identification of genetic modifiers in lrrk2 …

155
IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 PARKINSONISM by Joanne Trinh BSc, The University of British Columbia, 2012 A THESIS SUBMITTED IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY in The Faculty of Graduate and Postdoctoral Studies (Medical Genetics) THE UNIVERSITY OF BRITISH COLUMBIA (Vancouver) December 2016 © Joanne Trinh, 2016

Upload: others

Post on 09-Nov-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 PARKINSONISM

by

Joanne Trinh

BSc, The University of British Columbia, 2012

A THESIS SUBMITTED IN PARTIAL FULFILLMENT OF

THE REQUIREMENTS FOR THE DEGREE OF

DOCTOR OF PHILOSOPHY

in

The Faculty of Graduate and Postdoctoral Studies

(Medical Genetics)

THE UNIVERSITY OF BRITISH COLUMBIA

(Vancouver)

December 2016

© Joanne Trinh, 2016

Page 2: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

ii

Abstract

Genetic studies have been extremely informative to the pathophysiology of PD. The most

common pathogenic mutation discovered is LRRK2 p.G2019S which accounts for 30-40% of

Parkinson disease (PD) in North African Arab Berbers, 18-30% in Ashkenazi Jews and 1-3% in

Caucasians. Although LRRK2 p.G2019S parkinsonism is considered a monogenic form of

disease, disease penetrance of motor symptoms is variable. We hypothesize that genetic factors

can modulate the phenoconversion of LRRK2 p.G2019S which could lead to treatments that

prevent onset or delay disease progression.

Clinical characterization of LRRK2 p.G2019S carriers from Tunisia was performed by

analysis of motor and non-motor features. Genetic analysis of age of onset as a genetic trait was

performed in a cohort of Tunisian Arab Berbers with LRRK2 p.G2019S. Short-tandem repeat

genotyping (4cM resolution) and non-parametric and model-based genome-wide linkage was

evaluated in 41 multi-incident LRRK2 p.G2019S families. High-density locus-specific

genotyping and association analyses were also performed in 232 unrelated LRRK2 p.G2019S

carriers. Genome sequencing in a subset of 25 subjects informed imputation and haplotype

analyses. Validation analysis used Sanger sequencing and Taqman genotyping on additional

LRRK2 p.G2019S carriers originating from Algeria, France and Norway. Whole transcriptome,

candidate gene and protein expression was assessed in striatum from 60 human brains.

Significant linkage was identified on chromosome 1q23.3-24.3 (model-based LOD=4.99,

D1S2768). In the chromosome 1q23.3-24. interval higher-resolution SNP genotyping,

association and haplotype mapping nominated genetic variability within DNM3 as an age of

onset modifier of disease penetrance (rs2421947 nominal p<10-5

; haplotype p=1.67 x 10-7

). In

terms of age of onset the penetrance of parkinsonism in LRRK2 p.G2019S carriers varies as a

Page 3: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

iii

function of DNM3 genotype; rs2421947 is a haplotype-tag for which median onset in GG

carriers is 13 years younger than CC carriers (HR 1.63 CI=1.05-2.63, p=0.03). DNM3 rs2421947

variability is also directly correlated with dynamin 3 mRNA and protein expression in human

brain striatum (p<0.05).

Dynamin 3, shown to complex with endophilin A, LRRK2 and vacuolar protein sorting

35, localizes to the endocytic machinery of dendritic spines to modulate receptor recycling and

excitatory synaptic transmission, now suggests novel targets for therapeutic development in

Parkinson’s disease.

Page 4: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

iv

Preface

All of the work presented was conducted at the Centre for Applied Neurogenetics (CAN),

part of the Djavad Mowafaghian Centre for Brain Health (CBH) at the University of British

Columbia. CAN was established by Dr. Matthew Farrer (Principal Investigator). Study and

experimental approaches were approved by the University of British Columbia Ethics Board.

UBC Research Ethics (H10-02191) and ethics certificate (#5885 – 13) for Disease penetrance of

LRRK2 Gly2019Ser parkinsonism, LRRK2 G2019S disease penetrance modifiers and

Clinicogenetic studies of LRRK2 G2019S in Tunisia was obtained.

All manuscripts published or in preparation have been written under the guidance of Dr.

Matthew Farrer. I collected all genetic data and performed clinical and genetic analysis with

contributions from collaborators (Dr. Jan Aasly, Dr. Faycel Hentati, Dr. Suzanne Lesage, Dr.

Alexis Brice, Dr. Tatiana Foroud, Dr. Rick Myers), graduate student (Emil Gustavsson), and

committee members (Dr. Angie Brooks-Wilson, Dr. Denise Daley, Dr. Carolyn Brown).

Chapter 1: Parts of this chapter has been published as a review: Trinh et al (2013) Advances in

the genetics of Parkinson disease, Nature Neurology Reviews. All tables and figures have been

adapted and added on from Trinh et al 2013.

Chapter 2: Parts of chapter contains published data from Trinh et al (2014) Disease penetrance of

late-onset Parkinson disease, JAMA Neurology and Trinh et al (2014) and LRRK2

parkinsonism in Tunisia and Norway: A comparative analysis of disease penetrance (2014)

Neurology. Published work was done through collaborations with Dr. Jan Aasly from Trondheim

University and Dr. Faycel Hentati from Tunis Neurologie institute.

Page 5: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

v

Chapter 3: Parts of chapter contains published data Trinh et al (2013) A comparative study on

LRRK2 parkinsonism. Neurobiology of Aging. The collection of data and questionnaires was

funded by the Michael J Fox Foundation. Conference abstract: A comparative study of LRRK2

G2019S parkinsonism and idiopathic Parkinson’s disease in Tunisia. 3rd World Parkinson

Congress. October 1-4, 2013. Montreal, Canada. Conference abstract: Identification of LRRK2

p.G2019S disease modifiers. 62nd Annual Meeting of The American Society of Human

Genetics, November 6-10, 2012 in San Francisco, California.

Chapter 4: Written as a manuscript: DNM3 modifies age of onset in LRRK2 parkinsonism.

Lancet Neurology. 2015 (accepted). Conference abstract: DNM3; a genetic modifier of LRRK2

parkinsonism. 64th Annual Meeting of The American Society of Human Genetics . October 18-

22 2014, San Diego, California, USA.

Chapter 5: Section 1 has been used in a DFG grant, project title “Reduced penetrance in

hereditary movement disorders: elucidating mechanisms of endogenous disease protection”.

For the use of article, figures and tables, all copyright permissions have been obtained through

journal publishers.

Page 6: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

vi

Table of contents

Abstract ........................................................................................................................................... ii

Preface............................................................................................................................................ iv

Table of contents ............................................................................................................................ vi

List of tables ................................................................................................................................... ix

List of figures ................................................................................................................................. xi

List of abbreviations .................................................................................................................... xiii

Glossary of terms ........................................................................................................................ xvii

Acknowledgments........................................................................................................................ xix

1. Chapter 1: Introduction ....................................................................................................... 1

1.1. General features of Parkinson disease ........................................................................ 1

1.1.1. Motor features ..................................................................................................... 1

1.1.2. Non-motor features ............................................................................................. 2

1.1.3. Pathology ............................................................................................................ 2

1.2. Identification of genetic mutations in PD ................................................................... 3

1.2.1. Linkage analysis.................................................................................................. 3

1.2.2. Next generation sequencing ................................................................................ 4

1.2.3. Genome-wide case-control association............................................................... 5

1.3. Genes implicated in late-onset autosomal dominant PD ............................................ 6

1.3.1. SNCA .................................................................................................................. 6

1.3.2. LRRK2 ................................................................................................................ 7

1.3.3. MAPT ................................................................................................................. 8

1.3.4. EIF4G1 ................................................................................................................ 9

1.3.5. VPS35 and DNAJC13....................................................................................... 10

1.3.6. CHCHD2........................................................................................................... 11

1.3.7. Recessively inherited gene mutations ............................................................... 12

1.4. GWAS in PD............................................................................................................. 13

1.5. Neurobiological interactions: is there one pathway for PD? .................................... 20

1.6. Reduced penetrance .................................................................................................. 23

2. Chapter 2: Disease penetrance estimates of mutations in late-onset PD .......................... 25

2.1. Introduction: penetrance estimates ........................................................................... 25

Page 7: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

vii

2.2. Methods..................................................................................................................... 28

2.3. Results ....................................................................................................................... 29

2.3.1. SNCA: description of duplications, triplication and point mutations ............... 29

2.3.2. LRRK2 penetrance findings between populations ........................................... 36

2.3.3. Other autosomal dominantly-inherited mutations in familial PD ..................... 37

2.4. Discussion ................................................................................................................. 40

3. Chapter 3: A clinical comparison between LRRK2 parkinsonism and idiopathic PD ..... 53

3.1. General clinical features of LRRK2 parkinsonism ................................................... 53

3.2. Methods..................................................................................................................... 54

3.2.1. Motor symptom assessment .............................................................................. 54

3.2.2. Non-motor symptom assessment ...................................................................... 55

3.2.3. Genetic assessment and statistical analysis....................................................... 56

3.2.4. Michael J Fox Foundation (MJFF) database storage ........................................ 56

3.3. Results ....................................................................................................................... 60

3.3.1. Motor features ................................................................................................... 60

3.3.2. Non-motor features ........................................................................................... 68

3.3.3. Disease progression .......................................................................................... 74

3.4. Discussion ................................................................................................................. 75

4. Chapter 4: Dynamin 3 modifies age at onset in LRRK2 parkinsonism ............................ 79

4.1. Introduction ............................................................................................................... 79

4.2. Methods..................................................................................................................... 79

4.2.1. Discovery cohort and replication series ............................................................ 79

4.2.2. Linkage analysis and STR genotyping ............................................................. 80

4.2.3. Genome-wide SNP genotyping and association ............................................... 81

4.2.4. Whole genome sequencing and imputation ...................................................... 82

4.2.5. Sequencing and genotyping .............................................................................. 83

4.2.6. Brains, RNA, ampliseq transcriptome, antibodies ............................................ 83

4.3. Results ....................................................................................................................... 85

4.3.1. Linkage and association of LRRK2 p.G2019S families ................................... 85

4.3.2. Higher resolution mapping ............................................................................... 86

4.3.3. DNM3 expression in brain ................................................................................ 87

Page 8: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

viii

4.3.4. Replication cohorts ........................................................................................... 88

4.4. Discussion ................................................................................................................. 88

5. Chapter 5: Elucidating mechanisms of reduced penetrance in Mendelian disease ........ 113

5.1. The importance of reduced penetrance ................................................................... 113

5.2. Factors that influence penetrance............................................................................ 114

5.3. Methods and approaches to identify genetic modifiers .......................................... 116

5.4. Dynamin 3 as potential therapeutic target of LRRK2 parkinsonism ...................... 118

5.5. Conclusion .............................................................................................................. 119

References ................................................................................................................................... 121

Page 9: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

ix

List of tables

Table 1. Phenotypes associated with genes implicated in late-onset Lewy body PD ................... 15

Table 2. Selected genome-wide association studies in Parkinson disease.................................... 19

Table 3. Estimates of LRRK2 p.G2019S age-associated cumulative incidence ........................... 27

Table 4. Summary of patients included for each mutation into penetrance estimates .................. 31

Table 5. Demographics of unrelated patients and control subjects .............................................. 58

Table 6. Demographics of patients with a family history of parkinsonism within 1o .................. 59

Table 7. Clinical summary of patients .......................................................................................... 61

Table 8. Parkinsonism in LRRK2 p.G2019S carriers by gender ................................................. 62

Table 9. UPDRS Part IA Mentation, Behaviour and Mood ......................................................... 63

Table 10. UPDRS Part IB Mentation, Behaviour and Mood ........................................................ 64

Table 11. UPDRS Part II Activities of Daily Living .................................................................... 65

Table 12. UPDRS Part III ............................................................................................................. 66

Table 13. UPDRS Part IV Complications of Therapy .................................................................. 67

Table 14. Autonomic dysfunction (SCOPA-Aut) individual scores ............................................ 69

Table 15. Summary of autonomic assessments compared between LRRK2 parkinsonism and iPD

....................................................................................................................................................... 71

Table 16. Summary of cognitive assessment compared between iPD, LRRK2 parkinsonism and

control subjects ............................................................................................................................. 72

Table 17. Comparison of sleep scales among LRRK2 parkinsonism and iPD.............................. 73

Table 18. Rate of disease progression associated with age at onset in patients ............................ 74

Table 19. Demographics of discovery cohorts: Tunisian Arab-Berber LRRK2 p.G2019S carriers

....................................................................................................................................................... 96

Page 10: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

x

Table 20 . Demographics of LRRK2 p.G2019S carriers: replication series ................................. 97

Table 21. Demographics of healthy control brains for expression analysis ................................. 98

Table 22. Primer pairs and custom TaqMan probe design for different DNM3 transcript isoforms

in human striatum ......................................................................................................................... 99

Table 23. PLINK association underneath linkage regions.......................................................... 100

Table 24. DNM3 haplotypes associated with AAO.................................................................... 101

Table 25. DNM3 transcript levels correlate with LRRK2, VPS35 and SYNJ1 expression in striatal

tissue transcriptome data from normal controls (n=17). ............................................................. 102

Table 26. Sensitivity analysis for different age cut-offs on chromosome 1q23.3-24.3 using non-

parametric linkage ....................................................................................................................... 103

Page 11: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

xi

List of figures

Figure 1. Neurobiological Interactions between implicated genes for PD ................................... 22

Figure 2. Kaplan-Meier survival curves for SNCA mutations. .................................................... 33

Figure 3 Kaplan-Meier survival curves for SNCA ....................................................................... 35

Figure 4. Population-specific penetrance estimates of LRRK2 p.G2019S mutations. ................. 38

Figure 5. Kaplan-Meier survival curves for LRRK2 mutations. .................................................. 38

Figure 6. Kaplan-Meier survival curves for VPS35, EIF4G1 and DNAJC13 mutations. ............ 39

Figure 7. Comparison of SNCA and LRRK2 mutations. ............................................................. 45

Figure 8. Cumulative Incidence of SNCA triplication carriers. ................................................... 45

Figure 9. Cumulative Incidence of SNCA duplication carriers. ................................................... 46

Figure 10. Cumulative Incidence of LRRK2 p.N1437H carriers. ................................................ 46

Figure 11. Cumulative Incidence of LRRK2 p.R1441C carriers. ................................................. 47

Figure 12. Cumulative Incidence of LRRK2 p.R1441G carriers. ................................................ 47

Figure 13. Cumulative Incidence of LRRK2 p.Y1699C carriers. ............................................... 48

Figure 14. Cumulative Incidence of LRRK2 p.G2019S carriers. ................................................. 48

Figure 15. Cumulative Incidence of Ashkenazi Jewish LRRK2 p.G2019S carriers. ................... 49

Figure 16. Cumulative Incidence of Tunisian Arab-Berber LRRK2 p.G2019S carriers. ............. 49

Figure 17. Cumulative Incidence of Norwegian LRRK2 p.G2019S carriers. .............................. 50

Figure 18. Cumulative Incidence of EIF4G1 p.R1205H carriers. ................................................ 50

Figure 19. Cumulative Incidence of VPS35 p.D620N carriers. ................................................... 51

Figure 20. Cumulative Incidence of DNAJC13 p.N855S carriers................................................ 51

Figure 21. World map with LRRK2 mutations ............................................................................ 52

Figure 22. Chromosome 1 linkage peak ....................................................................................... 94

Page 12: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

xii

Figure 23. Age-associated cumulative incidence of LRRK2 p.G2019S carriers. ........................ 95

Figure 24. Whole genome sequencing and imputation workflow .............................................. 104

Figure 25. A schematic of the thirteen dynamin isoforms. ......................................................... 105

Figure 26. Multipoint model-based and non-parametric linkage analysis of Tunisian Arab-Berber

LRRK2 p.G2019S families. ........................................................................................................ 107

Figure 27 Chromosome 1 Q-Q plot values ................................................................................. 108

Figure 28. DNM3 transcript levels normalized by geometric mean of housekeeping genes ...... 109

Figure 29. Dynamin 3 protein levels normalized by GAPDH .................................................... 110

Figure 30. Dynamin 3 staining in cortical neurons ..................................................................... 111

Figure 31. Flow diagram of discovery and replication cohorts .................................................. 112

Page 13: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

xiii

List of abbreviations

AAO Age at onset

ABCA7 ATP-Binding Cassette, Sub-Family A (ABC1), Member 7

AD Alzheimer’s disease

ALS Amyotrophic lateral sclerosis

AOO Age of onset

APOC3 Apolipoprotein C-III

APOE Apolipoprotein class E

APP Amyloid Beta (A4) Precursor

ATP13A2 ATPase Type 13A2

BACE1 Beta-site amyloid precursor protein cleaving enzyme 1

BLBD Brainstem Lewy body disease

C9orf72 Chromosome 9 open reading frame 72

CF Cystic fibrosis

CFTR Cystic fibrosis transmembrane conductance regulator

CHCHD2 Coiled-coil-helix-coiled-coil-helix domain containing 2

CI Confidence interval

CRF Clinical research forms

CNV Copy number variation

DaT Dopamine transporters

DJ1 Protein deglycase peptidase C56 family

DLB Dementia with lewy bodies

DLBD Diffuse Lewy body disease

DNAJC13 DnaJ (Hsp40) Homolog, Subfamily C, Member 13

DNA Deoxyribonucleic acid

DNM3 Dynamin 3

DYT1 Torsion dystonia-1

EIF4G1 Eukaryotic Translation Initiation Factor 4 Gamma, 1

ESP Exome sequencing project

ET Essential tremor

Page 14: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

xiv

ExAC Exome aggregation consortium

F-DOPA Fluorodopa

FTD Frontotemporal dementia

GAK-DGKQ Cyclin G-associated kinase/ Diacylglycerol Kinase loci in GWAS

GAPDH Glyceraldehyde-3-phosphate dehydrogenase

GCH1 GTP cyclohydrolase 1

GDS Geriatric depression scale

GRS Genetic risk score

GSK GlaxoSmithKline

GTP Guanosine-5’-triphosphate

GWAS Genome-wide association studies

HD Huntington disease

hLOD heterogeneity LOD

HPRT Hypoxanthine phosphoribosyl-transferase

HTT Huntingtin

HWE Hardy-Weinberg equilibrium

IBD Identity by descent

IBS Identity by state

IPD Idiopathic PD

IPSC Induced human pluripotent stem cells

LD Linkage disequilibrium

LDL Low-density lipoprotein

L-dopa Levodopa

LOD Logarithm of odds

LRRK2 Leucine-rich repeat kinase 2

MAF Minor allele frequency

MAPT Microtubule-Associated Protein Tau

MDS-UPDRS Movement disorders society unified Parkinson disease rating scale

MJFF Michael J Fox Foundation

MMSE Mini-Mental State Examination

MOCA Montreal Cognitive Assessment

Page 15: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

xv

MSA Multiple system atrophy

NBIA Neurodegeneration with brain iron accumulation

NPC1L1 Niemann-Pick C1-Like 1

NPL Nonparametric linkage

NUCKS1 Nuclear Casein Kinase And Cyclin-Dependent Kinase Substrate 1

OR Odds Ratio

PCSK9 Proprotein convertase subtilisin/kexin type 9

PD Parkinson disease

PINK1 PTEN-induced putative kinase 1

PLA2G6 Phospholipase A2, Group VI

PRIMA Preferred Reporting Items for Systematic Reviews and Meta-analyses

PRKN Parkin; official name PARK2

RAB7L1 Member of the RAS Oncogenefamily; also known as RAB29

REM Rapid eye movement

REP1 Dinucleotide repeat sequence in promoter of SNCA

RIN RNA integrity number

RNA Ribonucleic acid

RME-8 Receptor mediated endocytosis 8

SCOPA-AUT Scales for Outcomes in Parkinson’s disease – Autonomic

SGCE Sarcoglycan, Epsilon

SLC41A1 Solute carrier family 41 magnesium transporter, member 1

SLC45A3 Solute carrier family 45, member 3

SNCA alpha-synuclein

SNP Single nucleotide polymorphism

STR Short tandem repeats

SYNJ1 Synaptojanin 1

SYP Synaptophysin

TDP TAR DNA-binding protein

THAP1 THAP Domain Containing, Apoptosis Associated Protein 1

TLBD Transitional Lewy body disease

TMEM175 Transmembrane protein 175

Page 16: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

xvi

TOR1A Torsin family 1, member A (torsin A)

UPDRS Unified parkinson disease rating scale

VCF Variant calling file

VPS26 Vacuolar protein sorting 26

VPS29 Vacuolar protein sorting 29

VPS35 Vacuolar protein sorting 35

WES Whole exome sequencing

WGS Whole genome sequencing

Page 17: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

xvii

Glossary of terms

1000 Genomes First project to sequence the genomes of a large number of people to

provide a comprehensive resource on human genetic variation

(www.1000genomes.org )

Apoptosis Process of programmed cell death

Bradykinesia Slowness of movement

ClinVar A freely accessible, public archive of reports of the relationships among

human variations and phenotype with supporting evidence

(www.ncbi.nlm.nih.gov/clinvar/)

dbSNP Database of single nucleotide polymorphisms

(www.ncbi.nlm.nih.gov/SNP)

DYT1 Dystonia caused by mutations in TOR1A

ENCODE The Encyclopedia of DNA Elements

(www.genome.ucsc.edu/ENCODE/)

Exome Part of the genome formed by exons, sequences which when

transcribed remain within the mature RNA after introns are removed by

RNA splicing

Familial With family history

Idiopathic Disease with unknown pathogenesis

Imprinting Epigenetic phenomenon by which certain genes are expressed in a

parent-of-origin-specific manner

Levodopa Drug treatment for symptoms of Parkinson disease

Low frequency variant Changes in the genome that deviates from the reference that is 1-5%

minor allele frequency in the general population

Modifiers Potential factors that influence the disease phenotype

Monogenic Inheritance of a phenotype controlled by one gene

Mutation Change in the genome that is rare <1%.

PLINK Free, open-source whole genome association analysis toolset, designed

to perform a range of basic, large-scale analyses in computationally

efficient manner

Page 18: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

xviii

(http://pngu.mgh.harvard.edu/~purcell/plink.index.shtml)

Polymorphism Common change in the genome (minor allele frequency >5%

Postural instability Disabling sign of Parkinson disease influenced by balance

Rare variant Changes in the genome that deviates from the reference that is <1%

minor allele frequency in the general population

Rigidity Stiffness of the body

Sporadic Disease with unknown pathogenesis and no family history

Transcriptomics Study of the transcriptome: the complete set of RNA transcripts that are

produced by the genome under specific circumstances or in a specific

cell, using high throughput technologies such as microarray analysis

Tremor Involuntary quivering movement

Variant Changes in the genome that deviates from the reference

Page 19: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

xix

Acknowledgments

I would like to thank the many patients and their families who volunteered, and the

longitudinal efforts of the clinical teams involved. Initial studies in Tunisia on familial

parkinsonism were in collaboration with Lefkos Middleton, Rachel Gibson and the

GlaxoSmithKline PD Programme Team (2002-2005). Subsequent clinical and molecular genetic

analysis was supported through Mayo Foundation, GlaxoSmithKline and National Institutes of

Health. The Michael J Fox Foundation generously supported clinical studies of LRRK2

p.G2019S in Tunisia and subsequent whole genome sequencing (2008-2011), Canada Excellence

Research Chairs program, CIHR/IRSC 275675 (2010-2017) and the Don Rix BC Leadership

Chair in Genetic Medicine. Replication series were made possible through the support of the

France-Parkinson Association, the Roger de Spoelberch Foundation (R12123DD), the French

program “Investissements d’avenir” (ANR-10-IAIHU-06), the Research Council of Norway,

Reberg’s legacy, the Norwegian Parkinson Foundation, Parkinson’s Study Group (PSG)

PROGENI Investigators.

I would like to thank members of my thesis committee, Dr. Angie Brooks-Wilson, Dr.

Denise Daley, Dr. Carolyn Brown, for their mentorship and a positive research environment. The

critical questions have allowed me to refocus and enhance this dissertation. Most importantly,

they have helped me mature into a more independent scientist.

A special thanks to Dr. Matthew Farrer for his knowledge, intellect and supervision.

Thank you for believing in this project and my abilities. The opportunities you have given me

have been helpful for my academic career. I would like to thank my closest friend, Dr. Carles

Vilarino-Guell for his ongoing intellectual and emotional support. I wouldn’t have been able to

do this without you. Matt and Carles, I will always look up to you and your scientific values. I

Page 20: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

xx

would also like to thank all the lab members at the Centre for Applied Neurogenetics, especially

Emil Gustavsson and Jas Khinda for their love and humor.

I am deeply grateful for the graduate scholarships/financial support I received from the

Canadian institutes of health reesearch (CIHR), UBC Faculty of Medicine (FoM), UBC Four

year fellowship (4YF), Michael Smith Foreign Exchange Supplement (MSFSS), James Miller

committee, Genome BC (LEEF), and the Simons Foundation.

Lastly, I would like to thank my mom, Linda Ninh, my dad, David Trinh, my uncle Thien

(Tommy) Ha Trinh and my sister, Angel Trinh for their unconditional love and encouragement. I

dedicate my thesis dissertation to my sister, Angel Trinh, who has been through a difficult battle

with neurological complications. You have motivated me in every way throughout my graduate

career. I owe an immense debt to my family who has sacrificed so much to give me every

opportunity to pursue my passions and fulfill my ambitions.

Page 21: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

1

1. Chapter 1: Introduction

1.1. General features of Parkinson disease

1.1.1. Motor features

Parkinson disease (PD) is the most common neurodegenerative movement disorder with

age-related prevalence (Bower, Maraganore, McDonnell, & Rocca, 1999). The mean age of

onset is 70 years although 4% of patients develop early-onset disease before the age of 50

(Schrag & Schott, 2006). Approximately 1% of the population is affected at 65 years, increasing

to 4–5% in 85-year-olds (de Lau & Breteler, 2006). The burden to patients, families, caregivers

and society is increasing steadily with population aging and the increased proportion of ‘baby

boomers’ aging.

Parkinsonism is characterized clinically by motor dysfunction; a triad of resting tremor,

bradykinesia, rigidity and postural instability (Fahn, 2003; L. W. Ferguson, Rajput, Muhajarine,

Shah, & Rajput, 2008). Initially, the symptoms are insidious and typically asymmetric, and most

patients suffer an inexorable decline. In diagnosed subjects ‘tremor-dominant’, ‘akinetic-rigid’,

or ‘mixed’ subtypes may dominate. However, an individual’s age-at-onset, disease course or

subsequent co-morbidities are difficult to predict. (Burn et al., 2006; L. W. Ferguson, et al.,

2008) A beneficial response to levodopa drug therapy (which treats the clinical motor features)

may remain late into the disease. However, with disease progression, optimizing the treatment to

patients is challenging and generally requires increased dosing. Side effects include troubling

‘on/off’ motor fluctuations and peak-dose dyskinesias (uncontrolled hyperkinetic movements).

(Fahn, 2000) The progressive loss of dopaminergic innervation to the striatum may be confirmed

using several imaging modalities, including DaTscan,18

F-DOPA positron emission tomography,

and via metabolic changes in brain glucose utilization and blood flow.

Page 22: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

2

1.1.2. Non-motor features

Non-motor features of PD include autonomic (constipation, cardiac denervation,

impotence, orthostatic hypotension and seborrhea), cognitive (bradyphrenia, cognitive decline

and dementia), psychiatric (depression, apathy, hallucinations and delusions), sensory problems

(hyposmia, anosmia and pain) and sleep disorders (REM sleep behavior disorder and excessive

daytime somnolence). (Chaudhuri, Healy, Schapira, & National Institute for Clinical, 2006)

These symptoms may be problematic long before the onset of movement disorder and are

difficult to treat. For a neuropathologic diagnosis of PD there must be evidence of neuronal loss

in the substantia nigra pars compacta accompanied by Lewy body pathology (alpha-

synucleinopathy; brainstem (BLBD) or more transitional Lewy body disease (TLBD)) in

surviving neurons. (Braak et al., 2003; Goedert, Spillantini, Del Tredici, & Braak, 2013a, 2013b;

Spillantini et al., 1997) With revised clinical criteria, the majority of patients with probable PD

that come to autopsy are now confirmed pathologically (Goedert, et al., 2013a). Fatigue is

difficult to treat and an important problem by patients. Similar to sleep disturbance, fatigue is

also almost universal in patients with PD (Alves, Wentzel-Larsen, & Larsen, 2004; Brown,

Dittner, Findley, & Wessely, 2005).

