15.08.2013 Views

Copyright Arati Ajinkya Inamdar 2009 - acumen - The University of ...

Copyright Arati Ajinkya Inamdar 2009 - acumen - The University of ...

Copyright Arati Ajinkya Inamdar 2009 - acumen - The University of ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

DISCOVERY OF A NITRIC OXIDE SYNTHASE-DEPENDENT RESPONSE AND A<br />

FUNCTIONAL ANALYSIS OF GENES REGULATING THE RESPONSE IN A<br />

DROSOPHILA MODEL OF PARKINSON’S DISEASE<br />

by<br />

ARATI AJINKYA INAMDAR<br />

A DISSERTATION<br />

Submitted in partial fulfillment <strong>of</strong> the requirements<br />

for the degree <strong>of</strong> Doctor <strong>of</strong> Philosophy in the<br />

Department <strong>of</strong> Biological Sciences<br />

in the Graduate School <strong>of</strong><br />

<strong>The</strong> <strong>University</strong> <strong>of</strong> Alabama<br />

TUSCALOOSA, ALABAMA<br />

<strong>2009</strong>


<strong>Copyright</strong> <strong>Arati</strong> <strong>Ajinkya</strong> <strong>Inamdar</strong> <strong>2009</strong><br />

ALL RIGHTS RESERVED


ABSTRACT<br />

<strong>The</strong> processes <strong>of</strong> neuroinflammation and oxidative stress are thought to be among the<br />

primary mechanisms playing roles in the etiology and pathophysiology <strong>of</strong> Parkinson’s disease<br />

(PD). Recently, it has been proposed that microglia, the innate immune cells <strong>of</strong> the mammalian<br />

brain, become hyperactivated and deregulated in response to neuronal dysfunction in PD and<br />

thereby, accelerate dopaminergic neuronal loss. <strong>The</strong> mechanisms for neuroinflammatory<br />

responses that exaggerate neurotoxicity are poorly understood. Moreover, the studies to date that<br />

investigate neuroinflammatory mechanisms have utilized primarily in vitro approaches. We have<br />

established a Drosophila PD model based on ingestion <strong>of</strong> the herbicide paraquat, which<br />

recapitulates most behavioral and patho-physiological features <strong>of</strong> PD, including loss <strong>of</strong><br />

dopaminergic neurons. Using this model, we have discovered that paraquat ingestion induces<br />

nitric oxide synthase (NOS) and a corresponding elevation <strong>of</strong> NO production in the adult<br />

Drosophila brain. Mammalian microglia have been shown to be a source <strong>of</strong> NOS, recognized as<br />

a major component/marker in neuron death, in mammalian PD models. <strong>The</strong>refore, the<br />

observation <strong>of</strong> NOS induction during Drosophila neurodegeneration parallels the mammalian<br />

process. <strong>The</strong> paraquat-induced stimulation <strong>of</strong> NOS was further confirmed using pharmacological<br />

approaches, which demonstrated that inhibition <strong>of</strong> NOS partially rescued adult Drosophila from<br />

the deleterious effects <strong>of</strong> paraquat. Minocycline, a tetracycline derivative, has been reported to<br />

act primarily on microglia, ameliorating excessive activation <strong>of</strong> NOS, in mammalian<br />

neurodegenerative disease models including PD. Similarly, ingestion <strong>of</strong> minocycline by adult<br />

ii


Drosophila ameliorates the effects <strong>of</strong> paraquat, including reduction <strong>of</strong> NOS activity and<br />

protection <strong>of</strong> dopaminergic neurons. <strong>The</strong> paraquat-induced PD model was also employed to<br />

identify the mechanisms <strong>of</strong> action that confer the neuroprotective properties <strong>of</strong> minocycline.<br />

Components <strong>of</strong> signaling pathways that potentially could mediate DA toxicity in this PD model<br />

were tested for their ability to modify the action <strong>of</strong> minocycline.<br />

<strong>The</strong> discovery <strong>of</strong> a NOS-dependent paraquat response in flies provides the foundation for<br />

future work to explore cellular and molecular mechanisms directing NOS induction and action in<br />

Drosophila, which exhibits features resembling neuroinflammation. This research also takes<br />

advantage <strong>of</strong> the ease <strong>of</strong> genetic and pharmacological methods in the Drosophila model to<br />

identify important genetic components <strong>of</strong> this process.<br />

iii


3-IT 3- iodo-tyrosine<br />

6-OHDA 6-hydroxydopamine<br />

o C Celsius<br />

Apaf-1 Apoptosis activating factor-1<br />

ATP Adenosine triphosphate<br />

LIST OF ABBEREVIATIONS AND SYMBOLS<br />

BDNF Brain -derived-neurotrophic factor<br />

BH4 6(R)L-erythro-5,6,7,8-tetrahydrobiopterin or Tetrahydrobiopterin<br />

Catsup Catecholamines up<br />

CD 14 Cluster <strong>of</strong> Differentiation 14<br />

cDNA Complementary deoxyribonucleic acid<br />

COX 1 Cyclooxygenase -1<br />

COX 2 Cyclooxygenase -2<br />

CSF Cerebrospinal fluid<br />

DA Dopamine<br />

DAT Dopamine transporter<br />

dDAT Drosophila dopamine transporter<br />

Ddc Dopa decarboxylase<br />

Diablo/Smac Direct IAP Binding protein with low pI/second mitochondria derived<br />

activators <strong>of</strong> caspases<br />

iv


DOPAC 3,4-Dihydroxy-phenylacetic acid<br />

dTH1 Drosophila tyrosine hydroxylase 1<br />

dTH2 Drosophila tyrosine hydroxylase 2<br />

dsRNAi double stranded RNA interference<br />

ERK Extracellular signal–regulated kinases<br />

FAD Flavin adenine dinucleotide<br />

FADD Fas associated death domain<br />

FMN Flavin mononucleotide<br />

GDNF Glial cell-line-derived-neurotrophic factor<br />

GFP Green fluorescent protein<br />

gmc Glial cells missing<br />

gmc2 Glial cells missing 2<br />

GSH Glutathione<br />

GTP Guanosine triphosphate<br />

GTPCH GTP cyclohydrolase<br />

HPLC High pressure liquid chromatography<br />

IAP Inhibitor <strong>of</strong> apoptosis<br />

ICE IL-1β converting enzyme<br />

IFN γ Interferon gamma<br />

IKK α NF-inhibitor<br />

iNOS Inducible nitric oxide synthase<br />

IL-6 Interleukin 6<br />

IL-1α Interleukin 1 alpha<br />

v


IL-1β Interleukin 1 beta<br />

JNK c-Jun NH2-terminal kinases<br />

kDa kilo Dalton<br />

L-DOPA 3, 4-dihydroxy-L-phenylalanine<br />

L-NAME N G -nitro-L-arginine methyl ester<br />

L-NMMA N G -monomethyl-L-arginine<br />

MAO Monoamine oxidase<br />

MAPKs Mitogen activated protein kinases<br />

MKKs Mitogen-activated protein kinase kinases<br />

MKKKs Mitogen-activated protein kinase kinase kinases<br />

mg Milligram<br />

MHC Major histo-compatibility complex<br />

ml Milliliter<br />

mM Millimolar<br />

MPP+ 1-methyl-4-phenylpyridinium<br />

MPTP 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine<br />

NADP Nicotinamide Adenine Dinucleotide Phosphate<br />

NF-κβ Nuclear factor kappa beta<br />

NH2PPP Dihydroneoperin triphosphate<br />

nm Nanometer<br />

NO Nitric oxide<br />

NOS Nitric oxide synthase<br />

NSAIDs Non steroidal anti inflammatory drugs<br />

vi


O2 − Superoxide radicals<br />

PBS Phosphate buffered saline<br />

PBT Phosphate buffered saline with Tween 20<br />

PCD Programmed cell death<br />

PD Parkinson’s disease<br />

PI3K Phosphoinositide 3-kinase<br />

PKB Protein kinase B<br />

PKC Protein kinase C<br />

ple Pale<br />

ppt Pointed<br />

PQ N,N'-dimethyl-4,4'-bipyridinium dichloride or Paraquat<br />

PQ .+ Paraquat radical<br />

PTP 6-pyruvoyl tetra-hydropterin<br />

Pu Punch<br />

rpr reaper<br />

repo Reverse polarity<br />

RNS Reactive nitrogen species<br />

ROI Reactive oxygen intermediate<br />

ROS Reactive oxygen species<br />

SN Substantia nigra<br />

SNpc Substantia nigra pars compacta<br />

TDC Tyrosine decarboxylase<br />

TH Tyrosine hydroxylase<br />

vii


TNF α Tumor necrotic factor alpha<br />

ttk Tramtrack<br />

VMAT Vesicular monoamine transporter<br />

VTA Ventral tegmental area<br />

μg Microgram<br />

μl Microliter<br />

μM Micromolar<br />

μmol Micromole<br />

μm Micrometer<br />

viii


ACKNOWLEDGMENTS<br />

It is my great pleasure to convey my acknowledgements to people whose valuable<br />

presence and support has made this journey successful. First <strong>of</strong> all, I would like to thank God to<br />

bless me with highly knowledgeable, intelligent and caring husband, Dr. <strong>Ajinkya</strong> C. <strong>Inamdar</strong>,<br />

without whom I would have never achieved this honor. I am so fortunate to be a mother <strong>of</strong> our<br />

son, Atharva A. <strong>Inamdar</strong>, who has made these years memorable with his smiles, babbles and big<br />

first footsteps.<br />

I am also thankful to God to fulfill my life with love and encouragements from my<br />

parents, Mangala and Arun Thoke throughout my life. I will always owe you for your teachings<br />

and blessings. My infinite thanks to my in-laws, Shilpa and Chintamani <strong>Inamdar</strong>, for their help,<br />

care and advice since I have become member <strong>of</strong> their family. <strong>Ajinkya</strong> and I thank you all for<br />

being so wonderful and loving grand-parents <strong>of</strong> Atharva. I would also like to thank my relatives<br />

in India, especially my brother Samir, my sister-in-law Priya and nephew Ishaan for their<br />

affection and support. .<br />

I would like to thank my advisor, Dr. Janis O’ Donnell, for her timely support, guidance<br />

and advice. Her talent, knowledge, positive criticism, persistence and understanding are<br />

invaluable to develop me as a scientist in addition to a physician. You have given a new direction<br />

to my life and I believe that these achievements will succeed in imparting unique contribution to<br />

the world.<br />

ix


I am thankful to my committee members, Drs. Guy and Kim Caldwell, Dr. Perry<br />

Churchill and Dr. Ed Stephenson, for their guidance in my research projects throughout these<br />

years. Many thanks to Guy for his timely help to obtain post-doctoral position at Rutgers<br />

<strong>University</strong>, New Jersey with Dr. Joan W. Bennett. I appreciate Joan’s support for my post-<br />

doctoral project and hope to make valuable contribution for the project.<br />

I appreciate past and present graduate and undergraduate students <strong>of</strong> Dr. O’Donnell lab<br />

for their friendship, advice and help. I also thank members <strong>of</strong> Dr. Bennett lab for their help<br />

during our initial period <strong>of</strong> adjustment in New Jersey. Finally, many thanks to faculty and staff<br />

members especially Mary Musumecci at the Department <strong>of</strong> Biological Sciences, UA and Liz<br />

Scarpa at the Department <strong>of</strong> Plant Biology and Pathology at Rutgers <strong>University</strong> for their<br />

friendliness, open-heartedness and time throughout these years.<br />

x


CONTENTS<br />

ABSTRACT……………………………………………………………..…………………….....ii<br />

LIST OF ABBEREVIATIONS AND SYMBOLS………………………………..…………......iv<br />

ACKNOWLEDGMENTS………………………………………………...………………..…....ix<br />

LIST OF TABLES……………………………………………………………………...............xiv<br />

LIST OF FIGURES……………………………………………………………………………..xv<br />

1. INTRODUCTION……………………………………………………………………………..1<br />

i. Dopamine and dopamine biosynthesis pathway in mammals………………………………….1<br />

ii. Drosophila dopamine biosynthesis pathway…………………………………………………..2<br />

iii. DA packaging and transportation……………………………………………………………..4<br />

iv. Drosophila DA biosynthesis pathway regulators……………………………………………..4<br />

v. Functions <strong>of</strong> DA in Drosophila………………………………………………………………..8<br />

vi. Parkinson’s disease……………………………………….………………………………….11<br />

vii. Parkinson’s disease and oxidative stress……………….…………………………………...12<br />

viii. Parkinson’s disease and Neuroinflammation……………………………………………....17<br />

ix. Glia in mammals and Drosophila…………………………………………………………...18<br />

x. Microglia…………………………………………………….……………………………….22<br />

xi. Neuroinflammation in experimental models <strong>of</strong> PD………………….……………………...25<br />

xii. Inhibition <strong>of</strong> neuroinflammation in PD models…………………………………………….25<br />

xiii. Nitric oxide………………………………………………………………………………...27<br />

xi


xiv. NO signaling in invertebrates……………………………………………………………....30<br />

xv. Drosophila NOS and NO signaling………………………………………………………....30<br />

xvi. Role <strong>of</strong> NO in Drosophila immune response……………………………………………....31<br />

xvii. Role <strong>of</strong> Signaling pathways in the pathogenesis <strong>of</strong> PD…………………………………...32<br />

xviii. Apoptosis signaling pathway……………………………………………………………..32<br />

xix. PCD in Drosophila…………………………………………………………………………35<br />

xx. Mitogen activation protein kinase signaling pathway ……………………………………...36<br />

xxi. JNK signaling pathway……………………………………………………….....................37<br />

xxii. Drosophila JNK…………………………………………………………………………...39<br />

xxiii. JNK signal transduction pathway and Parkinson’s disease………………………………39<br />

xxiv. ERK pathway in mammals………………………………………………………………..40<br />

xxv. ERK in Drosophila………………………………………………………… …………......41<br />

xxvi. ERK signaling pathway and Parkinson’s disease ………………………………...............41<br />

xxvii. Akt signaling pathway………………………………………………………....................42<br />

xxviii. Drosophila Akt signaling pathway……………………………………………………....43<br />

xxix. Akt signaling pathway and Parkinson’s disease……………………………………..........43<br />

2. DROSOPHILA MOUNTS A NITRIC OXIDE-DEPENDENT RESPONSE TO<br />

PARAQUAT-INDUCED DEGENERATION OF DOPAMINERGIC NEURONS…………...48<br />

i. Introduction…………………………………………………………………………………...49<br />

ii. Materials and Methods…………………………………………………………………..…...53<br />

iii. Results…………………………………………………………………………………..…...57<br />

iv. Discussion…………………………………………………..……………………………….73<br />

3. IDENTIFICATION OF A NEUROPROTECTIVE ROLE FOR MINOCYCLINE IN A<br />

DROSOPHILA MODEL OF PARKINSON’S DISEASE AND FUNCTIONAL<br />

ANALYSIS OF SIGNALING PATHWAYS MEDIATING PARAQUAT TOXICITY IN<br />

xii


A DROSOPHILA PD MODEL……………………………………………………………........78<br />

i. Introduction…………………………………………………………………………………...79<br />

ii. Materials and Methods……………………………………………………………………….83<br />

iii. Results……………………………………………………………………………………….88<br />

iv. Discussion…………………………………………………………………………………..106<br />

4. BRIEF EXPOSURE TO PARAQUAT DURING JUVENILE AND EARLY ADULT<br />

STAGES CAUSES PARKINSONIAN SYMPTOMS, INCREASED SENSITIVITY TO<br />

OXIDATIVE INSULT LATER IN LIFE AND A SHORTENED LIFE<br />

SPAN…………………………………………………………………………………………..112<br />

i. Introduction………………………………………………………………………………….113<br />

ii. Materials and Methods……………………………………………………………………...116<br />

iii. Results…...…………………………………………………………………………………118<br />

iv. Discussion...………………………………………………………………………………..135<br />

5. IDENTIFICATION OF A NITRIC OXIDE-DEPENDENT RESPONSE IN<br />

S. VENEZUELAE-INDUCED PARKINSON’S DISEASE IN DROSOPHILA<br />

MELANOGASTER..…………………………………………………………………………...140<br />

i. Introduction………………………………………………………………………………….141<br />

ii. Materials and Methods………………………………………...……………………..……..144<br />

iii. Results…………………………………………………………...………………………....146<br />

iv. Discussion………………………………………………………...………………………...159<br />

6. SUMMARY, FUTURE DIRECTIONS AND APPLICATIONS……..………....................164<br />

i. Drosophila and neuroinflammatory-like response…………………………………………..164<br />

ii. Drosophila and signal transduction pathway in PD………………………………………...171<br />

iii. Drosophila as a model to study Early Onset Parkinsonism………………………………..172<br />

7. REFERENCES………………………..…………………………………………………….174<br />

xiii


LIST OF TABLES<br />

1.1. Similarity in the function and distribution <strong>of</strong> vertebrate and Drosophila glia in<br />

CNS……………………………………………………………………………………………..21<br />

xiv


LIST OF FIGURES<br />

1.1. Dopamine Biosynthesis Pathway in Drosophila………………………………………….....6<br />

1.2. <strong>The</strong> BH4 biosynthesis pathway………………………………………………. ……………..7<br />

1.3. <strong>The</strong> schematic diagram showing the location and number <strong>of</strong> DA neurons in<br />

adult Drosophila brain viewed from anterior and posterior aspects…………………………….10<br />

1.4. <strong>The</strong> sagittal section <strong>of</strong> human brain showing major regions <strong>of</strong> the<br />

human brain…….……………………………………………………………………………….13<br />

1.5. <strong>The</strong> chemical structure <strong>of</strong> paraquat (PQ)……………………….…………………………..16<br />

1.6. <strong>The</strong> two step mechanism <strong>of</strong> PQ toxicity……………………………………….…………...16<br />

1.7. <strong>The</strong> Fenton reaction………………………………………………………………………...16<br />

1.8. An outcome <strong>of</strong> the sustained activation <strong>of</strong> microglia in chronic<br />

neurodegenerative disease ……………………………………………………………………..24<br />

1.9. A schematic diagram showing the components involved in the formation<br />

<strong>of</strong> nitric oxide (NO)………………………………………………………………………..........29<br />

1.10. Comparison <strong>of</strong> mammalian and Drosophila apoptotic signaling pathways……………....34<br />

1.11. <strong>The</strong> schematic diagram showing the components <strong>of</strong> MAPK signaling pathway in<br />

mammals and Drosophila……………………………………………………………………….38<br />

1.12. A schematic diagram showing the signaling pathways associated with<br />

activated Akt…………………………………………………………………………………….45<br />

2.1. Dopamine, tetrahydrobiopterin, and nitric oxide biosynthesis pathways………………......56<br />

2.2. L-NAME, an inhibitor <strong>of</strong> NOS, improves survival duration <strong>of</strong> adults<br />

exposed to paraquat………………………………………………………… ………………….58<br />

2.3. Effect <strong>of</strong> ingestion <strong>of</strong> minocycline on wild type flies with and without PQ on<br />

survival duration <strong>of</strong> wild type flies ……………………………………………………………..60<br />

xv


2.4. PQ induces, and minocycline suppresses, NOS activity in adult flies……………………..62<br />

2.5. Minocycline shows differential effects on paraquat-induced damage in dopamine<br />

regulatory mutants …………………...………………………………………………………....65<br />

2.6. Exposure to PQ causes induction <strong>of</strong> NOS in adult brain …………………………………..68<br />

2.7. Ingestion <strong>of</strong> PQ (1.5 mM) induces NOS-expression near and around<br />

dopaminergic neurons in TH-GAL4; UAS-eGFP adult brain……………………………….…..70<br />

2.8. Paraquat-induced NOS is expressed in non-glial cells in adult fly brain………..................72<br />

3.1. Effect <strong>of</strong> minocycline and doxycycline with 10 mM paraquat on survival <strong>of</strong> adult male<br />

flies. ………………..…………………………………………………...………........................90<br />

3.2. Minocycline protects against PQ-induced mobility defects…………………….………….92<br />

3.3. Minocycline confers protection to dopaminergic neurons…………………………….…...94<br />

3.4. Minocycline delays paraquat induced selective loss <strong>of</strong><br />

dopaminergic neurons...………………………..……………………………………………….95<br />

3.5. Minocycline blocks changes in DA pathway components indicative <strong>of</strong> PQ-induced<br />

oxidative stress……………………………………………………………….............................98<br />

3.6. Minocycline reduces PQ-generated reactive oxygen species……………………………..101<br />

3.7. Paraquat induced inflammatory response is regulated by JNK and Akt<br />

signaling pathways.…………………………………………………………………………….103<br />

3.8. Over-Expression <strong>of</strong> wild type JNK and Akt provide protection against PQ……………...105<br />

4.1. Exposure <strong>of</strong> juvenile (2nd instar larvae) and young adult (


5.2. Exposure to crude conditioned medium <strong>of</strong> S. venezualae induces<br />

Parkinsonian-like mobility defects……………………………………………..……………...149<br />

5.3. Exposure to crude conditioned medium <strong>of</strong> S. venezualae induces<br />

loss <strong>of</strong> DA neuron………………………………………………………………………..…….151<br />

5.4. Exposure <strong>of</strong> crude conditioned medium <strong>of</strong> S. venezualae induced NOS near DA<br />

neuron in TH-GAL4; UAS-eGFP transgenic strains………………………………….…….….155<br />

5.5. Exposure <strong>of</strong> S. venezuelae conditioned medium causes expression <strong>of</strong> NOS around<br />

glial cells in adult brain………………………………………………………...........................158<br />

xvii


CHAPTER 1<br />

INTRODUCTION<br />

Dopamine and dopamine biosynthesis pathway in mammals<br />

Dopamine (DA) is the major neurotransmitter and neurohormone known to be present in<br />

all invertebrates and vertebrates. A member <strong>of</strong> the catecholamine family, DA is chemically<br />

known as 4-(2-aminoethyl) benzene-1, 2-diol. It is a precursor to norepinephrine and epinephrine<br />

in the catecholamine biosynthesis pathway. DA neurons originate in the substantia nigra pars<br />

compacta (SNpc), ventral tegmental area (VTA), and the hypothalamus from where axons are<br />

projected into different regions <strong>of</strong> the brain. <strong>The</strong> four major pathways are the mesocortical,<br />

mesolimbic, nigrostriatal and tuberoinfundibular pathways (Iversen and Iversen, 2007; Björklund<br />

and Dunnett, 2007).<br />

<strong>The</strong> mesocortical pathway connects the ventral tegmentum to the frontal lobes <strong>of</strong> the<br />

cerebral cortex and is known to be involved in emotional and motivational responses.<br />

Deregulation <strong>of</strong> this pathway is associated with psychotic behavior, including schizophrenia as<br />

well as impairment <strong>of</strong> the memory (Berman et al., 1988; Weinberger et al., 1988).<br />

<strong>The</strong> mesolimbic pathway connects the ventral tegmentum to the nucleus accumbens in<br />

the striatum. This pathway is mainly involved in reward and pleasure related activities (Berridge<br />

and Robinson, 1998). <strong>The</strong> tendency <strong>of</strong> individuals towards addiction to certain drugs such as<br />

cocaine, morphine, amphetamines, and alcohol has been correlated with the dysfunction <strong>of</strong> the<br />

1


mesolimbic pathway (Beitner-Johnson and Nestler, 1991; Leyton et al., 2002; Boileau et al.,<br />

2003).<br />

<strong>The</strong> nigrostriatal pathway is crucial pathway for locomotor function and connects the<br />

substantia nigra to the striatum. This pathway forms a part <strong>of</strong> the basal ganglia motor loop and<br />

thus indirectly interconnects with the cerebral cortex, thalamus and brainstem and functions in<br />

the motor control, cognition, emotions, and learning. Loss <strong>of</strong> dopaminergic neurons in this<br />

circuit results in Parkinson’s disease, the most common movement disorder associated with<br />

motor deficits as well as cognitive and emotional disorders (Dunnett et al., 1981).<br />

<strong>The</strong> tuberoinfundibular pathway originates from the arcuate nucleus <strong>of</strong> the mediobasal<br />

hypothalamus and projects the axons to the infundibular region. <strong>The</strong> function <strong>of</strong> the<br />

tuberoinfundibular pathway is mainly implicated in lactation and sexual arousal (Gunnet et al.,<br />

1986; Moore et al., 1987).<br />

Drosophila dopamine biosynthesis pathway<br />

In Drosophila, as in mammals, DA is an essential neurotransmitter and neuromodulator<br />

functionally important for various biological activities such as learning, behavior, locomotion,<br />

memory, and development <strong>of</strong> the reproductive system (Neckameyer, 1996). <strong>The</strong> DA biosynthesis<br />

pathway <strong>of</strong> Drosophila is well-conserved with that <strong>of</strong> mammals. <strong>The</strong> genes encoding the<br />

enzymes regulating dopamine biosynthesis pathway in mammals also function in the similar<br />

manner in Drosophila (Livingstone and Tempel, 1983). Briefly, the first step in the<br />

catecholamine biosynthesis is the hydroxylation <strong>of</strong> tyrosine into 3,4-dihydroxy-L-phenylalanine<br />

(L-DOPA) via the enzyme tyrosine hydroxylase (TH). This is the rate limiting step in the<br />

pathway, and TH undergoes multiple levels <strong>of</strong> regulation as described below. L-DOPA is then<br />

2


converted into DA by dopa decarboxylase (Ddc) (Nagatsu et al., 1964). <strong>The</strong> catecholamine<br />

biosynthesis pathway terminates after formation <strong>of</strong> DA from tyrosine. In addition, tyrosine acts<br />

as a precursor for tyramine, another biogenic amine in Drosophila via tyrosine decarboxylase<br />

(TDC). Tyramine is converted into octopamine, another biogenic amine known to be a functional<br />

homolog <strong>of</strong> norepinephrine in Drosophila using tyramine β hydroxylase (Cole et al., 2005). <strong>The</strong><br />

genes regulating formation <strong>of</strong> DA and tyramine from tyrosine are shown in Fig. 1.1.<br />

A cDNA for the Drosophila gene encoding TH was isolated from adult head using a<br />

cDNA for rat TH as a probe, indicating a strong conservation between Drosophila and mammals.<br />

Further, Drosophila TH demonstrates 50 % amino-acid sequence identity and 80.5 % similarity<br />

to rat TH (Neckameyer and Quinn, 1989). TH is found to be expressed from embryonic stage 15<br />

onwards in Drosophila (Lundell and Hirsh, 1994). <strong>The</strong> mammalian TH exists as a homotetramer<br />

with C-terminal catalytic domain and N-terminal regulatory domain (Grima et al., 1985).<br />

Conservation <strong>of</strong> amino acid sequence with mammalian TH is restricted to the predicted catalytic<br />

domain <strong>of</strong> Drosophila TH; the presumptive N-terminal regulatory region is largely non-<br />

conserved. Nevertheless, fly neuronal TH can be detected by mammalian TH antibody and<br />

corresponds to a 58 kDa band in western blots (Vié et al., 1999)<br />

In contrast to human TH, which possesses four is<strong>of</strong>orms, Drosophila expresses two<br />

is<strong>of</strong>orms, both encoded by pale. Drosophila TH1 (dTH1) is expressed mainly in the CNS while<br />

Drosophila TH2 (dTH2) is expressed in non-neural tissues, predominantly in cuticular hypoderm<br />

(Birman et al., 1994). <strong>The</strong> dTH1 mRNA is 3.7 kb long, translating into an 80 kDa protein, while<br />

dTH2 mRNA is a 3.2 kb in length translating to a 56 kDa protein (Birman et al., 1994).<br />

3


DA packaging and transportation<br />

Once DA is synthesized in the neuron, Drosophila vesicular monoamine transporter<br />

(VMAT) mediates its packaging into synaptic vesicles. Drosophila VMAT shares high sequence<br />

similarity with mammalian VMAT (Greer et al., 2005). DA is recycled back into the cytoplasm<br />

<strong>of</strong> the presynaptic neuron after being released into the synaptic cleft for post-synaptic<br />

transmission by Drosophila Dopamine transporter (dDAT). dDAT is a twelve transmembrane<br />

domain protein functionally conserved with human DAT. <strong>The</strong> dDAT gene is located on the right<br />

arm <strong>of</strong> the second chromosome and encodes a single DAT is<strong>of</strong>orm (Pörzgen et al., 2001).<br />

In mammals, cytoplasmic DA is converted into 3, 4-Dihydroxy-Phenylacetic Acid<br />

(DOPAC) in the presynaptic region by Monoamine oxidase (MAO). However, in Drosophila,<br />

although MAO has been predicted by sequence analysis, its functional role is still unknown.<br />

Nevertheless, DA in the fly-brain is converted into DOPAC via N-acetylation <strong>of</strong> DA, and level<br />

<strong>of</strong> DOPAC can be detected by HPLC (Downer and Martin, 1987). <strong>The</strong> DA released at the<br />

presynaptic membrane interacts with DA receptors mediating the transmission <strong>of</strong> stimulus across<br />

post-synaptic region and hence play a key role in DA signaling. In Drosophila, there are two<br />

classes <strong>of</strong> DA receptors: D1-like and D2-like, based on their biochemical and pharmacological<br />

properties. <strong>The</strong>se two classes <strong>of</strong> receptors show functional homology with mammalian DA<br />

receptors (Hearn et al., 2002).<br />

Drosophila DA biosynthesis pathway regulators<br />

In Drosophila, DA biosynthesis is well-conserved with that <strong>of</strong> mammals. It involves two<br />

enzymatic pathways regulating the DA synthesis at different levels as mentioned above. DA<br />

4


synthesis is regulated by the rate-limiting enzyme, TH, encoded by pale (ple) (Neckameyer and<br />

White, 1993).<br />

TH activity is further regulated via tetrahydrobiopterin (BH4), a member <strong>of</strong> the pteridine<br />

family, which acts as a crucial co-factor for DA synthesis pathway (Nagatsu et al., 1964). BH4 is<br />

synthesized via two pathways, a de novo one where guanosine triphosphate (GTP) functions as a<br />

precursor and other one a salvage pathway utilizing enzymatically reducible dihydropterins (Fig.<br />

1.2). Briefly, GTP is converted into dihydroneoperin triphosphate (NH2PPP) via GTP<br />

cyclohydrolase (GTPCH), the first and rate-limiting enzyme <strong>of</strong> the pathway. NH2PPP is then<br />

converted into 6-pyruvoyl tetra-hydropterin (PTP) which in turn is converted into 6(R)L-erythro-<br />

5,6,7,8-tetrahydrobiopterin (BH4) in two successive steps utilizing PTP synthase and sepiapterin<br />

reductase, respectively (Thöny et al., 2000). GTPCH is encoded by the Punch (Pu) locus located<br />

on the right arm <strong>of</strong> the second chromosome (Mackey and O’Donnell, 1983). Both ple and Pu<br />

loss <strong>of</strong> function mutants are homozygous lethal, but the heterozygous mutants are viable and<br />

fertile. <strong>The</strong>se heterozygous mutants show dominant effects on DA and BH4 where Pu mutants<br />

have low DA and BH4 levels while ple mutants have only low DA levels (Chaudhuri et al.,<br />

2007).<br />

Catsup is another important regulator <strong>of</strong> the DA and BH4 biosynthesis pathways, acting<br />

as a negative regulator <strong>of</strong> both GTPCH and TH. <strong>The</strong> Catsup protein contains seven predicted<br />

transmembrane domains and interacts with both GTPCH and TH (<strong>The</strong> O’Donnell Lab,<br />

unpublished observations). <strong>The</strong> loss <strong>of</strong> function mutants <strong>of</strong> Catsup are homozygous lethal;<br />

however, heterozygous mutants show elevated DA (Stathakis et al., 1999).<br />

5


Catsup<br />

Tyrosine<br />

Tyrosine hydroxylase Tyramine decarboxylase<br />

L-DOPA BH4<br />

Tyramine<br />

Dopa decarboxylase Tyramine β Hydroxylase<br />

Dopamine Octapamine<br />

Figure 1.1: Dopamine Biosynthesis Pathway in Drosophila. Tyrosine acts as a precursor to two<br />

distinct pathways in Drosophila. <strong>The</strong> hydroxylation <strong>of</strong> tyrosine yields DA as the end product<br />

where L-DOPA acts as an intermediate product. Catecholamines up (Catsup) acts as a negative<br />

regulator <strong>of</strong> TH whereas tetrahydrobiopterin (BH4) functions as a c<strong>of</strong>actor for TH. Octopamine<br />

is generated from tyramine, which in turn, is obtained from decarboxylation <strong>of</strong> tyrosine.<br />

6


GTP<br />

GTP<br />

Cyclohydrolase<br />

(GTPCH)<br />

Dihydroneopterin<br />

triphosphate<br />

6-pyruvoyl<br />

tetrahydropterin<br />

synthase<br />

Figure 1.2: <strong>The</strong> BH4 biosynthesis pathway. GTPCH, the rate limiting enzyme converts GTP to<br />

dihydroneopterin triphosphate which is then dephosphorylated and reduced to 6- pyruvoyl<br />

tetrahydropterin (PTP) via PTP synthase. PTP is reduced to tetrahydrobiopterin (BH4) by<br />

sepiaterin reductase.<br />

7<br />

6-Pyruvoyl<br />

hydropterin<br />

Sepiaterin<br />

reductase<br />

Tetra-<br />

hydro-<br />

biopterin


Functions <strong>of</strong> DA in Drosophila<br />

In the adult Drosophila brain, there are 200 TH positive neurons, and expression <strong>of</strong> DA<br />

has been detected in the nerve terminals present in mushroom body, the center for memory and<br />

learning in flies (Neckameyer, 1998b). Cell bodies are present in specific clusters and their<br />

nomenclature is based on the region <strong>of</strong> brain where they localize (Fig. 1.3 A, B). <strong>The</strong>se DA<br />

neurons can be visualized by using anti-TH antibody or by driving the expression <strong>of</strong> Green<br />

Fluorescent Protein (GFP) in TH neurons using the GAL4-UAS system (Brand and Perrimon,<br />

1993, Friggi-Grelin et al., 2003).<br />

<strong>The</strong> functions <strong>of</strong> DA in Drosophila will be discussed here considering that DA mediates<br />

similar functions in invertebrates and vertebrates. In Drosophila, the level <strong>of</strong> DA controls<br />

locomotion both in adult and larvae (Neckameyer, 1996; Cooper and Neckameyer, 1999). In<br />

addition, depletion <strong>of</strong> DA levels results in defective learning in young males as assayed by the<br />

learned ability to distinguish between mature and immature females for courtship (Neckameyer,<br />

1998a). However, certain behavior features such as larval phototaxis, salt aversion, and heptanol<br />

preference were not altered by depletion <strong>of</strong> DA in larvae (Neckameyer, 1996).<br />

<strong>The</strong> loss <strong>of</strong> function mutations in the gene encoding TH, ple, are homozygous lethal.<br />

Homozygous ple mutants lack melanin, derived from catechols and die at late embryogenesis<br />

(Neckameyer and White, 1993). Pharmacological depletion <strong>of</strong> DA via DA inhibitor, 3-iodo-<br />

tyrosine (3-IT) or via loss-<strong>of</strong>-function mutations leads to defective ovarian development. DA<br />

depleted larvae also show developmental delay and decreased fertility suggesting an important<br />

role <strong>of</strong> DA during oogensis, embryogenesis and larval development (Neckameyer, 1996).<br />

Further, DA and L-DOPA function in cross-linking <strong>of</strong> cuticle proteins during cuticle formation<br />

8


(Wright, 1987). DA is also implicated in regulation <strong>of</strong> sleep cycle via DAT (Kume et al., 2005)<br />

and in responses to drugs such as amphetamine and cocaine (Pörzgen et al., 2001).<br />

9


A B<br />

Figure 1.3: <strong>The</strong> schematic diagram showing the location and number <strong>of</strong> DA neurons in adult<br />

Drosophila brain viewed from anterior and posterior aspects. (A) <strong>The</strong> anterior aspect <strong>of</strong> the brain<br />

consists <strong>of</strong> two subgroups, PAL (protocerebral anterolateral) and PAM (protocerebral<br />

anteromedial). (B) <strong>The</strong> posterior aspect consists <strong>of</strong> five subgroups, PPM1 (unpaired), PPM2<br />

(paired), PPM3 (paired) (protocerebral posterior medial); PPL1 and PPL2 (paired) (protocerebral<br />

posterolateral).<br />

10


Parkinson’s disease<br />

Parkinson’s disease (PD) is the second most common chronic neurodegenerative disease<br />

and the most common movement disorder (Dauer and Przedborski, 2003; Mayeux, 2003). PD is<br />

characterized by the progressive loss <strong>of</strong> dopaminergic neurons in the substantia nigra pars<br />

compacta (SNpc) along with a decrease in DA levels in their terminals within the dorsal striatum<br />

(Hornykiewicz and Kish, 1987). In addition, PD is associated with neuronal damage in regions<br />

<strong>of</strong> the brain stem such as the locus coeruleus, raphe nuclei, and the nucleus basalis <strong>of</strong> Meynert<br />

(Fig. 1.4). This progressive neurodegeneration <strong>of</strong> the nigrostriatal pathway causes the clinical<br />

manifestations <strong>of</strong> PD such as rigidity, resting tremor (a rhythmic involuntary 5–7 Hz tremor in<br />

patients at rest), slowness <strong>of</strong> voluntary movement, postural instability, and in some cases,<br />

dementia (Jellinger, 2001; Savitt et al., 2006).<br />

PD is an age-related neurodegenerative disease, mainly affecting the population over the<br />

age <strong>of</strong> 55, with an age-dependent increase in incidence. Recent studies indicate that PD afflicts<br />

about 3% <strong>of</strong> people over 65 years and 4–5% <strong>of</strong> people over 85 years <strong>of</strong> age; however, 5–10% <strong>of</strong><br />

PD patients are less than 40 years <strong>of</strong> age (Whitton, 2007). PD appears in two forms: familial and<br />

sporadic. <strong>The</strong> familial form <strong>of</strong> PD contributes to only 5% <strong>of</strong> PD cases and has been linked to<br />

mutations in several genes such as α-synuclein (Spillantini et al., 1997), parkin (Kitada et al.,<br />

1998), DJ1 (Bonifati et al., 2003), PINK1 (Velente et al., 2004), UCHL1 (Das et al., 1998),<br />

Omi/Htra2 (Strauss et al., 2005), and LRRK2 (Paisán-Ruíz et al., 2004). <strong>The</strong> remaining 95% <strong>of</strong><br />

PD cases are attributed to idiopathic causes with possible contributions from mitochondrial<br />

dysfunction, oxidative stress and protein aggregation (Zhou et al., 2008). In addition, recently the<br />

neuroinflammatory process has been recognized as an important factor in the onset as well as<br />

progression <strong>of</strong> PD (Mosley et al., 2006).<br />

11


Parkinson’s disease and oxidative stress<br />

Oxidative stress has been considered as a major contributor for the initiation <strong>of</strong> DA<br />

neuron loss in SN. It could be due to decreased endogenous anti-oxidant capacity and/or<br />

excessive production <strong>of</strong> reactive oxygen species (ROS), reactive oxygen intermediates (ROI) and<br />

reactive nitrogen species (RNS) in cells. <strong>The</strong> brain consumes almost 20% <strong>of</strong> the total oxygen in<br />

the body and is relatively sparse in its anti-oxidant defenses such as catalase, superoxide<br />

dismutase, glutathione, and glutathione peroxidase (Floyd, 1999). Especially, the higher<br />

metabolic rate but relatively lower content <strong>of</strong> anti-oxidant capacity <strong>of</strong> SN than other regions <strong>of</strong><br />

the brain renders it highly sensitive to the effects <strong>of</strong> ROS/ROI/RNS even in healthy individuals<br />

(Marshall et al., 1999).<br />

<strong>The</strong> “oxidative stress hypothesis” has been further supported by many experimental PD<br />

models generated using various neurotoxins such as rotenone, 1-methyl-4-phenyl-1,2,3,6-<br />

tetrahydropyridine (MPTP), 6-hydroxydopamine (6-OHDA) and N,N'-dimethyl-4,4'-<br />

bipyridinium dichloride (paraquat) (PQ), which recapitulate many <strong>of</strong> the pathological features <strong>of</strong><br />

PD (Mendez and Finn, 1975; Burns et al., 1983; Betarbet et al., 2000; McCormack et al., 2002).<br />

MPTP and rotenone are well-known mitochondrial toxins. MPTP is highly lipophilic and after<br />

crossing the blood brain barrier is converted into 1-methyl-4-phenylpyridinium (MPP+) in the<br />

presence <strong>of</strong> monoamine oxidase B in the glia and serotonergic neurons in the central nervous<br />

system (Heikkila et al., 1984). Once formed, MPP+ is released extracellularly and then<br />

internalized in the dopaminergic neurons via DAT (Javitch et al., 1985). Both MPP+ and<br />

rotenone can accumulate within the mitochondria, bind to complex I <strong>of</strong> the electron transport<br />

12


Figure 1.4: <strong>The</strong> sagittal section <strong>of</strong> human brain showing major regions <strong>of</strong> the human brain. <strong>The</strong><br />

cell bodies <strong>of</strong> dopaminergic neurons, targeted in PD, are located in Substantia nigra (SN), which<br />

is situated in the midbrain region <strong>of</strong> the brainstem. (Image source:<br />

http://content.answers.com/main/content/img/McGrawHill/Encyclopedia/images/CE093200FG0<br />

010.gif )<br />

13


chain (Nicklas et al., 1985) and subsequently lead to deficient formation <strong>of</strong> mitochondrial ATP<br />

and accumulation <strong>of</strong> ROS, such as superoxide, in mitochondria (Cleeter et al., 1992). Superoxide<br />

radical (O2 - ) is converted into hydrogen peroxide, H2O2, initiating the process <strong>of</strong> cellular<br />

destruction inside as well as outside the mitochondria. On the other hand, after entering into the<br />

brain, oxidation <strong>of</strong> 6-OHDA in the presence <strong>of</strong> iron or copper in the intraneuronal and<br />

extraneuronal regions, generates para-quinone, H2O2, superoxide and hydroxyl radicals (Saner<br />

and Thoenen, 1972). H2O2, superoxide and hydroxyl radicals bring about the oxidation <strong>of</strong><br />

cellular organelle leading to cell death. Para-quinone causes depletion <strong>of</strong> vital antioxidants such<br />

as glutathione, inactivation <strong>of</strong> critical enzymes such as catechol-O-methyltransferase and<br />

tyrosine hydroxylase, which are required for dopamine synthesis (Borchardt et al., 1976; Kuhn et<br />

al., 1999).<br />

PQ or N,N'-dimethyl-4-4'-bipyridinium ion is comprised <strong>of</strong> two pyridine<br />

rings (aromatic rings in which one carbon atom is replaced by a nitrogen atom) joined covalently<br />

to each other and possessing methyl groups attached to both nitrogens (Fig. 1.5). PQ undergoes<br />

a single electron, reduction-oxidation cycling with subsequent formation <strong>of</strong> superoxide radicals<br />

(O2 − ) (Bus et al., 1974). This process occurs in two steps: the first step is single-electron<br />

reduction <strong>of</strong> PQ, which occurs in the presence <strong>of</strong> diaphorases. PQ diaphorases are<br />

oxidoreductase enzymes which use NADPH as a source <strong>of</strong> electron(s) to donate to PQ,<br />

generating a PQ radical (PQ .+ ) (Dicker and Cederbaum, 1991). Recently, nitric oxide synthase<br />

(NOS) has been shown to be one <strong>of</strong> the diaphorases capable <strong>of</strong> reacting with PQ (Day et al.,<br />

1999). In the second step, oxidation <strong>of</strong> the PQ .+ obtained from the first step by molecular oxygen<br />

takes place, yielding oxidized PQ or PQ and O2 − (Fig. 1.6). <strong>The</strong> PQ radical (PQ .+ ) is a powerful<br />

reducing radical, capable not only <strong>of</strong> reacting with molecular oxygen to generate superoxide<br />

14


adicals, but also <strong>of</strong> reacting with transitional metals such as iron and thus generates hydroxyl<br />

radicals via the Fenton reaction as shown in Fig.1.7. Intrastriatal, intraperitoneal and intravenous<br />

injections <strong>of</strong> PQ in the various animal models <strong>of</strong> PD have demonstrated loss <strong>of</strong> dopaminergic<br />

neurons, possibly due to increased oxidation <strong>of</strong> DA to a greater extent than other subpopulation<br />

<strong>of</strong> neurons (Brooks et al., 1999; McCormack et al., 2002). Aside from oxidative stress, PQ<br />

indirectly brings about depletion <strong>of</strong> the intracellular stores <strong>of</strong> NADPH and NADH, and impairs<br />

vital metabolic pathways such as fatty acid synthesis, inhibition <strong>of</strong> fatty acid synthesis and<br />

increased oxidation <strong>of</strong> glucose by the pentose phosphate shunt all culminating in death <strong>of</strong> the<br />

cells (Fisher and Reicherter, 1984).<br />

15


Figure 1.5: <strong>The</strong> chemical structure <strong>of</strong> paraquat (PQ). PQ is comprised <strong>of</strong> two pyridine rings<br />

(aromatic rings in which one carbon atom is replaced by a nitrogen atom) joined covalently to<br />

each other and contain attached methyl groups to both nitrogen. (Image source: Przedborski and<br />

Ischiropolous, 2005).<br />

Figure 1.6: <strong>The</strong> two step mechanism <strong>of</strong> PQ toxicity. In the first step, PQ uses NADPH as a<br />

source <strong>of</strong> electron(s) to donate to PQ, generating PQ radical (PQ .+ ). In the second step, oxidation<br />

