215
Neural Substrates of Parkinsons Disease by Yuko Koshimori A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Institute of Medical Science University of Toronto © Copyright by Yuko Koshimori 2016

Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

  • Upload
    others

  • View
    8

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

Neural Substrates of Parkinson’s Disease

by

Yuko Koshimori

A thesis submitted in conformity with the requirements

for the degree of Doctor of Philosophy

Institute of Medical Science

University of Toronto

© Copyright by Yuko Koshimori 2016

Page 2: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

ii

Neural Substrates of Parkinson’s Disease

Yuko Koshimori

Doctor of Philosophy

Institute of Medical Science

University of Toronto

2016

Abstract

Parkinson’s disease is characterized by selective degeneration of dopaminergic neurons in the

substantia nigra, the presence of Lewy bodies and Lewy neurites containing alpha-synuclein

aggregates in such neurons, and neuroinflammation mediated by the activation of microglia and

astrocytes. These pathological changes collectively contribute to the cardinal motor symptoms of

Parkinson’s disease such as resting tremor, bradykinesia, rigidity, and postural instability. In

addition, dysfunction of non-dopaminergic neurons such as serotonergic, noradrenergic, and

cholinergic neurons, the Lewy pathology, and neuroinflammation outside of the nigro-striatal

regions are thought to play a major role in non-motor symptoms of Parkinson’s disease such as

psychiatric problems, cognitive dysfunction including dementia, autonomic symptoms, and

rapid-eye movement sleep disorder. Therefore, Parkinson’s disease is a multisystem disease,

affecting diffuse area of brain and producing various symptoms. Current trends toward use of

data driven analytic methods can facilitate to expand our knowledge about Parkinson’s disease

beyond the classic basal ganglia motor model and DA dysfunction. The general aim of this thesis

is to investigate a whole brain in vivo to further understand the complex neural substrates of the

Page 3: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

iii

disease using multimodal neuroimaging tools and techniques. This thesis has shed light on the

cortical and subcortical structural and functional brain changes underpinning for the clinical

manifestations in Parkinson’s disease. Changes in gray and white matter as well as functional

changes were evident using magnetic resonance imaging (MRI). Gray matter and functional

changes occurred in the frontal cortex while white matter changes occurred more diffuse area of

the brain including bilateral frontal and temporal regions and left parietal and occipital regions as

well as subcortical white matter tracts. These changes were also associated with motor and

cognitive dysfunction, suggesting that structural MRI and diffusion tensor imaging as well as

resting functional MRI may be promising as in vivo biomarkers for Parkinson’s disease. Our

investigation of neuroinflammation using Positron Emission Tomography and an improved

second generation radioligand did not find any changes in the striatum of patients with

Parkinson’s disease. However, demonstrated gray matter, white matter, and functional changes

warrant further investigations of neuroinflammation in extra-striatal and white matter regions.

Page 4: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

iv

Acknowledgements

My PhD experiences supported by amazing individuals have inspired me to further pursuit a

career in brain research. Foremost, I would like to thank my supervisor, Dr. Antonio Strafella

who provided me with this invaluable opportunity to completely my PhD degree in his lab.

Antonio is a role mode for a research scientist, supervisor, and a personal figure. He has been a

reliable and responsible supervisor, providing me with continued solid guidance. I am inspired

by his creative and passionate mind, diligence, positive attitude, warmth, and kindness. I greatly

appreciate his mentorship throughout my PhD years.

I would like to acknowledge my other mentors, Dr. Romina Mizrahi and Dr. Clement Hamani

who have provided me with indispensable feedback, guidance, and research ideas. Thank you to

Dr. Pablo Rusjan for teaching me the fundamentals of PET analyses and assisting me with the

analyses and to Dr. Nancy Lobaugh for her guidance for image quality control and imaging

analyses of MRI. Thanks to Dr. Ariel Graff for his time and effort as an internal examiner.

I would like to thank all of my current and past lab members who have created such a supportive

and enjoyable learning environment over the past years. Because of their knowledge, skills,

kindness, and friendship, my PhD experiences have been incredibly positive and inspiring.

Thanks to Leigh Christopher for providing continued support, laughs, and friendship throughout

my PhD years. Thanks to Sang Soo Cho for her invaluable support and advice, teaching me

every aspect of research. I would also like to acknowledge Marion Criaud, Mark Jacobs, Sarah

Coakley, and Crystal Li for their help and friendship. Thank you to Christine Ghadery, Alex

Mihaescu, and Jin-hee Kim for their warm support and friendship. Thank you to Ji Hyun Ko who

first taught me the PET analysis and provided me continued support throughout the project,

Giovanna Pellachia and Barbara Segura for their help with imaging data processing and analyses,

Kelly Aminian, and Rostom Mabrouk for their support and kindness. I would also like to thank

my friends I made through my graduate studies, Danielle DeSouza, Ivonne Suridjan, Aaron

Kucyi, and Shinichiro Nakajima for all of our shared experiences, friendship and laughter that I

will treasure.

Page 5: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

v

Thank you to Alvina Ng, Laura Nguyen, Anusha Ravichandran, Natalie Freeman, Sajid Shaikh,

Jun Parkes, and Irina Vitcu for all of the research and technical support. Thank you to Dr.

Anthony Lang and Dr. Sylvain Houle for the incredible opportunities and variable input.

I would like to acknowledge the Canadian Institute of Health Research and Parkinson Society

Canada, and Society for Neuroscience for providing financial support for this thesis work and for

providing opportunities to present the data, learn most updated research findings, and interact

with other fellow researchers. I also deeply appreciate time and effort of all the research

participants and their family members.

Lastly, I would like to extend my gratitude to my family in Japan and friends in Canada and

Japan. Thanks to my loving, supportive, and understanding mother and father. They encouraged

me to work hard, persevere, and achieve my goal. I also thank all of my friends for laughs,

kindness, and support over the years.

Without all of these people, my PhD would not have been possible. Thank you very much.

Page 6: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

vi

Contributions

Yuko Koshimori (author) was the sole author of this thesis. All work required for this thesis,

including the planning, writing, analysis and research was performed either in whole or in part by

the author. Contributions from other individuals are acknowledged:

Dr. Antonio Strafella (Supervisor) – provided guidance in planning and execution of projects;

laboratory resources; guidance in interpretation of results and preparation of manuscripts

Dr. Romina Mizrahi – provided mentorship and guidance in interpretation of results

Dr. Clement Hamani – provided mentorship and guidance in interpretation of results

Dr. Leigh Christopher – provided assistance with data collection and experiments and

preparation of manuscripts for Chapters 3, 4 & 5

Dr. Anthony Lang – provided guidance in the write-up of the manuscripts for Chapters 3, 4 & 5

Dr. Sylvain Houle – provided guidance in the write-up of the manuscripts for Chapters 3, 4 & 5

Dr. Sarah Duff-Canning – provided guidance for all of the neuropsychological testing for

Chapters 3

Dr. Barbara Segura – provided assistance with data analysis for Chapters 3

Dr. Nancy Lobaugh – provided guidance with the MRI imaging methods for Chapter 3

Kelly Aminian – provided assistance with preparation of manuscripts for Chapter 3

Page 7: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

vii

Mark Jacobs - provided assistance with preparation of manuscripts for Chapter 4 and 5

Dr. Sang-Soo Cho – provided assistance with data collection and experiments; preparation of

manuscripts for Chapters 4

Dr. Marion Criaud – provided assistance with image quality control for Chapters 4

Dr. Christine Ghadery – provided assistance with preparation of manuscripts for Chapters 4

Sarah Coakeley – provided assistance with preparation of manuscripts for Chapters 4

Madeleine Harris - provided assistance with preparation of manuscripts for Chapters 4

Dr. Ji-Hyun Ko – provided assistance with data collection and experiments and preparation of

manuscripts for Chapters 5

Dr. Pablo Rusjan – provided guidance with the PET imaging analysis for Chapters 5

Dr. Rostom Mabrouk - assistance with the PET imaging analysis for Chapters 5

Dr. Alan Wilson – provided assistance with experiments for Chapter 5

Page 8: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

viii

Table of Contents

Acknowledgements ............................................................................................................ iv

Contribution ....................................................................................................................... vi

Table of Contents ............................................................................................................. viii

List of Tables ................................................................................................................... xvi

List of Figures ................................................................................................................. xvii

List of Abbreviations ....................................................................................................... xix

1.0 Literature Review .......................................................................................................... 1

1.1 Parkinson’s disease ................................................................................................ 1

1.2 Clinical symptoms of Parkinson’s disease ............................................................. 2

1.3 Neuropathology of Parkinson’s disease ................................................................. 3

1.3.1 Dysfunction in dopamine pathways and basal ganglia-thalamocortical

circuits ................................................................................................................... 3

1.3.2 Dysfunction of non-dopaminergic systems ................................................. 5

1.3.3 Lewy pathologies and alpha-synuclein ........................................................ 7

1.3.4 Neuroinflammation .................................................................................... 10

1.3.4.1 Microglia.......................................................................................... 10

1.3.4.2 Astrocytes ........................................................................................ 11

1.3.4.3 Neuroinflammation in aging condition ............................................ 12

1.3.4.4 Neuroinflammation in Parkinson’s disease ..................................... 13

Page 9: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

ix

1.3.4.5 Potential mechanisms underlying for neuroinflammation in

Parkinson’s disease ...................................................................................... 14

1.3.5 Summary of Parkinson’s disease ............................................................... 15

1.3.6 Potential mechanisms underlying neuroimaging changes in Parkinson’s disease

............................................................................................................................ 16

1.4 Magnetic Resonance Imaging .............................................................................. 18

1.4.1 Principles of Magnetic Resonance Imaging .............................................. 18

1.4.2 Gray matter imaging .................................................................................. 19

1.4.2.1 Voxel based morphometry ............................................................... 19

1.4.2.2 Cortical thickness analysis ............................................................... 20

1.4.3 White matter imaging ................................................................................ 22

1.4.3.1 Diffusion-weighted imaging/Diffusion tensor imaging................... 23

1.4.3.2 Diffusion tensor tractography .......................................................... 27

1.4.3.3 Tract-based Spatial Statistics ........................................................... 28

1.4.4 Functional MRI ............................................................................................. 29

1.4.4.1 Blood oxygen level-dependent signal .............................................. 29

1.4.4.2 Resting state functional MRI ........................................................... 30

1.4.4.3 Seed-based functional connectivity analysis ................................... 30

1.4.4.4 Independent Components Analysis ................................................. 31

1.4.4.5 Graph theoretical analysis................................................................ 31

1.4.4.5.1 Processing and analysis steps ................................................ 32

Page 10: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

x

1.4.4.5.2 Graph measures ..................................................................... 33

1.5 Positron Emission Tomography ........................................................................... 36

1.5.1 Principles of Positron Emission Tomography imaging ............................. 36

1.5.2 PET radioligands ....................................................................................... 38

1.5.3 Quantification of PET radioligand binding in vivo ................................... 38

1.5.3.1 Kinetic modeling of time-activity curve .......................................... 38

1.5.3.2 Two-tissue compartment model and its outcome measures ............ 39

1.5.3.3 One-tissue compartment model ....................................................... 40

1.5.3.4 A simplified reference tissue model ................................................ 41

1.6 Neuroimaging studies in Parkinson’s disease ...................................................... 42

1.6.1 Structural changes in Parkinson’s disease ................................................. 42

1.6.2 Brain network changes in Parkinson’s disease using rsfMRI and graph

theoretical approach ............................................................................................ 46

1.6.3 Neuroinflammation in PD using Translocator protein 18 kDa imaging .... 48

1.6.3.1 Translocator protein 18 kDa ............................................................ 48

1.6.3.2 Prototypical TSPO radioligand: [11C]-PK11195 ............................. 50

1.6.3.3 Second-generation TSPO radioligands ............................................ 50

1.6.3.4 TSPO polymorphism and binding affinity of second generation TSPO

radioligands.................................................................................................. 52

1.6.3.5 [18F]-FEPPA .................................................................................... 54

1.6.3.5.1 Evaluations in vitro and ex vivo of [18F]-FEPPA .................. 54

Page 11: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xi

1.6.3.5.2 Evaluation of quantification in vivo of [18F]-FEPPA in human

gray matter ............................................................................................ 55

1.6.3.5.3 Evaluation of quantification in vivo of [18F]-FEPPA in human

white matter .......................................................................................... 56

1.6.3.5.4 TSPO polymorphism and [18F]-FEPPA VT ........................... 56

1.6.3.5.5 [18F]-FEPPA study on neuroinflammation and aging ........... 57

1.6.3.5.6 [18F]-FEPPA studies in clinical populations .......................... 57

1.6.3.5.7 TSPO imaging in Parkinson’s disease ................................... 58

2.0 Aims and Hypotheses ................................................................................................. 60

2.1 Study 1: Imaging changes associated with cognitive abnormalities in Parkinson’s

disease ........................................................................................................................ 60

2.2 Study 2: Disrupted nodal and hub organization account for abnormalities across

brain networks and clinical manifestations of Parkinson’s disease ........................... 61

2.3 Study 3: Imaging striatal microglial activation in patients with Parkinson’s

disease ........................................................................................................................ 61

3.0 Study 1: Imaging changes associated with cognitive abnormalities in Parkinson’s

disease ............................................................................................................................... 62

3.1 Introduction .......................................................................................................... 62

3.2 Methods ............................................................................................................... 65

3.2.1 Participants ................................................................................................ 65

Page 12: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xii

3.2.2 Neuropsychological Assessment ............................................................... 65

3.2.3 MRI Acquisition ........................................................................................ 66

3.2.3.1 Cortical Thickness Analysis ............................................................ 67

3.2.3.2 Subcortical Volume Analysis .......................................................... 67

3.2.3.3 TBSS ................................................................................................ 68

3.2.3.4 Statistical Analysis .......................................................................... 68

3.3 Results .................................................................................................................. 69

3.3.1 Demographic, Clinical, and Cognitive Characteristics .............................. 69

3.3.2 Cortical Thickness ..................................................................................... 71

3.3.3 Subcortical Volume ................................................................................... 74

3.3.4 White matter .............................................................................................. 74

3.4 Discussion ............................................................................................................ 77

4.0 Study 2: Disrupted nodal and hub organization account for brain network abnormalities in

Parkinson’s disease ........................................................................................................... 82

4.1 Introduction .......................................................................................................... 82

4.2 Methods ............................................................................................................... 85

4.2.1 Participants ................................................................................................ 85

4.2.2 MRI image acquisition .............................................................................. 85

4.2.3 rsfMRI preprocessing ................................................................................ 86

4.2.4 Network nodes ........................................................................................... 86

4.2.5 Computation of network measures ............................................................ 90

Page 13: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xiii

4.2.6 Seed-based functional connectivity analysis ............................................. 91

4.2.7 Statistical analysis ...................................................................................... 91

4.3 Results .................................................................................................................. 92

4.3.1 Demographic and clinical characteristics .................................................. 92

4.3.2 Head motion parameters and outliers ........................................................ 94

4.3.3 Group comparisons of graph measures ...................................................... 95

4.3.4 Hub analysis ............................................................................................... 99

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

4.4.1 Changes in the sensorimotor network ...................................................... 102

4.4.2 Changes in the cognitive subnetworks .................................................... 103

4.4.3 Hub reorganization .................................................................................. 104

4.4.4 Conclusions .............................................................................................. 106

5.0 Study 3: Imaging striatal microglial activation in patients with Parkinson’s

disease ............................................................................................................................. 107

5.1 Introduction ........................................................................................................ 107

5.2 Methods ............................................................................................................. 109

5.2.1 Subjects .................................................................................................... 109

5.2.2 PET data acquisition ................................................................................ 109

5.2.3 MRI acquisition ....................................................................................... 110

5.2.4 Input function measurement .................................................................... 111

Page 14: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xiv

5.2.5 Generation of ROI-based time activity curve ......................................... 111

5.2.6 Kinetic analysis ........................................................................................ 112

5.2.7 DNA extraction and polymorphism genotyping ...................................... 112

5.2.8 Statistical analysis .................................................................................... 113

5.3 Results ................................................................................................................ 113

5.3.1 Demographic and clinical characteristics ................................................ 113

5.3.2 Genotype and disease effects on TSPO binding ...................................... 114

5.4 Discussion .......................................................................................................... 117

6.0 General Discussion ................................................................................................... 120

6.1 Summary of Findings ......................................................................................... 120

6.2 Structural and functional changes in sensorimotor nodes in Parkinson’s

disease ...................................................................................................................... 121

6.3 Structural and functional changes in cognitive nodes in Parkinson’s disease ... 123

6.4 Dopaminergic modulation on resting-state functional connectivity .................. 126

6.5 White matter changes in Parkinson’s disease .................................................... 128

6.6 Neuroinflammation in Parkinson’s disease ....................................................... 130

6.6.1 Interindividual variability in total distribution volume .............................. 130

6.6.2 [18F]-FEPPA binding .................................................................................. 132

6.6.3 Neuroinflammation in Parkinson’s disease ................................................ 132

6.7 Neuroimaging biomarkers for Parkinson’s disease ........................................... 133

Page 15: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xv

6.8 Limitations ................................................................................................................ 135

7.0 Conclusion ................................................................................................................ 137

8.0 Future Directions ...................................................................................................... 138

References ....................................................................................................................... 144

Page 16: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xvi

List of Tables

Table 3-1. Demographic, clinical, and cognitive characteristics of PD patients and HCs

........................................................................................................................................... 70

Table 4-1. Node labels of four subnetworks and their MNI coordinates .......................... 88

Table 4-2. Demographic and clinical characteristics of patients with PD and HCs ......... 93

Table 4-3. Head motion parameters and outliers of patients with PD and HCs ............... 94

Table 4-4. Graph measure changes in patients with PD compared with

HCs ................................................................................................................................... 97

Table 4-5. FC of three nodes that showed group differences ........................................... 98

Table 4-6. Hub regions in HCs and patients with PD ..................................................... 101

Table 5-1. Demographic and clinical characteristics and PET measures of HC subjects and PD

patients ............................................................................................................................ 114

Table 5-2. Demographic and clinical characteristics of PD patients .............................. 114

Page 17: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xvii

List of Figures

Figure 1-1. BG thalamocortical model in normal state and parkinsonian state .................. 4

Figure 1-2. Three main cortico-BG thalamocortical circuits: motor, associative, and limbic

circuits. ................................................................................................................................ 5

Figure 1-3. Affected various neurotransmitter systems in PD ............................................ 6

Figure 1-4. Six stages of spread of α-synuclein inclusions across brain regions in idiopathic

PD ....................................................................................................................................... 9

Figure 1-5. Neuronal transmission of α-synuclein aggregates ........................................... 9

Figure 1-6. PD pathology. ................................................................................................. 16

Figure 1-7. Estimations of CTh using slice data. .............................................................. 21

Figure 1-8. Measurement of CTh. ..................................................................................... 22

Figure 1-9. Anisotropic diffusion in white matter ............................................................ 26

Figure 1-10. Spherical tensor shape with three orthogonal eigenvectors and their associated

eigenvalues (λ1, λ2, λ3) .................................................................................................... 26

Figure 1-11. Spherical tensor shapes illustrating various anisotropic degrees ................. 27

Figure 1-12. Mean FA skeleton projected on a skull-stripped DWI image. ..................... 29

Figure 1-13. Graph measures. ........................................................................................... 35

Figure 1-14. Principles of PET ......................................................................................... 37

Figure 1-15. 2TCM. .......................................................................................................... 40

Figure 1-16. 1TCM ........................................................................................................... 40

Page 18: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xviii

Figure 1-17. SRTM. .......................................................................................................... 41

Figure 3-1. a. Cortical areas showing significant cortical thinning in PD patients compared to

HCs. b. Bar graphs on extracted CTh values (mm) from the significant clusters in the left SFG

and left precentral gyrus between HCs and PD patients ................................................... 73

Figure 3-2. Partial correlations with age as a covariate between SFG thickness (extracted from

the significant cluster) and global composite z (left) and executive composite z (right) in 26 PD

patients showing the significant positive correlations.. .................................................... 74

Figure 3-3. a. Clusters of significantly increased MD in 16 patients with PD compared with 15

HCs in TBSS. b. Bar graphs on mean MD derived from the significant clusters in TBSS between

HC and PD groups ... ........................................................................................................ 76

Figure 4-1. Medial and lateral views of brain images with 120 regions of interest of four

subnetworks ...................................................................................................................... 87

Figure 4-2. Dorsal view of brain image presenting four nodes showing group differences

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

Figure 4-3. Caudal view of brain image presenting 10 hub nodes.... ............................. 100

Figure 5-1. Graphs of PVEC VT in the CN and in the putamen.... ................................. 116

Figure 5-2. Graphs of VT in the CN and in the putamen.... ............................................ 116

Figure 6-1. Neuroimaging biomarkers for Parkinson’s disease.... .................................. 135

Page 19: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xix

List of Abbreviations

%COV Percent coefficient of variation

1H Hydrogen

1HMRS Proton magnetic resonance spectroscopy

1TCM One-tissue compartment model

2D Two-dimensional

2TCM Two-tissue compartment model

3D Three-dimensional

α-synuclein alpha-synuclein

ACC Anterior cingulate cortex

ACR Anterior corona radiata

AD Alzheimer’s disease

ADC Apparent diffusion coefficient

AIC Akaike information criterion

AIns Anterior insula

ALIC Anterior limb of internal capsule

ANOVA Analysis of variance

ASSET Array spatial sensitivity encoding technique

ATR Anterior thalamic radiation

AUC Area under the curve

BA Brodmann area

BBB Blood-brain barrier

BC Betweenness centrality

BDI Beck depression inventory

BET Brain extraction tool

BG Basal ganglia

BIC Bayesian information criterion

BOLD Blood oxygen level dependent

BPND Binding potential non-displaceable

CAMH Centre for addiction and mental health

Page 20: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xx

CBR Central benzodiazepine receptors

CC Corpus callosum

CF Concentration of free radioligand in tissue

CN Caudate nucleus

CND Concentration of non-displaceable radioligand in tissue

CNS Central nervous system

CNS Concentration of non-specifically bound radioligand in tissue

CP Concentration of radioactivity in blood plasma

CS Concentration of specifically bound radioligand in tissue

CSF Cerebrospinal fluid

CTA Cortical thickness analysis

CTh Cortical thickness

CT Concentration of radioactivity in tissue

CVLT California verbal learning and memory test

D Effective diffusion tensor

D2 receptor Dopamine D2 receptor

DA Dopamine

DAD Disability assessment for dementia

DAN Dorsal attention network

DBI Diazepam binding inhibitor

D-KEFS Delis-kaplan executive function system

DLB Dementia with Lewy bodies

DLPFC Dorsolateral prefrontal cortex

DMN Default mode network

DRT Dopamine replacement therapy

DTI Diffusion tensor imaging

DWI Diffusion-weighted imaging

EC External capsule

EEG Electroencephalogram

FA Fractional anisotropy

FC Functional connectivity

FLIRT FSL linear registration tool

Page 21: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xxi

fMRI Functional magnetic resonance imaging

FNIRT FSL non-linear registration tool

FOV Field of view

fp Plasma free fraction

FPN Fronto-parietal network

FSL FMRIB software library

FWHM Full width half maximum

GABA γ-aminobutyric acid

GAT Graph-theoretical analysis toolbox

GE General electric

GFAP Glial fibrillary acid protein

GLM General linear model

Gln Glutamine

GM Gray matter

Glu Glutamate

GP Globus pallidus

GPi Globus pallidus pars interna

GTM Geometric transfer matrix

HAB High affinity binder

Hb Hemoglobin

HC Healthy control

HC-HAB Healthy control with high affinity binder

HC-MAB Healthy control with mixed-affinity binder

HIV Human immunodeficiency virus

HRRT High resolution research tomograph

Iba1 Ionized calcium binding adaptor molecule 1

ICA Independent components analysis

ICBM International consortium for brain mapping

ICC Interrater correlation coefficient

ICD Impulse control disorders

ICV Intracranial volume

IFO Inferior fronto-occipital fasciculus

Page 22: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xxii

IL Interleukin

ILF Inferior longitudinal fasciculus

Ins Insula

JHU Johns Hoskins University

JLO Judgment of Line Orientation

keV Kilo-electron volts

LAB Low affinity binder

LDL Low-density lipoprotein

LFP Local field potential

LEDD Levodopa equivalent daily dose

M1 Microglia M1

M1 Primary motor cortex

M2 Microglia M2

MAB Mixed affinity binder

MCI Mild cognitive impairment

MD Mean diffusivity

MDE Major depressive episodes

MDS Movement disorders society

MEG Magnetoencephalography

MFG Middle frontal gyrus

MHC Major histocompatibility complex

MIns Mid-insula

MMSE Mini-mental status examination

MNI Montreal neurological institution

MoCA Montreal cognitive assessment

mPEC Medial prefrontal cortex

MRI Magnetic resonance imaging

MS Multiple sclerosis

NA Nucleus accumbens

NEX Number of excitations

NMDA N-methyl-D-aspartate

NMS Non-motor symptoms

Page 23: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xxiii

NO Nitric oxide

NSAID Nonsteroidal anti-inflammatory drugs

PBR Peripheral benzodiazepine receptors

PCR Polymerase chain reaction

PD Parkinson’s disease

PDD Parkinson’s disease with dementia

PD-HAB Parkinson’s disease with high affinity binder

PD-MAB Parkinson’s disease with mixed affinity binder

PD-MCI Parkinson’s disease Mild Cognitive Impairment

PET Positron emission tomography

PFC Prefrontal cortex

PiB Pittsburgh compound B

PLIC Posterior limb of internal capsule

PostIns Posterior insula

PPN Pedunculopontine nucleus

PVE Partial volume error

PVEC Partial volume error correction

RBD Rapid-eye-movement sleep behavior disorder

RF Radiofrequency

ROI Region of Interest

ROS Reactive oxygen species

rsfMRI Resting state functional magnetic resonance imaging

RSN Resting state network

SAL Salience network

SCR Superior corona radiata

SCZ schizophrenia

SFG Superior frontal gyrus

SLF Superior longitudinal fasciculus

SMA Supplementary motor area

SMN Sensorimotor network

SN Substantial nigra

SNP Single-nucleotide polymorphism

Page 24: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

xxiv

SNpc Substantia nigra pars compacta

SNpr Substantia nigra pars reticulata

SPM Spatial parametric mapping

SPSS Statistical package for the social sciences

SRTM Simplified reference tissue model

STN Subthalamic nucleus

T1 Relaxation time 1

T2 Relaxation time 2

TAC Time-activity curve

TBSS Tract-based spatial statistics

TE Echo Time

TFCE Threshold free cluster enhancement

TI Inversion time

TLR2 Toll-like receptor2

TNF-α Tumor necrosis factor- α

TR Repetition Time

TSPO Translocator protein 18 kDa

UF Uncinate fasciculus

UPDRS Unified Parkinson’s disease rating scale

VBM Voxel based morphometry

Vim Vimentin

VND Non-displaceable distribution volume

VS Specific distribution volume

VT Total distribution volume

WM White matter

WMS Wechsler memory scale

Page 25: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

1

1.0 Literature Review

1.1 Parkinson’s disease

Parkinson’s disease (PD) was originally described as a shaking palsy by James Parkinson in

1817 and later named after him. PD is the second most common age-related

neurodegenerative disease after Alzheimer’s disease (AD) and the most common movement

disorder (Goedert, Spillantini, Del Tredici, & Braak, 2013). It affects about 1% of population

aged over 60 years and 4% of those over 80 in industrial countries. The onset of disease

primarily occurs during the sixth decade, but there is also a young onset between 20 and 50

years of age (Dexter & Jenner, 2013).

PD diagnosis is made based on the unilateral onset of the cardinal motor symptoms such as

tremor, rigidity, and bradykinesia, which later spread to become bilateral, levodopa

responsiveness, and absence of markers suggestive of other disease. Pathologic confirmation

includes alpha-synuclein (α-synuclein) deposition and dopamine (DA) neuronal loss in the

substantia nigra pars compacta (SNpc) (Berg et al., 2014).

Etiology of PD remains unknown. Familial PD accounts for approximately 10% (Dexter &

Jenner, 2013). Gene mutations causing familial PD include autosomal dominant and

autosomal recessive gene mutations in SNCA, LRRK2, and PAKIN (Houlden & Singleton,

2012). The rest comprises of idiopathic PD where both genetic risk and environmental

factors play roles in its etiology.

PD is classically characterized by the cardinal motor symptoms such as tremor, rigidity,

bradykinesia, and postural instability (Dexter & Jenner, 2013). However, it is recognized to

be a more complex disease producing a number of non-motor symptoms (NMS) such as

rapid-eye-movement sleep behavior disorder (RBD), hypomia, constipation, autonomic

dysfunction, visual disturbances, depression, and cognitive impairment. Some of these NMS

often occur well before PD diagnosis (Berg et al., 2014; Langston, 2006; Muzerengi,

Contrafatto, & Chaudhuri, 2007) and are generally associated with increased disability, poor

quality of life and entry into long-term care (Chaudhuri & Schapira, 2009).

Page 26: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

2

The treatment for motor symptoms of PD include DA replacement therapy employing

Levodopa and DA agonists, supported by the use of enzyme inhibitors such as peripheral

dopa decarboxylase inhibitors, catechol-O-methyl transferase inhibitors, and selective

monoamine oxidase type B inhibitors (selegiline and rasagiline) (Dexter & Jenner, 2013;

Tufekci, Meuwissen, Genc, & Genc, 2012). Besides these dopaminergic medications, the

weak N-methyl-D-aspartate receptor antagonist, amantadine and anticholinergics are also in

use. Alternative treatment strategies include deep brain stimulation in more advanced stages

of the disease. Unlike the motor symptoms of PD, the NMS such as cognitive impairment is

not effectively treated because of the limited knowledge of its pathology.

1.2 Clinical symptoms of Parkinson’s disease

In addition to the cardinal motor symptoms such as tremor, rigidity, bradykinesia, and

postural instability (Dexter & Jenner, 2013), other motor features include gait and posture

changes that manifest as festination (rapid shuffling steps with a forward-flexed posture

when walking), speech and swallowing difficulties, and a masklike facial expression and

micrographia (Jankovic, 2008). Among NMS, cognitive impairment frequently occurs in PD.

Twenty to 60% of patients may show impairment in one or more cognitive domains

(Aarsland, Bronnick, & Fladby, 2011; Williams-Gray, Foltynie, Brayne, Robbins, & Barker,

2007).

The most common cognitive abnormality in PD is executive dysfunction (Litvan et al., 2012)

such as problem solving, planning, set-shifting (switching goals or tasks) and inhibition.

Additionally, impairment in attention and working memory, visuospatial function, memory

and language are also reported in PD. PD patients showing these cognitive deficits, but no

significant impairment in daily function are termed Mild Cognitive Impairment in PD (PD-

MCI) (Caviness et al., 2007). The PD-MCI is classified in two ways based on a standard

cognitive assessment scale and/or the neuropsychological testing (Litvan et al., 2012). It is

now evident that PD patients with MCI have a six-fold increase in the risk of developing

dementia (Aarsland et al., 2011; Janvin, Larsen, Aarsland, & Hugdahl, 2006).

Page 27: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

3

1.3 Neuropathology of Parkinson’s disease

Neuropathology of PD is primarily characterized by loss of dopaminergic neurons in the

SNpc, resulting in depletion of dopaminergic input to the striatum as well as widespread

aggregation of α-synuclein, the main component of the Lewy pathologies. However, a

number of other neuropathological processes have been indicated such as dysfunction of

non-dopaminergic systems, mitochondrial dysfunction, impaired autophagy, glutamate (Glu)

excitotoxicity, and neuroinflammation (Dexter & Jenner, 2013). The following sub-sections

will discuss some of the neuropathology of PD including dopaminergic and non-

dopaminergic dysfunction and α-synuclein aggregation, and neuroinflammation that has been

investigated in the study 3 of this thesis.

1.3.1 Dysfunction in dopaminergic pathways and basal ganglia-

thalamocortical circuits

There are four DA pathways of the brain: nigrostriatal, mesocortical, mesolimbic, and

tuberoinfundibular pathways. The first three pathways are most relevant to PD. The

nigrostriatal pathway originates from the SNpc of the midbrain and innervates the striatal

structures including the caudate nucleus (CN) and putamen, which is involved in motor,

cognitive, and behavioural/emotional functions. At a time of the diagnosis of PD, the

majority of dopaminergic neurons in the SNpc have been lost. This causes severe depletion

of dopaminergic input to the striatum, leading to pathophysiological changes within the basal

ganglia (BG) thalamocortical circuits including motor, limbic, and cognitive circuits (Figure

1-1; Figure 1-2). The mesocortical and mesolimbic pathways originate in the ventral

tegmental area of the brainstem. The former pathway innervates most regions of the cortex

with most dense projections to the entorhinal, anterior cingulate, insular and prefrontal

cortices and is associated with learning, memory and cognition (Oades & Halliday, 1987).

The latter pathway innervates mostly the nucleus accumbens (NA) or the ventral striatum

with some projections to the hippocampus, amygdala and thalamus (Oades & Halliday,

1987), and is involved in reward and emotional processing (Oades & Halliday, 1987).

Dysfunctions of these DA pathways and the BG-thalamocortical circuits contribute to motor

Page 28: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

4

and cognitive/behavioural complications of PD such as cardinal motor symptoms (Stoessl,

Lehericy, & Strafella, 2014) as well as impairment in executive function (Floresco &

Magyar, 2006), reward-learning behaviour (Lee & Jeon, 2014), and other neuropsychiatric

complications (i.e., depression, visual hallucinations, and so forth) (Pagonabarraga,

Kulisevsky, Strafella, & Krack, 2015) (Figure 1-1; Figure 1-2).

Figure 1-1. BG thalamocortical model in normal state and parkinsonian state. In the

parkinsonian state, DA depletion in SNpc decreases input to the striatum, subthalamic

nucleus (STN), and globus pallidus pars interna-substantia nigra pars reticulata (GPi-SNpr)

illustrated in dotted lines, and the nuclei in the BG such as STN, GPi-SNpr, and

pedunculopontine nucleus (PPN) in darker color boxes become hyperactive (left).

Reproduced with permission from (Obeso & Lanciego, 2011).

Page 29: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

5

Figure 1-2. Three main cortico-BG thalamocortical circuits: motor, associative, and limbic

circuits. The diagram demonstrates the anatomical and functional organization of circuits.

DA depletion in the posterior putamen affects the motor circuit, producing motor

dysfunction, and the dysfunction in the more anterior and ventral regions subserving the

associative and limbic circuits can affect cognitive and limbic functions. Reproduced with

permission from (Galvan, Devergnas, & Wichmann, 2015).

1.3.2 Dysfunction of non-dopaminergic systems

In addition to the dysfunction of the dopaminergic system caused by the degeneration of

dopaminergic neurons in the SNpc, non-dopaminergic nuclei may also be affected such as

the locus coeruleus (producing nor-epinephirine), raphe nucleus (producing serotonin), basal

nucleus of the Meyert (producing acetylcholine), (Jellinger, 2012) (Figure 1-3). Thus, other

neurotransmitter systems are also affected in PD where the cell bodies can be depleted

anywhere between 30 and 90% (Jellinger, 1991). These neuropathological changes likely

contribute to producing a number of NMS (Dexter & Jenner, 2013).

Page 30: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

6

Figure 1-3. Affected various neurotransmitter systems in PD. Neurodegeneration in the

brainstem nuclei affecting widespread cortical regions as well as the cerebellum in PD.

Reproduced with permission from (Lang & Lozano, 1998), Copyright Massachusetts

Medical Society.

Page 31: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

7

1.3.3 Lewy pathologies and alpha-synuclein

Cellular inclusions in nerve cell bodies and in nerve cell processes were first described in

brain regions outside of the substantia nigra (SN) of post-mortem PD brains by Fritz

Heinrich Lewy in 1912 and later called Lewy body and Lewy neurites, respectively after his

name (Goedert et al., 2013). There are two types of Lewy bodies (i.e. brain stem type and

cortical type) and lewy neurites occurring in the axons and dendrites of affected neurons.

The significance of the inclusions was poorly understood until 1997 when a missense

mutation in alpha-synuclein (α-synuclein) gene was found to cause familial PD

(Polymeropoulos et al., 1997). The α-synuclein is the main component of the Lewy

pathologies in PD (Spillantini et al., 1997). The α-synuclein is composed of 140 amino-acid

proteins (Benskey, Perez, & Manfredsson, 2016) and widely expressed throughout central

and peripheral nervous system. It is primarily considered a presynaptic protein (Iwai et al.,

1995), and thought to play a role in synaptic transmission and vesicular transport (Uversky,

2007) although the exact function remains unclear. The aggregation of α-synuclein from

monomers into fibrils via oligomeric intermediates occurs in the soma and neurites of

neurons in PD (Spillantini et al., 1997; Spillantini, Crowther, Jakes, Hasegawa, & Goedert,

1998) and is thought to be part of the toxic mechanism causing the disease (Glass, Saijo,

Winner, Marchetto, & Gage, 2010).

A staging scheme on the temporal and regional distribution of α-synuclein inclusions

associated with idiopathic PD was proposed by Braak, Tredici, Rub, et al (2003) (Figure 1-

4). It includes six stages (Braak et al., 2003; Braak & Del Tredici, 2009): In stage 1, the

pathology is present in the olfactory bulb, the anterior olfactory nucleus and/or dorsal motor

nucleus of the glossopharyngeal and vagal nerves in the brainstem. In stage 2, it develops in

the medulla oblongata and the pontine tegmentum (locus coeruleus, magnocellular nucleus of

the reticular formation, and lower raphe nuclei). In stage 3, it reaches the pedunculopontine

nucleus, the cholinergic magnocellular nuclei of the basal forebrain, amygdala and the SNpc.

Generally, during this stage the cardinal motor symptoms of PD begin to appear. In stage 4,

it progresses to the hypothalamus, parts of the thalamus, and the anteromedical temporal

mesocortex. During stage 5 and 6, it appears in the cortical regions, from high-order

Page 32: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

8

association areas including the insular and cingulate cortices (stage 5) to secondary and

primary regions.

The α-synuclein deposits are also detected in the spinal cord, the enteric nervous system of

the peripheral nervous system connecting to the brain via the vagal nerve, plexuses in the

gastro-intestinal tract, sympathetic ganglia and trunk, the adrenal medulla, the sub-madibular

gland, and the heart. These α-synuclein deposits outside of the brain and those in the brain

from the stage 1-3 may provide pathological substrates for NMS (e.g., autonomic

dysfunction, hyposmia, depression and RBD) during the premotor stages of the disease

(Benskey et al., 2016; Langston, 2006; Schapira & Tolosa, 2010).

The transmission of α-synuclein between cells was evidenced in post-mortem PD brain

(Kordower, Chu, Hauser, Olanow, & Freeman, 2008; Li et al., 2008) and animals (Desplats

et al., 2009; Hansen et al., 2011). PD patients who received mesencephalic tissue transplants

showed that two to five percent of the grafted neurons displayed α-synuclein aggregates over

five years (Brundin, Li, Holton, Lindvall, & Revesz, 2008; Kordower et al., 2008; Kordower,

Chu, Hauser, Freeman, & Olanow, 2008; Li et al., 2008; Li et al., 2010). In transgenic mice

overexpressing human α-synuclein, the transgenic protein was transferred from the host

neurons to neural stem cells grafted into the hippocampus (Desplats et al., 2009), and to

dopaminergic neurons grafted into the striatum (Hansen et al., 2011).

The mechanisms underlying this cell-to-cell transmission of α-synuclein aggregates can be

exocytosis (Brundin, Melki, & Kopito, 2010; Lee, Bae, & Lee, 2014) and passive release

from cell death (Brundin et al., 2010), and endocytosis of neighboring neurons (Lee et al.,

2014). The spreading of α-synuclein aggregates across brain regions described in Braak’s

staging scheme may occur through trans-synaptic transmission (Brundin et al., 2010;

Hawkes, Del Tredici, & Braak, 2009; Volpicelli-Daley et al., 2011). However, this has not

been confirmed in in vivo models (Lee et al., 2014) (Figure 1-5).

Page 33: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

9

Figure 1-4. Six stages of spread of α-synuclein inclusions across brain regions in idiopathic

PD. Disease progresses with additional brain regions from lower to higher brain regions and

worsening of pathology compared to the previously affected brain regions illustrated from

lighter to darker colors. Reproduced with permission from (Goedert et al., 2013).

Figure 1-5. Neuronal transmission of α-synuclein aggregates. a. Intracellular α-synuclein

aggregates can be released from neurons by exocytosis or cell death. Then, the aggregates are

taken up by neighboring neuronal cell bodies and are either retained in the cell soma (local

spread of pathology) or transported anterogradely by axons. Alternatively, they are taken up

Page 34: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

10

by axon terminals and transported retrogradely to the cell soma. The protein aggregates can

spread between brain regions by axonal transport. b. Appearance of α-synuclein aggregates

from the initial stage in the dorsal motor nucleus of the vagal nerve in the brainstem and

anterior olfactory structures in darkest green to the final stage in the primary and secondary

cortical regions. Reproduced with permission from (Brundin et al., 2010).

1.3.4 Neuroinflammation

Inflammation is generated by an activated immune system, and is a strictly regulated self-

defensive mechanism against pathogenic stimuli or injury to protect the host organism by

clearing pathogenic stimuli or debris and promote the healing process (Khandelwal, Herman,

& Moussa, 2011; Stone, Reynolds, Mosley, & Gendelman, 2009). In the central nervous

system (CNS), glial cells including microglia and astrocytes are primary immune cells

protecting the brain against pathogenic stimuli (Glass et al., 2010).

1.3.4.1 Microglia

Microglia are the key immune cells of the CNS originated from primitive macrophage

progenitors in the yolk sac and migrate into the CNS during early embryogenesis (Saijo &

Glass, 2011). They constitute up to 5-15 % of the brain and 20 % of the glial cell population

(Kofler & Wiley, 2011; J. A. Smith, Das, Ray, & Banik, 2012). In humans, microglia are

present at a higher density in white matter (WM) than gray matter (GM) and localized mostly

in the medulla oblongata, pons, BG, and SN (Mittelbronn, Dietz, Schluesener, &

Meyermann, 2001). Under the normal physiological conditions, microglia have a unique

ramified morphology consisting of a small and round soma with numerous branching

processes (Smith et al., 2012). These “resting” microglia are constantly sampling their

surrounding environment by their processes undergoing repeated cycles of extension and

withdrawal while the cell bodies remaining relatively stable (Venneti, Wiley, & Kofler,

2009). Microglia mediate immune responses (Saijo & Glass, 2011), release neurotropic

factors, remove toxic substances, repair neurons, remodel and prune synapses, and guide

neural stem cells during synaptogenesis (Paolicelli et al., 2011; Ransohoff & Stevens, 2011;

Saijo & Glass, 2011).

Page 35: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

11

Microglia become activated in response to various environmental challenges such as

bacterial and viral molecules, disease proteins (beta-amyloid and α-synuclein) and soluble

mediators released by damaged neurons such as α-synuclein and neuromelanin (Lull &

Block, 2010). The resting ramified microglia shift to the intermediate hyper-ramified

morphology, characterized by a larger soma and an amoeboid morphology to initiate

phagocytosis (Graeber & Streit, 2010). Microglia upregulate cell surface markers of

inflammation, proliferate, migrate to the injured site and secrete pro- and anti-inflammatory

cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1B (IL-1B) and IL-10,

chemokines, oxidative and nitrodative stress-inducing factors such as nitric oxide (NO) and

superoxide (Lull & Block, 2010).

Activated microglia are often divided into two subsets: microglia M1 (M1) and microglia M2

(M2) based on the distinct molecular phenotype and effector function. The M1 phenotype

reflects classical activation where microglia produce oxidative metabolites, proteases, and

proinflammatory cytokines to defend the host organism against pathogens and tumor cells,

whereas the M2 phenotype reflects alternative activation where microglia secrete anti-

inflammatory cytokine IL-10 and transforming growth factor-β to repair and remodel tissue

(Czeh, Gressens, & Kaindl, 2011). However, this classification is overly simplistic in

diseased brains.

1.3.4. 2 Astrocytes

Astrocytes are abundant glial cells in the CNS and are five times greater in number than

neurons (Freeman, 2010). Astrocytes are dispersed throughout the CNS where each astrocyte

has own territorial region without overlapping with others. They exhibit numerous branches

extending through neurons and blood vessels to form functional networks via gap junctions,

called neurovascular units (Aarsland et al., 2011). Two main types of astrocytes in the CNS

include (1) protoplasmic astrocytes localized in the GM and envelope neuronal bodies and

synapses, and (2) fibrous astrocytes in the WM that interact with the nodes of Ranvier and

oligodendroglia (Halliday & Stevens, 2011; Oades & Halliday, 1987).

Astrocytes serve many essential functions: they perform regulatory and supportive roles in

the CNS such as biochemical and nutritional support for neurons, extracellular ion balance,

Page 36: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

12

and repair of scarring of brain and spinal cord tissue (Sofroniew & Vinters, 2010), and form

a bridge between the vascular system and neurons, which makes them a key regulatory

element of neuronal activity and cerebral blood flow. Additionally, astrocytes have immune

functions that limit the spreading of inflammatory cells and infectious agents from the

damaged area to healthy parenchyma (Hamby & Sofroniew, 2010).

When astrocytes become activated, they undergo reactive astrogliosis including scar

formation. During the astrogliosis, astrocytes undergo morphological changes, resulting in

cell hypertrophy (Hamby & Sofroniew, 2010; Sofroniew & Vinters, 2010); upregulate

proteins such as the intermediate filaments vimentin (Vim) and glial fibrillary acid protein

(GFAP) (Alvarez, Katayama, & Prat, 2013); act as immune regulatory cells by presenting

antigens and pro- and anti-inflammatory molecules such as cytokines, chemokines, and

neurotropic factors (Sofroniew & Vinters, 2010); and activate distant microglia (Liu, Tang,

& Feng, 2011).

1.3.4.3 Neuroinflammation in aging condition

Age-associated changes in microglial functions have been characterized in two different

ways: (1) primed, sensitized or reactive and (2) senescent or dystrophic (Norden & Godbout,

2013; Streit & Xue, 2010; Venneti et al., 2009). The primed microglia are an intermediate

activation state, evidenced by morphological activation (deramification) and upregulation of

the major histocompatibility complex (MHC) class II cell surface markers (von Bernhardi,

Tichauer, & Eugenin, 2010), as well as elevated levels of proinflammatory cytokines and

downregulated anti-inflammatory cytokines (Dilger & Johnson, 2008). This creates a chronic

low-level inflammation and increased reactivity to stimuli in aged brain (Streit & Xue,

2010). On the other hand, the senescent microglia was referred to as the functional

impairments of microglia in phagocytosis (Norden & Godbout, 2013), characterized by

reduced phagocytosis of protein by microglia (Hickman, Allison, & El Khoury, 2008; Lee et

al., 2010), delayed recruitment of phagocytic cells and less clearance of debris (Zhao, Li, &

Franklin, 2006), and slower velocity towards the injury site and aggregation at the injury site

for a longer duration (Damani et al., 2011).

Page 37: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

13

Astrocytes also become more inflammatory with age (Norden & Godbout, 2013). For

example, activated astrocytic profiles associated with inflammation (Eng, Ghirnikar, & Lee,

2000) such as increased expression of GFAP and Vim were found in brains of aged humans

(Finch, 2003; Nichols, Day, Laping, Johnson, & Finch, 1993; Porchet et al., 2003). These

activated astrocytes in aged brain may disrupt the communication with neurons and

microglia (Norden & Godbout, 2013).

The increased inflammatory markers on the glial cells in the aged brain set the stage for the

rapid and prolonged expression of pro-inflammatory molecules following a stimulus, which

can cause disease behaviour including cognitive impairment and depression (Norden &

Godbout, 2013; Streit, 2006). In addition, dysfunctional phagocytosis can lead to

accumulation of toxic materials and disease related proteins associated with

neurodegenerative disorders (Streit, 2006), which can induce more glial activation, creating

prolong inflammation that drives the chronic progression of neurodegenerative disorders

(Gao & Hong, 2008).

1.3.4.4 Neuroinflammation in Parkinson’s disease

In post-mortem PD brains, neuroinflammatory processes were confirmed by the presence of

microglia positive to MHC class II (Croisier, Moran, Dexter, Pearce, & Graeber, 2005;

Imamura et al., 2003; McGeer, Itagaki, Boyes, & McGeer, 1988), such as CR3/43 (Banati,

Daniel, & Blunt, 1998; Mirza, Hadberg, Thomsen, & Moos, 2000), ionized calcium binding

adaptor molecule 1 (Iba1) (Doorn, Moors et al., 2014b), as well as CD68, glycoprotein that

binds to low density lipoprotein and expressed on monocytes/macrophages (Doorn et al.,

2014b), and/or by the presence of astrocytes positive to GFAP (Banati et al., 1998; Mirza et

al., 2000; Mythri et al., 2011).

Activated microglial cells were first evidenced in the SN of PD patients (McGeer et al.,

1988). Subsequent studies corroborated their findings (Banati et al., 1998; Croisier et al.,

2005; Desai Bradaric, Patel, Schneider, Carvey, & Hendey, 2012; Doorn et al., 2014b;

Imamura et al., 2003; Mirza et al., 2000). Additionally, microglial activation occurred in

other brain regions such as putamen (Imamura et al., 2003), hippocampus (Doorn et al.,

2014b; Imamura et al., 2003), transentorhinal, cingulate, and temporal cortex (Imamura et

Page 38: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

14

al., 2003), frontal cortex (Vroon et al., 2007), olfactory bulb (Vroon et al., 2007), and

anterior olfactory nucleus (Doorn, Goudriaan et al., 2014) . Reactive astrocytes were also

confirmed in the CN and frontal cortex (Mythri et al., 2011), and temporal and mesocortex

(Braak, Sastre, & Del Tredici, 2007). However, these neuroiflammatory processes were not

associated with clinical measures such as duration of disease (Banati et al., 1998; Croisier et

al., 2005; McGeer et al., 1988) or severity of disease (Mythri et al., 2011).

Inflammatory markers were also elevated including the levels of TNF-α (Mogi et al., 1994;

Nagatsu & Sawada, 2005) as well as IL-1 (Blum-Degen et al., 1995; Nagatsu & Sawada,

2005) and IL-6 in the cerebrospinal fluid (CSF) of PD patients (Blum-Degen et al., 1995;

Mogi et al., 1994; Muller, Blum-Degen, Przuntek, & Kuhn, 1998; Nagatsu & Sawada, 2005).

In addition, there were significant inverse correlations between IL-6 CSF levels and disease

severity measured by Unified Parkinson’s Disease Rating Scale (UPDRS) I, II, III,

bradykinesia, resting tremor, and rigidity (Muller et al., 1998).

1.3.4.5 Potential mechanisms underlying for neuroinflammation in

Parkinson’s disease

Neuroinflammatory processes can be activated by injured neurons releasing noxious self-

compounds such as membrane breakdown products, aggregated α-synuclein, and

neuromelanin in the extracellular milieu in PD (Gao & Hong, 2008). They were evidenced in

post-mortem brains where activated microglia was in close proximity to melanin-containing

dopaminergic cells and to free melanin in the SN (McGeer et al., 1988), as well as α-

synuclein depositions in the SN (Croisier et al., 2005; Doorn et al., 2014b), hippocampus

(Doorn et al., 2014b), cingulate cortex (Croisier et al., 2005), and in damaged dopaminergic

and serotonergic neuritis (Croisier et al., 2005). In addition, α- synuclein-positive astrocytes

were detected in prosencephalic regions such as amygdala, thalamus, septum, striatum,

claustrum and cerebral cortex (Braak et al., 2007).

Recent data also suggest that extracellular α-synuclein oligomers released by exocytosis from

neuronal cells play an important role in initiating neuroinflammatory processes. For example,

they can directly activate microglia by binding Toll-like receptor2 (TLR2) on microglia (Kim

Page 39: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

15

et al., 2013). This response was not observed if microglia lacks TLR2. In addition,

microarray analysis of astrocytes exposed to neuron-released α synuclein induced specific

genes that are involved in proinflammatory responses, such as production of cytokines and

chemokines (Lee, Kim, & Lee, 2010). Proinflammatory reactions of astrocytes could also act

as a precursor for more extensive inflammation, possibly through recruitment and activation

of microglia (Lee et al., 2010). Both microglia and astrocytes were also activated to clear

extracellular α-synuclein aggregates although the former was shown to be principle

scavenger of extracellular α-synuclein aggregates (Lee, Suk, Bae, & Lee, 2008).