1.1.3. Pathology

A clinical diagnosis of dementia with Lewy bodies (DLB) is associated with much more

extensive cortical and limbic Lewy pathology and pathologically defined as diffuse Lewy body

disease (DLBD), that is often but not invariably associated with parkinsonism (Goedert, et al.,

2013a). Similarly, multiple system atrophy (MSA) has parkinsonism and prominent

dysautonomia but is characterized pathologically by glial cytoplasmic alpha-synuclein inclusions

Page 23: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

3

(Fellner, Wenning, & Stefanova, 2015; Koga et al., 2015). More sparse or ‘incidental’ Lewy

body pathology is often found in the healthy aged. Conversely, and more rarely, patients with

parkinsonism and clinically atypical ‘Parkinson-plus’ syndromes may have tauopathy (such as

post-encephalytic parkinsonism, cortico-basal degeneration, progressive supranuclear palsy

(Golbe, 1999; Papapetropoulos et al., 2005) and the parkinsonism-dementia complex of Guam

(lytico-bodig)), (Forman et al., 2002) ubiquitin or TDP-43 (TAR DNA-binding protein)

proteinopathy (such as Perry syndrome) (Tsuboi et al., 2008) or have non-specific findings such

as nigral neuronal cell loss with gliosis (including rapid-onset dystonia-parkinsonism, X-linked

dystonia-parkinsonism (Lubag) (Waters et al., 1993).

In PD, genetic mutations can now inform a diagnosis, disease-modeling and basic/pre-

clinical research. However, as the rare may inform the general, the text is also punctuated with

references to molecular findings from Parkinson-plus syndromes. Past discoveries, especially

within monogenic families, appear to coalesce about three interconnected processes: 1) synaptic

transmission (exo-, endocytosis) and endosomal receptor sorting and recycling; 2) lysosomal-

autophagy, and; 3) mitochondrial quality control and stress response. The emerging synthesis

may provide a unified molecular foundation for hypothesis-testing, pharmaceutical development

and future trials aimed at disease modification, not only symptomatic relief.

1.2. Identification of genetic mutations in PD

1.2.1. Linkage analysis

Linkage analysis methods were theoretically developed in the 1950-70s, but applied in

thel late 1990s and early 2000 to analyze rare traits influenced by a major variant or strong

genetic effects in families. Linkage analysis is a study of genetic markers and recombination in

Page 24: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

4

families with disease. Traditionally, microsatellites were used as genetic markers for linkage.

These include polymorphic sequences of DNA that are characterized by repeated sequences.

They can be repeats of 2(dinucleotide), 3(trinucleotide), 4(tetranucleotide), which makes them

highly heterozygous. The probability of genetic markers segregating with disease within families

is calculated and represented as a logarithm of odds (LOD) score (Dawn Teare & Barrett, 2005).

Two genetic loci are linked if transmitted from parent to offspring. This concept is used to

identify variants that segregate with a disease phenotype in a family. The LOD score is the

function of the recombination fraction, the higher the LOD score the higher the evidence of

cosegregation (of disease marker and phenotype). When a significant linkage region is identified

(LOD > 3.0) fine-mapping and sequencing underneath the linkage peak is often pursued to

elucidate the causal variant (Dawn Teare & Barrett, 2005). There are now many linkage marker

sets publicly available (DeCode, Marshfield resources). There are multiple methods for linkage.

One is the ‘parametric’ model which requires estimation of disease penetrance, mode of

inheritance (i.e. is it dominant or recessive), disease marker allele frequencies. Often times, these

allele frequencies are taken from population studies and inheritance is estimated from the

pedigree information. Parametric linkage uses identity by descent within a family. However, a

combined score across families is possible. Another is the non-parametric or ‘model-free’

linkage which does not require an input of inheritance and penetrance estimates. In PD, linked

regions to disease were often given a “PARK” locus designation. For example, before the gene

was identified, the region containing LRRK2 was named “PARK8”.

1.2.2. Next generation sequencing

Whole exome or genome sequencing is another approach to identify causal variants

segregating with disease. This involves massive parallel sequencing of ‘short-sequences’ that are

Page 25: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

5

aligned to a reference genome through computational methods. Variants that deviate from the

reference genome are identified and called. However, this leads to many variations which may or

may not have an impact on disease. Some more challenges involve the inability to detect

differences in repetitive elements, other large repeat expansions. In the case of exome

sequencing, only coding variations are captured and the initial hypothesis includes only protein-

coding variation (non-synonymous, deletions, frameshifts and loss of function mutations). The

advantages of looking at exome sequencing include the ability to determine pathogenic impact

through annotation and available protein crystal structures. Large publicly-available datasets can

be used as references. The ‘ExAC’ database (exome aggregation consortium) is one such

database to determine potential pathogenicity of a variant of interest. Over 60,000 individuals

have been exome sequenced in this consortium, which allows a reliable estimation of frequency

in the general population. Many prediction tools have allowed us to estimate how amino-acid

changes influence the folding. On the other hand, non-coding regulatory regions may also

contribute to disease pathogenesis. These regions are not covered in exome sequencing.

Although whole-genome sequencing databases are soon to be available, the annotation in these

regions is not as informative as coding regions.

1.2.3. Genome-wide case-control association

Genome-wide association studies test common variants’ association with common

disease. Linkage detects segments of inheritance within pedigrees, and association detects alleles

whose presence is correlated with a trait. Association makes use of linkage disequilibrium (the

likelihood of two markers traveling together in a population), thus association takes into account

the identity-by-state status rather than the identity-by-descent. Linkage analysis can localize 5-

10cM but association extends less than 1cM. There are two main types of genetic association:

Page 26: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

6

one is the population-based and the other family-based. Population-based association compares

genetic polymorphisms across case and controls. Genetic polymorphisms are variations in the

genome which can be common (frequency>5%), low (frequency<5% and >1%) or rare

(frequency<1%). Most recently, testing for association in GWAS is at an upwards of 500,000-5

million markers. Family-based association investigates transmission disequilibrium of alleles

through pedigrees. This method is not as commonly used, since it is easier to obtain independent

cases rather than families. Also case-control design was demonstrated to have greater power than

family based designs, provided the disease allele is common (Risch & Merikangas, 1996) . This

analysis has been a seminal driver for case-control designs (Risch & Merikangas, 1996) . Allelic

and genotypic frequencies between cases and controls are compared and the expected

contribution of these genetic polymorphisms are low (effect sizes or odds ratios = 1-1.5).

1.3. Genes implicated in late-onset autosomal dominant PD

A summary of pathogenic mutations and genes implicated in late-onset autosomal

dominant PD is described in Table 1.

1.3.1. SNCA

SNCA encodes for protein a-synuclein. Single nucleotide polymorphisms (SNPs) in

SNCA are found to be associated with PD across multiple ethnic populations: Caucasian,

Japanese, Tunisian Arab Berbers. A mega-meta GWAS of PD has replicated and shown SNCA

to have the most robust effect (Nalls, Pankratz, Lill, Do, Hernandez, Saad, DeStefano, Kara,

Bras, Sharma, Schulte, Keller, Arepalli, Letson, Edsall, Stefansson, Liu, Pliner, Lee, Cheng,

International Parkinson's Disease Genomics, et al., 2014). There is association of the promoter

(REP1) in the 5’ end of SNCA. REP1 associated SNPs also influence transcription of SNCA.

Page 27: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

7

However, the functional consequence of other associations within intron 4 and the 3’ end of

SNCA has yet to be discovered.

Many pathogenic mutations within SNCA have been found, traditionally through linkage

analysis and fine mapping. The first mutation identified was SNCA p.A53T (Polymeropoulos et

al., 1997). Since then, the field has identified more pathogenic point mutations such as p.A30P,

p.E46K, p.H50Q, p.G51D and copy number variations such as duplications and triplications

(Lesage, Anheim, Letournel, Bousset, Honore, Rozas, Pieri, Madiona, Durr, Melki, Verny,

Brice, et al., 2013; Trinh & Farrer, 2013). Alpha-synuclein is a key component of Lewy body

inclusion. Patients with these missense variations have predominantly DLBD pathology. In

addition to DLBD, duplication and triplication carriers have prominent nigral and hippocampal

neuronal loss. SNCA copy numbers lead to earlier onset and more fulminant LBD and dementia

is a prominent clinical feature. SNCA mutations are overall rare and p.A53T seems to be the

most frequent one.

1.3.2. LRRK2

LRRK2 (Leucine-rich repeat kinase 2) mutations confer the highest genotypic risk for

PD. Thus far, there are six pathogenic mutations identified in LRRK2: p.N1437H,

p.R1441C/G/H, p.Y1699C, p.G2019S, p.I2020T. LRRK2 p.G2019S is especially frequent in PD

patients of Ashkenazi Jewish or North African Arab-Berber origin, accounting for 13% and 30%

of cases in these populations respectively. Common risk factors include p.R1628P and G2385R.

Genome-wide association studies have highlighted and replicated LRRK2 as an associated gene

for PD. The effect of LRRK2 in GWAS studies has been smaller than that of SNCA. Large-scale

genotyping and gene sequencing of LRRK2 have identified risk factors associated with PD. One

Page 28: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

8

example is LRRK2 p.G2385R as a risk factor in Asian populations. There’s also evidence of a

haplotype in LRRK2 that is inversely associated with PD and other Parkinson-plus syndromes,

suggesting that LRRK2 variants in cis or in trans can have different influences on PD risk

(Heckman, Elbaz, et al., 2014; Heckman, Schottlaender, et al., 2014; Heckman et al., 2013; Trinh

& Farrer, 2013; Trinh, Farrer, Ross, & Guella, 1993)

LRRK2 parkinsonism has pleomorphic pathology. At autopsy, patients with LRRK2

parkinsonism typically have Lewy body or neurofibrillary tangle pathology, with nigral neuronal

loss and gliosis in some cases, TDP-43 proteinopathies have also been observed. Pleomorphic

pathology can be evident even within families with the same mutation. Intracellular Lewy bodies

and Lewy neurities, by definition the pathological hallmark of PD, are largely comprised of

aggregated a-synuclein. Clinically, patients with mutations in LRRK2 closely resemble patients

with idiopathic PD.

1.3.3. MAPT

There are two major haplotypes for MAPT (Tau): H1 and H2. The ancestral haplotype

for MAPT involves a paracentric inversion spanning 1.5 Mb (Zody et al., 2008). The H1 allele is

overrepresented in patients(Skipper et al., 2004; Spillantini & Goedert, 2001). Importantly, the

most significant associations of MAPT H1 and an H1-subtype (H1c; defined by the major allele

of rs242557) in neurodegeneration are with progressive supranuclear palsy, corticobasal

degeneration and Parkinson–dementia complex of Guam. These disorders are rare forms of

parkinsonism defined by their primary neurofibrillary tangle pathologies consisting of

hyperphosphorylated 4R tau—a tau protein isoform with four microtubule-binding domains that

results from alternative gene splicing and inclusion of MAPT exon 10. Similar to SNCA and

Page 29: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

9

LRRK2, MAPT has been consistently associated with PD in GWAS studies. (Simon-Sanchez &

Gasser, 2015) Postmortem studies of Lewy body disease in which patients had a longitudinal

clinical diagnosis of PD have also observed a MAPT H1 association. The H1 association in

Alzheimer’s disease is not as compelling but there are mutations in MAPT segregating in

frontal-temporal dementia (FTD)(Rademakers, Cruts, & van Broeckhoven, 2004; Rademakers et

al., 2003; Roks et al., 1999).

1.3.4. EIF4G1

EIF4G1 encodes for eukaryotic translation initiation factor 4 gamma 1. A dominantly

inherited p.R1205H is linked to late-onset PD. Although seen in multiple families in the initial

paper, there is incomplete penetrance of the mutation. Support for EIF4G1 in PD remains

equivocal. Some studies have shown that the p.R1205H mutation is present in more controls than

patients in Iceland (N. Nichols, Bras, Hernandez, Jansen, Lesage, Lubbe, Singleton, et al., 2015;

Siitonen et al., 2013). The study assessed the relevance of EIF4G1 in a large cohort by imputing

the p.R1205H mutation. They found 76 icelandic subjects older than 65 years of age that carried

the mutation. Another study with the NeuroX chip assayed over 12,000 patients and controls and

found 5 control subjects carrying the p.R1205H mutation. The control subjects range between

68-75 years of age. This led the studies to conclude that p.R1205H is a benign variant. However,

imputation accuracy needs to be taken into account and the evidence for p.R1205H is still

arguable. Reduced penetrance of the mutation could be an explanation for the observed

asymptomatic carriers.

Exome sequencing was performed for the original French families affected with

Parkinson disease. Importantly, EIF4G1 p.R1205H remains the only mutation identified in

Page 30: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

10

chromosome 3q26 linked to parkinsonism . It has not been identified by the Exome Aggregation

Consortium (ExAC; Jan 13th 2015 release) of 60,706 subjects, that includes contributions for the

1000 Genomes and NHLBI-GO Exome Sequencing Project (ESP). Of course this does not rule

out non-coding variation that is in close proximity to p.R1205H and could be the causal

mutation.

1.3.5. VPS35 and DNAJC13

VPS35 p.D620N causes autosomal dominantly inherited parkinsonism. The mutation

segregated in a Mendelian fashion. (Vilarino-Guell et al., 2011; Zimprich et al., 2011) The

mutation was found in large multi-incident families through exome sequencing. The families do

not share haplotypes and the mutation seems to have arisen de novo. Thus far, VPS35 p.D620N

is extremely rare and other pathogenic mutations in VPS35 have yet to be found. VPS26 and

VPS29 bind to VPS35 to form a functional retromer. However, variants identified thus far in

these genes do not seem to segregate with disease (Gustavsson, Guella, et al., 2015).

Nonetheless, VPS35 p.D620N has been independently replicated in large Dutch, French,

Japanese families and other sporadic patients has made a convincing case as a gene for PD

(Ando et al., 2012; Kumar et al., 2012; Lesage et al., 2012; Sharma et al., 2012) . VPS35 is also

implicated in other neurodegenerative diseases. Haploinsufficiency in VPS35 increases the

neuropathology with AD and the protein conforms to the spatiotemporal model of AD(Small et

al., 2005; Wen et al., 2011) . VPS35 could regulate Abeta peptide levels(Small, et al., 2005). The

retromer sorts cargo from endosomes in all cell types. VPS35 is expressed in axons and dendrites

of neurons, involved in retrograde tracking of APP, BACE1 and is important in plasma

membrane trafficking (Bhalla et al., 2012; Steinberg et al., 2013) . VPS35 may have neuron-

Page 31: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

11

specific functions: there is evidence that overexpressing VPS35 is neuroprotective to

dopaminergic neurons (Bi, Li, Huang, & Zhou, 2013).

DNAJC13 p.N855S has been found in one large Mennonite kindred by exome

sequencing. However, there is a phenocopy and unaffected carriers in the family. The mutation

needs to be independently replicated in PD. In the meantime, DNAJC13 p.N855S mutations have

also been found to be implicated in Essential Tremor (ET). Other variants in DNAJC13 besides

p.N855S are extremely rare and there is lack of segregation analysis done in families thus far

(Gustavsson, Trinh, et al., 2015; Rajput et al., 2015; Vilarino-Guell et al., 2014)

Both DNAJC13 and VPS35 are the first genes found to be implicated in PD through next

generation sequencing and open new methods for the application of exome and whole genome

sequencing in mapping genes for disease. Since the discovery of VPS35, genes in early-onset

parkinsonism have also been identified by exome sequencing. For example, SYNJ1 mutations

seem to segregate with early-onset disease in some families (Olgiati et al., 2014; Quadri et al.,

2013) . SYNJ1 was first discovered through homozygosity mapping and exome sequencing in an

Italian consanguineous family with parkinsonism and dystonia (Quadri, et al., 2013). However,

this thesis will focus on autosomal dominant Parkinson disease rather than early-onset Parkinson

disease/ Parkinson-plus syndrome.

1.3.6. CHCHD2

CHCHD2 (full name is coiled-coil-helix-coiled-coil-helix domain containing 2) has been

implicated in late-onset autosomal PD. CHCHD2 p.T61I was described in two large Japanese

families with autosomal dominant PD (Funayama et al., 2015). Furthermore, CHCHD2

p.R145Q, and 300+5G>A were also identified in other smaller Japanese families with autosomal

Page 32: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

12

dominant PD. There is no evidence of common variants in CHCHD2 being associated with PD.

Further sequencing revealed rare exonic mutations with unknown significance in LBD patients:

the majority of these rare variants were located within the gene’s mitochondrial targeting

sequence (Ogaki et al., 2015).

CHCHD2 contains cysteine-x9-cysteine motifs that are important for regulating enzymes

in the mitochondrial respiratory chain. Mutations in Parkin and PINK1 (PTEN-induced

putative kinase 1) in juvenile/early-onset parkinsonism are important in the mitochondria

respiratory chain. PINK1 knock-outs show reduced mitochondrial ATP synthesis (Grunewald et

al., 2009; Pilsl & Winklhofer, 2012; Rakovic et al., 2011; Vos, Verstreken, & Klein, 2015).

Thus, CHCHD2 is functionally compelling and replication of these three mutations in other

families is warranted.

1.3.7. Recessively inherited gene mutations

Recessively inherited mutations (homozygous or compound heterozygous loss of

function) have also been identified by linkage analysis in parkin (PARK2;PRKN) (Cookson et

al., 2003; Mata et al., 2005; Tan et al., 2003; West, Lockhart, O'Farell, & Farrer, 2003), PTEN-

induced putative kinase 1 (PINK1) (Ishihara-Paul et al., 2008; Lee et al., 2009; Toft et al., 2007)

and DJ-1 (PARK7) (Lockhart, Bounds, et al., 2004; Lockhart, Lincoln, et al., 2004; Maraganore

et al., 2004) , albeit clinical syndromes with juvenile (≤20 years at diagnosis) or early-onset

disease (≤45 years at diagnosis). While the majority of cases that have come to autopsy suffer

neuronal loss without Lewy body pathology there are noteworthy exceptions in compound

heterozygotes (Farrer et al., 2001; Samaranch et al., 2010) . PRKN loss-of-function may explain

Page 33: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

13

~15% of early-onset cases and the majority (~50%) in which there is a family history of

parkinsonism and/or parental consanguinity, albeit without Lewy pathology.

Early-onset parkinsonism accounts for <4% of PD in the community although it is more

frequently encountered in movement disorders neurology clinics. Recessively inherited

mutations have been also implicated in rare, rather atypical Parkinson-plus disorders including

Kufor-Rakeb syndrome due to mutations in ATP13A2, neuroaxonal dystrophy due to loss of

PLA2G6, (Morgan et al., 2006; Paisan-Ruiz, Washecka, Nath, Singleton, & Corder, 2009) and

neurodegeneration with brain iron accumulation (NBIA) due to mutations PANK2, C2orf37,

C19orf12, FA2H and WDR45 (Gregory & Hayflick, 2011; Haack, Hogarth, Gregory, Prokisch,

& Hayflick, 2013; Haack et al., 2012)

1.4. GWAS in PD

Genetic association study (GWAS) looks to find alleles that are observed more often than

expected by chance in individuals with a trait of interest than those without. There are many

strengths in this approach and GWAS have made important contributions in the scientific field.

Most notably, in neurodegeneration, the APOE association was identified for Alzheimer’s

disease (AD) (Harold et al., 2009; Lambert et al., 2013) . The APOE allele had a large and robust

effect on AD. The SNCA signal in PD is the most robust across populations in GWAS.

Furthermore, many genes that have been linked (identified through families) have also been

nominated in GWAS. A basic summary of associated genes found from GWAS is present in

Table 2. SNCA, LRRK2, GCH1 are a few genes that harbor genetic risk factors and pathogenic

mutations segregating in families (Nalls, Pankratz, Lill, Do, Hernandez, Saad, DeStefano, Kara,

Bras, Sharma, Schulte, Keller, Arepalli, Letson, Edsall, Stefansson, Liu, Pliner, Lee, Cheng,

Page 34: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

14

International Parkinson's Disease Genomics, et al., 2014; Simon-Sanchez et al., 2011). GWAS

findings can be followed up with additional sequencing to identify genetic variants of

pathogenicity. For example, loss-of function (LOF) mutations in ABCA7 in patients with AD

have been identified. In fact, an ABCA7 LOF mutation segregated in late-onset autosomal

dominant AD families (Cuyvers et al., 2015; Hollingworth et al., 2011) .

Like many studies, there are also caveats to GWAS. One main problem is difficulty in

pin-pointing the real associated genes and/or functional variants.The PARK16 locus contains

five genes (SLC45A3, NUCKS1, RAB7L1, SLC41A1) (Trinh, Vilarino-Guell, & Ross, 2015;

Vilarino-Guell et al., 2010). RAB7L1 seems to be the most studied and most compelling

candidate. RAB7L1 has been shown to co-immunoprecipitate with VPS35 and LRRK2 (D. A.

MacLeod et al., 2013). RAB7L1 seems to interact with common variants in LRRK2 to modify

risk. However, the effect of RAB7L1 is different across populations. Thus, elucidating the real

functional variant for RAB7L1 is challenging. Another example is the GAK-DGKQ locus has

also been nominated by GWAS and the genomic region consists of three genes. GAK is the most

interesting, due to its involvement in clathrin-mediated endocytosis. Even if the locus points to

one gene of interest, there may be multiple risk variants to consider. There are multiple variants

associated in SNCA and the effect between two variants very close together is difficult to

distinguish because they co-segregate during inheritance. However, each variant can alter

expression levels of SNCA differently. A novel strategy to identify such functional variants is

with human pluripotent stem cells (IPSC) and genome editing techniques. Through

CRISPR/Cas9, a common PD-associated risk variant in a non-coding distal enhancer element

(located in intron 4) was found to regulate the expression of α-synuclein (Soldner et al., 2016).

Page 35: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

15

Table 1. Phenotypes associated with genes implicated in late-onset Lewy body PD

Disease

OMIM

identifier

Gene Mutations Age at

onset

(range)

Synopsis of

clinical

features

Predominant

pathology

References

Dominantly inherited late-onset PD

168601 SNCA Missense:

Ala30Pro,

Glu46Lys,

His50Gln,

Gly51Asp,

Ala53Thr

60 years

(30–80)

Levodopa-

responsive

parkinsonis

m

Diffuse

LBD

(Kruger et

al., 1998;

Lesage,

Anheim,

Letournel,

Bousset,

Honore,

Rozas, Pieri,

Madiona,

Durr, Melki,

Verny, &

Brice, 2013;

Polymeropo

ulos, et al.,

1997;

Proukakis et

al., 2013;

Zarranz et

al., 2004)

605543 SNCA Locus

duplication

(and

triplication)

31–71 years

(24–48)

Levodopa-

responsive

parkinsonis

m, cognitive

decline,

autonomic

dysfunction

and

dementia;

progression

more rapid

in SNCA

triplication

cases

Diffuse

LBD, with

prominent

nigral and

hippocampal

(CA2–3)

neuronal

loss

(Chartier-

Harlin et al.,

2004; J.

Fuchs et al.,

2008;

Ibanez et al.,

2004;

Nishioka,

Wider, et

al., 2010;

Singleton et

al., 2003)

607060 LRRK2 Missense:

Asn1437His,

Arg1441Cys/

Gly/His,

Tyr1699Cys,

Gly2019Ser,

Ile2020Thr

60 years

(32–79)

Levodopa-

responsive

parkinsonis

m consistent

with

sporadic

PD;

Brainstem

LBD,

neurofibrilla

ry tangle or

TDP-43

pathology

and/or nigral

(Paisan-

Ruiz, Lang,

et al., 2005;

Ross et al.,

2011;

Zimprich et

al., 2004)

Page 36: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

16

Disease OMIM

identifier

Gene Mutations Age at onset

(range)

Synopsis of clinical

features

Predominant pathology

References

Common

polymorphis

ms:

Ala419Val,

Arg1628Pro,

Gly2385Arg

(Asia)

Protective

haplotype:

Asn551Lys–

Arg1398His

Lys1423Lys

occasionally

dystonia,

amyotrophy,

gaze palsy

and

dementia

neuronal

loss

614203 VPS35 Missense:

Asp620Asn

53 years

(40–68)

Tremor-

dominant

levodopa-

responsive

parkinsonis

m,

dyskinesia

and

dystonia,

occasionally

dementia

Inconclusive

, possibly

without

LBD

(Ando, et

al., 2012;

Kumar, et

al., 2012;

Nuytemans

et al., 2013;

Sheerin et

al., 2012;

Vilarino-

Guell, et al.,

2011;

Zimprich, et

al., 2011)

616361 DNAJC13 Missense:

Arg855Ser

67 years

(57.5-76.5)

Slowly

progressive,

late-onset

asymmetric

parkinsonis

m, good

response to

L-dopa.

Lewy body

inclusions in

carriers and

also

DNAJC13

staining

within these

inclusions

(Vilarino-

Guell, et al.,

2014)

616244 CHCHD2 Missense:

Thr61Ile

55.5 years

(48-61)

Typical

parkinsonso

nian features

(bradykinesi

a, rigidity,

gait). L-

dopa

responsive

NA: Post-

mortem yet

to be tested

for Lewy

body

inclusions

(Funayama,

et al., 2015)

Page 37: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

17

Disease OMIM

identifier

Gene Mutations Age at onset

(range)

Synopsis of clinical

features

Predominant pathology

References

Juvenile and early-onset recessively inherited parkinsonism

600116 PARK2 Numerous

missense,

exon

deletion and

duplication

mutations

<45 years

(12–58)

Levodopa-

responsive

parkinsonis

m, often

juvenile and

typically

slowly

progressive

Predominant

ly nigral

neuronal

loss,

occasionally

with

synuclein or

tau

pathology

(Kitada et

al., 1998)

605909 PINK1 Missense:

Gln129X,

Gln129fsX1

57,

Pro196Leu,

Gly309Asp

Trp437X,

Gly440Glu,

Gln456X

Rare: locus

and exon

deletion

Typically

<45 years

(18–56)

Levodopa-

responsive

parkinsonis

m, often

akinetic

with

postural

instability/g

ait

disturbance

with slow

progression;

sleep benefit

One case

with LBD

(Ishihara-

Paul, et al.,

2008;

Samaranch,

et al., 2010;

Valente,

Abou-

Sleiman, et

al., 2004;

Valente,

Salvi, et al.,

2004)

606324 DJ-1 Missense:

Glu163Lys,

Leu166Pro

Exon 1–5

deletion,

g.168–

185dup

<40 years

(24–39)

Levodopa-

responsive

parkinsonis

m,

psychologic

al and

behavioural

disturbances

,

amyotrophy

and

cognitive

impairment

Unknown (Annesi et

al., 2005;

Bonifati et

al., 2003)

Page 38: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

18

Disease OMIM

identifier

Gene Mutations Age at onset

(range)

Synopsis of clinical

features

Predominant pathology

References

606693 ATP13A2 Missense:

Phe182Leu,

Gly504Arg,

Gly877Arg,

1019GfsX10

21

Exon 13

1306+5G>A

Exon 16

22-bp

deletion

<20 years

(10–33)

Levodopa-

responsive

atypical

parkinsonis

m associated

with

supranuclear

gaze palsy,

spasticity

and

dementia

Neuroradiol

ogical

atrophy with

iron

accumulatio

n in basal

ganglia

(Di Fonzo et

al., 2007;

Ramirez et

al., 2006)

Abbreviations: fs, frameshift; LBD, Lewy body disease; OMIM, Online Mendelian Inheritance in Man;

PD, Parkinson disease; X, stop codon.

Page 39: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

19

Table 2. Selected genome-wide association studies in Parkinson disease

Gene Chromosome Population

SNCA 4q21 USA, UK, France, Japan

MAPT 17q21.1 USA, UK, France

LRRK2 12q12 USA, Japan

HLA-DRA 6q21.3 USA, UK

GAK–DGKQ 4p16 USA, UK

PARK16 1q32 USA, UK, Japan

BST1 4p15 France, USA

Page 40: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

20

1.5. Neurobiological interactions: is there one pathway for PD?

Many genes implicated in PD are expressed in the endosomes, synaptic vesicle sorting

and recycling and membrane curvature (Figure 1). Alpha-synuclein is important at the

presynaptic terminals and promotes exocytosis. Alpha-synuclein has roles in membrane

curvature and is expressed in endosomes, multi-vesicular bodies and lysosomes. There is

evidence that a-synuclein is also involved in endocytosis with dynamins during clathrin-

mediated endocytosis (Vargas et al., 2014). At the post-synapse (medium spiny neurons),

LRRK2 is involved with endocytosis by phosphorylating endophilin A at S75 (Matta et al.,

2012). Activation of LRRK2 and PINK1 (recessive mutations cause early-onset parkinsonism)

phosphorylate Rab family GTPases(Lai et al., 2015; Steger et al., 2016). An unbiased phospho-

proteomics approach identified Rabs with a pThr73 autophosphorylation site (Rab3 , Rab8 and

Rab 10) as LRRK2 substrates in vitro. Furthermore, Rab8A, Rab8B and Rab13 are indirectly

phosphorylated by PINK1 (Lai, et al., 2015) . Loss of Rab39B causes early-onset

parkinsonism(Wilson et al., 2014). Rabs are important for vesicular trafficking and cellular

compartmentalization(Clague & Rochin, 2016).

