role of spleen tyrosine kinase in tumor necrosis factor

315
Graduate eses and Dissertations Iowa State University Capstones, eses and Dissertations 2017 Role of spleen tyrosine kinase in tumor necrosis factor like weak inhibitor of apoptosis induced dopaminergic neuronal cell death and microglial activation in invitro and in vivo models of Parkinson disease Sri Harsha Kanuri Iowa State University Follow this and additional works at: hps://lib.dr.iastate.edu/etd Part of the Physiology Commons is Dissertation is brought to you for free and open access by the Iowa State University Capstones, eses and Dissertations at Iowa State University Digital Repository. It has been accepted for inclusion in Graduate eses and Dissertations by an authorized administrator of Iowa State University Digital Repository. For more information, please contact [email protected]. Recommended Citation Kanuri, Sri Harsha, "Role of spleen tyrosine kinase in tumor necrosis factor like weak inhibitor of apoptosis induced dopaminergic neuronal cell death and microglial activation in invitro and in vivo models of Parkinson disease" (2017). Graduate eses and Dissertations. 16389. hps://lib.dr.iastate.edu/etd/16389

Upload: others

Post on 20-Oct-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Graduate Theses and Dissertations Iowa State University Capstones, Theses andDissertations

2017

Role of spleen tyrosine kinase in tumor necrosisfactor like weak inhibitor of apoptosis induceddopaminergic neuronal cell death and microglialactivation in invitro and in vivo models ofParkinson diseaseSri Harsha KanuriIowa State University

Follow this and additional works at: https://lib.dr.iastate.edu/etd

Part of the Physiology Commons

This Dissertation is brought to you for free and open access by the Iowa State University Capstones, Theses and Dissertations at Iowa State UniversityDigital Repository. It has been accepted for inclusion in Graduate Theses and Dissertations by an authorized administrator of Iowa State UniversityDigital Repository. For more information, please contact [email protected].

Recommended CitationKanuri, Sri Harsha, "Role of spleen tyrosine kinase in tumor necrosis factor like weak inhibitor of apoptosis induced dopaminergicneuronal cell death and microglial activation in invitro and in vivo models of Parkinson disease" (2017). Graduate Theses andDissertations. 16389.https://lib.dr.iastate.edu/etd/16389

Role of spleen tyrosine kinase in tumor necrosis factor like weak inhibitor of apoptosis

induced dopaminergic neuronal cell death and microglial activation in invitro and in vivo

models of Parkinson disease

by

Sri Harsha Kanuri

A dissertation submitted to the graduate faculty

in partial fulfillment of the requirements for the degree of

DOCTOR OF PHILOSOPHY

Major: Biomedical Sciences (Physiology)

Program of Study Committee:

Arthi Kanthasamy, Major Professor

Anumantha Kanthasamy

Manju Reddy

Wilson K Rumbeiha

Bryan H Belliare

Jesse Paul Goff

The student author, whose presentation of the scholarship herein was approved by the

program of study committee, is solely responsible for the content of this dissertation.

The Graduate College will ensure this dissertation is globally accessible and will not

permit alterations after a degree is conferred

Iowa State University

Ames, Iowa

2018

Copyright © Sri Harsha Kanuri, 2018. All rights reserved.

ii

TABLE OF CONTENTS

Page

NOMENCLATURE .................................................................................................. iv

ABSTRACT………………………………. .............................................................. Vi

CHAPTER I: LITERATURE REVIEW .................................................................. 1

General Introduction .......................................................................................... 1

Dopamine Neurochemistry ............................................................................... 4

Basal Ganglia Neuroanatomy .......................................................................... 5

Lewy body pathology ....................................................................................... 6

Braak Hypothesis of Parkinson Disease .......................................................... 10

Cell Death in PD .............................................................................................. 12

Genetics and PD ............................................................................................... 17

Pathogenic Mechanisms in PD ......................................................................... 19

Inflammasome ................................................................................................. 31

Microglia and Neuro-inflammation in PD ....................................................... 40

Astrocytes ........................................................................................................ 45

Animal Models of PD ...................................................................................... 46

Spleen Tyrosine Kinase ...................................................................................... 57

Tweak Neuro-inflammation ................................................................................ 64

Bibliography ...................................................................................................... 76

CHAPTER II: INVOLVEMENT OF MITOCHONDRIA MEDIATED

OXIDATIVE STRESS DEPENDENT CELL SIGNALING EVENTS AND SYK

TYROSINE KINASE ACTIVATION IN TUMOR NECROSIS LIKE WEAK

STIMULATOR OF APOPTOSIS (TWEAK) INDUCED DOPAMINERGIC CELL

DEATH

......................................................................................................... 139

Abstract ......................................................................................................... 139

Introduction ......................................................................................................... 140

Methods ......................................................................................................... 145

Results ........................................................................................................ 151

Discussion ......................................................................................................... 162

Bibliography ........................................................................................................ 171

iii

CHAPTER III: ROLE OF SPLEEN TYROSINE KINASE (SYK) IN

MEDIATINGTUMOR NECROSIS FACTOR LIKE WEAK

INHIBITOROFAPOPTOSIS (TWEAK) INDUCED INFLAMMASOME

ACTIVATION IN BV2 MICROGIAL CELLS IN CELL CULTURE MODELS OF

PD

......................................................................................................... 197

Abstract ......................................................................................................... 197

Introduction ......................................................................................................... 198

Methods …..................................................................................................... 202

Results Section ..................................................................................................... 207

Discussion ......................................................................................................... 220

Bibliography ........................................................................................................ 228

CHAPTER IV: SPLEEN TYROSINE KINASE MEDIATES

NLRP3 INFLAMMASOME ACTIVATION IN LPS STIMULATED

MICROGLIA VIA NF-KB ACTIVATION, MAP KINASE ................................... 263

AND ROS GENERATION

Abstract ......................................................................................................... 263

Introduction ......................................................................................................... 264

Methods ......................................................................................................... 267

Results Section ..................................................................................................... 270

Discussion ......................................................................................................... 280

Bibliography ........................................................................................................ 282

CHAPTER V: CONCLUSIONS................................................................................ 291

Conclusions .......................................................................................................... 291

Bibliography ........................................................................................................ 297

CHAPTER VI : FUTURE AREAS OF INVESTIGATION ...................................... 299

Bibliography ........................................................................................................ 303

ACKNOWLEDGMENTS ......................................................................................... 306

iv

NOMENCLATURE

PD Parkinson Disease

AD Alzheimer Disease

TWEAK Tumor Necrosis Factor like Weak stimulator of Apoptosis

TNF, Tumor Necrosis Factor; ALS

Autophagy Lysosomal system

UPS Ubiquitin Proteasome system

SLE Systemic Lupus Erythematosis

BBB Blood brain barrier

MMP Mitochondrial membrane potential

LC3 Light Chain 3

Fn14 Fibroblast Growth Factor Inducible 14

TH Tyrosine Hydroxylase

MCP-1 Monocyte chemoattractant protein

EAE, Experimental Autoimmune Encephalitis

MPTP Methyl Phenyl Tetrahydroperidine

MAPK Mitogen activated Protein Kinase

ERK Extracellular signal regulated kinase

NF-kB Nuclear factor kappa-light-chain-enhancer of activated B cells

FPN Fipronil

GABA Gamma amino butyric acid

PKC-δ Protein Kinase C Delta

ER Endoplasmic Reticulum

MMP Mitochondrial Membrane Potential

SHSY Bone marrow biopsy-derived line SK-N-SH cell line

v

GSK-3B Glycogen Synthase Kinase – 3 Beta

N27 Rat dopaminergic neuronal cell line

BV2 Mouse microglial cell line

ROS Reactive oxygen species

RNS Reactive Nitrite species

GSH Glutathione

DA Dopaminergic neurons

PERK PKR-like ER kinase

CHOP CCAAT-enhancer-binding protein homologous protein

IRE1 Inositol Requiring 1

ATF-4 Activating Transcription Factor 4

Elf-2 Eukaryotic Initiation Factor 2

SYK Spleen Tyrosine Kinase

ZAP-70 Zeta-chain-associated protein kinase 70

BLINK B-cell Linker protein

PAMP Pathogen associated molecular patterns

DAMP Damage associated molecular patterns

LPS Lipo-polysaccaride

HBSS Hank Balanced Salt solution

G3BP1 G3BP Stress Granule Assembly Factor 1

NLRP3 nucleotide-binding domain, leucine-rich-containing family, pyrin domain-3

IL-1 B Interleukin-1 Beta

IL-18 Interleukin 18

R788 Spleen Tyrosine Kinase inhibitor

vi

ABSTRACT

Tweak and TNF family members are novel regulators of acute and chronic

inflammation. Tweak / Fn14 interaction appears to be involved in angiogenesis, inflammation,

proliferation, migration, cytokine production, cytotoxicity and apoptosis. Thus, TWEAK can

induce both cell death and proliferation. Recently, TWEAK has been shown to be associated with

neurodegenerative effects in an MPTP mouse model of PD. Despite numerous studies

demonstrating TWEAK’s ability to cause cell death in diverse cell types the mechanism(s) by

which TWEAK-induce dopaminergic cell death remain poorly defined. Therefore in an attempt to

better understand the molecular basis of TWEAK-induced dopaminergic neurotoxicity we

evaluated the apoptotic effect of TWEAK and associated molecular mechanisms using N27

dopaminergic neuronal cells. TWEAK-induced a time dependent increase in ROS generation,

mitochondrial dysfunction, caspase activation, and NF-kB activation. Additionally, a concurrent

activation of SYK and proteolytic cleavage of PKC delta was evidenced in TWEAK treated cells.

In contrast a marked down regulation of p-GSK 3b (Ser 9) and Akt activity was evidenced in

TWEAK treated cells. Intriguingly, inhibition of SYK activity via R708 attenuated TWEAK-

induced loss of dopaminergic cell viability. Likewise, SN50, NF-kB inhibitor and Quercetin, a

ubiquitous bioactive plant flavonoid attenuated TWEAK-induced apoptotic cell death further

highlighting the pivotal role of NF-kB and mitochondria dependent oxidative stress signaling

events in the mechanism of dopaminergic neurodegeneration. Taken together, our studies

demonstrate the involvement SYK/NF-kB signaling axis and mitochondrial dysfunction in

TWEAK-induced apoptotic cell death. In the next series of studies we determined the effects of

TWEAK on microglia. A growing body of evidence suggests that persistent microglial activation

and accompanying oxidative stress may act as co-conspirators in mediating dopaminergic

vii

neurodegeneration in the substantia nigra in PD pathogenesis. Previously we demonstrated that

PKC delta a redox sensitive kinase is a critical determinant of microglial activation response in

response to diverse inflammogens. More recently, SYK, has been implicated in the activation of

inflammatory cells in response to infection. Therefore, in the current study we hypothesized that

SYK may act as an upstream regulator of NLRP3 inflammasome thereby leading to a heightened

microglial activation response in TWEAK stimulated cells. In the present study we systematically

elucidated the signaling network underlying TWEAK-induced microglial activation response. A

concentration dependent increase in SYK activation and accompanying increase in kinase activity

was evidenced in TWEAK treated cells. Our results with pharmacological inhibitors and siRNA

mediated gene silencing revealed the regulatory role of SYK in ER stress response (ERS), NOX2

upregulation, GSK 3β activation as well as autophagolysosomal system (ALS) and mitochondrial

dysfunction. Taken together our findings demonstrate a role for SYK signaling network in

mediating TWEAK triggered inflammatory response by positively regulating NLRP3

inflammasome activation and ERS in an autophagy dependent manner. We have discovered the

pivotal role of SYK in mediating dopaminergic neurodegeneration as well as heightened

microglial activation response upon TWEAK treatment. By inhibiting SYK activation we can

limit dopaminergic neuronal degeneration as well as microglial activation response in response to

diverse inflammagens including TWEAK. Our studies further highlight the therapeutic advantage

of targeting SYK for the treatment of inflammation related disorders including PD.

