determination of the sub-cellular mechanisms underlying ... · toxins which mimic purported...

256
Determination of the Sub-Cellular Mechanisms Underlying Neurodegeneration in Parkinson’s Disease by Christopher J Yong-Kee A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Cell & Systems Biology University of Toronto © Copyright by Christopher J Yong-Kee 2013

Upload: others

Post on 27-Jul-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

Determination of the Sub-Cellular Mechanisms Underlying Neurodegeneration in Parkinson’s Disease

by

Christopher J Yong-Kee

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Cell & Systems Biology

University of Toronto

© Copyright by Christopher J Yong-Kee 2013

Page 2: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

ii

Determination of the Sub-Cellular Mechanisms Underlying

Neurodegeneration in Parkinson’s Disease

Christopher J Yong-Kee

Doctor of Philosophy

Department of Cell & Systems Biology University of Toronto

2013

Abstract

Parkinson’s disease (PD) is the second most common neurodegenerative disease affecting

approximately 1.8% of the population over 65 years of age. It is characterized by three cardinal

symptoms: bradykinesia, muscle rigidity and resting tremor. Symptoms are presented following

50% loss of dopaminergic neurons within the substantia nigra pars compacta (SNc).

Neurodegeneration is associated with reactive oxygen species (ROS) production, protein

aggregation, mitochondrial dysfunction, ubiquitin-proteasome system (UPS) inhibition and

lysosomal malfunction; however it is unclear if a single mechanism or multiple mechanisms lead

to disease onset. The primary aim of the studies described in this thesis was to elucidate the

interactions between various pathological mechanisms underlying PD pathology. An

examination of organelle function during exposure of SH-SY5Y neuroblastoma to a variety of

toxins which mimic purported pathological processes in PD reveal mitochondrial membrane

potential becomes depolarized, not only following mitochondrial impairment, but also after the

UPS and lysosome are inhibited. Given that mitochondrial dysfunction appeared to be central to

PD pathology, mitochondrial dysfunction was studied in more detail. Mitochondrial fission and

fusion maintains mitochondrial integrity, which is critical to neuronal health. Thus, we

examined mitochondrial dynamics in a common genetic variant linked with familial PD, known

Page 3: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

iii

as leucine-rich repeat kinase 2 (LRRK2). Upon the expression of wild-type and mutant LRRK2,

mitochondrial fusion was inhibited causing fragmentation of mitochondria. This inhibition of

fusion may be the initial step leading to mitochondrial dysfunction, since inhibition of fusion

occurs prior to the induction of cell stress. The findings that mitochondrial dysfunction appears

to be central to PD pathology, suggest that mitochondria may be an excellent therapeutic target

for PD. Thus, the potential neuroprotective function of a regulator of mitochondrial function,

known as SIRT3 was examined. In SH-SY5Y cells, over-expression of SIRT3 protected neurons

from degeneration associated with LRRK2 over-expression. The studies described in this thesis

provide evidence that multiple sub-cellular mechanisms converge to inhibit mitochondrial

function. Furthermore, mitochondrial dynamics which regulate mitochondrial function could be

a key mediator in the pathology associated with PD. The work herein suggests therapies which

target the mitochondria are likely to be successful in the treatment of PD.

Page 4: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

iv

Acknowledgments

I would first and foremost like to thank Dr. Joanne Nash for her constant guidance and

encouragment over the course of my PhD. My skills as an independent researcher have vastly

improved because of the lab environment you have provided. I would also like to thank my

committee members, Dr. Michelle Aarts, Dr. Janelle LeBoutillier and Dr. Mauricio Terebiznik

for giving me enormous insight into the finer details of my project. A special thank you goes to

Sherri Thiele for her support, friendship, many laughs and white girl dances over the years.

Vitali, Ruth, Kristin, I appreciate all of the little errands you ran for me, and especially the crazy

nights caused by the many drinks over lab dinners. Julie, Darren, Sam, Wajma, Suhail, Paul, and

Colin thank you for being excellent peers and for the stress relieving moments in the hallways.

Thank you to my loving parents for providing a stimulating childhood environment which

sparked my fondness of science, and showing interest in my work even though you had no clue

what I was talking about. Thank you to Kacy for being my personal mechanic, and Justin and

Matthew for being my personal punching bags. Do not worry I also appreciate the fun times

each of you have provided me over the years. To the boys, John, Ian, Daniel C., Daniel F. and

Mike, and the girls, Elena and Steph thank you for being great friends.

Page 5: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

v

Table of Contents

Acknowledgments ......................................................................................................................... iv  

Table of Contents ........................................................................................................................... v  

List of Tables ................................................................................................................................. ix  

List of Figures ................................................................................................................................ x

List of Abbreviaitons .................................................................................................................. xii

1   Introduction ............................................................................................................................... 2  

1.1   Parkinson’s disease etiology ................................................................................................ 2  

1.2   Genetics underlying Parkinson’s disease ............................................................................ 3  

1.2.1   α-synuclein .............................................................................................................. 4  

1.2.2   Parkin ....................................................................................................................... 6  

1.2.3   UCHL-1 ................................................................................................................... 7  

1.2.4   PINK1 ...................................................................................................................... 8  

1.2.5   DJ-1 ......................................................................................................................... 8  

1.2.6   LRRK2 ..................................................................................................................... 9  

1.2.7   ATP13A2 ............................................................................................................... 14  

1.3   Environmental toxins linked with Parkinson’s disease pathology .................................... 14  

1.4   Cell death mechanisms linked with PD pathology ............................................................ 15  

1.4.1   Mitochondrial dysfunction .................................................................................... 18  

1.4.2   Dysfunction of the ubiquitin-proteasome system .................................................. 19  

1.4.3   Lysosomal dysfunction .......................................................................................... 20  

1.4.4   Oxidative stress ...................................................................................................... 20  

1.4.5   Protein aggregation ................................................................................................ 21  

1.5   Interaction of cell death mechanisms ................................................................................ 22  

1.6   Mitochondrial dynamics .................................................................................................... 27  

Page 6: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

vi

1.6.1   Regulation of mitochondrial dynamics .................................................................. 30  

1.6.2   Function of mitochondrial dynamics ..................................................................... 31  

1.6.3   Mitochondrial dynamics and neurodegenerative disease ...................................... 31  

1.7   Disease modifying agents .................................................................................................. 32  

1.7.1   RGMa .................................................................................................................... 34  

1.7.2   SIRT3 ..................................................................................................................... 35  

1.8   Models of Parkinson’s disease .......................................................................................... 36  

1.8.1   In vitro models ....................................................................................................... 36  

1.8.2   In vivo models ........................................................................................................ 38  

1.9   Hypothesis and  aims .......................................................................................................... 41  

2   Mitochondrial Dysfunction Precedes other Sub-Cellular Abnormalities in an In Vitro Model Linked with Cell Death in Parkinson’s Disease ............................................. 45  

2.1   Introduction ....................................................................................................................... 45  

2.2   Materials and Methods ...................................................................................................... 47  

2.2.1   Materials ................................................................................................................ 47  

2.2.2   Cell culture ............................................................................................................ 48  

2.2.3   Cell viability assays ............................................................................................... 48  

2.2.4   Cell death assays .................................................................................................... 48  

2.2.5   JC-1 and lysosensor green assays .......................................................................... 49  

2.2.6   Proteasome sensor vector assay ............................................................................. 49  

2.2.7   Western blotting .................................................................................................... 49  

2.2.8   Statistical analysis .................................................................................................. 50  

2.3   Results ............................................................................................................................... 50  

2.3.1   Effect of toxin exposure on cell viability and cell death ....................................... 50  

2.3.2   Effect of toxin exposure on mitochondria, UPS and lysosomes ........................... 54  

2.3.3   Effect of toxins on ubiquitin levels ........................................................................ 63  

Page 7: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

vii

2.4   Discussion .......................................................................................................................... 66  

3   Development and Validation of a Screening Assay for the Evaluation of Putative Neuroprotective Agents in the Treatment of Parkinson’s Disease ..................................... 73  

3.1   Introduction ....................................................................................................................... 73  

3.2   Materials and Methods ...................................................................................................... 74  

3.2.1   Materials ................................................................................................................ 74  

3.2.2   Cell culture ............................................................................................................ 75  

3.2.3   Effect of toxins on cell viability and cell death ..................................................... 75  

3.2.4   Effect of putative neuroprotective agents on toxins .............................................. 76  

3.2.5   Statistical analysis .................................................................................................. 76  

3.3   Results ............................................................................................................................... 76  

3.4   Discussion .......................................................................................................................... 90  

4   Effect of Over-Expression of WT and Mutant LRRK2 on Mitochondrial Dynamics ..... 94  

4.1   Introduction ....................................................................................................................... 94  

4.2   Materials and Methods ...................................................................................................... 99  

4.2.1   Constructs .............................................................................................................. 99  

4.2.2   Cell culture ............................................................................................................ 99  

4.2.3   SDS-PAGE followed by Western blotting .......................................................... 100  

4.2.4   Expression profile of constructs .......................................................................... 100  

4.2.5   Mitochondrial morphology assay ........................................................................ 101  

4.2.6   Tetramethylrhodamine ethyl ester perchlorate (TMRE) assay ............................ 102  

4.2.7   Polyethylene glycol-induced (PEG) cellular fusion assay .................................. 102  

4.2.8   Mitochondrial fission assay ................................................................................. 103  

4.2.9   Statistical analysis ................................................................................................ 104  

4.3   Results ............................................................................................................................. 104  

4.3.1   Generation of ptracer-3xFLAG-LRRK2-mCherry construct .............................. 104  

Page 8: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

viii

4.3.2   Expression of LRRK2 in SH-SY5Y cells ............................................................ 112  

4.3.3   Effect of LRRK2 on mitochondrial morphology ................................................ 120  

4.3.4   Effect of LRRK2 on mitochondrial fusion and fission ........................................ 127  

4.4   Discussion ........................................................................................................................ 133  

5   Neuroprotective Actions of SIRT3 and RGM in Advanced In Vitro Models of Parkinson’s Disease .............................................................................................................. 138  

5.1   Introduction ..................................................................................................................... 138  

5.2   Materials and Methods .................................................................................................... 143  

5.2.1   SH-SY5Y cells .................................................................................................... 143  

5.2.2   Ventral mesencephalic primary cultures ............................................................. 144  

5.2.3   Nigro-striatal organotypic co-cultures ................................................................. 147  

5.2.4 Statistical analysis ................................................................................................ 149  

5.3   Results ............................................................................................................................. 149  

5.3.1   Assessment of neuroprotective effects of RGMa and SIRT3 .............................. 149  

5.3.2   Development of an in vitro α-synuclein model utilizing ventral mesencephalic primary cultures ................................................................................................... 159  

5.3.3   Development of a nigro-striatal organotypic co-culture model of PD ................ 167  

5.4   Discussion ........................................................................................................................ 177  

6   Summary and Future Directions ......................................................................................... 183  

7   References .............................................................................................................................. 188  

8   Appendices ............................................................................................................................. 212  

 

 

Page 9: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

ix

List of Tables

Table 1.1 Genes linked with familial PD .................................................................................. 4

Table 1.2 Agents tested by CINAPS for neuroprotective potential ........................................ 33

Page 10: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

x

List of Figures

Figure 1.1. Schematic diagram showing the LRRK2 gene structure. ......................................... 10  

Figure 1.2. Schematic diagram outlining genes and environmental toxins that induce

parkinsonian pathology. ................................................................................................................ 16  

Figure 1.3. Schematic diagram showing the interactions of various sub-cellular mechanisms. . 23  

Figure 1.4. Schematic diagram showing mitochondrial fission and fusion. . ............................... 28  

Figure 2.1. Effect of toxins on cell viability and cell death in SH-SY5Y cells. . ......................... 52  

Figure 2.2. Effect of compounds on mitochondrial membrane potential in SH-SY5Y cells. ..... 55  

Figure 2.3. Effect of toxins on proteasomal function in SH-SY5Y cells. . .................................. 58  

Figure 2.4. Effect of toxin exposure on lysosomal function. ....................................................... 61  

Figure 2.5. Western blots to show changes in ubiquitin levels following toxin exposure. ......... 64  

Figure 3.1. Effect of toxins on cell viability in SH-SY5Y cells. . ................................................ 78  

Figure 3.2. Effect of putative neuroprotective agents on toxin-induced decreases in cell viability.

. ...................................................................................................................................................... 82  

Figure 3.3. Effect of putative neuroprotective compounds on viability in SH-SY5Y cells. ....... 88

Figure 4.1. Genetic mutations and toxins that affect mitochondrial processes. .......................... 97

Figure 4.2. Generation of ptracer-mCherry construct. .............................................................. 106  

Figure 4.3. Generation of ptracer-3xFLAG-LRRK 2-mCherry construct. . ............................... 109  

Figure 4.4. Expression of wild-type LRRK2 and G2019S-LRRK2 in SH-SY5Y cells. ............ 113  

Figure 4.5. Wild-type LRRK2 and mutant G2019S-LRRK2 expression time course. . ............ 115  

Page 11: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

xi

Figure 4.6. Expression of mCherry tagged wild-type LRRK2 and G2019S-LRRK2 in SH-SY5Y

cells. ............................................................................................................................................. 118  

Figure 4.7. Changes in mitochondrial morphology in SH-SY5Y cells. .................................... 121  

Figure 4.8. Effect of wild-type LRRK2 and G2019S-LRRK2 on mitochondrial function. ...... 124  

Figure 4.9. Effect of wild-type LRRK2 and mutant G2019S-LRRK2 on mitochondrial fusion.

..................................................................................................................................................... 128  

Figure 4.10. Effect of wild-type LRRK2 and G2019S-LRRK2 on mitochondrial fission. ....... 131

Figure 5.1. The potential role of RGMa and SIRT3 in preventing PD cell death mechanisms.141

Figure 5.2. Effect of RGMa on toxin-induced reductions of cell viability. ............................... 151  

Figure 5.3. Effect of SIRT3 in an SH-SY5Y cell model of PD. ................................................ 153  

Figure 5.4. Effect of SIRT3 in differentiated SH-SY5Y cells. .................................................. 155

Figure 5.5. Effect of SIRT3 in primary cultures. ....................................................................... 157

Figure 5.6. Determination of MAPK and GFAP expression in primary culture. ...................... 160  

Figure 5.7. Determination of TH and GIRK2 expression in ventral mesencephalic primary

culture. ........................................................................................................................................ 162  

Figure 5.8. Effect of mutant A53T α-synuclein expression in ventral mesencephalic primary

cultures. ....................................................................................................................................... 160  

Figure 5.9. Representative images showing axon outgrowth projecting from the SNc to STR.

..................................................................................................................................................... 169  

Figure 5.10. Expression of TH and GIRK2 in nigro-striatal cultures. ...................................... 171  

Figure 5.11. Effect of naphthazarin, PSI and rotenone on toxin-induced cell death. . ............... 173  

Figure 5.12. Effect of toxin exposure on TH expression in nigro-striatal oranotypic co-cultures.

..................................................................................................................................................... 175

Page 12: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

xii

List of Abbreviations

6-OHDA 6-hydroxydopamine

AAV Adeno-associated virus

AMCA Aminomethylcoumarin acetate

ATP Adenosine triphosphate

ANOVA Analysis of variance

BCS Bovine calf serum

BFP Blue fluorescent protein

CMA Chaperone-mediated autophagy

cAMP 3’ -5’-cyclic adenosine monophosphate

CINAPS Committee to identify neuroprotective agents in Parkinson’s disease

CMV Cytomegalovirus

DAPI 4',6-diamidino-2-phenylindole

DAT Dopamine transporter

DMEM Dulbecco’s modified Eagle’s medium

Dlp1 Dynamic-like protein 1

DNA Deoxyribonucleic acid

Drp1 Dynamin-related protein 1

Dsred2 Discosoma red 2

ECL Enhance chemiluminescence

Page 13: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

xiii

Fis1 Fission 1

GABA Gamma-aminobutyric acid

GIRK2 G protein-activated inward rectifier potassium channel 2

GFAP Glial fibrillary acidic protein

GFP Green fluorescent protein

GPI Glycosylphosphatidylinositol

GTP Guanosine triphosphate

HRP Horseradish peroxidase

LRRK2 Leucine-rich repeat kinase 2

MAO-B Monoamine oxidase-B

MAPK Mitogen-activated protein kinase

Mfn1 Mitofusin 1

Mfn2 Mitofusin 2

mPTP Mitochondrial permeability transition pore

mRNA Messenger ribonucleic acid

MPTP 1-methyl-4-pheny-1,2,3,6-tetrahydropyridine

mtDNA Mitochondrial deoxyribonucleic acid

mtGFP Mitochondrial green fluorescent protein

Opa1 Optic atrophy 1

PBS Phosphate buffered saline

Page 14: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

xiv

PD Parkinson’s disease

PEG Polyethylene glycol

PFA Parformaldehyde

PGC1-α Peroxisome proliferator-activated receptor gamma coactivator-1-alpha

PI Propidium iodide

PINK1 PTEN-induced kinase 1

PSD95 Postsynaptic density 95

PSI Z-Ile-Glu(OBut)-Ala-Leu-H

RGMa Repulsive guidance molecule a

ROS Reactive oxygen species

SDS-PAGE Sodium dodecyl sulphate-polyacrylamide gel electrophoresis

SIRT3 Sirtuin 3

SNc Substantia nigra pars compacta

SNCA synuclein a

STR Striatum

TH Tyrosine hydroxylase

TKL Tyrosine kinase like

TMRE tetramethylrhodamine ethyl ester perchlorate

TTBS Tween tris buffered saline

UCHL-1 Ubiquitin carboxy-terminal hydrolase 1

Page 15: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

xv

UPDRS Unified Parkinson’s disease rating scale

UPS Ubiquitin proteasome system

VTA Ventral tegmental area

Page 16: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

1

Chapter 1

Introduction

Page 17: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

2

1 Introduction

1.1 Parkinson’s disease etiology

Parkinson’s disease (PD) was first described by Dr. James Parkinson as a shaking palsy defined

by involuntary motion and loss of muscle power (Parkinson, 2002). PD is now characterized by

three cardinal symptoms, bradykinesia or slowed movement, resting tremor and muscle rigidity

which is responsible for postural instability (Tolosa et al., 2006). Patients frequently suffer from

non-motor symptoms which may begin before motor symptoms are displayed (Koller, 1992).

Such non-motor symptoms include depression, sleep disorders and dementia (Chaudhuri et al.,

2006). Both motor and non-motor symptoms become more severe as the disease progresses.

PD is the second most common neurodegenerative disease next to Alzheimer’s disease. The

cause of PD is elusive and likely variable between patients. In idiopathic PD, interactions

between genes and environment are believed to be involved. Mutations in nine proteins have

been linked with familial PD. No matter what the type of PD, risk factors include exposure to

pesticides, positive family history and old age (Massano and Bhatia, 2012). The worldwide

prevalence of PD in patients over 65 years of age is 1.8% (de Rijk et al., 2000). The average age

of onset is 65 years of age with incidence increasing from 0.6% at age 65-69 years to 2.5-3.5% at

age 85-89 years (de Rijk et al., 2000; Coelho and Ferreira, 2012). Early onset PD is defined as

occuring in patients under the age of 50, and has an incidence rate of 1.5/100 000 persons

(Wickremaratchi et al., 2009). While less prevalent than late-onset PD, symptoms of early-onset

PD are very similar to late-onset PD (Wickremaratchi et al., 2009).

Page 18: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

3

PD arises from degeneration of the dopaminergic nigro-striatal terminals within the basal ganglia

pathway (Hornykiewicz, 1966; Obeso et al., 2000). There is severe loss of tyrosine hydroxylase

immunoreactive neurons from the ventrolateral portion of the substantia nigra pars compacta

(SNc). Approximately 50% of dopaminergic cells within the SNc have degenerated before

clinical symptoms are present (Murray et al., 1995). However, sub-clinical manifestations are

believed to be caused by pathology that affects non-motor brain areas in early stages of the

disease (Braak et al., 2003). Brain pathology is thought to begin in the dorsal IX/X motor

nucleus and/or adjoining intermediate reticular zone, which then advances into brain stem nuclei

and then extends into cortical areas (Braak et al., 2003). Patients typically do not suffer motor

inabilities prior to being diagnosed with PD however, once diagnosed, motor impairments

become more prominent. A patient’s quality of life is severely affected once diagnosed, but is

improved when medication is prescribed (Den Oudsten et al., 2011). Eventually motor

impairments become so severe that palliative care is necessary. The main treatment for PD is

dopamine replacement therapies which reduce symptoms (Olanow, 2008). L-dopa is the most

effective anti-parkinsonian treatment to date, but is often associated with debilitating side effects

(Tuite and Riss, 2003; Olanow et al., 2004). The ELLDOPA study indicates that in patients

taking L-dopa, 16% develop dyskinesia and 20% have motor fluctuations just nine months after

beginning treatment (Fahn et al., 2004; Stocchi et al., 2008).

1.2 Genetics underlying Parkinson’s disease

Although only 10% of PD cases arise from genetic abnormalities, these cases have helped

scientists understand the cell death mechanisms underlying idiopathic PD (Cannon and

Greenamyre, 2012). There are as many as seventeen genes suspected to be linked with PD,

however most are extremely rare or are questionable causative factors of PD (Houlden and

Page 19: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

4

Singleton, 2012). Most of these genes have an autosomal dominant mode of inheritance. Each

gene is designated the name PARK and given a name according to its genetic locus, but is

commonly referred to by its gene product (Table 1.1).

Table 1.1: Genes linked with familial PD.Designation Locus Gene InheritancePARK1 4q21-22 SNCA: misense mutation ADPARK2 6q25.2-27 Parkin ARPARK3 2p13 ? ADPARK4 4q21-22 SNCA: multiplication ADPARK5 4p14 UCHL-1 ADPARK6 1p35-1p36 PINK1 ARPARK7 1p36 DJ-1 ARPARK8 12p11.2-q13.1 LRRK2 ADPARK9 1p36 ATP13A2 AR

Abbreviations: AD, autosomal dominant; AR, autosomal recessive

Adapted from: (Hampshire et al., 2001; von Bohlen und Halbach et al., 2004; Poulopoulos et al., 2012)

Each gene is listed in choronological order by its PARK designation.

1.2.1 α-synuclein

α-synuclein is a member of a 15-20 kDa synuclein family of proteins which consist of three

members: α, β, and γ. All three proteins are expressed throughout the brains of humans and

rodents, but it is unclear whether the substantia nigra and striatum contain the highest or lowest

levels of α-synuclein (Solano et al., 2000; Rockenstein et al., 2001). α-synuclein is a soluble

cytoplasmic protein that is ubiquitously expressed throughout the body. The highest expression

of α-synuclein in the body is found in the liver and testes. In neurons it is predominately found

in synaptic terminals where it associates with synaptic vesicles (Cookson, 2005). The function

of α-synuclein was largely unknown at the time of its discovery as a causative factor in PD.

Page 20: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

5

More recently it has been implicated in turnover of synaptic vesicles, molecular chaperone

activity, golgi trafficking, regulation of dopamine transporter function and mitochondrial fusion

(Cooper et al., 2006; Vogiatzi et al., 2008; Kamp et al., 2010; Nemani et al., 2010; Lundblad et

al., 2012).

Missense mutations in SNCA were identified as the first genetic cause of PD (Polymeropoulos et

al., 1997). The first identified missense mutation was A53T, which was subsequently followed

by the discovery of A30P and E46K mutations. Patients with missense mutations have early

disease onset combined with severe parkinsonian symptoms (Golbe et al., 1990). SNCA

multiplications were soon found to be a causative factor in familial PD. Gene duplications and

triplications in SNCA have variable disease penetrances (Poulopoulos et al., 2012). SNCA

triplication leads to early onset PD with dementia, while duplication causes late onset PD with

dysautonomia (Poulopoulos et al., 2012). Studies suggest multiplication of SNCA increases the

severity of PD symptoms in a dose dependent manner (Singleton and Gwinn-Hardy, 2004).

Post-mortem examinations of brain tissue from PD patients reveal the presence of Lewy bodies,

which are large protein aggregates which form when misfolded α-synuclein oligomerizes and

then forms fibrils which then aggregate. Brain areas typically affected by Lewy body pathology

include the brain stem (gigantocellular reticular nucleus and caudal raphe nuclei) and cerebral

cortex, as well as the SNc (Braak et al., 2003; Farrer et al., 2004). Missense mutations and

multiplications increase the propensity for α-synuclein to fibrilize and form Lewy bodies (Rochet

et al., 2004). While both misense mutations and gene multiplications lead to fibril formation,

other factors such as oxidative/nitrative stress, posttranslational modifications and incubation

with metals can induce fibril formation (Lee and Trojanowski, 2006; Savitt et al., 2006). No

Page 21: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

6

matter what the pathology underlying PD, all forms express α-synuclein-positive LBs with the

exception of PD caused by mutations in Parkin.

1.2.2 Parkin

The human Parkin gene encodes a 465 amino acid RING-finger containing protein that has E3

ubiquitin ligase activity (von Bohlen und Halbach et al., 2004; Dodson and Guo, 2007). Parkin

is mainly localized to the cytosol and endoplasmic reticulum, however small amounts are

localized on the cytoplasmic surface of the outer membrane of mitochondria (Shimura et al.,

1999). It is highly expressed in the substantia nigra, striatum, hippocampal formation and

pallidal formation (Zarate-Lagunes et al., 2001). Aside from its role in attachment of ubiquitin to

proteins destined for degradation, Parkin is important for the regulation of mitochondrial

homeostasis. Indeed, Parkin plays a crucial role with PINK1 to control mitochondrial function

by directing mitochondrial dynamics (Kane and Youle, 2011).

Mutations in Parkin cause autosomal recessive juvenile parkinsonism, which has the earliest

onset. It is characterized by dopaminergic cell death and the absence of Lewy bodies (von

Bohlen und Halbach et al., 2004). Some rare cases occur within the first decade of life, while the

majority occur before the age of 40 (Lucking et al., 2000). The Parkin gene is the only gene

linked to autosomal recessive juvenile parkinsonism (Kitada et al., 1998). Despite misense

mutations and duplications to Parkin, autosomal recessive early-onset PD still occurs, however,

it has been shown that heterozygous mutations in Parkin increase the susceptibility to late-onset

PD (Oliveira et al., 2003). Moreover, this increased susceptibility can lead to the presence of

Lewy body pathology (Farrer et al., 2001).

Page 22: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

7

1.2.3 UCHL-1

Ubiquitin carboxy-terminal hydrolase 1 (UCHL-1) is a deubiquitinating enzyme that plays a role

in the ubiquitin-proteasome system (UPS) protein degradation pathway. UCHL-1 possesses

carboxy hydrolase activity which breaks peptide bonds between ubiquitin monomers so they may

be reused in the UPS pathway. While UCHL-1 is mainly located in the cytoplasm, there is a

significant proportion that resides within synaptic vesicles (Liu et al., 2003). UCHL-1 mRNA is

widely expressed in the brain, however a study employing in situ hybridization revealed the

highest expression is in the substantia nigra (Solano et al., 2000). More recently UCHL-1 was

discovered to have ATP-independent dimerization-dependent ubiquitin ligase activity and

regulate the degradation of free ubiquitin monomers (Osaka et al., 2003; Healy et al., 2004).

The discovery of a mutation in a German family with a history of PD first linked UCHL-1 with

familial PD. A missense mutation in the UCHL-1 gene changes isoleucine to a methionine at the

amino acid 93 of the fourth exon (Leroy et al., 1998). Symptoms of PD induced by UCHL-1 are

similar to idiopathic PD, however the age of symptom onset occurs earlier (Leroy et al., 1998).

UCHL-1 appears to be found in Lewy bodies, however, this finding is controversial since the

Lewy bodies used in the study did not originate from PD patients (Lowe et al., 1990). The

involvement of UCHL-1 in PD is contentious since mice having UCHL-1 loss of function

mutations fail to present neuronal degeneration in the substantia nigra (Saigoh et al., 1999).

Moreover, I93M mutations only cause a partial loss of UCHL-1 hydrolase activity (Nishikawa et

al., 2003). It is likely that an interplay between I93M mutations and environment leads to PD

onset.

Page 23: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

8

1.2.4 PINK1

The PTEN-induced kinase 1 (PINK1) gene encodes a 581 amino acid protein that is highly

expressed in the hippocampus, substantia nigra and cerebellum (Blackinton et al., 2007). The

gene contains a mitochondrial targeting sequence and a C-terminal serine-threonine kinase

domain (Kawajiri et al., 2011). Although PINK1 is a mitochondrial membrane protein whose

kinase domain is located in the outer membrane, studies have shown that part of PINK1 is

accessible from the cytosol (Haque et al., 2008; Zhou et al., 2008a). PINK1 is believed to

phosphorylate proteins linked with PD such as Parkin and HtrA2 that are involved in

mitochondrial function (Plun-Favreau et al., 2007; Sha et al., 2010). Another role of PINK1

includes promoting autophagy in SH-SY5Y cells by interacting with the pro-apoptotic protein

Beclin-1(Michiorri et al., 2010).

Mutations in the PINK1 gene lead to autosomal recessive early-onset PD associated with Lewy

body pathology (Samaranch et al., 2010). PINK1 is the second most prevalent cause of familial

PD having a prevalence of 1-8% in familial and early onset PD (Michiorri et al., 2010). Several

mutations occur in the serine-threonine kinase domain suggesting the pathological effects are due

to a loss of kinase activity. Gene expression levels affect penetrance since the mean age of onset

is higher in patients with heterozygous mutations than those with homozygous mutations

(Kumazawa et al., 2008). Moreover, it has been suggested that a heterozygous mutation may

make one more susceptible to PD (Klein et al., 2007).

1.2.5 DJ-1

DJ-1 is a member of the ThiJ/PfpI super family of proteins and is widely expressed in the brain

(Bonifati et al., 2003). It is expressed in the cytoplasm, intermembrane space and matrix of

mitochondria and can translocate from the intermembrane space to the outer membrane during

Page 24: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

9

periods of oxidative stress (Canet-Aviles et al., 2004). The biological function of DJ-1 remains

unknown, however it is believed to play a role in oxidative stress and mitochondrial function.

Indeed, in vitro studies reveal that DJ-1 prevents oxidative stress and restores mitochondrial

complex activity in neuroblastoma (Miyazaki et al., 2008).

Mutations in the DJ-1 gene are the least common cause of autosomal recessive parkinsonism,

comprising of only 1-2% of early-onset PD cases (Bonifati et al., 2003). There are a total of 11

different mutations in DJ-1 which include missense, truncating, splice site mutations and large

deletions (Bonifati et al., 2004). All of which suggest that loss of function of DJ-1 is the likely

cause of pathogenicity in PD (Abou-Sleiman et al., 2004). Unlike other recessive forms of PD,

the majority of patients with PD linked to DJ-1 are in their early 30s (Bonifati et al., 2004).

1.2.6 LRRK2

Leucine-rich repeat kinase 2 (LRRK2) is a large protein consisting of 2527 amino acids and is a

member of the ROCO family of proteins. LRRK2 is comprised of several domains including an

ankyrin repeat region, Roc GTPase domain, leucine-rich repeat domain, COR domain, kinase

domain of the tyrosine kinase like (TKL) domain and WD40 domain (Fig. 1.1) (Manning et al.,

2002; Bosgraaf and Van Haastert, 2003; Mata et al., 2006). These domains allow it to bind

various proteins from transcription factors to signaling proteins (Andrade et al., 2001). LRRK2

is an interesting protein in that it contains both a kinase and GTPase domain. However, it is

unclear if these domains play distinct roles or are supportive of each other. Members of the TKL

family are activated by small GTPases, suggesting LRRK2 activity might be regulated by its

GTPase domain (Bosgraaf and Van Haastert, 2003; Korr et al., 2006).

Page 25: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

10

Figure 1.1. Schematic diagram showing the LRRK2 protein structure. LRRK2 is a large

protein comprised of 2527 amino acids and various functional domains. The locations of

common pathogenic mutations are highlighted in red. Abbreviations: ANK: ankyrin repeat

region, COR: C-terminal of Ras, LRR: leucine-rich repeat domain, Roc: Ras of complex GTPase

domain.

Page 26: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

11

G2019SI2020TY1699C

R1441CR1441G

2527  aa

Page 27: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

12

In-situ hybridization experiments reveal the expression of LRRK2 mRNA in dopaminergic

neurons within the SNc (Higashi et al., 2007). LRRK2 mRNA was also detected in the cerebral

cortex and caudate putamen (Galter et al., 2006; Higashi et al., 2007). In neurons, LRRK2 is

mainly localized in perikarya and dendritic and axonal processes (Higashi et al., 2007). This is

also consistent with the localization of LRRK2 in cultured cells (Smith et al., 2005c). LRRK2

localizes to membranous and vesicular structures including lysosomes, mitochondria, transport

vesicles and endosomes (Biskup et al., 2006). Interestingly, 10% of LRRK2 exists on the outer

mitochondrial membrane and closely associates with mitochondrial proteins in this vicinity

(West et al., 2005).

Mutations in LRRK2 are the most common genetic cause of familial and sporadic PD. In certain

populations up to 40% of PD cases can be attributed to LRRK2 mutations (Lesage et al., 2006;

Ozelius et al., 2006). There are at least 20 known mutations in LRRK2 that are linked to

autosomal dominant PD (Mata et al., 2006). Among these, the most common mutant variant is

G2019S, while the more infrequent R1441C, R1441G, and I2020T mutations have been found in

a small number of patients (Kett and Dauer, 2012). These mutations vary in their position on the

LRRK2 gene, where I2020T and G2019S occur in the kinase domain, R1441C and R1441G

occur in the GTPase domain, and Y1699C occurs in the COR domain (Mata et al., 2005).

Moreover, there is varying pathology associated with each mutation. G2019S carriers typically

have Lewy body pathology and exhibit a late-onset phenotype that is indistinguishable from

those with idiopathic PD (Aasly et al., 2005; Ross et al., 2006). R1441C substitutions are linked

with synucleinopathy, tauopathy and neuronal loss in the substantia nigra (Zimprich et al., 2004).

Patients with Y1699C substitutions display neuronal loss and nuclear ubiquitin inclusions (Mata

et al., 2006). The G2019S mutation causes a toxic gain of function, whereby enhanced kinase

activity induces cell death (West et al., 2005; Greggio et al., 2006; Smith et al., 2006). Although

Page 28: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

13

a toxic gain of function is conferred by G2019S mutations, it appears other mutations in the same

domain do not consistently show increases in kinase activity due to a gain of function (Jaleel et

al., 2007; West et al., 2007).

The normal function of LRRK2 is largely unknown, but studies suggest it is involved in vesicle

trafficking, neurite outgrowth, mitochondrial function and apoptosis (Plowey et al., 2008; Ho et

al., 2009; Parisiadou et al., 2009; Mortiboys et al., 2010). Previous studies reveal the association

of LRRK2 with lipid rafts, and interestingly, it is suggested to regulate vesicle trafficking

(Hatano et al., 2007). Indeed, in cortical neurons where LRRK2 expression has been silenced,

vesicle redistribution and altered vesicle kinetics occur (Piccoli et al., 2011). Moreover,

transgenic mice expressing G2019S mutations experience lower striatal dopamine content,

dopamine release and uptake (Li et al., 2010). Similarly, primary cultures harbouring LRRK2

mutations display shorter neurite length and less branching (MacLeod et al., 2006).

Modifications to neurite outgrowth and branching may result from phosphorylation of the ezrin,

radixin and moesin proteins by LRRK2 (Parisiadou et al., 2009). Furthermore, a study in

differentiated SH-SY5Y cells suggests neurite shortening caused by LRRK2 is associated with a

dysregulation of autophagic processes (Plowey et al., 2008). Strong evidence for the

involvement of LRRK2 in mitochondrial function comes from a study where patients having

G2019S mutations have mitochondria with reduced membrane potential and ATP production

(Mortiboys et al., 2010). While LRRK2 has been suggested to play a role in apoptosis, it is

likely that LRRK2 induced mitochondrial dysfunction activates apoptosis. However, a study

utilizing primary cultures reveals the extrinsic cell death pathway, rather than the

mitcochondrially mediated cell death pathway, is activated by LRRK2 through interactions with

a death adapter FAS-associated protein (Ho et al., 2009).

Page 29: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

14

1.2.7 ATP13A2

ATP13A2 encodes a lysosomal protein that is a member of the P-type ATPase superfamily

(Ramirez et al., 2006). The function of ATP13A2 remains elusive, however it is believed to be

involved in lysosomal function and protein degradation (Usenovic et al., 2012). Real-time PCR

experiments demonstrate that ATP13A2 mRNA is highest in the ventral midbrain and lowest in

the cerebellum (Ramirez et al., 2006). Cellular expression of APT13A2 is confined to lysosomal

vesicles where it co-localizes with lysosomal membrane proteins, LAMP1 and LAMP2 (Ramirez

et al., 2006). Interestingly, restoration of ATP13A2 expression increases lysosomal function and

prevents accumulation of α-synuclein (Usenovic et al., 2012).

Mutations in ATP13A2 genes are associated with Kufor-Rakeb Syndrome which is a juvenile,

early-onset parkinsonism with pyramidal degeneration and dementia (Park et al., 2011).

Frameshift mutations in ATP13A2 genes remove three or six C-terminal transmembrane

domains which cause a loss of function of the protein (Ramirez et al., 2006). In addition to

homozygous mutations, heterozygous and compound heterozygous mutations can lead to onset

of parkinsonism (Park et al., 2011).

1.3 Environmental toxins linked with Parkinson’s disease pathology

Prolonged exposure to various environmental toxins is linked with sporadic PD, and while toxin

exposure alone is thought to cause PD, it is also believed that a combination of environmental

and genetic factors lead to disease onset. Toxins which are implicated in sporadic PD onset

generally affect sub-cellular areas by the same mechanisms as those found in familial PD.

Indeed, toxins such as 1-methyl-4-pheny-1,2,3,6-tetrahydropyridine (MPTP), rotenone, maneb

and paraquat, block mitochondrial function by inhibiting electron transport chain activity. MPTP

Page 30: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

15

and rotenone inhibit mitochondrial complex 1, while maneb blocks mitochondrial complex 3

activity, and paraquat causes non-selective inhibition of all the mitochondrial complexes (Zhang

et al., 2003; Richardson et al., 2005; Gomez et al., 2007; Sherer et al., 2007). Other than the

primary effect of rotenone, a secondary effect includes oxidative stress and α-synuclein

aggregation (Sherer et al., 2003a; Testa et al., 2005). Another toxin used in the study of PD is 6-

hydroxydopamine (6-OHDA). Although 6-OHDA is not an environmental toxin, it is has been

utilized in experiments to replicate the pathological and biochemical characteristics of PD. Such

characteristics include the induction of oxidative stress through intracellular and extracellular

mechanisms (Blum et al., 2001; Hanrott et al., 2006). Similarly, exposure of neurons to high

concentrations of dopamine hydrochloride is commonly used as a toxicant to emulate

pathological mechanisms in PD (Yong-Kee et al., 2012).

1.4 Cell death mechanisms linked with Parkinson’s disease pathology

Numerous sub-cellular deficits are believed to be the underlying cause of neurodegeneration

displayed in PD. Studies reveal that malfunction of the mitochondria (Bohlen und Halbach,

2004; Lannuzel et al., 2003; Sherer et al., 2007), the UPS (Ardely et al., 2004; Caneda-Feron et

al., 2008) and the lysosome (Cooper et al., 2006) appear to be sources triggering cell death linked

with PD. Moreover, genetic mutations and environmental toxins specifically target these

organelles (Fig. 1.2). In addition, reactive oxygen species (ROS) production and protein

aggregates are traditionally regarded as key mediators of dopaminergic neuron toxicity. Such

protein aggregates include Lewy bodies that are primarily composed of both α-synuclein and

ubiquitin.

Page 31: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

16

Figure 1.2. Schematic diagram outlining genes and environmental toxins that induce

parkinsonian pathology. All mutated genes are synthesized within the nucleus. All

environmental toxins that are known to cause parkinsonism inhibit mitochondrial function.

Arrows point to the organelle or molecule that is affected by each known trigger.

Page 32: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

17

Page 33: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

18

1.4.1 Mitochondrial dysfunction

Mitchondria are classically known as “powerhouses” of the cell because they supply a large

portion of ATP for cellular processes. However, mitochondria play many roles including

initiating apoptosis, integrating stress response pathways and regulating cellular metabolism. In

neurons, mitochondria are critical to synaptic transmission since mitochondrial density

influences the number and plasticity of spines and synapses (Li et al., 2004). Mitochondria are

comprised of an inner and outer membrane that contains the intermembrane space. A series of

intermembrane proteins or mitochondrial complexes located within the inner membrane form the

electron transport chain. Here, ATP is generated through a series of reactions mediated by

mitochondrial complexes 1-5.

Mitochondrial complexes that reside within the inner membrane of mitochondria are targets for

the different environmental toxins that have been implicated in PD pathogenesis; however

complex 1 is primarily targeted. Rotenone exhibits high affinity binding for the NUO D site

which sits at the junction between the membranous and peripheral domains of mitochondrial

complex 1 (Darrouzet et al., 1998). Binding of rotenone prevents electron flow through

mitochondrial complex 1 which inhibits ATP production by the electron transport chain and

reduces membrane potential (Ramsay et al., 1991). Moreover, studies suggest rotenone enhances

reactive oxygen species production by mitochondria, which further exacerbates the toxic effects

of rotenone (Sherer et al., 2003b). These changes within the mitochondria ultimately lead to the

activation of apoptosis and neurodegeneration in cells within the SNc. Interestingly, mutations

in genes which appear to affect mitochondria do not affect mitochondrial complexes. Rather,

mutations in genes such as DJ-1 induce mitochondrial oxidative stress by preventing

Page 34: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

19

mitochondrial uncoupling, while mutations in other genes such as Parkin and PINK1 affect

mitochondrial dynamics (Guzman et al., 2010; Kane and Youle, 2011).

1.4.2 Dysfunction of the ubiquitin-proteasome system

The UPS is one of the key protein degradation pathways within the cell. The UPS degrades

various short-lived proteins, including misfolded, mutant and oxidatively damaged proteins, but

not membranous and extracellular proteins (Betarbet et al., 2005). Another role of the UPS is to

maintain mitochondrial protein quality control by preventing the accumulation of intermembrane

space proteins which can be deleterious if left unregulated (Radke et al., 2008). Aside from

being part of the UPS mediated degradation pathway, ubiquitin plays various roles in DNA

repair, and intracellular signalling (Ikeda and Dikic, 2008). The 20S proteasome is constructed

from four heptameric rings that form a hollow core that contains catalytic sites. Chymoptrypsin–

like, trypsin-like and peptidylglutamyl-peptide catalytic sites are located within the two inner

rings to prevent indiscriminate degradation of proteins (Betarbet et al., 2005). A 19S subunit

resides on either end of the 20S proteasome to form the 26S proteasome. The 19S subunit

confers ATPase and ubiquitin ligase function to the 26S proteasome to regulate the entrance of

proteins into the catalytic core (Smith et al., 2005a). The 19S subunit has three functions which

include opening the 20S subunit so proteins can gain access to the core, unfolding ubiquitin

tagged proteins before entrance into the catalytic core and removing ubiquitin subunits from

tagged substrates. Enzymes such as UCHL-1 and Parkin respectively are involved in recycling

ubiquitin monomers and adding ubiquitin to proteins that will be degraded. The genes of such

enzymes become mutated and cause as much as a 50% decrease in UPS activity which leads to

the formation of protein aggregates. However, evidence also suggests that these mutations

promote aggregate formation which then inhibits the proteasome since mutated UCHL-1 and

Parkin were found in these aggregates (Ardley et al., 2003; Ardley et al., 2004). It appears UPS

Page 35: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

20

inactivity can promote protein aggregation or UPS inactivity can result from the presence of

aggregated proteins.