There is also evidence that suggests LRRK2 regulates chaperone-mediated autophagy,

microtubule stabilization, mitochondria and Golgi pathways. LRRK2 co-immunoprecipitates

with VPS35 and RME-8 (DNAJC13), and is involved in actin polymerization(Munsie et al.,

2015). VPS35 is part of the retromer, formed with VPS26 and VPS29. The retromer complex

mediates cargo recognition of early endosomes and membrane recruitment. VPS35 mediates

recycling from endosomes to the Golgi apparatus. LRRK2, VPS35 and RME-8 directly mediate

endosomal protein sorting and recycling, including the delivery of synaptic neurotransmitter

receptors and lysosomal proteins to either degradation or endosome to membrane recycling.

Page 41: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

21

VPS35 has been shown to interact with eIF4G1 in yeast to modulate alpha-synuclein

toxicity(Dhungel et al., 2015) .

Perhaps impairment in synaptic vesicle trafficking and recycling is central to the

pathophysiology of PD. When this process is perturbed, cargo retention in the endosome lead to

the formation of multivesicular bodies that are destined to fuse with lysosomes for exosomal

release. Cell-to-cell transmission of alpha-synuclein proteinopathy has been a highlight in recent

research (Luk, Kehm, Carroll, et al., 2012; Luk, Kehm, Zhang, et al., 2012; Wang et al., 2012)

and this may be a consequence of cargo retention and synaptic vesicle trafficking/recycling.

Page 42: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

22

Figure 1. Neurobiological Interactions between implicated genes for PD

Key molecular processes in neurons for important genes implicated in PD. Dopaminergic neuron

is in green, glutamatergic cortical neuron in blue and medium spiny neuron is in yellow.

Page 43: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

23

1.6. Reduced penetrance

It has been almost 20 years since the discovery of the first SNCA mutation in familial

PD. Before the discovery of genes implicated in PD, many scientists had thought that PD was

caused by environmental factors. Genetics have been extremely informative in the biology of

PD, which could lead to new therapies that could help all those with the disease. However,

treatment to prevent symptom onset or delay progression has yet to be developed.

Interestingly, large numbers of putative pathogenic mutation carriers are free of disease:

there is evidence of reduced penetrance in patients carrying known pathogenic mutations.

Penetrance is formally known as conditional probability of being affected with a disease given a

genotype. Penetrance can be age-dependent and may even border ‘variable expressivity’ in very

subtle disease manifestations (expressivity describes the extent a certain phenotype manifests).

Within genomic research, reduced penetrance has been neglected and is now emerging as a new

field of research. The identification of genetic, environmental, lifestyle and biological factors

influencing the phenomena of reduced penetrance is of great interest in neurodegeneration. The

idea behind discoveries of ‘protective’ genetic factors can help developing relevant therapeutic

targets to halt the development of disease. In large-scale 1000 Genomes and ExAC projects,

many pathogenic mutation carriers have been identified. For example, there are 47 LRRK2

p.G2019S mutation carriers in the ExAC database that are potentially asymptomatic, although

these individuals are not well phenotyped (exac.broadinstitute.org). In fact, an average genome

has 150 sites of protein truncating variants, 10-12,000 sites with protein-altering variants and

even up to 30 mutations implicated in rare disease (Auton et al., 2015) . Thus, penetrance

estimates may be more reduced than what is estimated in literature. Recognizing the potential of

disease modifiers or protective factors has already sparked research initiatives such as the

Page 44: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

24

‘resilience project’ (resilienceproject.me) at Mount Sinai, ‘wellderly’ (scripps.org) at Scripps

amongst others. These projects focus on healthy, elderly individuals. Studying modifier genes in

Parkinson disease and other neurodegenerative disorders are more difficult as this requires much

more stringent phenotyping from neurologists with clinical expertise.

LRRK2 p.G2019S is the most common gene mutation in familial PD and accounts for the

highest attributable risk in PD. The high frequency of LRRK2 p.G2019S in North African Arab

Berbers and Ashkenazi Jewish populations give a larger sample size to discover genetic

modifiers that can influence penetrance. Although LRRK 2 p.G2019S parkinsonism is

considered a monogenic form of disease, the mutation is not fully penetrant. We hypothesize that

genetic factors can modulate phenoconversion of LRRK2 p.G2019S.We postulate that the novel

modifier genes and DNA variants that are identified will advance our understanding of the

biological mechanisms of LRRK2. Second, these genetic factors may prove to be useful

therapeutic targets that could be used to delay the onset of PD among those with LRRK2

mutations. Third, screening of these genetic variants could be included as part of LRRK2 genetic

testing and results provided as part of genetic counseling to yield better estimates of the likely

onset of PD for a particular at-risk individual.

There are three main studies in this thesis 1) comparative analysis of disease penetrance

of mutations implicated in late-onset autosomal dominant PD 2) detailed clinical analysis of

LRRK2 p.G2019S carriers compared to idiopathic PD 3) Identification of a potential age-at-

onset modifier in LRRK2 parkinsonism.

Page 45: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

25

2. Chapter 2: Disease penetrance estimates of mutations in late-onset PD

2.1. Introduction: penetrance estimates

There have been many pathogenic mutations identified for PD. However, these mutations are

not 100% penetrant. Penetrance is defined as the probability of individuals with a given genotype

who exhibit a certain phenotype. There are many methods to assess penetrance in age-associated

diseases. In late-onset autosomal dominant PD, the disease onset, progression, pathology and

clinical features of mutation carriers can be vastly distinct. For example, SNCA has been

associated with LOPD in every population tested (Nalls, Pankratz, Lill, Do, Hernandez, Saad,

DeStefano, Kara, Bras, Sharma, Schulte, Keller, Arepalli, Letson, Edsall, Stefansson, Liu, Pliner,

Lee, Cheng, Ikram, et al., 2014; Simon-Sanchez & Gasser, 2015). However, a penetrance

comparison of point mutations and copy number variations in SNCA has not been assessed in

detail. On the other hand, the penetrance of LRRK2 p.G2019S has been explored through

various case sampling and statistical analyses (Table 1) (Healy et al., 2008; Marder et al., 2015;

Trinh, Amouri, et al., 2014; Trinh, Guella, & Farrer, 2014) . The two main methods that have

been used to assess the penetrance of LRRK2 p.G2019S are cumulative incidence plots and kin-

cohort. Some studies have inferred genotypes within families (Marder, et al., 2015). The

penetrance varies between 10-50% at age 60 (Table 3). The statistical analyses used are also

quite variable. The first study on LRRK2 p.G2019S published an age-dependent penetrance

within families and derived an estimation by a simple equation (proportion of affected/total

carriers) (Kachergus et al., 2005) . Kachergus et al report at age 50 the LRRK2 p.G2019S

mutation is 17% penetrant and at age 70 it is 85%. A world-wide consortium of LRRK2 carriers

in Europe have reported a similar estimation age 59-79 (28-74%). This was further replicated in

Page 46: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

26

Tunisian Arab Berbers (Hulihan et al., 2008) . Interestingly, the Ashkenazi Jewish LRRK2

carriers and Italian LRRK2 carriers had a more reduced estimation ranging from 15-32%.

Page 47: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

27

Table 3. Estimates of LRRK2 p.G2019S age-associated cumulative incidence

Ethnicity Sample Statistical Analysis Age range

(penetrance)

Reference

Norwegian,

American (United

States), Irish and

Polish

13 LRRK2

families

22 familial

affected

carriers

Proportion of

affected/total carriers

50-70 (17-85%) (Kachergus,

et al., 2005)

French and North

African families

2 LRRK2

families

6 familial

affected

carriers

Not reported 55-76 (33-100%)

(Lesage et

al., 2005)

Ashkenazi Jews 2975 familial

relatives of

459 probands

Kin-cohort (Wacholder

et al., 1998)

Relatives were not

genotyped for mutation:

probability of carrying

mutation was estimated

60-80 (12-24%)

(Clark et

al., 2006)

Ashkenazi Jews 22 affected

carriers

Penetrance calculated

from odds ratio

Lifetime risk = 35% (Ozelius et

al., 2006)

Italian (UK

Parkinson’s

Disease Brain

Bank)

36 familial

affected

carriers

Kaplan Meier 60-80 (15-32%)

(Goldwurm

et al., 2011;

Goldwurm

et al., 2007)

World-wide

(mostly

European)

133 LRRK2

families

327 affected

members

Kaplan Meier 59-79 (28-74%)

(Healy, et

al., 2008)

Page 48: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

28

2.2. Methods

In this study, we have created a large meta-analysis of published literature on disease onset

and clinical phenotypes of late-onset autosomal dominant Parkinson disease. Published literature

was included if there was information on ethnicity, mutation, age and age-at-onset/first motor

symptom and confirmation of mutation (not inferred). If age-at-onset or age at last contact were

not available in the published literature, we requested information from corresponding authors.

We also excluded autosomal recessive genes. Preferred Reporting Items for Systematic Reviews

and Meta-analyses (PRIMA) guidelines were followed (Vrabel, 2015) . Literature search

involved keywords: SNCA point mutation, SNCA duplication, VPS35, EIF4G1, LRRK2,

DNAJC13, Parkinson disease, autosomal dominant, late-onset parkinsonism. Published studies

were included if they have the following information: 1) ethnicity of patient or unaffected

Ethnicity Sample Statistical Analysis Age range

(penetrance)

Reference

Arab-Berber 72 affected

carriers

Kaplan Meier 60-80 (50-100%)

(Hulihan, et

al., 2008)

European

countries, mainly

Italy

154 first

degree

relatives and

190 second

degree

relatives of

10 probands

with

p.G2019S

Kin-cohort (Wacholder,

et al., 1998)

*No relatives were

genotyped for mutation:

probability of carrying

mutation was estimated

1st degree

60-80 (12-33%)

2nd

degree

60-80 (10-30%)

(Goldwurm,

et al., 2011)

Northern Spain

(Cantabria)

32 carriers Kaplan Meier 60-80 (12-47%)

(Sierra et

al., 2011)

Page 49: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

29

individual; 2) confirmation of mutation (not inferred); 3) age of patient or unaffected individual;

4) age of onset of patient; 5) gender of the patient or unaffected individual; 6) first motor

symptom of patient, and;7) non-motor symptom of patient .

When age of onset, age, gender and age-at-last contact data was not available in the

published article, information was requested through the corresponding authors. If this

information was not obtainable, the study subjects were excluded. We also excluded articles:1)

about autosomal recessive parkinsonism; 2) that reported duplicate data; 3) that were not written

in English, and; 4) genes for which significant genetic linkage was not reported.

The age-associated cumulative incidence (disease penetrance) was estimated using a Kaplan-

Meier method with age-at-onset as the time variable; asymptomatic carriers were right censored

at the age-at-last contact or age-at-death (JMP software, SAS Institute Inc., Cary, NC). Statistical

comparisons between survival curves were done with log-rank tests unless otherwise stated.

2.3. Results

2.3.1. SNCA: description of duplications, triplication and point mutations

SNCA harbors both copy number variation and point mutations in patients with PD.

Clinically, SNCA triplication carriers have an earlier onset, faster progression and more

fulminant disease compared with duplication carriers. These findings from duplication carriers

are more closely comparable to typical late-onset PD. SNCA triplication and duplication carriers

seldom have dementia. However, the frequency of SNCA mutation carriers are extremely rare

and thus, clinical comparisons are difficult to interpret.

We assessed the cumulative incidence of SNCA copy number and point mutation carriers.

Penetrance of triplications was higher than duplications and point mutations (log rank p<0.01)

Page 50: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

30

Penetrance of triplications had a lower quartile of 31 years, median of 39 years and upper

quartile of 46 years (Figure 2). Point mutations had a lower quartile of 42 years, median of 49

years and upper quartile of 60 (n=59). Point mutations were comparable to duplications (log rank

p=0.97) which had a lower quartile of 40 years, median of 48 years and upper quartile of 61

years (n=41). We observed penetrance differences in point mutations. However, the sample

sizes were too small for a meaningful interpretation. SNCA p.A53T (n=35) had a mean age-at-

onset of 45.9 years; p.A30P (n=5) had a mean age-at-onset 59.8 years; p.E46K had a mean age at

onset of 62.3 years; p.H50Q (n=3) had a mean age-at-onset 64.7 years and p.G51D (n=3) had a

mean age-at-onset of 32.7 years (Figures 3a-f). A summary of patients included for each

mutation is described in Table 4.

Cumulatively, SNCA mutations (triplications, duplications, point mutations) had an earlier

onset age (mean age at onset 38.5-49.5 years) compared to LRRK2 mutations (mean age at onset

46.8-68.8 years). Unlike SNCA, two copies of the mutant allele (i.e. homozygous G2019S

mutations) do not confer significantly higher risk or higher penetrance .

Page 51: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

31

Table 4. Summary of patients included for each mutation into penetrance estimates

Mutation Carriers

included (n)

Affected

carriers (n)

Unaffected

carriers (n)

Ethnic

backgrounds

References

SNCA point

mutation

47 43 4 Greek,

British,

Korean,

Polish,

Swedish,

English

(Golbe,

1999)

SNCA

duplications

41 39 2 French,

Korean,

Italian

(J. Fuchs et

al., 2007; J.

Fuchs, et al.,

2008;

Nishioka et

al., 2009)

SNCA

triplications

15 15 0 Swedish-

American,

Japanese

(Farrer et al.,

2004;

Nishioka, et

al., 2009)

LRRK2

N1437H

10 9 1 Norwegian (Johansen,

White,

Farrer, &

Aasly, 2011)

LRRK2

R1441C

27 17 10 Norwegian (Haugarvoll

et al., 2008)

LRRK2

R1441G

104 62 42 Basque (Haugarvoll

& Wszolek,

2009; Marti-

Masso et al.,

2009;

Pchelina,

Ivanova,

Emel'ianov,

&

Iakimovskii,

2011; Ruiz-

Martinez et

al., 2010)

LRRK2

Y1699C

7 7 0 Norwegian (Khan et al.,

2005;

Zimprich, et

al., 2004)

LRRK2

G2019S

330 291 39 Norwegian,

Tunisian,

Ashkenazi Jewish

(Healy, et al.,

2008; Trinh,

Amouri, et al., 2014)

Page 52: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

32

Mutation Carriers

included (n)

Affected

carriers (n)

Unaffected

carriers (n)

Ethnic

backgrounds

References

LRRK2 I2020T 29 23 6 Japanese (Tomiyama

et al., 2006)

VPS35 D620N 61 54 7 German,

Tunisian,

Yemen Jews,

Japanese,

French

(Ando, et al.,

2012;

Lesage, et al.,

2012;

Sharma, et

al., 2012;

Sheerin, et

al., 2012;

Vilarino-

Guell, et al.,

2011;

Zimprich, et

al., 2011)

EIF4G1

R1205H

20 20 0 French,

Tunisian,

Yemen Jews

(Chartier-

Harlin et al.,

2011; N.

Nichols,

Bras,

Hernandez,

Jansen,

Lesage,

Lubbe, &

Singleton,

2015;

Nuytemans,

et al., 2013;

Siitonen, et

al., 2013)

DNAJC13

N855S

18 12 6 Mennonites-

Canadian

(Vilarino-

Guell, et al.,

2014)

Total

Combined

709 592 117

Page 53: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

33

Figure 2. Kaplan-Meier survival curves for SNCA mutations.

The probability of being affected at median age 56 is 0.90 for SNCA triplication carriers, 0.70

for SNCA duplication and missense carriers.

Page 54: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

34

A

B

C

Page 55: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

35

Figure 3 Kaplan-Meier survival curves for SNCA

a) cumulative incidence for all SNCA point mutation carriers, b) p.A30P, c) p.A53T, d) p.E46K,

e) p.G51D, f) p.H50Q. The dotted lines represent confidence intervals.

D

E

F

Page 56: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

36

2.3.2. LRRK2 penetrance findings between populations

The LRRK2 p.G2019S mutations account for up to 15% in Ashkenazi Jewish populations,

30% in Arab Berber populations and 1% in Caucasian populations. We assessed the penetrance

of LRRK2 in the Arab Berber population with an expanding a Tunisian cohort since 2005

(Figure 4) (Hulihan, et al., 2008). The Arab Berber population cumulative incidence and mean

age at onset estimates were consistent with previous studies: mean age at onset 57.1 years;

95%CI, 45.5-68.7 years n=220 (Healy, et al., 2008; Hulihan, et al., 2008). However, the

Norwegian penetrance estimates (mean age at onset 63 years; 95% CI 51.4-74.6 years) were

reduced compared to Tunisia (p<0.0001). Lastly, the Ashkenazi Jewish population from Israel

were comparable to Tunisia (mean age at onset 57.9 years, 95% CI, 54-63 years) (Figure 4).

Furthermore, there are six pathogenic mutations in LRRK2 (p.N1437H, p.R1441C/G/H,

Y1699C, p.G2019S, p.I2020T). Higher penetrance for p.N1437H and p.Y1699C mutations,

may reflect the small sample size (n=10). The cumulative incidence of LRRK2 mutations are

significantly different from each other. Penetrance within the kinase domain (p.G2019S and

p.I2020T) are similar and significantly higher than the Roc domain mutations (p.R1441C/G/H)

(Figure 5). The p.N1437H is also in the Roc domain but hampered by small sample size. The

cumulative incidence of LRRK2 p.I2020T had a lower quartile of 51 years or younger, a median

of 55 years of age, and an upper quartile of 60 years or older (n = 29). The estimation was similar

to LRRK2 p.G2019S, which had a lower quartile of 49 years or younger, a median of 57 years,

and an upper quartile of 67 years or older (n = 330). Lastly, the cumulative incidence of LRRK2

p.R1441C and p.R1441G were the least penetrant. LRRK2 p.R1441C had a lower quartile of 65

years or younger, a median of 71 years, and an upper quartile of 77 years or older (n = 27).

p.R1441G had a lower quartile of 60 years or younger, a median of 65 years of age, and an upper

Page 57: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

37

quartile of 72 years or older (n = 104).The LRRK2 p.I2020T mutation is largely Japanese,

R1441G is largely Basque, R1441C is mostly Belgian. These estimates could also reflect

diagnostic or referral differences across regions. Perhaps there is better clinical care for

neurodegenerative diseases in different parts of the wold which could reflect higher reporting of

disease and earlier age-at-onset estimates.

2.3.3. Other autosomal dominantly-inherited mutations in familial PD

VPS35 p.D620N (lower quartile ≤ 45years, median 49 years and upper quartile ≥ 59 years;

n= 61) was significantly more penetrant than EIF4G1 p.R1205H (lower quartile ≤ 56 years,

median 62 years and upper quartile ≥ 69.5 years; n=20) and DNAJC13 p.N855S (lower quartile

≤ 61 years, median 68 years and upper quartile ≥ 76 years; n=18) (Figure 6).

The age-dependent cumulative incidence was significantly different across mutations

(p<0.0001) . Overall SNCA triplication carriers (n=15) had the highest cumulative incidence

(penetrance) and LRRK2 p.G2019S carriers in Norway (n=84) had the lowest (Figure 7).

Page 58: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

38

Figure 4. Population-specific penetrance estimates of LRRK2 p.G2019S mutations.

Figure 5. Kaplan-Meier survival curves for LRRK2 mutations.

Page 59: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

39

Figure 6. Kaplan-Meier survival curves for VPS35, EIF4G1 and DNAJC13 mutations.

Page 60: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

40

2.4. Discussion

This study summarizes and systematically compares the age-dependent cumulative incidence

of all known mutations leading to late-onset parkinsonism. Fifteen rare pathogenic variations in

five genes (SNCA, LRRK2, VPS35, EIF4G1, and DNAJC13) were assessed. All mutation

carriers were combined, whether from the literature or contributed by corresponding authors,

providing the most accurate penetrance estimates to date. Nevertheless, the study has many

limitations. These include cultural and environmental differences between populations, access to

health care and ascertainment bias. Various diagnostic criteria have to be considered, and the

movement disorders neurology expertise at different centres . Moreover, age at onset is

retrospective and subjective, dependent upon a variety of symptoms and signs, although well

correlated with age at diagnosis (Reider et al., 2003) .

All comparisons utilized the same statistical measure to estimate cumulative incidence which

simplifies comparisons between mutations. The Kaplan-Meier method is a reverse survival curve

analysis, ideally suited for sporadic patients and unrelated probands that censors for

asymptomatic carriers. In contrast, the kin-cohort method excludes probands, employing just

relatives with inferred genotypes to specifically avoid inflating penetrance estimates. However, a

disadvantage is that the phenotypic and genotypic information of the relatives may be inaccurate.

While analyses have been adapted to compensate for a variety of study designs, Kaplan-Meier

and kin-cohort are the major methods employed in penetrance estimates of PD. Bias from the

inclusion of probands and family members has been assessed using a variety of statistical

methods and sensitivity analyses for LRRK2 p.R1441G and p.G2019S show comparable results

(Trinh, Amouri, et al., 2014; Trinh, Guella, et al., 2014) . The sensitivity analysis compared

penetrance estimates derived from different methods and sample groups (kin-cohort methods

Page 61: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

41

versus Kaplan meier as well as families versus unrelated patients). Herein we are limited by

published data, by the relatedness of subjects and the total number of carriers/families with each

gene. With these caveats acknowledged, confidence intervals are provided for genetic counseling

(Figure 7-20).

Penetrance estimates for monogenic parkinsonism vary by gene, by mutation and by

ethnicity. SNCA triplications are more penetrant than duplications for which genomic dosage has

been directly correlated to mRNA and protein expression (Farrer, et al., 2004; Nishioka, et al.,

2009) . Clinically, SNCA triplication carriers have an earlier onset, faster progression and more

fulminant disease compared to duplication carriers which more closely resembles late-onset

idiopathic PD (Muenter et al., 1998; Nishioka, Kefi, et al., 2010; Nishioka, et al., 2009) . Seldom

do SNCA triplications or duplication carriers have dementia as a first symptom; typically

cognitive decline is noted several years after the onset of parkinsonism. Nevertheless, many have

a clinical diagnosis of dementia with Lewy bodies (DLB), with diffuse Lewy body disease on

autopsy (DLBD). Overall SNCA point mutations and SNCA duplications are quite similar in

penetrance. While the majority of missense carriers of SNCA p.A53T have been described with

young onset parkinsonism, with an aggressive course (Golbe, 1999) , and most duplication

carriers are described as DLB, they are most comparable. The frequency of SNCA

multiplications and point mutations is extremely rare (less than 1% in different populations), thus

meaningful comparisons of clinical features is problematic, although a global study of SNCA

multiplication and missense carriers has recently been initiated (The Parkinson Progression

Markers Initiative by The Michael J Fox Foundation for Parkinson’s Research).

LRRK2 mutations confer the highest population-attributable risk to PD but the function

of the encoded protein still remains unclear. The majority of pathogenic variants are within three

Page 62: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

42

contiguous domains: kinase, ROC and COR (Mills, Mulhern, Liu, Culvenor, & Cheng, 2014) .

Penetrance of mutations within the kinase domain (LRRK2 p.G2019S and p.I2020T) are similar,

and significantly higher than ROC domain mutation (p.R1441C and p.R1441G). LRRK2

p.R1441C and p.R1441G mutations have similar penetrance estimates (p=0.31). The sample size

was too small to compare p.R1441H. However, we observe higher penetrance of LRRK2

p.N1437H, which could be hampered by the rarity of this variant (n=10). COR domain mutations

are highly penetrant, but could also be due to a smaller sample size (n=7).

SNCA mutations (triplications, duplications and point mutations) had a larger effect, with

an earlier onset (AAO means were from 38.5-49.5 years) compared to LRRK2 mutations (AAO

means were from 46.8 to 68.8 years) . SNCA mutation carriers have a more aggressive

phenotype whereas LRRK2 carriers have a more benign clinical course compared to idiopathic

PD. In LRRK2 parkinsonism, there is less REM sleep behavior disorder and gastrointestinal

dysfunction (Trinh, Amouri, et al., 2014) which are two main clinical features affected by Braak

staging. SNCA mutation carriers primarily have Lewy-body-like inclusions of α-synuclein

aggregates (Conway et al 2000 oligomerization SNCA, Wood et al alpha-synuclein 1999). In

contrast, LRRK2 carriers (albeit p.N1437H, p.R1441C/G/H, p.G2019S, or p.I2020T) have

pleiomorphic pathologies including α-synuclein, 4-repeat-tau, or tar-dna binding-43

proteinopathies on a background of neuronal loss and gliosis. The clinical course probably

reflects the burden and type of end-stage pathology.

Age at onset is only one measure of variability across these pathogenic mutations. The

pathology in LRRK2 mutation carriers are extremely pleiomorphic. Furthermore, clinical

features such as cognitive decline/dementia, autonomic dysfunction can vary between mutation

carriers. The LRRK2 p.Y1699C mutation is most unusual with amotrophy, dementia and not just

Page 63: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

43

parkinsonism, compared to LRRK2 p.I2020T which has typical parkinsonism which is

comparable to idiopathic PD and tauopathies (Hasegawa et al., 2009; Ujiie et al., 2012).

This study highlights the role of ethnicity or environmental factors as a major contributor

of penetrance. Stratification of LRRK2 p.G2019S parkinsonism by ethnicity was possible

because of the large sample size. Israeli Ashkenazi Jews have a significantly higher penetrance

compared to Norwegian LRRK2 p.G2019S mutation carriers, and are comparable in penetrance

to Tunisian Arab-Berbers. In New York, the disease in Ashkenazi Jewish carriers is less

penetrant (24% penetrance at age 80) (Figure 21); these differences may reflect a sample of 7

carriers, the exclusion of family members (Clark, et al., 2006) and environmental factors such as

orthodox or unorthodox practices. The study by Clark et al has now been further expanded to 90

LRRK2 carriers and include a much larger cohort of Ashkenazi LRRK2 G2019S in New York.

The reduced penetrance estimate still stands (26% at age 80 years). However, the statistical

method used was different and the sampling included more unaffected LRRK2 p.G2019S

carriers with family history (Marder, et al., 2015) . In contrast, similarities in age of onset

between Israeli Jews and Tunisian Arab-Berbers carriers may reflect similar genetic and

environmental backgrounds (Nebel et al., 2000) . The environment may also play a role in the

differences. Furthermore, ascertainment bias in the patients sample sets may influence the data.

These cohorts are predominantly tertiary referral clinic-based samples from specialist hospitals,

which means age-at-onset can be inflated as patients from the same family may be more aware of

symptoms. Nevertheless, ethnic differences are an important consideration in genetic counseling.

Mutations in SNCA, LRRK2, VPS35, EIF4G1 and DNAJC13 have been directly

implicated in familial parkinsonism (Trinh & Farrer, 2013) . These proteins are centrally

involved in synaptic transmission, early endosomal sorting and recycling, and lysosomal

Page 64: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

44

autophagy. Indeed, LRRK2, VPS35, and DNAJC13 directly immunoprecipitate with members of

the WASH complex (Helfer et al., 2013) , which regulates actin remodelling and membrane

trafficking in these processes. Whether this network is similarly perturbed in idiopathic PD has

yet to be established. Differences in the penetrance estimates may reflect the type of substitution,

its location and functional consequence. Mutations may affect interactions with binding partners

and downstream signaling pathways, thus influencing expression (transcript or protein), and

ultimately compensatory mechanisms (genetic and non-genetic).

Age is considered the greatest risk factor for PD and genetic susceptibility is only one

influence. The penetrance of mutations in late-onset parkinsonism is also dependent on ethnicity

and potentially environmental factors. Thus, heterogeneity between mutation carriers may be an

important consideration when identifying modifiers of disease. Prospective, longitudinal

evaluation of carriers and further meta-analyses will be required for more precise penetrance

estimates, and provide the opportunity to inform therapeutic trials.

Page 65: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

45

Figure 7. Comparison of SNCA and LRRK2 mutations.

SNCA mutations are illustrated as red lines and LRRK2 mutations are illustrated as black lines.

The cumulative incidence for distinct pathogenic mutations in each gene are shown in figure 2

and figure 4.

Figure 8. Cumulative Incidence of SNCA triplication carriers.

Dotted lines represent confidence intervals.

Page 66: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

46

Figure 9. Cumulative Incidence of SNCA duplication carriers.

Dotted lines represent confidence intervals.

Figure 10. Cumulative Incidence of LRRK2 p.N1437H carriers.

Dotted lines represent confidence intervals.

Page 67: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

47

Figure 11. Cumulative Incidence of LRRK2 p.R1441C carriers.

Dotted lines represent confidence intervals.

