bsc. (hons)iron transport and liver injury in mouse models of hereditary haemochromatosis roheeth d....

199
IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy of The University of Western Australia in the School of Medicine and Pharmacology, April 2013.

Upload: others

Post on 27-Jun-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

IRON TRANSPORT AND LIVER

INJURY IN MOUSE MODELS OF

HEREDITARY HAEMOCHROMATOSIS

Roheeth D. Delima

BSc. (Hons)

This thesis is presented for the degree of Doctor of Philosophy of The University of Western Australia in the School of Medicine and Pharmacology, April 2013.

Page 2: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

”Somewhere, something incredible is waiting to be known.”

- Dr. Carl Sagan (American Astronomer, Writer and Scientist, 1934-1996)

Page 3: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

iii

Contents

Declaration .................................................................................................................................... viii

Acknowledgements ......................................................................................................................... ix

Abstract x

Publications and presentations arising from this thesis ................................................................ xiii

List of Figures ............................................................................................................................... xiv

List of Tables ................................................................................................................................. xiv

Abbreviations ................................................................................................................................. xv

Chapter 1..............................................................................................................................................2

Literature review ...................................................................................................................................2

Introduction...........................................................................................................................................3

1.1 Iron in the human body ...................................................................................................3

1.2 Iron absorption................................................................................................................4

1.2.1 Dcytb ............................................................................................................................5

1.2.2 DMT1 ............................................................................................................................5

1.2.3 Haem uptake ................................................................................................................6

1.3 Iron export ......................................................................................................................7

1.3.1 Ferroportin ..........................................................................................................................7

1.4 Plasma iron .....................................................................................................................9

1.4.1 Transferrin-bound iron ..................................................................................................9

1.4.2 Non-transferrin bound iron ........................................................................................ 10

1.4.3 Transferrin-bound iron uptake ................................................................................... 10

1.4.3.1 Transferrin Receptor 1 (TFR1) .......................................................................... 10 1.4.3.2 Transferrin Receptor 2 (TFR2) .......................................................................... 12

1.4.4 Non-transferrin bound iron uptake ............................................................................ 14

1.4.4.1 Zrt- and Irt-like Protein 14 (ZIP14) .................................................................... 15 1.4.4.2 Ferritin uptake ................................................................................................... 16 1.4.4.3 Haem/Haemoglobin uptake ............................................................................... 16

1.5 Cellular iron ................................................................................................................. 17

1.5.1 Haem synthesis ......................................................................................................... 18

1.6 Iron storage ................................................................................................................. 19

1.6.1 Ferritin ....................................................................................................................... 19

1.6.2 Haemosiderin ............................................................................................................ 20

1.7 Systemic Iron homeostasis ......................................................................................... 21

1.7.1 Hepcidin .................................................................................................................... 21

1.7.2 Iron-dependent -hepcidin signalling ......................................................................... 22

1.7.2.1 BMP/SMAD signalling ....................................................................................... 22 1.7.2.2 Haemojuvelin ..................................................................................................... 22 1.7.2.3 HFE/TFR2 ......................................................................................................... 24

1.7.3 Erythropoietic Hepcidin signalling ............................................................................. 25

Page 4: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

iv

1.7.4 Inflammatory Hepcidin signalling .............................................................................. 25

1.8 Cellular iron homeostasis ............................................................................................ 27

1.8.1 Transferrin Receptor 2 (TFR2) regulation ................................................................. 27

1.8.2 Zrt- and Irt-like Protein 14 (ZIP14) regulation ........................................................... 27

1.8.3 Iron Regulatory Element (IRE)/Iron Regulatory Protein (IRP) regulation ................. 28

1.8.4 Ferroportin internalisation by hepcidin ...................................................................... 29

1.9 Hereditary haemochromatosis .................................................................................... 29

1.9.1 Hereditary haemochromatosis (HH) type 1 .............................................................. 29

1.9.2 Hereditary haemochromatosis type 2 ...................................................................... 30

1.9.3 Hereditary haemochromatosis type 3 ...................................................................... 31

1.9.4 Hereditary haemochromatosis Type 4 ..................................................................... 32

1.10 Pathogenesis of HH .................................................................................................... 33

1.11 Iron induced liver injury ............................................................................................... 33

1.11.1 Generation of reactive oxygen species ..................................................................... 34

1.11.2 Lipid peroxidation ...................................................................................................... 34

1.11.3 Lysosmal fragility ....................................................................................................... 35

1.11.4 Mitochondrial damage ............................................................................................... 35

1.11.5 DNA damage ............................................................................................................. 35

1.11.6 Oxidative stress ......................................................................................................... 36

1.11.7 Inflammatory cytokines ............................................................................................. 36

1.12 Present study............................................................................................................... 36

Aims .......................................................................................................................................... 37

Hypothesis 1: ....................................................................................................................... 37 Aim 1: ................................................................................................................................... 37 Hypothesis 2: ....................................................................................................................... 38 Aim 2: ................................................................................................................................... 38 Hypothesis 3: ....................................................................................................................... 38 Aim 3: ................................................................................................................................... 38 Hypothesis 4: ....................................................................................................................... 39 Aim 4: ................................................................................................................................... 39

Chapter 2........................................................................................................................................... 40

Materials and Methods ...................................................................................................................... 40

Materials ............................................................................................................................................ 41

2.1.1 Tissue collection ......................................................................................................... 41

2.1.2 Experimental procedures ........................................................................................... 41

2.1.3 Molecular biology ....................................................................................................... 42

2.1.4 Protein extraction and Western blotting ..................................................................... 42

2.1.5 Equipment .................................................................................................................. 43

2.1.5.1 Balances ................................................................................................................ 43

2.1.5.2 Centrifugation ........................................................................................................ 43

2.1.5.2 Imaging system ..................................................................................................... 43

2.1.5.3 Microscope ............................................................................................................ 43

Page 5: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

v

2.1.5.4 Peristaltic pump ..................................................................................................... 44

2.1.5.5 pH measurement ................................................................................................... 44

2.1.5.6 Pipettes ................................................................................................................. 44

2.1.5.6 Powerpacks ........................................................................................................... 44

2.1.5.7 Radioactivity measurements ................................................................................. 44

2.1.5.8 Spectrophotometry ................................................................................................ 44

2.1.5.9 Real-time PCR ...................................................................................................... 45

2.1.5.10 Thermocycler ......................................................................................................... 45

2.1.5.11 Western blot transfer apparatus ............................................................................ 45

2.1.6 Location of suppliers .................................................................................................. 45

General methods ............................................................................................................................... 46

2.2.1 Animals ...................................................................................................................... 46

2.2.2 ICP-AES .................................................................................................................... 47

2.2.3 Plasma Iron Assay .................................................................................................... 47

2.2.4 Total Iron Binding Capacity (TIBC) ........................................................................... 48

2.2.5 Non-Transferrin Bound Iron (NTBI) Assay ................................................................ 49

2.2.5.1 Preparation of Tris-carbanatocobaltate(III) trihydrate ....................................... 49 2.2.6 RNA extraction .......................................................................................................... 50

2.2.7 DNase treatment of RNA .......................................................................................... 51

2.2.8 RNA quantification..................................................................................................... 51

2.2.9 Gel Electrophoresis ................................................................................................... 52

2.2.10 Reverse transcriptase-Polymerase Chain Reaction (RT-PCR) ................................ 52

2.2.11 Primers ...................................................................................................................... 53

2.2.12 Protein extraction ...................................................................................................... 55

2.2.12.1 Bicinchoninic acid (BCA) protein assay ........................................................... 56 2.2.13 Statistical analysis ..................................................................................................... 56

Chapter 3........................................................................................................................................... 57

Characterisation of mouse models of hereditary haemochromatosis ............................................... 57

3.1 Introduction .......................................................................................................................... 58

3.2 Methods ............................................................................................................................... 59

3.2.1 Animals ....................................................................................................................... 59

3.2.2 Tissue collection ......................................................................................................... 59

3.2.3 Haematology .............................................................................................................. 59

3.2.4 Plasma iron measurement ......................................................................................... 60

3.2.5 Liver function .............................................................................................................. 60

3.2.6 Hepatic metal measurement ...................................................................................... 60

3.2.7 Gene expression ........................................................................................................ 60

3.2.8 p-Smad 1/5/8 expression ........................................................................................... 60

3.2.9 Statistics ..................................................................................................................... 61

3.3 Results ................................................................................................................................. 61

Page 6: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

vi

3.3.1 Haematology .............................................................................................................. 63

3.3.2 Plasma Iron parameters ............................................................................................. 65

3.3.3 Liver function .............................................................................................................. 67

3.3.4 Liver metal content ..................................................................................................... 69

3.3.5 Liver expression of iron regulatory genes .................................................................. 71

3.3.6 Liver expression of SMAD1/5/8 .................................................................................. 73

3.3.7 Liver expression of iron transport genes .................................................................... 75

3.3.8 Duodenal expression of iron transport genes ............................................................ 77

3.4 Discussion ............................................................................................................................ 79

Chapter 4........................................................................................................................................... 85

Non-transferrin-bound iron transport in hereditary haemochromatosis ............................................ 85

4.1 Introduction .......................................................................................................................... 87

4.2 Methods ............................................................................................................................... 88

4.2.1 Animals ....................................................................................................................... 88

4.2.2 NTBI uptake ................................................................................................................ 88

4.2.3 Tissue collection ......................................................................................................... 89

4.2.4 Plasma iron measurement ......................................................................................... 89

4.2.5 Hepatic iron content ................................................................................................... 89

4.2.6 Statistics ..................................................................................................................... 90

4.3 Results ................................................................................................................................. 90

4.3.1 Tissue iron content ..................................................................................................... 90

4.3.2 Plasma iron parameters ............................................................................................. 91

4.3.3 Plasma NTBI clearance .............................................................................................. 93

4.3.4 Tissue NTBI uptake .................................................................................................... 94

4.4 Discussion .......................................................................................................................... 102

Chapter 5......................................................................................................................................... 107

Disruption of HFE and TFR2 causes iron-induced liver injury in mice............................................ 107

5.1 Introduction ........................................................................................................................ 109

5.2 Methods ............................................................................................................................. 110

5.2.1 Animals ..................................................................................................................... 110

5.2.2 Tissue collection ....................................................................................................... 110

5.2.3 Histology ................................................................................................................... 110

5.2.4 Perls' Prussian blue staining .................................................................................... 111

5.2.5 Haemotoylin & Eosin staining ................................................................................... 111

5.2.6 Immunofluorescence ................................................................................................ 111

5.2.7 Biochemical markers of liver injury ........................................................................... 112

5.2.8 Collagen staining ...................................................................................................... 112

5.2.9 Gene expression ...................................................................................................... 113

5.3 Results ............................................................................................................................... 114

Page 7: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

vii

5.3.1 Iron measurements .................................................................................................. 114

5.3.2 Liver histology........................................................................................................... 116

5.3.3 Biochemical markers of liver injury ........................................................................... 119

5.3.4 Collagen deposition .................................................................................................. 121

5.3.5 Hepatic expression of injury-related genes .............................................................. 125

5.4 Discussion .......................................................................................................................... 127

Chapter 6......................................................................................................................................... 131

Inflammation in mouse models of hereditary haemochromatosis ................................................... 131

6.1 Introduction ................................................................................................................ 132

6.2 Methods ............................................................................................................................. 133

6.2.1 Animals ..................................................................................................................... 133

6.2.2 Tissue collection ....................................................................................................... 133

6.2.3 Plasma iron parameters ........................................................................................... 134

6.2.4 Gene expression ...................................................................................................... 134

6.3 Results ............................................................................................................................... 135

6.3.1 Effect of LPS with time on hepcidin expression ....................................................... 135

6.3.2 Hepatic expression of inflammatory genes .............................................................. 136

6.3.3 Plasma iron parameters ........................................................................................... 138

6.3.4 Hepatic expression of iron regulatory genes ............................................................ 140

6.3.5 Hepatic expression of iron transport genes .............................................................. 142

6.4 Discussion .......................................................................................................................... 144

Chapter 7......................................................................................................................................... 149

General discussion .......................................................................................................................... 149

7.1 Future directions ................................................................................................................ 155

7.1.1 The role of HFE and TFR2 and erythropoiesis ........................................................ 155

7.1.2 NTBI Transporters .................................................................................................... 155

7.1.3 Iron-induction of liver injury ...................................................................................... 156

Chapter 8......................................................................................................................................... 157

Bibliography .................................................................................................................................... 157

Page 8: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

viii

Declaration

I, Roheeth Delima declare that this thesis is my own account of my research and contains

as its main content work which has not previously been submitted for a degree at any

tertiary education institution.

…................................................

Roheeth Delima

…................................................

Date

Page 9: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

ix

Acknowledgements

Just as it takes a village to raise a child, my Ph.D candidature would not have been

possible without the support and assistance of the following people, to whom I am

eternally grateful

To my supervisors; thank you Professor Debbie Trinder, for giving me this great

opportunity and for your unending support. Dr Anita Chua, my heartfelt thanks for the

lunches, guidance, friendship and for “kicking my arse” when required. To Professor John

Olynyk and Dr Janina Tirnitz-Parker, thank you for all your support and for the promise that

there is a life after a Ph.D.

Thank you Dr Jane Allan, for your concern and advice over the time of my candidature,

your willingness to drop your own work to answer my constant questions has made my

time at Fremantle much easier. To all the past and present members of the Medical

Science labs at Fremantle hospital, thank you for making this such a nuturing and friendly

place to work.

Thank you to Fremantle Hospital Medical Research Foundation for your continued support

of my Ph.D (Warren Jones Scholarship) and external projects (grants).

To my friends, who were always willing to provide a much needed distraction and who

kindly refrained from asking how my thesis was going, thank you for all your patience and

support.

Last but not least, thank you to my family. To my sisters, thank you for putting up with me

during those times (years) when I was not always a pleasure to be around. Finally, thank

you to my parents. Thank you for your un-ending support; both financially and emotionally,

for your guidance and constant offers of assistance, this achievement is just as much

yours as it is mine.

Page 10: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

x

Abstract Fundamental biochemical activities, such as, oxygen transport, energy production and

cellular proliferation are all dependent on iron-containing proteins. Although iron is

essential, excess is toxic due to its ability to catalyse the production of reactive oxygen

species and damage cellular macromolecules (Chua et al. 2007). Hereditary

haemochromatosis (HH) is an autosomal recessive disorder in which excessive absorption

of dietary iron leads to iron accumulation in the parenchymal tissues. Excessive iron

accumulation is most prominent in the liver as well as in the pancreas, pituitary, heart,

joints and skin and may lead to liver fibrosis, cirrhosis and hepatocellular carcinoma,

diabetes mellitus, impotence, cardiac failure, arthritis and skin hyperpigmentation. There

are five types of HH caused by mutations in genes that encode proteins involved in the

synthesis of hepatic regulatory peptide hepcidin, and its receptor, ferroportin that regulate

iron metabolism.

The general aim of this study was to characterise the roles of the proteins;

haemochromatosis protein (HFE) and transferrin receptor 2 (TFR2) which are mutated in

HH type 1 and 3, respectively, in iron transport and the regulation of iron metabolism.

Disruption of both HFE and TFR2 in mice (Hfe-/-xTfr2mut) resulted in a more severe iron

loaded phenotype with increased plasma iron, non-transferrin bound iron (NTBI)

concentration, transferrin saturation, and liver iron content compared with mice with

disruption in either HFE (Hfe-/-) or TFR2 (Tfr2mut) alone. Hfe-/-xTfr2mut mice had elevated

liver Bmp6 mRNA expression consistent with increased liver iron content. However,

disruption of Hfe and Tfr2 expression resulted in ineffective liver p-Smad 1,5,8 signalling

leading to reduced liver Hamp1 expression. The more severe iron-loaded phenotype in

Hfe-/-xTfr2mut mice compared with single mutant mice suggests a model of iron-dependent

regulation of hepcidin where both HFE and TFR2 act as plasma iron sensors via parallel

and possibly converging signalling pathways.

Page 11: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

xi

Decreased hepcidin expression results in excessive dietary iron absorption and iron

release from macrophages, which saturates plasma transferrin and leads to the increased

presence of a toxic from of iron known as NTBI. Measurement of in vivo NTBI transport in

mouse models of HH showed that NTBI was cleared rapidly from the circulation in all

mouse models of HH, with most of the NTBI taken up by the liver and to a lesser degree

by the kidneys, pancreas and heart. Uptake of NTBI was greater in Hfe-/-xTfr2mut mice than

in Hfe-/- and Tfr2mut mice, which in turn had greater uptake than wild-type mice. NTBI

uptake was positively correlated with both plasma NTBI levels and iron content in the liver,

kidney, pancreas and heart suggesting that NTBI uptake contributes to tissue iron

overload in HH.

Free iron can generate reactive oxygen species (ROS) which may cause oxidative tissue

damage. In association with the previously mentioned severe iron-loaded phenotype, Hfe-/-

xTfr2mut mice had elevated plasma alanine transaminase activity, mild hepatic

inflammatory cell infiltration with scattered foci of CD45+ leukocytes co-localised

predominately with ferritin in portal regions of the liver. Elevated hydroxyproline levels, and

Sirius red and Trichrome staining demonstrated marked portal tract collagen deposition

and portal bridging in Hfe-/-xTfr2mut mice. In addition, there was decreased SOD activity

and enhanced lipid peroxidation in the liver, indicative of increased hepatic oxidative stress

in the Hfe-/-xTfr2mut mouse and to a lesser extent in the Tfr2mut mouse. The evidence of

iron-mediated liver injury seen in the Hfe-/-xTfr2mut mouse is similar to what is reported in

human HH, with mild inflammation, increased collagen deposition and decreased SOD

activity common findings in liver biopsies.

Inflammation has been shown to have a significant effect on iron metabolism causing a

phenomenon known as anaemia of inflammation. Administration of the inflammatory

Page 12: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

xii

stimuli, LPS, to HH mice induced inflammation resulting in decreased plasma iron and

NTBI levels with a concurrent increase in liver mRNA expression of the iron importer Zip14

and a decrease in the iron exporter Fpn. Inflammation also increased Hamp1 expression,

though this effect was diminished in HH mice. The increase in Zip14 levels and decrease

in Fpn expression, resulted in liver iron retention consistent with the anaemia of

inflammation, reducing the bioavailability of iron for erythropoiesis. Iron sequestration in

the liver may also contribute to iron-induced liver injury evident in the Hfe-/-xTfr2mut mouse.

In summary, the disruption of HFE and TFR2 resulted in decreased synthesis of the

hepatic iron regulator hepcidin, resulting in elevated plasma iron levels. Excess iron

saturated circulating transferrin, resulting in the presence of NTBI which was rapidly

removed from the circulation and deposited in the liver, kidney, pancreas and heart.

Excess iron in the liver resulted in iron-induced liver injury and fibrosis in mice with

disruption in both HFE and TFR2. Systemic inflammation may also exacerbate iron

sequestration in the liver, enhancing liver iron overload and iron-induced injury in HH.

Page 13: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

xiii

Publications and presentations arising from this thesis

Publications

1. Delima RD, Chua AC, Tirnitz-Parker JE, Gan EK, Croft KD, Graham RM, Olynyk

JK, Trinder D. 2012. Disruption of hemochromatosis protein and transferrin

receptor 2 causes iron-induced liver injury in mice. Hepatology 56(2):585-93

Impact factor 11.66.

2. Delima RD, Chua AC, Ho D, Olynyk JK, Trinder D. 2013. In vivo non-transferrin-

bound iron uptake is upregulated in murine models of Hereditary

Haemochromatosis. AJP: Gastrointestinal and Liver Physiology (Under review)

Oral Presentations

1. Delima RD, Chua ACG, Herbison C, Graham R, Olynyk J, Trinder D. 2009.

Disruption of both Hfe and Tfr2 causes more severe hepatic iron overload in

hereditary haemochromatosis; presented at the Hepatology and Luminal Workshop

(Australian Liver Association & Australian Liver Foundation), Yarra Valley,

Australia.

2. Delima RD, Chua ACG, Graham R, Olynyk J, Trinder D. 2010. Disruption of both

Hfe and Tfr2 causes more severe hepatic iron overload in hereditary

haemochromatosis; presented at the American Association for the Study of Liver

Disease annual meeting, Boston, USA.

3. Delima RD, Chua ACG, Ho D, Graham RM, Olynyk JK and Trinder D. 2011. Non-

transferrin bound iron transport in vivo is iron regulated in hereditary

haemochromatosis, presented at presented at the 4th Congress of the International

BioIron Society, Vancouver, Canada.

Page 14: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

xiv

List of Figures Page

Figure 1.1: Iron in the human body .................................................................................... 4 Figure 1.2: Model of the pathways of iron transport in the duodenum. ............................... 7 Figure 1.3: Model of iron transport pathways in the hepatocyte. ...................................... 17 Figure 1.4: Model of hepcidin regulatory pathways in the hepatocyte. ............................. 23 Figure 3.1: Plasma iron parameters. ................................................................................ 66 Figure 3.2: Liver metal content ......................................................................................... 70 Figure 3.3: Liver expression of iron regulatory genes. ...................................................... 72 Figure 3.4: Hepatic p-Smad1/5/8 protein expression. ...................................................... 74 Figure 3.5: Liver expression of iron transporter genes. .................................................... 76 Figure 3.6: Duodenal expression of iron transport genes. ................................................ 78 Figure 4.1: Plasma iron parameters. ................................................................................ 92 Figure 4.2: Plasma NTBI clearance ................................................................................. 93 Figure 4.3: Tissue NTBI uptake. ...................................................................................... 94 Figure 4.4: Liver NTBI uptake. ......................................................................................... 96 Figure 4.5: Kidney NTBI uptake. ...................................................................................... 98 Figure 4.6: Pancreas NTBI uptake. .................................................................................. 99 Figure 4.7: Heart NTBI uptake. ...................................................................................... 100 Figure 4.8: Duodenum and femur NTBI uptake .............................................................. 101 Figure 5.1: Hepatic iron concentration. .......................................................................... 115 Figure 5.2: Liver histology. ............................................................................................. 117 Figure 5.3: CD45+ / ferritin double staining in Hfe-/-xTfr2mut mice. ................................... 118 Figure 5.4: Biochemical markers of liver injury. .............................................................. 120 Figure 5.5: Liver collagen deposition via Sirius red stain. ............................................... 122 Figure 5.6: Sirius red stain correlated with iron, collagen and lipid peroxidation

measurement. ........................................................................................... 123 Figure 5.7: Liver collagen deposition using Masson’s trichrome stain. ........................... 124 Figure 5.8: Liver expression of injury-related genes. ...................................................... 126 Figure 6.1: LPS time course. ......................................................................................... 135 Figure 6.2: Liver expression of inflammatory cytokine genes: LPS vs. saline treated mice.

.................................................................................................................. 137 Figure 6.3: Plasma iron parameters: LPS vs. saline treated mice. ................................. 139 Figure 6.4: Liver expression of iron regulatory genes: LPS vs. saline treated mice. ....... 141 Figure 6.5: Liver expression of iron transport genes in LPS and saline treated mice. ..... 143

List of Tables Page

Table 2.1: Reagents used for reverse transcription of RNA. ............................................ 52 Table 2.2: Reagents in the PCR Master Mix. ................................................................... 53 Table 2.3: Primer sequences and annealing temperatures. ............................................. 53 Table 2.4: PCR cycling parameters.................................................................................. 55 Table 3.1: Body and organ weights of HH and WT mice. ................................................. 62 Table 3.2: Haematological parameters in HH and wild type mice. .................................... 64 Table 3.3: Serum markers of liver function in HH and wild type mice. .............................. 68 Table 4.1: Tissue iron content. ......................................................................................... 91

Page 15: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

xv

Abbreviations

The following abbreviations are used throughout this thesis:

2-DHBA Dihydroxybenzoic acid 4-HNE 4-Hydroxynonenal ABC ATP-binding cassette ALA Aminolevulinic acid ALAS2 Delta-aminolevulinate synthase 2 ALT Alanine transaminase BCA Bicinchoninic acid BMP6 Bone morphogenic protein-6 BMPR BMP receptor BPS bathophenanthroline disulfonic acid BSA Bovine serum albumin CD45 Cluster of differentiation45 CEBP/α CCAAT/enhancer-binding protein α CHOP CCAAT/Enhancer-Binding Protein Homologous Protein CREB cAMP response element-binding DAPI 4',6-diamidino-2-phenylindole

DcytB Duodenal cytochrome B

DMT1 Divalent metal transporter1 EKLF Erythroid Krüppel-like Factor EPO Erythropoietin ER Endoplasmic reticulum FBXL5 F-box and leucine-rich repeat protein 5 FFPE Formalin fixed paraffin embedded FLVCR Feline leukaemia virus subgroup C cellular receptor FPN Ferroportin GAPDH Glyceraldehyde 3-phosphate dehydrogenase GDF15 Growth differentiation factor 15 gp130 glycoprotein 130 H&E Hematoxylin and eosin HAMP Hepcidin gene Hct Haematocrit HFE Haemochromatosis protein Hfe-/- Hfe knockout mouse HH Hereditary haemochromatosis HIC Hepatic iron concentration HIF-2α Hypoxia inducible factor2α HJV Haemojuvelin HO-1 Haem oxygenase 1 ICP-AES Inductively coupled plasma – atomic emission spectroscopy Id1 Inhibitor of DNA binding-1 IL-6 Interleukin-6 IRE Iron regulatory element IRP Iron regulatory protein ISC Iron-sulfur cluster JAK-STAT Janus kinase-Signal Transducer and Activator of Transcription LAMP1 Lysosomal-associated membrane protein 1 LPS Lipopolysaccharide MCH Mean cell haemoglobin MCHC Mean corpuscular haemoglobin concentration MCV Mean corpuscular volume

Page 16: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

xvi

MDA Malondialdehyde MHC Major histocompatability complex MOPS 3-(N-morpholino) propanesulfonic acid MVB Multivesicular bodies NF-κB Nuclear factor kappa-light-chain-enhancer of activated B cells NMR Nuclear magnetic resonance NO Nitric oxide NTBI Non-transferrin bound iron PBS Phosphate buffered saline PCFT Proton-coupled folate transporter RBC Red blood cell RDW Red blood cell distribution width ROS Reactive oxygen species Scara5 Scavenger receptor class A, member 5 SDS Sodium dodecyl sulphate SLC Solute carrier SOD Superoxide dismutase STEAP Six transmembrane epithelial antigen of the prostate TBA Thiobarbituric acid TBARS Thiobarbituric acid reactive substances TBI Transferrin bound iron TBST Tris buffered saline and Tween TCA Trichloroacetic acid Tfr2mut Tfr2 mutant mouse TFRs Transferrin receptors TGA Thioglycolic acid TGFβ Transforming growth factor β TIBC Total iron binding content TIM2 T cell immunoglobulin and mucin domain 2 TLR4 Toll-like receptor 4 TNF Tumour necrosis factor TWSG1 Twisted gastrulation protein homolog 1 UTR Untranslated region WT Wild-type ZIP14 Zrt- and Irt-like protein 14

Page 17: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

2

Chapter 1

Literature review

Page 18: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

3

Introduction The unique ability of iron to serve both as an electron donor and acceptor makes it

essential in many physiological and metabolic processes. Fundamental biochemical

activities, such as oxygen transport, energy production and cellular proliferation are all

dependent on iron-containing proteins. Although a necessity at normal levels, excess iron

may become toxic due to its ability to catalyse the production of reactive oxygen species

and damage cellular macromolecules. This is of particular importance in the iron-overload

disorders, Herediatry haemochromatosis (HH) and β-Thalassaemia (Chua et al. 2007).

1.1 Iron in the human body The adult human body contains approximately 3-5 g of iron, with more than two-thirds of

body iron incorporated in the haemoglobin of mature red blood cells and erythroid

precursors (>2 g) (Andrews 1999). The remaining body iron is predominantly found in

macrophages of the reticuloendothelial system (≈600 mg) or within the iron storage

protein, ferritin, in hepatocytes (1000 mg). Small amounts of iron are also present in

myoglobin, in the muscles (≈300 mg) and as a constituent of other cellular iron containing

proteins and enzymes (≈8 mg). In humans, dietary iron absorption is in the range of 1 to 2

mg daily, which is approximately the amount of iron lost by the body each day, through

excretion and blood loss (Fig 1.1), as humans lack a dedicated mechanism for the

excretion of iron (Andrews 1999).

Page 19: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

4

Figure 1.1: Iron in the human body Approximately ≈1–2 mg of iron is absorbed daily to account for obligatory losses of a similar amount of iron through sloughing of mucosal and skin cells, haemorrhage, and other losses. Approximately 4 mg of iron is found in circulation bound to Tf, with the majority of body iron found in the erythroid compartment of bone marrow and in mature erythrocytes. Splenic reticuloendothelial macrophages, which recycle iron from senescent red blood cells, provide iron for the synthesis of new red blood cells. Liver hepatocytes store iron in ferritin shells. During pregnancy, 250 mg of iron is transported across the placenta to the foetus.

1.2 Iron absorption The majority of dietary iron is absorbed by the duodenum. There are two forms of dietary

iron: non-haem iron, derived from vegetables and grains, and haem iron, derived from red

meat. The contribution of these two sources of iron varies according to diet. Uptake of iron

occurs predominantly in the enterocytes of the duodenum, with gastric acidity promoting

the chelation of non-haem iron to soluble compounds such as amines, amino acids and

sugars (Hershko et al. 2007). Ninety percent of dietary iron exists as inorganic (non-haem)

Page 20: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

5

iron, predominantly in the form of the insoluble oxidised ferric (Fe3+) form. Following

digestion, ferric iron must first be reduced to the ferrous (Fe2+) form by the brush border

ferrireductase, duodenal cytochrome B (Dcytb; (McKie et al. 2001).

1.2.1 Dcytb

The mammalian ferric reductase Dcytb (Fig. 1.2), was first identified in 2001 and was

isolated from hypotransferrinaemic mice (McKie et al. 2001). Dcytb is highly hydrophobic

and has six transmembrane domains and is identical to haem protein p30. It is highly

expressed in the duodenum (McKie et al. 2001) and in erythrocyte membranes (Su et al.

2006), but has also been identified in the liver (McKie et al. 2001), and in airway epithelial

cells (Turi et al. 2006). The expression of Dcytb is strongly regulated by iron with Dcytb

mRNA and protein levels up-regulated in iron-deficient duodenum (Frazer 2002). However,

iron status does not appear to modulate Dcytb expression in the liver and spleen (McKie et

al. 2001). Regulation of Dcytb is hypothesised to be controlled by yet unidentified

transcription factors similar to the iron-inducible transcriptional regulator, Aft1p, which

controls yeast ferric reductases, but has been shown to be a direct regulatory target of

Hypoxia Inducible Factor 2α (HIF-2α) in response to hypoxic conditions (Mastrogiannaki et

al. 2009; Shah et al. 2009). Though the disruption of Dcytb in mice did not significantly

alter body iron stores (Gunshin et al. 2005), studies injecting radioactive 59Fe into tied

duodenal segments, demonstrated decreased iron uptake into the mucosa of Dcytb-

knockout mice (Choi 2008), supporting the role of Dcytb in the reduction of dietary iron.

Once reduced, the ferrous iron is then transported across the cell membrane by divalent

metal transporter 1 (DMT1; Figure 1.2) (Gunshin et al. 1997).

1.2.2 DMT1

The human DMT1 gene consists of 167 exons spread over more than 36 kb (Lee 1998).

The DMT1 protein is highly hydrophobic, with twelve predicted transmembrane domains

and both the amino-terminus and carboxy-terminus are predicted to exist within the

Page 21: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

6

cytoplasm (Zhou et al. 1998). DMT1 is capable of transporting a variety of divalent metals

including copper (Arredondo et al. 2003), cobalt, cadmium (Picard et al. 2000), and lead

(Garrick et al. 2006). It has also been reported that DMT1 can co-transport protons

(Gunshin et al. 1997) and functions optimally at approximately pH 6 (Su 1998). The

expression of intestinal DMT1 has been shown to be inversely regulated by iron status via

a post-transcriptional mechanism (See section 1.8.3).

1.2.3 Haem uptake

In most non-vegetarian diets more than one-third of the total daily iron is supplied by

dietary haemoglobin and myoglobin (Carpenter 1992). Before haem iron can be utilised,

haem must be cleaved from the haemoglobin and myoglobin proteins by proteolytic activity

in the lumen of the stomach and small intestine (Conrad 1967). Haem binds to the brush

border membrane of duodenal enterocytes and is translocated across the membrane by a

yet to be identified haem transporter (Figure 1.2). The transporter Haem Carrier Protein 1

(HCP1) was initially identified as a putative haem transporter (Shayeghi et al. 2005),

however its low affinity for haem (Km 125µM) and subsequent studies demonstrating its

high affinity for folate and its derivatives, has resulted in HCP1 being re-identified as

Proton-Coupled Folate Transporter (PCFT) (Qiu et al. 2006). After transport across the

apical membrane, the haem is degraded by haem oxygenase to release the ferrous ion

(Raffin 1974), which enters the low-molecular-weight iron pool in the enterocyte. The iron

may be stored in the cell as ferritin or transported across the basolateral membrane to the

plasma. Iron export across the basal membrane of the enterocyte is dependent on the iron

exporter, ferroportin, and the ferroxidase, hephaestin, which converts Fe2+ to Fe3+ and is

then bound by circulating transferrin (Fig. 1.2) (Donovan et al. 2005).

Page 22: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

7

Figure 1.2: Model of the pathways of iron transport in the duodenum. Uptake of ionic iron and haem iron from the lumen into the enterocyte across the apical membrane and transport out across the basolateral membrane to the blood (Chua et al. 2007). DMT1: Divalent metal transporter 1. DCYTB: Duodenal cytochrome b. FPN: Ferroportin. TF: Transferrin.

1.3 Iron export

1.3.1 Ferroportin

To date, only one iron exporter has been identified: ferroportin (FPN) (Donovan et al.

2005). FPN also known as Ireg1 (McKie et al. 2000) and Mtp1 (Abboud and Haile 2000)

was initially identified as an iron-export protein located on the basolateral membrane of

enterocytes (Figure 1.1). Subsequently it has been shown that FPN is a ubiquitously

expressed cell surface protein with 12-predicted transmembrane domains (Canonne-

Hergaux et al. 2006). Studies using Xenopus oocytes have shown that over-expression of

FPN results in an increase in iron released from the cells accompanied by a reduction in

ferritin expression, indicating a reduction of intracellular iron levels (Chung 2003). FPN

mRNA has been found to be highly expressed in duodenal enterocytes, spleen, kidney,

and liver, particularly in Kupffer cells and to a lesser degree in hepatocytes (Philpott 2002).

FPN expression is regulated at a transcriptional level by hypoxia (McKie et al. 2000),

Fe2+

Fe3+

Ferritin

Fe2+

Fe

2+

Fe2+

Fe2+

Gut Lumen Enterocyte Blood

DMT1

FPN

Hephaestin

Haem Haem

Haem

Oxygenase

DCYTB

Haem

transporter

Transit Iron Pool

Non-haem

iron Fe3+

Tf

Fe2Tf

Fe2+

DMT1

Fe2+

Page 23: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

8

inflammation (Yang et al. 2002), and haem, and iron (Knutson et al. 2003) and zinc

(Troadec et al. 2010) concentration. FPN is also post-transcriptionally regulated by cellular

iron levels via a Iron Responsive Protein (IRP)/Iron Responsive Element (IRE) post-

transcriptional mechanism (Abboud and Haile 2000)(see Section 1.8.3). Finally, FPN is

also regulated via a post-translational hepcidin-dependent mechanism (Nemeth et al.

2004)(see Section 1.8.4) and a hepcidin-independent substrate-dependent mechanism

(De Domenico et al. 2011). Once the iron is released from the cell, it is oxidised by

multicopper ferroxidases: hephaestin at the basolateral surface of the enterocyte or by

caeruloplasmin in other types of cells (De Domenico et al. 2007).

The expression of FPN at the cell surface is regulated by the antimicrobial peptide,

hepcidin, which through the interaction of hepcidin and duodenal FPN inhibits iron release

from the enterocyte and subsequently reduces plasma iron levels (see Section 1.8.4;

(Nemeth et al. 2004). First isolated from blood (Krause et al. 2000) and then urine (Park et

al. 2001), hepcidin expression was found to be regulated by body iron levels, with mice

deficient in hepcidin developing iron overload (Nicolas et al. 2001) and mice over-

expressing hepcidin developing severe anaemia and iron deficiency (Nicolas et al. 2002)

Though the regulation of FPN by hepcidin is well documented in macrophages, recent

studies suggest that this is a cell-specific effect, as numerous studies suggest that in the

duodenum, hepcidin may interact with the iron transporter DMT1 (Yamaji et al. 2004;

Chaston et al. 2008; Brasse-Lagnel et al. 2011).

Iron not released from the enterocyte is eventually lost through cell sloughing. FPN is also

highly expressed in macrophages and plays a key role in iron recycling. Senescent red

blood cells are phagocytosed by the macrophages of the reticuloendothelial system

(Kondo et al. 1988) Macrophages degrade haemoglobin and catabolise haem via haem

oxygenases (HO-1 and HO-2), liberating inorganic Fe2+, before being exported from the

Page 24: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

9

cell by FPN and re-oxidised by caeruloplasmin, where it binds to the iron transport protein,

transferrin (Fig. 1.2), for delivery to various tissues.

1.4 Plasma iron

1.4.1 Transferrin-bound iron

Transferrin is a monomeric, iron-binding glycoprotein composed of two structurally similar

lobes, each containing a single iron-binding site (Lambert 2012). It is expressed

predominantly in the foetal and adult liver and is transcriptionally regulated by iron status

with iron deficiency resulting in a 2-4 fold increase in the rate of transferrin synthesis

(Theisen et al. 1993). Low amounts of transferrin are also synthesised by other tissues,

such as the brain and testis (Zakin 1992). Plasma transferrin is a powerful chelator,

capable of binding iron tightly but reversibly (Huebers and Finch 1987). A molecule of

transferrin can potentially bind two atoms of Fe3+ with high affinity, which is higher in the

extracellular pH of 7.4 and decreases in the acidified endosomes, allowing for the

dissociation of the Fe3+. Iron bound to plasma transferrin accounts for less that 0.1% (≈3

mg) of total body iron but represents the most active iron pool in the body. More than 2

million erythrocytes are produced every second by the bone marrow, requiring a daily

supply of at least 20-30 mg of iron. To meet the requirements of erythropoiesis, plasma

transferrin turns over more than 10 times a day. It has been calculated that atoms of iron

entering the plasma transferrin pool will remain in the circulation for only 90 minutes before

being taken up by the bone marrow (Cavill 2002), with more than 80% being incorporated

into erythroblasts (Ponka et al. 1998). The saturation of transferrin with iron is an indicator

of body iron stores, but also reflects the balance between dietary iron absorption,

reticuloendothelial iron release and uptake by the bone marrow. Under normal conditions,

approximately 30% of the transferrin iron-binding sites are saturated. Low transferrin

saturation in conjunction with the high-affinity for iron, allows transferrin to efficiently buffer

Page 25: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

10

alterations in plasma iron levels, removing free iron from the circulation and minimising its

potential toxicity. In humans, a transferrin saturation of less than 15% indicates iron

deficiency, whereas saturations greater than 45% may indicate iron overload (Hentze et al.

2010). In disorders of iron overload, where transferrin saturation exceeds 60%, levels of

the toxic, redox-active non-transferrin bound iron (NTBI) increases dramatically (to 5 µM

and even higher) leading to possible tissue damage (Craven et al. 1987).

1.4.2 Non-transferrin bound iron

NTBI is a low-molecular-weight form of iron that is thought to play a major role in

pathological iron overload, via its ability to catalyse the formation of reactive oxygen

species (Jomova and Valko 2011). Despite its importance in the pathophysiology of iron

overload, the exact chemical composition of NTBI is still poorly understood. In the blood

plasma citrate, acetate, pyruvate and phosphates are all potential ligands, but is

considered to be the most likely ligand (May 1977), with 1H NMR studies on the serum of

haemochromatosis patients conclusively demonstrating the involvement on citrate in NTBI

coordination (Grootveld et al. 1989).

1.4.3 Transferrin-bound iron uptake

1.4.3.1 Transferrin Receptor 1 (TFR1)

Nearly all cells acquire iron via the uptake of transferrin-bound iron mediated by transferrin

receptors (TFRs). There are two types of TFRs, known as TFR1 and TFR2 present on cell

membranes. TFR1 is expressed by most types of cells except mature erythrocytes and

TFR2 is predominantly expressed by hepatocytes and erythroid precursors. TFR1 consists

of two identical glycoprotein transmembrane subunits linked by a disulphide bond. The

efficiency of TFR1 to deliver transferrin-bound iron depends on the iron content of the

transferrin, with differic transferrin binding to TFR1 with an affinity 30- and 500-fold higher

than mono-ferric and apo-transferrin, respectively (Young et al. 1984). Each receptor

subunit is capable of binding one molecule of transferrin, thus TFR1 can bind 2 molecules

Page 26: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

11

of transferrin. The dimeric TFR1 can mediate the uptake of four atoms of iron if each

molecule is saturated with iron (Sheth 2000). The expression of TFR1 is regulated by

cellular iron levels by a post-transcriptional mechanism (Section 1.8.3). TFR1 expression

is inversely regulated by cellular iron levels; low iron upregulates TFR1 and high iron

downregulates TFR1 levels. This regulatory process ensures the iron supply meets the

iron requirements of the cell and occurs at the post-transcriptional level through the

binding of iron regulatory proteins (IRP) to the iron regulatory elements (IRE) present in

the 3’ untranslated region of the TFR1 mRNA (Section 1.8.3) (Muckenthaler et al. 2008).

TFR1 is upregulated by cytokines, such as interleukin-2 (Seiser et al. 1993), mitogens

(Ouyang et al. 1993) and growth factors (Miskimins et al. 1986), as well as via hypoxia-

inducible factor 1α (HIF-1α), which binds to a conserved hypoxia regulatory element

binding site within the TfR1 promoter (Tacchini et al. 1999).

