235
AN EEG INVESTIGATION OF PAIN PROCESSING IN PARKINSON’S DISEASE A thesis submitted to the University of Manchester for the degree of Doctor of Philosophy in Faculty of Biology, Medicine and Health 2019 Sarah L. Martin School of Biological Sciences

AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

AN EEG INVESTIGATION OF PAIN PROCESSING IN PARKINSON’S

DISEASE

A thesis submitted to the University of Manchester for the degree of

Doctor of Philosophy in Faculty of Biology, Medicine and Health

2019

Sarah L. Martin

School of Biological Sciences

Page 2: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2

Contents

LIST OF FIGURES 5

LIST OF TABLES 7

ABBREVIATIONS 8

ABSTRACT 9

LAY ABSTRACT 10

DECLARATION 12

COPYRIGHT STATEMENT 12

ACKNOWLEDGEMENTS 13

PREFACE 15

CHAPTER 1 17

INTRODUCTION 17

1.1. PARKINSON’S DISEASE AND CHRONIC PAIN 18

1. 2. PARKINSON’S DISEASE: AN OVERVIEW 20

1.3. BRAIN REGIONS INVOLVED IN PAIN PROCESSING 24

1.4. PATHOLOGICAL PREDISPOSITION OF PAIN IN PD 29

1.5. IMAGING STUDIES SHOWING ALTERED CENTRAL PAIN PROCESSING IN PARKINSON’S DISEASE 45

1.6. CONCLUSION 47

CHAPTER 2 48

GENERAL METHODS AND INTRODUCTION TO STUDY AIMS 48

2.1. GENERAL METHODS 49

2.2. INTRODUCTION TO STUDY OBJECTIVES 54

2.3. RECRUITMENT 60

Page 3: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3

CHAPTER 3 64

A NEUROPHYSIOLOGICAL INVESTIGATION OF PAIN ANTICIPATION IN PARKINSON’S DISEASE 64

3.1. ABSTRACT 65

3.2. INTRODUCTION 66

3.3. METHODS 68

3.4. RESULTS 76

3.5. DISCUSSION 85

3.6. CONCLUSION 89

3.7. SUPPLEMENTARY MATERIALS 90

CHAPTER 4 93

CENTRAL PAIN PROCESSING IN PEOPLE WITH PARKINSON’S DISEASE WHILST ON MEDICATION 93

4.1. ABSTRACT 94

4.2. INTRODUCTION 94

4.3. METHODS 95

4.4. RESULTS 101

4.5. DISCUSSION 104

4.6. CONCLUSION 106

CHAPTER 5 108

AN INVESTIGATION OF THE ROLE OF THE DOPAMINE D2 RECEPTOR IN PAIN PERCEPTION 108

5.1. ABSTRACT 109

5.2. INTRODUCTION 110

5.3. METHODS 113

5.4. RESULTS 120

5.5. DISCUSSION 132

5.6. CONCLUSION 139

CHAPTER 6 140

AN EEG INVESTIGATION OF AGE-RELATED DIFFERENCES IN ACUTE PAIN 140

6.1. ABSTRACT 141

6.2. INTRODUCTION 141

6.3. METHODS 144

Page 4: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4

6.4. RESULTS 150

6.5. DISCUSSSION 157

6.6. CONCLUSION 161

CHAPTER 7 162

GENERAL DISCUSSION 162

7.1. MAIN AIM OF THESIS 163

7.2. SUMMARY OF EXPERIMENTAL WORK 163

7.3. AN INTERPRETATION OF THESIS STUDIES 165

7.4. SUMMARY OF INTERPRETATION 171

7.5. NEUROIMAGING RESEARCH OF PAIN IN PARKINSON’S 173

7.6. FUTURE CONSIDERATIONS 175

7.7. FUTURE RESEARCH 179

7.8. FINAL CONCLUSION 181

REFERENCES 182

Page 5: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5

List of Figures

FIGURE 1.1: Regions of the pain network taken from Bushnell, Ceko and Low, 2013 25

FIGURE 1.2: The SN and its connections to the regions of the pain network…………… 31

FIGURE 1.3: A summary of the various roles of dopamine and how they are

involved in pain perception..................................................................... 38

FIGURE 1.4: Dopaminergic pathways…………………....................................................... 39

FIGURE 2.1: The pain rating scale that was used for the psychophysics and main

experiments………………………………………………………………………………………… 50

FIGURE 2.2: An EEG waveform of an SPN generated by a visual anticipatory cue,

auditory cues, and the LEP response following acute laser pain...……….. 53

FIGURE 2.3: A schematic diagram of the action of the dopamine D2 receptor and

how low and high doses of agonists and antagonists have different

effects on postsynaptic D2 receptor activity……………............................. 59

FIGURE 2.4: The inverted-U relationship between dopamine level and

performance………...............................................................................….. 61

FIGURE 2.5: Business cards given to participants of the D2 dopamine study……........ 63

FIGURE 3.1: A schematic diagram of a single trial of the experimental paradigm…... 71

FIGURE 3.2: The energy of the laser required to induce high (level 7) pain…............. 77

FIGURE 3.3: Graphs showing the information of the N2 and P2 components of the

laser-evoked potential (LEP)………………………………...………….………............ 79

FIGURE 3.4: Topography plots showing the average response for the mid- and late-

anticipation time windows for HC and PD…............................................ 80

FIGURE 3.5: Source estimates for significant F-contrasts at FWE correction using

MRIcroN software…………………………………………………………………............... 83

FIGURE 3.6: Diagram from Böcker et al., (2001) which reports the grand average of

the stimulus preceding negativity associated with anticipating

threatening stimuli……………………………………............................................ 87

FIGURE 4.1: The laser voltage required to induce a low and high pain

intensity…………………..…………………………………………………..……..……………... 102

Page 6: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6

FIGURE 4.2: The effect of certainty on the pain rating scores…………………………......... 103

FIGURE 5.1: A schematic diagram of a single trial of the experimental paradigm...... 117

FIGURE 5.2: The effect of certainty on each laser intensity calculated and plotted

for each drug…………………………….................................................…………. 123

FIGURE 5.3: The eye-blink rate (EBR) calculated for each dopamine manipulation

condition highlighted a monotonic relationship between dopamine

D2R activation and EBR…………………………………………………………………….... 125

FIGURE 5.4: Topography plots showing the average response for early-, mid– and

late-anticipation for all conditions…………………………………………………....... 126

FIGURE 5.5: Source estimates during mid-anticipation for T-contrast of Control

versus agonist (Cabergoline) and antagonist (Amisulpride)……………….... 129

FIGURE 5.6: Source estimates during mid-anticipation for the Expectation T-

contrast of known High versus known Low……………………………………....... 130

FIGURE 5.7: The drug effect reported using source localisation for post-stimulus

TWOI [200 600 ms]…………………………....................................................... 131

FIGURE 5.8: A schematic diagram to signify the inverted-U relationship between

Dopamine level and performance.……………………..………………………………. 135

FIGURE 6.1: A schematic diagram of a single trial of the experimental paradigm...... 146

FIGURE 6.2: Pain rating of laser stimuli…………..………………………………......................... 151

FIGURE 6.3: The pain rating for Known High stimuli was averaged across

participants within each group at each trial number…………................... 152

FIGURE 6.4: Waveforms at Cz electrode for all participants in the Young and Old

groups……................................................................................................. 153

FIGURE 6.5: ERP waveforms and topography plots………………………………………............ 154

FIGURE 6.6: Topographic plots of the SPM F-statistics for the main group effect

during anticipation and post-stimulus time windows………………………..... 155

FIGURE 7.1: A schematic diagram of the relationship between dopamine level and

performance of dopaminergic processes…………………………………………….. 172

Word Count: 43,028

Page 7: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7

List of Tables

TABLE 1.1: Motor symptoms of Parkinson’s disease……………………………………………… 21

TABLE 1.2: Non-motor symptoms of Parkinson’s disease.…………………………………….. 21

TABLE 1.3: Pathological findings of Lewy body Pathology in the Parkinsonian brain outlined by Braak et al., (2004)………………………………………………….. 23

TABLE 3.1: The table displays the reported measures for the assessments for the PD and HC groups.………………………………………………………………………………. 76

TABLE 3.2: Group effect on sources of early, mid and late anticipation-evoked responses.…………………………………………………………………………………………… 82

TABLE 3.3: Scalp-level SPM results for the F-contrast of Intensity (Low v High) for post-stimulus TWOI [200 600]…………………………………………………………….. 90

TABLE 3.4: The source localisation results for the significant within-subject contrast of Intensity (Low v High) during the post-stimulus time window [200 600 ms].............................................................................. 91

TABLE 3.5: A detailed version of Table 3.2 to include all regions highlighted by the AAL2 toolbox............................................................................................ 92

TABLE 4.1: The rating of the laser stimuli for each laser condition and each group. 103

TABLE 5.1: The pain rating scores for the experimental paradigm........................... 121

TABLE 5.2: The unpleasantness rating for each laser condition for each Drug Condition................................................................................................. 128

TABLE 5.3: Source localisation significant results within mid-anticipation and the post stimulus TWOI.................................................................................. 131

TABLE 6.1: Scalp-level statistical results for whole-scalp analysis............................. 156

Page 8: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

8

Abbreviations

AAL2 Anatomical Automatic Labelling 2 ANOVA Analysis of variance

BEM Boundary Element Model BL Baseline

CBT Cognitive Behavioural Therapy D2R Dopamine D2 Receptor EBR Eye Blink Rate EEG Electroencephalography ERP Event Related Potential

HADS Hospital Anxiety and Depression Scale HC Healthy Control

ICA Independent Component Analysis LEP Laser Evoked Potential

LORETA Low Resolution Electromagnetic Tomography MCC Mid-Cingulate Cortex

MDS-UPDRS Movement Disorder Society Unified Parkinson’s Disease Rating Scale MoCA Montreal Cognitive Assessment

PAG Periaqueductal Gray PCS Pain Catastrophising Scale PD Parkinson’s Disease

PET Positron Emission Tomography PFC Prefrontal Cortex

PwPD People with Parkinson’s Disease ROI Region of Interest

SASICA Semi-Automatic Selection of Independent Components for Artifact correction

S1/S2 Primary/Secondary Somatosensory Cortex SMA Supplementary Motor Area SPM Statistical Parametric Mapping

TWOI Time Window of Interest VAS Visual Analogue Scale

Page 9: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

9

Abstract

Chronic pain in Parkinson’s disease (PD) is significantly higher than in the general population, and is often overlooked to be a side effect of the impaired movement and stiffness associated with the disease. There is scientific rational to link the degeneration which occurs in the PD brain to the increased prevalence of chronic pain. Although acute pain thresholds have been shown to be lower in PD, there is a lack of neuroimaging research which investigates the role of central processing in PD. Therefore, this thesis documents one of the first neuroimaging studies to investigate whether central pain processing is abnormal in people with PD.

The perception of pain can be modulated by subject driven processing such as attention, salience assignment and cognition, which is regarded as top-down modulation and is often abnormal in chronic pain conditions. A technique to investigate top-down processing is to study the neural activity during the anticipation of acute pain stimulus. Hence, in this thesis, we used EEG to record the anticipatory response to acute pain stimuli delivered by a CO2 laser.

Our first experiment compared people with PD whilst off their medication to a healthy age-matched cohort and concluded that the PD patients showed an amplified anticipatory response and a higher sensitivity to the laser. In contrast, following dopamine increasing medication, no difference was shown between the PD and healthy controls.

To investigate the role of dopamine in altering pain perception in PD, the second study investigated the effect of manipulating the dopamine D2 receptor in young healthy volunteers. The study reported that the agonist and antagonist induced reductions in neural activity in the parietal and temporal lobes during pain perception, and the antagonist induced a reduction in the activity of the insula during pain perception, and intensified the effect of certainty during anticipation on the rating of pain.

The patient and the D2R manipulation studies demonstrated that dopamine may play a role in the top-down processing of pain and hence explain why chronic pain in PD is prevalent. Importantly the pain rating and sensitivity were not changed by Levodopa administration or D2R manipulation and supports the notion that dopamine is central to modulating top-down processing, rather than directly coding the intensity of pain. Therefore, together with further neuroimaging research conducted during the PhD, there is strong evidence of a disruption in the central processing of pain in PD and promotes the use of alternative therapies in the treatment of chronic pain in PD.

Page 10: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

10

Lay Abstract

It is very common that people with Parkinson’s disease also suffer from long lasting

pain, known as chronic pain. The pain can be caused by the symptoms of

Parkinson’s disease such as; rigidity, bad posture and impaired movement.

However, changes in pain perception have been seen very early in the disease

before these symptoms have occurred. In addition, there is little evidence that the

severity of the movement symptoms relates to the prevalence of chronic pain.

Therefore, our research aimed to investigate how the changes within the brain

could be involved in the increased susceptibility to chronic pain in Parkinson’s

disease. During the anticipation of pain the activity of brain regions associated with

pain have been shown to be different in people with chronic pain. Therefore, our

experiments also recorded the brain activity during the anticipation of a painful

stimulus.

Firstly, within people with Parkinson’s off their medication we recorded their brain

activity whilst they anticipated pain. In comparison to a cohort of healthy

participants, we discovered that the participants with Parkinson’s disease were

more sensitive to the pain, plus a region of the brain known as the midcingulate

cortex showed increased activity during anticipation. The midcingulate cortex is

involved in pain sensitivity and therefore shows that the region is working

differently in people with Parkinson’s. After the people with Parkinson’s had taken

their normal medication, they remained more sensitive to the painful stimulus;

however, the medication had stopped the increased brain activity within the

midcingulate cortex during anticipation that was seen whilst they were off their

medication.

The medication for Parkinson’s disease improves the activity of dopamine, a

chemical within the brain. Therefore, our second investigation aimed to understand

the role of dopamine in the anticipation and perception of pain within young

healthy people by using drugs which increase and decrease the level of dopamine

activity in the brain. The participants completed the same experiment which was

Page 11: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

11

used in the Parkinson’s study. Our results showed that dopamine did not change an

individual’s pain sensitivity; however it does alter the way we rate the pain

depending on the information we provided during anticipation. This may indicate

that long-term changes in dopamine in the brain within Parkinson’s disease could

alter how an individual feels pain.

Our final aim was to investigate the difference in brain activity during the

anticipation and perception of pain between young and old people. Our

comparison showed that the young cohort showed a much higher level of brain

activity during the experiment. This may have been due to a difference in pain

processing and our relationship with pain changing over time between the two

groups. The difference could also be due to changes in the quality of brain

recordings in young and older brains.

In summary, the research highlighted an increased sensitivity to pain in people with

Parkinson’s disease and some evidence for altered brain activity in Parkinson’s

disease. With better understanding of the source of the pain, there will be better

treatment for people with Parkinson’s disease and pain. Therefore, the evidence of

altered brain activity during anticipation of pain indicates that treatments aimed at

improving how the brain processes pain, such as meditation and cognitive

behavioural therapy (CBT), could be beneficial for people with Parkinson’s disease.

Page 12: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

12

Declaration

No portion of the work included in this thesis has been submitted in support of an application for another degree or qualification of this or any other university or other institute of learning.

Copyright Statement

i. The author of this thesis (including any appendices and/or schedules to this thesis) owns certain copyright or related rights in it (the “Copyright”) and s/he has given The University of Manchester certain rights to use such Copyright, including for administrative purposes.

ii. Copies of this thesis, either in full or in extracts and whether in hard or electronic copy, may be made only in accordance with the Copyright, Designs and Patents Act 1988 (as amended) and regulations issued under it or, where appropriate, in accordance with licensing agreements which the University has from time to time. This page must form part of any such copies made.

iii. The ownership of certain Copyright, patents, designs, trademarks and other intellectual property (the “Intellectual Property”) and any reproductions of copyright works in the thesis, for example graphs and tables (“Reproductions”), which may be described in this thesis, may not be owned by the author and may be owned by third parties. Such Intellectual Property and Reproductions cannot and must not be made available for use without the prior written permission of the owner(s) of the relevant Intellectual Property and/or Reproductions.

iv. Further information on the conditions under which disclosure, publication and commercialisation of this thesis, the Copyright and any Intellectual Property and/or Reproductions described in it may take place is available in the University IP Policy (see http://documents.manchester.ac.uk/display.aspx?DocID=24420), in any relevant Thesis restriction declarations deposited in the University Library, The University Library’s regulations (see http://www.library.manchester.ac.uk/about/regulations/) and in The University’s policy on Presentation of Theses.

Page 13: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Acknowledgements

I have been immensely lucky for the support that I have received throughout my

PhD from my supervisors, colleagues, family and friends. Firstly I would like to

extend huge thanks to my supervision team; Dr Monty Silverdale, Professor

Anthony Jones, Dr Christopher Kobylecki, Dr Christopher Brown and Professor Wael

El-Deredy, and my advisor Dr Joanna Neil. Together they have helped me to

develop as a researcher and overcome challenges that I have faced.

I am hugely thankful for the teaching and guidance I received from my supervisors,

including Anthony’s knowledge of all things pain related, Chris Brown’s EEG analysis

training & support, and Chris Kobylecki’s Parkinson’s knowledge & recruitment.

Special thanks go to Monty, who is without a doubt, scarily intelligent, both in

neuroscience and as a supervisor. His support has given me the confidence to be

independent in my research and excitement to continue research in Parkinson’s

disease.

Working in The Human Pain Research Group has been incredibly important to my

enjoyment of my PhD. Anthony has created a supportive and team-focused lab

where hard work and commitment are expected, yet a good work-life balance is

expected in equal measure. An immense thank you to the irreplaceable Tim Rainey;

I truly would have been lost without your EEG training, technical support, running

and cycling commentary, and of course the amazing cups of teas. I also want to give

huge thanks to Kate Lees, a fountain of knowledge and the greatest supplier of

kitten and puppy photos! And finally to Grace Whitaker and Emily Hird who guided

and trained me in my first couple of years. To be part of the group gave me an

identity that I am proud of, and truly hope that the saying “You never leave The

Human Pain Research Group” is true.

I am also indebted to the people at Salford Royal who have provided technical

support from Medical Physics; Stuart Watson, Prawin Samraj, and Donald Allan and

IT support from Adam Lounsbach.

Page 14: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Acknowledgements

14

It goes without saying that my family have provided fantastic support throughout

my PhD – as they do in everything that I do. Special thanks go to my dad, Ian

Martin, for his support. An extra thank you goes to my brother Paul Martin, for all

of his help with coding throughout the early stages of my PhD.

I am immensely grateful for the friendships outside the world of PhD. Special thanks

go to the South Manchester Mafia; Grace Bennett, Alex Bennett and Jon Robinson –

a home away from home, with Friday night film and food, and our superior skills of

escape rooms keeping me sane throughout my PhD. Huge thanks go to Ruth Ingram

– the dream of a house-mate and more-so greatest friend and PhD ally.

Further thanks must be extended to my Girlguiding family in Manchester who have

provided me with a break from the PhD bubble. Running the 1st Height Brownie unit

is a joy and I have been so lucky to have made so many friends, and had amazing

experiences through Girlguiding.

This research could not have happened without the funding from Parkinson’s UK,

who I am incredibly grateful for. And finally, I am immensely thankful to my

examiners, Dr Donna Lloyd and Dr Ellen Poliakoff, for taking the time to read and

assess my thesis.

This thesis is dedicated to my mum, Carran Martin, a fun -loving,

courageous and inspiring woman.

Page 15: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Preface

This thesis contains the research which stems from Monty Silverdale’s observations

of Parkinson’s disease patients suffering from chronic pain which did not appear to

correlate with the severity of their motor and peripheral symptoms. For better

understanding and thus better treatment, Monty began a collaboration with The

Human Pain Research Group at Salford Royal Hospital to further understand the

brain’s role in pain in Parkinson’s disease. Together with Professor Anthony Jones,

Dr Christopher Kobylecki, , Dr Christopher Brown and Professor Wael El-Deredy, a

series of studies were designed using EEG and laser-induced pain. The aim of the

PhD was to investigate the processing of pain within the Parkinson’s disease in

comparison to a healthy age-matched cohort, and to investigate the impact of the

altered neurotransmitter levels within Parkinson’s disease could have on pain

processing. I was lucky enough to be selected to undertake this research and knew

the projects were very exciting for a PhD student to carry out.

Initially, two studies were outlined for the thesis, including a comparison of

Parkinson’s patients with age-matched controls, and a second study investigating

the role of Dopamine and Serotonin in pain perception within a young healthy

cohort via the use of acute amino acid depletion. At the start of my PhD, an exciting

collaboration was set up with Dr Grace Whitaker who would be recruiting

participants for a pharmacological modulation of Dopamine. The research linked

perfectly with our research aims and the study was carried out in my second year of

the PhD.

Unfortunately, the building which the research took place, was being demolished

throughout my second and third year, and caused significant delays to data

collection of the amino acid depletion study due to the displacement, noise,

vibration and dust. Therefore, for the submission of this thesis, the second study

included in the thesis was switched to be the dopamine pharmacological

modulation study carried out in collaboration with Dr Grace Whitaker, instead of

the amino-acid depletion study. The theories behind the amino acid modulation

Page 16: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Preface

16

and dopamine modulation studies were both to investigate how altered

neurotransmitters in Parkinson’s disease impacts pain perception, and hence,

provide a similar narrative to the progression of this thesis.

Page 17: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Chapter 1

Introduction

Page 18: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

18

1.1. PARKINSON’S DISEASE AND CHRONIC PAIN

In comparison to the general population, the prevalence of chronic pain in

Parkinson’s disease (PD) has been reported to be significantly higher at between

40% and 85% (median = 57.78 %) (Valkovic et al., 2015; Allen et al., 2015; Beiske et

al., 2009; Lee et al., 2006; Goetz et al., 1986; Tinazzi et al., 2006; Nègre-Pagès et al.,

2008a; Hanagasi et al., 2011; Rana et al., 2013; Defazio et al., 2008; Silva, Viana and

Quagliato, 2008; Silverdale et al., 2018) whilst the prevalence within the general

population has been reported to be 22.75% [averaged from (Blyth et al., 2001b;

Johannes et al., 2010; Breivik et al., 2006)]. There is clearly a higher prevalence of

chronic pain reported in the PD population compared to the general population

which indicates that the development of PD may increase an individual’s

susceptibility to developing chronic pain.

The development and persistence of chronic pain is thought to be associated with

dysfunctional bottom-up and top-down mechanisms of pain modulation. The term

bottom-up refers to stimulus-driven modulation (i.e. stimulus intensity, stimulus

novelty etc) and modulation of ascending pain signals in the dorsal horn of the

spinal cord. In contrast, top-down modulation refers to subject-driven manipulation

of the perception of pain (i.e. emotional state, cognition, attention orientation,

memories etc). Both processes can either facilitate or inhibit pain perception and

abnormalities within these mechanisms are associated with chronic pain conditions

and pain hypersensitivity.

The presence of chronic pain in PD patients has often been considered to be a

consequence of the musculoskeletal symptoms, and to be primarily driven by

excessive nociceptive input due to bottom-up processes. Although musculoskeletal

symptoms are likely to contribute to chronic pain, the fact that pain can precede

the musculoskeletal symptoms and that the pain can be idiopathic, demonstrates

that heightened pain may be a syndrome caused by the pathological changes within

the brain associated with PD. There is a lack of neuroimaging research which

Page 19: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

19

investigates the role of impaired top-down processing. However, the degeneration

within the PD brain provides a scientific rationale to suggest that altered top-down

processing, rather than merely atypical peripheral nociceptive transmission, may be

the cause of the increased likelihood of chronic pain in PD.

To dispel theories that the pain is solely a result of peripheral musculoskeletal

symptoms a large scale investigation of 1957 PD patients reported that there was

no correlation between pain and motor impairments during early stage PD

(Silverdale et al., 2018). In addition, Valkovic et al., (2015) reported that a high

proportion of pain in PD is deemed to be central neuropathic pain which was

widespread and not associated with the musculoskeletal or dystonic symptoms.

Additionally, pain has been reported to be located in areas that are unlikely to be

affected by the PD muscular symptoms such as oral, genital, (Ford et al., 1996) and

abdominal pain (Nègre-Pagès et al., 2008a).

Finally, pain perception has been shown to be abnormal in PD patients irrespective

of whether they have chronic pain or not. For instance, PD patients have an

increased pain-induced cortical activation as measured by positron emission

tomography (PET) (Brefel-Courbon et al., 2005), reduced pain thresholds and

tolerance (Zambito Marsala et al., 2011), and reduced electrical and muscle

withdrawal reflex to pain (Mylius et al., 2008). Furthermore, numerous studies

which selectively investigate PD patients suffering from chronic pain have reported

reduced pain thresholds and an inability to habituate to stimuli compared to

healthy controls (Schestatsky et al., 2007a; Djaldetti et al., 2004a).

Chronic pain negatively impacts on daily life and has been considered to be more of

an impairment than the motor symptoms (Silverdale et al., 2018). Therefore, there

is clear evidence that further research is required to understand the cause of

chronic pain in people with PD to provide an improvement in treatment. Here, I

briefly review the pathology of PD, pain processing and the impairments within

shared central process which shed light on a potential link between PD and chronic

pain.

Page 20: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

20

1. 2. PARKINSON’S DISEASE: AN OVERVIEW

PD is a neurodegenerative disease affecting 127, 000 people in the UK and 1 million

in the US and is progressively more common with increasing age (Dorsey et al.,

2007). It is the second biggest neurological disease after Alzheimer’s disease,

affecting approximately 100/100,000 (Seidel et al., 2015b). PD is classically

regarded to be due to the loss of dopaminergic neurons located in the basal

ganglia, specifically the substantia nigra pars compacta (SNpc), however, there is a

multitude of additional pathological changes. The basal ganglia are the main

processors of voluntary motor movements and hence the main symptoms of PD are

regarded to be motor related. The characteristic features of PD are the presence of

a tremor, bradykinesia, and rigidity with symptoms including, shuffling gait, bad

posture and reduced facial expression. Nevertheless, PD is not solely a movement

disorder, and non-motor symptoms are seen to precede the motor aspects. The

widespread degeneration throughout the brain also leads to non-motor symptoms

including; cognitive impairments, dementia, pain, sleep disturbances, depression

and anxiety (Barone, 2010).

The three main motor symptoms of PD are tremor, bradykinesia and rigidity and

are normally the first point at which the disease can be clinically recognised. Below

Table 1.1 defines the motor related symptoms seen in PD. The severity of each

symptom varies between patients. The non-motor symptoms associated with PD

are outlined in Table 1.2. It is important to state that not all PD patients experience

all of the symptoms discussed, and the severity of the condition varies between

each person. The presence of cognitive decline has been related to the altered

neurotransmitter activity in the PD brain.

Page 21: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

21

Table 1.1: Motor symptoms of Parkinson’s disease.

Symptom Definition Location Reference

Tremor

Uncontrolled, somewhat rhythmic movements. The amplitude of the tremor can be as large as more than 10cm, as

well as being as low as below 1cm.

Upper and lower limbs.

Jaw/Lips

(Geraghty, Jankovic and Zetusky, 1985; Jankovic,

Schwartz and Ondo, 1999)

Bradykinesia Slow movement and freezing n/a (Berardelli et al., 2001)

Rigidity Impaired movement of the joints Joints (Berardelli, Sabra and

Hallett, 1983; Prochazka et al., 1997)

Akinesia Impaired control of voluntary movement (Pascual-Leone et al.,

1994a; b)

Dystonia Muscle spasms and abnormal muscle tone causing uncontrolled movements

Widespread (Tolosa and Compta, 2006;

Poewe, Lees and Stern, 1988)

Abnormal gait Gait is shorter than normal, and can

develop to become shuffling. n/a

(Bloem et al., 2004; Rogers, 1996; Olson, Lockhart and Lieberman, 2019; Morris et

al., 2019)

Impaired posture

A consequence of the rigidity, dytonia and akinesia

n/a (Benatru, Vaugoyeau and

Azulay, 2008; Dietz, Berger and Horstmann, 1988)

Table 1.2: Non-motor symptoms of Parkinson’s disease.

Symptom Definition Reference

Dementia Cognitive decline including memory and attention, and increased confusion. Reduced independence.

(Aarsland, Zaccai and Brayne, 2005; Gomperts et al., 2016)

Speech and communication

problems

Their speech may become slurred and lack of expression within their speech. Handwriting may

also become smaller and less-defined.

(Ho et al., 1999; Canter, 1963; Drotár et al., 2016)

Chronic pain Chronic pain is persistent pain for more than 3

months. Pain can be caused by musculoskeletal problems and can also be idiopathic.

(Silverdale et al., 2018; Nègre-Pagès et al., 2008b; Beiske et

al., 2009)

Fatigue Prolonged tiredness (Lou et al., 2001; Friedman and

Friedman, 1993; Friedman et al., 2007)

Anxiety Excessive and idiopathic sense of dread, worry and

with physical symptoms of breathlessness and heart palpitations

(Richard, Schiffer and Kurlan, 1996; Broen et al., 2016)

Depression Prolonged intense feeling of sadness and lack of

optimism. (Thobois et al., 2017; Maillet et

al., 2016; Hu et al., 2015)

Hallucinations Hallucinations can be visual, auditory, tactile and

illusions.

(Fenelon et al., 2000; Sanchez-Ramos, Ortoll and Paulson, 1996; Inzelberg, Kipervasser

and Korczyn, 1998; Barnes and David, 2001)

Delusions Delusions include paranoia, jealousy and

extravagance (Schrag, 2004)

Memory problems

Impaired storage of new memories or retrieval of old memories.

(Morris et al., 1988; Harrington et al., 1990; Lehrner et al., 2015)

Sleep problems Impaired sleep patterns. Can include insomnia,

nightmares, nocturia and sleep-apnoea.

(Faludi et al., 2015; Albers, Chand and Anch, 2017; Lin et

al., 2017)

Page 22: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

22

The development of PD is typically characterised by the loss of dopaminergic

neurons and in some cases, the presence of Lewy body pathology within the brain.

Although PD is considered a dopamine disorder, there is also prominent

dysfunction of other neurotransmitters such as serotonin, noradrenaline,

acetylcholine and glutamate (Xu et al., 2012; Mann and Yates, 1983; Reisine et al.,

1977; Hornykiewicz, 1981). They are discussed in further detail in regards to the

effect on pain perception in section 1.4.

In addition to the loss of dopamine and other neurotransmitters, a pathological

characteristic in PD is the presence of Lewy body pathology throughout the brain,

which are a type of protein aggregate and results in the reduction of

neurotransmitter release or neuronal death (Sulzer and Surmeier, 2013). The

presence of Lewy body pathology increases over time and has been classified by

Braak et al., (2004) to be in six stages which is outlined in Table 1.3.

Therefore, the pathological and neurotransmitter changes in the Parkinsonian brain

result in the diverse list of symptoms. Whilst the motor symptoms have been

researched extensively, the non-motor aspect of PD has been investigated less. One

of the least researched non-motor symptoms in PD is pain, yet the impact of

chronic pain on quality of life is substantial and contributes to the development of

depression, sleep disturbances and anxiety. The widespread pathophysiology and

diverse range of symptoms in PD therefore may predispose patients to

dysfunctional central processes including pain perception. First, section 1.3 will

briefly summarise the brain’s involvement in pain processing, and section 1.4 will

highlight how PD pathology could predispose patients to the development of

chronic pain.

Page 23: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

23

Table 1.3: Pathological findings of Lewy body Pathology in the Parkinsonian brain outlined by Braak et al.,

(2004)

Stage Brain location

Sub-region (if applicable)

Lewy body Pathology More details

Stage 1

Brain stem

Medulla Dorsal motor nucleus of the

vagal nerve Long unmyelinated preganglionic

fibers

Intermediate reticular zone (adjoins the dorsal motor

nucleus)

Stage 2

Brain stem

Reticular formation

Lower raphe nuclei Responsible for the synthesis of

serotonin Magnocellular portions of the

reticular portion (especially the gigantocellular reticular

nucleus)

Pons Coeruleus-subcoeruleus

complex

Descending tracts ‘Gain-setting’ system nuclei

Sparingly myelinated tracts. Involved in the pain control system

involved in the inhibition of somatosensory and viscerosensory

input.

Stage 3

Basal ganglia

Substantia Nigra pars compacta

Postero-lateral subnucelus

Melanised projection neurons. Neurons have little myelination and

are thin. Eventually 70% neurons lost. Lose

almost all melononeurons

Temporal lobe

Amygdala Central subnucleus

LNs extends throughout the central subnucleus of the amygdala and isolates it from the neighbouring

structures Basolateral nuclei

Brainstem

Pedunculopontine nucleus

Cholinergic tegmental nucleus Connections to the SN

Oral raphe nuclei Use serotonin and projects to the

striatum Basal

forebrain

Cholinergic magnocellular nuclei

Stage 4

Cerebral Cortex

The bridge between the allocortex and

the neocortex.

Anteromedial temporal mesocortex

Thick layer of LNs appear. Sparsely myelinated fibers.

Hippo-campus

Second sector Can be a marker of PD that a clinician

can use for diagnosis.

Stage 5

Cerebral cortex

Neocortex

Prefrontal High-order sensory association

regions

Premotor region

Stage 6

Cerebral cortex

Neocortex First order sensory association

regions

Primary fields (In some cases)

Page 24: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

24

1.3. BRAIN REGIONS INVOLVED IN PAIN PROCESSING

There are multiple regions of the brain involved in pain processing and which

constitute the pain network (see Figure 1.1). The main regions are regarded to be

the insula, anterior cingulate cortex (ACC), thalamus, somatosensory cortex (S1/S2),

prefrontal cortex (PFC) and the amygdala. The network integrates the

somatosensory, emotional and contextual components of the pain input. It is

important to note that these regions have multiple roles and are not unique to

nociceptive processing. The network is considered to be important for the

detection of salient stimuli within the environment (Legrain et al., 2011).

One theory of the organisation of the pain network within the brain is of two

defined pathways; the medial and lateral systems. Firstly, the medial pathway

ascends via the spinoreticulothalmic pathway and terminates in the amygdala,

hypothalamus, insula and the ACC, and is responsible for the affective and

emotional aspect of pain perception (Vogt and Sikes, 2000; Treede et al., 2000;

Scherder et al., 2005). The medial pain pathway also has projections to the PFC,

basal ganglia, and periaqueductal grey (PAG), which allows integration of the

nociceptive transmission and a link to the descending pain pathway. Secondly, the

lateral pathway ascends via the spinothalamic tract and terminates in the S1 and

S2, parietal operculum and the posterior insula. The lateral pathway reports the

intensity, duration and location of the pain stimuli (Sewards and Sewards, 2002;

Treede et al., 2000; Vogt and Sikes, 2000). However, the division of the pain

network into medial and lateral systems which have distinct roles in the perception

of pain is an oversimplification, and both systems have been shown to be involved

in the processing both sensory and affective aspects of pain.

In addition, there are descending modulatory tracts which originate in the

brainstem and can be modulated by higher cortical regions (Millan, 2002). The

descending modulation of pain is considered to be a type of top-down processing

(Pertovaara, 2013; Donaldson and Lumb, 2017). The descending tracts begin from

Page 25: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

25

the brainstem within the structures such as PAG and rostral ventromedial medulla

(RVM), with descending targets including the dorsoreticular nucleus, parabrachial

nucleus and the rostraventral medulla (Millan, 2002). The end target of the

descending pathways is the dorsal horn of the spinal cord, a site for modulation of

ascending pain transmission. The descending tracts are not independent of the

higher brain areas and receive modulatory input from the cortex, hypothalamus

and the amygdala and are therefore modulated by central processes (ie. Top-down

modulation).

Figure 1.1: Regions of the pain network taken from Bushnell, Ceko and Low, 2013. Abbreviations: PFC;

Prefrontal Cortex, ACC; Anterior Cingulate cortex, S1/S2; Primary/Secondary Somatosensory cortex, BG;

Basal Ganglia, AMY; Amygdala, PAG; Periaqueductal grey, PB; Parabrachial nucleus. The ascending tracts

from the spinal cord travel via the brainstem and terminate in regions of the midbrain, which connect to

cortical regions for the integration of pain processing.

Page 26: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

26

1.3.a. Top-Down modulation of pain

The focus of this thesis is regarding the top-down processing of pain in people with

PD. Here I will briefly summarise the different types of top-down processing and

their potential impact on the perception of pain and involvement in the

development of chronic pain.

The processing of pain is often divided into bottom-up and top-down aspects.

Bottom-up perception is stimulus-driven, whereby the stimulus influences our

perception. For example, the salience of a stimulus or the novelty of a noxious

stimulus can modulate the perceived pain intensity (Hauck et al., 2015a). In

contrast, top-down processing is subject-driven where prior knowledge influences

perception. An example of top-down processing in regards to pain perception is

attentional orientation or emotional state, or prior experience of pain. Top-down

pain processes are considered to be central to the development of chronic pain, yet

are also believed to be the key to treating chronic pain. For instance, whilst

negative expectation can cause an increase in pain (Bjørkedal and Flaten, 2012),

positive expectation such as the placebo effect (Dobrila-Dintinjana and Nacinović-

Duletić, 2011), and improving control by meditation (Zeidan et al., 2012; Brown and

Jones, 2010) can modulate pain perception via top-down processes.

For instance, the activity of the dorsolateral prefrontal cortex has been linked to the

ability to cope with chronic pain, and a reduced activity within the region has been

associated with chronic pain conditions such as FM and osteoarthritis (Brown, El-

Deredy and Jones, 2014). Abnormalities within this region are thought to contribute

to depression, Schizophrenia and other neurological mood disorders, which also

have an impact on pain management (Narasimhan and Campbell, 2010).

One of the main top-down processes which can modulate pain perception is the

orientation of attention (Miron, Duncan and Bushnell, 1989a). For instance, a

simple distraction can result in reduced pain, and focused attention can cause an

increase in pain intensity (Ruscheweyh et al., 2011). In addition, an unpleasant

Page 27: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

27

distraction has shown to alter the activity within the ACC during painful stimuli

(Phillips et al., 2003; Roy et al., 2009; Berna et al., 2010). Attention orientation also

modulates the descending pain pathways which can change the perception of pain.

The descending pain pathways receive input from regions associated with attention

processing such as the amygdala, PFC and ACC, and modulate the descending pain

systems, either facilitating or inhibiting pain transmission (Ong, Stohler and Herr,

2019)(Ossipov and Limitado, 2012). Therefore, dysfunction within the cortical and

subcortical regions, influences the descending modulatory system and can alter

pain perception (Ossipov and Limitado, 2012).

An example of how attention can alter the perception of pain is via the study of

anticipation of a painful stimulus. Anticipation results in the pain network to be

activated for the forthcoming pain as attention is oriented towards the stimulus,

and can modulate the resultant perception of the pain (Koyama et al., 2005; Wang

et al., 2008). For instance, when expectation of intensity is greater than what is

delivered, the pain perceived is closer to the expected rather than the stimulus

intensity. In contrast, a decreased pain expectation can reduce the pain intensity

perceived, thereby emphasising the influence of a positive outlook may have on

reducing pain (Schmid et al., 2013). The specific regions that are activated during

anticipation include the ACC, insula, S1/S2, PFC and the PAG (Koyama et al., 2005).

Within chronic pain conditions, such as FM and osteoarthritis, heightened

anticipation is common and correlated to increased pain sensitivity (Brown, El-

Deredy and Jones, 2014).

The mechanism by which expectation or anticipation of painful stimuli can alter the

perception of pain is yet to be fully understood. A functional magnetic resonance

imaging (fMRI) study has highlighted that the pain network regions including the

ACC, insula, thalamus, PFC were increased relative to the level of pain intensity

expected (Koyama et al., 2005). The study reported that the expectation of a lower

pain than administered resulted in a decreased level of activation of the insula, ACC

and S1, which is similar to that of a placebo response. The anticipation of a painful

Page 28: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

28

stimulus is a technique used to investigate central processing of pain, and has

highlighted abnormalities within chronic pain cohorts, and is therefore used in the

research conducted within this thesis.

1.3.b. Chronic pain

Finally, I will briefly discuss the central processing associated with chronic pain

conditions. Chronic pain is defined as persistent pain for at least 3 months and can

be linked to a primary condition such as osteoarthritis, or can have an idiopathic

cause. Living with chronic pain can be debilitating, with severity ranging from mild

to excruciating, and with persistent or relapsing states (Von Korff et al., 1992). The

development of chronic pain has been associated with atypical ascending and

descending pathways, along with abnormalities in top-down processing of

nociceptive information. For instance, the anti-nociceptive descending pathway of

pain is altered in many chronic pain states (Porreca, 2002; Banic et al., 2004;

Gebhart, 2004) and may be important in the persistence of the chronic pain

(Gebhart, 2004).

Numerous experiments have investigated pain processing within different pain

conditions and it is well-established that patients with chronic pain conditions such

as Fibromyalgia (FM), arthritis and back pain, have reduced pain thresholds

compared to controls (Wolfe et al., 1995; Huskisson and Hart, 1972; Imamura et al.,

2013). The establishment of reduced pain thresholds in chronic pain conditions, and

the persistence of the pain, is indicative of aberrant top-down processing. For

instance, FM, which is characterised by widespread pain with no pathophysiological

explanation, is considered to be a partly due to central sensitisation and abnormal

top-down processing (Guymer and Littlejohn, 2007). FM is the most prominently

researched chronic pain condition in regards to the role of top-down processing in

its development – therefore, it is important to review the current research for

comparison with pain processing in PD. The top-down processes include an

imbalance of neurotransmitters and neuroendocrine factors, and dysfunction of the

Page 29: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

29

hypothalamic-pituitary-axis (HPA) (Guymer and Littlejohn, 2007). FM patients have

also shown augmented brain activity within regions associated with pain perception

during the anticipation and perception of pain (Brown, El-Deredy and Jones, 2014).

Further evidence of central dysfunction includes; reduced dopamine release within

the basal ganglia (Wood et al., 2007b), reduced activation of the ACC and

descending inhibitory pain pathway (Jensen et al., 2009), and increased rates of

pain catastrophising within the FM population which correlates with altered brain

activity (Gracely et al., 2004). The treatment of FM is limited and an increasing

focus is being placed on the positive effects of alternative, non-pharmacological,

treatments such as meditation and cognitive behavioural therapy due to their

ability to improve top-down processes (Kaplan, Goldenberg and Galvin-Nadeau,

1993; Pleman et al., 2019; Montero-Marín et al., 2018; Aman et al., 2018; Amutio et

al., 2018). This emphasises the role of the brain in the manifestation of persistent

pain and why treatments for chronic pain which are targeted at central processing

are promising.

To summarise, the baseline state of the brain, be it healthy or negatively affected

by a condition such as depression or catastrophizing pain, effects the perception of

pain. Additional to the baseline state, the expectation and level of attention can

alter the pain perception, as does the context of the pain. Chronic pain states can

alter the brain’s structure and neurotransmitter levels which further impact on the

processing of pain, producing a negative cycle that can progress to worsening of the

chronic pain state. Therefore, irrespective of whether the central dysfunction is the

cause or consequence of the chronic pain state, the top-down modulation of pain is

central to deterioration, and yet also very important in the treatment of pain.

1.4. PATHOLOGICAL PREDISPOSITION OF PAIN IN PD

The pathophysiology of PD has widespread affects throughout the brain and

includes regions of the pain network and alteration in the level of

neurotransmitters which are also evident in chronic pain conditions. This section

Page 30: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

30

will highlight how the abnormalities seen in the PD brain can cause alteration in

pain processing and hence contribute to the development of central sensitisation

and chronic pain.

1.4.a. Pain network and Parkinson’s disease

Firstly, the regions associated with pain perception outlined in Figure 1.1 have been

shown to be abnormal in PD patients. For instance, the activation of the insula and

the ACC, regions of the medial pain system, are increased in PD patients following a

painful stimulus compared to healthy participants (Brefel-Courbon et al., 2005). The

augmented activation was shown to normalise following the administration of

Levodopa, a common drug used to treat PD, which increases the level of dopamine

and highlights the potential role of dopamine. The lateral pain pathway involves the

somatosensory cortex and has been associated with abnormal somatosensory

processing in PD patients, and affects their sensorimotor integration (Conte et al.,

2013; Lewis and Byblow, 2002; Rossini, Filippi and Vernieri, 1998).

Furthermore, the impaired function of the basal ganglia is central to PD motor

symptoms, however, there is extensive evidence of the basal ganglia’s involvement

pain processing and chronic pain states (see review: Borsook et al., 2010). The basal

ganglia receive input from the ascending pain spinothalamic tract and have diverse

connections with regions associated with pain processing; and is important for the

integration of motor, cognitive, emotional and autonomic aspects of pain

processing (Borsook et al., 2010). Additionally, there is an abundance of opioids and

their receptors, which are responsible for endogenous analgesia, within the basal

ganglia (Barceló, Filippini and Pazo, 2012). The basal ganglia also have roles in

attention (Van Schouwenburg, Den Ouden and Cools, 2010), emotion (Lanciego,

Luquin and Obeso, 2012), and avoidance behaviours (Hormigo, Vega-Flores and

Castro-Alamancos, 2016), all of which play a part in the perception of pain.

Aberrant function of the basal ganglia has been associated with chronic pain

conditions such as FM (Shokouhi et al., 2016), chronic regional pain syndrome

Page 31: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

31

(CRPS) (Azqueta-Gavaldon et al., 2017) and burning mouth syndrome (BMS)

(Jääskeläinen, 2012).

This section will summarise studies which have investigated the role of the basal

ganglia nuclei in pain processing, and hence highlight possible explanations of the

high prevalence of chronic pain in PD.

Firstly, the degeneration of the dopaminergic neurons in the substantia nigra pars

compacta (SNpc) within PD patients is the most significant change within the basal

ganglia and an early investigation reported that the SN consisted of approximately

50% nociceptive neurons (Pay and Barasi, 1982). The SN is connected to regions of

the pain network and is shown in Figure 1.2. The degeneration of the SNpc is

therefore likely to affect pain processing

Figure 1.2: The SN and its connections to the regions of the pain network. Figure taken from (Wasner and

Deuschl, 2012). S1: Primary Somatosensory Cortex, S2: Secondary Somatosensory Cortex. The Substantia

nigra is connected, either directly (black line) or indirectly (red lines), to regions of the pain matrix. The red

and yellow boxes highlight the pain-processing regions.

Page 32: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

32

The SN has also been associated with the descending inhibition of pain. The

stimulation of the SN has been shown to increase the descending inhibition, as did

the selective stimulation of PAG and the raphe nuclei (Barnes, Fung and Adams,

1979). The study also investigated the role of dopamine and serotonin on the

inhibitory effect and highlighted that it relied on dopaminergic neurons of the SN

innervating the serotonergic neurons within the raphe nuclei. Therefore, the

substantial loss of dopamine neurons within the SN in people with PD indicates a

possible impaired function of the descending inhibition of pain.

Within the basal ganglia, the striatum, made up of the putamen and caudate

nucleus, has been linked to pain transmission. The striatum is involved in cortical

and subcortical loops which are involved in both bottom-up and top-down

processing. A study using anaesthetised rats showed that the activation of the

peripheral nociceptors caused an increase in the activity of the nociceptive neurons

in the caudate nucleus and the putamen (Chudler, Sugiyama and Dong, 1993). The

caudate nucleus and the putamen of the rat have been analysed to show that there

are different types of nociceptors ranging from; wide-dynamic range, nociceptive

specific, and inhibited neurons (Chudler, Sugiyama and Dong, 1993). The blood

flow in the striatum has also been shown to increase when the contralateral hand

receives a painful stimulus (Barceló, Filippini and Pazo, 2012).

As previously mentioned, the basal ganglia is home to a high level of opioid activity;

this being especially high in the striatum. This leads to a strong belief that the

striatum is required for the endogenous analgesia generated by opioid activity.

Patients with arthritis have been shown to have a high abundance of opioid

receptors and a low binding availability within the striatum, which is thought to be

an adaptive mechanism (Brown et al., 2015).

Page 33: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

33

A high degree of dopaminergic transmission occurs within the striatum, yet is

impaired within people with PD. Further detail of how this impacts on pain

processing is discussed in section 1.4.

A study by Starr et al., 2011, highlights the putamen’s role in pain processing and

provides evidence that the putamen is involved in sensory processing related to

pain. The study reported that stroke patients with selective lesions of the putamen

had a reduced activation of the pain matrix and alterations of pain thresholds

depending on the type of stimulus. Within the healthy controls, it was shown that

the putamen was involved in the cortical-basal-ganglia-thalamic-cortical loop and

connections to the ACC, insula and the thalamus during painful stimulation. These

connections proposed the theory that the putamen was involved in the memory,

attention and emotional aspects of pain transmission (Starr et al., 2011). Therefore,

the putamen appears to play an important role in pain processing within the brain,

including the pain threshold levels and the cognitive aspects of pain perception.

Hence, the atypical striatal activity in PD may impact on the role of the putamen in

pain regulation.

The second region within striatum is the caudate nucleus and there is evidence that

the caudate nucleus is involved in both acute and chronic pain. Firstly, the

conscious act of suppressing electrically induced acute pain was shown to trigger a

consistent bilateral activation of the caudate along with contralateral activation of

the insula (Wunderlich et al., 2011). In contrast, heat pain did not show this strong

activation pattern (Wunderlich et al., 2011). The caudate nucleus’ role in pain

modulation is further supported by a study reporting its role in pain-avoidance

(Koyama, Kato and Mikami, 2000). The study involved macaque monkeys and

focused on the ACC, a well-known region of the pain network, and the caudate

nucleus (Koyama, Kato and Mikami, 2000). Both regions were activated during the

task, resulting in the idea that the caudate nucleus is functionally related to the ACC

during pain transmission. Hence, the caudate nucleus might be associated with the

ACC role of processing the affective component of pain (Besson, Guilbaud and Ollat,

Page 34: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

34

1995). Furthermore, there was early research within rats that demonstrate that the

caudate nucleus is involved in avoidance processing (Kirkby and Kimble, 1968;

White and Rebec, 1993).