1.4.3 Lysosomal dysfunction

The lysosome plays a major role in macroautophagy, microautophagy and chaperone-mediated

autophagy (CMA). In each of these three processes, the lysosome is responsible for degrading

stable long-lived proteins, damaged proteins, protein complexes, protein aggregates and

organelles. However, in CMA, lysosomal degradation is more specific in that a molecular

chaperone can only escort proteins with a lysosomal targeting motif to the lysosome for

degradation. The lysosome also compensates for the inability of the proteasome to breakdown

large membrane proteins and aggregates that cannot enter the proteasomal core (Cuervo et al.,

2004). Recently, mutations in the gene encoding ATP13A2, a lysosomal membrane protein,

were found in a small cohort of patients with PD (Ramirez et al., 2006). Mutations in the gene

encoding ATP13A2 localizes it to the endoplasmic reticulum where it is subsequently degraded

by the UPS (Park et al., 2011). Moreover, it is possible that this places additional stress on the

UPS pathway leading to protein aggregation (Ramirez et al., 2006). An alternative consequence

of lysosomal dysfunction due to mutations in the ATP13A2 gene is impaired lysosomal

degradation capacity which promotes aggregation of α-synuclein (Dehay et al., 2012; Usenovic

et al., 2012).

1.4.4 Oxidative stress

In the late 1980s and early 1990s, oxidative stress was viewed as the primary source of

pathology in PD because of the involvement of dopamine dysregulation and dopamine

metabolism in dopaminergic neurons (Goldstein et al., 1988; Brannan et al., 1990; Jenner, 1992).

However, oxidative stress is involved in a variety of dysfunctional mechanisms and can be

Page 36: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

21

regarded as both a primary and secondary source of pathology. Oxidative stress is mainly caused

by ROS and to a lesser degree, reactive nitrogen species. ROS arise from various sources

including dopamine metabolism, iron dysregulation, superoxide production, and lowered

antioxidant defenses (Jenner, 2003; Chen et al., 2008). Autooxidation of dopamine forms

quinone and semiquinone, and more importantly, the breakdown of dopamine by monoamine

oxidase A can produce H2O2 (Halliwell, 2006). If antioxidant molecules are unable to

metabolize H2O2, iron can react with H2O2 to produce the highly reactive hydroxyl radical

(Jenner and Olanow, 1998). It appears that chemical reactions producing ROS are linked, so that

one reaction may enhance another. ROS production is not only dependent on intracellular

dopamine alone, but extracellular dopamine which is transported into the cell can exacerbate

ROS production (Hanrott et al., 2006). Mutations in the gene encoding DJ-1 appear to be the

only cause of familial PD that is directly associated with oxidative stress. Although an exact

mechanism of DJ-1 action has yet to be delineated, there is substantial evidence that DJ-1 plays a

role in oxidative stress (Taira et al., 2004; Miyazaki et al., 2008).

1.4.5 Protein aggregation

The presence of Lewy bodies in post-mortem brain samples from PD patients has made protein

aggregates the defining feature of PD pathology. Protein aggregates are comprised of many

protein fibrils that are formed from several oligomers with β-pleated sheet conformation.

Oligomer formation is initiated by the misfolding of mutant or modified proteins (Lee and

Trojanowski, 2006). The presence of mutated α-synuclein in Lewy bodies suggests that

mutations in the α-synuclein gene play a role in PD pathology (Zhou et al., 2008b). Evidence

suggests A30P mutations accelerate oligomer formation while A53T enhances fibril formation

(Conway et al., 1998; Conway et al., 2000). The presence of oxidatively modified α-synuclein in

Lewy bodies suggests other factors like oxidative stress influence protein aggregation (Smith et

Page 37: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

22

al., 2005b; Gao et al., 2008). Moreover, wild-type α-synuclein without any oxidative

modifications can fibrilize to form protein aggregates (Volpicelli-Daley et al., 2011). Given

various forms of α-synuclein are found in aggregates, it is possible that overexpression of any α-

synuclein variant promotes aggregate formation by a concentration dependent manner. Although

some of the processes involved in protein aggregation are thought to be toxic to cells, it is

unclear whether the final products of protein aggregation serve as a neuroprotective strategy

where toxic proteins are sequestered to prevent them from affecting normal processes. Indeed,

post-mortem brain samples from asymptomatic patients exhibit Lewy bodies and neurons

containing Lewy bodies appear to be more healthy than their non-Lewy body containing

counterparts (Tompkins and Hill, 1997). Although the mechanisms of aggregate toxicity is

unclear, it is plausible that aggregation of proteins, like α-synuclein, prevents these proteins from

performing their normal functions within the cells, which ultimately affects neuronal function

(Lee and Trojanowski, 2006). Further evidence that Lewy bodies have neuroprotective function

is shown by the absence of Lewy bodies in patients displaying early-onset PD caused by

mutations in Parkin.

1.5 Interaction of cell death mechanisms

While the previously mentioned pathological mechanisms act independently to produce

pathology, it is believed that interactions between each can produce or even exacerbate disease

onset (Fig. 1.3). It appears that the interaction between each pathological mechanism could be

direct, however further elucidation of these mechanisms may reveal indirect interactions where

one dysfunctional mechanism induces another pathological mechanism by first acting upon an

intermediary mechanism.

Page 38: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

23

Figure 1.3. Schematic diagram showing the interactions of various sub-cellular

mechanisms. Mechanisms that are thought to become dysfunctional at later stages of cell stress

during PD. It is unknown if multiple sub-cellular mechanisms converge upon mitochondria and

if reciprocal interactions exist between them. Single-headed arrows indicate a unidirectional

interaction between sub-cellular domains. Double-headed arrows indicate a reciprocal

interaction between sub-cellular areas.

Page 39: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

24

Page 40: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

25

There is substantial evidence supporting the production of ROS by dysfunctional mitochondria.

Indeed, large amounts of ROS are by-products of the reactions mediated by mitochondrial

complexes of the electron transport chain (Lin and Beal, 2006). It also appears a link between

mitochondrial dysfunction and UPS inhibition exists since mitochondrial dysfunction at complex

1 promotes UPS inhibition. This is demonstrated by the accumulation of an exogenous protein

that is normally degraded by the UPS in neuroblastoma cells treated with rotenone (Yong-Kee et

al., 2012). The mechanism by which dysfunctional mitochondria affects the UPS is unclear, but

it is likely reduced ATP production prevents ubiquitin tagging and 19S cap opening (Hoglinger

et al., 2003a; Smith et al., 2005a). Moreover, it appears that a reciprocal interaction exists

between the two whereby UPS dysfunction can promote inactivity of mitochondrial complexes

(Sullivan et al., 2004). Studies suggest that protein quality control of mitochondria is regulated

by the UPS in order to prevent excessive accumulation of intermembrane space proteins and

collapse of the mitochondrial network (Radke et al., 2008). A link between mitochondrial

integrity and lysosome function is demonstrated by lysosome inhibition during the presence of

rotenone (Yong-Kee et al., 2012). Although there does not appear to be evidence of

mitochondrial dysfunction directly affecting lysosomes, it is possible that an indirect interaction

by way of UPS inhibition occurs.

Many studies have established a direct interaction of mechanisms between the UPS and

lysosome by demonstrating that inhibition of either system can affect the other. The lysosome

can compensate for the UPS when it is unable to efficiently degrade proteins (Rubinsztein,

2007). This interaction is supported by a study where multiphoton in vivo imaging of mouse

brain expressing α-synuclein reveals the autophagic-lysosome pathway becomes recruited when

UPS mediated degradation of α-synuclein is elevated (Ebrahimi-Fakhari et al., 2011). The UPS

can also compensate for lysosomal inhibition, as evidenced by the increase of ubiquinated

Page 41: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

26

proteins in mice lacking proteins necessary for autophagy (Hara et al., 2006). However, only

substrates intended for the lysosome that are small enough to enter the proteasomal core can be

degraded. Compensation for lysosomal inhibition comes at a cost however, since delayed

clearance of ubiquitinated proteins is observed when the lysosome is inhibited (Korolchuk et al.,

2009).

Protein aggregation is an end result of the inability of the UPS and lysosome to clear normal,

oxidatively modified and mutant proteins. It was traditionally thought that UPS and lysosome

dysfunction mediates the formation of aggregates containing ubiquitin and α-synuclein (Rideout

et al., 2001; McNaught et al., 2004; Komatsu et al., 2006). However, a reciprocal interaction

exists where aggregate formation decreases proteasome function (Bence et al., 2001).

Proteasome function is strongly inhibited by binding of α-synuclein aggregates to the 19S cap of

the proteasome (Snyder et al., 2003). Given the interactions and similarities between the UPS

and lysosome, it seems likely that protein aggregation should affect lysosome function as well.

A feature of the lysosome is to contain large aggregates from the cytosolic environment;

however some of the aggregates are insoluble, which would then lead to a failure of autophagy

(Cuervo et al., 2005).

ROS are by-products of many reactions and because of this, interactions with various sub-

cellular systems and proteins often occur. Frequent interactions with proteins combined with

failures in protein degradation pathways influence the formation of protein aggregates. Indeed,

α-synuclein is highly susceptible to oxidative modifications which stabilizes α-synuclein fibrils

and in turn promotes aggregate formation (Souza et al., 2000). Oxidative modification is not

limited to α-synuclein however, as other proteins like Parkin, aggregates once its cysteine rich

Page 42: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

27

domain is modified (LaVoie et al., 2007; Meng et al., 2011). Similarly, nitrative modification of

α-synuclein frequently occurs to induce aggregate formation (Stone et al., 2012).

1.6 Mitochondrial dynamics

Mitochondrial dysfunction is regarded as a classical mechanism causing cell death in PD.

Moreover, the first toxin-induced case of PD was caused by a mitochondrial inhibitor. Thus,

mitochondrial dysfunction may be a key mediator of disease pathogenesis in both sporadic and

familial cases. Mitochondrial dynamics consist of various processes that maintain the integrity

and function of mitochondria. Such processes include fission and fusion of mitochondria,

trafficking of mitochondria, and autophagy of mitochondria or mitophagy. Fission is when a

single mitochondrion divides into two mitochondria, while fusion is the process where two

mitochondria contact each other and fuse into one mitochondrion (Fig. 1.4).

Page 43: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

28

Figure 1.4. Schematic diagram showing mitochondrial fission and fusion. (a) Fis1 interacts

with Drp1 to promote self-association. Drp1 forms a spiral structure that wraps around

mitochondria during fission. Mitochondria are severed by Drp1 through a GTPase mediated

reaction. (b) Mfn1 and Mfn2 tether mitochondria together during outer mitochondrial membrane

fusion. Inner mitochondrial membrane fusion is then mediated by Opa1. GTP hydrolysis

initiates the reactions mediated by Mfn1, Mfn2 and Opa1. Abbreviations: Drp1: dynamin-

related protein 1, Mfn 1: Mitofusin 1, Mfn2: Mitofusin 2.

Page 44: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

29

Drp1

Fis1

a Fission

b Fusion

Mfn2

Mfn1

Opa1

Page 45: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

30

1.6.1 Regulation of mitochondrial dynamics

Large GTPase proteins mediate fission and fusion processes. Dynamin-related protein (Drp1)

and fission 1 (Fis1) mediate the mechanisms that orchestrate fission of mitochondria (Knott et

al., 2008). Drp1 is a cytosolic protein that contains an N-terminal GTPase domain. During

fission, Drp1 is believed to translocate from the cytosol to fission sites on mitochondria where it

self-associates to form spiral-like structures (Smirnova et al., 2001). These structures begin to

wrap around and constrict the mitochondria until the mitochondrial membrane is severed by a

GTPase dependent mechanism (Otera and Mihara, 2011). Fis1 is anchored to the outer

mitochondrial membrane with its N-terminus exposed to the cytoplasm. Studies suggest Fis1

plays a role in recruiting Drp1 to sites of fission on the mitochondria (Yoon et al., 2003). Three

large GTPases, mitofusin 1 (Mfn1), mitofusin 2 (Mfn2) and optic atrophy 1 (Opa1) mediate

mitochondrial fusion. Mfn1 and Mfn2 are anchored to the outer mitochondrial membrane with

their N-terminal GTPase and C-terminal coiled coil ends facing the cytoplasm. It is unclear how

Mfn1 and Mfn2 mediate outer membrane fusion, but they are believed to tether mitochondria

together in a GTPase dependent manner. Opa1 is situated in the inner mitochondrial membrane

with its GTPase domain facing the intermembrane space (Otera and Mihara, 2011). The process

of inner membrane fusion is unclear, but the GTPase domain of Opa1 appears to be essential

since mutations in this domain lead to mitochondrial fragmentation (Olichon et al., 2007). While

the proteins that regulate fission and fusion are controlled by GTP hydrolysis, it appears that the

activity of these proteins can also be regulated by phosphorylation by cyclin-dependent kinase 1

and cAMP-dependent protein kinase (Westermann, 2010).

Page 46: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

31

1.6.2 Function of mitochondrial dynamics

Mitochondria are constantly undergoing fission and fusion to adjust mitochondrial populations to

meet cellular demands. Processes such as axonal outgrowth and synaptic plasticity require

changes in mitochondrial content which is dependent on mitochondrial fission and fusion

(Morris and Hollenbeck, 1993; Li et al., 2004). Fission and fusion are antagonistic processes and

a delicate balance between the two must be maintained to ensure mitochondria are functional.

Indeed, damaged mitochondrial DNA is eliminated and depolarised mitochondrial membrane

potentials are restored upon completion of mitochondrial fusion. Fusion is also important for the

inheritance and maintenance of mtDNA, which in turn is key to the overall health of

mitochondrial populations (Rapaport et al., 1998). Mitochondrial fission is critical for synaptic

transmission between neurons since the proper function of synapses is reliant on functional

mitochondria at synaptic sites (Li et al., 2004; Ishihara et al., 2009). Fission is implicated in the

regulation of apoptotic processes in neurons since Drp1 is required for cytochrome c release and

caspase activation (Estaquier and Arnoult, 2007; Ishihara et al., 2009).

1.6.3 Mitochondrial dynamics and neurodegenerative disease

The functions performed by fission and fusion are critical to neuronal health which implies that

any perturbations to these processes may be linked with neurodegenerative disease. Several

neurodegenerative diseases are linked to aberrant mitochondrial dynamics. Mutations in Mfn2

cause Charcot-Marie-Tooth type 2A disease which affects sensory and motor neurons (Chen and

Chan, 2009). Dominant optic atrophy is caused by mutations in Opa1 and leads to degeneration

of retinal ganglion cells (Detmer and Chan, 2007). Interestingly, patients with Charcot-Marie-

Tooth type 2A disease display various symptoms including parkinsonism and psychiatric

disturbances (Verhoeven et al., 2006). Alterations to mitochondrial dynamics may be equally

Page 47: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

32

important in disorders such as PD where mitochondrial function is especially critical to the

health of nigral neurons.

1.7 Disease modifying agents

Presently, all treatments for PD are focused on treating symptoms of PD by using dopamine

replacement therapy, either by replacing lost dopamine in the form of the dopamine precursor L-

dopa or dopamine agonists, or by enhancing the half-life of remaining dopamine using mono

amine oxidase-B (MAO-B) inhibitors. Levodopa, rotigotine (dopamine agonist), selegiline and

rasagiline (MAO-B inhibitors) are all successful at reducing motor symptoms, however long-

term use is associated with debilitating side effects such as dyskinesias (Schapira, 2009). While

much research is focused on treating symptoms of PD, it would be much more beneficial to

patients if it were possible to stop or slow the progression of neurodegeneration by using a

neuroprotective agent. This would prevent PD progressing to more advanced stages where

patients become seriously disabled and palliative care is necessary. The Committee to Identify

Neuroprotective Agents in Parkinson’s disease (CINAPS) project was the first of its kind to

select a battery of neuroprotective agents to test in clinical trials (Heemskerk et al., 2002; Ravina

et al., 2003). Given the importance of various mechanisms such as oxidative stress and

mitochondrial dysfunction which ultimately cause apoptosis, it is conceivable that putative

neuroprotective agents should act on these pathways. Indeed, some of the various chemicals

tested by CINAPS are involved in the previously mentioned mechanisms (Table 1.2).

Page 48: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

33

Table 1.2: Agents tested by CINAPS for neuroprotective potential.Agent Mechanism of ActionCaffeine Adenosine A2A receptor antagonismCoenzyme Q10 Cofactor of electron transport chain; antioxidant propertiesCreatine Indirect antioxidant by enhancing energy transfer during periods of high energy demandEstrogen May act as an antioxidant through upregulation of Bcl-2, BDNF and GDNFGM-1 ganglioside UnknownMinocycline Inhibition of microglial activationNicotine Increases neurotrophic factorsNeuro-immunophilin A Immunophilin ligandSelegeline, Rasagiline MAO-B inhibitorRopinirole, Pramipexole Dopamine agonist with antioxidant properties

Adapted from: (Meissner et al., 2004; Hung and Schwarzschild, 2007)

Many of these agents were found to be neuroprotective in various rodent and primate models

before being considered for clinical trials, while others were tested in clinical trials without prior

testing in animal models (Meissner et al., 2004). Nicotine and caffeine were suggested to be

neuroprotective based on the low incidence rates of PD in humans that frequently consumed

them. Estrogen was selected as a putative neuroprotective agent because of the lower incidence

of PD in women, especially in women receiving estrogen replacement (Saunders-Pullman et al.,

1999). Coenzyme Q10 appears to be the most successful agent, having protective effects in

animal models and placebo-controlled clinical trials. Similarly, selegeline was found to be

beneficial when tested using the Unified Parkinson’s Disease Rating Scale (UPDRS). Indeed,

lower scores were given by patients receiving selegeline treatment, but these results were

questionable due to errors in clinical trial design (Hung and Schwarzschild, 2007). The effect of

creatine was assessed in a two year placebo-controlled clinical trial but protection was not

observed as UPDRS scores were not lowered (Bender et al., 2006). However, further study of

creatine is warranted as it was found that it could not be excluded as a possible neuroprotective

agent in a futility study (2006). It appears that compounds that are neuroprotective in animal

models are not always as successful in clinical trials. However, in cases such as with coenzyme

Page 49: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

34

Q10 and creatine, there was benefit in both animal models and clinical trials. Thus it seems there

is a disparity between the predictive capabilities of clinical trials, and improvements in clinical

trial designs are necessary to test the clinical relevance of putative neuroprotective agents.

Alternatively, improvements in cellular and animal models may be necessary so these models are

better reflective of the human disease.

1.7.1 RGMa

Repulsive guidance molecule A (RGMa) is a membrane-associated glycoprotein that was

originally isolated from chick optic tectum (Monnier et al., 2002). The C-terminal is anchored to

the plasma membrane by a glycosylphosphotidylinositol (GPI) domain and the N-terminal

contains a signal peptide sequence. A partial von Willebrand factor domain and RGD sequence

also exist to support membrane adhesion (Monnier et al., 2002). RGMa is part of a family of

RGM proteins (RGMa-c) with each having their own specific function in the central nervous

system. RGMa is localized to the soma in mature and immature neurons, while immature

neurons also contain RGMa in their axons (Brinks et al., 2004; Schwab et al., 2005). RGMa acts

as a guidance cue for retinal axons and induces the collapse of temporal growth cones during

neuronal development (Monnier et al., 2002). Other functions of RGMa include the ability to

regulate neuronal differentiation and prevent apoptosis through its interaction with neogenin, a

homologue of netrin-1 and DCC receptors. Studies reveal that neogenin is a dependence

receptor where activation of pro-apoptotic pathways normally occurs in the absence of a ligand

(Matsunaga and Chedotal, 2004; Matsunaga et al., 2004). Caspase-3 cleavage of neogenin

initiates these apoptotic signalling pathways, which are blocked in the presence of its ligand,

RGMa. Moreover, RGMa overexpression prevents pro-apoptotic signalling while RGMa siRNA

expression has no effect on cell survival (Matsunaga et al., 2004). Although RGMa has the

Page 50: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

35

ability to regulate axonal outgrowth and to modulate apoptotic pathways, surprisingly it has not

been studied in various disease models.

1.7.2 SIRT3

The novel family of protein deacetylases, sirtuins, consist of seven family members (SIRT 1-7),

each of which have shown efficacy in the treatment of a wide spectrum of diseases. Sirtuin1

(SIRT1) was the first member of the sirtuins to be extensively studied in mammals (Gan and

Mucke, 2008). Evidence suggests that SIRT1 elicits neuroprotective properties by its actions on

non-histone substrates in the nucleus (Lavu et al., 2008). Another member of the sirtuin family,

SIRT3, is less extensively studied; however, it is believed to be involved in mitochondrial

function. SIRT3 is encoded within the nucleus and then directed to the mitochondria by its N-

terminal mitochondrial localization sequence (Bao et al., 2010). Once inside the mitochondria,

SIRT3 becomes enzymatically active upon cleavage of its localization sequence by a protein

peptidase (Giralt and Villarroya, 2012). SIRT3 is primarily located in mitochondria. Studies

report SIRT3 is also localized in the nucleus and cytosol in its uncleaved form while the cleaved

form is localized in the mitochondria (Cooper and Spelbrink, 2008). Targets of SIRT3 include

acetyl-CoA, the mitochondrial permeability pore and mitochondrial complexes 1-5. Upon

regulation of the respiratory chain, SIRT3 influences energy production (Shimazu et al., 2010;

Verdin et al., 2010). The activation of these targets may be associated with SIRT3’s actions in

the cell’s stress response pathway. SIRT3 is also involved in mitochondrial biogenesis where it

mediates the effects of PGC-1α by acting as a downstream target (Kong et al., 2010). Although

SIRT3 plays many critical roles in mitochondrial function, and its family members are

efficacious in various diseases, most studies on SIRT3 have been performed in the heart, and

nothing is known about its therapeutic effect in neurons.

Page 51: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

36

1.8 Models of Parkinson’s disease

Various in vitro and in vivo models have been generated to replicate the mechanisms underlying

PD and the symptoms resulting from PD. Each model utilizes toxins or genetic manipulations to

induce parkinsonian features. Since each model does not fully replicate disease mechanisms or

symptoms, new models that incorporate these important aspects of PD will be invaluable for

therapeutic development. Although the present models lack certain characteristics of PD

mechanisms and symptoms, each have been useful in the development of therapeutics that are

currently available to treat PD.

1.8.1 In vitro models

In vitro models have been extremely valuable tools for determining the cellular mechanisms

underlying PD. Toxins have been applied to in vitro models to study idiopathic forms of PD,

while the insertion of exogenous DNA known to be mutated in individuals with PD into

individual cells has given insight into the mechanisms causing familial PD. Organotypic co-

cultures contain multiple cell types allowing for changes in one cell type to be directly compared

to changes in another cell type while they are in the same microenvironment. Such cultures can

be maintained for up to a few weeks in culture allowing the neurons to mature, which more

closely resembles neurons degenerating in PD. Other advantages of organotypic co-cultures

include neuronal and glial interactions, maintenance of neuronal connections and growth of

synapses while in culture (Lyng et al., 2007). More importantly, organotypic cultures express

dopamine, homovanillic acid and tyrosine hydroxylase from as little as 6 days to 17 days after

culturing (Lyng et al., 2007). Compared to other in vitro models, the production of organotypic

co-cultures is more complicated since slices of multiple brain areas must be obtained from

embryonic and post-natal rodents. Primary cultures have been employed in many studies to

Page 52: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

37

discover the biochemical pathways responsible for PD pathology (Hoglinger et al., 2003a;

Lannuzel et al., 2003). Synapses actively form between neurons in primary cultures, but the

intrinsic activity of synapses seen in the brain is not usually found in these cultures. Indeed,

these in vitro neurons were less spontaneously active and never fired in bursts, unlike in vivo

neurons (Rayport et al., 1992). While primary cultures benefit from having neurons and glia and

display the typical characteristics of mature neurons, such as TH and dopamine, generally very

few cells from the culture have these characteristics. However, the presence of dopamine

autoreceptors was identified by neuronal activity in response to quinpirole, a dopamine receptor

agonist (Cardozo, 1993). In vitro models such as cell lines have the advantage of being quickly

generated allowing reproducible effects to be shown. Even the differentiation of neuroblastoma

to neuronal-like cells can take place in a time frame as short as 72 hours, allowing for high-

throughput findings that can be inferred to actual neurons. Disadvantages of cell lines include

being dissimilar from actual neurons and having a microenvironment that is non-physiological.

Since the genes of cells in culture are easily manipulated and their protein products are rapidly

produced, the biochemical pathways and mechanisms involved in PD can be efficiently studied.

Indeed, specific molecular pathways underlying PD can be directly targeted in cellular models

(Dawson et al., 2010). In addition, specific cell types can be isolated to study changes that are

unique to that cell type. Studies show that the various toxins used to create models reliably act

upon their intended target and repeatedly produce cellular dysfunction and markers associated

with the disease state (Sherer et al., 2003b). The efficient nature of in vitro cultures enables their

use for rapid screening of therapeutic compounds. As a result, there will be a quicker “bench to

bedside” process enabling therapeutics to become available on the market more quickly.

However changes observed in cellular models do not exactly correlate with those seen in the

Page 53: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

38

disease state, thus more advanced in vivo models, which better emulate disease states, must be

utilized to further substantiate the findings obtained using in vitro models.

In vitro models lack the surrounding micro environment provided by in vivo models, but benefit

from having a more controllable environment (Alberio et al., 2012). Furthermore, most in vitro

models cannot recapitulate the complexity of multiple connections formed by cells from different

areas of the brain. This is highly disadvantageous considering PD pathology affects complex

networks organized between the SNc and striatum. Moreover, multiple cell types are affected,

which makes it difficult to model with most in vitro models, except for those models employing

organotypic cultures that are comprised of neurons from various brain areas (Snyder-Keller et

al., 2001). Given that in vitro models are isolated from organisms, they are unable to show how

changes mediated by therapeutic compounds affect disease symptoms and physiology of the

organism as a whole. Although in vitro models have many benefits, it is necessary to further

characterize and develop in vitro models which display the multitude of pathological

mechanisms found in PD, rather than modeling a single mechanism.

1.8.2 In vivo models

The use of animal models in PD research has been the main approach for developing therapeutic

compounds and studying disease symptoms. Toxin and transgenic animal models have been

used to replicate the pathology in sporadic and familial PD (Sherer et al., 2003a; Rockenstein et

al., 2007). The reserpine-treated rat model was developed in the late 1950’s, resulting in the

discovery of L-dopa as a potential treatment for parkinsonian symptoms. Reserpine application

in rodents induces muscle rigidity, postural flexion, and akinesia, closely resembling symptoms

observed in parkinsonian patients (Carlsson et al., 1957). However, administration of reserpine

produces a model that is acute, reversible and fails to replicate the pathology underlying

Page 54: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

39

idiopathic PD. A further disadvantage associated with the reserpine-treated rat is that serotonin

and noradrenaline levels are depleted to a greater extent compared to idiopathic PD. The

unilaterally-lesioned 6-OHDA rat model of PD has proved extremely useful in providing insight

into pathological, electrophysiological and pharmacological changes which occur following

degeneration of the nigrostriatal pathway, increasing our understanding of the mechanisms

underlying the generation of parkinsonian symptoms. Following stereotaxic injection of 6-

OHDA into the medial forebrain bundle, dopamine cell loss is more profound compared to that

observed following injections of 6-OHDA into the SNc (Carman et al., 1991). Metabolism of 6-

OHDA results in a phenomenal amount of oxidative stress, promoting deleterious effects of free

radicals on mitochondrial function, membrane stability and DNA integrity, which ultimately

results in cell death (Schwarting and Huston, 1996). Following unilateral administration of 6-

OHDA, rodents exhibit a spontaneous rotational behaviour ipsiversive to the side of the lesion

which represents an anti-parkinsonian action. Although exhibition of contraversive rotational

behavior in 6-OHDA lesioned animals is thought to represent an anti-parkinsonian action,

parkinsonian symptoms such as rigidity, tremor and bradykinesia are not apparent in these

animals (Ungerstedt, 1968). Furthermore adverse effects including adipsia, aphagia and high

mortality rates make this model less attractive (Ungerstedt, 1971). The MPTP mouse model is

widely used to evaluate potential therapeutic agents for PD (Cannon and Greenamyre, 2010).

This model shows progressive neurodegeneration of the nigro-striatal pathway, and sometimes

motor deficits. Thus, potential neuroprotective agents can be evaluated using post-mortem

measures of dopamine system integrity. The MPTP-lesioned mouse is easy to reproduce, and

more importantly, has proved to be one of the most useful models for assessing neuroprotective

potential of compounds in PD (Pothakos et al., 2009). One drawback of this model is that it does

Page 55: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

40

not result in the generation of protein aggregates, which are considered an important component

of PD pathology.

Upon the discovery of new genes linked with familial PD, classical toxin-induced and genetic

models became limited in their scientific value because they were unable to represent the various

forms of PD and their associated pathology. As a result, many animal models now express these

newly discovered genes that underlie PD (Li et al., 2010). This has led to the rapid identification

of the symptoms and pathology associated with certain genetic forms of PD. Models utilizing

AAV-mediated delivery and over-expression of α-synuclein in the rodent replicate the

progression of cell death and molecular pathology most closely to that observed in patients

(Koprich et al., 2010). The model also displays behavioral deficits consistent with a

parkinsonian phenotype. Following unilateral delivery of AAV-α-synuclein, dopaminergic

neurons become dysfunctional, exhibit abnormal axonal morphology and proteinase K-resistant

aggregations of α-synuclein in both terminals and nigral cell bodies, as well as increased striatal

dopamine transporter binding (DAT) (Koprich et al., 2010). These changes are accompanied by

behavioral abnormalities. At later stages, the pathology has progressed further, with marked

presence of dystrophic neurites, reductions in striatal TH, DAT and dopamine and a significant

loss of TH neurons within the nigra.

Animal models are critical for the development of neuroprotective strategies because they have

enabled the discovery of various sub-cellular deficits and various symptoms that arise from

mutations in the genes associated with PD and more importantly, reveal changes that occur in

live organisms. Moreover, these models enhance the clinical applicability of drugs since the

effect of therapeutic interventions on a live organism expressing genetic mutations closely

predicts the effect of the drug on patients. Although the various models replicate one particular

Page 56: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

41

aspect of PD, they do not emulate the multifaceted nature of the disease. This is demonstrated

by the inhibition of an individual mechanism by a particular toxin or genetic manipulation, rather

than a multitude of mechanisms as seen in PD patients. Furthermore, certain brain areas that are

not typically affected during PD appear to be vulnerable to degeneration (van der Putten et al.,

2000). Application of rotenone in animal models leads to global degeneration of neurons rather

than specific degeneration within the substantia nigra alone (Meredith et al., 2008). Markers

such as Lewy bodies that are typically found in patients exhibiting PD are not consistently

reproduced by the various toxin and genetic models (Dauer and Przedborski, 2003). Despite the

expression of these markers, many distinguishing features including nigral degeneration and loss

of tyrosine hydroxylase expression do not always occur. Although these models suffer from

disadvantages, they have been extremely useful in delineating gene specific pathology and

symptomatic treatments.

Given the advantages and disadvantages of in vitro and in vivo models with respect to

developing therapeutics to treat PD, these models must be further developed to better emulate all

the pathological mechanisms found in PD; then both models can be utilized to successfully

predict if a neuroprotective agent will be efficacious in clinical trials.

1.9 Hypothesis and aims

In PD, a variety of sub-cellular dysfunctions cause neurodegeneration of the SNc which

ultimately lead to the symptoms of PD. By understanding the function of mutations linked with

familial PD, we know that a number of separate mechanisms can trigger PD pathology.

Moreover, the utilization of environmental toxins in PD models has further highlighted that

multiple mechanisms induce PD pathology. Although a number of dysfunctional mechanisms

exist, the role of mitochondria in PD pathology is becoming more apparent. Thus, I

Page 57: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

42

hypothesized that altered mitochondrial function, due to the dysfunction of sub-cellular domains

and inhibition of mitochondrial dynamics, is a primary cause of disease pathology. Furthermore,

potential neuroprotective agents that target the mitochondria will be effective at protecting

degenerating neurons of the SNc. The initial hypothesis of this study was, that no matter what

the initial trigger, a single sub-cellular abnormality activates additional sub-cellular pathways,

such as the mitochondria, that further enhances the pathology of PD. Secondly, I hypothesized

that a correlation between the outcomes of neuroprotective agents in an in vitro model and those

of clinical trials can validate the effectiveness of an in vitro model at predicting potential

neurprotective agents to treat PD. Given the importance of mitochondrial function in the

pathology of PD, and the existence of LRRK2 on mitochondrial membranes, I hypothesized that

LRRK2 alters mitochondrial dynamics causing mitochondrial dysfunction. Moreover, a

potential neuroprotective agent such as SIRT3, which is involved in mitochondrial function, will

be effective at protecting neurons from degeneration.

The first aim of the studies described in Chapter Two was to understand how different triggers

associated with initiating the pathology of PD interact with other known components of the

neurodegenerative process associated with PD. The initial aim of these studies revealed that, no

matter what the initiating trigger, mitochondrial dysfunction occurs early in the cell death

pathway. This suggests that malfunction of mitochondria is central to all pathologies associated

with PD. The studies that were completed as part of the initial aim led to the generation of a cell

model of PD that took into account all known causes of PD. This model is quick, easy and

reliable to produce, thus, in Chapter Three my second aim of the studies validated this as a useful

model to test potential neuroprotective agents in PD using compounds that had previously passed

screening processes and were already in clinical trials. The third aim of the studies described in

Chapter Four was to further investigate how mitochondrial function is affected in PD. Since

Page 58: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

43

mitochondrial dynamics are required for physiological mitochondrial function, I investigated

changes in mitochondrial fission and fusion in cell models of PD. This aim revealed that

mitochondrial fusion is inhibited in a common genetic variant of PD. The fourth aim of the

studies outlined in Chapter Five was to evaluate the neuroprotective ability of two novel

compounds in our novel model of PD. This aim affirmed the importance of targeting the

mitochondria when trying to prevent the neurodegeneration associated with PD.

Page 59: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

44

Chapter 2

Mitochondrial Dysfunction Precedes other Sub-Cellular Abnormalities in an In Vitro Model

Linked with Cell Death in Parkinson’s Disease

A modified version of this chapter was previously published as:

Yong-Kee CJ, Sidorova E, Hanif A, Perera G, Nash JE (2012) Mitochondrial dysfunction precedes other sub-cellular abnormalities in an in vitro model linked with cell death in

Parkinson's disease. Neurotoxicity research 21:185-194.

Page 60: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

45

2 Mitochondrial Dysfunction Precedes other Sub-Cellular Abnormalities in an In Vitro Model Linked with Cell Death in Parkinson’s Disease

2.1 Introduction

By the time patients present with symptoms of PD, approximately 50% of the nigro-striatal

pathway has degenerated (Bernheimer et al., 1973), and a substantial proportion of the remaining

nigral neurons are undergoing some form of cell stress (Murray et al., 1995). Nevertheless

symptoms are typically not severely disabling and patients still have a good quality of life. Thus,

if it was possible to prescribe a disease-modifying agent that stopped or slowed down disease

progression at this stage of the disease, patients would not experience the advanced, severely

disabling stages of the disease. Development of such disease modifying strategy depends

critically upon understanding the sub-cellular mechanisms underlying neurodegeneration in PD.

Although 90-100% of cases of PD are believed to be sporadic, clues regarding the mechanisms

underlying cell death in PD have arisen from the 5-10% of cases that have been shown to be

genetically linked. All 15 loci on the 11 genes that have been linked to familial PD encode

proteins involved in the handling or removal of misfolded proteins, either at the level of the UPS,

macro-autophagy and CMA (Kitada et al., 1998; Ardley et al., 2004; Ramirez et al., 2006; Di

Fonzo et al., 2007; Lees and Singleton, 2007; Hatano et al., 2009), the regulation of

mitochondrial function (Lannuzel et al., 2003; von Bohlen und Halbach et al., 2004; Sherer et al.,

2007) and oxidative stress (Martinat et al., 2004; Shendelman et al., 2004). Furthermore, the

genetic mutations linked with familial PD cause dysfunction in at least one, if not all, of these

regulatory mechanisms (Hardy, 2010; Magen and Chesselet, 2010). For example, loss-of-

Page 61: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

46

function mutations in the E3 ubiquitin-protein ligase Parkin, a protein most commonly associated

with early-onset autosomal recessive PD (Kitada et al., 1998) was originally shown to play a role

in tagging proteins for degradation by the proteasome (Cookson, 2003; Sakata et al., 2003).

More recently, Parkin has also been shown to play a role in the regulation of mitochondrial

dynamics (Yun et al., 2008; Whitworth and Pallanck, 2009) as well as in CMA (Olzmann and

Chin, 2008). A second example of a protein associated with familial PD, which appears to cause

multiple problems at the sub-cellular level is α-synuclein. Mutations in α-synuclein cause

impaired UPS and lysosomal function (Stefanis et al., 2001; Tanaka et al., 2001; Cuervo et al.,

2004; Chu et al., 2009), and increased susceptibility of dopaminergic neurons to mitochondrial

toxins such as MPP+, as well as oxidative stress (Kanda et al., 2000; Qian et al., 2008). Finally,

post mortem studies in parkinsonian patients suggest that similar mechanisms are responsible for

causing cell death in both familial and idiopathic cases of this disease, since all show evidence of

impaired function of mitochondrial complex 1, UPS and lysosome, as well as increased oxidative

stress in the SNc and other affected brain regions (Parker et al., 1989; Jenner, 1993; Chu et al.,

2009). If it were possible to identify a common sub-cellular dysfunction that was initiated by all

known cell mechanisms associated with neurodegeneration in PD, this would represent a

valuable target for the development of a disease modifying agent since it would allow the rescue

of the remaining 50% of neurons in the SNc.

Given that multiple sub-cellular mechanisms are involved in Parkinson’s pathology, I

hypothesized that there is a single point of convergence of these mechanisms which results in

cellular stress. The studies described in this chapter sought to identify such a point of

convergence by using a catecholaminergic neuroblastoma cell line, which has previously been

used to study the mechanisms underlying cell death in PD (Dadakhujaev et al., 2010; Xie et al.,

Page 62: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

47

2010; Xie et al., 2011; Nonaka and Hasegawa, 2009; Wu et al., 2009), in combination with

toxins that induce cell death via the three mechanisms commonly associated with

neurodegeneration in PD: inhibition of mitochondrial complex 1 (rotenone), inhibition of the

UPS using Z-Ile-Glu(OBut)-Ala-Leu-H (PSI), and disruption of the lysosomal membrane via

5,8-dihydroxy-1,4-naphthoquinone (naphthazarin). In order to determine the effect of these

toxins in early, as compared to later stages of cell stress, when cell damage is unlikely to be

reversible, sub-cellular function was measured after three and twenty four hours of exposure to

toxin.

2.2 Materials and Methods

2.2.1 Materials

SH-SY5Y cells were purchased from ATCC (USA), Dulbecco’s Modified Eagle’s Medium

(DMEM), bovine calf serum, from Wisent (Canada) and trypsin from Sigma (USA). 5,8-

dihydroxy-1,4-naphthoquinone (naphthazarin) and rotenone were purchased from Sigma (USA),

Z-Ile-Glu(OBut)-Ala-Leu-H (PSI) from BIOMOL (USA) and alamar blueTM from Biosource

(Canada). Propidum iodide was purchased from Invitrogen (USA). Lysosensor green and JC-1

were purchased from Invitrogen (USA) and proteasome sensor vector from Clontech (USA).

Ubiquitin anti-rabbit antibody was purchased from Dako (USA), β-actin anti-mouse antibody

from Sigma (USA), anti-mouse and anti-rabbit HRP-conjugated secondary antibodies from

Jackson Laboratories (USA) and nitrocellulose membrane was purchased from BIORAD (USA).

PSI was dissolved in 0.1% dimethyl sulfoxide (DMSO) plus DMEM, and rotenone and

naphthazarin in DMEM only.

Page 63: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

48

2.2.2 Cell culture

Human dopaminergic neuroblastoma SH-SY5Y cells (ATCC) (P10-P30) were grown in DMEM

(consisting of 6400 mg/L NaCl, 3700 mg/L NaHCO3, 400 mg/L KCl and 584 mg/L L-glutamine)

supplemented with 5% bovine calf serum in a sterile humidified chamber (37 oC, 5% CO2, 95%

O2) (Incubator: MCO-20AIC, Sanyo, USA) until confluent. Cells were passaged using 0.1%

trypsin for 5 min, then pelleted by centrifuging at 340xg for 5 min (Allegra 6R Centrifuge,

Beckman Coulter, USA).

2.2.3 Cell viability assays

SH-SY5Y cells were grown in 96-well plates at 1.0 x 105 cells/ml. After 24 hours, cells were

exposed to full dose response curves of naphthazarin, PSI or rotenone (0 µM–1000 µM) in the

presence of the redox sensitive dye alamar blue (0.4% of final volume). Cell viability was

assessed by measuring the change in fluorescence of alamar blue (Ex. 544 nm, Em. 590 nm)

using a plate reader (FLUOstar OPTIMA, BMG Labtech, USA) at 3 and 24 hour time points.

Data are expressed as percentage cell viability compared to control ± SEM (n = 4).

2.2.4 Cell death assays

SH-SY5Y cells were grown in 6-well plates at a density of 2.0 x 105 cells/ml. After 48 hours,

naphthazarin (2.17 µM), PSI (80.0 µM) or rotenone (40.0 µM) was added to cells. Three or 24

hours following addition of toxin, propidium iodide (2 µM) was added to cells for 5 minutes, and

the number of propidium iodide positive (dead) cells quantified. Data are presented as

percentage of propidium iodide positive cells out of 150 counted cells ± SEM (n = 4).

Page 64: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

49

2.2.5 JC-1 and lysosensor green assays

Twenty-four hours post plating of SH-SY5Y cells, naphthazarin (2.17 µM), PSI (80 µM) or

rotenone (40 µM) were added. Following incubation with toxins for 3 or 24 hours, JC-1 (2 µM)

or lysosensor green (2 µM) was added to each well and incubated with the cells for 30 minutes,

then each well was aspirated and washed with DMEM before recording fluorescence intensity of

each probe (FLUOstar OPTIMA): JC-1 monomer: Ex. 485 nm, Em. 430 nm, J-aggregate: Ex.