PD brain releases a greater amount of α-synuclein aggregates than normal brain (Lee et al.,

2014). Glial overactivation results in increased release of pro-inflammatory cytokines, NO,

and reactive oxygen species (ROS) (Deshpande et al., 2005; Lee et al., 2010; Qian & Flood,

2008; Zhang et al., 2005), which can create a chronic inflammatory microenvironment in PD

patients (Lee et al., 2014).

1.3.5 Summary of Parkinson’s disease

To summarize, PD is characterized by selective degeneration of dopaminergic neurons in the

SN, the presence of Lewy bodies and Lewy neurites containing α-synuclein aggregates in

such neurons, and neuroinflammation mediated by the activation of microglia and astrocytes.

These pathological changes collectively contribute to the cardinal motor symptoms of PD

such as resting tremor, bradykinesia, rigidity, and postural instability. In addition,

dysfunction of non-dopaminergic neurons such as serotonergic, noradrenergic, and

cholinergic neurons, the Lewy pathology, and neuroinflammation outside of the nigro-striatal

regions are thought to play a major role in NMS of PD such as psychiatric problems,

cognitive dysfunction including dementia, autonomic symptoms, and RBD. Therefore, PD is

a multisystem disease, affecting diffuse area of brain and producing various symptoms

(Figure 1-6). The complex neural substrates of PD reflecting these pathological changes can

be studied in vivo using multimodal neuroimaging tools in the attempt of identifying

neuroimaging biomarkers associated with motor and cognitive symptoms.

Page 40: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

16

Figure 1-6. PD pathology. PD is characterized by degeneration of dopaminergic neurons and

non-dopaminergic neurons such as serotonergic, noradrenergic, and cholinergic neurons, as

well as aggregated α-synuclein, which can induce neuroinflammation mediated by the

activation of microglia and astrocytes. These pathological changes collectively contribute to

the cardinal motor symptoms of PD such as resting tremor, bradykinesia, rigidity, and

postural instability and NMS such as cognitive dysfunction, depression, impairment in

reward processing, hyposmia and RBD. Using in vivo neuroimaging tools, brain changes

such as structural as well as functional changes reflecting these pathological changes can be

quantified. Adapted from (Brudin, 2010).

1.3.6 Potential mechanisms underlying neuroimaging changes in

Parkinson’s disease

GM changes in PD reflect different neuropathologies such as dysfunction/degeneration of

different neurotransmitter systems, Lewy pathology and/or secondary effect of Lewy

pathology in subcortical regions, as well as neuroinflammation. These contribute to GM

changes varying from subtle changes such as reduction in size of cell bodies, dendritic

Page 41: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

17

arborisation and presynaptic terminals (Morrison & Hof, 1997; Pellicano et al., 2012) to

synaptic and cell loss. Similarly, WM changes are possibly due to gliosis, Lewy pathologies,

and neuronal loss (Braak et al., 2004). For example, neuronal losses in subcortical projection

nuclei such as the noradrenergic locus ceruleus, the serotonergic raphe nuclei, and the

cholinergic nucleus basalis of Meynert (Jellinger, 1991) likely lead to axonal degeneration.

Lewy pathology can lead to axonal dysfunction and/or altered axonal microstructure

(Bellucci et al., 2016). Postmortem brains with dementia with Lewy bodies (DLB) showed

axonal transport dysfunction leading to accumulation of axonal transported substances

(Katsuse, Iseki, Marui, & Kosaka, 2003). This can change axonal microstructure by swelling

as well as retrograde and anterograde degeneration of the axonal projections (Hattori et al.,

2012), which may eventually leads cell body damage. These GM and WM changes can be

detected using magnetic resonance imaging (MRI).

These structural deteriorations mentioned above would affect functional connectivity (FC) of

the brain regions. In fact, direct comparisons between functional and structural connectivity

in the same individuals suggest that the FC can be predicted from the anatomical structure

(Honey et al., 2009), and the microstructure of cingulum that interconnects key regions of the

default mode network (DMN) was associated with the level of FC between these regions

(van den Heuvel, Mandl, Luigjes, & Hulshoff Pol, 2008). In addition, PD neuropathologies

can directly affect functionality. For example, the level of CSF α-synuclein was associated

with reduced sensorimotor network (SMN) connectivity in PD patients (Campbell et al.,

2015). Furthermore, the associations between neuroinflammation and FC were demonstrated

in patients with multiple sclerosis (Colasanti et al., 2016).

One of the potential mechanisms underlying structural and functional changes detected by

MRI is neuroinflammation, which can be specifically investigated utilizing Positron

Emission Tomography (PET) with a radioligand targeting for translocator protein (TSPO), a

biomarker for microglia activation.

Page 42: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

18

1.4 Magnetic Resonance Imaging

1.4.1 Principles of Magnetic Resonance Imaging

MRI is primarily based on detection of the hydrogen (1H) signal from water because a 1H

nucleus or proton has a significant magnetic moment (meaning a small magnetic field like a

bar magnet generated from its positively charged and spin characteristics) and is abundant in

the body. When the protons are surrounded by a strong external magnetic field, they align in

either parallel or anti-parallel to the magnetic field. More nuclei take the parallel alignment,

as this requires lower energy than the other orientation. The protons also precess at some

frequency. This longitudinal magnetization then reaches thermal equilibrium. In this

condition, however, the individual protons creating the longitudinal magnetization do not

precess in phase. As the result, the longitudinal magnetization does not produce a measurable

signal.

To produce signal, a radiofrequency (RF) pulse in a 90° flip angle (that produces the

maximum signal) is introduced at the frequency at which the protons precess. As the RF

pulse continues, some of the protons in the low energy state absorb energy from the RF field

and make a transition into the high energy state, resulting in “tipping” the longitudinal

magnetization toward the transverse plane. The RF pulse results in the synchronized

precessing of proton spins in the transverse plane, producing an oscillating MR signal in a

receiver coil. After the RF transmitter is turned off, the protons start to lose precessing and

the transverse magnetization decays (T2 relaxation). Simultaneously, the longitudinal

magnetization increases toward its pre-excitation value over time (T1 relaxation). T2*

relaxation describes the decay in the transverse plane following a RF pulse due to

inhomogeneities in the magnetic field that are introduced by objects with magnetic

susceptibility.

The times required for the transverse and longitudinal magnetizations to return to their

equilibrium states are called relaxation times. T1 relaxation time (T1) indicates the time

required for the system to recover 63% of its equilibrium value (generally in the order of

Page 43: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

19

seconds) and the T2 relaxation time (T2) is the time it takes for dephasing to decay to 37% of

its original value (generally tens to hundreds of milliseconds).

Image contrast can be modified by varying how often RF pulses are applied. The interval

between 90° pulses is called the repetition time (TR). The magnitude of the signal detected

also depends on how slowly the transverse magnetization decays from its initial maximum

value. The amount of time allowed for decay to occur following the initial 90° flip is called

the echo time (TE). The TR is shorter than the time necessary for total longitudinal relaxation

produces T1-weighted images. Long TR and long TE create T2-weighted images. When TR

is long so that protons fully recover longitudinal magnetization between repetitions, but TE is

short when the effect of T2 decay is minimized, then the image will not be dependent either

on T1 or T2, but only on the tissue’s proton density and it is called a proton density weighted

image.

1.4.2 Gray matter imaging

Given the sensitivity of T1-weighted imaging in detecting differences between tissue types,

structural analyses of brain GM are based on T1-weighted MR images. The two most

common quantitative analysis methods, voxel based morphometry (VBM) and cortical

thickness analysis (CTA) are discussed.

1.4.2.1 Voxel based morphometry

VBM allows for the whole brain voxel-wise comparison of GM volume between groups. It is

a fully automated and operator-independent analysis method. The data processing is

relatively straightforward and involves spatial normalization of individual T1-weighted

images into the common stereotactic space (e.g., the Montreal Neurological Institute (MNI)

152 standard space), followed by segmentation of GM, and smoothing for noise reduction.

Voxel-wise parametric statistical tests are performed on the smoothed images from the

groups (Ashburner & Friston, 2000). It can be carried out using the Spatial Parametric

Mapping (SPM) or FMRIB Software Library (FSL) software. Because of these simple,

automated pipelines to investigate the entire GM in the brain, it has been widely used to

study diseased brains including PD (Pan, Song, & Shang, 2012). However, there are some

Page 44: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

20

limitations of VBM such as imperfect registration and tissue segmentation, which is

particularly a concern in abnormal brains. In addition, the statistical assumptions result in the

potential statistical errors (Duncan, Firbank, O'Brien, & Burn, 2013).

1.4.2.2 Cortical thickness analysis

Instead of measuring cortical GM in volume, an automated technique to accurately measure

cortical thickness (CTh) was developed using FreeSurfer software (Fischl & Dale, 2000),

i.e., CTA. CTA allows for the measure of CTh at any point in the submillimeter accuracy.

The detection of subtle cortical changes is important to study neuroplasticity due to

development and aging, neuropathology and treatment effects. CTA allows for the

quantification of GM in human cerebral cortex that is highly folded and has large regional

variations of the thickness by modeling the gray/white and pial surfaces. The identification of

these surfaces is needed for an accurate measure of CTh because the cortex contains many

folds that cannot be aligned with any of the cardinal axes along which slice data are viewed.

Figure 1-6 illustrates measuring CTh of one point from the coronal and axial slices and an

overestimation of the measurement using the coronal slice, resulting from the fact that the

surface is locally parallel to the coronal slice (Figure 1-7).

To measure CTh, the border between the WM and the GM (WM surface) is identified to

provide an estimate of the inner boundary of the cortex using a triangular tessellation over

this surface. This inner border is constructed first because resolution limitations innate to

MRI make it difficult to directly compute the outer border between the GM and the

meningeal tissue/CSF (pial surface). Once the inner cortical surface is identified, it is refined

for submillimeter accuracy and deformed outward to the pial surface. The corners of each

triangle on the surface become vertices. Thickness can then be computed as the

perpendicular distance measured between the inner and outer cortical boundaries at every

vertex in each hemisphere (Figure 1-8).

In addition to the measurement of CTh, FreeSurfer provides an automated technique to

measure subcortical volume, allowing for the investigation of whole brain GM changes.

Page 45: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

21

Figure 1-7. Estimations of CTh using slice data. Coronal (left) and horizontal (right) views of

a T1-weighted MR image of the left hemisphere with gray/white (yellow) and pial (red)

surfaces overlaid. The green crosses indicate a point at which would result in a dramatic

overestimation of the thickness of the cortex using the coronal view only. Reproduced with

permission from (Fischl & Dale, 2000), Copyright (2000) National Academy of Sciences.

Page 46: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

22

Figure 1-8. Measurement of CTh. The intersection of the tessellated WM surface in blue and

pial surface in red on MRI volume (left). CTh is the perpendicular distance measured

between the inner and outer cortical boundaries at every vertex (right). Reproduced with

permission from (Malpass, 2011).

1.4.3 White matter imaging

WM imaging for quantitative analyses is based on diffusion-weighted imaging (DWI).

Diffusion refers to the random movement of molecules due to their thermal energy (Le

Bihan, 2003; Mukherjee, Chung, Berman, Hess, & Henry, 2008; R. Watts, Liston, Niogi, &

Ulug, 2003), known as Brownian motion (Beaulieu, 2002; Le Bihan, Poupon, Amadon, &

Lethimonnier, 2006). In a free medium, during a given time interval (diffusion time),

molecules travel randomly in space over a distance depending on the size of the molecules,

the temperature and the viscosity of the medium, which is statistically well described by a

diffusion coefficient. Water diffusion in biological tissues, however, significantly differs

Page 47: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

23

from this Brownian motion because of the presence of obstacles such as cell membranes,

fibers and macromolecules (Beaulieu, 2002; Le Bihan et al., 2006; Mukherjee et al., 2008;

Neil, 2008). This reflects apparent diffusion coefficient (ADC), and allows us to infer

microstructure and geometric organization of neural tissues and importantly changes in these

features with pathological states (Le Bihan, 2003).

1.4.3.1 Diffusion-weighted imaging/Diffusion tensor imaging

DWI also relies on a proton signal from water. However, its contrast is based on water

displacements in a given time. In DWI, a pair of strong magnetic field gradient pulses

(diffusion-sensitizing gradients) is applied. The first pulse labels the spatial location of 1H

nuclei in water molecules, and the second pulse detects changes in the location due to

diffusion that occurred in the time in between the two pulses. The amplitude, duration, and

the interval between the onsets of the diffusion gradients, described by a parameter known as

the ‘b’ factor influences the signal intensities obtained in images (Mukherjee et al., 2008).

Multiple images are collected so that the signal can be sensitized to diffusion in many

different directions, building up multiple measurements for each voxel in the brain

(Johansen-Berg, 2009).

Water mobility in brain WM is anisotropic, reflecting the non-equal diffusivities depending

on direction (Basser, 1995; Pierpaoli & Basser, 1996; Watts et al., 2003). In WM, the

presence of structures in intracellular and extracellular space (Sullivan & Pfefferbaum, 2006)

including axonal membranes, myelin sheath (Beaulieu, 2002) and glial cells creates an

anisotropic environment (Assaf & Pasternak, 2008). Water diffuses mostly along the

direction of the axons rather than perpendicular to the axons (Figure 1-9). Under such

microstructure, diffusion tensor model is required to fully characterize diffusion including

the magnitude of diffusion, the degree of anisotropy, and the direction of diffusion

anisotropy (Alexander, Lee, Lazar, & Field, 2007). Diffusion tensor MR imaging or DTI is a

mathematical model of the three dimensional (3D) pattern of anisotropic diffusion of WM

tracts (Mukherjee et al., 2008) and was first introduced by Basser, Mattiello, & LeBihan

(1994). DTI estimates an effective diffusion tensor (D) with 3D measurements characterized

by a 3 x 3 matrix of nine components from DW images (Basser, 1995).

Page 48: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

24

Dxx Dyx Dzx

D = Dxy Dyy Dzy

Dxz Dyz Dzz

Each component denoted by Dij reflects the direction, i, j = {x, y, z}. Because of symmetry,

paired off-diagonal diffusion components represent the same (i.e., Dxy = Dyx, Dxz = Dzx,

and Dyz = Dzy). Thus, the six diffusivities need to be determined (Gulani & Sundgren,

2006) with diffusion gradients along at least six different non-collinear directions (Bammer,

2003; Basser et al., 1994).

In practice, the image acquisition with a large number of non-collinear directions (at least 30

directions, but 60-80 directions are recommended) is required for accuracy in the

reconstruction of fiber orientations and for reduction in the statistical rotational variance (i.e.,

the extent to which the variance in an estimate of a given parameter depends on the

orientation of the structure) as fibers are not all aligned along the same axis (Jones, Knosche,

& Turner, 2013).

From the measured tensor, principal diffusivities or eigenvalues, annotated 1, 2, and 3 in

order from largest to smallest magnitude of diffusivities (Neil, 2008; Nucifora, Verma, Lee,

& Melhem, 2007) (Figure 1-10) as well as eigenvectors of the system for the orientation

(Pierpaoli & Basser, 1996) of the eigenvalues can be calculated by a mathematical

transformation or diagonalization relating to the measured tensor to the principle

diffusivities. The eigenvector associated with the largest eigenvalue is oriented parallel to the

fiber direction while the two other eigenvectors are oriented perpendicular to it (Basser et al.,

1994; Neil, 2008).

1 0 0

Dprin = 0 2 0

0 0 3

Page 49: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

25

Once the diffusion tensor has been calculated, a range of quantities can be derived (Watts et

al., 2003). The mean diffusivity (MD) and trace of D are directionally averaged diffusivity of

water within a voxel, described as the mean of the three eigenvalues and the sum of the three

eigenvalues, respectively.

MD = (1 + 2 + 3 )/3 = trace (D)/3.

Fractional anisotropy (FA) characterizes the amount of diffusion anisotropy, and express the

anisotropic degree to which the diffusion tensor deviates from the directionally averaged

diffusion (Mukherjee et al., 2008).

When the primary eigenvalue is much larger than the other two eigenvalues, FA will be high,

indicating preferred direction of diffusion along the axon. FA rages from zero to one where

zero represents complete isotropic diffusion, and one represents complete anisotropic

diffusion (Le Bihan et al., 2001) (Figure 1-11).

The degree of orientational coherence of the axons is an important determinant of diffusion

anisotropy (Pierpaoli & Basser, 1996). Axotomy and loss of axons due to degeneration can

greatly contribute to decreased anisotropic diffusion. Myelin partially contributes to diffusion

anisotropy demonstrated in studies on multiple sclerosis (MS) or on neuronal development

(Horsfield, Larsson, Jones, & Gass, 1998; Huppi et al., 1998; Mukherjee et al., 2001;

Werring, Clark, Barker, Thompson, & Miller, 1999).

Page 50: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

26

Figure 1-9. Anisotropic diffusion in WM. Water diffuses mostly along the direction of the

axons rather than perpendicular to the axons due to the presence of microstructures such as

axonal membranes and myelin sheath. Reproduced with permission from (Mukherjee,

Berman, Chung, Hess, & Henry, 2008), AJNR. American Journal of Neuroradiology, 29,

636).

Figure 1-10. Spherical tensor shape with three orthogonal eigenvectors and their associated

eigenvalues (λ1, λ2, λ3). Reproduced with permission from (Johansen-Berg & Rushworth,

2009).

Page 51: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

27

Figure 1-11. Spherical tensor shapes illustrating various anisotropic degrees. FA = 0.1

indicates highly isotropic (left) and FA = 0.8 indicates with highly anisotropic (right).

Reproduced with permission from (Johansen-Berg & Rushworth, 2009).

1.4.3.2 Diffusion tensor tractography

There are two tractography techniques to characterize WM connectivity: (1) streamline

tractography (Mori, Crain, Chacko, & van Zijl, 1999) and (2) probabilistic tractography

using FSL software (Behrens et al., 2003; Parker & Alexander, 2005). The former follows

principal diffusion directions along adjacent voxels in WM to delineate WM connectivity.

Therefore, the only one streamline is created from one seed location (Mukherjee et al., 2008).

It uses arbitrary criteria to terminate tractography such as a low FA threshold that reflects

non WM, and a high curvature threshold, which is anatomically implausible. This approach

is often used to visualize WM connectivity. However, there are a few disadvantages of this

approach. First, modeling a single diffusion tensor in each voxel is problematic for voxels

that contain crossing fibers, which is often the case in most WM regions. Streamliners

typically terminate when fibers approach GM or areas of crossing fibers (i.e., areas of low

FA).

To address these limitations, probabilistic tractography has been developed. This technique

accounts for uncertainty in the likelihood that a WM pathway should continue and for the

possibility of crossing fibers within voxels (Behrens et al., 2003; Parker & Alexander, 2005).

In each voxel, a probability distribution of possible fiber orientations is created. From a seed

Page 52: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

28

location (e.g., one or many voxels in a region of interest (ROI)), many streamline samples

(typically thousands) are sent to trace pathways through the voxel probability distributions.

This results in a map of many streamlines, where the density at a given voxel putatively

represents the strength of connectivity between that voxel and the seed location. This

approach can fit multiple fiber populations in a voxel, allowing for modeling crossing fibers

in a voxel (Behrens, Berg, Jbabdi, Rushworth, & Woolrich, 2007; Parker & Alexander,

2005). It also allows for tractography to GM targets (Johansen-Berg, 2009). Lastly, it can

provide quantitative information about the likelihood of connections (percentage of

streamline samples from seed location reaching target voxel), which can be used to make

inferences about connectivity strength and to perform statistical analyses between groups.

However, the resulting quantified connectivity strength should be interpreted with caution,

given the lack of knowledge about the biological significance of tracts obtained with

probabilistic tractography (Jones et al., 2013).

1.4.3.3 Tract-based Spatial Statistics

Tract-based Spatial Statistics (TBSS) is a fully automated method that allows for the whole-

brain voxel-wise comparison of DTI measures between groups using FSL software (Smith et

al., 2006). It is developed to overcome the limitations of VBM analyses such as inaccurate

alignment as well as smoothing that involves the arbitrary choice of smoothing extent and

that can be another source of partial volume effect. For this purpose, a group mean WM

‘skeleton’ is generated in TBSS. It is one voxel in thickness and contains the voxel with

highest FA being the center of WM tracts common to the subjects included in a study (Figure

1-12). Once the individual FA values are mapped onto the skeleton, group-level statistics can

be performed on the skeleton-space FA data.

Page 53: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

29

Figure 1-12. Mean FA skeleton projected on a skull-stripped DWI image.

1.4.4 Functional MRI

1.4.4.1 Blood oxygen level-dependent signal

The blood oxygen level-dependent functional MRI (BOLD fMRI) was first described by

Ogawa and Lee (1990) in rat brains where deoxygenated venous blood resulted in vessels

appearing darker due to the reduction in signal. It is based on T2* signal and capitalized on

the fact that hemoglobin (Hb) in blood vessels in brain has different magnetic properties

depending on whether it is oxygenated or deoxygenated. While oxygenated Hb is

diamagnetic (weak repulsion from a magnetic field) and has little magnetic susceptibility,

deoxygenated Hb is paramagnetic (attracted to a magnetic filed). Deoxygenated Hb results in

greater field inhomogeneities and a faster decay and thus, reduction of the T2* signal

compared to oxygenated Hb (Heeger & Ress, 2002). Brain activation is reflected by an

increase in blood flow supplying neuronal activity in the active regions, and a decrease in

deoxygenated Hb, resulting in an increase in signal and image intensity. The decrease in

deoxygenated Hb is thought to be due to an oversupply of oxygenated blood when neuronal

activity increases, which could occur for a number of reasons including inefficient oxygen

Page 54: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

30

extraction from blood to surrounding tissue and/or non-oxidative metabolic processes

(Heeger & Ress, 2002).

Changes in blood flow following neuronal activity are thought to be related with synaptic

activity of Glu and recycling of astrocytes (Raichle & Mintun, 2006). At the

electrophysiological level, BOLD activity increases in anesthetized monkeys corresponded

best with changes in local field potential (LFP) signals, representing the compound activity

of neuronal populations compared with multiunit and single unit spiking (Logothetis, Pauls,

Augath, Trinath, & Oeltermann, 2001). As LFPs reflect synchronized neural activity, not

necessarily linked with firing rate, it has been proposed that the BOLD signal reflects

synaptic input to and local processing within a given area, rather than spiking output.

However, the underlying mechanisms for BOLD signal changes are still not fully understood

despite demonstrated metabolic and neuronal correlates of the BOLD signals.

1.4.4.2 Resting state functional MRI

Resting state functional MRI (rsfMRI) measures low frequency (0.01-0.1 Hz) BOLD signal

fluctuations in the resting brain. BOLD fluctuations lower than 0.01 Hz occur due to scanner-

related noise and greater than 0.1 Hz are contaminated with respiratory and cardiovascular

signals. Participants undergo a typically 5-10 min scan where they rest, and are usually

instructed to think about nothing in particular and to let their minds wander. rsfMRI is

analyzed by using the entire time course of a scan to investigate FC with an assumption that

FC does not change over the course of the scan. FC is a measure of temporal correlations of

physiological signal in anatomically distinct brain regions or voxels (Friston, Frith, Liddle, &

Frackowiak, 1993). Regions demonstrating stronger correlations are thought to have greater

FC. Functionally connected regions form patterns of synchronous activity, which make up a

number of uniquely identifiable intrinsic connectivity networks (see section 1.4.4.4). While

rsfMRI can provide a whole brain representation of FC and avoids the potential cofounder of

differential task performance in clinical populations (Greicius, 2008), it does not have

controlled behavior, making it difficult to associate the data with cognitive functions.

1.4.4.3 Seed-based functional connectivity analysis

Page 55: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

31

Seed-based analysis was the first and most straightforward method used to measure

correlations between resting state time-courses of different brain regions (Biswal, Yetkin,

Haughton, & Hyde, 1995). A time series extracted from a seed, which is a predefined ROI, is

correlated with that of every other voxel, resulting in correlation maps representing FC of

that particular seed with each individual voxel. This analysis method can also be applied to

many regions or all regions of the brain where the FC between all pairs of the regions is

calculated, generating an FC matrix. The resulting map or matrix provides information about

which regions the seed is functionally connected with and to what extent. Seed-based FC

analysis is most suitable when a researcher has a hypothesis-driven question about a specific

region. One advantage of this method is that the results are relatively easy to interpret, as

measured FC corresponds directly to the strength of correlated time-courses between specific

regions (Buckner & Vincent, 2007).

1.4.4.4 Independent Components Analysis

Independent Components Analysis (ICA) is a data-driven approach and separates

components of spatiotemporally coherent patterns of activity. Unlike the seed-based FC

analysis, ICA does not require any predefined ROIs as it uses multivariate data in all voxels

to separate the brain into groups of spatial regions with signals that have common properties.

This allows for ICA to detect structured noise components within the data, which can be

removed. This is one of the advantages of ICA over the seed-based FC analysis. ICA can

also generate multiple spatially independent networks of coherent activity, or resting-state

networks (RSNs). RSNs closely correspond to the organization of functional network derived

from co-activation analysis (Spreng, Sepulcre, Turner, Stevens, & Schacter, 2013). Examples

of these networks include default mode network (DMN), sensorimotor network (SMN),

visual network, and fronto-parietal network (FPN), and are largely consistent across healthy

people, and persist even during sleep and under anesthesia.

1.4.4.5 Graph theoretical analysis

Graph theory is a relatively old branch of mathematics that started back in the 1700 (Stam,

2014). The idea of graph theory is to represent a complex set of relationships and entity with

Page 56: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

32

a set of nodes and their connections. In the application of graph theory to

neuroscience/neuroimaging, a graph (i.e., brain network) is defined by a set of nodes (i.e.,

brain regions) and the edges (i.e., anatomical, functional or effective connections) between

them. Anatomical connections typically correspond to WM tracts connecting pairs of brain

regions. Functional connections correspond to the magnitudes of temporal correlations in

brain activity between pairs of regions. Effective connections represent direct or indirect

causal influences of one region on another (Friston, Harrison, & Penny, 2003). Graph

theoretical analysis can model and quantify topological properties of structural and

functional brain networks at the level of the entire graph, modules (subnetworks), or

individual nodes, which makes this approach one of the most powerful and flexible analytical

methods to represent a complex brain system (Power et al., 2011). Graph theoretical analysis

can be applied to MRI, Electroencephalogram (EEG) or Magnetoencephalography (MEG)

data (Rubinov & Sporns, 2010).

1.4.4.5.1 Processing and analysis steps

Here, the processing and analysis steps for undirected graphs used in study 2 are discussed.

First, nodes are defined (e.g., anatomically defined regions of MRI, functionally defined

regions of fMRI activation data). Second, an association matrix is generated by compiling

allpairwise associations between nodes (e.g., Pearson correlation, partial correlation). Third,

the association matrix is thresholded and binarized to produce a binary adjacency matrix or

binary undirected graph where links are either connected or unconnected over the certain

threshold values. The threshold values are often arbitrarily determined and thus network

properties are often explored over a range of plausible thresholds. Instead of binary graphs,

weighted graphs can be generated, which does not require the thresholding and contain more

information than the simpler binary graphs. Lastly, the network parameters of interest in the

graph are computed. Statistical testing of network parameters may be best conducted by non-

parametric permutation methods, as there is the lack of statistical theory concerning the

distribution of network measures (Rubinov & Sporns, 2010).

In our study, we used a binary adjacency matrix or undirected graph as it is simpler to

characterize and has a more easily defined null model for statistical comparison than a

Page 57: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

33

weighted network. In addition, weak and non-significant links of a weighted network tend to

obscure the topology of strong and significant connections and may also be spurious

connections (Rubinov & Sporns, 2010).

1.4.4.5.2 Graph measures

The most fundamental measure is the degree of a node, which is the number of connections

that a node has with the rest of the network. Nodes with high degree are often called hubs.

Hubs are also determined by their centrality. The centrality of a node or betweeness

centrality (BC) is measured by the number of shortest paths (i.e., direct connections) passing

through the node to connect with all other nodes in the network. A node with a high level of

centrality is often a bridging node connecting disparate parts of the network (Rubinov &

Sporns, 2010), and plays a key role in overall communication efficiency or integration of a

network (van den Heuvel & Hulshoff Pol, 2010). Creating a ‘lesioned’ network where a node

is deleted can assess the importance of an individual node to network efficiency.

If the nearest neighboring nodes of one node are also directly connected, they form a cluster

(Bullmore & Sporns, 2009). The clustering coefficient or local efficiency can be quantified

by the ratio of the number of connections that exit between the nearest neighbours of a node

and the maximum number of possible connections between the neighbouring nodes of a node

(Watts & Strogatz, 1998). High clustering coefficient indicates high local efficiency of

information transfer and robustness of the cluster/local function.

When a subset of nodes is densely connected to one another, they form a module. The nodes

in one module are sparsely connected to nodes in other modules of the network. It has been

demonstrated modules in graph correspond to RSNs of ICA (Crossley et al., 2013). The

degree to which the network is subdivided into distinct groups is quantified by modularity

(Newman, 2006). Hubs can also be defined in terms of their role in modules (Guimera &

Amaral, 2005). Nodes primarily connected to other nodes in the same module are referred to

as provincial hubs. They can be measured using the within-module degree z-score. On the

other hand, nodes linking nodes in other modules measured by participation coefficient are

referred to as connector hubs. They can be calculated by the proportion of edges linking it to

nodes in other modules (Power & Petersen, 2013). Thus, while provincial hubs play an

Page 58: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

34

important role in functional specialization in the module (local integration), connector hubs

are important for interactions or functional integration between modules (Meunier,

Lambiotte, & Bullmore, 2010). Both structural and functional neuroimaging demonstrated

that the connector hubs are typically located at the junctions between anatomically

segregated cortices and in regions of multimodal association cortex while the provincial hubs

are located within functionally specialized areas of cortex such as primary or unimodal

association areas (Meunier et al., 2010).

Brain function of a network can be assessed using several measures. A degree distribution

represents the degrees of all nodes in a network (Amaral, Scala, Barthelemy, & Stanley,

2000), which is a marker of network resilience. Another measure of network resilience is

assortativity, which is the correlation between the degrees of connected nodes. Positive

assortativity indicates that high-degree nodes tend to connect to each other. The mean

clustering coefficient for a network reflects on average the prevalence of clustered

connectivity around individual nodes (Rubinov & Sporns, 2010). Path length is the minimum

number of edges a node must traverse to reach another node. Characteristic path length of

the network is the mean shortest path length between all pairs of nodes in the network and is

related to global efficiency. Low/short mean path length or high global efficiency indicates a

highly integrated system. Connection density or cost is the actual number of edges in the

graph as a proportion of the total number of possible edges. Lastly, small-worldness is

characterized by high levels of both local clustering and short paths and can be quantified by

the ratio of the clustering coefficient to the path length. It presents an intermediate network

organization between the organization of random networks characterized by a short overall

path length and low level of local clustering, and that of regular networks characterized by a

high-level of clustering and a long path length (Watts & Strogatz, 1998). Complex networks

such as brain generally presents small-world properties with non-Gaussian degree

distribution with a long tail towards high degree to achieve an optimal balance between

regional segregation (specialization) and global integration (Bullmore & Sporns, 2012;

Tononi, Sporns, & Edelman, 1994). Major graph measures are illustrated in Figure 1-13.

Page 59: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

35

Figure 1-13. Graph measures. a. Representation of a graph with the collection of nodes and

the collection of edges (connections), describing the interactions between the nodes; b.

Clustering-coefficient of node i is given by the ratio of the number of connections between

the direct neighbors of node i in darker green (i.e., 2) and the maximum number of possible

connections between the neighbors of node i (i.e., 3), providing information about the level

of local connectedness in the graph; c. Path length of node i is given by the number of

connections that have to be crossed to travel from node i to node j in the graph, providing

information about the level of global communication efficiency of a network; d. BC of a

node i indicates how many of the shortest paths between the nodes of the network pass

g

Page 60: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

36

through node i. A high BC indicates a potential hub role of this node in the overall network;

e. Degree of node i is defined as its total number of connections; f. Modularity of a graph

describes the possible formation of communities in the network; g. Network topologies:

regular network (left) has a rather local character, characterized by a high clustering-

coefficient (C) and a high path length (L); random network (right) has a more global

character, with a low C and a much shorter path length L than the regular network; and

small-world network (middle) with both a high C and a low L, combining a high level of

local and global efficiency. Reproduced with permission from (van den Heuvel & Hulshoff

Pol, 2010).

1.5 Positron Emission Tomography

1.5.1 Principles of Positron Emission Tomography imaging

PET imaging is a nuclear medicine imaging technique to quantify the chemical/biological

processes of molecules in vivo by injecting radiolabelled molecules (i.e., radioligand)

(Venneti, Lopresti, & Wiley, 2013). The radioligands typically resemble endogenous

biological molecules with specific biological targets to which they bind. This allows for

mapping the distribution of the protein in the brain. The radioligand is synthesized by

labeling a precursor molecule with short-lived radionuclides such as carbon-11 (t1/2 = 20.4

min) and fluorine-18 (t1/2 = 109.8 min) (Figure 1-14). After the radioligand is injected

intravenously, it enters the bloodstream, crosses the blood-brain barrier (BBB) and binds to

target receptors or proteins in the brain.

When the radionuclide decays, positron is emitted. The positron quickly loses its kinetic

energy and reaches thermal equilibrium with its surroundings and then combines with a free

electron before they mutually annihilate. The annihilation produces two 511 kilo-electron

volts (keV) gamma ray or photons that travel along a line in opposite directions (Figure 1-

13). These 511 keV gamma emissions are detected by rings of radiation detectors of a PET

scanner (Figure 1-14). Detection of two photons within a coincidence window (~ 6 nsec)

results in a coincidence event that is plotted on a sinogram Upon the completion of scan, the

Page 61: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

37

sonogram is corrected for photon attenuation and scatter based on a transmission scan

acquired prior to the data acquisition. The PET image is reconstructed using a mathematical

algorithm based on the sinogram plots, radioligand dead time and decay for data

quantification.

Figure 1-14. Principles of PET. The synthesis of PET radioligand by labeling a precursor

Page 62: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

38

molecule with short-lived radionuclides (top). PET radionuclides decay by positron emission

to a stable nuclide. Once the positron reaches thermal equilibrium with its surroundings

(thermalization), it interacts with a free electron, briefly forming a bound state called

positronium before both particles mutually annihilate. The annihilation of the electron and

positron results in the emission of two 511 keV gamma ray photons, traveling in opposite

directions along a line (middle). PET scanner detects the coincident annihilation events with

the surrounding rings of radiation detectors, which is used for the data reconstruction

(bottom). Reproduced with permission from (Venneti et al., 2013).

1.5.2 PET radioligands

PET radioligand should binds with ≤5% of target proteins (Innis et al., 2007) that reflects the

entire population of target proteins without significantly perturbing the total number of

available target proteins. To image targets in the brain, a PET radioligand must meet a

number of criteria such as high affinity, selectivity for the target protein, low non-specific

binding, and suitable brain pharmacokinetics related with radiolabel half-life (i.e., observable

brain uptake and washout kinetics). They also must penetrate the BBB and reach their target

in vivo. Radioligands with high lipophilicity are considered undesirable because of slow

brain entry and high non-specific binding to brain fats and proteins, resulting in attenuation

of specific signal. Furthermore, they must lack of radiometabolites in the brain because PET

cannot discriminate between the chemical sources of detected radioactivity. The metabolism

should ideally occur outside of the brain and produce less lipophilic radiometabolites with

poor brain entry and little or no interaction with the target protein.

1.5.3 Quantification of PET radioligand binding in vivo

1.5.3.1. Kinetic modeling of time-activity curve

Kinetic modeling is a mathematical model to quantify specific radioligand binding to a target

protein from the time-activity curve of the radioligand. Kinetic modeling often uses

compartment models. The compartments can include those of the blood plasma (CP),

specifically bound to a target protein (CS), nonspecifically bound (CNS) or free in the tissue

(CF). A compartment model is based on an assumption that radioligands may move freely

Page 63: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

39

between compartments until they reach equilibrium. The movement of radioligand molecules

between the compartments is described in terms of rate constants. The rate constants are

estimated to fit the measured time-activity data to a compartment model using the

metabolite-corrected radioligand concentration in the plasma as an input function. The input

function is derived from estimations of a concentration of radioligand in the whole blood and

plasma, as well as fractions of radiolabeled metabolites in the plasma from a sequential series

of arterial blood samples obtained during the PET scan. Iterative non-linear fitting is

typically used to estimate the rate constants that best fit the measured time activity data. The

identifiability of the estimate of the rate constants can also be assessed using the percent

coefficient of variation (%COV), which is the standard error of the estimated parameters. An

estimate with a low %COV indicates a better fit of the chosen model and thus more accurate

parameter estimates. The goodness of fit and the appropriateness of a particular compartment

model for a given dataset can be quantitatively determined using the Akaike Information

Criterion (AIC) (Akaike, 1974) and Bayesian Information Criterion (BIC) (Schwarz, 1978).

Lower AIC and higher BIC values indicates better fit, respectively.

1.5.3.2. Two-tissue compartment model and its outcome measures

The two-tissue compartment model (2TCM) consists of three compartments: CP, CS, and

nondiplaceable compartment (CND) including both CF and CNS (CND = CF + CNS). (This

model is typically called three-compartment model in clinical pharmacology, but two-tissue

compartment in radioligand imaging) (Innis, et al., 2007). It is based on an assumption that

the CNS and CF have fast kinetics and reach equilibrium very rapidly, and thus they are

considered one compartment. (Figure 1-15). Rate constants include K1 from CP to CND, k2, in

a reverse direction, as well as k3 from CND to CS and k4, in a reverse direction. K1 uses upper

case whereas others use lower case. This distinction is primary because K1 uses different

units (mL* cm-3 mL-1 *min-1 representing a volume of blood per volume of tissue per minute)

than the other rate constants (min-1).

Using the 2TCM, three binding outcome measures can be estimated based on the kinetic

parameters including the total distribution volume (VT), distribution volume of the specific

compartment (VS), and the binding potential non-displaceable (BPND). VT refers to the ratio

Page 64: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

40

at equilibrium of total concentration of radioligand in the brain tissue (CT) to the

concentration of radioligand in CP (Innis et al., 2007). Therefore, VT = CT / CP where CT =

CND + CS. In 2TCM, VT value can be estimated by VT = K1/k2 (1+ k3/k4). VS refers to the

ratio at equilibrium of the specifically bound radioligand to that of total parent radioligand in

plasma and estimated by VS = K1/k2 k3/k4. BPND refers to the ratio at equilibrium of

specifically bound radioligand to that of nondisplaceable radioligand in tissue and estimated

by BPND= k3/k4.

Figure 1-15. 2TCM. The two compartments are located within the tissue including

nondisplaceble (free and nonspecific) and specific compartments. Input to the tissue derives

from the plasma compartment. The rate constants representing the exchange of radioligand

between compartments.

1.5.3.3. One-tissue compartment model

A one-tissue compartment model (1TCM) includes two compartments: one is CP and the

other has CF, CNS, and CS, which cannot be kinetically distinguished from one another and

therefore, it describes the bidirectional flux of radioligand between the plasma and tissue

compartments (Figure 1-13).

CP

CND

= CF + C

NS

CS

K1

k2

k3

k4

CP

CF + C

NS +

C

S

K1

k2

Page 65: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

41

Figure 1-16. 1TCM. The tissue has just one compartment, that is the free and nonspecifically

bound ligand cannot be kinetically distinguished from the specifically bound.

1.5.3.4 A simplified reference tissue model

A simplified reference tissue model (SRTM) does not require the arterial input function but

instead relies on the presence of a reference region (Lammertsma & Hume, 1996) (Figure 1-

17). It is based on four assumptions. First, the reference region is devoid of specific binding.

Second, target tissue and reference tissue compartments can be described as one

compartment based on an assumption that both compartments reach equilibrium rapidly.

Third, the level of nondisplaceble binding is the same in both reference and target tissues and

fourth, the blood volume contribution to both reference and target tissue is negligible

(Salinas, Searle, & Gunn, 2015). This model allows for an estimation of BPND, which refers

to the ratio at equilibrium of a specifically bound radioligand to that of a nondisplaceable

radioligand in tissue.

CREFERENCE

CP

K1

k2

K1

k2

CTARGET

CF + C

NS +

C

S

Page 66: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

42

Figure 1-17. SRTM. The free, non-specific and specifically bound radioligand reach

equilibrium rapidly and can be condensed into one compartment. CREFERENCE represents a

brain region that is devoid of target receptors.

1.6 Neuroimaging studies in Parkinson’s disease

PD is a complex disease with different clinical phenotypes, rates of progression, and

pathologies. Current trends toward use of data driven analytic methods can facilitate to

expand our knowledge about PD beyond the classic BG motor model (Weingarten,

Sundman, Hickey, & Chen, 2015) and DA dysfunction. Structural and functional changes

beyond the BG have been investigated in vivo using different neuroimaging techniques. My

thesis projects employed MRI and PET to identify PD-related brain changes accounting for

the clinical manifestations.

1.6.1 Structural changes in Parkinson’s disease

MRI is a non-invasive and safe approach with high spatial and temporal resolution. It is

widely available, leading to its widespread applications for structural investigations of PD

(Weingarten et al., 2015). Whole brain GM and WM changes can be quantified using VBM

and CTA combined with subcortical ROI analysis from an anatomical T1-weighted image

and TBSS from a DWI image, respectively.

Whole brain GM changes have been extensively investigated using VBM in PD patients.

One meta-analysis study including 17 studies involving 498 idiopathic PD patients and 375

healthy control (HC) subjects reported significant regional GM volume loss in the left

inferior frontal gyrus (Brodmann Area (BA)47) extending to the left superior temporal gyrus

(BA38) and the left insula (Ins) (BA13) (Pan et al., 2012). As suggested from these findings,

many of VBM studies found cortical changes but failed to find subcortical changes (Biundo

et al., 2011; Burton, McKeith, Burn, Williams, & O'Brien, 2004; Hong, Lee, Sohn, & Lee,

Page 67: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

43

2012; Kostic et al., 2010; Messina et al., 2011; Song et al., 2011; Tessitore et al., 2012;

Weintraub et al., 2011).

It is also noted that six of the 17 studies in the meta-analysis did not find any significant

group differences. While volume measurements like VBM often result in overestimations of

GM changes, they may underestimate subtle cortical changes occurring in submillimeter

(Fischl & Dale, 2000) (see section 1.4.2.2). Four PD studies using both CTA and VBM

suggested that CTA be more sensitive to detect changes in cortical regions in PD (Gerrits,

van Loenhoud, van den Berg, Berendse, Foncke, Klein, et al., 2016; Ibarretxe-Bilbao et al.,

2012; Pereira et al., 2012; Tessitore, Santangelo, De Micco, Vitale, Giordano, Raimo, et al.,

2016). In one study, faster rate of cortical thinning but not that of volume reduction was

detected in PD patients compared to HC subjects in a longitudinal study (Ibarretxe-Bilbao et

al., 2012), and in anther study CTA detected cortical thinning in a number of cortical regions

in PD patients compared with HC subjects whereas VBM found volume decrease only in one

cortical region (Pereira et al., 2012). In addition, CTA but not VBM differentiated PD

patients from HC subjects (Gerrits, et al., 2016) as well as PD patients with Impulse Control

Disorders (ICD) from those without ICD and HC subjects (Tessitore, et al., 2016).

CTA revealed that PD patients showed cortical thinning in various regions compared to HC

subjects. Cortical thinning occurred in the frontal regions including the motor (Pereira et al.,

2012), premotor (Zarei et al., 2013) and supplementary motor areas (SMA) (Jubault et al.,

2011; Zarei et al., 2013) as well as the orbitofrontal (Biundo et al., 2013; Hanganu et al.,

2013; Lyoo, Ryu, & Lee, 2010), superior (Zarei et al., 2013), middle (Lyoo et al., 2010),

dorsolateral (Zarei et al., 2013) and inferior frontal regions (Pereira et al., 2012; Tinaz,

Courtney, & Stern, 2011); in the temporal regions including the superior (Lyoo et al., 2010;

Pereira et al., 2012), middle (Pellicano et al., 2012; Pereira et al., 2012), and inferior

temporal or angular regions (Pereira et al., 2012; Zarei et al., 2013), fusiform (Lyoo et al.,

2010; Pellicano et al., 2012; Tinaz et al., 2011; Zarei et al., 2013) and lingual gyri (Hanganu

et al., 2013; Lyoo et al., 2010; Tinaz et al., 2011), and temporal pole (Lyoo et al., 2010;

Pereira et al., 2012); in the parietal regions including the precuneus (Zarei et al., 2013),

postcentral (Pereira et al., 2012) and the inferior parietal (Lyoo et al., 2010; Pereira et al.,

2012) or supramarginal (Lyoo et al., 2010; Zarei et al., 2013) regions; and in the occipital

Page 68: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

44

regions including the lateral occipital (Li et al., 2013; Pereira et al., 2012; Tinaz et al., 2011)

and cuneus regions (Tinaz et al., 2011).

Taken together, the most reported region of cortical thinning is the frontal region followed by

the temporal and parietal regions. A longitudinal study also supported these findings by

demonstrating that PD patients showed a faster rate of cortical thinning than HC subjects in

most cortical regions except in the occipital region and the frontal cortex was most affected

(Ibarretxe-Bilbao et al., 2012). The frontal regions included the superior, caudal middle

frontal and inferior gyri, as well as precentral and paracentral regions. The temporal regions

included the superior temporal and middle temporal gyri. The parietal region included the

supramarginal and inferior parietal regions.

Cortical thickening in small cortical regions was also reported in subtypes of PD patients.

For example, PD-MCI patients showed cortical thickening in a small region of the left

middle temporal gyrus compared with PD patients with normal cognition (Hanganu et al.,

2013). In addition, PD patients with Levodopa-induced dyskinesia also showed cortical

thickening in the inferior frontal region compared to HC subjects (Cerasa et al., 2013).

WM changes also occurred in PD patients (Hattori et al., 2012; Kim et al., 2013; Melzer et

al., 2013; Theilmann et al., 2013; Zhan et al., 2012; Zheng et al., 2014). They become severe

as the disease progresses (Agosta et al., 2013) and cognition declines (Hattori et al., 2012).

The early stage of patients and patients with normal cognition may spare WM changes

(Agosta et al., 2013; Agosta et al., 2014). However, one study identified changes in limited

frontal WM including the anterior corpus callosum (CC), superior corona radiata (SCR), and

cingulum in a larger sample of PD patients with normal cognition (Melzer et al., 2013). This

study employed the Movement Disorders Society (MDS) diagnostic criteria for PD-MCI

(Litvan et al., 2012) to group PD patients into those with normal cognition and those with

MCI. WM changes only in the frontal region were also found in PD-MCI such as the anterior

corona radiata (ACR) and SCR (Agosta et al., 2014). Other studies tend to show diffuse WM

changes in non-demented PD patients including the studies where PD group showed no

deficits on neuropsychological tests (Gallagher et al., 2013; Theilmann et al., 2013).

Page 69: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

45

CTh, subcortical volume, and WM measures were also investigated for correlations with

clinical/cognitive measures. CTh was associated with the duration (Hanganu et al., 2013;

Jubault et al., 2011; Lyoo, Ryu, & Lee, 2011), and stage of disease (Pereira et al., 2012;

Zarei et al., 2013), motor severity (Lyoo et al., 2011; Zarei et al., 2013), Mini-mental status

examination (MMSE) scores (Zarei et al., 2013), neuropsychological test scores (Biundo et

al., 2013; Pellicano et al., 2012), and facial emotion recognition (Baggio et al., 2012). GM

atrophy in subcortical regions was associated with poor memory, DA non-responsive motor

signs and executive function (Camicioli et al., 2009), global cognitive measure (aggregated

cognitive z score) (Melzer et al., 2012), as well as olfactory dysfunction (Wattendorf et al.,

2009). WM changes were associated with MMSE scores (Agosta et al., 2013; Gallagher et

al., 2013; Hattori et al., 2012; Melzer et al., 2013; Theilmann et al., 2013), cognitive

performance in different cognitive domains (Gallagher et al., 2013; Melzer et al., 2013;

Theilmann et al., 2013; Zhan et al., 2012).

Despite the evidence of structural abnormalities and variability of GM and WM measures

associated with clinical and cognitive manifestations in PD patients, whether the reported

structural abnormalities account for specific clinical sequelae in PD patients is still unclear.

This is largely because studies that used the whole brain approach typically investigated

group differences and correlations independently (Camicioli et al., 2009; Hanganu et al.,

2013; Jubault et al., 2011; Tinaz et al., 2011). For example, one study found group

differences in WM measured by increased MD in diffuse brain regions (Agosta et al., 2013).

However, correlations were found only between FA and MMSE scores. These findings make

the interpretations difficult and questions structural makers for PD pathology. Even though

brain regions demonstrating significant group differences have been shown to correlate with

cognition, PD patients did not show impairment on cognitive tasks (Theilmann et al., 2013).

The authors argued that WM changes might precede cognitive impairment in these patients,

which warrants further confirmation. Furthermore, only correlations were investigated in PD

patients (Lyoo et al., 2011; Zheng et al., 2014). Thus, whether or not these correlations were

unique to PD patients and whether or not these regions showed any structural differences

compared to HC subjects are unknown. Studies are needed to elucidate whether the structural

changes would account for specific clinical manifestations of PD by interrogating the regions

showing significant structural changes for correlations.

Page 70: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

46

For this purpose, we aimed to determine whether structural changes occurred in PD patients

and whether these structural abnormalities were associated with the motor and NMS by

investigating the whole brain GM and WM using CTA and TBSS (see section 2.1).

1.6.2 Brain network changes in Parkinson’s disease using resting

statefMRI and graph theoretical approach

To date, there are approximately 20 studies published using graph theoretical analyses to

fMRI, EEG, and MEG data in PD patients. In general, these studies included PD patients in

the earlier stage of disease.

On the global level, PD patients showed more segregated (Fogelson et al., 2013; Gottlich et

al., 2013; Ma et al., 2016; Sang et al., 2015; Skidmore et al., 2011) and less integrated

network (Fogelson et al., 2013; Gottlich et al., 2013; Ma et al., 2016; Sang et al., 2015;

Skidmore et al., 2011). These topological patterns were also observed in PD-MCI (Baggio et

al., 2014). On the contrary, one study showed that PD patients showed less segregated

network with preserved overall integration (Luo et al., 2015). A longitudinal study showed

that earlier pathology was limited to reduced local clustering/integration, which later became

more prominent and widespread, leading to a reduction in global integration (Olde

Dubbelink et al., 2014).

More segregated network measured by clustering coefficient (Lebedev et al., 2014; Tinaz,

Lauro, Hallett, & Horovitz, 2016) or less integrated network measured by BC (Olde

Dubbelink et al., 2014; Tinaz et al., 2016) were associated with more severe motor symptoms

as well as the overall disease severity measured by UPDRS total scores. In addition,

increased network segregation was associated with worse cognitive performance in

visuospatial/visuoperceptual and memory domains in PD-MCI patients (Baggio et al., 2014).

On the modular (subnetwork) level, PD patient did not differ from HC subjects (Ma et al.,

2016; Sang et al., 2015). However, PD-MCI patients showed increased modularity compared

with PD patients with normal cognition and HC subjects (Baggio et al., 2014), suggesting

more segregated brain system in PD-MCI. One study investigated intra- and inter-modular

connectivity and found increased local integration in medial PFC (mPFC) network, salience

Page 71: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

47

network (SAL), SMN, and FPN, as well as reduced interactions between mPFC network and

(1) SAL (2) SMN and (3) visual network in PD patients compared with HC subjects (Ma et

al., 2016). Furthermore, higher local integration within the mPFC network was associated

with better cognitive performance on MMSE (Ma et al., 2016).

