189
PREVALENCE, CLINICAL CHARACTERISTICS AND GENETIC MARKERS OF INCIPIENT DIABETIC NEPHROPATHY IN CHILDREN AND ADOLESCENTS WITH TYPE 1 DIABETES IN WESTERN AUSTRALIA Patricia Herold Gallego MD, MSc Paediatrics Fellow of the Royal Australasian College of Physicians 2010 This thesis is submitted for the degree of Doctor of Philosophy at The University of Western Australia School of Paediatics and Child Health

Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

PPRREEVVAALLEENNCCEE,, CCLLIINNIICCAALL CCHHAARRAACCTTEERRIISSTTIICCSS

AANNDD GGEENNEETTIICC MMAARRKKEERRSS OOFF IINNCCIIPPIIEENNTT

DDIIAABBEETTIICC NNEEPPHHRROOPPAATTHHYY IINN CCHHIILLDDRREENN AANNDD

AADDOOLLEESSCCEENNTTSS WWIITTHH TTYYPPEE 11 DDIIAABBEETTEESS IINN

WWEESSTTEERRNN AAUUSSTTRRAALLIIAA

Patricia Herold Gallego

MD, MSc Paediatrics

Fellow of the Royal Australasian College of Physicians

2010

This thesis is submitted for the degree of Doctor of Philosophy at The

University of Western Australia

School of Paediatics and Child Health

Page 2: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

ii

DDEEDDIICCAATTIIOONN

This thesis is dedicated to my parents,

MANOEL GUILHERME MEUL GALLEGO

AND

DULCI HEROLD GALLEGO

For their endless love, support and encouragement

Page 3: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

iii

EETTHHIICCSS CCLLEEAARRAANNCCEE

All studies in this thesis were approved by the Ethics Committee at Princess

Margaret Hospital for Children, Subiaco, Western Australia. All subjects, and

their parents, gave written, informed consent for their results to be analysed.

Page 4: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

iv

SSTTAATTEEMMEENNTT OOFF CCOONNTTRRIIBBUUTTIIOONN

The work presented in this thesis was performed primarily by Dr Patricia H

Gallego (the candidate) at the Department of Endocrinology and Diabetes at

Princess Margaret Hospital for Children, The University of Western Australia,

Perth, Australia. The candidate planned the design of all studies with Dr Tim

Jones (the candidate’s supervisor). The candidate undertook the planning,

the elaboration of ethics protocol, the structuring of the data, the data

collection and checking, the statistical analysis, the literature review, the

interpretation of results, discussions and conclusions included in this thesis

which comprises about 80-90% of the work. In the first paper (chapter 3), Dr

Fiona Frazer, Dr Antony Lafferty and Dr Elizabeth Davis were co-authors

and provided feedback for drafts of manuscripts. Dr Max Bulsara provided

statistical advice for all manuscripts. In the second paper (chapter 4), Andrea

Gilbey and Mary Grat collaborated in the recruitment of participants. Due to

the nature of the research of chaper 5, the manuscript had collaborators

from the research team at Princess Margaret Hospital, Western Australia

Instiitute for Medical Research and University of Western Australia, including

Neil Shepard, Frank von Bockxmeer, Brenda Powell, John Beilby, Gillian

Arscott, Michael Le Page, Lyle Palmer, Elizabeth Davis and Catherine

Choong, who are liested as co-authors. Permission was sought and granted

by all collaborators to include the publications in the thesis.

Dr Patricia H Gallego

(Candidate)

Dr Tim Jones

(Supervisor)

Page 5: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

v

AABBSSTTRRAACCTT

Diabetic nephropathy (DN) is an important cause of morbidity and mortality

among subjects with type 1 diabetes mellitus (T1DM). Early elevation of

albumin excretion rate (AER) and the presence of microalbuminuria (MA)

are early signs of DN and predictors for the progression of diabetic

nephropathy and the development of cardiovascular disease.

The overall aim of this work was to define potential clinical and genetic

markers for the appearance of persistent MA among children and

adolescents with T1DM. The main hypothesis was that both environmental

and genetic factors are contributors to the appearance of DN and that these

factors can be identified at very early stages of T1DM in childhood.

MA in children and adolescents with T1DM was examined in four different

settings:

1) In the first study (Chapter 3), the natural history of incipient nephropathy

was assessed in a population-based sample of 955 children and

adolescents aged less than 16 years at diabetes onset who were followed

longitudinally until the development of MA. The prevalence and the clinical

factors that predicted the development of MA were studied. MA was

prevalent in 13.4%. The major clinical contributors were glycaemic control as

reflected by mean HbA1c; age at diagnosis of T1DM and the presence of

puberty. HbA1c increased the risk for MA by 21% after adjusting for other

covariates. Age at diagnosis increased the risk by 25% whereas puberty

increased by 8-fold the risk for persistent MA. Furthermore, the contribution

of diabetes duration for the appearance of persistent MA was not uniform

over time with subjects diagnosed under the age of 5 years presenting a

Page 6: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

vi

prolonged survival time without MA in comparison with those diagnosed

between 5-11 and > 11 years.

2) In the second study (Chapter 4), the relationship between AER and 24-

hour blood pressure (BP) measurements was determined in a sub-group of

75 T1DM children with normal AER. By studying 24-hour BP, early changes

in both systolic and diastolic BP occurred concomitantly with a rise in AER

even prior to the development of MA.

3) The third study (Chapter 5) examined the relationship between changes

of AER over a decade in a cohort of 620 children and adolescents. A decline

in the prevalence of MA was noticed over time in parallel to improvements in

diabetes control.

4) The study number four (Chapter 6) investigated the genetic risk of

polymorphisms at the renin-angiotensin system (RAS) in relation to the

appearance of persistent MA. In this study, the angiotensinogen (AGT) and

angiotensin 1-converting enzyme (ACE) genes conferred a higher risk for

persistent MA. Young subjects homozygous for the T allele at the AGT

T235M gene presented a 4-fold increased risk for MA and the survival time

without persistent MA was shorter for those subjects homozygous for the

deletion (D) allele at the ACE insertion/deletion (ACE I/D) gene.

The results of this work highlight the effects of both environmental and

genetic variables identifiable at early stages of T1DM as main contributors to

the development of persistent MA. The contribution of pre and post pubertal

duration of diabetes to the development of persistent MA was non-uniform.

By identifying these predictors for incipient DN, prevention strategies can be

targeted to high-risk adolescents with T1DM earlier in the course of the

disease.

Page 7: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

vii

AACCKKNNOOWWLLEEDDGGEEMMEENNTTSS

I am extremely grateful to my supervisors Dr Timothy W Jones who has

always supported me and has provided me with encouragement and

strength for this work, and Dr Catherine C Choong who, as well, has guided

me and provided me with support over the last few years.

I have very much appreciated the endless assistance and support from my

colleagues from the department of Endocrinology at the Mater Children’s

Hospital, Dr Andrew Cotterill, Dr Mark Harris and Dr Gary Leong.

I would also like to thank Max Bulsara who has given me not only his

friendship but also the most valuable statistical advice through all this work.

Max has tirelessly helped me with data management and statistical analysis.

I have very much appreciated the assistance, support and friendship I have

had from the co-authors of the published manuscripts Fiona Frazer, Anthony

Lafferty, Elisabeth Davis, Andrea Gilbey, Mary Grant, Geoff Byrne, Neil

Shephard, Frank van Bockxmeer, Brenda Powell, John Beilby and Lyle

Palmer.

I would also like to thank the research assistants that helped performing the

DNA extraction and genetic polymorphisms, Michael Le Page and Gillian

Arscott.

I am also particularly grateful to all past and present staff of the Department

of Endocrinology and Diabetes at Princess Margaret Hospital for Children;

Jacqueline Curran, Sarah McMahon, Aveni Haynes, Niru Paramalingam,

Aris Siafarikas, Vanessa Baker, Alisha Thompson, Leanne Youngs, Glynis

Page 8: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

viii

Price, Stefan Riedl for their friendship and support. These people have

taught me new skills and have generously given me their advice and

friendship when I needed the most, which I have greatly valued.

I am extremely grateful to the children and families who have participated in

the studies described in this thesis, without which none of this research

could have been conducted. To them, I extend my heartfelt thanks.

Finally, I wish to thank to my parents, brother, sister-in-law and lovely

nephews who living geographically so far away have supported me at each

step of this remarkable experience. They gave me the light and the strength

I needed to overcome all the obstacles and to arrive at the end of this

journey.

Page 9: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

ix

CCOONNTTEENNTTSS

DEDICATION --------------------------------------------------------------------------------ii

ETHICS CLEARANCE--------------------------------------------------------------------iii

STATEMENT OF CONTRIBUTION---------------------------------------------------iv

ABSTRACT-----------------------------------------------------------------------------------v

ACKNOWLEDGEMENTS---------------------------------------------------------------vii

CONTENTS----------------------------------------------------------------------------------ix

LIST OF TABLES------------------------------------------------------------------------xvi

LIST OF FIGURES----------------------------------------------------------------------xvii

ABBREVIATIONS------------------------------------------------------------------------xix

DEFINITIONS------------------------------------------------------------------------------xxi

CHAPTER 1: INTRODUCTION---------------------------------------------------------2

1.1 Outline of thesis--------------------------------------------------------------- 3

1.2 Research aims-----------------------------------------------------------------4

1.3 Hypotheses---------------------------------------------------------------------4

1.4 Publications arising from this thesis--------------------------------------5

CHAPTER 2: BACKGROUND----------------------------------------------------------7

TYPE 1 DIABETES MELLITUS---------------------------------------------------------7

2.1 Definition and Classification of Diabetes in Childhood------------7

2.2 Global and Australian Incidence of Type 1 Diabetes----------------7

COMPLICATIONS OF TYPE 1 DIABETES----------------------------------------10

2.3 Overview of Complications in T1DM------------------------------------10

2.4 Long-term complications in T1DM---------------------------------------10

Page 10: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

x

DIABETIC NEPHROPATHY-----------------------------------------------------------15

2.5 Defining Diabetic Nephropathy (DN)------------------------------------15

2.5.1 Microalbuminuia (MA)-------------------------------------------15

2.5.2 Borderline Albuminuria------------------------------------------16

2.5.3 Macroalbuminuria------------------------------------------------16

2.6 Classification or Stages of DN--------------------------------------------16

2.6.1 Stage I - Early Hypertrophy-hyperfunction-----------------16

2.6.2 Stage II - Glomerular Lesions without Clinical Disease----

-------------------------------------------------------------------------------17

2.6.3 Stage III - Incipient DN-----------------------------------------17

2.6.4 Stage IV - Overt DN---------------------------------------------18

2.6.5 Stage V - End Stage Renal Disease (ESDR)-------------18

2.7 Pathogenesis of Diabetic Nephropathy--------------------------------19

2.7.1 HYPERGLYCAEMIA AND DN-------------------------------19

2.7.1.1Formation of Advanced Glycation End-products

(AGEs): The Glycation Hypothesis-------------------------20

2.7.1.2 Activation of Aldose Reductase (Polyol)

Pathway-----------------------------------------------------------21

2.7.1.3 Activation of Protein Kinase C (PKC) Theory-----

----------------------------------------------------------------------22

2.7.1.4 Other mechanisms-----------------------------------24

a) Oxidative stress-induced vascular pathology--------24

b) Changes in expression and action of growth factors--

----------------------------------------------------------------------24

2.7.2 HEAMODYNAMIC CHANGES AND DN------------------24

2.7.2.1 The Role of Systemic Hypertension-------------24

2.7.2.2 The Role of Glomerular Capillary Hypertension

and Glomerular Hyperfiltration in DN----------------------26

Page 11: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xi

2.7.2.3 Treatment of Glomerular Hyperfiltration--------28

2.8 Genetics of Diabetic Nephropathy---------------------------------------29

2.8.1 Genes of the Renin-Angiotensin System (RAS)---------30

2.8.1.1 The angiotensin I-converting enzyme (ACE)

gene and DN-----------------------------------------------------31

2.8.1.2 The angiotensinogen (AGT) gene and DN-----32

2.8.1.3 The angiotensin II receptor type 1 (AGTR1)

gene and DN-----------------------------------------------------33

2.8.2 Other candidate genes------------------------------------------34

2.9 DN in Children with T1DM------------------------------------------------37

2.9.1 Risk factors for DN and microvascular complications in children

with T1DM -------------------------------------------------------------------------37

Glycaemic control-------------------------------------------------------37

Duration of diabetes and the effect of puberty-------------------42

Blood pressure ----------------------------------------------------------44

Lipids------------------------------------------------------------------------46

Smoking--------------------------------------------------------------------46

Family history of vascular complications---------------------------46

Obesity---------------------------------------------------------------------46

2.10 Screening guidelines for diabetes complications in T1DM

children------------------------------------------------------------------------------47

CHAPTER 3: STUDY I: “Prevalence and risk factors for

microalbuminuria in a population-based sample of children and

adolescents with T1DM in Western Australia”---------------------------------49

3.1 PREFACE------------------------------------------------------------------------------49

3.2 Abstract---------------------------------------------------------------------------------50

3.3 Introduction-----------------------------------------------------------------------------51

Page 12: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xii

3.4 Methods---------------------------------------------------------------------------------52

3.4.1 Definition of micro albuminuria-----------------------------------------52

3.4.2 Laboratory evaluation----------------------------------------------------53

3.4.3 Statistical analysis--------------------------------------------------------54

3.5 Results----------------------------------------------------------------------------------55

3.5.1 Clinical characteristics---------------------------------------------------55

3.5.2 Microalbuminuria, metabolic control, age at diagnosis, gender,

duration of diabetes and puberty---------------------------------------------55

3.5.3 Contribution of the different risk factors for developing MA----60

3.6 Discussion------------------------------------------------------------------------------61

3.7 Acknowledgements------------------------------------------------------------------64

CHAPTER 4: STUDY II: “Early changes in 24-hour ambulatory blood

pressure are associated with high normal albumin excretion rate in

children with type 1 diabetes”-------------------------------------------------------66

4.1 PREFACE------------------------------------------------------------------------------66

4.2 Abstract---------------------------------------------------------------------------------67

4.3 Introduction-----------------------------------------------------------------------------68

4.4 Research design and methods----------------------------------------------------69

4.4.1 Urinary AER assay--------------------------------------------------------69

4.4.2 Mean HbA1c levels-------------------------------------------------------69

4.4.3 Assessment of BP--------------------------------------------------------70

4.4.4 Statistical analysis--------------------------------------------------------70

4.5 Results----------------------------------------------------------------------------------72

4.5.1 Characteristics of the patients at the time of enrolment---------72

4.5.2 Subgroup analysis according to AER--------------------------------72

Page 13: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xiii

4.5.3 Subgroup analysis according to glycosilated hemoglobin

level----------------------------------------------------------------------------------76

4.5.4 Subgroup analysis on dippers and non-dippers-------------------76

4.6 Discussion------------------------------------------------------------------------------77

CHAPTER 5: STUDY III: “Declining Trends of albumin excretion rate in

Children and Adolescents with Type 1 Diabetes Mellitus”----------------80

5.1 PREFACE------------------------------------------------------------------------------80

5.2 Abstract---------------------------------------------------------------------------------81

5.3 Introduction-----------------------------------------------------------------------------82

5.4 Patient and methods-----------------------------------------------------------------83

5.4.1 Study design and patients----------------------------------------------83

5.4.2 Laboratory tests-----------------------------------------------------------84

AER-------------------------------------------------------------------------84

HbA1c----------------------------------------------------------------------85

Total cholesterol---------------------------------------------------------85

Anthropometric data and blood pressure--------------------------85

5.4.3 Statistical analysis--------------------------------------------------------86

5.5 Results----------------------------------------------------------------------------------87

5.5.1 Patients’ characteristics-------------------------------------------------87

5.5.2 Albumin excretion rate and MA----------------------------------------89

5.5.3 Metabolic control, insulin management and hypoglycaemic

events--------------------------------------------------------------------------------90

5.5.4 Blood pressure, cholesterol levels and anthropometric data---91

5.5.5 Univariate and multiple logistic regression-------------------------91

5.6 Discussion------------------------------------------------------------------------------94

Page 14: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xiv

CHAPTER 6: STUDY IV: “Angiotensinogen gene T235 variant: a marker

for the development of microalbuminuria in children and adolescents

with type 1 diabetes mellitus”-------------------------------------------------------99

6.1 PREFACE------------------------------------------------------------------------------99

6.2 Abstract--------------------------------------------------------------------------------100

6.3 Introduction---------------------------------------------------------------------------101

6.4 Patients and methods--------------------------------------------------------------102

6.4.1 Design and procedures------------------------------------------------103

6.4.2 Laboratory tests----------------------------------------------------------103

6.4.3 Genotyping----------------------------------------------------------------104

6.4.4 Statistical analysis-------------------------------------------------------105

6.5 Results---------------------------------------------------------------------------------107

6.6 Discussion----------------------------------------------------------------------------112

6.7 Acknowledgment--------------------------------------------------------------------114

CHAPTER 7: GENERAL DISCUSSION-------------------------------------------116

7.1 Summary and Conclusions-------------------------------------------------------116

7.2 Limitations----------------------------------------------------------------------------119

7.3 Implications and Future Directions---------------------------------------------122

7.3.1 Intervention Directed to T1DM Children and Adolescents---123

7.3.1.1 Glycaemic control--------------------------------------------123

7.3.1.2 The use of antihypertensive treatment-----------------124

ACE Inhibitors (ACEI)----------------------------------------125

Angiotensin II receptor blocker (ARB)-------------------125

7.3.1.3 The use of lipid agents--------------------------------------126

7.3.2 Genetic Screening for DN---------------------------------------------126

Page 15: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xv

REFERENCES---------------------------------------------------------------------------128

Appendix A Information Sheets and Consent Forms-------------157

Appendix B Classification and characteristic features of T1DM

in children; screening guidelines for vascular

complications in the pediatric and adolescent T1DM

group------------------------------------------------------------163

Appendix C PDF published manuscripts related to this thesis----

---------------------------------------------------------------------166

Appendix D Other Diabetes Related PDF Publications----------190

Page 16: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xvi

LLIISSTT OOFF TTAABBLLEESS

Table 2.1 Candidate genes in which polymorphisms have

demonstrated association with T1DM diabetic nephropathy--

-----------------------------------------------------------------------------36

Table 2.2 Target indicators of glycaemic control in T1DM children and

adolescents-------------------------------------------------------------41

Table 3.1 Gender, HbA1c, incidence density and postpubertal

incidence density of MA for subjects aged < 5, 5-11 and > 11

years at diabetes onset----------------------------------------------56

Table 3.2 Proportional contribution of gender, puberty, age at diabetes

onset and HbA1c as risk factors for the development of MA

with duration of diabetes as the time variable (n= 949)------60

Table 4.1 Characteristics of children and adolescents with type 1

diabetes with low and high normal AER-------------------------73

Table 5.1 Characteristics of adolescents with T1DM in the three

study periods----------------------------------------------------------88

Table 5.2 Univariate logistic regression analysis for high AER---------91

Table 5.3 Univariate logistic regression analysis for microalbuminuria--

-----------------------------------------------------------------------------92

Table 5.4 Multiple logistic regression analysis for high AER------------92

Table 5.5 Multiple logistic regression analysis for micro albuminuria-----

-----------------------------------------------------------------------------93

Table 6.1 Clinical characteristics of participants----------------------------107

Table 6.2 ACE, AGT and AGTR1 genotypes of participants------------108

Table 6.3 Cox regression multivariate analysis on the risk of persistent

MA in T1DM children---------------------------------------------------111

Page 17: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xvii

LLIISSTT OOFF FFIIGGUURREESS

Figure 2.1 New cases of Type 1 Diabetes Mellitus in children 0-14 years

(per 100 000 population) in 2010--------------------------------------8

Figure 2.2 Incidence rate of T1DM per 100 000 population aged 0-14

years old in OECD countries. Data survey from late 1990s to

early 2000s------------------------------------------------------------------9

Figure 2.3 Decline in the cumulative proportion of severe (laser-treated)

retinopathy in a population of patients with T1DM

diagnosed before the age of 15 years according to the year of

onset of diabetes---------------------------------------------------------12

Figure 2.4 Three main pathways implicated in the link between

hyperglycaemia and diabetic microvascular complications---19

Figure 2.5 Activation of aldose reductase or polyol in diabetes------------22

Figure 2.6 Activation of diacylglycerol (DAG)-PKC pathway in the

pathogenesis of diabetic microvascular complications---------23

Figure 2.7 Mechanisms of extracellular matrix deposition in mesangial

cells caused by mechanical stretch in DN-------------------------28

Figure 2.8 The renin angiotensisn system (RAS) and the role of ACE---31

Figure 2.9 Potential genetic and environmental risk factors for the

development of long-term diabetes complications--------------37

Figure 2.10 Distribution of HbA1c concentration by randomized treatment

group at the end of the DCCT and in each year of EDIC study-

--------------------------------------------------------------------------------39

Page 18: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xviii

Figure 2.11 Prevalence and cumulative incidence of MA---------------------39

Figure 2.12 Cumulative percentage of borderline AER by age in T1DM

with diabetes duration of 6 years------------------------------------42

Figure 3.1 Cumulative probability (Kaplan-Meier survival curves, log-rank

test, P=0.03) for the development of microalbuminuria for boys

and girls during puberty (○ females, □ males)--------------------58

Figure 3.2 Cumulative probability (Kaplan-Meier survival curves, log-rank

test, P<0.001) for the development of microalbuminuria for

each year since diabetes onset in three groups divided by

age at diagnosis (+ < 5 years, □ 5 to 11 years and ○> 11

years)-----------------------------------------------------------------------59

Figure 4.1 Effect of increments of AER on systolic blood pressure

during daytime and nighttime-----------------------------------------75

Figure 4.2 Effect of increments of AER on diastolic

blood pressure during daytime and nighttime--------------------75

Figure 5.1 Percentage of subjects with high AER (1a) and

microalbuminuria (1b) over the three study periods------------89

Figure 5.2 Median AER by age, comparing P1+ P2 (1993-2000) versus

P3 (2001-2004)-----------------------------------------------------------90

Figure 6.1 Kaplan-Meier estimation for the development of persistent MA

according to ACE genotype in the recessive model (DD versus

ID/II)-----------------------------------------------------------------------109

Figure 6.2 Kaplan-Meier estimation for the development of persistent MA

according to AGT genotype in the recessive model (TT versus

MT/MM)-------------------------------------------------------------------110

Page 19: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xix

AABBBBRREEVVIIAATTIIOONNSS

ABP Ambulatory Blood Pressure

ACE Angiotensin 1-converting enzyme

ACEI Angiotensin 1-converting enzyme inhibitor

ACR Albumin Creatinine Ratio

AER Albumin excretion rate

AGE Advanced Glycation End Product

AGT Angiotensinogen

AGTR1 Angiotensin II receptor type 1

ARB Angiotensin II receptor blocker

BP Blood Pressure

CI 95% Confidence Intervals

CVD Cardiovascular Disease

DAN Diabetic Autonomic Neuropathy

DBP Diastolic Blood Pressure

DCCT Diabetes Control and Complications Trial

DKA Diabetes Ketoacidosis

DN Diabetic Nephropathy

DPN Diabetic Polineuropathy

DR Diabetic Retinopathy

ESRD End Stage Renal Disease

EDIC Epidemiology of Diabetes Interventions and

Complications

GFR Glomerular Filtration Rate

HbA1c Glycated Haemoglobin

Page 20: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xx

HR Hazard Ratio

I/D Insertion/Deletion

IQR Interquartile range

MA Microalbuminuria

ORPS Oxford Regional Prospective Study

PKC Protein Kinase C

RAS Renin-Angiotensin System

SBP Systolic Blood Pressure

T1DM Type 1 Diabetes Mellitus

T2DM Type 2 Diabetes Mellitus

UKPDS United Kingdom Prospective Diabetes Study

Page 21: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

xxi

DDEEFFIINNIITTIIOONNSS

Microalbuminuria

Presence of albumin excretion rate greater or equal to

20µg/min but lower than 200 µg/min in two out of three

overnight timed urine collections unless otherwise

stated.

High normal AER or Borderline AER

Mean albumin excretion rate greater than 7 and lower

than 20 µg/min collected from three overnight timed

urine collections.

Low normal AER

Mean albumin excretion rate lower or equal to 7 µg/min

collected from three overnight timed urine collections.

Page 22: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

CCHHAAPPTTEERR OONNEE

Page 23: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

2

CCHHAAPPTTEERR 11.. IINNTTRROODDUUCCTTIIOONN

This work contains a series of four related manuscripts dealing with the

development of MA or incipient DN (DN) in children and adolescents with

T1DM.

MA is an early sign and predictor of DN among people with T1DM. Although

improvements in diabetes management have occurred over the last

decades, DN is still the leading cause of end-stage renal disease (ESRD) in

US and Europe (1) and the second most common cause in Australia (2). DN

typically develops after 15 to 20 years of diabetes duration but the presence

of persistent MA, an early sign of DN, is a predictor of cardiovascular

disease and early mortality (3-7).

MA may be detected in adolescents who have T1DM but is rarely present in

pre-pubertal children who have diabetes. During adolescence, between 5%

and 25% of patients with T1DM have MA (8-13). Even though persistent MA

is predictive for overt nephropathy in adults with diabetes, its prognostic

value among children and adolescents with diabetes are less well defined.

The main objective of this study is to identify potential predictors for the

development of MA in children and adolescents with T1DM aiming to

minimize the development of diabetes microvascular complications,

particularly nephropathy, by early detection and possible intervention among

those considered at high risk.

Unfortunately, primary prevention and cure for T1DM is still not available.

Improvements in diabetes management with intensive insulin therapy for

optimizing glycaemic control is still the best available treatment for T1DM

and the most effective way of preventing long term vascular complications.

Page 24: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

3

However, several studies demonstrate that despite dramatic improvements

in diabetes care and glycaemic control, some still develop microvascular

disease.

Hence, this study provides a unique opportunity not only to explore the

natural history of persistent MA in a large population-based sample of

children with T1DM followed prospectively from diagnosis but also to identify

clinical and genetic markers for the development of this complication. The

results of this study will allow the identification of those at risk and the

development of prevention strategies for DN.

1.1 Outline of thesis

The following chapter outlines the background of T1DM starting with an

overview of the global incidence and in Australia then moving to an overview

of complications associated with T1DM. This is followed by literature review

on DN, the focus of this research.

The next four chapters following background are structured as papers.

Three of these chapters were published in peer-reviewd journals.

Chapter 3 describes the prevalence and the natural history of MA in a

population-based sample of children and adolescents with T1DM. This

paper highlights potential clinical predictors for the development of MA in a

large cohort of children followed prospectively from diagnosis.

In Chapter 4, the effect of blood pressure on AER is examined more closely

in a sub-group of children and adolescents randomly selected from the

Children’s Diabetic clinic at Princess Margaret Hospital for Children.

In Chapter 5 (not published), the changes in the prevalence of AER and MA

between the years 1993 and 2004 were examined in a large cohort of young

people with T1DM. A decline occurred at times of improvement in diabetes

management and particularly in glycaemic control. In Chapter 6, the

relationship between genetic variants at the renin-angiotensin system,

Page 25: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

4

mainly the angiotensin-converting enzyme, angiotensinogen and angiotensin

II receptor type 1 genes, were studied in a large cohort as candidate genes

implicated in the development of incipient DN.

1.2 Research aims

1) To examine the natural history and to identify clinical predictors for

the development of persistent MA in childhood onset diabetes;

2) To assess the relative risk of diabetes duration in relation to puberty

for the development of MA in children and adolescents with T1DM;

3) To examine the effect of blood pressure in relation to AER in a sub-

group of T1DM children and adolescents

4) To examine the changes of AER and persistent MA over time

5) To identify potential genetic markers that contribute to the

development of early DN early in the course of diabetes;

1.3 Hypotheses

1) That clinical and genetic risk factors for the development of incipient

nephropathy are recognizable in children and adolescents with T1DM

at early stages of the disease;

2) That the duration of diabetes before and after the onset of puberty

have a non-uniform effect towards the development of MA in children

diagnosed with T1DM;

3) That changes in BP precede the appearance of persistent MA;

Page 26: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

5

4) That changes in the prevalence of MA over time will parallel the

modifications in diabetes management and improvements in

glycaemic control;

5) That specific polymorphisms at genes from the renin-angiotensin

system are contributors to the development of incipient nephropathy

in children and adolescents with T1DM

1.4 Publications arising from this thesis

1) Prevalence and risk factors for MA in a population-based sample of

children and adolescents with T1DM in Western Australia (chapter 3).

Gallego PH, Bulsara MK, Frazer F, Lafferty AR, Davis EA, Jones TW.

Pediatric Diabetes 2006:7:165-172.

2) Early changes in 24-hour ambulatory blood pressure (ABP) are

associated with high normal AER in children with T1DM (chapter 4).

Gallego PH, Gilbey AJ, Grant MT, Bulsara MK, Byrne GC, Jones TW,

Frazer FL. Journal of Pediatric Endocrinology and Metabolism 2005:

3) Angiotensinogen gene T235 variant: a marker for the development of MA

in children and adolescents with T1DM mellitus (chapter 6). Gallego PH,

Shepard N, Bulsara MK, van Bockxmeer FM, Powell BL,Beilby JP, Arscott

G, Le Page M, Palmer LJ, Davis EA, Jones TW, Choong CSY. Jurnal of

Diabetes Complications 2007:

Page 27: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

6

CCHHAAPPTTEERR TTWWOO

Page 28: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

7

CCHHAAPPTTEERR 22.. BBAACCKKGGRROOUUNNDD

TYPE 1 DIABETES MELLITUS

2.1 Definition and Classification of Diabetes in Childhood

Diabetes Mellitus has a long history and it was first described in Egyptian

documents from 1552 BC. In the 1st century, Arateus described diabetes as

“the melting down of flesh and limbs into urine” but it was not until the 1940s

that the link between diabetes and long-term complications was stablished

(14).

Type 1 diabetes (T1DM) results from auto-immunity T–cell mediated

destruction of the insulin producing pancreatic beta cells causing insulin

deficiency and leading to chronic hyperglycaemia (15). T1DM is the most

prevalent form of diabetes among young people under the age of 10 years

and the most common chronic disease in childhood, affecting up to 1% of

the general population during their lifespan (16).

Although type 1 and type 2 diabetes can occur in childhood, they present

different disease patterns and require different management. In T1DM

children require daily insulin, which is a life-saving treatment while in T2DM,

depending on clinical parameters, children may not necessarily require

insulin and modifications in lifestyle as well as the use of oral hypoglycaemic

agents may be sufficient for clinical management.

The aetiological classification of disorders of glycaemia in childhood and

adolescence and the characteristic features of type 1 diabetes compared to

type 2 in childhood are shown in Appendix A.

2.2 Global and Australian Incidence of Type 1 Diabetes

T1DM is not restricted by geographical boundaries, nationality, age or

gender. It is been recognized in almost every country with no region being

exempt from the disease (Figure 2.1).

Page 29: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

8

Figure 2.1 New cases of T1DM in children 0-14 years (per 100 000

population) in 2010

[Source: Diabetes Atlas, International Federation]

The mean annual incidence rates for T1DM in children 0-14 years of age is

summarized in Figure 2.2. Between the years 1990 and 1994, low incidence

rates of 0.1 per 100 000 per annum were observed in countries like China

while higher rates close to 56 per 100 000 per annum were reported in

Finland (17). In 2007, the International Diabetes Federation estimated that

among children aged 0-14 years, some 70 000 children worldwide are

expected to develop T1DM annually. Increasing incidence rates ranging

from 2-5% per annum have been reported, particularly in the under 4-yr-old

age group (18). These changes in incidence rates have been described in

countries with both high and low prevalences but with some indication of a

more rapid increase in low-prevalence countries.

From the late 1990s to early 2000s, the Australian Institute of Health and

Welfare reported the Australia’s incidence rate for T1DM as in the upper end

of range. A survey covering the period 2000-2006 estimated the age-

standardized incidence rate of T1DM to be 22.4 per 100 000 population

aged 0-14 years (19). This incidence places Australia in a similar category to

other countries such as the United Kingdom and Canada (Figure 2.2).

Page 30: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

9

Figure 2.2 Incidence rate of T1DM per 100 000 population aged 0-14 years

old in OECD countries. Data survey from late 1990s to early 2000s.

[Source: Australian Institute of Health and Welfare, 2008]

Studies in the incidence of T1DM in childhood have reported that the trends

in Australia reflect what has been observed in other countries. In Sweden,

time trends analysis in the incidence T1DM from 1983 to 2007 in individuals

aged 0-34yrs revealed an increase by calendar year in both sexes under the

age of 15yrs (20). Similarly, a prospective study of twenty-one years in

Hungary reported that the overall incidence rate has doubled from 1989 to

2009, with the highest rate in the very young children (21).

Overall the annual incidence of type 1 diabetes has increased on average by

3.2% per year from 1990 to 1996 in New South Wales (22).

In Western Australia, the incidence increased on average by 3.1% a year

over the period between 1985 to 2002 (23).

In Victoria, the rate of increase of T1DM was much higher and reported as

close to 9% but it was measured over a shorter period of time in comparison

to the previous studies (24).

Page 31: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

10

COMPLICATIONS OF TYPE 1 DIABETES

2.3 Overview of Complications in T1DM

In T1DM, insulin therapy is essential for maintaining plasma glucose levels

within a normal range. Insulin is delivered subcutaneously by intermittent

injections or by continuous delivery via an insulin pump. Although there have

been advances in methods of insulin delivery as well as an improvement in

the quality of available insulin, injecting exogenous insulin does not perfectly

replicate the normal biological variation in insulin secretion and the

development of short and long-term complications is commomly seen

among those who suffer the disease.

Short-term complications result from either acute severe insulin deficiency

causing diabetes ketoacidosis or are due to relative insulin excess causing

hypoglycaemia. A discussion of acute complications associated with the

management of children with T1DM is beyond the scope of this work.

The long-term complications of diabetes include macrovascular disease

(cerebrovascular disease, ischaemic heart disease and peripheral arterial

disease) and microvascular complications (nephropathy, retinopathy and

neuropathy).

2.4 Long-term complications in T1DM

In adults and adolescents, it has been established that poor glycemic control

is causally associated with long-term vascular sequelae of T1DM. These

complications include nephropathy, retinopathy, neuropathy and

cardiovascular disease.

Although clinically evident diabetes–related vascular complications are rare

in childhood and adolescence, early signs of functional and structural

abnormalities may be detected a few years after the onset of the disease.

For example, increased vascular stiffness is seen in children with type 1

diabetes compared with healthy controls (25).

Page 32: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

11

In regards to macrovascular complications, T1DM is associated with a three

to fourfold increased mortality risk in all age-bands when compared to the

non-diabetic population (26). Cardiovascular disease (CVD) including

myocardial infarction, ischaemic heart disease and strokes accounts for half

of all deaths with similar mortality rates observed in men and women under

the age of 40 years (27). The incidence rates of CVD are substantially

higher in the T1DM population at 1-2% per year when compared with the

general population incidence rates of 0.1-0.5% per year (28, 29).

Despite improvements in diabetes care, the Pittsburgh Epidemiology of

Diabetes Complications (EDC) study, a prospective observational study of

childhood-onset diabetes, demonstrated a decreasing trend for

complications such as mortality, renal failure and neuropathy but no

changes in rates of coronary artery disease. Improvement in glycaemic

control was not associated with a decline in CVD over time. These data

suggest that focus on other risk factors for CVD such as blood pressure and

lipids are important in T1DM (29). Supporting these data, the EURODIAB

Prospective Complications Study showed that the strongest risk factors for

coronary heart disease were baseline albuminuria, blood pressure, fasting

tryglicerides and waist-to-hip circumference (30).

Persistent proteinuria is also a strong predictor for the development of CVD

(4, 29). Atherosclerosis has been documented to start in childhood with

increased thickness of intima-media of carotids and aorta; silent coronary

atherosclerosis has also been demonstrated in young adults with childhood-

onset diabetes (31). Cholesterol is an important factor for the initiation of

atherosclerosis. In diabetes, a more atherogenic profile is observed despite

well-controlled glycaemia (32) but poor glycaemic control is associated with

a more atherogenic profile (33).

Microvascular complications are a major cause of morbidity in young people

with T1DM. Diabetic retinopathy (DR) affects the majority of patients with

T1DM after 15 years duration (34). Although recent data indicates a

reduction in the incidence of severe retinopathy (Figure 2.3) (35), studies

suggest that 24% to 27% of children and adolescents with T1DM present

Page 33: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

12

early background retinopathy after 6 to 13 years of diabetes duration (36,

37).

Onset of diabetes: 1961-1965 (▲), 1966-1977 (◄), 1971-1975 (○), 1976-1980 (◊), 1981-1985 (●).

Figure 2.3 Decline in the cumulative proportion of severe (laser-treated)

retinopathy in a population of patients with T1DM diagnosed before the age

of 15 years according to the year of onset of diabetes

[Source: The Linköping Diabetes Complications Study, Diabetologia, 2004]

Hyperglycaemia plays a major role in the development of DR by impairing

the mechanism of retinal perfusion auto regulation, which protects the eye

from systemic hypertension (38). Two major clinical trials, the DCCT and

the UKPDS, have demonstrated the beneficial effects of intensive glycaemic

control in reducing the onset and progression of DR in type 1 (39-41) and

type 2 diabetes (42).

The Wiscosin Epidemiology Study of Diabetic Retinopathy (WESDR)

reported the 14-year incidence and progression of DR in a large cohort of

“younger-onset” persons with diabetes (diagnosed before 30 years of age).

This group showed a very high 14-yr incidence of any retinopathy (96%) and

a 86% incidence of progression of retinopathy (43). In addition to

hyperglycaemia being the main trigger for DR, higher diastolic blood

pressure and male gender were risk factors for DR in this study.

Page 34: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

13

In T1DM adolescents, long-term glycaemic control, both before and after

puberty, is the major risk factor for retinal changes. But other factors such as

age of onset of diabetes, puberty, lipids, blood pressure and genetic factors

will also have varying relevance at a individual level (44).

Diabetic neuropathy described the involvement of the peripheral and

autonomic nervous systems commonly seen in T1DM. The somatic

neuropathies associated with diabetes fall into two categories: focal

neuropathies and sensorimotor polyneuropathies.

One of the largest studies of diabetic neuropathy, the EURODIAB baseline

diabetic polyneuropathy (DPN) study, was conducted between 1989-1991

and involved 3250 subjects with T1DM from 16 countries (mean age of 33

years; mean duration of diabetes of 15 years). Diabetic neuropathy was

present in 19% of 15-29 yr olds; 28% of 30-44 yr olds and 57% of 45-61 yr

old subjects at baseline. Glycaemic control was the major correlate of

neuropathy, even after adjusting for duration of diabetes. A substantial

prevalence of DPN was also found in those whose control was very good

(A1C < 7%) (45).

The EURODIAB prospective study followed 1172 subjects for 7.3 years.

The cumulative incidence of neuropathy was 23.5% and risk factors included

glycaemic contro, diabetes duration, hypertension, smoking, BMI and

triglycerdides (45). The DCCT also shed light on the effect of glycaemic

control on the rates of neuropathy in young people with T1DM (41). The

EDIC study confirmed lower rates of diabetic neuropathy in the intensive

group with benefits extending for at least 8 years beyond the end of the

DCCT (46).

The prevalence of diabetic neuropathy in the paediatric population is less

clear probably due to the lack of large epidemiological studies in the

paediatric T1DM group. In epidemiological studies involving both adult and

paediatric patients, the prevalence of DPN varies, ranging from 7 to 57%

(47). This large difference is likely due to the lack of appropriate diagnostic

criteria. The most complete evaluation using electrophysiological testing

reported a prevalence of 57% of subclinical neuropathy in children and

adolescents with T1DM (48).

Page 35: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

14

Hereafter the literature review on microvascular complications will refer to

the development of DN detailed in the following pages of this review.

Page 36: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

15

DIABETIC NEPHROPATHY

2.5 Defining Diabetic Nephropathy (DN)

The earliest clinical evidence of DN is the appearance of low but abnormal

levels of albumin in the urine, known as micro albuminuria (MA). Patients

with MA are referred to as having incipient DN. All definitions of MA are

somewhat arbitrary.