535 nm, Em. 590 nm; lysosensor green: Ex. 440 nm, Em. 510 nm. Data from the JC-1 studies

are presented as the J-aggregate/monomer ratio in fluorescence intensity units ± SEM (n = 4).

Data from lysosensor green studies are presented as mean percent fluorescence intensity units

compared to control ± SEM (3 hours: N = 3; 24 hours: N = 4).

2.2.6 Proteasome sensor vector assay

SH-SY5Y cells were electroporated with ZsProSensor-1 using Nucleofector Kit V in A-23 mode

(Amaxa, USA) in 96-well plates. Naphthazarin (2.17 µM), PSI (80 µM) or rotenone (40 µM)

was added to the cells 48 hours after plating. Three or 24 hours following toxin addition,

fluorescence intensity was measured using a FLUOstar OPTIMA plate reader: Ex. 490 nm, Em.

520 nm. Data are presented as mean percent fluorescence intensity compared to control ± SEM

(3 and 24 hours: N = 4).

2.2.7 Western blotting

SH-SY5Y cells were exposed to naphthazarin (2.17 µM), PSI (80 µM) or rotenone (40 µM) for 3

or 24 hours. Cells were washed twice with ice-cold PBSx1 and scraped from 100 mm plates

with Laemmli buffer following toxin incubation. Samples were lysed for 15 minutes in water

(95 oC), and protein concentrations determined using the Bradford method (Bradford 1976).

Protein samples (10-20 µg) were loaded onto gels, and SDS-PAGE followed by Western blot

Page 65: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

50

was carried out. Nitrocellulose blots were blocked in 5% non-fat powdered milk (60 min) and

incubated with antibodies against β-actin (1:1000 µl) and ubiquitin (1:500 µl) overnight (4 oC).

Following 5 washes in 0.5% Triton-X in PBS, mouse (1:5000 µl) or rabbit (1:5000 µl) HRP-

conjugated secondary antibodies were incubated at room temperature (60 min) in a 1% non-fat

powdered milk/TTBS (0.1% Tween-20) solution. Protein levels were detected using enhanced

chemiluminescence (Immun-Star, Bio-Rad), then imaged (FluorChem, Alpha Innotech

Corporation) and optical density determined (FluorChem v2.0). Data are presented as mean

optical density ± SEM (n = 4).

2.2.8 Statistical analysis

For each experiment (n or N), each n/N was taken from a different plate of cells. For propidium

iodide assays, there was 1 replicate in each n. For alamar blue assays, there were 6 replicates for

each condition per N. For JC-1 and lysosensor green assays, there were 5 replicates in each N.

For proteasome sensor vector there was one replicate per N. Data generated from cell viability

were analysed using one-way ANOVA with Dunnett’s multiple comparison test post-hoc.

Propidium iodide assays were analysed using two-way ANOVA, using toxin and time of toxin

exposure as variables with Bonferroni test post-hoc. JC-1, proteasome sensor vector, lysosensor

green and Western blot experiments were analysed using one-way ANOVA with Tukey`s

multiple comparisons test post-hoc. For all studies, significance was assigned when P < 0.05.

2.3 Results

2.3.1 Effect of toxin exposure on cell viability and cell death

Exposure to naphthazarin significantly reduced cell viability over the ranges of 1 µM-1000 µM

at 3 hours (Fig. 2.1a) and 24 hours (Fig. 2.1b). The concentration of naphthazarin that decreased

cell viability by approximately 50% (EC50%) was 0.9 µM and 1.0 µM following 3 and 24 hours

Page 66: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

51

exposure respectively. PSI significantly reduced cell viability following incubation for 3 hours

and 24 hours. Post hoc analysis demonstrated a decrease in cell viability at concentrations

between 1 µM and 1000 µM following exposure for 3 hours (Fig. 2.1a) and 10 µM-1000 µM

following exposure for 24 hours (Fig. 2.1b) with an EC50% 6.04 µM and 100.0 µM respectively.

Rotenone also reduced cell viability after 3 hours and 24 hours exposure (Fig. 2.1). Post hoc

analysis showed a significant decrease in cell viability at concentrations between 1 µM and 1000

µM following 3 hours (Fig. 2.1a) and 24 hours exposure (Fig. 2.1b) with an EC50% 680.0 µM

and 210.1 µM respectively. Next, based on our findings in the cell viability assays and previous

studies in SH-SY5Y cells (Sapkota et al.; Jiang et al., 2004; Naoi et al., 2005; Chung et al., 2007;

Cho et al., 2008; Koch et al., 2009), approximate EC50% concentrations of each toxin were used

to determine the extent of cell death following 3 and 24 hours of toxin exposure to the cells.

Toxins were added to SH-SY5Y cells, and the amount of cell death was assessed using

propidium iodide. Following incubation of SH-SY5Y cells with toxins and propidium iodide,

there was a significant increase in the number of dead cells over time (Fig. 2.1c). Post-hoc

analysis showed that none of the toxins used significantly increased cell death after a 3 hour

exposure but all toxins caused a significant increase in cell death after 24 hours exposure.

Naphthazarin and rotenone increased cell death by 2-fold compared to vehicle (media), and PSI

increasing cell death by 4-fold compared to vehicle (DMSO).

Page 67: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

52

Figure 2.1. Effect of toxins on cell viability and cell death in SH-SY5Y cells. Cells were

incubated with naphthazarin, PSI or rotenone (0.1 µM–1000 µM) in the presence of alamar blue

for (a) 3 or (b) 24 hours, and cell viability was assessed. Data are expressed as mean percentage

fluorescence ± SEM compared to control (N = 4). One way ANOVA showed significant effects

of toxin exposure on cell viability following 3 and 24 hours exposure (naphthazarin: 3 hours: F5

= 84.5, 24 hours: F5 = 204.8; PSI: 3 hours: F5 (toxin) = 7.2, 24 hours: F5 (toxin) = 23.8;

rotenone: 3 hours: F5 (toxin) = 7.2, 24 hours: F5 (toxin) = 23.8). Dunnett’s multiple

comparisons test post-hoc showed that toxins caused a significant decrease in cell viability.

Naphthazarin: *** P < 0.001, compared to vehicle (media). PSI: $ P < 0.05, $$ P < 0.01, $$$ P

< 0.001 compared to vehicle (DMSO). Rotenone: & P < 0.05, && P < 0.01, &&& P < 0.001

compared to vehicle (media). (c) Cell death in SH-SY5Y was assessed following 3 or 24 hours

exposure to naphthazarin (2.17 µM), PSI (80 µM) or rotenone (40 µM) using propidium iodide

(PI). Data are presented as mean number of PI positive cells out of 150 ± SEM (n = 4). Two

way ANOVA showed a significant difference in propidium iodide uptake following incubation

with toxins for 3 and 24 hours compared to control (media for naphthazarin and rotenone;

DMSO for PSI) (F4 (toxin) = 26.49, F1 (time) = 56.76; F4 (interaction toxin x time) = 16.65).

Bonferroni test post-hoc showed a significant increase in propidium iodide uptake in the cells

following incubation with naphthazarin, PSI and rotenone for 24 hours compared to control, with

no significant effect of any toxin following incubation for 3 hours (*** P < 0.001).

Page 68: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

53

a

0 0.1 1 10 100 10000

20

40

60

80

100

120

*$

***$$$&&

***$$&

***$$$& ***

$$$&&&

concentration (τM)

cell viability (% control)

b

0 0.1 1 10 100 10000

20

40

60

80

100

120

***$$$&&&

***&& ***$

&&

***$$$&&&

concentration (τM)

cell viability (% control)

c

Napthazarin PSI RotenoneMedia DMSO

3 24 0

10

20

30

40

50

******

***

time (hr)

number of PI positive cells

3 hours

24 hours

Page 69: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

54

2.3.2 Effect of toxin exposure on mitochondria, UPS and lysosomes

An early marker of mitochondrial impairment is depolarisation of the mitochondrial membrane

potential. To assess the impact of toxins on mitochondrial function, naphthazarin, PSI or

rotenone were added to SH-SY5Y cells for 3 or 24 hours, and JC-1 was utilised to measure

mitochondrial membrane potential. After 3 hours exposure to all toxins, the ratio of JC-1

monomer to aggregates was significantly increased compared to controls (vehicle-treated cells),

indicating a depolarisation in mitochondrial membrane potential. Post hoc analysis showed that

the monomer to aggregate ratio following naphthazarin, PSI and rotenone treatment was

increased by 2.5-fold, 2-fold and 4.6-fold respectively compared to control (media for

naphthazarin and rotenone; 0.1% DMSO for PSI) (Fig. 2.2). Longer exposure to rotenone (24

hours) enhanced mitochondrial membrane depolarisation 6.1-fold compared to control. In

contrast, following 24 hours exposure to naphthazarin and PSI, there was no significant effect

compared to control (Fig. 2.2).

Page 70: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

55

Figure 2.2. Effect of compounds on mitochondrial membrane potential in SH-SY5Y cells.

SH-SY5Y cells were incubated with naphthazarin (2.17 µM), PSI (80 µM) or rotenone (40 µM)

for 3 hours or 24 hours. Changes in mitochondrial membrane potential were assessed using JC-

1. Data are expressed as ratio of mean fluorescence of aggregate to monomer ± SEM (N = 4).

One-way ANOVA showed a significant effect of toxins on mitochondrial membrane potential, 3

hours (F4 = 12.32) and 24 hours (F4 (toxin) = 22.85) following exposure. Tukey’s multiple

comparison test post-hoc: * P < 0.05, *** P < 0.001.

Page 71: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

56

Media DMSO NAP PSI ROT0.0

0.5

1.0

1.5 ******

*

3 hours

24 hours

ratio

of m

onom

er/a

ggre

gate

(ave

rage

FIU

)

*

Page 72: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

57

To determine the effect of naphthazarin, PSI and rotenone on proteasomal function, toxins were

incubated with cells previously transfected with the proteasome-sensitive fluorescent reporter

ZsProSensor-1 for 3 or 24 hours. When the proteasome is functioning normally, ZsProSensor-1

is broken down, however, when proteasomal function is compromised, ZsProSensor-1

accumulates, resulting in an increase in fluorescence intensity. Three hours of exposure to PSI

caused a significant increase (398%) in ZsProSensor-1 fluorescence compared to control,

indicating impaired proteasomal function however there was no significant effect of

naphthazarin or rotenone on proteasomal function. Following 24 hours exposure, impaired

proteasomal function was observed with all 3 toxins. Naphthazarin, PSI and rotenone increased

the fluorescence intensity of ZsProSensor-1 by 1135%, 2217% and 712% respectively (Fig. 2.3).

Page 73: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

58

Figure 2.3. Effect of toxins on proteasomal function in SH-SY5Y cells. Cells were

transfected with the proteasome-sensitive fluorescent reporter ZsProSensor-1 then incubated

with naphthazarin (2.17 µM), PSI (80 µM) or rotenone (40 µM) for (a) 3 hours or (b) 24 hours,

then fluorescence intensity measured. Data are expressed as mean fluorescence ± SEM (N = 4).

One-way ANOVA showed a significant effect of toxins on proteasomal function, 3 hours (F4

(toxin) = 3.15) and 24 hours (F4 = 90.61) following exposure compared to media control

(naphthazarin and rotenone) or DMSO control (PSI). Tukey’s multiple comparison post-hoc: * P

< 0.05, *** P < 0.001.

Page 74: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

59

3 hours

24 hours

Media DMSO NAP PSI ROT

aver

age f

luor

esce

nce (

%co

ntro

l)

050

100150200

5001000150020002500 ***

*******

Page 75: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

60

To determine the effect of naphthazarin, PSI and rotenone on lysosomes, cells were incubated

with the 3 toxins in the presence of lysosensor green, which fluoresces when taken into the

lysosome, serving as a marker of lysosomal function. Three hours of toxin exposure led to a

significant difference in lysosensor green fluorescence compared to control (Fig. 2.4). Post hoc

analysis showed that naphthazarin decreased fluorescence by 67.24% compared to vehicle,

whereas PSI and rotenone had no effect at this time. Following 24 hours of toxin exposure, there

was a significant effect of all toxins on fluorescence of lysosensor green compared to vehicle.

Post hoc analysis showed that naphthazarin, PSI and rotenone decreased fluorescence intensity

60.93%, 40.35% and 38.05% respectively.

Page 76: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

61

Figure 2.4. Effect of toxin exposure on lysosomal function. Lysosomal function was assessed

via measurement of fluorescence of lysosensor green 3 hours or 24 hours after the addition of

naphthazarin (2.17 µM), PSI (80 µM) or rotenone (40 µM). Data are expressed as mean

fluorescence ± SEM (N = 4). One-way ANOVA showed a significant effect of toxins on

lysosomal function, 3 hours (F4 = 3.63) and 24 hours (F4 = 15.31) following toxin exposure

compared to media control (naphthazarin and rotenone) or DMSO control (PSI). Tukey’s

multiple comparison test post-hoc * P < 0.05, ** P < 0.01, *** P < 0.001.

Page 77: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

62

0

40

80

120

**

***

3 hours24 hours

**

*

aver

age f

luor

esce

nce (

% co

ntro

l)

Media DMSO NAP PSI ROT

Page 78: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

63

2.3.3 Effect of toxins on ubiquitin levels

Post mortem studies have shown increased formation of ubiquitin aggregates in affected brain

regions of patients with PD (Leigh et al., 1989). However, it is not known whether such

pathologies arise due to dysfunction of the mitochondria, proteasome or lysosome, or indeed all

three. As shown in Figure 2.5a, b, a 3 hour exposure to naphthazarin, and PSI, but not to

rotenone altered ubiquitin expression. Following 3 hours of toxin exposure, naphthazarin and

PSI increased ubiquitin expression by 315.94%and 323.19% respectively compared to controls

(Fig. 2.5a,b). Following 24 hours of toxin exposure, ubiquitin expression was dramatically

increased compared to control. Post hoc analysis showed that naphthazarin, PSI and rotenone

increased ubiquitin levels by 7495%, 8954% and 10902% respectively compared to vehicle (Fig.

2.5a,c).

Page 79: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

64

Figure 2.5. Western blots to show changes in ubiquitin levels following toxin exposure. SH-

SY5Y cells were exposed to naphthazarin (2.17 µM), PSI (80 µM) or rotenone (40 µM) for 3

hours or 24 hours, harvested, and SDS-PAGE followed by Western blot was performed to detect

levels of ubiquitin. (a) Data are expressed as mean optical density ± SEM (n = 4). One-way

ANOVA showed significant effects following 3 hours (F4 = 5.17) and 24 hours (F4 = 253.1)

following toxin exposure compared to media control (naphthazarin and rotenone) or DMSO

control (PSI). Dunnett’s multiple comparison test post-hoc * P < 0.05, ** P < 0.01, *** P <

0.001. (b) Representative Western blot following 3 hours toxin exposure. (c) Representative

Western blot following 24 hours toxin exposure.

Page 80: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

65

a

0

200

400

6006000

8000

10000

12000

***

3 hours24 hours

**

Media DMSO NAP PSI ROT

***

***

*

aver

age o

ptic

al d

ensi

ty(%

con

trol)

bUbiquitin

ß-actin 42 kDa

200-250 kDa

MED

IA

DM

SO

NA

P

PSI

RO

T

c

DM

SO

NA

P

PSI

RO

T

Ubiquitin

ß-actin 42 kDa

200-250 kDa

3 hours

24 hours

Page 81: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

66

2.4 Discussion

SH-SY5Y cells were exposed to toxins that cause their effects via different mechanisms that

have been linked with neurodegeneration in PD, i.e., impairment of mitochondrial, lysosomal or

UPS function. The aim of this study was to determine a common cell stress mechanism

triggered by these different sub-cellular dysfunctions during the early stages of cell stress, so that

a target for a disease modifying agent may be revealed. Thus, it was necessary to utilise

concentrations of toxins that caused a significant level of cell stress (as measured by decreased

cell viability), while causing minimal levels of cell death, as this scenario recapitulates the

situation at the time of initial diagnosis of PD, when approximately 50% of neurons in the SNc

are alive, although may be undergoing some form of sub-cellular stress (Bernheimer et al.,

1973). Whilst in this study, cells were exposed to high concentrations of toxins for a relatively

short period of time (24 hrs) to induce acute toxicity. Previous studies have shown that this acute

model does reproduce the cell death processes implicated in PD (Yong-Kee et al., 2011a).

Following 3 hours exposure of all toxins at concentrations that caused an approximate 50%

decrease in cell viability (EC50%), no cell death was observed. Following 24 hours exposure,

the approximate EC50% of all toxins caused a significant increase (approximately 2-4 fold) in

cell death. Thus, following three hours of EC50% toxin exposure, 50% of SH-SY5Y cells

exhibit decreased cell viability, and the remaining 50% are likely to be undergoing some form of

sub-cellular stress, since there is a further decrease in cell viability after 24 hours. This scenario,

in which cells are under-going moderate to high levels of cell stress, although have not yet died,

is likely to be comparable to the situation in surviving neurons of SNc in parkinsonian patients.

Specifically, following 24 hours of toxin exposure, we see a loss of cell viability and an increase

in cell death. Thus, this represents a mixed population of cells, in which some are healthy, some

Page 82: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

67

are no longer viable, and some are dead (Bernheimer et al. 1973). Thus, exposure of cells to

toxins that mimic cell death mechanisms associated with PD for short periods (3 and 24 hours) is

a useful model for studying the early events which take place when cells are under cell stress.

Given that catecholaminergic neurons are the most susceptible to neurodegeneration in PD

(Hornykiewicz, 1972; Braak et al., 1996), the catecholaminergic neuroblastoma cell line, SH-

SY5Y, which has previously been used as a model for studying cell death mechanisms in PD

was selected for our studies (Dadakhujaev et al., 2010; Xie et al., 2010; Xie et al., 2011; Nonaka

and Hasegawa, 2009; Wu et al., 2009). Whilst primary mesencephalic cultures are a more

physiological representation of neurons of the substantia nigra pars compacta, it is difficult to

produce them in large quantities, and so it would not have been possible to perform the assays

utilised in the present studies. Previous studies have shown that lower concentrations of

rotenone (15-50 nM) were sufficient to cause cellular dysfunction (Liss & Roeper, 2001;

Shamoto-Nagai et al., 2003; Lannuzel et al., 2003; Lannuzel et al., 2006); in the present study

however, the concentration of rotenone required to decrease cell viability by 50% was 680.0 µM.

There are at least three possible explanations for these variations in the EC50% of rotenone:

firstly, cell type, secondly, the period of time in which rotenone was exposed to the cells, and

finally, the parameter utilised to measure cellular dysfunction. In one study, 15 nM of rotenone

was sufficient to inhibit mitochondrial complex 1 activity by 50% in acute mouse brain slices

(Liss & Roeper, 2001). Indeed, neurons are known to be much more sensitive to cell stress than

other cell types, such as SH-SY5Y cells that were used in the present study. Whilst SH-SY5Y

cells have a neuronal phenotype, they are not neuronal cells, and so are less sensitive to rotenone

as has been shown previously (Shamoto-Nagai et al., 2003; Lannuzel et al., 2003). Another

explanation for the difference in EC50s between the study carried out by Shamoto-Nagai and

colleagues and also Lannuzel et al., compared to the current study, is that in these two previously

Page 83: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

68

published studies, different markers of cellular dysfunction were used to measure the impact of

rotenone on cellular dysfunction. Shamoto-Nagai and colleagues measured inhibition of

mitochondrial complex 1 activity, whereas in the current study, changes in cell viability were

assessed. Since inhibition of mitochondrial complex 1 will precede a loss of cell viability, it is

most likely that the rotenone concentration required to cause a 50% inhibition of the

mitochondrial complex 1 is less than that required to cause a 50% decrease in cell viability.

Lannuzel and co-workers utilised loss of TH immunoreactivity as a marker of cell death.

Previously it has been shown that, in dopaminergic neurons, loss of TH phenotype precedes loss

of cell viability and neuronal cell death (Paul et al., 2004), thus providing a possible explanation

as to why the concentration of rotenone required was lower than in the present study.

Interestingly, Shamoto-Nagai and co-workers also used SH-SY5Y cells, and showed that 25-50

nM of rotenone was sufficient to kill 50% of the cells. However, in this study, 50% of cells died

following incubation with toxins for 72-120 hours. Indeed, as in our study, Shamoto-Nagai et

al., observe no cell death following incubation with rotenone for 24 or 48 hours. Finally, in the

present study, we describe an acute model of cellular stress, in which the initial sub-cellular

effects of toxic insult were evaluated. This justifies the use of higher concentrations of toxins for

a shorter time course.

When mitochondria, UPS and lysosomal function was assessed following 3 and 24 hours

exposure to naphthazarin, PSI or rotenone, all toxins caused depolarisation of the mitochondrial

membrane potential at 3 hours, whereas after 24 hours, only rotenone affected mitochondrial

function. This apparent recovery of mitochondrial function may represent a compensatory

mechanism, indeed it has been shown that pre-conditioning of the mitochondria occurs with mild

forms of toxic insults (Gidday, 2006; Busija et al., 2008). Three hours following toxin exposure,

only PSI inhibited proteasomal function, whereas following 24 hours exposure, all 3 toxins

Page 84: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

69

inhibited UPS function. Similarly, with respect to lysosomal function, after 3 hours of toxin

exposure, only naphthazarin decreased lysosomal function, whereas, following 24 hours of

exposure to toxins, all 3 toxins inhibited lysosomal function. The early effect of PSI on UPS

function and naphthazarin on lysosomal function confirms that the primary effects of these

toxins are via inhibition of the proteasome and lysosome respectively. Finally, following three

hours of toxin incubation, only naphthazarin and PSI increased levels of ubiquitination, whereas

following 24 hours treatment, all three toxins caused a significant increase in ubiquitin

expression. Both the UPS and lysosome are important for the breakdown of ubiquitinated

proteins (Kettern et al., 2010; Hochstrasser, 1992; Kubota, 2009; Todde et al., 2009), therefore,

the observed increase in ubiquitinated proteins shortly after incubation of cells with toxins that

inhibit these functions is not surprising. The finding that all 3 toxins increased ubiquitin levels

24 hours following toxin exposure is expected, since both lysosome and the proteasome-

mediated de-ubiquitination require ATP, thus extended periods of mitochondrial inhibition will

result in a lack of ATP, hindering physiological function of lysosomes and the UPS. Autophagic

processes require acidified lysosomal compartments that are maintained by ATP-dependent

proton pumps (Terman et al., 2010). Thus, reduced ATP levels may hinder lysosomal function,

by decreasing the rate of activity of the proton pump, which would slow down the ability of the

lysosome to degrade unwanted proteins. Similarly, UPS function relies heavily on a constant

supply of ATP. Access to the proteasomal core is regulated by the opening and closing of the

proteasomal cap (Betarbet et al., 2005). Therefore, since this process is ATP-dependent,

decreased ATP will also prevent protein degradation by the proteasome. Furthermore, excess

proteins in the cytosol may ultimately become deleterious to the cell because increased

misfolded proteins may form toxic aggregates which cause cell death (Mytilineou et al., 2004).

Page 85: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

70

Indeed, reduced ATP levels were found to hamper proteasomal activity in primary cultures

(Hoglinger et al., 2003).

Interestingly, both proteasomal and lysosomal inhibition reduced mitochondrial membrane

potential. Mitochondrial dysfunction caused by inhibition of the proteasome has been shown in

other in vitro studies (Qui et al., 2000; Ding and Keller, 2001; Hoglinger et al., 2003). Inhibition

of the proteasome results in an increase in the number of misfolded and aggregated proteins,

which has deleterious effects on the cell, by increasing the number of reactive oxygen species.

The resulting increase in oxidative stress further augments cell stress by increasing influx of Ca2+

into the cell, leading to excitotoxic conditions (Demuro et al., 2005; Danzer et al., 2007).

Increased excitotoxicity, enhances Ca2+ influx into the mitochondria, resulting in the

accumulation of free radicals within the mitochondria, which induces depolarisation of the

mitochondrial membrane potential (Ward et al., 2000) and loss of ATP (Budd and Nicholls,

1996). Whilst, there are no previous studies to show that inhibition of lysosomal function

impacts mitochondrial function, it has been shown that following over-expression of wild-type

α-synuclein in neuronal cultures, this protein accumulates in mitochondria leading to reduced

activity of mitochondrial complex 1 and subsequent mitochondrial dysfunction (Devi et al.,

2008). Whilst the reason why α-synuclein accumulates in the mitochondria is unclear; it is

known that α-synuclein is usually degraded by the lysosome, and that mutant α-synuclein

inhibits lysosome-mediated breakdown of misfolded and toxic proteins, including α-synuclein

(Cuervo et al., 2004; Sarkar et al., 2007). Impaired lysosomal function would also prevent

lysosome-mediated uptake of free radicals, which would in turn cause mitochondrial dysfunction

via similar mechanisms to those described for the proteasome (Kubota et al., 2010). Thus, these

studies indicate that, no matter what the mechanism of insult, mitochondrial dysfunction occurs

Page 86: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

71

early in the cell death process, indicating that targeting the mitochondria may be an effective

neuroprotective strategy against all causes of cell death linked with PD.

As mentioned above, previous in vitro studies in cell models of PD have shown a tight link

between mitochondrial and UPS function (Shamoto-Nagai et al., 2003; Sullivan et al., 2004;

Radke et al., 2008). It has also been shown previously that mitochondrial abnormalities result in

lysosomal dysfunction, suggesting that there is also a link between mitochondrial and lysosomal

integrity (Irrcher et al., 2010). The current study is the first to demonstrate the link between

UPS, lysosomal and mitochondrial abnormalities very early in the cell stress pathway. It is

known that lysosomes and the UPS work together to regulate protein degradation within the cell,

the present study also suggests that only after longer periods of impaired lysosomal function does

the UPS become impaired, and vice versa. This delayed impairment of the UPS or lysosome

following prolonged inhibition of lysosome or UPS respectively, is probably due to a build up of

aggregated proteins, as indicated by the increase in ubiquitin levels as early as 3 hours following

exposure to naphthazarin or PSI in the present study.

In conclusion, these studies show that with respect to mechanisms of cellular dysfunction linked

with PD, cells are most sensitive to mitochondrial dysfunction, since any form of cell stress,

whether caused by inhibition of mitochondria, proteasome or lysosome results in mitochondrial

dysfunction early in the cell death pathway. Finally, our findings suggest that protecting

mitochondrial function early in the disease process i.e., upon initial diagnosis could potentially

be a disease modifying strategy for preventing the cell death that characterises later stages of PD.

The assays utilised in these studies may prove useful for testing potential neuroprotective

treatments in the future.

Page 87: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

72

Chapter 3

Development and Validation of a Screening Assay for the Evaluation of Putative

Neuroprotective Agents in the Treatment of Parkinson’s Disease

A modified version of this chapter was previously published as:

Yong-Kee CJ, Salomonczyk D, Nash JE (2011) Development and validation of a screening assay for the evaluation of putative neuroprotective agents in the treatment of

Parkinson's disease. Neurotoxicity research 19:519-526.

Page 88: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

73

3 Development and Validation of a Screening Assay for the Evaluation of Putative Neuroprotective Agents in the Treatment of Parkinson’s Disease

3.1 Introduction

In order to prevent neurodegeneration, it is necessary to understand how cells die. Genetic and

post mortem studies suggest that inhibition of mitochondrial complex 1, aberrant protein

degradation caused by dysfunction of the UPS and lysosomes, as well as oxidative stress from

dopamine metabolism in neurons of the SNc contribute to neurodegeneration in Parkinson’s

disease (Ardely et al., 2004; Di Fonzo et al., 2007; Kitada et al., 1998; Ramirez et al., 2006).

Both the lysosome and UPS are required for the degradation of damaged proteins. Ubiquitin

motifs are attached to proteins by ubiquitin ligases, which target the protein to proteasomes,

while lysosomes employ chaperone-mediated autophagy for protein degradation. Abnormal

lysosome and UPS function leads to protein accumulation into toxic aggregates that eventually

cause cell death (Betarbet et al., 2005). Presently, there is no evidence that mutations in genes

involved in dopamine metabolism cause PD, however, in any neuron, dopamine metabolism is

inherently toxic, causing oxidative stress, due to the production of semiquinones, and other free

radicals. It is not known why dopaminergic neurons of the SNc are more susceptible to

oxidative stress than any other dopaminergic neuron however (Jenner, 2003).

It is probable that in most parkinsonian patients, a combination of some or all of the above

pathological processes contribute to cell death (Dagda et al., 2008; Canu et al., 2000; Hoglinger

et al., 2003; Pandey et al., 2007). Many animal and cell culture models of PD have been

developed to further understand how these cell death mechanisms interact (Hanrott et al., 2006;

Page 89: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

74

Panov et al., 2005) however, it is difficult to develop a model that takes into account the

numerous cell death mechanisms that are likely to cause idiopathic PD. Furthermore, such

models take months or years to develop. Unfortunately, without such a model, it is extremely

difficult to accurately predict the true potential of a putative disease modifying agent for the

treatment of PD.

Herein I hypothesized that a direct correlation between the assay used here, and the assays

conducted by NINDS can validate the effectiveness of this assay to test potential neurprotective

agents. The studies described in this chapter outline the development of an in vitro assay, which

involved exposure of catecholaminergic neuroblastoma cells (SH-SY5Y) to toxins that mimic

what are currently believed to be the main causes of neurodegeneration in PD. To assess the

validity of this assay, we tested five compounds that were previously chosen by the NINDS

sponsored CINAPS to move forward into clinical trials (Ravina et al., 2003; Heemskerk et al.,

2002). The results of this study directly correlate with the success of putative neuroprotective

agents in clinical trials. Therefore, it appears that this cell assay is a rapid and accurate predictor

of the usefulness of potential disease modifying agents for halting the progression of PD. We

propose that this assay will be a useful predictor of potential neuroprotective agents which may

be successful in clinical trials.

3.2 Materials and Methods

3.2.1 Materials

Z-Ile-Glu(OBut)-Ala-Leu-H (PSI) was purchased from BIOMOL (USA), alamar blueTM was

purchased from Biosource (Canada) and propidium iodide from Invitrogen (USA). Dopamine

hydrochloride, 5,8-dihydroxy- 1,4-naphthoquinone (naphthazarin), rotenone, coenzyme Q10,

caffeine, creatine, nicotine, R-(-)-deprenyl hydrochloride (deprenyl), salicylic acid and dimethyl

Page 90: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

75

sulfoxide (DMSO) were purchased from Sigma (USA). SH-SY5Y cells were purchased from

ATCC (USA), Dulbecco’s Modified Eagle’s Medium (DMEM), bovine calf serum, and trypsin

from Wisent (Canada).

3.2.2 Cell culture

SH-SY5Y cells were grown in DMEM (6400 mg/L NaCl, 3700 mg/L NaHCO3, 400 mg/L KCl

and 584 mg/L L-glutamine) and supplemented with 10% bovine calf serum in a sterile

humidified chamber (37 °C, 95% CO2, 5% O2) (MCO-20AIC, Sanyo, USA) until 80% confluent.

Cells were passaged using 0.1% trypsin for 5 min, then pelleted by centrifuging at 340xg

(Allegra 6R Centrifuge, Beckman Coulter, USA).

3.2.3 Effect of toxins on cell viability and cell death

To determine the effects of toxins on cell viability, SH-SY5Y cells (P20-P30) were plated at a

density of 1 x 105 per ml into 96 well plates 24 hours prior to the addition of dopamine

hydrochloride (0.1 µM–600 µM), naphthazarin (0.1 µM–1 mM), PSI (0.1 µM–500 µM) and

rotenone (0.1 µM–1 mM). The redox sensitive dye, alamar blue (0.4% of final volume) was

added to the cells immediately after the addition of toxin. Twenty four hours after treatment,

changes in alamar blue fluorescence were quantified (ex. 544 nm, em. 590 nm) using a plate

reader (FLUOstar OPTIMA, BMG Labtech, USA).

Cell death was determined using propidium iodide. SH-SY5Y cells were plated at a density of 1

x 105 per ml onto coverslips (diameter: 22 mm) in 6 well plates 24 hours prior to the addition of

dopamine (600 µM) and propidium iodide (2 µM). Cells were imaged at time 0, and 24 hours

later using confocal microscopy (LSM 510 META, Zeiss).

Page 91: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

76

3.2.4 Effect of putative neuroprotective agents on toxins

SH-SY5Y cells (P20-P30) were plated in 96 well plates at a density of 1 x 105 per ml 24 hours

prior to addition of compounds. Putative disease modifying agents were added to the cells 5

minutes before addition of toxins in the following concentrations: coenzyme Q10 (117 µM),

caffeine (140 µM), creatine (25 mM), nicotine (1 µM), R-(-)-deprenyl hydrochloride (10 µM)

and salicylic acid (10 mM). All compounds were dissolved in media. These concentrations

were chosen based on previous studies in cell lines (Andres et al., 2005; Lai & Yu, 1997; Menke

et al., 2003; Paterson et al., 1998; Xie et al., 2005). Approximate EC50 of toxins were added to

the cells; these were as follows: dopamine (30 µM), naphthazarin (2.17 µM), PSI (80 µM) and

rotenone (40 µM). Dopamine, naphthazarin and rotenone were dissolved in media; PSI was

dissolved in 0.1% DMSO. Alamar blue was added to the cells immediately after the addition of

toxin, and cell viability was assessed 24 hours later as described above.

3.2.5 Statistical analysis

A different passage of cells was used for each experiment (n = 1), fluorescence was expressed as

the mean of 6 replicates in each experiment (n = 6). Data are expressed as means ± SEM

compared to vehicle. Effect of toxin concentration on cell viability, and putative neuroprotective

agents on toxins was determined using two-way ANOVA with Bonferroni post hoc. Effect of

neuroprotective agents on cell viability was assessed using one-way ANOVA with Dunnett’s

multiple comparisons post hoc. Significance was assigned when P < 0.05.

3.3 Results

I first determined the EC50 of a battery of toxins that recapitulate the most likely cell death

mechanisms in PD, i.e. oxidative stress (dopamine), inhibition of the lysosome (naphthazarin),

inhibition of the mitochondria (rotenone) and proteasome chymotrypsin like activity inhibition

Page 92: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

77

(PSI). SH-SY5Y cells were exposed to a range of toxins for 24 hours, and cell viability assessed

(Fig. 3.1a). Two-way ANOVA, using treatment and concentration as factors, showed significant

effects of toxin treatment and concentration (F3,340 (toxin treatment) = 67.21, P < 0.0001, m =

11490; F16,340 (concentration) = 117.4, P < 0.001, m = 20080; F48,340 (interaction toxin treatment x

concentration) = 4.44, P < 0.001, m = 759.4) (n = 6). Bonferroni post hoc showed that dopamine

caused a significant decrease in cell viability compared to vehicle (media) at concentrations 10

µM–600 µM (P < 0.01–0.001, n = 6). The maximum decrease in cell viability was 83 ± 3.5%

SEM. Naphthazarin caused a significant decrease in cell viability compared to control (vehicle)

at concentrations 3 µM–1000 µM (P < 0.05–0.001, n = 6). The maximum decrease in cell

viability was 98 ± 2.7% SEM. PSI (0.1 µM -500 µM) caused a significant decrease in cell

viability compared to vehicle (0.1% DMSO) at concentrations 15 µM–500 µM (P < 0.05–0.001,

n = 6). Rotenone (0.1 µM–1000 µM) caused a significant decrease in cell viability compared to

vehicle (media) at concentrations 30 µM–1000 µM (P < 0.05–0.001, n = 6). The maximum

decrease in cell viability was 62 ± 5.6% SEM. The maximum decrease in cell viability was 82 ±

6.3% SEM. EC50’s for the toxins were as follows in order of decreasing potency: naphthazarin:

2.17 µM < dopamine hydrochloride: 30 µM < rotenone: 40 µM < PSI: 80 µM. To confirm that

changes in cell viability are a true correlate of cell death, cells were exposed to dopamine (600

µM) in the presence of propidium iodide for 24 hours. Immediately after addition of toxin, cells

appeared healthy, however following 24 hours incubation, the cells had absorbed the red dye,

due to cell membrane permeabilisation indicating that the cells were dead. Thus, measurement

of cell viability using alamar blue is a direct correlate of cell death (Fig. 3.1b).

Page 93: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

78

Figure 3.1. Effect of toxins on cell viability in SH-SY5Y cells. (a) SH-SY5Y cells were treated

with concentration response curves of toxin, which were as follows: dopamine hydrochloride

(0.1 µM–600 µM), naphthazarin (0.1 µM–1 mM), PSI (0.1 µM–500 µM) and rotenone (0.1 µM–

1 mM). After 24 hours of toxin treatment, cell viability was assessed using alamar blue.

Dopamine, naphthazarin, PSI and rotenone caused significant decreases in cell viability

compared to vehicle (media or DMSO) (two-way ANOVA with Bonferroni post hoc). Data are

expressed as mean ± SEM percent viability compared to vehicle (DMSO for PSI and media for

all other toxins) (n = 6). (b) SH-SY5Y cells were incubated with dopamine (600 µM) or vehicle

and propidium iodide (2 µM) and imaged immediately following addition of compounds and 24

hours later.

Page 94: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

79

b Control DA

0 hr

24 hr

0.0 0.1 1 10 100 10000

20

40

60

80

100

120

140DA

ROTPSINAP

[Toxins]µM

Cel

l Via

bilit

y (%

Con

trol)

a

Page 95: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

80

Following establishment of the EC50, putative disease modifying agents were added to the cells

immediately prior to the addition of each toxin. Following dopamine treatment alone, there was

an 88.3 ± 2.4% decrease in cell viability compared to vehicle-treated cells. ANOVA showed a

significant difference in cell viability following co-incubation of potential neuroprotective

compounds with dopamine compared to dopamine treatment alone (Bonferroni post hoc test

(F1,60 (toxin) = 149.2, P < 0.0001, m = 6194; F5,60 (concentration) = 17.0, P < 0.0001, m = 707.5;

F5,60 (interaction toxin x concentration) = 20.9, P < 0.0001, m = 868.7) (n = 6). Coenzyme Q10

(117 µM), creatine (25 mM), nicotine (1 µM) and salicylic acid (10 mM) reduced dopamine

induced toxicity by 16.3 ± 3.3%, 19.4 ± 4.2% 51.5 ± 3.6% and 10.4 ± 1.2% respectively (all P <

0.001, except salicylic acid P < 0.05, n = 6) (Fig. 3.2a). Caffeine (140 µM) and deprenyl (10

µM) had no significant effect on dopamine induced toxicity. There was a significant effect of

neuroprotective agents on naphthazarin-induced decreases in cell viability compared to

naphthazarin treatment alone (F1,60 (toxin) = 59.2, P < 0.0001, m = 958.6; F5,60 (concentration) =

169.3, P < 0.0001, m = 2743; F5,60 (interaction toxin x concentration) = 142.8, P < 0.0001, m =

2314) (n = 6). Following naphthazarin treatment, there was a 79.3 ± 0.6% decrease in cell

viability compared to vehicle-treated cells. When naphthazarin was combined with coenzyme

Q10 (117 µM) or salicylic acid (10 mM) there was a 10.8 ± 1.1% and 60.8 ± 5.2% increase in

cell viability, respectively, compared to naphthazarin alone (both P < 0.001, n = 6) (Fig. 3.2b).

Caffeine (140 µM), creatine (25 mM), nicotine (1 µM) and deprenyl (10 µM) did not attenuate

the reduction in cell viability caused by naphthazarin. Neuroprotective agents significantly

reduced PSI-induced decreases in cell viability compared to PSI treatment alone (F1,60 (toxin) =

161.7, P < 0.0001, m = 7080; F5,60 (concentration) = 11.9, P < 0.0001, m = 521.7; F5,60

(interaction toxin x concentration) = 10.1, P < 0.0001, m = 443.7) (n = 6). PSI caused a 26.6 ±

2.2% decrease in cell viability compared to vehicle-treated cells. Co-incubation of PSI with

Page 96: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

81

coenzyme Q10 (117 µM), creatine (25 mM), nicotine (1 µM), deprenyl (10 µM) and salicylic

acid (10 mM) led to a 16.2 ± 2.9%, 24.4 ± 3.5%, 24.5 ± 1.9%, 37.1 ± 2.1% and 16.5 ± 5.2%

increase in cell viability, respectively (all P < 0.001, n = 6) (Fig. 3.2c). Caffeine (140 µM) had

no significant effect on the decrease in cell viability caused by PSI. There was a significant

effect of neuroprotective agents on rotenone-induced decreases in cell viability compared to

rotenone treatment alone (F1,60 (toxin) = 531.1, P < 0.0001, m = 31590; F5,60 (concentration) =

18.1, P < 0.0001, m = 1074; F5,60 (interaction toxin x concentration) = 84.6, P < 0.0001, m =

5029) (n = 6). Following rotenone treatment there was a 37.6 ± 2.7% reduction of cell viability

compared to vehicle-treated cells. Coenzyme Q10 (117 µM), caffeine (140 µM), creatine (25

mM), nicotine (1 µM) and deprenyl (10 µM) resulted in a 53.2 ± 4.2%, 75.7 ± 2.1%, 63.4 ±

3.1%, 12.2 ± 3.7% and 73.9 ± 3.1% increase in cell viability respectively, compared to rotenone

treatment alone (all P < 0.001, except nicotine P < 0.05, n = 6) (Fig. 3.2d). Salicylic acid (10

mM) had no effect on the decrease in cell viability caused by rotenone.

Page 97: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

82

Figure 3.2. Effect of putative neuroprotective agents on toxin-induced decreases in cell

viability. Dopamine hydrochloride (30 µM), naphthazarin (2.17 µM), PSI (80 µM) or rotenone

(40 µM) were added to SH-SY5Y cells. Neuroprotective agents were added immediately prior to

toxins and incubated with cells for 24 hours. Data represent average cell viability as percent

control ± SEM of six independent experiments. * P < 0.05, *** P < 0.001 compared to untreated

control (two-way ANOVA with Bonferroni post-hoc test). (a) Coenzyme Q10, creatine, nicotine

and salicylic acid had significant protective effects compared to dopamine treatment alone. (b)

Reduced cell viability caused by naphthazarin was significantly attenuated by coenzyme Q10

and salicylic acid. (c) Coenzyme Q10, creatine, nicotine, deprenyl and salicylic acid had

significant protective effects compared to PSI treatment alone. (d). Rotenone-induced decreases

in cell viability were negated by coenzyme Q10, caffeine, creatine, nicotine and deprenyl.