TFR1 mediated uptake of diferric transferrin has been studied extensively (Graham et al.

2007). Initially it involves the high-affinity interaction of differic transferrin with TFR1 at the

cell surface at pH 7.4 (Morgan 1981). Formation of the transferrin and receptor complex

triggers internalisation of the complex into a clathrin-coated vesicle (Harding 1983) which

matures into a proton-pumping endosome (Fig. 1.3). As the pH of the endosome

approaches 5.5, a conformational change in transferrin results in the release of the iron

(Klausner et al. 1983) and the increased binding affinity of transferrin for TFR1. The iron

released from transferrin into the endosome is reduced by the ferrireductase, six-

transmembrane epithelial antigen of the prostate (STEAP) 3 before being exported to the

cytoplasm via either DMT1 (Ohgami et al. 2005) or the zinc transport protein, Zrt- and Irt-

like protein-14 (ZIP14) (Fig. 1.3) (Zhao et al. 2010). The iron enters the liable iron pool and

is used immediately for cellular processes or stored in the iron storage protein, ferritin. The

acidified compartment containing the iron-depleted transferrin (apo-transferrin) still bound

to TFR1 returns to the cell surface where at a pH of 7.4 due to a decrease in binding

Page 27: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

12

affinity, the apo-transferrin is released from the receptor to the circulation. This allows the

TFR1 to bind more diferric transferrin and the transferrin-cell cycle to continue and deliver

more iron to the cell. It has been shown that iron delivery via the transferrin-TFR1 cycles

can be completed in approximately 5-20 min depending on cell type (Morgan and Baker

1986; Qian and Morgan 1992).

Although the high-affinity transferrin-TFR1 pathway is the predominant mechanism by

which most cells take up transferrin-bound iron, studies in Huh7 hepatoma cells suggest

the existence of internalisation of transferrin-bound iron via a low-affinity, TFR1-

independent manner (Trinder et al. 1996). At low transferrin concentrations, less than 0.3

µmol/L, transferrin-bound iron uptake occurs predominantly through TFR1, whilst at higher

transferrin concentrations, low-affinity mechanisms predominate (Anderson et al. 1994).

Interestingly, the physiological concentration of differic transferrin in human plasma is ≈5

µmol/L (Ponka et al. 1998), suggesting that in many cell types the low-affinity TFR1-

independent pathway dominates the acquisition of transferrin-bound iron. The exact

mechanisms of TFR1-independent uptake of transferrin-bound iron are still not clearly

characterised, however, one possible mechanism may involve transferrin receptor 2

(TFR2).

1.4.3.2 Transferrin Receptor 2 (TFR2)

TFR2 is a 105 kDa membrane protein with significant homology to TFR1. The TFR2 gene

expresses two transcripts, an alpha form (2.9 kb) and a beta form (2.5 kb). The TFR2-α

protein is a type-II membrane protein that has 45% identity and 66% similarity with the

extracellular domain of TFR1, and is most significantly expressed by the liver, with much

lower expression in the spleen, lung, muscle, prostate (Kawabata et al. 1999) and in early

erythroid precursors (Kawabata et al. 2001). The TFR2-β protein has been shown to lack

the amino-terminal portion of the TFR2-α protein including the putative transmembrane

Page 28: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

13

domain. However its expression is much more common than TFR2-α with significant

expression not only in the liver, but also in the brain, heart and spleen (Kawabata et al.

1999).

Though significantly homologous to TFR1, TFR2 has a 30-fold lower affinity for differic

transferrin than TFR1 (West et al. 2000). In addition, TFR2 protein levels show a dose-

dependent response to transferrin saturation, with increasing concentrations of diferric-

transferrin increase receptor levels by stabilising TFR2 protein (Johnson and Enns 2004;

Robb and Wessling-Resnick 2004; Chen and Enns 2007). TFR2 is not regulated by

intracellular iron levels (Kawabata et al. 2001) unlike TFR1, which is inversely regulated by

cellular iron status via the posttranscriptional iron responsive element-iron regulatory

protein (IRE-IRP) mechanism (Section 1.8.3). TFR2 is also unable to compensate for the

loss of TFR1 function, as Tfr1-knockout mice display embryonic lethality, and TfR1+/− mice

have lower hepatic iron levels than wild-type mice (Kawabata et al. 2000), this in contrast

to the hepatic iron overload associated with human or murine mutations in TFR2

(Camaschella et al. 2000).

The uptake of iron-loaded transferrin by TFR2 in liver Huh-7 cells and rat hepatocytes is

characterised by a pattern of biphasic internalisation kinetics. With steady-state

internalisation of transferrin typically saturated at low concentrations (<0.5 µM) (Blight and

Morgan 1983), this atypical pathway displays a linear phase of uptake at ligand

concentrations >0.5 µM, the subcellular distribution of the internalised ligand indicated that

TFR2 delivers transferrin to the late endocytic pathway (Figure 1.2), where it accumulated

in multivesicular bodies (MVB) devoid of receptors and deficient in lysosome-associated

membrane protein 1 (LAMP1), resulting in transferrin not being degraded (Robb and

Wessling-Resnick 2004). This suggests a model in which the apparently nonsaturable

Page 29: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

14

linear phase of transferrin uptake by TFR2 can be explained by the intracellular

accumulation of transferrin.

However, as TFR2 tissue distribution is limited, it is unlikely that TFR2 plays a major role in

low-affinity, TFR1-independent iron uptake, with only a minor decrease in hepatic

transferrin-bound iron uptake reported in TFR2 mutant mice (Chua et al. 2010). Other

potential mediators for low-affinity TFR1-independent uptake of transferrin-bound iron

include cubulin, which operates conjugated to its co-receptor, megalin (Kozyraki et al.

2001), in the proximal tubules of the kidney. Cubulin is expressed on the apical membrane

of proximal tubule cells and mediates the re-absorption of filtered transferrin from the

glomerular filtrate (Smith and Thevenod 2009). Other mechanisms include endocytosis

mediated by glyceraldehyde-3-phosphate-dehydrogenase (GAPDH) in macrophages (Raje

et al. 2007) and proteoglycans in hepatocytes (Hu and Regoeczi 1992).

1.4.4 Non-transferrin bound iron uptake

A small fraction (<1%) of iron in the plasma circulates as a non-transferrin form conjugated

to small molecules such as citrate and amino acids. In cases of physiological iron overload

circulating transferrin binding sites are saturated and excess iron or NTBI may be found in

the plasma. NTBI is extremely toxic due to its potential to generate free radicals that can

damage essential biological molecules and it is rapidly cleared from the plasma by the liver

(Brissot et al. 1985). Studies have shown that, at least in the rat, more than 60% of NTBI is

removed from the circulation on its first past through the liver at a rate of approximately 30

nmol/min/g liver, whilst in the same experiment less than 1% of transferrin-bound iron was

cleared by the liver on its first pass (Wright et al. 1986). A subsequent study suggests that

the plasma half-life of NTBI may be less than 30s (Craven et al. 1987). These studies

indicate that the liver is part of an extremely efficient mechanism for the removal of NTBI

from the circulation, however the exact mechanism by which this occurs is still poorly

understood. DMT1 was initially proposed to be the major NTBI transporter in the liver

Page 30: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

15

(Figure 1.2), the principal site of NTBI deposition. However, liver-specific Dmt1-/- mice are

not protected against hepatic iron accumulation (Gunshin et al. 2005). This is consistent

with the knowledge that limited iron transport occurs by proton-dependent DMT1 at the

neutral pH of the extracellular fluid. These findings clearly suggest the existence of other

NTBI uptake pathways.

1.4.4.1 Zrt- and Irt-like Protein 14 (ZIP14)

Although its role in liver iron-loading remains to be established, ZIP14 has been shown to

facilitate the uptake of NTBI in cultured cells (Liuzzi et al. 2006). ZIP14 is a member of a

large family of metal-ion transporters, the SLC39 family (Lichten and Cousins 2009). ZIP14

is abundantly expressed in the liver, heart and pancreas (Liuzzi et al. 2006), the major

sites of tissue injury in iron overload. Further evidence for the involvement of ZIP14 in

NTBI transport (Figure 1.2) comes from studies that have utilised siRNA to reduce ZIP14

protein levels and in turn reduce iron and zinc uptake (Liuzzi et al. 2006). In the rat liver

perfusion model, it has been shown that zinc strongly inhibited the uptake of ferrous iron

(Wright et al. 1986). Similar observations have been made in primary hepatocytes from

mice (Chua et al. 2004) and rats (Baker et al. 1998), where the presence of zinc inhibited

the uptake of ferric citrate. ZIP14 has been shown to be specific for Fe2+ with Fe3+ not

transported (Pinilla-Tenas et al. 2011). Though most NTBI in the plasma exists as Fe3+-

citrate, a significant amount may be reduced to Fe2+ via the presence of plasma L-ascorbic

acid (May et al. 1999), superoxide (Ghio et al. 2003), the presence of mammalian cell

surface ferrireductases in hepatocytes and other cell types (Inman et al. 1994; Jordan and

Kaplan 1994), or by citrate chelating Fe2+ in addition to Fe3+.

NTBI can also be taken up by cardiomyocytes via L-type voltage-dependent calcium

channels. When mice with iron overload were treated with calcium channel blockers, there

was an attenuation of myocardial iron accumulation and a decrease in iron-associated

Page 31: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

16

oxidative stress (Oudit et al. 2003). A similar role for voltage-gated calcium channels has

been proposed for the delivery of iron to neuronal cells (Gaasch et al. 2007).

In the kidney, iron-loaded lipocalin 2 can be endocytosed by the receptors 24p3R

(Devireddy et al. 2005) and megalin (Hvidberg et al. 2005). These mechanisms for the

uptake of NTBI primarily operate during organ development (Yang et al. 2002), injury (Mori

et al. 2005), or inflammation (Devireddy et al. 2005). However, as lipocalin 2 deficient mice

do not exhibit any observable defects of iron metabolism (Flo et al. 2004), this excludes

lipocalin 2 from functioning as a major mechanism for iron uptake.

1.4.4.2 Ferritin uptake

Serum ferritin has also been implicated in the uptake of NTBI, particularly in pathological

conditions in which tissue damage results in the release of iron dense intracellular ferritin

into the plasma. The endocytosis of ferritin is mediated by a ferritin receptor and several

candidate molecules have been identified including T-cell immunoglobulin-domain and

mucin-domain 2 (TIM-2) in cells of the spleen, liver, bile duct, and kidney (Chen et al.

2005) and Scavenger receptor class A, member 5 (Scara5) in epithelial cells (Li et al.

2009). Studies have also shown that the H-subunit of ferritin can be internalised upon

specific binding to TFR1 (Li et al. 2009).

1.4.4.3 Haem/Haemoglobin uptake

A further mechanism of NTBI uptake occurs when intravascular haemolysis results in the

release of haemoglobin or free haem into the plasma. The liver-derived plasma protein

haptoglobin binds free haemoglobin and promotes its endocytosis by macrophages, upon

recognition and binding to the CD163 receptor (Kristiansen et al. 2001). Similarly,

haemopexin scavenges free haem and the resulting complex is endocytosed via the CD91

receptor (Figure 1.3) present on the surface or macrophages, hepatocytes and possibly

other cell types (Hvidberg et al. 2005).

Page 32: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

17

Figure 1.3: Model of iron transport pathways in the hepatocyte. Uptake of transferrin bound iron (Transferrin receptors 1 and 2: TfR1 and TfR2), non-tranferrin bound iron (Divalent metal transport 1: DMT1 and Zrt- and Irt-like protein 14: ZIP14), and haem (Cluster of differentiation 91 receptor: CD91R). Export of iron from the endosome (DMT1, Six transmembrane epithelial antigen of the prostate 3: STEAP3, and ZIP14) and iron release from the cell (Ferroportin and caeruloplasmin)(Figure modified from (Graham et al. 2007).

1.5 Cellular iron Once iron leaves the endosome, it becomes part of a transient cytosolic pool of iron,

presumably bound to low molecular weight intracellular chelators, such as citrate, various

peptides, ATP, AMP or pyrophosphate, which is known as the labile iron pool (Kakhlon

and Cabantchik 2002). Although it represents only a small fraction total cellular iron (≈3-

5%), the cytosolic labile iron pool is a reflection of cellular iron status (Kruszewski 2003),

with fluctuations triggering homeostatic adaptive responses.

Page 33: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

18

Cytosolic iron can enter the mitochondria via the SLC transporter mitoferrin, which is

localised to the inner mitochondrial membrane (Shaw et al. 2006). Studies also suggest

that 2,5-dihydroxybenzoic acid (2-DHBA), a ligand for lipocalin 2, may also play a role in

mitochondrial iron transport (Devireddy et al. 2005), as cells unable to synthesise this

mammalian siderophore accumulate excessive quantities of cytoplasmic iron. However,

there is still some controvery regarding this work has it has been difficult to reproduce

(Correnti et al. 2012).Further studies utilising erythroid cells suggest that iron may also be

transported directly from the endosomes to the mitochondria via a “kiss and run”

mechanism involving direct contact between the two organelles (Richardson et al. 2010).

Once within the mitochondria, iron is primarily used for the synthesis of haem and iron

sulphur clusters (ISCs).

1.5.1 Haem synthesis

The synthesis of haem from the precursor 5-aminolevulinic acid (ALA) occurs via an

evolutionarily conserved eight step pathway. Briefly, ALA is exported to the cytosol where

it is converted into the intermediate metabolites porphobilinogen, hydroxymethylbilane,

uroporphyrinogen III and copro-porphyrinogen III, which is oxidised to protoporphyringen

IX and imported into the mitochondria where it is further oxidised to protoporphyrin IX. In

the final step of the haem synthesis pathway, ferrochelatase catalyses the insertion of Fe2+

into protoporphyrin IX. The newly synthesised haem is then exported to the cytosol for

incorporation into haem-containing proteins (Ryter and Tyrrell 2000). It is suggested that

the transport of haem and its intermediates across mitochondrial membranes might involve

the ATP-binding cassette (ABC) family of transporters (Severance and Hamza 2009) and

the SLC transporter, SLC25A39 (Nilsson et al. 2009). In non-erythroid cells, the rate-

limiting step of haem synthesis is the production of ALA. This is in contrast to what occurs

in erythroid cells, where it is the synthesis of the porphyrin ring, a step that is iron-

dependent, that is rate limiting (Ponka 1997). Interestingly, the enzyme ALA synthase,

responsible for the production of ALA in erythroid precursor cells (Srivastava et al. 1988),

Page 34: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

19

contains an Iron Response Element and as described in section 1.8.3 is post-

transcriptionally regulated by Iron-Responsive Proteins, providing another point of

regulation for the haem biosynthesis pathway.

1.6 Iron storage

1.6.1 Ferritin

Cells may export excess intracellular iron by exporting Fe2+ via ferroportin or by secretion

of haem through the putative haem exporter, feline leukaemia virus, subgroup C, receptor

(FLVCR) (Keel et al. 2008). However, cells can also store and detoxify excess intracellular

iron in the cytosol by incorporating it into the iron storage protein, ferritin. Ferritin is able to

store large amounts of iron in a soluble, non-toxic form.

Synthesis of ferritin in the liver occurs primarily on free polyribosomes within the

hepatocyte (Konijn 1977) with the rate of ferritin production regulated by the iron status of

the cell by a post-transcriptional mechanism (Section 1.8.3). Ferritin is composed of an

apoprotein shell with a molecular weight of approximately 480 kDa which surrounds a core

of up to 4500 atoms of iron in the form of the mineral ferrihydrite (Harrison 1987). The

ferritin molecule is composed of 24 subunits of two structurally distinct subunit types. The

heavy or H-subunit has a more acidic isoelectric point and a molecular weight of

approximately 21 kDa and the light of L-subunit has a more basic isoelectric point and a

molecular weight of approximately 19 kDa. Differing proportions of each subunit give rise

to isoferritins which have characterisitic isoelectric points and tissue distribution. These

isoferritins (L- and H-type ferritin) are functionally different, with H-ferritin taking up iron

more rapidly than L-ferritin and storing iron in a more metabolically available form

(Harrison and Arosio 1996). In addition, H-type ferritin has biological effects that are

unrelated to iron binding, such as the capacity to inhibit cell growth (Guo et al. 1998),

Page 35: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

20

lymphocyte proliferation (Morikawa et al. 1995) and inflammatory signalling (Ramm and

Ruddell 2010). Non-parenchymal liver cells, mainly Kupffer and endothelial cells, of iron-

loaded rats, contained the acidic H-type isoferritin that is not present in hepatocyte ferritin

(Doolittle and Richter 1981).

Iron stored within ferritin is bioavailable and may be mobilised for metabolic purposes

during its lysosomal turnover (Zhang et al. 2010) and possibly also following structural

rearrangements of the ferritin subunits (Takagi et al. 1998). The induction of ferroportin

promotes mobilisation and export of ferritin-derived iron, followed by mono-ubiquitination

and degradation of the apo-ferritin by the proteasome (De Domenico et al. 2006).

1.6.2 Haemosiderin

With normal levels of cytosolic iron, soluble iron-containing ferritin is present in the cytosol

as randomly dispersed ferritin particles. As cytosolic iron levels increase, as in conditions

of pathological iron overload, the concentration of dispersed ferritin increases and small

clusters of ferritin particles appear, still soluble and spread throughout the cytosol. With

further increases in cytosolic iron, ferritin is collected in lysosomes by fusion of ferritin

clusters with lysosomal membranes or by autophagocytosis (Iancu 1992). It is suggested

that digestion of ferritin within secondary lysosomes (siderosomes) leads to denaturation

of ferritin protein subunits (Miyazaki et al. 2002) and to the aggregation of the ferritin iron

cores, resulting in the formation of amorphous, insoluble masses known as haemosiderin

(Iancu et al. 1997). Haemosiderin, which is most commonly found in macrophages, is

especially abundant in situations following haemorrhage but is also found in pathological

conditions in which the capacity of ferritin to store iron is overwhelmed. Ferritin iron may

have a pro-oxidant role and contribute to tissue damage (Arosio and Levi

2010). Production of haemosiderin, enclosed within siderosome membranes, may

sequester the excess iron away from the cytosol and help protect against iron toxicity. As

the total amount of tissue iron increases, the proportion stored as haemosiderin rises, from

Page 36: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

21

trace amounts in normal individuals to 90% or more in patients with severe iron overload

(Selden et al. 1980).

Depending upon cell type, iron supply and utilisation, the half-life of cellular ferritin may

range from ≈20 to 96 h (Truty et al. 2001) with haemosiderin having a much slower cellular

turnover than ferritin.

1.7 Systemic Iron homeostasis In mammals iron homeostasis is regulated at the level of iron absorption as there is no

physiological pathway for the excretion of iron. Impaired regulation of iron absorption leads

to either iron deficiency or overload. This complex task is accomplished by the liver-

derived peptide hormone, hepcidin, that responds to multiple regulatory signals, including,

iron availability, erythropoietic activity, inflammation, endoplasmic reticulum (ER) stress

and hypoxia (Hentze et al. 2010).

1.7.1 Hepcidin

Activated hepcidin is a 25 amino acid peptide (Park et al. 2001), synthesised primarily by

hepatocytes, but also at significantly lower levels in other cell types, upon cleavage of the

larger 84 amino acid pro-peptide by the pro-hormone convertase, furin (Valore and Ganz

2008). Mature hepcidin is secreted into the plasma and circulated bound to α2-

macroglobulin (Peslova et al. 2009). Hepcidin levels increase in response to iron (Pigeon

et al. 2001), ER stress (Vecchi et al. 2009), or inflammation (Nemeth et al. 2004). In

contrast, it has been shown that hepcidin levels decrease with iron deficiency, hypoxia and

increased erythropoiesis (Nicolas et al. 2002).

Page 37: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

22

1.7.2 Iron-dependent -hepcidin signalling

1.7.2.1 BMP/SMAD signalling

It is proposed that hepatic iron content induces hepcidin expression via bone morphogenic

protein (BMP) signalling (Babitt et al. 2006). Activation of the BMP receptor by BMP

binding (Steinbicker et al. 2011) leads to phosphorylation of the intracellular SMAD1/5/8

(mothers against decapentalegic; (Kautz et al. 2008). Subsequently, p-SMAD1/5/8 forms a

complexe with SMAD4 before translocating to the nucleus, where upon it binds to two

BMP-responsive elements (BMP-RE 1 and 2) at proximal and distal sites of the hepcidin

promoter, thereby inducing its transcription (Fig. 1.4)(Casanovas et al. 2009)) The

aforementioned model is validated by experiments in which SMAD4-/- mice have

decreased hepcidin expression and develop iron overload (Wang et al. 2005) and by

experiments that show that inhibitory SMAD7 prevents the interaction of pSMAD1/5/8 with

SMAD4 resulting in decreased hepcidin expression (Mleczko-Sanecka et al. 2010). BMP2,

5, 6, 7 and 9 have all been shown to induce hepcidin expression in vitro, however, BMP6

is the most physiologically relevant, with liver-specific disruption of BMP6 resulting in

nearly undetectable hepcidin levels with concomitant iron overload, excluding a

compensatory role for the other BMPs (Andriopoulos et al. 2009). The mechanism by

which BMP6 is induced by iron is still not fully understood, but as BMP6 mRNA levels have

been shown to correlate with hepatic iron content, it would suggest a role for BMP6 in

sensing alterations to hepatocellular iron (Kautz et al. 2008).

1.7.2.2 Haemojuvelin

Haemojuvelin (HJV) is a BMP co-receptor that can complex with Type I and II BMP

receptors, enhancing BMP/SMAD signal transduction and increasing hepcidin expression

(Figure 1.4)(Babitt et al. 2006). HJV is predominantly expressed in hepatocytes and

skeletal muscle and is bound to cell membranes via a C-terminal Glycophosphatidylinositol

anchor. Studies suggest that membrane-bound hepatocellular HJV can be inactivated

following cleavage by matriptase-2, thereby decreasing BMP/SMAD signalling and

Page 38: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

23

negatively regulating hepcidin expression (Silvestri et al. 2008). In genome-wide

association studies, mutations in matriptase-2 have been shown to correlate with serum

iron, transferrin saturation (Tanaka et al. 2010) and haemoglobin levels (Chambers et al.

2009). Expression of matriptase-2 has also been shown to be up-regulated by hypoxia and

BMP6 or iron, with BMP6 most likely working via induction of Id1 (inhibitor of DNA binding

1) (Meynard et al. 2011). Haemojuvelin cleaved my matriptase-2 results in soluble HJV

which has also been shown to directly compete with BMP6 for binding to the BMPR (Nili et

al. 2010), resulting in decreased BMP/SMAD signalling.

Figure 1.4: Model of hepcidin regulatory pathways in the hepatocyte. The figure shows the hepcidin regulation pathways (Receptor-regulated SMAD: R-SMAD; SMAD1/5/8, Janus kinase 1: JAK1, Signal transducer and activator of transcription 3: STAT3, and phophorylation: P) by which differic transferrin (Transferrin receptors 1 and 2: TfR1 and TfR2, Haemochromatosis protein: HFE), intracellular iron levels (Bone morphogenic protein 6: BMP6, BMP6 receptor: BMP6R, and Haemojuvelin: HJV) and inflammation (Interleukin 6: IL6, IL6 receptor: IL6R, and glycoprotein 130: gp130) regulate hepcidin mRNA.

Page 39: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

24

1.7.2.3 HFE/TFR2

The regulation of hepcidin occurs via two pathways, intracellular iron concentration affects

hepcidin signalling via the BMP/SMAD pathway and via a more dynamic pathway, plasma

transferrin saturation affects hepcidin via the HFE/TFR2. Haemochromatosis protein (HFE)

is an MHC-1 like molecule, and as such, interacts with β2-microglobulin (β2M; (Feder et al.

1996). The observation that loss of β2M leads to iron overload was the first suggestion that

linked HFE to iron metabolism (Porto et al. 1998). Subsequently, TFR1 (Parkkila et al.

1997) and its homologue TFR2 (Goswami and Andrews 2006) were also identified as

binding partners of HFE. Even though TFR1 and TFR2 have similar structures, it has been

shown that the domains through which they interact with HFE are different, since the

transferrin binding site of TFR2 does not overlap with the HFE binding site as it does with

TFR1 (West et al. 2001). It is proposed that when differic-transferrin binds to TFR1, HFE

dissociates and binds to TFR2, which has been shown to be capable of binding differic

transferrin and HFE simultaneously (Chen et al. 2007). In addition, it has been

demonstrated that TFR2 will compete with TFR1 for HFE binding regardless of differic

transferrin levels (Goswami and Andrews 2006), and that the HFE/TFR2 complex is

necessary for the induction of hepcidin expression (Fig. 1.4). However, recent studies

utilising combined Hfe and Tfr2 disruption in mice show that these animals develop more

severe iron overload than their single mutant counterparts, suggesting that HFE and TFR2

may exert some independent effects on hepcidin regulation (Wallace et al. 2009; Corradini

et al. 2011). The HFE/TFR2 complex is proposed to signal to hepcidin via the ERK/MAP

(extracellular signal regulated kinase/mitogen activated protein) kinase pathway (Fig. 1.4),

as this pathway has been shown to be activated when cultured murine hepatocytes are

treated with differic transferrin, with subsequent treatment with an ERK inhibitor blocking

the induction of hepcidin by diferric transferrin (Ramey et al. 2009). Similarly, Hfe-/-, Tfr2-/-,

and Hfe-/-xTfr2-/- mice all show significant decrease in hepatic p-ERK1/2 levels, suggesting

that both HFE and TFR2 signal hepcidin via the ERK1/2 pathway (Wallace et al. 2009).

Page 40: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

25

1.7.3 Erythropoietic Hepcidin signalling

Studies have shown that the injection of erythropoietin (EPO) in mice (Pak et al. 2006) and

the treatment of isolated mouse hepatocytes with EPO (Pinto et al. 2008) resulted in a

dose-dependent decrease in hepcidin. In addition, human volunteers treated with EPO

have greatly reduced urinary hepcidin levels (Robach et al. 2009). EPO has been shown in

vitro to directly suppress hepcidin expression, decreasing the binding of CEBP/α to the

hepcidin promoter (Pinto et al. 2008). However, in vivo studies indicate that the down-

regulation of hepcidin is triggered by erythropoietic activity and the increased iron demand

by erythroid precursor cells, rather than directly by EPO (Vokurka et al. 2006). EPO has

also been shown to modulate iron homeostasis by inducing TFR1 expression, and

increasing iron uptake and subsequently haem synthesis in erythroid progenitor cells

(Weiss et al. 1997), and by inhibiting pro-inflammatory immune pathways known to affect

hepcidin and ferritin expression (Nairz et al. 2011). Mechanisms by which erythroid cells in

the bone marrow induce hepcidin suppression are proposed to involve growth-

differentiating factor 15 (GDF15) and twisted gastrulation-1 (TWSG1). GDF15 is a member

of the transforming growth factor (TGF) β superfamily and is predominantly expressed by

erythroid precursors and serum levels of GDF15 are increased in patients with β-

thalassaemia (Tanno et al. 2007), congenital dyserythropoietic anaemias (Casanovas et

al. 2011) and sideroblastic anaemias (Ramirez et al. 2009). Similarly, TWSG1 is also

produced by erythroid precursors and has been shown to increase in thalassaemia and to

suppress hepcidin expression in vitro, by indirectly inhibiting the BMP/SMAD

transcriptional activation of hepcidin (Tanno et al. 2009).

1.7.4 Inflammatory Hepcidin signalling

Inflammatory cytokines and cell stress signals can also modulate hepcidin expression. The

inflammatory up-regulation of hepcidin is considered a mechanism by which the innate

immune system can deprive rapidly growing pathogens of iron. IL-6 has been shown to

promote the phosphorylation of signal transducer and activator of transcription (STAT) 3,

Page 41: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

26

which then translocates to the nucleus and upon binding to a proximal promoter element

induces hepcidin transcription (Figure 1.3; (Pietrangelo et al. 2007). In addition, mice

injected with bacterial lipopolysaccharide (LPS) demonstrate increased hepatic hepcidin

expression. This may occur directly via LPS binding to TLR4 (toll-like receptor 4) and

inducing hepcidin expression, or indirectly via increased IL-6 binding to its receptor and

activating the STAT3 pathway in response to LPS binding to TLR4 (De Domenico et al.

2010). IL-1β is also a potent inducer of hepcidin expression utilising both C/EBPα and

BMP/SMAD signalling pathways to induce expression (Matak et al. 2009). In contrast,

tumour necrosis factor (TNF) has been shown to downregulate hepcidin expression in two

ways. Firstly by binding to a HJV response element and inducing its downregulation

(Constante et al. 2007; Salama et al. 2012) and secondly by direct inhibition of SMAD1

protein (Shanmugam et al. 2012). However, previous studies have demonstrated that both

the SMAD1/5/8 and STAT3 pathways are linked by the protein complex Activin B, a TGF-β

protein superfamily and structurally similar to BMP (Kingsley 1994). Activin B has been

shown to induce SMAD1/5/8 phosphorylation in combination with IL-6/STAT3 signalling,

resulting in a marked increase in hepcidin expression (Besson-Fournier et al. 2012). The

induction of hepcidin transcription by endoplasmic reticulum (ER) stress has been

demonstrated to involve the transcription factors cyclic AMP response element-binding

protein (CREB) H and/or the stress-inducible C/EBP homologous protein (CHOP), with

CHOP inducing hepcidin expression through the modulation of C/EBPα activity (Oliveira et

al. 2009). In contrast, oxidative stress has been shown to decrease hepcidin expression,

with increased reactive oxygen species (ROS) promoting the hypoacetylation of histones

leading to decreased binding of C/EBPα and STAT3 to the hepcidin promoter (Choi et al.

2007).

Page 42: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

27

1.8 Cellular iron homeostasis Whilst the central role of hepcidin in the regulation of systemic iron homeostasis is well

established, distinct mechanisms that regulate cellular iron homeostasis have also been

identified, allowing for the safe transport and utilisation of iron. Recent studies suggest that

cellular and systemic iron regulatory mechanisms interact with each other to control iron

homeostasis at various levels within an organism (Hentze et al. 2010).

1.8.1 Transferrin Receptor 2 (TFR2) regulation

Iron homeostasis can also be controlled by the cellular regulation of various iron-related

proteins. The iron transporter and regulator, TFR2, has been shown to be regulated at

both a transcriptional and post-translational level, independent of its role in hepcidin

signalling. The murine TFR2 promoter can be activated by the erythroid/liver-related

transcription factors GATA, C/EBP, and erythroid Kruppel-like factor (EKLF). In contrast,

the GATA-1 cofactor, friend of GATA -1 (FOG-1), essential for erythrocyte maturation, was

show to repress the enhanced promoter activity induced by GATA-1 (Kawabata et al.

2001). Also, as mentioned previously, TFR2 protein can be post-translationally stabilised

by the binding of diferric transferrin (Robb and Wessling-Resnick 2004). The cellular

regulation of TFR2 is likely to have a major effect on systemic iron homeostasis via its

signalling to hepcidin, but only a modest effect on cellular iron transport due to its limited

involvement in this process (Chua et al. 2010).

1.8.2 Zrt- and Irt-like Protein 14 (ZIP14) regulation

The iron and zinc transporter ZIP14, is also regulated at a transcriptional level, with IL-6

inducing activation of the ZIP14 promoter (Liuzzi et al. 2005). ZIP14 has also been shown

to be transcriptionally upregulated by IL-1β, which through nitric oxide (NO) signalling

causes the transcription complex AP-1 to translocate to the nucleus where it may to bind

to two putative AP-1 binding sites in the ZIP14 promoter (Lichten et al. 2009). ZIP14 is

also regulated post-translationally, as it has been demonstrated that the presence of HFE

protein results in decreased ZIP14 protein levels, due to decreased stability of the ZIP14

Page 43: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

28

protein rather than decreased mRNA levels (Gao et al. 2008). The cellular upregulation of

ZIP14, may play an important role in iron homeostasis during the inflammatory response

and may be responsible for the anaemia of inflammation.

1.8.3 Iron Regulatory Element (IRE)/Iron Regulatory Protein (IRP) regulation

In contrast to TFR2 and ZIP14, many proteins involved in cellular iron uptake, utilisation,

storage and transport are controlled by post-transcriptional mechanisms involving the

IRE/IRP system. Several key proteins of iron metabolism are encoded by mRNAs

containing one or more IREs in their untranslated regions (UTRs). These evolutionary

conserved hairpin structures are binding sites for the two homologous iron regulatory

proteins, IRP1 and IRP2, which are activated for IRE-binding during cellular iron deficiency

(Wallander et al. 2006). IRE/IRP interactions inhibit translation of the mRNAs encoding H-

and L-ferritin, the haem synthesis enzyme, ALAS2, and Fpn, which contain a single IRE in

their 5’ UTR. In addition, IRE/IRP interactions stabilise TFR1 and DMT1 mRNA, which

contain IREs in their 3’ UTR. In conditions of cellular iron deficiency, the IRE/IRP

homeostatic mechanism allows for increased iron uptake and transport via TFR1 and

DMT1 whilst preventing haem synthesis by ALAS2 (Malicka-Blaszkiewicz and Kubicz

1979) and the storage of iron in ferritin and its efflux via FPN. With increasing iron

concentration, IRPs are prevented from binding to IREs. In IRP1, increased iron levels

leads to the reversible insertion of a cubane 4Fe-4S cluster that converts IRP into a

cytosolic aconitase (Wallander et al. 2006). In contrast, increased iron levels result in IRP2

undergoing iron- and oxygen-dependent degradation following ubiquitination by F-

box/LRR-repeat protein 5 (FBXL5), which senses iron levels via a Fe-O-Fe centre within

its haemerythrin domain (Moroishi et al. 2011). IRPs have also been shown to respond to

iron-independent signals, with both IRP1 and IRP2 induced upon exposure of cells to H2O2

(Hausmann et al. 2011) and NO (Wang et al. 2005), stimulating TFR1 expression and iron

uptake.

Page 44: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

29

1.8.4 Ferroportin internalisation by hepcidin

The post-translational regulation of FPN has been shown to occur through the binding of

hepcidin to FPN leading to the phosphorylation, internalisation, ubiquitination, and

eventual degradation of FPN (De Domenico et al. 2007). The binding site for hepcidin has

been identified as an extracellular loop in FPN, of which cysteine residue 236 has been

shown to be essential for hepcidin binding (Fernandes et al. 2009). With the dimeric nature

of FPN (De Domenico et al. 2007), each monomer must bind hepcidin for hepcidin-

mediated internalisation. The decrease in FPN protein expression results in decreased

iron release and iron sequesteration in the enterocyte, hepatocyte and macrophage.

1.9 Hereditary haemochromatosis Hereditary haemochromatosis (HH) is an autosomal recessive disorder in which

abnormally high absorption of dietary iron leads to iron accumulation in the parenchymal

tissues. However in the early stages of the disease, macrophages are spared from iron

loading. Excessive iron accumulation is most often observed after an individual reaches

the age of forty, and is most prominent in the liver, pancreas, pituitary, heart, joints and

skin. Accumulation of iron in these areas may lead to liver fibrosis, cirrhosis and

hepatocellular carcinoma, diabetes mellitus, impotence, cardiac failure, arthritis and skin

hyperpigmentation (Siddique and Kowdley 2012). Phlebotomy to remove excess iron is an

effective treatment during the early stages of the disease. HH is a genetically

heterogenous disorder, classified into four types (Olynyk et al. 2008).

1.9.1 Hereditary haemochromatosis (HH) type 1

Type 1 or classical HH is the most common form of the disease. HH type 1 is an

autosomal recessive disorder caused by two common missense mutations, C282Y and

H63D in the HFE gene on chromosome 6p (Feder et al. 1996). Most patients with HH type

1 are homozygous for a C282Y substitution or heterozygotes for C282Y/H63D mutations.

HH type 1 is one of the most prevalent genetic diseases and occurs at a frequency of 1 in

Page 45: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

30

200 individuals of Northern European descent (Olynyk 1999). Genetic factors, as well as

environmental factors such as alcohol consumption may contribute to the degree of iron

overload. Patients with HH type 1 lack the ability to downregulate iron absorption and iron

is absorbed via the duodenum at a high rate, irrespective of the body’s iron stores,

resulting in increased serum transferrin saturation and progressive accumulation of body

iron (Andersen et al. 2004). Patients with HH type 1 initially deposit iron in the

parenchymal tissues, primarily in the hepatocytes of the liver, and subsequently in other

organs such as the pancreas and heart leading to irreversible tissue damage. However,

studies have shown that the macrophages of the spleen are comparatively resistant to

dietary iron loading (Harrison 2003).

To study the in vivo consequences of HFE deletion or mutation, five different gene

disruption have been produced in the mouse: a C282Y knock-in (Levy et al. 1999), a H63D

knock-in (Tomatsu et al. 2003), an exon 2-3 knockout (Bahram et al. 1999), an exon 3

disruption/exon 4 knockout (Levy et al. 1999), and the exon 4 knockout (Zhou et al. 1998)

used in this study (Hfe-/-). HFE knockout mice exhibit decreased hepcidin expression

(Ahmad et al. 2002), elevated transferrin saturation (Zhou et al. 1998), and increased

intestinal iron absorption (Ajioka et al. 2002), and as with human HH Type 1 patients, HFE

knockout mice demonstrate relative sparing of iron accumulation in macrophages (Zhou et

al. 1998).

1.9.2 Hereditary haemochromatosis type 2

Juvenile or HH type 2 is characterised by an early onset of iron overload that leads to

severe organ impairment usually before 30 years of age (Camaschella 2002). The disorder

was first described in 1979 and is a recessive trait that affects both sexes.

Hypogonadotrophic hypogonadism and cardiac involvement are prominent features of the

clinical syndrome. The daily increase in iron absorption and the rate of iron accumulation

are higher in HH type 2 HH than in type 1. The first causative gene identified was HAMP1

Page 46: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

31

located on chromosome 19q13.1(Roetto et al. 2003) that encodes a peptide, which plays a

key role in the regulation of iron absorption. HH type 2 caused by mutations in the hepcidin

gene is referred to as Type 2B.

To investigate the effects of hepcidin disruption, a mouse with deletion in almost the entire

coding sequence of the Hamp1 gene was generated, with Hamp1 knockout mice

developing early and severe multi-organ iron overload (Lesbordes-Brion et al. 2006), with

the characteristic sparing of the splenic macrophages.

However, for many years it has been known that the most common cause of Type 2 HH

was linked to chromosome 1q, and in 2002 the causative gene was identified and named

haemojuvelin (HJV). A number of mutations in HJV have been identified. The most

common mutations are missense, frameshift, or the insertion of premature stop codons.

Type 2 HH caused by mutations in the HJV gene is referred to as Type 2A. Patients

suffering from Type 2A HH show the same pathology as Type 2B, with the premature

appearance of iron overload (Camaschella et al. 2002).

The Hjv knockout mouse is a model of HH type 2A, which exhibits decreased hepcidin

expression, increased FPN protein in intestinal enterocytes and macrophages (Huang et

al. 2005), and increased iron deposition in the liver, pancreas and heart (Niederkofler et al.

2005).

1.9.3 Hereditary haemochromatosis type 3

HH type 3 is a rare recessive disorder, which leads to iron overload and severe clinical

complications similar to those reported in HH type 1. HH type 3 is caused by mutations in

TFR2, located on chromosome 7q22 (Kawabata et al. 1999). HH type 3 was originally

characterised in two Sicilian families, where several members were found to be

homozygous for a nonsense mutation (Y250X) in TfR2 (Camaschella et al. 2000). In

Page 47: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

32

recent years other mutations in TFR2 have been characterised, spread along the entire

sequence of the gene. The clinical phenotype of HH type 3 is similar to HH type 1 with

increased plasma iron and transferrin saturation and parenchymal iron overload.

To investigate HH type 3, two mouse models have been generated. Firstly, as used in the

current study (Tfr2mut), a mouse with a Y245X mutation in Tfr2 (Fleming et al. 2002), a

murine orthologue of the human Y250X mutation, and secondly, a Tfr2 knockout mouse

(Tfr2-/-) (Wallace et al. 2005). Both Tfr2mut and Tfr2-/- mice exhibit inappropriately low

hepcidin levels and hepatic iron overload (Wallace et al. 2005; Drake et al. 2007). In

addition, Tfr2mut mice have been shown to have increased iron absorption, elevated levels

of iron transport genes in the duodenum, and increased liver iron uptake (Drake et al.

2007).

1.9.4 Hereditary haemochromatosis Type 4

HH type 4 is also referred to as FPN disease. Type 4 HH is due to heterozygous mutations

in the FPN gene on chromosome 2q32, which causes mutations in the FPN protein, which

in turn mediates cellular iron export. Type 4 HH differs from other forms of HH for several

reasons. Firstly, it is inherited as an autosomal dominant trait and secondly, patients with

Type 4 HH have high serum ferritin levels but low to normal transferrin saturation. Thirdly,

particularly in young patients there is iron deposition in the reticuloendothelial cells rather

than hepatocytes as found in other type of HH. Patients with Type 4 HH also exhibit

reduced tolerance for phlebotomy and have mild iron-deficient anaemia (Pietrangelo

2004). Of the two forms of HH Type 4, Type 4A is caused by mutations within ferroportin

itself, resulting in an inability to export iron from the cell. Whilst Type 4B is caused by

mutations in the hepcidin binding site of ferroportin, resulting in hepcidin being unable to

degrade ferroportin and as a consequence leading to excessive iron release (Pietrangelo

2004).

Page 48: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

33

Currently, there exists only one murine model of HH type 4 and deletion of ferroportin is

embryonically lethal (Donovan et al. 2005). The flatiron mouse, which has the H23R

missense mutation in FPN, exhibits defects in the localisation and export of iron by FPN

(Zohn et al. 2007), resulting in the iron loading of Kupffer cells, high serum ferritin levels,

and low transferrin saturations, similar to patients with HH type 4A.

