total

Inflammation

Lobular

Inflammation

Ballooning

Intensity NAS

HOMA-

IR BMI

Acetic Acid NS NS NS NS

rs=-0.44

p=0.012

n=32

NS NS NS

Formic Acid NS NS NS NS NS NS NS NS

2-

Hydroxybutyrate NS NS NS NS NS NS NS

rs=0.37

p=0.003

n=61

Ethanol NS NS NS NS NS NS NS NS

Choline

rs=-0.40

p=0.022

n=32

NS NS NS

rs=-0.40

p=0.022

n=32

rs=-0.48

p=0.005

n=32

NS NS

TMAO NS

rs=0.38

p=0.025

n=34

NS NS NS NS NS NS

Endotoxin NS NS NS NS NS NS NS NS

Correlations are for NAFLD patients only for all parameters except for BMI which was compared in all

patients.

58

Figure 17: Relationship between fecal formic acid and total inflammation in NAFLD patients

Figure 18: Relationship between fecal choline and body mass index in all subjects

59

Figure 19: Relationship between fecal trimethylamine and body mass index in all subjects

Figure 20: Relationship between serum acetic acid and hepatocyte ballooning intensity in

NAFLD patients

60

Figure 21: Relationship between serum choline and liver steatosis in NAFLD patients

Table 15: Correlation between intestinal microbiota by qPCR and NAFLD related parameters

Steatosis

(%) Fibrosis

Total

Inflammation

Lobular

Inflammation

Ballooning

Intensity NAS HOMA-IR BMI

Bacteroidetes NS NS NS NS NS NS NS NS

Lactobacilli NS NS NS NS NS NS NS NS

C. coccoides NS NS NS NS NS NS NS NS

C. leptum

rs=0.41

p=0.012

n=38

NS NS NS NS NS NS NS

Bifidobacteria NS NS NS NS NS NS NS NS

E. coli NS NS NS NS NS NS NS

rs=0.25

p=0.038

n=67

Archaea NS NS NS NS NS NS

rs=0.48

p=0.005

n=33

NS

Firmicutes

rs=0.47

p=0.003

n=38

NS NS NS NS NS NS NS

Correlations are for % of total bacteria except for E. coli and Archaea where count/g wet feces was used.

Correlations are for NAFLD patients only for all parameters except for BMI which was compared in all

patients.

61

Figure 22: Relationship between Firmicutes count and liver steatosis in NAFLD patients

Figure 23: Relationship between C. leptum count and fecal butyric acid in all subjects

62

Table 16: Correlations between intestinal microbiota and metabolites in all subjects

METABOLITE BACTERIAL TAXA OF INTEREST

Fecal (n=66) Bacteroidetes Firmicutes Lactobacilli C. coccoides C. leptum Bifidobacteria E. coli Archaea

Butyric Acid NS

rs=0.38

p=0.002 NS NS

rs=0.41

p=0.0006 NS NS NS

Propionate NS NS NS NS NS NS NS NS

Acetic Acid NS NS NS NS NS NS NS NS

Formic Acid NS NS NS NS

rs=0.27

p=0.031 NS NS NS

Isobutyric Acid NS NS NS NS NS NS NS NS

Isovalerate rs=-0.31

p=0.010 NS NS NS NS NS NS NS

Succinate NS NS NS NS NS NS NS NS

Choline NS NS NS

rs=0.27

p=0.026 NS NS NS NS

Trimethylamine NS NS NS NS NS NS NS NS

Blood (n=59) Bacteroidetes Firmicutes Lactobacilli C. coccoides C. leptum Bifidobacteria E. coli Archaea

Acetic Acid NS

rs=0.43

p=0.001 NS NS

rs=0.41

p=0.001 NS NS NS

Formic Acid NS NS NS

rs=0.33

p=0.011 NS NS NS NS

2-

Hydroxybutyrate NS NS NS

rs=0.28

p=0.029 NS

rs=-0.34

p=0.009 NS NS

Ethanol NS NS NS NS NS NS NS NS

Choline NS NS NS NS NS NS NS NS

TMAO NS NS NS NS NS NS NS NS

Endotoxin NS NS NS NS NS NS NS NS

Correlations with IM presented are for %bacteria except for Firmicutes, Archaea, C. leptum, E. coli, and C. coccoides where cell count/g wet feces was

used.

63

Table 17: Correlations between metabolites and diet in all subjects

METABOLITE REPORTED DIETARY INTAKE

Fecal (n=52) Fat

(%kcal/BMR)

Carbohydrate

(%kcal)

Protein

(%kcal)

Fibre

(g/1000kcal)

Vitamin A

(IU/1000kcal)

Vitamin E

(mg/1000kcal)

Vitamin C

(mg/1000kcal)

Calcium

(mg/1000kcal)

Butyric Acid NS NS NS rs =0.29

p=0.038

rs =0.31

p=0.027 NS NS NS

Propionate rs=-0.31

p=0.028 NS NS NS NS NS NS NS

Acetic Acid NS NS rs =0.32

p=0.021 NS NS NS NS NS

Isobutyric Acid rs =-0.29

p=0.037 NS NS NS NS NS NS

rs =-0.29

p=0.037

Isovalerate NS NS rs =-0.33

p=0.016 NS NS NS NS NS

Succinate NS NS rs =0.31

p=0.024 NS NS NS NS NS

Choline NS NS rs =0.43

p=0.002 NS NS NS NS NS

TMA NS NS rs =0.33

p=0.016 NS NS NS NS NS

Blood (n=48)†

Fat

(%kcal/BMR)

Carbohydrate

(%kcal)

Protein

(%kcal)

Fibre

(g/1000kcal)

Vitamin A

(IU/1000kcal)

Vitamin E

(mg/1000kcal)

Vitamin C

(mg/1000kcal)

Calcium

(mg/1000kcal)

Acetic Acid NS NS NS NS NS rs =0.39

p=0.007 NS NS

2-Hydroxybutyrate NS NS NS rs =0.29

p=0.042 NS NS NS NS

L-Lactic Acid NS NS NS NS rs =-0.32

p=0.027 NS NS NS

Ethanol NS NS NS NS NS NS rs =-0.29

p=0.045 NS

Choline NS NS NS NS NS NS rs =-0.38

p=0.008 NS

Endotoxin rs =-0.33

p=0.019

rs =0.39

p=0.005 NS NS NS NS NS NS

† n=50 for plasma endotoxin correlations only

64

Table 18: Correlations between intestinal microbiota by qPCR and diet in all subjects

Bacterial Taxa

of Interest

Fat

(%kcal/BMR)

Carbohydrate

(%kcal)

Protein

(%kcal)

Fibre

(g/1000kcal)

Vitamin A

(IU/1000kcal)

Vitamin E

(mg/1000kcal)

Vitamin C

(mg/1000kcal)

Calcium

(mg/1000kcal)

Bacteroidetes NS NS NS NS NS NS NS NS

Lactobacilli NS NS NS NS NS NS NS NS

C. coccoides NS NS NS NS NS NS NS NS

C. leptum NS NS NS rs=0.29

p=0.032

n=53

rs=0.33

p=0.015

n=53

NS

rs=0.38

p=0.005

n=53

rs=0.33

p=0.015

n=53

Bifidobacteria NS NS NS NS NS NS NS NS

E. coli NS NS NS NS NS NS NS NS

Archaea NS NS NS NS NS NS NS NS

Firmicutes NS NS NS NS

rs=0.38

p=0.005

n=53

NS

rs=0.33

p=0.016

n=53

NS

Correlations with IM displayed are for bacterial taxa as a percent of total bacteria except for Firmicutes, E. coli and Archea where count/g

wet feces is shown.

65

Figure 24: Relationship between relative abundance of C. leptum and reported dietary fibre

intake in all subjects

Figure 25: Relationship between plasma endotoxin and reported carbohydrate intake in all

subjects

66

6 Discussion

To our knowledge, this is the first human study that measures both the IM and bacterial

metabolites in adults with biopsy confirmed SS, NASH, and HC. The primary and secondary

study outcome, plasma endotoxin and fecal butyrate, did not differ between these groups.

However, other SCFA and metabolites showed differences between NAFLD patients and HC.

NAFLD patients had higher fecal total SCFA, propionate and isobutyric acid and higher serum

2-hydroxybutyrate than HC. Significant differences were also found between NAFLD and HC

for fecal compounds related to the bacterial metabolism of dietary choline. Fecal choline and

trimethylamine were higher in NAFLD patients but serum choline and TMAO did not differ

from HC. HC, SS, and NASH patients had similar total bacteria counts in stool samples as

measured by qPCR and the proportion of subjects who had detectable Archaea did not differ

between groups. NASH patients were found to have lower proportions of Clostridium leptum and

higher counts of Escherichia coli in their feces. Some bacterial metabolites and IM taxa were

correlated with histological findings and dietary intake, suggesting potential contribution to

NAFLD pathogenesis.

6.1 Endotoxin

Based on the findings of four human studies in NAFLD patients [129-132], three of which

measured blood endotoxin levels using a similar method to ours [129, 130, 132], we predicted

that NASH patients would have a significantly higher plasma endotoxin level than SS or HC. We

found no significant differences between the three disease groups, and no perceivable trend. This

was also found in a study released last month which found no difference in endotoxin measured

via LAL assay between HC and NAFLD patients [217]. Endotoxin levels in the entire study

population ranged from 4.54 EU/mL to 7.14 EU/mL with a slightly narrower range for NASH

patients (5.14-7.00 EU/mL). This was similar to the levels found by Harte et al. (~3.2-14.8

EU/mL) who used the same assay [130], but higher than the levels seen by Thuy et al. and

Volynets et al. who used the same assay type from a different company and whose values ranged

from about 0.001-0.28 EU/mL [129, 132]. Studies in other disease populations also appear to be

shifted up or down as an entire study population, but one would still expect to see a difference

between groups if it was present. There are two main factors which we believe may have played

67

a role in the lack of significant differences in endotoxin level between our groups: diet and

disease severity.

Dietary intake was measured in both Thuy et al. and Volynets et al. and in both of these studies

NAFLD patients were found to have a higher dietary intake of carbohydrates, particularly

fructose in Thuy’s study [129, 132]. Volynets also found a positive correlation between

carbohydrate intake and endotoxin level. Although the mechanism for this correlation is unclear

it is possible that in both of these studies the higher endotoxin level in the NAFLD group was

due to higher carbohydrate intake levels rather than disease state. Interestingly, we also found a

positive correlation between plasma endotoxin and dietary carbohydrate intake, but our disease

groups did not differ in levels of carbohydrate intake. It is conceivable therefore that we did not

see a difference in endotoxin between groups because their carbohydrate intake was equal.

Notably a negative correlation was detected in our patients between reported fat intake and

endotoxin level. This is in contradiction to previous animal studies that have found a positive

correlation between these two variables [127, 218-220]. This is thought to be due to a fat

mediated uptake of endotoxin whereby fats are incorporated into chylomicrons, which also have

a high affinity for endotoxin, and therefore endotoxin enters circulation along with chylomicrons

[221, 222]. It is important to note that the blood used for endotoxin analysis in our patients was

drawn after a 12 hour fast therefore recent fat intake would not have had a significant effect on

endotoxin intake. Fat absorption occurs in the small intestine rather than the colon so colonic

endotoxin as a result of microbiota should not be influenced by fat uptake. It is therefore

plausible that this is a superfluous correlation that reflects a dietary composition with high

carbohydrate intake rather than low fat intake being related to higher endotoxin levels.

The third study that measured plasma endotoxin in a NAFLD population did not account for

dietary intake, but endotoxin levels were positively correlated with HOMA-IR [130]. We did see

a difference in HOMA-IR between our HC and NASH group, but the magnitude of this

difference was far lower than that seen in Harte’s study [130]. In their study NASH patients were

much more insulin resistant with a median HOMA-IR of 8.6 whereas our NASH patients only

had a HOMA-IR of 4.26. This is likely because the diabetic individuals in our study were taking

medication and were well controlled. If our patients had poorer control and greater insulin

resistance we may have seen a greater level of endotoxin in this group. A study by Verdam et al.

68

that looked at IgG levels against endotoxin rather than blood endotoxin concentration found that

IgG levels were positively correlated with NASH severity [131]. In our study the NASH patients

had relatively mild disease with a median NAS of only 5 out of a possible 8. It is conceivable

that the severity of the disease was not great enough to cause endotoxin differences between

disease groups.

6.2 SCFA and Related Metabolites

This is the first human study to measure fecal butyric acid in biopsy confirmed NAFLD patients

versus HC. A study by Wong et al. found that NASH patients had a lower prevalence of butyrate

producing Faecalibacterium, but they did not measure butyrate levels [14]. Animal studies

showed beneficial effects of butyrate producing probiotic supplementation including

improvements in weight, steatosis, FFA, inflammation, and reduced intestinal permeability [154,

155]. One of these studies was also able to correlate these beneficial improvements with fecal

and plasma butyrate concentrations [155]. In our study there was no difference in fecal butyrate

between the three groups or between the combined NAFLD group and HC. There was, however,

a significant difference in the prevalence of the butyrate producing C. lepum bacteria, with a

significantly lower relative abundance in NASH patients. This will be discussed separately

below.

It is perhaps naïve to presume that we would see a clear connection between bacterial prevalence

of butyrate producers, fecal butyrate, and disease state. The concentration of fecal SCFA is

frequently used as a proxy for measuring the bacterial production of these compounds, but it

does not account for differences in colonic absorption. We assume that a higher concentration of

fecal butyrate means that more butyrate was produced and more butyrate will therefore be

absorbed in the colon. A study by Vogt and Wolever, however, found otherwise [223]. Vogt and

Wolever conducted a study where subjects infused a SCFA solution into their colon, retained it

for several minutes, and then voided. Absorption of the three main SCFA as well as

combinations thereof were measured and compared with SCFA composition in the subject’s

normal feces. They found that the acetate absorption from the colonic infusion was negatively

correlated with acetate concentration in normal stools and that other SCFA did not have

significant correlation between these two measures [223]. This could mean that a lower fecal

concentration of a certain SCFA indicates a greater absorption rather than a lower production

69

from the IM. The amount of SCFA produced and absorbed can also be influenced by fecal transit

time, which was not measured in this study [224]. Further research is needed to better understand

the production and uptake of butyrate in different disease states. It is also important to note that

the proposed mechanism for the beneficial role of butyrate uptake in healthy patients was in

preventing excessive intestinal permeability. Jiang et al. and Volynets et al. did find increased

intestinal permeability in NAFLD patients, but did not look at relationships between

permeability, IM, and SCFA production [15, 132].