<strong>of</strong> the PQ .+ obtained from the first step by molecular oxygen takes place, yielding oxidized PQ or<br />

PQ and superoxide radical (O2 − ). (Image source: Przedborski and Ischiropolous, 2005).<br />

Figure 1.7: <strong>The</strong> Fenton reaction. PQ radical generated after reacting with NADPH also reacts<br />

with transitional metals such as iron and thus generates hydroxyl radicals. (Image source:<br />

Przedborski and Ischiropolous, 2005).<br />

16


Parkinson’s disease and Neuroinflammation<br />

Neuroinflammation is proposed to play a crucial role in neurodegenerative. Neuro-<br />

inflammation can be broadly defined as the activation <strong>of</strong> immune cells such as microglia within<br />

the central nervous system in response to non-self agents or abnormal environment. In general,<br />

this response includes production <strong>of</strong> inflammatory mediators from neuronal and non-neuronal<br />

cells such a microglia, astrocytes. <strong>The</strong> initial study by McGeer et al. (1988) demonstrated the up-<br />

regulation <strong>of</strong> Major Histocompatibility Complex (MHC) molecules in PD patients, along with an<br />

increase in activated microglial cells, the resident immune cells <strong>of</strong> the brain, in the substantia<br />

nigra (SN). Elevated levels <strong>of</strong> proinflammatory cytokines such as tumor necrosis factor-alpha<br />

(TNF-α), interleukin (IL)-1beta and IL-6 in the cerebrospinal fluid and striatum in PD brains<br />

(Mogi et al., 1994; Muller et al., 1998). Further, up-regulation <strong>of</strong> inducible nitric oxide synthase<br />

(iNOS) and cyclooxygenase 2 (COX-2) was found in the SN <strong>of</strong> PD patients but not in control<br />

individuals (Knott et al., 2000). Enhanced expression <strong>of</strong> IL-1, IL-6 and TNF-α has also been<br />

shown in the cerebrospinal fluid as well as in the basal ganglia <strong>of</strong> PD patients (Nagatsu and<br />

Sawada, 2006). In order to understand the mechanisms <strong>of</strong> neuroinflammation, several animal<br />

models have been generated using MPTP, rotenone and 6-OHDA (Czlonkowska et al., 1996;<br />

Ciccetti et al., 2002; Gao et al., 2002) in which activated microglia have been observed.<br />

Increased pro-inflammatory cytokines/chemokines such as TNF-α, IL-1, IL-6 and enzymes such<br />

as COX-2 and NO are proposed to be toxic to dopaminergic neurons (Liberatore et al., 1999;<br />

Block et al., 2007). Further, the injured neurons recruit more microglia and astrocytes and<br />

influence the release <strong>of</strong> pro-inflammatory cytokines causing further activation <strong>of</strong> microglia (Le et<br />

al., 2001). <strong>The</strong>refore, neuroinflammatory response has been considered to amplify and/or<br />

accelerate the loss <strong>of</strong> DA neurons in PD. However, the mechanisms for neuroinflammatory<br />

17


esponses that exaggerate neurotoxicity are poorly understood. Moreover, the studies to date that<br />

investigate neuroinflammatory mechanisms have mostly utilized in vitro approaches. Further, it<br />

is difficult to identify the genes capable <strong>of</strong> modifying the neuroinflammatory response in<br />

mammalian studies. We have established a Drosophila PD model based on ingestion <strong>of</strong> the<br />

herbicide paraquat, which recapitulates most behavioral and patho-physiological features <strong>of</strong> PD,<br />

including loss <strong>of</strong> dopaminergic neurons (Chaudhuri et al., 2007). Data presented in Chapter 2<br />

demonstrates that paraquat ingestion induces a dramatic and rapid activation <strong>of</strong> nitric oxide<br />

synthase (NOS) and a corresponding elevation <strong>of</strong> NO production in adult Drosophila brain.<br />

Since the deleterious effects <strong>of</strong> paraquat are partially rescued by a NOS inhibitor, the data<br />

suggest that NO plays an important role in PQ toxicity in Drosophila. <strong>The</strong>se data set the<br />

preliminary results for further exploring the neuroinflammatory response in adult Drosophila<br />

brain.<br />

Glia in mammals and Drosophila<br />

In mammals, glial cells are non-neuronal cells constituting about 50 % <strong>of</strong> the volume <strong>of</strong><br />

the CNS where the glia to neuron ratio in CNS is about 10:1. Glia are also found in the<br />

peripheral nervous system. Mammalian glia play a key role in support and nutrition to CNS<br />

neurons, formation <strong>of</strong> myelin sheath and in signal transmission in the CNS. In addition, they also<br />

perform crucial developmental roles by guiding migration <strong>of</strong> neurons and generating molecules<br />

supporting the growth <strong>of</strong> axons and dendrites (Freeman and Doherty, 2006).<br />

Drosophila is emerging as an excellent model to study the molecular and functional and<br />

developmental aspects <strong>of</strong> glia. In Drosophila, there are mainly four types <strong>of</strong> glia, namely,<br />

cortex, neuropil, surface, and peripheral glia. Cortex glia, functionally homologous to vertebrate<br />

18


astrocytes, remain in close contact with neurons during the development <strong>of</strong> the brain (Ito et al.,<br />

1995; Pereanu et al., 2005). Neuropil glia, similar to vertebrate oligodendrocytes, provide<br />

insulation to the axons associated with larval and adult neurons and trophic support to the<br />

developing neurons (Booth et al., 2000). Peripheral glia ensheath and support the peripheral<br />

nerves mediating motor and sensory circuitry peripherally, like Schwann cells in vertebrates<br />

(Leiserson et al., 2000). However, there has been no indication <strong>of</strong> cells acting as a functional<br />

homolog <strong>of</strong> vertebrate microglial cells (Freeman and Doherty, 2006). <strong>The</strong> functions performed<br />

by vertebrate and Drosophila glia are summarized in Table 1.1.<br />

In Drosophila, almost all glial cells are derived from neuroectoderm, the peripheral<br />

ectoderm present lateral to the ventral midline (Ito et al., 1995). <strong>The</strong> longitudinal glia<br />

ensheathing the longitudinal axons reside along the ventral nerve cord. <strong>The</strong>se lateral glia express<br />

the glial cells missing (gmc) transcription factor, which regulates glial cell differentiation; gmc<br />

positive cells become glia while gmc negative cells become neurons. When gmc is ectopically<br />

expressed, neurons transform into glia (Hosoya et al., 1995). In addition to differentiation <strong>of</strong> glial<br />

cells, gmc together with its homolog, glial cells missing 2 (gmc2) is essential for the proper<br />

differentiation <strong>of</strong> hemocytes (Alfonso and Jones, 2002). It is to be noted that gmc expression is<br />

required for the early differentiation <strong>of</strong> glial cells, and terminal differentiation and maintenance<br />

<strong>of</strong> glial cell fate is carried out by other genes. Among these fate-determing genes, reverse<br />

polarity (repo), pointed (ppt) and tramtrack (ttk) are well-characterized (Klaes et al., 1994;<br />

Halter et al., 1995; Giesen et al., 1997). repo is a homeodomain transcription factor expressed in<br />

nearly all glial cells and mutation in the repo gene results in glial cell defects at later stages<br />

during embryonic development. <strong>The</strong> expression <strong>of</strong> repo and ppt follows the expression <strong>of</strong> gmc in<br />

different cell contexts.<br />

19


In PD, glial cell dysfunction has been reported. Microglia are mainly associated with the<br />

neuroinflammatory process, but other glial cells have also been known to function in the<br />

pathogenesis <strong>of</strong> PD (McGeer and McGeer, 2008). In mammalian PD models, astrocytes show<br />

reactive morphology and function to isolate the pathological site being attacked by microglia,<br />

thereby preventing microglial attack to the normal surrounding area. <strong>The</strong>y are also shown to<br />

secrete neurotrophic factors for DA neurons such as glial cell-line-derived-neurotrophic factor<br />

(GDNF) and brain-derived neurotrophic factors (BDNF) (Lin et al., 1993; Knott et al., 2002;<br />

Chen et al., 2006). Some studies have, however, reported that activation <strong>of</strong> astrocytes is<br />

associated with amplification <strong>of</strong> the inflammatory reaction through expression <strong>of</strong> chemokines<br />

and adhesion molecules (Miklossy et al., 2006; Ubogu et al., 2006). On the other hand, only a<br />

few studies have elucidated the functional role <strong>of</strong> the oligodendrocyte. Overall, these studies<br />

suggest the activation <strong>of</strong> oligodendrocytes in the SN <strong>of</strong> PD cases (Yamada et al., 1991; Yamada<br />

et al., 1992).<br />

20


Glial cell type in<br />

vertebrates/Drosophila<br />

Function Distribution in CNS<br />

Astrocytes/Cortex glia Trophic support <strong>of</strong> CNS cell cortex,<br />

neurons<br />

Synapse transmission<br />

synapse, CNS surface<br />

Oligodendrocytes/Neurophil glia Trophic support <strong>of</strong> Ensheathment <strong>of</strong> axons<br />

neurons,<br />

myelination<br />

<strong>of</strong> CNS<br />

Microglia/?? Immune response<br />

Macrophage function<br />

All around the CNS<br />

Schwann cells/peripheral glia Support <strong>of</strong> peripheral Ensheathment <strong>of</strong><br />

nerves and their<br />

myelination<br />

peripheral nerves<br />

Table 1.1: Similarity in the function and distribution <strong>of</strong> vertebrate and Drosophila glia in CNS.<br />

Drosophila is thought to have functional homologs for all types <strong>of</strong> vertebrate glia, except for<br />

microglia. (<strong>The</strong> table is presented as modified version <strong>of</strong> table published in Freeman and<br />

Doherty, 2006).<br />

21


Microglia<br />

Microglia are bone marrow derived macrophage-lineage cells which enter into the brain<br />

during embryogenesis and comprise approximately 12 % <strong>of</strong> the cells in human brain (Kaur et al.,<br />

2001). Microglia function as resident immune surveillance cells in the CNS. Cajal (1931)<br />

defined the cells in the brain: neurons as the first element, astrocytes as the second element and<br />

microglia as the third element. Rio-Hortega (1932) subsequently performed a systemic<br />

investigation <strong>of</strong> microglial cells. In the normal brain, microglia, are in a resting stage, with cell<br />

bodies having only a few fine ramified processes and low expression <strong>of</strong> surface antigens<br />

(Lawson et al., 1990). In pathological conditions or during invasion <strong>of</strong> pathogens, microglia<br />

become activated with morphological changes evident by an increase in cell body area and<br />

ramifying processes exhibiting amoeboid and spider-like appearance (Fig. 1.8). In addition, once<br />

activated, microglia increase in number and up-regulation <strong>of</strong> surface markers such as CD14,<br />

Major Histocompatibility Complex (MHC) molecules, chemokine receptors, pro-inflammatory<br />

cytokines, interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), interleukin 1-β (IL-1β), and<br />

enzymes such as inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX) 1 and 2<br />

occurs (McGeer et al., 1988; Mirza et al., 2001; Rock et al., 2004; Nimmerjahn et al., 2005). It is<br />

well known that activation <strong>of</strong> macrophages/microglia against invading microbes and other<br />

harmful pathogens serves as the first line <strong>of</strong> defense to the host organisms and serves as an<br />

essential function for the survival <strong>of</strong> organisms. It is proposed that mild to moderate activation <strong>of</strong><br />

microglia performs a homeostatic, neurotrophic and thus neuroprotective role in neuropathology<br />

(Batchelor et al., 1999; Batchelor et al., 2002), but sustained activation <strong>of</strong> microglia actively<br />

participates in the self-perpetuating inflammatory processes linked to neurodegeneration in<br />

various neurodegenerative and inflammatory diseases. <strong>The</strong>refore, the role <strong>of</strong> microglial cells in<br />

22


the pathogenesis <strong>of</strong> chronic neurodegenerative diseases including PD, discussed above, suggests<br />

that maintenance <strong>of</strong> the delicate balance for the microglial beneficial and deleterious functions is<br />

crucial (Wyss-Coray and Mucke, 2002; McGeer and McGeer, 2004) (Fig. 1.8). However, the<br />

stimulus causing the alteration in such balance is still not well understood. In Chapter 2,<br />

identification <strong>of</strong> NOS expressing structures in adult Drosophila brain in response to PQ ingestion<br />

have been identified which may appear analogous to NOS expressing mammalian microglia.<br />

23


Physiological condition<br />

(supporting regulatory immune<br />

and homeostatic functions)<br />

Microglia<br />

Figure 1.8: An outcome <strong>of</strong> the sustained activation <strong>of</strong> microglia in chronic neurodegenerative<br />

disease. Microglia activation is associated with a delicate balance <strong>of</strong> physiological and<br />

pathological conditions. In chronic neurodegenerative diseases, sustained activation <strong>of</strong> microglia<br />

leads to destruction <strong>of</strong> diseased as well and as healthy neurons and thus amplification and<br />

acceleration <strong>of</strong> neurodegenerative process. <strong>The</strong> morphology <strong>of</strong> becomes reactive one from<br />

resting stage as shown in figure. (Image source for images showing microglial morphology:<br />

Mosley et al., 2006).<br />

24<br />

Pathological condition<br />

(reactive to destructive role)<br />

-Excessive stimulation <strong>of</strong><br />

microglia<br />

-Up-regulation <strong>of</strong> microglial<br />

markers<br />

-Destruction <strong>of</strong> diseased and<br />

Resting microglia Reactive microglia


Neuroinflammation in experimental models <strong>of</strong> PD<br />

MPTP is a potent neurotoxin known to induce depletion <strong>of</strong> dopaminergic neurons in<br />

humans (Langston et al., 1983). Examination <strong>of</strong> post-mortem brains <strong>of</strong> patients 3-16 years after<br />

exposure to MPTP for relatively short period and with symptoms <strong>of</strong> PD showed pathological<br />

features <strong>of</strong> neurodegeneration and activated microglia (Langston et al., 1999). Similarly, 6-<br />

OHDA exposure in mice induces the proinflammatory cytokine TNF-α in the striatum and CSF<br />

<strong>of</strong> rats (Mogi et al., 1994). Signs <strong>of</strong> inflammation remained one month after 6-OHDA exposure<br />

as evaluated by increased expression levels <strong>of</strong> mRNA for IL-1α and IL-1β in lesioned tissues<br />

(Depino et al., 2003). Rotenone induced increased expression <strong>of</strong> Mac-1-positive reactive<br />

microglia in the striatum and SN, even in the absence <strong>of</strong> DA neuron loss (Sherer et al., 2003). PQ<br />

toxicity has been reported to cause an increase in cytokines, IL-1β, IL-6, IL-10 and TNF-α in<br />

serum <strong>of</strong> rats; however, these changes are associated with a high dose (120 mg/kg) (Jian et al.,<br />

2007). Similarly, acute poisoning with lethal and non-lethal doses <strong>of</strong> PQ involeved up-regulation<br />

<strong>of</strong> iNOS and IL-1β, respectively in rats (Tomita et al., 1999). In in vitro studies, PQ has been<br />

shown to generate excess superoxide radicals via NOS and NADPH oxidase enzymes in<br />

microglia and thereby proposed to mediate PQ induced oxidative damage to nearby neurons<br />

including DA neurons (Bonneh-Barkay et al., 2005; Wu et al., 2005). Exposure to relatively low<br />

dose (10 mg/kg) <strong>of</strong> PQ exposure also causes an increase in Mac-1 immunoreactive cells in in<br />

vivo mammalian model (Purisai et al., 2007).<br />

Inhibition <strong>of</strong> neuroinflammation in PD models<br />

Genetic and pharmacological inhibition <strong>of</strong> microglia has been shown to decrease the<br />

induction <strong>of</strong> microglia by MPTP, 6-OHDA and lipopolysaccharide (LPS). Although these<br />

25


studies have targeted is<strong>of</strong>orms <strong>of</strong> NOS and/or cytokines known to be up-regulated in various<br />

toxin-induced PD models, the results are somewhat controversial since these approaches may not<br />

produce complete inhibition <strong>of</strong> activated microglia or prevention <strong>of</strong> DA neuron loss. Especially,<br />

although activation <strong>of</strong> iNOS is mainly reported with reactive microgliosis in PD models,<br />

suppression <strong>of</strong> microglial activation has not been achieved with elimination iNOS genetically<br />

(Dehmer et al., 2000; Iravani et al., 2002). More recently, inhibition <strong>of</strong> nNOS has been reported<br />

to impart protection against MPTP induced mouse PD model (Watanabe et al., 2008).<br />

<strong>The</strong> daily intake <strong>of</strong> Non-Steroidal Anti-inflammatory Drugs (NSAIDs) has been shown to<br />

reduce the risk <strong>of</strong> PD in epidemiological studies (Chen et al., 2003). Inhibition <strong>of</strong> COX-2, the<br />

rate-limiting enzyme in prostaglandin E2 synthesis markedly diminishes dopaminergic<br />

neurodegeneration along the nigrostriatal axis when exposed to MPTP and 6-OHDA (Teismann<br />

et al., 2001; Sanchez-Pernaute et al., 2004).<br />

Minocycline, a second-generation semi-synthetic tetracycline derivative has been shown<br />

to act on multiple components involved with neuroinflammatory process in chronic<br />

neurodegenerative and neuronal injury. It has been shown to down-regulate iNOS, inhibit NOS-<br />

mediated phosphorylation <strong>of</strong> p38 mitogen-activated protein kinase (MAPKs), and reduce IL-1β<br />

converting enzyme (ICE) and IL-1β production, to decrease MPTP induced increase in IL-1 α,<br />

IL-6 and other microglial markers (Yrjänheikki et al., 1999; Lin et al., 2001; Wu et al., 2002;<br />

Sriram et al., 2006). For these properties, minocycline is currently being tested in Phase III<br />

clinical trials for PD. Minocycline has been used in many studies as an agent <strong>of</strong> microglial<br />

inhibition in mammalian models for PD (Peng et al., 2006; Purisai et al., 2007) and has been<br />

shown to increase the survival <strong>of</strong> PQ-fed flies (Bonilla et al., 2006). However, neuroprotective<br />

and anti-inflammatory roles for minocycline in flies have never been studied in detail.<br />

26


Identification <strong>of</strong> neuroprotective and anti-inflammatory functions <strong>of</strong> minocycline in a well<br />

known in vivo model that can be genetically manipulated, such as Drosophila melanogaster,<br />

could define a model that could be used to identify genes modifying this response and to identify<br />

signaling pathways that mediate this inflammatory response. We have demonstrated that<br />

minocycline is capable <strong>of</strong> suppressing NOS activity in adult Drosophila brain. In chapter three,<br />

we have further identified the neuroprotective properties <strong>of</strong> minocycline and presented the<br />

preliminary results for the signaling pathways mediating the PQ-induced DA toxicity in this in<br />

vivo model.<br />

Nitric oxide<br />

Nitric oxide is a gas that functions as a second messenger molecule in various metabolic<br />

processes (Bredt and Snyder, 1994; Murad, 1998). NO is generated when L-arginine is converted<br />

into citrulline via nitric oxide synthase (NOS) in the presence <strong>of</strong> molecular oxygen and NADPH.<br />

NOS, which is active as a dimer, consists <strong>of</strong> an oxygenase (catalytic) domain and a reductase<br />

domain. <strong>The</strong> catalytic domain <strong>of</strong> NOS (NOSox) contains a heme group and BH4, and binds the<br />

substrate, L-arginine (Mayer, 1994). <strong>The</strong> c<strong>of</strong>actors, flavin mononucleotide (FMN), flavin<br />

adenine dinucleotide (FAD) and NADPH bind to the reductase domain <strong>of</strong> the enzyme (NOSred)<br />

(Marletta, 1989; Bredt et al., 1991). Ca 2+ /calmodulin acts to promote electron transport between<br />

the NOSred and NOSox (Crane et al., 1999). NO generation involves a two-stage reaction via<br />

formation <strong>of</strong> hydroxyarginine resulting in the oxidation <strong>of</strong> the guanadino nitrogen group <strong>of</strong><br />

arginine, to form citrulline (Fig. 1.9). Three is<strong>of</strong>orms <strong>of</strong> NOS are encoded by separate genes,<br />

NOS1, NOS2 and NOS3, in vertebrate genomes. <strong>The</strong>se genes are further classified according to<br />

27


the tissue <strong>of</strong> origin: nNOS (neuronal NOS; encoded by NOS1); iNOS (inducible, macrophage<br />

NOS; encoded by NOS2), and eNOS (endothelial NOS; encoded by NOS3).<br />

In vertebrates, the function <strong>of</strong> NOS was recognized first in the maintenance <strong>of</strong> the<br />

vasodilation, where the absence <strong>of</strong> NO signaling was implicated in the erectile dysfunction in<br />

males (Burnett, 1995). Subsequently, various studies performed using biochemical, physiological<br />

and transgenic approaches in vertebrates, identified diverse physiological roles <strong>of</strong> NO. It is<br />

known that NO signaling is important in learning and memory (Hölscher, 1997), reproduction<br />

(Rosselli et al., 1998), and in host defense mechanisms (Liew et al., 1999). Activation <strong>of</strong> NO<br />

signaling is mediated by diverse stimuli and by rapid diffusion <strong>of</strong> the NO molecule across<br />

cellular membrane (Davies SA, 2000).<br />

28


Nitric oxide<br />

synthase<br />

Figure 1.9: A schematic diagram showing the components involved in the formation <strong>of</strong> nitric<br />

oxide (NO). NO is generated along with Citrulline from L-Arginine and molecular oxygen. <strong>The</strong><br />

catalytic domain <strong>of</strong> NOS (NOSox) contains a heme group and BH4, and binds the substrate, Larginine.<br />

<strong>The</strong> c<strong>of</strong>actors, FMN, FAD and NADPH binds to the reductase domain NOS.<br />

Ca 2+ /calmodulin promotes electron transport between the NOSred and NOSox. (Image source:<br />

modification <strong>of</strong> figure published in Davies SA, 2000).<br />

29<br />

NADPH


NO signaling in invertebrates<br />

Many comparative physiological and biochemical studies have presented strong evidence<br />

that NO signaling is evolutionarily and functionally conserved. Several insect NOS genes have<br />

been cloned, including those from Drosophila melanogaster (Regulski and Tully, 1995) and<br />

Anopheles stephensi (Luckhart and Rosenberg, 1999). Insect NOS, including Drosophila NOS,<br />

shows highest sequence similarity to NOS1 (49%), as compared with NOS2 (44% identity) and<br />

NOS3 (47% identity) (Luckhart and Rosenberg, 1999; Stasiv et al., 2001). <strong>The</strong> insect NOS<br />

protein sequence shows 100% sequence identity in regions corresponding to the c<strong>of</strong>actor binding<br />

sites for calmodulin, FAD, FMN and NADPH in vertebrate NOS. Similar to vertebrates, NO is<br />

known to function in learning and memory, axonal guidance, locomotion and olfaction in insects.<br />

Drosophila NOS and NO signaling<br />

Regulski and Tully (1995) first reported the cloning and molecular characterization <strong>of</strong> the<br />

Drosophila NOS gene. <strong>The</strong> gene consists <strong>of</strong> 19 exons extending over 34 kb <strong>of</strong> DNA, located on<br />

the second chromosome at cytological position 32B. Using in situ hybridization and quantitative<br />

RT-PCR, Stasiv et al. (2001) determined the expression <strong>of</strong> dNOS transcript during Drosophila<br />

development. dNOS transcripts were detected in lower levels in the embryonic stage but were<br />

abundantly present in the late larval and pupal stages. NO signaling in Drosophila has been<br />

shown to be involved in the regulation <strong>of</strong> cell proliferation during imaginal disc development<br />

(Kuzin et al., 1996; Kuzin et al., 2000), development <strong>of</strong> the nervous system (Truman et al., 1996;<br />

Wildemann and Bicker, 1999), retinal patterning in the optic lobe (Gibbs and Truman, 1998),<br />

epithelial fluid secretion (Dow et al., 1994), response to hypoxia (Wingrove and O’Farrel,1999;<br />

30


DiGregorio et al., 2001), behavior (Wingrove and O’Farrel, 1999), and immunity (Nappi et al.,<br />

2000).<br />

In Drosophila, different methods have been used to determine the expression pattern <strong>of</strong><br />

NOS. A universal NOS antibody has been utilized to detect all is<strong>of</strong>orms <strong>of</strong> NOS. This antibody,<br />

which recognizes conserved amino-acid sequences corresponding to amino acid residues 1113-<br />

1122 <strong>of</strong> murine iNOS and nNOS, has been used to detect Drosophila NOS in studies <strong>of</strong> visual<br />

system development and <strong>of</strong> malphigian tubules where it functions in osmoregulation and<br />

excretion (Davies, 2000; Gibbs, 2001). Similarly, the 150 kDA NOS polypeptide has been<br />

detected in malphigian tubules with western blot analysis using the universal antibody<br />

(Broderick et al., 2003). Although the expression pattern for NOS in adult brain has not been<br />

determined with this antibody, the expression <strong>of</strong> NOS in embryos and larval brain have been<br />

studied in detail using NADPH diaphorase staining, confirming its presence in the Drosophila<br />

nervous system (Wildemann and Bicker, 1999).<br />

Role <strong>of</strong> NO in Drosophila immune response<br />

Nappi and Vaas (1998) showed that wasp parasitoid L. boulardi induced production <strong>of</strong><br />

reactive oxygen intermediates (ROI) in larval hemocytes. Both hydrogen peroxide and<br />

superoxide function as important messengers capable <strong>of</strong> activating transcription factor NF-κβ<br />

(Schmidt et al., 1995). Induction <strong>of</strong> this innate immune response has been demonstrated against<br />

parasites and bacteria, but there has been no similar study with respect to a possible neurotoxin<br />

induction <strong>of</strong> the insect innate immune response that is comparable to microglial activation in<br />

adults. Nappi et al. (2000) demonstrated increased production <strong>of</strong> superoxide radical and H2O2 in<br />

immune-challenged Drosophila against the wasp parasitoid L. boulardi. <strong>The</strong>y showed that upon<br />

31


infection with wasp, D. melanogaster and D. teissieri generate reactive intermediate species <strong>of</strong><br />

oxygen and nitrogen, ROS and RNI, which constitutes an important step in induction <strong>of</strong><br />

antimicrobial peptides like Diptericin. Foley and O’Farrell (2003) used universal NOS antibody<br />

and an antibody generated against a C-terminal peptide <strong>of</strong> dNOS to detect increased expression<br />

<strong>of</strong> NOS protein in larval hemocytes, the innate immune cells <strong>of</strong> Drosophila in response to gram-<br />

negative infection. Similar results were obtained with both antibodies. <strong>The</strong> presence <strong>of</strong><br />

hemocytes in the adult Drosophila brains has been reported (Lanot et al., 2001; Holz et al.,<br />

2003). <strong>The</strong>se results indicate the role <strong>of</strong> Drosophila hemocytes in induction <strong>of</strong> NOS against<br />

infection suggesting future directions for determining whether these cells have a crucial role in<br />

the response to paraquat in adult brain.<br />

Role <strong>of</strong> signaling pathways in the pathogenesis <strong>of</strong> PD<br />

Apoptosis signaling pathway<br />

Apoptosis or programmed cell death (PCD) is an important and essential process in all<br />

multicellular organisms to prevent developmental disorders and tumorigenesis (Vaux and<br />

Korsmeyer, 1999; Yuan and Yankner, 2000). It is proposed that death signals induced by<br />

developmental and cellular damage cause activation <strong>of</strong> the cysteine proteases, caspases, which<br />

are known to play an important role in PCD including membrane blebbing and fragmentation <strong>of</strong><br />

DNA. Drosophila melanogaster has evolutionarily conserved mechanisms for PCD (Vernooy et<br />

al., 2000). Mammalian cells possess two types <strong>of</strong> PCD pathways, extrinsic and intrinsic death<br />

inducing pathways. <strong>The</strong> intrinsic pathway involves release <strong>of</strong> mitochondrial cytochrome c due to<br />

cellular stress and its binding to adaptor protein, Apaf-1, which in turn recruits pro-caspases 9<br />

and brings about autocatalytic activation (Li et al., 1997; Zou et al., 1999). <strong>The</strong> extrinsic pathway<br />

32


involves tumor necrosis factor family with activation <strong>of</strong> caspases via Fadd (Fas-associated death<br />

domain) (Ashkenazi and Dixit, 1998; Ashkenazi A, 2002). <strong>The</strong> Drosophila PCD pathway shares<br />

many <strong>of</strong> the signaling components found in mammals and provides an excellent opportunity <strong>of</strong><br />

dissecting the functions <strong>of</strong> signaling components <strong>of</strong> PCD in vivo. <strong>The</strong> extrinsic and intrinsic<br />

signaling pathways involved with mammalian apoptosis are compared with Drosophila apoptosis<br />

signaling pathway in Fig. 1.10.<br />

33


Extrinsic death pathway<br />

Tumor Necrotic Factor<br />

Receptor family<br />

FADD<br />

dFADD<br />

Diablo/Smac<br />

Rpr, hid, grim<br />

p53<br />

Dmp53<br />

Intrinsic death<br />

pathway<br />

Bcl-2<br />

Dabcl<br />

Apaf-1<br />

Dark<br />

Caspase-8<br />

Dredd<br />

Cytochrome c<br />

Initiator caspases<br />

Dronc, Dredd<br />

Figure 1.10: Comparison <strong>of</strong> mammalian and Drosophila apoptotic signaling pathways.<br />

Drosophila possesses homologues (shown in red) for the mammalian apoptosis signaling<br />

components (shown in black) (for details, see text). (Image source: Modification <strong>of</strong> the image<br />

published in Richardson and Kumar, 2002)<br />

34<br />

IAP<br />

Diap<br />

Effector caspase<br />

Drice, Caspase-3 Dcp-1<br />

PROGRAMMED<br />

CELL DEATH


PCD in Drosophila<br />

Caspases are cysteine proteases found in an inactive form, pro-caspases in cells, which<br />

upon receiving an apoptotic signal undergo proteolytic processing to generate active enzyme,<br />

caspase. Seven caspases have been identified: Dcp-1, Dredd/Dcp-2, Drice, Dronc, Decay,<br />

Strica/Dream and Damm/Daydream (Kumar and Doumanis, 2000). Out <strong>of</strong> the several PCD<br />

related genes, reaper (rpr) acts as a pro-death genes in Drosophila; the protein encoded by this<br />

gene is functionally homologous to mammalian Diablo/Smac (Direct IAP Binding protein with<br />

low pI/second mitochondria derived activator <strong>of</strong> caspases). Like the mammalian Diablo/Smac,<br />

Rpr, along with other Drosophila pro-death proteins, Hid and Grim, interacts with the IAP<br />

(Inhibitor <strong>of</strong> Apoptosis) and antagonizes the action <strong>of</strong> IAP, thereby causing PCD (Vucic et al.,<br />

1997; Vucic et al., 1998). In Drosophila, Dronc acts as an apoptosis/caspase initiator leading to<br />

activation <strong>of</strong> Drice, the executor <strong>of</strong> PCD.<br />

In PD, the death <strong>of</strong> DA neurons in SN has been proposed to occur through activation <strong>of</strong><br />

the apoptotic signaling pathway (Levy et al., <strong>2009</strong>). Several experimental models for PD have<br />

reported the involvement <strong>of</strong> up-regulation <strong>of</strong> Bax, Bak Bim and other apoptosis related genes<br />

(Biswas et al., 2005; Perier et al., 2007; Fei et al., 2008). Moreover, activation <strong>of</strong> PCD<br />

components, caspase-3 and caspase-8 in the DA neurons <strong>of</strong> SN in PD has been reported<br />

(Hartmann et al., 2001; Tatton et al., 2003). Furthermore, many <strong>of</strong> these studies have reported<br />

the reversal <strong>of</strong> apoptotic death using caspase-inhibitors suggesting the involvement <strong>of</strong> apoptosis<br />

in the pathogenesis <strong>of</strong> PD. <strong>The</strong> interaction <strong>of</strong> apoptotic signaling with other signal transduction<br />

pathways is described below.<br />

35


Mitogen activation protein kinase signaling pathway<br />

Mitogen activation protein kinases (MAPKs) are evolutionarily conserved signaling<br />

pathway components that mediate numerous fundamental cellular processes, including cell<br />

proliferation, survival, differentiation, apoptosis, motility and metabolism (Chang and Karin,<br />

2002). Various MAPK family members are activated by different stimuli and are involved in<br />

signaling by phosphorylating specific serines and threonines <strong>of</strong> target protein substrates such as<br />

protein kinases, phospholipases, transcription factors, and cytoskeletal proteins. MAPKs are a<br />

part <strong>of</strong> the phospho-relay system involving sequentially activated kinases regulated by<br />

phosphorylation (Fig 1.11). Cells receive different stimuli from their environment and depending<br />

on the stimuli, activation <strong>of</strong> downstream Mitogen-activated protein kinase kinase kinases<br />

(MKKKs) occurs. MKKKs phosphorylate and activate specific MKKs. MKKKs have distinct<br />

motifs in their sequences that selectively mediate their activation in response to different stimuli.<br />

MKKs, in turn, catalyze the phosphorylation <strong>of</strong> MAPKs. Activation <strong>of</strong> MAPK catalyzes the<br />

phosphorylation <strong>of</strong> its own substrates which are varied for the different MAPKs signaling<br />

pathways. MAPKs are regulated by MAPKs phosphatases which reverse phosphorylation and<br />

return the MAPKs to an inactive state (Johnson and Lapadat, 2002).<br />

<strong>The</strong>re are three well-characterized MAPKs signaling pathway subfamilies: extracellular<br />

signal–regulated kinases (ERK), c-Jun NH2-terminal kinases (JNK) and p38 enzymes.<br />

Activation <strong>of</strong> these signaling pathways depends on the stimuli and the cell type in which they are<br />

activated. In general, the ERK subfamily, which includes ERK 1 and ERK 2, is involved in the<br />

regulation <strong>of</strong> meiosis, mitosis, and postmitotic functions in differentiated cells. <strong>The</strong> JNK<br />

pathway participates in stress-related signaling while p38 is activated in immune cells by<br />

inflammatory cytokines (Kyriakis and Avruch, 2001).<br />

36


JNK signaling pathway<br />

JNK signaling pathway has been well studied (Davis RJ, 2000; Johnson and Lapadat,<br />

2002). <strong>The</strong> JNK serves as phosphorylation substrates for MAP kinase kinases (MKKs) such as<br />

MKEK4/7, which are activated in turn by phosphorylation via MAP kinase kinase kinases<br />

(MKKKs) such as MKK4/7 (Fig. 1.11). It has been proposed that activation <strong>of</strong> JNK involves<br />

release <strong>of</strong> cytochrome c or Smac/DIALBO into the cytoplasm and subsequent formation <strong>of</strong> a<br />

complex with apoptosis activating factor-1 (Apaf-1) and caspase-9 causing activation <strong>of</strong><br />

executioner caspase-3 and cleavage <strong>of</strong> cellular substrates eventually (Tournier et al., 2000;<br />

Chauhan et al., 2003). In addition, other studies have suggested that the pro-apoptotic cascade<br />

induces apoptosis via a mitochondria-dependent pathway involving JNK-activated pro-apoptotic<br />

members <strong>of</strong> the Bcl-2 family, Bim and Bmf, resulting in activation <strong>of</strong> Bax and Bak (Lei et al.,<br />

2002; Lei and Davis, 2003). Furthermore, JNK activation results in phosphorylation <strong>of</strong> anti-<br />

apoptotic genes, Bcl-2 and Bcl-xL and thus induction <strong>of</strong> apoptosis (Pandey et al., 1999;<br />

Yamamoto et al., 1999).<br />

37


Figure 1.11: A schematic diagram showing the components <strong>of</strong> MAPK signaling pathway in<br />

mammals and Drosophila. ERK and JNK signaling pathways are sub-families <strong>of</strong> MAPK and<br />

follow the phospho-relay system involving sequentially activated kinases regulated by<br />

phosphorylation and results in specific biological response (for details, see text). Drosophila<br />

possesses homologues (shown in red) for the mammalian apoptosis signaling components<br />

(shown in black) for each component <strong>of</strong> ERK and JNK signaling pathways. (Image source:<br />

modification <strong>of</strong> figure from www.cellsignaling.com).<br />

38


Drosophila JNK<br />

Drosophila JNK is a homolog <strong>of</strong> mammalian JNK and is encoded by the gene basket on<br />

the left arm <strong>of</strong> the second chromosome (Nüsslein-Volhard et al., 1984). <strong>The</strong> JNK pathway<br />

includes a MAPKKKs (mekk1, slipper, wallenda/MEKK, MLK3, ASK1) (Stronach and<br />

Perrimon, 2002) and a MAPKKs (hemipterous/MKK7) (Glise et al., 1995), as well as the JNK<br />

homolog basket. Basket, upon phosphorylation by Hemipterous, activates the AP-1 transcription<br />

complex consisting <strong>of</strong> Jun and Fos, modifying target gene expression. Drosophila JNK has been<br />

reported to play crucial roles in morphogenesis and mobility, immune response, wound healing<br />

and stress response in flies.<br />

JNK signal transduction pathway and Parkinson’s disease<br />

Recently, the role <strong>of</strong> JNK in the pathogenesis <strong>of</strong> PD has been proposed. In mouse PD<br />

models in which neurodegeneration is induced by MPTP or PQ, activation <strong>of</strong> JNK via<br />

phosphorylation <strong>of</strong> c-Jun is associated with apoptosis <strong>of</strong> dopaminergic neurons (Xia et al., 2001;<br />

Saporito et al., 2003; Peng et al., 2004). Peng et al. (2004) showed that intraperitoneal injection<br />

<strong>of</strong> PQ induces an increase in superoxide and related ROS, subsequently causing activation <strong>of</strong><br />

JNK thereby triggering up-regulation <strong>of</strong> caspases-3, contributing to the apoptotic response. A<br />

similar mechanism has been shown to be involved in the 6-OHDA induced loss <strong>of</strong> dopaminergic<br />

neurons in PC cells where activation <strong>of</strong> JNK culminates in the induction <strong>of</strong> caspase-3<br />

(Rodriguez-Blanco et al., 2008). More recently, using PC 12 cell culture and primary cultured<br />

dopaminergic neurons, Klintworth et al. (2007) found that treatment <strong>of</strong> these cultures with PQ<br />

causes the stimulation <strong>of</strong> p38 and JNK signal transduction pathways and, subsequently, loss <strong>of</strong><br />

dopaminergic neurons. Although these in vitro studies support the pro-apoptotic role <strong>of</strong> JNK<br />

39


signaling pathway in PD models, the genetic redundancy resulting from the three JNK genes in<br />

mammalian genomes, JNK1, 2 and 3 and the 10 or more is<strong>of</strong>orms <strong>of</strong> JNK expressed by these<br />

genes, creates complexities in interpretations <strong>of</strong> the roles <strong>of</strong> JNK in the pathogenesis <strong>of</strong> PD.<br />

In Drosophila, functional roles <strong>of</strong> JNK/Bsk have been studied in the context <strong>of</strong> neuronal<br />

connectivity and response to oxidative and pathogenic stress. Srahna et al. (2006) demonstrated<br />

that Bsk is required for the axonal connectivity <strong>of</strong> dorsal cluster neurons between the lobula and<br />

medulla during the development <strong>of</strong> the optic lobe. Wang et al. (2003b) showed that heterozygous<br />

loss <strong>of</strong> function mutant, bsk 2 , is highly sensitive to PQ-induced oxidative stress which could be<br />

overcome by pan-neuronal over-expression <strong>of</strong> bsk employing an elav-GAL4 driver. In addition,<br />

these investigators found that JNK is essential for longevity and combating oxidative stress. Cha<br />

et al. (2005) detected the up-regulation <strong>of</strong> JNK in loss-<strong>of</strong>-function <strong>of</strong> parkin mutants. Moreover,<br />

using genetic interaction between transgenic strains for signaling components <strong>of</strong> the JNK signal<br />

transduction pathway and parkin revealed a negative relationship between JNK and parkin.<br />

<strong>The</strong>se results suggest that cell type and the context <strong>of</strong> the stimulus play an important role for<br />

anti-apoptotic and pro-apoptotic property <strong>of</strong> JNK. Mammalian JNK appears to respond to<br />

oxidative stress by triggering apoptosis in DA neurons in in vitro mammalian models, but<br />

functional roles <strong>of</strong> JNK in in vivo PD models need further elucidation.<br />

ERK pathway in mammals<br />

ERK function is mainly concerned with cellular proliferation and differentiation <strong>of</strong><br />

various cells in response to growth factors and other stimuli. In addition, recently, ERK1/2 are<br />

reported to regulate neuronal responses to both functional (learning and memory) and pathologic<br />

40


(regulated cell death) stimuli. <strong>The</strong> cascade for the ERK mediated activation <strong>of</strong> transcription<br />

factors includes phosphorylation <strong>of</strong> MAPKKK, MAPKK and MAPK as shown in Fig 1.11.<br />

ERK in Drosophila<br />

Drosophila ERK is a homolog <strong>of</strong> mammalian ERK and is encoded by the gene rolled<br />

(Lim et al., 1999). <strong>The</strong> ERK pathway includes a MAPKKK (draf-1/RAF), and a MAPKK<br />

(dsor1/MEK1/2), as well as the ERK1/2 homolog rolled. Rolled, upon phosphorylation,<br />

activates the pointed transcription factor activating target gene expression (Brunner et al., 1994).<br />

In Drosophila, rolled has been shown to mediate key roles in synaptic transmission and<br />

regulation <strong>of</strong> developmental processes such as sensory organ development.<br />

ERK signaling pathway and Parkinson’s disease<br />

<strong>The</strong>re are numerous controversial studies presenting data for both neuroprotective and<br />

neurodegenerative roles <strong>of</strong> ERK in the pathogenesis <strong>of</strong> PD. <strong>The</strong> neuroprotective role <strong>of</strong> ERK1/2<br />

in neuronal cell lines and primary neuron cultures in mammalian toxin and genetic PD models<br />

has been reported (Hashimoto et al., 2003; Cavanaugh, 2004; Hetman and Gozdz, 2004).<br />

However, at the same time, aberrant up-regulation <strong>of</strong> phospho-ERK1/2 in substantia nigra<br />

neurons <strong>of</strong> patients with PD and other Lewy body diseases, as well as in the midbrains <strong>of</strong> these<br />

patients has been documented (Zhu et al., 2002a). Furthermore, such up-regulation <strong>of</strong> ERK1/2 is<br />

associated with excessive generation <strong>of</strong> ROS and RNS in PD (Kulich and Chu, 2001). More<br />

recently, Miller et al. (2007) demonstrated that NADPH oxidase mediates the PQ-induced<br />

elevation <strong>of</strong> ROS in microglial cells, which involves activation <strong>of</strong> the ERK dependent signaling<br />

pathway.<br />

41


In a PQ+ Maneb model for PD, association <strong>of</strong> phosphorylation at serine 112 <strong>of</strong> pro-<br />

apoptotic protein Bad with the phosphorylation <strong>of</strong> p 42 /p 44 serine sites <strong>of</strong> ERK in the midbrain<br />

extract has been reported. Such association suggests the up-regulation <strong>of</strong> ERK is inclined<br />

towards apoptotic death in oxidative stress (Thiruchelvam et al., 2005). <strong>The</strong>refore, in light <strong>of</strong><br />

these reports, the exact functional role <strong>of</strong> ERK is controversial in PD and could be elucidated<br />

using a in vivo Drosophila PD model.<br />

Akt signaling pathway<br />

Akt is a serine/threonine protein kinase acting as a cellular homolog <strong>of</strong> the viral oncogene<br />

v-Akt. Although the C-terminal catalytic domain <strong>of</strong> Akt is closely related to that <strong>of</strong> most <strong>of</strong> the<br />

members <strong>of</strong> the protein kinase C (PKC) family, the N-terminal domain is distant from that <strong>of</strong><br />

PKC family members, and therefore, Akt is categorized as a member <strong>of</strong> the Protein kinase B<br />

(PKB) family (Kandel and Hay, 1999). In mammals, there are three known is<strong>of</strong>orms <strong>of</strong> the Akt<br />

kinase, Akt1, Akt2, and Akt3, each activated by various growth and survival factors. Once a<br />

surface receptor is activated, secondary messengers are activated and in turn activate<br />

phosphoinositide 3-kinase (PI3K) located upstream <strong>of</strong> Akt. Once phosphorylated, upon<br />

activation <strong>of</strong> PDK1, Akt promotes cell survival through various distinct pathways. Among them,<br />

two are commonly implicated. In the first one, activation inhibits apoptosis by phosphorylating<br />

the Bad component <strong>of</strong> the Bad/Bcl-xL complex. <strong>The</strong> phosphorylated Bad binds to 14-3-3 causing<br />

dissociation <strong>of</strong> the Bad/Bcl-xL complex and mediating cell survival. In the second one, Akt<br />

directly antagonizes the activation <strong>of</strong> NF-κβ inhibitor, IKK-α, ultimately leading to NF- κβ<br />

activation and cell survival (Burke, 2007) (Fig. 1.13). Akt also regulates cell growth,<br />

42


proliferation, migration, glucose metabolism, transcription, protein synthesis and angiogenesis<br />

(Brazil & Hemmings, 2001).<br />

Drosophila Akt signaling pathway<br />

<strong>The</strong> Drosophila genome contains a single Akt1 gene encoding a protein that is 76.5%<br />

similar to mammalian Akt protein (Franke et al., 1994). All known components <strong>of</strong> the<br />

mammalian Akt signaling pathway are implicated in the Drosophila Akt signaling pathway<br />