1

CHAPTER 1. LITERATURE REVIEW

General Introduction

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

only next to Alzheimer's disease. The economic burden of PD was 14.4 billion dollars in United

States in year 2010, and this burden would increase over coming years as size of the elderly

population also, grows substantially (Kowal et al., 2013). The annual incidence rate of Parkinson

Disease varies from 1.5- 14.8 cases per 100,000 populations worldwide (de Lau & Breteler, 2006;

Elbaz et al., 2016; Kasten et al., 2007; Tanner & Goldman, 1996; Wright Willis et al., 2010).

Caucasians have higher prevalence and incidence of Parkinson’s Disease as compared to African

Americans and Asians (Wright Willis et al., 2010). United States, mid-west and northeastern

regions have a relatively higher incidence and prevalence of Parkinson’s disease as compared to

other regions (Wright Willis et al., 2010). Parkinson’s disease is slightly more common in men

than women (de Lau et al., 2006; Elbaz et al., 2016; Kasten et al., 2007). The incidence and

prevalence of Parkinson’s disease were more common after 50 years of age and increases steadily

until 9th decade (de Lau et al., 2006; Elbaz et al., 2016; Kasten et al., 2007; Tanner et al., 1996).

Approximately, 630,000 people have been diagnosed with PD in 2010 and it is estimated that, by

the year 2050 the disease prevalence would be doubled (Kowal et al., 2013). The prevalence of

Parkinson Disease is 1-2 per 1000 population (Tysnes & Storstein, 2017). The prevalence of

Parkinson Disease increases gradually after 60 years age and becomes highest after 80 years of

age (1663 per 100,000 population) (C. L. Ma et al., 2014) . The mean age of disease onset is 48

years while average disease duration is 21 years (S. Rocha et al., 2014). The annual incidence rate

for males and females with PD includes 207-396 and 127-259 per 100, 000 respectively (C. L.

Ma et al., 2014). The estimated worldwide population with PD by the year 2030 would be around

2

8.67 million (Allyson Jones et al., 2012). The national annual heath care costs per person with PD

for the year 2017 is around $25,000 with additional out of pocket expenses $15,137 per person

(Bohingamu Mudiyanselage et al., 2017). The annual health care health cost per person with

moderate disease is 4 fold very higher than person with mild disease ($63,579 Vs $17,537). The

patients with PD will have severe disability and decreased quality of life attributed to varied

disease symptomatology. These figures highlight the importance of developing appropriate

clinical interventions which arrest the disease progression, reduce disability, improve the quality

of life and alleviate the clinical symptoms in PD patients.

It is hypothesized that, PD pathogenesis originates from gastrointestinal tract and gradually

spreads through brain stem, mid brain and subsequently other regions of the brain according to the

the recently described BRAAK staging (Braak et al., 2003; Robert E. Burke et al., 2008; D. W.

Dickson et al., 2010). The pathophysiological cellular hallmarks of PD includes apoptosis, protein

misfolding, neuroinflammation, mitochondrial dysfunction, calcium dyshomeostasis, lysosomal

dysfunction and proteosomal dysfunction (Antony et al., 2013) among others. A well characterized

histological hallmark of Parkinson’s disease is Lewy bodies, are mainly composed of alpha

synuclein proteins in substantia nigra of midbrain which has been implicated in the prion like

disease progression in the affected brain regions (Baba et al., 1998; DelleDonne et al., 2008; J. L.

Eriksen et al., 2005a; Wakabayashi et al., 2007).

Although mechanisms like oxidative stress, proteosomal dysfunction, mitochondrial

dysfunction and alteration of calcium homeostasis are proposed to be instigating factors (J. L.

Eriksen et al., 2005a). Recent studies implicate a combination of environmental and genetic factors

in the mechanism of dopaminergic neurodegeneration (Riess & Kruger, 1999). Most of the PD

cases are idiopathic but upto 10% of them are caused due to genetic mutations of PD genes like

3

LRRK2, Parkin, PINK, DJ-1 and SNCA (Hardy et al., 2003; Christine Klein & Westenberger,

2012). Genetic PD cases usually have early age of onset with most cases having the disease onset

around 40 years (Hardy et al., 2003; Christine Klein et al., 2012).

The cardinal features of parkinsonism includes resting tremor, rigidity, bradykinesia and

loss of postural reflexes (Jankovic, 2008). Sometimes the diagnosis of PD patients gets delayed

because they do not present with primary motor symptoms always and they present with non-

specific motor symptoms. Few non-specific motor symptoms of PD patients include dysarthria,

dysphagia, sialorrhoea, micrographia, shuffling gait, festination, freezing and dystonia (Jankovic,

2008). Additionally, non-motor symptoms in PD patients includes autonomic dysfunction,

cognitive / neurobehavioral abnormalities, sleep disturbances and sensory abnormalities

(Jankovic, 2008). The most common sleep disturbances in PD patients includes nocturnal sleep

disturbance and excessive day time sleepiness (Davie, 2008). The differential diagnosis of patients

presenting with motor symptoms includes progressive Supra nuclear palsy, Dementia with Lewy

bodies, multiple system atrophy and drug induced Dementia (Frank et al., 2006). Depression and

psychosis can be associated with Parkinson disease (Dooneief et al., 1992; Naimark et al., 1996).

Cognition impairment and depression are very common in PD developing in 40% of the patients

and can develop as late as 10 years after symptoms have emerged (Davie, 2008). The most

commonly used drugs for Parkinson’s disease include levodopa, dopamine agonists

(Bromocriptine, Roprinirole and Pramipexole), MAO inhibitors (Selegiline and Rasagiline),

COMT inhibitors (Entacapone and Tolcapone), neuroleptics and cholinesterase inhibitors (Davie,

2008; Korczyn, 2004). Apart from medications, surgical options are also available for providing

symptomatic relief in PD patients. Bilateral subthalamic stimulation is one of the viable surgical

therapies which provides substantial symptomatic relief from bradykinesia, tremor and rigidity in

4

PD patients (Davie, 2008). Understanding the disease pathogenesis is very important for

developing pharmacological interventions. In this context, animal models of PD have been

beneficial in studying the mechanistic basis of dopaminergic neurodegeneration.

Dopamine Neurochemistry

Parkinson Disease is characterized by dopaminergic cell loss in the substantia nigra and

depletion of striatal dopamine content with associating motor abnormalities. Therefore, it is

important to understand the role of dopamine metabolism and its metabolites. Tyrosine is

converted into L-DOPA by tyrosine hydroxylase by a rate limiting enzyme of dopamine

biosynthesis (Bjorklund & Dunnett, 2007; Johannes Meiser et al., 2013b). Next, L-DOPA is

converted to dopamine by DOPA decarboxylase by removing the carboxyl group from L-DOPA

(Bjorklund et al., 2007; Johannes Meiser et al., 2013b). Additionally, Dopamine is converted to

norepinephrine by dopamine beta hydroxylase. Furthermore, norepinephrine is converted into

epinephrine by phenylethanolamine – S-methyl transferase (Bjorklund et al., 2007; Johannes

Meiser et al., 2013b). Dopamine is released from synaptic vesicles upon stimulation of

dopaminergic neurons which tends to act on the post synaptic receptors or presynaptic receptors

(Werkman et al., 2006; Hui Zhang & Sulzer, 2012). The dopamine which is present in the synaptic

cleft can be taken up by dopaminergic neurons by dopamine transporters (DAT) or taken up by

surrounding glial cells (J. Eriksen et al., 2010). The dopamine that is up taken by the dopaminergic

neurons can present in the cytosol or taken back into synaptic vesicles by vesicular monoamine

transporters (VMAT) (J. Eriksen et al., 2010; Werkman et al., 2006; H. Zhang et al., 2012). The

dopamine which is up taken by glial cells can be metabolized by various enzymes like MAO

(monamine oxidase) or COMT (Catecholamine o-methyl transferase) into various metabolites

(Johannes Meiser et al., 2013a). The main conjugation reactions of dopamine include sulfation and

5

glucuronidation (Eisenhofer et al., 1999; Uutela et al., 2009). The main end products of dopamine

metabolism that are excreted in the urine includes HVA (Homo-vanilic acid), DOPAC (3,4-

dihydroxyphenylacetic acid) and their conjugation metabolites (Swahn & Wiesel, 1976). It is

widely believed that, dopamine metabolites like catecholamine oxidation products can lead to

production of superoxide radicals, deplete glutathione levels and promote the formation of alpha

synuclein formation (Johannes Meiser et al., 2013a; Perez et al., 2002). Neuro-melanin is another

byproduct of metabolism of cytoplasmic dopamine in neurons that can activate microglia mediated

neuro-inflammation and ROS production (Johannes Meiser et al., 2013b; Wilms et al., 2003;

Zecca et al., 2006). These findings indicate the dopamine and its metabolites can accentuate

dopaminergic neurodegeneration in PD.

Basal Ganglia Neuroanatomy

The basal ganglia consist of striatum, pallidum, subthalamic nucleus and substantia nigra

(Groenewegen, 2003). There are two pathways of basal ganglia-thalamocortical circuit.

The direct pathway projects directly from striatum to globus pallidus internus / Substantia nigra

reticulata and it expresses mainly dopamine D1 receptor (Groenewegen, 2003). The indirect

pathway projects from striatum to globus pallidus externus and then to globus pallidus internus /

Substantia nigra reticulata and it expresses mainly dopamine D2 receptor (Groenewegen, 2003).

Generally, the direct pathway is inhibitory whereas the indirect pathway is stimulatory

(Groenewegen, 2003). In fact, direct and indirect pathways act in opposing manner to control the

neurak output from globus pallidus and substantia nigra. The synchronous integration of both the

pathways will help initiation and stoppage of movements (Calabresi et al., 2014). Asynchronous

interaction of direct and indirect pathways such as that occurs in PD results in impairment of start

and stop actions, including bradykinesia, rigidity and gait abnormalities (Calabresi et al., 2014;

6

Dennis W. Dickson, 2012; Ferrer, 2011; G. M. Halliday & McCann, 2010). There are other

functional characteristics of direct and indirect pathways. Mounting evidence suggests that, direct

pathway is associated with reward learning and indirect pathway is involved with aversive

behavior (Hikida et al., 2010). Additionally, activation of direct pathway causes positive

reinforcement whereas activation of indirect pathway leads to punishment (Kravitz et al., 2012).