Page 98: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

83

a

Coenz

yme Q

10 11

7 Mµ

Caffein

e 140

Creatin

e 25 m

M Mµ

Nicotin

e 1 Mµ

Depren

yl 10

Salicy

lic A

cid 10

mM

0

20

40

60

80

Vehicle + DopamineTreatment + Dopamine

*** ***

***

*

Cel

l Via

bilit

y (%

Con

trol

)

Page 99: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

84

b

Coenz

yme Q

10 11

7 Mµ

Caffein

e 140

Creatin

e 25 m

M Mµ

Nicotin

e 1 Mµ

Depren

yl 10

Salicy

lic A

cid 10

mM

0

20

40

60

80

100

Vehicle + NaphthazarinTreatment + Naphthazarin

***

***C

ell V

iabi

lity

(% C

ontr

ol)

Page 100: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

85

c

Coenz

yme Q

10 11

7 Mµ

Caffein

e 140

Creatin

e 25 m

M Mµ

Nicotin

e 1 Mµ

Depren

yl 10

Salicy

lic A

cid 10

mM

0

50

100

150

Vehicle + PSITreatment + PSI

****** *** ***

***

Cel

l Via

bilit

y (%

Con

trol

)

Page 101: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

86

d

Coenz

yme Q

10 11

7 Mµ

Caffein

e 140

Creatin

e 25 m

M Mµ

Nicotin

e 1 Mµ

Depren

yl 10

Salicy

lic A

cid 10

mM

0

50

100

150

Vehicle + RotenoneTreatment + Rotenone

*********

*

***C

ell V

iabi

lity

(% C

ontr

ol)

Page 102: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

87

It is possible that the apparent neuroprotective effects were caused by the compounds increasing

cell proliferation. To determine whether this was the case, the potential neuroprotective

compounds were incubated with SH-SY5Y cells alone, i.e. in the absence of toxins for 24 hours,

and their effect on cell viability was assessed using alamar blue. ANOVA showed a significant

effect of compound on cell viability. Dunnett’s multiple comparisons test showed that there was

a significant effect of nicotine compared to vehicle. Nicotine increased cell viability by 13.0 ±

2.4% compared to vehicle (P < 0.01, n = 6) (Fig. 3.3), indicating that nicotine has a mild

proliferative effect in SH-SY5Y cells. Coenzyme Q10, caffeine, creatine, deprenyl and salicylic

acid had no effect on cell viability compared to vehicle.

Page 103: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

88

Figure 3.3. Effect of putative neuroprotective compounds on viability in SH-SY5Y cells.

SH-SY5Y cells were treated with putative neuroprotective compounds for 24 hours in the

absence of toxins. Nicotine caused an increase in cell viability, while there was no effect of any

other agent on cell viability (one-way ANOVA with Dunnett’s multiple comparisons post hoc).

Data represent average cell viability as percent control ± SEM (n = 6). ** P < 0.01, compared to

untreated control.

Page 104: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

89

Vehicl

e Mµ

Coenz

yme Q

10 11

7 Mµ

Caffein

e 140

Creatin

e 25 m

M Mµ

Nicotin

e 1 Mµ

Depren

yl 10

Salicy

lic A

cid 10

mM

0

20

40

60

80

100

120 **C

ell V

iabi

lity

(% C

ontr

ol)

Page 105: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

90

3.4 Discussion

Here we demonstrate that exposure of SH-SY5Y cells to different toxins that mimic cell death

mechanisms associated with PD is a valid assay for testing potential disease modifying agents.

Coenzyme Q10 was chosen as one of the 12 putative neuroprotective agents for clinical trials, by

the NINDS-funded CINAPS group. This decision was made based on studies demonstrating that

coenzyme Q10 is neuroprotective, both in vitro and in vivo (Cleren et al., 2008; Moon et al.,

2005; Somayajulu-Nitu et al., 2009). Clinical trials showed that administration of high doses of

coenzyme Q10 to parkinsonian patients in the early stages of the disease significantly decreased

functional decline, as determined by the period of time before patients were required to take L-

dopa to control symptoms, suggesting that coenzyme Q10 slows disease progression (Shults et

al., 2002). In the current study, coenzyme Q10 was the most effective potential neuroprotective

agent tested. Coenzyme Q10 effectively protected against dysfunction of the mitochondria,

proteasome, lysosome and dopamine. The neuroprotective actions of coenzyme Q10 are thought

to be mediated by several mechanisms, which may explain the ability to protect against all four

toxins (Ernster and Dallner, 1995; Forsmark-Andrée et al., 1997). However, the primary

neuroprotective actions of coenzyme Q10 are on oxidative phosphorylation of the mitochondria,

where it transfers electrons from complex 1 and 2, to complex 3, thus facilitating ATP generation

(Schapira, 2006). In addition to this effect on mitochondrial function, coenzyme Q10-induced

increases in ATP production enables increased UPS function, since ATP is required for ubiquitin

conjugation to mis-folded proteins and opening of the proteasome channel for protein

degradation (Ciechanover, 2005). Furthermore, the reduced form of coenzyme Q10 is a strong

antioxidant which enables the effective removal of free radicals from mitochondria (Matthews et

Page 106: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

91

al., 1998). This in turn, may eliminate the toxic effects of free radicals produced by dopamine

metabolism and lysosomal destabilization.

To further evaluate the efficacy of this cell assay as a predictor of potential disease modifying

agents for PD, we also evaluated the ability of four other compounds that were chosen by the

CINAPS group: caffeine, creatine, nicotine and the MAO-B inhibitor deprenyl, to prevent

dopamine, naphthazarin, PSI and rotenone-induced toxicity in SH-SY5Y cells. Both creatine

and deprenyl have been shown to decrease cell death in animal models of PD (Hara et al., 2006;

Kragten et al., 1998; Yang et al., 2009). Initial clinical trials suggested that caffeine, creatine,

and deprenyl may delay the progression of parkinsonian symptoms, however, further studies

proved that none of these compounds were effective disease modifying agents (Bender et al.,

2006; Suchowersky et al., 2006; Simon et al., 2008). In this study, caffeine was only protective

against rotenone, having no significant effect on dopamine, naphthazarin and PSI-induced

decreases in cell viability. Creatine and deprenyl afforded significant protection against

dopamine-induced oxidative stress, as well as toxicity caused by mitochondria and proteasome

inhibition, however, neither had any significant effect on naphthazarin-induced decreases in cell

viability. Thus, the current studies suggest that caffeine, creatine and deprenyl were not effective

disease modifying agents in parkinsonian patients because they do not protect cells from all

forms of sub-cellular dysfunction associated with PD such as lysosome dysfunction.

Nicotine protected against dopamine, rotenone and PSI-induced toxicity, having no effect on

naphthazarin. While nicotine was chosen by the CINAPS group as a useful neuroprotective

agent for parkinsonian patients, to date, clinical trials have not been conducted on this

compound. The results from this study suggest that it would not be a good candidate as a disease

modifying agent for patients with PD.

Page 107: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

92

Previous studies have shown that both mitochondria and lysosome inhibition leads to the

increased presence of reactive oxygen species and oxidatively modified proteins within cells, and

that salicylic acid acts as a free radical scavenger, resulting in neuroprotection against such

insults (Kataoka et al., 1997; Kiffin et al., 2004; Testa et al., 2005). In addition, it has also been

shown that salicylic acid blocks the neurotoxic effect of MPTP in mice (Ferger et al., 1999;

Mohanakumar et al., 2000). The potential of salicylic acid as a disease modifying agent has not

been verified in the clinic however. Given that the current cell assay appears to be a good

predictor of the neuroprotective potential of agents to treat PD, we evaluated whether salicylic

acid may be such a candidate. Our findings that salicylic acid is effective in protecting against

cellular toxicity induced by dopamine, naphthazarin and PSI, but not rotenone suggests that this

would not be a useful disease modifying therapy for patients with PD.

In this study, we have shown that there is a direct correlation between the efficacy of disease

modifying agents in the clinic for the treatment of PD and the ability of these compounds to

protect cells against a battery of toxins that recapitulate cell death mechanisms associated with

the pathology of this disease. Thus, assessment of the ability of compounds to protect against a

battery of toxins in SH-SY5Y cells represents a simple, rapid, reliable and cost effective method

for evaluating their potential as neuroprotective agents in the treatment of PD. We believe that

using such an assay as a first step to assess purported disease modifying agents will reduce the

time taken for such agents to reach the clinic, as well as increasing the success of clinical trials.

We predict that the most effective agents will be those that protect against the multiple cellular

dysfunctions linked with Parkinson’s pathology.

Page 108: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

93

Chapter 4

Effect of Over-Expression of WT and Mutant LRRK2 on Mitochondrial Dynamics

Page 109: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

94

4 Effect of Over-Expression of WT and Mutant LRRK2 on Mitochondrial Dynamics

4.1 Introduction

Mutations in LRRK2 are responsible for the majority of familial and sporadic cases of PD (Mata

et al., 2006). There are numerous mutant variants of LRRK2; however, G2019S mutations are

the most prevalent in familial PD and in familial cases that were believed to be sporadic (Di

Fonzo et al., 2005; Farrer et al., 2005; Gilks et al., 2005). LRRK2 is a member of the ROCO

family of proteins that contains a Roc GTPase protein domain and kinase domain (Kett and

Dauer, 2012). The LRRK2 kinase domain is believed to be responsible for most of the protein’s

function, while the GTPase domain is less involved in mediating its function (Lee et al., 2012).

Interestingly, studies reveal the activity of the kinase domain is self-regulated by the GTPase

domain (Smith et al., 2006; Ito et al., 2007). These sites are flanked by multiple domains that

allow for numerous protein-protein interactions (Mata et al., 2006). The normal function of

LRRK2 is largely unknown, but has been implicated in neurite outgrowth, autophagy and

mitochondrial dynamics. Transgenic mice expressing mutant G2019S have impeded neurite

outgrowth (Parisiadou et al., 2009). Moreover, neurite shortening is associated with

dysfunctional autophagic processes in neuroblastoma cells (Plowey et al., 2008). Interestingly,

patients with G2019S mutations exhibited impaired mitochondrial function and reduced overall

intracellular ATP levels (Mortiboys et al., 2010). This Chapter is focused on determination of

the role of LRRK2 in the control of mitochondrial dynamics, in particular, fission and fusion.

Mitochondria undergo migration and trafficking, as well as fission and fusion to maintain their

energtic homeostasis and reduce mitochondrial damage; also to maintain quality control of

Page 110: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

95

mitochondrial proteins. Fission and fusion are of paramount importance to mitochondrial

function. Fission contributes to the distribution of mitochondria in response to localized ATP

demands, while fusion facilitates mtDNA exchange to rescue damaged mitochondria (Otera and

Mihara, 2011). Inhibition of fission machinery prevents autophagy of mitochondria and

promotes oxidation of mitochondrial proteins (Twig et al., 2008). Furthermore, inhibition of

mitochondrial fusion proteins leads to dysfunctional membrane potential and aberrant cellular

respiration (Olichon et al., 2003; Chen et al., 2005). These studies demonstrate the importance

of mitochondrial dynamics, not only for maintaining mitochondrial integrity, but also with

respect to the entire cell.

Alterations to mitochondrial dynamics have frequently been implicated in neurodegenerative

diseases (Baloh et al., 2007). Mutations in Mfn1 and Opa1 fusion proteins cause Charcot-Marie-

Tooth neuropathy and optic atrophy (Alexander et al., 2000; Zuchner et al., 2004).

Mitochondrial dysfunction is common in PD pathology; however while abnormalities in

mitochondrial dynamics are not believed to be responsible for the pathology of the disease,

mitochondrial dynamics has been shown to be dysfunctional in models of PD, suggesting that

while abnormal mitochondrial dynamics are not one of the initiating factors underlying the

pathology of PD, some of the known initiating factors that do trigger PD pathology do affect

mitochondrial dynamics, which probably exacerbates cell death. Nigral neurons have high

energy demands which make mitochondria critical to their survival (Vila et al., 2008). In

addition, mitochondrial dynamics are essential for distributing mitochondria across the long

processes of nigral neurons (Schon and Przedborski, 2011). Studies utilizing genetic and toxin

cell models of PD demonstrate the importance of mitochondrial dynamics in PD pathology.

Inhibition of mitochondrial fusion caused by α-synuclein is rescued by PINK-1, Parkin and DJ-1

overexpression (Kamp et al., 2010).

Page 111: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

96

Large GTPases regulate mitochondrial fission and fusion. Such proteins include Drp1 and Fis1

which mediate fission, and Mfn1, Mfn2 and Opa1 that regulate fusion. Fission is initiated when

cytosolic Drp1 binds to Fis1. Once bound to Fis1, Drp1 forms a multiple Drp1 complex which

leads to scission of the outer membrane (Westermann, 2010). Currently the mechanisms

controlling inner membrane division are unknown. Mfn1 and Mfn2 are transmembrane proteins

that tether opposing mitochondria during fusion (Otera and Mihara, 2011). Tethering brings the

mitochondria together to allow for fusion of the outer membrane (Otera and Mihara, 2011). The

mechanisms of inner membrane fusion is unclear, however Opa1 is believed to mediate this

process (Westermann, 2010). Altered mitochondrial fission and fusion are generally caused by

changes to fission and fusion proteins. Indeed, MPTP and rotenone, toxins used in models of

PD, are implicated in Drp-1 dependent mitochondrial fragmentation (Barsoum et al., 2006).

Mitochondria are important sources of dysfunctional mechanisms underlying familial and

idiopathic PD. Various toxins mediate their effects by inhibiting mitochondrial function, while

many mutations in genes linked with PD alter mitochondrial dynamics, however the effect of

LRRK2 on mitochondria is unknown (Fig. 4.1). Given that various genetic mutations and toxins

are implicated in mitochondrial dysfunction, and that LRRK2 mutations are common among PD

patients, I hypothesized that LRRK2 may alter fission and fusion, which causes mitochondrial

dysfunction linked with PD. The studies described in this chapter examine the effect of wild-

type and mutant LRRK2 (G2019S) on mitochondrial fission and fusion.

Page 112: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

97

Figure 4.1. Genetic mutations and toxins that affect mitochondrial processes. Mutations in

genes such as α-synuclein, DJ-1, Parkin and PINK1 have been implicated in the dysregulation of

mitochondrial dynamics. Environmental toxins such as rotenone block mitochondrial function.

Similarly, experimental toxins such as naphthazarin and PSI have been shown to block

mitochondrial function by first inhibiting the function of another organelle. The role of LRRK2

in mitochondrial dynamics and function is currently unknown.

Page 113: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

98

Mitochondrial Dynamics Mitochondrial Functionα-synuclein DJ-1 Parkin PINK1 Naphthazarin PSI Rotenone

??? LRRK2 ???

Page 114: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

99

4.2 Materials and Methods

4.2.1 Constructs

The pOCT-dsRed2 and pCMV-mitoGFP (kindly donated by Dr. Heidi McBride and Dr. Linda

Mills, respectively) constructs have CMV promoters which drive the expression of the

mitochondrially targeted fluorescent proteins. The pcDNA3.1-3xFLAG-LRRK2 and pcDNA3.1-

3xFLAG-G2019S-LRRK2 (kindly donated by Dr. Christopher Ross) constructs contain a CMV

promoter which drives the expression of the transgene. The 3xFLAG sequence is upstream of

the LRRK2/G2019S sequence. pcDNA3.1-3xFLAG-LRRK2 and pcDNA3.1-3xFLAG-G2019S-

LRRK2 have the same sequence except GGC was replaced by AGC at amino acid 2019. The

pcDNA3.1 empty vector was generated from the excision of the 3xFLAG-LRRK2 sequence at

Xho1 sites. The ptracer-3xFLAG-LRRK2-mCherry and ptracer-3xFLAG-G2019S-LRRK2-

mCherry constructs were generated from pcDNA3.1-3xFLAG-LRRK2, pcDNA3.1-3xFLAG-

G2019S-LRRK2, ptracer-CMV2, and pmCherry-C1 (Clontech, USA). The mCherry sequence

was excised, blunted and inserted into the Pml1 sites of ptracer-CMV2 so that expression of

mCherry was driven by the EF1 promoter. The LRRK2 or G2019S-LRRK2 sequence was then

ligated into the Kpn1 and Not1 sites of ptracer-CMV2-mCherry. LRRK2 and G2019S-LRRK2

was positioned upstream of the mCherry sequence and its expression was regulated by the

CMV2 promoter.

4.2.2 Cell culture

Human dopaminergic neuroblastoma SH-SY5Y cells (ATCC, USA) (P10-P25) were stored in

liquid nitrogen (-200 oC), rapidly thawed and grown in DMEM (consisting of 6400 mg/L NaCl,

3700 mg/L NaHCO3, 400 mg/L KCl and 584 mg/L L-glutamine) supplemented with 5% bovine

Page 115: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

100

calf serum in a sterile humidified chamber (37 oC, 5% CO2, 95% O2) (Incubator: MCO-20AIC,

Sanyo, USA) until confluent. Cells were passaged using 0.1% trypsin for 5 min, then pelleted by

centrifuging at 340xg for 5 min (Allegra 6R Centrifuge, Beckman Coulter, USA).

4.2.3 SDS-PAGE followed by Western blotting

SH-SY5Y cells were transfected with pcDNA3.1-3xFLAG-LRRK2 or pcDNA3.1-3xFLAG-

G2019S-LRRK2 (5µg). After 48 hrs, cells were washed twice with ice-cold PBSx1 and scraped

from 100 mm plates with Laemmli buffer. Samples were lysed for 15 minutes in water (95 oC),

and protein concentrations determined using the Bradford method. Protein samples (10-20µg)

were loaded onto gels, and SDS-PAGE followed by Western blotting was carried out.

Nitrocellulose blots were blocked in 5% non-fat powdered milk (60 min) and incubated with

antibodies against β-actin (1:1000 µl) and FLAG (1:1000 µl) (both Sigma-Aldrich, Canada)

overnight (4oC). Following 5 washes in 0.5% Triton-X in PBSx1, mouse (1:5000 µl) HRP-

conjugated secondary antibodies (Jackson Immunoresearch, USA) were incubated at room

temperature (60 min) in a 1% non-fat powdered milk/TTBS (0.1% Tween-20) solution. Protein

levels were detected using enhanced chemiluminescence (ECL Western Blotting Substrate,

Pierce), then imaged (SRX-101A, Konica Minolta). Three replicates were produced for each

condition.

4.2.4 Expression profile of constructs

SH-SY5Y cells were transfected with pcDNA3.1, pcDNA3.1-3xFLAG-LRRK2 or pcDNA3.1-

3xFLAG-G2019S-LRRK2 (all 8.6 µg) 24 hours after plating. Cells were fixed 8, 12, 24, 48 and

72 hours after transfection using 4% PFA, permeabilized with 0.1% Trion-X plus 100 mM

glycine, and blocked non-specific proteins with 5% bovine calf serum. Mouse anti-FLAG

(Sigma-Aldrich, Canada) was incubated with cells overnight at 4oC, which was then probed with

Page 116: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

101

anti-mouse cy2 (Jackson Immunoresearch, USA). In a separate experiment, SH-SY5Y cells

were transfected with ptracer-mCherry, ptracer-3xFLAG-LRRK2-mCherry or ptracer-3xFLAG-

G2019S-LRRK2-mCherry (all 5 µg) 24 hours after plating. Images were obtained 8, 12, 24, 48

and 72 hours after transfection. Images were captured using an epifluorescent microscope with

attached Apotome grid with 40x oil immersion objective (Axiovert 200, Carl Zeiss, Canada) for

fixed samples or with a 20x long distance objective for live imaging. One hundred cells were

counted for each of four replicates that were positively stained. Data are expressed as mean

number of positive cells ± SEM.

4.2.5 Mitochondrial morphology assay

SH-SY5Y cells were plated on glass coverslips in a 6 well dish 24 hours prior to transfection.

Cells were transiently transfected with pmCherry (0.768 µg), pCMV-mtGFP (1.86 µg) and

pcDNA3.1-3xFLAG-LRRK2 or pcDNA3.1-3xFLAG-G2019S-LRRK2 (both 8.6 µg). After 48

hours, cells were fixed with 4% parformaldehyde, permeabilized with 0.1% Triton-X plus 100

mM glycine and incubated with 5% bovine calf serum for 1 hour. In order to probe for 3xFLAG,

anti-mouse FLAG primary antibody was incubated at 4 oC overnight, followed by incubation

with anti-mouse cy5 secondary antibody (Jackson Immunoresearch, USA). Cells were imaged

using an epifluorescent microscope equipped with an Apotome grid with a 63x objective. To

determine the relative amounts of mitochondria that were fragmented, elongated or clustered,

one hundred cells from four independent experiments were analyzed in a blinded fashion.

Mitochondria were categorized as fragmented, elongated or clustered if 50% of the total

mitochondrial population in a cell displayed one morphology type. Data are expressed as mean

number of cells having a particular mitochondrial morphology ± SEM.

Page 117: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

102

4.2.6 Tetramethylrhodamine ethyl ester perchlorate (TMRE) assay SH-SY5Y cells were cultured on glass coverslips in a 6 well dish 24 hours prior to transfection

with pTag-BFP-C (0.78 µg) (Evrogen, USA), pcmv-mtGFP (1.86 µg) and pcDNA3.1,

pCDNA3.1-3xFLAG-LRRK2 or pCDNA3.1-3xFLAG-G2019S-LRRK2 (all 8.6 µg). On the

second day after transfection, cells were labelled with 20 nM TMRE (Invitrogen, USA) for 20

min in DMEM. TMRE is a lipophillic cationic dye that binds to the inner and outer portions of

the mitochondrial inner membrane upon proportion of membrane potential (Scaduto and

Grotyohann, 1999). Fluorescence intensity is reduced as a consequence of depolarisation in

membrane potential. Coverslips were placed in a live imaging chamber with imaging buffer

(116 mM NaCl, 5.4 mM KCl, 0.4 mM MgSO4, 20 mM HEPES, 0.9 mM Na2HPO4, 1.2 mM

CaCl2, 10 mM glucose, 5 mM pyruvate, pH 7.4) and imaged on an epifluorescent microscope

with Apotome with a 40x oil immersion objective. The fluorescence intensity of 30-50

randomly chosen cells from each of four independent experiments was measured using ZEN

2009 (Carl Zeiss, Canada). Then the average fluorescence intensity of all four independent

experiments was determined. Data are expressed as mean TMRE intensity ± SEM.

4.2.7 Polyethylene glycol (PEG)-induced cellular fusion assay

SH-SY5Y cells were plated in 6 well plates 24 hours prior to transfection. One well of cells was

transfected with pcmv-mtGFP (5.0 µg) and pcDNA3.1 (8.6 µg), or pcmv-mtGFP (5.0 µg) and

pcDNA3.1-3xFLAG-LRRK2 or pcDNA3.1-3xFLAG-G2019S-LRRK2 (both 8.6 ug). Another

well of cells was transfected with pOCT-dsRed2 (2.5 µg) and pcDNA3.1, or pOCT-dsRed2 (2.5

µg) and pcDNA3.1-3xFLAG-LRRK2 or pcDNA3.1-3xFLAG-G2019S-LRRK2 (both 8.6 µg).

After 8 hrs, cells from each well were co-plated (1:1 ratio) on coverslips and cultured for 16 hrs

in DMEM. Cycloheximide (40 µg/ml), an inhibitor of protein synthesis, was incubated with the

cells for 30 min. Cells were washed with PBSx1 (w/ 40 µg/ml cycloheximide) and incubated

Page 118: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

103

with a 50% w/v of PEG 3350 in PBSx1 for 90 sec. PEG induces membrane fusion between cells

by destabilizing lipid bilayers and neutralizing their charge gradient (Boni et al., 1984). Each

well was gently washed four times with PBSx1 (w/ 40 µg/ml cycloheximide). DMEM was

added to each well, and the cells were cultured for an additional 5 hrs prior to fixing with 4%

paraformaldehyde. Mouse anti-FLAG was incubated with cells overnight at 4 oC and incubated

with cy5 for 1 hr at room temperature. Images were acquired using a laser scanning confocal

microscope (Zeiss) with a 100x oil immersion objective. Twenty to forty cells from three

independent experiments were measured blindly for Pearson’s correlation coefficient (R) using

ZEN 2009. Pearson’s correlation coefficient was converted to R2 then to a percentage that

indicates the rate of mitochondrial fusion. Data are expressed as mean percent mitochondrial

fusion ± SEM.

4.2.8 Mitochondrial fission assay

SH-SY5Y cells were plated on glass coverslips in 6 well plates 24 hours before transfection.

Cells were transfected with pcmv-mtGFP (5 µg), or pcmv-mtGFP (5 µg) and ptracer-3xFLAG-

mCherry (8.6 µg), or pcmv-mtGFP (5 µg) and ptracer-3xFLAG-LRRK2-mCherry or ptracer-

3xFLAG-G2019S-mCherry (both 8.6 µg) using Lipofectamine LTX. After 24 hours, coverslips

were placed in a live imaging chamber with fresh DMEM. Images were acquired using a

spinning disk confocal microscope (WaveFX-X1, Quorum, Canada) with a 63x oil immersion

objective equipped with a temperature and CO2 controlled chamber. Images of five cells were

taken from five stage positions at each time point in four independent experiments. Z-stacks

consisting of 0.5 µm slices were acquired every 30 seconds over a 20 min period. Individual

slices were combined to create 3-D projections using Volocity 6.0.1 (Perkin Elmer, USA).

Images from each stage position were skeletonised using Metamorph 7.7.0.0 (Molecular

Page 119: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

104

Devices, USA). The number of mitochondria in each skeletonized image was counted using

Metamorph 7.7.0.0. Data are expressed as the number of mitochondria per 30 seconds.

4.2.9 Statistical analysis

A different cell passage was used for each replicate (n = 1). A minimum of three replicates were

used for each experiment. All data were analyzed used GraphPad Prism Version 5 (GraphPad

Software Inc, USA). The statistical tests used include Student’s t-test and one-way ANOVA’s

with associated post-tests. Significance was assigned when P < 0.05.

4.3 Results

4.3.1 Generation of ptracer-3xFLAG-LRRK2-mCherry construct

A variant of the LRRK2 construct with a fluorescent tag was required for live imaging

experiments. Thus, a construct was created which drives the expression of LRRK2 or G2019S-

LRRK2 and mCherry from different promoters within the same construct. The mCherry

sequence was first inserted into the Pml1 sites of ptracer-CMV2 (Fig. 4.2a). Restriction digest at

Bsrg1 and Pml1 sites show the insertion of the mCherry sequence in the correct location and

orientation (Fig. 4.2b). Digest at Bsrg1 and EcoR1 sites produced a 3.5 kb (ptracer sequence)

and a 2.5 kb (mCherry sequence) band. The ptracer-mCherry (5 µg) construct was transiently

transfected into SH-SY5Y cells to test its functionality (Fig. 4.2c). SH-SY5Y cells expressed

mCherry 48 hours after transfection. The 3xFLAG-LRRK2 or 3xFLAG-G2019S-LRRK2

sequence was inserted into ptracer-mCherry at Kpn1 and Not1 sites (Fig. 4.3a). Restriction

digest at Kpn1 and Not1 sites show that 3xFLAG-LRRK2 or 3xFLAG-G2019S-LRRK2

sequences are in the correct location and orientation (Fig. 4.3b). Digest at Kpn1 and Not1 sites

produced an 8 kb (3xFLAG-LRRK2/3xFLAG-G2019S-LRRK2 sequence) and 6 kb (ptracer-

mCherry sequence) band. The ptracer-3xFLAG-LRRK2-mCherry or ptracer-3xFLAG-G2019S-

Page 120: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

105

LRRK2-mCherry constructs were transiently transfected into SH-SY5Y cells and their

associated proteins were expressed after 48 hours (Fig. 4.3c). Western blotting also revealed the

expression of the proteins in SH-SY5Y cells (Fig. 4.3d).

Page 121: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

106

Figure 4.2. Generation of ptracer-mCherry construct. (a) The mCherry sequence was

inserted into Pml1 restriction sites and driven by the EF1 promoter region. Ampicillin resistance

(AmpR) is conferred to bacteria containing ptracer-mCherry. (b) Bsrg1 and EcoR1 restriction

enzymes were used to determine the correct orientation of the insert. The expected DNA bands

were approximately 3.5 kb and 2.5 kb if mCherry was inserted in the correct orientation. The 2.5

kb band contains the mCherry sequence. (c) Detection of mCherry in SH-SY5Y cell using live

imaging. Red fluorescence indicates mCherry expression driven by ptracer-mCherry construct.

Scale bar = 20 µm.

Page 122: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

107

a

Page 123: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

108

2.5 kb3.5 kb

b

c

Page 124: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

109

Figure 4.3. Generation of ptracer-3xFLAG-LRRK 2-mCherry construct. (a) The 3xFLAG-

LRRK2 or 3xFLAG-G2019S-LRRK2 sequence was inserted into the Kpn1 and Not1 restriction

sites. Bacteria containing ptracer-3xFLAG-LRRK2-mCherry has ampicillin resistance (AmpR).

(b) Restriction digest at Kpn1 and Not1 restriction sites to determine the orientation and location

of the insert. An expected DNA band size of 8 kb indicates the correct orientation of the LRRK2

sequence. (c) Detection of 3xFLAG-LRRK2-mCherry or 3xFLAG-G2019S-LRRK2-mCherry in

SH-SY5Y cells using live imaging. Red fluorescence indicates protein expression driven by

ptracer-3xFLAG-LRRK2-mCherry. Scale bars = 20 µm. (d) Detection of 3xFLAG-LRRK2-

mCherry or 3xFLAG-G2019S-LRRK2-mCherry in SH-SY5Y cells using SDS-PAGE followed

by Western blotting. The full length LRRK2 protein is approximately 250 kDa. – CNT: naive

cells; + CNT: cells transfected with pcDNA3.1.

Page 125: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

110

a

a

Page 126: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

111

250 kDa

3xFLAG-G2019S-mCherry 3xFLAG-LRRK2-mCherry

8 kb6 kb

c

d

b

Page 127: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

112

4.3.2 Expression of LRRK2 in SH-SY5Y cells

Since protein expression varies among cell types, it was first necessary to determine if wild-type

LRRK2 and G2019S-LRRK2 can be expressed in the neuroblastoma cell line SH-SY5Y.

Western blotting was used to detect the expression of these proteins 48 hours after transfection.

Wild-type LRRK2 and G2019S-LRRK2 were highly expressed compared to non-transfected and

empty vector controls (Fig. 4.4). Given that large proteins such as LRRK2 take longer times to

express, and different construct promoters have variable strengths, it was necessary to determine

the protein expression profile over a time course that will be used for future experiments. Here,

SH-SY5Y cells were transfected with wild-type LRRK2 and G2019S-LRRK2 constructs tagged

with 3xFLAG. Each protein was expressed in the cytoplasm of SH-SY5Y cells over the entire

time course (Fig. 4.5a). Wild-type and G2019S-LRRK2 expression was significantly increased

from 8 hours to 24 hours (P < 0.001) (Fig. 4.5b). At 8, 12, 24, and 48 hours, there was a 4.50 ±

0.87, 6.75 ± 0.85, 10.00 ± 1.08, and 14.50 ± 2.63 (P < 0.01, P < 0.001, P < 0.001, P < 0.001 and

P < 0.05, respectively) increase in cells positively stained with LRRK2. While cells expressing

G2019S-LRRK2 increased by 6.25 ± 0.85, 5.25 ± 0.85, 8.50 ± 2.10, and 13.50 ± 0.65 (P < 0.001,

P < 0.01, P < 0.001, P < 0.001 and P < 0.05, respectively) at 8, 12, 24, and 48 hours,

respectively. SH-SY5Y cells expressing wild-type LRRK2 and G2019S-LRRK2 tagged to

mCherry increased over a 24 to 48 hour time course (P < 0.001) (Fig. 4.6). Cells expressing

wild-type LRRK2 tagged with mCherry significantly increased by 7.50 ± 1.26 and 6.00 ± 0.50 at

24 and 48 hours, respectively (both P < 0.001). The number of SH-SY5Y cells expressing

G2019S-LRRK2 also increased by 7.75 ± 1.00 at 24 hours and 7.5 ± 0.76 at 48 hours post-

transfection (both P < 0.001).

Page 128: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

113

Figure 4.4. Expression of wild-type LRRK2 and G2019S-LRRK2 in SH-SY5Y cells. SH-

SY5Y cells were transfected with pcDNA3.1-3xFLAG-LRRK2 or pcDNA3.1-3xFLAG-

G2019S-LRRK2 (both 5 µg). Cells were lysed 48 hours after transfection and subjected to SDS-

PAGE followed by Western blotting. Wild-type LRRK2 and G2019S-LRRK2 expression were

determined by measuring 3xFLAG expression. Expression of wild-type LRRK2 and G2019S-

LRRK2 proteins are shown as bands with mass of 250 kDa. β-actin serves as a loading control.

CNT: naive cells; EV: cells transfected with pcDNA3.1; WT: wild-type LRRK2; G2019S:

G2019S-LRRK2.

Page 129: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

114

WTCNT EV

42 kDa

250 kDa

β-actin

3xFLAG

β-actin 42 kDa

250 kDa3xFLAG

G2019SCNT EV

Page 130: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

115

Figure 4.5. Wild-type LRRK2 and mutant G2019S-LRRK2 expression time course. SH-

SY5Y cells were transfected with pcDNA3.1-3xFLAG-LRRK2 or pcDNA3.1-3xFLAG-

G2019S-LRRK2 (both 8.6 µg) and were fixed 8, 12, 24, 48 and 72 hrs after transfection. Wild-

type LRRK2 and G2019S-LRRK2 expression was probed using 3xFLAG. (a) Wild-type LRRK2

and G2019S- LRRK2 were expressed in the soma and processes of SH-SY5Y cells. Scale bars =

20 µm. (b) Wild-type LRRK2 and G2019S-LRRK2 expression at 8, 12, 24, 48 and 72 hrs after

transfection. One-way ANOVA with Tukey’s multiple comparison test were performed. * P <

0.05, ** P < 0.01, *** P < 0.001 (WT), $ P < 0.05, $$ P < 0.01, $$$ P < 0.001 (G2019S).

Overall effect of LRRK2 treatment: F4,3 = 0.96, G2019S treatment: F 4,3 = 0.50. Control: naive

cells; empty vector: cells transfected with pcDNA3.1; WT: wild-type LRRK2; G2019S:

G2019S-LRRK2.

Page 131: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

116

Con

trol

Empt

y Ve

ctor

WT

G20

19S

8 Hrs 12 Hrs 24 Hrs 48 Hrs 72 Hrsa

Page 132: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

117

b

0 10 20 30 40 50 60 70 800

5

10

15

20Control

WTG2019S

Empty Vector

*****

***

***

*$$$$$

$$$

$$$

$

Time (Hrs)

Num

ber o

f Pos

itive

Cel

ls

Page 133: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

118

Figure 4.6. Expression of mCherry tagged wild-type LRRK2 and G2019S-LRRK2 in SH-

SY5Y cells. SH-SY5Y cells were transfected with ptracer-3xFLAG-LRRK2-mCherry or ptracer-

3xFLAG-G2019S-LRRK2-mCherry (both 5 µg) and were fixed 8, 12, 24 and 48 hours later.

The number of cells expressing wild-type LRRK2 and G2019S-LRRK2 were counted 8, 12, 24

and 48 hours after transfection. Two-way ANOVA with Bonferroni’s multiple comparison test

were performed. *** P < 0.001 (WT), $$$ P < 0.001 (G2019S), &&& P < 0.001 (vector).

Overall effect of treatment: F3,48 = 115.5. Control: naive cells; empty vector: cells transfected

with ptracer-mCherry; WT: wild-type LRRK2; G2019S: G2019S-LRRK2.

Page 134: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

119

0 10 20 30 40 50 600

10

20

30

40

50ControlVectorWTG2019S

*** ***$$$ $$$

&&&

&&&

Time (Hrs)

Num

ber o

f Pos

itive

Cel

ls

Page 135: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

120

4.3.3 Effect of LRRK2 on mitochondrial morphology

Since familial forms of PD lead to changes in mitochondrial morphology, and LRRK2 is

localized on the outer membrane of mitochondria, the effect of wild-type LRRK2 and G2019S-

LRRK2 on mitochondrial morphology was determined. SH-SY5Y cells were triple transfected

with pmCherry, pCMV-mitoGFP and pcDNA3.1-3xFLAG-LRRK2 or pcDNA3.1-3xFLAG-

G2019S-LRRK2. A triple transfection approach was used to determine the likelihood of a cell

expressing mtGFP, 3xFLAG-LRRK2 or 3xFLAG-G2019S-LRRK2. Since pmCherry had the

lowest molar ratio of the three plasmids, it is likely that one cell expresses LRRK2 if mCherry is

expressed. Control cells exhibit three types of mitochondrial morphology, clustered, fragmented

and elongated, each demonstrating a static state of mitochondrial dynamics (Fig. 4.7a). Over-

expression of wild-type LRRK2 and G2019S-LRRK2 had significant effects on mitochondrial

morphology (both P < 0.05). Wild-type LRRK2 expression led to a 14.50 ± 2.72 increase in

cells with fragmented mitochondria compared to control (P < 0.05) (Fig. 4.7b). G2019S-LRRK2

expression increased the number of cells having fragmented mitochondria by 18.75 ± 2.53 and

decreased the number of cells with elongated mitochondria by 16.00 ± 4.03 compared to control

(P < 0.001 and P < 0.05, respectively) (Fig. 4.7c). It is well known that mitochondria lose

function, and become fragmented in response to cell stress. In order to determine whether over-

expression of wild-type LRRK2 or G2019S-LRRK2 induces mitochondrial fragmentation by

activating stress response pathways, a TMRE assay was employed to assess mitochondrial

function. There was no difference in TMRE fluorescence intensity in LRRK2 and G2019S-

LRRK2 cells compared to control (P > 0.05) (Fig. 4.8a,b). Rotenone, the inhibitor of

mitochondrial complex 1, induced a significant decrease in TMRE fluorescence intensity (P <

0.05).

Page 136: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

121

Figure 4.7. Changes in mitochondrial morphology in SH-SY5Y cells. SH-SY5Y cells were

transfected with pcDNA3.1-3xFLAG-LRRK2 or pcDNA3.1-3xFLAG-G2019S-LRRK2 (both

8.6 µg) and fixed 48 hours after transfection. (a) Representative fluorescent images of

mitochondria expressing mtGFP, mCherry and wild-type LRRK2 or G2019S-LRRK2, showing

SH-SY5Y cells with clustered, elongated or fragmented mitochondria. Scale bars = 10 µm. (b)

Changes in mitochondrial morphology in cells expressing wild-type LRRK2 are shown. One-

hundred cells were counted and categorized as having clustered, elongated or fragmented

mitochondria. (c) Changes in mitochondrial morphology in cells expressing mutant G2019S-

LRRK2 are shown. One-hundred cells were counted and categorized as having clustered,

elongated or fragmented mitochondria. Paired two-tailed t-test was performed. * P < 0.05, ***

P < 0.001. Overall effect of wild-type LRRK2 treatment: t3 = 0.9 (clustered), t3 = 5.3

(fragmented), t3 = 1.4 (elongated); overall effect of G2019S-LRRK2 treatment: t3 = 1.0

(clustered), t3 = 13.6 (fragmented), t3 = 4.3 (elongated). Control: naive cells; Vector: cells

transfected with pcDNA3.1; WT: wild-type LRRK2; G2019S: G2019S-LRRK2.

Page 137: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

122

aG2019S

WT

Control

mtGFP FLAGVector

mCherry Merge

Page 138: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

123

b

Contro

lW

T0

20

40

60

ClusteredFragmented

*

Elongated

Mito

chon

dria

l Mor

phol

ogy

(# o

f cel

ls)

Contro

l

G2019

S0

20

40

60

ClusteredFragmentedElongated

****

Mito

chon

dria

l Mor

phol

ogy

(# o

f cel

ls)

c

Page 139: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

124

Figure 4.8. Effect of wild-type LRRK2 and G2019S-LRRK2 on mitochondrial function.

SH-SY5Y cells were transfected with pcDNA3.1-3xFLAG, pcDNA3.1-3xFLAG-LRRK2 or

pcDNA3.1-3xFLAG-G2019S-LRRK2 (all 8.6 µg), then incubated with TMRE (20 nM) 48 hours

later. (a) Fluorescent images of functional and non-functional mitochondria as shown by TMRE

fluorescence. Mitochondrial morphology is revealed by mtGFP fluorescence. BFP serves as a

transfection control for wild-type LRRK2 and G2019S-LRRK2. Rotenone serves as a positive

control to show changes in TMRE fluorescence. Scale bars = 10 µm. (b) Changes in

mitochondrial function of SH-SY5Y cells expressing wild-type LRRK2 and G2019S-LRRK2 are

shown. TMRE fluorescence intensity was quantified in 30-50 cells. One-way ANOVA with

Bonferroni’s multiple comparison test. * P < 0.05. Overall effect of rotenone treatment: F 4,15 =

3.0. Control: naive cells; Empty Vector: cells transfected with pcDNA3.1; WT: wild-type

LRRK2; G2019S: G2019S-LRRK2; mtGFP: mitochondrial GFP; BFP: blue fluorescent protein.

Page 140: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

125

Con

trol

Empt

y Ve

ctor

Rot

enon

eW

TG

2019

Sa

TMRE mtGFP BFP Merge

Page 141: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

126

Contro

l

Empty V

ector

Roteno

ne WT

G2019

S0

50

100

150

*

TMR

E (a

vera

ge in

tens

ity)

b

Page 142: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

127

4.3.4 Effect of LRRK2 on mitochondrial fusion and fission

Mitochondria are highly dynamic organelles that undergo repeated fusion and fission events.

Increased fragmentation could result from a loss of fusion which would produce many small

mitochondria instead of elongated forms. A classical fusion assay was employed to measure the

amount of fusion events that occur in the presence of wild-type LRRK2 and G2019S-LRRK2

(Fig. 4.9a). Wild-type LRRK2 and G2019S-LRRK2 expression decreased the percentage of

mitochondrial fusion (P < 0.0001). Wild-type LRRK2 and G2019S-LRRK2 reduced the

percentage of mitochondrial fusion by 15.82 ± 2.12% and 14.72 ± 2.77%, respectively (both P <

0.001) (Fig. 4.9b). Although reduced mitochondrial fusion may lead to fragmented

mitochondria, it is likely that excessive mitochondrial fission also fragments mitochondria.

Changes in the number of mitochondrial fission events were explored by utilizing live imaging

of fluorescently labeled mitochondria. Mitochondrial fission was not affected by the expression

of wild-type LRRK2 and G2019S-LRRK2 since there were no changes in the division or number

of mitochondria (P > 0.05) (Fig. 4.10).

Page 143: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

128

Figure 4.9. Effect of wild-type LRRK2 and mutant G2019S-LRRK2 on mitochondrial

fusion. SH-SY5Y cells were transfected with pcDNA3.1-3xFLAG-LRRK2 or pcDNA3.1-

3xFLAG-G2019S-LRRK2 (both 8.6 µg). Cell fusion was induced 24 hours later and SH-SY5Y

cells were fixed 5 hours after cell fusion. (a) Laser scanning confocal images showing fused

mitochondria. Different mitochondria populations are represented by mtGFP and dsRed2. Scale

bars = 20 µm. (b) Changes in the percentage of mitochondrial fusion in wild-type LRRK2 and

G2019S-LRRK2 expressing cells are shown. The percentage of mitochondrial fusion is derived

from the amount of colocalization in 20-40 cells per replicate. One-way ANOVA with

Bonferroni’s multiple comparison test was performed. *** P < 0.001. Overall effect of LRRK2

and G2019S: F3,263 = 17.5. Control: naive cells; Empty Vector: cells transfected with

pcDNA3.1; WT: wild-type LRRK2; G2019S: G2019S-LRRK2.