(Scanga et al., 2000). Genetic analysis <strong>of</strong> Akt demonstrated that this kinase mediates an anti-<br />

apoptotic mechanism that is regulated by caspases instead <strong>of</strong> the pro-apoptotic components,<br />

reaper, hid and grim (Staveley et al., 1998).<br />

Akt signaling pathway and Parkinson’s disease<br />

<strong>The</strong>re is compelling evidence supporting dysregulation <strong>of</strong> the mammalian Akt signaling<br />

pathway in genetic and toxin models for PD (Seo et al., 2002 ; Rodriguez-Blanco et al., 2008;<br />

Xiromerisiou et al., 2008 ). Yang et al. (2005) reported that knock-down <strong>of</strong> DJ1, one <strong>of</strong> the PD-<br />

associated genes, down-regulates the PI3K/Akt signaling pathway, thereby causing loss <strong>of</strong> DA<br />

neurons in Drosophila. <strong>The</strong>refore, these studies suggest that inactivation <strong>of</strong> Akt results in<br />

apoptotic death <strong>of</strong> DA neurons. It is proposed that Akt negatively regulates the phosphorylation<br />

and activation <strong>of</strong> c-jun, a component <strong>of</strong> the JNK signaling pathway known to induce apoptotic<br />

death in numerous PD mammalian models. Ries et al. (2006) proposed that vector based delivery<br />

<strong>of</strong> Akt is associated with neurotrophic effects in mammals. However, it is to be noted that most<br />

<strong>of</strong> these studies have utilized in vitro PD models and thus the functional role <strong>of</strong> Akt at the whole<br />

43


organism level has not been well elucidated. Moreover, its role in PQ-mediated DA toxicity is<br />

lacking.<br />

44


Figure 1.12: A schematic diagram showing the signaling pathways associated with activated Akt.<br />

Stimulus from growth and survival factors results in activation <strong>of</strong> Akt via PI3K. Activated Akt<br />

promotes cell survival by inhibiting pro-apoptotic gene, Bad, and by stimulating activation <strong>of</strong><br />

NF-κβ (also, see text).<br />

45


Gaps in understanding the mechanism <strong>of</strong> induction <strong>of</strong> an inflammatory response in Parkinson’s<br />

disease and its importance in the pathogenesis <strong>of</strong> Parkinson’s disease<br />

<strong>The</strong>re are several studies correlating the induction <strong>of</strong> the inflammatory process and its<br />

role in the pathogenesis <strong>of</strong> Parkinson’s disease in mammalian models. However, there are<br />

several unanswered key questions. <strong>The</strong>se questions are presented below briefly with possible<br />

directions to answer them.<br />

1. While there are compelling studies linking the process <strong>of</strong> inflammation to the pathogenesis<br />

and progression <strong>of</strong> Parkinson’s disease, most <strong>of</strong> these studies have been performed using in vitro<br />

approaches. It is still debated whether activation <strong>of</strong> microglia in PD is deleterious or beneficial to<br />

the individual. Moreover, studies exploring the importance <strong>of</strong> genetic interaction in the<br />

inflammatory process are lacking. A simple and genetically modifiable model that would allow<br />

in vivo approaches could provide novel insights into the process <strong>of</strong> inflammation in the<br />

Parkinson’s disease. Chapter 2 describes the discovery <strong>of</strong> the first in vivo Drosophila model<br />

capable <strong>of</strong> inducing increased expression <strong>of</strong> NOS against PQ exposure. We have used this model<br />

to study the role <strong>of</strong> genes modulating the inflammatory response in Parkinson’s disease using a<br />

well-known anti-inflammatory drug, minocycline. This chapter presents the data which could be<br />

further explored to determine the source <strong>of</strong> NOS in response to paraquat and may support the<br />

induction <strong>of</strong> NOS expressing mammalian microglial cells in PQ-induced in vivo Drosophila PD<br />

model.<br />

2. A variety <strong>of</strong> signal transductions pathways have been implicated in processes leading to<br />

pathological conditions in neurons and to the neuroinflammatory response. Most components <strong>of</strong><br />

these pathways are evolutionary conserved, exhibiting functional similarities in invertebrates and<br />

vertebrates. <strong>The</strong> activation <strong>of</strong> signaling pathways in PD have been extensively studied in<br />

46


mammalian genetic and toxic PD models. However, the functional redundancy associated with<br />

the existence <strong>of</strong> multiple genes and numerous is<strong>of</strong>orms in mammals creates difficulties in<br />

understanding the exact role <strong>of</strong> signaling pathway in the pathogenesis <strong>of</strong> PD and could be<br />

eliminated in the simple in vivo Drosophila PD model, since most signaling proteins are encoded<br />

by single genes. In Chapter 3, identification <strong>of</strong> the neuroprotective role <strong>of</strong> minocycline in a<br />

Drosophila PD model are described. Further, this chapter also reports preliminary studies for the<br />

signaling pathways involved with DA toxicity in this Drosophila PD model.<br />

3. <strong>The</strong> occurrence <strong>of</strong> Parkinson’s disease in the population <strong>of</strong> less than 40 years <strong>of</strong> age is referred<br />

to as Early Onset Parkinsonism. Chapter 3 investigates the correlation <strong>of</strong> brief exposure to<br />

paraquat during the juvenile and young ages with the pathogenesis <strong>of</strong> PD in Drosophila PD<br />

model. This study opens new avenues to understand the progressive alteration in dopamine<br />

homeostasis during the entire period <strong>of</strong> life in a simple and efficient manner.<br />

4. Although the exact etiological agents for PD pathogenesis is still not known, experimental<br />

studies have resulted in the hypothesis that multiple factors including exposure to insecticides,<br />

pesticides, and other environmental toxins could act as substantial etiological causes for sporadic<br />

PD. It is noteworthy that increased incidence <strong>of</strong> PD is associated reported in some agricultural<br />

populations. Recently, the role <strong>of</strong> bacterial pathogens in neuronal degeneration has been tested<br />

(<strong>The</strong> Caldwell Lab, UA). <strong>The</strong>y have identified Streptomyces venezuale as a pathogen responsible<br />

for the loss <strong>of</strong> dopaminergic neurons in C. elegans., In Chapter 5, one <strong>of</strong> the possible<br />

mechanisms <strong>of</strong> action <strong>of</strong> this toxin via induction <strong>of</strong> NOS positive structures in adult Drosophila<br />

brain in the pathogenesis <strong>of</strong> PD has been elucidated.<br />

47


CHAPTER 2<br />

DROSOPHILA MOUNTS A NITRIC OXIDE-DEPENDENT RESPONSE TO PARAQUAT-<br />

INDUCED DEGENERATION OF DOPAMINERGIC NEURONS<br />

This work is under revision for submission to Nature Neuroscience with the following authors:<br />

<strong>Inamdar</strong> A., Lawal H., Ferdousy F., Chaudhuri A., and O’Donnell J. Drs. Hakeem Lawal and<br />

Faiza Ferdousy collected data presented in Fig. 2.6 and Fig. 2.8. Dr. Anathbandhu Chaudhuri<br />

helped <strong>Arati</strong> <strong>Inamdar</strong> to collect data presented in Fig. 2.3. <strong>The</strong> remaining data were collected and<br />

analyzed by <strong>Arati</strong> <strong>Inamdar</strong>. Dr. Janis O’Donnell and <strong>Arati</strong> <strong>Inamdar</strong> wrote the manuscript.<br />

48


INTRODUCTION<br />

Excessive activation <strong>of</strong> the innate inflammatory response is considered a crucial and key<br />

factor in the pathogenesis <strong>of</strong> neurodegenerative diseases as well as in neurological conditions<br />

associated with injury and compromised blood supply to brain. Recently, evidence has been<br />

presented in support <strong>of</strong> an escalated neuroinflammatory response in Parkinson’s disease (PD)<br />

both in patients and in experimental models. <strong>The</strong>se studies include documentation <strong>of</strong> increased<br />

levels <strong>of</strong> inflammatory cytokines/chemokines in PD patients and PD models (Hunot and Hirsch,<br />

2003; Liu, 2006). In addition, generation <strong>of</strong> nitric oxide, reactive nitrogen species (RNS) and<br />

peroxynitrites has been observed in experimental animals modeling Parkinsonian features and<br />

symptoms (Hunot et al., 1996; Hunot et al., 1999; Mosley et al., 2006; Block et al., 2007).<br />

Further, an increase in cyclooxygenase 2 (COX-2), the enzyme responsible for the formation <strong>of</strong><br />

prostanoids and thus mediating inflammatory responses in the substantia nigra and around DA<br />

neurons in PD patients and in MPTP and 6-OHDA-induced PD animal models, has been<br />

reported (Di Matteo et al., 2006; Tyurina et al., 2006; Vijitruth et al., 2006). Moreover,<br />

epidemiological studies have found a correlation between long-term consumption <strong>of</strong> Non-<br />

Steroidal-Anti-Inflammatory Drugs (NSAIDS) and a decreased incidence <strong>of</strong> PD (Esposito et al.,<br />

2007). Most importantly, a pr<strong>of</strong>ound increase has been observed in the number and the activity<br />

<strong>of</strong> the microglial cells, innate immune surveillance cells resident in the CNS, in the nigral<br />

regions <strong>of</strong> PD brains. <strong>The</strong>se microglial cells normally are present in small numbers in the brain,<br />

where they are maintained in a resting state in and around the SN (Sugama et al., 2003; Zhang et<br />

al., 2005).<br />

Under pathological conditions such as injury and infection, these cells mediate an inflammatory<br />

process as a normal protective response. In addition, they perform numerous cellular<br />

49


maintenance functions essential for neuronal survival, releasing trophic and anti-inflammatory<br />

factors, and clearing cellular debris by initiating programmed cell death in neurons (Liao et al.,<br />

2004; Morgan et al., 2004; Cho et al., 2006; Simard et al., 2006).<br />

However, microglia also are considered to be the main culprits responsible for converting<br />

their normal surveillance functions into functions detrimental to normal healthy neurons in the<br />

brain, thereby amplifying the effects <strong>of</strong> neurodegenerative disease or neuronal injury. Under<br />

pathological conditions such as neuronal injury or immunological stimuli, microglia are activated<br />

and release excessive chemokines/cytoxins such as TNF-α, IL-1, IL-6 and enzymes such as<br />

iNOS, COX-2 to combat the altered cells in such disease states (Block et al., 2007). <strong>The</strong> failure<br />

<strong>of</strong> the normal surveillance function <strong>of</strong> microglia in neurological diseases including PD and the<br />

derangement <strong>of</strong> the balance between neuroprotective and neurodegenerative roles is still not well<br />

understood. Further, it is unknown whether signals from dying neuron or activated microglia<br />

initiate a self-perpetuating condition resulting into amplified microgliosis in PD. Most studies<br />

suggesting an adverse role for microglia in PD pathogenesis are performed in in vitro conditions,<br />

thereby lacking strong evidence for such detrimental effects <strong>of</strong> microglia at the whole organism<br />

level. Moreover, in the absence <strong>of</strong> a simple in vivo model in which powerful genetic<br />

manipulation methods can applied to understand the functional role <strong>of</strong> microglia in Parkinson’s<br />

disease, the modulatory effect <strong>of</strong> genes participating in the neuroinflammatory response cannot<br />

be studied.<br />

Among the several microglial-derived pro-inflammatory cytokines and enzymes<br />

contributing to the dysfunction <strong>of</strong> DA neurons, the enzyme inducible NOS (iNOS) has been<br />

recently recognized as a major component in DA neuron degeneration. Various mammalian PD<br />

models have established that upon activation by neurotoxic and excitotoxic agents, glial<br />

50


(astrocytes and microglia)-derived inducible NOS (iNOS) and NO production facilitated the<br />

inhibition <strong>of</strong> the mitochondrial respiratory chain via cytochrome c inhibition, generation <strong>of</strong><br />

peroxynitrites capable <strong>of</strong> causing protein nitration, depletion <strong>of</strong> anti-oxidant pools, and<br />

modification <strong>of</strong> cellular reactions by acting as a diaphorase (Moncada and Bolanos, 2006).<br />

We have shown previously that upon ingestion <strong>of</strong> paraquat (PQ), Drosophila exhibits<br />

mobility defects, loss <strong>of</strong> DA neurons, and truncation <strong>of</strong> its lifespan, the key features <strong>of</strong> PD. In<br />

addition, mutations in genes regulating DA homeostasis, Punch (Pu), Catecholamines up<br />

(Catsup) and pale (ple) caused differential responses to PQ (Chaudhuri et al., 2007). Pu encodes<br />

GTP cyclohydrolase (GTPCH), the rate-limiting enzyme for the synthesis <strong>of</strong> tetrahydrobiopterin<br />

(BH4), which acts as a c<strong>of</strong>actor for both DA and NO biosynthesis (Fig.2.1). <strong>The</strong> Drosophila<br />

genome possesses only one NOS gene (compared to three in mammals), and the enzyme is<br />

known to have a variety <strong>of</strong> physiological and developmental functions in insects, including<br />

Drosophila.<br />

In this report, we demonstrate with biochemical and immunolocalization data that NOS<br />

synthesis is up-regulated in adult fly brain in response to PQ. To confirm the role <strong>of</strong> NOS in PQ<br />

toxicity, we co-fed PQ with a well-known anti-inflammatory drug, minocycline, which is<br />

capable <strong>of</strong> suppressing microglial and NOS activation (Wu et al., 2002; Ryu and McLarnon,<br />

2006) and found that the drug improved survival duration and suppressed the induction <strong>of</strong> NOS<br />

activity in adult. Moreover, we demonstrate that pharmacological inhibition <strong>of</strong> NOS activity<br />

similarly increases the life span during paraquat exposure.<br />

In an effort to characterize these NOS positive cells, we also present preliminary<br />

evidence that NOS is not induced in glial cells, but further work is necessary to further<br />

characterize NOS-positive structures. <strong>The</strong>refore, we present results demonstrating the capacity <strong>of</strong><br />

51


Drosophila to mount a NOS-dependent response that has some similarities to mammalian<br />

neuroinflammation mediated by microglial under neurodegenerative conditions.<br />

52


MATERIALS AND METHODS<br />

Drosophila strains and culture maintenance. Strains utilized in all experiments were<br />

Canton S, a wild type strain, the Df(1)w; y, a white-eyed strain that carries wild type alleles <strong>of</strong> all<br />

DA-regulating genes, a Pu (GTPCH) mutant strain Df(1)w, y; dp cn Pu Z22 a px sp/SM1, Cy cn 2<br />

sp 2 , a ple (tyrosine hydroxylase) mutant strain, ple 2 /TM3, Sb e, and a Catsup mutant strain,<br />

Catsup 26 /CyO. For all experiments, the mutant strains were mated to the appropriate wild type<br />

strain, and all assays were performed on mutants heterozygous for the wild type genes. All<br />

stocks were maintained at 25º C.<br />

A second chromosome UAS-eGFP transgenic strain was obtained from the Bloomington<br />

Drosophila Stock Center. <strong>The</strong> following Gal4 transgenes were employed to drive cell-specific<br />

GFP expression: tyrosine hydroxylase (TH)-GAL4 (dopaminergic neuron expression) was a gift<br />

from Jay Hirsh (<strong>University</strong> <strong>of</strong> Virginia). Reversed polarity (Repo)-GAL4 (glial expression line)<br />

was obtained from the Bloomington Stock Center.<br />

Feeding experiments. Male flies at 24 hr post-eclosion were fed on filter paper saturated<br />

with 5% sucrose only or 5% sucrose containing the following chemicals separately or in<br />

combinations as described in the Results: paraquat (1 mM, 1.5 mM, 3 mM or 10mM),<br />

minocycline HCl (200 μM or 1mM), NG-nitro-L-arginine methyl ester (L-NAME) (200μM or 2<br />

mM). For minocycline and L-NAME, preliminary dose response tests confirmed that the<br />

concentrations employed with PQ co-feeding were not toxic alone within the time constraints <strong>of</strong><br />

each experiment. Concentrations <strong>of</strong> PQ between wild type and the Punch mutant strain were<br />

varied because the Punch mutant strain is hypersensitive to PQ and the progression <strong>of</strong><br />

degeneration is too rapid at 10 mM paraquat to achieve measurable rescue with either L-NAME<br />

53


or minocycline at non-toxic concentrations. <strong>The</strong> rate <strong>of</strong> degeneration <strong>of</strong> the wild type strain on 1<br />

mM is slowed to the point that secondary effects from lack <strong>of</strong> normal medium become apparent.<br />

Concentrations <strong>of</strong> minocycline and L-NAME were varied to test responses to different<br />

concentration ratios <strong>of</strong> PQ to therapeutic drug. All reagents were obtained from Sigma. For<br />

assays <strong>of</strong> survival, feeding was continued until all flies had died, with daily supplementation with<br />

fresh solutions. For confocal microscopic analysis <strong>of</strong> adult brains, exposure to the appropriate<br />

chemicals continued for 6 hrs prior to dissection <strong>of</strong> brains.<br />

Nitric oxide synthase assay. Male and female adult flies at 24-48 hrs post-eclosion, were<br />

fed PQ, minocycline and L-NAME according to the doses indicated in the Results for up to 24<br />

hr. Head extracts were prepared by homogenizing in 0.1M phosphate buffer, pH 7.2, followed<br />

by centrifugation for 10 min at 10,000 g at 4°C. <strong>The</strong> supernatants were mixed with freshly<br />

prepared Modified Griess reagent (Sigma) in proportion <strong>of</strong> 1: 1 and incubated for 15 min. Nitrite<br />

levels were measured spectrophotometrically at 595 nm, with concentrations <strong>of</strong> nitrite calculated<br />

against a silver nitrite-derived standard curve and data was presented as a concentration <strong>of</strong> nitrite<br />

in micromolars for 50 fly-heads.<br />

Immunocytochemistry and confocal microscopy. Brains from TH-GAL4; UAS-eGFP<br />

adults (untreated or exposed to 1.5 mM or 3 mM paraquat for 6 or 12 hrs) were fixed in 4%<br />

paraformaldehyde for 3.5 h and washed extensively in 1x phosphate buffered saline (PBS) and<br />

then in 0.1% Triton X-100, 0.2% bovine serum albumin, in PBS (PBT). Brains were then<br />

blocked in 5% normal goat serum in PBT overnight at 4°C, followed by overnight incubation<br />

with a 1:500 dilution <strong>of</strong> rabbit anti-universal NOS antiserum (Catalogue No. N127, Sigma) and<br />

54


mouse anti-GFP antibody (Abcam). After additional washing, the brains were incubated at room<br />

temperature for 2 h in a 1: 5000 dilutions <strong>of</strong> Cy-3-conjugated goat anti-rabbit IgG and FITC<br />

conjugated rabbit anti-mouse IgG. Confocal studies were performed using a Leica TCS SP2<br />

AOBS confocal microscope (Wetzlar, Germany). Each brain was scanned to include 10-15<br />

sections for optimum visualization <strong>of</strong> NOS-positive structures. For capturing magnified images<br />

(63X and 126X) about 5-8 Z-sections were used to obtain average image <strong>of</strong> all sections. <strong>The</strong>se<br />

imaging criteria were utilized for all the images mentioned in the Results section.<br />

Statistical Analysis. All data were analyzed by one way ANOVA with Dunnett’s post<br />

test or two way ANOVA or by two-tailed Student’s t-test, assuming equal variances wherever<br />

applicable, using GraphPad Prism (San Diego, CA). Details <strong>of</strong> the analyses are described in the<br />

figure legends.<br />

55


Tyrosine hydroxylase<br />

Tyrosine<br />

L-Dopa Dopamine<br />

GTP cyclohydrolase<br />

GTP H2NPPP PTP BH4<br />

Arginine<br />

Nitric oxide synthase<br />

Citrulline +<br />

Figure 2.1: Dopamine, tetrahydrobiopterin, and nitric oxide biosynthesis pathways. BH4<br />

functions as an essential c<strong>of</strong>actor for the generation <strong>of</strong> DA from tyrosine, and NO from citrulline<br />

in separate pathways. Tyrosine hydroxylase converts tyrosine into L-Dopa which is converted<br />

into dopamine. GTP cyclohydrolase acts as a rate limiting enzyme for synthesis <strong>of</strong> BH4 which is<br />

formed from guanosine triphosphate (GTP) through the formation <strong>of</strong> two intermediates,<br />

dihydroneopterin triphosphate (H2NPPP) and 6- pyruvoyl tetrahydropterin (PTP). Nitric oxide<br />

synthase converts citrulline to arginine with generation <strong>of</strong> nitric oxide.<br />

56<br />

Nitric oxide


RESULTS<br />

A NOS inhibitor, L-NAME, improves survival duration <strong>of</strong> adults exposed to paraquat<br />

Recently, nitric oxide was proposed to be involved in PQ-induced neurotoxicity in<br />

mammals (Djukic et al., 2007). We therefore asked if the PD-like effects from PQ exposure in<br />

our Drosophila model are also mediated via nitric oxide. To test this, we co-fed wild type flies 2<br />

mM L-NAME, a non-selective NOS inhibitor and 10 mM PQ continuously until all flies were<br />

dead. We found that addition <strong>of</strong> L-NAME results in improvement <strong>of</strong> survival duration by<br />

approximately 1.5 days when compared to the survival duration <strong>of</strong> PQ-fed wild type flies,<br />

suggesting that truncation <strong>of</strong> life span with PQ exposure is at least partially due to NO-induced<br />

damage and prevention <strong>of</strong> PQ-mediated NOS induction could be neuroprotective (Fig. 2.2).<br />

57


Survival (days)<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

PQ<br />

Figure 2.2: L-NAME, an inhibitor <strong>of</strong> NOS, improves survival duration <strong>of</strong> adults exposed to<br />

paraquat. 48 hr old post eclosion flies were fed 10 mM PQ alone or 10 mM PQ with 2 mM L-<br />

NAME continuously until death and the average survival duration was calculated. Co-feeding <strong>of</strong><br />

PQ and L-NAME extends the average survival duration by about 1.5 days, compared to flies fed<br />

PQ alone. ** =P


Minocycline prevents PQ-induced truncation <strong>of</strong> life span in wild type flies<br />

It is also reported recently that minocycline enhances survival <strong>of</strong> paraquat (PQ)-fed<br />

Drosophila (Bonilla et al., 2006). Because minocycline toxicity has been reported in some<br />

mammalian disease models, we first tested increasing concentrations <strong>of</strong> minocycline on non-PQ<br />

treated flies. We found that ingestion <strong>of</strong> concentrations <strong>of</strong> 5 mM and above affected viability,<br />

while lower concentrations caused no observable deleterious effects (Fig. 2.3 A). <strong>The</strong>refore, all<br />

subsequent experiments utilized minocycline concentrations <strong>of</strong> 1 mM or less. We asked whether<br />

minocycline is able to rescue PQ- induced toxicity in flies. To test this, we determined the<br />

average survival duration <strong>of</strong> wild type adult flies continuously fed PQ alone (10 mM), or 10 mM<br />

PQ with 1 mM minocycline. We found that flies survived about 4 days when exposed to PQ<br />

alone, but addition <strong>of</strong> 1 mM minocycline with the PQ extended the survival duration by about 2<br />

days (Fig. 2.3 B). A similar response was detected by pre-feeding flies 1 mM minocycline for 5<br />

days and then feeding only 10 mM paraquat until death (Fig. 2.3 B). <strong>The</strong>refore, as in the<br />

mammalian PD model studies, minocycline ameliorates PQ-induced truncation <strong>of</strong> life span in<br />

our Drosophila model.<br />

59


A B<br />

Mortality (%)<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

0 24 48 72 96 120 144 168 192 216 240<br />

Duration (hr)<br />

100uM<br />

250uM<br />

500uM<br />

1mM<br />

5mM<br />

10mM<br />

25mM<br />

50mM<br />

Figure 2.3: Effect <strong>of</strong> ingestion <strong>of</strong> minocycline, with and without PQ, on survival duration <strong>of</strong> wild<br />

type flies. (A) Effect <strong>of</strong> increasing concentrations <strong>of</strong> minocycline (100 μM-50 mM) on survival<br />

<strong>of</strong> adult male flies. Data were collected every 24 hrs for each group until 100 % mortality was<br />

noted with 50mM minocycline. (B) Wild type flies at 48 hrs post-eclosion were fed 10 mM PQ<br />

alone, after 5 days <strong>of</strong> continuous ingestion <strong>of</strong> minocycline (1 mM) and along with minocycline<br />

(1 mM). <strong>The</strong> co-feeding group was compared to the PQ alone group and the pre-feeding group<br />

was compared with the same aged group pre-fed 5% sucrose and then PQ (10 mM). Both c<strong>of</strong>eeding<br />

and post-feeding regimens improved the survival duration compared to PQ alone. **=<br />

P


Minocycline reduces the PQ-induced increase in NOS activity in wild type adult flies<br />

It is proposed that minocycline in some PD models, to be neuroprotective, prevents up-<br />

regulation <strong>of</strong> inducible nitric oxide synthase (iNOS), an important enzyme mediating<br />

inflammatory response in an MPTP-induced mammalian model for Parkinson’s disease (Du et<br />

al., 2001; Wu et al., 2002). We hypothesized that minocycline could reduce the PQ-induced<br />

toxicity by preventing the induction <strong>of</strong> NOS. We sought to test this by quantifying the level <strong>of</strong><br />

nitrites, the product <strong>of</strong> NO breakdown, after exposure to PQ alone or with minocycline, using<br />

modified Griess reagent (Green et al., 1982). We found that NOS activity was induced to about<br />

4.5 to 5 mM within the first 24 hrs <strong>of</strong> exposure to PQ in both males and females against 3 mM in<br />

control flies. However, minocycline, as in mammalian models, inhibits production <strong>of</strong> NO in flies<br />

that were co-fed PQ and minocycline (Fig. 2.4 A, B). <strong>The</strong>se data confirm similarities in the<br />

mechanism <strong>of</strong> action between L-NAME and minocycline in Drosophila and mammals, and they<br />

provide evidence <strong>of</strong> the anti-inflammatory property <strong>of</strong> minocycline via suppression <strong>of</strong> NOS in<br />

flies.<br />

61


A<br />

B<br />

Nitrite (M)<br />

Nitrite (M)<br />

5% sucrose<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

5% sucrose<br />

*<br />

*<br />

PQ<br />

##<br />

PQ+Min<br />

5% sucrose<br />

PQ<br />

Male Female<br />

PQ<br />

##<br />

PQ+Min<br />

5% sucrose<br />

*<br />

PQ<br />

Male Female<br />

*<br />

NS<br />

PQ+Min<br />

##<br />

PQ+Min<br />

Figure 2.4: PQ induces, and minocycline suppresses, NOS activity in adult flies. (A) After 24 hrs<br />

<strong>of</strong> PQ exposure, the specific activity <strong>of</strong> NOS is five-fold higher in both males and females.<br />

Minocycline, co-fed with PQ, suppresses NOS induction in males but not in females. (B)<br />

Induction <strong>of</strong> NOS and suppression by minocycline was noted in both males and females after 48<br />

hrs <strong>of</strong> ingestion <strong>of</strong> PQ and minocycline. All NOS activity values are averages <strong>of</strong> three<br />

independent assays per condition. * = P < 0.05 and ** = P< 0.005 and represent the significant<br />

difference between control and PQ fed groups; ##= P< 0.005 and represents the significant<br />

difference between PQ, and minocycline and PQ co-fed group. Error bars represent standard<br />

error <strong>of</strong> means and n= 250-300 fly heads for all groups.<br />

62


Minocycline increases the life span <strong>of</strong> ple and Catsup mutants but cannot rescue Pu mutants<br />

We previously reported the differential PQ sensitivity <strong>of</strong> Drosophila strains heterozygous<br />

for mutations in DA-regulating genes. <strong>The</strong>se included mutations in the rate-limiting genes for<br />

BH4 and DA synthesis, Punch (Pu; GTPCH) (Mackay and O'Donnell, 1983) and pale (ple; TH)<br />

(Neckameyer and White, 1993), respectively. <strong>The</strong>se mutations result in decreased DA pools in<br />

adult heads, even in the heterozygous state in which a copy <strong>of</strong> a wild type allele <strong>of</strong> each gene is<br />

also present. Further, these strains are highly sensitive to PQ (Chaudhuri et al., 2007).<br />

Conversely, heterozygous strains carrying one mutant and one wild type allele <strong>of</strong><br />

Catecholamines up (Catsup), a negative regulator <strong>of</strong> TH and GTPCH (Stathakis et al.,1999),<br />

have elevated BH4 and DA pools and a strong resistance to PQ (Chaudhuri et al., 2007). We<br />

asked whether these mutants showed altered responses to PQ in the presence <strong>of</strong> minocycline.<br />

Wild type adult males survive approximately 4.5 days on 10 mM PQ, while Catsup<br />

heterozygotes survive 24 hrs longer, on average (Fig.2.5 A). In contrast, the low DA Pu and ple<br />

heterozygotes survive only 2 days (Fig. 2.5 A). Survival <strong>of</strong> Catsup and ple mutants, as well as<br />

wild type, was extended by minocycline, by approximately the same increments. <strong>The</strong>refore, we<br />

detected no DA-specific interactions with minocycline in these mutants. Strikingly, however,<br />

minocycline was unable to improve the survival <strong>of</strong> Pu mutants under these conditions. One<br />

possibility for this result could be that oxidative damage in Pu mutants progresses too rapidly for<br />

minocycline to impart its protective effect. We therefore, reduced the dose <strong>of</strong> PQ fed to the Pu<br />

mutant animals to 1 mM, which, though eventually causing similar neurological symptoms,<br />

extends life span and the time period before onset <strong>of</strong> movement disorders, consistent with a<br />

reduction in oxidative stress compared to 10 mM paraquat. We then tested 1 mM and 200 μM<br />

minocycline to determine whether a slower accumulation <strong>of</strong> oxidative damage with the lower PQ<br />

63


dose would allow some amelioration <strong>of</strong> symptoms by minocycline. We found, however, that co-<br />

feeding at these concentrations also failed to rescue the mutant flies (Fig. 2.5 B). We further<br />

detected elevated NO production with PQ treatment in wild type and Pu mutants which was<br />

alleviated by minocycline in wild type flies but not in Pu mutants (Fig. 2.5 C, D). Since Pu<br />

mutants have limited BH4 production (Krishnakumar et al., 2000) and limiting BH4 is known to<br />

cause catalytic uncoupling <strong>of</strong> NOS, producing elevated peroxides and peroxynitrites in in vitro<br />

conditions, we hypothesized that the failure <strong>of</strong> minocycline protection in Pu mutants was linked<br />

to BH4 deficits leading to more catastrophic oxidative damage (Presta et al., 1998; Werner et al.,<br />

2003; Foxton et al., 2007). If this hypothesis is correct, then inhibition <strong>of</strong> NOS activity should<br />

limit the production <strong>of</strong> ROS and RNS, and thereby, improve survival <strong>of</strong> the Pu mutant flies. We<br />

therefore, co-fed PQ and L-NAME to Pu mutants and found extension <strong>of</strong> survival <strong>of</strong> the Pu<br />

mutants by 3 days (Fig. 2.5 E). In addition, we observed that co-feeding <strong>of</strong> PQ and L-NAME for<br />

24 hr resulted in a decrease in NO production, in contrast to the absence <strong>of</strong> NOS suppression<br />

with minocycline (Fig. 2.5 F). <strong>The</strong>se results suggest that the failure <strong>of</strong> minocycline to rescue Pu<br />

mutants was due to its inability to control excessive NO production when BH4 production is<br />

compromised (Fig 2.5 E, F). This interpretation is supported by the observation that untreated<br />

heterozygous Pu mutants possessed low nitrite levels relative to wild type adults (Figure 2.5 F)<br />

and stimulation <strong>of</strong> NOS activity by PQ, which would exacerbate the uncoupling effects <strong>of</strong><br />

limiting BH4 (Fig. 2.5 D). <strong>The</strong>refore, these results show that our in vivo model could be used to<br />

identify the modulatory effect <strong>of</strong> genes on the protective properties <strong>of</strong> minocycline or<br />

minocycline-like therapeutic agents in simple and efficient manner..<br />

64


Figure 2.5: Minocycline shows differential effects on paraquat-induced damage in dopamine<br />

regulatory mutants. (A) Effect <strong>of</strong> 1 mM minocycline co-fed with 10 mM PQ on DA regulatory<br />

mutants, Catsup 26 /+, Pu Z22 /+ and ple 2 /+. Mutant adult males were continuously exposed to<br />

either 10 mM PQ only or 10 mM PQ with 1 mM minocycline. Catsup and ple mutant flies<br />

showed significant extension <strong>of</strong> life span, incrementally similar to the wild type control strain,<br />

while Pu mutants did not. (B) Neither 200 μM nor 1 mM minocycline improved the survival <strong>of</strong><br />

Pu mutants exposed to 1 mM PQ. (C) After 24 hr <strong>of</strong> PQ, or PQ with minocycline exposure,<br />

suppression <strong>of</strong> NOS was detected in wild type heads. (D) <strong>The</strong> NO levels <strong>of</strong> non-PQ treated Pu<br />

mutants, assayed at 48 hr post-eclosion, are significantly lower than NO levels in non-PQ treated<br />

wild type heads. Minocycline, which prevents PQ-induced elevation <strong>of</strong> NO in wild type heads<br />

(see C), could not modify NO levels induced by PQ in Pu. (E) <strong>The</strong> survival <strong>of</strong> Pu mutants was<br />

improved by co-feeding <strong>of</strong> L-NAME with PQ when compared with survival <strong>of</strong> Pu mutants on<br />

PQ alone. (F) Addition <strong>of</strong> L-NAME to PQ reduced the NO production in Pu mutants. In contrast,<br />

200 μM L-NAME prevented PQ-induced decrease in survival duration. NS = not significant. **<br />

= P < 0.005 and represent significant differences between PQ and PQ and minocycline in Fig<br />

2.7A. *, ** and *** =P < 0.05, P=< 0.005 and P=


A<br />

Survival (days)<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

**<br />

PQ (1mM)<br />

NS<br />

**<br />

**<br />

Wild type-PQ<br />

Wild-type-PQ+Min<br />

Pu/+ -PQ<br />

Pu/+ -PQ+Min<br />

Catsup/+ -PQ<br />

Catsup/+ -PQ+Min<br />

Pale/+ -PQ<br />

Pale/+ -PQ+Min<br />

C D<br />

E<br />

Survival (days)<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

***<br />

PQ+L-NAME (1mM+200 uM)<br />

66<br />

F<br />

B<br />

Survival (days)<br />

Nitrite (M)<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

PQ (1mM)<br />

5% sucrose (wild type)<br />

NS<br />

PQ+Min (1 mM+200 uM)<br />

*<br />

5% sucrose (Pu/+)<br />

*<br />

PQ+Min (1 mM+1 mM)<br />

*<br />

PQ (Pu/+)<br />

PQ+Min (Pu/+)


PQ exposure causes activation <strong>of</strong> NOS as evidenced by expression <strong>of</strong> NOS positive structures in<br />

adult fly brain<br />

Since exposure to PQ with L-NAME and minocycline delayed PQ-induced truncation <strong>of</strong><br />

life span in flies, we hypothesized that the activation <strong>of</strong> NOS was contributing to PQ-induced<br />

damage to the CNS. To test this, we sought to detect the expression <strong>of</strong> NOS in the adult brain<br />

using universal anti-NOS antibody. This antibody was raised against a conserved amino-acid<br />

sequences corresponding to amino acid residues 1113-1122 <strong>of</strong> murine iNOS and nNOS. A<br />

polyclonal antibody raised against this epitope from a different commercial source has been<br />

previously used to detect the expression <strong>of</strong> NOS during Drosophila visual system development<br />

and in malphigian tubules (Davies, 2000; Gibbs, 2001). Similarly, a protein <strong>of</strong> 150 kDa, the<br />

predicted molecular mass <strong>of</strong> the Drosophila NOS polypeptide was detected by immunoblotting<br />

using another polyclonal antibody independently raised against this epitope (Broderick et al.,<br />

2003). We detected little NOS-positive signal in normal, untreated brain (Fig. 2.6 A) with the<br />

exception <strong>of</strong> expression restricted to the junction <strong>of</strong> the central brain complex and optic lobes<br />

(data not shown). In contrast, upon exposure to PQ, a significant increase in signal was noted in<br />

the central complex (Fig. 2.6 B). Further, these NOS-positive structures exhibited an array-like<br />

pattern suggesting that they might be aligning along processes <strong>of</strong> neuronal or non-neuronal<br />

structures. <strong>The</strong> size for these NOS-positive structures ranged from 0.5 to 2 μm.<br />

67


5% sucrose PQ<br />

A B<br />

C D<br />

Figure 2.6: Exposure to PQ causes induction <strong>of</strong> NOS in adult brain. (A) Little NOS expression<br />

was detected in normal, 5% sucrose brains. (B) Upon exposure to 3 mM PQ for 12 hrs, a<br />

dramatic increase in the expression <strong>of</strong> NOS was detected by anti-NOS antibody. Scale bars in A,<br />

B=10 μm.<br />

68


PQ increases the expression <strong>of</strong> NOS-positive structures near dopaminergic neurons<br />

Having found that PQ increases expression <strong>of</strong> NOS in adult Drosophila brain, we sought<br />

out to determine any association between NOS-positive cells and DA neurons in response to PQ<br />

exposure. Using the transgenic reporter strain, TH-GAL4; UAS-eGFP, we found that ingestion <strong>of</strong><br />

PQ induces NOS signal in structures near the DA neuron and or processes (Fig. 2.7 A).<br />

Moreover, we detected clusters <strong>of</strong> NOS signal around the dopaminergic neurons in the adult<br />

brains exposed to PQ (Fig. 2.7 B). Since, we found that NOS suppression by L-NAME also<br />

improves the survival <strong>of</strong> PQ-exposed flies; we propose that NOS mediates the loss <strong>of</strong> DA<br />

neurons in adult Drosophila brain.<br />

69


A B<br />

Figure 2.7: Ingestion <strong>of</strong> PQ (1.5 mM) induces NOS-expression near and around dopaminergic<br />

neurons in TH-GAL4; UAS-eGFP adult brain. (A) Arrows indicate NOS positive structures (red)<br />

near the DA neurons and or processes (green) (B) <strong>The</strong> arrowhead indicates the cell body <strong>of</strong> a<br />

dopaminergic neuron (green) surrounded by NOS positive structures (red) (arrows). Scale bar for<br />

A= 15.87 μm and for B= 5 μm.<br />

70


Paraquat induces expression <strong>of</strong> NOS in non-glial cells in adult brain<br />

Induction <strong>of</strong> NOS activity in response to bacterial invasion in Drosophila has been<br />

reported previously (Nappi et al., 2000; Foley and O’Farrell, 2003). However, there have been<br />

no published reports <strong>of</strong> an association <strong>of</strong> NO with neurodegeneration in Drosophila models <strong>of</strong><br />

neurodegenerative disease. Our data suggest that generation <strong>of</strong> NO by PQ is deleterious to<br />

Drosophila as observed in mammalian PD models. In mammals, iNOS, induced in microglia and<br />

glia is associated with their activation and proliferation in response to inflammation (Block et al.,<br />

2007). In Drosophila, glial cells perform various functions including phagocytosis <strong>of</strong> harmful<br />

non-self invaders, but the presence <strong>of</strong> microglial-like cells in the CNS <strong>of</strong> Drosophila has not yet<br />

been reported (Freeman and Doherty, 2006). <strong>The</strong>refore, we hypothesized that glial cells could be<br />

the source <strong>of</strong> NOS induced in response to PQ in flies. We used Repo-GAL4; UAS-eGFP flies,<br />

where all differentiated glial cells express reversed polarity (repo), and are therefore, identified<br />

by repo-driven expression <strong>of</strong> GFP. We found that NOS, detected by a universal anti-NOS<br />

antibody, was not expressed in glia (Fig. 2.8 A). <strong>The</strong>se results indicate that NOS-expressing cells<br />

are a distinct population from glial cells in adult Drosophila brain.<br />

71


A B<br />

Figure 2.8: Paraquat-induced NOS is expressed in non-glial cells in adult fly brain. (A) NOS<br />

expression was induced in Drosophila brain by treatment <strong>of</strong> young Repo-Gal4; UAS-GFP flies<br />

with 1.5 mM PQ. Glial cells (green, thick arrows) are distinct from NOS-expressing cells (red,<br />

thin arrows). Note that NOS-positive structures were seen <strong>of</strong> varied sizes, the one indicated with<br />

notched arrow measures about 5 μm in diameter. Scale bar in A= 31.75 μm.<br />

72<br />

C<br />

D


DISCUSSION<br />

Exposure to paraquat causes induction <strong>of</strong> NOS in the adult Drosophila brain<br />

In this report, we demonstrate for the first time, induction <strong>of</strong> NOS in adult Drosophila<br />

brain in response to PQ. Although NOS activity has been previously reported, the response was<br />

induced via bacterial and wasp infection in larvae (Nappi et al., 2001; Foley and O'Farrell, 2003).<br />

PQ is one <strong>of</strong> the most potent redox-cyclers capable <strong>of</strong> generating toxicity in lung and<br />

brain tissue in mammals (Ilett et al., 1974; Prasad et al., 2007). PQ, in fact, induces the<br />

degeneration <strong>of</strong> DA neurons in invertebrate and vertebrate models, and such models are being<br />

used to identify neurodegenerative mechanisms for PD pathogenesis as well as to identify<br />

genetic susceptibility factors for PD (McCormack et al., 2002; Chaudhuri et al., 2007; Kuter et<br />

al., 2007). In the presence <strong>of</strong> molecules with diaphorase activity, PQ transfers single electrons<br />

and generates superoxide radicals upon reacting with oxygen (Bus et al., 1974). NOS, which<br />

catalyzes the formation <strong>of</strong> NO from L-arginine (Palmer et al., 1987), acts as a diaphorase<br />

molecule that also reacts with generated superoxide radicals to form highly toxic and cell<br />

membrane permeable peroxynitrite (Nemery and van Klaveren, 1995). N G -nitro-L-arginine<br />

methyl ester (L-NAME), but not N G -monomethyl-L-arginine (L-NMMA), has been shown to<br />

block the PQ-induced increase in NADPH oxidation by inhibiting the formation <strong>of</strong> superoxide<br />

radicals (Pou et al., 1992). We found that addition <strong>of</strong> L-NAME (2 mM) to PQ (10 mM)<br />

improved the survival duration relative to age-matched PQ (10 mM) alone fed flies (Fig. 2.2).<br />

We also found that PQ (10 mM) ingestion for 24 hrs stimulated NOS activity in adult fly<br />

heads, as reflected in the increase <strong>of</strong> nitrite levels determined with modified Griess reagent (Fig.<br />

2.4 A, B; 2.5 C). Moreover, our results demonstrate that co-feeding 2 mM L-NAME decreased<br />

NOS activity levels in wild type flies (Fig.2.5 F). Our data parallel reports that L-NAME is<br />

73


protective against PQ toxicity in rats that had been pre-fed L-NAME for 30 min prior to PQ<br />

injections (Djukic et al., 2008).<br />

Drosophila possesses only one NOS gene compared to three NOS genes present in<br />

mammals and shows highest similarity with the NOS1 is<strong>of</strong>orm <strong>of</strong> mammalian NOS (Regulski<br />

and Tully, 1995). However, the single is<strong>of</strong>orm in Drosophila appears to have functional<br />

properties that are distributed amongst the three mammalian is<strong>of</strong>orms. We employed an antibody<br />

that was raised against a conserved epitope in the C-terminus <strong>of</strong> the NOS protein to detect the<br />

expression <strong>of</strong> NOS in brains <strong>of</strong> non-PQ treated and PQ treated adult flies (Fig. 2.6 A, B). NOS<br />

positive signal was found to be localized primarily at the junction <strong>of</strong> the central brain complex<br />

and the optic lobes in the absence <strong>of</strong> PQ exposure (data not shown). However, upon exposure to<br />

PQ, expression <strong>of</strong> NOS significantly increases and the location <strong>of</strong> the signal changes to regions<br />

near or surrounding DA neurons in the central brain as demonstrated in a reporter strain<br />

expressing GFP in dopaminergic neurons (Fig. 2.7 A, B). <strong>The</strong> size <strong>of</strong> the NOS-positive structures<br />

ranged from 0.5 to 2 μm. In an effort to determine the source <strong>of</strong> NOS induced in response to PQ,<br />

we exposure Repo-GAL4; UAS-eGFP adult flies to 1.5 mM <strong>of</strong> PQ. We found that glial cells<br />

identified by GFP expression driven by the glia-specific driver reverse polarity (repo)-GAL 4<br />

transgenic line, were devoid <strong>of</strong> NOS expression, demonstrating that glial cells are not <strong>of</strong> the<br />

source <strong>of</strong> NOS in PQ treated brain (Fig. 2.8 A). <strong>The</strong>se findings suggest that these small NOS-<br />

positive structures could be cellular inclusions or extracellular particles, bacteria.<br />

It is to be noted that the increased expression <strong>of</strong> NOS-positive structures and their<br />

variability in expression needs further validation by employing other staining techniques such as<br />

the NADPH diaphorase colorimetric reaction. Furthermore, the increased expression <strong>of</strong> the<br />

NOS-positive structures requires quantification to confirm the expression pattern. Nevertheless,<br />

74


our data have pioneered the demonstration <strong>of</strong> a NOS-dependent response to PQ in adult<br />

