© 2016 Richard Coffey - University of...

155
MECHANISMS OF NON-TRANSFERRIN-BOUND IRON UPTAKE BY HUMAN β CELLS AND THE ROLE OF IRON IN DIABETIC PATHOGENESIS By RICHARD COFFEY A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2016

Transcript of © 2016 Richard Coffey - University of...

Page 1: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

MECHANISMS OF NON-TRANSFERRIN-BOUND IRON UPTAKE BY HUMAN β CELLS AND THE ROLE OF IRON IN DIABETIC PATHOGENESIS

By

RICHARD COFFEY

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2016

Page 2: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

© 2016 Richard Coffey

Page 3: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

To my parents

Page 4: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

4

ACKNOWLEDGMENTS

I would first and foremost like to thank my major advisor Dr. Mitchell Knutson

who took a chance and gave me the opportunity to do research all those years ago. I

would not be where I am right now if I did not end up in your undergraduate lab

techniques class.

Also, I would like to thank my past and current labmates Chia-Yu Wang, Supak

Jenkitkasemwong, Wei Zhang, Hyeyoung Nam, Lin Zhang, Katie Sullivan, Lizzie

Paulus, Laura Diez-Ricote, Nike Akinyode, and Emily Mejia who have either taught me

most of what I know about lab work today or have helped me in innumerable other

ways. I wish you the best wherever you find yourself in life and know that you will be

successful.

I would also like to thank my committee members Dr. Clayton Mathews, who has

been instrumental in the design and execution of several experiments discussed in this

dissertation, Dr. James Collins, and Dr. Michelle Gumz who have all taken time out of

their busy lives to help me during this process.

Finally I would like to thank my parents who have always given me the

opportunity to pursue my interests and have been unconditionally supportive of my

decisions.

Page 5: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

5

TABLE OF CONTENTS

page

ACKNOWLEDGMENTS .................................................................................................. 4

LIST OF TABLES ............................................................................................................ 7

LIST OF FIGURES .......................................................................................................... 8

LIST OF ABBREVIATIONS ........................................................................................... 10

ABSTRACT ................................................................................................................... 13

CHAPTER

1 INTRODUCTION .................................................................................................... 15

2 LITERATURE REVIEW .......................................................................................... 17

Basics of Iron Metabolism ....................................................................................... 17 General Information .......................................................................................... 17

Dietary Iron Absorption ..................................................................................... 18 Disorders of Iron Metabolism ............................................................................ 21

Non-Transferrin-Bound Iron Import Proteins .................................................... 25 The Iron and Diabetes Connection ......................................................................... 34

Elevated Iron Stores and Diabetic Pathology ................................................... 34 Pancreatic Iron Accumulation ........................................................................... 39

Mechanistic Evidence from Human Studies ..................................................... 43 Mechanistic Evidence from Animal Studies ...................................................... 47

Iron and Autoimmune Diabetes ........................................................................ 49

3 MATERIALS AND METHODS ................................................................................ 53

Animals and Diets ............................................................................................ 53 Iron Status Parameters and Blood Glucose Concentrations ............................ 54

Pancreatic Mineral Concentrations ................................................................... 55 Histological Analysis ......................................................................................... 55

RNA Isolation and Assessment of RNA Integrity .............................................. 55 Microarray Analysis .......................................................................................... 56

Relative mRNA Quantification .......................................................................... 56 Western Blotting ............................................................................................... 57

Cell Culture and Treatments ............................................................................. 58 In Vitro Glucose Stimulated Insulin Secretion ................................................... 59

Mouse Islet Isolation ......................................................................................... 60 Determination of DMT1, ZIP8, and ZIP14 mRNA Copy Numbers .................... 60

siRNA Knockdown of DMT1, ZIP8, and ZIP14 ................................................. 61

Page 6: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

6

Overexpression of DMT1, ZIP8, and ZIP14 ...................................................... 61 Immunofluorescencse ...................................................................................... 62

Cellular NTBI Uptake ........................................................................................ 63 Generation of Transgenic MIP-Zip14-HA Mice ................................................. 64

Statistical Analysis ............................................................................................ 65

4 TRANSCRIPTIONAL PROFILING OF PANCREATIC GENE EXPRESSION IN RESPONSE TO DIETARY IRON LOADING OR DEFICIENCY ............................. 66

Results .................................................................................................................... 67

Body Weight, Iron Status, and Blood Glucose Concentrations ......................... 67 Pancreatic Mineral Concentrations ................................................................... 67

Identification and Classification of Differentially Expressed Genes by Microarray Analysis ....................................................................................... 68

Confirmation of Up-Regulation of Alox15 Expression by QRT-PCR and Western Blotting ............................................................................................ 69

Confirmation of Reg Family mRNA Levels by QRT-PCR ................................. 70 Discrepancies Between Microarray and QRT-PCR Analysis Results ............... 70

Discussion .............................................................................................................. 71

5 MECHANISMS OF NTBI UPTAKE BY HUMAN β CELLS ...................................... 86

Results .................................................................................................................... 87 Overexpression of NTBI Transporters in Human β Cells .................................. 87

siRNA Knockdown of NTBI Transporters in Human β Cells ............................. 88 Cellular Localization of NTBI Transporters in Human Islets ............................. 89

Modulation of ZIP14 Expression by Iron in Human β Cells .............................. 89 Modulation of ZIP14 Expression By IL-1β in Human β Cells ............................ 90

Discussion .............................................................................................................. 90

6 THE INFLUENCE OF IRON STATUS ON DIABETIC PATHOLOGY AND β-CELL FUNCTION ................................................................................................. 103

Results .................................................................................................................. 105

Effect of Iron Status on Spontaneous Autoimmune Diabetes in NOD Mice.... 105 Effect of Dietary Iron on Rate of Growth and Systemic Iron Status ................ 105

Pancreatic Mineral Concentrations ................................................................. 107 Testing of β cell function During the Prediabetic Period ................................. 107

Effect of Iron Status on Human Islet GSIS ..................................................... 108 Generation of Mice Selectively Overexpressing Zip14 in β Cells ................... 108

Discussion ............................................................................................................ 109

7 CONCLUSIONS, LIMITATIONS, AND FUTURE DIRECTIONS ........................... 124

LIST OF REFERENCES ............................................................................................. 131

BIOGRAPHICAL SKETCH .......................................................................................... 155

Page 7: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

7

LIST OF TABLES

Table page 4-1 Body weight, iron status, and blood glucose concentration of rats ..................... 75

4-2 Pancreatic mineral concentrations ...................................................................... 76

4-3 Top 10 most up-regulated and down-regulated genes in FeD pancreata ........... 77

4-4 Top 10 most up-regulated and down-regulated genes in FeO pancreata ........... 78

4-5 Primers for qRT-PCR ......................................................................................... 79

4-6 Functional categories of pancreatic genes differentially expressed in response to iron deficiency ................................................................................. 80

4-7 Functional categories of pancreatic genes differentially expressed in response to iron overload ................................................................................... 81

6-1 Iron parameters of type 1 diabetes-prone NOD mice ....................................... 115

6-2 Pancreatic mineral concentrations in NOD mice .............................................. 116

Page 8: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

8

LIST OF FIGURES

Figure page 4-1 Functional classification of pancreatic genes up- or down-regulated in

response to iron deficiency and iron overload.. .................................................. 83

4-2 Effect of iron deficiency and overload on rat pancreatic Alox15 expression.. ..... 84

4-3 Effect of iron deficiency and overload on the expression of pancreatic Reg family genes. ...................................................................................................... 85

5-1 ZIP14 and ZIP8, but not DMT1, overexpression increases iron uptake by βlox5 cells ........................................................................................................... 95

5-2 When overexpressed in βlox5 cells, ZIP14 localizes to the plasma membrane whereas DMT1 mainly localizes intracellularly. .................................................. 96

5-3 Endogenous iron uptake by βlox5 cells is decreased by siRNA knockdown of ZIP14, but not ZIP8. ........................................................................................... 97

5-4 siRNA knockdown of ZIP14 decreases NTBI uptake by primary human islets ... 98

5-5 ZIP14 is detected in human pancreatic β cells by immunofluorescent analysis. ............................................................................................................. 99

5-6 Cellular iron levels and treatment with IL-1β increase ZIP14 levels in βlox5 cells but not primary human islets.. .................................................................. 100

5-7 mRNA copy numbers of NTBI transporters in primary human islets. qRT-PCR measurement of DMT1, ZIP14, and ZIP8 mRNA copy numbers in total RNA isolated from nondiabetic human islets. ................................................... 101

5-8 DMT1, but not ZIP8, is detected in human β cells by immunoflourescence staining.. ........................................................................................................... 102

6-1 Dietary iron deficiency, but not iron overload, results in a trend towards an increased incidence of spontaneous diabetes in female NOD mice.. ............... 117

6-2 NOD mice fed an iron-loaded diet initially experience diminished growth. ....... 118

6-3 Iron stores of mice fed an iron-deficient diet increase with age.. ...................... 119

6-4 Glucose tolerance and insulin secretory capacity is not affected by iron status in prediabetic NOD mice.. ................................................................................. 120

6-5 Iron-deficient prediabetic NOD mice show no differences in β cell iron status or insulitis compared with iron-adequate mice.. ................................................ 121

Page 9: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

9

6-6 Iron status does not affect glucose-stimulated insulin secretion by human islets in vitro.. .................................................................................................... 122

6-7 Generation of mice selectively overexpressing Zip14 in β cells.. ...................... 123

Page 10: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

10

LIST OF ABBREVIATIONS

Alox15 Arachidonate 15-lipoxygenase

AMPK 5’ adenosine monophosphate-activated protein kinase

Apoa1 Apolipoprotein A-1

BMI Body mass index

CCS Copper chaperone for superoxide dismutase

CRP C-reactive protein

CS Cell surface

DAVID Database for annotations, visualization, and integrated discovery

DCT1 Divalent cation transporter 1

DcytB Duodenal cytochrome B

DFO Deferoximine mesylate

DMT1 Divalent metal-ion transporter 1

ELISA Enzyme-linked immunosorbent assay

EV Empty vector

Fabp1 Fatty acid binding protein 1

Fabp2 Fatty acid binding protein 2

FAC Ferric ammonium citrate

FeA Iron adequate

FeD Iron deficient

Fe-NTA Ferric nitrilotriacetate

FeO Iron loaded

GFP Green fluorescent protein

GSIS Glucose-stimulated insulin secretion

HA Hemagglutinin antigen tag

Page 11: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

11

HAMP Hepcidin antimicrobial peptide

HETE Hydroxyeicosatetraenoic acid

hGH Human growth hormone intron region

HJV Hemojuvelin

ICP-MS Inductively coupled plasma mass spectrometry

IgG Immunoglobulin G

IL-1β Interleukin 1β

IP-GTT Intraperitoneal glucose tolerance testing

IRE Iron response element

KRB Kreb’s-Ringer Buffer

LPS Lipopolysaccharide

LTCC L-type calcium channel

MIP Mouse insulin 1 promoter

Mn-SOD Manganese superoxide dismutase

Na+/K+ ATPase Sodium-potassium adenosine triphosphatase

NOD Non-obese diabetic

NRAMP2 Natural-resistance associated macrophage protein 2

NTBI Non-transferrin bound iron

PPM Parts per million

qRT-PCR Quantitative reverse transcriptase polymerase chain reaction

Reg Rengenerating islet-derived gene

ROS Reactive oxygen speices

siRNA Small interfering ribonucleic acid

SLC11a2 Solute carrier family 11 member 2

SLC39a14 Solute transporter family 39 member 14

Page 12: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

12

SLC39a8 Solute transporter family 39 member 8

STEAP3 Six-Transmembrane epithelial antigen of prostate family member 3

sTFR Soluble transferrin receptor

TBI Transferrin bound iron

TCL Total cell lysate

TF Transferrin

TF sat Transferrin saturation

TFR Transferrin receptor

TFR1 Transferrin receptor 1

TFR2 Transferrin receptor 2

Tg Transgenic

TIBC Total iron binding capacity

TTCC T-type calcium channel

UTR Untranslated region

Wt Wild type

ZIP14 ZRT/IRT-Like protein 14

ZIP8 ZRT/IRT-Like protein 8

Page 13: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

13

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

MECHANISMS OF NON-TRANSFERRIN-BOUND IRON UPTAKE BY HUMAN β

CELLS AND THE ROLE OF IRON IN DIABETIC PATHOGENESIS

By

Richard Coffey

May 2016

Chair: Mitchell D. Knutson Major: Nutritional Sciences

The relationship between iron and diabetes has long been recognized as

individuals with iron overload display an increased prevalence of diabetes and iron

depletion is thought to protect against the development of diabetes. This link is

attributed to the accumulation of iron in beta cells, which may impair cellular function.

However, the mechanisms by which beta cells take up iron, as well as the specifics of

how iron status affects diabetic pathogenesis, are undetermined. During iron overload

plasma iron levels exceed the carrying capacity of transferrin resulting in non-transferrin

bound iron (NTBI), which is rapidly taken up by tissues. Currently 3 mammalian proteins

which transport NTBI have been identified: Divalent metal-ion transporter 1 (DMT1),

ZRT/IRT-Like transporter 14 (ZIP14), and ZRT/IRT-Like transporter 8 (ZIP8). The aims

of this project were to determine the contribution of DMT1, ZIP14, and ZIP8 to iron

uptake by human beta cells and to investigate the influence of iron status on various

aspects of diabetic pathology. I found that overexpression of ZIP14 and ZIP8, but not

DMT1, resulted in increased iron uptake by Betalox5 cells, a human beta-cell line.

siRNA-mediated knockdown of ZIP14, but not ZIP8, resulted in 50% lower iron uptake

in Betalox5 cells. In primary human islets, knockdown of ZIP14 also reduced iron

Page 14: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

14

uptake by 50%. Immunofluorescence analysis of human pancreatic sections localized

ZIP14 and DMT1, but not ZIP8, to beta cells. To determine how iron status may affect

diabetic pathology I examined pancreatic gene expression in iron-deficient, iron-

adequate, and iron-loaded rats. Iron overload and deficiency were associated with

increased pancreatic expression of genes associated with pancreatic stress and linked

to the development of autoimmune diabetes. Additionally, non-obese diabetic mice fed

an iron-deficient, but not iron-loaded, diet trended towards an increased incidence of

diabetes compared with iron-adequate mice. However, this trend was not associated

with a reduction in beta-cell function during the prediabetic period. Iron depletion or

loading of isolated human islets also had no effect on beta-cell function. Overall, results

from these studies indicate that ZIP14 contributes to beta-cell NTBI uptake and suggest

that iron deficiency may not protect against the development of diabetes.

Page 15: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

15

CHAPTER 1 INTRODUCTION

Iron is an essential trace mineral that is necessary for many biological functions

including, but not limited to, mitochondrial respiration (1), hemoglobin production (2),

drug metabolism (3), and DNA synthesis (4). Disruptions of normal iron homeostasis,

due to either iron deficiency or iron overload, are some of the most common nutrition-

related issues worldwide. One complication of iron overload is an increased prevalence

of diabetes, as evidenced by humans with hemochromatosis or β-thalassemia major,

which result in systemic iron accumulation (5, 6).

Despite the well-documented association between excess iron and an increased

incidence of diabetes several questions remain unanswered. For example, little is

known regarding how pancreatic β cells take up iron, which is found in the plasma

bound to transferrin, under normal conditions, and as non-transferrin bound iron (NTBI)

during disorders of iron overload, when the carrying capacity of transferrin becomes

exceeded. NTBI is known to be a major contributor to iron loading of hepatocytes and

pancreatic acinar cells (7), but its contribution to iron loading of pancreatic β cells is not

well understood. Additionally, the details of how iron deficiency and overload may affect

the pathogenesis of diabetes have yet to be fully elucidated. The experiments described

in this dissertation sought to answer questions concerning the link between iron

metabolism and the pathogenesis of diabetes. Specifically, by determining the

mechanisms by which NTBI is taken up by pancreatic β cells and by evaluating the

impact of iron status on pancreatic gene expression and on aspects of diabetic

pathogenesis.

Page 16: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

16

To determine the molecular mechanisms of NTBI uptake by β cells I performed in

vitro experiments testing how altering the expression of established mammalian NTBI

transport proteins affects iron uptake by human β cells. Additionally I determined the

cellular localization of these transporters within the human pancreas to identify the cell

populations in which they may function. To evaluate the role of iron in the pathogenesis

of diabetes I investigated potential mechanisms by which iron status could affect the

development of diabetes by using microarray analysis to identify the altered expression

of genes previously linked to various aspects of diabetic pathogenesis or β cell function,

as described by other reports, in the pancreata of rats fed iron-deficient or -loaded diets.

To test the proposed relationship between iron status and the risk of developing type 1

diabetes I also determined the effect of systemic iron status on the development of

autoimmune diabetes in type 1-diabetes-prone NOD mice and measured the effect of

cellular iron status on insulin secretion by human islets in vitro. Lastly, I document the

generation of a novel transgenic mouse that overexpresses the iron transporter ZIP14,

which may provide a model to study β cell iron loading in vivo.

Page 17: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

17

CHAPTER 2 LITERATURE REVIEW

Basics of Iron Metabolism

General Information

Iron in biological systems has two main oxidation states, ferrous (Fe2+) or ferric

(Fe3+). These two oxidation states allow iron to readily exchange electrons and to

participate in oxidation-reduction reactions within biological systems. Iron is generally

categorized as either heme iron, in which iron is incorporated into a protoporphyrin ring

as found in hemoglobin and myoglobin, or non-heme iron, a broad term used to

describe all iron not incorporated into heme. The majority of dietary iron consumed is

non-heme iron (8), which is found in both plant and animal foods whereas heme iron is

only present in animal sources. Despite the greater bioavailablity of heme iron, the

majority of iron absorbed by the body is obtained from non-heme sources (8, 9), due to

the abundance of dietary non-heme iron. On average the human body contains 2.5 to 4

g of iron, for females and males respectively. The majority of body iron exists as

hemoglobin or myoglobin, which function in oxygen transport, or as stored iron within

ferritin, the iron-storage protein. The daily requirement for iron to support biological

functions, such as erythropoiesis and the production of other iron-containing proteins, is

approximately 24 mg/day. However, the majority of iron used daily is obtained from the

catabolism of senescent erythrocytes by splenic and hepatic macrophages, which can

then recycle the iron contained within erythrocytes for the production of new red blood

cells and iron-containing proteins by the bone marrow and systemic tissues. While this

process is efficient and provides approximately 90% of the daily iron requirement, minor

quantities of iron are lost through various routes including sweating and the sloughing of

Page 18: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

18

skin cells. Therefore, 1-2 mg of dietary iron is absorbed from the diet to offset daily

losses and maintain systemic iron homeostasis.

Dietary Iron Absorption

Iron metabolism in humans differs from that of other minerals, such as copper or

zinc, in that there is no major route of excretion by which substantial amounts of iron

can be eliminated from the body. Rodents are able to remove some excess iron through

biliary excretion, although this is insufficient to prevent iron overload (10). Therefore, the

absorption of iron from food sources is tightly regulated. The majority of dietary iron is

non-heme iron, which is solubilized at the acidic pH of gastric secretions and absorbed

in the proximal small intestine via Divalent Metal-ion Transporter 1 (DMT1), a

transmembrane protein that couples the transport of ferrous iron to a proton gradient

(11). DMT1 is indispensable for intestinal NTBI uptake as mice lacking DMT1 in the

intestine develop severe iron deficiency and display ablated iron absorption (12, 13). To

ensure that dietary iron exists as ferrous iron, the form which can be transported by

DMT1, ferric iron is thought to be reduced at the enterocyte brush border by the

reductase duodenal cytochrome B (Dcytb). However mice lacking Dcytb show no

difference in the uptake of radiolabeled ferric iron compared wild-type mice suggesting

that Dcytb is dispensable for the reduction of ferric iron in vivo (14). It is possible that

the absence of Dcytb can be compensated by the presence of unidentified brush-border

reductases or reducing agents, such as ascorbate either consumed in the diet or

secreted within digestive juices (15), to reduce iron within the gut lumen. After the

uptake of ferrous iron by the enterocyte iron can either be stored within the enterocyte

within ferritin, and eventually lost when the enterocyte is sloughed off into the gut lumen,

or exported into the portal circulation. If systemic iron stores are adequate or elevated,

Page 19: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

19

much of the iron taken up by enterocytes will be incorporated into the iron storage

protein ferritin, a multisubunit spherical protein with ferroxidase activity containing a core

composed of ferric iron. If systemic iron stores are low, or in response to stimuli

including anemia (16, 17) or hypoxia (18), enterocyte iron is exported into the portal

circulation by ferroportin, the only identified mammalian iron export protein. Ferroportin

is located on the enterocyte basolateral membrane and is essential for iron export as

mice selectively lacking intestinal ferroportin accumulate iron within enterocytes and

develop severe anemia (19). After export from the enterocyte, iron in the plasma binds

to the plasma iron transport protein transferrin (TF). However, ferrous iron exported

from enterocytes must be oxidized to the ferric state before binding to transferrin. The

oxidation of iron exported via ferroportin may be accomplished by the action of the

ferroxidase hephaestin, located at the basolateral membrane of the enterocyte. In mice

the loss of hephaestin function, due to genetic mutation (20) or genetic deletion (21),

results in decreased dietary iron absorption and iron accumulation in enterocytes

despite elevated levels of ferroportin (21, 22). Impaired iron transport by ferroportin in

response to the loss of functional hephaestin may be due to a necessary interaction

between ferroportin and hephaestin during iron export from the enterocyte. Hephaestin

and ferroportin have been reported to physically interact in rat enterocytes (23)

supporting this hypothesis. However, ferroportin is able to transport iron when

overexpressed in xenopus oocytes, independent of hephaestin overexpression,

suggesting that the interaction of ferroportin with hephaestin is not necessary for iron

export activity (24). Ferric iron circulating as TBI in the plasma is taken up by cells

through the binding of TF to transferrin receptor (TFR) on the cell surface, forming a

Page 20: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

20

complex which is then endocytosed. Iron is released from TF within the endosome by

endosomal acidification after which ferric iron is reduced to ferrous iron by STEAP3

(25), allowing for the export into the cytosol of ferrous iron via DMT1 and, potentially,

ZRT/IRT-like Protein 14 (ZIP14) and ZRT/IRT-like Protein 8 (ZIP8) (26-28). In the bone

marrow, the major site of transferrin-TFR iron acquisition, the export of endosomal iron

is accomplished by the action of DMT1, as evidenced by loss-of-function mutations in

DMT1 leading to impaired reticulocyte iron delivery by the endocytosed TF-TFR

complex (29) and microcytic anemia (26, 30).

In humans the majority of iron released into the circulation and delivered to

tissues by TF is provided by the release of iron by the macrophages of the

reticuloendothelial system and hepatocytes, with newly absorbed dietary iron being a

minor contributor to plasma iron under normal conditions (31). To ensure that

appropriate amounts of iron are provided for bodily functions, such as erythropoiesis,

the release of stored iron from macrophages and hepatocytes is tightly regulated. The

export of tissue iron stores into circulation is mediated by ferroportin (19, 32), similar to

the release of iron from enterocytes, allowing iron mobilization from body stores and

enterocytes to be regulated by a similar mechanism. The control of systemic iron

homeostasis centers on the regulation of ferroportin levels through the action of

hepcidin, a peptide hormone primarily produced by hepatocytes (33). Hepcidin binds to

ferroportin leading to endocytosis and subsequent degradation (34), thus preventing the

release of iron from cells. The expression of hepcidin is normally linked to systemic iron

status and decreases during iron deficiency (35) to increase dietary iron absorption and

the release of stored iron from macrophages and hepatocytes, while increasing during

Page 21: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

21

iron loading (36), to prevent excess dietary iron uptake and the release of stored iron.

The expression of hepcidin can also be regulated by factors including anemia, hypoxia,

and inflammatory stimuli (37).

Disorders of Iron Metabolism

While under normal conditions iron homeostasis is tightly regulated by hepcidin

to ensure adequate iron for cellular functions while preventing excess iron

accumulation, conditions can arise during which iron homeostasis becomes perturbed.

Dietary iron deficiency is the most common nutrient deficiency worldwide, with over 2

billion people estimated to be iron deficient (38). The greatest estimated incidence of

dietary iron deficiency is seen in infants, children (39), and pregnant women in

developing countries (38), but women of child-bearing age still suffer from iron

deficiency in industrialized countries. In the United States 10-15% of women of child-

bearing age are iron deficient (40). Iron deficiency in men, as well as post-menopausal

women, is less common in the United States (40). The negative effects of iron

deficiency include fatigue (41), impaired cognitive function (42), and pica (43).

On the opposite end of the spectrum from iron deficiency are disorders of iron

overload, characterized by iron accumulation in various organs resulting in iron-

mediated tissue damage. Iron overload disorders can most often be linked to genetic

mutations. One such disorder is hemochromatosis, a disease characterized by

excessive iron loading in the liver, pancreas, and heart, resulting in hepatic

fibrosis/cirrhosis (44), diabetes (45), and cardiomyopathy (46). The majority of

hemochromatosis cases result from a single point mutation in the hemochromatosis

gene, HFE (47) (48), and are referred to as type 1 hemochromatosis. HFE is involved in

plasma-iron sensing by hepatocytes and interacts with transferrin receptor 2 (TFR2) to

Page 22: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

22

regulate the hepatic production of hepcidin (48, 49). Homozygosity for this mutation

occurs at a frequency of 1/200 in individuals of northern European descent but has

incomplete penetrance (50). More severe and rapid iron accumulation is observed in

humans with juvenile hemochromatosis, or type 2 hemochromatosis, resulting from

mutations in the hemochromatosis type 2 gene (HFE2) (51), encoding Hemojuvelin

(HJV) involved in hepatocyte iron sensing (type 2A) (52), or in the hepcidin antimicrobial

peptide gene (HAMP) (53), encoding hepcidin itself (type 2B). Symptoms of iron

overload in juvenile hemochromatosis occur rapidly and are often detected in patients

with juvenile hemochromatosis before 30 years of age (54), earlier than type 1

hemochromatosis, which is usually diagnosed in middle-aged patients (55). The

majority of juvenile hemochromatosis cases are due to mutations in HFE2, with HAMP

mutations being less commonly documented (56). Type-3 hemochromatosis due to

mutations in the gene transferrin receptor 2 (TFR2) (57), another protein involved in

hepatocyte iron sensing (48), has also been documented to produce iron overload of an

intermediate degree, with iron symptoms of iron overload appearing before those seen

in with type 1 but later than type 2 hemochromatosis (58, 59).While these mutations

occur in various genes, including HFE, HFE2, TFR2, and HAMP, the end result is a

deficiency of hepcidin production by hepatocytes, resulting in the impaired down-

regulation of ferroportin and increased dietary iron absorption.

A unique form of iron overload linked to mutations in ferroportin, rather than a

dysregulation of hepcidin, is referred to as ferroportin disease or type-4

hemochromatosis. Mutations that inhibit the ability of hepcidin to bind ferroportin result

in a phenotype similar to other forms of hemochromatosis (60, 61), in which ferroportin

Page 23: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

23

expression is elevated, characterized by elevated dietary iron absorption, macrophage

iron export, transferrin saturation, and liver iron accumulation. While gain-of-function

mutations in ferroportin lead to iron overload, paradoxically, loss-of-function mutations in

ferroportin have also been reported to result in iron accumulation. Mutations that impair

the targeting of ferroportin to the plasma membrane (62) have been reported in humans

with ferroportin disease, characterized by iron accumulation in macrophages, elevated

serum ferritin prior to elevated transferrin saturation, and liver iron deposition (63).

Currently it remains to be clarified how a loss-of-function mutation in ferroportin can

simultaneously result in the loss of ferroportin activity in iron export by macrophages

while allowing for dietary iron uptake and the establishment of elevated liver iron levels.

Studies of intestinal iron absorption in humans or animals with loss-of-function

ferroportin mutations are needed.

Another genetic disorder that results in iron overload is β-thalassemia major,

caused by mutations in the β globin gene (64) leading to impaired hemoglobin

production. Treatment for β-thalassemia major involves regular blood transfusions that

produce transfusional iron overload. Additionally, the failure to produce adequate

hemoglobin results in persistent anemia that can suppress hepcidin production in

response to iron overload, (65, 66), resulting in elevated intestinal iron absorption

exacerbating transfusional iron loading (67). While patients with β-thalassemia major

are usually treated with iron chelators, iron overload still occurs characterized by severe

iron accumulation in peripheral tissues, often associated with the development of

endocrine complications and heart failure (68).

Page 24: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

24

Iron overload resulting from high dietary iron is far less common than

hemochromatosis or β-thalassemia major and is best documented in individuals living in

rural Sub-Saharan Africa. In this region, the practice of brewing beer in metal containers

results in iron leaching into the beverage, which is often heavily consumed (69). Iron

loading in response to this highly bioavailable iron affects some individuals to a greater

degree than others indicating that there may be a genetic predisposition to iron

accumulation (70). Mutations in ferroportin, similar to that observed in ferroportin

disease, have been associated with African iron overload suggesting that this may

explain the susceptibility of some individuals to dietary iron loading (71). However, other

reports indicate that the presence of this mutation is not associated with markers of iron

overload in African families with previously identified iron overload (72). The interplay

between dietary iron consumption and genetic susceptibility to iron overload has yet to

be elucidated regarding African iron overload.

While many conditions lead to iron overload, a common phenotype of excess iron

deposition in peripheral tissues, to varying degrees, is observed in response to systemic

iron accumulation. During severe iron overload the amount of plasma iron exceeds the

binding capacity of TF leading to the appearance of non-transferrin bound iron (NTBI).

NTBI is cleared rapidly from the circulation by tissues, leading to iron deposition in

organs such as the liver, pancreas, and heart (73, 74), potentially leading to tissue

damage and dysfunction (60). While the deposition of iron in these tissues during iron

overload is well established, the mechanisms by which NTBI is initially taken up remain

to be elucidated for many cell types.

Page 25: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

25

Non-Transferrin-Bound Iron Import Proteins

The study of cellular iron transport in mammals has in large part centered on the

action of transmembrane proteins that demonstrate the ability to transport free iron. The

first discovered mammalian iron transport protein identified was DMT1, formerly referred

to as divalent cation transporter 1 (DCT1) or natural resistance-associated macrophage

protein 2 (NRAMP2), a transmembrane protein encoded by the solute carrier family 11,

member 2 gene (SLC11A2) (11). DMT1 was originally identified by screening a cDNA

library from iron-deficient rat duodenum for iron transport activity (11). DMT1 expression

was also found to be strongly induced in the duodenum of rats fed a low-iron diet, a

treatment which greatly induces intestinal iron uptake. Intestinal expression of DMT1 is

most abundant in the proximal small intestine, which has an acidic microenvironment

near the brush border due to the presence of gastric secretions. In line with this

localization, DMT1 functions optimally at an acidic pH with iron transport ability

substantially decreasing, displaying only residual activity, at physiologic pH (75). The

pH-dependent nature of DMT1-mediated NTBI transport is due to the coupling of iron

transport with protons, necessitating a low pH for efficient activity. Since the initial

discovery and characterization of DMT1, further studies have reported multiple isoforms

of human DMT1 that differ at both the N and C-terminal regions, allowing for 4 isoforms.