2.5.1 Microalbuminuia (MA)

Definitions of MA include an AER of 20 to 200 μg per minute in urine

collected over 24 hours.; an AER of 15 to 150 μg/min in urine collected

overnight; or an AER of 30 to 300 mg/24hr in 24-h urine collections (49).

Classically, and by consensus of many investigators, MA has been defined

as an overnight urinary AER of 20-200 μg/min (50).

According to ISPAD guidelines, the accepted definitions for MA in childhood

onset diabetes are:

1) AER 20-200 μg/min in timed overnight urine collection; or AER 30-300

mg/24h in 24-h urine collections;

2) Albumin concentration of 30-300 mg/l (early morning urine sample);

3) Albumin/creatinine ratio (ACR) 2.5 – 25 mg/mmol or 30-300 mg/g (spot

urine) in males and 3.5 - 25 mg/mmol in females (because of their lower

creatinine excretion)

Irrespective of the definition used, MA needs to be confirmed by the finding

of two out of three abnormal samples collected over a 3-6 months period. In

addition to its being the earliest manifestation of DN, persistent MA is also a

marker of end-stage renal disease (ESRD) and of increased cardiovascular

morbidity and mortality in T1DM (4, 51, 52).

Page 37: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

16

2.5.2 Borderline Albuminuria

Borderline albuminuria is considered an AER between the upper 95th

percentile of normal AER (levels reported between 7.2 to 7.6 μg/min) and

the lowest value predictive of nephropathy (20 μg/min) in at least one of

three consecutive timed collections (53, 54).

In childhood, early elevations of AER within the normal range, or borderline

AER, also identify patients at risk of progression to renal damage and is

associated with changes in glomerular filtration rates (54, 55).

2.5.3 Macroalbuminuria

Macroalbuminuria, clinical albuminuria or overt nephropathy is defined as

AER above or equal to 200 μg/min or 300 mg/24h.

Overt nephropathy is associated with a gradual reduction in the glomerular

filtration rates, which is variable from individual to individual in the course of

several years. End stage renal disease (ESRD) is characterized by uraemia

and by glomerular filtration rates less than 15mL/min per 1.73 m2 or when

treatment by dyalisis is required (56). End stage renal disease (ESRD)

develops in about 50% of T1DM individuals with overt nephropathy within 10

years (57).

2.6 Classification or Stages of DN

Alterations in renal function in T1DM follow a very characteristic pattern and

five different stages have been described in the course of development of

DN (56).

2.6.1 Stage I - Early Hypertrophy-hyperfunction

This stage, characterized by early hyperfunction and hypertrophy, is present

at diagnosis of T1DM even before the start of insulin treatment. The main

structural changes are increased kidney volume, increased glomerular size,

nephron hypertrophy and hyperplasia (56). Insulin treatment leads to

reduction in both renal size and glomerular filtration rate (GFR) suggesting

Page 38: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

17

that these changes are due to metabolic effects and partly reversible by

insulin treatment (58).

Both increased kidney volume and faster GFR during the first years of

diagnosis are associated with poor renal prognosis (59-62). Two hypotheses

have been proposed to explain these early renal dysfunctions induced by

diabetes. The “vascular hypothesis” claims that hyperfiltration is the primary

event due to abnormalities in vascular control, including increments in

plasma flow and intra-glomerular pressure. Hypertrophy would follow, after

compensatory tubular changes, aiming to prevent loss of water and

electrolytes (63-65). The “tubular hypothesis” suggests that the primary

event is renal hypertrophy, which is a consequence of hyperglycaemia and

hyperglycaemia-induced overproduction of growth factors and cytokines (66-

68).

2.6.2 Stage II - Glomerular Lesions without Clinical Disease

The second stage develops silently over many years. It is characterized by

histological changes without signs of clinical disease. However, renal

function tests and kidney biopsy reveal some changes. There is evidence of

increased glomerular and tubular basement membrane thickness and

mesangium expansion. Hyperperfusion may be the initiating event with

increased GFR and increased mesangial matrix production in a focal or

diffuse distribution (69). Characteristically, GFR is increased by 20 to 30%,

AER and blood pressure are normal at rest but physical exercise unmasks

changes in albuminuria (56).

2.6.3 Stage III - Incipient DN

The earliest clinical evidence of DN is the appearance of low but abnormal

levels of albumin in the urine (≤ 30 mg/day or 20 μg/min), or MA. A slow, but

gradual increase of albuminuria over the years is a prominent characteristic

of this phase of renal disease when blood pressure is rising. MA and

diabetic renal disease are closely linked (10, 49, 59, 70-75). The presence of

Page 39: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

18

MA usually indicates the beginning of DN but it is also often found in

essential hypertension as first described by Parving et al (76).

Persistent MA is involved in early renal and vascular disorders and predicts

the development of overt DN and increased cardiovascular mortality (77).

MA is considered an early sign of damage of the kidney and the

cardiovascular system (75, 78-80).

Diabetic nephropathy prevention strategies may involve the use of early

angiotensin-converting enzyme (ACE) inhibitor treatment and the control of

diabetes to reduce glomerular hypertension and hyperfiltration. It is a

general clinical practice to screen subjects with T1DM for MA. In adults with

diabetes andMA, studies suggest that early blockade of the renin-

angiotensin system prevent the progression to renal disease and the

progression of other microvascular complications (81-84). In the adolescent

T1DM population a large randomized controlled clinical trial is currently

being developed to address the benefits of early intervention witn

angiotensin converting enzyme inhibitors (ACEI) and statins in prevention of

diabetic nephropathy (85).

2.6.4 Stage IV - Overt DN

Without specific interventions, approximately 80% of T1DM subjects who

develop persistent MA have their albumin excretion increase at a rate of 10

to 20% per year to the stage of overt nephropathy or clinical albuminuria (≥

300mg/ day or ≥ 200μg/min) over a period of 10-15 years (1). In this stage,

diffuse and focal glomerulosclerosis develop. Once overt nephropathy

occurs, and without specific interventions, GFR gradually declines over a

period of several years at a rate that is highly variable from individual to

individual (2-20 ml/min/year) and hypertension develops along the way (56).

2.6.5 Stage V - End Stage Renal Disease (ESRD)

The fifth stage is the end stage renal failure with uraemia characterized by

glomerular closure and GFR less than 10ml/min (56). ESRD develops in

50% of T1DM subjects with overt nephropathy within 10 years and in more

Page 40: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

19

than 75% by 20 years (1). Subjects with nephropathy had a much poorer

survival than those without proteinuria. Forty years after onset of diabetes,

only 10% of patients who developed nephropathy were alive, whereas

greater than 70% of patients who did not develop nephropathy survived (3).

2.7 Pathogenesis of Diabetic Nephropathy

2.7.1 HYPERGLYCAEMIA AND DN

The true mechanism by which lack of glycaemic control predisposes to

vacular damage is not fully understood. Among the proposed mechanisms

linking hyperglycaemia and vascular disease, two contributing factors are

the advanced glycation end-products (AGEs) and the acceleration of the

aldose reductase or polyol pathway. Other mechanisms may also be

important including the activation of protein kinase C (PKC), enhanced

oxidative stress, altered expression and action of growth factors and

vasoactive mediators in the glomerular tissues (86).

Three of these pathways (Figure 2.4) are further described in the following

sessions:

Figure 2.4 Three main pathways implicated in the link between

hyperglycaemia and diabetic microvascular complications

[Source: The Journal of the American Osteopathic Association, Vol 100, No 10, October

2000]

Page 41: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

20

2.7.1.1 Formation of Advanced Glycation End-products (AGEs): The

Glycation Hypothesis Chronic hyperglycaemia and oxidative stress in

diabetes result in the formation and accumulation of advanced glycation

end-products (AGEs). In intracellular hyperglycaemia, these products are

formed primarily through non-enzymatic reactions between protein and

glucose, a process named glycation or non-enzymatic glycation (87).

In diabetes, AGEs accumulates in sites of microvascular injury, including the

kidney, the retina and within vasculature (88). Studies in diabetic populations

have shown that serum and skin AGEs are powerful predictors for the

development of retinopathy, neuropathy and nephropathy (89).

For diabetic nephropathy, the role of AGEs is highly supported by clinical

studies where serum levels of AGEs are significantly increased with the

progression to microalbuminuria and subsequently overt nephropathy (90).

Of note, the proximal tubule is the main site for reabsorption of filtered

AGEs. Glucose uptake in the proximal tubule is not under the effect of

insulin meaning that hyperglycaemia is directly translated into high

intracellular glucose levels which in turn lead to AGEs production and

oxidative stress (91).

AGEs also interact directly with cell surface proteins, activating intracellular

pathways to alter vascular biology. These cell surface proteins, named

RAGEs (scavenger receptor class A) are also involved in the development of

microvascular complications. In the mice model, overexpression of RAGEs

in vascular endothelial cells promote nephromegaly, mesangial expansion,

glomerular hypertrophy and sclerosis (92).

Glycation is not the only mechanism by which vascular damage happens in

diabetes. Nevertheless, AGEs contribute to diabetic pathology both on their

own and in combination with other pathways. Advanced glycation is only one

of the pathways by which renal injury develops in diabetes. An interaction

between metabolic and hemodynamic factors is likely to promote the

deleterious effects of the diabetic milieu in the pathogenesis of diabetes

complications.

Page 42: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

21

It seems likely that therapeutic agents that target multiple pathways be more

effective than an individual therapy in preventing diabetes associated renal

injury. In fact in rat models, a synergistic effect toward prevention of diabetic

nephropathy was demonstrated by blockade of the renin-angiotensin system

and inhibition of advanced glycation, This combined therapy was more

effective in preventing glomerulosclerosis and tubulointerstitial damage and

in reducing albuminuria (93). In the clinical setting, it remains to be

determined whether a combination of hemodynamic and metabolic

pathways is more effective than any individual pathway in preventing renal

injury.

2.7.1.2 Activation of Aldose Reductase (Polyol) Pathway This theory

postulates that during hyperglycaemic states, there is activation of the polyol

pathway. The polyol pathway reduces toxic aldehydes generated by reactive

oxygen species to inactive alcohols via aldose reductase (94). In parallel,

sorbitol dehydrogenase reduces glucose to sorbitol, and eventually into

fructose. Elevated intracellular glucose can increase the activity of aldose

reductase. The aldose reductase reaction uses NADPH (nicotinamide

adenine dinucleotide phosphate) as a co-factor, decreasing the availability of

this enzyme for other pathways, which are required to reduce intracellular

oxidative stress. The combination of decreased NADPH and increased

oxidative stress activate other pathways that promote cellular damage,

which in turn causes vasoconstriction and compromises blood supply (95,

96) (Figure 2.5). Therefore, the increased activity of the aldose

reductase/polyol pathway is likely to result in vascular damage secondary to

osmotic changes, induction of oxidative stress and reduced Na+K+-ATPase

activity.

Based on the polyol pathway, one would expect that the use of aldose

reductase inhibition would be an appropriate target for prevention of

microvascular complications. However, initial studies with aldose reductase

inhibitors in the 1980’s ans 1990’s suggested lack of significant effects for

neuropathy, retinopathy and nephropathy (97, 98). More recently, preliminary

Page 43: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

22

data on new generations of aldose reductase inhibitorsm is promising in the

management of diabetic peripheral neuropathy (99).

Figure 2.5 Activation of aldose reductase or polyol in diabetes. NADPH =

nicotinamide adenine dinucleotide phosphate

[Source: The Journal of the American Osteopathic Association, Vol 100, No 10, October

2000]

2.7.1.3 Activation of Protein Kinase C (PCK) Theory Protein kinase C is

a family of 12 structurally and functionally related serine/threonine kinases

involved in intracellular signalling related to vascular, cardiac, immunologic

and other systemic functions (94). Pathological activation of PKC can occur

in diabetes.

Some of its isoforms, particularly the β and δ, are expressed in the

vasculature. Abnormalities in PKC activity result in vascular changes in

retinal and renal blood flow, contractility, permeability and cell proliferation

(92).

Activation of PKC in blood vessels of the retina, kidney and nerves can

produce vascular damage tha includes increased permeability, increased

leucocyte adhesion and alterations in blood flow (100, 101).

Page 44: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

23

The proposed mechanisms to explain PKC activation in diabetes patients

and its link to diabetes microvascular complications are shown in Figure 2.6.

The use of β-selective PKC inhibitors has been shown to attenuate the

progression of experimental DN in rodents in the setting of continued

hyperglycemia, hypertension, and activation of RAS (102).

Oral Ruboxistaurin treatment, a selective PKC- β inhibitor, has sjown to

reduce vision loss, need for laser treatment, and macular edema

progression in patients with diabetes (103). In type 2 diabetes, treatment

with ruboxistaurin reduced albuminuria and maintained eGFR over 1 year

suggesting some benefit to patients with diabetic nephropathy (104).

Figure 2.6 Activation of diacylglycerol (DAG)-PKC pathway in the

pathogenesis of diabetic microvascular complications. Activation of PKC,

particularly β and δ isoforms, will promote vascular changes and changes in

gene expression, which contribute to the development of microangiopathy

[Source: Endocrinology and Metabolism of North America, Vol 33 (March), 2004)

Page 45: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

24

2.7.1.4 Other mechanisms

a) Oxidative stress-induced vascular pathology Hyperglycaemia

generates increased activity of the polyol pathway, AGE formation and

activation of the PKC pathway. All these biochemical changes result in the

production of reactive oxygen species and oxidative stress. Oxidative stress

is implicated in the pathogenesis of microvascular complications by causing

endothelium dysfunction, vascular leakage and altered motor tone (92).

b) Changes in expression and action of growth factors Many growth

factors, which regulate vascular biology and tissue fibrosis, have been

implicated in the pathogenesis of microvascular complications. Vascular

endothelial growth factor (VEGF) has been associated with microvascular

complications in children and adolescents with T1DM (105), and particularly

associated with DR (106). Profibrotic factors, transforming growth factor β1

(TGF-β1) and connective tissue growth factor (CTGF), are increased in

diabetic kidneys and are likely to contribute to the accumulation of

extracellular matrix and mesangium expansion (92).

In addition to the role of hyperglycaemia in DN, haemodynamic disturbances

promote the development of glomerulosclerosis and proteinuria. Among

these factors, systemic and intraglomerular hypertension and in particular

the role of vasoactive hormone such as angiotensin II will be discussed in

further detail.

2.7.2 HAEMODYNAMIC CHANGES AND DN

Both metabolic and haemodynamic insults trigger intracellular signalling

pathways which promote the release of cytokines and growth factors

mediating renal damage.

2.7.2.1 The Role of Systemic Hypertension

The impact of hypertension on DN is independent of other contributing

factors in children, adolescents and adults with T1DM (107).

Page 46: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

25

A sustained reduction in blood pressure seems to be the most important

single intervention to slow the progression of nephropathy in type 1 and type

2 diabetes. Diabetic patients with blood pressure < 130/80 mmHg rarely

develop MA and have a decline of GFR similar to age-matched normal

population. On the contrary, diabetic patients with blood pressure between

130/80 and 140/90 mmHg have a greater decline in GFR and 30% of them

develop MA or proteinuria over the subsequent 15 years (108).

Strict blood pressure control is important for preventing progression of

diabetic nephropathy and other complications in patients with type 2

diabetes. In the United Kingdom Prospective Diabetes Study (UKPDS),

each 10 mmHg reduction in systolic pressure was associated with a 12

percent risk reduction in diabetic complications; the lowest risk occurred at a

systolic pressure below 120 mmHg (109). Therefore, normotension is crucial

in the management of diabetes and in preventing DN.

Genetic factors determine a patient’s risk of hypertension and subsequent

DN. There is considerable evidence of familial clustering of long-term

complications of diabetes (110). A higher frequency of hypertension is

observed among subjects with DN and their first-degree relatives suggesting

that susceptibility to develop nephropathy may be influenced by an inherited

predisposition to essential hypertension (111). The susceptibility to DN was

investigated in a subset of 98 young T1DM adults from the Oxford Regional

Prospective Study showing that a higher paternal night to day ratio of ABP

and albumin to creatinine ratio were associated with the presence of

incipient DN (112).

In children with T1DM, parental hypertension is associated with higher 24-h

diastolic blood pressure prior to development of DN and has an independent

effect on longitudinal AER (113).

In microalbuminuric and proteinuric adults with diabetes, several studies

have demonstrated a benefifical effect on microalbuminuria and blood

pressure with the use of agents that block the renin-angiotensin system

(114-119). A more aggressive target for blood pressure control has shown to

reduce the development of incipient and overt nephropathy (120).

Page 47: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

26

A meta-analysis based on 698 normotensive adults with T1DM and MA who

received agents that block the renin-angiotensin system (RAS) concluded

that these agents have a clear beneficial effect on AER with a one-third

reduction in the progression to macroalbuminuria and a threefold regression

to normoalbuminuria (121).

Because of the central role of the RAS in the development of DN and the

beneficial effects of agents that interrupt the RAS in this condition, a number

of candidate genes encoding for various components of the RAS may

predict DN. This topic will be explored in more detail in the session

Genetics of Diabetes Nephropathy.

2.7.2.2 The Role of Glomerular Capillary Hypertension and Glomerular

Hyperfiltration in DN

Glomerular hypertension is central to the initiation and progression of DN.

Classical studies performed more than 20 years ago using renal

micropunctures suggested that glomerular hypertension, as observed in

experimental diabetes, has a central role in the initiation and progression of

diabetic nephropathy, even in the setting of normal systemic blood pressure

(122).

Elevations of 25-50% in the whole-renal glomerular filtration rate (GFR),

measured by creatinine clearance or using filtration markers, have been

described in many T1DM patients at diagnosis and during the first years

after diabetes onset preceding the appearance of proteinuria (123).

The mechanisms by which diabetic patients develop glomerular

hyperfiltration are unclear. Animal studies suggest that afferent glomerular

arterioles dilate more than the efferent ones, raising GFR and

intraglomerular pressure. These hemodynamic changes have been

proposed to mediate diabetic glomerulopathy in streptozotocin-induced

diabetic rats (124). Moreover, increased GFR and renal plasma flow have

been reported in newly onset type 1 and type 2 diabetes patients,

suggestive of afferent renal vasodilation. These studies showed elevated

filtration fraction, indicating glomerular hypertension (123, 125-127). Several

Page 48: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

27

mechanisms have been proposed as to leading to the afferent glomerular

vasodilation including elevated insulin levels (123), insulin-like growth factor

1 (128), advanced glycation products (129) and a direct effect of

hyperglycaemia (130).

In diabetes, glomerular capillary hypertension happens when the blood

pressure within the glomerulus increases due to impaired autoregulation of

the glomerular microcirculation. Vasodilatation of both afferent and efferent

arteriole occurs, more pronounced on the afferent side, leading to an

increase in the intraglomerular capillary pressure (131). In experimental

diabetes, glomerular hyperfiltration is also a result of increased tubular

reabsorption of glucose and sodium, which causes vasodilation due to

decreased tubuloglomerular feedback (132).

Glomerular hyperfiltration has been associated with the development of

diabetic nephropathy. A meta-analysis of 10 cohort studies involving 780

subjects followed for a median of 11 years suggest an increased risk for

diabetic nephropathy in patients with early glomerular hyperfiltration

compared to those with normal GFR (130). Conversely, a series of 426

T1DM patients followed for 15 years was unable to detect an association

between glomerular hyperfiltration and progression to incipient DN

suggesting that other factors, mainly poor metabolic control, might play a

more important role compared to glomerular hyperfiltration itself (133).

The intra-glomerular hyperfiltration and hypertension in diabetes will lead to

mechanical stretching of all glomerular components, including mesangial

cells and podocytes.

This mechanical injury promotes extracellular matrix accumulation through a

variety of mechanisms summarized in Figure 2.7, including the increased

synthesis of extracellular matrix, decreased activity of degradative enzymes,

induction of gene and protein expression of TGF-β1 (a profibrotic factor),

upregulation of glucose transporter GLUT-1 and angiotensin II (powerful

vasoconstrictor and trophic hormone), production of VEGF and an increased

inflammatory response in the mesangial cells (108).

Moreover, in DN mechanical stretch decreases podocyte proliferation,

increases apoptosis and promotes podocyte detachment from the

Page 49: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

28

glomerular basement membrane (108). Mechanical stretching of the the

glomeruli, and stretching-induced podocyte loss will ultimately lead to to

progressive podocyte loss and glomerulosclerosis in diseases associated

with intra-glomerular hypertension, including diabetic nephropathy (108).

Figure 2.7 Mechanisms of extracellular matrix deposition in mesangial cells

caused by mechanical stretch in DN

[Source: Hypertension, Vol 48, 2006]

2.7.2.3 Treatment of Glomerular Hyperfiltration

Diabetic renal injury is mediated by increased renal blood flow, increased

fitration factor, vasodilation of afferent arteriole and increased proximal

tubular sodium absorption which promote glomerular hypertension and

consequent activation of the RAS (134). Therefore, iIncreasd glomerular

pressure could be attenuated by the use of agents that block the

vasoconstrictive effects of angiotensin II on the glomerular efferent arteriole.

In fact, animal data support the beneficial effects of angiotensin 1

converting-enzyme inhibitors (ACEI) or angiotensin II receptor blockers

(ARB) in causing egression in glomerulosclerosis even in advanced phases

of renal disease (135).

Page 50: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

29

Several randomized controlled trials in patients with T1DM and T2DM

confirm that ACEI and ARB therapy can prevent the development of ESRD

(136-138), overt nephropathy (114, 139) and microalbuminuria (81, 140).

However, the effect of blocking the activity of the RAS with the use of ACEI,

ARB or even a combination of these therapies is partially limited due to the

angiotensin II and aldosterone breakthrough (141).

2.8 Genetics of Diabetes Nephropathy

Patients with T1DM face a 20-50% risk of developing ESRD requiring

dialysis or renal transplantation (142). As not all individuals with diabetes

develop nephropathy, it is likely that an interaction of environmental factors

and genetic susceptibility play an important role in the development of this

complication. There is evidence of familial clustering of diabetes nephropathy

and a higher incidence of kidney complications among siblings of probands

with DN indicating a strong genetic component (143, 144).

Several groups of genes associated with different metabolic pathways have

been proposed as candidates for influencing the development of DN.

Because the RAS is an important regulator of systemic and local circulation

and is involved in the pathogenesis of arterial hypertension (145), it has been

postulated that genetic polymorphisms of its various components are

important in the pathogenesis of this complication. Systemic and intra-renal

RAS plays an important role in the pathogenesis of diabetic nephropathy

(146). Local production of angiotensin II promotes intra-gomerular

hypertension via constriction of the efferent arterioles contributing to to the

development and progression of glomerulosclerosis. Angiotensin II

stimulates the production of several cytokines, such as TGF-β (147) or

reactive oxygen species (148) mediating accumulation of extra-cellular matrix

proteins and causing cellular dysfunction under diabetic conditions.

Clinical trials have demonstrated that blockage of angiotensin II with either

ACEI or ARB prevent or delay the progression of renal injury associated with

diabetes (136). Therefore, the impoprtance of RAS with respect to the

pathogenesis of diabetic nephropathy is well established.

Page 51: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

30

2.8.1 Genes of the Renin-Angiotensin System (RAS)

As accumulating evidence suggests that genetic susceptibility plays an

important role in the development of diabetic nephropathy (149-151), genes

encoding for some components of RAS, such as angiotensin-converting

enzyme, angiotensinogen and angiotensin II receptor type 1, have been

reported to be most probable candidate genes for DN.

The RAS comprises the renin, angiotensinogen (AGT), angiotensin I-

converting enzyme (ACE) and angiotensin II receptor type 1 and 2 (AGTR1,

AGTR2) genes (Figure 2.8)..

The renin gene, located on chromosome 1q32, it is expressed in the

juxtaglomerular cells of the kidney. Circulating renin catalyzes the conversion

of the AGT to angiotensin I.

The AGT gene, located on chromosome 1q42-43, is expressed in the liver.

Angiotensin I is vasoinactive and the conversion of angiotensin I to

angiotensin II is the key reaction of the RAS pathway. This reaction is

catalyzed by the enzyme ACE.

The ACE gene is located on chromosome 17q23. Angiotensin II, the major

bioactive peptide of the RAS, exerts its main effects by binding to two

transmembrane G protein receptors, namely angiotensin II receptors type 1

(AGTR1) and type 2 (AGTR2).

The physiological actions of angiotensin II on vasoconstriction, hyperthrophy

and catecholamine liberation from sympathetic nerve endings (152) are

mostly mediated by the subtype AGTR1 expressed in cardiovascular cells,

such as the vascular smooth muscle cells. The activation of AGTR1 by

angiotensin II triggers a number of cytoplasmatic siganling pathways which

will exert its functional significance in mediating vascular remodelling (153).

Genetic variants of the RAS components, AGT, ACE and AGTR1 genes,

have been implicated in the aetiology of hypertension, coronary artery

disease, myocardial infarction and non-diabetic renal disease. Therefore,

these genes are considered potential candidates involved in the

pathogenesis of diabetic renal complication (152, 154-160).

Page 52: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

31

Figure 2.8 The renin angiotensin system (RAS) and the role of ACE

[Source: The Journal of Molecular Diagnostics, Vol 2, August 2000]

2.8.1.1 The angiotensin 1-converting enzyme (ACE) gene and DN

ACE plays a key role in maintaining blood pressure and kidney function

through modulation of RAS and the kinin-kallikrein systems (152). Rigat et al

first described the relationship between the ACE insertion/deletion

polymorphism and its role in controlling ACE levels. In this study, an

insertion/deletion of a 287 base pair Alu sequence in intron 16 of the ACE

gene, the ACE I/D gene, accounted for almost 50% of the variance of serum

ACE and was the major determinant of serum ACE concentration. Subjects

with a DD genotype for the ACE gene had higher serum concentrations while

II individuals had the lower serum concentrations (161).

The D allele has been associated with increased serum ACE activity together

with enhanced conversion of angiotensin I to angiotensin II (162), inactivation

of bradykinin (163) and more rapid progression of renal disease (164).

The possible casual association between the ACE I/D polymorphism and the

development of DN was studied in mice. Diabetes was induced in mice

genetically engineered to have 1, 2, or 3 copies of the gene. After 12 weeks,

the 3-copy diabetic mice developed higher blood pressure and proteinuria,

which also correlated with increased plasma ACE levels (165).

Page 53: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

32

In humans, the genetic variation at the ACE gene, mainly the

insertion/deletion (ACE I/D) variant, has been found to be associated with the

development of DN in both type 1 and type 2 diabetes, as demonstrated by a

meta-analysis of almost 15,000 subjects between 1994 and 2004. Subjects

homozygous for the insertion allele (ACE I/I) had a 22% lower risk of DN

when compared to carriers of the D allele (166). More recently, a meta-

analysis on 63 studies from 1994 to 2010 involving 14,108 DN cases and

12,472 controls demonstrated a significant association between the ACE I/D

polymorphism and the risk of DN for all genetic models, particularly for the

development of DN in the Asian group with T2DM (167).

The prognostic value of the ACE I/D polymorphism for nephropathy in T1DM

was confirmed in 310 patients followed for 6 years. The D allele

independently favoured the development of incipient nephropathy (168).

Supporting these data, the DCCT/EDIC groups followed 1,365 T1DM

subjects and reported that presence of I/I genotype conferred a lower risk for

both persistent MA and severe nephropathy (169).

Most studies in the paediatric population have a cross-sectional design and

include only small numbers. Some have found that in T1DM normotensive

and normoalbuminuria children, the DD genotype was associated with higher

diastolic mean 24-h BP and lower diurnal variation (170) while others found

no correlation between ACE I/D genotype and BP or AER (171).

2.8.1.2 The angiotensinogen (AGT) gene and DN

Angiotensinogen is the only precursor of angiotensin II, a vasoconstrictor and

growth factor implicated in the development of glomerulosclerosis and

mesangial cell hypertrophy (172). Cleavage of angiotensinogen by renin is

the rate-limiting step in activation of the RAS.

A molecular variant involving a methionine-to-threonine transition at

aminoacid position 235 (the AGT M235T polymorphism) has a functional

effect. This polymorphism has been correlated not only with plasma levels,

with highest AGT levels observed in subjects homozygous for the T allele

compared to MT or MM, but also with essential hypertension and

Page 54: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

33

cardiovascular disease (173-175).The association between the AGT M235T

variant of the AGT gene has also been recognized among individuals with

early onset of familial hypertension (176).

Examination of the AGT M235T gene polymorphismIn for association with

DN has shown conflicting results. The presence of the TT genotype for AGT

was found to increase the risk for DN by almost 3-fold in a case-control study

involving 95 T1DM patients with nephropathy compared with 100 T1DM

patients without DN (177). An interactive effect between the AGT and ACE

genes was reported in T1DM subjects who developed DN (178). Others have

found that the T allele of the AGT M235T gene is associated with elevated

AER, but only when combined with the D allele of the ACE I/D polymorphism

(179). A gender-specific association of M235T polymorphism has been

described for DN in subjects with T2DM where the M235T variant contributed

to the risk of DN in men but not in women (180). Moreover, the TT genotype

was associated with a faster progression to renal failure in diabetic patients

as described in a large study involving 327 controls and 260 patients with

ESRD (181).

On the other hand, other studies have have failed to report an association

between AGT polymorphisms and DN (182-185) but demonstrated an

association between the TT genotype and elevated blood pressure (183).

Others have found that homozyosity for the M allele of the AGT gene

increased the risk for nephropathy either independently (186, 187) or in a

combinational effect with the D allele of the ACE gene (188).

2.8.1.3 The angiotensin II receptor type 1 (AGTR1) gene and DN

AGTR1 subtype, expressed in the vascular smooth muscle cells, is a

membrane-bound G-protein-coupled receptor that mediates the most

relevant biological properties of angiotensinogen II.

Bonnardeaux et al identified five silent polymorphisms at the AGTR1 gene,

the T573C, A1062G, A1166C, G1517T and A1878G and subsequently

described a significant allelic frequency of the C1166 polymorphism in

hypertensive individuals compared to normotensive controls (189). Since

Page 55: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

34

then, an association between the AGTR1 A1166C polymorphism and the

predisposition to hypertension, left ventricular hypertrophy, coronary artery

disease and myocardial infarction has been described (152). A study of 708

adults found a significant associtation between the presence of C1166 and

left ventricular mass (190). A recent meta-analysis of 16,474 subjects from a

total of 22 studies revealed that C1166 allele confered a higher risk of

hypertension (191).

In diabetes, a possible link between A1166C variant of the AGTR1 gene and

the increased risk for microalbuminuria was reported in T2DM patients (192).

More recently, the AGTR1 1166C allele was associated with lower

glomerular filtration rates, kidney dysfunction and with coronary heart

disease in patients with T2DM (193). In addition, the presence of CC at the

AGTR1 gene was more prevalent in T2DM subjects with microalbuminuria

compared to normoalbuminuric ones; CC AGTR1 significantly increased the

incidence of albuminuria after 10 years of diabetes duration (194).

In T1DM, a synergistic effect of the AGTR1 polymorphism and poor

glycaemic control increased the risk of DN in a case-control study of 152

patients, 79 being normoalbuminuric. The risk of nephropathy in the group

with poor glycaemic control during the first decade of diabetes was higher

among the C 1166 carriers compared with A 1166 carriers (195). More

recently, a meta-analysis was performed to estimate the risk of

polymorphisms at the AGT and AGTR1 genes associated with the

development of DN on 4,377 cases and 4,905 controls from 34 published

studies. Overall, a two-fold increased risk of DN was demonstrated for

subjcts with the AGTR1 CC 1166 compared to AA 1166 supporting that

AGTR1 polymorphism may contribute to DN development (196).

2.8.2 Other candidate genes

In addition to genes involved in BP regulation and the RAS, any gene that

could potentially mediate the functional changes observed in the

pathogenesis of DN can be regarded as a potential candidate.

Page 56: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

35

Among these genes are those involved with 1) synthesis and degradation of

glomerular basement membrane and mesangial matrix; 2) metabolic

pathways involved in glucose metabolism and transport; 3) cytokines, growth

factors and transcription factors; and 4) advanced glycation among others.

Polymorphisms in the aldose reductase gene have been implicated in DN

indicating a role of the polyol pathway in the pathogenesis of diabetic kidney

disease (197).

Genes involved in lipid oxidation, such as paraoxonase 1 and 2 genes, have

been described in association with increased risk for persistent MA in a large

group of T1DM adolescents (198).

Polymorphisms in the apolipoprotein E gene, a glycoprotein known to play a

key role in lipid metabolism, has also been associated with an increased risk

of DN in T1DM (199).

Genes involved in insulin resistance (ecto-nucleotide pyrophosphatase gene,

ENPP1 gene), glucose transport (GLUT1 gene), and inflammation

(interleukin-6, interleukin-6 receptor, fibrinogen genes) have been linked to

DN and vascular complications, but still need to be closely examined in the

paediatric population (200-202).

A summary of some of the candidate genes in which polymorphisms have

demonstrated in association with T1DM diabetic nephropathy are

summarized in Table 2.1.

Page 57: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

36

Table 2.1 Candidate genes in which polymorphisms have demonstrated

association with T1DM diabetic nephropathy

Class Gene Symbol Location

Renin-

angiotensin

system

Angiotensin-converting enzyme

Angiotensinogen

Angiotensin II receptor type 1

ACE

AGT

AGTR1

17q23

1q42-43

3q21-25

Glucose

metabolism

Aldose reductase

Glucose transporter-1

Receptor for advanced glycation end

products

AKR1B1

SCL2A1

RAGE

7q35

1p35

6p21.3

Growth

Factors

Transforming growth factor β1

Transformer growth factor β –

receptors I-III

Vascular endothelial growth factor

TGFB1

TGFBR1/2/3

VEGF

19q13.1

9q22,3p22,1

p33

6p12

Oxidative

stress

Paraoxonase 1 and 2

Superoxide dismutase 1 and 2

Haptoglobin

Catalase

PON1/2

SOD1/2

HP

CAT

7q21-22

21q22,

6q25.3

16q22

11p13

Lipid

metabolism

Apolipoprotein E

Adiponectin

Peroximase proliferator activated

receptor gamma

APOE

ADIPOQ

PPRγ

19q13.2

3q27

3p25

Inflammatory

factors

Interleukin-1

Intercellular adhesion molecule-1

IL1B

ICAM1

2q14

19p13.3

Others Endothelial nitric oxide synthase

Protein kinase C, β1

NOS3

PRKCB1

7q36

16p11.2

[Table adapted from Clinical Practice Nephron (203)].

Page 58: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

37

2.9 DN in Children with T1DM

Both genetic and environmental risk factors have been described in

association with DN (Figure 2.9).

Figure 2.9 Potential genetic and environmental risk factors for the

development of long-term diabetes complications

[Source: Pediatric Diabetes, Vol 8 (suppl 6), 2007]

Among the environmental risk factors involved in the appearance of DN in

T1DM children and adolescents are glycaemic control, diabetes duration,

puberty, hypertension, smoking, lipid disorders, family history of vascular

complications and obesity. These factors are commonly associated with the

development of macro and microvascular complications in both adults and

children with T1DM.

2.9.1 Risk factors for DN and microvascular complications in children

with T1DM

Glycaemic control Glycated haemoglobin is presently the best parameter to

predict the risk of developing long-term complications. Several Interventional

studies provide clear evidence in adults and adolescents that better

glycaemic control as measured by lower HbA1c levels is associated with fewr

and delayed microvascular complications (41, 204, 205).

Page 59: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

38

The Diabetes Control and Complications Trial (DCCT) was a randomized,

multicenter, prospective study involving 1441 patients with T1DM from 1983

to 1993 (41). In this trial, 195 subjects were pubertal adolescents (age 13-17

years) at recruitment (39). The DCCT demonstrated that intensive diabetes

treatment and lower HbA1c levels conferred a significant reduction risk for

both microvascular and macrovascular complications compared to

conventional treatment. In the adolescent cohort, there was a risk reduction

of background retinopathy of 53%, clinical neuropathy of 60% and MA of

54%. The difference in HbA1c after completion of the DCCT (median 7.4

year of follow-up) in the adolescent group was 8.1% vs. 9.8% (intensive vs.

conventional). The results observed in the adolescent group occurred

despite a higher HbA1c in the adolescent cohort treated by intensified

regimens compared to the total cohort in which the great majority were

adults (8.1% vs. 7.2%).

The Epidemiology of Diabetes Interventions and Complications (EDIC) study

continued to follow the DCCT cohort for another 8 years after the end of the

trial. Despite no differences in HbA1c at 4 years after completion of the trial,

the benefits of the intensive treatment and improved glycaemic control

during the DCCT persited and delayed the progression of microvascular

complications (Figure 2.10 and 2.11). In the former adolescent cohort there

was a 74% reduction in retinopathy, 48% reduction in MA and 85%

reduction in macroalbuminuria in those who received intensive treatment

(206). In addition, the EDIC study also showed a positive effect of intensive

therapy for reduction in of macrovascular disease (207).

Page 60: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

39

DCCT indicates Diabetes Control and Complications Trial; EDIC, Epidemiology of Diabetes Interventions and

Complications; HbA1c, glycosylated haemoglobin. Boxes indicate 25th and 75th percentiles of HbA1c level;

whiskers, 5th and 95th percentiles; heavy horizontal lines, medians; thin horizontal lines, means.

Figure 2.10 Distribution of HbA1c Concentration by Randomized Treatment

Group at the end of the DCCT and in Each Year of the EDIC Study

[Source: The Epidemiology of Diabetes Interventions and Complications (EDIC) Study,

JAMA, 2003]

MA defined as AER 28 µg/min, equivalent to 40 mg/24 h. A, Prevalence at the end of the Diabetes Control and

Complications Trial (DCCT) and during the Epidemiology of Diabetes Interventions and Complications (EDIC)

study. The differences between the 2 treatment groups are significant at each time point after DCCT closeout

(P<.001). B, Cumulative incidence of new cases in the EDIC study for those participants in the intensive- and

conventional-treatment groups with normal albuminuria at the beginning and end of the DCCT. The difference in

cumulative incidences is significant by the log-rank test (P<.001).

Figure 2.11 Prevalence and cumulative incidence of MA

[Source: The Epidemiology of Diabetes Interventions and Complications (EDIC) Study,

JAMA, 2003]

Page 61: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

40

The sustained effect reported by EDIC study suggests that the influence of

previous glycaemic control can leave an imprint on the cells of the

vasculature and target organs, favouring the future development of diabetes

complications, a phenomenon defined as “metabolic memory” (208). The

term glycaemic exposure, encompassing both the degree of hyperglycaemia

and time, and the concept of “metabolic memory” are supported by the long-

term influence of early metabolic control on clinical outcomes (208).

Although MA is rare before puberty, even in subjects with long diabetes

duration (12, 61, 209), the concept that pre-pubertal glycaemic control does

not affect the development of microvascular complications has changed over

the years. Several groups have demonstrated that glycaemic exposure

during the pre-pubertal years confers additional risk to the development of

MA and retinopathy (210, 211) . Schultz et al showed in a large cohort that

children diagnosed with T1DM before puberty had a latent period of time to

developing MA, which was then followed by a rapid development of MA after

the onset of puberty. In contrast, those diagnosed at or after puberty had a

constant rate of MA over time (210). Early glycaemic exposure, particularly in

the first years of diabetes, seems to accelerate the occurrence of

microvascular complications in childhood onset T1DM (212).

Experts agree that at present, the safest recommendation for improving

glycaemic control in all T1DM children is to achieve the lowest HbA1c that

can be sustained without causing severe hypoglycaemia while avoiding

prolonged periods of significant hyperglycaemia (blood glucose levels > 15-

20 mmol/L) and episodes of diabetes ketoacidosis (213). There is little age-

related scientific evidence for strict, i.e.nearl normal, glucose targets. Each

child should have their targets individually determined aiming at values as

close to normal as possible while avoiding severe hypoglycaemia as weel as

frequent mild to moderate hypoglycaemia. The targets in Table 2.2 are

proposed as guideline by the International Diabetes Federation and

International Society for Pediatric and Adolescent Diabetes (213).