Drosophila brain, that appears to be similar to responses observed in mammalian PD models and<br />

further studies will be performed to determine the source <strong>of</strong> NOS after confirming the results<br />

obtained so far.<br />

Minocycline imparts anti-inflammatory functions against PQ<br />

In order to confirm the neuroinflammatory response against PQ, we took a<br />

pharmacological approach by using a potent anti-inflammatory agent reported to delay the<br />

inflammation-mediated pathological process in neurodegenerative diseases including PD (Kim<br />

and Suh, 2008). We found that co-feeding and 5 days <strong>of</strong> pre-feeding 1 mM minocycline with 10<br />

mM PQ are effective in increasing the survival duration <strong>of</strong> flies by about 2-3 days compared to<br />

an average survival <strong>of</strong> 4 days on PQ alone after finding determining that 1 mM minocycline and<br />

below are non-toxic in flies (Fig. 2.3 A, B). Since minocycline has been reported to inhibit the<br />

activation <strong>of</strong> NOS in mammalian neurodegenerative models (Chen et al., 2000; Du et al., 2001),<br />

we tested the levels <strong>of</strong> NOS induction by PQ with and without minocycline treatment. We found<br />

that co-feeding minocycline with PQ decreases NOS activity in both males and females although<br />

a significant effect was evident in females only after 48 hrs, while response in males was<br />

observed after 24 hrs <strong>of</strong> the feeding regimen, implying possible gender-specificity in the action<br />

<strong>of</strong> minocycline (Fig. 2.4 A, B). Hence, our biochemical data suggest that, as in vertebrate PD<br />

models, minocycline imparts NOS inhibitory functions in this invertebrate model for PD, one <strong>of</strong><br />

the important inflammatory marker in PD.<br />

75


Minocycline causes differential responses to DA regulatory gene mutations in PQ exposure<br />

After establishing the protective role <strong>of</strong> minocycline during PQ exposure in wild type<br />

Drosophila, we next tested its effects in strains mutant for DA regulatory genes. We tested three<br />

key genes that we previously found to modulate DA synthesis and affect PQ sensitivity, pale,<br />

Punch and Catsup, the TH, GTPCH and Catsup encoding genes, respectively (Neckameyer and<br />

White, 1993; Mackay and O’Donnell, 1983; Stathakis et al., 1999; Chaudhuri et al., 2007). Since<br />

these mutations are homozygous lethal, we tested heterozygous mutants <strong>of</strong> ple, Pu and Catsup to<br />

determine whether their effects on DA homeostasis affected the efficacy <strong>of</strong> minocycline. We<br />

found that co-feeding 1 mM minocycline with PQ extended the survival duration <strong>of</strong> ple and<br />

Catsup mutants (low and high DA levels, respectively) to the same extent as it did wild type<br />

Drosophila (Fig. 2.5 A). We conclude from this result that perturbations <strong>of</strong> DA pools per se do<br />

not affect the ameliorative properties <strong>of</strong> minocycline. Interestingly, Punch mutants were not<br />

responsive to minocycline. We further decreased the concentration <strong>of</strong> PQ to equalize the<br />

proportion <strong>of</strong> PQ and minocycline, hypothesizing that the severity <strong>of</strong> Pu mutant phenotypes<br />

required additional minocycline for observable rescue. However, we were still unable to detect<br />

any protective effect <strong>of</strong> minocycline on Pu mutants. This result contrasts with the ability <strong>of</strong> L-<br />

NAME to improve the survival duration <strong>of</strong> Pu mutants (Fig 2.5 E). Pu mutants are deficient in<br />

BH4 synthesis and this c<strong>of</strong>actor is required by NOS as well as by TH. Because it has been<br />

reported that limiting BH4 can result in the uncoupling <strong>of</strong> electron transfer in NOS under in vitro<br />

conditions (Presta et al., 1998; Werner et al., 2003), we asked whether limiting c<strong>of</strong>actor<br />

produced an elevated oxidative/nitrosative background that enhanced PQ sensitivity including<br />

high nitrite concentrations. However, in the non-PQ treated Pu mutants, Greiss reagent assays<br />

detected lower than normal nitrite levels consistent with lower than normal NOS activity<br />

76


expected in Pu mutants (Fig. 2.5 D). <strong>The</strong>refore, the 50% decrease in BH4 pools observed in<br />

heterozygous Pu mutants (Krishnakumar et al., 2000) is not sufficient to produce elevated<br />

peroxynitrites in the absence <strong>of</strong> PQ. We found, however, that, as in wild type flies, NOS activity<br />

was further enhanced in the presence <strong>of</strong> PQ in Pu mutants (Fig. 2.5 D). This result suggests that<br />

the BH4 deficit, which we previously found to be exacerbated by PQ treatment (Chaudhuri et al.,<br />

2007) might lead to a greater imbalance between the c<strong>of</strong>actor and NOS and, in consequence,<br />

more severe uncoupling <strong>of</strong> Drosophila NOS, leading to excessive production <strong>of</strong> highly reactive<br />

superoxide radicals. This scenario is supported by our observation that L-NAME, but not<br />

minocycline, was able to reduce NOS levels and extend the survival duration <strong>of</strong> Pu mutants (Fig.<br />

2.5 E, F). A similar response is reported in BH4 limited samples <strong>of</strong> vascular smooth muscles<br />

resulting in the elevation <strong>of</strong> blood pressure (Wang et al., 2008).<br />

<strong>The</strong>refore, this report presents our preliminary results <strong>of</strong> NOS-dependent response in<br />

adult Drosophila PD model that was further supported by pharmacological study.<br />

77


CHAPTER 3<br />

IDENTIFICATION OF A NEUROPROTECTIVE ROLE FOR MINOCYCLINE IN A<br />

DROSOPHILA MODEL OF PARKINSON’S DISEASE AND FUNCTIONAL ANALYSIS OF<br />

SIGNALING PATHWAYS MEDIATING PARAQUAT TOXICITY IN A DROSOPHILA PD<br />

MODEL<br />

This work is a manuscript in preparation for submission to Journal <strong>of</strong> Neuroscience. Co-authors<br />

for this work were <strong>Arati</strong> <strong>Inamdar</strong>, Anathbandhu Chaudhuri and Janis O’ Donnell.<br />

Dr. Anathbandhu Chaudhuri contributed in collecting data presented in figures 3.1, 3.2, 3.3, 3.4<br />

and 3.5. <strong>Arati</strong> <strong>Inamdar</strong> collected the remaining data.<br />

78


INTRODUCTION<br />

Parkinson disease (PD) is a debilitating neurodegenerative disease insulting initially<br />

dopaminergic neurons in the substantia nigra (SN). Although the exact causes <strong>of</strong> PD are<br />

unknown, pathogenic mechanisms include protein aggregation, defective mitochondrial function<br />

and damage to other cellular membranes, associated with increased oxidative stress within the<br />

neurons themselves. Pesticides such as rotenone and paraquat (PQ) have been implicated in the<br />

oxidative pathogenesis <strong>of</strong> idiopathic PD (Betarbet et al., 2000; McCormack et al., 2002;<br />

Uversky, 2004), while mutations in various genes are associated with familial PD (Mizuno et al.,<br />

2008).<br />

Minocycline, a second generation tetracycline drug known to be clinically safe<br />

(MacDonald et al., 1973), has shown promising ameliorative effects in animal models for<br />

chronic neurodegenerative diseases, including neurotoxin-induced PD models (Wu et al., 2002;<br />

Zhu et al., 2002b; Lin et al., 2003; Wang et al., 2003a). Minocycline appears to have anti-<br />

inflammatory property that mediates neuroprotection in PD animal models (Wu et al., 2002).<br />

Further, antioxidant property <strong>of</strong> minocycline has been reported (Kraus et al., 2005).<br />

It was reported recently that minocycline enhances survival <strong>of</strong> paraquat (PQ)-fed<br />

Drosophila (Bonilla et al., 2006), but the precise mechanism <strong>of</strong> minocycline action is not well-<br />

understood in flies. We have developed a Drosophila model based upon ingestion <strong>of</strong> PQ, which<br />

recapitulates characteristic symptoms <strong>of</strong> PD with modulation <strong>of</strong> dopamine (DA) pools,<br />

degeneration <strong>of</strong> dopaminergic neurons, and accompanying neurological symptoms including<br />

resting tremors and postural instability (Chaudhuri et al., 2007). Moreover, we demonstrated that<br />

mutations that directly alter the regulation <strong>of</strong> DA production dramatically alter susceptibility to<br />

79


PQ, and more recently, we discovered that nitric oxide synthase (NOS) expression is induced in<br />

response to PQ (<strong>Inamdar</strong> et al., <strong>2009</strong>, under revision).<br />

<strong>The</strong> mitogen activated protein kinases (MAPKs) consist <strong>of</strong> three subfamilies, namely<br />

extracellular signal regulated kinase (ERK), c-Jun N-terminal kinases (SAPK/JNK), and p38<br />

kinases which are known to be activated by a wide range <strong>of</strong> stimuli including hormones and<br />

growth factors, inflammatory cytokines, and diverse environmental stresses. <strong>The</strong>se signal<br />

transduction pathways mediate a variety <strong>of</strong> downstream effects such as the regulation <strong>of</strong> gene<br />

transcription, the cell cycle, cellular differentiation and cell death (Gotoh and Nishida, 1995;<br />

Chang and Karin, 2001). <strong>The</strong>se MAPK-mediated downstream effects depend on the stimulus and<br />

the cell type in which they are activated. Moreover, experimental conditions employed to<br />

investigate the roles <strong>of</strong> these signaling pathways, in particular developmental processes, or in<br />

pathogenic responses are generally manipulated in mammalian cell culture systems.<br />

Nevertheless, the dysregulation <strong>of</strong> MAPK has been reported in many neurodegenerative<br />

diseases including PD (Burke et al., 2007). In these mammalian models for neuronal injury and<br />

neurodegenerative diseases, pharmacological approaches have provided evidence that p38 and<br />

JNK are mainly implicated in the neuronal death processes, while ERK may promote cellular<br />

recovery/ survival from neuronal death implicated in these conditions (Xia et al., 1995; Harper<br />

and LoGrasso, 2001). Immortalized rat brain neuroblasts (E18 cells) exposed to low doses <strong>of</strong> PQ,<br />

up regulate phosphorylated forms <strong>of</strong> ERK, PKB and JNK, but exhibit no measurable increase in<br />

phospho-p38. Further, when these kinases were blocked pharmacologically, only inhibition <strong>of</strong><br />

JNK prevented PQ-induced cell death (Niso-Santano et al., 2006). Peng et al. (2004) reported<br />

that activated JNK induced caspase-3 dependent apoptosis in Primary Mesencephalic<br />

dopaminergic neuron cultures, in response to PQ but detected no role for ERK in this system.<br />

80


Although these studies present strong evidence for the key roles <strong>of</strong> activated kinases in<br />

neuronal survival or death after exposure to PQ, there are discrepancies due to variations in the<br />

doses, cell types and cultural conditions employed. Further, in vitro cultural conditions lack<br />

appropriate cell-cell interactions from neighboring cells which could modulate the response to<br />

external stimuli. Moreover, many <strong>of</strong> the commercially available pharmacological inhibitors <strong>of</strong><br />

kinases employed in these experiments are not well-defined with respect to their exact modes <strong>of</strong><br />

action and may give rise to non-specific effects due to blockage <strong>of</strong> unknown target molecules<br />

(Sekiguchi et al., 1999; Learish et al., 2000; Waetzig and Herdegen, 2005).<br />

All these limitations <strong>of</strong> in vitro mammalian PD models necessitate the development <strong>of</strong> a<br />

simple in vivo model to further define the exact roles <strong>of</strong> these kinases in PD pathogenesis.<br />

Drosophila signaling pathways are highly conserved with those <strong>of</strong> mammals (Blenis, 1993; Tan<br />

and Kim, 1999). Among the many signal transduction pathways investigated, receptor tyrosine<br />

kinases are the best characterized. Unlike vertebrates, the Drosophila genome contains only one<br />

gene encoding each <strong>of</strong> the MAPK subfamilies and PKB/Akt1 (FlyBase), eliminating the<br />

possibility <strong>of</strong> functional redundancy due to the presence <strong>of</strong> multiple genes and is<strong>of</strong>orms for these<br />

kinases in vertebrates. Further, modulation <strong>of</strong> the inflammatory response to PQ exposure in<br />

mammalian models by these signaling pathways has not been reported.<br />

PQ toxicity is associated with nitric oxide (NO) production and is alleviated when NO<br />

synthesis is reduced by a competitive inhibitor <strong>of</strong> NOS, L-NAME (Djukic et al., 2007; <strong>Inamdar</strong><br />

et al., <strong>2009</strong>, under revision). Further, we have shown that minocycline suppresses the PQ-<br />

induced activation <strong>of</strong> NOS (<strong>Inamdar</strong> et al., <strong>2009</strong>, under revision).<br />

In this report, we demonstrate that minocycline prolongs survival <strong>of</strong> PQ-exposed adult<br />

Drosophila, diminishes PQ-induced mobility defects, blocks associated changes in the DA<br />

81


pathway, diminishes levels <strong>of</strong> reactive oxygen species (ROS), and prolongs DA neuron survival.<br />

We further sought to identify signaling pathways that mediate the PQ-induced toxicity in this in<br />

vivo Drosophila model, as well as the signaling pathways modifying the protective response <strong>of</strong><br />

anti-oxidant and anti-inflammatory drug, minocycline against PQ. Using loss <strong>of</strong> function mutant<br />

and over-expression transgenic lines, we found that JNK and Akt1 per se play an important role<br />

in protecting DA neurons against PQ toxicity while caspase (dronc, Drosophila homolog <strong>of</strong><br />

caspase) mediates PQ toxicity in Drosophila. We further report the identification <strong>of</strong> signaling<br />

pathways capable <strong>of</strong> modifying the neuroinflammatory response against PQ using<br />

pharmacological and genetic approaches.<br />

82


MATERIALS AND METHODS<br />

Drosophila strains and culture maintenance. Strains utilized in all experiments were<br />

Canton S, a wild type strain, Df(1)w, y, a white-eyed strain that carries otherwise wild type<br />

genes, and mutant alleles <strong>of</strong> the following signal transduction genes: rolled (rl 1 ) (Morgan et al.,<br />

1925), a weak loss-<strong>of</strong>-function allele <strong>of</strong> the gene encoding ERK, basket, bsk 1 /Cyo, a loss-<strong>of</strong>-<br />

function allele <strong>of</strong> the gene encoding JNK (Nüsslein-Volhard et al., 1984), PKB/Akt1, ry 506<br />

P{PZ}Akt 104226 /TM3, ry RK Sb 1 Ser 1 , carrying a loss-<strong>of</strong>-function mutation <strong>of</strong> Akt1 (Perrimon et al.,<br />

1996), a loss-<strong>of</strong>-function allele for the gene encoding Diablo/Smac, reaper, y 1 w 67c23 ; P{SUPor-<br />

P}KG07184 ry 506 (White et al., 1994), and a null mutant allele <strong>of</strong> Nc or Dronc, encoding a<br />

caspase, y 1 w*; Nc 51 /TM3, Sb 1 (Chew et al., 2004). Transgenic strains employed for wild type<br />

expression <strong>of</strong> kinases were: JNK (bsk), y 1 w 1118 ; P{UAS-bsk.A-Y}1 (Adachi-Yamada, personal<br />

communication to Bloomington Stock Center) and Akt1, y 1 w 1118 ; P{UAS-Akt1}/Cyo (from Dr.<br />

Tien Hsu, Medical <strong>University</strong> <strong>of</strong> South Carolina, Charleston, SC). All mutants and transgenic<br />

strains were mated to the appropriate wild type strain, and all assays were performed on mutants<br />

heterozygous for the wild type genes. All stocks were maintained at 25º C.<br />

<strong>The</strong> transgenic strain UAS-2X eGFP (Chromosome II) and the above mutant strains for<br />

signal transduction pathways were obtained from the Bloomington, IN Drosophila stock center<br />

and a TH-Gal4 strain (Friggi-Grelin et al., 2003) was obtained from Jay Hirsh (<strong>University</strong> <strong>of</strong><br />

Virginia).<br />

Feeding experiments. Separated male and female flies, 48-96 hr post-eclosion, were fed<br />

on filter paper saturated with one <strong>of</strong> the following solutions: 5% sucrose only or 5% sucrose<br />

with 1 or 10mM paraquat only, minocycline HCl only at varying concentrations, 10mM paraquat<br />

83


with 1mM minocycline HCl, 10mM doxycycline only or with 10mM paraquat. Feedings were<br />

continued for up to 48 hr. All chemicals were obtained from Sigma (St. Louis, MO).<br />

Locomotion assay. <strong>The</strong> mobility <strong>of</strong> adult male and female flies from each treatment<br />

group was assessed using a negative geotaxis climbing assay. A single fly was placed in an<br />

empty plastic vial, tapped to the bottom and the time required to climb 5 cm was recorded three<br />

times sequentially with 10 min rest periods between each measurement. Each replication value<br />

recorded is an average <strong>of</strong> the three trials; each assay was conducted on 10 flies per test group.<br />

HPLC analysis. Monoamine and pteridine levels were determined using an ESA<br />

CoulArray Model 5600A high performance liquid chromatography system. Fifty adult heads<br />

were extracted in 60 μl 0.1M perchloric acid, followed by centrifugation. Ten μl <strong>of</strong> each extract<br />

was injected per extract. Analyses were conducted on 3 extracted replicas <strong>of</strong> each test set.<br />

Amines and pteridines were separated on a Phenomenex Synergi 4μm Hydro-RP column (4.5 X<br />

150 mm) according to the method <strong>of</strong> McClung and Hirsh (1999). Separations were performed<br />

with isocratic flow at 1ml/min.<br />

Amines were detected with the ESA CoulArray electrochemical analytical call, Model<br />

5011 (channel 1 at -50 mV, channel 2 at 300 mV). Pteridines were detected with a Linear Model<br />

LC305 fluorescence detector (excitation wavelength 360 nm and emission wavelength 456 nm).<br />

Analysis was performed using ESA CoulArray s<strong>of</strong>tware.<br />

GTPCH assay. GTPCH activity was assayed by modification <strong>of</strong> a method described by<br />

Viveros et al. (1981). Thirty heads <strong>of</strong> 3-5 days old adult males were collected and homogenized<br />

84


in 100μl <strong>of</strong> lysis buffer (50mM Tris, 2.5 mM EDTA, pH 8.0). Extracts were centrifuged at<br />

10,000 rpm for 10 min and the protein concentrations <strong>of</strong> supernatants were determined using the<br />

BioRad Protein Assay Reagent. Seven µl <strong>of</strong> 2mM GTP and extract corresponding to 45 μg <strong>of</strong><br />

protein were brought to a volume <strong>of</strong> 70 μl. <strong>The</strong> mixture was incubated for 1 hr at 37° C to<br />

convert GTP to dihydroneopterin triphosphate (dNP3), followed by the addition <strong>of</strong> 30μl <strong>of</strong> 1%<br />

iodine and 2% potassium iodide in 1M HCl. <strong>The</strong> mixture was then incubated in the dark at room<br />

temperature for 1 hr, which terminates the reaction and oxidizes dNP3, converting it to a<br />

florescent form. <strong>The</strong> reaction was decolorized in 3% absorbic acid and centrifuged at 14,000 rpm<br />

for 5 min. <strong>The</strong> supernatant were dephosphorylated with 2 units <strong>of</strong> alkaline phosphatase (Roche)<br />

in 10X dephosphorylation buffer and incubated at 37°C for 1 hr. Neopterin peaks, which were<br />

detected by fluorescence at excitation wavelength <strong>of</strong> 353 nm and emission wavelength <strong>of</strong> 438<br />

nm, were identified and quantified relative to a neopterin (Sigma) standard.<br />

Confocal microscopy. Whole mounts <strong>of</strong> dissected brains from TH-Gal4; UAS-eGFP<br />

adults fed with sucrose alone, or with paraquat, as described in the Results were examined for<br />

dopaminergic neuron morphology and number, detected by visualizing GFP-expressing neurons.<br />

Each brain was scanned to include 15-18 sections for optimum visualization <strong>of</strong> DA neurons. <strong>The</strong><br />

Z-sections were then utilized to get the average <strong>of</strong> all sections using a Leica TCS SP2 AOBS<br />

confocal microscope (Wetzlar, Germany).<br />

Catalase assay. Separated males and female flies at 2-4 days post-eclosion were fed 5%<br />

sucrose alone, 10mM paraquat, 1mM minocycline or a combination <strong>of</strong> 10mM paraquat and 1mM<br />

minocycline in 5% sucrose solution for 24 hr prior to sacrifice. Crude enzyme extracts were<br />

85


prepared by homogenizing 10 heads from each treatment group in 150 μl 0.1M sodium–<br />

potassium phosphate buffer containing 0.1 M Triton X-100 (pH 7.0) following the method <strong>of</strong><br />

Bewley et al (1983). After homogenization, all the samples were incubated at 4ºC for 10 min<br />

prior to centrifugation at 12,000 x g for 10 min. Assays were performed at 30 ºC following the<br />

methods <strong>of</strong> Lubinsky and Bewley (1979). <strong>The</strong> reaction was initiated by adding 20 μl head tissue<br />

extract to the reaction mixture containing 100 μl (48.6mM) H2O2 in 0.1M sodium-potassium<br />

phosphate buffer (pH 6.8) to a final volume <strong>of</strong> 2.55 ml. <strong>The</strong> reaction <strong>of</strong> head extract with H2O2<br />

was determined at absorbance wavelength 230 nm and calculated using a molar extinction<br />

coefficient for H2O2 <strong>of</strong> 62.4. One unit <strong>of</strong> catalase activity was defined as 1μmole <strong>of</strong> H2O2<br />

decomposed per min. All values represent the average <strong>of</strong> 6-8 replications from independently<br />

prepared extracts.<br />

Lipid peroxidation assay. Head extracts were prepared from female flies at 2-4 days post-<br />

eclosion fed 5% sucrose alone, 10mM paraquat, 1mM minocycline or a combination <strong>of</strong> 10mM<br />

paraquat and 1mM minocycline in 5% sucrose solution after 24 hr <strong>of</strong> feeding were homogenized<br />

with 0.1M phosphate buffer and centrifuged for 10 min at 10,000g at 4°C. Two ml <strong>of</strong> reagent<br />

TCA-TBA (thiobarbituric acid)-HCl was added to 1ml <strong>of</strong> head extract and heated for 15 min in a<br />

boiling water bath to allow malondialdehyde, the product <strong>of</strong> the lipid peroxidase reaction, to<br />

develop a red chromophore, detected spectrophotometrically at 535 nm, as described by Beuge<br />

and Aust (1978).<br />

86


Statistical Analysis. All data were analyzed by one way ANOVA with Dunnett’s post test<br />

or by two-tailed Student’s t-test, assuming equal variances wherever applicable, using GraphPad<br />

Prism (San Diego, CA). Details <strong>of</strong> the analyses are described in the figure legends.<br />

87


RESULTS<br />

Effect <strong>of</strong> minocycline on paraquat-induced truncation <strong>of</strong> life span<br />

Minocycline has been reported to improve survival duration in animal models for<br />

amyotrophic lateral sclerosis, Huntington’s disease, and Parkinson’s disease (Hersch et al., 2003;<br />

Van Den Bosch et al., 2002; Quintero et al., 2005). However, this antibiotic has been reported to<br />

have detrimental effects in some systems (Diguet et al., 2004). <strong>The</strong>refore, we first assessed<br />

whether ingestion <strong>of</strong> minocycline alone, at concentrations ranging from 100μM to 50 mM,<br />

affected viability <strong>of</strong> adult Drosophila. <strong>The</strong> results for male flies are shown in Fig. 2.3A, Chapter<br />

2; females gave comparable results (data not shown).<br />

Employing non-toxic levels <strong>of</strong> minocycline, we then asked whether this antibiotic could<br />

rescue the toxic effects <strong>of</strong> PQ. We first co-fed minocycline doses from 100 μM to 1 mM with 10<br />

mM PQ and determined the average survival duration (Fig. 3.1 A). We found that co-feeding <strong>of</strong><br />

1 mM minocycline with 10 mM PQ extended average survival time an additional 48 hr, from<br />

three to five days (Fig. 3.1.A). Minocycline at concentrations <strong>of</strong> 500 μM and below were unable<br />

to modify the effects <strong>of</strong> 10 mM PQ. We then tested the efficacy <strong>of</strong> minocycline under three<br />

different regimens, co-feeding 10 mM PQ and 1 mM minocycline, pre-feeding minocycline for 2<br />

days prior to PQ exposure, and pre-feeding 10 mM PQ for 2 days prior to exposure to<br />

minocycline alone. We found that neither pre-feeding nor post-treatment <strong>of</strong> minocycline were<br />

able to modify survival duration <strong>of</strong> flies ingesting 10 mM PQ (data not shown). However,<br />

extending the minocycline pre-feeding period to 5 days resulted in extension <strong>of</strong> life span to<br />

almost the same degree as the co-feeding regimen (Fig. 2.3 B, Chapter 2). We also co-fed<br />

another semi-synthetic tetracycline derivative belonging to minocycline group, doxycycline, at 1<br />

mM concentration, to determine if the protective effect is a general property <strong>of</strong> tetracycline drugs<br />

88


or specific to minocycline. Although minocycline and doxycycline belong to semi-synthetic<br />

tetracycline group, minocycline possesses an extra diethylamino group at C7 and lacks<br />

functional group at C6 (Fig. 3.1 C, D) (Kim et al., <strong>2009</strong>). We tested both 10 mM and 5 mM<br />

doses for PQ. However, co-feeding doxycycline with 10 mM and 5 mM was unable to affect the<br />

survival duration <strong>of</strong> 10 mM (data not shown) and 5 mM PQ fed adults (Fig.3.1B).<br />

Thus, altogether the above data indicates that minocycline at lower doses is not toxic to<br />

flies and able to extend PQ-induced truncation <strong>of</strong> life span. It also suggests that this effect is not<br />

a general effect <strong>of</strong> the whole family <strong>of</strong> tetracycline molecules, since doxycycline could not<br />

provide such protection.<br />

89


A B<br />

Survival (days)<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

PQ 10mM<br />

PQ 10mM+Min 100 uM<br />

NS<br />

**<br />

PQ 10mM+Min 250 uM<br />

PQ 10mM+Min 500 uM<br />

PQ 10mM+Min 1mM<br />

C D<br />

Figure 3.1: Effect <strong>of</strong> minocycline and doxycycline with 10 mM paraquat on survival <strong>of</strong> adult<br />

male flies. (A) Effect <strong>of</strong> different doses <strong>of</strong> minocycline co-fed with 10mM PQ on the life span <strong>of</strong><br />

wild type adult males. Male flies were co-fed 100 µM, 250 µM, 500 µM and 1mM minocycline<br />

with 10mM PQ and their survival duration was monitored. Co-feeding <strong>of</strong> 1mM minocycline with<br />

10 mM PQ significantly extends the survival duration compared with PQ alone. Addition <strong>of</strong> 100<br />

µM, 250 μM and 500 µM minocycline with 10mM PQ had no effect on life span. ** represents<br />

the difference between PQ-fed flies and those fed PQ with 1mM minocycline at P< 0.005. (B)<br />

Effect <strong>of</strong> doxycycline on PQ-treated flies. Addition <strong>of</strong> doxycycline had no effect on survival<br />

duration suggesting that the action <strong>of</strong> minocycline is specific. NS= non-significant. Error bars<br />

represent the standard error <strong>of</strong> the mean. Each data point represents at least 10 replications <strong>of</strong> 15<br />

flies each. (C,D) Chemical structures <strong>of</strong> minocycline and doxycycline, respectively.<br />

90<br />

Survival (days)<br />

6 NS<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

PQ PQ+Doxycycline


Minocycline protects against PQ-induced mobility defects<br />

We employed a climbing assay to assess whether minocycline could provide rescue <strong>of</strong><br />

the mobility deficits induced by PQ, similar to the improvement in motor performance after<br />

minocycline treatment reported in ALS and Huntington’s disease models (Kriz et al., 2002; Zhu<br />

et al., 2002b; Hersch et al., 2003) and in a 6-OHDA PD model (Quintero et al., 2003). Within 24<br />

hr <strong>of</strong> the initiation <strong>of</strong> 10 mM PQ feeding in the absence <strong>of</strong> minocycline, tremors and<br />

bradykinesia were apparent. At 48 hr, these flies were unable to climb and appeared frozen at<br />

the bottom <strong>of</strong> the vial. Within 72-84 hr, 40-50% mortality was observed. In contrast, when PQ<br />

was co-fed with 1 mM minocycline, no movement defects were apparent until after 72 hr,<br />

establishing that minocycline is capable <strong>of</strong> rescuing the behavioral syndrome associated with PQ<br />

ingestion (Fig.3.2).<br />

91


Time to cross 5 cm distance (sec)<br />

13<br />

12<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

**<br />

#<br />

24 hrs 48 hrs<br />

Figure 3.2: Minocycline protects against PQ-induced mobility defects. <strong>The</strong> ability <strong>of</strong><br />

minocycline to prevent the PQ-induced mobility defect was determined by negative geotaxis<br />

assay. <strong>The</strong> time required by 10 flies to cross 5 cm distance at 24 and 48 hr after the initiation <strong>of</strong><br />

ingestion <strong>of</strong> 10mM PQ and 10mM PQ with 1mM minocycline in female flies. <strong>The</strong> ingestion <strong>of</strong><br />

1mM minocycline has no effect on mobility, while PQ ingestion alone adversely affects<br />

mobility. Co-feeding minocycline with PQ results in mobility performance near control levels.<br />

Ten flies were scored per replication and the mean values represent the average <strong>of</strong> 15<br />

independent replications. <strong>The</strong> * and # indicate the significance <strong>of</strong> differences between control<br />

and PQ-fed flies, and between PQ only and co-fed flies, respectively. ***/###=P< 0.001 ** = P<br />

< 0.005 and # = P < 0.05. Error bars represent standard error <strong>of</strong> the mean. Each data point<br />

represents at least 10 replications <strong>of</strong> 15 flies each.<br />

92<br />

***<br />

###<br />

Control<br />

PQ<br />

PQ+ Min<br />

Min


Minocycline delays PQ-induced loss <strong>of</strong> dopaminergic neurons<br />

We recently established that the onset <strong>of</strong> movement dysfunction upon PQ ingestion<br />

coincides with the loss <strong>of</strong> specific subsets dopaminergic neurons in the adult brain (Chaudhuri et<br />

al., 2007). In light <strong>of</strong> the ability <strong>of</strong> minocycline to ameliorate PQ-induced tremors and mobility<br />

deficits, we next asked whether this effect is mediated through protection <strong>of</strong> at-risk dopaminergic<br />

neurons. Dopaminergic neurons were detected by the TH promoter-directed expression <strong>of</strong> GFP<br />

in the transgenic strain, TH-Gal4; UAS-eGFP (Friggi-Grelin et al., 2003). We compared<br />

dopaminergic neuron morphology and numbers in brains dissected every 24 hr after the initiation<br />

<strong>of</strong> feeding 5% sucrose only, 10 mM PQ alone, 10 mM minocycline alone or PQ in combination<br />

with 1 mM minocycline (Fig.3.3). As we had observed previously (using both the GFP reporter<br />

and immunolocalization <strong>of</strong> TH to identify dopaminergic neurons), these neurons display<br />

characteristic patterns <strong>of</strong> neuronal sensitivity. Upon exposure <strong>of</strong> 10 mM PQ for 24 hr, we<br />

observed that the PAL subgroup in the anterior region and the PPL1, PPM2 and PPM3<br />

subgroups in the posterior brain exhibited statistically significant neuron loss relative to controls.<br />

In the animals that were co-fed PQ and minocycline, there was no neuron loss in the PPM1 or<br />

PPL2 subgroups at 24 hr, and neuron numbers and morphology in other clusters were almost<br />

identical to those in control brains (Fig. 3.4). After 48 hr <strong>of</strong> PQ feeding, previously affected<br />

clusters continued to deteriorate, while neurodegeneration was noted in the previously unaffected<br />

PPM1 and PPL2 clusters (data not shown). At this time point, flies that ingested minocycline<br />

with PQ exhibited loss <strong>of</strong> neurons, especially PAL (data not shown). <strong>The</strong>refore, we conclude that<br />

the extension <strong>of</strong> life span observed when flies were fed minocycline along with PQ correlates<br />

with both delay <strong>of</strong> the onset <strong>of</strong> movement deficits and protection against PQ-induced<br />

neurodegeneration.<br />

93


A PPM2<br />

B<br />

Control<br />

C PPM2<br />

D<br />

PQ<br />

Figure 3.3: Minocycline confers protection to dopaminergic neurons. (A-D) <strong>The</strong> effect <strong>of</strong> 24 hr<br />

exposure <strong>of</strong> sucrose, 1mM minocycline alone, 10mM paraquat alone, and 10mM paraquat co-fed<br />

with 1mM minocycline on the dopaminergic neurons <strong>of</strong> TH-GAL4; UAS-eGFP adult brain. <strong>The</strong><br />

inset in each image demonstrates the change in the morphology and number <strong>of</strong> the PPM2<br />

subgroup <strong>of</strong> neurons. <strong>The</strong> exposure to minocycline alone (B) does not induce any change in<br />

number and morphology <strong>of</strong> the dopaminergic neuron when compared to the control image in<br />

Panel A. <strong>The</strong> addition <strong>of</strong> minocycline to PQ (D) delays the loss and alteration in the shape and<br />

neuronal process <strong>of</strong> dopaminergic neuron as opposed to paraquat only (C). Scale bar for A-<br />

D=100 μm.<br />

94<br />

PPM2<br />

PPM2<br />

Min<br />

PQ+Min


No <strong>of</strong> neurons<br />

16<br />

14<br />

12<br />

10<br />

8<br />

6<br />

4<br />

2<br />

0<br />

* #<br />

*<br />

#<br />

PAL PPM1 PPM2 PPM3 PPL1 PPL2<br />

Subgroups <strong>of</strong> DA neurons<br />

*<br />

Figure 3.4: Minocycline delays paraquat induced selective loss <strong>of</strong> dopaminergic neurons. <strong>The</strong><br />

average number <strong>of</strong> neurons per subset was determined 24 hr after the initiation <strong>of</strong> feeding. All<br />

scoring was done on TH-Gal4:UAS-GFP adults. Each subset <strong>of</strong> dopaminergic neurons was<br />

scored separately (n=15-25). PAL, PPM2, PPM3 and PPL1 subsets show significant reduction in<br />

the number <strong>of</strong> neurons in the PQ-treated groups, while the number <strong>of</strong> corresponding neurons in<br />

the co-fed animals show significantly elevated neuron numbers when compared to the brains <strong>of</strong><br />

animals fed PQ only, except in the most sensitive cluster, PPL1. No significant difference in the<br />

neuron counts was noted between control, minocycline only, and co-fed individuals. <strong>The</strong><br />

significance <strong>of</strong> difference in each neuron cluster between the PQ-treated and control groups, and<br />

between the PQ-treated and co-fed groups are indicated as * and #, respectively, where * = P<<br />

0.05 and #= P < 0.05. Error bars represent standard error <strong>of</strong> the mean. Each data point represents<br />

at least 10 replications <strong>of</strong> 15 flies each.<br />

95<br />

#<br />

*<br />

Control<br />

PQ<br />

PQ+Min<br />

Min


Minocycline blocks changes in DA pathway components indicative <strong>of</strong> PQ-induced oxidative<br />

stress<br />

<strong>The</strong> production <strong>of</strong> DA is rate-limited by two enzymes; tyrosine hydroxylase (TH), which<br />

converts tyrosine to L-DOPA, and GTP cyclohydrolase (GTPCH), which is rate-limiting for the<br />

production <strong>of</strong> tetrahydrobiopterin (BH4), a c<strong>of</strong>actor for and regulator <strong>of</strong> TH catalysis. We<br />

previously observed that sensitivity to PQ is at least partially defined by the activity <strong>of</strong> the BH4<br />

and DA biosynthesis pathways (Chaudhuri et al., 2007). Although the precise mechanism<br />

remains unclear, we found that mutants synthesizing abnormally low amounts <strong>of</strong> BH4 and DA<br />

and wild type flies in which DA was depleted pharmacologically exhibited increased sensitivity<br />

to PQ. Conversely, mutants with elevated pools <strong>of</strong> DA and wild type flies that have ingested L-<br />

DOPA or DA exhibit increased resistance to PQ. Moreover, we found that following PQ<br />

ingestion, but prior to loss <strong>of</strong> dopaminergic neurons (and the accompanying decrease in BH4 and<br />

DA levels and corresponding increase in their oxidative products), there is a transient stimulation<br />

<strong>of</strong> DA pathway activity. <strong>The</strong> observed protection by minocycline against the effects <strong>of</strong> PQ might<br />

be mediated through interactions with the DA homeostasis machinery in at least two possible<br />

ways. Minocycline itself might stimulate the activity <strong>of</strong> the DA biosynthetic pathway and<br />

homeostasis components, creating a cellular environment that emulates those generated<br />

genetically or pharmacologically. Alternatively, minocycline, which reportedly can act as a<br />

scavenger <strong>of</strong> ROS (Kraus et al., 2006), might prevent the PQ-induced oxidative depletion <strong>of</strong> DA,<br />

which is expected to elevate oxidative load. As shown in Fig. 5, ingestion <strong>of</strong> minocycline alone<br />

for 24 hr has no significant effect on the production <strong>of</strong> pathway metabolites or on GTPCH<br />

activity, ruling out the possibility that minocycline activates DA synthesis. As observed in<br />

Chaudhuri et al. (2007), ingestion <strong>of</strong> PQ alone resulted in dynamic changes <strong>of</strong> enzyme activity,<br />

pathway products and oxidative products. After 24 hr <strong>of</strong> PQ exposure, L-DOPA pools are<br />

96


significantly elevated, relative to controls, indicating an increase in TH activity; however, the<br />

oxidative environment results in depletion <strong>of</strong> DA and a corresponding increase in the oxidative<br />

product, DOPAC (Fig. 3.5 A, B). Similarly, GTPCH activity increases (Fig. 3.5 C), and the<br />

oxidized product biopterin increases in concentration as BH4 pools are diminished (Fig.3.5 A).<br />

In contrast, when minocycline was co-fed with PQ for 24 hr, the effect <strong>of</strong> PQ on each <strong>of</strong> these<br />

components was significantly less severe. Stimulation <strong>of</strong> GTPCH and TH activity with these<br />

components was minimal and levels <strong>of</strong> the oxidized products, biopterin and DOPAC, were<br />

indistinguishable from those in sucrose only or minocycline only controls (Fig.3.5 A, B, C).<br />

97


Figure 3.5: Minocycline blocks changes in DA pathway components indicative <strong>of</strong> PQ-induced<br />

oxidative stress. Minocycline blocks the changes induced by PQ the DA and BH4 biosynthesis<br />

pathways. (A) Changes in the DA and BH4 metabolites after 24 hr <strong>of</strong> exposure <strong>of</strong> adult males to<br />

PQ and PQ with minocycline. <strong>The</strong> addition <strong>of</strong> minocycline prevents the reduction in the PQinduced<br />

DA and BH4 levels. <strong>The</strong> increase in L-DOPA levels, indicative <strong>of</strong> PQ-stimulated TH<br />

activity, is reduced by minocycline. (B) <strong>The</strong> oxidized DA metabolite,DOPAC, is elevated by PQ<br />

exposure and is significantly decreased by the co-feeding <strong>of</strong> minocycline to male adults for 24<br />

hr. (C) <strong>The</strong> compensatory increase in GTPCH activity in PQ-fed adult males is reduced in PQminocycline<br />

co-fed males after the 24 hr <strong>of</strong> ingestion. <strong>The</strong> significance <strong>of</strong> differences in each<br />

subset between the PQ-treated and control groups, and PQ-treated and co-fed groups are<br />

indicated as * and # respectively where * and #= P < 0.05, ** and ## = P < 0.005. Error bars<br />

represent the standard error <strong>of</strong> the mean. Each data point represents at least 10 replications <strong>of</strong> 15<br />

flies each.<br />

98


A<br />

B<br />

C<br />

DOPAC<br />

ng/head<br />

ng/head<br />

2.25<br />

2.00<br />

1.75<br />

1.50<br />

1.25<br />

1.00<br />

0.75<br />

0.50<br />

0.25<br />

0.00<br />

0.0175<br />

0.0150<br />

0.0125<br />

0.0100<br />

0.0075<br />

0.0050<br />

0.0025<br />

0.0000<br />

GTPCH activity<br />

(nmole/min/mg)<br />

*<br />

#<br />

**<br />

#<br />

L-Dopa Dopamine BH4 Biopterin<br />

Control<br />

*<br />

PQ<br />

9 **<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

99<br />

*<br />

#<br />

PQ+Min<br />

#<br />

*<br />

Min<br />

Control PQ PQ+Min Min<br />

#<br />

Control<br />

PQ<br />

PQ+Min<br />

Min


Minocycline reduces PQ-generated reactive oxygen species<br />

<strong>The</strong> ability <strong>of</strong> minocycline to block the PQ-induced changes in the DA and BH4<br />

biosynthesis pathways indicative <strong>of</strong> oxidative stress led us to hypothesize that minocycline was<br />

serving principally as a scavenger <strong>of</strong> PQ- generated ROS in Drosophila. In order to test this<br />

hypothesis, we assayed several markers <strong>of</strong> PQ-induced oxidative stress that are known to be<br />

elevated in mammalian systems. Elevation in malondialdehyde, formed from the breakdown <strong>of</strong><br />

polyunsaturated fatty acids and the accompanying increase lipid peroxidase level, is an important<br />

and specific cellular response <strong>of</strong> PQ-mediated toxicity due to the ability <strong>of</strong> PQ to produce<br />

hydroxyl radicals which react with cellular membranes (Bus et al., 1974). <strong>The</strong>refore, we<br />

employed a lipid peroxidation assay to test the ability <strong>of</strong> minocycline to specifically counteract<br />

the PQ toxicity in Drosophila. As seen in Fig. 3.6A, PQ ingestion results in a two-fold elevation<br />

<strong>of</strong> lipid peroxidation within the first 24 hr. Co-feeding <strong>of</strong> minocycline with PQ almost<br />

completely blocks these indicators <strong>of</strong> lipid damage; lipid peroxidation levels in these flies were<br />

indistinguishable from levels in sucrose only and minocycline only controls. Stimulation <strong>of</strong><br />

catalase activity in neural cell cultures (Röhrdanz et al., 2001; Yang and Tiffany-Castiglioni.,<br />

2005) and in Drosophila (Chaudhuri et al., 2007) is another indicator <strong>of</strong> elevated ROS. We,<br />

therefore, compared catalase activities in the heads <strong>of</strong> PQ only and PQ and minocycline co-fed<br />

flies. Elevated catalase activities were detected in the heads <strong>of</strong> both groups (Fig. 3.6 B);<br />

however, the catalase activity in flies that had ingested minocycline with PQ was significantly<br />

lower than those exposed to PQ only. <strong>The</strong>se results strongly suggest that minocycline has a<br />

strong ROS suppression capacity.<br />

100


A<br />

B<br />

Lipid peroxidation<br />

Moles/mg head tissue(x10 -8 )<br />

Catalase Specific activity<br />

Units (10 6 )/mg protein<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

5<br />

4<br />

3<br />

2<br />

1<br />

0<br />

**<br />

Control PQ PQ+ Min Min<br />

**<br />

Figure 3.6: Minocycline reduces PQ-generated reactive oxygen species. (A) <strong>The</strong> level <strong>of</strong><br />

malondialdehyde, formed from the breakdown <strong>of</strong> polyunsaturated fatty acids by PQ, was<br />

determined by adding thiobarbituric acid to head extracts <strong>of</strong> males that had ingested PQ or PQ<br />

with minocycline for 24 hr. <strong>The</strong> reaction results in a red colored product absorbing at 535nm.<br />

Minocycline significantly decreases the amount <strong>of</strong> lipid peroxidation induced by PQ. (B)<br />

Minocycline reduces the specific activity <strong>of</strong> catalase after 24 hr <strong>of</strong> ingestion in co-fed male flies.<br />

Specific activity is expressed in 10 6 units/mg protein, where 1 unit is defined as 1 μmole <strong>of</strong> H2O2<br />

decomposed per minute. <strong>The</strong> significance <strong>of</strong> differences between the PQ-treated and control<br />

groups, and between the PQ-treated and co-fed groups was indicated as * and #, respectively,<br />

where * and # = P < 0.05. Error bars represent standard error <strong>of</strong> the mean. <strong>The</strong> experiments were<br />

done as 5-8 replicas <strong>of</strong> 10 head extracts.<br />

101<br />

##<br />

Control PQ PQ+Min Min<br />

#


Loss <strong>of</strong> function mutants <strong>of</strong> the genes encoding JNK and Akt are sensitive to PQ but involvement<br />

<strong>of</strong> reaper, caspase and rolled in PQ-induced toxicity was not detected<br />

We next used this PD model to investigate the signaling pathways associated with PQ<br />

toxicity. Specifically, we tested JNK, encoded by the gene basket and Akt, associated with PKB<br />

signaling pathway and encoded by Akt1. In addition, rolled, dronc and reaper which encode<br />

ERK, caspase-9 and the pro-apoptotic protein, reaper, respectively, were tested. Mutations in<br />

rolled gene are homozygous viable while others are homozygous lethal. <strong>The</strong>se kinases function<br />

in myriad biological processes, but are particularly known to respond to various cellular stresses.<br />