Finally, the globus pallidus, a region of the striatum affected by PD, is involved

primarily in the control of voluntary movement and has been shown to be involved

in pain transmission (Braz et al., 2005; Chudler, 1998). By using a tracing agent, the

globus pallidus was shown to be a target for peripheral non-peptide nociceptors,

whereas the peptide nociceptors did not terminate within the globus pallidus,

indicating two separate ascending pathways depending on nociceptor type. There is

evidence that the globus pallidus within the PD brain responds to thermal and

mechanical noxious stimuli (Belasen et al., 2016). The deep brain stimulation of the

globus pallidus (Loher et al., 2002), or the bilateral removal of the region in PD

patients results in pain relief (Favre et al., 2000).

1.4.b. Basal ganglia connections to regions of the pain matrix

The basal ganglia have numerous connections to other regions which include those

that are part of the pain network such as; the thalamus, insula, ACC and the S1/S2.

There are also regions that are involved in the descending modulation of pain

transmission which the basal ganglia is connected with. The following section

discusses the important connections that the basal ganglia make with these regions

of the pain network, and how they can be disrupted in PD.

The thalamus, a main output of the basal ganglia, is dysfunctional in PD and hence

important to discuss its role in pain processing (Halliday, 2009; Pifl, Kish and

Hornykiewicz, 2012). The thalamus receives input from the globus pallidus and

substantia nigra pars reticulatar (SNpr), and is a major output of the basal ganglia.

The basal ganglia afferents innervate the ventral nuclear and the intralaminar group

(centromedian) of the thalamus. The ventral nuclear group projects to the motor,

pre-motor and supplementary motor area (SMA), whereas the intralaminar group

projects to the motor cortex and the striatum within the basal ganglia. The

Page 35: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

35

thalamus receives input from the ascending nociceptive tract such as the

spinothalamic tract which terminates in the lateral nuclei of the thalamus, and is

deemed to be part of both the medial and lateral pain pathways within the brain.

The thalamus innervates higher cortical regions including the insula, ACC and the

S1/S2, along with the striatum of the basal ganglia and descending afferents to the

brainstem. Thus, due to the thalamus being a major output of the basal ganglia and

the altered activity of the thalamus in PD patients (Halliday, 2009) may impact on

the thalamic role in nociception.

The basal ganglia is also connected to the insula which is central pain perception

(Flynn, 1999). The insula projects to the basal ganglia for the integration of motor,

somatosensory and vestibular processing (Flynn, 1999). The insula specifically

targets the striatum and the globus pallidus, and the afferents which terminate in

the dorsolateral striatum are deemed to be for somatosensory integration which is

involved in pain processing (Starr et al., 2009; Kim et al., 2017). Hence, the reduced

connectivity between the substantia nigra pars compacta (SNc), a region of the

basal ganglia, and the insula in PD patients is likely to have an impact on the ability

to integrate information during pain processing (Flynn, 1999; Wu et al., 2012). In

addition, the pain processing region of the ACC innervates the ventral striatum

(Alexander, DeLong and Strick, 1986) and has shown to be functionally correlated

with the basal ganglia (Margulies et al., 2007).

The basal ganglia is also connected to the periaqueductal gray, a region of the

midbrain and the main control centre for descending pain modulation (Northoff,

2013; Behbehani, 1995). Within a rat model of PD, a defective pathway between

the Substantia nigra, PAG and rostral medulla oblongata, has been shown (Lima et

al., 2018) and indicates that the disruption of the basal ganglia afferents may lead

to altered nociceptive modulation via the descending pain pathway.

Page 36: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

36

1.4.c. The Brainstem changes in PD and pain processing

The brainstem is one of the earliest regions to be affected by Lewy body pathology

in PD (Braak et al., 2004), and as pain has been reported to precede motor

symptoms, the brainstem could be involved in the development of chronic pain in

PD.

Firstly, descending tracts are affected by Lewy body pathology during Braak stage 2

which has been linked with pain hypersensitivity and indicates that the descending

pain modulation could be altered in PD patients (Latremoliere and Woolf, 2009).

The pons, medulla (Gebhart, 2004) and reticular formation (Casey, 1980; Bowsher,

1976) are also involved in the descending modulation pain system and thus the

Lewy body pathology in these regions may contribute to atypical pain processing.

Additionally, the nucleus raphe magnus (NRM) is highly populated with

serotoninergic neurons and is involved in both movement and endogenous

analgesia within the healthy brain. However, in PD, the NRM has been shown to

degenerate and thus likely to inhibit the endogenous analgesic response to

nociceptive input (Gai, Blessing and Blumbergs, 1995). For instance, a lesion within

the regions of the brainstem can alter pain sensitivity and rodent studies have

shown how lesions impact the action of opiate related analgesia (Abbott and

Melzack, 1982; Basbaum and Fields, 1984). For instance, the lesion of the median

raphe nucleus or central tegmental nucleus induced a potentiation of the morphine

response; whereas the loss of the nucleus raphe magnus lead to an attenuation of

the morphine response (Abbott and Melzack, 1982).

Additionally, an interesting link between pain and PD is how the activation of the

motor cortex is believed to disinhibit the PAG by inhibiting the GABAergic

interneurons and thus stopping their inhibition on the descending anti-nociception

pathway (Pagano et al., 2012). Therefore, this interesting finding implies that the

reduced motor cortex activity in PD patients may lead to a reduced disinhibition of

the PAG, and thus a reduced activation of the descending anti-nociception

Page 37: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

37

pathway. This would also help to explain the reason why Levodopa improves pain

states in PD as it increases the motor cortex output.

1.4.d. Cortical changes and pain processing

The final stages of PD involves the deterioration of the cerebral cortex leading to

cognitive decline and damage to the hippocampus contributing to loss of memory

and the development of PD dementia (Braak et al., 2003). The changes in the higher

cortical regions will impact on the complex emotional and attention related aspects

of nociception processing. As discussed in section 1.3, the top-down processes

involved in pain have a significant impact on the development of chronic pain.

1.4.e. Neurotransmitter changes in Parkinson’s disease linked with

pain

There are prominent changes in the levels of neurotransmitters in PD and abnormal

neurotransmitter levels are related to chronic pain states such as Fibromyalgia

(FM), Burning Mouth Syndrome (BMS) and Chronic Regional Pain Syndrome (CRPS)

(Stahl, 2009). As such, this section will look at the role of neurotransmitters in pain

processing and how abnormalities in neurotransmitter levels within PD may impact

pain transmission.

1.4.e.i. Dopamine

PD is characterised by the degeneration of the SNpc, a highly dopamine abundant

region, and the resulting decrease in dopamine produces both the motor and non-

motor symptoms. In addition to dopamine’s role in regulating movement, there are

theories of dopamine being central to the precision of prediction and the salience

value of stimuli, and is considered to be involved in the integration of bottom-up

and top-down processing (Friston et al., 2012; FitzGerald, Dolan and Friston, 2015;

Shiner et al., 2015). Dopamine is therefore important in the top-down modulation

of pain perception and dysfunctional transmission in PD may lead to impaired pain

processing. Figure 1.3 summaries the importance of dopamine in pain perception.

Page 38: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

38

Figure 1.3: A summary of the various roles of dopamine and how they are involved in pain perception. DA:

Dopamine, BG: Basal ganglia, SN: Substantia Nigra.

Dopamine is a modulatory neurotransmitter due to its role in both excitation and

inhibition of neural activity. There are two main post-synaptic dopaminergic

receptors, the D1 (subtypes D1 and D5) (excitatory) and D2 (subtypes D2, D3 and D4)

(inhibitory) receptors, plus pre-synaptic D2 receptors which regulate the release of

dopamine into the synapse. Dopamine receptors are found widespread, however,

there are most abundant in the basal ganglia. Within a human study, the D1Rs were

shown to be more widespread than the D2Rs and more abundant within the

dopaminergic pathways, especially in the cortex and limbic system (Hall et al.,

1994). The D2Rs have a higher abundance in the thalamus in comparison to the

D1Rs, whilst both receptors are co-localised and highly abundant in the striatum

Page 39: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

39

(Hall et al., 1994). Furthermore, a rodent study revealed higher levels of D2Rs in the

midbrain compared to D1Rs (Meador-Woodruff et al., 1991). There are four well

defined dopaminergic pathways within the brain, which originate from the SN,

ventral tegmental area (VTA) and hypothalamus (see Figure 1.4).

Figure 1.4: Dopaminergic pathways. [Anatomical locations not precise]. Blue: Mesocortical; Ventral

Tegmental area (VTA) to Prefrontal cortex (PFC). Orange: Mesolimbic; Ventral Tegmental area (VTA) to

Nucleus Accumbens (NAc). Pink: Nigrostriatal; Substantia Nigra pars compacta (SNpc) to the Striatum (STR).

Purple: Tuberinfundibular; Arcuate nucleus (Arc) of the Hypothalamus (Hyp) to the Pituitary gland.

Within healthy subjects, a PET study detected an increased D2R dopaminergic

activity in the basal ganglia and connecting pathways during pain perception (Scott

et al., 2006). The study identified different roles of the nigrostriatal and mesolimbic

pathways; the nigrostriatal pathway was associated with the pain intensity

perceived, whilst the mesolimbic pathway was associated with the affective

processing such as unpleasantness ratings and the emotional aspect of the pain

(Scott et al., 2006). In addition, the binding capacity of D2R/D3Rs within the

Page 40: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

40

putamen of the striatum have been shown to be inversely correlated to individual

pain thresholds (Pertovaara et al., 2004). Therefore, dopamine has the potential to

modulate the pain intensity perceived whilst also being susceptible to affective

aspects on pain perception.

The dopaminergic nigrostriatal pathway has been regarded to be involved sensory

gating of nociceptive stimuli and the development of chronic pain (Jääskeläinen et

al., 2001). The nigrostriatal pathway originates in the substantia nigra and

terminates in the dorsal striatum; both regions are within the basal ganglia. Altered

dopaminergic transmission within the nigrostriatal pathway has been shown to

contribute to burning mouth syndrome (BMS) (Hagelberg et al., 2003b) and atypical

facial pain (Hagelberg et al., 2003a). BMS patients have also been reported to have

a reduced level of presynaptic dopaminergic function within the right putamen, a

nuclei of the striatum (Jääskeläinen et al., 2001).

Chemical lesions of the substantia nigra to reduce dopamine levels in rats has

shown to lead to an increase in pain sensitivity (Saadé et al., 1997). Furthermore,

lesions of the substantia nigra in rats was shown to induce allodynia following a

mild stimulation to the orofacial region (Dieb et al., 2014), and subsequent

administration of a D2R agonist (Bromocriptine) remediated the allodynia. The

study also demonstrated how the loss of the dopaminergic nuclei of the substantia

nigra caused a reduced level of neurons within the striatum and the VTA (Dieb et

al., 2014). Therefore, the degeneration of the dopaminergic neurons of the

substantia nigra pars compacta within PD has an impact on the abundance of

neurons within other regions, and there is evidence that this will lead to pain

sensitivity.

Furthermore, the dopaminergic mesolimbic and mesocortical pathways have been

linked to nociceptive processing. The main focus of research of the mesolimbic and

mesocortical pathways in PD is in regards to their role in regulating reward and

gambling behaviours (Berridge and Robinson, 1998; Anselme and Robinson, 2013).

Page 41: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

41

It is common that PD patients have impaired hedonistic regulation and suffer from

pathological gambling due a dysfunction of the pathways (Torta and Castelli, 2008).

However, chemical lesions of the VTA (the origin of both pathways) have also

proven to increase the pain sensitivity in rats during standard pain tests (Saadé et

al., 1997). Analgesics which increase pain thresholds, such as opioids and

amphetamine, increase the abundance of dopamine in the nucleus accumbens

(Altier and Stewart, 1999). Hence, a low endogenous level of dopamine in PD may

result in the opposite and lower pain thresholds.

The mesolimbic pathway is also known as the ‘reward pathway’ and has also been

reported to be involved in pain. The naming of the pathway as the ‘reward

pathway’ is an oversimplification as the pathway is involved in the regulation of

both aversive and pleasurable stimuli (Leknes and Tracey, 2008; Taylor et al., 2016).

The mesolimbic pathway has been shown to activate during noxious thermal stimuli

(Leknes and Tracey, 2008). An fMRI study highlighted that the activation of regions

within the mesolimbic pathway preceded the somatosensory activation during

thermal pain (Becerra et al., 2001). Furthermore, the mesolimbic pathway is

involved in the placebo response as the dopaminergic reward system is involved in

the successful placebo effect (Borsook et al., 2010) and dopaminergic activity within

the ventral basal ganglia has been reported to increase during a successful placebo

response of pain (Scott et al., 2008; Schweinhardt et al., 2009). There is also a

positive correlation between the level of dopamine activity and effectiveness of the

placebo (Scott et al., 2008).

The final dopaminergic pathway, the mesocortical pathway, has reduced dopamine

levels in PD (Ford, 2010). The mesocortical dopaminergic pathway is associated

with emotional, attention and motivational processing (Levy, 1991; Bertolucci-

D’Angio, Serrano and Scatton, 1990; Bromberg-Martin, Matsumoto and Hikosaka,

2010). All three aspects are involved in pain processing and can manipulate the

perception of pain. For instance, a rodent study concluded that the mesocortical

Page 42: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

42

pathway increased the release of dopamine within the PFC via D2Rs mechanisms,

and resulted in a reduction in pain (Sogabe et al., 2013).

Therefore, dopamine transmission within the four pathways has been associated

with a negative effect on pain perception. Thus, the reduced production of

dopamine in PD is likely to contribute to the dysfunctional pain processing and a

susceptibility of chronic pain in the disease.

In addition to the altered activity within the dopaminergic pathways, the altered

level of dopamine leads to an altered abundance and activation of the

dopaminergic receptors within PD (Ryoo, Pierrotti and Joyce, 1998; Brooks et al.,

1992) and further disrupts the role of dopamine in pain processing.

For instance, the D2 subtype of dopamine receptors has been associated with pain

transmission and the administration of D2R agonists and antagonists have been

shown to modulate pain perception. For example, pain was reduced in rats

following an injection of a D2R agonist (Quinpirole) into the nucleus accumbens

(Taylor, Joshi and Uppal, 2003), which was blocked if preceded by a selective D2

antagonist (Raclopride) (Taylor, Joshi and Uppal, 2003). In addition, the abundance

of D2 receptors within the putamen and ratio of D1 and D2 receptors within the

striatum have been suggested to have a relationship with atypical facial pain

syndrome (Hagelberg et al., 2003a).

The abundance of D2-receptors is altered in PD, with studies reporting variability

(Brooks et al., 1992), and a 15% increase in abundance (Ryoo, Pierrotti and Joyce,

1998). The level of D2 binding potential in the putamen and the right medial

temporal cortex has shown an inverse correlation to cold pain thresholds

(Hagelberg et al., 2002). Participants who had a low D2 binding potential reported a

high pain threshold, whereas subjects with high D2 binding potential reported low

pain thresholds. In contrast, when heat and cold induced pain was administered

concurrently, low binding potential correlated with a low pain threshold and vice

versa (Hagelberg et al., 2002). Although the study’s conclusions were speculative,

Page 43: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

43

they suggested that low D2 binding potential was due to a high level of dopamine,

and the high dopamine abundance correlated with a high pain threshold.

Therefore, the study supports the fact that in PD, a low level of dopamine would be

correlated with subsequent low pain threshold for cold stimuli.

Further evidence of how abnormal dopamine signalling may contribute to chronic

pain is how patients with FM have atypical dopamine signalling. A PET study which

recorded D2 and D3 receptor activity reported that the FM patients did not show

dopamine release within the basal ganglia in response to pain, whilst the control

subjects showed a release of dopamine which was positively correlated to the

intensity of pain (Wood et al., 2007b). This study highlights how dopamine

transmission within the basal ganglia is abnormal in FM patients, and this provides

strong evidence that the low dopamine levels and dysfunctional basal ganglia

activity in the PD basal ganglia contributes to the development of persistent pain.

1.4.e.ii. Serotonin

Similar to dopamine, serotonin is a modulatory neurotransmitter with effects seen

in a diverse range of processes such as cognition, mood and stress (see review:

Olivier, 2015). Serotonin dysfunction is associated with the manifestation of

depressive and anxiety related disorders (Young et al., 1985; Cannon et al., 2006,

2007). It is well-known that low mood is correlated with chronic pain and

exacerbates the intensity and unpleasantness of pain (Bair et al., 2003; Giesecke et

al., 2005).

Serotonin dysfunction is present in PD and has been associated with the

development of non-motor symptoms (Poewe and Luginger, 1999; Politis and

Niccolini, 2015; Langston, 2006; Kish, 2003; Bernheimer, Birkmayer and

Hornykiewicz, 1961). The raphe-nuclei which produces serotonin degenerates in PD

and the neuronal loss contributes the low serotonin levels in the PD brain

(Hornung, 2003; Braak et al., 2003; Halliday et al., 1990). A PET study has shown

that serotonin dysfunction is widespread and amongst other regions, was present

Page 44: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

44

in regions associated with the pain network, including the thalamus, ACC, PFC,

insula and amygdala (Politis et al., 2010). PD patients also have a lower serotonin

level within the cerebrospinal fluid which has been correlated to the presence of

depression (Mayeux et al., 1988). Serotonin is involved in the modulation of pain

via the descending inhibitory pain pathways (Ossipov, Morimura and Porreca, 2014;

Millan, 2002; Bowker et al., 1983) and the development of chronic pain has been

associated with reduced serotonergic activity within the descending pathways

(Kwon et al., 2013; Ossipov, Morimura and Porreca, 2014). Serotonin has a

modulatory role on pain processing, and depending on the receptor subtype, can

induce either analgesia or algesia (Sommer, 2004). For instance, selective-

serotonergic reuptake inhibitors (SSRIs), a common anti-depressant, increases

serotonin transmission and can evoke an analgesic effect (Sansone and Sansone,

2008). Additionally, a lower level of serotonin has been linked with FM (Wolfe et al.,

1997).

Therefore, the dysfunction of serotoninergic transmission within PD is likely to

impair the anti-nociceptive descending pain pathways, and negatively affect the

contextual-affective aspect of pain processing. Importantly, there is a link between

depression and the prevalence of chronic pain PD, however, it is difficult to

differentiate between cause or consequence (Ehrt, Larsen and Aarsland, 2009).

1.4.e.iii. Noradrenaline

In addition to Dopamine and Serotonin, the level of noradrenaline is reduced in PD

(Scatton et al., 1983) due to degeneration of the locus coeruleus, the main source

of noradrenaline production (Reches and Meiner, 1992; Mavridis et al., 1991;

Mann, 1983; Benarroch, 2009; Del Tredici and Braak, 2013). In addition, there is

widespread Lewy body pathology in the brain stem nuclei including the medulla,

pons and descending tracts which are involved in descending pain inhibition (Braak

et al., 2003). A reduction in the abundance of noradrenaline due to the presence of

Lewy body pathology in the locus coeruleus has been reported in the PD brain (Del

Tredici and Braak, 2013).

Page 45: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

45

Noradrenaline is involved in nociceptive processing within the spinal cord,

brainstem nuclei and higher brain regions (see review: Pertovaara, 2006).

Noradrenaline is present in the nociceptive processing nuclei within the brainstem

which are important for descending pain pathways, and noradrenergic efferents to

the amygdala are involved in the effect of emotion on pain perception (Strobel et

al., 2014). Within the spinal cord, noradrenaline can suppress the nociceptive input

via the descending pain pathways (Pertovaara, 2006).

Noradrenaline is theorised to mainly play a role in persistent pain, whereby

sustained pain activates noradrenergic mechanisms to inhibit pain transmission.

Research has also highlighted how chronic pain can induce atypical noradrenergic

transmission (Alba-Delgado et al., 2013). Furthermore, the role of noradrenaline is

involved with the modulation of behavioural states and as such is considered to be

involved in top-down modulation of pain (Strobel et al., 2014; Pertovaara, 2006;

Hirata, Aguilar and Castro-Alamancos, 2006; Onur et al., 2009). The combination of

chronic pain and noradrenaline dysfunction can lead to negative emotional state

such as anxiety and depression (Alba-Delgado et al., 2013), which also increases

pain sensitivity and reduces the ability to cope with chronic pain (Geisser et al.,

1994).

Hence, the abnormal noradrenergic transmission triggered by PD results in a

disruption in its normal role in the pain processing and predisposes PD patients to

be more susceptible to develop chronic pain.

1.5. IMAGING STUDIES SHOWING ALTERED CENTRAL PAIN

PROCESSING IN PARKINSON’S DISEASE

The changes in the PD brain discussed in Section 1.4. demonstrate how PD patients

may be predisposed to the development of chronic pain. Therefore, there has been

a clear scientific rationale to research the mechanisms of pain in the Parkinsonian

Page 46: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

46

brain via imaging techniques. Here I will review the current neuroimaging results of

research into central pain processing in PD.

Firstly, PET imaging research has shown that PD patients had a higher activation

within the prefrontal cortex, insula and ACC during thermal pain (Brefel-Courbon et

al., 2005, 2013). PET has also highlighted reduced activity within the PFC and insula

during the cold pressor test, and an increase within the ACC (Brefel-Courbon et al.,

2013). Within both experiments, the differences in activation were normalised

after taking L-DOPA.

fMRI has also been useful in understanding pain processing in PD. A recent fMRI

study investigating the descending pain pathway in drug-naive PD patients and

reported that the PD patients had dysfunctional activity and connectivity during

anticipation of pain. Regions affected included the PFC, ACC and MCC which are

associated with the top-down modulation of descending pain inhibition (Forkmann

et al., 2017). The study by Forkmann supports the theory that the descending pain

pathway is impaired in PD. Furthermore, the latest investigation by Tessitore et al.,

(2018) showed significant BOLD signal changes in the drug-naïve PD brain following

heat stimulation. The study concluded that there was an increased activation within

the cerebellum, somatosensory cortex and pons of the brainstem compared to

healthy controls (Tessitore et al., 2018).

Finally, a study used EEG to assess the brain response to a range of nociceptive

stimuli in PD patients, off and on their medication, in comparison to healthy

controls (Priebe et al., 2016). Priebe showed that brain activations did not correlate

with pain rating in the PD group, and concluded that amplitude and frequencies of

brain responses were lower in PD. In contrast, the latency of brain activation was

increased in the PD cohort and together with increased pain sensitivity, supports

the theory that central pain processing is dysfunctional in the PD.

Page 47: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

1. Introduction

47

1.6. CONCLUSION

Although the general consensus is often that chronic pain is driven by peripheral

nociceptive dysfunction, there is strong evidence that many chronic pain states are

due to disruption to central mechanisms. The high prevalence of chronic pain in PD

patients and the presence of idiopathic and widespread pain unrelated to the

musculoskeletal symptoms present a foundation to investigate the central

mechanisms involved in PD pain. There are numerous regions that are mutually

affected by PD pathology and are involved in pain processing within the healthy

brain. Furthermore, the development of pain in PD has been shown to precede the

onset of motor symptoms which may be correlated to the early Lewy body

pathology within the brainstem. Research has shown that the perception of pain is

significantly modulated by central processing and disruption to the somatosensory,

emotional and cognitive networks have shown to lead to hyperalgesia,

catastrophizing states and hence, the development of chronic pain. The fact that

neurotransmitters such as dopamine, serotonin and noradrenaline are involved in

pain processing and are also irregular in PD, strengthens the theory that central

mechanisms are likely to be involved in PD pain. Therefore, to conclude, the

combination of Lewy body pathology and neurotransmitter changes within regions

involved in pain processing are likely to increase the likelihood of a chronic pain

state in PD patients.

Page 48: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Chapter 2

General Methods and Introduction to study aims

Page 49: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

49

This section will discuss the rationale behind the choice of methodology and outline

the studies which will be discussed in this thesis.

2.1. GENERAL METHODS

The research included in this thesis used electroencephalography (EEG) to record

brain activity during the anticipation and perception of laser induced acute pain.

The study design of the research was based on the protocol carried out in Brown et

al., (2014) and Brown et al., (2008). Firstly, the Brown et al., (2014) investigated the

brain activity during the anticipation and perception of pain in people with

Fibromyalgia, Osteoarthritis and healthy age-matched participants. The research

used EEG source localisation analysis to conclude regional differences in the patient

groups during the anticipation of pain, and hence demonstrated that the protocol

was sensitive to distinguishing differences in the processing of pain within patients

groups. Secondly, the Brown et al., (2008) study demonstrated the effect of

certainty on the rating of pain and neural activity. There is evidence that dopamine

is involved in the processing of uncertainty and hence was included as an aspect of

the protocol (Fiorillo, 2017).

The same experimental protocol was applied in each investigation, and as such, the

rationale for the study design is introduced in the following sections, with further

detail included in the individual papers of the thesis.

2.1.a. Experimental pain

The common techniques used to induce pain in research are via thermal,

mechanical, chemical and electrical stimulation. The choice of stimulation is

dependent on the experimental design and for the studies included in this thesis,

thermal stimulation via a CO2 laser was used (Brown, El-Deredy and Jones, 2014)

due to its quick pulse duration, small stimulation area and selective activation of

the nociceptive A and C-fibres. The laser induced pain has been used to evaluate

pain thresholds in healthy and patient groups (Gibson et al., 2001; Brown, El-Deredy

and Jones, 2014; Brown et al., 2008a; Derbyshire et al., 2002). In addition, laser

Page 50: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

50

induced pain stimuli have been used in numerous pain experiments investigating

pain anticipation (Brown, El-Deredy and Jones, 2014; Clark et al., 2008a; Wang et

al., 2008; Brown and Jones, 2010, 2008; Forkmann et al., 2017). The fast onset and

offset of the laser produces well-defined evoked responses which can be time-

locked in neuroimaging techniques and are referred as laser evoked potentials

(LEPs).

A limitation of the laser is the potential of skin sensitivity and damage due to the

heating of the skin. To limit the potential, the laser was moved in a pattern within a

grid on the forearm of the participant. The laser was also limited to a maximum

voltage that could be delivered to avoid skin damage. Nevertheless, the skin

sensitivity of the individual must be considered and the maximum voltage adjusted

accordingly, or stimulation stopped altogether in rare instances.

Pain is a subjective experience and the rating of pain can therefore be variable

between individuals. To limit the subjective interpretation of how to rate the laser

stimuli, a pain rating scale was presented to the participants and was explained

using a script by the researcher (see figure 2.1).

Figure 2.1: The pain rating scale that was used for the psychophysics and main experiments. The explanation

of the rating scale was identical for all participants to limit subjective interpretation.

2.1.b. EEG

EEG is a method used to record the neural activity by monitoring the voltage

oscillations generated by the brain. The post-synaptic activity of neighbouring

neurons produces local dipoles which are recorded by electrodes placed on the

Moderate

Pain

Severe

Pain

No

Sensation

Just

Painful

Unbearable

pain

Page 51: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

51

scalp (Baillet, Mosher and Leahy, 2001). The pyramidal neurons are the main

generators of the EEG signal due to their uniform orientation to the surface and

their long length allowing dipoles to form. EEG detects the synchronised activity of

neurons with a high temporal resolution that is significantly higher than other

imaging techniques.

As such, EEG has allowed for a deeper understanding of cognitive processing and

disease states, along with providing a diagnostic tool for epilepsy, sleep disorders

and many other diseases. The non-invasive nature of EEG allows researchers to

record the activity in a broad range of people, irrespective of their health or age.

EEG has also helped to compare the differences of electrical activity between

disease states and healthy volunteers.

2.1.b.i. The Forward and Inverse EEG Problems

EEG has two main confines, the forward and inverse problem. The forward problem

consists of how the activity within the brain is recorded on the scalp electrodes,

whilst the inverse problem is in regards to calculating the precise source of the

potential generated at the scalp. Over decades of research, more complex models

have been developed to address the forward problem, which take into

consideration; head shape, tissue heterogeneity and the difference in conductivity

(Hallez et al., 2007). There are also several techniques to resolve the inverse

problem [see review: (Grech et al., 2008)]. The LORETA (Low resolution

electromagnetic tomography) technique which is used within the research of this

thesis has been reviewed to be able to accurately identify sources, even of deep

regions (Grech et al., 2008).

The LORETA technique overcomes the inverse problem using a tomographic

solution whereby it calculates the current source density in a large cortical area to

produce a 3D volume image of neural activity. The solutions created by LORETA are

based on the assumption that neighbouring neurons respond similarly, such as the

same orientation, latency and activation. An advantage of using the LORETA

technique is that it does not require a priori estimates of sources, which are a

Page 52: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

52

requirement in other inverse solutions such as the equivalent current dipole (ECD)

technique. Therefore, LORETA is not restricted by a limited number of expected

sources.

Research into the accuracy of the LORETA technique has shown that the source

estimates are analogous to other imaging techniques which have a higher spatial

resolution. Concurrent recording of EEG and fMRI has shown that LORETA

successfully detects current source densities which are consistent and are on

average 14.5/16 mm within the fMRI loci (Mulert et al., 2004; Vitacco et al., 2002).

Similar validation has been shown using PET and LORETA (Zumsteg et al., 2005), as

have intra-cerebral recordings (Seeck et al., 1998; Trébuchon-Da Fonseca et al.,

2005). LORETA is also able to localise activity within deep brain structures such as

the insula (Mulert et al., 2004).

2.1.b.ii. Anticipatory-evoked potentials

A cue indicating a forthcoming stimulus can evoke an anticipatory response which is

detected in EEG as a negative waveform known as the stimulus-preceding

negativity (SPN) (Figure 2.2). The SPN can be triggered by both innocuous and

noxious stimuli. There is evidence to show that the SPN induced by a noxious

stimulus may be generated within the cingulate cortex, especially the anterior- and

mid- regions (Böcker et al., 2001). Therefore a more negative response during the

anticipation of noxious stimuli indicates a higher anticipatory response. The

characteristic topographic map of anticipation is a negative response over the

dorsal regions and is shown in Figure 2.2.

For the research conducted within this thesis two baselining methods were applied

to investigate the neural activity during the anticipation of pain. Initially, a single

baseline of -3500 -3000 ms was used to compare with the Early (-2500 -2000ms),

Mid (-1500ms -1000ms) and Late (-500 0ms) time windows of interest. However, as

this time window used for the single baseline was between trials and no cue was

presented yet, observations of participants highlighted that they were using this

time period to readjust their position, relax or stretch. Furthermore, the tremor of

Page 53: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

53

the participants with PD was more prominent during the inter-trial periods.

Therefore, the single baseline of -3500 to -3000ms was potentially variable and

affected by artefacts, and a secondary baselining method was applied.

Therefore, primary baselining method was adjusted to use individual baselines for

each time window of interest set as -500ms prior to the auditory beeps. The

baselines (BL) for each time window of interest (TWOI) were as follows: Early (BL: -

3500 -3000ms, TWOI: -2500 -2000ms), Mid (BL: -2500 -2000, TWOI: -1500 -

1000ms), Late (BL: -1500 -1000ms, TWOI: -500 0ms). An aim of baselining uniquely

for each anticipation window was to reduce variability in the data as the

anticipation phase progressed, such that the analysis of each phase of anticipation

was unique to that phase and not subject to variability arising from neural activity

occurring in the previous phase.

The analysis using single baseline of -3500 -3000ms was also conducted to enable

comparison to previous studies (Brown et al., 2008a; Brown, El-Deredy and Jones,

2014) that used the same baselining method. All statistical analysis for the

anticipation phase was adjusted for multiple comparisons.

Figure 2.2: (Left) An EEG waveform of an SPN generated by a visual anticipatory cue, auditory cues, and the

LEP response following acute laser pain. Diagram from (Jones, Brown and El-Deredy, 2013). (Right) An

example of a characteristic anticipatory response recorded via EEG displayed on a topographic map. Plot

displays the grand mean response of a healthy cohort during the late SPN (-500 ms) preceding a noxious laser

stimulus.

Page 54: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

54

2.1.b.iii. Suitability of EEG for research aims

EEG was chosen as the method to record brain activity for the two studies due to its

suitability in assessing pain perception which was used in Brown et al., (2014) and

other pain research studies (Clark, Brown, Jones, et al., 2008; Shao et al., 2012;

Jones et al., 2013; Hauck et al., 2015).

A benefit of EEG is that the setup is quick and does not require restricted

movement compared to fMRI. Therefore, the technique was suitable for volunteers

with PD as movement was allowed between trials and did not present possible

complications of claustrophobia. The movement constraints of other imaging

techniques meant that EEG was the optimal tool for recording brain activity in

people with Parkinson’s disease. In addition the use of EEG for pain processing has

been used extensively within pain research due to its good temporal resolution. The

research conducted in The Human Pain Research Group has developed protocol

and analysis methods using EEG to investigate the top-down modulation of pain via

the anticipation signal.

The good temporal resolution of EEG is in contrast to its poor spatial resolution in

comparison to fMRI and PET. Therefore, source localisation was a key focus for the

research to establish the sources of the brain activity recorded that would allow a

deeper interpretation of the EEG data. The LORETA source localisation technique is

used to solve the inverse problem with EEG recording (Pascual-Marqui, 2002). The

technique was selected due to its accuracy in localisation activity. In addition, due

to the LORETA technique being used in previous research conducted by The Human

Pain Research Group, the findings can be comparable to prior research.

2.2. INTRODUCTION TO STUDY OBJECTIVES

The research within this thesis was planned to investigate central pain processing in

Parkinson’s disease with the aim to improve understanding of the cause of the

higher rate of chronic pain in the disease. The first study explored pain processing

in PD patients and age-matched healthy volunteers (Chapter 3 & 4). This study was

Page 55: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

55

followed by an investigation into the role of dopamine in pain processing and was

carried out in young healthy volunteers (Chapter 5). An additional analysis was

carried out between young and old healthy volunteers to establish the age-related

differences in the response to the protocol (Chapter 6).

2.2.a. Chapters 3 & 4: Study 1: PD vs Healthy Controls

The first study investigated pain processing in volunteers with PD and age-matched

healthy volunteers. We wanted to investigate pain processing both on and off

medication within the PD group. The benefit of investigating the ‘off’ state of PD

was to remove any effect of the medication and to understand the ‘normal’ state of

the brain in PD patients. The additional study of ‘on’ medication was to highlight

whether the dopaminergic medication used for PD changed pain processing as L-

DOPA had previously been shown to have analgesic qualities. The experimental

paradigm explained below was carried out in the morning following the withdrawal

of PD medication and repeated on the same day 1 hour after PD patients had taken

their medication. The order of the two recordings could not be randomised due to

the need of 12 hours withdrawal of the medication within the ‘off’ condition. The

healthy controls also repeated the study in the afternoon after a 1 hour break. The

decision to complete the two recordings on the same day was after discussions with

potential PD volunteers and their limited willingness to complete two separate

visits. Furthermore, although the order of the visits could have been randomised,

there would have been no blinding to the study due to the use of personal

medication and the researcher’s interaction with the participant. Therefore, the

two groups completed the paradigm twice on the same day, and the healthy

control group was a control for each time-point.

The limitations of this study design became apparent after data collection as a high

proportion of the EEG datasets needed to be excluded due to high levels of noise in

the data. The reason is likely to be due to the length of the EEG cap being

connected, and the reduced quality of EEG data in the aged population. In addition,

there was a clear difference in both the PD and HC groups’ tolerance to the laser

due to the repetition of the paradigm. For this reason, the morning ‘off’ and

Page 56: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

56

afternoon ‘on’ sessions were analysed and discussed individually, without

comparison between time-points. Therefore, Chapter 3 includes the main report of

PD ‘off’ state, and Chapter 4 summarises the results of the ‘on’ state.

The protocol was based on the previous research carried out in The Human Pain

Research Group within chronic pain groups and healthy volunteers (Brown, El-

Deredy and Jones, 2014; Brown et al., 2008a). The original protocol used a CO2 laser

to induce acute pain to the forearm. The laser stimuli were preceded by a visual cue

of Low, Mid, High or Unknown which informed the participant of the forthcoming

intensity of the laser. The Unknown visual cue indicated that either intensity would

be given and allowed investigation into the effect of certainty on pain perception.

The visual cue was also concurrently presented with auditory beeps for an accurate

3 second countdown to the stimulus. The participants completed the paradigm

whilst EEG recording was taken, and both sensor level and source localisation was

used to analyse the data.

The original laser paradigm was slightly adjusted to be suitable for the PD patient

group and the protocol was consistent between studies to allow for comparison.

The number of conditions was reduced to be Low, High and Unknown, by removing

the Mid intensity condition, to reduce the time needed for the PD participants to be

sat still. The reduced time was also required to allow time to repeat the study once

the PD patients had taken their PD medication. The three main conditions were

sufficient for analysis as the inclusion of the Low and High conditions allowed for

analysis on for the effect of expectation on brain activity, and the Unknown

condition was used to establish an effect of certainty on brain activity and pain

rating. The protocol is outlined in full within the papers of the thesis.

2.2.b. Chapter 5: Study 2: D2 Dopamine Study

The second study investigated the manipulation of striatal dopamine due to its

strong links with pain and PD (discussed in section 1.4). Therefore, an agonist and

antagonist of dopamine D2Rs were selected to study both high and low levels of

dopamine transmission. The D2R was selected as the target of modulation due to

Page 57: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

57

the high abundance in the striatum in comparison to the more widespread

distribution of other dopamine receptors. The use of D2 modulation via drugs has

been shown to be successful in numerous studies (Nandam et al., 2013; Mehta et

al., 2005; Becker et al., 2013). A D2R agonist and antagonist were selected via Dr

Grace Whitaker for their selectivity, safety and efficacy. The agonist Cabergoline

and the antagonist Amisulpride were selected. The in depth comparison between

other drugs and the reason for the final choice is reported in her thesis (Whitaker,

2017).

Cabergoline is often used as a Parkinson’s treatment and has one of the highest

binding affinity to D2Rs compared with other agonists (Gerlach et al., 2003). Also,

Cabergoline has shown to be highly tolerable and safe to administer in patient

groups and experimental studies (Vilar et al., 2018; Webster et al., 1993; Gibson et

al., 2012; Nomoto et al., 1998).

Amisulpride is a selective antagonist of D2 receptors (subtypes D2 and D3) and

prescribed as an anti-psychotic for conditions such as schizophrenia (Silveira da

Mota Neto, Soares and Silva de Lima, 2002). Amisulpride has a good tolerability and

safety profile (Rosenzweig et al., 2002; Juruena, de Sena and de Oliveira, 2010),

however, Amisulpride been associated with prolongation of the QTc interval in an

electrocardiogram (ECG) (Wenzel-Seifert, Wittmann and Haen, 2011; Isbister et al.,

2010; Isbister and Page, 2013). The risk of QTc prolongation is low and will only

occur in combination with additional risk factors such as a pre-existing heart

condition or concurrent administration with other drugs (Wenzel-Seifert, Wittmann

and Haen, 2011). Therefore, all participants recruited to the study underwent an

ECG assessment to ensure that no underlying heart condition was present.

The D2R is a G-coupled receptor and activation induces an inhibitory response to

the post-synaptic neuron. The D2R is mainly located on non-dopaminergic neurons,

however, is also present on dopaminergic neurons and acts as an autoreceptor

which regulate the activity of the dopaminergic neurons (see Figure 2.3). The

activation of the autoreceptors causes a reduction in dopamine transmission and

dopamine production, and previous studies have demonstrated that low doses of

Page 58: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

58

D2R drugs favours the activation of the autoreceptors. Therefore, due to the

possible conflicting outcome of the activation of the autoreceptors (reducing

dopamine transmission), rather than the desired activation of the post-synaptic

receptors, the two drugs were administered at a relatively high dose. On the basis

of previous research, the agonist, Cabergoline was given at 1.25mg (Nandam et al.,

2013; Frank and O’Reilly, 2006), and the antagonist, Amisulpride, was given at

400mg (Rosenzweig et al., 2002).

The modulation of the dopamine levels was assessed via eye-blink rate (EBR) which

has previously been positively correlated to striatal dopamine activity and

associated with the D2R (Taylor et al., 1999; KARSON, 1983). The method to

quantify the blink rate was via the ICA function within the EEGLAB toolbox in

MATLAB. The upward and downward deflection, shape and topography of the blink

was searched for within the resting state data. The blink-rate has been shown to

reduce in Parkinson’s disease and is hypothesised to be due to the reduced

dopaminergic activity within the nigrostriatal pathway. Therefore, the blink rate of

each participant was calculated using the EEG recording during a period of resting.

In addition to the selection of the drugs, the recruitment of participants and health-

screening was completed by Dr Whitaker. The running of the study was carried out

by Dr Whitaker and myself in equal measure. All data presented in this thesis

related to the D2 Dopamine study were collected and analysed by myself. The

collaboration was organised due to the central role of dopamine in PD and the thus

a suitable follow-on to the Parkinson’s study.

2.2.c. Chapter 6: Age comparison

The same protocol was completed in Study 1 (PD v age-matched healthy controls)

and Study 2 (D2 dopamine modulation in young healthy volunteers). Therefore, a

comparison was made between the brain activity from the older healthy controls in

Study 1, and the young volunteers in the control condition in Study 2. The rationale

to compare the two age-groups was that there is limited research in the effect of

age on pain processing, especially top-down modulation.

Page 59: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

59

R

R

R

opam

ine

D2R ac va on

in ibits

postsynap c

ac va on

A to

-

re eptor

D2 Auto

-receptor

ac va on decreases

dopam

ine synthesis

and release

Presynap

ne ron

Postsynap

Ne ron

A onist

Anta onist

o doses favours

presynap c D2

auto

-receptor ac va on

i doses ac vate

postsynap c D2Rs in

addi on to presynap c

auto

-receptors

A onist

Decrease in dopam

ine

release and decrease in

postsynap c D2R

ac va on

Anta onist

Increase in dopam

ine

release and increase in

postsynap c D2R

ac va on

A onist

Increase in postsynap c

D2R ac va on despite

presynap c dopam

ine

down

-regula on.

Anta onist

Decrease in postsynap c

D2R ac va on despite

presynap c dopam

ine

up

-regula on.

Figure 2.3: A schematic diagram of the action of the dopamine D2 receptor and how low and high doses of

agonists and antagonists have different effects on postsynaptic D2 receptor activity.

Page 60: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

60

2.3. RECRUITMENT

2.3.a. Recruitment: Study 1: PD vs Healthy Controls

The recruitment of the PD group was carried out by Dr Monty Silverdale and Dr

Christopher Kobylecki at their clinics held at Salford Royal NHS Foundation Trust.

The study was introduced to people who could be temporarily withdrawn from

their PD medication and pain relief on the study visit. Patients presenting a severe

tremor or dystonia were not recruited to the study due to the EEG recording being

sensitive to movement. The contact details of those interested in taking part were

passed over to me for a follow-up phone-call. Approximately 50% of the patients

who showed interest took part in the research.

The recruitment for the HC group was via the Citizen Scientist website and adverts

within local golf clubs. The HC group were age-matched to the PD group. The

recruitment of the partners of PD patients was avoided due to previous research

demonstrating an altered pain perception within partners who had a caring role

(Romano et al., 2000).

The PD study recruited age-matched controls to act as the health control group for

the study. The HC group carried out the experiment twice to replicate the

experience of the PD group completing the study off and on their medication. The

HC also repeated the study and acted as a control for the ‘on’ condition of the PD

group.

2.3.b. Recruitment: Study 2: D2 Dopamine Study

The recruitment of the Dopamine D2 Study was via the email announcements at

The University of Manchester, and advert posters circulated in the University

buildings. The age range was selected to be 18-35 years old to limit age-related

changes which may affect the results. In addition, the criteria for the study required

a healthy cohort and as such a younger age range was appropriate. Prior to

selection for the study, the participants completed the Barratt impulsiveness scale

(BIS-11 (provided by Dr Grace Whitaker) to establish their baseline level of striatal

Page 61: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

61

dopamine as the (BIS-11) has previously been correlated with individual dopamine

levels, specifically striatal dopamine (Costa et al., 2013; Buckholtz et al., 2010;

Korponay et al., 2017). Final recruitment to the experiment was restricted to the

participants who scored in the mid-range on the questionnaire as this indicated that

their dopamine levels were also mid-range and within the peak of the inverted-u

relationship of dopamine and performance (see figure 2.4). The inclusion of this

assessment was aimed to reduce the range of the variability in baseline dopamine

levels in the participants which may have affected the outcome of the D2R

manipulation. A study by Cools et al (2009) demonstrated the importance of

baseline dopamine and D2R manipulation. The study assessed the relationship

between reward- and punishment-based learning and baseline dopamine level and

concluded using PET imaging that participants’ with high striatal baseline dopamine

performed better in the paradigm than those with low dopamine levels. They

further highlighted that the baseline dopamine level affected the result of a D2R

agonist on punishment driven outcomes; such that performance was improved by

the agonist in participants’ with low baseline dopamine, whilst impairing

performance in those with high baseline dopamine (Cools et al., 2009).

Figure 2.4: The inverted-U relationship between dopamine level and performance. There is an optimal range

of dopamine level at the peak of the curve, and a low or high level of dopamine has been shown to impair

performance in cognitive responses. The BIS-11 score of impulsivity has been correlated with baseline

dopamine levels and was used as a screening technique to recruit participants with dopamine levels within

the optimal range. This recruitment criterion aimed to limit the variability in the effect of the D2R

manipulation on performance output.

Per

form

ance

Dopamine level/BIS-11 score

Page 62: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

62

Furthermore, the Dopamine D2R antagonist Amisulpride required pre-health

screening due to it being dangerous for people with extended QTc intervals (Täubel

et al., 2017). Therefore, an electrocardiogram (ECG) was performed on all potential

volunteers by either Dr Monty Silverdale or Dr Christopher Kobylecki. All ECGs were

assessed prior to recruitment to the study and if a volunteer showed abnormalities

they would have been withdrawn from the study and their GP informed.

The study was designed to be repeated-measures and all participants completed

the experiment three times over a six week period. Each visit was separated by 10-

days to allow for the skin the recover between each laser session. The study was

pseudo-randomised and double-blinded. The randomisation was pseudo-

randomised so that near equivalent numbers were in each of the six possible orders

of the drug conditions.

The benefit of a repeated-measures design is that there is less variability within

condition groups and the highly subjective and individuality of pain perception

would have increased the variability within a between-subject design. Therefore, a

between-subject design would require a much higher n number to account for the

high degree of variability.

After each study visit the participants were given specially made business cards

with information about the study. The participants were advised to keep it on their

person as it provided information to medical personnel if an emergency arose

(Figure 2.5).

Page 63: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

2. General Methods and Introduction to Study Aims

63

Figure 2.5: Business cards given to participants of the D2 dopamine study. The participants were instructed to

carry the cards on them for 24 hours in the unlikely case that they needed medical attention and were not

able to explain their involvement in the study.

Page 64: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Chapter 3

A neurophysiological investigation of pain anticipation in Parkinson’s

disease

Sarah Martin, Anthony Jones, Christopher Brown,

Christopher Kobylecki, Wael El-Deredy, Monty Silverdale

Page 65: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

65

3.1. ABSTRACT

Chronic pain is common in people with Parkinson’s disease, and is often considered

to be caused by the motor impairments associated with the disease. Altered top-

down processing of pain characterises several chronic pain conditions and occurs

when the cortex modifies nociceptive processing in the brain and spinal cord. This

contrasts with bottom-up modulation of pain whereby nociceptive processing is

modified on its way up to the brain. Although several studies have demonstrated

altered bottom-up pain processing in Parkinson’s, the contribution of enhanced

pain anticipation and enhanced top-down processing of pain has not been fully

explored.

In the present study, EEG was recorded whilst noxious stimuli were delivered by a

carbon dioxide laser to the forearm. Participants with Parkinson’s disease were

compared with a Healthy Control group.

EEG source localisation reported an increased activation in the mid-cingulate cortex

and supplementary motor area in the Parkinson’s disease group compared to the

healthy control group during mid [-1500 -1000 ms] and late anticipation [-500 0

ms], indicating enhanced cortical activity before noxious stimulation. The

Parkinson’s disease group was also more sensitive to the laser and required a lower

energy level to induce pain.

This study provides evidence supporting the hypothesis that enhanced top-down

processing of pain may contribute to the development of chronic pain in

Parkinson’s. With further research to confirm these findings, our results inform a

scientific rationale for novel treatment strategies of pain in Parkinson’s disease,

including mindfulness, cognitive therapies and other approaches targeted at

reducing top down processing of pain.

Page 66: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

66

3.2. INTRODUCTION

3.2.a. Pain in Parkinson’s disease

Chronic pain is a highly prominent symptom in people with Parkinson’s disease

(PwPD), yet there is a limited understanding of whether the pain is primarily a

consequence of motor impairment, including muscle rigidity, or whether

Parkinson’s disease (PD) causes a centrally produced heightened sensitivity to pain.

Whilst the percentage of the general population living with chronic pain is

approximately 20% (van Hecke, Torrance and Smith, 2013; Breivik et al., 2006),

there is a significantly higher prevalence within the PD population of approximately

two-thirds (Ozturk et al., 2016; Nègre-Pagès et al., 2008a; Skogar and Lokk, 2016;

Silverdale et al., 2018). There is evidence that PwPD have lower threshold and

tolerance of pain compared to age-matched healthy cohorts (Brefel-Courbon et al.,

2005; Chaudhuri and Schapira, 2009; Djaldetti et al., 2004a; Schestatsky et al.,

2007a), and EEG and functional imaging studies have demonstrated an altered

central response to pain in PD (Tinazzi et al., 2009; Schestatsky et al., 2007b; Brefel-

Courbon et al., 2005). These abnormalities provide strong evidence that altered

central pain processing contributes to the development of pain in PD (Silverdale et

al., 2018). Therefore, an increased understanding of the pathophysiological

mechanisms causing the chronic pain is imperative to make an informed

improvement in the treatment of pain in Parkinson’s.

3.2.b. Top down alteration in pain processing

Research into chronic pain has largely focused on bottom-up mechanisms

amplifying the pain signal on its way from the peripheries to the brain (Reicherts et

al., 2017; Watson et al., 2009). However, there is a developing field in the role of

top-down modulation of pain, whereby the cortical activity modulates the incoming

pain signal as it reaches the brain.

Previous research within our group has used EEG source localisation to investigate

the anticipatory processing of painful stimuli. Our previous research has shown that

the anticipation of pain leads to changes in the activity of the same brain regions

Page 67: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

67

that subsequently respond to nociception (Brown and Jones, 2008; Brown, El-

Deredy and Jones, 2014; Clark et al., 2008b; Brown et al., 2008a). Examples of

regions which have been seen to activate during anticipation include the cingulate

cortex, insula, primary and secondary somatosensory cortex (SI/SII), prefrontal

cortex (PFC) and the periaqueductal gray (PAG) (Koyama et al., 2005; Babiloni et al.,

2004). The degree of anticipation has been shown to be correlated with subsequent

pain perception and the EEG laser-evoked potential (LEP) (Brown et al., 2008a).