1.10 Pathogenesis of HH In HH types 1, 2A and 3, one or more components of the plasma iron-sensing machinery

fail, and adequate levels of hepcidin are not produced in response to increased levels of

iron, resulting in increased intestinal and macrophage iron release. When a functional form

of HAMP is expressed at appropriate levels and HFE, TFR2 and HJV are functional, the

amount of iron released into the blood is appropriate for cellular requirements. Disruption

of HFE or TFR2 in HH type 1 and 3 increases the amount of iron that enters the

bloodstream. However, BMP/SMAD signalling is still sufficient to enable some expression

of hepcidin. Therefore, plasma iron loading will proceed at a slower rate and the build up of

iron in parenchymal tissues will be more gradual. The phenotype of iron overload

associated with loss of HJV (HH Type 2B), which is required for hepcidin signalling, is

more severe and similar to that associated with loss of hepcidin itself (HH Type 2A). When

hepcidin expression is normal, mutations in FPN, either in domains that interact with

hepcidin or those that allow FPN to be internalised following hepcidin binding, can result in

a insensitivity to hepcidin and HH type 4.

1.11 Iron induced liver injury Studies have shown that when hepatic iron concentration exceeds 60 µmol/g, hepatic

stellate cells begin to exhibit early signs of activation, an integral event in the initiation of

hepatic fibrosis. As hepatic iron levels increase further, the risk of significant liver fibrosis

Page 49: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

34

and ultimately cirrhosis increases (Ramm et al. 1997). Although the exact mechanisms of

liver injury induced by iron overload have not yet been fully elucidated, it is thought that the

accumulation of excess iron-catalysed ROS plays a significant role (Sochaski et al. 2002).

1.11.1 Generation of reactive oxygen species

In almost all cases of oxidative stress the initial reactive oxygen intermediate is the

superoxide radical (O2

•-) which is rapidly converted to hydrogen peroxide (H2O2) by the

action of superoxide dismutases (SODs). Neither O2

•- nor H2O2 are strong oxidising agents

and can usually only interact directly with iron and iron-containing molecules. However,

when redox-active iron is available, the Fenton and Haber-Weiss reactions take place,

where O2

•- reduces Fe3+ to Fe2+, which then reacts with H2O2, resulting in the reactive and

highly damaging hydroxyl radical (OH•)(Aruoma et al. 1991). In the presence of the radical

NO. (produced by nitric oxide synthase) and elevated O2

•-, the peroxynitrite radical (ONOO-

) is formed, which can then react with iron to produce the reactive NO2

+ species (Videla et

al. 2003). Both OH• and NO2

+ are highly reactive and central to the process of lipid

peroxidation and damage to DNA and cellular protein.

1.11.2 Lipid peroxidation

It is suggested that the OH• radical damages the polyunsaturated fatty acids within lipid

membranes resulting in carbon-centred lipid radicals. The intramolecular rearrangement of

the double bonds within these radicals results in the formation of conjugated dienes, which

in the presence of oxygen, form lipid peroxyl radicals (Halliwell and Gutteridge 1984). Lipid

peroxyl radicals can auto-catalyse by reacting with more fatty acids and by forming lipid

hydroperoxide, which is susceptible to cleavage by ferrous and ferric iron molecules, then

decomposing to alkoxyl and peroxyl free radicals, respectively. Lipid hydroperoxides

undergo intramolecular cyclisation and decomposition to generate thiobarbituric acid

(TBA)-reactants and breakdown by-products malondialdehyde (MDA), 4-hydroxynonenal

(4-HNE), ketones, alcohol, ethane and pentane (Bacon and Britton 1990).

Page 50: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

35

1.11.3 Lysosmal fragility

In patients with iron overload, the accumulation of iron within lysosomes is commonly

observed (Iancu and Shiloh 1994) and is believed to result from the receptor-mediated

uptake of transferrin (Iacopetta et al. 1983) and ferritin-bound iron (Britton et al. 1994).

Sequestration of iron within the lysosome is believed to be a protective mechanism,

removing iron from redox-sensitive sites and providing a mechanism of excretion via the

biliary system (Britton et al. 1994). Peroxidation of lipids is believed to play a major role in

lysosomal fragility with both ferritin and haemosiderin shown to induce lipid peroxidation in

vitro (O'Connell et al. 1985).

1.11.4 Mitochondrial damage

Lipid peroxidation can also affect the membranes of mitochondria. Chronic iron overload in

vivo in rats (Bacon et al. 1993) and in isolated rat mitochondria (Bacon et al. 1986) has

been shown to have an inhibitory effect on the mitochondrial electron transport chain.

Studies have also shown that lysosomal enzymes can induce mitochondrial oxidant

production and cytochrome c release (Zhao et al. 2003).

1.11.5 DNA damage

Britton and colleagues (Britton et al. 2002; Fleming et al. 2002) have shown that excess

iron can induce DNA damage in both animal models of iron overload and in hepatocytes in

vitro. It has also been demonstrated in haemochromatotic liver that there are increased

levels of etheno-DNA adducts derived from the interaction of lipid peroxidation products

and DNA (Nair et al. 1998), which are associated with the increased frequency of

mutations in the tumour suppressor gene p53 (Hussain et al. 2000). During chronic iron

overload lipid peroxidation produces DNA-reactive aldehydes, which can deregulate

cellular homeostasis and induce malignancy (Nair et al. 1995).

Page 51: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

36

1.11.6 Oxidative stress

Previous studies have demonstrated increased hepatic levels of antioxidants, such as

glutathione (Reardon and Allen 2009; Mizukami et al. 2010), and decreased levels of non-

enzymatic antioxidants such as ascorbate, β-carotene and vitamins E and A in iron

overload conditions (Livrea et al. 1996). It has also been demonstrated that iron induced

oxidative stress can initiate apoptosis and necrosis, promoting the synthesis and release

of proinflammatory and fibrogenic factors that alter Kupffer cell (Lin et al. 1997; Xiong et al.

2003) and hepatocyte functions, triggering the activation of hepatic stellate cells and

fibrogenesis (Parola and Robino 2001).

1.11.7 Inflammatory cytokines

Studies have shown that iron overload results in mild hepatic inflammation in response to

apoptotic and necrotic hepatocytes (Deugnier et al. 1992). Inflammatory cells produce a

variety of cytokines involved in the hepatic response to injury. Among these, the

proinflammatory molecule TNF-α , and the anti-inflammatory cytokine IL-10, have emerged

as key factors in iron-induced liver disease (Ramm and Ruddell 2010).

1.12 Present study HH is a primary genetic disorder of iron homeostasis, which results in the hyperabsorption

of dietary iron, leading to iron accumulation in tissues of the body and iron-induced injury

and organ dysfunction (Pietrangelo 2006). Although the aberrant absorption of iron occurs

in the duodenum, mouse models have demonstrated that in most types of HH, the primary

cause of the disorder is the hepatocyte (Verga Falzacappa et al. 2007). It is well

established that hepatocytes take up iron from TBI and NTBI. TBI is taken up by

hepatocytes by TFR1 with high-affinity and to a lesser degree by TFR2 with low-affinity

(Trinder et al. 1996; Chua et al. 2008). NTBI is potentially very toxic as it can generate

ROS. In HH, NTBI is found mainly in the form of iron citrate (Grootveld et al. 1989). In iron

Page 52: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

37

overload diseases, such as HH, NTBI uptake becomes important as transferrin saturation

reaches 100% and co-exists with iron citrate in the serum of untreated patients. It is likely

to be an important source of iron accumulation by the liver in iron overload.

Aims

The general aim of this project is to characterise the role of HFE and TFR2 in iron

transport and the regulation of iron metabolism as well as the mechanisms of iron-induced

liver injury caused when these proteins are impaired in HH. The study will utilise the Hfe

knockout (Hfe-/-), Tfr2 Y245X mutant (Tfr2mut) and double mutant (Hfe-/-xTfr2mut) mouse

models of HH.

Hypothesis 1:

Disruption of HFE and TFR2 in combination will result in a form of iron overload that is

more severe than disruption of HFE or TFR2 alone. HFE and TFR2 act in separate as well

as the same signalling pathways as mutations in both genes have an additive inhibitory

effect on hepcidin synthesis causing a more severe iron loading.

Aim 1:

To determine if disruption of HFE and TFR2 in combination results in a form of iron

overload that is more severe than disruption of HFE or TFR2 alone. This aim will be

addressed by cross-breeding Hfe-/- and Tfr2mut mice to produce a Hfe-/-xTfr2mut mouse. The

phenotype including physical characteristics, haematology, iron status (liver iron, plasma

iron and NTBI levels) and expression of key iron metabolism genes will be examined in the

Hfe-/-xTfr2mut mouse and compared with the levels in Hfe-/-,Tfr2mut, wild-type animals (WT)

and dietary iron-supplemented WT mice (WT+Fe).

Page 53: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

38

Hypothesis 2:

TFR2 and HFE play an important role in sensing body iron levels, and control iron

metabolism by regulating hepcidin expression, which is impaired in Hfe-/-, Tfr2mut, and Hfe-/-

xTfr2mut mice, leading to increased plasma iron levels and iron deposition in the liver. The

excess iron is taken up by the hepatocyte in the form of NTBI.

Aim 2:

To characterise the mechanisms of hepatic NTBI uptake in Hfe-/-, Tfr2mut, and Hfe-/-xTfr2mut

mice.

This aim will be addressed by measuring plasma NTBI clearance and tissue uptake in vivo

Hfe-/-, Tfr2mut, and Hfe-/-xTfr2mut mice. Tissue NTBI uptake will be correlated with plasma

NTBI levels and tissue iron content, measured by Inductively coupled plasma atomic

emission spectroscopy (ICP-AES).

Hypothesis 3:

In HH mouse models, increasing hepatic iron deposition leads to iron-induced injury, which

recapitulates the injury seen in human HH.

Aim 3:

To examine the iron-induced hepatic pathology associated in the murine models of HH.

This aim will be addressed using Hfe-/-, Tfr2mut, and Hfe-/-xTfr2mut mouse models of HH.

Measurement of iron parameters will be correlated with expression of iron metabolism and

liver injury genes measured by reverse transcription polymerase chain reaction (PCR). In

conjunction, histological, biochemical, and immunofluorescent techniques will be used to

assess lipid peroxidation, collagen deposition and inflammation and the degree of hepatic

injury in the murine models of HH.

Page 54: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

39

Hypothesis 4:

Inflammation will increase hepatic NTBI transport in Hfe-/-, Tfr2mut, and Hfe-/-xTfr2mut mouse

models by activation of the IL-6 cytokine pathway, resulting in increased levels of

inflammation-regulated iron transporters and increased iron loading of the liver.

Aim 4:

To investigate the effect of inflammation on the regulation of iron metabolism and iron

transporters in mouse models of HH.

This aim will be addressed using Hfe-/-, Tfr2mut, and Hfe-/-xTfr2mut mice treated with

lipopolysaccharide (LPS) to stimulate IL-6 production. Iron status levels (plasma iron,

transferrin saturation and NTBI levels) will be correlated with iron metabolism gene

expression measured by reverse transcription-PCR.

Page 55: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

40

Chapter 2

Materials and Methods

Page 56: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

41

Materials

All materials used in this investigation are listed with their supplier below. All reagents and

chemicals are of analytical grade unless specified.

2.1.1 Tissue collection

Material Supplier

Anaesthetics (Ketamine; Xylazine) Troy Laboratories

Heparin Pharmacia and Upjohn

Mouse chow (70 mg or 200mg iron/kg) Specialty Feeds

2.1.2 Experimental procedures

Material Supplier

Acetic acid, glacial BDH

Bathophenanthroline disulfonic acid Sigma

BCA Protein Assay Kit Pierce

Centricon ultrafilters Millipore

HCl BDH

Iron (III) chloride Sigma

Iron (II) sulphate.7H20 BDH

Microfuge tubes Quantum Scientific

MOPS Sigma

Nitriloacetic acid Sigma

Pipette tips Greiner

Serological pipettes Sarstedt

Sodium bicarbonate BDH

Syringes BD Sciences

Thioglycollic acid Sigma

Trisodium Citrate BDH

Triton X-100 BDH

Page 57: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

42

2.1.3 Molecular biology

Material Supplier

Agarose Promega

Superscript III Invitrogen

1-Bromo-3-chloropropane Sigma

DNA freeTM kit Ambion

Ethanol Sigma

Isopropanol Sigma

Microfuge tubes (DNAse/RNAse free) Quantum scientific

dNTP Invitrogen

Nuclease free water Invitrogen

oligoDT Invitrogen

Pipette tips (DNAse/RNAse free) Axygen/Fisherbiotec

SYBR Green Master Mix Roche

Primers Geneworks

Invitrogen

Tri-reageent Ambion

RNAsin Promega

2.1.4 Protein extraction and Western blotting

Material Supplier

Complete Mini tablets Roche

PhosphoSTOP Roche

Tris-HCl Sigma

SDS Biorad

Glycerol BDH

Β-Mercaptoethanol Sigma

Bromophenol Blue Sigma

MagicMark XP Invitrogen

Novex Sharp Protein Standard Invitrogen

NuPage Bis/Tris 4-12% gel Invitrogen

MOPS SDS running buffer Invitrogen

NuPAGE transfer buffer Invitrogen

Page 58: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

43

Methanol

Nitrocellulose Membrane Fisherbiotec

Antibodies

Rabbit anti-human ferritin DAKO

Goat polyclonal actin Santa Cruz

Donkey anti-goat IgG horseradish peroxidase Santa Cruz

Goat anti-mouse IgG horseradish peroxidase Santa Cruz

Goat anti-rabbit IgG horseradish peroxidase Santa Cruz

Coomassie brilliant blue R-250 Sigma

Western Lightening Detection Perkin-Elmer

PBS ThermoElectron

Novablot electrode paper Amersham

Tween 20 BDH

2.1.5 Equipment

2.1.5.1 Balances

All analytical and biochemical reagents were measured using either an A&D ER-120A or

FX-2000 balances (A&D Mercury Pty Ltd, SA, Australia).

2.1.5.2 Centrifugation

Eppendorf microfuges were used for molecular biology techniques.

2.1.5.2 Imaging system

Images of RNA gels and immunoblots were captured using a VersaDoc imaging system

(Bio-Rad, NSW, Australia).

2.1.5.3 Microscope

A Nikon Eclipse TE-2000U phase contrast microscope (Coherent Scientific, SA, Australia)

and a CK-2 Olympus inverted microscope (Olympus Australia Pty Ltd, Mt Waverley, NSW,

Australia) was used to observe liver pathology.

Page 59: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

44

2.1.5.4 Peristaltic pump

A Gilson Minipuls 3 pump (John Morris Scientific, Willoughby, NSW, Australia) was used

for tissue perfusion.

2.1.5.5 pH measurement

A Corning 220 pH meter (Crown Scientific Pty Ltd, Moorebank, NSW, Australia) was used

for all pH measurements. Radiometer buffer solutions were used to calibrate the pH meter.

2.1.5.6 Pipettes

Volumes over 10 mL were measured using a Drummond pipette aid (Drummond Sci Co,

Broomall, PA, USA) and graduated serological pipettes or measuring cylinders. Gilson

(John Morris Scientific, WA, Australia) or Eppendorf (Eppendorf, NSW, Australia)

micropipettors were used to measure volumes of 1 mL or less. A Genex-Beta (Unimed

Australia Pty Ltd, Jandakot, WA, Australia) multichannel variable volume pipette was used

in bicinchoninic acid (BCA) protein assay.

2.1.5.6 Powerpacks

An EPS-301 electrophoresis power supply unit (Amersham Pharmacia Biotech, Castle Hill,

NSW, Australia) was used in conjuction with a semi-dry electroblotter to transfer proteins

onto nitrocellulose membranes. Agarose gels were run in Bio-Rad gel tanks using a Power

Pac 300 power supply (Bio-Rad, NSW, Australia).

2.1.5.7 Radioactivity measurements

Iron-59 (59Fe) samples were counted for 10 min on a Wallac 1480 WizardTM γ-counter

(Perkin Elmer, VIC, Australia).

2.1.5.8 Spectrophotometry

Plasma iron and NTBI concentrations were measured on a Beckman-Coulter DU-640

spectrophotometer (Beckman Coulter Pty Ltd, Gladesville, NSW, Australia). BCA protein

assay measurements were performed spectrophotometrically on a BMG Fluoristar Optima

microplate reader (BMG Labtechnologies Pty Ltd, Mt Eliza, VIC, Australia) and RNA was

quantitated by measurement of absorbance at 260 and 280 nm using a Nanodrop 1000

(Thermo Scientific).

Page 60: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

45

2.1.5.9 Real-time PCR

Real-time PCR assays were performed on a Rotor-Gene (QTM or R-G3000TM; Qiagen,

NSW, Australia).

2.1.5.10 Thermocycler

cDNA was amplified on a PTC-100 thermocycler (MJ Research Inc, San Francisco, CA,

USA).

2.1.5.11 Western blot transfer apparatus

Protein samples were electroblotted onto nitrocellulose membranes using the PantherTM

Semi-dry Hep-1 Electroblotter (Owl, Portsmouth, NH, USA).

2.1.6 Location of suppliers

Amersham Pharmacia Biotech, SA, Australia

Axygen Scientific Inc, Mt Martha, VIC, Australia

BDH AnalaR, Kilsyth, VIC, Australia

BD Sciences, Sydney, NSW Australia

Bio-Rad, Regents Park, NSW, Australia

Boehringer Mannheim, Castle Hill, NSW, Australia

DAKO, Glostrup, Denmark

Eppendorf, Clayton, VIC, Australia

Fisons Scientific Equipments, Loughborough, England, UK

GeneWorks, Thebarton, NSW, Australia

GibcoBRL, Invitrogen, Mount Waverley, VIC, Australia

Glen Forrest Stockfeeders, WA, Australia

Greiner Labortechnik Ltd, Kremsmünster, Germany

Invitrogen, Mount Waverley, VIC, Australia

Millipore, North Ryde, NSW, Australia

Pharmacia and Upjohn, WA, Australia

Pierce, Rockford, IL, USA

Promega, Annandale, NSW, Australia

QSP, Quantum Scientific, Milton, QLD, Australia

Santa Cruz Biotechnology; Santa Cruz, CA, USA

Sarstedt AG & Co, Technology Park, SA, Australia

Page 61: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

46

Sigma Chemical Co, St Louis, MO, USA

ThermoElectron Co, Melbourne, VIC, Australia

Worthington Biochem Corp, Freehold, NJ, USA

Zymed Laboratories Inc, San Francisco, CA, USA

General methods

2.2.1 Animals

Hfe-/- mice were generated by disruption of the Hfe gene using homologous recombination,

as described by Zhou et al (Zhou et al. 1998). Tfr2mut mice were generated using targeted

mutagenesis to introduce a premature stop codon (C735G) into the murine Tfr2 coding

sequence. The resulting Y245X mutation is orthologous to the Y250X mutation identified in

some patients with HH type 3 (Fleming et al. 2002). Hfe-/- and Tfr2mut mice were

backcrossed for ten generations on to an AKR background at the Animal Resource Centre

(Murdoch, WA, Australia). Hfe-/- and Tfr2mut mice were then crossed to generate Hfe-/-

xTfr2mut double mutant mice. Female Hfe-/-, Tfr2mut, Hfe-/-xTfr2mut and wild-type mice were

fed standard mouse chow (100 mg iron/kg diet; Specialty Feeds, Glen Forrest, WA,

Australia) ad libitum from four weeks of age. An additional group of wild-type mice were

fed an iron-supplemented diet (20 g carbonyl iron/kg diet; Specialty Feeds) for three weeks

from 8-10 weeks of age. Following overnight fasting, blood was collected by cardiac

puncture and organs were perfused in situ with isotonic saline. Tissues were collected and

snap frozen in liquid nitrogen, OCT or fixed in formalin. This study was approved by The

University of Western Australia Animal Ethics Committee.

Page 62: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

47

2.2.2 ICP-AES

1 mL of liver homogenate (whole liver homogenised with 1 mL saline), or whole kidney,

pancreas and heart from HH (Hfe-/-, Tfr2mut and Hfe-/-xTfr2mut), non-iron loaded and iron-

loaded WT mice were digested in concentrated 1 mL 12M nitric acid and 1 mL 35%

hydrogen peroxide at 110°C, until volume was halved. The digestion was repeated three

times before samples were made up to 10 mL with Milli-Q water. Measurement of tissue

metals content was performed by Inductively Coupled Plasma Atomic Emission

Spectroscopy using a Varian (Vista AX) ICP-AES CCD Simultaneous, according to AS

3641.2-1999 (Standards Australia International, Strathfield) by the Marine and Freshwater

Research Laboratory (Murdoch University, Perth, WA, Australia).

2.2.3 Plasma Iron Assay

Plasma iron was measured in all samples according to the method described by Fielding

(Fielding 1980). Samples were removed from -80ºC storage and placed on ice until used.

An iron standard solution was prepared from a 20 mM iron stock solution of ferric chloride

dissolved in 1 M HCL. The standard was then diluted 1:4 with Milli-Q water to give a

solution of 280 µL/mL iron in 250 mM HCl, this was then further diluted 1:70 with Milli-Q

water to give a final concentration of 4 µg/mL iron in 7 mM HCl. Dilutions of the stock

solution in Milli-Q water were made to give concentrations ranging from 0-4 µg/mL iron.

50 µL of the plasma or standard were transferred to iron free tubes and an equal volume of

protein precipitate solution consisting of 50% TCA, TGA, HCl (12M) in Milli-Q water was

added to each tube. Samples and standards were mixed thoroughly by vortexing for 30

seconds and incubated at room temperature for 15 min. The samples and standards were

then centrifuged at 14,000 rpm for 15 min. 50 µL of the supernatant was transferred to new

tubes and 50 µL of chromogen solution consisting of sodium acetate, BPS, and Milli-Q

Page 63: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

48

water was added. The samples and standards were mixed briefly and allowed to stand at

room temperature for 20 min. Absorbance of each sample and standard was measured

using the Beckman DU 640 spectrophotometer at a wavelength of 535 nm, using Milli-Q

water as a blank. Plasma iron concentrations were then calculated using the standard

curve.

2.2.4 Total Iron Binding Capacity (TIBC)

TIBC was determined in all plasma samples. 50 µL of plasma was transferred to an iron

free tube and 50 µL of saturating iron solution consisting of stock ferric chloride solution

(20 mM) and Milli-Q water was added slowly, to saturate the transferrin with iron. Samples

were mixed thoroughly by vortexing and allowed to stand at room temperature for 15 min.

The samples were centrifuged briefly and 7.5 mg of light magnesium carbonate added to

precipitate any unbound iron. Samples were then mixed gently and placed on a rotating

turntable for 60 min. The samples were then centrifuged at 14,000 rpm for 10 min to

precipitate the magnesium carbonate and the supernatant was transferred to a new tube

and centrifuged at 14, 000 rpm for 7 min. The iron content in 50 µL of the supernatant was

then assayed by the method used in the Plasma Iron Assay.

TIBC concentrations were calculated using the same standard curve generated in the

Plasma iron assay. The iron concentration (µg/mL) of the TIBC samples must be multiplied

by a factor of 2 to account for the 1:2 dilution of the plasma with iron saturating solution in

the TIBC assay, prior to the Plasma Iron Assay.

The data obtained from both the Plasma Iron Assay and the TIBC assay were used to

determine the level of transferrin saturation in the sample using the following formula:

Transferrin Saturation (%) = [Plasma Iron] X 100

[TIBC] 1

Page 64: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

49

Levels of transferrin saturation were then corrected for non-transferrin bound iron (NTBI)

by subtracting the NTBI concentration from the transferrin saturation.

2.2.5 Non-Transferrin Bound Iron (NTBI) Assay

Analysis of NTBI levels in HH and wild-type mice was conducted according to the methods

described by Gosriwatana et al (Gosriwatana 1999).

2.2.5.1 Preparation of Tris-carbanatocobaltate(III) trihydrate

A cold slurry of 2.1 g NaHCO3 and 2.5 mL Milli-Q water was prepared at 4ºC. 1.45 g of

cobalt(II)nitrate.6H20 was dissolved in 2.5 mL Milli-Q water and 0.5 mL of 30% H202 was

added dropwise. Cobalt(II)nitrate solution was added to the NaHCO3 solution and

continuously stirred for 1 h at 4ºC. A green precipitate developed and this was separated

by filtration and the precipitate was washed 3 times with 10 mL cold Milli-Q water. After

washing, 1 mL of 100% ethanol was added drop-wise to the precipitate. After all the liquid

was filtered, the precipitate was transferred to a clean filter paper and allowed to dry for 1

h at room temperature, and then for another 1 h at 37ºC. The dry precipitate was removed

from the filter paper and placed in a clean bottle with 10 mL 1M NaHCO3 and stirred for 2 h

at room temperature. The solution was filtered again and the filtrate allowed to stand at

4ºC for 2 days before use. Prior to use the solution was diluted 1:3 with additional NaHCO3

solution.

Frozen plasma samples were removed from -80ºC storage and placed in a water bath at

37ºC for 10 min and the thawed samples were then briefly centrifuged at 13,000 rpm for 30

seconds. 80 µL of plasma was transferred to a clean tube and 17.8 µL of the Tris-

carbanatocobaltate solution was added to each sample and incubated for 60 min in a 37ºC

water bath to saturate all unoccupied iron binding sites with cobalt. 10.85 µL of 800 mM

NTA was added to each sample and incubated at room temperature for 30 min to chelate

Page 65: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

50

all non-transferrin bound iron. The Amicon Microcon-30 Centrifugal Filter Devices with a

30,000 molecular weight cutoff were washed with 500 µL of Milli-Q water and then

centrifuged at 14,000 rpm for 12 min. The filters were then turned upside down and

centrifuged again at 2000 rpm for 6 min taking care not to dry out the filters. Samples were

briefly centrifuged before being transferred to the filters and centrifuged for 12 min at 4,000

rpm to separate transferrin bound iron from non-transferrin bound iron. A standard curve

was produced ranging from 0-10 µM iron, using a 50 µM FeCl3 stock solution, made

freshly each time from a 20 mM stock solution.

30 µL of 5mM MOPS buffer at pH 7.4 was added to an equal volume of samples and

standards, followed by 7.5 µL of 120 mM thioglycolic acid. Finally, 7.5 µL of 60 mM BPS

was added and the samples and standard allowed to stand for 30 min at room

temperature. For each sample, a duplicate was prepared containing no BPS and used as

a reagent blank. After 30 min the optical density of the samples was measured using the

Beckman DU 640 spectrophotometer at a wavelength of 535nm, with Milli-Q water used as

a blank.

The line of best fit derived from the standard curve was used to calculate the concentration

of each sample and the concentration of the reagent blank was subtracted from the

concentration in each sample.

2.2.6 RNA extraction

Total RNA was isolated from tissues and gene expression was measured using RT-PCR.

Tissues were homogenised with cold TRI reagent to release RNA from the cells. The

lysates were transferred to 1.5 mL low adhesion tubes and left for 5 min at room

temperature. 1-bromo-3-chloropropane was added to each tube and shaken vigorously for

Page 66: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

51

30 seconds. Tubes were then incubated (room temperature, 10 mins) prior to

centrifugation (16100 g, 4oC, 15 mins) to separate RNA (aqueous phase) from DNA and

protein (organic phase). After centrifugation, the aqueous phase was collected and

transferred to fresh 0.6 mL low adhesion tubes. An equal volume of isopropanol was

added to the tubes to precipitate the RNA (room temperature, 10 mins) then centrifuged

(16100 g, 4oC, 15 mins). The supernatant was removed, and the pellet was washed once

with 75% ethanol and centrifuged (16100 g, 4oC, 15 mins). The supernatant was removed

and the pellet was air-dried and dissolved in 15 µL of nuclease free water.

2.2.7 DNase treatment of RNA

RNA samples were treated with DNase to remove any contaminating DNA. 0.1 volume of

10X DNase 1 buffer and 0.5 uL rDNase 1 (2 U/µL) were added to each sample, mixed and

incubated (37oC, 30 mins). Then 0.1 volume DNase inactivating reagent was added to

each sample and incubated (room temperature, 2 mins) with occasional mixing prior to

centrifuging (10,000 g, room temperature, 1.5 min) to pellet the inactivating agent. The

supernatant, containing the RNA, was transferred to fresh low adhesion tubes for storage

at -20oC for short term storage or -80oC for long term storage.

2.2.8 RNA quantification

The concentration and purity of RNA was determined by the measurement of absorbance

at 260 and 280 nm using a Nanodrop 1000 (Thermo Scientific) spectrophotometer using 1

µL of sample. The concentration of RNA was read at A260 where A260 = 1 defined a

concentration of 40 µg/mL. The purity of RNA was determined by the ratio A260/A280 with a

ratio of 1.8-2 representing pure RNA.

Page 67: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

52

2.2.9 Gel Electrophoresis

PCR products from samples were run together with 1 Kb Plus Molecular Weight Marker on

a 1.5% agarose gel at 70V for one h. Bands were visualised using a UV transilluminator

and a Kodak digital camera with KDS 120 software.

2.2.10 Reverse transcriptase-Polymerase Chain Reaction (RT-

PCR)

RNA extracted was reverse transcribed to produce complementary DNA (cDNA) and

amplified using polymerase chain reaction. Nuclease free water was added to 1 µg RNA to

a total volume of 5 µL and mixed with RT-Master Mix (Table 2.1) and the tubes were

placed in a PTC-100TM Programmable thermal controller (MJ Research INC.) thermocycler

at 65oC for 5 min to denature RNA. After 5 min, the tubes were placed in a cooling block

for 4 min followed by the addition of the RT-Enzyme Mix (Table 2.1) and incubated at 50oC

for one h. Reverse transcriptase was inactivated by incubating the samples at 70oC for 15

min. After the cycle has completed, the cDNA was stored at 4oC and used within 3 days of

synthesis.

Table 2.1: Reagents used for reverse transcription of RNA.

RT reagents Final Concentration

RT-Master Mix (6.75 µL total volume per reaction)

Oligo dT 25 µg/ mL

dNTPs 0.25 mM

Nuclease free water To volume

RT-Enzyme Mix (3.25 µL total volume per reaction)

RT Reaction buffer 1x

DTT 2.5 mM

RNaseOUTTM 1 U/ µL SuperscriptTM III Reverse

Transcriptase 10 U/ µL

Page 68: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

53

Quantification of mRNA transcripts was performed using the Rotorgene RG3000TM

(Corbett Research). Each PCR reaction contained 1 µL of template cDNA and 19 µL of

PCR Master Mix (Table 2.2). The cycling parameters are shown in Table 2.4. During each

cycle, double stranded DNA was denatured, followed by the annealing of primers to the

single stranded DNA template and extension of the primer catalysed by Taq polymerase.

Table 2.2: Reagents in the PCR Master Mix.

PCR reagents Final Concentration

PCR Master Mix (19µL) per reaction

Nuclease free water To volume

FastStart SYBR Green master (Roche)

0.5 X

Primer Mix containing forward and

reverse primer (5µM) (see Table 2.3)

0.25 µM

2.2.11 Primers

Table 2.3: Primer sequences and annealing temperatures.

GENE Primer sequence (5'-3') anneal. temp (°C)

m Beta-actin F-CTGGCACCACACCTTCTA 59

R-GGTGGTGAAGCTGTAGCC

mBMP6 F-ATGGCAGGACTGGATCATTGC 58

R-CCATCACAGTAGTTGGCAGCG

mDcytb F-CATCCTCGCCATCATCTC 56

R-GGCATTGCCTCCATTTAGCTG

mDMT F-TCTATCGCCATCATCCCCACCC 60

R-TCCACAGTCCAGGAAAGACAGACCC

mFPN F-GTCATCCTCTGCGGAATCATCCTGA 58

R-GAGACCCATCCATCTCGGAAAGTGC

mGDF15 F-GAGCTACGGGGTCGCTTC 58

R-K64GGGACCCCAATCTCACCT

mHJV F-TGGTTCTATCAATGGGGGCG 59

R-CACAGTAAAGTTGGGGTCACCG

mHamp1 F-TTGCGATACCAATGCAGAAGA 58

Page 69: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

54

R-GATGTGGCTCTAGGCTATGTT

mHFE F-CAGCTGAAACGGCTCCTG 58

R-CGAGTCACTTTCACCAAAGTAGG

mMetallothionein F-CACCAGATCTCGGAATGGAC 60

R-AGGAGCAGCAGCTCTTCTTG

mTransferrin F-CATAACTATGTCACTGCCATTCG 60

R-TCACTGGCGAGTTGTCGAT

mTfR2 F-CCGCTATGGAGACGTGGTT 60

R-TGGCGACACATACTGGGACAG

mZip14A F-TTCCTCAGTGTCTCACTGATTAA 54

R-GGAAAAGGGCGTTAGAGAGC

IL-6 F-GTATGAACAACGATGATGCACTTG 58

R-ATGGTACTCCAGAAGACCAGAGGA

IL-11 F-CTGCACAGATGAGAGACAAATTCC 58

R-GAAGCTGCAAAGATCCCAATG

Il-17a F-GCTCCAGAAGGCCCTCAG 58

R-CTTTCCCTCCGCATTGACA

TGF beta F-TGGAGCAACATGTGGAACTC 60

R-GTCAGCAGCCGGTTACCA

IL-1 alpha F-TTGGTTAAATGACCTGCAACA 60

R-GAGCGCTCACGAACAGTTG

TNF alpha F-CTGTAGCCCACGTCGTAGC 60

R-TTGAGATCCATGCCGTTG

IFN gamma F-ATCTGGAGGAACTGGCAAAA 60

R-TTCAAGACTTCAAAGAGTCTGAGGTA

Id1 F-AACGGCGAGCTCAGTGCCTT 58

R-GAGTCCATCTGGTCCCTCAGTG

Smad7 F'-ACCCCCATCACCTTAGTCG 58

R-GAAAATCCATTGGGTATCTGGA

c-myc F-CCTAGTGCTGCATGAGGAGA 58

R-TCCACAGACACCACATCAATTT

Page 70: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

55

Table 2.4: PCR cycling parameters.

Steps Temperature (oC) Time (sec) Number of cycles

Denature 95 600 1

Cycling

– Denaturation 95 15

– Annealing See Table 2.3 20 40

– Extension 72 20

Hold 72 30 1

Melting Curve 72-99 5

Hold 40 30 1

Plasmids containing full-length cDNA or PCR products of the gene of interest were

generated. Standard curves were measured from serial dilutions of known copy numbers

(calculated using known factors: 1 µg of 1000 bp DNA contains 1.52 pmol and 1 mole of

DNA has 6.23 x 1023 molecules) of the plasmids to quantify gene expression in HH and

wild-type mice. The specificity of the PCR product was determined by the melting curve

peak which represents the temperature at which the DNA strands separate for each gene

of interest. This was measured by heating the PCR product and observing the

fluorescence at 1°C increments from 72 to 99°C. mRNA expression of all iron transporters

and regulatory molecules were normalised against β-actin mRNA expression.

2.2.12 Protein extraction

Protein was extracted by homogenisation of tissue samples in 150 mM NaCl, 25 mM Tris-

HCl, pH 7.5 and 0.5% Triton X-100 plus Complete mini protease inhibitors (Roche,

Australia; 1 tablet per 10 mL lysis buffer). The homogenates were left for 1-2 h on ice

before being centrifuged for 15 min at 16000 g at 4°C and the supernatant collected.

Protein concentration of the cytoplasmic extract was then measured using BCA protein

assay.

Page 71: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

56

2.2.12.1 Bicinchoninic acid (BCA) protein assay

The Pierce BCA protein assay involves a two-step reaction that combines the reduction of

Cu2+ to Cu+ by protein in an alkaline environment and the selective colourimetric detection

of the Cu+-protein complex with BCA (Smith et al. 1985). Standards were prepared from

known concentrations of bovine serum albumin (BSA) solutions (0-600 µg/mL). Aliquots of

protein and standards (20 µL) were added to the wells of a 96-well tissue culture plate,

followed by the addition of 200 µL of the BCA working reagent (prepared fresh by mixing

49 parts Reagent A to 1 part Reagent B) to each well. The plates were then incubated at

37°C for 1 h and the absorbance measured in a microplate reader.

2.2.13 Statistical analysis

Results are expressed as mean ± SEM where n = 5-11 mice per group, unless otherwise

stated. Differences between groups were analysed using analysis of variance with Tukey’s

multiple comparison post-test or an unpaired Student’s t-test (GraphPad PRISM, La Jolla,

CA, USA). Differences between groups were defined as statistically significant for p < 0.05.

Page 72: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

57

Chapter 3

Characterisation of mouse models of hereditary

haemochromatosis

Page 73: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

58

3.1 Introduction Iron is an essential trace element with both iron deficiency and iron overload having severe

pathological consequences. For this reason, iron homeostasis is tightly controlled through

the regulation of duodenal iron absorption and iron release from macrophages and other

cell types, with the liver playing a central role in iron storage. A number of the proteins

involved in the regulation of iron homeostasis including HFE, TFR2 and hepcidin are highly

expressed in the liver and are mutated in the various types of HH as outlined in Chapter 1.

A characteristic finding in HH caused by mutations in either HFE or in TFR2 genes is the

decrease in levels of the liver iron regulatory peptide hepcidin compared with livers with

the same degree of liver iron loading caused by secondary iron overload in both humans

and mice (Bridle et al. 2003). This observation suggests that both HFE and TFR2 play a

role in the regulation of hepcidin expression. Recent studies have shown that HFE and

TFR2 are capable of forming a complex (Chen et al. 2007), and evidence suggests this

complex is involved in hepcidin regulation (Gao et al. 2009).

The development of murine HH models has enabled the investigation of the molecular

mechanism responsible for the dysregulation of iron metabolism in HH. The Hfe knockout

model (Hfe-/-) of HH Type 1 (Zhou et al. 1998) and the Tfr2 Y245X knock-in model of HH

Type 3 (Fleming et al. 2002), both have decreased hepcidin levels and increased iron

absorption (Trinder et al. 2002; Drake et al. 2007) and demonstrate how disruption of

either Hfe or Tfr2 genes can result in disturbed iron metabolism akin to that evidenced in

HH patients. Though not classified in its own HH subgroup, there have been reports of

patients with mutations in both HFE and TFR2 that have a form of HH more severe than

either Type 1 or Type 3 (Pietrangelo et al. 2005). The generation of the HFExTFR2 double

mutant model (Hfe-/-xTfr2mut)(Delima et al. 2012) will enable further investigation of the

Page 74: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

59

interactions between HFE and TFR2 and their joint roles in iron metabolism and the

pathogenesis of HH.

In this chapter Hfe-/-, Tfr2mut and Hfe-/-xTfr2mut murine models of HH were used as well as

wild-type (WT) mice fed either a control or iron supplemented diet. The aim of this study

was to characterise the phenotypic characteristics of these mice including body and organ

weight, haematological iron parameters, liver metal content as a means of investigating

possible shared transporters of iron, and liver function. Furthermore the effects of

disruption of Hfe and Tfr2 alone or in combination on the expression of duodenal and liver

iron metabolism genes and hepcidin signalling pathways will be examined.

3.2 Methods

3.2.1 Animals Animal models were generated and raised as previously described (Materials and

methods 2.2.1)

3.2.2 Tissue collection Tissues used in this study were collected as stated in Materials and methods 2.2.1.

3.2.3 Haematology Blood was collected by cardiac puncture and placed in Microtainer K2EDTA lined tubes

(BD Medical, NJ, USA). Haemoblobin, haematocrit, red blood cell count (RBC), mean

corpuscular volume (MCV), mean corpuscular haemoglobin concentration (MCHC), mean

corpuscular haemoglobin (MCH), red blood cell distribution width (RDW), platelet count

and white blood cell count assays were conducted according to standard diagnostic

methods by PathWest, Fremantle Hospital, Australia.

Page 75: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

60

3.2.4 Plasma iron measurement Blood was collected by cardiac puncture according to the method previously described

(Materials and Methods 2.2.1). Plasma iron assays were conducted according to the

methods described in Materials and Methods 2.2.3-5.

3.2.5 Liver function Blood was collected by cardiac puncture and placed in Microtainer serum separating tubes

(BD Medical, NJ, USA). Total protein, albumin, globulin, alanine transaminase and alkaline

phosphatase assays were conducted according to standard diagnostic methods by

PathWest, Fremantle Hospital, Australia.

3.2.6 Hepatic metal measurement Hepatic metal (iron, copper, zinc and manganese) content was measured by ICP-AES as

previously described (Materials and methods 2.2.2)

3.2.7 Gene expression Total RNA from liver tissue was isolated as described in Materials and methods 2.2.6.

Measurement of liver gene expression was performed by RT-PCR according to the

manufacturer’s instructions as outlined in Materials and methods 2.2.10. Real-time PCR

was conducted for liver β-Actin, Bmp6, DcytB, Dmt1, Fpn, Hamp1, Hfe, Id1, Smad7, Tfr1,

Tfr2, Transferrin, Zip14 using the appropriate forward and reverse primers (Table 2.3) on a

Rotorgene 3000 as previously described (Materials and methods 2.2.10-11).

3.2.8 p-Smad 1/5/8 expression Measurement of p-Smad 1/5/8 was performed by Western blotting. Briefly, liver

homogenates (100 µg) were electrophoresed on 4%-12% Novex Bis-Tris gradient gels

(Invitrogen) with a MOPS-SDS buffering system, prior to being transferred onto BioTrace

NT nitrocellulose membrane (Pall, NY, USA) Membranes were blocked in 5% skim milk

powder in TBST (tris-buffered saline and 0.5% Tween 20) for 30 min at room temperature.

Membranes were incubated sequentially in anti-phospho-Smad1/5/8 (1:500, Cell

Page 76: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

61

Signalling, Boston, MA,USA), anti-Smad1/5/8 (1:500, Santa Cruz Biotechnology, Santa

Cruz, CA, USA) and anti-β-actin (1:1000, Millipore, Billerica, MA, USA) in PBST, overnight

at 4ºC. Membranes were washed in TBST before incubation in anti-rabbit (Millipore) or

anti-mouse (Millipore) horseradish peroxidase for 4 h at room temperature. After

incubation, membranes were washed in 3 changes of TBST. Following a final wash in

TBST, Western Lightning Chemiluminescent reagent Plus (PerkinElmer, MA, USA) was

applied to membranes and images were captured using a VersaDoc Model 3000 (BioRad,

Hercules, CA, USA) and protein levels were quantified using Quantity One software

(BioRad).