At the N terminus DMT1 isoforms can be identified as either 1A or 1B, differentiated by

an additional amino acid sequence present on the 1A isoform proceeding the shared

sequence by both 1A and 1B isoforms (76). 1A/1B isoforms of DMT1 display differential

expression patterns, with 1B isoforms being expressed to some degree in all tissues

examined whereas 1A-DMT1 expression is restricted to the duodenum and kidney (77).

Isoforms of DMT1 differing in the C-terminal region can be classified as those translated

Page 26: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

26

from mRNA sequences with or without an iron response element (IRE), identified as

+IRE or –IRE isoforms. 3’ iron response elements allow for posttranscriptional

regulation of mRNA levels, increasing mRNA stability during iron deficiency (78). While

the IREs are located in the 3’UTR, DMT1 translated from –IRE transcripts differs from

that translated from +IRE due to the substitution of the terminal 18 amino acids with a

different 25 amino acid sequence (79). Tissue characterization of DMT1 expression

between +IRE and –IRE isoforms indicate that most tissues examined express both

isoforms, with the exception of the liver, testis, and duodenum in which +IRE isoforms

were more abundant (77). Differences in the C-terminal region between DMT1 isoforms

have been documented to affect the intracellular targeting of DMT1 in a cell-type

specific manner (76, 80). However, intracellular targeting attributed to differences

between isoforms at the N-terminal region have yet to be reported. The isoform of

DMT1 that functions in the duodenum to upregulate iron uptake in response to iron

deficiency is likely DMT1+IRE, as the induction of DMT1+IRE, but not –IRE, has been

reported in the iron deficient mouse intestine (81), and 1A but not 1B isoforms, of DMT1

are regulated by iron status in Caco2 cells, an intestinal epithelial cell line (77).

While the role of DMT1 in intestinal iron uptake and the export of iron from

endosomes into the cytosol within developing erythrocytes is well established and

previously discussed, the contribution of DMT1 to NTBI uptake by other cell types

during iron overload is unclear. The initial characterization of DMT1 in rats reported low

level mRNA expression of Dmt1 in the liver, pancreas, and heart, relative to the level of

Dmt1 expressed in the kidney or intestine (11), suggesting that NTBI uptake via DMT1

may be limited in these tissues. DMT1 has been detected in rat liver at the protein level;

Page 27: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

27

however, hepatic DMT1 protein levels are strongly reduced in response to liver iron

loading (82), suggesting that this pathway of NTBI uptake is unlikely to promote hepatic

iron accumulation. A study of mice selectively lacking DMT1 in hepatocytes also argues

against a role for DMT1 in NTBI uptake by the liver, as DMT1 was found to be

dispensable for hepatic NTBI uptake and hepatocyte iron accumulation in a mouse

model of hemochromatosis (83). Cardiac NTBI uptake through the action of DMT1 also

appears unlikely as Dmt1 expression in the heart is observed to decrease in response

to cardiac iron loading, similar to the trend observed in the liver (82). However,

mechanistic studies regarding the role that DMT1 plays in cardiac NTBI uptake have yet

to be carried out.

Unlike DMT1 expression in the liver or heart, DMT1 expression is reportedly

unchanged in response to pancreatic iron accumulation in rats (82). Acinar cells

comprise the majority of the pancreas and therefore the unaltered DMT1 expression

during iron loading likely reflects the state of this cell population. However, pancreatic

iron loading is often viewed in the context of the pathogenesis of diabetes, requiring the

study of pancreatic islets which constitute 1-2% of the pancreas. Due to the small

contribution of islets to overall pancreatic mass, techniques that measure whole-tissue

expression will be unable to draw accurate conclusions about islet gene expression as

mRNA or protein from islets will be diluted by that of acinar cells. Techniques that are

able to discern cell-type-specific changes in gene expression have reported that the

expression of Dmt1 within pancreatic islets in mice injected with iron decreases (84).

Additionally, the pattern of DMT1 expression within the cell types of the pancreas is

disputed in the literature. In humans pancreatic DMT1 is reported to be primarily

Page 28: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

28

restricted to islets (85), whereas in mice contradictory reports exist demonstrating Dmt1

expression restricted to islets (60, 84) or to a similar level between islets and acinar

cells (86). Mice selectively lacking Dmt1 in the β cell have been generated but no

measure of NTBI uptake has been made in islets or β cells from this model (86).

The second identified mammalian NTBI transporter is ZIP14, encoded by the

gene SLC39A14. ZIP14 stands for ZRT/IRT-like Protein 14, named after the similarity

between this protein and both zinc-regulated transporters (ZRT) and iron-regulated

transporters (IRT). Members of the ZRT gene family transport zinc in Saccharomyces

cerevisiae (87, 88), and IRT1 was identified as a route of iron transport in the roots of

iron-deficient Arabidopsis thaliana (89). ZIP14 was originally identified as a zinc

transporter and the iron transport capability of ZIP14 was not assessed until later, when

ZIP14 expression was reported to affect NTBI uptake and iron accumulation in

mammalian cells (90). Unlike DMT1, iron transport by ZIP14 is electrically neutral (75)

indicating iron uptake is accompanied by either the co-transport of anionic or the export

of a cationic species but the specifics of this have not yet been elucidated. ZIP14 has

been demonstrated to transport iron optimally at a neutral pH, with a loss of iron

transport as pH decreases (27, 75). The ability of Zip14 to transport iron at a physiologic

pH, similar to that of plasma, is consistent with a role for ZIP14 in the clearance of NTBI

at the plasma membrane. ZIP14 in mouse hepatocytes has been localized to the

plasma membrane (82, 91), as well as intracellular locations (27). The expression of

ZIP14 at the cell surface and intracellularly has also been reported in cell lines

overexpressing ZIP14 (90-92) and those expressing ZIP14 at endogenous levels (27).

Despite decreased iron transport ability at an acidic pH, ZIP14 has been demonstrated

Page 29: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

29

to colocalize with transferrin within endosomes and promote the assimilation of iron

from transferrin (27). More than 50% of TBI has been reported to dissociate from TF at

pH 6.5 (93), at which ZIP14 still demonstrates iron transport activity (75), indicating that

ZIP14 may contribute to the export of iron from the endosome into the cytosol. Four

mRNA transcript variants predicted to encode three unique protein isoforms of human

ZIP14 have been recorded within the UniGene database. Transcript variants 1, 2, and 3

encode isoforms of human ZIP14 comprised of 492 amino acids. However, the

predicted protein product of mRNA variant 2 differs mid-sequence from the protein

products of mRNA variants 1 and 3, which are identical in amino-acid sequence.

Transcript variant 4 encodes a 481-amino-acid peptide with a sequence identical to that

of variants 1 and 3 with the exception of the C-terminal region. Currently,

characterization of the differences in iron transport capabilities or intracellular targeting

between isoforms of human ZIP14 has not been performed but investigation into the

iron transport kinetics of mouse ZIP14 isoforms has been carried out in Xenopus

oocytes. Three transcript variants of mouse Zip14 encoding 2 protein isoforms, both

containing 489 amino acids but differing mid-sequence, have been reported and

identified as isoforms A and B. Isoform B is reported to demonstrate a greater affinity for

iron as well as a greater maximal rate of transport compared with isoform A (94).

However, this study did not control for differences in the expression of individual ZIP14

isoforms complicating the interpretation of these results as similar sequences in the

same expression vector may demonstrate differences in expression (76).

Early characterization of ZIP14 identified this protein as a potential contributor to

tissue NTBI uptake. Human ZIP14 mRNA expression was reportedly highest in the liver,

Page 30: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

30

pancreas, and heart, tissues known to accumulate iron during iron overload (92). The

cellular/subcellular localization of ZIP14 in human tissues has not yet been performed.

However, in rats Zip14 is detected at the hepatocyte sinusoidal membrane and

throughout pancreatic acinar cells, at the plasma membrane as well as intracellular

locations, but not β cells (82). Recent determination of the role ZIP14 plays in tissue

NTBI uptake using mice lacking Zip14 has indicated that Zip14 is required for iron

loading in hepatocytes and pancreatic acinar cells in response to both genetic and

dietary iron overload (7). In the absence of Zip14, iron deposits are only observed in the

non-parenchymal cells of the liver and within pancreatic connective tissue indicating that

ZIP14 is likely the sole route of NTBI uptake within hepatocytes and acinar cells. ZIP14

is unlikely to contribute to cardiac NTBI accumulation as hearts from ZIP14 knockout

mice display no difference in NTBI uptake compared with mice with intact ZIP14. In

addition to providing a route of NTBI uptake within the liver and pancreas, ZIP14

expression has also been reported to increase in response to iron loading within these

tissues (82). Mechanistic studies of the regulation of ZIP14 by cellular iron status have

shown that iron regulates ZIP14 posttranscriptionally, by preventing the proteosomal

degradation of ZIP14 (95). The upregulation of hepatic and pancreatic ZIP14 in

response to iron loading suggests that iron accumulation in the liver and pancreas may

increase the future rate of iron uptake in these tissues. In agreement with this

hypothesis, iron loaded HepG2 cells (96) and rodent hepatocytes (97, 98) demonstrate

increased NTBI uptake. However, while iron loading increases total-cell ZIP14

expression the abundance of ZIP14 on the plasma membrane is not increased, relative

to non-iron treated HepG2 cells (95), arguing against increased plasma NTBI clearance

Page 31: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

31

by ZIP14 during iron overload (although NTBI uptake in response to iron loading was

not measured in this study). Discrepancies in the subcellular distribution of ZIP14

between studies may be attributed to differences in the degree of iron loading achieved

(95, 96). Future studies will be required to determine if the increased rate of NTBI

uptake associated with cellular iron loading is attributed to ZIP14 upregulation.

The most recently described mammalian NTBI transporter is ZIP8, ZRT/IRT-Like

Protein 8, another member of the ZIP protein family encoded by the gene SLC39A8.

Within the ZIP protein family ZIP8 is the most similar to ZIP14 in amino-acid sequence,

with mouse ZIP8 and ZIP14 having approximately 50% shared identity (99). ZIP8 was

originally referred to as BIGM103 and identified as a protein induced in monocytes in

response to LPS or TNFα (100). Similar to ZIP14, ZIP8 was originally found to transport

zinc, as overexpression of ZIP8 increased zinc accumulation by CHO cells (100). In

light of the ability of ZIP14 to transport iron and the similarity between ZIP14 and ZIP8,

the iron transport activity of ZIP8 was measured revealing that ZIP8 overexpression and

suppression increase and decrease NTBI uptake, respectively, in mammalian cells (28).

ZIP8 is reported to transport iron optimally at pH 7.5, with a loss of transport activity with

decreasing pH, and is localized to the plasma membrane supporting the role of this

protein in iron uptake from the plasma (28). ZIP8 is also detected in endosomes

suggesting that ZIP8 may be able to contribute to iron export from endosomes into the

cytosol as ZIP8 demonstrates iron transport within mammalian cells at pH 6.5 (28, 101),

at which iron will dissociate from transferrin within endosomes (93). However, iron

transport in Xenopus oocytes is abrogated at pH 6.5, complicating the interpretation of

the role ZIP8 plays in endosomal iron transport (28). ZIP8 reportedly functions as an

Page 32: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

32

electrically neutral symporter (102), coupling the transport of cations with the transport

of bicarbonate, as cells overexpressing ZIP8 demonstrate increased ion uptake in the

presence of added bicarbonate and decreased metal uptake when bicarbonate

transport is inhibited (101). However, the possibility that the effect of bicarbonate on

metal transport was mediated through the action of a transporter other than ZIP8 was

not accounted for in this experiment.

ZIP8 mRNA is detected in many tissues but concentrated in the pancreas, lung,

placenta, liver, and thymus (100). Detection of ZIP8 at the protein level within these

tissues, from either human or animal sources, has been carried out to a very limited

degree, both in regards to tissue protein expression and the cellular/subcellular

localization of ZIP8 in vivo. The lack of data regarding the expression profile of ZIP8

currently limits the ability to make conclusions as to the role of ZIP8 in tissue NTBI

uptake during iron overload. ZIP8 is reportedly expressed at the plasma membrane of

rat β cells (103) but experiments to determine the contribution of ZIP8 to β cell iron

uptake have not been performed. Study of the role ZIP8 plays in tissue NTBI uptake has

also proved difficult due to the embryonic lethality of Slc39a8 disruption in mice. Mice

with Slc39a8 alleles disrupted by the neomycin-resistance cassette display

hypomorphic ZIP8 expression and fail to survive past post-natal day 3 (104). The

embryonic lethality observed in response to the disruption of ZIP8 is thought to be due

to impaired hematopoiesis during embryonic development (104). Mice with selective

deletion of Zip8 in tissues that accumulate iron during iron-overload disorders have yet

to be investigated. However, it is unlikely that Zip8 plays a role in NTBI uptake by

hepatocytes or pancreatic acinar cells as these cell populations display no iron loading

Page 33: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

33

in the absence of ZIP14 (7). ZIP8 may play a role in NTBI uptake by other hepatic or

pancreatic cell types as well as in other organs (e.g. heart) that are unaffected by the

loss of ZIP14. Similar to ZIP14, ZIP8 expression is upregulated by cellular iron loading.

However, unlike ZIP14 that demonstrates an increase in intracellular rather than cell

surface expression in iron-treated HepG2 cells (95), ZIP8 levels were observed to

increase at the cell surface in response to iron loading in H4IIE cells, a rat hepatoma

cell line (28). However, an increase in the protein level of ZIP8 in response to iron

loading in vivo has yet to be confirmed. More research is necessary to determine the

pattern of ZIP8 expression in human tissues and the contribution of ZIP8 to iron

transport.

Some reports indicate that NTBI can be taken up into cells through both L-type

Ca2+ channels (LTCC) and T-type Ca2+ channels (TTCC). Currently, the study of NTBI

transport by calcium channels has been restricted to cardiomyoctes, exploring

mechanisms by which iron may accumulate in cardiac tissue during iron overload.

LTCC agonists increase iron uptake by the mouse heart and LTCC blockers decrease

iron accumulation in mouse heart tissue perfused with ferrous sulfate (105).

Additionally, treatment with LTCC or TTCC blockers in vivo reduces cardiac iron

accumulation in mice injected with iron dextran and in a mouse model of β thalassemia

(106, 107). While Ca2+ channels show promising evidence for a role in cardiac NTBI

uptake, the contribution of Ca2+ channels to NTBI uptake by other cell populations has

not been investigated. However, other cell types, such as pancreatic β cells, express

both LTCC (108) and TTCC (109) and therefore may take up via calcium channels. The

Page 34: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

34

contribution of both LTCC and TTCC in various cell populations should be a topic of

future study.

The Iron and Diabetes Connection

Elevated Iron Stores and Diabetic Pathology

A link between iron overload and the development of diabetes dates back to the

initial case report of the disorder which would later become known as hemochromatosis.

Autopsy of an individual who died due to diabetic complications in 1865 noted the

bronze coloration of skin and organs referring to the condition as “bronze diabetes.”

Many years later it was established that the bronze discoloration observed was due to

excess iron deposition in tissues. The initial link between excess iron accumulation and

diabetes has been strengthened since the initial discovery by studying the prevalence of

diabetes in patients with pathological iron overload disorders.

In patients with hemochromatosis the incidence of diabetes varies considerably

among reports but is substantially greater than the incidence reported in the general

population of middle-aged Americans and Northern Europeans, in which

hemochromatosis is prevalent (110, 111). A general trend is that the reported incidence

of diabetes in hemochromatotic patients declines with the passage of time, with the

highest prevalence being reported in earlier manuscripts and a lower prevalence in

more recent reports. Initial reports indicate that diabetes is observed in approximately

80% of patients with hemochromatosis (112), although the diagnostic methods used

were not discussed. A later report based on data collected by physicians from an

unspecified time until 1972 reported that 63% of patients with hemochromatosis, as

defined by serum iron indices and liver biopsy, were diagnosed with diabetes (113). A

study of hospital records from 1977 to 1997 found that 40% of patients admitted to the

Page 35: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

35

hospital with hemochromatosis were diabetic (114). However, a substantially lower

incidence of diabetes was reported in a study of French and Canadian

hemochromatosis patients between 1970 and 1997 which only identified 15.9% of men

compared with 7.4% of women to have diabetes (115). However this study may have

under reported the prevalence of diabetes as patients were not tested for undiagnosed

diabetes in this study. Glucose tolerance testing in hemochromatosis patients between

2000 and 2006 determined that 23% of patients were diabetic and 30% had impaired

glucose tolerance or elevated fasting glucose (45). The same study corroborated these

findings by examination of the medical records from the study center from 1975 to 2006

which reported that 26% of hemochromatosis patients were diabetic, as defined by a

measurement of elevated fasting blood glucose.

The general decrease in the reported prevalence of diabetes in patients with

hemochromatosis over time may reflect improvements in diagnosis, such as genetic

testing, and improved implementation of treatments. In hemochromatosis patients

diagnosed after the advent of genetic testing, post 1995, the prevalence of diabetes was

17.7% compared with 35.6% in patients diagnosed with hemochromatosis by elevated

iron indices between 1983-1995, prior to genetic testing (116). However, it should be

noted that there was no difference in the age of diabetic diagnosis between the groups

in this study allowing the older patients, diagnosed with hemochromatosis by iron

parameters rather than by genetic testing, a longer time period to develop diabetes. In

line with this concept the prevalence of impaired glucose tolerance is greater in patients

diagnosed with hemochromatosis in recent years, 13%, compared with those diagnosed

pre-1995 (6.7%) (116). It is difficult to interpret from this study whether impaired glucose

Page 36: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

36

tolerance will progress to diabetes, resulting in a similar prevalence of diabetes as was

observed in patients diagnosed with hemochromatosis by traditional metrics of iron

overload, rather than by genetic testing, if given the same amount of time. The study

carried out by Bussychard et al. (114) is in agreement with the concept that diabetes

incidence likely is inversely related to the management of hemochromatosis and

lifestyle choices as diabetes was prevalent in 53% of patients with cirrhosis but only

25% of those without cirrhosis. Ferritin levels were significantly higher in patients with

cirrhosis suggesting poor disease management relative to those with no cirrhosis and a

lower incidence of diabetes. The addition of other factors associated with diabetes risk,

such as obesity, may also affect the development of diabetes in hemochromatosis

patients. One report has documented that all patients with hemochromatosis who were

identified as diabetic by glucose tolerance testing were overweight or obese (45),

suggesting that the influence of iron overload on diabetic pathology may increase when

coupled with additional risk factors for diabetes, such as obesity. However, another

study reported no difference in BMI between patients with hemochromatosis alone, 24.2

kg/m2, or with hemochromatosis accompanied by diabetes, 25 kg/m2, (117). The

development of diabetes in non-obese hemochromatosis patients indicates that obesity

is not required for the development of diabetes in patients with iron overload.

Evidence linking pathological iron overload to the development of diabetes is also

provided by studies measuring the prevalence of diabetes in patients with thalassemia

major. Thalassemic patients frequently develop diabetes, often at a young age when

diabetes is rare in the general population. The prevalence of diabetes varies among

studies likely reflecting differences in demographic factors including frequency of

Page 37: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

37

transfusion, age of patients, and improvements in disease management or treatment

options. One study reported an initial diabetic prevalence of 26%, increasing to 30%

after a 2-year follow up (118). Another study documented a 14% prevalence of diabetes

out of all cases seen over the course of a 30-year study (119). However, diabetes was

only documented in patients over the age of 23, yielding a prevalence of 30% in this

demographic within the study. The observed increase in the prevalence of diabetes with

age supports the concept of longer-duration iron accumulation increasing diabetes risk

in patients with thalassemia as well as novel treatment options potentially improving

disease prognosis, as evidenced by improved overall survival with increased chelator

treatment (119). A lower prevalence of diabetes, 6.5%, has been reported in patients

from various centers in Italy which followed patients diagnosed with thalassemia

between 1970 and 1983. The lower prevalence in this study may be partially by more

stringent diagnostic criteria for diabetes, fasting glucose higher than 140 mg/dL for

several consecutive days or overt symptoms of diabetes, such as glycosuria. It is

possible that asymptomatic or mild diabetes may have been overlooked in this study

(120).

While an increased incidence of diabetes in pathological disorders resulting in

iron overload is well established, elevated iron stores remaining within the normal range

may also contribute to the pathogenesis of diabetes. Many prospective epidemiological

studies have investigated the link between markers of iron status, most often

determined by serum ferritin, and the risk of developing diabetes in the future (121-125).

Nearly all studies report increased baseline serum ferritin levels in subjects that develop

diabetes at a later time point in the study supporting the link between elevated normal

Page 38: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

38

iron stores and glucose dyshomeostasis. However, the use of serum ferritin as an

indicator of iron stores is problematic due to the nature of ferritin as an acute-phase

reactant induced by inflammatory signaling pathways (126). Chronic systemic

inflammation is hypothesized to contribute to diabetic pathology (127) and therefore it is

difficult to determine whether ferritin is elevated in subjects who develop diabetes due to

increased systemic iron or simply in response to low-grade inflammation preceding the

development of diabetes. In line with the concern that serum ferritin is a marker of

inflammation rather than a barometer of iron stores, markers of inflammation, such as

C-reactive protein (CRP), are often higher in the diabetic cohort at baseline relative to

the control patients which did not develop diabetes during follow up (121, 123, 125).

However, when relative risk is adjusted to account for differences in CRP, serum ferritin

levels remain an independent risk factor for developing diabetes (123).

Fewer studies make use of soluble transferrin receptor (sTFR) levels in serum to

measure iron status, either alone or in combination with serum ferritin (123-125). sTFR

is reported to inversely correlate with iron stores independently of inflammatory stimuli

(128, 129). Unlike serum ferritin, sTFR as an indicator of iron status does not suggest

elevated baseline iron stores in patients who developed diabetes arguing against the

modulation of diabetes risk by iron stores within normal levels (123). Some studies do

not directly report the value for sTFR, instead referencing the ratio of sTFR to serum

ferritin (130). However, the data for ferritin listed individually in these studies suggest

that sTFR is largely similar at baseline between normal patients and future diabetics.

Elevated sTFR, suggesting lower iron stores, has even been reported in baseline

measurements from patients who developed diabetes compared with controls (125). In

Page 39: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

39

light of the confounding influence of inflammation on ferritin levels and the lack of a

clear trend observed with sTFR levels, the link between variation in non-pathological

iron stores and the development of diabetes appears unsubstantiated, despite being

often mentioned in the literature.

Pathological iron accumulation leads to transferrin saturation and the appearance

of NTBI in circulation. Additionally, plasma NTBI has been reported in type 2 diabetics

despite normal levels of transferrin saturation and without pathological iron

accumulation (131). Plasma NTBI present in diabetics or in patients with severe iron

loading can be taken up by tissues such as the pancreas.

Pancreatic Iron Accumulation

Due to the role of the pancreas, specifically the pancreatic β cells, in glucose

homeostasis and the pathogenesis of diabetes, pancreatic iron accumulation is thought

to potentially account for the increased prevalence of diabetes in humans with

pathological iron overload.

Reports of iron loading in the human pancreas from patients with either

hemochromatosis or thalassemia indicate that iron deposits heavily within the acinar

cells of the exocrine pancreas and to a somewhat lesser degree within islets of the

endocrine pancreas as determined by various iron-staining techniques (132-134).

Within the islet, iron staining has been determined to be primarily restricted to β cells,

with α cells remaining relatively free of iron deposits (132, 134). No studies have

reported iron accumulation in the other cell populations of the pancreatic islet (e.g. δ

cells, ε cells). Due to the invasive nature of pancreatic biopsy, all data published on

human pancreatic iron loading at this point are derived from autopsy cases. Therefore,

the current reports indicate the pattern of pancreatic iron loading at end of life usually

Page 40: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

40

after long periods of iron overload. Magnetic resonance imaging has been used to

detect pancreatic iron accumulation and could be used to measure the progression of

pancreatic iron overload over long periods in patients with iron overload (135). However,

magnetic resonance imaging is not currently able to distinguish between cell

populations within the pancreas precluding the use of this method to discern between

iron loading in the exocrine versus endocrine pancreas. New non-invasive methods of

measuring pancreatic iron in vivo will need to be developed before the nuanced

characterization of pancreatic iron loading over time in humans is feasible.

While the cell type-specific examination of pancreatic iron loading is not possible

in humans, extensive studies in rodents have been carried out with both genetic and

dietary models of iron overload. However, rodent models fail to appropriately model

human pancreatic iron loading as both mouse and rat islets remain largely free of iron

deposition even during severe iron overload (60, 134, 136, 137). Studies using Hjv

(138) and Hamp (136) knockout mice, resulting in severe pancreatic iron accumulation,

clearly show that β cells/islets fail to accumulate iron despite iron building up in the

surrounding exocrine pancreas. In accord with the lack of islet iron deposition in these

models no diabetic phenotype, characterized by impaired glucose tolerance resulting

from insufficient insulin secretion, is detected even up to a year of age in HAMP

knockout mice (136). The resistance of mice islets to iron loading and subsequent

diabetic pathology during severe iron overload is best evidenced by a novel mouse

model with a mutation in ferroportin resulting in impaired regulation by hepcidin leading

to severe tissue iron loading (60). Massive iron accumulation in this mouse model

results in death due to exocrine pancreas failure, a phenotype not reported in other

Page 41: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

41

mouse models of iron overload, suggesting that this model potentially represents the

most severe model of pancreatic iron loading. Yet, even in this mouse model islets are

spared from iron deposition and glucose homeostasis is unchanged compared with

mice that have the wild-type ferroportin allele.

Rats also fail to recapitulate the human condition with regard to substantial islet

iron loading and the development of a diabetic phenotype. Several feeding studies with

high-iron diets at various concentrations and durations have been performed using rats

(137, 139-142). However, none report a diabetic phenotype even when pancreatic iron

loading was observed. Studies that have achieved pancreatic iron loading in rats

through prolonged feeding with high-iron diets report that, similar to mice, iron

deposition occurs within the exocrine pancreas, in both acinar cells and interstitial

areas, but islets were spared from iron loading (140). High dietary iron in rats has also

been reported to lead to pancreatic atrophy, of both the exocrine and endocrine

pancreas, during which pancreatic tissue is replaced by adipose tissue (141, 142).

However, iron staining or the measurement of pancreatic iron was not reported in these

studies to demonstrate that iron deposits were localized to atrophic cells. Currently it

remains unclear whether islet atrophy in response to high dietary iron is due to islet iron

accumulation or simply the loss of normal surrounding pancreatic morphology, as islets

are reported to resist atrophy longer than acinar cells (142). The most extreme

incidence of pancreatic iron loading through diet combined the surgical bypass of the

liver by the portal circulation in conjunction with feeding a high-iron diet for an extended

period of time resulted in severe pancreatic iron overload (137). However, within the

islet very little iron deposition was observed relative to the surrounding acinar cells. No

Page 42: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

42

determination of glucose homeostasis was carried out in this study, but the authors

reported no overt diabetic phenotype despite an extreme treatment to induce pancreatic

iron loading. Additionally pancreatic atrophy was only reported in one rat that received

this treatment. It is possible that genetic differences between rat strains account for

differences in susceptibility to pancreatic atrophy in response to iron loading.

Iron loading of the pancreas has also been reported in rats injected with high

doses of iron. Rats administered iron dextran demonstrate detectable iron staining

within the exocrine pancreas, with the most prominent staining reportedly detected

within macrophages, while islets remained free of iron deposits. Injection of ferric

nitrilotriacetate (Fe-NTA) daily for 120 days in rats resulted in heavy iron deposits visible

by Perl’s staining in acinar cells, macrophages, and pancreatic connective tissue but

iron loading was sparse within islets (143). Similar experiments repeated by the same

group produced similar results reporting iron deposits in the exocrine pancreas, while

the islet remained free of detectable iron accumulation until 6 months after the start of

injections, when faint staining was detected within islets (144). Experiments that use Fe-

NTA injections to produce iron overload are unique in that they report a diabetic

phenotype, characterized by hyperglycemia and reduced insulin secretion (143, 144).

However, these studies report the development of diabetes within 1-2 (143, 144)

months of iron injections while iron deposits are not detectable in islets until 6 months

after the start of injections (144) making it difficult to conclude that it is β cell iron loading

that directly leads to impaired insulin production in this model. Additionally Fe-NTA

administered via injection represents both a non-physiologic source and method of iron

loading.

Page 43: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

43

Mechanistic Evidence from Human Studies

Currently there is a lack of an established physiologically relevant rodent model

that accurately models the pancreas of humans with iron overload, characterized by

significant iron deposition in β cells in response to genetic, transfusional, or dietary iron

overload. Due to the limitation of in vivo models, mechanistic investigation into the

pathogenesis of iron-induced diabetes has been difficult; however, studies using

humans with iron-overload disorders provide some insight into the pathology of iron-

induced diabetes.

Diabetes in humans with iron overload can result from either impaired insulin

production by β cells, decreased peripheral insulin sensitivity, or a combination of these

factors (145). The study of humans with hemochromatosis suggests that impaired

glucose homeostasis is due, at least in part, to impaired insulin secretory capacity.

Hemochromatosis is associated with a trend toward decreased first and second phase

insulin secretion during glucose tolerance testing and individuals with hemochromatosis

and diabetes show drastically reduced first phase insulin secretion (117). This finding

has been corroborated in the literature as middle-aged, non-obese patients with

hemochromatosis demonstrate lower glucose-stimulated insulin secretion compared

with relatives without hemochromatosis (45). Somewhat conflicting evidence exists for

the role of insulin sensitivity in glucose homeostasis during hemochromatosis. One

report indicates that insulin sensitivity is not different between control subjects and

patients with hemochromatosis without cirrhosis or diabetes (117). However, in patients

with hemochromatosis and either cirrhosis or diabetes, insulin sensitivity is significantly

impaired, potentially attributable to greater iron loading, as indicated by higher serum

ferritin levels, compared with non-symptomatic hemochromatosis. Another study

Page 44: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

44

reported that hemochromatosis patients with normal glucose tolerance maintain greater

insulin sensitivity, despite similar age and body mass indices, compared with normal

patients, although again this was lost in patients with diabetic complications (45).

Studies from animal models of hemochromatosis support the position of unchanged

insulin sensitivity in human hemochromatosis as insulin sensitivity in both Hamp-/- and

Hfe-/- mice is similar to that in wild-type mice (136, 146). Conflicting evidence also exists

regarding the effect of iron depletion by phlebotomy on β cell function and insulin

sensitivity in hemochromatosis. The removal of systemic iron stores, as indicated by

diminished plasma ferritin levels, by phlebotomy is reported to improve insulin secretory

capacity to varying degrees (147, 148), although this is interestingly associated with

unchanged glucose tolerance attributed to either decreased insulin sensitivity or

increased hepatic insulin clearance. The extent of improvement in insulin secretion, post

phlebotomy, may be due to the amount of iron removed and the severity of initial iron

accumulation, as a more prominent improvement in insulin secretory capacity was seen

in individuals with greater initial iron loading (147) compared with more mild iron

accumulation (148). However, normalization of iron stores by phlebotomy has also been

reported to have no effect on either first-phase insulin secretion, although longer time

points were not examined and insulin sensitivity was not measured (117). In diabetic

patients without hemochromatosis but with elevated ferritin levels, phlebotomy improved

both insulin secretory capacity and insulin sensitivity, supporting the idea that elevated

iron levels negatively affect glucose homeostasis by modulating both β cell function and

peripheral tissue insulin sensitivity (149).