The ratio of SCFA in our NAFLD patients and HC (approximately 62:23:15

acetate:propionate:butyrate) was fairly similar to the commonly quoted ratio of 60:20:20 and

60:25:15 noted in the literature [224]. When looking at the other main SCFA and related

carboxylic acids in fecal and serum samples we found a significantly greater level of total fecal

SCFA, fecal propionate, fecal isobutyric acid, and serum 2-hydroxybutyrate in NAFLD patients

compared to HC. A greater amount of total fecal SCFA, particularly propionate, was previously

found in overweight and obese adults compared to lean controls [118, 225]. As discussed in

section 2.2.1 it has been suggested that the human obese IM is enriched with genes that code for

the bacterial metabolism of macronutrients leading to an increased energy absorption of up to

150 kcal per day [119, 121]. The increased energy harvest from SCFA is one potential

mechanism through which these metabolites may affect NAFLD pathogenesis. However, in

order to confirm this we would have needed to conduct bomb calorimetry on stool samples,

which was not included in our study protocol.

The primary role of propionate, once absorbed and taken up by the liver, is to function as a

substrate for gluconeogenesis [226]. As an additional energy substrate propionate could increase

adiposity and potentially contribute to steatosis. Further studies on the mechanistic role of

propionate on steatosis and energy balance, including gene expression would be helpful.

Fecal isobutyric acid is a branched chain fatty acid that is formed by the bacterial catabolism of

branched chain amino acids [224]. Isobutyric acid is an isomer of butyric acid and is also closely

related to propionate (with the addition of a methyl branch) [227]. The functional role of

isobutyric acid is not well described, but as it is an intermediate in protein breakdown it is

possible that elevated levels in the NAFLD group are secondary to the increased protein intake

that was observed in these individuals.

70

2-hydroxybutyrate (alpha-hydroxybutyrate) is a metabolite formed during amino acid catabolism

and glutathione anabolism [228]. Although this compound has been mentioned as an

intermediary in the bacterial metabolism of amino acids, it is not clear how much is actually

produced and absorbed in the human colon and incorporated into the blood [229]. It is therefore

more likely that the serum levels measured in our study, which were found to be significantly

higher in NAFLD patients, are due to the presence of NAFLD and its related metabolic

syndrome. Oxidative stress can increase the production of glutathione in the liver and may

therefore increase the amount of 2–hydroxybutyrate produced [228]. Research has recently

suggested that the oxidative stress triggered by insulin resistance, a primary mechanism in

NAFLD pathogenesis, can increase circulating levels of 2–hydroxybutyrate [230, 231]. Li et al.

conducted metabolic profiling in 48 diabetic patients and 31 healthy controls [230]. They found 5

biomarkers that accurately distinguished diabetic patients from healthy controls, including 2–

hydroxybutyrate [230]. Similarly, a study by Gall et al. conducted non-targeted biochemical

profiling on 399 subjects who also underwent the gold-standard hyperinsulinemic euglycemic

clamp testing and oral glucose tolerance testing for identification and grading of insulin

resistance [231]. After statistical analysis, 2–hydroxybutyrate was identified as the best candidate

biochemical to separate insulin resistant from insulin sensitive individuals independent of age,

sex, or BMI [231]. It is therefore likely that the higher 2–hydroxybutyrate levels that we see in

NAFLD patients are due to insulin resistance and resulting oxidative stress rather than alterations

in IM metabolism. Indeed, 2-hydroxybutyrate was the only serum metabolite correlated with

BMI, pointing to its potential relationship to the metabolic state of NAFLD patients.

Two SCFA were also correlated with markers of disease severity in NAFLD patients. Fecal

formic acid, the smallest of the SCFA, was positively correlated with total inflammation. This is

a novel finding in NAFLD and little research exists on the physiological effects of formic acid.

One study found an increased level of serum formic acid in patients with inflammatory bowel

disease in comparison to healthy controls, which may also support a pro-inflammatory role, but

the mechanism remains unknown [232]. Serum acetic acid seemed to have a more beneficial

effect in NAFLD patients with a negative correlation with hepatocyte ballooning intensity.

Recently it was suggested that acetate may have an anti-inflammatory function through

interaction with GPR43, but the exact mechanism through which acetic acid may reduce

ballooning intensity is not known [169].

71

In relation to dietary intake, some of the fecal and serum metabolites measured were positively

correlated with potential substrates for their production. Fecal isovalerate and succinate, products

of the catabolism of protein by IM, were positively correlated with protein intake [224]. Fecal

butyric acid was correlated with fibre and vitamin A intake. Although not all fibres are

fermentable, many of the substrates used for butyrate production would be categorized as a fibre

by the data analysis software. Vitamin A may also be a marker of these substrates as they are

both primarily found in plant-based foods. Serum acetic acid was positively correlated with

Vitamin E, which is again a primarily plant-derived vitamin which may be an indicator of a

relationship between this SCFA and plant-based substrates available for fermentation.

6.3 Metabolites Related to the Bacterial Metabolism of Choline

Another proposed mechanism for IM in NAFLD pathogenesis is through the bacterial

metabolism of dietary choline to trimethylamine. We hypothesized that NAFLD patients would

have a lower circulating choline level due to an increased bacterial breakdown of choline which

would also result in increased fecal trimethylamine and serum TMAO. We did observe a higher

fecal trimethylamine level as well as a higher fecal choline level. Unfortunately we cannot link

fecal choline with dietary choline intake as this dietary information was incomplete in our

database. We did, however, see a slightly higher intake of proteins in NAFLD patients which

may include choline rich foods such as eggs, meat, and fish. This may have led to a higher

amount of choline reaching the colon. This is supported by the positive correlation that we found

between both fecal choline and fecal trimethylamine and reported dietary protein intake. We

could therefore suggest that a higher fecal choline (substrate) level resulted in a higher fecal

trimethylamine (product) level. Serum choline and TMAO, however, did not differ between

groups.

Previous human studies looking at the role of choline in NAFLD pathogenesis have not

consistently measured serum choline. The only study that did so was by Nehra et al. They were

looking at a potential role for choline deficiency as a result of celiac sprue in NAFLD

pathogenesis [233]. They found no correlations between disease severity and serum choline

levels. They did not compare choline levels in NAFLD patients to HC. It is important to note that

the proposed role for choline deficiency in NAFLD pathogenesis is through a decreased

production and export of VLDL from the liver as a result of lower phosphatidylcholine

72

availability. A previous study from our research group did find that SS and NASH patients had

lower hepatic phosphatidylcholine than HC [180]. It is plausible therefore that although serum

levels are not different between our groups, phosphatidylcholine levels are reduced as a result of

reduced choline bioavailability. The issue with this theory is that fecal choline levels were higher

in NAFLD patients. Like SCFA, however, we are unsure of whether a high fecal level reflects a

high colonic absorption or a low colonic absorption.

Our study is the first in humans to measure both serum choline and NAFLD characteristics on

liver histology. Interestingly, serum choline was negatively correlated with liver steatosis and

hepatic ballooning intensity in NAFLD patients. The role of dietary choline deficiency in

steatosis is well known [10, 119, 154] and a potential role of low choline availability in disease

progression through oxidative and endoplasmic reticulum stress has also been discussed [175-

178]. Despite no significant difference in levels between disease groups our findings still support

the role of reduced choline availability in NAFLD pathogenesis.

As mentioned previously, trimethylamine is absorbed in the colon and processed in the liver to

form TMAO. This conversion is catalyzed by flavin monooxygenase 3 (FMO3) [234]. The

expression of FMO, however, can vary from person to person and result in varying levels of

serum TMAO [235]. It is therefore plausible that a significantly greater amount of fecal TMA is

absorbed in NAFLD patients but may not have resulted in a significantly higher level of TMAO.

The effect of high TMA levels in the liver and circulation appear to be benign. Individuals who

lack FMO3 due to a genetic mutation develop a condition called trimethylaminuria where high

levels of TMA results in a fishy odour in the patient’s sweat, urine, and saliva, however no other

physiological consequences are mentioned [235]. Serum TMAO levels, however, has been

associated with several chronic diseases including cardiovascular disease [236-238] and it is

known that NAFLD patients are at increased risk of cardiovascular disease [239]. However, our

current findings do not show higher levels of TMAO in NALFD patients or associations with

NAFLD pathogenesis.

Lastly, it is important to note that both fecal choline and fecal trimethylamine were correlated

with BMI. It is possible that as individuals with NAFLD are heavier, greater BMI but not disease

state is associated with higher levels of these two metabolites. Unfortunately as our groups differ

significantly by BMI it is not possible to decipher the effect of disease versus BMI. Further

73

studies with a sufficient sample size of obese healthy controls would better distinguish the

difference between BMI and NAFLD.

6.4 Ethanol

The last metabolite of particular interest in our study was serum ethanol. We predicted, in line

with the findings of Zhu et al., that patients with NASH would have a significantly higher

ethanol level than HC and SS patients and that this would be related to a higher prevalence of

ethanol producing bacteria such as E. coli [11]. We did not find a significant difference in serum

ethanol levels between disease groups, but our mean levels were quite similar to the mean level

for the HC and obese patients in the Zhu et al. study which was approximately 25 µM compared

to their NASH group which was approximately 35 µM [11]. It is important to consider, however,

that the Zhu study was conducted on children who were assumed to have zero alcohol intake. It

is known that increased exposure to ethanol leads to increased metabolic tolerance, meaning that

ethanol is more readily metabolized and blood ethanol levels decrease more quickly after

repeated exposures [240]. It is therefore possible that as our NAFLD patients were adults they

were better able to tolerate ethanol exposure and therefore their serum levels quickly returned to

the normal levels seen in the HC, whereas in the children a slight increase in ethanol exposure

had a more lasting effect and continued to be elevated in serum. This does not mean that ethanol

exposure from bacterial metabolism in adult NAFLD patients is benign as the ethanol will still

travel directly from the colon to the liver, potentially causing an increased production of toxic

metabolic intermediates such as acetaldehyde or increased intestinal permeability [173, 241].

6.5 Intestinal Microbiota

Disease groups did not differ significantly in the relative abundance or counts of Bacteroidetes,

Firmicutes, Archaea, Bifidobacteria, Lactobacilli, or C. coccoides. Two main differences,

however, were found in the bacterial composition of fecal samples in NASH patients compared

to healthy controls. NASH patients had a significantly lower proportion of the C. leptum

bacterial group as a percent of total bacteria and a significantly higher count of E. coli compared

to healthy controls.

The C. leptum bacterial group was lower in NASH patients. It is also known as Clostridial

Cluster IV which compose approximately 16-25% of the intestinal bacteria [242, 243]. The

74

bacteria contained in this group ferment carbohydrates in the colon to produce SCFA,

particularly the most abundant bacteria in this group, Faecalibacterium prausnitzii which is a

butyrate producer [153]. Indeed, C. leptum was positively correlated with dietary fibre intake

which would include potential substrates for this fermentation. One of the few human studies on

NAFLD and microbiota by Wong et al. similarly found that patients with NASH had a

significantly lower abundance of Faecalibacterium [14]. They suggested that F. prausnitzii may

have protective anti-inflammatory properties, preventing NASH [244]. Another possible

mechanism could be that lower Faecalibacterium in that study’s NASH group could reduce the

production of butyrate which is known to have anti-inflammatory action [150, 151] and

strengthening effects on the intestinal epithelium [145-147]. Although differences in fecal

butyric acid between disease groups was not significant in our study, we did see a positive

correlation between C. leptum abundance and fecal butyric acid levels.

The significantly higher abundance of E. coli in NASH patients in comparison to healthy

controls is also supported by the literature. Zhu et al. found that children with NASH had a

significantly higher abundance of Escherichia than both HC and obese children [11]. This was

associated with higher serum ethanol in NASH patients [11]. As discussed above our study did

not find a significant difference in ethanol levels between groups and additionally serum ethanol

was not correlated with E. coli abundance. This again may be due to adults’ capacity to clear

ethanol efficiently. Future metagenomic studies may be able to look at the genetic capacity of the

entire IM to better understand if the bacterial production of ethanol is associated with NASH.

A previous study by our research group, using some of the same subjects, found a significantly

lower relative abundance of Bacteroidetes in NASH patients compared to HC [13]. Although we

saw a trend towards this relationship, it did not reach significance. It is possible that this is

simply because the relationship was lost with a larger sample size, particularly of healthy

controls, or this may be influenced by length of stool sample storage. Research has shown that

with extended storage of stool samples at -80 °C the ratio of Firmicutes:Bacteroidetes increases

[245]. It is therefore possible that the relationship that was previously seen is no longer

significant due to changes that occurred in storage. A sub-analysis comparing results from the

subjects with qPCR from DNA extracted in 2012 and DNA extracted in 2014 is planned to

further investigate differences in IM that may occur during storage.

75

Lastly, although Firmicutes did not differ between groups, Firmicutes count was positively

correlated with hepatic steatosis. The Firmicutes phylum contains many fermenting bacteria and

is therefore responsible for much of the SCFA production [111]. It is therefore possible that the

difference in the amount of total fecal SCFA in the feces of the NAFLD patients compared to HC

is due, in part, to a higher count of Firmicutes which produce an influx of energy to the liver,

contributing to fat accumulation.

6.6 Diet and Physical Activity

The dietary data used in this study was collected from a 7-day food record with the assistance of

validated 2D visual models for portion estimates and clear instructions provided by a nutrition

expert. All food records were reviewed and follow-up questions were asked of the subject to

clarify any missing or unclear details regarding method of preparation or ingredients consumed.

The use of a 7-day food record in this study is superior to the food frequency questionnaires used

in many studies because it accounts for recent dietary intake which is of greatest importance for

the IM and metabolites and is a better estimate of individual micro and macronutrient intake

[246]. In addition, 7-day food records are completed at the time of consumption and therefore

reduce the risk of memory-related errors.

Dietary intake between the three disease groups was similar with the exception of energy intake.

A trend was seen for higher protein intake in SS patients but was not significant after Bonferroni

correction for multiple comparisons. Reported kilocalorie intake was significantly lower in

NASH patients compared to HC, however, there was no difference between HC and SS patients

or NASH and SS patients. The EER (energy requirements including needs for reported physical

activity) and BMR (energy required for basic physiological functioning) were then compared

with reported energy intake, a method used to estimate underreporting [247]. The proportion of

EER reported was not different between groups, but the proportion of BMR reported was. The

NASH patients reported consuming a significantly smaller proportion of their BMR than the HC

with a median that was below 100%. Physiologically someone who is consuming less than their

BMR should be losing weight, however, none of our subjects reported weight loss. There are two

possible explanations for this discrepancy. One is that the BMR did not accurately estimate these

patients’ metabolic rate, which may be depressed secondary to metabolic disturbances, reduced

lean body mass or a sedentary lifestyle. The second is that NASH patients may be underreporting

76

their dietary intake. Underreporting is particularly suspected as this has been reported

disproportionally in overweight and obese individuals [65, 248, 249].