Figure 12. Cumulative Incidence of LRRK2 p.R1441G carriers.

Dotted lines represent confidence intervals.

Page 68: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

48

Figure 13. Cumulative Incidence of LRRK2 p.Y1699C carriers.

Dotted lines represent confidence intervals.

Figure 14. Cumulative Incidence of LRRK2 p.G2019S carriers.

Dotted lines represent confidence intervals.

Page 69: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

49

Figure 15. Cumulative Incidence of Ashkenazi Jewish LRRK2 p.G2019S carriers.

Dotted lines represent confidence intervals.

Figure 16. Cumulative Incidence of Tunisian Arab-Berber LRRK2 p.G2019S carriers.

Dotted lines represent confidence intervals.

Page 70: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

50

Figure 17. Cumulative Incidence of Norwegian LRRK2 p.G2019S carriers.

Dotted lines represent confidence intervals.

Figure 18. Cumulative Incidence of EIF4G1 p.R1205H carriers.

Dotted lines represent confidence intervals.

Page 71: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

51

Figure 19. Cumulative Incidence of VPS35 p.D620N carriers.

Dotted lines represent confidence intervals.

Figure 20. Cumulative Incidence of DNAJC13 p.N855S carriers.

Dotted lines represent confidence intervals.

Page 72: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

52

Figure 21. World map with LRRK2 mutations

Page 73: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

53

3. Chapter 3: A clinical comparison between LRRK2 parkinsonism and idiopathic PD

3.1. General clinical features of LRRK2 parkinsonism

PD is characterized by four cardinal signs: resting tremor with asymmetry at onset,

bradykinesia, rigidity, postural instability and positive response to Levodopa (Postuma et al.,

2015) . LRRK2 p.G2019S has the highest genotypic and population attributable risk. The

mutation was first shown to segregate with PD in a Norwegian family (Kachergus et al 2005).

Overall, the clinical presentation of idiopathic PD (iPD) is similar to LRRK2 parkinsonism

(Aasly et al., 2005) . However, there is heterogeneity in the cohorts, sample size and

methodology.

Some reports suggest a more severe phenotype in LRRK2 mutation carriers compared to

idiopathic PD. LRRK2 p.G2019S carriers were reported to have more severe motor symptoms

and dyskinesias (Nishioka, et al., 2009) (Oosterveld et al., 2015) . Depression, hallucinations,

sleep issues were reported to be more common in LRRK2 p.G2019S carriers (Pchelina, et al.,

2011) . Furthermore, postural instability and gait problems were more common in early-onset

LRRK2 carriers (Alcalay et al., 2009) (Alcalay et al., 2015; Marras et al., 2016) . On the other

hand, LRRK2 carriers have reported to have slower disease progression, less cognitive

impairment, lower depression, less autonomic dysfunction and UPDRS scores (Alcalay, et al.,

2009; Healy, et al., 2008) (Marras, et al., 2016) (Tijero et al., 2013) . A better characterization of

LRRK2 carriers is warranted.

Furthermore, collecting and analyzing a large database on clinical features of LRRK2

p.G2019S carriers can lead to a better understanding of the progression of the disease and study

of endophenotypes (both motor and non-motor). In the present study, we have expanded the

Page 74: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

54

Tunisian Arab-Berber LRRK2 cohort over a period of six years to compare the disease onset,

clinical symptoms and disease progression.

3.2. Methods

All patients were recruited at the same neurological centre: Mongi Ben Hamida National

Institute of Neurology, Tunis. The center provides out-patient and in-patient services for

neurological disorders in Tunisia. Local on-site monitoring was independently performed by

PRN clinical research (www.prnservices.co.uk) every 18 months. Clinical examinations were

performed and questionnaires were administered by movement disorder specialists Dr. Faycal

Hentati, Dr. Samia Ben Sassi, Dr. Fatma Nabli, Dr. Emna Farhat. Diagnoses of PD were made

according to the UK PD Society brain bank criteria. Enrollment information including additional

family medical history and origin was also collected. Patients and control subjects completed

standardized clinical research forms (CRFs), all medical history of patients and families were

recorded. Clinical data and blood samples were collected for 778 patients and 580 unaffected

subjects (Table 5-6).

3.2.1. Motor symptom assessment

Movement disorders society unified Parkinson disease rating scale (MDS-UPDRS) were

performed on patients with symptoms of parkinsonism. The MDS-UPDRS consists of four parts:

Non-motor experiences of daily living, motor experiences of daily living, motor examination and

motor complications (Goetz, Nutt, & Stebbins, 2008; Goetz et al., 2008). All assessments in the

MDS-UPDRS have five responses: 0=normal, 1=slight, 2=mild, 3=moderate, 4=severe. “Slight”

refers to symptoms with low frequency or intensity that have no impact on function, “mild” has

Page 75: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

55

modest impact on function, “moderate” has considerable impact on function and lastly, “severe”

refers to symptoms that prevent function. Medication status of L-dopa “on” and “off” stages

were recorded. Hoehn and Yahr staging (1-5), to objectively rate the patient’s disability at a

certain time (stage 0 means no signs of disease and stage 5 is wheelchair bound or bedridden),

was also recorded.

3.2.2. Non-motor symptom assessment

Multiple questionnaires were used for non-motor symptoms. The Schwab and England

questionnaire was used for activities of daily living (McRae, Diem, Vo, O'Brien, & Seeberger,

2000) . The rating was performed by the neurologist and ranges from 0% (only vegetative

functions are working, bedridden and helpless) to 100% (completely independent, able to do all

chores, no slowness or difficulty). Autonomic dysfunction was assessed with Scales for

Outcomes in Parkinson’s disease – Autonomic (SCOPA-AUT) (Visser, Marinus, Stiggelbout, &

Van Hilten, 2004). There are 25 items that assess gastrointestinal, urinary, cardiovascular,

thermoregulatory, and sexual dysfunction. Autonomic problems increase significantly with

disease severity (Visser, Marinus, Stiggelbout, et al., 2004; Visser, Marinus, van Hilten,

Schipper, & Stiggelbout, 2004) . Geriatric depression scale and Epworth daytime sleepiness

scale was used to assess depression and sleep function, respectively (Johns, 1991; Yesavage et

al., 1982). REM sleep behavior disorder was noted as 50% of all REM sleep disordered patients

by polysonography develop PD. A specially modified “sniffin’ test” was created with the help of

neurologist Dr. John Duda for odorant descriptors and distractors for the Arab Berber population.

“Sniffin tests” is a validated test consisting of odor detection, discrimination and sensitivity. It

was adopted a trial of 100 control subjects to be ‘culturally’ appropriate. Simplified, culturally

appropriate tests were created for largely illiterate population. Cognition was measured using the

Page 76: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

56

mini-mental state examination (MMSE), Montreal cognitive assessment (MOCA), six picture

test or frontal assessment battery. The latter two were adapted for a largely illiterate population.

3.2.3. Genetic assessment and statistical analysis

DNA was extracted by standard procedures (Miller et al 1988) and LRRK2 c.6055G>A

(p.G2019S) was genotyped with a TaqMan probe on an ABI7900 analyzer and then verified by

sequencing, as previously described (Hulihan, et al., 2008) . Patients with pathogenic mutations

in PINK1 or Parkin were excluded from this study (Bonifati, et al., 2003; Valente, Salvi, et al.,

2004).

Multivariate regression models were used to investigate and compare different

questionnaires and clinical assessments, adjusted for age at onset, disease duration, gender and

medical state (on or off levodopa) (JMP software version 10). Cumulative incidence was

assessed with Kaplan Meier or kin-cohort analysis and significant differences were detected with

either log-rank or Wilcoxon tests. The log-rank test gives equal weight to all time points in a

cumulative incidence plot, whereas the Wilcoxon test gives more weight to earlier time points

and requires one group consistently have a higher risk than the other.

3.2.4. Michael J Fox Foundation (MJFF) database storage

Each patient and control subjects had a clinical research form ID, linked with a MJFF

family ID and individual ID. The data was imported into under six categories: 1) enrollment, 2)

UPDRS, 3) medications, 4) non-motor, 5) cognitive testing, 6) environment and lifestyle. The

collected data was stored in a database under the LRRK2 cohort consortium (https://www.inn-

tunisia.com/) webpage titled “Parkinson’s disease in Tunisia”. However, the database is no

Page 77: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

57

longer maintained and is warehoused in Tunisia and UBC. It has also been submitted to the

MJFF LRRK2 cohort consortium to be made broadly accessible. However, it may be important

to make this publicly available.

Page 78: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

58

Table 5. Demographics of unrelated patients and control subjects

Non-LRRK2 p.G2019S LRRK2 p.G2019S carriers

Patients Controls Patients Controls

N 350 399 220 (38%) 12 (3%)

Number of men (%) 187 (53%) 203 (51%) 124 (56%) 6 (50%)

Mean age (SD) years 66.6 (12.9) 61.1 (11.1) 67.6 (12.6) 56.7 (10.9)

Median age (IQR) 69 (59–76) 59 (53–69) 69 (48–90)

54.5 (38–

72)

Mean age of onset (SD) 55.3 (14.4) - 57.1 (11.6) -

Median age at onset (IQR) 58 (46–66) - 57 (40–74) -

Mean disease duration 8.10 (5.2) 9.07 (5.03)

Range disease duration 2-23 3-23

Page 79: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

59

Table 6. Demographics of patients with a family history of parkinsonism within 1o

Affected

probands

Affected family

members

Unaffected family

members

N 162 126 169

Number of men (%) 89 (55%) 73 (58%) 74 (44%)

Mean age (SD) 67.0 (14.2) 76.6 (14.0) 59.4 (17.9)

Median age (IQR) 68 (46–90) 80 (66–95) 57 (26.5–87.5)

Mean age at onset (SD) 55.0 (14.0) 59.2 (14.4) -

Median age at onset

(IQR) 56 (38–74) 60 (42–79) -

LRRK2 p.G2019S

carriers 80 (49%) 46 (36%) 71 (42%)

Page 80: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

60

3.3. Results

3.3.1. Motor features

The motor features were largely indistinguishable between iPD or LRRK2 carriers. Age and

age-at-onset were similar between LRRK2 p.G2019S homozygotes (n=32), heterozygotes

(n=177) and idiopathic PD (n=324) (Table 7). Comparison of first symptoms between idiopathic

PD and LRRK2 carriers was similar. Tremor was the most predominant first symptom across all

genotypes (range from 71.9% to 81.5%) (Table 7). When stratifying by gender, the results

remain comparable across LRRK2 parkinsonism and iPD (Table 8).

Unified Parkinson disease rating scale (UPDRS) has also been assessed and comparisons

were performed with regression modeling. There were no remarkable differences between

LRRK2 parkinsonism and iPD (Table 7-13). However, early-onset LRRK2 carriers tend to suffer

more rigidity than late-onset carriers (UPDRS III rigidity score, p=0.05). Although this value is

not significant after Bonferroni correction.

Page 81: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

61

Table 7. Clinical summary of patients

LRRK2 p.G2019S carrier status Homozygous Heterozygous iPD p-values

N 32 177 324

Mean age of onset (SD) 54.5 (12.4) 57.3 (11.9) 56.2 (13.8) 0.46

Median age of onset (IQR) 56 (40–72) 58 (41–75) 58 (38–78)

First symptom (%)

Tremor 23 (71.9%) 135 (76.3%) 265 (81.5%) 0.53

Gait or balance deterioration 4 (12.5%) 23 (13.0%) 26 (8.0%) Muscle cramping or dystonia 2 (6.2%) 5 (2.8%) 7 (2.2%)

Shoulder stiffness 0 5 (2.8%) 8 (2.5%) Other 3 (9.4%) 7 (4.0%) 10 (3.1%)

NA 0 2 (1.1%) 8 (2.5%) PD phenotype (%)

Mixed 22 (69%) 101 (57%) 192 (59.1%) 0.29

Akinetic rigid 3 (9.4%) 34 (19.2%) 68 (21.0%)

Tremor dominant 6 (19%) 31 (17.5%) 62 (19.1%) NA 1 (3.1%) 11 (6.2%) 2 (0.6%)

Hoehn and Yahr (SD)

On (SD, n) 2.5 (0.9, 20) 2.1 (0.8, 70) 1.9 (1.0, 126) 0.21

Off (SD, n) 1.6 (0.9, 8) 2.3 (1.1, 84) 2.3 (0.9, 123) UPDRS-III score (SD)

On (SD, n)

34.0 (21.1,

20) 36.7 (16.4, 70) 33.3 (19.5, 128) 0.45

Off (SD, n) 45.2 (19.8, 8) 49.0 (21.9, 84) 47.4 (17.2, 123)

Page 82: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

62

Table 8. Parkinsonism in LRRK2 p.G2019S carriers by gender

LRRK2 p.G2019S iPD

Female Male Female Male

N 103 106 142 182

Mean age of onset (SD) 60.0 (12.0) 57.8 (12.0) 56.4 (13.4) 56.2 (14.1)

Median age of onset (IQR) 56 (40-72) 60 (43.5-

76.5)

58 (39.5-76.5) 58 (38-78)

First Symptom

Tremor 73 (70.9%) 85 (80.1%) 122 (85.9%) 143 (78.6%)

Gait or balance

deterioration 14 (13.6%) 13 (12.3%) 8 (5.6%) 18 (9.9%)

Muscle cramping or

dystonia 7 (6.8%) 0 4 (2.8%) 3 (1.6%)

Shoulder stiffness 1 (0.97%) 4 (3.8%) 2 (1.4%) 6 (3.3%)

Other 8 (0.78%) 2 (1.9%) 2 (1.4%) 8 (4.4%)

NA 0 2 (1.9%) 4 (2.8%) 4 (2.2%)

PD phenotype

Akinetic rigid 20 (19.4%) 17 (16.0%) 28 (19.7%) 40 (22.0%)

Mixed 58 (56.3%) 65 (61.3%) 84 (59.2%) 108 (59.3%)

Tremor dominant 17 (16.5%) 20 (18.9%) 29 (20.4%) 33 (18.1%)

NA 8 (7.8%) 4 (3.8%) 1 (0.70%) 1 (0.54%)

Hoehn &Yahr (SD)

On 2.3 (0.93)

(n=48)

2.0 (0.73)

(n=42)

1.9 (0.89)

(n=58)

2.0 (1.1)

(n=68)

Off 2.3 (1.2) (n=40) 2.2 (1.1)

(n=52)

2.5 (0.97)

(n=51)

2.1 (0.80)

(n=72)

UPDRS-III score (SD)

On 39.2 (19.4)

(n=48)

32.7 (14.4)

(n=43)

32.4 (18.2)

(n=60)

34.1 (20.7)

(n=68)

Off 50.2(21.2)

(n=40)

47.5(21.6)

(n=52)

51.6(16.9)

(n=51)

44.4(16.9)

(n=72)

NA = not available. Other = change in facial expression, change in speech or voice, decreased

dexterity, stooped posture or bradykinesia.

Page 83: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

63

Table 9. UPDRS Part IA Mentation, Behaviour and Mood

LRRK2 p.G2019S iPD p-value

Cognitive impairment 0.28 (0.67) 0.33 (0.74) 0.66

Hallucinations and psychosis 0.14 (0.45) 0.23 (0.57) 0.35

Depressed mood 1.28 (1.00) 1.40 (1.03) 0.08

Anxious mood 1.06 (1.11) 1.02 (1.07) 0.32

Apathy 1.22 (1.03) 1.19 (1.01) 0.53

Features of dopamine

dysregulation syndrome

0.05 (0.25) 0.06 (0.35) 0.95

Quantitative scales are from 0-4: 0 (normal) – 4 (most severe). Mean values (standard deviation)

are given.

Page 84: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

64

Table 10. UPDRS Part IB Mentation, Behaviour and Mood

LRRK2 p.G2019S iPD p-value

Sleep problems 0.96 (1.15) 0.87 (1.17) 0.30

Daytime sleepiness 0.96 (0.96) 0.85 (0.92) 0.14

Pain and other sensations 1.53 (1.21) 1.45 (1.12) 0.75

Urinary problems 1.20 (1.27) 1.19 (1.20) 0.68

Constipation problems 1.15 (1.21) 1.20 (1.14) 0.95

Light headedness on standing 0.92 (1.10) 1.01 (1.06) 0.11

Fatigue 1.86 (1.09) 1.74 (0.97) 0.57

Quantitative scales are from 0-4: 0 (normal) - 4 (most severe). Mean values (standard deviation)

are given.

Page 85: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

65

Table 11. UPDRS Part II Activities of Daily Living

LRRK2 p.G2019S iPD p-value

Speech 1.01 (0.98) 1.00 (0.96) 0.34

Saliva and drooling 0.97 (1.13) 1.19 (1.24) 0.02

Chewing 0.72 (0.90) 0.70 (0.87) 0.95

Eating tasks 1.18 (0.89) 1.04 (0.82) 0.24

Dressing 1.60 (1.08) 1.48 (1.05) 0.79

Hygiene 1.71 (1.13) 1.59 (1.14) 0.34

Handwriting 1.54 (1.19) 1.53 (1.17) 0.76

Doing hobbies and other activities 1.86 (1.30) 1.71 (1.11) 0.99

Turning in bed 1.56 (1.17) 1.44 (1.10) 0.80

Tremor 2.00 (1.00) 1.98 (1.00) 0.74

Getting out of bed, a car, or a deep chair 1.67 (1.15) 1.32 (1.05) 0.01

Walking and balance 1.66 (1.04) 1.37 (0.98) 0.10

Freezing 0.85 (1.13) 0.72 (1.08) 0.92

Quantitative scales are from 0-4: 0 (normal) - 4 (most severe). Mean values (standard deviation)

are given.

Page 86: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

66

Table 12. UPDRS Part III

Quantitative scales are from 0-4: 0 (normal) - 4 (most severe). Mean values (standard deviation)

are given.

LRRK2 p.G2019S iPD P-value

Mean (SD)

Part III. Total Rigidity Subscale 1.59 (0.71) 1.53 (0.76) 0.83

Part III. Total Bradykinesia Subscale 1.65 (0.96) 1.49 (0.98) 0.63

Part III. Total Tremor Subscale 0.76 (0.60) 0.65 (0.60) 0.31

Part III. Total Postural Instability and

Gait Disorder Subscale

1.33 (0.79) 1.27 (0.85) 0.40

Page 87: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

67

Table 13. UPDRS Part IV Complications of Therapy

LRRK2 p.G2019S iPD p-value

Time spent with dyskinesias 0.47 (1.00) 0.30 (0.75) 0.83

Functional impact of dyskinesias 0.46 (1.02) 0.27 (0.80) 0.52

Time spent in the off state 1.34 (1.18) 1.14 (1.03) 0.34

Functional impact of fluctuations 1.51 (1.50) 1.34 (1.36) 0.62

Complexity of motor fluctuation 0.95 (0.97) 0.88 (0.91) 0.21

Painful off-state dystonia 0.30 (0.75) 0.23 (0.60) 0.42

Quantitative scales are from 0-4: 0 (normal) - 4 (most severe). Mean values (standard deviation)

are given.

Page 88: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

68

3.3.2. Non-motor features

To assess the validity of the non-motor questionnaires we compared control subjects to iPD

for SCOPA-AUT, MOCA, Epworth sleepiness scale, and olfactory assessments (Table 14-17).

SCOPA-autonomic assessments can be clearly distinguished between control subjects and iPD

(p<0.0001). Interestingly, there is a trend for affected LRRK2 carriers with less gastrointestinal

dysfunction (mean score 0.64 in LRRK2 carriers compared to mean 0.74, p=0.04). For example,

the score for constipation is 0.64 for LRRK2 carriers compared to 0.74 in idiopathic PD.

Cognitive assessments were done using the Mini-Mental State Examination (MMSE) and

Montreal Cognitive Assessment (MOCA), six picture test and frontal assessment battery.

However, when looking into control subjects vs iPD these scores were comparable in the

Tunisian Arab Berber population. This suggests that cognitive assessments using these scales

may not be appropriate, and are not sensitive enough for measuring differences between patients

and control subjects (Table 16).

Of interest, LRRK2 carriers displayed less REM sleep behavior disorder compared to iPD

(16% for LRRK2 p.G2019S carriers compared to 29%, p<0.0001) (Table 17). Other sleep

disorders: Epworth sleepiness score, sleep apnea and restless legs syndrome were

indistinguishable between LRRK2 carriers and iPD.

Page 89: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

69

Table 14. Autonomic dysfunction (SCOPA-Aut) individual scores

Control

subjects

iPD p-value LRRK2

p.G2019S

iPD p-value

Swallowing/choking 0.20 (0.45) 0.45 (0.65) <0.0001 0.41 (0.63) 0.45 (0.65) 0.09

Sialorrea 0.13 (0.36) 0.84 (0.93) <0.0001 0.72 (0.90) 0.84 (0.93) 0.10

Dysphagia 0.14 (0.39) 0.47 (0.68) <0.0001 0.47 (0.57) 0.47 (0.68) 0.09

Early abdominal

fullness

0.33 (0.74) 0.82 (1.00) <0.0001 0.78 (0.91) 0.82 (1.00) 0.09

Constipation 0.48 (0.73) 1.40 (1.2) <0.0001 1.12 (1.09) 1.40 (1.2) 0.09

Straining for

defecation

0.41 (0.78) 1.10 (1.2) <0.0001 0.90 (1.1) 1.10 (1.2) 0.09

Faecal incontinence

0.0078

(0.09)

0.08 (0.32) 0.0035 0.08 (0.88) 0.08 (0.32) 0.09

Straining for

urination

0.33 (0.73) 0.90 (1.02) <0.0001 0.75 (0.87) 0.90 (1.02) 0.43

Urinary incontinence 0.20 (0.48) 0.65 (0.85) <0.0001 0.68 (0.92) 0.65 (0.85) 0.47

Incomplete emptying 0.23 (0.62) 0.78 (1.04) <0.0001 0.70 (0.97) 0.78 (1.04) 0.44

weak stream of urine 0.25 (0.65) 0.70 (1.00) <0.0001 0.68 (0.95) 0.70 (1.00) 0.45

frequency of urine

passing

0.33 (0.70) 0.98 (1.13) <0.0001 0.86 (1.1) 0.98 (1.13) 0.45

Nocturia 0.63 (0.88) 1.25 (1.13) <0.0001 1.11 (1.14) 1.25 (1.13) 0.45

Light headed when

standing up

0.48 (0.75) 0.85 (0.90) 0.0001 0.71 (0.87) 0.85 (0.90) 0.79

Light headed

standing some time

0.30 (0.61) 0.63 (0.82) <0.0001 0.61 (0.81) 0.63 (0.82) 0.81

Syncope 0.09 (0.28) 0.14 (0.40) 0.09 0.16 (0.45) 0.14 (0.40) 0.81

Hyperhidrosis during

day

0.53 (0.88) 1.06 (1.08) <0.0001 0.98 (1.1) 1.06 (1.08) 0.10

Hyperhidrosis during

night

0.46 (0.81) 0.90 (1.03) <0.0001 0.95 (1.1) 0.90 (1.03) 0.18

Oversensitive to

bright light

0.24 (0.67) 0.50 (0.85) <0.0001 0.55 (0.90) 0.50 (0.85) 0.56

Cold tolerance 0.49 (0.92) 0.67 (0.96) 0.0302 0.73 (0.92) 0.67 (0.96) 0.82

Heat tolerance 0.58 (0.92) 0.87 (1.00) 0.0013 0.93 (0.99) 0.87 (1.00) 0.34

Men: erection

problem

- 0.51 (1.01) 0.53 (1.02) 0.51 (1.01) 0.97

Men: ejaculation

problem

- 0.41 (0.91) 0.43 (0.94) 0.41 (0.91) 0.95

Medication for

erection disorder (%)

- 2 (0.8) 3 (1.8) 2 (0.8) 0.30

Women: vaginal

lubrication

- 1.50 (0.53) 1.4 (0.51) 1.50 (0.53) 0.38

Women: orgasm - 1.58 (0.67) 1.4 (0.51) 1.58 (0.67) 0.03

Page 90: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

70

Control

subjects

iPD p-value LRRK2

p.G2019S

iPD p-value

Constipation

medications (%)

- 37 (16%) 23 (14%) 37 (16%) 0.89

Urinary medications

(%)

- 10 (4.3%) 4 (2.5%) 10 (4.3%) 0.39

PD medications (%) - 40 (17%) 34 (21%) 40 (17%) 0.17

L-Dopa ON State

(%)

- 119 (51%) 76 (47%) 119 (51%) 0.46

Scale: 0-4. 0=never, 1=sometimes, 2=regularly, 3=often, 4=use catheter. Mean values (standard

deviation or %) are given.

Page 91: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

71

Table 15. Summary of autonomic assessments compared between LRRK2 parkinsonism

and iPD

LRRK2 p.G2019S iPD p-value

Gastrointestinal (SD) 0.64 (0.51) 0.74 (0.52) 0.04

Urinary (SD) 0.80 (0.80) 0.87 (0.83) 0.10

Cardiovascular (SD) 0.50 (0.59) 0.54 (0.54) 0.35

Thermoregulatory (SD) 0.89 (0.76) 0.87 (0.73) 0.92

SCOPA-Aut subdomain scores were compared between patients. Quantitative scales are from

from 0-4: 0 (normal) - 4 (most severe). SD: Standard deviation

Page 92: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

72

Table 16. Summary of cognitive assessment compared between iPD, LRRK2 parkinsonism

and control subjects

Control

subjects

iPD p-value LRRK2

p.G2019S

iPD p-value

MMSE (SD) 27.1 (3.29) 25.4 (3.9) 0.6898 25.7 (3.6) 25.4 (3.9) 0.42

FAB (SD) 12.7 (4.60) 10.6 (4.5) 0.2728 10.8 (4.6) 10.6 (4.5) 0.27

MOCA (SD) 17.10 (10.8) 19.3 (8.5) 0.3550 21.7 (6.8) 19.3 (8.5) 0.03

Mini Mental State Examination (MMSE), Frontal Assessment Battery (FAB), Montreal

Cognitive Assessment (MoCA).

Page 93: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

73

Table 17. Comparison of sleep scales among LRRK2 parkinsonism and iPD.

Only a subset of patients had levodopa state recorded .

LRRK2 p.G2019S iPD p-value

Epworth total score

On state (SD) 5.35 (4.67) 5.25 (4.90) 0.98

Off state (SD) 4.75 (4.48) 4.88 (5.19)

Restless legs

On state (%) 9/75 (12%) 15/118 (13%) 0.75

Off state (%) 6/81 (7.4%) 11/113 (9.7%)

REM sleep disorder

On state (%) 12/75 (16%) 34/118 (29%) 0.001

Off state (%) 14/81 (17%) 40/113 (35%)

Sleep apnea

On state (%) 7/75 (9.3%) 10/118 (8.5%) 0.65

Off state (%) 9/81 (11%) 15/113 (13%)

Page 94: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

74

3.3.3. Disease progression

The rate of disease progression was determined by taking motor and autonomic scores

over the disease duration. Age at onset was highly correlated with motor and non-motor

progression scores in idiopathic PD (R=0.20-0.31, p<0.0001). However, LRRK2 parkinsonism

was more uniform in progression (Table 18).

Table 18. Rate of disease progression associated with age at onset in patients

iPD LRRK2 p.G2019S

Correlation to age at onset Correlation to age at onset

R p-value R p-value

Hoehn and Yahr progression 0.29 <0.0001* 0.00 0.99

GI progression 0.30 <0.0001* 0.09 0.27

Urinary progression 0.22 0.0020* 0.05 0.53

Cardiovascular progression 0.20 0.0054* 0.06 0.51

Thermoregulatory progression 0.24 0.0006* 0.08 0.33

Rigidity progression 0.30 <0.0001* 0.02 0.85

Bradykinesia progression 0.29 <0.0001* 0.07 0.45

Tremor progression 0.22 0.0017* 0.07 0.19

Postural instability and gait

disorder progression

0.31 <0.0001* 0.05 0.99

R =pearson’s correlation coefficient to age at onset. Disease progression is measured by severity

score/disease duration. *=significant after Bonferroni

Page 95: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

75

3.4. Discussion

The main motor feature of LRRK2 p.G2019S parkinsonism in 220 sporadic patients and

126 familial patients was tremor-predominant parkinsonism with bradykinesia and rigidity that

responds to dopamine replacement therapy. Patients with LRRK2 p.G2019S parkinsonism are

generally indistinguishable from patients with iPD cross-sectionally, however, our data suggests

temporal distinction and trending differences in non-motor features. Earlier studies highlighted

tremor as the predominant feature of LRRK2 carriers which is supported by more recent meta-

analysis (‘dardarin’ the Basque word for tremor remains a colloquial term for LRRK2 protein)

(Aasly, et al., 2005; Healy, et al., 2008; Paisan-Ruiz, Lang, et al., 2005; Paisan-Ruiz, Saenz, et

al., 2005) . In this study, tremor was observed less in 220 Arab-Berber patients with LRRK2

p.G2019S than in iPD.