On the local level, as expected, PD patients showed nodal changes (both increased and

decreased FC) within SMN (Gottlich et al., 2013; Luo et al., 2015; Nagano-Saito, Martinu, &

Monchi, 2014; Tinaz et al., 2016; Wei et al., 2014; Wu, Wang et al., 2009) and the changes

correlated (both positive and negative) with UPDRS scores (Cao, Xu, Zhao, Long, & Zhang,

2011; Wei et al., 2014; Wu et al., 2009). In addition, nodal changes occurred in the fronto-

parietal (Fogelson et al., 2013; Ma et al., 2016; Tinaz et al., 2016), visual or occipital

(Gottlich et al., 2013; Luo et al., 2015), orbitofrontal (Gottlich et al., 2013; Luo et al., 2015;

Olde Dubbelink et al., 2014; Tinaz et al., 2016), and temporal regions (Luo et al., 2015; Olde

Dubbelink et al., 2014). Increased connectivity in the fronto-parietal nodes was associated

with better executive function (Lebedev et al., 2014).

Hub reorganization was also observed in PD patients (Baggio et al., 2014; Sang et al., 2015).

Notable alternations in hub organization included lost hubness in nodes of temporal-limbic

regions as well as in the putamen in PD patients (Sang et al., 2015) and increased hubness in

nodes of the prefrontal region in PD-MCI patients (Baggio et al., 2014). It is noted that the

definition of hubness is arbitrary and these studies used different measures for hubness: The

former study used BC and tested the values against the null hypothesis using a non-

parametric one-tail test across different thresholds and the latter study defined nodes as hubs

when they ranked at 20% highest sum of BC and degree out of 90 ROIs.

These findings suggest that in the earlier disease, functional changes occur locally and a loss

of centrality of hub nodes leads to disconnection of interacting modules. As the disease

progresses, the functional changes on inter- and intra-modular levels become more prominent

and widespread, affecting global integration of the network.

Using the graph theoretical approach, an investigation of hub regions has become the focus

of attention in pathological brains because of its widespread effects on network structure and

function (Zalesky, Fornito, Cocchi, Gollo, & Breakspear, 2014). Empirical studies also

Page 72: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

48

suggest that hub regions were disproportionately affected by brain disorders (Buckner et al.,

2009; Crossley et al., 2014). This may be related to the high baseline activity and metabolic

activity of hub regions such as high rates of cerebral blood flow, aerobic glycolysis, and

oxidative glucose metabolism (Buckner et al., 2009; Liang, Zou, He, & Yang, 2013; Saxena

& Caroni, 2011; Tomasi, Wang, & Volkow, 2013; Vaishnavi et al., 2010), which make them

particularly vulnerable to a wide range of pathologic factors such as metabolic (oxidative)

stress or oxidative stress (Crossley et al., 2014). Network hubs are almost certainly

vulnerable in PD (Stam, 2014) with dysfunction of multiple neurotransmitters (Gratwicke,

Jahanshahi, & Foltynie, 2015; Pagonabarraga et al., 2015), deposition of α-synuclein (Braak,

Rub, Jansen Steur, Del Tredici, & de Vos, 2005), and neuroinflammation (Gao & Hong,

2008; Hirsch & Hunot, 2009), and the selective deterioration of hubs may account for the

distributed abnormalities across the brain in PD (McColgan et al., 2015) with distinct clinical

manifestations. Therefore, the Study 2 hypothesized selective node changes (see section 2.2)

and their association with motor and cognitive symptoms.

1.6.3 Neuroinflammation in PD using Translocator protein 18 kDa

imaging

1.6.3.1.Translocator protein 18 kDa

TSPO (18 kDa) has been investigated as a in vivo biomarker for neuroinflammation (Chen &

Guilarte, 2008; Venneti et al., 2013). TSPO was first discovered as an alternative binding site

for the benzodiazepines in 1977 (Braestrup & Squires, 1977). It was initially called

Peripheral Benzodiazepine Receptor (PBR) and renamed TSPO that reflects its molecular

role in cholesterol synthesis and the molecular weight of the protein (Papadopoulos et al.,

2006). TSPO consists of 169 amino acids and highly hydrophobic and rich in tryptophan

(Casellas, Galiegue, & Basile, 2002). There is an 80% sequence homology among various

species such as rodents, bovines and humans. The TSPO gene is located in the q13.3 regions

of the long arm of human chromosome 22 (Riond et al., 1991).

TSPO is distinct from the central benzodiazepine receptor (CBR) in pharmacological profile,

structure, subcellular localization (located on the plasma membrane of GABAergic (γ-

Page 73: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

49

aminobutyric acid) neurons), and tissue distribution (Gatliff & Campanella, 2012).

Benzodiazepines such as clonazepam binds with high affinity to CBR, but with extremely

low affinity to TSPO (Benavides et al., 1983) although diazepam binds to both TSPO and

CBR with high affinity.

Endogenous molecules that bind to TSPO include the diazepam binding inhibitor (DBI) or

endozepine (Guidotti et al., 1983), cholesterol and porphyrins. DBI is widely distributed in

the CNS (predominantly in glial cells) and in peripheral organs, especially steroidogenic

cells.

TSPO is located primarily in the outer mitochondrial membrane (Antkiewicz-Michaluk,

Mukhin, Guidotti, & Krueger, 1988; Bribes et al., 2004; Garnier et al., 1994). It shows high

levels in glandular and secretory tissues such as the pineal gland, adrenal gland, salivary

glands, olfactory epithelium and gonads, intermediate levels in renal and myocardial tissue

and relatively low levels in the liver and brain. In the brain parenchyma, TSPO is located in

the outer mitochondrial membrane of glial cells (Chen & Guilarte, 2008). However, it was

also detected in the nucleus and perinuclear area in glial cells in the CNS (Kuhlmann &

Guilarte, 2000). Additionally, TSPO is expressed in circulating blood cells with the highest

concentrations in monocytes and neurotrophils (Canat et al., 1993).

TSPO are thought to be involved in a lot of physiological functions including cell growth and

proliferation, steroidogenesis, mitochondrial respiration, and apoptosis. The fact that TSPO

knockout mice die at an early embryonic stage (Papadopoulos et al., 1997) suggests that

TSPO is involved in basic house-keeping functions and is essential for embryonic

development (Chen & Guilarte, 2008). Among many potential physiological functions of

TSPO, steroidogenesis is best characterized. Abundantly expressed in steroidogenetic tissues,

TSPO mediates cholesterol transport into mitochondria (Papadopoulos et al., 1997). TSPO

can bind cholesterol and facilitate the transport of cholesterol from the outer to the inner

mitochondrial membrane, the rate-determining step of steroidogenesis. The side chain

cleavage cytochrome P-450 enzyme, located in the inner mitochondrial membrane, can

convert cholesterol to pregnenolone (steroid precursor), and initiate steroidogenesis such as

neurosteroid production in glial cells (Rupprecht et al., 2009; Rupprecht et al., 2010).

Page 74: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

50

1.6.3.2 Prototypical TSPO radioligand: [11C]-PK11195

[11C]-PK11195 was the first and has been most commonly used PET radioligand for the

investigation of TSPO density (Chauveau, Boutin, Van Camp, Dolle, & Tavitian, 2008).

Normal brain expresses minimal [3H]-PK11195 binding (Banati et al., 2000) and TSPO

expression (Cosenza-Nashat et al., 2009). In various pathological post mortem brains,

elevated [3H]-PK11195 binding was associated primarily with activated microglia (Banati et

al., 2000; Venneti, Wang, & Wiley, 2008). Microglial activation in vitro increased the

number of TSPO without changing binding affinity of PK11195 (Banati et al., 2000).

Although increased [11C]-PK11195 binding in vivo has been reported in various neurological

diseases such as MS (Banati et al., 2000), ischemic stroke (Gerhard et al., 2000),

amyotrophic lateral sclerosis (Turner et al., 2004), Human immunodeficiency virus (HIV)

dementia (Hammoud et al., 2005), frontotemporal dementia (Cagnin, Rossor, Sampson,

Mackinnon, & Banati, 2004), PD (Gerhard et al., 2006), and AD (Cagnin et al., 2001), [11C]-

PK11195 binding was not different between HC subjects and patients with mild cognitive

impairment (Schuitemaker et al., 2004; Schuitemaker et al., 2006), suggesting that the

radioligand may not be sensitive to detect mild forms of neuroinflammation (Venneti et al.,

2008).

In fact, this radioligand has several important limitations including a high nonspecific

binding (Chen, Baidoo, Verina, & Guilarte, 2004), low total brain uptake (Maeda et al.,

2004; Petit-Taboue et al., 1991), and a poor signal-noise ratio (Banati et al., 2000; Kropholler

et al., 2006; Pappata et al., 1991; Venneti et al., 2013). These limitations of [11C]-PK11195

have led to developing new radioligands with greater sensitivity and specificity to detecting

TSPO binding since neuroinflammatory processes are common pathologies in various brain

diseases and disorders (James, Selleri, & Kassiou, 2006; Okubo, Yoshikawa, Chaki,

Okuyama, & Nakazato, 2004).

1.6.3.3 Second generation TSPO radioligands

To date, there are a number of second generation TSPO radioligands have developed

including [11C]PBR28 (Imaizumi, Kim et al., 2007), [18F]-FEPPA (discussed in section

Page 75: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

51

1.6.3.5) (Wilson et al., 2008), [11C]-DAA1106 (Maeda et al., 2004), [11C]-DAA1106 (Zhang

et al., 2003), [18F]-FEDAA1106 (Zhang, Maeda et al., 2003), [11C]-DPA 713 (Boutin,

Chauveau, Thominiaux, Gregoire et al., 2007), [11C]-CLINME (Boutin, Chauveau,

Thominiaux, Kuhnast et al., 2007), [18F]-PBR06 (Imaizumi, Briard et al., 2007), [18F]-GE180

(Wadsworth et al., 2012), [18F]-DPA 714 (James et al., 2008), [18F]-F-VUIIS1008 (Tang,

Nickels, Tantawy, Buck, & Manning, 2014) and [18F]DPA-C5yne (Medran-Navarrete et al.,

2014).

Compared with [11C]-PK11195, [11C]-DAA1106 (Venneti, Lopresti et al., 2007; Venneti,

Wagner et al., 2007; Venneti, Wang, Nguyen, & Wiley, 2008), [18F]-FEDAA1106 (Fujimura

et al., 2006), [11C]-PBR28 (Parente et al., 2016), [18F]-PBR06 (Imaizumi et al., 2007a), [18F]-

DPA 714 (James et al., 2008), [18F]-GE180 (Wadsworth et al., 2012), and [18F]-DPA-C5yne

(Medran-Navarrete et al., 2014) showed higher affinity; [11C]-PBR28 (Parente et al., 2016)

and [18F]-DPA 714 (James et al., 2008) showed higher brain uptake; as well as [11C]-DPA

713 (Boutin et al., 2007a; Doorduin et al., 2009), [11C]-CLINME (Boutin et al., 2007b),

[18F]-PBR111 (Van Camp et al., 2010), [18F]-DPA714 (Corcia et al., 2012) and [18F]-GE180

(Boutin et al., 2015; Liu et al., 2015) demonstrated a better contrast between lesioned and

non-lesioned sites of the brain.

Some of these radioligands have been evaluated in vivo in healthy human individuals.

Compared to [11C]-PK11195, [11C]-PBR28 and [11C]-DPA-713 showed a larger brain uptake

(Endres et al., 2009; Fujita et al., 2008) and signal (Endres et al., 2009). However, shorter

half-life of [11C] radiolabelled ligands limits their dissemination and thus, [18F] radiolabelled

ligands are preferred for wide clinical applications. [18F]-DPA 714 demonstrated the

favorable pharmacology and biodistribution (Arlicot et al., 2012). However, it was quantified

using the cerebellum as a reference region, which is likely to produce the underestimation of

specific binding (Arlicot et al., 2012), as there is no brain region devoiding TSPO (see

section 1.6.3.5). Despite of improvements of sensitivity and specificity for TSPO, other

second-generation TSPO radioligands also showed some limitations such as a slow

accumulation of radiometaboliltes of [18F]-PBR06 in the brain, which compromises the

accurate estimation of TSPO (Fujimura et al., 2006; Fujimura et al., 2009), and slow kinetic

Page 76: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

52

behavior of [18F]-FEDAA1106 (Fujimura et al., 2006), which likely requires a longer scan

and thus may not be suitable for clinical populations.

Interestingly, the [11C]-PBR28 study reported “non-binders” who appeared to lack the

binding site or lack of the TSPO proteins in both brain and peripheral organs (Fujita et al.,

2008). In this study, there were two such individuals out of 10 participants. The authors also

reported that since the time of the study, there were four such participants out of 29. This

study led to finding of three binding affinity patterns of second-generation TSPO

radioligands depending on the TSPO polymorphism. The null findings of the studies in

clinical populations using [18F]- FEDAA1106 (Takano et al., 2013; Varrone et al., 2013) and

[18F]-DPA714 (Golla et al., 2015) may have been due to the lack of information about the

binding affinity of study participants.

1.6.3.4 TSPO polymorphism and binding affinity of second generation

TSPO radioligands

Following the finding of “non-binders” of [11C]-PBR28 (Fujita et al., 2008), binding

characteristics of PBR28 was compared with PK11195 in autoradiographic studies using MS

human brain tissue (Owen et al., 2010) and revealed that 5 of the 22 donors (23%) did not

show a measurable binding signal of [3H]PBR28 while they showed strong binding with

[3H]PK11195. These non-binding tissue samples showed a significant reduction in the

affinity of [3H]-PBR28 compared to those showing normal signal. In addition, 40% of those

with normal signal presented both high-affinity and low-affinity sites. It is noted that there

was no difference in TSPO density (Kd or Bmax) for [3H]-PK11195 among the study

samples. These binding patterns were characterized by high-affinity, low-affinity, and

mixed-affinity (two-site) binding. The subsequent study extended the findings to further

confirm that the three binding affinity patterns existed (1) in normal brain tissue of 20

subjects, (2) using other TSPO radioligands such as PBR06, DAA1106, DPA713, and

PBR111 in the brain tissue although differences in affinity between high affinity binders

(HABs) and low affinity binders (LABs) varied, (3) in the platelets of healthy subjects and

(4) in the outcome measures of [11C]-PBR28 PET scans of healthy subjects (Owen, Gunn,

Rabiner, Bennacef, Fujita, Kreisl, Innis, Pike, Reynolds, Matthews, & Parker, 2011).

Page 77: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

53

These different binding affinity patterns can be predicted by the TSPO polymorphism (Owen

et al., 2012). Twenty polymorphisms in TSPO gene were analyzed for association with the

binding phenotype of PBR28 in platelets of 41 healthy subjects (Owen et al., 2012). HABs

and LABs were defined as subjects with a single binding site with Ki <15 or > 100 nmol/L,

respectively because there were no HABs with Ki >15 or no LABs with Ki > 100 (Owen et

al., 2011). Mixed- affinity binders (MABs) were defined as subjects with two binding sites.

Only one polymorphism (rs6971) survived the multiple testing P value threshold, which is

located in exon 4 of the TSPO gene. It results in a nonconservative amino-acid substitution at

position 147 from alanine to threonine (Ala147Thr) in the fifth transmembrane domain of the

TSPO protein. This study also showed the complete agreement between rs6971 genotype and

PBR28 binding phenotype in the platelets. This polymorphism also predicted PBR28 binding

affinity class in the leukocytes membranes as well as [11C]-PBR28 binding affinity class in

brain where HABs showed significantly higher mean VT values than MABs in the whole

brain and cortical regions including the medial temporal, cingulate, temporal, frontal, parietal

and occipital regions (Kreisl et al., 2013). Similarly, significant group differences in [18F]-

PBR111 VT values between HABs and MABs were reported in the global cortical region,

cerebellum, hippocampus, thalamus, brain stem and CN in healthy individuals (Guo et al.,

2013). Furthermore, [3H]-PBR28 binding in postmortem brain tissues from patients with

schizophrenia (SCZ) and HC subjects showed significant group difference after binding

affinity was included as a covariate in the statistical model. Taken together, accounting for

the TSPO polymorphism likely increases the chance to detect group differences.

The prevalence of HAB, MAB, and LAB depends on ethnicity. According to the HapMap

project, HAB, MAB, and LAB account for 46%, 38%, and 15% in Caucasian population; in

African American population, 78%, 20%, and 2%; in both Japanese and Han Chinese

population, 98%, 2%, and less than 0.1%, respectively. Thus, this binding affinity variation

has the greatest impact on studies of Caucasians.

The interindividual variability in binding affinity observed in second-generation TSPO

radioligands was not evident in [11C]-PK11195. This may be because second-generation

TSPO radioligands and [11C]-PK11195 bind to different sites (Kreisl et al., 2010; Owen et

al., 2010; Owen, et al., 2011).

Page 78: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

54

1.6.3.5 [18F]-FEPPA

[18F]-FEPPA is also one of these second generation TSPO radioligands developed to

overcome the limitations of the prototypical [11C]-PK11195 at the Centre for Addiction and

Mental Health (CAMH) (Wilson et al., 2008). The study 3 presented the preliminary data

using this radioligand to investigate neuroinflammation in the disease-associated regions in

PD patients.

1.6.3.5.1 Evaluations in vitro and ex vivo of [18F]-FEPPA

Evaluation studies in vitro and ex vivo demonstrated that [18F]-FEPPA is a promising TSPO

radioligand (Wilson et al., 2008). The affinity of [18F]-FEPPA (Ki = 0.07 nM) was threefold

more potent than [11C]-PBR28 (Ki = 0.22 nM) and an order of magnitude more potent than

[11C]-DPA713 (Ki = 0.87 nM) and the prototypical PK11195 (Ki = 1.29 nM). A

lipophilicity of [18F]-FEPPA (logD = 2.99) was suitable for brain penetration. Unmetabolized

[18F]-FEPPA accounted for 95.4% while unmetabolized [11C]-PBR28, only 84.6% at 40

minutes post injection. The metabolites were less lipophilic than [18F]-FEPPA in plasma and

brain extracts of rats as well as in human brains (Rusjan et al., 2011). The biodistribution of

[18F]-FEPPA study demonstrated a reasonable BBB penetration, brain uptake and slow

washout, as well as ubiquitous distribution of TSPO with high levels of radioactivity in the

olfactory bulb and hypothalamus, which is consistent with previous literature (Benavides et

al., 1983; Chaki et al., 1999; Venneti et al., 2007). Furthermore, in pig brains, [18F]-FEPPA

showed good and rapid brain uptake as well as appropriate regional distribution with the

highest uptake found in the thalamus while the lowest, in the cerebellum and frontal cortex.

Pretreatment with PK11195 resulted in a significant reduction in binding, confirming the

specific binding and reversible kinetics of [18F]-FEPPA (Bennacef et al., 2008). Lastly, in rat

model of PD, [18F]-FEPPA demonstrated greater sensitivity and selectivity to detect

inflammation compared with [11C]-PK11195. [18F]-FEPPA uptake was higher in the lesion

site, which was also more closely correlated with inflammatory cytokines, and lower in the

non-lesion site than [11C]-PK11195 (Hatano et al., 2010).

Page 79: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

55

1.6.3.5.2 Evaluation of quantification in vivo of [18F]-FEPPA in human

gray matter

Quantification of [18F]-FEPPA in human GM was evaluated to determine the appropriate (1)

kinetic modeling (1TCM or 2TCM), (2) outcome measures (VT, VS, or BPND), and (3) scan

length, as well as to determine the capacity for the quantification with increased specific

binding (Rusjan et al., 2011). Twelve healthy individuals consisting of four men and eight

women aged from 24 to 72 years old underwent a 180-min [18F]-FEPPA scan with arterial

sampling that generates a metabolite corrected plasma curve to be used as input function for

kinetic analyses. ROIs included the cerebellum, CN, putamen, frontal, temporal occipital,

insular, and ACC, and thalamus. %COV estimated the identifiability of the outcome

measures. Monte Carlo simulations were performed by increasing k3 in the thalamus that

showed the highest biding to determine the capacity of [18F]-FEPPA quantification.

Consistent with known distribution of TSPOs in human brain (Doble et al., 1987), the

radioactivity was widespread, which excludes reference region modeling for the kinetic

analysis of [18F]-FEPPA data. The 2TCM model provided better fit than the 1TCM model in

all of the ROIs, demonstrated by significantly lower AIC and higher Model Selection

Criterion. A two-hour scan appeared to be appropriate from the perspective of both

quantification of the outcome measures and the tolerance of subjects.

Using the two-hour scan data with 2TCM model, VT was the most appropriate binding

outcome measure as it showed better identifiability and less variability of the identifiability

across the ROIs compared to VS and BPND. It can also measure high level of TSPO density

(up to 500% increase in specific binding) with better identifiability than other outcome

measures. The highest VT binding was found in the thalamus followed by the temporal,

occipital, insular, and frontal cortex, cerebellum, ACC, putamen, and the lowest values were

found in the CN.

The study also demonstrated that radioactivity of skull was negligible, excluding a partial

volume effect in cortical ROIs. Furthermore, changes in blood blow did not affect the VT

quantification. This is important to study aging and pathological brains because increased

Page 80: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

56

blood flow can occur due to increased vascular permeability of disrupted BBB by

neuroinflammation in diseased brains (Winkeler, Boisgard, Martin, & Tavitian, 2010) or

because reduced blood flow can occur by aging (Iwata & Harano, 1986; Takahashi,

Yamaguchi, Kobayashi, & Yamamoto, 2005).

1.6.3.5.3 Evaluation of quantification in vivo of [18F]-FEPPA in human

white matter

Unlike the quantification of TSPO density in human GM (Rusjan et al., 2011), that of the

WM is challenging. This is due to the low level of TSPO in WM and its very slow washout,

making the identification of the small k4 value difficult (Rusjan et al., 2011). All of these

make the TSPO quantification in WM susceptible to noise. However, given the importance

of studying neuroinflammation in WM in various pathological brains, the quantification of

[18F]-FEPPA binding to TSPO in WM was investigated under several noise levels in 32HC

subjects (Suridjan, Rusjan, Kenk et al., 2014). The WM ROIs included the CC, cingulum,

superior longitudinal fasciculus (SLF), and posterior limb of internal capsule (PLIC). Using

the 2TCM, identifiability for VT was reported to be moderate ranging from %COV 15-19%.

Approximately 10% of the subjects showed very low VT identifiability (%COV > 30 %)

although these data did not affect the results. The effects of noise increased VT variability

and decreased its identifiability. However, the noise-induced bias in VT was relatively small.

Under the highest noise level, 6% of the data did not fit reliably. A simulation of increased

TSPO density showed minimal effect on VT variability and identifiability regardless noise

level, supporting the potential utility of [18F]-FEPPA in the WM (Suridjan et al., 2014).

1.6.3.5.4 TSPO polymorphism and [18F]-FEPPA VT

The relationship between the TSPO polymorphism and VT was investigated in 20 healthy

individuals (Mizrahi et al., 2012). ROIs included the frontal, temporal, occipital, insular, and

ACC, cerebellum, putamen, CN, and thalamus. Genotyping the rs6971 polymorphism

revealed that the study sample consisted of 13 HABs, six MABs, and one LAB. There were

significant ROI and genetic effects on VT without significant interactions. MABs showed a

Page 81: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

57

reduction of 27% (23-29%) in VT values on average across the ROIs compared to HABs.

However, there was overlap in VT values between MABs and HABs across the ROIs.

In contrast, there were no significant genetic effects on VT in any WM ROIs although MABs

showed on average 15% reduction in VT values than HABs (Suridjan et al., 2014). However,

excluding one MAB outlier, significant group differences appeared in the SLF, PLIC, and

cingulum, but not in CC. Considering differences in microstructure (e.g., myelin sheath in

WM), TSPO density, and kinetics of [18F]-FEPPA between GM and WM, a larger sample

size may be needed to test the polymorphism effects on VT (Suridjan et al., 2014).

1.6.3.5.5 [18F]-FEPPA study on neuroinflammation and aging

There was no significant age effect on [18F]-FEPPA VT values n 33 healthy individuals aged

from 19 to 82 years (22 HABs and 11 MABs) in the prefrontal cortex (PFC), dorsolateral

prefrontal cortex (DLPFC), and temporal cortex or hippocampus (Suridjan, Rusjan,

Voineskos et al., 2014) or in 32 healthy individuals aged from 19 to 78 years in the CC,

cingulum, SLF, and PLIC adjusted for the TSPO polymorphism (Suridjan et al., 2014). There

were also no interactions between age and the GM ROIs or between age and binding affinity

status in any of the GM ROIs.

1.6.3.5.6 [18F]-FEPPA studies in clinical populations

In addition to a case study of a patient with tumor (Ko et al., 2013), clinical populations such

as patients with SCZ (Kenk et al., 2015), AD (Suridjan et al., 2015), and major depression

episodes (MDE) (Setiawan et al., 2015) were investigated using [18F]-FEPPA. In two later

studies, the patients showed elevated TSPO binding in the disease/disorder related ROIs

compared to HC subjects and the TSPO binding was associated with clinical measures in

these patients (cognitive performance in AD patients and depression severity in MDE

patients). These two studies demonstrated the promise of [18F]-FEPPA to detect

neuroinflammation in clinical populations although there was the lack of evidence of

neuroinflammation in patients with SCZ, possibly due to the effects of antipsychotic

medications that can exert inhibitory effects on proinflammatory processes, and thus future

studies in medication-naïve patients are warranted (Kenk et al., 2015).

Page 82: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

58

1.6.3.5.7 TSPO imaging in Parkinson’s disease

To date, eight studies have been published for the investigation of neuroinflammation in PD.

All of these studies used [11C]-PK11195 and the results were inconclusive. Some studies

found that drug-naïve, early stage PD patients showed increased neuroinflammation in the

midbrain/SN (Iannaccone et al., 2013; Ouchi et al., 2005) and putamen (Iannaccone et al.,

2013) compared to HC subjects. The elevated neuroinflammation in the midbrain was also

associated with more severe motor symptoms as well as with more DA dysfunction in the

putamen (Ouchi et al., 2005). On the other hand, another study did not find significantly

elevated neuroinflammation in a similar sample size of PD patients with the clinical

characteristics (Bartels et al., 2010).

Two studies included more advanced stages of PD patients (Hoehn and Yahr stage 2 and 2-3)

did not find increased neuroinflammation in the nigro-striatal regions (Bartels et al., 2010;

Kobylecki et al., 2013). Instead, one of the studies found increased neuroinflammation in the

pons in the PD patients (Kobylecki et al., 2013). Another study including PD subjects with

moderate severity showed significant increase in neuroinflammation in the cortical regions

including the temporal and occipital regions using the ROI analysis (Edison et al., 2013) . A

voxel-based whole brain analysis further revealed that the PD patients showed increased

neuroinflammation in the frontal, temporal and occipital regions.

In a relatively larger sample size of 18 PD patients with varied severity, duration, and stages

of disease (including four drug naïve patients), neuroinflammation occurred in the striatum,

palllidum, thalamus, frontal and cingular regions, and pons while a whole brain region

analysis additionally found neuroinflammation in the hippocampus and cerebellum in these

patients (Gerhard et al., 2006). Interestingly, neuroinflammation decreased with longer

duration of disease in the cingular regions and thalamus.

Two of the studies mentioned above followed up some of the PD patients. In the study by

Ouchi, et al (2005), four PD patients were followed up four years later and showed the

consistent results (i.e., elevated neuroinflammation level in the midbrain, and the correlation

with motor severity and presynaptic DA dysfunction in the putamen). Additionally, higher

neuroinflammation was detected in the putamen and in the thalamus compared to the HC

Page 83: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

59

subjects and compared to their first assessment, suggesting a spread of neuroinflammation

within and beyond the nigrostriatal pathway (Ouchi et al., 2005). On the other hand, in the

other study where eight of the patients were followed up 18-28 months later, there were no

changes in neuroinflammation whereas the disease progressed, evidenced by an increase in

motor severity and further reduction in presynaptic DA dysfunction in the putamen in these

patients (Gerhard et al., 2006).

In more severe cases of PD patients with dementia, neuroinflammation primarily occurred in

cortical regions, which was also associated with MMSE scores (Edison et al., 2013; Fan,

Lindemann, Feuilloley, & Papadopoulos, 2012) suggesting that this outcome measure be

used to assess cognitive symptoms in PD. Interestingly, however, not all PD with dementia

(PDD) showed increased neuroinflammation (Edison et al., 2013). The authors argued that

the finding might be due to insufficient sensitivity of [11C]-PK11195.

As mentioned earlier, neuroinflammation has been studied in PD patients using [11C]-

PK11195 only. Studies using a second generation TSPO radioligand warrant elucidating

whether neuroinflammation occurs in PD patients. For this purpose, we used [18F]-FEPPA

(see section 2.3) to investigate the striatal regions in PD patients accounting for the TSPO

polymorphism.

Page 84: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

60

2.0 Aims and hypotheses

The general aim of this thesis was to use multimodal imaging tools to investigate structural,

functional, and molecular changes in PD and the associations with clinical manifestations.

The specific aim of each study was (1) to detect structural changes using CTA, which is

more sensitive for subtle cortical changes than the most commonly used VBM for gray

matter changes, and TBSS for white matter changes to elucidate the relationships between

structural abnormalities and clinical manifestations in PD, (2) to investigate changes in brain

network properties using the most advanced FC analysis method, graph theoretical analysis,

and (3) to first assess whether neuroinflammatory changes occur in PD using a second-

generation TSPO radioligand, developed to overcome the limitations of the prototype TSPO

radioligand.

2.1: Study1: Imaging changes associated with cognitive

abnormalities in Parkinson’s disease

The study 1 aimed to determine whether structural changes occurred in PD patients and

whether observed structural abnormalities were associated with the motor and cognitive

symptoms by investigating the whole brain GM and WM using CTA and TBSS. We

hypothesized that PD patients would show GM and WM abnormalities compared to age-

matched HC subjects and these changes would be correlated with clinical and cognitive

measures in PD patients.

Page 85: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

61

2.2 Study 2: Disrupted nodal and hub organization account for

brain network abnormalities in Parkinson’s disease

The study 2 aimed to determine whether the disease-associated nodal connections and

network hubs were affected in PD patients, and whether these changes were associated with

motor and cognitive symptoms using the graph theoretical approaches to rsfMRI data. We

hypothesized that PD would affect the nodal organization of regions such as SMA, Ins,

DLPFC, as well as the striatum, and the nodal and hub changes would account for motor and

cognitive symptoms of PD.

2.3: Study 3: Imaging striatal microglial activation in patients

with Parkinson’s disease

The study 3 aimed to evaluate whether a second generation TSPO radioligand, [18F]-FEPPA

could be used to detect neuroinflammation in PD patients. It would determine whether there

would be significant differences in VT values between HABs and MABs in PD patients like

those observed in HC subjects and in other clinical populations, and whether [18F]-FEPPA

VT values could differentiate PD patients from HC subjects. Using the striatum as our main

ROI, we hypothesized that PD-HABs would show higher VT values than PD-MABs, PD

patients would show increased neuroinflammation than HC subjects accounting for the

TSPO polymorphism, and the neuroinflammation would correlate with clinical measures of

PD.

Page 86: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

62

3.0 Study 1: Imaging changes associated with cognitive

abnormalities in Parkinson’s disease

This study was published in Brain Structure and Function.

This chapter has been modified from the following:

Koshimori, Y., Segra, B., Christopher, L., Lobaugh, N., Duff-Canning, S., Mizrahi, R.,

… Strafella, A. (2015). Imaging changes associated with cognitive abnormalities in

Parkinson’s disease. Brain Structure and Function, 220(4), 2249-61.

3.1 Introduction

PD is a multisystem neurodegenerative disease, affecting not only dopaminergic nerve cells

of the SN, but also other brain regions and neurotransmitters (Braak et al., 2006). PD

presents with the cardinal motor symptoms such as tremor, rigidity, bradykinesia, and loss of

postural stability along with a set of NMS (Bonnet, Jutras, Czernecki, Corvol, & Vidailhet,

2012) such as cognitive impairment, depression, sleep disturbances, and autonomic

dysfunction (Barnum & Tansey, 2012; Chaudhuri, Odin, Antonini, & Martinez-Martin,

2011; Ferrer, Martinez, Blanco, Dalfo, & Carmona, 2011). For this reason, PD research has

expanded its investigation beyond the nigrostriatal region to the whole brain in order to

characterize the different symptoms. In particular, neuroimaging investigation of cognitive

impairment is a topic of a growing interest (Christopher & Strafella, 2013). It is prevalent

and present regardless of the disease stage (Litvan et al., 2012). It ranges from mild deficits

demonstrable by means of comprehensive neuropsychological testing, to dementia (Jellinger,

2012), and MCI increases risk of developing dementia (Janvin, Larsen, Aarsland, &

Hugdahl, 2006). The neural substrates of motor and cognitive symptoms has been

investigated using non-invasive brain imaging such as structural MRI, which can offer the

opportunity of identifying early biomarkers. In fact, different MRI techniques are providing

Page 87: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

63

amounting evidence of both GM and WM changes in PD (Cochrane & Ebmeier, 2013; Pan et

al., 2012).

Whole brain GM changes have been investigated using VBM as well as surface-based

analyses including CTh and surface analyses combined with subcortical volumetric analysis.

PDD typically show bilateral diffuse GM changes (Beyer, Janvin, Larsen, & Aarsland, 2007;

Compta et al., 2012; Melzer et al., 2012; Song et al., 2011; Zarei et al., 2013). On the other

hand, non-demented PD patients show regional GM changes in areas such as frontal (Biundo

et al., 2011; Burton et al., 2004; Ibarretxe-Bilbao, Ramirez-Ruiz et al., 2010; Jubault et al.,

2009), temporal and/or limbic regions (Feldmann et al., 2008; Ibarretxe-Bilbao et al., 2009;

Pellicano et al., 2012; Tinaz et al., 2011; Wattendorf et al., 2009), or posterior regions

including the parieto-occipital cortex (Pereira et al., 2012). Furthermore, non-demented PD

patients show faster progression of GM changes including atrophy and cortical thinning than

healthy controls (HCs) in diffuse areas including frontal, temporal and parietal regions (Hu et

al., 2001; Ibarretxe-Bilbao et al., 2012), restricted regions to cortical motor areas and

cerebellum (Ibarretxe-Bilbao, Junque et al., 2010), or limbic, paralimbic, and temporo-

occipital regions (Ramirez-Ruiz et al., 2007). Among PD patients, variability in CTh and/or

subcortical volume is also associated with cognitive measures (Biundo et al., 2011; Camicioli

et al., 2009; Ibarretxe-Bilbao et al., 2009; Melzer et al., 2012; Pellicano et al., 2012; Zarei et

al., 2013), facial emotion recognition (Baggio et al., 2012), duration of disease (Hanganu et

al., 2013; Jubault et al., 2011; Lyoo et al., 2011), motor severity (Lyoo et al., 2011; Melzer et

al., 2012; Zarei et al., 2013) and stage (Zarei et al., 2013) as well as DA non-responsive

symptoms (Brenneis et al., 2003; Camicioli et al., 2009).

WM changes have been investigated using DTI. The two most common indices derived from

DTI are FA and or MD (Cochrane & Ebmeier, 2013). FA estimates the degree of anisotropic

directionality of water diffusion while MD estimates the magnitude/size of water diffusion.

FA and MD in the whole brain WM can be assessed using voxel-based analysis (VBA) or

TBSS. Contrary to GM findings, in which regional changes are more common in non-

demented PD, WM changes are reported in more diffuse brain areas even in non-demented

PD (Hattori et al., 2012; Kim et al., 2013; Melzer et al., 2013; Theilmann et al., 2013; Zheng

et al., 2014), and the frontal region was one of the most consistently reported regions for

Page 88: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

64

WM changes (Agosta et al., 2014; Deng et al., 2013; Gattellaro et al., 2009; Rae et al., 2012;

Zhan et al., 2012; K. Zhang et al., 2011). Moreover, variability in FA and/or MD was

primarily associated with cognitive measures (Agosta et al., 2014; Gallagher et al., 2013; Rae

et al., 2012; Theilmann et al., 2013).

Despite the evidence of structural abnormalities and the variability associated with clinical

and cognitive manifestations in non-demented PD, whether structural abnormalities account

for specific clinical sequelae in PD patients is still unclear. This is largely due to the lack of

imaging studies investigating both structural group differences, and relationships between

these differences and various clinical and cognitive measures. For example, the anatomical

regions displaying significant group differences in structure have not been specifically

investigated for correlation analyses (Camicioli et al., 2009; Hanganu et al., 2013; Jubault et

al., 2011; Tinaz et al., 2011). Even though brain regions demonstrating significant group

differences have been shown to correlate with cognition, PD patients did not show

impairment on cognitive tasks (Theilmann et al., 2013). Furthermore, only correlations were

demonstrated in PD without comparing structural data of PD with those of HCs (Lyoo et al.,

2011; Zheng et al., 2014). To date, only a handful of studies have addressed which structural

abnormalities account for different PD symptoms by investigating whole brain GM and WM

changes (Agosta et al., 2013; Agosta et al., 2014; Hattori et al., 2012). Using both VBM and

TBSS analyses, these studies have consistently concluded that WM and not GM changes

underlie cognitive impairment in PD (Agosta et al., 2013; Agosta et al., 2014). This

discrepancy may be due to the fact that VBM may not be highly sensitive for detecting subtle

cortical atrophy in early stages of PD (Agosta et al., 2013; Jubault et al., 2011). In fact, CTA

appeared more prone to detect cortical GM changes in PD than VBM when both analysis

methods were compared (Pereira et al., 2012). Thus based on these preliminary observations,

the current study aimed at (1) investigating both GM and WM changes using CTA and

TBSS, and thereby also demonstrating which MRI technique can be a promising biomarker

for PD and (2) further elucidating the relationships between observed structural

abnormalities and clinical and cognitive manifestations in non-demented PD patients.

Page 89: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

65

3.2 Methods

3.2.1 Participants

Twenty-six patients meeting UK Brain Bank criteria for the diagnosis of idiopathic PD

(Defer, Widner, Marie, Remy, & Levivier, 1999; Langston et al., 1992) and 15 HCs

participated in the study. PD patients were recruited from the Movement Disorders Clinic of

the Toronto Western Hospital. The HCs were recruited from friends and spouses of the

patients or through advertisements posted at the hospital and the affiliated university.

Exclusion criteria included (1) history of a head injury, psychiatric, neurological or major

medical diseases; (2) dementia assessed by a modified Disability Assessment for Dementia

(DAD) with an additional question regarding whether any reported impairment was related to

cognitive difficulties or the physical impairments of PD; (3) contraindications for MRI

scanning; and (4) for HCs evidence of cognitive impairment as assessed by a

neuropsychological test battery. All participants underwent a cognitive assessment by means

of an extended neuropsychological test battery and the Montreal Cognitive Assessment

(MoCA) (Nasreddine et al., 2005; Tison et al., 1995) as well as the Beck Depression

Inventory (BDI) (Beck, Ward, Mendelson, Mock, & Erbaugh, 1961) that assesses levels of

depression. PD patients were additionally evaluated for motor severity of the disease using

the motor subset of the UPDRS (UPDRS-III). All participants underwent structural MRI

scans and 35 participants had additional DTI scans. Among the 35 participants, five subjects

(four PD and one HC) were excluded due to motion artifacts resulting in 16 PD patients and

14 HCs included in the DTI analysis. PD patients underwent all the study procedures in an

“on-medication” state”. All participants gave informed consent following full explanation of

the study procedures. This study was approved by the institutional ethics committee of the

CAMH and the University Health Network.

3.2.2 Neuropsychological Assessment

Cognitive function in the domains of executive function, attention/working memory,

language, visuospatial function and memory were assessed using the following

neuropsychological tests (Litvan et al., 2012). For executive function: Visual Verbal Test

Page 90: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

66

total number of shifts (Wicklund, Johnson, & Weintraub, 2004), Delis-Kaplan Executive

Function System (D-KEFS) Color-Word Interference (Stroop task), time to complete

condition 3: inhibition, and D-KEFS Verbal Fluency, total score for category fluency

(Delis, Kaplan, & Kramer, 2001). For attention and working memory: Wechsler Memory

Scale-III (WMS-III) Digit Span and Letter-Number Sequencing total score (Wechsler,

1997), D-KEFS Color-Word Interference (Stroop task), and time to complete condition 1:

color naming (attention). For language: the category fluency from the Verbal Fluency

subtest (Delis et al., 2001). For visuospatial function: Judgment of Line Orientation (JLO)

total score (Benton, Sivan, Hamsher, Varney, & Spreen, 1994). For memory: California

Verbal Learning Test-II (CVLT-II) long delay free recall score (Delis, Kramer, Kaplan, &

Ober, 2002). Global composite z-scores were calculated for each domain of cognition.

When the cognitive domain had only one test, the individual test score was used. For

executive function the average of the z-scores from the visual verbal test, inhibition

segment of the Stroop task, and verbal fluency was used, while for attention/working

memory, the average of the z-scores from the digit span, letter-number sequencing, and

color-naming segment of the Stroop was used.

3.2.3 MRI Acquisition

Whole-brain T1-weighted and DWI images were acquired using a 3.0 T General Electric

(GE) Signa HD x MRI system (General Electric, Milwaukee, WI) equipped with an eight-

channel phased array head coil. For GM analysis, a high-resolution 3D anatomical scan was

acquired with a T1-weighted 3D IR-Fast Spoied Gradient-Recalled-Echo sequence

(TR/TE/inversion time (TI), 7.8/min full/450 ms; matrix, 256 x 256; voxel size, 1 x 1 x 1

mm; field of view (FOV), 256 x 256 mm; flip angle, 15°; 180 axial slices). For WM analysis,

a DWI scan was acquired with spin-echo single-shot echo planar imaging with diffusion

encoding in 60 noncolinear directions (TR/TE, 17,000/min ms; FOV, 230 x 230 mm; matrix,

128 x 128, voxel size, 1.8 x 1.8 x 2.4 mm; b value, 1000 s/mm2; 64 slices). Parallel imaging

was employed using the Array Spatial Sensitivity Encoding Technique (ASSET) with an

acceleration factor of two. DWI images were acquired in the axial plane. Additionally, ten

non-DWI scans were acquired at the beginning of each scan. The DWI scans were repeated

Page 91: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

67

three times to increase signal-to-noise ratio. Two PD patients and two HCs underwent only

two DWI scans due to inability to lie down in the scanner for an extended period of time.

3.2.3.1 Cortical Thickness Analysis

CTA was performed using the FreeSurfer image analysis suite (version 5.1; available at

http://surfer.nmr.mgh.harvard.edu). The processing of T1 high-resolution images for the

cortical surface reconstruction involved several steps (Dale, Fischl, & Sereno, 1999):

automated Talairach transformation, intensity normalization (Sled, Zijdenbos, & Evans,

1998), skull stripping, and WM segmentation, tessellation of the GM/WM boundary,

automated topology correction (Fischl, Liu, & Dale, 2001; Segonne, Pacheco, & Fischl,

2007), and surface deformation following intensity gradients to optimally place the

gray/white and gray/CSF borders at the location (Dale & Sereno, 1993; Dale et al., 1999;

Fischl & Dale, 2000). All surface models were visually inspected for accuracy. CTh was

calculated as the closest distance from the gray/white boundary to the gray/CSF boundary at

each vertex on the tessellated surface (Fischl & Dale, 2000).

3.2.3.2 Subcortical Volume Analysis

Segmentation of brain volume was obtained based on the automatic procedure included in

FreeSurfer (version 5.1; available at: http://surfer.nmr.harvard.edu) (Fischl & Dale, 2000).

The labels were created using an automated subcortical labeling algorithm based on a

probabilistic atlas obtained from a manually labeled training set. The image was rigid-body

registered to the probabilistic brain atlas, followed by non-linear morphing to the atlas. Then,

an automated segmentation procedure assigned a label to each voxel in a dataset based on

signal intensity information and the spatial relationship of the subcortical labels in the

training sets. Volumetric measures from 18 structures in each hemisphere as well as

intracranial volume (ICV) were automatically obtained. Among these structures, we selected

the thalamus, caudate, putamen, pallidum, NA, hippocampus, amygdala and brainstem for

our ROIs.

Page 92: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

68

3.2.3.3 TBSS

Individual FA and MD images were generated to conduct TBSS analysis using FSL tools

from the FMRIB software library (FSL version 4.1.5, http://www.fmrib.ox.ac.uk/) (Smith et

al., 2004; Woolrich et al., 2009). First, each volume upsampled to create isotropic voxel size

(2.4 x 2.4 x 2.4 mm) was affine registered to the 9th b0 volume using FSL Linear

Registration Tool (FLIRT) to correct motion and eddy current distortion (Jenkinson & Smith,

2001; Jenkinson, Bannister, Brady, & Smith, 2002). Then, an averaged DWI image was

generated from the three sets of DWI images. Non-brain tissue of the averaged DWI image

was removed using BET, brain extraction tool (Smith, 2002), and FA and MD images were

derived using DTIfit from FMRIB’s Diffusion Toolbox. The FA maps of all participants

were warped to the FMRIB58_FA template using FSL non-linear registration tool (FNIRT)

(Andersson, Jenkinson, & Smith, 2007a; Andersson, Jenkinson, & Smith, 2007b). A mean

FA map of all subjects was created and thinned to generate a mean FA skeleton, which

represents the centres of WM tracts common to all subjects included in the present study.

The mean skeleton was thresholded and binarized at FA value of 0.2 to minimize partial

voluming. The individual FA and MD maps were projected onto the mean skeleton resulting

in a skeletonised FA and MD maps.

3.2.3.4 Statistical Analysis

The CTA was performed using Freesurfer’s QDEC application that fits a general linear

model (GLM) at each surface vertex. CTh maps were smoothed using a circularly symmetric

Gaussian kernel across the surface with a full width at half maximum (FWHM) of 15 mm. Z

Monte Carlo simulations with 10,000 iterations were applied to CTh maps to provide

clusterwise correction for multiple comparisons, and the results were thresholded at a

corrected P value of 0.05 (Z = 1.3). Mean thickness was calculated for each significant

cluster.

Voxelwise statistics were performed on the skeletonized FA and MD maps using the

Threshold Free Cluster Enhancement (TFCE) (S. M. Smith & Nichols, 2009). A P < 0.05

voxelwise correction for multiple comparisons was considered significant. The Johns

Page 93: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

69

Hopkins University (JHU) DTI-based WM atlases including the JHU WM tractography atlas

and the International Consortium for Brain Mapping (ICBM)-DTI WM labels (Hua et al.,

2008; Mori, Wakana, Nagae-Poetscher, & van Zijl, 2005; Wakana et al., 2007) were used to

identify WM tracts that showed significant group differences and correlations in FA and MD

values.

Age, years of education, MoCA, BDI and neuropsychological tests were compared between

groups using independent samples t tests. Gender and handedness were compared using

Pearson’s chi-square tests. Subcortical volume in a priori ROIs (thalamus, putamen, caudate,

nucleus accumbens, globus pallidus (GP), hippocampus, amygdala, and brainstem) in each

hemisphere was compared between groups using independent t test or Analysis of Variance

(ANOVA) after testing the effects of ICV on the subcortical volume in each ROI.

From the clusters displaying significant group differences mean individual values were

extracted to investigate the relationships between structural changes (CTh, subcortical

volume, and DTI) and (1) cognitive data (MoCA and neuropsychological test scores) and (2)

clinical data (UPDRS-III scores and duration of disease). Prior to the correlation analyses,

bivariate correlation was performed among all relevant covariates including demographic

(age, gender, years of education, handedness), clinical (symptom-dominant side, UPDRS-III

scores, disease duration, Levodopa equivalent daily dose (LEDD), BDI), cognitive (MoCA,

visual verbal test, JLO, and global and executive composite z) and MRI findings from the

group analysis using Pearson’s correlation tests to determine the effects of nuisance

variables, which would be then controlled for in the correlations, if necessary. All statistical

analyses were two-sided and statistical significance was set at P < 0.05 corrected for multiple

comparisons. All of the statistical analyses were conducted using Statistical Package for the

Social Sciences (SPSS 13.0).

3.3 Results

3.3.1 Demographic, Clinical, and Cognitive Characteristics

Page 94: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

70

Table 3-1 shows the demographic and clinical characteristics as well as neuropsychological

data of all of 26 PD patients and 15 HCs. There was no difference between the two groups

regarding age, gender, education, BDI, and handedness (P > 0.05). However, they were

significantly different on MoCA scores (t = 2.88, P = 0.006), global composite z (t = 2.82, P

= 0.007), executive composite z scores (t = 3.48, P = 0.001), visual verbal test (t = 3.6, P <

0.001), and JLO (t = 2.3, P = 0.027).

Table 3-1. Demographic, clinical, and cognitive characteristics of PD patients and HCs.

PD (n = 26) HC (n = 15)

Age (years) 70.5 (5.6) 67.13 (5.1)

Sex (male/female) 13/13 4/11

Handedness (right/left) 24/2 14/1

Education (years) 15.6 (2.1) 17.0 (2.5)

MoCA 25.2 (2.8) 27.6 (2.2)**

BDI 6.5 (5.5) 3.8 (3.6)

Disease duration (years) 6.7 (4.2) -

Symptom-dominant side (right/left) 17/9

UPDRS-III (on-medication) 25.3 (15.3) -

Total LEDDa (mg/day) 731.3 (459.8) -

Neuropsychological tests

Global composite z -0.22 (0.60) 0.30 (0.50)**

Attention/working memory

composite z

0.12 (0.63) 0.27 (0.70)

Executive composite z -0.64 (0.89) 0.25 (0.60)**

Page 95: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

71

Digit span forward 0.31 (0.73) 0.46 (1.02)

California verbal test 0.19 (1.06) 0.23 (0.92)

Letter-number sequencing 0.20 (0.77) 0.15 (0.68)

Visual verbal test -2.21 (1.93) -0.380 (0.48)**

Judgment of line orientation -0.78 (1.57) 0.24 (0.88)*

STROOP

Color naming -1.44 (0.86) 0.19 (1.17)

Inhibition 0.13 (0.72) 0.29 (1.00)

Category fluency 0.21 (1.21) 0.86 (0.83)

Data are presented in mean (standard deviation) and neuropsychological data are presented in

group based mean Z-scores (standard deviation). aLEDD: Levodopa dose + 1-Levodopa-CR

x 0.75 + pramipexole (mg) x 67 (Evans et al., 2004). * P < 0.03, ** P < 0.01.

3.3.2 Cortical Thickness

We found that the PD patients showed significant reduction in CTh in the left superior-

frontal gyrus (SFG) (cluster size: 1585 mm2; P = 0.03, Talairach coordinates of maxima: x =

-18, y = 35.2, z = 39.3; HC = 2.72 ± 0.03 and PD = 2.51 ± 0.04) and the left precentral gyrus

extending back into the postcentral gyrus (cluster size: 2555.42 mm2; P = 0.01, Talairach

coordinates of maxima: x = -58.8, y = -5.4, z = 9.5; HC = 2.76 ± 0.03 and PD = 2.6 ± 0.03)

compared with the HCs (Figure 3-1). The PD patients did not show significant increase in

CTh in any brain regions compared with the HCs.

We further investigated whether the CTh in these two significant clusters was correlated with

clinical and cognitive measures that showed significant group difference (i.e., MoCA, visual

verbal test, JLO, and global and executive composite z scores) in PD patients. To this end,

Page 96: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

72

first, we ran bivariate correlations among different covariates including the CTh values in the

SFG and precentral gyrus clusters and demographic, clinical and cognitive measures to

determine the effects of nuisance variables. In particular, age showed significant negative

correlation both with SFG CTh (r = -0.497, P = 0.01) and cognitive scores including MoCA

(r = -0.423, P = 0.031), visual verbal tests (r = -0.499, P = 0.009), and executive z scores (r =

-0.541, P = 0.004). Thus controlling for the effect of age, we found significant positive

correlations between the SFG thickness and global composite z (r = 0.597, P = 0.002), and

executive composite z (r = 0.430, P = 0.032) scores (Figure 3-2). Similarly, we performed

the partial correlations between the SFG CTh and the cognitive scores, controlling for age in

HCs. In HCs, we found a significant positive correlation between the SFG thickness and

visual verbal test scores (r = 0.607, P = 0.016). These findings suggest that the SFG thinning

was uniquely associated with poor executive and global cognitive performance in PD

patients.