To address the possibility of underreporting in study subjects we chose to focus on dietary

composition rather than energy intake. We have therefore expressed nutrients as a proportion of

energy intake and in the case of fat have also adjusted for age and weight by dividing the percent

of kilocalories from fat by BMR as was done in one of our previous publications [13]. These

energy-controlled values were used for all statistical analyses and dietary intake remained similar

between groups. Notably dietary quality was quite poor in all patient groups, reflecting a typical

Western-style diet with low dietary fibre intake and high intakes of saturated fat and sodium.

This was also seen in a previous study from our lab [60].

Physical activity was recorded in a self-reported 7-day activity record. Average daily physical

activity units completed and average daily minutes of strenuous and very strenuous physical

activity did not differ between disease groups, however, a trend was observed for a lower level of

high intensity physical activity in SS and NASH patients. Previous work from our team saw a

significantly greater proportion of NASH patients than HC who were classified as sedentary

[60]. This is significant as physical activity can reduce excess body weight and improve insulin

resistance and other metabolic abnormalities that contribute to NAFLD [56, 250-252]. However,

only vigorous activity was reported to be associated with decreased risk of developing NASH

and advanced fibrosis [58]. Like dietary intake, it is possible that physical activity was

overestimated in our obese subjects who may have perceived some physical activities as more

strenuous than HC [253, 254]. Literature in the field of IM only saw differences in bacteria based

on physical activity between sedentary individuals and athletes therefore it is unlikely that any

small, statistically insignificant, differences between physical activity in our groups affected IM

and metabolites [255].

6.7 Potential Confounders

Aside from the variables already discussed above there were a few other demographic and

clinical variables that differed between disease groups: age, country of origin, BMI, insulin

resistance, chronic disease diagnosis and medication use. Many of these differences were

expected as they are risk factors for or part of the pathogenesis of NAFLD. The potentially

confounding effect of each is discussed below.

77

Patients in both the SS and NASH group were significantly older than HC. This was expected

because age is a risk factor for the development of NAFLD [1]. Age may affect the production of

bacterial metabolites secondary to an increase in colonic transit time [224, 256, 257], however,

the research regarding the effect of age on transit time is mixed [258, 259]. It is also unclear at

what age transit time slows with the majority of studies focusing on seniors who were older than

our patient population. IM composition has also been shown to change with age, but remains

relatively stable in adults until they are elderly meaning that age is unlikely to affect the IM

composition of our subjects [260].

Although the ethnicity of our patients does not differ significantly between groups, we did see a

significantly higher proportion of Canadian-born individuals in the HC group than in the SS and

NASH groups. The intestinal microbiota of children and adults has been shown to differ

significantly between countries [261-264]. The differences between these populations, however,

have been largely attributed to dietary differences such as an increased amount of fermentable

fibre in the diet of African children versus those in Europe [261]. What is unknown is whether

individuals who immigrate to Canada develop a ‘Westernized IM’ or retain their original IM

composition. Further studies are needed to understand this transition.

BMI was significantly higher in the NASH group than HC and trended higher for SS versus HC

but this difference did not reach statistical significance. This was expected as BMI is a primary

risk factor for NAFLD [1]. This was also expected because although the HC group did have a

median BMI in the overweight range, the BMI of these patients was somewhat limited secondary

to the nature of their recruitment. HC were recruited from a group of healthy living liver donors.

Due to the high prevalence of NAFLD in obese individuals, typically obese people are not

considered as potential donors. Due to the fact that BMI and disease status were closely related

in our patient samples, the representation of diagnostic groups within each BMI category was

very unbalanced, which prevented the possibility of stratified analysis or ANCOVA to control

for BMI in order to distinguish the effect of BMI from that of disease diagnosis. Interestingly

only fecal choline, fecal trimethylamine, and serum 2-hydroxybutyrate were significantly

correlated with BMI.

Biochemical differences between disease groups were also secondary to disease state including

higher AST and ALT in SS and NASH patients than in HC and higher insulin, HOMA-IR, and

78

blood triglyceride level in NASH patients. NASH patients also had an increased prevalence of

hypertension, which is unsurprising as obesity is a known risk factor for both diseases [1, 265].

Medication use differed in accordance with clinical conditions with a significantly higher

proportion of NASH and SS patients taking cardiovascular medications, primarily

antihypertensives, than HC and with significantly more NASH patients taking lipid lowering

drugs than HC. Interestingly, SS patients took significantly more NSAIDS than HC. Only

patients taking low dose NSAIDS were included in this study, but it is possible that without these

anti-inflammatory medications some of these patients may have progressed on to NASH. No

studies to date have investigated this potential preventative treatment.

79

7 Strengths and Limitations

This was the first study in humans to measure both intestinal microbiota and bacterial products in

biopsy confirmed SS and NASH patients in comparison to healthy controls. This is one step

forward in our understanding of the role of IM in NAFLD pathogenesis through various bacterial

products. Given the high prevalence of NAFLD and limited treatment options it is worthwhile to

consider the role of the IM and its metabolites for further research on potential therapies or

preventative strategies.

With the exception of one previous study from our group [13] no other human study has

compared the IM of NAFLD patients with HC where all patient groups had liver biopsies

available for histological analysis. Liver biopsies are currently the gold standard for NASH

diagnosis and disease grading [2] but previous studies have been limited by the ethical issue for

obtaining a liver biopsy from a healthy patient. Our healthy living liver donor group allows us to

obtain this vital data without a separate invasive procedure. Even in HC who were unable to

undergo surgery and therefore did not provide a liver biopsy, analysis of liver health was

significantly more detailed than previous studies which relied primarily on normal liver enzymes.

Although dietary and physical activity data may have been limited by self-reporting, it is a

strength of this study that we were able to compare these lifestyle factors between groups and

examine the relationship between diet, IM, and metabolite composition. This data therefore

allowed us to make connections that were limited in other studies.

There were some limitations in this study as well. As this was a cross-sectional study, causal

relationships cannot be drawn and further work is therefore necessary to understand the

physiological mechanisms behind the relationships that we observed. A significant limitation

was the small sample size in our SS group which was half of what was determined to be required

a priori. This was due to the fact that SS is largely a benign and asymptomatic disease that is

usually detected by ultrasound and does not require a liver biopsy. Although our SS patients had

elevated liver enzymes that necessitated a referral to a hepatologist and subsequent liver biopsy,

those patients are now generally followed with new surrogate markers such as Fibrotest [266] or

Fibroscan [267] as per the American Gastroenterological Association guidelines [2], so liver

biopsies are no longer performed unless patients demonstrate features suggestive of NASH or

80

fibrosis. Despite this limitation ours is one of few studies on NAFLD and IM that diagnosed SS

via liver biopsy.

In this study IM composition was limited to the taxa selected for qPCR, meaning that an

understanding of the full IM community is not possible. The value of having bacterial counts,

however, is not to be underestimated as the actual amount of bacteria present is of particular

interest when comparing bacteria and metabolites. Future analyses that compare the full IM

community using Illumina 16s rRNA sequencing and metagenomics are in progress.

81

8 Conclusions

The findings of this study provide further evidence for a relationship between the IM, bacterial

products, and disease severity in NAFLD. More specifically we have observed in NAFLD

patients: a higher presence of some fermentation products, the production of potentially harmful

metabolites, the altered metabolism of an essential nutrient (choline), and differences in IM

composition. In relation to the study aims, hypotheses, and proposed mechanisms, the study

results were as follows:

The study did not find a significant difference in plasma endotoxin or fecal butyric acid

levels between HC, SS, and NASH patients.

NAFLD patients had higher fecal total SCFA, propionate, isobutyric acid, choline, and

trimethylamine, and serum 2-hydroxybutyrate than HC. Fecal choline and trimethylamine

and serum 2-hydroxybutyrate were positively correlated with BMI.

Metabolites including fecal formic acid, serum choline, and serum acetic acid were

correlated with histological features of NAFLD.

NASH patients had lower prevalence of C. leptum and higher amounts of E. coli in their

stool. No other differences existed for IM composition.

Reported energy intake was significantly lower in NASH patients than in HC and tended

to be higher in protein derived energy for SS and NASH patients than for HC.

Positive correlations were observed between substrates and products of bacterial

metabolism: protein with succinate and isovalerate, and sources of fermentable

carbohydrates with butyric and acetic acid.

Bacterial taxa were also correlated with substrates and in some cases, products of their

metabolic processes, including: C. leptum with sources of fermentable carbohydrates and

serum acetic acid, and Firmicutes with sources of fermentable carbohydrates, fecal

butyric acid, and serum acetic acid.

This study contributes to growing evidence suggesting the potential benefit of therapeutic

alteration of the IM in NAFLD prevention and treatment. Future research is necessary to further

evaluate the role of the IM and bacterial products in NAFLD pathogenesis and to investigate the

potential exploitation of this relationship for disease prevention and treatment.

82

9 Future Directions

There are several future research projects that could expand on the findings presented in this

thesis. Of primary interest would be the metagenomics analysis of the bacterial IM of these

patients. This data would allow us to fill in some of the gaps in the mechanistic information

coming from the current study. We would, for instance, be able to look at the gene expression for

carbohydrate fermentation and butyrate production to better understand if the higher relative

abundance of butyrate producers that we observed actually results in a higher butyrate

production. Current methods are limited by our incomplete understanding of the relevance of

fecal SCFA content in relation to SCFA production and absorption. Metagenomic studies would

also be able to determine if a higher E. coli count results in a greater production of ethanol or if

the overall metagenome results in differences in ethanol production between NASH patients and

HC. Planning for this type of study is underway.

Another important step to undertake in order to better understand the potential role of butyrate

would be to measure the abundance of butyrate producers, or ideally the bacterial gene

expression for butyrate production, while at the same time measuring fecal butyrate and

intestinal permeability/integrity. Similar studies, although not as comprehensive, have been able

to look at the role of butyrate producing probiotics with fecal butyrate and intestinal permeability

in mice, but this has not been examined in humans [154, 155]. Intestinal permeability can be

measured in a variety of ways in adults with NAFLD. Previous studies have used transmission

electron microscopy to observe microvilli and tight junctions or immunohistochemical analysis

of zona occludins-1 expression in intestinal biopsies [15, 134] or less invasive techniques

including the measurement of lactulose/mannitol or (51)Cr-ethylene diamine tetraacetate in urine

[132, 134].

Of particular interest in the role of bacterial metabolism of choline would be to look at the

expression of flavin monooxygenases in the liver, which is responsible for the conversion of

trimethylamine to TMAO. This may help us to better understand whether trimethylamine

metabolism after absorption is related to NAFLD disease status. Comparisons of fecal and serum

choline to phosphatidylcholine and expression of genes involved in the conversion of choline to

this active form, e.g. PEMT, would also be of interest since we did not see the expected lower

serum choline level in NAFLD patients, despite a higher production of trimethylamine.

83

A useful method for future studies would be to match NAFLD and HC by BMI. As BMI is so

closely tied to the NAFLD disease pathogenesis this will be a difficult task, but larger centers, or

multicenter trials may be able to accomplish this matching which would allow for a better

understanding of factors involved in NAFLD pathogenesis irrespective of weight.

The use of chemostat models, recently adapted to allow for the growth and manipulation of

human IM communities may be of particular use in future research [268]. Chemostats may be

particularly interesting when trialing the effects of potential pre or probiotic supplements or

dietary modifications as researchers can observe how these interventions alter IM and bacterial

metabolites directly. These basic science studies may then provide the data needed to plan for

larger human intervention trials with supplementation or potentially fecal transplantation. Fecal

transplantation has been successfully used for the treatment of Clostridium difficile infection

[269, 270] and was recently shown to improve insulin sensitivity [271].

84

References

1. LaBrecque, D.R., et al., World Gastroenterology Organisation global guidelines:

Nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. J Clin Gastroenterol,

2014. 48(6): p. 467-73.

2. Chalasani, N., et al., The diagnosis and management of non-alcoholic fatty liver disease:

Practice guideline by the American Association for the Study of Liver Diseases, American

College of Gastroenterology, and the American Gastroenterological Association. Am J

Gastroenterol, 2012. 107(6): p. 811-26.

3. Brunt, E.M., Pathology of nonalcoholic fatty liver disease. Nat Rev Gastroenterol

Hepatol, 2010. 7(4): p. 195-203.

4. Yu, A.S. and E.B. Keeffe, Nonalcoholic fatty liver disease. Rev Gastroenterol Disord,

2002. 2(1): p. 11-9.

5. Wanless, I.R. and J.S. Lentz, Fatty liver hepatitis (steatohepatitis) and obesity: an

autopsy study with analysis of risk factors. Hepatology, 1990. 12(5): p. 1106-10.

6. Festi, D., et al., Hepatic steatosis in obese patients: clinical aspects and prognostic

significance. Obes Rev, 2004. 5(1): p. 27-42.

7. Charlton, M.R., et al., Frequency and outcomes of liver transplantation for nonalcoholic

steatohepatitis in the United States. Gastroenterology, 2011. 141(4): p. 1249-53.

8. Gastrointestinal Surgery for Severe Obesity. NIH Consensus Statment Online, 1991. 9.

9. Vernon, G., A. Baranova, and Z.M. Younossi, Systematic review: the epidemiology and

natural history of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis in

adults. Aliment Pharmacol Ther, 2011. 34(3): p. 274-85.

10. Spencer, M.D., et al., Association between composition of the human gastrointestinal

microbiome and development of fatty liver with choline deficiency. Gastroenterology,

2011. 140(3): p. 976-86.

11. Zhu, L., et al., Characterization of gut microbiomes in nonalcoholic steatohepatitis

(NASH) patients: a connection between endogenous alcohol and NASH. Hepatology,

2013. 57(2): p. 601-9.

12. Raman, M., et al., Fecal microbiome and volatile organic compound metabolome in

obese humans with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol, 2013.

11(7): p. 868-75.e1-3.

13. Mouzaki, M., et al., Intestinal microbiota in patients with nonalcoholic fatty liver disease.

Hepatology, 2013. 58(1): p. 120-7.

85

14. Wong, V.W., et al., Molecular characterization of the fecal microbiota in patients with

nonalcoholic steatohepatitis--a longitudinal study. PLoS One, 2013. 8(4): p. e62885.

15. Jiang, W., et al., Dysbiosis gut microbiota associated with inflammation and impaired

mucosal immune function in intestine of humans with non-alcoholic fatty liver disease.