It is important to know that abnormalities of direct and indirect pathways are seen in other

psyhiatric diseases where dopamine metabolism is altered such as obsessive control disorders and

depression (Wichmann & DeLong, 2011). Dysregulation of direct and indirect pathways due to

dopamine metabolism, can lead to impulse control disorders in PD like pathological gambling,

hypersexuality, shopping and compulsive eating (Voon et al., 2009; G. Zhang et al., 2014). Apart

from PD, other neurodegenerative disorders that are associated with imbalance of direct and

indirect pathways includes Huntington disease (Galvan et al., 2012; Vonsattel et al., 1985). Deep

brain stimulation affords symptomatic relief in PD due to modulation of imbalance of direct /

indirect pathways in PD (DeLong & Wichmann, 2015; Tronnier et al., 2002; Wichmann et al.,

2011). Further studies to characterize the signaling pathways underlying dopamine interactions

with direct and indirect pathways are needed to facilitate the discovery of therapeutic options for

PD.

Lewy body pathology

The pathological hallmark and diagnostic marker of PD includes Lewy bodies which are

intra-neuronal inclusions in the substantia nigra of mid brain (Braak & Braak, 1998; Del Tredici

et al., 2002; Gibb & Lees, 1988; Wakabayashi & Takahashi, 1997). An earlier study, revealed that

Lewy bodies are most prevalent in PD and the brain regions encompass trans-entorhinal, entorhinal

regions, the limbic thalamic nuclei, the anterior cingulated areas, agranular insular cortex, and

7

components of the amygdala (Braak et al., 1998). The other specific brain regions where Lewy

bodies can be located in PD includes hypothalamus, thalamus, cerebral cortex and autonomic

ganglia (Gibb et al., 1988). In PD, the formation of Lewy bodies occurs first in areas like dorsal

vagus-glossophayrngeal complex, locus ceruleus, reticular nucleus, olfactory bulb, olfactory tract

and anterior olfactory nucleus before its spread to the substantia nigra of the mid brain (Del Tredici

et al., 2002). In PD, the systemic sites where Lewy bodies can also be found include sympathetic

nervous system, parasympathetic system, alimentary tract, cardiac plexus, adrenal plexus and

adrenal medulla (Wakabayashi et al., 1997). Lewy body related disorders include Lewy body

dementia, idiopathic Lewy body disease, Parkinson disease, subacute sclerosing pan encephalitis,

ataxia telangiectasia, motor neuron disease and juvenile Parkinson syndrome (Gibb et al., 1988).

The significance of Lewy bodies is difficult to predict as they can be neuroprotective in some

patients whereas in other patients they might represent pre-symptomatic cases that can progress to

full blown PD cases in the near future (Wakabayashi et al., 2007). Pathologically, they are

characterized by intracytoplasmic eosinophilic dark inclusions surrounded by paler halo

structurally composed of abnormally processed neurofilaments (Gibb et al., 1988; Wakabayashi

et al., 2007).

The specific age related prevalence of Lewy body in the general population is as follows

10% in age group 60 -69 years, 12% in age group 70 -79 years, 18% in age group 80 - 89 years

and 16% in age group 90 -99 years (Gibb et al., 1988). The prevalence of finding Lewy bodies

increases gradually from 6th to 9th decade. Mounting evidence suggests that, neural expression

of alpha synuclein leads to Lewy body like inclusions in neocortex, hippocampus and substantia

nigra and is associated with loss of dopamine in basal ganglia and motor abnormalities (Masliah

et al., 2000). It is interesting to know that, mutations of autosomal recessive gene like parkin can

8

cause early onset Parkinson Disease with Lewy bodies (Farrer et al., 2001). The major component

of Lewy body bodies in sporadic PD and Lewy body dementia is mainly alpha Synuclein (Duda

et al., 2000). Accordingly, it is widely believed that, accumulation of Lewy bodies occurs due to

aggregation of poorly degraded alpha-synuclein due to pertubations in autophago-lysosomal

system (ALS), ubiquitin-protesomal system (UPS) and induction of ER stress pathways (Y. Chu

et al., 2009; Mark R. Cookson, 2009; Cuervo et al., 2004; Fujiwara et al., 2002; Smith et al., 2005;

Stefanis et al., 2001).

The ability of alpha-synuclein to form beta pleated structures has been identified as a

critical factor that promotes aggregation and formation of Lewy bodies and Lewy neurites (Vilar

et al., 2008). Phosphorylation, truncation and nitration are the important predisposing factors that

accentuate beta pleated oligomerization and Lewy body formation (Mark R. Cookson, 2009).

Formation of dopamine-alpha-synuclein adducts results in Alpha-Synuclein profibril stabilization,

aggregation and accentuation of dopaminergic neurotoxicity (Conway et al., 2001). Aggregation

of Alpha-Synuclein is the most important factor that leads to formation of inclusion bodies,

accumulation of high molecular weight Alpha-Synuclein and enhanced neurotoxicity (Periquet et

al., 2007). It is interesting to know that serine 129 phosphorylation of Alpha-Synuclein is

neurotoxic and promotes neurodegeneration whereas tyrosine 125 phosphorylation is

neuroprotective and protects against neurodegeneration (L. Chen et al., 2009). Alpha-Synuclein

induced dysfunction of ubiquitin-proteosome system and chaperone mediated autophagy creates a

vicious loop that enhances its own aggregation and subsequent neurodegeneration (Alvarez-Erviti,

Rodriguez-Oroz, Cooper, Caballero, et al., 2010; Snyder et al., 2003). Furthermore, Alpha-

Synuclein causes mitochondrial dysfunction, golgi fragmentation, ER stress and impairs ER-golgi

transport in dopaminergic neuronal cells (Cooper et al., 2006; Devi et al., 2008; Gosavi et al., 2002;

9

Smith et al., 2005). In animal models of PD, it was shown that alpha Synuclein causes activation

of caspase-9 and caspase-3 in substantia nigra of midbrain ultimately leading to neurodegeneration

(L. J. Martin et al., 2006; Yamada et al., 2004). Alpha synuclein also tend to have neuro-

inflammatory effects on the microglia which is associated with the production of pro-inflammatory

mediators, regulation of of NADPH oxidase which ultimately leads to dopaminergic

neurodegeneration (Béraud et al., 2011; Marques & Outeiro, 2012; W. Zhang, Wang, et al., 2005).

Alpha synuclein can be transmitted from cell to cell via prion like mechanism leading to

pathological consequences characterized by cell death and associated neuronal injury (Desplats et

al., 2009; K. Luk et al., 2012; Recasens & Dehay, 2014). In this regard, direct intracerebral

injection of recombinant alpha synuclein into striatum of wild type mice resulted in its gradual

spreading to the neighboring substantia nigra ultimately leading to neuronal loss and behavioral

abnormalities (Desplats et al., 2009; K. Luk et al., 2012; Recasens et al., 2014). Furthermore, alpha

synuclein which is located within the neuronal cells can be transferred to neighboring microglia

and astrocytes and can lead to neuro-inflammation (H. J. Lee et al., 2010; W. Zhang, Wang, et al.,

2005). Alpha synuclein also tends to accumulate in the presynaptic terminals and thus might lead

to defects in neurotransmitter release (Spinelli et al., 2014). Taken together accumulation of alpha

synuclein into proteinaceous deposits known as Lewy body in mid brain region is deleterious and

can lead to progressive neurodegeneration and pathological consequences in PD patients. Studying

the mechanisms that underlie accumulation of these intra cellular inclusions in brain tissues and

developing pharmacological interventions against their formation would improve the survival of

dopaminergic neurons and survival of PD patients.

10

Braak Hypothesis of Parkinson Disease

Alpha Synuclein aggregation and Lewy body inclusions are pathological features of

Parkinson Disease (PD) (Spillantini et al., 1998). Dr Heiko Braak has proposed a hypothesis that

describes mode of spread; with pathology that starts peripherally and sequentially entering central

nervous system (Braak et al., 2003). 51-73% of the PD patients tend to follow the staging system

of Braak regarding initiation, propagation and spread of Lewy body and alpha-synuclein

aggregates into the central nervous system system (Rietdijk et al., 2017). According to Braak

hypothesis, pathological process starts systemically within the gut, spreads through vagus nerve

and olfactory bulb; sequentially entering the substantia nigra (Mid brain) in PD (Braak et al., 2003;

Brooks, 2010; R. E. Burke et al., 2008). In Braak staging, Stages I & II represent early Stage of

disease whereas as stages III, IV, V & VI represent late stages of disease with the presence of

Lewy neuritis and Lewy bodies within the tissues (Braak et al., 2003; Kurt A. Jellinger, 2009). In

Stage I, Lewy body pathology is present within olfactory bulb, vagus nerve and enteric nervous

system (Braak et al., 2003; Brooks, 2010; Kurt A. Jellinger, 2009). Non-motor symptoms like

constipation and loss of smell signifying the involvement of gastrointestinal and olfactory systems

with presence of Lewy bodies is also evidenced (Y. Saito & Murayama, 2012). According to the

dual hit hypothesis, neutrophic pathogen can enter the central nervous system through nasal or

gasterointestinal route thereby leading to pathological changes in the brain (Mulak & Bonaz, 2015;

Visanji et al., 2013). In the nasal route, there is early involvement of anterior olfactory nucleus or

olfactory bulb ultimately spreading to the central nervous system (Chandra et al., 2017; Mulak et

al., 2015; Visanji et al., 2013). In gastric route, the pathogen enters the gastrointestinal system and

localizes in the enteric nervous system (ENS) (Chandra et al., 2017; Mulak et al., 2015; Visanji et

al., 2013). The visceral motor projections of the dorsal motor nucleus of vagus nerve (DMV)

11

synapse with ENS (Chandra et al., 2017; Visanji et al., 2013). So the pathogen spreads retrograde

from ENS through dorsal motor nucleus of vagus nerve into central nervous system (Chandra et

al., 2017; Mulak et al., 2015; Visanji et al., 2013). In central nervous system, the pathology initially

localizes to medulla oblongata (Stage II) and gradually progresses to substantia nigra (Stage III).

(Braak et al., 2003; W. J. Burke, 2003; K. A. Jellinger, 2010). Stages IV, V and VI represent late

Stage of PD with the presence of Lewy body pathology in dorsal motor nucleus of mesocortex /

thalamus, high order neocortex and primary neocortex respectively (Braak et al., 2003; W. J.

Burke, 2003; K. A. Jellinger, 2010). There are few pitfalls that were associated with Braak

hypothesis. It is very highly unlikely that all the patients will have pattern of spread as described

according to Braak staging (Kalaitzakis & Pearce, 2009). It is possible that Lewy body pathology

is absent in dorsal motor nucleus of vagus nerve but still the patient might be presenting with wide

distribution of pathology within the brain (Kalaitzakis et al., 2009). There might be no correlation

between the pathology between the ENS and olfactory bulb which contradicts the dual hit

hypothesis (Lebouvier et al., 2011). Braak hypothesis is based on patterns of neurodegeneration in

different post mortem PD patients at various stages of diagnosis and spread of pathology was not

based in a single case of PD (Koprich et al., 2017). Furthermore, there is no conclusive evidence

that alpha-synuclein pathology undergoes spread during physiological conditions in humans and

is not always associated with onset of symptoms (Walsh & Selkoe, 2016). Additionally, there is

no specific in-vivo marker that can track the progression of disease spread sequentially from

gastrointestinal tract to brain stem (G. Halliday et al., 2012). Only Lewy body pathology is

considered for Braak staging while other earlier events like neuronal loss and microglial activation

were not considered thus creating a selection bias for staging of disease pathology (Milber et al.,

2012; Rietdijk et al., 2017).