Page 45: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

45

In patients with β thalassemia major glucose dyshomeostasis may result from

either decreased insulin sensitivity, impaired β cell function, or some degree of both of

these factors. In thalassemics over the age of 18, insulin secretion in response to

glucose was elevated but glucose tolerance was unchanged relative to control subjects,

indicating diminished insulin sensitivity but increased β cell function (150). Decreased

insulin sensitivity compensated by increased insulin production in patients with

thalassemia has been reported elsewhere and it was documented that the phenotype

became more pronounced with the extent of transfusional iron overload (151). With the

onset of adulthood, glucose tolerance worsened and a loss of first-phase insulin

secretion was observed, indicating impaired β cell function after initial insulin resistance

(151). The finding of eventual β cell failure corroborates other reports of decreased

insulin sensitivity and a lack of compensatory insulin secretion leading to impaired

glucose tolerance, attributed to β cell failure (152). Impaired glucose tolerance in

patients with thalassemia has also been attributed to impaired insulin secretion by other

studies, however insulin sensitivity was not directly measured in these reports (153,

154). In patients with thalassemia improved treatment with iron chelators is reported to

marginally improve glucose tolerance, attributed to improved insulin sensitivity rather

than increased insulin secretion (155). Currently, the specifics of the relative

contribution between systemic insulin resistance and declining β cell function to the

development of diabetes in patients with thalassemia are still undefined. Variation in

these reports likely reflects differences between the subjects studied possibly including

lifestyle factors such as effectiveness of chelation therapy, extent of iron overload, or

age of participants.

Page 46: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

46

While the clinical characteristics of hemochromatosis and thalassemia differ with

regard to insulin sensitivity, both disorders are linked to a diminished insulin secretory

capacity suggesting impaired β cell function. Several reports indicate that islets from

humans, as well as rodents, express low levels of or display low activity of antioxidant

proteins, relative to other tissues such as the liver, suggesting that islets may be easily

damaged by oxidative stress (156-158). During iron overload it has been hypothesized

that iron accumulation catalyzes the formation of reactive oxygen species (ROS), which

damage islets and leads to a loss of insulin secretory capacity. However, the effect of

iron loading on β cell function, evidenced by markers of oxidative stress and indicators

of β cell function such as glucose-stimulated insulin secretion, has not been examined

in human islets.

In addition to iron accumulating directly in β cells leading to impaired function it

has also been suggested that iron loading of the exocrine pancreas can result in

pancreatic atrophy, compromising β cell function due to the loss of normal pancreatic

morphology. Diminished pancreatic mass and an increased proportion of fatty tissue in

the pancreas has been reported in patients with hemochromatosis that have developed

diabetes (132). However, it is possible that in patients with both iron overload and

diabetes diminished pancreatic mass is a result, rather than the cause, of decreased β

cell function as insulin is thought to promote acinar cell growth (159) and a lack of

insulin is associated with decreased pancreatic mass (160). Compelling evidence

against the “degeneration-dysfunction” hypothesis is provided by a mouse model of iron

overload that demonstrates severe degeneration of the exocrine pancreas and the

failure to produce digestive enzymes resulting in death. However, glucose tolerance is

Page 47: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

47

intact in this model despite pancreatic degradation (60) suggesting that iron-mediated

damage of the exocrine pancreas has little effect on β cell function.

Mechanistic Evidence from Animal Studies

Studies carried out using mice are complicated by the lack of substantial β cell

iron accumulation and the development of a diabetic phenotype. However, several

studies have still been performed in mouse models of iron overload which have reported

differences in β cell function and/or mass. Islets from Hfe knockout (Hfe-/-) mice on a

129/SvEvTac genetic background are reported to have marginally elevated iron levels

compared with Hfe+/+ mice and this is associated with diminished islet mass, pancreatic

insulin content, and in vitro GSIS by isolated islets (146) attributed to increased β cell

oxidative damage and apoptosis. However, in vivo glucose tolerance was improved in

Hfe-/- mice compared with Hfe+/+ controls (146, 161), despite slightly reduced insulin

secretory capacity during the first 30 min following intraperitoneal glucose injection

(146). In aged Hfe-/- mice, on a C57BL6 background, glucose tolerance was reportedly

reduced attributed to a failure to increase insulin secretion compared with Hfe+/+ mice,

suggesting the burden of added iron results in increased β cell dysfunction with time.

Follow up experiments with the Hfe-/- mice have suggested that the phenotype observed

is due to a cellular manganese deficiency in response to elevated iron levels leading to

insufficient Mn-SOD activity and impaired mitochondrial function (162). Supplementation

with manganese improved insulin secretion and glucose tolerance in Hfe-/- mice. The

Hfe-/- mouse represents a relatively mild model of iron overload, accumulating little

additional iron in the pancreas (7), and it would be expected that the non-significant

trend towards reduced insulin secretion in this model, on both 129/SvEvTac and

C57BL6, backgrounds would manifest as an overt phenotype in a more drastic model of

Page 48: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

48

iron overload, such as Hamp knockout mice. However, aged mice lacking hepcidin,

resulting in massive iron overload, report no difference in either glucose tolerance or

GSIS (136), suggesting the slight differences observed in Hfe-/- mice may be attributable

to a factor other than iron overload. Additionally the phenotype of improved glucose

tolerance, attributed to increased glucose disposal as a result of increased AMPK

activity in Hfe-/- mice, was not reported in Hamp-/- mice, again suggesting other signaling

pathways besides iron sensing may be altered in Hfe-/- mice.

Leptin-deficient, Ob/Ob mice on a C57BL/6J background have also been used as

a model to reveal the possible contribution of iron status to dysglycemia during

conditions when β cell function is stressed. It has been reported that feeding of a diet

containing 500 ppm iron vs 35 ppm results in increased insulin resistance and a loss of

insulin secretory capacity in response to obesity (163). Similar findings were also

reported in response to iron chelation in the same study. However, in this study no iron

measurements were performed for any tissue making it difficult to conclude whether the

observed differences were due to the impact of iron on the β cell or rather systemic

differences.

Dietary iron deficiency and phlebotomy have also been reported to increase

pancreatic insulin content in obese rats compared with those fed a control diet, in which

pancreatic insulin levels were unaffected by iron deficiency, (164) attributed to lower

levels of islet fibrosis. However, it unlikely that the fibrosis was due to islet iron

accumulation as iron staining did not reveal any iron deposits in the islets, with iron only

accumulating in macrophages, and islet iron was not otherwise quantified in any

manner. Rats in this study were not fed an iron-loaded diet but plasma ferritin levels

Page 49: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

49

were elevated suggesting systemic inflammation, potentially accounting for the

observed islet fibrosis and loss of function.

Rodents are poor models of β cell iron loading, as previously discussed,

complicating the interpretation that changes in β cell function in response to iron loading

in vivo are due to β cell iron accumulation rather than other systemic factors as a result

of systemic iron accumulation. Currently, investigation into the influence of islet iron

status on function in vitro, without the confounding factors potentially introduced by

changes in systemic iron status in vivo, has not been carried out. However, treatment of

rat islets with iron has been demonstrated to modestly decrease islet viability, attributed

to β cell death, and increase markers of oxidative stress (165). Future studies will be

needed to determine if increased oxidative stress due to iron loading results in impaired

islet function.

Iron and Autoimmune Diabetes

The vast majority of research examining the relationship between iron status and

diabetic pathology has focused on the impact of iron on exacerbating β cell stress or

worsening insulin resistance. The failure to produce adequate insulin to regulate

glucose levels due to either β cell exhaustion, insulin resistance, or a combination of

these factors is associated with type 2 diabetes; however, less is known about the

impact of iron status on the development of autoimmune type 1 diabetes.

Epidemiological evidence is less prevalent and provides a less compelling link between

iron and autoimmune diabetes compared with type 2 diabetes. The consumption of

greater dietary iron within the first 4 months of life has been associated with an

increased risk of developing type 1 diabetes before the age of 6 (166). However, the

collection of data for this study relied on 1 retrospective survey, administered 6 to 10

Page 50: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

50

years after the feeding period of interest, in which the parents were asked about the

child’s food consumption. Therefore, the data obtained in this study, and the

conclusions drawn, may be questionable. Elevated transferrin saturation, indicating

higher iron status, was also associated with an increased risk of type1 diabetes in adults

(167). However, it is also a possibility that in this study elevated transferrin saturation

was a result of diabetes, rather than a causative factor, as hepatic insulin signaling has

been demonstrated to regulate hepcidin production and iron absorption (168). One

caveat in these epidemiological studies is that type 1 diabetes is defined as “insulin-

dependent” diabetes rather than directly indicated as type 1A diabetes, indicating

autoimmune involvement, or type 1B diabetes indicating an idiopathic origin. No

indicators of the mechanistic origin of type 1 diabetes are measured in the current

studies and therefore it is unclear whether insulin deficiency is attributable to the

autoimmune destruction of β cells. Additionally it has been suggested that some cases

of late-onset type 1 diabetes may be the result of undiagnosed hemochromatosis

leading to the loss of β cell function. The prevalence of HFE C282Y homozygosity was

greater in type1 diabetics who were diagnosed after the age of 30, allowing time for

systemic iron loading, compared with the general population and that follow up

investigation of these patients indicated iron overload (169). However this study did not

determine the mechanism of diabetic development in patients with type 1 diabetes and

iron overload and therefore it cannot be established if hemochromatosis was directly

responsible for the loss of β cell function or if iron had an effect on autoimmunity in

these patients. Currently, the epidemiological evidence linking iron status with

autoimmune diabetes is tentative and very limited; a more compelling case can be

Page 51: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

51

made from studies that treated rodent models of autoimmune diabetes with factors

capable of modulating aspects of iron metabolism.

Several reports provide indirect evidence that iron restriction may limit critical

aspects of β cell death during autoimmune diabetes. Injection of the iron chelator

desferrioxamine prolongs the survival of islets grafts in NOD mice, suggesting that iron

restriction may limit T cell mediated β cell destruction (170). However, no measure of

either systemic or cellular iron status or further mechanistic investigation was carried out

in this study, thus complicating the interpretation of these results. Treatment with anti-

TFR antibodies has also been demonstrated to impair T cell proliferation and

cytotoxicity in vitro (171). The mechanism by which TFR interference affects T cell

activation is unclear but could potentially be linked to impaired iron acquisition limiting

activity or proliferation. However, the antibody used to bind TFR is reported to not

interfere with TF-TFR interaction, arguing against the blockade of iron uptake as an

explanation for impaired the impaired T cell response. It should be noted that in this

study the actual measure of cellular iron uptake was not carried out and it was only

assumed that if binding between TFR and TF was able to occur then successful iron

acquisition was also unhindered (172). In line with TFR signaling playing a role in

immune activation, apotransferrin has demonstrated a protective effect towards islets

during diabetic pathogenesis. Treatment of cultured mouse islets with apotransferrin

reduces the loss of viability observed in response to incubation with proinflammatory

cytokines and administration of apotransferrin to rodent models of spontaneous

autoimmune diabetes decreases the incidence and delays the onset of diabetes (173).

The protective benefits of apotransferrin in these models have been attributed to

Page 52: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

52

decreased production of proinflammatory cytokines and reduced insulitis, characterized

by the infiltration of pancreatic islets by immune cells. The mechanism through which

apotransferrin elicits this shift in the immune response has not been elucidated but

could potentially affect cellular iron acquisition by interfering with cellular iron uptake via

TF. However the binding affinity for apotransferrin is considerably lower compared with

holotransferrin arguing against this theoretical mechanism (174, 175). While current

findings indirectly suggest that iron depletion may be protective against the

development of autoimmune diabetes no investigation into the direct influence of

systemic or cellular iron status on diabetic pathology has been performed at this time.

Future studies will need to be carried out to determine the impact of iron status on the

development of autoimmune diabetes.

Page 53: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

53

CHAPTER 3 MATERIALS AND METHODS

Animals and Diets

Weanling male Sprague-Dawley rats (Charles River Laboratories), used in

experiments detailed within chapter 4, were randomized (n=6/group) to receive either

iron-deficient (FeD), iron-adequate (FeA), or iron-overloaded (FeO) diets. Purified diets

were prepared according to the AIN-93G formulation, but with no added iron (FeD), 35

mg/kg ferric citrate (FeA), or 2% carbonyl iron (Sigma-Aldrich) (FeO). Iron contents of

the diets, as determined by inductively coupled plasma mass spectroscopy (ICP-MS),

were 5 ppm (FeD), 36 ppm (FeA), and 20,275 ppm (FeO). Diets were also modified to

contain Avicel® microcrystalline cellulose instead of cellulose and 20% sucrose instead

of 10% sucrose (while reducing the amount of cornstarch accordingly) (176). After 3

weeks of feeding, overnight-fasted rats were sacrificed by exsanguination from the

descending aorta. Blood was collected into heparinized syringes and then centrifuged

to obtain plasma. Pancreata were quickly harvested, immediately frozen in liquid

nitrogen, and maintained at -80 °C for subsequent analyses. Weanling female

NOD/ShiLtJ mice (The Jackson Laboratory) ,used in experiments detailed within

chapter 6, were randomized to receive either iron-deficient (FeD), iron-adequate (FeA),

or iron-overloaded (FeO) diets. Purified diets were prepared according to the AIN-76A

formulation, but with no added iron (FeD), 270 mg/kg ferric citrate, or 10000 mg/kg

carbonyl iron. Diets were modified to contain 5% wheat, to increase the diabetic

potential of the diet, and avicel microcrystalline cellulose, replacing cellulose, to reduce

contaminate iron. Additionally diets were heated in a convection oven at 125°C for 30

m, as this treatment has previously been reported to increase the incidence of diabetes

Page 54: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

54

in mice fed a purified diet, and subsequently irradiated. Iron contents of the finalized

diets, as determined inductively coupled plasma mass spectroscopy (ICP-MS) analysis

were 14 mg/kg (FeD), 359 mg/kg (FeA), and 6629 (FeO) mg/kg dry matter. To account

for potential heat degradation of vitamins during diet preparation an additional 10 g/kg

vitamin mix was added to all diets. Animals were fed their respective diets until removal

from the study, due to either assignment to prediabetic analysis at 10 wk of age, study

termination at 30 wk of age, or detection of diabetes during the study. Testing for

diabetes was carried out biweekly starting at 8 wk of age by using a handheld

glucometer (Accu-Chek). Animals were considered to be diabetic if glycosuria was

detected on 2 consecutive days followed by 2 consecutive blood glucose readings of

>250 mg/dL. Upon removal from the study, animals were fasted overnight and sacrificed

by isofluorane inhalation.

Iron Status Parameters and Blood Glucose Concentrations

Hemoglobin levels in heparinized blood were measured by using a HemoCue Hb

201+ Hemoglobin Analyzer (HemoCue). Liver non-heme iron levels were determined by

colorimetric analysis of acid-digested tissues, as described previously (177). Briefly,

tissues were weighed and digested in a solution of HCl, trichloroacetic acid. After

incubation at 65ᵒ C for 20 h, the iron content of dissolved samples was measured by the

addition of the dissolved sample to a chromogen reagent containing

bathophenanthroline disulphonate, thioglycolic acid, and saturated sodium acetate.

Color change, indicating non-heme iron concentration, was measured by using a

spectrophotometer and compared with a dilution series made from an iron-reference

solution (Fisher). Plasma iron and total iron-binding capacity (TIBC) were measured

colorimetrically by the methods described by Cook (1985). Briefly, for plasma iron

Page 55: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

55

determination a solution of trichloroacetic acid, HCl, and thioglycolic acid was added to

plasma followed by centrifugation to precipitate proteins and reduce plasma iron.

Plasma supernatant was then added to a chromogen solution composed of sodium

acetate and ferrozine and iron concentration of unknown samples and a reference iron

dilution series (Fisher) measured by using a spectrophotometer. TIBC was calculated

by saturating plasma transferrin with iron followed by addition of magnesium carbonate

to remove excess iron prior to the measurement of plasma iron. Blood glucose

concentrations were determined in freshly collected heparinized blood by using a

handheld glucometer (Accu-Chek).

Pancreatic Mineral Concentrations

Concentrations of iron, zinc, manganese, copper, and cobalt in pancreas

samples were determined by using ICP-MS. Analyses were performed by the Michigan

State University Diagnostic Center for Population and Animal Health.

Histological Analysis

10% buffered formalin phosphate-fixed and paraffin embedded pancreases from

10 wk NOD were stained with hematoxylin and eosin and scored for insulitis. Insulitis

was scored based on the scale 0, no insulitis; 1, peripheral-insulitis, 2, mild-insulitis

(<50% of islet infiltrated); 3, severe insulitis (≥ 50% islet infiltrated). An average of 35

islets from at least 3 unique pancreatic sections per animal were scored and the

average insulitis score reported.

RNA Isolation and Assessment of RNA Integrity

Frozen pancreas samples were submerged in liquid nitrogen and finely ground

by using a mortar and pestle. After grinding, total RNA was isolated by using the

RNeasy Mini Kit (Qiagen) following the manufacturer’s protocol. Integrity of isolated

Page 56: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

56

RNA was confirmed by denaturing agarose gel electrophoresis followed by visualization

of 18S and 28S ribosomal RNA bands. Prior to microarray analysis, RNA integrity was

additionally assessed by using the Agilent 2100 Bioanalyzer and RNA 6000 Nano Kit

(Agilent).

Microarray Analysis

RNA samples (n=6) from each dietary group were pooled and analyzed by using

a Rat GE 4x44K v3 Microarray (Agilent). Both FeD and FeO pooled cDNA samples

were individually compared with FeA in duplicate measurements. The assignment of

fluorescent Cy3 or Cy5 dye to each comparison group was alternated between the

duplicates. To identify differential gene expression, values of signal intensity were log2

transformed and normalized before the Student’s t-test was performed for probe-

specific comparisons. Genes showing a statistically significant (P<0.05) log2-

transformed fold change of at least ± 2 were analyzed to identify functional biological

categories by using the Database for Annotation, Visualization and Integrated Discovery

(DAVID) (178). Microarray analysis was conducted at the Interdisciplinary Center for

Biotechnology at the University of Florida. The microarray data discussed herein have

been deposited in NCBI's Gene Expression Omnibus (179) and are accessible through

GEO Series accession number GSE44699

Relative mRNA Quantification

cDNA was synthesized from total RNA by using the High Capacity cDNA

Reverse Transcription Kit (Applied Biosystems). Specific primers for genes of interest

were generated (Table 4-5) and confirmed for specificity by using the NCBI Basic Local

Alignment and Search Tool (BLAST) (180). Quantitative reverse transcriptase

polymerase chain reaction (qRT-PCR) was performed by using Power SYBR Green

Page 57: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

57

PCR Mastermix (Applied Biosystems) and an Applied Biosystems 7300 Real-Time PCR

System. Dissociation curve analysis of PCR products revealed single products and all

PCR amplification efficiencies were 100 ± 10%. Quantitation of mRNA was determined

by comparison to standard curves generated by four 10-fold serial dilutions of standard

cDNA. Transcript levels were normalized to those of cyclophilin B (peptidylprolyl

isomerase B, PPIB).

Western Blotting

Pancreas samples were homogenized in ice-cold buffer containing 0.05 M Tris-

HCl (pH 7.4), 0.05 M NaCl, 0.001 M EDTA, 0.25% Tween-20, and Complete, Mini

Protease Inhibitor Cocktail (Roche). Tissue homogenates were clarified by

centrifugation at 10,000 x g for 10 minutes at 4 ºC, followed by sonication of the

supernatant. Protein concentrations were determined by using the RC DC Protein

Assay Kit (Bio-Rad). Proteins were mixed with Laemmli buffer, incubated at 70 °C for

10 minutes, and then electrophoretically separated by sodium dodecyl sulfate-

polyacrylamide gel electrophoresis (SDS-PAGE) on a 7.5% gel. Separated proteins

were transferred to a polyvinyl difluoride (PVDF) membrane (Bio-Rad), and incubated in

blocking buffer (5% nonfat dry milk in Tris-buffered saline (TBS)-Tween 20 (TBS-T)) for

1 hour. The blot was then incubated with rabbit anti-rat Alox15 antibody (kindly provided

by Dr. James F. Collins, University of Florida), 1:8,000 dilution for 2 hour. After washing

with TBS-T, the blot was incubated with horseradish peroxidase (HRP)-conjugated

donkey anti-rabbit IgG secondary antibody (Amersham Biosciences), 1:10,000 dilution

for 45 minutes. After washing with TBS-T and TBS, antibody binding was observed by

using enhanced chemiluminescence (SuperSignal West Pico, Pierce) and the

Fluorchem E imaging system (ProteinSimple). To indicate lane loading, the blot was

Page 58: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

58

stripped and reprobed with a mouse anti-α-tubulin antibody (Sigma) at a 1:5,000

dilution, followed by an HRP-conjugated goat anti-mouse IgG secondary antibody

(Santa Cruz) at a 1:10,000 dilution. Densitometry was performed by using AlphaView

software (ProteinSimple). Cell samples were lysed and sonicated in RIPA buffer

containing 150 mM sodium chloride, 1% IGEPAL, .5% sodium deoxycholate, .1%

sodium dodecyl sulfate, and 50 mM tris-base, and Complete, Mini Protease Inhibitor

Cocktail (Roche). The RC DC Protein Assay Kit (Bio-Rad) was used to determine lysate

protein concentrations and samples were mixed with Laemmli buffer and incubated at

37 °C for 20 minutes prior to western blotting analysis of ZIP14, ZIP8, and DMT1 or

incubated at 95 °C for 10 min for other proteins. The procedure of western blotting was

performed as previously discussed with the exception of nitrocellulose replacing PVDF

membranes (139). Primary antibodies used were rabbit anti-mDMT1 antibody at a

concentration of 1:1,000 (generously contributed by Dr. Francois Cannone-Hergaux),

rabbit anti-hZIP8 at 1:5,000 concentration (Prestige Antibodies,Sigma Aldrich), rabbit

anti-hZIP14 at 1:5,000 concentration (Prestige Antibodies, Sigma Aldrich), rabbit anti-

CCS at 1:200 (Santa Cruz), mouse anti-Na+/K+-ATPase at 1:200 (Santa Cruz), Goat

anti-ferritin light chain 1:4,000 (Novus Biologicals), mouse anti-α tubulin 1:5,000 (Sigma

Aldrich).

Cell Culture and Treatments

βlox5 cells were cultured in low glucose (1g/L) Dulbecco’s Modified Eagle’s

Medium (Cellgro) containing 100 units/ml penicillin, 100 ug/ml streptomycin, 10% fetal

bovine serum (FBS) (Atlanta Biologicals), 1% Minimum Eagle’s Medium Non-Essential

Amino Acids (Corning), and 15 mM HEPES (Cellgro). βlox5 cells were treated with 100

µM ferric ammonium citrate (MP Biomedicals) and 1 mM ascorbate (EMD Millipore) for

Page 59: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

59

24 h to promote iron loading. Recombinant interleukin 1β (IL-1β) (Peprotech) was

added to media at a concentration of 100 U/ml for specified times. Primary human islets

of at least 90% purity and viability were obtained from the Integrated Islet Distribution

Program (IIDP) and were cultured in Ultra-Low attachment plates (Corning) with

CMRL1066 media containing 10% FBS, 100 units/ml penicillin, and 100 ug/ml

streptomycin. Depletion of cellular iron or iron loading was performed by treating islets

with 50 µM Deferoxamine (Hospira), or 100 µM ferric ammonium citrate (MP

Biomedicals) and 1 mM ascorbate (EMD Millipore), respectively, for 48 h. Prior to

transfection islets were dissociated by incubating in Accutase (Life Technologies) for 15

min at 37 °C followed by pipetting to ensure a single cell suspension. Dissociated islets

were transfected for 48 h. All cells were maintained in 5% CO2 at 37 °C.

In Vitro Glucose Stimulated Insulin Secretion

Triplicate groups of approximately 10 human islets were selected for glucose–

stimulated insulin secretion (GSIS) testing and incubated in low glucose 2.8 mM Kreb’s-

ringer buffer (KRB) containing 25 mM HEPES, 115 mM NaCl, 24 mM NaHCO3, 5 mM

KCl, 1 mM MgCl26H20, .1% BSA, 2.5 mM CaCl2, adjusted to pH 7.4 for 30 m. After

incubation islets were transferred to fresh 2.8 mM glucose KRB and incubated for 1 hr

after which KRB was sampled for determination of basal insulin secretion and replaced

with high-glucose KRB containing 22 mM glucose. Islets were again incubated for 1 hr

before high-glucose KRB was sampled and removed. Insulin concentrations in sampled

KRB were measured by using the ALPCO Ultra-sensitive ELISA KIT (Alpco) and used

to determine total insulin secretion during each time period. Results were normalized to

levels of islet DNA measured by using the Quant-IT PicoGreen dsDNA assay kit (Life

Technologies).

Page 60: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

60

Mouse Islet Isolation

Islets from 10 wk mice used in glucose-stimulated insulin secretion testing were

isolated by using a collagenase infiltration method with Liberase TL (Roche) as

previously detailed (181). In brief, pancreases were incubated at 37°C for 15 m and

islets released by manually agitating the tissues with ice-cold HBSS followed by

centrifugation at 300 g at 4°C to pellet the tissue. Manual agitation and centrifugation

was repeated 5 times. After dissolution of the pancreas, islets were handpicked and

transferred to RPMI1640 medium for culture prior to analysis. Islets from 2-3 mice from

each dietary group were pooled and lysed in RIPA buffer (50 mM Tris-HCl, 1% NP-40,

0.25% Na-deoxycholate, 150 mM NaCl, and 1 mM EDTA, adjusted to pH 7.4) for

western blot analysis. Islet iron status was determined by western blotting for transferrin

receptor by using mouse anti-transferrin receptor (Life Technologies) at a concentration

of 1:4000 ug/ml.

Determination of DMT1, ZIP8, and ZIP14 mRNA Copy Numbers

Total RNA was isolated from primary human islets by using RNAzol (Molecular

Research center) following the manufacturer’s protocol. cDNA synthesis from isolated

RNA was carried out by using the High Capacity cDNA Archive Kit (Life Technologies).

Quantitative RT-PCR was performed by using SYBR Select Master Mix (Life

Technologies) and a CFX96 Real Time PCR Detection System (Bio-Rad). Copy

numbers of DMT1, ZIP8, and ZIP14 were calculated by comparing the Ct values from

human islet cDNA samples to standard curves constructed from known quantities of the

plasmids pBluescriptR-hDMT1 (BC100014; Addgene), pCMV-Sport6-hZIP8 (BC012125;

Open Biosystems), and pCMV-XL4-hZIP14 (BC015770; Open Biosystems). The

primers used to detect DMT1 (F, 5’- TGCATCTTGCTGAAGTATGTCACC-3’ and R, 5’-

Page 61: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

61

CTCCACCATCAGCCACAGGAT-3’), ZIP14 (F, 5’-CAAGTCTGCAGTGGTGTTTG-3’

and R 5’-GTGTCCATGATGATGCTCATTT-3’), and ZIP8 (F, 5’-

CAGTGTGGTATCTCTACAGGATGGA-3’ and R, 5’-CAGTTTGGGCCCCTTCAAA-3’)

were designed to detect all known mRNA transcripts.

siRNA Knockdown of DMT1, ZIP8, and ZIP14

SMARTpool siRNA targeting either human DMT1 or ZIP14 (Thermo Scientific)

and Flexitube siRNA targeting ZIP8 (Qiagen) were used to suppress mRNA levels.

Transfection was performed by using Lipofectamine siRNAiMAX (Life Technologies)

and Opti-MEM Media (Life Technologies) for siRNA and reagent suspension following

the manufacturer’s protocol to yield a final concentration of 12 nM siRNA after addition

to the complex to plated cells. In brief Opti-MEM media was added to separate vials of

either siRNA or Lipofectamine siRNAiMAX after which the contents of each vial were

combined and allowed an incubation period of 15 min. After incubation 500 µL of the

transfection mixture was then added to each well of a 6 well plate containing 2 ml of cell

culture media and cultured for 48 h prior to collection. Successful knockdown was

confirmed by immunoblotting.

Overexpression of DMT1, ZIP8, and ZIP14

Cultured βlox5 cells were transiently transfected with either pcDNA3.1hDMT1-

(1A/+IRE) flag, generously contributed by Dr. Natascha Wolff, pCMV-Sport6-hZip14

(BC015770), pCMV-Sport6-hZIP8 (BC012125), or pCMV-Sport6-empty vector, by using

Effectene Transfection Reagent (Qiagen) according to the manufacture’s protocol. After

24 h cells were harvested for confirmation of overexpression or used in iron uptake

experiments.

Page 62: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

62

Immunofluorescencse

Paraffin-embedded tail sections of human pancreas were obtained through the

Network for Pancreatic Organ Donors with Diabetes (nPOD). Paraffin was cleared with

xylene and tissues rehydrated in stages. After hydration slides underwent heat-induced

epitope retrieval in sodium citrate buffer containing 10 mM sodium citrate, .05% tween

20, and adjusted to pH 6.0 with HCl. Slides were then briefly cooled in distilled H20 and

washed with TBS to remove residual sodium citrate buffer. Washed slides were then

incubated in blocking buffer containing 2% goat serum for 30 min to prevent nonspecific

binding of secondary antibody. During the primary antibody incubation human sections

were triple stained for insulin, glucagon, and either DMT1, ZIP8, or ZIP14 by using

guinea pig anti-insulin (1:200, Abcam), mouse anti-glucagon (1:1,000, Abcam), and

either rabbit anti-DMT1 (1:1,000, Prestige Antibodies Sigma Aldrich), rabbit anti-Zip8

(1:250 Peprotech), and rabbit anti-ZIP14 (1:1,000, Prestige antibodies Sigma Aldrich)

antibodies. During staining for ZIP8 slides were also permeabilized with 5% triton x-100

for 15 min after antigen retrieval. To control for nonspecific primary antibody binding

serial sections were also triple stained with nonimmune rabbit IgG replacing the primary

antibody for DMT1, ZIP8, or ZIP14 at the same concentration. The incubation with

primary antibodies was carried out at 4 degrees C overnight in a humidified chamber.

After the primary incubation slides were washed with TBS and incubated for 2 h at room

temperature with the secondary antibodies goat anti-guinea pig Alexa Flour 594 (1:250,

Life Technologies) for insulin, goat anti-mouse Pacific Blue (1:250, Life Technologies)

for glucagon, and goat anti-rabbit Alexa Flour 488 (1:250, Life Technologies) for either

DMT1, ZIP8, or ZIP14 to obtain a fluorescent signal. After this incubation period slides

were washed with TBS and cover slips carefully mounted. Confocal microscopy was

Page 63: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

63

performed and images obtained by using an Olympus IX2-DSU spinning disk confocal

fluorescent microscope equipped with a Hamamatsu ORCA-AG camera and 3i

SlideBook v4.2 software.

Cellular NTBI Uptake

Cultured βlox5 cells or isolated human islets were washed twice with serum–free

media (SFM) and incubated for 1 h in SFM containing 2% bovine serum albumin to bind

residual transferrin and prevent iron uptake via transferrin-bound iron endocytosis.