Page 144: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

129

mtGFP dsRed2 FLAG MergeC

ontro

lEm

pty

Vect

orW

TG

2019

Sa

Page 145: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

130

Contro

l

Empty V

ector W

T

G2019

S0

20

40

60

80

*** ***

% M

itoch

ondr

ial F

usio

nb

Page 146: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

131

Figure 4.10. Effect of wild-type LRRK2 and G2019S-LRRK2 on mitochondrial fission. SH-

SY5Y cells were transfected with pcmv-mtGFP (5 µg) and ptracer-mCherry, or pcmv-mtGFP (5

µg) and ptracer-3xFLAG-LRRK2-mCherry or ptracer-3xFLAG-G2019S-mCherry (8.6 µg).

After 24 hours, cells were imaged live using a spinning disk confocal microscope. The effect of

wild-type LRRK2 and G2019S-LRRK2 on mitochondrial number was measured. One-way

ANOVA with Bonferroni’s multiple comparison test was performed. P > 0.05. Overall effect of

LRRK2 and G2019S: F3,492 = 1.03. Control: naive cells; Empty Vector: cells transfected with

ptracer-mCherry; LRRK2: wild-type LRRK2; G2019S: G2019S-LRRK2.

Page 147: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

132

0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 448

9

10

11

12

13

ControlEmpty VectorLRRK2G2019S

Time (30 s)

Num

ber o

f Mito

chon

dria

(Nor

mal

ized

)

Page 148: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

133

4.4 Discussion

Regulation of mitochondrial dynamics, such as fission and fusion are of paramount importance

to cells, especially neurons that have high energy needs. Dysregulation of mitochondrial

dynamics is believed to contribute to the pathology of many neurodegenerative diseases

(Alexander et al., 2000; Zuchner et al., 2004; Trushina et al., 2012). To this end, changes in

mitochondrial dynamics were examined in an in vitro model of PD.

Aberrant mitochondrial dynamics are associated with changes in the morphology of

mitochondrial populations. Here, the morphology of mitochondria in response to over-

expression of wild-type LRRK2 and a mutant form of LRRK2, G2019S-LRRK2, was examined.

Interestingly, wild-type LRRK2 expression increased the number of fragmented mitochondria,

while G2019S-LRRK2 reduced the number of elongated mitochondria and increased the number

of fragmented mitochondria. This suggests that wild-type and mutant LRRK2 could regulate

rates of fission or fusion which alters mitochondrial morphology. These findings are in contrast

to previous studies showing an increase in elongated mitochondria in fibroblasts expressing

G2019S-LRRK2 (Mortiboys et al., 2010). This disparity may be due to the different cell type

used. Given that G2019S-LRRK2 leads to a toxic gain of function, it makes sense that over-

expression of this mutation leads to pathological changes, such as an increase in the number of

fragmented mitochondria. Moreover, this finding suggests that a pathological mechanism

involving G2019S-LRRK2 may decrease fusion or increase fission to promote these changes in

mitochondrial morphology. Indeed, studies have shown that mitochondria fragment in response

to abnormal mitochondrial dynamics caused by MPTP administration and over-expression of

mutated forms of Parkin (Lutz et al., 2009; Wang et al., 2011).

Page 149: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

134

Changes in mitochondrial morphology could be caused by cell stress and subsequent changes in

mitochondrial function. Indeed, many studies have shown that mitochondria become fragmented

as a consequence of mitochondrial dysfunction (Barsoum et al., 2006; Twig et al., 2008). To

address this possibility I performed TMRE experiments to examine the functional state of

mitochondria following over-expression of wild-type or mutant LRRK2. These studies showed

that there is no loss of mitochondrial function in cells over-expressing wild-type LRRK2 or

G2019S-LRRK2, suggesting that a secondary cell stress pathway is not activated by LRRK2

overexpression. Moreover, mitochondrial fragmentation is not a result of cell stress, but of

altered mitochondrial dynamics. This is consistent with studies where mitochondrial dynamics

altered mitochondrial function following over-expression of DJ-1 or α-synuclein (Kamp et al.,

2010; Wang et al., 2012a). Since cell stress does not precede mitochondrial fragmentation, it is

likely that altered mitochondrial dynamics induced by wild-type LRRK2 or G2019S-LRRK2

cause fragmentation, and could potentially cause mitochondrial dysfunction at later time points.

Indeed, measurements of mitochondrial fusion were taken at time points earlier than those of the

mitochondrial morphology and TMRE experiments. It is possible if TMRE measurements were

taken at time points later than those of the mitochondrial morphology experiments,

mitochondrial dysfunction could have been observed. Thus, altered mitochondrial dynamics

may cause fragmentation which promotes cell stress eventually causing cell death.

In the experiments examining mitochondrial morphology, it was shown that more fragmented

mitochondria were present in cells over-expressing wild-type LRRK2 or G2019S-LRRK2, while

less elongated mitochondria were present in cells expressing G2019S-LRRK2. These changes in

mitochondrial morphology can be attributed to either increased fission or decreased fusion, since

each would make mitochondria more fragmented or less elongated. Studies have shown the

involvement of PD related genes such as α-synuclein in mitochondrial fusion, and LRRK2 could

Page 150: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

135

also participate in the fusion process (Kamp et al., 2010; Xie and Chung, 2012). Both wild-type

LRRK2 and G2019S-LRRK2 expressing cells have mitochondria that appear less likely to

undergo fusion. Thus, it appears wild-type LRRK2 and G2019S-LRRK2 directly inhibit

mitochondrial fusion. This finding is supported by a study that found LRRK2 interacts with

membranous and vesicular structures such as mitochondria and vesicles (Biskup et al., 2006).

Here we see equivalent reductions of fused mitochondria following over-expression of wild-type

LRRK2 and G2019S-LRRK2. However, it remains unclear why G2019S-LRRK2 has a more

severe effect on mitochondrial morphology than wild-type LRRK2. It could be that

mitochondria take longer to fuse in the presence of G2019S-LRRK2 than wild-type LRRK2.

This is supported by experiments in stable cells which show that G2019S-LRRK2 increases the

time required for mitochondria to fuse (Wang et al., 2012b). However, the effect of G2019S-

LRRK2 on elongated mitochondria may be caused by enhanced mitochondrial fission.

In cells over-expressing G2019S-LRRK2, increased mitochondrial fission could reduce

elongated mitochondria. Live imaging of mitochondria revealed no change in mitochondrial

number, suggesting wild-type LRRK2 and G2019S-LRRK2 do not alter fission. This is

contrasted by studies that show wild-type LRRK2 and G2019S-LRRK2 interact with DLP1,

however whether altered fission occurred was not determined (Niu et al., 2012; Wang et al.,

2012b). Although DLP1 is essential for fission, it’s interaction with LRRK2 alone may not be

sufficient to induce alterations of fission rates. Indeed, Fis1 was found to be an imperative

component of the complex required for mitochondrial fission in mammalian cells (James et al.,

2003; Stojanovski et al., 2004). Given that mitochondrial fragmentation was observed 48 hours

after transfection in the mitochondrial morphology experiments, if mitochondrial fission was

measured at a similar time point, a significant change in mitochondrial fission may have been

observed.

Page 151: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

136

These experiments suggest wild-type LRRK2 plays an important role in mitochondrial fusion

and may play an equally important role in fission, and that G2019S-LRRK2 may further

exacerbate the effect of wild-type LRRK2 on fusion and fission which could potentially affect

mitochondrial function. Given the importance of mitochondria to the cell, mitochondrial

dysfunction induced by abnormal fusion could be linked to cell stress pathways which may

ultimately lead to cell death. This study suggests a new pathological process linked with a model

of PD, and in turn, suggests a potential therapeutic avenue for patients with PD since kinase

inhibitors of LRRK2 could be used to block the adverse effects of LRRK2 on mitochondrial

dynamics.

Page 152: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

137

Chapter 5

Neuroprotective Actions of SIRT3 and RGM in Advanced In Vitro Models of Parkinson’s

Disease

Page 153: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

138

5 Neuroprotective Actions of SIRT3 and RGM in Advanced In Vitro Models of Parkinson’s Disease

5.1 Introduction

In order to develop an effective neuroprotective treatment for PD, it is necessary to use a model

that mimics cell death mechanisms of PD as closely as possible. A number of in vivo and in

vitro models that mimic pathology of PD to different levels have been developed. Several in

vitro or cell models derived from human or animal tissue have been developed and include cell

lines, organotypic cultures and primary cultures. Toxin or genetic manipulation techniques are

used to generate cell models of sporadic and familial forms of PD (Sherer et al., 2003b; Testa et

al., 2005; Yong-Kee et al., 2012). Indeed, toxin and genetic models are useful for studying

cellular and biochemical mechanisms underlying pathology (Alberio et al., 2012). Furthermore,

human derived cell models benefit by having a human genetic background and may be more

useful for initial drug screening (Biedler et al., 1978). Various toxins have been applied to cells

to study the underlying mechanisms of sporadic and familial PD. Such toxins include MPTP and

rotenone which inhibit mitochondrial complex 1, proteasome inhibitors which cause UPS

dysfunction and 6-OHDA that causes oxidative stress (Hoglinger et al., 2003a; Mytilineou et al.,

2004; Hanrott et al., 2006). Genetic manipulation of α-synuclein and mutant variants represents

a classical model of familial PD, whilst manipulation of LRRK2 reproduces sporadic and

familial PD (Liu et al., 2005; Healy et al., 2008). While these models are advantageous for

examining individual mechanisms and for testing drugs in a high throughput methodology, they

lack the physiological environment provided by in vivo models.

Page 154: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

139

As discussed in detail in the introduction (Chapter 2), many animal models of PD have been

generated. While earlier models, in particular 6-OHDA lesioned rat, replicate symptoms, such

models do not reproduce the mechanisms underlying pathology (Betarbet et al., 2002). The

discovery of genetic mutations associated with PD that were linked with genes encoding proteins

linked with mitochondrial function verified that the MPTP lesioned primate or mouse probably

mimics some of the cell death mechanisms associated with PD pathogenesis (Dauer and

Przedborski, 2003; Meredith et al., 2008). More recently genetic mouse models that induce

over-expression or knock-down of proteins linked with familial PD have been developed

(Chesselet et al., 2012). These animal models excel as tools for determining the effects of drugs

on PD physiology and for understanding the pathophysiology of PD because they better model

the human condition (Betarbet et al., 2002). Genetic manipulation allows for protein expression

over the life time of the animal model, which better mimics the progression of the disease state.

Animal models show variations in their ability to replicate disease pathology. The level of α-

synuclein expression in transgenic mice influences whether Lewy bodies form (Meredith et al.,

2008). Furthermore, rotenone rat models do not consistently display a loss of dopaminergic

terminals or loss of TH (Lapointe et al., 2004). Animal models excel at providing a

physiological environment for studying PD, however they are inconsistent at producing disease

pathology.

As has been shown in Chapter 2, and in previous studies, oxidative stress and mitochondrial

dysfunction play a pivotal role in the degeneration associated with PD. Indeed, various studies

reveal the effectiveness of ROS scavengers and mitochondrial rescuing drugs at protecting PD

models (Yang et al., 2009; Yong-Kee et al., 2011b). Mitochondrial complex 1 inhibitors such as

MPTP and rotenone, induce neurodegeneration by initiating mitochondrial dysfunction, which

leads to other downstream dysfunction, such as oxidative stress, impaired UPS function and

Page 155: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

140

lysosome dysfunction (Perier et al., 2007). Oxidative stress and α-synuclein aggregation

activates cell death mechanisms in multiple cell lines (Hanrott et al., 2006). Thus, potential

neuroprotective agents require the ability to block oxidative stress and rescue mitochondria.

RGMa is a glyocosylphosphotidylinositol anchored protein involved in the collapse of growth

cones during retinal neuron development (Tassew et al., 2008). More recently, it was found to

interact with the dependence receptor, neogenin, during cell survival (Rajagopalan et al., 2004).

Neogenin receptors activate apoptosis when its ligand RGMa is unbound (Matsunaga et al.,

2004; Rajagopalan et al., 2004). In the presence of the RGMa, neuronal survival is promoted

(Matsunaga et al., 2004). Thus, RGMa may be an effective treatment in certain forms of PD.

SIRT3 is a member of the family of sirtuins that serve as NAD-dependent deacetylases and

ADP-ribosyl transferases (Onyango et al., 2002). SIRT3 is localized to the mitochondria where

it is involved in metabolism and biogenesis. Other functions include protein regulation through

PGC1α up-regulation, reduction of excitoxicity and modulation of stress response pathways

(Kong et al., 2010; Kim et al., 2011). The purported functions of SIRT3 make it a strong

candidate for a neuroprotective agent for PD.

Studies from previous chapters highlighted mitochondria as an important source of PD

pathology. Mitochondrial impairment by various toxins and mutations may impair energy

production and generate ROS which may result in apoptosis (Fig. 5.1). Given the potential cell

survival enhancing properties of RGMa and SIRT3, I hypothesized that these proteins may be

neuroprotective in models of PD, through the enhancement of energy production and prevention

of oxidative stress or apoptosis. The studies outlined in this chapter also describe the generation

of in vitro models that mimic key pathological processes known to occur in degenerating cells in

PD patients.

Page 156: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

141

Figure 5.1. The potential role of RGMa and SIRT3 in preventing PD cell death

mechanisms. Toxins and genetic mutations which directly or indirectly inhibit mitochondria

reduce energy production and increase ROS production. These processes may result in the

activation of apoptotic mechanisms which ultimately lead to cell death in PD. RGMa may

prevent mitochondrial induced cell death by directly blocking apoptosis. SIRT3 may prevent

reductions of ATP and block increases of ROS to protect cells from mitochondrial impairment

and subsequent cell death.

Page 157: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

142

Page 158: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

143

5.2 Materials and Methods

5.2.1 SH-SY5Y cells

5.2.1.1 Generation and maintenance of SH-SY5Y cell lines

Human dopaminergic neuroblastoma SH-SY5Y cells (ATCC, USA) (P10-P25) were stored in

liquid nitrogen (-200oC) and quickly thawed in a water bath. SH-SY5Y cells were grown in

DMEM (6400 mg/L NaCl, 3700 mg/L NaHCO3, 400 mg/L KCl and 584 mg/L L-glutamine)

supplemented with 5% bovine calf serum in a sterile humidified chamber (37 oC, 5% CO2, 95%

O2) (Incubator: MCO-20AIC, Sanyo, USA) until confluent. Cells were passaged using 0.1%

trypsin for 5 min, then pelleted by centrifuging at 340xg for 5 min (Allegra 6R Centrifuge,

Beckman Coulter, USA).

5.2.1.2 Neuroprotective effect of repulsive guidance molecule A (RGMa)

SH-SY5Y cells were plated at 1.0x105 cells/ml in 96-well plates. Twenty four hours later,

RGMa (0.1 µM) was incubated with the cells for 5 minutes. Then dopamine hydrochloride (300

µM), naphthazarin (7.26 µM), PSI (40 µM) or rotenone (10 µM) were added. Twenty-four hours

following addition of toxin plus the redox-sensitive dye alamar blue (0.03% final volume) cell

viability was assessed. Mean fluorescent intensity units of three independent experiments are

expressed as a percentage of control ± SEM.

5.2.1.3 Assessment of the neuroprotective effect of SIRT3 in SH-SY5Y cells

SH-SY5Y cells were plated at 5.0x105 cells/ml. Twenty-four hours later, cells were transiently

transfected with ptracer-SIRT3-GFP (0.6 µg) using Lipofectamine 2000. Dopamine

hydrocholoride (30 µM), naphthazarin (2.17 µM), PSI (80 µM) or rotenone (40 µM) and the

Page 159: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

144

redox-sensitive dye, alamar blue, were added 48 hours after transfection (time 48 hrs). Cell

viability was measured 24 hours after toxin treatment (time 72 hrs) using a fluorescent plate

reader (FLUOstar OPTIMA, BMG Labtech, USA). Data from four independent experiments are

expressed as mean percentage of control (vehicle) ± SEM.

5.2.1.4 Assessment of the neuroprotective effect of SIRT3 in differentiated SH-SY5Y cells

SH-SY5Y cells were plated at 5.0x105 cells/ml. Twenty-four hours later, cells were

differentiated with retinoic acid (10 µM) (Sigma, Canada) for three days. Cells were then

transiently transfected with pcDNA3.1-3xFLAG-LRRK2 (5 µg) and ptracer-GFP (0.6 µg) or

pcDNA3.1-3xFLAG-LRRK2 (5 µg) and ptracer-SIRT3-GFP (0.6 µg) using Lipofectamine 2000.

After 48 hours, propidum iodide (1.5 µM) was incubated with the cells for 30 min and

fluorescence was measured (time 48.5 hrs) using a fluorescent plate reader. Data from two

independent experiments are expressed as mean percentage of control ± SEM.

5.2.2 Ventral mesencephalic primary cultures

5.2.2.1 Solutions

Borate buffer (0.1 M) consists of 0.05 M boric acid, 0.01 M sodium tetraborate, 400 ml H2O.

Plating medium was made by combining 460 ml Neurobasal media (Gibco, Invitrogen, Canada),

10 ml B27 Supplement (Gibco, Invitrogen, Canada), 25 ml bovine calf serum (Wisent, Canada),

and 5 ml 1 % L-glutamine (Sigma, Canada). Growth media consists of 475 ml Neurobasal

media, 10 ml B27 supplement and 15 ml BCS.

5.2.2.2 Generation of ventral mesencephalic primary cultures

Poly-D-lysine hydrobromide (0.1 mg/ml) (Sigma, Canada) in 0.1 M borate buffer was pipetted

onto the centre of each coverslip and incubated overnight at 37 oC. Twenty-four hours later,

Page 160: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

145

poly-D-lysine was removed and coverslips were soaked in sterile H2O for 1 hr followed by a 5

min rinse with sterile H2O. Immediately after aspirating the final wash, 2 ml of plating medium

was added to each dish.

Sprague Dawley rats (embryonic day 16) were anesthetised with isoflurane and euthanized by

cervical dislocation. The uterus was removed from the cavity and the embryos were transferred

to a Petri dish filled with Hank’s Buffered Salt Solution (HBSS) (Gibco, Invitrogen, Canada).

Brains were carefully removed and transferred to a Petri dish containing ice-cold HBSS. After

removing the meninges, the SNc was dissected. The SNc was transferred to a 15 ml falcon tube

containing 5ml of ice-cold HBSS. The tissue was incubated with 10% Trypsin/EDTA solution

(Gibco, Invitrogen, Canada) for 15 min at 37 oC. Plating medium was added to the tissue and

dissociated firstly using a 1000 µl pipette tip, followed by a 200 µl pipette tip. Dead cells were

excluded following addition of trypan blue. Cells were counted and plated at a density of

1.0x104 per 2 ml and maintained at 37oC with 5% CO2 in a humidified chamber. Cells were fed

every four days with growth media. To inhibit glia growth on division 4 (D4), 1 µm cytosine β-

D-arabinofuranoside (Sigma, Canada) was added to each dish.

5.2.2.3 Characterization of ventral mesencephalic primary cultures

Primary cultures were grown for three weeks before being fixed with 4% PFA, permeabilized

with 0.1% Triton-X, and blocked with 5% BCS. Cultures were incubated with the following

antibodies: tyrosine hydroxylase (TH) (1:200 µl) (Chemicon, USA), G protein-activated inward

rectifier potassium channel 2 (GIRK2) (Alomone Labs, Jerusalem) (1:200 µl), post-synaptic

density 95 (PSD95) (1:500 µl) (Affinity BioReagents, USA), synaptophysin (1:250 µl)

(Chemicon, USA), glial fibrillary acidic protein (GFAP) (1:500 µl) (Cell Signalling, USA) and

mitogen-activated protein kinase (MAPK) (1:500 µl) (Cell Signalling, USA). Secondary

Page 161: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

146

antibodies, cy2 (1:1000 µl) and cy3 (1:2000 µl) (both Jackson ImmunoResearch Laboratories

Inc, USA) were incubated with cultures overnight at 4 oC. Images were captured using an

epifluorescent microscope at 40x magnification (Axiovert 200). Images are representative of

three independent experiments.

5.2.2.4 Generation of an in vitro α-synuclein model of PD

Primary ventral mesencephalic cultures were plated on glass coverslips and grown in vitro for

one week. AAV1/2-A53T α-syn, AAV1/2-empty vector and AAV1/2-GFP (all 3.4x109 gc/well)

were added to the cultures. After 6 weeks of virus exposure, fluorescent reactive dye (1 µl)

(Fixable Live/Dead Kit, Life Technologies, USA) was incubated with the cells for 30 min. This

was followed by fixation with 4% PFA, and permeabilization with 0.1% Triton-X and blocking

with 5% BCS. GIRK2 (1:500 µl) and ubiquitin (1:500 µl) (Santa Cruz Biotechnology, USA)

was incubated overnight at 4 oC. This was followed by a 1 hour incubation with cy3 (1:1000 µl)

and aminomethylcoumarin acetate (AMCA) (Jackson ImmunoResearch Laboratories Inc., USA).

Images were captured using an epiflourescent microscope equipped with a 63x objective

(Axiovert 200). Data consists of 30-40 cells in each of three independent experiments and is

expressed as mean percent positive cells ± SEM.

5.2.2.5 Assessment of the neuroprotective effect of SIRT3 in primary culture

Primary ventral mesencephalic cultures were maintained in vitro for three weeks (D21) prior to

transient transfection with ptracer-GFP (0.6 µg) or ptracer-SIRT3-GFP (0.6 µg). After 48 hours,

rotenone (20 nm) was incubated with the cells (time 48 hrs). After 48 hours of rotenone

exposure, propidum iodide (1.5 µM) was incubated with the cells for 30 min and fluorescence

was measured using a fluorescent plate reader (time 96.5 hrs). Data from two independent

experiments are expressed as mean percent change in cell death above control ± SEM.

Page 162: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

147

5.2.3 Nigro-striatal organotypic co-cultures

5.2.3.1 Generation of nigro-striatal organotypic co-cultures

Pregnant Sprague Dawley rats (Charles River, Canada) were obtained so that embryos would be

used at specific time points (E21-E23 and E14-E16). E21-E23 embryos were used for striatal

sections and E14-E16 embryos for SNc sections. A fourteen day pregnant rat was anesthetised

with isoflurane and euthanized by cervical dislocation. Embryos were quickly removed and

placed in a Petri dish containing ice cold F12 medium (Gibco, Invitrogen, Canada). Embryos

were decapitated and placed in a new Petri dish containing ice cold F-12 medium. The SNc was

isolated and placed in a new Petri dish containing ice cold F-12 medium. A twenty-one day

pregnant rat was anesthetised with isoflurane and cervical dislocation was performed. Embryos

were decapitated, brains removed, and placed in a Petri dish containing ice-cold F-12 medium.

Coronal brain slices (250 µM) were generated using a vibratome (VT1000S, Leica, USA).

Using a dissecting microscope, the striatum was removed from each coronal slice using a scalpel

and fine forceps and placed in a Petri dish containing ice-cold F-12 medium. The dorsolateral

area of the striatum was removed and incubated in a Petri dish containing ice-cold F-12 medium.

One dorsolateral area of striatum was then placed 1mm above hand-cut sections of SNc on PTFE

membranes (Millipore, USA). Cultures were incubated at 37 oC in 5% CO2 for 3 to 24 days.

Culture medium was changed every 3-4 days and treated at division 10 (D10) in vitro.

5.2.3.2 Characterization of cell types in nigro-striatal organotypic co-cultures

On D10, organotypic co-culture neurite outgrowth was examined. Sections were imaged in

phase contrast using an epifluorescent microscope equipped with a 20x long distance objective

(Axiovert 200, Carl Zeiss, Canada). For subsequent analysis, cultures were fixed and

immunostaining was performed. Co-cultures were excised by cutting around slices through

Page 163: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

148

PTFE membrane. Cells were fixed using 4% PFA/4% sucrose for 24 hrs at 4 oC. After fixation,

co-cultures were washed for 30 min and then incubated in 0.2% Triton-X for 30 min. Co-

cultures were incubated with 5% BCS for 2hrs. TH (1:200 µl), GIRK2 (1:200 µl) and PSD95

(1:500 µl) primary antibodies were diluted in 1% BCS and incubated with cultures overnight at 4

oC. Co-cultures were incubated with cy2 (1:1000 µL) or cy3 (1:2000 µL) secondary antibodies

diluted in 1% BCS for 1 hr at room temperature. Nuclei were stained by incubating co-cultures

with 4',6-diamidino-2-phenylindole (DAPI) (1:40000 µl) (Invitrogen, Canada) for 15 min. Phase

contrast images were captured using an epifluorescent microscope equipped with a 20x long-

distance objective (Axiovert 200, Carl Zeiss, Canada). Fluorescent images were captured using

an epifluorescent microscope equipped with an Apotome grid with a 63x objective (Axiovert

200) or without a deconvolution grid and 40x objective (IX81, Olympus, Canada). A total of

three replicates were generated for each experiment.

5.2.3.3 Assessment of cell death in organotypic co-cultures

To assess cell death, cultures were exposed to toxins that mimic cell death processes linked with

the pathogenesis of PD. Organotypic co-cultures (24 days in vitro) were treated with the

lysosome inhibitor, naphthazarin (60 µM, 80 µM), UPS inhibitor, PSI (80 µM, 100 µM) and

mitochondria complex 1 inhibitor, rotenone (50 µM, 70 µM) for 24 hrs. Propidium iodide (150

µM) (Invitrogen, Canada) was incubated with each co-culture for 1 hr, then removed from the

wells and replaced with fresh media. Propidium iodide fluorescence was then measured using a

plate reader (FLUOstar OPTIMA, BMG Labtech, USA). Mean fluorescence intensity units from

three independent experiments are expressed as mean percentage compared to control (vehicle-

no toxin) ± SEM.

Page 164: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

149

5.2.3.4 Assessment of TH/GIRK2 expression following exposure to toxins that mimic cell death processes in PD

Organotypic co-cultures (24 days in vitro) were treated with naphthazarin (60 µM, 80 µM) PSI

(80 µM, 100 µM) and rotenone (50 µM, 70 µM) for 24 hrs then fixed with 4% PFA/4% sucrose

for 24 hrs at 4 oC. After fixation, co-cultures were washed for 30 min and then incubated in

0.2% Triton-X in PBSx1 for 30 min. Co-cultures were blocked for 2 hrs using 5% BCS and

incubated with TH (1:200 µl) or GIRK2 (1:200 µl) antibodies overnight at 4 oC. Co-cultures

were incubated with cy2 (1:1000 µL) or cy3 (1:2000 µL) secondary antibodies diluted in 1%

BCS for 1 hr at room temperature. Nuclei were stained by incubating co-cultures with DAPI

(1:40000 µl) for 15 min. Co-cultures were imaged using an epifluorescent microscope with

attached Apotome grid and 40x objective (Axiovert 200). Images represent neurons from two or

three independent experiments.

5.2.4 Statistical analysis

A different cell passage was used for each replicate (n = 1). A minimum of two replicates were

used for each experiment. All data were analyzed used GraphPad Prism Version 5 (GraphPad

Software Inc, USA). The statistical tests used include one-way and two-way ANOVAs with

associated post-tests. Significance was assigned when P < 0.05.

5.3 Results

5.3.1 Assessment of the neuroprotective effects of RGMa and SIRT3

An intial screening of RGMa and SIRT3 was conducted to determine if there were any protective

effects of these proteins. Various toxins that mimic purported cell death mechanisms were

applied to SH-SY5Y cells and cell viability was measured in the presence of RGMa or SIRT3.

The presence of RGMa afforded significant protection to SH-SY5Y cells (Fig. 5.2). Pre-

Page 165: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

150

treatment with RGMa led to a 40.25 ± 10.72% increase in cell viability when exposed to

dopamine (P < 0.05). The presence of RGMa during rotenone toxicity increased cell viability by

21.55 ± 6.55% (P < 0.05). Similarly, the effectiveness of SIRT3 at protecting cells was also

tested in SH-SY5Y cells. Over-expression of SIRT3 reduced rotenone (40 µM) induced toxicity

by 17.48 ± 6.29% (P < 0.01) (Fig. 5.3). Given SIRT3 protected cells in a model that mimics

idiopathic PD, it was necessary to determine whether SIRT3 could protect cells in a model that

recapitulates familial PD. A differentiated SH-SY5Y model expressing LRRK2 was used to

asses the effectivenes of SIRT3. Over-expression of LRRK2 increased cell death by 31.60 ± 0%

compared to control (P < 0.001) (Fig. 5.4). Interestingly, SIRT3 reduced cell death by 9.15 ±

0.85% compared to control (P < 0.01). It was necessary to further confirm the neurprotective

potential of SIRT3 by utilizing a neuronal model. Ventral mesencephalic primary cultures were

challenged with a toxic dose of rotenone, and the level of cell death in the presence of SIRT3

was determined. SIRT3 protected ventral mesencephalic primary cultures from cell death

induced by rotenone (20 nm). The presence of rotenone increased cell death by 21.70 ± 0%

compared to control (P < 0.01), however, over-expression of SIRT3 prevented rotenone induced

cell death by 19.07 ± 1.57% (P < 0.01) (Fig. 5.5).

Page 166: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

151

Figure 5.2. Effect of RGMa on toxin-induced reductions of cell viability. SH-SY5Y cells

were plated 24 hours before treatment with RGMa, toxins and alamar blue. A fluorescent plate

reader was used to measure alamar blue fluorescence. The effect of RGMa on dopamine

hydrochloride (300 µM), naphthazarin (7.26 µM), PSI (40 µM) and rotenone (10 µM) toxicity is

shown. Student’s t-test. * P < 0.05. Overall effect of RGMa on dopamine: F5,5 = 1.38, overall

effect of RGMa on rotenone: F5,5 = 1.86.

Page 167: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

152

CONTROLRGM

DMSO DA

DA+RGMROT

ROT+RGMNAP

NAP+RGM PSI

PSI+RGM0

25

50

75

100

125

*

*C

ell V

iabi

lity

(% c

ontr

ol)

Page 168: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

153

Figure 5.3. Effect of SIRT3 in an SH-SY5Y cell model of PD. SH-SY5Y cells were plated 24

hours before being transiently transfected with ptracer-SIRT3-GFP (0.6 µg). Toxins were

incubated with the cells 24 hours prior to measuring cell viability. The effect of SIRT3 on cell

viability during rotenone (40 µM) toxicity is shown. Two-way ANOVA with Bonferroni post-

test. ** P < 0.05. Overall effect of SIRT3 on rotenone: F5,36 = 2.11.

Page 169: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

154

Media

DMSO DANAP PSI

ROT0

50

100

150ControlSIRT3

**

Cel

l V

iabi

lity

(% c

ontr

ol)

Page 170: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

155

Figure 5.4. Effect of SIRT3 in differentiated SH-SY5Y cells. SH-SY5Y cells were

differentiated with retinoic acid then transiently transfected with pcDNA3.1-3xFLAG-LRRK2

and ptracer-SIRT3-GFP. Cell death was measured using propidium iodide 48 hours after

transfection. The effect of SIRT3 on cell death associated with LRRK2 over-expression is

shown. One-way ANOVA with Bonferroni multiple-comparison test post hoc. ** P < 0.05, ***

P < 0.001. Overall effect of SIRT3 on LRRK2: F2,5 = 10.98.

Page 171: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

156

Contro

l

LRRK2+EV

LRRK2+SIR

T30

10

20

30

40*****

Cel

l Dea

th (%

con

trol

)

Page 172: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

157

Figure 5.5. Effect of SIRT3 in primary cultures. Ventral mesencephalic primary cultures

were grown for 21 days before transient transfection with SIRT3. Rotenone (20 nm) was

incubated with cultures for 48 hours before measuring propidium iodide fluorescence. The effect

of SIRT3 on rotenone induced increases in cell death is shown. One-way ANOVA with

Bonferroni multiple-comparison test post hoc. ** P < 0.05. Overall effect of SIRT3 on rotenone:

F2,6 = 79.13

Page 173: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

158

Contro

l

ROT+EV

ROT+SIRT3

0

5

10

15

20

25 ****C

ell D

eath

(% c

ontr

ol)

Page 174: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

159

5.3.2 Development of an in vitro α-synuclein model utilizing ventral mesencephalic primary cultures

In order to create a valid and effective in vitro model of PD, the characteristics of neurons that

degenerate in PD must be recapitulated. To this end, the relative expression of glia and neurons

in our ventral mesencephalic cultures were determined. Immuno-labelling was performed using

antibodies against MAPK and GFAP to distinguish between dendrites and glia. Ventral

mesencephalic primary cultures contain glia and neurons as demonstrated by strong GFAP and

MAPK expression (Fig. 5.6a,b). Since only SNc neurons from the ventral mesencephalon

degenerate in PD, it was necessary to determine if these cultures consist of SNc or VTA neurons.

Strong GIRK2 expression was found in these cultures, revealing the existence of SNc neurons

(Fig. 5.7a). Furthermore, these populations contain TH, indicating that these neurons are indeed

functional. These primary cultures also form synaptic connections as demonstrated by PSD95

and synaptophysin expression (Fig. 5.7b). Large puncta are seen at sites where processes contact

each other. There are a number of genetic mutations that lead to PD pathology. The most

thoroughly examined of which are mutations in the gene that encodes the protein, α-synuclein.

Thus, an in vitro model was generated by over-expressing a mutated form of α-synuclein that is

linked with PD. Ventral mesencephalic primary cultures were subjected to recombinant AAV

expressing mutant α-synuclein mutant gene expression. Six weeks after virus exposure, A53T

mutant α-synuclein expression caused a 58.54 ± 7.52% increase in cell death compared to

control (PBSx1 vehicle) (Fig. 5.8a,b). Ubiquitin expression was also examined in A53T α-

synuclein expressing cells. Ubiquitin aggregates did not increase in neurons exposed to the

A53T mutation (Fig. 5.8b).

Page 175: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

160

Figure 5.6. Determination of MAPK and GFAP expression in primary culture. Ventral

mesencephalic primary cultures (D21) were fixed and immuno-labelled with antibodies against

GFAP and MAPK. These cultures express both neurons and glia. (a) GFAP expression is

localized around the entire soma of glia. (b) Neurons strongly express MAPK within their cell

bodies and processes. Scale bars = 20 µm.

Page 176: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

161

GFAP DAPI Mergea

MAPK DAPI Mergeb

Page 177: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

162

Figure 5.7. Determination of TH and GIRK2 expression in ventral mesencephalic primary

culture. Ventral mesencephalic primary cultures (D21) were fixed and immuno-labelled with

antibodies against TH and GIRK2. These neurons express both TH and GIRK2. (a) TH and

GIRK2 expression is localized in the neuronal bodies and processes. (b) Strong PSD95 and

synaptophysin expression is found in neuronal processes. Scare bars = 20 µm.

Page 178: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

163

GIRK2TH MergeDAPIa

SynaptophysinPSD95 MergeDAPIb

Page 179: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

164

Figure 5.8. Effect of mutant A53T α-synuclein expression in ventral mesencephalic primary

cultures. Cultures were transduced with recombinant AAV expressing mutant (A53T) α-

synuclein for 6 weeks. After 6 weeks exposure, cell death was assessed using a live/dead kit

which exhibits bright fluorescence upon binding internal/external amines of dead cells. Cells

were then fixed and immuno-labelled with antibodies against GIRK2 and ubiquitin. (a) Effect of

mutant A53T α-synuclein expression on cell death. Cell death is represented by positive staining

of the live/dead kit. (b) Representative images of GIRK2 cells with positive staining of

internal/external amines of dead cells by the live/dead kit. Scale bars = 20 µm. One-way

ANOVA with Tukey’s multiple comparison test post-hoc. ** P < 0.01. Overall effect of α-

synuclein A53T: F3,11 = 10.30.

Page 180: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

165

Contro

l

Empty V

ector

GFP

-synu

clein

α

0

20

40

60

80 **C

ell

Dea

th (%

pos

itive

cel

ls)

a

Page 181: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

166

A53

T α-

synu

clei

nG

FPEm

pty

Vect

orC

ontro

lGIRK2 Live/DeadA53T α-synuclein Ubiquitin Mergeb

Page 182: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

167

5.3.3 Development of a nigro-striatal organotypic co-culture model of PD

PD symptoms result from the dysregulation of complex neuronal networks that consist of

specialized cell types. Thus, it was necessary to identify synaptic connections and characterize

specific cell types in these nigro-striatal organotypic co-cultures. After 14 days, axons from SNc

neurons traverse the space between slices and innervate the striatal section (Fig. 5.9a). Synapse

formation is shown by the presence of synaptic markers, such as PSD95. Here, neurons from

these co-cultures express PSD95 within their projections (Fig. 5.9b). Degeneration of

dopaminergic neurons occurs selectively in the SNc, leaving the VTA intact. To determine

whether the neurons in our co-cultures were from the SNc or the VTA, we measured GIRK2

expression as this is expressed specifically in the SNc. As shown in Fig. 5.10a, a large

proportion of neurons in these cultures strongly express GIRK2. Since these neurons also

express TH (Fig. 5.10b), it appears that these neurons represent dopaminergic nigro-striatal

neurons, rather than neurons originating from the VTA or GABAergic interneurons which are

also found within the SNc. In order to generate a cell model that mimics PD, it was first

necessary to assess the effect of toxins that are believed to emulate cell death mechanisms in PD.

Nigro-striatal organotypic co-cultures were treated with naphthazarin (60 µM, 80 µM), PSI (80

µM, 100 µM) and rotenone (50 µM, 70 µM) for 24 hrs. Each toxin, at varying concentrations,

increased the number of dead cells. Naphthazarin concentrations of 60 µM and 80 µM increased

propidium iodide fluorescence by 67.54 ± 20.96% and 183.94 ± 23.42%, respectively (both P <

0.05) (Fig. 5.11a). Neurons exposed to PSI (100 µM) exhibited a 55.81 ± 16.57% increase in

propidium iodide fluorescence (P < 0.01) (Fig. 5.11b). Rotenone (70 µM) exposure increased

propidium iodide fluorescence by 66.58 ± 15.71% (P < 0.05) (Fig. 5.11c). Neurons of the SNc

experiencing dysfunctional sub-cellular mechanisms associated with PD often lose their

dopaminergic phenotype. Given cells degenerate when exposed to all three toxins, it was

Page 183: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

168

necessary to determine if these neurons also loss their dopaminergic phenotype. Nigro-striatal

organotypic co-cultures were labelled with TH and GIRK2. Interestingly, neurons expressing

GIRK2 had reduced TH expression when exposed to naphthazarin, PSI and rotenone (Fig. 5.12).

Page 184: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

169

Figure 5.9. Representative images showing axon outgrowth projecting from the SNc to

STR. SNc and STR slices were plated on day 0 (D0) and imaged 24 days later (D24) (a) Phase

contrast image to show multiple axons originating from the SNc (top-right) projecting to the STR

(bottom-left). (b) On D24, cultures were fixed and immunolabelled with the synaptic marker

PSD-95. Neurons forming networks have strong punctate expression of PSD95 in their

processes. Scale bars = 20 µm.

Page 185: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

170

Phase Contrasta

PSD95 DAPI Mergeb

SNc

STR

Page 186: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

171

Figure 5.10. Expression of TH and GIRK2 in nigro-striatal cultures. Nigro-striatal co-

cultures were grown for 24 days before being fixed and immuno-labelled with antibodies against

GIRK2 and TH. (a) Nigral slices of the co-cultures strongly express GIRK2 within the soma.

Scale bar = 40 µm (b) Nigro-striatal co-cultures are fully functional as shown by the expression

of TH within the cell bodies and projections. Scale bars = 20 µm.

Page 187: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

172

aGIRK2 DAPI Merge

THb

DAPI Merge

Page 188: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

173

Figure 5.11. Effect of naphthazarin, PSI and rotenone on toxin-induced cell death. Co-

cultures (D24) were exposed to toxins for 24 hours. Cell death was measured using propidium

iodide (PI). PI fluorescence was measured using a fluorescent plate reader. (a) Effect of

naphthazarin (60 µM and 80 µM) on cell death. (b) Effect of PSI (80 µM and 100 µM) on cell

death. (c) Effect of rotenone (50 µM and 70 µM) on cell death. One-way ANOVA with

Dunnett’s multiple comparison test post-hoc. * P < 0.05, ** P < 0.01, *** P < 0.001. Overall

effect naphthazarin treatment: F2,1 = 6.01, PSI treatment: F2,11 = 8.10, rotenone treatment: F2,11 =

6.27.

Page 189: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

174

a b

c

Naphthazarin

Vehicle 60 µM 80 µM0

50

100

150

200

250

**

Prop

idiu

m Io

dide

Flu

ores

cenc

e (%

con

trol

)

Rotenone

Vehicle 50 µM 70 µM0

50

100

150

200 *

Prop

idiu

m Io

dide

Flu

ores

cenc

e (%

con

trol

)PSI

Vehicle 80 µM 100 µM0

50

100

150

200**

Prop

idiu

m Io

dide

Flu

ores

cenc

e (%

con

trol

)

Page 190: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

175

Figure 5.12. Effect of toxin exposure on TH expression in nigro-striatal organotypic co-

cultures. Co-cultures (D24) were incubated with toxins 24 hours prior to being fixed and

immunolabelled with antibodies against GIRK2 and TH. Images show the effect of naphthazarin

(80 µM), PSI (100 µM) and rotenone (70 µM) on TH expression in GIRK2 expressing cells.

Scale bars = 20 µm.

Page 191: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

176

DMSO

DAPIGIRK2 TH MergeNAP

PSI

ROT

Page 192: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

177

5.4 Discussion

Familial and sporadic PD are likely to have similar mechanisms underlying their pathology,

however a treatment that halts, slows or reverses PD has yet to be developed. To this end, I

developed and characterized cell models of PD that enable more rapid screening of potential

neuroprotective compounds in a relatively high throughput and cost effective manner. Whilst

these models are less physiological than in vivo models, I have shown that they are likely to have

clinical relevance.

Previously I validated an SH-SY5Y cell model of PD which proved to be effective in testing

potential neuroprotective agents (Yong-Kee et al., 2011b). Here, I utilized this same model to

test the neuroprotective effect of RGMa. Interestingly, RGMa was protective during dopamine

and rotenone toxicity, both highly relevant in PD pathology. Given that only rotenone and

dopamine toxicity was blocked, it is likely that RGMa protects cells by preventing ROS

generation. Here, the greatest protective effect of RGMa was during rotenone toxicity. Since

mitochondria are highly active organelles that generate large amounts of ROS, inhibition of

mitochondrial complex 1 would lead to a large rapid increase in ROS (Lin and Beal, 2006).