12

Cell Death in PD

Broadly, the programmed cell death of dopaminergic neurons can be of three types namely,

apoptotic cell death, autophagic cell death and necrotic cell death (Kostrzewa, 2000; Venderova

& Park, 2012).

Apoptotic Cell Death

Evidence of apoptotic cell death like DNA cleavage, chromatin condensation positive

TUNEL staining, caspase-3 activation caspase-8 activation, TNF-alpha upregulation were

demonstrated in substantia nigra of human PD brains and MPTP treated mice brains (Hartmann,

Troadec, et al., 2001; K. A. Jellinger, 2000; Mochizuki et al., 1996; Mogi et al., 2000; N. A. Tatton

& Kish, 1997; Venderova et al., 2012). Moreover, sometimes apoptotic cell death can be

accompanied with autophagic degeneration in substantia nigra of PD patients (Anglade et al.,

1997; W. G. Tatton et al., 2003).

MPP+ induced apoptotic cell death is the most common form of cell death and is

characterized by positive TUNEL staining, DNA fragmentation, chromatin condensation, caspase

3,8 & 9 activation, endoplasmic reticulum (ER) dysfunction and mitochondrial fragmentation (R.

E. Burke, 2008; Hartmann et al., 2000; Suen et al., 2008; N. A. Tatton, 2000; N. A. Tatton et al.,

1998; Yasuda et al., 2013). Apoptotic cell death is also characteristic of 6-OHDA and alpha-

synulcein induced dopaminergic degeneration in animal models of PD (Saha et al., 2000; Zuch et

al., 2000). In MPTP treated mice, activation of caspase-8, 9& 3 precedes the apoptotic death of

dopaminergic neurons (Mark R. Cookson, 2009; Hartmann, Michel, et al., 2001; Turmel et al.,

2001). In N27 dopaminergic neuronal cells, alpha-synuclein treatment resulted in oxidative stress,

caspase-3 & 9 activation and apoptotic cell death (Chong et al., 2017). In MPTP treated animal

models, excessive BAX (pro-apoptotic protein) activation leads to mitochondrial membrane

13

permeability, excessive ROS production, release of cytochrome C into cytosol leading to apoptotic

cell death of dopaminergic neurons (Hassouna et al., 1996; Venderova et al., 2012; Vila et al.,

2001). MPTP treated mice have decreased expression of Bcl2 (Anti-apoptotic protein) and it’s

over expression affords protection against apoptosis of dopaminergic neurons (Offen et al., 2000;

Vila et al., 2001).

Previous studies support a role for ER stress in PD (Michel et al., 2016). MPP+, rotenone

and 6-OHDA cause ER stress associated apoptosis of dopaminergic neurons (E. J. Ryu et al.,

2002). MPP+ treatment causes ER stress via disruption of calcium metabolism and leads to

apoptotic cell death in human neuroblastoma cell lines (Selvaraj et al., 2012). Disruption of

calcium metabolism leads to apoptosis through pro-apoptotic proteins BAX / BAK and caspase-

12 activation (Nakagawa & Yuan, 2000; R. Sano & J. C. Reed, 2013). Some of the mediators of

ER stress induced apoptosis includes ATF-4, CHOP, GADD34, IRE1 alpha and caspase-12

(Szegezdi et al., 2006). Another study mentions that, ER stress plays a pivotal role in the alpha-

synuclein induced apoptosis and progressive degeneration of dopaminergic neurons in substantia

nigra (Smith et al., 2005). Interestingly, administration of ER stress antagonist protected against

dopaminergic neurodegeneration in alpha-synulcein over expressed mice (Colla et al., 2012b).

Perturbations of the protein degradation machinery namely autophago-lysosomal system

(ALS) and ubiquitin – proteosomal system (UPS) can lead to accumulation of Lewy bodies and

Lewy neurites containing aggregated a-synuclein, a distinctive hallmark of PD which can lead to

oxidative stress and cell death of dopaminergic neurons (Moore et al., 2005; Pan et al., 2008;

Przedborski, 2005; Tan et al., 2009). Formation of inclusion bodies have been believed to have a

deleterious effect on the survival of dopaminergic neurons. It has been shown that, matrix

metalloproteinase-3 (MMP-3) induced cleavage of alpha-synuclein leads to generation of several

14

truncated peptide fragments (1-78, 1-91and 1-93) which accentuates proto-fibril like aggregation

of alpha-synuclein, accumulation of Lewy Bodies and dopaminergic neurotoxicity (D. H. Choi et

al., 2011). It is hypothesized that, alpha synuclein causes downregulation of 14-3-3 chaperones

and upregulation of proapoptotic signals like BAD ultimately leading to apoptotic cell death of

dopaminergic neurons (Yasuda & Mochizuki, 2010).

Necrotic Cell Death

Necrotic cell death is associated with activation of PARP-1 [poly (ADP ribose)

polymersase-1] activation and translocation of AIF (Apoptosis Inducing Factor) from the outer

mitochondrial membrane to the nucleus (Venderova et al., 2012; H. Wang et al., 2003). Migration

of AIF to nucleus is associated with selective degeneration of dopaminergic neurons in substantia

nigra (Burguillos et al., 2011). Nuclear translocation of AIF was previously demonstrated in

ventral mesencephalon of human PD brains (Burguillos et al., 2011; Venderova et al., 2012). Poly

(ADP-ribose) polymerase-1 activation and nuclear AIF translocation were primarily implicated in

the LPS induced dopaminergic cell loss in rat ventral mesencephalon (Burguillos et al., 2011).

PARP activation is responsible for dopaminergic neurodegeneration in MPTP treated rats (Mandir

et al., 1999). Following PARP activation, there is activation of nuclear cell death signaling with

depletion of NAD+ and ATP followed by release of PAR polymers into cytosol and mitochondria

resulting in migration of AIF into the nucleus (S. J. Hong et al., 2004; Y. Wang et al., 2009). PARG

is an important metabolic enzyme that degrades PAR polymers from the cytosol and regulates

PARP induced neuronal cell death (Y. Wang et al., 2009). Previous studies overexpressing PARG

metabolic enzyme resulted in decreased AIF translocation and prevented necrotic cell death (Y.

Wang et al., 2009; Yu et al., 2006). Treatment of MN9D cells (dopaminergic neuronal cell line)

with MPP+ resulted in nuclear translocation of AIF and wide spread DNA fragmentation (C. T.

15

Chu et al., 2005). The predisposing factors that favor nuclear translocation of AIF include

decreased Bcl2 and increased BAX (E. C. Cheung et al., 2005; Cregan et al., 2002).

The mechanistic aspects of programmed cell necrosis includes PARP activation of calpain

followed by BAX upregulation, mitochondrial outer membrane permeabilization (MOMP)

resulting in AIF translocation to the nucleus leading to DNA breaks and chromatin condensation

(S. J. Hong et al., 2004; Moubarak et al., 2007). Previous studies show that, PARP causes

mitochondrial calpain activation through calcium dysregulation and JNK signaling pathway

(Douglas & Baines, 2014; Vosler et al., 2009; Xu et al., 2006). It is important to know that calpain

activation results in mitochondrial release of AIF into nucleus resulting in necrotic cell death

(Polster et al., 2005; Y. Wang et al., 2009). Accumulating evidence suggests that, heat shock

protein (Hsc-70) and Bcl2 are protective factors that delay and impede the migration of AIF into

nucleus preventing necrotic cell death (S. J. Hong et al., 2004; Y. Wang et al., 2009). The proposed

mechanism through which nuclear translocation of AIF induces chromatin condensation and DNA

fragmentation is through recruitment of nucleases and proteases (S. J. Hong et al., 2004). Inhibition

of PARP, calpain, BAX and AIF resulted in neuroprotection against dopaminergic cell loss and

behavioral abnormalities in MPTP mouse models of PD (Burguillos et al., 2011; Crocker et al.,

2003; S. J. Hong et al., 2004; C. Ma et al., 2016; Yokoyama et al., 2010). PARP and AIF are also

implicated in excitotoxic neuronal death induced by excess glutamate levels in ischemia and acute

neurotrauma (E. C. Cheung et al., 2005).

Autophagic Cell Death

Marked reduction of lysosomal markers like LAMP2A and Hsc-70 along with

upregulation of LC3-II levels and accumulation of autophagy vacuoles were demonstrated in

dopaminergic neurons of substantia nigra of PD brains highlighting the involvement of autophagic

16

process in dopaminergic neurodegeneration in PD (Alvarez-Erviti, Rodriguez-Oroz, Cooper,

Caballero, et al., 2010; B. Wang et al., 2016). Autophagic type of cell death in PD is second most

common form of death and is characterized by formation of large cytoplasmic vacuoles and

expression of molecular markers of autophagy (R. A. Nixon & D.-S. Yang, 2012; Venderova et

al., 2012). Mounting evidence suggests that, autophagic cell death is associated with commonly

associated neurotoxins like MPP+, rotenone and paraquat (Dadakhujaev et al., 2010; Niso-Santano

et al., 2011; J. H. Zhu et al., 2007). Alpha-synuclein accumulation or deletion of PD genes like

DJ1, LRRK2, PINK and Parkin can lead to dysfunctional autophagy and dopaminergic cell death

(Gomez-Suaga et al., 2012; McCoy & Cookson, 2011; Scarffe et al., 2014; Winslow &

Rubinsztein, 2011). Alpha-synuclein induced downregulation of chaperone mediated autophagy

leads to excess activation of autophagy resulting in accumulation of autophagosomes and

autophagy-induced cell death (Melinda A. Lynch-Day et al., 2012). Sometimes, autophagy

alterations can also be accompanied with apoptotic cell death in the substantia nigra of the PD

brains (Anglade et al., 1997). Accordingly, in a recent study, MPP+ induced apoptotic cell death

in dopaminergic neuronal cells is associated with activation of autophagy process by decreasing

the interaction of beclin-bcl2 complex (Nopparat et al., 2014). Inhibition of key autophagic

proteins like Atg5, Atg7 & Atg8 has offered neuroprotection against MPP+ induced dopaminergic

neuronal cell death (C. T. Chu et al., 2007). Furthermore, deletion of beclin and Atg16L1 resulted

in decreased accumulation of autophagosomes and offered neuroprotection against protein

aggregate induced neurotoxicity in neurodegenerative disorders like PD (Button et al., 2017).

Alternatively, overexpression of autophagy proteins like LAMP2 resulted in neuroprotection

against alpha-synuclein toxicity (B. Wang et al., 2016; Xilouri et al., 2013).