Non-motor features occurred at similar frequencies in LRRK2 p.G2019S patients and in

iPD. However, affected LRRK2 carriers have less REM sleep disorder and gastrointestinal

dysfunction. Less REM sleep disorder and olfactory impairment has also been seen in

Ashkenazi Jewish LRRK2 carriers (Saunders-Pullman et al., 2015; Saunders-Pullman et al.,

2014) . Patients with iPD have Lewy body disease in the periphery, most notably the dorsal

motor nucleus, vagal nerve, cardiac sympathetic and enteric nervous systems, as well as in the

olfactory bulb, brainstem (midbrain, pons, medulla) and cortex (Braak, et al., 2003). Nonmotor

features of cognitive impairment and hypotension has been correlated with presence of Lewy

bodies (Kalia et al., 2015) . While most LRRK2 p.G2019S patients have similar Lewy body

disease some develop alternative 4R-tauopathy or TDP43 proteinopathy (Marras, et al., 2016) .

Hence, we speculate marginal differences in REM sleep and gastrointestinal function in LRRK2

p.G2019S carriers may reflect less concomitant alpha-synucleinopathy. Less REM sleep

Page 96: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

76

behavioural disorder and peripheral dysfunction can also imply that the background of LRRK2

p.G2019S does not follow Braak staging (Goedert, et al., 2013a) .

The disease penetrance of LRRK2 p.G2019S ranges from 24% to 100% (Marder, et al.,

2015; Trinh, Amouri, et al., 2014) (Latourelle et al., 2008)(Latourelle et al., 2008)(Latourelle et

al., 2008)(Latourelle, Sun, et al.)(Latourelle et al., 2008)(Latourelle et al., 2008)(Latourelle et al.,

2008)(Latourelle et al., 2008)(Latourelle et al., 2008) (Latourelle et al., 2008) , the accuracy of

figures is disputed and some of the disparity may reflect differences in population ethnicity, bias

in patient recruitment, and differences in statistical analysis. Accurate penetrance estimates of

LRRK2 p.G2019S in Arab-Berbers are important given the highest frequency of LRRK2

p.G2019S carriers and the prevalence of parkinsonism in this region of the world (Hulihan, et al.,

2008; Lesage, Anheim, Letournel, Bousset, Honore, Rozas, Pieri, Madiona, Durr, Melki, Verny,

& Brice, 2013) . The population of Tunisia may also offer greater ethnic, genetic and

environmental homogeneity than prior North American, European and Israeli studies. The range

of age at onset in LRRK2 p.G2019S carriers is broad spanning 50 years. Ascertainment bias

appears an unlikely explanation as the kin-cohort analysis of LRRK2 p.G2019S pedigrees

supported the Kaplan Meier findings, which means the familial penetrance estimates are

comparable to the unrelated LRRK2 p.G2019S carriers. If there were an ascertainment bias, we

would expect higher penetrance estimates in LRRK2 families. Hence, penetrance modifiers that

modulate motor symptom onset in LRRK2 p.G2019S carriers appear likely but remain to be

defined.

Hoehn & Yahr scores are a composite measure of dysfunction encompassing activities of

daily living, motor and cognitive disability. In cross-sectional analysis LRRK2 parkinsonism and

iPD cannot be distinguished; the range and distributions of component symptoms and scores

Page 97: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

77

overlap. In iPD, mild progressors have an earlier onset age and faster progressors have a later

onset age. Patients with LRRK2 parkinsonism appear to have the same rate of progression

regardless of onset age. Age of onset has always been a reasonable predictor of disease

progression and morbidity in PD (Diamond, et al., 1989). Interestingly, onset age does not

predict progression for LRRK2 parkinsonism. Nevertheless, a more uniform rate of progression

in LRRK2 p.G2019S carriers may aid biomarker discovery and clinical trials focused on disease-

modification (neuroprotection). However, this might reflect a sample size and lack of test

sensitivity effect in the LRRK2 patient group.

Our objective was to compare the clinical features of iPD and LRRK2 parkinsonism and

estimate the risk in carriers as an aid for genetic counselling. Kaplan-Meier and kin-cohort

methods were used to estimate the risk of parkinsonism in sporadic and familial LRRK2 carriers.

Clinic-based and volunteer patient proband series may lead to an overestimate of the penetrance

of LRRK2 p.G2019S. However, the kin-cohort method, which does not take the proband into

consideration, gave similar results to Kaplan-Meier analyses. A weakness of our study is that

samples were only drawn from Tunisia; while LRRK2 p.G2019S carriers generally inherit the

same ancestral haplotype (Kachergus, et al., 2005) our penetrance findings may not be

universally applicable and comparative clinical and genetic studies in different ethnic

backgrounds are needed.

Presently, carrier status of this pathogenic mutation does not influence a patient’s choice

of treatment, although the discovery of LRRK2 biomarkers and specific molecular interventions

are actively sought (Cookson, 2010; R. J. Nichols et al., 2009). The range and severity of motor

and non-motor features in idiopathic PD and LRRK2 parkinsonism are comparable which

suggests therapies for LRRK2 parkinsonism might be readily generalizable to iPD. Future

Page 98: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

78

studies might include comparative whole genome sequencing (WGS) of LRRK2 patients with

divergent ages of onset in an attempt to find novel genetic variants that modulate

phenoconversion, to symptoms that warrant diagnosis and therapeutic interventions. WGS in

large multi-incident pedigrees with the p.G2019S mutation would be a good start in identifying

novel genetic modifiers as a rare variant segregating with age at onset in a family tree can be a

good indication of a modifier. Another possibility can be common polymorphisms influencing

age at onset, which can be identified in larger case cohorts. Similarly, environmental exposures

that influence risk of parkinsonism might be more readily identified in a relatively more

homogeneous patient sample.

Page 99: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

79

4. Chapter 4: Dynamin 3 modifies age at onset in LRRK2 parkinsonism

4.1. Introduction

Genetic variability in leucine-rich kinase 2 (LRRK2) has been linked to familial

parkinsonism and associated with idiopathic Parkinson disease (PD): LRRK2 c.6055G>A

(p.G2019S) confers the highest genotypic and population attributable risk (Kachergus, et al.,

2005; Ross, et al., 2011; Zimprich, et al., 2011) . Penetrance estimates are variable with a wide

range in age of onset (AOO) influenced by ethnicity (Healy, et al., 2008; Hentati et al., 2014;

Trojano, Moretta, Estraneo, & Santoro, 2010). The relatively homogeneous North-African Arab-

Berber population has the highest frequency of LRRK2 p.G2019S carriers, between 30-40% of

patients with PD (Lesage, et al., 2005; Trinh, Amouri, et al., 2014) and provides a unique

opportunity to identify genetic modifiers of AOO.

LRRK2 is a large multi-domain protein with GTPase (Roc) and kinase activities that appear

to modulate cytoskeletal outgrowth and vesicular dynamics, including synaptic transmission,

endosomal trafficking and lysosomal autophagy (Orenstein et al., 2013) . Although many

binding partners and substrates have been identified, it remains uncertain which are clinically

relevant to disease pathophysiology. Herein, a genome-wide approach was used to identify

genetic variability that directly influences LRRK2 p.G2019S penetrance.

4.2. Methods

4.2.1. Discovery cohort and replication series

Arab-Berber subjects were recruited between 2006 to 2012 by movement disorders

neurologists (FH, SBS, FN, EF) at the Mongi Ben Hamida National Institute of Neurology,

Tunis. Community-based samples consisted of 41 multi-incident LRRK2 p.G2019S families

Page 100: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

80

(150 affected and 103 unaffected LRRK2 carriers), and 232 unrelated LRRK2 p.G2019S carriers

(Table 19). All subjects were older than ≥18 years at neurological assessment and provided

informed consent prior to their participation. Specific approvals obtained from the local ethics

committee at the National Institute and Ministry of Health in Tunis were reviewed by

GlaxoSmithKline (GSK), the Institutional Review Board of Mayo Foundation and the Research

Ethics Board of the University of British Columbia. Additional replication cohorts included 263

LRRK2 p.G2019S carriers from Algeria (MT), France (AB), Norway (JAA) and North America

(PSG–Progeni GenePD Investigators (Latourelle et al., 2011) (Table 20). Human biological

samples were sourced ethically and their research use was in accord with the terms of the

informed consents. An overview of the discovery and replication samples is depicted in Figure

31.

4.2.2. Linkage analysis and STR genotyping

Genome wide linkage analysis was performed on 41 LRRK2 p.G2019S families from

Tunisia using deCODE’s 4cM density STR (short tandem repeat) marker set, with standard

approaches (Abecasis, Cherny, Cookson, & Cardon, 2002) . Allele frequencies derived from

Tunisian unrelated, non-carrier, control subjects were used for STRs. Consanguineous loops are

noted in ~1/3rd of the families but were split to maximize information content (Abecasis, et al.,

2002). Both non-parametric (NPL) and model-based linkage analyses were performed

considering early-onset and late-onset groups, dichotomized by median AOO: < or ≥ 56 years.

Linkage was performed with merlin. We had two categories: an early PD onset group (all

affected carriers with an age at onset <56 years) and a late PD onset group (all affected carriers

with age at onset ≥56 years and all unaffected carriers with an age at examination of ≥56 years).

Alternatively, AOO in patients and age at recruitment/examination of unaffected carriers were

Page 101: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

81

assessed as a quantitative trait. Model-based linkage used an additive model with incomplete

penetrance to provide LOD (logarithm of odds) and hLOD (heterogeneity LOD) scores.

4.2.3. Genome-wide SNP genotyping and association

Single nucleotide polymorphisms (SNP) were genotyped for the Tunisian Arab-Berber

cohort using Affymetrix 500K NspI and StyI (n=101) and Illumina Multi-Ethnic Genome Arrays

(MEGA) (n=131). Affymetrix genotypes were extracted from .cel intensity files using three

algorithms, BBRML, JAPL and CHIAMO, and only nominated when there was consensus as

previous (Trinh, Gustavsson, et al., 2014) ; GenomeStudio® was used to provide genotypes for

Ilumina data. Samples with genotype call rate below 99% were excluded from further analysis.

Genotype distributions for all SNPs within control subjects, and all cases combined, satisfied

Hardy-Weinberg equilibrium (HWE) expectations (p>0.001) . PLINK was used to assess IBS,

IBD and population stratification as quality measures for the MEGA and Affymetrix data

(Purcell et al., 2007). Extraction of the DNM3 locus region was performed with PLINK on

MEGA and Affymetrix merged datasets. Quality control of MEGA and Affymetrix data was

performed. A subset of consistent genotypes/individuals was assessed for population

stratification using Eigenstrat, as previously described. Prior to case-control association,

genome-wide IBS/IBD (identity by state/identify by descent) estimates were used to identify and

exclude sample contamination, duplicates and individuals with unknown relationship (e.g.

sibling-pairs in the unrelated case-control series). We assessed IBS and IBD in detail since the

unrelated carriers share the same G2019S haplotype. Within regions of linkage, PLINK

association analyses were performed (Howey & Cordell, 2014; Purcell, et al., 2007) . Quantile-

quantile plots of p-values were employed to highlight potential confounders (R package qqman).

Page 102: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

82

4.2.4. Whole genome sequencing and imputation

Whole genome sequencing (WGS) was accomplished for 14 Tunisian Arab-Berber

patients. All are LRRK2 p.G2019S carriers with a family history of parkinsonism, half had early-

onset disease (mean onset 34.9 years, SD±7.2, range 22-42) and the remainder are clinically

asymptomatic elderly carriers (mean age 77 years, SD±6.9, range 68-90). Sequencing was

carried out using Illumina 2x100 nucleotide paired-end reads, with minimum 50-fold mean depth

using standard methods for sequence alignment and variant calling (Figure 24).

SNP genotypes in the chromosome 1q23.3-24.3 region of linkage were imputed with

Beagle 4.0, (Browning & Browning, 2008, 2009) employing 14 Tunisian WGS and phased 1000

Genomes data as a reference for MEGA and Affymetrix data (n=232 LRRK2 carriers).

Subsequently, haplotype associations were assessed within the linked interval using a variable-

length Markov-chain Monte Carlo method (Browning & Browning, 2008, 2009). Affymetrix and

MEGA genotype calls were previously merged together. PLINK files were then converted into

VCF files with PLINK/SEQ and Beagle 4.0 was used for imputation. The genotype file (gt) was

designated as the merged file and 1000 Genomes data was used as the reference VCF file (ref).

There were 15075 reference markers, 1595 target markers and 2504 reference samples. Burn-in,

phase and imputation iterations were set at 10, to maximize genotype imputation accuracy. The

haplotype association was performed using Beagle 3.3 on affected unrelated individuals. Phasing

iterations and then haplotype association was performed on allelic, recessive, over-dominant and

dominant models. Corrected p-values for haplotype association and multiple-testing were

estimated by permutation analyses, randomizing case-control status. The beagle haplotype

association p-value was significant after permutation analyses, (p=0.002).

Page 103: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

83

4.2.5. Sequencing and genotyping

All subjects were screened for LRRK2 p.G2019S by Sanger sequencing or TaqMan SNP

assays-on-demand (Life Technologies, Inc, Foster City, CA), and excluded for other pathogenic

mutations implicated in PD (Gustavsson, Trinh, et al., 2015; Ishihara-Paul, et al., 2008).

Subsequent genotyping was carried out by a combination of Sequenom MassArray iPLEX

system (Sequenom, San Diego, CA) and TaqMan genotyping. Cumulative incidence plots

(Kaplan Meier) and hazard ratios (Cox proportional hazard regression models) were used to

stratify age of initial symptom by genotypes using JMP® software (SAS Institute Inc., Cary,

NC). These models were adjusted for family relatedness, gender and population series (Tunisia,

Algeria, France, Norway, and North America). Right censoring for asymptomatic carriers was

performed at age of examination. Meta-analyses of all populations was performed with R-

package ‘metafor’.

4.2.6. Brains, RNA, ampliseq transcriptome, antibodies

Brain tissue from 61 healthy control subjects without any neurological symptoms was

obtained from the Oxford Brain Bank, University of Oxford (LP); any with neurodegenerative

vascular pathology were excluded (LP). Full ethical approval (REC 07/Q2707/98) and written

informed consent are obtained for all participants. Gender, age-at-death and post mortem delay

was available for all subjects (Table 21). DNA was prepared from ~20mg of frozen tissue

samples of striatum with an Autogen NA1000 and quantified using standard methods. Prism 6.0

(GraphPad Software, Inc) was used for RNA/protein analysis. Total RNA was also extracted

(RNeasy Qiagen Minikit) from duplicate samples, and DNase I digested prior to assessing the

Page 104: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

84

concentration, quality and integrity (RIN) with an Agilent 2100 bioanalyzer, RNA 6000

LabChip kit and associated software (Agilent). After extraction, RNA integrity numbers for

samples was good quality (mean RIN = 8.9). Out of the 61 human control striatum, 38 have a

RIN > 7.0 and were used for TaqMan expression. A subset of the striatum was used for

AmpliseqTM whole human transcriptome analysis (n=17) was performed with an Ion Proton

(Life Technologies, Inc). High-quality, total RNA was reverse transcribed and amplified using

TaqMan One Step RT-PCR kit following manufacture’s protocol (ABI). Sequencing analysis

resulted in an average of over 12 million reads per sample and a read length of 114 bases.

AmpliseqRNA was used to map reads and generate absolute/normalized gene expression values

(reads per million, RPM). RNA expression analyses were adjusted by RIN quality. Expression

levels were quantified by dividing 2-Ct by the geometric mean of the expression levels of three

commonly used “housekeeping” genes: hypoxanthine phosphoribosyl-transferase (HPRT;

Hs02800695_m1), glyceraldehyde-3-phosphate dehydrogenase (GAPDH; Hs02758991_g1) and

synaptophysin (SYP; Hs00300531_m1)). DNM3 expression was measured using Taqman probe

expression assay ID Hs00399015_m1 (all transcripts) and Hs00927940_m1 (NM_001136127.2

and NM_015569.4 only). Likewise, Ampliseq Transcriptome data was normalized with a variety

of housekeeping genes (GAPDH, HPRT, SYP, YWHAZ), including those primarily expressed in

neurons (TH, MAP2, ENO2, SV2A, SV2B, SYN1, SYN2) and the expression findings were

robust. For protein analysis, 20mg brain tissue (n=17) was lysed with buffer containing 1% NP-

40, 20mM HEPES, 125mM NaCl, 50mM NaF and protease inhibitor cocktail (Roche). The

lysates were put on ice for 1 hour. Blotting of dynamin-3 was done with a polyclonal rabbit

antibody (Synaptic Systems [115 302], 1:1000) and anti-for GAPDH a mouse monoclonal

antibody was used (Thermo Scientific [MA5-15738], 1:1000). MAP2 antibodies were used in

Page 105: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

85

immunofluorescence (Abcam, 1:1000).LRRK2 p.G2019S mice, primary neuronal littermate

cultures, immunostaining and image analysis were as previously described (Beccano-Kelly et al.,

2014) .

4.3. Results

4.3.1. Linkage and association of LRRK2 p.G2019S families

Linkage analysis of AOO in 41 Tunisian LRRK2 p.G2019S pedigrees identified

chromosome 12q12 using non-parametric (LOD NPL = 3.3, θ=0 at D12S85) and model-based

methods (maximum LOD = 7.6, θ =0 at D12S85 under a dominant model of inheritance), which

encompasses the LRRK2 locus. Genome-wide analysis using similar approaches, with allele-

dependent penetrances, also identified chromosome 1q23.3-24.3 (LOD NPL =2.90, maximum

LOD & hLOD = 4.99, θ =0 at D1S2768 with a recessive model, and LOD = 2.81 and hLOD=

3.81, θ =0 at D1S2768 with a dominant-additive model) (Figure 22). Significant linkage was

obtained using AOO as a dichotomous trait and was robust to subsequent ordered subset analyses

over a range of divisions (Hauser et al., 2004) and implications of significant familial

heterogeneity. The highest LOD score across all models was obtained on chromosome 1.

However, there was suggestive linkage on chromosome 6, 17 and 21 (Figure 26).

Evidence for association within the chromosome 1q23.3-24.3 linkage region (170.8-

172.5Mb, the maximum LOD -1 support interval) was assessed in unrelated LRRK2 p.G2019S

carriers (n=232). Only affected individuals were included in this association, the unaffected

carriers were excluded. Association with dichotomized AOO revealed three associated SNPs

(rs742510, rs2421947 and rs2206543, r2= 0·98; pnominal=2·6 x 10-5,) within the dynamin 3

locus (DNM3)(Table 23). Within the chromosome 1q23.3-24.3 linkage region, 634 SNPs were

Page 106: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

86

assessed. A Bonferroni correction was applied to account for multiple testing (corrected

p=0.016). A QQ-plot for association analyses on chromosome 1 deviated from the line of

equality but the DNM3 rs2421947 association was confirmed by TaqMan probe genotyping

(Figure 27).

Subsequently, all 21 coding exons of DNM3 gene were sequenced in LRRK2 p.G2019S

carriers with divergent AOO (n=25) and three rare (MAF<0.01) synonymous variants were

identified (p.A81A, p.H128H and p.V609V). Carriers of divergent AOO refer to LRRK2

p.G2019S carriers who have early onset PD (<45 onset year) or were elderly (>75years) without

motor signs of PD.

4.3.2. Higher resolution mapping

WGS of 14 LRRK2 p.G2019S Tunisian Arab-Berber subjects and 1000 Genomes data

provided references for SNP imputation, to improve haplotype analysis and identify specific

variability associated with AOO. Within and flanking the DNM3 locus (chr1:171,810,018-

172,382,057) a dense framework of informative markers (MAF>0.05) was imputed in all

unrelated LRRK2 carriers (n=232) with Affymetrix, MEGA and Sequenom iPLEX genotypes.

The shortest, most significant haplotype associated with AOO was subsequently defined between

chr1:171,832,491-171,833,094 (rs77565020 to rs2421947, 603bp), using variable-length

Markov-chain Monte Carlo methods (p=1.07 x 10-7, Text S1). Within the disease-associated

haplotype allelic association with rs2421947 was most significant (p=1.07 x 10-7) (Figure 22,

Table 24).

The Kaplan-Meier method was used to calculate median/IQR censoring at age of last

examination for unaffected carriers by DNM3 genotype. rs2421947 CC homozygous carriers had

Page 107: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

87

a median AOO of 64 years (IQR: 48-67); CG heterozygotes had a median 57 (IQR: 50.5-64

years), and GG homozygotes had a median 51.5 (IQR: 46-61.5 years) (Kaplan Meier log-rank p-

value=0.03) (Figure 23). The median onset of LRRK2 parkinsonism in DNM3 rs2421947 GG

homozygotes is 12.5 years younger than CC homozygotes. DNM3 rs2421947 has a minor allele

frequency (MAF) C= 0·42 in unrelated control subjects from Caucasian populations (HapMap-

CEU, n=226), and 0·42 in unrelated control subjects from Tunisia (n=321). In LRRK2 p.G2019S

carriers the MAF C=0·39 overall, irrespective of affection status, but increases to C=0·46 with

disease onset ≥56 years.

To fully estimate the effect of DNM3 rs2421947 in the Tunisian population, we

combined the unrelated individuals and families in a Cox proportional hazard model censoring

unaffected individuals while adjusting for family relatedness and gender (HR 1.63, CI=1.05-

2.63, p=0.03 for alternate homozygous genotypes).

4.3.3. DNM3 expression in brain

DNM3 rs2421947 was genotyped in striatal brain tissue (n=61) to assess any influence on

expression. The rs2421947 GG genotype was correlated with higher DNM3 mRNA levels

(r=0.25, p=0.006) (Figure 28), 1.25-fold higher for the GG genotype compared to CC. Results

were confirmed using Ampliseq whole transcriptome analysis in a subset of samples (n=17;

transcriptome data available on request). The findings were robust to normalization with a

variety of housekeeping genes. DNM3 total transcript expression was correlated with LRRK2

expression (r2=0.65 p=0.004)(Table 25). Dynamin-3 protein levels in striatum stratified by

rs2421947 genotype (n=17) are higher for the GG genotype (1.6 fold higher, p=0.08, Figure 29).

Page 108: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

88

4.3.4. Replication cohorts

The DNM3 association with AOO was examined in additional LRRK2 p.G2019S carriers

including subjects originating from Algeria (n=46), France (n=65), Norway (n=64) and North

America (n=88). DNM3 rs2421947 was imputed for the American series using 1000 Genomes as

a reference , or was otherwise genotyped. Of note, the MAF for LRRK2 carriers in each

population series is different. Cox proportional hazard ratios are provided for each population,

censoring unaffected individuals, adjusting for family relatedness and gender as covariates, and

combined within a meta-analysis also adjusting for population in the model (HR 1.46 CI=1.04-

2.04, p=0.02 for alternate homozygous genotypes) (Figure 23).

4.4. Discussion

Unbiased genome-wide linkage analyses and locus–specific association, with replication

of that association in an unrelated series, nominate DNM3 as a genetic modifier of AOO in

LRRK2 p.G2019S parkinsonism. The frequency of LRRK2 p.G2019S carriers is higher in North

Africa than in any other region reported to date (Kachergus, et al., 2005; Ross, et al., 2011).

Hence a strength of our study is the large number of patients and family members with LRRK2

p.G2019S originating from the same population. Clinical exams applied longitudinally by the

same team of movement disorder specialists ensure accurate diagnoses and consistent data

reporting. Inclusion of unrelated, incident cases at one site also avoids potential selection biases

in referrals from multiple centers. The Arab-Berber population of Tunisia provides ethnic,

genetic and environmental homogeneity to increase power for discovery. However, there are also

many study limitations. In general, AOO is broadly defined and subjective: its variance is large

even within LRRK2 families although highly correlated with age of a motor diagnosis.

Page 109: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

89

Nevertheless, the variance in AOO in families is less than the variance in unrelated LRRK2

p.G2019S carriers suggestive of penetrance modifiers . AOO is a fixed albeit temporal measure

of disease pathophysiology. Hence, in our initial linkage and association analyses a dichotomized

approach was used, using AOO about 56 years as a categorical variable. Key findings were

assessed using Cox proportional hazards regression models censoring unaffected individuals,

adjusting for family relatedness, gender and population series. It would be worthwhile to

examine disease onset and progression in other ways. Longitudinal follow up of these families,

additional patients and asymptomatic carriers is warranted.

In general, AOO is broadly defined and subjective: its variance is large even within

LRRK2 families in this study although highly correlated with age of a motor diagnosis.

Nevertheless, the variance in AOO in families is less than the variance in unrelated LRRK2

p.G2019S carriers suggestive of penetrance modifiers (Table 19, median interquartile range).

AOO is a fixed albeit temporal measure of disease pathophysiology. Hence, it would be

worthwhile to assess onset and disease progression in other ways. Longitudinal follow up of

these families, additional patients and asymptomatic carriers is warranted.

Genome-wide linkage analysis to AOO was performed in large LRRK2 p.G2019S

pedigrees employing informative STRs. The highest linkage peak identified is on chromosome

12 and explained by p.G2019S; however, there was no evidence of genetic variability in cis or

trans within this region influencing AOO. In Tunisia, several heterozygous ‘married in’ relatives

and homozygous carriers are observed in highly consanguineous multi-incident pedigrees, and

within the families 150 (59%) of carriers are affected (Table 19). While the incidence of

idiopathic PD is generally low (~2% at >65 years), and biologically carrier status and AOO may

be independent, in our dataset this does not appear to be the case. Hence, we took a careful look

Page 110: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

90

at both the cis/trans effects of the LRRK2 haplotype and association between AOO and common

polymorphisms. Nevertheless, no significant effects were identified after a Bonferroni correction

(data available on request). However, lack of evidence for association to AOO should not be

considered evidence against. The second highest linkage peak is on chromosome 1q23.3-24.3

and remained robust when considering different models and allele frequencies. Other linkage

peaks were also present on chromosome 6, 17 and 21; while none showed evidence for

association further investigation is warranted in larger datasets. LRRK2 p.G2019S is a relatively

rare, pathogenic mutation for disease. Thus our study was limited by the number of LRRK2

p.G2019S carriers available, in families and in population-based series of idiopathic PD. As a

continuous trait the distribution of affected carriers was too sparse for AOO analysis; unaffected

carriers were not included and there was insufficient information for linkage analysis. However,

as a dichotomized trait, we were able to include unaffected carriers’ age greater than or equal to

the median AOO. In addition, unaffected carriers younger than the median AOO for the

pedigrees were marked as ‘unknown’ status in pedigree analyses and thus contribute their

genotype information. Hence, the significance of the DNM3 finding may be driven by the

inclusion of unaffected carriers older than the median AOO, not only affected carriers. Overall,

rs2421947 appears to have an effect on AAO of LRRK2 p.G2019S parkinsonism. Nevertheless,

confidence intervals are wide and span 1.0 for several replication series, albeit relative to sample

size, and the effect appears to be in the opposite direction for the French series. In addition, in

replication series, a major caveat is that convenience samples suffer an intrinsic ascertainment

bias – as they are from patients with PD of which a subset were found to be p.G2019S carriers.

Worldwide LRRK2 p.G2019S is generally inherited from the same ancestral haplotype

(Kachergus, et al., 2005) but the influence of modifiers and their associated allele frequencies

Page 111: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

91

may be population specific. There is suggestive linkage (LOD = 2.43) for AOO on chromosome

1q32.1 in predominantly North American LRRK2 p.G2019S families, albeit with no evidence

for association in that region in those samples (Latourelle, et al., 2011) . Nevertheless, genome-

wide association analysis of idiopathic PD in Japan robustly implicates PARK16 within 1q32

(Satake et al., 2009), which is reproducibly observed albeit with low effect size (OR ~1.1) in a

mega meta-analysis of Caucasian samples (Nalls, Pankratz, Lill, Do, Hernandez, Saad,

DeStefano, Kara, Bras, Sharma, Schulte, Keller, Arepalli, Letson, Edsall, Stefansson, Liu, Pliner,

Lee, Cheng, Ikram, et al., 2014) . PARK16 includes RAB29 (formerly RAB7L1) investigated as

a candidate gene and associated with reduced risk of idiopathic and monogenic parkinsonism

(LRRK2 p.G2019S and GBA p.N370S) in Ashkenazi (Gan-Or et al., 2008) . Functional studies

also support an interaction between RAB7L1 and LRRK2 (Beilina et al., 2014; D. A. MacLeod,

et al., 2013) . Nevertheless, chromosome 1 linkage results in this and the previous study appear

independent. Patterns of linkage disequilibrium in Tunisian Arab-Berber and Israeli Jewish

population samples are also different thus additional tagging SNPs may be required to evaluate

DNM3 or other loci as penetrance modifiers.