Page 97: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

73

Figure 3-1. a. Cortical areas showing significant cortical thinning in PD patients compared

to HCs. Color bar indicates the significance levels in the clusters in Z values. b. Bar

graphs on extracted CTh values (mm) from the significant clusters in the left SFG and left

precentral gyrus between HCs and PD patients. Error bars represent s.e.m. Figure

reproduced with permission from (Koshimori, Segura et al., 2015).

Page 98: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

74

Figure 3-2. Partial correlations with age as a covariate between SFG thickness (extracted

from the significant cluster) and global composite z (left) and executive composite z (right)

in 26 PD patients showing the significant positive correlations. Figure reproduced with

permission from (Koshimori, Segura et al., 2015).

3.3.3 Subcortical Volume

We found no significant group differences between PD patients and HCs in any subcortical

ROIs and therefore, no correlation analysis was pursued.

3.3.4 White Matter

We found that PD patients showed WM changes with significantly higher MD values

compared to HCs (PD: 0.86 ± 0.01 and HC: 0.81 ± 0.006). The significant cluster with

increased MD was detected in widespread cortical WM regions including the frontal,

temporal, parietal and occipital regions as well as subcortical white matter regions (Figure 3-

3). These changes were located primarily in a larger area of bilateral frontal and temporal

regions and smaller areas of the left parietal and occipital regions. More specifically, WM

tracts with MD changes included forceps minor, cingulum, anterior thalamic radiation

(ATR), SCR, external capsule (EC), body of CC, uncinate fasciculus (UF), inferior fronto-

occipital fasciculus (IFO), SLF and inferior longitudinal fasciculus (ILF), and forceps major.

PD patients did not show significantly lower MD values or any differences in FA values

compared with HCs.

Page 99: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

75

We extracted mean MD values from the significant cluster resulting from the group analysis

for each subject and investigated correlations between the MD values and (1) cognitive

measures that showed significant group differences (i.e., visual verbal test, JLO, global

composite and executive composite z scores) in both PD patients and HCs as well as clinical

measures (duration of disease and UPDRS-III scores) in PD patients. We first ran bivariate

correlations among all relevant covariates. In PD patients we found a significant negative

correlation between age and executive z scores (r = -0.622, P = 0.01) and a positive

correlation between age and the mean MD values in the significant cluster (r = 0.525, P =

0.037). In this group of patients, we also observed a significant correlation between gender

and cognitive measures (global composite z score (r = 0.667, P = 0.005) and executive

composite z score (r = 0.583, P = 0.018)). Thus, controlling for the effects of age and gender,

we found significant negative correlations between the MD values and global composite z

score (r = -0.39, P < 0.05) and executive composite z score (r = -0.44, P = 0.018) in PD

patients, suggesting that WM damage (mainly in frontal and temporal regions) was

associated with cognitive impairment. In HCs, we found no correlations among any of the

considered variables.

Page 100: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

76

Page 101: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

77

Figure 3-3. a. Clusters of significantly increased MD in 16 patients with PD compared

with 15 HCs in TBSS. Result images are overlaid on the MNI152 template. b. Bar graphs

on mean MD derived from the significant clusters in TBSS between HC and PD groups.

Error bars represent s.e.m. Reproduced with permission from (Koshimori, Segura et al.,

2015).

3.4 Discussion

The present study corroborated structural changes in PD patients and demonstrated specific

relationships between GM and WM damages and cognitive deficits in PD, suggesting that

these abnormalities may represent a sensitive biomarker for detecting brain changes

associated with cognitive changes.

Our results provided evidence of GM changes at the cortical level in PD. PD patients showed

significant cortical thinning in the left superior frontal, caudal middle frontal, precentral and

postcentral gyri compared to HCs. GM changes in these regions have previously been

reported in non-demented PD patients (Kostic et al., 2010; Melzer et al., 2012; Pereira et al.,

2012; Zarei et al., 2013). In addition, the superior frontal, caudal middle frontal gyrus (MFG)

and precentral sulcus were among the areas showing significantly greater progression of

cortical thinning in PD patients compared to HCs (Ibarretxe-Bilbao et al., 2012).

We further investigated whether these cortical abnormalities would explain the clinical

sequelae including motor and cognitive manifestations in PD and found that cortical thinning

in the dorsolateral SFG was associated with global and executive cognitive measures. The

dorsolateral SFG including BA8 and BA9 that showed significant thinning, is involved in a

variety of cognitive functions including WM (Levy & Goldman-Rakic, 2000; Owen et al.,

1998), attention (Corbetta, Patel, & Shulman, 2008), episodic memory (Desgranges, Baron,

& Eustache, 1998), and spatial cognition (Courtney, Petit, Maisog, Ungerleider, & Haxby,

1998; du Boisgueheneuc et al., 2006). In particular, the left SFG is involved in higher levels

of cognitive processing (du Boisgueheneuc et al., 2006) or executive functions. This may

well explain the significant correlation between the SFG thinning and global composite and

Page 102: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

78

executive z scores. We also found a significant positive correlation between the left SFG

thickness and visual verbal test scores in HCs but not in PD patients. This is most likely due

to a lack of variability in the scores of the PD patients.

We did not find any correlation between the reduced CTh in the sensorimotor area and

UPDRS-III scores or duration of disease. These observations seem to be consistent with

previous studies where cortical thinning was found in motor areas including the left medial

SMA and right dorsal pre-SMA without any correlation with UPDRS-III scores or disease

duration in those regions (Jubault et al., 2011). On the other hand, correlations between

cortical GM measures in the sensorimotor area and motor symptoms (Lyoo et al., 2011;

Rosenberg-Katz et al., 2013) as well as between CTh in several cortical areas and duration of

disease (Lyoo et al., 2011; Rosenberg-Katz et al., 2013) have been reported in other studies.

However, it is unknown whether PD patients had cortical abnormalities in those areas

compared to HCs, as those studies did not include control data. The lack of a significant

correlation between cortical abnormality in the sensorimotor area and UPDRS-III scores in

our patients may well be due to the fact that UPDRS evaluations were performed during an

on-medication state (instead of during an off-medication state), and this could have very

likely diminished the possibility of detecting a significant relationship with the cortical

thinning in those regions.

We did not find any changes in subcortical volume between groups. Previous studies using

the same method also failed to find significant group differences (Tinaz et al., 2011; Zarei et

al., 2013). These consistent observations suggest that current imaging analysis may lack

sensitivity for detecting subtle subcortical GM changes.

Our TBSS analysis revealed that PD patients showed WM damage in multiple WM tracts in

widespread areas, more extensively in the bilateral frontal and temporal regions compared to

HCs. WM changes in these regions were consistently reported in previous TBSS findings

(Agosta et al., 2013; Agosta et al., 2014; Deng et al., 2013; Hattori et al., 2012; Matsui,

Nishinaka, Oda, Niikawa, Kubori et al., 2007; Rae et al., 2012). We detected group

differences with an increase in MD (but not in FA) in our patients. MD appears to be more

sensitive in detecting subtle WM changes than FA as suggested in other studies in early PD

Page 103: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

79

(Melzer et al., 2013), early AD patients (Acosta-Cabronero, Williams, Pengas, & Nestor,

2010) and individuals with concussion (Cubon, Putukian, Boyer, & Dettwiler, 2011).

We further found that the fronto-temporal WM damage significantly correlated with the

global and executive cognitive measures. Our findings combined with previous studies

(Agosta et al., 2013; Gallagher et al., 2013; Hattori et al., 2012; Melzer et al., 2013) suggest

that frontal WM damage is a core pathological substrate of mild cognitive deficits in PD.

WM damage did not correlate UPDRS-III scores. This is very likely because UPDRS-III

evaluation was representative of the on-medication state as the same explanation provided

above.

Our GM and WM data consistently showed structural changes in the frontal region in PD.

The frontal cortex includes part of the fronto-striatal loops (Alexander, DeLong, & Strick,

1986), which have important implications for motor and NMS of PD. Prefronto-striatal

dysfunction is thought to underlie the basis for the most prominent executive impairment in

PD (Nagano-Saito et al., 2013; Owen, 2004; Pagonabarraga & Kulisevsky, 2012; Zgaljardic

et al., 2006). Abnormality in the SFG in particular can be an early indicator for further

decline of cognitive function. For example, PD patients who converted to dementia showed

cortical thinning in the frontal regions including the SFG, precentral gyrus, and anterior

cingulate at a baseline assessment and showed wider areas of cortical thinning in temporal,

parietal and occipital regions at a follow-up assessment (Compta et al., 2013). The WM

underlying the PFC contains projections from striatum via thalamus, to the striatum via

thalamus and directly to striatum. Although the TBSS does not allow for specific

identification of the fronto-striatal pathway, it reveals that the WM comprising the fronto-

striato-thalamic loop was affected in our PD patients. For example, the ATR includes WM

tracts from thalamus to PFC and vice versa. The lateral anterior ventral and medial dorsal

nuclei of thalamus, in particular, receive input from the BG. WM damage found in our study

extended beyond the frontal region and thus, it is still to be determined whether WM changes

in the prefrontal region alone can contribute to cognitive impairment. However, prefrontal

WM alone appears to be able to contribute to executive cognitive functions in PD (Gallagher

et al., 2013).

Page 104: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

80

Our PD patients also showed extensive WM changes in bilateral temporal regions. A few

studies consistently showed that PD with MCI may be associated with GM atrophy in limited

regions of frontal and temporal regions (Hanganu et al., 2013; Melzer et al., 2012; Song et

al., 2011) while one of them additionally showed parietal volume loss (Melzer et al., 2012)

and others, occipital volume loss (Hanganu et al., 2013; Song et al., 2011). A longitudinal

study also demonstrated higher rate of cortical thinning in the frontal and temporal regions,

extending to parietal cortex (Ibarretxe-Bilbao et al., 2012). Although our PD patients did not

show GM abnormalities in the temporal region, the WM changes may have preceded GM

changes. The topography of WM changes in our study is similar to the previous GM findings

mentioned above with extensive frontal and temporal abnormalities as core changes and

unilateral focal parietal and occipital changes. Abnormality in the temporal region may be

associated with developing dementia in PD patients. For example, compared to the PD with

normal cognition, PD-MCI and PDD had significantly smaller hippocampal volumes, and

PDD additionally showed the medial temporal lobe atrophy (Weintraub et al., 2011).

Furthermore, PDD showed significant atrophy in the entorhinal cortex compared with PD

with normal cognition (Goldman et al., 2012).

Our findings in GM and WM changes in PD are in line with previous studies showing

widespread WM abnormalities but limited (Agosta et al., 2014) or absent (Agosta et al.,

2013; Hattori et al., 2012) cortical GM changes. However, differently from those studies

using VBM, the significant GM abnormalities reported here seem to suggest that CTA may

be a better approach. Thus, the combination of DTI and CTA appear to be sensitive

approaches for detecting subtle WM and GM changes associated with PD.

Although the MRI techniques used in the present study are validated methods to assess

structural changes, the biological underpinnings of these changes are not fully understood.

Subtle cortical thinning may reflect changes in size of cell bodies, dendritic arborisation,

and/or presynaptic terminals (Morrison & Hof, 1997; Pellicano et al., 2012). Changes in DTI

indices can result from a number of processes including neuronal loss and gliosis, as well as

disturbances in axonal membranes, myelin sheath, microtubules, and neurofilaments

(Shenton et al., 2012). PD has been associated with cytoskeletal damage of various neuronal

cells including dopaminergic, glutamatergic, cholinergic, tryptaminergic, GABAergic,

Page 105: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

81

noradrenergic and adrenergic neurons (Braak et al., 1994; Braak et al., 1995; Braak, de Vos,

Jansen, Bratzke, & Braak, 1998; Foley & Riederer, 1999; Jellinger, 1991). The cytoskeletal

damage leads to Lewy pathologies including Lewy bodies and Lewy neuritis mostly located

in presynaptic terminals and in axons of affected nerve cells respectively (Braak,

Ghebremedhin, Rub, Bratzke, & Del Tredici, 2004). The major components of Lewy

pathologies include aggregations of misfolded α-synuclein (Braak et al., 2004) and

abnormally phosphorylated neurofilaments (Braak & Braak, 2000). Thus, MRI changes may

reflect the Lewy pathologies and/or neuronal degeneration secondary to the Lewy

pathologies.

The current study has a few potential limitations. First, PD patients included in the present

study had been taking parkinsonian medications for quite some time. To date, the effects of

chronic dopaminergic medication on brain structures remain to be determined. Second, our

PD patients underwent all study procedures in a on-medication state. It is well known that

dopaminergic medications in general can influence cognition (Kehagia, Barker, & Robbins,

2010). For example, while they can ameliorate certain cognitive deficits (e.g. executive

functions), dopaminergic medications can also worsen other cognitive abilities (Kehagia et

al., 2010; MacDonald et al., 2013; Ryterska, Jahanshahi, & Osman, 2013). Our decision not

to study them in an off-medication state was justified by the risk of worsening their motor

symptoms, increasing the risk of motion artifacts during MRI acquisitions.

In conclusion, the current study employed the CTA and TBSS to demonstrate that both GM

and WM are affected in PD patients and these anatomical changes may represent the neural

substrates underlying mild cognitive deficits in non-demented PD patients. Our findings

suggest that the dorsolateral SFG and fronto-temporal WM integrity play an important role in

global and executive cognitive performance, which are potential biomarkers for earlier

cognitive impairment in PD patients. In addition, distributed WM pathology (in contrast to

localized GM pathology in the frontal region) suggests that WM changes may precede

cortical GM changes and that WM changes be a potential biomarker for the early stage of

pathology. Taken together, the structural changes in the frontal region in particular may be an

early pathological substrates of cognitive impairment of PD, and may represent a sensitive

biomarker for brain changes in PD patients.

Page 106: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

82

4.0 Study 2: Disrupted nodal and hub organization

account for brain network abnormalities in Parkinson’s

disease

Yuko Koshimori, Sang-Soo Cho, Marion Criaud, Leigh Christopher, Mark Jacobs, Christine

Ghadery, … Antonio P. Strafella.

4.1 Introduction

The clinical phenotypes of PD include cardinal motor symptoms such as tremor, rigidity,

bradykinesia, and loss of postural stability, along with a set of NMS such as depression, sleep

disturbances, autonomic dysfunction (Bonnet et al., 2012), and cognitive impairment

(Barnum & Tansey, 2012; Bonnet et al., 2012; Chaudhuri et al., 2011; Ferrer et al., 2011).

Cognitive impairment ranging from MCI in different cognitive domains (e.g., attention,

executive, visuospatial, and memory) to dementia is derived from dysfunction of different

neurotransmitter systems/brain networks (Gratwicke et al., 2015). The recent application of

graph theory to brain networks (Bullmore & Sporns, 2009) may shed light on complex

diseases such as PD.

The graph theoretical approaches have revealed that human brains show the following

topological characteristics (Bullmore & Sporns, 2009; Bullmore & Sporns, 2012). First,

individual brain regions termed ‘nodes’ typically have disproportionate connections with

other nodes. Highly connected nodes are considered as hubs, which play a crucial role in

integrating information. The anterior and posterior cingulate cortices, Ins, and superior

frontal and parietal cortex are identified as both structural and functional hubs (Sporns, 2014;

van den Heuvel & Sporns, 2013). These are heteromodal areas that are involved in a broad

range of cognitive processes (Achard, Salvador, Whitcher, Suckling, & Bullmore, 2006;

Mesulam, 1998). Second, some nodes are more closely connected to one another and form

modules/subnetworks, which promote specialized processing and functional segregation.

Large-scale modules correspond to well-established resting-state functional networks (Engel,

Gerloff, Hilgetag, & Nolte, 2013; Stam, 2014). Third, these modules are also interconnected

Page 107: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

83

via hubs with long-distance pathways and thereby promote functional integration. Brain

networks display small-world properties of efficient global and local parallel information

transfer (i.e., integration and segregation) at relatively low connection cost (Achard &

Bullmore, 2007), which reflects an optimal organization through evolution and development

(Bullmore & Sporns, 2012; Gong et al., 2009). The coexistence and balance of segregation

and integration of subnetworks are fundamental for brain function (Bullmore & Sporns,

2012; Sporns, 2013).

There is evidence that the pathological course of neurodegenerative diseases, such as AD,

target network hubs (Seeley, Crawford, Zhou, Miller, & Greicius, 2009; Stam et al., 2009),

and selective damage to hubs can significantly change the integration of brain networks

(Bullmore & Sporns, 2012; Gong et al., 2009). With the dysfunction of multiple

neurotransmitters (Gratwicke et al., 2015; Pagonabarraga et al., 2015) and deposition of α-

synuclein (Braak et al., 2005), network hubs are almost certainly vulnerable in PD (Stam,

2014), and the selective deterioration of these network hubs may account for the distributed

abnormalities across the brain in PD (McColgan et al., 2015) with distinct clinical

correlations.

The premotor area and pre-supplementary motor area (SMA) have been identified as key

regions connecting multiple networks in healthy adults (Spreng et al., 2013). These regions

are important for self-initiated movements and preparation for actions (Passingham,

Bengtsson, & Lau, 2010). The SMA consisting of SMA proper and pre-SMA, is part of the

BC thalamo-cortical motor circuit associated with DA (Alexander et al., 1986; Krack, Hariz,

Baunez, Guridi, & Obeso, 2010; Obeso & Lanciego, 2011) and the changes in FC have been

well documented in PD patients, along with other cortical motor areas such as the primary

motor cortex (M1) and premotor area (Haslinger et al., 2001; Sabatini et al., 2000; Wu, Long

et al., 2009; Yu, Sternad, Corcos, & Vaillancourt, 2007).

DLPFC has been implicated as an anatomical and a functional node associated with

executive functions (Gong et al., 2009; Spreng et al., 2013; van den Heuvel & Sporns, 2013).

This brain region is part of FPN or executive control network (Dosenbach et al., 2007;

Seeley et al., 2007; Trujillo et al., 2015), as well as the fronto-striatal DA network

Page 108: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

84

(Gratwicke et al., 2015). PD patients often show executive dysfunction due to a disruption of

the fronto-striatal DA system (Gratwicke et al., 2015). In addition, structural (Koshimori,

Segura et al., 2015) and functional (Li, Liang, Jia, & Li, 2016; Trujillo et al., 2015; Wu et al.,

2015) changes in this cortical area and their association with cognitive impairment have been

reported consistently in these patients (Koshimori, Segura et al., 2015).

Another important disease-associated hub in PD is the insular cortex, which connects and

integrates several functional systems: the anterior area is involved in cognitive and

behavioral/emotional functions, and the mid to posterior area involved in sensorimotor

functions (Kurth, Zilles, Fox, Laird, & Eickhoff, 2010). The insular cortex has extensive

connections with the striatum in a connectivity gradient from posterior to anterior

(Christopher, Koshimori, Lang, Criaud, & Strafella, 2014). In post-mortem PD brains, α-

synuclein depositions become evident throughout the Ins at Braak’s stage V (Braak et al.,

2005). In addition, the level of CSF α-synuclein concentration was significantly associated

with anterior insular network disruption (Madhyastha et al., 2015). The anterior insula (AIns)

is a core region of the SAL that guides behaviour (Menon & Uddin, 2010), and this brain

network can affect other cognitive subnetworks such as the DMN (Bonnelle et al., 2012;

Chiong et al., 2013). Graph theoretical analyses have revealed a decreased hub role in the left

dorsal AIns in PD patients compared to HCs (Tinaz et al., 2016). Furthermore, recent data

from our group have demonstrated that dopamine D2 receptor (D2 receptor) availability in

the AIns was associated with executive and memory dysfunction in PD-MCI (Christopher,

Duff-Canning et al., 2014; Christopher et al., 2015). Thus, the AIns, in particular, seems to

be a critical “hub” for cognitive impairment in parkinsonian patients (Christopher, Koshimori

et al., 2014; Criaud et al., 2016).

The present study aimed to investigate whether nodal connections and network hubs were

affected in PD patients, and whether these changes were associated with motor and NMS

employing graph theoretical approaches to rs-fMRI data. We hypothesized that (1) PD would

affect the nodal organization of regions such as the SMA, Ins, DLPFC, as well as the

striatum, and (2) the nodal and hub changes would account for some of the motor and

cognitive symptoms of PD.

Page 109: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

85

4.2 Methods

4.2.1 Participants

Forty-five patients meeting the UK Brain Bank criteria for the diagnosis of idiopathic PD and

25 HCs participated in the study. During the screening session, all participants provided

written informed consent following a full explanation of the study procedures. Subsequently,

they were assessed for their global cognitive performance using the MoCA and their level of

depression using the BDI II. Additionally, patients were assessed for their motor severity of

the disease using the motor subset of the UPDRS (UPDRS-III) and for disease stage using

the Hoehn and Yahr. All the participants underwent a structural and rsfMRI scan. Patients

underwent the study procedures in an on-medication state. We reasoned that (1) scanning

patients in an on-medication state would minimize participants’ motion effects on rs-fMRI

data (Mowinckel, Espeseth, & Westlye, 2012; Tahmasian, Bettray, van Eimeren, Drzezga,

Timmermann, Eickhoff, et al., 2015) and (2) studying chronically medicated patients would

permit us to capture the complex neural substrates of cognitive impairment because cognitive

impairment in patients is mostly likely to be derived from dysfunction of multiple

neurotransmitters (Gratwicke et al., 2015). Exclusion criteria for the participants included:

(1) history of a head injury, psychiatric or neurological diseases (except PD for the patients),

(2) alcohol or drug dependency or abuse, (3) contraindications for MRI scanning, (4)

dyskinesia and dystonia for PD patients, and (5) for HCs, MoCA scores < 26 and BDI II

scores > 13. The study was approved by the CAMH Research Ethics Board.

4.2.2 MRI image acquisition

MR images were acquired with a GE Discovery MR 750 3T scanner with 8-channel head

coil. The protocol included whole-brain anatomic T1-weighted MRI images (Fast Spoiled

Gradient Echo pulse sequence; 200 sagittal slices; matrix of 256 x 256; TR: 6.7 ms; TE: 3.0

ms; slice thickness: 0.9 mm; FOV: 23 cm; Inversion Time: 650 ms; flip angle = 8°) and rs-

fMRI images (Gradient Echo/Fast Gradient Echo pulse sequence, 31 axial slices; matrix of

64 x 64; TR: 2000 ms; TE: 30 ms; flip angle = 60°, FOV: 22 cm; slice thickness: 5 mm;

duration: approximately 8min). During the rsfMRI scan, the participants were instructed to

Page 110: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

86

keep their eyes open, let their mind wander, not to think about anything in particular, and not

to fall asleep. The first two volumes of the rsfMRI images were removed to allow for

magnetization equilibrium, resulting in the preprocessing of 240 volumes.

4.2.3 rsfMRI preprocessing

The functional data were preprocessed for each subject using the Conn FC toolbox version

15a (Whitfield-Gabrieli & Nieto-Castanon, 2012) including slice timing correction, motion

correction using a 6 degree rigid spatial transformation, which provided the spatial deviation

for each time point for translational (x, y, z) and rotational (roll, pitch, yaw) directions of

movement, normalization into standard MNI space using non-linear transformations, and

smoothing with a Gaussian smoothing kernel of 6 mm FWHM. Outliers in the global signal

of brain activation and movement were examined using the artifact detection toolbox. Time

points were considered as outliers if the global signal of brain activation exceeded three

standard deviations of the mean or if movement exceeded 0.5 mm across translational and

rotational directions of scan-to-scan deviation. Participants were excluded from the analyses

if outliers accounted for greater than 20 % of the entire dataset. Among the 70 participants,

five participants (two HCs and three patients) were excluded due to excessive head motion

and artifacts, resulting in 23 HCs and 42 patients included in the graph theoretical analysis.

Nuisance signals including six motion parameters as well as the signal from WM and CSF

voxels were regressed using aCompCor, the component–based noise correction method

(Behzadi, Restom, Liau, & Liu, 2007). The residual datasets were then temporally filtered

(0.008 < f < 0.09).

4.2.4 Network nodes

Among 160 regions of interest, of six different neural networks, derived from a series of five

meta-analyses of fMRI activation studies combined with cognitive control nodes identified

during error-processing (Dosenbach et al., 2007; 2008), we selected 120 regions of interest in

spheres with a radius of 5 mm in four networks (Figure. 4-1 A-D and Table 4-1) to

investigate the hub regions associated with motor and cognitive symptoms of PD. These

Page 111: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

87

included 34 nodes of the DMN, 21 nodes of the FPN, 32 nodes of the cingulo-opercular

network (CON), and 33 nodes of the SMN.

Figure 4-1. Medial and lateral views of brain images with 120 regions of interest of four

subnetworks (Dosenbach et al., 2010). 34 nodes in red are affiliated with DMN (default

mode network); 21 nodes in blue, with FPN (fronto-parietal network); 32 nodes in green,

with CON (cinculo-opercular network); and 33 nodes in purple, with SMN (sensorimotor

network). Brain image and nodes are visualized using the BrainNet Viewer (NKLCNL,

Beijing Normal University).

DMN FPN

CON SMN

R R L L

Page 112: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

88

Table 4-1 Node labels of four networks and their MNI coordinates

Network Node Coordinate#

X Y Z

Default mode

network

R ventromedial PFC 6 64 3

Medial PFC 0 51 32

L anterior PFC -25 51 27

R ventromedial PFC 9 51 16

L ventromedial PFC -6 50 -1

L ventromedial PFC -11 45 17

R ventromedial PFC 8 42 -5

R anterior cingulate cortex 9 39 20

R ventrolateral PFC 46 39 -15

R superior frontal cortex 23 33 47

L superior frontal cortex -16 29 54

R inferior temporal cortex 52 -15 -13

L inferior temporal cortex -59 -25 -15

R posterior cingulate cortex 1 -26 31

R fusiform gyrus 28 -37 -15

L precuneus -3 -38 45

L posterior cingulate cortex -8 -41 3

L inferior temporal cortex -61 -41 -2

L occipital -28 -42 -11

L posterior cingulate cortex -5 -43 25

R precuneus 9 -43 25

R precuneus 5 -50 33

L posterior cingulate cortex -5 -52 17

R posterior cingulate cortex 10 -55 17

L precuneus -6 -56 29

L posterior cingulate cortex -11 -58 17

R angular gyrus 51 -59 34

L angular gyrus -48 -63 35

R precuneus 11 -68 42

L intraparietal sulcus -36 -69 40

L occipital cortex -9 -72 41

R occipital cortex 45 -72 29

L occipital cortex -2 -75 32

L occipital cortex -42 -76 26

Fronto-parietal

network

R anterior PFC 29 57 18

L anterior PFC -29 57 10

R ventral anterior PFC 42 48 -3

L ventral anterior PFC -43 47 2

R ventrolateral PFC 39 42 16

R dorsolateral PFC 40 36 29

L anterior cingulate cortex -1 28 40

Page 113: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

89

R dorsolateral PFC 46 28 31

L ventral PFC -52 28 17

L dorsolateral PFC -44 27 33

R dorsal frontal cortex 40 17 40

R dorsal frontal cortex 44 8 34

L dorsal frontal cortex -42 7 36

L inferior parietal lobule -41 -40 42

R inferior parietal lobule 54 -44 43

L posterior parietal cortex -35 -46 48

L inferior parietal lobule -48 -47 49

L inferior parietal lobule -53 -50 39

R inferior parietal lobule 44 -52 47

L intraparietal sulcus -32 -58 46

R intraparietal sulcus 32 -59 41

Cingulo-opercular

network

R anterior PFC 27 49 26

R ventral PFC 34 32 7

L anterior cingulate cortex -2 30 27

R ventral FC 51 23 8

R anterior insula 38 21 -1

R dorsal anterior cingulate cortex 9 20 34

L anterior insula -36 18 2

L basal ganglia -6 17 34

Medial frontal cortex 0 15 45

L ventral frontal cortex -46 10 14

L basal ganglia -20 6 7

R basal ganglia 14 6 7

L ventral frontal cortex -48 6 1

R mid insula 37 -2 -3

L thalamus -12 -3 13

L thalamus -12 -12 6

R thalamus 11 -12 6

R mid insula 32 -12 2

L mid insula -30 -14 1

R basal ganglia 11 -24 2

L posterior insula -30 -28 9

R temporal cortex 51 -30 5

L posterior cingulate -4 -31 -4

R fusiform gyrus 54 -31 -18

R precuneus 8 -40 50

R parietal cortex 58 -41 20

R temporal cortex 43 -43 8

L parietal cortex -55 -44 30

R superior temporal cortex 42 -46 21

L angular gyrus -41 -47 29

L temporal cortex -59 -47 11

Page 114: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

90

L: left; PFC: prefrontal cortex; R: right; # the Montreal Neurological Institute coordinates

describe the center of node area.

4.2.5 Computation of network measures

Network measures were computed with age as a covariate using a Graph-Theoretical

Analysis Toolbox (GAT) (Hosseini, Hoeft, & Kesler, 2012). This toolbox constructs a binary

L temporoparietal junction -52 -63 15

Sensorimotor

network

R frontal cortex 58 11 14

R dorsal frontal cortex 60 8 34

L ventral frontal cortex -55 7 23

R pre-supplementary motor area 10 5 51

R ventral frontal cortex 43 1 12

Supplementary motor area 0 -1 52

R frontal cortex 53 -3 32

R precentral gyrus 58 -3 17

L MIns -42 -3 11

L precentral gyrus -44 -6 49

L parietal cortex -26 -8 54

R precentral gyrus 46 -8 24

L precentral gyrus -54 -9 23

R precentral gyrus 44 -11 38

L parietal cortex -47 -12 36

R MIns 33 -12 16

L MIns -36 -12 15

R temporal cortex 59 -13 8

L parietal cortex -38 -15 59

L parietal cortex -47 -18 50

R parietal cortex 46 -20 45

L parietal cortex -55 -22 38

L precentral gyrus -54 -22 22

L temporal cortex -54 -22 9

R parietal cortex 41 -23 55

R PostIns 42 -24 17

R parietal cortex 18 -27 62

L parietal cortex -38 -27 60

L parietal cortex -24 -30 64

L poterior parietal cortex -41 -31 48

L temporal cortex -41 -37 16

L temporal cortex -53 -37 13

R superior parietal cortex 34 -39 65

Page 115: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

91

undirected graph G that has a network degree of E equal to the number of edges, and a

network density (cost) of D = E/[N x (N-1)]/2 representing the ratio of existing edges relative

to all possible edges (Bruno, Hosseini, & Kesler, 2012). It also identifies network fragment.

Network measures were computed over a range of connection densities. We examined those

graphs where all nodes were fully connected in the networks for both groups, which ensured

that network comparison between groups was meaningful with the same number of nodes in

each subnetwork, and where small-world properties were displayed for both groups. Network

fragment was observed at the density of 40%. Animal research has indicated that densities

above 50% are unlikely to be biological (Kaiser & Hilgetag, 2006). Therefore, the densities

ranging from 41% to 50% with an increment of 1% were investigated. Both HCs and patients

displayed small worldness properties where normalized characteristic path length is close to

one and normalized clustering coefficient is greater than one. Our interest was to detect

changes in nodal characteristics including hubness. Therefore, we investigated the graph

measures of local efficiency, as well as of nodal degree and BC (Bullmore & Sporns, 2009;

Rubinov & Sporns, 2010). Local efficiency measures the efficiency of local communication

of a given node, taking account for the number of the shortest paths between its neighbouring

nodes (Latora & Marchiori, 2001). It measures the ability of functional integration to

combine information across distributed brain regions. Nodal degree is defined as the total

number of connections that a node has with other nodes in the network, and BC is defined as

the fraction of all shortest paths in the network that pass through a given node. Bridging

nodes that connect disparate parts of the network often have high BC, making a potential hub

role of these nodes in the network (van den Heuvel & Hulshoff Pol, 2010) that interact with

many other regions and facilitate functional integration (Rubinov & Sporns, 2010).

4.2.6 Seed-based functional connectivity analysis

The only nodes showing group differences were further investigated using seed-based FC

analyses to elucidate changes in connectivity with other nodes in the network using the Conn

FC toolbox version 15a.

4.2.7 Statistical analysis

Page 116: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

92

Statistical analyses for demographic measures were performed using SPSS Statistics 20.0.0

(Chicago, IL, http://www-01.ibm.com/software/analytics/spss/). Statistical significance

threshold was set at P ≤ 0.05. Independent student’s t-test (two-tailed) was used to compare

the means in head motion parameters and demographic measures between patients and HCs.

Area under the curve (AUC) analyses was performed to test group differences in graph

measures using a two-tailed non-parametric permutation test with 1000 repetitions over the

density range without network fragmentation using GAT. The AUC analyses are less

sensitive for thresholding processes as they yield a summarized scalar independent of single

threshold selection. Statistical significance threshold was set at P ≤ 0.005.

As a hub is characterized both by high connectivity and centrality (Sporns, Honey, & Kotter,

2007), the nodes whose degree and BC were one standard deviation above the mean network

degree and BC were considered hubs. Spearman’s correlation analyses were also performed

between the only network measures that yielded significant group differences, and different

disease measures (UPDRS III total scores, rigidity, bradykinesia, and resting tremor sub-

scores, LEDD, MoCA and BDI II scores) using SPSS Statistics 20.0.0 (Chicago, IL,

http://www-01.ibm.com/software/analytics/spss/). Statistical significance threshold was set at

P ≤ 0.05. General linear model was used to obtain between-subjects contrasts of seed-based

FC analysis. Statistical significance threshold was set at P ≤ 0.005.

4.3 Results

4.3.1 Demographic and clinical characteristics

Table 4-2 shows the demographic and clinical characteristics of the patients and HCs. There

was a significant difference in MoCA (t = 3.216, P = 0.002) and BDI II scores (t = -4.27, P <

0.001) between the two groups. PD patients showed lower global cognitive performance and

higher depression level than HCs. There was no significant difference regarding age (t =

0.59, P = 0.56) or education (t = 0.18, P = 0.86) between HCs and patients. The LEDD was

calculated by Levodopa (mg/day) + Controlled Levodopa (Levodopa x 0.75 mg/day) +

Page 117: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

93

Entacapone/Comtan (Levodopa x 0.33 mg/day) + Praipexole/Mirapex (mg/day) +

Ropinirole/Requiq x 20 (mg/day) + Rasagilline/Azilect x 100 (mg/day) + Selegillne x 10

(mg) + Amantadine (mg/day) (Tomlinson et al., 2010).

Table 4-2 Demographic and clinical characteristics of patients with PD and HCs

Patients

(n = 42)

Healthy controls

(n = 23)

Age (years) 65.4 (6.5) 64.3 (8.3)

Sex (male/female) 31/11 10/13

Handedness (right/left) 34/8 23/0

Education (years) 16.0 (3.2) 16.1 (3.1)

MoCA 25.8 (2.9)* 27.5 (1.4)

BDI II 8.0 (4.6) ** 3.2 (3.4)

Disease duration (years) 5.9 (4.5) -

Symptom-dominant side (right/left) 26/16 -

UPDRS III

(on-medication)

27.5 (10.1) -

Rigidity 5.1 (2.2) -

Bradykinesia 1.9 (0.6) -

Resting tremor 1.3 (1.3) -

Hoehn and Yahr 2 (median) -

LEDD (mg/day) 644.4 (298.5) -

Data are presented in mean (standard deviation) unless otherwise indicated; * P = 0.002. **

P < 0.001.

Page 118: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

94

4.3.2 Head motion parameters and outliers

The mean of realignment parameters for head motion and outliers of patients and HCs are

presented in Table 4-3. Realignment parameters for head motion including x (t = 1.70, P =

0.1), y (t = 0.69, P = 0.49), z (t = 0.31, P = 0.76), pitch (t = 1.87, P = 0.07), roll (t = 0.06, P =

0.95), and yaw (t = 1.43, P = 0.16) as well as the total number of outliers (t = 0.040, P =

0.97) were not significantly different between HCs and patients.

Table 4-3 Head motion parameters and outliers of patients with PD and HCs

Patients

(n = 42)

Healthy controls

(n = 23)

Head motion parameters

Liner motion parameters (mm)

X 0.013 (0.005) 0.011 (0.005)

Y 0.018 (0.119) 0.016 (0.009)

Z 0.058 (0.035) 0.061 (0.036)

Rotational motion parameters (radians)

Pitch 0.0006 (0.0003) 0.0005 (0.0002)

Roll 0.0002 (0.0001) 0.0002 (0.0001)

Yaw 0.0003 (0.0001) 0.00002 (0.0001)

Outliers (# of volume) 7.6 (7.4) 7.7 (7.9)

Data are presented in mean (standard deviation).

Page 119: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

95

4.3.3 Group comparisons of graph measures

Compared to HCs, patients showed increased numbers of connections in the right and left

DLPFC of the FPN measured by nodal degree (P = 0.005 and P = 0.003, respectively)

(Figure 4-2; Table 4-4). In contrast, patients showed a reduction in intra-connectivity of the

right mid-insula (MIns) of the SMN measured by local efficiency (P = 0.005), as well as a

reduction in bridging role of the right pre-SMA of the SMN (P = 0.005) measured by nodal

BC.

We interrogated whether the graph measures of these four nodes would account for any of

the clinical measures (i.e., disease duration, UPDRS III scores as well as rigidity,

bradykinesia, and resting tremor subscores, Hoehn and Yahr, and LEDD, as well as MoCA

and BDI scores) by conducting Spearman’s correlation analyses. We found that BC of the

right pre-SMA showed a significant negative correlation with bradykinesia scores

(Spearman’s rho = -0.307, P = 0.048), suggesting that the diminished bridging role (i.e.

impaired integration) of the right pre-SMA may contribute to more severe bradykinesia.

We then applied the seed-based FC analyses to explore where these four affected areas

changed their connectivity. The results are summarized in Table 4-5. First, nodes in the right

DLPFC of the FPN showed increased intra-connectivity with a node in the homologous

anatomical region (t = 2.95, P = 0.002) and increased inter-connectivity with nodes of the

CON including the right MIns (t = 4.01, P < 0.001) and left PostIns (t= 3.3, P < 0.001).

Furthermore, the left DLPFC of the FPN showed increased intra-connectivity with a node in

the contralateral anatomical region (t = 2.68, P < 0.005). These increased regional intra- and

inter-connectivity may have occurred as a compensatory mechanism. Second, the right pre-

SMA of the SMN showed reduced inter-connectivity with nodes in the DMN including the

left angular gyrus (t = 3.29, P < 0.001) and left inferior temporal cortex (t = 2.81, P = 0.003),

suggesting its bridging and integrative role between cognitive and motor functions. The right

MIns did not show any significant changes in functional connectivity with other nodes.

Page 120: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

96

Figure 4-2. Dorsal view of brain image presenting four nodes showing group differences.

Increased degree in the bilateral DLPFC of FPN (fronto-parietal network), as well as

decreased between centrality in the right pre-SMA and decreased local efficiency in the right

mid-insula of SMN (sensorimotor network) in PD patients compared with healthy controls (P

≤ 0.005, uncorrected). The nodes are presented in equal size for visualization purposes. Brain

image and nodes are visualized using the BrainNet Viewer (NKLCNL, Beijing Normal

University). Bar graphs show the averaged values of each graph measure for HC (healthy

controls) and PD (patients with Parkinson’s disease). Error bars represent standard error of

the mean.

Page 121: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

97

Table 4-4 Graph measure changes in patients with Parkinson’s disease compared with HCs

# the MNI coordinates describe the center of node area.

Graph measure Node Subnetwork BA

Coordinate#

X Y Z

HCs < patients Degree Right DLPFC FPN 9 46 28 31

Left DLPFC FPN 9 -42 7 36

HCs > patients Local efficiency Right MIns SMN - 33 -12 16

BC Right pre-SMA SMN 6 10 5 51

Page 122: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

98

Table 4-5. FC of three nodes that showed group differences.

Graph

measur

e

Seed

region

Subnetwork Coordinate# Target region Subnetwork Coordinate# T (63) P

(uncorrecte

d) X Y Z X Y Z

HCs <

Patients

Degree Right

DLPFC

(BA9)

FPN 46 28 3

1

Right MIns CON 32 -12 2 4.01 < 0.001

Left PostIns CON -30 -28 9 3.23 < 0.001

Right DLPFC

(BA9)

FPN 40 17 40 2.95 0.002

Left putamen CON -20 6 7 2.7 0.004

Left

DLPFC

(BA9)

FPN -42 7 3

6

Right DLPFC

(BA9)

FPN 40 17 40 2.68 <0.005

HCs >

Patients

BC Right pre-

SMA

(BA6)

SMN 10 5 5

1

Left angular

gyrus (BA40)

DMN -48 -63 35 3.29 < 0.001

Left inferior

temporal cortex

(BA20)

DMN -59 -25 -15 2.81 0.003

# the MNI coordinates describe the center of node area.

Page 123: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

99

4.3.4 Hub analysis

Ten hub nodes, their affiliated networks, and MNI coordinates of both patients and HCs are

presented in Figure 4-3 and Table 4-5. The disease affected hub organization: Patients

presented eight regions acting as hubs while HCs presented nine regions. Seven of these

regions were shared by both patients and HCs, which included the right ACC and left

precuneus of the DMN, the left ACC of the FPN, as well as the left ACC, left thalamus, and

two nodes in the right thalamus of the CON. The patients lost hub properties in the left AIns

of the CON and the left MIns of the SMN, while the CN of the CON acted as a new hub in

the patients. We explored whether any of these three hub regions were associated with the

clinical measures (i.e., disease duration, UPDRS III scores as well as rigidity, bradykinesia,

and resting tremor scores, Hoehn and Yahr, and LEDD, as well as MoCA and BDI scores) in

PD patients. We found a significant positive correlation between hubness (i.e., the mean of

degree and BC) in the left MIns of the SMN and dopaminergic medication (Spearman’s rho

= 0.303, P = 0.05), suggesting that dopaminergic medication may influence hubness in the

MIns in these patients.

Page 124: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

100

Figure 4-3 Caudal view of brain image presenting 10 hub nodes. ACC: anterior cingulate cortex; AIns: anterior insula; CN: caudate

nucleus; CON: cingulo-opercular network; DMN: default mode network; FPN: fronto-parietal network; MIns: mid insula. Seven hubs

in red represent common hubs to both healthy controls and patients with Parkinson’s disease. Two in yellow represent hubs identified

only in healthy controls and one in green, only in patients. Brain image and hubs are visualized using the BrainNet Viewer (NKLCNL,

Beijing Normal University).

ACC (FPN)

ACC (CON)

Precuneus (DMN) ACC (DMN)

Thalamus

(CON)

CN (CON) AIns (CON)

L R

MIns (SMN)

Hub – Common

Hub – HCs

Hub – Patients with PD

Page 125: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

101

Table 4-6. Hub regions in HCs and patients with PD.

# The MNI coordinates describe the center of node area.

Hub region Subnetwork Coordinate#

X Y Z

Common Right ACC DMN 9 39 20

Left precuneus DMN -3 -38 45

Left ACC FPN -1 28 40

Left ACC CON -2 30 27

Left thalamus CON -12 -12 6

Right thalamus CON 11 -12 6

Right thalamus CON 11 -24 2

HC only Left AIns CON -36 18 2

Left MIns SMN -36 -12 15

Patient only Right CN CON 14 6 7

Page 126: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

102

4.4 Discussion

In the current study, we aimed to investigate functional changes in the SMN and cognitive

networks in PD with a particular focus on changes in the disease-associated nodes and hubs

using graph theoretical analyses. In general, patients showed weakened connectivity in nodes

of the SMN and enhanced connectivity in nodes associated with cognitive networks. Patients

showed functional changes in the right pre-SMA and right MIns, as well as the bilateral

DLPFC (BA9) compared with HCs. PD also affected hub functions and organization. In

particular, the left AIns of the CON and left MIns of the SMN lost their hub properties and a

node in the CN was identified as a new hub in these patients.

4.4.1 Changes in the sensorimotor network

As hypothesized, we found functional changes in the nodes of the SMN. More specifically,

patients showed a reduction in intra-connectivity of the right MIns and inter-connectivity of

the right pre-SMA with the nodes of the DMN. Furthermore, the diminished inter-

connectivity of the right pre-SMA was associated with more severe bradykinesia. In addition,

the left MIns lost its hub properties in the patients and showed a positive interaction with

dopaminergic medication.

Reduced FC in the SMA including the pre-SMA was consistently demonstrated in PD

patients compared to HCs using the graph theoretical approaches. (Nagano-Saito et al., 2014;

Sang, et al., 2015; Skidmore et al., 2011; Tinaz et al., 2016; Wei et al., 2014; Wu et al.,

2009). For example, nodal degree, efficiency, and BC were all reduced in the SMA in PD

patients (Sang, et al., 2015). Reduced connectivity of the SMA was also associated with

more severe motor symptoms (Cao et al., 2011; Wu, et al., 2009).

The pre-SMA is the anterior part of the SMA and is separated from the SMA proper by the

vertical anterior commissural line (Picard & Strick, 2001). These two areas are different in

terms of anatomical connections and functions. The SMA proper is connected to the M1,

projected to the spinal cord and is involved in movement generation, while the pre-SMA has

extensive connections with the PFC (Wang, Shima, Sawamura, & Tanji, 2001) and is

Page 127: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

103

involved in cognition (Picard & Strick, 2001). For example, the pre-SMA has been

implicated to play an important role in cognitive motor control involving integration of

sensory information and decision-making about actions or motor selection (Ikeda et al.,

1999).

It is not surprising that the decreased interconnectivity of the pre-SMA with a cognitive

subnetwork can account for the slowness of movement, as it is postulated that bradykinesia

could arise from slowness in formulating the instructions to move before the onset of and

during the actions (Berardelli, Rothwell, Thompson, & Hallett, 2001). In the present study,

the weakened connectivity between the right pre-SMA and the nodes of the DMN, in the

context of DA deficiency, may affect the redirection of attentional processes from self-

reflection (i.e., internal reference) to goal-directed behavior, thus contributing to

bradykinesia.

4.4.2 Changes in the cognitive networks

We also found functional changes in the DLPFC (BA9) associated with the FPN and the left

AIns of the CON in patients compared with HCs. Patients showed an increased level of

connectivity in bilateral DLPFC (BA9). The subsequent FC analysis revealed that the

enhanced connectivity occurred within and between cognitive networks (i.e., FPN and

CON). The hub analysis revealed that while the left AIns lost its hub properties, a new hub

region was identified in the striatal region (i.e. CN) associated with the same network (i.e.

CON) in PD patients.

Literature showed mixed results in FC associated with the fronto-parietal network and the

DLPFC in PD patients compared with HC subjects using graph theoretical analysis. In line

with our findings, increased degree of DLPFC (Cao et al., 2011) and increased FC in the

fronto-parietal module (Baggio et al., 2014) were reported in PD patients. On the other hand,

reduced FC in the DLPFC (Nagano-Saito et al., 2014) and in the fronto-parietal regions

(Fogelson et al., 2013) was also reported. The discrepancy in these findings may be in part

due to capturing different mechanisms in response to the disease process in this anatomical

region (i.e., compensatory or pathological mechanism).

Page 128: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

104

Increased FC of task-related regions (i.e. DLPFC) is often interpreted as a compensatory

mechanism (Grady, 2012) when cognitive/behavioural performance starts failing. Our

patients had lower MoCA scores compared with HCs, suggesting that the enhanced

connectivity and the new hub of the CN may be parallel “attempted compensation” (Grady,

2012) for trying to maintain an efficient utilization of neural resources (Grady, 2008), in the

context of DA deficiency. In fact, AIns of CON lost its hub properties in our PD patients.

The insular cortex is associated with dopaminergic dysfunction PD (Christopher, Duff-

Canning et al., 2014; Christopher et al., 2015) and deposition of aggregated α-synuclein

(Braak et al., 2005). Dopamine dysfunction in the AIns in particular was associated with

poor executive and memory impairment in PD patients (Christopher, Duff-Canning et al.,

2014; Christopher et al., 2015). The right DLPFC enhanced FC with MIns and PostIns of the

CON. Thus, enhanced FC in the DLPFC and CN might be counteracting the reduced or

reducing hubness of the AIns. It may also be explained by pathological cortical disinhibition

(de Haan, Mott, van Straaten, Shceltens, Stam, 2012). Another possible explanation is that

the brain changes may have resulted from the effects of dopaminergic medication although

there was no association between the changes and LEDD.

4.4.3 Hub reorganization

Hub nodes are characterized with high connectivity and centrality (Sporns et al., 2007).

Therefore, we used both BC and degree to identify hub regions. Our HCs showed nine hub

regions: two DMN nodes including ACC and precuneus, one FPN node - ACC, five CON

nodes in the regions of ACC, thalamus, and AIns, and one SMN node – MIns. As we

hypothesized, PD patients showed hub reorganization characterized by the loss of hub

properties in the insular regions and an emergence of a new hub in the CN. Consistent with

the literature, our findings further support that (1) the insular cortex is an important

integrative node in brain network communication (Menon & Uddin, 2010), and (2) it is

affected by PD, possibly due to aggregated alpha synuclein (Braak et al., 2005) and

dopamine dysfunction (Christopher, Duff-Canning et al., 2014; Christopher et al., 2015).

To date, there were two studies investigated hub regions in PD patients and HCs using

rsfMRI data (Baggio et al., 2014; Sang et al., 2015). One study identified 18 nodes as hub

regions that showed the 20% highest scoring of the sum of clustering coefficient and BC

Page 129: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

105

(Baggio et al., 2014). Consistent with our data, the precuneus was one of the hub regions in

the DMN in both HCs and PD patients. This study also identified the medial SFG node as a

hub in HCs and PD-MCI patients; SFG in HCs and PD patients; and middle SFC in non-MCI

PD patients. The other study used BC and tested the values against the null hypothesis using

a non-parametric one-tail test across different thresholds and found 13 hubs in both PD

patients and HCs (Sang et al., 2015). There was no consistency in hub regions in HCs

between this study and our study while this study also identified the CN as a hub in PD

patients.

In addition to the functional hubs, two studies investigated structural hubs using cortical

thickness and subcortical volume (Pereira et al., 2015) and using DTI tractography (Nigro et

al., 2016). The first study employed the same classification to identify hub regions as our

study (i.e, one standard deviation higher than the mean of both degree and BC) (Pereira et al.,

2015). It identified 19 hubs in HCs, 13 hubs in non-MCI PD patients, and 16 hubs in PD-

MCI patients. The hubs identified across the three groups were located in distributed cortical

regions, primarily in the frontal, temporal and parietal regions. The nodes in the DLPFC

region were also identified as hubs in both HCs and non-MCI PD patients. Consistent with

our data, the precuneus was identified as a hub region in both HC and two PD groups.

Interestingly, only PD-MCI patients showed the insula region as a hub region. The second

study used four different graph measures to score each node from 0 to 4. If the node showed

the highest 20 % of one of these four graph measures, it gets one score and if the node had a

score of 2 or higher, it was identified as a hub. PD patients and HCs shared 14 hubs, which

were located in the cortical regions except the occipital region. The precuneus, DLPFC

region and insula were identified as hub regions in both groups, and the paracentral lobule

was identified as a hub region only in HCs.

Some of these studies identified the DLPFC and SMA regions as hubs in HCs, whose

function was altered in PD patients, which we hypothesized, but we did not find in our data.

The discrepancy in hub regions between our study and the previous studies largely arises

from the definition of hub regions and types of nodes (e.g., functional nodes derived from

tasks used in our study or anatomically parceled nodes used in all other studies) investigated.

Other possible factors contributing to the differences include types of MRI data (e.g.,

Page 130: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

106

structural or function), the number of nodes investigated, as well as processing and analysis

methods.

4.4.4 Conclusions

Using graph theoretical approach, we further characterized PD-related functional changes

and highlight the diffuse changes in the nodal organization along with the regional hub

disruption, which accounts for the distributed abnormalities across brain networks and

clinical manifestations of PD. PD patient showed both increased and decreased FC, which

occurred within the same network and across different networks. The decreased FC found in

the pre-SMA of the SMN is most likely pathological changes evidenced by its association

with severe motor symptom. Increased FC and a new hub in cognitive networks such as FPN

and CON may be adaptive and counteracting the influence of neurotransmitter dysfunction

and deposition of α-synuclein associated with cognitive function. Lastly, the insula plays an

important integrative role in brain network communication, susceptible to PD pathology.

Page 131: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

107

5.0 Study 3: Imaging striatal microglial activation in

patients with Parkinson’s disease

This study was published in PLoSOne.