Page 62: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

41

Table 2.2 Target indicators of glycaemic control in T1DM children and

adolescents

Level of

control

Ideal

(non-diabetic)

Optimal Suboptimal High Risk

Clinical assessment

Raised blood

glucose (BG)

Not raised No symptoms Polyuria, polydipsia

and enuresis

Blurred vision, poor

weight gain, poor

growth, delayed

puberty, signs of

microvascular

complications

Low BG Not low Mild and no

severe

hypoglycaemias

Severe

hypoglycaemias

(unconscious

and/or convulsions

Biochemical assessment – targets must be adjusted according to individual circumstances

Self monitoring blood glucose levels in mmol/L

AM fasting or

preprandial

plasma

glucose

3.6 – 5.6 5 -8 >8 >9

Postprandial

plasma

glucose

4.5 - 7 5 - 10 10 - 14 >14

Nocturnal

plasma

glucose

3.6 – 5.6 4.5 - 9 < 4.2 or >9 <4.0 or >11

HbA1c% DCCT standardized

DCCT <0.05 <7.5 7.7 – 9.0 >9.0

[Table adapted from Global IDF/ISPAD guideline for Diabetes in Childhood and Adolescence

(213)].

Page 63: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

42

Duration of diabetes and the effect of puberty In an incident cohort of

childhood onset T1DM, the effect of diabetes duration could be removed by

examining all patients at a median duration of 6 years (214). Increasing age

and later pubertal staging were associated with increased risk for early

elevation of albumin excretion (AER>7.5 μg/min) (Figure 2.12). In this study,

one year of age increased the risk of borderline AER (AER 7.5 – 20μg/min)

by 27% (OR 1.27, CI 1.12 – 1.45). Pubertal stages 4-5 conferred a five-fold

increased risk for borderline AER (OR 5.3, CI 1.71 – 15.99) over pubertal

stages 1-3 (215).

Cumulative pecentage of borderline AER

0

20

40

60

80

100

5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21

Age (years)

Pe

rce

nta

ge

(%

)

Figure 2.12 Cumulative percentage of borderline AER by age in T1DM with

diabetes duration of 6 years

[Source: Pediatric Diabetes, Vol 8 (suppl 6), 2007]

Postpubertal years of diabetes contribute more heavily to the risk of

developing microvascular complications (8, 210, 211, 216, 217). For the

same number of years of diabetes duration, children in puberty will have

increased risk for incipient DN and for retinopathy in comparison to pre-

pubertal children (36). By comparing adolescents with pre-pubertal and

postpubertal onset of diabetes, retinopathy developed at a shorter period of

diabetes duration among those with pubertal onset of diabetes indicating

that pubertal years have a greater impact in developing complications

compared to prepubertal years (211).

A recent multicentre study involving 105 patients aged 16-40 years

compared the effect of prepubertal duration on the occurrence of

Page 64: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

43

complications in T1DM patients after the same number of years of the

disease. The authors found that patients diagnosed in early infancy were

protected from microvascular complications but this effect disappeared if

lifetime metabolic control was poor. Instead, the onset of diabetes in puberty

was itself an independent risk factor for complications which was less

dependent of metabolic control suggesting that other factors such as blood

pressure influence these complications in pubertal years (218). Other have

reported similar findings for both retinopathy (217) and for MA (210).

The duration of glycaemic exposure in pre-pubertal years give a strong

additional risk to the development of MA and retinopathy. The Berlin

Retinopathy Study compared time to onset of retinopathy in children and

adolescents with pre and postpubertal onset of diabetes. The postpubertal

duration until retinopathy was shorter by 3 years in those with prepubertal

diabetes onset (211). In Sweden, a population-based study followed 94

T1DM children (age 0-14 years at diabetes onset) for a median of 12 years.

Inadequate glycaemic control in the first 5 years of diabetes accelerated

time to occurrence of DN (212).

The mechanism by which puberty confers a higher risk for DN is

multifactorial. Glycaemic haemoglobin levels during puberty are higher than

rcommended levels for prevention of complications. In the DCCT,

adolescents showed the same benefits from intensified therapy as adults but

HbA1c levels were generaly 1% higher and weight gain was more frenquent

(39). Children of pubertal age at T1DM onset have poorer glycaemic control

when compared to those diagnosed at a younger age (210). The association

between deterioration in glycaemic control and higher prevalence of MA

observed in puberty is consistent with a decrease in sensitivity to insulin

action observed during the pubertal stages (219).

The relationship between puberty and MA is only partly explained by poor

metabolic control and there is evidence that puberty itself may be an

independent factor (220). A more pronounced progression of albuminuria

occurs during puberty when compared to before or after this period

supporting the concept that other changes in pubertal hormones lead to

kidney damage (221).

Page 65: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

44

Growth hormone and IGF-1 have been associated with the pathogenesis of

early DN (222). Derangements in growth hormone and IGF1 are described

in adolescents with T1DM. In comparison to normal controls, diabetic

children excretes more urinary growth hormone at every pubertal stage

(223). Persistent MA in T1DM adolescents occurs more frequently during the

first stages of puberty compared to late pubertal stages (224). Adolescents

girls with MA have lower IGF-I and elevated testorenone levels when

compared to normoalbuminuric controls (225). In addition, girls have been

reported to have an increased risk of MA during puberty in comparison to

boys (210, 224). The observation that hyperandrogenism and low sex

hormone-binding globulin levels in T1DM adolescent girls accompany the

development of MA suggest the role of ovarian hormonal changes as a

contributor to the development of incipient DN and may, at least in part,

account for the sexual dimorphism in MA noticed in puberty (225).

In adolescence, the development of MA has also been associated with

hyperlipidemia (226), elevation of arterial BP (227), decline in renal function

(55) and retinal changes (228). Moreover, MA may represent the first

evidence of a generalized endotheliopathy (229). Markers of endothelial

function, measured by flow mediated dilation, and of early atherosclerosis,

measured by carotid artery intima-media thickness, are abnormal in

adolescents with T1DM (230).

Inflammation is a condition that is common to obesity (231) and T1DM (232),

and systemic inflammation is associated with the development of diabetic

nephropathy (233) and macrovacular complications in T1DM (234).

Increased systemic inflammation, measured by higher levels of hsCRP, IL-6

and fibrinogen, were demonstrated in youth with T1DM compared to non-

diabetic youth regardless of adiposity and glycaemic control (235).

Blood pressure Hypertension contributes more to cardiovascular disease in

diabetic patients than in non-diabetic individuals and is a major factor in the

development of all vascular complications including atherosclerosis,

nephropathy and retinopathy (236). Hypertension has emerged as a risk

Page 66: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

45

factor for the appearance and progression of microvascular complications

T1DM adults (43, 237, 238).

In normotensive children with T1DM, elevations in both systolic and diastolic

blood pressure as a continuous variable increase the probability of early

retinopathy (228). Interventional studies with Angiotensin Converting

Enzyme (ACE) inhibitors have shown a reduction in the progression of

retinopathy in adults with T1DM (239). Moreover, intensive blood pressure

control in normotensive T1DM adults slows the progression of nephropathy

(240) and retinopathy (241).

The prevalence of hypertension among patients with T1DM with normal

AER, determined on the basis of causal BP assessment at routine clinical

visits, does not differ from that in the general population (242).

In normotensive individuals, the systolic and diastolic BP variation follows a

circadian pattern in which values are lower at nighttime than during daytime.

In T1DM adults, abnormalities in this rhythm, or non-dipping status, precede

the development of clinical hypertension and have been associated with

increased risk of overt DN and early mortality (243). In addition, the

measurements of ABP give additional information about diurnal variation

during normal activities. Patients with T1DM and MA who are normotensive

on casual blood pressure assessments present with elevated ABP in

comparison to normoalbuminuric individuals (244). In T1DM adolescents,

higher 24-h ABPvalues , still in the normotensive range, have been

associated with persistent MA and subclinical signs of autonomic

neuropathy compared to normoalbuminuric patients and normal controls

(245).

In T1DM normotensive children, the non-dipping pattern has been reported

in association with incipient DN (246, 247). Of note, abnormalities of ABP

were also reported in association with high AER and family history of

hypertension suggesting that this measure may be helpful in detecting

T1DM children prone to develop DN (248).

Page 67: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

46

Lipids Lipoproteins contribute to the appearance of microvascular

complications in T1DM subjects. Reports from the DCCT/EDIC studies

confirmed dyslipoproteinaemia to be associated with the development of DN

in individuals with T1DM (32). In a cross-sectional study, lipoproteins from

968 subjects were analysed and renal dysfunction was associated with a

more atherogenic lipoprotein profile: higher total and low density lipoprotein

(LDL) cholesterol and higher triglycerides (32). The severity of retinopathy

was positively associated with triglycerides and negatively associated with

HDL cholesterol implying the involvement of dyslipoproteinaemia in the

pathogenesis of DR (249).

Hyperlipidaemia is commonly reported in adolescents with T1DM (226). A

one-third reduction of major vascular events is observed among T1DM

adults treated with HMG-CoA reductase inhibitors (statins) (250).The role of

statin treatment in children and adolescents with T1DM is unclear as there

have been no studies of HGM-CoA therapy in this population. Short-term

studies in children with familial hypercholesterolaemia show treatment to be

beneficial and safe (251). However, the effects of early statin intervention

on the development of micro and macrovascular complications in T1DM

children need to be addressed through longer-term trials.

Smoking Smoking is correlated with higher AER, MA and borderline

albuminuria in T1DM adolescents (252). Smoking is also associated with

poorer glycaemic control. In addition, an increased prevalence of retinopathy

has been reported in smoking versus non-smoking patients with T1DM

(253).

Family history of vascular complications The DCCT study provided

evidence that familial factors, independent of metabolic control, influence the

presence of DN (110). Despite poor glycaemic control, there are some

T1DM subjects who do not develop microvascular complications (254). This

and the observation of familial clustering of diabetes complications (110)

imply that hereditary factors modify susceptibility to microvascular

complications.

Obesity Higher BMI has been documented as a risk factor for MA (255)

retinopathy, neuropathy (256) as well as associated with for cardiovascular

Page 68: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

47

disease (257). In patients with T1DM, correlations among central obesity,

insulin resistance and MA have been reported (258, 259) suggesting a role

of central obesity in the pathogenesis of renal complications in T1DM.

Central obesity was found to be a strong predictor for the development of

MA in the EDIC study (260).

2.10 Screening guidelines for diabetes complications in T1DM children

The current guidelines for screening for diabetes vascular complications

recommended by the International Society for Pediatric and Adolescent

Diabetes (ISPAD) are summarized in Appendix A. ISPAD guidelines are

used by diabetes centres in Australia and New Zealand and are in

agreement with the position statement from the Australasian Paediatric

Endocrinology Group (APEG). The ISPAD Consensus Clinical Practice

Guidelines recommend annually screening from age 11 yrs with 2 yrs of

diabetes duration or from 9 yrs with 5 yrs duration will capture most children

and adolescents developing retinopathy and MA. For macrovascular

disease, the recommended strategy proposes that lipid profiles should be

assessed from age 12 yrs and then be performed every 5 yrs; blood

pressure measurements are recommended to be performed annually (261).

The American Diabetes Association (ADA) Annual screening suggest that

screening for MA should be initiated at age 10 yrs for children who have

diabetes for longer than 5 years; for DR this guidleines suggest the first

ophthalmologic examination once the child is 10 yrs or older and has had

diabetes for 3-5 yrs. For dyslipidaemia, ADA guidelines advise that

prepubertal children older than 2 yrs should have an initial screening at

diagnosis and for this to be repeated every 5 yrs. In pubertal children older

than 12 years, a fasting lipid profile should be performed at diagnosis and

repeated every 5 yrs (262).

The American Academy of Pediatrics recommends an initial eye

examination 3-5 years after diagnosis of diabetes in subjects older than 9

yrs, with annual follow-ups thereafter (263).

Page 69: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

48

CCHHAAPPTTEERR TTHHRREEEE

Page 70: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

49

CCHHAAPPTTEERR 33 SSTTUUDDYY II

““PPrreevvaalleennccee aanndd rriisskk ffaaccttoorrss ffoorr

mmiiccrrooaallbbuummiinnuurriiaa iinn aa ppooppuullaattiioonn--bbaasseedd

ssaammppllee ooff cchhiillddrreenn aanndd aaddoolleesscceennttss wwiitthh TT11DDMM

iinn WWeesstteerrnn AAuussttrraalliiaa

3.1 PREFACE This chapter was published and the full PDF is presented in

Appendix C

Page 71: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

50

3.2 Abstract

Objective. To define the prevalence and describe the natural history of MA

(MA) in a population-based sample of children with T1DM.

Methods. Children with T1DM diagnosed 16 years and screened for MA

were identified through the Western Australia diabetes register created in

1991. The diabetes register provides clinical information of all patients

followed routinely at the service. An audit from all data collected from 1991

and 2003 was used for the analysis. Three-monthly HbA1c was performed

from diagnosis. MA screening (AER in 3 timed overnight samples) was

yearly performed after 10 years of age or after 5 years from diagnosis. Cox

proportional model assessed the risk of different variables on the occurrence

of MA. Kaplan-Meier survival analyses (log-rank test) estimated the

probability of developing MA.

Results. Nine hundred and fifty-five T1DM children (462 male), mean

diabetes duration of 7.6 years, mean age at onset of diabetes of 8.5 years

were selected for the study. MA, mean AER 20 and < 200μg/min,

developed in 128 subjects (13.4%) at mean diabetes duration of 7.6 years.

Cumulative probability for MA was 16% after 10 years. Determinants for MA

were HbA1c (HR 1.21; 95% CI 1.05 -1.38; p=0.007), onset of puberty (HR

8.01; 95% CI 3.18 - 20.16; p<0.001) and age at diagnosis (HR 1.25; 95% CI

1.18 -1.33; P<0.001). Females had a higher probability for MA during

puberty than males (p=0.03). The total incidence of MA (subjects with

MA/100 person-years) was 1.26, 1.85 and 2.44 for those who developed

diabetes at ages < 5 years, 5-11 years and > 11 years, respectively.

Conclusions. Onset of puberty, diabetes duration and metabolic control are

major factors predisposing the development of MA. Children diagnosed with

T1DM at younger ages have a prolonged time for developing MA.

Page 72: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

51

3.3 Introduction

In subjects with T1DM, persistent MA (MA) is a predictor of nephropathy and

is associated with an increased risk of cardiovascular disease and early

mortality (3-7). Although a decline in incidence of DN has been reported

(264), between 20 to 80% of adults with T1DM and MA will progress to overt

nephropathy (5-7, 51).

Long-term sub-optimal glycaemic control is an important but not the only risk

factor for development of microvascular complications in T1DM (265, 266).

Other positive predictive factors for the appearance of DN include increasing

age, and duration of diabetes (10, 12, 210, 267), poor metabolic control

within the first five years (10, 268), dyslipidaemia, hypertension (267, 268),

smoking (253) and familial and genetic factors (269-272).

Although persistent MA is predictive for overt nephropathy in adults with

diabetes, its prognostic implications among children and adolescents are

less well defined. Duration of diabetes is a risk factor for MA in children with

T1DM (267, 273, 274), however, MA is uncommon before the onset of

puberty (61, 209, 224, 275). Puberty, characterised by changes both in body

composition, metabolism and hormones, is frequently associated with

deterioration in metabolic control in subjects with T1DM, and all of these

factors may be implicated in the development of incipient nephropathy (9,

222).

Princess Margaret Hospital for Children is the only referral centre for children

diagnosed with T1DM in the state of Western Australia and case

ascertainment levels of 99% are consistently achieved (23, 276). This

prospectively assessed cohort provides a unique opportunity to report the

characteristics and natural history of MA in a population-based sample of

childhood onset T1DM.

Page 73: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

52

3.4 Methods

Western Australia is the largest state in Australia with an estimated

population of 1.9 million. Aboriginal people account for 3.5% of the state’s

population and the remainder consists mainly of people of European

descent. This population is mainly distributed in the metropolitan areas with

approximately 25% of people living in the remote/rural areas. The

Endocrinology and Diabetes service at Princess Margaret Hospital for

children provides centralised medical care for all children with diabetes in the

state.

Nine hundred and sixty-nine children aged 0-16 years at onset of T1DM

were initially identified for this study through the Western Australia Children’s

Diabetes database created in 1991. All participants were screened for MA

between 1991 and 2003. Fourteen subjects had only been screened for

AER through one sample of spot urine, and were excluded from the

analysis. The remaining 955 subjects were studied.

As part of our standardised diabetes care program, subjects are seen 3

monthly at clinic with HbA1c being measured at every visit. Height and weight

are measured at each visit and blood pressure at every second visit.

Screening for MA, as by the estimation of AER from three consecutive

overnight urine samples, was performed yearly before puberty when

diabetes duration was more than 5 years or after 10 years of age. The age

of 11 years was used as a definition for the onset of puberty for both sexes

in accordance with other reports in literature (210, 224)

Ethics approval was obtained from the Princess Margaret Hospital Ethics

Committee. Informed consent was obtained from all subjects prior to their

data being stored on the diabetes database.

3.4.1 Definition of microalbuminuria

The classic definition of MA is the presence of AER 20 and 200 μg/min in

2 out of 3 consecutive urine samples. For this study, MA was defined as

mean AER, from three consecutive overnight urine samples, being 20

Page 74: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

53

μg/min and < 200 μg/min. A mean of 3.6 2.1 MA screenings/subject

(minimum 1, maximum 12) was performed.

The collection of urine samples was routinely performed at a time interval of

6-12 months and the results were stored in the database. AER collected at

longer intervals (more than 1 year apart) were also included. Urinalysis was

routinely performed to exclude infection and urine collections were not

performed in the presence of any intercurrent illness. Onset of MA was

considered the first occasion when an abnormal AER screening was

observed. Clinically, subjects that present an abnormal screening are

requested a second MA screening performed 6 months apart. In this case,

persistent MA was defined as the presence of a second positive screening

with mean AER 20 μg/min and < 200 μg/min.

All definitions of MA are somewhat arbitrary. By using the classic definition

of AER 20 and 200 μg/min in 2 out of 3 consecutive urine samples

twelve subjects would not be included in our analysis. All twelve subjects

presented with at least one AER in the MA range. The use of the mean AER

as the definition of MA did not affect the final results and therefore this

parameter was chosen to facilitate the analysis.

3.4.2 Laboratory evaluation

HbA1c – The mean HbA1c level for the entire follow-up period was used as a

reflection of metabolic control. HbA1c was followed 3 monthly from diagnosis

to either the first event of MA or during the entire follow-up period. Prior to

1993, glycosylated haemoglobin measurements were performed using an

HPLC (high performance liquid chromatography) assay. The assay was

changed from HPLC to Ames DCA 2000 early in 1993. In this method, HbA1c

is assayed by latex agglutination inhibition using DCA 2000 analyser (Bayer

Ltd, Indianapolis, IN, USA). The inter-assay coefficient of variability (CV) was

4.69% and 2.81% at HbA1c levels of 5% and 11% respectively. On the

changeover, the first 400 samples were analysed by both methods, which

allowed correlation between methods.

Page 75: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

54

AER assay - All overnight urine samples were collected and stored at

temperatures between +2 to +8C prior to testing. Urine analysis until 1997

were performed using timed overnight urine AER through Randox

Microalbumin competitive enzyme-linked immunosorbent assay (ELISA)

using rabbit antibodies to human albumin (Cat No MA1410), manufactured

by Randox Labs Ltd, Diamond Rd, Crumlin, County Antrim, N Ireland, UK.

The inter-assay CV was 8.5% and 8.9% for measurements of 9.7 mg/L and

97.6 mg/L respectively. From 1997 to 1999, the method was changed to

nephelometry on the Behring Nephelometer Analyser (BNA). The inter-

assay CV was 3.3% at 10.4 mg/L and 2.7% at 91.2 mg/L. A comparison

between the methods was done using regression equations (Randox ELISA

= 0.9472 Nephelometer + 1.39). From 1999 the AER method was changed

to the Tina-quant Albumin, an immunoturbidimetric assay using

Roche/Hitachi 917. The intra-assay CV was 2.2% at 38.43 mg/L. The inter-

assay CV was 6.4% at 14.15 mg/L and 1.5% at 83.78 mg/L. The latter two

methods were compared and the slopes were virtually identical with a

correlation coefficient close to 1.

3.4.3 Statistical analysis

SPSS 11.5 for Windows was used for statistical analysis, which included

independent two-tailed Student’s t test for differences in mean values

between two groups and Yate’s corrected chi-square for testing differences

in proportions. One-way ANOVA was used for differences in means within

more than two groups. Differences in incidence density were calculated

using the exact binomial probability. Kaplan- Meier survival curves were

used to estimate the probability of developing MA. The log-rank test was

performed to compare survival time. Cox Regression was used for modelling

time-to-event data. It estimated the contribution of each variable to the risk of

developing the outcome, after adjusting for known covariates. Duration of

diabetes was the time variable (unless otherwise stated) and MA was

considered the outcome. Gender, mean HbA1c, puberty and age at diagnosis

were included in the model as covariables. A P value of less than 0.05 was

considered to indicate statistical significance.

Page 76: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

55

3.5 Results

3.5.1 Clinical characteristics

For all 955 subjects, the mean diabetes duration at follow-up was 7.6 4.1

years (range 0.9-21.4). The mean age at onset of diabetes was 8.5 3.8

years (range 0.8-15.9) and the mean HbA1c was 9.2 1.4% (range 5.6-

13.9).

One hundred and twenty-eight subjects (13.4%) in the total cohort

developed MA at mean diabetes duration of 7.6 ± 3.8 years (range 1.3 –

17.2).

Forty-four subjects (4.6%) had persistent MA (second positive screening

within 6 months). MA regressed in 61 subjects (47.6% MA cases). In twenty-

three subjects no subsequent AER measurements were yet available until

the end of the study.

Six subjects (mean age of 12.0 ± 1.9 years; mean duration 7.0 ± 4.3 years

(0.6%) presented with macroalbuminuria as the first abnormal screening and

were not included in the analysis for MA. Among the 44 subjects with

persistent MA, three developed macroalbuminuria with a mean follow-up

period of 2.2 ± 0.8 years.

Hereafter, the term MA is used to refer to only the first abnormal values of

AER.

3.5.2 Microalbuminuria, metabolic control, age at diagnosis, gender,

duration of diabetes and puberty

Mean HbA1c was significantly higher among those who developed MA

compared to those who did not (10.1% 1.7 and 9.1% 1.3 respectively,

P<0.001) (Table 3.1).

Page 77: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

56

Table 3.1 Gender, HbA1c, incidence density and postpubertal incidence

density of MA for subjects aged < 5, 5-11 and > 11 years at diabetes onset

< 5 yrs 5-11 yrs > 11 yrs Total

n 197 475 277 949

M/F 99/98 212/263 149/128 460/489

Age (years) 15.1 3.5 15.7 2.9 17.7 2.3 16.2 3.1

Age at onset (years) 2.9 1.2 8.2 1.7 12.9 1.3 8.5 3.8

Duration (years) 12.1 3.4 7.5 3.4 4.7 2.5 7.6 4.1

MA subjects (M/F) 14/16 30/36 16/16 60/68

HbA1c (%) Non-MA

subjects

9.4 1.1 9.1 1.2 8.9 1.5 9.1 1.3

HbA1c (%) MA subjects 10.7 1.5 a

10.3 1.6 b

9.3 1.6

10.1 1.7 c

Total person-years (from

diabetes onset to the last

follow-up)

2386.5 3560.2 1306.2 7251.9

Postpubertal years 829.2

2261.2 1306.2 4396.6

Total incidence density of

MA

1.26

1.85 2.44 d

1.77

Postpubertal incidence

density of MA

3.25 2.78 2.44 2.77

HbA1c, glycated haemoglobin; MA, MA. Data are presented as mean SD or n unless

otherwise stated.

a P < 0.001 versus < 5 years non-MA group.

b P < 0.001 versus 5-11 years non-MA group.

c P < 0.001 versus whole non-MA group.

d P = 0.009 versus < 5 years total incidence density of MA.

Page 78: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

57

Mean HbA1c was higher among those who developed MA when age at

diagnosis was less than 5 years or between 5 - 11 years (10.7% 1.5 vs

9.4% 1.1 and 10.3 % 1.6 vs 9.1 % 1.2 respectively, P<0.001). There

was no difference in mean HbA1c between children with and without MA

aged > 11 years at diabetes onset.

Considering MA as the outcome and duration of diabetes as the time

variable, the probability for developing MA was 16% after 10 years and 37%

after 15 years of diabetes duration.

Girls represented 51.5% of the total cohort and no difference in gender was

seen among those with and without MA.

Similar age, duration of diabetes and mean HbA1c were observed between

boys and girls who developed MA. The Kaplan-Meier curve for the

appearance of MA was similar between males and females either using

duration of diabetes or age attained as the survival time.

Girls were more likely to develop MA during pubertal years (11 – 15 years)

(n=269) when age attained was used as the time variable (log-rank test,

P=0.03) (Figure 3.1).

Page 79: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

58

Figure 3.1 Cumulative probability (Kaplan-Meier survival curves, log-rank

test, P=0.03) for the development of MA for boys and girls during puberty (○

females, □ males).

Using 11 years of age as the defined age for onset of puberty, only 6 (4.7%)

of 128 individuals developed MA before puberty (mean age SD of 9.8 1.1

years) and three of them developed diabetes before 5 years of age.

Dividing the subjects into three groups: those who were aged < 5 years

(n=197), 5 - 11 years (n=475) and > 11 years at diagnosis (n=277) enabled

the evaluation of the effect of diabetes duration and puberty on the

development of MA. Kaplan-Meier survival curves as a function of age at

onset showed a highly statistical difference in the cumulative probability for

the appearance of MA across the 3 groups (log-rank test, P<0.001) (Figure

3.2).

Page 80: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

59

Figure 3.2 Cumulative probability (Kaplan-Meier survival curves, log-rank

test, P<0.001) for the development of MA for each year since diabetes onset

in three groups divided by age at diagnosis (+ < 5 years, □ 5 to 11 years and

○> 11 years)

Among those aged < 5 years at diabetes onset the probability for MA was

5% after 10 years of diabetes duration. This same probability was achieved

after only 2.5 years among those who were diagnosed with diabetes > 11

years of age. In addition, the increase in the probability of developing MA

remained constant over time among the oldest age-at-onset group while in

the youngest age-at-onset group, the cumulative probability for MA started

to increase after a period of 8 years of diabetes.

The total incidence density of MA, defined as the number of subjects who

developed MA per 100 person-years of follow-up, was highest among those

who were > 11 years of age at diagnosis of diabetes when compared to

those diagnosed < 5 years of age (P = 0.009). There was a trend in the

incidence rates of MA to be higher in the 5-11 group compared to those

Page 81: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

60

diagnosed < 5 years (P = 0.07) but no difference in the incidence rate of MA

was observed between the 5-11 and > 11 years groups at diagnosis. The

two groups who were prepubertal at diagnosis demonstrated a higher

incidence rate of MA after puberty (Table 3.1).

3.5.3 Contribution of the different risk factors for developing of MA

The proportional contribution of gender, puberty, age at diagnosis and mean

HbA1c (glycaemic exposure) for the development of incipient nephropathy

was measured using the Cox proportional hazard model. Modelling MA as a

function of glycaemic control up to the onset of MA or during the entire

follow-up and adjusting the model for gender, age at diabetes onset and

onset of puberty, an increase in hazard ratio (HR) of 21% per one

percentage unit increase in mean HbA1c was observed (HR 1.21; 95% CI

1.05 – 1.38, P=0.007). The probability of developing MA was higher after the

onset of puberty by a factor of 8. Also, an increase in HR of 25% was

noticed per one-year increase in the age of diabetes onset for a given

duration of diabetes (HR 1.25; 95% CI 1.18 -1.33; P<0.001) (Table 3.2).

Table 3.2 Proportional contribution of gender, puberty, age at diabetes onset

and HbA1c as risk factors for the development of MA with duration of diabetes

as the time variable (n= 949)

Variable Baseline

value

P value Hazard Ratio

Exp ()

95% CI for HR

Gender Female 0.72 1.07 0.74 – 1.53

Puberty Prepubertal 0.000 8.01 3.18 – 20.16

Age at diagnosis 0.000 1.25 a

1.18 – 1.33

Mean HbA1c (%) 0.007 1.21 b 1.05 – 1.38

HbA1c, glycated haemoglobin; HZ, hazard ratio; MA, micro albuminuria.

For gender and puberty, the probability for developing MA is expressed as a ratio relative to

the baseline values, female and prepubertal years.

a Hazard ratio per one year increase in age of diabetes onset.

b Hazard ratio per one percentage unit increase in HbA1c.

Page 82: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

61

3.6 Discussion

We report the association of MA with gender, duration of diabetes, age at

onset, metabolic control and puberty in a population-based cohort of children

with T1DM followed from diagnosis to a mean age of 16 years and a mean

duration of 8 years. In this cohort, the prevalence of MA of 13 percent was

similar to prevalence rates of 8- 18% that have been reported in cross-

sectional studies (8, 277-279)

As one of the main determinants for the development of microvascular

complications, our results confirmed that an increase in the mean HbA1c

increased the risk for developing MA over time for the total cohort. Poor

metabolic control directly correlates with hyperfiltration and renal

hyperperfusion in early stages of T1DM (280) and it has been reported as a

major risk factor for MA even among young subjects with diabetes (281). In

our cross sectional analysis between those with and without MA, subjects

diagnosed > 11 years presented no differences in the mean HbA1c. This

group had a shorter duration of follow-up and consequently a shorter

duration of exposure to high glucose levels, which could partially explain

similar mean HbA1c between those with and without MA. These results do

not exclude the effect of higher HbA1c as a risk factor over time in the

development of MA. For the total group a difference in mean HbA1c was

observed between those with and without MA. When adjustments for other

variables related to MA were performed, including duration of diabetes and

onset of puberty, mean HbA1c was still a remarkable risk factor in the

appearance of MA in this cohort.

In addition, the effect of diabetes duration on the risk of MA was not constant

over time. Subjects who developed diabetes at younger ages presented a

latent period of time with little risk for MA followed by an abrupt increase in

this risk, which coincided with the onset of puberty. By contrast, in subjects

developing diabetes after puberty, an increased risk for MA occurred at

earlier time after the onset of diabetes and at a constant rate.

Epidemiological data suggests that postpubertal years of diabetes contribute

more heavily to the risk of developing DN (8, 9, 278, 282) compared to the

prepubertal ones and persistent MA has been reported in T1DM adolescents

Page 83: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

62

during the early stages of puberty (224). Our findings suggest that children

developing diabetes at younger ages have a prolonged time to the

development of renal complication and may be relatively protected during

their early years of diabetes. However, once puberty is achieved, the

prepubertal years of diabetes contribute as an additional risk for MA. The

relative protection of development of MA in prepubertal years seems to be

overcome by a longer duration of diabetes and longer glycaemic exposure,

which give additional risk for MA after puberty. A higher incidence rate of MA

in postpubertal years among those diagnosed with T1DM earlier in life

suggests that a similar cumulative probability for MA across the age groups

might occur with a longer duration of diabetes, but longer follow-up is

needed to confirm these observations in our cohort. Similar findings were

reported by Donaghue et al who described an increasing delay in the onset

of retinopathy and MA in subjects with T1DM with a longer prepubertal

duration (217). A study by the Danish group revealed that prepubertal and

postpubertal duration of diabetes are contributors for the development of

retinopathy. Similar results were not observed for the development of

increased AER or MA. Our study involves a population-based cohort with a

different study design and different age for puberty onset which may have

contributed for the discrepancies in findings (283). Other factors other than

glycaemic control and duration of diabetes play a role in the development of

DN. The DCCT/EDIC and Pittsburgh epidemiology of diabetes studies have

demonstrated that multiple genetic susceptibilities, insulin resistance,

atherogenic lipoprotein profiles and inflammatory markers are associated

with increased risk of DN (258, 284, 285). The concept of microhypertension

or incipient hypertension has also been associated with the development of

MA. Early disturbances of blood pressure in normotensive subjects with

T1DM have been reported and increments of blood pressure occur in

parallel to AER even within the normal range (246, 286).

The mechanisms by which puberty increases the risk of the development

and progression of MA are not completely understood. Both the increase in

insulin requirements and insulin resistance observed in puberty might result

in poor glycaemic control (287) a well established risk factor for

Page 84: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

63

microvascular complication (265). Our data showed that the total incidence

of MA was higher among those who started with diabetes after puberty and

a rise in postpubertal incidence of MA was observed among those with

prepubertal age at diabetes onset, as described in the Oxford Regional

Prospective Study (210). As the group diagnosed with T1DM after puberty

who developed MA differed from the prepubertal one by having a better

glycaemic control, the proportional hazard model was applied and the

estimate probability for puberty was corrected for mean HbA1c. Still, puberty

was associated with an 8-fold increase in the likelihood of developing MA,

representing a strong independent risk factor for the development of MA.

Additional hormonal changes may contribute to kidney damage during

puberty other than deterioration in metabolic control. Experimental models of

DN have implicated sex steroids in its pathogenesis (288, 289). This study

did not show any differences in gender regarding the total number of

subjects who developed MA. When age was used as the time for the

development of MA, our results demonstrated that girls are at increased risk

of developing MA during puberty. A possible explanation for this finding is

the high androgen and low sex hormone-binding globulin levels that have

been associated with increased risk of MA during puberty, particularly in

females (225, 290). The development of hyperandrogenism and insulin

resistance secondary to polycystic ovarian syndrome is more common

among female adolescents with T1DM and could also explain the gender

discrepancies of MA during puberty. Other factors including changes in

growth hormone release and low IGF-I levels, more commonly reported in

girls with T1DM (222, 225, 278, 291-293), and glomerular hypertrophy and

hypertension (10, 61, 268, 279, 294) may also be involved in the

pathogenesis of MA during the pubertal years. In addition, as onset of

puberty at the age of 11 years was arbitrarily chosen, the discrepancies in

gender during pubertal years might reflect the fact that girls start puberty

earlier and have it for a longer period of time compared to boys. No

differences in gender were observed in the cumulative probability for MA

after puberty.

Page 85: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

64

In summary, the cumulative probability of MA in this cohort of children and

adolescents is strongly associated to the onset of puberty. Poorer metabolic

control, age at diagnosis and diabetes duration are also determinants for

MA. Children diagnosed at younger ages have a prolonged time to the

appearance of MA but once puberty is achieved, prepubertal years increase

the risk for renal complication. Although MA seems to regress in

approximately 50% during childhood, there is need for longitudinal follow-up

particularly of this high risk group for the development of overt DN and

cardiovascular complications later in life.

3.7 Acknowledgements

This study was supported by grants from the Diabetes Research

Foundation, Perth, Western Australia. We wish to thank Maree Grant,

endocrine nurse and clinical data manager at the Department of

Endocrinology and Diabetes, Princess Margaret Hospital for Children,

Western Australia for her invaluable advice.

Page 86: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

65

CCHHAAPPTTEERR FFOOUURR

Page 87: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

66

CCHHAAPPTTEERR 44 SSTTUUDDYY IIII

““Early changes in 24-hour ambulatory blood

pressure are associated with high normal

albumin excretion rate in children with type 1

diabetes”

4.1 PREFACE This chapter was published and the full PDF is presented in

Appendix C

Page 88: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

67

4.2 Abstract

Objective. The relationship between urinary AER and elevated blood

pressure (BP) is unclear as a cause-effect phenomenon in the development

of DN. The aim of this study was to examine the association between AER,

HbA1c and BP in children with normoalbuminuria.

Methods. 24-hour ambulatory BP assessment was performed in 78 children

with T1DM, mean age ± SD of 13.4 ± 2.7 yrs, range 7.3-18.3 yrs, mean

diabetes duration ± SD of 6.6 ± 2.9 yrs, range 2.1-11.9 yrs. Using

generalised linear mixed models with systolic (SBP) and diastolic (DBP)

blood pressure as dependent variables, the effects of AER and HbA1c were

examined, adjusting for age, gender, diabetes duration and insulin dose.

Results. Subjects with high normal AER (7-20 mcg/min) had higher SBP

during daytime and nighttime compared to the low normal AER ( 7

mcg/min) (mean SD of 118.20 7.98 and 110.33 7.08 mmHg, P= 0.02;

mean SD of 108.76 9.21 and 100.20 7.75 mmHg, P= 0.03,

respectively). DBP was also higher both during day and nighttime when

compared to the 7 mcg/min group (mean SD of 73.40 6.50 and 64.86

5.67 mmHg, P= 0.002; mean SD of 62.50 6.75 and 56.30 5.56 mmHg,

P=0.03 day and nighttime respectively).

Conclusion. A rise in SBP and DBP is associated with increased levels of

AER even within the normal range.

Page 89: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

68

4.3 Introduction

Nephropathy and hypertension remain major complications of T1DM mellitus

(T1DM). Persistent MA in diabetic patients is a risk factor not only for renal

disease but also for cardiovascular morbidity and mortality (4, 28, 295). The

cumulative incidence of DN in patients with T1DM is approximately 25-30%,

although a decline has been reported probably as a result of improved

glycaemic control (3, 296, 297). Several risk factors have been described

associated with the onset and progression of MA in children and adolescents

with T1DM including gender, HbA1c, duration of diabetes, pubertal status

(11, 61, 210, 268), as well as elevated BP and increased levels of AER (268,

298).

The prevalence of hypertension among patients with T1DM with normal

urinary AER, determined on the basis of blood pressure (BP) readings at

clinic visits, is similar to that in the general population (242).

Measurements of ABP (ABP) give additional information about the

differences between daytime and nighttime BP and about the diurnal

variation during normal activities. Patients with T1DM and MA often present

with elevated BP (detected using ABP monitoring) but it is unclear whether

the elevation precedes the appearance of MA or is concomitant with its

development (245, 299-301). Systolic and diastolic blood pressures follow a

circadian pattern in normotensive individuals with values being lower at night

than during the day. The loss of diurnal rhythm has been shown to correlate

with kidney disease (246, 247, 302), with a higher incidence of

cardiovascular disease and as an early sign of DN (247, 303, 304). The non-

dipping status (loss of the normal BP circadian pattern) has been related to

renal morphologic changes and to long-term hyperfiltration in

normoalbuminuric subjects with T1DM independent of duration of diabetes

(305).

In this study, we examined the relationship between blood pressure and

albumin excretion in a cohort of normoalbuminuric children with T1DM.

Page 90: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

69

4.4 Research design and methods

A total of 78 children with T1DM were randomly selected from the Children’s

Diabetic Clinic at Princess Margaret Hospital for Children, Perth, Western

Australia. All children with T1DM in the State are currently followed in this

service. None of the patients had evidence of other chronic illness nor had

received anti-hypertensive treatment. All study participants were

normoalbuminuric (urine AER <20 mcg/min in at least 2 out of three timed

overnight collections) and had no evidence of other microvascular

complications of T1DM.

Height, weight and pubertal status were assessed in all patients by the

clinician at the time of the visit. Pre-pubertal subjects were classified as

stage 1, early pubertal as stage 2, mid-pubertal as stage 3 and finally, when

puberty was complete as stage 4-5 according to Tanner (306).

4.4.1 Urinary AER assay

Three overnight urine samples were collected and stored at temperatures

between +2 and +8C prior to testing and were analysed within one week of

arrival at the laboratory. Urinary albumin concentrations were measured by a

Randox Microalbumin competitive enzyme-linked immunosorbent assay

using rabbit antibodies to human albumin (Cat No MA1410), manufactured

by Randox Labs Ltd, Diamond Rd, Crumlin, County Antrim, N Ireland, UK.

The interassay coefficient of variation was 8.5% and 8.9% for measurements

of 9.7 mg/L and 97.6 mg/L respectively. The AER method is sensitive

enough to detect levels as low as 1 mcg/min. The values less than 1 are still

detectable, but results are described as < 1 mcg/min. For the analyses

where the mean AER was used, AER’s reported as < 1 mcg/min were

considered 1 mcg/min.