Following this incubation period cells were again washed with SFM before the addition

of media containing 2 μM ferric ammonium citrate (MP Biomedicals) radiolabeled with

59Fe and 1 mM ascorbate. Cells were incubated with radiolabeled media for 2 h during

siRNA experiments and for 1 h during overexpression experiments. After the incubation

period media was aspirated and cells were washed 3 times with an iron chelator

solution containing 1 mM diethylenetriaminepentaaetic acid and 1mM

bathophenanthroline disulfonate to remove residual radiolabeled iron. Cells were then

lysed in SDS lysis buffer and the lysate collected measured for radioactivity by using a

WIZARD2 gamma counter (PerkinElmer). Counts for each sample were then

normalized to cellular protein concentrations. To account for variation in 59Fe uptake

between independent experiments, NTBI uptake relative to cellular protein was

measured in separate groups of βlox5 cells and dissociated human islets to establish a

reference level of 59Fe uptake in each independent experiment. Reference levels from

separate independent experiments were compared and used to generate adjustment

ratios by which experimental groups would be multiplied by to produce a control groups

with a relative 59Fe uptake value of 1 while preserving variance in the experimental

control group for statistical analysis.

Page 64: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

64

Generation of Transgenic MIP-Zip14-HA Mice

The transgenic construct previously used to generate mice selectively

overexpressing green fluorescent protein (GFP) in β cells under control of the mouse

insulin 1 promoter, MIP-GFP was generously contributed by Drs. Graeme Bell and

Manami Hara (University of Chicago, Chicago, IL) and has been detailed previously

(182). In brief the construct consisted of a fragment of the mouse insulin 1 promoter,

the GFP coding region, and an intron region of the human growth hormone gene to

enhance expression. The region of this vector encoding GFP was excised by restriction

digestion with Xho1 and hemagglutinin antigen (HA) tagged Zip14 was subcloned into

the MIP vector, resulting in the generation of a MIP-ZIP14-HA construct. The MIP-

Zip14-HA-hGH fragment was isolated from the vector backbone by restriction digestion

with HindIII and SfiI followed by agarose gel separation, extraction, and purification.

DNA was introduced into fertilized embryos from C57BL/6J mice by pronuclear

microinjection carried out at the Mouse Models Core at the University of Florida.

Founders were screened based on the presence of HA tag using the primers 5’

TACCCTTACGACGTGCCT 3’ and 5’ AGGAGAGAGGCCAGGTTAAT 3’ to differentiate

between endogenous Zip14 and successful transgene insertion. Transgene copy

numbers were determined by the comparison of genomic DNA to a standard curve

constructed from known amounts of MIP-Zip14-HA plasmid DNA. The primer set used

was 5’ CCTTACGACGTGCCTGATTA 3’ and 5’ TTCAGCTGTGTCAGGGTAAG 3’,

targeting HA tag. Founders were bred with wild-type C57BL/6J mice to establish

transgenic lines. Overexpression of Zip14-HA in β cells was confirmed by

immunofluorescence detection of HA tag in mouse pancreatic sections (Mouse anti-HA

Page 65: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

65

1:100, Roche) following the protocol previously detailed for IF in human pancreatic

sections.

Statistical Analysis

Data were analyzed for statistical significance by using one-way ANOVA and

Tukey’s multiple comparison post-hoc test or Student’s T-test (GraphPad Prism) where

indicated. Unequal variance between groups was accounted for by log transformation,

where applicable, to normalize variance before statistical analysis. Survival curves were

analyzed for significance by using the Gehan-Breslow-Wilcoxon Test (Graphpad Prism).

Page 66: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

66

CHAPTER 4 TRANSCRIPTIONAL PROFILING OF PANCREATIC GENE EXPRESSION IN

RESPONSE TO DIETARY IRON LOADING OR DEFICIENCY1

The association between excess iron and pancreatic dysfunction has long been

observed in the iron overload disorder hereditary hemochromatosis (183). Patients with

hemochromatosis have a higher prevalence of diabetes, decreased insulin secretory

capacity, and impaired glucose tolerance relative to the normal population (45). The Hfe

knockout mouse, the animal model of hemochromatosis, also displays alterations in

pancreatic function, including decreased insulin secretory capacity (146). In humans,

insulin secretory capacity and glucose tolerance improves after iron stores are

normalized by phlebotomy, suggesting that tissue iron levels are an important

determinant of insulin action (184). Consistent with this idea are animal studies

showing that a decrease in iron stores (in response to phlebotomy or a low-iron diet)

can increase insulin secretion and pancreatic insulin levels (163, 164). However, iron

depletion to the point of iron deficiency and anemia has been shown to negatively affect

glucose homeostasis by increasing blood glucose concentrations (185).

The effects of iron overload and deficiency on glucose homeostasis are likely

mediated, at least in part, by iron-related changes in the expression of genes involved in

glucose metabolism. For example, iron deficiency has been reported to be associated

with higher levels of rate-limiting gluconeogenic enzymes in rat liver (186) and iron-

loaded Hfe knockout mice display increased glucose uptake by isolated soleus muscle

1Reprinted with permission from Coffey R, Nam H, Knutson MD. Microarray analysis of rat pancreas

reveals altered expression of Alox15 and regenerating islet-derived genes in response to iron deficiency and overload. PLoS One. 2014;9:e86019.

Page 67: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

67

and decreased glucose oxidation by isolated hepatic mitochondria (161, 187). Little

information, however, exists regarding iron-related gene expression in the pancreas.

Given that the pancreas hormonally controls whole-body glucose homeostasis, the aim

of the present study was to examine global changes in pancreatic gene expression in

response to iron deficiency and overload. Identification of pancreatic genes that are

regulated by iron status may offer insight not only into how iron status perturbs glucose

homeostasis, but also how iron overload may contribute to β cell destruction and

diabetes.

Results

Body Weight, Iron Status, and Blood Glucose Concentrations

After 3 weeks of feeding the experimental diets, body weights were significantly

lower in the FeD and FeO groups relative to FeA controls, but did not differ between

FeD and FeO animals (Table 4-1). Liver non-heme iron concentrations, an indicator of

body iron stores, confirmed that rats fed the FeD diet became iron deficient whereas

rats fed the FeO diet became iron overloaded. In FeO animals, liver non-heme iron

concentrations were nearly 40 times higher than controls. FeD rats also became

anemic with hemoglobin levels that were 41% lower than normal (Table 4-1). Blood

glucose concentrations were elevated in FeD rats compared with FeA controls, whereas

those in FeO animals did not differ from controls (Table 4-1).

Pancreatic Mineral Concentrations

In FeO rats, pancreatic iron concentrations were 155% higher than those in FeA

animals, whereas in FeD rats, iron concentrations were 40% lower than controls (Table

4-2). Given that iron deficiency and overload can affect tissue concentrations of other

trace minerals (176), I measured pancreatic concentrations of zinc, manganese, copper,

Page 68: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

68

and cobalt (Table 4-2). Pancreatic zinc concentrations were found to be 26% higher in

FeD rats, and copper concentrations were 74% lower in FeO rats, when compared with

FeA controls. By contrast, pancreatic manganese and cobalt concentrations did not

differ among groups. It should be noted that the concentrations of zinc, manganese,

copper, and cobalt did not differ among the experimental diets (data not shown).

Identification and Classification of Differentially Expressed Genes by Microarray Analysis

Microarray analysis was used to identify candidate genes that are differentially

expressed in FeD and FeO pancreas, especially those that may influence the risk for

diabetes. Using a log2 fold change of ± 2 and P < 0.05 as a cutoff, I identified a total of

230 genes as differentially expressed in FeD and FeO pancreas relative to FeA

pancreas. In FeD pancreas 66 genes were differentially expressed (56 down-regulated

and 10 up-regulated) (Figure 4-1A). In FeO pancreas 164 genes were differentially

expressed (82 down-regulated and 82 up-regulated) (Figure 4-1B). The differentially

expressed genes were analyzed by using DAVID bioinformatics resources to identify

gene ontology categories. In FeD pancreas, the category with the highest number of

genes was “lipid transport” (7 genes), followed by “antimicrobial” (4 genes),

“neuropeptide” (4 genes), and “pancreatitis-associated protein” (3 genes) (Figure 4-1A).

All but two of the genes in these categories were down-regulated in FeD pancreas. By

contrast, in FeO pancreas, most gene ontology categories were enriched with up-

regulated genes (Figure 4-1B). For example, 6 of 8 genes were up-regulated in the

“pattern binding” category in FeO pancreas. Of note, the gene ontology category

“pancreatitis-associated protein” was identified in both FeD and FeO pancreas. Lists of

the genes in each category along with fold change are provided in Table 4-6 and Table

Page 69: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

69

4-7. The top 10 most up-regulated and down-regulated genes in FeD and FeO

pancreas, ordered by mean magnitude change (P< 0.05), are shown in Table 4-3 and

Table 4-4. The genes listed in Table 4-6, Table 4-7, Table 4-3, and Table 4-4 were

surveyed in the literature to identify those with reported associations with diabetes

and/or glucose homeostasis, and several of these were subsequently selected for

validation by qRT-PCR of individual rat samples (n=6 group).

Confirmation of Up-Regulation of Alox15 Expression by QRT-PCR and Western Blotting

According to the microarray analysis, the most up-regulated gene in FeD

pancreas was Alox15 (arachidonate 15-lipoxygenase) (Table 4-3). Alox15 catalyzes the

oxidation of polyunsaturated fatty acids, such as arachidonic acid, during the formation

of inflammatory mediators and has been linked to the development of type 1 diabetes

(188, 189). QRT-PCR analysis confirmed the up-regulation of Alox15 mRNA levels in

FeD pancreas (Figure 4-2A), and western blot analysis revealed higher Alox15 protein

levels in FeD pancreas (Figure 4-2B). Alox15 protein levels were also found to be

higher in FeO pancreas compared with FeA controls despite no increase in Alox15

mRNA levels. As western blotting controls for rat Alox15, jejunum samples from iron-

adequate (JA) and iron-deficient (JD) rats were analyzed in parallel with the rat

pancreas samples. Consistent with a previous study by Collins et al. (190), Alox15 was

detected at approximately 70 kDa and was markedly up-regulated in iron-deficient

jejunum (JD) (Figure 4-2B). Densitometric analysis of the western blots indicated that

Alox15 protein levels were approximately 8- and 9-fold higher (P<0.001) in FeD and

FeO pancreas, respectively, compared with FeA controls (n=6/group; data not shown).

Page 70: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

70

Confirmation of Reg Family mRNA Levels by QRT-PCR

Of the genes showing positive regulation during FeO, the most elevated

belonged to the Reg family of regenerating islet-derived genes (Table 4-4). The genes

of the Reg family, most notably Reg1a, have been linked to pancreatic regeneration as

well as cellular growth and survival during oxidative stress (191-194). Consistent with

the microarray data, qRT-PCR analysis revealed that mRNA levels of these genes were

up-regulated in FeO pancreas. Mean mRNA levels of Reg1a, Reg3a, and Reg3b were

found to be 21, 37, and 18 times higher, respectively, in FeO pancreas than FeA

controls (Figure 4-3). Also consistent with the microarray, qRT-PCR analysis found that

Reg1a mRNA levels were up-regulated in FeD pancreas. Reg mRNA levels varied

considerably among rats, particularly in the FeO and FeD groups in which two or three

values were notably higher than the others. Repeated analyses confirmed that the high

values do not represent analytical artifacts. In the FeD, FeA, and FeO groups, the high

values for Reg1a and Reg3a (but not Reg3b) mRNA are from the same animals,

suggesting that Reg1a and Reg3a are up-regulated in parallel.

Discrepancies Between Microarray and QRT-PCR Analysis Results

According to the microarray, both FeD and FeO pancreas showed large down-

regulations in the expression of Fabp1, Fabp2, and Apoa1 (Tables 3 and 4), which

clustered in the gene ontology category of “lipid transport” (Table S3). As all of these

genes have been associated with diabetes in the surveyed literature (195-197) they

were selected for follow-up study. However, the expression levels of these genes in

pancreas were found to be below the detection limit of qRT-PCR, similar to previous

studies that have failed to detect the expression of these genes in pancreas (198, 199).

Page 71: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

71

Discussion

Because iron status can affect glucose homeostasis, I sought to identify glucose

metabolism-related genes in rat pancreas whose expression might be affected by iron

deficiency or overload. Unexpectedly, our microarray data, and subsequent functional

enrichment analysis by DAVID, did not identify any changes in the expression of genes

known to be involved in glucose metabolism. A limitation to our study is that many of the

glucose-responsive genes are found in islet cells (200), which constitute only 1-2% of

the mass of the pancreas, and therefore changes in islet-cell gene expression may not

be readily detectable in whole pancreas tissue. The most notable finding from our

microarray analyses was the identification of differentially expressed genes that are

associated with diabetes and/or pancreatic stress. More specifically, Alox15 was

identified as the most up-regulated mRNA in iron deficiency, and Reg family transcripts

Reg1a, Reg3a, and Reg3b, were found to be markedly up-regulated in iron overload.

Alox15 encodes arachidonate 15-lipoxygenase, a non-heme iron-containing

enzyme that catalyzes the oxygenation of polyunsaturated fatty acids to form

inflammatory mediators (201). Despite the name suggesting 15-lipoxygenase activity,

Alox15 in rodents has been demonstrated to function primarily as a 12-lipoxygenase

with secondary 15-lipoxygenase function (202). Therefore the term leukocyte 12-

lipoxgenase, as well as the hybrid term 12/15-lipoxygenase, is often used in reference

towards Alox15. A link between iron deficiency and Alox15 was first reported in a

microarray study by Collins et al. (190), who identified Alox15 as the most strongly

induced gene in the intestine of iron-deficient rats. Alox15 has also been identified as

the most highly induced gene in microarray studies of iron-deficient rat liver (186) and

brain (203). Similar to Collins et al. (190), I found that elevated Alox15 mRNA levels

Page 72: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

72

were associated with higher Alox15 protein levels. Protein levels of Alox15 were also

found to be elevated in iron-loaded rat pancreas, despite no up-regulation of Alox15

mRNA levels, suggesting post-transcriptional regulation under iron-overload conditions.

In the pancreas, Alox15 is present in β cells (204) where it appears to play a role

in the pathogenesis of diabetes. Genetic deletion of a locus containing Alox15 has

been shown to protect nonobese diabetic (NOD) mice from developing autoimmune

diabetes, with knockout mice exhibiting superior islet mass and glucose tolerance (188).

Recent experiments using siRNA against Alox15 provide evidence that diminished

Alox15 levels are responsible for the protective phenotype (189). Resistance to the

development of a diabetic phenotype induced via streptozotocin was also observed in

mice lacking Alox15 (205). It has been proposed that Alox15 contributes to the

development of diabetes via its ability to catalyze the formation of inflammatory

mediators such as 12-HETE (hydroxyeicosatetraenoic acid), which causes β cell

dysfunction and death (204, 206, 207), recently linked to excessive production of

reactive oxygen species (208). Our observation that iron deficiency causes a marked

elevation in Alox15 mRNA and protein levels in the pancreas raises the possibility that

iron deficiency—in addition to iron overload—may increase the risk of developing

diabetes through up-regulation of Alox15. Such a possibility appears opposite to recent

studies showing a protective effect of iron restriction on diabetes risk. For example,

Cooksey et al. (163) observed that an iron-restricted diet enhanced β cell function and

insulin sensitivity in the ob/ob mouse model of type 2 diabetes. Similarly, Minamiyama

et al. (164) found that feeding an iron-restricted diet to type 2 diabetic rats normalized

Page 73: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

73

plasma insulin levels. It should be noted, however, that in the study by Cooksey et al.

(163), iron-restriction did not result in iron deficiency or anemia in contrast to our study.

Although it is well known that individuals with iron overload are susceptible to

developing diabetes (183), the molecular mechanisms involved remain poorly

understood. Our observation that iron-overloaded rats have highly elevated Alox15

protein levels in the pancreas suggests that Alox15 may contribute to β cell loss and β

cell dysfunction in iron overload. Indeed, the pancreases of iron-loaded rats appear to

be under stress as indicated by the elevated expression of the regenerating islet-

derived gene family members Reg1a, Reg3a, and Reg3b. As indicated by their name,

Reg genes were first identified by their strong induction in regenerating pancreatic islets

in response to stress/damage (209). Reg1a is a 165-a.a secreted protein that has been

shown to play an important role in β cell function in vivo (191). Disruption of murine

Reg1 (the ortholog of rat Reg1a) resulted in decreased proliferative capacity of

pancreatic β cells (210), whereas administration of recombinant rat Reg1a resulted in β

cell regeneration and reversal of diabetes in rats after surgical resection of 90% of the

pancreas (191). Similar to Reg1, Reg3a and Reg3b have been associated with islet

regeneration and protection against diabetes (193, 211). Reg3 proteins are also known

as pancreatitis-associated proteins (PAP) that become highly expressed in acinar cells

in response to injury (212). Our observation of elevated Reg3 expression in iron-loaded

rat pancreas is consistent with a previous report of hypotransferrinemic mice, which

displayed pancreatic iron loading and markedly elevated expression of Reg3 mRNA

(213). However, in that study, a time course analysis of pancreatic iron loading

indicated that Reg3/PAP mRNA became detectable only when pancreatic non-heme

Page 74: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

74

iron concentrations had reached levels that were ~50 times normal. In our study of iron-

loaded rats, I found that even modest elevations in pancreatic iron concentrations (2.5

times normal) are associated with enhanced expression of Reg3 mRNA, suggesting

that Reg mRNA levels could serve as an early biomarker of iron-related pancreatic

stress/damage in rats. The apparent discrepancy in pancreatic iron load required to

elicit increased Reg3 expression between mice and rats is likely attributable to

interspecies variability. Mice are largely resistant to the degenerative effects of

pancreatic iron loading whereas rats exhibit acinar cell degradation, indicative of

pancreatic damage, following dietary iron overload (136, 142). One caveat is that the

elevated pancreatic Reg expression in iron-loaded rats could be confounded by the

abnormally low (i.e., ~25% of normal) copper concentrations in these animals. Copper

deficiency in rats has been shown to result in pronounced atrophy of the exocrine

pancreas (214). Pancreatic atrophy is observed during pancreatitis, a state which

promotes extensive expression of Reg family genes (215). Also, during copper

deficiency islet hyperplasia and β cell neogenesis have been documented (216) in line

with the islet-regenerating properties of Reg proteins. More research is needed to

determine if low copper levels induce the expression of these genes.

In conclusion, microarray analysis of rat pancreas has revealed that iron

deficiency and overload increase the expression of one or more genes strongly

associated with diabetes and pancreatic stress, thus highlighting the importance of iron

status in the pancreas.

Page 75: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

75

Table 4-1. Body weight, iron status, and blood glucose concentration of rats

Group Body weight

(g) Liver non-heme iron

(μg/g) Hemoglobin

(g/dL) Glucose (mg/dL)

FeD 193.3 ± 20.2a 3.5 ± 3.6a 7.5 ± 2.2a 154.5 ± 39.0b

FeA 224.3 ± 11.1b 25.4 ±17.7b 12.8 ± 0.4b 99.0 ± 16.0a

FeO 170.3 ± 23.6a 980.6 ± 310.2c 13.6 ± 0.6b 117.7 ± 18.5a

FeD, iron deficient; FeA, iron adequate; FeO, iron overloaded. Values represent means ± SD, n = 6. Means without a common superscript are significantly different P< 0.05

Page 76: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

76

Table 4-2. Pancreatic mineral concentrations

Mineral concentrations (μg/g dry weight) were measured by using ICP-MS. Values represent means ± SD, n = 6 Means without a common superscript are significantly different P<0.05

Group Iron Zinc Manganese Copper Cobalt

FeD 38.2 ± 5.7a 107.5 ± 18.0b 8.7 ± 1.9 4.7 ± 1.0b 0.05 ± 0.02

FeA 63.7 ± 14.3b 85.0 ± 16.0a 6.2 ± 1.9 3.8 ± 0.7b 0.03 ± 0.01

FeO 162.7 ± 59.5c 75.2 ± 8.5a 7.0 ± 1.2 1.0 ± 0.0a 0.04 ± 0.01

Page 77: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

77

Table 4-3. Top 10 most up-regulated and down-regulated genes in FeD pancreata

Gene Name Symbol Accession Fold changea

arachidonate 15-lipoxygenase Alox15 NM_031010 4.1

L-threonine dehydrogenase Tdh NM_001106044 3.3

RT1 class I, locus CE5 RT1-CE5 NM_001008843 3.3

S100 calcium binding protein A9* S100a9 NM_053587 2.7 transient receptor potential cation channel, subfamily C, member 3

Trpc3 NM_021771 2.4

vascular endothelial growth factor B Vegfb NM_053549 2.4

regenerating islet-derived 1 alpha* Reg1a NM_012641 2.2

secretoglobin, family 2A, member 1 Scgb2a1 NM_080770 2.2

potassium intermediate/small conductance Ca-activated channel, subfamily N, member 1

Kcnn1 NM_019313 2.0

alanine-glyoxylate aminotransferase 2 Agxt2 NM_031835 1.9

fatty acid binding protein 1, liver* Fabp1 NM_012556 -6.1

fatty acid binding protein 2, intestinal* Fabp2 NM_013068 -6.0

LOC494499 protein LOC494499 NM_001010921 -5.0

proline-rich acidic protein 1 Prap1 NM_031669 -5.0

s100 calcium binding protein G S100g NM_012521 -4.9

monoacylglycerol O-acyltransferase 2*

Mogat2 NM_001109436 -4.9

apolipoprotein A-I* Apoa1 NM_012738 -4.6

cAMP responsive element binding protein 3-like 3

Creb3l3 NM_001012115 -4.5

similar to carboxylesterase 5 LOC679368 XM_001056053 -4.3

carboxylesterase 5-like LOC688542 XR_086144 -4.2

Fold change log2 relative to iron-adequate rat pancreas.* In gene ontology category in Figure 2-1 and supplemental Table S2.

Page 78: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

78

Table 4-4. Top 10 most up-regulated and down-regulated genes in FeO pancreata

Gene name Symbol Accession Fold changea

regenerating islet-derived 3 alpha* Reg3a NM_172077 4.8

regenerating islet-derived 3 beta* Reg3b NM_053289 4.3

extracellular proteinase inhibitor Expi NM_133537 4.3

regenerating islet-derived 1 alpha* Reg1a NM_012641 4.2

prepronociceptin Pnoc NM_013007 3.8

beta-galactosidase-like protein Bin2a NM_001009524 3.6

calmodulin-like 3 Calml3 NM_001012054 3.5

vascular endothelial growth factor B* Vegfb NM_053549 3.5

phospholipase A2, group IIA* Pla2g2a NM_031598 3.5

upper zone of growth plate and cartilage matrix associated

Ucma NM_001106121 3.2

similar to Robo-1 LOC691352 NM_001109638 -8.7

fatty acid binding protein 2, intestinal * Fabp2 NM_013068 -7.0

fatty acid binding protein 1, liver* Fabp1 NM_012556 -7.0

proline-rich acidic protein 1 Prap1 NM_031669 -6.6

lectin, galactoside-binding, soluble, 4 Lgals4 NM_012975 -6.0

apolipoprotein A-I * Apoa1 NM_012738 -5.8

hydroxysteroid (17-beta) dehydrogenase 2

Hsd17b2 NM_024391 -5.6

S100 calcium binding protein G S100g NM_012521 -5.6

LOC494499 protein* LOC494499 NM_001010921 -5.4

hypothetical protein LOC691259 LOC691259 NM_001109632 -5.2

Fold change log2 relative to iron-adequate rat pancreas. *In gene ontology category in Figure 1 and supplemental Table S3.

Page 79: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

79

Table 4-5. Primers for qRT-PCR

Symbol Name GenBank Accession No.

Forward primer (5'-3')

Reverse primer (5'-3')

Alox15 arachidonate 15-lipoxygenase

NM_031010 CCCTGTCGGGACTCGGAAGC

CCAGTGCCCTCAGGGAGGCT

Reg1a regenerating islet-derived 1 alpha

NM_012641 TTGTCTCAGCCTGCAGAGATTG

CATGATGAGCAGCAGACTGTCTT

Reg3a regenerating islet-derived 3 alpha

NM_172077 CCGTGGTAACTGTGGCAGTCT

GTGATGGTCTCCCCACTTCAG

Reg3b regenerating islet-derived 3 beta

NM_053289 AAAGATGATGAGAGTTAAGATGTTGCA

AGCAGCATCCAGGACATGACT

Fabp1 fatty acid binding protein 1, liver

NM_012556 AGGTCAAGGCAGTGGTTAAGAT

TGTCATGGTATTGGTGATTGTGT

Fabp2 fatty acid binding protein 2, intestinal

NM_013068 TCACTGGGACCTGGACCATG

CATATGTGTAGGTCTGGATTAGT

Apoa1 apolipoprotein A-I

NM_012738 TCCACTTTGGGCAAACAGCTGAAC

TCCTGTAGGCGACCAACAGTTGAA

Page 80: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

80

Table 4-6. Functional categories of pancreatic genes differentially expressed in response to iron deficiency

Functional category Gene symbol Description GenBank number Fold change

Lipid transport Npc1l1 NPC1 (Niemann-Pick disease, type C1, gene)-like 1

NM_001002025 -2.27

Mttp microsomal triglyceride transfer protein

NM_001107727 -2.97

Apoc3 apolipoprotein C-III NM_012501 -3.19

Apoa1 apolipoprotein A-I NM_012738 -4.57

Mogat2 monoacylglycerol O-acyltransferase 2

NM_001109436 -4.88

Fabp2 fatty acid binding protein 2, intestinal

NM_013068 -5.96

Fabp1 fatty acid binding protein 1, liver

NM_012556 -6.10

Antimicrobial S100a9 S100 calcium binding protein A9

NM_053587 2.69

Defa6 defensin alpha 6 NM_001033076 -2.71

Defa-rs1 defensin alpha-related sequence 1

NM_001033073 -2.82

Defa8 defensin alpha 8 NM_001033077 -3.11

Neuropeptide Calca calcitonin-related polypeptide alpha

NM_017338 -2.08

Cartpt CART prepropeptide NM_017110 -2.48

Gal galanin prepropeptide

NM_033237 -3.08

Vip vasoactive intestinal peptide

NM_053991 -3.51

Pancreatitis-associated protein

Reg1a regenerating islet-derived 1 alpha

NM_012641 2.24

Reg3g regenerating islet-derived 3 gamma

NM_173097 -2.48

Reg4 regenerating islet-derived family, member 4

NM_001004096 -3.08

Fold change log2 relative to iron-adequate rat pancreas

Page 81: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

81

Table 4-7. Functional categories of pancreatic genes differentially expressed in

response to iron overload

Functional category Gene symbol Description Genbank number Fold change

Pattern binding Vegfb vascular endothelial growth factor B

NM_053549 3.45

pla2g5 phospholipase A2, group V NM_017174 3.11

Itgam integrin, alpha M NM_012711 2.65

Ccl7 chemokine (C-C motif) ligand 7

NM_001007612 2.65

Prg4 proteoglycan 4, (megakaryocyte stimulating factor, articular superficial zone protein, camptodactyly, arthropathy, coxa vara, pericarditis syndrome)

NM_001105962 2.14

Tpsb2 tryptase beta 2 NM_019180 2.07

Abp1 amiloride binding protein 1 (amine oxidase, copper-containing)

NM_022935 -2.29

Colq collagen-like tail subunit (single strand of homotrimer) of asymmetric acetylcholinesterase

NM_019274 -2.42

Glutathione and drug metabolism

Cyp2e1 cytochrome P450, family 2, subfamily e, polypeptide 1

NM_031543 3.16

Aox1 aldehyde oxidase 1 NM_019363 3.10

Fmo1 flavin containing monooxygenase 1

NM_012792 2.72

Gpx2 glutathione peroxidase 2 NM_183403 -2.61

Gsta5 glutathione S-transferase Yc2 subunit

NM_001159739 -3.57

Gsta2 glutathione S-transferase A2 NM_017013 -4.55

Loc494499 LOC494499 protein NM_001010921 -5.44

Pancreatitis-associated protein

Reg3a regenerating islet-derived 3 alpha

NM_172077 4.83

Reg3b regenerating islet-derived 3 beta

NM_053289 4.29

Reg1a regenerating islet-derived 1 alpha

NM_012641 4.18

Reg3g regenerating islet-derived 3 gamma

NM_173097 -2.26

Reg4 regenerating islet-derived family, member 4

NM_001004096 -4.38

Page 82: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

82

Table 4-7. Continued

Functional category Gene symbol Description Genbank number Fold change

Digestive system process

Tff1 trefoil factor 1 NM_057129 -3.86

Mogat2 monoacylglycerol O-acyltransferase 2

NM_001109436 -3.99

Fabp1 fatty acid binding protein 1, liver

NM_012556 -7.00

Fabp2 fatty acid binding protein 2, intestinal

NM_013068 -7.01

Defensin propeptide

Defa24 defensin, alpha, 24 NM_001013053 -2.57

Defa-rs1 defensin alpha-related sequence 1

NM_001033073 -3.14

Defa8 defensin alpha 8 NM_001033077 -3.54

Regulation of lipid transport

Adipoq adiponectin, C1Q and collagen domain containing

NM_144744 2.22

Apoc3 apolipoprotein C-III NM_012501 -2.19

Apoa1 apolipoprotein A-I NM_012738 -5.77

Protease activity Cma1 chymase 1, mast cell NM_013092 2.58

Mcpt1l3 mast cell protease 1-like 4

ENSRNOT00000043182 2.32

Tmprss8 transmembrane protease, serine 8 (intestinal)

NM_199371 -2.02

Spink4 serine peptidase inhibitor, Kazal type 4

NM_001008871 -2.48

Mep1b meprin 1 beta NM_013183 -3.81

Mmp7 matrix metallopeptidase 7

NM_012864 -5.10

Complement activation

Cfd complement factor D (adipsin)

NM_001077642 2.37

C4bpa complement component 4 binding protein, alpha

NM_012516 2.42

C6 complement component 6

NM_176074 3.05

Fold change Log2 relative to iron-adequate rat pancreas

Page 83: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

83

Figure 4-1. Functional classification of pancreatic genes up- or down-regulated in

response to iron deficiency and iron overload. Microarray analysis identified a total of 66 differentially expressed genes in response to iron deficiency (Panel A) and 164 genes in response to iron overload (Panel B). Genes were then subjected to DAVID analysis to identify functional categories. A) Functional gene categories identified in iron-deficient pancreas and the number of genes in each category. B) Functional gene categories identified in iron-overloaded pancreas and the number of genes in each category.

Page 84: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

84

Figure 4-2. Effect of iron deficiency and overload on rat pancreatic Alox15 expression.

A) Total RNA was isolated from rat pancreas and the relative transcript abundance of Alox15 was measured by using qRT-PCR. Transcript abundances were normalized to the housekeeping transcript cyclophilin B and are expressed relative to the FeA group mean (set to 1). B) Immunoblot analysis of Alox15 from a representative sample of FeD, FeA, and FeO rats. Jejunum from iron-adequate (JA) and iron-deficient (JD) rats were analyzed in parallel to serve as negative and positive controls respectively for immunodetection of Alox15. The blot was stripped and reprobed for tubulin to indicate protein loading among lanes. Values are expressed as the mean ± SEM, n=6. Asterisks indicate a significant difference relative to FeA controls, **P<0.01.