3.2.9 Statistics Results are expressed as mean ± SEM, n=6 and were calculated as previously described

(Materials and methods 2.2.13).

3.3 Results Total body and organ weights in HH mice (Hfe

-/-, Tfr2mut, Hfe

-/-xTfr2mut) and wild-

type (WT) mice (iron-loaded and non-iron-loaded) are shown in Table 3.1. Total

body weight was approximately ten percent lower in Hfe-/-xTfr2

mut, Hfe

-/- and iron-

loaded WT mice compared with their non-iron loaded wild-type counterparts.

Organ weight is expressed relative to total body weight. Liver weight was similar in

Hfe-/-, Tfr2

mut and WT mice and significantly (p<0.05) increased in Hfe-/-xTfr2

mut

compared with Tfr2mut and WT mice. Splenic weight was unchanged in single

mutant mice and iron-loaded mice compared with non-iron-loaded WT mice.

However, splenic weight was 33% higher in Hfe-/-xTfr2

mut than single mutant and

WT mice. The kidney weight was increased in all HH mice compared with WT mice

but was not significantly different between HH mice. There was no variation

observed in heart, pancreas and brain weight between all types of mice.

Page 77: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

62

Table 3.1: Body and organ weights of HH and WT mice.

WT WT+Fe Hfe-/- Tfr2

mut Hfe-/-xTfr2mut

Body weight (g)

25.8±0.5 22.9±0.4a 23.9±0.4a 25.5±0.4 23.3±0.5a

Liver (mg/g body weight)

48.3±0.9 48.5±1.3 48.5±1.9 50.1±0.9 53.4±0.7a,b,d

Kidneys (mg/g body weight)

12.4±0.2 12.3±0.2 13.1±0.3a,b 12.8±0.2a,b 12.7±0.2a,b

Spleen (mg/g body weight)

3.1±0.1 3.1±0.1 3.1±0.2 3.2±0.1 3.7±0.2a,b,c,d

Heart (mg/g body weight)

6.0±0.5 5.6±0.2 5.9±0.1 5.6±0.1 5.8±0.1

Pancreas (mg/g body weight)

12.2±0.2 12.3±0.4 12.6±0.5 12.3±0.3 11.9±0.3

Brain (mg/g body weight)

17.7±0.3 18.7±0.3 18.7±0.3 17.7±0.2 18.9±0.6

NOTE. Results are expressed as mean ± SEM (n=5-15). ap<0.05 vs WT; bp<0.05 vs WT+Fe; cp<0.05 vs Hfe-/-; dp<0.05 vs Tfr2mut denote significance between groups.

Page 78: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

63

3.3.1 Haematology Both plasma haemoglobin and haematocrit (Hct) levels were significantly (p<0.05)

increased in Tfr2mut and Hfe-/-xTfr2mut mice compared with Hfe-/- and WT mice (Table 3.2).

Hfe-/-xTfr2mut and Tfr2mut mice also displayed approximately a 10% increase in red blood

cells (RBC) when compared with Hfe-/- and non-iron-loaded WT mice. Mean corpuscular

volume (MCV) was not significantly altered in Hfe-/-xTfr2mut, Hfe-/-, iron-loaded WT mice

compared with non-iron-loaded, but was increased in Tfr2mut mice when compared to all

other types of mice. Mean cell haemoglobin concentration (MCHC) was increased in Hfe-/-

xTfr2mut, Hfe-/-, and iron-loaded WT mice compared with non-iron-loaded mice and Tfr2mut

mice. No significant changes in mean cell haemoglobin (MCH), red blood cell distribution

width (RDW), or platelet number were observed (Table 3.2). White cell counts were

unlatered in Hfe-/-xTfr2mut, Hfe-/-, and iron-loaded WT mice compared with non-iron-loaded

mice but were elevated in Tfr2mut mice compared with all other mice (Table 3.2).

Page 79: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

64

Table 3.2: Haematological parameters in HH and wild type mice.

WT WT+Fe Hfe-/- Tfr2

mut Hfe-/-xTfr2mut

Haemoglobin (g/L)

137.0±2.5 147.4±3.0a 142.3±2.1 158.1±2.0a,b,c 158.6±2.6a,b,c

Hct 0.45±0.01 0.48±0.01 0.44±0.01 0.52±0.01a,b,c 0.50±0.01a,b,c

RBC (x1012/L)

8.47±0.18 9.33±0.24a 8.56±0.17 9.55±0.11a,c 9.58±0.17a,c

MCV (fL)

52.67±0.21 50.57±0.20a 51.30±0.54 54.45±0.37a,b,c 52.44±0.24b,d

MCHC (g/L)

303.4±4.2 317.0±3.4a 324.3±4.4a 303.6±3.3c 316.9±1.1a,d

MCH (pg)

16.32±0.40 16.10±0.10 16.61±0.19 16.54±0.10 16.58±0.08

RDW 14.32±0.31 13.81±0.21 13.75±0.23 14.24±0.14 14.10±0.23

Platelets (x109/L)

365.2±104.1 317.0±91.7 413.6±58.1 440.6±48.3 237.3±38.1

White cells (x109/L)

3.85±0.41 3.71±0.34 4.10±0.37 5.14±0.29a,b,c 3.47±0.34d

NOTE. Results are expressed as mean ± SEM (n=5-15). ap<0.05 vs WT; bp<0.05 vs WT+Fe; cp<0.05 vs Hfe-/-; dp<0.05 vs Tfr2mut denote significance between groups. RBC, red blood cell count; Hct, haematocrit; MCV, mean corpuscular volume; MCH, mean cell haemoglobin; MCHC, mean cell haemoglobin concentration; RDW, red blood cell distribution width.

Page 80: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

65

3.3.2 Plasma Iron parameters Plasma iron concentration and transferrin saturation were higher in Hfe-/-xTfr2mut, Tfr2mut,

Hfe-/- and iron-loaded WT mice compared with non-iron-loaded WT mice (p<0.05; Fig.

3.1A,B). Plasma iron concentration and transferrin saturation were highest in Hfe-/-xTfr2mut

mice (p<0.05, Fig. 3.1A,B). Plasma iron concentration in Tfr2mut mice was increased

compared to Hfe-/- mice (p<0.05). Plasma NTBI concentration was also elevated in all iron-

loaded mice (p<0.05). In Hfe-/-xTfr2mut mice, NTBI levels were 7-fold higher than non-iron-

loaded wild-type mice and more than 2-fold higher than Hfe-/-, Tfr2mut and iron-loaded WT

mice (p<0.001; Fig. 3.1C).

Page 81: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

66

Figure 3.1: Plasma iron parameters. Plasma iron concentration (A), transferrin saturation (B) and non-transferrin bound iron (NTBI) concentration (C) were measured in wild-type (WT), iron-loaded wild-type (WT+Fe), Hfe-/-, Tfr2mut and Hfe-/-xTfr2mut mice. Results are expressed as mean ± SEM (n=5-15). a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut.

Page 82: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

67

3.3.3 Liver function

Plasma total protein and globulin levels were only increased in Hfe-/-xTfr2mut and Tfr2mut

mice compared with WT mice. Tfr2mut mice had increased levels of plasma albumin when

compared to Hfe-/- and WT mice, whilst no change was observed in Hfe-/-xTfr2mut mice

(Table 3.3). Serum alanine transaminase (ALT) and alkaline phosphatase are markers of

liver injury. ALT levels were increased in all HH mice and iron-loaded WT mice. Hfe-/-

xTfr2mut ALT levels were also significantly (p<0.05) increased in compared with single

mutant HH mice and were more than 3-fold higher than WT mice (Table 3.3). Alkaline

phosphatase levels were unaltered in Hfe-/-xTfr2mut and Tfr2mut mice but were increased in

Hfe-/- and iron-loaded WT mice compared with non-iron-loaded WT mice.

Page 83: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

68

Table 3.3: Serum markers of liver function in HH and wild type mice.

WT WT+Fe Hfe-/- Tfr2

mut Hfe

-/-xTfr2mut

Total protein (g/L) 46.83±1.05 45.25±0.45 47.17±1.51 54.27±0.76a,b,c 51.70±0.54a,b,c

Albumin (g/L) 26.83±0.75 26.88±0.40 26.33±1.02 30.09±0.56a,b,c 27.09±0.48d

Globulins (g/L) 20.00±1.00 18.38±0.18 20.83±0.54b 24.18±0.63a,b,c 24.30±0.37a,b,c

Alanine transaminase (U/L)

87.80±7.27 170.75±11.99a 162.40±8.56a 142.00±7.90a 294.00±55.70a,b,c,d

Alkaline Phosphatase (U/L)

102.83±8.17 130.25±5.28a 137.40±2.34a 110.09±2.03b,c 103.91±4.02b,c

NOTE. Results are expressed as mean ± SEM (n=5-15). ap<0.05 vs WT; bp<0.05 vs WT+Fe; cp<0.05 vs Hfe-/-; dp<0.05 vs Tfr2mut denote

significance between groups.

Page 84: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

69

3.3.4 Liver metal content Hepatic iron content (HIC) was more 20-fold higher than liver zinc content which was in

turn more than 10-fold higher than liver copper and manganese content (Fig. 3.2). HIC

was higher in Hfe-/-xTfr2mut mice compared with either Hfe-/- or Tfr2mut mice (Fig. 3.2A,

p<0.01) and approximately 5-fold greater than non-iron-loaded WT mice. In Tfr2mut mice

HIC was approximately 20% higher than in Hfe-/- mice which were similar to the iron-

loaded WT and 3-fold higher than non-iron-loaded WT (Fig. 3.2A, p<0.001). Hepatic zinc

concentration was increased only in Hfe-/-xTfr2mut mice (Fig. 3.2B, p<0.01), whilst copper

concentration was increased in both Tfr2mut and Hfe-/-xTfr2mut mice compared with other

types of mice (Fig. 3.2C, p<0.05). Hepatic manganese concentration was only altered in

Hfe-/-xTfr2mut mice, where it was increased by approximately 2-fold compared with WT

mice (Fig. 3.2D, p<0.05).

Page 85: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

70

Figure 3.2: Liver metal content Liver metal content was measured by ICP-AES. Iron (A), zinc (B), copper (C), and manganese (D) concentrations were measured in WT, WT+Fe, Hfe-/-, Tfr2mut and Hfe-/-

xTfr2mut mice. Results are expressed as mean ± SEM (n=5-15). a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut denote significance between groups.

Page 86: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

71

3.3.5 Liver expression of iron regulatory genes Hfe expression was undetectable in Hfe-/- and Hfe-/-xTfr2mut mice (p<0.001) while

expression was similar in Tfr2mut and WT mice (Fig 3.3A). Tfr2 mRNA expression in Tfr2mut

and Hfe-/-xTfr2mut mice was decreased by more than 80% compared with non-iron-loaded

WT mice (p<0.001). Tfr2 mRNA expression in Hfe-/- and iron-loaded WT mice was also

lower than non-iron-loaded wild-type mice (p<0.05; Fig 3.3B). Expression of Hamp1 was

decreased to approximately 40% in Hfe-/- and Tfr2mut mice compared with non-iron-loaded

WT mice. In Hfe-/-xTfr2mut mice, Hamp1 expression was almost abolished, being further

reduced to approximately 1% or 3% of that observed in non-iron-loaded wild-type mice

(p<0.01) or Hfe-/- and Tfr2mut mice (p<0.05), respectively. Hamp1 expression, as expected,

was increased in iron-loaded wild-type mice compared with non-iron-loaded wild-type mice

(p<0.05) and HH mice (p<0.001; Fig 3.3C). Bmp6 mRNA expression was similar in Hfe-/-

xTfr2mut, Tfr2mut and Hfe-/- mice and approximately double that of the non-iron-loaded WT

mice. However, Bmp6 expression was greatest in the iron-loaded WT mice which were

almost 60% higher than the Hfe-/-xTfr2mut, Tfr2mut and Hfe-/- and were more than 2-fold

higher than in non-iron-loaded WT mice (p<0.001; Fig 3.3D). Liver expression of Id1

mRNA was similar and decreased in Hfe-/-xTfr2mut, Tfr2mut and Hfe-/- mice but was markedly

upregulated in iron-loaded WT compared with non-iron-loaded WT (p<0.001; Fig 3.3E).

Smad7 expression was 30% lower in Hfe-/-xTfr2mut than in Tfr2mut, Hfe-/- and non-iron-

loaded WT mice which were similar and 3-fold lower than in the iron-loaded WT mice

(p<0.001; Fig 3.3F).

Page 87: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

72

Figure 3.3: Liver expression of iron regulatory genes. mRNA expression was determined by real-time qPCR. Hfe (A), Tfr2 (B), Hamp1 (C), Bmp6 (D), and Id1 (D) mRNA expression were measured in WT), WT+Fe, Hfe-/-, Tfr2mut and Hfe-/-

xTfr2mut mice. Results are expressed as mean ± SEM (n=5-15). a, p<0.05 vs. WT; b, p<0.05vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut denote significance between groups.

Page 88: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

73

3.3.6 Liver expression of SMAD1/5/8 Hepatic phosphorlyation of SMAD1/5/8 protein (p-Smad1/5/8) was reduced by almost 50%

in Hfe-/-xTfr2mut mice compared with non-iron-loaded WT mice (Fig 3.4A+B). pSmad1/5/8

levels were unchanged in Hfe-/- and Tfr2mut mice compared to non-iron loaded WT mice but

was decreased by 30% compared with iron-loaded WT mice. pSmad1/5/8 levels in iron-

loaded wild-type mice were 30% higher than non-iron-loaded WT mice (Fig 4A+B). No

change was detected in total SMAD1/5/8 protein (SMAD1/5/8) in any groups.

Page 89: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

74

Figure 3.4: Hepatic p-Smad1/5/8 protein expression. Total liver protein from WT, WT+Fe, Hfe-/-, Tfr2mut and Hfe-/-xTfr2mut mice was immunoblotted with antibodies against phospho-Smad1/5/8, and total Smad1/5/8, with actin as a loading control (A). Protein levels were quantified by densitometry and phospho-Smad levels were expressed relative to actin and normalised to WT levels. Results are expressed as mean ± SEM (n=6). a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut denote significance between groups (B).

Page 90: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

75

3.3.7 Liver expression of iron transport genes Liver Tfr1 mRNA expression was reduced in all types of HH mice and iron-loaded WT

mice compared with non-loaded WT mice (p<0.05), consistent with liver iron loading.

There was no difference in the level of Tfr1 expression between Hfe-/-xTfr2mut, Hfe-/- and

Tfr2mut mice (Fig 3.5A). Similarly Dmt1 mRNA expression was decreased by almost 50% in

HH and iron-loaded WT mice (p<0.05; Fig. 3.5B). Liver mRNA expression of the

transmembrane metal transporter Zip14 was decreased in all iron-loaded mice. It was

decreased by approximately 50% in Hfe-/-, Tfr2mut and iron-loaded WT mice and was

further decreased in Hfe-/-xTfr2mut mice to approximately 30% of non-iron loaded WT levels

(Fig. 3.5C). Expression of the iron transport protein Tf was increased in HH and iron

loaded WT mice compared with non-iron loaded WT mice. Tf expression was significantly

(p<0.05) greater in HH mice than iron-loaded WT mice (Fig. 3.5D). mRNA levels of the iron

exporter Fpn were unchanged in HH and WT mice (Fig. 3.5E).

Page 91: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

76

Figure 3.5: Liver expression of iron transporter genes. mRNA expression was determined by real-time qPCR. Tfr1 (A), Dmt1 (B), Zip14A (C), Transferrin (D) and Fpn mRNA expression (E) were measured in WT, WT+Fe, Hfe-/-, Tfr2mut and Hfe-/-xTfr2mut mice. Results are expressed as mean ± SEM (n=5-15). a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut denote significance between groups.

Page 92: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

77

3.3.8 Duodenal expression of iron transport genes Duodenal Tfr1 expression was unchanged in Tfr2 mutant and Hfe-/-xTfr2mut mice but was

decreased in Hfe-/- mice when compared with WT mice (Fig. 3.6A). Duodenal Dmt1 mRNA

expression was increased by 2-fold in Tfr2mut and Hfe-/-xTfr2mut mice but was unchanged in

Hfe-/- and iron-loaded WT mice compared with non-iron loaded WT mice (Fig. 3.6B).

Expression of the duodenal iron exporter Fpn was significantly (p<0.05) increased in

Tfr2mut and Hfe-/-xTfr2mut mice compared with WT mice but was unchanged in Hfe-/- and

iron-loaded WT mice compared with non-iron loaded WT mice (Fig. 3.6C). Duodenal

expression of DcytB mRNA was increased by more than 10-fold in Tfr2mut and Hfe-/-xTfr2mut

mice and by 2-fold in Hfe-/- mice when compared with WT mice (Fig. 3.6D).

Page 93: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

78

Figure 3.6: Duodenal expression of iron transport genes. Gene expression was determined by real-time qPCR. Tfr1 (A), Dmt1 (B), Fpn (C), and DcytB (D) mRNA expression were measured in WT, WT+Fe, Hfe-/-, Tfr2mut and Hfe-/-

xTfr2mut mice. Results are expressed as mean ± SEM (n=5-15). a, p<0.05 vs. WT; b, p<0.05vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut denote significance between groups.

Page 94: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

79

3.4 Discussion In this study it was shown that Hfe-/-xTfr2mut mice develop a more severe iron loading

phenotype than their Hfe-/- or Tfr2mut single mutant counterparts. Hfe-/-xTfr2mut mice were

found to have decreased body weight and increased liver, kidney and spleen weights

when compared with non-iron-loaded WT mice. Haematological parameters (RBC, Hb,

and Hct), iron status (plasma iron, transferrin saturation and NTBI concentration) and liver

metal content (iron, zinc, copper and manganese) were all increased in Hfe-/-xTfr2mut mice.

Hfe-/-xTfr2mut mice had increased Bmp6 levels consistent with increased HIC but

decreased Hfe and Tfr2 expression lead to ineffective p-Smad signaling and reduced

Hamp1 expression.

The observation that the murine models of HH; Hfe-/-xTfr2mut and Tfr2mut had increased

numbers of RBCs with elevated haemoglobin and haematocrit levels consistent with

several other studies in HH patients (Barton et al. 2000; McLaren et al. 2007) and mice

(Ramos et al. 2011) that have observed haematological changes. It is thought that HH

increases erythropoiesis in two ways; firstly, the down regulation of hepcidin in HH results

in increased iron absorption by the intestine and increased recycling of iron from the

macrophage increasing the bioavailability of iron for erythropoiesis (Zhang et al. 2011).

Secondly, TFR2, is abundantly expressed in the early erythron (Sposi et al. 2000) and it

has been shown that the binding of TFR2 to the erythropoietin receptor (EpoR) is required

for efficient erythropoiesis (Forejtnikova et al. 2010). Other studies have shown that

erythroid progenitor cells from Tfr2 deficient mice are less responsive to erythropoietin

(Epo), resulting in increased serum Epo levels further stimulating erythropoiesis. The

increase in erythropoiesis in Hfe-/-xTfr2mut mice is further supported by the increase in

spleen weight (Table 3.1), as splenomegaly is often associated with increased workload by

the spleen in response to proliferation of the bone marrow (Pozo et al. 2009).

Page 95: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

80

Plasma iron concentration and transferrin saturation were increased in Hfe-/- and Tfr2mut

mice in congruence with the characterisation studies of these mice by Fleming et al (Zhou

et al. 1998; Fleming et al. 2002) and were further increased in Hfe-/-xTfr2mut mice (Wallace

et al. 2009; Corradini et al. 2011). HH mice also had increased NTBI levels providing

further evidence that HH is a disease of dysregulated iron absorption, where absorbed iron

exceeds the iron binding capacity of transferrin and the excess iron in present in the serum

as NTBI.

Increased serum total protein, globulin and ALT levels observed in Hfe-/-xTfr2mut mice

suggest that Hfe-/-xTfr2mut mice have liver injury. This is further supported by the

observation of hepatomegaly in the Hfe-/-xTfr2mut mice, which is a common occurrence in

the advanced liver disease caused by HH (Brissot et al. 2000). The effect of excess

hepatic iron deposition on liver injury in HH mice will be described in more detail in

Chapter 5.

In this study it has been shown that Hfe-/-xTfr2mut mice develop a more severe hepatic iron

loading than Hfe-/- and Tfr2mut mice (Fig. 3.1). This observation is in agreement with reports

of a severe form of HH with an early onset akin to that seen in juvenile HH, in probands

with mutations in both HFE and TFR2 (Pietrangelo et al. 2005; Rueda Adel et al. 2011).

Other studies in mice with disruptions in both HFE and TFR2 (Wallace et al. 2009;

Corradini et al. 2011) have shown increases in hepatic iron concentration that are similar

to that observed in the current study. The increase in liver zinc, copper and manganese

content in Hfe-/-xTfr2mut mice suggests that there is a shared transporter for these divalent

metals and iron which is upregulated in the absence of HFE and TFR2, possible

transporters include DMT1 and ZIP14 (Hansen et al. 2009). Further evidence of a shared

transporter for the metals iron, zinc, copper and managense is the study by Chua et al

(Chua et al. 2004) who found that the divalent metals manganese, zinc, and copper

Page 96: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

81

inhibited ferric citrate uptake by hepatocytes from both Hfe-/- and wild-type mice. Inhibition

of NTBI uptake has also been demonstrated by zinc and manganese in the perfused rat

liver (Wright et al. 1986), and by zinc in both isolated rat hepatocytes (Baker et al. 1998),

and hepatoma cells (Randell et al. 1994). Increased levels of zinc, copper and manganese

in Hfe-/-xTfr2mut mice may play a role in oxidative stress as copper, zinc and manganese

are important components of the free radical scavenger, superoxide dismutase (McCord

and Fridovich 1988).

The reduction in liver hepcidin (Hamp1) expression in the presence of disruption of either

Hfe or Tfr2 in the liver emphasises the role that HFE and TFR2 play in the regulation of

hepcidin expression and iron homeostasis. Furthermore, the additive effect on iron loading

by the near abrogation of hepcidin in response to the disruption of HFE and TFR2,

suggests they may work in parallel or possibly converging signalling pathways. Other

studies have demonstrated that the iron-dependent regulation of hepcidin is controlled by

HFE and TFR2, and BMP6/SMAD cell signalling pathways (Kautz et al. 2008; Wallace et

al. 2009). It has been shown that HFE can interact with TFR1 and TFR2 to form a complex

that is hypothesised to sense plasma transferrin saturation and modulate hepcidin

synthesis accordingly (Olynyk et al. 2008). However, the exact nature of this mechanism is

yet to be fully elucidated. These findings support previous studies that suggest there may

be cross-talk between HFE/TFR2 and BMP6/SMAD signalling pathways, as disruption of

Hfe in combination with Tfr2 results in decreased levels of pSMAD1/5/8, which leads to a

decrease in hepcidin synthesis.

Duodenal expression of the iron transporters Dmt1 and Fpn was unchanged in Hfe-/- mice

but was significantly (p<0.05) increased in Tfr2mut and Hfe-/-xTfr2mut mice. This result is

consistent with other studies that show Dmt1 and Fpn are unchanged in Hfe-/- mice

(Herrmann et al. 2004) but are both increased in Tfr2mut mice (Drake et al. 2007). Reports

Page 97: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

82

of increased levels of duodenal Dmt1 and Fpn mRNA in Hfe-/- (Dupic et al. 2002) and

Tfr2mut mice (Kawabata et al. 2005) are most likely due to genetic differences in the

background stains of the mutant mice. Increased iron absorption has been demonstrated

in Hfe-/- mice (Ajioka et al. 2002) and Tfr2mut mice (Drake et al. 2007) and it is likely to be

similar in Hfe-/-xTfr2mut mice. The enterocytes of the duodenum have been shown to be

relatively iron deficient in HH (Francanzani et al. 1989) due to the excessive export of iron

to the circulation. The increase in Dmt1 expression in the Tfr2mut and Hfe-/-xTfr2mut mice is

likely to be due to an increase in DMT1 transcripts containing an IRE that is increased in

iron deficiency (Hubert and Hentze 2002). This is supported by reports of high duodenal

Dmt1 expression in iron deficient mice (Gunshin et al. 1997). The reason for an increase in

Fpn in Tfr2mut and Hfe-/-xTfr2mut mice is unclear but as mentioned previously, similar

findings have been reported (Drake et al. 2007; Wallace et al. 2009).

Hepatic TfR1 expression was decreased in all HH mice (Fig. 3.3A). This finding was

expected and is supported by previous studies in Hfe-/- (Chua et al. 2008), Tfr2mut (Chua et

al. 2010) and iron-loaded wild-type mice (Chua et al. 2008; Chua et al. 2010). TfR1

expression is inversely regulated by cellular iron levels by the IRE/IRP post-transcriptional

regulation as described in Chapter 1 (section 1.8.3), wherein Tfr1 mRNA undergoes

enhanced degradation under conditions of high HIC found in HH mice (Rouault 2006). The

fact that liver Fpn mRNA levels were unchanged by high HIC in the current study (Fig.

3.3B), is consistent with the findings of previous studies (Chua et al. 2008; Chua et al.

2010); (Constante et al. 2006) and support the theory that FPN regulation occurs by a

post-translational mechanism, with hepcidin binding to FPN protein and inducing its

degradation (Nemeth et al. 2004).

The increased expression of liver transferrin in Hfe-/-xTfr2mut, Hfe-/- and Tfr2mut mice is an

unexpected finding as the hepatic synthesis of transferrin has been shown to be positively

Page 98: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

83

regulated by iron deficiency, not iron overload. However, proximal region II, adjacent to the

transferrin promoter of transferrin contains a CCAAT sequence capable of binding CCAAT

enhancer-binding proteins (C/EBP) (Theisen et al. 1993). One particular form of C/EBP,

known as C/EBPα has been shown to be positively regulated by increased iron levels

(Harrison-Findik et al. 2007) and may possibly contribute to the iron dependent regulation

of transferrin seen in the HH mice.

Hepatic expression of the zinc and iron transporter Zip14 was decreased in Hfe-/- and

Tfr2mut mice and to a greater extent in Hfe-/-xTfr2mut mice. The decrease in Zip14A

expression with increasing HIC may be due to in the increased hepatic zinc content

evident in Hfe-/-xTfr2mut mice. Zip14 is a member of slc39 transporter family, other

members of the family such as Zip10 are known to possess metal-response elements

which in response to zinc-induced metal-regulatory transcription factors result in the down-

regulation of the target mRNA (Zheng et al. 2008). Zip14 may also be regulated in a

manner similar to Zip4, where zinc excess results in reduced mRNA stability leading to

protein degradation (Mao et al. 2007). It has also been shown that in the absence of HFE,

the stability of ZIP14 protein is increased (Gao et al. 2008) allowing for ZIP14 to contribute

to liver iron and zinc transport in HH despite a decrease in mRNA expression.

The disruption of HFE and TFR2 results in an inability to sense plasma iron levels and

impairment of Bmp/Smad signaling resulting in reduced hepcidin synthesis. With the loss

of hepcidin synthesis, duodenal iron absorption continues unabated and the excess iron

that is responsible for the alterations in phenotype that were evident particularly in Tfr2mut

and Hfe-/-xTfr2mut mice. Increased liver iron and other metal levels in Hfe-/-xTfr2mut mice

result in hepatomegaly with increased erythropoiesis leading to elevations in RBCs and Hb

levels and ultimately splenomegaly (Spivak 2000). The results of this study suggest that

Page 99: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

84

disruption of HFE and TFR2 whilst primarily causing liver iron overload also induced other

phenotypic changes that should be considered in human HH.

Page 100: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

85

Chapter 4

Non-transferrin-bound iron transport in hereditary

haemochromatosis

Page 101: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

86

The following chapter "In vivo liver non-transferrin-bound iron uptake is upregulated in

murine models of Hereditary Hemochromatosis" has been submitted to American Journal

of Physiology - Gastrointestinal and Liver Physiology on 4th Apr 2013 (GI-00113-2013)

Page 102: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

87

4.1 Introduction

Under normal conditions, the plasma iron transport protein, transferrin, is 20-35%

saturated with iron (Morgan 1996). However, in conditions where transferrin approaches

saturation, such as hereditary haemochromatosis and in transfusion-dependent

thalassaemias, excess iron in the body is unable to bind to transferrin and exists in the

circulation as non-transferrin-bound iron (NTBI). NTBI can produce reactive oxygen

species (ROS) in tissues and bodily fluids through catalysis of the Fenton reaction

(Aruoma et al. 1988).

Since the early studies by Hersko et al. (Hershko et al. 1978), NTBI has been found to be

present in several other iron-overload conditions such as sickle cell anaemia (Koren et al.

2010) and bone marrow failure during cancer treatments (Sahlstedt et al. 2009). Though

the presence of NTBI usually correlates with high transferrin saturation, it has also been

found in patients with only partially saturated transferrin (Gutteridge et al. 1985). Increased

NTBI levels have been implicated in abnormal iron deposition in the liver, heart and

endocrine glands (Oudit et al. 2003) and are thought to be a major cause of organ

dysfunction in iron-overload disorders.

Though the presence of NTBI has been known for many years, there is still a poor

understanding of its chemical composition in various iron-overload disorders. NTBI in the

plasma may exist as iron bound to various non-protein ligands such as citrate, acetate,

pyruvates and phosphates (Silva and Hider 2009), though a precise understanding of the

chemical composition of NTBI has yet to be elucidated. However, studies using serum

from haemochromatosis patients, implicates iron-citrate as the predominant form of

circulating NTBI in haemochromatosis (Grootveld et al. 1989).

Page 103: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

88

The uptake and distribution of NTBI in hereditary haemochromatosis is still poorly

understood. In this condition, excess circulating iron in the form of transferrin-bound iron

(TBI) and NTBI deposits primarily in the parenchymal cells of the liver. Previous studies

have demonstrated that NTBI uptake is increased in isolated hepatocytes from Hfe

knockout mice (Chua et al. 2004) and decreased in the presence of HFE in Chinese

Hamster Ovary cells (Carlson et al. 2005) and HepG2 hepatoma cells (Gao et al. 2008).

Furthermore, it has recently been demonstrated that there is a link between plasma NTBI

levels and the degree of liver injury in mouse models of HH (Delima et al. 2012). Thus the

relationship between plasma NTBI levels, hepatic NTBI uptake and the pathogenesis of

liver injury in HH is of importance.

In the current study Hfe knockout (Hfe-/-)(Zhou et al. 1998), Tfr2 mutant (Tfr2mut)(Fleming et

al. 2002) and double mutant (Hfe-/-xTfr2mut)(Delima et al. 2012) mouse models of HH were

used to investigate the transport of NTBI in vivo.

4.2 Methods

4.2.1 Animals Animal models were generated and raised as previously described (Materials and

methods 2.2.1)

4.2.2 NTBI uptake NTBI uptake was measured in vivo using a modified method described previously by

Craven et al. (Craven et al. 1987). Based on pre-experiment measurement of plasma total

iron binding capacity and transferrin saturation (Fig 4.1A), plasma transferrin was

saturated with non-radioactive ferric citrate (4-15 nmoles; 1:5 ratio of iron to citrate in 50 µL

Page 104: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

89

isotonic saline) in Hfe-/-, Tfr2mut and WT mice via lateral tail vein injection. Hfe-/-xTfr2mut

double mutant mice received an equivolume injection of isotonic saline as plasma

transferrin was already saturated with iron. After 15 min, the mice were injected with NTBI

in the form of 59Fe-citrate (0.6 nmole; 1:5 ratio of iron to citrate in 30 µL isotonic saline) via

lateral tail vein and blood samples (50 µL) were collected at various time points. After 30

min, blood was collected by cardiac puncture, and mice were perfused with 15-20 mL ice-

cold 0.15 M saline in situ after which the liver, kidneys, pancreas, heart, duodenum and

femurs were collected and counted for radioactivity using a Wizard gamma counter

(PerkinElmer, Massachusetts USA). Specific activity of 59Fe-NTBI was corrected to

account for the different pool sizes of circulating NTBI in HH and WT mice using the post-

experiment plasma NTBI concentration (Fig. 4.1C). The amount of 59Fe-NTBI in the

plasma samples taken at various time intervals after 59Fe-citrate injection was expressed

as a percentage of the zero time value to determine the plasma NTBI clearance. Tissue

NTBI uptake was calculated using tissue 59Fe content and corrected specific activity and

expressed as pmoles NTBI per total organ or g wet weight.

4.2.3 Tissue collection Tissues used in this study were collected as stated in Materials and methods 2.2.1.

4.2.4 Plasma iron measurement Blood was collected by cardiac puncture according to the method previously described

(Materials and Methods 2.2.1). Plasma iron assays were conducted according to the

methods described in Materials and Methods 2.2.3-5.

4.2.5 Hepatic iron content Hepatic iron content was measured by ICP-AES as previously described (Materials and

methods 2.2.2)

Page 105: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

90

4.2.6 Statistics Results are expressed as mean ± SEM, n=6 and were calculated as previously described

(Materials and methods 2.2.13).

4.3 Results

4.3.1 Tissue iron content Liver iron concentration was elevated in all HH and dietary iron-loaded mice compared

with non-iron-loaded WT mice. Liver iron levels in Hfe-/- and iron-loaded WT mice were

similar and more than 3-fold higher than non-iron-loaded WT mice (p<0.001; Table 4.1).

Tfr2mut mice exhibited significantly (p<0.01) higher HIC than Hfe-/- mice. Furthermore, iron

concentration in the liver of Hfe-/-xTfr2mut mice was approximately 1.5-fold greater than Hfe-

/- or Tfr2mut mice (Table 4.1), and approximately 5-fold higher than non-iron-loaded WT

mice, consistent with previously reported results using a colorimetric assay (Delima et al.

2012). Iron concentration in the kidneys was similar in Hfe-/-, Tfr2mut and non-iron loaded

WT mice but was less than iron-loaded WT mice (p<0.05; Table 4.1). In Hfe-/-xTfr2mut mice,

kidney iron concentration was increased compared with all other types of mice (p<0.05;

Table 4.1). Pancreatic iron concentration was increased in Hfe-/- and iron-loaded WT mice

(p<0.01), whereas iron concentration in the heart was not increased in these mice

compared with non-iron-loaded WT mice (Table 4.1). In Tfr2mut mice, pancreatic and

cardiac iron levels were more than 4-fold higher than Hfe-/- and WT mice (p<0.01; Table

4.1). Most notably, Hfe-/-xTfr2mut mice had pancreatic and cardiac iron levels approximately

3-fold higher than Tfr2mut mice and more than 10 to 20-fold higher than other types of mice

(p<0.01; Table 4.1).

Page 106: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

91

Table 4.1: Tissue iron content.

Tissue WT WT+Fe Hfe-/- Tfr2

mut Hfe-/-xTfr2

mut

Liver 2.77 ± 0.04 10.41 ± 0.29a 9.45 ± 0.29

a 11.25 ± 0.57

a,c 14.39 ± 1.00

a,b,c,d

Kidney 1.29 ± 0.05 1.77 ± 0.07a 1.00 ± 0.07

b 1.21 ± 0.02

b 2.79 ± 0.23

a,b,c,d

Pancreas 0.50 ± 0.02 0.80 ± 0.09a 0.88 ± 0.09

a 3.52 ± 0.29

a,b,c 10.02 ± 0.73

a,b,c,d

Heart 0.43 ± 0.04 0.32 ± 0.04 0.43 ± 0.04 1.91 ± 0.09a,b,c

4.70 ± 0.32a,b,c,d

NOTE. Results are expressed as mean ± SEM µmol Fe/g wet weight tissue (n=4-12). a, p<0.05 versus WT; b, p<0.05 versus WT+Fe; c, p<0.05 versus Hfe-/-; d, p<0.05 versus Tfr2mut denote significance between groups.

4.3.2 Plasma iron parameters Plasma TS and NTBI concentrations were increased in all HH and iron-loaded WT mice

compared with non-iron-loaded WT mice (non-hatched bars, p<0.05; Fig. 4.1A, B). The

increases in plasma TS and NTBI levels were greatest in Hfe-/-xTfr2mut mice (non-hatched

bars, p<0.05, Fig. 4.1A, B). NTBI concentration was approximately 4-fold higher than in

either Hfe-/- or Tfr2mut mice (p<0.05, Fig. 4.1B). There was a strong positive correlation

between plasma TS and NTBI concentration (r=0.91, p=0.03; Fig. 4.1C). At the conclusion

of the experiment, TS was approximately 100% in all groups of mice (Fig. 4.1A; hatched

bars). There were also no changes in plasma NTBI levels post-experiment in HH and iron-

loaded WT mice except in non-iron-loaded WT mice (Fig. 4.1B; non-hatched bars versus

hatched bars).

Page 107: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

92

Figure 4.1: Plasma iron parameters. Transferrin saturation (A) and non-transferrin bound iron (NTBI) concentration (B) were measured pre- (non-hatched bars) and post-NTBI uptake experiment (hatched bars) in wild-type (WT), iron-loaded wild-type (WT+Fe), Hfe knockout (Hfe-/-), Tfr2 mutant (Tfr2mut) and Hfe-/-xTfr2mut mice. Correlation between plasma transferrin saturation and plasma NTBI concentration (C). Results are expressed as mean ± SEM (n=5-11). a, p<0.05 versus WT; b, p<0.05 versus WT+Fe; c, p<0.05 versus Hfe-/-; d, p<0.05 versus Tfr2mut denote significance between groups. *, p<0.05 versus pre-experiment (non-hatched bars).

r=0.91 p=0.03

Page 108: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

93

4.3.3 Plasma NTBI clearance NTBI was rapidly cleared from the plasma in Hfe-/-, Tfr2mut and Hfe-/-xTfr2mut, with the

majority of 59Fe-NTBI cleared from the plasma by 2 mins (Fig. 4.2). The clearance on NTBI

was significantly (p<0.001) increased in HH and iron-loaded mice compared with non-iron-

loaded WT mice (Fig. 4.2). Clearance on NTBI from the plasma was most efficient in Hfe-/-

xTfr2mut and Tfr2mut mice was up to 5-fold greater than in non-iron-loaded WT mice at 30

mins. NTBI clearance was approximately 2.5-fold higher in Hfe-/- and iron-loaded WT mice

than in non-iron-loaded WT mice at 30 mins (Fig. 4.2).

Figure 4.2: Plasma NTBI clearance Plasma NTBI clearance was measured at 2 and 30 min in wild-type (WT), iron-loaded wild-type (WT+Fe), Hfe knockout (Hfe-/-), Tfr2 mutant (Tfr2mut) and Hfe-/-xTfr2mut mice. Results are expressed as mean ± SEM (n=5-8). Clearance at time point: a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut. *, p<0.05 30 min vs 2 min.

Page 109: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

94

4.3.4 Tissue NTBI uptake In WT and Hfe-/-xTfr2mut mice, NTBI was taken up predominately by the liver with less

taken up by the kidneys, pancreas, heart and duodenum (p<0.05; Fig. 4.3A), with a similar

pattern of distribution in Hfe-/- and Tfr2mut mice (p<0.05; Fig. 4.3B).

Figure 4.3: Tissue NTBI uptake. Tissue distribution of NTBI uptake was measured in WT mice (dark green bars) and Hfe-/-

xTfr2mut mice (red bars) (A), and Hfe-/- (orange bars) and Tfr2mut mice (light green bars) (B). Results are expressed as mean ± SEM (n=5-11). a, p<0.05 versus liver; b, p<0.05 versus kidney; c, p<0.05 versus pancreas; d, p<0.05 versus heart denote significance between groups. *, p<0.05 versus WT (green bars) or Hfe-/- (orange bars).

Page 110: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

95

Liver NTBI uptake was elevated in all iron-loaded mice compared with non-iron-loaded WT

mice (p<0.001). In Hfe-/- mice, liver NTBI uptake was similar to iron-loaded WT mice and

more than 2-fold higher than non-iron-loaded WT mice (p<0.001; Fig. 4.4A). NTBI uptake

was increased by 30% in Tfr2mut mice and by more than 5-fold in Hfe-/-xTfr2mut mice

compared with Hfe-/- mice (p<0.05; Fig. 4.4A). There was a significant positive correlation

between liver NTBI uptake and both liver iron concentration (r=0.75, p<0.0001; Fig. 4.4B)

and plasma NTBI concentration post-experiment (r=0.92, p<0.0001; Fig. 4.4C).

Page 111: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

96

Figure 4.4: Liver NTBI uptake. Liver NTBI uptake (A) was measured in WT, WT+Fe, Hfe-/-, Tfr2mut and Hfe-/-

xTfr2mut mice and plotted against liver iron concentration (B), and plasma NTBI concentration (C). Results are expressed as mean ± SEM (n=5-11). a, p<0.05 versus WT; b, p<0.05 versus WT+Fe; c, p<0.05 versus Hfe-/-; d, p<0.05 versus Tfr2mut denote significance between groups.

r=0.75 p<0.0001

r=0.92 p<0.0001

Page 112: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

97

NTBI uptake by the kidneys and pancreas was increased in all iron-loaded mice compared

with non-iron-loaded WT mice (Fig. 4.5A and 4.6A; p<0.05). In both kidney and pancreas,

NTBI uptake was similar in Hfe-/- and iron-loaded WT mice, and was increased by 1.5- fold

in Tfr2mut and by 4-5-fold in Hfe-/-xTfr2mut mice compared with Hfe-/- mice (Fig. 4.5A and

4.6A; p<0.05). Cardiac NTBI uptake was similar in Hfe-/- and WT mice, and was

significantly (p<0.01) increased by approximately 70% in Tfr2mut mice, and 4-fold in Hfe-/-

xTfr2mut mice compared with non-iron-loaded mice (Fig. 4.7A). Duodenal NTBI uptake was

similar in Hfe-/-, Tfr2mut and WT mice and increased by more than 3-fold in Hfe-/-xTfr2mut

mice compared with all other groups (Fig. 4.8A; p<0.001). NTBI uptake by the femurs was

considerably lower than NTBI uptake by the liver, kidney, pancreas and heart. Femur NTBI

uptake in Hfe-/- and Tfr2mut mice was reduced by 50%, and increased in Hfe-/-xTfr2mut by 4-

fold compared with non-iron-loaded WT mice (Fig. 4.8B). NTBI uptake was strongly

correlated with plasma NTBI and tissue iron content in the kidneys (r=0.84, p<0.0001

versus iron content, Fig. 4.5B; r=0.86, p<0.0001 versus plasma NTBI, Fig. 4.5C;),

pancreas (r=0.98, p<0.0001 versus iron content, Fig. 4.6B; r=0.89, p<0.0001 versus

plasma NTBI, Fig. 4.6C) and heart (r=0.89, p<0.0001 versus iron content, Fig. 4.7B;

r=0.95, p<0.0001 versus plasma NTBI, Fig. 4.7C) in all groups of mice.