4.4.2 Mean HbA1c levels

HbA1c is measured 3 monthly in all subjects and the mean HbA1c was

calculated from the onset of diabetes. HbA1c was assayed by latex

agglutination inhibition using DCA 2000 analyser (Bayer Ltd). Inter-assay

Page 91: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

70

coefficient of variation was 4.69 and 2.81% at HbA1c measurements of 5

and 11.5% respectively. The non-diabetic reference range is < 6.2%.

4.4.3 Assessment of BP

The 24-h ABP was monitored using an oscillometric portable automatic

monitor, the Suntech Accutracker® II (Suntech Medical Instruments,

Raleigh, North Caroline USA). The monitor was programmed to perform

systolic BP (SBP) and diastolic BP (DBP) measurements at 30-min intervals.

Patients followed their usual insulin regimen, avoided excessive physical

activity and none reported symptoms of hypoglycaemia. The use of the

monitor was explained to each participant and the appropriate cuff was

chosen and attached to the non-dominant arm. Subjects were instructed to

relax the arm during the blood pressure measurement.

For the purpose of ABP monitoring, two different periods were defined:

daytime which included all readings obtained from the time the subject woke

up to the time he went to bed and nighttime which included all readings from

the time he went to bed until the time he woke up.

The 24-h ABP measurements were statistically examined for mean systolic

and diastolic BP and respective standard deviations.

In this study, normal circadian variation was defined as a difference of 10/5

mmHg or more between mean daytime and nighttime blood pressure.

Patients were classified into “dippers” or “non-dippers” based on the

presence or absence of nocturnal dip (304).

4.4.4 Statistical analysis

Using the SPSS software package (version 11.5 for Windows), the

independent Student’s t test was used to compare means between the low

normal AER and the high normal AER groups.

Logistic regression analysis was used to examine the association between

several variables when considering dippers or non-dippers as the final

outcome.

Page 92: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

71

In order to use all repeated measurements, a generalised linear mixed

model with SBP and DBP as dependent variables was applied. The effects

of AER and HbA1c were examined adjusting for age, gender, duration of

diabetes, body mass index (BMI) and insulin/kg/day (SAS software

package). A P value of less than 0.05 was considered to indicate statistical

significance.

Page 93: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

72

4.5 Results

4.5.1 Characteristics of the subjects at the time of enrolment

Among the 78 subjects (40 males, 38 females) the mean ± SD age was 13.4

± 2.7 years (range, 7.4-18.3 years) and the mean ± SD duration of diabetes

of 6.6 ± 2.9 years (range, 2.1-11.9 years). Fourteen patients were pre-

pubertal, 27 were in early puberty (Tanner stage 2 and 3) and 37 were in

late puberty (Tanner stage 4). AER was performed in 75 subjects. Mean

AER, based on the mean of the 3 overnight urine samples, was within the

normal range in all of them (mean SD 3.58 2.88; range 1.00 – 18.57

mcg/min).

ABP was completely assessed in 65 subjects with measurements during

daytime and nighttime. The remaining subjects had some BP readings either

during daytime or nighttime and all measurements were included in the

mixed model.

The mean values for the 24-h ABP for each individual was in the normal

range. The 24-h BP mean ± SD for the SBP was 106.92 ± 12.08 and for the

DBP was 62.07 ± 11.12 mmHg. Mean ± SD daytime SBP and DBP were

110.92 7.39 and 65.49 6.12 mmHg, respectively. Mean ± SD nighttime

SBP and DBP were 100.72 8.03 and 56.68 5.78 mmHg, respectively

(307).

4.5.2 Subgroup analysis according to AER

Seventy-five subjects had a complete assessment of AER based on 3

overnight urine samples. Subjects were classified into 2 subgroups based on

the mean AER from the 3 samples. Mean AER less or equal to 7.0 mcg/min

were considered low normal albuminuria and mean AER above 7.0 mcg/min

but lower than 20 mcg/min were considered high normal albuminuria. The

cut off value of 7.0 mcg/min was used as this value corresponded to the 95th

percentile of normal for this cohort.

Page 94: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

73

The clinical characteristics of the 2 groups are shown on Table 4.1. Seven

patients presented with a “high normal” AER (4 males; 3 females) and all

were in puberty (Tanner stage 3 and 4).

Table 4.1 Characteristics of children and adolescents with T1DM with low

and high normal AER

Low Normal AER

(n=68)

High Normal AER

(n=7)

t test

Age (yrs) 13.4 2.7 15.2 1.6 NS

Duration (yrs) 6.6 2.7 7.5 4.3 NS

AER (mcg / min) 2.8 1.4 10.7 3.1 P = 0.002

Mean HbA1c (%) 8.6 0.8 8.9 1.0 NS

24-h SBP (mmHg) 106.7 ± 12.0 111.1 ± 12.6 P < 0.001

24-h DBP (mmHg) 61.7 ± 11.0 69.0 ± 12.4 P = 0.006

SBP –daytime (mmHg) 110.3 7.1 118.2 8.0 P = 0.02

DBP – daytime (mmHg) 64.8 5.7 73.4 6.5 P = 0.002

SBP-nighttime (mmHg) 100.2 7.7 108.7 9.2 P = 0.03

DBP–nighttime (mmHg) 56.3 5.6 62.5 6.7 P = 0.03

Results are expressed in mean ± SD. SBP, systolic blood pressure; DBP, diastolic blood

pressure

Mean AER was higher in the high normal AER group, as expected (mean

SD of 10.66 4.14 and 2.85 1.37 mcg/min respectively, P=0.002). No

differences were observed between the 2 groups regarding age, duration of

diabetes, gender, mean HbA1c or BMI.

The 24-h SBP and DBP were higher among those with high normal AER

levels compared to those with low normal AER (mean ± SD of 111.07 ±

Page 95: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

74

12.59 and 106.72 ± 12.02 for SBP, P=0.006 and 68.99 ± 12.43 and 61.74 ±

10.95 for DPB, P<0.001)

Daytime SBP was higher among those with high normal AER (mean SD of

118.20 7.98 and 110.33 7.08 mmHg, P= 0.02) and so was nighttime

SBP (mean SD of 108.76 9.21 and 100.20 7.75 mmHg, P= 0.03).

Diastolic BP was also higher among subjects with high normal AER (mean

SD of 73.40 6.50 and 64.86 5.67 mmHg at daytime, P= 0.002; mean

SD of 62.50 6.75 and 56.30 5.56 mmHg at night-time, P=0.03).

In addition, a significant increase in both SBP (P=0.02) and DBP (P=0.01)

was observed comparing the high normal AER group to the low normal AER,

adjusting for age, gender, mean HbA1c, duration of diabetes, body mass

index (BMI) and insulin/kg/day.

In order to analyse in depth the effect of increasing AER on SBP and DBP,

subjects were further stratified in 3 categories of AER: < 2.0 mcg/min (n=10),

2–7 mic/min (n=58) and 7–20 mic/min (n=7). The effect of increasing AER

was not present on SBP comparing the 7-20 mcg/min group to the 2-7

mcg/min during daytime but it was still significant at nighttime (daytime

P=0.09 and nighttime P=0.03). The effect of increasing AER both at daytime

and nighttime DBP was still observed between the 7-20 and the 2-7 group

(daytime P=0.01 and nighttime P=0.03). There was no significant effect of

increased AER on either SBP or DBP during daytime or nighttime between

the < 2 and 2-7 groups (Figures 4.1 and 4.2).

Page 96: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

75

AER (mcg/min)

7-202-7< 2

SB

P (

mm

Hg

)

130

120

110

100

90

Nighttime

Daytime

*

**

**

*

Figure 4.1 Effect of increments of AER on systolic blood pressure (SBP)

during daytime (■) and nighttime (●). Results are expressed as means and

95% CIs. P values represent the differences between the 2-7 and 7-20

mcg/min groups (* P=0.03; ** P=0.09).

AER (mcg/min)

7-202-7< 2

DB

P (

mm

Hg

)

90

80

70

60

50

Nighttime

Daytime

*

** *

**

Figure 4.2 Effect of increments of AER on diastolic blood pressure (DBP)

during daytime (■) and nighttime (●).Results are expressed as means and

95% CIs. P values represent the differences between the 2-7 and 7-20

mcg/min groups (* P<0.01; ** P <0.03).

Page 97: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

76

4.5.3 Subgroup analysis according to glycosylated hemoglobin level

Subjects were grouped according to their metabolic control based on the

mean HbA1c in 4 categories: HbA1c < 7.5%, between 7.5 and 8.3%,

between 8.3 and 9% and finally >9.0%. There was no effect of increasing

the mean HbA1c and changes in SBP. A significant effect was observed on

DBP, but only when comparing the group with mean HbA1c >9% with the

others groups (P=0.02).

4.5.4 Subgroup analysis on dippers and non-dippers

Among 65 subjects (36 males, 29 females) whose ABP was completely

assessed, 29 (44.63%) had non- SBP dipping and 18 (27.6%) had non-

diastolic dipping. Non-dipping in both systolic and diastolic blood pressure

was observed in 15 (23.1%) subjects.

Non-dipping for systolic and diastolic BP was more common among males

than females (P=0.05 and P=0.03, respectively) adjusting for age, duration

of diabetes, mean HbA1c and mean AER. Neither long-term glycaemic

control nor mean AER at the time of the study increased the risk for non-

dipping BP.

Page 98: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

77

4.6 Discussion

The relationship between elevated BP and incipient DN is important not only

to understanding the natural history of diabetic kidney disease but also when

considering intervention strategies that might prevent and delay the

development of overt nephropathy.

In this study, we found an increase in SBP and DBP both during daytime

and nighttime, detected by 24-h ABP monitoring, among normoalbuminuric

children with T1DM with higher AER levels. Even though borderline AER has

been defined differently by different authors, its development is associated

with a higher risk for developing incipient nephropathy (53, 54). In the Oxford

regional Prospective Study for Childhood Diabetes, higher tertiles of the

albumin excretion phenotype in children aged 11-15 years were predictive of

subsequent risk for the development of MA. Tracking the AER phenotypes in

a second independent cohort from Perth, Australia showed similar results.

All subjects who developed proteinuria had an AER in the upper tertile or

levels of HbA1c above 9%. (308).

Our findings are in accordance with studies that showed that higher 24-h BP

is present before the development of persistent MA among children and

young adults with T1DM (245, 247, 300, 301). Lafferty et al demonstrated an

incremental increase in all parameters of the 24-h BP from nondiabetic

controls through diabetic subjects with normoalbuminuria, intermittent and

persistent MA (245). Although our observations come from a cross-sectional

analysis of normoalbuminuric T1DM children who had never developed MA

(the classification of intermittent or persistent MA can not be applied in our

case) we still observed a simultaneous rise in BP and AER even in the

normal range. In spite of the lack a healthy matched control group in this

study, our data suggest that there is an early association between increased

AER and BP, which precedes MA in subjects with T1DM. These findings are

consistent with the observations of Garg et al who studied teenagers with

T1DM and concluded that 24-h ABP parameters were significantly higher

among those with borderline AER. In the same study, all BP parameters on

the 24-h ABP were similar for subjects with diabetes having AER’s less than

Page 99: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

78

7.6 mic/min when compared to matched healthy controls (301). Of note, in

our study, there was no significant effect of increasing AER on the ABP

when < 2 and 2-7 mcg/min groups were compared suggesting that BP rises

only when borderline AER levels are achieved.

Giving credence to the observations described by Schultz et al (227), our

findings are not consistent with the hypothesis that elevations in systemic BP

are the initiating factor for DN but support the idea of a simultaneous rise of

both BP and AER. However, it is clear that longitudinal evaluation is needed

to clarify if the changes in BP precede those in AER.

The loss of circadian rhythm of BP has been related to albuminuria (305,

309) and it has been associated with an increase in mortality rate of adults

with diabetes and overt nephropathy (243). Diabetes duration, long-term

metabolic control, BMI and mean AER were not associated with abnormal

systolic or diastolic dipping in this study. These subjects will need

prospective assessment to investigate the persistence of abnormal BP

dipping and its real impact on the development of DN.

We conclude that early changes in both systolic and diastolic BP occur

concomitantly with a rise in AER prior to the development of MA. Early

structural changes have been described in borderline MA and these subjects

require further attention to minimise associated factors such as higher blood

pressure in the development of nephropathy. The use of 24-h ABP is a

sensitive method that should be used to identify T1DM subjects at risk of

developing nephropathy.

Page 100: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

79

CCHHAAPPTTEERR FFIIVVEE

Page 101: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

80

CCHHAAPPTTEERR 55 SSTTUUDDYY IIIIII

““Declining Trends of Albumin Excretion Rate in

Children and Adolescents with Type 1 Diabetes

Mellitus”

5.1 PREFACE This chapter has not been published. It is presented in full in

the following pages.

Page 102: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

81

5.2 Abstract

Aim. The aim of this study is to examine the changes in prevalence of MA in

a population-based cohort of adolescents with T1DM between 1993 and

2004.

Methods. T1DM subjects (n=620, median age 14.8 years, median duration

6.6 years) matched by diabetes duration were selected for a cross-sectional

study of MA in three study periods (P1= 1993-1996; P2= 1997-2000 and

P3= 2001-2004). MA was defined as AER 20g/min and < 200g/min (2

out of 3 consecutive timed overnight urine samples). High AER was defined

as mean AER 7.5 g/min.

Results. High AER (P1 26%; P2 20% and P3 10%; P<0.001) and MA

prevalence (P1 5%; P2 6% and P3 2%; P=0.06; P2 6% vs. P3 2%; P=0.03)

declined over time. Metabolic control improved (mean HbA1c SD, P1 10.6

1.1; P2 9.4 1.1 and P3 8.4 0.8, P<0.001). Systolic and diastolic BP Z

scores decreased (mean SBP Z score SD, P1 0.46 1.13, P2 0.43 0.93,

P3 0.08 1.00, P<0.001 and mean DBP Z score SD, P1 0.16 0.73, P2 -

0.08 0.70 and P3 0.02 0.70; P=0.004) over time. More patients were

treated using intensive insulin management (P1 7%, P2 26% and P3 53%,

P<0.001) over time.

Conclusion. Prevalence of high AER and MA decreased in this cohort. Over

a 12-year period, intensified management and better metabolic control are

likely to have influenced changes in AER.

Page 103: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

82

5.3 Introduction

Diabetes management has been strongly influenced by the Diabetes Control

and Complications Trial (DCCT), which confirmed a relationship between

optimal metabolic control achieved by intense diabetes management and

reduced microvascular complications both in adults and adolescents with

T1DM (265, 266, 310). Although a secular decline in the incidence of DN

has been reported among T1DM adults over the past years in some western

countries (264, 311), others have found no trend toward a decrease in DN

despite evidence of better glycaemic control (312, 313)

In adults with T1DM, MA is a marker for incipient nephropathy and is

considered a risk factor for renal disease, proliferative retinopathy, and

cardiovascular morbidity and mortality (295, 314).

Persistent MA is rare before the onset of puberty (209, 210, 224). The

prevalence rates vary between 4 and 21% in childhood (12, 61, 278, 279,

294). Reports suggest that the presence of MA in the first decade of disease

will persist or progress into the second decade in approximately two thirds of

children with T1DM (315). Even those with borderline increases in

albuminuria have a relatively high rate of progression to persistent MA, and

at higher risk of developing incipient DN (54).

We have reported an overall prevalence of MA of 13% in our population and

a cumulative probability for MA of 16% after 10 years of diabetes duration .

Risk factors involved in the appearance of MA during childhood included

poor metabolic control, onset of puberty, duration of diabetes, age at

diagnosis and gender discrepancies, among others (210, 212, 279, 316,

317).

We hypothesised that improvements in glycaemic control and modifications

in diabetes management have reduced the prevalence of MA over time. The

aim of this study was to assess the changes in AER in a population-based

cohort of adolescents with T1DM screened for DN from 1993 to 2004.

Page 104: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

83

5.4 Patients and methods

5.4.1 Study design and patients

The Endocrinology and Diabetes service at Princess Margaret Hospital for

Children is the only referral centre for subjects diagnosed with T1DM under

the age of 16 years in the state of Western Australia with a case

ascertainment of over 99% (23). Clinical information has been collected from

all subjects since diabetes onset and data have been stored in the Western

Australia Children’s Diabetes database since 1987. All cases of T1DM have

been confirmed by auto-antibody testing.

Information on clinical features, on the results of blood tests and urine

protein of the patients was obtained from the diabetes database. All

individuals were seen three monthly in clinic. Records of glycosylated

haemoglobin (HbA1c), height, weight, body mass index (BMI), systolic (SBP)

and diastolic blood pressures (DBP) were collected at each visit. Information

on number of injections, total daily insulin doses and number of

hypoglycaemic episodes in the previous 3 months was also collected.

The estimation of AER was obtained from three consecutive overnight urine

samples following ISPAD guidelines. For children with prepubertal onset of

diabetes the AER screening was performed annually after 5 years of

diabetes or from the age of 11 years (whichever came first). For those

children diagnosed during or after puberty the AER screening was

performed annually after 2 years of diabetes onset (318).

For this study, adolescents aged above 11 years diagnosed with T1DM

between the years of 1986 and 2000 (n=1058) were initially identified. Those

with T1DM for at least 5 years were selected for this cross-sectional study

(n=620).

All participants were matched by diabetes duration. Participants were

randomly selected without replacement in three study periods based on the

year of AER screening: 1993-1996 (P1, n=169), 1997-2000 (P2, n=238) and

Page 105: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

84

2001-2004 (P3, n=213). Hence, subjects allocated in the first period were

not included in the second period or third one; patients allocated to the

second period were not included in the third one. One random AER

screening consisting of three consecutive overnight urine samples was

selected for each participant.

The mean of all HbA1c levels for each subject since diabetes onset was

used to measure long-term metabolic control. Persistent MA was defined as

a minimum of two out of three consecutive urine specimens with AER >= 20

and < 200 g/min. High AER was defined as the mean AER 7.5 g/min

and < 20 g/min. This cut-off was used based on findings that 95th centime

of urinary AER in healthy non-diabetic adolescents have been shown to be

less than 7.5 g/min (53, 252). The mean AER of the three timed overnight

urine collection for each subject was used as a continuous variable for all

statistical analysis.

The number of subjects with severe hypoglycaemic events in the 12 months

preceding the AER screening was used for analysis. Severe hypoglycaemia

was defined as an event in which the child was found semi-conscious or

unconscious (coma/convulsions).

Intense insulin management was defined as 3 insulin injections per day or

pump therapy. Conventional insulin management was defined as 1-2 insulin

injections per day.

Patients and their parents gave informed consent prior to their data being

stored on the diabetes database. Ethics approval for this study was obtained

from the Princess Margaret Hospital Ethics Committee.

5.4.2 Laboratory tests

AER Urine samples were stored at temperatures between + 2 to + 8C. Until

1997, AER was analysed through Randox Microalbumin competitive

enzyme-linked immunosorbent assay (ELISA) using rabbit antibodies to

human albumin (Cat N MA1410), manufactured by Randox Labs Ltd,

Diamond Rd, Crumlin, County Antrim, UK. The inter-assay coefficient of

Page 106: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

85

variability (CV) was 8.5% and 8.9% for measurements of 9.7 mg/L and 97.6

mg/L respectively. From 1997 to 1999, the method was changed to

nephelometry on the Boehring Nephelometer Analyser (BNA). The inter-

assay CV was 3.3% and 2.7% at 10.4 mg/L and 91.2 mg/L respectively. In

the changeover, 80 urine samples were analysed by the two assays with

measurements being virtually the same (Randox ELISA = 0.9472

Nephelometer + 1.39, r2=0.9727).

From 1999 the AER method was changed to the Tina-quant Albumin, an

immunoturbidimetric assay using Roche/Hitachi 917. The intra-assay CV

was 2.2% at 38.43 mg/L. The inter-assay CV was 6.4% at 14.15 mg/L and

1.5% at 83.78 mg/L. Sixty-two urine samples with measurements between 0

and 3030 mg/L were analysed by the two assays and highly correlated

(BNA=0.989 x Hitachi 917 – 1.20, r2=0.999).

HbA1c Prior to 1993, HbA1c measurements were performed using a high-

performance liquid chromatography (HPLC) assay. The assay was changed

to Ames DCA 2000 early in 1993, in which HbA1c is assayed by latex

agglutination inhibition using DCA 2000 analyser (Bayer Ltd, Indianapolis,

IN, USA). The inter-assay CV was 4.69% and 2.81% at HbA1c levels of 5%

and 11 % respectively. On the changeover, the first 400 samples were

analysed by both methods and the correlation coefficient was close to 1.

Total cholesterol Non-fasting cholesterol levels were assayed using a

colorimetric assay at 540nm using cholesterol esterase and oxidase as

adapted to the dry slide technology used in the Vitros 500 and 250 chemistry

analysers.

Anthropometric data and blood pressure Height (Harpenden stadiometer)

and weight (electronic weighing scales) were assessed and BMI was

calculated in kg/m2. Height, weight and BMI were transformed in to Z scores

for age and gender based on published standards (319). Measurements of

blood pressure were performed by auscultation with a sphygmomanometer.

Appropriate cuff sizes were chosen ensuring that the cuff bladder length

Page 107: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

86

would cover 80% of the circumference of the arm. Blood pressures Z scores

for gender, age and height were computed using published standards (320).

5.4.3 Statistical analysis

Summary statistics are expressed as median [interquartile range (IQR)],

number (%) and mean standard deviation (SD). SPSS version 13.0 (SPSS

for Windows, Rel. 11.0.1.2001. Chicago: SPSS Inc.) was used for statistical

analysis. Kruskal-Wallis test was used to compare continuous variables not

normally distributed across the three study groups (P1, P2 and P3).

One-way ANOVA was used for analysis of continuous variables with normal

distribution. For categorical variables, a chi-square (2) test was performed

to establish the association with the three time periods. Where statistically

significant results were achieved, the difference between individual groups

was assessed using the Student’s t-test or Mann-Whitney test.

Univariate analysis for predictors of high AER and MA was performed using

simple logistic regression. The variables studied were gender, age, age at

diagnosis, diabetes duration, mean HbA1c, total cholesterol, SBP and DBP

Z scores, BMI Z scores, insulin dose (U/kg), insulin management (intense

vs. conventional) and study period.

Multiple logistic regressions was performed to assess the combined effect of

different predictors of the development of MA and high AER. A strong

multicolinearity effect between mean HbA1c and the 3-study periods was

noticed (Pearson correlation –0.64, P<0.001). Therefore, two models were

used to assess the association of multiple variables and the outcome. In the

first model, mean HbA1c was included and the variable study-period was

removed from analysis. In the second model, study-period was included

instead of mean HbA1c, as stated in footnotes of Tables 5.4 and 5.5. A P-

value of <0.05 was considered statistically significant.

Page 108: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

87

5.5 Results

5.5.1 Patients’ characteristics

A total of 620 subjects (M=292; F=328) were studied. The overall median

(IQR) age and duration of diabetes were 14.8 years (12.9-17.1) and 6.6

years (5.7-8.2). The median (IQR) age at diagnosis was 8.3 years (5.7-10.7).

Mean HbA1c% SD was 9.4 1.4 and the median (IQR) AER was 3.0

g/min (1.5-5.7).

Duration of diabetes was similar across the three study groups. Group P3

was younger when compared to P1 and P2 and had an earlier onset of

diabetes when compared to P1 but not to P2. The clinical characteristics of

the subjects are summarised in Table 5.1.

Page 109: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

88

Table 5.1 Characteristics of T1DM adolescents the three study periods

P1 (93 – 96)

n=169

P2 (97 – 00)

n=238

P3 (01 – 04)

n=213

P value

Gender (M/F) 68/101 a

111/127 113/100 a

Year of Diagnosis 1986 - 1989

1990 - 1994

1995 - 2000

125

44

_

34

194

10

_

57

156

CSII subjects 0 2 (1%) 27 (13%)

Age (years) b 15.5 2.7

15.4 2.9

b 14.6 2.4 0.002

Duration (years) 6.7 (5.8 – 8.0) 6.7 (5.7 – 8.3) 6.6 (5.6 – 8.5) 0.428

Age at onset (years) c 8.5 2.9

8.1 3.6

c 7.6 3.5

0.01

Last HbA1c (%) 9.6 1.7 8.9 1.5 8.4 1.2 <0.001

Mean HbA1c (%) * 10.7 1.1 9.4 1.1 8.5 0.8 <0.001

AER (g/min) 4.2 (2.8 – 7.8) 3.5 (2.4 – 6.5) 2.0 (1.0 – 3.5) <0.001

Total cholesterol

(mmol/L)

4.64 1.04 4.54 1.03 4.38 0.79 0.05

BMI Z score 0.59 0.67 0.64 0.76 0.74 0.79 0.157

Weight Z score 0.55 ± 0.78 0.59 ± 0.82 0.74 ± 0.89 0.06

SBP Z score 0.46 1.13 0.41 0.93 0.07 1.01 <0.001

DBP Z score 0.16 0.74 0.13 0.73 0.00 0.72 0.001

N injections/day 1-2

3-5

157(93%)

12 (7%)

176 (74%)

62 (26%)

100 (47%)

113 (53%)

<0.001

Non-MA

MA

161 (95.3%)

8 (4.7%)

223 (93.7%)

15 (6.3%)

209 (98.1%)

4 (1.9%)

0.06

Low AER

High AER

125 (74%)

44 (26%)

190 (80%)

48 (20%)

191 (90%)

22 (10%)

<0.001

Number (%) subjects with severe

hypoglycaemia (12 months)

9 (5%) 32 (13%) 25 (12%) 0.03

Results are expressed in n (%), mean SD or median (IQR). * Mean of all HbA1c

measurements since the onset of diabetes. CSII, subjects on continuous subcutaneous

Page 110: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

89

insulin infusion. a

P1 vs. P3, -square test, P=0.01. b P1 vs. P3, Student’s t test, P=0.001.

c

P1 vs. P3, Student’s t test, P=0.002.

5.5.2 Albumin excretion rate and MA

The proportion of subjects with high AER decreased across the three

periods (Figure 5.1a). There was a declining trend in MA over the three time

points, mainly observed between P2 and P3 (Figure 5.1b).

01 - 0497 - 0093 - 96

STUDY PERIOD

30

25

20

15

10

5

0

% H

IGH

AE

R

01 - 0497 - 0093 - 96

STUDY PERIOD

7

6

5

4

3

2

1

0

% M

ICR

OC

AS

ES

Figure 5.1 Percentage of subjects with high AER (1a) and MA (1b) over the three

study periods

Page 111: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

90

To minimise the effect of age differences, subjects were grouped by age in

two-year intervals. The median AER was consistently lower in the third study

period compared to the first two study groups (p<0.01) (Figure 5.2).

>2018-2016-1814-1612-14< 12

Age (years)

5.00

4.00

3.00

2.00

1.00

0.00

Med

ian

AE

R (

mic

/min

)

2001-2004

1993-2000

Study period

Figure 5.2 Median AER by age, comparing P1+ P2 (1993-2000) vs. P3

(2001-2004)

5.5.3 Metabolic control, insulin management and hypoglycaemic

events

Improvements in long-term glycaemic control and glycaemic control on the

day of assessment were noticed across the three groups.

A higher proportion of subjects were intensively treated across the three

periods. The difference was significant both when P2 was compared to P1

(P<0.001) and when P3 was compared to P2 (P<0.001). There was a

parallel increase in the percentage of subjects on continuous subcutaneous

insulin infusion.

A higher number of subjects presented severe hypoglycaemia events across

the three time points but no difference was noticed in the number of subjects

with severe hypoglycaemia between the last two study periods (p=0.35)

(Table 5.1).

Page 112: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

91

5.5.4 Blood pressure, cholesterol levels and anthropometric data

There was an overall decrease in SBP and DBP Z scores and total

cholesterol across the three study-groups (Table 1). SBP Z score was lower

in P3 when compared to P1 (mean SD 0.07 1.01 vs. 0.46 1.13,

P=0.001) and to P2 (mean SD 0.07 1.01 vs 0.41 0.93, P<0.001). DBP

Z score was lower in P2 compared to P1 (mean SD -0.13 0.74 vs 0.16

0.74, P<0.001). No differences were observed between P2 and P3.

Total cholesterol was lower when P3 was compared to P2 (mean SD 4.38

0.79 vs 4.64 1.04, P=0.02) and compared to P1 (mean SD 4.38 0.79

vs 4.54 1.02, P=0.01).

No differences were noticed in height, weight and BMI Z scores over the

three periods.

5.5.5 Univariate and multiple logistic regression

The associated risk factors for high AER and persistent MA in the univariate

analysis are shown in Tables 5.2 and 5.3.

Table 5.2 Univariate logistic regression analysis for high AER

Outcome Variable Odd ratio

(95% CI)

P value

High AER

(AER 7.5 g/min

Age (years) 1.19 (1.11 – 1.28) < 0.001

Age at onset (years) 1.13 (1.06 – 1.20) <0.001

Mean HbA1c % 1.44 (1.24 – 1.68) <0.001

SBP Z score 1.24 (1.01 – 1.52) 0.04

Total cholesterol (mmol/L) 1.39 (1.10 – 1.76) 0.005

Study period

1993-1996 vs. 2001-2004

1997-2000 vs. 2001-2001

3.05 (1.75 – 5.35)

2.19 (1.27 – 3.77)

<0.001

0.005

Page 113: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

92

Table 5.3 Univariate logistic regression analysis for MA

Outcome Variable Odd ratio

(95% CI)

P value

Microalbuminuria Age (years) 1.27 (1.12 – 1.46) <0.001

Duration (years) 1.24 (1.02 – 1.49) 0.03

Mean HbA1c % 1.43 (1.08 – 1.89) 0.01

SBP Z score 1.46 (1.01 – 2.13) 0.04

DBP Z score 1.77 (1.01 – 3.10) 0.05

Total cholesterol (mmol/L) 1.62 (1.12 – 2.34) 0.01

Study period

1997-2000 vs. 2001-2001

3.51 (1.14 – 10.76)

0.03

In the multivariate analysis for risk factors associated with high/low AER,

older age at diagnosis and mean HbA1c was associated with high AER in

the first model. In the second model when study period was included and

mean HbA1c removed, the most recent study period was an independent

risk factor associated with high AER (Table 5.4).

Table 5.4 Multiple logistic regression analysis for high AER

Outcome Variable Odds Ratio (95% CI) P value

High AER *

(AER 7.5 g/min)

Age at diagnosis (years) 1.13 (1.01 – 1.28) 0.03

Mean HbA1c % 1.42 (1.17 – 1.73) <0.001

High AER **

(AER 7.5 g/min)

Study period

1993 -1996 vs. 2001-

2004

1997-2000 vs. 2001-2004

3.23 (1.66 – 6.23)

2.04 (1.07 – 3.91)

0.001

0.03

* Adjusting for: gender, age at diagnosis, diabetes duration, total cholesterol, SBP and DBP

Z scores and mean HbA1c. **Adjusting for: gender, age at diagnosis, diabetes duration, total

cholesterol, SBP and DBP Z scores and study period.

Page 114: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

93

The predictors for MA were older age at diagnosis and longer diabetes

duration both when the first and the second model were applied (Table 5.5).

Table 5.5 Multiple logistic regression analysis for MA

Outcome Variable Odds Ratio (95% CI) P value

Microalbuminuria * Age at diagnosis (years) 1.34 (1.11 – 1.62) 0.002

Duration (years) 1.74 (1.23 – 2.47) 0.002

Microalbuminuria ** Age at diagnosis (years) 1.35 (1.12 – 1.62) 0.002

Duration (years) 1.76 (1.25 – 2.49) 0.001

* Adjusting for gender, age at diagnosis, diabetes duration, SBP and DBP Z scores and

mean HbA1c.** Adjusting for gender, age at diagnosis, diabetes duration, SBP and DBP Z

scores and study period.

Page 115: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

94

5.6 Discussion

In this population-based cohort of adolescents with T1DM, we observed a

decline in the median AER in parallel to improvements in metabolic control

between 1993 and 2004. A trend towards a lower prevalence of MA was

noticed across the three study periods, particularly after the year 2000.

In view of the relative small number of MA cases, we analysed the

prevalence of high AER, which has been shown to confer a higher risk of

progression to diabetes nephropathy and to share the same risk

associations as MA (53, 54). A lower prevalence of high AER was seen

across the three periods supporting the finding of a decrease in AER over

time.

We have recently reported a prevalence of 13% of persistent MA in our

population of T1DM children followed prospectively from diagnosis (20). The

present study shows a lower prevalence of MA. However, this analysis

includes a smaller number of participants selected from the whole

population. In addition, it had a cross-sectional design and included one

AER screening (3 overnight samples) randomly selected for each subject

reflecting one single instant in time. As a cross-sectional study, it was less

likely to reflect the true prevalence of MA for the whole population.

Nevertheless, it clearly demonstrated declining trends of AER over time.

Different risk factors have been associated with the development of

microavascular complications among subjects with T1DM. Improvements in

metabolic control have been reported as contributing to the decline in

incidence of severe retinopathy and nephropathy in longitudinal follow-up

studies (35, 264, 311). A close relationship between glycaemic control and

early glomerular morphological changes is observed among young T1DM

subjects (321, 322). Better glycaemic control, as reflected by lower mean

HbA1c, was shown in this cohort across the three periods. Despite the fact

that the observed mean HbA1c was still above the target HbA1c level of

7.0% recommended by the DCCT (310), the decrease in the prevalence of

both high AER and MA suggests that minor changes in glycaemic control

Page 116: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

95

may affect AER. In addition, an increase in mean HbA1c was an

independent risk factor associated with high AER adjusting for different

covariates.

All participants were matched by diabetes duration but subjects in the last

study period were younger and had an earlier onset of diabetes. These

factors could be confounders for the lower prevalence of high AER and MA

observed in the last study period. However, the observation of a significant

decrease in median AER between the first two study periods and the last

one, when subjects were grouped by age in two-year intervals, gives support

to our findings of a decreased prevalence of high AER and MA specially

after the year 2000.

In spite of carefully matching the three study groups by diabetes duration

and selecting only adolescents for this study, our data showed that older age

at diagnosis and longer duration of diabetes were still independently

associated with MA. Others have also reported that children that develop

diabetes at younger ages have an increased time to the development of

incipient nephropathy compared to those diagnosed in pubertal years.

Postpubertal years of diabetes contribute more heavily to the risk of

developing DN (8, 9, 210, 317). The lack of detailed information on puberty

status for the whole study group did not allow us to analyse closely the effect

of different stages of puberty as risk factors associated with the development

of high AER and MA. The endocrine changes associated with puberty,

particularly in its later stages, lead to an accelerated process of early kidney

damage in diabetes and have been associated with deterioration in

glycaemic control during the pubertal years (221, 225, 323, 324).

Changes in the AER assays occurred three times over the study period of 12

years, which might have interfered with the MA screening. However, at each

changeover, methods were carefully validated with correlation coefficients

close to 1 and therefore were virtually identical in measuring AER.

Consequently, the declining trends observed in this study are unlikely to

reflect eventual fluctuations of AER secondary to changes in AER assays.

Page 117: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

96

Concomitantly to the decrease in mean HbA1c and median AER, insulin

therapy was intensified over time with a higher proportion of adolescents

intensively managed. Intensified insulin regimens have not always been

described as having an impact on metabolic control among adolescents with

diabetes and have been associated with increments in insulin dosage and

greater weight gain particularly in girls (325). Mohzin et al showed a decline

in the prevalence of retinopathy and MA among T1DM adolescents over

time in parallel to an increase in intensified insulin management but the

authors were not able to show changes in metabolic control (326).

Of note, our study was able to show improvements in metabolic control but

there was no evidence of increase either in weight Z scores or in BMI Z

scores across the three study periods. Insulin dosage only increased

between the first and second study-groups but not between the last two

periods.

Hypoglycaemia is the major obstacle to tight metabolic control in T1DM

children and adolescents (327). An increasing number of subjects with

severe hypoglycaemic events were seen between the first and second study

periods but no changes were observed between the second and third

periods despite improvements in metabolic control. These findings are in

accordance to previous reports in our population showing an increased

incidence of severe hypoglycaemia over the last decade but with a plateau

in the incidence rates after 1997 (328). This plateau may have been

achieved as a result of improved skills of the diabetes carers in preventing

hypoglycaemia and the introduction of better systems of insulin delivery such

as the use of continuous subcutaneous insulin infusion which is known to

reduce the risk of hypoglycaemic events (329).

Elevated BP and increased levels of AER have previously been described

being associated with the onset and progression of MA in children and

adolescents with T1DM (268, 298). A decrease in both SBP and DBP Z

scores was seen in parallel to the decrease in AER rates over time. This

finding gives credit to the hypothesis that changes in SBP and DBP occur

simultaneously with changes in AER and illustrates that improvements in

Page 118: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

97

metabolic control may have an impact not only on AER but also on BP levels

early in the course of diabetes (227, 286). It also supports the finding of a

true decline on AER over time because of changes in diabetes management

and unlikely to be an artefact of modifications on AER assays.

In conclusion, our results suggest a decline in the prevalence of high AER

and MA in a population-based cohort of adolescents with T1DM over time.

This decline appears to be directly related to improvements in metabolic

control and changes in diabetes management in the last decade but further

longitudinal evaluation is recommended.

Page 119: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

98

CCHHAAPPTTEERR SSIIXX

Page 120: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

99

CCHHAAPPTTEERR 66 SSTTUUDDYY IIVV

““AAnnggiiootteennssiinnooggeenn ggeennee TT223355 vvaarriiaanntt:: aa mmaarrkkeerr

ffoorr tthhee ddeevveellooppmmeenntt ooff mmiiccrrooaallbbuummiinnuurriiaa iinn

cchhiillddrreenn aanndd aaddoolleesscceennttss wwiitthh ttyyppee 11 ddiiaabbeetteess

mmeelllliittuuss””

6.1 PREFACE This chapter was published and the full PDF version is

presented in Appendix C

Page 121: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

100

6.2 Abstract

Aim. We examined genetic polymorphisms in the renin-angiotensin system

(RAS) coding for angiotensin-converting enzyme (ACE) insertion/deletion

(I/D), for angiotensinogen (AGT) M235T and for angiotensin II receptor type

1(AGTR1) A1166C as predictors for the development of MA (MA) in children

with T1DM.

Methods. 453 T1DM children (215 M, 238 F), median (IQR) age 16.7 yrs

(13.9 – 18.3), diabetes duration 6.9 yrs (3.3 – 10.8) and age at diagnosis 9.1

yrs (5.8 – 11.8) were followed prospectively from diagnosis until the

development of MA (2 out of 3 consecutive overnight urine samples with

AER 20 and < 200 g/min). Kaplan-Meier survival curves and Cox

proportional multivariate model estimated the probability of developing MA

and the relative risk for MA among different variables.

Results. MA developed in 41 (9.1%) subjects. The frequencies of the

genotypes were ACE-II 112 (25%), ACE-ID 221 (49%) and ACE-DD 117

(26%) (n=450); AGT-MM 144 (32%), AGT-MT 231 (51%) and AGT-TT 77

(17%) (n=452); AGTR1-AA 211 (47%), AGTR1-AC 204 (45%) and AGTR1-

CC 37 (8%) (n=452). The cumulative risk for the development of MA was

higher in ACE-DD vs. ACE-ID/II groups (log rank test, P=0.05) and a trend

was noticed when AGT-TT was compared to AGT-MT/MM groups (log rank

test, P=0.08). The AGT-TT polymorphism conferred a 4-fold increased risk

for MA compared to AGT-MM/MT (HR 3.8, 95% CI 1.43 – 10.3, P= 0.008).

Interpretation. Our findings suggest that RAS gene polymorphism at AGT

M235T is a strong predictor for early MA in young T1DM subjects.

Page 122: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

101

6.3 I Introduction

DN is a common complication in T1DM and is characterised by the

development of proteinuria, hypertension and decline in renal function.

Persistent MA is a predictor of overt nephropathy and an independent risk

factor for cardiovascular disease and early mortality in subjects with T1DM

(4, 5). In T1DM children the presence of MA in the first decade of disease

persists and progresses in approximately two thirds of T1DM subjects (330).

We have recently observed a cumulative risk for MA of 16% after 10 years of

diabetes duration in our paediatric population of T1DM. The main predictors

in this study were poor metabolic control, onset of puberty, duration of

diabetes and age at diagnosis (317).