Sci Rep, 2015. 5: p. 8096.

16. Bugianesi, E., et al., Fibrosis in genotype 3 chronic hepatitis C and nonalcoholic fatty

liver disease: Role of insulin resistance and hepatic steatosis. Hepatology, 2006. 44(6): p.

1648-55.

17. Fong, D.G., et al., Metabolic and nutritional considerations in nonalcoholic fatty liver.

Hepatology, 2000. 32(1): p. 3-10.

18. Van Steenbergen, W. and S. Lanckmans, Liver disturbances in obesity and diabetes

mellitus. Int J Obes Relat Metab Disord, 1995. 19 Suppl 3: p. S27-36.

19. Fisher, R.L., Hepatobiliary abnormalities associated with total parenteral nutrition.

Gastroenterol Clin North Am, 1989. 18(3): p. 645-66.

20. Fabbrini, E., et al., Alterations in adipose tissue and hepatic lipid kinetics in obese men

and women with nonalcoholic fatty liver disease. Gastroenterology, 2008. 134(2): p. 424-

31.

21. Bradbury, M.W., Lipid metabolism and liver inflammation. I. Hepatic fatty acid uptake:

possible role in steatosis. Am J Physiol Gastrointest Liver Physiol, 2006. 290(2): p.

G194-8.

22. Donnelly, K.L., et al., Sources of fatty acids stored in liver and secreted via lipoproteins

in patients with nonalcoholic fatty liver disease. J Clin Invest, 2005. 115(5): p. 1343-51.

23. McGarry, J.D. and D.W. Foster, Regulation of hepatic fatty acid oxidation and ketone

body production. Annu Rev Biochem, 1980. 49: p. 395-420.

24. Angulo, P., Nonalcoholic fatty liver disease. N Engl J Med, 2002. 346(16): p. 1221-31.

25. Browning, J.D. and J.D. Horton, Molecular mediators of hepatic steatosis and liver

injury. J Clin Invest, 2004. 114(2): p. 147-52.

26. Monetti, M., et al., Dissociation of hepatic steatosis and insulin resistance in mice

overexpressing DGAT in the liver. Cell Metab, 2007. 6(1): p. 69-78.

27. McClain, C.J., S. Barve, and I. Deaciuc, Good fat/bad fat. Hepatology, 2007. 45(6): p.

1343-6.

28. Yamaguchi, K., et al., Inhibiting triglyceride synthesis improves hepatic steatosis but

exacerbates liver damage and fibrosis in obese mice with nonalcoholic steatohepatitis.

Hepatology, 2007. 45(6): p. 1366-74.

86

29. Baeuerle, P.A. and T. Henkel, Function and activation of NF-kappa B in the immune

system. Annu Rev Immunol, 1994. 12: p. 141-79.

30. Feldstein, A.E., et al., Free fatty acids promote hepatic lipotoxicity by stimulating TNF-

alpha expression via a lysosomal pathway. Hepatology, 2004. 40(1): p. 185-94.

31. Libermann, T.A. and D. Baltimore, Activation of interleukin-6 gene expression through

the NF-kappa B transcription factor. Mol Cell Biol, 1990. 10(5): p. 2327-34.

32. Farrell, G.C. and C.Z. Larter, Nonalcoholic fatty liver disease: from steatosis to cirrhosis.

Hepatology, 2006. 43(2 Suppl 1): p. S99-s112.

33. Sanyal, A.J., et al., Nonalcoholic steatohepatitis: association of insulin resistance and

mitochondrial abnormalities. Gastroenterology, 2001. 120(5): p. 1183-92.

34. Dowman, J.K., J.W. Tomlinson, and P.N. Newsome, Pathogenesis of non-alcoholic fatty

liver disease. Qjm, 2010. 103(2): p. 71-83.

35. Jou, J., S.S. Choi, and A.M. Diehl, Mechanisms of disease progression in nonalcoholic

fatty liver disease. Semin Liver Dis, 2008. 28(4): p. 370-9.

36. Malhi, H., et al., Free fatty acids induce JNK-dependent hepatocyte lipoapoptosis. J Biol

Chem, 2006. 281(17): p. 12093-101.

37. Feldstein, A.E., et al., Hepatocyte apoptosis and fas expression are prominent features of

human nonalcoholic steatohepatitis. Gastroenterology, 2003. 125(2): p. 437-43.

38. Susca, M., et al., Liver inflammatory cells, apoptosis, regeneration and stellate cell

activation in non-alcoholic steatohepatitis. Dig Liver Dis, 2001. 33(9): p. 768-77.

39. Ribeiro, P.S., et al., Hepatocyte apoptosis, expression of death receptors, and activation

of NF-kappaB in the liver of nonalcoholic and alcoholic steatohepatitis patients. Am J

Gastroenterol, 2004. 99(9): p. 1708-17.

40. Kong, J.Y. and S.W. Rabkin, Palmitate-induced apoptosis in cardiomyocytes is mediated

through alterations in mitochondria: prevention by cyclosporin A. Biochim Biophys

Acta, 2000. 1485(1): p. 45-55.

41. Wang, D., Y. Wei, and M.J. Pagliassotti, Saturated fatty acids promote endoplasmic

reticulum stress and liver injury in rats with hepatic steatosis. Endocrinology, 2006.

147(2): p. 943-51.

42. Mari, M., et al., Mitochondrial free cholesterol loading sensitizes to TNF- and Fas-

mediated steatohepatitis. Cell Metab, 2006. 4(3): p. 185-98.

43. Wei, Y., et al., Saturated fatty acids induce endoplasmic reticulum stress and apoptosis

independently of ceramide in liver cells. Am J Physiol Endocrinol Metab, 2006. 291(2):

p. E275-81.

87

44. Malhotra, J.D. and R.J. Kaufman, The endoplasmic reticulum and the unfolded protein

response. Semin Cell Dev Biol, 2007. 18(6): p. 716-31.

45. Ozcan, U., et al., Endoplasmic reticulum stress links obesity, insulin action, and type 2

diabetes. Science, 2004. 306(5695): p. 457-61.

46. Kantartzis, K., et al., Dissociation between fatty liver and insulin resistance in humans

carrying a variant of the patatin-like phospholipase 3 gene. Diabetes, 2009. 58(11): p.

2616-23.

47. Kotronen, A., et al., A common variant in PNPLA3, which encodes adiponutrin, is

associated with liver fat content in humans. Diabetologia, 2009. 52(6): p. 1056-60.

48. Sookoian, S. and C.J. Pirola, Meta-analysis of the influence of I148M variant of patatin-

like phospholipase domain containing 3 gene (PNPLA3) on the susceptibility and

histological severity of nonalcoholic fatty liver disease. Hepatology, 2011. 53(6): p.

1883-94.

49. Romeo, S., et al., Genetic variation in PNPLA3 confers susceptibility to nonalcoholic

fatty liver disease. Nat Genet, 2008. 40(12): p. 1461-5.

50. Valenti, L., et al., Homozygosity for the patatin-like phospholipase-3/adiponutrin I148M

polymorphism influences liver fibrosis in patients with nonalcoholic fatty liver disease.

Hepatology, 2010. 51(4): p. 1209-17.

51. Rotman, Y., et al., The association of genetic variability in patatin-like phospholipase

domain-containing protein 3 (PNPLA3) with histological severity of nonalcoholic fatty

liver disease. Hepatology, 2010. 52(3): p. 894-903.

52. Speliotes, E.K., et al., PNPLA3 variants specifically confer increased risk for histologic

nonalcoholic fatty liver disease but not metabolic disease. Hepatology, 2010. 52(3): p.

904-12.

53. Speliotes, E.K., et al., Genome-wide association analysis identifies variants associated

with nonalcoholic fatty liver disease that have distinct effects on metabolic traits. PLoS

Genet, 2011. 7(3): p. e1001324.

54. Petersen, K.F., et al., Apolipoprotein C3 gene variants in nonalcoholic fatty liver disease.

N Engl J Med, 2010. 362(12): p. 1082-9.

55. Perseghin, G., et al., Habitual physical activity is associated with intrahepatic fat content

in humans. Diabetes Care, 2007. 30(3): p. 683-8.

56. Krasnoff, J.B., et al., Health-related fitness and physical activity in patients with

nonalcoholic fatty liver disease. Hepatology, 2008. 47(4): p. 1158-66.

57. Zelber-Sagi, S., et al., Role of leisure-time physical activity in nonalcoholic fatty liver

disease: a population-based study. Hepatology, 2008. 48(6): p. 1791-8.

88

58. Kistler, K.D., et al., Physical activity recommendations, exercise intensity, and

histological severity of nonalcoholic fatty liver disease. Am J Gastroenterol, 2011.

106(3): p. 460-8; quiz 469.

59. Church, T.S., et al., Association of cardiorespiratory fitness, body mass index, and waist

circumference to nonalcoholic fatty liver disease. Gastroenterology, 2006. 130(7): p.

2023-30.

60. Da Silva, H.E., et al., A cross-sectional study assessing dietary intake and physical

activity in Canadian patients with nonalcoholic fatty liver disease vs healthy controls. J

Acad Nutr Diet, 2014. 114(8): p. 1181-94.

61. Anderson, E.L., et al., Childhood energy intake is associated with nonalcoholic Fatty

liver disease in adolescents. J Nutr, 2015. 145(5): p. 983-9.

62. Ferolla, S.M., et al., Dietary patterns in Brazilian patients with nonalcoholic fatty liver

disease: a cross-sectional study. Clinics (Sao Paulo), 2013. 68(1): p. 11-7.

63. Capristo, E., et al., Nutritional aspects in patients with non-alcoholic steatohepatitis

(NASH). Eur Rev Med Pharmacol Sci, 2005. 9(5): p. 265-8.

64. Dhurandhar, N.V., et al., Energy balance measurement: when something is not better

than nothing. Int J Obes (Lond), 2014.

65. Bedard, D., B. Shatenstein, and S. Nadon, Underreporting of energy intake from a self-

administered food-frequency questionnaire completed by adults in Montreal. Public

Health Nutr, 2004. 7(5): p. 675-81.

66. Vos, M.B., et al., Correlation of vitamin E, uric acid, and diet composition with

histologic features of pediatric NAFLD. J Pediatr Gastroenterol Nutr, 2012. 54(1): p. 90-

6.

67. Cankurtaran, M., et al., Serum vitamin-E levels and its relation to clinical features in

nonalcoholic fatty liver disease with elevated ALT levels. Acta Gastroenterol Belg, 2006.

69(1): p. 5-11.

68. Canbakan, B., et al., Clinical, biochemical and histological correlations in a group of

non-drinker subjects with non-alcoholic fatty liver disease. Acta Gastroenterol Belg,

2007. 70(3): p. 277-84.

69. Solga, S., et al., Dietary composition and nonalcoholic fatty liver disease. Dig Dis Sci,

2004. 49(10): p. 1578-83.

70. Ricci, G., et al., Nutrient intake in Italian obese patients: relationships with insulin

resistance and markers of non-alcoholic fatty liver disease. Nutrition, 2011. 27(6): p.

672-6.

71. Ma, J., et al., Sugar-sweetened beverage, diet soda, and fatty liver disease in the

Framingham Heart Study cohorts. J Hepatol, 2015.

89

72. Abid, A., et al., Soft drink consumption is associated with fatty liver disease independent

of metabolic syndrome. J Hepatol, 2009. 51(5): p. 918-24.

73. Mager, D.R., et al., Dietary and physical activity patterns in children with fatty liver. Eur

J Clin Nutr, 2010. 64(6): p. 628-35.

74. Ouyang, X., et al., Fructose consumption as a risk factor for non-alcoholic fatty liver

disease. J Hepatol, 2008. 48(6): p. 993-9.

75. Lim, J.S., et al., The role of fructose in the pathogenesis of NAFLD and the metabolic

syndrome. Nat Rev Gastroenterol Hepatol, 2010. 7(5): p. 251-64.

76. Morino, K., K.F. Petersen, and G.I. Shulman, Molecular mechanisms of insulin

resistance in humans and their potential links with mitochondrial dysfunction. Diabetes,

2006. 55 Suppl 2: p. S9-s15.

77. Wei, Y., et al., Fructose-mediated stress signaling in the liver: implications for hepatic

insulin resistance. J Nutr Biochem, 2007. 18(1): p. 1-9.

78. Musso, G., et al., Dietary habits and their relations to insulin resistance and postprandial

lipemia in nonalcoholic steatohepatitis. Hepatology, 2003. 37(4): p. 909-16.

79. Cummings, J.H. and G.T. Macfarlane, The control and consequences of bacterial

fermentation in the human colon. J Appl Bacteriol, 1991. 70(6): p. 443-59.

80. Solah, V.A., et al., Dose-response effect of a novel functional fibre, PolyGlycopleX((R)),

PGX((R)), on satiety. Appetite, 2014. 77: p. 72-6.

81. Pentikainen, S., et al., Enrichment of biscuits and juice with oat beta-glucan enhances

postprandial satiety. Appetite, 2014. 75: p. 150-6.

82. Blundell, J.E. and V.J. Burley, Satiation, satiety and the action of fibre on food intake. Int

J Obes, 1987. 11 Suppl 1: p. 9-25.

83. Bhupathiraju, S.N., et al., Glycemic index, glycemic load, and risk of type 2 diabetes:

results from 3 large US cohorts and an updated meta-analysis. Am J Clin Nutr, 2014.

100(1): p. 218-32.

84. Rocha, R., et al., [Non alcoholic fatty liver disease: treatment with soluble fibres]. Arq

Gastroenterol, 2007. 44(4): p. 350-2.

85. Chan, W.K., et al., Low physical activity and energy dense Malaysian foods are

associated with non-alcoholic fatty liver disease in centrally obese but not in non-

centrally obese patients with diabetes mellitus. Asia Pac J Clin Nutr, 2015. 24(2): p. 289-

98.

86. Mollard, R.C., et al., Dietary determinants of hepatic steatosis and visceral adiposity in

overweight and obese youth at risk of type 2 diabetes. Am J Clin Nutr, 2014. 99(4): p.

804-12.

90

87. Sathiaraj, E., et al., A case-control study on nutritional risk factors in non-alcoholic fatty

liver disease in Indian population. Eur J Clin Nutr, 2011. 65(4): p. 533-7.

88. Vilar, L., et al., High-fat diet: a trigger of non-alcoholic steatohepatitis? Preliminary

findings in obese subjects. Nutrition, 2008. 24(11-12): p. 1097-102.

89. Cortez-Pinto, H., et al., How different is the dietary pattern in non-alcoholic

steatohepatitis patients? Clin Nutr, 2006. 25(5): p. 816-23.