Hence, we have previously demonstrated that the cortex can modulate the

perception of pain of the incoming pain signal via top-down modulation. Within

chronic pain conditions, such as fibromyalgia and osteoarthritis, heightened

anticipation within the insula cortex is correlated with the extent and severity of

chronic pain symptoms (Brown, El-Deredy and Jones, 2014).

Unlike fMRI and PET, the higher temporal resolution of EEG allows for a more

accurate recording of the anticipatory processes. Prior to a noxious stimulus, during

anticipation, there is cortical activity from a network of regions including the

cingulate cortex, basal ganglia and thalamic structures (Brunia and van Boxtel,

2001; Vogt, Finch and Olson, 1992). In this study we used EEG with source

localisation analysis because of the temporal advantage over neuroimaging

techniques reliant on slow haemodynamic responses. The accuracy and reliability of

EEG source localisation has been verified by research which shows high similarity to

other neuroimaging techniques such as PET (Christoph M. Michel, 2004; Lantz et al.,

2003), fMRI (Mulert et al., 2004; Vitacco et al., 2002) and intracerebral recordings

(Seeck et al., 1998; Trébuchon-Da Fonseca et al., 2005). EEG source localisation has

reliably reported activations within the pain network during anticipation (Brown

and Jones, 2012; Brown et al., 2008a; b). For instance, impaired processing within

the parietal and frontal regions during anticipation has been reported using source

localisation in patients with the chronic pain associated with Fibromyalgia and

Osteoarthritis (Brown, El-Deredy and Jones, 2014).

Page 68: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

68

3.2.c. Aim of study

Here we investigated anticipatory processing in PwPD in the “off” medication state

in comparison to age-matched healthy volunteers. We used a CO2 laser to induce

acute noxious stimuli and monitored brain activity throughout via EEG. We

hypothesised that the anticipatory phase would be abnormal in the PD group and

would provide evidence that top-down mechanisms are key to explaining the

mechanisms of chronic pain in PD.

3.3. METHODS

The study was approved by the local ethics committee and all participants gave

informed consent according to the Declaration of Helsinki to participate in the

study.

3.3.a. Participants

Twenty-four participants with Parkinson’s were recruited (sixteen males). PD

patients were recruited via correspondence with their neurologist (MS or CK).

Participants were screened to ensure safe withdrawal of medication for the study,

and to exclude cases of severe tremors which might interfere with EEG recording.

Clinically significant peripheral neuropathy was excluded by clinical examination

and neuropathy scale. Symptom duration ranged from 1-month to 16 years, with a

mean duration of 5.13 years. One PD participant was unable to complete the study

due to severe symptoms after medication withdrawal. Twenty-three participants

with PD were included for analysis of behavioural measures. Twenty PD participants

were included for EEG analysis due to two datasets being removed because of noisy

data and one for a low MoCA (Montreal Cognitive Assessment) score (12/30).

Twenty-five age-matched healthy controls (HCs) were recruited (fourteen males).

One HC participant was unable to complete the study due to the laser failing to

induce a sufficient pain level, and one HC dataset was removed due to noisy data.

Twenty-four HCs were included in the behavioural measures and twenty-three HC

Page 69: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

69

participants were included for EEG data analysis. The participants with PD [age

range; 63.3 ± 8.27] were age matched to the HCs [age range; 65.5 ± 8.59].

3.3.b. Medication

The PD group omitted their evening medication and were studied in the practically-

defined OFF state (after 12 hours withdrawal of anti-Parkinsonian medication). The

motor section of the Movement Disorder Society Unified Parkinson's Disease Rating

Scale (MDS-UPDRS) was completed to report the current severity of their

parkinsonian disability off their medication.

3.3.c. Assessments

The participants were assessed for; PD motor severity, pain, mood and cognitive

state. PD motor severity: The motor section (III) of the MDS-UPDRS was completed

to evaluate the motor disability due to PD (Goetz et al., 2008). The assessment in

the PD group was completed off their medication. Pain: All participants rated their

current pain state via a number rating scale (NRS) prior to starting the laser

protocol. The PD participants with chronic pain reported their minimum and

maximum degree of pain over the last 6 months via a visual analogue scale (VAS).

All participants completed the pain catastrophising scale (PCS) (Sullivan et al., 1995)

to report their psychological coping ability when experiencing pain. Mood: All

participants completed the Hospital Anxiety and Depression Scale (HADS) (Zigmond

& Snaith, 1983) to dissociate results from anxiety or depression. Cognitive state:

The Montreal Cognitive Assessment (MoCA) (Nasreddine et al., 2005) test was

carried out to assess the participants’ cognitive ability. Participants with low scores

(<25) were removed from the study to avoid cognitive decline affecting the EEG

signal.

3.3.d. Experimental design

3.3.d.i. Pain stimuli

A CO₂ laser [50W Synrad 48-5 J-series (J-48-5(S)W) Wavelength: 10600nm] was

used to deliver acute pain to the dorsal surface of the right forearm. The CO₂ laser

Page 70: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

70

delivered a beam with a diameter of 15 mm and 150 ms duration. The voltage (V) of

the laser is linearly related to the laser energy delivered to the forearm. For each

test, the stimuli were delivered in an area measuring 4 x 5cm and was delivered in a

predetermined randomised path (Brown et al., 2008a). This was to avoid

habituation, sensitization, or skin damage.

3.3.d.ii. Psychophysics

Before starting the experimental protocol, psychophysics was used to calibrate the

laser to the individual’s pain sensitivity. An ascending method of limits procedure

was used, starting from 0.6 V with 0.06 increments each time. The participant used

an eleven point NRS (0 - 10) to rate the intensity of the pain perceived for each

stimulus and the following description was provided; 0=no sensation, 4=pain

threshold, 7=moderately painful, 10=unbearably painful. The rating scale was

introduced to the participant via these standardised descriptives to ensure that no

explanation altered their interpretation of the scale. The procedure was repeated

three times to allow participants to get used to the laser and was used to calculate

the average voltage to induce level 4 (low) and level 7 (moderate) pain. These two

levels provided ‘low’ and ‘high’ stimuli intensity for the main laser experiment

protocol.

3.3.d.iii. Main experiment

The participants received 120 laser stimuli at the two intensities (low and high)

separated into four conditions; Low (level 4), High (level 7), Unknown Low (level 4)

and Unknown High (level 7). To investigate the anticipation of a painful stimulus, a

3 second auditory countdown preceded the laser stimuli (see Figure 3.1). The first

auditory cue was presented concurrently with an anticipatory cue to indicate the

forthcoming laser stimuli and to maintain attention. The participant was either

shown ‘Low’, ‘High’ or ‘Unknown’. The presentation of the word ‘Unknown’

indicated that the laser stimulus has an equal chance of being low or high. This was

to investigate the importance of certainty in the anticipation of the laser stimuli.

The image was also used as a visual fixation cue to discourage eye movements.

After the laser stimuli, the 0-10 numerical rating scale was shown on the screen and

Page 71: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

71

the participant rated the intensity of the pain. The order of the stimuli was

randomised and separated into three blocks with short breaks in-between.

Figure 3.1: A schematic diagram of a single trial of the experimental paradigm. A computer monitor showed

the participant a visual cue of; Low, High or Unknown from -3s to +2s. A three second countdown of beeps at

-3, -2 and -1 allowed accurate anticipation of the laser stimuli at time 0s (red bar). The presentation of the

visual cue at -3s was concurrent with the first auditory cue. The visual cue was consistent throughout the

anticipation and laser stimulus. At +2s, an eleven-point number rating scale (NRS) was presented for the

participant to rate the laser stimulus. For each condition (Low, High, Unknown Low and Unknown High) there

were thirty trials. The trials were presented in a randomised order and divided into three blocks of forty

trials.

3.3.d.iv. EEG recording:

A BrainVision MR EEG cap was used to record from 63 scalp electrodes using a

BrainVision-cap system [Standard BrainCap-MR with Multitrodes]. The arrangement

of the electrodes was modelled on the extended 10-20 system. Recording

parameters were set at: Filter (DC to 70 Hz), Sampling rate (1000 Hz), Gain (500). To

reduce electrical interference, a 50Hz notch filter was applied. Prior to starting the

laser protocol, resting states were recorded with eyes open and closed for two

minutes in all participants. This ensured that the experience prior to the experiment

was identical. The three experimental blocks were recorded separately to allow for

better artefact rejection of the EEG data.

Page 72: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

72

3.3.e. Analysis Methods

3.3.e.i. Statistical analysis of behavioural data

Statistical analyses of the behavioural measures were carried out using IBM SPSS

Statistics 22 software. The questionnaires were all investigated for significant group

differences. Prior to using statistical tests, the data was assessed for normality using

a combination of Q-Q plots, histograms, and the values of skew and kurtosis.

Normally distributed data was analysed using independent t-tests and ANOVA tests,

whilst data reported to be not normally distributed, the appropriate non-

parametric test was utilised. Specific statistical tests for each analysis step are

reported in the results section.

3.3.e.ii. EEG analysis method

EEG pre-processing was carried out using EEGLAB toolbox (Delorme and Makeig,

2004) in MATLAB version R2015a (The Mathworks Inc) whilst statistical analysis was

carried out using SPM12 toolbox (Wellcome Department of Imaging Neuroscience,

Institute of Neurology, UCL, London, United Kingdom) running in MATLAB. The EEG

data was pre-processed for scalp and source localisation analysis. The main

motivation of the analysis was to establish the anatomical origin of the brain

activity using (Low Resolution Electromagnetic Tomography) LORETA source

localisation.

3.3.e.iii. EEGLAB Pre-processing

Pre-processing consisted of; removal and interpolation of bad channels, down-

sample to 500, low-pass filter of 20Hz and re-reference to the common average.

The four conditions were separated and -3500ms to 2000ms epochs extracted and

Linear detrend applied. Independent Component Analysis (ICA) was carried out on

all datasets using the SemiAutomatic Selection of Independent Components for

Artifact correction (SASICA) toolbox to select components to remove via pre-

determined thresholds. The thresholds were set to; Autocorrelation (threshold =

0.35 r, lag = 20 ms), Focal (threshold = 3.5 z), Focal trial (threshold 5.5 z), Signal to

noise (period of interest (POI) = [0 Inf], baseline (BL) [-Inf 0], threshold ratio = 0.5),

Page 73: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

73

and Adjust Selection enabled. The thresholds were sufficient to remove artefacts

from the majority of the datasets; however, a number of datasets required further

manual removal of eye-blink components where not picked up by SASICA.

3.3.e.iv. SPM EEG Analysis

The pre-processed datasets were converted to Statistical Parametric Mapping

(SPM) compatible files. Statistical analysis was carried out to investigate the

anticipation evoked potentials and the post-stimulus LEPs using scalp-level and

source localisation analysis techniques available in the SPM toolbox.

SPM scripts for batch processing were used to analyse the EEG data at the scalp

level and source localisation. Two baselining methods were applied to the data

analysis for the anticipation phase and were applied for scalp-level and source

localisation analysis (see 2.1.b.ii). Primary analysis applied distinct baselines (BLs) of

500 ms, occurring prior to each of the three auditory cues respectively, to analyse

each of the three anticipation phases. The BLs and time window of interest (TWOI)

were as follows; Early [BL: -3500 ms -3000 ms: TWOI: -2500 ms -2000 ms], Mid [BL:

-2500 ms -2000 ms: TWOI: -1500 -1000 ms] and Late [BL: -1500 -1000 ms: TWOI: -

500 0 ms] anticipation phases. The secondary analysis method applied a single

baseline of 500 ms prior to the first auditory cue [BL: -3500 ms -3000 ms] that was

common to every TWOI anticipation phase (early, mid and late). This second

analysis was conducted to enable comparison to previous studies (Brown et al.,

2008a; Brown, El-Deredy and Jones, 2014). All statistical analysis for the

anticipation phase was adjusted for multiple comparisons.

The SPM whole-head analysis for the post-stimulus phase TWOI [200 ms 600 ms],

centred on the LEP, was baseline corrected to -500 ms prior to the laser stimulus

[BL: -500 ms 0 ms].

Two further analysis steps were carried out to investigate the LEP further. Firstly,

the N2 and P2 components of the LEP were calculated for the Cz electrode at scalp

level (chosen for showing highest amplitude during LEP). The time window was

restricted to 0 600 ms and the amplitude and latency of the most negative (N2-

Page 74: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

74

peak) and positive (P2-peak) peaks were extracted for statistical analysis. All laser

conditions were averaged. The values extracted for the N2-peak was restricted to

be prior to the P2-peak due to the N2 peak occurring prior to the P2 peak.

Secondly, a focussed analysis of the P2 peak using source localisation and individual

time windows was carried out. A time window of 80ms centred on each

participant’s P2 peak latency was used to calculate source estimates, and was

assessed for group differences.

We also calculated the SD (Standard Deviation) of the EEG potential over trials for

every time sample across the whole-epoch [-3500 1500 ms] and compared the

results between the two groups to evaluate for possible differences in variability of

the data over trials. This was to test whether any group differences found in ERP

amplitudes and sources from the main analyses might have resulted from

differences in data variability; such variability can arise from noise in the EEG signal

(including motion and other artefact) rather than from neural signals. Such noise

was expected to be greater in the PD group and therefore required assessing in

order to interpret the results.

3.3.e.v. Source Localisation analysis parameters

SPM12 EEG and MATLAB scripts were used to estimate the sources of the

anticipation- and laser-evoked potentials using LORETA. The forward model was

created using an 8196 vertex template cortical mesh coregistered to the electrode

positions of the standard 10-20 EEG system. A three-shell boundary element model

(BEM) EEG head model available in SPM12 was used to compute the forward-

model. The images were smoothed with a 12mm full-width-at-half-maximum

(FWHM).

3.3.e.vi. EEG Analysis Statistical analysis

For the analysis of the anticipatory TWOIs, a three way repeated measures ANOVA

was applied, with one between-subject factor of Group (HC vs PD) and two within-

subject factors of Certainty [Known (Low/High) v Unknown] and Expectation [Low v

High (Known)]. For the analysis of the post-stimuli TWOI, a three way repeated

Page 75: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

75

measures ANOVA was applied, with one between-subject factor of Group (HC v PD)

and two within-subject factors of Certainty [Known (Low/High) v Unknown] and

Intensity [Low v High (Known and Unknown)].

Source localisation results were reported as follows. To control for multiple

comparisons, a cluster-forming threshold of p<0.001 was used and resulting

clusters were considered significant at FWE (p<0.05). Significant clusters were also

restricted to >100 voxels in size and regions labelled using the Anatomical

Automatic Labelling (AAL2) toolbox in SPM. We also extracted the eigenvariate data

from source estimates to investigate possible correlations with behavioural

measures including MDS-UPDRS (PD group only), HADS, PCS, chronic pain maximum

VAS score and laser voltage for high pain (V). The p value was adjusted for multiple

comparisons, p<0.01.

Page 76: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

76

3.4. RESULTS

3.4.a. Behavioural Results

3.4.a.i. Questionnaires

Group comparisons were carried out using an independent t-test between

questionnaire scores and are reported in Table 3.1. There was a significant

difference reported in the PCS, such that the PD group reported a higher level of

catastrophising behaviours towards pain compared to the HC group. In contrast,

there was no significant difference seen in the HADS, although a trend is seen

towards higher scores in the PD group compared to the HC group. The OFF MDS-

UPDRS-III motor score recorded in the PD group ranged from 15 to 78, with a mean

of 38.3±16.56. For reference, a high MDS-UPDRS-III score indicates more severe

movement impairments.

Table 3.1: T e table displays t e reported meas res for t e assessments for t e P and C ro ps. ^ evene’s

Test for Equality of variance p<0.05 hence equal variances not assumed and significance adjusted. * =

Si nifi ant res lt p<0.05. P : Parkinson’s disease, C: ealt y Control, PCS: Pain Catastrop isin S ale, A S:

Hospital Anxiety and Depression Scale, SD: Standard deviation.

3.4.a.ii. Laser behavioural results

The psychophysics ramping procedure was used to calculate the participants’

individual energy level required to induce a High pain score. A Welch t-test was run

to determine if there were differences between the HC and PD groups in the energy

required to induce a High pain score due to the assumption of homogeneity of

variances being violated, as assessed by Levene's test for equality of variances (p =

Assessment Group N Mean SD Significance

(2-tailed T-test)

PCS PD 23 11.59 10.85 0.019^*

HC 24 5.29 5.67

HADS PD 23 13.83 9.89 0.070

HC 24 9.21 6.76

Page 77: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

77

0.010). There was a statistically significant difference in energy required to induce a

High pain between HC and PD (Figure 3.2), with PD requiring a lower energy level

(1.99±0.44 V) compared to HC (2.20±0.25 V), (95% CI, 0.003 to 0.432), t(34.75) =

2.060, p = 0.047.

Figure 3.2: The energy of the laser required to induce high (level 7) pain. The PD group required a

lower energy level compared to the HC group to induce the equivalent pain. The blue and red

boxes show the median (bold horizontal line) (HC: 2.32V, PD: 2.24V), lower and upper quartiles.

The vertical lines show the minimum (HC: 1.68V, PD: 1.30V) and maximum (HC: 2.50V, PD: 2.48V)

values. Laser voltage was delivered from 0.8V to a maximum of 2.5V in increments of 0.06V. * =

p<0.05. C: ealt y Control, P : Parkinson’s disease.

The mean and standard error (SE) of the pain rating scores for all conditions were

calculated; Low (PD: 2.54±0.23, HC: 2.0±0.18), High (PD: 4.94±0.30, HC: 4.7±0.26),

Unknown Low (PD: 3.06±0.25, HC: 2.4±0.22) and Unknown High (PD 4.53±0.33, HC:

4.1±0.26). The pain rating scores for the laser stimuli during the main experiment

were investigated using a three-way mixed ANOVA with a between-subject factor

of group, and within-subject factors of certainty (Known v Unknown) and Intensity

Page 78: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

78

(Low v High). The data met homogeneity threshold (p>0.05) calculated by the

Levene’s Test of Equality of Variances. There was no effect of group F(1, 43)=2.347,

p = 0.133, η2 = 0.52. There was an effect of intensity F(1, 43)=290.992, p=0.000, η2 =

0.871, yet no effect of certainty F(1, 43)=0.006, p= 0.937, η2 = 0.000. However,

there was a significant two-way interaction between certainty and intensity F(1,

43)=59.188, p=0.000, η2 = 0.579. A follow-up paired t-test comparing the difference

between known and unknown for Low and High pain rating reported a significant

result; T(1, 43)=-7.820, p<0.000, d=42, such that the pain ratings of unknown low

were higher (+0.52) compared to known low, whilst ratings of unknown high were

lower (-0.46) compared to known high. This result acts as a manipulation check that

demonstrates that the experiment was successful in inducing expectations (via

anticipation cues) that modulated pain in the expected direction as seen in previous

research (Brown et al., 2008b). There was no difference in the effect of intensity or

certainty on pain ratings between the two groups (HC v PD), nor was there any

interaction between intensity, certainty and group.

3.4.b. EEG Results

3.4.b.i. LEP component analysis

The N2 and P2 components of the LEP at the Cz electrode were assessed for

differences between the two groups using independent t-tests. Outliers were

assessed via boxplot function in SPSS and one data point was excluded from the

statistical analysis due to being more than 1.5 box-lengths away from the box edge.

The statistical analysis concluded that there were no significant differences

between the HC and PD groups in the amplitude or latency of the N2 and P2 peaks

(see Figure 3.3).

Page 79: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

79

Figure 3.3: Graphs showing the information of the N2 and P2 components of the laser-evoked

potential (LEP). The N2 peak was deemed to be the most negative amplitude prior to the P2 peak

latency and within the post-stimulus time-window. The P2 peak was the most positive amplitude

within the time window of [0 600ms]. A) and B) show the amplitude of the N2 and P2 peaks

respectively, whilst C) and D) show the latency of the N2 and P2 peaks respectively. All data is

presented as mean and error bars of standard deviation. Statistical analysis to assess group

differences between HC and PD, reported no significant differences.

3.4.b.ii. Scalp-level Whole head analysis

SPM whole-head statistical analysis of the early [-2500 -2000 ms], mid [-1500 -1000

ms] and late [-500 0 ms] anticipatory phases, reported no significant difference of

group, certainty or expectation. This was true for both baseline methods (see

Methods section). Similarly, the post-stimulus TWOIs [200 600 ms] reported no

significant group or certainty differences. The intensity factor in the post-stimulus

A)

B)

C)

D)

Page 80: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

80

TWOI reported increased activity in high pain condition compared to low pain (SPM

results reported in Supplementary materials Table 3.3).

The anticipatory response over time was calculated for each group and is displayed

as topography plots in Figure 3.4. The difference between HC and PD group was

also determined by subtracting the HC amplitudes from the PD data. Despite the

SPM scalp-level analysis not reporting a significant difference, an anticipation

response is seen in the central scalp region which is more prominent in PD than HC.

Because scalp responses are a summation of deeper brain events from different

sources, the scalp-level analysis was hence followed up by source localisation

analysis.

Figure 3.4: Topography plots showing the average response for the mid- and late-anticipation time

windows for HC and PD. Mid-anticipation is baseline corrected to [-2500 -2000 ms] and late-

anticipation of [-1500 -1000]. The difference between the PD and the HC group was calculated via

subtracting the HC data from the PD data. The same scale has been used for all topography plots.

In the group difference plot, the red highlights a more positive response in the PD participants,

whilst blue indicates a more negative amplitude in PD.

Page 81: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

81

3.4.b.iii. Source EEG analysis

Source localisation analysis using the distinct baselines prior to the auditory cues

revealed that the PD group showed a higher degree of activity compared to the HC

group during mid and late anticipation phases (see Table 3.2). In addition, a trend

was also seen in the early anticipation phase. Cluster-level statistics which showed

a trend within regions previously associated with pain anticipation including the

hippocampus (Reicherts et al., 2017), the postcentral gyrus (Yang, Jackson and

Huang, 2016), and the cingulate cortex (Shackman et al., 2011) and so these were

also reported at the uncorrected p value, 0.001. Figure 3.5, shows the significant

clusters which are significant F-contrasts after FWE and adjustment of the p value

(p<0.025) due to multiple analysis techniques. The clusters are denoted with ‘◊’ in

Table 3.2 and were used for correlation analysis with behavioural parameters.

Page 82: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

82

Table 3.2: Group effect on sources of early, mid and late anticipation-evoked responses. A threshold of

clusters >100 voxels was set and results were restricted to FWE correction and the p value adjusted for

m ltiple omparisons (p<0.0 5). Res lts labelled it ‘◊’ are used for figure 3.5 and correlation analysis.

Res lts s o in trend are reported and si nified it †, and t e n orre ted val e (0.001) reported belo .

The Anatomical Automatic Labelling (AAL2) atlas was used to report the regions within the significant cluster

for the group effect. The region with the highest percentage overlap is shown, unless an equivalent share of

per enta e overlap as seen. A label of ‘Unkno n’ as not reported. T e f ll report of all of t e per enta e

overlaps can be seen in the Supplementary Materials Table 3.5.

Group Difference

Brain Region Cluster-Level Peak-Level MNI

coordinates

P(FWE) K F/T Z x y z

Earl

y

F-contrast 0.092†

(0.033 Uncorr) 413 13.79 3.45 16 -22 -16 R Hippocampus (47.5%)

T-contrast

PD>HC 0.028 757 3.71 3.63 16 -22 -16 R Hippocampus (46.8%)

Mid

F-contrast

0.021◊ 763 27.41 4.89 14 -22 52

R

R

Supplementary Motor Area (52.7%)

Mid Cingulate (33.6%)

0.003◊ 1233 19.77 4.16 -16 -38 38

L

L

Mid Cingulate (41.0%)

Supplementary Motor Area (35.2%)

0.059†

(0.022 Uncorr) 529 19.02 4.08 -26 -34 38 L Postcentral (67.5%)

T-contrast

PD>HC 0.019 922 5.24 5.03 14 -22 52 R

L

Supplementary Motor Area (52.5%)

Mid Cingulate (42.1%)

0.002 1706 4.45 4.31 -16 -38 38 L

L

Mid Cingulate (42.1%)

Supplementary Motor Area (31.4%)

0.022 877 4.36 4.24 -26 -34 38 L Postcentral (66.5%)

0.074†

(0.033 Uncorr) 457 4.13 4.03 14 -36 -14

R

R

R

Lingual (19.2%)

ParaHippocampal (16.3%)

Posterior Cingulate (12.1%)

0.068†

(0.031 Uncorr) 568 4.05 3.95 14 -60 36 R Precuneus (56.0%)

Late

F-contrast 0.000

◊ 4001 22.15 4.40 -2 -22 54

L+R

L+R

Mid Cingulate (32.6%)

Precuneus (17.0%)

T-contrast

PD>HC 0.000 6223 4.71 4.55 -2 -22 54 R

R

Mid Cingulate (27.7%)

Precuneus (17.7%)

Page 83: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

83

Figure 3.5: Source estimates for significant F-contrasts at FWE correction using MRIcroN software.

Red represents the clusters of increased activity in PD compared to HC. A) Shows the increased

activation in the PD group compared to the HC group within the mid-anticipation phase: i) Clusters

are within the Right Supplementary Motor Area and Right Mid Cingulate Cortex. ii) Clusters are

within the Left Mid Cingulate Cortex and Left Supplementary Motor Area. B) Shows the increased

activation within the PD group compared to the HC group during the late anticipatory phase: i)

Clusters are within the bilateral Mid Cingulate and Precuneus. The image is aligned by the peak-

voxel at peak-level and reported as x y z (mm).

The certainty (Known v Unknown) contrast reported a trend within the early

anticipatory TWOI, [(F-contrast: p = 0.053 F=19.87, kE = 520), (T-contrast:

Unknown>Known: p = 0.013, T= 4.32, kE = 967)], with peak voxel at x:48 y:16 z:-16

(mm). The T-contrast indicated an increased activity within the cluster in the

unknown condition in contrast to the known condition. The cluster is within the

right superior temporal pole (37.5%), right mid temporal pole (18.3%), right

superior temporal (14.6%) and the right insula (3.7%). Contrasts for expectation

Page 84: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

84

(Low v High) during anticipation, and certainty (Known v Unknown) within the post-

stimulus TWOI, showed no differences in source estimates.

During the post-stimuli TWOI there was a significant difference in the intensity,

such that a greater response was seen in the High intensity condition compared to

Low, (see supplementary materials Table 3.4). The additional source localisation

analysis of the P2 component which applied individual time-windows centred on

the P2-peak latency, reported no group differences.

3.4.b.iv. Source correlations with behavioural measures

The eigenvariate data from the source estimates (denoted with ‘◊’ in Table 3.2) for

the mid and late anticipation group effects were extracted for correlation analysis.

Spearman rank correlation of mid-anticipation clusters (A: i & ii) and the late

anticipation cluster (B: i) were investigated for correlation with MDS-UPDRS motor

score (PD only), HADS, PCS, maximum chronic VAS score and the energy of the

laser. The P value was adjusted to account for multiple comparison and no

significant correlations were highlighted.

3.4.b.v. Standard Deviation analysis

The analysis of the SD-over-trials across the whole epoch at sensor-level EEG

revealed no significant differences between the HC and PD groups, nor within-

subject contrasts, certainty or expectation.

ii) -16 –38 38

Page 85: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

85

3.5. DISCUSSION

In the present study, we used EEG source localisation techniques to investigate the

pain processing in PwPD in the practically-defined OFF state (after 12 hours

withdrawal of anti-Parkinsonian medication). There were three main findings.

Firstly, the PD group were more sensitive to the laser and required a lower energy

level to evoke painful stimuli. Secondly, EEG source localisation showed that the PD

group had a heightened anticipatory signal during the anticipation phase prior to

acute heat pain. And finally, the heightened anticipatory response was independent

of PD motor severity (MDS-UPDRS), mood (HADS), pain coping mechanisms (PCS)

and severity of chronic pain (max chronic VAS score). This present study provides

evidence to support the hypothesis that there is an augmented top-down

processing during the anticipation of pain in PwPD which may help us to

understand the high prevalence of pain in PwPD.

3.5.a. Altered Top-Down control

The amplified anticipatory signal seen in the PD group is evidence of altered top-

down modulation within the pain network prior to the pain signal reaching the

brain. This opens the possibility that the heightened pain sensitivity in PD is not

solely due to impaired peripheral, bottom-up, factors, but may also be due to the

modulation within the pain network of the brain.

The significance of altered top-down processing is well established in the pain field:

different attentional states can directly and substantially alter pain intensity and

unpleasantness (Miron, Duncan and Bushnell, 1989b); anxious anticipation of

aversive stimuli activates regions of the pain network, resulting in altered

perception of the stimuli (Reicherts et al., 2017). Furthermore, the degree of

anticipation of a noxious stimulus has been shown to directly correlate with the

perceived stimuli intensity (Pfingsten et al., 2001; Fairhurst et al., 2007), and

abnormal anticipation of a painful stimulus has been shown in a number of chronic

pain conditions such as Fibromyalgia (Burgmer et al., 2011; González-Roldán et al.,

2016; Brown, El-Deredy and Jones, 2014).

Page 86: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

86

Here we have shown increased activation in the PD group within the pain network,

including, the Mid-cingulate cortex (MCC), Supplementary Motor Area (SMA) and

Precuneus. Importantly, the group effects seen during anticipation were

independent of the MDS-UPDRS, PCS and HADS emphasising that it is likely that the

underlying PD pathological process is causing the greater anticipatory activity

rather than the consequence of motor impairment, coping mechanisms and mood

state. The fact that the abnormal processing is independent of the MDS-UPDRS

supports the hypothesis reported in Silverdale et al., (2018) that the high

prevalence of chronic pain in PD is independent of the severity of the motor

impairments and could be due to top-down modulation of the pain signal.

Our results are consistent with a single previous fMRI study, using a very different

protocol which demonstrated that PwPD showed a significantly reduced activation

within the inhibitory descending pain pathway during the anticipation of pain and

an increased activation within the MCC during pain perception (Forkmann et al.,

2017). In combination with the results presented in this current study, we reason

that PwPD display maladaptive top-down modulation during pain processing which

may help to explain the high prevalence of pain within the PD community.

3.5.b. Regions activated during the anticipatory response

The location of the amplified anticipatory signal within the PD group during mid and

late anticipation was mainly, but not limited to, the SMA and the MCC. The SMA

and MCC have a role in the selection and planning of movements (Russo et al.,

2002; Morecraft and Van Hoesen, 1998; Rushworth et al., 2004), and are indicated

to be involved in the prediction of aversive events and the movement response

required (Morrison, Peelen and Downing, 2007). The SMA and the MCC have shown

overlapping functional connectivity during pain-processing and motor control

(Misra and Coombes, 2015), and enhanced activation within the SMA has been

associated with patients with phantom limb pain (Dettmers et al., 2001).

The MCC is a region involved in cognitive processing and is associated with the fear

response, pain processing and specific motor outcomes to painful stimuli (Vogt,

Page 87: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

87

Berger and Derbyshire, 2003; Shackman et al., 2011; Böcker et al., 2001). The MCC

has previously been shown by Böcker et al., (2001) to be activated during

anticipation of threats and suggested to be the source of generating the slow-wave

anticipatory response (see figure 3.6.

Figure 3.6: Diagram from Böcker et al., (2001) which reports the grand average of the stimulus preceding

negativity associated with anticipating threatening stimuli. The data is representative of eleven participants

and show conclusions drawn from a 1-dipole source model. Böcker et al., (2001) concluded that the dipoles

clustered within the posterior of the medial frontal cortex, and show high similarity with loci of the LORETA

source localisation results shown in this study. Thereby validates the role of the MCC in the anticipation of

noxious stimuli.

Abnormal connectivity within the MCC is associated with patients with migraines

(Hubbard et al., 2014) and a heightened response within the MCC has been

reported in PwPD during pain perception (Forkmann et al., 2017). In addition, in an

extensive rodent study, the MCC showed to be central to the development of pain

hypersensitivity in the absence of peripheral noxious stimuli (Tan et al., 2017). The

study also demonstrated that silencing the MCC via optogenetic techniques,

partially reversed inflammatory hypersensitivity, thus highlighting the role of MCC

in development of chronic pain conditions (Tan et al., 2017).

Page 88: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

88

Additionally, the MCC is also associated with reward processing (Hayden et al.,

2008; Pearson et al., 2009; Shackman et al., 2011), a process highly dependent on

dopaminergic signalling and hence may be important due to the depletion of

dopamine seen in PD. The reward pathway is linked to pain perception as it

encodes the inverse of a reward signal and is closely correlated to the opioid

system. Abnormal reward processing is a possible predictor of the development of

increased pain sensitivity (Nees and Becker, 2017; Nees et al., 2017), and provides a

potential explanation of why abnormalities in the MCC during pain perception are

present in PwPD.

Thus, the MCC and SMA are important for the processing of the pain signal and

provide a putative anatomical substrate for enhanced top-down modulation of

incoming pain signals.

3.5.c. Laser Evoked Potential

The LEP was not significantly different between the two groups. The subjective

experience of pain was standardised in both groups, such that participants

experienced what they subjectively considered to be a low and high pain. We

therefore did not expect a group difference in the LEP.

Although research has previously shown that an altered degree of anticipation can

modulate the LEP amplitude and pain unpleasantness, there is research by Clark et

al., (2008) which reported that the characteristics of the LEP, specifically the P2

peak, was not affected by the duration of the anticipation period, nor correlated to

the behavioural pain ratings. They concluded that the anticipatory response is more

closely aligned with attentional processing rather than intensity coding of the

stimulus. Hence the research is a potential explanation of how a difference during

the anticipatory period was reported, yet not the LEP.

3.5.d. Considerations of interpretation

The present study has used source localisation to successfully quantify the

anatomical origin of the activity recorded at scalp electrodes. These differences

were not directly correlated to significant changes at scalp-level EEG and thus needs

Page 89: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

89

to be considered when interpreting these results. It is known that there is no simple

relationship between scalp and source activity, as a single electrode summates the

potentials generated within neural sources and is dependent on the combination of

the orientation, strengths and location of these potentials. Hence, as scalp

topographies are the product of the addition of multiple brain sources, it is difficult

to interpret them directly in relation to source localisation estimates. The

comparison of SD-over-trials between the groups did not report any differences,

and indicates that group differences in data variability (e.g. due to noise) is unlikely

to be a factor driving group effects at the source level. Hence we are confident that

despite not seeing scalp-level group differences, the significant differences reported

via source localisation are valid.

3.5.e. Clinical impact

The current perception of pain in PwPD is often considered to be a consequence of

peripheral symptoms such as rigidity and stooped posture. However, our findings

suggest the possibility that the treatment for chronic pain in PD could beneficially

incorporate alternative treatments such as mindfulness and cognitive behavioural

therapy (CBT). These treatments have been shown to reduce pain anticipation

(Brown and Jones, 2010), improve cognitive control of pain over time and reduce

the severity of chronic pain (Kabat-Zinn, Lipworth and Burney, 1985; Kabat-Zinn,

1982). Nevertheless, further investigations are essential to establish whether

cognitive therapies are a suitable treatment for pain in Parkinson’s disease.

3.6. CONCLUSION

In conclusion, Parkinson’s patients demonstrated enhanced anticipatory activity in

the pain network before an acute pain stimulus. Thus we have provided evidence

for enhanced top-down processing of pain in Parkinson’s disease, increasing the

evidence for abnormal central processing in this and other chronic pain conditions.

Our results contribute to the building knowledge of the relationship between

chronic pain and Parkinson’s disease; and inform a possible scientific rational for

novel treatment strategies in Parkinson’s pain, including mindfulness, cognitive

therapies and other treatments targeted at reducing top down processing of pain.

Page 90: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

90

3.7. SUPPLEMENTARY MATERIALS

Table 3.3: Scalp-level SPM results for the F-contrast of Intensity (Low v High) for post-stimulus

TWOI [200 600]. Significant results were restricted to clusters of >100 voxels and significant

following FWE-correction, During the TWOI centred on the laser evoked potential (LEP) the data

shows an increased activation in the High condition compared to the low condition. Results are

presented at cluster- and peak-level, plus the coordinates of the peak-voxel within the cluster.

Cluster-Level Peak-Level Coordinates

P(FWE-corr) kE p(unc) p(FWE-corr) F Z p(unc) x,y,z {mm}

0.000 30153 0.000 0.000 86.31 Inf 0.000 8.5 -40.88 458

0.000 82.53 Inf 0.000 12.75 -40.88 478

0.000 81.25 Inf 0.000 -8.5 -51.63 538

0.000 2869 0.000 0.000 49.48 6.48 0.000 -42.5 -8.63 256

0.004 28.54 5.00 0.000 -51 -30.13 216

0.004 28.32 4.98 0.000 -55.25 -24.75 210

0.000 3096 0.000 0.000 45.45 6.23 0.000 -68 7.50 488

0.000 43.41 6.10 0.000 -68 7.50 472

0.000 42.59 6.05 0.000 -68 7.50 462

0.000 1833 0.000 0.000 43.34 6.10 0.000 4.25 -73.13 264

0.000 879 0.006 0.000 41.76 5.99 0.000 55.25 -67.75 428

0.000 37.83 5.72 0.000 63.75 2.13 434

0.000 35.11 5.52 0.000 59.5 -24.75 434

0.005 272 0.094 0.000 35.84 5.58 0.000 63.75 2.13 536

0.001 32.82 5.35 0.000 55.25 18.25 536

0.001 729 0.010 0.000 34.68 5.49 0.000 -51 34.38 898

0.000 1090 0.003 0.001 31.98 5.28 0.000 -59.5 18.25 740

0.001 31.49 5.24 0.000 -59.5 18.25 698

0.000 1174 0.002 0.001 31.58 5.25 0.000 -12.75 -73.13 954

0.003 29.65 5.09 0.000 -8.5 -73.13 976

0.002 493 0.030 0.001 31.57 5.25 0.000 -55.25 23.63 638

0.011 143 0.214 0.005 28.09 4.96 0.000 -55.25 29.00 796

0.007 211 0.136 0.006 27.69 4.93 0.000 55.25 18.25 674

0.020 24.48 4.64 0.000 63.75 2.13 704

0.005 275 0.092 0.007 27.30 4.89 0.000 59.5 7.50 772

0.008 186 0.159 0.010 26.29 4.80 0.000 55.25 18.25 832

0.011 139 0.221 0.010 26.19 4.79 0.000 17 -8.63 192

Page 91: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

91

Table 3.4: The source localisation results for the significant within-subject contrast of Intensity

(Low v High) during the post-stimulus time window [200 600 ms]. A threshold of clusters >100

voxels was set and results were restricted to FWE correction. The Anatomical Automatic Labelling

(AAL2) atlas was used to report the regions within the significant cluster for the group effect. All

re ions are reported in l din ‘Unkno n’. T e T-contrast shows that High intensity evoked a

higher activation in comparison to Low intensity.

Cluster-Level Peak-Level Brain Region

P(FWE) K F/T Z mm mm mm

Po

st-S

tim

ulu

s [2

00

60

0 m

s]

F-contrast

0.000 70986 55.34 6.80 20 -60 -12

Unknown (17.0%), Occipital_Mid_L (3.3%), Precuneus_R (3.0%), Fusiform_L (2.9%), Parietal_Inf_L (2.8%), Lingual_R (2.8%),

Lingual_L (2.6%), Fusiform_R (2.5%), Calcerine_L (2.2%), Postcentral_L(2.2%),

Parietal_Sup_L (2.2%), Calcerine_R (1.9%), Parietal_Sup_R (1.9%), Postcentral_R (1.8%),

Cingulate_Mid_L (1.7%), Temporal_Inf_L(1.7%), Occipital_Sup_R

(1.6%), Occipital_Sup_L (1.6%), Cuneus_L(1.5%), Occipital_Mid_R (1.5%),

SupraMarginal_L (1.5%), Precentral_L (1.5%), Angular_L (1.4%), Temporal_Sup_L

(1.4%), Insula_L (1.4%), Rolandic_Oper_R(1.3%), Cingulate_Mid_R (1.3%), Frontal_Inf_Tri_L (1.2%), Cuneus_R

(1.2%), Temporal_Inf_R(1.1%), Hippocampus_L (1.1%), Parietal_Inf_R (1.1%), Paracentral_Lobule_L (1.0%),

Rolandic_Oper_L (1.0%)

0.002 1290 21.29 4.32 26 24 28

Frontal_Mid_2_R (59.2%), Unknown (22.1%), Frontal_Sup_2_R (13.3%),

Precentral_R(5.3%)

T-contrast High>Low

0.000 43912 5.63 5.38 -32 -38 12

Unknown (20.0%), Precuneus_L (4.7%), Parietal_Inf_L (4.1%), Lingual_R (4.0%), Postcentral_L(3.8%), Fusiform_L (3.0%),

Postcentral_R (2.8%), Parietal_Sup_L (2.7%), Preentral_R (2.7%), Frontal_Inf_Tri_L (2.6%),

Calcerine_R (2.6%), Insula_L (2.4%), Lingual_L (2.3%), Precentral_L (2.2%), Cingulate_Mid_L (2.2%), Precuneus_R

(1.9%), Fusiform_R (1.9%), Angular_L(1.8%), Calcerine_L (1.7%), Paracentral_Lobule_L

(1.7%), Frontal_Inf_Oper_L (1.7%), Occipital_Mid_L(1.7%), Cingulate_Mid_R

Page 92: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

3. A neurophysiological investigation of pain anticipation in Parkinson’s disease

92

Table 3.5: A detailed version of Table 3.2 to include all regions highlighted by the AAL2 toolbox.

The table displays the group effect on sources of early, mid and late anticipation-evoked

responses. A threshold of clusters >100 voxels was set and results were restricted to FWE

orre tion and t e p val e adj sted for m ltiple omparisons (p<0.0 5). Res lts labelled it ‘◊’

are used for figure 3.5 and correlation analysis. Results showing a trend are reported and signified

it †, and the uncorrected value (0.001) reported below. The Anatomical Automatic Labelling

(AAL2) atlas was used to report the regions within the significant cluster for the group effect. All

re ions are reported in l din ‘Unkno n’. = eft, R = Ri t, S p = Superior, Inf = Inferior.

Group Difference

Cluster-Level Peak-Level Brain Region

P(FWE) K F/T Z mm mm mm

Ea

rly

F-contrast 0.092

(0.033 Uncorr) 413 13.79 3.45 16 -22 -16

Unknown (48.4%), Hippocampus_R (47.5%),

Parahippocampal_R (4.1%)

T-contrast

PD>HC 0.028 757 3.71 3.63 16 -22 -16

Hippocampus_R (46.8%), Unknown (46.1%),

Amygdala (4.0%), Parahippocampal_R (2.9%),

Linqual_R (0.3%)

Mid

F-contrast 0.021◊ 763 27.41 4.89 14 -22 52

Supp_Motor_Area_R (52.7%) Cingulate_Mid_R

(33.6%), Paracentral_Lobule_R (10.1%),

Unknown (2.6%), Precuneus_R (1,0%)

0.003◊ 1233 19.77 4.16 -16 -38 38

Cingulate_Mid_L (41.0%) Supp_Motor_Area_L

(35.2%), Unknown (23.1%), Precuneus_L (0.2%),

Paracentral_Lobule_L (0.2%), Frontal_Sup_2_L

(0.1%), Cingulate_Ant_L (0.1%)

0.059†

(0.022 Uncorr) 529 19.02 4.08 -26 -34 38

Postcentral_L (67.5%), Unknown (19.3%),

Parietal_Inf_L (12.3%), Supramarginal_L (0.9%)

T-contrast

PD>HC 0.019 922 5.24 5.03 14 -22 52

Supp_Motor_Area_R (52.5%) Cingulate_Mid_L

(42.1%), Paracentral_Lobule_R (11.0%),

Unknown (3.7%), Precuneus (2.8%)

0.002 1706 4.45 4.31 -16 -38 38

Cingulate_Mid_L (42.1%) Supp_Motor_Area_L

(31.4%), Unknown (24.6%), Precuneus_L (0.7%),

Cingulate_Ant_L (0.4%), Frontal_Sup_Medial_L

(0.4%), Parecentral Lobule (0.2%),

Frontal_Sup_2_L (0.2%)

0.022 877 4.36 4.24 -26 -34 38 Postcentral_L (66.5%), Unknown (18.9%),

Parietal_Inf_L (13.0%), SupraMarginal_L (1.6%)

0.074†

(0.022 Uncorr) 457 4.13 4.03 14 -36 -14

Unknown (30.3%), Lingual_R (19.2%)

ParaHippocampal_R (16.3%) Cingulate_Post_R

(12.1%) Cerebellum_R (9.7%),

Hippocampus_R(4.9%), Precuneus_R (3.5%),

Thalamus_R (2.2%), Cerebelum_3_R (1.3%),

Vermis_4_5 (0.5%)

0.068†

(0.022 Uncorr) 568 4.05 3.95 14 -60 36

Precuneus_R (56.0%) Cingulate_Mid_R (10.4%)

Cingulate_Post_R (9.9%) Cuneus_R (7.4%),

Unknown (6.0%), Calcerine_R (5.6%),

Precuneus_L (3.0%), Cingulate_Post_L (0.7%),

Cuneus_L (0.7%), Calcerine_L (0.2%),

Occipital)Sup_R (0.2%)

La

te

F-contrast

0.000◊ 4001 22.15 4.40 -2 -22 54

Unknown (17.2%), Cingulate_Mid_R (16.5%)

Cingulate_Mid_L (16.1%) Precuneus (11.4%)

Supp_Motor_Area_L (10.3%) Cingulate_Post_L

(5.8%) Precuneus_L (5.6%) Paracentral_Lobule_L

(4.5%) Supp_Motor_Area_R (4.3%)

Paracentral_Lobule_R (4.1%) Cingulate_Post_R

(3.8%), Postcentral_R (0.2%), Parietal_Sup_R

(0.0%).

T-contrast

PD>HC

0.000 6223 4.71 4.55 -2 -22 54

Unknown (21.0%), Cingulate_Mid_R (14.5%)

Cingulate_Mid_L (13.2%) Precuneus_R (9.6%)

Precuneus_L (8.1%) Supp_Motor_Area_L (7.5%)

Cingulate_Post_L (4.7%), Calcerine_L (4.6%),

Supp_Motor_Area_R (4.1%),

Parecentral_Lobule_L (4.0%),

Paracentral_Lobule_R (2.9%), Cingulate_Post_R

(2.8%), Postcentral_R (1.3%), SupraMarginal_R

(0.8%), Cuneus_L (0.5%), Thalamus_L (0.2%),

Occipital_Sup_L (0.1%), Lingua_L (0.1%),

Parietal_Inf_R (0.0%), Parietal_Sup_R (0.0%).

Page 93: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Chapter 4

Central pain processing in people with Parkinson’s disease whilst on

medication

Sarah Martin, Anthony Jones, Christopher Brown,

Christopher Kobylecki, Wael El-Deredy, Monty Silverdale

Page 94: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

94

4.1. ABSTRACT

The administration of dopaminergic medication for Parkinson’s disease has

previously been shown to have analgesic qualities. Here we investigated the pain

processing of people with Parkinson’s disease whilst on their medication via EEG

recording and receiving acute noxious stimuli. We have previously shown that in

the same participants, whilst off their medication, there was a significant

amplification of the anticipatory processing within the mid-cingulate cortex

compared to healthy age-matched volunteers. This study reported that following

medication administration, the people with Parkinson’s disease remained

significantly more sensitive to the noxious stimulus in comparison to a healthy

cohort. However, no difference was seen in the brain activations between the PD

and healthy groups post medication administration. Therefore, in the PD

medication normalised the augmented anticipatory processing that was reported in

the patient cohort whilst off their medication.

4.2. INTRODUCTION

There is a higher prevalence of chronic pain in Parkinson’s disease (PD) than the

general population (Nègre-Pagès et al., 2008; Ozturk et al., 2016; Skogar & Lokk,

2016; Silverdale et al., 2018). Pain is often considered to be secondary to muscle

stiffness and reduced movement characteristic to PD, however, there is growing

evidence that altered brain activity during pain processing contributes to the

development of chronic pain.

The severity of pain in Parkinson’s has been shown to be worse in the ‘off’ state,

when medication is wearing off, and improved after taking medication (Skogar and

Lokk, 2016). Hence, the stabilisation of the dopamine levels following Parkinson’s

medication may regulate the dopamine-dependent processes involved in pain

processing. L-DOPA, a commonly used drug in PD, has been associated with

analgesic qualities (Shimizu et al., 2004a; Kernbaum and Hauchecorne, 1981; Ertas

et al., 1998; Nixon, 1975; Minton, n.d.; Sandyk et al., 1987; Cotzias et al., 1970). The

Page 95: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

95

role of dopamine in the brain is widespread and contributes to the affective and

attentional processing in pain (de Wied and Verbaten, 2001; Tiemann et al., 2014).

Dopamine has also shown to have analgesic properties (Wood, 2014) and is

considered to be a potential treatment of chronic pain conditions (Lawson, 2016;

Haddad et al., 2018).

Hence, we aim to investigate the processing of acute pain in people with

Parkinson’s disease (PwPD) whilst on their medication in comparison to healthy

age-matched controls. This investigation follows the report that PwPD whilst off

their medication were more sensitive to the noxious stimuli and showed

augmented anticipatory processing prior to acute pain (see Chapter 3). On the basis

that dopamine has shown analgesic qualities, we hypothesised that the Parkinson’s

medication, which increases dopamine, would reduce pain sensitivity and normalise

the amplified central processing seen whilst off their medication.

4.3. METHODS

4.3.i. Participants

Nineteen PD participants and twenty-one HC participants repeated the experiment

outlined in Chapter 3. However, within the PD cohort, five datasets were removed

due to noisy EEG data, and within the HC cohort, two datasets were removed due

to habituation to the laser, and eight datasets removed due to noisy EEG data. The

high degree of noise seen in the EEG was likely to be due to the EEG cap being kept

on for a long time. Therefore, fourteen PD participants and eleven HC participants

were included in the final analysis.