We hypothesized that these kinases also play key roles in the DA associated toxic response<br />

triggered by PQ. To test this hypothesis, we exposed heterozygous loss-<strong>of</strong>-function mutants,<br />

bsk 1 , Akt 1 , rolled, dronc and reaper, to 1 mM PQ alone and to a combined dose <strong>of</strong> 1mM PQ and<br />

1mM minocycline. When compared to wild type flies, heterozygous bsk 1 and Akt 1 mutants<br />

showed increased sensitivity to PQ and died on average 2 days before wild type flies (Fig. 3.7).<br />

However, heterozygous reaper and rolled mutants were indistinguishable from the wild type<br />

controls for the effect <strong>of</strong> PQ on their survival. In contrast, the heterozygous caspase mutants,<br />

dronc, survived 2 days more than the wild type controls on PQ (Fig.3.7). Increasing the dose <strong>of</strong><br />

PQ to 10 mM did not alter the outcome for these mutant strains (data not shown). Further, we<br />

found that, unlike wild type flies, the lifespan <strong>of</strong> bsk 1 and Akt 1 mutants exposed to PQ could not<br />

be extended by minocycline treatment, suggesting that JNK and Akt signaling pathways are<br />

important in the protective response <strong>of</strong> minocycline in flies. In addition, the result shows lack <strong>of</strong><br />

regulatory effects by ERK and reaper on PQ toxicity since these heterozygous mutants were<br />

indistinguishable from wild type flies with respect to sensitivity to paraquat and rescue by<br />

minocycline (Fig. 3.7).<br />

102


Figure 3.7: Paraquat induced inflammatory response is regulated by JNK and Akt signaling<br />

pathways. Effect <strong>of</strong> 1 mM PQ and 1 mM minocycline co-fed with 1 mM PQ on loss-<strong>of</strong>-function mutants,<br />

JNK/+ (bsk 1 /+), Akt1/+, reaper/+ , caspase/+ (dronc/+), ERK/+ (rolled/+) . rolled (ERK), reaper and<br />

caspase mutants showed an extension <strong>of</strong> life span with minocycline treatment, while the survival <strong>of</strong> bsk 1<br />

(JNK) and Akt 1 mutants was unmodified in the presence <strong>of</strong> minocycline. **= P < 0.005 represents<br />

significant difference between PQ, and PQ with minocycline fed groups. ##=P< 0.005 and shows<br />

significant difference between PQ-fed wild type, and PQ-fed bsk and Akt loss-<strong>of</strong>-function mutants NS=<br />

not significant. Error bars represent standard error <strong>of</strong> the mean. n= 180 and each data point represents at<br />

least three independent replications <strong>of</strong> 50-60 flies each.<br />

103


JNK and Akt confer protection against paraquat-induced toxicity in dopaminergic neurons<br />

<strong>The</strong> observations that JNK and Akt loss-<strong>of</strong>-function heterozygous mutants increase the<br />

sensitivity to PQ and that these genes are important for minocycline mediated protective<br />

responses against PQ led us to test whether this effect is reversed when JNK and Akt are over-<br />

expressed. Since PQ toxicity is associated with DA (Peng et al., 2004), we first drove the<br />

expression <strong>of</strong> JNK and Akt in DA neurons using the GAL4-UAS system (Brand and Perrimon,<br />

1993). Expression <strong>of</strong> JNK and Akt in dopaminergic neurons resulted in a two-fold increase in the<br />

survival duration in the presence <strong>of</strong> PQ (Fig. 3.8). Furthermore, over-expression <strong>of</strong> JNK and Akt<br />

in DA neurons resulted in protection by minocycline against PQ. <strong>The</strong> exposure <strong>of</strong> 1 mM<br />

minocycline with 10 mM PQ improved the lifespan by 30% compared to 10 mM PQ alone in<br />

control and over-expressed strains. <strong>The</strong>se results suggest that both Akt and JNK have pro-<br />

survival function in PQ-induced DA toxicity.<br />

104


Figure 3.8: Over-expression <strong>of</strong> wild type JNK and Akt provide protection against PQ. Adult<br />

males <strong>of</strong> genotypes TH-GAL4/+, UAS-bsk 1 /+, UAS-Akt 1 /+, TH-GAL4;UAS-bsk 1 /+ and TH-GAL4;<br />

UAS-Akt 1 /+ flies were fed, beginning at 48 hr post-eclosion, 10 mM PQ or 10 mM PQ and 1 mM<br />

minocycline. <strong>The</strong> average survival duration for each group was determined. Expression <strong>of</strong> JNK<br />

and Akt in TH neurons increased the survival duration by two fold with respect to control flies.<br />

** =P < 0.005 and represent significant differences between the PQ and PQ with minocycline groups.<br />

##=P< 0.005, indicating a significant difference between control and JNK or Akt expressing flies fed only<br />

PQ. Error bars represent standard error <strong>of</strong> the mean. Each data point represents at least three independent<br />

replications <strong>of</strong> 50-60 flies each.<br />

105


DISCUSSION<br />

Minocycline imparts anti-oxidant effects in a paraquat induced Drosophila model for<br />

Parkinson’s disease<br />

PQ is considered an oxidative stressor, generating super oxide and hydroxyl radicals (Bus<br />

et al., 1974). Moreover, epidemiological and experimental studies point to PQ as an etiological<br />

agent for PD (Dinis-Oliveira et al., 2006). We have used PQ ingestion to establish an in vivo<br />

Drosophila PD model (Chaudhuri et al., 2007). This model was employed to study the gene-<br />

environmental interactions for DA regulatory genes regulating the dopamine biosynthesis and<br />

homeostasis that modified susceptibility to PQ.<br />

Minocycline, a second generation semi-synthetic antibiotic derived from tetracycline, is<br />

gaining attention due to its anti-inflammatory and anti-oxidant properties in numerous<br />

neurodegenerative and injury induced mammalian models such as such as cerebral ischemia,<br />

traumatic brain injury, amyotrophic lateral sclerosis, Parkinson's diseases, kainic acid treatment,<br />

Huntington' disease, multiple sclerosis and Alzheimer’s disease (Jordan et al., 2007; Kim and<br />

Suh, <strong>2009</strong>). Tetracyclines are available in three groups, tetracycline as original, semi-synthstic<br />

compounds, and modified tetracyclines. Minocycline and doxycycline belong to the semi-<br />

synthetic group. Minocycline differs from doxycycline due to the presence <strong>of</strong> diethylamino<br />

group at C7 carbon ring and lack <strong>of</strong> a functional group at C6 (Fig. 3.1 C, D). <strong>The</strong> hydroxyl group<br />

at C10 and diethylamino group at C7 on the fourth carbon ring have been proposed to play a<br />

crucial role in scavenging free radicals (Kraus et al., 2005; Kim and Suh, <strong>2009</strong>). Moreover,<br />

minocycline has been found to be a more potent radical scavenger than doxycycline in Fe 3+<br />

induced oxidative stress in in vitro neuron culture (Kraus et al., 2005). Although the exact<br />

biological targets for minocycline are still not well known, certain results have demonstrated that<br />

minocycline causes the inhibition <strong>of</strong> the mitochondrial permeability-transition mediated<br />

106


cytochrome c release from mitochondria, the inhibition <strong>of</strong> caspase-1 and -3 expression, and the<br />

suppression <strong>of</strong> microglial activation (Chen et al., 2000; Wu et al., 2002; Zhu et al., 2002b).<br />

More recently, controversial studies on the protective effects <strong>of</strong> minocycline in various<br />

disease models have been reported. Diguet et al. (2004) reported that the protective or deleterious<br />

effects <strong>of</strong> minocycline depend on the mode <strong>of</strong> administration and dose <strong>of</strong> minocycline. Along the<br />

same line, we first tested different doses <strong>of</strong> minocycline ranging from 100 µm to 50 mM to wild<br />

type flies and conducted experiments subsequently using 1 mM minocycline as this was a highly<br />

efficient and non-toxic dose in wild type Drosophila (Fig. 2.3 A, Chapter 2). We found that this<br />

concentration <strong>of</strong> minocycline rescued the effects <strong>of</strong> 10 mM PQ (Fig.3.1 A), and that pre-feeding<br />

and co-feeding regimens for minocycline were equally protective (Fig. 2.3 B, Chapter 2). In<br />

contrast, doxycycline could not extend the PQ induced truncation <strong>of</strong> life span, suggesting the<br />

specificity <strong>of</strong> the protective role <strong>of</strong> minocycline in this PD model mainly due to favorable<br />

chemical groups at fourth carbon ring (Fig. 3.1 B, C, D). Bonelli et al. (2006) also reported the<br />

prevention <strong>of</strong> PQ- induced reduction <strong>of</strong> survival duration in Drosophila but the mechanism<br />

through which minocycline imparts this effect in Drosophila was not investigated. We first<br />

assessed the behavioral, neurochemical and neuroprotective abilities <strong>of</strong> minocycline.<br />

Minocycline delayed PQ induced mobility defects and the loss <strong>of</strong> dopaminergic neurons (Fig. 3.2<br />

and Fig. 3.3). Further, this neuroprotective effect was observed in all the dopaminergic<br />

subgroups found in Drosophila (Fig. 3.4). Similar protective effects <strong>of</strong> minocycline have been<br />

reported with respect to different PD mimetics in mammalian in vivo and in vitro models (Wu et<br />

al., 2002; Zhu et al., 2002b; Lin et al., 2003; Wang et al., 2003a). We also assessed the<br />

antioxidant effects <strong>of</strong> minocycline and detected a reduction in the generation <strong>of</strong> ROS as assayed<br />

by changes in catalase activity and lipid peroxidation (Fig. 3.6 A, B). We have shown previously<br />

107


that PQ modulates the DA biosynthesis pathway by inducing rapid oxidative turn-over <strong>of</strong> the<br />

products (Fig. 3.5 and Chaudhuri et al. 2007). Our current results indicate that minocycline<br />

prevents the rapid turn-over <strong>of</strong> key DA pathway metabolites induced by PQ and thus acts as a<br />

strong anti-oxidant (Fig. 3.5).<br />

We recently found that minocycline causes suppression <strong>of</strong> PQ-mediated increase in NOS<br />

activity in the adult fly head (<strong>Inamdar</strong> et al., <strong>2009</strong>, under revision). After this initial validation <strong>of</strong><br />

the anti-oxidant in addition to NOS-associated anti-inflammatory properties <strong>of</strong> minocycline<br />

against PQ in Drosophila, we then turned to an investigation <strong>of</strong> the potential involvement <strong>of</strong><br />

signal transduction pathways associated with PQ-induced in vivo PD model.<br />

Identification <strong>of</strong> signal transduction pathways mediating PQ-induced neurotoxic and<br />

neuroinflammatory responses in Drosophila<br />

Our investigation <strong>of</strong> signaling pathways mediating PQ-induced neurotoxic and NOS<br />

associated neuroinflammatory response in Drosophila is unique in that it could be get insight<br />

into signal transduction pathway involved in PQ mediated toxicity at the whole organism level.<br />

Most <strong>of</strong> the previous mammalian studies have utilized in vitro (i.e., cell culture) approaches to<br />

address this question. Moreover, these mammalian studies have used pharmacological inhibitors<br />

to block the proposed functions <strong>of</strong> the signaling pathway, which may lack complete specificity <strong>of</strong><br />

function. Finally, the variations in the type <strong>of</strong> inhibitor used, inhibitor concentrations, time <strong>of</strong><br />

addition, as well as cell lines may affect the outcome <strong>of</strong> these experiments (Sekiguchi et al.,<br />

1999; Learish et al., 2000; Waetzig and Herdegen, 2005).<br />

We have tested the role <strong>of</strong> kinases known to be functionally conserved with mammals in<br />

PQ-induced Drosophila PD. We found that heterozygous loss-<strong>of</strong>-function mutants for JNK,<br />

ERK, Akt, caspase and reaper (pro-apoptosis gene) show differential responses to PQ. <strong>The</strong><br />

108


heterozygous loss-<strong>of</strong>-function mutants for JNK/bsk and Akt1exhibit extreme sensitivity to PQ<br />

and fail to respond to minocycline, indicating crucial roles <strong>of</strong> these signaling pathways in either<br />

the anti-inflammatory or neuronal stress responses to PQ. On the contrary, heterozygous loss-<strong>of</strong>-<br />

function mutants for ERK and reaper mutants were equivalent to wild type flies in their<br />

sensitivity to PQ (Fig. 3.7).<br />

In Drosophila, Akt1, which encodes phosphoinositide-3-OH-kinase-dependent<br />

serine/threonine protein kinase, affects cell and organ size in a cell autonomous manner (Verdu<br />

et al., 1999). Akt1 has been shown to be regulated via the Drosophila PI(3)K, in a mechanism<br />

similar to that <strong>of</strong> its mammalian homolog, indicating the functional conservation <strong>of</strong> Akt1’s mode<br />

<strong>of</strong> action. In mammals, Akt1 plays a crucial role in cell survival. It is regulated by the PI3K-<br />

mediated signaling pathway and negatively regulates JNK and caspase-mediated apoptosis<br />

(Burke, 2007). Deregulation <strong>of</strong> Akt1 mediated signaling pathway has been well documented in<br />

familial and sporadic forms <strong>of</strong> PD models (Dudek et al., 1997; Yang et al., 2005; Rodriguez-<br />

Blanco et al., 2008; Xiromerisiou et al., 2008). Stimulation <strong>of</strong> the Akt1 signaling pathway in in<br />

vitro or in vivo models resulted into neurotrophic, anti-apoptotic effects (Ries et al., 2006; Burke,<br />

2007). Yang et al. (2005) found the suppression <strong>of</strong> ROS and survival <strong>of</strong> DA neurons in<br />

transgenic strains over-expressing Drosophila Akt1 in DJ1 RNAi strains. <strong>The</strong>refore, our<br />

preliminary results indicate that, like mammalian Akt1, Drosophila Akt1 appears to have<br />

functions in promoting survival <strong>of</strong> DA neurons in PQ-induced Drosophila PD model (Fig. 3.7,<br />

Fig. 3.8).<br />

In mammalian PD models, JNK has been documented to initiate PCD (programmed cell<br />

death) by inactivating anti-apoptotic protein Bcl-xl. Moreover, phospho-JNK is detected in DA<br />

neurons in MPTP and 6-OHDA- induced PD model (Gearan et al., 2001; Ganguly et al., 2004).<br />

109


Furthermore, Peng et al. (2004) detected up-regulation <strong>of</strong> phospho-JNK in TH neurons in a PQ-<br />

induced mammalian PD model. Similarly, activated JNK has been detected in parkin mutants in<br />

Drosophila (Yang et al., 2005). However, in Drosophila, JNK, encoded by bsk, has also been<br />

reported to play an important role in morphogenesis and innate immune response (Noselli and<br />

Agnès, 1999; Boutros et al., 2002). Moreover, Wang et al. (2003b) also demonstrated important<br />

roles for JNK in longevity and resistance to PQ induced-oxidative stress. Our preliminary data<br />

suggest a specific role <strong>of</strong> JNK in the survival response <strong>of</strong> DA neurons in our PD model and could<br />

raise the possibility that JNK may function in both neurons and innate immune cells in PQ<br />

response (Fig. 3.7, Fig. 3.8).<br />

ERK is proposed to play an important neuroprotective role in PD (Cavanaugh et al.,<br />

2006). However, there are controversies since evidence for a neurodegenerative role <strong>of</strong> ERK in<br />

PD also has been reported (Chu et al., 2004). <strong>The</strong> elucidation <strong>of</strong> a direct functional role for ERK<br />

in in vivo PD models has not been reported. We, therefore, used the heterozygous mutant form<br />

for ERK/rolled in PQ- induced PD model to investigate potential functions, and we found that<br />

heterozygous loss-<strong>of</strong>-function mutant form for ERK lack any detectable functional involvement<br />

in our model, although the result could also infer the lack <strong>of</strong> sufficient knock-down due to<br />

heterozygous strain (Fig. 3.7). It would be interesting to test for the dominant negative form for<br />

ERK in DA neurons to correlate direct role <strong>of</strong> ERK in our PD model.<br />

We further analyzed the role <strong>of</strong> PCD in PQ neurotoxicity by testing the heterozygous<br />

mutants for pro-apoptotic gene, reaper and PCD initiator, caspase-9 ortholog, dronc. We found<br />

that the in heterozygous loss-<strong>of</strong>-function mutants for reaper and dronc, reaper mutants lack<br />

comparable survival to wild type flies on PQ, while dronc mutants showed increase in lifespan.<br />

<strong>The</strong>se results suggest that the activation <strong>of</strong> caspase mediates apoptosis/PCD in PQ toxicity which<br />

110


lack direct interaction with reaper gene known to inhibit the Inhibitor <strong>of</strong> Apoptosis (IAPs) in<br />

Drosophila and mammals (Fig. 3.7) (Steller, 2008).<br />

Thus, our preliminary results here provide in vivo evidence for essential role for kinases<br />

in a PQ induced PD model. Furthermore, we performed primary screening using minocycline, an<br />

agent known to possess anti-inflammatory and anti-oxidant in mammalian and Drosophila model<br />

for PD in wild type flies (<strong>Inamdar</strong> et al., <strong>2009</strong>, under revision). Further, we have detected the<br />

modification <strong>of</strong> protective properties <strong>of</strong> minocycline by DA regulatory genes (<strong>Inamdar</strong> et al.,<br />

<strong>2009</strong>, under revision). In this report, we present data showing the failure <strong>of</strong> heterozygous Akt1<br />

and JNK loss-<strong>of</strong>-function mutants to respond to minocycline, suggesting that minocycline might<br />

act via these signaling pathways. However, over-expression <strong>of</strong> wild type Akt1 and JNK in DA<br />

neurons does not provide additional levels <strong>of</strong> protection. Thus, these pathways may function<br />

parallel to minocycline, or alternatively, their role in the minocycline response is mediated in a<br />

cell type other than dopaminergic neurons (Fig. 3.8). We must perform genetic interactions<br />

between Atk1 and JNK signaling pathways by testing double mutants for Akt1 and JNK for PQ<br />

and PQ with minocycline sensitivity to further assess the mechanisms by which these signaling<br />

pathways are involved in this neurodegeneration model. In addition, it will be necessary to test<br />

the association <strong>of</strong> these signaling pathway with the PQ-induced NOS response in CNS by<br />

expressing the loss-<strong>of</strong>-function and/or dominant negative, and over-expression transgenic lines in<br />

the cells inducing the NOS against PQ.<br />

111


CHAPTER 4<br />

BRIEF EXPOSURE TO PARAQUAT DURING JUVENILE AND EARLY STAGES CAUSES<br />

PARKINSONIAN SYMPTOMS, INCREASED SENSITIVITY TO OXIDATIVE INSULT<br />

LATER IN LIFE AND A SHORTENED LIFE SPAN<br />

This work is a manuscript in preparation for submission to Journal <strong>of</strong> Neuroscience. Co-authors<br />

for this work were <strong>Arati</strong> <strong>Inamdar</strong>, Shane Welch, Russ Alexander and Janis O’Donnell. Shane<br />

Welch and Russ Alexander contributed in collecting data presented in figure 4.2 A, C. <strong>Arati</strong><br />

<strong>Inamdar</strong> collected the remaining data.<br />

112


INTRODUCTION<br />

Parkinson’s disease (PD), characterized by loss <strong>of</strong> dopaminergic neurons and presence <strong>of</strong><br />

Lewy bodies in the substantia nigra pars compacta (SNpc), is the second most common chronic<br />

neurodegenerative disease (Schiller, 2000). <strong>The</strong> loss <strong>of</strong> dopaminergic neurons along with<br />

degeneration <strong>of</strong> nerve terminals in the striatum eventually develops into Parkinsonian symptoms,<br />

resting tremor, rigidity, bradykinesia, and gait disturbance (Jellinger, 2001). Unfortunately, the<br />

exact cause <strong>of</strong> sporadic Parkinson’s disease is unknown. However, epidemiological and<br />

experimental studies have suggested multifactorial etiology with potential roles for<br />

environmental toxins, genetic susceptibility and aging in the pathogenesis <strong>of</strong> PD. Upon exposure<br />

to environmental toxins such as MPTP, 6-OHDA, rotenone, and paraquat in animal models,<br />

typical Parkinsonian features have been noted, implying that gene-environment interactions can<br />

be modeled to understand the mechanisms <strong>of</strong> etio-pathogenesis for PD (Mendez and Finn, 1975;<br />

Burns et al., 1983; Betarbet et al., 2000; McCormack et al., 2002; Uversky, 2004).<br />

PD previously has been considered to be associated with geriatric populations, but it has<br />

also been reported in populations below age 40. <strong>The</strong> appearance <strong>of</strong> Parkinsonian symptoms in<br />

the patients below the age <strong>of</strong> 40 (in some studies the cut <strong>of</strong>f age is considered as 50 yrs) are<br />

classified as Early-Onset Parkinsonism (EOP). <strong>The</strong> incidence <strong>of</strong> EOP ranges from 0.8 per<br />

100000 per year to 3.0 per 100000 per year in population between 0-49 yrs (Schrag and Schott,<br />

2006). Early-onset Parkinsonism has two subsets. Juvenile Parkinsonism patients represent that<br />

subset <strong>of</strong> EOP who present with the Parkinsonian features under age 21 while those with onset <strong>of</strong><br />

Parkinsonian features at or above 21 yrs but under 40/50 yrs are classified as Young-Onset PD<br />

(YOPD). <strong>The</strong> causes for Juvenile and YOPDs are largely proposed to be genetic but the definite<br />

cause is still unknown. Epidemiological studies have not yet reported exposure to pesticides as<br />

113


the risk factor for the pathogenesis <strong>of</strong> YOPD although rural living associated with well water<br />

drinking and head injury early in life have been proposed to be important factors accelerating the<br />

occurrences <strong>of</strong> Parksinonism symptoms in such individuals (Tsai et al., 2002). However,<br />

correlation between early exposure to toxins and early onset <strong>of</strong> PD has been proposed<br />

(Logroscino, 2005).<br />

Paraquat is one <strong>of</strong> the well-known Parkinson mimetic agents proposed to induce<br />

generation <strong>of</strong> ROS, thereby causing loss <strong>of</strong> dopaminergic neurons in both mammalian and<br />

Drosophila PD models (McCormack et al., 2002; McCormack et al., 2005; Chaudhuri et al.,<br />

2007). Various studies have recently demonstrated that like other neurotoxins, paraquat toxicity<br />

is partially mediated by nitric oxide where it acts as a diaphorase and increases the production <strong>of</strong><br />

oxygen radicals and lipid peroxidation, thereby causing dysfunction <strong>of</strong> membrane potential<br />

(Djukic et al., 2007, Day et al., 1999). In vitro studies demonstrated that PQ toxicity is induced<br />

in microglial cultures, but not in neuron-glia cultures implying that microglia mainly contributed<br />

to PQ-induced ROS in PQ-treated microglia enriched cultures (Wu et al., 2005). Further, single<br />

paraquat exposure is capable <strong>of</strong> activating microglia without DA neuron loss in mammalian PQ<br />

induced PD model (Purisai et al., 2007).<br />

<strong>The</strong>re has been a growing incidence <strong>of</strong> YOPD and higher mortality and morbidity in<br />

YOPD patients than the normal population (Schrag et al., 1998; Schrag A and Schott, 2006).<br />

<strong>The</strong>refore, it is still unknown whether a single exposure to Parkinson mimetics early in life plays<br />

any role in etio-pathogenesis <strong>of</strong> early onset PD. Here, we have used a PQ induced Drosophila<br />

PD model, which, as in mammalian PD models generated by continuous exposure <strong>of</strong> PQ,<br />

shortens life-span and induces mobility defects correlating with the loss <strong>of</strong> DA neurons<br />

(Chaudhuri et al., 2007). Further, we showed that PQ toxicity in our Drosophila model is<br />

114


associated with NO production. Finally, we showed that NO is induced in microglial-like cells,<br />

hemocytes, that mediate the engulfment and demise <strong>of</strong> dopaminergic neurons (<strong>Inamdar</strong> et al.,<br />

<strong>2009</strong>, under revision).<br />

In the present study, we present a comprehensive study employing our in vivo Drosophila<br />

PQ-induced PD model showing that brief exposure to paraquat in young adult and juvenile/larval<br />

stages is accompanied by loss <strong>of</strong> dopaminergic neurons, mobility defects and modified survival<br />

duration. Moreover, differential PQ sensitivity seems to be dependent upon the age at exposure,<br />

as well as dose <strong>of</strong> exposure. <strong>The</strong> results suggest that sporadic or brief exposure is capable <strong>of</strong><br />

initiating the toxic process influencing the sensitivity towards paraquat upon subsequent<br />

exposure.<br />

115


MATERIALS AND METHODS<br />

Drosophila strains and culture maintenance. A transgenic reporter strain, TH-GAL4;<br />

UAS-eGFP was employed in all experiments. <strong>The</strong> transgenic strain UAS-2X eGFP<br />

(Chromosome II) was obtained from the Bloomington, IN Drosophila stock center and a TH-<br />

GAL-4 strain (Friggi-Grelin et al., 2003) was obtained from Jay Hirsh (<strong>University</strong> <strong>of</strong><br />

Virginia). All stocks were maintained at 25º C, and all the experiments were performed at the<br />

same temperature.<br />

Feeding experiments. Separated males and females, less than six hours old post-<br />

eclosion and second instar larvae were fed paraquat at1 mM or 10 mM concentrations soaked<br />

on filter paper with 5% sucrose, and in 1 % agar with 5% sucrose and yeast for 12 hr for<br />

young adult flies and larvae, respectively. <strong>The</strong> Drosophila food medium was changed every<br />

15 days after exposure to PQ.<br />

Locomotion assay. <strong>The</strong> mobility <strong>of</strong> larvae was performed in petri dishes containing<br />

1% agar. <strong>The</strong> time required for the larvae to crawl a 5 cm distance towards a filter paper<br />

piece soaked in acetone was recorded. <strong>The</strong> average <strong>of</strong> three recordings per larva was noted<br />

for about 15-20 larvae. <strong>The</strong> mobility <strong>of</strong> adult male and female flies from each treatment<br />

group was assessed using a negative geotaxis climbing assay. A single fly was placed in an<br />

empty plastic vial, tapped to the bottom and the time required to climb 5 cm was recorded<br />

three times sequentially with 10 min rest periods between each measurement. Each<br />

replication value recorded is an average <strong>of</strong> the three trials; each assay was conducted on 10<br />

adult flies per test group.<br />

116


Confocal microscopy. Whole mounts <strong>of</strong> dissected brains from age matched TH-Gal4;<br />

UAS-eGFP adults and larvae fed with sucrose alone, or with paraquat, as described in the<br />

Results were examined for dopaminergic neuron morphology and number, detected by<br />

visualizing GFP-expressing neurons. Each brain was scanned to include 10-15 sections for<br />

optimum visualization <strong>of</strong> DA neurons. <strong>The</strong> Z-sections were then utilized to get the average <strong>of</strong><br />

all sections using a Leica TCS SP2 AOBS confocal microscope (Wetzlar, Germany).<br />

Statistical Analysis. All data were analyzed by one way ANOVA with Dunnett’s post<br />

test or by one-tailed Student’s t-test, assuming equal variances wherever applicable, using<br />

GraphPad Prism (San Diego, CA). Details <strong>of</strong> the analyses are described in the figure legends.<br />

117


RESULTS<br />

High and low doses <strong>of</strong> PQ, fed to juvenile (2nd instar larvae) and young (


in contrast to chronic PQ exposure <strong>of</strong> sexually mature adults, where females were less<br />

sensitive to PQ than males (Chaudhuri et al., 2007). <strong>The</strong>refore, as is the case for chronic PQ<br />

exposure, a single episode <strong>of</strong> PQ-exposure earlier in life has long-term consequences for<br />

survival.<br />

119


Figure 4.1: Exposure <strong>of</strong> juvenile (2 nd instar larvae) and young adult (


A<br />

B<br />

121


Exposure <strong>of</strong> juvenile and young adult (


Figure 4.2: Exposure <strong>of</strong> larva and young adult flies to high and low doses <strong>of</strong> PQ causes<br />

mobility defects immediately after exposure to PQ and in later stages <strong>of</strong> life. (A) Second<br />

instar stage larvae were exposed to 10 mM and 1 mM PQ, or to 5% sucrose for 12 hr and<br />

then transferred to the normal food. <strong>The</strong> mobility <strong>of</strong> the larvae was assayed by the rate at<br />

which they crawled towards an acetone soaked filter paper kept in a petri-dish 5 cm away<br />

from the start-line. <strong>The</strong> larvae exposed to PQ in the larval stage demonstrate mobility defects.<br />

(B) <strong>The</strong> PQ (10 mM and 1 mM) induced mobility defect in adult males exposed to PQ during<br />

the larval stage and aged to one and a half months post-eclosion, was determined by the<br />

negative geotaxis assay. (C) <strong>The</strong> PQ-mobility defect in the young adult males performed<br />

immediately after 12 hr exposure to PQ (1 mM and 10 mM) was determined by the negative<br />

geotaxis assay. (D) <strong>The</strong> PQ-induced mobility defects <strong>of</strong> males exposed to 1 mM PQ for 12 hr<br />

as newly eclosed adults and subsequently aged for one and a half months on normal food.<br />

<strong>The</strong> * indicates the significance <strong>of</strong> differences between control and PQ-fed flies. NS = not<br />

significant ***=P< 0.005, ** = P < 0.005 and * = P < 0.05. Error bars represent standard<br />

error <strong>of</strong> the mean. Each data point represents at least 5 replications <strong>of</strong> 15 flies each.<br />

123


A<br />

B<br />

124


C<br />

D<br />

125


Exposure <strong>of</strong> juvenile and young adult (


for each subgroup <strong>of</strong> neurons following exposure to both PQ concentrations is shown in Fig<br />

4.3P.<br />

<strong>The</strong>se results show that there is dose-dependent loss <strong>of</strong> DA neurons which correlates<br />

well with the mobility defects associated with PQ exposure reported in the previous section.<br />

127


Figure 4.3: Exposure to high and low doses <strong>of</strong> PQ induces dose-dependent loss <strong>of</strong> DA<br />

neurons in larval and young adult brains. (A), (B). Schematic diagrams showing the positions<br />

<strong>of</strong> DA subgroups <strong>of</strong> neurons in larval and adult brains, respectively. (C, D, E) <strong>The</strong> effect <strong>of</strong> 5<br />

% sucrose, 1 mM PQ and 10 mM PQ on the dopaminergic neurons <strong>of</strong> w; TH-GAL4; UASeGFP<br />

larval brain. PQ causes severe loss <strong>of</strong> almost all regions <strong>of</strong> DA neurons in the larval<br />

brain exposed to 10 mM PQ (E) but not with 1 mM PQ (D) compared to control larval<br />

brain(C). (F, G, H) Insets enlarged for larval DM, DL1 subgroups <strong>of</strong> DA neurons for control,<br />

PQ (1 mM) and PQ 10 mM. (I) <strong>The</strong> average number <strong>of</strong> DA neurons after 12 hr <strong>of</strong> continuous<br />

feeding <strong>of</strong> 5 % sucrose, 1 mM PQ and 10 mM PQ performed on <strong>of</strong> w; TH-GAL4; UAS-eGFP<br />

larval brain. <strong>The</strong>re was significant reduction in the DA neurons in DM, DL1, DL2 subgroups<br />

in the larval brain fed with 10 mM PQ but no difference between the neuron counts between<br />

the larval brains fed with 5 % sucrose and 1 mM PQ. (J,K,L) <strong>The</strong> effect <strong>of</strong> 5 % sucrose, 1<br />

mM PQ and 10 mM PQ on the dopaminergic neurons <strong>of</strong> w; TH-GAL4; UAS-eGFP adult<br />

brain. 10 mM PQ (L), but not 1 mM (K), causes alteration in the number and morphology <strong>of</strong><br />

DA neurons compared to control DA neurons (J), suggesting a dose-dependent loss <strong>of</strong> DA<br />

neurons induced by PQ in both larval and adult brain. (M,N,O) Insets enlarged for PPM2<br />

subgroup <strong>of</strong> DA neurons in adult brain for control, PQ (1 mM) and PQ (10 mM). (P)<strong>The</strong><br />

average number <strong>of</strong> DA neurons after 12 hr <strong>of</strong> continuous feeding <strong>of</strong> 5 % sucrose, 1 mM PQ<br />

and 10 mM PQ, performed on <strong>of</strong> w; TH-GAL4; UAS-eGFP adult brain, shows that PAL,<br />

PPM3 and PPL2 subgroups <strong>of</strong> DA neurons are affected by 10 mM PQ but not with 1 mM<br />

PQ. <strong>The</strong> significance <strong>of</strong> differences between PQ-treated and control groups in each subgroup<br />

were indicated as *, where * = P< 0.05. Error bars represent standard error <strong>of</strong> the mean. Each<br />

data point represents at least 5-7 brains. Scale bars for C,D,E=50 μm; J,K,L =100 μm. (Image<br />

source for A, B: Images published in Friggi-Grenin et al., 2003)<br />

128


Wild type<br />

F<br />

129


I<br />

130


J<br />

Wild-type<br />

K<br />

PQ 1 mM<br />

L<br />

PQ 10 mM<br />

131<br />

M<br />

N<br />

O


P<br />

132


Exposure <strong>of</strong> juvenile and young adult (


Figure 4.4: Exposure to 1 mM PQ at early stages <strong>of</strong> life sensitizes adults to subsequent PQ<br />

exposure. A reduction in average survival duration <strong>of</strong> flies pre-exposed to 1 mM PQ at larval<br />

and young adult age for 12 hr and continuously exposed to 1 mM PQ after 2 months on<br />

normal food was detected. This result demonstrates that early exposure to PQ sensitizes the<br />

flies for a second exposure <strong>of</strong> PQ. <strong>The</strong> significance <strong>of</strong> the difference between 5 % sucrose<br />

pre-exposed and 1 mM PQ pre-exposed at larval and young adult stages are represented with<br />

* where, P


DISCUSSION<br />

Brief exposure to PQ early in life causes truncation <strong>of</strong> life span and mobility defects in later<br />

stages <strong>of</strong> life<br />

We report here the long-term effect <strong>of</strong> brief exposure early in life to PQ, one <strong>of</strong> the<br />

potential Parkinson mimetics. <strong>The</strong> etiology <strong>of</strong> sporadic forms <strong>of</strong> PD is considered to be<br />

multifactorial. <strong>The</strong> processes <strong>of</strong> oxidative stress and inflammation are considered the most<br />

important factors amplifying the degenerative changes in PD. <strong>The</strong> exact stimuli capable <strong>of</strong><br />

initiating or enhancing this degenerative process are still unknown. However, current studies<br />

have suggested that exposure to insecticides, pesticides, brain injury, and brain infections<br />

early in life may serve key roles in the pathogenesis <strong>of</strong> PD (Liu et al., 2003; Liu and Hong,<br />

2003). Idiopathic PD affects 3% <strong>of</strong> the population between ages 65-85, 4-5% <strong>of</strong> the<br />

population over 85 years and, surprisingly, 5-10 % <strong>of</strong> all PD patients are being reported to be<br />

less than 40 years <strong>of</strong> age. <strong>The</strong>se epidemiological data suggest that the pathogenesis <strong>of</strong><br />

idiopathic form <strong>of</strong> PD may be associated with certain risk factors capable <strong>of</strong> altering the<br />

homeostasis <strong>of</strong> DA in the brain via multiple mechanisms. In PD patients, the SN shows a<br />

reduction in the levels <strong>of</strong> Glutathione (GSH), increase in lipid peroxidases, oxidative<br />

products <strong>of</strong> damaged RNA and other products, indicating the detrimental role <strong>of</strong> oxidative<br />

damage for DA neuron degeneration (Halliwell, 2001). Several toxin-induced PD models<br />

have further supported the association <strong>of</strong> oxidative stress to PD.<br />

Among many neurotoxins capable <strong>of</strong> inducing PD-like symptoms, PQ is considered<br />

to be the most potent redox cycler and is capable <strong>of</strong> inducing oxidative damage to almost all<br />

cells including DA neurons (McCormack et al., 2002). Noteworthy, epidemiological studies<br />

support an increase in the incidence <strong>of</strong> PD in agricultural populations known to be exposed to<br />

PQ (Liou et al., 1997). We have generated a Drosophila PD model based on the continuous<br />

135


ingestion <strong>of</strong> PQ, which is able to reproduce Parkinsonian features including mobility deficits<br />

(Chaudhuri et al., 2007). Further, we found that PQ toxicity is dependent on the dosage <strong>of</strong><br />

PQ, and these results are supported by the observations reported here. In this report, we have<br />

shown that brief exposure <strong>of</strong> high (10 mM) or low (1 mM) doses <strong>of</strong> PQ is capable <strong>of</strong><br />

initiating processes detrimental to the organism and ultimately results in early death,<br />

although the immediate impact <strong>of</strong> low dose <strong>of</strong> PQ was not evident immediately after<br />

exposure as assessed by the status <strong>of</strong> the DA neurons. In contrast, feeding the higher dose <strong>of</strong><br />

PQ (10 mM) not only caused DA neuron loss even after only a 12 hr exposure to larvae and<br />

young adults but also resulted in a significant reduction in average survival duration in both<br />

groups. Similarly, although the 1 mM PQ dose did not cause degeneration <strong>of</strong> DA neurons in<br />

larvae or young adult brains, the treatment strongly decreased their survival duration (~65-70<br />

days) relative to control flies (~90 days) (Fig. 4.1 A, B and Fig 4.3 C, D, E, F, G, H,I, J, K, L,<br />

M, N, O, P). Surprisingly, flies exposed to 10 mM PQ for 12 hrs soon after eclosion could<br />

only survive about 30 days (Fig. 4.1 B). Flies eclosed after larval exposure to 10 mM PQ<br />

fared somewhat better, surviving about 50 days (Fig. 4.1 A). It has been observed that during<br />

metamorphosis further development and differentiation <strong>of</strong> DA neurons occurs and this could<br />

be the reason for the differences in the total survival durations for the larvae and young flies<br />

exposure to same concentration <strong>of</strong> PQ (Budnik and White, 1988). Moreover, loss <strong>of</strong> DA<br />

neurons has been reported in aged flies (Neckameyer et al., 2000). Interestingly, we did not<br />

find a significant difference in the survival duration between males and females in these<br />

experiments as opposed to the differences we found when 20 mM PQ was exposed<br />

continuously to 24-48 hr old flies (Chaudhuri et al., 2007) (Fig. 4.1 A, B). However, it has<br />

previously been reported that response to the PQ-induced oxidative stress alters the TH<br />

136


activity differentially depending upon the gender, age, reproductive status and sexual<br />

maturity (Neckameyer and Weinstein, 2005). Our results are consistent with their<br />

observations.<br />

Both larvae and young adults exposed to 10 mM PQ demonstrated mobility defects as<br />

assessed by crawling and negative geotaxis assays, respectively. Although 1 mM PQ induced<br />

bradykinesis in larvae after 12 hr <strong>of</strong> continuous feeding, young adult flies ingesting the same<br />

dose for 12 hr lacked significant mobility defects (Fig 4.2 A, C). Interestingly, upon transfer<br />

to normal food these flies had obvious mobility defects until about 35 days post-eclosion,<br />

when we noted the onset <strong>of</strong> tremors in the flies exposed to 1 mM PQ at either the larval and<br />

young adult stages. Gradually these features became worse as assessed by negative geotaxis<br />

assay performed after one and a half months being on normal food medium on the flies<br />

exposed to 1 mM PQ at larval and young adult age (Fig.4.2 B, D). Surprisingly, the flies pre-<br />

exposed to PQ during the larval stage survived longer than those exposed as young adults,<br />

suggesting that metamorphosis may be a favorable event for removing dead DA neurons,<br />

development and differentiation <strong>of</strong> DA neurons and DA homeostasis (Budnik and White,<br />

1988).<br />

Brief exposure to PQ sensitizes the DA neurons for the subsequent exposure <strong>of</strong> paraquat<br />

Although several studies have presented evidence supporting the exposure <strong>of</strong> PQ as<br />

an etiological agent for PD, most <strong>of</strong> these studies employ continuous exposure <strong>of</strong> PQ.<br />

Considering the growing incidence <strong>of</strong> PD in a comparatively young population, it may be<br />

speculated that exposure to proposed PD inducers briefly at early points in life increases the<br />

vulnerability <strong>of</strong> individual towards exposure to PD inducers at later stages.<br />

137


Purisai et al. (2007) found that a low dose <strong>of</strong> PQ is capable <strong>of</strong> microglial activation in SN,<br />

even though no DA neurons are lost. However, this even sensitizes the DA neurons for<br />

degeneration after a subsequent PQ exposure. Our experiments demonstrate the generality <strong>of</strong><br />

these effects. <strong>The</strong> brief exposure <strong>of</strong> PQ without any detectable <strong>of</strong> DA neuron loss by 1 mM<br />

PQ amplifies the death process after the flies are exposed to another 1 mM dose around 2<br />

months later possibly via generation <strong>of</strong> ROS and ROS mediated DA neuron death (Fig. 4.4).<br />

It is to be noted that the difference in the survival <strong>of</strong> control groups (from larvae and young<br />

adult) upon exposed to 1 mM PQ after 2 months could be due to two different time points at<br />

which these experiments were performed. It will be important to pursue the question <strong>of</strong><br />

whether this early event also perturbs DA homeostasis in the adult brain as well as other<br />

metabolic processes that may lead to increased sensitivity towards such PD inducers as well<br />

as early death. Whether, brief exposure <strong>of</strong> PQ-like neurotoxins are capable for inducing<br />

NOS, one <strong>of</strong> the important inflammatory mediators known to be activated in inflammatory<br />

response in mammalian models are not known. Since, we have detected the induction <strong>of</strong><br />

NOS in the adult brain after exposure to PQ for 6 hr (<strong>Inamdar</strong> et al., <strong>2009</strong>, under revision); it<br />

would be interesting to known if exposure to high and low doses <strong>of</strong> PQ for brief (12 hr)<br />

period <strong>of</strong> time is also associated with induction <strong>of</strong> NOS in larvae and young adults at any<br />

time-point. Moreover, Langston et al. (1999) reported a study performed on three patients<br />

that were known to be exposed to MPTP in which activated microglia were found around the<br />

DA neurons after 3-16 years suggesting a correlation between inflammatory processes and<br />

DA neuron loss at later stages <strong>of</strong> life. However, it is unknown if a similar response will be<br />

evident with PQ exposure at early stages <strong>of</strong> life. <strong>The</strong>refore, whether the sustained induction<br />

<strong>of</strong> the NOS dependent response leads to long-term induction <strong>of</strong> pro-inflammatory molecules<br />

138


as seen in mammalian models is currently unknown and will be an important topic for further<br />

experiments.<br />

139


CHAPTER 5<br />

IDENTIFICATION OF A NITRIC OXIDE-DEPENDENT RESPONSE IN S. VENEZUELAE-<br />

INDUCED PARKINSON’S DISEASE IN DROSOPHILA MELANOGASTER<br />

This work has been in progress in O’Donnell Lab. All the data were collected by <strong>Arati</strong> <strong>Inamdar</strong>.<br />

140


INTRODUCTION<br />

<strong>The</strong> processes <strong>of</strong> neuroinflammation and oxidative stress are thought to be among the<br />

primary mechanisms playing roles in the etiology and pathophysiology <strong>of</strong> Parkinson’s disease<br />

(PD). In addition to genetic causes, environmental factors are proposed to contribute to the<br />

sporadic, and possibly hereditary, forms <strong>of</strong> PD. Among the various candidate environmental<br />

factors, recent concern has been raised regarding exposure to the soil bacterium, Actinobacteria,<br />

in agricultural populations, who are known to exhibit increased incidence <strong>of</strong> PD (Semchuk et al.,<br />

1992; Kamel et al., 2007). Actinobacteria, which consist <strong>of</strong> pathogenic species like Streptomyces<br />

and Nocardia, are gram-positive bacteria. Actinobacteria are known to produce secondary<br />

metabolites, which are composed <strong>of</strong> various products balanced in necessary biological elements<br />

such as carbon, nitrogen, and phosphorus that can be reutilized by the bacteria to survive in<br />

nutrient depleted environment. Further, these secondary metabolites could contain products toxic<br />

to other microorganisms or possess therapeutically important properties (Shapiro, 1989).<br />

<strong>The</strong> etiological agents responsible for Parkinsonian symptoms in patients and<br />

experimental models provide growing evidence for an infectious cause for some cases <strong>of</strong> PD,<br />

particularly various viruses, especially the Epstein Barr virus, influenza virus, coronavirus and<br />

Japanese encephalitis virus (Duvoisin and Yahr, 1965; Fazzini et al., 1992; Pradhan et al., 1999).<br />

Since the 1980’s, several additional microorganisms such as Borrelia burgdorferi, Haemophilus<br />

influenzae, Corynebacterium diphtheriae, Cryptococcus ne<strong>of</strong>ormans, Mycoplasma pneumoniae<br />

and Helicobacter pylori have been reported to be associated with PD features (Wszolek et al.,<br />

1988; Kim et al., 1995; Altschuler, 1996).<br />

141


Recently, the role <strong>of</strong> Nocardia in the pathogenesis <strong>of</strong> Parkinson’s disease has been<br />

reported (Broxmeyer, 2002). Exposure <strong>of</strong> neuronal cell culture and a mouse model to media<br />

containing the secreted metabolites from growing Nocardia caused apoptosis in dopaminergic<br />

neurons and induced Parkinsonian symptoms such as bradykinesia and stooped posture. In<br />

another study, it was observed that exposure to Nocardia caused inhibition <strong>of</strong> the ubiquitin<br />

proteosome pathway, leading to apoptotic death <strong>of</strong> neurons in substantia nigra without induction<br />