90. Charlton, M., et al., Fast food diet mouse: novel small animal model of NASH with

ballooning, progressive fibrosis, and high physiological fidelity to the human condition.

Am J Physiol Gastrointest Liver Physiol, 2011. 301(5): p. G825-34.

91. Chai, W. and Z. Liu, p38 mitogen-activated protein kinase mediates palmitate-induced

apoptosis but not inhibitor of nuclear factor-kappaB degradation in human coronary

artery endothelial cells. Endocrinology, 2007. 148(4): p. 1622-8.

92. Calder, P.C., n-3 polyunsaturated fatty acids, inflammation, and inflammatory diseases.

Am J Clin Nutr, 2006. 83(6 Suppl): p. 1505s-1519s.

93. Yasutake, K., et al., Nutritional investigation of non-obese patients with non-alcoholic

fatty liver disease: the significance of dietary cholesterol. Scand J Gastroenterol, 2009.

44(4): p. 471-7.

94. Oya, J., et al., Intake of n-3 polyunsaturated fatty acids and non-alcoholic fatty liver

disease: a cross-sectional study in Japanese men and women. Eur J Clin Nutr, 2010.

64(10): p. 1179-85.

95. Araya, J., et al., Increase in long-chain polyunsaturated fatty acid n - 6/n - 3 ratio in

relation to hepatic steatosis in patients with non-alcoholic fatty liver disease. Clin Sci

(Lond), 2004. 106(6): p. 635-43.

96. Puri, P., et al., A lipidomic analysis of nonalcoholic fatty liver disease. Hepatology, 2007.

46(4): p. 1081-90.

97. Arendt, B.M., et al., Altered hepatic gene expression in nonalcoholic fatty liver disease is

associated with lower hepatic n-3 and n-6 polyunsaturated fatty acids. Hepatology, 2015.

61(5): p. 1565-78.

98. Allard, J.P., et al., Nutritional assessment and hepatic fatty acid composition in non-

alcoholic fatty liver disease (NAFLD): a cross-sectional study. J Hepatol, 2008. 48(2): p.

300-7.

99. Clarke, S.D., Polyunsaturated fatty acid regulation of gene transcription: a molecular

mechanism to improve the metabolic syndrome. J Nutr, 2001. 131(4): p. 1129-32.

100. Nakamura, M.T., H.P. Cho, and S.D. Clarke, Regulation of hepatic delta-6 desaturase

expression and its role in the polyunsaturated fatty acid inhibition of fatty acid synthase

gene expression in mice. J Nutr, 2000. 130(6): p. 1561-5.

91

101. Sanyal, A.J., et al., Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis.

N Engl J Med, 2010. 362(18): p. 1675-85.

102. Mai, V. and P.V. Draganov, Recent advances and remaining gaps in our knowledge of

associations between gut microbiota and human health. World J Gastroenterol, 2009.

15(1): p. 81-5.

103. Frank, D.N., et al., Molecular-phylogenetic characterization of microbial community

imbalances in human inflammatory bowel diseases. Proc Natl Acad Sci U S A, 2007.

104(34): p. 13780-5.

104. Eckburg, P.B., et al., Diversity of the human intestinal microbial flora. Science, 2005.

308(5728): p. 1635-8.

105. Zhang, H., et al., Human gut microbiota in obesity and after gastric bypass. Proc Natl

Acad Sci U S A, 2009. 106(7): p. 2365-70.

106. Gosalbes, M.J., et al., Metatranscriptomic approach to analyze the functional human gut

microbiota. PLoS One, 2011. 6(3): p. e17447.

107. Arumugam, M., et al., Enterotypes of the human gut microbiome. Nature, 2011.

473(7346): p. 174-80.

108. Wu, G.D. and J.D. Lewis, Analysis of the human gut microbiome and association with

disease. Clin Gastroenterol Hepatol, 2013. 11(7): p. 774-7.

109. Backhed, F., et al., The gut microbiota as an environmental factor that regulates fat

storage. Proc Natl Acad Sci U S A, 2004. 101(44): p. 15718-23.

110. Stappenbeck, T.S., L.V. Hooper, and J.I. Gordon, Developmental regulation of intestinal

angiogenesis by indigenous microbes via Paneth cells. Proc Natl Acad Sci U S A, 2002.

99(24): p. 15451-5.

111. Turnbaugh, P.J., et al., An obesity-associated gut microbiome with increased capacity for

energy harvest. Nature, 2006. 444(7122): p. 1027-31.

112. Backhed, F., et al., Mechanisms underlying the resistance to diet-induced obesity in

germ-free mice. Proc Natl Acad Sci U S A, 2007. 104(3): p. 979-84.

113. Cani, P.D., et al., Changes in gut microbiota control metabolic endotoxemia-induced

inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes, 2008.

57(6): p. 1470-81.

114. Bergheim, I., et al., Antibiotics protect against fructose-induced hepatic lipid

accumulation in mice: role of endotoxin. J Hepatol, 2008. 48(6): p. 983-92.

115. Le Roy, T., et al., Intestinal microbiota determines development of non-alcoholic fatty

liver disease in mice. Gut, 2013. 62(12): p. 1787-94.

92

116. Ley, R.E., et al., Microbial ecology: human gut microbes associated with obesity. Nature,

2006. 444(7122): p. 1022-3.

117. Armougom, F., et al., Monitoring bacterial community of human gut microbiota reveals

an increase in Lactobacillus in obese patients and Methanogens in anorexic patients.

PLoS One, 2009. 4(9): p. e7125.

118. Schwiertz, A., et al., Microbiota and SCFA in lean and overweight healthy subjects.

Obesity (Silver Spring), 2010. 18(1): p. 190-5.

119. Turnbaugh, P.J., et al., A core gut microbiome in obese and lean twins. Nature, 2009.

457(7228): p. 480-4.

120. Furet, J.P., et al., Differential adaptation of human gut microbiota to bariatric surgery-

induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes,

2010. 59(12): p. 3049-57.

121. Jumpertz, R., et al., Energy-balance studies reveal associations between gut microbes,

caloric load, and nutrient absorption in humans. Am J Clin Nutr, 2011. 94(1): p. 58-65.

122. Gregor, M.F. and G.S. Hotamisligil, Inflammatory mechanisms in obesity. Annu Rev

Immunol, 2011. 29: p. 415-45.

123. Cai, D., et al., Local and systemic insulin resistance resulting from hepatic activation of

IKK-beta and NF-kappaB. Nat Med, 2005. 11(2): p. 183-90.

124. Song, M.J., et al., Activation of Toll-like receptor 4 is associated with insulin resistance

in adipocytes. Biochem Biophys Res Commun, 2006. 346(3): p. 739-45.

125. Rivera, C.A., et al., Toll-like receptor-4 signaling and Kupffer cells play pivotal roles in

the pathogenesis of non-alcoholic steatohepatitis. J Hepatol, 2007. 47(4): p. 571-9.

126. Miyake, K., Innate immune sensing of pathogens and danger signals by cell surface Toll-

like receptors. Semin Immunol, 2007. 19(1): p. 3-10.

127. Cani, P.D., et al., Metabolic endotoxemia initiates obesity and insulin resistance.

Diabetes, 2007. 56(7): p. 1761-72.

128. Brun, P., et al., Increased intestinal permeability in obese mice: new evidence in the

pathogenesis of nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol,

2007. 292(2): p. G518-25.

129. Thuy, S., et al., Nonalcoholic fatty liver disease in humans is associated with increased

plasma endotoxin and plasminogen activator inhibitor 1 concentrations and with fructose

intake. J Nutr, 2008. 138(8): p. 1452-5.

130. Harte, A.L., et al., Elevated endotoxin levels in non-alcoholic fatty liver disease. J

Inflamm (Lond), 2010. 7: p. 15.

93

131. Verdam, F.J., et al., Novel evidence for chronic exposure to endotoxin in human

nonalcoholic steatohepatitis. J Clin Gastroenterol, 2011. 45(2): p. 149-52.

132. Volynets, V., et al., Nutrition, intestinal permeability, and blood ethanol levels are

altered in patients with nonalcoholic fatty liver disease (NAFLD). Dig Dis Sci, 2012.

57(7): p. 1932-41.

133. Spruss, A., et al., Toll-like receptor 4 is involved in the development of fructose-induced

hepatic steatosis in mice. Hepatology, 2009. 50(4): p. 1094-104.

134. Miele, L., et al., Increased intestinal permeability and tight junction alterations in

nonalcoholic fatty liver disease. Hepatology, 2009. 49(6): p. 1877-87.

135. Bergman, E.N., Energy contributions of volatile fatty acids from the gastrointestinal tract

in various species. Physiol Rev, 1990. 70(2): p. 567-90.

136. Parker, D.S., The measurement of production rates of volatile fatty acids in the caecum of

the conscious rabbit. Br J Nutr, 1976. 36(1): p. 61-70.

137. Topping, D.L. and P.M. Clifton, Short-chain fatty acids and human colonic function:

roles of resistant starch and nonstarch polysaccharides. Physiol Rev, 2001. 81(3): p.

1031-64.

138. Cummings, J.H., et al., The effect of meat protein and dietary fiber on colonic function

and metabolism. II. Bacterial metabolites in feces and urine. Am J Clin Nutr, 1979.

32(10): p. 2094-101.

139. Hijova, E. and A. Chmelarova, Short chain fatty acids and colonic health. Bratisl Lek

Listy, 2007. 108(8): p. 354-8.

140. Binder, H.J., Role of colonic short-chain fatty acid transport in diarrhea. Annu Rev

Physiol, 2010. 72: p. 297-313.

141. Lin, H.V., et al., Butyrate and propionate protect against diet-induced obesity and

regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One,

2012. 7(4): p. e35240.

142. Gao, Z., et al., Butyrate improves insulin sensitivity and increases energy expenditure in

mice. Diabetes, 2009. 58(7): p. 1509-17.

143. Roediger, W.E., Role of anaerobic bacteria in the metabolic welfare of the colonic

mucosa in man. Gut, 1980. 21(9): p. 793-8.

144. Roediger, W.E., Utilization of nutrients by isolated epithelial cells of the rat colon.

Gastroenterology, 1982. 83(2): p. 424-9.

145. Mariadason, J.M., D.H. Barkla, and P.R. Gibson, Effect of short-chain fatty acids on

paracellular permeability in Caco-2 intestinal epithelium model. Am J Physiol, 1997.

272(4 Pt 1): p. G705-12.

94

146. Peng, L., et al., Effects of butyrate on intestinal barrier function in a Caco-2 cell

monolayer model of intestinal barrier. Pediatr Res, 2007. 61(1): p. 37-41.

147. Peng, L., et al., Butyrate enhances the intestinal barrier by facilitating tight junction

assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J

Nutr, 2009. 139(9): p. 1619-25.

148. Hinnebusch, B.F., et al., The effects of short-chain fatty acids on human colon cancer cell

phenotype are associated with histone hyperacetylation. J Nutr, 2002. 132(5): p. 1012-7.

149. Comalada, M., et al., The effects of short-chain fatty acids on colon epithelial

proliferation and survival depend on the cellular phenotype. J Cancer Res Clin Oncol,

2006. 132(8): p. 487-97.

150. Inan, M.S., et al., The luminal short-chain fatty acid butyrate modulates NF-kappaB

activity in a human colonic epithelial cell line. Gastroenterology, 2000. 118(4): p. 724-

34.

151. Yin, L., G. Laevsky, and C. Giardina, Butyrate suppression of colonocyte NF-kappa B

activation and cellular proteasome activity. J Biol Chem, 2001. 276(48): p. 44641-6.

152. Hamer, H.M., et al., Butyrate modulates oxidative stress in the colonic mucosa of healthy

humans. Clin Nutr, 2009. 28(1): p. 88-93.

153. De Vuyst, L. and F. Leroy, Cross-feeding between bifidobacteria and butyrate-producing

colon bacteria explains bifdobacterial competitiveness, butyrate production, and gas

production. Int J Food Microbiol, 2011. 149(1): p. 73-80.

154. Endo, H., et al., Butyrate-producing probiotics reduce nonalcoholic fatty liver disease

progression in rats: new insight into the probiotics for the gut-liver axis. PLoS One,

2013. 8(5): p. e63388.

155. Yadav, H., et al., Beneficial metabolic effects of a probiotic via butyrate-induced GLP-1

hormone secretion. J Biol Chem, 2013. 288(35): p. 25088-97.

156. Puertollano, E., S. Kolida, and P. Yaqoob, Biological significance of short-chain fatty

acid metabolism by the intestinal microbiome. Curr Opin Clin Nutr Metab Care, 2014.

17(2): p. 139-44.

157. Cummings, J.H., et al., Short chain fatty acids in human large intestine, portal, hepatic

and venous blood. Gut, 1987. 28(10): p. 1221-7.

158. Bloemen, J.G., et al., Short chain fatty acids exchange across the gut and liver in humans

measured at surgery. Clin Nutr, 2009. 28(6): p. 657-61.

159. Hong, Y.H., et al., Acetate and propionate short chain fatty acids stimulate adipogenesis

via GPCR43. Endocrinology, 2005. 146(12): p. 5092-9.

95

160. Ge, H., et al., Activation of G protein-coupled receptor 43 in adipocytes leads to

inhibition of lipolysis and suppression of plasma free fatty acids. Endocrinology, 2008.

149(9): p. 4519-26.

161. Demigne, C., et al., Effect of propionate on fatty acid and cholesterol synthesis and on

acetate metabolism in isolated rat hepatocytes. Br J Nutr, 1995. 74(2): p. 209-19.

162. Nishina, P.M. and R.A. Freedland, Effects of propionate on lipid biosynthesis in isolated

rat hepatocytes. J Nutr, 1990. 120(7): p. 668-73.

163. Bos, C.L., et al., Prostanoids and prostanoid receptors in signal transduction. Int J

Biochem Cell Biol, 2004. 36(7): p. 1187-205.

164. Al-Lahham, S.H., et al., Regulation of adipokine production in human adipose tissue by

propionic acid. Eur J Clin Invest, 2010. 40(5): p. 401-7.

165. Tedelind, S., et al., Anti-inflammatory properties of the short-chain fatty acids acetate

and propionate: a study with relevance to inflammatory bowel disease. World J

Gastroenterol, 2007. 13(20): p. 2826-32.

166. Xiong, Y., et al., Short-chain fatty acids stimulate leptin production in adipocytes

through the G protein-coupled receptor GPR41. Proc Natl Acad Sci U S A, 2004. 101(4):

p. 1045-50.