4.3.ii. Medication

The PD group were studied in their ON state after taking their medication at least 1

hour prior to experimental assessment. All participants were prescribed their usual

levodopa medication which increases dopamine activity.

Page 96: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

96

4.3.ii. Assessments

The motor section (III) of the Movement Disorder Society Unified Parkinson’s Rating

Scale (MDS-UPDRS) was completed to assess the PD participants’ symptom severity

whilst on their medication (Goetz et al., 2008). Other assessments also carried out

were the Hospital Anxiety and Depression HADS, Pain Catastrophizing Scale (PCS),

Montreal Cognitive Assessment (MoCA) and PD participants used a visual analogue

scale (VAS) to report their minimum and maximum intensity of chronic pain over

the last 6 months.

4.3.iii. Experimental design

4.3.iii.a. Pain stimuli

A CO₂ laser [50W Synrad 48-5 J-series (J-48-5(S)W) Wavelength: 10600nm] was

used to deliver acute pain to the dorsal surface of the right forearm. The CO₂ laser

delivered a beam with a diameter of 15 mm and 150 ms duration. The voltage (V) of

the laser is linearly related to the laser energy delivered to the forearm. For each

test, the stimuli were delivered in an area measuring 4 x 5cm and were delivered in

a predetermined randomised path (Brown et al., 2008a). This was to avoid

habituation, sensitization, or skin damage.

4.3.iii.b. Psychophysics

Before starting the experimental protocol, psychophysics was used to calibrate the

laser to the individual’s pain sensitivity. The procedure was carried out 1 hour post

medication administration. An ascending method of limits procedure was used,

starting from 0.6 V with 0.06 increments each time the laser was delivered. The

participant used an eleven point NRS (0 -10) to rate the intensity of the pain

perceived for each stimuli and the following description was provided; 0=no

sensation, 4=pain threshold, 7=moderately painful, 10=unbearably painful. The

rating scale was introduced to the participant via these standardised descriptives to

ensure that no explanation altered their interpretation of the scale. The procedure

was repeated three times to allow participants to get used to the laser and was

Page 97: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

97

used to calculate the average voltage to induce level 4 (just painful) and level 7

(moderate pain) . These two levels provided low and high stimuli intensity for the

main laser experimental protocol.

4.3.iii.c. Main experiment

The participants received 120 laser stimuli at the two intensities (low and high)

separated into four conditions; Low (level 4), High (level 7), Unknown Low (level 4)

and Unknown High (level 7). The unknown condition was to investigate the effect of

certain or uncertain anticipatory cues on neural activity and pain perception. To

investigate the anticipation of a painful stimulus, a 3 second auditory countdown

preceded the laser stimuli (see Figure 1). The first auditory cue was presented

concurrently with an anticipatory cue to indicate the forthcoming laser intensity

and to maintain focused attention. The participant was either shown ‘Low’, ‘High’

or ‘Unknown’. The presentation of the word ‘Unknown’ indicated that the laser

stimulus has an equal chance of being low or high. This was to investigate the

importance of certainty in the anticipation of the laser stimuli. The image was also

used as a visual fixation cue to discourage eye movements. After the laser stimuli,

the 0-10 numerical rating scale was shown on the screen and the participant rated

the intensity of the pain. The order of the stimuli was randomised and separated

into three blocks with short breaks in-between.

4.3.iii.d. EEG recording:

A BrainVision MR EEG cap was used to record from 63 scalp electrodes using a

BrainVision-cap system [Standard BrainCap-MR with Multitrodes]. The arrangement

of the electrodes was modelled on the extended 10-20 system. Recording

parameters were set at: Filter (DC to 70 Hz), Sampling rate (1000 Hz), Gain (500). To

reduce electrical interference, a 50Hz notch filter was applied. Prior to starting the

laser protocol, resting states were recorded with eyes open and closed for two

minutes in all participants. This ensured that the experience prior to the experiment

was identical. The three experimental blocks were recorded separately to allow for

better artefact rejection of the EEG data.

Page 98: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

98

4.3.iv. Analysis Methods

4.3.iv.a. Statistical analysis of behavioural data

Statistical analyses of the behavioural measures were carried out using IBM SPSS

Statistics 22 software. The questionnaires were all investigated for significant group

differences. Prior to using statistical tests, the data was assessed for normality using

a combination of Q-Q plots, histograms, and the values of skew and kurtosis.

Normally distributed data was analysed using independent t-tests and ANOVA tests,

whilst data reported to be not normally distributed, the appropriate non-

parametric test was utilised. Specific statistical tests for each analysis step are

reported in the results section.

4.3.iv.b. EEG analysis method

EEG pre-processing was carried out using EEGLAB toolbox (Delorme and Makeig,

2004) in MATLAB version R2015a (The Mathworks Inc) whilst statistical analysis was

carried out using SPM12 toolbox (Wellcome Department of Imaging Neuroscience,

Institute of Neurology, UCL, London, United Kingdom) running in MATLAB. The EEG

data was pre-processed for scalp and source localisation analysis. The main

motivation of the analysis was to establish the anatomical origin of the brain

activity using LORETA (Low Resolution Electromagnetic Tomography) source

localisation.

4.3.iv.c. EEGLAB Pre-processing

Pre-processing consisted of; removal and interpolation of bad channels, down-

sample to 500, low-pass filter of 20Hz and re-reference to the common average.

The four conditions were separated and -3500ms to 2000ms epochs extracted and

Linear detrend applied. Independent Component Analysis (ICA) was carried out on

all datasets using the SemiAutomatic Selection of Independent Components for

Artifact correction (SASICA) toolbox to select components to remove via pre-

determined thresholds. The thresholds were set to; Autocorrelation (threshold =

0.35 r, lag = 20 ms), Focal (threshold = 3.5 z), Focal trial (threshold 5.5 z), Signal to

Page 99: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

99

noise (period of interest (POI) = [0 Inf], baseline (BL) [-Inf 0], threshold ratio = 0.5),

and Adjust Selection enabled. The thresholds were sufficient to remove artefacts

from the majority of the datasets; however, a number of datasets required further

manual removal of eye-blink components where not picked up by SASICA.

4.3.iv.d. SPM EEG Analysis:

The pre-processed datasets were converted to Statistical Parametric Mapping

(SPM) compatible files. Statistical analysis was carried out to investigate the

anticipation evoked potentials and the post-stimulus LEPs using scalp-level and

source localisation analysis techniques available in the SPM toolbox.

SPM scripts for batch processing were used to analyse the EEG data at the scalp

level and source localisation. Two baselining methods were applied to the data

analysis for the anticipation phase and were applied for scalp-level and source

localisation analysis (see 2.1.b.ii). Primary analysis applied distinct baselines (BLs) of

500 ms, occurring prior to each of the three auditory cues respectively, to analyse

each of the three anticipation phases. The BLs and time window of interest (TWOI)

were as follows; Early [BL: -3500 ms -3000 ms: TWOI: -2500 ms -2000 ms], Mid [BL:

-2500 ms -2000 ms: TWOI: -1500 -1000 ms] and Late [BL: -1500 -1000 ms: TWOI: -

500 0 ms] anticipation phases. The secondary analysis method applied a single

baseline of 500 ms prior to the first auditory cue [BL: -3500 ms -3000 ms] that was

common to every TWOI anticipation phase (early, mid and late). This second

analysis was conducted to enable comparison to previous studies (Brown et al.,

2008a; Brown, El-Deredy and Jones, 2014). All statistical analysis for the

anticipation phase was adjusted for multiple comparisons.

All analysis for the post-stimulus phase TWOI [200 ms 600 ms], centred on the LEP,

was baseline corrected to -500 ms prior to the laser stimulus [BL: -500 ms 0 ms].

4.3.iv.e. Source Localisation analysis parameters

SPM12 EEG and MATLAB scripts were used to estimate the sources of the

anticipation- and laser-evoked potentials using LORETA. The forward model was

Page 100: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

100

created using an 8196 vertex template cortical mesh co-registered to the electrode

positions of the standard 10-20 EEG system. A three-shell boundary element model

(BEM) EEG head model available in SPM12 was used to compute the forward-

model. The images were smoothed with a 12mm full-width-at-half-maximum

(FWHM).

4.3.iv.f. EEG Analysis Statistical analysis

For the analysis of the anticipatory TWOIs, a three way repeated measures mixed

ANOVA was applied, with one between-subject factor of Group (HC vs PD) and two

within-subject factors of Certainty [Known (Low/High) v Unknown] and Expectation

[Low v High (Known)]. For the analysis of the post-stimuli TWOI, a three way

repeated measures ANOVA was applied, with one between-subject factor of Group

(HC v PD) and two within-subject factors of Certainty [Known (Low/High) v

Unknown] and Intensity [Low v High (Known and Unknown)].

Source localisation results were reported as follows. To control for multiple

comparisons, a cluster-forming threshold of p<0.001 was used and resulting

clusters were considered significant at FWE (p<0.05). Significant clusters were also

restricted to >100 voxels in size and regions labelled using the Anatomical

Automatic Labelling (AAL2) toolbox in SPM.

Page 101: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

101

4.4. RESULTS

4.4.i. Behavioural Results

4.4.i.a. Questionnaires

The PCS was significantly higher in the PD group (11.18±10.66, mean±SD) compared

to the HC group (4.36±4.11), a statistically significant difference of 6.81(95% CI,

13.35 to 0.28), t(23)=-2.194, p=0.042. Hence, indicating a higher level of pain

catastrophizing behaviours in PD. In contrast, there were no statistically significant

differences in the scores of the HADS or MoCA between the HC and PD groups.

The MDS-UPDRS Motor score (III) was recorded 1 hour after medication

administration within the PD group to be 20.75±7.70 (mean±SD). The score had

reduced from the score recorded during the PD participants’ ‘off’ state

(36.42±15.84) demonstrating symptom improvement.

Within the PD group, the level of current pain recorded via a VAS was compared to

their baseline current pain level whilst they were off their medication. A 2-tailed

paired t-test showed that within those with chronic pain n=8, there was a

significant reduction in their current pain levels (Mean±SD: 15.85±23.15 to

8.75±19.63) T(1, 7)=2.486, p=0.041. .

4.4.i.b. Laser Sensitivity

The voltage of the laser was calibrated to the participants’ subjective perception of

the intensity. An independent-samples t-test was carried out on the two pain

intensities to determine any differences in sensitivity between the HC and PD group

(Figure 4.1). The voltage to induce low pain was lower in PD (1.48±0.36; mean±SD)

than the HC group (1.75±0.24), a statistically significant difference of 0.27 (95% CI,

0.01 to 0.52), t(23)=2.13, p=0.045. The voltage to induce high pain was assessed via

a Welch test due to equal variances not assumed assessed by Levene’s Test for

Equal Variances (p=0.033). The result showed that the voltage required to induce

high pain was also lower in PD (mean±SE: 1.95±0.44) compared to HC (2.25±0.25).

Page 102: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

102

The comparison was a statistically significant difference of 0.14 (95% CI, 0.01 to

0.59), t(21.32)=2.12, p=0.046.

Figure 4.1: The laser voltage required to induce a low and high pain intensity. The PD group were more

sensitive to the laser stimuli as they required a significantly lower voltage level of the laser to induce a high

pain. The data is separated into HC (blue) and PD (red), and for Low and High pain. The boxes show the

median (bold horizontal line), and lower and upper quartiles. The vertical lines show the minimum and

maximum values. * denotes significance p<0.05.

4.4.i.c. Pain rating

The rating of the laser stimuli (Table 4.1) was assessed using a mixed-ANOVA to

determine any effect of the certainty of the visual cues and the intensity of the

laser, and any differences between the PD and HC group. Firstly, there was no

difference in pain rating between the PD and HC groups [F(1, 23)=0.738, p=0.992],

nor an effect of certainty on pain ratings. As expected, the rating of the high

intensity laser stimuli was significantly higher than low intensity [F(1, 23)= 108.7,

p<0.000].

There was a significant effect of Certainty (Known v Unknown) on Intensity (Low v

High), [F(1, 23)=34.91, p<0.000], such that the “Unknown” visual cue resulted in a

higher rating of the Low intensity, yet a lower rating of the high intensity of the

0

1

2

3

L a s e r v o lta g e to in d u c e L o w a n d H ig h

in te n s ity in H C a n d P D

La

se

r V

olt

ag

e (

V)

L o w H ig h

*

*

H C

P D

Page 103: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

103

laser (Figure 4.2). This manipulation of pain rating has previously been shown

(Chapter 3, (Brown et al., 2008a)) and is thus a good validation of the experimental

design. There was no difference seen between the two groups in the effect of

Certainty and Intensity.

Table 4.1: The rating of the laser stimuli for each laser condition and each group.

Figure 4.2: The effect of certainty on the pain rating scores. The graph shows the averaged pain

ratings for both HC and PD groups. The effect of the uncertain visual cue ‘Unknown’ resulted in a

higher rating of the low intensity compared to the known ‘Low’ visual cue. In contrast, the rating

of the high laser intensity was lower when preceded by the ‘Unknown’ cue rather than the certain

‘ i ’ vis al e. T is manip lation of pain ratings by certainty has been reported previously and

is a good validation of experiment design. The boxes show the median (bold horizontal line), and

lower and upper quartiles. The vertical lines show the minimum and maximum values. The

patterned boxes denote the Unknown cues which denote uncertain anticipation. UnLow =

Unknown Low, UnHigh = Unknown High. *** denotes significance p<0.001.

0

2

4

6

8

T h e c o n tra s t in g e ffe c t o f c e r ta in ty o n lo w a n d h ig h in te n s ity

La

se

r V

olt

ag

e (

V)

L o w H ig hU n L o w U n H ig h

* * *

HC PD

Laser Condition Mean SD Mean SD

Pai

n R

atin

g

Low 2.34 0.55 2.17 0.99

High 4.92 1.09 4.90 1.87

Unknown Low 2.76 0.81 2.56 1.16

Unknown High 4.25 1.03 4.63 1.65

Page 104: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

104

4.4.ii. EEG Results

The EEG recording was analysed to determine whether there were any differences

between the HC and PD whilst anticipating and perceiving the laser stimuli. During

anticipation, scalp-level analysis reported no difference in Group (HC v PD),

Certainty (Known v Unknown) or Expectation (Known Low v Known High). During

the post-stimulus time window centred on the LEP, there was no difference

between Group (HC v PD), or an effect of Certainty (Known v Unknown). However,

there was a significant effect of Intensity (Low v High), such that the higher laser

intensity evoked a higher degree of activity [kE=5101, p(FWE)=0.000, t=5.99, [-4 -52

mm], 518ms]. The cluster was located on the top of the head over dorsal regions.

The source localisation analysis showed no group differences in brain activity

between the PD and HC groups during both the anticipation and perception of the

laser stimuli. An expectation effect was seen in both PD and HC groups, such that a

higher degree of activity was seen during anticipation of high stimuli compared to

low stimuli. The increased activity during expectation of high stimuli was seen

within the left superior parietal lobe [kE=656, p(FWE)=0.042, t=4.63 [-26 -54

64mm]] during early anticipation, and higher activity within the left postcentral

gyrus during mid anticipation [kE=1620, p(FWE)=0.002, t=4.24 [-36 -22 36mm]]

(baselined to -2500 -2000 ms).

4.5. DISCUSSION

The results presented in this report contribute further to the findings described in

the Chapter 3. Here we have shown that the PD patients whilst on their medication

did not show any significant differences in central pain processing compared to

healthy controls, yet were more sensitive to the laser stimuli than controls.

The results in this study indicate that the Parkinson’s disease medication could

normalise the amplified anticipatory response seen following withdrawal of their

medication (see Chapter 3). The reduction in their current pain levels demonstrate

a potential analgesic quality and supports the theory that the lack of dopamine

Page 105: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

105

results in heightened pain sensitivity. Nevertheless, the direct comparison of

behavioural and neural responses whilst PD participants were off and on mediation

was not completed due to the confounding effect of protocol repetition alongside

medication administration. Thereby caution must be taken when suggesting

normalisation of the PD response to the HC response. The result was in agreement

with our prediction that the Parkinson’s medication, which increases dopamine,

would normalise the amplified central processing seen in the off state. This finding

suggests that the amplification of the anticipatory processing seen in the mid-

cingulate cortex whilst PD patients are off their medication is due to the reduced

level of dopamine.

Interestingly, we have shown that the PD participants were significantly more

sensitive to the laser stimuli as they required a lower laser voltage to induce both

low and high pain. This further supports the same finding in Chapter 3, and

contributes to the evidence of increased pain sensitivity in Parkinson’s and that the

increased pain sensitivity in PwPD is due to the dysfunction of other

neurotransmitter systems (Silverdale et al., 2018; Tinazzi et al., 2009). In contrast,

previous research on the effect of PD medication on pain tolerance of

experimentally induced pain has shown a reduction of sensitivity. Assessments of

cold pain tolerance and the nociceptive flexion reflex was shown to be lower in PD

patients off their medication, yet equal to healthy controls following taking their

medication (Brefel-Courbon et al., 2005; Gerdelat-Mas et al., 2007).

Dopamine’s role in pain processing is not considered to be solely specific to

intensity coding, instead it is known to be fundamental to the precision of

prediction coding of changing sensory inputs (Schwartenbeck et al., 2015; Friston et

al., 2014, 2012), which includes cognitive processing (Nieoullon, 2002) and salience

assignment (Borsook et al., 2013; Shiner et al., 2015; Mitsi and Zachariou, 2016).

The role of dopamine in the development of chronic pain has been shown to be due

to dysfunctional dopaminergic pathways affecting these processes (Wood, 2004;

Jarcho et al., 2012; Bushnell, Ceko and Low, 2013; Hagelberg et al., 2004). The

anticipatory response to the pain is highly likely to be dependent on dopaminergic

Page 106: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

106

transmission due to the role of attention, cognition and salience assignment

processes occurring prior to pain perception. Therefore, the repletion of the

dopamine via PD medication has shown to be sufficient to normalise the

anticipatory response seen in the ‘off-state’ (Chapter 3)

The high prevalence of chronic pain in the PD population, and the consistent

increase in pain sensitivity that we have seen in this study, demonstrates that PD

medication is not sufficient to prevent or reverse chronic pain in PD. It could be

argued that the fluctuating dopamine levels caused by PD physiology and the on-off

states caused by the PD medication causes long-term changes within key brain

regions involved in pain processing, predisposing and maintaining chronic pain.

Nevertheless, a confident conclusion is not possible when interpreting the results

from this study due to the limitations of the numerous uncontrolled variables which

may have resulted in a lack of a significant difference between the PD and HC

groups. Firstly, the type of PD medication was variable within the PD participants,

and no placebo medication was administered to the HC group to account for a

placebo effect of medication administration. In addition, the current level of pain

within the PD cohort was reduced following medication and may have had an effect

on neural activation. Furthermore, the number of participants who successfully

completed the study on their medication was lower than the cohort within the off-

medication study (Chapter 3). One reason of not taking part of the on-medication

aspect of the study was tiredness due to being off their medication. Hence, the final

cohort for the on-medication aspect of the study may have been biased to patients

with less severe symptoms as they did not tire as much as those with severe off-

state symptoms. This will have resulted in an imperfect representation of the

disease within the PD group.

4.6. CONCLUSION

The anticipation of pain is regarded to be a useful technique to investigate top-

down modulation of pain and has been shown to be dysfunctional in chronic pain

Page 107: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

4. Central pain processing in people with Parkinson’s disease whilst on medication

107

conditions. The processes associated with top-down modulation of pain include

processes such as salience assignment, the orientation of attention, and cognitive

processing, which are all modulated be dopaminergic transmission. Therefore, it

would be expected that PD patients show altered anticipatory neural activity to

pain whilst off their medication, which is normalised following dopaminergic

medication. This study provides potential evidence to support this theory and

further highlights the role of dopamine dysfunction in the development of

persistent pain in PD. This study also concludes that the PD patients had a reduced

tolerance to the pain stimuli, even with being on their medication, and contributes

to the prior knowledge of increased sensitivity in the PD population.

Page 108: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Chapter 5

An investigation of the role of the dopamine D2 receptor in pain

perception

Sarah Martin, Anthony Jones, Christopher Brown,

Christopher Kobylecki, Monty Silverdale

Page 109: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

109

5.1. ABSTRACT

Striatal dopamine dysfunction has been shown in a number of chronic pain

conditions and is associated with altered top-down modulation of pain processing.

Unlike the widespread abundance of the dopamine D1 receptor, the dopamine D2

receptor is confined primarily to the striatum and therefore, a focus within our

research. Here we investigated the effect of modulating striatal dopamine D2

receptor activity on the anticipation and perception of acute pain stimuli whilst

brain activity was recorded by EEG. Participants completed the experiment under

three conditions; control (Sodium Chloride, 20 mg), D2 receptor agonist

(Cabergoline, 1.25 mg) and D2 receptor antagonist (Amisulpride, 400 mg) in a

repeated-measures, double-blind study. The main experiment included the

presentation of cues which allowed for certain or uncertain prediction of two pain

intensity levels. Our results demonstrate that the agonist and antagonist did not

affect pain sensitivity. However, the antagonist amplified the effect of certainty on

pain rating. The EEG recording showed that the agonist and antagonist induced a

significant reduction in brain activation within the right temporal and parietal

regions during the anticipation of the pain. In addition, during the perception of

pain, the antagonist evoked a reduction in activity within the right insula and

inferior temporal lobe in comparison to the control and agonist conditions. This

study highlights that the D2 receptors are involved in the anticipatory processing

which occurs prior to the receipt of pain stimuli, and supports the notion that

dopamine has a role in the processing of uncertain predictions.

Page 110: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

110

5.2. INTRODUCTION

5.2.a. Dopamine and pain

There is evidence to show that there is a relationship between pain processing and

dopaminergic transmission. However, the role of dopamine in coding the intensity

of acute pain is unlikely to be its main role in pain perception. The majority of

dopaminergic neurons do not respond directly to aversive stimuli, with only a small

percentage (5-15%) reportedly activated during pain perception (Taylor et al.,

2016). Instead, the role of dopamine has been theorised to be involved in

processing salient stimuli within the environment (Cooper and Knutson, 2008; Wulff

et al., 2019; Gentry, Schuweiler and Roesch, 2018; Roughley and Killcross, 2019;

Nieoullon, 2002), assignment of emotional valence (Jensen et al., 2003), and to

modulate sensorimotor and motivation-related processes (Horvitz, 2002;

Bromberg-Martin, Matsumoto and Hikosaka, 2010).

There is evidence that dopamine has endogenous analgesic qualities (Bhagyashree

et al., 2017; Wood, 2014; Esposito et al., 1986). However, although drugs which

increase dopamine have been demonstrated to have analgesic properties (Shimizu

et al., 2004b; Nixon, 1975; Cobacho, de la Calle and Paíno, 2014), they are rarely

prescribed to treat pain, especially not for acute pain. On the basis of dopamine’s

widespread modulatory role in higher cortical processing, it is likely that the

analgesic qualities of dopamine are related to the improvement in the cognitive and

affective aspects of pain processing, rather than directly reducing pain intensity.

Central processing (i.e. attention, emotional state, and motivational state) can

manipulate pain perception and are regarded as top-down processes, whereby

subjective factors can modulate pain perception. The top-down modulation

(subject-driven) occurs in parallel with the bottom-up factors of pain processing

(stimulus-driven), including stimulus novelty and physical saliency. A prominent

theory of dopamine function outside of pain processing, is that it acts within a

Bayesian framework which codes the precision in predicting desired or expected

outcomes based on prior beliefs, thereby serving a top-down function (Friston et

Page 111: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

111

al., 2014; FitzGerald, Dolan and Friston, 2015). Altered top-down processing has

been associated with chronic pain conditions such as Fibromyalgia (Miron, Duncan

and Bushnell, 1989a; Burgmer et al., 2011; Brown, El-Deredy and Jones, 2014;

González-Roldán et al., 2016; Reicherts et al., 2017). Dopamine plays a central role

in modulating a wide variety of processes such as attention flexibility (van Holstein

et al., 2011; Klanker, Feenstra and Denys, 2013; Whitaker, 2017), salience

assignment (Nieoullon, 2002; Kapur, Mizrahi and Li, 2005; Howes and Nour, 2016;

Shiner et al., 2015; Parr and Friston, 2017), motivational states (Pultorak et al.,

2018; Elliot and Covington, 2001) and emotional processing (Salimpoor et al., 2011;

Baixauli, 2017; Peciña et al., 2013), and thus demonstrates the potential

detrimental affect dopamine dysfunction would have on top-down modulation of

pain. For example, impaired dopaminergic transmission has been associated with

chronic pain conditions (Jääskeläinen et al., 2001; Wood et al., 2007b; a) such as

Fibromyalgia (Häuser, 2016; Albrecht et al., 2016) and atypical facial pain

(Hagelberg et al., 2003a), plus it is likely to contribute to pain in Parkinson’s disease

(Blanchet and Brefel-Courbon, 2018; Silverdale et al., 2018).

5.2.b. Dopamine Receptors & Pain Perception

The dopaminergic activity within the striatum has been associated with modulation

of pain perception, especially the activity of the D2 dopamine receptor (D2R)

(Barceló, Filippini and Pazo, 2012; Hagelberg et al., 2002). For instance, direct

injection of the D2R agonists apomorphine or quinpirole into the striatum has

shown to reduce pain related behaviours within rodents, whilst a D1R agonist did

not induce any change in response (Barceló, Filippini and Pazo, 2012; Cobacho, de

la Calle and Paíno, 2014). Within humans, PET imaging has shown that striatal D2R

binding potential is inversely correlated with an individuals’ pain tolerance of the

cold pressor test (CPT) (Hagelberg et al., 2002) and heat thermode (Martikainen et

al., 2005) and their ability to suppress pain (Hagelberg et al., 2002).

The research into D2R’s role in pain has primarily been focused on pain tolerance

and brain activity during the receipt of noxious stimuli. Therefore, the role of the

D2R in the top-down modulation of pain processing is yet to be investigated. While

Page 112: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

112

the role of the D2Rs during the anticipation of noxious stimuli has not been

investigated, their role in anticipating rewarding stimuli has been conducted.

Research by Ye et al., (2011), targeted the striatal presynaptic D2/D3 autoreceptors

to reduce dopamine release via the agonist Pramopexole1, and highlighted an

increase in striatal activity during the anticipation of rewarding stimuli. They also

reported a reduction in the connection between the striatum and prefrontal cortex,

and concluded that manipulation of dopamine induced a reduction in top-down

control of impulsive behaviours.

5.2.c. Aim of study

In summary, the purpose of this study was to establish whether the D2R had a role

in top-down modulation of pain perception. To investigate this, a D2R agonist

(Cabergoline) and antagonist (Amisulpride) was chosen for their selectivity for the

D2R. Using EEG source localisation analysis we aimed to investigate changes in

brain activity during the anticipation and perception of a noxious stimuli following

D2R manipulation. Due to the role of dopamine in salience assignment, predicting

outcomes and attentional flexibility, the experimental paradigm was configured to

evaluate the effect of expectation of both low and high pain stimuli, and certain

and uncertain anticipation of the forthcoming stimuli intensity.

1 Pramopexole was given at a dose which activated the presynaptic auto-receptors and resulted in a

reduction of dopamine synthesis and release from the presynaptic neuron. In turn, there was a reduction in the activation of the post-synaptic dopamine receptors.

Page 113: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

113

5.3. METHODS

5.3.a. Ethical approval

The study was approved by the University of Manchester Ethics Committee and was

the UK Health Research Authority for the use of a National Health Service (NHS) site

(Salford Royal NHS Foundation Trust Hospital).

5.3.b. Drug selection

To target the striatum, we selected a D2R agonist, Cabergoline, and a D2R

antagonist, Amisulpride, which both have a high affinity for D2Rs. Previous studies

have shown that at low doses of D2R drugs, the binding of the presynaptic D2

autoreceptor is favoured and produces the opposite of the desired results (Maruya

et al., 2003; Ford, 2014). Therefore, a sufficiently high dose was selected to

modulate the postsynaptic D2R despite the action of the autoreceptors.

5.3.c. Participant recruitment

A total of twenty-nine healthy participants (16 females) were recruited for the

study (mean age 22.4 years, SD 3.2 years). All subjects gave written informed

consent. The exclusion criteria of the study was as follows; history of significant

head injury or seizures, diagnosed or taking medication for any neurological or

psychiatric condition, history of drug or alcohol dependence, use of psychotropic

medication within the past 6 months, use of dopaminergic drug within the past

month or lifetime use exceeding 3 months, pregnant or breastfeeding or

attempting to conceive (females only), suffering from chronic pain.

All participants also completed the Barratt Impulsivity Scale (BIS-11, Patton et al.,

1995) subscale of ‘cognitive instability’. The degree of impulsivity has shown to be

related to dopaminergic transmission and receptor abundance within the striatum

(Bucker and Theeuwes, 2014; Costa et al., 2013; Buckholtz et al., 2010; Dalley and

Robbins, 2017). The participants’ were only recruited if their score was within 1.5

standard deviations of the mean reported from a large control sample (1,577

healthy adults) (Stanford et al., 2009).

Page 114: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

114

We used a repeated-measures, double-blind and triple-crossover design such that

all participants were recruited to attend three visits and complete the experimental

protocol following ingestion of the agonist, antagonist and control substance. Two

participants did not complete all three visits of the study, and one participant was

removed due to an adverse reaction to Amisulpride. Therefore, twenty-six datasets

were included for behavioural and EEG analyses (mean age 22.2 years, SD 3.7 years,

14 females).

5.3.d. Health Screening

Before the first experimental visit, the participants attended a health screening to

deem them safe to take part in the study. A medical doctor assessed the

participants’ heart rate, blood pressure, temperature and recorded an

electrocardiogram (ECG). Due to the potential risk of Amisulpride causing

arrhythmias, all participants were assessed for QTc abnormalities in the ECG prior

to inclusion in the study. No participant presented with any abnormalities resulting

in the exclusion from the study. A letter to the participants’ general practitioner

(GP) was also organised to inform them of the participants’ involvement in the

study.

5.3.e. Prior to experimental visits

Participants were instructed to not consume alcohol for 24 hrs prior to the visit, to

only drink their normal intake of coffee or tea on the morning of each visit, and to

refrain from consuming other caffeinated drinks 2 hours prior to each visit.

Participants were also informed to not consume any psychoactive substances for

the duration of the study, or any over-the-counter medicine 48 hrs prior to each

visit.

5.3.f. Experimental Visit Protocol

5.3.f.i. Drug administration

One of three substances was administered to the participant at each experimental

visit; Cabergoline (Agonist) (1.25 mg), Amisulpride (Antagonist) (400 mg), or Sodium

Page 115: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

115

Chloride (Control) (20mg). All drugs hold a full product licence (EU). The

administration of the drug2 was double-blinded and each participant had each drug

condition once over the three visits in a randomised order. The three visits were

separated by at least ten days. The administration of the drug was recorded as time

0 hr and experimental testing commenced at 3 hr 30 mins post drug administration.

5.3.f.ii. Resting state

Previous research has indicated a potential correlation between the level of striatal

dopamine and the rate of blinks per minute (KARSON, 1983; Elsworth et al., 1991).

The blink rates of the participants were recorded using frontal EEG electrodes

during a resting state of nine minutes at +2 hours after taking the drug and prior to

any experimental testing. The participants were not informed that their blink rate

was being assessed to avoid affecting their spontaneous blink rate.

5.3.f.iii. Pain stimuli

A CO₂ laser [50W Synrad 48-5 J-series (J-48-5(S)W) Wavelength: 10600nm] was

used to deliver acute pain to the dorsal forearm surface (Brown, El-Deredy and

Jones, 2014). The CO₂ laser delivered a beam with a diameter of 15 mm and 150 ms

duration. For each test, the stimuli were delivered in an area measuring 4 x 5cm

and were delivered in a predetermined randomised path (Brown et al., 2008a). This

was to avoid habituation, sensitization, or skin damage.

5.3.f.iv. Psychophysics

Before starting the experimental protocol, psychophysics was used to calibrate the

laser to the individual’s pain sensitivity. An ascending method of limits procedure

was used, starting from 0.6 V with 0.06 increments each time. The participant used

an eleven point number rating scale (NRS) (0 - 10) to rate the intensity of the pain

perceived for each laser stimuli (0=no sensation, 4=pain threshold, 7=moderately

painful, 10=unbearably painful). The rating scale was introduced to the participant

2 The term drug or drug condition is in reference to all three substances; Control, Agonist and

Antagonist, unless specified.

Page 116: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

116

via these standardised descriptives to ensure that no explanation altered their

interpretation of the scale. The procedure was repeated three times to allow

participants to get used to the laser and was used to calculate the average voltage

to induce level 4 (low) and level 7 (moderate) pain. These two levels provided ‘low’

and ‘high’ pain stimuli for the laser protocol.

5.3.f.v. Main experiment

The participants received 120 laser stimuli at the two intensities (low and high)

separated into four conditions; Low (level 4), High (level 7), Unknown Low (level 4)

and Unknown High (level 7). To investigate the anticipation of a painful stimulus, a

3 second auditory countdown preceded the laser stimuli (see Figure 5.1). The first

auditory cue was presented concurrently with a visual anticipatory cue to indicate

the forthcoming laser stimuli. The participant was either shown ‘Low’, ‘High’ or

‘Unknown’. The presentation of the word ‘Unknown’ indicated that the laser

stimulus has an equal chance of being low or high. This was to investigate the

importance of certainty in the anticipation of the laser stimuli. The image was also

used as a visual fixation cue to discourage eye movements. After the laser stimuli,

the 0-10 numerical rating scale was shown on the screen and the participant rated

the intensity of the pain via a numerical keypad. The order of the stimuli was

randomised and separated into three blocks with short breaks in-between.

Following each block, the participants rated the unpleasantness on average for

each laser condition. Unpleasantness was scored using an 11-point NRS whereby 0

is not unpleasant and 10 is most unpleasant sensation.

Page 117: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

117

Figure 5.1: A schematic diagram of a single trial of the experimental paradigm. A computer monitor showed

the participant a visual cue of; Low, High or Unknown from -3s to +2s. A three second countdown of beeps at

-3, -2 and -1 allowed accurate anticipation of the laser stimuli at time 0s (red bar). The presentation of the

visual cue at -3s was concurrent with the first auditory cue. The visual cue was consistent throughout the

anticipation and laser stimulus. At +2s, an eleven-point number rating scale (NRS) was presented for the

participant to rate the laser stimulus. For each condition (Low, High, Unknown Low and Unknown High) there

were thirty trials. The trials were presented in a randomised order and divided into three blocks of forty

trials.

5.3.f.vi. EEG recording

A BrainVision MR EEG cap was used to record from 63 scalp electrodes using a

BrainVision-cap system [Standard BrainCap-MR with Multitrodes]. The arrangement

of the electrodes was modelled on the extended 10-20 system. Recording

parameters were set at: Filter (DC to 70 Hz), Sampling rate (1000 Hz), Gain (500). To

reduce electrical interference, a 50Hz notch filter was applied. Prior to starting the

laser protocol, resting states were recorded with eyes open and closed for two

minutes in all participants. This ensured that the experience prior to the experiment

was identical. The three experimental blocks were recorded separately to allow for

better artefact rejection of the EEG data.

5.3.g. Analysis Methods

5.3.g.i. Statistical analysis of behavioural data

Statistical analyses of the behavioural measures were carried out using IBM SPSS

Statistics 22 software. Prior to using statistical tests, the data was assessed for

normality using a combination of Q-Q plots, histograms, and the values of skew and

kurtosis. Normally distributed data was analysed using independent t-tests and

ANOVA tests, whilst for data reported to be not normally distributed, the

Visual Cue: Low / High / Unknown

-3 -2 -1 0 2 8

Rating Scale

Time (s)

Page 118: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

118

appropriate non-parametric test was utilised. Specific statistical tests for each

analysis step are reported in the results section.

5.3.g.ii. EEG analysis method

EEG pre-processing was carried out using EEGLAB toolbox (Delorme and Makeig,

2004) in MATLAB version R2015a (The Mathworks Inc) whilst statistical analysis was

carried out using SPM12 toolbox (Wellcome Department of Imaging Neuroscience,

Institute of Neurology, UCL, London, United Kingdom) running in MATLAB.

5.3.g.iii. Blink rate

For each participant and visit, the number of blinks per minute was calculated using

a MATLAB script3. The shape of waveform and topography was used to detect

blinks within the resting state data. A script which used the ICA function within the

EEGLAB toolbox was applied to count the blinks. The blinks per minute were

reported as the blink rate and were assessed for differences via repeated measures

ANOVA.

5.3.g.iv. EEGLAB Pre-processing

Pre-processing consisted of: removal and interpolation of bad channels, down-

sample to 500, low-pass filter of 20Hz and re-reference to the common average.

The four conditions were separated and -3500ms to 2000ms epochs extracted and

Linear detrend applied. Independent Component Analysis (ICA) was carried out on

all datasets using the SemiAutomatic Selection of Independent Components for

Artifact correction (SASICA) toolbox to select components to remove via pre-

determined thresholds. The thresholds were set to; Autocorrelation (threshold =

0.35 r, lag = 20 ms), Focal (threshold = 3.5 z), Focal trial (threshold 5.5 z), Signal to

noise (period of interest (POI) = [0 Inf], baseline (BL) [-Inf 0], threshold ratio = 0.5),

and Adjust Selection enabled. The thresholds were sufficient to remove artefacts

3 The MATLAB script was written and executed by Dr Grace Whitaker and Professor Wael El-Deredy.

All participants within this experiment were part of a wider study in collaboration with Dr Whitaker. The resting state data and assessment of the blink rate was a validation step in the all elements of the study and hence the data shared.

Page 119: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

119

from the majority of the datasets; however, a number of datasets required further

manual removal of eye-blink components where not picked up by SASICA.

5.3.g.v. SPM EEG Analysis

The pre-processed datasets were converted to Statistical Parametric Mapping

(SPM) compatible files. Statistical analysis was carried out to investigate the

anticipation-evoked potentials and the post-stimulus LEPs using scalp-level and

source localisation analysis techniques available in the SPM toolbox.

SPM scripts for batch processing were used to analyse the EEG data at the scalp

level and source localisation. Two baselining methods were applied to the data

analysis for the anticipation phase and were applied for scalp-level and source

localisation analysis (see 2.1.b.ii). Primary analysis applied distinct baselines (BLs) of

500 ms, occurring prior to each of the three auditory cues respectively, to analyse

each of the three anticipation phases. The BLs and time window of interest (TWOI)

were as follows; Early [BL: -3500 ms -3000 ms: TWOI: -2500 ms -2000 ms], Mid [BL:

-2500 ms -2000 ms: TWOI: -1500 -1000 ms] and Late [BL: -1500 -1000 ms: TWOI: -

500 0 ms] anticipation phases. The secondary analysis method applied a single

baseline of 500 ms prior to the first auditory cue [BL: -3500 ms -3000 ms] that was

common to every TWOI anticipation phase (early, mid and late). This second

analysis was conducted to enable comparison to previous studies (Brown et al.,

2008a; Brown, El-Deredy and Jones, 2014). All statistical analysis for the

anticipation phase was adjusted for multiple comparisons.

All analysis for the post-stimulus phase TWOI [200 ms 600 ms], centred on the LEP,

was baseline corrected to -500 ms prior to the laser stimulus [BL: -500 ms 0 ms].

5.3.g.vi. Source Localisation analysis parameters

SPM12 EEG and MATLAB scripts were used to estimate the sources of the

anticipation- and laser-evoked potentials using Low Resolution Electromagnetic

Tomography (LORETA). The forward model was created using an 8196 vertex

template cortical mesh coregistered to the electrode positions of the standard 10-

Page 120: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

120

20 EEG system. A three-shell boundary element (BEM) EEG head model available in

SPM12 was used to compute the forward-model. The images were smoothed with

a 12mm full-width-at-half-maximum (FWHM).

Source localisation results were reported as follows; to control for multiple

comparisons, a cluster-forming threshold of p<0.001 was used and resulting

clusters were considered significant at FWE (p<0.05). Significant clusters were also

restricted to >100 voxels in size and regions labelled using the Anatomical

Automatic Labelling (AAL2) toolbox in SPM. Small volume correction (SVC) was

applied using a 25 mm sphere to further investigate effects within regions of the

pain matrix, specifically the Insula, Anterior Cingulate Cortex (ACC), Thalamus and

Amygdala, using coordinates from the Brede Database (Årup Nielsen, 2003).

5.3.g.vii. EEG Analysis Statistical analysis

The anticipatory time windows of interest (TWOIs) were analysed using a repeated-

measure three-way ANOVA, with within subject factors of Drug (Control v Agonist v

Antagonist), Certainty (Known v Unknown), and Expectation (Known Low v Known

High). The analysis of the post-stimuli TWOI was conducted using a repeated

measure three-way ANOVA, with within subject factors of Drug (Control v Agonist v

Antagonist), Certainty (Known v Unknown), and Intensity (Low v High).

5.4. RESULTS

5.4.a. Behavioural results

5.4.a.i. Laser Sensitivity

To assess whether dopamine modulation evoked changes in sensitivity to the laser

stimuli, the participants’ tolerance to the laser was compared between each drug

condition. The voltage (V) required to induce a high (level 7) pain rating that was

determined by the psychophysics procedure was analysed for within-subject

differences using a repeated measures one-way ANOVA. The mean ±SD laser

energy (V) was calculated for control (1.93±0.43 V), agonist (1.98±0.49 V) and

Page 121: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

121

antagonist (1.97±0.41 V). There was no significant effect reported between drug

conditions F(2, 50)=0.465, p=0.631, signifying no drug-induced effect on sensitivity

to the nociception.

5.4.a.ii. Pain rating

Throughout the experiment, the participants rated the pain of the laser stimuli to

establish whether dopaminergic manipulation and anticipatory cues affected the

rating of the stimuli (see Table 5.1). The mean pain rating score for each drug

condition was calculated and analysed using a repeated measures three-way

ANOVA with within-subject factors of drug (control, agonist and antagonist),

certainty (known and unknown), and intensity (low and high). The pain rating scores

were normally distributed, as assessed by Shapiro-Wilk's test of normality (p > .05).

The within-subject comparison of the three Drug Conditions (Control, Agonist &

Antagonist) reported no effect on pain rating F(2, 50) = 0.60, p=0.552. This outcome

was expected as the laser intensity was calibrated to the participants’ individual

pain sensitivity on each visit to induce a score of 7 on the pain NRS. The calibration

was completed +3hr30 mins post drug administration and thus pain rating was not

expected to differ between drug conditions.

Table 5.1: The pain rating scores for the experimental paradigm. The rating is separated into laser conditions

and reported as mean and SD.

As we expected, there was a significant effect of intensity [F(1, 25)=262.98, p<

0.001], such that high intensity laser stimuli were rated more painful than low

intensity. Although the assessment of the effect of Certainty (Known v Unknown) on

Page 122: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

122

the pain rating did not show a significant result F(1, 25)= 3.33, p=0.80, a two-way

interaction between Certainty and Intensity was shown [F(2, 50)=47.82, p<0.001].

Pain ratings increased from Low (2.95±0.94) < Unknown Low (6.31±0.70) <

Unknown High (5.73±0.70) < to High (6.31±0.70). This shows that when participants

received a certain cue of Low, they rated the pain less than when they received the

Unknown cue and received a low intensity stimulus. In contrast, when the

participants were presented with a certain cue of High, they rated the pain higher

than when they received the Unknown cue and a high intensity stimulus was

delivered. This demonstrates how the rating of the low and high intensity stimuli

was affected differently by the certainty during the anticipation period. This

manipulation of pain perception by anticipatory cues replicates previous findings

(Brown et al., 2008b) and acts to validate the experimental paradigm.

Interestingly, there was a three-way interaction between drug, certainty and

intensity [F(2, 50)=5.60, p=0.006]. This indicates that the modulation of the D2

dopamine receptors via the agonist and antagonist affected the interaction

between certainty and intensity. Follow up two-way ANOVAs established how the

drug condition affected the interaction between certainty and intensity. Firstly, the

comparison of control and agonist revealed no three way interaction between drug,

certainty and intensity [F(1,25)=2.658, p=0.116]. However, the comparison of

control versus the antagonist did reveal a significant three way interaction, [F(1,

25)=9.535, p<0.005], such that the effect of uncertainty was increased within the

antagonist condition. The significant interaction was followed-up with paired t-tests

to establish the differences observed between the placebo and antagonist

condition. To investigate the degree of modulation via certainty, the Unknown pain

rating was deducted from the Known pain rating for low and high laser intensity. In

contrast to the placebo condition, the Antagonist showed a significantly higher

degree of certainty modulation for the high laser stimuli T(1,25)=3.525, p=0.002,

and did not show evidence of a difference within the low laser condition

T(1,25)=1.009, p=0.322. The degree of effect that the certainty condition had on the

pain rating for each Drug Condition, is shown in Figure 5.2.

Page 123: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

123

Figure 5.2: The effect of certainty on each laser intensity was calculated and plotted for each drug. The

difference between Known and Unknown pain rating of each laser intensity was calculated by subtracting the

Known rating from the Unknown rating. A positive value is indicative of a higher rating in the Unknown

condition, whereas a negative value is indicative of a lower rating in the Unknown condition. The bigger the

number, the bigger the effect of certainty had on pain rating. Therefore, the graph shows that for Low

intensity, there was an increase in pain rating following the Unknown visual cue within all drugs. In contrast,

for High intensity, there was a decrease in pain rating following the Unknown visual cue within all drugs. Both

the agonist and antagonist caused a bigger effect of certainty on pain rating compared to the control

condition, with the antagonist showing a significantly bigger difference in comparison to the Control

condition. **=p<0.005.

5.4.a.iii. Unpleasantness rating

The rating of unpleasantness was recorded for each laser condition (Low, High,

Unknown Low & Unknown High) and documented at three time points during the

laser experiment. The mean unpleasantness rating for each drug condition (Control,

Agonist & Antagonist) (Table 5.2) was compared and reported no significant

difference [F(2, 30) = 2.766, p=0.079]. In contrast, there was an expected effect of

Intensity, such that high intensity stimuli were rated more unpleasant than low

intensity stimuli [F(1, 15) = 197.75, p=0.000]. There was also an effect of Certainty

Low High

-2

-1

0

1

2

The effect of certainty on Low and High laser

intensity within each drug cond ition.

Laser Intensity

Un

kno

wn -

Kn

ow

n P

ain

Ratin

g

**

Agonist

Control

Antagonist

Page 124: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

124

(Known v Unknown) [F(1, 15) = 11.924, p=0.004] on unpleasantness ratings which

did not differ between the three drug conditions. The effect demonstrated that the

presentation of the Unknown cue caused a higher rating in unpleasantness

compared to the Known cues (‘Low’ and ‘High’).

Table 5.2: The unpleasantness rating for each laser condition for each Drug Condition. There was a significant

difference between low and high laser intensity, and the unknown conditions were rated significantly more

unpleasant than the known conditions. There was no significant difference between the three drug

conditions. Unpleasantness score is reported as mean and SD.

5.4.b. EEG results

5.4.b.i. Blink rate analysis

Blink rate has been shown to have a potential relationship with striatal dopamine

activity. As such, we analysed blink rate in order to test whether the agonist and

antagonist had modulated striatal dopamine. The analysis of the blink rate during

the resting state recording highlighted a linear relationship between the eye blink

rate and the expected dopamine D2R activation (Figure 5.3). The eye blink rate

(mean blinks per minute ±SD) was lowest in the Antagonist condition (23.58±13.13)

and highest in the Agonist condition (28.70±12.27), with the control eye blink rate

(26.77±13.35) in the middle of the Agonist and Antagonist. This monotonic

relationship was not significant, F(2, 46)=1.806, p=0.176. Nevertheless, the

monotonic relationship between the dopamine level and eye blink rate is consistent

with previous reports (Karson et al., 1984; Shukla, 1985; Taylor et al., 1999; Kleven

Unpleasantness Rating (VAS score /10)

Low High Unknown Low Unknown High

Drug Condition

Mean SD Mean SD Mean SD Mean SD

Control 2.39 1.30 6.49 0.91 2.56 1.44 7.08 1.30

Agonist 2.25 1.07 6.49 0.67 2.35 1.02 6.98 0.90

Antagonist 1.94 1.22 5.42 2.37 2.22 1.55 5.91 2.67

Page 125: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

125

and Koek, 1996) and demonstrated that the dosage of the agonist and antagonist

was sufficient to induce an increase and decrease of striatal dopamine respectively.

Figure 5.3: The eye-blink rate (EBR) calculated for each dopamine manipulation condition highlighted a

monotonic relationship between dopamine D2R activation and EBR. The D2 receptor agonist increased the

EBR and the antagonist reduced the EBR in comparison to the control condition. Data is presented as mean

and 95% confidence-intervals (cf. Cousineau, 2005).

5.4.b.ii. Scalp-level EEG analysis

The analysis of the EEG recording via whole head scalp-level analysis of the

anticipation TWOIs (early, mid and late) reported no main effect of Drug,

Expectation or Certainty for both baselining methods. The mean activity at each

electrode during anticipation is plotted as topographies for all drug conditions in

Figure 5.4.

Ag o n is t C o n tr o l An ta g o n is t

1 0

2 0

3 0

4 0

Bli

nk

s p

er m

inu

te

D o p a m in e M a n ip u la t io n

Page 126: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

126

Figure 5.4: Topography plots showing the average response for early-, mid– and late-anticipation for all

conditions. Data is presented as µV. All plot baselined to a common baseline of [-3500 –3000 ms] pre laser

stimulus.

In addition, the analysis of the post-stimulus TWOI [200 600 ms] at scalp-level, also

reported no significant effects of drug or certainty. A main effect of intensity

produced two clusters (p<0.000, kE =20418, x:-13mm y:-36mm, 464ms, and

p<0.000, kE = 3510, x:55mm y:-68mm, 450ms) with the T-contrast indicating an

enhanced activation during high pain in contrast to low pain (p<0.000, kE = 17732, x:

-13mm y: -41mm, 460ms).

5.4.b.iii. Source EEG analysis

The source localisation analysis reported novel results which we did not predict.