Page 113: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

98

Figure 4.5: Kidney NTBI uptake. Kidney NTBI uptake (A) was measured in WT, WT+Fe, Hfe-/-, Tfr2mut and Hfe-/-

xTfr2mut mice and plotted against liver iron concentration (B), and plasma NTBI concentration (C). Results are expressed as mean ± SEM (n=5-11). a, p<0.05 versus WT; b, p<0.05 versus WT+Fe; c, p<0.05 versus Hfe-/-; d, p<0.05 versus Tfr2mut denote significance between groups.

r=0.84 p<0.0001

r=0.86 p<0.0001

Page 114: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

99

Figure 4.6: Pancreas NTBI uptake. Pancreas NTBI uptake (A) was measured in WT, WT+Fe, Hfe-/-, Tfr2mut and Hfe-/-

xTfr2mut mice and plotted against liver iron concentration (B), and plasma NTBI concentration (C). Results are expressed as mean ± SEM (n=5-11). a, p<0.05 versus WT; b, p<0.05 versus WT+Fe; c, p<0.05 versus Hfe-/-; d, p<0.05 versus Tfr2mut denote significance between groups.

r=0.98 p<0.0001

r=0.89 p<0.0001

Page 115: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

100

Figure 4.7: Heart NTBI uptake. Heart NTBI uptake (A) was measured in WT, WT+Fe, Hfe-/-, Tfr2mut and Hfe-/-

xTfr2mut mice and plotted against liver iron concentration (B), and plasma NTBI concentration (C). Results are expressed as mean ± SEM (n=5-11). a, p<0.05 versus WT; b, p<0.05 versus WT+Fe; c, p<0.05 versus Hfe-/-; d, p<0.05 versus Tfr2mut denote significance between groups.

r=0.89 p<0.0001

r=0.95 p<0.0001

Page 116: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

101

Figure 4.8: Duodenum and femur NTBI uptake Duodenum NTBI uptake (A) and Femur NTBI uptake (B) were measured in WT, WT+Fe, Hfe-/-, Tfr2mut and Hfe-/-xTfr2mut mice. Results are expressed as mean ± SEM (n=5-11). a, p<0.05 versus WT; b, p<0.05 versus WT+Fe; c, p<0.05 versus Hfe-/-; d, p<0.05 versus Tfr2mut denote significance between groups.

Page 117: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

102

4.4 Discussion In this study, NTBI transport and tissue uptake in mouse models of HH were examined.

NTBI was cleared rapidly from the circulation in all mouse models of HH, with most of the

NTBI taken up by the liver and to a lesser degree by the kidneys, pancreas and heart. The

current study describes for the first time increased in vivo tissue NTBI uptake in mouse

models of HH with the greatest increase observed in Hfe-/-xTfr2mut mice followed by Tfr2mut

and Hfe-/- mice (Hfe-/-xTfr2mut >> Tfr2mut > Hfe-/- mice) compared with non-iron-loaded wild-

type mice. There was a significant positive correlation between NTBI uptake and both

plasma NTBI levels and iron content in the liver, kidney, pancreas and heart. This

suggests that NTBI uptake is likely to contribute to excessive iron deposition primarily in

the liver as well as in the kidney, pancreas and heart in HH.

In vivo transport of NTBI was determined in murine HH models in the presence of

circulating transferrin. The observation that the iron-binding capacity of transferrin was

saturated post-experiment demonstrated that the injection of known quantities of non-

radioactive ferric citrate saturated circulating plasma transferrin. The absence of significant

changes in plasma NTBI levels pre- and post-experiment in HH mice suggested that the

injection of non-radioactive ferric citrate prior to the experiment was sufficient to saturate

the transferrin without donating additional NTBI to the circulation. As iron citrate is the

predominant form of circulating NTBI in HH (Grootveld et al. 1989), citrate was chosen as

the ligand to examine in vivo NTBI transport in this study. The clearance of plasma 59Fe-

NTBI was rapid in HH mice and occurred significantly faster than in WT mice. This is much

shorter than the plasma half-life for TBI of approximately 50-60 mins that has been

reported previously in both HH (Hfe-/- and Tfr2mut) and WT mice (Chua et al. ; Frazer 2002).

The rapid clearance of plasma NTBI compared with TBI is likely to reflect the relatively

lower concentration of plasma NTBI and the higher capacity of tissue NTBI transporters

compared with transferrin receptors that mediate TBI uptake. Most of the 59Fe-NTBI was

Page 118: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

103

primarily taken up by the liver, followed by the kidneys, pancreas and heart consistent with

the tissue distribution of NTBI uptake observed previously in vivo in hypotransferrinemic

mice (Craven et al. 1987) and Dmt1flox mice (Wang and Knutson 2013). A greater

proportion of 59Fe was deposited in the livers of HH mice compared with non-iron-loaded

wild-type mice. 59Fe uptake by the femur was very low suggesting that 59Fe was present in

the circulation in the form of NTBI and not TBI, which is cleared predominately by the bone

marrow for erythropoiesis.

HH mice exhibited increased plasma NTBI levels and NTBI clearance, liver NTBI uptake

and liver iron content. These features were most marked in Hfe-/-xTfr2mut mice compared

with either Hfe-/- or Tfr2mut mice. Liver NTBI uptake was strongly associated with both liver

iron content and plasma NTBI levels. However, the marked elevation of these parameters

in the Hfe-/-xTfr2mut mice may influence the observed statistical correlation between plasma

NTBI levels, tissue iron content and NTBI uptake. It have previously demonstrated that

liver TBI uptake in vivo was also increased in the presence of elevated plasma TBI levels

in both Hfe-/- (Frazer 2002) and Tfr2mut mice (Chua et al. 2010). Liver expression of

transferrin receptor 1 (TFR1) is downregulated in HH in response to iron-loading (Chua et

al. 2010) and although TFR2 expression is upregulated in HH in the presence of increased

levels of diferric transferrin (Robb and Wessling-Resnick 2004), previous studies have

shown that TFR2 accounts for only approximately 20% of TBI uptake in vivo (Chua et al.

2010). Hence TBI uptake is likely to play a lesser role than NTBI uptake in liver iron-

loading in HH.

The efficient clearance of NTBI by the liver is well documented, with studies in mice

indicating that 58 to 75% of NTBI is cleared by the liver on its first pass from the portal

circulation and stored in the liver as ferritin (Brissot et al. 1985). Studies by Craven et al. in

transferrin-iron saturated rats (Craven et al. 1987) are consistent with the findings of the

Page 119: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

104

current study where NTBI delivered intravenously was cleared rapidly from the circulation

and the majority was found deposited in the liver, demonstrating efficient clearance of

plasma NTBI by the liver. The ability for hepatocytes to take up NTBI has also been

demonstrated in vitro in isolated hepatocytes from rats (Baker et al. 1998) and mice, with

an upregulation of NTBI uptake by hepatocytes in Hfe-/- mice (Chua et al. 2004). These

studies demonstrated that hepatocytes take up NTBI via a low-affinity membrane

transporter that is saturable at elevated non-physiological iron concentrations, such as

those found in HH (Barisani et al. 1995). The current study, suggests that liver NTBI

transport in vivo may be positively regulated by iron in HH, consistent with in vitro studies

in fibroblast (Craven et al. 1987) and hepatoma (Randell et al. 1994) cell lines that also

demonstrated increased NTBI uptake with iron-loading. As in HH, NTBI also plays a role in

beta-thalassemia, alcoholism, haematological malignancies, diabetes mellitus (De Feo et

al. 2001; Pootrakul et al. 2004; Lee et al. 2006; Sahlstedt et al. 2009) and end stage renal

disease (Prakash et al. 2005) where excess NTBI is deposited in the liver. Excessive iron

deposition leads to the production of ROS which are linked to liver injury through the

promotion of lipid peroxidation and oxidative damage to DNA and proteins and the

production of factors that promote inflammation and fibrosis in human HH (Olynyk et al.

2008; Ramm and Ruddell 2010).

It is well established that divalent metal transporter 1 (DMT1) is integral in the uptake of

dietary NTBI in the duodenum (Gunshin et al. 1997). In the liver, several NTBI transporters

have been identified, with uptake of NTBI in hepatocytes and hepatoma cells mediated by

DMT1(Shindo et al. 2006) and Zrt-Irt-like protein 14 (ZIP14) (Liuzzi et al. 2006). The

presence of HFE has been shown to decrease ZIP14 levels by reducing protein stability

resulting in decreased NTBI uptake (Gao et al. 2008). The current observation in HFE

deficient mice (Hfe-/- and Hfe-/-xTfr2mut) of increased liver NTBI uptake in vivo are

consistent with the previous in vitro data and a negative regulatory role for HFE in NTBI

Page 120: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

105

uptake. Dmt1 protein expression has been reported to be increased (Trinder et al. 2002) or

decreased (Nam et al. 2013), whilst Zip14 protein expression was increased (Nam et al.

2013) in the liver of mice with iron loading. Specific deletion of Dmt1 in the liver however,

did not alter liver NTBI uptake or liver iron levels suggesting that DMT1 is not essential for

hepatic NTBI uptake (Wang and Knutson 2013). Global deletion of Zip14 in mice has been

reported to have unchanged (Hojyo et al. 2011) or increased liver iron levels (Beker

Aydemir et al. 2012) however liver NTBI uptake was not measured in these studies.

Typically the kidney is unaffected in HH (Pietrangelo 2004), however there are reports of

kidney iron overload in cases of severe and lethal idiopathic neonatal haemochromatosis

(Herrmann et al. 2004), The Hfe-/-xTfr2mut mouse model represents a severe form of HH

(Chua et al. 2010) which exhibits higher NTBI uptake and greater deposition of iron in the

kidney than WT mice. Under normal physiological conditions, the kidney acquires iron by

receptor-mediated endocytosis of TBI (Zhang et al. 2007). However, in HH as transferrin

becomes saturated, free iron will bind to low-molecular weight ligands which may be

filtered by the glomerulus in the kidney. The iron transporter DMT1 is highly expressed in

the renal medulla and at the brush border and apical pole endothelial cells in the proximal

tubule (Abboud and Haile 2000). However, it has been shown that the kidney is comprised

predominantly of DMT1 that contains an iron regulatory element (Canonne-Hergaux and

Gros 2002), which is likely to result in the downregulation of DMT1 protein in the presence

of high renal iron levels. Therefore, it is unlikely that DMT1 plays a major role in renal iron-

loading in HH and is more likely a mechanism for iron scavenging from the urine in

conditions of iron deficiency. Lipocalin 2 or m24p3 is a component of the innate immune

system that binds and sequesters bacterial iron compounds in the blood and urine and is

heavily expressed in the kidney and epithelial tissues (Schmidt-Ott et al. 2006). Though its

role in times of infection is well characterised (Yang et al. 2002), it may also play a role in

the transport of NTBI during normal iron homeostasis and possibly in HH (Nairz et al.

Page 121: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

106

2009).

As in chronically transfused thalassemia major patients (Noetzli et al. 2009), HH patients

with severe iron overload may develop iron-related dysfunction of the heart and pancreas

(Camaschella and Poggiali 2009). Similar results are found in the hypotransferrinemic

mouse where NTBI is the predominant form of circulating iron (Craven et al. 1987). NTBI

uptake by the pancreas and heart is thought to share a similar mechanism. ZIP14 is

expressed in the heart and pancreas (Taylor et al. 2005) and may play a role in pancreatic

and cardiac NTBI uptake. Furthermore, L-type voltage-dependent calcium channels are

highly expressed in the heart and have been shown to facilitate NTBI uptake by

cardiomyocytes (Oudit et al. 2003). L-type calcium channels are also highly expressed in

the pancreas and may contribute to NTBI uptake by pancreatic cells (Lipscombe et al.

2004).

In conclusion, this study clearly demonstrates that plasma NTBI concentration and

clearance as well as in vivo tissue NTBI uptake were significantly increased in all three

mouse models of HH. Changes in these NTBI parameters were greatest in Hfe-/-xTfr2mut

mice followed by Tfr2mut and Hfe-/- mice (Hfe-/-xTfr2mut >> Tfr2mut > Hfe-/- mice) compared

with non-iron-loaded wild-type mice. The positive linear relationship between NTBI uptake

and both plasma NTBI and tissue iron concentrations in the liver, kidney, pancreas and

heart suggests that elevated NTBI uptake from the plasma contributes to the excessive

iron deposition in the liver and to a lesser degree in the kidney, pancreas and heart in HH.

Page 122: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

107

Chapter 5

Disruption of HFE and TFR2 causes iron-induced liver

injury in mice

Page 123: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

108

The following chapter “Disruption of HFE and TFR2 causes iron-induced liver injury in

mice” has been published as:

Delima, R. D., A. C. Chua, et al. (2012). "Disruption of HFE and TFR2 causes iron-induced

liver injury in mice." Hepatology. 56(2):585-93.

Page 124: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

109

5.1 Introduction

In hereditary haemochromatosis dysrgulated iron homeostasis results in increased

intestinal iron absorption, leading to increased cellular uptake of the excess circulating iron

which is stored as ferritin and haemosiderin, primarily in the liver, but with increased

severity of iron loading it may also involve other organs. Excessive iron deposition has

been linked to tissue damage and cellular dysfunction. Progressive iron deposition in the

liver leads to fibrosis, cirrhosis and hepatocellular carcinoma with the duration of iron

loading increasing the risk of developing significant liver injury (Olynyk et al. 2005).

Studies have shown that when hepatic iron concentration exceeds 60 µmol/g, hepatic

stellate cells begin to exhibit early signs of activation, an integral event in the initiation of

hepatic fibrosis (Ramm et al. 1997). As hepatic iron levels increase further, the risk of

significant liver fibrosis and ultimately cirrhosis increases (Adams 2001). Although the

exact mechanisms of liver injury induced by iron overload have not yet been fully

elucidated, it is thought that the accumulation of excess iron-catalyzed reactive oxygen

species (ROS) plays a significant role. Previous studies have demonstrated decreased

hepatic levels of antioxidants such as superoxide dismutase (SOD), ascorbate, β-carotene

and vitamins E and A in iron overload conditions (Livrea et al. 1996; Brown et al. 1998).

Furthermore, iron increases the level of lipid peroxidation products, such as

malondialdehyde and F2-isoprostanes (Matayatsuk et al. 2007), which can cause

mutagenesis in DNA (el Ghissassi et al. 1995). Lipid peroxidation-induced DNA lesions are

increased two- to three-fold in the livers of HH patients and, together with the iron overload

seen in HH are associated with an approximately 20-fold increased risk of hepatocellular

carcinoma (Elmberg et al. 2003). Oxidative stress has been shown to activate apoptosis

and necrosis, promoting the synthesis and release of pro-inflammatory and fibrogenic

Page 125: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

110

factors that alter Kupffer cell and hepatocyte functions, triggering the activation of hepatic

stellate cells and fibrogenesis (Olynyk et al. 2008).

There is a clear association between increased hepatic iron concentration (HIC) and

fibrosis stage (Olynyk et al. 2005), and whilst several hypotheses exist on the mechanisms

responsible for hepatocellular injury, the precise pathways associated with liver cell

dysfunction and fibrogenesis remain to be fully elucidated, in part due to an absence of

suitable models.

5.2 Methods

5.2.1 Animals Animal models were generated and raised as previously described (Materials and

methods 2.2.1)

5.2.2 Tissue collection Liver tissue was collected as described in Materials and methods 2.2.1

5.2.3 Histology Liver tissue for immunohistochemistry was fixed in 10% neutral buffered formalin overnight

before being subjected to routine histological processing. Four µm liver sections were cut

by microtome and mounted on SuperFrost Plus (Menzel-Gläser, Germany) glass slides.

Liver tissue for immunofluorescence was immediately embedded in Tissue-Tek® OCT™

Compound (Sakura, The Netherlands) and frozen in liquid nitrogen. Seven µm liver

sections were cut by cryostat and mounted on SuperFrost Plus (Menzel-Gläser, Germany)

glass slides and stored at -80°C.

Page 126: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

111

5.2.4 Perls' Prussian blue staining Liver sections were dewaxed in two changes of xylene for 5 min, and then two changes of

100% ethanol for 2 min, and two changes of 75% ethanol for 5 min before being placed in

running water. Slides were then rinsed in distilled water and then placed in the prussian

blue reaction solution (2% potassium ferrocyanide : 2% hydrochloric acid) for 10-20 min.

After incubation in Prussian blue solution, the slides were rinsed in distilled water and

counterstained in 1% neutral red for 30 seconds. Once counterstained, the slides were

rinsed in distilled water and then dehydrated in two changes of 75% ethanol for 2 min

each, two changes of 100% ethanol for 2 min each, followed two changes of xylene for 5

min each. The slides were then mounted using the DePeX Mounting Medium and

coverslips.

5.2.5 Haemotoylin & Eosin staining Staining was performed on formalin-fixed paraffin embedded (FFPE) sections according to

standard histopathological methods.

5.2.6 Immunofluorescence Cluster of differentiation 45–positive (CD45+) staining was performed on methanol/acetone

(1:1) fixed liver cryosections using a rat anti-CD45 antibody (Ly-5, 1:150; BD Pharmingen,

SanDiego, CA) and detected with goat anti-rat Alexa Fluor 594 or goat antirat Alexa Fluor

488 (1:200; Invitrogen, Mulgrave, Victoria, Australia) and mounted with Long Gold antifade

reagent, containing 40,6-diamidino-2-phenylindole (DAPI; Invitrogen), for nuclear

quantitation. Quantification was performed by the acquisition of six random, non-

overlapping fields of view per tissue sample, followed by colocalization analysis of CD45

and DAPI (nuclear quantification) using the AnalySIS Life Science Professional program

(Olympus, Melbourne, Victoria, Australia). Ferritin staining was performed using a rabbit

anti-ferritin antibody (1:800; Dako, Glostrup, Denmark) and detected using a goat anti-

rabbit Alexa Fluor 594 (1:200; Invitrogen).

Page 127: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

112

5.2.7 Biochemical markers of liver injury Plasma alanine aminotransferase (ALT) was measured as an indicator of liver injury using

a kit according to the manufacturer’s instructions (Sigma Chemical Company, MO). Liver

F2-isoprostanes, a marker of lipid peroxidation, was measured using gas chromatography-

mass spectrometry using a deuterium-labeled internal standard as previously described

(Mori et al. 1999) by Professor Kevin Croft (School of Medicine and Pharmacology, Royal

Perth Hospital, UWA). The antioxidant butylated hydroxyl toluene was added to the liver

tissue to scavenge any ROS generated during tissue storage and processing. Lipid

peroxidation was also examined by measuring liver thiobarbituric acid reactive species

(TBARS), which consists predominately of the lipid peroxidation product, malondialdehyde

(MDA), using a kit according to the manufactures’ instructions (Cayman Chemical,

Sydney, Australia). As an indicator of oxidative stress the activities of the antioxidant

enzymes copper/zinc and manganese superoxide dismutase (SOD) were measured in

liver homogenate using a kit according to the manufacturer’s instructions (Cayman

Chemical, Sydney, Australia). Liver hydroxyproline content was measured colourimetrically

as a biochemical marker of liver collagen using acid hydrolyzed liver samples, according to

the manufacturer’s instructions (QuickZyme Biosciences, Leiden, Netherlands).

5.2.8 Collagen staining Staining for collagen deposition was performed on FFPE liver sections. Slides were heated

at 60°C for 1hr. Sections were then deparaffinised in Xylene and brought to water via a

decreasing ethanol series. For Sirius Red staining, sections are initially immersed in a

0.1% solution of Sirius Red (Sigma-aldrich, Australia) in saturated aqueous picric acid

(Sigma-aldrich, Australia) for 1 h. Sections were rinsed briefly in distilled water before

immersion in a 0.1% solution of Fast Green (Sigma-aldrich, Australia). Slides were rinsed

in distilled water, dehydrated in ethanol and cleared in xylene, before being mounted with

DePex (VWR, Australia).

Page 128: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

113

For Masson’s Trichrome stain for collagen deposition, after being brought to water,

sections were stained in Weigert’s Iron Haematoxylin for 10 mins. Sections were washed

in warm tap water and then distilled water, followed by staining in Biebrich Scarlet-Acid

Fuchsin, washing in distilled water and differentiation in Phosphomolybdic-

Phosphotungstic acid solution (5% Phosphomolybdic acid : 5% Phosphotungstic acid) for

15 mins. Sections were immediately transferred to Aniline Blue solution for 5 mins,

differentiated in 1% acetic acid for 2 mins and washed in distilled water. Sections were

quickly dehydrated in ethanol to avoid removal of Biebrich Scarlet-Acid Fuchsin stain,

cleared in xylene and mounted in DePex (VWR, Australia). Stained sections were digitised

using an Aperio Scanscope XT using a positive pixel count algorithm supplied by the

manufacturer (Aperio Technologies, Vista, CA). Pixel positivity was determined by the

number of pixels representing stained tissue divided by the total number of pixels in the

whole liver section.

5.2.9 Gene expression Measurement of liver gene expression was performed by RT-PCR as described in

Materials and methods 2.2.6-11.

Page 129: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

114

5.3 Results

5.3.1 Iron measurements Plasma iron levels were measured in Hfe-/-xTfr2mut, Hfe-/-, Tfr2mut, and wild-type mice and

described previously in Chapter 3. Briefly, plasma iron concentration and transferrin

saturation were higher in Hfe-/-xTfr2mut, Tfr2mut, Hfe-/- and iron-loaded WT mice compared

with non-iron-loaded WT mice (Chapter 3; Fig. 1A,B). Plasma iron levels were highest in

Hfe-/-xTfr2mut mice (Chapter 3, Fig. 1A,B) and were significantly greater than Tfr2mut and

Hfe-/- mice. Perls’ Prussian blue staining of liver sections from Hfe-/-xTfr2mut mice

demonstrated a periportal distribution of iron, similar to that seen in Hfe-/-, Tfr2mut and iron-

loaded wild-type mice. However, the intensity of iron staining was greater in Hfe-/-xTfr2mut

than in the other types of mice (Fig. 5.1B-D) as measured by Pixel positivity. These results

indicate an increased iron burden in Hfe-/-xTfr2mut mice and are corroborated by non-haem

iron measurements (Fig. 5.1A).

Page 130: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

115

Figure 5.1: Hepatic iron concentration. Hepatic iron concentration was determined biochemically and by Perls’ Prussian blue staining. (A) Results are expressed as mean ± SEM (n=5-15). a, p<0.001 vs. WT; b, p<0.001 vs. WT+Fe; c, p<0.01 vs. Hfe-/-; d, p<0.001 vs. Tfr2mut. Staining was conducted on wild-type (WT; B), iron-loaded wild-type (WT+Fe; C), Hfe-/- (D), Tfr2mut (E), and Hfe-/-

xTfr2mut (F) mice. Each panel is representative of staining from 6-8 animals.

Page 131: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

116

5.3.2 Liver histology H&E-stained liver sections from Hfe-/-xTfr2mut mice demonstrated mild inflammation with

evidence of scattered foci of infiltrating inflammatory cells throughout the liver parenchyma

(Fig. 5.2, indicated by arrows). Immunofluorescent detection of the pan leukocyte marker,

CD45, revealed that the cell aggregates consisted mainly of CD45+ inflammatory cells (Fig.

5.2A, E) that colocalized predominately, but not exclusively, with the iron storage protein,

ferritin, in periportal regions of the liver (Fig. 5.3). The number of CD45+ inflammatory cells

was significantly increased in the livers from Hfe-/-xTfr2mut mice, compared with the other

groups of mice (P<0.05), whereas the number of CD45+ cells in Hfe-/-, Tfr2mut, and iron-

loaded WT mice was not significantly different from those in non-iron-loaded WT mice (Fig.

5.2F). Another unique feature of Hfe-/-xTfr2mut mice was the evidence of inflammatory

sideronecrosis of hepatocytes, which was not observed in any other group of mice (Fig.

5.2E).

Page 132: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

117

Figure 5.2: Liver histology. H&E staining of liver sections from WT (left panel, A), iron-loaded WT (WT+Fe; B), Hfe-/-

(C), Tfr2mut (D), and Hfe-/-xTfr2mut (left panel, E) mice. Arrows indicate inflammatory sideronecrosis of hepatocytes (left panel, E). CD45-stained (red) liver sections from WT (right panel, A) and Hfe-/-xTfr2mut (right panel, E) mice. Each panel shows a representative photomicrograph of staining from 6 animals. The number of CD45+ cells (F) is expressed as mean ± SEM (n=6). a, P<0.05 versus WT; b, P<0.05 versus WT+Fe; c, P<0.05 versus Hfe-/-; d, P<0.05 versus Tfr2mut.

Page 133: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

118

Figure 5.3: CD45+ / ferritin double staining in Hfe-/-xTfr2mut mice. Regions of high hepatic iron concentration were identified in the periportal regions of the liver by immunofluorescent staining for the iron storage protein ferritin (Fn; A, D). Clusters of CD45+ inflammatory cells (B, E) were predominantly but not exclusively seen in close spatial contact with Fn+ cells (C, F; merged with DAPI for nuclear staining).

Page 134: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

119

5.3.3 Biochemical markers of liver injury Liver injury was assessed by examining plasma ALT and hepatic SOD levels. Also lipid

peroxidation was assessed by measuring liver F2-isoprostane and TBARS levels. Plasma

ALT activity was increased in Hfe-/-xTfr2mut mice by at least 1.8-fold, compared with all

other types of mice (P<0.001; Fig. 5.4A). Both hepatic copper/zinc (cytosolic) and

manganese (mitochondrial) SOD activities were decreased significantly in all HH mice. In

Hfe-/-xTfr2mut mice copper/zinc SOD levels were similar, whereas manganese SOD levels

were significantly (p<0.01) lower than Hfe-/-and Tfr2mut mice (Fig. 5.4B). Liver F2-

isoprostanes were elevated in all groups of HH mice, compared with non-iron-loaded WT

mice (p<0.01), with Hfe-/-xTfr2mut mice having similar liver F2-isoprostane levels to iron-

loaded WT mice and significantly (p<0.01) higher levels than either Hfe-/- or Tfr2mut mice

(Fig. 5.4C). Hfe-/-xTfr2mut mice had TBARS levels 30% higher than Hfe-/- and Tfr2mut mice,

which were in turn 25% higher than the wild-type mice (Fig. 5.4D).

Page 135: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

120

Figure 5.4: Biochemical markers of liver injury. Plasma ALT (A), liver copper/zinc (non-hatched bars) and manganese (hatched bars) SOD (B), liver F2-isoprotane (C), liver TBARS (D), and hydroxyproline (E) levels were measured in WT, WT+Fe, Hfe-/-, Tfr2mut, and Hfe-/-

xTfr2mut mice. Results are expressed as mean ± SEM (n=5-15). a, P<0.05 versus WT; b, P<0.05 versus WT+Fe; c, P<0.05 versus Hfe-/-; d, P<0.05 versus Tfr2mut. For manganese SOD: 1, P<0.05 versus WT; 2, P<0.01 versus WT+Fe; 3, P<0.01 versus Hfe-/-; 4, P<0.01 versus Tfr2mut.

Page 136: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

121

5.3.4 Collagen deposition Hepatic collagen deposition, a marker of fibrosis, was examined by histology using Sirius

red and Masson’s Trichrome staining and by biochemical measurement of hydroxyproline

levels. Hydroxyproline levels were increased in all iron-loaded mice. In Hfe-/-xTfr2mut mice,

hydroxyproline levels were significantly increased compared with Tfr2mut mice and both

were elevated compared with Hfe-/- and iron-loaded wild-type mice (Fig. 5.5E; p<0.05).

Likewise, Hfe-/-xTfr2mut mice had significantly (p<0.05) increased Sirius red staining

compared with Hfe-/-, Tfr2mut and iron-loaded wild-type mice, which in turn exhibited greater

collagen deposition than non-iron-loaded wild-type mice (p<0.01; Fig. 5.5A-F). Sirius red

staining revealed portal tract thickening and periportal fibrosis, and there was evidence of

portal tract bridging in Hfe-/-xTfr2mut mice, which was not evident in other groups.

Quantification of Sirius red staining correlated with HIC (r2=0.98, p=0.001; Fig. 5.6A),

plasma NTBI (r2=0.82, p=0.033; Fig. 5.6B) as well as hydroxyproline (r2=0.89, p=0.015;

Fig. 5.6C) and F2-isoprotane levels (r2=0.77, p=0.048; Fig. 5.6D) in HH mice. This

suggests that biochemical measurements of collagen levels measured were consistent

with histological observations using Sirius red staining and were dependent on both

plasma NTBI and HIC in HH mice. Furthermore, the intensity of Trichrome staining, a

commonly used but less sensitive marker of fibrosis, was also significantly enhanced in

Hfe-/-xTfr2mut and Tfr2mut mice (Fig. 5.7F) with evidence of collagen thickening in the

periportal region of the liver (Fig. 5.7A-E).

Page 137: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

122

Figure 5.5: Liver collagen deposition via Sirius red stain. Sirius red staining of liver sections from WT (A), iron-loaded WT (WT+Fe; B), Hfe-/-(C), Tfr2mut (D), and Hfe-/-xTfr2mut (E) mice. Staining intensity is quantified in (F). There was increased collagen deposition in the portal tracts of WT+Fe, Hfe-/-, and Tfr2mut mice with advanced thickening of the portal tract in Hfe-/-xTfr2mut mice. Results are expressed as mean ± SEM (n=6). a, P < 0.01 versus WT; b, P<0.05 versus WT+Fe; c, P<0.01 versus Hfe-/-; d, P<0.05 versus Tfr2mut. Each panel is a representative photomicrograph of staining from 6 animals.

Page 138: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

123

Figure 5.6: Sirius red stain correlated with iron, collagen and lipid peroxidation measurement. Sirius red staining of liver sections from WT, iron-loaded WT, Hfe-/-, Tfr2mut, and Hfe-/-

xTfr2mut mice was correlated with hepatic iron concentration (A), plasma non-tranferrin bound iron concentration (B), liver hydroxyproline content (C), and F2-Isoprostane levels (D). Each data point is expressed as mean ± SEM (n=6).

r2=0.98

p=0.001 r2=0.82

p=0.033

r2=0.77

p=0.048 r2=0.89

p=0.015

Page 139: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

124

Figure 5.7: Liver collagen deposition using Masson’s trichrome stain. Trichrome staining of liver sections from WT (A), WT+Fe B), Hfe-/-(C), Tfr2mut (D), and Hfe-/-

xTfr2mut (E) mice. Staining intensity is quantified in (F). There was increased collagen deposition in the portal tracts of WT+Fe, Hfe-/-, and Tfr2mut mice with advanced thickening of the portal tract in Hfe-/-xTfr2mut mice. Results are expressed as mean ± SEM (n=6). a, P < 0.01 versus WT; b, P<0.05 versus WT+Fe; c, P<0.01 versus Hfe-/-; d, P<0.05 versus Tfr2mut. Each panel is a representative photomicrograph of staining from 6 animals.

Page 140: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

125

5.3.5 Hepatic expression of injury-related genes The hepatic expression of the injury-related genes c-Myc, IL-10, and Tnfα were measured

via RT-PCR as were the cytokines (described in detail in Chapter 6; Fig. 6.2A, B, C) IL-1α,

IL-6 and IL-17. c-Myc expression was significantly (p<0.05) higher in Hfe-/-xTfr2mut mice

compared with the other HH and wild-type mice. c-Myc expression in Hfe-/-xTfr2mut mice

was 2-fold higher than Tfr2 mice, which in turn, was 2-fold higher than Hfe and non-iron

loaded wild-type mice and 3-fold higher than non-iron-loaded wild-type mice (Fig. 5.8A).

IL-10 mRNA in Hfe-/-xTfr2mut andTfr2mut mice was very low compared with wild-type mice.

Hfe-/- mice had IL-10 levels similar to that of wild-type mice, whilst expression in iron-

loaded wild-type mice was significantly (p<0.05) higher than all other groups of mice (Fig.

5.8B). As described in detail in Chapter 6; Fig. 6.2D liver expression of Tnfα was similar in

Hfe-/, Tfr2mut and wild-type mice and was more than 2-fold lower than in Hfe-/-xTfr2mut mice

(p<0.05; Fig. 5.8C).

Page 141: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

126

Figure 5.8: Liver expression of injury-related genes.

mRNA expression was determined by real-time qPCR for c-Myc (A), IL-10 (B) and Tnfα (C) in wild-type (WT), WT+Fe, Hfe-/-, (Tfr2mut) and Hfe-

/-xTfr2mut mice. Results are expressed as mean ± SEM (n=4-8). Saline: a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut.

Page 142: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

127

5.4 Discussion In this study, Hfe-/- and Tfr2mut mouse models of HH types 1 and 3, respectively, and a Hfe-

/-xTfr2mut mouse model were used to examine the effects of disruption of Hfe and Tfr2,

either alone or in combination, on iron-induced liver injury. The Hfe-/-xTfr2mut mouse is the

first report of a genetic HH mouse model of iron-induced liver injury, which reflects both the

iron-loaded phenotype and increased liver injury seen in HH patients.

As previously discussed Hfe-/-xTfr2mut mice had elevated plasma and hepatic iron levels,

determined by both biochemical (Chapter 3; Fig. 3.1-2) and histological methods (Chapter

5; Fig. 5.1), compared with Hfe-/- and Tfr2mut mice. In association with increased liver iron

loading, there was a pronounced elevation of plasma ALT activity, a marker of liver injury,

in Hfe-/-xTfr2mut mice. There was also mild hepatic inflammatory cell infiltration with

scattered foci of CD45+ leukocytes and some evidence of hepatocyte sideronecrosis in

Hfe-/-xTfr2mut mice. Elevated hydroxyproline levels, and Sirius red and Trichrome staining

demonstrating marked portal tract collagen deposition and portal bridging in Hfe-/-xTfr2mut

mice strongly supports the presence of liver fibrosis and was consistent with areas of high

iron accumulation. In comparison, Hfe-/- and Tfr2mut mice had less collagen deposition and

inflammation. Histological evidence of a more pronounced liver damage in Hfe-/-xTfr2mut

mice was corroborated by decreased SOD activity and enhanced lipid peroxidation in the

liver, indicating elevated hepatic oxidative stress.

Mice with deletions in both Hfe and Tfr2 have been generated on other genetic

backgrounds (Corradini et al. 2011; Wallace et al. 2011). Other models of HFE/TFR2

disruption, similar to the Hfe-/-xTfr2mut murine model, exhibited elevated plasma and liver

iron levels compared with mice with the appropriate deletion of Hfe or Tfr2. The degree of

iron overload however, varies between strains, which is consistent with previous

observations that iron metabolism is modified by genetic background (McLachlan et al.

Page 143: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

128

2011). The Hfe-/-xTfr2mut HH mouse was generated on an AKR background, which has

relatively high iron levels (McLachlan et al. 2011) and this is likely to contribute to the more

advanced pathology of iron overload-induced liver injury seen in the Hfe-/-xTfr2mut mice

compared with HFE/TFR2 mouse models on other background strains.

Rodents are generally relatively resistant to iron-induced liver injury. Dietary carbonyl iron

loading of rats for 3 months produced iron loading in hepatocytes, similar to the levels

seen in the Hfe-/-xTfr2mut mice in the present study, but demonstrated only early signs of

liver injury including increased lipid peroxidation and collagen gene expression. Long-term

iron loading was required for up to 12 months before morphological evidence of fibrosis

was observed (Pietrangelo et al. 1990; Britton et al. 1994). Dietary iron supplementation in

combination with hepatotoxins such as ethanol and carbon tetrachloride was required to

accelerate liver injury (Mackinnon et al. 1995; Lakshmi Devi and Anuradha 2010). In the

present study, the degree of liver fibrosis seen in Hfe-/-xTfr2mut mice at 3 months of age

was similar to that observed after dietary iron-loading of rats for 12 months (Pietrangelo et

al. 1990; Britton et al. 1994). In the Hfe-/-xTfr2mut mice hepatic inflammation, fibrosis and

lipid peroxidation occurred in the presence of marked elevation of both plasma NTBI and

HIC similar to those observed in human HFE-HH (Bacon et al. 1999; Breuer et al. 2000).

Furthermore, the degree of fibrosis seen in the HH mice was dependent on both HIC and

NTBI levels.

The observation that Hfe-/-xTfr2mut mice have increased plasma ALT levels is consistent

with previous observations in HH patients, where the majority of patients had mildly

elevated ALT levels (Lin and Adams 1991). Levels of the anti-oxidant enzymes, cytosolic

copper/zinc and mitochondrial manganese SOD, were both decreased in Hfe-/-xTfr2mut

mice consistent with increased oxidative stress. Earlier studies have also reported

decreased copper/zinc SOD in dietary iron-loaded animals while manganese SOD was

Page 144: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

129

decreased in Hfe knockout and increased in iron-loaded rodents (Brown et al. 1998;

Jouihan et al. 2008; Tan et al. 2011). Furthermore, lipid peroxidation was increased in HH

mice. Unexpectedly, the level of F2-isoprostanes in dietary iron-loaded mice was greater

than in HH mice with similar HIC. This may be due to differences between dietary iron

(high hepcidin) and genetic HH (low hepcidin) models of liver iron overload where variation

in cellular iron distribution between parenchymal and Kupffer cells occurs despite similar

total HIC. Though both F2-isoprostanes and TBARS show elevated levels of lipid

peroxidation in Hfe-/-xTfr2mut, Hfe-/- and Tfr2mut mice, differences in their findings may be

due the nature of the assay, where TBARS is a measure of varying lipid peroxidation

products, F2-isoprostane meaurements assay a specific lipid peroxidation product.

Mild liver inflammation was observed only in Hfe-/-xTfr2mut mice, suggesting there was an

iron concentration threshold effect. Mild inflammation has been documented in human HH

studies during the development of fibrosis and cirrhosis (Brunt 2005). Deugnier and

colleagues reported inflammatory infiltrates in approximately 50% of liver biopsies from HH

patients (Deugnier et al. 1992). Inflammation was predominantly present in portal and

periportal regions and correlated with histological iron scores, sideronecrotic changes in

hepatocytes and hepatic fibrosis. Another study showed that approximately 25% of liver

biopsies from untreated HH patients displayed moderate inflammatory infiltration (Stal et

al. 1995). Bridle et al. also reported that 60% of liver biopsies from HH patients showed

mild inflammation consisting of scattered inflammatory foci. Furthermore, patients with

hepatic inflammation had a higher incidence of hepatic fibrosis (Bridle et al. 2003). Iron-

loaded and apoptotic/necrotic hepatocytes are purported to induce the activation of hepatic

stellate cells via various signaling mechanisms resulting in enhanced production of

proinflammatory (IL-6, IL-1β, and TNFα) and profibrogenic cytokines (such as TGF-β1) as

well as the recruitment of inflammatory cells (Ramm and Ruddell 2010). This inflammatory

cytokine response has also been shown to stimulate the transcription of c-Myc, through

Page 145: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

130

the phosphorylation of Smad3 (Matsuzaki 2012), resulting in increased proliferation and

regeneration of hepatocytes, in response to liver injury. The absence of antifibrogenic IL-

10 in Hfe-/-xTfr2mut mice is likely to exacerbate liver injury in these mice. This study

provides further support for direct hepatotoxic effects of iron overload, which results from

the disruption of Hfe and Tfr2, manifesting as increased inflammation and increased

collagen deposition.

Iron plays an important part in the progression of hepatic injury, and it does this via its

ability to catalyze the formation of highly reactive and damaging ROS. ROS induces tissue

injury by promoting lipid peroxidation as well as protein and DNA modification leading

ultimately to apoptosis and necrosis. Further investigation into the molecular mechanisms

of iron toxicity and how it causes liver injury will provide a better understanding of the role

iron plays in the progression of liver disease. The Hfe-/-xTfr2mut mouse represents a model

of HH that mimics both iron overload and consequent liver injury observed in humans with

HH.

Page 146: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

131

Chapter 6

Inflammation in mouse models of hereditary

haemochromatosis

Page 147: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

132

6.1 Introduction

Iron plays important roles in both pathogen virulence and host antimicrobial resistance

(Drakesmith and Prentice 2012). Consequently, disturbances in iron metabolism may lead

to changes in host susceptibility to infection, with both primary (Ashrafian 2003) and

secondary (Gangaidzo et al. 2001) iron overload, predisposing an individual to

salmonellosis, tuberculosis and other infections. Conversely, iron deficiency is associated

with relative resistance to infection (Murray et al. 1978).