This chapter has been modified from the following:

Yuko Koshimori, Ji-Hyun Ko, Romina Mizrahi, Pablo Rusjan, Rostom Mabrouk, Mark F.

Jacobs, … Antonio P. Strafella (2015). Imaging Striatal Microglial Activation in Patients

with Parkinson's Disease. PLoSOne, 10(9), e0138721.

5.1 Introduction

Neuroinflammation is considered to play an important role in the progression of Parkinson’s

PD. Neuroinflammation in PD was first evidenced in a postmortem study where activated

microglia were found in the SN (McGeer et al., 1988). A subsequent postmortem study

further identified activated microglia in the extended brain areas such as putamen,

hippocampus, as well as trans-entorhinal, cingulate, and temporal cortices (Imamura et al.,

2003). Neuroinflammatory processes were also confirmed by increased concentration of

inflammatory cytokines such TNF-α and IL-1β and IL-6 in the striatum at postmortem (Mogi

et al., 1994) as well as in vivo studies using the serum (Dobbs et al., 1999) and cerebrospinal

fluid (Blum-Degen et al., 1995) of PD patients.

TSPO has been studied as a potential in vivo biomarker of reactive gliosis and inflammation

associated with a variety of neuropathological conditions (Chen & Guilarte, 2008). TSPO is

located in the outer mitochondrial membrane of glial cells. While TSPO levels are very low

in healthy brains, they markedly increase co-localizing activated microglia in brains affected

by various diseases such as amyotrophic lateral sclerosis, AD, frontotemporal dementia and

MS (Venneti et al., 2013). This elevated TSPO expression was primarily quantified using

[11C]-PK11195 PET, the first and most commonly used TSPO radioligand.

Page 132: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

108

To date, a few studies have investigated neuroinflammation in PD patients using [11C]-

PK11195 PET. However, the findings have been inconclusive. While some studies have

found elevated TSPO binding in the nigro-striatal regions (Ouchi et al., 2005), others did not

support these observations (Kobylecki et al., 2013). Additionally, an association between

elevated TSPO binding in the nigro-striatal regions and clinical measures has been

demonstrated in some studies (Ouchi et al., 2005) but not in others (Kobylecki et al., 2013).

One possible explanation for these inconsistent findings may be due to the limitations

inherent to this prototypical radioligand ([11C]-PK11195) such as low signal-to-noise ratio,

high non-specific binding, low brain penetration, and high plasma protein binding. These

limitations as well as its limited dissemination for wider clinical use due to the short-lived

carbon-11 labeling have prompted the development of new TSPO radioligands.

[18F]-FEPPA is a new, second-generation TSPO radioligand that has shown a high affinity

for TSPO, an appropriate metabolic profile, high brain penetration and good

pharmacokinetics (Wilson et al., 2008). Its quantification has been validated using the 2TCM

with VT as reliable outcome measure (Rusjan et al., 2011). [18F]-FEPPA demonstrated

superior specificity to the affected side of the striatum and stronger correlation with

inflammatory cytokines compared with [11C]-PK11195 using an animal model of PD

(Hatano et al., 2010). Further, our group recently demonstrated a three-fold higher [18F]-

FEPPA VT in a tumor site compared to the healthy contralateral area in an individual subject

(Ko et al., 2013), as well as a significant increase in individuals with MDE (Setiawan et al.,

2015) and patients with AD (Suridjan et al., 2015).

Second-generation TSPO radioligands have been known to present three patterns of binding

affinity based on genetic polymorphism: HABs, MABs, and LABs (Owen et al., 2012).

These different phenotype patterns can be predicted by a single-nucleotide polymorphism

(SNP), rs6971 located in the exon 4 of the TSPO gene resulting in a nonconservative amino-

acid substitution at position 147 from alanine to threonine (Ala147Thr) in the fifth

transmembrane domain of the TSPO protein. This polymorphism accounts for some of the

large inter-individual variability in the outcome measures (Owen et al., 2012). Therefore,

accounting for this polymorphism in the statistical analyses will likely increase sensitivity in

detecting neuropathological changes (Mizrahi et al., 2012).

Page 133: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

109

In the present study, we investigated for the first time whether TSPO imaging with [18F]-

FEPPA, could be used as a potential biomarker for neuroinflammation in PD. Using the

striatum as our main ROI, we sought to determine whether (1) the polymorphisms would

reflect on TSPO binding affinity also in PD patients; (2) [18F]-FEPPA VT could differentiate

the PD group from the HC group; and (3) the levels of TSPO binding would correlate with

clinical measures of PD.

5.2 Methods

5.2.1 Subjects

Nineteen patients meeting UK Brain Bank criteria for the diagnosis of idiopathic PD and 17

HC subjects participated in the study. Exclusion criteria for all participants included (1)

history of a head injury, psychiatric or neurological (except PD for the patients) diseases, (2)

alcohol or drug dependency or abuse, (3) contraindications for MRI scanning, and (4) use of

nonsteroidal anti-inflammatory drugs (NSAID). PD patients were assessed for their cognitive

ability using the MoCA, the level of depression using the BDI II, as well as for motor

severity of the disease using the motor subset of the UPDRS-III. All participants underwent

PET and structural MRI scans. All participants provided written informed consent following

full explanation of the study procedures. The study was approved by the CAMH Research

Ethics Board and the University Health Network Research Ethics Board, and conformed to

the Declaration of Helsinki.

5.2.2 PET data acquisition

The synthesis of [18F]-FEPPA has been described in detail elsewhere (Wilson et al., 2008). It

can be reliably and quickly labeled with [18F] by nucleophilic displacement of a tosylate,

leaving group in a fast one-step reaction yielding a sterile and pyrogen-free product after

purification and formulation.

Page 134: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

110

The PET images were obtained using a 3D High Resolution Research Tomograph (HRRT)

(CPS/Siemens, Knoxville, TN, USA), which measures radioactivity in 207 slices with an

inter-slice distance of 1.22 mm. A custom-fitted thermoplastic mask was made for each

subject and used with a head fixation system during the PET scans to minimize head

movement. Following a transmission scan, intravenous [18F]-FEPPA was administered as a

bolus. The mean injected amount, specific activity at the time of injection and mass injected

of all the participants were 4.926 ± 0.265 (mCi), 3127.723 ± 2026.481 (mCi/μmol), and 0.9 ±

0.676 (μg), respectively. The scan duration was 125 min. The images were reconstructed into

34 time frames: 1 frame of variable length until the radioactivity appears in the FOV, 5

frames of 30s, 1 frame of 45s, 2 frames of 60s, 1 frame of 90s, 1 frame of 120s, 1 frame of

210s, and 22 frames of 300s.

All PET images were corrected for attenuation using a single photon point source, 137Cs

(T50 = 30.2 years, Eγ = 662 keV) and were reconstructed by filtered back projection

algorithm using a HANN filter at Nyquist cutoff frequency. The reconstructed image has 256

× 256 × 207 cubic voxels measuring 1.22 × 1.22 × 1.22 mm and the resulting reconstructed

resolution is close to isotropic 4.4 mm, FWHM in plane and 4.5 mm FWHM axially,

averaged over measurements from the center of the transaxial FOV to 10 cm off-center in 1.0

cm increments. In addition, for frame realignment for head motion correction, each image

was reconstructed without attenuation correction using three iterations of iterative

reconstruction (Rusjan et al., 2011).

5.2.3 MRI acquisition

MRI images for all the subjects were acquired for co-registration with the corresponding

PET images and the anatomical delineation of the ROIs (i.e., CN and putamen). Proton

density weighted MR images were chosen for better identification of the striatum (Rusjan et

al., 2006). Two dimensional (2D) oblique proton density-weighted MR images were acquired

with a GE Discovery 3.0 T MRI scanner (slice thickness = 2 mm, TR = 6000 ms, TE = Min

Full, flip angle = 90°, number of excitations (NEX) = 2, acquisition matrix = 256 × 192, and

FOV = 22 cm).

Page 135: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

111

5.2.4 Input function measurement

Dispersion- and metabolite-corrected plasma input function was generated as previously

described (Rusjan et al., 2011). Briefly, arterial blood was taken continuously at a rate 2.5

mL/min for the first 22.5 minutes after radioligand injection and the blood radioactivity

levels were measured using an automatic blood sampling system (Model # PBS-101 from

Veenstra Instruments, Joure, The Netherlands). In addition, 4 to 8 ml manual arterial blood

samples were obtained at 2.5, 7, 12, 15, 30, 45, 60, 90, and 120 min relative to time of

injection (Rusjan et al., 2011). A bi-exponential function was used to fit the blood-to-plasma

ratios. A hill function was used to fit the percentage of unmetabolized radioligand. The

dispersion effect was modeled as to the convolution with a mono-exponential with dispersion

coefficient of 16 seconds and corrected with iterative deconvolution (Rusjan et al., 2014).

5.2.5 Generation of ROI-based time activity curve

[18F]-FEPPA PET images were preprocessed and ROIs were automatically generated using

in-house software, ROMI (Rusjan et al., 2011). Briefly, ROMI fits a standard template of

ROIs to an individual PD-weighted MR image based on the probability of GM, WM, and

CSF. The individual MR images are then co-registered to each summed [18F]-FEPPA PET

image using the normalized mutual information algorithm so that individual refined ROI

template can be transferred to the PET image space to generate the time activity curve (TAC)

for each ROI. Our a priori ROIs included the CN and putamen, which are disease-affected

regions and whose quantification was validated (Rusjan et al., 2011). Dynamical series of

images of [18F]-FEPPA PET were visually checked for head motion and corrected using

frame-by-frame realignment. Low noise, nonattenuation-corrected images (created with

iterative reconstruction) were used to optimize the frame-by-frame realignment process. A

normalized mutual information algorithm was applied with SPM8 (Wellcome Trust Centre

for Neuroimaging, London, UK) to co-register each frame to the frame that showed a high

signal-to-noise ratio. Parameters from the normalized mutual information were applied to the

corresponding attenuation-corrected dynamic images to generate a movement-corrected

dynamic image.

Page 136: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

112

To address the potential issues of bias from the volume loss in the older subjects, time

activity data for all subjects was corrected for the effect of partial volume error (PVE) using

the Mueller-Gartner partial volume error correction (PVEC) algorithm as implemented in

Bencherif, Stumpf, Links, & Frost (2004).

5.2.6 Kinetic analysis

VT values in each ROI were derived from a 2TCM using [18F]-FEPPA radioactivity in

arterial plasma as an input function and a 5% vascular contribution (Rusjan et al., 2011). VT

is a ratio at equilibrium of the radioligand concentration in tissue to that in plasma (i.e.

specific binding and non-displaceable uptake including non-specifically bound and free

radioligand in tissue) and can be expressed in terms of kinetic rate parameters as: VT = K1 /

k2 (1 + k3 / k4) where K1 and k2 are influx and efflux rates for radiotracer passage across the

blood brain barrier and k3 and k4 describe the radioligand transfer between the free and non-

specific compartments and the specific binding compartment. We also measured %COV

(100% x standard error/mean), where standard error was estimated from the diagonal of the

covariance matrix of nonlinear least-squares fitting (Rusjan et al., 2011). From the different

ROIs, we included VT with %COV of ≤ 20, which assured less data noise.

5.2.7 DNA extraction and polymorphism genotyping

Genomic DNA was obtained from peripheral leukocytes using high salt extraction methods

(Lahiri & Nurnberger, 1991). The polymorphism rs6971 was genotyped variously using a

TaqMan® assay on demand C_2512465_20 (AppliedBiosystems, CA, USA). The allele

T147 was linked to Vic and the allele A147 was linked FAM. Polymerase chain reactions

(PCR) were performed in a 96-well microtiter-plate on a GeneAmp PCR System 9700

(Applied Biosystems, CA, USA). After PCR amplification, end point plate read and allele

calling was performed using an ABI 7900 HT (Applied Biosystems, CA, USA) and the

corresponding SDS software (v2.2.2). Individuals with genotype Ala147/Ala147 were

classified as HABs, Ala147/Thr147 as MABs, and Thr147/Thr147 as LABs (Owen et al.,

2012).

Page 137: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

113

5.2.8 Statistical analysis

Demographic and clinical measures were compared using factorial ANOVA, independent,

two-tailed student t tests, or Fisher’s exact tests. Group differences in VT values were

analyzed using factorial ANOVA with TSPO genotype and disease as fixed factors in the CN

and the putamen. A second level of analysis with student t tests were performed among four

groups: (1) between HC with MAB (HC-MAB) and HC with HAB (HC-HAB), (2) between

PD with MAB (PD-MAB) and PD with HAB (PD-HAB), (3) between HC-MAB and PD-

MAB, and (4) between HC-HAB and PD-HAB. Correlations were investigated between

clinical measures and VT values in the ROIs using Pearson’s correlation tests. All of the

statistical analyses were performed using SPSS Statistics version 20.0. The threshold for

significance was set at P < 0.05.

5.3 Results

5.3.1 Demographic and clinical characteristics

Based on the rs6971 polymorphism, there were 16 MABs consisting of 8 HC subjects and 8

PD patients, 16 HABs consisting of 8 HC subjects and 8 PD patients, as well as 4 LABs

consisting of 3 PD patients and 1 HC subject. [18F]-FEPPA signals are too low to be

quantifiable in these LABs and for this reason these subjects were not included in the

analyses. In general, LAB subjects in a Caucasian sample represent less than 5% (Mizrahi et

al., 2012), and only very few are identified in any given study. The demographic and clinical

characteristics of HC subjects and PD patients included in the analyses were presented in

Table 5-1 and 5-2.

The factorial ANOVA showed that there were no significant differences in age (F(3, 28) =

0.86, P = 0.47), as well as in the [18F]-FEPPA injected amount (F (3, 28) = 1.30, P = 0.30),

specific activity at the time of injection (F (3, 28) = 0.94, P = 0.44) or mass injected (F (3,

28) = 1.68, P = 0.19). Chi-square analysis showed that there was no significant difference in

the composition of gender (χ2 = 3.14, P = 0.08). In addition, the PD-MAB and PD-HAB

Page 138: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

114

groups were not significantly different in years of education (t(14) = 0.50, P = 0.63), MoCA

scores (t(14) = 0.82, P = 0.43), BDI scores (t(14)= 0.65, P = 0.53), UPDS III (t(14) = 0.88, P

= 0.40), duration of disease (t(14) = 0.18, P = 0.87), total LEDD (t(14) = 0.03, P = 0.98) or

the composition of symptom dominant side (Fisher’s exact tests, P = 0.61).

Table 5-1. Demographic and clinical characteristics and PET measures of HC subjects and

PD patients

HC

(n = 16)

PD

(n = 16)

Age 62.1 (8.7) 64.3 (9.0)

Gender (M:F) 6:10 11:5

Genotype (HAB:MAB) 8:8 8:8

Injected amount (mCi) 4.85 (0.26) 5.00 (0.26)

Specific activity

at the time of injection (mCi/µmol)

3006.91 (2435.27) 3248.54 (1589.13)

mass injected (µg) 1.05 (0.83) 0.75 (0.45)

Data was presented in mean (standard deviation).

Table 5-2. Demographic and clinical characteristics of PD patients

PD-MAB

(n = 8)

PD-HAB

(n = 8)

Education (years) 15.5 (3.8) 16.4 (3.2)

MoCA 27.4 (2.3) 26.5 (1.9)

BDI 6.4 (4.1) 7.6 (4.3)

Symptom dominant side (R:L) 6:2 4:4

UPDRS III 28.4 (9.6) 25.0 (5.1)

Duration of disease (years) 5.6 (2.1) 5.9 (4.7)

Total LEDDa (mg/day) 623.8 (596.4) 525.1 (267.6)

Data was presented in mean (standard deviation). aLEDD: Levodopa dose + 1-Levodopa-CR

x 0.75 + pramipexole (mg) x 67 (Evans et al., 2004).

5.3.2 Genotype and disease effects on TSPO binding

Page 139: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

115

Levene’s tests of equality of error variances were not significant in either CN (P = 0.449) or

putamen (P = 0.284). There was a significant main effect of genotype on VT values in the CN

(F(1, 28) = 14.62, P = 0.001) and in the putamen (F(1, 28) = 27.15, P < 0.001). There was no

main effect of disease in the CN (F(1, 28) = 0.37, P = 0.55) or in the putamen (F(1, 28) =

1.27, P = 0.269). There was also no disease x genotype interaction in the CN (F(1, 28) =

1.38, P = 0.25) or in the putamen (F(1, 28) = 0.86, P = 0.362).

Using two-tailed student t tests, when comparing the effect of genotype in HC groups, HC-

HAB group showed significantly higher VT values in in the putamen (10.05 ± 0.97; t(14) =

3.04, P = 0.009), but not in the CN (9.29 ± 0.94; t(14) = 1.86, P = 0.084) compared with HC-

MAB group (putamen: 6.31 ± 0.76; CN: 6.95 ± 0.91).

The effect of genotype in PD groups showed that PD-HAB had significantly higher VT

values in both putamen (11.84 ± 1.12; t(14) = 4.30, P = 0.001) and CN (11.02 ± 1.14; t(14) =

3.56, P = 0.003) and compared with PD-MAB (putamen: 6.49 ± 0.52; CN: 6.43 ± 0.61;)

(Figure 5-1A).

When testing the effect of the disease, neither PD-MAB (putamen: 6.49 ± 0.52; CN: 6.43 ±

0.61) nor PD-HAB (putamen: 11.84 ± 1.12; CN: 11.02 ± 1.14) group showed significant

increase in VT values in any ROI compared with HC-MAB (putamen: 6.31 ± 0.76; CN: 6.95

± 0.91) and HC-HAB (putamen: 10.05 ± 0.97; CN: 9.39 ± 0.94) groups, respectively (Figure

5-1B). The percentage differences of the mean VT values between PD-HAB and HC-HAB

groups were 16% in both putamen and CN, while they were 3% and -8%, respectively in the

MAB groups.

There was no correlation between LEDD, UPDRS scores or duration of disease with the VT

values in the striatum in either PD-HAB, PD-MAB, or PD groups, thus excluding any role of

these variables in the VT findings.

We conducted as well an additional analysis on the data without PVEC to investigate

whether this played any role in the results. The analysis confirmed the main effect of

genotype on VT values (Figure 5-2A), but no main effect of disease (Figure 5-2B), or disease

x genotype interaction.

Page 140: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

116

Figure 5.1. Graphs of PVEC VT in the CN and in the putamen. A. HC-MAB and HC-HAB

groups as well as PD-MAB and PD-HAB groups. Asterisks indicate that the HAB groups

show significantly higher VT mean values compared with the MAB groups (** P < 0.01). B.

HC-MAB and PD-MAB groups as well as HC-HAB and PD-HAB groups. In the HAB

group, the percentage difference between PD and HC was 16% in both CN and putamen, and

in the MAB group, it was -8% and 3%, respectively. Reproduced with permission from

(Koshimori, Ko et al., 2015).

Figure 5.2. Graphs of VT in the CN and in the putamen. A. HC-MAB and HC-HAB groups

as well as PD-MAB and PD-HAB groups. Asterisks indicate that the HAB groups show

significantly higher VT mean values compared with the MAB groups (**P < 0.01). B. HC-

MAB and PD-MAB groups as well as HC-HAB and PD-HAB groups. In the HAB group, the

percentage difference between PD and HC was 11% in the CN and 14% in the putamen, and

in the MAB group, it was -3% and 5%, respectively. Reproduced with permission from

Page 141: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

117

(Koshimori, Ko et al., 2015)

5.4 Discussion

This is the first study to investigate the potential use of [18F]-FEPPA as a new radioligand to

measure neuroinflammation in PD patients. As already observed in normal controls, we

found that in PD patients as well, the rs6971 polymorphism influenced TSPO binding

affinity. Although, we did not find a significant disease effect on TSPO expression in the

striatum, an interesting observation was a trend towards elevated TSPO binding in the PD-

HAB group (16% in both CN and putamen), but not in the PD-MAB group. These

observations imply that while the genotype of the rs6791 polymorphism certainly plays a

role in TSPO expression also in PD, a larger sample size may be needed to investigate the

interactions between rs6791polymorphism and neuroinflammation in PD patients, although

other studies with similar sample size did show a significant increase in [18F]-FEPPA VT in

individuals with MDE (Setiawan et al., 2015) and patients with AD (Suridjan et al., 2015).

In those studies, such differences in TSPO expression were reported mainly in the HAB

groups. In particular in AD patients (Suridjan et al., 2015), elevated microglia was detected

throughout the brain, in both GM and WM suggesting that neuroinflammation may certainly

play a role in the cognitive decline.

The same dissociation and potential role of the rs6971 polymorphism on TSPO binding has

also been suggested in studies using other second-generation TSPO radioligands. For

example using [3H]PBR28, postmortem brains of patients with SCZ classified as HABs

showed significantly greater specific binding in the DLPFC while those categorized as

MABs did not (Kreisl et al., 2013). Similarly using [18F]-FEMPA in AD, the disease effect

on TSPO expression was stronger in different cerebral regions in HAB patients (Varrone et

al., 2015). On the other hand, cocaine abusers did not seem to demonstrate the same

dissociation (Narendran et al., 2014).

The rs6791 polymorphism can be associated with the susceptibility, progression or disease

protection (Rone et al., 2012). For instance, healthy individuals with Ala147/Ala147 (i.e.,

HABs) have been associated with significantly higher level of low-density lipoprotein (LDL)

Page 142: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

118

than those with Ala147/Thr147 (i.e., MABs) and with Thr147/Thr147 (i.e., LABs) (Costa et

al., 2009), and LDL has been associated with the risk of PD (Huang et al., 2015). While these

reports are highly suggestive, the clinical significance of this polymorphism in PD is yet to

be determined. Similarly, despite the well-documented increase in the TSPO expression as a

result of brain insult in ex-vivo brain tissue, the functional significance of the upregulated

TSPO is still unclear (Chen & Guilarte, 2008). One possibility is that this upregulation may

be related to glial proliferation, migration, and phagocytosis (Streit, Sammons, Kuhns, &

Sparks, 2004) or secretion of inflammatory cytokines. [11C]-PK11195 reduced the expression

of proinflammatory cytokines in cultured human microglia (Choi et al., 2002). Furthermore,

increased TSPO levels in microglia and astrocytes may possibly increase neurosteroid

synthesis at injury sites to promote neurotropic and neuroprotective activity (Schumacher et

al., 2000). While the observations regarding the potential role of the polymorphism on TSPO

binding may be quite intriguing, they are speculative and warrant further investigations.

We investigated microglial activation only in the CN and putamen as a first evaluation study

of a second generation TSPO radioligand, [18F]-FEPPA, and did not find a significant disease

effect on [18F]-FEPPA VT in the striatum. We selected these two regions because they are

most affected by dopaminergic dysfunction in PD and [18F]-FEPPA VT is quantifiable while

the SN is also severely affected in PD by DA neurons undergoing degeneration, and is most

studied and evidenced for microglial activation in the postmortem PD brains, but it is hard to

accurately quantify the VT value of the SN due to its low and unstable identifiability.

Postmortem and neuroimaging studies demonstrated evidence of elevated neuroiflammatory

processes in the striatal region although they yielded mixed results. In one study, the tissues

in the putamen from 12 PD brains showed significantly higher number of ramified microglia

compared with those from 4 HC brains (Imamura et al., 2003). On the other hand, two

postmortem studies showed no activated microglia in the putamen, comparing five PD brains

with five HC brains (Mira et al., 2000) and comparing brains of nine PD, six incidental LBD,

four progressive supranuclear palsy, and 10 HCs (Bradaric et al., 2012) although these two

studies found significantly higher level of microglial activation in the SN in PD brains. To

our knowledge, the CN has not been investigated for microglial activation in postmortem PD

brains. However, there is evidence for significantly increased reactive astrocytes in the

Page 143: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

119

region in postmortem PD brains (Mythri et al., 2011). The expression of GFAP in the CN of

five PD brains was approximately two fold higher than that of six HC brains. In addition,

GFAP immunoreactive astrocytes were reported in the striatum of 12 PD brains although the

immunoactivity varied from slight in seven cases to moderate in five cases while two cases

showed no detectable immunoactivity (Braak et al., 2007).

Similarly, some of the neuroimaging studies using [11C]-PK11195 found microglial

activation in the putamen (Iannacone et al., 2012; Ouchi et al., 2009) and striatum (Gerhard

et al., 2006) while others did not in the striatum (Bartel et al., 2010; Edison et al., 2013;

Kobylecki et al., 2013).

We did not observe any relationships between clinical measures such as antiparkinsonian

medication, disease severity and duration of disease and [18F]-FEPPA VT in the striatum,

which is consistent with previous neuroimaging studies (Gerhard et al., 2006; Kobylecki et

al., 2013). These findings thus imply that these variables are not associated with TSPO

expression in PD patients.

Our findings did not reveal a significant disease effect on [18F]-FEPPA VT in the striatum.

Postmortem and neuroimaging studies showed evidence of neuroinflammatory processes in

PD while the degree of neuroinflammation might vary. Further investigations warrant to

determine whether [18F]-FEPPA could be used as a biomarker of neuroinflammation in PD

by investigating a large sample size and to understand the interactions between the

rs6791polymorphism, neuroinflammation, and clinical subtypes as well as disease prognosis.

Page 144: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

120

6.0 General Discussion

6.1 Summary of Findings

The studies included in this thesis used multimodal imaging tools and techniques to

investigate structural, functional, and molecular changes and their associations with clinical

manifestations in PD. The specific aims of each study were (1) to elucidate the relationships

between structural abnormalities and clinical manifestations of PD, (2) to investigate changes

in brain network properties with focus on the disease-associated nodes and hubs using the

most advanced graph theoretical approach, and (3) to assess TSPO polymorphism and

neuroinflammation using a second generation TSPO radioligand, [18F]-FEPPA in PD.

Overall, the main findings were:

1. Compared to HC subjects, PD patients showed structural changes including cortical

thinning in the left SFG (BA8 and BA9) and in the left M1 extending anteriorly to the

MFG and posteriorly to the S1, as well as diffuse WM abnormalities in bilateral

frontal and temporal regions, in the left parietal and occipital regions as well as

subcortical regions in a subgroup of the patients.

2. The cortical thinning in the left SFG and the WM abnormalities were associated with

poor performance on the overall cognition as well as executive function measured by

neuropsychological tests in PD patients. The significant cortical thinning in the left

SFG solely accounted for the associations with cognitive function, suggesting that

this region play an important role in cognition and be a potential biomarker for earlier

cognitive impairment in PD.

3. Diffuse WM changes occurred in the subgroup of PD patients, suggesting that WM

changes precede cortical GM changes and that WM changes measured by TBSS be a

biomarker for the early stage of disease.

4. Within the SMN, PD patients showed weakened connectivity in key nodes including

the right pre-SMA and right MIns compared to HC subjects. Additionally, the left

MIns lost its hub properties in PD patients.

5. The reduced bridging role of the right pre-SMA was associated with more severe

Page 145: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

121

bradykinesia, which specifically impaired network integration with DMN, suggesting

that the nodal change may affect the redirection of attentional processes from self-

reflection (i.e., internal reference) to goal-directed behavior. The finding also

suggests that pre-SMA be an important node possibly for both motor and cognitive

symptoms.

6. The hubness in the left MIns was positively correlated with dopaminergic medication,

suggesting the role of dopaminergic medication in the connectivity of the node.

7. Within the executive cognitive subnetworks, the left AIns lost its hub properties,

while a new hub region was identified in the CN in PD patients paralleled by

increased levels of inter- and intra-connectivity in the DLPFC compared to HC

subjects.

8. The increased connectivity in the cognitive subnetworks might reflect “attempted

compensatory” or dedifferentiation in our PD patients.

9. The insula plays an important integrative role in brain network communication,

which is susceptible to PD pathology.

10. Using [18F]-FEPPA, PD-HABs showed significantly higher VT values than PD-MABs

in the putamen and CN. However, there was no evidence of elevated levels of

neuroinflammation in the striatal regions in PD patients compared to HC subjects or

no correlations between TSPO density and clinical measures in PD patients.

6.2 Structural and functional changes in sensorimotor nodes in

Parkinson’s disease

In study 1, we demonstrated cortical thinning in the M1 in our patients. The cortical thinning

and faster rate of thinning in the M1 in PD has been reported in several studies (Compta et

al., 2013; Huang, Lou, Xuan, Gu, Guan, Xu, et al., 2016; Ibarretxe-Bilbao et al., 2012; Kim

et al., 2014; Pereira et al., 2012; Segura et al., 2014; Uribe et al., 2016). Similar to the

findings of four of these studies, we found cortical thinning in the face area of M1. This

atrophy may reflect impairment in speech activity as the ventral sensorimotor area is

Page 146: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

122

associated with speech performance (Yeo et al., 2011). In addition, over 90% of PD patients

develop speech disorder (i.e. hypophonia, slurred speech), and PD patients had significantly

reduced brain activation during a speech task compared to HC subjects (Pinto et al., 2011).

In study 2, we found functional changes in the MIns and pre-SMA in the SMN in our

patients. (Note that in this section, “mid-posterior Ins” is used to discuss the findings of

different studies because different studies used the MIns or the posterior insula (PostIns) for

the similar MNI coordinates.) Insular cortex is an important area to PD. First, the Ins is

highly interconnected with the striatum in a connectivity gradient from posterior to anterior,

with PostIns projecting to the dorsal/posterior striatum, and AIns progressively towards

anterior and ventral regions of the striatum. This organization is highly consistent with the

functional roles of both dorsal/posterior Ins and striatum in sensorimotor processes, and

anterior/ventral regions of these structures in cognitive and affective processing (Chikama,

McFarland, Amaral, & Haber, 1997). Second, α-synuclein aggregates were found throughout

insular cortex starting at Braak stage 5. Third, PD-MCI patients showed reduced D2 receptor

availability in the bilateral mid-posterior Ins compared with HC subjects (Christopher, Duff-

Canning et al., 2014; Christopher et al., 2015). Fourth, the meta-analysis study confirmed

one of the important insular subregions associated with PD was the dorsal mid-posterior Ins,

which was associated with an off-medication state, suggesting the vulnerability of this

subregion without dopaminergic medication (Criaud et al., 2016). Lastly, the meta-analysis

of VBM studies in PD reported volume loss in the Ins (Pan, et al., 2012).

Study 2 further corroborated the insular involvement in PD. First, the right mid-posterior Ins

showed reduced FC within the SMN compared to HC subjects. Second, the left mid-posterior

Ins lost hub function and its reduced hub role was associated with lower dose of

dopaminergic medication. The mid-posterior Ins is implicated in the processing of position,

movement, and sensation of the body (Cerasa et al., 2006; Chang, Yarkoni, Khaw, & Sanfey,

2013). Thus, our findings of abnormalities in the sensorimotor Ins may be associated with

postural instability, which is one of the cardinal motor symptoms but typically appears at

more advanced stage.

Page 147: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

123

Furthermore, we demonstrated diminished bridging role of the right pre-SMA with the DMN

nodes in PD patients compared to HC subjects. SMA or pre-SMA has been implicated as a

hub in a few studies in healthy adults (de Pasquale et al., 2013; de Pasquale, Della Penna,

Sporns, Romani, & Corbetta, 2015; Spreng et al., 2013; Yeo et al., 2011). SMA was

identified as a hub region along with other hubs in the DMN and dorsal attention network

(DAN) in healthy adults (de Pasquale et al., 2015). As hubs tend to connect with one another,

the authors speculated that these hubs allow for the integration of internal cognitive

processes such as memory and self-referential behavior medicated by the DMN, the selection

of environmental and body information mediated by the DAN, and the motor planning and

execution mediated by the SMN. Thus, it is possible that pre-SMA plays an important role in

funneling the internal cognitive processes and passing on to the SMA proper for self-initiated

movements.

Importantly, the diminished bridging role of the right pre-SMA measured by BC was

associated with more severe bradykinesia in the patients. Bradykinesia could arise from

slowness in formulating the instructions to move before the onset of and during the actions

(Berardelli et al., 2001), suggesting the cognitive involvement in the generation of the

symptom. The pre-SMA is involved in motor planning based on internally generated thought

(Chung, Han, Jeong, & Jack, 2005). Thus, compromised FC between the pre-SMA and

cognitive nodes might lead to slowness to move and slow movement. The DMN nodes

(BA40 and BA20) that had reduced connectivity with the pre-SMA in our study are

functionally connected to the posterior putamen and the posterior CN in PD patients and HC

subjects (Helmich et al., 2010). Thus, these DMN nodes are also involved in sensorimotor

function. In fact, imagery of movements activates pre-SMA as well as the inferior parietal

cortex (Macuga & Frey, 2011).

6.3 Structural and functional changes in cognitive nodes in

Parkinson’s disease

We found structural and functional changes in the DLPFC in addition to functional changes

Page 148: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

124

in the AIns in our PD patients. DLPFC is a core region of FPN or executive control network

(Dosenbach et al., 2007; Seeley et al., 2007; Trujillo et al., 2015), as well as the fronto-

striatal DA network (Gratwicke et al., 2015), and has been also implicated as an anatomical

and a functional hub associated with executive functions in healthy brains (Gong et al., 2009;

Spreng et al., 2013; van den Heuvel & Sporns, 2013). The most common cognitive

impairment of PD patients is executive dysfunction due to a disruption of the fronto-striatal

DA system as well as the FPN possibly due to cholinergic and/or noradrenergic dysfunction

(Gratwicke et al., 2015).

Importantly, we demonstrated structural changes in the left SFG (BA8 and BA9) (Koshimori,

Segura et al., 2015) and functional changes in bilateral DLPFC (BA9) compared to HC

subjects in study 1 and study 2, respectively. In study 1, we also demonstrated that cortical

thinning in the left SFG was associated with poor executive and overall cognitive

performance on a neuropsychological test battery (Koshimori, Segura et al., 2015).

Supporting the important role of this region in cognition, the SFG is one of the regions

shown cortical thinning at the baseline assessment in PD patients who were later converted to

PDD (Compta et al., 2013). Similarly, multiple protein pathologies (tau, beta-amyloid, α-

synuclein) in BA9 along with the superior and middle temporal gyrus were associated with

the development of dementia in PDD and DLB (Howlett et al., 2015). The lateral SFG is

thought to be involved in executive processing such as monitoring and manipulation (Postle,

Stern, Rosen, & Corkin, 2000). However, the left SFC in particular is functionally connected

not only to a number of distributed FPN regions but also to most of DMN regions, making

this area a key region for internally-focused, goal-oriented cognition (Spreng et al., 2013).

Thus, abnormalities in this region could have substantial effects on cognitive function in PD

patients.

In addition, we demonstrated increased FC in DLPFC (BA9 in MFG) of the FPN within and

between cognitive subnetworks compared to HC subjects. Increased FC in the fronto-parietal

module was also reported in PD-MCI patients using graph theoretical analysis (Baggio et al.,

2014) and during cognitive tasks in PD patients (Gerrits et al., 2015; Madhyastha et al.,

2015; Trujillo et al., 2015). In aging or neuropathologic literature, hyper-activations are

interpreted as compensation when older adults or patients perform at the same level as

Page 149: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

125

younger adults or HC subjects (Cabeza, Anderson, Locantore, & McIntosh, 2002; Clement,

Gauthier, & Belleville, 2013; de Vries et al., 2014), or when increased brain activity is

positively correlated with cognitive performance in older adults or patients, but not in

younger adults or HC subjects (Davis, Dennis, Daselaar, Fleck, & Cabeza, 2008; Della-

Maggiore et al., 2000; Madden et al., 1999). However, when hyper-activations are not related

to clinical or cognitive measures, this refers to as “attempted compensation” (Grady, 2012),

which applied to our findings. Our findings can be also interpreted as pathological responses

called dedifferentiation defined by inefficient use of neural resources or reduction in

specific/selective recruitment of brain regions (Fornito, Zalesky, & Breakspear, 2015; Grady,

2008). It may be possible that at early disease compensation occurs, but as the disease

progresses, brain changes such as dedifferentiation appear before a full-blown dysfunction

develops (Gratwicke et al., 2015).

One possible pathological mechanism for higher FC is due to GABA deficiency resulting in

glutamatergic disinhibition and excessive Glu release (Krystal et al., 2003; Lewis, Pierri,

Volk, Melchitzky, & Woo, 1999) or the impaired N-methyl-D-aspartate (NMDA)

conductance/Glu neurotransmission onto GABAergic interneurons and thereby induces

cortical disinhibition (Anticevic et al., 2012; Krystal et al., 2003). Glu overactivity in the BG

is well documented in PD (DeLong & Wichmann, 2015). Postmortem PD brains showed

impairment of GABAergic neurotransmission in BA9 (Lanoue, Dumitriu, Myers, &

Soghomonian, 2010). On the other hand, significantly reduced vesicular Glu transporters

were found in the BA9 and anterior part of the superior temporal gyrus of postmortem PD

brains (Kashani, Betancur, Giros, Hirsch, & El Mestikawy, 2007). To date, there were two

studies using proton magnetic resonance spectroscopy (1HMRS) at high magnetic field

strengths demonstrated reduced Glu (reductions in Glu/Cr) in the posterior cingulate cortex

in PD patients (Griffith, Okonkwo, O'Brien, & Hollander, 2008) and in PDD patients

(Griffith et al., 2008).

We also found that the left dorsal AIns lost its hub role, which was consistent with another

study using graph theoretical analysis in PD patients (Tinaz et al., 2016). The hub role of the

dorsal AIns is also supported from a meta-analysis study showing that it is the overlap region

for multimodal functions including cognition, interoception, empathy, emotion, pain,

Page 150: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

126

olfaction, and gustation (Kurth et al., 2010). Furthermore, the AIns is known as a core region

of the SAL, which is involved in switching between cognitive subnetworks (i.e., central

executive network and DMN) (Seeley et al., 2007). In PD-MCI patients, reduced D2 receptor

availability in the right dorsal AIns was associated with executive and memory dysfunction

(Christopher, Duff-Canning et al., 2014; Christopher et al., 2015). The meta-analysis study

also showed that the bilateral AIns was associated with cognitive function in PD patients

(Criaud et al., 2016).

Furthermore, in the CON, the right CN emerged as a new hub while the left dorsal AIns lost

its hub role in our patients. This may be explained by the “hub failure” at the chronic stage of

disease proposed by Stam (2014). This model postulates that in the acute stage of brain

disease, hubs take up connections from nodes that redirect from the affected nodes. If this

abnormal redirection is severe and sustained, hubs themselves may deteriorate and diminish

their role of handling information traffic. In the chronic stage of disease, the affected hubs

reroute their traffic to nodes at lower levels of hierarchy, resulting in the decreased centrality

of the original hubs and local emergence of new connections, which may also be an

emergence of new hubs.

6.4 Dopaminergic modulation on resting-state functional

connectivity

Abnormalities in resting state FC have been found in PD patients both in ON and OFF states

compared with HC subjects (Tahmasian et al., 2015 for review). However, there are

significant effects of dopamine replacement therapy (DRT) on FC at rest, which was

demonstrated by contrasting ON and OFF states using various FC methods. In general,

administration of DRT normalizes abnormalities in FC in PD patients. For example, there is

one study that investigated functional changes in a motor network using graph theoretical

analysis in both ON and OFF states compared with HC subjects (Wu, et al., 2009). This

study included 18 nodes associated with a motor network and measured their nodal degree

(i.e., the number of connections each node has). There were six nodes showing significant

Page 151: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

127

group differences after correcting multiple comparisons between PD patients in an OFF state

and HC subjects including the SMA, cerebellum, M1, parietal cortex, DLPFC, and putamen

(P ≤ 0.003) and group differences in the globus pallidum and premotor cortex to lesser extent

(0.003 < P < 0.05). After the administration of DRT, the five nodes except the left parietal

cortex were normalized. However, PD patients in an ON state still showed some decrease in

nodal degree in the SMA (P = 0.02), putamen (P = 0.016), and cerebellum (P = 0.042)

compared with HC subjects. The nodal degree in the SMA, putamen, and thalamus showed

negative correlations with UPDRS III scores while the nodal degree in the M1, cerebellum,

premotor cortex, and parietal cortex showed positive correlations with UPDRS III scores.

Whether or not these correlations persist in an ON state is unknown, as they were

investigated only in an OFF state.

Dopaminergic effects are also known to affect cognitive performance primarily in executive

function and modulate brain activation/deactivation in PD patients (Poletti and Bonuccelli,

2013 for review). However, the effects of dopaminergic medication on cognitive networks at

rest were little studied in PD patients. In one study, FC at rest as well as the spectral

compositions of rsfMRI oscillations were investigated before and after the administration of

DRT in drug-naïve PD patients and compared with those in a placebo group of drug-naïve

PD patients as well as HC subjects (Esposito et al., 2013). The levodopa-induced spectral

changes in low-frequency band were observed in visual network and bilateral FPN.

However, cognitive performance associated with the changes is unknown.

There were more studies on dopaminergic modulation on resting state FC in healthy subjects.

One study found liner effects of dopaminergic modulation (L-dopa > placebo > haloperidol)

on RSNs including between bilateral midbrain and DMN, between dorsal caudate and right

FPN, and between the ventro-medial thalamus and left FPN, as well as between the ventral

striatum and salience/executive network (Cole, Oei, Soeter, Both, van Gerven, Rombouts, et

al., 2013). However, differential effects of dopamine were also reported depending on nodes

within networks. For example, haloperidol group showed greater FC in the left precentral

and middle frontal gyri while it showed reduced FC in supramarginal gyrus/intraparietal

sulcus within the DMN (Cole, Beckmann, Oei, Both, Gerven, and Rombouts, 2013),

suggesting the complex effects of dopamine on cognitive networks.

Page 152: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

128

Dopamine modulation on FC and cognitive performance may be more complicated in PD

patients than healthy subjects. It can depend on differential effects of different medication

such as levodopa, dopamine agonists, or different dopamine agonists when PD patients are in

an ON state, acute and chronic effects of the medication (Poletti and Bonucelli, 2013), and

the interactions of these factors with COMT Val58Met polymorphism of individual PD

patients.

Most studies including the current studies enrolled PD patients who already have been

medicated. The chronic effects of dopaminergic medication on cognitive status in PD

patients are largely unknown due to paucity of literature (Poletti and Bonucelli, 2013).

However, there were three randomized longitudinal studies (Kulisevsky et al., 2000;

Rektorová et al., 2005; Relja and Klepac, 2006). They consistently concluded while

dopaminergic medication significantly improved motor symptoms, it had differential or no

effects on cognitive performance, and was not sufficient to compensate their cognitive

impairment and decline.

Taken together, our findings of reduced FC in the SMN nodes including the pre-SMA and

MIns likely reflected the abnormalities that were not completely restored by dopaminergic

medication in our PD patients with moderate severity. Similarly, our patients showed

significant cognitive impairment even in an ON state and thus dopaminergic medication had

not compensated for the cognitive impairment, which is consistent with the findings of the

three studies above. We found increased FC in the DLPFC nodes in an ON state. As FC in

the DLPFC is generally affected by dopaminergic medication, it cannot totally exclude the

medication effects on these brain changes even though we did not find the correlation

between the changes and LEDD.

6.5 White matter changes in Parkinson’s disease

We found more extensive WM changes in the bilateral frontal and temporal areas and limited

changes in the left parietal and occipital areas. The altered WM tracts and ROIs in the frontal

region included (1) the forceps minor connecting the lateral and medial parts of the frontal

Page 153: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

129

lobes including the anterior CC, (2) cingulum, (3) ATR, and (4) SCR containing motor WM

tracts, and body of CC connecting motor areas. The affected association fibers included (1)

the UF containing cholinergic fibers (Selden, Gitelman, Salamon-Murayama, Parrish, &

Mesulam, 1998) and connecting the orbital cortex and the anterior temporal lobe, (2) SLF in

the frontal, parietal, and temporal regions, (3) ILF in the temporal and occipital region, and

(4) IFO in the frontal, temporal and occipital regions. The Forceps major excluding the

splenium of the CC was affected in the occipital region. Subcortically, MD changes were

also detected in the anterior limb of internal capsule (ALIC) containing ATR, which lies

between the head of the CN medially and the putamen laterally as well as in the EC adjacent

to the putamen and Ins, which containing association fibers such as SLF and IFO,

commissural fibers as well as ascending cholinergic fibers (Selden et al., 1998).

Combined with the findings of previous studies, frontal WM tracts are vulnerable in PD

(Agosta et al., 2013; Agosta et al., 2014; Deng et al., 2013; Hattori et al., 2012; Matsui,

Nishinaka, Oda, Niikawa, Komatsu et al., 2007; Melzer et al., 2013; Rae et al., 2012). We

also found bilateral temporal WM changes. In postmortem PD brains, the reduction of

serotonergic axonal density was most severe in the temporal and prefrontal regions compared

to normal brains (Azmitia & Nixon, 2008). In addition, the axons in these regions displayed

enlarged vesicles, splayed ending, clustering and degenerating profile. We also found that

WM abnormalities were associated with executive and global cognitive dysfunction. This is

not surprising as association fibers connecting different cortical regions were disrupted in our

patients.

A majority of studies investigated WM changes using both FA and MD. Our study

demonstrated WM changes measured by MD. Only MD changes but not FA changes were

detected in PD patients with normal cognition (Melzer et al., 2013). Other studies also

showed changes in MD, but not in FA (Agosta et al., 2013; Kim et al., 2013). In addition,

MD showed a larger area of WM changes than FA (Gallagher et al., 2013; Theilmann et al.,

2013). Furthermore, MD values were more often correlated with neuropsychological test

scores in different domains than FA values (Melzer et al., 2013; Zheng et al., 2014). FA and

MD are thought to reflect different tissue characteristics where FA is more sensitive to tissue

directionality and organization (architecture) and MD to tissue density (Farina et al., 2000;

Page 154: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

130

Wiltshire et al., 2010). Depending on WM region, disease stage, and the presence of

cognitive impairment, either MD or FA can be more sensitive detecting WM changes

(Agosta et al., 2014; Rae et al., 2012; Theilmann et al., 2013; Zhan et al., 2012). Thus

investigating both DTI indices are most likely to capture changes in WM microstructure.

6.6 Neuroinflammation in Parkinson’s disease

6.6.1 Interindividual variability in total distribution volume

It is now known that it is important to account for TSPO binding affinity classes based on the

rs6971 polymorphism in the statistical analysis to reduce interindividual variability in VT

values. Yet, within the same genetic group, there is still a large variability in VT values,

resulting in a considerable overlap in VT values between HABs and MABs. The previous

study reported that most MABs expressed their two binding sites in approximately equal

proportions with the mean fraction of high affinity sites was 58% ± 6.6% in vitro and 63% ±

9.8% in platelets (Owen et al., 2011). However, the fraction of high-affinity PBR28 binding

sites within MABs ranges from 38% to 83% in vitro. Thus, the authors discussed the

possibility of a continuum in the expression of the HAB and LAB sites, which may explain

some of the variability VT values within the same genetic group.

Age may be another factor accounting for the variability of VT values. As discussed in

section 1.3.4.3, there are age-related changes in microglia and astrocytes. So far, the effect of

age on TSPO outcome measures in healthy subjects has yielded inconsistent results using

second-generation TSPO radioligands. There was no significant age effect using [11C]-

DAA1106 (Yasuno et al., 2008), [18F]-FEPPA (Suridjan et al., 2014) or using [11C]-DPA713

(Yokokura et al., 2016). On the other hand, the age effect was detected in the whole brain

using [11C]-vinpocentine (Gulyas et al., 2011), in most GM brain regions using [11C]-

PBR111 (Guo et al., 2013), as well as frontal and temporal lobes using [11C]-PBR28

(Bloomfield et al., 2016). To avoid the potential age effect on TSPO outcome measures,

study groups should be age-matched and age can also be included in the analysis as a

covariate.

Page 155: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

131

Another possible factor may be the variability of plasma free fraction (fp) (Bloomfield et al.,

2016). Although previous [18F]-FEPPA studies in AD and MDE demonstrated significant

group differences without accounting for the effect of the fp (Setiawan et al., 2015; Suridjan

et al., 2015), future studies are needed to determine whether the inclusion of fp would

improve the sensitivity detecting neuroinflammation in clinical populations (Suridjan et al.,

2015).

The reproducibility of the outcome measures may play a role in generating the variability.

One study tested the reproducibility of VT values in six HABs including three HC subjects

and three HIV patients. They underwent [11C]-DPA713 PET scans twice in the morning and

afternoon on the same day and showed significant systematic differences in VT values

between the two scans (Coughlin et al., 2014). VT values obtained from the afternoon scans

were consistently higher than those obtained from the morning scans in all of the 16 ROIs

investigated with interater correlation coefficient (ICC) being 0.028 ± 0.057. The authors

also reported the inclusion of fp did not change the results. The underlying mechanisms for

the systematic differences are unknown. However, the authors discussed possible

contributing factors such as hormone-medicated phasic changes in TSPO expression, tonic

changes due to anxiety associated with the study procedure, or changes in the level of blood

cholesterol due to food intake between the two scans (Coughlin et al., 2014). These factors

may also contribute to the variability and obscure the true group differences. The

reproducibility of VT values using [18F]-FEPPA needs to determine.

In an effort to reduce the interindividual variability, some recent studies using the second-

generation TSPO radioligands have used normalized outcome measures such as VT ratio (the

ratio of the VT in the ROI to VT in the whole brain or GM), and found significant group

differences between clinical populations and HC subjects (Bloomfield et al., 2016; Coughlin

et al., 2014). However, there are some issues about using the normalized outcome measure.

In such approach, VT value in the ROI is also included in the denominator to calculate the

ratio. In addition, normalization using the whole brain or GM can compromise the accurate

estimate of neuroinflammation in pathological brains where neuroinflammation can occur in

diffuse areas of brain, which may be the case of PD. Furthermore, as discussed in section

1.6.3.5.3, TSPO density is different between in GM and WM, and the pharmacological

Page 156: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

132

properties of [18F]-FEPPA in WM are different from those in GM. Thus, normalization using

the whole brain including WM or WM poses a problem.

Taken together, there are some factors that can be taken into consideration in the study and

analysis procedures to reduce noise. This may lead to the reduction of the exiting

interindividual variability in VT values and thereby increase the sensitivity of [18F]-FEPPA

detecting neuroinflammation in PD patients.

6.6.2 [18F]-FEPPA binding

VT value is a reliable measure of [18F]-FEPPA and other second-generation TSPO

radioligands. However, specific binding of most of these radioligands is unknown. There was

no suitable pharmacological agent to perform blocking studies to estimate non-displaceable

volume of distribution (VND) in human subjects. However, more recently, [11C]-PBR28 VND

and BPND (VT/VND-1) were estimated using the TSPO agonist XBD173 (emapunil),

medication for generalized anxiety (Rupprecht et al., 2009) in the healthy human subjects

and reported specific binding accounted for a substantial portion of VT (Owen et al., 2014).

Population VND was estimated to be 1.98 (95% Confidence Interval 1.69, 2.26).

Both microglia and astrocytes are the major cellular sources of TSPO expression in the brain

(Lavisse et al., 2012; Venneti et al., 2009). Unlike acute insults such as encephalitis, stroke,

or traumatic brain injury in which peripheral immune cells enter through damaged BBB,

microglia and astrocytes are mostly likely key immune response cells in neurodegenerative

diseases such as PD (Ouchi, Yagi, Yokokura, & Sakamoto, 2009). [3H]-DAA1106 binding

overlapped mostly with activated microglia compared with activated astrocytes in various

pathological conditions such as cerebral infarcts, amyotrophic lateral sclerosis, AD,

frontotemporal dementia, and MS (Venneti et al., 2008). However, due to lack of

autoradiography and immunohistochemistry studies of [18F]-FEPPA, the cellular sources of

[18F]-FEPPA binding is unknown.

6.6.3 Neuroinflammation in Parkinson’s disease

Our preliminary investigation of neuroinflammation using [18F]-FEPPA showed genetic

Page 157: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

133

effects on VT values in both PD patients and HC subjects in the putamen and CN. HABs

showed significantly higher VT values than MABs in these regions except HC group in the

CN. On the other hand, there were no significant disease effects in either region. PD-HABs

showed 16% increase in TSPO binging compared to PD-MABs in these regions.