There were significant contrasts within the anticipatory and post-stimulus time

Page 127: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

127

window only when using pre-auditory cue baselines. Firstly, the mid-anticipation

TWOI [-1500 -1000 ms] reported significant clusters within the main contrasts of

Drug and Expectation (see Table 5.2). The subsequent T-contrasts of the drug effect

during mid-anticipation reported a lower degree of activity within both the agonist

and antagonist in comparison to the control condition. The clusters in both the

agonist and antagonist condition were located within the right hemisphere. The

lower activity in the agonist condition was reported in the right mid-temporal

region and angular gyrus, whilst the antagonist condition induced reduced activity

within the postcentral, mid temporal and inferior parietal regions (see Figure 5.5).

In addition to the main effect of drug condition, all drug conditions showed an

effect of Expectation (Known Low v Known High) during mid-anticipation. The

significant differences in activity were located in the left hemisphere, contralateral

to the pain, and showed augmented activity when participants were presented with

the visual cue of High in comparison to the visual cue of Low. The clusters were

located in the contralateral mid-temporal, mid-occipital and insula regions and are

shown in Figure 5.6.

The post-stimulus TWOI [200 600 ms] produced a main effect of intensity (Low v

High) and drug condition (control v agonist v antagonist) (see Table 5.3). The effect

of intensity during the post-stimuli time window was as expected such that the high

condition induced a higher degree of activation in contrast to the low condition (T-

contrast: High>Low: p = 0.000 F=8.44, kE =98792, x: 8, y: -14, z: -16). The main effect of

the drug condition highlighted a difference within the right insula, a main region of

pain perception. Subsequent T-contrast tests specified that the differences were

between the control and antagonist conditions, and the agonist and antagonist

conditions. Firstly, the antagonist was reported to cause a reduced level of

activation within the insula in comparison to the control condition (see Figure 5.7).

The agonist condition showed a significantly higher activation than the antagonist

within multiple regions; right hippocampus, mid- and inferior temporal region,

insula and the Heschl (auditory processing) (see Figure 5.7).

Page 128: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

128

Table 5.3: Source localisation significant results within mid-anticipation and the post stimulus TWOI. The mid-

anticipation TWOI is baseline corrected to the distinct pre-auditory cue time window. The significant clusters

were restricted to >100 voxels, reported at FWE correction with a threshold of p<0.025 to account for

multiple comparison. Results are divided into main F-contrast and post-hoc T-contrasts. The MNI coordinates

are reported as the peak-voxel response within the cluster. The brain regions were labelled using the

Anatomical Automatic Labelling (AAL2) atlas. The percentage overlap of the significant cluster with the brain

region is reported. The region with the highest percentage overlap is shown, unless an equivalent share of

per enta e overlap as observed. A label of ‘Unkno n’ as not reported. FWE=Family Wise Error,

K=number of voxels, F/T=F-contrast/T-contrast, Z=Z-score, Expectation=Known Low v Known High visual

cues, Antag = Antagonist, R = Right, L = Left.

Cluster-Level Peak-Level MNI coordinates

Brain Region P(FWE) K F/T Z x y z

Mid

An

tici

pa

tio

n

F-contrast

Drug 0.004 909 11.62 4.20 32 -44 34 R Inferior Parietal (48.0%)

0.013 700 9.79 3.79 34 -54 14 R Mid Temporal (56.3%)

Expectation

0.000 4098 25.90 4.85 -44 -48 6 L

L

Mid Temporal (35.3%)

Mid Occipital (23.1%)

0.013 783 18.64 4.09 -56 2 -16 L Mid Temporal (69.1%)

0.007 931 15.28 3.69 -14 -86 2 L

L

Calcarine (52.8%)

Lingual (37.2%)

0.005 390 15.65 3.73 -30 -18 8 L Insula (60.7%)

T-contrast

Drug:

Control > Agonist 0.031 693 3.92 3.87 52 -58 10

R

R

Mid Temporal (49.9%)

Angular gyrus (29.4%)

Drug:

Control > Antag 0.000 4184 4.60 4.52 34 -40 40

R

R

R

Postcentral (17.2%)

Mid Temporal (16.9%)

Inferior Parietal (15.0%)

Expectation:

High > Low

0.000 8555 5.09 4.98 -44 -48 6 L

L

Mid Temporal (30.4%)

Mid Occipital (17.9%)

0.003◊ 641 3.96 3.90 -30 -18 8 L Insula (58.8%)

Po

st S

tim

ulu

s

F-Contrast

Drug 0.022◊ 164 10.82 4.02 28 -26 14 R Insula (40.9%)

T-Contrast

Drug:

Control > Antag 0.033

◊ 131 3.73 3.68 28 -26 14 R Insula (45.0%)

Drug:

Agonist > Antag

0.022 748 3.93 3.88 30 -20 -16 R Hippocampus (54.3%)

0.010 940 3.80 3.75 64 -30 -6 R

R

Mid Temporal (52.0%)

Inferior Temporal (38.7%)

0.008◊ 347 4.14 4.07 28 -26 14

R

R

Insula (32.6%)

Heschl (18.9%) ◊

Small volume correction (SVC) has been applied using a sphere of 25 mm radius

> Symbolises that the left-sided condition showed a greater response in comparison to the right-sided condition.

Page 129: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

129

Figure 5.5: Source estimates during mid-anticipation for T-contrast of Control versus agonist (Cabergoline)

and antagonist (Amisulpride). The time window of mid anticipation [–1500 –1000 ms] was baseline corrected

to [–2500 –2000 ms]. Clusters are shown at FWE correction using MRIcroN software and regions labelled

using the AAL2 atlas. The agonist (blue) reported reduced activity within the right mid-temporal and angular

regions in contrast to the control. The antagonist (pink) reported reduced activity within the right

postcentral, mid-temporal and inferior parietal regions. The image is aligned via the peak-voxel at peak-level

and reported as x y z (mm).

Page 130: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

130

Figure 5.6: Source estimates during mid-anticipation for the Expectation T-contrast of known High versus

known Low. The time window of mid anticipation [–1500 –1000 ms] was baseline corrected to [–2500 –2000

ms]. Clusters are shown at FWE correction using MRIcroN software and regions labelled using the AAL2 atlas.

Two clusters of enhanced activity were seen in the high condition versus the low condition and were

contralateral to the noxious stimuli. The regions included the left mid temporal and the occipital lobe, and

the left insula. The image is aligned via the peak-voxel at peak-level and reported as x y z (mm).

Page 131: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

131

Figure 5.7: The drug effect reported using source localisation for post-stimulus TWOI [200 600 ms]. The TWOI

was baseline corrected to -500ms prior to the noxious stimuli. There was a significant difference between the

control and antagonist conditions, showing a lower degree of activity within the right insula in the antagonist

condition. The antagonist condition also reported a lower activity in comparison to the agonist within the

right hippocampus, mid/inferior temporal lobe, insula and Heschl. The alignment of the images is fixated on

the peak-voxel MNI coordinates from the peak-level results.

Page 132: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

132

5.5. DISCUSSION

In this study, we aimed to address the role of D2 dopaminergic receptors during

pain anticipation and pain perception by using EEG source localisation. The D2

receptor agonist Cabergoline and the antagonist Amisulpride were used to

modulate striatal dopaminergic activity within healthy volunteers. The agonist and

antagonist amplified the effect of anticipation on pain ratings, with the antagonist

inducing a significantly larger effect in comparison to the control condition. Using

EEG source localisation, we have shown that both an increase and decrease in

striatal dopamine D2R activity results in a reduced degree of activity during mid-

anticipation within regions of the right mid-temporal and parietal lobes in

comparison to the control condition. In addition, EEG source localisation

highlighted that the modulation of D2 receptor activity induced altered processing

during the perception of the laser stimuli, yet did not change behavioural outcomes

of pain sensitivity or pain rating. This latter result is comparable to the research by

Becker et al., (2013), whereby dopamine manipulation did not change the rating of

acute pain.

Importantly, the manipulation of striatal dopaminergic activity via the Agonist

(Cabergoline) and Antagonist (Amisulpride) was shown via the calculation of the eye

blink rate during resting state. Although the differences between the drug

conditions were not significant, the eye blink rate showed a monotonic relationship

with the level of striatal dopamine D2R activity and indicates that the dose of the

agonist and antagonist were sufficient to increase and decrease striatal dopamine

respectively (Karson et al., 1984; Shukla, 1985; Taylor et al., 1999; Kleven and Koek,

1996). Nevertheless, caution should be taken when interpreting eye blink rate as

there is recent evidence that eye blink rate does not correlate with dopamine

synthesis capacity or D2R availability, or that it is an inverse relationship (Sescousse

et al., 2018; Dang et al., 2017).

Page 133: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

133

5.5.a. Behavioural results

5.5.a.i. Pain sensitivity

Subjective pain intensity within this study was deliberately maintained at similar

levels across all drug conditions in order to maintain similar levels of anticipation.

Adjusting the laser intensity to each condition allowed us to identify that the

dopamine manipulation did not affect the participants’ sensitivity to the laser,

consistent with a previous study by Becker et al., (2013).

5.5.a.ii. Pain unpleasantness

The unpleasantness rating was not affected by dopamine manipulation. This result

is in contrast to previous research which has shown that acute reduction in global

dopamine results in the increase in unpleasantness ratings (Tiemann et al., 2014).

Furthermore, an investigation of dopamine neurotransmission in chronic back pain

has shown that a low abundance of D2/D3 receptors within the striatum is

correlated with the unpleasantness rating. Therefore, we would have expected that

the agonism and antagonism of the D2Rs would modulate the rating of

unpleasantness. Several reasons may explain the difference reported in this study.

Firstly the method of recording the rating of unpleasantness may not have been

sufficiently accurate due to it being recorded as a mean after each experimental

block. Also, the acute action of the D2R agonist and antagonist within this study

could have been insufficient to evoke changes in the affective processing and a

more persistent change in dopamine may be required to alter affective processing.

Another explanation could be that the feeling of unpleasantness is not a D2R-

dependent process.

5.5.a.iii. Pain rating

The rating of the pain intensity within this study was also not altered by dopamine

manipulation. As mentioned, this was expected as the stimulus intensity was

calibrated to the individuals’ subjective perception after drug administration.

However, the dopamine manipulation did cause a difference on the effect of

Page 134: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

134

Certainty (known v unknown) on Intensity (low v high), such that the effect of

uncertainty was amplified within both the agonist and antagonist condition, with

the largest difference seen between the control and antagonist conditions. Thus,

following an ‘Unknown’ cue, participants demonstrated reduced attentional

flexibility during both the antagonist condition when compared to placebo (i.e. a

reduced ability to perceive the strength of the subsequent stimulus following the

‘Unknown’ cue).

The activity of the midbrain dopaminergic neurons has previously been shown to be

involved in the anticipation of uncertain cues of motivational-related outcomes

(Bromberg-Martin, Matsumoto and Hikosaka, 2010). Thus one population of

dopamine neurons has been demonstrated to code the uncertainty of a

forthcoming reward whereas a different population codes the reward prediction

errors (Fiorillo, Tobler and Schultz, 2003). By coding uncertainty as well as reward

prediction errors, phasic and tonic increases in dopamine neurotransmission may

encourage exploratory ‘risk-taking’ behaviour as well as attentional flexibility

(Whitaker 2017). We propose that the cue of Unknown evoked a more uncertain

state which requires a higher salience value (Seidel et al., 2015a). It could then be

predicted that the coding of both uncertainty and reward prediction errors is

impaired in the antagonist condition (due to blocking dopamine responses). This

impairment in both the coding of uncertainty and of reward prediction might

explain the reduced attentional flexibility of our participants in the antagonist

condition (Whitaker 2017). Hence, the interaction of the dopamine manipulation

with the effect of Certainty on pain rating provides further evidence for a role of

dopamine in salience processing.

5.5.b. EEG results

5.5.b.i. Drug induced effect during anticipation

One of the present study’s main findings was a drug-induced reduction in neural

activity during the mid-anticipation phase. The agonist and antagonist both induced

a region-specific reduction in activity during the anticipation of pain when

Page 135: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

135

compared to the control condition. Although the result being in the same direction

could be considered to be unexpected, this result could be explained by the fact

that dopamine signalling is well-known to have an inverted-U type relationship

(Cools and D’Esposito, 2011; Vijayraghavan et al., 2007; Levy, 2009; Floresco, 2013;

Beggs and Plenz, 2003; Finke et al., 2010; Li and Backman, 2010; Goto, Otani and

Grace, 2007) (See Figure 5.8). The peak of the inverted-U curve is indicative to be

the optimum level of dopamine, with both low and high levels of dopamine being

sub-optimal. This relationship has been demonstrated in cognitive and memory

tasks (Cools and D’Esposito, 2011; Floresco, 2013), and understood further with

help from research into Parkinson’s disease and Schizophrenia.

Figure 5.8: A schematic diagram to signify the inverted-U relationship between Dopamine level and

performan e. T e verti al das ed lines are representative of t e ‘optim m’ ran e of dopamine for many

cortical processes.

During mid-anticipation, both the agonist and antagonist conditions induced a

reduced activity in the right mid-temporal lobe, a region involved in sensory

integration and semantic processing (Herath, Kinomura and Roland, 2001; Ishai et

al., 1999; Chao, Haxby and Martin, 1999; Tranel, Damasio and Damasio, 1997).

D2Rs are located within the mid-temporal lobe (Goldsmith and Joyce, 1996) and the

Page 136: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

136

binding potential of D2Rs within the right medial temporal cortex has been

inversely correlated with cold pain tolerance (Hagelberg et al., 2002). In addition, a

reduced abundance has been proposed to explain the aberrant information

processing in cognitive tasks in dementia (Joyce, Myers and Gurevich, 1998).

Therefore, the region has previously been demonstrated to have a role in the

integration of sensory and cognitive processing, and be central to an individual’s

pain tolerance. Therefore, this study’s finding of altered processing prior to pain

perception following dopamine D2R manipulation may indicate a potential role of

the mid-temporal lobe in top-down mechanisms associated with pain processing.

We have also highlighted differences within regions of the parietal lobe in both the

agonist and the antagonist conditions. The agonist condition showed a reduced

activity within the right angular gyrus, a region located within the parietal lobule

and connected with the mid-temporal lobe. Whilst the left angular gyrus is

responsible for language processing (Seghier, 2013; Binder et al., 1996; Pugh et al.,

2000), the right angular gyrus is associated with body representation (Spitoni et al.,

2013), attentional reorientation (Taylor et al., 2011), and the integration of

vestibular and somatosensory input (Blanke et al., 2002). Hence, we propose that

the dopamine manipulation may have altered these processes.

The differences seen in the parietal and temporal regions following dopamine

modulation may be linked with the orienting attention which is associated with

salience detection (Legrain et al., 2011). The network of regions associated with

pain processing is often referred to a salience detection system, and includes the

temporal and parietal regions (Legrain et al., 2011). The participants of this study

were also assessed for their attentional reorientation in a separate assessment

(Whitaker, 2017). The results of this assessment showed that both the agonist and

antagonist reduced attentional flexibility, and this aligns with the reduced

activation seen within the right parietal lobe in this study. The results also support

the inverted-u relationship between dopamine and performance. Hence, our

results indicate a potential disruption in the salience and orientating attention

network which is activated during the anticipation of the noxious stimuli.

Page 137: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

137

The antagonist induced a lower activity within the right postcentral gyrus, located

within the inferior parietal lobule and the location of the primary somatosensory

cortex. This area is responsible for tactile and pain processing (Backonja, 1996;

Bushnell et al., 1999; Corkin, Milner and Rasmussen, 1970) and has previously been

seen to be involved in the anticipatory response to tactile and painful stimuli

(Roland, 1981; Brown, El-Deredy and Jones, 2014). A reduced activation within the

right somatosensory cortex has been shown in Fibromyalgia patients during the

anticipation of pain (Brown, El-Deredy and Jones, 2014; Loggia et al., 2014). In

addition, an fMRI study has previously shown a reduced BOLD response in the right

somatosensory cortex within Fibromyalgia patients in comparison to controls

during painful pressure (Gracely et al., 2002). Hence a reduced activity within the

right somatosensory cortex has been associated with abnormal pain processing and

we have shown that the D2Rs could be involved in this process.

5.5.b.ii. Drug induced effect during laser perception

During the post-stimulus time window, the antagonist induced a reduced degree of

activity in the right insula in comparison to the control condition, and when

compared to the agonist, also showed reduced activity in the right hippocampus,

mid- and inferior-temporal lobe, and the Heschl. The change in activity during the

perception of pain was not correlated to any change in pain rating of intensity or

unpleasantness. Thus indicates that the processes affected were not involved in the

coding of these aspects of pain, and instead could be related to the integration of

sensory information.

The activation of the contralateral insula during pain perception has previously

been shown to be increased in patient groups whom have a reduced level of

baseline dopamine; Fibromyalgia (FM) patients (Gracely et al., 2002; Bradley et al.,

2000), Parkinson’s disease (Brefel-Courbon et al., 2005) and people with cluster

headaches (Hsieh et al., 1995). It would be predicted that the D2R antagonist,

would induce a response in healthy controls that is similar to that seen in these

patient groups. However, we have shown a contrasting result whereby the

antagonism of the D2Rs reduced the activity within the ipsilateral (right) insula

Page 138: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

138

during nociception. The specific manipulation of D2Rs is thus affecting the insula’s

processing during pain differently than the effect of global loss of dopamine. Our

results may be explained by the differences between acute manipulation of

dopamine receptors as seen in our study compared with the chronic reduction in

dopamine levels seen in PD and FM. It is well recognised that chronic, but not

acute, alteration in dopamine levels can lead to long term changes in synaptic

plasticity and may explain this difference (Jay, 2003; Shen et al., 2008; Arbuthnott,

Ingham and Wickens, 2000).

The insula has been considered to be involved in intensity coding (Derbyshire et al.,

1997), however, there is more evidence to show that the insula may play a much

more complex role in integrating both the sensory and affective aspects of pain

(Mazzola et al., 2010; Lu et al., 2016; Singer et al., 2004a), and integrating cognitive

and emotional processing. This parallels with the developing story for the role of

dopamine and D2Rs in pain, such that dopamine is important for correct

interpretation and integration of sensory stimuli. Hence, a persistent dysfunction of

the D2R (Martikainen et al., 2015) could amplify the altered insula activity observed

in this study and result in an impaired integration of sensory processing.

5.5.c. Future Considerations

An important consideration of the interpretation of this study is whether the

changes in the activity during anticipation are directly linked with pain processing

and whether long term dopamine dysfunction would impair the top-down

modulation of pain. The reduction in the activity within the aforementioned regions

could be independent of pain processing. Therefore, a future investigation which

uses salient (i.e. noxious) and non-salient (i.e. visual) stimuli could further decipher

D2Rs role in pain perception. In addition, the acute administration of D2R agonist

and antagonist does not fully enable us to understand how long term dopaminergic

transmission (with resultant changes in synaptic plasticity) can impact pain

processing. The fact that we have shown significant differences following acute D2R

manipulation within multiple aspects of pain processing, suggests that prolonged

changes in dopamine might magnify these changes.

Page 139: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

5. An investigation of the role of the dopamine D2 receptor in pain processing

139

5.6. CONCLUSION

In summary, these data demonstrate that manipulating the activity of D2Rs alters

aspects of pain processing. Firstly, we have shown for the first time, that D2R

antagonism induced a greater effect of uncertainty on pain rating, and that neither

agonism nor antagonism affected pain sensitivity, pain unpleasantness or pain

rating. The EEG recording highlighted that sub-optimal dopamine activity following

the agonist and antagonist evoked a reduction in activity within the right parietal

and temporal lobes during the anticipation of pain. The antagonist also induced a

reduction in the activity within the right insula during the perception of pain.

Therefore, we propose that the dopaminergic activity via the D2Rs is likely to be

involved in the top-down processes associated with pain perception, rather than

directly coding the intensity of pain. The strong role of dopamine neurons in

attention and salience, plus the participants’ showing impaired attentional

flexibility (Whitaker, 2017), and the importance of these factors in top-modulation

supports D2R neurotransmission as a strong candidate for involvement in the top-

down modulation of noxious stimuli.

Page 140: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Chapter 6

An EEG investigation of age-related differences in acute pain

Sarah Martin, Anthony Jones, Christopher Brown,

Christopher Kobylecki, Monty Silverdale

Page 141: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

141

6.1. ABSTRACT

The tolerance to experimentally induced pain has previously been shown to

increase with age, however, chronic pain rates increase with age. Therefore, within

this study we investigated whether age-related changes in central processes of pain

can help to explain the increased prevalence of chronic pain in the older

population. Previous research has shown that pain tolerance increases with age,

however, there is limited information on changes within the central processing of

pain. Participants were divided into a Young and Old group and underwent an EEG

recording whilst acute heat stimuli were delivered to their forearm. Our results

concluded that the Older cohort were significantly more tolerant of the pain

stimuli, and showed a reduced degree of brain activity during the anticipation and

perception of the pain stimuli. We have highlighted a possible age-related change in

central processing of nociception which will help to further understand the natural

changes in pain processing over time. In addition, the age-related difference in pain

processing highlights the importance for chronic pain conditions to be researched

within age-groups in which the disease is prevalent. Nevertheless, additional

research is required to confirm our findings due to differing protocol efficacy

between the two age groups.

6.2. INTRODUCTION

There is a substantial increase of chronic pain with age (Andersson et al., 1993;

Blyth et al., 2001a; Rustøen et al., 2005). Whilst the increase in chronic pain in the

aged population can be partly explained due to an accumulation of negative

lifestyle choices, such as a sedentary lifestyle, and age-related diseases such as

arthritis, there is emerging evidence of the deterioration of the pain system leading

to a heightened susceptibility to chronic pain (see review: Yezierski, 2012).

Studies investigating the age-induced changes in pain perception have mainly

focused on acute pain tolerance. In contrast to the increased rates of chronic pain,

the sensitivity of acute pain has been shown to reduce with age in the majority of

Page 142: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

142

studies demonstrating that with age comes a higher pain tolerance (see review:

Gibson and Farrell, 2004). The cause of altered pain tolerance could be explained by

the age-related changes in the peripheral nervous system such as slower

conductance of sensory nerves (Voitenkov et al., 2017; Palve and Palve, 2018), and

changes in the Aδ- and C-fibres (Chakour et al., 1996). Therefore, the development

of chronic pain within the older population is not merely due to increased

sensitivity to noxious stimuli, and is likely to be linked with altered central top-down

processing.

The processing of pain is multidimensional and depends on the integration of a

number of cortical processes. Abnormalities in central processing have been

associated with chronic pain conditions (Brown, El-Deredy and Jones, 2014; Dick,

Eccleston and Crombez, 2002; Gracely et al., 2004; Baliki et al., 2006; Latremoliere

and Woolf, 2009) which demonstrate the importance of cortical processing prior

and during pain processing. There is evidence that maladaptive top-down

modulation of pain, which takes into account the subjective aspect of pain

processing, is central to the development of chronic pain conditions. Hence, the

age-related changes seen in the central nervous system may negatively affect the

top-down modulation of pain and help to explain the increased susceptibility of

chronic pain.

Age-related changes have been demonstrated within regions associated with pain

processing, including a reduced brain volume within the prefrontal cortex,

hippocampus and brain stem structures, whilst the insula and putamen have shown

to reduce in activity during pain perception (Farrell, 2012). In addition, there is

evidence that there is an age-related deterioration in the cortical networks

associated with sensory processing, cognition and salience (Onoda, Ishihara and

Yamaguchi, 2012; La Corte et al., 2016; Eckert et al., 2012). Furthermore, the

efficacy of the endogenous pain control systems such as the descending pain

pathway and the diffuse noxious inhibitory controls (DNICs) reduce with age

(Lariviere et al., 2007; Grashorn et al., 2013) and potentially contributes to the

Page 143: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

143

development of chronic pain (Ossipov, Dussor and Porreca, 2010). These findings

indicate an age-related alteration in central top-down modulation of pain.

The investigation of the top-down modulation can be investigated by studying the

anticipation of noxious stimuli (Brown et al., 2008b; Brown, El-Deredy and Jones,

2014; Brown and Jones, 2008; Clark et al., 2008a; Jones, Brown and El-Deredy,

2013). The anticipation of pain has previously shown to be abnormal within chronic

pain conditions (Brown, El-Deredy and Jones, 2014; Jones et al., 2012) and is

indicative of a maladaptive pain network. In addition, the activity during the

anticipation of a noxious stimulus is associated with the assignment of a salience

value to the forthcoming stimulus (Seidel et al., 2015a; Wiech et al., 2010; Menon

and Uddin, 2010) and has been shown to be correlated to the pain perceived.

Therefore, for the first time, we have compared the anticipatory processing to a

noxious stimulus in young and old cohorts via EEG recording and the delivery of

noxious stimuli. Any differences observed aim to improve our understanding of how

pain perception changes over time. Also, it will improve the translatability of pain

studies using EEG conducted in young healthy volunteers to chronic pain conditions

which are more prevalent in the older population.

Importantly, we will address methodological considerations when using laser

induced pain stimuli and EEG recordings in the different age groups. We will also

discuss the possible limitations of interpretation of EEG recordings between age

groups due the changes seen in the aging nervous system.

Page 144: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

144

6.3. METHODS

The study was approved by the local ethics committee and all participants gave

informed consent according to the Declaration of Helsinki to participate in the

study.

6.3.a. Participants

A total of fifty healthy participants took part in the study and were divided into two

age groups. The young age group consisted of twenty-six participants aged between

18 and 32 years old (mean age 22.2 years, SD 3.7 years). The older age group

included twenty-four participants aged between 45 and 82 years old (mean age

62.92 years old, SD 8.76 years).

The datasets included in this report were from two separate studies using identical

experimental methods. The young group reported in this paper are the participants

within Chapter 5 of this thesis during the control condition of the study4. The old

group in this study are the healthy control participants reported in Chapter 3 of this

thesis. The data used is from their first session completing the experimental

protocol. A difference within the recruitment of the participants is that the Young

cohort was selected for a middle score of the BIS-11 questionnaire with the aim of

limiting baseline dopamine variability, whereas the Old cohort did not complete the

questionnaire. Therefore, there is a potential impact of different variability within

the baseline dopamine levels within the two groups.

6.3.b. Experimental design

6.3.b.i. Pain Stimuli

A CO₂ laser was used to deliver acute pain to the dorsal forearm surface. The CO₂

laser delivered a beam with a diameter of 15 mm and 150 ms duration. For each

4 Participants were part of randomised, repeated measure, double-blinded study design. The study

involved the administration of a D2 dopamine receptor agonist (Cabergoline) or antagonist (Amisulpride), or an inactive control (Sodium Chloride) (20mg) on three separate visits. The data collected from the control condition was used in this analysis. The control visit was visit 1 for 9 participants, visit 2 for 11 participants and visit 3 for 8 participants.

Page 145: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

145

test, the stimuli were delivered in an area measuring 4 x 5cm and was delivered in a

predetermined randomised path (Brown et al., 2008a). This was to avoid

habituation, sensitization, or skin damage.

6.3.b.ii. Psychophysics

Before starting the experimental protocol, psychophysics was used to calibrate the

laser to the individual’s pain sensitivity. An ascending method of limits procedure

was used, starting from 0.6 V with 0.06 increments each time. The participant used

an eleven point NRS (0 - 10) to rate the intensity of the pain perceived for each

stimuli (0=no sensation, 4=pain threshold, 7=moderately painful, 10=unbearably

painful). The rating scale was introduced to the participant via these standardised

descriptives to ensure that no explanation altered their interpretation of the scale.

The procedure was repeated three times to allow participants to get used to the

laser and was used to calculate the voltage used for the Low and High laser

intensities in the main experiment.

The High laser intensity was calibrated to be the voltage required to induce level 7

(moderate pain) and the Low laser intensity was calculated to be level 4 (just

painful) in both groups.

6.3.b.iii. Main experiment

The participants received 120 laser stimuli at the two intensities (low and high)

separated into four conditions; Low (level 4), High (level 7), Unknown Low (level 4)

and Unknown High (level 7). To investigate the anticipation of a painful stimulus, a

3 second auditory countdown preceded the laser stimuli (see Figure 6.1). The first

auditory cue was presented concurrently with an anticipatory cue to indicate the

forthcoming laser stimuli. The participant was either shown ‘Low’, ‘High’ or

‘Unknown’. The presentation of the word ‘Unknown’ indicated that the laser

stimulus has an equal chance of being low or high. This was to investigate the

importance of certainty in the anticipation of the laser stimuli. The image was also

used as a visual fixation cue to discourage eye movements. After the laser stimuli,

the 0-10 numerical rating scale was shown on the screen and the participant rated

Page 146: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

146

the intensity of the pain. The Young group used a handheld keypad to enter a pain

rating, whilst the Old group verbally reported their pain rating5. The order of the

stimuli was randomised and separated into three blocks with short breaks in-

between.

Figure 6.1: A schematic diagram of a single trial of the experimental paradigm. A computer monitor showed

the participant a visual cue of; Low, High or Unknown from -3s to +2s. A three second countdown of beeps at

-3, -2 and -1 allowed accurate anticipation of the laser stimuli at time 0s (red bar). The presentation of the

visual cue at -3s was concurrent with the first auditory cue. The visual cue was consistent throughout the

anticipation and laser stimulus. At +2s, an eleven-point number rating scale (NRS) was presented for the

participant to rate the laser stimulus. For each condition (Low, High, Unknown Low and Unknown High) there

were thirty trials. The trials were presented in a randomised order and divided into three blocks of forty

trials.

6.3.b.iv. EEG recording

A BrainVision MR EEG cap was used to record from 63 scalp electrodes using a

BrainVision-cap system [Standard BrainCap-MR with Multitrodes]. The arrangement

of the electrodes was modelled on the extended 10-20 system. Recording

parameters were set at: Filter (DC to 70 Hz), Sampling rate (1000 Hz), Gain (500). To

reduce electrical interference, a 50Hz notch filter was applied. Prior to starting the

laser protocol, resting states were recorded with eyes open and closed for two

minutes in all participants. This ensured that the experience prior to the experiment

5 The difference in pain rating methodology was due to the participants within the Old group were the control group for the investigation of pain processing in people with Parkinson’s disease. As the Parkinson’s patients had limited movement, the pain rating was completed verbally in both groups. The addition of the keypad for pain rating within the Young cohort was the return to standard practice of the experimental protocol.

Page 147: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

147

was identical. The three experimental blocks were recorded separately to allow for

better artefact rejection of the EEG data.

6.3.c. Analysis Methods

6.3.c.i. Statistical analysis of behavioural data

Statistical analyses of the behavioural measures were carried out using IBM SPSS

Statistics 22 software. Prior to using statistical tests, the data was assessed for

normality using a combination of Q-Q plots, histograms, and the values of skew and

kurtosis. Normally distributed data was analysed using independent t-tests and

ANOVA tests, whilst data reported to be not normally distributed, the appropriate

non-parametric test was utilised. Specific statistical tests for each analysis step are

reported in the results section.

6.3.c.ii. EEG analysis method

EEG pre-processing was carried out using EEGLAB toolbox (Delorme and Makeig,

2004) in MATLAB version R2015a (The Mathworks Inc) whilst statistical analysis was

carried out using SPM12 toolbox (Wellcome Department of Imaging Neuroscience,

Institute of Neurology, UCL, London, United Kingdom) running in MATLAB.

6.3.c.ii.a. EEGLAB Pre-processing

Pre-processing consisted of; removal and interpolation of bad channels, down-

sample to 500, low-pass filter of 20Hz and re-reference to the common average.

The four conditions were separated and -3500ms to 2000ms epochs extracted and

Linear detrend applied. Independent Component Analysis (ICA) was carried out on

all datasets using the SemiAutomatic Selection of Independent Components for

Artifact correction (SASICA) toolbox to select components to remove via pre-

determined thresholds. The thresholds were set to; Autocorrelation (threshold =

0.35 r, lag = 20 ms), Focal (threshold = 3.5 z), Focal trial (threshold 5.5 z), Signal to

noise (period of interest (POI) = [0 Inf], baseline (BL) [-Inf 0], threshold ratio = 0.5),

and Adjust Selection enabled. The thresholds were sufficient to remove artefacts

Page 148: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

148

from the majority of the datasets; however, a number of datasets required further

manual removal of eye-blink components where not picked up by SASICA.

6.3.c.ii.b. SPM EEG Analysis

The pre-processed datasets were converted to Statistical Parametric Mapping

(SPM) compatible files. Statistical analysis was carried out to investigate the

anticipation evoked potentials and the post-stimulus LEPs using scalp-level and

source localisation analysis techniques available in the SPM toolbox.

SPM scripts for batch processing were used to analyse the EEG data at the scalp

level and source localisation. Two baselining methods were applied to the data

analysis for the anticipation phase and were applied for scalp-level and source

localisation analysis (see 2.1.b.ii). Primary analysis applied distinct baselines (BLs)

of 500 ms, occurring prior to each of the three auditory cues respectively, to

analyse each of the three anticipation phases. The BLs and time window of interest

(TWOI) were as follows; Early [BL: -3500 ms -3000 ms: TWOI: -2500 ms -2000 ms],

Mid [BL: -2500 ms -2000 ms: TWOI: -1500 -1000 ms] and Late [BL: -1500 -1000 ms:

TWOI: -500 0 ms] anticipation phases. The secondary analysis method applied a

single baseline of 500 ms prior to the first auditory cue [BL: -3500 ms -3000 ms] that

was common to every TWOI anticipation phase (early, mid and late). This second

analysis was conducted to enable comparison to previous studies (Brown et al.,

2008a; Brown, El-Deredy and Jones, 2014). All statistical analysis for the

anticipation phase was adjusted for multiple comparisons.

All analysis for the post-stimulus phase TWOI [200 ms 600 ms], centred on the LEP,

was baseline corrected to -500 ms prior to the laser stimulus [BL: -500 ms 0 ms].

6.3.c.ii.c. Source Localisation analysis parameters

SPM12 EEG and MATLAB scripts were used to estimate the sources of the

anticipation- and laser-evoked potentials using Low Resolution Electromagnetic

Tomography (LORETA). The forward model was created using an 8196 vertex

template cortical mesh coregistered to the electrode positions of the standard 10-

Page 149: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

149

20 EEG system. A three-shell boundary element model (BEM) EEG head model

available in SPM12 was used to compute the forward-model. The images were

smoothed with a 12mm full-width-at-half-maximum (FWHM).

6.3.c.ii.d. EEG Analysis Statistical analysis

The EEG analysis was analysed via an analysis of covariance (ANCOVA) with

covariates of laser energy and average pain rating. All data was assessed for outliers

using SPM’s boxplot function and only outliers labelled as extreme outliers were

removed. The linear relationship between the covariates and the EEG data was

assessed via extraction of non-adjusted Eigenvariate data from the significant

clusters reported in the SPM statistics output. The homogeneity of regression

slopes was assessed between age and each covariate for each group. The data had

no outliers and the assumptions required for an ANCOVA were met.

For the analysis of the anticipatory TWOIs, a three way repeated measures ANOVA

was applied, with one between-subject factor of Group (Young vs Old) and two

within-subject factors of Certainty [Known (Low/High) v Unknown] and Expectation

[Low v High (Known)]. For the analysis of the post-stimuli TWOI, a three way

repeated measures ANOVA was applied, with one between-subject factor of Group

(Young vs Old) and two within-subject factors of Certainty [Known (Low/High) v

Unknown] and Intensity [Low v High (Known and Unknown)].

Source localisation results were reported as follows. To control for multiple

comparisons, a cluster-forming threshold of p<0.001 was used and resulting

clusters were considered significant at FWE (p<0.05). Significant clusters were also

restricted to >100 voxels in size and regions labelled using the Anatomical

Automatic Labelling (AAL2) toolbox in SPM.

Page 150: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

150

6.4. RESULTS

6.4.a. Laser behavioural results

The previous research showing an age-related reduction in sensitivity to

experimentally induced pain was investigated via comparing the sensitivity to the

laser in the Young and Old groups. An independent t-test reported a significantly

higher tolerance in the older group (Mean ±SD: 2.20±0.25) in comparison to the

young group (Mean ±SD: 1.93±0.43), F(1,48)=9.52, p=0.009.

To assess any differences in the rating of pain between the two groups, a mixed

three way ANOVA was carried out on the pain rating with a between factor of Age

(Young v Old), and within factors of Certainty (Known v Unknown) and Intensity

(Low v High). A group difference was reported showing that the old group had a

lower rating of the pain stimuli than the young group [F(1, 48)=34.90, p<0.000]

(Figure 6.2). This indicates that although the laser was calibrated to induce a

subjective moderate pain prior to the main experiment via the psychophysics

procedure, the older group significantly reduced their rating for the same intensity

when it was delivered in the main protocol.

The effect of certainty on the rating of the Low and High pain stimuli was consistent

between the two groups [F(1,48) = 55.27, p<0.000] such that the certainty (Known

(Low or High) v Unknown) of the visual cue affected the rating of Low and High

stimulation with contrasting results. The known Low intensity was rated lower than

the unknown Low condition, whilst known High intensity was rated higher than the

unknown High condition (Figure 6.2).

Page 151: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

151

Figure 6.2: Pain rating of laser stimuli. A) The pain rating of the laser stimuli is shown for each laser condition.

The Young (blue) group rated the pain significantly higher than the Old (red) group. B) The effect of certainty

on the rating of Low and High was consistent in both groups, such that the Unknown Low was rated higher

than known Low, whilst Unknown High was rated lower than known High. NRS: Number rating scale, Low:

‘ o ’ vis al e and lo laser intensity, i : ‘ i ’ vis al e and i laser intensity, Un o : ‘Unkno n’

visual cue and lo laser intensity, Un i : ‘Unkno n’ vis al e and i laser intensity. ata is plotted as

mean ± SD.

To assess whether either group habituated to the laser stimuli within the main

experiment, a linear regression was applied to establish whether the rating of the

Known High stimuli could be predicted by trial number. The known high stimuli was

selected for the habituation analysis due to it being calculated by identical

protocols within the psychophysics in each group, and so that the effect of

uncertainty did not need to be accounted for. We concluded that trial number did

not predict pain rating for the Young [F(1,28) = 1.042, p=0.316] or Old [F(1, 28) =

3.325, p=0.079] (see Figure 6.3).

Lo

w

Un

Lo

w

Hig

h

Un

Hig

h

0

2

4

6

8

T h e c o n tra s t in g e ffe c t o f c e r ta in ty

o n lo w a n d h ig h in te n s ity

Pa

in

Ra

tin

g (

VA

S /

10

)

* * *

Lo

w

Un

Lo

w

Hig

h

Un

Hig

h

0

2

4

6

8

D iffe re n c e in p a in ra tin g b e tw e e n

th e Y o u n g a n d O ld g ro u p s

Pa

in

Ra

tin

g (

VA

S /

10

)

Y o u n g

O ld

***

A) Difference in pain rating between the Young and Old groups

B) The contrasting effect of certainty on low and high intensity

Pai

n R

atin

g (N

RS/

10

)

Pai

n R

atin

g (N

RS/

10

)

Page 152: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

152

Figure 6.3: The pain rating for Known High stimuli was averaged across participants within each group at each

trial number. There was no association between the trial number and pain rating within either group which is

indicative of no habituation to the laser stimuli during the main experiment. NRS: Number rating Scale.

6.4.b. EEG Results

6.4.b.i. Sensor-level

The waveforms at Cz are presented in Figure 6.4 and present the individual

waveforms for participants in the Young and Old groups. The plots present the

spread of the data and the average for each group (black line). The averaged

waveform for each group is shown in Figure 6.5 and shows that the Old group had a

distinctively lower degree of activity during the stimulus preceding negativity (SPN),

a representation of anticipation, and a smaller LEP. This difference in the activity

between the Young and Old groups can also be seen in the topography plots in

Figure 6.5.

No habituation of pain rating was reported for high stimuli across trials

Pai

n R

atin

g (N

RS/

10

)

Page 153: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

153

Figure 6.4: Waveforms at Cz electrode for all participants in the Young and Old groups. Both plots

show the stimulus preceding negativity (SPN) during the anticipation of the laser stimuli (0 ms)

and t e s bseq ent laser evoked potential ( EP). Ea aveform represents a parti ipant’s

waveform averaged across all trials and the solid black line shows the averaged response within

each group. Data is baselined to -3500 -3000.

Page 154: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

154

Figure 6.5: ERP waveforms and topography plots. A) The average waveform at Cz electrode of the

anticipatory and post-stimulus response. Red: Old, Blue: Young. The data is presented as an average of all

laser-conditions and baselined to [–3500 –3000 ms]. The waveforms of the young and old groups are

distinctively different, with the young group showing a higher stimulus preceding negativity (SPN), and larger

laser-evoked potential (LEP). B) The topoplots represent the average activity at each electrode for all

participants in each group. The difference between Young and Old is also shown.

) rand avera e topo rap i plots for ea ro p

o n Old o n - Old

Early

Mid

ate

Time (ms)

-3000 -2000 -1000 0 1000

Pote

ntia

l (

V)

-4

-2

0

2

4

Old Young

A) rand avera e aveforms for ea ro p

Page 155: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

155

6.4.b.i.a. SPM analysis

Both baselining methods reported the same sensor-level differences between the

young and old groups. For clarity and to allow reference to results from Chapters 3,

4 and 5, here we will show statistical results using the primary baseline method

using baselines prior to auditory cues.

The Young group showed a significantly higher degree of activity during

anticipation. The characteristic topography of anticipatory processing is a negative

response over dorsal regions and a positive response over posterior regions. Our

results of comparing the Young and Old groups show two distinct locations of

significant clusters which are located in the centre of the top of the head (dorsal)

and the back of the head (posterior (Figure 6.6). Hence, the statistical results along

with the topographic plots (Figure 6.5) show that the Young group have a

significantly higher anticipatory response compared to the Old group. This

interpretation is supported by the SPM T-contrasts which report which factor (i.e.

Age group) had a more positive EEG amplitude in the significant clusters (Table 6.1).

The Young group showed a more negative dorsal response (as the Old group

showed a more positive response: Old>Young), and a more positive response over

posterior regions (Young>Old).

Figure 6.6: Topographic plots of the SPM F-statistics for the main group effect during anticipation and post-

stimulus time windows. The results are restricted to FWE correction and clusters of >100 voxels. The red

arrow indicates the highest peak value.

Page 156: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

156

Table 6.1: Scalp-level statistical results for whole-scalp analysis. Group differences were found within all

time-windows. The significant clusters were reported at FWE correction with a threshold of p<0.025 to

account for multiple comparison (due to two baseline methods). The results are divided into main F-contrast

and post-hoc T-contrasts. The MNI coordinates are reported as the peak voxel response within the cluster

and described with x and y coordinates and time (ms). The interpretation of the T-contrast comparisons are

such that the > sign denotes which factor (i.e. Age group) had a more positive EEG amplitude within the

cluster. Group = Young v Old, FWE = Family-wise error K = Number of voxels, F/T = F-contrast/T-contrast, Z =

Z-score.

6.4.b.ii. Source localisation

The source localisation analysis using the LORETA technique did not highlight any

significant contrasts between the two groups.

Cluster-Level Peak-Level

MNI coordinates

(mm)

Time

(ms)

P(FWE) K F/T Z x y

Early

F-contrast

Group 0.000 42426 47.99 6.42 13 13 -2420

0.000 29119 42.06 6.04 -9 -79 -2450

T-contrast

Group:

Young>Old 0.000 31969 6.49 6.15 -9 -79 -2450

Group:

Old>Young 0.000 47905 6.93 6.53 13 13 -2420

Mid

F-contrast

Group

0.000 40878 73.28 7.76 -9 2 -1462

0.000 14418 34.50 5.50 0 -79 -1480

0.000 5027 21.02 4.31 -4 -89 -1058

T-contrast

Group:

Young>Old

0.000 16324 5.87 5.62 0 -79 -1480

0.000 6607 4.58 4.46 -4 -89 -1058

Group:

Old>Young 0.000 44388 8.56 7.84 -9 2 -1462

Late

F-contrast

Group 0.000 39799 72.23 7.71 -4 2 -232

0.000 10846 28.02 4.97 -4 -95 -240

T-contrast

Group:

Young>Old 0.000 13905 5.29 5.10 -4 -95 -240

Group:

Old>Young 0.000 42859 8.50 7.80 -4 2 -232

Post-

Stim

F-contrast

Group 0.000 12166 49.16 6.49 0 -30 376

Intensity 0.000 21047 74.71 7.82 9 -36 462

0.000 4951 25.90 4.78 55 -68 492

Group*Intensity 0.013 1829 16.97 3.86 -4 -25 368

T-contrast

Group:

Young>Old 0.000 13636 7.01 6.60 0 -30 376

Intensity:

Low>High 0.000 6354 5.09 4.92 55 -68 492

Intensity:

High>Low 0.000 22348 8.64 Inf 9 -36 462

Page 157: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

157

6.5. DISCUSSSION

Here we have indicated that there may be an age related difference in pain

processing. We have shown that the younger cohort were more sensitive to the

laser stimuli and rated the pain higher than the older group. We have also shown

via scalp-level EEG analysis that the younger cohort showed a higher degree of

anticipation related activity and larger response during perception of pain.

Although caution must be taken when interpreting the results due to several

limitations of the investigation (see section 6.4.c) the results are indicative that the

central processing of pain may change with age.

6.5.a. Pain sensitivity

Firstly, the older group were less sensitive to the laser stimuli during the

psychophysics procedure and required a higher voltage of the laser. The finding

that the older cohort was less sensitive to the laser stimulation supports previous

knowledge of increased tolerance of a range of experimentally induced stimuli. The

experiments are reviewed in detail by Gibson and Farrell, 2004, and collates

research to show either an age related increase, or no change, in pain threshold to

heat, electrical and mechanical stimuli.

The psychophysics procedure allowed for the calibration of the laser voltage to the

individuals’ subjective sensitivity. Although the psychophysics protocol was

successful within both groups to induce a high (level 7) pain, the Old group reduced

their rating of the pain when the main experiment was started using the previously

calibrated voltage. Therefore, we have demonstrated that age can affect the

efficacy of the psychophysics protocol used to calibrate to a subjective experience.

This highlights the benefit of using a strict age range in pain research to limit the

variability. The reduction in the rating of the laser stimuli must be considered when

interpreting the results and are discussed in Section 6.4.c.

Page 158: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

158

6.5.b. Interpretation of anticipation phase in EEG results

Here we have shown a reduced degree of activity during anticipation in the older

group compared to the younger group. One potential explanation of this finding is

to consider the age-related changes in an individual’s relationship with pain

stimulation. The relationship with pain changes with the experiences that we have,

and as such our response to pain will ultimately change as we grow older. The top-

down processes such as context, prior experience and emotional state, are all able

to modulate the perception of pain and are likely to change with age. There is a

theory coined as the ‘positivity effect’ whereby older people are more likely to

favour positive stimuli over negative stimuli within cognitive tasks (Reed and

Carstensen, 2012). An fMRI study reported a reduced activation within the striatum

and insula during the anticipation of monetary loss in the older cohort in

comparison to the younger group (Samanez-Larkin et al., 2007). This age-related

reduced activity was not observed during the anticipation of monetary gain.

Therefore, the reduction in brain activity during anticipation of the laser stimuli that

we have seen in this study may be analogous to the age-related difference seen

associated with the ‘positivity effect’ (Reed and Carstensen, 2012; Samanez-Larkin

et al., 2007), i.e. older subjects have a tendency to pay less attention to negative

cognitive input. In addition, there is evidence to show that psychological resilience

is increased within the older population (Cicchetti and Cohen, 2006; Ong, Bergeman

and Boker, 2009). As such, within the older population, the noxious stimuli may

have been assigned a lower salience value in comparison to the younger cohort,

and resulted in a reduced level of activity during anticipation.

The development of chronic pain has been associated with dysfunctional brain

activity during the anticipation of pain (Gracely et al., 2004; Brown, El-Deredy and

Jones, 2014; Loggia et al., 2014). Therefore, in people with chronic pain, the normal

age-related reduction in the anticipatory activity to pain stimuli could be different

and represent abnormal top-down processing of pain. It would be interesting to

establish whether older people with chronic pain show the same ‘positivity effect’

seen in the healthy population. It could be hypothesised that older people with

Page 159: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

159

chronic pain would not show the typical disfavour for negative stimuli which is seen

in the healthy population. As such, if a higher salience was assigned to the negative

stimuli, and a higher degree of activity occurred, this may help to explain the

persistence of chronic pain.

6.5.c. Limitation of interpretation

The interpretations of these results are constrained by several limitations

associated with the comparison. Here we will outline the limitations and provide

suggestions of interpretation.

6.5.c.i. Recruitment

One constraint of interpretation includes the recruitment of the participants being

from two different studies. The participants within the Young age group were part

of a repeated-measures, double-blind and placebo-controlled study which

investigated the role of dopamine D2 receptors in pain perception. The data used

for this comparison is taken from the control condition when they received the

inactive compound Sodium Chloride (20 mg). Although this would have no direct

physiological effect, there is potential that the knowledge that it could be a

dopamine modulating drug could induce a difference in their response to the

experiment. In contrast, the Old group were the control group in an investigation of

pain perception in Parkinson’s disease, and no intervention was administered.

6.5.c.ii. Difference in Pain Rating

Although the psychophysics procedure successfully calibrated the laser voltage to

induce a high pain in both age groups, the resulting pain rating during the main

experiment was significantly lower within the older group and demonstrates that

the older participants habituated to the laser stimuli between the psychophysics

and main experiment. Importantly, the rating of the stimuli within the main

experiment did not show any habituation within either group. The drop in pain

rating was not shown in the Young group and the pain rating was hence used as a

covariate within the EEG data analysis. Although the covariate was included, it is

Page 160: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

160

difficult to conclude whether the decrease in activity during anticipation in the Old

group was not merely due to the participants anticipating a lower intensity of pain.

Whilst the young group rated the pain via a numeric keypad, the older group

reported their ratings verbally. The different techniques may have led to the

difference in pain rating due to the impact of psychological and social factors when

rating verbally in comparison to via a personal keypad. There is evidence that the

environment and social context of pain experiments can influence pain rating. For

instance, males are likely to reduce their pain rating when the researcher is female

in comparison to a male researcher (Levine and De Simone, 1991). Furthermore,

stoic behaviours of hiding pain severity have been shown to be a good predictor of

reductions in self-reported pain intensity (Yong, 2006; Cantwell et al., 2013).