Synthesis of the key iron regulatory hormone; hepcidin, is up-regulated with iron loading

(Pigeon et al. 2001) and inflammation and down-regulated with anaemia, hypoxia and

increased erythropoiesis (Nicolas et al. 2002). However, in many pathological states,

hepcidin synthesis can be regulated by various synergistic or antagonistic signals. In iron-

loading anaemias such as, thalassaemia and sideroblastic anaemias there is a strong but

inefficient erythropoietic response (Piperno 1998) with the development of anaemia and

hypoxia (Nemeth and Ganz 2006); synergistic signals for the down-regulation of hepcidin

(Nicolas et al. 2002). Conversely, in the anaemia of chronic disease, anaemia occurs

concurrently with inflammation; a condition often seen in autoimmune diseases, chronic

infectious diseases and cancer. In the anaemia of chronic disease, the excessive

production of certain cytokines, in particular IL-6, IL-1β, TNFα and INFγ, resulting in iron

being sequestered by reticuloendothelial macrophages and hepatocytes due to impaired

iron mobilisation (Weiss and Goodnough 2005).

Hepcidin is an antimicrobial peptide of the β-defensin family found in urine (Park et al.

2001) and blood (Krause et al. 2000) and its increased synthesis in response to

lipopolysaccharide (LPS) induced inflammation is mediated via the inflammatory cytokine

IL-6 (Ganz 2003). Although the up-regulation of hepcidin by iron requires the action of

Page 148: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

133

HFE, TFR2 and HJV, inflammation regulates hepcidin expression via the IL-6 / Janus

kinase 2 (JAK2) -signal transducer and activator of transcription 3 (STAT3) pathway. IL-6

binding to its receptor activates JAK2, which in turn phosphorylates the transcription factor,

STAT3. Phosphorylated STAT3 translocates to the nucleus and binds to the STAT3-

binding site in the proximal region of the hepcidin promoter and upregulates hepcidin gene

expression (Wrighting and Andrews 2006; Pietrangelo et al. 2007; Verga Falzacappa et al.

2007). There is also evidence of crosstalk between this pathway and that of the BMP-

SMAD signaling pathway, as a functional SMAD4 (Wang et al. 2005) and a SMAD-binding

site on the hepcidin promoter has been shown to be a requirement for IL-6-mediated

hepcidin expression (Huang et al. 2009).

The aim of this study was to examine the roles of HFE and TFR2 in the inflammatory

regulation of hepcidin in Hfe knockout (Hfe-/-), Tfr2 Y245X knock-in (Tfr2mut) and HfexTfr2

double mutant (Hfe-/-xTfr2mut) mice and how inflammation together with the dysregulation

of hepcidin synthesis in HH affects iron status.

6.2 Methods

6.2.1 Animals Animal models were generated and raised as previously described (Materials and

methods 2.2.1). Mice were injected with either 0.25mg/kg LPS from Escherichia coli

055:B5 (Sigma-Aldrich)(Yeh et al. 2004) in isotonic saline or 250 µL of saline via

intraperitoneal injection.

6.2.2 Tissue collection Liver tissue was collected 4-18 h after LPS or saline injection as described in Materials

and methods 2.2.1.

Page 149: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

134

6.2.3 Plasma iron parameters Blood was collected by cardiac puncture according to the method previously described

(Materials and Methods 2.2.1). Plasma iron, transferrin saturation and non-transferrin

bound iron assays were conducted according to the methods described in Materials and

Methods 2.2.3-5.

6.2.4 Gene expression Measurement of liver gene expression was performed by RT-PCR as described in

Materials and methods 2.2.6-11.

Page 150: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

135

6.3 Results

6.3.1 Effect of LPS with time on hepcidin expression Liver tissue from wild-type mice was collected at 0, 1, 4, and 18 h post LPS injection to

determine the time point at which hepcidin gene expression was most influenced by LPS

stimulation. One h after administration of 0.25 mg/kg LPS (Yeh et al. 2004), hepatic

Hamp1 mRNA expression was increased two-fold and after 4 h Hamp1 levels were 3-fold

higher compared to base-line levels. After 18 h, Hamp1 expression was decreased to

below initial levels (Fig. 6.1). Based on this data, all subsequent analyses were conducted

on liver and blood samples collected 4 h post-LPS administration.

0 1 4 18

0

20

40

60

80

Hamp1

/β-A

ctin

mR

NA

co

py

nu

mb

er

Figure 6.1: LPS time course. Liver Hamp1 mRNA expression was determined by real-time qPCR in wild-type (WT) mice at 0, 1, 4, 18 h after injection with LPS.

Page 151: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

136

6.3.2 Hepatic expression of inflammatory genes The hepatic inflammatory response induced by LPS administration was examined by

measuring the inflammatory cytokines IL-1α, IL-6, IL-17, and TNFα gene expression by

RT-PCR. Expression of IL-1α was similar in HH and wild-type untreated mice but was

approximately 30% higher in iron-loaded wild-type mice (p<0.05; Fig. 6.2A). IL-1α

expression was increased by more than 2-fold in all LPS treated mice compared with

untreated mice (p<0.01; hatched vs. non-hatched bars). LPS treated Hfe-/-xTfr2mut mice

had significantly (p<0.05) increased IL-1α expression compared with LPS treated wild-type

and iron-loaded wild-type mice.

Hepatic expression of IL-6 was similar in LPS treated HH (Hfe-/-xTfr2mut, Tfr2mut, Hfe-/-) and

iron-loaded wild-type mice and was increased by approximately 23-fold compared with

untreated mice (p<0.01). In LPS-treated mice, IL-6 expression in HH and iron-loaded wild-

type mice was more than 2-fold higher than in non-iron-loaded mice (p<0.05; Fig. 6.2B,

hatched bars vs. non-hatched bars).

LPS treatment resulted in 14-, 9- and 35-fold increases in IL-17a mRNA expression in Hfe-

/-, Tfr2mut and Hfe-/-xTfr2mut mice, respectively (p<0.05; Fig. 6.2C, hatched bars vs. non-

hatched bars). No change in IL-17a expression was observed in LPS treated non-iron

loaded wild-type mice.

Treatment with LPS resulted in an 8-fold increase in TNFα mRNA expression in Hfe-/-

xTfr2mut mice and approximately a 7-fold increase in Hfe-/- and Tfr2mut mice when

compared with untreated mice (p<0.05; Fig. 6.2D, hatched bars vs. non-hatched bars).

LPS treated Hfe-/-xTfr2mut mice were 30% higher than LPS treated Hfe-/- and Tfr2mut mice,

and 2-fold higher than LPS treated wild-type mice (p<0.05; Fig. 6.2D).

Page 152: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

137

Figure 6.2: Liver expression of inflammatory cytokine genes: LPS vs. saline treated mice. mRNA expression was determined by real-time qPCR. IL-1α (A), IL-6 (B), IL-17 and (C) and TNFα (D) mRNA expression was measured in wild-type (WT), iron-loaded wild-type (WT+Fe), Hfe knockout (Hfe-/-), Tfr2 mutant (Tfr2mut) and Hfe-/-xTfr2mut mice after intraperitoneal injection of saline (non-hatched bars) or LPS (hatched bars). Results are expressed as mean ± SEM (n=4-8). Saline: a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut. LPS: a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut. *, p<0.05 LPS vs saline.

Page 153: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

138

6.3.3 Plasma iron parameters Plasma iron concentration in LPS and saline treated mice was higher in HH and iron-

loaded wild-type mice compared with non-iron-loaded wild-type mice (p<0.05; Fig. 6.3A).

Administration of LPS resulted in a 30% reduction of plasma iron levels in non-iron-loaded

and iron-loaded wild-type, and Hfe-/- mice compared with their saline counterparts (p<0.05;

Fig. 6.3A, hatched vs. non-hatched bars) while plasma iron levels in Tfr2mut and Hfe-/-

xTfr2mut mice were unchanged. Similarly, transferrin saturation in LPS and saline treated

mice was higher in all HH and iron-loaded wild-type mice compared with non-iron-loaded

wild-type mice (Fig. 6.3B). However, LPS treatment reduced transferrin saturation in wild-

type and Tfr2mut mice only (p<0.05; Fig. 6.3B, hatched vs. non-hatched bars). Plasma

NTBI concentration in LPS and saline treated mice was elevated in all HH and iron-loaded

wild-type mice (p<0.05; Fig. 6.3C). LPS treatment significantly (p<0.05) reduced plasma

NTBI levels in all types of mice compared with their saline treated counterparts, with a

reduction of almost 40% observed in Hfe-/-xTfr2mut mice (Fig. 6.3C, hatched vs. non-

hatched bars).

Page 154: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

139

Figure 6.3: Plasma iron parameters: LPS vs. saline treated mice. Plasma iron concentration (A), transferrin saturation (B) and non-transferrin bound iron (NTBI) concentration (C) were measured in wild-type (WT), iron-loaded wild-type (WT+Fe), Hfe-/-, Tfr2mut and Hfe-/-xTfr2mut mice after intraperitoneal injection of saline (non-hatched bars) or LPS (hatched bars). Results are expressed as mean ± SEM (n=5-10). Saline: a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut. LPS: a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut. *, p<0.05 LPS vs saline.

Page 155: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

140

6.3.4 Hepatic expression of iron regulatory genes The hepatic expression of the iron regulatory genes Hfe, Tfr2, Bmp6 and Hamp1 were

measured via RT-qPCR. Hfe expression was unchanged in LPS treated Hfe-/-xTfr2mut,

Tfr2mut, and Hfe-/- mice compared with untreated mice, but was increased by more than

20% in LPS treated wild-type and iron-loaded wild-type mice (p<0.05; Fig. 6.4A, hatched

vs. non-hatched bars). Hepatic Tfr2 expression was decreased by more than 60% in LPS

treated Hfe-/-xTfr2mut, Tfr2mut, Hfe-/- and iron-loaded wild-type mice, and decreased by 40%

in non-iron-loaded wild-type mice (p<0.05; Fig. 6.4B, hatched vs. non-hatched bars).

Bmp6 expression was decreased in all LPS treated mice compared with untreated mice

(p<0.05; Fig. 6.4C, hatched vs. non-hatched bars). Bmp6 expression was similar in all LPS

treated HH mice but lower than LPS treated iron-loaded wild-type mice (p<0.05). Hamp1

expression was increased by approximately 80-, 10-, and 6-fold in LPS treated Hfe-/-

xTfr2mut, Tfr2mut and Hfe-/- mice, respectively (p<0.05; Fig. 6.4D, hatched vs. non-hatched

bars). In LPS treated Hfe-/-xTfr2mut mice Hamp1 expression was more than 50% lower than

in LPS treated Tfr2mut and Hfe-/- mice, and more than 70% lower than LPS treated wild-

type mice. In Hfe-/- and Tfr2mut mice, LPS treatment resulted in Hamp1 levels similar to LPS

treated non-iron–loaded wild-type mice but lower than LPS treated iron-loaded wild-type

mice (p<0.05; Fig. 6.4D, hatched bars vs. non-hatched bars).

Page 156: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

141

Figure 6.4: Liver expression of iron regulatory genes: LPS vs. saline treated mice. mRNA expression was determined by real-time qPCR. Hfe (A), Tfr2 (B), Bmp6 (C), and Hamp1 (D) mRNA expression were measured in wild-type (WT), iron-loaded wild-type (WT+Fe), Hfe knockout (Hfe-/-), Tfr2 mutant (Tfr2mut) and Hfe-/-xTfr2mut mice after intraperitoneal injection of saline (non-hatched bars) or LPS (hatched bars). Results are expressed as mean ± SEM (n=4-8). Saline: a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut. LPS: a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut. *, p<0.05 saline vs. LPS.

Page 157: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

142

6.3.5 Hepatic expression of iron transport genes Tfr1 mRNA expression was decreased in all HH and iron-loaded wild-type mice by more

than 40% compared with wild-type mice. Tfr1 gene expression was further decreased by

more than 30-50% in Hfe-/-, Tfr2mut mice and Hfe-/-xTfr2mut mice treated with LPS (p<0.05;

Fig. 6.5A, hatched bars vs. non-hatched bars). Hepatic expression of Zip14 was increased

markedly in all mice after administration of LPS with a 6-8 -fold increase in Zip14 mRNA in

Hfe-/- and Tfr2mut mice and a 5-fold increase in Hfe-/-xTfr2mut mice when compared to

untreated mice (p<0.05; Fig. 6.5B, hatched bars vs. non-hatched bars). Expression of Fpn

was similar in all LPS treated mice and was reduced by more than 70% when compared to

the saline injected mice (p<0.05; Fig. 6.5C, hatched bars vs. non-hatched bars).

Page 158: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

143

Figure 6.5: Liver expression of iron transport genes in LPS and saline treated mice. mRNA expression was determined by real-time qPCR. Tfr1 (A), Zip14 (B), and Fpn (C) mRNA expression were measured in wild-type (WT), iron-loaded wild-type (WT+Fe), Hfe knockout (Hfe-/-), Tfr2 mutant (Tfr2mut) and Hfe-/-

xTfr2mut mice after intraperitoneal injection of saline (non-hatched bars) or LPS (hatched bars). Results are expressed as mean ± SEM (n=4-8). Saline: a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut. LPS: a, p<0.05 vs. WT; b, p<0.05 vs. WT+Fe; c, p<0.05 vs. Hfe-/-; d, p<0.05 vs. Tfr2mut. *, p<0.05 saline vs. LPS.

Page 159: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

144

6.4 Discussion HFE and TFR2 play important roles in the regulation of the iron regulatory hormone

hepcidin, that controls iron absorption, recycling and storage. However, hepcidin is also

regulated by pro-inflammatory cytokines during inflammation and has been implicated in

the anaemia of chronic disease (Andrews 2004). In the current study, administration of

0.25 mg/kg LPS resulted in an increase in liver Hamp1, IL-1α, IL-6, IL-17a and TNFα

expression after four h in both wild-type and HH mice, and that this was associated with

reductions in plasma iron and transferrin saturation levels in Hfe-/-, Tfr2mut and wild-type

mice compared with untreated mice, but were unchanged in LPS treated Hfe-/-xTfr2mut

mice. Furthermore, there were significant reductions in plasma NTBI levels in all LPS

treated groups of mice with approximately a 30% decrease in Hfe-/-xTfr2mut mice. LPS

treatment also decreased hepatic expression of the iron sensors Tfr2 and Bmp6, and

increased Hamp1 in both HH and wild-type mice. Expression of the iron transport genes

Tfr1 and Fpn was decreased with LPS expression, whereas Zip14 expression was

markedly increased in all mice.

In the current study, inflammation was assessed by measuring liver mRNA levels of IL-1α,

IL-6, TNFα and IL-17, all of which were markedly increased in LPS treated HH and wild-

type mice. The administration of bacterial LPS induces inflammation by binding to Toll-like

receptor 4 (TLR4), activating the transcription factor nuclear factor-kappa B (NF-

κB)(Beutler and Poltorak 2001) and inducing the production of the proinflammatory

cytokines including IL-1α, IL-1β, IL-6, IL-17 and TNFα. The activation of NF-κB has been

shown to be stimulated by iron, with iron loading increasing the expression of NF-κB

responsive genes in macrophages (Xiong et al. 2003). LPS-mediated IL-6 production

occurs via two mechanisms, firstly, IL-6 may be stimulated by the increased synthesis of

IL-1β which with or without IL-1α can complex with the IL-1 receptor and IL-1 receptor

accessory protein to stimulate IL-6 synthesis. IL-6 production may also be stimulated by

Page 160: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

145

the binding of TNFα to its p55 and p75 receptors resulting in increased IL-6 production

(Zetterstrom et al. 1998).

IL-17a is secreted by T helper 17 cells (Miossec et al. 2009) and plays an important role in

regulating tissue inflammation through the induction of proinflammatory cytokines, IL-1β,

IL-6 and TNFα (Aggarwal and Gurney 2002). IL-17a mRNA was measured in this study as

another marker of the inflammation in LPS treated mice. IL-17a and TNFα expression in

Hfe-/-xTfr2mut mice was markedly increased compared with Hfe-/- and Tfr2mut mice and this

may be due to the increased iron burden in these mice accentuating the activation of NF-

κB and in turn IL-17a and TNFα, or it may reflect the early signs of liver injury evident in

Hfe-/-xTfr2mut mice (see Chapter 5)(Delima et al. 2012). IL-17a also activates signal

transducer and activator of transcription 3 (STAT3)(Kim et al. 2012), which then

translocates to the nucleus, binds to the proximal promoter element of the hepcidin gene

(Hamp1), and activates transcription.

Similarly to IL-17a, the cytokine IL-6 promotes the phosphorylation of STAT3 protein,

resulting in enhanced hepcidin expression. In the current study Hfe-/- and Tfr2mut mice

demonstrated the appropriate upregulation of hepcidin (Hamp) in response to IL-6 mice

(Verga Falzacappa et al. 2007). However, this mechanism was impaired in Hfe-/-xTfr2mut

mice as LPS did not upregulate Hamp1 expression to the same extent as in wild-type, Hfe-

/- or Tfr2mut mice. These findings are consistent with the observations by Wallace et al, who

also described a blunted response to inflammation in the absence of both Hfe and Tfr2

(Wallace et al. 2011). It has been suggested that the decreased levels of hepcidin in Hfe-/-

and Tfr2mut mice and even more so in Hfe-/-xTfr2mut (Wallace et al. 2009; Corradini et al.

2011; Delima et al. 2012), is due to impaired BMP-SMAD signaling.

Page 161: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

146

In the current study the upregulation of Bmp6 mRNA in iron-loaded wild-type mice was the

appropriate response to increased hepatic iron levels (Corradini et al. 2011), whereas this

response was blunted in HH mice despite higher liver iron levels in Hfe-/-xTfr2mut mice than

found in the iron-loaded wild-type mice (Chapter 5, Fig. 3.3D). BMP6 expression was

downregulated in LPS treated HH and wild-type mice. Although the exact mechanism by

which LPS administration causes BMP6 downregulation is not known, it may occur

through the LPS induced downregulation of the BMP6 co-receptor HJV (Krijt et al. 2004). It

has been shown that LPS administration can downregulate HJV expression by two

mechanisms, firstly, through TLR4 signaling, and secondly via TNFα independently of IL-6

(Constante et al. 2007). Bmp6 expression in LPS treated HH mice was down-regulated to

similar extent (approximately 30%) in Hfe-/-xTfr2mut, Hfe, Tfr2 and wild-type mice

suggesting that there was no defect in the inflammatory regulation of BMP6 in HH mice,

but that the lower levels of Bmp6 in LPS treated HH mice compared with iron-loaded wild-

type mice was merely due to the lower basal level of the untreated HH mice. This is in turn

reflected in the Hamp1 expression in Hfe-/-xTfr2mut mice although able to upregulate

Hamp1 levels after LPS treatment, were not able to do so to the same extent as Hfe, Tfr2

and wild-type mice, resulting in an impaired expression of Hamp1 in Hfe-/-xTfr2mut mice.

In the current study, LPS administration decreased plasma iron levels in Hfe-/- and wild-

type mice. The reduction in plasma iron levels after LPS administration is part of the

physiological response to infectious stimuli. LPS administration upregulated the iron

regulatory hormone hepcidin. Increased hepcidin levels result in a decrease in the levels of

the iron exporter, ferroportin (Nemeth et al. 2004), resulting in decreased iron release from

hepatocytes and reticuloendothelial macrophages, ultimately leading to a decrease in

plasma iron levels. Plasma iron levels were unaltered in Hfe-/-xTfr2mut and Tfr2 mice after

LPS administration despite upregulation of hepcidin, this may be due to the extremely low

basal levels of hepcidin in these mice, that even when upregulated by LPS is unable to

Page 162: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

147

change ferroportin expression to the extent that altered plasma iron levels within the time

frame of this experiment.

LPS administration resulted in dramatic decrease in plasma NTBI levels in both HH and

wild-type mice, suggesting that LPS stimulated the removal of NTBI from the plasma,

possibly mediated by the NTBI transporter Zip14 (Liuzzi et al. 2006). In the current study

Zip14 expression was dramatically upregulated by LPS administration, this is in agreement

with previous studies which reported that liver Zip14 expression was regulated strongly by

IL-6 (Liuzzi et al. 2005). Furthermore liver Zip14 expression was increased in Hfe-/-xTfr2mut

and Tfr2mut mice suggesting a synergistic effect of liver iron loading and inflammation on

Zip14 expression. Also Fpn gene expression was found to be dramatically downregulated

in HH and wild-type mice by LPS treatment. Fpn is not regulated by iron at a

transcriptional level but by a post-translational level by hepcidin. However, the

downregulation of Fpn gene expression by LPS has also previously been shown to involve

the proinflammatory cytokines IL-1 and IL-6 (Liu et al. 2005), in a manner independent of

TNFα (Yang et al. 2002).

The anaemia of chronic disease is typically characterised by immune cell activation and an

inflammatory cytokine response via IL-6, and to lesser extent IL-1 and TNFα (Weiss and

Goodnough 2005), ultimately resulting in dysregulated iron homeostasis and impaired

erythropoiesis. This is supported by findings in the current study where LPS induced

inflammation resulted in increased expression of the iron importer, ZIP14 resulting in

decreased plasma levels of iron and NTBI and a concurrent decrease in the mRNA

expression of the iron exporter Fpn. Inflammation also resulted in an increase in hepcidin

mRNA, though this effect was diminished in HH mice. The concurrent increase in iron

importer levels and decrease in iron exporter levels, results in an iron sequestration

Page 163: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

148

phenotype evident in the anaemia of chronic disease reducing the bioavailability of iron for

erythropoiesis.

In patients with HH, the immune response to infection is altered, with increased

susceptibility to pathogens such as Vibrio fulnificus (Bullen et al. 1991) as well as

concurrent resistance to macrophage-resident pathogens (Paradkar et al. 2008). The

increased susceptibility to certain pathogens is suggested to be due to attenuated

inflammatory cytokine production in HH by iron deficient macrophages (Wang et al. 2008).

Conversely, it has been proposed that the increased resistance to bacterial infection in

Hfe-/- mice, is due to the enhanced production of the iron sequestering (Flo et al. 2004)

enterochelin-binding peptide, lipocalin-2, in response to HFE disruption (Nairz et al. 2009).

As evident in this study, mutations in iron regulatory genes HFE and TFR2 has a

significant effect on the immune response to bacterial pathogens. Iron plays important

roles in both pathogen virulence and host antimicrobial resistance (Drakesmith and

Prentice 2012). Consequently, disturbances in iron metabolism may lead to changes in

host susceptibility to infection, either via iron sequestration, as in the anaemia of chronic

disease, or by the attenuation of the innate immune response.

Page 164: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

149

Chapter 7

General discussion

Page 165: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

150

In recent years significant advances have been made in the understanding of hereditary

haemochromatosis (HH), and iron biology, in general. Medical and biological research has

been focused on several areas including firstly, the cellular and molecular mechanisms of

iron metabolism including the regulation of dietary iron absorption, transport, and storage;

secondly, the pathophysiological mechanisms of chronic iron overload; and finally, the

genetic basis of HH, diagnosis, and clinical management of iron overload diseases. The

current study addresses these areas using mouse models HH and a dietary iron overload

mouse model to examine iron transport and iron regulatory mechanisms (Chapter 3, 4, 6)

and the pathophysiological mechanisms of iron overload and iron-induced liver injury

(Chapter 5, 6).

Major breakthroughs in the understanding of HH occurred in 1996 with the discovery of the

haemochromatosis gene, HFE (Feder et al. 1996), and the development of the first HFE

knockout mouse in 1998 (Zhou et al. 1998) Further advances occurred in 2000, with the

discovery of another form of HH, caused by mutation of the TFR2 gene (Camaschella et

al. 2000) and the subsequent development of a TFR2 mutant mouse (Fleming et al. 2002).

Currently it is thought, that HFE and TFR2, form a complex on the cell surface of

hepatocytes to sense plasma transferrin saturation (Gao et al. 2009), whilst BMP6 through

its co-receptor HJV (Xia et al. 2008) and its negative regulator matriptase (Folgueras et al.

2008), sense liver iron levels (Kautz et al. 2008). HFE/TFR2 and BMP6/HJV, possibly as a

multi-protein membrane complex (D'Alessio et al. 2012), signal via the SMAD pathway to

modulate dietary iron absorption and cellular iron release, through the regulation of the

iron regulatory hormone, hepcidin.

In Chapter 3, it was shown that mice with disruption in both HFE and TFR2 (Hfe-/-xTfr2mut)

develop a more severe iron loading phenotype than mice with disruption of either HFE

(Hfe-/-) or TFR2 (Tfr2mut) alone. Haematological parameters (RBC, Hb, and Hct) were all

Page 166: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

151

higher in Hfe-/-xTfr2mut and Tfr2mut mice than in Hfe-/- mice. Iron status (plasma iron,

transferrin saturation and NTBI concentration) and liver iron content were all increased in

Hfe-/-xTfr2mut mice, and significantly higher than in Tfr2mut and Hfe-/- mice, indicative of a

more severe iron loading phenotype. These findings are consistent with the evidence

obtained in other models of HFE and TFR2 disruption (Wallace et al. 2009; Corradini et al.

2011). Hfe-/-xTfr2mut mice had elevated liver Bmp6 levels consistent with increased liver

iron content, however disruption of Hfe and Tfr2 expression resulted in ineffective p-Smad

1,5,8 signalling leading to reduced liver Hamp1.

The disruption of HFE and TFR2 in the Hfe-/-xTfr2mut mouse resulted in a form of HH

comparable to HH type 2, a disorder that is characterised by a severe, early onset iron

overload, with increased iron absorption and liver iron accumulation that is greater than

HH type 1 (HFE mutations) or type 3 (TFR2 mutations), leading to severe organ

impairment with hypogonadism and cardiac involvement prominent features of the clinical

syndrome. HH type 2 occurs as a result of mutations in HJV or the gene encoding the iron

regulatory hepcidin, known as HAMP. The observation that the combined disruption of

HFE and TFR2 resulted in a more severe phenotype than disruption of either HFE or

TFR2 alone suggests a model of iron-dependent regulation of hepcidin where both HFE

and TFR2 act as plasma iron sensors via parallel and possibly converging signalling

pathways that is as important as BMP6/HJV in the regulation of hepcidin.

Decreased hepcidin expression results in enhanced release of iron from intestinal

enterocytes and macrophages, which saturates plasma transferrin leading to the increased

presence of plasma NTBI as observed in Chapter 3 (Fig 3.1). As previously mentioned, the

predominant form of NTBI in HH is iron-citrate (Grootveld et al. 1989), however the uptake

and tissue distribution of NTBI in HH is still poorly understood. The kinetics of hepatic

NTBI transport have been predominantly studied in cell lines and isolated cells, utilising

Page 167: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

152

the human hepatoma Huh7 cell line (Trinder and Morgan 1998), and isolated rat (Baker et

al. 1998) and mouse (Chua et al. 2004) hepatocytes. These studies have provided

invaluable information regarding the mechanisms of NTBI uptake. The use of isolated cells

to the study of NTBI transport however, neglects the importance of complex interactions

between the circulation and various cells and tissues in vivo. In Chapter 4, in vivo NTBI

transport was determined in the mouse models of HH. It was shown that NTBI was cleared

rapidly from the circulation in all mouse models of HH, with most of the NTBI taken up by

the liver and to a lesser degree by the kidneys, pancreas and heart. This Chapter

describes for the first time increased in vivo tissue NTBI uptake in mouse models of HH,

and the positive correlation between NTBI uptake and both plasma NTBI levels and iron

content in the liver, kidney, pancreas and heart. This data suggest that NTBI uptake is

likely to contribute to excessive tissue iron deposition in HH. The only previously

documented studies of in vivo NTBI transport used wild-type rats with iron-saturated

transferrin and hypotransferrinaemic mice. In that study, NTBI was also shown to be

cleared rapidly from the plasma in rats with iron-saturated transferrin and

hypotransferrinaemic mice, with the majority of radiolabeled iron citrate taken up by the

liver (Craven et al. 1987).

Free iron is redox-active and can generate ROS via the Fenton and Haber-Weiss

reactions, leading to lipid peroxidation, lysosomal fragility and mitochondrial and DNA

damage (Britton et al. 2002). Although other models of combined HFE and TFR2

disruption display a similar iron overload phenotype to that of the Hfe-/-xTfr2mut mouse

(Wallace et al. 2009; Corradini et al. 2011), to date, there has been no report of iron-

induced liver injury in murine models of HH (Subramaniam et al. 2012), in stark contrast to

the well described injury in the human disease (Deugnier et al. 1992). In Chapter 5, the

first HH mouse model of iron-induced liver injury, the Hfe-/-xTfr2mut mouse, is described. In

association with increased plasma iron levels and elevated hepatic periportal iron

Page 168: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

153

deposition (Chapter 3), there was significant elevation of plasma ALT activity in Hfe-/-

xTfr2mut mice. There was also mild hepatic inflammatory cell infiltration with scattered foci

of CD45+ leukocytes colocalised predominately with ferritin in portal regions of the liver and

evidence of hepatocyte sideronecrosis in Hfe-/-xTfr2mut mice. Elevated hydroxyproline

levels, and Sirius red and Trichrome staining demonstrated marked portal tract collagen

deposition and portal bridging in Hfe-/-xTfr2mut mice, strongly supporting the presence of

liver fibrosis in areas of high iron accumulation. Histological evidence of a more

pronounced liver damage in Hfe-/-xTfr2mut mice was corroborated by increased expression

of TNFα, and decreased SOD activity and enhanced lipid peroxidation in the liver,

indicating elevated hepatic oxidative stress.

As previously described the genetic background of HH mice may contribute to the iron-

induced liver injury in murine HH. The AKR mouse strain is often described as an iron-

loading strain, with liver iron content, transferrin saturation (McLachlan et al. 2011), and in

turn lipid peroxidation product malondialdehyde levels (Gerhard et al. 2002) twice that of

the commonly used C57BL/6 strain, such that the liver injury described in Chapter 5 may

result from higher basal levels of iron in the AKR genetic strain than observed in

comparable models of HFE and TFR2 disruption on C57BL/6 (Wallace et al. 2009) and

FVB (Corradini et al. 2011) genetic backgrounds. Interestingly, AKR mice also have higher

levels of liver TBARS than the higher-iron-loading mouse strain, CBA (Sverko et al. 2002),

suggesting that iron levels alone may not be the only factor influencing liver injury in the

Hfe-/-xTfr2mut mouse. In fact, studies in human HH have shown a correlation between

genetic dimorphisms in the antioxidant enzymes SOD2 and myeloperoxidase and

increased rates of cirrhosis and hepatocellular carcinoma, suggesting that iron-induced

liver injury may be influenced by an inability to effectively ameliorate the effects of ROS,

this may also influence the degree of iron-induced injury evident in the Hfe-/-xTfr2mut

mouse.

Page 169: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

154

Although HH is not typically considered an inflammatory disease, evidence of mild

inflammation (Stal et al. 1995; Bridle et al. 2003) and periportal sideronecrosis (Deugnier

et al. 1992) has been observed in human HH liver and, as mentioned previously, in the

Hfe-/-xTfr2mut mouse (Chapter 5). In addition, inflammatory stimuli have been shown to

have a significant effect on the regulation of hepcidin (Drakesmith and Prentice 2012) and

a number of iron transporters causing anaemia of inflammation where iron required for

erythropoiesis is sequestered in the liver. In Chapter 6, the administration of LPS to HH

mice induced inflammation resulting in decreased plasma iron and NTBI levels and a

concurrent increase in the mRNA expression of liver iron importer Zip14 and decrease in

the iron exporter Fpn. Inflammation also increased hepcidin mRNA, though this effect was

diminished in HH mice. Increased hepcidin levels would result in decreased FPN protein

expression and hepatic iron export. The concurrent increase in iron importer levels and

decrease in iron exporter levels, results in liver iron retention evident in the anaemia of

inflammation, reducing the bioavailability of iron for erythropoiesis. In chronic inflammatory

conditions, with iron sequesteration in the liver, iron overload may occur over time and

cause liver injury as described in Chapter 5.

In conclusion, the disruption of both HFE and TFR2 results in an inability to sense plasma

iron levels leading to decreased synthesis of the hepatic iron regulator hepcidin, resulting

in elevated iron levels, and unmitigated iron absorption. Excess iron saturated circulating

transferrin, resulting in the presence of NTBI. Plasma NTBI was rapidly removed from the

circulation and predominantly deposited in the liver, kidney, pancreas and heart with NTBI

uptake positively correlated with both plasma NTBI and tissue iron content. The deposition

of excess iron in the liver likely contributes to the iron-induced liver injury and fibrosis in

Hfe-/-xTfr2mut mice, with liver NTBI uptake positively correlating with collagen deposition.

Systemic inflammation may also exacerbate the iron sequestration contributing further to

liver iron overload and iron-induced injury. The use of the Hfe-/-xTfr2mut mouse model of HH

Page 170: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

155

provides not only greater insight into the mechanisms of iron-induced liver injury, but also

a model for the screening of new therapeutics such as modifiers of the hepcidin pathway

and inhibitors of inflammatory pathways, to treat iron overload disorders and the anaemia

of chronic disease.

7.1 Future directions

7.1.1 The role of HFE and TFR2 and erythropoiesis An interesting finding from this study was the observation of increased haematological

parameters in conjunction with splenomegaly, in Tfr2mut and Hfe-/-xTfr2mut mice indicating a

role for TFR2 in erythropoiesis. Apart from stimulating erythropoiesis by increasing

available iron through hepcidin inhibition, TFR2 is proposed to play a role in early

erythropoiesis and erythropoietin sensitivity. However, the exact mechanism by which this

occurs is not fully understood and deserves further investigation. Experiments should

focus on measurement of serum erythropoietin and examination of the bone marrow in

Tfr2mut and Hfe-/-xTfr2mut mouse to confirm increased erythropoiesis and identify any

abnormalities in erythropoietic function.

7.1.2 NTBI Transporters Though much is known about the clearance, uptake, and distribution of NTBI, the

molecular mechanisms by which uptake occurs are yet to be fully elucidated. DMT1 and

ZIP14 have been proposed as candidate NTBI transporters, with studies showing

increased NTBI uptake in overexpressing cell lines (Liuzzi et al. 2006; Shindo et al. 2006)

and HFE knockout hepatocytes (Chua et al. 2004). However, the hepatic expression and

localisation of DMT1 and ZIP14 in murine HH has been hampered by the lack of suitable

antibodies. Studies in mice with deletion of hepatic DMT1 are still capable of accumulating

hepatic iron (Gunshin et al. 2005), in addition, the knock-out of ZIP14 in mice did not result

Page 171: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

156

in decreased iron levels (Nam et al. 2013). These results suggest that DMT1 and ZIP14

may not play a major role in NTBI uptake and that other NTBI transporters exist. Future

experiments should also examine the expression and localisation of ZIP8 and L-type

calcium channels in the pancreas and heart and lipocalin in the kidney, to determine if their

expression is regulated by iron levels in murine HH.

7.1.3 Iron-induction of liver injury Studies in the Hfe-/-xTfr2mut mouse have shown a strong positive correlation between liver

iron content, plasma NTBI concentration and liver injury. Also, in unpublished studies from

this laboratory, dietary iron-loading and increasing age of Hfe-/-xTfr2mut mice exacerbated

liver injury. However, to make a definitive statement on whether iron was indeed the

causative agent of liver injury a study should be undertaken in which Hfe-/-xTfr2mut mice are

raised on an iron-deficient diet, to confirm whether liver injury in the Hfe-/-xTfr2mut mouse is

reduced in the absence of iron. In addition, the mechanisms by which iron-induced

oxidative stress and inflammation contribute to liver injury should be examined by

quantifying and delineating the effects of ROS and by examining how lipid peroxidation

products affect mitochondrial function and in turn cell death.

Page 172: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

157

Chapter 8

Bibliography

Page 173: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

158

Abboud, S. and D. J. Haile (2000). "A novel mammalian iron-regulated protein involved in intracellular iron metabolism." J Biol Chem 275(26): 19906-19912. Adams, P. C. (2001). "Is there a threshold of hepatic iron concentration that leads to cirrhosis in C282Y hemochromatosis?" Am J Gastroenterol 96(2): 567-569. Aggarwal, S. and A. L. Gurney (2002). "IL-17: prototype member of an emerging cytokine family." J Leukoc Biol 71(1): 1-8. Ahmad, K. A., J. R. Ahmann, M. C. Migas, A. Waheed, R. S. Britton, B. R. Bacon, W. S. Sly and R. E. Fleming (2002). "Decreased liver hepcidin expression in the Hfe knockout mouse." Blood Cells Mol Dis 29(3): 361-366. Ajioka, R. S., J. E. Levy, N. C. Andrews and J. P. Kushner (2002). "Regulation of iron absorption in Hfe mutant mice." Blood 100(4): 1465-1469. Andersen, R. V., A. Tybjaerg-Hansen, M. Appleyard, H. Birgens and B. G. Nordestgaard (2004). "Hemochromatosis mutations in the general population: iron overload progression rate." Blood 103(8): 2914-2919. Anderson, G. J., L. W. Powell and J. W. Halliday (1994). "The endocytosis of transferrin by rat intestinal epithelial cells." Gastroenterology 106(2): 414-422. Andrews, N. C. (1999). "Disorders of iron metabolism." N Engl J Med 341(26): 1986-1995. Andrews, N. C. (2004). "Anemia of inflammation: the cytokine-hepcidin link." J Clin Invest 113(9): 1251-1253. Andriopoulos, B., Jr., E. Corradini, Y. Xia, S. A. Faasse, S. Chen, L. Grgurevic, M. D. Knutson, A. Pietrangelo, S. Vukicevic, H. Y. Lin and J. L. Babitt (2009). "BMP6 is a key endogenous regulator of hepcidin expression and iron metabolism." Nat Genet 41(4): 482-487. Arosio, P. and S. Levi (2010). "Cytosolic and mitochondrial ferritins in the regulation of cellular iron homeostasis and oxidative damage." Biochim Biophys Acta 1800(8): 783-792. Arredondo, M., P. Munoz, C. V. Mura and M. T. Nunez (2003). "DMT1, a physiologically relevant apical Cu1+ transporter of intestinal cells." Am J Physiol Cell Physiol 284(6): C1525-1530. Aruoma, O. I., A. Bomford, R. J. Polson and B. Halliwell (1988). "Nontransferrin-bound iron in plasma from hemochromatosis patients: effect of phlebotomy therapy." Blood 72(4): 1416-1419. Aruoma, O. I., H. Kaur and B. Halliwell (1991). "Oxygen free radicals and human diseases." J R Soc Health 111(5): 172-177. Ashrafian, H. (2003). "Hepcidin: the missing link between hemochromatosis and infections." Infect Immun 71(12): 6693-6700. Babitt, J. L., F. W. Huang, D. M. Wrighting, Y. Xia, Y. Sidis, T. A. Samad, J. A. Campagna, R. T. Chung, A. L. Schneyer, C. J. Woolf, N. C. Andrews and H. Y. Lin (2006). "Bone

Page 174: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

159

morphogenetic protein signaling by hemojuvelin regulates hepcidin expression." Nat Genet 38(5): 531-539. Bacon, B. R., R. O'Neill and C. H. Park (1986). "Iron-induced peroxidative injury to isolated rat hepatic mitochondria." J Free Radic Biol Med 2(5-6): 339-347. Bacon, B. R. and R. S. Britton (1990). "The pathology of hepatic iron overload: a free radical--mediated process?" Hepatology 11(1): 127-137. Bacon, B. R., R. O'Neill and R. S. Britton (1993). "Hepatic mitochondrial energy production in rats with chronic iron overload." Gastroenterology 105(4): 1134-1140. Bacon, B. R., J. K. Olynyk, E. M. Brunt, R. S. Britton and R. K. Wolff (1999). "HFE genotype in patients with hemochromatosis and other liver diseases." Ann Intern Med 130(12): 953-962. Bahram, S., S. Gilfillan, L. C. Kuhn, R. Moret, J. B. Schulze, A. Lebeau and K. Schumann (1999). "Experimental hemochromatosis due to MHC class I HFE deficiency: immune status and iron metabolism." Proc Natl Acad Sci U S A 96(23): 13312-13317. Baker, E., S. M. Baker and E. H. Morgan (1998). "Characterisation of non-transferrin-bound iron (ferric citrate) uptake by rat hepatocytes in culture." Biochim Biophys Acta 1380(1): 21-30. Barisani, D., C. L. Berg, M. Wessling-Resnick and J. L. Gollan (1995). "Evidence for a low Km transporter for non-transferrin-bound iron in isolated rat hepatocytes." Am J Physiol 269(4 Pt 1): G570-576. Barton, J. C., L. F. Bertoli and B. E. Rothenberg (2000). "Peripheral blood erythrocyte parameters in hemochromatosis: evidence for increased erythrocyte hemoglobin content." J Lab Clin Med 135(1): 96-104. Beker Aydemir, T., S. M. Chang, G. J. Guthrie, A. B. Maki, M. S. Ryu, A. Karabiyik and R. J. Cousins (2012). "Zinc transporter ZIP14 functions in hepatic zinc, iron and glucose homeostasis during the innate immune response (endotoxemia)." PLoS One 7(10): e48679. Besson-Fournier, C., C. Latour, L. Kautz, J. Bertrand, T. Ganz, M. P. Roth and H. Coppin (2012). "Induction of activin B by inflammatory stimuli up-regulates expression of the iron-regulatory peptide hepcidin through Smad1/5/8 signaling." Blood 120(2): 431-439. Beutler, B. and A. Poltorak (2001). "The sole gateway to endotoxin response: how LPS was identified as Tlr4, and its role in innate immunity." Drug Metab Dispos 29(4 Pt 2): 474-478. Blight, G. D. and E. H. Morgan (1983). "Ferritin and iron uptake by reticulocytes." Br J Haematol 55(1): 59-71. Brasse-Lagnel, C., Z. Karim, P. Letteron, S. Bekri, A. Bado and C. Beaumont (2011). "Intestinal DMT1 cotransporter is down-regulated by hepcidin via proteasome internalization and degradation." Gastroenterology 140(4): 1261-1271 e1261.