167. Al-Lahham, S.H., et al., Biological effects of propionic acid in humans; metabolism,

potential applications and underlying mechanisms. Biochim Biophys Acta, 2010.

1801(11): p. 1175-83.

168. den Besten, G., et al., The role of short-chain fatty acids in the interplay between diet, gut

microbiota, and host energy metabolism. J Lipid Res, 2013. 54(9): p. 2325-40.

169. Maslowski, K.M., et al., Regulation of inflammatory responses by gut microbiota and

chemoattractant receptor GPR43. Nature, 2009. 461(7268): p. 1282-6.

170. Fukuda, S., et al., Bifidobacteria can protect from enteropathogenic infection through

production of acetate. Nature, 2011. 469(7331): p. 543-7.

171. Blomstrand, R., Observations of the formation of ethanol in the intestinal tract in man.

Life Sci II, 1971. 10(10): p. 575-82.

172. Krebs, H.A. and J.R. Perkins, The physiological role of liver alcohol dehydrogenase.

Biochem J, 1970. 118(4): p. 635-44.

173. Ferrier, L., et al., Impairment of the intestinal barrier by ethanol involves enteric

microflora and mast cell activation in rodents. Am J Pathol, 2006. 168(4): p. 1148-54.

174. Noga, A.A. and D.E. Vance, A gender-specific role for phosphatidylethanolamine N-

methyltransferase-derived phosphatidylcholine in the regulation of plasma high density

and very low density lipoproteins in mice. J Biol Chem, 2003. 278(24): p. 21851-9.

96

175. Guo, W.X., et al., Mitochondrial dysfunction in choline deficiency-induced apoptosis in

cultured rat hepatocytes. Free Radic Biol Med, 2005. 39(5): p. 641-50.

176. Teodoro, J.S., et al., Differential alterations in mitochondrial function induced by a

choline-deficient diet: understanding fatty liver disease progression. Mitochondrion,

2008. 8(5-6): p. 367-76.

177. Pacelli, C., et al., Dietary choline deprivation impairs rat brain mitochondrial function

and behavioral phenotype. J Nutr, 2010. 140(6): p. 1072-9.

178. Soon, R.K., Jr., et al., Stress signaling in the methionine-choline-deficient model of

murine fatty liver disease. Gastroenterology, 2010. 139(5): p. 1730-9, 1739.e1.

179. Dumas, M.E., et al., Metabolic profiling reveals a contribution of gut microbiota to fatty

liver phenotype in insulin-resistant mice. Proc Natl Acad Sci U S A, 2006. 103(33): p.

12511-6.

180. Arendt, B.M., et al., Nonalcoholic fatty liver disease is associated with lower hepatic and

erythrocyte ratios of phosphatidylcholine to phosphatidylethanolamine. Appl Physiol

Nutr Metab, 2013. 38(3): p. 334-40.

181. Song, J., et al., Polymorphism of the PEMT gene and susceptibility to nonalcoholic fatty

liver disease (NAFLD). Faseb j, 2005. 19(10): p. 1266-71.

182. Li, Z., et al., Probiotics and antibodies to TNF inhibit inflammatory activity and improve

nonalcoholic fatty liver disease. Hepatology, 2003. 37(2): p. 343-50.

183. Seo, M., et al., Clostridium butyricum MIYAIRI 588 improves high-fat diet-induced non-

alcoholic fatty liver disease in rats. Dig Dis Sci, 2013. 58(12): p. 3534-44.

184. Mencarelli, A., et al., VSL#3 resets insulin signaling and protects against NASH and

atherosclerosis in a model of genetic dyslipidemia and intestinal inflammation. PLoS

One, 2012. 7(9): p. e45425.

185. Daubioul, C.A., et al., Dietary oligofructose lessens hepatic steatosis, but does not

prevent hypertriglyceridemia in obese zucker rats. J Nutr, 2000. 130(5): p. 1314-9.

186. Alisi, A., et al., Randomised clinical trial: The beneficial effects of VSL#3 in obese

children with non-alcoholic steatohepatitis. Aliment Pharmacol Ther, 2014. 39(11): p.

1276-85.

187. Loguercio, C., et al., Beneficial effects of a probiotic VSL#3 on parameters of liver

dysfunction in chronic liver diseases. J Clin Gastroenterol, 2005. 39(6): p. 540-3.

188. Aller, R., et al., Effect of a probiotic on liver aminotransferases in nonalcoholic fatty liver

disease patients: a double blind randomized clinical trial. Eur Rev Med Pharmacol Sci,

2011. 15(9): p. 1090-5.

97

189. Vajro, P., et al., Effects of Lactobacillus rhamnosus strain GG in pediatric obesity-related

liver disease. J Pediatr Gastroenterol Nutr, 2011. 52(6): p. 740-3.

190. Malaguarnera, M., et al., Bifidobacterium longum with fructo-oligosaccharides in

patients with non alcoholic steatohepatitis. Dig Dis Sci, 2012. 57(2): p. 545-53.

191. Mohammed, S.S., et al., HIV-positive patients with nonalcoholic fatty liver disease have

a lower body mass index and are more physically active than HIV-negative patients. J

Acquir Immune Defic Syndr, 2007. 45(4): p. 432-8.

192. Madill, J., et al., Oxidative stress and nutritional factors in hepatitis C virus-positive liver

recipients, controls, and hepatitis C virus-positive nontransplant patients. Transplant

Proc, 2010. 42(5): p. 1744-9.

193. Pan, X.R., et al., Effects of diet and exercise in preventing NIDDM in people with

impaired glucose tolerance. The Da Qing IGT and Diabetes Study. Diabetes Care, 1997.

20(4): p. 537-44.

194. Roza, A.M. and H.M. Shizgal, The Harris Benedict equation reevaluated: resting energy

requirements and the body cell mass. Am J Clin Nutr, 1984. 40(1): p. 168-82.

195. Matthews, D.R., et al., Homeostasis model assessment: insulin resistance and beta-cell

function from fasting plasma glucose and insulin concentrations in man. Diabetologia,

1985. 28(7): p. 412-9.

196. Brunt, E.M., et al., Nonalcoholic steatohepatitis: a proposal for grading and staging the

histological lesions. Am J Gastroenterol, 1999. 94(9): p. 2467-74.

197. Kleiner, D.E., et al., Design and validation of a histological scoring system for

nonalcoholic fatty liver disease. Hepatology, 2005. 41(6): p. 1313-21.

198. Dehghan, P., et al., Inulin controls inflammation and metabolic endotoxemia in women

with type 2 diabetes mellitus: a randomized-controlled clinical trial. Int J Food Sci Nutr,

2014. 65(1): p. 117-23.

199. Clemente-Postigo, M., et al., Endotoxin increase after fat overload is related to

postprandial hypertriglyceridemia in morbidly obese patients. J Lipid Res, 2012. 53(5):

p. 973-8.

200. Monte, S.V., et al., Reduction in endotoxemia, oxidative and inflammatory stress, and

insulin resistance after Roux-en-Y gastric bypass surgery in patients with morbid obesity

and type 2 diabetes mellitus. Surgery, 2012. 151(4): p. 587-93.

201. Basu, S., et al., Pregravid obesity associates with increased maternal endotoxemia and

metabolic inflammation. Obesity (Silver Spring), 2011. 19(3): p. 476-82.

202. Psychogios, N., et al., The human serum metabolome. PLoS One, 2011. 6(2): p. e16957.

98

203. Saude, E.J., C.M. Slumpsky, and B.D. Sykes, Optimization of NMR analysis of biological

fluids for quantitative accuracy. Metabolomics, 2006(2): p. 113-123.

204. Wishart, D.S., et al., The human cerebrospinal fluid metabolome. J Chromatogr B Analyt

Technol Biomed Life Sci, 2008. 871(2): p. 164-73.

205. Qin, J., et al., A human gut microbial gene catalogue established by metagenomic

sequencing. Nature, 2010. 464(7285): p. 59-65.

206. Le Gall, G., et al., Metabolomics of fecal extracts detects altered metabolic activity of gut

microbiota in ulcerative colitis and irritable bowel syndrome. J Proteome Res, 2011.

10(9): p. 4208-18.

207. Furet, J.P., et al., Comparative assessment of human and farm animal faecal microbiota

using real-time quantitative PCR. FEMS Microbiol Ecol, 2009. 68(3): p. 351-62.

208. Suzuki, M.T., L.T. Taylor, and E.F. DeLong, Quantitative analysis of small-subunit

rRNA genes in mixed microbial populations via 5'-nuclease assays. Appl Environ

Microbiol, 2000. 66(11): p. 4605-14.

209. Haarman, M. and J. Knol, Quantitative real-time PCR analysis of fecal Lactobacillus

species in infants receiving a prebiotic infant formula. Appl Environ Microbiol, 2006.

72(4): p. 2359-65.

210. Trumbo, P., et al., Dietary reference intakes for energy, carbohydrate, fiber, fat, fatty

acids, cholesterol, protein and amino acids. J Am Diet Assoc, 2002. 102(11): p. 1621-30.

211. Krauss, R.M., et al., Revision 2000: a statement for healthcare professionals from the

Nutrition Committee of the American Heart Association. J Nutr, 2001. 131(1): p. 132-46.

212. Institute of Medicine Standing Committee on the Scientific Evaluation of Dietary

Reference, I., The National Academies Collection: Reports funded by National Institutes

of Health, in Dietary Reference Intakes for Calcium, Phosphorus, Magnesium, Vitamin

D, and Fluoride. 1997, National Academies Press (US). National Academy of Sciences.:

Washington (DC).

213. Institute of Medicine Standing Committee on the Scientific Evaluation of Dietary

Reference, I., O.B.V. its Panel on Folate, and Choline, The National Academies

Collection: Reports funded by National Institutes of Health, in Dietary Reference Intakes

for Thiamin, Riboflavin, Niacin, Vitamin B6, Folate, Vitamin B12, Pantothenic Acid,

Biotin, and Choline. 1998, National Academies Press (US). National Academy of

Sciences.: Washington (DC).

214. Institute of Medicine Panel on, M., in Dietary Reference Intakes for Vitamin A, Vitamin

K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel,

Silicon, Vanadium, and Zinc. 2001, National Academies Press (US). Copyright 2001 by

the National Academy of Sciences. All rights reserved.: Washington (DC).

99

215. Medicine, I.o., Dietary Reference Intakes for Water, Potassium, Sodium, Chloride, and

Sulfate. 2004, Washington, DC: National Academies Press.

216. Institute of Medicine Panel on Dietary, A. and C. Related, in Dietary Reference Intakes

for Vitamin C, Vitamin E, Selenium, and Carotenoids. 2000, National Academies Press

(US). Copyright 2000 by the National Academy of Sciences. All rights reserved.:

Washington (DC).

217. Wong, V.W., et al., Bacterial endotoxin and non-alcoholic fatty liver disease in the

general population: a prospective cohort study. Aliment Pharmacol Ther, 2015.

218. Serino, M., et al., Metabolic adaptation to a high-fat diet is associated with a change in

the gut microbiota. Gut, 2012. 61(4): p. 543-53.

219. de La Serre, C.B., et al., Propensity to high-fat diet-induced obesity in rats is associated

with changes in the gut microbiota and gut inflammation. Am J Physiol Gastrointest

Liver Physiol, 2010. 299(2): p. G440-8.

220. Amar, J., et al., Energy intake is associated with endotoxemia in apparently healthy men.

Am J Clin Nutr, 2008. 87(5): p. 1219-23.

221. Miller, M.A., et al., Ethnic and sex differences in circulating endotoxin levels: A novel

marker of atherosclerotic and cardiovascular risk in a British multi-ethnic population.

Atherosclerosis, 2009. 203(2): p. 494-502.

222. Dixon, A.N., et al., Effect of the orlistat on serum endotoxin lipopolysaccharide and

adipocytokines in South Asian individuals with impaired glucose tolerance. Int J Clin

Pract, 2008. 62(7): p. 1124-9.

223. Vogt, J.A. and T.M. Wolever, Fecal acetate is inversely related to acetate absorption

from the human rectum and distal colon. J Nutr, 2003. 133(10): p. 3145-8.

224. Macfarlane, S. and G.T. Macfarlane, Regulation of short-chain fatty acid production.

Proc Nutr Soc, 2003. 62(1): p. 67-72.

225. Fernandes, J., et al., Adiposity, gut microbiota and faecal short chain fatty acids are

linked in adult humans. Nutr Diabetes, 2014. 4: p. e121.

226. den Besten, G., et al., Gut-derived short-chain fatty acids are vividly assimilated into host

carbohydrates and lipids. Am J Physiol Gastrointest Liver Physiol, 2013. 305(12): p.

G900-10.

227. Isobutyric Acid. National Center for Biotechnology Information. PubChem Compound

Database; CID=6590, https://pubchem.ncbi.nlm.nih.gov/compound/6590 (accessed July

20, 2015).

228. 2-Hydroxybutyric acid. HMDB Version 3.6.,

http://www.hmdb.ca/metabolites/hmdb00008 (accessed July 20, 2015).

100

229. Doelle, H.W., Bacterial Metabolism. 1969, New York: Academic Press, Inc.

230. Li, X., et al., Comprehensive two-dimensional gas chromatography/time-of-flight mass

spectrometry for metabonomics: Biomarker discovery for diabetes mellitus. Anal Chim

Acta, 2009. 633(2): p. 257-62.

231. Gall, W.E., et al., alpha-hydroxybutyrate is an early biomarker of insulin resistance and

glucose intolerance in a nondiabetic population. PLoS One, 2010. 5(5): p. e10883.

232. Schicho, R., et al., Quantitative metabolomic profiling of serum, plasma, and urine by

(1)H NMR spectroscopy discriminates between patients with inflammatory bowel disease

and healthy individuals. J Proteome Res, 2012. 11(6): p. 3344-57.

233. Nehra, V., et al., Nutritional and metabolic considerations in the etiology of nonalcoholic

steatohepatitis. Dig Dis Sci, 2001. 46(11): p. 2347-52.

234. Baker, J.R. and S. Chaykin, The biosynthesis of trimethylamine-N-oxide. J Biol Chem,

1962. 237: p. 1309-13.

235. Messenger, J., et al., A review of trimethylaminuria: (fish odor syndrome). J Clin Aesthet

Dermatol, 2013. 6(11): p. 45-8.

236. Tang, W.H., et al., Intestinal microbial metabolism of phosphatidylcholine and

cardiovascular risk. N Engl J Med, 2013. 368(17): p. 1575-84.

237. Tang, W.H., et al., Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway

contributes to both development of renal insufficiency and mortality risk in chronic

kidney disease. Circ Res, 2015. 116(3): p. 448-55.