Improvements in glycaemic control achieved through intensive management

reduce the development and progression of microvascular complications in

young T1DM subjects (39). However long-term follow-up studies show that

despite poor glycaemic control some subjects do not develop DN (254). In

addition to familial clustering, these findings imply hereditary factors may be

relevant in the aetiology of DN (144).

The renin-angiotensin system (RAS) is important in blood pressure

homeostasis and involved in the pathogenesis of arterial hypertension (145).

Genetic variants of its major components, angiotensinogen (AGT),

angiotensin I-converting enzyme (ACE) and angiotensin II receptor type 1

(AGTR1) have been implicated in the aetiology of hypertension, coronary

artery disease, myocardial infarction and non-diabetic renal disease and

therefore could be considered candidate genes involved in the pathogenesis

of diabetic renal complication (152, 154-160). An insertion/deletion of a 287

base pair Alu sequence in intron 16 of the ACE gene on chromosome 17q23

(ACE I/D) accounts for approximately 50% of the variability of serum ACE

levels between individuals. Subjects homozygous for the D allele exhibit fifty

per cent higher ACE levels than those homozygous for the I allele (161).

Associations between AGT gene polymorphism, plasma AGT levels and

essential hypertension have been described (174, 331). The AGTR1 is a

membrane-bound, G protein-coupled receptor that mediates the

Page 123: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

102

vasoconstrictive effects of angiotensin II. The AGTR1 A1166C polymorphism

has been associated with essential hypertension, increased left ventricular

mass and myocardial infarction (152) and is reported as a predictor for renal

injury in the presence of inadequate glucose control in adults with T1DM

(195, 332).

We hypothesized that genetic variations in the RAS contribute to the

development of persistent MA in children with T1DM. We examined specific

gene polymorphisms coding for ACE insertion/deletion (I/D), for AGT M235T

(rs5186) and for AGTR1 A1166C (rs11568053) in children with T1DM

followed from diagnosis.

6.4 Patients and methods

The Endocrinology and Diabetes service at Princess Margaret Hospital for

Children is the referral centre for subjects diagnosed with T1DM under the

age of 16 years in the state of Western Australia. Cases of T1DM have been

confirmed by autoantibody testing. Clinical information has been collected

prospectively from all subjects since diabetes onset and data has been

stored in the Western Australia Children’s Diabetes database since 1987.

Four hundred and fifty-three Caucasian children with T1DM (215 males, 238

females) with median age of 16.8 years (IQR 13.9 – 18.4) and median

duration of diabetes of 6.9 years (IQR 3.3 – 10.8) were randomly selected

from subjects regularly attending the diabetes outpatient clinic at our

institute. Case ascertainment for T1DM children diagnosed under the age of

16 years is 99% (23). Children with secondary diabetes, T2DM and with

other causes of nephropathy were excluded from this study. None of the

selected participants has had any previous treatment with ACE inhibitors.

Informed consent was obtained from parents prior to the child’s data being

stored on the diabetes database. Ethical approval for this study was

obtained from the Princess Margaret Hospital Ethics Committee.

Page 124: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

103

6.4.1 Design and procedures

Participants were seen once every three months in clinic. Records of

glycated haemoglobin (HbA1c), height, weight, systolic (SBP) and diastolic

blood pressures (DBP) and insulin regimen were collected at each visit.

In children with prepubertal onset of diabetes, the estimation of AER was

obtained from three consecutive overnight urine samples performed

annually after 5 years of diabetes onset or at the age of 11 years. In those

children with pubertal onset of diabetes, AER screening was performed

annually after 2 years of diabetes onset as recommended by the

International Society for Pediatric and Adolescent Diabetes guidelines

(ISPAD). Persistent MA was defined as the presence of a minimum of two

out of three consecutive urine specimens with AER >= 20 and < 200 g/min

(333).

All subjects were followed from diagnosis until the development of persistent

MA or until the end of the study in July 2005. The mean HbA1c since

diabetes onset until the end of the study was used as an indication of long-

term metabolic control. Information on anthropometric data, BP, lipids and

insulin dose from the last visit were used for statistical analysis. All

participants were screened for the ACE I/D, AGT M235T and AGTR1

A1166C polymorphisms.

6.4.2 Laboratory tests

Urine samples were stored at temperatures between +2 to +8C. Until 1997,

AER was analysed through Randox Microalbumin competitive enzyme-

linked immunosorbent assay (ELISA) using rabbit antibodies to human

albumin (Cat N MA1410), manufactured by Randox Labs Ltd, Diamond Rd,

Crumlin, County Antrim, UK. The inter-assay coefficient of variability (CV)

was 8.5% and 8.9% for measurements of 9.7 mg/L and 97.6 mg/L

respectively. From 1997 to 1999, the method was changed to nephelometry

on the Boehring Nephelometer Analyser (BNA). The inter-assay CV was

3.3% and 2.7% at 10.4 mg/L and 91.2 mg/L respectively. During the

transition period, measurements were made on the same samples by the

two assays and the two methods were compared and validated (ELISA=

Page 125: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

104

0.9472 x BNA +1.39, r2=0.9727). From 1999 the AER method was changed

to the Tina-quant Albumin, an immunoturbidimetric assay using

Roche/Hitachi 917. The intra-assay CV was 2.2% at 38.43 mg/L. The inter-

assay CV was 6.4% at 14.15 mg/L and 1.5% at 83.78 mg/L. Comparison

between the two methods was made and validated (BNA= 0.989 x Hitachi

917 - 1.20, r2=0.999).

Prior to 1993, HbA1c measurements were performed using a high-

performance liquid chromatography (HPLC) assay. The assay was changed

to Ames DCA 2000 early in 1993 in which HbA1c is assayed by latex

agglutination inhibition using DCA 2000 analyser (Bayer Ltd, Indianapolis,

IN, USA). The inter-assay CV was 4.69% and 2.81% at HbA1c levels of 5%

and 11%. Both methods have been DCCT standardised.

Cholesterol levels were assayed using a colorimetric assay at 540nm using

cholesterol esterase and oxidase as adapted to the dry slide technology

used in the Vitros 500 and 250 chemistry analysers.

Height (Harpenden stadiometer), weight (electronic weighing scales) and

BMI (kg/m2) were transformed to age and gender adjusted Z scores (319).

Blood pressure, measured after 5 minutes rest through auscultation with a

sphygmomanometer, was transformed to gender, age and height adjusted Z

scores (334).

6.4.3 Genotyping

Blood was collected into EDTA blood tubes and DNA was extracted from the

buffy coats using a salting out method (335). The 287bp Alu insertion in ACE

was genotyped directly by polymerase chain reaction (PCR) amplification,

followed by separation on a 2% agarose gel. The insertion allele was

identified by the presence of a 480bp band, the deletion allele by a 193bp

band (154). Samples that were identified as being DD homozygous were

confirmed using an insertion specific forward primer (5’–

TTTGAGACGGAGTCTCGCTC–3’). The DD genotype was confirmed by the

absence of PCR product.

The M235T AGT polymorphism was genotyped using a tetra-primer ARMS

assay (336) with forward outer primer 5’–

AGTCCTAGGGCCAGAGCCAGCAGAGAGG–3’, reverse outer primer 5’–

Page 126: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

105

CCGTTTGTGCAGGGCCTGGCTCTCTATA–3’, forward inner primer 5’–

CAGGGTGCTGTCCACACTGGCTCCGA–3’, reverse inner primer 5’-

GACAGGATGGAAGACTGGCTGCTCCCTTAC–3’. The T allele was

identified by the presence of a 166bp band, the C allele by a 121bp band.

PCR products were separated on a 2% agarose gel.

The A1166C polymorphism in AGTR1 was identified using mutagenically

separated-PCR, with A allele specific forward primer 5’–

TCTGCAGCACTTCACTACCAAATGAGTA–3’, C allele specific forward

primer 5’-

AAGGAGCAAGAGAACATTCCCTTGCAGCACTTCACTACCAAATGAACC–

3’, reverse primer 5’–TCAGAGCTTTAGAAAAGTCGG–3’. A 3% agarose gel

was used to separate the PCR products. The A allele specific product was

indicated by the presence of a 97bp band, the C allele specific product by a

117bp band.

6.4.4 Statistical analysis

SPSS version 13.0 (SPSS for Windows, Rel. 11.0.1.2001. Chicago: SPSS

Inc.) and SimHaP (337) version Beta 2.1 were used for statistical analysis.

Results were reported as mean standard deviation (SD); or median and

interquartile ranges (IQR) when distributions were skewed. Mean AER was

log-transformed. Groups were compared by the use of the chi-square (2)

test for categorical variables. Differences in continuous variables were

evaluated using ANOVA or Student’s t tests if normally distributed, or using

Kruskal-Wallis or Mann-Whitney U tests if distributions were skewed.

Kaplan-Meier survival curves estimated the probability for the development

of persistent MA. Duration of diabetes was used as the time variable.

Comparisons between the curves were performed using the log-rank test.

Genotypes at each locus were tested for deviations from Hardy-Weinberg

equilibrium separately in individuals with and without MA. A Cox regression

method was used to calculate the hazard ratios and 95% confidence

intervals for genotypes at the ACE, AGT and AGTR1 loci under additive,

dominant and recessive models adjusting for gender, age at diagnosis,

mean HbA1c, total cholesterol, insulin dose (U/kg), BMI Z score, SBP Z

score and DBP Z score.

Page 127: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

106

Genotypic models were tested by recoding genotypes with the most

common genotype coded as zero for the baseline. Under additive models,

heterozygous were coded as 1 and homozygous for the rarer allele as 2.

Under dominant models, both the heterozygous and homozygous for the

rarer allele were coded as 1 whilst under recessive models heterozygous

were also coded as zero and homozygous for the rarer allele were coded as

1. The threshold for declaring statistical significance was set at alpha=0.05.

Page 128: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

107

6.5 Results

Forty-one subjects developed persistent MA (9.1%). The overall cumulative

probability for persistent MA in this cohort was 11% after 10 years of

diabetes duration.(data not showed) Subjects in the MA group were younger

at diagnosis, had poorer metabolic control, required higher insulin doses and

had higher SBP and DBP Z scores and higher total cholesterol levels. There

was a trend for subjects with MA to have a longer duration of diabetes

(Table 6.1).

Table 6.1 Clinical characteristics of participants

Results expressed as median ± SD and median (Interquartie range).

The distribution of ACE, AGT and AGTR1 genotypes in this cohort of T1DM

children with and without MA was consistent with Hardy-Weinberg

equilibrium (Table 2). There were no significant differences in the genotype

frequencies between subjects with and without MA under additive, dominant

or recessive models. The allele frequencies of ACE, AGT and AGTR1

genotype were similar to previous data reported in other studies both in non-

diabetic and diabetic subjects (Table 6.2) (195, 338, 339). The distribution

of the three genotypes studied was similar between genders.

Page 129: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

108

Table 6.2 ACE, AGT and AGTR1 genotype distribution of participants

There was no difference among the ACE genotypes in terms of age at

diagnosis, duration of diabetes, mean HbA1c, log mean AER, SBP and DBP

Z scores or insulin dose (U/kg) in any of the models. Subjects identified as

ACE-DD had higher total cholesterol levels compared to ACE-II (mean SD

4.6 1.1 vs. 4.2 0.9 mmol/L, P=0.03) and higher BMI Z scores (mean

SD 0.84 0.67 vs. 0.55 0.90, P=0.02) but no differences between ACE-

DD and ACE-ID groups in the additive model suggesting a recessive effect.

However when ACE-DD and ACE-ID+II were compared, no differences were

observed in total cholesterol and BMI Z scores between the two groups.

Similarly, we found no differences in age at diagnosis, duration of diabetes,

mean HbA1c, log mean AER, SBP and DBP Z scores, BMI Z scores, insulin

dose (U/kg) and total cholesterol across the three AGT and AGTR1

genotypes under additive, dominant and recessive models. For the ACE

genotype, the Kaplan-Meier survival curves for duration of diabetes to the

development of MA showed a higher cumulative probability for persistent MA

for subjects ACE-DD in comparison to ACE-ID/II implying a recessive model

of action (log-rank test, P=0.05) (Figure 6.1). Under the recessive model,

thirty per cent of subjects homozygous for the ACE-D allele developed MA

after 12 years of diabetes while only 15% of those genotyped as ACE-ID/II

Page 130: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

109

developed MA after the same duration of follow-up. There was no difference

in the cumulative risk for persistent MA between the ACE genotypes under

the additive or dominant models.

Figure 6.1 Kaplan-Meier estimation for the development of persistent MA

according to ACE genotype in the recessive model (DD versus ID/II

There was a trend towards statistical significance of higher risk for MA for

subjects homozygous for the T allele of the AGT M235T polymorphism

under a recessive model (log-rank test, P=0.08) (Figure 6.2). Dominant and

additive models for AGT M235T showed no significant effect in the

cumulative probability for the appearance of MA. None of the models applied

to the AGTR1 genotype showed a significant effect on the cumulative risk for

MA.

Page 131: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

110

Figure 6.2 Kaplan-Meier estimation for the development of persistent MA

according to AGT genotype in the recessive model (TT versus MT/MM).

The multivariate Cox regression showed that subjects carrying two risk

alleles for the AGT polymorphism (AGT-TT) had a 4-fold increase risk for

persistent MA (Table 6.3). Epistatic interactions between loci were

investigated but no significant interactions were found. There was no

interaction between ACE, AGT or AGTR1 polymorphisms and gender or

long-term glycaemic control. In addition, we have analysed the interaction

between different genotypes, which lacked statistical significance in this

study. There was a positive correlation between insulin dose (U/kg) and

mean HbA1c (Pearson’s correlation, P<0.001) but no significant effect was

found between the interaction of these variables and persistent MA.

Page 132: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

111

Table 6.3 Cox regression multivariate analysis on the risk of persistent MA

in T1DM children

Page 133: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

112

6.6 Discussion

We identified an association between AGT M235T polymorphism and the

development of persistent MA in children with T1DM. This is the first

longitudinal study in an ethnically large and homogenous population-based

sample of T1DM children followed prospectively from diagnosis that has

demonstrated an increase risk for persistent MA, an early sign of DN and a

marker for cardiovascular disease, among subjects that are homozygous for

AGT T235. Although the relatively small number of subjects who developed

persistent MA (41/453) and the short follow-up time (median 6.9 years) need

to be carefully considered when these findings are interpreted, the 4-fold

increased risk for MA with the AGT-TT genotype strongly indicates an

association between polymorphisms in the RAS and the development of

incipient nephropathy in young T1DM. Subjects that developed MA had a

poorer glycaemic control and were older at diagnosis. There was a trend

towards a longer duration of diabetes among subjects with MA. Of note, in

this cohort, glycaemic control was not a predictor for MA in the multivariate

analysis. However, a strong correlation between mean HbA1c and insulin

dose was observed and a higher insulin dose increased by almost 3 times

the risk for MA. In addition, the overall glycaemic control for the total cohort

was above the recommendations from the DCCT (DCCT 1994) guidelines.

Doria et al have reported a synergistic effect of genetic polymorphisms at the

RAS and poor glycaemic control on risk of nephropathy in IDDM (195) and it

is likely that specific DNA sequence differences at the RAS genes may

modify the already existent noxious effects of hyperglycaemia on the kidney.

AGT is the only precursor of angiotensin II, a vasoconstrictor and growth

factor implicated in the development of glomerulosclerosis and mesangial

cell hypertrophy (172). Systemic and glomerular hypertension play a role in

the pathogenesis of DN (340-342). Cleavage of angiotensinogen by renin is

the rate-limiting step in the activation of the RAS and molecular variants of

the AGT gene, M235T in particular, have been shown to correlate with

higher plasma angiotensinogen levels and to be associated with essential

hypertension and cardiovascular disease (173, 174, 343). Some have found

that differences between genders in the genotype frequency confer different

Page 134: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

113

risk to DN for males and females (180), our study could not replicate these

findings and similar genotype distribution was observed between genders.

It seems unlikely that the 4-fold increase risk for persistent MA conferred by

the AGT-TT genotype is exclusively mediated by systemic hypertension as

the multivariate analysis showed that AGT T235 was an independent

predictor for incipient nephropathy, adjusting for systolic and diastolic blood

pressures. Although there was a trend in SBP Z score to be an independent

predictor for MA, there was no significant difference in SBP or DBP Z scores

across the three AGT genotypes for the whole group or for the MA and non-

MA groups, individually. However, changes in 24-hour blood pressure have

been associated with high levels of AER (286) and with different ACE

genotypes (170). Thus it is possible that minor blood pressure abnormalities

not detected as a clinical BP measurement could at least in part mediate the

relationship between AGT T235M polymorphism and the development of

persistent MA.

In adults with T1DM, reports on the association of AGT gene polymorphisms

and the development of nephropathy show discrepant results mainly due to

differences in study design, study population and measured outcomes.

Some have found the TT genotype of the AGT M235T polymorphism to be

more common among subjects with overt nephropathy and persistent MA

(177, 344) while others have failed to reproduce the same results (183, 184)

but described an association between higher blood pressure among AGT-TT

subjects with DN (183). More recently, a Dutch group found the T allele to be

associated with an increased risk of elevated AER but only when interaction

with the D allele of the ACE I/D polymorphism was considered (179). A

family-based study involving diabetic subjects and their parents found that

the T allele of the M235T polymorphism was transmitted preferentially to

those with the most severe manifestation of nephropathy, end-stage renal

disease (345).

In T1DM children, there is little information on the association of AGT

polymorphism and persistent MA. Most studies in the paediatric population

have a cross-sectional design and focus on the association between ACE

genotype, nephropathy and BP abnormalities (170, 171, 338). Barkai et al

found the DD genotype to be associated with higher diastolic BP in

Page 135: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

114

normoalbuminuric children with T1DM (170) while Pavlovic at al described

nocturnal BP abnormalities in children homozygous for the ACE-I allele

(338) but reported no association between I/D polymorphism and

nephropathy (171).

The higher cumulative risk for persistent MA found in ACE-DD subjects is in

agreement with most reports in the literature in which a causal relationship

between ACE gene function and DN in both type 1 and type 2 diabetes

mellitus is described (166, 169). In our study, there was no difference in SBP

or DBP across the three ACE genotypes neither for the whole group nor for

the MA and non-MA groups. Nevertheless we found an increase in total

cholesterol and BMI Z scores among those homozygous for the ACE-D

allele suggesting an association between the ACE polymorphism and lipid

profiles as probable contributors to cardiovascular disease in children with

T1DM(346, 347).

Finally, there was no significant association between the AGTR1 gene

A1166C polymorphism and the development of persistent MA. Although

some have reported a synergistic effect of AGTR1 genotype and poor

glycaemic control on the risk of DN (195) others failed to reproduce these

results in subjects with T1DM (348, 349).

In conclusion, our data gives evidence that genetic variability at the RAS, in

particular the AGT 235T allele, is a predictor for persistent MA in the T1DM

paediatric population. These findings provide a potential tool for identifying a

subgroup of children who may benefit from strategies targeted to early

prevention of DN.

6.7 Acknowledgement

This study was funded by the School of Paediatrics and Child Health, the

University of Western Australia, 2004 Channel Seven Telethon Grant

awarded to Professor CSY Choong.

Page 136: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

115

CCHHAAPPTTEERR SSEEVVEENN

Page 137: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

116

CCHHAAPPTTEERR 77 GGEENNEERRAALL DDIISSCCUUSSSSIIOONN

77..11 SSUUMMMMAARRYY &&CCOONNCCLLUUSSIIOONNSS

Although improvements in glycaemic control in T1DM adolescents reduce

the risk of appearance and progression of DN (350), MA remains the best

predictive marker for future development of overt nephropathy (351) and is

an independent risk factor for the development of cardiovascular disease

and early mortality in T1DM (3, 4).

Identifying children and adolescents at greatest risk for developing MA,

provides a unique opportunity for selecting individuals for early intervention.

The studies comprising this thesis have identified relevant clinical and

genetic predictors for the development of MA in a large population-based

cohort of children and adolescents with T1DM. These markers allow

selection of an at-risk group of children in whom intervention strategies

aimed at reducing the rate of micro and macrovascular long-term

complications can be trialled.

In the first study, outlined in Chapter 3, the natural history of MA was

examined in almost a thousand T1DM children followed longitudinally for a

mean duration of diabetes of 7.6 years. The overall prevalence of MA was

13%, similar to previous rates reported in the paediatric population (8, 210).

The cumulative incidence after 10 years of diabetes duration was 16%. The

major determinants for the development of MA were the onset of puberty,

older age at diagnosis and higher mean HbA1c levels since diagnosis. We

demonstrated that the effect of diabetes duration on the risk of MA was not

constant over time. Children diagnosed with T1DM after puberty had a

shorter time until the appearance of MA, shown as higher total incidence

density of MA, compared to the younger age-at-onset groups. These

findings are consistent with the hypothesis that the effect diabetes duration

Page 138: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

117

before and after puberty is non-linear in relation to the appearance of MA.

Similarly, Donaghue et al found a delay in the onset of MA and retinopathy

in children with longer prepubertal duration (217). However, it is important to

highlight that in the current study, the post-pubertal incidence of MA among

the young age-at-onset group was higher when compared with the older

age-onset groups. Our results indicate that the relative protection from MA

observed in subjects with pre-pubertal onset of diabetes is overcome by a

prolonged duration of diabetes and glycaemic exposure once puberty starts.

These findings are in agreement with the Berlin Retinopathy Study in which,

for the outcome of incipient retinopathy, the median post-pubertal duration

was shorter by 3 years in those with pre-pubertal onset of diabetes (211);

and with the Oxford Regional Prospective Study (ORPS) group, which

reported a strong contribution of prepubertal duration of diabetes and

hyperglycaemia to the appearance of postpubertal MA (210).

The effect of BP on the development of MA was studied in Chapter 4.

Elevated BP has been reported in patients with T1DM monitored with 24-

hour ABP but it is unclear whether this elevation precedes the appearance

of MA or is concomitant to its development (245, 299, 300). We addressed

this question by analysing the relationship between changes in AER and 24-

hour ABP in a cohort of almost 80 normoalbuminuric and normotensive

T1DM children. We were able to demonstrate that changes in 24-hour ABP

preceded the onset of MA. Children with borderline AER (AER 7.0-20

μg/min) had higher 24-hour ABP when compared to those with

normoalbuminuria. After adjusting for age, duration of diabetes, BMI,

gender and glycaemic control, subjects with borderline AER had higher

systolic and diastolic BP when compared with the normoalbuminuric group.

Our results support the hypothesis that the elevation in systemic BP parallels

the rise in AER and that these changes are detectable prior to development

of persistent MA. The ORPS group also found that the onset of MA was

associated with elevations in systolic and diastolic BP, measured in a clinical

setting, in a large cohort of T1DM children followed longitudinally (227).

Hence, we concluded from Chapter 4 that early changes in SBP and DBP

Page 139: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

118

occur prior to the development of MA; and that assessing 24-hour ABP is a

sensitive screening tool that could be used to identify T1DM children that

would benefit from intervention with antihypertensive agents.

In Chapter 5, we explored the changes over time in the prevalence of MA

and of borderline AER in a population-based cohort of 620 T1DM

adolescents screened for DN between the years of 1993 and 2004. Although

a secular decline in the incidence of DN has been reported among T1DM

patients over the past years in some western countries (264, 311), results

are controversial and others have found no trend toward a decrease in DN

despite evidence of better glycaemic control (312, 313). In a report from

Amin et al, the prevalence of MA was unchanged in a UK based incipient

cohort of T1DM children diagnosed from 1986 to 1996 despite

improvements in glycaemic control (352). In our study, we showed a decline

in the prevalence of incipient DN and borderline AER, which appeared to be

directly related to a decrease in mean HbA1c and to a more intensified

insulin therapy regimen. In addition, we noticed a similar decline in both

systolic and diastolic BP along with the changes in AER over time. Others

have described a decline in the prevalence of retinopathy and MA among

T1DM children over time in parallel to a more intensified diabetes

management but were not able to show improvements in glycaemic control

(326). Intensified insulin regimens have been reported in association with

greater weight gain (325) and hypoglycaemia is still a major obstacle in

achieving a better glycaemic control among T1DM children and adolescents

(327). In our study, despite improvements in glycaemic control, there was

no increase in BMI Z scores across the study periods. In addition, although

the prevalence of individuals with severe hypoglycaemia was increased

between the first and second study-periods, a plateau was observed after

1997 as previously shown by our group (328). In summary, the decline in the

prevalence of incipient DN occurred concomitantly with improvements in

metabolic control, to a decline BP Z scores and to a more intensified

diabetes management.

Page 140: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

119

In Chapter 6, we studied the relationship between specific genetic variants

in the RAS and the development of incipient DN. We investigated three gene

polymorphisms coding for ACE I/D, AGT M235T and AGTR1 A1166C in 465

T1DM children followed from diagnosis for a median of 6.9 years. The

cumulative risk for persistent MA was higher for subjects ACE-DD in

comparison to ACE-ID/II. After 12 years of diabetes duration 30% of subjects

genotyped ACE-DD developed persistent MA compared to only 15% of

those ACE-ID/II. Our findings support a casual relationship between ACE

gene function and the development of DN which has also been confirmed by

a meta-analysis of several studies comprising almost 15,000 T1DM and

T2DM subjects between 1994 and 2004 (166). In adults with T1DM,

discrepant results in relation to the association between AGT gene

polymorphisms and the risk for DN have been reported (184, 344). In T1DM

children, there is limited available information on the effect of AGT

polymorphism and the development of MA. Some have reported its

relationship with 24-hour BP changes in the T1DM paediatric population

(338) but most studies in the paediatric group focus on the association

between ACE genotype and DN in T1DM children (170, 171). We were able

to demonstrate that T1DM children homozygous for the T allele at the AGT

M235T gene have a four-fold increased risk for persistent MA; and that this

effect was independent of changes in systolic or diastolic BP Z scores.

Hence, our data give evidence to the hypothesis that genetic variants at the

RAS, particularly the presence of the AGT 235T allele, are predictors for

persistent MA and can be identifiable early in the course of diabetes.

77..22 LLIIMMIITTAATTIIOONNSS

Although this thesis has contributed to a better understanding of the natural

history of MA, and of the clinical and genetic predictors for the development

of incipient DN in T1DM children, a number of limitations must be

acknowledged.

Page 141: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

120

Firstly, identifying children at-risk of incipient DN based on the outcome of

the first MA event is not fully accurate as MA will regress in around 50% of

adolescents by the end of puberty; and a smaller proportion of these

subjects will progress to overt nephropathy (330). In Study I, we reported

that almost 50% of our MA cases regressed and approximately 35% of those

with one episode of MA had a second episode detected at least 6 months

later. However, the finding of increasing rates of AER identified as early as

one year from diagnosis, has been associated with increased risk of DN in

childhood T1DM (298). Moreover, recent evidence suggests that higher AER

in the upper tertile of the normal range strongly predicts those children who

go on to develop persistent MA (353).

In addition, although puberty was clearly a strong predictor for MA, the lack

of detailed information on puberty status for all participants did not allow an

analysis of the effect of different pubertal stages in the development of MA.

By using the chronologic age of 11 years as cut off for definition of onset of

puberty likely overestimated the effect of onset of puberty as an independent

predictor for MA. Neverhtless, the auhors have analysed the data using

older ages of 12 and 13 years for onset of puberty and the effect was

maintained (data not showed).

Although involving a complete cohort in Study I, we were unable to

document accurately information on other potential predictors for the

development of MA such as the effect of the lipoprotein profile or the

presence of inflammatory markers as predictors for the development of MA.

Hyperlipidaemia has been reported among adolescents with T1DM (226,

354). There is emerging data supporting the use of lowering lipid agents in

T1DM adults with MA in preventing cardiovascular events (250).

Inflammation appears to be involved in the pathogenesis of both macro and

microvascular complications. Inflammation occurs in the vasculature as a

response to lipid oxidation. Various others risk factors such as hypertension

and diabetes are amplified by the effects of oxidized low-density-lipoprotein

cholesterol initiating a chronic inflammatory reaction, which is associated

with increased risk of cardiovascular disease (355). Elevated CRP

concentrations have been found in association with increasing HbA1c

Page 142: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

121

suggesting a link between glycaemic control and systemic inflammation in

people with established diabetes (356). Low-grade inflammatory markers are

associated with DN in T1DM patients (357) and serum inflammatory markers

are elevated in subjects with severe DR (358). Therefore, the assessment of

inflammatory state would have provided a more complete picture of the

relative contribution of various factors to MA.

Due to constraints of a cross-sectional study design and a relatively small

group of children with T1DM involved in Study II, interpretation of results

should be done with caution. We were able to identify an effect of increasing

AER on both systolic and diastolic BP by stratifying the patients according to

AER in three normal categories. However, although our findings indicate a

simultaneous rise of BP and AER, blood pressure assessment of these

patients was measured in mmHg and are not reported as Z

scores/percentieles after appropriate adjustments for height and gender

which has been recommended in the pediatric population (359).

Abnormal SBP and DBP dipping has been previously reported in association

with albuminuria in adolescents (305). In our study, the lack of association

between abnormal BP dipping status and mean AER or glycaemic control

may be due to limited number of studied children. The study also have

limitations as power calculation indicated that a minimum of 80 paticipants

should have been recruited for detecting 50% of changes between dippers

and non-dippers.. In addition, from the studied participants, only 9% (7/75

participants) presented with high AER. Only a longitudinal evaluation of

these participants in would clarify whether these changes in BP precede

those in AER.

As discussed in Study III, in spite of carefully matching subjects of the three

study–periods by diabetes duration and selecting only adolescents,

participants in the last study period were younger and had earlier onset of

diabetes, factors that could be confounders for the lower prevalence of MA

or borderline AER observed in the last study-period. Similar to the

discussion in Study I, the lack of information on different stages of puberty

Page 143: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

122

did not allow us to assess the risk of each pubertal stage in the development

of MA or borderline MA.

Study IV, which addressed the effect of genetic variants in the RAS as

predictors for the development of MA, has a number of limitations and these

were detailed in Chapter 6. Only a small proportion of adolescents (41 of

453) developed MA over a short period of follow-up time (median 6.9 years).

We were unable to measure serum angiotensinogen levels in order to

correlate it to the variants at the AGT gene. Higher serum levels of

angiotensinogen are associated with polymorphisms at the AGT gene,

particularly with M235T, and are also associated with the development of

essential hypertension and cardiovascular disease (173). Additionally, we

failed to demonstrate the effects conferred by the AGT-TT genotype in the

development of MA being exclusively mediated by systemic hypertension as

no changes in systolic or diastolic BP were observed across the three AGT

genotypes, when measured in the clinical setting. The possibility of minor BP

abnormalities detected through more sensitive methods such as the use of

24-hour ABP could not be excluded from this study.

Similar to the lack of angiotensinogen serum measurements, we were

unable to correlate the ACE I/D genotype with ACE serum enzymatic

activity. Finally, a more prolonged follow-up and a larger study group would

probably have shown a stronger effect of the studied ACE and AGT

genotypes in the cumulative risk for MA.

77..33 IIMMPPLLIICCAATTIIOONNSS AANNDD FFUUTTUURREE DDIIRREECCTTIIOONNSS

The studies comprising this thesis have provided further insights into the

natural history of MA in children and adolescents with T1DM. The knowledge

gained from these studies contributes largely to identifying children at risk for

this complication. A combination of clinical and genetic factors is involved in

the pathogenesis of DN. Despite the decline in the prevalence of DN,

Page 144: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

123

improvements in glycaemic control do not eliminate the risk of this

complication.

Our studies highlight the importance of having discussions on prevention of

complications in T1DM, mainly DN, earlier in the clinical practice. Although

DN is rarely seen during childhood, poor glycaemic control since diabetes

onset contributes to an increased risk for MA and the child with diabetes

who receives limited care is more likely to develop diabetes long-term

complications at an earlier age.

Although screening programs for diabetes complications are costly to set up,

attention to risk factors and the impact of improved glycaemic control should

reduce long-term morbidity and mortality from diabetes complications.

Nevertheless, the development of screening programs for subclinical

complications is only appropriate if treatment intervention is available. There

is need for the development of intervention therapies in T1DM children and

adolescents with the ultimate goal of reducing the rates of DN and other

complications.

77..33..11 IInntteerrvveennttiioonn DDiirreecctteedd ttoo TT11DDMM CChhiillddrreenn aanndd AAddoolleesscceennttss

77..33..11..11 GGllyyccaaeemmiicc CCoonnttrrooll

Glycaemic control remains the most important risk factor for DN and

glycosilated heamglobin levels during puberty are invariably higher than

those levels recommended for prevention of complications (325).

Although the adolescent subgroup of the DCCT provided unquestionable

evidence for the superiority of an intensified insulin regimen in reducing the

risk for DN, excess weight gain and hypoglycaemia were more frequent

compared to adults (39). In addition, the beneficial effects of intensified

insulin therapy during puberty seem to be less evident than in younger

children and older individuals with T1DM (360).

Achieving near-normal glycaemia without increasing the risk of severe

hypoglycaemia in young T1DM children and adolescents require a great

Page 145: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

124

deal of effort on the part of both patients and those who care for them.

Nevertheless, based on our findings, one should recognize the effect of

prepubertal glycaemic exposure in increasing the risk for the future

development of MA and consider the concept of “metabolic memory”, in

which there is an imprinting phenomenon related to previous levels of

exposure to hyperglycaemia in determining the future development of DN

(206).

Therefore, in providing good quality diabetes care, every effort should go

into targeting a near-normal glycaemic control since the onset of diabetes,

with benefits and risks balanced and treatment individually tailored.

From our findings, we concluded that early signs of DN occur in puberty but

the relationship between incipient DN and puberty is partly mediated by poor

glycaemic control. Puberty itself exerts an independent effect in the

development of MA. Intervention on other modifiable factors in addition to

poor glycaemic control should be targeted in children and adolescents with

T1DM, particularly BP and lipid control.

77..33..11..22.. TThhee uussee ooff aannttiihhyyppeerrtteennssiivvee ttrreeaattmmeenntt

It is now over 30 years since Mogensen showed that the use of

antihypertensive treatment attenuates the rate of decline in renal function in

T1DM (361). In adults with T1DM and MA with or without hypertension, there

is a universal agreement in the usage of ACEI or ARB as these have been

shown to reduce the risk of progression of MA to overt DN and of mortality

from CVD (362).

In the paediatric population, although screening guidelines for MA are

routinely recommended (261, 262), recommendations on who should

receive treatment with renoprotective agents and when are not consistent.

There are still some concerns regarding the use of ACEI in protecting long-

term renal function in young people without hypertension. Some clinicians

treat only if retinopathy and hypertension coincide, others will treat patients

with a familial history of hypertension or CVD, and still others who will treat

all patients that develop MA (363).

Page 146: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

125

There is no consensus as to the use of either ACEI or ARB during

adolescence.

ACE Inhibitors (ACEI)

As mentioned previously, our findings showed a strong association between

ACE I/D and AGT M235T variants with increased risk for DN as well as

changes of ambulatory BP preceding the appearance of MA. Due to the

known renoprotective effect associated with antihypertensive action, drugs

that affect the RAS are potentially the first choice in preventing DN.

In a meta-analysis of 12 studies involving 698 adults with T1DM, patients

receiving ACEI had a reduction in AER of 50% compared to those receiving

placebo. In the normotensive T1DM patients with MA, ACEI reduced

progression to overt nephropathy (121).

In the paediatric group, only a small number of studies have been performed

suggesting the potential benefits of ACEI in the adolescents T1DM group

(364, 365) However, most of these studies have included small number of

subjects followed for a short period of time. There is an urgent call for long-

term randomized control trials involving the use of ACEI the paediatric

population. Since the elaboration of this thesis and publication of our results,

a large multicentric trial aiming for prevention of long-term complications in

T1DM adolescents, the Adolescent T1DM Cardio-Renal Intervention Trial

(AdDIT) is under development coordinated primarily by Prof Dunger in the

United Kingdom (Cambridge). This study involved several groups from

United Kingdom, Australia and Canada. In Australia, Western Australia has

become the National Coordinator Centre. In this study, a large cohort of

adolescents at high-risk for DN, based on AER in the upper tertile, will

receive intervention with ACEI, statins or a combination of both and will be

compared with the use of placebo.

Angiotenisn II receptor blocker (ARB)

The ARBs are selective inhibitors of the angiotensin II type I receptor. These

drugs have not been used widely until now. However, studies from in adults

Page 147: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

126

with T1DM and T2DM suggest that this may be a valuable new drug in

reducing the risk for DN and other diabetes complications (366, 367).

Up to now, there has been no clinical trial in the paediatric population in

order to exploit the pharmacological profile of this drug in this age group.

77..33..11..33.. TThhee uussee ooff lliippiidd aaggeennttss

Although hyperlipidaemia is commonly reported in the paediatric T1DM

population (226) there is no longitudinal study using lipid lowering agents in

this population. Based on a 10-year incidence data, the Pittsburgh

Epidemiology of Diabetes Complications Study suggests a vigorous control

of lipids and BP in terms of reducing mortality and CVD (226). Treatment

with lipid lowering agents, statins, has been increasingly used in adults in

order to reduce vascular events (250).

The AdDit study, as previously mentioned, is the first long-term randomized

control trial in the paediatric population addressing the effects of statins in

preventing microvascular complications, mainly DN, in the adolescents

group.

77..33..22 GGeenneettiicc SSccrreeeenniinngg ffoorr DDNN

The pathogenesis of DN involves an interplay between environmental

factors and an individual genetic predisposition (mono or polygenic factors),

which determines a phenotypic expression. No particular mutation has been

identified, which could explain the development of DN in the majority of

patients; the use of genetic markers as a population screening in assessing

the risk of DN is somehow limited.

However, the discovery of susceptibility genes for DN identifiable at early

stages in the course of the disease in childhood implicates on the

development of novel modalities of prevention and treatment of this

complication. For example, several studies have reported that genetic

variants at the ACE gene seem to modulate the response to the use of ACEI

Page 148: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

127

in DN (166). By having a better understanding of the genetic profile in

relation to the development of DN may be useful in selecting different

antihypertensive agents for patients with different genetic profile.

In addition, the future development of better technologies, such as the

genomic and proteinomic approaches, will possibly unravel the genetics of

DN, identify new mechanisms of renal disease and generate target

molecules for prevention and treatment of DN.

In summary, we have studied clinical and genetic markers for DN in children

and adolescents with T1DM. Based on these findings and other studies,

future protocols could be developed to prevent incipient DN including:

Longitudinal Intervention trials comparing the use of ACEI and ARB in

the progression of DN in childhood;

Longitudinal studies assessing the long-term effects of ACEI and the

use of lipid agents in the development of DN (currently under

development, the AdDIT study);

The use of genetic markers of RAS in assessing not only the risk for

DN but also the response to different antihypertensive agents used in

the adolescent group.

Page 149: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

128

RReeffeerreenncceess

Page 150: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

129

RREEFFEERREENNCCEESS

1. Molitch ME, DeFronzo RA, Franz MJ, Keane WF, Mogensen CE, Parving HH, et al. Nephropathy in diabetes. Diabetes Care. 2004 Jan;27 Suppl 1:S79-83.

2. ANZDATA Registry. Australian and New Zealand Dialysis and Transplant Registry. 1998.

3. Andersen AR, Christiansen JS, Andersen JK, Kreiner S, Deckert T. Diabetic nephropathy in Type 1 diabetes: an epidemiological study. Diabetologia. 1983;25(6):496-501.

4. Borch-Johnsen K, Andersen PK, Deckert T. The effect of proteinuria on relative mortality in type 1 diabetes mellitus. Diabetologia. 1985;28(8):590-6.

5. Mathiesen ER, Ronn B, Storm B, Foght H, Deckert T. The natural course of microalbuminuria in insulin-dependent diabetes: a 10-year prospective study. Diabetic Medicine. 1995;12(6):482-7.

6. Mogensen CE, Christensen CK. Predicting diabetic nephropathy in insulin-dependent patients. New England Journal of Medicine. 1984;311(2):89-93.