Page 85: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

85

Figure 4-3. Effect of iron deficiency and overload on the expression of pancreatic Reg family genes.Total RNA was isolated from rat pancreas and the relative transcript abundances of Reg family genes were determined by qRT-PCR. Transcript abundances were normalized to levels of cyclophilin B and are expressed relative to the FeA group mean (set to 1). Statistical significance was determined by one-way ANOVA. Asterisks indicate a significant difference relative to FeA controls *P<0.05, **P<0.01, ***P<0.001.

Page 86: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

86

CHAPTER 5 MECHANISMS OF NTBI UPTAKE BY HUMAN β CELLS

Iron is an essential trace mineral necessary for numerous biological functions,

including oxidation-reduction reactions, due, in part, to the ability of iron to exist in

multiple oxidation states. While these reactions are required for normal physiologic

processes, iron can also catalyze the generation of hydroxyl radicals, which can

damage lipids, protein, and DNA (217). Due to the duality of iron redox chemistry, iron

transport and homeostasis are tightly regulated in vivo to prevent the production of

reactive oxygen species by free iron. However in genetic disorders such as

hemochromatosis, in which excessive amounts of dietary iron are absorbed, or β-

thalassemia major, which requires blood transfusions, excess iron overwhelms the

normal mechanisms of iron transport. One such consequence is the appearance of

plasma non-transferrin-bound iron (NTBI), a form of iron that appears when the carrying

capacity of transferrin, the circulating iron transport protein, becomes exceeded. The

exact chemical nature of NTBI in the plasma is not known, but is thought to consist

mainly of ferric citrate and other low-molecular-weight iron species (218, 219). Although

it is generally believed that NTBI is a pathologic species that appears only when

transferrin saturation exceeds 75% (220), plasma NTBI has been reported to be

commonly present in diabetics with transferrin saturations below 60% (131).

Studies in mice have shown that plasma NTBI is rapidly cleared mostly by the liver, and

to a lesser extent, the pancreas, kidney, and heart (73, 74, 83). Accordingly, NTBI is a

major contributor to iron loading of the liver and other tissues in iron overload disorders.

In the liver and pancreas, NTBI is taken up mainly by hepatocytes and acinar cells via

Page 87: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

87

ZRT/IRT-Like Protein 14, ZIP14 (SLC39A14) (7). How NTBI is taken up by the kidney,

heart, and other organs/cell types remains to be established.

Studies of iron-loaded human pancreas have revealed that iron not only accumulates in

acinar cells, but also in β cells of the islets (132-134). Iron loading of the β cell has

been proposed to contribute to the well-known β cell dysfunction and diabetes in

individuals with clinical iron overload (85, 133). Given the known role of NTBI uptake to

iron loading of various organs and cells, we hypothesize that human β cells are able to

take up NTBI. The aim of the present study was to examine the potential roles of the

transmembrane transporters DMT1 (divalent metal-ion transporter 1), ZIP14, and ZIP8

in NTBI uptake by human β cells. We focused on these three transporters because of

their well-documented roles in NTBI uptake/iron metabolism (7, 11, 13, 28, 90), and in

the case of DMT1 and ZIP8, also because DMT1 has been reported to be expressed in

human islets and ZIP8 has been reported in rat β cells (85, 103).

Results

Overexpression of NTBI Transporters in Human β Cells

To determine whether the expression of established NTBI transporters could

promote iron uptake in β cells, ZIP14, ZIP8, and DMT1 were overexpressed in βlox5

cells, a human β cell line (221), and NTBI uptake was measured. NTBI uptake was

assessed at pH 7.4, the pH of blood plasma. I found that overexpression of ZIP14 or

ZIP8, but not DMT1, increased the ability of βlox5 cells to take up NTBI when compared

with cells transfected with empty vector control (Figure 5-1). To explore the possibility

that the lack of DMT1-mediated NTBI transport in βlox5 cells results from poor DMT1

expression at the cell surface, I isolated cell-surface proteins from cells overexpressing

DMT1. Western blotting analysis of total-cell lysate and isolated cell-surface proteins

Page 88: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

88

revealed that the majority of DMT1 was indeed intracellular with little expression at the

cell surface, thus potentially accounting for the lack of additional NTBI uptake during

DMT1 overexpression (Figure 5-2A). In contrast to DMT1, overexpressed ZIP14 was

enriched at the cell surface (Figure 5-2B). The proteins copper chaperone for

superoxide dismutase (CCS) and Na+/K+ ATPase were measured to indicate

intracellular and cell-surface protein fractions, respectively.

siRNA Knockdown of NTBI Transporters in Human β Cells

To define the contribution of endogenous NTBI transporter expression to iron

uptake by human β cells, siRNA was used to suppress the expression of ZIP14, ZIP8,

and DMT1 in βlox5 cells. siRNA-mediated suppression of ZIP14 expression decreased

cellular iron uptake by approximately 50% (Figure 5-3A). By contrast, siRNA knockdown

of ZIP8 did not affect iron uptake (Figure 5-3B), suggesting that endogenous ZIP8-

mediated NTBI uptake is negligible in βlox5 cells. I was unable to achieve successful

knockdown of DMT1 in this cell line because the cells died shortly after transfection.

Interestingly, cell death could be prevented by supplementing the cell culture medium

with 50 µM ferric ammonium citrate (FAC), suggesting that decreased cellular viability

was related to cellular iron deficiency (data not shown).

Similar to βlox5 cells (Figure 5-3A), knockdown of ZIP14 in primary human islets

decreased NTBI uptake by approximately 50% (Figure 5-4), suggesting that ZIP14 is a

major route of NTBI uptake in human β cells. Analysis of mRNA copy numbers in

human primary islets (Figure 5-7) indicates that the number of mRNA transcripts

encoding ZIP14 is approximately 2 and 4 times the number of ZIP8 and DMT1

transcripts, respectively (Figure 5-7).

Page 89: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

89

Cellular Localization of NTBI Transporters in Human Islets

As pancreatic islets represent a non-homogenous population of cells, consisting

primarily of β and α cells, my methods using whole islets are unable to discern the

contribution of individual cell types to mRNA expression and iron uptake. Therefore,

immunofluorescence analysis was used to determine if ZIP14, ZIP8, and DMT1 are

expressed at the protein level in β cells or in other cells comprising pancreatic islets. In

the case of ZIP14 I found that protein expression is largely restricted to β cells with

negligible expression in α cells (Figure 5-5). ZIP14 staining in β cells displayed a diffuse

speckled pattern throughout the cytosol (Figure 5-5B).

Staining for DMT1 in the human pancreas indicated that its expression was

restricted to β cells with no signal detected from α cells (Figure 5-8A). DMT1 displayed

a punctate, granular staining pattern suggesting an intracellular localization, consistent

with the known role of DMT1 in endosomal iron transport in some cell types (26).

Staining for ZIP8 in the human pancreas revealed only low-level diffuse staining in

pancreatic acinar cells. No islet staining was observed beyond non-specific levels

detected with non-immuned IgG substituted for anti-ZIP8 primary antibody (Figure 5-

8B).

Modulation of ZIP14 Expression by Iron in Human β Cells

Previous reports have indicated that ZIP14 protein levels are modulated by

cellular iron status. For example, in human hepatoma HepG2 cells, ZIP14 protein levels

are induced by iron loading with ferric ammonium citrate (FAC) (82, 95). ZIP14 protein

levels are also elevated in iron-loaded rat liver and pancreas (82). To determine if ZIP14

levels are induced by iron loading in human β cells, I treated βlox5 cells and primary

human islets with FAC and measured ZIP14 protein expression. I found that cellular iron

Page 90: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

90

loading, confirmed by elevated ferritin protein levels, increased ZIP14 protein

expression in βlox5 cells (Figure 5-6A) but not primary islets (Figure 5-6B). Depletion of

cellular iron levels by the iron chelator desferrioxamine (DFO) has been documented to

decrease ZIP14 protein levels in HepG2 cells (95). However in primary human islets

treated with DFO, I detected no difference in ZIP14 protein levels after DFO-induced

iron depletion, as confirmed by elevated TFR1 protein levels (Figure 5-6B). I was unable

to test the effect of iron deficiency on ZIP14 expression in βlox5 cells as DFO treatment

did not successfully alter TFR1 levels in this cell line (data not shown).

Modulation of ZIP14 Expression By IL-1β in Human β Cells

IL-1β levels are elevated in primary islets isolated from type 2 diabetics.

Additionally, ZIP14 mRNA levels have been observed to increase in response to IL-1β

in isolated mouse hepatocytes (222). To determine if IL-1β induces ZIP14 expression in

human β cells, βlox5 cells were treated with IL-1β for either 8 or 24 h. Both of these

treatment times increased ZIP14 protein levels to a similar degree (Figure 5-6C).

However, treatment of human islets with IL-1β (for 24 h) resulted in no induction of

ZIP14 protein (Figure 5-6D).

Discussion

Disorders of iron overload in humans are associated with β cell iron accumulation

(132-134), which is currently thought to impair β cell function (45). While β cell iron

loading is has been documented during these disorders, little is known regarding the

mechanisms by which β cells take up iron. In the present study I examined the

contribution of the established NTBI transport proteins DMT1, ZIP14, and ZIP8 to β cell

NTBI uptake. The observation that suppression of ZIP14 expression decreased NTBI

uptake by approximately 50% in the human pancreatic β cell line βlox5 suggests that

Page 91: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

91

ZIP14 is a major route of NTBI uptake by human β cells. A similar reduction in NTBI

uptake was observed after suppression of ZIP14 expression in isolated primary human

islets, which I found express ZIP14 in β but not cells. Iron loading in human islets is

reported to be restricted to β cells, in line with the pattern of ZIP14 expression in human

islets, suggesting that the lack of iron accumulation in α cells may be due to a lack of

ZIP14 expression (132, 134). Although ZIP14 in the human pancreas is detected in β

cells, more robust ZIP14 staining was observed in surrounding acinar cells, similar to

our previous studies of ZIP14 expression in rat pancreas (82). Indeed, the more robust

expression of ZIP14 in acinar cells likely explains why iron loads in the exocrine

pancreas during iron overload (7). However, in contrast to the pattern of ZIP14

expression in human pancreas, ZIP14 in rat pancreas was not detectable in β cells (82).

Based on these observations, I speculate that the lack of β cell ZIP14 in rodents

accounts for the fact that rodent β cells do not load iron, even in the context of massive

iron overload (60, 136, 138, 223, 224). I am aware of only 2 studies that have

demonstrated iron loading by Perls’ staining in rodent β cells, both of which have utilized

non-physiologic models of iron loading (e.g, portacaval shunting and iron dextran

injections) (137, 144).

Recently it has been reported that plasma NTBI levels are elevated in type 2

diabetics, even in the absence of systemic iron overload in which plasma iron levels

exceed the binding capacity of transferrin (131). Due to the ability of cellular iron loading

to increase ZIP14 expression in other cell populations (82, 95), plasma NTBI could

initially be taken up by β cells, leading to an upregulation of ZIP14 and therefore an

increased capacity for subsequent β cell NTBI uptake. Excess β cell iron is proposed to

Page 92: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

92

decrease insulin secretory capacity (45). Thus, the mechanism by which iron uptake

and accumulation increases subsequent iron loading may be relevant in the context of

diabetic pathology. While I found that iron loading increased ZIP14 levels in βlox5 cells

this trend was not observed in primary human islets arguing against a cyclic mechanism

of iron uptake and ZIP14 upregulation. The upregulation of ZIP14 observed in βlox5

cells but not in primary human islets may be due to the iron status of these cell

populations under normal culture conditions. Isolated islets are reported to be quiescent

in vitro (225) whereas βlox5 cells rapidly proliferate resulting in a basal state of iron

deficiency, evidenced by a lack of induction in TFR1 levels after treatment with DFO

(data not shown). Therefore, it is possible that the upregulation of ZIP14 in βlox5 cells

after treatment with iron is not due to cellular iron loading but rather the restoration of

adequate iron status, a change which does not occur in cultured primary islets as iron

status may be adequate to support cellular function, even after temporary iron chelation

with DFO, due to a lack of proliferation.

β cells from individuals with type 2 diabetes display increased levels of the

cytokine IL-1β, attributed to exposure to elevated levels of glucose (226). Given that IL-

1β was previously demonstrated to increase Zip14 expression in isolated mouse

hepatocytes (222), I hypothesized that IL-1β may increase β cell ZIP14 levels which, in

diabetics with plasma NTBI, could increase β cell NTBI uptake. While ZIP14 levels

increased in βlox5 cells following IL-1β treatment no effect was observed in primary

human islets suggesting that IL-1β is unlikely to upregulate ZIP14 in islets of patients

with diabetes. Differences in gene expression between βlox5 cells and primary human β

Page 93: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

93

cells have previously been reported (221), potentially accounting for the differential

effect of IL-1β treatment observed in primary islets and βlox5 cells.

The expression of DMT1 in human islets has been reported previously and it has

been hypothesized that DMT1 may be responsible for β cell iron loading (85). While I

observed that DMT1 is expressed in β cells, and is also absent from α cells reflecting

the pattern of islet iron loading in humans, other results from the present study argue

against a role of DMT1 in the process of NTBI uptake by β cells. For example, I found

that overexpression of DMT1 fails to increase NTBI uptake in βlox5 cells, likely due to

the intracellular localization of DMT1 precluding iron uptake at the cell surface.

Immunofluorescense analysis of DMT1 in human islets also suggests that DMT1 is

localized intracellularly, due to the punctate, granular staining pattern observed. In

addition to the intracellular localization of DMT1, the functional properties of DMT1,

specifically the coupling of efficient iron transport to a proton gradient, argue against

DMT1 contributing substantially to the uptake of plasma NTBI by β cells. DMT1

functions optimally at pH 5.5 (11), in line with the established function of DMT1 in

intestinal (13) and endosomal NTBI transport (26), and transports iron relatively poorly

at the physiologic pH of 7.4 for plasma.

ZIP8 is reported to be expressed at the plasma membrane of rat β cells (103)

and ZIP8 mRNA is abundantly expressed in the human pancreas, relative to other

tissues (100), suggesting that ZIP8 may contribute to β cell iron uptake. In the present

study I found that the overexpression of ZIP8 in βlox5 cells increased NTBI uptake but

that suppression of endogenous ZIP8 expression had no effect on NTBI uptake,

suggesting that ZIP8 levels are not abundant in human β cells. Additionally, I detected

Page 94: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

94

modest amounts of ZIP8 protein in acinar cells but not in β cells strengthening the

finding that ZIP8 protein expression is negligible, and therefore is unlikely to contribute

to NTBI uptake in human β cells.

In conclusion I have identified ZIP14 as a major contributor to NTBI uptake by

human β cells. The identification of ZIP14 as a route of β cell NTBI uptake provides a

target for inhibitors that could be used to prevent β cell iron accumulation during iron

overload. Future study of the role ZIP14 plays in in-vivo NTBI uptake by β cells, and β

cell function, will need to be carried out using rodent models which successfully reflect

the human phenotype characterized by β cell ZIP14 expression and β cell iron

accumulation.

Page 95: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

95

Figure 5-1. ZIP14 and ZIP8, but not DMT1, overexpression increases iron uptake by

βlox5 cells. A) Western blot analysis of cell lysates from blox5 cells transfected with pCMV-Sport6-empty vector (EV), DMT1, ZIP14, or ZIP8. Tubulin is shown to indicate lane loading. B) Effect of ZIP14, ZIP8, or DMT1 overexpression on the uptake of iron by βlox5 cells. To measure iron uptake,

cells were incubated for 1 h in serum-free medium containing 2 μM [59

Fe]

ferric citrate and 1 mM ascorbate and the cellular uptake of 59

Fe was measured by gamma counting. Data represent the mean ± S.E. of 3 independent experiments performed in triplicate. Group means were compared by unpaired Student’s t-test. Asterisks indicate differences relative to cells transfected with EV (*P < 0.05).

Page 96: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

96

Figure 5-2. When overexpressed in βlox5 cells, ZIP14 localizes to the plasma membrane whereas DMT1 mainly localizes intracellularly. Western blot

analysis of ZIP14, DMT1, Na+/K

+-ATPase, and copper chaperone for

superoxide dismutase (CCS) in total-cell lysate (TCL) or cell-surface (CS) proteins isolated from βlox5 cells transfected with either empty vector (EV), A) ZIP14, or B) DMT1. Plasma membrane proteins were labeled with Sulfo-NHS-SS-Biotin and affinity purified by using streptavidin-agarose columns

prior to western blotting. Na+/K

+-ATPase and CCS serve as markers for

plasma membrane and cytosolic proteins, respectively.

Page 97: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

97

Figure 5-3. Endogenous iron uptake by βlox5 cells is decreased by siRNA knockdown of ZIP14, but not ZIP8. A) Western blot analysis of lysates from βlox5 cells transfected with negative control siRNA (siNC) or siRNA targeting either ZIP14 (siZIP14, left panel) or ZIP8 (siZIP8, right panel). B) To measure NTBI uptake, cells were incubated for 2 h in serum-free medium containing 2 μM

[59

Fe] ferric citrate and 1 mM ascorbate and the cellular uptake of 59

Fe was measured by gamma counting. Data represent the mean ± S.E. of 3 independent experiments performed in triplicate. Group means were compared by unpaired Student’s t-test. Asterisks indicate differences relative to cells transfected with siNC (*P < 0.05).

Page 98: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

98

Figure 5-4. siRNA knockdown of ZIP14 decreases NTBI uptake by primary human islets. A) Western blot analysis of cell lysates from isolated human islets transfected with either negative control siRNA (siNC) or siRNA targeting ZIP14 (siZIP14). B) To measure iron uptake, cells were incubated for 2 h in

serum-free medium containing 2 μM [59

Fe] ferric citrate and 1 mM ascorbate

and the cellular uptake of 59

Fe was measured by gamma counting. Data represent the mean ± S.E. of 3 independent experiments performed in triplicate. Group means were compared by unpaired Student’s t-test. Asterisks indicate differences relative to cells transfected with siNC (**P < 0.01).

Page 99: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

99

Figure 5-5. ZIP14 is detected in human pancreatic β cells by immunofluorescent analysis. Immunofluorescent images taken at either A) 20x or B) 60x of human pancreatic tail sections co-stained for ZIP14 (green), insulin (β cell marker, red), and glucagon (α cell marker, blue). Panels show the same tissue region as stained for ZIP14 only (I), ZIP14 with insulin (II), or ZIP14 with insulin and glucagon (III). Serial sections co-stained in parallel, but with non-immune IgG replacing the ZIP14 primary antibody are shown to indicate background staining (IV).

Page 100: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

100

Figure 5-6. Cellular iron levels and treatment with IL-1β increase ZIP14 levels in βlox5 cells but not primary human islets. A) Western blot analysis of βlox5 cell lysates for ZIP14 and ferritin after 24 h incubation in control (CON) medium or medium supplemented with 100 μM ferric ammonium citrate +1 mM ascorbate (FAC). B) Western blot analysis of ZIP14, TFR1, and ferritin in human-islet lysates 48 h after treatment with CON medium or medium containing 50 µM deferoxamine (DFO) or 100 µM ferric ammonium citrate + 1mM ascorbate (FAC). Lysates from βlox5 cells transfected with either siNC or siZIP14 siRNA are shown to confirm the band size of ZIP14 protein. C) Western blot analysis of ZIP14 in βlox5 lysates after incubation in CON medium or medium supplemented with 100 U/ml recombinant human IL-1β for either 8 or 24 h. D) Western blot analysis for ZIP14 in human-islet lysates after incubation in CON medium or medium containing 100 U/ml recombinant human IL-1β for 24 h. Lysates from βlox5 cells transfected with either siNC or siZIP14 siRNA are shown to confirm the band size. Tubulin is shown to indicate lane loading.

Page 101: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

101

Figure 5-7. mRNA copy numbers of NTBI transporters in primary human islets. qRT-PCR measurement of DMT1, ZIP14, and ZIP8 mRNA copy numbers in total RNA isolated from nondiabetic human islets. Copy numbers were calculated based on standard curves constructed from known concentrations of plasmid DNA encoding either DMT1, ZIP14, or ZIP8. Data represent the mean mRNA copy numbers ± S.E. obtained from 4 independent donors.

Page 102: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

102

Figure 5-8. DMT1, but not ZIP8, is detected in human β cells by immunoflourescence

staining. A) Immunoflourescence images (60x) of human pancreatic tail sections co-stained for DMT1 (green), insulin (red), and glucagon (blue). Panels show the same tissue region as DMT1 signal only (I),DMT1 with insulin signal (II), or DMT1 with insulin and glucagon signal (III). Serial sections co-stained in parallel, but with non-immune IgG replacing the DMT1 primary antibody are shown to indicate background DMT1 signal (IV). B) Immunoflourescence images (20x) of human pancreatic tail sections co-stained for ZIP8 (green), insulin (red), and glucagon (blue). Panels show the same tissue region as ZIP8 signal only (I), ZIP8 with insulin signal (II), or ZIP8 with insulin and glucagon signal (III). Serial sections co-stained in parallel, but with non-immune IgG replacing the ZIP8 primary antibody are shown to indicate background ZIP8 signal (IV).

Page 103: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

103

CHAPTER 6 THE INFLUENCE OF IRON STATUS ON DIABETIC PATHOLOGY AND β-CELL

FUNCTION

The link between iron status and diabetes has been extensively documented in

patients with iron overload disorders, in which the prevalence of diabetes is elevated

compared with the general population (45, 113, 114, 118). Patients with the iron

overload disorder hemochromatosis are reported to display diminished insulin secretion

in response to glucose (45), suggesting that excess iron accumulation impairs the ability

of β cells to regulate glucose homeostasis. Diabetes has historically been categorized

as either type 2 diabetes, resulting from systemic insulin resistance, or type 1 diabetes,

resulting from a loss of insulin secretory capacity. Type 1 diabetes most often results

from the autoimmune-mediated destruction of pancreatic β cells. While the link between

systemic iron status and diabetes has been extensively documented, little is currently

known about the influence of iron in the pathogenesis of autoimmune diabetes.

The evidence linking iron status to autoimmune diabetes in humans is currently

limited to retrospective epidemiology demonstrating a potential link between iron and

diabetes risk. Increased iron consumption during infancy is reported to be associated

with a greater risk for developing diabetes during childhood (166) and elevated

transferrin saturation, an indicator of iron status, is associated with an increased

prevalence of type 1 diabetes in adults (167). However, direct mechanistic evidence for

a causative role of iron in autoimmune diabetes is currently lacking. Studies carried out

in animal models have indirectly suggested that iron depletion may be protective against

autoimmune diabetes. Treatment with the iron chelator desferroxiamine protects islet

grafts from autoimmune destruction in NOD mice (170), a mouse model of autoimmune

diabetes. Also, the administration of apotransferrin to NOD mice reduces the incidence

Page 104: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

104

of spontaneous diabetes (173), an effect hypothesized to be attributed to the binding of

plasma NTBI, which may be elevated in diabetics (131). T-cell proliferation and

cytotoxicity are also reduced by treatment with anti-transferrin receptor antibodies (171),

although the ability of T cells to acquire iron in response to antibody treatment was not

determined. Currently no controlled trials have determined the influence of systemic iron

status on the development of autoimmune diabetes.

Evidence for β cell iron status directly affecting β cell function is also lacking as

studies evaluating glucose homeostasis during iron deficiency or overload have done so

in the context of systemic changes in iron status (45, 146, 147, 163). Changes in whole-

body iron levels may affect β cell function through the modulation of known or unknown

systemic factors, such as erythropoietin, which promotes β cell proliferation and

prevents apoptosis (227). Current in vitro studies investigating the direct impact of iron

status on islets in isolation have been limited to rat islets (165) and have not determined

the effect of iron status on islet function, as evidenced by insulin secretion, directly. To

date no studies have determined the effect of iron depletion or loading on the insulin

secretory capacity of human islets. Additionally, the study of β cell iron loading in vivo

has been complicated by the lack of a mouse model which accumulates substantial iron

within β cells, similar to the pattern observed in humans with iron overload (132, 133). β

cells in mouse models of iron overload demonstrate a remarkable resistance to iron

loading (60, 136, 138) making it difficult to determine the effect of progressive β cell iron

accumulation on β cell function.

In the current study I aimed to examine the role of iron in diabetic pathology by

determining the influence of systemic iron status on the development of autoimmune

Page 105: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

105

diabetes in NOD mice and the effect of iron status on glucose-stimulated insulin

secretion (GSIS) by isolated human islets. I hypothesized that iron deficiency would be

protective against the development of diabetes and increase GSIS by human islets

while iron loading would result in an increased incidence of autoimmune diabetes in

NOD mice and impair GSIS by human islets. Additionally I produced a transgenic

mouse expressing the mammalian iron transporter Zip14 under control of the mouse

insulin 1 promoter with the aim of generating a mouse model of β cell iron loading.

Results

Effect of Iron Status on Spontaneous Autoimmune Diabetes in NOD Mice

To determine whether differences in systemic iron status could affect the

development of autoimmune diabetes female NOD mice were fed either iron-deficient

(FeD), iron-adequate (FeA), or iron-loaded (FeO) diets from weaning until 30 wk of age.

FeD mice demonstrated a trend towards a greater incidence of diabetes relative to FeA

mice, 80% diabetic at 30 wk of age compared with 60%, although this difference did not

reach statistical significance (P=0.06) (Figure 6-1). The initial onset of diabetes was

also earlier in FeD mice, first detected at 11 wk of age, compared with FeA mice in

which diabetes was first detected at 14 wk of age. No differences were observed

between the incidences of diabetes in FeO compared with FeA mice and the

development of diabetes was initially detected at similar ages in these groups.

Effect of Dietary Iron on Rate of Growth and Systemic Iron Status

Iron deficient and loaded diets have previously been demonstrated to affect

growth in rodents (176, 228, 229) and higher body weights are associated with

increased susceptibility to autoimmune diabetes in BioBreeding rats (230). However, no

differences were observed in the rate of growth between FeD and FeA mice at any point

Page 106: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

106

in the study. Mice in the FeO group gained weight at a reduced rate initially but the

difference was corrected during the prediabetic period, before 11 wk of age, and body

weights between groups did not differ after this point (Figure 6-2). To confirm that

systemic iron status was successfully altered by dietary treatments prior to the

development of diabetes I measured indices of iron status in 10-wk-old mice that had

consumed FeD, FeA, or FeO diets since weaning (Table 6-1). FeD mice had lower 12%

lower hemoglobin levels compared with FeA mice but plasma iron levels were not

diminished in response to dietary iron deficiency. FeO mice had elevated transferrin

saturation, attributed primarily to diminished total iron binding capacity (TIBC), whereas

no difference was detected between FeD and FeA mice, in line with the similar plasma

iron concentrations between these groups. Iron stores, indicated by liver non-heme iron

concentrations, were depleted in FeD mice and 10 times greater in FeO animals

compared with FeA mice, indicating that iron status was successfully altered by dietary

intervention during the period preceding the development of diabetes.

Plasma iron was found to be greater in 30 wk-old-FeO mice compared with other

groups, although transferrin saturation was similar between age groups. Iron status in

FeD mice normalized with age, as evidenced by the recovery of hemoglobin values in

30-wk-old FeD mice (Table 6-1). Additionally liver iron stores increased in 30-wk-old

FeD mice relative to those measured in FeD mice at 10 wk of age. However, iron stores

were found to still be significantly greater in FeA mice compared with FeD at 30 wk of

age. Unlike at 10 wk of age, plasma iron was significantly greater in 30-wk-old FeO

mice compared with the other groups. Measurement of liver non-heme iron levels from

Page 107: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

107

FeD mice at various ages between 10 and 30 wk of age indicate that iron stores

gradually increase in a linear fashion after 10 wk of age (Figure 6-3).

Pancreatic Mineral Concentrations

Our lab has demonstrated that alterations in dietary iron can alter pancreatic

mineral concentrations, which may contribute to pancreatic dysfunction (139). To

explore this possibility I measured trace minerals in the pancreases from prediabetic

FeD, FeA, and FeO mice by using ICP-MS analysis. In 10-wk-old mice pancreatic iron

levels in FeD and FeO mice were 52% and 172% of FeA levels, respectively (Table 6-

2). Modest differences were also detected in pancreatic zinc and copper, which were

elevated in FeD mice compared with the FeA and FeO animals, and in selenium which

was lower in FeO mice. Pancreatic Iron levels in 30-wk-old mice were not different

between FeD and FeA animals but were 3.5 times greater in FeO mice. Differences in

pancreatic zinc levels were not detected between groups at 30 wk of age but FeD mice

had slightly elevated copper levels.

Testing of β cell function During the Prediabetic Period

To investigate whether systemic iron status has an effect on β cell function in

NOD mice I performed glucose tolerance testing using 10 wk prediabetic mice. No

differences in glucose tolerance or fasting glucose levels between groups were detected

(Figure 6-4A). However, all groups reported poor glucose tolerance, maintaining blood

glucose levels >300 even 2 h post injection. GSIS capacity was also measured in

conjunction with glucose tolerance and no significant differences in plasma insulin levels

were detected between groups at any time point (Figure 6-4B).

As β cell iron status has been hypothesized to affect β cell function, islets from

mice used in glucose tolerance testing were isolated and iron status was determined by

Page 108: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

108

measuring transferrin receptor 1 (TFR1) expression, which is well documented to

inversely reflect cellular iron levels (78). TFR1 levels in FeO mice were lower compared

with FeD and FeA mice, indicating increased levels of islet iron (Figure 6-5A). However,

no difference was observed between FeD and FeA mice indicating that islet iron status

was not different between FeD and FeA mice at 10 wk. Also, histological analysis of

pancreas sections from 10 wk mice revealed no difference in the degree of insulitis

between groups, with all groups reporting only mild insulitis (Figure 6-5B).

Effect of Iron Status on Human Islet GSIS

The feeding of NOD mice with an iron-deficient did not result in diminished β cell

iron status in vivo. To determine the effect of iron status on β cell function I altered the

iron status of isolated primary human islets in vitro prior to GSIS testing to indicate islet

function. Islets were treated for 48 h with either control medium (CON), 50 μM

deferoxamine mesylate (DFO), an iron chelator to deplete islets of iron, or 100 μM ferric

ammonium citrate and 1 mM ascorbate (FAC), to load islets with iron. The successful

alteration of islet iron status was confirmed by western blot analysis for both TFR1 and

ferritin, an indicator of cellular iron stores. TFR1 expression was elevated in islets

treated with DFO relative to CON islets, indicating a reduction in cellular iron levels, and

decreased in islets treated with FAC, indicating cellular iron loading (Figure 6-6A).

However, no differences in insulin secretion between groups, during either basal or

glucose stimulated conditions, were measured during GSIS testing indicating that

altered iron status does not affect GSIS (Figure 6-6B).

Generation of Mice Selectively Overexpressing Zip14 in β Cells

Previous studies using mouse models of severe iron overload have

demonstrated that mouse islets are resistant to substantial iron loading in vivo (60, 136,

Page 109: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

109

138). Recent findings by our lab have indicated that ZIP14 is required for iron loading of

the exocrine pancreas (7) and that ZIP14 contributes to β cell iron uptake by human

islets (unpublished results). In light of these findings I aimed to generate transgenic

mice overexpressing ZIP14 in pancreatic β cells to create a novel mouse model

predisposed to β cell iron loading. I generated a vector construct containing mZip14

tagged with human influenza hemagglutinin antigen (HA) under control of the mouse

insulin 1 promoter (MIP) and containing a downstream intronic region of human growth

hormone (Figure 6-7A). Four founder animals were obtained and bred to establish 4

individual transgenic mouse lines. Analysis of the number of transgene copies per

genome by using qRT-PCR revealed that 2 founders possessed approximately 5

copies, referred to as low-copy lines, while other founders had approximately either 13

or 500 copies, referred to as moderate- or high-copy lines. Comparison between

transgene copy numbers in founder animals and subsequent generations indicate that

the transgenes are completely inherited in the lines carrying low- and moderate-copy

lines. However, incomplete inheritance of the transgene was detected within the high-

copy line with some offspring inheriting a low number of copies. Successful expression

of the transgene in β cells was confirmed by immunostaining for HA in pancreatic

sections (Figure 6-7B). Animals from 3 out of 4 transgenic lines demonstrated

transgene expression to various degrees in line with the number of transgene copies

detected.