Page 175: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

160

Breuer, W., C. Hershko and Z. I. Cabantchik (2000). "The importance of non-transferrin bound iron in disorders of iron metabolism." Transfus Sci 23(3): 185-192. Bridle, K. R., D. M. Frazer, S. J. Wilkins, J. L. Dixon, D. M. Purdie, D. H. Crawford, V. N. Subramaniam, L. W. Powell, G. J. Anderson and G. A. Ramm (2003). "Disrupted hepcidin regulation in HFE-associated haemochromatosis and the liver as a regulator of body iron homoeostasis." Lancet 361(9358): 669-673. Bridle, K. R., D. H. Crawford, L. M. Fletcher, J. L. Smith, L. W. Powell and G. A. Ramm (2003). "Evidence for a sub-morphological inflammatory process in the liver in haemochromatosis." J Hepatol 38(4): 426-433. Brissot, P., T. L. Wright, W. L. Ma and R. A. Weisiger (1985). "Efficient clearance of non-transferrin-bound iron by rat liver. Implications for hepatic iron loading in iron overload states." J Clin Invest 76(4): 1463-1470. Brissot, P., D. Guyader, O. Loreal, F. Laine, A. Guillygomarc'h, R. Moirand and Y. Deugnier (2000). "Clinical aspects of hemochromatosis." Transfus Sci 23(3): 193-200. Britton, R. S., G. A. Ramm, J. Olynyk, R. Singh, R. O'Neill and B. R. Bacon (1994). "Pathophysiology of iron toxicity." Adv Exp Med Biol 356: 239-253. Britton, R. S., K. L. Leicester and B. R. Bacon (2002). "Iron toxicity and chelation therapy." Int J Hematol 76(3): 219-228. Brown, K. E., M. T. Kinter, T. D. Oberley, M. L. Freeman, H. F. Frierson, L. A. Ridnour, Y. Tao, L. W. Oberley and D. R. Spitz (1998). "Enhanced gamma-glutamyl transpeptidase expression and selective loss of CuZn superoxide dismutase in hepatic iron overload." Free Radic Biol Med 24(4): 545-555. Brunt, E. M. (2005). "Pathology of hepatic iron overload." Semin Liver Dis 25(4): 392-401. Bullen, J. J., P. B. Spalding, C. G. Ward and J. M. Gutteridge (1991). "Hemochromatosis, iron and septicemia caused by Vibrio vulnificus." Arch Intern Med 151(8): 1606-1609. Camaschella, C., A. Roetto, A. Cali, M. De Gobbi, G. Garozzo, M. Carella, N. Majorano, A. Totaro and P. Gasparini (2000). "The gene TFR2 is mutated in a new type of haemochromatosis mapping to 7q22." Nat Genet 25(1): 14-15. Camaschella, C., A. Roetto and M. De Gobbi (2002). "Juvenile hemochromatosis." Semin Hematol 39(4): 242-248. Camaschella, C. and E. Poggiali (2009). "Rare types of genetic hemochromatosis." Acta Haematol 122(2-3): 140-145. Camaschella, C., Roetto, A., Gobbi, M. (2002). "Juvenile hemochromatosis." Seminars in Hematology 39: 242-248. Canonne-Hergaux, F. and P. Gros (2002). "Expression of the iron transporter DMT1 in kidney from normal and anemic mk mice." Kidney Int 62(1): 147-156.

Page 176: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

161

Canonne-Hergaux, F., A. Donovan, C. Delaby, H. J. Wang and P. Gros (2006). "Comparative studies of duodenal and macrophage ferroportin proteins." Am J Physiol Gastrointest Liver Physiol 290(1): G156-163. Carlson, H., A. S. Zhang, W. H. Fleming and C. A. Enns (2005). "The hereditary hemochromatosis protein, HFE, lowers intracellular iron levels independently of transferrin receptor 1 in TRVb cells." Blood 105(6): 2564-2570. Carpenter, C. E., and Mahoney, A.W. (1992). "Contributions of heme and non-heme iron to human nutrition." Critical Reviews in Food Science and Nutrition 31: 333-367. Casanovas, G., K. Mleczko-Sanecka, S. Altamura, M. W. Hentze and M. U. Muckenthaler (2009). "Bone morphogenetic protein (BMP)-responsive elements located in the proximal and distal hepcidin promoter are critical for its response to HJV/BMP/SMAD." J Mol Med (Berl) 87(5): 471-480. Casanovas, G., D. W. Swinkels, S. Altamura, K. Schwarz, C. M. Laarakkers, H. J. Gross, M. Wiesneth, H. Heimpel and M. U. Muckenthaler (2011). "Growth differentiation factor 15 in patients with congenital dyserythropoietic anaemia (CDA) type II." J Mol Med (Berl) 89(8): 811-816. Cavill, I. (2002). "Erythropoiesis and iron." Best Pract Res Clin Haematol 15(2): 399-409. Chambers, J. C., W. Zhang, Y. Li, J. Sehmi, M. N. Wass, D. Zabaneh, C. Hoggart, H. Bayele, M. I. McCarthy, L. Peltonen, N. B. Freimer, S. K. Srai, P. H. Maxwell, M. J. Sternberg, A. Ruokonen, G. Abecasis, M. R. Jarvelin, J. Scott, P. Elliott and J. S. Kooner (2009). "Genome-wide association study identifies variants in TMPRSS6 associated with hemoglobin levels." Nat Genet 41(11): 1170-1172. Chaston, T., B. Chung, M. Mascarenhas, J. Marks, B. Patel, S. K. Srai and P. Sharp (2008). "Evidence for differential effects of hepcidin in macrophages and intestinal epithelial cells." Gut 57(3): 374-382. Chen, J. and C. A. Enns (2007). "The Cytoplasmic domain of transferrin receptor 2 dictates its stability and response to holo-transferrin in Hep3B cells." J Biol Chem 282(9): 6201-6209. Chen, J., M. Chloupkova, J. Gao, T. L. Chapman-Arvedson and C. A. Enns (2007). "HFE modulates transferrin receptor 2 levels in hepatoma cells via interactions that differ from transferrin receptor 1-HFE interactions." J Biol Chem 282(51): 36862-36870. Chen, T. T., L. Li, D. H. Chung, C. D. Allen, S. V. Torti, F. M. Torti, J. G. Cyster, C. Y. Chen, F. M. Brodsky, E. C. Niemi, M. C. Nakamura, W. E. Seaman and M. R. Daws (2005). "TIM-2 is expressed on B cells and in liver and kidney and is a receptor for H-ferritin endocytosis." J Exp Med 202(7): 955-965. Choi, J. H., Y. Latunde-Dada, A.T. Laftah, and Simpson, R.J. (2008). The role of DcytB in intestinal iron absorption, (abstract). Choi, S. O., Y. S. Cho, H. L. Kim and J. W. Park (2007). "ROS mediate the hypoxic repression of the hepcidin gene by inhibiting C/EBPalpha and STAT-3." Biochem Biophys Res Commun 356(1): 312-317.

Page 177: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

162

Chua, A. C., J. K. Olynyk, P. J. Leedman and D. Trinder (2004). "Nontransferrin-bound iron uptake by hepatocytes is increased in the Hfe knockout mouse model of hereditary hemochromatosis." Blood 104(5): 1519-1525. Chua, A. C., R. M. Graham, D. Trinder and J. K. Olynyk (2007). "The regulation of cellular iron metabolism." Crit Rev Clin Lab Sci 44(5-6): 413-459. Chua, A. C., C. E. Herbison, S. F. Drake, R. M. Graham, J. K. Olynyk and D. Trinder (2008). "The role of Hfe in transferrin-bound iron uptake by hepatocytes." Hepatology 47(5): 1737-1744. Chua, A. C., R. D. Delima, E. H. Morgan, C. E. Herbison, J. E. Tirnitz-Parker, R. M. Graham, R. E. Fleming, R. S. Britton, B. R. Bacon, J. K. Olynyk and D. Trinder (2010). "Iron uptake from plasma transferrin by a transferrin receptor 2 mutant mouse model of haemochromatosis." J Hepatol 52(3): 425-431. Chung, J., Wessling-Resnick, M. (2003). "Molecular mechanisms and regulation of iron transport." Critical Reviews in Clinical Laboratory Sciences 40: 151-182. Conrad, M. E., Benjamin, B.I., Williams, H.L., Foy, A.L. (1967). "Human absorption of hemoglobin iron." Gastroenterology 53: 5-10. Constante, M., W. Jiang, D. Wang, V. A. Raymond, M. Bilodeau and M. M. Santos (2006). "Distinct requirements for Hfe in basal and induced hepcidin levels in iron overload and inflammation." Am J Physiol Gastrointest Liver Physiol 291(2): G229-237. Constante, M., D. Wang, V. A. Raymond, M. Bilodeau and M. M. Santos (2007). "Repression of repulsive guidance molecule C during inflammation is independent of Hfe and involves tumor necrosis factor-alpha." Am J Pathol 170(2): 497-504. Corradini, E., M. Rozier, D. Meynard, A. Odhiambo, H. Y. Lin, Q. Feng, M. C. Migas, R. S. Britton, J. L. Babitt and R. E. Fleming (2011). "Iron regulation of hepcidin despite attenuated Smad1,5,8 signaling in mice without transferrin receptor 2 or Hfe." Gastroenterology. Correnti, C., V. Richardson, A. K. Sia, A. D. Bandaranayake, M. Ruiz, Y. Suryo Rahmanto, Z. Kovacevic, M. C. Clifton, M. A. Holmes, B. K. Kaiser, J. Barasch, K. N. Raymond, D. R. Richardson and R. K. Strong (2012). "Siderocalin/Lcn2/NGAL/24p3 does not drive apoptosis through gentisic acid mediated iron withdrawal in hematopoietic cell lines." PLoS One 7(8): e43696. Craven, C. M., J. Alexander, M. Eldridge, J. P. Kushner, S. Bernstein and J. Kaplan (1987). "Tissue distribution and clearance kinetics of non-transferrin-bound iron in the hypotransferrinemic mouse: a rodent model for hemochromatosis." Proc Natl Acad Sci U S A 84(10): 3457-3461. D'Alessio, F., M. W. Hentze and M. U. Muckenthaler (2012). "The hemochromatosis proteins HFE, TfR2, and HJV form a membrane-associated protein complex for hepcidin regulation." J Hepatol 57(5): 1052-1060. De Domenico, I., M. B. Vaughn, L. Li, D. Bagley, G. Musci, D. M. Ward and J. Kaplan (2006). "Ferroportin-mediated mobilization of ferritin iron precedes ferritin degradation by the proteasome." Embo J 25(22): 5396-5404.

Page 178: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

163

De Domenico, I., D. M. Ward, G. Musci and J. Kaplan (2007). "Evidence for the multimeric structure of ferroportin." Blood 109(5): 2205-2209. De Domenico, I., D. M. Ward, C. Langelier, M. B. Vaughn, E. Nemeth, W. I. Sundquist, T. Ganz, G. Musci and J. Kaplan (2007). "The molecular mechanism of hepcidin-mediated ferroportin down-regulation." Mol Biol Cell 18(7): 2569-2578. De Domenico, I., D. M. Ward, M. C. di Patti, S. Y. Jeong, S. David, G. Musci and J. Kaplan (2007). "Ferroxidase activity is required for the stability of cell surface ferroportin in cells expressing GPI-ceruloplasmin." Embo J 26(12): 2823-2831. De Domenico, I., T. Y. Zhang, C. L. Koening, R. W. Branch, N. London, E. Lo, R. A. Daynes, J. P. Kushner, D. Li, D. M. Ward and J. Kaplan (2010). "Hepcidin mediates transcriptional changes that modulate acute cytokine-induced inflammatory responses in mice." J Clin Invest 120(7): 2395-2405. De Domenico, I., E. Lo, B. Yang, T. Korolnek, I. Hamza, D. M. Ward and J. Kaplan (2011). "The role of ubiquitination in hepcidin-independent and hepcidin-dependent degradation of ferroportin." Cell Metab 14(5): 635-646. De Feo, T. M., S. Fargion, L. Duca, B. M. Cesana, L. Boncinelli, P. Lozza, M. D. Cappellini and G. Fiorelli (2001). "Non-transferrin-bound iron in alcohol abusers." Alcohol Clin Exp Res 25(10): 1494-1499. Delima, R. D., A. C. Chua, J. E. Tirnitz-Parker, E. K. Gan, K. D. Croft, R. M. Graham, J. K. Olynyk and D. Trinder (2012). "Disruption of HFE and TFR2 causes iron-induced liver injury in mice." Hepatology. Deugnier, Y. M., O. Loreal, B. Turlin, D. Guyader, H. Jouanolle, R. Moirand, C. Jacquelinet and P. Brissot (1992). "Liver pathology in genetic hemochromatosis: a review of 135 homozygous cases and their bioclinical correlations." Gastroenterology 102(6): 2050-2059. Devireddy, L. R., C. Gazin, X. Zhu and M. R. Green (2005). "A cell-surface receptor for lipocalin 24p3 selectively mediates apoptosis and iron uptake." Cell 123(7): 1293-1305. Donovan, A., C. A. Lima, J. L. Pinkus, G. S. Pinkus, L. I. Zon, S. Robine and N. C. Andrews (2005). "The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis." Cell Metab 1(3): 191-200. Doolittle, R. L. and G. W. Richter (1981). "Isoferritins in rat Kupffer cells, hepatocytes, and extrahepatic macrophages. Biosynthesis in cell suspensions and cultures in response to iron." Lab Invest 45(6): 567-574. Drake, S. F., E. H. Morgan, C. E. Herbison, R. Delima, R. M. Graham, A. C. Chua, P. J. Leedman, R. E. Fleming, B. R. Bacon, J. K. Olynyk and D. Trinder (2007). "Iron absorption and hepatic iron uptake are increased in a transferrin receptor 2 (Y245X) mutant mouse model of hemochromatosis type 3." Am J Physiol Gastrointest Liver Physiol 292(1): G323-328. Drakesmith, H. and A. M. Prentice (2012). "Hepcidin and the iron-infection axis." Science 338(6108): 768-772.

Page 179: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

164

Dupic, F., S. Fruchon, M. Bensaid, N. Borot, M. Radosavljevic, O. Loreal, P. Brissot, S. Gilfillan, S. Bahram, H. Coppin and M. P. Roth (2002). "Inactivation of the hemochromatosis gene differentially regulates duodenal expression of iron-related mRNAs between mouse strains." Gastroenterology 122(3): 745-751. el Ghissassi, F., A. Barbin, J. Nair and H. Bartsch (1995). "Formation of 1,N6-ethenoadenine and 3,N4-ethenocytosine by lipid peroxidation products and nucleic acid bases." Chem Res Toxicol 8(2): 278-283. Elmberg, M., R. Hultcrantz, A. Ekbom, L. Brandt, S. Olsson, R. Olsson, S. Lindgren, L. Loof, P. Stal, S. Wallerstedt, S. Almer, H. Sandberg-Gertzen and J. Askling (2003). "Cancer risk in patients with hereditary hemochromatosis and in their first-degree relatives." Gastroenterology 125(6): 1733-1741. Feder, J. N., A. Gnirke, W. Thomas, Z. Tsuchihashi, D. A. Ruddy, A. Basava, F. Dormishian, R. Domingo, Jr., M. C. Ellis, A. Fullan, L. M. Hinton, N. L. Jones, B. E. Kimmel, G. S. Kronmal, P. Lauer, V. K. Lee, D. B. Loeb, F. A. Mapa, E. McClelland, N. C. Meyer, G. A. Mintier, N. Moeller, T. Moore, E. Morikang, C. E. Prass, L. Quintana, S. M. Starnes, R. C. Schatzman, K. J. Brunke, D. T. Drayna, N. J. Risch, B. R. Bacon and R. K. Wolff (1996). "A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis." Nat Genet 13(4): 399-408. Fernandes, A., G. C. Preza, Y. Phung, I. De Domenico, J. Kaplan, T. Ganz and E. Nemeth (2009). "The molecular basis of hepcidin-resistant hereditary hemochromatosis." Blood 114(2): 437-443. Fielding, J. (1980). Serum iron and iron binding capacity. Methods in Hematology, IRON. J. D. Cook, Churchill Livingstone: 15-43. Fleming, R. E., J. R. Ahmann, M. C. Migas, A. Waheed, H. P. Koeffler, H. Kawabata, R. S. Britton, B. R. Bacon and W. S. Sly (2002). "Targeted mutagenesis of the murine transferrin receptor-2 gene produces hemochromatosis." Proc Natl Acad Sci USA 99(16): 10653-10658. Flo, T. H., K. D. Smith, S. Sato, D. J. Rodriguez, M. A. Holmes, R. K. Strong, S. Akira and A. Aderem (2004). "Lipocalin 2 mediates an innate immune response to bacterial infection by sequestrating iron." Nature 432(7019): 917-921. Folgueras, A. R., F. M. de Lara, A. M. Pendas, C. Garabaya, F. Rodriguez, A. Astudillo, T. Bernal, R. Cabanillas, C. Lopez-Otin and G. Velasco (2008). "Membrane-bound serine protease matriptase-2 (Tmprss6) is an essential regulator of iron homeostasis." Blood 112(6): 2539-2545. Forejtnikova, H., M. Vieillevoye, Y. Zermati, M. Lambert, R. M. Pellegrino, S. Guihard, M. Gaudry, C. Camaschella, C. Lacombe, A. Roetto, P. Mayeux and F. Verdier (2010). "Transferrin receptor 2 is a component of the erythropoietin receptor complex and is required for efficient erythropoiesis." Blood 116(24): 5357-5367. Francanzani, A. L., S. Fargion, R. Romano, A. Piperno, P. Arosio, G. Ruggeri, G. Ronchi and G. Fiorelli (1989). "Immunohistochemical evidence for a lack of ferritin in duodenal absorptive epithelial cells in idiopathic hemochromatosis." Gastroenterology 96(4): 1071-1078.

Page 180: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

165

Frazer, D. A., Wilkins, S.J., Becker, E.M., Vulpe, C.D., McKie, A.T., Trinder, D. and Anderson, G.J. (2002). "Hepcidin expression inversely correlates with the expression of duodenal iron transporters and iron absorption in rats." Gastroenterology 123: 835-844. Gaasch, J. A., W. J. Geldenhuys, P. R. Lockman, D. D. Allen and C. J. Van der Schyf (2007). "Voltage-gated calcium channels provide an alternate route for iron uptake in neuronal cell cultures." Neurochem Res 32(10): 1686-1693. Gangaidzo, I. T., V. M. Moyo, E. Mvundura, G. Aggrey, N. L. Murphree, H. Khumalo, T. Saungweme, I. Kasvosve, Z. A. Gomo, T. Rouault, J. R. Boelaert and V. R. Gordeuk (2001). "Association of pulmonary tuberculosis with increased dietary iron." J Infect Dis 184(7): 936-939. Ganz, T. (2003). "Hepcidin, a key regulator of iron metabolism and mediator of anemia of inflammation." Blood 102(3): 783-788. Gao, J., N. Zhao, M. D. Knutson and C. A. Enns (2008). "The hereditary hemochromatosis protein, HFE, inhibits iron uptake via down-regulation of Zip14 in HepG2 cells." J Biol Chem 283(31): 21462-21468. Gao, J., J. Chen, M. Kramer, H. Tsukamoto, A. S. Zhang and C. A. Enns (2009). "Interaction of the hereditary hemochromatosis protein HFE with transferrin receptor 2 is required for transferrin-induced hepcidin expression." Cell Metab 9(3): 217-227. Garrick, M. D., S. T. Singleton, F. Vargas, H. C. Kuo, L. Zhao, M. Knopfel, T. Davidson, M. Costa, P. Paradkar, J. A. Roth and L. M. Garrick (2006). "DMT1: which metals does it transport?" Biol Res 39(1): 79-85. Gerhard, G. S., E. J. Kaufmann, X. Wang, K. M. Erikson, J. Abraham, M. Grundy, J. L. Beard and M. J. Chorney (2002). "Genetic differences in hepatic lipid peroxidation potential and iron levels in mice." Mech Ageing Dev 123(2-3): 167-176. Ghio, A. J., E. Nozik-Grayck, J. Turi, I. Jaspers, D. R. Mercatante, R. Kole and C. A. Piantadosi (2003). "Superoxide-dependent iron uptake: a new role for anion exchange protein 2." Am J Respir Cell Mol Biol 29(6): 653-660. Gosriwatana, I., Loreal, O., Lu, S., Brissot, P., Porter, J., Hider, R.C. (1999). "Quantification of non-transferrin-bound iron in the presence of unsaturated transferrin." Analytical Biochemistry 273: 212-220. Goswami, T. and N. C. Andrews (2006). "Hereditary hemochromatosis protein, HFE, interaction with transferrin receptor 2 suggests a molecular mechanism for mammalian iron sensing." J Biol Chem 281(39): 28494-28498. Graham, R. M., A. C. Chua, C. E. Herbison, J. K. Olynyk and D. Trinder (2007). "Liver iron transport." World J Gastroenterol 13(35): 4725-4736. Grootveld, M., J. D. Bell, B. Halliwell, O. I. Aruoma, A. Bomford and P. J. Sadler (1989). "Non-transferrin-bound iron in plasma or serum from patients with idiopathic hemochromatosis. Characterization by high performance liquid chromatography and nuclear magnetic resonance spectroscopy." J Biol Chem 264(8): 4417-4422.

Page 181: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

166

Gunshin, H., B. Mackenzie, U. V. Berger, Y. Gunshin, M. F. Romero, W. F. Boron, S. Nussberger, J. L. Gollan and M. A. Hediger (1997). "Cloning and characterization of a mammalian proton-coupled metal-ion transporter." Nature 388(6641): 482-488. Gunshin, H., C. N. Starr, C. Direnzo, M. D. Fleming, J. Jin, E. L. Greer, V. M. Sellers, S. M. Galica and N. C. Andrews (2005). "Cybrd1 (duodenal cytochrome b) is not necessary for dietary iron absorption in mice." Blood 106(8): 2879-2883. Gunshin, H., Y. Fujiwara, A. O. Custodio, C. Direnzo, S. Robine and N. C. Andrews (2005). "Slc11a2 is required for intestinal iron absorption and erythropoiesis but dispensable in placenta and liver." J Clin Invest 115(5): 1258-1266. Guo, J. H., S. H. Juan and S. D. Aust (1998). "Suppression of cell growth by heavy chain ferritin." Biochem Biophys Res Commun 242(1): 39-45. Gutteridge, J. M., D. A. Rowley, E. Griffiths and B. Halliwell (1985). "Low-molecular-weight iron complexes and oxygen radical reactions in idiopathic haemochromatosis." Clin Sci (Lond) 68(4): 463-467. Halliwell, B. and J. M. Gutteridge (1984). "Oxygen toxicity, oxygen radicals, transition metals and disease." Biochem J 219(1): 1-14. Hansen, S. L., N. Trakooljul, H. C. Liu, A. J. Moeser and J. W. Spears (2009). "Iron transporters are differentially regulated by dietary iron, and modifications are associated with changes in manganese metabolism in young pigs." J Nutr 139(8): 1474-1479. Harding, C., Heuser, J., Stahl, P. (1983). "Receptor-mediated endocytosis of transferrin and recylcing of the transferrin receptor in rat reticulocytes." Journal of Cell Biology 97: 329-339. Harrison-Findik, D. D., E. Klein, C. Crist, J. Evans, N. Timchenko and J. Gollan (2007). "Iron-mediated regulation of liver hepcidin expression in rats and mice is abolished by alcohol." Hepatology 46(6): 1979-1985. Harrison, P. M. and P. Arosio (1996). "The ferritins: molecular properties, iron storage function and cellular regulation." Biochim Biophys Acta 1275(3): 161-203. Harrison, P. M., Ford, G.C., Rice, D.W., Smith, J.M., Treffry, A., White, J.L. (1987). "Structural and functional studies on ferritins." Biochemical Society Transactions 15: 744-748. Harrison, S. A., and Bacon, B.R. (2003). "Hereditary haemochromatosis: update for 2003." Journal of Hepatology 38: S14-S23. Hausmann, A., J. Lee and K. Pantopoulos (2011). "Redox control of iron regulatory protein 2 stability." FEBS Lett 585(4): 687-692. Hentze, M. W., M. U. Muckenthaler, B. Galy and C. Camaschella (2010). "Two to tango: regulation of Mammalian iron metabolism." Cell 142(1): 24-38. Herrmann, T., M. Muckenthaler, F. van der Hoeven, K. Brennan, S. G. Gehrke, N. Hubert, C. Sergi, H. J. Grone, I. Kaiser, I. Gosch, M. Volkmann, H. D. Riedel, M. W. Hentze, A. F. Stewart and W. Stremmel (2004). "Iron overload in adult Hfe-deficient mice independent of

Page 182: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

167

changes in the steady-state expression of the duodenal iron transporters DMT1 and Ireg1/ferroportin." J Mol Med (Berl) 82(1): 39-48. Hershko, C., G. Graham, G. W. Bates and E. A. Rachmilewitz (1978). "Non-specific serum iron in thalassaemia: an abnormal serum iron fraction of potential toxicity." Br J Haematol 40(2): 255-263. Hershko, C., J. Patz and A. Ronson (2007). "The anemia of achylia gastrica revisited." Blood Cells Mol Dis 39(2): 178-183. Hojyo, S., T. Fukada, S. Shimoda, W. Ohashi, B. H. Bin, H. Koseki and T. Hirano (2011). "The zinc transporter SLC39A14/ZIP14 controls G-protein coupled receptor-mediated signaling required for systemic growth." PLoS One 6(3): e18059. Hu, W. L. and E. Regoeczi (1992). "Hepatic heparan sulphate proteoglycan and the recycling of transferrin." Biochem Cell Biol 70(7): 535-538. Huang, F. W., J. L. Pinkus, G. S. Pinkus, M. D. Fleming and N. C. Andrews (2005). "A mouse model of juvenile hemochromatosis." J Clin Invest 115(8): 2187-2191. Huang, H., M. Constante, A. Layoun and M. M. Santos (2009). "Contribution of STAT3 and SMAD4 pathways to the regulation of hepcidin by opposing stimuli." Blood 113(15): 3593-3599. Hubert, N. and M. W. Hentze (2002). "Previously uncharacterized isoforms of divalent metal transporter (DMT)-1: implications for regulation and cellular function." Proc Natl Acad Sci U S A 99(19): 12345-12350. Huebers, H. A. and C. A. Finch (1987). "The physiology of transferrin and transferrin receptors." Physiol Rev 67(2): 520-582. Hussain, S. P., K. Raja, P. A. Amstad, M. Sawyer, L. J. Trudel, G. N. Wogan, L. J. Hofseth, P. G. Shields, T. R. Billiar, C. Trautwein, T. Hohler, P. R. Galle, D. H. Phillips, R. Markin, A. J. Marrogi and C. C. Harris (2000). "Increased p53 mutation load in nontumorous human liver of wilson disease and hemochromatosis: oxyradical overload diseases." Proc Natl Acad Sci U S A 97(23): 12770-12775. Hvidberg, V., M. B. Maniecki, C. Jacobsen, P. Hojrup, H. J. Moller and S. K. Moestrup (2005). "Identification of the receptor scavenging hemopexin-heme complexes." Blood 106(7): 2572-2579. Hvidberg, V., C. Jacobsen, R. K. Strong, J. B. Cowland, S. K. Moestrup and N. Borregaard (2005). "The endocytic receptor megalin binds the iron transporting neutrophil-gelatinase-associated lipocalin with high affinity and mediates its cellular uptake." FEBS Lett 579(3): 773-777. Iacopetta, B. J., E. H. Morgan and G. C. Yeoh (1983). "Receptor-mediated endocytosis of transferrin by developing erythroid cells from the fetal rat liver." J Histochem Cytochem 31(2): 336-344. Iancu, T. C. (1992). "Ferritin and hemosiderin in pathological tissues." Electron Microsc Rev 5(2): 209-229.

Page 183: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

168

Iancu, T. C. and H. Shiloh (1994). "Morphologic observations in iron overload: an update." Adv Exp Med Biol 356: 255-265. Iancu, T. C., Y. Deugnier, J. W. Halliday, L. W. Powell and P. Brissot (1997). "Ultrastructural sequences during liver iron overload in genetic hemochromatosis." J Hepatol 27(4): 628-638. Inman, R. S., M. M. Coughlan and M. Wessling-Resnick (1994). "Extracellular ferrireductase activity of K562 cells is coupled to transferrin-independent iron transport." Biochemistry 33(39): 11850-11857. Johnson, M. B. and C. A. Enns (2004). "Diferric transferrin regulates transferrin receptor 2 protein stability." Blood 104(13): 4287-4293. Jomova, K. and M. Valko (2011). "Advances in metal-induced oxidative stress and human disease." Toxicology 283(2-3): 65-87. Jordan, I. and J. Kaplan (1994). "The mammalian transferrin-independent iron transport system may involve a surface ferrireductase activity." Biochem J 302 ( Pt 3): 875-879. Jouihan, H. A., P. A. Cobine, R. C. Cooksey, E. A. Hoagland, S. Boudina, E. D. Abel, D. R. Winge and D. A. McClain (2008). "Iron-mediated inhibition of mitochondrial manganese uptake mediates mitochondrial dysfunction in a mouse model of hemochromatosis." Mol Med 14(3-4): 98-108. Kakhlon, O. and Z. I. Cabantchik (2002). "The labile iron pool: characterization, measurement, and participation in cellular processes(1)." Free Radic Biol Med 33(8): 1037-1046. Kautz, L., D. Meynard, A. Monnier, V. Darnaud, R. Bouvet, R. H. Wang, C. Deng, S. Vaulont, J. Mosser, H. Coppin and M. P. Roth (2008). "Iron regulates phosphorylation of Smad1/5/8 and gene expression of Bmp6, Smad7, Id1, and Atoh8 in the mouse liver." Blood 112(4): 1503-1509. Kawabata, H., R. Yang, T. Hirama, P. T. Vuong, S. Kawano, A. F. Gombart and H. P. Koeffler (1999). "Molecular cloning of transferrin receptor 2. A new member of the transferrin receptor-like family." J Biol Chem 274(30): 20826-20832. Kawabata, H., R. S. Germain, P. T. Vuong, T. Nakamaki, J. W. Said and H. P. Koeffler (2000). "Transferrin receptor 2-alpha supports cell growth both in iron-chelated cultured cells and in vivo." J Biol Chem 275(22): 16618-16625. Kawabata, H., R. S. Germain, T. Ikezoe, X. Tong, E. M. Green, A. F. Gombart and H. P. Koeffler (2001). "Regulation of expression of murine transferrin receptor 2." Blood 98(6): 1949-1954. Kawabata, H., R. E. Fleming, D. Gui, S. Y. Moon, T. Saitoh, J. O'Kelly, Y. Umehara, Y. Wano, J. W. Said and H. P. Koeffler (2005). "Expression of hepcidin is down-regulated in TfR2 mutant mice manifesting a phenotype of hereditary hemochromatosis." Blood 105(1): 376-381. Keel, S. B., R. T. Doty, Z. Yang, J. G. Quigley, J. Chen, S. Knoblaugh, P. D. Kingsley, I. De Domenico, M. B. Vaughn, J. Kaplan, J. Palis and J. L. Abkowitz (2008). "A heme export

Page 184: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

169

protein is required for red blood cell differentiation and iron homeostasis." Science 319(5864): 825-828. Kim, G., P. Khanal, S. C. Lim, H. J. Yun, S. G. Ahn, S. H. Ki and H. S. Choi (2012). "Interleukin-17 induces AP-1 activity and cellular transformation via upregulation of tumor progression locus 2 activity." Carcinogenesis 34(2): 341-350. Kingsley, D. M. (1994). "The TGF-beta superfamily: new members, new receptors, and new genetic tests of function in different organisms." Genes Dev 8(2): 133-146. Klausner, R. D., G. Ashwell, J. van Renswoude, J. B. Harford and K. R. Bridges (1983). "Binding of apotransferrin to K562 cells: explanation of the transferrin cycle." Proc Natl Acad Sci U S A 80(8): 2263-2266. Knutson, M. D., M. R. Vafa, D. J. Haile and M. Wessling-Resnick (2003). "Iron loading and erythrophagocytosis increase ferroportin 1 (FPN1) expression in J774 macrophages." Blood 102(12): 4191-4197. Kondo, H., K. Saito, J. P. Grasso and P. Aisen (1988). "Iron metabolism in the erythrophagocytosing Kupffer cell." Hepatology 8(1): 32-38. Konijn, A. M., Hershko, C. (1977). "Ferritin synthesis in inflammation. Pathogenesis of impaired iron release." British Journal of Haematology 37: 7-16. Koren, A., D. Fink, O. Admoni, Y. Tennenbaum-Rakover and C. Levin (2010). "Non-transferrin-bound labile plasma iron and iron overload in sickle-cell disease: a comparative study between sickle-cell disease and beta-thalassemic patients." Eur J Haematol 84(1): 72-78. Kozyraki, R., J. Fyfe, P. J. Verroust, C. Jacobsen, A. Dautry-Varsat, J. Gburek, T. E. Willnow, E. I. Christensen and S. K. Moestrup (2001). "Megalin-dependent cubilin-mediated endocytosis is a major pathway for the apical uptake of transferrin in polarized epithelia." Proc Natl Acad Sci U S A 98(22): 12491-12496. Krause, A., S. Neitz, H. J. Magert, A. Schulz, W. G. Forssmann, P. Schulz-Knappe and K. Adermann (2000). "LEAP-1, a novel highly disulfide-bonded human peptide, exhibits antimicrobial activity." FEBS Lett 480(2-3): 147-150. Krijt, J., M. Vokurka, K. T. Chang and E. Necas (2004). "Expression of Rgmc, the murine ortholog of hemojuvelin gene, is modulated by development and inflammation, but not by iron status or erythropoietin." Blood 104(13): 4308-4310. Kristiansen, M., J. H. Graversen, C. Jacobsen, O. Sonne, H. J. Hoffman, S. K. Law and S. K. Moestrup (2001). "Identification of the haemoglobin scavenger receptor." Nature 409(6817): 198-201. Kruszewski, M. (2003). "Labile iron pool: the main determinant of cellular response to oxidative stress." Mutat Res 531(1-2): 81-92. Lakshmi Devi, S. and C. V. Anuradha (2010). "Mitochondrial damage, cytotoxicity and apoptosis in iron-potentiated alcoholic liver fibrosis: amelioration by taurine." Amino Acids 38(3): 869-879.

Page 185: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

170

Lambert, L. A. (2012). "Molecular evolution of the transferrin family and associated receptors." Biochim Biophys Acta 1820(3): 244-255. Lee, D. H., D. Y. Liu, D. R. Jacobs, Jr., H. R. Shin, K. Song, I. K. Lee, B. Kim and R. C. Hider (2006). "Common presence of non-transferrin-bound iron among patients with type 2 diabetes." Diabetes Care 29(5): 1090-1095. Lee, P. L., Gelbart, T., West, C., Halloran, C., Beutler, E. (1998). "The human Nramp2 gene: characterization of the gene structure, alternative splicing, promoter region and polymorphisms." Blood Cells, Molecules, and Diseases 24: 199-215. Lesbordes-Brion, J. C., L. Viatte, M. Bennoun, D. Q. Lou, G. Ramey, C. Houbron, G. Hamard, A. Kahn and S. Vaulont (2006). "Targeted disruption of the hepcidin 1 gene results in severe hemochromatosis." Blood 108(4): 1402-1405. Levy, J. E., L. K. Montross, D. E. Cohen, M. D. Fleming and N. C. Andrews (1999). "The C282Y mutation causing hereditary hemochromatosis does not produce a null allele." Blood 94(1): 9-11. Li, J. Y., N. Paragas, R. M. Ned, A. Qiu, M. Viltard, T. Leete, I. R. Drexler, X. Chen, S. Sanna-Cherchi, F. Mohammed, D. Williams, C. S. Lin, K. M. Schmidt-Ott, N. C. Andrews and J. Barasch (2009). "Scara5 is a ferritin receptor mediating non-transferrin iron delivery." Dev Cell 16(1): 35-46. Li, L., C. J. Fang, J. C. Ryan, E. C. Niemi, J. A. Lebron, P. J. Bjorkman, H. Arase, F. M. Torti, S. V. Torti, M. C. Nakamura and W. E. Seaman (2009). "Binding and uptake of H-ferritin are mediated by human transferrin receptor-1." Proc Natl Acad Sci U S A 107(8): 3505-3510. Lichten, L. A. and R. J. Cousins (2009). "Mammalian zinc transporters: nutritional and physiologic regulation." Annu Rev Nutr 29: 153-176. Lichten, L. A., J. P. Liuzzi and R. J. Cousins (2009). "Interleukin-1beta contributes via nitric oxide to the upregulation and functional activity of the zinc transporter Zip14 (Slc39a14) in murine hepatocytes." Am J Physiol Gastrointest Liver Physiol 296(4): G860-867. Lin, E. and P. C. Adams (1991). "Biochemical liver profile in hemochromatosis. A survey of 100 patients." J Clin Gastroenterol 13(3): 316-320. Lin, M., R. A. Rippe, O. Niemela, G. Brittenham and H. Tsukamoto (1997). "Role of iron in NF-kappa B activation and cytokine gene expression by rat hepatic macrophages." Am J Physiol 272(6 Pt 1): G1355-1364. Lipscombe, D., T. D. Helton and W. Xu (2004). "L-type calcium channels: the low down." J Neurophysiol 92(5): 2633-2641. Liu, X. B., N. B. Nguyen, K. D. Marquess, F. Yang and D. J. Haile (2005). "Regulation of hepcidin and ferroportin expression by lipopolysaccharide in splenic macrophages." Blood Cells Mol Dis 35(1): 47-56. Liuzzi, J. P., L. A. Lichten, S. Rivera, R. K. Blanchard, T. B. Aydemir, M. D. Knutson, T. Ganz and R. J. Cousins (2005). "Interleukin-6 regulates the zinc transporter Zip14 in liver

Page 186: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

171

and contributes to the hypozincemia of the acute-phase response." Proc Natl Acad Sci U S A 102(19): 6843-6848. Liuzzi, J. P., F. Aydemir, H. Nam, M. D. Knutson and R. J. Cousins (2006). "Zip14 (Slc39a14) mediates non-transferrin-bound iron uptake into cells." Proc Natl Acad Sci U S A 103(37): 13612-13617. Livrea, M. A., L. Tesoriere, A. M. Pintaudi, A. Calabrese, A. Maggio, H. J. Freisleben, D. D'Arpa, R. D'Anna and A. Bongiorno (1996). "Oxidative stress and antioxidant status in beta-thalassemia major: iron overload and depletion of lipid-soluble antioxidants." Blood 88(9): 3608-3614. Mackinnon, M., C. Clayton, J. Plummer, M. Ahern, P. Cmielewski, A. Ilsley and P. Hall (1995). "Iron overload facilitates hepatic fibrosis in the rat alcohol/low-dose carbon tetrachloride model." Hepatology 21(4): 1083-1088. Malicka-Blaszkiewicz, M. and A. Kubicz (1979). "Partial purification and some properties of a liver alkaline ribonuclease from the frog Rana esculenta." Acta Biochim Pol 26(3): 275-283. Mao, X., B. E. Kim, F. Wang, D. J. Eide and M. J. Petris (2007). "A histidine-rich cluster mediates the ubiquitination and degradation of the human zinc transporter, hZIP4, and protects against zinc cytotoxicity." J Biol Chem 282(10): 6992-7000. Mastrogiannaki, M., P. Matak, B. Keith, M. C. Simon, S. Vaulont and C. Peyssonnaux (2009). "HIF-2alpha, but not HIF-1alpha, promotes iron absorption in mice." J Clin Invest 119(5): 1159-1166. Matak, P., T. B. Chaston, B. Chung, S. K. Srai, A. T. McKie and P. A. Sharp (2009). "Activated macrophages induce hepcidin expression in HuH7 hepatoma cells." Haematologica 94(6): 773-780. Matayatsuk, C., C. Y. Lee, R. W. Kalpravidh, P. Sirankapracha, P. Wilairat, S. Fucharoen and B. Halliwell (2007). "Elevated F2-isoprostanes in thalassemic patients." Free Radic Biol Med 43(12): 1649-1655. Matsuzaki, K. (2012). "Smad phosphoisoform signals in acute and chronic liver injury: similarities and differences between epithelial and mesenchymal cells." Cell Tissue Res 347(1): 225-243. May, J. M., Z. C. Qu and S. Mendiratta (1999). "Role of ascorbic acid in transferrin-independent reduction and uptake of iron by U-937 cells." Biochem Pharmacol 57(11): 1275-1282. May, P., Linder, P., and D. Williams (1977). "Computer simulation of metal-ion equilibria in biofluids: models for the low-molecular-weight complex distribution of calcium(II), magnesium(II), manganese(II), iron(III), copper(II), zinc(II), and lead(II) ions in human blood plasma." Journal of the Chemical Society, Dalton Transactions(6): 588-595. McCord, J. M. and I. Fridovich (1988). "Superoxide dismutase: the first twenty years (1968-1988)." Free Radic Biol Med 5(5-6): 363-369.