Therefore, the verbal pain rating in the Old group may have been sensitive to the

additional psychological factors such as resilience (Ong, Bergeman and Boker,

2009), stoicism and rapport with the researcher, in comparison to the Young groups

use of the keypad.

6.5.c.iii. Age-related changes in EEG recording

Another consideration of comparing younger and older participants is the

differences in the EEG recording. A study by Duffy et al., (1984) reported no age-

related change in alpha frequency or amplitude, however, did report a dysfunction

of EEG desynchronization. In addition, a study which investigated the visual evoked

potential (VEP) in EEG recordings of young and middle aged adults concluded that

the older cohort showed a lower VEP amplitude and delayed latency (Dustman et

al., 1990). Although there is limited information regarding EEG age-related changes,

there are widely discussed differences in fMRI recording. A comparison in the

overall functional response to cerebral activity in fMRI highlighted that the older

participants showed a lower amplitude in the BOLD response compared to the

younger participants (Ross et al., 1997; Madden et al., 2004; Juckel et al., 2012).

Therefore, an age-related decline in neuroimaging may have produced the reduced

response seen in the older cohort. Further investigation is needed to establish

Page 161: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

6. An EEG investigation of age-related differences in acute pain

161

whether the reduced response is a consequence of reduced anticipation of pain, or

a result of a decline in neuroimaging quality.

6.6. CONCLUSION

Our investigation further strengthens the knowledge that tolerance to

experimentally induced pain is increased with age. We have also highlighted a

potential age-related reduction in brain activity during the anticipation and

perception of pain, and highlighted methodological considerations for experimental

paradigms in different ages. Although there are limitations which impair a confident

conclusion of the comparison, the reduced response may be highlighting the

‘positivity-effect’ within older cohorts whereby they show a reduced neural

response to negative outcomes. We have also shown that the current

psychophysics procedure is sensitive to age and is an important protocol choice for

future studies. Further investigations are required to understand the differences in

pain perception in young and older cohorts.

Page 162: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

Chapter 7

General Discussion

Page 163: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

163

7.1. MAIN AIM OF THESIS

The main aim of the research conducted in this thesis was to further understand

pain processing in Parkinson’s disease and to establish whether abnormal top-down

processing could help to explain the high prevalence of chronic pain seen in

Parkinson’s disease. The main focus of the research was to investigate the brain

activity during anticipation of pain as a technique to research top-down modulation

during pain processing.

The first study investigated pain perception in people with Parkinson’s disease

whilst off (Chapter 3) and on (Chapter 4) their medication. The second study

explored the role of dopaminergic transmission in pain perception in young healthy

volunteers (Chapter 5). And finally, age-related changes in pain perception were

explored (Chapter 6).

7.2. SUMMARY OF EXPERIMENTAL WORK

Chapters 3 and 4 describe the Parkinson’s disease patient study which investigated

the brain activity during anticipation and perception of pain in the patient group

and healthy age-matched controls.

Firstly, Chapter 3 reviews the results from the study conducted whilst the patients

were withdrawn from their medication. The behavioural results reported that the

PD group, compared to the healthy age-matched controls, were more sensitive to

the laser stimuli, showed higher scores in pain catastrophising, and yet showed no

difference in their mood or cognitive states (assessed via HADS and MoCA

respectively). Brain activity was assessed using LORETA source localisation and

concluded that there was an increased activation in the midcingulate region within

the PD group during mid and late anticipation in comparison to the control group.

This amplified response was independent of the PD motor severity, PCS, HADS and

chronic pain severity.

Chapter 4 (on) summarises the results from when the experimental protocol was

repeated in participants from the study reported in Chapter 3 (off) whilst the PD

Page 164: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

164

patients were on their Parkinson’s medication. The behavioural results showed a

higher sensitivity to the laser in the PD group, which is consistent with conclusions

drawn in Chapter 3 (off). The EEG data reported no differences between the PD

group on their medication and the healthy controls. Caution must be taken in

drawing firm conclusions due to the following limitations of the study: no

randomisation of experiment order, repetition of the experiment on the same day,

a potential time-of-day effect and a low n number. However, the study indicates

that the higher pain sensitivity seen in the PD participants is not affected by

increasing dopamine and suggests that dysfunction of other neurotransmitters such

as Serotonin or Noradrenaline may be to blame. The lack of difference in the EEG

results between the HC and PD on their medication could indicate that the increase

in dopamine ‘normalised’ the anticipatory response in PwPD seen following

withdrawal of their medication, but it did not ‘normalise’ their pain sensitivity.

Chapter 5 (D2R) reviews the pharmacological manipulation of dopamine D2

receptors to assess the role of dopamine in pain processing. The investigation was

carried out in young healthy volunteers and conducted using a repeated-measures,

randomised, double-blind, placebo-controlled protocol. The dopamine

manipulation did not change sensitivity to the laser, but EEG source localisation

analysis did show a significant effect on brain activity during the anticipation and

the perception of pain. The agonist (Cabergoline) and antagonist (Amisulpride)

induced similar reductions in the parietal and temporal lobes during anticipation in

comparison to the control condition. In addition, the antagonist induced a reduced

activation in the insula during the receipt of pain stimuli in comparison to the

control and agonist. Interestingly, the dopamine manipulation evoked a larger

effect of certainty on pain rating, with the antagonist inducing a significantly

amplified effect of the Unknown cue on the rating of the pain.

Chapter 6 (Age) reviews the differences in the behavioural and neural responses to

the experimental protocol. The aim of the analysis was to shed light on the age-

related changes in pain processing due to the higher degree of chronic pain within

the aging population. The comparison highlighted a higher pain sensitivity and

Page 165: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

165

anticipatory response in the Young group compared to the Old group. The reduced

pain rating in the Old group and age-related difference in EEG recording may have

influenced the results, and caution is needed when interpreting the results.

Nevertheless, the study indicated that there is an age-related reduction in

anticipatory processing to noxious stimuli.

The conclusions drawn from the EEG analysis were mainly from the pre-auditory

cue baselining method. The method was carried out in conjunction with a single

baselining method which did not highlight significant differences within the studies

(except for the Age comparison: Chapter 6). A potential reason for this is due to the

timing of the single baseline period (-3500 -3000 ms) being when a higher degree of

movement of the participants would be expected which increased variability in the

baseline time window. The PD participants especially were likely to use the interval

between the trials to relax their suppression of tremors or to move into more

comfortable condition, prior to the visual/auditory cue. The pre-auditory cue

baselining method, which highlighted significant differences, may have shown

differences due to the mid- and late- baselines being during focused attention on

the experiment (i.e. low variability). Due to two baselining methods being used, the

statistical significance was adjusted accordingly, and ensured reliability of the

results.

The conclusions drawn from each chapter and how they contribute to the

understanding of pain processing, both in Parkinson’s disease and the general

population, will be discussed in the following sections. Considerations for future

research and details of potential further research will also be discussed.

7.3. AN INTERPRETATION OF THESIS STUDIES

We propose that the dysfunctional dopaminergic system within the Parkinson’s

brain results in a disruption in the top-down modulation of pain, which predisposes

PwPD to develop chronic pain.

Page 166: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

166

The three main studies (Chapters 3, 4 & 5) which investigate pain in Parkinson’s

patients off and on dopamine medication, and the effect of dopamine modulation

on healthy participants, contribute to the understanding that dopamine plays a role

in top-down modulation in pain processing, whilst demonstrating that it is unlikely

to be central in coding the intensity of pain. This thesis can draw this conclusion

because we have shown no change in pain sensitivity following dopamine

manipulation, yet have shown changes in brain activity prior to pain stimulation, a

representation of top-down processing, following dopamine modulation. Top-down

modulation encompasses various neural processes associated with processing

nociceptive information such as: cognition, attentional orientation and emotional

aspects. Therefore, we conclude that changes in dopamine levels present in

Parkinson’s disease is likely to cause disruption within these processes and may

result in long-term changes in the brain’s ability to integrate nociceptive

information accurately and increase susceptibility to the development of chronic

pain.

7.3.a. Behavioural response

Firstly, the sensitivity to the laser stimuli was not shown to be modulated by

dopamine levels in the PD study or D2R study. The PD participants had a higher

sensitivity to the CO2 laser whilst both off and on medication. This finding was also

shown by Tinazzi et al., (2008) using CO2 laser stimulation, and Djaldetti et al.,

(2004) using a heat thermode. In contrast, (Brefel-Courbon et al., 2005) showed a

reduction in sensitivity to the cold pressor test (CPT) in PwPD following Levodopa

medication. The difference in the effect of dopamine medication may be due to the

differences in the method of inducing experimental pain. The CO2 laser stimulation

is comparable to an acute pain, whereas the CPT is regarded to be more analogous

to chronic pain (Rainville et al., 1992). Therefore, due to dopamine’s potential role

in modulating top-down processes during pain perception, it would be more likely

that the PD dopaminergic medication would improve CPT tolerance in contrast to

acute CO2 laser stimulation.

Page 167: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

167

In addition, the manipulation of D2Rs, via Cabergoline and Amisulpride, in healthy

participants also did not modulate the individual’s sensitivity to the laser. The

higher sensitivity to the laser in the PD participants (both off and on medication),

and no differences seen in the D2R study participants, indicates that the sensitivity

in PD patients is likely to be associated with other neurotransmitters such as

serotonin, noradrenaline and/or opioids.

Furthermore, the D2R study concluded that the manipulation of dopamine changed

the effect of certainty on pain rating of the two pain intensities. This shows that

there is dopamine-dependent processing during the anticipation of pain which

results in the alteration of the pain rating and strengthens our theory that

dopamine is involved in the cognitive/top-down processes related to pain

perception. However, it is important to state that the PD group did not show

evidence of an altered effect of certainty on pain ratings which was present

following the D2R manipulation. The translatability of results from a young healthy

cohort to Parkinson’s disease is limited due to age-related differences in

nociceptive processing, and the multitude of neurological changes present in the PD

brain. Yet a better understanding of dopaminergic transmission in nociceptive

processing will help to better understand, and treat, dysfunctions of pain

processing.

7.3.b. Neural response

A difference in brain activity during the anticipation of the laser stimuli was present

in the PD patients off their medication, and the D2R agonist and antagonist

conditions, and thus indicates that there are dopamine dependent processes prior

to the perception of pain. As previously discussed in Chapter 2, brain activity which

occurs prior to the perception of pain can represent top-down modulation of pain

perception, whereby factors such as cognition, attention and emotional state can

manipulate the perception of the pain.

Page 168: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

168

7.3.b.i. Neural response in PD

The increased anticipatory activity within the MCC/SMA of the PD patients whilst

off their medication is hence highlighting an underlying dysfunction of pain-related

processing. The amplified response was not observed following levodopa and

suggests that dopaminergic hypofunction may be responsible for the amplified

response. Therefore, the degeneration and long-term disruption in the

dopaminergic pathways seen in the PD brain are likely to result in persistent

disruption of top-down processes. In addition to the deterioration of the

dopaminergic systems potentially impairing central processing of pain, a possible

‘side-effect’ of dopamine treatments for patients in the early stages of PD is

dopamine hyper-function following L-DOPA administration (Poletti, 2018).

Dopamine hyper-function has been linked with aberrant salience assignment

whereby a neutral stimulus is attributed as a salient event (Poletti, 2018; Nagy et

al., 2012; Poletti et al., 2014). Therefore, the fluctuations between hypo- and hyper-

function of dopaminergic processes are likely to disrupt the interpretation of pain.

7.3.b.ii. Neural response following D2R manipulation

The manipulation of the dopamine D2R in young healthy participants also induced

changes in central processing during the anticipation of the laser stimuli and

highlights a potential role of dopamine activity in the central processes related to

pain. A common compensatory response to the loss of dopamine in PD is a

significant increase in the abundance of striatal D2 (and D1) receptors (Hassan and

Thakar, 1988; Ryoo, Pierrotti and Joyce, 1998), and drug-naïve PD patients have

shown supersensitivity in the D2R binding. A study by Christopher et al., (2014)

have also shown a reduction in D2R in the striatum and insula, within PwPD who

also presented with mild cognitive impairment (MCI). In theory, the change in the

anticipatory activity within regions associated with sensory integration and pain

perception in young healthy volunteers following D2R manipulation may highlight

that the pathophysiological changes in the D2R in the PD brain could also disrupt

pain processing within these regions. The potential reasons for why the neural

Page 169: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

169

response during anticipation was different in the PD patients and young healthy

volunteers following D2R manipulation are discussed in the forthcoming section.

The D2R study also showed a reduction in the Insula during the perception of the

laser stimuli within the antagonist group. The Insula is a region sometimes

considered to be involved in coding pain intensity, however, there is also evidence

that it has a wider role in the integration of sensory and affective processing in pain

(Derbyshire et al., 1997; Mazzola et al., 2010; Lu et al., 2016; Singer et al., 2004b).

The fact that the altered activity within the Insula within the antagonist condition

did not alter pain rating or pain sensitivity, supports the theory that the Insula is not

solely intensity coding, and further supports our theory that dopamine is not

central to intensity coding.

7.3.c. Parkinson’s Disease vs D2R manipulation in healthy participants

The D2R manipulation in the young healthy group induced a different alteration in

neural activity that was shown in PD patients whilst off their medication (Chapter

3). Therefore, the amplified response in the MCC/SMA within PwPD is likely to not

be solely due to altered D2R activity. This would be expected as PD is not a disorder

of D2Rs, and in reality is the combination of impaired signalling of numerous

neurotransmitters and structural changes across the brain. In addition, the acute

manipulation of D2Rs in healthy volunteers cannot be directly compared to the

chronic changes that have occurred in the PD brain. In addition, the action of taking

long-term medication, and the action of medication withdrawal will have also

produced unique differences in the PD group.

The PD participants off their medication have low striatal dopaminergic processing

and Chapter 3 concluded that they have an increased anticipatory activity within

the MCC/SMA prior to the pain stimuli. In contrast, the dopamine D2R antagonist in

young healthy volunteers evoked a reduction in activation in the temporal/parietal

regions during the anticipation of pain stimuli. The global loss of dopamine and the

antagonism of the D2R hence resulted in opposing effects on the degree of activity

during anticipation, and within different regions. There are numerous reasons for

the differences observed. Firstly, the global loss of dopamine in PwPD is different to

Page 170: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

170

the specific agonism/antagonism of the D2R, and therefore, cannot be directly

compared. In addition, the neurodegeneration associated with the PD brain such as

the deterioration of the dopaminergic, serotonergic and noradrenergic systems,

along with any age-related differences, mean that a direct comparison cannot be

carried out. Nevertheless, as stated previously, the D2R-related changes in the

healthy brain during anticipation of pain, and the changes in the D2R within the PD

brain, may further suggest an impaired central processing of pain in the PD brain.

7.3.d. Age, Pain and Dopamine

Chapter 6 discussed the finding of reduced brain activation in the older cohort

compared to the young control participants. Due to the focus of this thesis being on

the dopamine’s role in pain perception, an extra consideration to discuss is the age-

related decline in dopamine (Volkow et al., 2000, 1996b; a; Suhara et al., 1991).

There is evidence that dopaminergic activity changes with age. For instance, the D2

(Volkow et al., 1996b) and D1 (Suhara et al., 1991) receptors, and dopamine

transporters (Volkow et al., 1996a) decline with age, and the positive correlation

between midbrain dopamine level and frontal cortex activity changes to a negative

correlation (Dreher et al., 2008). In addition, within healthy aging, there is a

reduction in the D2R within the putamen and caudate, which correlates with a

reduction in motor and cognitive tasks (Volkow et al., 1998). Thus demonstrates

that the age-related reduction in the dopaminergic system can result in the changes

in the performance of dopamine-dependent processes.

We can only speculate that the natural reduction in dopamine and the decline of

dopaminergic processes may indicate that if the dopamine level in the healthy aged

population becomes too low, and drops below the ‘optimal’ level for performance,

there is an impairment in the top-down aspects of pain processing. Theoretically,

this could explain why chronic pain is more prevalence in the older population

however, dopamine is only one of many age-related changes in the brain and no

evidence as yet has investigated this relationship.

Page 171: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

171

To the best of my knowledge, there are no investigations which specifically

investigate the relationship of age-related changes in dopamine and its effect on

pain processing. Future investigations would be valuable to establish whether age-

related decline in dopamine contributes to the higher susceptibility of chronic pain

in the older population.

7.4. SUMMARY OF INTERPRETATION

In summary, acute modulation of dopamine transmission affects the processing

prior to pain perception. And although no change was seen in the pain rating, long

term changes in dopaminergic transmission and the top-down processes which are

dependent on dopamine, is likely to result in an impaired pain system which creates

persistent and augmented perception of pain. Previous research has shown an

inverted-u relationship between dopamine and the performance of central

processes. Therefore, Figure 7.1 summarises the potential effect that dopamine

level has had on central processing.

Page 172: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

172

Figure 7.1: A schematic diagram of the relationship between dopamine level and performance of

dopaminer i pro esses. It is important to state t at t e ‘performan e’ is not dire tly orrelated to t e brain

activity during the anticipation of t e laser stim li; rat er t at ‘performan e’ is t e

effectiveness/efficacy/accuracy of dopamine neurotransmission. We suggest that a dopamine level outside

of the optimal may induce an impaired top-down processing of pain.

Firstly, t e lo dopamine level in t e Parkinson’s patients red es t e optimal performan e of t e

dopaminergic processes, such as the precise prediction of stimuli, which may have resulted in the

a mentation of t e anti ipatory a tivity prior to pain stim lation ( an’t be for s re ntil e r le o t

serotonin, noradrenaline etc). However, following the administration of the dopaminergic medication, the

dopamine levels were increased and anticipatory processing did not differ to the HC group. The age-related

decline in dopamine has been taken into account such that the older Healthy controls (Blue Square) within

t e Parkinson’s st dy (C apters 3 & 4) are positioned on the lower side of the optimal dopamine level. We

propose that a de novo PD patient group would show a similar response to our off-medication group; yet

variability would be expected based on the severity of their diagnosis, and the unknown impact of

medication withdrawal and compensation mechanisms has had on our results.

The modulation of dopamine level via the D2 receptor agonist and antagonist is likely to have resulted in an

increase and decrease of dopamine respectively. Both low and high dopamine has been reported to reduce

performance in cognition, memory, attention due to sub-optimal dopamine levels. Therefore, the sub-

optimal dopamine level induce a change in the central top-down processing of the forthcoming stimuli and

also alter the effect of certainty on pain ratings.

Dopamine Level

Pe

rfo

rman

ce

Optimum range PD Off medication

PD On medication

Healthy Controls (Old)

Young Healthy Controls

Agonist

Antagonist

Page 173: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

173

7.5. NEUROIMAGING RESEARCH OF PAIN IN PARKINSON’S

The motivation of the research within this thesis was to address the concerning

high prevalence of chronic pain in people with Parkinson’s disease, and to establish

whether dysfunctional central processing within PD contributes to the persistence

of pain. To establish the potential significance of the research within this thesis in

the clinical setting, we need to understand where it fits in with the wider research

and how together the research could be clinically important. Therefore, here we

will summarise the research studies which shaped our theory of altered central

processing prior to the commencement of the PhD (pre-2015), and also discuss the

research which has been published during the PhD (post-2015).

At the beginning of the PhD, there was limited neuroimaging research published on

central pain processing in PD. An early publication by Schott, 1985 highlighted the

potential role of the dysfunctional dopaminergic pathways in altered pain in

Parkinson’s disease, however, focused research on central pain processing is limited

until the last decade.

7.5.a. Pre-PhD

Prior to starting the research within this thesis (pre-2015), there were few

neuroimaging studies which focused on the central processing of pain in PwPD. A

PET imaging study by Brefel-Courbon et al., (2005) showed that PwPD had an

increased activity within the right insula and PFC, and the left ACC, during the cold

pressor test compared to the healthy age-matched group. The augmented activity

was attenuated following Levodopa administration. The research conducted by

Schestatsky et al., (2007) and Tinazzi et al., (2008) investigated the LEP response to

CO2 laser stimulation using single or pairs of electrodes on the scalp. Schestatsky et

al., (2007) concluded that the LEP response at the electrode position of Cz was

amplified in PD patients who suffered from chronic pain, but not in those who did

not have pain and controls. The difference was attenuated or disappeared following

L-Dopa medication. In contrast, Tinazzi et al., (2008), recorded LEPs from electrodes

at Cz and Fz positions, from pain-free PD patients, and reported a reduction in LEP

Page 174: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

174

amplitude which was not affected by Levodopa administration. Therefore, the

study by Brefel-Courbon et al., (2005) highlighted a difference in central pain

processing in PD patients during the perception of pain. The research by

(Schestatsky et al., 2007a) and (Tinazzi et al., 2008) applied similar techniques to

conclude differences in the LEP in PD, however, the LEP is unlikely to shed-light on

alterations in top-down processing.

7.5.b. Post-PhD start

Following the start of the PhD (2015) there have been several neuroimaging studies

published which strengthen the theory that PD pathophysiology alters pain

processing and increase the likelihood of developing chronic pain. fMRI has been

the most widely used technique and has highlighted differences in regions

associated with pain processing in PwPD. Tan et al., (2015a) showed a reduced

functional connectivity in the putamen and midcingulate cortex, and between the

basal ganglia network and the salience network, during heat pain stimuli in drug-

naïve PwPD without pain. Also a reduced activation of the temporal gyrus and

insula was shown in subsequent analysis (Tan et al., 2015b). An fMRI and DTI

investigation of PwPD with chronic pain indicated a thinning in several regions

which are associated with pain processing (i.e. PFC, cingulate cortex & parietal

areas), and an amplified response within the cerebellum and right inferior temporal

areas, plus a reduction in the activity within the left frontal inferior orbital cortex

(Polli et al., 2016). There was one study which also investigated the anticipatory

response to laser pain in PD, and concluded that PD patients showed a reduced

activation in the ACC and dorsal lateral PFC during anticipation and an augmented

response in the MCC during the receipt of the pain (Forkmann et al., 2017). And

finally, a study by Tessitore et al., (2018) showed that drug-naïve, pain-free PD

patients showed an increase in neural activity within the left somatosensory cortex,

left cerebellum and right pons, during heat pain stimulation.

Therefore, the research within this thesis in combination with other research also

concluding altered central processing in PwPD (with or without pain) strongly

indicates a role of altered central processing in chronic pain in PwPD. There is

Page 175: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

175

therefore, a scientific rationale to promote the use of treatments which aim to

improve central processing (i.e. meditation, cognitive behavioural therapy etc) in

the treatment of pain in PwPD. For example, mindfulness meditation has

successfully been used to reduce pain and is considered to improve cognitive

processing important in modulating pain (Zeidan et al., 2012). Furthermore, EEG

imaging has shown that meditation can alter anticipatory neural responses to

painful stimuli and reduce the resultant unpleasantness ratings of the pain (Brown

and Jones, 2010). In addition, there is increasing evidence of Cognitive Behavioural

Therapy (CBT) as a treatment option for chronic pain. Along with improvements in

pain severity, fMRI has highlighted the beneficial changes in connectivity between

regions associated with nociceptive processing (Shpaner et al., 2014). Hence there

is converging evidence that non-pharmacological ‘alternative’ treatments may be a

valid treatment option for chronic pain which is considered to be due to

dysfunctional top-down processes such as cognition, expectations, attention and

emotion.

7.6. FUTURE CONSIDERATIONS

The limitations within the studies have been discussed within the individual

chapters, however, there are general considerations which should be taken into

account for future research.

7.6.a. Selection of pain stimuli

The use of the CO2 laser for pain stimulation was chosen for its optimal delivery

speed and activation of nociceptive fibres and previous use in pain research (Brown

and Jones, 2008; Clark et al., 2008b; Brown, El-Deredy and Jones, 2014). However,

the technique does not come without limitations. Although the laser selectively

activates the Aδ- and C-fibres which process pain, the activation of the C-fibres can

sometimes result in a dull burning sensation which reduces the ability to distinguish

the laser stimuli resulting in habituation. In contrast, due to the laser heating up the

skin over time, there is also the potential development of sensitisation to the laser.

The movements within a grid aim to avoid the development of sensitisation,

Page 176: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

176

however, due to the limited size of the grid, it is likely that the same area of skin will

receive more than one laser stimulus.

A further limitation of the laser which needs to be considered is the potential to

cause long-lasting pigment change or skin damage. All participants included in laser

experiments are informed of the likelihood of temporary pigment changes which

last up to 6 weeks. However, there is a small chance that the pigment changes can

last longer than this in a very small number of cases. Although there is a maximum

of energy that the laser can be set to, the individual differences in skin sensitivity

results in different reactions to the laser. There were no long-term changes in

pigment change or skin damage within the studies included in this thesis.

There are several other options to induce pain experimentally which could be

considered for future research. Firstly, the use of an electric shock allows for event

related potentials (ERPs) analogous to the laser evoked potentials (LEPs). However,

the stimulation activates all nerves and muscles below the electrodes and is not

specific to pain fibres. Also, the anticipatory response to electric pain is different to

that evoked by laser stimuli (Hird et al., 2018), plus the habituation of electrical

stimuli is common (Bingel et al., 2007).

Another consideration is whether the use of acute pain stimulation is optimal to

investigate chronic pain. A benefit of using acute pain is that it allows for the

investigation of anticipation prior to the stimuli, a technique to assess the top-down

processes that occur prior to pain which have been shown to be abnormal in

chronic pain conditions (Brown, El-Deredy and Jones, 2014). In addition to acute

pain, tonic pain induced via the cold pressor test (CPT) is also beneficial to research

chronic pain due to it inducing persistent pain which requires motivational,

attentional and emotional control (Verhoeven et al., 2010; Ahles, Blanchard and

Leventhal, 1983), and can investigate coping strategies of individuals (Berntzen,

1987; Efran et al., 1989; Geisser, Robinson and Pickren, 1992). The CPT involves the

submersion of the hand into cold water and has been used for the investigation of

affective and cognitive factors which influence pain perception (Birnie et al., 2014).

However, there is a high variability in the protocol of CPT between researchers and

Page 177: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

177

leads to a limited ability to compare results between studies. The CPT has

previously shown that Parkinson’s patients have a difference cardiovascular

response during the CPT, and the tolerance of the CPT has been shown to be

reduced in PD patients. However, to the best of our knowledge there have been no

investigations of neural activity during CPT in Parkinson’s disease. Therefore, it

would be beneficial to include the CPT in further neuroimaging investigations into

chronic pain in PD.

7.6.b. Protocol Design

The laser paradigm has been used in both experiments and no changes were made

to allow direct comparison between studies. The laser paradigm is a replication of

previous research into chronic pain conditions; however, improvements could be

made to the paradigm.

The current laser paradigm would be improved by including both innocuous and

noxious stimuli to allowing conclusions to be drawn that the neural response is

unique to the anticipation of noxious stimuli. For instance, the innocuous stimuli

could be a tactile stimulus and be cued by a visual cue of ‘No Pain’.

In addition, the reduction in the pain rating observed in the older participants

highlighted that an improvement to the protocol is needed so that the perception

of the pain is maintained throughout the experiment so that the anticipation is to a

consistent perceived intensity. Firstly, the psychophysics procedure could include a

more randomised up/down method to calibrate the laser to the participants’

subjective level 7. Also, the voltage of the laser could be updated throughout the

main experiment. Experience with the laser has led to the development of a

formula (see below) which allows for a standardised change in laser voltage that is

related to the average pain rating within each block of 30 laser stimulations.

𝒘 ∗ (𝒙 − 𝒚) = 𝒛 𝑤 = 𝑇𝑤𝑜 𝑖𝑛𝑐𝑟𝑒𝑚𝑒𝑛𝑡𝑠 𝑜𝑓 𝑙𝑎𝑠𝑒𝑟 𝑣𝑜𝑙𝑡𝑎𝑔𝑒 (0.12 𝑉) 𝑥 = 𝐴𝑣𝑒𝑟𝑎𝑔𝑒 𝑝𝑎𝑖𝑛 𝑟𝑎𝑡𝑖𝑛𝑔 𝑜𝑓 𝐻𝑖𝑔ℎ 𝑦 = 𝐷𝑒𝑠𝑖𝑟𝑒𝑑 𝑝𝑎𝑖𝑛 𝑟𝑎𝑡𝑖𝑛𝑔 (𝑖. 𝑒. 7) 𝑧 = 𝑉𝑜𝑙𝑡𝑎𝑔𝑒 𝑖𝑛𝑐𝑟𝑒𝑎𝑠𝑒

Page 178: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

178

7.6.c. Study Recruitment

7.6.c.i. Parkinson’s patients

For PD versus HC study, a possible improvement of the study design would have

been to use additional control groups which divided the PD group into people with

and without chronic pain, and healthy control group with no pain. This would allow

any differences observed in the PD group to be independent of the presence of

chronic pain and any abnormal brain response would hence be unique to PD

physiology. An additional comparison with a chronic pain group would also be

interesting to establish whether any maladaptive central processing within PD is

common to all chronic pain conditions (such as FM) or whether the abnormalities

are unique to PD.

In addition, the PD group could be better controlled by solely recruiting drug-naïve

participants, single medication usage, and/or similar disease progression or

duration. However, to remove the variability within the PD group would remove the

true representation of the variety seen in the PD population. The recruitment of a

small/specific participant is timely and often very difficult at a single-site

experiment. The recruitment criteria being restricted to people able to be

withdrawn from their medication already created a limited recruitment pool, and

hence a further restriction of recruitment would have resulted in delays and a false

representation of the PD population.

7.6.c.ii. D2R study

Within the Dopamine D2R modulation study, the participants attended three visits

and carried out the same laser protocol. The order of the drug condition (agonist,

antagonist or control) was randomised; however, there is a potential effect on

neural processing when a protocol is repeated. A possible effect is that the

participants’ response to the laser stimulation would be different on each visit,

especially due to the stimuli being novel to the participant on their first visit. This

potential effect was controlled by the participants receiving the three drug

conditions in a randomised order. However, for future research, we propose the

Page 179: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

179

use of a pre-study assessment to introduce the pain stimuli to the participants. This

may reduce potential chance of a higher sensitivity to the laser being seen on their

first visit compared to subsequent visits.

The choice to use a within-subject repeated-measure study design was selected

over a between-subject design as the positives outweighed the negatives. For

instance, a significantly higher n number would have been required for a between-

subject study design. Plus there is a high degree of variability within individuals in

their baseline dopamine, D2R properties, and pain perception, which could not

have been controlled for and could have led to high variability within and between

drug condition groups.

7.7. FUTURE RESEARCH

7.7.a. The role of Dopamine, Serotonin and Noradrenaline in pain

perception

The focus of the research within this thesis is on dopamine’s role in pain processing

due to PD being characterised by low dopamine levels. However, PD is also a result

of other neurotransmitter changes, such as serotonin, noradrenaline, choline, and

glutamate transmission. Therefore, understanding of the role of these

neurotransmitters in pain perception will increase our understanding of what

causes the potential dysfunction of central processes in PD.

Future research to compliment the findings within this thesis includes the

investigation of tryptophan (precursor of Serotonin) and tyrosine (precursor of

Dopamine & Noradrenaline) depletion in healthy volunteers. The design will be a

repeated-measure, triple-crossover, placebo-controlled, double-blind study. The

aim is to use the same laser and EEG protocol to investigate the effect of low levels

of Serotonin and Dopamine/Noradrenaline, with additional testing of the CPT

response due to its potential translatability to central processing associated with

chronic pain. Results of the study will hopefully help to explain how low levels of

Page 180: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

180

serotonin and noradrenaline (in addition to low dopamine), could contribute to

impaired central pain processing in PD.

Preliminary findings of the study indicates that the depletion of serotonin and

dopamine via their precursors Tryptophan and Tyrosine respectively, does not

change pain tolerance to the laser stimuli or the cold pressor test. The lack of

difference induced in pain perception in dopamine depletion is in line with our

results presented in the D2 study within this thesis. However, the serotonin

depletion has previously been shown to result in a decreased pain tolerance to

laser stimuli (Martin et al, 2017). Therefore, our results may indicate that there is a

degree of variability in the role of serotonin pain tolerance. EEG imaging indicated

that serotonin depletion evokes amplification in the anticipation-evoked neural

response to pain during the late anticipation time-window, plus increased activity

during the receipt of the laser. In addition, there was evidence that serotonin and

dopamine depletion reduced expectation- and certainty-evoked neural responses

compared to the control condition. Therefore, preliminary findings are in

agreement with our conclusions drawn within this thesis that dopamine and

serotonin are involved in the central processing associated with processing

nociceptive information, rather than coding the intensity of acute pain.

Page 181: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

7. General Discussion

181

7.8. FINAL CONCLUSION

In summary, the research conducted within this thesis supports the theory of

altered central pain processing in PD; and is likely to contribute to the high

prevalence of pain in PD. The role of dopamine in causing the abnormalities is not

confirmed, however, a possible normalisation of anticipation following dopamine

medication, and altered pain anticipation during D2R manipulation, may indicate

that dopamine is involved in the disruption in top-down processing seen in PD. Our

conclusions are strengthened by the recent neuroimaging studies investigating pain

in PD which have also confirmed altered central processing. Further investigations

of global loss of dopamine, noradrenaline and serotonin will help to further

understand the cause of these abnormalities. A better understanding of pain in PD

is essential to provide better treatment and improve the quality of life for people

with PD.

Page 182: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

182

References

Aarsland, D., Zaccai, J. and Brayne, C., 2005. A systematic review of prevalence

studies of dementia in Parkinson’s disease. Movement Disorders, 20(10), pp.1255–

1263.

Abbott, F. V. and Melzack, R., 1982. Brainstem lesions dissociate neural

mechanisms of morphine analgesia in different kinds of pain. Brain Research,

251(1), pp.149–155.

Ahles, T.A., Blanchard, E.B. and Leventhal, H., 1983. Cognitive control of pain:

Attention to the sensory aspects of the cold pressor stimulus. Cognitive Therapy

and Research, 7(2), pp.159–177.

Alba-Delgado, C., Llorca-Torralba, M., Horrillo, I., Ortega, J.E., Mico, J.A., Sánchez-

Blázquez, P., Meana, J.J. and Berrocoso, E., 2013. Chronic pain leads to concomitant

noradrenergic impairment and mood disorders. Biological psychiatry, 73(1), pp.54–

62.

Albers, J.A., Chand, P. and Anch, A.M., 2017. Multifactorial sleep disturbance in

Parkinson’s disease. Sleep Medicine, 35, pp.41–48.

Albrecht, D.S., MacKie, P.J., Kareken, D.A., Hutchins, G.D., Chumin, E.J., Christian,

B.T. and Yoder, K.K., 2016. Differential dopamine function in fibromyalgia. Brain

Imaging and Behavior, 10(3), pp.829–839.

Alexander, G.E., DeLong, M.R. and Strick, P.L., 1986. Parallel organization of

functionally segregated circuits linking basal ganglia and cortex. Annual review of

neuroscience, 9, pp.357–81.

Allen, N.E., Wong, C.M., Canning, C.G. and Moloney, N., 2015. The Association

Between Parkinson’s Disease Motor Impairments and Pain. Pain medicine (Malden,

Mass.) 17(3), pp.456-462.

Altier, N. and Stewart, J., 1999. The role of dopamine in the nucleus accumbens in

Page 183: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

183

analgesia. Life Sciences, 65(22), pp.2269–2287.

Aman, M.M., Jason Yong, R., Kaye, A.D. and Urman, R.D., 2018. Evidence-Based

Non-Pharmacological Therapies for Fibromyalgia. Current Pain and Headache

Reports, 22(5), p.33.

Amutio, A., Franco, C., Sánchez-Sánchez, L.C., Pérez-Fuentes, M. del C., Gázquez-

Linares, J.J., Van Gordon, W. and Molero-Jurado, M. del M., 2018. Effects of

Mindfulness Training on Sleep Problems in Patients With Fibromyalgia. Frontiers in

Psychology, 9, p.1365.

Andersson, H.I., Ejlertsson, G., Leden, I. and Rosenberg, C., 1993. Chronic pain in a

geographically defined general population: studies of differences in age, gender,

social class, and pain localization. The Clinical journal of pain, 9(3), pp.174–82.

Anselme, P. and Robinson, M.J.F., 2013. What motivates gambling behavior? Insight

into dopamine’s role. Frontiers in Behavioral Neuroscience, 7.

Arbuthnott, G.W., Ingham, C.A. and Wickens, J.R., 2000. Dopamine and synaptic

plasticity in the neostriatum. J. Anat, 196(4), pp.587-596.

Årup Nielsen, F., 2003. The Brede database: a small database for functional

neuroimaging. New York.

Azqueta-Gavaldon, M., Schulte-Göcking, H., Storz, C., Azad, S., Reiners, A., Borsook,

D., Becerra, L. and Kraft, E., 2017. Basal ganglia dysfunction in complex regional

pain syndrome - A valid hypothesis? European Journal of Pain, 21(3), pp.415–424.

Babiloni, C., Brancucci, A., Arendt-Nielsen, L., Del Percio, C., Babiloni, F., Pascual-

Marqui, R.D., Sabbatini, G., Rossini, P.M. and Chen, A.C.N., 2004. Cortical

Sensorimotor Interactions During the Expectancy of a Go/No-Go Task: Effects of

Painful Stimuli. Behavioral Neuroscience, 118(5), pp.925–935.

Backonja, M.-M., 1996. Primary somatosensory cortex and pain perception. Pain

Forum, 5(3), pp.174–180.

Page 184: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

184

Baillet, S., Mosher, J.C. and Leahy, R.M., 2001. Electromagnetic brain mapping. IEEE

Signal Processing Magazine, 18(6), pp.14–30.

Bair, M.J., Robinson, R.L., Katon, W. and Kroenke, K., 2003. Depression and Pain

Comorbidity. Archives of Internal Medicine, 163(20), p.2433.

Baixauli, E., 2017. Happiness: Role of Dopamine and Serotonin on Mood and

Negative Emotions. Emergency Medicine: Open Access, 07(02), pp.1–3.

Baliki, M.N., Chialvo, D.R., Geha, P.Y., Levy, R.M., Harden, R.N., Parrish, T.B. and

Apkarian, A.V., 2006. Chronic pain and the emotional brain: specific brain activity

associated with spontaneous fluctuations of intensity of chronic back pain. The

Journal of neuroscience : the official journal of the Society for Neuroscience, 26(47),

pp.12165–73.

Banic, B., Petersen-Felix, S., Andersen, O.K., Radanov, B.P., Villiger, M.P., Arendt-

Nielsen, L. and Curatolo, M., 2004. Evidence for spinal cord hypersensitivity in

chronic pain after whiplash injury and in fibromyalgia. Pain, 107(1), pp.7–15.

Barceló, A.C., Filippini, B. and Pazo, J.H., 2012. The striatum and pain modulation.

Cellular and molecular neurobiology, 32(1), pp.1–12.

Barnes, C.D., Fung, S.J. and Adams, W.L., 1979. Inhibitory effects of substantia nigra

on impulse transmission from nociceptors. Pain, 6(2), pp.207–215.

Barnes, J. and David, A.S., 2001. Visual hallucinations in Parkinson’s disease: a

review and phenomenological survey. Journal of neurology, neurosurgery, and

psychiatry, 70(6), pp.727–33.

Barone, P., 2010. Neurotransmission in Parkinson’s disease: beyond dopamine.

European journal of neurology : the official journal of the European Federation of

Neurological Societies, 17(3), pp.364–76.

Basbaum, A. and Fields, H., 1984. Endogenous pain control systems: Brainstem

Spinal Pathways and Endorphin Circuitry. Neuroscience, 7, pp.309-38.

Page 185: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

185

Becerra, L., Breiter, H.C., Wise, R., Gonzalez, R.G. and Borsook, D., 2001. Reward

Circuitry Activation by Noxious Thermal Stimuli. Neuron, 32(5), pp.927–946.

Becker, S., Ceko, M., Louis-Foster, M., Elfassy, N.M., Leyton, M., Shir, Y. and

Schweinhardt, P., 2013. Dopamine and Pain Sensitivity: Neither Sulpiride nor Acute

Phenylalanine and Tyrosine Depletion Have Effects on Thermal Pain Sensations in

Healthy Volunteers. PLoS ONE, 8(11), p.80766.

Beggs, J.M. and Plenz, D., 2003. Neuronal avalanches in neocortical circuits. The

Journal of neuroscience : the official journal of the Society for Neuroscience, 23(35),

pp.11167–77.

Behbehani, M.M., 1995. Functional characteristics of the midbrain periaqueductal

gray. Progress in Neurobiology, 46(6), pp.575–605.

Beiske, A.G., Loge, J.H., Rønningen, A. and Svensson, E., 2009. Pain in Parkinson’s

disease: Prevalence and characteristics. Pain, 141(1–2), pp.173–7.

Belasen, A., Youn, Y., Gee, L., Prusik, J., Lai, B., Ramirez-Zamora, A., Rizvi, K., Yeung,

P., Shin, D.S., Argoff, C. and Pilitsis, J.G., 2016. The Effects of Mechanical and

Thermal Stimuli on Local Field Potentials and Single Unit Activity in Parkinson’s

Disease Patients. Neuromodulation: Technology at the Neural Interface, 19(7),

pp.698–707.

Benarroch, E.E., 2009. The locus ceruleus norepinephrine system: functional

organization and potential clinical significance. Neurology, 73(20), pp.1699–704.

Benatru, I., Vaugoyeau, M. and Azulay, J.-P., 2008. Postural disorders in Parkinson’s

disease. Neurophysiologie Clinique/Clinical Neurophysiology, 38(6), pp.459–465.

Berardelli, A., Rothwell, J.C., Thompson, P.D. and Hallett, M., 2001. Pathophysiology

of bradykinesia in Parkinson’s disease. Brain, 124(11), pp.2131–2146.

Berardelli, A., Sabra, A.F. and Hallett, M., 1983. Physiological mechanisms of rigidity

in Parkinson’s disease. Journal of neurology, neurosurgery, and psychiatry, 46(1),

Page 186: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

186

pp.45–53.

Berna, C., Leknes, S., Holmes, E.A., Edwards, R.R., Goodwin, G.M. and Tracey, I.,

2010. Induction of depressed mood disrupts emotion regulation neurocircuitry and

enhances pain unpleasantness. Biological psychiatry, 67(11), pp.1083–90.

Bernheimer, H., Birkmayer, W. and Hornykiewicz, O., 1961. Distribution of 5-

hydroxytryptamine (serotonin) in the human brain and its behavior in patients with

Parkinson’s disease. Klinische Wochenschrift, 39(20), pp.1056–1059.

Berntzen, D., 1987. Effects of multiple cognitive coping strategies on laboratory

pain. Cognitive Therapy and Research, 11(6), pp.613–623.

Berridge, K.C. and Robinson, T.E., 1998. What is the role of dopamine in reward:

hedonic impact, reward learning, or incentive salience? Brain Research Reviews,

28(3), 309-69.

Bertolucci-D’Angio, M., Serrano, A. and Scatton, B., 1990. Mesocorticolimbic

dopaminergic systems and emotional states. Journal of Neuroscience Methods,

34(1–3), pp.135–142.

Besson, J.-M., Guilbaud, G. and Ollat, H., 1995. Forebrain Areas Involved in Pain

Processing. John Libbey Eurotext.

Bhagyashree, A., Manikkoth, S., Sequeira, M., Nayak, R. and Rao, S.N., 2017. Central

dopaminergic system plays a role in the analgesic action of paracetamol: Preclinical

evidence. Indian journal of pharmacology, 49(1), pp.21–25.

Binder, J.R., Frost, J.A., Hammeke, T.A., Cox, R.W., Rao, S.M. and Prieto, T., 1996.

Human Brain Language Areas Identified by Functional Magnetic Resonance

Imaging. Journal of Neuroscience, 17(1), pp.353-62.

Bingel, U., Schoell, E., Herken, W., Büchel, C. and May, A., 2007. Habituation to

painful stimulation involves the antinociceptive system. Pain, 131(1–2), pp.21–30.

Birnie, K.A., Caes, L., Wilson, A.C., Williams, S.E. and Chambers, C.T., 2014. A

Page 187: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

187

practical guide and perspectives on the use of experimental pain modalities with

children and adolescents. Pain management, 4(2), pp.97–111.

Bjørkedal, E. and Flaten, M.A., 2012. Expectations of increased and decreased pain

explain the effect of conditioned pain modulation in females. Journal of pain

research, 5, pp.289–300.

Blanchet, P.J. and Brefel-Courbon, C., 2018. Chronic pain and pain processing in

Parkinson’s disease. Progress in Neuro-Psychopharmacology and Biological

Psychiatry, 87, pp.200–206.

Blanke, O., Ortigue, S., Landis, T. and Seeck, M., 2002. Stimulating illusory own-body

perceptions. Nature, 419(6904), pp.269–270.

Bloem, B.R., Hausdorff, J.M., Visser, J.E. and Giladi, N., 2004. Falls and freezing of

gait in Parkinson’s disease: A review of two interconnected, episodic phenomena.

Movement Disorders, 19(8), pp.871–884.

Blyth, F.M., March, L.M., Brnabic, A.J., Jorm, L.R., Williamson, M. and Cousins, M.J.,

2001a. Chronic pain in Australia: a prevalence study. Pain, 89(2–3), pp.127–34.

Blyth, F.M., March, L.M., Brnabic, A.J.M., Jorm, L.R., Williamson, M. and Cousins,

M.J., 2001b. Chronic pain in Australia: a prevalence study. Pain, 89(2), pp.127–134.

Böcker, K.B.., Baas, J.M.., Kenemans, J.. and Verbaten, M.., 2001. Stimulus-

preceding negativity induced by fear: a manifestation of affective anticipation.

International Journal of Psychophysiology, 43(1), pp.77–90.

Borsook, D., Edwards, R., Elman, I., Becerra, L. and Levine, J., 2013. Pain and

analgesia: the value of salience circuits. Progress in neurobiology, 104, pp.93–105.

Borsook, D., Upadhyay, J., Chudler, E.H. and Becerra, L., 2010. A key role of the

basal ganglia in pain and analgesia - insights gained through human functional

imaging. Molecular Pain, 6(1), p.27.

Bowker, R.M., Westlund, K.N., Sullivan, M.C., Wilber, J.F. and Coulter, J.D., 1983.

Page 188: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

188

Descending serotonergic, peptidergic and cholinergic pathways from the raphe

nuclei: a multiple transmitter complex. Brain research, 288(1–2), pp.33–48.

Bowsher, D., 1976. Role of the reticular formation in responses to noxious

stimulation. Pain, 2(4), pp.361–378.

Braak, H., Ghebremedhin, E., Rüb, U., Bratzke, H. and Del Tredici, K., 2004. Stages in

the development of Parkinson’s disease-related pathology. Cell and tissue research,

318(1), pp.121–34.

Braak, H., Tredici, K. Del, Rüb, U., de Vos, R.A.., Jansen Steur, E.N.. and Braak, E.,

2003. Staging of brain pathology related to sporadic Parkinson’s disease.

Neurobiology of Aging, 24(2), pp.197–211.

Bradley, L.A., Mckendree-Smith, N.L., Alberts, K.R., Alarcón, G.S., Mountz, J.M. and

Deutsch, G., 2000. Use of Neuroimaging to Understand Abnormal Pain Sensitivity in

Fibromyalgia. Current Rheumatology Reports, 2, pp.141–148.

Braz, J.M., Nassar, M.A., Wood, J.N. and Basbaum, A.I., 2005. Parallel ‘pain’

pathways arise from subpopulations of primary afferent nociceptor. Neuron, 47(6),

pp.787–93.

Brefel-Courbon, C., Ory-Magne, F., Thalamas, C., Payoux, P. and Rascol, O., 2013.

Nociceptive brain activation in patients with neuropathic pain related to

Parkinson’s disease. Parkinsonism & Related Disorders, 19(5), pp.548–552.

Brefel-Courbon, C., Payoux, P., Thalamas, C., Ory, F., Quelven, I., Chollet, F.,

Montastruc, J.L. and Rascol, O., 2005. Effect of levodopa on pain threshold in

Parkinson’s disease: A clinical and positron emission tomography study. Movement

Disorders, 20(12), pp.1557–1563.

Breivik, H., Collett, B., Ventafridda, V., Cohen, R. and Gallacher, D., 2006. Survey of

chronic pain in Europe: prevalence, impact on daily life, and treatment. European

journal of pain (London, England), 10(4), pp.287–333.

Page 189: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

189

Broen, M.P.G., Narayen, N.E., Kuijf, M.L., Dissanayaka, N.N.W. and Leentjens,

A.F.G., 2016. Prevalence of anxiety in Parkinson’s disease: A systematic review and

meta-analysis. Movement Disorders, 31(8), pp.1125–1133.

Bromberg-Martin, E.S., Matsumoto, M. and Hikosaka, O., 2010. Dopamine in

Motivational Control: Rewarding, Aversive, and Alerting. Neuron, 68(5), pp.815–

834.

Brooks, D.J., Ibanez, V., Sawle, G. V., Playford, E.D., Quinn, N., Mathias, C.J., Lees,

A.J., Marsden, C.D., Bannister, R. and Frackowiak, R.S.J., 1992. Striatal D2 receptor

status in patients with Parkinson’s disease, striatonigral degeneration, and

progressive supranuclear palsy, measured with11C-raclopride and positron

emission tomography. Annals of Neurology, 31(2), pp.184–192.

Brown, C.A., El-Deredy, W. and Jones, A.K.P., 2014. When the brain expects pain:

common neural responses to pain anticipation are related to clinical pain and

distress in fibromyalgia and osteoarthritis. The European journal of neuroscience,

39(4), pp.663–72.

Brown, C.A. and Jones, A.K., 2008. A role for midcingulate cortex in the interruptive

effects of pain anticipation on attention. Clinical Neurophysiology, 119(10),

pp.2370–2379.

Brown, C.A. and Jones, A.K.P., 2010. Meditation experience predicts less negative

appraisal of pain: Electrophysiological evidence for the involvement of anticipatory

neural responses. Pain, 150(3), pp.428–438.

Brown, C.A. and Jones, A.K.P., 2012. Psychobiological Correlates of Improved

Mental Health in Patients With Musculoskeletal Pain After a Mindfulness-based

Pain Management Program. Clinical Journal of Pain, 29(3), pp.233-44.