Discussion

Excess iron accumulation is associated with an increased prevalence of diabetes

and is believed to influence aspects of diabetic pathology (45, 113, 114, 118). Previous

experiments have suggested that systemic iron levels may be a risk factor for the

Page 110: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

110

development of autoimmune diabetes (167, 169) and that β cell iron accumulation

results in diminished insulin secretory capacity (45, 146). However, direct evidence for

these claims is currently lacking. The present study using NOD mice produced the

unexpected results that iron overload did not increase the incidence of diabetes and that

dietary iron restriction was not protective, potentially even promoting to the development

of autoimmune diabetes. Diabetes was detected earlier in FeD mice and the overall

incidence of diabetes trended strongly towards being increased by dietary iron

restriction, suggesting that iron deficiency may increase susceptibility to the

development of autoimmune diabetes. Furthermore, dietary iron overload failed to

increase the incidence of diabetes compared with mice fed FeA diets, arguing against

the hypothesis that elevated iron stores increase the risk of developing type 1 diabetes.

While iron deficiency may negatively impact the development of diabetes in NOD

mice, experiments in prediabetic mice failed to explain the trend toward an increased

incidence of diabetes in in iron-restricted mice. Glucose tolerance testing in prediabetic

NOD mice did not reveal any differences between groups regarding either glucose

tolerance or GSIS, indicating that systemic iron status did not affect glucose

homeostasis or β cell function during the prediabetic period. All groups investigated

reported poor glucose tolerance, potentially attributed to the high sucrose diet used in

this study (231). Other studies have reported that feeding an iron-deficient or loaded

diet can affect insulin sensitivity in rodents (232-234). In this study I did not detect any

differences in insulin sensitivity as both glucose tolerance and insulin secretion were

similar for all groups measured. Discrepancies between the current study and previous

studies regarding the influence of iron loading or depletion on insulin sensitivity in

Page 111: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

111

rodents may result from differences in either dietary iron content or feeding duration. In

general the iron content of the diets and the duration of feeding used in the present

study were less extreme compared with diets and timelines used in previous studies

reporting differences in insulin sensitivity in response to dietary iron deficiency or

overload (232-234).

The lack of difference in GSIS between groups during glucose tolerance testing

indicates that β cell function was not affected by altering systemic iron stores during the

prediabetic period. While iron stores were lower in 10-wk-old FeD mice relative to FeA

mice, as evidenced by diminished liver non-heme iron and mild anemia in FeD animals,

islet TFR1 levels were not different between these groups suggesting that the trend

toward increased diabetic development was not attributable to β cell iron deficiency.

Serum iron was also similar between FeD and FeA mice suggesting that circulating iron

levels were adequate to supply islets with iron, thus preventing islets from becoming

iron deficient. Additionally, dietary iron overload increased islet iron status but did not

result in altered insulin secretory capacity or glucose tolerance, suggesting that islet iron

loading, at least to the degree observed in this study, does not have an effect on β cell

function.

The finding that islet iron status does not affect GSIS is supported by the testing

of iron-loaded and iron-depleted human islets in vitro, also carried out in this study. I

determined that there was no effect of iron status on GSIS, during either basal or high

glucose conditions, arguing against the hypothesis that β cell iron status impacts insulin

secretory capacity. To my knowledge this is the first report of the effect of iron depletion

or iron loading on human β cell function in an isolated cell-culture system, eliminating

Page 112: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

112

the potential influence of systemic factors, on β cell function. The lack of effect on GSIS

by both iron loading and iron depletion was unexpected based on previous reports

demonstrating a role for ROS in the process of glucose stimulated insulin secretion

(235) and the hypothesized role of cellular iron in the generation of intracellular ROS

(86). It is possible that in the current study the degree of iron loading of human islets did

not exceed the ability of the β cell to sequester iron within ferritin, preventing the buildup

of free intracellular iron which would be capable of catalyzing ROS formation. While iron

status was clearly different between control, iron-depleted, and iron-loaded islets in the

current study, as indicated by TFR1 and ferritin levels, future investigation of the

influence β cell iron status has on insulin secretory capacity may benefit from longer

term or higher-dose iron loading. Additionally it is possible that the alteration of β cell

iron status affects first-phase insulin secretion, an early indicator of impaired β cell

function (236, 237), which would not be detected by the methods used in this study.

Future studies may benefit from the use of more nuanced methods of GSIS testing,

such as islet perifusion, which is capable of discerning differences in GSIS at individual

time points.

The study of long-term, progressive β cell iron accumulation would benefit from

the availability of a mouse model that accumulates iron in β cells, similar to what has

been observed in humans with iron overload disorders (132, 133). In this study I also

detailed the generation of transgenic mice that overexpress mZIP14 in pancreatic β

cells under the control of the mouse insulin 1 promoter. Previous reports by our lab

have indicated that ZIP14 is required for iron loading of pancreatic acinar cells (7) and

that ZIP14 contributes to non-transferrin bound iron uptake by human islets (publication

Page 113: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

113

under review), suggesting that overexpressing ZIP14 in mouse β cells may result in

increased iron uptake and accumulation. In this study I reported the successful

overexpression of mZIP14 in β cells, localized to the plasma membrane and intracellular

locations within β cells of transgenic mice. Future testing is needed to confirm that this

novel model demonstrates β cell iron loading during systemic iron overload.

A limitation of the current study is that iron deficiency resolved in the FeD mice

with age, potentially attributable to the plateau of growth after mice reached

approximately 10 wk of age. Due to the recovery of iron status it is possible that early

trends observed regarding the incidence of diabetes in FeD mice were somewhat

ablated in older mice and could potentially be more dramatic under conditions of more

severe iron deficiency. In the current study the iron-deficient diet contained 14 ppm iron,

a greater concentration than traditional iron-deficient diets (163, 238), due to the

inclusion of 5% wheat in the diet. Wheat was added to the diets as additional wheat has

been reported to increase the diabetic potential of purified diets (239), which usually

result in a low incidence of diabetes in NOD mice (240, 241). Future studies

investigating the role of iron deficiency in autoimmune diabetes may benefit from the

use of BioBreeding rats, which also develop spontaneous autoimmune diabetes and in

which iron deficiency may be easier to induce due to the greater increase in rat body

weight relative to that of mice.

In conclusion I report that iron deficiency, but not iron overload, may increase the

development of autoimmune diabetes in NOD mice. Additionally β cell iron status did

not affect β cell function, calling into question the long-hypothesized mechanism thought

to account for the link between iron loading and diabetes. Future studies will be required

Page 114: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

114

to determine the influence of both systemic and β cell iron content in the pathogenesis

of diabetes.

Page 115: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

115

Table 6-1. Iron parameters of type 1 diabetes-prone NOD mice

Age Group Hemoglobin

(g/dL)

Liver non-heme iron (μg Fe/g)

Plasma Iron (μg/dL)

TIBC (μg/dL)

TF Sat (%)

10 wk FeD 12.7 ± 1.3a 16.7 ± 3.8a 158.1 ± 61.7a 383.3 ± 43.7a 37.4 ± 15.5a

10 wk FeA 14.5 ± 1.1b 247.2 ± 41.8b 198.9 ± 46.7a 402.2 ± 22.8a 49.6 ± 12.1ab

10 wk FeO 14.5 ± 0.6b 2490.4 ± 410.1c 231.4 ± 51.2a 313.9 ± 38.9b 74.4 ± 18.3b

30 wk FeD 14.1 ± 0.6a 118.4 ± 27.5a# 133.3 ± 16.7a 392.8 ± 22.5a 33.9 ± 2.4a

30 wk FeA 14.1 ± 0.9a 424.2 ± 23.3b# 136.1 ± 32.9a 366.2 ± 24.1a 37.1 ± 8.4a

30 wk FeO 15.1 ± 0.4a 3124.2 ± 1027.3c 281.5 ± 97.0b 385.6 ± 74.2a 71.6 ± 13.1b

Liver non-heme iron levels are reported as μg Fe/g wet tissue. Liver non-heme iron, plasma iron, and total iron-binding capacity (TIBC) were determined colorimetrically. Transferrin saturation (TF Sat) was calculated as plasma iron as a percentage of TIBC. Values are presented as means ± SD, n=3-8. Statistical significance was determined by one-way ANOVA. Means without a common superscript are significantly different compared with other groups at the same age. Values at 30 wk of age that are significantly different from those of the same group at 10 wk of age are indicated by # (P<0.05).

Page 116: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

116

Table 6-2. Pancreatic mineral concentrations in NOD mice

Age Group Iron Zinc Manganese Copper Cobalt Selenium Molybdenum

10 wk FeD 77.7 ± 13.4a 259.0 ± 57.6a 7.6 ± 1.1a 7.6 ± 1.3a 0.16 ± 0.01a 1.4 ± 0.20a 0.5 ± 0.04a

10 wk FeA 148.8 ± 26.6b 194.8 ± 24.8b 7.4 ± 1.3a 6.3 ± 0.5b 0.16 ± 0.02a 1.3 ± 0.08a 0.5 ± 0.08a

10 wk FeO 256.7 ± 67.3c 164.4 ± 25.0b 7.9 ± 1.1a 5.5 ± 0.5b 0.14 ± 0.01a 1.0 ± 0.20b 0.5 ± 0.05a

30 wk FeD 251.5 ± 58.0a# 183.0 ± 40.6a# 10.4 ± 0.5a 6.9 ± 0.4a 0.18 ± 0.03a 1.3 ± 0.1a 0.5 ± 0.03a

30 wk FeA 260.7 ± 25.5a 161.5 ± 40.6a 8.5 ± 0.7a 5.4 ± 0.5b 0.14 ± 0.01a 1.3 ± 0.2a 0.4 ± 0.04a

30 wk FeO 931.69.7 ± 654.5b# 153.7 ± 17.4a 17.7 ± 11.8a# 5.1 ± 0.5b 0.13 ± 0.03a 1.3 ± 0.3a 0.5 ± 0.07a

Mineral concentrations (mg/g dry weight) were measured by using ICP-MS. Values represent means ± SD, n = 4-6. Means without a common superscript are significantly different P<0.05. Statistical significance was determined by one-way ANOVA. Means without a common superscript are significantly different compared with other groups at the same age. Values at 30 wk of age that are significantly different from those of the same group at 10 wk of age are indicated by # (P<0.05).

Page 117: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

117

0 5 10 15 20 25 300

20

40

60

80

100FeD (n=20)

FeA (n=20)

FeO (n=20)

P=0.06

Age (weeks)

% W

ith

ou

t D

iab

ete

s

Figure 6-1. Dietary iron deficiency, but not iron overload, results in a trend towards an

increased incidence of spontaneous diabetes in female NOD mice. Cumulative diabetes incidence in female NOD mice fed either iron deficient (FeD), iron adequate (FeA), or iron loaded (FeO) diets starting at 3 wk of age. Spontaneous development of diabetes was monitored by glycosuria starting at 8 wk of age. Survival curves were compared to FeA by using the Gehan-Breslow-Wilcoxon test.

Page 118: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

118

3 6 9 12 15 18 21 24 27 300

5

10

15

20

25

30

35

FeD (n=20)

FeA (n=20)

FeO (n=20)** ***

****

Age (Weeks)

Bo

dy

We

igh

t (g

)

Figure 6-2. NOD mice fed an iron-loaded diet initially experience diminished growth.

Body weights of female NOD mice fed either iron-deficient (FeD), iron-adequate (FeA) or Iron-loaded (FeO) diets from 3 to 30 wk of age. Animals were weighed every 3 days until 12 wk of age, at which point body weights were recorded weekly. Body weights at individual time points were compared by one-way ANOVA. Asterisks indicate significant differences in bodyweights of FeO animals compared with FeA animals (*P<0.05, **P<0.01, ***P<0.001)

Page 119: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

119

10 15 20 25 300

50

100

150

200

r=0.92

Age (Weeks)

Liv

er

No

nh

em

e Iro

n (

g/g

tis

su

e)

Figure 6-3. Iron stores of mice fed an iron-deficient diet increase with age. Liver non-

heme iron concentrations of FeD mice were measured colorimetrcally in 10-wk-old prediabetic mice, diabetic mice of various ages, and 30 wk nondiabetic mice fed an iron-deficient diet. The correlation coefficient (r) was calculated by using a linear model.

Page 120: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

120

Figure 6-4. Glucose tolerance and insulin secretory capacity is not affected by iron

status in prediabetic NOD mice. A) Results of intraperitoneal glucose tolerance testing in fasted 10-wk-old prediabetic female NOD mice. Values reported as mean ± SEM, n=5 per group. B) Plasma insulin levels in mice used in glucose tolerance testing. Values reported as mean ± SEM, n=3-5 per group. Blood glucose and plasma insulin levels between groups at individual time points were compared by one-way ANOVA.

Page 121: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

121

Figure 6-5. Iron-deficient prediabetic NOD mice show no differences in β cell iron status

or insulitis compared with iron-adequate mice. A) Western blot analysis of mouse islet total-cell lysate from 10-wk-old prediabetic iron-deficient (FeD), iron-adequate (FeA), and iron-loaded (FeO) NOD mice for transferrin receptor (TFR1). Islets were pooled from 2-3 mice per group. Tubulin is shown to indicate lane loading. B) Average insulitis score from FeD, FeA, and FeO 10-wk-old prediabetic female NOD mice. Values are expressed as the mean ± SEM, n=6. Statistical significance was determined by one-way ANOVA.

Page 122: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

122

Figure 6-6. Iron status does not affect glucose-stimulated insulin secretion by human

islets in vitro. A) Western blotting of human islet total cell lysate for transferrin receptor (TFR1) and ferritin. Islets were treated for 48 h with either control medium (CON), 50 μM deferoxamine mesylate (DFO) or 100 μM ferric ammounium citrate and 1 mM ascorbate (FAC) prior to analysis. Tubulin is shown to indicate lane loading. B) Ability of islets to secrete insulin after DFO or FAC treatment. Total insulin secreted by islets during a 1-h incubation in media containing 2.8 mM D-Glucose followed by a 1-h incubation in media containing 22 mM D-glucose was measured and normalized to islet DNA. Data represent the mean ± SEM of 4 independent experiments performed in triplicate. Treatment group means were compared by one-way ANOVA.

Page 123: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

123

Figure 6-7. Generation of mice selectively overexpressing Zip14 in β cells. A) Vector

map of the construct used to generate MIP-Zip14-HA transgenic mice consisting of a region of the mouse insulin 1 promoter (MIP), HA-tagged ZIP14 (ZIP14-HA), and an intronic region of human growth hormone (hGH Intron). DNA was digested with HindIII and SfiI, to remove the vector backbone, purified, and introduced to fertilized embryos by pronuclear injection. B) Confirmation of transgene expression by Immunofluorescense analysis. Mouse pancreas sections were co-stained for HA (green), insulin (red), and DAPI (blue). Panels show the same tissue section as HA with insulin and DAPI (panel I), HA with DAPI (panel II), insulin with DAPI (panel III). A pancreatic section from a wild-type mouse co-stained in parallel for HA, insulin, and DAPI is shown to indicate non-specific background HA signal (panel IV).

Page 124: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

124

CHAPTER 7 CONCLUSIONS, LIMITATIONS, AND FUTURE DIRECTIONS

The experiments detailed in this dissertation explore the connection between iron

status and diabetes and can be categorized into 2 general topics: the study of NTBI

uptake by pancreatic β cells and the effect of iron status on diabetic pathology.

Experiments investigating the process of β cell NTBI uptake evaluated the evidence for

the involvement of the 3 identified mammalian NTBI transport proteins, DMT1, ZIP14,

and ZIP8 in β cell iron uptake. As iron loading is more extensively documented in

human than in rodent islets, experiments were carried out using human β cell lines,

isolated primary human islets, and human pancreas samples. Experiments examining

both the cellular and subcellular localization, as well as effect of expression on iron

transport, provided evidence for the contribution of ZIP14, but not DMT1 or ZIP8 to the

process of NTBI uptake by human β cells. Previous investigation into iron uptake, either

as TBI or NTBI, by β cells has not been carried out making this finding a novel

contribution to the literature. However, certain limitations in the experiments performed

leave unanswered questions and provide direction for future studies.

Investigation of iron uptake by β cells in this study focused on mechanisms of

NTBI uptake. Although NTBI is thought to be the primary form of iron taken up by the

pancreas during iron overload (7, 242), studies have not determined the contribution of

TBI uptake in iron accumulation by pancreatic β cells. A prominent role for TBI uptake in

the process of β cell iron loading seems unlikely, due to the negative regulation of TFR

by cellular iron status. However, human β cells express TFR (243) and the possibility

cannot be ruled out as the contribution of TBI to β cell iron uptake was not directly

determined by experiments in this dissertation. Another unexplored aspect of β cell iron

Page 125: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

125

loading is the discrepancy between human β cells, which accumulate iron deposits

(132, 133), and mouse β cells, which resist iron accumulation during iron overload (60,

136). The findings in this report indicating that ZIP14 contributes to NTBI uptake in

human β cells, as well as the recently published finding by our lab that ZIP14 is required

for iron loading in mouse pancreatic acinar cells (7), pose the obvious question of

whether the resistance of mouse β cells to iron loading is due to the absence or reduced

abundance of ZIP14 in mouse β cells. However, despite repeated efforts, I have been

unable to successfully detect ZIP14 in the mouse pancreas by immunofluorescence

analysis or immunohistochemistry. Future experiments confirming that ZIP14 is either

not expressed, or far less abundantly expressed, in mouse β cells compared with

human β cells will be necessary to conclude that the discrepancy in β cell iron loading

between species is due to differential ZIP14 expression.

Investigation into potential mechanisms by which iron status may influence

diabetic pathology was initiated by the analysis of pancreatic gene expression in rats

fed either iron-deficient or iron-loaded diets. The primary finding in this study was that

both iron overload and deficiency increased the expression of Reg family genes,

associated with pancreatic stress (244) and β cell regeneration (209), as well as Alox15,

a gene linked to the development of autoimmune diabetes in NOD mice (188). The idea

that iron deficiency, as well as iron overload, can elicit changes in gene expression

associated with diabetic pathology is novel and at odds with what has been previously

reported suggesting that iron deficiency is protective against the development of

diabetes ((163, 164). However, this study was observational in nature and did not

provide further mechanistic evidence for a role of either changes in the expression of

Page 126: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

126

Reg family genes Alox15 in pancreatic function in response to altered iron status.

Additionally attributing the observed changes in gene expression directly to pancreatic

iron status is problematic due to differences in pancreatic mineral concentrations

between groups. Iron-loaded animals displayed markedly reduced pancreatic copper

levels and copper deficiency has been reported to result in pancreatic degradation

(214), potentially contributing to pancreatic stress that may have induced the expression

of Reg family genes. Unpublished observations indicate that iron-loaded rats in this

study demonstrate lower copper levels in the liver as well, possibly resulting from

impaired dietary copper absorption when placed on a high iron diet, as lower tissue

copper levels were not observed in genetic models of iron overload fed normal diets.

Future studies are needed to determine the effect of altered copper status on pancreatic

stress and to determine if the differences observed in response to dietary iron overload

are due to elevated iron or rather depleted pancreatic copper levels. Another caveat is

that due to the nature of microarray analysis this study was only designed to detect

differences in gene expression at the mRNA level. Differences in the posttranscriptional

regulation of pancreatic gene expression, such as the upregulation of ALOX15 during

iron overload which was fortunately measured by western blotting, will usually go

undetected by using this methodology. Finally, due to the low level of pancreatic iron,

even in response to a high iron diet, I was unable to successfully stain pancreatic

sections from these rats to determine the location of iron deposition as the level of

pancreatic iron was below the threshold for enhanced Perls’ staining in my hands.

Therefore, it is difficult to conclude which cell populations are being affected by

differential iron status and are thus likely to be responsible for the alterations in gene

Page 127: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

127

expression. Future studies may benefit from the isolation of islets or acinar cells prior to

gene expression analysis to eliminate the issue of dealing with heterogeneous cell

populations.

In line with the concept of determining the effect of iron status on isolated

populations of cells, I performed experiments testing the ability of isolated human islets

to secrete insulin in response to glucose after either iron depletion or loading. Previous

studies suggesting that iron status affects β cell function have been carried out in vivo

(146, 147, 163), where other factors besides β cell iron status could potentially alter

insulin secretion. In contrast to what has been suggested by other studies I found that

the alteration of islet iron status had no effect on GSIS. This finding was unexpected

and in disagreement with the existing literature reporting differences in insulin secretory

capacity in response to changes in whole body iron status. However, there are obvious

limitations with this experiment as I only measured total insulin secretion after 1 h

exposure to either low or high glucose. It is possible that differences in first-phase

insulin secretion, indicative of reduced β cell function (236, 237), manifest in response

to altered iron status but could be corrected by subsequent increased insulin secretion

at later time points. Future experiments would benefit from the use a perifusion system

to measure the rate of insulin secretion continuously throughout testing.

Another limitation of the in vitro islet function testing I carried out is that the

treatments may have been insufficient, in either duration or severity, to induce the

physiologic changes necessary to alter insulin secretory capacity. Time points and

doses of supplemental iron or deferoxamine in the present study were selected as they

successfully altered the expression of TFR1, a well-established indicator of cellular iron

Page 128: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

128

status (78). It is possible that treatment with increased concentrations of iron chelators

or additional iron for longer time periods may affect insulin secretion but due to limited

resources these possibilities were not investigated. Additionally, it is possible that

alterations in iron status alone are not sufficient to perturb β cell function but may

exacerbate a loss of function when combined with metabolic stress due to chronically

elevated glucose and/or free-fatty acids, as observed during diabetic conditions (245).

Future experiments should investigate these possibilities before concluding that iron

status does not affect GSIS. It would also be beneficial to measure other indicators of

cellular damage, similar to what has been published using isolated rat islets (165).

Generation of MIP-Zip14-HA transgenic mice overexpressing ZIP14 in β cells

should allow for the future study of β cell iron loading on β cell function in an in vivo

model of systemic iron overload. However, it has not yet been confirmed that these

mice successfully load iron in β cells during iron overload conditions. Future

experiments confirming this either by crossing MIP-Zip14-HA mice with models of

genetic iron overload or by injecting MIP-Zip14-HA mice with iron dextran will be

needed to determine if the overexpression of ZIP14 in mouse β cells results in mouse β

cell iron accumulation.

The investigation of the effect of iron status on the development of autoimmune

diabetes in NOD mice was carried out as a pilot study to determine whether the

hypothesized connection between iron status and type 1 diabetes would manifest as a

clear trend in a controlled trial using an animal model. Contrary to what is suggested by

epidemiological studies (166, 167) and preliminary cell culture or animal experiments

(170, 171, 173), iron overload did not increase the incidence of diabetes in NOD mice.

Page 129: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

129

Additionally mice fed an iron-deficient diet, which was hypothesized to be protective

against diabetes, trended towards an increased incidence of diabetes. This novel

finding suggests that the relationship between iron status and autoimmune diabetes

may be different than what has been been previously proposed. However, as a

preliminary study there are many limitations which will need to be addressed by future

experiments.

Preliminary testing in this study did not reveal any mechanisms by which iron

deficiency may affect the development of diabetes in NOD mice. In prediabetic NOD

mice I detected no difference between groups in either glucose tolerance, GSIS, or the

degree of insulitis leaving the trend towards an increased incidence of diabetes in iron-

deficient NOD mice unexplained. Future studies will need to further investigate the

influence of iron status on aspects of diabetic pathology in NOD mice to elucidate

potential mechanisms by which iron deficiency may be detrimental. Another limitation of

this study is fact that iron-deficient mice slowly recovered their iron status over the

duration of the study. Due to the gradual buildup of iron stores it cannot be said that

FeD mice were in fact iron deficient after 10 wk of age, although their iron stores were

still depleted compared with other groups. In line with this concern plasma iron levels

were not different between FeD and FeA mice, even at 10 wk of age. The relative

ineffectiveness of the iron-deficient diet at inducing iron deficiency over a prolonged

period is likely due to the extra iron provided by the addition of wheat to the diet,

necessary to increase the prevalence of diabetes in NOD mice fed a purified diet (239).

The iron content of the diet is unlikely to be further reduced mandating the use of other

methods to increase the severity of iron depletion. Future studies may benefit from the

Page 130: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

130

use of BioBreeding rats in place of NOD mice, as rats are more susceptible to iron

deficiency. Additionally animals could be housed in wire-bottom cages long term to

prevent coprophagy, although this assumes that adequate iron could not be obtained

from the diet alone. Periodic bleeding to reduce iron stores is another option but may be

complicated by altered glucose metabolism due to erythropoietin production in response

to bleeding (227, 246).

In summary, the results of these experiments provide new insight into both

mechanisms of NTBI uptake by human β cells and the role of iron in diabetic pathology.

The results of the experiments previously discussed question many aspects of the

proposed link between iron status, β cell function, and the development of autoimmune

diabetes. Future studies will hopefully clarify these issues.

Page 131: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

131

LIST OF REFERENCES

1. Cartier LJ, Ohira Y, Chen M, Cuddihee RW, Holloszy JO. Perturbation of

mitochondrial composition in muscle by iron deficiency. Implications regarding regulation of mitochondrial assembly. J Biol Chem 1986;261(29):13827-32.

2. Davies KJ, Maguire JJ, Brooks GA, Dallman PR, Packer L. Muscle mitochondrial bioenergetics, oxygen supply, and work capacity during dietary iron deficiency and repletion. Am J Physiol 1982;242(6):E418-27.

3. Hoensch H, Woo CH, Raffin SB, Schmid R. Oxidative metabolism of foreign compounds in rat small intestine: cellular localization and dependence on dietary iron. Gastroenterology 1976;70(6):1063-70.

4. Hoffbrand AV, Ganeshaguru K, Hooton JW, Tattersall MH. Effect of iron deficiency and desferrioxamine on DNA synthesis in human cells. Br J Haematol 1976;33(4):517-26.

5. Pietrangelo A. Hereditary hemochromatosis--a new look at an old disease. N Engl J Med 2004;350(23):2383-97. doi: 10.1056/NEJMra031573.

6. Angelucci E, Barosi G, Camaschella C, Cappellini MD, Cazzola M, Galanello R, Marchetti M, Piga A, Tura S. Italian Society of Hematology practice guidelines for the management of iron overload in thalassemia major and related disorders. Haematologica 2008;93(5):741-52. doi: 10.3324/haematol.12413.

7. Jenkitkasemwong S, Wang CY, Coffey R, Zhang W, Chan A, Biel T, Kim JS, Hojyo S, Fukada T, Knutson MD. SLC39A14 Is Required for the Development of Hepatocellular Iron Overload in Murine Models of Hereditary Hemochromatosis. Cell Metab 2015;22(1):138-50. doi: 10.1016/j.cmet.2015.05.002.

8. Carpenter CE, Mahoney AW. Contributions of heme and nonheme iron to human nutrition. Crit Rev Food Sci Nutr 1992;31(4):333-67. doi: 10.1080/10408399209527576.

9. Bjorn-Rasmussen E, Hallberg L, Isaksson B, Arvidsson B. Food iron absorption in man. Applications of the two-pool extrinsic tag method to measure heme and nonheme iron absorption from the whole diet. J Clin Invest 1974;53(1):247-55. doi: 10.1172/JCI107545.

10. LeSage GD, Kost LJ, Barham SS, LaRusso NF. Biliary excretion of iron from hepatocyte lysosomes in the rat. A major excretory pathway in experimental iron overload. J Clin Invest 1986;77(1):90-7. doi: 10.1172/JCI112307.

Page 132: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

132

11. Gunshin H, Mackenzie B, Berger UV, Gunshin Y, Romero MF, Boron WF, Nussberger S, Gollan JL, Hediger MA. Cloning and characterization of a mammalian proton-coupled metal-ion transporter. Nature 1997;388(6641):482-8. doi: 10.1038/41343.

12. Shawki A, Anthony SR, Nose Y, Engevik MA, Niespodzany EJ, Barrientos T, Ohrvik H, Worrell RT, Thiele DJ, Mackenzie B. Intestinal DMT1 is critical for iron absorption in the mouse but is not required for the absorption of copper or manganese. Am J Physiol Gastrointest Liver Physiol 2015;309(8):G635-47. doi: 10.1152/ajpgi.00160.2015.

13. Gunshin H, Fujiwara Y, Custodio AO, Direnzo C, Robine S, Andrews NC. Slc11a2 is required for intestinal iron absorption and erythropoiesis but dispensable in placenta and liver. J Clin Invest 2005;115(5):1258-66. doi: 10.1172/JCI24356.

14. Choi J, Masaratana P, Latunde-Dada GO, Arno M, Simpson RJ, McKie AT. Duodenal reductase activity and spleen iron stores are reduced and erythropoiesis is abnormal in Dcytb knockout mice exposed to hypoxic conditions. J Nutr 2012;142(11):1929-34. doi: 10.3945/jn.112.160358.

15. Muto N, Ohta T, Suzuki T, Itoh N, Tanaka K. Evidence for the involvement of a muscarinic receptor in ascorbic acid secretion in the rat stomach. Biochem Pharmacol 1997;53(4):553-9.

16. McKie AT, Marciani P, Rolfs A, Brennan K, Wehr K, Barrow D, Miret S, Bomford A, Peters TJ, Farzaneh F, et al. A novel duodenal iron-regulated transporter, IREG1, implicated in the basolateral transfer of iron to the circulation. Mol Cell 2000;5(2):299-309.

17. Chiabrando D, Fiorito V, Marro S, Silengo L, Altruda F, Tolosano E. Cell-specific regulation of Ferroportin transcription following experimentally-induced acute anemia in mice. Blood Cells Mol Dis 2013;50(1):25-30. doi: 10.1016/j.bcmd.2012.08.002.

18. Taylor M, Qu A, Anderson ER, Matsubara T, Martin A, Gonzalez FJ, Shah YM. Hypoxia-inducible factor-2alpha mediates the adaptive increase of intestinal ferroportin during iron deficiency in mice. Gastroenterology 2011;140(7):2044-55. doi: 10.1053/j.gastro.2011.03.007.

19. Donovan A, Lima CA, Pinkus JL, Pinkus GS, Zon LI, Robine S, Andrews NC. The iron exporter ferroportin/Slc40a1 is essential for iron homeostasis. Cell Metab 2005;1(3):191-200. doi: 10.1016/j.cmet.2005.01.003.

Page 133: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

133

20. Pinkerton PH, Bannerman RM. Hereditary defect in iron absorption in mice. Nature 1967;216(5114):482-3.

21. Fuqua BK, Lu Y, Darshan D, Frazer DM, Wilkins SJ, Wolkow N, Bell AG, Hsu J, Yu CC, Chen H, et al. The multicopper ferroxidase hephaestin enhances intestinal iron absorption in mice. PLoS One 2014;9(6):e98792. doi: 10.1371/journal.pone.0098792.