Non-synonymous variability in DNM3 was not observed in LRRK2 carriers which

allowed us to focus on polymorphic non-coding eQTLs (expression quantitative trait loci).

Variability in DNM3 expression correlates with genotype whether quantified by Ampliseq

transcriptome or TaqMan methods. A specific isoform (Dyn3b) co-localizes with clathrin (Cao,

Garcia, & McNiven, 1998) and appears more highly expressed in DNM3 rs2421947 GG

homozygotes (Figure 28, Figure 29). In striatum DNM3 and LRRK2 expression are correlated

suggesting they are involved in the same process. DNM3 rs2421947 does not appear to

Page 112: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

92

contribute to risk of idiopathic PD, neither susceptibility nor AOO, but the influence of DNM3

rare variability has yet to be explored.

LRRK2 has been implicated in neurite outgrowth (D. MacLeod et al., 2006; Parisiadou et

al., 2009) , synaptic vesicle trafficking and neurotransmitter release , and via kinase-dependent

mechanisms (Arranz et al., 2015) . Much of the underlying mechanistic biology in these

processes remains enigmatic, as does their clinical relevance to PD. However, our genetic study

shows DNM3 is an AOO modifier of LRRK2 p.G2019S parkinsonism. LRRK2 co-

immunoprecipitates with the dynamin family GTPases that drive membrane fission (DNM1-3

and dynamin-related proteins). LRRK2 co-immunoprecipitates with the dynamin family

GTPases that drive membrane fission (DNM1-3, and dynamin-related proteins) (Stafa et al.,

2014) . Amphiphysin recruits dynamin and endophilin A (a LRRK2 kinase substrate) , and

recruits synaptojanin (SYNJ1) for endocytic vesicle fission (S. M. Ferguson & De Camilli, 2012)

. Recessive mutations in SYNJ1 have been implicated in seizure disorders and early-onset

parkinsonism (Krebs et al., 2013; Quadri, et al., 2013) . In neurons, dynamin 3 localizes to the

endocytic machinery of dendritic spines to modulate receptor recycling and excitatory synaptic

transmission (Gray, Kruchten, Chen, & McNiven, 2005) . In this process ‘Dyn3b’ isoform

expression is also centrally involved in the regulation of actin polymerization, filopodia and

spine formation (Cao, et al., 1998; Gray, et al., 2005) . Intriguingly, a significant reduction and

redistribution of dendritic dynamin 3 staining is observed in LRRK2 p.G2019S murine cortical

cultures (Figure 30), although it may also reflect elevated glutamateric synaptic transmission

(Beccano-Kelly, et al., 2014) .

We postulated LRRK2 p.G2019S activates kinase activity (Kachergus, et al., 2005), an

outcome of which has been the pursuit of competitive LRRK2 inhibitors. Based on similarly

Page 113: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

93

unbiased genetic data, we postulate lower levels of DNM3, and perhaps specific dynamin 3

isoforms, will delay the onset of LRRK2 p.G2019S parkinsonism. The crystal structure of the

dynamin tetramer has just been elucidated (Reubold et al., 2015) and might accelerate the

development of dynamin GTPase inhibitors (dynasores). These anticonvulsants repress synaptic

transmission in seizure disorder (Li et al., 2015) and delay alpha-synuclein uptake by neuronal

and oligodendroglial cells (Konno et al., 2012) . At autopsy, most LRRK2 p.G2019S carriers

have alpha-synucleinopathy and Lewy body disease (Ross et al., 2006) . Thus DNM3 expression

represents a target for neuroprotection in LRRK2 p.G2019S carriers, and potentially for disease-

modification in LRRK2 parkinsonism.

Page 114: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

94

Figure 22. Chromosome 1 linkage peak

a. (LOD score = 4.99). b. Region of association within the LOD -1 linkage interval: Plink SNPs

(10-5

) and Beagle haplotype (p=1.06 x 10-7

) associations

Page 115: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

95

Figure 23. Age-associated cumulative incidence of LRRK2 p.G2019S carriers.

a. Replication cohorts: Algerian, French, Norwegian and North American, stratified by

rs2421947 genotype (log rank p=0.0001). b. All populations combined (Algerian, French,

Norwegian, North American and Tunisian Arab Berber) stratified by rs2421947 genotype (log

rank p<0.0001).

B

A

Page 116: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

96

Table 19. Demographics of discovery cohorts: Tunisian Arab-Berber LRRK2 p.G2019S

carriers

Unrelated

patients

Unrelated

control

subjects

Familial

patients

Unaffected

family

members

N 220 12 150 103

Number of men

(%)

124 (56%) 6 (50%) 77 (51.3%) 48 (46.6%)

Mean age (SD)

years

67.6 (12.6) 56.7 (10.9) 68.6 (15.8) 56.1 (17.5)

Median age

(IQR)

69 (48-90) 54.5 (38-72) 70.5 (57-81) 53 (43-72.5)

Mean age of

onset (SD)

57.1 (11.6) - 56.1 (12.8) -

Median age of

onset (IQR)

57 (40-74) - 56 (47-65) -

Page 117: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

97

Table 20 . Demographics of LRRK2 p.G2019S carriers: replication series

Norway France Algeria North America Total

Patient Unaffected Patient Unaffected Patient Unaffected Patient Unaffected

N 19 45 48 17 45 1 88 - 263

Number

of men

(%)

8 (42%) 18 (40%) 26 (60%) 7 (39%) 19 (42%) - 41(47%) -

Mean

age

(SD)

years

67.6

(17.5)

63.6 (12.4) 57.7 (13.8) 67.4 (11.8) 55.5

(11.3)

54 NA -

Median

age

(IQR)

73 (52-82) 62 (54.5-

70)

59 (46.8-

67.3)

67 (59.5-

76.8)

55 (45.3-

63)

54 NA -

Mean

age of

onset

(SD)

62.6

(13.0)

- 52.1 (13.5) - 49.6

(10.3)

- 61.5

(10.1)

-

Median

age of

onset

(IQR)

65 (49-74) - 51 (41.3-62) - 50 (43-

56)

- 63 (56-

70)

-

Page 118: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

98

Table 21. Demographics of healthy control brains for expression analysis

Control subjects

N 61

Number of men (%) 30 (49.2%)

Mean age at death (SD) years 80.6 (12.1)

Median age at death (IQR) 85 (71-89)

Tissue type Striatal

Average RIN (RNA integrity

number) (SD)

8.6 (1.2)

Average PMI (Post-mortem

interval) (SD)

48.8 (33.2)

Page 119: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

99

Table 22. Primer pairs and custom TaqMan probe design for different DNM3 transcript

isoforms in human striatum

Names 5'->3' Primers Amino acid sequence

Region 1

DNM3_Reg1F aaacggaaaggattgttgc

DNM3_Reg1Fprobe tctcttacatcaacaccaacc

DNM3_Reg1BR cccttgcgaatcacaatttg GTNLPPSRQI

DNM3_Reg1AR ttgcgaatcacctgatttc

DNM3_Reg1C_F gcaaattgtacgagctaagttc VRAKFCKLYCCFFI

DNM3_Reg1_R ttcaggttgtccaagggaag

Region 2

DNM3_Reg2A_F tatcctgacaaatctgtagctg SVAEN

DNM3_Reg2_R ggtcctctgaagaatacaac

DNM3_Reg2B_F tctgtagggaacaacaaagc SVGNNKAEN

DNM3_Reg2_R ggtcctctgaagaatacaac

Region 3

DNM3_Reg3_F aaaggaggccaacactaag SRRPPPSPTRPTIIRP

DNM3_Reg3B_R attatagtgggacgagttgg

DNM3_Reg3_F aaaggaggccaacactaag RFGAMKDEAAEP

DNM3_Reg3A_R cagcagcttcatccttcatgg

Probe

TaqMan Probe design attggcttcgcaaatgctcagcagag

Page 120: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

100

Table 23. PLINK association underneath linkage regions

CHR SNP BP (hg19) A1 F_A F_U A2 CHISQ P OR

1 rs742510 171858930 A 0.5 0.1974 G 18.13 2.06 x 10-5

** 4.067

1 rs2421947 171833094 C 0.5 0.1974 G 18.13 2.06 x 10-5

** 4.067

1 rs2206543 171835493 G 0.5 0.1974 A 18.13 2.06 x 10-5

** 4.067

** Values significant after Bonferroni correction for all SNP association tests within the LOD-1 linkage interval on chr 1

Page 121: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

101

Table 24. DNM3 haplotypes associated with AAO

rs77565020

rs75848807

rs192895361

rs74673993

rs142760983

rs559149705

rs185844670

rs541736672

rs563254497

rs530428455

rs190417579

rs74777828

rs192302781

rs146042960

rs566301333

rs376575981

rs183688167

rs114979811

rs56237038

rs72713714

rs2421947

count p-value

Major haplotypes:

G G G T A G G A A C A T A T G A G T A G G 238 1.07E-07**

G G G T A G G A A C A T A T G A G T A G C 138 0.122

Minor haplotypes:

G A G T A G G A A C A T A T G A G T A G C 1 NA

G G G T A G G A A C A A A T G A G T A G C 5 NA

G G G T A G G A A C A T A T G A G T A C C 3 NA

G A G T A G G A A C A T A T G A G T A C C 1 NA

G A G T A G G A A C A T A T G A G T A G G 2 0.515

G G G T A G G A A C A T A T G A G T A C G 1 0.411

** Values significant after Bonferroni correction all haplotype associations within the LOD-1 linkage interval on chr 1

Page 122: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

102

Table 25. DNM3 transcript levels correlate with LRRK2, VPS35 and SYNJ1 expression in

striatal tissue transcriptome data from normal controls (n=17).

Gene DNM3 expression

level correlation

coefficient

p-value

Genes implicated in

Late-onset autosomal dominant

LRRK2 0.65 0.004**

VPS35 0.65 0.008

SNCA 0.65 0.04

DNAJC13 0.21 0.14

Genes implicated in

Early-onset recessive

SYNJ1 0.41 0.008

PINK1 0.53 0.02

PARK2 0.21 0.04

FBXO7 0.51 0.12

**Values significant after Bonferroni correction

Page 123: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

103

Table 26. Sensitivity analysis for different age cut-offs on chromosome 1q23.3-24.3 using

non-parametric linkage

Age at onset dichotomization

Chromosome 1q23.3-24.3

NPL LOD score

45 years 2.3

50 years 2.5

55 years 2.9

60 years 1.8

65 years 1.2

Page 124: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

104

Clinical characteristics of subjects with WGS

7 early onset LRRK2 p.G2019S carriers 7 asymptomatic LRRK2 p.G2019S carriers

Mean Age of Onset (SD): 34.8 (7.2) Mean Age (SD): 77 (6.9) Mean Sequencing Depth

50X

Align to NCBI hg19 Build38

Compare SNPs against dbSNP

(91% of variants represented in dbSNP)

Average number of SNP variants ~3,000,000

Average number of insertion variants ~325,750

Average number of deletion variants ~349,189

Imputation of chromosome 1 linkage region (DNM3 locus)

Use imputed data for Beagle haplotype association

.Figure 24. Whole genome sequencing and imputation workflow

Page 125: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

105

Figure 25. A schematic of the thirteen dynamin isoforms.

Refer to table 22 for primer designs to capture different amino acid sequences. Figure adapted

from (Cao, et al., 1998).

Page 126: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

106

0

1

2

3

4

5

6

7

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22

A. Multipoint model-based linkage LOD (blue) and HLOD (black) dominant model

0

2

4

6

8

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22

B. Multipoint model-based linkage LOD (blue) and HLOD (black) recessive model

0

2

4

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22

C. Multipoint non-parametric linkage (NPL) LOD (cumulative)

LO

D

score

LO

D s

core

Chromosome

Chromosome

LO

D

score

Chromosome

Page 127: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

107

Figure 26. Multipoint model-based and non-parametric linkage analysis of Tunisian Arab-Berber LRRK2 p.G2019S families. A. Parametric linkage with divergent ages at onset (<56 or ≥56 years) , using a dominant model with incomplete penetrance; B

Parametric linkage hLOD cumulative scores; C. Non-parametric linkage D. Continuous trait analysis

0

1

2

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24

D. Continuous trait linkage analysis (NPL) LOD

LO

D s

core

Page 128: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

108

Figure 27 Chromosome 1 Q-Q plot values

Page 129: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

109

.

Figure 28. DNM3 transcript levels normalized by geometric mean of housekeeping genes

Total DNM3 RNA levels

DNM3 rs2421947

DN

M3/G

eo

metr

ic M

ean

CC

CG

GG

0

1

2

3

4 ** p=0.006

CC CG GG

Dyn3A/Dyn3B 1.53 1.46 0.98

No

rmal

ize

d D

NM

3

Page 130: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

110

Figure 29. Dynamin 3 protein levels normalized by GAPDH

DNM3 protein levels

DNM3 rs2421947 genotypes

DN

M3/G

AP

DH

CC

GG

0.0

0.5

1.0

1.5

2.0 p=0.08

Page 131: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

111

Figure 30. Dynamin 3 staining in cortical neurons

A. representative confocal microscopic images of dynamin-3 (red) and MAP2 (blue) staining in

wild-type (WT) and GKI (LRRK2 p.G2019S) murine cortical neurons, cultured as previously

described (Beccano-Kelly, et al., 2014)Left: 60X 2-times zoom of individual neuron staining.

Right: expanded region of interest with and without MAP2; B. Quantification of dynamin-3

intensity in cortical cultures (DIV=21). Scale bars, 50um, n=3 cultures per group; C.

Quantification of dynamin-3 cluster density in cortical cultures (DIV=21) **=p<0.05.

A

B

C

Page 132: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

112

Figure 31. Flow diagram of discovery and replication cohorts

Page 133: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

113

5. Chapter 5: Elucidating mechanisms of reduced penetrance in Mendelian disease

5.1. The importance of reduced penetrance

It has been over 10 years since the discovery of LRRK2 mutations in PD. The penetrance

of LRRK2 p.G2019S parkinsonism is complex and varies across ethnicities and environments.

Other pathogenic mutations in LRRK2 also show variable penetrance estimates. The penetrance

estimates for p.G2019S are relevant for genetic counseling, but treatment and prognosis for

these patients are the same as typical PD. The lack of a definitive cure for PD drives the search

for modifier genes that are informative for genetic counseling, disease severity and potential new

avenues for therapeutics. However, discovering penetrance modifiers in monogenic forms of

disease (albeit genetic or environmental) requires large numbers of mutation carriers (both in

families and sporadic unrelated patients) for sufficient power. Here we have a large

homogeneous Tunisian population with a high frequency of the LRRK2 p.G2019S mutation. Our

study was limited by the rarity of this Mendelian form of parkinsonism, in families and in

population-based series of idiopathic PD, and it has taken 10 years of research to build this

valuable resource of clinical and genetic data.

In PD, homozygous or compound heterozygous mutations in Parkin and PINK1 are

highly penetrant genotypes in early onset parkinsonism, heterozygous mutations in LRRK2,

VPS35, EIF4G1, have reduced penetrance estimates and heterozygous states of Parkin/PINK1

mutations may be regarded as pathogenic but with very low penetrance, although this is

debatable (Klein & Ziegler, 2011) . Thus far, there has not been a genetic modifier identified for

genes implicated in monogenic forms of PD. Although few genetic modifiers have been

identified and validated, there are many biological candidates. Mutations of GBA are an

important risk factor for PD which reduce enzyme activity leading to ER associated degradation.

Page 134: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

114

The GBA enzyme, GCase, interacts with alpha synuclein. Reduced GCase in GBA mutation is

associated with increased SNCA (Schapira, 2015) .

There are several examples of genetic modifiers in movement disorders and

neurodegeneration. Mutations in SGCE lead to development of myoclonus-dystonia. However,

maternal imprinting of SGCE does not lead to disease in the offspring when transmitted through

the mother (Guettard et al., 2008) . The finding is extremely relevant for young female patients

with an SGCE mutation, as their children will not suffer from the disease. Likewise, DYT1

dystonia is caused by a TOR1A GAG deletion. However, when p.D216H polymorphism in

TOR1A is present in trans, there is reduced penetrance of the TOR1A GAG deletion to 3%

(Bruggemann et al., 2009; Kamm et al., 2008; Klein, 2014).

Herein, we have the unique opportunity of a large homogeneous cohort with one identical

mutation to study age-at-onset genetic modifiers. The work in this thesis has made use of

collected detailed clinical research forms to study and characterize endophenotypes in LRRK2

parkinsonism. The work has demonstrated the usefulness of families in linkage analysis,

withsubsequent use of whole-genome sequencing and haplotype analysis. We have identified a

potential age-at-onset modifier for the most common mutation in familial PD.

5.2. Factors that influence penetrance

The phenotypic manifestation of mutations in neurodegenerative diseases are age-

dependent, e.g. c9orf72 in FTD/ALS, LRRK2 mutations in PD, HTT expansion in HD. The risk

of developing the disease increases with age. The mutation type can also influence penetrance.

Some mutations are more penetrant than others. For example, we have found that SNCA point

Page 135: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

115

mutations, duplications and triplications are more highly penetrant in comparison to LRRK2

point mutations in PD(Trinh, Guella, et al., 2014). This may be due to mutation type (duplication

or triplications can severely influence the patient compared to point mutations). It can also be

due to gene-specific differences. Perhaps perturbations in SNCA have a larger effect on disease

processes compared to LRRK2. But even within the same gene, the penetrance estimates are

vastly different. For example, LRRK2 p.G2019S is more penetrant compared to the

R1441G/C/H mutations. Perhaps penetrance correlates with the kinase or GTPase activity in

LRRK2. This phenomena is not specific to PD. Cis and trans elements that control gene

expression can also influence the penetrance of mutations. If there is unequal expression of the

wild-type and the mutant allele, then there would be an influence on expression levels. The

ethnic or environmental background can influence penetrance. In our study, we have shown that

cumulative incidence estimates are significantly different between Norwegians and Tunisians

with the LRRK2 p.G2019S mutation (Hentati, et al., 2014) . Furthermore, new studies with

larger sample sizes show that there are differences between Ashkenazi Jews from New York

(n=90 LRRK2 p.G2019Scarriers) and Tunisian Arab Berbers (n=220 LRRK2 p.G2019S

carriers)with the same LRRK2 p.G2019S mutation (Marder, et al., 2015; Trinh, Guella, et al.,

2014). Gender can also influence penetrance, although controversial, females may have higher

risk of disease compared to males in LRRK2 p.G2019S (Cilia et al., 2014) (Trinh, Amouri, et al.,

2014) . This is in contrast to the higher risk of idiopathic PD in males compared to females (de

Lau & Breteler, 2006) .

Page 136: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

116

5.3. Methods and approaches to identify genetic modifiers

The most obvious candidates for genetic modifiers may be the top GWAS hits. Genes

such as SNCA, MAPT, RAB7L1 that contribute to risk of PD could also modify age-at-onset in

idiopathic PD. In fact, there are a few polymorphisms in SNCA and the TMEM175/GAK loci that

may influence age-at-onset (Lill et al., 2015; Ritz, Rhodes, Bordelon, & Bronstein, 2012). A

combined genetic risk score for age-at-onset of all significantly associated SNPs revealed that

the signal was mostly driven by SNCA and the TMEM175/GAK. There was a reduction of the

effect when these two top SNPs were removed and thus other PD risk loi besides SNCA and

TMEM175/GAK have a relatively small contribution to AAO variability (Lill, et al., 2015) .

Another study has shown that SNCA rs356165 and rs356219 modifies age-at-onset in idiopathic

PD (Brockmann et al., 2013) . However, we have found that SNCA polymorphisms do not have

an effect on disease risk or onset age in LRRK2 p.G2019S carriers (Trinh, Gustavsson, et al.,

2014), suggesting that PD GWAS risk loci may have a relatively small contribution to modifying

endophenotypes in Mendelian forms of PD.

A combination of linkage analyses, genome-wide association studies, meta-analyses and

exome sequencing of ‘extreme’ cases have been used to identify modifiers of disease severity

and comorbidities in the field of complex genetic disorders (Wright et al., 2011) (Emond et al.,

2012) which requires good phenotyping, especially in the context of movement disorders and

longitudinal follow up on families and patients. It also requires large Mendelian families or sib-

pairs segregating with disease. Many modifiers of endophenotypes in cystic fibrosis have been

explored with linkage analysis of phenotypes such as lung disease severity (Corvol et al., 2015;

Emond, et al., 2012; Wright, et al., 2011). A genome-wide study on 486 sib-pairs identified

linkage on chromosome 20q13.2 that modifies lung disease severity (Wright, et al., 2011) .

Page 137: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

117

Another method is looking for ‘protective’ alleles. These are alleles that lower risk of

getting disease. One example is in the context of cholesterol low-density lipoprotein, loss of

function variants in PCSK9 were found in individuals with low levels of LDL cholesterol (Cohen

et al., 2005) . Other examples include inactivating mutations in NPC1L1 and APOC3 protecting

from coronary heart disease (Crosby et al., 2014; "Inactivating mutations in NPC1L1 and

protection from coronary heart disease," 2014; Jorgensen, Frikke-Schmidt, Nordestgaard, &

Tybjaerg-Hansen, 2014) . Protein inactivating mutations in NPC1L1 such as p.Arg406X were

more associated with lower LDL cholesterol levels and lower risk of coronary heart disease.

Protective alleles may also exist in the LRRK2 p.G2019S carriers. The Norwegian population

may carry ‘protective’ alleles that the Tunisian Arab-Berber population does not carry.

Interestingly, the DNM3 rs2421947 GG genotype is almost absent in the Norwegian LRRK2

p.G2019S carriers.

Penetrance of mutations can be modified by expression levels. Using translational models

of human stem cells or other mammalian models to look for transcriptomic differences may be

one important step to test potential candidate modifiers or look for novel modifiers. THAP1

mutations can cause early onset primary torsion dystonia, with an autosomal-dominant

inheritance and 40% penetrance (T. Fuchs et al., 2009) . THAP1 encodes a transcription factor

that regulates expression of TOR1A and also autoregulates its own expression levels (Erogullari

et al., 2014) .

Based on this multiplicity of mechanisms and their conceivable interactions, it appears

unlikely that a single approach will suffice to arrive at a comprehensive understanding of the

molecular mechanisms underlying reduced penetrance of movement disorders. In this respect, a

Page 138: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

118

number of DNA and RNA based genetic methods, complemented by functional models may be

required for thorough investigations of potential modifier genes.

5.4. Dynamin 3 as potential therapeutic target of LRRK2 parkinsonism

LRRK2 has been found to interact with various presynaptic proteins: AP3, clathrin,

dynamin-1 (Schreij et al., 2015; Stafa, et al., 2014; Waschbusch et al., 2014) .These presynaptic

proteins are important for maintaining reserve vesicle pools and membrane fusion. LRRK2 binds

to purified synaptic vesicles and perhaps regulates exocytosis, modulating vesicle pool

mobilization (Piccoli et al., 2014) . We identified a genetic modifier of LRRK2 parkinsonism

that is heavily involved in synaptic vesicle fission and release of clathrin (S. M. Ferguson & De

Camilli, 2012; Raimondi et al., 2011; Wu et al., 2014) . DNM3 rs2421947 GG is associated with

earlier age at onset and higher gene expression in human control striatum. The dynamin 3b

isoform which is involved in regulation of actin polymerization, filopodia and spine formation is

also more highly expressed. Lastly, a significant redistribution of dendritic dynamin 3 staining is

observed in LRRK2 p.G2019S murine cortical culture. The discovery directs therapeutic

development to dynamin 3, as a neuroprotective strategy for LRRK2 parkinsonism or subjects

with Parkinson disease. Diagnostics/therapeutics targeting (a) DNM3 nucleic acid , (b) reducing

the levels of DNM3 GTPase activity, protein or mRNA may help delay the onset of and prevent

symptom progression since the higher gene expression is associated with earlier age of onset.

The therapeutic target may even be generalizable to treat other neurodegenerative disorders with

similar pathogenesis such as Alzheimer's disease, Huntington disease, immune and inflammatory

disorders. Suppression of dynamin GTPase decreases a-synuclein uptake by neuronal and

oligodendroglial cells and amyloid-beta internalization (Konno, et al., 2012; Yu, Nwabuisi-

Page 139: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

119

Heath, Laxton, & Ladu, 2010) . Drp-1 (dynamin-related protein like 1) inhibitors were shown to

protect against ischemic neuronal injury through inhibiting mitochondrial calcium uptake (Tian

et al., 2014). Preliminary evidence has shown that small molecule dynamin inhibitors can also be

an anticonvulsant drug, acting to control synaptic transmission as a novel target for epilepsy.

A limitation is the therapeutic potential of dynamin 3. Thus far, there has not been a

specific drug to readily target dynamin 3, although non-specific inhibitors such as dynasore

(which inhibits GTPase activity of dynamin I and dynamin II but not dynamin III) exist. Other

more potent series include: dimeric tyrphostins, long chain amines and ammonium salts

(myristyl trimethyl ammonium bromides), dynoles, iminodyns and pthaladyns are other drugs

that inhibit dynamin. These drugs have been considered in cancer treatments to induce apoptosis

following cytokinesis failure in a concentration-dependent manner (Chircop et al., 2011; Joshi,

Braithwaite, Robinson, & Chircop, 2011) . Another disadvantage is that pharmacological

inhibition of dynamin in mice has reduced long-term potentiation and resulted in memory loss

(Fa, Staniszewski, Saeed, Francis, & Arancio, 2014) . This limitation can be overcome with

careful monitoring of dynamin inhibitor levels. Perhaps a moderate to low level of inhibitor will

be beneficial and neuroprotective whereas more potent levels lead to apoptosis. Alternatively,

allosteric modulators of dynamin GTPase activity might be considered.

5.5. Conclusion

The identification of risk loci, genes and mutations in PD has provided new insights into

disease aetiology and highlighted new study approaches. Several biological processes involved

in PD pathogenesis have been highlighted, and the discovery of novel PD-associated genes in

families with Mendelian disease has been particularly informative in this regard. Historically,

Page 140: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

120

each major discovery has defined a major theme for translational neuroscience. For example, the

discovery of α‑synuclein as a key component of Lewy bodies highlighted protein aggregation

and propagation, and the discovery of parkin highlighted protein ubiquitination and the

proteosome. Each discovery generally led to a change and/or replacement of focus. Recently,

some pathways have emerged that relate to mitochondrial metabolism (PINK1, PARK2) and

lysosomal-autophagy (ATP13A2, GBA, LRRK2). Nevertheless, the ultimate focus must be on

late-onset Lewy body PD as, clinically and pathologically, this phenotype describes the vast

majority of patients.

On the basis of the finding of DNM3 as a penetrance modifier of LRRK2 parkinsonism,

we postulate a unifying synthesis whereby deficits in synaptic exocytosis and endocytosis

involving DNAJC6, DNAJC13, VPS35, SNCA and LRRK2 are relevant for the clinical

phenotype of disease onset. With the advent of next-generation sequencing, we anticipate

genetic advances in PD will continue to flourish, and our understanding of the molecular

mechanisms underlying susceptibility, progression and response to treatment will continue to

evolve.

Page 141: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

121

References

Aasly, J. O., Toft, M., Fernandez-Mata, I., Kachergus, J., Hulihan, M., White, L. R., et al.

(2005). Clinical features of LRRK2-associated Parkinson's disease in central Norway.

Ann Neurol, 57(5), 762-765.

Abecasis, G. R., Cherny, S. S., Cookson, W. O., & Cardon, L. R. (2002). Merlin--rapid analysis

of dense genetic maps using sparse gene flow trees. Nat Genet, 30(1), 97-101.

Alcalay, R. N., Mejia-Santana, H., Mirelman, A., Saunders-Pullman, R., Raymond, D., Palmese,

C., et al. (2015). Neuropsychological performance in LRRK2 G2019S carriers with

Parkinson's disease. Parkinsonism Relat Disord, 21(2), 106-110.

Alcalay, R. N., Mejia-Santana, H., Tang, M. X., Rosado, L., Verbitsky, M., Kisselev, S., et al.

(2009). Motor phenotype of LRRK2 G2019S carriers in early-onset Parkinson disease.

Arch Neurol, 66(12), 1517-1522.

Alves, G., Wentzel-Larsen, T., & Larsen, J. P. (2004). Is fatigue an independent and persistent

symptom in patients with Parkinson disease? Neurology, 63(10), 1908-1911.

Ando, M., Funayama, M., Li, Y., Kashihara, K., Murakami, Y., Ishizu, N., et al. (2012). VPS35

mutation in Japanese patients with typical Parkinson's disease. Mov Disord, 27(11),

1413-1417.

Annesi, G., Savettieri, G., Pugliese, P., D'Amelio, M., Tarantino, P., Ragonese, P., et al. (2005).