238. Bae, S., et al., Plasma choline metabolites and colorectal cancer risk in the Women's

Health Initiative Observational Study. Cancer Res, 2014. 74(24): p. 7442-52.

239. Athyros, V.G., et al., Cardiovascular risk across the histological spectrum and the

clinical manifestations of non-alcoholic fatty liver disease: An update. World J

Gastroenterol, 2015. 21(22): p. 6820-6834.

240. Tabakoff, B., N. Cornell, and P.L. Hoffman, Alcohol tolerance. Ann Emerg Med, 1986.

15(9): p. 1005-12.

241. Zakhari, S., Overview: how is alcohol metabolized by the body? Alcohol Res Health,

2006. 29(4): p. 245-54.

242. Sghir, A., et al., Quantification of bacterial groups within human fecal flora by

oligonucleotide probe hybridization. Appl Environ Microbiol, 2000. 66(5): p. 2263-6.

243. Lay, C., et al., Colonic microbiota signatures across five northern European countries.

Appl Environ Microbiol, 2005. 71(7): p. 4153-5.

101

244. Sokol, H., et al., Faecalibacterium prausnitzii is an anti-inflammatory commensal

bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl

Acad Sci U S A, 2008. 105(43): p. 16731-6.

245. Bahl, M.I., A. Bergstrom, and T.R. Licht, Freezing fecal samples prior to DNA extraction

affects the Firmicutes to Bacteroidetes ratio determined by downstream quantitative PCR

analysis. FEMS Microbiol Lett, 2012. 329(2): p. 193-7.

246. Basiotis, P.P., et al., Number of days of food intake records required to estimate

individual and group nutrient intakes with defined confidence. J Nutr, 1987. 117(9): p.

1638-41.

247. Park, H.A., J.S. Lee, and L.H. Kuller, Underreporting of dietary intake by body mass

index in premenopausal women participating in the Healthy Women Study. Nutr Res

Pract, 2007. 1(3): p. 231-6.

248. Mendez, M.A., et al., Under- and overreporting of energy is related to obesity, lifestyle

factors and food group intakes in Jamaican adults. Public Health Nutr, 2004. 7(1): p. 9-

19.

249. Hill, R.J. and P.S. Davies, The validity of self-reported energy intake as determined using

the doubly labelled water technique. Br J Nutr, 2001. 85(4): p. 415-30.

250. York, L.W., S. Puthalapattu, and G.Y. Wu, Nonalcoholic fatty liver disease and low-

carbohydrate diets. Annu Rev Nutr, 2009. 29: p. 365-79.

251. Gill, H.K. and G.Y. Wu, Non-alcoholic fatty liver disease and the metabolic syndrome:

effects of weight loss and a review of popular diets. Are low carbohydrate diets the

answer? World J Gastroenterol, 2006. 12(3): p. 345-53.

252. Thoma, C., C.P. Day, and M.I. Trenell, Lifestyle interventions for the treatment of non-

alcoholic fatty liver disease in adults: a systematic review. J Hepatol, 2012. 56(1): p. 255-

66.

253. Warner, E.T., et al., Differential accuracy of physical activity self-report by body mass

index. Am J Health Behav, 2012. 36(2): p. 168-78.

254. Brown, B.B. and C.M. Werner, Using accelerometer feedback to identify walking

destinations, activity overestimates, and stealth exercise in obese and nonobese

individuals. J Phys Act Health, 2008. 5(6): p. 882-93.

255. Clarke, S.F., et al., Exercise and associated dietary extremes impact on gut microbial

diversity. Gut, 2014. 63(12): p. 1913-20.

256. Madsen, J.L. and J. Graff, Effects of ageing on gastrointestinal motor function. Age

Ageing, 2004. 33(2): p. 154-9.

257. Madsen, J.L., Effects of gender, age, and body mass index on gastrointestinal transit

times. Dig Dis Sci, 1992. 37(10): p. 1548-53.

102

258. Orr, W.C. and C.L. Chen, Aging and neural control of the GI tract: IV. Clinical and

physiological aspects of gastrointestinal motility and aging. Am J Physiol Gastrointest

Liver Physiol, 2002. 283(6): p. G1226-31.

259. Metcalf, A.M., et al., Simplified assessment of segmental colonic transit.

Gastroenterology, 1987. 92(1): p. 40-7.

260. Lakshminarayanan, B., et al., Compositional dynamics of the human intestinal microbiota

with aging: implications for health. J Nutr Health Aging, 2014. 18(9): p. 773-86.

261. De Filippo, C., et al., Impact of diet in shaping gut microbiota revealed by a comparative

study in children from Europe and rural Africa. Proc Natl Acad Sci U S A, 2010.

107(33): p. 14691-6.

262. Lin, A., et al., Distinct distal gut microbiome diversity and composition in healthy

children from Bangladesh and the United States. PLoS One, 2013. 8(1): p. e53838.

263. Nam, Y.D., et al., Comparative analysis of Korean human gut microbiota by barcoded

pyrosequencing. PLoS One, 2011. 6(7): p. e22109.

264. Yatsunenko, T., et al., Human gut microbiome viewed across age and geography. Nature,

2012. 486(7402): p. 222-7.

265. National High Blood Pressure Education, P., in The Seventh Report of the Joint National

Committee on Prevention, Detection, Evaluation, and Treatment of High Blood Pressure.

2004, National Heart, Lung, and Blood Institute (US): Bethesda (MD).

266. Poynard, T., et al., Validation of liver fibrosis biomarker (FibroTest) for assessing liver

fibrosis progression: proof of concept and first application in a large population. J

Hepatol, 2012. 57(3): p. 541-8.

267. Pathik, P., et al., Fibroscan versus simple noninvasive screening tools in predicting

fibrosis in high-risk nonalcoholic fatty liver disease patients from Western India. Ann

Gastroenterol, 2015. 28(2): p. 281-286.

268. McDonald, J.A., et al., Evaluation of microbial community reproducibility, stability and

composition in a human distal gut chemostat model. J Microbiol Methods, 2013. 95(2): p.

167-74.

269. Cammarota, G., et al., Randomised clinical trial: faecal microbiota transplantation by

colonoscopy vs. vancomycin for the treatment of recurrent Clostridium difficile infection.

Aliment Pharmacol Ther, 2015. 41(9): p. 835-43.

270. Stollman, N., et al., Frozen encapsulated stool in recurrent Clostridium difficile:

exploring the role of pills in the treatment hierarchy of fecal microbiota transplant

nonresponders. Am J Gastroenterol, 2015. 110(4): p. 600-1.

103

271. Vrieze, A., et al., Transfer of intestinal microbiota from lean donors increases insulin

sensitivity in individuals with metabolic syndrome. Gastroenterology, 2012. 143(4): p.

913-6.e7.

104

Appendices

APPENDIX 1

Patient Information Sheet & Consent Form

Study Title: Non-alcoholic Steato-hepatitis (NASH) Versus Simple Hepatic Steatosis: Is

There a Difference in the Nutritional Factors Influencing Lipid Peroxidation and

Inflammation?

PRINCIPAL INVESTIGATOR: Dr. Johane P. Allard Tel: (416) 340-5159

SPONSOR: Canadian Liver Foundation & Canadian Institutes of Health Research

Introduction

You were referred to a liver specialist because of elevated liver enzymes and/or fat infiltration in

your liver detected by blood work or abdominal ultrasound. As part of standard medical care, a

hepatologist (liver specialist) ordered a liver biopsy to make a diagnosis. A biopsy consists of a

very small sample of your liver taken with a needle. Prior to having the liver biopsy, a research

study coordinator will contact you to ask you if you would be interested in participating in a

research study. The only way to determine if you are eligible for the study is to examine the

sample of tissue taken from your liver biopsy, but, we first need your permission to be able to

look at the tissue.

If you agree to allowing us to look at a portion of the liver biopsy, and if the tissue confirms the

diagnosis of liver steatosis (fat in the liver) or non-alcoholic steatohepatitis (fat with

inflammation) and no other causes are found or you have a completely normal liver biopsy

result, the research coordinator will contact you to see if you agree to have further assessments.

It is important that you understand the purpose, procedures, benefits, discomfort, risks and

precautions associated with the study before you agree to participate, so that you can make an

informed decision. This is known as the informed consent process. If there is anything in this

form that you do not understand, please consult the study coordinator. Make sure all your

questions have been answered to your satisfaction before signing this document.

Of course, you have the right to refuse to participate and also the right to withdraw from the

study at any time.

Purpose of the Study

Fatty infiltration of the liver can be associated with alcohol, obesity, diabetes and abnormal fat

and sugar handling by your body. When alcohol intake is excluded, this is called a “Non-

Alcoholic Fatty Liver” or “NAFLD”. If it is associated with inflammation (cell damage), it is

called “Non-Alcoholic Steato-Hepatitis” or “NASH”. No one knows why some patients with

excess fat in their liver remain stable while others progress to a more advanced disease like

cirrhosis.

Therefore, the purpose of this study is to determine if nutritional factors measured in your blood

and diet or the kind of bacteria that are found in the intestinal tract can have an effect on liver

tissue damage, the type of fat deposited in the liver and also the genes involved in fat and sugar

metabolism. We are also measuring bacterial products in stool and blood samples.

105

About 190 patients with liver-biopsy proven simple steatosis (fat infiltration), NASH (fat with

inflammation) or people with minimal findings on liver biopsy will participate in this study at

The Toronto General Hospital and The Toronto Western Hospital. For the purpose of this study,

nutritional assessment and blood work will be done only in one occasion, and we will ask the

participants for one single stool sample. Fifty patients with healthy liver will serve as a control

group.

Study Procedures

If you consent to participate in this study, the following study procedures are to be performed at

each study visit.

First Visit: (Approximately 30 minutes)

This visit is the present clinical appointment with your liver specialist where the decision is made

to do the liver biopsy. You are now presented with the consent form for the study and meet with

the study coordinator who explains the study and ask you to sign the consent. You will be

instructed to record your food intake and physical activity for a period of 7 days. You will also

receive instructions and the material to collect a stool sample.

Second Visit

According to standard care, you need a liver biopsy. This is an in-hospital procedure performed

by a liver specialist using local anesthesia. Local anesthesia means that a small area of your skin

over the liver will be frozen with a very small needle and then a specially designed needle will be

inserted between the ribs into the liver. The biopsy takes only a moment and there is slight

discomfort during the procedure. In some cases, the liver specialist cannot perform the liver

biopsy due to anatomical reasons. In this case, you will be referred to the ultrasound department

for your liver biopsy. The procedure is the same, but ultrasound is used to locate the best place to

obtain the liver sample, and the person performing the procedure will be an interventional

radiologist.

During the procedure, usually 1 – 2 small standard samples (3.0 cm long x 1 cm thickness (liver

specialist) or 2.5 cm long x 1 mm thickness (radiology)) will be taken from your liver to assess

for the presence of fat and inflammation (you will be provided with the standard consent form

routinely used for performing liver biopsy procedure). The study coordinator is present during

the liver biopsy and takes a portion of the sample for future tests.

After the biopsy you will be kept under observation for 2 to 4 hours.

We will ask you to collect a stool sample the day before your liver biopsy and bring the sample

with you to the hospital, where you give it to the study coordinator. We will ask you to fill out an

environmental questionnaire that inquires about factors that could influence the bacteria

composition in your stool.

The saved portion of the liver biopsy and the stool sample will be identified by a code and if you

decide not to participate in the study, they will be destroyed.

This entire visit will last 4-5 hours.

Third Visit: Follow-up with liver specialist for liver biopsy results (1-2 months after)

After the liver biopsy, you will need a follow-up appointment with the liver specialist to discuss

the results of the liver biopsy and possible treatments. You will be contacted by the study

106

coordinator about 10 days before the follow-up visit if the diagnosis of simple steatosis or NASH

is made and you qualify for the rest of the study. You will be instructed to record your food

intake for a period of 7 days (forms will be given to you in the first visit). Before your visit, you

will need to be in a fasting state for 12 hours (nothing to eat or drink overnight) for blood work

and nutritional assessment.

During this visit, you will return the 7-days food and activity record. The study coordinator will

record your medical history and review your medical chart for your drug regimen and previous

blood results. Your body fat will be measured (by taking the circumferences of your waist and

hip). Using a device, your skinfold thickness will be measured at several sites on your body

(mainly at your arms and at the back of your shoulders). You will have blood taken (5 table

spoons) for different measurements.

This visit will take 30 minutes with the study coordinator.

Participation and Withdrawal

Your participation in this study is voluntary. You can choose not to participate and you may

withdraw at any time without affecting your medical care. During the course of the study, you

will be kept informed of any new treatments or finding that may influence your participation in

the study. It is your responsibility to follow the directions and rules of the study. The data

gathered from this study is being monitored on an on-going basis by the study coordinator. You

will be notified of the results of the study once the study is completed.

Risks

There is always a small risk involved in invasive procedures such as the liver biopsy. These

risks are not related to research procedures and they are associated with your routine clinical

care. To minimize this risk, the biopsies will be done in the hospital premises by a very

experienced liver specialist or interventional radiologist. There may be some pain (in about 10%

of cases), which is usually mild and does not last long. Occasionally the pain is more severe and

requires painkillers. However, this also usually passes within a few hours. Bleeding may occur

in about 1 in 5000 to 1 in 10,000 cases. Rarely the needle will penetrate another organ, such as

the gall bladder. All such complications are usually managed without requiring surgery.

There may also be a risk associated with drawing blood with a needle which include discomfort,

bruising, or infection where the needle goes in.

Benefits

You may not receive any medical benefit from your participation in this study. However,

depending on the results of your tests, you may be asked in the near future (2-3 months) to

participate in another study where an intervention will be proposed to improve your liver

condition.

Remuneration

There will be no costs to you for participating in the study and you will not be charged for any

research procedure. You will receive $50.00 for participating in the study to compensate for

your time and traveling expenses once you complete the study. If you leave the study early, you

will be reimbursed for the time you have spent in the study.

107

Compensation

If you become ill or are physically injured as a result of participation in this study, medical

treatment will be provided. The reasonable costs of such treatment will be covered by your

health insurance for any injury or illness that is directly a result of participation in this trial. In

no way does signing this consent form waive your legal rights nor does it relieve the

investigators, sponsors or involved institutions from their legal and professional responsibilities”.

Confidentiality

All information obtained during the study will be held in strict confidence. All the samples will

be identified by a code. No names or identifying information will be used in any publication or

presentations. For the purpose of verification of clinical trial procedures and/or data, members of

specific agencies in the presence of the investigator or his/her staff may look at sections of your

medical records that are related to the study without participant’s names attached. These

agencies include: members of the Research Ethic Board and a representative from the Canadian

Liver Foundation and the Canadian Institutes of Health Research. By signing this form, you are

agreeing to make your study records available to the above-mentioned individuals. The above-

mentioned individuals will not see information that is not related to the study. No information

identifying you will be transferred outside the investigators in this study.