22. Chen H, Su T, Attieh ZK, Fox TC, McKie AT, Anderson GJ, Vulpe CD. Systemic regulation of Hephaestin and Ireg1 revealed in studies of genetic and nutritional iron deficiency. Blood 2003;102(5):1893-9. doi: 10.1182/blood-2003-02-0347.

23. Yeh KY, Yeh M, Glass J. Interactions between ferroportin and hephaestin in rat enterocytes are reduced after iron ingestion. Gastroenterology 2011;141(1):292-9, 9 e1. doi: 10.1053/j.gastro.2011.03.059.

24. Mitchell CJ, Shawki A, Ganz T, Nemeth E, Mackenzie B. Functional properties of human ferroportin, a cellular iron exporter reactive also with cobalt and zinc. Am J Physiol Cell Physiol 2014;306(5):C450-9. doi: 10.1152/ajpcell.00348.2013.

25. Ohgami RS, Campagna DR, Greer EL, Antiochos B, McDonald A, Chen J, Sharp JJ, Fujiwara Y, Barker JE, Fleming MD. Identification of a ferrireductase required for efficient transferrin-dependent iron uptake in erythroid cells. Nat Genet 2005;37(11):1264-9. doi: 10.1038/ng1658.

26. Fleming MD, Romano MA, Su MA, Garrick LM, Garrick MD, Andrews NC. Nramp2 is mutated in the anemic Belgrade (b) rat: evidence of a role for Nramp2 in endosomal iron transport. Proc Natl Acad Sci U S A 1998;95(3):1148-53.

27. Zhao N, Gao J, Enns CA, Knutson MD. ZRT/IRT-like protein 14 (ZIP14) promotes the cellular assimilation of iron from transferrin. J Biol Chem 2010;285(42):32141-50. doi: 10.1074/jbc.M110.143248.

28. Wang CY, Jenkitkasemwong S, Duarte S, Sparkman BK, Shawki A, Mackenzie B, Knutson MD. ZIP8 Is an Iron and Zinc Transporter Whose Cell-surface Expression Is Up-regulated by Cellular Iron Loading. J Biol Chem 2012;287(41):34032-43. doi: 10.1074/jbc.M112.367284.

29. Garrick MD, Gniecko K, Liu Y, Cohan DS, Garrick LM. Transferrin and the transferrin cycle in Belgrade rat reticulocytes. J Biol Chem 1993;268(20):14867-74.

30. Fleming MD, Trenor CC, 3rd, Su MA, Foernzler D, Beier DR, Dietrich WF, Andrews NC. Microcytic anaemia mice have a mutation in Nramp2, a candidate iron transporter gene. Nat Genet 1997;16(4):383-6. doi: 10.1038/ng0897-383.

Page 134: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

134

31. Ganz T, Nemeth E. Hepcidin and iron homeostasis. Biochim Biophys Acta 2012;1823(9):1434-43. doi: 10.1016/j.bbamcr.2012.01.014.

32. Knutson MD, Oukka M, Koss LM, Aydemir F, Wessling-Resnick M. Iron release from macrophages after erythrophagocytosis is up-regulated by ferroportin 1 overexpression and down-regulated by hepcidin. Proc Natl Acad Sci U S A 2005;102(5):1324-8. doi: 10.1073/pnas.0409409102.

33. Zumerle S, Mathieu JR, Delga S, Heinis M, Viatte L, Vaulont S, Peyssonnaux C. Targeted disruption of hepcidin in the liver recapitulates the hemochromatotic phenotype. Blood 2014;123(23):3646-50. doi: 10.1182/blood-2014-01-550467.

34. Nemeth E, Tuttle MS, Powelson J, Vaughn MB, Donovan A, Ward DM, Ganz T, Kaplan J. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 2004;306(5704):2090-3. doi: 10.1126/science.1104742.

35. Frazer DM, Wilkins SJ, Becker EM, Vulpe CD, McKie AT, Trinder D, Anderson GJ. Hepcidin expression inversely correlates with the expression of duodenal iron transporters and iron absorption in rats. Gastroenterology 2002;123(3):835-44.

36. Pigeon C, Ilyin G, Courselaud B, Leroyer P, Turlin B, Brissot P, Loreal O. A new mouse liver-specific gene, encoding a protein homologous to human antimicrobial peptide hepcidin, is overexpressed during iron overload. J Biol Chem 2001;276(11):7811-9. doi: 10.1074/jbc.M008923200.

37. Nicolas G, Chauvet C, Viatte L, Danan JL, Bigard X, Devaux I, Beaumont C, Kahn A, Vaulont S. The gene encoding the iron regulatory peptide hepcidin is regulated by anemia, hypoxia, and inflammation. J Clin Invest 2002;110(7):1037-44. doi: 10.1172/JCI15686.

38. Zimmermann MB, Hurrell RF. Nutritional iron deficiency. Lancet 2007;370(9586):511-20. doi: 10.1016/S0140-6736(07)61235-5.

39. Lutter CK. Iron deficiency in young children in low-income countries and new approaches for its prevention. J Nutr 2008;138(12):2523-8. doi: 10.3945/jn.108.095406.

40. Iron deficiency--United States, 1999-2000. MMWR Morb Mortal Wkly Rep 2002;51(40):897-9.

41. Hunt JR, Zito CA, Erjavec J, Johnson LK. Severe or marginal iron deficiency affects spontaneous physical activity in rats. Am J Clin Nutr 1994;59(2):413-8.

42. Murray-Kolb LE, Beard JL. Iron treatment normalizes cognitive functioning in young women. Am J Clin Nutr 2007;85(3):778-87.

Page 135: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

135

43. Khan Y, Tisman G. Pica in iron deficiency: a case series. J Med Case Rep 2010;4:86. doi: 10.1186/1752-1947-4-86.

44. Loreal O, Deugnier Y, Moirand R, Lauvin L, Guyader D, Jouanolle H, Turlin B, Lescoat G, Brissot P. Liver fibrosis in genetic hemochromatosis. Respective roles of iron and non-iron-related factors in 127 homozygous patients. J Hepatol 1992;16(1-2):122-7.

45. McClain DA, Abraham D, Rogers J, Brady R, Gault P, Ajioka R, Kushner JP. High prevalence of abnormal glucose homeostasis secondary to decreased insulin secretion in individuals with hereditary haemochromatosis. Diabetologia 2006;49(7):1661-9. doi: 10.1007/s00125-006-0200-0.

46. Niederau C, Fischer R, Sonnenberg A, Stremmel W, Trampisch HJ, Strohmeyer G. Survival and causes of death in cirrhotic and in noncirrhotic patients with primary hemochromatosis. N Engl J Med 1985;313(20):1256-62. doi: 10.1056/NEJM198511143132004.

47. Feder JN, Gnirke A, Thomas W, Tsuchihashi Z, Ruddy DA, Basava A, Dormishian F, Domingo R, Jr., Ellis MC, Fullan A, et al. A novel MHC class I-like gene is mutated in patients with hereditary haemochromatosis. Nat Genet 1996;13(4):399-408. doi: 10.1038/ng0896-399.

48. Goswami T, Andrews NC. Hereditary hemochromatosis protein, HFE, interaction with transferrin receptor 2 suggests a molecular mechanism for mammalian iron sensing. J Biol Chem 2006;281(39):28494-8. doi: 10.1074/jbc.C600197200.

49. Gao J, Chen J, Kramer M, Tsukamoto H, Zhang AS, Enns CA. Interaction of the hereditary hemochromatosis protein HFE with transferrin receptor 2 is required for transferrin-induced hepcidin expression. Cell Metab 2009;9(3):217-27. doi: 10.1016/j.cmet.2009.01.010.

50. Phatak PD, Ryan DH, Cappuccio J, Oakes D, Braggins C, Provenzano K, Eberly S, Sham RL. Prevalence and penetrance of HFE mutations in 4865 unselected primary care patients. Blood Cells Mol Dis 2002;29(1):41-7.

51. Papanikolaou G, Samuels ME, Ludwig EH, MacDonald ML, Franchini PL, Dube MP, Andres L, MacFarlane J, Sakellaropoulos N, Politou M, et al. Mutations in HFE2 cause iron overload in chromosome 1q-linked juvenile hemochromatosis. Nat Genet 2004;36(1):77-82. doi: 10.1038/ng1274.

52. Niederkofler V, Salie R, Arber S. Hemojuvelin is essential for dietary iron sensing, and its mutation leads to severe iron overload. J Clin Invest 2005;115(8):2180-6. doi: 10.1172/JCI25683.

Page 136: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

136

53. Roetto A, Papanikolaou G, Politou M, Alberti F, Girelli D, Christakis J, Loukopoulos D, Camaschella C. Mutant antimicrobial peptide hepcidin is associated with severe juvenile hemochromatosis. Nat Genet 2003;33(1):21-2. doi: 10.1038/ng1053.

54. De Gobbi M, Caruso R, Daraio F, Chianale F, Pinto RM, Longo F, Piga A, Camaschella C. Diagnosis of juvenile hemochromatosis in an 11-year-old child combining genetic analysis and non-invasive liver iron quantitation. Eur J Pediatr 2003;162(2):96-9. doi: 10.1007/s00431-002-1114-6.

55. Adams PC, Deugnier Y, Moirand R, Brissot P. The relationship between iron overload, clinical symptoms, and age in 410 patients with genetic hemochromatosis. Hepatology 1997;25(1):162-6. doi: 10.1002/hep.510250130.

56. Lee PL, Beutler E, Rao SV, Barton JC. Genetic abnormalities and juvenile hemochromatosis: mutations of the HJV gene encoding hemojuvelin. Blood 2004;103(12):4669-71. doi: 10.1182/blood-2004-01-0072.

57. Camaschella C, Roetto A, Cali A, De Gobbi M, Garozzo G, Carella M, Majorano N, Totaro A, Gasparini P. The gene TFR2 is mutated in a new type of haemochromatosis mapping to 7q22. Nat Genet 2000;25(1):14-5. doi: 10.1038/75534.

58. Majore S, Milano F, Binni F, Stuppia L, Cerrone A, Tafuri A, De Bernardo C, Palka G, Grammatico P. Homozygous p.M172K mutation of the TFR2 gene in an Italian family with type 3 hereditary hemochromatosis and early onset iron overload. Haematologica 2006;91(8 Suppl):ECR33.

59. Le Gac G, Mons F, Jacolot S, Scotet V, Ferec C, Frebourg T. Early onset hereditary hemochromatosis resulting from a novel TFR2 gene nonsense mutation (R105X) in two siblings of north French descent. Br J Haematol 2004;125(5):674-8. doi: 10.1111/j.1365-2141.2004.04950.x.

60. Altamura S, Kessler R, Grone HJ, Gretz N, Hentze MW, Galy B, Muckenthaler MU. Resistance of ferroportin to hepcidin binding causes exocrine pancreatic failure and fatal iron overload. Cell Metab 2014;20(2):359-67. doi: 10.1016/j.cmet.2014.07.007.

61. Sham RL, Phatak PD, Nemeth E, Ganz T. Hereditary hemochromatosis due to resistance to hepcidin: high hepcidin concentrations in a family with C326S ferroportin mutation. Blood 2009;114(2):493-4. doi: 10.1182/blood-2009-04-216226.

62. Schimanski LM, Drakesmith H, Merryweather-Clarke AT, Viprakasit V, Edwards JP, Sweetland E, Bastin JM, Cowley D, Chinthammitr Y, Robson KJ, et al. In vitro functional analysis of human ferroportin (FPN) and hemochromatosis-associated FPN mutations. Blood 2005;105(10):4096-102. doi: 10.1182/blood-2004-11-4502.

Page 137: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

137

63. Montosi G, Donovan A, Totaro A, Garuti C, Pignatti E, Cassanelli S, Trenor CC, Gasparini P, Andrews NC, Pietrangelo A. Autosomal-dominant hemochromatosis is associated with a mutation in the ferroportin (SLC11A3) gene. J Clin Invest 2001;108(4):619-23. doi: 10.1172/JCI13468.

64. Jarjour RA, Murad H, Moasses F, Al-Achkar W. Molecular update of beta-thalassemia mutations in the Syrian population: identification of rare beta-thalassemia mutations. Hemoglobin 2014;38(4):272-6. doi: 10.3109/03630269.2014.912661.

65. Hendy OM, Allam M, Allam A, Attia MH, El Taher S, Eldin MM, Ali A. Hepcidin levels and iron status in beta-thalassemia major patients with hepatitis C virus infection. Egypt J Immunol 2010;17(2):33-44.

66. Kattamis A, Papassotiriou I, Palaiologou D, Apostolakou F, Galani A, Ladis V, Sakellaropoulos N, Papanikolaou G. The effects of erythropoetic activity and iron burden on hepcidin expression in patients with thalassemia major. Haematologica 2006;91(6):809-12.

67. Pippard MJ, Callender ST, Warner GT, Weatherall DJ. Iron absorption and loading in beta-thalassaemia intermedia. Lancet 1979;2(8147):819-21.

68. Borgna-Pignatti C, Cappellini MD, De Stefano P, Del Vecchio GC, Forni GL, Gamberini MR, Ghilardi R, Origa R, Piga A, Romeo MA, et al. Survival and complications in thalassemia. Ann N Y Acad Sci 2005;1054:40-7. doi: 10.1196/annals.1345.006.

69. Bothwell TH, Seftel H, Jacobs P, Torrance JD, Baumslag N. Iron Overload in Bantu Subjects; Studies on the Availability of Iron in Bantu Beer. Am J Clin Nutr 1964;14:47-51.

70. Moyo VM, Mandishona E, Hasstedt SJ, Gangaidzo IT, Gomo ZA, Khumalo H, Saungweme T, Kiire CF, Paterson AC, Bloom P, et al. Evidence of genetic transmission in African iron overload. Blood 1998;91(3):1076-82.

71. Gordeuk VR, Caleffi A, Corradini E, Ferrara F, Jones RA, Castro O, Onyekwere O, Kittles R, Pignatti E, Montosi G, et al. Iron overload in Africans and African-Americans and a common mutation in the SCL40A1 (ferroportin 1) gene. Blood Cells Mol Dis 2003;31(3):299-304.

72. McNamara L, Gordeuk VR, MacPhail AP. Ferroportin (Q248H) mutations in African families with dietary iron overload. J Gastroenterol Hepatol 2005;20(12):1855-8. doi: 10.1111/j.1440-1746.2005.03930.x.

73. Bradbury MW, Raja K, Ueda F. Contrasting uptakes of 59Fe into spleen, liver, kidney and some other soft tissues in normal and hypotransferrinaemic mice. Influence of an antibody against the transferrin receptor. Biochem Pharmacol 1994;47(6):969-74.

Page 138: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

138

74. Craven CM, Alexander J, Eldridge M, Kushner JP, Bernstein S, Kaplan J. 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 1987;84(10):3457-61.

75. Pinilla-Tenas JJ, Sparkman BK, Shawki A, Illing AC, Mitchell CJ, Zhao N, Liuzzi JP, Cousins RJ, Knutson MD, Mackenzie B. 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 2011;301(4):C862-71. doi: 10.1152/ajpcell.00479.2010.

76. Mackenzie B, Takanaga H, Hubert N, Rolfs A, Hediger MA. Functional properties of multiple isoforms of human divalent metal-ion transporter 1 (DMT1). Biochem J 2007;403(1):59-69. doi: 10.1042/BJ20061290.

77. Hubert N, Hentze MW. Previously uncharacterized isoforms of divalent metal transporter (DMT)-1: implications for regulation and cellular function. Proc Natl Acad Sci U S A 2002;99(19):12345-50. doi: 10.1073/pnas.192423399.

78. Casey JL, Hentze MW, Koeller DM, Caughman SW, Rouault TA, Klausner RD, Harford JB. Iron-responsive elements: regulatory RNA sequences that control mRNA levels and translation. Science 1988;240(4854):924-8.

79. Lee PL, Gelbart T, West C, Halloran C, Beutler E. The human Nramp2 gene: characterization of the gene structure, alternative splicing, promoter region and polymorphisms. Blood Cells Mol Dis 1998;24(2):199-215. doi: 10.1006/bcmd.1998.0186.

80. Tabuchi M, Tanaka N, Nishida-Kitayama J, Ohno H, Kishi F. Alternative splicing regulates the subcellular localization of divalent metal transporter 1 isoforms. Mol Biol Cell 2002;13(12):4371-87. doi: 10.1091/mbc.E02-03-0165.

81. Canonne-Hergaux F, Gruenheid S, Ponka P, Gros P. Cellular and subcellular localization of the Nramp2 iron transporter in the intestinal brush border and regulation by dietary iron. Blood 1999;93(12):4406-17.

82. Nam H, Wang CY, Zhang L, Zhang W, Hojyo S, Fukada T, Knutson M. 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 2013. doi: 10.3324/haematol.2012.072314.

83. Wang CY, Knutson MD. Hepatocyte divalent metal-ion transporter-1 is dispensable for hepatic iron accumulation and non-transferrin-bound iron uptake in mice. Hepatology 2013;58(2):788-98. doi: 10.1002/hep.26401.

84. Giorgi G, Roque ME. Iron overload induces changes of pancreatic and duodenal divalent metal transporter 1 and prohepcidin expression in mice. Acta Histochem 2014;116(2):354-62. doi: 10.1016/j.acthis.2013.08.013.

Page 139: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

139

85. Koch RO, Zoller H, Theuri I, Obrist P, Egg G, Strohmayer W, Vogel W, Weiss G. Distribution of DMT 1 within the human glandular system. Histol Histopathol 2003;18(4):1095-101.

86. Hansen JB, Tonnesen MF, Madsen AN, Hagedorn PH, Friberg J, Grunnet LG, Heller RS, Nielsen AO, Storling J, Baeyens L, et al. Divalent Metal Transporter 1 Regulates Iron-Mediated ROS and Pancreatic beta Cell Fate in Response to Cytokines. Cell Metab 2012;16(4):449-61. doi: 10.1016/j.cmet.2012.09.001.

87. Zhao H, Eide D. The ZRT2 gene encodes the low affinity zinc transporter in Saccharomyces cerevisiae. J Biol Chem 1996;271(38):23203-10.

88. Zhao H, Eide D. The yeast ZRT1 gene encodes the zinc transporter protein of a high-affinity uptake system induced by zinc limitation. Proc Natl Acad Sci U S A 1996;93(6):2454-8.

89. Eide D, Broderius M, Fett J, Guerinot ML. A novel iron-regulated metal transporter from plants identified by functional expression in yeast. Proc Natl Acad Sci U S A 1996;93(11):5624-8.

90. Liuzzi JP, Aydemir F, Nam H, Knutson MD, Cousins RJ. Zip14 (Slc39a14) mediates non-transferrin-bound iron uptake into cells. Proc Natl Acad Sci U S A 2006;103(37):13612-7. doi: 10.1073/pnas.0606424103.

91. Liuzzi JP, Lichten LA, Rivera S, Blanchard RK, Aydemir TB, Knutson MD, Ganz T, Cousins RJ. Interleukin-6 regulates the zinc transporter Zip14 in liver and contributes to the hypozincemia of the acute-phase response. Proc Natl Acad Sci U S A 2005;102(19):6843-8. doi: 10.1073/pnas.0502257102.

92. Taylor KM, Morgan HE, Johnson A, Nicholson RI. Structure-function analysis of a novel member of the LIV-1 subfamily of zinc transporters, ZIP14. FEBS Lett 2005;579(2):427-32. doi: 10.1016/j.febslet.2004.12.006.

93. Nunez MT, Gaete V, Watkins JA, Glass J. Mobilization of iron from endocytic vesicles. The effects of acidification and reduction. J Biol Chem 1990;265(12):6688-92.

94. Nebert DW, Galvez-Peralta M, Hay EB, Li H, Johansson E, Yin C, Wang B, He L, Soleimani M. ZIP14 and ZIP8 zinc/bicarbonate symporters in Xenopus oocytes: characterization of metal uptake and inhibition. Metallomics 2012;4(11):1218-25. doi: 10.1039/c2mt20177a.

95. Zhao N, Zhang AS, Worthen C, Knutson MD, Enns CA. An iron-regulated and glycosylation-dependent proteasomal degradation pathway for the plasma membrane metal transporter ZIP14. Proc Natl Acad Sci U S A 2014;111(25):9175-80. doi: 10.1073/pnas.1405355111.

Page 140: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

140

96. Parkes JG, Randell EW, Olivieri NF, Templeton DM. Modulation by iron loading and chelation of the uptake of non-transferrin-bound iron by human liver cells. Biochim Biophys Acta 1995;1243(3):373-80.

97. Scheiber-Mojdehkar B, Zimmermann I, Dresow B, Goldenberg H. Differential response of non-transferrin bound iron uptake in rat liver cells on long-term and short-term treatment with iron. J Hepatol 1999;31(1):61-70.

98. Chua AC, Olynyk JK, Leedman PJ, Trinder D. Nontransferrin-bound iron uptake by hepatocytes is increased in the Hfe knockout mouse model of hereditary hemochromatosis. Blood 2004;104(5):1519-25. doi: 10.1182/blood-2003-11-3872.

99. Jenkitkasemwong S, Wang CY, Mackenzie B, Knutson MD. Physiologic implications of metal-ion transport by ZIP14 and ZIP8. BioMetals 2012;25(4):643-55. doi: 10.1007/s10534-012-9526-x.

100. Begum NA, Kobayashi M, Moriwaki Y, Matsumoto M, Toyoshima K, Seya T. Mycobacterium bovis BCG cell wall and lipopolysaccharide induce a novel gene, BIGM103, encoding a 7-TM protein: identification of a new protein family having Zn-transporter and Zn-metalloprotease signatures. Genomics 2002;80(6):630-45.

101. He L, Girijashanker K, Dalton TP, Reed J, Li H, Soleimani M, Nebert DW. ZIP8, member of the solute-carrier-39 (SLC39) metal-transporter family: characterization of transporter properties. Mol Pharmacol 2006;70(1):171-80. doi: 10.1124/mol.106.024521.

102. Liu Z, Li H, Soleimani M, Girijashanker K, Reed JM, He L, Dalton TP, Nebert DW. Cd2+ versus Zn2+ uptake by the ZIP8 HCO3--dependent symporter: kinetics, electrogenicity and trafficking. Biochem Biophys Res Commun 2008;365(4):814-20. doi: 10.1016/j.bbrc.2007.11.067.

103. Pae EK, Kim G. Insulin production hampered by intermittent hypoxia via impaired zinc homeostasis. PLoS One 2014;9(2):e90192. doi: 10.1371/journal.pone.0090192.

104. Galvez-Peralta M, He L, Jorge-Nebert LF, Wang B, Miller ML, Eppert BL, Afton S, Nebert DW. ZIP8 zinc transporter: indispensable role for both multiple-organ organogenesis and hematopoiesis in utero. PLoS One 2012;7(5):e36055. doi: 10.1371/journal.pone.0036055.

105. Tsushima RG, Wickenden AD, Bouchard RA, Oudit GY, Liu PP, Backx PH. Modulation of iron uptake in heart by L-type Ca2+ channel modifiers: possible implications in iron overload. Circ Res 1999;84(11):1302-9.

Page 141: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

141

106. Oudit GY, Sun H, Trivieri MG, Koch SE, Dawood F, Ackerley C, Yazdanpanah M, Wilson GJ, Schwartz A, Liu PP, et al. L-type Ca2+ channels provide a major pathway for iron entry into cardiomyocytes in iron-overload cardiomyopathy. Nat Med 2003;9(9):1187-94. doi: 10.1038/nm920.

107. Kumfu S, Chattipakorn S, Chinda K, Fucharoen S, Chattipakorn N. T-type calcium channel blockade improves survival and cardiovascular function in thalassemic mice. Eur J Haematol 2012;88(6):535-48. doi: 10.1111/j.1600-0609.2012.01779.x.

108. Taylor JT, Huang L, Keyser BM, Zhuang H, Clarkson CW, Li M. Role of high-voltage-activated calcium channels in glucose-regulated beta-cell calcium homeostasis and insulin release. Am J Physiol Endocrinol Metab 2005;289(5):E900-8. doi: 10.1152/ajpendo.00101.2005.

109. Bhattacharjee A, Whitehurst RM, Jr., Zhang M, Wang L, Li M. T-type calcium channels facilitate insulin secretion by enhancing general excitability in the insulin-secreting beta-cell line, INS-1. Endocrinology 1997;138(9):3735-40. doi: 10.1210/endo.138.9.5390.

110. Cheng YJ, Imperatore G, Geiss LS, Wang J, Saydah SH, Cowie CC, Gregg EW. Secular changes in the age-specific prevalence of diabetes among U.S. adults: 1988-2010. Diabetes Care 2013;36(9):2690-6. doi: 10.2337/dc12-2074.

111. Lusignan S, Sismanidis C, Carey IM, DeWilde S, Richards N, Cook DG. Trends in the prevalence and management of diagnosed type 2 diabetes 1994-2001 in England and Wales. BMC Fam Pract 2005;6(1):13. doi: 10.1186/1471-2296-6-13.

112. Finch SC, Finch CA. Idiopathic hemochromatosis, an iron storage disease. A. Iron metabolism in hemochromatosis. Medicine (Baltimore) 1955;34(4):381-430.

113. Dymock IW, Cassar J, Pyke DA, Oakley WG, Williams R. Observations on the pathogenesis, complications and treatment of diabetes in 115 cases of haemochromatosis. Am J Med 1972;52(2):203-10.

114. Buysschaert M, Paris I, Selvais P, Hermans MP. Clinical aspects of diabetes secondary to idiopathic haemochromatosis in French-speaking Belgium. Diabetes Metab 1997;23(4):308-13.

115. Moirand R, Adams PC, Bicheler V, Brissot P, Deugnier Y. Clinical features of genetic hemochromatosis in women compared with men. Ann Intern Med 1997;127(2):105-10.

116. O'Sullivan EP, McDermott JH, Murphy MS, Sen S, Walsh CH. Declining prevalence of diabetes mellitus in hereditary haemochromatosis--the result of earlier diagnosis. Diabetes Res Clin Pract 2008;81(3):316-20. doi: 10.1016/j.diabres.2008.05.001.

Page 142: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

142

117. Hramiak IM, Finegood DT, Adams PC. Factors affecting glucose tolerance in hereditary hemochromatosis. Clin Invest Med 1997;20(2):110-8.

118. Li MJ, Peng SS, Lu MY, Chang HH, Yang YL, Jou ST, Lin DT, Lin KH. Diabetes mellitus in patients with thalassemia major. Pediatr Blood Cancer 2014;61(1):20-4. doi: 10.1002/pbc.24754.

119. Gamberini MR, Fortini M, De Sanctis V, Gilli G, Testa MR. Diabetes mellitus and impaired glucose tolerance in thalassaemia major: incidence, prevalence, risk factors and survival in patients followed in the Ferrara Center. Pediatr Endocrinol Rev 2004;2 Suppl 2:285-91.

120. De Sanctis V, Zurlo MG, Senesi E, Boffa C, Cavallo L, Di Gregorio F. Insulin dependent diabetes in thalassaemia. Arch Dis Child 1988;63(1):58-62.

121. Jehn ML, Guallar E, Clark JM, Couper D, Duncan BB, Ballantyne CM, Hoogeveen RC, Harris ZL, Pankow JS. A prospective study of plasma ferritin level and incident diabetes: the Atherosclerosis Risk in Communities (ARIC) Study. Am J Epidemiol 2007;165(9):1047-54. doi: 10.1093/aje/kwk093.

122. Le TD, Bae S, Ed Hsu C, Singh KP, Blair SN, Shang N. Effects of Cardiorespiratory Fitness on Serum Ferritin Concentration and Incidence of Type 2 Diabetes: Evidence from the Aerobics Center Longitudinal Study (ACLS). Rev Diabet Stud 2008;5(4):245-52. doi: 10.1900/RDS.2008.5.245.

123. Montonen J, Boeing H, Steffen A, Lehmann R, Fritsche A, Joost HG, Schulze MB, Pischon T. Body iron stores and risk of type 2 diabetes: results from the European Prospective Investigation into Cancer and Nutrition (EPIC)-Potsdam study. Diabetologia 2012;55(10):2613-21. doi: 10.1007/s00125-012-2633-y.

124. Salonen JT, Tuomainen TP, Nyyssonen K, Lakka HM, Punnonen K. Relation between iron stores and non-insulin dependent diabetes in men: case-control study. BMJ 1998;317(7160):727.

125. Rajpathak SN, Wylie-Rosett J, Gunter MJ, Negassa A, Kabat GC, Rohan TE, Crandall J. Biomarkers of body iron stores and risk of developing type 2 diabetes. Diabetes Obes Metab 2009;11(5):472-9. doi: 10.1111/j.1463-1326.2008.00985.x.

126. Rogers JT. Ferritin translation by interleukin-1and interleukin-6: the role of sequences upstream of the start codons of the heavy and light subunit genes. Blood 1996;87(6):2525-37.

127. Pitsavos C, Tampourlou M, Panagiotakos DB, Skoumas Y, Chrysohoou C, Nomikos T, Stefanadis C. Association Between Low-Grade Systemic Inflammation and Type 2 Diabetes Mellitus Among Men and Women from the ATTICA Study. Rev Diabet Stud 2007;4(2):98-104. doi: 10.1900/RDS.2007.4.98.

Page 143: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

143

128. Looker AC, Loyevsky M, Gordeuk VR. Increased serum transferrin saturation is associated with lower serum transferrin receptor concentration. Clin Chem 1999;45(12):2191-9.

129. Ferguson BJ, Skikne BS, Simpson KM, Baynes RD, Cook JD. Serum transferrin receptor distinguishes the anemia of chronic disease from iron deficiency anemia. J Lab Clin Med 1992;119(4):385-90.

130. Jiang R, Manson JE, Meigs JB, Ma J, Rifai N, Hu FB. Body iron stores in relation to risk of type 2 diabetes in apparently healthy women. JAMA 2004;291(6):711-7. doi: 10.1001/jama.291.6.711.

131. Lee DH, Liu DY, Jacobs DR, Jr., Shin HR, Song K, Lee IK, Kim B, Hider RC. Common presence of non-transferrin-bound iron among patients with type 2 diabetes. Diabetes Care 2006;29(5):1090-5. doi: 10.2337/diacare.2951090.

132. Rahier J, Loozen S, Goebbels RM, Abrahem M. The haemochromatotic human pancreas: a quantitative immunohistochemical and ultrastructural study. Diabetologia 1987;30(1):5-12.

133. Kishimoto M, Endo H, Hagiwara S, Miwa A, Noda M. Immunohistochemical findings in the pancreatic islets of a patient with transfusional iron overload and diabetes: case report. J Med Invest 2010;57(3-4):345-9.

134. Lu JP, Hayashi K. Selective iron deposition in pancreatic islet B cells of transfusional iron-overloaded autopsy cases. Pathol Int 1994;44(3):194-9.

135. Noetzli LJ, Papudesi J, Coates TD, Wood JC. Pancreatic iron loading predicts cardiac iron loading in thalassemia major. Blood 2009;114(19):4021-6. doi: 10.1182/blood-2009-06-225615.