Page 187: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

172

McKie, A. T., P. Marciani, A. Rolfs, K. Brennan, K. Wehr, D. Barrow, S. Miret, A. Bomford, T. J. Peters, F. Farzaneh, M. A. Hediger, M. W. Hentze and R. J. Simpson (2000). "A novel duodenal iron-regulated transporter, IREG1, implicated in the basolateral transfer of iron to the circulation." Mol Cell 5(2): 299-309. McKie, A. T., D. Barrow, G. O. Latunde-Dada, A. Rolfs, G. Sager, E. Mudaly, M. Mudaly, C. Richardson, D. Barlow, A. Bomford, T. J. Peters, K. B. Raja, S. Shirali, M. A. Hediger, F. Farzaneh and R. J. Simpson (2001). "An iron-regulated ferric reductase associated with the absorption of dietary iron." Science 291(5509): 1755-1759. McLachlan, S., S. M. Lee, T. M. Steele, P. L. Hawthorne, M. A. Zapala, E. Eskin, N. J. Schork, G. J. Anderson and C. D. Vulpe (2011). "In silico QTL mapping of basal liver iron levels in inbred mouse strains." Physiol Genomics 43(3): 136-147. McLaren, C. E., J. C. Barton, V. R. Gordeuk, L. Wu, P. C. Adams, D. M. Reboussin, M. Speechley, H. Chang, R. T. Acton, E. L. Harris, A. M. Ruggiero and O. Castro (2007). "Determinants and characteristics of mean corpuscular volume and hemoglobin concentration in white HFE C282Y homozygotes in the hemochromatosis and iron overload screening study." Am J Hematol 82(10): 898-905. Meynard, D., V. Vaja, C. C. Sun, E. Corradini, S. Chen, C. Lopez-Otin, L. Grgurevic, C. C. Hong, M. Stirnberg, M. Gutschow, S. Vukicevic, J. L. Babitt and H. Y. Lin (2011). "Regulation of TMPRSS6 by BMP6 and iron in human cells and mice." Blood 118(3): 747-756. Miossec, P., T. Korn and V. K. Kuchroo (2009). "Interleukin-17 and type 17 helper T cells." N Engl J Med 361(9): 888-898. Miskimins, W. K., A. McClelland, M. P. Roberts and F. H. Ruddle (1986). "Cell proliferation and expression of the transferrin receptor gene: promoter sequence homologies and protein interactions." J Cell Biol 103(5): 1781-1788. Miyazaki, E., J. Kato, M. Kobune, K. Okumura, K. Sasaki, N. Shintani, P. Arosio and Y. Niitsu (2002). "Denatured H-ferritin subunit is a major constituent of haemosiderin in the liver of patients with iron overload." Gut 50(3): 413-419. Mizukami, S., R. Ichimura, S. Kemmochi, L. Wang, E. Taniai, K. Mitsumori and M. Shibutani (2010). "Tumor promotion by copper-overloading and its enhancement by excess iron accumulation involving oxidative stress responses in the early stage of a rat two-stage hepatocarcinogenesis model." Chem Biol Interact 185(3): 189-201. Mleczko-Sanecka, K., G. Casanovas, A. Ragab, K. Breitkopf, A. Muller, M. Boutros, S. Dooley, M. W. Hentze and M. U. Muckenthaler (2010). "SMAD7 controls iron metabolism as a potent inhibitor of hepcidin expression." Blood 115(13): 2657-2665. Morgan, E. H. (1981). "Transferrin, biochemistry, physiology and clinical significance." Molecular Aspects of Medicine 4(1): 1-123. Morgan, E. H. and E. Baker (1986). "Iron uptake and metabolism by hepatocytes." Fed Proc 45(12): 2810-2816. Morgan, E. H. (1996). Iron metabolism and transport. Hepatology: A Textbook of Liver Disease. Z. D. a. B. TD. Philadelphia, Saunders Company.

Page 188: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

173

Mori, K., H. T. Lee, D. Rapoport, I. R. Drexler, K. Foster, J. Yang, K. M. Schmidt-Ott, X. Chen, J. Y. Li, S. Weiss, J. Mishra, F. H. Cheema, G. Markowitz, T. Suganami, K. Sawai, M. Mukoyama, C. Kunis, V. D'Agati, P. Devarajan and J. Barasch (2005). "Endocytic delivery of lipocalin-siderophore-iron complex rescues the kidney from ischemia-reperfusion injury." J Clin Invest 115(3): 610-621. Mori, T. A., K. D. Croft, I. B. Puddey and L. J. Beilin (1999). "An improved method for the measurement of urinary and plasma F2-isoprostanes using gas chromatography-mass spectrometry." Anal Biochem 268(1): 117-125. Morikawa, K., F. Oseko and S. Morikawa (1995). "A role for ferritin in hematopoiesis and the immune system." Leuk Lymphoma 18(5-6): 429-433. Moroishi, T., M. Nishiyama, Y. Takeda, K. Iwai and K. I. Nakayama (2011). "The FBXL5-IRP2 axis is integral to control of iron metabolism in vivo." Cell Metab 14(3): 339-351. Muckenthaler, M. U., B. Galy and M. W. Hentze (2008). "Systemic iron homeostasis and the iron-responsive element/iron-regulatory protein (IRE/IRP) regulatory network." Annu Rev Nutr 28: 197-213. Murray, M. J., A. B. Murray, M. B. Murray and C. J. Murray (1978). "The adverse effect of iron repletion on the course of certain infections." Br Med J 2(6145): 1113-1115. Nair, J., A. Barbin, Y. Guichard and H. Bartsch (1995). "1,N6-ethenodeoxyadenosine and 3,N4-ethenodeoxycytine in liver DNA from humans and untreated rodents detected by immunoaffinity/32P-postlabeling." Carcinogenesis 16(3): 613-617. Nair, J., P. L. Carmichael, R. C. Fernando, D. H. Phillips, A. J. Strain and H. Bartsch (1998). "Lipid peroxidation-induced etheno-DNA adducts in the liver of patients with the genetic metal storage disorders Wilson's disease and primary hemochromatosis." Cancer Epidemiol Biomarkers Prev 7(5): 435-440. Nairz, M., I. Theurl, A. Schroll, M. Theurl, G. Fritsche, E. Lindner, M. Seifert, M. L. Crouch, K. Hantke, S. Akira, F. C. Fang and G. Weiss (2009). "Absence of functional Hfe protects mice from invasive Salmonella enterica serovar Typhimurium infection via induction of lipocalin-2." Blood 114(17): 3642-3651. Nairz, M., A. Schroll, A. R. Moschen, T. Sonnweber, M. Theurl, I. Theurl, N. Taub, C. Jamnig, D. Neurauter, L. A. Huber, H. Tilg, P. L. Moser and G. Weiss (2011). "Erythropoietin contrastingly affects bacterial infection and experimental colitis by inhibiting nuclear factor-kappaB-inducible immune pathways." Immunity 34(1): 61-74. Nam, H., C. Y. Wang, L. Zhang, W. Zhang, S. Hojyo, T. Fukada and M. Knutson (2013). "ZIP14 and DMT1 in the liver, pancreas, and heart are differentially regulated by iron deficiency and overload: implications for tissue iron uptake in iron-related disorders." Haematologica. Nemeth, E., M. S. Tuttle, J. Powelson, M. B. Vaughn, A. Donovan, D. M. Ward, T. Ganz and J. Kaplan (2004). "Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization." Science 306(5704): 2090-2093.

Page 189: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

174

Nemeth, E., S. Rivera, V. Gabayan, C. Keller, S. Taudorf, B. K. Pedersen and T. Ganz (2004). "IL-6 mediates hypoferremia of inflammation by inducing the synthesis of the iron regulatory hormone hepcidin." J Clin Invest 113(9): 1271-1276. Nemeth, E. and T. Ganz (2006). "Hepcidin and iron-loading anemias." Haematologica 91(6): 727-732. Nicolas, G., M. Bennoun, I. Devaux, C. Beaumont, B. Grandchamp, A. Kahn and S. Vaulont (2001). "Lack of hepcidin gene expression and severe tissue iron overload in upstream stimulatory factor 2 (USF2) knockout mice." Proc Natl Acad Sci U S A 98(15): 8780-8785. Nicolas, G., M. Bennoun, A. Porteu, S. Mativet, C. Beaumont, B. Grandchamp, M. Sirito, M. Sawadogo, A. Kahn and S. Vaulont (2002). "Severe iron deficiency anemia in transgenic mice expressing liver hepcidin." Proc Natl Acad Sci U S A 99(7): 4596-4601. Nicolas, G., C. Chauvet, L. Viatte, J. L. Danan, X. Bigard, I. Devaux, C. Beaumont, A. Kahn and S. Vaulont (2002). "The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation." J Clin Invest 110(7): 1037-1044. Niederkofler, V., R. Salie and S. Arber (2005). "Hemojuvelin is essential for dietary iron sensing, and its mutation leads to severe iron overload." J Clin Invest 115(8): 2180-2186. Nili, M., U. Shinde and P. Rotwein (2010). "Soluble repulsive guidance molecule c/hemojuvelin is a broad spectrum bone morphogenetic protein (BMP) antagonist and inhibits both BMP2- and BMP6-mediated signaling and gene expression." J Biol Chem 285(32): 24783-24792. Nilsson, R., I. J. Schultz, E. L. Pierce, K. A. Soltis, A. Naranuntarat, D. M. Ward, J. M. Baughman, P. N. Paradkar, P. D. Kingsley, V. C. Culotta, J. Kaplan, J. Palis, B. H. Paw and V. K. Mootha (2009). "Discovery of genes essential for heme biosynthesis through large-scale gene expression analysis." Cell Metab 10(2): 119-130. Noetzli, L. J., J. Papudesi, T. D. Coates and J. C. Wood (2009). "Pancreatic iron loading predicts cardiac iron loading in thalassemia major." Blood 114(19): 4021-4026. O'Connell, M. J., R. J. Ward, H. Baum and T. J. Peters (1985). "The role of iron in ferritin- and haemosiderin-mediated lipid peroxidation in liposomes." Biochem J 229(1): 135-139. Ohgami, R. S., D. R. Campagna, E. L. Greer, B. Antiochos, A. McDonald, J. Chen, J. J. Sharp, Y. Fujiwara, J. E. Barker and M. D. Fleming (2005). "Identification of a ferrireductase required for efficient transferrin-dependent iron uptake in erythroid cells." Nat Genet 37(11): 1264-1269. Oliveira, S. J., J. P. Pinto, G. Picarote, V. M. Costa, F. Carvalho, M. Rangel, M. de Sousa and S. F. de Almeida (2009). "ER stress-inducible factor CHOP affects the expression of hepcidin by modulating C/EBPalpha activity." PLoS One 4(8): e6618. Olynyk, J. K., T. G. St Pierre, R. S. Britton, E. M. Brunt and B. R. Bacon (2005). "Duration of hepatic iron exposure increases the risk of significant fibrosis in hereditary hemochromatosis: a new role for magnetic resonance imaging." Am J Gastroenterol 100(4): 837-841.

Page 190: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

175

Olynyk, J. K., D. Trinder, G. A. Ramm, R. S. Britton and B. R. Bacon (2008). "Hereditary hemochromatosis in the post-HFE era." Hepatology 48(3): 991-1001. Olynyk, J. K., Cullen, D.J., Aquilia, S., Rossi, E., Summerville, L., Powell, L.W. (1999). "A population based study of the clinical expression of haemochromatosis gene." The New England Journal of Medicine 341: 718-724. Oudit, G. Y., H. Sun, M. G. Trivieri, S. E. Koch, F. Dawood, C. Ackerley, M. Yazdanpanah, G. J. Wilson, A. Schwartz, P. P. Liu and P. H. Backx (2003). "L-type Ca2+ channels provide a major pathway for iron entry into cardiomyocytes in iron-overload cardiomyopathy." Nat Med 9(9): 1187-1194. Ouyang, Q., M. Bommakanti and W. K. Miskimins (1993). "A mitogen-responsive promoter region that is synergistically activated through multiple signalling pathways." Mol Cell Biol 13(3): 1796-1804. Pak, M., M. A. Lopez, V. Gabayan, T. Ganz and S. Rivera (2006). "Suppression of hepcidin during anemia requires erythropoietic activity." Blood 108(12): 3730-3735. Paradkar, P. N., I. De Domenico, N. Durchfort, I. Zohn, J. Kaplan and D. M. Ward (2008). "Iron depletion limits intracellular bacterial growth in macrophages." Blood 112(3): 866-874. Park, C. H., E. V. Valore, A. J. Waring and T. Ganz (2001). "Hepcidin, a urinary antimicrobial peptide synthesized in the liver." J Biol Chem 276(11): 7806-7810. Parkkila, S., A. Waheed, R. S. Britton, B. R. Bacon, X. Y. Zhou, S. Tomatsu, R. E. Fleming and W. S. Sly (1997). "Association of the transferrin receptor in human placenta with HFE, the protein defective in hereditary hemochromatosis." Proc Natl Acad Sci U S A 94(24): 13198-13202. Parola, M. and G. Robino (2001). "Oxidative stress-related molecules and liver fibrosis." J Hepatol 35(2): 297-306. Peslova, G., J. Petrak, K. Kuzelova, I. Hrdy, P. Halada, P. W. Kuchel, S. Soe-Lin, P. Ponka, R. Sutak, E. Becker, M. L. Huang, Y. Suryo Rahmanto, D. R. Richardson and D. Vyoral (2009). "Hepcidin, the hormone of iron metabolism, is bound specifically to alpha-2-macroglobulin in blood." Blood 113(24): 6225-6236. Philpott, C. C. (2002). "Molecular aspects of iron absorption: insights into the role of HFE in haemochromatosis." Hepatology 35: 993-1001. Picard, V., G. Govoni, N. Jabado and P. Gros (2000). "Nramp 2 (DCT1/DMT1) expressed at the plasma membrane transports iron and other divalent cations into a calcein-accessible cytoplasmic pool." J Biol Chem 275(46): 35738-35745. Pietrangelo, A., E. Rocchi, L. Schiaffonati, E. Ventura and G. Cairo (1990). "Liver gene expression during chronic dietary iron overload in rats." Hepatology 11(5): 798-804. Pietrangelo, A. (2004). "The ferroportin disease." Blood Cells, Molecules & Diseases 32: 131-138. Pietrangelo, A. (2004). "The ferroportin disease." Blood Cells Mol Dis 32(1): 131-138.

Page 191: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

176

Pietrangelo, A. (2004). "Hereditary hemochromatosis--a new look at an old disease." N Engl J Med 350(23): 2383-2397. Pietrangelo, A., A. Caleffi, J. Henrion, F. Ferrara, E. Corradini, H. Kulaksiz, W. Stremmel, P. Andreone and C. Garuti (2005). "Juvenile hemochromatosis associated with pathogenic mutations of adult hemochromatosis genes." Gastroenterology 128(2): 470-479. Pietrangelo, A. (2006). "Hereditary hemochromatosis." Biochim Biophys Acta 1763(7): 700-710. Pietrangelo, A., U. Dierssen, L. Valli, C. Garuti, A. Rump, E. Corradini, M. Ernst, C. Klein and C. Trautwein (2007). "STAT3 is required for IL-6-gp130-dependent activation of hepcidin in vivo." Gastroenterology 132(1): 294-300. Pigeon, C., G. Ilyin, B. Courselaud, P. Leroyer, B. Turlin, P. Brissot and O. Loreal (2001). "A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload." J Biol Chem 276(11): 7811-7819. Pinilla-Tenas, J. J., B. K. Sparkman, A. Shawki, A. C. Illing, C. J. Mitchell, N. Zhao, J. P. Liuzzi, R. J. Cousins, M. D. Knutson and B. Mackenzie (2011). "Zip14 is a complex broad-scope metal-ion transporter whose functional properties support roles in the cellular uptake of zinc and nontransferrin-bound iron." Am J Physiol Cell Physiol 301(4): C862-871. Pinto, J. P., S. Ribeiro, H. Pontes, S. Thowfeequ, D. Tosh, F. Carvalho and G. Porto (2008). "Erythropoietin mediates hepcidin expression in hepatocytes through EPOR signaling and regulation of C/EBPalpha." Blood 111(12): 5727-5733. Piperno, A. (1998). "Classification and diagnosis of iron overload." Haematologica 83(5): 447-455. Ponka, P. (1997). "Tissue-specific regulation of iron metabolism and heme synthesis: distinct control mechanisms in erythroid cells." Blood 89(1): 1-25. Ponka, P., C. Beaumont and D. R. Richardson (1998). "Function and regulation of transferrin and ferritin." Semin Hematol 35(1): 35-54. Pootrakul, P., W. Breuer, M. Sametband, P. Sirankapracha, C. Hershko and Z. I. Cabantchik (2004). "Labile plasma iron (LPI) as an indicator of chelatable plasma redox activity in iron-overloaded beta-thalassemia/HbE patients treated with an oral chelator." Blood 104(5): 1504-1510. Porto, G., H. Alves, P. Rodrigues, J. M. Cabeda, C. Portal, A. Ruivo, B. Justica, R. Wolff and M. De Sousa (1998). "Major histocompatibility complex class I associations in iron overload: evidence for a new link between the HFE H63D mutation, HLA-A29, and non-classical forms of hemochromatosis." Immunogenetics 47(5): 404-410. Pozo, A. L., E. M. Godfrey and K. M. Bowles (2009). "Splenomegaly: investigation, diagnosis and management." Blood Rev 23(3): 105-111. Prakash, M., S. Upadhya and R. Prabhu (2005). "Serum non-transferrin bound iron in hemodialysis patients not receiving intravenous iron." Clin Chim Acta 360(1-2): 194-198.

Page 192: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

177

Qian, Z. M. and E. H. Morgan (1992). "Changes in the uptake of transferrin-free and transferrin-bound iron during reticulocyte maturation in vivo and in vitro." Biochim Biophys Acta 1135(1): 35-43. Qiu, A., M. Jansen, A. Sakaris, S. H. Min, S. Chattopadhyay, E. Tsai, C. Sandoval, R. Zhao, M. H. Akabas and I. D. Goldman (2006). "Identification of an intestinal folate transporter and the molecular basis for hereditary folate malabsorption." Cell 127(5): 917-928. Raffin, S. B., Woo, C.H., Roost, K.T., Price, D.C., Schmid, R. (1974). "Intestinal absorption of hemoglobin iron-heme cleavage by mucosal heme oxygenase." Journal of Clinical Investigation 54: 1344-1352. Raje, C. I., S. Kumar, A. Harle, J. S. Nanda and M. Raje (2007). "The macrophage cell surface glyceraldehyde-3-phosphate dehydrogenase is a novel transferrin receptor." J Biol Chem 282(5): 3252-3261. Ramey, G., J. C. Deschemin and S. Vaulont (2009). "Cross-talk between the mitogen activated protein kinase and bone morphogenetic protein/hemojuvelin pathways is required for the induction of hepcidin by holotransferrin in primary mouse hepatocytes." Haematologica 94(6): 765-772. Ramirez, J. M., O. Schaad, S. Durual, D. Cossali, M. Docquier, P. Beris, P. Descombes and T. Matthes (2009). "Growth differentiation factor 15 production is necessary for normal erythroid differentiation and is increased in refractory anaemia with ring-sideroblasts." Br J Haematol 144(2): 251-262. Ramm, G. A., D. H. Crawford, L. W. Powell, N. I. Walker, L. M. Fletcher and J. W. Halliday (1997). "Hepatic stellate cell activation in genetic haemochromatosis. Lobular distribution, effect of increasing hepatic iron and response to phlebotomy." J Hepatol 26(3): 584-592. Ramm, G. A. and R. G. Ruddell (2010). "Iron homeostasis, hepatocellular injury, and fibrogenesis in hemochromatosis: the role of inflammation in a noninflammatory liver disease." Semin Liver Dis 30(3): 271-287. Ramos, P., E. Guy, N. Chen, C. C. Proenca, S. Gardenghi, C. Casu, A. Follenzi, N. Van Rooijen, R. W. Grady, M. de Sousa and S. Rivella (2011). "Enhanced erythropoiesis in Hfe-KO mice indicates a role for Hfe in the modulation of erythroid iron homeostasis." Blood 117(4): 1379-1389. Randell, E. W., J. G. Parkes, N. F. Olivieri and D. M. Templeton (1994). "Uptake of non-transferrin-bound iron by both reductive and nonreductive processes is modulated by intracellular iron." J Biol Chem 269(23): 16046-16053. Reardon, T. F. and D. G. Allen (2009). "Iron injections in mice increase skeletal muscle iron content, induce oxidative stress and reduce exercise performance." Exp Physiol 94(6): 720-730. Richardson, D. R., D. J. Lane, E. M. Becker, M. L. Huang, M. Whitnall, Y. Suryo Rahmanto, A. D. Sheftel and P. Ponka (2010). "Mitochondrial iron trafficking and the integration of iron metabolism between the mitochondrion and cytosol." Proc Natl Acad Sci U S A 107(24): 10775-10782.

Page 193: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

178

Robach, P., S. Recalcati, D. Girelli, C. Gelfi, N. J. Aachmann-Andersen, J. J. Thomsen, A. M. Norgaard, A. Alberghini, N. Campostrini, A. Castagna, A. Vigano, P. Santambrogio, T. Kempf, K. C. Wollert, S. Moutereau, C. Lundby and G. Cairo (2009). "Alterations of systemic and muscle iron metabolism in human subjects treated with low-dose recombinant erythropoietin." Blood 113(26): 6707-6715. Robb, A. and M. Wessling-Resnick (2004). "Regulation of transferrin receptor 2 protein levels by transferrin." Blood 104(13): 4294-4299. Roetto, A., G. Papanikolaou, M. Politou, F. Alberti, D. Girelli, J. Christakis, D. Loukopoulos and C. Camaschella (2003). "Mutant antimicrobial peptide hepcidin is associated with severe juvenile hemochromatosis." Nat Genet 33(1): 21-22. Rouault, T. A. (2006). "The role of iron regulatory proteins in mammalian iron homeostasis and disease." Nat Chem Biol 2(8): 406-414. Rueda Adel, C., N. C. Grande, E. A. Fernandez, R. Enriquez de Salamanca, L. A. Sala and M. J. Jimenez (2011). "Mutations in HFE and TFR2 genes in a Spanish patient with hemochromatosis." Rev Esp Enferm Dig 103(7): 379-382. Ryter, S. W. and R. M. Tyrrell (2000). "The heme synthesis and degradation pathways: role in oxidant sensitivity. Heme oxygenase has both pro- and antioxidant properties." Free Radic Biol Med 28(2): 289-309. Sahlstedt, L., L. von Bonsdorff, F. Ebeling, J. Parkkinen, E. Juvonen and T. Ruutu (2009). "Non-transferrin-bound iron in haematological patients during chemotherapy and conditioning for autologous stem cell transplantation." Eur J Haematol 83(5): 455-459. Salama, M. F., H. K. Bayele and S. S. Srai (2012). "Tumour necrosis factor alpha downregulates human hemojuvelin expression via a novel response element within its promoter." J Biomed Sci 19: 83. Schmidt-Ott, K. M., K. Mori, A. Kalandadze, J. Y. Li, N. Paragas, T. Nicholas, P. Devarajan and J. Barasch (2006). "Neutrophil gelatinase-associated lipocalin-mediated iron traffic in kidney epithelia." Curr Opin Nephrol Hypertens 15(4): 442-449. Seiser, C., S. Teixeira and L. C. Kuhn (1993). "Interleukin-2-dependent transcriptional and post-transcriptional regulation of transferrin receptor mRNA." J Biol Chem 268(18): 13074-13080. Selden, C., M. Owen, J. M. Hopkins and T. J. Peters (1980). "Studies on the concentration and intracellular localization of iron proteins in liver biopsy specimens from patients with iron overload with special reference to their role in lysosomal disruption." Br J Haematol 44(4): 593-603. Severance, S. and I. Hamza (2009). "Trafficking of heme and porphyrins in metazoa." Chem Rev 109(10): 4596-4616. Shah, Y. M., T. Matsubara, S. Ito, S. H. Yim and F. J. Gonzalez (2009). "Intestinal hypoxia-inducible transcription factors are essential for iron absorption following iron deficiency." Cell Metab 9(2): 152-164.

Page 194: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

179

Shanmugam, N. K., S. Ellenbogen, E. Trebicka, L. Wang, S. Mukhopadhyay, A. Lacy-Hulbert, C. A. Gallini, W. S. Garrett and B. J. Cherayil (2012). "Tumor necrosis factor alpha inhibits expression of the iron regulating hormone hepcidin in murine models of innate colitis." PLoS One 7(5): e38136. Shaw, G. C., J. J. Cope, L. Li, K. Corson, C. Hersey, G. E. Ackermann, B. Gwynn, A. J. Lambert, R. A. Wingert, D. Traver, N. S. Trede, B. A. Barut, Y. Zhou, E. Minet, A. Donovan, A. Brownlie, R. Balzan, M. J. Weiss, L. L. Peters, J. Kaplan, L. I. Zon and B. H. Paw (2006). "Mitoferrin is essential for erythroid iron assimilation." Nature 440(7080): 96-100. Shayeghi, M., G. O. Latunde-Dada, J. S. Oakhill, A. H. Laftah, K. Takeuchi, N. Halliday, Y. Khan, A. Warley, F. E. McCann, R. C. Hider, D. M. Frazer, G. J. Anderson, C. D. Vulpe, R. J. Simpson and A. T. McKie (2005). "Identification of an intestinal heme transporter." Cell 122(5): 789-801. Sheth, S., and Brittenham, G.M. (2000). "Genetic disorders affecting proteins or iron metabolism: clinical implications." Annual Review of Medicine 51: 443-464. Shindo, M., Y. Torimoto, H. Saito, W. Motomura, K. Ikuta, K. Sato, Y. Fujimoto and Y. Kohgo (2006). "Functional role of DMT1 in transferrin-independent iron uptake by human hepatocyte and hepatocellular carcinoma cell, HLF." Hepatol Res 35(3): 152-162. Siddique, A. and K. V. Kowdley (2012). "Review article: the iron overload syndromes." Aliment Pharmacol Ther 35(8): 876-893. Silva, A. M. and R. C. Hider (2009). "Influence of non-enzymatic post-translation modifications on the ability of human serum albumin to bind iron. Implications for non-transferrin-bound iron speciation." Biochim Biophys Acta 1794(10): 1449-1458. Silvestri, L., A. Pagani, A. Nai, I. De Domenico, J. Kaplan and C. Camaschella (2008). "The serine protease matriptase-2 (TMPRSS6) inhibits hepcidin activation by cleaving membrane hemojuvelin." Cell Metab 8(6): 502-511. Smith, C. P. and F. Thevenod (2009). "Iron transport and the kidney." Biochim Biophys Acta 1790(7): 724-730. Smith, P. K., R. I. Krohn, G. T. Hermanson, A. K. Mallia, F. H. Gartner, M. D. Provenzano, E. K. Fujimoto, N. M. Goeke, B. J. Olson and D. C. Klenk (1985). "Measurement of protein using bicinchoninic acid." Anal Biochem 150(1): 76-85. Sochaski, M. A., W. J. Bartfay, S. R. Thorpe, J. W. Baynes, E. Bartfay, D. C. Lehotay and P. P. Liu (2002). "Lipid peroxidation and protein modification in a mouse model of chronic iron overload." Metabolism 51(5): 645-651. Spivak, J. L. (2000). "The blood in systemic disorders." Lancet 355(9216): 1707-1712. Sposi, N. M., L. Cianetti, E. Tritarelli, E. Pelosi, S. Militi, T. Barberi, M. Gabbianelli, E. Saulle, L. Kuhn, C. Peschle and U. Testa (2000). "Mechanisms of differential transferrin receptor expression in normal hematopoiesis." Eur J Biochem 267(23): 6762-6774.

Page 195: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

180

Srivastava, G., I. A. Borthwick, D. J. Maguire, C. J. Elferink, M. J. Bawden, J. F. Mercer and B. K. May (1988). "Regulation of 5-aminolevulinate synthase mRNA in different rat tissues." J Biol Chem 263(11): 5202-5209. Stal, P., U. Broome, A. Scheynius, R. Befrits and R. Hultcrantz (1995). "Kupffer cell iron overload induces intercellular adhesion molecule-1 expression on hepatocytes in genetic hemochromatosis." Hepatology 21(5): 1308-1316. Steinbicker, A. U., T. B. Bartnikas, L. K. Lohmeyer, P. Leyton, C. Mayeur, S. M. Kao, A. E. Pappas, R. T. Peterson, D. B. Bloch, P. B. Yu, M. D. Fleming and K. D. Bloch (2011). "Perturbation of hepcidin expression by BMP type I receptor deletion induces iron overload in mice." Blood 118(15): 4224-4230. Su, D., J. M. May, M. J. Koury and H. Asard (2006). "Human erythrocyte membranes contain a cytochrome b561 that may be involved in extracellular ascorbate recycling." J Biol Chem 281(52): 39852-39859. Su, M. A., Trenor, C.C., Fleming, J.C., Fleming, M.D. and Andrews, N.C. (1998). "The G185R mutation disrupts function of the iron transporter Nramp2." Blood 92: 2157-2163. Subramaniam, V. N., C. J. McDonald, L. Ostini, P. E. Lusby, L. F. Wockner, G. A. Ramm and D. F. Wallace (2012). "Hepatic iron deposition does not predict extrahepatic iron loading in mouse models of hereditary hemochromatosis." Am J Pathol 181(4): 1173-1179. Sverko, V., T. Balog, S. Sobocanec, M. Gavella and T. Marotti (2002). "Age-associated alteration of lipid peroxidation and superoxide dismutase activity in CBA and AKR mice." Exp Gerontol 37(8-9): 1031-1039. Tacchini, L., L. Bianchi, A. Bernelli-Zazzera and G. Cairo (1999). "Transferrin receptor induction by hypoxia. HIF-1-mediated transcriptional activation and cell-specific post-transcriptional regulation." J Biol Chem 274(34): 24142-24146. Takagi, H., D. Shi, Y. Ha, N. M. Allewell and E. C. Theil (1998). "Localized unfolding at the junction of three ferritin subunits. A mechanism for iron release?" J Biol Chem 273(30): 18685-18688. Tan, T. C., D. H. Crawford, L. A. Jaskowski, T. M. Murphy, M. L. Heritage, V. N. Subramaniam, A. D. Clouston, G. J. Anderson and L. M. Fletcher (2011). "Altered lipid metabolism in Hfe-knockout mice promotes severe NAFLD and early fibrosis." Am J Physiol Gastrointest Liver Physiol 301(5): G865-876. Tanaka, T., C. N. Roy, W. Yao, A. Matteini, R. D. Semba, D. Arking, J. D. Walston, L. P. Fried, A. Singleton, J. Guralnik, G. R. Abecasis, S. Bandinelli, D. L. Longo and L. Ferrucci (2010). "A genome-wide association analysis of serum iron concentrations." Blood 115(1): 94-96. Tanno, T., N. V. Bhanu, P. A. Oneal, S. H. Goh, P. Staker, Y. T. Lee, J. W. Moroney, C. H. Reed, N. L. Luban, R. H. Wang, T. E. Eling, R. Childs, T. Ganz, S. F. Leitman, S. Fucharoen and J. L. Miller (2007). "High levels of GDF15 in thalassemia suppress expression of the iron regulatory protein hepcidin." Nat Med 13(9): 1096-1101. Tanno, T., P. Porayette, O. Sripichai, S. J. Noh, C. Byrnes, A. Bhupatiraju, Y. T. Lee, J. B. Goodnough, O. Harandi, T. Ganz, R. F. Paulson and J. L. Miller (2009). "Identification of

Page 196: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

181

TWSG1 as a second novel erythroid regulator of hepcidin expression in murine and human cells." Blood 114(1): 181-186. Taylor, K. M., H. E. Morgan, A. Johnson and R. I. Nicholson (2005). "Structure-function analysis of a novel member of the LIV-1 subfamily of zinc transporters, ZIP14." FEBS Lett 579(2): 427-432. Theisen, M., R. R. Behringer, G. G. Cadd, R. L. Brinster and G. S. McKnight (1993). "A C/EBP-binding site in the transferrin promoter is essential for expression in the liver but not the brain of transgenic mice." Mol Cell Biol 13(12): 7666-7676. Tomatsu, S., K. O. Orii, R. E. Fleming, C. C. Holden, A. Waheed, R. S. Britton, M. A. Gutierrez, S. Velez-Castrillon, B. R. Bacon and W. S. Sly (2003). "Contribution of the H63D mutation in HFE to murine hereditary hemochromatosis." Proc Natl Acad Sci U S A 100(26): 15788-15793. Trinder, D., O. Zak and P. Aisen (1996). "Transferrin receptor-independent uptake of differic transferrin by human hepatoma cells with antisense inhibition of receptor expression." Hepatology 23(6): 1512-1520. Trinder, D. and E. Morgan (1998). "Mechanisms of ferric citrate uptake by human hepatoma cells." Am J Physiol 275(2 Pt 1): G279-286. Trinder, D., J. K. Olynyk, W. S. Sly and E. H. Morgan (2002). "Iron uptake from plasma transferrin by the duodenum is impaired in the Hfe knockout mouse." Proc Natl Acad Sci U S A 99(8): 5622-5626. Trinder, D., C. Fox, G. Vautier and J. K. Olynyk (2002). "Molecular pathogenesis of iron overload." Gut 51(2): 290-295. Troadec, M. B., D. M. Ward, E. Lo, J. Kaplan and I. De Domenico (2010). "Induction of FPN1 transcription by MTF-1 reveals a role for ferroportin in transition metal efflux." Blood 116(22): 4657-4664. Truty, J., R. Malpe and M. C. Linder (2001). "Iron prevents ferritin turnover in hepatic cells." J Biol Chem 276(52): 48775-48780. Turi, J. L., X. Wang, A. T. McKie, E. Nozik-Grayck, L. B. Mamo, K. Crissman, C. A. Piantadosi and A. J. Ghio (2006). "Duodenal cytochrome b: a novel ferrireductase in airway epithelial cells." Am J Physiol Lung Cell Mol Physiol 291(2): L272-280. Valore, E. V. and T. Ganz (2008). "Posttranslational processing of hepcidin in human hepatocytes is mediated by the prohormone convertase furin." Blood Cells Mol Dis 40(1): 132-138. Vecchi, C., G. Montosi, K. Zhang, I. Lamberti, S. A. Duncan, R. J. Kaufman and A. Pietrangelo (2009). "ER stress controls iron metabolism through induction of hepcidin." Science 325(5942): 877-880. Verga Falzacappa, M. V., M. Vujic Spasic, R. Kessler, J. Stolte, M. W. Hentze and M. U. Muckenthaler (2007). "STAT3 mediates hepatic hepcidin expression and its inflammatory stimulation." Blood 109(1): 353-358.

Page 197: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

182

Videla, L. A., V. Fernandez, G. Tapia and P. Varela (2003). "Oxidative stress-mediated hepatotoxicity of iron and copper: role of Kupffer cells." Biometals 16(1): 103-111. Vokurka, M., J. Krijt, K. Sulc and E. Necas (2006). "Hepcidin mRNA levels in mouse liver respond to inhibition of erythropoiesis." Physiol Res 55(6): 667-674. Wallace, D. F., L. Summerville, P. E. Lusby and V. N. Subramaniam (2005). "First phenotypic description of transferrin receptor 2 knockout mouse, and the role of hepcidin." Gut 54(7): 980-986. Wallace, D. F., L. Summerville, E. M. Crampton, D. M. Frazer, G. J. Anderson and V. N. Subramaniam (2009). "Combined deletion of Hfe and transferrin receptor 2 in mice leads to marked dysregulation of hepcidin and iron overload." Hepatology 50(6): 1992-2000. Wallace, D. F., C. J. McDonald, L. Ostini and V. N. Subramaniam (2011). "Blunted hepcidin response to inflammation in the absence of Hfe and transferrin receptor 2." Blood 117(10): 2960-2966. Wallander, M. L., E. A. Leibold and R. S. Eisenstein (2006). "Molecular control of vertebrate iron homeostasis by iron regulatory proteins." Biochim Biophys Acta 1763(7): 668-689. Wang, C. Y. and M. D. Knutson (2013). "Hepatocyte divalent metal-ion transporter-1 is dispensable for hepatic iron accumulation and non-transferrin-bound iron uptake in mice." Hepatology. Wang, J., G. Chen and K. Pantopoulos (2005). "Nitric oxide inhibits the degradation of IRP2." Mol Cell Biol 25(4): 1347-1353. Wang, L., E. E. Johnson, H. N. Shi, W. A. Walker, M. Wessling-Resnick and B. J. Cherayil (2008). "Attenuated inflammatory responses in hemochromatosis reveal a role for iron in the regulation of macrophage cytokine translation." J Immunol 181(4): 2723-2731. Wang, R. H., C. Li, X. Xu, Y. Zheng, C. Xiao, P. Zerfas, S. Cooperman, M. Eckhaus, T. Rouault, L. Mishra and C. X. Deng (2005). "A role of SMAD4 in iron metabolism through the positive regulation of hepcidin expression." Cell Metab 2(6): 399-409. Weiss, G., T. Houston, S. Kastner, K. Johrer, K. Grunewald and J. H. Brock (1997). "Regulation of cellular iron metabolism by erythropoietin: activation of iron-regulatory protein and upregulation of transferrin receptor expression in erythroid cells." Blood 89(2): 680-687. Weiss, G. and L. T. Goodnough (2005). "Anemia of chronic disease." N Engl J Med 352(10): 1011-1023. West, A. P., Jr., M. J. Bennett, V. M. Sellers, N. C. Andrews, C. A. Enns and P. J. Bjorkman (2000). "Comparison of the interactions of transferrin receptor and transferrin receptor 2 with transferrin and the hereditary hemochromatosis protein HFE." J Biol Chem 275(49): 38135-38138. West, A. P., Jr., A. M. Giannetti, A. B. Herr, M. J. Bennett, J. S. Nangiana, J. R. Pierce, L. P. Weiner, P. M. Snow and P. J. Bjorkman (2001). "Mutational analysis of the transferrin

Page 198: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

183

receptor reveals overlapping HFE and transferrin binding sites." J Mol Biol 313(2): 385-397. Wright, T. L., P. Brissot, W. L. Ma and R. A. Weisiger (1986). "Characterization of non-transferrin-bound iron clearance by rat liver." J Biol Chem 261(23): 10909-10914. Wrighting, D. M. and N. C. Andrews (2006). "Interleukin-6 induces hepcidin expression through STAT3." Blood 108(9): 3204-3209. Xia, Y., J. L. Babitt, Y. Sidis, R. T. Chung and H. Y. Lin (2008). "Hemojuvelin regulates hepcidin expression via a selective subset of BMP ligands and receptors independently of neogenin." Blood 111(10): 5195-5204. Xiong, S., H. She, H. Takeuchi, B. Han, J. F. Engelhardt, C. H. Barton, E. Zandi, C. Giulivi and H. Tsukamoto (2003). "Signaling role of intracellular iron in NF-kappaB activation." J Biol Chem 278(20): 17646-17654. Xiong, S., H. She, C. K. Sung and H. Tsukamoto (2003). "Iron-dependent activation of NF-kappaB in Kupffer cells: a priming mechanism for alcoholic liver disease." Alcohol 30(2): 107-113. Yamaji, S., P. Sharp, B. Ramesh and S. K. Srai (2004). "Inhibition of iron transport across human intestinal epithelial cells by hepcidin." Blood 104(7): 2178-2180. Yang, F., X. B. Liu, M. Quinones, P. C. Melby, A. Ghio and D. J. Haile (2002). "Regulation of reticuloendothelial iron transporter MTP1 (Slc11a3) by inflammation." J Biol Chem 277(42): 39786-39791. Yang, J., D. Goetz, J. Y. Li, W. Wang, K. Mori, D. Setlik, T. Du, H. Erdjument-Bromage, P. Tempst, R. Strong and J. Barasch (2002). "An iron delivery pathway mediated by a lipocalin." Mol Cell 10(5): 1045-1056. Yeh, K. Y., M. Yeh and J. Glass (2004). "Hepcidin regulation of ferroportin 1 expression in the liver and intestine of the rat." Am J Physiol Gastrointest Liver Physiol 286(3): G385-394. Young, S. P., A. Bomford and R. Williams (1984). "The effect of the iron saturation of transferrin on its binding and uptake by rabbit reticulocytes." Biochem J 219(2): 505-510. Zakin, M. M. (1992). "Regulation of transferrin gene expression." Faseb J 6(14): 3253-3258. Zetterstrom, M., A. K. Sundgren-Andersson, P. Ostlund and T. Bartfai (1998). "Delineation of the proinflammatory cytokine cascade in fever induction." Ann N Y Acad Sci 856: 48-52. Zhang, D., E. Meyron-Holtz and T. A. Rouault (2007). "Renal iron metabolism: transferrin iron delivery and the role of iron regulatory proteins." J Am Soc Nephrol 18(2): 401-406. Zhang, D. L., T. Senecal, M. C. Ghosh, H. Ollivierre-Wilson, T. Tu and T. A. Rouault (2011). "Hepcidin regulates ferroportin expression and intracellular iron homeostasis of erythroblasts." Blood 118(10): 2868-2877.

Page 199: BSc. (Hons)IRON TRANSPORT AND LIVER INJURY IN MOUSE MODELS OF HEREDITARY HAEMOCHROMATOSIS Roheeth D. Delima BSc. (Hons) This thesis is presented for the degree of Doctor of Philosophy

184

Zhang, Y., M. Mikhael, D. Xu, Y. Li, S. Soe-Lin, B. Ning, W. Li, G. Nie, Y. Zhao and P. Ponka (2010). "Lysosomal proteolysis is the primary degradation pathway for cytosolic ferritin and cytosolic ferritin degradation is necessary for iron exit." Antioxid Redox Signal 13(7): 999-1009. Zhao, M., F. Antunes, J. W. Eaton and U. T. Brunk (2003). "Lysosomal enzymes promote mitochondrial oxidant production, cytochrome c release and apoptosis." Eur J Biochem 270(18): 3778-3786. Zhao, N., J. Gao, C. A. Enns and M. D. Knutson (2010). "ZRT/IRT-like protein 14 (ZIP14) promotes the cellular assimilation of iron from transferrin." J Biol Chem 285(42): 32141-32150. Zheng, D., G. P. Feeney, P. Kille and C. Hogstrand (2008). "Regulation of ZIP and ZnT zinc transporters in zebrafish gill: zinc repression of ZIP10 transcription by an intronic MRE cluster." Physiol Genomics 34(2): 205-214. Zhou, X. Y., S. Tomatsu, R. E. Fleming, S. Parkkila, A. Waheed, J. Jiang, Y. Fei, E. M. Brunt, D. A. Ruddy, C. E. Prass, R. C. Schatzman, R. O'Neill, R. S. Britton, B. R. Bacon and W. S. Sly (1998). "HFE gene knockout produces mouse model of hereditary hemochromatosis." Proc Natl Acad Sci USA 95(5): 2492-2497. Zohn, I. E., I. De Domenico, A. Pollock, D. M. Ward, J. F. Goodman, X. Liang, A. J. Sanchez, L. Niswander and J. Kaplan (2007). "The flatiron mutation in mouse ferroportin acts as a dominant negative to cause ferroportin disease." Blood 109(10): 4174-4180.