Preventing this increase may explain the higher protective effect seen during rotenone toxicity

than in dopamine toxicity. Studies revealed an interaction between RGMa and neogenin

receptors prevents the activation of proapoptotic pathways (Matsunaga and Chedotal, 2004). It

could be possible that RGMa blocks apoptosis given that toxic effects of dopamine and rotenone

are mediated by apoptotic processes (Hanrott et al., 2006; Imamura et al., 2006). However, a

study using neural precursor cells revealed the activation of apoptosis in response to lysosome

inhibition (Walls et al., 2010). Thus, if RGMa blocked apoptotic processes, it would be expected

that it would be effective during naphthazarin toxicity as well.

Page 193: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

178

The role of SIRT3 in mitochondria and cell survival made it a potential candidate for a

neuroprotective agent in PD. The effectiveness of SIRT3 as a neuroprotective agent was tested

in various in vitro models of PD. SIRT3 protected neuroblastoma and neurons from rotenone

toxicity. A study by Ahn et al. (2008) revealed the deacetylase activity of SIRT3 can enhance

the activity of mitochondrial complex 1. This in turn regulates the production of ATP levels in

the cell. Since mitochondrial activity at complex 1 is reduced by rotenone, it is feasible that the

actions of SIRT3 can reverse these effects on mitochondria. This explains how effective SIRT3

is at negating the effects of rotenone and not any of the other toxins tested. An alternative

function of SIRT3 can warrant the protective effect of SIRT3. A study in myotubes revealed the

SIRT3 dependent up-regulation of PGC1α leads to mitochondria biogenesis (Kong et al., 2010).

An up-regulation of mitochondria may compensate for damaged mitochondria which ultimately

prevents cell loss. These results prompted the examination of the effectiveness of SIRT3 during

LRRK2 induced toxicity. Given the effects of LRRK2 on mitochondria, SIRT3 would be a

prime candidate for neuroprotection in PD (Wang et al., 2012b). Indeed, SIRT3 prevented

LRRK2 induced cell death in differentiated SH-SY5Y cells. This necessitates further study of

SIRT3 to protect cells from dysfunction associated with mutant LRRK2. SIRT3 is effective at

protecting mitochondria which infers that it may be a valuable tool for other mitochondrial

disorders.

In vitro models utilizing cell lines are useful for high throughput experiments where multiple

compounds can be tested, but they lack physiological elements that are essential for more

rigorous testing of compounds. Thus, the more complex in vitro models, such as the organotypic

and primary cultures were characterized for various neuronal markers. Both the organotypic and

primary cultures used here express TH, which is a key component in the production of

dopamine. Other studies in ventral mesencephalic and organotypic cultures have shown the

Page 194: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

179

expression of TH and homovanillic acid, proteins involved in dopamine production (Lyng et al.,

2007; Zhang et al., 2007). Furthermore, these cultures are composed of neurons from the SNc,

as shown by prominent GIRK2 expression. Neurons that degenerate in the early stages of PD

are specific to the SNc and have a dopamine producing phenotype (Hornykiewicz, 1966; Obeso

et al., 2000). These findings make them an appropriate model, from a physiological standpoint,

to study PD. In vivo models benefit from the retention of synaptic networks when compared to

in vitro models. Indeed, the cultures used here form synaptic connections and have key

components of synapse formation. This is demonstrated by the expression of the synaptic

proteins PSD95 and synaptophysin. This model is in agreement with other studies that found

PSD95 and synaptophysin expression in organotypic and primary cultures (Buckby et al., 2004;

Wakita et al., 2010). The models studied here display characteristics similar to those of in vivo

models and benefit from relatively faster production. This in turn enables testing of various

compounds in an environment that is more physiological than those provided by cell lines, while

retaining easier production than animal models.

Toxin based in vitro and in vivo models have been used as tools to mimic the pathology seen in

sporadic PD. Most in vitro models lack the physiological environment that resembles PD, while

in vivo models are inconsistent at obtaining a complete loss of dopaminergic neurons (Betarbet et

al., 2000; Hoglinger et al., 2003b). It is apparent here that a variety of toxins produced a loss of

cells at specific concentrations in organotypic cultures. Cell death induced by naphthazarin, PSI

and rotenone is associated with a loss of TH specifically in SNc neurons. A similar study in

ventral mesencephalic cultures shows a loss of TH after MPTP exposure (Jakobsen et al., 2005).

Interestingly, rat models which fail to produce behavioural changes similar to those found in PD,

do not show a reduction in TH because these behavioural changes are associated with losses of

dopamine (Lapointe et al., 2004). A retraction of SNc axons and terminals occurs in patients, but

Page 195: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

180

the toxins used here do not lead to similar morphological changes. This could be a consequence

of the time cultures were exposed to the toxins, thus extended exposure times may allow for a

loss of axons. Indeed, multiple studies have shown axon degeneration in organotypic cultures

using rotenone incubation periods from three days to more than a week (Testa et al., 2005;

Ullrich and Humpel, 2009). This organotypic co-culture model is advantageous for studying

mechanisms related to PD because it employs toxins linked with PD pathology to consistently

produce a loss of dopaminergic neurons.

Current transgenic models suffer from the inability to consistently produce dopaminergic

degeneration (Meredith et al., 2008). Furthermore the only transgenic animals expressing α-

synuclein are mice; which makes it extremely difficult to dissect the nigra. Thus, I generated an

in vitro primary culture model that expresses α-synuclein. The α-synuclein primary culture

model used here repeatedly shows degeneration of GIRK2 expressing cells. This specificity is

important because it replicates the early stages of disease pathology, whereby nigral cells begin

to degenerate (Hornykiewicz, 1966; Obeso et al., 2000). Lewy bodies, which are composed of

α-synuclein and ubiquitin, do not form in these neurons. Lewy bodies are often associated with

PD linked with α-synuclein (Crowther et al., 2000; Saito et al., 2004). It is likely that increased

culture periods would allow for the formation of Lewy bodies because α-synuclein would have

more time to accumulate and aggregate. Since this model does not require transgenic animals, it

is a relatively cost-effective way to study pathology linked with PD.

These studies reveal that in vitro models are a valuable tool for studying PD because they are

easily produced and are similar to cells affected in PD. Furthermore, they display typical losses

of function and viability when presented with toxin and gene induced cell stressors. Granted

there are disadvantages to these models, their advantages are highly beneficial when used in a

Page 196: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

181

high throughput environment. This is demonstrated by the elucidation of the protective effect of

RGMa and SIRT3. The rescue of cells from mitochondrial dysfunction caused by toxins and

genetic over-expression implies that PD could be a mitochondrial disorder. Furthermore, RGMa

and SIRT3 may be important agents for treating other mitochondrial disorders.

Page 197: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

182

Chapter 6

Summary and Future Directions

Page 198: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

183

6 Summary and Future Directions

To conclude, following exposure to toxins that mimic PD pathology, UPS and lysosome function

are inhibited, consequently reducing mitochondrial function. Following genetic over-expression

of LRRK2 and G2019S-LRRK2, mitochondrial fusion becomes inhibited, resulting in

fragmentation of mitochondria. Compounds that primarily target mitochondrial function in

models that both mimic PD pathology and contain neurons that are similar to those which

degenerate in PD are essential for developing therapeutics to treat PD. The findings described

here suggest that alterations to mitochondria may be a central factor contributing to PD onset,

and that enhancing the function of mitochondria is a potential avenue for preventing

neurodegeneration causing PD.

Recently it has been shown that ROS production, as well as malfunctions to the UPS and

lysosome are causative factors of the disease (Cuervo et al., 2004; Betarbet et al., 2005). In

Chapter Two, studies reveal that malfunction of multiple sub-cellular systems inhibit

mitochondrial function early on during cell stress. More specifically, these experiments reveal a

tight link between the UPS, lysosomes and mitochondria, and that convergence of these

dysfunctional processes can be disastrous for cellular health. These experiments not only reveal

the convergence of multiple mechanisms, but they also present evidence that mitochondria are

sensitive to abnormalities in other sub-cellular systems, such as the UPS and lysosome. These

studies are the first to show a link between the mitochondria, UPS and lysosome. Moreover, the

link between the UPS and lysosome is further established here since prolonged inhibition of the

UPS or lysosome can impair the lysosome or UPS, respectively (Sherer et al., 2002). In future,

studies should examine the pathways leading to the interactions between the mitochondria, UPS

Page 199: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

184

and lysosome. The regulation of mitochondrial protein quality control by the UPS and the

removal of dysfunctional mitochondria by lysosomes are possible pathways interconnecting

these sub-cellular domains. These pathways can be studied by measuring the accumulation of

ubiquitnated proteins in mitochondrial fractions from cell lysates during proteasomal inhibition

or by measuring the number of mitochondria encapsulated by autophagosomes during lysosomal

inhibition. However, it is still unclear why mitochondria are specifically affected and why

mitochondria are affected so early during cellular stress when processes such as fission and

fusion are in place to rescue mitochondria. Since mitochondria provide energy to the cell, it is

possible that slight perturbations to mitochondrial function can have profound effects on neurons

that have high energy demands. Moreover, nigral cells are highly susceptible to changes in

homeostasis because they are constantly metabolizing dopamine, which produces large quantities

of ROS, thus, further production of ROS by mitochondrial damage may bring imbalance to

nigral cell homeostasis. This would also account for nigral neurons being unable to quickly

compensate for losses of mitochondrial function and ROS production. Thus, future studies

should compare the function of mitochondria and their effects on ROS in nigral and non-nigral

neurons to determine if nigral cells are more susceptible to these changes. It is clear from these

studies that mitochondria play an important role in disease pathogenesis.

Since multiple sub-cellular mechanisms are responsible for disease pathogenesis, it is necessary

for a model to incorporate all of these mechanisms. Moreover, such a model can be deemed

representative of the condition, if when tested against a battery of potential neuroprotective

agents, has a high degree of similarity when correlated to the outcomes of clinical trials. Indeed,

the outcomes of the neuroprotective assay used in Chapter Three are very similar to the results of

clinical trials where various neuroprotective agents were tested. In particular, coenzyme Q10

was highly protective during stress induced by multiple mechanisms, such as mitochondrial, UPS

Page 200: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

185

and lysosomal inhibition. A correlation between this study and clinical trials can be drawn since

coenzyme Q10 showed promise in clinical trials, thus validating the in vitro assay used here.

Substantial evidence for a causative role of mitochondria in PD pathology is provided by the

neuroprotective effect of coenzyme Q10. Since mitochondrial dysfunction results from UPS and

lysosomal inhibition, it is expected that a potential therapeutic which acts directly on

mitochondria would be highly protective, and indeed, coenzyme Q10 did protect cells from the

toxic effects of UPS and lysosomal inhibition. Coenzyme Q10 is an integral part of the electron

transport chain, and the protective effects seen here could be attributed to an increase in ATP

production. However, increased ROS production associated with inhibited respiratory activity

could be prevented when coenzyme Q10 restores oxidative phosphorylation. Thus, future

studies can examine if the aforementioned pathways are affected by blocking key molecules

essential for each pathway in the presence of coenzyme Q10 and then determine if cell viability

is enhanced. Alternatively, ATP and ROS levels can be measured when coenzyme Q10 is

present. These studies are the first to describe an in vitro model that is easy to produce and can

effectively test potential neuroprotective agents to treat PD. Moreover, it will enable rapid and

efficient testing of a wide range of compounds and make available more compounds for testing

in clinical trials. These studies suggest that for a therapeutic to have any potential to treat PD

patients it must enhance mitochondrial function or induce the biogenesis of mitochondria to

compensate for mitochondria that have lost function due to cell stress.

Recently research has redirected its focus back to the field of mitochondria by suggesting

mitochondrial dynamics is an important mechanism in neurodegenerative disease (Chen and

Chan, 2009). Thus, given the findings of my initial study, subsequent studies in my thesis

focused on elucidating the role of mitochondrial dynamics in PD. Mitochondrial dysfunction in

PD may arise from alterations to fission and fusion since these processes are imperative for

Page 201: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

186

proper mitochondrial function (Otera and Mihara, 2011). This hypothesis is confirmed in

Chapter Four by finding that mitochondrial fusion becomes inhibited by LRRK2 and G2019S-

LRRK2 expression which ultimately fragments mitochondria. Furthermore, it appears that

mitochondrial fragmentation occurs before cell stress occurs. Although mitochondrial

dysfunction was not shown during LRRK2 and G2019S-LRRK2 expression, it may be that

altered fusion is the first step leading to reduced membrane potential, and hence, dysfunctional

mitochondria. Indeed, mitochondrial fragmentation is a prerequisite for mitochondrial

dysfunction and eventual cell death (Frank et al., 2001; Breckenridge et al., 2003). While

increased mitochondrial fission could lead to fragmented mitochondria, it was not observed in

this study. This result however, may be specific to the time point used. At later time points

fission may indeed play a role in the fragmentation caused by LRRK2 and G2019S-LRRK2.

Given the discovery of altered fusion, future studies should focus on elucidating the changes that

occur in fusion proteins. It is possible that changes in the activity of Mfn1, Mfn2 and Opa1 are

regulated by the kinase function of LRRK2. Moreover, the GTPase activity of LRRK2 could

limit the available GTP required for fusion protein function since LRRK2, which is located on

the outer mitochondrial membrane, would be situated close to Mfn1 and Mfn2. Future studies

can delineate if the kinase or GTPase domain of LRRK2 is responsible for the inhibition of

fusion protein activity by knocking down these specific domains of LRRK2 and subsequently

measuring fusion rates. While studies suggest mitochondrial fission is key to abnormal

mitochondrial function, this study is the first to suggest that inhibition of mitochondrial fusion by

LRRK2 could affect mitochondrial function. It is possible that patients having a LRRK2 genetic

background may suffer neurodegeneration caused by abnormal mitochondrial fusion.

It is imperative for in vitro models to replicate the pathology found in PD as precisely as possible

so as to foster the efficient production of therapeutics. Various studies, including the ones

Page 202: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

187

described here, suggest compounds which target the mitochondria are highly effective at treating

PD. In Chapter Five, upon characterization of various in vitro models, neuronal cultures were

found to be very similar to the neurons that degenerate in the SNc of parkinsonian patients.

Moreover, the novel mitochondrial protein, SIRT3, protected neurons from degeneration

associated with cell stress induced by LRRK2 over-expression. The high therapeutic value of

SIRT3 can be concluded for two reasons. Firstly, the feasibility of SIRT3 as a neuroprotective

agent is confirmed since it was able to protect mature neurons having a dopaminergic phenotype

from degeneration. Secondly, since SIRT3 protected neurons from the toxic effects of LRRK2, a

genetic mutation that is common among PD patients, it has the potential to treat a broad

spectrum of patients with PD. The next logical study is to test SIRT3 in various animal models

that closely mimic the pathological mechanisms underlying PD. In addition to mitochondrial

dysfunction, these models should display ROS production and protein aggregation as these are

classical markers of PD pathology. Further studies should be conducted to examine the

mechanisms by which SIRT3 provides neuroprotection. Indeed two main roles of SIRT3 are

likely responsible for its therapeutic effect, one being the regulation of mitochondrial complex 1

function, and two being its role in mitochondrial biogenesis (Ahn et al., 2008; Cimen et al.,

2010; Kong et al., 2010). These pathways can be studied by measuring changes in ATP levels or

by quantifying levels of mitochondrial DNA during SIRT3 expression. These studies are the

first to show the protective effect of SIRT3 in a neuronal model that has a high degree of

similarity to the neurons that degenerate in the SNc. Furthermore, SIRT3 could have a broad use

in the clinic since it could be used to treat patients having either familial PD caused by LRRK2,

or idiopathic forms caused by environmental toxins.

Page 203: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

188

7 References

(2006) A randomized, double-blind, futility clinical trial of creatine and minocycline in early Parkinson disease. Neurology 66:664-671.

Aasly JO, Toft M, Fernandez-Mata I, Kachergus J, Hulihan M, White LR, Farrer M (2005) Clinical features of LRRK2-associated Parkinson's disease in central Norway. Ann Neurol 57:762-765.

Abou-Sleiman PM, Healy DG, Wood NW (2004) Causes of Parkinson's disease: genetics of DJ-1. Cell and tissue research 318:185-188.

Ahn BH, Kim HS, Song S, Lee IH, Liu J, Vassilopoulos A, Deng CX, Finkel T (2008) A role for the mitochondrial deacetylase Sirt3 in regulating energy homeostasis. Proc Natl Acad Sci U S A 105:14447-14452.

Alberio T, Lopiano L, Fasano M (2012) Cellular models to investigate biochemical pathways in Parkinson's disease. The FEBS journal 279:1146-1155.

Alexander C, Votruba M, Pesch UE, Thiselton DL, Mayer S, Moore A, Rodriguez M, Kellner U, Leo-Kottler B, Auburger G, Bhattacharya SS, Wissinger B (2000) OPA1, encoding a dynamin-related GTPase, is mutated in autosomal dominant optic atrophy linked to chromosome 3q28. Nature genetics 26:211-215.

Andrade MA, Perez-Iratxeta C, Ponting CP (2001) Protein repeats: structures, functions, and evolution. Journal of structural biology 134:117-131.

Ardley HC, Scott GB, Rose SA, Tan NG, Robinson PA (2004) UCH-L1 aggresome formation in response to proteasome impairment indicates a role in inclusion formation in Parkinson's disease. J Neurochem 90:379-391.

Ardley HC, Scott GB, Rose SA, Tan NG, Markham AF, Robinson PA (2003) Inhibition of proteasomal activity causes inclusion formation in neuronal and non-neuronal cells overexpressing Parkin. Molecular biology of the cell 14:4541-4556.

Baloh RH, Schmidt RE, Pestronk A, Milbrandt J (2007) Altered axonal mitochondrial transport in the pathogenesis of Charcot-Marie-Tooth disease from mitofusin 2 mutations. J Neurosci 27:422-430.

Bao J, Lu Z, Joseph JJ, Carabenciov D, Dimond CC, Pang L, Samsel L, McCoy JP, Jr., Leclerc J, Nguyen P, Gius D, Sack MN (2010) Characterization of the murine SIRT3 mitochondrial localization sequence and comparison of mitochondrial enrichment and deacetylase activity of long and short SIRT3 isoforms. Journal of cellular biochemistry 110:238-247.

Page 204: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

189

Barsoum MJ, Yuan H, Gerencser AA, Liot G, Kushnareva Y, Graber S, Kovacs I, Lee WD, Waggoner J, Cui J, White AD, Bossy B, Martinou JC, Youle RJ, Lipton SA, Ellisman MH, Perkins GA, Bossy-Wetzel E (2006) Nitric oxide-induced mitochondrial fission is regulated by dynamin-related GTPases in neurons. The EMBO journal 25:3900-3911.

Bence NF, Sampat RM, Kopito RR (2001) Impairment of the ubiquitin-proteasome system by protein aggregation. Science 292:1552-1555.

Bender A, Koch W, Elstner M, Schombacher Y, Bender J, Moeschl M, Gekeler F, Muller-Myhsok B, Gasser T, Tatsch K, Klopstock T (2006) Creatine supplementation in Parkinson disease: a placebo-controlled randomized pilot trial. Neurology 67:1262-1264.

Bernheimer H, Birkmayer W, Hornykiewicz O, Jellinger K, Seitelberger F (1973) Brain dopamine and the syndromes of Parkinson and Huntington. Clinical, morphological and neurochemical correlations. J Neurol Sci 20:415-455.

Betarbet R, Sherer TB, Greenamyre JT (2002) Animal models of Parkinson's disease. BioEssays : news and reviews in molecular, cellular and developmental biology 24:308-318.

Betarbet R, Sherer TB, Greenamyre JT (2005) Ubiquitin-proteasome system and Parkinson's diseases. Exp Neurol 191 Suppl 1:S17-27.

Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT (2000) Chronic systemic pesticide exposure reproduces features of Parkinson's disease. Nature neuroscience 3:1301-1306.

Biedler JL, Roffler-Tarlov S, Schachner M, Freedman LS (1978) Multiple neurotransmitter synthesis by human neuroblastoma cell lines and clones. Cancer research 38:3751-3757.

Biskup S, Moore DJ, Celsi F, Higashi S, West AB, Andrabi SA, Kurkinen K, Yu SW, Savitt JM, Waldvogel HJ, Faull RL, Emson PC, Torp R, Ottersen OP, Dawson TM, Dawson VL (2006) Localization of LRRK2 to membranous and vesicular structures in mammalian brain. Ann Neurol 60:557-569.

Blackinton JG, Anvret A, Beilina A, Olson L, Cookson MR, Galter D (2007) Expression of PINK1 mRNA in human and rodent brain and in Parkinson's disease. Brain research 1184:10-16.

Blum D, Torch S, Lambeng N, Nissou M, Benabid AL, Sadoul R, Verna JM (2001) Molecular pathways involved in the neurotoxicity of 6-OHDA, dopamine and MPTP: contribution to the apoptotic theory in Parkinson's disease. Progress in neurobiology 65:135-172.

Boni LT, Hah JS, Hui SW, Mukherjee P, Ho JT, Jung CY (1984) Aggregation and fusion of unilamellar vesicles by poly(ethylene glycol). Biochimica et biophysica acta 775:409-418.

Bonifati V, Oostra BA, Heutink P (2004) Linking DJ-1 to neurodegeneration offers novel insights for understanding the pathogenesis of Parkinson's disease. J Mol Med (Berl) 82:163-174.

Page 205: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

190

Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, Krieger E, Dekker MC, Squitieri F, Ibanez P, Joosse M, van Dongen JW, Vanacore N, van Swieten JC, Brice A, Meco G, van Duijn CM, Oostra BA, Heutink P (2003) Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 299:256-259.

Bosgraaf L, Van Haastert PJ (2003) Roc, a Ras/GTPase domain in complex proteins. Biochimica et biophysica acta 1643:5-10.

Braak H, Braak E, Yilmazer D, de Vos RA, Jansen EN, Bohl J (1996) Pattern of brain destruction in Parkinson's and Alzheimer's diseases. J Neural Transm 103:455-490.

Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E (2003) Staging of brain pathology related to sporadic Parkinson's disease. Neurobiology of aging 24:197-211.

Brannan T, Bhardwaj A, Martinez-Tica J, Weinberger J, Yahr M (1990) Striatal L-dopa metabolism studied in vivo in rats with nigrostriatal lesions. Journal of neural transmission Parkinson's disease and dementia section 2:15-22.

Breckenridge DG, Stojanovic M, Marcellus RC, Shore GC (2003) Caspase cleavage product of BAP31 induces mitochondrial fission through endoplasmic reticulum calcium signals, enhancing cytochrome c release to the cytosol. J Cell Biol 160:1115-1127.

Brinks H, Conrad S, Vogt J, Oldekamp J, Sierra A, Deitinghoff L, Bechmann I, Alvarez-Bolado G, Heimrich B, Monnier PP, Mueller BK, Skutella T (2004) The repulsive guidance molecule RGMa is involved in the formation of afferent connections in the dentate gyrus. J Neurosci 24:3862-3869.

Buckby LE, Mummery R, Crompton MR, Beesley PW, Empson RM (2004) Comparison of neuroplastin and synaptic marker protein expression in acute and cultured organotypic hippocampal slices from rat. Brain research Developmental brain research 150:1-7.

Busija DW, Gaspar T, Domoki F, Katakam PV, Bari F (2008) Mitochondrial-mediated suppression of ROS production upon exposure of neurons to lethal stress: mitochondrial targeted preconditioning. Adv Drug Deliv Rev 60:1471-1477.

Canet-Aviles RM, Wilson MA, Miller DW, Ahmad R, McLendon C, Bandyopadhyay S, Baptista MJ, Ringe D, Petsko GA, Cookson MR (2004) The Parkinson's disease protein DJ-1 is neuroprotective due to cysteine-sulfinic acid-driven mitochondrial localization. Proc Natl Acad Sci U S A 101:9103-9108.

Cannon JR, Greenamyre JT (2010) Neurotoxic in vivo models of Parkinson's disease recent advances. Progress in brain research 184:17-33.

Cannon JR, Greenamyre JT (2012) Gene-environment interactions in Parkinson's disease: Specific evidence in humans and mammalian models. Neurobiol Dis.

Cardozo DL (1993) Midbrain dopaminergic neurons from postnatal rat in long-term primary culture. Neuroscience 56:409-421.

Page 206: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

191

Carlsson A, Lindqvist M, Magnusson T (1957) 3,4-Dihydroxyphenylalanine and 5-hydroxytryptophan as reserpine antagonists. Nature 180:1200.

Carman LS, Gage FH, Shults CW (1991) Partial lesion of the substantia nigra: relation between extent of lesion and rotational behavior. Brain research 553:275-283.

Chaudhuri KR, Healy DG, Schapira AH (2006) Non-motor symptoms of Parkinson's disease: diagnosis and management. Lancet neurology 5:235-245.

Chen H, Chan DC (2009) Mitochondrial dynamics--fusion, fission, movement, and mitophagy--in neurodegenerative diseases. Human molecular genetics 18:R169-176.

Chen H, Chomyn A, Chan DC (2005) Disruption of fusion results in mitochondrial heterogeneity and dysfunction. J Biol Chem 280:26185-26192.

Chen L, Ding Y, Cagniard B, Van Laar AD, Mortimer A, Chi W, Hastings TG, Kang UJ, Zhuang X (2008) Unregulated cytosolic dopamine causes neurodegeneration associated with oxidative stress in mice. J Neurosci 28:425-433.

Chesselet MF, Richter F, Zhu C, Magen I, Watson MB, Subramaniam SR (2012) A progressive mouse model of Parkinson's disease: the Thy1-aSyn ("Line 61") mice. Neurotherapeutics 9:297-314.

Cho HS, Kim S, Lee SY, Park JA, Kim SJ, Chun HS (2008) Protective effect of the green tea component, L-theanine on environmental toxins-induced neuronal cell death. Neurotoxicology 29:656-662.

Chu Y, Dodiya H, Aebischer P, Olanow CW, Kordower JH (2009) Alterations in lysosomal and proteasomal markers in Parkinson's disease: relationship to alpha-synuclein inclusions. Neurobiol Dis 35:385-398.

Chung WG, Miranda CL, Maier CS (2007) Epigallocatechin gallate (EGCG) potentiates the cytotoxicity of rotenone in neuroblastoma SH-SY5Y cells. Brain Res 1176:133-142.

Cimen H, Han MJ, Yang Y, Tong Q, Koc H, Koc EC (2010) Regulation of succinate dehydrogenase activity by SIRT3 in mammalian mitochondria. Biochemistry 49:304-311.

Coelho M, Ferreira JJ (2012) Late-stage Parkinson disease. Nature reviews Neurology.

Conway KA, Harper JD, Lansbury PT (1998) Accelerated in vitro fibril formation by a mutant alpha-synuclein linked to early-onset Parkinson disease. Nature medicine 4:1318-1320.

Conway KA, Lee SJ, Rochet JC, Ding TT, Williamson RE, Lansbury PT, Jr. (2000) Acceleration of oligomerization, not fibrillization, is a shared property of both alpha-synuclein mutations linked to early-onset Parkinson's disease: implications for pathogenesis and therapy. Proc Natl Acad Sci U S A 97:571-576.

Cookson MR (2003) Parkin's substrates and the pathways leading to neuronal damage. Neuromolecular Med 3:1-13.

Page 207: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

192

Cookson MR (2005) The biochemistry of Parkinson's disease. Annual review of biochemistry 74:29-52.

Cooper AA, Gitler AD, Cashikar A, Haynes CM, Hill KJ, Bhullar B, Liu K, Xu K, Strathearn KE, Liu F, Cao S, Caldwell KA, Caldwell GA, Marsischky G, Kolodner RD, Labaer J, Rochet JC, Bonini NM, Lindquist S (2006) Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's models. Science 313:324-328.

Cooper HM, Spelbrink JN (2008) The human SIRT3 protein deacetylase is exclusively mitochondrial. The Biochemical journal 411:279-285.

Crowther RA, Daniel SE, Goedert M (2000) Characterisation of isolated alpha-synuclein filaments from substantia nigra of Parkinson's disease brain. Neuroscience letters 292:128-130.

Cuervo AM, Stefanis L, Fredenburg R, Lansbury PT, Sulzer D (2004) Impaired degradation of mutant alpha-synuclein by chaperone-mediated autophagy. Science 305:1292-1295.

Cuervo AM, Bergamini E, Brunk UT, Droge W, Ffrench M, Terman A (2005) Autophagy and aging: the importance of maintaining "clean" cells. Autophagy 1:131-140.

Dadakhujaev S, Noh HS, Jung EJ, Cha JY, Baek SM, Ha JH, Kim DR (2010) Autophagy protects the rotenone-induced cell death in alpha-synuclein overexpressing SH-SY5Y cells. Neurosci Lett 472:47-52.

Darrouzet E, Issartel JP, Lunardi J, Dupuis A (1998) The 49-kDa subunit of NADH-ubiquinone oxidoreductase (Complex I) is involved in the binding of piericidin and rotenone, two quinone-related inhibitors. FEBS letters 431:34-38.

Dauer W, Przedborski S (2003) Parkinson's disease: mechanisms and models. Neuron 39:889-909.

Dawson TM, Ko HS, Dawson VL (2010) Genetic animal models of Parkinson's disease. Neuron 66:646-661.

de Rijk MC, Launer LJ, Berger K, Breteler MM, Dartigues JF, Baldereschi M, Fratiglioni L, Lobo A, Martinez-Lage J, Trenkwalder C, Hofman A (2000) Prevalence of Parkinson's disease in Europe: A collaborative study of population-based cohorts. Neurologic Diseases in the Elderly Research Group. Neurology 54:S21-23.

Dehay B, Ramirez A, Martinez-Vicente M, Perier C, Canron MH, Doudnikoff E, Vital A, Vila M, Klein C, Bezard E (2012) Loss of P-type ATPase ATP13A2/PARK9 function induces general lysosomal deficiency and leads to Parkinson disease neurodegeneration. Proc Natl Acad Sci U S A 109:9611-9616.

Den Oudsten BL, Lucas-Carrasco R, Green AM (2011) Perceptions of persons with Parkinson's disease, family and professionals on quality of life: an international focus group study. Disability and rehabilitation 33:2490-2508.

Page 208: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

193

Detmer SA, Chan DC (2007) Functions and dysfunctions of mitochondrial dynamics. Nature reviews Molecular cell biology 8:870-879.

Di Fonzo A, Rohe CF, Ferreira J, Chien HF, Vacca L, Stocchi F, Guedes L, Fabrizio E, Manfredi M, Vanacore N, Goldwurm S, Breedveld G, Sampaio C, Meco G, Barbosa E, Oostra BA, Bonifati V (2005) A frequent LRRK2 gene mutation associated with autosomal dominant Parkinson's disease. Lancet 365:412-415.

Di Fonzo A et al. (2007) ATP13A2 missense mutations in juvenile parkinsonism and young onset Parkinson disease. Neurology 68:1557-1562.

Dodson MW, Guo M (2007) Pink1, Parkin, DJ-1 and mitochondrial dysfunction in Parkinson's disease. Current opinion in neurobiology 17:331-337.

Ebrahimi-Fakhari D, Cantuti-Castelvetri I, Fan Z, Rockenstein E, Masliah E, Hyman BT, McLean PJ, Unni VK (2011) Distinct roles in vivo for the ubiquitin-proteasome system and the autophagy-lysosomal pathway in the degradation of alpha-synuclein. J Neurosci 31:14508-14520.

Estaquier J, Arnoult D (2007) Inhibiting Drp1-mediated mitochondrial fission selectively prevents the release of cytochrome c during apoptosis. Cell death and differentiation 14:1086-1094.

Fahn S, Oakes D, Shoulson I, Kieburtz K, Rudolph A, Lang A, Olanow CW, Tanner C, Marek K (2004) Levodopa and the progression of Parkinson's disease. The New England journal of medicine 351:2498-2508.

Farrer M, Stone J, Mata IF, Lincoln S, Kachergus J, Hulihan M, Strain KJ, Maraganore DM (2005) LRRK2 mutations in Parkinson disease. Neurology 65:738-740.

Farrer M, Kachergus J, Forno L, Lincoln S, Wang DS, Hulihan M, Maraganore D, Gwinn-Hardy K, Wszolek Z, Dickson D, Langston JW (2004) Comparison of kindreds with parkinsonism and alpha-synuclein genomic multiplications. Ann Neurol 55:174-179.

Farrer M, Chan P, Chen R, Tan L, Lincoln S, Hernandez D, Forno L, Gwinn-Hardy K, Petrucelli L, Hussey J, Singleton A, Tanner C, Hardy J, Langston JW (2001) Lewy bodies and parkinsonism in families with parkin mutations. Ann Neurol 50:293-300.

Frank S, Gaume B, Bergmann-Leitner ES, Leitner WW, Robert EG, Catez F, Smith CL, Youle RJ (2001) The role of dynamin-related protein 1, a mediator of mitochondrial fission, in apoptosis. Dev Cell 1:515-525.

Galter D, Westerlund M, Carmine A, Lindqvist E, Sydow O, Olson L (2006) LRRK2 expression linked to dopamine-innervated areas. Ann Neurol 59:714-719.

Gan L, Mucke L (2008) Paths of convergence: sirtuins in aging and neurodegeneration. Neuron 58:10-14.

Page 209: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

194

Gao HM, Kotzbauer PT, Uryu K, Leight S, Trojanowski JQ, Lee VM (2008) Neuroinflammation and oxidation/nitration of alpha-synuclein linked to dopaminergic neurodegeneration. J Neurosci 28:7687-7698.

Gidday JM (2006) Cerebral preconditioning and ischaemic tolerance. Nat Rev Neurosci 7:437-448.

Gilks WP, Abou-Sleiman PM, Gandhi S, Jain S, Singleton A, Lees AJ, Shaw K, Bhatia KP, Bonifati V, Quinn NP, Lynch J, Healy DG, Holton JL, Revesz T, Wood NW (2005) A common LRRK2 mutation in idiopathic Parkinson's disease. Lancet 365:415-416.

Giralt A, Villarroya F (2012) SIRT3, a pivotal actor in mitochondrial functions: metabolism, cell death and aging. The Biochemical journal 444:1-10.

Golbe LI, Di Iorio G, Bonavita V, Miller DC, Duvoisin RC (1990) A large kindred with autosomal dominant Parkinson's disease. Ann Neurol 27:276-282.

Goldstein M, Lieberman AN, Helmer E, Koslow M, Ransohoff J, Elsworth JD, Roth RH, Deutch AY (1988) Biochemical analysis of caudate nucleus biopsy samples from parkinsonian patients. Ann Neurol 24:685-688.

Gomez C, Bandez MJ, Navarro A (2007) Pesticides and impairment of mitochondrial function in relation with the parkinsonian syndrome. Frontiers in bioscience : a journal and virtual library 12:1079-1093.

Greenamyre JT, Hastings TG (2004) Biomedicine. Parkinson's--divergent causes, convergent mechanisms. Science 304:1120-1122.

Greggio E, Jain S, Kingsbury A, Bandopadhyay R, Lewis P, Kaganovich A, van der Brug MP, Beilina A, Blackinton J, Thomas KJ, Ahmad R, Miller DW, Kesavapany S, Singleton A, Lees A, Harvey RJ, Harvey K, Cookson MR (2006) Kinase activity is required for the toxic effects of mutant LRRK2/dardarin. Neurobiol Dis 23:329-341.

Guzman JN, Sanchez-Padilla J, Wokosin D, Kondapalli J, Ilijic E, Schumacker PT, Surmeier DJ (2010) Oxidant stress evoked by pacemaking in dopaminergic neurons is attenuated by DJ-1. Nature 468:696-700.

Halliwell B (2006) Oxidative stress and neurodegeneration: where are we now? J Neurochem 97:1634-1658.

Hampshire DJ, Roberts E, Crow Y, Bond J, Mubaidin A, Wriekat AL, Al-Din A, Woods CG (2001) Kufor-Rakeb syndrome, pallido-pyramidal degeneration with supranuclear upgaze paresis and dementia, maps to 1p36. Journal of medical genetics 38:680-682.

Hanrott K, Gudmunsen L, O'Neill MJ, Wonnacott S (2006) 6-hydroxydopamine-induced apoptosis is mediated via extracellular auto-oxidation and caspase 3-dependent activation of protein kinase Cdelta. J Biol Chem 281:5373-5382.

Page 210: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

195

Haque ME, Thomas KJ, D'Souza C, Callaghan S, Kitada T, Slack RS, Fraser P, Cookson MR, Tandon A, Park DS (2008) Cytoplasmic Pink1 activity protects neurons from dopaminergic neurotoxin MPTP. Proc Natl Acad Sci U S A 105:1716-1721.

Hara T, Nakamura K, Matsui M, Yamamoto A, Nakahara Y, Suzuki-Migishima R, Yokoyama M, Mishima K, Saito I, Okano H, Mizushima N (2006) Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441:885-889.

Hardy J (2010) Genetic analysis of pathways to Parkinson disease. Neuron 68:201-206.

Hatano T, Kubo S, Sato S, Hattori N (2009) Pathogenesis of familial Parkinson's disease: new insights based on monogenic forms of Parkinson's disease. J Neurochem 111:1075-1093.

Hatano T, Kubo S, Imai S, Maeda M, Ishikawa K, Mizuno Y, Hattori N (2007) Leucine-rich repeat kinase 2 associates with lipid rafts. Human molecular genetics 16:678-690.

Healy DG, Abou-Sleiman PM, Wood NW (2004) Genetic causes of Parkinson's disease: UCHL-1. Cell and tissue research 318:189-194.

Healy DG et al. (2008) Phenotype, genotype, and worldwide genetic penetrance of LRRK2-associated Parkinson's disease: a case-control study. Lancet neurology 7:583-590.

Heemskerk J, Tobin AJ, Ravina B (2002) From chemical to drug: neurodegeneration drug screening and the ethics of clinical trials. Nature neuroscience 5 Suppl:1027-1029.

Higashi S, Biskup S, West AB, Trinkaus D, Dawson VL, Faull RL, Waldvogel HJ, Arai H, Dawson TM, Moore DJ, Emson PC (2007) Localization of Parkinson's disease-associated LRRK2 in normal and pathological human brain. Brain research 1155:208-219.

Ho CC, Rideout HJ, Ribe E, Troy CM, Dauer WT (2009) The Parkinson disease protein leucine-rich repeat kinase 2 transduces death signals via Fas-associated protein with death domain and caspase-8 in a cellular model of neurodegeneration. J Neurosci 29:1011-1016.

Hochstrasser M (1992) Ubiquitin and intracellular protein degradation. Curr Opin Cell Biol 4:1024-1031.

Hoglinger GU, Carrard G, Michel PP, Medja F, Lombes A, Ruberg M, Friguet B, Hirsch EC (2003a) Dysfunction of mitochondrial complex I and the proteasome: interactions between two biochemical deficits in a cellular model of Parkinson's disease. J Neurochem 86:1297-1307.

Hoglinger GU, Feger J, Prigent A, Michel PP, Parain K, Champy P, Ruberg M, Oertel WH, Hirsch EC (2003b) Chronic systemic complex I inhibition induces a hypokinetic multisystem degeneration in rats. Journal of neurochemistry 84:491-502.

Hornykiewicz O (1966) Dopamine (3-hydroxytyramine) and brain function. Pharmacological reviews 18:925-964.

Page 211: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

196

Hornykiewicz O (1972) Dopamine and extrapyramidal motor function and dysfunction. Res Publ Assoc Res Nerv Ment Dis 50:390-415.

Houlden H, Singleton AB (2012) The genetics and neuropathology of Parkinson's disease. Acta neuropathologica.

Hung AY, Schwarzschild MA (2007) Clinical trials for neuroprotection in Parkinson's disease: overcoming angst and futility? Current opinion in neurology 20:477-483.

Ikeda F, Dikic I (2008) Atypical ubiquitin chains: new molecular signals. 'Protein Modifications: Beyond the Usual Suspects' review series. EMBO reports 9:536-542.

Imamura K, Takeshima T, Kashiwaya Y, Nakaso K, Nakashima K (2006) D-beta-hydroxybutyrate protects dopaminergic SH-SY5Y cells in a rotenone model of Parkinson's disease. Journal of neuroscience research 84:1376-1384.

Irrcher I et al. (2010) Loss of the Parkinson's disease-linked gene DJ-1 perturbs mitochondrial dynamics. Hum Mol Genet 19:3734-3746.

Ishihara N, Nomura M, Jofuku A, Kato H, Suzuki SO, Masuda K, Otera H, Nakanishi Y, Nonaka I, Goto Y, Taguchi N, Morinaga H, Maeda M, Takayanagi R, Yokota S, Mihara K (2009) Mitochondrial fission factor Drp1 is essential for embryonic development and synapse formation in mice. Nature cell biology 11:958-966.

Ito G, Okai T, Fujino G, Takeda K, Ichijo H, Katada T, Iwatsubo T (2007) GTP binding is essential to the protein kinase activity of LRRK2, a causative gene product for familial Parkinson's disease. Biochemistry 46:1380-1388.

Jakobsen B, Gramsbergen JB, Moller Dall A, Rosenblad C, Zimmer J (2005) Characterization of organotypic ventral mesencephalic cultures from embryonic mice and protection against MPP toxicity by GDNF. The European journal of neuroscience 21:2939-2948.

Jaleel M, Nichols RJ, Deak M, Campbell DG, Gillardon F, Knebel A, Alessi DR (2007) LRRK2 phosphorylates moesin at threonine-558: characterization of how Parkinson's disease mutants affect kinase activity. The Biochemical journal 405:307-317.

James DI, Parone PA, Mattenberger Y, Martinou JC (2003) hFis1, a novel component of the mammalian mitochondrial fission machinery. J Biol Chem 278:36373-36379.

Jenner P (1992) What process causes nigral cell death in Parkinson's disease? Neurologic clinics 10:387-403.

Jenner P (1993) Altered mitochondrial function, iron metabolism and glutathione levels in Parkinson's disease. Acta Neurol Scand Suppl 146:6-13.

Jenner P (2003) Oxidative stress in Parkinson's disease. Ann Neurol 53 Suppl 3:S26-36; discussion S36-28.

Page 212: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

197

Jenner P, Olanow CW (1998) Understanding cell death in Parkinson's disease. Ann Neurol 44:S72-84.

Jiang H, Ren Y, Zhao J, Feng J (2004) Parkin protects human dopaminergic neuroblastoma cells against dopamine-induced apoptosis. Hum Mol Genet 13:1745-1754.

Kamp F, Exner N, Lutz AK, Wender N, Hegermann J, Brunner B, Nuscher B, Bartels T, Giese A, Beyer K, Eimer S, Winklhofer KF, Haass C (2010) Inhibition of mitochondrial fusion by alpha-synuclein is rescued by PINK1, Parkin and DJ-1. The EMBO journal 29:3571-3589.

Kanda S, Bishop JF, Eglitis MA, Yang Y, Mouradian MM (2000) Enhanced vulnerability to oxidative stress by alpha-synuclein mutations and C-terminal truncation. Neuroscience 97:279-284.

Kane LA, Youle RJ (2011) PINK1 and Parkin flag Miro to direct mitochondrial traffic. Cell 147:721-723.

Kawajiri S, Saiki S, Sato S, Hattori N (2011) Genetic mutations and functions of PINK1. Trends in pharmacological sciences 32:573-580.

Kett LR, Dauer WT (2012) Leucine-rich repeat kinase 2 for beginners: six key questions. Cold Spring Harbor perspectives in medicine 2:a009407.