Brown, C.A., Matthews, J., Fairclough, M., McMahon, A., Barnett, E., Al-Kaysi, A., El-

Deredy, W. and Jones, A.K.P., 2015. Striatal opioid receptor availability is related to

acute and chronic pain perception in arthritis: does opioid adaptation increase

Page 190: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

190

resilience to chronic pain? Pain, 156(11), pp.2267–75.

Brown, C.A., Seymour, B., Boyle, Y., El-Deredy, W. and Jones, A.K.P.P., 2008a.

Modulation of pain ratings by expectation and uncertainty: Behavioral

characteristics and anticipatory neural correlates. Pain, 135(3), pp.240–50.

Brown, C.A., Seymour, B., El-Deredy, W. and Jones, A.K.P., 2008b. Confidence in

beliefs about pain predicts expectancy effects on pain perception and anticipatory

processing in right anterior insula. Pain, 139(2), pp.324–32.

Brunia, C.H. and van Boxtel, G.J., 2001. Wait and see. International journal of

psychophysiology : official journal of the International Organization of

Psychophysiology, 43(1), pp.59–75.

Bucker, B. and Theeuwes, J., 2014. The effect of reward on orienting and

reorienting in exogenous cuing. Cognitive, Affective, & Behavioral Neuroscience,

14(2), pp.635–646.

Buckholtz, J.W., Treadway, M.T., Cowan, R.L., Woodward, N.D., Li, R., Ansari, M.S.,

Baldwin, R.M., Schwartzman, A.N., Shelby, E.S., Smith, C.E., Kessler, R.M. and Zald,

D.H., 2010. Dopaminergic Network Differences in Human Impulsivity. Science,

329(5991), pp.532–532.

Burgmer, M., Petzke, F., Giesecke, T., Gaubitz, M., Heuft, G. and Pfleiderer, B., 2011.

Cerebral Activation and Catastrophizing During Pain Anticipation in Patients With

Fibromyalgia. Psychosomatic Medicine, 73(9), pp.751–759.

Bushnell, M.C., Ceko, M. and Low, L.A., 2013. Cognitive and emotional control of

pain and its disruption in chronic pain. Nature reviews. Neuroscience, 14(7),

pp.502–11.

Bushnell, M.C., Duncan, G.H., Hofbauer, R.K., Ha, B., Chen, J.-I. and Carrier, B., 1999.

Pain perception: Is there a role for primary somatosensory cortex? Proceedings of

the National Academy of Sciences, 96(14), pp.7705–7709.

Page 191: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

191

Cannon, D.M., Ichise, M., Fromm, S.J., Nugent, A.C., Rollis, D., Gandhi, S.K., Klaver,

J.M., Charney, D.S., Manji, H.K. and Drevets, W.C., 2006. Serotonin Transporter

Binding in Bipolar Disorder Assessed using [11C]DASB and Positron Emission

Tomography. Biological Psychiatry, 60(3), pp.207–217.

Cannon, D.M., Ichise, M., Rollis, D., Klaver, J.M., Gandhi, S.K., Charney, D.S., Manji,

H.K. and Drevets, W.C., 2007. Elevated Serotonin Transporter Binding in Major

Depressive Disorder Assessed Using Positron Emission Tomography and [11C]DASB;

Comparison with Bipolar Disorder. Biological Psychiatry, 62(8), pp.870–877.

Canter, G.J., 1963. Speech Characteristics of Patients with Parkinson’s Disease: I.

Intensity, Pitch, and Duration. Journal of Speech and Hearing Disorders, 28(3),

pp.221–229.

Cantwell, M., Leong, L., Cano, A. and Moghaddam, M., 2013. Pain stoicism is related

to the expression of pain behaviors in women, but not men, during an acute pain

task. The Journal of Pain, 14(4), pp.S95.

Casey, K.L., 1980. Reticular formation and pain: toward a unifying concept.

Research publications - Association for Research in Nervous and Mental Disease, 58,

pp.93–105.

Chakour, M.C., Gibson, S.J., Bradbeer, M. and Helme, R.D., 1996. The effect of age

on Aδ- and C-fibre thermal pain perception. Pain, 64(1), pp.143–152.

Chao, L.L., Haxby, J. V. and Martin, A., 1999. Attribute-based neural substrates in

temporal cortex for perceiving and knowing about objects. Nature Neuroscience,

2(10), pp.913–919.

Chaudhuri, R. and Schapira, A.H., 2009. Non-motor symptoms of Parkinson’s

disease: dopaminergic pathophysiology and treatment. The Lancet Neurology, 8,

pp.464–474.

Christoph M. Michel, G.L.L.S.R.G. de P.T.L.M.S., 2004. 128-channel Eeg Source

Imaging in Epilepsy: Clinical Yield and Localization Precision. Journal of Clinical

Page 192: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

192

Neurophysiology, 21(2), pp.71–83.

Christopher, L., Marras, C., Duff-Canning, S., Koshimori, Y., Chen, R., Boileau, I.,

Segura, B., Monchi, O., Lang, A.E., Rusjan, P., Houle, S. and Strafella, A.P., 2014.

Combined insular and striatal dopamine dysfunction are associated with executive

deficits in Parkinson’s disease with mild cognitive impairment. Brain, 137(2),

pp.565–575.

Chudler, E.H., 1998. Response properties of neurons in the caudate–putamen and

globus pallidus to noxious and non-noxious thermal stimulation in anesthetized

rats. Brain Research, 812(1–2), pp.283–288.

Chudler, E.H., Sugiyama, K. and Dong, W.K., 1993. Nociceptive responses in the

neostriatum and globus pallidus of the anesthetized rat. J Neurophysiol, 69(6),

pp.1890–1903.

Cicchetti, D. and Cohen, D.J., 2006. DEVELOPMENTAL PSYCHOPATHOLOGY SECOND

EDITION Volume Three: Risk, Disorder, and Adaptation.

Clark, J.A., Brown, C.A., Jones, A.K.P. and El-Deredy, W., 2008a. Dissociating

nociceptive modulation by the duration of pain anticipation from unpredictability in

the timing of pain. Clinical Neurophysiology, 119(12), pp.2870–2878.

Clark, J.A., Brown, C.A., Jones, A.K.P. and El-Deredy, W., 2008b. Dissociating

nociceptive modulation by the duration of pain anticipation from unpredictability in

the timing of pain. Clinical Neurophysiology.

Cobacho, N., de la Calle, J.L. and Paíno, C.L., 2014. Dopaminergic modulation of

neuropathic pain: Analgesia in rats by a D2-type receptor agonist. Brain Research

Bulletin, 106, pp.62–71.

Conte, A., Khan, N., Defazio, G., Rothwell, J.C. and Berardelli, A., 2013.

Pathophysiology of somatosensory abnormalities in Parkinson disease. Nature

Reviews Neurology, 9(12), pp.687–697.

Page 193: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

193

Cools, R. and D’Esposito, M., 2011. Inverted-U–Shaped Dopamine Actions on

Human Working Memory and Cognitive Control. Biological Psychiatry, 69(12),

pp.e113–e125.

Cools, R., Frank, M.J., Gibbs, S.E., Miyakawa, A., Jagust, W. and D’Esposito, M.,

2009. Striatal dopamine predicts outcome-specific reversal learning and its

sensitivity to dopaminergic drug administration. The Journal of neuroscience : the

official journal of the Society for Neuroscience, 29(5), pp.1538–43.

Cooper, J.C. and Knutson, B., 2008. Valence and salience contribute to nucleus

accumbens activation. NeuroImage, 39(1), pp.538–547.

Corkin, S., Milner, B. and Rasmussen, T., 1970. Somatosensory Thresholds. Archives

of Neurology, 23(1), p.41.

La Corte, V., Sperduti, M., Malherbe, C., Vialatte, F., Lion, S., Gallarda, T.,

Oppenheim, C. and Piolino, P., 2016. Cognitive Decline and Reorganization of

Functional Connectivity in Healthy Aging: The Pivotal Role of the Salience Network

in the Prediction of Age and Cognitive Performances. Frontiers in aging

neuroscience, 8, p.204.

Costa, A., la Fougère, C., Pogarell, O., Möller, H.-J., Riedel, M. and Ettinger, U., 2013.

Impulsivity is related to striatal dopamine transporter availability in healthy males.

Psychiatry Research: Neuroimaging, 211(3), pp.251–256.

Cotzias, G.C., Papavasiliou, P.S., Fehling, C., Kaufman, B. and Mena, I., 1970.

Similarities between Neurologic Effects of L-Dopa and of Apomorphine. New

England Journal of Medicine, 282(1), pp.31–33.

Cousineau, D., 2005. Confidence intervals in within-subject designs: A simpler

solution to Loftus and Masson’s method. Tutorials in Quantitative Methods for

Psychology, .

Dalley, J.W. and Robbins, T.W., 2017. Fractionating impulsivity: neuropsychiatric

implications. Nature Reviews Neuroscience, 18(3), pp.158–171.

Page 194: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

194

Defazio, G., Berardelli, A., Fabbrini, G., Martino, D., Fincati, E., Fiaschi, A., Moretto,

G., Abbruzzese, G., Marchese, R., Bonuccelli, U., Del Dotto, P., Barone, P., De Vivo,

E., Albanese, A., Antonini, A., Canesi, M., Lopiano, L., Zibetti, M., Nappi, G.,

Martignoni, E., Lamberti, P. and Tinazzi, M., 2008. Pain as a nonmotor symptom of

Parkinson disease: evidence from a case-control study. Archives of neurology, 65(9),

pp.1191–4.

Delorme, A. and Makeig, S., 2004. EEGLAB: an open source toolbox for analysis of

single-trial EEG dynamics including independent component analysis. Journal of

Neuroscience Methods, 134(1), pp.9–21.

Derbyshire, S.W.G., Jones, A.K.P., Gyulai, F., Clark, S., Townsend, D. and Firestone,

L.L., 1997. Pain processing during three levels of noxious stimulation produces

differential patterns of central activity, 73(3), pp.431-45.

Derbyshire, S.W.G., Nichols, T.E., Firestone, L., Townsend, D.W. and Jones, A.K.P.,

2002. Gender differences in patterns of cerebral activation during equal experience

of painful laser stimulation. The Journal of Pain, 3(5), pp.401–411.

Dettmers, C., Adler, T., Rzanny, R., van Schayck, R., Gaser, C., Weiss, T., Miltner,

W.H., Brückner, L. and Weiller, C., 2001. Increased excitability in the primary motor

cortex and supplementary motor area in patients with phantom limb pain after

upper limb amputation. Neuroscience letters, 307(2), pp.109–12.

Dick, B., Eccleston, C. and Crombez, G., 2002. Attentional functioning in

fibromyalgia, rheumatoid arthritis, and musculoskeletal pain patients. Arthritis and

rheumatism, 47(6), pp.639–44.

Dieb, W., Ouachikh, O., Durif, F. and Hafidi, A., 2014. Lesion of the dopaminergic

nigrostriatal pathway induces trigeminal dynamic mechanical allodynia. Brain and

behavior, 4(3), pp.368–80.

Dietz, V., Berger, W. and Horstmann, G.A., 1988. Posture in Parkinson’s disease:

Impairment of reflexes and programming. Annals of Neurology, 24(5), pp.660–669.

Page 195: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

195

Djaldetti, R., Shifrin, A., Rogowski, Z., Sprecher, E., Melamed, E. and Yarnitsky, D.,

2004a. Quantitative measurement of pain sensation in patients with Parkinson

disease. Neurology, 62(12), pp.2171–5.

Djaldetti, R., Shifrin, A., Rogowski, Z., Sprecher, E., Melamed, E. and Yarnitsky, D.,

2004b. Quantitative measurement of pain sensation in patients with Parkinson

disease. Neurology, 62(12), pp.2171–5.

Dobrila-Dintinjana, R. and Nacinović-Duletić, A., 2011. Placebo in the treatment of

pain. Collegium antropologicum, 35 Suppl 2, pp.319–23.

Donaldson, L.F. and Lumb, B.M., 2017. Top-down control of pain. The Journal of

physiology, 595(13), pp.4139–4140.

Dorsey, E.R., Constantinescu, R., Thompson, J.P., Biglan, K.M., Holloway, R.G.,

Kieburtz, K., Marshall, F.J., Ravina, B.M., Schifitto, G., Siderowf, A. and Tanner, C.M.,

2007. Projected number of people with Parkinson disease in the most populous

nations, 2005 through 2030. Neurology, 68(5), pp.384–6.

Dreher, J.-C., Meyer-Lindenberg, A., Kohn, P. and Berman, K.F., 2008. Age-related

changes in midbrain dopaminergic regulation of the human reward system.

Proceedings of the National Academy of Sciences of the United States of America,

105(39), pp.15106–11.

Drotár, P., Mekyska, J., Rektorová, I., Masarová, L., Smékal, Z. and Faundez-Zanuy,

M., 2016. Evaluation of handwriting kinematics and pressure for differential

diagnosis of Parkinson’s disease. Artificial Intelligence in Medicine, 67, pp.39–46.

Duffy, F.H., Albert, M.S., McAnulty, G. and Garvey, A.J., 1984. Age-related

differences in brain electrical activity of healthy subjects. Annals of Neurology,

16(4), pp.430–438.

Dustman, R.E., Emmerson, R.Y., Ruhling, R.O., Shearer, D.E., Steinhaus, L.A.,

Johnson, S.C., Bonekat, H.W. and Shigeoka, J.W., 1990. Age and fitness effects on

EEG, ERPs, visual sensitivity, and cognition. Neurobiology of Aging, 11(3), pp.193–

Page 196: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

196

200.

Eckert, M.A., Cute, S.L., Vaden, K.I., Kuchinsky, S.E., Dubno, J.R. and Dubno, J.R.,

2012. Auditory cortex signs of age-related hearing loss. Journal of the Association

for Research in Otolaryngology : JARO, 13(5), pp.703–13.

Efran, J.S., Chorney, R.L., Ascher, L.M. and Lukens, M.D., 1989. Coping styles,

paradox, and the cold pressor task. Journal of Behavioral Medicine, 12(1), pp.91–

103.

Ehrt, U., Larsen, J.P. and Aarsland, D., 2009. Pain and its relationship to depression

in Parkinson disease. The American journal of geriatric psychiatry : official journal of

the American Association for Geriatric Psychiatry, 17(4), pp.269–75.

Elliot, A.J. and Covington, M. V, 2001. Approach and Avoidance Motivation.

Educational Psychology Review, 13(2), pp.73-92.

Elsworth, J.D., Lawrence, M.S., Roth, R.H., Taylor, J.R., Mailman, R.B., Nichols, D.E.,

Lewis, M.H. and Redmond, D.E., 1991. D1 and D2 dopamine receptors

independently regulate spontaneous blink rate in the vervet monkey. Journal of

Pharmacology and Experimental Therapeutics, 259(2), pp.595-600.

Ertas, M., Sagduyu, A., Arac, N., Uludag, B. and Ertekin, C., 1998. Use of levodopa to

relieve pain from painful symmetrical diabetic polyneuropathy. Pain, 75(2–3),

pp.257–9.

Esposito, E., Romandini, S., Merlo-Pich, E., Mennini, T. and Samanin, R., 1986.

Evidence of the involvement of dopamine in the analgesic effect of nefopam.

European journal of pharmacology, 128(3), pp.157–64.

Fairhurst, M., Wiech, K., Dunckley, P. and Tracey, I., 2007. Anticipatory brainstem

activity predicts neural processing of pain in humans. Pain, 128, pp.101–110.

Faludi, B., Janszky, J., Komoly, S. and Kovács, N., 2015. Alvászavarok Parkinson-

kórban: megjelenés, kivizsgálás, terápiás lehetőségek. Orvosi Hetilap, 156(27),

Page 197: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

197

pp.1091–1099.

Farrell, M.J., 2012. Age-Related Changes in the Structure and Function of Brain

Regions Involved in Pain Processing. Pain Medicine, 13(suppl 2), pp.S37–S43.

Favre, J., Burchiel, K.J., Taha, J.M. and Hammerstad, J., 2000. Outcome of unilateral

and bilateral pallidotomy for Parkinson’s disease: patient assessment.

Neurosurgery, 46(2), pp.344-53;

Fenelon, G., Mahieux, F., Huon, R. and Ziégler, M., 2000. Hallucinations in

Parkinson’s disease: Prevalence, phenomenology and risk factors. Brain, 123(4),

pp.733–745.

Finke, K., Dodds, C.M., Bublak, P., Regenthal, R., Baumann, F., Manly, T. and Müller,

U., 2010. Effects of modafinil and methylphenidate on visual attention capacity: a

TVA-based study. Psychopharmacology, 210(3), pp.317–329.

Fiorillo, C.D., 2017. Neuroscience: Rationality, uncertainty, dopamine. Nature

Human Behaviour, 1(8), p.0158.

Fiorillo, C.D., Tobler, P.N. and Schultz, W., 2003. Discrete Coding of Reward

Probablilty and Uncertainty by Dopamine Neurons. Science, 299, pp.1898–1902.

FitzGerald, T.H.B., Dolan, R.J. and Friston, K., 2015. Dopamine, reward learning, and

active inference. Frontiers in computational neuroscience, 9, p.136.

Floresco, S.B., 2013. Prefrontal dopamine and behavioral flexibility: shifting from an

“inverted-U” toward a family of functions. Frontiers in Neuroscience, 7, p.62.

Flynn, F.G., 1999. Anatomy of the insula functional and clinical correlates.

Aphasiology, 13(1), pp.55–78.

Ford, B., 2010. Pain in Parkinson’s disease. Movement disorders : official journal of

the Movement Disorder Society, 25 Suppl 1(S1), pp.S98-103.

Ford, B., Louis, E.D., Greene, P. and Fahn, S., 1996. Oral and genital pain syndromes

Page 198: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

198

in Parkinson’s disease. Movement disorders : official journal of the Movement

Disorder Society, 11(4), pp.421–6.

Ford, C.P., 2014. The role of D2-autoreceptors in regulating dopamine neuron

activity and transmission. Neuroscience, 282, pp.13–22.

Forkmann, K., Grashorn, W., Schmidt, K., Undt, O.F., Buhmann, C. and Bingel, U.,

2017. Altered neural responses to heat pain in drug-naive patients with Parkinson

disease. Pain, 158, pp.1408–1416.

Frank, M.J. and O’Reilly, R.C., 2006. A mechanistic account of striatal dopamine

function in human cognition: Psychopharmacological studies with cabergoline and

haloperidol. Behavioral Neuroscience, 120(3), pp.497–517.

Friedman, J. and Friedman, H., 1993. Fatigue in Parkinson’s disease. Neurology,

43(10), pp.2016–8.

Friedman, J.H., Brown, R.G., Comella, C., Garber, C.E., Krupp, L.B., Lou, J.-S., Marsh,

L., Nail, L., Shulman, L. and Taylor, C.B., 2007. Fatigue in Parkinson’s disease: A

review. Movement Disorders, 22(3), pp.297–308.

Friston, K., Schwartenbeck, P., FitzGerald, T., Moutoussis, M., Behrens, T. and

Dolan, R.J., 2014. The anatomy of choice: dopamine and decision-making.

Philosophical Transactions of the Royal Society B: Biological Sciences, 369(1655),

pp.20130481–20130481.

Friston, K.J., Shiner, T., FitzGerald, T., Galea, J.M., Adams, R., Brown, H., Dolan, R.J.,

Moran, R., Stephan, K.E. and Bestmann, S., 2012. Dopamine, Affordance and Active

Inference. PLoS Computational Biology, 8(1), p.e1002327.

Gai, W.P., Blessing, W.W. and Blumbergs, P.C., 1995. Ubiquitin-positive

degenerating neurites in the brainstem in Parkinson’s disease. Brain, 118(6),

pp.1447–1459.

Gebhart, G.F., 2004. Descending modulation of pain. Neuroscience and

Page 199: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

199

biobehavioral reviews, 27(8), pp.729–37.

Geisser, M.E., Robinson, M.E., Keefe, F.J. and Weiner, M.L., 1994. Catastrophizing,

depression and the sensory, affective and evaluative aspects of chronic pain. Pain,

59(1), pp.79–83.

Geisser, M.E., Robinson, M.E. and Pickren, W.E., 1992. Differences in cognitive

coping strategies among pain-sensitive and pain-tolerant individuals on the cold-

pressor test. Behavior Therapy, 23(1), pp.31–41.

Gentry, R.N., Schuweiler, D.R. and Roesch, M.R., 2018. Dopamine signals related to

appetitive and aversive events in paradigms that manipulate reward and

avoidability. Brain Research.

Geraghty, J.J., Jankovic, J. and Zetusky, W.J., 1985. Association between essential

tremor and Parkinson’s disease. Annals of Neurology, 17(4), pp.329–333.

Gerdelat-Mas, A., Simonetta-Moreau, M., Thalamas, C., Ory-Magne, F., Slaoui, T.,

Rascol, O. and Brefel-Courbon, C., 2007. Levodopa raises objective pain threshold in

Parkinson’s disease: a RIII reflex study. Journal of neurology, neurosurgery, and

psychiatry, 78(10), pp.1140–2.

Gerlach, M., Double, K., Arzberger, T., Leblhuber, F., Tatschner, T. and Riederer, P.,

2003. Dopamine receptor agonists in current clinical use: comparative dopamine

receptor binding profiles defined in the human striatum. Journal of Neural

Transmission, 110(10), pp.1119–1127.

Gibson, C.D., Karmally, W., McMahon, D.J., Wardlaw, S.L. and Korner, J., 2012.

Randomized pilot study of cabergoline, a dopamine receptor agonist: effects on

body weight and glucose tolerance in obese adults. Diabetes, obesity & metabolism,

14(4), pp.335–40.

Gibson, S.J. and Farrell, M., 2004. A review of age differences in the

neurophysiology of nociception and the perceptual experience of pain. Clinical

Journal of Pain, 20(4), pp.227–239.

Page 200: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

200

Gibson, S.J., Voukelatos, X., Ames, D., Flicker, L. and Helme, R.D., 2001. An

Examination of Pain Perception and Cerebral Event-Related Potentials following

Carbon Dioxide Laser Stimulation in Patients with Alzheimer’s Disease and Age-

Matched Control Volunteers. Pain Research and Management, 6(3), pp.126–132.

Giesecke, T., Gracely, R.H., Williams, D.A., Geisser, M.E., Petzke, F.W. and Clauw,

D.J., 2005. The relationship between depression, clinical pain, and experimental

pain in a chronic pain cohort. Arthritis & Rheumatism, 52(5), pp.1577–1584.

Goetz, C.G., Tanner, C.M., Levy, M., Wilson, R.S. and Garron, D.C., 1986. Pain in

Parkinson’s disease. Movement disorders : official journal of the Movement Disorder

Society, 1(1), pp.45–9.

Goetz, C.G., Tilley, B.C., Shaftman, S.R., Stebbins, G.T., Fahn, S., Martinez-Martin, P.,

Poewe, W., Sampaio, C., Stern, M.B., Dodel, R., Dubois, B., Holloway, R., Jankovic, J.,

Kulisevsky, J., Lang, A.E., Lees, A., Leurgans, S., LeWitt, P.A., Nyenhuis, D., Olanow,

C.W., Rascol, O., Schrag, A., Teresi, J.A., van Hilten, J.J., LaPelle, N. and Movement

Disorder Society UPDRS Revision Task Force, 2008. Movement Disorder Society-

sponsored revision of the Unified Parkinson’s Disease Rating Scale (MDS-UPDRS):

Scale presentation and clinimetric testing results. Movement Disorders, 23(15),

pp.2129–2170.

Goldsmith, S.. and Joyce, J.., 1996. Dopamine D2 receptors are organized in bands

in normal human temporal cortex. Neuroscience, 74(2), pp.435–451.

Gomperts, S.N., Marquie, M., Locascio, J.J., Bayer, S., Johnson, K.A. and Growdon,

J.H., 2016. PET Radioligands Reveal the Basis of Dementia in Parkinson’s Disease

and Dementia with Lewy Bodies. Neurodegenerative Diseases, 16(1–2), pp.118–

124.

González-Roldán, A.M., Bomba, I.C., Diesch, E., Montoya, P., Flor, H. and Kamping,

S., 2016. Controllability and hippocampal activation during pain expectation in

fibromyalgia syndrome. Biological Psychology, 121, pp.39–48.

Page 201: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

201

Goto, Y., Otani, S. and Grace, A.A., 2007. The Yin and Yang of dopamine release: a

new perspective. Neuropharmacology, 53(5), pp.583–587.

Gracely, R.H., Geisser, M.E., Giesecke, T., Grant, M.A.B., Petzke, F., Williams, D.A.

and Clauw, D.J., 2004. Pain catastrophizing and neural responses to pain among

persons with fibromyalgia. Brain : a journal of neurology, 127(Pt 4), pp.835–43.

Gracely, R.H., Petzke, F., Wolf, J.M. and Clauw, D.J., 2002. Functional magnetic

resonance imaging evidence of augmented pain processing in fibromyalgia. Arthritis

& Rheumatism, 46(5), pp.1333–1343.

Grashorn, W., Sprenger, C., Forkmann, K., Wrobel, N. and Bingel, U., 2013. Age-

dependent decline of endogenous pain control: exploring the effect of expectation

and depression. PloS one, 8(9), p.e75629.

Grech, R., Cassar, T., Muscat, J., Camilleri, K.P., Fabri, S.G., Zervakis, M.,

Xanthopoulos, P., Sakkalis, V. and Vanrumste, B., 2008. Review on solving the

inverse problem in EEG source analysis. Journal of neuroengineering and

rehabilitation, 5, p.25.

Guymer, E.K. and Littlejohn, G.O., 2007. Fibromyalgia: top down or bottom up?

APLAR Journal of Rheumatology, 10(3), pp.174–177.

Haddad, M., Pud, D., Treister, R., Suzan, E. and Eisenberg, E., 2018. The effects of a

dopamine agonist (apomorphine) on experimental and spontaneous pain in

patients with chronic radicular pain: A randomized, double-blind, placebo-

controlled, cross-over study. PLOS ONE, 13(4), p.e0195287.

Hagelberg, N., Forssell, H., Aalto, S., Rinne, J.O., Scheinin, H., Taiminen, T., Någren,

K., Eskola, O. and Jääskeläinen, S.K., 2003a. Altered dopamine D2 receptor binding

in atypical facial pain. Pain, 106(1–2), pp.43–8.

Hagelberg, N., Forssell, H., Rinne, J.O., Scheinin, H., Taiminen, T., Aalto, S.,

Luutonen, S., Någren, K. and Jääskeläinen, S., 2003b. Striatal dopamine D1 and D2

receptors in burning mouth syndrome. Pain, 101(1–2), pp.149–54.

Page 202: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

202

Hagelberg, N., Jääskeläinen, S.K., Martikainen, I.K., Mansikka, H., Forssell, H.,

Scheinin, H., Hietala, J. and Pertovaara, A., 2004. Striatal dopamine D2 receptors in

modulation of pain in humans: a review. European journal of pharmacology, 500(1–

3), pp.187–92.

Hagelberg, N., Martikainen, I.K., Mansikka, H., Hinkka, S., Någren, K., Hietala, J.,

Scheinin, H. and Pertovaara, A., 2002. Dopamine D2 receptor binding in the human

brain is associated with the response to painful stimulation and pain modulatory

capacity. Pain, 99(1–2), pp.273–9.

Hall, H., Sedvall, G., Magnusson, O., Kopp, J., Halldin, C. and Farde, L., 1994.

Distribution of D1- and D2-Dopamine Receptors, and Dopamine and Its Metabolites

in the Human Brain. Neuropsychopharmacology, 11(4), pp.245–256.

Hallez, H., Vanrumste, B., Grech, R., Muscat, J., De Clercq, W., Vergult, A., D’Asseler,

Y., Camilleri, K.P., Fabri, S.G., Van Huffel, S. and Lemahieu, I., 2007. Review on

solving the forward problem in EEG source analysis. Journal of NeuroEngineering

and Rehabilitation, 4(1), p.46.

Halliday, G.M., 2009. Thalamic changes in Parkinson’s disease. Parkinsonism &

Related Disorders, 15, pp.S152–S155.

Halliday, G.M., Blumbergs, P.C., Cotton, R.G., Blessing, W.W. and Geffen, L.B., 1990.

Loss of brainstem serotonin- and substance P-containing neurons in Parkinson’s

disease. Brain research, 510(1), pp.104–7.

Hanagasi, H.A., Akat, S., Gurvit, H., Yazici, J. and Emre, M., 2011. Pain is common in

Parkinson’s disease. Clinical neurology and neurosurgery, 113(1), pp.11–3.

Harrington, D.L., Haaland, K.Y., Yeo, R.A. and Marder, E., 1990. Procedural memory

in Parkinson’s disease: Impaired motor but not visuoperceptual learning. Journal of

Clinical and Experimental Neuropsychology, 12(2), pp.323–339.

Hassan, M.N. and Thakar, J.H., 1988. Dopamine receptors in Parkinson’s disease.

Progress in neuro-psychopharmacology & biological psychiatry, 12(2–3), pp.173–82.

Page 203: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

203

Hauck, M., Domnick, C., Lorenz, J., Gerloff, C. and Engel, A.K., 2015a. Top-down and

bottom-up modulation of pain-induced oscillations. Frontiers in human

neuroscience, 9, p.375.

Hauck, M., Domnick, C., Lorenz, J., Gerloff, C. and Engel, A.K., 2015b. Top-down and

bottom-up modulation of pain-induced oscillations. Frontiers in Human

Neuroscience, 9, p.375.

Häuser, W., 2016. Fibromyalgia syndrome: Basic knowledge, diagnosis and

treatment. Medizinische Monatsschrift fur Pharmazeuten, 39(12), pp.504–11.

Hayden, B.Y., Nair, A.C., McCoy, A.N. and Platt, M.L., 2008. Posterior Cingulate

Cortex Mediates Outcome-Contingent Allocation of Behavior. Neuron, 60(1), pp.19–

25.

van Hecke, O., Torrance, N. and Smith, B.H., 2013. Chronic pain epidemiology and

its clinical relevance. British Journal of Anaesthesia, 111(1), pp.13–18.

Herath, P., Kinomura, S. and Roland, P.E., 2001. Visual recognition: Evidence for two

distinctive mechanisms from a PET study. Human Brain Mapping, 12(2), pp.110–

119.

Hirata, A., Aguilar, J. and Castro-Alamancos, M.A., 2006. Noradrenergic activation

amplifies bottom-up and top-down signal-to-noise ratios in sensory thalamus. The

Journal of neuroscience : the official journal of the Society for Neuroscience, 26(16),

pp.4426–36.

Hird, E.J., Jones, A.K.P., Talmi, D. and El-Deredy, W., 2018. A comparison between

the neural correlates of laser and electric pain stimulation and their modulation by

expectation. Journal of Neuroscience Methods, 293, pp.117–127.

Ho, A.K., Iansek, R., Marigliani, C., Bradshaw, J.L. and Gates, S., 1999. Speech

Impairment in a Large Sample of Patients with Parkinson’s Disease. Behavioural

Neurology, 11(3), pp.131–137.

Page 204: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

204

van Holstein, M., Aarts, E., van der Schaaf, M.E., Geurts, D.E.M., Verkes, R.J., Franke,

B., van Schouwenburg, M.R. and Cools, R., 2011. Human cognitive flexibility

depends on dopamine D2 receptor signaling. Psychopharmacology, 218(3), pp.567–

578.

Hormigo, S., Vega-Flores, G. and Castro-Alamancos, M.A., 2016. Basal Ganglia

Output Controls Active Avoidance Behavior. The Journal of neuroscience : the

official journal of the Society for Neuroscience, 36(40), pp.10274–10284.

Hornung, J.-P., 2003. The human raphe nuclei and the serotonergic system. Journal

of Chemical Neuroanatomy, 26(4), pp.331–343.

Hornykiewicz, O., 1981. Brain neurotransmitter changes in Parkinson’s disease.

Movement Disorders, pp.41–58.

Horvitz, J.C., 2002. Dopamine gating of glutamatergic sensorimotor and incentive

motivational input signals to the striatum. Behavioural Brain Research, 137(1–2),

pp.65–74.

Howes, O.D. and Nour, M.M., 2016. Dopamine and the aberrant salience hypothesis

of schizophrenia. World psychiatry : official journal of the World Psychiatric

Association (WPA), 15(1), pp.3–4.

Hsieh, J.-C., Belfrage, M., Stone-Elander, S., Hansson, P. and Ingvar, M., 1995.

Central representation of chronic ongoing neuropathic pain studied by positron

emission tomography. PAIN®, 63(2), pp.225–236.

Hu, X., Song, X., Yuan, Y., Li, E., Liu, J., Liu, W. and Liu, Y., 2015. Abnormal functional

connectivity of the amygdala is associated with depression in Parkinson’s disease.

Movement Disorders, 30(2), pp.238–244.

Hubbard, C.S., Khan, S.A., Keaser, M.L., Mathur, V.A., Goyal, M. and Seminowicz,

D.A., 2014. Altered Brain Structure and Function Correlate with Disease Severity

and Pain Catastrophizing in Migraine Patients. eNeuro, 1(1), p.e20.14.

Page 205: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

205

Huskisson, E.C. and Hart, F.D., 1972. Pain threshold and arthritis. British medical

journal, 4(5834), pp.193–5.

Imamura, M., Chen, J., Matsubayashi, S.R., Targino, R.A., Alfieri, F.M., Bueno, D.K.

and Hsing, W.T., 2013. Changes in pressure pain threshold in patients with chronic

nonspecific low back pain. Spine, 38(24), pp.2098–107.

Inzelberg, R., Kipervasser, S. and Korczyn, A.D., 1998. Auditory hallucinations in

Parkinson’s disease. Journal of neurology, neurosurgery, and psychiatry, 64(4),

pp.533–5.

Isbister, G.K., Balit, C.R., Macleod, D. and Duffull, S.B., 2010. Amisulpride Overdose

Is Frequently Associated With QT Prolongation and Torsades de Pointes. Journal of

Clinical Psychopharmacology, 30(4), pp.391–395.

Isbister, G.K. and Page, C.B., 2013. Drug induced QT prolongation: the

measurement and assessment of the QT interval in clinical practice. British journal

of clinical pharmacology, 76(1), pp.48–57.

Ishai, A., Ungerleider, L.G., Martin, A., Schouten, J.L. and Haxby, J. V, 1999.

Distributed representation of objects in the human ventral visual pathway.

Proceedings of the National Academy of Sciences of the United States of America,

96(16), pp.9379–84.

Jääskeläinen, S.K., 2012. Pathophysiology of primary burning mouth syndrome.

Clinical neurophysiology : official journal of the International Federation of Clinical

Neurophysiology, 123(1), pp.71–7.

Jääskeläinen, S.K., Rinne, J.O., Forssell, H., Tenovuo, O., Kaasinen, V., Sonninen, P.

and Bergman, J., 2001. Role of the dopaminergic system in chronic pain -- a

fluorodopa-PET study. Pain, 90(3), pp.257–60.

Jankovic, J., Schwartz, K.S. and Ondo, W., 1999. Re-emergent tremor of Parkinson’s

disease. Journal of neurology, neurosurgery, and psychiatry, 67(5), pp.646–50.

Page 206: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

206

Jarcho, J.M., Mayer, E.A., Jiang, Z.K., Feier, N.A. and London, E.D., 2012. Pain,

affective symptoms, and cognitive deficits in patients with cerebral dopamine

dysfunction. Pain, 153(4), pp.744–54.

Jay, T.M., 2003. Dopamine: a potential substrate for synaptic plasticity and memory

mechanisms. Progress in Neurobiology, 69(6), pp.375–390.

Jensen, J., McIntosh, A.R., Crawley, A.P., Mikulis, D.J., Remington, G. and Kapur, S.,

2003. Direct activation of the ventral striatum in anticipation of aversive stimuli.

Neuron, 40(6), pp.1251–7.

Jensen, K.B., Kosek, E., Petzke, F., Carville, S., Fransson, P., Marcus, H., Williams,

S.C.R., Choy, E., Giesecke, T., Mainguy, Y., Gracely, R. and Ingvar, M., 2009. Evidence

of dysfunctional pain inhibition in Fibromyalgia reflected in rACC during provoked

pain. Pain, 144(1), pp.95–100.

Johannes, C.B., Le, T.K., Zhou, X., Johnston, J.A. and Dworkin, R.H., 2010. The

prevalence of chronic pain in United States adults: results of an Internet-based

survey. The journal of pain : official journal of the American Pain Society, 11(11),

pp.1230–9.

Jones, A., Brown, C. and El-Deredy, W., 2013. Chapter 5 – How does EEG Contribute

to Our Understanding of the Placebo Response?: Insights from the Perspective of

Bayesian Inference. In: Placebo and Pain. pp.37–43.

Jones, A.K.P., Huneke, N.T.M., Lloyd, D.M., Brown, C. a and Watson, A., 2012. Role

of functional brain imaging in understanding rheumatic pain. Current rheumatology

reports, 14(6), pp.557–67.

Joyce, J.N., Myers, A.J. and Gurevich, E., 1998. Dopamine D2 receptor bands in

normal human temporal cortex are absent in Alzheimer’s disease. Brain Research,

784(1–2), pp.7–17.

Juckel, G., Karch, S., Kawohl, W., Kirsch, V., Jäger, L., Leicht, G., Lutz, J., Stammel, A.,

Pogarell, O., Ertl, M., Reiser, M., Hegerl, U., Möller, H.J. and Mulert, C., 2012. Age

Page 207: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

207

effects on the P300 potential and the corresponding fMRI BOLD-signal.

NeuroImage, 60(4), pp.2027–2034.

Juruena, M.F., de Sena, E.P. and de Oliveira, I.R., 2010. Safety and tolerability of

antipsychotics: focus on amisulpride. Drug, healthcare and patient safety, 2,

pp.205–11.

Kabat-Zinn, J., 1982. An outpatient program in behavioral medicine for chronic pain

patients based on the practice of mindfulness meditation: Theoretical

considerations and preliminary results. General Hospital Psychiatry, 4(1), pp.33–47.

Kabat-Zinn, J., Lipworth, L. and Burney, R., 1985. The clinical use of mindfulness

meditation for the self-regulation of chronic pain. Journal of Behavioral Medicine,

8(2), pp.163–190.

Kaplan, K.H., Goldenberg, D.L. and Galvin-Nadeau, M., 1993. The impact of a

meditation-based stress reduction program on fibromyalgia. General Hospital

Psychiatry, 15(5), pp.284–289.

Kapur, S., Mizrahi, R. and Li, M., 2005. From dopamine to salience to psychosis—

linking biology, pharmacology and phenomenology of psychosis. Schizophrenia

Research, 79(1), pp.59–68.

KARSON, C.N., 1983. SPONTANEOUS EYE-BLINK RATES AND DOPAMINERGIC

SYSTEMS. Brain, 106(3), pp.643–653.

Karson, C.N., Burns, R.S., LeWitt, P.A., Foster, N.L. and Newman, R.P., 1984. Blink

rates and disorders of movement. Neurology, 34(5), pp.677–8.

Kernbaum, S. and Hauchecorne, J., 1981. Administration of Levodopa for Relief of

Herpes Zoster Pain. JAMA: The Journal of the American Medical Association, 246(2),

p.132.

Kim, J.-H., Choi, S.-H., Jang, J.H., Lee, D.-H., Lee, K.-J., Lee, W.J., Moon, J.Y., Kim, Y.C.

and Kang, D.-H., 2017. Impaired insula functional connectivity associated with

Page 208: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

208

persistent pain perception in patients with complex regional pain syndrome. PloS

one, 12(7), p.e0180479.

Kirkby, R.J. and Kimble, D.P., 1968. Avoidance and escape behavior following striatal

lesions in the rat. Experimental Neurology, 20(2), pp.215–227.

Kish, S.J., 2003. Biochemistry of Parkinson’s disease: is a brain serotonergic

deficiency a characteristic of idiopathic Parkinson’s disease? Advances in neurology,

91, pp.39–49.

Klanker, M., Feenstra, M. and Denys, D., 2013. Dopaminergic control of cognitive

flexibility in humans and animals. Frontiers in neuroscience, 7, p.201.

Kleven, M.S. and Koek, W., 1996. Differential effects of direct and indirect

dopamine agonists on eye blink rate in cynomolgus monkeys. The Journal of

pharmacology and experimental therapeutics, 279(3), pp.1211–9.

Von Korff, M., Ormel, J., Keefe, F.J. and Dworkin, S.F., 1992. Grading the severity of

chronic pain. Pain, 50(2), pp.133–149.

Korponay, C., Dentico, D., Kral, T., Ly, M., Kruis, A., Goldman, R., Lutz, A. and

Davidson, R.J., 2017. Neurobiological correlates of impulsivity in healthy adults:

Lower prefrontal gray matter volume and spontaneous eye-blink rate but greater

resting-state functional connectivity in basal ganglia-thalamo-cortical circuitry.

NeuroImage, 157, pp.288–296.

Koyama, T., Kato, K. and Mikami, A., 2000. During pain-avoidance neurons activated

in the macaque anterior cingulate and caudate. Neuroscience Letters, 283(1),

pp.17–20.

Koyama, T., McHaffie, J.G., Laurienti, P.J. and Coghill, R.C., 2005. The subjective

experience of pain: where expectations become reality. Proceedings of the National

Academy of Sciences of the United States of America, 102(36), pp.12950–5.

Kwon, M., Altin, M., Duenas, H. and Alev, L., 2013. The Role of Descending

Page 209: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

209

Inhibitory Pathways on Chronic Pain Modulation and Clinical Implications.

Lanciego, J.L., Luquin, N. and Obeso, J.A., 2012. Functional neuroanatomy of the

basal ganglia. Cold Spring Harbor perspectives in medicine, 2(12), p.a009621.

Langston, J.W., 2006. The parkinson’s complex: Parkinsonism is just the tip of the

iceberg. Annals of Neurology, 59(4), pp.591–596.

Lantz, G., Grave de Peralta, R., Spinelli, L., Seeck, M. and Michel, C.., 2003. Epileptic

source localization with high density EEG: how many electrodes are needed?

Clinical Neurophysiology, 114(1), pp.63–69.

Lariviere, M., Goffaux, P., Marchand, S. and Julien, N., 2007. Changes in Pain

Perception and Descending Inhibitory Controls Start at Middle Age in Healthy

Adults. The Clinical Journal of Pain, 23(6), pp.506–510.

Latremoliere, A. and Woolf, C.J., 2009. Central Sensitization: A Generator of Pain

Hypersensitivity by Central Neural Plasticity. Journal of Pain, 10(9), pp.895-926.

Lawson, K., 2016. Potential drug therapies for the treatment of fibromyalgia. Expert

Opinion on Investigational Drugs, 25(9), pp.1071–1081.

Lee, M.A., Walker, R.W., Hildreth, T.J. and Prentice, W.M., 2006. A survey of pain in

idiopathic Parkinson’s disease. Journal of pain and symptom management, 32(5),

pp.462–9.

Legrain, V., Iannetti, G.D., Plaghki, L. and Mouraux, A., 2011. The pain matrix

reloaded: A salience detection system for the body. Progress in Neurobiology, 93(1),

pp.111–124.

Lehrner, J., Kogler, S., Lamm, C., Moser, D., Klug, S., Pusswald, G., Dal-Bianco, P.,

Pirker, W. and Auff, E., 2015. Awareness of memory deficits in subjective cognitive

decline, mild cognitive impairment, Alzheimer’s disease and Parkinson’s disease.

International Psychogeriatrics, 27(03), pp.357–366.

Leknes, S. and Tracey, I., 2008. A common neurobiology for pain and pleasure.

Page 210: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

210

Nature reviews. Neuroscience, 9(4), pp.314–20.

Levine, F. and De Simone, L., 1991. The effects of experimenter gender on pain

report in male and female subjects. Pain, 44, pp.69–72.

Levy, F., 1991. The Dopamine Theory of Attention Deficit Hyperactivity Disorder

(ADHD). Australian & New Zealand Journal of Psychiatry, 25(2), pp.277–283.

Levy, F., 2009. Dopamine vs Noradrenaline: Inverted-U Effects and ADHD Theories.

Australian & New Zealand Journal of Psychiatry, 43(2), pp.101–108.

Lewis, G.N. and Byblow, W.D., 2002. Altered sensorimotor integration in

Parkinson’s disease. Brain : a journal of neurology, 125(Pt 9), pp.2089–99.

Li, S.-Ch. and Backman, L., 2010. Dopaminergic modulation of cognition across the

life span. Neuroscience & Biobehavioral Reviews, 34, pp.625–630.

Lima, J.C., Oliveira, L.M., Botelho, M.T., Moreira, T.S. and Takakura, A.C., 2018. The

involvement of the pathway connecting the substantia nigra, the periaqueductal

gray matter and the retrotrapezoid nucleus in breathing control in a rat model of

Parkinson’s disease. Experimental Neurology, 302, pp.46–56.

Lin, Y.-Y., Chen, R.-S., Lu, C.-S., Huang, Y.-Z., Weng, Y.-H., Yeh, T.-H., Lin, W.-Y. and

Hung, J., 2017. Sleep disturbances in Taiwanese patients with Parkinson’s disease.

Brain and Behavior, 7(10), p.e00806.

Loggia, M.L., Berna, C., Kim, J., Cahalan, C.M., Gollub, R.L., Wasan, A.D., Harris, R.E.,

Edwards, R.R. and Napadow, V., 2014. Disrupted brain circuitry for pain-related

reward/punishment in fibromyalgia. Arthritis and Rheumatology, 66(1).

Loher, T.J., Burgunder, J.-M., Weber, S., Sommerhalder, R. and Krauss, J.K., 2002.

Effect of chronic pallidal deep brain stimulation on off period dystonia and sensory

symptoms in advanced Parkinson’s disease. Journal of neurology, neurosurgery, and

psychiatry, 73(4), pp.395–9.

Lou, J.-S., Kearns, G., Oken, B., Sexton, G. and Nutt, J., 2001. Exacerbated physical

Page 211: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

211

fatigue and mental fatigue in Parkinson’s disease. Movement Disorders, 16(2),

pp.190–196.

Lu, C., Yang, T., Zhao, H., Zhang, M., Meng, F., Fu, H., Xie, Y. and Xu, H., 2016. Insular

Cortex is Critical for the Perception, Modulation, and Chronification of Pain.

Neuroscience bulletin, 32(2), pp.191–201.

Madden, D.J., Whiting, W.L., Provenzale, J.M. and Huettel, S.A., 2004. Age-related

Changes in Neural Activity during Visual Target Detection Measured by fMRI.

Cerebral Cortex, 14(2), pp.143–155.

Maillet, A., Krack, P., Lhommée, E., Météreau, E., Klinger, H., Favre, E., Le Bars, D.,

Schmitt, E., Bichon, A., Pelissier, P., Fraix, V., Castrioto, A., Sgambato-Faure, V.,

Broussolle, E., Tremblay, L. and Thobois, S., 2016. The prominent role of

serotonergic degeneration in apathy, anxiety and depression in de novo Parkinson’s

disease. Brain, 139(9), pp.2486–2502.

Mann, D.M., 1983. The locus coeruleus and its possible role in ageing and

degenerative disease of the human central nervous system. Mechanisms of ageing

and development, 23(1), pp.73–94.

MANN, D.M.A. and YATES, P.O., 1983. PATHOLOGICAL BASIS FOR

NEUROTRANSMITTER CHANGES IN PARKINSON’S DISEASE. Neuropathology and

Applied Neurobiology, 9(1), pp.3–19.

Margulies, D.S., Kelly, A.M.C., Uddin, L.Q., Biswal, B.B., Castellanos, F.X. and

Milham, M.P., 2007. Mapping the functional connectivity of anterior cingulate

cortex. NeuroImage, 37(2), pp.579–88.

Martikainen, I.K., Hagelberg, N., Mansikka, H., Hietala, J., Någren, K., Scheinin, H.

and Pertovaara, A., 2005. Association of striatal dopamine D2/D3 receptor binding

potential with pain but not tactile sensitivity or placebo analgesia. Neuroscience

Letters, 376(3), pp.149–153.

Martikainen, I.K., Nuechterlein, E.B., Peciña, M., Love, T.M., Cummiford, C.M.,

Page 212: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

212

Green, C.R., Stohler, C.S. and Zubieta, J.-K., 2015. Chronic Back Pain Is Associated

with Alterations in Dopamine Neurotransmission in the Ventral Striatum. The

Journal of neuroscience : the official journal of the Society for Neuroscience, 35(27),

pp.9957–65.

Maruya, H., Watanabe, Y., Okita, M., Lawlor, G.F., Utsumi, H. and Niitsuma, T.,

2003. Inhibitory effects of D2 agonists by striatal injection on excessive release of

dopamine and hyperactivity induced by Bay K 8644 in rats. Neuroscience, 118(4),

pp.1091–8.

Mavridis, M., Degryse, A.D., Lategan, A.J., Marien, M.R. and Colpaert, F.C., 1991.

Effects of locus coeruleus lesions on parkinsonian signs, striatal dopamine and

substantia nigra cell loss after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine in

monkeys: a possible role for the locus coeruleus in the progression of Parkinson’s

disease. Neuroscience, 41(2–3), pp.507–23.

Mayeux, R., Stern, Y., Sano, M., Williams, J.B. and Cote, L.J., 1988. The relationship

of serotonin to depression in Parkinson’s disease. Movement disorders : official

journal of the Movement Disorder Society, 3(3), pp.237–44.

Mazzola, V., Latorre, V., Petito, A., Gentili, N., Fazio, L., Popolizio, T., Blasi, G.,

Arciero, G. and Bondolfi, G., 2010. Affective response to a loved one’s pain: insula

activity as a function of individual differences. PloS one, 5(12), p.e15268.

Meador-Woodruff, J.H., Mansour, A., Healy, D.J., Kuehn, R., Zhou, Q.Y., Bunzow,

J.R., Akil, H., Civelli, O. and Watson, S.J., 1991. Comparison of the distributions of D1

and D2 dopamine receptor mRNAs in rat brain. Neuropsychopharmacology : official

publication of the American College of Neuropsychopharmacology, 5(4), pp.231–42.

Mehta, M.A., Hinton, E.C., Montgomery, A.J., Bantick, R.A. and Grasby, P.M., 2005.