7. Parving HH, Oxenboll B, Svendsen PA, Christiansen JS, Andersen AR. Early detection of patients at risk of developing diabetic nephropathy. A longitudinal study of urinary albumin excretion. Acta Endocrinologica. 1982;100(4):550-5.

8. Bognetti E, Calori G, Meschi F, Macellaro P, Bonfanti R, Chiumello G. Prevalence and correlations of early microvascular complications in young type I diabetic patients: role of puberty. Journal of Pediatric Endocrinology & Metabolism. 1997;10(6):587-92.

9. Dahlquist G, Rudberg S. The prevalence of microalbuminuria in diabetic children and adolescents and its relation to puberty. Acta Paediatrica Scandinavica. 1987;76(5):795-800.

10. Jones CA, Leese GP, Kerr S, Bestwick K, Isherwood DI, Vora JP, et al. Development and progression of microalbuminuria in a clinic sample of patients with insulin dependent diabetes mellitus. Archives of Disease in Childhood. 1998;78(6):518-23.

11. Moore TH, Shield JP. Prevalence of abnormal urinary albumin excretion in adolescents and children with insulin dependent diabetes: the MIDAC study. Microalbinuria in Diabetic Adolescents and Children (MIDAC) research group. Archives of Disease in Childhood. 2000;83(3):239-43.

12. Mortensen HB, Marinelli K, Norgaard K, Main K, Kastrup KW, Ibsen KK, et al. A nation-wide cross-sectional study of urinary albumin excretion rate, arterial blood pressure and blood glucose control in Danish children with type 1 diabetes mellitus. Danish Study Group of Diabetes in Childhood. Diabetic Medicine. 1990;7(10):887-97.

Page 151: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

130

13. Olsen BS, Johannesen J, Sjolie AK, Borch-Johnsen K, Hougarrdss P, Thorsteinsson B, et al. Metabolic control and prevalence of microvascular complications in young Danish patients with Type 1 diabetes mellitus. Danish Study Group of Diabetes in Childhood. Diabetic Medicine. 1999;16(1):79-85.

14. Northon D. Relationship between metabolic control and long-term complications of diabetes. Joslin's Diabetes Mellitus 14th edition. 2005 2005:809-20.

15. World Health Organization. Definition, Diagnosis and Classification of Diabetes Mellitus and its Complications. . Report of a WHO Consultation 1999;Part 1: Diagnosis and Classification.(WHO/NCD/NCS/99.2):Geneva, World Health Organization.

16. Onkamo P, Väänänen S, Karvonen M, Tuomilehto J. Worldwide increase in incidence of Type I diabetes – the analysis of the data on published incidence trends. Diabetologia. 1999;42(12):1395-403.

17. Craig ME, Hattersley A, Donaghue KC. Definition, epidemiology and classification of diabetes in children and adolescents. Pediatric Diabetes. 2009;10(s12):3-12.

18. IDF. International Diabetes Federation (IDF) World Atlas of Diabetes 2006:www.eatlas.idf.org.

19. Australian Institute of Health Wefare. http://www.aihw.gov.au/publications/. 2008.

20. Dahlquist GG, Nystrom L, Patterson CC. Incidence of type 1 diabetes in Sweden among individuals aged 0-34 years, 1983-2007: an analysis of time trends. Diabetes Care. 2011 Aug;34(8):1754-9.

21. Gyurus EK, Patterson C, Soltesz G. Twenty-one years of prospective incidence of childhood type 1 diabetes in Hungary - the rising trend continues (or peaks and highlands?). Pediatr Diabetes. 2011 Nov 8.

22. Craig ME, oward NJ, Silink M, Chan A. The rising incidence of childhood type 1 diabetes in New South Wales, Australia. J Pediatr Endocrinol Metab. 2000;13:363 - 72.

23. Haynes A, Bower C, Bulsara MK, Jones TW, Davis EA. Continued increase in the incidence of childhood Type 1 diabetes in a population-based Australian sample (1985-2002). Diabetologia. 2004;47(5):866-70.

24. Chong JW, Craig ME, Cameron FJ, Clarke CF, Rodda CP, Donath SM, et al. Marked increase in type 1 diabetes mellitus incidence in children aged 0 to 14 yr in Victoria, Australia, from 1999 to 2002. Pediatric Diabetes. 2007;8(2):67-73.

25. Haller MJ, Samyn M, Nichols WW, Brusko T, Wasserfall C, Schwartz RF, et al. Radial artery tonometry demonstrates arterial stiffness in children with type 1 diabetes. Diabetes Care. 2004 Dec;27(12):2911-7.

26. Soedamah-Muthu S, Fuller J, Mulnier H, Raleigh V, Lawrenson R, Colhoun H. All-cause mortality rates in patients with type 1 diabetes mellitus compared with a non-diabetic population from the UK general practice research database, 1992–1999. Diabetologia. 2006;49(4):660-6.

Page 152: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

131

27. Laing SP, Swerdlow AJ, Slater SD, Burden AC, Morris A, Waugh NR, et al. Mortality from heart disease in a cohort of 23,000 patients with insulin-treated diabetes. Diabetologia. 2003;46(6):760-5.

28. Soedamah-Muthu SS, Chaturvedi N, Toeller M, Ferriss B, Reboldi P, Michel G, et al. Risk Factors for Coronary Heart Disease in Type 1 Diabetic Patients in Europe: The EURODIAB Prospective Complications Study Diabetes Care. 2004 February 1, 2004;27(2):530-7.

29. Pambianco G, Costacou T, Ellis D, Becker DJ, Klein R, Orchard TJ. The 30-Year Natural History of Type 1 Diabetes Complications. Diabetes. 2006 May 2006;55(5):1463-9.

30. Soedamah-Muthu SS, Chaturvedi N, Witte DR, Stevens LK, Porta M, Fuller JH. Relationship Between Risk Factors and Mortality in Type 1 Diabetic Patients in Europe. Diabetes Care. 2008 July 2008;31(7):1360-6.

31. Larsen J, Brekke M, Sandvik L, Arnesen H, Hanssen KF, Dahl-Jorgensen K. Silent Coronary Atheromatosis in Type 1 Diabetic Patients and Its Relation to Long-Term Glycemic Control. Diabetes. 2002 August 2002;51(8):2637-41.

32. Jenkins AJ, Lyons TJ, Zheng D, Otvos JD, Lackland DT, McGee D, et al. Lipoproteins in the DCCT/EDIC cohort: associations with diabetic nephropathy. Kidney International. 2003;64(3):817-28.

33. Jenkins A, Lyons T, Zheng D, Otvos J, Lackland D, McGee D, et al. Serum Lipoproteins in the Diabetes Control and Complications Trial/Epidemiology of Diabetes Intervention and Complications Cohort: Associations with gender and glycemia. Diabetes Care March. 2003;26(3):810-8.

34. Klein R, Klein BE, Moss SE, Davis MD, DeMets DL. The Wisconsin epidemiologic study of diabetic retinopathy. II. Prevalence and risk of diabetic retinopathy when age at diagnosis is less than 30 years. Arch Ophthalmol. 1984 Apr;102(4):520-6.

35. Nordwall M, Bojestig M, Arnqvist HJ, Ludvigsson J, Linkoping Diabetes Complications S. Declining incidence of severe retinopathy and persisting decrease of nephropathy in an unselected population of Type 1 diabetes-the Linkoping Diabetes Complications Study. Diabetologia. 2004 Jul;47(7):1266-72.

36. Donaghue KC, Craig ME, Chan AK, Fairchild JM, Cusumano JM, Verge CF, et al. Prevalence of diabetes complications 6 years after diagnosis in an incident cohort of childhood diabetes. Diabetic Medicine. 2005 Jun;22(6):711-8.

37. Nordwall M, Hyllienmark L, Ludvigsson J. Early diabetic complications in a population of young patients with type 1 diabetes mellitus despite intensive treatment. Journal of Pediatric Endocrinology. 2006 Jan;19(1):45-54.

38. Rassam SM, Patel V, Kohner EM. The effect of experimental hypertension on retinal vascular autoregulation in humans: a mechanism for the progression of diabetic retinopathy. Experimental Physiology. 1995 Jan;80(1):53-68.

Page 153: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

132

39. DCCT. Effect of intensive diabetes treatment on the development and progression of long-term complications in adolescents with insulin-dependent diabetes mellitus: Diabetes Control and Complications Trial. Diabetes Control and Complications Trial Research Group. Journal of Pediatrics. 1994;125(2):177-88.

40. Writing Team for the DCCT and EDIC Research Group. Effect of intensive therapy on the microvascular complications of type 1 diabetes mellitus. Jama. 2002;287(19):2563-9.

41. DCCT. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group.[comment]. New England Journal of Medicine. 1993;329(14):977-86.

42. Stratton IM, Kohner EM, Aldington SJ, Turner RC, Holman RR, Manley SE, et al. UKPDS 50: risk factors for incidence and progression of retinopathy in Type II diabetes over 6 years from diagnosis. Diabetologia. 2001 Feb;44(2):156-63.

43. Klein R, Klein BE, Moss SE, Cruickshanks KJ. The Wisconsin Epidemiologic Study of Diabetic Retinopathy: XVII. The 14-year incidence and progression of diabetic retinopathy and associated risk factors in type 1 diabetes.[see comment]. Ophthalmology. 1998 Oct;105(10):1801-15.

44. Danne T, Kordonouri O, Enders I, Havener G, Weber B. Factors Modifying the Effect of Hyperglycemia on the Development of Retinopathy in Adolescents with Diabetes. Hormone Research in Paediatrics. 1998;50(Suppl. 1):28-32.

45. Tesfaye S, Selvarajah D. The Eurodiab study: What has this taught us about diabetic peripheral neuropathy? Current Diabetes Reports. 2009;9(6):432-4.

46. Martin CL, Albers J, Herman WH, Cleary P, Waberski B, Greene DA, et al. Neuropathy Among the Diabetes Control and Complications Trial Cohort 8 Years After Trial Completion. Diabetes Care. 2006 February 2006;29(2):340-4.

47. Trotta D, Verrotti A, Salladini C, Chiarelli F. Diabetic neuropathy in children and adolescents. Pediatric Diabetes. 2004;5(1):44-57.

48. Hyllienmark L, Brismar T, Ludvigsson J. Subclinical nerve dysfunction in children and adolescents with IDDM. Diabetologia. 1995;38(6):685-92.

49. Mogensen CE, Chachati A, Christensen CK, Close CF, Deckert T, Hommel E, et al. Microalbuminuria: an early marker of renal involvement in diabetes. Uremia Investigation. 1985;9(2):85-95.

50. Mogensen CE, Keane WF, Bennett PH, Jerums G, Parving HH, Passa P, et al. Prevention of diabetic renal disease with special reference to microalbuminuria.[see comment]. Lancet. 1995 Oct 21;346(8982):1080-4.

51. Viberti GC, Jarrett RJ, Keen H. Microalbuminuria as prediction of nephropathy in diabetics. Lancet. 1982;2(8298):11.

Page 154: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

133

52. Borch-Johnsen K, Kreiner S. Proteinuria: value as predictor of cardiovascular mortality in insulin dependent diabetes mellitus. British Medical Journal Clinical Research Ed. 1987;294(6588):1651-4.

53. Chase HP, Marshall G, Garg SK, Harris S, Osberg I. Borderline increases in albumin excretion rate and the relation to glycemic control in subjects with type I diabetes. Clinical Chemistry. 1991;37(12):2048-52.

54. Couper JJ, Clarke CF, Byrne GC, Jones TW, Donaghue KC, Nairn J, et al. Progression of borderline increases in albuminuria in adolescents with insulin-dependent diabetes mellitus. Diabetic Medicine. 1997;14(9):766-71.

55. Amin R, Turner C, van Aken S, Konopelska Bahu K, Watts A, Lindsell DRM, et al. The relationship between microalbuminuria and glomerular filtration rate in young type 1 diabetic subjects: The Oxford Regional Prospective Study. Kidney Int. 2005;68(4):1740-9.

56. Mogensen CE, Christensen CK, Vittinghus E. The stages in diabetic renal disease. With emphasis on the stage of incipient diabetic nephropathy. Diabetes. 1983 May;32 Suppl 2:64-78.

57. ADA. Diabetic Nephropathy. Diabetes Care. 2002 January 2002;25(suppl 1):s85-s9.

58. Christiansen JS, Gammelgaard J, Tronier B, Svendsen PA, Parving HH. Kidney function and size in diabetics before and during initial insulin treatment. Kidney International. 1982 May;21(5):683-8.

59. Bognetti E, Zoja A, Meschi F, Paesano PL, Chiumello G. Relationship between kidney volume, microalbuminuria and duration of diabetes mellitus. Diabetologia. 1996;39(11).

60. Dahlquist G, Stattin EL, Rudberg S. Urinary albumin excretion rate and glomerular filtration rate in the prediction of diabetic nephropathy; a long-term follow-up study of childhood onset type-1 diabetic patients. Nephrology Dialysis Transplantation. 2001;16(7):1382-6.

61. Lawson ML, Sochett EB, Chait PG, Balfe JW, Daneman D. Effect of puberty on markers of glomerular hypertrophy and hypertension in IDDM. Diabetes. 1996;45(1):51-5.

62. Mogensen CE. Early glomerular hyperfiltration in insulin-dependent diabetics and late nephropathy. Scandinavian Journal of Clinical & Laboratory Investigation. 1986 May;46(3):201-6.

63. Fine L. The biology of renal hypertrophy. Kidney International. 1986 Mar;29(3):619-34.

64. Hostetter TH. Hypertrophy and hyperfunction of the diabetic kidney.[see comment]. Journal of Clinical Investigation. 2001 Jan;107(2):161-2.

65. O'Bryan GT, Hostetter TH. The renal hemodynamic basis of diabetic nephropathy. Seminars in Nephrology. 1997 Mar;17(2):93-100.

66. Vallon V, Blantz RC, Thomson S. Glomerular hyperfiltration and the salt paradox in early type 1 diabetes mellitus: a tubulo-centric view. Journal of the American Society of Nephrology. 2003 Feb;14(2):530-7.

Page 155: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

134

67. Wolf G, Ziyadeh FN. Molecular mechanisms of diabetic renal hypertrophy. Kidney International. 1999 Aug;56(2):393-405.

68. Zerbini G, Bonfanti R, Meschi F, Bognetti E, Paesano PL, Gianolli L, et al. Persistent renal hypertrophy and faster decline of glomerular filtration rate precede the development of microalbuminuria in type 1 diabetes. Diabetes. 2006 Sep;55(9):2620-5.

69. Osterby R, Gundersen HJ, Horlyck A, Kroustrup JP, Nyberg G, Westberg G. Diabetic glomerulopathy. Structural characteristics of the early and advanced stages. Diabetes. 1983 May;32 Suppl 2:79-82.

70. Drummond K, Mauer M. The Early Natural History of Nephropathy in Type 1 Diabetes: II. Early Renal Structural Changes in Type 1 Diabetes Diabetes. 2002 May 1, 2002;51(5):1580-7.

71. Feldt-Rasmussen B, Norgaard K, Jensen T, Deckert T. Microalbuminuria, clinical nephropathy and hypertension in diabetes. Journal of Human Hypertension. 1991;5(4):255-63.

72. Gatling W, Mullee MA, Knight C, Hill RD. Microalbuminuria in diabetes: relationships between urinary albumin excretion and diabetes-related variables. Diabetic Medicine. 1988;5(4):348-51.

73. Hovind P, Tarnow L, Rossing P, Jensen BR, Graae M, Torp I, et al. Predictors for the development of microalbuminuria and macroalbuminuria in patients with type 1 diabetes: inception cohort study. BMJ. 2004;328(7448):8.

74. Koulouridis E. Diabetic nephropathy in children and adolescents and its consequences in adults. Journal of Pediatric Endocrinology & Metabolism. 2001;5:1367-77.

75. Mogensen CE. Microalbuminuria, blood pressure and diabetic renal disease: origin and development of ideas. Diabetologia. 1999;42(3):263-85.

76. Parving HH, Mogensen CE, Jensen HA, Evrin PE. Increased urinary albumin-excretion rate in benign essential hypertension. Lancet. 1974 Jun 15;1(7868):1190-2.

77. Mogensen CE, Poulsen PL. Microalbuminuria, glycemic control, and blood pressure predicting outcome in diabetes type 1 and type 2. Kidney International - Supplement. 2004 Nov(92):S40-1.

78. Janssen WM, de Jong PE, de Zeeuw D. Hypertension and renal disease: role of microalbuminuria. Journal of Hypertension Supplement. 1996;14(5).

79. Wachtell K, Ibsen H, Olsen MH, Borch-Johnsen K, Lindholm LH, Mogensen CE, et al. Albuminuria and cardiovascular risk in hypertensive patients with left ventricular hypertrophy: the LIFE study. Annals of Internal Medicine. 2003 Dec 2;139(11):901-6.

80. Mogensen CE, Hansen KW, Sommer S, Klebe J, Christensen CK, Marshall S, et al. Microalbuminuria: studies in diabetes, essential hypertension, and renal diseases as compared with the background population. Advances in Nephrology From the Necker Hospital. 1991;20:191-228.

Page 156: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

135

81. Mauer M, Zinman B, Gardiner R, Suissa S, Sinaiko A, Strand T, et al. Renal and retinal effects of enalapril and losartan in type 1 diabetes. N Engl J Med. 2009 Jul 2;361(1):40-51.

82. Shlipak M. Diabetic nephropathy: preventing progression. Clin Evid (Online). Epub date Jan, 2010.

83. Salardi S, Balsamo C, Zucchini S, Maltoni G, Scipione M, Rollo A, et al. High rate of regression from micro-macroalbuminuria to normoalbuminuria in children and adolescents with type 1 diabetes treated or not with enalapril: the influence of HDL cholesterol. Diabetes Care. 2011 Feb;34(2):424-9.

84. Vejakama P, Thakkinstian A, Lertrattananon D, Ingsathit A, Ngarmukos C, Attia J. Reno-protective effects of renin-angiotensin system blockade in type 2 diabetic patients: a systematic review and network meta-analysis. Diabetologia. 2012 Mar;55(3):566-78.

85. Adolescent type 1 Diabetes Cardio-renal Intervention Trial Research Group. Adolescent type 1 Diabetes Cardio-renal Intervention Trial (AdDIT). BMC Pediatr. 2009;9:79.

86. Cooper ME. Pathogenesis, prevention, and treatment of diabetic nephropathy. Lancet. 1998 Jul 18;352(9123):213-9.

87. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature, 2001; 414(6865):813-20.

88. Soulis T, Thallas V, Youssef S, Gilbert RE, McWilliam BG, Murray-McIntosh RP, et al. Advanced glycation end products and their receptors co-localise in rat organs susceptible to diabetic microvascular injury. Diabetologia. 1997 Jun;40(6):619-28.

89. Monnier VM, Sell DR, Genuth S. Glycation products as markers and predictors of the progression of diabetic complications. Ann N Y Acad Sci. 2005 Jun;1043:567-81.

90. Beisswenger PJ. Glycation and biomarkers of vascular complications of diabetes. Amino Acids. 2010 Nov 2.

91. Asano M, Fujita Y, Ueda Y, Suzuki D, Miyata T, Sakai H, et al. Renal proximal tubular metabolism of protein-linked pentosidine, an advanced glycation end product. Nephron. 2002 Aug;91(4):688-94.

92. He Z, King GL. Microvascular Complications of Diabetes. Endocrinology and Metabolism Clinics of North America. 2004;33(March):215 - 38.

93. Davis BJ, Forbes JM, Thomas MC, Jerums G, Burns WC, Kawachi H, et al. Superior renoprotective effects of combination therapy with ACE and AGE inhibition in the diabetic spontaneously hypertensive rat. Diabetologia. 2004 Jan;47(1):89-97.

94. Sheetz MJ, King GL. Molecular understanding of hyperglycemia's adverse effects for diabetic complications. Jama. 2002 Nov 27;288(20):2579-88.

95. Gugliucci A. Glycation as the glucose link to diabetic complications. J Am Osteopath Assoc. 2000 October 1, 2000;100(10):621-34.

Page 157: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

136

96. Cumbie BC, Hermayer KL. Current concepts in targeted therapies for the pathophysiology of diabetic microvascular complications. Vasc Health Risk Manag. 2007;3(6):823-32.

97. Sorbinil Retinopathy Trial Research Group. A randomized trial of sorbinil, an aldose reductase inhibitor, in diabetic retinopathy. Sorbinil Retinopathy Trial Research Group. Arch Ophthalmol. 1990 Sep;108(9):1234-44.

98. Sorbinil Retinopathy Trial Research Group. The sorbinil retinopathy trial: neuropathy results. Sorbinil Retinopathy Trial Research Group. Neurology. 1993 Jun;43(6):1141-9.

99. Schemmel KE, Padiyara RS, D'Souza JJ. Aldose reductase inhibitors in the treatment of diabetic peripheral neuropathy: a review. J Diabetes Complications. 2010 Sep-Oct;24(5):354-60.

100. Nagpala PG, Malik AB, Vuong PT, Lum H. Protein kinase C beta 1 overexpression augments phorbol ester-induced increase in endothelial permeability. J Cell Physiol. 1996 Feb;166(2):249-55.

101. Nonaka A, Kiryu J, Tsujikawa A, Yamashiro K, Miyamoto K, Nishiwaki H, et al. PKC-beta inhibitor (LY333531) attenuates leukocyte entrapment in retinal microcirculation of diabetic rats. Invest Ophthalmol Vis Sci. 2000 Aug;41(9):2702-6.

102. Kelly DJ, Zhang Y, Hepper C, Gow RM, Jaworski K, Kemp BE, et al. Protein Kinase C β Inhibition Attenuates the Progression of Experimental Diabetic Nephropathy in the Presence of Continued Hypertension. Diabetes. 2003 February 1, 2003;52(2):512-8.

103. Aiello LP, Davis MD, Girach A, Kles KA, Milton RC, Sheetz MJ, et al. Effect of ruboxistaurin on visual loss in patients with diabetic retinopathy. Ophthalmology. 2006 Dec;113(12):2221-30.

104. Tuttle KR, Bakris GL, Toto RD, McGill JB, Hu K, Anderson PW. The effect of ruboxistaurin on nephropathy in type 2 diabetes. Diabetes Care. 2005 Nov;28(11):2686-90.

105. Chiarelli F, Spagnoli A, Basciani F, Tumini S, Mezzetti A, Cipollone F, et al. Vascular endothelial growth factor (VEGF) in children, adolescents and young adults with Type 1 diabetes mellitus: relation to glycaemic control and microvascular complications. Diabetic Medicine. 2000;17(9):650-6.

106. Ray D, Mishra M, Ralph S, Read I, Davies R, Brenchley P. Association of the VEGF gene with proliferative diabetic retinopathy but not proteinuria in diabetes. Diabetes. 2004 Mar;53(3):861-4.

107. Raile K, Galler A, Hofer S, Herbst A, Dunstheimer D, Busch P, et al. Diabetic nephropathy in 27,805 children, adolescents, and adults with type 1 diabetes: effect of diabetes duration, A1C, hypertension, dyslipidemia, diabetes onset, and sex. Diabetes Care. 2007 Oct;30(10):2523-8.

108. Giunti S, Barit D, Cooper ME. Mechanisms of Diabetic Nephropathy: Role of Hypertension. Hypertension. 2006 October 1, 2006;48(4):519-26.

109. UK Prospective Diabetes Study Group. Tight blood pressure control and risk of macrovascular and microvascular complications in type 2

Page 158: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

137

diabetes: UKPDS 38. UK Prospective Diabetes Study Group. Bmj. 1998 Sep 12;317(7160):703-13.

110. DCCT. Clustering of long-term complications in families with diabetes in the diabetes control and complications trial. The Diabetes Control and Complications Trial Research Group. Diabetes 1997;46(11):1829.

111. Chowdhury TA, Dyer PH, Kumar S, Barnett AH, Bain SC. Genetic determinants of diabetic nephropathy. Clinical Science. 1999;96(3):221-30.

112. Schultz CJ, Dalton RN, Selwood M, Dunger DB, Neil HAW. Paternal phenotype is associated with microalbuminuria in young adults with Type 1 diabetes mellitus of short duration. Diabetic Medicine. 2004;21(3):246-51.

113. Guntsche Z, Saravi FD, Reynals EA, Rauek B, Rauek M, Guntsche EM. Parental hypertension and 24 h-blood pressure in children prior to diabetic nephropathy. Pediatric Nephrology. 2002;17(3):157-64.

114. Heart Outcomes Prevention Evaluation (HOPE) Study Investigators. Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: results of the HOPE study and MICRO-HOPE substudy. Heart Outcomes Prevention Evaluation Study Investigators.Lancet. 2000;355(9200):253-9.

115. Dalla Vestra M, Simioni N, Masiero A. Renal effects of dual renin-angiotensin-aldosterone system blockade in patients with diabetic nephropathy. Int Urol Nephrol. 2009;41(1):119-26.

116. Mogensen CE. ACE inhibitors and antihypertensive treatment in diabetes: focus on microalbuminuria and macrovascular disease. J Renin Angiotensin Aldosterone Syst. 2000 Sep;1(3):234-9.

117. Robinson DM, Curran MP, Lyseng-Williamson KA. Imidapril: a review of its use in essential hypertension, Type 1 diabetic nephropathy and chronic heart failure. Drugs. 2007;67(9):1359-78.

118. Schjoedt KJ, Hansen HP, Tarnow L, Rossing P, Parving HH. Long-term prevention of diabetic nephropathy: an audit. Diabetologia. 2008 Jun;51(6):956-61.

119. Thomas MC, Atkins RC. Blood pressure lowering for the prevention and treatment of diabetic kidney disease. Drugs. 2006;66(17):2213-34.

120. Mogensen CE. Microalbuminuria and hypertension with focus on type 1 and type 2 diabetes. Journal of Internal Medicine. 2003;254(1):45-66.

121. The ACE Inhibitors in Diabetic Nephropathy Trialist Group. Should All Patients with Type 1 Diabetes Mellitus and Microalbuminuria Receive Angiotensin-Converting Enzyme Inhibitors? Annals of Internal Medicine. 2001 March 6, 2001;134(5):370-9.

122. Hostetter TH, Rennke HG, Brenner BM. The case for intrarenal hypertension in the initiation and progression of diabetic and other glomerulopathies. Am J Med. 1982 Mar;72(3):375-80.

123. Bank N. Mechanisms of diabetic hyperfiltration. Kidney Int. 1991 Oct;40(4):792-807.

Page 159: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

138

124. Zatz R, Meyer TW, Rennke HG, Brenner BM. Predominance of hemodynamic rather than metabolic factors in the pathogenesis of diabetic glomerulopathy. Proc Natl Acad Sci U S A. 1985 Sep;82(17):5963-7.

125. Keller CK, Bergis KH, Fliser D, Ritz E. Renal findings in patients with short-term type 2 diabetes. J Am Soc Nephrol. 1996 Dec;7(12):2627-35.

126. Levine DZ. Can rodent models of diabetic kidney disease clarify the significance of early hyperfiltration?: recognizing clinical and experimental uncertainties. Clin Sci (Lond). 2008 Jan;114(2):109-18.

127. Vora JP, Dolben J, Dean JD, Thomas D, Williams JD, Owens DR, et al. Renal hemodynamics in newly presenting non-insulin dependent diabetes mellitus. Kidney Int. 1992 Apr;41(4):829-35.

128. Hirschberg R, Brunori G, Kopple JD, Guler HP. Effects of insulin-like growth factor I on renal function in normal men. Kidney Int. 1993 Feb;43(2):387-97.

129. Sabbatini M, Sansone G, Uccello F, Giliberti A, Conte G, Andreucci VE. Early glycosylation products induce glomerular hyperfiltration in normal rats. Kidney Int. 1992 Oct;42(4):875-81.

130. Magee GM, Bilous RW, Cardwell CR, Hunter SJ, Kee F, Fogarty DG. Is hyperfiltration associated with the future risk of developing diabetic nephropathy? A meta-analysis. Diabetologia. 2009 Apr;52(4):691-7.

131. Hostetter TH, Rennke HG, Brenner BM. The case for intrarenal hypertension in the initiation and progression of diabetic and other glomerulopathies. The American Journal of Medicine. 1982;72(3):375-80.

132. Vervoort G, Veldman B, Berden JH, Smits P, Wetzels JF. Glomerular hyperfiltration in type 1 diabetes mellitus results from primary changes in proximal tubular sodium handling without changes in volume expansion. Eur J Clin Invest. 2005 May;35(5):330-6.

133. Ficociello LH, Perkins BA, Roshan B, Weinberg JM, Aschengrau A, Warram JH, et al. Renal hyperfiltration and the development of microalbuminuria in type 1 diabetes. Diabetes Care. 2009 May;32(5):889-93.

134. Helal I, Fick-Brosnahan GM, Reed-Gitomer B, Schrier RW. Glomerular hyperfiltration: definitions, mechanisms and clinical implications. Nat Rev Nephrol. 2012 Feb 21.

135. van der Meer IM, Cravedi P, Remuzzi G. The role of renin angiotensin system inhibition in kidney repair. Fibrogenesis Tissue Repair. 2010;3:7.

136. Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, et al. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med. 2001 Sep 20;345(12):861-9.

137. Lewis EJ, Hunsicker LG, Bain RP, Rohde RD. The effect of angiotensin-converting-enzyme inhibition on diabetic nephropathy. The Collaborative Study Group. N Engl J Med. 1993 Nov 11;329(20):1456-62.

138. Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, et al. Renoprotective effect of the angiotensin-receptor antagonist irbesartan in

Page 160: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

139

patients with nephropathy due to type 2 diabetes. N Engl J Med. 2001 Sep 20;345(12):851-60.

139. Parving HH, Lehnert H, Brochner-Mortensen J, Gomis R, Andersen S, Arner P. The effect of irbesartan on the development of diabetic nephropathy in patients with type 2 diabetes. N Engl J Med. 2001 Sep 20;345(12):870-8.

140. Ruggenenti P, Fassi A, Ilieva AP, Bruno S, Iliev IP, Brusegan V, et al. Preventing microalbuminuria in type 2 diabetes. N Engl J Med. 2004 Nov 4;351(19):1941-51.

141. Nobakht N, Kamgar M, Rastogi A, Schrier RW. Limitations of angiotensin inhibition. Nat Rev Nephrol. 2011 Jun;7(6):356-9.

142. Nordwall M, Bojestig M, Arnqvist HJ, Ludvigsson J, Linkoping Diabetes Complications Study. Declining incidence of severe retinopathy and persisting decrease of nephropathy in an unselected population of Type 1 diabetes-the Linkoping Diabetes Complications Study. Diabetologia. 2004;47(7):1266-72.

143. Seaquist ER, Goetz FC, Rich S, Barbosa J. Familial clustering of diabetic kidney disease. Evidence for genetic susceptibility to diabetic nephropathy.[comment]. New England Journal of Medicine. 1989;320(18):1161-5.

144. Borch-Johnsen K, Norgaard K, Hommel E, Mathiesen ER, Jensen JS, Deckert T, et al. Is diabetic nephropathy an inherited complication? Kidney International. 1992;41(4):719-22.

145. Miller JA, Scholey JW. The impact of renin-angiotensin system polymorphisms on physiological and pathophysiological processes in humans. Current Opinion in Nephrology & Hypertension. 2004;13(1):101-6.

146. Cooper ME, Bonnet F, Oldfield M, Jandeleit-Dahm K. Mechanisms of diabetic vasculopathy: an overview. Am J Hypertens. 2001 May;14(5 Pt 1):475-86.

147. Kagami S, Border WA, Miller DE, Noble NA. Angiotensin II stimulates extracellular matrix protein synthesis through induction of transforming growth factor-beta expression in rat glomerular mesangial cells. J Clin Invest. 1994 Jun;93(6):2431-7.

148. Jaimes EA, Galceran JM, Raij L. Angiotensin II induces superoxide anion production by mesangial cells. Kidney Int. 1998 Sep;54(3):775-84.

149. Bain SC, Chowdhury TA. Genetics of diabetic nephropathy and microalbuminuria. Journal of the Royal Society of Medicine. 2000;93(2):62-6.

150. Merta M, Reiterova J, Rysava R, Kmentova D, Tesar V. Genetics of diabetic nephropathy. Nephrology Dialysis Transplantation. 2003 Jul;18 Suppl 5:v24-5.

151. Parving HH, Tarnow L, Rossing P. Genetics of diabetic nephropathy. Journal of the American Society of Nephrology. 1996;7(12):2509-17.

152. Crisan D, Carr J. Angiotensin I-Converting Enzyme - Genotype and Disease Associations. Journal of Molecular Diagnostics. 2000;2(3):105-15.

Page 161: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

140

153. Ohtsu H, Suzuki H, Nakashima H, Dhobale S, Frank GD, Motley ED, et al. Angiotensin II signal transduction through small GTP-binding proteins: mechanism and significance in vascular smooth muscle cells. Hypertension. 2006 Oct;48(4):534-40.

154. Abbud ZA, Wilson AC, Cosgrove NM, Kostis JB. Angiotensin-converting enzyme gene polymorphism in systemic hypertension. American Journal of Cardiology. 1998;81(2):244-6.

155. Bedir A, Arik N, Adam B, Kilinc K, Gumus T, Guner E. Angiotensin converting enzyme gene polymorphism and activity in Turkish patients with essential hypertension. American Journal of Hypertension. 1999;12(1O Pt 1):1038-43.

156. Butler R, Morris AD, Burchell B, Struthers AD. DD angiotensin-converting enzyme gene polymorphism is associated with endothelial dysfunction in normal humans. Hypertension. 1999;33(5):1164-8.

157. Di Pasquale P, Cannizzaro S, Paterna S. Does angiotensin-converting enzyme gene polymorphism affect blood pressure? Findings after 6 years of follow-up in healthy subjects. European Journal of Heart Failure. 2004;6(1):11-6.

158. Huang XH, Rantalaiho V, Wirta O, Pasternack A, Koivula T, Hiltunen TP, et al. Angiotensin-converting enzyme gene polymorphism is associated with coronary heart disease in non-insulin-dependent diabetic patients evaluated for 9 years. Metabolism: Clinical & Experimental. 1998;47(10):1258-62.

159. Teranishi M, Ono H, Ishimitsu T, Matsuoka H. Insertion/deletion angiotensin converting enzyme gene polymorphism affects the microvascular structure of the kidney in patients with nondiabetic renal disease. Journal of Hypertension. 1999;17(3):351-6.

160. Fabris B, Bortoletto M, Candido R, Barbone F, Cattin MR, Calci M, et al. Genetic polymorphisms of the renin-angiotensin-aldosterone system and renal insufficiency in essential hypertension. Journal of Hypertension. 2005;23(2):309-16.

161. Rigat B, Hubert C, Alhenc-Gelas F, Cambien F, Corvol P, Soubrier F. An insertion/deletion polymorphism in the angiotensin I-converting enzyme gene accounting for half the variance of serum enzyme levels. Journal of Clinical Investigation. 1990;86(4):1343-6.

162. Ueda S, Elliott HL, Morton JJ, Connell JM. Enhanced pressor response to angiotensin I in normotensive men with the deletion genotype (DD) for angiotensin-converting enzyme. Hypertension. 1995 Jun;25(6):1266-9.

163. Brown NJ, Blais C, Jr., Gandhi SK, Adam A. ACE insertion/deletion genotype affects bradykinin metabolism. J Cardiovasc Pharmacol. 1998 Sep;32(3):373-7.

164. Rudberg S, Rasmussen LM, Bangstad HJ, Osterby R. Influence of insertion/deletion polymorphism in the ACE-I gene on the progression of diabetic glomerulopathy in type 1 diabetic patients with microalbuminuria. Diabetes Care. 2000 Apr;23(4):544-8.

Page 162: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

141

165. Huang W, Gallois Y, Bouby N, Bruneval P, Heudes D, Belair M-F, et al. Genetically increased angiotensin I-converting enzyme level and renal complications in the diabetic mouse. Proceedings of the National Academy of Sciences of the United States of America. 2001 November 6, 2001;98(23):13330-4.

166. Ng DPK, Tai BC, Koh D, Tan KW, Chia K. Angiotensin-I converting enzyme insertion/deletion polymorphism and its association with diabetic nephropathy: a meta-analysis of studies reported between 1994 and 2004 and comprising 14,727 subjects. Diabetologia. 2005;48:1008-16.

167. Wang F, Fang Q, Yu N, Zhao D, Zhang Y, Wang J, et al. Association between genetic polymorphism of the angiotensin-converting enzyme and diabetic nephropathy: a meta-analysis comprising 26,580 subjects. J Renin Angiotensin Aldosterone Syst. 2012 Mar;13(1):161-74.

168. Hadjadj S, Belloum R, Bouhanick B, Gallois Y, Guilloteau G, Chatellier G, et al. Prognostic value of angiotensin-I converting enzyme I/D polymorphism for nephropathy in type 1 diabetes mellitus: a prospective study. Journal of the American Society of Nephrology. 2001 Mar;12(3):541-9.

169. Boright AP, Paterson AD, Mirea L, Bull SB, Mowjoodi A, Scherer SW, et al. Genetic Variation at the ACE Gene Is Associated With Persistent Microalbuminuria and Severe Nephropathy in Type 1 Diabetes: The DCCT/EDIC Genetics Study. Diabetes April. 2005;54(4):1238-44.

170. Barkai L, Soos A, Vamosi I. Association of angiotensin-converting enzyme DD genotype with 24-h blood pressure abnormalities in normoalbuminuric children and adolescents with Type 1 diabetes. Diabetic Medicine. 2005 Aug;22(8):1054-9.

171. Pavlovic M, Reile M, Haeberle U, Schwarz K, Heinze E, Teller W, et al. Angiotensin I-converting enzyme-gene-polymorphism: relationship to albumin excretion and blood pressure in pediatric patients with type-I-diabetes mellitus. Experimental & Clinical Endocrinology & Diabetes. 1997;105(5):248-53.

172. Anderson PW, Do YS, Hsueh WA. Angiotensin II causes mesangial cell hypertrophy. Hypertension. 1993;21(1):29-35.

173. Jeunemaitre X. Renin-angiotensin-aldosterone system polymorphisms and essential hypertension: where are we? Journal of Hypertension. 2003;21(12):2219-22.

174. Jeunemaitre X, Soubrier F, Kotelevtsev YV, Lifton RP, Williams CS, Charru A, et al. Molecular basis of human hypertension: Role of angiotensinogen. Cell. 1992 1992/10/2;71(1):169-80.

175. Winkelmann BR, Russ AP, Nauck M, Klein B, Bohm BO, Maier V, et al. Angiotensinogen M235T polymorphism is associated with plasma angiotensinogen and cardiovascular disease. American Heart Journal. 1999 1999/4;137(4):698-705.

176. Schmidt S, Sharma AM, Zilch O, Beige J, Walla-Friedel M, Ganten D, et al. Association of M235T variant of the angiotensinogen gene with familial hypertension of early onset. Nephrol Dial Transplant. 1995;10(7):1145-8.

Page 163: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

142

177. Fogarty DG, Harron JC, Hughes AE, Nevin NC, Doherty CC, Maxwell AP. A molecular variant of angiotensinogen is associated with diabetic nephropathy in IDDM. Diabetes. 1996 Sep;45(9):1204-8.

178. Marre M, Jeunemaitre X, Gallois Y, Rodier M, Chatellier G, Sert C, et al. Contribution of genetic polymorphism in the renin-angiotensin system to the development of renal complications in insulin-dependent diabetes: Genetique de la Nephropathie Diabetique (GENEDIAB) study group. Journal of Clinical Investigation. 1997 Apr 1;99(7):1585-95.

179. van Ittersum FJ, de Man AM, Thijssen S, de Knijff P, Slagboom E, Smulders Y, et al. Genetic polymorphisms of the renin-angiotensin system and complications of insulin-dependent diabetes mellitus. Nephrology Dialysis Transplantation. 2000;15(7):1000-7.

180. Freire MB, Ji L, Onuma T, Orban T, Warram JH, Krolewski AS. Gender-specific association of M235T polymorphism in angiotensinogen gene and diabetic nephropathy in NIDDM. Hypertension. 1998 Apr;31(4):896-9.