136. Ramey G, Faye A, Durel B, Viollet B, Vaulont S. Iron overload in Hepc1(-/-) mice is not impairing glucose homeostasis. FEBS Lett 2007;581(5):1053-7. doi: 10.1016/j.febslet.2007.02.002.

137. Horne WI, Tandler B, Dubick MA, Niemela O, Brittenham GM, Tsukamoto H. Iron overload in the rat pancreas following portacaval shunting and dietary iron supplementation. Exp Mol Pathol 1997;64(2):90-102. doi: 10.1006/exmp.1997.2212.

138. Nick H, Allegrini PR, Fozard L, Junker U, Rojkjaer L, Salie R, Niederkofler V, O'Reilly T. Deferasirox reduces iron overload in a murine model of juvenile hemochromatosis. Exp Biol Med (Maywood) 2009;234(5):492-503. doi: 10.3181/0811-RM-337.

Page 144: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

144

139. Coffey R, Nam H, Knutson MD. Microarray analysis of rat pancreas reveals altered expression of Alox15 and regenerating islet-derived genes in response to iron deficiency and overload. PLoS One 2014;9(1):e86019. doi: 10.1371/journal.pone.0086019.

140. Iancu TC, Ward RJ, Peters TJ. Ultrastructural changes in the pancreas of carbonyl iron-fed rats. J Pediatr Gastroenterol Nutr 1990;10(1):95-101.

141. Whittaker P, Hines FA, Robl MG, Dunkel VC. Histopathological evaluation of liver, pancreas, spleen, and heart from iron-overloaded Sprague-Dawley rats. Toxicol Pathol 1996;24(5):558-63.

142. Whittaker P, Dunkel VC, Bucci TJ, Kusewitt DF, Thurman JD, Warbritton A, Wolff GL. Genome-linked toxic responses to dietary iron overload. Toxicol Pathol 1997;25(6):556-64.

143. Awai M, Narasaki M, Yamanoi Y, Seno S. Induction of diabetes in animals by parenteral administration of ferric nitrilotriacetate. A model of experimental hemochromatosis. Am J Pathol 1979;95(3):663-73.

144. Lu JP, Hayashi K, Okada S, Awai M. Transferrin receptors and selective iron deposition in pancreatic B cells of iron-overloaded rats. Acta Pathol Jpn 1991;41(9):647-52.

145. Weyer C, Tataranni PA, Bogardus C, Pratley RE. Insulin resistance and insulin secretory dysfunction are independent predictors of worsening of glucose tolerance during each stage of type 2 diabetes development. Diabetes Care 2001;24(1):89-94.

146. Cooksey RC, Jouihan HA, Ajioka RS, Hazel MW, Jones DL, Kushner JP, McClain DA. Oxidative stress, beta-cell apoptosis, and decreased insulin secretory capacity in mouse models of hemochromatosis. Endocrinology 2004;145(11):5305-12. doi: 10.1210/en.2004-0392.

147. Abraham D, Rogers J, Gault P, Kushner JP, McClain DA. Increased insulin secretory capacity but decreased insulin sensitivity after correction of iron overload by phlebotomy in hereditary haemochromatosis. Diabetologia 2006;49(11):2546-51. doi: 10.1007/s00125-006-0445-7.

148. Hatunic M, Finucane FM, Norris S, Pacini G, Nolan JJ. Glucose metabolism after normalization of markers of iron overload by venesection in subjects with hereditary hemochromatosis. Metabolism 2010;59(12):1811-5. doi: 10.1016/j.metabol.2010.06.005.

149. Fernandez-Real JM, Penarroja G, Castro A, Garcia-Bragado F, Hernandez-Aguado I, Ricart W. Blood letting in high-ferritin type 2 diabetes: effects on insulin sensitivity and beta-cell function. Diabetes 2002;51(4):1000-4.

Page 145: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

145

150. Dmochowski K, Finegood DT, Francombe W, Tyler B, Zinman B. Factors determining glucose tolerance in patients with thalassemia major. J Clin Endocrinol Metab 1993;77(2):478-83. doi: 10.1210/jcem.77.2.8345055.

151. Merkel PA, Simonson DC, Amiel SA, Plewe G, Sherwin RS, Pearson HA, Tamborlane WV. Insulin resistance and hyperinsulinemia in patients with thalassemia major treated by hypertransfusion. N Engl J Med 1988;318(13):809-14. doi: 10.1056/NEJM198803313181303.

152. Messina MF, Lombardo F, Meo A, Miceli M, Wasniewska M, Valenzise M, Ruggeri C, Arrigo T, De Luca F. Three-year prospective evaluation of glucose tolerance, beta-cell function and peripheral insulin sensitivity in non-diabetic patients with thalassemia major. J Endocrinol Invest 2002;25(6):497-501.

153. Jaruratanasirikul S, Chareonmuang R, Wongcharnchailert M, Laosombat V, Sangsupavanich P, Leetanaporn K. Prevalence of impaired glucose metabolism in beta-thalassemic children receiving hypertransfusions with a suboptimal dosage of iron-chelating therapy. Eur J Pediatr 2008;167(8):873-6. doi: 10.1007/s00431-007-0602-0.

154. Soliman AT, el Banna N, alSalmi I, Asfour M. Insulin and glucagon responses to provocation with glucose and arginine in prepubertal children with thalassemia major before and after long-term blood transfusion. J Trop Pediatr 1996;42(5):291-6.

155. Farmaki K, Angelopoulos N, Anagnostopoulos G, Gotsis E, Rombopoulos G, Tolis G. Effect of enhanced iron chelation therapy on glucose metabolism in patients with beta-thalassaemia major. Br J Haematol 2006;134(4):438-44. doi: 10.1111/j.1365-2141.2006.06203.x.

156. Lenzen S, Drinkgern J, Tiedge M. Low antioxidant enzyme gene expression in pancreatic islets compared with various other mouse tissues. Free Radic Biol Med 1996;20(3):463-6.

157. Robertson RP, Harmon JS. Pancreatic islet beta-cell and oxidative stress: the importance of glutathione peroxidase. FEBS Lett 2007;581(19):3743-8. doi: 10.1016/j.febslet.2007.03.087.

158. Tonooka N, Oseid E, Zhou H, Harmon JS, Robertson RP. Glutathione peroxidase protein expression and activity in human islets isolated for transplantation. Clin Transplant 2007;21(6):767-72. doi: 10.1111/j.1399-0012.2007.00736.x.

159. Mossner J, Logsdon CD, Williams JA, Goldfine ID. Insulin, via its own receptor, regulates growth and amylase synthesis in pancreatic acinar AR42J cells. Diabetes 1985;34(9):891-7.

Page 146: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

146

160. Fonseca V, Berger LA, Beckett AG, Dandona P. Size of pancreas in diabetes mellitus: a study based on ultrasound. Br Med J (Clin Res Ed) 1985;291(6504):1240-1.

161. Huang J, Gabrielsen JS, Cooksey RC, Luo B, Boros LG, Jones DL, Jouihan HA, Soesanto Y, Knecht L, Hazel MW, et al. Increased glucose disposal and AMP-dependent kinase signaling in a mouse model of hemochromatosis. J Biol Chem 2007;282(52):37501-7. doi: 10.1074/jbc.M703625200.

162. Jouihan HA, Cobine PA, Cooksey RC, Hoagland EA, Boudina S, Abel ED, Winge DR, McClain DA. Iron-mediated inhibition of mitochondrial manganese uptake mediates mitochondrial dysfunction in a mouse model of hemochromatosis. Mol Med 2008;14(3-4):98-108. doi: 10.2119/2007-00114.Jouihan.

163. Cooksey RC, Jones D, Gabrielsen S, Huang J, Simcox JA, Luo B, Soesanto Y, Rienhoff H, Abel ED, McClain DA. Dietary iron restriction or iron chelation protects from diabetes and loss of beta-cell function in the obese (ob/ob lep-/-) mouse. Am J Physiol Endocrinol Metab 2010;298(6):E1236-43. doi: 10.1152/ajpendo.00022.2010.

164. Minamiyama Y, Takemura S, Kodai S, Shinkawa H, Tsukioka T, Ichikawa H, Naito Y, Yoshikawa T, Okada S. Iron restriction improves type 2 diabetes mellitus in Otsuka Long-Evans Tokushima fatty rats. Am J Physiol Endocrinol Metab 2010;298(6):E1140-9. doi: 10.1152/ajpendo.00620.2009.

165. Masuda Y, Ichii H, Vaziri ND. At pharmacologically relevant concentrations intravenous iron preparations cause pancreatic beta cell death. Am J Transl Res 2013;6(1):64-70.

166. Ashraf AP, Eason NB, Kabagambe EK, Haritha J, Meleth S, McCormick KL. Dietary iron intake in the first 4 months of infancy and the development of type 1 diabetes: a pilot study. Diabetol Metab Syndr 2010;2:58. doi: 10.1186/1758-5996-2-58.

167. Ellervik C, Mandrup-Poulsen T, Andersen HU, Tybjaerg-Hansen A, Frandsen M, Birgens H, Nordestgaard BG. Elevated transferrin saturation and risk of diabetes: three population-based studies. Diabetes Care 2011;34(10):2256-8. doi: 10.2337/dc11-0416.

168. Wang H, Li H, Jiang X, Shi W, Shen Z, Li M. Hepcidin is directly regulated by insulin and plays an important role in iron overload in streptozotocin-induced diabetic rats. Diabetes 2014;63(5):1506-18. doi: 10.2337/db13-1195.

169. Ellervik C, Mandrup-Poulsen T, Nordestgaard BG, Larsen LE, Appleyard M, Frandsen M, Petersen P, Schlichting P, Saermark T, Tybjaerg-Hansen A, et al. Prevalence of hereditary haemochromatosis in late-onset type 1 diabetes mellitus: a retrospective study. Lancet 2001;358(9291):1405-9. doi: 10.1016/S0140-6736(01)06526-6.

Page 147: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

147

170. Nomikos IN, Prowse SJ, Carotenuto P, Lafferty KJ. Combined treatment with nicotinamide and desferrioxamine prevents islet allograft destruction in NOD mice. Diabetes 1986;35(11):1302-4.

171. Bayer AL, Baliga P, Woodward JE. Transferrin receptor in T cell activation and transplantation. J Leukoc Biol 1998;64(1):19-24.

172. Lesley JF, Schulte RJ. Inhibition of cell growth by monoclonal anti-transferrin receptor antibodies. Mol Cell Biol 1985;5(8):1814-21.

173. Mangano K, Fagone P, Di Mauro M, Ascione E, Maiello V, Milicic T, Jotic A, Lalic NM, Saksida T, Stojanovic I, et al. The immunobiology of apotransferrin in type 1 diabetes. Clin Exp Immunol 2012;169(3):244-52. doi: DOI 10.1111/j.1365-2249.2012.04619.x.

174. Young SP, Bomford A, Williams R. The effect of the iron saturation of transferrin on its binding and uptake by rabbit reticulocytes. Biochem J 1984;219(2):505-10.

175. Tsunoo H, Sussman HH. Characterization of transferrin binding and specificity of the placental transferrin receptor. Arch Biochem Biophys 1983;225(1):42-54.

176. Nam H, Knutson MD. Effect of dietary iron deficiency and overload on the expression of ZIP metal-ion transporters in rat liver. BioMetals 2012;25(1):115-24. doi: 10.1007/s10534-011-9487-5.

177. Torrance JD, Bothwell TH. A simple technique for measuring storage iron concentrations in formalinised liver samples. S Afr J Med Sci 1968;33(1):9-11.

178. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc 2009;4(1):44-57. doi: 10.1038/nprot.2008.211.

179. Peyssonnaux C, Zinkernagel AS, Schuepbach RA, Rankin E, Vaulont S, Haase VH, Nizet V, Johnson RS. Regulation of iron homeostasis by the hypoxia-inducible transcription factors (HIFs). J Clin Invest 2007;117(7):1926-32. doi: 10.1172/JCI31370.

180. Altschul SF, Madden TL, Schaffer AA, Zhang J, Zhang Z, Miller W, Lipman DJ. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res 1997;25(17):3389-402.

181. MacGregor RR, Williams SJ, Tong PY, Kover K, Moore WV, Stehno-Bittel L. Small rat islets are superior to large islets in in vitro function and in transplantation outcomes. Am J Physiol Endocrinol Metab 2006;290(5):E771-9. doi: 10.1152/ajpendo.00097.2005.

Page 148: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

148

182. Hara M, Wang X, Kawamura T, Bindokas VP, Dizon RF, Alcoser SY, Magnuson MA, Bell GI. Transgenic mice with green fluorescent protein-labeled pancreatic beta -cells. Am J Physiol Endocrinol Metab 2003;284(1):E177-83. doi: 10.1152/ajpendo.00321.2002.

183. Moritz W, Meier F, Stroka DM, Giuliani M, Kugelmeier P, Nett PC, Lehmann R, Candinas D, Gassmann M, Weber M. Apoptosis in hypoxic human pancreatic islets correlates with HIF-1alpha expression. FASEB J 2002;16(7):745-7. doi: 10.1096/fj.01-0403fje.

184. Shin D, Jeon JH, Jeong M, Suh HW, Kim S, Kim HC, Moon OS, Kim YS, Chung JW, Yoon SR, et al. VDUP1 mediates nuclear export of HIF1alpha via CRM1-dependent pathway. Biochim Biophys Acta 2008;1783(5):838-48. doi: 10.1016/j.bbamcr.2007.10.012.

185. Borel MJ, Smith SH, Brigham DE, Beard JL. The impact of varying degrees of iron nutriture on several functional consequences of iron deficiency in rats. J Nutr 1991;121(5):729-36.

186. Kamei A, Watanabe Y, Ishijima T, Uehara M, Arai S, Kato H, Nakai Y, Abe K. Dietary iron-deficient anemia induces a variety of metabolic changes and even apoptosis in rat liver: a DNA microarray study. Physiol Genomics 2010;42(2):149-56. doi: 10.1152/physiolgenomics.00150.2009.

187. Huang J, Jones D, Luo B, Sanderson M, Soto J, Abel ED, Cooksey RC, McClain DA. Iron overload and diabetes risk: a shift from glucose to Fatty Acid oxidation and increased hepatic glucose production in a mouse model of hereditary hemochromatosis. Diabetes 2011;60(1):80-7. doi: 10.2337/db10-0593.

188. McDuffie M, Maybee NA, Keller SR, Stevens BK, Garmey JC, Morris MA, Kropf E, Rival C, Ma K, Carter JD, et al. Nonobese diabetic (NOD) mice congenic for a targeted deletion of 12/15-lipoxygenase are protected from autoimmune diabetes. Diabetes 2008;57(1):199-208. doi: 10.2337/db07-0830.

189. Leconet W, Petit P, Peraldi-Roux S, Bresson D. Nonviral Delivery of Small Interfering RNA Into Pancreas-associated Immune Cells Prevents Autoimmune Diabetes. Mol Ther 2012;20(12):2315-25. doi: 10.1038/mt.2012.190.

190. Collins JF, Hu Z, Ranganathan PN, Feng D, Garrick LM, Garrick MD, Browne RW. Induction of arachidonate 12-lipoxygenase (Alox15) in intestine of iron-deficient rats correlates with the production of biologically active lipid mediators. Am J Physiol Gastrointest Liver Physiol 2008;294(4):G948-62. doi: 10.1152/ajpgi.00274.2007.

191. Watanabe T, Yonemura Y, Yonekura H, Suzuki Y, Miyashita H, Sugiyama K, Moriizumi S, Unno M, Tanaka O, Kondo H, et al. Pancreatic beta-cell replication and amelioration of surgical diabetes by Reg protein. Proc Natl Acad Sci U S A 1994;91(9):3589-92.

Page 149: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

149

192. Lim JW, Song JY, Seo JY, Kim H, Kim KH. Role of pancreatitis-associated protein 1 on oxidative stress-induced cell death of pancreatic acinar cells. Ann N Y Acad Sci 2009;1171:545-8. doi: 10.1111/j.1749-6632.2009.04702.x.

193. Xiong X, Wang X, Li B, Chowdhury S, Lu Y, Srikant CB, Ning G, Liu JL. Pancreatic islet-specific overexpression of Reg3beta protein induced the expression of pro-islet genes and protected the mice against streptozotocin-induced diabetes mellitus. Am J Physiol Endocrinol Metab 2011;300(4):E669-80. doi: 10.1152/ajpendo.00600.2010.

194. Choi JH, Lee MY, Kim Y, Shim JY, Han SM, Lee KA, Choi YK, Jeon HM, Baek KH. Isolation of genes involved in pancreas regeneration by subtractive hybridization. Biol Chem 2010;391(9):1019-29. doi: 10.1515/BC.2010.101.

195. Weiss EP, Brown MD, Shuldiner AR, Hagberg JM. Fatty acid binding protein-2 gene variants and insulin resistance: gene and gene-environment interaction effects. Physiol Genomics 2002;10(3):145-57. doi: 10.1152/physiolgenomics.00070.2001.

196. Riaz S, Alam SS, Akhtar MW. Proteomic identification of human serum biomarkers in diabetes mellitus type 2. J Pharm Biomed Anal 2010;51(5):1103-7. doi: 10.1016/j.jpba.2009.11.016.

197. Mansego ML, Martinez F, Martinez-Larrad MT, Zabena C, Rojo G, Morcillo S, Soriguer F, Martin-Escudero JC, Serrano-Rios M, Redon J, et al. Common variants of the liver fatty acid binding protein gene influence the risk of type 2 diabetes and insulin resistance in Spanish population. PLoS One 2012;7(3):e31853. doi: 10.1371/journal.pone.0031853.

198. Bogan RL, Hennebold JD. The reverse cholesterol transport system as a potential mediator of luteolysis in the primate corpus luteum. Reproduction 2010;139(1):163-76. doi: 10.1530/REP-09-0005.

199. Gordon JI, Elshourbagy N, Lowe JB, Liao WS, Alpers DH, Taylor JM. Tissue specific expression and developmental regulation of two genes coding for rat fatty acid binding proteins. J Biol Chem 1985;260(4):1995-8.

200. Marin P, Rebuffe-Scrive M, Smith U, Bjorntorp P. Glucose uptake in human adipose tissue. Metabolism 1987;36(12):1154-60.

201. Brash AR. Lipoxygenases: occurrence, functions, catalysis, and acquisition of substrate. J Biol Chem 1999;274(34):23679-82.

202. Griffin ME, Marcucci MJ, Cline GW, Bell K, Barucci N, Lee D, Goodyear LJ, Kraegen EW, White MF, Shulman GI. Free fatty acid-induced insulin resistance is associated with activation of protein kinase C theta and alterations in the insulin signaling cascade. Diabetes 1999;48(6):1270-4.

Page 150: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

150

203. Baron AD, Brechtel G, Wallace P, Edelman SV. Rates and tissue sites of non-insulin- and insulin-mediated glucose uptake in humans. Am J Physiol 1988;255(6 Pt 1):E769-74.

204. Chen M, Yang ZD, Smith KM, Carter JD, Nadler JL. Activation of 12-lipoxygenase in proinflammatory cytokine-mediated beta cell toxicity. Diabetologia 2005;48(3):486-95. doi: 10.1007/s00125-005-1673-y.

205. Bleich D, Chen S, Zipser B, Sun D, Funk CD, Nadler JL. Resistance to type 1 diabetes induction in 12-lipoxygenase knockout mice. J Clin Invest 1999;103(10):1431-6. doi: 10.1172/JCI5241.

206. Dongiovanni P, Valenti L, Ludovica Fracanzani A, Gatti S, Cairo G, Fargion S. Iron depletion by deferoxamine up-regulates glucose uptake and insulin signaling in hepatoma cells and in rat liver. Am J Pathol 2008;172(3):738-47. doi: 10.2353/ajpath.2008.070097.

207. Le Jan S, Le Meur N, Cazes A, Philippe J, Le Cunff M, Leger J, Corvol P, Germain S. Characterization of the expression of the hypoxia-induced genes neuritin, TXNIP and IGFBP3 in cancer. FEBS Lett 2006;580(14):3395-400. doi: 10.1016/j.febslet.2006.05.011.

208. Bertrand L, Ginion A, Beauloye C, Hebert AD, Guigas B, Hue L, Vanoverschelde JL. AMPK activation restores the stimulation of glucose uptake in an in vitro model of insulin-resistant cardiomyocytes via the activation of protein kinase B. Am J Physiol Heart Circ Physiol 2006;291(1):H239-50. doi: 10.1152/ajpheart.01269.2005.

209. Terazono K, Yamamoto H, Takasawa S, Shiga K, Yonemura Y, Tochino Y, Okamoto H. A novel gene activated in regenerating islets. J Biol Chem 1988;263(5):2111-4.

210. Unno M, Nata K, Noguchi N, Narushima Y, Akiyama T, Ikeda T, Nakagawa K, Takasawa S, Okamoto H. Production and characterization of Reg knockout mice: reduced proliferation of pancreatic beta-cells in Reg knockout mice. Diabetes 2002;51 Suppl 3:S478-83.

211. Kubota N, Terauchi Y, Yamauchi T, Kubota T, Moroi M, Matsui J, Eto K, Yamashita T, Kamon J, Satoh H, et al. Disruption of adiponectin causes insulin resistance and neointimal formation. J Biol Chem 2002;277(29):25863-6. doi: 10.1074/jbc.C200251200.

212. Funda DP, Kaas A, Tlaskalova-Hogenova H, Buschard K. Gluten-free but also gluten-enriched (gluten+) diet prevent diabetes in NOD mice; the gluten enigma in type 1 diabetes. Diabetes Metab Res Rev 2008;24(1):59-63. doi: 10.1002/dmrr.748.

Page 151: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

151

213. Beker Aydemir T, Chang SM, Guthrie GJ, Maki AB, Ryu MS, Karabiyik A, Cousins RJ. Zinc transporter ZIP14 functions in hepatic zinc, iron and glucose homeostasis during the innate immune response (endotoxemia). PLoS One 2012;7(10):e48679. doi: 10.1371/journal.pone.0048679.

214. Tosh D, Shen CN, Alison MR, Sarraf CE, Slack JM. Copper deprivation in rats induces islet hyperplasia and hepatic metaplasia in the pancreas. Biol Cell 2007;99(1):37-44. doi: 10.1042/BC20060050.

215. Zenilman ME, Tuchman D, Zheng Q, Levine J, Delany H. Comparison of reg I and reg III levels during acute pancreatitis in the rat. Ann Surg 2000;232(5):646-52.

216. Al-Abdullah IH, Ayala T, Panigrahi D, Kumar AM, Kumar MS. Neogenesis of pancreatic endocrine cells in copper-deprived rat models. Pancreas 2000;21(1):63-8.

217. Walling C, Partch RE, Weil T. Kinetics of the decomposition of hydrogen peroxide catalyzed by ferric ethylenediaminetetraacetate complex. Proc Natl Acad Sci U S A 1975;72(1):140-2.

218. Evans RW, Rafique R, Zarea A, Rapisarda C, Cammack R, Evans PJ, Porter JB, Hider RC. Nature of non-transferrin-bound iron: studies on iron citrate complexes and thalassemic sera. J Biol Inorg Chem 2008;13(1):57-74. doi: 10.1007/s00775-007-0297-8.

219. Grootveld M, Bell JD, Halliwell B, Aruoma OI, Bomford A, Sadler PJ. 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 1989;264(8):4417-22.

220. Le Lan C, Loreal O, Cohen T, Ropert M, Glickstein H, Laine F, Pouchard M, Deugnier Y, Le Treut A, Breuer W, et al. Redox active plasma iron in C282Y/C282Y hemochromatosis. Blood 2005;105(11):4527-31. doi: 10.1182/blood-2004-09-3468.

221. de la Tour D, Halvorsen T, Demeterco C, Tyrberg B, Itkin-Ansari P, Loy M, Yoo SJ, Hao E, Bossie S, Levine F. Beta-cell differentiation from a human pancreatic cell line in vitro and in vivo. Mol Endocrinol 2001;15(3):476-83. doi: 10.1210/mend.15.3.0604.

222. Lichten LA, Liuzzi JP, Cousins RJ. 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 2009;296(4):G860-7. doi: 10.1152/ajpgi.90676.2008.

Page 152: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

152

223. Simpson RJ, Deenmamode J, McKie AT, Raja KB, Salisbury JR, Iancu TC, Peters TJ. Time-course of iron overload and biochemical, histopathological and ultrastructural evidence of pancreatic damage in hypotransferrinaemic mice. Clin Sci (Lond) 1997;93(5):453-62.

224. Pelot D, Zhou XJ, Carpenter P, Vaziri ND. Effects of experimental hemosiderosis on pancreatic tissue iron content and structure. Dig Dis Sci 1998;43(11):2411-4.

225. Parnaud G, Bosco D, Berney T, Pattou F, Kerr-Conte J, Donath MY, Bruun C, Mandrup-Poulsen T, Billestrup N, Halban PA. Proliferation of sorted human and rat beta cells. Diabetologia 2008;51(1):91-100. doi: 10.1007/s00125-007-0855-1.

226. Maedler K, Sergeev P, Ris F, Oberholzer J, Joller-Jemelka HI, Spinas GA, Kaiser N, Halban PA, Donath MY. Glucose-induced beta cell production of IL-1beta contributes to glucotoxicity in human pancreatic islets. J Clin Invest 2002;110(6):851-60. doi: 10.1172/JCI15318.

227. Choi D, Schroer SA, Lu SY, Wang L, Wu X, Liu Y, Zhang Y, Gaisano HY, Wagner KU, Wu H, et al. Erythropoietin protects against diabetes through direct effects on pancreatic beta cells. J Exp Med 2010;207(13):2831-42. doi: 10.1084/jem.20100665.

228. Kochanowski BA, Sherman AR. Cellular growth in iron-deficient rats: effect of pre- and postweaning iron repletion. J Nutr 1985;115(2):279-87.

229. Cusack RP, Brown WD. Iron Deficiency in Rats: Changes in Body and Organ Weights, Plasma Proteins, Hemoglobins, Myoglobins, and Catalase. J Nutr 1965;86:383-93.

230. Pedersen CR, Bock T, Hansen SV, Hansen MW, Buschard K. High juvenile body weight and low insulin levels as markers preceding early diabetes in the BB rat. Autoimmunity 1994;17(4):261-9.

231. Wilson M, Hughes SJ. Impaired glucose tolerance and mild hyperglycemia in sucrose-fed rats does not impair insulin secretion. Acta Diabetol 1996;33(3):211-5.

232. Yu F, Hao S, Yang B, Zhao Y, Zhang R, Zhang W, Yang J, Chen J. Insulin resistance due to dietary iron overload disrupts inner hair cell ribbon synapse plasticity in male mice. Neurosci Lett 2015;597:183-8. doi: 10.1016/j.neulet.2015.04.049.

233. Dongiovanni P, Ruscica M, Rametta R, Recalcati S, Steffani L, Gatti S, Girelli D, Cairo G, Magni P, Fargion S, et al. Dietary iron overload induces visceral adipose tissue insulin resistance. Am J Pathol 2013;182(6):2254-63. doi: 10.1016/j.ajpath.2013.02.019.

Page 153: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

153

234. Farrell PA, Beard JL, Druckenmiller M. Increased insulin sensitivity in iron-deficient rats. J Nutr 1988;118(9):1104-9.

235. Pi J, Bai Y, Zhang Q, Wong V, Floering LM, Daniel K, Reece JM, Deeney JT, Andersen ME, Corkey BE, et al. Reactive oxygen species as a signal in glucose-stimulated insulin secretion. Diabetes 2007;56(7):1783-91. doi: 10.2337/db06-1601.

236. Ize-Ludlow D, Lightfoot YL, Parker M, Xue S, Wasserfall C, Haller MJ, Schatz D, Becker DJ, Atkinson MA, Mathews CE. Progressive erosion of beta-cell function precedes the onset of hyperglycemia in the NOD mouse model of type 1 diabetes. Diabetes 2011;60(8):2086-91. doi: 10.2337/db11-0373.

237. Brunzell JD, Robertson RP, Lerner RL, Hazzard WR, Ensinck JW, Bierman EL, Porte D, Jr. Relationships between fasting plasma glucose levels and insulin secretion during intravenous glucose tolerance tests. J Clin Endocrinol Metab 1976;42(2):222-9. doi: 10.1210/jcem-42-2-222.

238. Gibson JN, Jellen LC, Unger EL, Morahan G, Mehta M, Earley CJ, Allen RP, Lu L, Jones BC. Genetic analysis of iron-deficiency effects on the mouse spleen. Mamm Genome 2011;22(9-10):556-62. doi: 10.1007/s00335-011-9344-4.

239. Mueller DB, Koczwara K, Mueller AS, Pallauf J, Ziegler AG, Bonifacio E. Influence of early nutritional components on the development of murine autoimmune diabetes. Ann Nutr Metab 2009;54(3):208-17. doi: 10.1159/000220416.

240. Coleman DL, Kuzava JE, Leiter EH. Effect of diet on incidence of diabetes in nonobese diabetic mice. Diabetes 1990;39(4):432-6.

241. Marietta EV, Gomez AM, Yeoman C, Tilahun AY, Clark CR, Luckey DH, Murray JA, White BA, Kudva YC, Rajagopalan G. Low incidence of spontaneous type 1 diabetes in non-obese diabetic mice raised on gluten-free diets is associated with changes in the intestinal microbiome. PLoS One 2013;8(11):e78687. doi: 10.1371/journal.pone.0078687.

242. Brissot P, Ropert M, Le Lan C, Loreal O. Non-transferrin bound iron: A key role in iron overload and iron toxicity. Biochim Biophys Acta 2012;1820(3):403-10. doi: 10.1016/j.bbagen.2011.07.014.

243. Gatter KC, Brown G, Trowbridge IS, Woolston RE, Mason DY. Transferrin receptors in human tissues: their distribution and possible clinical relevance. J Clin Pathol 1983;36(5):539-45.

Page 154: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

154

244. Vasseur S, Folch-Puy E, Hlouschek V, Garcia S, Fiedler F, Lerch MM, Dagorn JC, Closa D, Iovanna JL. p8 improves pancreatic response to acute pancreatitis by enhancing the expression of the anti-inflammatory protein pancreatitis-associated protein I. J Biol Chem 2004;279(8):7199-207. doi: 10.1074/jbc.M309152200.

245. Reaven GM, Hollenbeck C, Jeng CY, Wu MS, Chen YD. Measurement of plasma glucose, free fatty acid, lactate, and insulin for 24 h in patients with NIDDM. Diabetes 1988;37(8):1020-4.

246. Caillaud C, Mechta M, Ainge H, Madsen AN, Ruell P, Mas E, Bisbal C, Mercier J, Twigg S, Mori TA, et al. Chronic erythropoietin treatment improves diet-induced glucose intolerance in rats. J Endocrinol 2015;225(2):77-88. doi: 10.1530/JOE-15-0010.

Page 155: © 2016 Richard Coffey - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/04/97/92/00001/COFFEY_R.pdfsTFR Soluble transferrin receptor TBI Transferrin bound iron TCL Total cell

155

BIOGRAPHICAL SKETCH

Richard Coffey was born in New York, New York in 1988. He completed his B.S.

in nutritional sciences at the University of Florida in the spring of 2011. After graduation

he joined Dr. Mitchell Knutson’s laboratory at the University of Florida focusing on

mechanisms of iron transport and the influence of iron on aspects of diabetic pathology.

He received his Ph.D. in nutritional sciences from the University of Florida in the spring

of 2016.