17

Genetics and PD

PD is a multifactorial disease caused by environmental factors, susceptible genetic factors

and ageing brain (Christine Klein et al., 2012). Although considered rare, these genetic cases

account for few documented cases of reported PD. In sporadic cases of PD, genetic mutations

account for 5-10% of the cases (Pirkevi et al., 2009). In contrast, the incidence of PD in the family

members of documented cases of PD is usually little higher between 10%-30% (von Coelln et al.,

2004). The autosomal dominant genes that was most commonly associated with PD includes alpha

synuclein (SNCA) and Leucine rich repeat kinase (LRRK2) (Christine Klein et al., 2012; Pirkevi

et al., 2009). The autosomal recessive genes that was most commonly associated with PD includes

Parkin, DJ1, PINK1 (PTEN induced kinase-1) and ATPase type 13A (Christine Klein et al., 2012;

Pirkevi et al., 2009). The exact mechanism due to which these genetic mutations lead to occurrence

of PD is not very clear but might be related to induction of specific cell signaling mechanisms.

Parkin

Parkin is cytosolic protein that is mainly located near mitochondrial membrane and it helps

in processing and degrading damaged proteins through ubiquitin proteasome system (von Coelln

et al., 2004). More specifically, it acts as an E3 ubiquitin ligase which attaches ubiquitin to

damaged proteins so that they will be recognized by proteasome complex and then degraded later

(Shimura et al., 2000). Parkin helps in processing alpha synuclein through UPS (ubiquitin-

proteasome system) and prevents the formation of Lewy bodies in PD (Lo Bianco et al., 2004).

Accordingly, parkin associated PD is associated with absence of Lewy bodies (Mori et al., 1998;

Yamamura et al., 1993). Parkin associated PD closely resembles idiopathic Parkinson Disease in

clinical findings although the age of onset relatively earlier with some cases occurring as early as

38-40 years (C. Klein et al., 2000; Lucking et al., 2000). In animal studies, parkin knock out mice

18

show some clinical and behavioral abnormalities which might due to neurodegeneration seen in

PD (Goldberg et al., 2003; Itier et al., 2003).

PINK

PINK is normally a cytosolic protein and gets localized to dysfunctional and depolarized

mitochondria to facilitate their removal through mitophagy (Lazarou et al., 2012). It is important

to know that, PINK migration to depolarized mitochondria leads to activation and recruitment of

parkin to mitochondria for their removal through autophagy (A. W. Greene et al., 2012; Matsuda

et al., 2010; Narendra et al., 2008). Accordingly, loss of PINK physiological function will result

in accumulation of dysfunctional mitochondria thereby leading to cell death. In addition, loss of

PINK1 function leads to abnormalities of mitochondrial morphology and loss of mitochondrial

membrane potential (Exner et al., 2007). Genetic mutations which lead to loss of PINK1 function

leads to clinical phenotype which is very similar to early onset parkin associated PD (Ibanez et al.,

2006). PINK1 knock out studies done in drosophila were associated mitochondrial abnormalities

implying their physiological role in mitochondrial dynamics (Ibanez et al., 2006).

DJ1

Mounting evidence suggests that, in autosomal recessive early onset PD, mutations of DJ1

are least common and are of less clinical significance in neurological practice (Healy et al., 2004).

DJ1 is widely expressed in neurons and glial cells in the brain (Ariga et al., 2013; Bonifati et al.,

2003). It is neuroprotective because, it is involved in physiological functions like mitochondrial

regulation and antioxidative stress reaction (Hao et al., 2010; Hayashi et al., 2009; Junn et al.,

2009; Taira et al., 2004; Yanagida et al., 2009; T. Yokota et al., 2003). Accordingly, genetic

mutations of DJ1 lead to loss of its neuroprotective functions thus making the cells vulnerable to

oxidative stress and cell death (Canet-Avilés et al., 2004). Loss of neuroprotective DJ1 function

19

might render the dopaminergic neurons more vulnerable to environmental toxins like Paraquat and

rotenone in DJ1 drosophila mutants (Meulener et al., 2005). Additionally, DJ1 deficient mice show

increased striatal denervation and dopaminergic neuronal loss emphasizing its neuroprotective

function (R. H. Kim et al., 2005). Finally, another study done on DJ1 null mice showed significant

behavioral abnormalities and alteration of dopaminergic neuronal function (J.-Q. Li et al., 2014).

LRRK2

The physiological functions associated with LRRK2 gene includes neuronal overgrowth,

cytoskeletal maintenance, vesicular trafficking and autophagic protein degradation (Rideout &

Stefanis, 2014). The various mechanisms involved with LRRK2 gene associated PD includes

oxidative stress, mitochondrial dysfunction, synaptic dysfunction, tau phosphorylation and alpha

synuclein accumulation (J.-Q. Li et al., 2014). Most important histopathological features of

LRRK2 gene associated PD includes hyperphosphorylated tau, ubiquitin inclusions and lewy body

neurities (Santpere & Ferrer, 2009). Over expression of LRRK2 gene in drosophila resulted in

behavioral abnormalities and dopaminergic neuronal cell death (Z. Liu et al., 2008). Additionally,

LRRK2 transgenic mice showed substantial behavioral abnormalities with minimal dopaminergic

neurodegeneration (Y. Li et al., 2009).

Pathogenic Mechanisms in PD

Oxidative stress

Parkinson Disease (PD) is a multifactorial disease with wide variety of molecular

mechanisms converging together leading to dopaminergic neurodegeneration. Oxidative stress is

proposed to be one of the most important molecular mechanisms that can potentially attenuate

dopaminergic degeneration in PD. Examination of post mortem brains of PD revealed lipid

peroxidation and GSH (Glutathione) depletion; both signifying the role of oxidative stress in

20

dopaminergic cell death (Hwang, 2013; P. Jenner et al., 1992; Sian et al., 1994). In normal

circumstances, ROS (Reactive Oxygen Species) production within the cell is dissipated by cellular

antioxidant defense mechanisms. Oxidative stress occurs when excess ROS generation is

associated with diminished function of antioxidant enzymes; allowing ROS to accumulate and

ultimately leading to deleterious consequences (Hwang, 2013; John Betteridge, 2000; Sies, 2015).

ROS and RNS can cause DNA fragmentation, lipid peroxidation and protein oxidation ultimately

leading to cell death (Hirsch & Hirsch, 1993; Peter Jenner, 2003).

The external factors that can cause oxidative stress includes parkinsonian neurotoxins like

MPTP, rotenone and paraquat (Arif & Khan, 2010). The internal causative factors that mediate

oxidative stress include alpha-synuclein and genetic PD mutations (PINK1, Parkin, DJ-1&

LRRK2) (Arduíno et al., 2009; Hauser & Hastings, 2013). Notably, the most common molecular

mechanisms that can lead to oxidative stress in PD include, dopamine metabolism, mitochondrial

dysfunction, neuro-inflammation, iron metabolism, calcium metabolism, ubiquitin-proteosome

dysfunction and GSH depletion (Carvalho et al., 2012; Ermak & J.A Davies, 2002; Hwang, 2013;

P. Jenner et al., 1992; J. Meiser et al., 2013)

Dopamine metabolism can yield intermediate products such as DA quinone which can be

detrimental to neuronal survival (Hwang, 2013; C. Zhou et al., 2008). DA quinone can induce

production of superoxide and hydrogen peroxide radicals thereby contributing to oxidative stress.

Mitochondrial dysfunction is the second most important factor contributing to oxidative stress in

PD. Neurotoxins can potentially cause disruption of complex I of ETC (Electron Transport Chain)

thereby leading to ROS accumulation and oxidative stress (Hwang, 2013; Marella et al., 2009;

Perier & Vila, 2012; Winklhofer & Haass, 2010). Mutations of mitochondrial genes (PINK, Parkin

and DJ-1) can cause mitochondrial damage and dysfunctioning of the ETC; both of which can

21

possibly mediate oxidative stress (M. R. Cookson, 2012; Hwang, 2013). Alpha-synuclein,

neuromelanin and MMP-3 are important mediators which are released from degenerating

dopaminergic neurons can stimulate the neighboring microglia to produce ROS and NOS (Nitric

oxide species) ultimately leading to oxidative stress (Hwang, 2013; C. Zhou et al., 2008).

Abnormal iron metabolism is associated with neurodegenerative diseases like PD and there is

excessive iron accumulation in post mortem brain samples of PD patients (Peter Jenner & Olanow,

1998; Perier et al., 2012). The various mechanisms through which iron accumulation can mediate

oxidative stress include dysregulation of mitochondrial function, attenuation of antioxidant

defense mechanism, GSH depletion and lipid peroxidation (Bresgen & Eckl, 2015; Marcio S.

Medeiros et al., 2016; Puntarulo, 2005).

Calcium metabolism is related to mitochondrial function and can be regarded as an

important contributor factor for oxidative stress (Ermak & Davies, 2002; Peng & Jou, 2010). L-

type calcium channels, which are preferentially concentrated in dopaminergic neurons of

substantia nigra can be activated by alpha-synuclein thereby leading to oxidative stress and death

of neurons (Venda et al., 2010).

Pathological aggregation of misfolded and toxic proteins is the one of the important

triggering factor for progression of neurodegenerative diseases like PD (J. Blesa et al., 2015; V.

Dias et al., 2013; Jha et al., 2014). Ubiquitin proteasome system (UPS) mainly functions to remove

unwanted accumulation of oxidized proteins (K.-L. Lim & Tan, 2007). Malfunctioning of

ubiquitin proteasome system can cause accumulation of misfolded, aggregated and oxidized

proteins ultimately leading to excessive ROS production and cell death (Di Domenico et al., 2014).

Recent reports suggest that, UPS dysfunction is followed by mitochondrial dysfunction,

accumulation of ROS and neuronal death (Sunita Maharjan et al., 2014). It is reported that, Lewy

22

body accumulation, impairment of mitochondrial complex I and Parkin gene mutations associated

with PD pathogenesis trigger impairment of proteosome system ultimately resulting in

accumulation of misfolded proteins and oxidative stress (McNaught et al., 2001; Olanow &

McNaught, 2006)

GSH (Glutathione) is generally regarded an antioxidant and mitochondrial protectant in

physiological conditions (Lushchak, 2012). The ratio of reduced glutathione (GSH) to oxidized

glutathione (GSSH) is regarded as indicator of oxidative stress (Zitka et al., 2012). GSH depletion

leads to oxidative stress due to compromise of antioxidant and cellular defense mechanisms and

is commonly associated with Parkinson Disease (Mytilineou et al., 2002). Examination of

postmortem brains revealed depleted GSH levels in substantia nigra of PD patients (Peter Jenner,

2003; Sian et al., 1994). GSH deficiency has been shown be associated with excessive

mitochondrial ROS production, oxidative stress and death of dopaminergic neurons (Peter Jenner

et al., 1998).