DJ-1 mutations and parkinsonism-dementia-amyotrophic lateral sclerosis complex. Ann

Neurol, 58(5), 803-807.

Arranz, A. M., Delbroek, L., Van Kolen, K., Guimaraes, M. R., Mandemakers, W., Daneels, G.,

et al. (2015). LRRK2 functions in synaptic vesicle endocytosis through a kinase-

dependent mechanism. J Cell Sci, 128(3), 541-552.

Auton, A., Brooks, L. D., Durbin, R. M., Garrison, E. P., Kang, H. M., Korbel, J. O., et al.

(2015). A global reference for human genetic variation. Nature, 526(7571), 68-74.

Beccano-Kelly, D. A., Kuhlmann, N., Tatarnikov, I., Volta, M., Munsie, L. N., Chou, P., et al.

(2014). Synaptic function is modulated by LRRK2 and glutamate release is increased in

cortical neurons of G2019S LRRK2 knock-in mice. Front Cell Neurosci, 8, 301.

Beilina, A., Rudenko, I. N., Kaganovich, A., Civiero, L., Chau, H., Kalia, S. K., et al. (2014).

Unbiased screen for interactors of leucine-rich repeat kinase 2 supports a common

pathway for sporadic and familial Parkinson disease. Proc Natl Acad Sci U S A, 111(7),

2626-2631.

Bhalla, A., Vetanovetz, C. P., Morel, E., Chamoun, Z., Di Paolo, G., & Small, S. A. (2012). The

location and trafficking routes of the neuronal retromer and its role in amyloid precursor

protein transport. Neurobiol Dis, 47(1), 126-134.

Bi, F., Li, F., Huang, C., & Zhou, H. (2013). Pathogenic mutation in VPS35 impairs its

protection against MPP(+) cytotoxicity. Int J Biol Sci, 9(2), 149-155.

Bonifati, V., Rizzu, P., van Baren, M. J., Schaap, O., Breedveld, G. J., Krieger, E., et al. (2003).

Mutations in the DJ-1 gene associated with autosomal recessive early-onset

parkinsonism. Science, 299(5604), 256-259.

Bower, J. H., Maraganore, D. M., McDonnell, S. K., & Rocca, W. A. (1999). Incidence and

distribution of parkinsonism in Olmsted County, Minnesota, 1976-1990. Neurology,

52(6), 1214-1220.

Page 142: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

122

Braak, H., Del Tredici, K., Rub, U., de Vos, R. A., Jansen Steur, E. N., & Braak, E. (2003).

Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging,

24(2), 197-211.

Brockmann, K., Schulte, C., Hauser, A. K., Lichtner, P., Huber, H., Maetzler, W., et al. (2013).

SNCA: major genetic modifier of age at onset of Parkinson's disease. Mov Disord, 28(9),

1217-1221.

Brown, R. G., Dittner, A., Findley, L., & Wessely, S. C. (2005). The Parkinson fatigue scale.

Parkinsonism Relat Disord, 11(1), 49-55.

Browning, B. L., & Browning, S. R. (2008). Haplotypic analysis of Wellcome Trust Case

Control Consortium data. Hum Genet, 123(3), 273-280.

Browning, B. L., & Browning, S. R. (2009). A unified approach to genotype imputation and

haplotype-phase inference for large data sets of trios and unrelated individuals. Am J

Hum Genet, 84(2), 210-223.

Bruggemann, N., Kock, N., Lohmann, K., Konig, I. R., Rakovic, A., Hagenah, J., et al. (2009).

The D216H variant in the DYT1 gene: a susceptibility factor for dystonia in familial

cases? Neurology, 72(16), 1441-1443.

Burn, D. J., Rowan, E. N., Allan, L. M., Molloy, S., O'Brien, J. T., & McKeith, I. G. (2006).

Motor subtype and cognitive decline in Parkinson's disease, Parkinson's disease with

dementia, and dementia with Lewy bodies. J Neurol Neurosurg Psychiatry, 77(5), 585-

589.

Cao, H., Garcia, F., & McNiven, M. A. (1998). Differential distribution of dynamin isoforms in

mammalian cells. Mol Biol Cell, 9(9), 2595-2609.

Chartier-Harlin, M. C., Dachsel, J. C., Vilarino-Guell, C., Lincoln, S. J., Lepretre, F., Hulihan,

M. M., et al. (2011). Translation initiator EIF4G1 mutations in familial Parkinson

disease. Am J Hum Genet, 89(3), 398-406.

Chartier-Harlin, M. C., Kachergus, J., Roumier, C., Mouroux, V., Douay, X., Lincoln, S., et al.

(2004). Alpha-synuclein locus duplication as a cause of familial Parkinson's disease.

Lancet, 364(9440), 1167-1169.

Chaudhuri, K. R., Healy, D. G., Schapira, A. H., & National Institute for Clinical, E. (2006).

Non-motor symptoms of Parkinson's disease: diagnosis and management. Lancet Neurol,

5(3), 235-245.

Chircop, M., Perera, S., Mariana, A., Lau, H., Ma, M. P., Gilbert, J., et al. (2011). Inhibition of

dynamin by dynole 34-2 induces cell death following cytokinesis failure in cancer cells.

Mol Cancer Ther, 10(9), 1553-1562.

Cilia, R., Siri, C., Rusconi, D., Allegra, R., Ghiglietti, A., Sacilotto, G., et al. (2014). LRRK2

mutations in Parkinson's disease: confirmation of a gender effect in the Italian population.

Parkinsonism Relat Disord, 20(8), 911-914.

Clague, M. J., & Rochin, L. (2016). Parkinson's Disease: A Traffic Jam? Curr Biol, 26(8), R332-

334.

Clark, L. N., Wang, Y., Karlins, E., Saito, L., Mejia-Santana, H., Harris, J., et al. (2006).

Frequency of LRRK2 mutations in early- and late-onset Parkinson disease. Neurology,

67(10), 1786-1791.

Cohen, J., Pertsemlidis, A., Kotowski, I. K., Graham, R., Garcia, C. K., & Hobbs, H. H. (2005).

Low LDL cholesterol in individuals of African descent resulting from frequent nonsense

mutations in PCSK9. Nat Genet, 37(2), 161-165.

Page 143: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

123

Cookson, M. R. (2010). The role of leucine-rich repeat kinase 2 (LRRK2) in Parkinson's disease.

Nat Rev Neurosci, 11(12), 791-797.

Cookson, M. R., Lockhart, P. J., McLendon, C., O'Farrell, C., Schlossmacher, M., & Farrer, M.

J. (2003). RING finger 1 mutations in Parkin produce altered localization of the protein.

Hum Mol Genet, 12(22), 2957-2965.

Corvol, H., Blackman, S. M., Boelle, P. Y., Gallins, P. J., Pace, R. G., Stonebraker, J. R., et al.

(2015). Genome-wide association meta-analysis identifies five modifier loci of lung

disease severity in cystic fibrosis. Nat Commun, 6, 8382.

Crosby, J., Peloso, G. M., Auer, P. L., Crosslin, D. R., Stitziel, N. O., Lange, L. A., et al. (2014).

Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J

Med, 371(1), 22-31.

Cuyvers, E., De Roeck, A., Van den Bossche, T., Van Cauwenberghe, C., Bettens, K.,

Vermeulen, S., et al. (2015). Mutations in ABCA7 in a Belgian cohort of Alzheimer's

disease patients: a targeted resequencing study. Lancet Neurol, 14(8), 814-822.

Dawn Teare, M., & Barrett, J. H. (2005). Genetic linkage studies. Lancet, 366(9490), 1036-1044.

de Lau, L. M., & Breteler, M. M. (2006). Epidemiology of Parkinson's disease. Lancet Neurol,

5(6), 525-535.

Dhungel, N., Eleuteri, S., Li, L. B., Kramer, N. J., Chartron, J. W., Spencer, B., et al. (2015).

Parkinson's disease genes VPS35 and EIF4G1 interact genetically and converge on alpha-

synuclein. Neuron, 85(1), 76-87.

Di Fonzo, A., Chien, H. F., Socal, M., Giraudo, S., Tassorelli, C., Iliceto, G., et al. (2007).

ATP13A2 missense mutations in juvenile parkinsonism and young onset Parkinson

disease. Neurology, 68(19), 1557-1562.

Emond, M. J., Louie, T., Emerson, J., Zhao, W., Mathias, R. A., Knowles, M. R., et al. (2012).

Exome sequencing of extreme phenotypes identifies DCTN4 as a modifier of chronic

Pseudomonas aeruginosa infection in cystic fibrosis. Nat Genet, 44(8), 886-889.

Erogullari, A., Hollstein, R., Seibler, P., Braunholz, D., Koschmidder, E., Depping, R., et al.

(2014). THAP1, the gene mutated in DYT6 dystonia, autoregulates its own expression.

Biochim Biophys Acta, 1839(11), 1196-1204.

Fa, M., Staniszewski, A., Saeed, F., Francis, Y. I., & Arancio, O. (2014). Dynamin 1 is required

for memory formation. PLoS One, 9(3), e91954.

Fahn, S. (2000). The spectrum of levodopa-induced dyskinesias. Ann Neurol, 47(4 Suppl 1), S2-

9; discussion S9-11.

Fahn, S. (2003). Description of Parkinson's disease as a clinical syndrome. Ann N Y Acad Sci,

991, 1-14.

Farrer, M., Chan, P., Chen, R., Tan, L., Lincoln, S., Hernandez, D., et al. (2001). Lewy bodies

and parkinsonism in families with parkin mutations. Ann Neurol, 50(3), 293-300.

Farrer, M., Kachergus, J., Forno, L., Lincoln, S., Wang, D. S., Hulihan, M., et al. (2004).

Comparison of kindreds with parkinsonism and alpha-synuclein genomic multiplications.

Ann Neurol, 55(2), 174-179.

Fellner, L., Wenning, G. K., & Stefanova, N. (2015). Models of multiple system atrophy. Curr

Top Behav Neurosci, 22, 369-393.

Ferguson, L. W., Rajput, M. L., Muhajarine, N., Shah, S. M., & Rajput, A. (2008). Clinical

features at first visit and rapid disease progression in Parkinson's disease. Parkinsonism

Relat Disord, 14(5), 431-435.

Page 144: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

124

Ferguson, S. M., & De Camilli, P. (2012). Dynamin, a membrane-remodelling GTPase. Nat Rev

Mol Cell Biol, 13(2), 75-88.

Forman, M. S., Schmidt, M. L., Kasturi, S., Perl, D. P., Lee, V. M., & Trojanowski, J. Q. (2002).

Tau and alpha-synuclein pathology in amygdala of Parkinsonism-dementia complex

patients of Guam. Am J Pathol, 160(5), 1725-1731.

Fuchs, J., Nilsson, C., Kachergus, J., Munz, M., Larsson, E. M., Schule, B., et al. (2007).

Phenotypic variation in a large Swedish pedigree due to SNCA duplication and

triplication. Neurology, 68(12), 916-922.

Fuchs, J., Tichopad, A., Golub, Y., Munz, M., Schweitzer, K. J., Wolf, B., et al. (2008). Genetic

variability in the SNCA gene influences alpha-synuclein levels in the blood and brain.

FASEB J, 22(5), 1327-1334.

Fuchs, T., Gavarini, S., Saunders-Pullman, R., Raymond, D., Ehrlich, M. E., Bressman, S. B., et

al. (2009). Mutations in the THAP1 gene are responsible for DYT6 primary torsion

dystonia. Nat Genet, 41(3), 286-288.

Funayama, M., Ohe, K., Amo, T., Furuya, N., Yamaguchi, J., Saiki, S., et al. (2015). CHCHD2

mutations in autosomal dominant late-onset Parkinson's disease: a genome-wide linkage

and sequencing study. Lancet Neurol, 14(3), 274-282.

Gan-Or, Z., Giladi, N., Rozovski, U., Shifrin, C., Rosner, S., Gurevich, T., et al. (2008).

Genotype-phenotype correlations between GBA mutations and Parkinson disease risk

and onset. Neurology, 70(24), 2277-2283.

Goedert, M., Spillantini, M. G., Del Tredici, K., & Braak, H. (2013a). 100 years of Lewy

pathology. Nat Rev Neurol, 9(1), 13-24.

Goedert, M., Spillantini, M. G., Del Tredici, K., & Braak, H. (2013b). 100 years of Lewy

pathology. [Research Support, Non-U.S. Gov't]. Nature reviews. Neurology, 9(1), 13-24.

Goetz, C. G., Nutt, J. G., & Stebbins, G. T. (2008). The Unified Dyskinesia Rating Scale:

presentation and clinimetric profile. Mov Disord, 23(16), 2398-2403.

Goetz, C. G., Tilley, B. C., Shaftman, S. R., Stebbins, G. T., Fahn, S., Martinez-Martin, P., et al.

(2008). Movement Disorder Society-sponsored revision of the Unified Parkinson's

Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results.

Mov Disord, 23(15), 2129-2170.

Golbe, L. I. (1999). Alpha-synuclein and Parkinson's disease. Mov Disord, 14(1), 6-9.

Goldwurm, S., Tunesi, S., Tesei, S., Zini, M., Sironi, F., Primignani, P., et al. (2011). Kin-cohort

analysis of LRRK2-G2019S penetrance in Parkinson's disease. Mov Disord, 26(11),

2144-2145.

Goldwurm, S., Zini, M., Mariani, L., Tesei, S., Miceli, R., Sironi, F., et al. (2007). Evaluation of

LRRK2 G2019S penetrance: relevance for genetic counseling in Parkinson disease.

Neurology, 68(14), 1141-1143.

Gray, N. W., Kruchten, A. E., Chen, J., & McNiven, M. A. (2005). A dynamin-3 spliced variant

modulates the actin/cortactin-dependent morphogenesis of dendritic spines. J Cell Sci,

118(Pt 6), 1279-1290.

Gregory, A., & Hayflick, S. J. (2011). Genetics of neurodegeneration with brain iron

accumulation. Curr Neurol Neurosci Rep, 11(3), 254-261.

Grunewald, A., Gegg, M. E., Taanman, J. W., King, R. H., Kock, N., Klein, C., et al. (2009).

Differential effects of PINK1 nonsense and missense mutations on mitochondrial

function and morphology. Exp Neurol, 219(1), 266-273.

Page 145: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

125

Guettard, E., Portnoi, M. F., Lohmann-Hedrich, K., Keren, B., Rossignol, S., Winkler, S., et al.

(2008). Myoclonus-dystonia due to maternal uniparental disomy. Arch Neurol, 65(10),

1380-1385.

Gustavsson, E. K., Guella, I., Trinh, J., Szu-Tu, C., Rajput, A., Rajput, A. H., et al. (2015).

Genetic variability of the retromer cargo recognition complex in parkinsonism. Mov

Disord, 30(4), 580-584.

Gustavsson, E. K., Trinh, J., Guella, I., Vilarino-Guell, C., Appel-Cresswell, S., Stoessl, A. J., et

al. (2015). DNAJC13 genetic variants in parkinsonism. Mov Disord, 30(2), 273-278.

Haack, T. B., Hogarth, P., Gregory, A., Prokisch, H., & Hayflick, S. J. (2013). BPAN: the only

X-linked dominant NBIA disorder. Int Rev Neurobiol, 110, 85-90.

Haack, T. B., Hogarth, P., Kruer, M. C., Gregory, A., Wieland, T., Schwarzmayr, T., et al.

(2012). Exome sequencing reveals de novo WDR45 mutations causing a phenotypically

distinct, X-linked dominant form of NBIA. Am J Hum Genet, 91(6), 1144-1149.

Harold, D., Abraham, R., Hollingworth, P., Sims, R., Gerrish, A., Hamshere, M. L., et al. (2009).

Genome-wide association study identifies variants at CLU and PICALM associated with

Alzheimer's disease. Nat Genet, 41(10), 1088-1093.

Hasegawa, K., Stoessl, A. J., Yokoyama, T., Kowa, H., Wszolek, Z. K., & Yagishita, S. (2009).

Familial parkinsonism: study of original Sagamihara PARK8 (I2020T) kindred with

variable clinicopathologic outcomes. Parkinsonism Relat Disord, 15(4), 300-306.

Haugarvoll, K., Rademakers, R., Kachergus, J. M., Nuytemans, K., Ross, O. A., Gibson, J. M., et

al. (2008). Lrrk2 R1441C parkinsonism is clinically similar to sporadic Parkinson

disease. Neurology, 70(16 Pt 2), 1456-1460.

Haugarvoll, K., & Wszolek, Z. K. (2009). Clinical features of LRRK2 parkinsonism.

Parkinsonism Relat Disord, 15 Suppl 3, S205-208.

Hauser, E. R., Watanabe, R. M., Duren, W. L., Bass, M. P., Langefeld, C. D., & Boehnke, M.

(2004). Ordered subset analysis in genetic linkage mapping of complex traits. Genet

Epidemiol, 27(1), 53-63.

Healy, D. G., Falchi, M., O'Sullivan, S. S., Bonifati, V., Durr, A., Bressman, S., et al. (2008).

Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated

Parkinson's disease: a case-control study. Lancet Neurol, 7(7), 583-590.

Heckman, M. G., Elbaz, A., Soto-Ortolaza, A. I., Serie, D. J., Aasly, J. O., Annesi, G., et al.

(2014). Protective effect of LRRK2 p.R1398H on risk of Parkinson's disease is

independent of MAPT and SNCA variants. Neurobiol Aging, 35(1), 266 e265-214.

Heckman, M. G., Schottlaender, L., Soto-Ortolaza, A. I., Diehl, N. N., Rayaprolu, S., Ogaki, K.,

et al. (2014). LRRK2 exonic variants and risk of multiple system atrophy. Neurology,

83(24), 2256-2261.

Heckman, M. G., Soto-Ortolaza, A. I., Aasly, J. O., Abahuni, N., Annesi, G., Bacon, J. A., et al.

(2013). Population-specific frequencies for LRRK2 susceptibility variants in the Genetic

Epidemiology of Parkinson's Disease (GEO-PD) Consortium. Mov Disord, 28(12), 1740-

1744.

Helfer, E., Harbour, M. E., Henriot, V., Lakisic, G., Sousa-Blin, C., Volceanov, L., et al. (2013).

Endosomal recruitment of the WASH complex: active sequences and mutations

impairing interaction with the retromer. Biol Cell, 105(5), 191-207.

Hentati, F., Trinh, J., Thompson, C., Nosova, E., Farrer, M. J., & Aasly, J. O. (2014). LRRK2

parkinsonism in Tunisia and Norway: a comparative analysis of disease penetrance.

Neurology, 83(6), 568-569.

Page 146: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

126

Hollingworth, P., Harold, D., Sims, R., Gerrish, A., Lambert, J. C., Carrasquillo, M. M., et al.

(2011). Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP

are associated with Alzheimer's disease. Nat Genet, 43(5), 429-435.

Howey, R., & Cordell, H. J. (2014). Imputation without doing imputation: a new method for the

detection of non-genotyped causal variants. Genet Epidemiol, 38(3), 173-190.

Hulihan, M. M., Ishihara-Paul, L., Kachergus, J., Warren, L., Amouri, R., Elango, R., et al.

(2008). LRRK2 Gly2019Ser penetrance in Arab-Berber patients from Tunisia: a case-

control genetic study. Lancet Neurol, 7(7), 591-594.

Ibanez, P., Bonnet, A. M., Debarges, B., Lohmann, E., Tison, F., Pollak, P., et al. (2004). Causal

relation between alpha-synuclein gene duplication and familial Parkinson's disease.

Lancet, 364(9440), 1169-1171.

Inactivating mutations in NPC1L1 and protection from coronary heart disease. (2014). N Engl J

Med, 371(22), 2072-2082.

Ishihara-Paul, L., Hulihan, M. M., Kachergus, J., Upmanyu, R., Warren, L., Amouri, R., et al.

(2008). PINK1 mutations and parkinsonism. Neurology, 71(12), 896-902.

Johansen, K. K., White, L. R., Farrer, M. J., & Aasly, J. O. (2011). Subclinical signs in LRRK2

mutation carriers. Parkinsonism Relat Disord, 17(7), 528-532.

Johns, M. W. (1991). A new method for measuring daytime sleepiness: the Epworth sleepiness

scale. Sleep, 14(6), 540-545.

Jorgensen, A. B., Frikke-Schmidt, R., Nordestgaard, B. G., & Tybjaerg-Hansen, A. (2014). Loss-

of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med,

371(1), 32-41.

Joshi, S., Braithwaite, A. W., Robinson, P. J., & Chircop, M. (2011). Dynamin inhibitors induce

caspase-mediated apoptosis following cytokinesis failure in human cancer cells and this

is blocked by Bcl-2 overexpression. Mol Cancer, 10, 78.

Kachergus, J., Mata, I. F., Hulihan, M., Taylor, J. P., Lincoln, S., Aasly, J., et al. (2005).

Identification of a novel LRRK2 mutation linked to autosomal dominant parkinsonism:

evidence of a common founder across European populations. Am J Hum Genet, 76(4),

672-680.

Kalia, L. V., Lang, A. E., Hazrati, L. N., Fujioka, S., Wszolek, Z. K., Dickson, D. W., et al.

(2015). Clinical correlations with Lewy body pathology in LRRK2-related Parkinson

disease. JAMA Neurol, 72(1), 100-105.

Kamm, C., Fischer, H., Garavaglia, B., Kullmann, S., Sharma, M., Schrader, C., et al. (2008).

Susceptibility to DYT1 dystonia in European patients is modified by the D216H

polymorphism. Neurology, 70(23), 2261-2262.

Khan, N. L., Jain, S., Lynch, J. M., Pavese, N., Abou-Sleiman, P., Holton, J. L., et al. (2005).

Mutations in the gene LRRK2 encoding dardarin (PARK8) cause familial Parkinson's

disease: clinical, pathological, olfactory and functional imaging and genetic data. Brain,

128(Pt 12), 2786-2796.

Kitada, T., Asakawa, S., Hattori, N., Matsumine, H., Yamamura, Y., Minoshima, S., et al.

(1998). Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism.

Nature, 392(6676), 605-608.

Klein, C. (2014). Genetics in dystonia. Parkinsonism Relat Disord, 20 Suppl 1, S137-142.

Klein, C., & Ziegler, A. (2011). From GWAS to clinical utility in Parkinson's disease. Lancet,

377(9766), 613-614.

Page 147: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

127

Koga, S., Aoki, N., Uitti, R. J., van Gerpen, J. A., Cheshire, W. P., Josephs, K. A., et al. (2015).

When DLB, PD, and PSP masquerade as MSA: an autopsy study of 134 patients.

Neurology, 85(5), 404-412.

Konno, M., Hasegawa, T., Baba, T., Miura, E., Sugeno, N., Kikuchi, A., et al. (2012).

Suppression of dynamin GTPase decreases alpha-synuclein uptake by neuronal and

oligodendroglial cells: a potent therapeutic target for synucleinopathy. Mol

Neurodegener, 7, 38.

Krebs, C. E., Karkheiran, S., Powell, J. C., Cao, M., Makarov, V., Darvish, H., et al. (2013). The

Sac1 domain of SYNJ1 identified mutated in a family with early-onset progressive

Parkinsonism with generalized seizures. Hum Mutat, 34(9), 1200-1207.

Kruger, R., Kuhn, W., Muller, T., Woitalla, D., Graeber, M., Kosel, S., et al. (1998). Ala30Pro

mutation in the gene encoding alpha-synuclein in Parkinson's disease. Nat Genet, 18(2),

106-108.

Kumar, K. R., Weissbach, A., Heldmann, M., Kasten, M., Tunc, S., Sue, C. M., et al. (2012).

Frequency of the D620N mutation in VPS35 in Parkinson disease. Arch Neurol, 69(10),

1360-1364.

Lai, Y. C., Kondapalli, C., Lehneck, R., Procter, J. B., Dill, B. D., Woodroof, H. I., et al. (2015).

Phosphoproteomic screening identifies Rab GTPases as novel downstream targets of

PINK1. EMBO J, 34(22), 2840-2861.

Lambert, J. C., Grenier-Boley, B., Harold, D., Zelenika, D., Chouraki, V., Kamatani, Y., et al.

(2013). Genome-wide haplotype association study identifies the FRMD4A gene as a risk

locus for Alzheimer's disease. Mol Psychiatry, 18(4), 461-470.

Latourelle, J. C., Hendricks, A. E., Pankratz, N., Wilk, J. B., Halter, C., Nichols, W. C., et al.

(2011). Genomewide linkage study of modifiers of LRRK2-related Parkinson's disease.

Mov Disord, 26(11), 2039-2044.

Latourelle, J. C., Sun, M., Lew, M. F., Suchowersky, O., Klein, C., Golbe, L. I., et al. (2008).

The Gly2019Ser mutation in LRRK2 is not fully penetrant in familial Parkinson's

disease: the GenePD study. BMC Med, 6, 32.

Lee, M. J., Mata, I. F., Lin, C. H., Tzen, K. Y., Lincoln, S. J., Bounds, R., et al. (2009).

Genotype-phenotype correlates in Taiwanese patients with early-onset recessive

Parkinsonism. Mov Disord, 24(1), 104-108.

Lesage, S., Anheim, M., Letournel, F., Bousset, L., Honore, A., Rozas, N., et al. (2013). G51D

alpha-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann Neurol,

73(4), 459-471.

Lesage, S., Anheim, M., Letournel, F., Bousset, L., Honore, A., Rozas, N., et al. (2013). G51D

alpha-synuclein mutation causes a novel parkinsonian-pyramidal syndrome. Ann Neurol,

73(4), 459-471.

Lesage, S., Condroyer, C., Klebe, S., Honore, A., Tison, F., Brefel-Courbon, C., et al. (2012).

Identification of VPS35 mutations replicated in French families with Parkinson disease.

Neurology, 78(18), 1449-1450.

Lesage, S., Ibanez, P., Lohmann, E., Pollak, P., Tison, F., Tazir, M., et al. (2005). G2019S

LRRK2 mutation in French and North African families with Parkinson's disease. Ann

Neurol, 58(5), 784-787.

Li, Y. Y., Chen, X. N., Fan, X. X., Zhang, Y. J., Gu, J., Fu, X. W., et al. (2015). Upregulated

dynamin 1 in an acute seizure model and in epileptic patients. Synapse, 69(2), 67-77.

Page 148: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

128

Lill, C. M., Hansen, J., Olsen, J. H., Binder, H., Ritz, B., & Bertram, L. (2015). Impact of

Parkinson's disease risk loci on age at onset. Mov Disord, 30(6), 847-850.

Lockhart, P. J., Bounds, R., Hulihan, M., Kachergus, J., Lincoln, S., Lin, C. H., et al. (2004).

Lack of mutations in DJ-1 in a cohort of Taiwanese ethnic Chinese with early-onset

parkinsonism. Mov Disord, 19(9), 1065-1069.

Lockhart, P. J., Lincoln, S., Hulihan, M., Kachergus, J., Wilkes, K., Bisceglio, G., et al. (2004).

DJ-1 mutations are a rare cause of recessively inherited early onset parkinsonism

mediated by loss of protein function. J Med Genet, 41(3), e22.

Luk, K. C., Kehm, V., Carroll, J., Zhang, B., O'Brien, P., Trojanowski, J. Q., et al. (2012).

Pathological alpha-synuclein transmission initiates Parkinson-like neurodegeneration in

nontransgenic mice. Science, 338(6109), 949-953.

Luk, K. C., Kehm, V. M., Zhang, B., O'Brien, P., Trojanowski, J. Q., & Lee, V. M. (2012).

Intracerebral inoculation of pathological alpha-synuclein initiates a rapidly progressive

neurodegenerative alpha-synucleinopathy in mice. J Exp Med, 209(5), 975-986.

MacLeod, D., Dowman, J., Hammond, R., Leete, T., Inoue, K., & Abeliovich, A. (2006). The

familial Parkinsonism gene LRRK2 regulates neurite process morphology. Neuron,

52(4), 587-593.

MacLeod, D. A., Rhinn, H., Kuwahara, T., Zolin, A., Di Paolo, G., McCabe, B. D., et al. (2013).

RAB7L1 interacts with LRRK2 to modify intraneuronal protein sorting and Parkinson's

disease risk. Neuron, 77(3), 425-439.

Maraganore, D. M., Wilkes, K., Lesnick, T. G., Strain, K. J., de Andrade, M., Rocca, W. A., et

al. (2004). A limited role for DJ1 in Parkinson disease susceptibility. Neurology, 63(3),

550-553.

Marder, K., Wang, Y., Alcalay, R. N., Mejia-Santana, H., Tang, M. X., Lee, A., et al. (2015).

Age-specific penetrance of LRRK2 G2019S in the Michael J. Fox Ashkenazi Jewish

LRRK2 Consortium. Neurology, 85(1), 89-95.