Some of the previous studies using [11C]-PK11195 found elevated microglial activation in

the putamen (Iannacone, et al., 2012; Ouchi, et al., 2009) and striatum (Gerhard, et al., 2006)

while others did not in the striatum (Bartel, et al., 2010; Edison, et al., 2013; Kobylecki, et

al., 2013). In fact, intervariability in neuroinflammation in PD patients was also reported in

postmortem (Graeber, Bise, & Mehraein, 1994) and neuroimaging studies (Edison et al.,

2013). It can depend on the disease stage, proximity to the lesion (Cosenza-Nashat et al.,

2009), disease severity, the rate of disease progression, and polymorphism in immune-related

genes (International Parkinson Disease Genomics Consortium et al., 2011). The investigation

of neuroinflammation will be extended to include a larger sample size or PD patients with

different clinical characteristics in the whole brain regions. Such study will determine

whether [18F]-FEPPA can be a biomarker for PD.

6.7 Neuroimaging biomarkers for Parkinson’s disease

This thesis utilized different neuroimaging tools to investigate structural, functional and

molecular changes in PD. The three studies included different cohorts of PD patients and HC

subjects. Therefore, the conclusions integrating all the findings should be interpreted with

caution. Our results suggest that (1) frontal motor (i.e., pre-SMA) and cognitive (i.e.,

DLPFC) regions may be early biomarker for motor and cognitive symptoms of PD, (2)

increased FC in the DLPFC may precede more substantial brain changes such as atrophy

associated with cognitive impairment, and (3) WM changes may precede GM changes and

functional changes as it revealed more pathological changes in diffuse area of the brain or

DTI with TBSS be the most promising biomarker detecting pathological changes compared

with other methods. Lastly, there was no evidence of neuroinflammation in the striatum of

PD. TSPO density was not correlated with any clinical measures, either. However, further

studies are needed to conclude whether neuroinflammation can be a biomarker for PD by

Page 158: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

134

extending ROIs beyond the striatum, in particular in the frontal cortex and white matter

where we found brain changes using the other methods (Figure 6-1).

To date, there has been no single study using the multimodal neuroimaging tools and

analysis methods employed in this thesis. However, the findings of the two studies that

investigated GM, WM, and FC in the same cohort of PD patients are in line with our

findings. One study used VBM, TBSS and tractography for pedenculopontine tract, as well

as ICA to compare them between PD patients with freezing of gait with HC subjects (Canu et

al., 2015). The other study used CTA, TBSS and seed-based FC analysis with bilateral

posterior parietal cortex and CN as seed regions to compare these changes among PD

patients with normal cognition, PD patients who developed MCI in earlier disease and those

who developed MCI in later disease (Shin et al., 2016). Neither of the studies found

significant group differences in gray matter changes. On the other hand, both studies found

significant group differences in WM and FC changes. Using the whole brain analysis

methods including TBSS and ICA, WM revealed distributed changes in cortical and

subcortical regions including midbrain as well as cellebrum while FC changes were found

primarily in the sensorimotor network in PD patients with freezing of gait in an OFF

medication compared with HC subjects (Canu, et al., 2015). In addition, severe motor

symptoms measured by UPDRS III scores were associated with greater axial diffusivity of

the right pedunculopontine tract in these patients. In the other study, TBSS revealed

extensive changes in the frontal region, which corroborated our findings with the frontal

white matter most affected. Thus, consistent with our findings, these studies suggest that

WM and FC changes may precede GM changes or that DTI with TBSS be more sensitive

neuroimaging method to detect brain abnormalities associated with PD symptoms than

structural MRI with CTA or rsfMRI with ICA in PD.

Page 159: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

135

Figure 6-1. Neuroimaging biomarkers for Parkinson’s disease. Our findings suggest that

TBSS be most promising biomarker for PD. However, further investigation on

neuroinflammation in the extra-striatal GM regions and white matter needs to confirm this

conclusion.

6.8 Limitations

Some limitations must be considered when interpreting the results of the studies included in

this thesis. First, the sample sizes were relatively small due to the inherent difficulty in

recruiting patients and “healthy” older adults. The replication of the findings in a larger

sample size is needed. Small sample sizes also limit us from subdividing patients into

subgroups with different disease stages, motor symptoms (e.g., right or left dominant,

tremor-dominant), cognitive phenotypes, and types of dopaminergic medication to further

dismantle the complexity of PD. Second, the study procedures were performed in an ON

medication state for patients except for the [18F]-FEPPA scan. This is due to the concerns

Page 160: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

136

about the side effects and worsening of motor symptoms that patients may experience

without medication, which also can compromise the data quality. Thus, the findings in the

studies are associated with medicated PD patients. Third, the exact mechanisms underlying

for the changes in MRI and rsfMRI data are unknown. Similarly, the exact roles of activated

microglia and astrocytes (i.e., neurotoxic or neuroprotective) are unknown. Behavioral

correlates may help to interpret them. Fourth, the [18F]-FEPPA scans were performed using

HRRT, which features high spatial image resolution and high sensitivity (Wienhard et al.,

2002) and thereby allows for the accurate quantitative measurement of the radioligand

concentration even in small regions and regions with low tracer concentrations, such as the

striatal regions. However in aging and disease brains, where brain atrophy and enlarged

ventricles can be a significant issue for the accurate quantification, we analyzed our data both

with and without PVEC using the Mueller-Gartner PVEC algorithm. However, geometric

transfer matrix (GTM) (Rousset, Ma, & Evans, 1998) is often used for subcortical structures

such as BG to address spill-over effects due to the close proximity of structures (e.g. CN and

putamen) (Erlandsson, Buvat, Pretorius, Thomas, & Hutton, 2012). Lastly, there are general

limitations of second-generation TSPO radioligands including the inability to assess LABs

and the requirement of serial arterial blood sampling for quantification of TSPO, which poses

a challenge for the clinical use (Vivash & O'Brien, 2016). However, alternative methods for

the serial arterial blood sampling to derive input function have been proposed (Lavisse et al.,

2015; Mabrouk et al., 2014).

Page 161: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

137

7.0 Conclusion

This thesis has shed light on both structural and functional brain changes underlying for the

clinical manifestations in PD patients using multimodal neuroimaging tools and techniques.

We demonstrated that cortical thinning in the left SFG (BA8 & 9) and diffused WM

abnormalities with frontal and temporal WM tracts mostly affected accounted for general

cognition as well as executive function in PD patients. We also demonstrated that the

diminished bridging role of pre-SMA with DMN is associated with more severe

bradykinesia. Taken together, we demonstrated the frontal changes as earlier sign of disease

associated with motor and cognitive dysfunction, and that structural MRI using CTA, DTI

with TBSS, and rsfMRI with graph theoretical analysis are potential in vivo biomarker for

PD. Among these brain changes, white matter changes were observed in diffuse area of the

brain, suggesting DTI with TBSS as the most promising neuroimaging method to detect

pathological changes in PD. Finally, using a second generation TSPO radioligand, [18F]-

FEPPA, the percentage of increase in TSPO binding in PD patients was 16 % in the putamen

as well as in the CN, which did not yield a statistically significant group difference. The

TSPO density was not correlated with any clinical measures, either. However, further studies

are needed to conclude whether neuroinflammation measured by [18F]-FEPPA can be a

biomarker for PD by extending ROIs beyond the striatum and by investigating a larger

sample size of PD patients as well as PD patients with different clinical characteristics.

Page 162: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

138

8.0 Future Directions

In addition to addressing the limitations listed above, several possible future directions

arising from the studies in this thesis are as follows:

Proposed study 1: Whole brain investigation of neuroinflammation

We did not find any evidence of neuroinflammation in the striatum in 16 PD patients

compared with 16 HC subjects. However, we demonstrated pathological changes measured

by CTA and graph theoretical analysis in the frontal cortex in PD patients. Postmortem

studies showed more extensive neuroinflammation in the frontal cortex (Mythri et al., 2011)

and cortical regions (Braak et al., 2007) than in the striatal region. Thus, a pertinent

investigation of the extra-striatal GM regions would be needed. Similarly, we demonstrated

diffuse WM pathological changes in the cortical and subcortical regions, and postmortem PD

brains showed extensive axonal neuropathology, suggesting that further investigations

warrant in WM.

Proposed study 2: Neuroinflammation in subtypes of patients with Parkinson’s disease

Investigation in PD patients with different clinical characteristics may reveal more specific

role of neuroinflammation in different symptoms such as cognitive impairment, psychiatric

problems, and fatigue. Inflammatory markers in CSF samples from 87 PD patients including

16 PDD were compared with those form 33 HC subjects and studied for their association

with the severity of depression, anxiety, fatigue, and cognitive impairment (Lindqvist et al.,

2013). Only PDD patients showed significantly elevated levels of some of these markers

compared with non demented PD patients and HC subjects. However, higher levels of the

inflammatory markers were significantly associated with more severe depression, anxiety,

fatigue, and cognitive impairment, which was not observed in HC subjects. In addition, two

studies using [11C]-PK11195 demonstrated significantly elevated level of microglial

activation in distributed cortical regions (Edison et al., 2013; Fan et al., 2015), and the

amygdala and hippocampus (Fan et al., 2015) in PDD compared with HC. Both studies

showed inverse correlations between the level of microglial activation and MMSE scores.

Page 163: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

139

Furthermore, neuroinflammation was detected in patients with AD (Suridgan, et al., 2015)

and with MDS (Setiawan, et al., 2015) using [18F]-FEPPA. Therefore, PD patients presenting

different NMS mentioned above would be investigated whether these patients show

neuroinflammation compared to PD patients without these NMS and to HC subjects using

[18F]-FEPPA. Furthermore, as the role of neuroinflammation has been implicated to

contribute to disease progression, PD patients with faster disease progression and those with

slower disease progression would be studied and followed up longitudinally to determine

whether TSPO imaging using [18F]-FEPPA can be a predictive measure for disease

progression.

Proposed study 3: Association between neuroinflammation and atrophy

We demonstrated that PD patients had structural changes in the study 1. The one of the

potential mechanisms underlying for GM and WM atrophy is neuroinflammation. Recently

published study demonstrated that volume loss in the hippocampus was associated with

elevated levels of neuroinflammation in the hippocampus measured by [11C]-PK11195 in AD

and PDD combined (Femminella et al., 2016). It also showed that the volume loss in the

hippocampus was associated with elevated levels of neuroinflammation in the diffuse

cortical and other subcortical regions. The authors suggested that neuronal degeneration

mediated by microglial activation follows the cortical connection from hippocampus because

of anatomical connections between hippocampal neurons project and the whole cortex via

entorhinal cortex. A few studies have investigated WM changes measured by DTI and

neuroinflammation using [11C]-PK11195 (Ramlackhansingh et al., 2011; Scott et al., 2015;

Thiel et al., 2010). However, to date, the relationships between structural and

neuroinflammation using the second generation TSPO radioligands have been little studied.

One study investigated TSPO binding using [11C]-DPA-713 as well as GM changes using the

CTA and subcortical volume analysis in the former NHL football players (Coughlin et al.,

2015). However, the relationships between GM changes and TSPO findings were not

investigated or not reported. Thus, multimodal approach combining structural MRI using

CTA, DTI, and TSPO imaging with [18F]-FEPPA may be able to elucidate such

relationships. Neuroinflammation has been implicated to contribute to the disease

progression of PD and it is also possible that positive findings of TSPO imaging may precede

Page 164: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

140

GM or WM atrophy detected by MRI. Thus, a longitudinal study would reveal whether

TSPO density at one point could predict the disease progression that would be manifested on

structural changes at a following up.

Proposed study 4: Modeling Parkinson’s disease using hub changes

The current study identified nodes as hubs using network measures combined degree and

BC. However, as we demonstrated, diminished bridging role of a node between networks

measured by BC had an important implication for one of the cardinal motor symptoms of

PD. Thus, hub roles of nodes can be further investigated using graph measures such as

within-module degree z-score and participation coefficient. A node with higher within-

module degree z-score indicates that the node is a provincial hub while a node with higher

participation coefficient is a connector hub that has higher proportion of edges connecting

with nodes in other modules. To date, only one study investigated participation coefficient in

37 PD patients compared to 20 HC subjects (Gottlich et al., 2013). This study investigated

modular participation coefficient. It identified seven modules in HC subjects, and PD

patients showed significantly reduced modular participation coefficient in the visual module

compared to HC subjects. It further interrogated which nodes in the visual module

significantly reduced participation coefficient in PD patients compared with HC subjects, and

found that the calcarine and cuneus showed significant reduction in connectivity with frontal

regions including the ACC, superior orbital region and rolandic operculum. However, this

study used anatomically parcellated ROIs, which is known to compromise functional

boundaries. In addition, nodal participation coefficient was not investigated in other modules.

Furthermore, which nodes are high in participation coefficient in each module was not

investigated. Therefore, in the proposed project, data-driven nodes and modules would be

used. To this end, a group of older HC subjects would undergo fMRI scans, during which

they perform motor and cognitive tasks such as executive, memory, working memory tasks

to derive functional nodes. Additionally, PD patients and another group of older HC subjects

would undergo rsfMRI scans and these two groups would be interrogated for modular and

hub organizations including modularity, within-module degree z-score and participation

coefficient. Affected hubs in PD would be further investigated for their association with

clinical measures and for their target connections using a seed-based analysis as we

Page 165: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

141

performed in study 2. This study would be longitudinally followed up. In the proposed

studies, it is hypothesized that pathology of connector hubs would (1) have greater impact on

symptoms than that of provincial hubs, (2) predict spread of disease depending on where the

hubs have connection to, and (3) predict faster progression of disease affecting distributed

brain regions. Furthermore, individual differences in robustness in such hub regions would

be able to predict the variability of disease progression. Longitudinal studies including early-

diagnosed PD patients may be able to confirm these models.

Proposed study 5: Investigation of increased functional connectivity as a pathological

response using MR Spectroscopy

We found increased FC within and between cognitive subnetworks in bilateral DLPFC in PD

patients compared to HC subjects, which is consistent with some of the PD literature.

Increased cortical FC was also reported in other diseases/disorders such as AD, depressive

and bipolar disorders, and SCZ. This enhanced FC is typically interpreted as a compensatory

response to the disease. However, it may also be pathological. Literature suggests that

increased FC in certain regions or increased FC in wider area of regions in cognitive

subnetworks might be due to altered glutamatergic/GABAergic activity (Anticevic et al.,

2012; Fornito et al., 2015; Krystal et al., 2003). In fact, postmortem PD brains showed

impairment of GABAergic neurotransmission in BA9 (Lanoue et al., 2010). The recent

development of 1HMRS at high magnetic field strengths allows for more reliable estimation

of the amino acids glutamine (Gln), Glu, and GABA (Ciurleo, Di Lorenzo, Bramanti, &

Marino, 2014) and thus may be able to elucidate the mechanisms underlying for cortico-

cortical FC changes in vivo. To this end, resting-state 1HMRS data would be acquired along

with fMRI data in PD patients and HC subjects. During the fMRI scans, the participants

would perform an executive task associated with the activation of DLPFC (BA9). Although

increased FC in bilateral DLPFC was not statistically associated with dopaminergic

medication in study 2, since DA exerts effects on NMDA and GABA activities, PD patients

would undergo fMRI scans in both on- and off-medication states. This study would allow to

determine (1) whether the resting-state Glu and GABA activities in the DLPFC would be

altered in PD patients compared to HC subjects, (2) whether these activities would predict

brain activation in the DLPFC and executive performance in PD patients, and (3) how

Page 166: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

142

dopaminergic medication would affect cortical Glu and GABA activities as well as brain

activation in the DLPFC and executive function in PD patients.

Proposed study 6: Investigation of structural and functional brain network changes

using graph theoretical approach

Interpreting both structural and functional MRI data can be challenging even in the same

cohort due to different processing and analysis methods as well as outcome measures (e.g.,

gray matter thickness or volume; white matter microstructure; or connectivity strength or

characteristics). Both structural and function MRI data can be analyzed using the graph

theoretical approach. To date, published PD studies and our study applied such analysis

method solely to functional neuroimaging data such as rsfMRI, EEG, and MEG, structural

data including cortical thickness and subcortical volume (Pereira et al., 2015) or DTI data

with tractrography (Nigro et al., 2016). Therefore, by applying the graph theoretical approach

to structural, DTI, and rsfMRI would improve direct comparisons of the findings and thereby

elucidate their relationships with one another, as well as with the clinical symptoms in PD

patients.

Proposed study 7: Multimodal neuroimaging approach using PET and MRI to study

subtypes of Parkinson’s disease

PET can investigate more specific neuropathological changes than MRI, and therefore, such

findings can lead to the opportunities for pharmacological interventions. On the other hand,

MRI can be more preferred in a clinical setting as it is more widely available, more

accessible, less invasive and requires less scan time to monitor the course of disease

compared with PET. To utilize the advantages of both techniques, more studies using

multimodal neuroimaging approach are needed to investigate the relationships among the

outcome measures, and their associations with clinical symptoms. Such studies also need to

conduct longitudinally. In addition to the proposed study 2 (i.e., neuroinflammation and

structural changes), other PET radioligands such as [11C]-MP4A and [18F]FP-TZTP to

measure cholinergic function, and Pittsburgh compound B (PiB) to measure β amyloid

combined with MRI would also be used to study particular PD subpopulation such as PD-

MCI and PDD. PET with [18F]-Setoperone and [11C]-DASB to measure serotoninergic

Page 167: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

143

function combined with MRI would be studied in PD patients with visual hallucinations.

Furthermore, PET findings can be used to group PD patients into subtypes instead of using

clinical/behavioral measures. In such study, PD patients would be first classified into

subgroups, such as high vs. low neuroinflammation, PiB positive vs negative, and PD with

normal vs abnormal cholinergic or serotonergic dysfunction based on their PET findings, and

these two PD patient subgroups would be compared with each other and with HC subjects,

and would be monitored using MRI to further characterize neuropathology in PD subtypes

and NMS. This study would also potentially pave the way for pharmacological interventions

for NMS.

Page 168: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

144

References

Aarsland, D., Bronnick, K., & Fladby, T. (2011). Mild cognitive impairment in parkinson's

disease. Current Neurology and Neuroscience Reports, 11(4), 371-378.

Achard, S., & Bullmore, E. (2007). Efficiency and cost of economical brain functional

networks. PLoS Computational Biology, 3(2), e17.

Achard, S., Salvador, R., Whitcher, B., Suckling, J., & Bullmore, E. (2006). A resilient, low-

frequency, small-world human brain functional network with highly connected

association cortical hubs. The Journal of Neuroscience : The Official Journal of the

Society for Neuroscience, 26(1), 63-72.

Acosta-Cabronero, J., Williams, G. B., Pengas, G., & Nestor, P. J. (2010). Absolute

diffusivities define the landscape of white matter degeneration in alzheimer's disease.

Brain : A Journal of Neurology, 133(Pt 2), 529-539.

Agosta, F., Canu, E., Stefanova, E., Sarro, L., Tomic, A., Spica, V., . . . Filippi, M. (2014).

Mild cognitive impairment in parkinson's disease is associated with a distributed pattern

of brain white matter damage. Human Brain Mapping, 35(5), 1921-1929.

Agosta, F., Canu, E., Stojkovic, T., Pievani, M., Tomic, A., Sarro, L., . . . Filippi, M. (2013).

The topography of brain damage at different stages of parkinson's disease. Human Brain

Mapping, 34(11), 2798-2807.

Akaike, H. (1974). A new look at the statistical model identification. IEEE Trans Automat

Contr, 19, 716-723.

Alexander, A. L., Lee, J. E., Lazar, M., & Field, A. S. (2007). Diffusion tensor imaging of

the brain. Neurotherapeutics : The Journal of the American Society for Experimental

NeuroTherapeutics, 4(3), 316-329.

Alexander, G. E., DeLong, M. R., & Strick, P. L. (1986). Parallel organization of

functionally segregated circuits linking basal ganglia and cortex. Annual Review of

Neuroscience, 9, 357-381.

Alvarez, J. I., Katayama, T., & Prat, A. (2013). Glial influence on the blood brain barrier.

Glia, 61(12), 1939-1958.

Amaral, L. A., Scala, A., Barthelemy, M., & Stanley, H. E. (2000). Classes of small-world

networks. Proceedings of the National Academy of Sciences of the United States of

America, 97(21), 11149-11152.

Page 169: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

145

Andersson, J. L. R., Jenkinson, M., & Smith, S. (2007a). Non-linear optimisation. FMRIB

technical report TR07JA1 from www.fmrib.ox.ac.uk/analysis/techrep.

Andersson, J. L. R., Jenkinson, M., & Smith, S. (2007b). Non-linear registration, aka spatial

normalisation FMRIB technical report TR07JA2 from

www.fmrib.ox.ac.uk/analysis/techrep.

Anticevic, A., Gancsos, M., Murray, J. D., Repovs, G., Driesen, N. R., Ennis, D. J., . . .

Corlett, P. R. (2012). NMDA receptor function in large-scale anticorrelated neural

systems with implications for cognition and schizophrenia. Proceedings of the National

Academy of Sciences of the United States of America, 109(41), 16720-16725.

Antkiewicz-Michaluk, L., Mukhin, A. G., Guidotti, A., & Krueger, K. E. (1988). Purification

and characterization of a protein associated with peripheral-type benzodiazepine binding

sites. The Journal of Biological Chemistry, 263(33), 17317-17321.

Arlicot, N., Vercouillie, J., Ribeiro, M. J., Tauber, C., Venel, Y., Baulieu, J. L., . . .

Guilloteau, D. (2012). Initial evaluation in healthy humans of [18F]DPA-714, a

potential PET biomarker for neuroinflammation. Nuclear Medicine and Biology, 39(4),

570-578.

Ashburner, J., & Friston, K. J. (2000). Voxel-based morphometry--the methods.

NeuroImage, 11(6 Pt 1), 805-821.

Assaf, Y., & Pasternak, O. (2008). Diffusion tensor imaging (DTI)-based white matter

mapping in brain research: A review. Journal of Molecular Neuroscience : MN, 34(1),

51-61.

Azmitia, E. C., & Nixon, R. (2008). Dystrophic serotonergic axons in neurodegenerative

diseases. Brain Research, 1217, 185-194.

Baggio, H. C., Sala-Llonch, R., Segura, B., Marti, M. J., Valldeoriola, F., Compta, Y., . . .

Junque, C. (2014). Functional brain networks and cognitive deficits in parkinson's

disease. Human Brain Mapping, 35(9), 4620-4634.

Baggio, H. C., Segura, B., Ibarretxe-Bilbao, N., Valldeoriola, F., Marti, M. J., Compta, Y., . .

. Junque, C. (2012). Structural correlates of facial emotion recognition deficits in

parkinson's disease patients. Neuropsychologia, 50(8), 2121-2128.

Bammer, R. (2003). Basic principles of diffusion-weighted imaging. European Journal of

Radiology, 45(3), 169-184.

Banati, R. B., Daniel, S. E., & Blunt, S. B. (1998). Glial pathology but absence of apoptotic

nigral neurons in long-standing parkinson's disease. Movement Disorders : Official

Journal of the Movement Disorder Society, 13(2), 221-227.

Page 170: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

146

Banati, R. B., Newcombe, J., Gunn, R. N., Cagnin, A., Turkheimer, F., Heppner, F., . . .

Myers, R. (2000). The peripheral benzodiazepine binding site in the brain in multiple

sclerosis: Quantitative in vivo imaging of microglia as a measure of disease activity.

Brain, 123(Pt 11), 2321-2337.

Barnum, C. J., & Tansey, M. G. (2012). Neuroinflammation and non-motor symptoms: The

dark passenger of parkinson's disease? Current Neurology and Neuroscience Reports,

12(4), 350-358.

Bartels, A. L., Willemsen, A. T., Doorduin, J., de Vries, E. F., Dierckx, R. A., & Leenders,

K. L. (2010). [11C]-PK11195 PET: Quantification of neuroinflammation and a monitor

of anti-inflammatory treatment in parkinson's disease? Parkinsonism & Related

Disorders, 16(1), 57-59.

Basser, P. J. (1995). Inferring microstructural features and the physiological state of tissues

from diffusion-weighted images. NMR in Biomedicine, 8(7-8), 333-344.

Basser, P. J., Mattiello, J., & LeBihan, D. (1994). MR diffusion tensor spectroscopy and

imaging. Biophysical Journal, 66(1), 259-267.

Beaulieu, C. (2002). The basis of anisotropic water diffusion in the nervous system - a

technical review. NMR in Biomedicine, 15(7-8), 435-455.

Beck, A. T., Ward, C. H., Mendelson, M., Mock, J., & Erbaugh, J. (1961). An inventory for

measuring depression. Archives of General Psychiatry, 4, 561-571.

Behrens, T. E., Berg, H. J., Jbabdi, S., Rushworth, M. F., & Woolrich, M. W. (2007).

Probabilistic diffusion tractography with multiple fibre orientations: What can we gain?

NeuroImage, 34(1), 144-155.

Behrens, T. E., Woolrich, M. W., Jenkinson, M., Johansen-Berg, H., Nunes, R. G., Clare, S.,

. . . Smith, S. M. (2003). Characterization and propagation of uncertainty in diffusion-

weighted MR imaging. Magnetic Resonance in Medicine, 50(5), 1077-1088.

Behzadi, Y., Restom, K., Liau, J., & Liu, T. T. (2007). A component based noise correction

method (CompCor) for BOLD and perfusion based fMRI. NeuroImage, 37(1), 90-101.

Bellucci, A., Mercuri, N. B., Venneri, A., Faustini, G., Longhena, F., Pizzi, M., . . . Spano, P.

(2016). Review: Parkinson's disease: From synaptic loss to connectome dysfunction.

Neuropathology and Applied Neurobiology, 42(1), 77-94.

Benavides, J., Quarteronet, D., Imbault, F., Malgouris, C., Uzan, A., Renault, C., . . . Le Fur,

G. (1983). Labelling of "peripheral-type" benzodiazepine binding sites in the rat brain

by using [3H]PK 11195, an isoquinoline carboxamide derivative: Kinetic studies and

autoradiographic localization. Journal of Neurochemistry, 41(6), 1744-1750.

Page 171: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

147

Bencherif, B., Stumpf, M. J., Links, J. M., & Frost, J. J. (2004). Application of MRI-based

partial-volume correction to the analysis of PET images of mu-opioid receptors using

statistical parametric mapping. Journal of Nuclear Medicine : Official Publication,

Society of Nuclear Medicine, 45(3), 402-408.

Bennacef, I. S. C., Horvath, G., Gunn, R., Bonasera, T., Wilson, A., Gee. A., & Laruelle, M.

(2008). Comparison of [11C]PBR28 and [18F]-FEPPA as CNS peripheral

benzodiazepine receptor PET ligands in the pig. J Nucl Med Meeting, 49, 81P-b.

Benskey, M. J., Perez, R. G., & Manfredsson, F. P. (2016). The contribution of alpha

synuclein to neuronal survival and function - implications for parkinson's disease.

Journal of Neurochemistry, 137(3), 331-359.

Benton, A. B., Sivan, K. D., Hamsher, N. R., Varney, N. R., & Spreen, O. (1994).

Contributions to neuropsychological assessment-2nd edition. Orland, FL: Psychological

Assessment Resources.

Berardelli, A., Rothwell, J. C., Thompson, P. D., & Hallett, M. (2001). Pathophysiology of

bradykinesia in parkinson's disease. Brain : A Journal of Neurology, 124(Pt 11), 2131-

2146.

Berg, D., Postuma, R. B., Bloem, B., Chan, P., Dubois, B., Gasser, T., . . . Deuschl, G.

(2014). Time to redefine PD? introductory statement of the MDS task force on the

definition of parkinson's disease. Movement Disorders : Official Journal of the

Movement Disorder Society, 29(4), 454-462.

Beyer, M. K., Janvin, C. C., Larsen, J. P., & Aarsland, D. (2007). A magnetic resonance

imaging study of patients with parkinson's disease with mild cognitive impairment and

dementia using voxel-based morphometry. Journal of Neurology, Neurosurgery, and

Psychiatry, 78(3), 254-259.

Biswal, B., Yetkin, F. Z., Haughton, V. M., & Hyde, J. S. (1995). Functional connectivity in

the motor cortex of resting human brain using echo-planar MRI. Magnetic Resonance in

Medicine, 34(4), 537-541.

Biundo, R., Calabrese, M., Weis, L., Facchini, S., Ricchieri, G., Gallo, P., & Antonini, A.

(2013). Anatomical correlates of cognitive functions in early parkinson's disease

patients. PloS One, 8(5), e64222.

Biundo, R., Formento-Dojot, P., Facchini, S., Vallelunga, A., Ghezzo, L., Foscolo, L., . . .

Antonini, A. (2011). Brain volume changes in parkinson's disease and their relationship

with cognitive and behavioural abnormalities. Journal of the Neurological Sciences,

310(1-2), 64-69.

Bloomfield, P. S., Selvaraj, S., Veronese, M., Rizzo, G., Bertoldo, A., Owen, D. R., . . .

Howes, O. D. (2016). Microglial activity in people at ultra high risk of psychosis and in

Page 172: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

148

schizophrenia: An [(11)C]PBR28 PET brain imaging study. The American Journal of

Psychiatry, 173(1), 44-52.

Blum-Degen, D., Muller, T., Kuhn, W., Gerlach, M., Przuntek, H., & Riederer, P. (1995).

Interleukin-1 beta and interleukin-6 are elevated in the cerebrospinal fluid of alzheimer's

and de novo parkinson's disease patients. Neuroscience Letters, 202(1-2), 17-20.

Bonnelle, V., Ham, T. E., Leech, R., Kinnunen, K. M., Mehta, M. A., Greenwood, R. J., &

Sharp, D. J. (2012). Salience network integrity predicts default mode network function

after traumatic brain injury. Proceedings of the National Academy of Sciences of the

United States of America, 109(12), 4690-4695.

Bonnet, A. M., Jutras, M. F., Czernecki, V., Corvol, J. C., & Vidailhet, M. (2012). Nonmotor

symptoms in parkinson's disease in 2012: Relevant clinical aspects. Parkinson's

Disease, 2012, 198316.

Boutin, H., Chauveau, F., Thominiaux, C., Gregoire, M. C., James, M. L., Trebossen, R., . . .

Kassiou, M. (2007a). 11C-DPA-713: A novel peripheral benzodiazepine receptor PET

ligand for in vivo imaging of neuroinflammation. Journal of Nuclear Medicine : Official

Publication, Society of Nuclear Medicine, 48(4), 573-581.

Boutin, H., Chauveau, F., Thominiaux, C., Kuhnast, B., Gregoire, M. C., Jan, S., . . . Katsifis,

A. (2007b). In vivo imaging of brain lesions with [(11)C]CLINME, a new PET

radioligand of peripheral benzodiazepine receptors. Glia, 55(14), 1459-1468.

Boutin, H., Murray, K., Pradillo, J., Maroy, R., Smigova, A., Gerhard, A., . . . Trigg, W.

(2015). 18F-GE-180: A novel TSPO radiotracer compared to 11C-R-PK11195 in a

preclinical model of stroke. European Journal of Nuclear Medicine and Molecular

Imaging, 42(3), 503-511.

Braak, H., Bohl, J. R., Muller, C. M., Rub, U., de Vos, R. A., & Del Tredici, K. (2006).

Stanley fahn lecture 2005: The staging procedure for the inclusion body pathology

associated with sporadic parkinson's disease reconsidered. Movement Disorders :

Official Journal of the Movement Disorder Society, 21(12), 2042-2051.

Braak, H., & Braak, E. (2000). Pathoanatomy of parkinson's disease. Journal of Neurology,

247 Suppl 2, II3-10.

Braak, H., Braak, E., Yilmazer, D., de Vos, R. A., Jansen, E. N., Bohl, J., & Jellinger, K.

(1994). Amygdala pathology in parkinson's disease. Acta Neuropathologica, 88(6), 493-

500.

Braak, H., Braak, E., Yilmazer, D., Schultz, C., de Vos, R. A., & Jansen, E. N. (1995). Nigral

and extranigral pathology in parkinson's disease. Journal of Neural

Transmission.Supplementum, 46, 15-31.

Page 173: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

149

Braak, H., de Vos, R. A., Jansen, E. N., Bratzke, H., & Braak, E. (1998). Neuropathological

hallmarks of alzheimer's and parkinson's diseases. Progress in Brain Research, 117,

267-285.

Braak, H., & Del Tredici, K. (2009). Neuroanatomy and pathology of sporadic parkinson's

disease. Advances in Anatomy, Embryology, and Cell Biology, 201, 1-119.

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. Neurobiology of

Aging, 24(2), 197-211.

Braak, H., Ghebremedhin, E., Rub, U., Bratzke, H., & Del Tredici, K. (2004). Stages in the

development of parkinson's disease-related pathology. Cell and Tissue Research, 318(1),

121-134.

Braak, H., Rub, U., Jansen Steur, E. N., Del Tredici, K., & de Vos, R. A. (2005). Cognitive

status correlates with neuropathologic stage in parkinson disease. Neurology, 64(8),

1404-1410.

Braak, H., Sastre, M., & Del Tredici, K. (2007). Development of alpha-synuclein

immunoreactive astrocytes in the forebrain parallels stages of intraneuronal pathology in

sporadic parkinson's disease. Acta Neuropathologica, 114(3), 231-241.

Braestrup, C., & Squires, R. F. (1977). Specific benzodiazepine receptors in rat brain

characterized by high-affinity (3H)diazepam binding. Proceedings of the National

Academy of Sciences of the United States of America, 74(9), 3805-3809.

Brenneis, C., Seppi, K., Schocke, M. F., Muller, J., Luginger, E., Bosch, S., . . . Wenning, G.

K. (2003). Voxel-based morphometry detects cortical atrophy in the parkinson variant of

multiple system atrophy. Movement Disorders : Official Journal of the Movement

Disorder Society, 18(10), 1132-1138.

Bribes, E., Carriere, D., Goubet, C., Galiegue, S., Casellas, P., & Simony-Lafontaine, J.

(2004). Immunohistochemical assessment of the peripheral benzodiazepine receptor in

human tissues. The Journal of Histochemistry and Cytochemistry : Official Journal of

the Histochemistry Society, 52(1), 19-28.

Brundin, P., Li, J. Y., Holton, J. L., Lindvall, O., & Revesz, T. (2008). Research in motion:

The enigma of parkinson's disease pathology spread. Nature Reviews.Neuroscience,

9(10), 741-745.

Brundin, P., Melki, R., & Kopito, R. (2010). Prion-like transmission of protein aggregates in

neurodegenerative diseases. Nature Reviews.Molecular Cell Biology, 11(4), 301-307.

Page 174: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

150

Bruno, J., Hosseini, S. M., & Kesler, S. (2012). Altered resting state functional brain network

topology in chemotherapy-treated breast cancer survivors. Neurobiology of Disease,

48(3), 329-338.

Buckner, R. L., Sepulcre, J., Talukdar, T., Krienen, F. M., Liu, H., Hedden, T., . . . Johnson,

K. A. (2009). Cortical hubs revealed by intrinsic functional connectivity: Mapping,

assessment of stability, and relation to alzheimer's disease. The Journal of Neuroscience

: The Official Journal of the Society for Neuroscience, 29(6), 1860-1873.

Buckner, R. L., & Vincent, J. L. (2007). Unrest at rest: Default activity and spontaneous

network correlations. NeuroImage, 37(4), 1091-6; discussion 1097-9.

Bullmore, E., & Sporns, O. (2009). Complex brain networks: Graph theoretical analysis of

structural and functional systems. Nature Reviews.Neuroscience, 10(3), 186-198.

Bullmore, E., & Sporns, O. (2012). The economy of brain network organization. Nature

Reviews.Neuroscience, 13(5), 336-349.

Burton, E. J., McKeith, I. G., Burn, D. J., Williams, E. D., & O'Brien, J. T. (2004). Cerebral

atrophy in parkinson's disease with and without dementia: A comparison with

alzheimer's disease, dementia with lewy bodies and controls. Brain : A Journal of

Neurology, 127(Pt 4), 791-800.

Cabeza, R., Anderson, N. D., Locantore, J. K., & McIntosh, A. R. (2002). Aging gracefully:

Compensatory brain activity in high-performing older adults. NeuroImage, 17(3), 1394-

1402.

Cagnin, A., Brooks, D. J., Kennedy, A. M., Gunn, R. N., Myers, R., Turkheimer, F. E., . . .

Banati, R. B. (2001). In-vivo measurement of activated microglia in dementia. Lancet,

358(9280), 461-467.

Cagnin, A., Rossor, M., Sampson, E. L., Mackinnon, T., & Banati, R. B. (2004). In vivo

detection of microglial activation in frontotemporal dementia. Annals of Neurology,

56(6), 894-897.

Camicioli, R., Gee, M., Bouchard, T. P., Fisher, N. J., Hanstock, C. C., Emery, D. J., &

Martin, W. R. (2009). Voxel-based morphometry reveals extra-nigral atrophy patterns

associated with dopamine refractory cognitive and motor impairment in parkinsonism.

Parkinsonism & Related Disorders, 15(3), 187-195.

Campbell, M. C., Koller, J. M., Snyder, A. Z., Buddhala, C., Kotzbauer, P. T., & Perlmutter,

J. S. (2015). CSF proteins and resting-state functional connectivity in parkinson disease.

Neurology, 84(24), 2413-2421.

Page 175: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

151

Canat, X., Carayon, P., Bouaboula, M., Cahard, D., Shire, D., Roque, C., . . . Casellas, P.

(1993). Distribution profile and properties of peripheral-type benzodiazepine receptors

on human hemopoietic cells. Life Sciences, 52(1), 107-118.

Canu, E., Agosta, F., Sarasso, E., Volontè, M.A., Basaia, S., Stojkovic, T., … Filippi, M.

(2015). Brain structural and functional connectivity in Parkinson's disease with freezing

of gait. Human Brain Mapping, 36(12):5064-78.

Cao, H., Xu, X., Zhao, Y., Long, D., & Zhang, M. (2011). Altered brain activation and

connectivity in early parkinson disease tactile perception. AJNR.American Journal of

Neuroradiology, 32(10), 1969-1974.

Casellas, P., Galiegue, S., & Basile, A. S. (2002). Peripheral benzodiazepine receptors and

mitochondrial function. Neurochemistry International, 40(6), 475-486.

Caviness, J. N., Driver-Dunckley, E., Connor, D. J., Sabbagh, M. N., Hentz, J. G., Noble, B.,

. . . Adler, C. H. (2007). Defining mild cognitive impairment in parkinson's disease.

Movement Disorders : Official Journal of the Movement Disorder Society, 22(9), 1272-

1277.

Cerasa, A., Hagberg, G. E., Peppe, A., Bianciardi, M., Gioia, M. C., Costa, A., . . . Sabatini,

U. (2006). Functional changes in the activity of cerebellum and frontostriatal regions

during externally and internally timed movement in parkinson's disease. Brain Research

Bulletin, 71(1-3), 259-269.

Cerasa, A., Morelli, M., Augimeri, A., Salsone, M., Novellino, F., Gioia, M. C., . . .

Quattrone, A. (2013). Prefrontal thickening in PD with levodopa-induced dyskinesias:

New evidence from cortical thickness measurement. Parkinsonism & Related Disorders,

19(1), 123-125.

Chaki, S., Funakoshi, T., Yoshikawa, R., Okuyama, S., Okubo, T., Nakazato, A., . . .

Tomisawa, K. (1999). Binding characteristics of [3H]DAA1106, a novel and selective

ligand for peripheral benzodiazepine receptors. European Journal of Pharmacology,

371(2-3), 197-204.

Chang, L. J., Yarkoni, T., Khaw, M. W., & Sanfey, A. G. (2013). Decoding the role of the

insula in human cognition: Functional parcellation and large-scale reverse inference.

Cerebral Cortex, 23(3), 739-749.

Chaudhuri, K. R., Odin, P., Antonini, A., & Martinez-Martin, P. (2011). Parkinson's disease:

The non-motor issues. Parkinsonism & Related Disorders, 17(10), 717-723.

Chaudhuri, K. R., & Schapira, A. H. (2009). Non-motor symptoms of parkinson's disease:

Dopaminergic pathophysiology and treatment. The Lancet.Neurology, 8(5), 464-474.

Page 176: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

152

Chauveau, F., Boutin, H., Van Camp, N., Dolle, F., & Tavitian, B. (2008). Nuclear imaging

of neuroinflammation: A comprehensive review of [11C]PK11195 challengers.

European Journal of Nuclear Medicine and Molecular Imaging, 35(12), 2304-2319.

Chen, M. K., Baidoo, K., Verina, T., & Guilarte, T. R. (2004). Peripheral benzodiazepine

receptor imaging in CNS demyelination: Functional implications of anatomical and

cellular localization. Brain : A Journal of Neurology, 127(Pt 6), 1379-1392.

Chen, M. K., & Guilarte, T. R. (2008). Translocator protein 18 kDa (TSPO): Molecular

sensor of brain injury and repair. Pharmacology & Therapeutics, 118(1), 1-17.

Chikama, M., McFarland, N. R., Amaral, D. G., & Haber, S. N. (1997). Insular cortical

projections to functional regions of the striatum correlate with cortical cytoarchitectonic

organization in the primate. The Journal of Neuroscience : The Official Journal of the

Society for Neuroscience, 17(24), 9686-9705.

Chiong, W., Wilson, S. M., D'Esposito, M., Kayser, A. S., Grossman, S. N., Poorzand, P., . .

. Rankin, K. P. (2013). The salience network causally influences default mode network

activity during moral reasoning. Brain : A Journal of Neurology, 136(Pt 6), 1929-1941.

Choi, H. B., Khoo, C., Ryu, J. K., van Breemen, E., Kim, S. U., & McLarnon, J. G. (2002).

Inhibition of lipopolysaccharide-induced cyclooxygenase-2, tumor necrosis factor-alpha

and [Ca2+]i responses in human microglia by the peripheral benzodiazepine receptor

ligand PK11195. Journal of Neurochemistry, 83(3), 546-555.

Christopher, L., Duff-Canning, S., Koshimori, Y., Segura, B., Boileau, I., Chen, R., . . .

Strafella, A. P. (2015). Salience network and parahippocampal dopamine dysfunction in

memory-impaired parkinson disease. Annals of Neurology, 77(2), 269-280.

Christopher, L., Koshimori, Y., Lang, A. E., Criaud, M., & Strafella, A. P. (2014).

Uncovering the role of the insula in non-motor symptoms of parkinson's disease. Brain :

A Journal of Neurology, 137(Pt 8), 2143-2154.

Christopher, L., Marras, C., Duff-Canning, S., Koshimori, Y., Chen, R., Boileau, I., . . .

Strafella, A. P. (2014). Combined insular and striatal dopamine dysfunction are

associated with executive deficits in parkinson's disease with mild cognitive impairment.

Brain : A Journal of Neurology, 137(Pt 2), 565-575.

Christopher, L., & Strafella, A. P. (2013). Neuroimaging of brain changes associated with

cognitive impairment in parkinson's disease. Journal of Neuropsychology, 7(2), 225-

240.

Chung, G. H., Han, Y. M., Jeong, S. H., & Jack, C. R.,Jr. (2005). Functional heterogeneity of

the supplementary motor area. AJNR.American Journal of Neuroradiology, 26(7), 1819-

1823.

Page 177: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

153

Ciurleo, R., Di Lorenzo, G., Bramanti, P., & Marino, S. (2014). Magnetic resonance

spectroscopy: An in vivo molecular imaging biomarker for parkinson's disease? BioMed

Research International, 2014, 519816.

Clement, F., Gauthier, S., & Belleville, S. (2013). Executive functions in mild cognitive

impairment: Emergence and breakdown of neural plasticity. Cortex; a Journal Devoted

to the Study of the Nervous System and Behavior, 49(5), 1268-1279.

Cochrane, C. J., & Ebmeier, K. P. (2013). Diffusion tensor imaging in parkinsonian

syndromes: A systematic review and meta-analysis. Neurology, 80(9), 857-864.

Colasanti, A., Guo, Q., Giannetti, P., Wall, M.B., Newbould, R.D., Bishop, C., … Rabiner,

E.A. (2016). Hippocampal Neuroinflammation, Functional Connectivity, and

Depressive Symptoms in Multiple Sclerosis. Biological Psychiatry, 80(1), 62-72.

Cole, D.M., Beckmann, C.F., Oei, N.Y., Both, S., van Gerven, J.M., Rombouts. S.A. (2013).

Differential and distributed effects of dopamine neuromodulations on resting-state

network connectivity. Neuroimage, 78, 59-67.

Cole, D.M., Oei, N.Y., Soeter, R.P., Both, S., van Gerven, J.M., Rombouts, S.A., Beckmann,

C.F. (2013). Dopamine-dependent architecture of cortico-subcortical network

connectivity. Cereb Cortex, 23(7), 1509-16.

Compta, Y., Ibarretxe-Bilbao, N., Pereira, J. B., Junque, C., Bargallo, N., Tolosa, E., . . .

Marti, M. J. (2012). Grey matter volume correlates of cerebrospinal markers of

alzheimer-pathology in parkinson's disease and related dementia. Parkinsonism &

Related Disorders, 18(8), 941-947.

Compta, Y., Pereira, J. B., Rios, J., Ibarretxe-Bilbao, N., Junque, C., Bargallo, N., . . . Marti,

M. J. (2013). Combined dementia-risk biomarkers in parkinson's disease: A prospective

longitudinal study. Parkinsonism & Related Disorders, 19(8), 717-724.

Corbetta, M., Patel, G., & Shulman, G. L. (2008). The reorienting system of the human

brain: From environment to theory of mind. Neuron, 58(3), 306-324.

Corcia, P., Tauber, C., Vercoullie, J., Arlicot, N., Prunier, C., Praline, J., . . . Ribeiro, M. J.

(2012). Molecular imaging of microglial activation in amyotrophic lateral sclerosis.

PloS One, 7(12), e52941.

Cosenza-Nashat, M., Zhao, M. L., Suh, H. S., Morgan, J., Natividad, R., Morgello, S., &

Lee, S. C. (2009). Expression of the translocator protein of 18 kDa by microglia,

macrophages and astrocytes based on immunohistochemical localization in abnormal

human brain. Neuropathology and Applied Neurobiology, 35(3), 306-328.

Costa, B., Pini, S., Gabelloni, P., Da Pozzo, E., Abelli, M., Lari, L., . . . Martini, C. (2009).

The spontaneous Ala147Thr amino acid substitution within the translocator protein

Page 178: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

154

influences pregnenolone production in lymphomonocytes of healthy individuals.

Endocrinology, 150(12), 5438-5445.

Coughlin, J. M., Wang, Y., Ma, S., Yue, C., Kim, P. K., Adams, A. V., . . . Pomper, M. G.

(2014). Regional brain distribution of translocator protein using [(11)C]DPA-713 PET

in individuals infected with HIV. Journal of Neurovirology, 20(3), 219-232.

Coughlin, J. M., Wang, Y., Munro, C. A., Ma, S., Yue, C., Chen, S., . . . Pomper, M. G.

(2015). Neuroinflammation and brain atrophy in former NFL players: An in vivo

multimodal imaging pilot study. Neurobiology of Disease, 74, 58-65.

Courtney, S. M., Petit, L., Maisog, J. M., Ungerleider, L. G., & Haxby, J. V. (1998). An area

specialized for spatial working memory in human frontal cortex. Science, 279(5355),

1347-1351.

Criaud, M., Christopher, L., Boulinguez, P., Ballanger, B., Lang, A. E., Cho, S. S., . . .

Strafella, A. P. (2016). Contribution of insula in parkinson's disease: A quantitative

meta-analysis study. Human Brain Mapping, 37(4), 1375-1392.

Croisier, E., Moran, L. B., Dexter, D. T., Pearce, R. K., & Graeber, M. B. (2005). Microglial

inflammation in the parkinsonian substantia nigra: Relationship to alpha-synuclein

deposition. Journal of Neuroinflammation, 2, 14.

Crossley, N. A., Mechelli, A., Scott, J., Carletti, F., Fox, P. T., McGuire, P., & Bullmore, E.

T. (2014). The hubs of the human connectome are generally implicated in the anatomy

of brain disorders. Brain : A Journal of Neurology, 137(Pt 8), 2382-2395.

Crossley, N. A., Mechelli, A., Vertes, P. E., Winton-Brown, T. T., Patel, A. X., Ginestet, C.

E., . . . Bullmore, E. T. (2013). Cognitive relevance of the community structure of the

human brain functional coactivation network. Proceedings of the National Academy of

Sciences of the United States of America, 110(28), 11583-11588.

Cubon, V. A., Putukian, M., Boyer, C., & Dettwiler, A. (2011). A diffusion tensor imaging

study on the white matter skeleton in individuals with sports-related concussion. Journal

of Neurotrauma, 28(2), 189-201.

Czeh, M., Gressens, P., & Kaindl, A. M. (2011). The yin and yang of microglia.

Developmental Neuroscience, 33(3-4), 199-209.

Dale, A. M., Fischl, B., & Sereno, M. I. (1999). Cortical surface-based analysis. I.

segmentation and surface reconstruction. NeuroImage, 9(2), 179-194.

Dale, A. M., & Sereno, M. I. (1993). Improved localizadon of cortical activity by combining

EEG and MEG with MRI cortical surface reconstruction: A linear approach. Journal of

Cognitive Neuroscience, 5(2), 162-176.

Page 179: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

155

Damani, M. R., Zhao, L., Fontainhas, A. M., Amaral, J., Fariss, R. N., & Wong, W. T.

(2011). Age-related alterations in the dynamic behavior of microglia. Aging Cell, 10(2),

263-276.

Davis, S. W., Dennis, N. A., Daselaar, S. M., Fleck, M. S., & Cabeza, R. (2008). Que PASA?

the posterior-anterior shift in aging. Cerebral Cortex, 18(5), 1201-1209.

de Pasquale, F., Della Penna, S., Sporns, O., Romani, G. L., & Corbetta, M. (2015). A

dynamic core network and global efficiency in the resting human brain. Cerebral

Cortex, Sep 6.

de Pasquale, F., Sabatini, U., Della Penna, S., Sestieri, C., Caravasso, C. F., Formisano, R., &

Peran, P. (2013). The connectivity of functional cores reveals different degrees of

segregation and integration in the brain at rest. NeuroImage, 69, 51-61.

de Vries, F. E., de Wit, S. J., Cath, D. C., van der Werf, Y. D., van der Borden, V., van

Rossum, T. B., . . . van den Heuvel, O. A. (2014). Compensatory frontoparietal activity

during working memory: An endophenotype of obsessive-compulsive disorder.

Biological Psychiatry, 76(11), 878-887.

Defer, G. L., Widner, H., Marie, R. M., Remy, P., & Levivier, M. (1999). Core assessment

program for surgical interventional therapies in parkinson's disease (CAPSIT-PD).

Movement Disorders : Official Journal of the Movement Disorder Society, 14(4), 572-

584.

Delis, D. C., Kaplan, E., & Kramer, J. H. (2001). Examiner's manual for the delis-kaplan

executive function system. San Antonio, TX: The Psychological Corporation.

Delis, D. C., Kramer, J. H., Kaplan, B. A., & Ober, B. A. (2002). Manual for the california

verbal learning test-second edition. San Antonio TX: The psychological Corportaion.

Della-Maggiore, V., Sekuler, A. B., Grady, C. L., Bennett, P. J., Sekuler, R., & McIntosh, A.

R. (2000). Corticolimbic interactions associated with performance on a short-term

memory task are modified by age. The Journal of Neuroscience : The Official Journal of

the Society for Neuroscience, 20(22), 8410-8416.

DeLong, M. R., & Wichmann, T. (2015). Basal ganglia circuits as targets for

neuromodulation in parkinson disease. JAMA Neurology, 72(11), 1354-1360.

Deng, B., Zhang, Y., Wang, L., Peng, K., Han, L., Nie, K., . . . Wang, J. (2013). Diffusion

tensor imaging reveals white matter changes associated with cognitive status in patients

with parkinson's disease. American Journal of Alzheimer's Disease and Other

Dementias, 28(2), 154-164.

Desai Bradaric, B., Patel, A., Schneider, J. A., Carvey, P. M., & Hendey, B. (2012).

Evidence for angiogenesis in parkinson's disease, incidental lewy body disease, and

Page 180: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

156

progressive supranuclear palsy. Journal of Neural Transmission (Vienna, Austria :

1996), 119(1), 59-71.