Autophagy is a physiological dynamic process that helps in degradation and recycling of

cellular proteins and organelles for maintaining homeostasis within the cell (Hansen & Johansen,

2011; Levine et al., 2011). Autophagy induction is generally used a compensatory defense

mechanism against accumulation of oxidized proteins and dysfunctional mitochondria (Dasuri et

al., 2013). Alpha-synuclein (characteristic feature of PD) has been shown to be associated with

downregulation of autophagy process (Dasuri et al., 2013). Dysregulation of autophagy is usually

associated with the accumulation ROS generated from damaged mitochondria thereby

accentuating oxidative stress and contributing to the demise of dopaminergic neurons in PD

(Dasuri et al., 2013). Caplains are cysteine proteases that are mainly involved in degradation of

cytoskeleton and signaling proteins in the presence of calcium (Dasuri et al., 2013). Caplain

23

activation along with calcium accumulation can lead to impairment of mitochondrial respiratory

function ultimately leading excess ROS production and oxidative stress (Dasuri et al., 2013; V.

Dias et al., 2013).

The ends products or biochemical sequelae of oxidative stress in substantia nigra of PD

patients can include lipid hyper-peroxides, malondialdehyde and 4-hydroxy nonenal (4-HNE)

(Peter Jenner, 2003; P. Jenner et al., 1992; C. Zhou et al., 2008). Various mechanisms by which

these end products potentiate dopaminergic neurodegeneration can include caspase activation,

PARP cleavage, DNA fragmentation, NF-kB inhibition, JNK activation, depletion of

peroxiredoxin-2 (Prx-2), BAX activation and disruption of electron transport chain (Peter Jenner,

2003; C. Zhou et al., 2008)

Therapeutic interventions aimed at oxidative stress might be beneficial in rescuing

dopaminergic neurons from neurodegeneration in PD. Doxycycline has been shown to be effective

in reducing RNS and MMP3 production; thus, offering protection in MPTP model of PD (Hwang,

2013). NQO1 (Quinone reductase) which converts pathogenic quinone into more stable metabolite

has shown therapeutic benefit in MPTP model of PD (Hwang, 2013). Coenzyme Q (CoQ) was

beneficial against oxidative stress in MPTP induced PD model and in double blinded randomized

clinical trials (Henchcliffe & Beal, 2008). The other therapeutic modalities that are effective

against oxidative stress in animal models of PD include dopamine antagonists and creatinine

through improvement of mitochondrial function and restoration of GSH levels (V. Dias et al.,

2013; Henchcliffe et al., 2008).

24

ER stress

The Endoplasmic reticulum (ER) is the largest organelle composed of nuclear envelope

and peripheral ER which are connected with each other through cisternae (Schwarz & Blower,

2016). The major significant functions of ER include protein synthesis, protein folding, post

translational modification, include lipid metabolism, calcium storage and carbohydrate

metabolism (Breckenridge et al., 2003; Lindholm et al., 2006; Paschen & Frandsen, 2001; Rao et

al., 2004; Schwarz et al., 2016). Rough ER (RER) with high concentration of ribosomes works in

conjunction with Golgi apparatus in synthesis of proteins and shuttling proteins to their appropriate

destinations (Chaudhari et al., 2014). Ribosome free smooth ER (RER) ribosomes is mainly

involved in carbohydrate metabolism, lipid metabolism and drug metabolism (Bravo et al., 2013).

Sarcoplasmic reticulum is specialized type of smooth ER that is specifically present in the muscle

cells that is associated with excitation contraction coupling and muscle contraction (Rossi & T.

Dirksen, 2006).

Aberrant ER functions can result in accumulation of misfolded proteins and mounting an

unfolded protein response (UPR) in the ER ultimately leading to pathological consequences

(Breckenridge et al., 2003; Paschen et al., 2001; Rao et al., 2004). Unfolded protein response

(UPR) in the ER includes simultaneous activation of three proteins in the ER membrane namely,

IRE1, PERK and ATF-6 in a three-pronged approach (Cullinan & Diehl, 2004; Oyadomari &

Mori, 2004; Ron & Hubbard, 2008; R. Sano et al., 2013; Yamaguchi & Wang, 2004). Primarily,

IRE1 mediated XBP1 mRNA splicing leads to stimulation JNK pathway and activation of

autophagy (Urano et al., 2000). Secondarily, PERK mediated Elf-2 alpha phosphorylation leads to

increased transcription of antioxidant enzymes for protection against oxidative stress (Cullinan et

al., 2004; DuRose et al., 2006). Lastly, ATF-6 activation by proteolytic activation in Golgi

25

apparatus leads to increased production of ER chaperones which prevent misfolding of proteins in

ER (Renata Sano & John C. Reed, 2013; L. Wu et al., 2013). Short term UPR response helps to

restore normal physiological function of ER whereas long term and sustained UPR response leads

deleterious consequences in the cell (Lindholm et al., 2006). It is interesting to note that, ER stress

stimulates autophagy to promote the clearance of misfolded proteins as a compensatory

mechanism thereby preventing their aggregation and further deleterious consequences (Ogata et

al., 2006). It is hypothesized that, excessive and prolonged UPR response leads to increased

production of ROS, mitochondrial depolarization and subsequent release of pro-apoptotic factors

into cytosol causing deleterious consequences namely cell death (Cao & Kaufman, 2014).

ER stress is implicated in the pathogenesis of neurodegenerative diseases like Parkinson

Disease, Alzheimer Disease and Prion Diseases (Mercado et al., 2015). Accumulating evidence

suggests that, pathological sequelae of ER stress were commonly witnessed in postmortem tissue

analysis of sporadic cases and animal models of PD (Mercado et al., 2015). Activation of UPR

(unfolded response) pathway was shown to be associated with human and cellular models of PD.

Studies had shown that there is activation of PERK-Elf-2 alpha pathway in the substantia nigra of

human PD brains (Hoozemans et al., 2007). Activation of PERK-Elf-2 pathway leads to CHOP

activation, ATF-4 upregulation and apoptosis of dopaminergic neurons in PD (Hoozemans et al.,

2007). Accumulation of ER stress markers including CHOP, IRE and PERK were observed in the

neuronal cultures treated with neurotoxins like MPP+, 6-OHDA and Rotenone (Holtz & O'Malley,

2003; Elizabeth J. Ryu et al., 2002). MPP+ induced dopaminergic neurotoxicity involves

transcriptional activation of ATF-6 and upregulation of ER chaperones via MAPK P38

phosphorylation (Egawa et al., 2010). It is interesting to note that, apoptosis in tunicamycin

induced ER stress in PC12 cells is regulated by combination of signaling molecules including

26

CHOP, AKT, P38 MAPK, TRB3 (Novel target gene of CHOP), FOXO 3a (Important transcription

factor) and PUMA (BH3 only subgroup of Bcl2 family) (C. G. Zou et al., 2009). It should be noted

that, ER stress and disturbance of calcium homeostasis are involved in pathogenesis of Parkinson

Disease (Arduíno et al., 2009). Studies show interaction of genes and ER stress in pathogenesis of

Parkinson Disease. It is proposed that, downregulation of DJ-1 leads to chronic ER stress,

oxidative stress and UPS dysfunction there leading to early onset juvenile Parkinson Disease

(Takanori Yokota et al., 2004). Decreased expression of Parkin leads to accumulation of Pear R

receptor in ER ultimately leading to sustained ER stress and dopaminergic cell death in autosomal

recessive early onset PD (Imai et al., 2001).

Previous reports indicate that, Alpha-Synuclein accumulation leads to the impairment in

processing of ATF-6 in COP-II vesicles through ER-golgi transit ultimately resulting in decreased

associated degradation and pro-apoptotic signaling (Credle, Forcelli, et al., 2015). Aggregation of

Alpha-Synuclein in ER results in sustained and prolonged ER stress induced UPR response thereby

leading to dopaminergic neurodegeneration and neuronal cell death in Alpha- Synuclein induced

PD cases (Colla et al., 2012a; Hugo J. R. Fernandes et al.; Jiang et al., 2010). Furthermore, ER

stress and autophagy dysfunction are together responsible for Alpha-Synuclein accumulation in

independent pleuri-potent stem cell derived dopaminergic neurons from PD patients heterozygous

for GBA-N370S mutation (Hugo J. Fernandes et al., 2016). Additionally, previously published

reports indicate that ER stress and mitochondrial dysfunction can potentiate accumulation of

Alpha-Synuclein in PC12 cell lines (W Smith et al., 2006). ER stress along with impaired clearance

of toxic protein aggregates by ubiquitin-proteosome system contributes to dopaminergic

neurodegeneration (Calì et al., 2011). In paraquat induced PD, there is marked upregulation of

CHOP and GRP-78 along with stimulation of IRE1/ASK/JNK pathway thereby leading to

27

neuronal cell death (W. Yang et al., 2009). Another report reveals that, altered calcium signaling

and inhibition of AKT / mTOR signaling leads to elevated ER stress in neurotoxin induced PD

cases (S et al., 2012). In the rat model of PD, overexpression of ATF-4 resulted in severe

dopaminergic cell loss and neurodegeneration via apoptotic cell death (Gully et al., 2016). Reports

suggest that, over expression of XBP-1 (X-box binding protein) results in dopaminergic neuronal

survival whereas deficiency of XBP-1 leads to sustained ER stress and dopaminergic

neurodegeneration (Valdés et al., 2014). Additionally, over expression of ER chaperone GRP-78

resulted in diminished Alpha-Synuclein toxicity, decreased apoptosis and increased survival of

dopaminergic neurons in rat model of PD (Gorbatyuk et al., 2012).

In 6-OHDA induced PD, basic fibroblast growth factor offered neuroprotection against ER

stress through activation of AKT and ERK ½ pathways (P. Cai et al., 2016). ER stress protective

agents like salubrinol were shown to provide neuroprotection in Alpha-synuclein over expressing

experimental models of PD (Colla et al., 2012a). Methoxyflavones are anti-inflammatory agents

which are present in citrus peel were shown offer protection against ER stress in MPTP induced

mice (Takano et al., 2007). Lastly, Insulin like growth factor also offered protection ER stress

against tunicamycin induced ER stress in PC12 cells through activation of AKT and P38 pathways

(C. G. Zou et al., 2009). Zonasimide (Antiepileptic drug) was proven to afford therapeutic benefit

in human PD by protection against ER stress (Tsujii et al., 2015). In rat model of PD, Telmisartan

and Candesartan celexitil offered protective benefits by protecting dopaminergic neurons against

ER stress (Tong et al., 2016; J. Wu et al., 2007). Taken together, it is likely that neuroprotective

strategies against ER stress would be beneficial in preventing dopaminergic neurodegeneration in

PD.

28

Autophagy

The hallmark of Parkinson Disease is the accumulation of protein aggregates in

dopaminergic neuronal cells which promote cell death through release of toxic intermediates.

Autophagy is a protective mechanism that helps in the clearance of the protein aggregates and

prevents their accumulation which can lead to detrimental outcomes (Eskelinen & Saftig, 2009;

Hansen et al., 2011; Nedelsky et al., 2008). They are three types of autophagy namely

Microautophagy, Macroautophagy and Chaperone mediated Autophagy (Boya et al., 2013).