<strong>of</strong> an inflammatory response in that region (Tam et al., 2002). Díaz-Corrales et al. (2004)<br />

showed that intravenous injection <strong>of</strong> the Nocardia species isolated from a patient suffering from<br />

Actinomycetoma in mice reproduced Parkinsonian symptoms with alterations in dopamine (DA)<br />

levels in the substantia nigra (SN).<br />

Given the possibility <strong>of</strong> an infectious etiology <strong>of</strong> Parkinson’s disease, our collaborators in<br />

<strong>The</strong> Caldwell Lab (<strong>University</strong> <strong>of</strong> Alabama), recently tested for effects <strong>of</strong> Streptomyces<br />

venezuelae on the dopaminergic neurons <strong>of</strong> C. elegans and found that exposure to media<br />

containing bacterial secondary metabolites from this strain <strong>of</strong> common soil bacteria cause loss <strong>of</strong><br />

dopaminergic neurons. <strong>The</strong>y also found that ingestion <strong>of</strong> the proteosome inhibitor, MG132, also<br />

induces loss <strong>of</strong> dopaminergic neurons in C. elegans. <strong>The</strong> results were also confirmed in human<br />

dopaminergic neuron cell culture, SHSY5Y, employing culture media obtained from growing S.<br />

colicolor as the control against S. venezuelae culture media (J. Armagost, K. Caldwell, and G.<br />

Caldwell, unpublished results).<br />

Despite these results, the mechanism through which S. venezuelae induces DA neuron<br />

loss is still unknown. As discussed in Chapter 2, paraquat (PQ), a well-known herbicide that is<br />

also considered an environmental risk factor, was previously found in our laboratory to produce<br />

Parkinsonian symptoms in Drosophila, including resting tremors and region-specific<br />

142


degeneration <strong>of</strong> dopaminergic neurons (Chaudhuri et al., 2007). Further, we have recently<br />

discovered that upon exposure to PQ, the adult brain <strong>of</strong> Drosophila also shows induction <strong>of</strong> nitric<br />

oxide synthase (NOS) as assayed biochemically and by immunolocalization. Further NOS-<br />

positive structures found to be surrounding the dopaminergic neurons were not in glial cells<br />

(<strong>Inamdar</strong> et al., <strong>2009</strong>, under revision).<br />

Here, I demonstrate that the ingestion <strong>of</strong> a crude conditioned medium <strong>of</strong> S. venezuelae<br />

also causes Parkinsonian symptoms in Drosophila, similar to the behavioral and cellular changes<br />

induced by PQ. In addition, we found that exposure to such medium induces NOS in the adult<br />

brain. In particular, unlike PQ exposure, the induced NOS cells do not seem to surround the<br />

dopaminergic cell bodies; rather they associate closely with glial cells suggesting that the<br />

mechanism <strong>of</strong> action <strong>of</strong> this toxin on CNS may be distinct from PQ-associated mechanisms. I<br />

report results that begin the process <strong>of</strong> defining the mechanism <strong>of</strong> action <strong>of</strong> this toxin, which has<br />

the propensity to cause Parkinsonian symptoms, and may be one <strong>of</strong> the environmental causes <strong>of</strong><br />

PD.<br />

143


MATERIALS AND METHODS<br />

Drosophila strains and culture maintenance. <strong>The</strong> strain, Df (1)w; y, which is normal for<br />

all genes functioning in DA homeostasis, was used as the wild type strain. <strong>The</strong> transgenic strains<br />

UAS-2X eGFP (Chromosome II), Repo (reversed polarity), GAL4 (a glial cell driver) were<br />

obtained from the Bloomington, IN Drosophila stock center and a TH-GAL4 (a DA neuron<br />

driver) (Friggi-Grelin et al., 2003) was obtained from Jay Hirsh (<strong>University</strong> <strong>of</strong> Virginia).<br />

Cultures were maintained at 25º C.<br />

Feeding experiments. Separated male and female flies, 48 hr post-eclosion, were fed on<br />

filter paper saturated with one <strong>of</strong> the following solutions: 5% sucrose only or 5% sucrose with<br />

crude conditioned culture media collected from S. venezuelae and S. colicolor bacterial strains.<br />

Feedings were continued until all flies were dead. <strong>The</strong> crude conditioned culture media were<br />

obtained from Caldwell Lab, UA.<br />

Locomotion assay. <strong>The</strong> mobility <strong>of</strong> adult male and female flies from each treatment<br />

group was assessed using a negative geotaxis climbing assay. A single fly was placed in an<br />

empty plastic vial, tapped to the bottom and the time required to climb 5 cm was recorded three<br />

times sequentially with 10 min rest periods between each measurement. Each replication value<br />

recorded is an average <strong>of</strong> the three trials; each assay was conducted on 10 flies per test group.<br />

Immunohistochemistry. Brains from TH-GAL4; UAS-2XeGFP and Repo-GAL4; UAS-<br />

eGFP adults (untreated or exposed to crude conditioned medium from S. venezuelae and S.<br />

colicolor bacterial strains for 24 or 48 hr) were fixed in 4% paraformaldehyde for 3.5 hr and<br />

144


washed extensively in 1x phosphate buffered saline (PBS) and then in 0.1% Triton X-100, 0.2%<br />

bovine serum albumin, in PBS (PBT). Brains were then blocked in 5% normal goat serum in PBT<br />

overnight at 4°C, followed by overnight incubation with a 1: 500 dilution <strong>of</strong> rabbit anti-NOS<br />

antiserum (N217, Sigma Aldrich) and mouse anti-GFP antibody (Abcam). After additional<br />

washing, the brains were incubated at room temperature for 2 hr in a 1: 5000 and 1: 5000 dilution<br />

<strong>of</strong> Cy-3-conjugated goat anti-rabbit IgG and FITC conjugated rabbit anti-mouse IgG,<br />

respectively. Confocal studies were performed using a Leica TCS SP2 AOBS confocal<br />

microscope (Wetzlar, Germany). <strong>The</strong> whole mounts <strong>of</strong> dissected brains from TH-Gal4; UAS-<br />

eGFP adults for the dopaminergic neuron morphology and number, detected by visualizing GFP-<br />

expressing neurons was performed on the above mentioned confocal microscope. Each brain was<br />

scanned to include 10-15 sections for optimum visualization <strong>of</strong> cells being studied. <strong>The</strong>se Z-<br />

sections were then used to get the average image <strong>of</strong> all sections as presented in Results section.<br />

Statistical Analysis. All data were analyzed by one way ANOVA with Dunnett’s post test<br />

or by one-tailed Student’s t-test, assuming equal variances wherever applicable, using GraphPad<br />

Prism (San Diego, CA). Details <strong>of</strong> the analyses are described in the figure legends.<br />

145


RESULTS<br />

Exposure to S. venezuelae conditioned medium causes truncation <strong>of</strong> life span in wild type flies<br />

<strong>The</strong> crude conditioned medium is derived from the media containing secondary<br />

metabolites from the growing S. venezuelae. In order to determine whether crude conditioned<br />

medium is functionally active and imparts detrimental effects in our Drosophila model, we<br />

hypothesized that exposure <strong>of</strong> the conditioned medium causes truncation <strong>of</strong> life span <strong>of</strong> wild type<br />

flies. Wild type flies were exposed to crude conditioned medium from S. venezuelae and S.<br />

colicolor and their survival was noted every 24 hr intervals until all flies were dead. <strong>The</strong><br />

conditioned medium <strong>of</strong> S. colicolor, which was non-toxic in C. elegans and a human<br />

dopaminergic cell line, SHSY5Y was used as a control (J. Armagost, K. Caldwell, and G.<br />

Caldwell, unpublished results). We also found that exposure to S. colicolor had limited toxicity<br />

in flies, while flies exposed to S. venezuelae showed a gradual decline in survival with a sudden<br />

peak in the death rate after 96 hr <strong>of</strong> exposure with death <strong>of</strong> all flies by 168 hr. <strong>The</strong> death trend<br />

observed with S. venezuelae toxin exposed was different from that <strong>of</strong> PQ exposure, in which the<br />

rate the wild type flies died was almost the same at each time point (Fig. 5.1). This suggests that<br />

upon exposure to conditioned medium <strong>of</strong> S. venezuelae, an accumulation <strong>of</strong> toxic substances<br />

occur, under the influence <strong>of</strong> which flies succumb in a delayed response.<br />

146


Figure 5.1: Exposure to crude conditioned medium <strong>of</strong> S. venezuelae causes truncation <strong>of</strong> life<br />

span in wild type flies. <strong>The</strong> crude conditioned media from S. venezuelae and S. colicolor cultures<br />

were fed to wild type flies aged to 48 hr post-eclosion continuously until all flies on S.<br />

venezuelae conditioned medium were dead. <strong>The</strong>re was an initial increase in death on both<br />

conditioned mediums until 96 hr. However, no further death was seen on S. colicolor<br />

conditioned medium while the flies on S. venezuelae exhibited a continuous death trend after 96<br />

hr, culminating in death <strong>of</strong> all flies in the next three days. <strong>The</strong> SEM are not clearly visible in the<br />

graph due to small error numbers. Each data point represents at least 10 replications <strong>of</strong> 10 flies<br />

each.<br />

147


Exposure to S. venezuelae conditioned medium causes mobility defects in wild type flies<br />

Since our collaborators had noted the loss <strong>of</strong> DA neurons in both C. elegans and cell<br />

culture after exposure to S. venezuelae conditioned medium, we next asked whether this bacterial<br />

strain had a similar effect on Drosophila DA neurons. We previously found that wild type flies<br />

exposed to 20 mM PQ began exhibiting strong tremors and bradykinesia within 24 hr, preceding<br />

death by approximately one to two days (See supplementary videos <strong>of</strong> tremors and mobility after<br />

PQ ingestion in Chaudhuri et al., 2007). Flies exposed to the S. venezuelae conditioned medium<br />

for four days (a point at which approximately 50% <strong>of</strong> the flies had died) exhibited bradykinesia<br />

(Fig. 5.2) as in PQ exposure. However, there were also significant differences between the<br />

effects <strong>of</strong> PQ and the bacterial medium. Under PQ exposure tremors preceded the initiation <strong>of</strong><br />

increased mortality, while flies exposed to the bacterial medium only exhibited tremors well after<br />

the initial increase in mortality, generally between 3-4 days after exposure to the medium (Fig.<br />

5.2). Moreover, these tremors were not as severe as seen with PQ exposure. No obvious mobility<br />

defects were observed in flies exposed to S. colicolor conditioned medium. <strong>The</strong>se data show that<br />

exposure to crude conditioned medium from S. venezuelae induces mobility defects, a<br />

characteristic feature <strong>of</strong> PD.<br />

148


Figure 5.2: Exposure to crude conditioned medium <strong>of</strong> S. venezuelae induces Parkinsonian-like<br />

mobility defects. <strong>The</strong> movement defects induced by feeding S. venezuelae medium to wild type<br />

flies were determined using a negative geotaxis assay, performed on the fourth day after the<br />

initiation <strong>of</strong> continuous exposure started at 48 hr post-eclosion. <strong>The</strong> average time required by a<br />

single fly to climb a 5 cm distance is presented. <strong>The</strong> flies exposed to S. venezuelae conditioned<br />

medium showed predominant bradykinesia along with tremors at this time while these features<br />

were not evident in flies fed conditioned medium <strong>of</strong> S. colicolor or 5% sucrose. <strong>The</strong>re was no<br />

significant difference in the climbing ability <strong>of</strong> wild type flies on the conditioned medium <strong>of</strong> the<br />

non-toxic S. colicolor conditioned medium and 5% sucrose. Each data point represents at least<br />

10 replications <strong>of</strong> 10 flies each. NS= non significant, *= P


Exposure to crude conditioned medium <strong>of</strong> S. venezuelae causes loss <strong>of</strong> dopaminergic neurons in<br />

wild type flies<br />

I next asked whether exposure to S. venezuelae conditioned medium was associated with<br />

loss <strong>of</strong> DA neurons, employing the transgenic reporter strain, TH-GAL4; UAS-eGFP to monitor<br />

the DA neurons (Friggi-Grelin et al., 2003). I observed morphological changes in each subgroup<br />

<strong>of</strong> DA neurons at 24 hr intervals and found no significant loss <strong>of</strong> DA neurons (data not shown).<br />

At 48 hr <strong>of</strong> continuous exposure, differential responses <strong>of</strong> DA neuron subgroups were found<br />

(Fig. 5.3 A, B, C and Fig. 5.3 G). For example, loss <strong>of</strong> the PPM2 subgroup <strong>of</strong> DA neurons<br />

exposed to S. venezuelae conditioned medium was found relative to the same subgroup <strong>of</strong><br />

neurons exposed to 5% sucrose and crude conditioned medium <strong>of</strong> S. colicolor (Fig. 5.3 D, E, F).<br />

DA neurons exposed to S. colicolor conditioned medium showed no decrease in the number or<br />

morphology <strong>of</strong> neurons relative to control (5 % sucrose fed) brains up to 48 hr after initiation <strong>of</strong><br />

exposure (Fig. 5.3 G). <strong>The</strong>refore, exposure to crude conditioned medium <strong>of</strong> S. venezuelae is<br />

associated with loss <strong>of</strong> DA neurons, and thus provides evidence for its potency to induce<br />

Parkinsonism in flies.<br />

150


Figure 5.3: Exposure to crude conditioned medium <strong>of</strong> S. venezuelae induces loss <strong>of</strong> DA neurons.<br />

<strong>The</strong> effect <strong>of</strong> 48 hr <strong>of</strong> continuous exposure <strong>of</strong> 5% sucrose, crude conditioned media <strong>of</strong> S.<br />

colicolor and S. venezuelae on the dopaminergic neurons <strong>of</strong> TH-GAL4; UAS-eGFP adult brain.<br />

(A, B) <strong>The</strong> number and normal asymmetric neuron morphology was seen in brains exposed to<br />

5% sucrose and crude conditioned medium <strong>of</strong> S. colicolor (B). (C) <strong>The</strong> brains treated with S.<br />

venezuelae conditioned medium induced loss <strong>of</strong> DA neurons in different subgroups <strong>of</strong> neurons.<br />

(D, E, F) <strong>The</strong> insets show the changes in the number <strong>of</strong> the PPM2 subgroup <strong>of</strong> neurons in A,B,C,<br />

respectively. Scale in A, B, C= 100 μm. (G) <strong>The</strong> average number <strong>of</strong> neurons per subgroup was<br />

determined 48 hr after the initiation <strong>of</strong> feeding. All scoring was done on TH-Gal4:UAS-GFP<br />

adults. Each subset <strong>of</strong> dopaminergic neurons was scored separately (n=10-15). No difference in<br />

the neuron counts between 5% sucrose and S. colicolor conditioned medium were found but<br />

there was significant loss <strong>of</strong> DA neurons in PPM2, PPM3, and PPL1 was detected in S.<br />

venezuelae conditioned medium. **=P < 0.005 and represents significant difference between S.<br />

venezuelae conditioned medium and 5% sucrose or S. colicolor conditioned medium.<br />

151


A<br />

152<br />

D<br />

E<br />

F


G<br />

153


Exposure to conditioned medium <strong>of</strong> S. venezuelae causes activation <strong>of</strong> nitric oxide synthase<br />

(NOS) near DA neurons in adult fly brain<br />

Upon exposure to PQ, we found NOS expressing-structures surrounding the individual DA<br />

cell bodies. <strong>The</strong>se are postulated to be cells engulfing debris from dying neurons (<strong>Inamdar</strong> et al.,<br />

<strong>2009</strong>, under revision; Fig. 2.6A, chapter 2). I hypothesized that exposure <strong>of</strong> conditioned medium<br />

<strong>of</strong> S. venezuelae causes activation <strong>of</strong> NOS like in mammalian PD model, leading to death <strong>of</strong> DA<br />

neurons, a response we have proposed to be crucial for the death <strong>of</strong> DA neurons upon PQ<br />

exposure (<strong>Inamdar</strong> et al., <strong>2009</strong>, under revision). I therefore exposed TH-GAL4; UAS-eGFP flies<br />

to the conditioned medium <strong>of</strong> S. venezuelae and S. colicolor for 48 hr and immunohistochemistry<br />

using anti-NOS and anti-GFP antibodies to detect NOS and DA neurons expression,<br />

respectively. I found that normal DA neuron number and morphology were retained in brains<br />

exposed to S. colicolor conditioned medium with no induction <strong>of</strong> NOS near DA neurons (Fig.<br />

5.3 G, Fig. 5.4 A). In contrast, S. venezuelae exposed brains exhibited a gradual reduction in the<br />

number <strong>of</strong> DA neurons in each subgroup from 48 hr until 72-96 hr exposure when complete loss<br />

<strong>of</strong> DA neurons occurred (data not shown). Notably, unlike the close association <strong>of</strong> NOS signal,<br />

which surrounded DA neurons in PQ-treated brain (Fig. 2.7 B, Chapter 2), the activated NOS<br />

was never found to completely surround DA neurons upon exposure to S. venezuelae (Fig. 5.4B).<br />

A similar response was detected in the brains exposed to crude conditioned medium <strong>of</strong> S.<br />

venezuelae for 24 hr (data not shown). <strong>The</strong> size <strong>of</strong> the NOS-positive structures roughly ranges<br />

between 0.6-2 μm in diameter. <strong>The</strong> difference observed in distribution <strong>of</strong> the NOS signal in PQ<br />

vs. toxin exposed brains suggests the possibility that the toxin is inducing neuron death by a<br />

different mechanism.<br />

154


Figure 5.4: Exposure <strong>of</strong> crude conditioned medium <strong>of</strong> S. venezuelae induced NOS near DA<br />

neurons in TH-GAL4; UAS-eGFP transgenic strains. TH-GAL4; UAS-eGFP flies were exposed<br />

to conditioned media <strong>of</strong> S. colicolor and S. venezuelae for 48 hr. Exposure to S. colicolor<br />

conditioned medium (A), failed to induce NOS, while exposure to S. venezuelae conditioned<br />

medium caused induction <strong>of</strong> NOS detected by anti-NOS antibody (arrows, red) near GFP<br />

expressing DA neurons detected using anti-GFP antibody. However, these NOS-positive<br />

structures, while occurring in the approximate vicinity <strong>of</strong> DA neurons, were neither found to be<br />

in close proximity to the neuronal cell bodies nor surrounding the DA neurons. <strong>The</strong> PPM1<br />

subgroup <strong>of</strong> DA neuron is shown in the images A and B. Scale bar=15.87 μm.<br />

155


A<br />

B<br />

S. colicolor<br />

S. venezuelae<br />

156


Exposure to conditioned medium <strong>of</strong> S. venezuelae induced NOS near glial cells<br />

In Drosophila, glial cells constitute a significant volume <strong>of</strong> adult brain, where among<br />

other functions they provide trophic support, metabolic and homeostatic functions necessary for<br />

neuron survival. Death <strong>of</strong> glial cells has been proposed to result in death <strong>of</strong> neurons in<br />

Drosophila (Parker and Auld, 2006). <strong>The</strong> distinctive pattern <strong>of</strong> NOS expression in toxin-exposed<br />

brains suggested the possibility that NOS was localized on or in glia under these conditions.<br />

<strong>The</strong>refore, I tested the hypothesis that the toxin was causing glial death and thus indirectly<br />

affecting neuron survival. I employed a GAL4 driver controlled by the promoter <strong>of</strong> the gene<br />

reversed polarity (repo) (Halter et al., 1995) to direct the expression <strong>of</strong> GFP in all glial cells to<br />

test this hypothesis. I exposed Repo-GAL4; UAS-eGFP flies to conditioned mediums <strong>of</strong> S.<br />

colicolor and S. venezuelae for 24 hr. I found that in the presence <strong>of</strong> the control conditioned<br />

medium <strong>of</strong> S. colicolor, glial cells, identified by GFP expression, showed mild induction <strong>of</strong> NOS<br />

(Fig. 5.5 A, thin arrow). In contrast, the brains exposed to crude conditioned medium <strong>of</strong> S.<br />

venezuelae showed the presence <strong>of</strong> NOS signal in the close vicinity <strong>of</strong> glial cell bodies or even<br />

surrounding the glial cell bodies, but I did not observe a widespread co-localization <strong>of</strong> signal<br />

(Fig. 5.5 B and Fig.5.5 D, dotted arrow). Occasionally, however, I did observe apparent co-<br />

localized GFP and NOS signal, which suggests either a regionally specific induction <strong>of</strong> NOS in<br />

glia, or alternatively, the possible engulfment <strong>of</strong> glial cells by NOS-expressing structures (Fig.<br />

5.5 D, arrows). In either case, the positioning <strong>of</strong> the NOS signal and the occasional co-<br />

localization <strong>of</strong> signal support the hypothesis that toxin-induced neuronal death is a by-product <strong>of</strong><br />

damaged glia.<br />

157


S. colicolor<br />

S. venezuelae<br />

Figure 5.5: Exposure <strong>of</strong> S. venezuelae conditioned medium causes expression <strong>of</strong> NOS around<br />

glial cells in adult brain. Repo-GAL4;UAS-eGFP flies were exposed to conditioned mediums <strong>of</strong><br />

S. colicolor (A) and S. venezuelae (B) for 24 hr. (A) <strong>The</strong>re was minimal expression <strong>of</strong> NOS in<br />

the presence <strong>of</strong> crude conditioned medium <strong>of</strong> S. colicolor. However, there was a increased<br />

expression <strong>of</strong> NOS signal in brains exposed to conditioned medium <strong>of</strong> S. venezuelae (B). Arrows<br />

(thin) in A,B indicate NOS expression. (C, D) Enlarged images <strong>of</strong> insets for A,B. Arrows<br />

indicate co-localization (yellow) for NOS-signal (red) and glial cells (green) suggesting that<br />

NOS may be associated with induced by glial cells or affecting glia. <strong>The</strong> dotted arrow shows the<br />

expression <strong>of</strong> NOS around glial cell(s). Scale bar= 31.75 μm.<br />

B<br />

158


DISCUSSION<br />

In this report, I have used our robust Drosophila model to identify the mechanism <strong>of</strong><br />

action <strong>of</strong> the crude preparation <strong>of</strong> secondary metabolites from Streptomyces venezuelae on<br />

dopaminergic neurons in Drosophila. S. venezuelae belongs to a class <strong>of</strong> gram-positive bacteria,<br />

Actinomycetes, well known to play a crucial role in the degradation <strong>of</strong> organic matter,<br />

maintaining the ecosystem by forming an important soil microbial community (Kennedy, 1999;<br />

Buckley and Schmidt, 2003). S. venezuelae is also an important species in the phylum<br />

Actinobacteria, known to produce many bioactive molecules, many <strong>of</strong> which are currently used<br />

as antibiotics (Bradley and Ritzi, 1968). <strong>The</strong>se are chloramphenicol (He, 2001), methymycin and<br />

neomethymycin (Donin et al., 1954; Perlman and O'Brien, 1954). Although many <strong>of</strong> the soil-<br />

residing bacteria are commercially important for their ability to produce antibiotics, recently<br />

some subspecies <strong>of</strong> Actinobacteria, such as Nocardia and even Streptomyces, were found to<br />

produce neurotoxic secondary metabolites (Barry and Beaman, 2007; Tam et al., 2002;<br />

unpublished findings <strong>of</strong> Caldwell Lab, UA). <strong>The</strong> Caldwell Lab recently discovered an effect <strong>of</strong><br />

S. venezuelae on the dopaminergic neurons <strong>of</strong> C. elegans and found that exposure to media<br />

containing bacterial secondary metabolites from this strain <strong>of</strong> common soil bacteria caused loss<br />

<strong>of</strong> dopaminergic neurons. Subsequently human DA culture cells, SHSY5Y were shown to be<br />

similarly affected (<strong>The</strong> Standaert Lab, unpublished observations). However, the mechanism<br />

through which the crude conditioned medium acts on DA neurons is still unknown.<br />

Exposure to crude conditioned medium <strong>of</strong> S. venezuelae causes truncation <strong>of</strong> life and<br />

Parkisonian features in wild type flies<br />

We recently established a Drosophila model for PD that recapitulates essential features <strong>of</strong><br />

PD, such as tremors, bradykinesia, postural instability and loss <strong>of</strong> DA neurons (Chaudhuri et al.,<br />

159


2007). I, therefore, used our in vivo fly model to test whether exposure to crude conditioned<br />

medium induced Parkinsonian feature equivalent to those caused by PQ exposure by assessing<br />

lifespan, mobility defects and loss <strong>of</strong> DA neurons. We used the S. colicolor species and 5%<br />

sucrose-fed flies as the controls, since they are devoid <strong>of</strong> DA neurotoxicity in C. elegans and<br />

human DA cell lines, SHSY5Y (unpublished data, Caldwell Lab and Standaert Lab). I also found<br />

a similar lack <strong>of</strong> toxic effects <strong>of</strong> crude conditioned medium <strong>of</strong> S. colicolor in flies. I found that<br />

exposure to the crude conditioned medium <strong>of</strong> S. venezuelae drastically reduces survival duration<br />

relative to the crude conditioned medium <strong>of</strong> S. colicolor. <strong>The</strong> average survival <strong>of</strong> adults exposed<br />

to S. venezuelae was only 4 days (Fig. 5.1). Moreover, the flies exposed to S. venezuelae<br />

demonstrated tremors and bradykinesia, as assayed by a negative geotaxis assay (Chaudhuri et<br />

al., 2007). <strong>The</strong> tremors we found in flies exposed to high (20 mM) dose <strong>of</strong> PQ were severe and<br />

were seen within 6 hr <strong>of</strong> exposure to 20 mM PQ (Chaudhuri et al., 2007). Interestingly, the flies<br />

exposed to the crude conditioned medium <strong>of</strong> S. venezuelae showed mild tremors and<br />

bradykinesia only after 72-96 hr <strong>of</strong> exposure to the toxic conditioned medium when lethal effects<br />

were also evident (Fig. 5.1, Fig. 5.2). This implies that the mechanism <strong>of</strong> bacterial-induced<br />

neurotoxicity could be different from PQ-generated toxicity. In addition to the Parkinsonian<br />

symptoms, we also found that upon continuous exposure <strong>of</strong> the crude conditioned medium from<br />

S. venezuelae and S. colicolor up to 48 hr, a significant loss <strong>of</strong> DA neuron loss in PPM2, PPM3<br />

and PPL1 was apparent. However, neuron loss in the PPM1 and PPL2 subgroups was evident<br />

only after 48 hr <strong>of</strong> exposure to S. venezuelae conditioned medium relative to S. colicolor<br />

conditioned medium treated and 5% sucrose-fed brains (data not shown). We used crude<br />

conditioned medium in these experiments; it is possible that exposure to more purified extract<br />

possessing a higher concentration <strong>of</strong> the biologically active toxic molecule may induce loss <strong>of</strong><br />

160


DA neuron loss much earlier. Our result and the studies performed on mice inoculated with<br />

Nocardia asteroides strain GUH-2 show typical Parkinsonian features upon chronic exposure.<br />

<strong>The</strong> results demonstrate parallel development <strong>of</strong> Parkinsonian-like symptoms by certain species<br />

in the Actinobacteria phylum and suggest a possibility that these species play an etiological role<br />

in PD (Kohbata and Beaman, 1991). Moreover, many other bacterial species are reported to<br />

produce Parkinsonian symptoms (Wszolek et al., 1988; Kim et al., 1995).<br />

Exposure to the crude conditioned medium <strong>of</strong> S. venezuelae induces NOS dependent response in<br />

adult brain<br />

<strong>The</strong> processes <strong>of</strong> neuroinflammation are recently being considered as a primary<br />

mechanism in the etiology and pathophysiology <strong>of</strong> PD. We recently found that exposure to PQ is<br />

associated with the induction <strong>of</strong> NOS activity and expression <strong>of</strong> NOS-positive cells in adult<br />

Drosophila brains. <strong>The</strong> NOS-positive cells were also found to surround the DA neurons,<br />

apparently mediating the loss <strong>of</strong> DA neurons in vivo (<strong>Inamdar</strong> et al., <strong>2009</strong>, under revision).<br />

<strong>The</strong>refore, I hypothesized that like PQ, exposure to crude conditioned medium <strong>of</strong> S. venezuelae<br />

also would mediate DA neuron loss by inducing NOS. To test this, I used universal NOS<br />

antibody (Sigma) known to recognize conserved domains in NOS is<strong>of</strong>orms among different<br />

species including Drosophila, human and mouse. As discussed in Chapter 2, this antibody has<br />

been previously used to detect the role <strong>of</strong> NOS in the development <strong>of</strong> visual system during<br />

metamorphosis, Drosophila malphigian tubules and hemocytes (Davies, 2000; Gibbs, 2001;<br />

Foley and O’Farrell, 2003).<br />

I found a significant level <strong>of</strong> induction <strong>of</strong> NOS in response to exposure to crude<br />

conditioned medium <strong>of</strong> S. venezuelae in adult Drosophila brain, but almost no induction <strong>of</strong> NOS<br />

with S. colicolor crude conditioned medium near DA neurons but never found to be surrounding<br />

161


the DA neurons (Fig. 5.4 A, B). Although further quantification data for NOS expression is<br />

needed to confirm the observation, based on these results it is to be speculated that various<br />

species in the Actinomyces genus mediate DA toxicity with different mechanism(s).<br />

Exposure to crude conditioned medium <strong>of</strong> S. venezuelae induces NOS near glial cells in adult<br />

brain<br />

Upon exposure to PQ, NOS-signal was found surrounding the DA neurons and seem to<br />

be correlated with neuronal death (<strong>Inamdar</strong> et al., <strong>2009</strong>, under revision). However, we never<br />

found similar patterns around DA neurons during exposure to crude conditioned medium <strong>of</strong> S.<br />

venezuelae (Fig. 5.4 B). Like mammalian brain, Drosophila brain contains a large population <strong>of</strong><br />

glial cells that play important roles in axonal guidance, ensheathment and neuronal trophic<br />

support (Booth et al., 2000; Hidalgo and Booth, 2000). It has been proposed that in adult<br />

Drosophila brain, glial cells generate different bio- active molecules to support neuronal growth<br />

and development (Freeman and Doherty, 2006). We found that the NOS signal induced by the<br />

crude conditioned medium <strong>of</strong> S. venezuelae for 24 hr surrounded glial cells and some instances<br />

<strong>of</strong> co-localized signal suggests the possibility that NOS-expressing cells are engulfing glia or that<br />

a regionally-specific subset <strong>of</strong> glia are producing NOS (Fig. 5.5 B, D). Even though NOS<br />

expression was found occasionally in brains exposed to S. colicolor crude conditioned medium, I<br />

never observed co-localization <strong>of</strong> signal (Fig. 5.5 A, C). In mammalian in vitro studies, survival<br />

<strong>of</strong> transplanted DA neurons has been shown to be significantly improved by glial subtype,<br />

oligodendrocyte-type 2 astrocyte-derived trophic factors (Sortwell et al., 2000). Although<br />

reactive gliosis has been reported in PD induced via different neurotoxic agents including PQ,<br />

the mechanism through which S. venezuelae induces PD features seems to be different and<br />

possibly may occur indirectly via degeneration <strong>of</strong> glia. However, further studies will be required<br />

162


to directly address the glial link to neurodegeneration in this case. Finally, it is possible that<br />

altered response may be seen against the purified extract/component from crude conditioned<br />

medium <strong>of</strong> S. venezuelae. Nevertheless, so far these results present preliminary data implying the<br />

crucial role <strong>of</strong> NOS in the pathogenesis <strong>of</strong> S. venezuelae crude conditioned medium- induced DA<br />

neuron loss.<br />

163


CHAPTER 6<br />

SUMMARY, FUTURE DIRECTIONS AND APPLICATIONS<br />

Drosophila and neuroinflammatory-like response<br />

Neuroinflammation is considered a crucial component <strong>of</strong> chronic neurodegenerative<br />

diseases including PD. Neuroinflammation is characterized by activation <strong>of</strong> immune cells in<br />

mammals where microglia act as a primary line <strong>of</strong> defense against non-self and pathological<br />

invaders. Microglia are bone marrow-derived macrophage-lineage cells and constitute about<br />

12% <strong>of</strong> the total number <strong>of</strong> cells in the human brain. <strong>The</strong>se cells appear in small numbers in the<br />

resting state, in certain regions <strong>of</strong> the brain (Sugam et al., 2003; Zhang et al., 2005). Under<br />

potentially pathological conditions such as injury and infection, these cells alter their<br />

morphology and number, and express myriad inflammatory cytokines/chemokines and other<br />

enzymes to combat such pathological conditions. Moreover, neuronal and non-neuronal cell<br />

populations are proposed to induce inflammatory mediators and further interact with each other<br />

during the process <strong>of</strong> neuroinflammation (Mosley et al., 2006; Block et al., 2007).<br />

Parkinson’s disease, the second most common chronic neurodegenerative disease, has<br />

been demonstrated to be associated with altered microglial morphology and number along with<br />

induction <strong>of</strong> cytokines/chemokines such as IL-1, IL-6, TNF-α and enzymes COX 1/2, NOS in<br />

mammalian model and PD patients (Mogi et al., 1994; Blum-Dgen et al., 1995; Knott et al.,<br />

2000). It is still debated whether the neuroinflammation/activation <strong>of</strong> microglia is a beneficial or<br />

detrimental process in PD (Wyss-Coray and Mucke, 2002; McGeer and McGeer, 2004). <strong>The</strong><br />

current notion is that in acute pathological conditions, the mild to moderate activation <strong>of</strong><br />

164


microglia play a crucial role in maintaining homeostasis in CNS by scavenging toxins, removing<br />

dying cells and debris, and thereby, promoting wound healing. However, in chronic pathological<br />

conditions, especially in chronic neurodegenerative diseases including PD, the activated<br />

microglia display elevated levels <strong>of</strong> response ultimately causing damage to viable host tissue in<br />

addition to dead one. In several mammalian PD models, pharmacological and genetic<br />

suppression <strong>of</strong> the inflammatory mediators known to be expressed by activated microglia are<br />

reported to delay the DA neuron loss in different mammalian models suggesting that induction <strong>of</strong><br />

the neuroinflammatory process is detrimental in pathogenesis <strong>of</strong> PD (Wu et al., 2002; Esposito et<br />

al., 2007; Vafeiadou et al., 2007). However, it appears that sustained activation <strong>of</strong> microglia in<br />

PD is a downstream event. <strong>The</strong> factors or stimuli that trigger the events leading to alteration <strong>of</strong><br />

the beneficial roles <strong>of</strong> microglia to detrimental responses are not well understood. Moreover, it is<br />

unknown whether the genes known to play a crucial role in the pathogenesis <strong>of</strong> PD also show<br />

modulatory roles in the PD-associated neuroinflammatory process. Furthermore, the key signal<br />

transduction pathways controlling the different aspects <strong>of</strong> microglia activation such as alteration<br />

in morphology, proliferation, up-regulation <strong>of</strong> inflammatory mediators, phagocytosis etc. are not<br />

yet identified. Finally, the characterization <strong>of</strong> the overall impact <strong>of</strong> any triggering event for<br />

microglial activation in the development and survival <strong>of</strong> an organism is difficult in mammalian<br />

models. <strong>The</strong>refore, a simple genetic model for this response would be <strong>of</strong> great benefit for<br />

analysis <strong>of</strong> the role <strong>of</strong> neuroinflammation in neurodegenerative disease.<br />

My research project has laid the foundation for future projects for the exploration <strong>of</strong> a<br />

previously unknown process in the Drosophila Parkinson’s disease model that seems to parallel<br />

in some respects, mammalian neuroinflammation. My research has pioneered crucial preliminary<br />

data supporting the presence <strong>of</strong> a PQ-induced NOS-dependent response in the adult Drosophila<br />

165


ain. As discussed above, we recently established a PQ-induced PD model based on ingestion <strong>of</strong><br />

PQ in Drosophila (Chaudhuri et al., 2007). PQ has been proposed to act as a potent redox cycler<br />

in the presence <strong>of</strong> diaphorases, electron donors, to generate superoxide radicals, which further<br />

give rise to other reactive oxygen and nitrogen species (Dinis-Oliveira et al., 2006). NOS also is<br />

known to function as a diaphorase to mediate PQ toxicity in vitro (Day et al., 1999); however,<br />

the function <strong>of</strong> NOS at the whole organism level in PQ-mediated neurotoxicity has never been<br />

identified in Drosophila. Drosophila possesses only one NOS gene (Regulski and Tully, 1995).<br />

Since mammalian studies have identified NOS as a well-known marker for microglial activation<br />

and as a mediator <strong>of</strong> potentially deleterious activity in PD pathogenesis, I hypothesized that NOS<br />

also plays an important toxic effect in the PQ-induced Drosophila PD model. I found that PQ-<br />

induced toxicity was at least partly mediated via generation <strong>of</strong> nitric oxide as L-NAME, an<br />

inhibitor <strong>of</strong> NOS, was able to improve survival during PQ exposure. I further found that PQ<br />

ingestion activates NOS in adult Drosophila brain. Interestingly, in control (non-treated) adult<br />

brain, NOS signal is mainly confined to the junction <strong>of</strong> the central complex and optic lobe.<br />

However, in PQ-treated brain, this NOS signal was found to be closely associated with the DA<br />

neurons in the central complex in many instances. Further, we found that NOS signal was<br />

surrounding the DA neurons and the presence <strong>of</strong> such NOS signal appeared to correlate with<br />

neuronal death as such neurons were almost uniformly abnormal in morphology. We further<br />

determined that NOS was not expressed in glial cells, taking advantage <strong>of</strong> reports expressed only<br />

in glial cells. I also used minocycline, a well known drug reported to suppress the induction <strong>of</strong><br />

inducible NOS in mammalian models to further characterize the toxic role <strong>of</strong> NOS in PQ-<br />

mediated toxicity in Drosophila. Moreover, minocycline was found to mediate both anti-oxidant<br />

and anti-inflammatory properties against PQ in Drosophila. Although these results are<br />

166


promising, further experiments are required to confirm the NOS expression in Drosophila CNS.<br />

NO has been reported to play a important role in several biological processes including<br />

development <strong>of</strong> the nervous system (Truman et al., 1996; Wildemann and Bicker, 1999),<br />

synaptogenesis (Enikolopov et al., 1999) and immunity (Nappi et al., 2000). However, detection<br />

<strong>of</strong> NOS response in the Drosophila PD model, which is seemingly analogous to the mammalian<br />

microglial response in PD pathogenesis, has opened new avenues to explore the previously<br />

unknown NOS mediated, presumably neuroinflammatory response, in an in vivo Drosophila<br />

model <strong>of</strong> neurodegeneration. However, confirmation <strong>of</strong> these results by employing other NOS<br />

detection methods such as NADPH diaphorase histochemical assay is needed. Furthermore,<br />

increased NOS signal close to DA neurons needs quantification data to determine whether PQ<br />

also causes increase in the number <strong>of</strong> NOS positive structures, since I was unable to determine<br />

whether these structures were comprised <strong>of</strong> individual cells. <strong>The</strong> most likely candidate for this<br />

activity is the Drosophila innate immune cells, the hemocytes (see below), which are known to<br />

express NOS in response to bacterial infection and which frequently form cellular aggregates in<br />

such responses (Foley and O’Farrell, 2003). Further studies are in progress and will set the stage<br />

for future studies to further explore the role <strong>of</strong> NOS in our Drosophila PD model. <strong>The</strong><br />

preliminary result showing that glial cells lack the expression <strong>of</strong> NOS by performing<br />

immunolocalization studies to detect the co-localization signal for glial and NOS signal has also<br />

provided the directions for identifying the source <strong>of</strong> activated NOS in response to PQ.<br />

Like mammals, Drosophila possesses a robust host defense system against<br />

microorganisms. <strong>The</strong> anti-microbial peptides are present in the epidermis <strong>of</strong> the digestive system,<br />

genital tract, tracheal and malphigian tubules. <strong>The</strong>se peptides inhibit microbial growth<br />

(Ferrandon et al., 1998; Tzou et al., 2000; Onfelt Tingvall et al., 2001). Microbes that escape the<br />

167


ody wall-mediated first line <strong>of</strong> defense are subject to further innate immune responses after<br />

entering into the general body cavity or hemocoele. In general, Drosophila exhibits two types <strong>of</strong><br />

immune responses: a humoral response involving production <strong>of</strong> antimicrobial peptides in the fat<br />

body (equivalent to the mammalian liver), and cellular immunity dependent on phagocytosis by a<br />

macrophage-like class <strong>of</strong> hemocytes (Drosophila blood cells), the plasmatocytes, and<br />

encapsulation by crystal cells and/or lamellocytes, the remaining two classes <strong>of</strong> hemocytes<br />

(Rizki, 1962; H<strong>of</strong>fmann and Reichhart, 2002; Kim and Kim, 2005; Lemaitre and H<strong>of</strong>fmann,<br />

2007). Drosophila cellular immune response has been studied in detail at embryonic and larval<br />

stages. Three classes <strong>of</strong> hemocytes corresponding to mammalian hematopoietic lineage cells<br />

including microglia also arise from mesoderm and mediate cellular immune response in<br />

Drosophila. <strong>The</strong> three cell types, the plasmatocytes, lamellocytes and crystal cells, are distinct in<br />

morphology and function (Evans et al., 2003; Jung et al., 2005). Plasmatocytes comprise 90-95%<br />

<strong>of</strong> all mature hemocytes and appear as small rounded cells with alteration to size in response to<br />

phagocytosis. <strong>The</strong> developmental and functional similarity <strong>of</strong> plasmatocytes, known to represent<br />

the only class <strong>of</strong> hemocytes in adult Drosophila, could be hypothesized to act like mammalian<br />

microglia in response to chronic neurodegenerative diseases including PD. This hypothesis could<br />

be tested by employing transgenic lines generated using the promoter regions for the genes<br />

associated with hemocytes, hemese, collagen and hemolectin (Yasothornsrikul et al., 1997; Goto<br />

et al., 2002; Kurucz et al., 2002; Asha et al., 2003; Zettervall et al., 2004); these experiments<br />

have been initiated. Furthermore, detection <strong>of</strong> plasmatocytes could be confirmed by using P1<br />

antibody (Asha et al., 2003). We hope to identify the source <strong>of</strong> NOS induction in response to PQ<br />

so that mentioned unanswered questions discussed above from the mammalian studies could be<br />

further determined in our in vivo Drosophila model.<br />

168


In addition, we can further explore with this model the role <strong>of</strong> other inflammatory<br />

mediators such as TNF and cytokines and COX-1/2 enzyme. Eiger, upd-3 and dhf, and pxt are<br />

the homologs for TNF, cytokine and COX-1/2, respectively, known to present in Drosophila<br />

(Igaki et al., 2002; Malagoli et al., 2008; Tootle and Spalding, 2008). It would be interesting to<br />

know the response <strong>of</strong> these genes to PQ by using loss-<strong>of</strong>-function mutants and transgenic lines<br />

available at Bloomington Stock center.<br />

Having found that PQ ingestion induces activation <strong>of</strong> NOS, one <strong>of</strong> the important<br />

inflammatory mediators for inflammatory response, I further performed studies showing the<br />

implementation <strong>of</strong> this model to understand the NOS-associated disease mechanisms in our in<br />

vivo disease model. In C. elegans and SHSY5Y cell lines, loss <strong>of</strong> DA neurons by medium<br />

containing secondary metabolites (or crude conditioned medium) <strong>of</strong> S. venezuelae has been<br />

detected (<strong>The</strong> Caldwell lab, UA, and <strong>The</strong> Standaert Lab, UAB). I further confirmed the observed<br />

neurotoxic effects <strong>of</strong> crude conditioned medium <strong>of</strong> S. venezuelae in Drosophila. <strong>The</strong> ingestion <strong>of</strong><br />

crude conditioned medium <strong>of</strong> S. venezuelae to flies caused truncation <strong>of</strong> life and mobility defects<br />

seen with PQ ingestion. However, the observed death trend with S. venezuelae crude conditioned<br />

medium on wild type flies was different from those on PQ. <strong>The</strong> S. venezuelae crude conditioned<br />

medium exposed flies showed a gradual decline in survival with a sudden peak in the death rate<br />

at later half <strong>of</strong> total survival duration whereas in PQ exposed flies, the rate the wild type flies<br />

died was almost the same at each time point (Fig. 5.1). This suggests that upon exposure to<br />

conditioned medium <strong>of</strong> S. venezuelae, an accumulation <strong>of</strong> toxic substances occur, under the<br />

influence <strong>of</strong> which flies succumb in a delayed response.<br />

Interestingly, the flies exposed to the crude conditioned medium <strong>of</strong> S. venezuelae showed<br />

mild tremors and bradykinesia only in the latter half <strong>of</strong> the total survival duration when lethal<br />