Desgranges, B., Baron, J. C., & Eustache, F. (1998). The functional neuroanatomy of

episodic memory: The role of the frontal lobes, the hippocampal formation, and other

areas. NeuroImage, 8(2), 198-213.

Deshpande, M., Zheng, J., Borgmann, K., Persidsky, R., Wu, L., Schellpeper, C., &

Ghorpade, A. (2005). Role of activated astrocytes in neuronal damage: Potential links to

HIV-1-associated dementia. Neurotoxicity Research, 7(3), 183-192.

Desplats, P., Lee, H. J., Bae, E. J., Patrick, C., Rockenstein, E., Crews, L., . . . Lee, S. J.

(2009). Inclusion formation and neuronal cell death through neuron-to-neuron

transmission of alpha-synuclein. Proceedings of the National Academy of Sciences of

the United States of America, 106(31), 13010-13015.

Dexter, D. T., & Jenner, P. (2013). Parkinson disease: From pathology to molecular disease

mechanisms. Free Radical Biology & Medicine, 62, 132-144.

Dilger, R. N., & Johnson, R. W. (2008). Aging, microglial cell priming, and the discordant

central inflammatory response to signals from the peripheral immune system. Journal of

Leukocyte Biology, 84(4), 932-939.

Dobbs, R. J., Charlett, A., Purkiss, A. G., Dobbs, S. M., Weller, C., & Peterson, D. W.

(1999). Association of circulating TNF-alpha and IL-6 with ageing and parkinsonism.

Acta Neurologica Scandinavica, 100(1), 34-41.

Doble, A., Malgouris, C., Daniel, M., Daniel, N., Imbault, F., Basbaum, A., . . . Le Fur, G.

(1987). Labelling of peripheral-type benzodiazepine binding sites in human brain with

[3H]PK 11195: Anatomical and subcellular distribution. Brain Research Bulletin, 18(1),

49-61.

Doorduin, J., Klein, H. C., Dierckx, R. A., James, M., Kassiou, M., & de Vries, E. F. (2009).

11C]-DPA-713 and [18F]-DPA-714 as new PET tracers for TSPO: A comparison with

[11C]-(R)-PK11195 in a rat model of herpes encephalitis. Molecular Imaging and

Biology : MIB : The Official Publication of the Academy of Molecular Imaging, 11(6),

386-398.

Doorn, K. J., Goudriaan, A., Blits-Huizinga, C., Bol, J. G., Rozemuller, A. J., Hoogland, P.

V., . . . van Dam, A. M. (2014a). Increased amoeboid microglial density in the olfactory

bulb of parkinson's and alzheimer's patients. Brain Pathology, 24(2), 152-165.

Doorn, K. J., Moors, T., Drukarch, B., van de Berg, W. D., Lucassen, P. J., & van Dam, A.

M. (2014b). Microglial phenotypes and toll-like receptor 2 in the substantia nigra and

hippocampus of incidental lewy body disease cases and parkinson's disease patients.

Acta Neuropathologica Communications, 2, 90-014-0090-1.

Page 181: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

157

Dosenbach, N. U., Fair, D. A., Cohen, A. L., Schlaggar, B. L., & Petersen, S.E. (2008). A

dual-networks architecture of top-down control. Trendsin Cognitive Sciences, 12(3), 99-

105.

Dosenbach, N. U., Fair, D. A., Miezin, F. M., Cohen, A. L., Wenger, K. K., Dosenbach, R.

A., . . . Petersen, S. E. (2007). Distinct brain networks for adaptive and stable task

control in humans. Proceedings of the National Academy of Sciences of the United

States of America, 104(26), 11073-11078.

Dosenbach, N. U., Nardos, B., Cohen, A. L., Fair, D. A., Power, J. D., Church, J. A., . . .

Schlaggar, B. L. (2010). Prediction of individual brain maturity using fMRI. Science,

329(5997), 1358-1361.

du Boisgueheneuc, F., Levy, R., Volle, E., Seassau, M., Duffau, H., Kinkingnehun, S., . . .

Dubois, B. (2006). Functions of the left superior frontal gyrus in humans: A lesion

study. Brain : A Journal of Neurology, 129(Pt 12), 3315-3328.

Duncan, G. W., Firbank, M. J., O'Brien, J. T., & Burn, D. J. (2013). Magnetic resonance

imaging: A biomarker for cognitive impairment in parkinson's disease? Movement

Disorders : Official Journal of the Movement Disorder Society, 28(4), 425-438.

Edison, P., Ahmed, I., Fan, Z., Hinz, R., Gelosa, G., Ray Chaudhuri, K., . . . Brooks, D. J.

(2013). Microglia, amyloid, and glucose metabolism in parkinson's disease with and

without dementia. Neuropsychopharmacology : Official Publication of the American

College of Neuropsychopharmacology, 38(6), 938-949.

Endres, C. J., Pomper, M. G., James, M., Uzuner, O., Hammoud, D. A., Watkins, C. C., . . .

Kassiou, M. (2009). Initial evaluation of 11C-DPA-713, a novel TSPO PET ligand, in

humans. Journal of Nuclear Medicine : Official Publication, Society of Nuclear

Medicine, 50(8), 1276-1282.

Eng, L. F., Ghirnikar, R. S., & Lee, Y. L. (2000). Glial fibrillary acidic protein: GFAP-thirty-

one years (1969-2000). Neurochemical Research, 25(9-10), 1439-1451.

Engel, A. K., Gerloff, C., Hilgetag, C. C., & Nolte, G. (2013). Intrinsic coupling modes:

Multiscale interactions in ongoing brain activity. Neuron, 80(4), 867-886.

Erlandsson, K., Buvat, I., Pretorius, P. H., Thomas, B. A., & Hutton, B. F. (2012). A review

of partial volume correction techniques for emission tomography and their applications

in neurology, cardiology and oncology. Physics in Medicine and Biology, 57(21), R119-

59.

Esposito, F., Tessitore, A., Giordano, A., De Micco, R., Paccone, A., Conforti, R., …

Tedeschi, G. (2013). Rhythm-specific modulation of the sensorimotor network in

drug-naive patients with Parkinson's disease by levodopa. Brain,136(Pt 3):710-725.

Evans, A. H., Katzenschlager, R., Paviour, D., O'Sullivan, J. D., Appel, S., Lawrence, A. D.,

Page 182: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

158

& Lees, A. J. (2004). Punding in parkinson's disease: Its relation to the dopamine

dysregulation syndrome. Movement Disorders : Official Journal of the Movement

Disorder Society, 19(4), 397-405.

Fan, J., Lindemann, P., Feuilloley, M. G., & Papadopoulos, V. (2012). Structural and

functional evolution of the translocator protein (18 kDa). Current Molecular Medicine,

12(4), 369-386.

Farina, E., Gattellaro, G., Pomati, S., Magni, E., Perretti, A., Cannata, A. P., . . . Mariani, C.

(2000). Researching a differential impairment of frontal functions and explicit memory

in early parkinson's disease. European Journal of Neurology, 7(3), 259-267.

Feldmann, A., Illes, Z., Kosztolanyi, P., Illes, E., Mike, A., Kover, F., . . . Nagy, F. (2008).

Morphometric changes of gray matter in parkinson's disease with depression: A voxel-

based morphometry study. Movement Disorders : Official Journal of the Movement

Disorder Society, 23(1), 42-46.

Femminella, G. D., Ninan, S., Atkinson, R., Fan, Z., Brooks, D. J., & Edison, P. (2016).

Does microglial activation influence hippocampal volume and neuronal function in

alzheimer's disease and parkinson's disease dementia? Journal of Alzheimer's Disease :

JAD, 51(4), 1275-1289.

Ferrer, I., Martinez, A., Blanco, R., Dalfo, E., & Carmona, M. (2011). Neuropathology of

sporadic parkinson disease before the appearance of parkinsonism: Preclinical parkinson

disease. Journal of Neural Transmission, 118(5), 821-839.

Finch, C. E. (2003). Neurons, glia, and plasticity in normal brain aging. Neurobiology of

Aging, 24 Suppl 1, S123-7; discussion S131.

Fischl, B., & Dale, A. M. (2000). Measuring the thickness of the human cerebral cortex from

magnetic resonance images. Proceedings of the National Academy of Sciences of the

United States of America, 97(20), 11050-11055.

Fischl, B., Liu, A., & Dale, A. M. (2001). Automated manifold surgery: Constructing

geometrically accurate and topologically correct models of the human cerebral cortex.

IEEE Transactions on Medical Imaging, 20(1), 70-80.

Floresco, S. B., & Magyar, O. (2006). Mesocortical dopamine modulation of executive

functions: Beyond working memory. Psychopharmacology, 188(4), 567-585.

Fogelson, N., Li, L., Li, Y., Fernandez-Del-Olmo, M., Santos-Garcia, D., & Peled, A.

(2013). Functional connectivity abnormalities during contextual processing in

schizophrenia and in parkinson's disease. Brain and Cognition, 82(3), 243-253.

Foley, P., & Riederer, P. (1999). Pathogenesis and preclinical course of parkinson's disease.

Journal of Neural Transmission.Supplementum, 56, 31-74.

Page 183: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

159

Fornito, A., Zalesky, A., & Breakspear, M. (2015). The connectomics of brain disorders.

Nature Reviews.Neuroscience, 16(3), 159-172.

Freeman, M. R. (2010). Specification and morphogenesis of astrocytes. Science, 330(6005),

774-778.

Friston, K. J., Frith, C. D., Liddle, P. F., & Frackowiak, R. S. (1993). Functional

connectivity: The principal-component analysis of large (PET) data sets. Journal of

Cerebral Blood Flow and Metabolism : Official Journal of the International Society of

Cerebral Blood Flow and Metabolism, 13(1), 5-14.

Friston, K. J., Harrison, L., & Penny, W. (2003). Dynamic causal modelling. NeuroImage,

19(4), 1273-1302.

Fujimura, Y., Ikoma, Y., Yasuno, F., Suhara, T., Ota, M., Matsumoto, R., . . . Ito, H. (2006).

Quantitative analyses of 18F-FEDAA1106 binding to peripheral benzodiazepine

receptors in living human brain. Journal of Nuclear Medicine : Official Publication,

Society of Nuclear Medicine, 47(1), 43-50.

Fujimura, Y., Zoghbi, S. S., Simeon, F. G., Taku, A., Pike, V. W., Innis, R. B., & Fujita, M.

(2009). Quantification of translocator protein (18 kDa) in the human brain with PET and

a novel radioligand, (18)F-PBR06. Journal of Nuclear Medicine : Official Publication,

Society of Nuclear Medicine, 50(7), 1047-1053.

Fujita, M., Imaizumi, M., Zoghbi, S. S., Fujimura, Y., Farris, A. G., Suhara, T., . . . Innis, R.

B. (2008). Kinetic analysis in healthy humans of a novel positron emission tomography

radioligand to image the peripheral benzodiazepine receptor, a potential biomarker for

inflammation. NeuroImage, 40(1), 43-52.

Gallagher, C., Bell, B., Bendlin, B., Palotti, M., Okonkwo, O., Sodhi, A., . . . Alexander, A.

(2013). White matter microstructural integrity and executive function in parkinson's

disease. Journal of the International Neuropsychological Society : JINS, 19(3), 349-354.

Galvan, A., Devergnas, A., & Wichmann, T. (2015). Alterations in neuronal activity in basal

ganglia-thalamocortical circuits in the parkinsonian state. Frontiers in Neuroanatomy, 9,

5.

Gao, H. M., & Hong, J. S. (2008). Why neurodegenerative diseases are progressive:

Uncontrolled inflammation drives disease progression. Trends in Immunology, 29(8),

357-365.

Garnier, M., Dimchev, A. B., Boujrad, N., Price, J. M., Musto, N. A., & Papadopoulos, V.

(1994). In vitro reconstitution of a functional peripheral-type benzodiazepine receptor

from mouse leydig tumor cells. Molecular Pharmacology, 45(2), 201-211.

Page 184: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

160

Gatliff, J., & Campanella, M. (2012). The 18 kDa translocator protein (TSPO): A new

perspective in mitochondrial biology. Current Molecular Medicine, 12(4), 356-368.

Gattellaro, G., Minati, L., Grisoli, M., Mariani, C., Carella, F., Osio, M., . . . Bruzzone, M. G.

(2009). White matter involvement in idiopathic parkinson disease: A diffusion tensor

imaging study. AJNR.American Journal of Neuroradiology, 30(6), 1222-1226.

Gerhard, A., Neumaier, B., Elitok, E., Glatting, G., Ries, V., Tomczak, R., . . . Reske, S. N.

(2000). In vivo imaging of activated microglia using [11C]PK11195 and positron

emission tomography in patients after ischemic stroke. Neuroreport, 11(13), 2957-2960.

Gerhard, A., Pavese, N., Hotton, G., Turkheimer, F., Es, M., Hammers, A., . . . Brooks, D. J.

(2006). In vivo imaging of microglial activation with [11C](R)-PK11195 PET in

idiopathic parkinson's disease. Neurobiology of Disease, 21(2), 404-412.

Gerrits, N. J., van der Werf, Y. D., Verhoef, K. M., Veltman, D. J., Groenewegen, H. J.,

Berendse, H. W., & van den Heuvel, O. A. (2015). Compensatory fronto-parietal

hyperactivation during set-shifting in unmedicated patients with parkinson's disease.

Neuropsychologia, 68, 107-116.

Gerrits, N. J., van Loenhoud A.C., van den Berg, S. F., Berendse, H. W., Foncke, E. M.,

Klein, M., … van den Heuvel, O. A. (2016). Cortical thickness. Surface are and

subcoritical volume differntially contribute to cognitive heterogeneity in Parkinson's

disease. PLoS One, 11(2), e0148852.

Glass, C. K., Saijo, K., Winner, B., Marchetto, M. C., & Gage, F. H. (2010). Mechanisms

underlying inflammation in neurodegeneration. Cell, 140(6), 918-934.

Goedert, M., Spillantini, M. G., Del Tredici, K., & Braak, H. (2013). 100 years of lewy

pathology. Nature Reviews.Neurology, 9(1), 13-24.

Goldman, J. G., Stebbins, G. T., Bernard, B., Stoub, T. R., Goetz, C. G., & deToledo-

Morrell, L. (2012). Entorhinal cortex atrophy differentiates parkinson's disease patients

with and without dementia. Movement Disorders : Official Journal of the Movement

Disorder Society, 27(6), 727-734.

Golla, S. S., Boellaard, R., Oikonen, V., Hoffmann, A., van Berckel, B. N., Windhorst, A.

D., . . . Rinne, J. O. (2015). Quantification of [18F]DPA-714 binding in the human

brain: Initial studies in healthy controls and alzheimer's disease patients. Journal of

Cerebral Blood Flow and Metabolism : Official Journal of the International Society of

Cerebral Blood Flow and Metabolism, 35(5), 766-772.

Gong, G., Rosa-Neto, P., Carbonell, F., Chen, Z. J., He, Y., & Evans, A. C. (2009). Age- and

gender-related differences in the cortical anatomical network. The Journal of

Neuroscience : The Official Journal of the Society for Neuroscience, 29(50), 15684-

15693.

Page 185: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

161

Gottlich, M., Munte, T. F., Heldmann, M., Kasten, M., Hagenah, J., & Kramer, U. M.

(2013). Altered resting state brain networks in parkinson's disease. PloS One, 8(10),

e77336.

Grady, C. (2012). The cognitive neuroscience of ageing. Nature Reviews.Neuroscience,

13(7), 491-505.

Grady, C. L. (2008). Cognitive neuroscience of aging. Annals of the New York Academy of

Sciences, 1124, 127-144.

Graeber, M. B., Bise, K., & Mehraein, P. (1994). CR3/43, a marker for activated human

microglia: Application to diagnostic neuropathology. Neuropathology and Applied

Neurobiology, 20(4), 406-408.

Graeber, M. B., & Streit, W. J. (2010). Microglia: Biology and pathology. Acta

Neuropathologica, 119(1), 89-105.

Gratwicke, J., Jahanshahi, M., & Foltynie, T. (2015). Parkinson's disease dementia: A neural

networks perspective. Brain : A Journal of Neurology, 138(Pt 6), 1454-1476.

Greicius, M. (2008). Resting-state functional connectivity in neuropsychiatric disorders.

Current Opinion in Neurology, 21(4), 424-430.

Griffith, H. R., den Hollander, J. A., Okonkwo, O. C., O'Brien, T., Watts, R. L., & Marson,

D. C. (2008). Brain metabolism differs in alzheimer's disease and parkinson's disease

dementia. Alzheimer's & Dementia : The Journal of the Alzheimer's Association, 4(6),

421-427.

Griffith, H. R., Okonkwo, O. C., O'Brien, T., & Hollander, J. A. (2008). Reduced brain

glutamate in patients with parkinson's disease. NMR in Biomedicine, 21(4), 381-387.

Guidotti, A., Forchetti, C. M., Corda, M. G., Konkel, D., Bennett, C. D., & Costa, E. (1983).

Isolation, characterization, and purification to homogeneity of an endogenous

polypeptide with agonistic action on benzodiazepine receptors. Proceedings of the

National Academy of Sciences of the United States of America, 80(11), 3531-3535.

Guimera, R., & Amaral, L. A. (2005). Cartography of complex networks: Modules and

universal roles. Journal of Statistical Mechanics, 2005(P02001), nihpa35573.

Gulani, V., & Sundgren, P. C. (2006). Diffusion tensor magnetic resonance imaging. Journal

of Neuro-Ophthalmology : The Official Journal of the North American Neuro-

Ophthalmology Society, 26(1), 51-60.

Gulyas, B., Vas, A., Toth, M., Takano, A., Varrone, A., Cselenyi, Z., . . . Halldin, C. (2011).

Age and disease related changes in the translocator protein (TSPO) system in the human

Page 186: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

162

brain: Positron emission tomography measurements with [11C]vinpocetine.

NeuroImage, 56(3), 1111-1121.

Guo, Q., Colasanti, A., Owen, D. R., Onega, M., Kamalakaran, A., Bennacef, I., . . . Gunn,

R. N. (2013). Quantification of the specific translocator protein signal of 18F-PBR111

in healthy humans: A genetic polymorphism effect on in vivo binding. Journal of

Nuclear Medicine : Official Publication, Society of Nuclear Medicine, 54(11), 1915-

1923.

Halliday, G. M., & Stevens, C. H. (2011). Glia: Initiators and progressors of pathology in

parkinson's disease. Movement Disorders : Official Journal of the Movement Disorder

Society, 26(1), 6-17.

Hamby, M. E., & Sofroniew, M. V. (2010). Reactive astrocytes as therapeutic targets for

CNS disorders. Neurotherapeutics : The Journal of the American Society for

Experimental NeuroTherapeutics, 7(4), 494-506.

Hammoud, D. A., Endres, C. J., Chander, A. R., Guilarte, T. R., Wong, D. F., Sacktor, N. C.,

. . . Pomper, M. G. (2005). Imaging glial cell activation with [11C]-R-PK11195 in

patients with AIDS. Journal of Neurovirology, 11(4), 346-355.

Hanganu, A., Bedetti, C., Jubault, T., Gagnon, J. F., Mejia-Constain, B., Degroot, C., . . .

Monchi, O. (2013). Mild cognitive impairment in patients with parkinson's disease is

associated with increased cortical degeneration. Movement Disorders : Official Journal

of the Movement Disorder Society, 28(10), 1360-1369.

Hansen, C., Angot, E., Bergstrom, A. L., Steiner, J. A., Pieri, L., Paul, G., . . . Brundin, P.

(2011). Alpha-synuclein propagates from mouse brain to grafted dopaminergic neurons

and seeds aggregation in cultured human cells. The Journal of Clinical Investigation,

121(2), 715-725.

Haslinger, B., Erhard, P., Kampfe, N., Boecker, H., Rummeny, E., Schwaiger, M., . . .

Ceballos-Baumann, A. O. (2001). Event-related functional magnetic resonance imaging

in parkinson's disease before and after levodopa. Brain : A Journal of Neurology, 124(Pt

3), 558-570.

Hatano, K., Yamada, T., Toyama, H., Kudo, G., Nomura, M., Suzuki, H., . . . Ito, K. (2010).

Correlation between FEPPA uptake and microglia activation in 6-OHDA injured rat

brain. NeuroImage, 52, S138-S.

Hattori, T., Orimo, S., Aoki, S., Ito, K., Abe, O., Amano, A., . . . Mizusawa, H. (2012).

Cognitive status correlates with white matter alteration in parkinson's disease. Human

Brain Mapping, 33(3), 727-739.

Hawkes, C. H., Del Tredici, K., & Braak, H. (2009). Parkinson's disease: The dual hit theory

revisited. Annals of the New York Academy of Sciences, 1170, 615-622.

Page 187: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

163

Heeger, D. J., & Ress, D. (2002). What does fMRI tell us about neuronal activity? Nature

Reviews.Neuroscience, 3(2), 142-151.

Helmich, R. C., Derikx, L. C., Bakker, M., Scheeringa, R., Bloem, B. R., & Toni, I. (2010).

Spatial remapping of cortico-striatal connectivity in parkinson's disease. Cerebral

Cortex, 20(5), 1175-1186.

Hickman, S. E., Allison, E. K., & El Khoury, J. (2008). Microglial dysfunction and defective

beta-amyloid clearance pathways in aging alzheimer's disease mice. The Journal of

Neuroscience : The Official Journal of the Society for Neuroscience, 28(33), 8354-8360.

Hirsch, E. C., & Hunot, S. (2009). Neuroinflammation in parkinson's disease: A target for

neuroprotection? Lancet Neurology, 8(4), 382-397.

Honey, C. J., Sporns, O., Cammoun, L., Gigandet, X., Thiran, J. P., Meuli, R., & Hagmann,

P. (2009). Predicting human resting-state functional connectivity from structural

connectivity. Proceedings of the National Academy of Sciences of the United States of

America, 106(6), 2035-2040.

Hong, J. Y., Lee, J. E., Sohn, Y. H., & Lee, P. H. (2012). Neurocognitive and atrophic

patterns in parkinson's disease based on subjective memory complaints. Journal of

Neurology, 259(8), 1706-1712.

Horsfield, M. A., Larsson, H. B., Jones, D. K., & Gass, A. (1998). Diffusion magnetic

resonance imaging in multiple sclerosis. Journal of Neurology, Neurosurgery, and

Psychiatry, 64 Suppl 1, S80-4.

Hosseini, S. M., Hoeft, F., & Kesler, S. R. (2012). GAT: A graph-theoretical analysis

toolbox for analyzing between-group differences in large-scale structural and functional

brain networks. PloS One, 7(7), e40709.

Houlden, H., & Singleton, A. B. (2012). The genetics and neuropathology of parkinson's

disease. Acta Neuropathologica, 124(3), 325-338.

Howlett, D. R., Whitfield, D., Johnson, M., Attems, J., O'Brien, J. T., Aarsland, D., . . .

Francis, P. T. (2015). Regional multiple pathology scores are associated with cognitive

decline in lewy body dementias. Brain Pathology (Zurich, Switzerland), 25(4), 401-408.

Hu, M. T., White, S. J., Chaudhuri, K. R., Morris, R. G., Bydder, G. M., & Brooks, D. J.

(2001). Correlating rates of cerebral atrophy in parkinson's disease with measures of

cognitive decline. Journal of Neural Transmission, 108(5), 571-580.

Hua, K., Zhang, J., Wakana, S., Jiang, H., Li, X., Reich, D. S., . . . Mori, S. (2008). Tract

probability maps in stereotaxic spaces: Analyses of white matter anatomy and tract-

specific quantification. NeuroImage, 39(1), 336-347.

Page 188: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

164

Huang, P., Lou, Y., Xuan, M., Gu, Q., Guan, X., Xu, X., et al., … Zhang, M. (2016). Cortical

abnormalities in Parkinson's disease patients and relationship to depression: A surface-

based morphometry study. Psychiatry Reserch: Neuroimaging, 250, 24-28.

Huang, X., Alonso, A., Guo, X., Umbach, D. M., Lichtenstein, M. L., Ballantyne, C. M., . . .

Chen, H. (2015). Statins, plasma cholesterol, and risk of parkinson's disease: A

prospective study. Movement Disorders : Official Journal of the Movement Disorder

Society, 30(4), 552-559.

Huppi, P. S., Maier, S. E., Peled, S., Zientara, G. P., Barnes, P. D., Jolesz, F. A., & Volpe, J.

J. (1998). Microstructural development of human newborn cerebral white matter

assessed in vivo by diffusion tensor magnetic resonance imaging. Pediatric Research,

44(4), 584-590.

Iannaccone, S., Cerami, C., Alessio, M., Garibotto, V., Panzacchi, A., Olivieri, S., . . . Perani,

D. (2013). In vivo microglia activation in very early dementia with lewy bodies,

comparison with parkinson's disease. Parkinsonism & Related Disorders, 19(1), 47-52.

Ibarretxe-Bilbao, N., Junque, C., Marti, M. J., Valldeoriola, F., Vendrell, P., Bargallo, N., . . .

Tolosa, E. (2010). Olfactory impairment in parkinson's disease and white matter

abnormalities in central olfactory areas: A voxel-based diffusion tensor imaging study.

Movement Disorders : Official Journal of the Movement Disorder Society, 25(12),

1888-1894.

Ibarretxe-Bilbao, N., Junque, C., Segura, B., Baggio, H. C., Marti, M. J., Valldeoriola, F., . . .

Tolosa, E. (2012). Progression of cortical thinning in early parkinson's disease.

Movement Disorders : Official Journal of the Movement Disorder Society, 27(14),

1746-1753.

Ibarretxe-Bilbao, N., Junque, C., Tolosa, E., Marti, M. J., Valldeoriola, F., Bargallo, N., &

Zarei, M. (2009). Neuroanatomical correlates of impaired decision-making and facial

emotion recognition in early parkinson's disease. The European Journal of

Neuroscience, 30(6), 1162-1171.

Ibarretxe-Bilbao, N., Ramirez-Ruiz, B., Junque, C., Marti, M. J., Valldeoriola, F., Bargallo,

N., . . . Tolosa, E. (2010). Differential progression of brain atrophy in parkinson's

disease with and without visual hallucinations. Journal of Neurology, Neurosurgery,

and Psychiatry, 81(6), 650-657.

Ikeda, A., Yazawa, S., Kunieda, T., Ohara, S., Terada, K., Mikuni, N., . . . Shibasaki, H.

(1999). Cognitive motor control in human pre-supplementary motor area studied by

subdural recording of discrimination/selection-related potentials. Brain : A Journal of

Neurology, 122(Pt5), 915-931.

Page 189: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

165

Imaizumi, M., Briard, E., Zoghbi, S. S., Gourley, J. P., Hong, J., Musachio, J. L., . . . Fujita,

M. (2007a). Kinetic evaluation in nonhuman primates of two new PET ligands for

peripheral benzodiazepine receptors in brain. Synapse, 61(8), 595-605.

Imaizumi, M., Kim, H. J., Zoghbi, S. S., Briard, E., Hong, J., Musachio, J. L., . . . Fujita, M.

(2007b). PET imaging with [11C]PBR28 can localize and quantify upregulated

peripheral benzodiazepine receptors associated with cerebral ischemia in rat.

Neuroscience Letters, 411(3), 200-205.

Imamura, K., Hishikawa, N., Sawada, M., Nagatsu, T., Yoshida, M., & Hashizume, Y.

(2003). Distribution of major histocompatibility complex class II-positive microglia and

cytokine profile of parkinson's disease brains. Acta Neuropathologica, 106(6), 518-526.

Lindqvist, D., Hall, S., Surova, Y., Henrietta, M., Nielsen, M., Janelidze, S…. Hansson.

(2013). Cerebrospinal fluid inflammatory markers in Parkinson's disease - associations

with depression, fatigue, and cognitive impairment. Brain, Behavior, and Immunity, 33,

183-189.

Innis, R. B., Cunningham, V. J., Delforge, J., Fujita, M., Gjedde, A., Gunn, R. N., . . .

Carson, R. E. (2007). Consensus nomenclature for in vivo imaging of reversibly binding

radioligands. Journal of Cerebral Blood Flow and Metabolism : Official Journal of the

International Society of Cerebral Blood Flow and Metabolism, 27(9), 1533-1539.

International Parkinson Disease Genomics Consortium, Nalls, M. A., Plagnol, V.,

Hernandez, D. G., Sharma, M., Sheerin, U. M., . . . Wood, N. W. (2011). Imputation of

sequence variants for identification of genetic risks for parkinson's disease: A meta-

analysis of genome-wide association studies. Lancet, 377(9766), 641-649.

Iwai, A., Masliah, E., Yoshimoto, M., Ge, N., Flanagan, L., de Silva, H. A., . . . Saitoh, T.

(1995). The precursor protein of non-A beta component of alzheimer's disease amyloid

is a presynaptic protein of the central nervous system. Neuron, 14(2), 467-475.

Iwata, K., & Harano, H. (1986). Regional cerebral blood flow changes in aging. Acta

Radiologica.Supplementum, 369, 440-443.

James, M. L., Fulton, R. R., Vercoullie, J., Henderson, D. J., Garreau, L., Chalon, S., . . .

Kassiou, M. (2008). DPA-714, a new translocator protein-specific ligand: Synthesis,

radiofluorination, and pharmacologic characterization. Journal of Nuclear Medicine :

Official Publication, Society of Nuclear Medicine, 49(5), 814-822.

James, M. L., Selleri, S., & Kassiou, M. (2006). Development of ligands for the peripheral

benzodiazepine receptor. Current Medicinal Chemistry, 13(17), 1991-2001.

Jankovic, J. (2008). Parkinson's disease: Clinical features and diagnosis. Journal of

Neurology, Neurosurgery, and Psychiatry, 79(4), 368-376.

Page 190: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

166

Janvin, C. C., Larsen, J. P., Aarsland, D., & Hugdahl, K. (2006). Subtypes of mild cognitive

impairment in parkinson's disease: Progression to dementia. Movement Disorders :

Official Journal of the Movement Disorder Society, 21(9), 1343-1349.

Jellinger, K. A. (1991). Pathology of parkinson's disease. changes other than the nigrostriatal

pathway. Molecular and Chemical Neuropathology / Sponsored by the International

Society for Neurochemistry and the World Federation of Neurology and Research

Groups on Neurochemistry and Cerebrospinal Fluid, 14(3), 153-197.

Jellinger, K. A. (2012). Neurobiology of cognitive impairment in parkinson's disease. Expert

Review of Neurotherapeutics, 12(12), 1451-1466.

Jenkinson, M., Bannister, P., Brady, M., & Smith, S. (2002). Improved optimization for the

robust and accurate linear registration and motion correction of brain images.

NeuroImage, 17(2), 825-841.

Jenkinson, M., & Smith, S. (2001). A global optimisation method for robust affine

registration of brain images. Medical Image Analysis, 5(2), 143-156.

Johansen-Berg, H. & Rushworth, M.F. (2009). Using diffusion imaging to study human

connectional anatomy. Annual Review Neuroscience, 32, 75-94.

Jones, D. K., Knosche, T. R., & Turner, R. (2013). White matter integrity, fiber count, and

other fallacies: The do's and don'ts of diffusion MRI. NeuroImage, 73, 239-254.

Jubault, T., Brambati, S. M., Degroot, C., Kullmann, B., Strafella, A. P., Lafontaine, A. L., . .

. Monchi, O. (2009). Regional brain stem atrophy in idiopathic parkinson's disease

detected by anatomical MRI. PloS One, 4(12), e8247.

Jubault, T., Gagnon, J. F., Karama, S., Ptito, A., Lafontaine, A. L., Evans, A. C., & Monchi,

O. (2011). Patterns of cortical thickness and surface area in early parkinson's disease.

NeuroImage, 55(2), 462-467.

Kaiser, M., & Hilgetag, C. C. (2006). Nonoptimal component placement, but short

processing paths, due to long-distance projections in neural systems. PLoS

Computational Biology, 2(7), e95.

Kashani, A., Betancur, C., Giros, B., Hirsch, E., & El Mestikawy, S. (2007). Altered

expression of vesicular glutamate transporters VGLUT1 and VGLUT2 in parkinson

disease. Neurobiology of Aging, 28(4), 568-578.

Katsuse, O., Iseki, E., Marui, W., & Kosaka, K. (2003). Developmental stages of cortical

lewy bodies and their relation to axonal transport blockage in brains of patients with

dementia with lewy bodies. Journal of the Neurological Sciences, 211(1-2), 29-35.

Page 191: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

167

Kehagia, A. A., Barker, R. A., & Robbins, T. W. (2010). Neuropsychological and clinical

heterogeneity of cognitive impairment and dementia in patients with parkinson's disease.

The Lancet.Neurology, 9(12), 1200-1213.

Kenk, M., Selvanathan, T., Rao, N., Suridjan, I., Rusjan, P., Remington, G., . . . Mizrahi, R.

(2015). Imaging neuroinflammation in gray and white matter in schizophrenia: An in-

vivo PET study with [18F]-FEPPA. Schizophrenia Bulletin, 41(1), 85-93.

Khandelwal, P. J., Herman, A. M., & Moussa, C. E. (2011). Inflammation in the early stages

of neurodegenerative pathology. Journal of Neuroimmunology, 238(1-2), 1-11.

Kim, H. J., Kim, S. J., Kim, H. S., Choi, C. G., Kim, N., Han, S., . . . Lee, C. S. (2013).

Alterations of mean diffusivity in brain white matter and deep gray matter in parkinson's

disease. Neuroscience Letters, 550, 64-68.

Kim, J. S., Yang, J. J., Lee, J. M., Youn, J., Kim, J. M., & Cho, J. W. (2014). Topographic

pattern of cortical thinning with consideration of motor laterality in parkinson disease.

Parkinsonism & Related Disorders, 20(11), 1186-1190.

Ko, J. H., Koshimori, Y., Mizrahi, R., Rusjan, P., Wilson, A. A., Lang, A. E., . . . Strafella,

A. P. (2013). Voxel-based imaging of translocator protein 18 kDa (TSPO) in high-

resolution PET. Journal of Cerebral Blood Flow and Metabolism : Official Journal of

the International Society of Cerebral Blood Flow and Metabolism, 33(3), 348-350.

Kobylecki, C., Counsell, S. J., Cabanel, N., Wachter, T., Turkheimer, F. E., Eggert, K., . . .

Gerhard, A. (2013). Diffusion-weighted imaging and its relationship to microglial

activation in parkinsonian syndromes. Parkinsonism & Related Disorders, 19(5), 527-

532.

Kofler, J., & Wiley, C. A. (2011). Microglia: Key innate immune cells of the brain.

Toxicologic Pathology, 39(1), 103-114.

Kordower, J. H., Chu, Y., Hauser, R. A., Freeman, T. B., & Olanow, C. W. (2008). Lewy

body-like pathology in long-term embryonic nigral transplants in parkinson's disease.

Nature Medicine, 14(5), 504-506.

Kordower, J. H., Chu, Y., Hauser, R. A., Olanow, C. W., & Freeman, T. B. (2008).

Transplanted dopaminergic neurons develop PD pathologic changes: A second case

report. Movement Disorders : Official Journal of the Movement Disorder Society,

23(16), 2303-2306.

Koshimori, Y., Ko, J. H., Mizrahi, R., Rusjan, P., Mabrouk, R., Jacobs, M. F., . . . Strafella,

A. P. (2015). Imaging striatal microglial activation in patients with parkinson's disease.

PloS One, 10(9), e0138721.

Page 192: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

168

Koshimori, Y., Segura, B., Christopher, L., Lobaugh, N., Duff-Canning, S., Mizrahi, R., . . .

Strafella, A. P. (2015). Imaging changes associated with cognitive abnormalities in

parkinson's disease. Brain Structure & Function, 220(4), 2249-2261.

Kostic, V. S., Agosta, F., Petrovic, I., Galantucci, S., Spica, V., Jecmenica-Lukic, M., &

Filippi, M. (2010). Regional patterns of brain tissue loss associated with depression in

parkinson disease. Neurology, 75(10), 857-863.

Krack, P., Hariz, M. I., Baunez, C., Guridi, J., & Obeso, J. A. (2010). Deep brain stimulation:

From neurology to psychiatry? Trends in Neurosciences, 33(10), 474-484.

Kreisl, W. C., Fujita, M., Fujimura, Y., Kimura, N., Jenko, K. J., Kannan, P., . . . Innis, R. B.

(2010). Comparison of [(11)C]-(R)-PK 11195 and [(11)C]PBR28, two radioligands for

translocator protein (18 kDa) in human and monkey: Implications for positron emission

tomographic imaging of this inflammation biomarker. NeuroImage, 49(4), 2924-2932.

Kreisl, W. C., Jenko, K. J., Hines, C. S., Lyoo, C. H., Corona, W., Morse, C. L., . . .

Biomarkers Consortium PET Radioligand Project Team. (2013). A genetic

polymorphism for translocator protein 18 kDa affects both in vitro and in vivo

radioligand binding in human brain to this putative biomarker of neuroinflammation.

Journal of Cerebral Blood Flow and Metabolism : Official Journal of the International

Society of Cerebral Blood Flow and Metabolism, 33(1), 53-58.

Kropholler, M. A., Boellaard, R., Schuitemaker, A., Folkersma, H., van Berckel, B. N., &

Lammertsma, A. A. (2006). Evaluation of reference tissue models for the analysis of

[11C](R)-PK11195 studies. Journal of Cerebral Blood Flow and Metabolism : Official

Journal of the International Society of Cerebral Blood Flow and Metabolism, 26(11),

1431-1441.

Krystal, J. H., D'Souza, D. C., Mathalon, D., Perry, E., Belger, A., & Hoffman, R. (2003).

NMDA receptor antagonist effects, cortical glutamatergic function, and schizophrenia:

Toward a paradigm shift in medication development. Psychopharmacology, 169(3-4),

215-233.

Kuhlmann, A. C., & Guilarte, T. R. (2000). Cellular and subcellular localization of

peripheral benzodiazepine receptors after trimethyltin neurotoxicity. Journal of

Neurochemistry, 74(4), 1694-1704.

Kulisevsky, J., García-Sánchez, C., Berthier, M.L., Barbanoj, M., Pascual-Sedano, B.,

Gironell, A., & Estévez-González, A. (2000). Chronic effects of dopaminergic

replacement on cognitive function in Parkinson's disease: a two-year follow-up study

of previously untreated patients. Movement Disorders : Official Journal of the

Movement Disorder Society, 15(4), 613-626.

Page 193: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

169

Kurth, F., Zilles, K., Fox, P. T., Laird, A. R., & Eickhoff, S. B. (2010). A link between the

systems: Functional differentiation and integration within the human insula revealed by

meta-analysis. Brain Structure & Function, 214(5-6), 519-534.

Lahiri, D. K., & Nurnberger, J. I.,Jr. (1991). A rapid non-enzymatic method for the

preparation of HMW DNA from blood for RFLP studies. Nucleic Acids Research,

19(19), 5444.

Lammertsma, A. A., & Hume, S. P. (1996). Simplified reference tissue model for PET

receptor studies. NeuroImage, 4(3 Pt 1), 153-158.

Lang, A. E., & Lozano, A. M. (1998). Parkinson's disease. first of two parts. The New

England Journal of Medicine, 339(15), 1044-1053.

Langston, J. W. (2006). The parkinson's complex: Parkinsonism is just the tip of the iceberg.

Annals of Neurology, 59(4), 591-596.

Langston, J. W., Widner, H., Goetz, C. G., Brooks, D., Fahn, S., Freeman, T., & Watts, R.

(1992). Core assessment program for intracerebral transplantations (CAPIT). Movement

Disorders : Official Journal of the Movement Disorder Society, 7(1), 2-13.

Lanoue, A. C., Dumitriu, A., Myers, R. H., & Soghomonian, J. J. (2010). Decreased glutamic

acid decarboxylase mRNA expression in prefrontal cortex in parkinson's disease.

Experimental Neurology, 226(1), 207-217.

Latora, V., & Marchiori, M. (2001). Efficient behavior of small-world networks. Physical

Review Letters, 87(19), 198701.

Lavisse, S., Garcia-Lorenzo, D., Peyronneau, M. A., Bodini, B., Thiriez, C., Kuhnast, B., . . .

Bottlaender, M. (2015). Optimized quantification of translocator protein radioligand

18F-DPA-714 uptake in the brain of genotyped healthy volunteers. Journal of Nuclear

Medicine : Official Publication, Society of Nuclear Medicine, 56(7), 1048-1054.

Lavisse, S., Guillermier, M., Herard, A. S., Petit, F., Delahaye, M., Van Camp, N., . . .

Escartin, C. (2012). Reactive astrocytes overexpress TSPO and are detected by TSPO

positron emission tomography imaging. The Journal of Neuroscience : The Official

Journal of the Society for Neuroscience, 32(32), 10809-10818.

Le Bihan, D. (2003). Looking into the functional architecture of the brain with diffusion

MRI. Nature Reviews.Neuroscience, 4(6), 469-480.

Le Bihan, D., Mangin, J. F., Poupon, C., Clark, C. A., Pappata, S., Molko, N., & Chabriat, H.

(2001). Diffusion tensor imaging: Concepts and applications. Journal of Magnetic

Resonance Imaging : JMRI, 13(4), 534-546.

Page 194: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

170

Le Bihan, D., Poupon, C., Amadon, A., & Lethimonnier, F. (2006). Artifacts and pitfalls in

diffusion MRI. Journal of Magnetic Resonance Imaging : JMRI, 24(3), 478-488.

Lebedev, A. V., Westman, E., Simmons, A., Lebedeva, A., Siepel, F. J., Pereira, J. B., &

Aarsland, D. (2014). Large-scale resting state network correlates of cognitive

impairment in parkinson's disease and related dopaminergic deficits. Frontiers in

Systems Neuroscience, 8, 45.

Lee, H. J., Bae, E. J., & Lee, S. J. (2014). Extracellular alpha--synuclein-a novel and crucial

factor in lewy body diseases. Nature Reviews.Neurology, 10(2), 92-98.

Lee, H. J., Kim, C., & Lee, S. J. (2010). Alpha-synuclein stimulation of astrocytes: Potential

role for neuroinflammation and neuroprotection. Oxidative Medicine and Cellular

Longevity, 3(4), 283-287.

Lee, H. J., Suk, J. E., Bae, E. J., & Lee, S. J. (2008). Clearance and deposition of

extracellular alpha-synuclein aggregates in microglia. Biochemical and Biophysical

Research Communications, 372(3), 423-428.

Lee, J. Y., & Jeon, B. S. (2014). Maladaptive reward-learning and impulse control disorders

in patients with parkinson's disease: A clinical overview and pathophysiology update.

Journal of Movement Disorders, 7(2), 67-76.

Lee, S., Varvel, N. H., Konerth, M. E., Xu, G., Cardona, A. E., Ransohoff, R. M., & Lamb,

B. T. (2010). CX3CR1 deficiency alters microglial activation and reduces beta-amyloid

deposition in two alzheimer's disease mouse models. The American Journal of

Pathology, 177(5), 2549-2562.

Levy, R., & Goldman-Rakic, P. S. (2000). Segregation of working memory functions within

the dorsolateral prefrontal cortex. Experimental Brain Research.Experimentelle

Hirnforschung.Experimentation Cerebrale, 133(1), 23-32.

Lewis, D. A., Pierri, J. N., Volk, D. W., Melchitzky, D. S., & Woo, T. U. (1999). Altered

GABA neurotransmission and prefrontal cortical dysfunction in schizophrenia.

Biological Psychiatry, 46(5), 616-626.

Li, J. Y., Englund, E., Holton, J. L., Soulet, D., Hagell, P., Lees, A. J., . . . Brundin, P.

(2008). Lewy bodies in grafted neurons in subjects with parkinson's disease suggest

host-to-graft disease propagation. Nature Medicine, 14(5), 501-503.

Li, J. Y., Englund, E., Widner, H., Rehncrona, S., Bjorklund, A., Lindvall, O., & Brundin, P.

(2010). Characterization of lewy body pathology in 12- and 16-year-old intrastriatal

mesencephalic grafts surviving in a patient with parkinson's disease. Movement

Disorders : Official Journal of the Movement Disorder Society, 25(8), 1091-1096.

Page 195: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

171

Li, W., Qin, W., Liu, H., Fan, L., Wang, J., Jiang, T., & Yu, C. (2013). Subregions of the

human superior frontal gyrus and their connections. NeuroImage, 78, 46-58.

Li, Y., Liang, P., Jia, X., & Li, K. (2016). Abnormal regional homogeneity in parkinson's

disease: A resting state fMRI study. Clinical Radiology, 71(1), e28-34.

Liang, X., Zou, Q., He, Y., & Yang, Y. (2013). Coupling of functional connectivity and

regional cerebral blood flow reveals a physiological basis for network hubs of the

human brain. Proceedings of the National Academy of Sciences of the United States of

America, 110(5), 1929-1934.

Litvan, I., Goldman, J. G., Troster, A. I., Schmand, B. A., Weintraub, D., Petersen, R. C., . . .

Emre, M. (2012). Diagnostic criteria for mild cognitive impairment in parkinson's

disease: Movement disorder society task force guidelines. Movement Disorders :

Official Journal of the Movement Disorder Society, 27(3), 349-356.

Liu, B., Le, K. X., Park, M. A., Wang, S., Belanger, A. P., Dubey, S., . . . Lemere, C. A.

(2015). In vivo detection of age- and disease-related increases in neuroinflammation by

18F-GE180 TSPO MicroPET imaging in wild-type and alzheimer's transgenic mice. The

Journal of Neuroscience : The Official Journal of the Society for Neuroscience, 35(47),

15716-15730.

Liu, W., Tang, Y., & Feng, J. (2011). Cross talk between activation of microglia and

astrocytes in pathological conditions in the central nervous system. Life Sciences, 89(5-

6), 141-146.

Logothetis, N. K., Pauls, J., Augath, M., Trinath, T., & Oeltermann, A. (2001).

Neurophysiological investigation of the basis of the fMRI signal. Nature, 412(6843),

150-157.

Lull, M. E., & Block, M. L. (2010). Microglial activation and chronic neurodegeneration.

Neurotherapeutics : The Journal of the American Society for Experimental

NeuroTherapeutics, 7(4), 354-365.

Luo, C. Y., Guo, X. Y., Song, W., Chen, Q., Cao, B., Yang, J., . . . Shang, H. F. (2015).

Functional connectome assessed using graph theory in drug-naive parkinson's disease.

Journal of Neurology, 262(6), 1557-1567.

Lyoo, C. H., Ryu, Y. H., & Lee, M. S. (2010). Topographical distribution of cerebral cortical

thinning in patients with mild parkinson's disease without dementia. Movement

Disorders : Official Journal of the Movement Disorder Society, 25(4), 496-499.

Lyoo, C. H., Ryu, Y. H., & Lee, M. S. (2011). Cerebral cortical areas in which thickness

correlates with severity of motor deficits of parkinson's disease. Journal of Neurology,

258(10), 1871-1876.

Page 196: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

172

Ma, Q., Huang, B., Wang, J., Seger, C., Yang, W., Li, C., . . . Huang, R. (2016). Altered

modular organization of intrinsic brain functional networks in patients with parkinson's

disease. Brain Imaging and Behavior, Feb 10.

Mabrouk, R., Rusjan, P. M., Mizrahi, R., Jacobs, M. F., Koshimori, Y., Houle, S., . . .

Strafella, A. P. (2014). Image derived input function for [18F]-FEPPA: Application to

quantify translocator protein (18 kDa) in the human brain. PloS One, 9(12), e115768.

MacDonald, A. A., Monchi, O., Seergobin, K. N., Ganjavi, H., Tamjeedi, R., & MacDonald,

P. A. (2013). Parkinson's disease duration determines effect of dopaminergic therapy on

ventral striatum function. Movement Disorders : Official Journal of the Movement

Disorder Society, 28(2), 153-160.

Macuga, K. L., & Frey, S. H. (2011). Selective responses in right inferior frontal and

supramarginal gyri differentiate between observed movements of oneself vs. another.

Neuropsychologia, 49(5), 1202-1207.

Madden, D. J., Turkington, T. G., Provenzale, J. M., Denny, L. L., Hawk, T. C., Gottlob, L.

R., & Coleman, R. E. (1999). Adult age differences in the functional neuroanatomy of

verbal recognition memory. Human Brain Mapping, 7(2), 115-135.

Madhyastha, T. M., Askren, M. K., Zhang, J., Leverenz, J. B., Montine, T. J., & Grabowski,

T. J. (2015). Group comparison of spatiotemporal dynamics of intrinsic networks in

parkinson's disease. Brain : A Journal of Neurology, 138(Pt 9), 2672-2686.

Maeda, J., Suhara, T., Zhang, M. R., Okauchi, T., Yasuno, F., Ikoma, Y., . . . Suzuki, K.

(2004). Novel peripheral benzodiazepine receptor ligand [11C]DAA1106 for PET: An

imaging tool for glial cells in the brain. Synapse, 52(4), 283-291.

Malpass, K. (2011). MRI may predict the onset of alzheimer disease. Nature

Reviews.Neurology, 7(6), 302.

Matsui, H., Nishinaka, K., Oda, M., Niikawa, H., Komatsu, K., Kubori, T., & Udaka, F.

(2007). Depression in parkinson's disease. diffusion tensor imaging study. Journal of

Neurology, 254(9), 1170-1173.

Matsui, H., Nishinaka, K., Oda, M., Niikawa, H., Kubori, T., & Udaka, F. (2007). Dementia

in parkinson's disease: Diffusion tensor imaging. Acta Neurologica Scandinavica,

116(3), 177-181.

McColgan, P., Seunarine, K. K., Razi, A., Cole, J. H., Gregory, S., Durr, A., . . . Track-HD

Investigators. (2015). Selective vulnerability of rich club brain regions is an

organizational principle of structural connectivity loss in huntington's disease. Brain : A

Journal of Neurology, 138(Pt 11), 3327-3344.

Page 197: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

173

McGeer, P. L., Itagaki, M. D., Boyes, B. E., & McGeer, E. G. (1988). Reactive microglia are

positiver for HLA-DR in the substantia nigra of parkinson's and alzheimer's disease

brains. Neurology, 38, 1285-1291.

Medran-Navarrete, V., Bernards, N., Kuhnast, B., Damont, A., Pottier, G., Peyronneau, M.

A., . . . Dolle, F. (2014). 18F]DPA-C5yne, a novel fluorine-18-labelled analogue of

DPA-714: Radiosynthesis and preliminary evaluation as a radiotracer for imaging

neuroinflammation with PET. Journal of Labelled Compounds &

Radiopharmaceuticals, 57(6), 410-418.

Melzer, T. R., Watts, R., MacAskill, M. R., Pitcher, T. L., Livingston, L., Keenan, R. J., . . .

Anderson, T. J. (2012). Grey matter atrophy in cognitively impaired parkinson's disease.

Journal of Neurology, Neurosurgery, and Psychiatry, 83(2), 188-194.

Melzer, T. R., Watts, R., Macaskill, M. R., Pitcher, T. L., Livingston, L., Keenan, R. J., . . .

Anderson, T. J. (2013). White matter microstructure deteriorates across cognitive stages

in parkinson disease. Neurology, 80(20), 1841-1849.

Menon, V., & Uddin, L. Q. (2010). Saliency, switching, attention and control: A network

model of insula function. Brain Structure & Function, 214(5-6), 655-667.

Messina, D., Cerasa, A., Condino, F., Arabia, G., Novellino, F., Nicoletti, G., . . . Quattrone,

A. (2011). Patterns of brain atrophy in parkinson's disease, progressive supranuclear

palsy and multiple system atrophy. Parkinsonism & Related Disorders, 17(3), 172-176.

Mesulam, M. M. (1998). From sensation to cognition. Brain : A Journal of Neurology, 121

(Pt 6), 1013-1052.

Meunier, D., Lambiotte, R., & Bullmore, E. T. (2010). Modular and hierarchically modular

organization of brain networks. Frontiers in Neuroscience, 4, 200.

Mirza, B., Hadberg, H., Thomsen, P., & Moos, T. (2000). The absence of reactive

astrocytosis is indicative of a unique inflammatory process in parkinson's disease.

Neuroscience, 95(2), 425-432.