Macroautophagy involves formation of isolation membrane which engulfs cytoplasmic material

which proceeds through autophagosome, auto-phagolysososome and degradation (A Lynch-Day

et al., 2012). Chaperone mediated autophagy involves receptor mediated uptake of cytoplasmic

contents by lysosome and their subsequent degradation (Kaushik et al., 2011). Microautophagy

involves nonreceptor mediated uptake of cytoplasmic aggregates into lysosomal lumen (W. W. Li

et al., 2012). Autophagic reflux describes the whole process of autophagy which begins with

formation of phagopore in cytoplasm and proceeds through the following stages; Autophagosome

and Autolysosomesome and finally ending with complete degradation of cargo material (Hansen

et al., 2011; Loos et al., 2014; X. J. Zhang et al., 2013).

The mechanism of autophagy is a complex process which includes three steps namely

autophagosome biogenesis, nucleation step and elongation step (Eskelinen et al., 2009; J. Kaur &

Debnath, 2015; Levine et al., 2011). In autophagosome biogenesis step, the ULK complex helps

to initiate the formation of autophagosome (J. Kaur et al., 2015). In the nucleation step, ULK1

complex migrates to the ER membrane and associates with PI-3K complex (Phosphotidylinositol-

3OH Kinase) which eventually get phosphorylated (PtdIns3-P) (Eskelinen et al., 2009; Levine et

al., 2011). Next, in the elongation step, ATG5-12 and LC3 present on the isolation membrane will

29

help in elongation and maturation of autophagosome (Eskelinen et al., 2009; Levine et al., 2011).

Finally, autophagosome gradually fuses with lysosome resulting in formation of auto-

phagolysosome thereby resulting in degradation and clearance of cargo (Eskelinen et al., 2009;

Hansen et al., 2011; J. Kaur et al., 2015; Levine et al., 2011; Loos et al., 2014; X. J. Zhang et al.,

2013). The most important regulatory factors for autophagy includes availability of aminoacids

and cytosolic calcium through TOR protein kinases (Eskelinen et al., 2009). The major

physiological functions of autophagy includes stress response, housekeeping function, innate

immunity, cell death and ageing (Eskelinen et al., 2009). Dysfunction of autophagy is associated

with accumulation of protein aggregation as seen in neuro-degenerative diseases like Parkinson

Disease, Alzheimer Disease and Prion diseases (Eskelinen et al., 2009; Hansen et al., 2011; Loos

et al., 2014; Nah et al., 2015; X. J. Zhang et al., 2013). Neurodegenerative diseases like Huntington

Disease and Alzheimer disease is associated with increased accumulation of autophagic vacuoles

implying activation of autophagic process (Nixon, 2013). It is interesting to note that, short lived

proteins are usually degraded by Ubiquitin Proteasome System (UPS) whereas long lived proteins

are degraded by autophagy (Fuertes et al., 2003; Kraft et al., 2010; D. Li, 2006; Lilienbaum, 2013).

It is believed that, autophagy process becomes stimulated when UPS is overwhelmed and there is

accumulation of misfolded proteins in the cytoplasm (Iwata et al., 2005; Lilienbaum, 2013; Taylor

et al., 2003; Yamamoto et al., 2006). Additionally, it is hypothesized that, impairment of UPS

leads to stimulation of autophagic process to clear the excess aggregation of protein aggregates in

the cytoplasm (Lilienbaum, 2013; Rideout et al., 2014; Yamamoto et al., 2006).

P62 is an important cytosolic protein that is frequently associated with ubiquitin positive

aggregates so that they will be degraded by autophagic process (Bjorkoy et al., 2009; Pankiv et al.,

2007). During oxidative stress, parkin induced ubiquitination of damaged mitochondria causes

30

migration of P62 which ultimately leads to their degradation through autophagic process (M. A.

Lynch-Day et al., 2012; Narendra et al., 2010). It has been reported that, P62 plays an important

role in autophagic degradation of ubiquition tagged cytoplasmic inclusions and Lewy bodies in

Parkinson Disease (Kuusisto et al., 2003; Watanabe et al., 2013; Zatloukal et al., 2002). Recently,

it was reported that, parkin degrades P62 associated with ubiquitinated positive inclusions and

deficiency of parkin leads to accumulation of P62 in dopaminergic neuronal cells and increases

their vulnerability to dopaminergic neurodegeneration (P. Song et al., 2016). Another study

mentions that, depletion of P62 resulted in decreased mitophagy and increased vulnerability to cell

death (Huang et al., 2011).

In paraquat induced PD models, production of ROS and loss of mitochondrial membrane

potential leads to parkin relocation to depolarized mitochondria and subsequent removal

autophagic removal of damaged mitochondria (Mitophagy) (A Lynch-Day et al., 2012). Previous

reports indicate that, autophagic rflux helps in removal of misfolded proteins and relieving ER

stress in neurodegenerative disorders like PD (Y. Cai et al., 2016). Autophagy cannot be

considered always neuroprotective. Chronic and excessive activation of autophagic process could

be detrimental and can be associated with neuronal loss in PD (Bredesen et al., 2006; Chung et al.,

2009).

Autophagy is usually protective mechanism that promotes cell survival through clearance

of aggregate proteins. But excessive activation of autophagy can be detrimental by

causing autophagic cell death (Kroemer & Levine, 2008). Excessive activation of autophagy can

engulf massive amount of cytosol and organelles leading cellular demise (Maiuri et al., 2007). In

other circumstances, excessive autophagy activation can degrade key antioxidant enzymes like

catalase which ultimately results in cell death (Maiuri et al., 2007). Previous reports have shown

31

that, over expression of ATG can lead to excessive activation of autophagy and cell death (R. C.

Scott et al., 2007). Autophagic cell death is defined as cell death accompanied by excessive

accumulation of autophagic vacuoles within the cell in the absence of chromatin condensation

(Kroemer et al., 2008). Reports indicate that, there is excess accumulation of autophagosomes with

co-localization of of LC3-II with Lewy bodies and alpha-synuclein inclusions in brain of PD

patients as compared to normal population (Alvarez-Erviti, Rodriguez-Oroz, Cooper, & et al.,

2010; Z. H. Cheung & Ip, 2009). Anticancer treatments often cause excessive activation of

autophagy ultimately leading to autophagic cell death of tumor cells (Shen et al., 2011).

Inflammasome

NLRP3 inflammasome is an intracellular multi-meric protein complex composed of

three proteins namely, NLR containing N-terminal pyrin domain, apoptosis associated speck like

protein containing CARD (ASC) and pro-caspase-1 (Haitao Guo et al., 2015; Schroder & Tschopp,

2010). Upon NLRP3 complex formation, it leads to its activation via caspase-1 mediated cleavage

of pro-inflammatory cytokines into active IL-1 beta and IL-18. Secreted pro-inflammatory

cytokines mount an innate immune response as a defense mechanism (Haitao Guo et al., 2015;

Kate Schroder et al., 2010). Their assembly usually occurs upon stimulation of external stimuli

by wide variety of pathogens, environmental irritants, toxins and PAMPs (Pathogen associated

molecular patterns) (Haitao Guo et al., 2015; Kate Schroder et al., 2010). The most common

microorganisms that are hypothesized to induce NLRP3 inflammasome activation include candida

albicans, saccharomyces cerevisiae, Listeria monocytogenes, Staphylococcus aureus, adenovirus

and Influenze virus (Latz et al., 2013). Inflammasomes are implicated in initiation and progression

of neurodegerative disorders like Alzheimer Disease and Parkinson Disease (Freeman & P.-Y.

32

Ting, 2015). Using experimental models of PD, previous studies reported NLRP3 inflammasome

activation as a potential mechanism in PD associated neuro-inflammatory response (Lamkanfi et

al., 2010; Mao et al., 2017; Mustafa et al., 2016a).

There are two types of inflammasome namely, canonical inflammasome which leads to

activation of procaspase-1 whereas non-canonical leads to activation of procaspase-11 (Lamkanfi

et al., 2010). Both types of inflammasome leads to production of pro-inflammatory cytokines like

IL-1 beta and IL-18. Canonical type of inflammasome is activated in a 2 step mechanism. The

two steps for NLRP3 inflammasome activation include, priming and activation. The priming step

involves NF-kB activation which results in production of pro-IL-1 beta which is a pre-requisite for

the activation step (E. I. Elliott & F. S. Sutterwala, 2015; Haitao Guo et al., 2015; Lamkanfi et al.,

2010; Kate Schroder et al., 2010). The underlying mechanisms that are responsible for priming

step includes mainly, IRAK1 activation, ERK activation and spleen tyrosine kinase dependent

ASC phosphorylation (E. I. Elliott et al., 2015). Next, activation step involves the assembly of

NLRP3 scaffold, ASC adaptor and caspase-1 together to form a multimeric protein complex that

is capable is activating pro-IL-1 beta and Peo-IL-18 to active IL-1 beta and active IL-18

respectively (E. I. Elliott et al., 2015; Haitao Guo et al., 2015; Lamkanfi et al., 2010; Kate Schroder

et al., 2010).

Non canonical-inflammasome is usually activated by gram negative bacteria due to

the presence of Lipopolysaccharide (LPS) in their plasma membrane (P. Broz & Monack, 2013).

Gram negative bacteria engulfed by macrophages can trigger immune response via caspase-11

dependent pyroptosis (P. Broz et al., 2013; Ding & Shao, 2017; Pellegrini et al., 2017). Some of

the important functions of non-canonical inflammasome includes clearing of bacterial infections,

spread of infection, sepsis and activation of canonical inflammasome (P. Broz et al., 2013; Ding

33

et al., 2017; Pellegrini et al., 2017). Non-canonical inflammasome is activated via three step

process (Ding et al., 2017; Latz et al., 2013). In the first step, LPS and IFN-gamma induced

expression of pro-caspase-11 through NF-kB pathway (Petr Broz & Dixit, 2016; Ding et al., 2017;

Latz et al., 2013; V. A. Rathinam & Fitzgerald, 2016). In the second step, lipid A component of

the LPS binds to pro-caspase-11 and promotes oligomerization and activation of caspase-11 (Ding

et al., 2017). In the third step, activated caspase-11 leads to cleavage of gasdermin D (GSDMD)

resulting its migration to the plasma membrane and formation of pores resulting in osmotic lysis

(Petr Broz et al., 2016; Ding et al., 2017; Kayagaki et al., 2015; Latz et al., 2013; V. A. Rathinam

et al., 2016; Shi et al., 2015). Non-canonical inflammasome might be specifically associated with

production of pro-inflammatory cytokines like IL-1 alpha an HMGB1 (Lamkanfi et al., 2010).

Some of the important functional roles of non-canonical inflammasome include cell death,

cytokine maturation, actin remodeling, enterocolitis, renal fibrosis and clinical sepsis (Diamond et

al., 2015; Lorenz et al., 2014).

Both types of inflammasome activation ultimately results in pyroptosis. Pyroptosis is

caspase-1 mediated cell death and is regarded as an important hallmark of inflammasome

activation in tissue damage (W. Chang et al., 2013; Lamkanfi et al., 2010). The most important

distinguishing features include loss of cell membrane, DNA fragmentation, water influx, cellular

swelling and release of pro-inflammatory content into surrounding areas to mount an inflammatory

response (W. Chang et al., 2013; Lamkanfi et al., 2010).