169


effects were also evident. This result further indicates that the mechanism <strong>of</strong> bacterial-induced<br />

neurotoxicity could be different from PQ-generated toxicity. In addition to the Parkinsonian<br />

symptoms, we also found that continuous exposure <strong>of</strong> the crude conditioned medium from S.<br />

venezuelae caused loss <strong>of</strong> DA neurons, but this loss was not as rapid as that observed during<br />

exposure to 10 mM PQ. However, it is to be noted that I used crude conditioned medium in these<br />

experiments; it is possible that exposure to more purified extract possessing a higher<br />

concentration <strong>of</strong> the biologically active toxic molecule may induce loss <strong>of</strong> DA neuron much<br />

earlier. Studies to determine the active toxic molecule responsible for the neurotoxic effects by S.<br />

venezuelae crude conditioned medium are in progress.<br />

I further explored the ability <strong>of</strong> such media to induce a NOS-dependent response in adult<br />

fly brain. My preliminary results suggest that, as in PQ treatment, S. venezuelae secreted<br />

secondary metabolites mediate DA neuron loss partly by causing induction <strong>of</strong> NOS. <strong>The</strong> NOS<br />

signal was, however, not found closely associated with DA neuron cell bodies. Interestingly, the<br />

positioning <strong>of</strong> the NOS signal and the occasional co-localization <strong>of</strong> signal with the glial cells, the<br />

cells required for the maintaining trophic support and homeostasis for neurons including DA<br />

neurons supports the hypothesis that toxin-induced neuronal death is a by-product <strong>of</strong> damaged<br />

glia. Although further studies are required to directly link the DA neurodegeneration and glia in<br />

response to crude conditioned medium <strong>of</strong> S. venezuelae by performing quantification data for<br />

glial and the NOS positive structures, my results clearly indicate that further exploration <strong>of</strong><br />

disease mechanisms <strong>of</strong> S. venezuelae crude conditioned medium associated DA toxicity in in<br />

vivo Drosophila model will be a pr<strong>of</strong>itable avenue for further study. <strong>The</strong>se results are <strong>of</strong><br />

particular interest because, unlike the degenerative process in C. elegans which does not have<br />

comparable glial populations or in DA neurons cultured in the absence <strong>of</strong> glia, they suggest a<br />

170


potential involvement <strong>of</strong> non-neuronal cells in the CNS and thus add a new dimension to this<br />

research.<br />

Drosophila and signal transduction pathway in PD<br />

Signal transduction pathways mediate myriad downstream effects such as the regulation<br />

<strong>of</strong> gene transcription, the cell cycle, cellular differentiation and cell death (Nishida and Gotoh,<br />

1993; Chang and Karin, 2001). More recently, the roles <strong>of</strong> MAPK and Akt mediated signal<br />

transduction pathways have been elucidated in mammalian PD model. <strong>The</strong> results are somewhat<br />

controversial regarding whether these roles <strong>of</strong> MAPK are neuroprotective or neurotoxic in the<br />

pathogenesis <strong>of</strong> PD (Xia et al., 1995; Harper and LoGrasso, 2001; Peng et al., 2004; Niso-<br />

Santano et al., 2006; Burke, 2007). Moreover, these models have mainly employed in vitro (cell-<br />

culture) approach where the experimental conditions employed to investigate the roles <strong>of</strong> these<br />

signaling pathways, in particular pathogenic responses, are generally manipulated. Further, in<br />

vitro culture conditions lack appropriate cell-cell interactions from neighboring cells which could<br />

modulate the response to external stimuli. Moreover, many <strong>of</strong> the commercially available<br />

pharmacological inhibitors <strong>of</strong> kinases employed in these experiments are not well-defined with<br />

respect to their exact modes <strong>of</strong> action and may give rise to non-specific effects due to blockage<br />

<strong>of</strong> unknown target molecules (Sekiguchi et al., 1999; Learish et al., 2000; Waetzig and<br />

Herdegen, 2005).<br />

I used heterozygous loss-<strong>of</strong>-function mutants for JNK, Akt, ERK and the pro-apoptotic<br />

genes, reaper and caspase-9 to determine the role <strong>of</strong> these important genes in PQ toxicity in our<br />

in vivo Drosophila PD model. As opposed to mammalian studies, we found that ERK and reaper<br />

lack any observable regulatory roles while Akt and JNK provide protective role in our PD model.<br />

171


<strong>The</strong>se studies could be further confirmed by expressing dominant negative form <strong>of</strong> these in<br />

dopaminergic neurons. <strong>The</strong> protective effect <strong>of</strong> Akt parallels mammalian findings, however,<br />

interestingly indicate a protective role for JNK in this PD model, in contrast to mammalian PD<br />

models using PQ, 6-OHDA (Gearan et al., 2001; Ganguly et al., 2004; Peng et al., 2004). I also<br />

present data showing the failure <strong>of</strong> herterozygous Akt1 and JNK loss-<strong>of</strong>-function mutants to<br />

respond to minocycline action, while Akt1 and JNK wild type over-expression in DA neurons<br />

fails to provide additional benefit, suggesting that JNK and Akt 1 may operate in separate<br />

pathway(s) and act independently from those <strong>of</strong> minocycline. Generating the double mutants for<br />

Akt, JNK together and with caspase-9 may provide further insight into the regulatory effects <strong>of</strong><br />

these pathways in PQ toxicity. It is to be noted that in Drosophila, JNK has also been reported to<br />

play an important role in innate immune response and longevity (Boutros et al., 2002; Wang et<br />

al., 2003b). My results present the preliminary studies to further explore the role <strong>of</strong> JNK, Akt and<br />

other MAPK-related signaling pathway components in the NOS-dependent response against PQ<br />

ingestion.<br />

Drosophila as a model to study Early Onset Parkinsonism<br />

Recently, idiopathic form <strong>of</strong> PD has been diagnosed in the under 40 age group. Such<br />

cases are classified as Early Onset Parkinsonian cases. I postulated that brief exposure to PQ<br />

early in life might contribute to the pathogenesis <strong>of</strong> PD at later stage <strong>of</strong> life. We used larvae and<br />

adult flies <strong>of</strong> less that 6 hr age to represent the juvenile and young aged population. Exposure to<br />

10 mM and 1 mM PQ for even a brief period (12 hr) at larval and young age resulted in<br />

truncation <strong>of</strong> life span, mobility defects and DA neuron loss in a dose dependent manner at later<br />

stage <strong>of</strong> life. It will be important to pursue the question <strong>of</strong> whether this early event also perturbs<br />

172


DA homeostasis in the adult brain as well as other metabolic processes that may lead to<br />

increased sensitivity towards such PD inducers as well as early death. Further, taking into<br />

account our discovery <strong>of</strong> the NOS-dependent response in PQ treated adult brains, it would be<br />

interesting to know if a similar response is seen in larvae, and if so whether a sustained response<br />

is seen at the later time point. As discussed above, this NOS dependent response needs other<br />

confirmatory experiments, and these are to be completed before further exploration <strong>of</strong> the effects<br />

<strong>of</strong> juvenile exposure to this toxin. However, my data strongly support the current findings for the<br />

role <strong>of</strong> exposure <strong>of</strong> neurotoxins as one <strong>of</strong> the insults inflicted early in life in triggering or<br />

accelerating the pathogenic events for PD (Liu et al., 2003; Liu and Hong, 2003; Logroscino,<br />

2005).<br />

173


REFERENCES<br />

Alfonso TB, Jones BW (2002) gcm2 promotes glial cell differentiation and is required with glial<br />

cells missing for macrophage development in Drosophila. Dev Biol 248:369-383.<br />

Altschuler E (1996) Gastric Helicobacter pylori infection as a cause <strong>of</strong> idiopathic Parkinson<br />

disease and non-arteric anterior optic ischemic neuropathy. Med Hypotheses 47:413-414.<br />

Asha H, Nagy I, Kovacs G, Stetson D, Ando I, Dearolf CR (2003) Analysis <strong>of</strong> ras-induced<br />

overproliferation in Drosophila hemocytes. Genetics 163:203-215.<br />

Ashkenazi A (2002) Targeting death and decoy receptors <strong>of</strong> the tumour-necrosis factor<br />

superfamily. Nat Rev Cancer 2:420-430.<br />

Ashkenazi A, Dixit VM (1998) Death receptors: signaling and modulation. Science 281:1305-<br />

1308.<br />

Barry DP, Beaman BL (2007) Nocardia asteroides strain GUH-2 induces proteasome inhibition<br />

and apoptotic death <strong>of</strong> cultured cells. Res Microbiol 158:86-96.<br />

Batchelor PE, Liberatore GT, Wong JY, Porritt MJ, Frerichs F, Donnan GA, Howells DW<br />

(1999) Activated macrophages and microglia induce dopaminergic sprouting in the injured<br />

striatum and express brain derived neurotrophic factor and glial cell line-derived<br />

neurotrophic factor. J Neurosci 19:1708-1716.<br />

Batchelor PE, Porritt MJ, Martinello P, Parish CL, Liberatore GT, Donnan GA, Howells DW<br />

(2002) Macrophages and microglia produce local trophic gradients that stimulate axonal<br />

sprouting toward but not beyond the wound edge. Mol Cell Neurosci 21:436-453.<br />

174


Beaman BL, Canfield D, Anderson J, Pate B, Calne D (2000) Site-specific invasion <strong>of</strong> the basal<br />

ganglia by Nocardia asteroides GUH-2. Med. Microbiol Immunol 188:161-168.<br />

Beitner-Johnson D, Nestler EJ (1991) Morphine and cocaine exert common chronic actions on<br />

tyrosine hydroxylase in dopaminergic brain reward regions. J Neurochem 57:344-347.<br />

Berman KF, Illowsky BP, Weinberger DR (1988) Physiological dysfunction <strong>of</strong> dorsolateral<br />

prefrontal cortex in schizophrenia. IV. Further evidence for regional and behavioral<br />

specificity Arch Gen Psychiatry 45:616-622.<br />

Berridge KC, Robinson TE (1998) What is the role <strong>of</strong> dopamine in reward: hedonic impact,<br />

reward learning, or incentive salience? Brain Res Brain Res Rev 28:309-369.<br />

Betarbet R, Sherer TB, MacKenzie G, Garcia-Osuna M, Panov AV, Greenamyre JT (2000)<br />

Chronic systemic pesticide exposure reproduces features <strong>of</strong> Parkinson's disease. Nat<br />

Neurosci 3:1301-1306.<br />

Beuge J, Aust SD (1978) Microsomal lipid peroxidation. Methods Enzymol 52:302-310.<br />

Bewley GC, Nahmias JA, Cook JL (1983) Developmental and tissue-specific control <strong>of</strong> catalase<br />

expression in Drosophila melanogaster: correlations with rates <strong>of</strong> enzyme synthesis and<br />

degradation. Dev Genet 4:49-60.<br />

Birman S, Morgan B, Anzivino M, Hirsh J (1994) A novel and major is<strong>of</strong>orm <strong>of</strong> tyrosine<br />

hydroxylase in Drosophila is generated by alternative RNA processing. J Biol Chem<br />

269:26559-26567.<br />

Biswas SC, Ryu E, Park C, Malagelada C, Greene LA (2005) Puma and p53 play required roles<br />

in death evoked in a cellular model <strong>of</strong> Parkinson disease. Neurochem Res 30:839-845.<br />

Björklund A, Dunnett SB (2007) Dopamine neuron systems in the brain: an update. Trends<br />

Neurosci 30:194-202.<br />

175


Blenis J (1993) Signal transduction via the MAP kinases: proceed at your own RSK. Proc Natl<br />

Acad Sci USA 90:5889-5892.<br />

Block ML, Zecca L, Hong JS (2007) Microglia-mediated neurotoxicity: uncovering the<br />

molecular mechanisms. Nat Rev Neurosci 8:57-69.<br />

Boileau I, Assaad JM, Pihl RO, Benkelfat C, Leyton M, Diksic M, Tremblay RE, Dagher A<br />

(2003) Alcohol promotes dopamine release in the human nucleus accumbens Synapse<br />

49:226-231.<br />

Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, Krieger E, Dekker MC, Squitieri F,<br />

Ibanez P, Joosse M, van Dongen JW, Vanacore N, van Swieten JC, Brice A, Meco G, van<br />

Duijn CM, Oostra BA, Heutink P (2003) Mutations in the DJ-1 gene associated with<br />

autosomal recessive early-onset parkinsonism. Science 299:256-259.<br />

Bonilla E, Medina-Leendertz S, Villalobos V, Molero L, Bohórquez A (2006) Paraquat-induced<br />

oxidative stress in Drosophila melanogaster: effects <strong>of</strong> melatonin, glutathione, serotonin,<br />

minocycline, lipoic acid and ascorbic acid. Neurochem Res 31:1425-32.<br />

Bonneh-Barkay D, Reaney SH, Langston WJ, Di Monte DA (2005) Redox cycling <strong>of</strong> the<br />

herbicide paraquat in microglial cultures. Brain Res Mol Brain Res 134:52-56.<br />

Booth GE, Kinrade EF, Hidalgo A (2000) Glia maintain follower neuron survival during<br />

Drosophila CNS development. Development 127:237-244.<br />

Borchardt RT, Reid JR, Thakker DR (1976) Catechol Omethyltransferase. Mechanism <strong>of</strong><br />

inactivation by 6- hydroxydopamine. J Med Chem 19:1201-1209.<br />

Boutros M, Agaisse H, Perrimon N (2002) Sequential activation <strong>of</strong> signaling pathways during<br />

innate immune responses in Drosophila. Dev Cell 3:711-722.<br />

176


Bradley SG, Ritzi D (1968) Composition and ultrastructure <strong>of</strong> Streptomyces venezuelae. J<br />

Bacteriol 95:2358-2364.<br />

Brand AH, Perrimon N (1993) Targeted gene expression as a means <strong>of</strong> altering cell fates and<br />

generating dominant phenotypes. Development 118:401–415.<br />

Brazil DP, Hemmings BA (2001) Ten years <strong>of</strong> protein kinase B signalling: a hard Akt to follow.<br />

Trends Biochem Sci 26:657-664.<br />

Bredt DS, Hwang PM, Glatt CE, Lowenstein C, Reed RR, Snyder SH (1991) Cloned and<br />

expressed nitric oxide synthase structurally resembles cytochrome P-450 reductase. Nature<br />

351:714-718.<br />

Bredt DS, Snyder SH (1994) Nitric oxide: a physiologic messenger molecule. Annu Rev<br />

Biochem 63:175-195.<br />

Broderick KE, MacPherson MR, Regulski M, Tully T, Dow JA, Davies SA (2003) Interactions<br />

between epithelial nitric oxide signaling and phosphodiesterase activity in Drosophila. Am<br />

J Physiol Cell Physiol 285:C1207-C1218.<br />

Brooks AI, Chadwick CA, Gelbard HA, Cory-Slechta DA, Feder<strong>of</strong>f HJ (1999) Paraquat elicited<br />

neurobehavioral syndrome caused by dopaminergic neuron loss. Brain Res 823:1-10.<br />

Broxmeyer L (2002) Parkinson's: another look. Med Hypotheses 59:373-377.<br />

Brunner D, Dücker K, Oellers N, Hafen E, Scholz H, Klämbt C (1994) <strong>The</strong> ETS domain protein<br />

pointed-P2 is a target <strong>of</strong> MAP kinase in the sevenless signal transduction Nature 370:386-<br />

389.<br />

Buckley DH, Schmidt TM (2003) Diversity and dynamics <strong>of</strong> microbial communities in soils<br />

from agro-ecosystems. Environ Microbiol 5:441-452.<br />

177


Budnik V, White K (1988) Catecholamine-containing neurons in Drosophila melanogaster:<br />

distribution and development. J Comp Neurol 268:400-413.<br />

Burke RE (2007) Inhibition <strong>of</strong> mitogen-activated protein kinase and stimulation <strong>of</strong> Akt kinase<br />

signaling pathways: Two approaches with therapeutic potential in the treatment <strong>of</strong><br />

neurodegenerative disease. Pharmacol <strong>The</strong>r 114:261-277.<br />

Burnett AL (1995) Role <strong>of</strong> nitric oxide in the physiology <strong>of</strong> erection. Biol Reprod 52:485-489.<br />

Burns RS, Chiueh CC, Markey SP, Ebert MH, Jacobowitz DM, Kopin IJ (1983) A primate<br />

model <strong>of</strong> parkinsonism: selective destruction <strong>of</strong> dopaminergic neurons in the pars compacta<br />

<strong>of</strong> the substantia nigra by N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. Proc Natl Acad<br />

Sci USA 80:4546-4550.<br />

Bus JS, Aust SD, Gibson JE (1974) Superoxide- and singlet oxygen-catalyzed lipid peroxidation<br />

as a possible mechanism for paraquat (methyl viologen) toxicity. Biochem Biophys Res<br />

Commun 58:749-755.<br />

Cavanaugh JE (2004) Role <strong>of</strong> extracellular signal regulated kinase 5 in neuronal survival. Eur J<br />

Biochem 271:2056-2059.<br />

Cavanaugh JE, Jaumotte JD, Lakoski JM, Zigmond MJ (2006) Neuroprotective role <strong>of</strong> ERK1/2<br />

and ERK5 in a dopaminergic cell line under basal conditions and in response to oxidative<br />

stress. J Neurosci Res 84:1367-1375.<br />

Cha GH, Kim S, Park J, Lee E, Kim M, Lee SB, Kim JM, Chung J, Cho KS (2005) Parkin<br />

negatively regulates JNK pathway in the dopaminergic neurons <strong>of</strong> Drosophila. Proc Natl<br />

Acad Sci USA 102:10345-10350.<br />

Chang L, Karin M (2001) Mammalian MAP kinase signaling cascades. Nature 410:37-40.<br />

178


Chaudhuri A, Bowling K, Funderburk C, Lawal H, <strong>Inamdar</strong> A, Wang Z, O'Donnell JM (2007)<br />

Interaction <strong>of</strong> genetic and environmental factors in a Drosophila parkinsonism model. J<br />

Neurosci 27:2457-2467.<br />

Chauhan D, Li G, Hideshima T, Podar K, Mitsiades C, Mitsiades N, Munshi N, Kharbanda S,<br />

Anderson KC (2002) JNK-dependent release <strong>of</strong> mitochondrial protein, Smac, during<br />

apoptosis in multiple myeloma (MM) cells. J Biol Chem 278:17593-17596.<br />

Chen H, Zhang SM, Hernán MA, Schwarzschild MA, Willett WC, Colditz GA, Speizer FE,<br />

Ascherio A (2003) Nonsteroidal anti-inflammatory drugs and the risk <strong>of</strong> Parkinson disease.<br />

Arch Neurol 60:1059-1064.<br />

Chen M, Ona VO, Li M, Ferrante RJ, Fink KB, Zhu S, Bian J, Guo L, Farrell LA, Hersch SM,<br />

Hobbs W, Vonsattel JP, Cha JH, Friedlander RM (2000) Minocycline inhibits caspase-1<br />

and caspase-3 expression and delays mortality in a transgenic mouse model <strong>of</strong> Huntington<br />

disease. Nat Med 6:797-801.<br />

Chen PS, Peng GS, Li G, Yang S, Wu X, Wang CC, Wilson B, Lu RB, Gean PW, Chuang DM,<br />

Hong JS (2006) Valproate protects dopaminergic neurons in midbrain neuron/glia cultures<br />

by stimulating the release <strong>of</strong> neurotrophic factors from astrocytes. Mol Psychiatry 11:1116<br />

-1125.<br />

Chew SK, Akdemir F, Chen P, Lu WJ, Mills K, Daish T, Kumar S, Rodriguez A, Abrams JM<br />

(2004) <strong>The</strong> apical caspase Dronc governs programmed and unprogrammed cell death in<br />

Drosophila. Dev Cell 7:897-907.<br />

Cho BP, Song DY, Sugama S, Shin DH, Shimizu Y, Kim SS, Kim YS, Joh TH (2006)<br />

Pathological dynamics <strong>of</strong> activated microglia following medial forebrain bundle<br />

transaction. Glia 53:92-102.<br />

179


Chu CT, Levinthal DJ, Kulich SM, Chalovich EM, DeFranco DB (2004) Oxidative neuronal<br />

injury. <strong>The</strong> dark side <strong>of</strong> ERK1/2. Eur J Biochem 271:2060-2066.<br />

Ciccetti F, Brownell AL, Williams K, Chen YI, Livni E, Isacson O (2002) Neuroinflammation <strong>of</strong><br />

the nigrostriatal pathway during progressive 6-OHDA dopamine degeneration in rats<br />

monitored by immunohistochemistry and PET imaging. Eur J Neurosci 15:991-998.<br />

Cleeter MW, Cooper JM, and Schapira AH (1992) Irreversible inhibition <strong>of</strong> mitochondrial<br />

complex I by 1-methyl-4- phenylpyridinium: evidence for free radical involvement. J<br />

Neurochem 58:786-789.<br />

Cole SH, Carney GE, McClung CA, Willard SS, Taylor BJ, Hirsh J (2005) Two functional but<br />

noncomplementing Drosophila tyrosine decarboxylase genes: distinct roles for neural<br />

tyramine and octopamine in female fertility. J Biol Chem 280:14948-14955.<br />

Cooper RL, Neckameyer WS (1999) Dopaminergic modulation <strong>of</strong> motor neuron activity and<br />

neuromuscular function in Drosophila melanogaster. Comp Biochem Physiol B Biochem<br />

Mol Biol 122:199-210.<br />

Crane BR, Rosenfeld, RJ, Arvai AS, Ghosh DK, Ghosh S, Tainer JA, Stuehr DJ, Getz<strong>of</strong>f ED<br />

(1999) N-terminal domain swapping and metal ion binding in nitric oxide synthase<br />

dimerization. EMBO J 18:6271-6281.<br />

Czlonkowska A, Kohuknika M, Kurkowska-Jatrzebska I, Czlonkowski A (1996) Microglial<br />

reaction in MPTP (1-methyl-4phenyl-1,2,3, 6-tetrahydropyridine) induced Parkinson’s<br />

disease mice model. Neurodegeneration 5:137-143.<br />

Das C, Hoang QQ, Kreinbring CA, Luchansky SJ, Meray RK, Ray SS, Lansbury PT, Ringe D,<br />

Petsko GA (1998) Structural basis for conformational plasticity <strong>of</strong> the Parkinson's disease-<br />

associated ubiquitin hydrolase UCH-L1. Proc Natl Acad Sci USA 103:4675-4680.<br />

180


Dauer W, Przedborski S (2003) Parkinson's disease: mechanisms and models. Neuron 39:889-<br />

909.<br />

Davies S (2000) Nitric oxide signaling in insects. Insect Biochem Mol Biol 30:1123-1138.<br />

Davis RJ (2000) Signal transduction by the JNK group <strong>of</strong> MAP kinases. Cell 103:239-252.<br />

Day BJ, Patel M, Calavetta L, Chang LY, and Stamler JS (1999) A mechanism <strong>of</strong> paraquat<br />

toxicity involving nitric oxide synthase. Proc Natl Acad Sci USA 96:12760-12765.<br />

Dehmer T, Lindenau J, Haid S, Dichgans J, Schulz JB (2000) Deficiency <strong>of</strong> inducible nitric<br />

oxide synthase protects against MPTP toxicity in vivo. J Neurochem 74:2213-2216.<br />

Depino AM, Earl C, Kaczmarczyk E, Ferrari C, Besedovsky H, del Rey A, et al. (2003)<br />

Microglial activation with atypical proinflammatory cytokine expression in a rat model <strong>of</strong><br />

Parkinson's disease. Eur J Neurosci 18:2731-2742.<br />

Di Matteo V, Pierucci M, Di Giovanni G, Di Santo A, Poggi A, Benigno A, Esposito E (2006)<br />

Aspirin protects striatal dopaminergic neurons from neurotoxin-induced degeneration: an in<br />

vivo microdialysis study. Brain Res 1095:167-177.<br />

Díaz-Corrales FJ, Colasante C, Contreras Q, Puig M, Serrano JA, Hernández L, Beaman BL.<br />

(2004) Nocardia otitidiscaviarum (GAM-5) induces parkinsonian-like alterations in mouse.<br />

Braz J Med Biol Res 37:539-548.<br />

Dicker E, Cederbaum AI (1991) NADH-dependent generation <strong>of</strong> reactive oxygen species by<br />

microsomes in the presence <strong>of</strong> iron and redox cycling agents. Biochem Pharmacol 42:529-<br />

535.<br />

DiGregorio PJ, Ubersax JA, O'Farrell PH (2001) Hypoxia and nitric oxide induce a rapid,<br />

reversible cell cycle arrest <strong>of</strong> the Drosophila syncytial divisions. J Biol Chem 276:1930-<br />

1937.<br />

181


Diguet E, Fernagut PO, Wei X, Du Y, Rouland R, Gross C, Bezard E, Tison F (2004)<br />

Deleterious effects <strong>of</strong> minocycline in animal models <strong>of</strong> Parkinson's disease and<br />

Huntington's disease. Eur J Neurosci 19:3266-3276.<br />

Dinis-Oliveira RJ, Remião F, Carmo H, Duarte JA, Navarro AS, Bastos ML, Carvalho F (2006)<br />

Paraquat exposure as an etiological factor <strong>of</strong> Parkinson's disease. Neurotoxicology<br />

27:1110-1122.<br />

Djukic M, Jovanovic MC, Ninkovic M, Vasiljevic I, Jovanovic M (2008) <strong>The</strong> role <strong>of</strong> nitric oxide<br />

in paraquat-induced oxidative stress in rat striatum. Ann Agric Environ Med 14:247-252.<br />

Donin MN, Pagano J, Dutcher JD, McKee CM (1954) Methymycin, a new crystalline antibiotic.<br />

Antobiot Annu 1:179-185.<br />

Dow JA, Maddrell SH, Davies SA, Skaer NJ, Kaiser K (1994) A novel role for the nitric oxide-<br />

cGMP signaling pathway: the control <strong>of</strong> epithelial function in Drosophila. Am J Physiol.<br />

266:R1716-R1719.<br />

Downer RG, Martin RJ (1987) Analysis <strong>of</strong> monoamines and their metabolites by high<br />

performance liquid chromatography with coulometric electrochemical detection. Life Sci<br />

41:833-836.<br />

Du Y, Ma Z, Lin S, Dodel RC, Gao F, Bales KR, Triarhou LC, Chernet E, Perry KW, Nelson<br />

DL, Luecke S, Phebus LA, Bymaster FP, Paul SM (2001) Minocycline prevents<br />

nigrostriatal dopaminergic neurodegeneration in the MPTP model <strong>of</strong> Parkinson's disease.<br />

Proc Natl Acad Sci USA 98:14669-14674.<br />

Dudek H, Datta SR, Franke TF, Birnbaum MJ, Yao R, Cooper GM, Segal RA, Kaplan DR,<br />

Greenberg ME (1997) Regulation <strong>of</strong> neuronal survival by the serine-threonine protein<br />

kinase Akt. Science 275:661-665.<br />

182


Dunnett SB, Björklund A, Stenevi U, Iversen SD (1981) Behavioural recovery following<br />

transplantation <strong>of</strong> substantia nigra in rats subjected to 6-OHDA lesions <strong>of</strong> the nigrostriatal<br />

pathway. I. Unilateral lesions. Brain Res 215:147-61.<br />

Duvoisin RC, Yahr MD (1965). Encephalitis and parkinsonism. Archives <strong>of</strong> Neurology 12:227-<br />

239.<br />

Enikolopov G, Banerji J, Kuzin B (1999) Nitric oxide and Drosophila development. Cell Death<br />

Differ 6:956-963.<br />

Esposito E, Di Matteo V, Benigno A, Pierucci M, Crescimanno G, Di Giovanni G (2007) Non-<br />

steroidal anti-inflammatory drugs in Parkinson's disease. Exp Neurol 205:295-312.<br />

Evans CJ, Hartenstein V, Banerjee U (2003) Thicker than blood: conserved mechanisms in<br />

Drosophila and vertebrate hematopoiesis. Dev Cell 5:673-690.<br />

Fazzini E, Fleming J & Fahn S (1992) Cerebrospinal fluid antibodies to coronavirus in patients<br />

with Parkinson’s disease. Movement Disorders 7:153-158.<br />

Fei Q, McCormack AL, Di Monte DA, Ethell DW (2008) Paraquat neurotoxicity is mediated by<br />

a Bak-dependent mechanism. J Biol Chem 283:3357-3364.<br />

Ferrandon D, Jung AC, Criqui M, Lemaitre B, Uttenweiler-Joseph S, Michaut L, Reichhart J,<br />

H<strong>of</strong>fmann JA (1998) A drosomycin-GFP reporter transgene reveals a local immune<br />

response in Drosophila that is not dependent on the Toll pathway. EMBO J 17:1217-1227.<br />

Fisher AB, Reicherter J (1984) Pentose pathway <strong>of</strong> glucose metabolism in isolated granular<br />

pneumocytes. Metabolic regulation and stimulation by paraquat. Biochem Pharmacol<br />

33:1349-1353.<br />

Floyd RA (1999) Antioxidants, oxidative stress, and degenerative neurological disorders. Proc<br />

Soc Exp Biol Med 222:236-245.<br />

183


Foley E, O'Farrell PH (2003) Nitric oxide contributes to induction <strong>of</strong> innate immune responses to<br />

gram-negative bacteria in Drosophila. Genes Dev 17:115-125.<br />

Foxton RH, Land JM, Heales SJ (2007) Tetrahydrobiopterin availability in Parkinson's and<br />

Alzheimer's disease; potential pathogenic mechanisms. Neurochem Res 32:751-756.<br />

Franke TF, Tart<strong>of</strong> KD, Tsichlis PN (1994) <strong>The</strong> SH2-like Akt homology (AH) domain <strong>of</strong> c-akt is<br />

present in multiple copies in the genome <strong>of</strong> vertebrate and invertebrate eukaryotes. Cloning<br />

and characterization <strong>of</strong> the Drosophila melanogaster c-akt homolog Dakt1. Oncogene<br />

9:141-148.<br />

Freeman MR, Doherty J (2006) Glial cell biology in Drosophila and vertebrates. Trends<br />

Neurosci 29:82-90.<br />

Friggi-Grelin F, Coulom H, Meller M, Gomez D, Hirsh J, Birman S (2003) Targeted gene<br />

expression in Drosophila dopaminergic cells using regulatory sequences from tyrosine<br />

hydroxylase. J Neurobiol 54:618-627.<br />

Ganguly A, Oo TF, Rzhetskaya M, Pratt R, Yarygina O, Momoi T, Kholodilov N, Burke RE<br />

(2004) CEP11004, a novel inhibitor <strong>of</strong> the mixed lineage kinases, suppresses apoptotic<br />

death in dopamine neurons <strong>of</strong> the substantia nigra induced by 6-hydroxydopamine. J<br />

Neurochem 88:469-480.<br />

Gao HM, Hong JS, ZhangW, Liu B (2002) Distinct role for microglia in rotenone-induced<br />

degeneration <strong>of</strong> dopaminergic neurons. J Neurosci 22:782-790.<br />

Gearan T, Castillo OA, Schwarzschild MA (2001) <strong>The</strong> parkinsonian neurotoxin, MPP+ induces<br />

phosphorylated c-Jun in dopaminergric neurons <strong>of</strong> mesencephalic cultures. Parkinsonism<br />

Relat Disord 8:19-22.<br />

184


Gibbs SM (2001) Regulation <strong>of</strong> Drosophila Visual System Development by Nitric Oxide and<br />

cyclic GMP. Amer Zool 41:268-281.<br />

Gibbs SM, Truman JW (1999) Nitric oxide and cyclic GMP regulate retinal patterning in the<br />

optic lobe <strong>of</strong> Drosophila. Neuron 20:83-93.<br />

Giesen K, Hummel T, Stollewerk A, Harrison S, Travers A, Klämbt C (1997) Glial development<br />

in the Drosophila CNS requires concomitant activation <strong>of</strong> glial and repression <strong>of</strong> neuronal<br />

differentiation genes. Development 124:2307-2316.<br />

Glise B, Bourbon H, Noselli S (1995) hemipterous encodes a novel Drosophila MAP kinase<br />

kinase, required for epithelial cell sheet movement. Cell 83:451-461.<br />

Goto A, Kumagai T, Kumagai C, Hirose J, Narita H, Mori H, Kadowaki T, Beck K, Kitagawa Y<br />

(2002) A Drosophila hemocyte-specific protein, hemolectin, similar to human von<br />

Willebrand factor. Biochem J 359:99-108.<br />

Gotoh Y, Nishida E (1995) Activation mechanism and function <strong>of</strong> the MAP kinase cascade. Mol<br />

Reprod Dev 42:486-492.<br />

Green LC, Wagner DA, Glogowski J, Skipper PL, Wishnok JS, Tannenbaum SR (1982)<br />

Analysis <strong>of</strong> nitrate, nitrite, and [15N] nitrate in biological fluids. Anal Biochem 126:131-<br />

138.<br />

Greer CL, Grygoruk A, Patton DE, Ley B, Romero-Calderon R, Chang HY, Houshyar R,<br />

Bainton RJ, Diantonio A, Krantz DE (2005) A splice variant <strong>of</strong> the Drosophila vesicular<br />

monoamine transporter contains a conserved trafficking domain and functions in the<br />

storage <strong>of</strong> dopamine, serotonin, and octopamine. J Neurobiol 64:239-258.<br />

Grima B, Lamouroux A, Blanot F, Biguet NF, Mallet J (1985) Complete coding sequence <strong>of</strong> rat<br />

tyrosine hydroxylase mRNA. Proc Natl Acad Sci USA 82:617-621.<br />

185


Gunnet JW, Lookingland KJ, Moore KE (1986) Effects <strong>of</strong> gonadal steroids on<br />

tuberoinfundibular and tuberohypophysial dopaminergic neuronal activity in male and<br />

female rats. Proc Soc Exp Biol Med 183:48-53.<br />

Halliwell B (2001) Role <strong>of</strong> free radicals in the neurodegenerative diseases: therapeutic<br />

implications for antioxidant treatment. Drugs Aging 18:685-716.<br />

Halter DA, Urban J, Rickert C, Ner SS, Ito K, Travers AA, Technau GM (1995) <strong>The</strong> homeobox<br />

gene repo is required for the differentiation and maintenance <strong>of</strong> glia function in the<br />

embryonic nervous system <strong>of</strong> Drosophila melanogaster. Development 121:317-332.<br />

Harper SJ, LoGrasso P (2001) Signalling for survival and death in neurones: the role <strong>of</strong> stress-<br />

activated kinases, JNK and p38. Cell Signal 13:299-310.<br />

Hartmann A, Troadec JD, Hunot S, Kikly K, Faucheux BA, Mouatt-Prigent A, Ruberg M, Agid<br />

Y, Hirsch EC (2001) Caspase-8 Is an effector in apoptotic death <strong>of</strong> dopaminergic neurons<br />

in Parkinson’s disease, but pathway inhibition results in neuronal necrosis. J Neurosci<br />

21:2247-2255.<br />

Hashimoto M, Takenouchi T, Rockenstein E, Masliah E (2003) Alpha-synuclein up-regulates<br />

expression <strong>of</strong> caveolin-1 and down-regulates extracellular signal-regulated kinase activity<br />

in B103 neuroblastoma cells: role in the pathogenesis <strong>of</strong> Parkinson’s disease. J Neurochem<br />

85:1468-1479.<br />

He J, Magarvey N, Piraee M, Vining LC (2001) <strong>The</strong> gene cluster for chloramphenicol<br />

biosynthesis in Streptomyces venezuelae ISP5230 includes novel shikimate pathway<br />

homologues and a monomodular non-ribosomal peptide synthetase gene. Microbiology<br />

147:2817-2829.<br />

186


Hearn MG, Ren Y, McBride EW, Reveillaud I, Beinborn M, Kopin AS (2002) A Drosophila<br />

dopamine 2-like receptor: Molecular characterization and identification <strong>of</strong> multiple<br />

alternatively spliced variants. Proc Natl Acad Sci USA 99:14554-14559.<br />

Heikkila RE, Hess A, and Duvoisin RC (1984) Dopaminergic neurotoxicity <strong>of</strong> 1-methyl-4-<br />

phenyl-1,2,3,6-tetrahydropyridine in mice. Science 224:1451-1453.<br />

Hersch S, Fink K, Vonsattel JP, Friedlander RM (2003) Minocycline is protective in a mouse<br />

model <strong>of</strong> Huntington's disease. Ann Neurol 54:841.<br />

Hetman M, Gozdz A (2004) Role <strong>of</strong> extracellular signal regulated kinases 1 and 2 in neuronal<br />

survival. Eur J Biochem 271:2050-2055.<br />

Hidalgo, A. and Booth, G.E. (2000) Glia dictate pioneer axon trajectories in the Drosophila<br />

embryonic CNS. Development 127:393-402.<br />

Hirsh EC, Hunot S, Damier P, Faucheux B (1998). Glial cells and inflammation in Parkinson’s<br />

disease: a role in neurodegeneration? Ann Neurol 44:S115–S120<br />

H<strong>of</strong>fmann JA, Reichhart JM (2002) Drosophila innate immunity: an evolutionary perspective.<br />

Nat Immunol 3:121-126.<br />

Hölscher C (1997) Nitric oxide, the enigmatic neuronal messenger: its role in synaptic plasticity.<br />

Trends Neurosci. 20:298-303.<br />

Holz A, Bossinger B, Strasser T, Janning W, Klapper R (2003) <strong>The</strong> two origins <strong>of</strong> hemocytes in<br />

Drosophila. Development 130:4955-4962.<br />

Hornykiewicz O, Kish SJ (1987) Biochemical pathophysiology <strong>of</strong> Parkinson's disease. Adv<br />

Neurol 45:19-34.<br />

Hosoya T, Takizawa K, Nitta K, Hotta Y (1995) glial cells missing: a binary switch between<br />

neuronal and glial determination in Drosophila. Cell 82:1025-1036.<br />

187


Hunot S, Boissiere F, Faucheux B, Brugg B, Mouatt-Prigent A, Agid Y (1996) Nitric oxide<br />

synthase and neuronal vulnerability in Parkinson’s disease. Neuroscience 72:355-363.<br />

Hunot S, Dugas N, Faucheux B, Hartmann A, Tardieu M, Debré P, Agid Y, Dugas B, Hirsch EC<br />

(1999) FcepsilonRII/CD23 is expressed in Parkinson's disease and induces, in vitro,<br />

production <strong>of</strong> nitric oxide and tumor necrosis factor-alpha in glial cells. J Neurosci<br />

19:3440-3447.<br />

Hunot S, Hirsch EC (2003) Neuroinflammatory processes in Parkinson's disease. Ann Neurol. 53<br />

:S49-S58, discussion S58-S60.<br />

Igaki T, Kanda H, Yamamoto-Goto Y, Kanuka H, Kuranaga E, Aigaki T, Miura M (2002) Eiger,<br />

a TNF superfamily ligand that triggers the Drosophila JNK pathway. EMBO J 21:3009-<br />

3018.<br />

Ilett KF, Stripp B, Menard RH, Reid WD, Gillette JR (1974) Studies on the mechanism <strong>of</strong> the<br />

lung toxicity <strong>of</strong> paraquat: comparison <strong>of</strong> tissue distribution and some biochemical<br />

parameters in rats and rabbits. Toxicol Appl Pharmacol 28:216-226.<br />

<strong>Inamdar</strong> A, Lawal H, Ferdousy F, Chaudhuri A, O’ Donnell (<strong>2009</strong>) Drosophila mounts a<br />

microglial-like neuroinflammatory response to paraquat-induced degeneration <strong>of</strong><br />

dopaminergic neurons (under revision for Nature Neuroscience).<br />

Iravani MM, Kashefi K, Mander P, Rose S, Jenner P (2002) Involvement <strong>of</strong> inducible nitric<br />

oxide synthase in inflammation-induced dopaminergic neurodegeneration. Neuroscience<br />

110:49–58.<br />

Ito K, Urban J, Technau, GM (1995) Distribution, classification and development <strong>of</strong> Drosophila<br />

glial cells in the late embryonic and early larval ventral nerve cord. Roux’s Arch Develop<br />

Biol 209:289–307.<br />

188


Iversen SD, Iversen LL (2007) Dopamine: 50 years in perspective. Trends Neurosci 30:188-193.<br />

Javitch JA, D’Amato RJ, Strittmatter SM, and Snyder SH (1985) Parkinsonism-inducing<br />

neurotoxin, N-methyl-4-phenyl- 1,2,3,6-tetrahydropyridine: uptake <strong>of</strong> the metabolite N-<br />

methyl- 4-phenylpyridinium by dopamine neurons explain selective toxicity. Proc Natl<br />

Acad Sci USA 82:2173-2177.<br />

Jellinger KA (2001) <strong>The</strong> pathology <strong>of</strong> Parkinson’s disease. Adv Neurol 86:55-72.<br />

Jian XD, Sui H, Chu ZH, Zhang ZW, Kan BT, Zhang L, Zhang HT (2007) Changes <strong>of</strong> serum<br />

cytokine caused by acute paraquat poisoning. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing<br />

Za Zhi 25:230-232.<br />

Johnson GL, Lapadat R (2002) Mitogen-Activated Protein Kinase Pathways Mediated by ERK,<br />

JNK, and p38 Protein Kinases. Science 298:1911-1912.<br />

Jordan J, Fernandez-Gomez FJ, Ramos M, Ikuta I, Aguirre N, Galindo MF (2007) Minocycline<br />

and cytoprotection: shedding new light on a shadowy controversy. Curr Drug Deliv 4:225-<br />

231.<br />

Jung SH, Evans CJ, Uemura C, Banerjee U (2005) <strong>The</strong> Drosophila lymph gland as a<br />

developmental model <strong>of</strong> hematopoiesis. Development 132:2521-2533.<br />

Kamel F, Tanner C, Umbach D, Hoppin J, Alavanja M, Blair A, Comyns K, Goldman S, Korell<br />

M, Langston J, Ross G, Sandler D. (2007) Pesticide Exposure and Self-reported<br />

Parkinson's Disease in the Agricultural Health Study. Am J Epidemiol 165:364-374.<br />

Kandel ES, Hay N (1999) <strong>The</strong> regulation and activities <strong>of</strong> the multifunctional serine/threonine<br />

kinase Akt/PKB. Exp Cell Res 253:210-229.<br />

Kaur C, Hao AJ, Wu CH, Ling EA (2001) Origin <strong>of</strong> microglia. Microsc Res Tech 54:2-9.<br />

Kennedy AC (1999) Bacterial diversity in agroecosystems. Agric Ecosyst Environ 74:65-76.<br />

189


Kim HS, Suh YH (<strong>2009</strong>) Minocycline and neurodegenerative diseases. Behav Brain Res<br />

196:168-179.<br />

Kim JS, Choi IS, Lee MC (1995). Reversible parkinsonism and dystonia following probable<br />

Mycoplasma pneumoniae infection. Mov Disord 10:510-512.<br />

Kim T, Kim YJ (2005) Overview <strong>of</strong> innate immunity in Drosophila. J Biochem Mol Biol 38:<br />

121-127.<br />

Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M,<br />

Mizuno Y, Shimizu N (1998) Mutations in the parkin gene cause autosomal recessive<br />

juvenile parkinsonism. Nature 392:605-608.<br />

Klaes A, Menne T, Stollewerk A, Scholz H, Klämbt C (1994) <strong>The</strong> Ets transcription factors<br />

encoded by the Drosophila gene pointed direct glial cell differentiation in the embryonic<br />

CNS. Cell 78:149-160.<br />

Klintworth H, Newhouse K, Li T, Choi WS, Faigle R, Xia Z (2007) Activation <strong>of</strong> c-Jun N-<br />

terminal protein kinase is a common mechanism underlying paraquat- and rotenone-<br />

induced dopaminergic cell apoptosis. Toxicol Sci 97:149-162.<br />

Knott C, Stern G, Kingsbury A, Welcher AA, Wilkin GP (2002) Elevated glial brain-derived<br />

neurotrophic factor in Parkinson’s diseased nigra. Parkinsonism Rel Disord 8:329-341.<br />

Knott C, Stern G, Wilkin GP (2000) Inflammatory regulators in Parkinson’s disease: iNOS,<br />

Lipocortin-1, and cyclooxygenase-1 and-2. Mol cell Neurosci 16:724-739.<br />

Kohbata S, Beaman BL (1991) L-DOPA-responsive movement disorder caused by Nocardia<br />

asteroides localized in the brains <strong>of</strong> mice. Infect Immun 59:181-191.<br />

190


Kraus RL, Pasieczny R, Lariosa-Willingham K, Turner MS, Jiang A, Trauger JW (2005)<br />

Antioxidant properties <strong>of</strong> minocycline: neuroprotection in an oxidative stress assay and<br />

direct radical-scavenging activity. J Neurochem 94:819-827.<br />

Krishnakumar S, Burton D, Rasco J, Chen X, O'Donnell J (2000) Functional interactions<br />

between GTP cyclohydrolase I and tyrosine hydroxylase in Drosophila. J Neurogenet 14:1-<br />

23.<br />

Kriz J, Nguyen MD, Julien JP (2002) Minocycline slows disease progression in a mouse model<br />

<strong>of</strong> amyotrophic lateral sclerosis. Neurobiol Dis 10:268-278.<br />

Kuhn DM, Arthur RE Jr, Thomas DM, Elferink LA (1999) Tyrosine hydroxylase is inactivated<br />

by catechol-quinones and converted to a redox-cycling quinoprotein: possible relevance to<br />

Parkinson’s disease. J Neurochem 73:1309-1317.<br />

Kulich SM, Chu CT (2001) Sustained extracellular signal-regulated kinase activation by 6-<br />

hydroxydopamine: implications for Parkinson's disease. J Neurochem 77:1058-1066.<br />

Kumar S, Doumanis J (2000) <strong>The</strong> fly caspases. Cell Death Differ 7:1039-1044.<br />

Kume K, Kume S, Park SK, Hirsh J, Jackson FR (2005) Dopamine is a regulator <strong>of</strong> arousal in<br />

the fruit fly. J Neurosci 25:7377-7384.<br />

Kurucz E, Zettervall CJ, Sinka R, Vilmos P, Pivarcsi A, Ekengren S, Hegedüs Z, Ando I,<br />