Sulpiride and mnemonic function: effects of a dopamine D2 receptor antagonist on

working memory, emotional memory and long-term memory in healthy volunteers.

Journal of Psychopharmacology, 19(1), pp.29–38.

Page 213: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

213

Menon, V. and Uddin, L.Q., 2010. Saliency, switching, attention and control: a

network model of insula function. Brain Structure and Function, 214(5–6), pp.655–

667.

Millan, M.J., 2002. Descending control of pain. Progress in Neurobiology, 66(6),

pp.355–474.

Minton, J.P., n.d. T H E RESPONSE OF BREAST CANCER PATIENTS WITH BONE PAIN T

O L-DOPA.

Miron, D., Duncan, G.H. and Bushnell, C.M., 1989a. Effects of attention on the

intensity and unpleasantness of thermal pain. Pain, 39(3), pp.345–352.

Miron, D., Duncan, G.H. and Bushnell, M.C., 1989b. Effects of attention on the

intensity and unpleasantness of thermal pain. Pain, 39(3), pp.345–52.

Misra, G. and Coombes, S.A., 2015. Neuroimaging Evidence of Motor Control and

Pain Processing in the Human Midcingulate Cortex. Cerebral Cortex, 25(7), pp.1906–

1919.

Mitsi, V. and Zachariou, V., 2016. Modulation of pain, nociception, and analgesia by

the brain reward center. Neuroscience, 338, pp.81–92.

Montero-Marín, J., Navarro-Gil, M., Puebla-Guedea, M., Luciano, J. V., Van Gordon,

W., Shonin, E. and García-Campayo, J., 2018. Efficacy of “Attachment-Based

Compassion Therapy” in the Treatment of Fibromyalgia: A Randomized Controlled

Trial. Frontiers in Psychiatry, 8, p.307.

Morecraft, R.J. and Van Hoesen, G.W., 1998. Convergence of Limbic Input to the

Cingulate Motor Cortex in the Rhesus Monkey. Brain Research Bulletin, 45(2),

pp.209–232.

Morris, R., Martini, D.N., Madhyastha, T., Kelly, V.E., Grabowski, T.J., Nutt, J. and

Horak, F., 2019. Overview of the cholinergic contribution to gait, balance and falls in

Parkinson’s disease. Parkinsonism & Related Disorders, 63, pp.20-30.

Page 214: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

214

Morris, R.G., Downes, J.J., Sahakian, B.J., Evenden, J.L., Heald, A. and Robbins, T.W.,

1988. Planning and spatial working memory in Parkinson’s disease. Journal of

neurology, neurosurgery, and psychiatry, 51(6), pp.757–66.

Morrison, I., Peelen, M. V. and Downing, P.E., 2007. The Sight of Others’ Pain

Modulates Motor Processing in Human Cingulate Cortex. Cerebral Cortex, 17(9),

pp.2214–2222.

Mulert, C., Jäger, L., Schmitt, R., Bussfeld, P., Pogarell, O., Möller, H.-J., Juckel, G.

and Hegerl, U., 2004. Integration of fMRI and simultaneous EEG: towards a

comprehensive understanding of localization and time-course of brain activity in

target detection. NeuroImage, 22(1), pp.83–94.

Mylius, V., Engau, I., Teepker, M., Stiasny-Kolster, K., Schepelmann, K., Oertel, W.H.,

Lautenbacher, S. and Moller, J.C., 2008. Pain sensitivity and descending inhibition of

pain in Parkinson’s disease. Journal of Neurology, Neurosurgery & Psychiatry, 80(1),

pp.24–28.

Nagy, H., Levy-Gigi, E., Somlai, Z., Takáts, A., Bereczki, D. and Kéri, S., 2012. The

Effect of Dopamine Agonists on Adaptive and Aberrant Salience in Parkinson’s

Disease. Neuropsychopharmacology, 37(4), pp.950–958.

Nandam, L.S., Hester, R., Wagner, J., Dean, A.J., Messer, C., Honeysett, A., Nathan,

P.J. and Bellgrove, M.A., 2013. Dopamine D 2 Receptor Modulation of Human

Response Inhibition and Error Awareness. Journal of Cognitive Neuroscience, 25(4),

pp.649–656.

Narasimhan, M. and Campbell, N., 2010. A tale of two comorbidities:

Understanding the neurobiology of depression and pain. Indian journal of

psychiatry, 52(2), pp.127–30.

Nasreddine, Z.S., Phillips, N.A., Bédirian, V., Charbonneau, S., Whitehead, V., Collin,

I., Cummings, J.L. and Chertkow, H., 2005. The Montreal Cognitive Assessment,

MoCA: a brief screening tool for mild cognitive impairment. Journal of the American

Page 215: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

215

Geriatrics Society, 53(4), pp.695–9.

Nees, F. and Becker, S., 2017. Psychological processes in chronic pain: Influences of

reward and fear learning as key mechanisms – Behavioral evidence, neural circuits,

and maladaptive changes. Neuroscience, 387, pp.72-84.

Nees, F., Becker, S., Millenet, S., Banaschewski, T., Poustka, L., Bokde, A., Bromberg,

U., Büchel, C., Conrod, P.J., Desrivières, S., Frouin, V., Gallinat, J., Garavan, H., Heinz,

A., Ittermann, B., Martinot, J.-L., Papadopoulos Orfanos, D., Paus, T., Smolka, M.N.,

Walter, H., Whelan, R., Schumann, G., Flor, H. and IMAGEN consortium, 2017. Brain

substrates of reward processing and the μ-opioid receptor. PAIN, 158(2), pp.212–

219.

Nègre-Pagès, L., Regragui, W., Bouhassira, D., Grandjean, H. and Rascol, O., 2008a.

Chronic pain in Parkinson’s disease: the cross-sectional French DoPaMiP survey.

Movement disorders : official journal of the Movement Disorder Society, 23(10),

pp.1361–9.

Nègre-Pagès, L., Regragui, W., Bouhassira, D., Grandjean, H., Rascol, O. and

DoPaMiP Study Group, 2008b. Chronic pain in Parkinson’s disease: The cross-

sectional French DoPaMiP survey. Movement Disorders, 23(10), pp.1361–1369.

Nieoullon, A., 2002. Dopamine and the regulation of cognition and attention.

Progress in neurobiology, 67(1), pp.53–83.

Nixon, D.W., 1975. Letter: Use of L-dopa to relieve pain from bone metastases. The

New England journal of medicine, 292(12), p.647.

Nomoto, M., Kita, S., Iwata, S.I., Kaseda, S. and Fukuda, T., 1998. Effects of acute or

prolonged administration of cabergoline on parkinsonism induced by MPTP in

common marmosets. Pharmacology, biochemistry, and behavior, 59(3), pp.717–21.

Northoff, G., 2013. Unlocking the Brain: Volume 2: Consciousness. OUP USA.

Olivier, B., 2015. Serotonin: A never-ending story. European Journal of

Page 216: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

216

Pharmacology, 753, pp.2–18.

Olson, M., Lockhart, T.E. and Lieberman, A., 2019. Motor Learning Deficits in

Parkinson’s Disease (PD) and Their Effect on Training Response in Gait and Balance:

A Narrative Review. Frontiers in Neurology, 10, p.62.

Ong, A.D., Bergeman, C.S. and Boker, S.M., 2009. Resilience Comes of Age: Defining

Features in Later Adulthood. Journal of Personality, 77(6), pp.1777–1804.

Ong, W.-Y., Stohler, C.S. and Herr, D.R., 2019. Role of the Prefrontal Cortex in Pain

Processing. Molecular Neurobiology, 56(2), pp.1137–1166.

Onoda, K., Ishihara, M. and Yamaguchi, S., 2012. Decreased Functional Connectivity

by Aging Is Associated with Cognitive Decline. Journal of Cognitive Neuroscience,

24(11), pp.2186–2198.

Onur, O.A., Walter, H., Schlaepfer, T.E., Rehme, A.K., Schmidt, C., Keysers, C., Maier,

W. and Hurlemann, R., 2009. Noradrenergic enhancement of amygdala responses

to fear. SCAN, 4, pp.119–126.

Ossipov, M.H., Dussor, G.O. and Porreca, F., 2010. Central modulation of pain. The

Journal of clinical investigation, 120(11), pp.3779–87.

Ossipov, M.H. and Limitado, A.-A., 2012. The Perception and Endogenous

Modulation of Pain. Scientifica, 2012, pp.33–37.

Ossipov, M.H., Morimura, K. and Porreca, F., 2014. Descending pain modulation and

chronification of pain. Current opinion in supportive and palliative care, 8(2),

pp.143–51.

Ozturk, E.A., Gundogdu, I., Kocer, B., Comoglu, S. and Cakci, A., 2016. Chronic pain

in Parkinson’s disease: Frequency, characteristics, independent factors, and

relationship with health-related quality of life. Journal of Back and Musculoskeletal

Rehabilitation, 30(1), pp.101–108.

Pagano, R.L., Fonoff, E.T., Dale, C.S., Ballester, G., Teixeira, M.J. and Britto, L.R.G.,

Page 217: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

217

2012. Motor cortex stimulation inhibits thalamic sensory neurons and enhances

activity of PAG neurons: possible pathways for antinociception. Pain, 153(12),

pp.2359–69.

Palve, S.S. and Palve, S.B., 2018. Impact of Aging on Nerve Conduction Velocities

and Late Responses in Healthy Individuals. Journal of neurosciences in rural

practice, 9(1), pp.112–116.

Parr, T. and Friston, K.J., 2017. Working memory, attention, and salience in active

inference. Scientific Reports, 7(1), p.14678.

Pascual-Leone, A., Valls-Solé, J., Brasil-Neto, J.P., Cammarota, A., Grafman, J. and

Hallett, M., 1994a. Akinesia in Parkinson’s disease. II. Effects of subthreshold

repetitive transcranial motor cortex stimulation. Neurology, 44(5), pp.892–8.

Pascual-Leone, A., Valls-Solé, J., Brasil-Neto, J.P., Cohen, L.G. and Hallett, M., 1994b.

Akinesia in Parkinson’s disease. I. Shortening of simple reaction time with focal,

single-pulse transcranial magnetic stimulation. Neurology, 44(5), pp.884–91.

Pascual-Marqui, R.D., 2002. Standardized low resolution brain electromagnetic

tomography (sLORETA): technical details.

Patton, J.H., Stanford, M.S. and Barratt, E.S., 1995. Factor structure of the Barratt

impulsiveness scale. Journal of clinical psychology, 51(6), pp.768–74.

Pay, S. and Barasi, S., 1982. A Study of the Connections of Nociceptive Substantia

Nigra Neurones. Pain, 12(1), pp.75–89.

Pearson, J.M., Hayden, B.Y., Raghavachari, S. and Platt, M.L., 2009. Neurons in

Posterior Cingulate Cortex Signal Exploratory Decisions in a Dynamic Multioption

Choice Task. Current Biology, 19(18), pp.1532–1537.

Peciña, M., Mickey, B.J., Love, T., Wang, H., Langenecker, S.A., Hodgkinson, C.,

Shen, P.-H., Villafuerte, S., Hsu, D., Weisenbach, S.L., Stohler, C.S., Goldman, D. and

Zubieta, J.-K., 2013. DRD2 polymorphisms modulate reward and emotion

Page 218: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

218

processing, dopamine neurotransmission and openness to experience. Cortex,

49(3), pp.877–890.

Pertovaara, A., 2006. Noradrenergic pain modulation. Progress in neurobiology,

80(2), pp.53–83.

Pertovaara, A., 2013. Descending Modulation and Persistent Pain. In: Encyclopedia

of Pain. Berlin, Heidelberg: Springer Berlin Heidelberg, pp.927–930.

Pertovaara, A., Martikainen, I.K., Hagelberg, N., Mansikka, H., Någren, K., Hietala, J.

and Scheinin, H., 2004. Striatal dopamine D2/D3 receptor availability correlates

with individual response characteristics to pain. The European journal of

neuroscience, 20(6), pp.1587–92.

Pfingsten, M., Leibing, E., Harter, W., Kröner-Herwig, B., Hempel, D., Kronshage, U.

and Hildebrandt, J., 2001. Fear-Avoidance Behavior and Anticipation of Pain in

Patients With Chronic Low Back Pain: A Randomized Controlled Study. Pain

Medicine, 2(4), pp.259–266.

Phillips, M.L., Gregory, L.J., Cullen, S., Coen, S., Ng, V., Andrew, C., Giampietro, V.,

Bullmore, E., Zelaya, F., Amaro, E., Thompson, D.G., Hobson, A.R., Williams, S.C.R.,

Brammer, M., Aziz, Q. and Cohen, S., 2003. The effect of negative emotional

context on neural and behavioural responses to oesophageal stimulation. Brain : a

journal of neurology, 126(Pt 3), pp.669–84.

Pifl, C., Kish, S.J. and Hornykiewicz, O., 2012. Thalamic noradrenaline in Parkinson’s

disease: deficits suggest role in motor and non-motor symptoms. Movement

disorders : official journal of the Movement Disorder Society, 27(13), pp.1618–24.

Pleman, B., Park, M., Han, X., Price, L.L., Bannuru, R.R., Harvey, W.F., Driban, J.B.

and Wang, C., 2019. Mindfulness is associated with psychological health and

moderates the impact of fibromyalgia. Clinical Rheumatology, 38(6), pp.1737-1745.

Poewe, W. and Luginger, E., 1999. Depression in Parkinson’s disease: impediments

to recognition and treatment options. Neurology, 52(7 Suppl 3), pp.S2-6.

Page 219: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

219

Poewe, W.H., Lees, A.J. and Stern, G.M., 1988. Dystonia in parkinson’s disease:

Clinical and pharmacological features. Annals of Neurology, 23(1), pp.73–78.

Poletti, M., 2018. The dark side of dopaminergic therapies in Parkinson’s disease:

shedding light on aberrant salience. CNS Sp, 23(347–351), pp.347–351.

Poletti, M., Frosini, D., Pagni, C., Baldacci, F., Lucetti, C., Del Dotto, P., Ceravolo, R.

and Bonuccelli, U., 2014. A pilot psychometric study of aberrant salience state in

patients with Parkinson’s disease and its association with dopamine replacement

therapy. Neurological Sciences, 35(10), pp.1603–1605.

Politis, M. and Niccolini, F., 2015. Serotonin in Parkinson’s disease. Behavioural

brain research, 277, pp.136–45.

Politis, M., Wu, K., Loane, C., Kiferle, L., Molloy, S., Brooks, D.J. and Piccini, P., 2010.

Staging of serotonergic dysfunction in Parkinson’s Disease: An in vivo 11C-DASB PET

study. Neurobiology of Disease, 40(1), pp.216–221.

Polli, A., Weis, L., Biundo, R., Thacker, M., Turolla, A., Koutsikos, K., Chaudhuri, K.R.

and Antonini, A., 2016. Anatomical and functional correlates of persistent pain in

Parkinson’s disease. Movement Disorders, 31(12), pp.1854–1864.

Porreca, F., 2002. Chronic pain and medullary descending facilitation. Trends in

Neurosciences, 25(6), pp.319–325.

Priebe, J.A., Kunz, M., Morcinek, C., Rieckmann, P. and Lautenbacher, S., 2016.

Electrophysiological assessment of nociception in patients with Parkinson’s disease:

A multi-methods approach. Journal of the Neurological Sciences, 368, pp.59–69.

Prochazka, A., Bennett, D.J., Stephens, M.J., Patrick, S.K., Sears-Duru, R., Roberts, T.

and Jhamandas, J.H., 1997. Measurement of rigidity in Parkinson’s disease.

Movement Disorders, 12(1), pp.24–32.

Pugh, K.R., Mencl, W.E., Shaywitz, B.A., Shaywitz, S.E., Fulbright, R.K., Todd

Constable, R., Skudlarski, P., Marchione, K.E., Jenner, A.R., Fletcher, J.M., Liberman,

Page 220: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

220

A.M., Shankweiler, D.P., Katz, L., Lacadie, C. and Gore, J.C., 2000. THE ANGULAR

GYRUS IN DEVELOPMENTAL DYSLEXIA: Task-Specific Differences in Functional

Connectivity Within Posterior Cortex, 11(1), pp.51-6.

Pultorak, K.J., Schelp, S.A., Isaacs, D.P., Krzystyniak, G. and Oleson, E.B., 2018. A

Transient Dopamine Signal Represents Avoidance Value and Causally Influences the

Demand to Avoid. eNeuro, 5(2).

Rainville, P., Feine, J.S., Bushnell, M.C. and Duncan, G.H., 1992. A psychophysical

comparison of sensory and affective responses to four modalities of experimental

pain. Somatosensory & motor research, 9(4), pp.265–77.

Rana, A.Q., Kabir, A., Jesudasan, M., Siddiqui, I. and Khondker, S., 2013. Pain in

Parkinson’s disease: analysis and literature review. Clinical neurology and

neurosurgery, 115(11), pp.2313–7.

Reches, A. and Meiner, Z., 1992. The locus coeruleus and dopaminergic function in

rat brain: implications to parkinsonism. Brain research bulletin, 28(5), pp.663–6.

Reed, A.E. and Carstensen, L.L., 2012. The Theory Behind the Age-Related Positivity

Effect. Frontiers in Psychology, 3, p.339.

Reicherts, P., Wiemer, J., Gerdes, A.B.M., Schulz, S.M., Pauli, P. and Wieser, M.J.,

2017. Anxious anticipation and pain: the influence of instructed vs conditioned

threat on pain. Social cognitive and affective neuroscience, 12(4), pp.544–554.

Reisine, T.D., Fields, J.Z., Yamamura, H.I., Bird, E.D., Spokes, E., Schreiner, P.S. and

Enna, S.J., 1977. Neurotransmitter receptor alterations in Parkinson’s disease. Life

Sciences, 21(3), pp.335–343.

Richard, I.H., Schiffer, R.B. and Kurlan, R., 1996. Anxiety and Parkinson’s disease.

The Journal of Neuropsychiatry and Clinical Neurosciences, 8(4), pp.383–392.

Rogers, M.W., 1996. Disorders of Posture, Balance, and Gait in Parkinson’s Disease.

Clinics in Geriatric Medicine, 12(4), pp.825–845.

Page 221: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

221

Roland, P.E., 1981. Somatotopical Tuning of Postcentral Gyrus Dir A Regiona ng

Focal Attention in Man. Cerebral Blood Flow Study, Journal of Neurophysiology,

46(4), pp.744-54

Romano, J.M., Jensen, M.P., Turner, J.A., Good, A.B. and Hops, H., 2000. Chronic

pain patient-partner interactions: Further support for a behavioral model of chronic

pain. Behavior Therapy, 31(3), pp.415–440.

Rosenzweig, P., Canal, M., Patat, A., Bergougnan, L., Zieleniuk, I. and Bianchetti, G.,

2002. A review of the pharmacokinetics, tolerability and pharmacodynamics of

amisulpride in healthy volunteers. Human Psychopharmacology: Clinical and

Experimental, 17(1), pp.1–13.

Ross, M.H., Yurgelun-Todd, D.A., Renshaw, P.F., Maas, L.C., Mendelson, J.H., Mello,

N.K., Cohen, B.M. and Levin, J.M., 1997. Age-related reduction in functional MRI

response to photic stimulation. Neurology, 48(1), pp.173–6.

Rossini, P.M., Filippi, M.M. and Vernieri, F., 1998. Neurophysiology of sensorimotor

integration in Parkinson’s disease. Clinical neuroscience (New York, N.Y.), 5(2),

pp.121–30.

Roughley, S. and Killcross, S., 2019. Differential involvement of dopamine receptor

subtypes in the acquisition of Pavlovian sign-tracking and goal-tracking responses.

Psychopharmacology, 236(6), pp.1853-1862.

Roy, M., Piché, M., Chen, J.-I., Peretz, I. and Rainville, P., 2009. Cerebral and spinal

modulation of pain by emotions. Proceedings of the National Academy of Sciences

of the United States of America, 106(49), pp.20900–5.

Ruscheweyh, R., Kreusch, A., Albers, C., Sommer, J. and Marziniak, M., 2011. The

effect of distraction strategies on pain perception and the nociceptive flexor reflex

(RIII reflex). Pain, 152(11), pp.2662–71.

Rushworth, M.F.S., Walton, M.E., Kennerley, S.W. and Bannerman, D.M., 2004.

Action sets and decisions in the medial frontal cortex. Trends in Cognitive Sciences,

Page 222: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

222

8(9), pp.410–417.

Russo, G.S., Backus, D.A., Ye, S. and Crutcher, M.D., 2002. Neural Activity in Monkey

Dorsal and Ventral Cingulate Motor Areas: Comparison with the Supplementary

Motor Area. Journal of Neurophysiology, 88(5), pp.2612–2629.

Rustøen, T., Wahl, A.K., Hanestad, B.R., Lerdal, A., Paul, S. and Miaskowski, C., 2005.

Age and the experience of chronic pain: differences in health and quality of life

among younger, middle-aged, and older adults. The Clinical journal of pain, 21(6),

pp.513–23.

Ryoo, H.L., Pierrotti, D. and Joyce, J.N., 1998. Dopamine D3 receptor is decreased

and D2 receptor is elevated in the striatum of Parkinson’s disease. Movement

Disorders, 13(5), pp.788–797.

Saadé, N.E., Atweh, S.F., Bahuth, N.B. and Jabbur, S.J., 1997. Augmentation of

nociceptive reflexes and chronic deafferentation pain by chemical lesions of either

dopaminergic terminals or midbrain dopaminergic neurons. Brain Research, 751(1),

pp.1–12.

Salimpoor, V.N., Benovoy, M., Larcher, K., Dagher, A. and Zatorre, R.J., 2011.

Anatomically distinct dopamine release during anticipation and experience of peak

emotion to music. Nature Neuroscience, 14(2), pp.257–262.

Samanez-Larkin, G.R., Gibbs, S.E.B., Khanna, K., Nielsen, L., Carstensen, L.L. and

Knutson, B., 2007. Anticipation of monetary gain but not loss in healthy older

adults. Nature Neuroscience, 10(6), pp.787–791.

Sanchez-Ramos, J.R., Ortoll, R. and Paulson, G.W., 1996. Visual Hallucinations

Associated With Parkinson Disease. Archives of Neurology, 53(12), pp.1265–1268.

Sandyk, R., Bernick, C., Lee, S.M., Stern, L.Z., Iacono, R.P. and Bamford, C.R., 1987. L-

Dopa in Uremic Patients with the Restless Legs Syndrome. International Journal of

Neuroscience, 35(3–4), pp.233–235.

Page 223: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

223

Sansone, R.A. and Sansone, L.A., 2008. Pain, pain, go away: antidepressants and

pain management. Psychiatry (Edgmont (Pa. : Township)), 5(12), pp.16–9.

Scatton, B., Javoy-Agid, F., Rouquier, L., Dubois, B. and Agid, Y., 1983. Reduction of

cortical dopamine, noradrenaline, serotonin and their metabolites in Parkinson’s

disease. Brain Research, 275(2), pp.321–328.

Scherder, E., Wolters, E., Polman, C., Sergeant, J. and Swaab, D., 2005. Pain in

Parkinson’s disease and multiple sclerosis: Its relation to the medial and lateral pain

systems. Neuroscience & Biobehavioral Reviews, 29(7), pp.1047–1056.

Schestatsky, P., Kumru, H., Valls-Solé, J., Valldeoriola, F., Marti, M.J., Tolosa, E. and

Chaves, M.L., 2007a. Neurophysiologic study of central pain in patients with

Parkinson disease. Neurology, 69(23), pp.2162–9.

Schestatsky, P., Kumru, H., Valls-Solé, J., Valldeoriola, F., Marti, M.J., Tolosa, E. and

Chaves, M.L., 2007b. Neurophysiologic study of central pain in patients with

Parkinson disease. Neurology, 69(23), pp.2162–9.

Schmid, J., Theysohn, N., Gaß, F., Benson, S., Gramsch, C., Forsting, M., Gizewski,

E.R. and Elsenbruch, S., 2013. Neural mechanisms mediating positive and negative

treatment expectations in visceral pain: A functional magnetic resonance imaging

study on placebo and nocebo effects in healthy volunteers. PAIN®, 154(11),

pp.2372–2380.

Schott, G.D., 1985. Pain in Parkinson’s disease. Pain, 22(4), pp.407–411.

Van Schouwenburg, M.R., Den Ouden, H.E.M. and Cools, R., 2010. The Human Basal

Ganglia Modulate Frontal-Posterior Connectivity during Attention Shifting.

Schrag, A., 2004. Psychiatric aspects of Parkinson?s disease. Journal of Neurology,

251(7), pp.795–804.

Schwartenbeck, P., FitzGerald, T.H.B., Mathys, C., Dolan, R. and Friston, K., 2015.

The Dopaminergic Midbrain Encodes the Expected Certainty about Desired

Page 224: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

224

Outcomes. Cerebral Cortex, 25(10), pp.3434–3445.

Schweinhardt, P., Seminowicz, D.A., Jaeger, E., Duncan, G.H. and Bushnell, M.C.,

2009. The anatomy of the mesolimbic reward system: a link between personality

and the placebo analgesic response. The Journal of neuroscience : the official

journal of the Society for Neuroscience, 29(15), pp.4882–7.

Scott, D.J., Heitzeg, M.M., Koeppe, R.A., Stohler, C.S. and Zubieta, J.-K., 2006.

Variations in the human pain stress experience mediated by ventral and dorsal

basal ganglia dopamine activity. The Journal of neuroscience : the official journal of

the Society for Neuroscience, 26(42), pp.10789–95.

Scott, D.J., Stohler, C.S., Egnatuk, C.M., Wang, H., Koeppe, R.A. and Zubieta, J.-K.,

2008. Placebo and nocebo effects are defined by opposite opioid and dopaminergic

responses. Archives of general psychiatry, 65(2), pp.220–31.

Seeck, M., Lazeyras, F., Michel, C.M., Blanke, O., Gericke, C.A., Ives, J., Delavelle, J.,

Golay, X., Haenggeli, C.A., de Tribolet, N. and Landis, T., 1998. Non-invasive

epileptic focus localization using EEG-triggered functional MRI and electromagnetic

tomography. Electroencephalography and clinical neurophysiology, 106(6), pp.508–

12.

Seghier, M.L., 2013. The angular gyrus: multiple functions and multiple subdivisions.

The Neuroscientist : a review journal bringing neurobiology, neurology and

psychiatry, 19(1), pp.43–61.

Seidel, E.-M., Pfabigan, D.M., Hahn, A., Sladky, R., Grahl, A., Paul, K., Kraus, C.,

Küblböck, M., Kranz, G.S., Hummer, A., Lanzenberger, R., Windischberger, C. and

Lamm, C., 2015a. Uncertainty during pain anticipation: The adaptive value of

preparatory processes. Human Brain Mapping, 36(2), pp.744–755.

Seidel, K., Mahlke, J., Siswanto, S., Krüger, R., Heinsen, H., Auburger, G., Bouzrou,

M., Grinberg, L.T., Wicht, H., Korf, H.-W., den Dunnen, W. and Rüb, U., 2015b. The

brainstem pathologies of Parkinson’s disease and dementia with Lewy bodies. Brain

Page 225: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

225

pathology (Zurich, Switzerland), 25(2), pp.121–35.

Sewards, T. V and Sewards, M.A., 2002. The medial pain system: neural

representations of the motivational aspect of pain. Brain research bulletin, 59(3),

pp.163–80.

Shackman, A.J., Salomons, T. V., Slagter, H.A., Fox, A.S., Winter, J.J. and Davidson,

R.J., 2011. The integration of negative affect, pain and cognitive control in the

cingulate cortex. Nature Reviews Neuroscience, 12(3), pp.154–167.

Shao, S., Shen, K., Yu, K., Wilder-Smith, E.P.V. and Li, X., 2012. Frequency-domain

EEG source analysis for acute tonic cold pain perception. Clinical Neurophysiology,

123(10), pp.2042–2049.

Shen, W., Flajolet, M., Greengard, P. and Surmeier, D.J., 2008. Dichotomous

dopaminergic control of striatal synaptic plasticity. Science (New York, N.Y.),

321(5890), pp.848–51.

Shimizu, T., Iwata, S., Morioka, H., Masuyama, T., Fukuda, T. and Nomoto, M.,

2004a. Antinociceptive mechanism of l-DOPA. Pain, 110(1), pp.246–249.

Shimizu, T., Iwata, S., Morioka, H., Masuyama, T., Fukuda, T. and Nomoto, M.,

2004b. Antinociceptive mechanism of l-DOPA. Pain, 110(1–2), pp.246–249.

Shiner, T., Symmonds, M., Guitart-Masip, M., Fleming, S.M., Friston, K.J. and Dolan,

R.J., 2015. Dopamine, Salience, and Response Set Shifting in Prefrontal Cortex.

Cerebral Cortex, 25(10), pp.3629–3639.

Shokouhi, M., Davis, K.D., Moulin, D.E., Morley-Forster, P., Nielson, W.R., Bureau,

Y., St. Lawrence, K. and St Lawrence, K., 2016. Basal Ganglia Perfusion in

Fibromyalgia is Related to Pain Disability and Disease Impact: An Arterial Spin

Labeling Study. The Clinical journal of pain, 32(6), pp.495–505.

Shpaner, M., Kelly, C., Lieberman, G., Perelman, H., Davis, M., Keefe, F.J. and

Naylor, M.R., 2014. Unlearning chronic pain: A randomized controlled trial to

Page 226: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

226

investigate changes in intrinsic brain connectivity following Cognitive Behavioral

Therapy. NeuroImage: Clinical, 5, pp.365–376.

Shukla, D., 1985. Blink rate as clinical indicator. Neurology, 35(2), p.286.

Silva, E.G. da, Viana, M.A. and Quagliato, E.M.A.B., 2008. Pain in Parkinson’s

disease: analysis of 50 cases in a clinic of movement disorders. Arquivos de neuro-

psiquiatria, 66(1), pp.26–9.

Silveira da Mota Neto, J.I., Soares, B.G. and Silva de Lima, M., 2002. Amisulpride for

schizophrenia. Cochrane Database of Systematic Reviews, (2), p.CD001357.

Silverdale, M.A., Kobylecki, C., Kass-Iliyya, L., Martinez-Martin, P., Lawton, M.,

Cotterill, S., Chaudhuri, K.R., Morris, H., Baig, F., Williams, N., Hubbard, L., Hu, M.T.,

Grosset, D.G. and UK Parkinson’s Pain Study Collaboration, 2018. A detailed clinical

study of pain in 1957 participants with early/moderate Parkinson’s disease.

Parkinsonism & Related Disorders, 56, pp.27–32.

Singer, T., Seymour, B., O’Doherty, J., Kaube, H., Dolan, R.J. and Frith, C.D., 2004a.

Empathy for pain involves the affective but not sensory components of pain.

Science (New York, N.Y.), 303(5661), pp.1157–62.

Singer, T., Seymour, B., O’Doherty, J., Kaube, H., Dolan, R.J. and Frith, C.D., 2004b.

Empathy for Pain Invovles the Affective but not Sensory Components of Pain.

Science, 303, pp.1157–1161.

Skogar, O. and Lokk, J., 2016. Pain management in patients with Parkinson’s

disease: challenges and solutions. Journal of multidisciplinary healthcare, 9, pp.469–

479.

Sogabe, S., Yagasaki, Y., Onozawa, K. and Kawakami, Y., 2013. Mesocortical

dopamine system modulates mechanical nociceptive responses recorded in the rat

prefrontal cortex. BMC neuroscience, 14, p.65.

Sommer, C., 2004. Serotonin in pain and analgesia: actions in the periphery.

Page 227: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

227

Molecular neurobiology, 30(2), pp.117–25.

Spitoni, G.F., Pireddu, G., Cimmino, R.L., Galati, G., Priori, A., Lavidor, M., Jacobson,

L. and Pizzamiglio, L., 2013. Right but not left angular gyrus modulates the metric

component of the mental body representation: a tDCS study. Experimental Brain

Research, 228(1), pp.63–72.

Stahl, S.M., 2009. Fibromyalgia--pathways and neurotransmitters. Human

psychopharmacology, 24 Suppl 1, pp.S11-7.

Stanford, M.S., Mathias, C.W., Dougherty, D.M., Lake, S.L., Anderson, N.E. and

Patton, J.H., 2009. Fifty years of the Barratt Impulsiveness Scale: An update and

review. Personality and Individual Differences, 47(5), pp.385–395.

Starr, C.J., Sawaki, L., Wittenberg, G.F., Burdette, J.H., Oshiro, Y., Quevedo, A.S. and

Coghill, R.C., 2009. Roles of the insular cortex in the modulation of pain: insights

from brain lesions. The Journal of neuroscience : the official journal of the Society

for Neuroscience, 29(9), pp.2684–94.

Starr, C.J., Sawaki, L., Wittenberg, G.F., Burdette, J.H., Oshiro, Y., Quevedo, A.S.,

McHaffie, J.G. and Coghill, R.C., 2011. The contribution of the putamen to sensory

aspects of pain: insights from structural connectivity and brain lesions. Brain : a

journal of neurology, 134(Pt 7), pp.1987–2004.

Strobel, C., Hunt, S., Sullivan, R., Sun, J. and Sah, P., 2014. Emotional regulation of

pain: the role of noradrenaline in the amygdala. Science China. Life sciences, 57(4),

pp.384–90.

Suhara, T., Fukuda, H., Inoue, O., Itoh, T., Suzuki, K., Yamasaki, T. and Tateno, Y.,

1991. Age-related changes in human D1 dopamine receptors measured by positron

emission tomography. Psychopharmacology, 103(1), pp.41–5.

Sulzer, D. and Surmeier, D.J., 2013. Neuronal vulnerability, pathogenesis, and

Parkinson’s disease. Movement disorders : official journal of the Movement Disorder

Society, 28(6), pp.715–24.

Page 228: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

228

Tan, L.L., Pelzer, P., Heinl, C., Tang, W., Gangadharan, V., Flor, H., Sprengel, R.,

Kuner, T. and Kuner, R., 2017. A pathway from midcingulate cortex to posterior

insula gates nociceptive hypersensitivity. Nature Neuroscience, 20(11), pp.1591–

1601.

Tan, Y., Tan, J., Deng, J., Cui, W., He, H., Yang, F., Deng, H., Xiao, R., Huang, Z.,

Zhang, X., Tan, R., Shen, X., Liu, T., Wang, X., Yao, D. and Luo, C., 2015a. Alteration

of Basal Ganglia and Right Frontoparietal Network in Early Drug-Naïve Parkinson’s

Disease during Heat Pain Stimuli and Resting State. Frontiers in human

neuroscience, 9, p.467.

Tan, Y., Tan, J., Luo, C., Cui, W., He, H., Bin, Y., Deng, J., Tan, R., Tan, W., Liu, T.,

Zeng, N., Xiao, R., Yao, D. and Wang, X., 2015b. Altered Brain Activation in Early

Drug-Naive Parkinson’s Disease during Heat Pain Stimuli: An fMRI Study.

Parkinson’s Disease, 2015, pp.1–8.

Täubel, J., Ferber, G., Fox, G., Fernandes, S., Lorch, U. and Camm, A.J., 2017.

Thorough QT study of the effect of intravenous amisulpride on QTc interval in

Caucasian and Japanese healthy subjects. British journal of clinical pharmacology,

83(2), pp.339–348.

Taylor, A.M.W., Becker, S., Schweinhardt, P. and Cahill, C., 2016. Mesolimbic

dopamine signaling in acute and chronic pain: implications for motivation,

analgesia, and addiction. Pain, 157(6), pp.1194–8.

Taylor, B.K., Joshi, C. and Uppal, H., 2003. Stimulation of dopamine D2 receptors in

the nucleus accumbens inhibits inflammatory pain. Brain Research, 987(2), pp.135–

143.

Taylor, J.R., Elsworth, J.D., Lawrence, M.S., Sladek, J.R., Roth, R.H. and Redmond,

D.E., 1999. Spontaneous Blink Rates Correlate with Dopamine Levels in the Caudate

Nucleus of MPTP-Treated Monkeys. Experimental Neurology, 158(1), pp.214–220.

Taylor, P.C.J., Muggleton, N.G., Kalla, R., Walsh, V. and Eimer, M., 2011. TMS of the

Page 229: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

229

right angular gyrus modulates priming of pop-out in visual search: combined TMS-

ERP evidence. Journal of Neurophysiology, 106(6), pp.3001–3009.

Tessitore, A., Russo, A., De Micco, R., Fratello, M., Caiazzo, G., Giordano, A., Cirillo,

M., Tedeschi, G. and Esposito, F., 2018. Central pain processing in “drug-naïve”

pain-free patients with Parkinson’s disease. Human Brain Mapping, 39(2), pp.932–

940.

Thobois, S., Prange, S., Sgambato-Faure, V., Tremblay, L. and Broussolle, E., 2017.

Imaging the Etiology of Apathy, Anxiety, and Depression in Parkinson’s Disease:

Implication for Treatment. Current Neurology and Neuroscience Reports, 17(10),

p.76.

Tiemann, L., Heitmann, H., Schulz, E., Baumkötter, J. and Ploner, M., 2014.

Dopamine precursor depletion influences pain affect rather than pain sensation.

PloS one, 9(4), p.e96167.

Tinazzi, M., Recchia, S., Simonetto, S., Defazio, G., Tamburin, S., Moretto, G.,

Fiaschi, A., Miliucci, R. and Valeriani, M., 2009. Hyperalgesia and laser evoked

potentials alterations in hemiparkinson: Evidence for an abnormal nociceptive

processing. Journal of the Neurological Sciences, 276(1–2), pp.153–158.

Tinazzi, M., Del Vesco, C., Defazio, G., Fincati, E., Smania, N., Moretto, G., Fiaschi, A.,

Le Pera, D. and Valeriani, M., 2008. Abnormal processing of the nociceptive input in

Parkinson’s disease: A study with CO2 laser evoked potentials. Pain, 136(1),

pp.117–124.

Tinazzi, M., Del Vesco, C., Fincati, E., Ottaviani, S., Smania, N., Moretto, G., Fiaschi,

A., Martino, D. and Defazio, G., 2006. Pain and motor complications in Parkinson’s

disease. Journal of neurology, neurosurgery, and psychiatry, 77(7), pp.822–5.

Tolosa, E. and Compta, Y., 2006. Dystonia in Parkinson’s disease. Journal of

Neurology, 253(S7), pp.vii7-vii13.

Torta, D.M.E. and Castelli, L., 2008. Reward pathways in Parkinson’s disease: Clinical

Page 230: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

230

and theoretical implications. Psychiatry and Clinical Neurosciences, 62(2), pp.203–

213.

Tranel, D., Damasio, H. and Damasio, A.R., 1997. A neural basis for the retrieval of

conceptual knowledge. Neuropsychologia, 35(10), pp.1319–1327.

Trébuchon-Da Fonseca, A., Giraud, K., Badier, J.-M., Chauvel, P. and Liégeois-

Chauvel, C., 2005. Hemispheric lateralization of voice onset time (VOT) comparison

between depth and scalp EEG recordings. NeuroImage, 27(1), pp.1–14.

Del Tredici, K. and Braak, H., 2013. Dysfunction of the locus coeruleus-

norepinephrine system and related circuitry in Parkinson’s disease-related

dementia. Journal of neurology, neurosurgery, and psychiatry, 84(7), pp.774–83.

Treede, R.D., Apkarian, A. V, Bromm, B., Greenspan, J.D. and Lenz, F.A., 2000.

Cortical representation of pain: functional characterization of nociceptive areas

near the lateral sulcus. Pain, 87(2), pp.113–9.

Valkovic, P., Minar, M., Singliarova, H., Harsany, J., Hanakova, M., Martinkova, J.

and Benetin, J., 2015. Pain in Parkinson´s Disease: A Cross-Sectional Study of Its

Prevalence, Types, and Relationship to Depression and Quality of Life. PloS one,

10(8), p.e0136541.

Verhoeven, K., Crombez, G., Eccleston, C., Van Ryckeghem, D.M.L., Morley, S. and

Van Damme, S., 2010. The role of motivation in distracting attention away from

pain: An experimental study. PAIN®, 149(2), pp.229–234.

Vijayraghavan, S., Wang, M., Birnbaum, S.G., Williams, G. V and Arnsten, A.F.T.,

2007. Inverted-U dopamine D1 receptor actions on prefrontal neurons engaged in

working memory. Nature Neuroscience, 10(3), pp.376–384.

Vilar, L., Vilar, C., Albuquerque, L., The, A.C., Trovao, E., Gadelha, P., Sampaio, I.,

Gomes, B., Ferreira, L., Aroucha, P., Lyra, R., Fonseca, M. and Lyra, R., 2018. Efficacy

and tolerability of cabergoline in a cohort of 160 prolactinomas at weekly doses of

up to 9 mg. Endocrine Abstracts, 56, pp.882.

Page 231: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

231

Vitacco, D., Brandeis, D., Pascual-Marqui, R. and Martin, E., 2002. Correspondence

of event-related potential tomography and functional magnetic resonance imaging

during language processing. Human Brain Mapping, 17(1), pp.4–12.

Vogt, B.A., Berger, G.R. and Derbyshire, S.W.G., 2003. Structural and functional

dichotomy of human midcingulate cortex. The European journal of neuroscience,

18(11), pp.3134–44.

Vogt, B.A., Finch, D.M. and Olson, C.R., 1992. Functional heterogeneity in cingulate

cortex: the anterior executive and posterior evaluative regions. Cerebral cortex

(New York, N.Y. : 1991), 2(6), pp.435–43.

Vogt, B.A. and Sikes, R.W., 2000. The medial pain system, cingulate cortex, and

parallel processing of nociceptive information. Progress in brain research, 122,

pp.223–35.

Voitenkov, V.B., Ekusheva, E. V, Komancev, V.N., Skripchenko, N. V, Grigoryev, S.G.,

Klimkin, A. V and Aksenova, A.I., 2017. [Age-related changes of sensory peripheral

nerve system in healthy subjects.]. Advances in gerontology = Uspekhi gerontologii,

30(6), pp.802–808.

Volkow, N.D., Ding, Y.S., Fowler, J.S., Wang, G.J., Logan, J., Gatley, S.J., Hitzemann,

R., Smith, G., Fields, S.D. and Gur, R., 1996a. Dopamine transporters decrease with

age. Journal of nuclear medicine : official publication, Society of Nuclear Medicine,

37(4), pp.554–9.

Volkow, N.D., Gur, R.C., Wang, G.J., Fowler, J.S., Moberg, P.J., Ding, Y.S., Hitzemann,

R., Smith, G. and Logan, J., 1998. Association between decline in brain dopamine

activity with age and cognitive and motor impairment in healthy individuals. The

American journal of psychiatry, 155(3), pp.344–9.

Volkow, N.D., Logan, J., Fowler, J.S., Wang, G.-J., Gur, R.C., Wong, C., Felder, C.,

Gatley, S.J., Ding, Y.-S., Hitzemann, R. and Pappas, N., 2000. Association Between

Age-Related Decline in Brain Dopamine Activity and Impairment in Frontal and

Page 232: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

232

Cingulate Metabolism. American Journal of Psychiatry, 157(1), pp.75–80.

Volkow, N.D., Wang, G.J., Fowler, J.S., Logan, J., Gatley, S.J., MacGregor, R.R.,

Schlyer, D.J., Hitzemann, R. and Wolf, A.P., 1996b. Measuring age-related changes

in dopamine D2 receptors with 11C-raclopride and 18F-N-methylspiroperidol.

Psychiatry research, 67(1), pp.11–6.

Wang, J.-Y., Zhang, H.-T., Chang, J.-Y., Woodward, D.J., Baccalá, L.A. and Luo, F.,

2008. Anticipation of pain enhances the nociceptive transmission and functional

connectivity within pain network in rats. Molecular pain, 4(1), p.34.

Wasner, G. and Deuschl, G., 2012. Pains in Parkinson disease--many syndromes

under one umbrella. Nature reviews. Neurology, 8(5), pp.284–94.

Watson, A., El-Deredy, W., Iannetti, G.D., Lloyd, D., Tracey, I., Vogt, B.A., Nadeau, V.

and Jones, A.K.P., 2009. Placebo conditioning and placebo analgesia modulate a

common brain network during pain anticipation and perception. Pain, 145(1),

pp.24–30.

Webster, J., Piscitelli, G., Polli, A., D’Alberton, A., Falsetti, L., Ferrari, C., Fioretti, P.,

Giordano, G., L’Hermite, M. and Ciccarelli, E., 1993. The efficacy and tolerability of

long-term cabergoline therapy in hyperprolactinaemic disorders: an open,

uncontrolled, multicentre study. European Multicentre Cabergoline Study Group.

Clinical endocrinology, 39(3), pp.323–9.

Wenzel-Seifert, K., Wittmann, M. and Haen, E., 2011. QTc prolongation by

psychotropic drugs and the risk of Torsade de Pointes. Deutsches Arzteblatt

international, 108(41), pp.687–93.

Whitaker, G., 2017. Investigating the Role of Striatal Dopamine in Attentional

Inhibition. The University of Manchester (PhD Thesis)

White, I.M. and Rebec, G. V., 1993. Responses of rat striatal neurons during

performance of a lever-release version of the conditioned avoidance response task.

Brain Research, 616(1–2), pp.71–82.

Page 233: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

233

Wiech, K., Lin, C.-S., Brodersen, K.H., Bingel, U., Ploner, M. and Tracey, I., 2010.

Behavioral/Systems/Cognitive Anterior Insula Integrates Information about Salience

into Perceptual Decisions about Pain, Journal of Neuroscience, 30(48), pp.16324-

16331

de Wied, M. and Verbaten, M.N., 2001. Affective pictures processing, attention, and

pain tolerance. Pain, 90(1–2), pp.163–172.

Wolfe, F., Ross, K., Anderson, J. and Russell, I.J., 1995. Aspects of fibromyalgia in the

general population: sex, pain threshold, and fibromyalgia symptoms. The Journal of

rheumatology, 22(1), pp.151–6.

Wolfe, F., Russell, I.J., Vipraio, G., Ross, K. and Anderson, J., 1997. Serotonin levels,

pain threshold, and fibromyalgia symptoms in the general population. The Journal

of rheumatology, 24(3), pp.555–9.

Wood, P.B., 2004. Stress and dopamine: implications for the pathophysiology of

chronic widespread pain. Medical hypotheses, 62(3), pp.420–4.

Wood, P.B., 2014. Role of central dopamine in pain and analgesia. Expert Review of

Neurotherapeutics, 8(5), pp.781-97.

Wood, P.B., Patterson, J.C., Sunderland, J.J., Tainter, K.H., Glabus, M.F. and Lilien,

D.L., 2007a. Reduced Presynaptic Dopamine Activity in Fibromyalgia Syndrome

Demonstrated With Positron Emission Tomography: A Pilot Study. The Journal of

Pain, 8(1), pp.51–58.

Wood, P.B., Schweinhardt, P., Jaeger, E., Dagher, A., Hakyemez, H., Rabiner, E.A.,

Bushnell, M.C. and Chizh, B.A., 2007b. Fibromyalgia patients show an abnormal

dopamine response to pain. The European journal of neuroscience, 25(12),

pp.3576–82.

Wu, T., Wang, J., Wang, C., Hallett, M., Zang, Y., Wu, X. and Chan, P., 2012. Basal

ganglia circuits changes in Parkinson’s disease patients. Neuroscience letters,

524(1), pp.55–9.

Page 234: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

234

Wulff, S., Nielsen, M.Ø., Rostrup, E., Svarer, C., Jensen, L.T., Pinborg, L. and

Glenthøj, B.Y., 2019. The relation between dopamine D2 receptor blockade and the

brain reward system: a longitudinal study of first-episode schizophrenia patients.

Psychological Medicine, pp.1–9.

Wunderlich, A.P., Klug, R., Stuber, G., Landwehrmeyer, B., Weber, F. and Freund,

W., 2011. Caudate nucleus and insular activation during a pain suppression

paradigm comparing thermal and electrical stimulation. The open neuroimaging

journal, 5, pp.1–8.

Xu, Y., Yan, J., Zhou, P., Li, J., Gao, H., Xia, Y. and Wang, Q., 2012. Neurotransmitter

receptors and cognitive dysfunction in Alzheimer’s disease and Parkinson’s disease.

Progress in Neurobiology, 97(1), pp.1–13.

Yang, Z., Jackson, T. and Huang, C., 2016. Neural Activation during Anticipation of

Near Pain-Threshold Stimulation among the Pain-Fearful. Frontiers in neuroscience,

10, p.342.

Ye, Z., Hammer, A., Camara, E. and Münte, T.F., 2011. Pramipexole modulates the

neural network of reward anticipation. Human Brain Mapping, 32(5), pp.800–811.

Yezierski, R.P., 2012. The effects of age on pain sensitivity: preclinical studies. Pain

medicine (Malden, Mass.), 13 Suppl 2(Suppl 2), pp.S27-36.

Yong, H.-H., 2006. Can attitudes of stoicism and cautiousness explain observed age-

related variation in levels of self-rated pain, mood disturbance and functional

interference in chronic pain patients? European Journal of Pain, 10(5), pp.399–407.

Young, S.N., Smith, S.E., Pihp ’, R.O. and Ervin, F.R., 1985. Tryptophan depletion

causes a rapid lowering of mood in normal males. Psychopharmacology, 87,

pp.173–177.

Zambito Marsala, S., Tinazzi, M., Vitaliani, R., Recchia, S., Fabris, F., Marchini, C.,

Fiaschi, A., Moretto, G., Giometto, B., Macerollo, A. and Defazio, G., 2011.

Spontaneous pain, pain threshold, and pain tolerance in Parkinson’s disease.

Page 235: AN INV STIATION O PAIN PRO SSIN IN PARKINSON’S ISAS

References

235

Journal of neurology, 258(4), pp.627–33.

Zeidan, F., Grant, J.A., Brown, C.A., McHaffie, J.G. and Coghill, R.C., 2012.

Mindfulness meditation-related pain relief: evidence for unique brain mechanisms

in the regulation of pain. Neuroscience letters, 520(2), pp.165–73.

Zumsteg, D., Wennberg, R.A., Treyer, V., Buck, A. and Wieser, H.G., 2005. H215O or

13NH3 PET and electromagnetic tomography (LORETA) during partial status

epilepticus. Neurology, 65(10), pp.1657–1660.