Kettern N, Dreiseidler M, Tawo R, Hohfeld J (2010) Chaperone-assisted degradation: multiple paths to destruction. Biol Chem 391:481-489.

Kim SH, Lu HF, Alano CC (2011) Neuronal Sirt3 protects against excitotoxic injury in mouse cortical neuron culture. PLoS One 6:e14731.

Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N (1998) Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 392:605-608.

Klein C, Lohmann-Hedrich K, Rogaeva E, Schlossmacher MG, Lang AE (2007) Deciphering the role of heterozygous mutations in genes associated with parkinsonism. Lancet neurology 6:652-662.

Knott AB, Perkins G, Schwarzenbacher R, Bossy-Wetzel E (2008) Mitochondrial fragmentation in neurodegeneration. Nature reviews Neuroscience 9:505-518.

Koch A, Lehmann-Horn K, Dachsel JC, Gasser T, Kahle PJ, Lucking CB (2009) Proteasomal inhibition reduces parkin mRNA in PC12 and SH-SY5Y cells. Parkinsonism Relat Disord 15:220-225.

Koller WC (1992) When does Parkinson's disease begin? Neurology 42:27-31; discussion 41-28.

Page 213: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

198

Komatsu M, Waguri S, Chiba T, Murata S, Iwata J, Tanida I, Ueno T, Koike M, Uchiyama Y, Kominami E, Tanaka K (2006) Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature 441:880-884.

Kong X, Wang R, Xue Y, Liu X, Zhang H, Chen Y, Fang F, Chang Y (2010) Sirtuin 3, a new target of PGC-1alpha, plays an important role in the suppression of ROS and mitochondrial biogenesis. PloS one 5:e11707.

Koprich JB, Johnston TH, Reyes MG, Sun X, Brotchie JM (2010) Expression of human A53T alpha-synuclein in the rat substantia nigra using a novel AAV1/2 vector produces a rapidly evolving pathology with protein aggregation, dystrophic neurite architecture and nigrostriatal degeneration with potential to model the pathology of Parkinson's disease. Molecular neurodegeneration 5:43.

Korolchuk VI, Mansilla A, Menzies FM, Rubinsztein DC (2009) Autophagy inhibition compromises degradation of ubiquitin-proteasome pathway substrates. Molecular cell 33:517-527.

Korr D, Toschi L, Donner P, Pohlenz HD, Kreft B, Weiss B (2006) LRRK1 protein kinase activity is stimulated upon binding of GTP to its Roc domain. Cellular signalling 18:910-920.

Kubota H (2009) Quality control against misfolded proteins in the cytosol: a network for cell survival. J Biochem 146:609-616.

Kumazawa R, Tomiyama H, Li Y, Imamichi Y, Funayama M, Yoshino H, Yokochi F, Fukusako T, Takehisa Y, Kashihara K, Kondo T, Elibol B, Bostantjopoulou S, Toda T, Takahashi H, Yoshii F, Mizuno Y, Hattori N (2008) Mutation analysis of the PINK1 gene in 391 patients with Parkinson disease. Archives of neurology 65:802-808.

Lannuzel A, Michel PP, Hoglinger GU, Champy P, Jousset A, Medja F, Lombes A, Darios F, Gleye C, Laurens A, Hocquemiller R, Hirsch EC, Ruberg M (2003) The mitochondrial complex I inhibitor annonacin is toxic to mesencephalic dopaminergic neurons by impairment of energy metabolism. Neuroscience 121:287-296.

Lapointe N, St-Hilaire M, Martinoli MG, Blanchet J, Gould P, Rouillard C, Cicchetti F (2004) Rotenone induces non-specific central nervous system and systemic toxicity. FASEB journal : official publication of the Federation of American Societies for Experimental Biology 18:717-719.

LaVoie MJ, Cortese GP, Ostaszewski BL, Schlossmacher MG (2007) The effects of oxidative stress on parkin and other E3 ligases. J Neurochem 103:2354-2368.

Lavu S, Boss O, Elliott PJ, Lambert PD (2008) Sirtuins--novel therapeutic targets to treat age-associated diseases. Nature reviews Drug discovery 7:841-853.

Lee BD, Dawson VL, Dawson TM (2012) Leucine-rich repeat kinase 2 (LRRK2) as a potential therapeutic target in Parkinson's disease. Trends in pharmacological sciences 33:365-373.

Page 214: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

199

Lee VM, Trojanowski JQ (2006) Mechanisms of Parkinson's disease linked to pathological alpha-synuclein: new targets for drug discovery. Neuron 52:33-38.

Lees AJ, Singleton AB (2007) Clinical heterogeneity of ATP13A2 linked disease (Kufor-Rakeb) justifies a PARK designation. Neurology 68:1553-1554.

Leigh PN, Probst A, Dale GE, Power DP, Brion JP, Dodson A, Anderton BH (1989) New aspects of the pathology of neurodegenerative disorders as revealed by ubiquitin antibodies. Acta Neuropathol 79:61-72.

Leroy E, Boyer R, Auburger G, Leube B, Ulm G, Mezey E, Harta G, Brownstein MJ, Jonnalagada S, Chernova T, Dehejia A, Lavedan C, Gasser T, Steinbach PJ, Wilkinson KD, Polymeropoulos MH (1998) The ubiquitin pathway in Parkinson's disease. Nature 395:451-452.

Lesage S, Durr A, Tazir M, Lohmann E, Leutenegger AL, Janin S, Pollak P, Brice A (2006) LRRK2 G2019S as a cause of Parkinson's disease in North African Arabs. The New England journal of medicine 354:422-423.

Li X, Patel JC, Wang J, Avshalumov MV, Nicholson C, Buxbaum JD, Elder GA, Rice ME, Yue Z (2010) Enhanced striatal dopamine transmission and motor performance with LRRK2 overexpression in mice is eliminated by familial Parkinson's disease mutation G2019S. J Neurosci 30:1788-1797.

Li Z, Okamoto K, Hayashi Y, Sheng M (2004) The importance of dendritic mitochondria in the morphogenesis and plasticity of spines and synapses. Cell 119:873-887.

Lin MT, Beal MF (2006) Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature 443:787-795.

Liu CW, Giasson BI, Lewis KA, Lee VM, Demartino GN, Thomas PJ (2005) A precipitating role for truncated alpha-synuclein and the proteasome in alpha-synuclein aggregation: implications for pathogenesis of Parkinson disease. J Biol Chem 280:22670-22678.

Liu Y, Lashuel HA, Choi S, Xing X, Case A, Ni J, Yeh LA, Cuny GD, Stein RL, Lansbury PT, Jr. (2003) Discovery of inhibitors that elucidate the role of UCH-L1 activity in the H1299 lung cancer cell line. Chemistry & biology 10:837-846.

Lowe J, McDermott H, Landon M, Mayer RJ, Wilkinson KD (1990) Ubiquitin carboxyl-terminal hydrolase (PGP 9.5) is selectively present in ubiquitinated inclusion bodies characteristic of human neurodegenerative diseases. The Journal of pathology 161:153-160.

Lucking CB, Durr A, Bonifati V, Vaughan J, De Michele G, Gasser T, Harhangi BS, Meco G, Denefle P, Wood NW, Agid Y, Brice A (2000) Association between early-onset Parkinson's disease and mutations in the parkin gene. The New England journal of medicine 342:1560-1567.

Page 215: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

200

Lundblad M, Decressac M, Mattsson B, Bjorklund A (2012) Impaired neurotransmission caused by overexpression of alpha-synuclein in nigral dopamine neurons. Proc Natl Acad Sci U S A 109:3213-3219.

Lutz AK, Exner N, Fett ME, Schlehe JS, Kloos K, Lammermann K, Brunner B, Kurz-Drexler A, Vogel F, Reichert AS, Bouman L, Vogt-Weisenhorn D, Wurst W, Tatzelt J, Haass C, Winklhofer KF (2009) Loss of parkin or PINK1 function increases Drp1-dependent mitochondrial fragmentation. J Biol Chem 284:22938-22951.

Lyng GD, Snyder-Keller A, Seegal RF (2007) Dopaminergic development of prenatal ventral mesencephalon and striatum in organotypic co-cultures. Brain research 1133:1-9.

MacLeod D, Dowman J, Hammond R, Leete T, Inoue K, Abeliovich A (2006) The familial Parkinsonism gene LRRK2 regulates neurite process morphology. Neuron 52:587-593.

Magen I, Chesselet MF (2010) Genetic mouse models of Parkinson's disease The state of the art. Prog Brain Res 184:53-87.

Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S (2002) The protein kinase complement of the human genome. Science 298:1912-1934.

Martinat C, Shendelman S, Jonason A, Leete T, Beal MF, Yang L, Floss T, Abeliovich A (2004) Sensitivity to oxidative stress in DJ-1-deficient dopamine neurons: an ES- derived cell model of primary Parkinsonism. PLoS Biol 2:e327.

Massano J, Bhatia KP (2012) Clinical approach to Parkinson's disease: features, diagnosis, and principles of management. Cold Spring Harbor perspectives in medicine 2:a008870.

Mata IF, Wedemeyer WJ, Farrer MJ, Taylor JP, Gallo KA (2006) LRRK2 in Parkinson's disease: protein domains and functional insights. Trends in neurosciences 29:286-293.

Mata IF, Kachergus JM, Taylor JP, Lincoln S, Aasly J, Lynch T, Hulihan MM, Cobb SA, Wu RM, Lu CS, Lahoz C, Wszolek ZK, Farrer MJ (2005) Lrrk2 pathogenic substitutions in Parkinson's disease. Neurogenetics 6:171-177.

Matsunaga E, Chedotal A (2004) Repulsive guidance molecule/neogenin: a novel ligand-receptor system playing multiple roles in neural development. Development, growth & differentiation 46:481-486.

Matsunaga E, Tauszig-Delamasure S, Monnier PP, Mueller BK, Strittmatter SM, Mehlen P, Chedotal A (2004) RGM and its receptor neogenin regulate neuronal survival. Nature cell biology 6:749-755.

McNaught KS, Perl DP, Brownell AL, Olanow CW (2004) Systemic exposure to proteasome inhibitors causes a progressive model of Parkinson's disease. Ann Neurol 56:149-162.

Meissner W, Hill MP, Tison F, Gross CE, Bezard E (2004) Neuroprotective strategies for Parkinson's disease: conceptual limits of animal models and clinical trials. Trends in pharmacological sciences 25:249-253.

Page 216: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

201

Meng F, Yao D, Shi Y, Kabakoff J, Wu W, Reicher J, Ma Y, Moosmann B, Masliah E, Lipton SA, Gu Z (2011) Oxidation of the cysteine-rich regions of parkin perturbs its E3 ligase activity and contributes to protein aggregation. Molecular neurodegeneration 6:34.

Meredith GE, Sonsalla PK, Chesselet MF (2008) Animal models of Parkinson's disease progression. Acta neuropathologica 115:385-398.

Michiorri S, Gelmetti V, Giarda E, Lombardi F, Romano F, Marongiu R, Nerini-Molteni S, Sale P, Vago R, Arena G, Torosantucci L, Cassina L, Russo MA, Dallapiccola B, Valente EM, Casari G (2010) The Parkinson-associated protein PINK1 interacts with Beclin1 and promotes autophagy. Cell death and differentiation 17:962-974.

Miyazaki S, Yanagida T, Nunome K, Ishikawa S, Inden M, Kitamura Y, Nakagawa S, Taira T, Hirota K, Niwa M, Iguchi-Ariga SM, Ariga H (2008) DJ-1-binding compounds prevent oxidative stress-induced cell death and movement defect in Parkinson's disease model rats. J Neurochem 105:2418-2434.

Monnier PP, Sierra A, Macchi P, Deitinghoff L, Andersen JS, Mann M, Flad M, Hornberger MR, Stahl B, Bonhoeffer F, Mueller BK (2002) RGM is a repulsive guidance molecule for retinal axons. Nature 419:392-395.

Morris RL, Hollenbeck PJ (1993) The regulation of bidirectional mitochondrial transport is coordinated with axonal outgrowth. J Cell Sci 104 ( Pt 3):917-927.

Mortiboys H, Johansen KK, Aasly JO, Bandmann O (2010) Mitochondrial impairment in patients with Parkinson disease with the G2019S mutation in LRRK2. Neurology 75:2017-2020.

Murray AM, Weihmueller FB, Marshall JF, Hurtig HI, Gottleib GL, Joyce JN (1995) Damage to dopamine systems differs between Parkinson's disease and Alzheimer's disease with parkinsonism. Ann Neurol 37:300-312.

Mytilineou C, McNaught KS, Shashidharan P, Yabut J, Baptiste RJ, Parnandi A, Olanow CW (2004) Inhibition of proteasome activity sensitizes dopamine neurons to protein alterations and oxidative stress. J Neural Transm 111:1237-1251.

Naoi M, Maruyama W, Shamoto-Nagai M, Yi H, Akao Y, Tanaka M (2005) Oxidative stress in mitochondria: decision to survival and death of neurons in neurodegenerative disorders. Mol Neurobiol 31:81-93.

Nemani VM, Lu W, Berge V, Nakamura K, Onoa B, Lee MK, Chaudhry FA, Nicoll RA, Edwards RH (2010) Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron 65:66-79.

Nicklas WJ, Vyas I, Heikkila RE (1985) Inhibition of NADH-linked oxidation in brain mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite of the neurotoxin, 1-methyl-4-phenyl-1,2,5,6-tetrahydropyridine. Life sciences 36:2503-2508.

Page 217: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

202

Nishikawa K, Li H, Kawamura R, Osaka H, Wang YL, Hara Y, Hirokawa T, Manago Y, Amano T, Noda M, Aoki S, Wada K (2003) Alterations of structure and hydrolase activity of parkinsonism-associated human ubiquitin carboxyl-terminal hydrolase L1 variants. Biochemical and biophysical research communications 304:176-183.

Niu J, Yu M, Wang C, Xu Z (2012) Leucine-rich repeat kinase 2 disturbs mitochondrial dynamics via Dynamin-like protein. Journal of neurochemistry 122:650-658.

Nonaka T, Hasegawa M (2009) A cellular model to monitor proteasome dysfunction by alpha-synuclein. Biochemistry 48:8014-8022.

Obeso JA, Rodriguez-Oroz MC, Rodriguez M, Lanciego JL, Artieda J, Gonzalo N, Olanow CW (2000) Pathophysiology of the basal ganglia in Parkinson's disease. Trends in neurosciences 23:S8-19.

Olanow CW (2008) Levodopa/dopamine replacement strategies in Parkinson's disease--future directions. Movement disorders : official journal of the Movement Disorder Society 23 Suppl 3:S613-622.

Olanow CW et al. (2004) Levodopa in the treatment of Parkinson's disease: current controversies. Movement disorders : official journal of the Movement Disorder Society 19:997-1005.

Olichon A, Baricault L, Gas N, Guillou E, Valette A, Belenguer P, Lenaers G (2003) Loss of OPA1 perturbates the mitochondrial inner membrane structure and integrity, leading to cytochrome c release and apoptosis. J Biol Chem 278:7743-7746.

Olichon A, Landes T, Arnaune-Pelloquin L, Emorine LJ, Mils V, Guichet A, Delettre C, Hamel C, Amati-Bonneau P, Bonneau D, Reynier P, Lenaers G, Belenguer P (2007) Effects of OPA1 mutations on mitochondrial morphology and apoptosis: relevance to ADOA pathogenesis. Journal of cellular physiology 211:423-430.

Oliveira SA et al. (2003) Parkin mutations and susceptibility alleles in late-onset Parkinson's disease. Ann Neurol 53:624-629.

Olzmann JA, Chin LS (2008) Parkin-mediated K63-linked polyubiquitination: a signal for targeting misfolded proteins to the aggresome-autophagy pathway. Autophagy 4:85-87.

Onyango P, Celic I, McCaffery JM, Boeke JD, Feinberg AP (2002) SIRT3, a human SIR2 homologue, is an NAD-dependent deacetylase localized to mitochondria. Proc Natl Acad Sci U S A 99:13653-13658.

Osaka H, Wang YL, Takada K, Takizawa S, Setsuie R, Li H, Sato Y, Nishikawa K, Sun YJ, Sakurai M, Harada T, Hara Y, Kimura I, Chiba S, Namikawa K, Kiyama H, Noda M, Aoki S, Wada K (2003) Ubiquitin carboxy-terminal hydrolase L1 binds to and stabilizes monoubiquitin in neuron. Human molecular genetics 12:1945-1958.

Otera H, Mihara K (2011) Molecular mechanisms and physiologic functions of mitochondrial dynamics. Journal of biochemistry 149:241-251.

Page 218: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

203

Ozelius LJ, Senthil G, Saunders-Pullman R, Ohmann E, Deligtisch A, Tagliati M, Hunt AL, Klein C, Henick B, Hailpern SM, Lipton RB, Soto-Valencia J, Risch N, Bressman SB (2006) LRRK2 G2019S as a cause of Parkinson's disease in Ashkenazi Jews. The New England journal of medicine 354:424-425.

Parisiadou L, Xie C, Cho HJ, Lin X, Gu XL, Long CX, Lobbestael E, Baekelandt V, Taymans JM, Sun L, Cai H (2009) Phosphorylation of ezrin/radixin/moesin proteins by LRRK2 promotes the rearrangement of actin cytoskeleton in neuronal morphogenesis. J Neurosci 29:13971-13980.

Park JS, Mehta P, Cooper AA, Veivers D, Heimbach A, Stiller B, Kubisch C, Fung VS, Krainc D, Mackay-Sim A, Sue CM (2011) Pathogenic effects of novel mutations in the P-type ATPase ATP13A2 (PARK9) causing Kufor-Rakeb syndrome, a form of early-onset parkinsonism. Human mutation 32:956-964.

Parker WD, Jr., Boyson SJ, Parks JK (1989) Abnormalities of the electron transport chain in idiopathic Parkinson's disease. Ann Neurol 26:719-723.

Parkinson J (2002) An essay on the shaking palsy. 1817. The Journal of neuropsychiatry and clinical neurosciences 14:223-236; discussion 222.

Perier C, Bove J, Wu DC, Dehay B, Choi DK, Jackson-Lewis V, Rathke-Hartlieb S, Bouillet P, Strasser A, Schulz JB, Przedborski S, Vila M (2007) Two molecular pathways initiate mitochondria-dependent dopaminergic neurodegeneration in experimental Parkinson's disease. Proc Natl Acad Sci U S A 104:8161-8166.

Piccoli G, Condliffe SB, Bauer M, Giesert F, Boldt K, De Astis S, Meixner A, Sarioglu H, Vogt-Weisenhorn DM, Wurst W, Gloeckner CJ, Matteoli M, Sala C, Ueffing M (2011) LRRK2 controls synaptic vesicle storage and mobilization within the recycling pool. J Neurosci 31:2225-2237.

Plowey ED, Cherra SJ, 3rd, Liu YJ, Chu CT (2008) Role of autophagy in G2019S-LRRK2-associated neurite shortening in differentiated SH-SY5Y cells. J Neurochem 105:1048-1056.

Plun-Favreau H, Klupsch K, Moisoi N, Gandhi S, Kjaer S, Frith D, Harvey K, Deas E, Harvey RJ, McDonald N, Wood NW, Martins LM, Downward J (2007) The mitochondrial protease HtrA2 is regulated by Parkinson's disease-associated kinase PINK1. Nature cell biology 9:1243-1252.

Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, Pike B, Root H, Rubenstein J, Boyer R, Stenroos ES, Chandrasekharappa S, Athanassiadou A, Papapetropoulos T, Johnson WG, Lazzarini AM, Duvoisin RC, Di Iorio G, Golbe LI, Nussbaum RL (1997) Mutation in the alpha-synuclein gene identified in families with Parkinson's disease. Science 276:2045-2047.

Pothakos K, Kurz MJ, Lau YS (2009) Restorative effect of endurance exercise on behavioral deficits in the chronic mouse model of Parkinson's disease with severe neurodegeneration. BMC neuroscience 10:6.

Page 219: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

204

Poulopoulos M, Levy OA, Alcalay RN (2012) The neuropathology of genetic Parkinson's disease. Movement disorders : official journal of the Movement Disorder Society 27:831-842.

Qian JJ, Cheng YB, Yang YP, Mao CJ, Qin ZH, Li K, Liu CF (2008) Differential effects of overexpression of wild-type and mutant human alpha-synuclein on MPP+-induced neurotoxicity in PC12 cells. Neurosci Lett 435:142-146.

Radke S, Chander H, Schafer P, Meiss G, Kruger R, Schulz JB, Germain D (2008) Mitochondrial protein quality control by the proteasome involves ubiquitination and the protease Omi. J Biol Chem 283:12681-12685.

Rajagopalan S, Deitinghoff L, Davis D, Conrad S, Skutella T, Chedotal A, Mueller BK, Strittmatter SM (2004) Neogenin mediates the action of repulsive guidance molecule. Nat Cell Biol 6:756-762.

Ramirez A, Heimbach A, Grundemann J, Stiller B, Hampshire D, Cid LP, Goebel I, Mubaidin AF, Wriekat AL, Roeper J, Al-Din A, Hillmer AM, Karsak M, Liss B, Woods CG, Behrens MI, Kubisch C (2006) Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nat Genet 38:1184-1191.

Ramsay RR, Krueger MJ, Youngster SK, Gluck MR, Casida JE, Singer TP (1991) Interaction of 1-methyl-4-phenylpyridinium ion (MPP+) and its analogs with the rotenone/piericidin binding site of NADH dehydrogenase. J Neurochem 56:1184-1190.

Rapaport D, Brunner M, Neupert W, Westermann B (1998) Fzo1p is a mitochondrial outer membrane protein essential for the biogenesis of functional mitochondria in Saccharomyces cerevisiae. J Biol Chem 273:20150-20155.

Ravina BM, Fagan SC, Hart RG, Hovinga CA, Murphy DD, Dawson TM, Marler JR (2003) Neuroprotective agents for clinical trials in Parkinson's disease: a systematic assessment. Neurology 60:1234-1240.

Rayport S, Sulzer D, Shi WX, Sawasdikosol S, Monaco J, Batson D, Rajendran G (1992) Identified postnatal mesolimbic dopamine neurons in culture: morphology and electrophysiology. J Neurosci 12:4264-4280.

Richardson JR, Quan Y, Sherer TB, Greenamyre JT, Miller GW (2005) Paraquat neurotoxicity is distinct from that of MPTP and rotenone. Toxicological sciences : an official journal of the Society of Toxicology 88:193-201.

Rideout HJ, Larsen KE, Sulzer D, Stefanis L (2001) Proteasomal inhibition leads to formation of ubiquitin/alpha-synuclein-immunoreactive inclusions in PC12 cells. J Neurochem 78:899-908.

Rochet JC, Outeiro TF, Conway KA, Ding TT, Volles MJ, Lashuel HA, Bieganski RM, Lindquist SL, Lansbury PT (2004) Interactions among alpha-synuclein, dopamine, and

Page 220: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

205

biomembranes: some clues for understanding neurodegeneration in Parkinson's disease. Journal of molecular neuroscience : MN 23:23-34.

Rockenstein E, Crews L, Masliah E (2007) Transgenic animal models of neurodegenerative diseases and their application to treatment development. Advanced drug delivery reviews 59:1093-1102.

Rockenstein E, Hansen LA, Mallory M, Trojanowski JQ, Galasko D, Masliah E (2001) Altered expression of the synuclein family mRNA in Lewy body and Alzheimer's disease. Brain research 914:48-56.

Ross OA, Toft M, Whittle AJ, Johnson JL, Papapetropoulos S, Mash DC, Litvan I, Gordon MF, Wszolek ZK, Farrer MJ, Dickson DW (2006) Lrrk2 and Lewy body disease. Ann Neurol 59:388-393.

Rubinsztein DC (2007) Autophagy induction rescues toxicity mediated by proteasome inhibition. Neuron 54:854-856.

Saigoh K, Wang YL, Suh JG, Yamanishi T, Sakai Y, Kiyosawa H, Harada T, Ichihara N, Wakana S, Kikuchi T, Wada K (1999) Intragenic deletion in the gene encoding ubiquitin carboxy-terminal hydrolase in gad mice. Nat Genet 23:47-51.

Saito Y, Ruberu NN, Sawabe M, Arai T, Kazama H, Hosoi T, Yamanouchi H, Murayama S (2004) Lewy body-related alpha-synucleinopathy in aging. Journal of neuropathology and experimental neurology 63:742-749.

Sakata E, Yamaguchi Y, Kurimoto E, Kikuchi J, Yokoyama S, Yamada S, Kawahara H, Yokosawa H, Hattori N, Mizuno Y, Tanaka K, Kato K (2003) Parkin binds the Rpn10 subunit of 26S proteasomes through its ubiquitin-like domain. EMBO Rep 4:301-306.

Samaranch L, Lorenzo-Betancor O, Arbelo JM, Ferrer I, Lorenzo E, Irigoyen J, Pastor MA, Marrero C, Isla C, Herrera-Henriquez J, Pastor P (2010) PINK1-linked parkinsonism is associated with Lewy body pathology. Brain : a journal of neurology 133:1128-1142.

Sapkota K, Kim S, Park SE, Kim SJ Detoxified extract of Rhus verniciflua stokes inhibits rotenone-induced apoptosis in human dopaminergic cells, SH-SY5Y. Cell Mol Neurobiol 31:213-223.

Saunders-Pullman R, Gordon-Elliott J, Parides M, Fahn S, Saunders HR, Bressman S (1999) The effect of estrogen replacement on early Parkinson's disease. Neurology 52:1417-1421.

Savitt JM, Dawson VL, Dawson TM (2006) Diagnosis and treatment of Parkinson disease: molecules to medicine. The Journal of clinical investigation 116:1744-1754.

Scaduto RC, Jr., Grotyohann LW (1999) Measurement of mitochondrial membrane potential using fluorescent rhodamine derivatives. Biophysical journal 76:469-477.

Schapira AH (2009) Neurobiology and treatment of Parkinson's disease. Trends in pharmacological sciences 30:41-47.

Page 221: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

206

Schon EA, Przedborski S (2011) Mitochondria: the next (neurode)generation. Neuron 70:1033-1053.

Schwab JM, Monnier PP, Schluesener HJ, Conrad S, Beschorner R, Chen L, Meyermann R, Mueller BK (2005) Central nervous system injury-induced repulsive guidance molecule expression in the adult human brain. Archives of neurology 62:1561-1568.

Schwarting RK, Huston JP (1996) The unilateral 6-hydroxydopamine lesion model in behavioral brain research. Analysis of functional deficits, recovery and treatments. Progress in neurobiology 50:275-331.

Sha D, Chin LS, Li L (2010) Phosphorylation of parkin by Parkinson disease-linked kinase PINK1 activates parkin E3 ligase function and NF-kappaB signaling. Human molecular genetics 19:352-363.

Shamoto-Nagai M, Maruyama W, Kato Y, Isobe K, Tanaka M, Naoi M, Osawa T (2003) An inhibitor of mitochondrial complex I, rotenone, inactivates proteasome by oxidative modification and induces aggregation of oxidized proteins in SH-SY5Y cells. J Neurosci Res 74:589-597.

Shendelman S, Jonason A, Martinat C, Leete T, Abeliovich A (2004) DJ-1 is a redox-dependent molecular chaperone that inhibits alpha-synuclein aggregate formation. PLoS Biol 2:e362.

Sherer TB, Kim JH, Betarbet R, Greenamyre JT (2003a) Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp Neurol 179:9-16.

Sherer TB, Betarbet R, Stout AK, Lund S, Baptista M, Panov AV, Cookson MR, Greenamyre JT (2002) An in vitro model of Parkinson's disease: linking mitochondrial impairment to altered alpha-synuclein metabolism and oxidative damage. J Neurosci 22:7006-7015.

Sherer TB, Richardson JR, Testa CM, Seo BB, Panov AV, Yagi T, Matsuno-Yagi A, Miller GW, Greenamyre JT (2007) Mechanism of toxicity of pesticides acting at complex I: relevance to environmental etiologies of Parkinson's disease. J Neurochem 100:1469-1479.

Sherer TB, Betarbet R, Testa CM, Seo BB, Richardson JR, Kim JH, Miller GW, Yagi T, Matsuno-Yagi A, Greenamyre JT (2003b) Mechanism of toxicity in rotenone models of Parkinson's disease. J Neurosci 23:10756-10764.

Shimazu T, Hirschey MD, Hua L, Dittenhafer-Reed KE, Schwer B, Lombard DB, Li Y, Bunkenborg J, Alt FW, Denu JM, Jacobson MP, Verdin E (2010) SIRT3 deacetylates mitochondrial 3-hydroxy-3-methylglutaryl CoA synthase 2 and regulates ketone body production. Cell metabolism 12:654-661.

Shimura H, Hattori N, Kubo S, Yoshikawa M, Kitada T, Matsumine H, Asakawa S, Minoshima S, Yamamura Y, Shimizu N, Mizuno Y (1999) Immunohistochemical and subcellular localization of Parkin protein: absence of protein in autosomal recessive juvenile parkinsonism patients. Ann Neurol 45:668-672.

Page 222: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

207

Singleton A, Gwinn-Hardy K (2004) Parkinson's disease and dementia with Lewy bodies: a difference in dose? Lancet 364:1105-1107.

Smirnova E, Griparic L, Shurland DL, van der Bliek AM (2001) Dynamin-related protein Drp1 is required for mitochondrial division in mammalian cells. Molecular biology of the cell 12:2245-2256.

Smith DM, Kafri G, Cheng Y, Ng D, Walz T, Goldberg AL (2005a) ATP binding to PAN or the 26S ATPases causes association with the 20S proteasome, gate opening, and translocation of unfolded proteins. Molecular cell 20:687-698.

Smith WW, Pei Z, Jiang H, Dawson VL, Dawson TM, Ross CA (2006) Kinase activity of mutant LRRK2 mediates neuronal toxicity. Nature neuroscience 9:1231-1233.

Smith WW, Margolis RL, Li X, Troncoso JC, Lee MK, Dawson VL, Dawson TM, Iwatsubo T, Ross CA (2005b) Alpha-synuclein phosphorylation enhances eosinophilic cytoplasmic inclusion formation in SH-SY5Y cells. J Neurosci 25:5544-5552.

Smith WW, Pei Z, Jiang H, Moore DJ, Liang Y, West AB, Dawson VL, Dawson TM, Ross CA (2005c) Leucine-rich repeat kinase 2 (LRRK2) interacts with parkin, and mutant LRRK2 induces neuronal degeneration. Proc Natl Acad Sci U S A 102:18676-18681.

Snyder-Keller A, Costantini LC, Graber DJ (2001) Development of striatal patch/matrix organization in organotypic co-cultures of perinatal striatum, cortex and substantia nigra. Neuroscience 103:97-109.

Snyder H, Mensah K, Theisler C, Lee J, Matouschek A, Wolozin B (2003) Aggregated and monomeric alpha-synuclein bind to the S6' proteasomal protein and inhibit proteasomal function. J Biol Chem 278:11753-11759.

Solano SM, Miller DW, Augood SJ, Young AB, Penney JB, Jr. (2000) Expression of alpha-synuclein, parkin, and ubiquitin carboxy-terminal hydrolase L1 mRNA in human brain: genes associated with familial Parkinson's disease. Ann Neurol 47:201-210.

Souza JM, Giasson BI, Chen Q, Lee VM, Ischiropoulos H (2000) Dityrosine cross-linking promotes formation of stable alpha -synuclein polymers. Implication of nitrative and oxidative stress in the pathogenesis of neurodegenerative synucleinopathies. J Biol Chem 275:18344-18349.

Stefanis L, Larsen KE, Rideout HJ, Sulzer D, Greene LA (2001) Expression of A53T mutant but not wild-type alpha-synuclein in PC12 cells induces alterations of the ubiquitin-dependent degradation system, loss of dopamine release, and autophagic cell death. J Neurosci 21:9549-9560.

Stocchi F, Tagliati M, Olanow CW (2008) Treatment of levodopa-induced motor complications. Movement disorders : official journal of the Movement Disorder Society 23 Suppl 3:S599-612.

Page 223: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

208

Stojanovski D, Koutsopoulos OS, Okamoto K, Ryan MT (2004) Levels of human Fis1 at the mitochondrial outer membrane regulate mitochondrial morphology. Journal of cell science 117:1201-1210.

Stone DK, Kiyota T, Mosley RL, Gendelman HE (2012) A model of nitric oxide induced alpha-synuclein misfolding in Parkinson's disease. Neuroscience letters 523:167-173.

Sullivan PG, Dragicevic NB, Deng JH, Bai Y, Dimayuga E, Ding Q, Chen Q, Bruce-Keller AJ, Keller JN (2004) Proteasome inhibition alters neural mitochondrial homeostasis and mitochondria turnover. J Biol Chem 279:20699-20707.

Taira T, Saito Y, Niki T, Iguchi-Ariga SM, Takahashi K, Ariga H (2004) DJ-1 has a role in antioxidative stress to prevent cell death. EMBO reports 5:213-218.

Tanaka Y, Engelender S, Igarashi S, Rao RK, Wanner T, Tanzi RE, Sawa A, V LD, Dawson TM, Ross CA (2001) Inducible expression of mutant alpha-synuclein decreases proteasome activity and increases sensitivity to mitochondria-dependent apoptosis. Hum Mol Genet 10:919-926.

Tassew NG, Chestopolava L, Beecroft R, Matsunaga E, Teng H, Chedotal A, Monnier PP (2008) Intraretinal RGMa is involved in retino-tectal mapping. Molecular and cellular neurosciences 37:761-769.

Testa CM, Sherer TB, Greenamyre JT (2005) Rotenone induces oxidative stress and dopaminergic neuron damage in organotypic substantia nigra cultures. Brain research Molecular brain research 134:109-118.

Todde V, Veenhuis M, van der Klei IJ (2009) Autophagy: principles and significance in health and disease. Biochim Biophys Acta 1792:3-13.

Tolosa E, Wenning G, Poewe W (2006) The diagnosis of Parkinson's disease. Lancet neurology 5:75-86.

Tompkins MM, Hill WD (1997) Contribution of somal Lewy bodies to neuronal death. Brain research 775:24-29.

Trushina E, Nemutlu E, Zhang S, Christensen T, Camp J, Mesa J, Siddiqui A, Tamura Y, Sesaki H, Wengenack TM, Dzeja PP, Poduslo JF (2012) Defects in mitochondrial dynamics and metabolomic signatures of evolving energetic stress in mouse models of familial Alzheimer's disease. PloS one 7:e32737.

Tuite P, Riss J (2003) Recent developments in the pharmacological treatment of Parkinson's disease. Expert opinion on investigational drugs 12:1335-1352.

Twig G, Elorza A, Molina AJ, Mohamed H, Wikstrom JD, Walzer G, Stiles L, Haigh SE, Katz S, Las G, Alroy J, Wu M, Py BF, Yuan J, Deeney JT, Corkey BE, Shirihai OS (2008) Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. The EMBO journal 27:433-446.

Page 224: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

209

Ullrich C, Humpel C (2009) Rotenone induces cell death of cholinergic neurons in an organotypic co-culture brain slice model. Neurochemical research 34:2147-2153.

Ungerstedt U (1968) 6-Hydroxy-dopamine induced degeneration of central monoamine neurons. European journal of pharmacology 5:107-110.

Ungerstedt U (1971) Adipsia and aphagia after 6-hydroxydopamine induced degeneration of the nigro-striatal dopamine system. Acta physiologica Scandinavica Supplementum 367:95-122.

Usenovic M, Tresse E, Mazzulli JR, Taylor JP, Krainc D (2012) Deficiency of ATP13A2 leads to lysosomal dysfunction, alpha-synuclein accumulation, and neurotoxicity. J Neurosci 32:4240-4246.

van der Putten H, Wiederhold KH, Probst A, Barbieri S, Mistl C, Danner S, Kauffmann S, Hofele K, Spooren WP, Ruegg MA, Lin S, Caroni P, Sommer B, Tolnay M, Bilbe G (2000) Neuropathology in mice expressing human alpha-synuclein. J Neurosci 20:6021-6029.

Verdin E, Hirschey MD, Finley LW, Haigis MC (2010) Sirtuin regulation of mitochondria: energy production, apoptosis, and signaling. Trends in biochemical sciences 35:669-675.

Verhoeven K et al. (2006) MFN2 mutation distribution and genotype/phenotype correlation in Charcot-Marie-Tooth type 2. Brain : a journal of neurology 129:2093-2102.

Vila M, Ramonet D, Perier C (2008) Mitochondrial alterations in Parkinson's disease: new clues. Journal of neurochemistry 107:317-328.

Vogiatzi T, Xilouri M, Vekrellis K, Stefanis L (2008) Wild type alpha-synuclein is degraded by chaperone-mediated autophagy and macroautophagy in neuronal cells. J Biol Chem 283:23542-23556.

Volpicelli-Daley LA, Luk KC, Patel TP, Tanik SA, Riddle DM, Stieber A, Meaney DF, Trojanowski JQ, Lee VM (2011) Exogenous alpha-synuclein fibrils induce Lewy body pathology leading to synaptic dysfunction and neuron death. Neuron 72:57-71.

von Bohlen und Halbach O, Schober A, Krieglstein K (2004) Genes, proteins, and neurotoxins involved in Parkinson's disease. Progress in neurobiology 73:151-177.

Wakita S, Izumi Y, Matsuo T, Kume T, Takada-Takatori Y, Sawada H, Akaike A (2010) Reconstruction and quantitative evaluation of dopaminergic innervation of striatal neurons in dissociated primary cultures. Journal of neuroscience methods 192:83-89.

Walls KC, Ghosh AP, Franklin AV, Klocke BJ, Ballestas M, Shacka JJ, Zhang J, Roth KA (2010) Lysosome dysfunction triggers Atg7-dependent neural apoptosis. J Biol Chem 285:10497-10507.

Page 225: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

210

Wang X, Petrie TG, Liu Y, Liu J, Fujioka H, Zhu X (2012a) Parkinson's disease-associated DJ-1 mutations impair mitochondrial dynamics and cause mitochondrial dysfunction. Journal of neurochemistry 121:830-839.

Wang X, Su B, Liu W, He X, Gao Y, Castellani RJ, Perry G, Smith MA, Zhu X (2011) DLP1-dependent mitochondrial fragmentation mediates 1-methyl-4-phenylpyridinium toxicity in neurons: implications for Parkinson's disease. Aging cell 10:807-823.

Wang X, Yan MH, Fujioka H, Liu J, Wilson-Delfosse A, Chen SG, Perry G, Casadesus G, Zhu X (2012b) LRRK2 regulates mitochondrial dynamics and function through direct interaction with DLP1. Hum Mol Genet 21:1931-1944.

West AB, Moore DJ, Biskup S, Bugayenko A, Smith WW, Ross CA, Dawson VL, Dawson TM (2005) Parkinson's disease-associated mutations in leucine-rich repeat kinase 2 augment kinase activity. Proc Natl Acad Sci U S A 102:16842-16847.

West AB, Moore DJ, Choi C, Andrabi SA, Li X, Dikeman D, Biskup S, Zhang Z, Lim KL, Dawson VL, Dawson TM (2007) Parkinson's disease-associated mutations in LRRK2 link enhanced GTP-binding and kinase activities to neuronal toxicity. Human molecular genetics 16:223-232.

Westermann B (2010) Mitochondrial fusion and fission in cell life and death. Nature reviews Molecular cell biology 11:872-884.

Whitworth AJ, Pallanck LJ (2009) The PINK1/Parkin pathway: a mitochondrial quality control system? J Bioenerg Biomembr 41:499-503.

Wickremaratchi MM, Ben-Shlomo Y, Morris HR (2009) The effect of onset age on the clinical features of Parkinson's disease. European journal of neurology : the official journal of the European Federation of Neurological Societies 16:450-456.

Wu F, Poon WS, Lu G, Wang A, Meng H, Feng L, Li Z, Liu S (2009) Alpha-synuclein knockdown attenuates MPP+ induced mitochondrial dysfunction of SH-SY5Y cells. Brain Res 1292:173-179.

Xie H, Chang M, Hu X, Wang D, Tian M, Li G, Jiang H, Wang Y, Dong Z, Zhang Y, Hu L (2011) Proteomics analysis of MPP(+)-induced apoptosis in SH-SY5Y cells. Neurol Sci 32:221-228.

Xie HR, Hu LS, Li GY (2010) SH-SY5Y human neuroblastoma cell line: in vitro cell model of dopaminergic neurons in Parkinson's disease. Chin Med J (Engl) 123:1086-1092.

Xie W, Chung KK (2012) Alpha-synuclein impairs normal dynamics of mitochondria in cell and animal models of Parkinson's disease. Journal of neurochemistry.

Yang L, Calingasan NY, Wille EJ, Cormier K, Smith K, Ferrante RJ, Beal MF (2009) Combination therapy with coenzyme Q10 and creatine produces additive neuroprotective effects in models of Parkinson's and Huntington's diseases. Journal of neurochemistry 109:1427-1439.

Page 226: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

211

Yong-Kee CJ, Salomonczyk D, Nash JE (2011a) Development and Validation of a Screening Assay for the Evaluation of Putative Neuroprotective Agents in the Treatment of Parkinson's Disease. Neurotox Res 19:7.

Yong-Kee CJ, Salomonczyk D, Nash JE (2011b) Development and validation of a screening assay for the evaluation of putative neuroprotective agents in the treatment of Parkinson's disease. Neurotoxicity research 19:519-526.

Yong-Kee CJ, Sidorova E, Hanif A, Perera G, Nash JE (2012) Mitochondrial dysfunction precedes other sub-cellular abnormalities in an in vitro model linked with cell death in Parkinson's disease. Neurotoxicity research 21:185-194.

Yoon Y, Krueger EW, Oswald BJ, McNiven MA (2003) The mitochondrial protein hFis1 regulates mitochondrial fission in mammalian cells through an interaction with the dynamin-like protein DLP1. Molecular and cellular biology 23:5409-5420.

Yun J, Cao JH, Dodson MW, Clark IE, Kapahi P, Chowdhury RB, Guo M (2008) Loss-of-function analysis suggests that Omi/HtrA2 is not an essential component of the PINK1/PARKIN pathway in vivo. J Neurosci 28:14500-14510.

Zarate-Lagunes M, Gu WJ, Blanchard V, Francois C, Muriel MP, Mouatt-Prigent A, Bonici B, Parent A, Hartmann A, Yelnik J, Boehme GA, Pradier L, Moussaoui S, Faucheux B, Raisman-Vozari R, Agid Y, Brice A, Hirsch EC (2001) Parkin immunoreactivity in the brain of human and non-human primates: an immunohistochemical analysis in normal conditions and in Parkinsonian syndromes. The Journal of comparative neurology 432:184-196.

Zhang D, Anantharam V, Kanthasamy A, Kanthasamy AG (2007) Neuroprotective effect of protein kinase C delta inhibitor rottlerin in cell culture and animal models of Parkinson's disease. The Journal of pharmacology and experimental therapeutics 322:913-922.