Hultmark D (2002) Hemese, a hemocyte-specific transmembrane protein affects the<br />

cellular immune response in Drosophila. Proc Natl Acad Sci USA 100:2622-2627.<br />

Kuter K, Smiałowska M, Wierońska J, Zieba B, Wardas J, Pietraszek M, Nowak P, Biedka I,<br />

Roczniak W, Konieczny J, Wolfarth S, Ossowska K (2007) Toxic influence <strong>of</strong> subchronic<br />

paraquat administration on dopaminergic neurons in rats. Brain Res 1155:196-207.<br />

191


Kuzin B, Regulski M, Stasiv Y, Scheinker V, Tully T, Enikolopov G (2000) Nitric oxide<br />

interacts with the retinoblastoma pathway to control eye development in Drosophila. Curr<br />

Biol 10:459-462.<br />

Kuzin B, Roberts I, Peunova N, Enikolopov G (1996) Nitric oxide regulates cell proliferation<br />

during Drosophila development. Cell 87:639-649.<br />

Kyriakis JM, Avruch J (2001) Mammalian mitogen-activated protein kinase signal transduction<br />

pathways activated by stress and inflammation. Physiol. Rev 81:807-869.<br />

Langston JW, Ballard P, Tetrud JW, Irwin I (1983) Chronic Parkinsonism in humans due to a<br />

product <strong>of</strong> meperidine-analog synthesis. Science 219:979-980.<br />

Langston JW, Forno LS, Tetrud J, Reeves AG, Kaplan JA, Karluk D (1999) Evidence <strong>of</strong> active<br />

nerve cell degeneration in the substantia nigra <strong>of</strong> humans years after 1-methyl-4-phenyl-<br />

1,2,3,6- tetrahydropyridine exposure. Ann Neurol 46:598-605.<br />

Lanot R, Zachary D, Holder F, Meister M (2001) Postembryonic hematopoiesis in Drosophila.<br />

Dev Biol 230:243-257.<br />

Lawson LJ, Perry VH, Dri P, Gordon S (1990) Heterogeneity in the distribution and morphology<br />

<strong>of</strong> microglia in the normal adult mouse brain. Neuroscience 39:151-170.<br />

Le W, Rowe D, Xie W, Ortiz I, He Y, Appel SH (2001) Microglial activation and dopaminergic<br />

cell injury: an in vitro model relevant to Parkinson’s disease. J Neurosci 21:8447–8455.<br />

Learish RD, Bruss MD, Haak-Frendscho M (2000) Inhibition <strong>of</strong> mitogen-activated protein<br />

kinase kinase blocks proliferation <strong>of</strong> neural progenitor cells. Brain Res Dev Brain Res 122:<br />

97-109.<br />

Lei K and Davis RJ (2003) JNK phosphorylation <strong>of</strong> Bim-related members <strong>of</strong> the Bcl2 family<br />

induces Bax-dependent apoptosis. Proc Natl Acad Sci USA 100:2432-2437.<br />

192


Lei K, Nimnual A, Zong WX, Kennedy NJ, Flavell RA, Thompson CB, Bar-Sagi D, Davis RJ<br />

(2002) <strong>The</strong> Bax subfamily <strong>of</strong> Bcl2-related proteins is essential for apoptotic signal<br />

transduction by c-Jun NH(2)-terminal kinase. Mol Cell Biol 22:4929-4942.<br />

Leiserson WM, Harkins EW, Keshishian H (2000) Fray, a Drosophila serine/threonine kinase<br />

homologous to mammalian PASK, is required for axonal ensheathment. Neuron 28:793-<br />

806.<br />

Lemaitre B, H<strong>of</strong>fmann J (2007) <strong>The</strong> host defense <strong>of</strong> Drosophila melanogaster. Annu Rev<br />

Immunol 25:697-743.<br />

Levy OA, Malagelada C, Greene LA (<strong>2009</strong>) Cell death pathways in Parkinson's disease:<br />

proximal triggers, distal effectors, and final steps. Apoptosis 14:478-500.<br />

Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, Wang X (1997)<br />

Cytochrome c and dATP-dependent formation <strong>of</strong> Apaf-1/caspase-9 complex initiates an<br />

apoptotic protease cascade. Cell 91:479-489.<br />

Liao H, Bu WY, Wang TH, Ahmed S, Xiao ZC (2005) Tenascin-R plays a role in<br />

neuroprotection via its distinct domains that coordinate to modulate the microglia function.<br />

J Biol Chem 280:8316-8323.<br />

Liberatore GT, Jackson-Lewis V, Vukosavic S, Mandir AS, Vila M, McAuliffe WG, Dawson<br />

VL, Dawson TM, Przedborski S (1999) Inducible nitric oxide synthase stimulates<br />

dopaminergic neurodegeneration in the MPTP model <strong>of</strong> Parkinson disease. Nat Med<br />

5:1403-1409.<br />

Liew FY, Xu D, Chan WL (1999) Immune effector mechanism in parasitic infections. Immunol<br />

Lett. 65:101-104.<br />

193


Lim YM, Nishizawa K, Nishi Y, Tsuda L, Inoue YH, Nishida Y (1999) Genetic analysis <strong>of</strong><br />

rolled, which encodes a Drosophila mitogen-activated protein kinase. Genetics 153:763-<br />

771.<br />

Lin LF, Doherty DH, Lile JD, Bektesh S, Collins F (1993) GDNF: a glial cell line-derived<br />

neurotrophic factor for midbrain dopaminergic neurons. Science 260:1130-1132.<br />

Lin S, Zhang Y, Dodel R, Farlow MR, Paul SM, Du Y (2001) Minocycline blocks nitric oxide-<br />

induced neurotoxicity by inhibition p38 MAP kinase in rat cerebellar granule neurons.<br />

Neurosci Lett 315:61-64.<br />

Liou HH, Tsai MC, Chen CJ, Jeng JS, Chang YC, Chen SY, Chen RC (1997) Environmental<br />

risk factors and Parkinson’s disease: a case-control study in Taiwan. Neurology 48:1583-<br />

1588.<br />

Liu B (2006) Modulation <strong>of</strong> microglial pro-inflammatory and neurotoxic activity for the<br />

treatment <strong>of</strong> Parkinson's disease. AAPS J 8:E606-E621.<br />

Liu B, Gao HM, Hong JS (2003) Parkinson’s Disease and Exposure to Infectious Agents and<br />

Pesticides and the Occurrence <strong>of</strong> Brain Injuries: Role <strong>of</strong> Neuroinflammation. Environ<br />

Health Perspect 111:1065-1073.<br />

Liu B, Hong JS (2003) Role <strong>of</strong> microglia in inflammation-mediated neurodegenerative diseases:<br />

mechanisms and strategies for therapeutic intervention. J Pharmacol Exp <strong>The</strong>r 304:1-7.<br />

Livingstone MS, Tempel BL (1983) Genetic dissection <strong>of</strong> monoamine neurotransmitter synthesis<br />

in Drosophila. Nature 303:67-70.<br />

Logroscino G (2005) <strong>The</strong> role <strong>of</strong> early life environmental risk factors in Parkinson disease: what<br />

is the evidence? Environ Health Perspect 113:1234-1238.<br />

194


Lubinsky S, Bewley GC (1979) Genetics <strong>of</strong> catalase in Drosophila melanogaster: rates <strong>of</strong><br />

synthesis and degradation <strong>of</strong> the enzyme in flies aneuploid and euploid for the structural<br />

gene. Genetics 91:723-749.<br />

Luckhart S, Rosenberg R (1999) Gene structure and polymorphism <strong>of</strong> an invertebrate nitric<br />

oxide synthase gene. Gene 232:25-34.<br />

Lundell MJ, Hirsh J (1994) Temporal and spatial development <strong>of</strong> serotonin and dopamine<br />

neurons in the Drosophila CNS. Dev Biol 165:385-396.<br />

Macdonald H, Kelly RG, Allen ES, Noble JF, Kanegis LA (1973) Pharmacokinetic studies on<br />

minocycline in man. Clin Pharmacol <strong>The</strong>r 14:852-861.<br />

Mackey WJ, O’Donnell JM (1983) A genetic analysis <strong>of</strong> the pteridine biosynthetic enzyme,<br />

guanosine triphosphate cyclohydrolase, in Drosophila melanogaster. Genetics 105:35–53.<br />

Malagoli D, Sacchi S, Ottaviani E (2008) unpaired (upd)-3 expression and other immune-related<br />

functions are stimulated by interleukin-8 in Drosophila melanogaster SL2 cell line.<br />

Cytokine 44:269-274.<br />

Marletta MA (1989) Nitric oxide: biosynthesis and biological significance. Trends Biochem Sci<br />

14:488-492.<br />

Marshall KA, Reist M, Jenner P, Halliwell B (1999) <strong>The</strong> neuronal toxicity <strong>of</strong> sulfite plus<br />

peroxynitrite is enhanced by glutathione depletion: implications for Parkinson's disease.<br />

Free Radic Biol Med 27:515-520.<br />

Mayer B (1994) Regulation <strong>of</strong> nitric oxide synthase and soluble guanylyl cyclase. Cell Biochem<br />

Funct 12:167-177.<br />

Mayeux R (2003) Epidemiology <strong>of</strong> neurodegeneration. Annu Rev Neurosci 26:81-104.<br />

195


McClung C, Hirsh J (1999) <strong>The</strong> trace amine tyramine is essential for sensitization to cocaine in<br />

Drosophila. Curr Biol 9:853-860.<br />

McCormack AL, Atienza JG, Johnston LC, Andersen JK, Vu S, Di Monte DA (2005) Role <strong>of</strong><br />

oxidative stress in paraquat-induced dopaminergic cell degeneration. J Neurochem<br />

93:1030-1037.<br />

McCormack AL, Thiruchelvam M, Manning-Bog AB, Thiffault C, Langston JW, Cory-Slechta<br />

DA, Di Monte DA (2002) Environmental risk factors and Parkinson’s disease: selective<br />

degeneration <strong>of</strong> nigral dopaminergic neurons caused by the herbicide paraquat. Neurobiol<br />

Dis 10:119-127.<br />

McGeer PL, Itagaki S, Botes BE, McGeer EG (1988) Reactive microglia are positive for HLA-<br />

DR in the substantia nigra <strong>of</strong> Parkinson’s and Alzheimer’s disease brains. Neurology 38:<br />

1285-1291.<br />

McGeer PL, McGeer EG (2004) Inflammation and the degenerative diseases <strong>of</strong> aging. Ann N Y<br />

Acad Sci 1035:104-116.<br />

McGeer PL, McGeer EG (2008) Glial reactions in Parkinson's disease. Mov Disord 23: 474-483.<br />

Mendez JS, Finn BW (1975) Use <strong>of</strong> 6-hydroxydopamine to create lesions in catecholamine<br />

neurons in rats. J Neurosurg 42:166-173.<br />

Miklossy J, Doudet DD, Schwab C, Yu S, McGeer EG, McGeer PL (2006) ICAM-1 in<br />

persisting inflammation in Parkinson disease and MPTP monkeys. Exp Neurol 197:275-<br />

283.<br />

Miller RL, Sun GY, Sun AY (2007) Cytotoxicity <strong>of</strong> paraquat in microglial cells: Involvement <strong>of</strong><br />

PKCdelta- and ERK1/2-dependent NADPH oxidase. Brain Res 1167: 129-139.<br />

196


Mirza B, Hadberg H, Thomsen P, Moos T (2001) <strong>The</strong> absence <strong>of</strong> reactive astrocytosis is<br />

indicative <strong>of</strong> a unique inflammatory process in Parkinson's disease. Neuroscience 95:425-<br />

432.<br />

Mizuno Y, Hattori N, Mochizuki H (2008) Genetic aspects <strong>of</strong> Parkinson's disease. Handb Clin<br />

Neurol 83:217-44.<br />

Mogi M, Harada M, Riederer P, Narabyashi H, Fujita J, Nagatsu T (1994). Interleukin-1 beta<br />

growth factor and transforming growth factor-alpha are elevated in the brain from<br />

Parkinsonian patients. Neurosci Lett 180:147–150.<br />

Moncada S, Bolanos JP (2005) Nitric oxide, cell bioenergetics and neurodegeneration J<br />

Neurochem 97:1676–1689.<br />

Moore KE, Demarest KT, Lookingland KJ (1987) Stress, prolactin and hypothalamic<br />

dopaminergic neurons. Neuropharmacology 26:801-808.<br />

Morgan SC, Taylor DL, Pocock JM (2004) Microglia release activators <strong>of</strong> neuronal proliferation<br />

mediated by activation <strong>of</strong> mitogen-activated protein kinase, phosphatidylinositol-3-<br />

kinase/Akt and delta-Notch signalling cascades. J Neurochem 90:89-101.<br />

Mosley RL, Benner EJ, Kadiu I, Thomas M, Boska MD, Hasan K, Laurie C, Gendelman HE<br />

(2006) Neuroinflammation, Oxidative Stress and the Pathogenesis <strong>of</strong> Parkinson's Disease<br />

Clin Neurosci Res 6: 261-281.<br />

Muller T, Blum-Degen D, Przuntek H, Kuhn W (1998) Interleukin-6 levels in cerebrospinal fluid<br />

inversely correlate to severity <strong>of</strong> Parkinson’s disease. Acta Neurol Scand 98:142-144.<br />

Murad F (1998) Nitric oxide signaling: would you believe that a simple free radical could be a<br />

second messenger, autacoid, paracrine substance, neurotransmitter, and hormone? Recent<br />

Prog Horm Res 53:43-59; discussion 59-60.<br />

197


Nagatsu T, Levitt M, Udenfriend S (1964) Tyrosine hydroxylase. <strong>The</strong> initial step in<br />

norepinephrine biosynthesis, J Biol Chem 239:2910–2917.<br />

Nagatsu T, Sawada M (2006) Cellular and molecular mechanisms <strong>of</strong> Parkinson's disease:<br />

neurotoxins, causative genes, and inflammatory cytokines. Cell Mol Neurobiol 26:781-802.<br />

Nappi AJ, Vass E (1998) Hydrogen peroxide production in immune-reactive Drosophila<br />

melanogaster. J Parasitol 84:1150-1157.<br />

Nappi AJ, Vass E, Frey F, Carton Y (2000) Nitric oxide involvement in Drosophila immunity.<br />

Nitric Oxide 4:423-430.<br />

Neckameyer WS (1996) Multiple roles for dopamine in Drosophila development. Dev Biol<br />

176:209-219.<br />

Neckameyer WS (1998a) Dopamine modulates female sexual receptivity in Drosophila<br />

melanogaster. J Neurogenet 12:101-114.<br />

Neckameyer WS (1998b) Dopamine and mushroom bodies in Drosophila: experience-dependent<br />

and -independent aspects <strong>of</strong> sexual behavior. Learn Mem 5:157-165.<br />

Neckameyer WS, Quinn WG (1989) Isolation and characterization <strong>of</strong> the gene for Drosophila<br />

tyrosine hydroxylase. Neuron 2:1167-1175.<br />

Neckameyer WS, Weinstein JS (2005) Stress affects dopaminergic signaling pathways in<br />

Drosophila melanogaster. Stress 8:117-131.<br />

Neckameyer WS, White K (1993) Drosophila tyrosine hydroxylase is encoded by the pale locus.<br />

J Neurogenet 8:189-199.<br />

Neckameyer WS, Woodrome S, Holt B, Mayer A (2000) Dopamine and senescence in<br />

Drosophila melanogaster. Neurobiol Aging 21:145-152.<br />

198


Nemery B, van Klaveren RJ (1995). NO wonder paraquat is toxic. Hum Exp Toxicol 14:308-<br />

309.<br />

Nicklas WJ, Vyas I, and Heikkila RE (1985) Inhibition <strong>of</strong> NADH-linked oxidation in brain<br />

mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite <strong>of</strong> the neurotoxin, 1-methyl-4-<br />

phenyl-1,2,5,6-tetrahydropyridine. Life Sci 36:2503-2508.<br />

Nimmerjahn A, Kirchh<strong>of</strong>f F, Helmchen F (2005) Resting microglial cells are highly dynamic<br />

surveillants <strong>of</strong> brain parenchyma in vivo. Science. 308:1314-1318.<br />

Niso-Santano M, Morán JM, García-Rubio L, Gómez-Martín A, González-Polo RA, Soler G,<br />

Fuentes JM (2006) Low concentrations <strong>of</strong> paraquat induces early activation <strong>of</strong> extracellular<br />

signal-regulated kinase 1/2, protein kinase B, and c-Jun N-terminal kinase 1/2 pathways:<br />

role <strong>of</strong> c-Jun N-terminal kinase in paraquat-induced cell death. Toxicol Sci 92:507-515.<br />

Noselli S, Agnès F (1999) Roles <strong>of</strong> the JNK signaling pathway in Drosophila morphogenesis.<br />

Curr Opin Genet Dev 9:466-472.<br />

Nüsslein-Volhard C, Wieschaus E, Kluding H (1984) Mutations affecting the pattern <strong>of</strong> the<br />

larval cuticle in Drosophila melanogaster. I. Zygotic loci on the second chromosome.<br />

Onfelt Tingvall T, Roos E, Engström Y (2001) <strong>The</strong> imd gene is required for local Cecropin<br />

expression in Drosophila barrier epithelia. EMBO Rep 2:239-243.<br />

Paisán-Ruíz et al. (2004) Cloning <strong>of</strong> the gene containing mutations that cause PARK8-linked<br />

Parkinson's disease. Neuron 44:595-600.<br />

Palmer RM, Ferrige AG, Moncada S (1987) Nitric oxide release accounts for the biological<br />

activity <strong>of</strong> endothelium-derived relaxing factor. Nature 32:524-526.<br />

Pandey P, Avraham S, Place A, Kumar V, Majumder PK, Cheng K, Nakazawa A, Saxena S and<br />

Kharbanda S (1999) Bcl-xL blocks activation <strong>of</strong> related adhesion focal tyrosine<br />

199


kinase/proline-rich tyrosine kinase 2 and stress activated protein kinase/c-Jun N-terminal<br />

protein kinase in the cellular response to methylmethane sulfonate. J. Biol Chem 274:<br />

8618-8623.<br />

Parker and Auld (2006) Roles <strong>of</strong> glia in the Drosophila nervous system. Semin Cell Dev Biol<br />

17:66-77.<br />

Peng J, Xiao OM, Stevenson FF, Hsu M, Anderson JK (2004). <strong>The</strong> Herbicide Paraquat Induces<br />

Dopaminergic Nigral Apoptosis through Sustained Activation <strong>of</strong> the JNK Pathway. J Bio<br />

Chem 279:32626-32632.<br />

Peng J, Xie L, Stevenson FF, Melov S, Di Monte DA, Andersen JK (2006) Nigrostriatal<br />

dopaminergic neurodegeneration in the weaver mouse is mediated via neuroinflammation<br />

and alleviated by minocycline administration. J Neurosci 26:11644-11651.<br />

Pereanu W, Shy D, Hartenstein V (2005) Morphogenesis and proliferation <strong>of</strong> the larval brain glia<br />

in Drosophila. Dev Biol 283:191-203.<br />

Perier C, Bové J, Wu DC, Dehay B, Choi DK, Jackson-Lewis V, Rathke-Hartlieb S, Bouillet P,<br />

Strasser A, Schulz JB, Przedborski S, Vila M (2007) Two molecular pathwaysinitiate<br />

mitochondria-dependent dopaminergic neurodegeneration in experimental Parkinson’s<br />

disease. Proc Natl Acad Sci USA 104:8161-8166.<br />

Perlman D, O'Brien E (1954) Microbiological production <strong>of</strong> methymycin and related antibiotics.<br />

Antibiot Chemother 4:894-898.<br />

Perrimon N, Lanjuin A, Arnold C, Noll E (1996) Zygotic lethal mutations with maternal effect<br />

phenotypes in Drosophila melanogaster. II. Loci on the second and third chromosomes<br />

identified by P-element-induced mutations. Genetics 144:1681-1692.<br />

200


Pörzgen P, Park SK, Hirsh J, Sonders MS, Amara SG (2001) <strong>The</strong> antidepressant-sensitive<br />

dopamine transporter in Drosophila melanogaster: a primordial carrier for catecholamines.<br />

Mol Pharmacol 59:83-95.<br />

Pou S, Pou WS, Bredt DS, Snyder SH, Rosen GM (1992) Generation <strong>of</strong> superoxide by purified<br />

brain nitric oxide synthase. J Biol Chem 267:24173-24176.<br />

Pradhan S, Pandey N, Shashank S, Gupta RK & Mathur A (1999) Parkinsonism due to<br />

predominant involvement <strong>of</strong> substantia nigra in Japanese encephalitis. Neurology 53:1781-<br />

1786.<br />

Prasad K, Winnik B, Thiruchelvam MJ, Buckley B, Mirochnitchenko O, Richfield EK (2007)<br />

Prolonged toxicokinetics and toxicodynamics <strong>of</strong> paraquat in mouse brain. Environ Health<br />

Perspect 115:1448-1453.<br />

Presta A, Siddhanta U, Wu C, Sennequier N, Huang L, Abu-Soud HM, Erzurum S, Stuehr DJ<br />

(1998) Comparative functioning <strong>of</strong> dihydro- and tetrahydropterins in supporting electron<br />

transfer, catalysis, and subunit dimerization in inducible nitric oxide synthase.<br />

Biochemistry 37:298-310.<br />

Purisai MG, McCormack AL, Cumine S, Li J, Isla MZ, Di Monte DA (2007) Microglial<br />

activation as a priming event leading to paraquat-induced dopaminergic cell degeneration.<br />

Neurobiol Dis 25: 392-400.<br />

Quintero EM, Willis L, Singleton R, Harris N, Huang P, Bhat N, Granholm AC (2006)<br />

Behavioral and morphological effects <strong>of</strong> minocycline in the 6- hydroxydopamine rat model<br />

<strong>of</strong> Parkinson’s disease. Brain Res 1093:198-207.<br />

Regulski M, Tully T (1995) Molecular and biochemical characterization <strong>of</strong> dNOS: a Drosophila<br />

Ca2+/calmodulin-dependent nitric oxide synthase. Proc Natl Acad Sci USA 92:9072-9076.<br />

201


Ries V, Henchcliffe C, Kareva T, Rzhetskaya M, Bland R, During MJ, Kholodilov N, Burke RE<br />

(2006) Oncoprotein Akt/PKB induces trophic effects in murine models <strong>of</strong> Parkinson's<br />

disease. Proc Natl Acad Sci USA 103:18757-18762.<br />

Rio-Hortega Pd (1932) Microglia. In: Cytology and cellular pathology <strong>of</strong> the nervous system (W<br />

Penfield, ed), pp 482-534, New York: Hoeber.<br />

Rizki TM (1962) Experimental analysis <strong>of</strong> hemocyte morphology in insects. Am Zool 2:247-<br />

256.<br />

Rock RB, Gekker G, Hu S, Sheng WS, Cheeran M, Lokensgard JR, Peterson PK (2004) Role <strong>of</strong><br />

microglia in central nervous system infections. Clin Microbiol Rev 17:942-964.<br />

Rodriguez-Blanco J, Martín V, Herrera F, García-Santos G, Antolín I, Rodriguez C (2008)<br />

Intracellular signaling pathways involved in post-mitotic dopaminergic PC12 cell death<br />

induced by 6-hydroxydopamine. J Neurochem 107:127-140.<br />

Röhrdanz E, Schmuck G, Ohler S, Kahl R (2001) <strong>The</strong> influence <strong>of</strong> oxidative stress on catalase<br />

and MnSOD gene transcription in astrocytes. Brain Res 900:128-136.<br />

Rosselli M, Keller PJ, Dubey RK (1998) Role <strong>of</strong> nitric oxide in the biology, physiology and<br />

pathophysiology <strong>of</strong> reproduction. Hum Reprod Update 4:3-24.<br />

Ryu JK, McLarnon JG (2006) Minocycline or iNOS inhibition block 3-nitrotyrosine increases<br />

and blood-brain barrier leakiness in amyloid beta-peptide-injected rat hippocampus. Exp<br />

Neurol 198:552-557.<br />

Sanchez-Pernaute R, Ferree A, Cooper O, Yu M, Brownell AL, Isacson O (2004) Selective<br />

COX-2 inhibition prevents progressive dopamine neuron degeneration in a rat model <strong>of</strong><br />

Parkinson's disease. J Neuroinflammation 1:6.<br />

202


Saner A, Thoenen H (1972) Model experiments on the molecular mechanism <strong>of</strong> action <strong>of</strong> 6-<br />

hydroxydopamine. Mol Pharmacol 7:147-154.<br />

Saporito MS, Thomas BA, Scott RW (2000) MPTP activates c-Jun NH(2)-terminal kinase (JNK)<br />

and its upstream regulatory kinase MKK4 in nigrostriatal neurons in vivo. J Neurochem<br />

75:1200-1208.<br />

Savitt JM, Dawson VL, Dawson TM (2006) Diagnosis and treatment <strong>of</strong> Parkinson disease:<br />

molecules to medicine. J Clin Invest 116:1744-1754.<br />

Scanga SE, Ruel L, Binari RC, Snow B, Stambolic V, Bouchard D, Peters M, Calvieri B, Mak<br />

TW, Woodgett JR, Manoukian AS (2000) <strong>The</strong> conserved PI3'K/PTEN/Akt signaling<br />

pathway regulates both cell size and survival in Drosophila. Oncogene 19:3971-3977.<br />

Schiller F (2000) Fritz Lewy and his bodies. J Hist Neurosci 9:148-151.<br />

Schrag A, Ben-Shlomo Y, Brown R, Marsden CD, Quinn N (1998) Young-onset Parkinson's<br />

disease revisited--clinical features, natural history, and mortality. Mov Disord 13:885-894.<br />

Schrag A, Schott JM (2006) Epidemiological, clinical, and genetic characteristics <strong>of</strong> early-onset<br />

parkinsonism. Lancet Neurol. 5:355-363.<br />

Sekiguchi K, Yokoyama T, Kurabayashi M, Okajima F, Nagai R (1999)<br />

Sphingosylphosphorylcholine induces a hypertrophic growth response through the<br />

mitogen-activated protein kinase signaling cascade in rat neonatal cardiac myocytes. Circ<br />

Res 85:1000-1008.<br />

Semchuk KM, Love EJ, Lee RG (1992) Parkinson's disease and exposure to agricultural work<br />

and pesticide chemicals. Neurology 42:1328-1335.<br />

203


Seo JH, Rah JC, Choi SH, Shin JK, Min K, Kim HS, Park CH, Kim S, Kim EM, Lee SH, Lee S,<br />

Suh SW, Suh YH (2002) Alpha-synuclein regulates neuronal survival via Bcl-2 family<br />

expression and PI3/Akt kinase pathway. FASEB J 16:1826-1828.<br />

Shapiro S (1989) Regulation <strong>of</strong> Secondary Metabolism in Actinomycetes. CRC Press<br />

Sherer TB, Betarbet R, Kim JH, Greenamyre JT (2003) Selective microglial activation in the rat<br />

rotenone model <strong>of</strong> Parkinson's disease. Neurosci Lett 341:87-90.<br />

Simard AR, Soulet D, Gowing G, Julien JP, Rivest S (2006) Bone marrow-derived microglia<br />

play a critical role in restricting senile plaque formation in Alzheimer’s disease. Neuron<br />

49:489-502.<br />

Sortwell CE, Daley BF, Pitzer MR, McGuire SO, Sladek JR Jr, Collier TJ (2000)<br />

Oligodendrocyte-type 2 astrocyte-derived trophic factors increase survival <strong>of</strong> developing<br />

dopamine neurons through the inhibition <strong>of</strong> apoptotic cell death. J Comp Neurol 426:143-<br />

153.<br />

Spillantini MG, Schmidt ML, Lee VM, Trojnowski JQ, Jakes R, Goedert M (1997) Alpha-<br />

synuclein in Lewy bodies. Nature 388:839-840.<br />

Srahna M, Leyssen M, Choi CM, Fradkin LG, Noordermeer JN, Hassan BA (2006) A signaling<br />

network for patterning <strong>of</strong> neuronal connectivity in the Drosophila brain. PLoS Biol 4:e348.<br />

Sriram K, Miller DB, O'Callaghan JP (2006) Minocycline attenuates microglial activation but<br />

fails to mitigate striatal dopaminergic neurotoxicity: role <strong>of</strong> tumor necrosis factor-alpha. J<br />

Neurochem 96:706-718.<br />

Stasiv Y, Regulski M, Kuzin B, Tully T, Enikolopov G (2001) <strong>The</strong> Drosophila nitric-oxide<br />

synthase gene (dNOS) encodes a family <strong>of</strong> proteins that can modulate NOS activity by<br />

acting as dominant negative regulators. J Biol Chem 276:42241-42251.<br />

204


Stathakis DG, Burton DY, McIvor WE, Krishnakumar S, Wright TR, O'Donnell JM (1999) <strong>The</strong><br />

catecholamines up (Catsup) protein <strong>of</strong> Drosophila melanogaster functions as a negative<br />

regulator <strong>of</strong> tyrosine hydroxylase activity. Genetics 153:361-382.<br />

Staveley BE, Ruel L, Jin J, Stambolic V, Mastronardi FG, Heitzler P, Woodgett JR, Manoukian<br />

AS (1998) Genetic analysis <strong>of</strong> protein kinase B (AKT) in Drosophila. Curr Biol 8:599-602.<br />

Steller H (2008) Regulation <strong>of</strong> apoptosis in Drosophila. Genes Dev 22:2256-2266.<br />

Strauss KM, Martins LM, Plun-Favreau H, Marx FP, Kautzmann S, Berg D, Gasser T, Wszolek<br />

Z, Müller T, Bornemann A, Wolburg H, Downward J, Riess O, Schulz JB, Krüger R<br />

(2005) Loss <strong>of</strong> function mutations in the gene encoding Omi/HtrA2 in Parkinson's disease.<br />

Hum Mol Genet 14:2099-2111.<br />

Stronach B, Perrimon N (2002) Activation <strong>of</strong> the JNK pathway during dorsal closure in<br />

Drosophila requires the mixed lineage kinase, slipper. Genes Dev 16:377-387.<br />

Sugama S, Yang L, Cho BP, De Giorgio LA, Lorenzl S, Albers DS, Beal MF, Volpe BT, Joh TH<br />

(2003) Age-related microglial activation in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine<br />

(MPTP)-induced dopaminergic neurodegeneration in C57BL/6 mice. Brain Res 964:288-<br />

294.<br />

Tam S, Barry DP, Beaman L, Beaman BL (2002) Neuroinvasive Nocardia asteroides GUH-2<br />

induces apoptosis in the substantia nigra in vivo and dopaminergic cells in vitro. Exp<br />

Neurol 177:453-460.<br />

Tan PB, Kim SK (1999) Signaling specificity: the RTK/RAS/MAP kinase pathway in<br />

metazoans. Trends Genet 15:145-149.<br />

Tatton WG, Chalmers-Redman R, Brown D, Tatton N (2003) Apoptosis in Parkinson’s disease:<br />

signals for neuronal degradation. Ann Neurol 53:S61–S70. discussion S70-S72.<br />

205


Teismann P, Ferger B (2001) Inhibition <strong>of</strong> the cyclooxygenase isoenzymes COX-1 and COX-2<br />

provide neuroprotection in the MPTP-mouse model <strong>of</strong> Parkinson's disease. Synapse<br />

39:167-174.<br />

Thiruchelvam M, Prokopenko O, Cory-Slechta DA, Buckley B, Mirochnitchenko O (2005)<br />

Overexpression <strong>of</strong> superoxide dismutase or glutathione peroxidase protects against the<br />

paraquat + maneb-induced Parkinson disease phenotype. J Biol Chem 280:22530-22539.<br />

Thöny B, Auerbach G, Blau N (2000) Tetrahydrobiopterin biosynthesis, regeneration and<br />

functions. Biochem J 347:1-16.<br />

Tomita M, Okuyama T, Hidaka K (1999) Changes in mRNAs <strong>of</strong> inducible nitric oxide synthase<br />

and interleukin-1 beta in the liver, kidney and lung tissues <strong>of</strong> rats acutely exposed to<br />

paraquat. Leg Med (Tokyo) 1:127-34.<br />

Tootle TL, Spradling AC (2008) Drosophila Pxt: a cyclooxygenase-like facilitator <strong>of</strong> follicle<br />

maturation. Development 135:839-847.<br />

Tournier C, Hess P, Yang DD, Xu J, Turner TK, Nimnual A, Bar-Sagi D, Jones SN, Flavell RA,<br />

Davis RJ (2000) Requirement <strong>of</strong> JNK for stress-induced activation <strong>of</strong> the cytochrome c-<br />

mediated death pathway. Science 288:870-874.<br />

Truman JW, De Vente J, Ball EE (1996) Nitric oxide-sensitive guanylate cyclase activity is<br />

associated with the maturational phase <strong>of</strong> neuronal development in insects. Development<br />

122:3949-3958.<br />

Tsai CH, Lo SK, See LC, Chen HZ, Chen RS, Weng YH, Chang FC, Lu CS (2002)<br />

Environmental risk factors <strong>of</strong> young onset Parkinson's disease: a case-control study. Clin<br />

Neurol Neurosurg 104:328-33<br />

206


Tyurina YY, Kapralov AA, Jiang J, Borisenko GG, Potapovich AI, Sorokin A, Kochanek PM,<br />

Graham SH, Schor NF, Kagan VE (2006) Oxidation and cytotoxicity <strong>of</strong> 6-OHDA are<br />

mediated by reactive intermediates <strong>of</strong> COX-2 overexpressed in PC12 cells. Brain Res<br />

1093:71-82.<br />

Tzou P, Ohresser S, Ferrandon D, Capovilla M, Reichhart JM, Lemaitre B, H<strong>of</strong>fmann JA, Imler<br />

JL (2000) Tissue-specific inducible expression <strong>of</strong> antimicrobial peptide genes in<br />

Drosophila surface epithelia. Immunity 13:737-748.<br />

Ubogu EE, Cossoy MB, Ransoh<strong>of</strong>f RM (2006) <strong>The</strong> expression and function <strong>of</strong> chemokines<br />

involved in CNS inflammation. Trends Pharmacol Sci 27:48-55.<br />

Uversky VN (2004) Neurotoxicant-induced animal models <strong>of</strong> Parkinson's disease: understanding<br />

the role <strong>of</strong> rotenone, maneb and paraquat in neurodegeneration. Cell Tissue Res 318:225-<br />

241.<br />

Vafeiadou K, Vauzour D, Spencer JP (2007) Neuroinflammation and its modulation by<br />

flavonoids. Endocr Metab Immune Disord Drug Targets 7:211-224.<br />

Valente et al. (2004) Hereditary early-onset Parkinson's disease caused by mutations in PINK1.<br />

Science 304:1158-1160.<br />

Van Den Bosch L, Tilkin P, Lemmens G, Robberecht Wm (2002) Minocycline delays disease<br />

onset and mortality in a transgenic model <strong>of</strong> ALS. Neuroreport 13:1067-1070.<br />

Vaux DL, Korsmeyer SJ (1999) Cell death in development. Cell 96:245-254.<br />

Verdu J, Buratovich MA, Wilder EL, Birnbaum MJ (1999) Cell-autonomous regulation <strong>of</strong> cell<br />

and organ growth in Drosophila by Akt/PKB. Nat Cell Biol 1:500-506.<br />

Vernooy SY, Copeland J, Ghaboosi N, Griffin EE, Yoo SJ, Hay BA (2000) Cell death regulation<br />

in Drosophila: conservation <strong>of</strong> mechanism and unique insights. J Cell Biol 150: F69-76.<br />

207


Vié A, Cigna M, Toci R, Birman S (1999) Differential regulation <strong>of</strong> Drosophila tyrosine<br />

hydroxylase is<strong>of</strong>orms by dopamine binding and cAMP-dependent phosphorylation. J Biol<br />

Chem 274:16788-16795.<br />

Vijitruth R, Liu M, Choi DY, Nguyen XV, Hunter RL, Bing G (2006) Cyclooxygenase-2<br />

mediates microglial activation and secondary dopaminergic cell death in the mouse MPTP<br />

model <strong>of</strong> Parkinson's disease. J Neuroinflammation 3:6.<br />

Viveros OH, Lee CL, Abou-Donia MM, Nixon JC, Nichol CA (1981) Biopterin c<strong>of</strong>actor<br />

biosynthesis: independent regulation <strong>of</strong> GTP cyclohydrolase in adrenal medulla and cortex.<br />

Science 213:349-350.<br />

Vucic D, Kaiser WJ, Harvey AJ, Miller LK (1997) Inhibition <strong>of</strong> reaper-induced apoptosis by<br />

interaction with inhibitor <strong>of</strong> apoptosis proteins (IAPs). Proc Natl Acad Sci USA 94:10183-<br />

10188.<br />

Vucic D, Kaiser WJ, Miller LK (1998) Inhibitor <strong>of</strong> apoptosis proteins physically interact with<br />

and block apoptosis induced by Drosophila proteins hid and grim. Mol Cell Biol 18:3300-<br />

3309.<br />

Waetzig V, Herdegen T (2005) MEKK1 controls neurite regrowth after experimental injury by<br />

balancing ERK1/2 and JNK2 signaling. Mol Cell Neurosci 30:67-78.<br />

Wang MC, Bohmann D, Jasper H (2003b) JNK signaling confers tolerance to oxidative stress<br />

and extends lifespan in Drosophila. Dev Cell 5:811-816.<br />

Wang S, Xu J, Song P, Wu Y, Zhang J, Chul Choi H, Zou MH (2008) Acute inhibition <strong>of</strong><br />

guanosine triphosphate cyclohydrolase 1 uncouples endothelial nitric oxide synthase and<br />

elevates blood pressure. Hypertension 52:484-490.<br />

208


Wang X, Zhu S, Drozda M, Zhang W, Stavrovskaya IG, Cattaneo E, Ferrante RJ, Kristal BS,<br />

Friedlander RM (2003a) Minocycline inhibits caspase-independent and -dependent<br />

mitochondrial cell death pathways in models <strong>of</strong> Huntington's disease. Proc Natl Acad Sci<br />

USA100:10483-10487.<br />

Watanabe Y, Kato H, Araki T (2008) Protective action <strong>of</strong> neuronal nitric oxide synthase inhibitor<br />

in the MPTP mouse model <strong>of</strong> Parkinson's disease. Metab Brain Dis 23:51-69.<br />

Weinberger DR, Berman KF, Illowsky BP (1988) Physiological dysfunction <strong>of</strong> dorsolateral<br />

prefrontal cortex in schizophrenia. III. A new cohort and evidence for a monoaminergic<br />

mechanism. Arch Gen Psychiatry 45:609-615<br />

Werner ER, Gorren AC, Heller R, Werner-Felmayer G, Mayer B (2003) Tetrahydrobiopterin and<br />

nitric oxide: mechanistic and pharmacological aspects. Exp Biol Med (Maywood)<br />

228:1291-302.<br />

White K, Tahaoglu E, Steller H (1996) Cell killing by the Drosophila gene reaper. Science<br />

271:805-807.<br />

Whitton PS (2007) Inflammation as a causative factor in the aetiology <strong>of</strong> Parkinson’s disease. Br<br />

J Pharmacol 150:963-976.<br />

Wildemann B, Bicker G (1999) Nitric oxide and cyclic GMP induce vesicle release at<br />

Drosophila neuromuscular junction. J Neurobiol 39:337-346.<br />

Wilhelm Roux's Arch. Dev Biol 193:267-282.<br />

Wingrove JA, O'Farrell PH (1999) Nitric oxide contributes to behavioral, cellular, and<br />

developmental responses to low oxygen in Drosophila. Cell 98:105-114.<br />

Wright TR (1987) <strong>The</strong> genetics <strong>of</strong> biogenic amine metabolism, sclerotization, and melanization<br />

in Drosophila melanogaster. Adv Genet 24:127-222.<br />

209


Wszolek Z, Monsour H, Smith P, Pfeiffer R (1988) Cryptococcal meningoencephalitis with<br />

parkinsonian features. Mov Disord 3:271-273.<br />

Wu DC, Jackson-Lewis V, Vila M, Tieu K, Teismann P, Vadseth C, Choi DK, Ischiropoulos H,<br />

Przedborski S (2002) Blockade <strong>of</strong> microglial activation is neuroprotective in the 1-methyl-<br />

4-phenyl-1,2,3,6-tetrahydropyridine mouse model <strong>of</strong> Parkinson disease. J Neurosci<br />

22:1763-1771.<br />

Wu XF, Block ML, Zhang W, Qin L, Wilson B, Zhang WQ, Veronesi B, Hong JS (2005) <strong>The</strong><br />

role <strong>of</strong> microglia in paraquat induced dopaminergic neurotoxicity. Antioxid Redox Signal<br />

7: 654-661.<br />

Wyss-Coray T, Mucke L (2002) Inflammation in neurodegenerative disease--a double-edged<br />

sword. Neuron 35:419-432.<br />

Xia XG, Harding T, Weller M, Bieneman A, Uney JB, Schulz JB (2001) Gene transfer <strong>of</strong> the<br />

JNK interacting protein-1 protects dopaminergic neurons in the MPTP model <strong>of</strong><br />

Parkinson's disease. Proc Natl Acad Sci USA 98:10433-10438.<br />

Xiromerisiou G, Hadjigeorgiou GM, Papadimitriou A, Katsarogiannis E, Gourbali V, Singleton<br />

AB (2008) Association between AKT1 gene and Parkinson's disease: a protective<br />

haplotype. Neurosci Lett 436:232-234.<br />

Yamada T, McGeer PL, McGeer EG (1991) Relationship <strong>of</strong> complement- activated<br />

oligodendrocytes to reactive microglia and neuronal pathology in neurodegenerative<br />

disease. Dementia 2:71-77.<br />

Yamada T, McGeer PL, McGeer EG (1992) Lewy bodies in Parkinson’s disease are recognized<br />

by antibodies to complement proteins. Acta Neuropath 84:100-104.<br />

210


Yamamoto K, Ichijo H and Korsmeyer SJ (1999) Bcl-2 is phosphorylated and inactivated by an<br />

ASK1/Jun N-terminal protein kinase pathway normally activated at G(2)/M. Mol Cell Biol<br />

19:8469-8478.<br />

Yang W, Tiffany-Castiglioni E (2005) <strong>The</strong> bipyridyl herbicide paraquat produces oxidative<br />

stress-mediated toxicity in human neuroblastoma SH-SY5Y cells: relevance to the<br />

dopaminergic pathogenesis. J Toxicol Environ Health A 68:1939-1961.<br />

Yang Y, Gehrke S, Haque ME, Imai Y, Kosek J, Yang L, Beal MF, Nishimura I, Wakamatsu K,<br />

Ito S, Takahashi R, Lu B (2005) Inactivation <strong>of</strong> Drosophila DJ-1 leads to impairments <strong>of</strong><br />

oxidative stress response and phosphatidylinositol 3-kinase Akt signaling. Proc Natl Acad<br />

Sci USA 102:13670-13675.<br />

Yasothornsrikul S, Davis WJ, Cramer G, Kimbrell DA, Dearolf CR (1997) viking: identification<br />

and characterization <strong>of</strong> a second type IV collagen in Drosophila. Gene 198:17-25.<br />

Yrjänheikki J, Tikka T, Keinänen R, Goldsteins G, Chan PH, Koistinaho J (1999) A tetracycline<br />

derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia<br />

with a wide therapeutic window. Proc Natl Acad Sci USA 96:13496-13500.<br />

Yrjänheikki J, Tikka T, Keinänen R, Goldsteins G, Chan PH, Koistinaho J (1999) A tetracycline<br />

derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia<br />

with a wide therapeutic window. Proc Natl Acad Sci USA 96: 13496-13500.<br />

Yuan J, Yankner BA (2000) Apoptosis in the nervous system. Nature 407:802-809.<br />

Zettervall CJ, Anderl I, Williams MJ, Palmer R, Kurucz E, Ando I, Hultmark D (2004) A<br />

directed screen for genes involved in Drosophila blood cell activation. Proc Natl Acad Sci<br />

USA 101:14192-14197.<br />

211


Zhang W, Wang T, Pei Z, Miller DS, Wu X, Block ML, Wilson B, Zhang W, Zhou Y, Hong JS,<br />

Zhang J (2005) Aggregated alpha synuclein activates microglia: a process leading to<br />

disease progression in Parkinson's disease. FASEB J 19:533-542.<br />

Zhou C, Huang Y, Przedborski S (2008) Oxidative stress in Parkinson's disease: a mechanism <strong>of</strong><br />

pathogenic and therapeutic significance. Ann N Y Acad Sci 1147:93-104.<br />

Zhu JH, Kulich SM, Oury TD, Chu CT (2002a) Cytoplasmic aggregates <strong>of</strong> phosphorylated<br />

extracellular signal-regulated protein kinases in Lewy body diseases. Am J Pathol 161:<br />

2087-2098.<br />

Zhu S, Stavrovskaya IG, Drozda M, Kim BY, Ona V, Li M, Sarang S, Liu AS, Hartley DM, Wu<br />

DC, Gullans S, Ferrante RJ, Przedborski S, Kristal BS, Friedlander RM. (2002b)<br />

Minocycline inhibits cytochrome c release and delays progression <strong>of</strong> amyotrophic lateral<br />

sclerosis in mice. Nature 417:74-78.<br />

Zou H, Henzel WJ, Liu X, Lutschg A, Wang X (1999) Apaf-1, a human protein homologous to<br />

C. elegans CED-4, participates in cytochrome c-dependent activation <strong>of</strong> caspase-3. Cell 90:<br />

405-413.<br />

212

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!