181. Lovati E, Richard A, Frey BM, Frey FJ, Ferrari P. Genetic polymorphisms of the renin-angiotensin-aldosterone system in end-stage renal disease. Kidney International. 2001 Jul;60(1):46-54.

182. Schmidt S, Giessel R, Bergis KH, Strojek K, Grzeszczak W, Ganten D, et al. Angiotensinogen gene M235T polymorphism is not associated with diabetic nephropathy. Nephrology Dialysis Transplantation. 1996 Sep;11(9):1755-61.

183. Tarnow L, Cambien F, Rossing P, Nielsen FS, Hansen BV, Ricard S, et al. Angiotensinogen gene polymorphisms in IDDM patients with diabetic nephropathy. Diabetes. 1996;45(3):367-9.

184. Chowdhury TA, Dronsfield MJ, Kumar S, Gough SL, Gibson SP, Khatoon A, et al. Examination of two genetic polymorphisms within the renin-angiotensin system: no evidence for an association with nephropathy in IDDM. Diabetologia. 1996;39(9):1108-14.

185. Zychma MJ, Zukowska-Szczechowska E, Lacka BI, Grzeszczak W. Angiotensinogen M235T and chymase gene CMA/B polymorphisms are not associated with nephropathy in type II diabetes. Nephrology Dialysis Transplantation. 2000 Dec;15(12):1965-70.

186. Osawa N, Koya D, Araki S, Uzu T, Tsunoda T, Kashiwagi A, et al. Combinational effect of genes for the renin-angiotensin system in conferring susceptibility to diabetic nephropathy. J Hum Genet. 2007;52(2):143-51.

187. Mollsten A, Kockum I, Svensson M, Rudberg S, Ugarph-Morawski A, Brismar K, et al. The effect of polymorphisms in the renin-angiotensin-aldosterone system on diabetic nephropathy risk. J Diabetes Complications. 2008 Nov-Dec;22(6):377-83.

188. Jacobsen P, Tarnow L, Carstensen B, Hovind P, Poirier O, Parving HH. Genetic variation in the Renin-Angiotensin system and progression of diabetic nephropathy. Journal of the American Society of Nephrology. 2003;14(11):2843-50.

Page 164: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

143

189. Bonnardeaux A, Davies E, Jeunemaitre X, Fery I, Charru A, Clauser E, et al. Angiotensin II type 1 receptor gene polymorphisms in human essential hypertension. Hypertension. 1994 Jul;24(1):63-9.

190. Jin Y, Kuznetsova T, Thijs L, Schmitz B, Liu Y, Asayama K, et al. Association of Left Ventricular Mass With the AGTR1 A1166C Polymorphism. Am J Hypertens. 2012 Jan 12.

191. Niu W, Qi Y. Association of the angiotensin II type I receptor gene +1166 A>C polymorphism with hypertension risk: evidence from a meta-analysis of 16474 subjects. Hypertens Res. 2010 Nov;33(11):1137-43.

192. Fradin S, Goulet-Salmon B, Chantepie M, Grandhomme F, Morello R, Jauzac P, et al. Relationship between polymorphisms in the renin-angiotensin system and nephropathy in type 2 diabetic patients. Diabetes Metab. 2002 Feb;28(1):27-32.

193. Lin J, Hu FB, Qi L, Curhan GC. Genetic polymorphisms of angiotensin-2 type 1 receptor and angiotensinogen and risk of renal dysfunction and coronary heart disease in type 2 diabetes mellitus. BMC Nephrol. 2009;10:9.

194. Fradin S, Goulet-Salmon B, Chantepie M, Grandhomme F, Morello R, Jauzac P, et al. Relationship between polymorphisms in the renin-angiotensin system and nephropathy in type 2 diabetic patients. Diabetes & Metabolism. 2002 Feb;28(1):27-32.

195. Doria A, Onuma T, Warram JH, Krolewski AS. Synergistic effect of angiotensin II type 1 receptor genotype and poor glycaemic control on risk of nephropathy in IDDM. Diabetologia. 1997;40(11):1293-9.

196. Ding W, Wang F, Fang Q, Zhang M, Chen J, Gu Y. Association between two genetic polymorphisms of the renin-angiotensin-aldosterone system and diabetic nephropathy: a meta-analysis. Mol Biol Rep. 2012 Feb;39(2):1293-303.

197. Lajer M, Tarnow L, Fleckner J, Hansen BV, Edwards DG, Parving HH, et al. Association of aldose reductase gene Z+2 polymorphism with reduced susceptibility to diabetic nephropathy in Caucasian Type 1 diabetic patients. Diabetic Medicine. 2004 Aug;21(8):867-73.

198. Kao Y, Donaghue KC, Chan A, Bennetts BH, Knight J, Silink M. Paraoxonase gene cluster is a genetic marker for early microvascular complications in type 1 diabetes. Diabetic Medicine. 2002;19(3):212-5.

199. Araki S, Moczulski DK, Hanna L, Scott LJ, Warram JH, Krolewski AS. APOE polymorphisms and the development of diabetic nephropathy in type 1 diabetes: results of case-control and family-based studies. Diabetes. 2000 Dec;49(12):2190-5.

200. Klein RL, Hunter SJ, Jenkins AJ, Zheng D, Semler AJ, Clore J, et al. Fibrinogen is a marker for nephropathy and peripheral vascular disease in type 1 diabetes: studies of plasma fibrinogen and fibrinogen gene polymorphism in the DCCT/EDIC cohort. Diabetes Care. 2003;26(5):1439-48.

Page 165: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

144

201. Ng DPK, Canani L, Araki S-i, Smiles A, Moczulski D, Warram J, et al. Minor effect of GLUT1 polymorphisms on susceptibility to diabetic nephropathy in type 1 diabetes. Diabetes. 2002 Jul;51(7):2264-9.

202. Canani LH, Ng DPK, Smiles A, Rogus JJ, Warram JH, Krolewski AS. Polymorphism in ecto-nucleotide pyrophosphatase/phosphodiesterase 1 gene (ENPP1/PC-1) and early development of advanced diabetic nephropathy in type 1 diabetes. Diabetes. 2002 Apr;51(4):1188-93.

203. Conway BR, Maxwell AP. Genetics of diabetic nephropathy: are there clues to the understanding of common kidney diseases? Nephron Clin Pract. 2009;112(4):c213-21.

204. Ellis D, Avner ED, Kurs-Lasky M, Richards M, Becker DJ. Effects of improved glycemic control on microalbuminuria in adolescents with insulin-dependent diabetes mellitus. International Journal of Pediatric Nephrology. 1986;7(1):31-8.

205. Microalbuminuria Collaborative Study Group. Intensive therapy and progression to clinical albuminuria in patients with insulin dependent diabetes mellitus and microalbuminuria. Microalbuminuria Collaborative Study Group, United Kingdom. BMJ 1995;311(7011):973-7.

206. DCCT and EDIC Writing Research Group. Sustained effect of intensive treatment of type 1 diabetes mellitus on development and progression of diabetic nephropathy: the Epidemiology of Diabetes Interventions and Complications (EDIC) study. Jama. 2003;290(16):2159-67.

207. Nathan DM, Cleary PA, Backlund JY, Genuth SM, Lachin JM, Orchard TJ, et al. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. New England Journal of Medicine. 2005;353(25):2643-53.

208. Ihnat MA, Thorpe JE, Ceriello A. Hypothesis: the 'metabolic memory', the new challenge of diabetes. Diabetic Medicine. 2007;24(6):582-6.

209. Rudberg S, Ullman E, Dahlquist G. Relationship between early metabolic control and the development of microalbuminuria--a longitudinal study in children with type 1 (insulin-dependent) diabetes mellitus. Diabetologia. 1993;36(12):1309-14.

210. Schultz CJ, Konopelska-Bahu T, Dalton RN, Carroll TA, Stratton I, Gale EA, et al. Microalbuminuria prevalence varies with age, sex, and puberty in children with type 1 diabetes followed from diagnosis in a longitudinal study. Oxford Regional Prospective Study Group. Diabetes Care. 1999;22(3):495-502.

211. Danne T, Kordonouri O, Hovener G, Weber B. Diabetic angiopathy in children. Diabetic Medicine. 1997;14(12):1012-25.

212. Svensson M, Eriksson JW, Dahlquist G. Early Glycaemic Control, Age at Onset, and Development of Microvascular Complications in Childhood-Onset Type 1 diabetes. Diabetes Care. 2004;27(4):955-62.

213. Global International Diabetes Federation (IDF)/International Society for Pediatric and Adolescent Diabetes (ISPAD). Guideline for Diabetes in Childhood and Adolescence. http://wwwidforg/guidelines. 2011.

Page 166: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

145

214. Donaghue KC, Craig ME, Chan AKF, Fairchild JM, Cusumano JM, Verge CF, et al. Prevalence of diabetes complications 6 years after diagnosis in an incident cohort of childhood diabetes. Diabetic Medicine. 2005 Jun 22(6):711-8.

215. Gallego PH, Wiltshire E, Donaghue KC. Identifying children at particular risk of long-term diabetes complications. Pediatric Diabetes. 2007;8:40-8.

216. Donaghue KC, Fung AT, Hing S, Fairchild J, King J, Chan A, et al. The effect of prepubertal diabetes duration on diabetes. Microvascular complications in early and late adolescence. Diabetes Care. 1997 Jan;20(1):77-80.

217. Donaghue KC, Fairchild JM, Craig ME, Chan AK, Hing S, Cutler LR, et al. Do all prepubertal years of diabetes duration contribute equally to diabetes complications?[comment]. Diabetes Care. 2003;26(4):1224-9.

218. Salardi S, Porta M, Maltoni G, Rubbi F, Rovere S, Cerutti F, et al. Infant and Toddler Type 1 Diabetes: Complications after 20 years' duration. Diabetes Care. 2012 Feb 8.

219. Yip J, Mattock MB, Morocutti A, Sethi M, Trevisan R, Viberti G. Insulin resistance in insulin-dependent diabetic patients with microalbuminuria. Lancet. 1993;342(8876):883-7.

220. Schultz CJ, Amin R, Dunger DB. Markers of microvascular complications in insulin dependent diabetes. Archives of Disease in Childhood. 2002;87(1):10-2.

221. Barkai L, Vamosi I, Lukacs K. Enhanced progression of urinary albumin excretion in IDDM during puberty. Diabetes Care. 1998;21(6):1019-23.

222. Cummings EA, Sochett EB, Dekker MG, Lawson ML, Daneman D. Contribution of growth hormone and IGF-I to early diabetic nephropathy in type 1 diabetes. Diabetes. 1998;47(8):1341-6.

223. Hourd P, Edge JA, Dunger DB, Dalton N, Edwards R. Urinary growth hormone excretion during puberty in type 1 (insulin-dependent) diabetes mellitus. Diabetic Medicine. 1991;8(3):237-42.

224. Janner M, Knill SE, Diem P, Zuppinger KA, Mullis PE. Persistent microalbuminuria in adolescents with type I diabetes mellitus is associated to early rather than late puberty. Results of a prospective longitudinal study. European Journal of Pediatrics. 1994;153(6):403-8.

225. Amin R, Schultz C, Ong K, Frystyk J, Dalton RN, Perry L, et al. Low IGF-I and Elevated Testosterone During Puberty in Subjects With Type 1 Diabetes Developing Microalbuminuria in Comparison to Normoalbuminuric Control Subjects: The Oxford Regional Prospective Study Diabetes Care. 2003 May 1, 2003;26(5):1456-61.

226. Orchard TJ, Forrest KY, Kuller LH, Becker DJ, the Pittsburgh Epidemiology of Diabetes Complications Study. Lipid and blood pressure treatment goals for type 1 diabetes: 10-year incidence data from the Pittsburgh Epidemiology of Diabetes Complications Study. Diabetes Care. 2001;24(6):1053-9.

Page 167: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

146

227. Schultz CJ, Neil HA, Dalton RN, Konopelska Bahu T, Dunger DB, Oxford Regional Prospective Study Group. Blood pressure does not rise before the onset of microalbuminuria in children followed from diagnosis of type 1 diabetes. Oxford Regional Prospective Study Group. Diabetes Care. 2001;24(3):555-60.

228. Gallego PH, Craig ME, Hing S, Donaghue KC. Role of blood pressure in development of early retinopathy in adolescents with type 1 diabetes: prospective cohort study. BMJ 2008 August 26, 2008;337(aug26_1):a918-.

229. Deckert T, Feldt-Rasmussen B, Borch-Johnsen K, Jensen T, Kofoed-Enevoldsen A. Albuminuria reflects widespread vascular damage. Diabetologia. 1989;32(4):219-26.

230. Jarvisalo MJ, Jartti L, Nanto-Salonen K, Irjala K, Ronnemaa T, Hartiala JJ, et al. Increased aortic intima-media thickness: a marker of preclinical atherosclerosis in high-risk children. Circulation. 2001 Dec 11;104(24):2943-7.

231. de Ferranti S, Mozaffarian D. The perfect storm: obesity, adipocyte dysfunction, and metabolic consequences. Clin Chem. 2008 Jun;54(6):945-55.

232. Devaraj S, Cheung AT, Jialal I, Griffen SC, Nguyen D, Glaser N, et al. Evidence of increased inflammation and microcirculatory abnormalities in patients with type 1 diabetes and their role in microvascular complications. Diabetes. 2007 Nov;56(11):2790-6.

233. Lin J, Glynn RJ, Rifai N, Manson JE, Ridker PM, Nathan DM, et al. Inflammation and progressive nephropathy in type 1 diabetes in the diabetes control and complications trial. Diabetes Care. 2008 Dec;31(12):2338-43.

234. Mangge H, Schauenstein K, Stroedter L, Griesl A, Maerz W, Borkenstein M. Low grade inflammation in juvenile obesity and type 1 diabetes associated with early signs of atherosclerosis. Exp Clin Endocrinol Diabetes. 2004 Jul;112(7):378-82.

235. Snell-Bergeon JK, West NA, Mayer-Davis EJ, Liese AD, Marcovina SM, D'Agostino RB, Jr., et al. Inflammatory markers are increased in youth with type 1 diabetes: the SEARCH Case-Control study. J Clin Endocrinol Metab. 2010 Jun;95(6):2868-76.

236. Hsueh WA, Anderson PW. Hypertension, the endothelial cell, and the vascular complications of diabetes mellitus. Hypertension. 1992 Aug;20(2):253-63.

237. Agardh CD, Agardh E, Torffvit O. The association between retinopathy, nephropathy, cardiovascular disease and long-term metabolic control in type 1 diabetes mellitus: a 5 year follow-up study of 442 adult patients in routine care. Diabetes Res Clin Pract. 1997 Mar;35(2-3):113-21.

238. Marshall G, Garg SK, Jackson WE, Holmes DL, Chase HP. Factors influencing the onset and progression of diabetic retinopathy in subjects with insulin-dependent diabetes mellitus. Ophthalmology. 1993 Aug;100(8):1133-9.

239. Chaturvedi N, Sjolie AK, Stephenson JM, Abrahamian H, Keipes M, Castellarin A, et al. Effect of lisinopril on progression of retinopathy in

Page 168: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

147

normotensive people with type 1 diabetes. The EUCLID Study Group. EURODIAB Controlled Trial of Lisinopril in Insulin-Dependent Diabetes Mellitus.Lancet. 1998 Jan 3;351(9095):28-31.

240. Strippoli GF, Craig M, Schena FP, Craig JC. Antihypertensive agents for primary prevention of diabetic nephropathy. Journal of the American Society of Nephrology. 2005 Oct;16(10):3081-91.

241. Schrier RW, Estacio RO, Esler A, Mehler P. Effects of aggressive blood pressure control in normotensive type 2 diabetic patients on albuminuria, retinopathy and strokes. Kidney Int. 2002 Mar;61(3):1086-97.

242. Norgaard K, Feldt-Rasmussen B, Borch-Johnsen K, Saelan H, Deckert T. Prevalence of hypertension in type 1 (insulin-dependent) diabetes mellitus. Diabetologia. 1990;33(7):407-10.

243. Sturrock ND, George E, Pound N, Stevenson J, Peck GM, Sowter H. Non-dipping circadian blood pressure and renal impairment are associated with increased mortality in diabetes mellitus. Diabetic Medicine. 2000;17(5):360-4.

244. Poulsen PL, Juhl B, Ebbehoj E, Klein F, Christiansen C, Mogensen CE. Elevated ambulatory blood pressure in microalbuminuric IDDM patients is inversely associated with renal plasma flow. A compensatory mechanism? Diabetes Care. 1997;20(3):429-32.

245. Lafferty AR, Werther GA, Clarke CF. Ambulatory blood pressure, microalbuminuria, and autonomic neuropathy in adolescents with type 1 diabetes. Diabetes Care. 2000;23(4):533-8.

246. Benhamou P, Halimi S, De Gaudemaris R, Boizel R, Pitiot M, Siche J, et al. Early disturbances of ambulatory blood pressure load in normotensive type I diabetic patients with microalbuminuria. Diabetes Care. 1992 November 1, 1992;15(11):1614-9.

247. Moore WV, Donaldson DL, Chonko AM, Ideus P, Wiegmann TB. Ambulatory blood pressure in type I diabetes mellitus. Comparison to presence of incipient nephropathy in adolescents and young adults. Diabetes. 1992 September 1, 1992;41(9):1035-41.

248. Darcan S, Goksen D, Mir S, Serdaroglu E, Buyukinan M, Coker M, et al. Alterations of blood pressure in type 1 diabetic children and adolescents. Pediatric Nephrology. 2006 May;21(5):672-6.

249. Lyons TJ, Jenkins AJ, Zheng D, Lackland DT, McGee D, Garvey WT, et al. Diabetic Retinopathy and Serum Lipoprotein Subclasses in the DCCT/EDIC Cohort. Invest Ophthalmol Vis Sci. 2004 March 1, 2004;45(3):910-8.

250. MRC/BHF. Heart Protection Study of cholesterol lowering with simvastatin in 20[punctuation space]536 high-risk individuals: a randomised placebocontrolled trial. Lancet. 2002;360(9326):7-22.

251. Avis HJ, Vissers MN, Stein EA, Wijburg FA, Trip MD, Kastelein JJP, et al. A Systematic Review and Meta-Analysis of Statin Therapy in Children With Familial Hypercholesterolemia. Arterioscler Thromb Vasc Biol. 2007 August 1, 2007;27(8):1803-10.

Page 169: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

148

252. Couper JJ, Staples AJ, Cocciolone R, Nairn J, Badcock N, Henning P. Relationship of smoking and albuminuria in children with insulin-dependent diabetes. Diabetic Medicine. 1994;11(7):666-9.

253. Chaturvedi N, Stephenson JM, Fuller JH. The relationship between smoking and microvascular complications in the EURODIAB IDDM Complications Study. Diabetes Care. 1995;18(6):785-92.

254. Borch-Johnsen K, Nissen H, Henriksen E, Kreiner S, Salling N, Deckert T, et al. The natural history of insulin-dependent diabetes mellitus in Denmark: 1. Long-term survival with and without late diabetic complications. Diabetic Medicine. 1987;4(3):201-10.

255. Stone ML, Craig ME, Chan AK, Lee JW, Verge CF, Donaghue KC. Natural history and risk factors for microalbuminuria in adolescents with type 1 diabetes: a longitudinal study. Diabetes Care. 2006 Sep;29(9):2072-7.

256. De Block CE, De Leeuw IH, Van Gaal LF. Impact of overweight on chronic microvascular complications in type 1 diabetic patients. Diabetes Care. 2005 Jul;28(7):1649-55.

257. Koivisto VA, Stevens LK, Mattock M, Ebeling P, Muggeo M, Stephenson J, et al. Cardiovascular disease and its risk factors in IDDM in Europe. EURODIAB IDDM Complications Study Group. Diabetes Care. 1996 Jul;19(7):689-97.

258. Orchard TJ, Chang Y-F, Ferrell RE, Petro N, Ellis DE. Nephropathy in type 1 diabetes: A manifestation of insulin resistance and multiple genetic susceptibilities? Further evidence from the Pittsburgh Epidemiology of Diabetes Complication Study. Kidney International September. 2002;62(3):963-70.

259. Yip J, Mattock M, Sethi M, Morocutti A, Viberti G. Insulin resistance in family members of insulin-dependent diabetic patients with microalbuminuria. Lancet. 1993;341(8841):369-70.

260. de Boer IH, Sibley SD, Kestenbaum B, Sampson JN, Young B, Cleary PA, et al. Central Obesity, Incident Microalbuminuria, and Change in Creatinine Clearance in the Epidemiology of Diabetes Interventions and Complications Study. J Am Soc Nephrol. 2007 January 1, 2007;18(1):235-43.

261. Donaghue KC, Chiarelli F, Trotta D, Allgrove J, Dahl-Jorgensen K. Microvascular and macrovascular complications associated with diabetes in children and adolescents. Pediatric Diabetes. 2009;10(s12):195-203.

262. ADA. Summary of Revisions for the 2006 Clinical Practice Recommendations. Diabetes Care. 2006 January 2006;29(suppl 1):s3-s.

263. American Academy of Pediatrics. Screening for retinopathy in the pediatric patient with type 1 diabetes mellitus. American Academy of Pediatrics. Sections on Endocrinology and Ophthalmology. Pediatrics. 1998 Feb;101(2):313-4.

264. Bojestig M, Arnqvist HJ, Hermansson G, Karlberg BE, Ludvigsson J. Declining incidence of nephropathy in insulin-dependent diabetes mellitus.New England Journal of Medicine. 1994;330(1):15-8.

Page 170: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

149

265. DCCT. The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. The Diabetes Control and Complications Trial Research Group. New England Journal of Medicine. 1993;329(14):977-86.

266. DCCT Effect of intensive diabetes treatment on the development and progression of long-term complications in adolescents with insulin-dependent diabetes mellitus: Diabetes Control and Complications Trial. Diabetes Control and Complications Trial Research Group.[comment]. Journal of Pediatrics. 1994;125(2):177-88.

267. Coonrod BA, Ellis D, Becker DJ, Bunker CH, Kelsey SF, Lloyd CE, et al. Predictors of microalbuminuria in individuals with IDDM. Pittsburgh Epidemiology of Diabetes Complications Study. Diabetes Care. 1993;16(10):1376-83.

268. Rudberg S, Dahlquist G. Determinants of progression of microalbuminuria in adolescents with IDDM. Diabetes Care. 1996;19(4):369-71.

269. Doria A, Warram JH, Krolewski AS. Genetic predisposition to diabetic nephropathy. Evidence for a role of the angiotensin I--converting enzyme gene. Diabetes. 1994;43(5):690-5.

270. Quinn M, Angelico MC, Warram JH, Krolewski AS. Familial factors determine the development of diabetic nephropathy in patients with IDDM. Diabetologia. 1996;39(8):940-5.

271. Earle K, Viberti GC. Familial, hemodynamic and metabolic factors in the predisposition to diabetic kidney disease. Kidney International. 1994;45(2):434-7.

272. Earle K, Walker J, Hill C, Viberti G. Familial clustering of cardiovascular disease in patients with insulin-dependent diabetes and nephropathy. New England Journal of Medicine. 1992;326(10):673-7.

273. Orchard TJ, Dorman JS, Maser RE, Becker DJ, Drash AL, Ellis D, et al. Prevalence of complications in IDDM by sex and duration. Pittsburgh Epidemiology of Diabetes Complications Study II. Diabetes. 1990;39(9):1116-24.

274. Mangili R, Deferrari G, Di Mario U, Giampietro O, Navalesi R, Nosadini R, et al. Arterial hypertension and microalbuminuria in IDDM: the Italian Microalbuminuria Study. Diabetologia. 1994;37(10):1015-24.

275. Norgaard K, Storm B, Graae M, Feldt-Rasmussen B. Elevated albumin excretion and retinal changes in children with type 1 diabetes are related to long-term poor blood glucose control. Diabetic Medicine. 1989;6(4):325-8.

276. Kelly HA, Byrne GC. Incidence of IDDM in Western Australia in children aged 0-14 yr from 1985 to 1989. Diabetes Care. 1992;15:515-7.

277. Bravo Rios EL, Cardoso Saldana G, Torres Tamayo M, Lerman Garber I, Zamora Gonzalez J, Garcia Bulnes G, et al. [Microalbuminuria and macrovascular risk factors in insulin-dependent diabetic children]. Revista de Investigacion Clinica. 1996;48(1):19-25.

Page 171: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

150

278. Salardi S, Cacciari E, Pascucci MG, Giambiasi E, Tacconi M, Tazzari R, et al. Microalbuminuria in diabetic children and adolescents. Relationship with puberty and growth hormone. Acta Paediatrica Scandinavica. 1990;79(4):437-43.

279. Quattrin T, Waz WR, Duffy LC, Sheldon MW, Campos SP, Albini CH, et al. Microalbuminuria in an adolescent cohort with insulin-dependent diabetes mellitus. Clinical Pediatrics. 1995;34(1):12-7.

280. Soper CP, Barron JL, Hyer SL. Long-term glycaemic control directly correlates with glomerular filtration rate in early Type 1 diabetes mellitus before the onset of microalbuminuria. Diabetic Medicine. 1998;15(12):1010-4.

281. Kalk WJ, Osler C, Taylor D, Panz VR. Prior long term glycaemic control and insulin therapy in insulin-dependent diabetic adolescents with microalbuminuria. Diabetes Research & Clinical Practice. 1990;9(1):83-8.

282. Mathiesen ER, Saurbrey N, Hommel E, Parving HH. Prevalence of microalbuminuria in children with type 1 diabetes mellitus. Diabetologia. 1986;29(9):640-3.

283. Olsen BS, Sjolie AK, Hougaard P, Johannesen J, Marinelli K, Jacobsen BB, et al. The significance of the prepubertal diabetes duration for the development of retinopathy and nephropathy in patients with type 1 diabetes. Journal of Diabetes & its Complications. 2004;18(3):160-4.

284. Jenkins AJ, Lyons TJ, Zheng D, Otvos JD, Lackland DT, McGee D, et al. Lipoproteins in the DCCT/EDIC cohort: associations with diabetic nephropathy. Kidney International. 2003;64(3):817-28.

285. Klein RL, Hunter SJ, Jenkins AJ, Zheng D, Semler AJ, Clore J, et al. Fibrinogen is a marker for nephropathy and peripheral vascular disease in type 1 diabetes: studies of plasma fibrinogen and fibrinogen gene polymorphism in the DCCT/EDIC cohort. Diabetes Care. 2003;26(5):1439-48.

286. Gallego PH, Gilbey AJ, Bulsara MK, Byrne GC, Jones TW, Frazer FL. Early Changes in 24-Hour Ambulatory Blood Pressure are Associated with High Normal Albumin Excretion Rate in Children with Type 1 Diabetes Mellitus. Journal of Pediatric Endocrinology. 2005;18(9):879-88.

287. Amiel SA, Sherwin RS, Simonson DC, Lauritano AA, Tamborlane WV. Impaired insulin action in puberty. A contributing factor to poor glycemic control in adolescents with diabetes. New England Journal of Medicine. 1986;315(4):215-9.

288. Williamson JR, Chang K, Tilton RG, Prater C, Jeffrey JR, Weigel C, et al. Increased vascular permeability in spontaneously diabetic BB/W rats and in rats with mild versus severe streptozocin-induced diabetes. Prevention by aldose reductase inhibitors and castration. Diabetes. 1987;36(7):813-21.

289. Williamson JR, Rowold E, Chang K, Marvel J, Tomlinson M, Sherman WR, et al. Sex steroid dependency of diabetes-induced changes in polyol metabolism, vascular permeability, and collagen cross-linking. Diabetes. 1986;35(1):20-7.

Page 172: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

151

290. Rudberg S, Persson B. Indications of low sex hormone binding globulin (SHBG) in young females with type 1 diabetes, and an independent association to microalbuminuria. Diabetic Medicine. 1995;12(9):816-22.

291. Sen A, Buyukgebiz A. Albumin excretion rate, serum insulin-like growth factor-I and glomerular filtration rate in type I diabetes mellitus at puberty. Journal of Pediatric Endocrinology & Metabolism. 1997;10(2):209-15.

292. Rudberg S, Persson B. Association between lipoprotein(a) and insulin-like growth factor I during puberty and the relationship to microalbuminuria in children and adolescents with IDDM. Diabetes Care. 1995;18(7):933-9.

293. Barkai L, Tombacz A. Alterations in insulin-like growth factor binding protein-1 and sex hormone binding globulin levels in type 1 diabetic adolescents with microalbuminuria. Diabetes Care. 2001;24(3):605-6.

294. Gibb DM, Dunger D, Levin M, Shah V, Smith C, Barratt TM. Early markers of the renal complications of insulin dependent diabetes mellitus.[comment]. Archives of Disease in Childhood. 1989;64(7):984-91.

295. Messent JW, Elliott TG, Hill RD, Jarrett RJ, Keen H, Viberti GC. Prognostic significance of microalbuminuria in insulin-dependent diabetes mellitus: a twenty-three year follow-up study. Kidney International. 1992;41(4):836-9.

296. Krolewski AS, Warram JH, Christlieb AR, Busick EJ, Kahn CR. The changing natural history of nephropathy in type I diabetes. American Journal of Medicine. 1985;78(5):785-94.

297. Kofoed-Enevoldsen A, Borch-Johnsen K, Kreiner S, Nerup J, Deckert T. Declining incidence of persistent proteinuria in type I diabetic patients in Denmark. Diabetes. 1987;36(2):205-9.

298. Schultz CJ, Neil HA, Dalton RN, Dunger DB, Oxforn Regional Prospective Study Group. Risk of nephropathy can be detected before the onset of microalbuminuria during the early years after diagnosis of type 1 diabetes. Diabetes Care. 2000;23(12):1811-5.

299. Poulsen PL, Hansen KW, Mogensen CE. Increase in nocturnal blood pressure and progression to microalbuminuria in diabetes.New England Journal of Medicine. 2003;348(3):260-4; author reply -4.

300. Lurbe E, Redon J, Kesani A, Pascual JM, Tacons J, Alvarez V, et al. Increase in nocturnal blood pressure and progression to microalbuminuria in type 1 diabetes. New England Journal of Medicine. 2002;347(11):797-805.

301. Garg SK, Chase HP, Icaza G, Rothman RL, Osberg I, Carmain JA. 24-hour ambulatory blood pressure and renal disease in young subjects with type I diabetes. Journal of Diabetes & its Complications. 1997;11(5):263-7.

302. Gilbert R, Phillips P, Clarke C, Jerums G. Day-night blood pressure variation in normotensive, normoalbuminuric type I diabetic subjects. Dippers and non-dippers. Diabetes Care. 1994 August 1, 1994;17(8):824-7.

Page 173: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

152

303. Jensen JS, Feldt-Rasmussen B, Strandgaard S, Schroll M, Borch-Johnsen K. Arterial hypertension, microalbuminuria, and risk of ischemic heart disease. Hypertension. 2000;35(4):898-903.

304. O'Brien E, Sheridan J, O'Malley K. Dippers and non-dippers. Lancet. 1988;2(8607):397.

305. Torbjornsdotter TB, Jaremko GA, Berg UB. Nondipping and Its Relation to Glomerulopathy and Hyperfiltration in Adolescents With Type 1 Diabetes. Diabetes Care. 2004 February 1, 2004;27(2):510-6.

306. Tanner JM, Whitehouse RH. Clinical longitudinal standards for height, weight, height velocity, weight velocity, and stages of puberty. Archives of Disease in Childhood. 1976;51(3):170-9.

307. Simckes AM, Srivastava T, Alon US. Ambulatory blood pressure monitoring in children and adolescents. Clinical Pediatrics. 2002;41(8):549-64.

308. Dunger DB, Schwarze CP, Cooper JD, Widmer B, Neil HA, Shield J, et al. Can we identify adolescents at high risk for nephropathy before the development of microalbuminuria? Diabet Med. 2007 Feb;24(2):131-6.

309. Poulsen PL, Ebbehoj E, Hansen KW, Mogensen CE. 24-h blood pressure and autonomic function is related to albumin excretion within the normoalbuminuric range in IDDM patients. Diabetologia. 1997;40(6):718-25.

310. Writing Team for the Diabetes C, Complications Trial/Epidemiology of Diabetes I, Complications Research G. Effect of intensive therapy on the microvascular complications of type 1 diabetes mellitus. Jama. 2002;287(19):2563-9.

311. Hovind P, Tarnow L, Rossing K, Rossing P, Eising S, Larsen N, et al. Decreasing incidence of severe diabetic microangiopathy in type 1 diabetes. Diabetes Care. 2003;26(4):1258-64.

312. Tryggvason G, Indridason OS, Thorsson AV, Hreidarsson AB, Palsson R. Unchanged incidence of diabetic nephropathy in Type 1 diabetes: a nation-wide study in Iceland. Diabetic Medicine February. 2005;22(2):182-7.

313. Pambianco G, Costacou T, Ellis D, Becker DJ, Klein R, Orchard TJ. The 30-Year Natural History of Type 1 Diabetes Complications - The Pittsburgh Epidemiology of Diabetes Complications Study Experience. Diabetes. 2006;55(May):1463-9.

314. Deckert T, Kofoed-Enevoldsen A, Norgaard K, Borch-Johnsen K, Feldt-Rasmussen B, Jensen T. Microalbuminuria. Implications for micro- and macrovascular disease. Diabetes Care. 1992;15(9):1181-91.

315. Gorman D, Sochett E, Daneman D. The natural history of microalbuminuria in adolescents with type 1 diabetes. Journal of Pediatrics. 1999;134(3):333-7.

316. Verrotti A, Cieri F, Petitti MT, Morgese G, Chiarelli F. Growth hormone and IGF-I in diabetic children with and without microalbuminuria. Diabetes, Nutrition & Metabolism Clinical & Experimental. 1999;12(4):271-6.

Page 174: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

153

317. Gallego PH, Bulsara MK, Frazer F, Lafferty AR, Davis EA, Jones TW. Prevalence and risk factors for microalbuminuria in a population-based sample of children and adolescents with T1DM in Western Australia. Pediatric Diabetes. 2006;7:165-72.

318. ISPAD. Consensus Guidelines for the Management of Type 1 Diabetes Mellitus in Children and Adolescents. International Society for Pediatric and Adolescent Diabetes. 2000:99-100.

319. Vidmar S, Carlin J, Hesketh K, Cole T. Standardizing anthropometric measures in children and adolescents with new functions for egen. The Stata Journal. 2004;4(1):1-6.

320. Falkner B, Daniels SR. Summary of the Fourth Report on the Diagnosis, Evaluation, and Treatment of High Blood Pressure in Children and Adolescents. Hypertension October. 2004;44(4):387-8.

321. Bangstad HJ, Osterby R, Hartmann A, Berg TJ, Hanssen KF. Severity of glomerulopathy predicts long-term urinary albumin excretion rate in patients with type 1 diabetes and microalbuminuria. Diabetes Care. 1999;22(2):314-9.

322. Bangstad HJ, Osterby R, Dahl-Jorgensen K, Berg KJ, Hartmann A, Hanssen KF. Improvement of blood glucose control in IDDM patients retards the progression of morphological changes in early diabetic nephropathy. Diabetologia. 1994;37(5):483-90.

323. Levy-Marchal C, Sahler C, Cahane M, Czernichow P, Gecer Study Group. Risk factors for microalbuminuria in children and adolescents with type 1 diabetes. Journal of Pediatric Endocrinology & Metabolism. 2000;13(6):613-20.

324. Riihimaa PH, Knip M, Hirvela H, Tapanainen P. Metabolic characteristics and urine albumin excretion rate in relation to pubertal maturation in Type 1 diabetes. Diabetes/Metabolism Research Reviews. 2000;16(4):269-75.

325. Holl RW, Swift PG, Mortensen HB, Lynggaard H, Hougaard P, Aanstoot HJ, et al. Insulin injection regimens and metabolic control in an international survey of adolescents with type 1 diabetes over 3 years: results from the Hvidore study group. European Journal of Pediatrics. 2003;162(1):22-9.

326. Mohsin F, Craig ME, Cusumano J, Chan AK, Hing S, Lee JW, et al. Discordant trends in microvascular complications in adolescents with type 1 diabetes from 1990 to 2002. Diabetes Care. 2005 Aug;28(8):1974-80.

327. Davis EA, Keating B, Byrne GC, Russell M, Jones TW. Hypoglycemia: incidence and clinical predictors in a large population-based sample of children and adolescents with IDDM. Diabetes Care. 1997 January 1, 1997;20(1):22-5.

328. Bulsara MK, Holman CD, Davis EA, Jones TW. The impact of a decade of changing treatment on rates of severe hypoglycemia in a population-based cohort of children with type 1 diabetes. Diabetes Care. 2004;27(10):2293-8.

Page 175: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

154

329. Pickup J, Keen H. Continuous subcutaneous insulin infusion at 25 years: evidence base for the expanding use of insulin pump therapy in type 1 diabetes. Diabetes Care. 2002;25(3):593-8.

330. Gorman D, Sochett E, Daneman D. The natural history of microalbuminuria in adolescents with type 1 diabetes. Journal of Pediatrics. 1999;134(3):333-7.

331. Caulfield M, Lavender P, Farrall M, Munroe P, Lawson M, Turner P, et al. Linkage of the angiotensinogen gene to essential hypertension. New England Journal of Medicine. 1994 Jun 9;330(23):1629-33.

332. Miller JA, Thai K, Scholey JW. Angiotensin II type 1 receptor gene polymorphism and the response to hyperglycemia in early type 1 diabetes. Diabetes. 2000;49(9):1585-9.

333. ISPAD. ISPAD Consensus Guidelines for the Management of Type 1 Diabetes Mellitus in Children and Adolescents. ISPAD - International Society for Pediatric and Adolescent Diabetes. 2000:99-100.

334. Falkner B, Daniels SR. Summary of the Fourth Report on the Diagnosis, Evaluation, and Treatment of High Blood Pressure in Children and Adolescents. Hypertension 2004;44(4):387-8.

335. Miller SA, Dykes DD, Polesky HF. A simple salting out procedure for extracting DNA from human nucleated cells. Nucleic Acids Research. 1988 1988;16(3):1215.

336. Ye S, Dhillon S, Ke X, Collins AR, Day IN. An efficient procedure for genotyping single nucleotide polymorphisms. Nucleic Acids Research. 2001;29(E88).

337. McCaskie PA, Crarker KW, Palmer LJ. SimHap: A comprehensive modelling framework and simulation-based approach to haplotypic analysis of population-based data. 2006.

338. Pavlovic M, Holl RW, Haeberle U, Schwarz K, Heinze E, Debatin KM, et al. Angiotensin I converting enzyme and angiotensinogen gene polymorphisms related to 24-h blood pressure in paediatric type I diabetes mellitus. European Journal of Pediatrics. 1999;158(1):18-23.

339. van Bockxmeer FM, Mamotte CD, Burke V, Taylor RR. Angiotensin-converting enzyme gene polymorphism and premature coronary heart disease. Clinical Science. 2000;99(3):247-51.

340. Feldt-Rasmussen B, Borch-Johnsen K, Mathiesen ER. Hypertension in diabetes as related to nephropathy. Early blood pressure changes. Hypertension. 1985;7(6 Pt 2):Nov-Dec.

341. Thomas W, Shen Y, Molitch ME, Steffes MW. Rise in albuminuria and blood pressure in patients who progressed to diabetic nephropathy in the Diabetes Control and Complications Trial. Journal of the American Society of Nephrology. 2001;12(2):333-40.

342. Yip JW, Jones SL, Wiseman MJ, Hill C, Viberti G. Glomerular hyperfiltration in the prediction of nephropathy in IDDM: a 10-year follow-up study. Diabetes. 1996;45(12):1729-33.

Page 176: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

155

343. Winkelmann BR, Russ AP, Nauck M, Klein B, Bohm B, Maier V, et al. Angiotensinogen M235T polymorphism is associated with plasma angiotensinogen and cardiovascular disease. American Heart Journal. 1999 1999/4;137(4):698-705.

344. Doria A, Onuma T, Gearin G, Freire MB, Warram JH, Krolewski AS. Angiotensinogen polymorphism M235T, hypertension, and nephropathy in insulin-dependent diabetes. Hypertension. 1996;27(5):1134-9.