Mittelbronn, M., Dietz, K., Schluesener, H. J., & Meyermann, R. (2001). Local distribution

of microglia in the normal adult human central nervous system differs by up to one order

of magnitude. Acta Neuropathologica, 101(3), 249-255.

Mizrahi, R., Rusjan, P. M., Kennedy, J., Pollock, B., Mulsant, B., Suridjan, I., . . . Houle, S.

(2012). Translocator protein (18 kDa) polymorphism (rs6971) explains in-vivo brain

binding affinity of the PET radioligand [(18)F]-FEPPA. Journal of Cerebral Blood Flow

and Metabolism : Official Journal of the International Society of Cerebral Blood Flow

and Metabolism, 32(6), 968-972.

Page 198: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

174

Mogi, M., Harada, M., Kondo, T., Riederer, P., Inagaki, H., Minami, M., & Nagatsu, T.

(1994). Interleukin-1 beta, interleukin-6, epidermal growth factor and transforming

growth factor-alpha are elevated in the brain from parkinsonian patients. Neuroscience

Letters, 180(2), 147-150.

Mori, S., Wakana, S., Nagae-Poetscher, L. M., & van Zijl, P. C. M. (2005). MRI atlas of

human white matter. Amsterdam, The Netherlands: Elsevier.

Mori, S., Crain, B. J., Chacko, V. P., & van Zijl, P. C. (1999). Three-dimensional tracking of

axonal projections in the brain by magnetic resonance imaging. Annals of Neurology,

45(2), 265-269.

Morrison, J. H., & Hof, P. R. (1997). Life and death of neurons in the aging brain. Science,

278(5337), 412-419.

Mowinckel, A. M., Espeseth, T., & Westlye, L. T. (2012). Network-specific effects of age

and in-scanner subject motion: A resting-state fMRI study of 238 healthy adults.

NeuroImage, 63(3), 1364-1373.

Mukherjee, P., Berman, J. I., Chung, S. W., Hess, C. P., & Henry, R. G. (2008). Diffusion

tensor MR imaging and fiber tractography: Theoretic underpinnings. AJNR.American

Journal of Neuroradiology, 29(4), 632-641.

Mukherjee, P., Miller, J. H., Shimony, J. S., Conturo, T. E., Lee, B. C., Almli, C. R., &

McKinstry, R. C. (2001). Normal brain maturation during childhood: Developmental

trends characterized with diffusion-tensor MR imaging. Radiology, 221(2), 349-358.

Muller, T., Blum-Degen, D., Przuntek, H., & Kuhn, W. (1998). Interleukin-6 levels in

cerebrospinal fluid inversely correlate to severity of parkinson's disease. Acta

Neurologica Scandinavica, 98(2), 142-144.

Muzerengi, S., Contrafatto, D., & Chaudhuri, K. R. (2007). Non-motor symptoms:

Identification and management. Parkinsonism & Related Disorders, 13 Suppl 3, S450-6.

Mythri, R. B., Venkateshappa, C., Harish, G., Mahadevan, A., Muthane, U. B., Yasha, T. C.,

. . . Shankar, S. K. (2011). Evaluation of markers of oxidative stress, antioxidant

function and astrocytic proliferation in the striatum and frontal cortex of parkinson's

disease brains. Neurochemical Research, 36(8), 1452-1463.

Nagano-Saito, A., Habak, C., Mejia-Constain, B., Degroot, C., Monetta, L., Jubault, T., . . .

Monchi, O. (2013). Effect of mild cognitive impairment on the patterns of neural

activity in early parkinson's disease. Neurobiology of Aging, 35(1), 223-231.

Nagano-Saito, A., Martinu, K., & Monchi, O. (2014). Function of basal ganglia in bridging

cognitive and motor modules to perform an action. Frontiers in Neuroscience, 8, 187.

Page 199: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

175

Nagatsu, T., & Sawada, M. (2005). Inflammatory process in parkinson's disease: Role for

cytokines. Current Pharmaceutical Design, 11(8), 999-1016.

Narendran, R., Lopresti, B. J., Mason, N. S., Deuitch, L., Paris, J., Himes, M. L., . . .

Nimgaonkar, V. L. (2014). Cocaine abuse in humans is not associated with increased

microglial activation: An 18-kDa translocator protein positron emission tomography

imaging study with [11C]PBR28. The Journal of Neuroscience : The Official Journal of

the Society for Neuroscience, 34(30), 9945-9950.

Nasreddine, Z. S., Phillips, N. A., Bedirian, V., Charbonneau, S., Whitehead, V., Collin, I., . .

. Chertkow, H. (2005). The montreal cognitive assessment, MoCA: A brief screening

tool for mild cognitive impairment. Journal of the American Geriatrics Society, 53(4),

695-699.

Neil, J. J. (2008). Diffusion imaging concepts for clinicians. Journal of Magnetic Resonance

Imaging : JMRI, 27(1), 1-7.

Newman, M. E. (2006). Modularity and community structure in networks. Proceedings of

the National Academy of Sciences of the United States of America, 103(23), 8577-8582.

Nichols, N. R., Day, J. R., Laping, N. J., Johnson, S. A., & Finch, C. E. (1993). GFAP

mRNA increases with age in rat and human brain. Neurobiology of Aging, 14(5), 421-

429.

Nigro, S., Riccelli, R., Passamonti, L., Arabia, G., Morelli, M., Nistico, R., …Quattrone, A.

(2016). Characterizing structural neural networks in de novo Parkinson Disease patients

using diffusion tensor imaging. Human Brain Mapping, in press.

Norden, D. M., & Godbout, J. P. (2013). Review: Microglia of the aged brain: Primed to be

activated and resistant to regulation. Neuropathology and Applied Neurobiology, 39(1),

19-34.

Nucifora, P. G., Verma, R., Lee, S. K., & Melhem, E. R. (2007). Diffusion-tensor MR

imaging and tractography: Exploring brain microstructure and connectivity. Radiology,

245(2), 367-384.

Oades, R. D., & Halliday, G. M. (1987). Ventral tegmental (A10) system: Neurobiology. 1.

anatomy and connectivity. Brain Research, 434(2), 117-165.

Obeso, J. A., & Lanciego, J. L. (2011). Past, present, and future of the pathophysiological

model of the basal ganglia. Frontiers in Neuroanatomy, 5, 39.

Ogawa, S., & Lee, T. M. (1990). Magnetic resonance imaging of blood vessels at high fields:

In vivo and in vitro measurements and image simulation. Magnetic Resonance in

Medicine, 16(1), 9-18.

Page 200: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

176

Okubo, T., Yoshikawa, R., Chaki, S., Okuyama, S., & Nakazato, A. (2004). Design,

synthesis, and structure-activity relationships of novel tetracyclic compounds as

peripheral benzodiazepine receptor ligands. Bioorganic & Medicinal Chemistry, 12(13),

3569-3580.

Olde Dubbelink, K. T., Hillebrand, A., Stoffers, D., Deijen, J. B., Twisk, J. W., Stam, C. J.,

& Berendse, H. W. (2014). Disrupted brain network topology in parkinson's disease: A

longitudinal magnetoencephalography study. Brain : A Journal of Neurology, 137(Pt 1),

197-207.

Ouchi, Y., Yagi, S., Yokokura, M., & Sakamoto, M. (2009). Neuroinflammation in the living

brain of parkinson's disease. Parkinsonism & Related Disorders, 15 Suppl 3, S200-4.

Ouchi, Y., Yoshikawa, E., Sekine, Y., Futatsubashi, M., Kanno, T., Ogusu, T., & Torizuka,

T. (2005). Microglial activation and dopamine terminal loss in early parkinson's disease.

Annals of Neurology, 57(2), 168-175.

Owen, A. M. (2004). Cognitive dysfunction in parkinson's disease: The role of frontostriatal

circuitry. The Neuroscientist : A Review Journal Bringing Neurobiology, Neurology and

Psychiatry, 10(6), 525-537.

Owen, A. M., Stern, C. E., Look, R. B., Tracey, I., Rosen, B. R., & Petrides, M. (1998).

Functional organization of spatial and nonspatial working memory processing within the

human lateral frontal cortex. Proceedings of the National Academy of Sciences of the

United States of America, 95(13), 7721-7726.

Owen, D. R., Gunn, R. N., Rabiner, E. A., Bennacef, I., Fujita, M., Kreisl, W. C., . . . Parker,

C. A. (2011). Mixed-affinity binding in humans with 18-kDa translocator protein

ligands. Journal of Nuclear Medicine : Official Publication, Society of Nuclear

Medicine, 52(1), 24-32.

Owen, D. R., Guo, Q., Kalk, N. J., Colasanti, A., Kalogiannopoulou, D., Dimber, R., . . .

Rabiner, E. A. (2014). Determination of [(11)C]PBR28 binding potential in vivo: A first

human TSPO blocking study. Journal of Cerebral Blood Flow and Metabolism :

Official Journal of the International Society of Cerebral Blood Flow and Metabolism,

34(6), 989-994.

Owen, D. R., Howell, O. W., Tang, S. P., Wells, L. A., Bennacef, I., Bergstrom, M., . . .

Parker, C. A. (2010). Two binding sites for [3H]PBR28 in human brain: Implications for

TSPO PET imaging of neuroinflammation. Journal of Cerebral Blood Flow and

Metabolism : Official Journal of the International Society of Cerebral Blood Flow and

Metabolism, 30(9), 1608-1618.

Owen, D. R., Yeo, A. J., Gunn, R. N., Song, K., Wadsworth, G., Lewis, A., . . . Rubio, J. P.

(2012). An 18-kDa translocator protein (TSPO) polymorphism explains differences in

binding affinity of the PET radioligand PBR28. Journal of Cerebral Blood Flow and

Page 201: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

177

Metabolism : Official Journal of the International Society of Cerebral Blood Flow and

Metabolism, 32(1), 1-5.

Pagonabarraga, J., & Kulisevsky, J. (2012). Cognitive impairment and dementia in

parkinson's disease. Neurobiology of Disease, 46(3), 590-596.

Pagonabarraga, J., Kulisevsky, J., Strafella, A. P., & Krack, P. (2015). Apathy in parkinson's

disease: Clinical features, neural substrates, diagnosis, and treatment. The

Lancet.Neurology, 14(5), 518-531.

Pan, P. L., Song, W., & Shang, H. F. (2012). Voxel-wise meta-analysis of gray matter

abnormalities in idiopathic parkinson's disease. European Journal of Neurology : The

Official Journal of the European Federation of Neurological Societies, 19(2), 199-206.

Paolicelli, R. C., Bolasco, G., Pagani, F., Maggi, L., Scianni, M., Panzanelli, P., . . . Gross, C.

T. (2011). Synaptic pruning by microglia is necessary for normal brain development.

Science (New York, N.Y.), 333(6048), 1456-1458.

Papadopoulos, V., Amri, H., Boujrad, N., Cascio, C., Culty, M., Garnier, M., . . . Drieu, K.

(1997). Peripheral benzodiazepine receptor in cholesterol transport and steroidogenesis.

Steroids, 62(1), 21-28.

Papadopoulos, V., Baraldi, M., Guilarte, T. R., Knudsen, T. B., Lacapere, J. J., Lindemann,

P., . . . Gavish, M. (2006). Translocator protein (18kDa): New nomenclature for the

peripheral-type benzodiazepine receptor based on its structure and molecular function.

Trends in Pharmacological Sciences, 27(8), 402-409.

Pappata, S., Cornu, P., Samson, Y., Prenant, C., Benavides, J., Scatton, B., . . . Syrota, A.

(1991). PET study of carbon-11-PK 11195 binding to peripheral type benzodiazepine

sites in glioblastoma: A case report. Journal of Nuclear Medicine : Official Publication,

Society of Nuclear Medicine, 32(8), 1608-1610.

Parente, A., Feltes, P. K., Vallez Garcia, D., Sijbesma, J. W., Moriguchi Jeckel, C. M.,

Dierckx, R. A., . . . Doorduin, J. (2016). Pharmacokinetic analysis of 11C-PBR28 in the

rat model of herpes encephalitis: Comparison with (R)-11C-PK11195. Journal of

Nuclear Medicine : Official Publication, Society of Nuclear Medicine, 57(5), 785-791.

Parker, G. J., & Alexander, D. C. (2005). Probabilistic anatomical connectivity derived from

the microscopic persistent angular structure of cerebral tissue. Philosophical

Transactions of the Royal Society of London.Series B, Biological Sciences, 360(1457),

893-902.

Passingham, R. E., Bengtsson, S. L., & Lau, H. C. (2010). Medial frontal cortex: From self-

generated action to reflection on one's own performance. Trends in Cognitive Sciences,

14(1), 16-21.

Page 202: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

178

Pellicano, C., Assogna, F., Piras, F., Caltagirone, C., Pontieri, F. E., & Spalletta, G. (2012).

Regional cortical thickness and cognitive functions in non-demented parkinson's disease

patients: A pilot study. European Journal of Neurology : The Official Journal of the

European Federation of Neurological Societies, 19(1), 172-175.

Pereira, J. B., Aarsland, D., Ginestet, C. E., Lebedev, A.V., Wahlund L., Simmons, A., …

Westman, E. (2015). Aberrant cerebral network topology and mild cognitive impairment

in early Parkinson's disease. Human Brain Mapping, 36(8), 2980-2995.

Pereira, J. B., Ibarretxe-Bilbao, N., Marti, M. J., Compta, Y., Junque, C., Bargallo, N., &

Tolosa, E. (2012). Assessment of cortical degeneration in patients with parkinson's

disease by voxel-based morphometry, cortical folding, and cortical thickness. Human

Brain Mapping, 33(11), 2521-2534.

Petit-Taboue, M. C., Baron, J. C., Barre, L., Travere, J. M., Speckel, D., Camsonne, R., &

MacKenzie, E. T. (1991). Brain kinetics and specific binding of [11C]PK 11195 to

omega 3 sites in baboons: Positron emission tomography study. European Journal of

Pharmacology, 200(2-3), 347-351.

Picard, N., & Strick, P. L. (2001). Imaging the premotor areas. Current Opinion in

Neurobiology, 11(6), 663-672.

Pierpaoli, C., & Basser, P. J. (1996). Toward a quantitative assessment of diffusion

anisotropy. Magnetic Resonance in Medicine, 36(6), 893-906.

Pinto, S., Mancini, L., Jahanshahi, M., Thornton, J.S., Tripoliti, E., Yousry, T.A., Limousin,

P. (2010). Functional magnetic resonance imaging exploration of combined hand and

speech movements in Parkinson's disease. Movement Disorders : Official Journal of the

Movement Disorder Society, 26(12), 2212-2219.

Poletti, M., & Bonuccelli, U. (2013). Acute and chronic cognitive effects of levodopa and

dopamine agonists on patients with parkinson's disease: A review. Therapeutic

Advances in Psychopharmacology, 3(2), 101-113.

Polymeropoulos, M. H., Lavedan, C., Leroy, E., Ide, S. E., Dehejia, A., Dutra, A., . . .

Nussbaum, R. L. (1997). Mutation in the alpha-synuclein gene identified in families

with parkinson's disease. Science, 276(5321), 2045-2047.

Porchet, R., Probst, A., Bouras, C., Draberova, E., Draber, P., & Riederer, B. M. (2003).

Analysis of glial acidic fibrillary protein in the human entorhinal cortex during aging

and in alzheimer's disease. Proteomics, 3(8), 1476-1485.

Postle, B. R., Stern, C. E., Rosen, B. R., & Corkin, S. (2000). An fMRI investigation of

cortical contributions to spatial and nonspatial visual working memory. NeuroImage,

11(5 Pt 1), 409-423.

Page 203: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

179

Power, J. D., Cohen, A. L., Nelson, S. M., Wig, G. S., Barnes, K. A., Church, J. A., . . .

Petersen, S. E. (2011). Functional network organization of the human brain. Neuron,

72(4), 665-678.

Power, J. D., & Petersen, S. E. (2013). Control-related systems in the human brain. Current

Opinion in Neurobiology, 23(2), 223-228.

Qian, L., & Flood, P. M. (2008). Microglial cells and parkinson's disease. Immunologic

Research, 41(3), 155-164.

Rae, C. L., Correia, M. M., Altena, E., Hughes, L. E., Barker, R. A., & Rowe, J. B. (2012).

White matter pathology in parkinson's disease: The effect of imaging protocol

differences and relevance to executive function. NeuroImage, 62(3), 1675-1684.

Raichle, M. E., & Mintun, M. A. (2006). Brain work and brain imaging. Annual Review of

Neuroscience, 29, 449-476.

Ramirez-Ruiz, B., Marti, M. J., Tolosa, E., Gimenez, M., Bargallo, N., Valldeoriola, F., &

Junque, C. (2007). Cerebral atrophy in parkinson's disease patients with visual

hallucinations. European Journal of Neurology : The Official Journal of the European

Federation of Neurological Societies, 14(7), 750-756.

Ramlackhansingh, A. F., Brooks, D. J., Greenwood, R. J., Bose, S. K., Turkheimer, F. E.,

Kinnunen, K. M., . . . Sharp, D. J. (2011). Inflammation after trauma: Microglial

activation and traumatic brain injury. Annals of Neurology, 70(3), 374-383.

Ransohoff, R. M., & Stevens, B. (2011). Neuroscience. how many cell types does it take to

wire a brain? Science, 333(6048), 1391-1392.

Rektorová, I., Rektor, I., Bares, M., Dostál, V., Ehler, E., Fanfrdlová, Z., … Velísková, J.

(2005). Cognitive performance in people with Parkinson's disease and mild or

moderate depression: effects of dopamine agonists in an add-on to L-dopa therapy.

European Journal of Neurology, 12(1):9-15.

Relja, M., & Klepac, N. (2006). A dopamine agonist, pramipexole, and cognitive functions

in Parkinson's disease. Neurological Sciences, 248(1-2), 251-254.

Riond, J., Mattei, M. G., Kaghad, M., Dumont, X., Guillemot, J. C., Le Fur, G., . . . Ferrara,

P. (1991). Molecular cloning and chromosomal localization of a human peripheral-type

benzodiazepine receptor. European Journal of Biochemistry / FEBS, 195(2), 305-311.

Rone, M. B., Midzak, A. S., Issop, L., Rammouz, G., Jagannathan, S., Fan, J., . . .

Papadopoulos, V. (2012). Identification of a dynamic mitochondrial protein complex

driving cholesterol import, trafficking, and metabolism to steroid hormones. Molecular

Endocrinology, 26(11), 1868-1882.

Page 204: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

180

Rosenberg-Katz, K., Herman, T., Jacob, Y., Giladi, N., Hendler, T., & Hausdorff, J. M.

(2013). Gray matter atrophy distinguishes between parkinson disease motor subtypes.

Neurology, 80(16), 1476-1484.

Rousset, O. G., Ma, Y., & Evans, A. C. (1998). Correction for partial volume effects in PET:

Principle and validation. Journal of Nuclear Medicine : Official Publication, Society of

Nuclear Medicine, 39(5), 904-911.

Rubinov, M., & Sporns, O. (2010). Complex network measures of brain connectivity: Uses

and interpretations. NeuroImage, 52(3), 1059-1069.

Rupprecht, R., Papadopoulos, V., Rammes, G., Baghai, T. C., Fan, J., Akula, N., . . .

Schumacher, M. (2010). Translocator protein (18 kDa) (TSPO) as a therapeutic target

for neurological and psychiatric disorders. Nature Reviews.Drug Discovery, 9(12), 971-

988.

Rupprecht, R., Rammes, G., Eser, D., Baghai, T. C., Schule, C., Nothdurfter, C., . . . Kucher,

K. (2009). Translocator protein (18 kD) as target for anxiolytics without

benzodiazepine-like side effects. Science, 325(5939), 490-493.

Rusjan, P., Mamo, D., Ginovart, N., Hussey, D., Vitcu, I., Yasuno, F., . . . Kapur, S. (2006).

An automated method for the extraction of regional data from PET images. Psychiatry

Research, 147(1), 79-89.

Rusjan, P. M., Wilson, A. A., Bloomfield, P. M., Vitcu, I., Meyer, J. H., Houle, S., &

Mizrahi, R. (2011). Quantitation of translocator protein binding in human brain with the

novel radioligand [18F]-FEPPA and positron emission tomography. Journal of Cerebral

Blood Flow and Metabolism : Official Journal of the International Society of Cerebral

Blood Flow and Metabolism, 31(8), 1807-1816.

Rusjan, P. M., Wilson, A. A., Miler, L., Fan, I., Mizrahi, R., Houle, S., . . . Meyer, J. H.

(2014). Kinetic modeling of the monoamine oxidase B radioligand [11C]SL25.1188 in

human brain with high-resolution positron emission tomography. Journal of Cerebral

Blood Flow and Metabolism : Official Journal of the International Society of Cerebral

Blood Flow and Metabolism, 34(5), 883-889.

Ryterska, A., Jahanshahi, M., & Osman, M. (2013). What are people with parkinson's

disease really impaired on when it comes to making decisions? A meta-analysis of the

evidence. Neuroscience and Biobehavioral Reviews, 37(10 Pt 2), 2836-2846.

Sabatini, U., Boulanouar, K., Fabre, N., Martin, F., Carel, C., Colonnese, C., . . . Rascol, O.

(2000). Cortical motor reorganization in akinetic patients with parkinson's disease: A

functional MRI study. Brain : A Journal of Neurology, 123(Pt 2), 394-403.

Saijo, K., & Glass, C. K. (2011). Microglial cell origin and phenotypes in health and disease.

Nature Reviews.Immunology, 11(11), 775-787.

Page 205: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

181

Salinas, C. A., Searle, G. E., & Gunn, R. N. (2015). The simplified reference tissue model:

Model assumption violations and their impact on binding potential. Journal of Cerebral

Blood Flow and Metabolism : Official Journal of the International Society of Cerebral

Blood Flow and Metabolism, 35(2), 304-311.

Sang, L., Zhang, J., Wang, L., Zhang, J., Zhang, Y., Li, P., . . . Qiu, M. (2015). Alteration of

brain functional networks in early-stage parkinson's disease: A resting-state fMRI study.

PloS One, 10(10), e0141815.

Saxena, S., & Caroni, P. (2011). Selective neuronal vulnerability in neurodegenerative

diseases: From stressor thresholds to degeneration. Neuron, 71(1), 35-48.

Schapira, A. H., & Tolosa, E. (2010). Molecular and clinical prodrome of parkinson disease:

Implications for treatment. Nature Reviews.Neurology, 6(6), 309-317.

Schuitemaker, A., Van Berckel, B. N., Kropholler, M., Boellaard, R., Jonker, C., Scheltens,

P., & Lammertsma, A. A. (2004). Microglia activation in mild cognitive impairment.

Neurobiol. Aging, 25(Suppl. 2), S286.

Schuitemaker, A., VanBerckel, B. N., Boellaard, R., Kropholler, M., Boellaard, R., Jonker,

C., . . . Lammertsma, A. A. (2006). Assessment of microglial activation in mild

cognitive impairment using [11C](R)-PK11195 and PET. NeuroImage, 31, T159.

Schumacher, M., Akwa, Y., Guennoun, R., Robert, F., Labombarda, F., Desarnaud, F., . . .

Baulieu, E. E. (2000). Steroid synthesis and metabolism in the nervous system: Trophic

and protective effects. Journal of Neurocytology, 29(5-6), 307-326.

Schwarz, G. E. (1978). Estimating the dimension of a model. Annals of Statistics, 6(2),

10789-10795.

Scott, G., Hellyer, P. J., Ramlackhansingh, A. F., Brooks, D. J., Matthews, P. M., & Sharp,

D. J. (2015). Thalamic inflammation after brain trauma is associated with thalamo-

cortical white matter damage. Journal of Neuroinflammation, 12, 224.

Seeley, W. W., Crawford, R. K., Zhou, J., Miller, B. L., & Greicius, M. D. (2009).

Neurodegenerative diseases target large-scale human brain networks. Neuron, 62(1), 42-

52.

Seeley, W. W., Menon, V., Schatzberg, A. F., Keller, J., Glover, G. H., Kenna, H., . . .

Greicius, M. D. (2007). Dissociable intrinsic connectivity networks for salience

processing and executive control. The Journal of Neuroscience : The Official Journal of

the Society for Neuroscience, 27(9), 2349-2356.

Segonne, F., Pacheco, J., & Fischl, B. (2007). Geometrically accurate topology-correction of

cortical surfaces using nonseparating loops. IEEE Transactions on Medical Imaging,

26(4), 518-529.

Page 206: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

182

Segura, B., Baggio, H. C., Marti, M. J., Valldeoriola, F., Compta, Y., Garcia-Diaz, A. I., . . .

Junque, C. (2014). Cortical thinning associated with mild cognitive impairment in

parkinson's disease. Movement Disorders : Official Journal of the Movement Disorder

Society, 29(12), 1495-1503.

Selden, N. R., Gitelman, D. R., Salamon-Murayama, N., Parrish, T. B., & Mesulam, M. M.

(1998). Trajectories of cholinergic pathways within the cerebral hemispheres of the

human brain. Brain : A Journal of Neurology, 12 (Pt 12), 2249-2257.

Setiawan, E., Wilson, A. A., Mizrahi, R., Rusjan, P. M., Miler, L., Rajkowska, G., . . .

Meyer, J. H. (2015). Role of translocator protein density, a marker of

neuroinflammation, in the brain during major depressive episodes. JAMA Psychiatry,

72(3), 268-275.

Shenton, M. E., Hamoda, H. M., Schneiderman, J. S., Bouix, S., Pasternak, O., Rathi, Y., . . .

Zafonte, R. (2012). A review of magnetic resonance imaging and diffusion tensor

imaging findings in mild traumatic brain injury. Brain Imaging and Behavior, 6(2), 137-

192.

Shin, N.Y., Shin, Y.S., Lee, P.H., Yoon, U., Han, S., Kim, D.J., & Lee, S.K. (2016).

Different Functional and Microstructural Changes Depending on Duration of Mild

Cognitive Impairment in Parkinson Disease. AJNR Ameican Journal of

Neuroradiology, 37(5), 897-903.

Skidmore, F., Korenkevych, D., Liu, Y., He, G., Bullmore, E., & Pardalos, P. M. (2011).

Connectivity brain networks based on wavelet correlation analysis in parkinson fMRI

data. Neuroscience Letters, 499(1), 47-51.

Sled, J. G., Zijdenbos, A. P., & Evans, A. C. (1998). A nonparametric method for automatic

correction of intensity nonuniformity in MRI data. IEEE Transactions on Medical

Imaging, 17(1), 87-97.

Smith, J. A., Das, A., Ray, S. K., & Banik, N. L. (2012). Role of pro-inflammatory cytokines

released from microglia in neurodegenerative diseases. Brain Research Bulletin, 87(1),

10-20.

Smith, S. M. (2002). Fast robust automated brain extraction. Human Brain Mapping, 17(3),

143-155.

Smith, S. M., Jenkinson, M., Johansen-Berg, H., Rueckert, D., Nichols, T. E., Mackay, C. E.,

. . . Behrens, T. E. (2006). Tract-based spatial statistics: Voxelwise analysis of multi-

subject diffusion data. NeuroImage, 31(4), 1487-1505.

Smith, S. M., Jenkinson, M., Woolrich, M. W., Beckmann, C. F., Behrens, T. E., Johansen-

Berg, H., . . . Matthews, P. M. (2004). Advances in functional and structural MR image

analysis and implementation as FSL. NeuroImage, 23 Suppl 1, S208-19.

Page 207: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

183

Smith, S. M., & Nichols, T. E. (2009). Threshold-free cluster enhancement: Addressing

problems of smoothing, threshold dependence and localisation in cluster inference.

NeuroImage, 44(1), 83-98.

Sofroniew, M. V., & Vinters, H. V. (2010). Astrocytes: Biology and pathology. Acta

Neuropathologica, 119(1), 7-35.

Song, S. K., Lee, J. E., Park, H. J., Sohn, Y. H., Lee, J. D., & Lee, P. H. (2011). The pattern

of cortical atrophy in patients with parkinson's disease according to cognitive status.

Movement Disorders : Official Journal of the Movement Disorder Society, 26(2), 289-

296.

Spillantini, M. G., Crowther, R. A., Jakes, R., Hasegawa, M., & Goedert, M. (1998). Alpha-

synuclein in filamentous inclusions of lewy bodies from parkinson's disease and

dementia with lewy bodies. Proceedings of the National Academy of Sciences of the

United States of America, 95(11), 6469-6473.

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.

Sporns, O. (2013). Network attributes for segregation and integration in the human brain.

Current Opinion in Neurobiology, 23(2), 162-171.

Sporns, O. (2014). Contributions and challenges for network models in cognitive

neuroscience. Nature Neuroscience, 17(5), 652-660.

Sporns, O., Honey, C. J., & Kotter, R. (2007). Identification and classification of hubs in

brain networks. PloS One, 2(10), e1049.

Spreng, R. N., Sepulcre, J., Turner, G. R., Stevens, W. D., & Schacter, D. L. (2013). Intrinsic

architecture underlying the relations among the default, dorsal attention, and

frontoparietal control networks of the human brain. Journal of Cognitive Neuroscience,

25(1), 74-86.

Stam, C. J. (2014). Modern network science of neurological disorders. Nature

Reviews.Neuroscience, 15(10), 683-695.

Stam, C. J., de Haan, W., Daffertshofer, A., Jones, B. F., Manshanden, I., van Cappellen van

Walsum, A. M., . . . Scheltens, P. (2009). Graph theoretical analysis of

magnetoencephalographic functional connectivity in alzheimer's disease. Brain : A

Journal of Neurology, 132(Pt 1), 213-224.

Stoessl, A. J., Lehericy, S., & Strafella, A. P. (2014). Imaging insights into basal ganglia

function, parkinson's disease, and dystonia. Lancet, 384(9942), 532-544.

Page 208: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

184

Stone, D. K., Reynolds, A. D., Mosley, R. L., & Gendelman, H. E. (2009). Innate and

adaptive immunity for the pathobiology of parkinson's disease. Antioxidants & Redox

Signaling, 11(9), 2151-2166.

Streit, W. J. (2006). Microglial senescence: Does the brain's immune system have an

expiration date? Trends in Neurosciences, 29(9), 506-510.

Streit, W. J., Sammons, N. W., Kuhns, A. J., & Sparks, D. L. (2004). Dystrophic microglia in

the aging human brain. Glia, 45(2), 208-212.

Streit, W. J., & Xue, Q. S. (2010). The brain's aging immune system. Aging and Disease,

1(3), 254-261.

Sullivan, E. V., & Pfefferbaum, A. (2006). Diffusion tensor imaging and aging.

Neuroscience and Biobehavioral Reviews, 30(6), 749-761.

Suridjan, I., Pollock, B. G., Verhoeff, N. P., Voineskos, A. N., Chow, T., Rusjan, P. M., . . .

Mizrahi, R. (2015). In-vivo imaging of grey and white matter neuroinflammation in

alzheimer's disease: A positron emission tomography study with a novel radioligand,

[18F]-FEPPA. Molecular Psychiatry, 20(12), 1579-1587.

Suridjan, I., Rusjan, P. M., Kenk, M., Verhoeff, N. P., Voineskos, A. N., Rotenberg, D., . . .

Mizrahi, R. (2014). Quantitative imaging of neuroinflammation in human white matter:

A positron emission tomography study with translocator protein 18 kDa radioligand,

[18F]-FEPPA. Synapse (New York, N.Y.), 68(11), 536-547.

Suridjan, I., Rusjan, P. M., Voineskos, A. N., Selvanathan, T., Setiawan, E., Strafella, A. P., .

. . Mizrahi, R. (2014). Neuroinflammation in healthy aging: A PET study using a novel

translocator protein 18kDa (TSPO) radioligand, [(18)F]-FEPPA. NeuroImage, 84, 868-

875.

Tahmasian, M., Bettray, L.M., van Eimeren, T., Drzezga, A., Timmermann, L., Eickhoff, C.

R., … Eggers, C. (2015). A systematic review on the applications of resting-state fMRI

in Parkinson's disease: Does dopamine replacement therapy play a role? Cortex, 73, 80-

105.

Takahashi, K., Yamaguchi, S., Kobayashi, S., & Yamamoto, Y. (2005). Effects of aging on

regional cerebral blood flow assessed by using technetium tc 99m

hexamethylpropyleneamine oxime single-photon emission tomography with 3D

stereotactic surface projection analysis. AJNR.American Journal of Neuroradiology,

26(8), 2005-2009.

Takano, A., Piehl, F., Hillert, J., Varrone, A., Nag, S., Gulyas, B., . . . Halldin, C. (2013). In

vivo TSPO imaging in patients with multiple sclerosis: A brain PET study with

[18F]FEDAA1106. EJNMMI Research, 3(1), 30-219X-3-30.

Page 209: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

185

Tang, D., Nickels, M. L., Tantawy, M. N., Buck, J. R., & Manning, H. C. (2014). Preclinical

imaging evaluation of novel TSPO-PET ligand 2-(5,7-diethyl-2-(4-(2-

[(18)F]fluoroethoxy)phenyl)pyrazolo[1,5-a]pyrimidin-3-yl)- N,N-diethylacetamide ([

(18)F]VUIIS1008) in glioma. Molecular Imaging and Biology : MIB : The Official

Publication of the Academy of Molecular Imaging, 16(6), 813-820.

Tessitore, A., Amboni, M., Cirillo, G., Corbo, D., Picillo, M., Russo, A., . . . Tedeschi, G.

(2012). Regional gray matter atrophy in patients with parkinson disease and freezing of

gait. AJNR.American Journal of Neuroradiology, 33(9), 1804-1809.

Tessitore, A., Santangelo, G., De Micco, R., Vitale, C., Giordano, A., Raimo, S., …

Tedeschi, G. (2016). Cortical thickness changes in patients with Parkinson's disease and

impulse control disorders. Parkinsonism & Related Disorders, 24, 119-126.

Theilmann, R. J., Reed, J. D., Song, D. D., Huang, M. X., Lee, R. R., Litvan, I., &

Harrington, D. L. (2013). White-matter changes correlate with cognitive functioning in

parkinson's disease. Frontiers in Neurology, 4, 37.

Thiel, A., Radlinska, B. A., Paquette, C., Sidel, M., Soucy, J. P., Schirrmacher, R., & Minuk,

J. (2010). The temporal dynamics of poststroke neuroinflammation: A longitudinal

diffusion tensor imaging-guided PET study with 11C-PK11195 in acute subcortical

stroke. Journal of Nuclear Medicine : Official Publication, Society of Nuclear Medicine,

51(9), 1404-1412.

Tinaz, S., Courtney, M. G., & Stern, C. E. (2011). Focal cortical and subcortical atrophy in

early parkinson's disease. Movement Disorders : Official Journal of the Movement

Disorder Society, 26(3), 436-441.

Tinaz, S., Lauro, P., Hallett, M., & Horovitz, S. G. (2016). Deficits in task-set maintenance

and execution networks in parkinson's disease. Brain Structure & Function, 221(3),

1413-1425.

Tison, F., Dartigues, J. F., Auriacombe, S., Letenneur, L., Boller, F., & Alperovitch, A.

(1995). Dementia in parkinson's disease: A population-based study in ambulatory and

institutionalized individuals. Neurology, 45(4), 705-708.

Tomasi, D., Wang, G. J., & Volkow, N. D. (2013). Energetic cost of brain functional

connectivity. Proceedings of the National Academy of Sciences of the United States of

America, 110(33), 13642-13647.

Tomlinson, C. L., Stowe, R., Patel, S., Rick, C., Gray, R., & Clarke, C. E. (2010). Systematic

review of levodopa dose equivalency reporting in parkinson's disease. Movement

Disorders : Official Journal of the Movement Disorder Society, 25(15), 2649-2653.

Page 210: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

186

Tononi, G., Sporns, O., & Edelman, G. M. (1994). A measure for brain complexity: Relating

functional segregation and integration in the nervous system. Proceedings of the

National Academy of Sciences of the United States of America, 91(11), 5033-5037.

Trujillo, J. P., Gerrits, N. J., Vriend, C., Berendse, H. W., van den Heuvel, O. A., & van der

Werf, Y. D. (2015). Impaired planning in parkinson's disease is reflected by reduced

brain activation and connectivity. Human Brain Mapping, 36(9), 3703-3715.

Tufekci, K. U., Meuwissen, R., Genc, S., & Genc, K. (2012). Inflammation in parkinson's

disease. Advances in Protein Chemistry and Structural Biology, 88, 69-132.

Turner, M. R., Cagnin, A., Turkheimer, F. E., Miller, C. C., Shaw, C. E., Brooks, D. J., . . .

Banati, R. B. (2004). Evidence of widespread cerebral microglial activation in

amyotrophic lateral sclerosis: An [11C](R)-PK11195 positron emission tomography

study. Neurobiology of Disease, 15(3), 601-609.

Uribe, C., Segura, B., Baggio, H. C., Abos, A., Marti, M. J., Valldeoriola, F., . . . Junque, C.

(2016). Patterns of cortical thinning in nondemented parkinson's disease patients.

Movement Disorders : Official Journal of the Movement Disorder Society, 31(5), 699-

708.

Uversky, V. N. (2007). Neuropathology, biochemistry, and biophysics of alpha-synuclein

aggregation. Journal of Neurochemistry, 103(1), 17-37.

Vaishnavi, S. N., Vlassenko, A. G., Rundle, M. M., Snyder, A. Z., Mintun, M. A., & Raichle,

M. E. (2010). Regional aerobic glycolysis in the human brain. Proceedings of the

National Academy of Sciences of the United States of America, 107(41), 17757-17762.

Van Camp, N., Boisgard, R., Kuhnast, B., Theze, B., Viel, T., Gregoire, M. C., . . . Tavitian,

B. (2010). In vivo imaging of neuroinflammation: A comparative study between

[(18)F]PBR111, [ (11)C]CLINME and [ (11)C]PK11195 in an acute rodent model.

European Journal of Nuclear Medicine and Molecular Imaging, 37(5), 962-972.

van den Heuvel, M., Mandl, R., Luigjes, J., & Hulshoff Pol, H. (2008). Microstructural

organization of the cingulum tract and the level of default mode functional connectivity.

The Journal of Neuroscience : The Official Journal of the Society for Neuroscience,

28(43), 10844-10851.

van den Heuvel, M. P., & Hulshoff Pol, H. E. (2010). Exploring the brain network: A review

on resting-state fMRI functional connectivity. European Neuropsychopharmacology :

The Journal of the European College of Neuropsychopharmacology, 20(8), 519-534.

van den Heuvel, M. P., & Sporns, O. (2013). Network hubs in the human brain. Trends in

Cognitive Sciences, 17(12), 683-696.

Page 211: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

187

Varrone, A., Mattsson, P., Forsberg, A., Takano, A., Nag, S., Gulyas, B., . . . Halldin, C.

(2013). In vivo imaging of the 18-kDa translocator protein (TSPO) with

[18F]FEDAA1106 and PET does not show increased binding in alzheimer's disease

patients. European Journal of Nuclear Medicine and Molecular Imaging, 40(6), 921-

931.

Varrone, A., Oikonen, V., Forsberg, A., Joutsa, J., Takano, A., Solin, O., . . . Rinne, J. O.

(2015). Positron emission tomography imaging of the 18-kDa translocator protein

(TSPO) with [18F]FEMPA in alzheimer's disease patients and control subjects.

European Journal of Nuclear Medicine and Molecular Imaging, 42(3), 438-446.

Venneti, S., Lopresti, B. J., Wang, G., Slagel, S. L., Mason, N. S., Mathis, C. A., . . . Wiley,

C. A. (2007). A comparison of the high-affinity peripheral benzodiazepine receptor

ligands DAA1106 and (R)-PK11195 in rat models of neuroinflammation: Implications

for PET imaging of microglial activation. Journal of Neurochemistry, 102(6), 2118-

2131.

Venneti, S., Lopresti, B. J., & Wiley, C. A. (2013). Molecular imaging of

microglia/macrophages in the brain. Glia, 61(1), 10-23.

Venneti, S., Wagner, A. K., Wang, G., Slagel, S. L., Chen, X., Lopresti, B. J., . . . Wiley, C.

A. (2007). The high affinity peripheral benzodiazepine receptor ligand DAA1106 binds

specifically to microglia in a rat model of traumatic brain injury: Implications for PET

imaging. Experimental Neurology, 207(1), 118-127.

Venneti, S., Wang, G., Nguyen, J., & Wiley, C. A. (2008). The positron emission

tomography ligand DAA1106 binds with high affinity to activated microglia in human

neurological disorders. Journal of Neuropathology and Experimental Neurology,

67(10), 1001-1010.

Venneti, S., Wang, G., & Wiley, C. A. (2008). The high affinity peripheral benzodiazepine

receptor ligand DAA1106 binds to activated and infected brain macrophages in areas of

synaptic degeneration: Implications for PET imaging of neuroinflammation in lentiviral

encephalitis. Neurobiology of Disease, 29(2), 232-241.

Venneti, S., Wiley, C. A., & Kofler, J. (2009). Imaging microglial activation during

neuroinflammation and alzheimer's disease. Journal of Neuroimmune Pharmacology :

The Official Journal of the Society on NeuroImmune Pharmacology, 4(2), 227-243.

Vivash, L., & O'Brien, T. J. (2016). Imaging microglial activation with TSPO PET: Lighting

up neurologic diseases? Journal of Nuclear Medicine : Official Publication, Society of

Nuclear Medicine, 57(2), 165-168.

Volpicelli-Daley, L. A., Luk, K. C., Patel, T. P., Tanik, S. A., Riddle, D. M., Stieber, A., . . .

Lee, V. M. (2011). Exogenous alpha-synuclein fibrils induce lewy body pathology

leading to synaptic dysfunction and neuron death. Neuron, 72(1), 57-71.

Page 212: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

188

von Bernhardi, R., Tichauer, J. E., & Eugenin, J. (2010). Aging-dependent changes of

microglial cells and their relevance for neurodegenerative disorders. Journal of

Neurochemistry, 112(5), 1099-1114.

Vroon, A., Drukarch, B., Bol, J. G., Cras, P., Breve, J. J., Allan, S. M., . . . Van Dam, A. M.

(2007). Neuroinflammation in parkinson's patients and MPTP-treated mice is not

restricted to the nigrostriatal system: Microgliosis and differential expression of

interleukin-1 receptors in the olfactory bulb. Experimental Gerontology, 42(8), 762-771.

Wadsworth, H., Jones, P. A., Chau, W. F., Durrant, C., Fouladi, N., Passmore, J., . . . Trigg,

W. (2012). (1)(8)F]GE-180: A novel fluorine-18 labelled PET tracer for imaging

translocator protein 18 kDa (TSPO). Bioorganic & Medicinal Chemistry Letters, 22(3),

1308-1313.

Wakana, S., Caprihan, A., Panzenboeck, M. M., Fallon, J. H., Perry, M., Gollub, R. L., . . .

Mori, S. (2007). Reproducibility of quantitative tractography methods applied to

cerebral white matter. NeuroImage, 36(3), 630-644.

Wang, Y., Shima, K., Sawamura, H., & Tanji, J. (2001). Spatial distribution of cingulate

cells projecting to the primary, supplementary, and pre-supplementary motor areas: A

retrograde multiple labeling study in the macaque monkey. Neuroscience Research,

39(1), 39-49.

Wattendorf, E., Welge-Lussen, A., Fiedler, K., Bilecen, D., Wolfensberger, M., Fuhr, P., . . .

Westermann, B. (2009). Olfactory impairment predicts brain atrophy in parkinson's

disease. The Journal of Neuroscience : The Official Journal of the Society for

Neuroscience, 29(49), 15410-15413.

Watts, D. J., & Strogatz, S. H. (1998). Collective dynamics of 'small-world' networks.

Nature, 393(6684), 440-442.

Watts, R., Liston, C., Niogi, S., & Ulug, A. M. (2003). Fiber tracking using magnetic

resonance diffusion tensor imaging and its applications to human brain development.

Mental Retardation and Developmental Disabilities Research Reviews, 9(3), 168-177.

Wechsler, D. (1997). Manual for the wechsler memeory scale-3rd edition. New York: The

Psychological Corporation.

Wei, L., Zhang, J., Long, Z., Wu, G. R., Hu, X., Zhang, Y., & Wang, J. (2014). Reduced

topological efficiency in cortical-basal ganglia motor network of parkinson's disease: A

resting state fMRI study. PloS One, 9(10), e108124.

Weingarten, C. P., Sundman, M. H., Hickey, P., & Chen, N. K. (2015). Neuroimaging of

parkinson's disease: Expanding views. Neuroscience and Biobehavioral Reviews, 59,

16-52.

Page 213: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

189

Weintraub, D., Doshi, J., Koka, D., Davatzikos, C., Siderowf, A. D., Duda, J. E., . . . Clark,

C. M. (2011a). Neurodegeneration across stages of cognitive decline in parkinson

disease. Archives of Neurology, 68(12), 1562-1568.

Werring, D. J., Clark, C. A., Barker, G. J., Thompson, A. J., & Miller, D. H. (1999).

Diffusion tensor imaging of lesions and normal-appearing white matter in multiple

sclerosis. Neurology, 52(8), 1626-1632.

Whitfield-Gabrieli, S., & Nieto-Castanon, A. (2012). Conn: A functional connectivity

toolbox for correlated and anticorrelated brain networks. Brain Connectivity, 2(3), 125-

141.

Wicklund, A. H., Johnson, N., & Weintraub, S. (2004). Preservation of reasoning in primary

progressive aphasia: Further differentiation from alzheimer's disease and the behavioral

presentation of frontotemporal dementia. Journal of Clinical and Experimental

Neuropsychology, 26(3), 347-355.

Wienhard, K., Schmand, M., Casey, M. E., Baker, K., Bao, J., Eriksson, L., . . . Nutt, R.

(2002). The ECAT HRRT: Performance and first clinical application of the new high

resolution research tomograph. IEEE Trans Nucl Sci, 49, 104-110.

Williams-Gray, C. H., Foltynie, T., Brayne, C. E., Robbins, T. W., & Barker, R. A. (2007).

Evolution of cognitive dysfunction in an incident parkinson's disease cohort. Brain : A

Journal of Neurology, 130(Pt 7), 1787-1798.

Wilson, A. A., Garcia, A., Parkes, J., McCormick, P., Stephenson, K. A., Houle, S., &

Vasdev, N. (2008). Radiosynthesis and initial evaluation of [18F]-FEPPA for PET

imaging of peripheral benzodiazepine receptors. Nuclear Medicine and Biology, 35(3),

305-314.

Wiltshire, K., Concha, L., Gee, M., Bouchard, T., Beaulieu, C., & Camicioli, R. (2010).

Corpus callosum and cingulum tractography in parkinson's disease. The Canadian

Journal of Neurological Sciences.Le Journal Canadien Des Sciences Neurologiques,

37(5), 595-600.

Winkeler, A., Boisgard, R., Martin, A., & Tavitian, B. (2010). Radioisotopic imaging of

neuroinflammation. Journal of Nuclear Medicine : Official Publication, Society of

Nuclear Medicine, 51(1), 1-4.

Woolrich, M. W., Jbabdi, S., Patenaude, B., Chappell, M., Makni, S., Behrens, T., . . . Smith,

S. M. (2009). Bayesian analysis of neuroimaging data in FSL. NeuroImage, 45(1 Suppl),

S173-86.

Wu, T., Liu, J., Zhang, H., Hallett, M., Zheng, Z., & Chan, P. (2015). Attention to automatic

movements in parkinson's disease: Modified automatic mode in the striatum. Cerebral

Cortex, 25(10), 3330-3342.

Page 214: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

190

Wu, T., Long, X., Zang, Y., Wang, L., Hallett, M., Li, K., & Chan, P. (2009). Regional

homogeneity changes in patients with parkinson's disease. Human Brain Mapping,

30(5), 1502-1510.

Wu, T., Wang, L., Chen, Y., Zhao, C., Li, K., & Chan, P. (2009). Changes of functional

connectivity of the motor network in the resting state in parkinson's disease.

Neuroscience Letters, 460(1), 6-10.

Yasuno, F., Ota, M., Kosaka, J., Ito, H., Higuchi, M., Doronbekov, T. K., . . . Suhara, T.

(2008). Increased binding of peripheral benzodiazepine receptor in alzheimer's disease

measured by positron emission tomography with [11C]DAA1106. Biological

Psychiatry, 64(10), 835-841.

Yeo, B. T., Krienen, F. M., Sepulcre, J., Sabuncu, M. R., Lashkari, D., Hollinshead, M., . . .

Buckner, R. L. (2011). The organization of the human cerebral cortex estimated by

intrinsic functional connectivity. Journal of Neurophysiology, 106(3), 1125-1165.

Yokokura, M., Terada, T., Bunai, T., Nakaizumi, K., Takebayashi, K., Iwata, Y., . . . Ouchi,

Y. (2016). Depiction of microglial activation in aging and dementia: Positron emission

tomography with [11C]DPA713 versus [11C](R)PK11195. Journal of Cerebral Blood

Flow and Metabolism : Official Journal of the International Society of Cerebral Blood

Flow and Metabolism, April 26.

Yu, H., Sternad, D., Corcos, D. M., & Vaillancourt, D. E. (2007). Role of hyperactive

cerebellum and motor cortex in parkinson's disease. NeuroImage, 35(1), 222-233.

Zalesky, A., Fornito, A., Cocchi, L., Gollo, L. L., & Breakspear, M. (2014). Time-resolved

resting-state brain networks. Proceedings of the National Academy of Sciences of the

United States of America, 111(28), 10341-10346.

Zarei, M., Ibarretxe-Bilbao, N., Compta, Y., Hough, M., Junque, C., Bargallo, N., . . . Marti,

M. J. (2013). Cortical thinning is associated with disease stages and dementia in

parkinson's disease. Journal of Neurology, Neurosurgery, and Psychiatry,

Zgaljardic, D. J., Borod, J. C., Foldi, N. S., Mattis, P. J., Gordon, M. F., Feigin, A., &

Eidelberg, D. (2006). An examination of executive dysfunction associated with

frontostriatal circuitry in parkinson's disease. Journal of Clinical and Experimental

Neuropsychology, 28(7), 1127-1144.

Zhan, W., Kang, G. A., Glass, G. A., Zhang, Y., Shirley, C., Millin, R., . . . Schuff, N.

(2012). Regional alterations of brain microstructure in parkinson's disease using

diffusion tensor imaging. Movement Disorders : Official Journal of the Movement

Disorder Society, 27(1), 90-97.

Page 215: Neural Substrates of Parkinson s Disease · Neural Substrates of Parkinson’s Disease Yuko Koshimori Doctor of Philosophy Institute of Medical Science University of Toronto 2016

191

Zhang, K., Yu, C., Zhang, Y., Wu, X., Zhu, C., Chan, P., & Li, K. (2011). Voxel-based

analysis of diffusion tensor indices in the brain in patients with parkinson's disease.

European Journal of Radiology, 77(2), 269-273.

Zhang, M. R., Kida, T., Noguchi, J., Furutsuka, K., Maeda, J., Suhara, T., & Suzuki, K.

(2003). [(11)C]DAA1106: Radiosynthesis and in vivo binding to peripheral

benzodiazepine receptors in mouse brain. Nuclear Medicine and Biology, 30(5), 513-

519.

Zhang, M. R., Maeda, J., Furutsuka, K., Yoshida, Y., Ogawa, M., Suhara, T., & Suzuki, K.

(2003). 18F]FMDAA1106 and [18F]FEDAA1106: Two positron-emitter labeled ligands

for peripheral benzodiazepine receptor (PBR). Bioorganic & Medicinal Chemistry

Letters, 13(2), 201-204.

Zhang, X., Haaf, M., Todorich, B., Grosstephan, E., Schieremberg, H., Surguladze, N., &

Connor, J. R. (2005). Cytokine toxicity to oligodendrocyte precursors is mediated by

iron. Glia, 52(3), 199-208.

Zhao, C., Li, W. W., & Franklin, R. J. (2006). Differences in the early inflammatory

responses to toxin-induced demyelination are associated with the age-related decline in

CNS remyelination. Neurobiology of Aging, 27(9), 1298-1307.

Zheng, Z., Shemmassian, S., Wijekoon, C., Kim, W., Bookheimer, S. Y., & Pouratian, N.

(2014). DTI correlates of distinct cognitive impairments in parkinson's disease. Human

Brain Mapping, 35(4), 1325-1333.