Zhang J, Fitsanakis VA, Gu G, Jing D, Ao M, Amarnath V, Montine TJ (2003) Manganese ethylene-bis-dithiocarbamate and selective dopaminergic neurodegeneration in rat: a link through mitochondrial dysfunction. J Neurochem 84:336-346.

Zhou C, Huang Y, Shao Y, May J, Prou D, Perier C, Dauer W, Schon EA, Przedborski S (2008a) The kinase domain of mitochondrial PINK1 faces the cytoplasm. Proc Natl Acad Sci U S A 105:12022-12027.

Zhou W, Milder JB, Freed CR (2008b) Transgenic mice overexpressing tyrosine-to-cysteine mutant human alpha-synuclein: a progressive neurodegenerative model of diffuse Lewy body disease. J Biol Chem 283:9863-9870.

Zimprich A et al. (2004) Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 44:601-607.

Zuchner S et al. (2004) Mutations in the mitochondrial GTPase mitofusin 2 cause Charcot-Marie-Tooth neuropathy type 2A. Nat Genet 36:449-451.

Page 227: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

212

8 Appendices

A modified version of this appendix was previously published as:

Yong-Kee CJ, Warre R, Monnier PP, Lozano AM, Nash JE (2012) Evidence for synergism between cell death mechanisms in a cellular model of neurodegeneration in

Parkinson's disease. Neurotoxicity research 22(4):355-364.

My contributions to the following appendix include data analyses and design of the

figures listed in the text.

Abstract

Delineation of how cell death mechanisms associated with PD interact and whether they

converge would help identify targets for neuroprotective therapies. The purpose of this study

was to use a cellular model to address these issues. Catecholaminergic SH-SY5Y neuroblastoma

cells were exposed to a range of compounds (dopamine, rotenone, naphthazarin, and PSI) that

are neurotoxic when applied to these cells for extended periods of times at specific

concentrations. At the concentrations used, these compounds cause cellular stress via

mechanisms that mimic those associated with causing neurodegeneration in PD, namely

oxidative stress (dopamine), mitochondrial dysfunction (rotenone), lysosomal dysfunction

(naphthazarine), and proteasomal dysfunction (PSI). The compounds were applied to the SH-

SY5Y cells either alone or in pairs. When applied separately, the compounds produced a

significant decrease in cell viability confirming that oxidative stress, mitochondrial, proteosomal,

or lysosomal dysfunction can individually result in catecholaminergic cell death. When the

compounds were applied in pairs, some of the combinations produced synergistic effects.

Analysis of these interactions indicates that proteasomal, lysosomal and mitochondrial

Page 228: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

213

dysfunction is exacerbated by dopamine-induced oxidative stress. Furthermore, inhibition of the

proteasome or lysosome or increasing oxidative stress had a synergistic effect on cell viability

when combined with mitochondrial dysfunction, suggesting that all cell death mechanisms

impair mitochondrial function. Finally, we show that there are reciprocal relationships between

oxidative stress, proteasomal dysfunction, and mitochondrial dysfunction, whereas lysosomal

dysfunction appears to mediate cell death via an independent pathway. Given the highly

interactive nature of the various cell death mechanisms linked with PD, we predict that effective

neuroprotective strategies would target multiple sites in these pathways, for example oxidative

stress and mitochondria.

Introduction

The primary pathologies underlying PD are a loss of dopaminergic cells within the SNc as well

as additional catecholaminergic brain stem nuclei (Braak et al. 2003), combined with the

appearance of aggregates of protein, known as Lewy bodies, in affected brain regions (Goldman

et al. 1983). Upon diagnosis, patients are relatively mobile and are able to perform most motor

tasks. As the disease progresses, symptoms become severely disabling such that palliative care

is necessary. If, upon diagnosis of PD, it were possible to prescribe a treatment capable of

halting, slowing or even reversing the progression of degeneration of the SNc, patients would not

reach the advanced stages of the disease, and so could maintain a better quality of life (Olanow

2008).

The development of effective neuroprotective or neurorestorative agents requires an

understanding of the sub-cellular mechanisms underlying neurodegeneration in PD. To this end,

studies in animal and cell models of PD and post-mortem brain tissue from patients with PD

have shown that oxidative stress (Jenner 2003; Testa et al. 2005), malfunction of mitochondrial

Page 229: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

214

complex 1 (Lannuzel et al. 2003; Sherer et al. 2007; von Bohlen Und Halbach et al. 2004), and

dysfunction of the UPS contribute to neurodegeneration in PD. In addition, dysfunction of

trafficking within and between the endoplasmic reticulum and golgi network (Cooper et al.

2006), presynaptic function (Wislet-Gendebien et al. 2006; Wislet-Gendebien et al. 2008), and

chaperone mediated autophagy (CMA) have also been associated with cell death in PD (Ardley

et al. 2004; Caneda-Ferron et al. 2008; Li et al. 2004). These findings are confounded by the fact

that all mutated proteins linked with familial cases of PD cause at least one of the above

mechanisms to function aberrantly (Pan et al. 2008). Recent studies have shown that different

cell death mechanisms associated with PD are closely linked, whereby one cell stressor causes

abnormalities in at least one other cell death mechanism linked with PD (Betarbet et al. 2006;

Greenamyre and Hastings 2004; Hoglinger et al. 2003; Sherer et al. 2007; Testa et al. 2005).

While many studies in animal and cell culture models of PD have revealed the sub-cellular

mechanisms responsible for cell death, and indicated how these mechanisms interact, all of these

models are either difficult to produce or take months or years to develop the pathologies or

phenotypes reminiscent of PD (Nuber et al. 2008; Richfield et al. 2002). Because of this, the

discovery of potential novel targets as neuroprotective or neurorestorative therapies has been

significantly delayed. Previously we have used a heterologous catecholaminergic neuroblastoma

cell line, SH-SY5Y, which has both dopamine β-hydroxylase and dopamine transporter activity,

and thus may be used as a model of dopaminergic neurons, to show that inhibition of either

mitochondria complex 1 using rotenone, inhibition of the proteasome using PSI, inhibition of

lysosomes using naphthazarin, or the induction of oxidative stress using high concentrations of

dopamine causes a decrease in cell viability over a 24 hour time period (Yong-Kee et al. 2011).

Page 230: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

215

The mechanisms underlying this increase in cell stress include early changes in mitochondrial

dysfunction followed by increased ubiquitin levels (Yong-Kee et al. 2012).

In the present study we extend our line of investigation by using combinations of these

compounds to study the interactions between the multiple cell-death mechanisms associated with

PD. The aim of the current study was to identify whether cell death mechanisms thought to be

responsible for neurodegeneration in PD converge. We show that proteasomal, lysosomal, and

mitochondrial dysfunction form independent cell death pathways. Furthermore, proteasomal and

mitochondrial dysfunction are exacerbated by oxidative stress. In addition there are reciprocal

links between oxidative stress, proteasomal dysfunction, and mitochondrial dysfunction, and also

between mitochondrial and lysosomal dysfunction. These findings suggest that the most

effective neuroprotective treatment strategies for PD should target multiple sites, for example

oxidative stress and mitochondria.

Materials and Methods

Materials

Dopamine hydrochloride, 5,8-dihydroxy-1,4-naphthoquinone (naphthazarin), rotenone and

dimethyl sulfoxide (DMSO) were purchased from Sigma (USA), Z-Ile-Glu(OBut)-Ala-Leu-al

(PSI) from BIOMOL (USA) and alamar blueTM from Biosource (Canada). SH-SY5Y cells were

purchased from ATCC, Dulbecco’s Modified Eagle’s Medium (DMEM), bovine calf serum,

trypsin and L-glutamine from Wisent (Canada).

Cell culture

Human catecholaminergic neuroblastoma SH-SY5Y cells (Imashuku et al. 1973) (P10 to P30)

were cultured in DMEM (6400 mg/L NaCl, 3700 mg/L NaHCO3, 400 mg/L KCl, and 584 mg/L

Page 231: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

216

L-glutamine) supplemented with 5% bovine calf serum in a sterile humidified chamber (37 °C,

95% CO2, 5% O2) (MCO-20AIC, Sanyo, USA) until confluent. Cells were passed using 0.1%

trypsin for 5 min, then pelleted by centrifuging at 1400 rpm (Allegra 6R Centrifuge, Beckman

Coulter, USA).

Cell viability assays

Twenty four hours following addition to 96-well plates, SH-SY5Y cells were exposed to

concentration response curves of either, dopamine hydrochloride (0.1 µM–600 µM),

naphthazarin (0.1 µM–1 mM), PSI (0.1 µM–500 µM), or rotenone (0.1 µM–1 mM). Rotenone

and naphthazarin were dissolved in 0.1% DMSO and all other compounds were dissolved in cell

media for the concentration response curves. For the compound pair experiments naphthazarin

was dissolved in cell media instead of DMSO. Where DMSO was used, a vehicle control group

for the effect of DMSO on cell viability was included in the protocol. The redox sensitive dye

alamar blue (0.4% of final volume) was added, and cells were incubated for 24 hours. Cell

viability was assessed by measuring the change in fluorescence of alamar blue (ex. 544 nm, em.

590 nm) using a FLUOstar OPTIMA (BMG Labtech, USA) in comparison with control wells

containing SH-SY5Y cells, alamar blue and either media or 0.1% DMSO depending on the

compounds used. To determine the effect of combinations of compounds, cells were exposed to

a full concentration response curve of one compound in the presence of a concentration of the

second compound that produced 10 to 20% of its maximal effect (EC10–20%; see Results and Fig.

1). Cells were exposed to pairs of compounds for 24 hours and cell viability was measured as

described above. For all experiments n = 6, where each n is the mean of six replicates.

Statistical Analysis

Page 232: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

217

For each replicate (n), fluorescence was expressed as the mean of 6 replicates in each

experiment. To determine EC10%, EC15%, and EC20% values, the concentration response data

were analysed using the least squares method of nonlinear regression to fit curves constrained by

a Hill slope of -1 and a curve bottom of 0. The top of the curve was unconstrained. Differences

in the means ± SEM for 6 experiments comparing the response to one compound with a pair of

compounds were analysed using two-way ANOVA and Bonferroni post hoc. All data were

analyzed using Graphpad Prism 5.0 statistical software (Graphpad Software Inc.).

Results

Determination of EC10–20% values

Concentration response curves for dopamine (0.01 µM–600 µM), rotenone (0.1 µM–1 mM), PSI

(0.1 µM–500 µM), and naphthazarin (0.1 µM–1 mM) were performed (Fig. 8.1). To investigate

potential synergism between cell death pathways, for each compound, an appropriate

concentration that could be applied to SH-SY5Y cells in combination with a range of

concentrations of a second compound was determined. The criteria for choosing the

concentration was that it was an active concentration, i.e., induced toxicity (as measured by a

decrease in cell viability), but not to such an extent that, when added in combination with a

second compound it would mask any possible synergistic effects, which would be the case if the

chosen concentration caused a large decrease in cell viability.

Following exposure of SH-SY5Y cells to a range of dopamine concentrations (0.1 µM – 600

µM), and measurement of cell viability, a curve fit generated an R2 value of 0.90 and indicated

that the EC20% of dopamine was 11.5 µM (logEC20% = 1.06 ± 0.06, 95% CI [0.94, 1.17]), thus a

concentration of 10 µM dopamine was used in combination with concentration response curves

Page 233: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

218

of the other three compounds. The curves fitted to the rotenone and PSI concentration response

curves generated R2 values of 0.64 and 0.71, respectively, indicating a poorer fit than observed

with dopamine, and the need to err on the side of caution with any predicted EC values. Indeed,

visual inspection of the curves indicated that in contrast to dopamine, decreases in cell viability

induced by rotenone and PSI followed an ‘all or none’ pattern, in which there was either no

significant loss of cell viability or almost maximal loss of cell viability, rather than a

concentration-dependent effect. Thus, in the presence of rotenone or PSI, cell viability dropped

from approximately 80 to 90% when cells were exposed to 100 µM of either compound, to 30 to

45% when cells were exposed to 1000 µM. Therefore, in order to avoid causing a large decrease

in cell viability when pairing rotenone or PSI with a second compound, which would mask

synergistic effects, an EC10% concentration of rotenone and PSI was chosen, rather than an EC20%

as with dopamine. Analysis of the concentration response curves of rotenone and PSI indicated

that the EC10% values were 103.3 µM (logEC10% = 2.01 ± 0.09, 95% CI [1.83, 2.20]) and 42.4

µM (logEC10% = 1.63 ± 0.11, 95% CI [1.41, 1.84]) respectively, thus 100 µM rotenone and 50

µM PSI was used when cells were exposed to combinations of compounds. The concentration

response curve for naphthazarin had an R2 value of 0.95 and the EC20% concentration was

determined to be 4.5 µM (logEC20% = 0.65 ± 0.05, 95% CI [0.54, 0.76]). However, in order to

avoid the effects of adding extra DMSO to the cell cultures naphthazarin was dissolved in cell

media for the compound pair experiments. Problems with the solubility of naphthazarin in cell

media led us to use 3 µM naphthazarin in the paired compound experiments, which equates to an

approximate EC value of 15% (logEC15% = 0.50 ± 0.05, 95% CI [0.39, 0.60]).

Page 234: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

219

Figure 8.1. Determination of EC10–20% concentrations of dopamine, rotenone, PSI, and

naphthazarin on cell viabililty in SH-SY5Y cells. SH-SY5Y cells were incubated with

dopamine (0.01 µM–600 µM), rotenone (0.1 µM–1 mM), PSI (0.1 µM–500 µM), or naphthazarin

(0.1 µM–1 mM) for 24 hours. Cell viability (% control) was assayed using alamar blue and the

resulting concentration response data were plotted (mean ± SEM). Curves were fitted to the data

using the using the least squares method constrained by a Hill slope of -1 and a curve bottom of

0. The top of the curve was unconstrained. The fitted curves were then used to determine the

EC values and concentrations indicated for each compound.

Page 235: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

220

-2 -1 0 1 2 30

20

40

60

80

100

120

Rotenone, EC10 = 103.3 µM, R2 = 0.64

PSI, EC10 = 42.4 µM, R2 = 0.71Naphthazarin, EC15 = 3.1 µM, R2 = 0.95

Dopamine, EC20 = 11.5 µM, R2 = 0.90

0.01 1001010.1 1000

[Toxin] µM

Cel

l via

bilit

y (%

con

trol

)

Page 236: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

221

Characterisation of interactions between cell death mechanisms

Concentration response curves were performed to assess cell viability in response to a dose

response curve of each compound (dopamine, rotenone, PSI, naphthazarin), either alone or in the

presence of an EC10–20% concentration of a second compound. The decrease in the percentage of

viable cells from 100% in the presence of a compound alone (compound 1) was compared with

the decrease in the percentage of viable cells from 100% in the presence of compound 1 plus an

approximate EC10–20% concentration of compound 2. As described above, the EC10-20%

concentrations used were 10 µM dopamine (EC20%), 100 µM rotenone (EC10%), 50 µM PSI

(EC10%), and 3 µM naphthazarin (EC15%).

As an EC20% of compound 2 should produce an additional decrease in cell viability of 20% if the

relationship is simply additive, synergism between the compounds was identified as an

additional decrease in the percentage of viable cells of greater than 25% in the presence of

compounds 1 and 2 in comparison with compound 1 alone for at least two points within the first

five on the concentration response curve. Similarly, where an EC10% or EC15% of compound 2

was added in combination with compound 1, an additive effect would be expected to decrease

cell viability by 10% and 15%, respectively. In these cases, synergism was defined as an

additional decrease in cell viability of 12.5% and 18.75%, respectively, for at least two points on

the concentration response curve for compounds 1 and 2 when compared with compound 1

alone.

Synergism is indicated in Figures 8.2 to 8.5 by diagonal shading. The occurrence of synergism

suggests that the two cell death mechanisms activated by the compounds are part of the same

pathway. An additive effect was defined when the decrease in the percentage of viable cells in

Page 237: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

222

the presence of compound 1 and 2 was 20%, 15%, 10% or less depending on the identity of

compound 2, indicating that the compounds cause cell death by independent pathways. In some

cases the combination of compound 1 and 2 produced no additional decrease in cell viability

over compound 1 indicating that the effects of compound 1 occlude the effects of compound 2.

Effect of rotenone, PSI, or naphthazarin on dopamine-induced toxicity

SH-SY5Y cells were exposed to a range of dopamine concentrations (0.01 µM–600 µM) either

alone, or in the presence of an EC10% of rotenone, an EC10% of PSI, or an EC15% of naphthazarin.

Incubation with dopamine alone caused a concentration-dependent decrease in cell viability (Fig.

8.2). The combination of rotenone (100 µM) with dopamine caused a significant additional

decrease in the percentage of viable cells of 25.7%, 27.1%, and 26.2% for 0.1, 1, and 10 µM

dopamine (P < 0.01), respectively, indicating that dopamine and rotenone work synergistically

(Fig 8.2a). Similarly, the addition of PSI (50 µM) to the cells in the presence of dopamine also

caused a synergistic decrease in cell viability, which ranged from 19.0% to 32.6%, for all

concentrations of dopamine between 0.1 µM and 300 µM (P < 0.001) (Fig. 8.2b). In contrast, the

addition of naphthazarin (3 µM) to the dopamine dose response curve only caused an additive

effect with a significant additional decrease in the cell viability of 24.6% only observed for 1 µM

dopamine. For the remaining concentrations of dopamine in the concentration-response curve

the addition of naphthazarin caused additional decreases in the percentage of viable cells of

11.5% to 18.5% (Fig. 8.2c).

Page 238: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

223

Figure 8.2. Effect of rotenone, PSI, or naphthazarin on dopamine-induced toxicity. SH-

SY5Y cells were incubated with dopamine (DA, 0.01 µM–600 µM), either alone, or in the

presence of approximate EC10–20% concentrations of (a) rotenone (ROT, 100 µM ≈ EC10%), (b)

PSI (50 µM ≈ EC10%), or (c) naphthazarin (NAP, 3 µM ≈ EC15%) for 24 hours. Cell viability (%

control) was assayed using alamar blue and concentration-response curves were plotted (mean ±

SEM). Diagonal shading indicates a difference of greater than 25% between the two curves for

dopamine and rotenone (a) and a difference of greater than 12.5% between the two curves for

dopamine and PSI (b), which indicates the presence of synergy between the pairs of compounds.

The effect of naphthazarin in combination with dopamine was additive (c). Differences between

the curves were analyzed using two-way ANOVA with Bonferroni’s multiple comparison test

post-hoc. * indicates P < 0.05, ** P < 0.01, and *** P < 0.001. Overall effect of rotenone

treatment: F1,70 = 28.11, P < 0.0001, PSI treatment: F1,70 = 123.9, P < 0.0001, and napthazarin

treatment: F1,70 = 18.20, P < 0.0001.

Page 239: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

224

0.01 0.1 1 10 100 10000

20

40

60

80

100

120 DA aloneDA + ROT 100 µM

****

**

[Dopamine]µM

Cel

l via

bilit

y (%

con

trol

)

0.01 0.1 1 10 100 10000

20

40

60

80

100

120 DA aloneDA + PSI 50 µM

***

***

******

***

[Dopamine]µM

Cel

l via

bilit

y (%

con

trol

)

ba

c

0.01 0.1 1 10 100 10000

20

40

60

80

100

120 DA aloneDA + NAP 3 µM

*

[Dopamine]µM

Cel

l via

bilit

y (%

con

trol

)

Page 240: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

225

Effect of dopamine, PSI, or naphthazarin on rotenone-induced

mitochondrial dysfunction

SH-SY5Y cells were exposed to a range of rotenone concentrations (0.01 µM–1000µM) either

alone, or in the presence of an approximate EC20% of dopamine, an EC10% of PSI, or an EC15% of

naphthazarin. Incubation with rotenone alone caused a concentration-dependent decrease in cell

viability (Fig. 8.3). The addition of dopamine (10 µM) to the rotenone concentration-response

curve caused a significant additional decrease in cell viability at all concentrations of rotenone

between 0.01 µM and 100 µM (P < 0.01). The additional decrease in the percentage of viable

cells ranged from 24.8% to 34.0% for the concentrations of rotenone between 0.01 µM and 100

µM indicating that the effects of dopamine and rotenone were synergistic (Fig. 8.3a). The

addition of PSI (50 µM) (Fig. 8.3b) or naphthazarin (3 µM) (Fig. 8.3c) to the rotenone

concentration-response curve both produced a synergistic effect at 1 µM and 10 µM rotenone

producing a decrease in cell viability of 18.5% and 15.8%, respectively with PSI, and 20.3% and

22.6%, respectively with naphthazarin (P < 0.01) (Fig. 8.3 b,c).

Page 241: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

226

Figure 8.3. Effect of dopamine, PSI, or naphthazarin on rotenone-induced mitochondrial

dysfunction. SH-SY5Y cells were incubated with rotenone (ROT, 0.01 µM–1000 µM), either

alone, or in the presence of approximate EC10–20% concentrations of (a) dopamine (DA, 10 µM ≈

EC20%), (b) PSI (50 µM ≈ EC10%), or (c) naphthazarin (NAP, 3 µM ≈ EC15%) for 24 hours. Cell

viability (% control) was assayed using alamar blue and concentration-response curves were

plotted (mean ± SEM). Diagonal shading indicates a difference of greater than 25% between the

two curves for rotenone and dopamine (a), a difference of greater than 12.5% between the two

curves for rotenone and PSI (b), and a difference of greater than 18.75% between the two curves

for rotenone and naphthazarin (c), which indicates the presence of synergy between the pairs of

compounds. Differences between the curves were analyzed using two-way ANOVA with

Bonferroni’s multiple comparison test post-hoc. * indicates P < 0.05, ** P < 0.01, and *** P <

0.001. Overall effect of dopamine treatment: F1,70 = 84.01, P < 0.0001, PSI treatment: F1,70 =

16.41, P = 0.0001, and naphthazarin treatment: F1,70 = 37.89, P < 0.0001.

Page 242: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

227

ba

c

0.01 0.1 1 10 100 1000

20

40

60

80

100

120ROT aloneROT + DA 10 µM

********* **

***

[Rotenone]µM

Cel

l via

bilit

y (%

con

trol

)

0.01 0.1 1 10 100 1000

20

40

60

80

100

120ROT aloneROT + PSI 50 µM

[Rotenone]µM

Cel

l via

bilit

y (%

con

trol

)

0.01 0.1 1 10 100 10000

20

40

60

80

100

120ROT aloneROT + NAP 3 µM

** **

[Rotenone]µM

Cel

l via

bilit

y (%

con

trol

)

Page 243: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

228

Effect of dopamine, rotenone, or naphthazarin on PSI-induced proteasome

dysfunction

SH-SY5Y cells were exposed to a range of PSI concentrations (0.1 µM–500 µM) in combination

with an approximate EC20% of dopamine, an EC10% of rotenone, or an EC15% of naphthazarin.

Incubation with PSI alone caused a concentration-dependent decrease in cell viability (Fig. 8.4).

Incubation of PSI with dopamine (10 µM) caused a significant additional decrease in cell

viability compared to PSI alone at all concentrations of PSI between 0.1 µM and 500 µM (P <

0.01 for 0.1 µM–10 µM, P < 0.05 for 50 µM and 150 µM, P > 0.05 for 500 µM). The additional

decrease in the percentage of viable cells ranged from 15.5% to 29.8% indicating that PSI and

dopamine act synergistically (Fig. 8.4a). The combination of PSI and rotenone (100 µM) also

produced a synergistic decrease in the percentage of viable cells that ranged from 19.1% to

25.0% for the concentrations of PSI between 0.1 µM and 150 µM (Fig. 8.4b). The addition of

naphthazarin (3 µM) to the PSI concentration-response curve produced additional decreases in

the percentage of viable cells of 16.7% to 19.6% between 0.1 µM and 10 µM PSI indicating that

the effects of napthazarin and PSI were additive, rather than synergistic as the additional

decrease in cell viability was not sustained at 18.75% or greater (Fig. 8.4c).

Page 244: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

229

Figure 8.4. Effect of dopamine, rotenone, or naphthazarin on PSI-induced proteasomal

dysfunction. SH-SY5Y cells were incubated with PSI (PSI, 0.1 µM–500 µM), either alone, or

in the presence of approximate EC10–20% concentrations of (a) dopamine (DA, 10 µM ≈ EC20%),

(b) rotenone (ROT, 100 µM ≈ EC10%), or (c) naphthazarin (NAP, 3 µM ≈ EC15%) for 24 hours.

Cell viability (% control) was assayed using alamar blue and concentration-response curves were

plotted (mean ± SEM). Diagonal shading indicates a difference of greater than 25% between the

two curves for PSI and dopamine (a) and a difference of greater than 12.5% between the two

curves for PSI and rotenone (b), which indicates the presence of synergy between the pairs of

compounds. The effect of naphthazarin in combination with PSI was additive (c). Differences

between the curves were analyzed using two-way ANOVA with Bonferroni’s multiple

comparison test post-hoc. * indicates P < 0.05, ** P < 0.01, and *** P < 0.001. Overall effect of

dopamine treatment: F1,70 = 71.06, P < 0.0001, rotenone treatment: F1,70 = 31.73, P < 0.0001, and

naphthazarin treatment: F1,70 = 32.62, P < 0.0001.

Page 245: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

230

ba

c

0.1 1 10 100 10000

20

40

60

80

100

120PSI alonePSI + NAP 3 µM

*

[PSI]µM

Cel

l via

bilit

y (%

con

trol

)

0.1 1 10 100 10000

20

40

60

80

100

120PSI alonePSI + DA 10 µM

** **** **

**

PSI[µM]

Cel

l via

bilit

y (%

con

trol

)

0.1 1 10 100 10000

20

40

60

80

100

120PSI alonePSI + ROT 100 µM

[PSI]µM

Cel

l via

bilit

y (%

con

trol

)

Page 246: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

231

Effect of dopamine, rotenone, or PSI on naphthazarin-induced lysosome

dysfunction

SH-SY5Y cells were incubated with a range of naphthazarin concentrations (0.01 µM–1000

µM), either alone, or in combination with an approximate EC20% of dopamine, an EC10% of

rotenone, or an EC10% of PSI. Incubation of the cells with naphthazarin alone caused a

concentration-dependent decrease in cell viability (Fig. 8.5). The addition of dopamine (10 µM)

to the naphthazarin concentration-response curve caused a significant additional decrease in cell

viability at all concentrations of naphthazarin between 0.01 µM and 30 µM (P < 0.001). For 0.01

µM and 0.1 µM naphthazarin the addition of dopamine (10 µM) caused an additional decrease in

the percentage of viable cells of 49.8% and 25.6%, respectively, indicating that the two

compounds were acting synergistically. Although the difference between the concentrations

curves for 1 µM naphthazarin with and without dopamine (10 µM) was only 18.5%, an

additional decrease in the percentage of viable cells of 23.4% was observed for dopamine (10

µM) combined with 10 µM naphthazarin, which suggests that there is synergism between

naphthazarin and dopamine (Fig. 8.5a). The addition of either rotenone (100 µM) or PSI (50

µM) to the naphthazarin concentration-response curve had little effect on the naphthazarin-

induced decrease in cell viability indicating the effects of rotenone and PSI are occluded by the

presence of naphthazarin (Fig. 8.5b,c).

Page 247: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

232

Figure 8.5. Effect of dopamine, rotenone, or PSI on naphthazarin-induced lysosomal

dysfunction. SH-SY5Y cells were incubated with naphthazarin (NAP, 0.01 µM–1000 µM),

either alone, or in the presence of approximate EC10–20% concentrations of (a) dopamine (DA, 10

µM ≈ EC20%), (b) rotenone (ROT, 100 µM ≈ EC10%), or (c) PSI (PSI, 50 µM ≈ EC10%) for 24

hours. Cell viability (% control) was assayed using alamar blue and concentration-response

curves were plotted (mean ± SEM). Diagonal shading indicates a difference of greater than 25%

between the two curves and the presence of synergy between naphthazarin and dopamine (a).

The effect of rotenone or PSI was occluded by the presence of naphthazarin, (b) and (c).

Differences between the curves were analyzed using two-way ANOVA with Bonferroni’s

multiple comparison test post-hoc. * indicates P < 0.05, ** P < 0.01, and *** P < 0.001. Overall

effect of dopamine treatment: F1,70 = 247.4, P < 0.0001, rotenone treatment: F1,70 = 2.377, P =

0.1276, and PSI treatment: F1,70 = 1.121, P = 0.2934.

Page 248: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

233

ba

c

0.01 0.1 1 10 100 10000

20

40

60

80

100

120 NAP aloneNAP + DA 10 µM

****** *** ***

***

[Naphthazarin]µM

Cel

l via

bilit

y (%

con

trol

)

0.01 0.1 1 10 100 10000

20

40

60

80

100

120 NAPNAP + ROT 100 µM

***

[Naphthazarin]µM

Cel

l via

bilit

y (%

con

trol

)

0.01 0.1 1 10 100 10000

20

40

60

80

100

120 NAPNAP + PSI 50 µM

***

[Naphthazarin]µM

Cel

l via

bilit

y (%

con

trol

)

Page 249: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

234

Discussion

Many studies have shown that dysfunction of the mitochondria, proteasome, and lysosome, as

well as increased oxidative stress and elevated protein aggregate formation contribute to cell

death in PD (Hoglinger et al. 2003; Rideout et al. 2001; Sherer et al. 2003). Furthermore, these

studies showed that some of these sub-cellular processes interact to accelerate

neurodegeneration. While previous studies have enabled a better understanding of the

mechanisms underlying neurodegeneration in PD, all the models used require lengthy

preparation times. In order to address this issue we developed a cell model that is simple and

quick to produce with characteristics that appear to mimic those in more sophisticated chronic

models of PD (Yong-Kee et al. 2011). However, due to the cellular nature of this model, caution

should be used in the extrapolation of results to processes underlying neurodegeneration in

patients with PD. Nonetheless, we predict that this simplified system is a useful method for

understanding interactions between intracellular mechanisms that are difficult to study in the

whole animal.

In this study we used our cell model to further characterise the interactions between the various

cell death mechanisms that destroy catecholaminergic neurons. In particular we were interested

in any potential synergistic relationships between different compounds that activate cell death

mechanisms associated with PD. As an EC10–20% of a second compound was added to a full

concentration response curve of the first compound, we defined an additive effect as an

additional decrease in cell viability of 10% to 20% or less, and synergism as an additional

decrease in cell viability of 12.5% to 25% or more for at least two points on the concentration

response curve, depending on the EC value of the second compound. According to this

definition, when combined the two compounds needed to produce an effect that was at least 25%

Page 250: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

235

greater than a simple addition of the effects of the two compounds to be identified as synergistic.

The definition also required that the synergistic effect be present for at least two points within the

first five of the concentration curve. Thus, we were able to identify pairs of compounds where

there was synergism, and also whether this synergism was reciprocally mediated.

The results of this study indicate that oxidative stress has synergistic effects on all other cell

death mechanisms, causing a greater decrease in cell viability than would be expected from

combining the individual effects of activating each pathway. For mitochondrial and proteasomal

dysfunction the synergism with oxidative stress was reciprocal but the relationship between

lysosomal dysfunction and oxidative stress was only unidirectional (Fig. 8.6), with an EC20%

concentration of dopamine enhancing cellular impairment caused by naphthazarin, but not vice

versa. A possible explanation for this is that, as well as being involved in protein degradation,

the lysosome also accumulates free radicals, to reduce oxidative stress. Thus, when the

lysosome is inhibited, addition of even low levels of dopamine may have a more dramatic effect

because the cells have less ability to remove the quinones and reactive oxygen species generated

through dopamine metabolism, enhancing the sensitivity of the cell to dopamine. This finding

also suggests that oxidative stress may occur as a consequence of lysosomal dysfunction. The

uni-directionality of the effect indicates that lysosomal-independent oxidative stress through

addition of dopamine does not affect lysosome function. This lack of reciprocality is probably

due to the ‘all or nothing’ effect of naphthazarin on cell death, meaning that once the lysosome is

significantly damaged, the cell has probably reached a point of no return, with regards to loss of

cell viability, thus inhibition of proteasomal or mitochondrial function will have relatively little

impact. Thus, whilst there appears to be close relationships between mitochondrial and

proteasomal dysfunction and oxidative stress, decreased cell viability as a consequence of

lysosome dysfunction is a relatively independent cell death pathway.

Page 251: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

236

Figure 8.6. Schematic diagram to show the interactions between the different mechanisms

in relation to changes in cell viability. Proteasomal, lysosomal, and mitochondrial dysfunction

form independent cell death pathways. Furthermore, proteasomal, lysosomal and mitochondria

dysfunction are exacerbated by oxidative stress. In addition there are reciprocal links between

oxidative stress, proteasomal dysfunction, and mitochondrial dysfunction.

Page 252: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

237

Mitochondriadysfunction(rotenone)

Proteasome dysfunction

(PSI)

Oxidative stress

(dopamine)

Lysosomedysfunction

(naphthazarin)

Page 253: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

238

Previous studies have shown that cell death caused by mitochondrial and proteasomal

dysfunction, as well as oxidative stress synergise to further enhance cell death (Hoglinger et al.

2003; Rideout et al. 2001; Sherer et al. 2003). The current studies take our understanding of cell

death mechanisms in PD further to show the relationship between these cell death mechanisms

and lysosomal dysfunction. Based on these studies, it appears that oxidative stress enhances

mitochondrial, lysosomal and proteasomal dysfunction in a synergistic manner. Furthermore, as

we have previously described, whether the site of the primary insult is the lysosome, proteasome,

mitochondria or increased oxidative stress, the mitochondria always become impaired (Yong-

Kee et al., 2012). Thus, all cell death mechanisms appear to converge on mitochondrial

dysfunction (Greenamyre and Hastings, 2004; Yong-Kee et al., 2012).

These studies suggest that an effective neuroprotective therapy in PD would need to target

multiple cell death pathways. Given that oxidative stress has a universal synergistic effect, and

that mitochondria are affected by all cell death mechanisms, perhaps decreasing oxidative stress

combined with enhancing mitochondrial health may prove to be the two most effective targets

for neuroprotective agents. Exposure of SH-SY5Y cells to a battery of compounds that

recapitulate the cell death mechanisms in PD, as was done here, in combination with potential

neuroprotective therapies may be a rapid and reliable method for screening such compounds.

Page 254: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

239

References

Ardley HC, Scott GB, Rose SA, Tan NG, Robinson PA (2004) UCH-L1 aggresome formation in response to proteasome impairment indicates a role in inclusion formation in Parkinson's disease. J Neurochem 90:379-391. doi:10.1111/j.1471-4159.2004.02485.x

Betarbet R, Canet-Aviles RM, Sherer TB, Mastroberardino PG, McLendon C, Kim JH, Lund S, Na HM, Taylor G, Bence NF, Kopito R, Seo BB, Yagi T, Yagi A, Klinefelter G, Cookson MR, Greenamyre JT (2006) Intersecting pathways to neurodegeneration in Parkinson's disease: Effects of the pesticide rotenone on DJ-1, alpha-synuclein, and the ubiquitin-proteasome system. Neurobiol Dis 22:404-420

Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E (2003) Staging of brain pathology related to sporadic Parkinson's disease. Neurobiol Aging 24:197-211

Brenner-Lavie H, Klein E, Ben-Shachar D (2009) Mitochondrial complex I as a novel target for intraneuronal DA: modulation of respiration in intact cells. Biochem Pharmacol 78:85-95

Caneda-Ferron B, De Girolamo LA, Costa T, Beck KE, Layfield R, Billett EE (2008) Assessment of the direct and indirect effects of MPP+ and dopamine on the human proteasome: implications for Parkinson's disease aetiology. J Neurochem 105:225-238. doi:10.1111/j.1471-4159.2007.05130.x

Cooper AA, Gitler AD, Cashikar A, Haynes CM, Hill KJ, Bhullar B, Liu K, Xu K, Strathearn KE, Liu F, Cao S, Caldwell KA, Caldwell GA, Marsischky G, Kolodner RD, Labaer J, Rochet JC, Bonini NM, Lindquist S (2006) Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron loss in Parkinson's models. Science 313:324-328

Goldman JE, Yen SH, Chiu FC, Peress NS (1983) Lewy bodies of Parkinson's disease contain neurofilament antigens. Science 221:1082-1084

Greenamyre JT, Hastings TG (2004) Biomedicine. Parkinson's--divergent causes, convergent

mechanisms. Science 304:1120-1122

Hoglinger GU, Carrard G, Michel PP, Medja F, Lombes A, Ruberg M, Friguet B, Hirsch EC (2003) Dysfunction of mitochondrial complex I and the proteasome: interactions between two biochemical deficits in a cellular model of Parkinson's disease. J Neurochem 86:1297-1307

Imashuku S, Inui A, Nakamura T, Tanaka J, Miyake S (1973) Catecholamine metabolism in tissue culture cells of a neuroblastoma. J Clin Endocrinol Metab 36:931-936

Jana S, Maiti AK, Bagh MB, et al. (2007) Dopamine but not 3,4-dihydroxy phenylacetic acid (DOPAC) inhibits brain respiratory chain activity by autoxidation and mitochondrea

Page 255: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

240

catalyzed oxdiation to quinone products: implications in Parkinson's disease. Brain Res 1139:195-200

Jenner P (2003) Oxidative stress in Parkinson's disease. Ann Neurol 53 Suppl 3:S26-36; discussion S36-28. doi:10.1002/ana.10483

Khan FH, Sen T, Maiti AK, Jana S, Chatterjee U, Chakrabati S (2005) Inhibition of rat brain mitochondrial electron transport chain activity by dopamine oxidation products during extended in vitro incubation: implications for Parkinson's disease. Biochim Biophys Acta 1741:65-74

Lannuzel A, Michel PP, Hoglinger GU, Champy P, Jousset A, Medja F, Lombes A, Darios F, Gleye C, Laurens A, Hocquemiller R, Hirsch EC, Ruberg M (2003) The mitochondrial complex I inhibitor annonacin is toxic to mesencephalic dopaminergic neurons by impairment of energy metabolism. Neuroscience 121:287-296

Li Z, Arnaud L, Rockwell P, Figueiredo-Pereira ME (2004) A single amino acid substitution in a proteasome subunit triggers aggregation of ubiquitinated proteins in stressed neuronal cells. J Neurochem 90:19-28. doi:10.1111/j.1471-4159.2004.02456.x

Massey AC, Follenzi A, Kiffin R, Zhang C, Cuervo AM (2008) Early cellular changes after blockage of chaperone-mediated autophagy. Autophagy 4:442-456

Nuber S, Petrasch-Parwez E, Winner B, Winkler J, von Horsten S, Schmidt T, Boy J, Kuhn M, Nguyen HP, Teismann P, Schulz JB, Neumann M, Pichler BJ, Reischl G, Holzmann C, Schmitt I, Bornemann A, Kuhn W, Zimmermann F, Servadio A, Riess O (2008) Neurodegeneration and motor dysfunction in a conditional model of Parkinson's disease. J Neurosci 28:2471-2484. doi:10.1523/JNEUROSCI.3040-07.2008

Olanow WC (2008) Levodopa/dopamine replacement strategies in Parkinson's disease--future directions. Movement Disorders 23:S613-S622

Pan T, Kondo S, Le W, Jankovic J (2008) The role of autophagy-lysosome pathway in neurodegeneration associated with Parkinson's disease. Brain 131:1969-1978. doi:10.1093/brain/awm318

Reaney SH, Johnston LC, Langston WJ, Di Monte DA (2006) Comparison of the neurotoxic effects of proteasomal inhibitors in primary mesencephalic cultures. Exp Neurol 202:434-440. doi:10.1016/j.expneurol.2006.07.010

Richfield EK, Thiruchelvam MJ, Cory-Slechta DA, Wuertzer C, Gainetdinov RR, Caron MG, Di Monte DA, Federoff HJ (2002) Behavioral and neurochemical effects of wild-type and mutated human alpha-synuclein in transgenic mice. Exp Neurol 175:35-48. doi:10.1006/exnr.2002.7882

Rideout HJ, Larsen KE, Sulzer D, Stefanis L (2001) Proteasomal inhibition leads to formation of ubiquitin/alpha-synuclein-immunoreactive inclusions in PC12 cells. J Neurochem 78:899-908

Page 256: Determination of the Sub-Cellular Mechanisms Underlying ... · toxins which mimic purported pathological processes in PD reveal mitochondrial membrane potential becomes depolarized,

241

Rockenstein E, Schwach G, Ingolic E, Adame A, Crews L, Mante M, Pfragner R, Schreiner E, Windisch M, Masliah E (2005) Lysosomal pathology associated with alpha-synuclein accumulation in transgenic models using an eGFP fusion protein. J Neurosci Res 80:247-259. doi:10.1002/jnr.20446

Sherer TB, Kim JH, Betarbet R, Greenamyre JT (2003) Subcutaneous rotenone exposure causes highly selective dopaminergic degeneration and alpha-synuclein aggregation. Exp Neurol 179:9-16

Sherer TB, Richardson JR, Testa CM, Seo BB, Panov AV, Yagi T, Matsuno-Yagi A, Miller GW, Greenamyre JT (2007) Mechanism of toxicity of pesticides acting at complex I: relevance to environmental etiologies of Parkinson's disease. J Neurochem 100:1469-1479. doi:10.1111/j.1471-4159.2006.04333.x

Testa CM, Sherer TB, Greenamyre JT (2005) Rotenone induces oxidative stress and dopaminergic neuron damage in organotypic substantia nigra cultures. Brain Res Mol Brain Res 134:109-118. doi:10.1016/j.molbrainres.2004.11.007

Vogiatzi T, Xilouri M, Vekrellis K, Stefanis L (2008) Wild type alpha-synuclein is degraded by chaperone-mediated autophagy and macroautophagy in neuronal cells. J Biol Chem 283:23542-23556. doi:10.1074/jbc.M801992200

von Bohlen Und Halbach O, Krieglstein K, Schober A, Schulz JB (2004) The dopaminergic nigrostriatal system: development, physiology, disease. Cell Tissue Res 318:3. doi:10.1007/s00441-004-0963-x

Wislet-Gendebien S, D'Souza C, Kawarai T, St George-Hyslop P, Westaway D, Fraser P, Tandon A (2006) Cytosolic proteins regulate alpha-synuclein dissociation from presynaptic membranes. J Biol Chem 281:32148-32155. doi:10.1074/jbc.M605965200

Wislet-Gendebien S, Visanji NP, Whitehead SN, Marsilio D, Hou W, Figeys D, Fraser PE, Bennett SA, Tandon A (2008) Differential regulation of wild-type and mutant alpha-synuclein binding to synaptic membranes by cytosolic factors. BMC Neurosci 9:92. doi:10.1186/1471-2202-9-92

Yong-Kee C, Salomonczyk D, Nash J (2011) Development and validation of a screening assay for the evaluation of putative neuroprotective agents in the treatment of Parkinson's disease. Neurotox Res 19:519-526

Yong-Kee CJ, Sidorova E, Hanif A, Perera G, Nash JE (2012) Mitochondrial dysfunction precedes other sub-cellular abnormalities in an in vitro model linked with cell death in Parkinson’s disease. Neurotox Res 21: 185-194