Contact Person

If you have any questions about the study, please contact Bianca Arendt (Study Coordinator) at

(416) 340-4104 or Dr. Johane Allard at (416) 340-5159.

If you have any questions about your rights as a research subjects, please call the chair of the

University Health Network Research Ethics Board at (416) 340-4557.

Consent

I have had the opportunity to discuss this study and my questions have been answered to my

satisfaction. I consent to take part in the study with the understanding that I may withdraw at any

time without affecting my medical care. I have received a signed copy of this consent form. I

voluntarily consent to participate in this study. I also understand that there is no guarantee that

this study will provide any benefit to me.

_______________________ __________________________ ____________

Patient’s Name (Please Print) Patient’ Signature Date

The potential benefits, risks and procedures associated with this study have been fully explained

to the volunteer and he/she has had ample time and opportunity to ask questions and to decide

whether or not to participate in the study. A copy of the signed consent form has been given to

the participant.

______________________ _________________________ __________

Name of Person Obtaining Signature Date

Consent

108

Patient Information Sheet & Consent Form

Healthy Liver Donors

Study Title: Non-alcoholic Steato-hepatitis (NASH) Versus Simple Hepatic Steatosis: Is

There a Difference in the Nutritional Factors Influencing Lipid Peroxidation and

Inflammation?

PRINCIPAL INVESTIGATOR: Dr. Johane P. Allard Tel: (416) 340-5159

SPONSOR: Canadian Liver Foundation & Canadian Institutes of Health Research

Introduction

You were seen in the Transplant Clinic of the University Health Network and you have

consented to operative and treatment procedures to donate part of your liver for liver

transplantation. In addition, you have signed consent to store a sample of your liver tissue for

future research. We are seeking your permission to use a portion of your liver, and samples of

your subcutaneous (fat under your skin) and mesenteric fat (fat around the internal organs) tissue

(0.5-1 g) as a control for a nutritional study.

If you agree to allow us to look at portions of your liver and fat tissue, the research coordinator

will contact you to see if you agree to have further nutritional assessments.

It is important that you understand the purpose, procedures, benefits, discomfort, risks and

precautions associated with the study before you agree to participate, so that you can make an

informed decision. This is known as the informed consent process. If there is anything in this

form that you do not understand, please consult the study coordinator. Make sure all your

questions have been answered to your satisfaction before signing this document.

Of course, you have the right to refuse to participate and also the right to withdraw from the

study at any time.

Purpose of the Study

Fatty infiltration of the liver can be associated with alcohol, obesity, diabetes and abnormal fat

and sugar handling by your body. When alcohol intake is excluded, this is called a “Non-

Alcoholic Fatty Liver” or “NAFLD”. If it is associated with inflammation (cell damage), it is

called “Non-Alcoholic Steato-Hepatitis” or “NASH”. No one knows why some patients with

excess fat in their liver remain stable while others progress to a more advanced disease like

cirrhosis and liver failure requiring liver transplantation. The purpose of this study is to

determine if nutritional factors measured in the blood and diet or the kind of bacteria that are

found in the intestinal tract can have an effect on liver tissue damage, the type of fat deposited in

the liver and also the genes involved in fat and sugar metabolism. We are also measuring

bacterial products in stool and blood samples.

You are asked to serve as a healthy control subject for this study. We would like to have similar

measurements from people with a healthy liver for comparison.

About 190 patients with liver-biopsy proven simple steatosis (fat infiltration), NASH (fat with

inflammation) or people with minimal findings on liver biopsy will participate in this study at

The Toronto General Hospital and The Toronto Western Hospital. For the purpose of this study,

nutritional assessment and blood work will be done only in one occasion, and we will ask the

109

participants for one single stool sample. Fifty patients with healthy liver will serve as a control

group.

Study Procedures

If you consent to participate in this study, the following study procedures are to be performed.

First Visit: (Approximately 30 minutes)

This visit is the present clinical appointment in the Transplant Clinic. You are now presented

with the consent form for the study and meet with the study coordinator who explains the study

and ask you to sign the consent. The study coordinator will review your medical chart and may

ask you some questions regarding your medical history and medications that you are taking. You

will then be instructed to record your food intake and physical activity for a period of 7 days.

Your body fat will be measured (by taking the circumferences of your waist and hip). Using a

device, your skinfold thickness will be measured at several sites on your body (mainly at your

arms and at the back of your shoulders).

You will need to be in a fasting state for 12 hours (nothing to eat or drink overnight) for blood

work. If you are fasting for this visit, you will have blood taken (5 table spoons) for different

measurements. If you are not in a fasting state, this will be done in the next visit. You will also

receive instructions and the material to collect a stool sample and an environmental questionnaire

that inquires about factors that could influence the bacteria composition in your stool. This form

should be filled out on the same day you are collecting the stool.

Second Visit (The day of the surgery)

During this visit, you will return the 7-days food, the activity record, the environmental

questionnaire and the stool sample. The study coordinator is present during the surgery and

takes the samples for future tests. The stool sample and the saved tissue portions will be

identified by a code and if you decide not to participate in the study, they will be destroyed.

Participation and Withdrawal

Your participation in this study is voluntary. You can choose not to participate and you may

withdraw at any time without affecting your medical care. It is your responsibility to follow the

directions and rules of the study. The data gathered from this study is being monitored on an

on-going basis by the study coordinator.

Risks

There may be a small risk associated with drawing blood with a needle which include

discomfort, bruising, or infection where the needle goes in.

Benefits

You may not receive any medical benefit personally from your participation in this study.

However, your participation will help understand the mechanism by which fatty liver disease can

develop and progress.

Remuneration

There will be no costs to you for participating in the study and you will not be charged for any

research procedure. You will receive $50.00 for participating in the study to compensate for

your time once you complete the study. If you leave the study early, you will be reimbursed for

the time you have spent in the study.

Compensation

If you become ill or are physically injured as a result of participation in this study, medical

treatment will be provided. The reasonable costs of such treatment will be covered by your

110

health insurance for any injury or illness that is directly a result of participation in this trial. In

no way does signing this consent form waives your legal rights nor does it relieve the

investigators, sponsors or involved institutions from their legal and professional responsibilities”.

Confidentiality

All information obtained during the study will be held in strict confidence. All the samples will

be identified by a code. No names or identifying information will be used in any publication or

presentations. For the purpose of verification of clinical trial procedures and/or data, members of

specific agencies in the presence of the investigator or his/her staff may look at sections of your

medical records that are related to the study without participant’s names attached. These

agencies include: members of the Research Ethic Board and a representative from the Canadian

Liver Foundation and the Canadian Institutes of Health Research. By signing this form, you are

agreeing to make your study records available to the above-mentioned individuals. The above-

mentioned individuals will not see information that is not related to the study. No information

identifying you will be transferred outside the investigators in this study.

Contact Person

If you have any questions about the study, please contact Bianca Arendt (Study Coordinator) at

(416) 340-4104 or Dr. Johane Allard at (416) 340-5159.

If you have any questions about your rights as a research subjects, please call the chair of the

University Health Network Research Ethics Board at (416) 340-4557.

Consent

I have had the opportunity to discuss this study and my questions have been answered to my

satisfaction. I consent to take part in the study with the understanding that I may withdraw at any

time without affecting my medical care. I have received a signed copy of this consent form. I

voluntarily consent to participate in this study. I also understand that there is no guarantee that

this study will provide any benefit to me.

_______________________ __________________________ ____________

Patient’s Name (Please Print) Patient’ Signature Date

The potential benefits, risks and procedures associated with this study have been fully explained

to the volunteer and he/she has had ample time and opportunity to ask questions and to decide

whether or not to participate in the study. A copy of the signed consent form has been given to

the participant.

______________________ _________________________ __________

Name of Person Obtaining Signature Date

Consent

111

APPENDIX 2

Subject Instructions for Stool Collection

You will be provided by a Plastic Buff Bag with: 1- Collection Bowl

2- Re-sealable plastic bag

3- Storage Ziploc container

4- Two ice bags

5- Cooler bag

Please follow these instructions carefully.

Important: As soon as you are at home, please put the ice bags in the freezer.

Collection Bowl and Package of Plastic Bag

You will note that the collection bowl has no bottom.

This is so you can insert the re-sealable plastic bag through it and

Fold the edge of the bag over top of the collection bowl.

The collection bowl is then lowered into centre of the toilet insert.

Plastic Toilet Insert

-Insert the collection bowl with bag into the toilet insert.

-Raise the toilet seat and place insert directly on porcelain rim of bowl.

-Lower seat and sit as usual.

Collection of Stool samples:

-Collect feces in a plastic bag folded over the top of a no-bottom collection bowl placed into the

toilet insert

-Close the bag tightly then put it in the storage Ziploc container

-Keep the sample at 4C (Freezer) until the date of liver biopsy.

-For sample transportation to the Toronto General Hospital, maintain the sample between the 2

frozen ice bags in the cooler bag during transportation to the Toronto General Hospital ( the stool

should be collected as early as possible to the liver biopsy date, ideally within 24 hours)

112

APPENDIX 3

Environmental Questionnaire

Patient ID ____________ Visit Date _____________

1. What country were you born in? _____________________________

2. If you were not born in Canada, when did you immigrate? ___________

3. What is your ethnic background? (Check all that apply)

□ African-Canadian □ Asian □ Caucasian

□ First Nation □ Hispanic □ Hispanic-Black

□ Middle Eastern □ Other _________________ (specify)

4. How were you born? □ Caesarian section (C-section) □ Natural birth □ I don’t know

5a. As an infant, were you ever breastfed?

□ Yes □ Never □ I don’t know

5b. If “Yes”, how long were you breastfed?

□ Less than 6 month □ More than 6 months □ I don’t know

6a. Do you have any allergies or food intolerances?

□ Yes □ No □ I don’t know

6b. If “Yes”, please specify: ________________________

7a. Do you have any history of gastro-intestinal diseases (e.g. inflammatory

bowel disease, celiac disease, gastric ulcer)?

□ Yes □ No □ I don’t know

7b. If “Yes”, please specify: _____________________

8a. Did you ever have any major abdominal surgery?

□ Yes □ No □ I don’t know

113

8b. If “Yes”, please specify: ________________________

Date of surgery: _____________________ (mm/yyyy)

9a. During the last 6 months, did you use any antibiotics?

□ Yes □ No □ I don’t know

9b. If “Yes”, when was the last time you used antibiotics?

________________________ (dd/mm/yy)

10a. During the last 6 months, did you use any laxatives?

□ Yes □ No □ I don’t know

10b. If “Yes”, how often do you use a laxative? __________________

10c. If “Yes”, when was the last time you used a laxative?

_________________ (dd/mm/yy)

11. Are you using any pre- or probiotic or synbiotic products on a regular basis (as

a food product or as a supplement such as tablets, powder, etc.)?

□ Yes □ No □ I don’t know

Name(s) of the product(s): ________________________

12. Did you travel outside of North-America during the last 6 months?

□ Yes Country: __________________ □ No

13a. Did you have any gastrointestinal infections during the last 6 months

(e.g. traveller’s diarrhea)

□ Yes □ No □ I don’t know

13b. If “Yes”, when did you have the last infection?

________________________ (dd/mm/yy)

14. Do you have any pets at home?

□ Yes What animal(s)? __________________ □ No

15a. Female Participants ONLY: Are you using any contraceptives?

□ Yes Specify: _______________________ □ No

114

15b. When was the first day of your last period? ____________________ (dd/mm/yy) □ I am past my menopause

15c. Usual length of your menstrual cycle? ___ days □ I don’t know

115

APPENDIX 4

Diet Intake Records

General Information:

Please read instructions before beginning to record your food intake.

Please keep an accurate record of everything you eat and drink for 3 days – 1 day should be on the

week-end. Include all snacks, alcoholic beverages, cigarette smoking, and condiments.

List each food item in as much detail as possible.

Try to describe the amount of food eaten.

Include the portion size model for each food item consumed.

For each food, briefly describe how it was prepared.

Include brand names where possible.

Recommendations of 2D Models Used To Describe Foods

Volume Models (A Side) Weight Model (B Side)

Beverages Beef, lamb, pork, fish

Cakes, quiches, pies Luncheon meats

Canned goods, vegetables, fruits, fish, etc. Solid cheese

Casseroles, stews, potted meats, cottage cheese Sliced chicken, turkey

Cereals, beans, nuts, chips Biscuits, muffins, cakes, some cookies

Fresh cooked vegetables, fruits

Frozen cooked vegetables, fruits

Lettuce and other salad items

Noodles, macaroni, spaghetti, rice, etc.

Sauces, soups, gravy, salad dressing, etc.

Sugar and other condiments

E.g. Beef and Broccoli Stir Fry – should be recorded as:

Food Item Description No. Model

Beef strips Lean, stir fry 1.0 A15

Broccoli Fresh, stir fry 0.5 A15

Pea Pods Fresh, stir fry 0.25 A15

Beef Sauce Thick salty 0.25 A15

Checklist: Have you included the amount and kind of:

Spread on bread, toast, vegetables, rice, etc…..

Milk and/or LAct-aid, LActaeeze, soy milk in cereal and beverages

Salad dressing; fats, oils in cooling, frying, etc

Sugar, jams, jellies, syrups

Candy, chocolate

Snack foods such as potato chips, pretzels, etc.

All fluids/beverages

116

DAILY FOOD INTAKE

ID: _________________________ Day of Week: ____________ Date: ________________

Time Food Item Description (frozen, fried, broiled; salted; brand name; etc.)

No. of serving

Amount/Model

Multiple/ B side Thickness

117

Activity Log Form

Please complete this activity log sheet for a period of the 7 days before your clinic visit.

According to the table provided below, please let us know how many units of exercise you

have performed in a day.

Activities required for one unit of exercise:

Intensity Time (min) Type of activity

Mild 30 Slow walking, Traveling by bus, Shopping,

Housecleaning

Moderate 20 Faster walking or walking down stairs, Cycling, Doing

heavy laundry, Ballroom dancing (slow)

Strenuous 10 Slow running, Climbing stairs, Disco dancing for the

elderly, Playing volley ball or table tennis

Very Strenuous 5 Jumping rope, Playing basketball, Swimming

ID: ________________________ DAY: _____________ DATE: _____________

Activity Log Sheet

Type of Activity Start

Time

Stop

Time

Intensity Level Duration of

Activity

Unit of

Exercise