Marras, C., Alcalay, R. N., Caspell-Garcia, C., Coffey, C., Chan, P., Duda, J. E., et al. (2016).

Motor and nonmotor heterogeneity of LRRK2-related and idiopathic Parkinson's disease.

Mov Disord.

Marti-Masso, J. F., Ruiz-Martinez, J., Bolano, M. J., Ruiz, I., Gorostidi, A., Moreno, F., et al.

(2009). Neuropathology of Parkinson's disease with the R1441G mutation in LRRK2.

Mov Disord, 24(13), 1998-2001.

Mata, I. F., Alvarez, V., Coto, E., Blazquez, M., Guisasola, L. M., Salvador, C., et al. (2005).

Homozygous partial genomic triplication of the parkin gene in early-onset parkinsonism.

Neurosci Lett, 380(3), 257-259.

Matta, S., Van Kolen, K., da Cunha, R., van den Bogaart, G., Mandemakers, W., Miskiewicz, K.,

et al. (2012). LRRK2 controls an EndoA phosphorylation cycle in synaptic endocytosis.

Neuron, 75(6), 1008-1021.

McRae, C., Diem, G., Vo, A., O'Brien, C., & Seeberger, L. (2000). Schwab & England:

standardization of administration. Mov Disord, 15(2), 335-336.

Mills, R. D., Mulhern, T. D., Liu, F., Culvenor, J. G., & Cheng, H. C. (2014). Prediction of the

repeat domain structures and impact of parkinsonism-associated variations on structure

and function of all functional domains of leucine-rich repeat kinase 2 (LRRK2). Hum

Mutat, 35(4), 395-412.

Page 149: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

129

Morgan, N. V., Westaway, S. K., Morton, J. E., Gregory, A., Gissen, P., Sonek, S., et al. (2006).

PLA2G6, encoding a phospholipase A2, is mutated in neurodegenerative disorders with

high brain iron. Nat Genet, 38(7), 752-754.

Muenter, M. D., Forno, L. S., Hornykiewicz, O., Kish, S. J., Maraganore, D. M., Caselli, R. J., et

al. (1998). Hereditary form of parkinsonism--dementia. Ann Neurol, 43(6), 768-781.

Munsie, L. N., Milnerwood, A. J., Seibler, P., Beccano-Kelly, D. A., Tatarnikov, I., Khinda, J., et

al. (2015). Retromer-dependent neurotransmitter receptor trafficking to synapses is

altered by the Parkinson's disease VPS35 mutation p.D620N. Hum Mol Genet, 24(6),

1691-1703.

Nalls, M. A., Pankratz, N., Lill, C. M., Do, C. B., Hernandez, D. G., Saad, M., et al. (2014).

Large-scale meta-analysis of genome-wide association data identifies six new risk loci

for Parkinson's disease. Nat Genet, 46(9), 989-993.

Nalls, M. A., Pankratz, N., Lill, C. M., Do, C. B., Hernandez, D. G., Saad, M., et al. (2014).

Large-scale meta-analysis of genome-wide association data identifies six new risk loci

for Parkinson's disease. Nat Genet, 46(9), 989-993.

Nebel, A., Filon, D., Weiss, D. A., Weale, M., Faerman, M., Oppenheim, A., et al. (2000). High-

resolution Y chromosome haplotypes of Israeli and Palestinian Arabs reveal geographic

substructure and substantial overlap with haplotypes of Jews. Hum Genet, 107(6), 630-

641.

Nichols, N., Bras, J. M., Hernandez, D. G., Jansen, I. E., Lesage, S., Lubbe, S., et al. (2015).

EIF4G1 mutations do not cause Parkinson's disease. Neurobiol Aging, 36(8), 2444

e2441-2444.

Nichols, N., Bras, J. M., Hernandez, D. G., Jansen, I. E., Lesage, S., Lubbe, S., et al. (2015).

EIF4G1 mutations do not cause Parkinson's disease. Neurobiol Aging, 36(8), 2444

e2441-2444.

Nichols, R. J., Dzamko, N., Hutti, J. E., Cantley, L. C., Deak, M., Moran, J., et al. (2009).

Substrate specificity and inhibitors of LRRK2, a protein kinase mutated in Parkinson's

disease. Biochem J, 424(1), 47-60.

Nishioka, K., Kefi, M., Jasinska-Myga, B., Wider, C., Vilarino-Guell, C., Ross, O. A., et al.

(2010). A comparative study of LRRK2, PINK1 and genetically undefined familial

Parkinson's disease. J Neurol Neurosurg Psychiatry, 81(4), 391-395.

Nishioka, K., Ross, O. A., Ishii, K., Kachergus, J. M., Ishiwata, K., Kitagawa, M., et al. (2009).

Expanding the clinical phenotype of SNCA duplication carriers. Mov Disord, 24(12),

1811-1819.

Nishioka, K., Wider, C., Vilarino-Guell, C., Soto-Ortolaza, A. I., Lincoln, S. J., Kachergus, J.

M., et al. (2010). Association of alpha-, beta-, and gamma-Synuclein with diffuse lewy

body disease. Arch Neurol, 67(8), 970-975.

Nuytemans, K., Bademci, G., Inchausti, V., Dressen, A., Kinnamon, D. D., Mehta, A., et al.

(2013). Whole exome sequencing of rare variants in EIF4G1 and VPS35 in Parkinson

disease. Neurology, 80(11), 982-989.

Ogaki, K., Koga, S., Heckman, M. G., Fiesel, F. C., Ando, M., Labbe, C., et al. (2015).

Mitochondrial targeting sequence variants of the CHCHD2 gene are a risk for Lewy body

disorders. Neurology, 85(23), 2016-2025.

Olgiati, S., De Rosa, A., Quadri, M., Criscuolo, C., Breedveld, G. J., Picillo, M., et al. (2014).

PARK20 caused by SYNJ1 homozygous Arg258Gln mutation in a new Italian family.

Neurogenetics, 15(3), 183-188.

Page 150: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

130

Oosterveld, L. P., Allen, J. C., Jr., Ng, E. Y., Seah, S. H., Tay, K. Y., Au, W. L., et al. (2015).

Greater motor progression in patients with Parkinson disease who carry LRRK2 risk

variants. Neurology, 85(12), 1039-1042.

Orenstein, S. J., Kuo, S. H., Tasset, I., Arias, E., Koga, H., Fernandez-Carasa, I., et al. (2013).

Interplay of LRRK2 with chaperone-mediated autophagy. Nat Neurosci, 16(4), 394-406.

Ozelius, L. J., Senthil, G., Saunders-Pullman, R., Ohmann, E., Deligtisch, A., Tagliati, M., et al.

(2006). LRRK2 G2019S as a cause of Parkinson's disease in Ashkenazi Jews. N Engl J

Med, 354(4), 424-425.

Paisan-Ruiz, C., Lang, A. E., Kawarai, T., Sato, C., Salehi-Rad, S., Fisman, G. K., et al. (2005).

LRRK2 gene in Parkinson disease: mutation analysis and case control association study.

Neurology, 65(5), 696-700.

Paisan-Ruiz, C., Saenz, A., Lopez de Munain, A., Marti, I., Martinez Gil, A., Marti-Masso, J. F.,

et al. (2005). Familial Parkinson's disease: clinical and genetic analysis of four Basque

families. Ann Neurol, 57(3), 365-372.

Paisan-Ruiz, C., Washecka, N., Nath, P., Singleton, A. B., & Corder, E. H. (2009). Parkinson's

disease and low frequency alleles found together throughout LRRK2. Ann Hum Genet,

73(Pt 4), 391-403.

Papapetropoulos, S., Scaravilli, T., Morris, H., An, S. F., Henderson, D. C., Quinn, N. P., et al.

(2005). Young onset limb spasticity with PSP-like brain and spinal cord NFT-tau

pathology. Neurology, 64(4), 731-733.

Parisiadou, L., Xie, C., Cho, H. J., Lin, X., Gu, X. L., Long, C. X., et al. (2009). Phosphorylation

of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin

cytoskeleton in neuronal morphogenesis. J Neurosci, 29(44), 13971-13980.

Pchelina, S. N., Ivanova, O. N., Emel'ianov, A. K., & Iakimovskii, A. F. (2011). [Clinical

features of LRRK2-associated Parkinson's disease]. Zh Nevrol Psikhiatr Im S S

Korsakova, 111(12), 56-62.

Piccoli, G., Onofri, F., Cirnaru, M. D., Kaiser, C. J., Jagtap, P., Kastenmuller, A., et al. (2014).

Leucine-rich repeat kinase 2 binds to neuronal vesicles through protein interactions

mediated by its C-terminal WD40 domain. Mol Cell Biol, 34(12), 2147-2161.

Pilsl, A., & Winklhofer, K. F. (2012). Parkin, PINK1 and mitochondrial integrity: emerging

concepts of mitochondrial dysfunction in Parkinson's disease. Acta Neuropathol, 123(2),

173-188.

Polymeropoulos, M. H., Lavedan, C., Leroy, E., Ide, S. E., Dehejia, A., Dutra, A., et al. (1997).

Mutation in the alpha-synuclein gene identified in families with Parkinson's disease.

Science, 276(5321), 2045-2047.

Postuma, R. B., Berg, D., Stern, M., Poewe, W., Olanow, C. W., Oertel, W., et al. (2015). MDS

clinical diagnostic criteria for Parkinson's disease. Mov Disord, 30(12), 1591-1601.

Proukakis, C., Dudzik, C. G., Brier, T., MacKay, D. S., Cooper, J. M., Millhauser, G. L., et al.

(2013). A novel alpha-synuclein missense mutation in Parkinson disease. Neurology,

80(11), 1062-1064.

Purcell, S., Neale, B., Todd-Brown, K., Thomas, L., Ferreira, M. A., Bender, D., et al. (2007).

PLINK: a tool set for whole-genome association and population-based linkage analyses.

Am J Hum Genet, 81(3), 559-575.

Quadri, M., Fang, M., Picillo, M., Olgiati, S., Breedveld, G. J., Graafland, J., et al. (2013).

Mutation in the SYNJ1 gene associated with autosomal recessive, early-onset

Parkinsonism. Hum Mutat, 34(9), 1208-1215.

Page 151: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

131

Rademakers, R., Cruts, M., & van Broeckhoven, C. (2004). The role of tau (MAPT) in

frontotemporal dementia and related tauopathies. Hum Mutat, 24(4), 277-295.

Rademakers, R., Dermaut, B., Peeters, K., Cruts, M., Heutink, P., Goate, A., et al. (2003). Tau

(MAPT) mutation Arg406Trp presenting clinically with Alzheimer disease does not share

a common founder in Western Europe. Hum Mutat, 22(5), 409-411.

Raimondi, A., Ferguson, S. M., Lou, X., Armbruster, M., Paradise, S., Giovedi, S., et al. (2011).

Overlapping role of dynamin isoforms in synaptic vesicle endocytosis. Neuron, 70(6),

1100-1114.

Rajput, A., Ross, J. P., Bernales, C. Q., Rayaprolu, S., Soto-Ortolaza, A. I., Ross, O. A., et al.

(2015). VPS35 and DNAJC13 disease-causing variants in essential tremor. Eur J Hum

Genet, 23(6), 887-888.

Rakovic, A., Grunewald, A., Kottwitz, J., Bruggemann, N., Pramstaller, P. P., Lohmann, K., et

al. (2011). Mutations in PINK1 and Parkin impair ubiquitination of Mitofusins in human

fibroblasts. PLoS One, 6(3), e16746.

Ramirez, A., Heimbach, A., Grundemann, J., Stiller, B., Hampshire, D., Cid, L. P., et al. (2006).

Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a

lysosomal type 5 P-type ATPase. Nat Genet, 38(10), 1184-1191.

Reider, C. R., Halter, C. A., Castelluccio, P. F., Oakes, D., Nichols, W. C., & Foroud, T. (2003).

Reliability of reported age at onset for Parkinson's disease. Mov Disord, 18(3), 275-279.

Reubold, T. F., Faelber, K., Plattner, N., Posor, Y., Ketel, K., Curth, U., et al. (2015). Crystal

structure of the dynamin tetramer. Nature, 525(7569), 404-408.

Risch, N., & Merikangas, K. (1996). The future of genetic studies of complex human diseases.

Science, 273(5281), 1516-1517.

Ritz, B., Rhodes, S. L., Bordelon, Y., & Bronstein, J. (2012). alpha-Synuclein genetic variants

predict faster motor symptom progression in idiopathic Parkinson disease. PLoS One,

7(5), e36199.

Roks, G., Dermaut, B., Heutink, P., Julliams, A., Backhovens, H., Van de Broeck, M., et al.

(1999). Mutation screening of the tau gene in patients with early-onset Alzheimer's

disease. Neurosci Lett, 277(2), 137-139.

Ross, O. A., Soto-Ortolaza, A. I., Heckman, M. G., Aasly, J. O., Abahuni, N., Annesi, G., et al.

(2011). Association of LRRK2 exonic variants with susceptibility to Parkinson's disease:

a case-control study. Lancet Neurol, 10(10), 898-908.

Ross, O. A., Toft, M., Whittle, A. J., Johnson, J. L., Papapetropoulos, S., Mash, D. C., et al.

(2006). Lrrk2 and Lewy body disease. Ann Neurol, 59(2), 388-393.

Ruiz-Martinez, J., Gorostidi, A., Ibanez, B., Alzualde, A., Otaegui, D., Moreno, F., et al. (2010).

Penetrance in Parkinson's disease related to the LRRK2 R1441G mutation in the Basque

country (Spain). Mov Disord, 25(14), 2340-2345.

Samaranch, L., Lorenzo-Betancor, O., Arbelo, J. M., Ferrer, I., Lorenzo, E., Irigoyen, J., et al.

(2010). PINK1-linked parkinsonism is associated with Lewy body pathology. Brain,

133(Pt 4), 1128-1142.

Satake, W., Nakabayashi, Y., Mizuta, I., Hirota, Y., Ito, C., Kubo, M., et al. (2009). Genome-

wide association study identifies common variants at four loci as genetic risk factors for

Parkinson's disease. Nat Genet, 41(12), 1303-1307.

Saunders-Pullman, R., Alcalay, R. N., Mirelman, A., Wang, C., Luciano, M. S., Ortega, R. A., et

al. (2015). REM sleep behavior disorder, as assessed by questionnaire, in G2019S

LRRK2 mutation PD and carriers. Mov Disord, 30(13), 1834-1839.

Page 152: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

132

Saunders-Pullman, R., Mirelman, A., Wang, C., Alcalay, R. N., San Luciano, M., Ortega, R., et

al. (2014). Olfactory identification in LRRK2 G2019S mutation carriers: a relevant

marker? Ann Clin Transl Neurol, 1(9), 670-678.

Schapira, A. H. (2015). Glucocerebrosidase and Parkinson disease: Recent advances. Mol Cell

Neurosci, 66(Pt A), 37-42.

Schrag, A., & Schott, J. M. (2006). Epidemiological, clinical, and genetic characteristics of

early-onset parkinsonism. Lancet Neurol, 5(4), 355-363.

Schreij, A. M., Chaineau, M., Ruan, W., Lin, S., Barker, P. A., Fon, E. A., et al. (2015). LRRK2

localizes to endosomes and interacts with clathrin-light chains to limit Rac1 activation.

EMBO Rep, 16(1), 79-86.

Sharma, M., Ioannidis, J. P., Aasly, J. O., Annesi, G., Brice, A., Bertram, L., et al. (2012). A

multi-centre clinico-genetic analysis of the VPS35 gene in Parkinson disease indicates

reduced penetrance for disease-associated variants. J Med Genet, 49(11), 721-726.

Sheerin, U. M., Charlesworth, G., Bras, J., Guerreiro, R., Bhatia, K., Foltynie, T., et al. (2012).

Screening for VPS35 mutations in Parkinson's disease. Neurobiol Aging, 33(4), 838

e831-835.

Sierra, M., Gonzalez-Aramburu, I., Sanchez-Juan, P., Sanchez-Quintana, C., Polo, J. M.,

Berciano, J., et al. (2011). High frequency and reduced penetrance of LRRK2 G2019S

mutation among Parkinson's disease patients in Cantabria (Spain). Mov Disord, 26(13),

2343-2346.

Siitonen, A., Majounie, E., Federoff, M., Ding, J., Majamaa, K., & Singleton, A. B. (2013).

Mutations in EIF4G1 are not a common cause of Parkinson's disease. Eur J Neurol,

20(4), e59.

Simon-Sanchez, J., & Gasser, T. (2015). Parkinson disease GWAS: the question of lumping or

splitting is back again. Neurology, 84(10), 966-967.

Simon-Sanchez, J., van Hilten, J. J., van de Warrenburg, B., Post, B., Berendse, H. W., Arepalli,

S., et al. (2011). Genome-wide association study confirms extant PD risk loci among the

Dutch. Eur J Hum Genet, 19(6), 655-661.

Singleton, A. B., Farrer, M., Johnson, J., Singleton, A., Hague, S., Kachergus, J., et al. (2003).

alpha-Synuclein locus triplication causes Parkinson's disease. Science, 302(5646), 841.

Skipper, L., Wilkes, K., Toft, M., Baker, M., Lincoln, S., Hulihan, M., et al. (2004). Linkage

disequilibrium and association of MAPT H1 in Parkinson disease. Am J Hum Genet,

75(4), 669-677.

Small, S. A., Kent, K., Pierce, A., Leung, C., Kang, M. S., Okada, H., et al. (2005). Model-

guided microarray implicates the retromer complex in Alzheimer's disease. Ann Neurol,

58(6), 909-919.

Soldner, F., Stelzer, Y., Shivalila, C. S., Abraham, B. J., Latourelle, J. C., Barrasa, M. I., et al.

(2016). Parkinson-associated risk variant in distal enhancer of alpha-synuclein modulates

target gene expression. Nature.

Spillantini, M. G., & Goedert, M. (2001). Tau and Parkinson disease. JAMA, 286(18), 2324-

2326.

Spillantini, M. G., Schmidt, M. L., Lee, V. M., Trojanowski, J. Q., Jakes, R., & Goedert, M.

(1997). Alpha-synuclein in Lewy bodies. Nature, 388(6645), 839-840.

Stafa, K., Tsika, E., Moser, R., Musso, A., Glauser, L., Jones, A., et al. (2014). Functional

interaction of Parkinson's disease-associated LRRK2 with members of the dynamin

GTPase superfamily. Hum Mol Genet, 23(8), 2055-2077.

Page 153: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

133

Steger, M., Tonelli, F., Ito, G., Davies, P., Trost, M., Vetter, M., et al. (2016).

Phosphoproteomics reveals that Parkinson's disease kinase LRRK2 regulates a subset of

Rab GTPases. Elife, 5.

Steinberg, F., Gallon, M., Winfield, M., Thomas, E. C., Bell, A. J., Heesom, K. J., et al. (2013).

A global analysis of SNX27-retromer assembly and cargo specificity reveals a function in

glucose and metal ion transport. Nat Cell Biol, 15(5), 461-471.

Tan, L. C., Tanner, C. M., Chen, R., Chan, P., Farrer, M., Hardy, J., et al. (2003). Marked

variation in clinical presentation and age of onset in a family with a heterozygous parkin

mutation. Mov Disord, 18(7), 758-763.

Tian, Y., Li, B., Shi, W. Z., Chang, M. Z., Zhang, G. J., Di, Z. L., et al. (2014). Dynamin-related

protein 1 inhibitors protect against ischemic toxicity through attenuating mitochondrial

Ca2+ uptake from endoplasmic reticulum store in PC12 cells. Int J Mol Sci, 15(2), 3172-

3185.

Tijero, B., Gomez Esteban, J. C., Somme, J., Llorens, V., Lezcano, E., Martinez, A., et al.

(2013). Autonomic dysfunction in parkinsonian LRRK2 mutation carriers. Parkinsonism

Relat Disord, 19(10), 906-909.

Toft, M., Myhre, R., Pielsticker, L., White, L. R., Aasly, J. O., & Farrer, M. J. (2007). PINK1

mutation heterozygosity and the risk of Parkinson's disease. J Neurol Neurosurg

Psychiatry, 78(1), 82-84.

Tomiyama, H., Li, Y., Funayama, M., Hasegawa, K., Yoshino, H., Kubo, S., et al. (2006).

Clinicogenetic study of mutations in LRRK2 exon 41 in Parkinson's disease patients from

18 countries. Mov Disord, 21(8), 1102-1108.

Trinh, J., Amouri, R., Duda, J. E., Morley, J. F., Read, M., Donald, A., et al. (2014).

Comparative study of Parkinson's disease and leucine-rich repeat kinase 2 p.G2019S

parkinsonism. Neurobiol Aging, 35(5), 1125-1131.

Trinh, J., & Farrer, M. (2013). Advances in the genetics of Parkinson disease. Nat Rev Neurol,

9(8), 445-454.

Trinh, J., Farrer, M., Ross, O. A., & Guella, I. (1993). LRRK2-Related Parkinson Disease. In R.

A. Pagon, M. P. Adam, H. H. Ardinger, S. E. Wallace, A. Amemiya, L. J. H. Bean, T. D.

Bird, C. T. Fong, H. C. Mefford, R. J. H. Smith & K. Stephens (Eds.), GeneReviews(R).

Seattle (WA).

Trinh, J., Guella, I., & Farrer, M. J. (2014). Disease penetrance of late-onset parkinsonism: a

meta-analysis. JAMA Neurol, 71(12), 1535-1539.

Trinh, J., Gustavsson, E. K., Guella, I., Vilarino-Guell, C., Evans, D., Encarnacion, M., et al.

(2014). The role of SNCA and MAPT in Parkinson disease and LRRK2 parkinsonism in

the Tunisian Arab-Berber population. Eur J Neurol, 21(11), e91-92.

Trinh, J., Vilarino-Guell, C., & Ross, O. A. (2015). A commentary on fine mapping and

resequencing of the PARK16 locus in Parkinson's disease. J Hum Genet, 60(8), 405-406.

Trojano, L., Moretta, P., Estraneo, A., & Santoro, L. (2010). Neuropsychologic assessment and

cognitive rehabilitation in a patient with locked-in syndrome and left neglect. Arch Phys

Med Rehabil, 91(3), 498-502.

Tsuboi, Y., Dickson, D. W., Nabeshima, K., Schmeichel, A. M., Wszolek, Z. K., Yamada, T., et

al. (2008). Neurodegeneration involving putative respiratory neurons in Perry syndrome.

Acta Neuropathol, 115(2), 263-268.

Ujiie, S., Hatano, T., Kubo, S., Imai, S., Sato, S., Uchihara, T., et al. (2012). LRRK2 I2020T

mutation is associated with tau pathology. Parkinsonism Relat Disord, 18(7), 819-823.

Page 154: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

134

Valente, E. M., Abou-Sleiman, P. M., Caputo, V., Muqit, M. M., Harvey, K., Gispert, S., et al.

(2004). Hereditary early-onset Parkinson's disease caused by mutations in PINK1.

Science, 304(5674), 1158-1160.

Valente, E. M., Salvi, S., Ialongo, T., Marongiu, R., Elia, A. E., Caputo, V., et al. (2004). PINK1

mutations are associated with sporadic early-onset parkinsonism. Ann Neurol, 56(3), 336-

341.

Vargas, K. J., Makani, S., Davis, T., Westphal, C. H., Castillo, P. E., & Chandra, S. S. (2014).

Synucleins regulate the kinetics of synaptic vesicle endocytosis. J Neurosci, 34(28),

9364-9376.

Vilarino-Guell, C., Rajput, A., Milnerwood, A. J., Shah, B., Szu-Tu, C., Trinh, J., et al. (2014).

DNAJC13 mutations in Parkinson disease. Hum Mol Genet, 23(7), 1794-1801.

Vilarino-Guell, C., Ross, O. A., Aasly, J. O., White, L. R., Rajput, A., Rajput, A. H., et al.

(2010). An independent replication of PARK16 in Asian samples. Neurology, 75(24),

2248-2249.

Vilarino-Guell, C., Wider, C., Ross, O. A., Dachsel, J. C., Kachergus, J. M., Lincoln, S. J., et al.

(2011). VPS35 mutations in Parkinson disease. Am J Hum Genet, 89(1), 162-167.

Visser, M., Marinus, J., Stiggelbout, A. M., & Van Hilten, J. J. (2004). Assessment of autonomic

dysfunction in Parkinson's disease: the SCOPA-AUT. Mov Disord, 19(11), 1306-1312.

Visser, M., Marinus, J., van Hilten, J. J., Schipper, R. G., & Stiggelbout, A. M. (2004). Assessing

comorbidity in patients with Parkinson's disease. Mov Disord, 19(7), 824-828.

Vos, M., Verstreken, P., & Klein, C. (2015). Stimulation of electron transport as potential novel

therapy in Parkinson's disease with mitochondrial dysfunction. Biochem Soc Trans,

43(2), 275-279.

Vrabel, M. (2015). Preferred Reporting Items for Systematic Reviews and Meta-Analyses. Oncol

Nurs Forum, 42(5), 552-554.

Wacholder, S., Hartge, P., Struewing, J. P., Pee, D., McAdams, M., Brody, L., et al. (1998). The

kin-cohort study for estimating penetrance. Am J Epidemiol, 148(7), 623-630.

Wang, Y., Shi, M., Chung, K. A., Zabetian, C. P., Leverenz, J. B., Berg, D., et al. (2012).

Phosphorylated alpha-synuclein in Parkinson's disease. Sci Transl Med, 4(121),

121ra120.

Waschbusch, D., Michels, H., Strassheim, S., Ossendorf, E., Kessler, D., Gloeckner, C. J., et al.

(2014). LRRK2 transport is regulated by its novel interacting partner Rab32. PLoS One,

9(10), e111632.

Waters, C. H., Faust, P. L., Powers, J., Vinters, H., Moskowitz, C., Nygaard, T., et al. (1993).

Neuropathology of lubag (x-linked dystonia parkinsonism). Mov Disord, 8(3), 387-390.

Wen, L., Tang, F. L., Hong, Y., Luo, S. W., Wang, C. L., He, W., et al. (2011). VPS35

haploinsufficiency increases Alzheimer's disease neuropathology. J Cell Biol, 195(5),

765-779.

West, A. B., Lockhart, P. J., O'Farell, C., & Farrer, M. J. (2003). Identification of a novel gene

linked to parkin via a bi-directional promoter. J Mol Biol, 326(1), 11-19.

Wilson, G. R., Sim, J. C., McLean, C., Giannandrea, M., Galea, C. A., Riseley, J. R., et al.

(2014). Mutations in RAB39B cause X-linked intellectual disability and early-onset

Parkinson disease with alpha-synuclein pathology. Am J Hum Genet, 95(6), 729-735.

Wright, F. A., Strug, L. J., Doshi, V. K., Commander, C. W., Blackman, S. M., Sun, L., et al.

(2011). Genome-wide association and linkage identify modifier loci of lung disease

severity in cystic fibrosis at 11p13 and 20q13.2. Nat Genet, 43(6), 539-546.

Page 155: IDENTIFICATION OF GENETIC MODIFIERS IN LRRK2 …

135

Wu, Y., O'Toole, E. T., Girard, M., Ritter, B., Messa, M., Liu, X., et al. (2014). A dynamin 1-,

dynamin 3- and clathrin-independent pathway of synaptic vesicle recycling mediated by

bulk endocytosis. Elife, 3, e01621.

Yesavage, J. A., Brink, T. L., Rose, T. L., Lum, O., Huang, V., Adey, M., et al. (1982).

Development and validation of a geriatric depression screening scale: a preliminary

report. J Psychiatr Res, 17(1), 37-49.

Yu, C., Nwabuisi-Heath, E., Laxton, K., & Ladu, M. J. (2010). Endocytic pathways mediating

oligomeric Abeta42 neurotoxicity. Mol Neurodegener, 5, 19.

Zarranz, J. J., Alegre, J., Gomez-Esteban, J. C., Lezcano, E., Ros, R., Ampuero, I., et al. (2004).

The new mutation, E46K, of alpha-synuclein causes Parkinson and Lewy body dementia.

Ann Neurol, 55(2), 164-173.

Zimprich, A., Benet-Pages, A., Struhal, W., Graf, E., Eck, S. H., Offman, M. N., et al. (2011). A

mutation in VPS35, encoding a subunit of the retromer complex, causes late-onset

Parkinson disease. Am J Hum Genet, 89(1), 168-175.

Zimprich, A., Biskup, S., Leitner, P., Lichtner, P., Farrer, M., Lincoln, S., et al. (2004).

Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic

pathology. Neuron, 44(4), 601-607.

Zody, M. C., Jiang, Z., Fung, H. C., Antonacci, F., Hillier, L. W., Cardone, M. F., et al. (2008).

Evolutionary toggling of the MAPT 17q21.31 inversion region. Nat Genet, 40(9), 1076-

1083.