345. Rogus JJ, Moczulski D, Freire MB, Yang Y, Warram JH, Krolewski AS. Diabetic nephropathy is associated with AGT polymorphism T235: results of a family-based study. Hypertension. 1998;31(2):627-31.

346. Alsaeid M, Moussa MA, Haider MZ, Refai TM, Abdella N, Al-Sheikh N, et al. Angiotensin-converting enzyme gene polymorphism and lipid profiles in Kuwaiti children with type 1 diabetes. Pediatric Diabetes. 2004;5(2):87-94.

347. Kobayashi K, Amemiya S, Mochizuki M, Matsushita K, Sawanobori E, Ishihara T, et al. Association of angiotensin-converting enzyme gene polymorphism with lipid profiles in children and adolescents with insulin-dependent diabetes mellitus. Hormone Research. 1999;51(4):201-4.

348. Tarnow L, Cambien F, Rossing P, Nielsen FS, Hansen BV, Ricard S, et al. Angiotensin-II type 1 receptor gene polymorphism and diabetic microangiopathy. Nephrology Dialysis Transplantation. 1996;11(6):1019-23.

349. Tarnow L, Kjeld T, Knudsen E, Major-Pedersen A, Parving HH. Lack of synergism between long-term poor glycaemic control and three gene polymorphisms of the renin angiotensin system on risk of developing diabetic nephropathy in type I diabetic patients. Diabetologia. 2000 Jun;43(6):794-9.

350. White NH, Cleary PA, Dahms W, Goldstein D, Malone J, Tamborlane WV, et al. Beneficial effects of intensive therapy of diabetes during adolescence: outcomes after the conclusion of the Diabetes Control and Complications Trial (DCCT). Journal of Pediatrics. 2001;139(6):804-12.

351. Hovind P, Tarnow L, Rossing P, Graae M, Torp I, Binder C, et al. Predictors for the development of microalbuminuria and macroalbuminuria in patients with type 1 diabetes: inception cohort study. Bmj. 2004 May 8, 2004;328(7448):1105.

352. Amin R, Widmer B, Dalton RN, Dunger DB. Unchanged incidence of microalbuminuria in children with type 1 diabetes since 1986: a UK based inception cohort. Archives of Disease in Childhood. 2009 April 1, 2009;94(4):258-62.

353. Dunger DB, Schwarze CP, Cooper JD, Widmer B, Neil HAW, Shield J, et al. Can we identify adolescents at high risk for nephropathy before the development of microalbuminuria? Diabetic Medicine. 2007;24(2):131-6.

354. Abraha A, Schultz C, Konopelska-Bahu T, James T, Watts A, Stratton IM, et al. Glycaemic control and familial factors determine hyperlipidaemia in early childhood diabetes. Oxford Regional Prospective Study of Childhood Diabetes. Diabetic Medicine. 1999;16(7):598-604.

Page 177: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

156

355. Willerson JT, Ridker PM. Inflammation as a cardiovascular risk factor. Circulation. 2004;109(21 Suppl 1):1.

356. King DE, Mainous AG, 3rd, Buchanan TA, Pearson WS. C-reactive protein and glycemic control in adults with diabetes.Diabetes Care. 2003;26(5):1535-9.

357. Saraheimo M, Teppo A-M, Forsblom C, Fagerudd J, Groop P-H, FinnDiane Study Group. Diabetic nephropathy is associated with low-grade inflammation in Type 1 diabetic patients. Diabetologia. 2003 2003/10;46(10):1402-7.

358. Meleth AD, Agron E, Chan CC, Reed GF, Arora K, Byrnes G, et al. Serum inflammatory markers in diabetic retinopathy. Investigative Ophthalmology & Visual Science. 2005;46(11):4295-301.

359. Soergel M, Kirschstein M, Busch C, Danne T, Gellermann J, Holl R, et al. Oscillometric twenty-four-hour ambulatory blood pressure values in healthy children and adolescents: a multicenter trial including 1141 subjects. Journal of Pediatrics. 1997;130(2):178-84.

360. Danne T, Mortensen HB, Hougaard P, Lynggaard H, Aanstoot HJ, Chiarelli F, et al. Persistent differences among centers over 3 years in glycemic control and hypoglycemia in a study of 3,805 children and adolescents with type 1 diabetes from the Hvidore Study Group. Diabetes Care. 2001;24(8):1342-7.

361. Mogensen CE. Progression of nephropathy in long-term diabetics with proteinuria and effect of initial anti-hypertensive treatment. Scandinavian Journal of Clinical & Laboratory Investigation. 1976 Jul;36(4):383-8.

362. Mogensen CE. New treatment guidelines for a patient with diabetes and hypertension. Journal of Hypertension - Supplement. 2003 Mar;21(1):S25-30.

363. Chiarelli F, Trotta D, Verrotti A, Mohn A. Treatment of hypertension and microalbuminuria in children and adolescents with type 1 diabetes mellitus. Pediatric Diabetes. 2002;3(2):113-24.

364. Cook J, Daneman D, Spino M, Sochett E, Perlman K, Balfe JW. Angiotensin converting enzyme inhibitor therapy to decrease microalbuminuria in normotensive children with insulin-dependent diabetes mellitus. Journal of Pediatrics. 1990;117(1 Pt 1):39-45.

365. Rudberg S, Aperia A, Freyschuss U, Persson B. Enalapril reduces microalbuminuria in young normotensive type 1 diabetic patients irrespective of its hypotensive effect. Diabetologia. 1990;33(8):470-6.

366. Laverman GD, Andersen S, Rossing P, Navis G, de Zeeuw D, Parving HH. Renoprotection with and without blood pressure reduction. Kidney International Supplement. 2005;94(9).

367. Andersen S, Tarnow L, Cambien F, Rossing P, Juhl TR, Deinum J, et al. Renoprotective effects of losartan in diabetic nephropathy: interaction with ACE insertion/deletion genotype? Kidney International. 2002;62(1):192-8.

Page 178: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

157

Appendix A

Information Sheet and Consent Forms

Page 179: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

158

MICROALBUMINURIA STUDY: PREVALENCE, CLINICAL CHARACTERISTICS AND

GENETIC MARKERS OF NEPHROPATHY IN CHILDREN AND ADOLESCENTS WITH

TYPE 1 DIABETES IN WESTERN AUSTRALIA.

Introduction: Among subjects with diabetes, the risk of developing complications from the

disease is greatly reduced by an improvement in the control of blood glucose. However, some

children and adolescents still develop problems with their kidneys as adults despite good control of

their blood glucose levels. As well as being related to glycaemic control, the risk of complications

may be partly inherited. Some genes may also influence the development of kidney complications.

Small leaks of protein in the urine of children and adolescents with diabetes (microalbuminuria) may

be the first marker of a later risk of developing complications. We plan to investigate the relationship

between the amount of protein in the urine, microalbuminuria, in adolescents with Type 1 diabetes

and the blood pressure.

What does the study involve? This study aims to find a relationship between blood pressure

and the amount of protein in the urine.

Your child will also be asked to provide three overnight urine samples for measurements of protein in

the urine and a blood test for assessing the genes and lipids. Blood pressure assessment will be

performed by using a 24-hour blood pressure machine.

What will happen to the data and the samples? Blood and urine samples will be

analysed at the laboratories in Princess Margaret Hospital for Children. Results will be kept

confidential and only available to the investigators involved in the study.

How will I find out the results of the tests? We will inform you and your child about all

the test results so that any necessary therapeutic adjustments can be performed. In the unlikely event

that the results are outside the normal range, we will advise you so that the necessary steps can be

taken.

If I agree to join the study: We will arrange dates for your child’s assessments. We will also

organise a convenient time to review your test results. Your involvement is entirely voluntary and

you are free to withdraw at any stage without affecting your child’s care at our department in any

way.

Confidentiality: Of note, we remind you that all the information will be kept under

confidentiality. A number will codify the data sheets. Only the principal investigators and supervisor

of the study will have access to the data.

Whom should I contact if I have any questions or concerns about the study? If you have any questions regarding this study, do not hesitate to contact:

Dr Timothy W Jones

Head of Department Endocrinology and Diabetes

Princess Margaret Hospital for Children

Roberts Road, Subiaco, WA 6008

Phone: (08) 9340-8090; FAX: (08) 9340 8605

E-mail: [email protected]

Dr Patricia Herold Gallego

Paediatric Endocrinologist Fellow

Department of Endocrinology and Diabetes

Princess Margaret Hospital for Children

Roberts Road, Subiaco

WA 6008

Phone: (08) 9340-8744/ 9340-8671

FAX: (08) 9340-8605

Email: [email protected]

Page 180: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

159

FORM OF CONSENT - PARTICIPANT

PLEASE NOTE THAT PARTICIPATION IN RESEARCH STUDIES IS

VOLUNTARY AND SUBJECTS CAN WITHDRAW AT ANY TIME WITH

NO IMPACT ON CURRENT OR FUTURE CARE.

I

....................................................................................................................................

have read and understood the information explaining the study entitled

“Microalbuminuria: Prevalence, Clinical Characteristics and Genetic Markers

of Nephropathy in Children and Adolescents with Type 1 Diabetes Mellitus in

Western Australia”

I agree to participate and I understand that I may withdraw from the study at

any stage and withdrawal will not interfere with routine care.

I agree that research data gathered from the results of this study may be

published, provided that names are not used.

Dated ................................. day of ............................................................ 20 ..........

Participant’s Signature ..................................................………………………………..

I,……............................................... …………………………………………………...

(investigator’s name)

have explained the above to the signatories who stated that he/she understood the

same.

Signature:.......................................................................................…………………….

Page 181: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

160

FORM OF CONSENT TO PARENTS AND CHILD

PLEASE NOTE THAT PARTICIPATION IN RESEARCH STUDIES IS

VOLUNTARY AND SUBJECTS CAN WITHDRAW AT ANY TIME WITH

NO IMPACT ON CURRENT OR FUTURE CARE.

I

....................................................................................................................................

have read and understood the information explaining the study “ Microalbuminuria:

Prevalence, Clinical Characteristics and Genetic Markers of Nephropathy in

Children and Adolescents with Type 1 Diabetes Mellitus in Western Australia”

Any questions I have asked have been answered to my satisfaction. I agree to

participate in this study and I understand my child may withdraw from the study at

any stage and withdrawal will not interfere with routine care.

I agree that research data gathered from the results of this study may be

published, provided that names are not used.

Dated.................... day of........................................................ 20 ........…………………

Child’s Signature ...................................………………………………………………..

Parent or Guardian’s

Signatures…………………………………………………………

I, ....................................................... …………………..have explained the above to

the (Investigator’s full name)

signatory who stated that he/she understood the same.

Signature

.............................................................................................................................

Page 182: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

161

DNA CONSENT FORM FOR GENE STUDIES

I consent to the collection from my child of (blood – via venepuncture, thumb

prick; cells - mouthwash) from which DNA will be extracted and stored for the gene

studies that have been explained to me as part of the study: “Microalbuminuria:

Prevalence, Clinical Characteristics and Genetic Markers of Nephropathy in

Children and Adolescents with Type 1 Diabetes Mellitus in Western Australia”

I consent to my child’s DNA being used for gene research into ……………………..

(condition)

I understand that the DNA will not be used for purposes other than that specified

above and will not be used for diagnostic purposes.

Name of child: ………………………. …………………………………………….

Name of Parent / Guardian: ………………………………………………………...

Signature: …………………………………………….Date: ……………………..

Witness: ……………………………………………....Date: ……………………..

Page 183: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

162

DNA CONSENT FORM FOR GENE STUDIES

I consent to the collection of my (blood – via venepuncture, thumb prick; cells -

mouthwash) from which DNA will be extracted and stored for the gene studies that

have been explained to me as part of the study “Microalbuminuria: Prevalence,

Clinical Characteristics and Genetic Markers of Nephropathy in Children and

Adolescents with Type 1 Diabetes Mellitus in Western Australia”

I consent to my DNA being used for gene research into ……………………..

(condition)

I understand that the DNA will not be used for purposes other than that specified

above and will not be used for diagnostic purposes.

Name of Participant: ……………………………………………………………….

Signature: ……………………………………………….Date: ……………………

Witness: ………………………………………………..Date: …………………...

Page 184: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

[Type text]

163

Appendix B

Classification and characteristic features of T1DM in children

Screening guidelines for vascular complications in the

paediatric and adolescent T1DM group

Page 185: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

Association Between p.Leu54MetPolymorphism at the Paraoxonase-1 Geneand Plantar Fascia Thickness in YoungSubjects With Type 1 DiabetesPATRICIA H. GALLEGO, MD, MSC

1

MARIA E. CRAIG, MBBS, PHD, FRACP1,2,3

ANTHONY C. DUFFIN, PHD1

BRUCE BENNETTS, PHD, FHGSA4

ALICIA J. JENKINS, MBBS, MD, FRACP, FRCP5

SABINE HOFER, MD6

ALBERT LAM, MD, FRACR7

KIM C. DONAGHUE, MBBS, PHD, FRACP1,2

OBJECTIVE — In type 1 diabetes, plantar fascia, a collagen-rich tissue, is susceptible toglycation and oxidation. Paraoxonase-1 (PON1) is an HDL-bound antioxidant enzyme. PON1polymorphisms have been associated with susceptibility to macro- and microvascular compli-cations. We investigated the relationship between plantar fascia thickness (PFT) and PON1 genevariants, p.Leu54Met, p.Gln192Arg, and c.-107C�T, in type 1 diabetes.

RESEARCH DESIGN AND METHODS — This was a cross-sectional study of 331adolescents with type 1 diabetes (162 male and 169 female). PFT was assessed by ultrasound,PON1 was assessed by genotyping with PCR and restriction fragment–length polymorphism,and serum PON1 activity was assessed by rates of hydrolysis of paraoxon and phenylacetate.

RESULTS — Median (interquartile range) age was 15.4 (13.5–17.3) years, and diabetes du-ration was 7.6 (4.9–10.6) years. The distribution of p.Leu54Met genotypes was LL 135 (40.8%),ML 149 (45%), and MM 47 (14.2%). PFT was abnormal (�1.7 mm) in 159 adolescents (48%).In multivariate analysis, predictors of abnormal PFT were ML/LL versus MM p.Leu54Met poly-morphism (odds ratio 3.84 [95% CI 1.49–9.82], P � 0.005); BMI (percentile) (1.02 [1.01–1.03], P � 0.007); systolic blood pressure (percentile) (1.01 [1.00–1.02], P � 0.03); and malesex (3.29 [1.98–5.46], P � 0.001).

CONCLUSIONS — Thickening of the plantar aponeurosis occurs predominantly in over-weight and male adolescents with type 1 diabetes. The MM genotype at PON1 p.Leu54Met isassociated with a reduced risk of abnormal PFT.

Diabetes Care 31:1585–1589, 2008

P araoxonase-1 (PON1) is a calcium-dependent HDL-associated enzymethat protects LDLs from oxidation.

In type 1 diabetic patients, the serumparaoxonase concentration is lower andHDL is a less efficient antioxidant than inhealthy individuals (1). Oxidized LDL isimplicated in the pathogenesis of athero-

sclerosis, diabetic retinopathy, and ne-phropathy (2). Variations in lipoprotein-related enzymes and genotypes may alsopromote diabetic microvascular damage(3).

Soft-tissue thickening is associatedwith chronic hyperglycemia and is hy-pothesized to be due to collagen glycation

(4). With use of ultrasound techniques tomeasure plantar aponeurosis, a collagen-rich tissue, researchers demonstrated pre-viously that people with diabetes haveincreased plantar fascia thickness (PFT)(5). Recently, this group reported that in-creased PFT predicted the developmentof microvascular complications in adoles-cents with type 1 diabetes and proposedabnormal PFT as a putative marker ofsoft-tissue glycation (6).

The PON gene cluster maps to chro-mosome 7q21-22 and influences gene ex-pression and serum activity. There is anestablished link between PON1 and mac-rovascular disease (7) and emerging evi-dence linking PON1 to microvascularcomplications (8,9). In this study we in-vestigated whether the variants c.-107C�T at the promoter region andp.Leu54Met and p.Gln192Arg at thecoding regions of PON1 are associatedwith PFT in type 1 diabetes.

RESEARCH AND METHODS —The cohort consisted of 331 Caucasianadolescents and young adults (162 maleand 169 female) with a median (inter-quartile range) age of 15.4 (13.5–17.3)years and type 1 diabetes duration of 7.6(4.9–10.6) years, who presented for rou-tine complications assessment at the Chil-dren’s Hospital at Westmead (Sydney,Australia) between 1998 and 2004. Thestudy was approved by the hospital’s eth-ics committee, and written informed con-sent was obtained.

PFT measurementA single investigator (A.D.) measured PFTusing ultrasound (Acuson 128 gray scaleimager; Acuson, Mountain View, CA).For aponeurosis measurements, a linear-array, 7-MHz high-resolution transducerwas placed longitudinally over the centerof the arch at least 3 cm from the calcanealinsertion. To assess test-retest variability,four individuals were assessed 10 times inthe same session. The assessor wasmasked to the measurements until theywere completed and stored. The intra-class coefficient was 0.89. Abnormal PFT

● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ● ●

From the 1Institute of Endocrinology and Diabetes, The Children’s Hospital at Westmead, Sydney, NewSouth Wales, Australia; the 2Discipline of Paediatrics and Child Health, University of Sydney, Sydney,New South Wales, Australia; the 3School of Women’s and Children’s Health, University of New SouthWales, Sydney, New South Wales, Australia; the 4Department of Molecular Genetics, The Children’sHospital at Westmead, Sydney, New South Wales, Australia; the 5Department of Medicine (St. Vincent’s),University of Melbourne, Melbourne, Victoria, Australia; the 6Department of Paediatrics, Medical Uni-versity of Innsbruck, Innsbruck, Austria; and the 7Department of Radiology, The Children’s Hospital atWestmead, Sydney, New South Wales, Australia.

Corresponding author: Patricia H. Gallego, [email protected] 25 November 2007 and accepted 2 May 2008.Published ahead of print at http://care.diabetesjournals.org on 9 May 2008. DOI: 10.2337/dc07-2236.© 2008 by the American Diabetes Association. Readers may use this article as long as the work is properly

cited, the use is educational and not for profit, and the work is not altered. See http://creativecommons.org/licenses/by-nc-nd/3.0/ for details.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be herebymarked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

P a t h o p h y s i o l o g y / C o m p l i c a t i o n sO R I G I N A L A R T I C L E

DIABETES CARE, VOLUME 31, NUMBER 8, AUGUST 2008 1585

Page 186: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

(�1.7 mm) was defined as 2 SDs abovethe mean measurement of 57 age-matched unrelated nondiabetic controlsubjects (27 male, median age 15.6 years)(5).

PON1 genotypingDNA was extracted from peripheral whiteblood cells collected in lithium-heparintubes using a modified salting out proto-col. Polymorphisms were analyzed byPCR-restriction fragment–length poly-morphism using a slightly modified ver-sion of the procedure described byHumbert et al. (10). DNA (100 ng) wasdenatured (94°C, 12 min) and then am-plified for 35 cycles using PCR primers.Each cycle consisted of denaturation(94°C, 30 s), annealing (55°C, 30 s), andextension (72°C, 30 s) with a final exten-sion of 5 min. For p.Leu54Met geno-typing, the 171-bp PCR product wasdigested (37°C, 2 h) with Hsp92II (Pro-mega, Madison, WI), and products wereseparated by PAGE (10%) and stained byethidium bromide. The leucine allele cor-responded to the presence of a non-digested fragment of 171 bp; themethionine allele corresponded to twodigestion fragments of 127 and 44 bp.

For p.Gln192Arg genotyping, the99-bp PCR product was digested withDpnII, and products were separated byPAGE (10%). The arginine allele corre-sponded to the presence of three diges-tion fragments (40, 31, and 28 bp); theglutamine allele corresponded to two di-gestion fragments of 59 and 40 bp.

For c.-107C�T genotyping, the240-bp PCR product was digested withBsrBI, and products were separated byPAGE (10%). The TT genotype corre-sponded to a nondigested 240-bp frag-

ment, the CC genotype corresponded totwo digestion fragments of 212 and 28bp, and the CT genotype corresponded tothree digestion fragments of 240, 212,and 28 bp.

PON1 activityIn a representative subgroup of subjects(n � 144), PON1 activity of lithium-heparin plasma was measured by the ratesof hydrolysis of paraoxon and phenylace-tate as described previously (9).

Other variablesA1C was measured by a Bio-Rad Diamatanalyzer (nondiabetic range of 4 – 6%;Bio-Rad Laboratories, Hercules, CA).Nonfasting plasma cholesterol was mea-sured by Cobas Mira. Systolic blood pres-sure (SBP), diastolic blood pressure(DBP), and BMI percentiles were deter-mined using age- and sex-related refer-ence standards.

Statistical analysisDescriptive statistics are reported asmeans � SD or median (interquartilerange) for skewed distributions. Groupswere compared by �2 test for categoricalvariables. Differences between indepen-dent samples were evaluated using Stu-dent’s t test and one-way ANOVA fornormally distributed data or the Mann-Whitney U test for skewed data.

Multiple logistic regression was usedto evaluate the association between ab-normal PFT and biological and geneticvariables. Explanatory variables were sex,age, diabetes duration, A1C, BMI, SBP,cholesterol, and PON1 genotypes (MMvs. ML/LL for p.Leu54Met; QQ vs. RQ/RRfor p.Gln192Arg, and TT vs. CT/CC forc.-107C�T). Results are expressed as

odds ratios and 95% CI. P � 0.05 indi-cated statistical significance.

RESULTS — Clinical characteristicsare summarized in Table 1. Patients withabnormal PFT had higher SBP and BMIpercentiles and were more likely to bemale. PON1 activity was not significantlydifferent between those with or withoutabnormal PFT for any substrate.

Table 2 shows genotype distributionsand allele frequencies (all mutations werein Hardy-Weinberg equilibrium). Activi-ties for phenylacetate and paraoxon sub-strates were higher for patients carryingLL alleles at PON1 p.Leu54Met versusthose with the MM genotype. Similarly,those with CC alleles at c.-107C�T hadhigher enzyme activity for both substratesversus those with the TT genotype. Atp.Gln192Arg, enzyme activity for hydro-lysis of paraoxon was higher in those withthe RR versus the QQ genotype, but phe-nylacetate activities did not differ.

The p.Leu54Met genotype distribu-tion differed between the two PFT groups.The MM genotype was less frequentamong those with thickened aponeurosis(normal PFT 20% vs. abnormal PFT 8%,�2, P � 0.01). There was no difference infrequency of p.Gln192Arg or c.-107C�Tgenotype distribution by PFT (Table 3).

In the multiple regression, LL/ML atp.Leu54Met, male sex, and BMI and SBPpercentiles were associated with in-creased PFT (Table 4). There was no in-teraction between sex and BMI, sex andPON1 variants, or BMI and PON1 vari-ants. There was strong linkage disequilib-r ium be tween p .Leu54Met andp.Gln192Arg and between p.Leu54Metand c.-107C�T genotypes (not shown).

Table 1—Clinical characteristics of young subjects with type 1 diabetes with and without abnormal PFT

Normal PFT Abnormal PFT Total P value

n 172 159 331Age (years) 15.7 (13.5–17.3) 15.1 (13.5–17.3) 15.4 (13.5–17.3) 0.46Sex (male/female) 62/110 100/59 172/159 �0.001Diabetes duration (years) 7.6 (5.0–10.3) 7.9 (4.3–11.0) 7.6 (4.9–10.6) 0.77A1C (%) 8.6 � 1.3 8.5 � 1.1 8.6 � 1.3 0.69Total cholesterol (mmol/l) 4.3 (3.9–5.0) 4.3 (3.8–4.8) 4.3 (3.8–4.9) 0.42SBP (percentile) 54 (27–82) 71 (38–85) 59 (32–82) 0.004DBP (percentile) 73 (48–83) 73 (48–87) 73 (48–84) 0.49BMI (percentile) 74 (56–87) 78 (63–91) 76 (61–88) 0.02PON1 activity*

Paraoxon (units/ml) 39.6 (28.3–79.3) 45.4 (28.1–85.4) 41.4 (28.4–79.9) 0.41Phenylacetate (units/ml) 64.9 (44.6–81.6) 70.6 (56.7–80.4) 66.7 (52.8–80.5) 0.26

Data are means � SD or median (interquartile range). *PON1 activity was measured in 144 subjects.

Plantar fascia thickness and PON1 gene

1586 DIABETES CARE, VOLUME 31, NUMBER 8, AUGUST 2008

Page 187: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

CONCLUSIONS — This is the firstreport demonstrating an association be-tween PFT, a marker of tissue glycationand/or oxidation, and PON1 gene poly-morphisms in type 1 diabetes. In thiscross-sectional study, the L allele at thep.Leu54Met gene was associated with ab-normal PFT, or, conversely, the M allelehad a protective effect. These results sup-port the association between PON1 genepolymorphisms and plantar fasciachanges. Previously, our group showedthat the LL genotype was closely associ-ated with microvascular complications(9,11).

After a relatively short median diabe-tes duration of 7 years, approximately halfof the cohort had abnormal plantar fasciameasurements. The early appearance ofabnormal PFT, although unrelated toA1C measured at the time of the assess-ment, does not exclude the possibilitythat early glycemic variability could leavean imprinting in target organs, includingchanges in collagen, predisposing theseorgans to the future development of com-plications. In support of this possibility,we have recently demonstrated that PFTpredicts retinopathy, early elevation of al-bumin excretion rate, and nerve abnor-malities in young people with type 1diabetes (6).

Oxidative stress and abnormalities oflipoprotein quantity and quality are im-plicated in the pathogenesis of microvas-cular complications (3). We hypothesizethat genetic variants at the PON1 gene, foran antioxidant enzyme, could also predis-pose to collagen abnormalities via ad-vanced glycation end product (AGE)

formation, which involves glycation andoxidation. Tissue glycation and/or oxida-tion via intra- and extracellular generationof AGEs may promote diabetes complica-tions. The Diabetes Control and Compli-cations Trial/Epidemiology of DiabetesInterventions and Complications (DCCT/EDIC) demonstrated in skin biopsies thatcollagen AGEs predict microvascular dis-ease (12). Skin collagen methionine sul-foxide, a marker of oxidative damage

independent of glycemia, has also beenassociated with type 1 diabetes complica-tions (13).

There are divergent reports of PON1activity and genotype and their relation-ship to microvascular complications. Ourgroup described a higher allelic frequencyof leucine 54 among type 1 diabetic sub-jects with versus without retinopathy(14). Similar findings were subsequentlyconfirmed in a larger cohort of 372 type 1

Table 2—Allele frequencies and enzyme activity of PON1 genotypes for hydrolysis of paraoxon and phenylacetate as substrates

Polymorphism Genotype (n)Frequency

(Hardy-Weinberg)Paraoxon(units/ml) Phenylacetate (units/ml)

n 331 144 144p.Leu53Met LL (135) 0.40 (0.39) 77.8 (46.2–106.5) 73.4 (64.9–87.7)

ML (149) 0.45 (0.46) 39.1 (28.4–64.7) 66.2 (53.0–80.9)MM (47) 0.14 (0.13) 21.0 (16.7–30.4) 56.0 (39.5–63.0)

P value �0.001 �0.001p.Gln191Arg RR (29) 0.09 (0.08) 99.8 (73.0–185.0) 68.2 (32.2–85.8)

RQ (127) 0.38 (0.39) 80.3 (63.4–106.1) 68.7 (54.6–80.1)QQ (175) 0.53 (0.52) 30.5 (24.5–39.8) 66.7 (53.1–81.7)

P value �0.001 0.81c.-107C�T* CC (90) 0.27 (0.26) 61.4 (37.9–90.3) 77.4 (68.4–90.2)

CT (154) 0.49 (0.49) 40.3 (28.7–82.3) 70.3 (59.4–81.6)TT (75) 0.24 (0.24) 34.6 (20.8–73.6) 54.0 (39.7–63.2)

P value 0.017 �0.001

Data are median (interquartile range) unless otherwise indicated. *For c.-107C�T, genotyping was performed in 319 individuals and PON1 activity measured in139 subjects.

Table 3—Distribution of PON1 genotype in subjects with type 1 diabetes with and withoutabnormal PFT

Normal PFT Abnormal PFT P value

p.Leu54MetLL 66 (38) 69 (43)ML 72 (42) 77 (49) 0.01MM 34 (20) 13 (8)

p.Leu54MetMM 34 (20) 13 (8)ML/LL 138 (80) 146 (92) 0.003

p.Gln192ArgRR 14 (8) 15 (9)RQ 64 (37) 63 (40) 0.81QQ 94 (55) 81 (51)

p.Gln192ArgQQ 94 (55) 81 (51)RQ/RR 78 (45) 78 (49) 0.58

c.-107C�TTT 41 (25) 34 (22)CT 76 (46) 78 (51) 0.65CC 49 (29) 41 (27)

c.-107C�TTT 41 (25) 34 (22)CT/CC 125 (75) 119 (78) 0.69

Data are n (%).

Gallego and Associates

DIABETES CARE, VOLUME 31, NUMBER 8, AUGUST 2008 1587

Page 188: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

diabetic adolescents in whom the LL ge-notype increased risk for early retinopa-thy by almost threefold (8). The presenceof the LL genotype and two other PON1polymorphisms was found to influenceurinary albumin excretion in 156 Cauca-sian type 1 diabetic adolescents (9). Incontrast, others found no correlation be-tween microvascular complications andPON1 polymorphisms in adults with type1 diabetes (15). Divergent results may beexplained by differences in ethnicity, age,sample size, smoking, diet, concomitantmedications (16), PON1 assays, and dif-fering relationships between PON activityor genotype for initiation and progressionof complications.

Conversely, there is strong evidenceshowing that “high-expressor” PON1 al-leles influence macrovascular disease.Homozygosity for the L allele doubledcardiovascular disease risk after adjust-ment for other risk factors in diabetes(17).

If PON1 protects lipids from oxida-tion, one might expect a higher expressorallele and a higher enzymatic activity to beprotective against oxidative stress, AGEformation, and diabetes complications.However, the artificial nature of the sub-strates in this study is recognized and maynot represent physiological substrates. Arecent report demonstrated that the pri-mary activity of the paraoxonases is thatof a lactonase (18). As yet, there are nostudies relating lactonase activity to dia-betes complications.

Although PON1 activity was mea-sured in a subgroup of subjects, PON1polymorphisms influenced at least one ofthe two measured activities, in agreementwith published data (19). PON1 activitywas higher in the abnormal PFT groupbut did not achieve statistical significance.There is a large interindividual variationin PON activity that mostly depends onfunctional and promoter variations at thePON gene. Lower PON activity has beenreported in type 1 diabetic subjects versuscontrol subjects regardless of differences

in PON1 phenotype (20). Hyperglycemiamay influence the in vivo concentrationand in vitro activity of PON1. Kordonouriet al. (11) showed that higher PON1 ac-tivity was associated with high glucoselevels but not with A1C. With adjustmentfor blood glucose and diabetes duration,PON1 activity was higher in subjects withdifferent stages of retinopathy versusthose without retinopathy.

Our findings suggest that high-expressor alleles and a higher enzymaticactivity are associated with increased sus-ceptibility to microvascular complica-tions (8,9,11). One explanation could bethat although paraoxonase hydrolyzesharmful lysolipids produced by peroxida-tion, it might also increase their produc-tion from phospholipid peroxidationproducts and create a more harmful li-poprotein profile. The LL genotype hasbeen shown to be least protective againstoxidation (21). Another possibility is thatrelatively reduced enzyme activity ratherthan increased absolute PON1 activitypromotes vascular complications. Somehave reported that a higher LDL choles-terol–to-PON concentration ratio may in-dicate a reduced capacity of the enzyme tolimit LDL oxidation (22). The low-activityallele at the PON1 gene has also been as-sociated with a less harmful lipoproteinprofile (23). Data on the LDL cholesterol–to-PON concentration ratio or levels ofLDL cholesterol, triglycerides, and apoli-poprotein B were not available for thisstudy.

Male sex presented a threefold in-creased risk for thickened aponeurosis in-dependent of other risk factors. Thereasons for this are unclear, but men havea greater risk of lower limb diabetes com-plications (24). Hormonal variation anddifferent recreational activities may becontributors. Although level of physicalactivity was not measured, it is unlikelythat plantar fasciitis, a common conditionamong athletes, is the cause of fasciathickness. Fasciitis is associated with heelpain, and its ultrasonographic changes

are limited to the proximal insertion ofthe aponeurosis (25).

Higher BMI and SBP were associatedwith abnormal PFT. Although BMI mayincrease mechanical load on the plantaraponeurosis, overweight is associatedwith microvascular complications in dia-betes, insulin resistance, hypertension,and an atherogenic profile.

In summary, this study underlinesthe association between p.Leu54Met vari-ants and PFT, implicating PON1 in thepathogenesis of diabetes-related collagenchanges. Although relationships betweenPON1 genes and collagen abnormalitiesmerit further investigation, these initialfindings support the concept of PON1 ge-netic variants as a link predisposing to de-velopment of complications in type 1diabetes.

Acknowledgments— This study was sup-ported by a Diabetes Australia Research TrustNew Millennium Grant.

We thank Connie Karschimkus and CheeLee for PON1 activity measurements.

References1. Ferretti G, Bacchetti T, Busni D, Rabini

RA, Curatola G: Protective effect of para-oxonase activity in high-density lipopro-teins against erythrocyte membranesperoxidation: a comparison betweenhealthy subjects and type 1 diabetic pa-tients. J Clin Endocrinol Metab 89:2957–2962, 2004

2. Kennedy AL, Lyons TJ: Glycation, oxida-tion, and lipoxidation in the developmentof diabetic complications. Metabolism 46:14–21, 1997

3. Jenkins AJ, Rowley KG, Lyons TJ, Best JD,Hill MA, Klein RL: Lipoproteins and dia-betic microvascular complications. CurrPharm Des 10:3395–3418, 2004

4. James VJ, Delbridge L, McLennan SV, YueDK: Use of X-ray diffraction in study ofhuman diabetic and aging collagen. Dia-betes 40:391–394, 1991

5. Duffin AC, Lam A, Kidd R, Chan AKF,Donaghue KC: Ultrasonography of plan-tar soft tissues thickness in young peoplewith diabetes. Diabet Med 19:1009–1013,2002

6. Craig ME, Duffin AC, Gallego PH, Lam A,Cusumano J, Hing S, Donaghue K: Plantarfascia thickness, a measure of tissue gly-cation, predicts the development of com-plications in adolescents with type 1diabetes. Diabetes Care In press

7. Mackness MI, Mackness B, DurringtonPN: Paraoxonase and coronary heart dis-ease. Atheroscler Suppl 3:49–55, 2002

8. Kao Y, Donaghue KC, Chan A, BennettsBH, Knight J, Silink M: Paraoxonase gene

Table 4—Factors associated with abnormal PFT in young subjects with type 1 diabetes usingmultiple regression analysis

Odds ratio (95% CI) P value

p.Leu54Met (ML/LL vs. MM) 3.84 (1.49–9.82) 0.005Sex (male) 3.29 (1.98–5.46) �0.001BMI (percentile) 1.02 (1.01–1.03) 0.007SBP (percentile) 1.01 (1.001–1.02) 0.03

The variables explained 19% of variation of abnormal PFT.

Plantar fascia thickness and PON1 gene

1588 DIABETES CARE, VOLUME 31, NUMBER 8, AUGUST 2008

Page 189: Patricia Herold Gallego - the UWA Profiles and Research ... · Gallego (the candidate) at the Department of Endocrinology and Diabetes at Princess Margaret Hospital for Children ,

cluster is a genetic marker for early micro-vascular complications in T1DM.DiabetMed 19:212–215, 2002

9. Hofer SE, Bennetts B, Chan AK, HollowayB, Karschimkus C, Jenkins AJ, Silink M,Donaghue KC: Association between PON1 polymorphisms, PON activity and dia-betes complications. J Diabetes Complica-tions 20:322–328, 2006

10. Humbert R, Adler DA, Disteche CM, Has-sett C, Omiecinski CJ, Furlong CE: Themolecular basis of the human serum para-oxonase activity polymorphism. NatGenet 3:73–76, 1993

11. Kordonouri O, James RW, Bennetts B,Chan A, Kao YL, Danne T, Silink M,Donaghue K: Modulation by blood glu-cose levels of activity and concentration ofparaoxonase in young patients with type1 diabetes mellitus. Metabolism 50:657–660, 2001

12. Genuth S, Sun W, Cleary P, Sell DR,Dahms W, Malone J, Sivitz W, MonnierVM: Glycation and carboxymethyllysinelevels in skin collagen predict the risk offuture 10-year progression of diabetic ret-inopathy and nephropathy in the DCCTand epidemiology of diabetes interven-tions and complications participants withtype 1 diabetes. Diabetes 54:3103–3111,2005

13. Yu Y, Thorpe SR, Jenkins AJ, Shaw JN,Sochaski MA, McGee D, Aston CE, Or-chard TJ, Silvers N, Peng YG, McKnightJA, Baynes JW, Lyons TJ: Advanced gly-

cation end-products and methionine sul-phoxide in skin collagen of patients withT1DM. Diabetologia 49:2488–2498,2006

14. Kao YL, Donaghue K, Chan A, Knight J,Silink M: A variant of paraoxonase(PON1) gene is associated with diabeticretinopathy in IDDM. J Clin EndocrinolMetab 83:2589–2592, 1998

15. Araki S, Makita Y, Canani L, Ng D, War-ram JH, Krolewski AS: Polymorphisms ofhuman paraoxonase 1 gene (PON1) andsusceptibility to diabetic nephropathy intype I diabetes mellitus. Diabetologia 43:1540–1543, 2000

16. Aviram M, Rosenblat M: Paraoxonasesand cardiovascular diseases: pharmaco-logical and nutritional influences. CurrOpin Lipidol 16:393–399, 2005

17. Garin MC, James RW, Dussoix P, BlancheH, Passa P, Froguel P, Ruiz J: Paraoxonasepolymorphism Met-Leu54 is associatedwith modified serum concentrations ofthe enzyme. A possible link between theparaoxonase gene and increased risk ofcardiovascular disease in diabetes. J ClinInvest 99:62–66, 1997

18. Khersonsky O, Tawfik DS: Structure-re-activity studies of serum paraoxonasePON1 suggest that its native activity islactonase. Biochemistry 44:6371–6382,2005

19. Brophy VH, Hastings MD, ClendenningJB, Richter RJ, Jarvik GP, Furlong CE:Polymorphisms in the human paraoxo-

nase (PON1) promoter. Pharmacogenetics11:77–84, 2001

20. Abbott CA, Mackness MI, Kumar S, Boul-ton AJ, Durrington PN: Serum paraoxo-nase activity, concentration, and phe-notype distribution in diabetes mellitusand its relationship to serum lipids andlipoproteins. Arterioscler Thromb Vasc Biol15:1812–1818, 1995

21. Mackness MI, Durrington PN, MacknessMI, Durrington PN: HDL, its enzymesand its potential to influence lipid peroxi-dation. Atherosclerosis 115:243–253,1995

22. James RW, Leviev I, Ruiz J, Passa P,Froguel P, Garin MC, James RW, Leviev I,Ruiz J, Passa P, Froguel P, Garin MC: Pro-moter polymorphism T(�107)C of theparaoxonase PON1 gene is a risk factorfor coronary heart disease in type 2 dia-betic patients. Diabetes 49:1390–1393,2000

23. Hegele RA, Brunt JH, Connelly PW: Apolymorphism of the paraoxonase geneassociated with variation in plasma li-poproteins in a genetic isolate. ArteriosclerThromb Vasc Biol 15:89–95, 1995

24. Duffin AC, Donaghue KC, Potter M,McInnes A, Chan AK, King J, Howard NJ,Silink M: Limited joint mobility in thehands and feet of adolescents with T1DM.Diabet Med 16:125–130, 1999

25. Wall JR, Harkness MA, Crawford A: Ul-trasound diagnosis of plantar fasciitis.Foot Ankle 14:465–470, 1993

Gallego and Associates

DIABETES CARE, VOLUME 31, NUMBER 8, AUGUST 2008 1589