Different models proposed to inflammasome activation

A wide variety of host derived factors can activate the NLRP3 inflammasome including

ATP, Asbestos, Silica, uric acid and amyloid (Halle et al., 2008; L Cassel et al., 2008; Sanjeev

34

Mariathasan et al., 2006; Martinon et al., 2006). Phosphorylation of ASC adaptor protein is the

most important factor leading to inflammasome activation (H. Hara, K. Tsuchiya, I. Kawamura,

R. Fang, E. Hernandez-Cuellar, Y. Shen, J. Mizuguchi, E. Schweighoffer, V. Tybulewicz, et al.,

2013). The most common models that are proposed for NLRP3 activation step includes the

following, K+ efflux, Lysosomal damage, ROS production, ER stress, calcium signaling and

mitochondrila damage (Cristiane M. Cruz et al., 2007a; Catherine Dostert, Virginie Pétrilli, Robin

Van Bruggen, Chad Steele, Brooke Mossman, et al., 2008; Halle et al., 2008; Hornung et al., 2009;

T.-D. Kanneganti et al., 2006; L Cassel et al., 2008).

K+ efflux

Extracellular ATP, pore forming toxins, Nigericin and MSU crystals are the most important

triggering factors for K+ efflux model of NLRP3 inflammasome activation (E. I. Elliott et al.,

2015; Haitao Guo et al., 2015; Lamkanfi et al., 2010; Kate Schroder et al., 2010). Activation of

P2X7 purinergic receptor by ATP results in increase in potassium efflux and decrease in

intracellular K+ (Abais-Battad et al., 2014). Additionally, ATP can also induce pore formation

through Pannexin-1 which mediates inflammasome activation and release of proinflammatory

cytokines (Pelegrin & Surprenant, 2006).

Lysosomal Damage

Particulate structures like silica, alum, amyloid and monosodium urate are engulfed by

lysosomes which is followed by lysosomal rupture and release of cathepsin-B into cytosol thereby

leading to inflammasome activation (E. Elliott & F. Sutterwala, 2015; Haitao Guo et al., 2015;

Lamkanfi et al., 2010; Kate Schroder et al., 2010). Accordingly, use of acidificaton inhibitors

which neutralize the acidification of lysosomal PH decrease the release of lysosomal cathepsin-B

and inflammasome activation (Hornung et al., 2009). Lysosomal calcium metabolism is also

35

involved in stabilization of pro-IL-1 beta mRNA synthesis; thus, emphasizing its role in regulation

of assembly and activation of inflammasome (Kassandra Weber & Joel D. Schilling, 2014).

Lysosomal membrane permeabilization (LMP) together with release of cathepsin-B and NADPH

oxidases (NOXs) can cause mitochondrial destabilization, thereby leading to mitochondrial ROS

production and NLRP3 inflammasome activation (Heid et al., 2013). In PD disease models, alpha-

synuclein aggregates induced production of cathepsin-B is responsible for triggering NLRP3

inflammasome activation (Codolo et al., 2013; Haitao Guo et al., 2015).

Reactive Oxygen Species

In the third model, ROS production is implicated in NLRP3 inflammasome activation.

Mitochondrial and non-mitochondrial ROS generation has been implicated in NLRP3

inflammasome (Catherine Dostert, Virginie Pétrilli, Robin Van Bruggen, Chad Steele, Brooke T.

Mossman, et al., 2008; K. Schroder et al., 2010; R. Zhou et al., 2011). The source of major ROS

production is usually mitochondria with minor contribution from NADP oxidases (Y. He et al.,

2016; Rodgers et al., 2014; Kate Schroder et al., 2010). Co-localization studies demonstrate that,

there is increased migration of NLRP3 near to damaged ROS generating mitochondria upon

treatment with inflammasome stimulants thus highlighting the importance of mitochondrial ROS

in NLRP3 inflammasome activation (Rodgers et al., 2014; R. Zhou et al., 2011). Complex I

inhibitors like rotenone leads to mitochondrial membrane depolarization, ROS production, NLRP3

inflammasome activation and pro-inflammatory cytokine production (Won et al., 2015; R. Zhou

et al., 2011). Accordingly, ROS inhibitors efficiently block the NLRP3 inflammasome activation

in rotenone treated cells (R. Zhou et al., 2011). Alternatively blockage of mitophagy leads to

accumulation of damaged ROS producing mitochondria and sustained NLRP3 inflammasome

activation (C. S. Shi et al., 2012). Voltage dependent calcium channels (VDCC), which are the

36

important channels of communication between ER and mitochondria for exchange of metabolites

and ions play an important role in mitochondrial ROS production and NLRP3 inflammsome

activation (R. Zhou et al., 2011). Inhibition of voltage dependent calcium channels resulted in

decreased caspase-1 activation and reduced IL-1 beta secretion (Rodgers et al., 2014; R. Zhou et

al., 2011). Additionally, phagocytosis of asbestosis and silica particles can lead to activation of

NADPH oxidase complex in the plasma membrane which triggers ROS production and ultimately

NLRP3 inflammasome activation (Catherine Dostert, Virginie Pétrilli, Robin Van Bruggen, Chad

Steele, Brooke T. Mossman, et al., 2008). Moreover, in ATP treated macrophages elevated

intracellular calcium can lead to enhanced ROS production and NLRP3 inflammasome activation

(Cristiane M. Cruz et al., 2007b). ROS can lead to loss of mitochondrial membrane potential and

opening of mitochondrial permeability transition pore causing the release of mitochondrial DNA;

a potent inducer of NLRP3 inflammsome activation (E. Elliott et al., 2015). Mitochondrial ROS

can also lead to lysosomal damage resulting in increasing in lysosomal membrane permeabilizaton

(LMP), release of lysosomal proteases and inflammasome activation (Heid et al., 2013). In PD

disease models, alpha- Synucluein aggregates induced production ofROS is responsible for

triggering NLRP3 inflammasome activation (Codolo et al., 2013; Haitao Guo et al., 2015).

Autophagy Dysfunction

Autophagy and inflammasome regulate each other thus playing a mutually important role

in disease progression and pathogenesis of many inflammatory diseases (Martins et al., 2015; S.

Qian et al., 2017). Autophagy induction is one of the main cellular defense mechanism that helps

in limiting and destruction of NLRP3 inflammasome assembly and activation (Harris et al., 2017;

S. Qian et al., 2017; Tatsuya Saitoh & Shizuo Akira, 2016; C.-S. Shi et al., 2012; Zhenyu Zhong

et al., 2016). The various mechanisms through which autophagy suppresses NLRP3

37

inflammasome activation includes removing mitochondrial DAMPs, degrading inflammasome

complexes and negatively regulating IL-1 beta synthesis, release and assembly (S. Qian et al.,

2017). Accumulation of damaged mitochondria during NLRP3 inflammasome activation leads to

compensatory upregulation of mitophagy and increased synthesis of P62 (M. J. Kim et al., 2016).

Migration of P62 to parkin associated depolarized mitochondria will ultimately facilitate their

removal by autophagy process (M. J. Kim et al., 2016; S. Qian et al., 2017; Tatsuya Saitoh et al.,

2016; Zhenyu Zhong et al., 2016). Similarly, P62 molecule migrates to ubiquitinated ASC proteins

thereby resulting in their destruction through autophagy process (S. Qian et al., 2017; Tatsuya

Saitoh et al., 2016). Interestingly, ubiquitinated NLRP3 proteins are made inactive until they are

deubiquitinated by BRCC3 enzyme but they are not targeted by autophagosomes for destruction

(Py et al., 2013; Rodgers et al., 2014). NLR proteins like NOD2 and NLRX1 interact with

autophagy proteins like Atg5-12 and Atg16 thus favoring autophagosome formation and clearance

of inflammatory proteins (Homer et al., 2012; Lei et al., 2012; Rodgers et al., 2014). In a similar

fashion, autophagy inducer sestrin2 (SESN2) protein is also upregulated and promotes removal of

damaged mitochondria during inflammasome activation (M. J. Kim et al., 2016). Non-canonical

inflammasome components, caspase-11 can also promotes fusion of lysosomes and phagosomes

and will aid in clearance of bacteria during infections (S. Qian et al., 2017). Administration of

autophagy inducers like rapamycin leads to recruitment of pro-IL-1 beta in autophagosomes and

subsequent degradation (S. Qian et al., 2017). Administration of autophagy inhibitors like

revasterol and 3-MA can lead increased mitochondrial ROS production and IL-1 beta production

(Y. P. Chang et al., 2015; S. Qian et al., 2017; Rodgers et al., 2014). Similarly loss of autophagy

proteins like LC3, beclin and Atg5 can lead to increase caspase-1 activation and IL-1 beta

production in macrophages (Nakahira et al., 2011; S. Qian et al., 2017; R. Zhou et al., 2011).

38

Capase-1 activation can result in feedback inhibition of autophagy through cleavage of parkin thus

amplifying the inflammasome activation (Alvarez-Arellano et al., 2018; S. Qian et al., 2017; J. Yu

et al., 2014). NLRP3 inflammasome assembly can lead to dysfunctional of autophagy process via

downregulating PINK1 (S. Qian et al., 2017; Y. Zhang et al., 2014). Lysosomal cathepsin-B can

trigger autophagy dysfunction and NLRP3 inflammasome activation in manganese treated BV2

microglia (Diya Wang et al., 2017). Obviously, autophagy dysfunction leads to accumulation of

depolarized mitochondria and damaged ROS producing mitochondria, release of oxidized

mitochondrial DNA into cytosol thereby leading to NLRP3 inflammasome activation in microglia

(E. Elliott et al., 2015; Harris et al., 2017; Tatsuya Saitoh et al., 2016; C.-S. Shi et al., 2012).

ER Stress

ER stress can cause activation of NLRP3 inflammasome assembly and activation through

various mechanisms. ER stress proteins PERK and IRE1 alpha leads to upregulation of CHOP

ultimately leading to NLRP3 inflammasome activation (D. N. Bronner et al., 2015; E. I. Elliott et

al., 2015; Lebeaupin et al., 2015b). Furthermore, ER stress can lead to activation of thioredoxin

interacting protein (TXNIP) which triggers NLRP3 inflammasome activation and secretion of pro-

inflammatory cytokines (Lerner et al., 2012; J. Song et al., 2015; Yoshihara et al., 2014).

Moreover, ER stress induced cleavage of caspase-2 and release of pro-apoptotic factor Bid can

trigger mitochondrial ROS production and NLRP3 inflammasome activation (D. N. Bronner et al.,

2015). Additionally, as ER is a major reservoir of intracellular calcium, it plays a major role in

regulating intracellular calcium levels, calcium signaling, NLRP3 inflammasome assembly and

activation (Yuan He et al.; G. S. Lee et al., 2012). Another study mentions that, deficiency of

uncoupling protein (UCP) in astrocytes leads to exacerbation of ER stress and accompanying

NLRP3 inflammasome activation (M. Lu et al., 2014). Alternatively, ER stress can disrupt the

39

physiological steady state communication between ER and mitochondria thereby triggering

mitochondrial dysfunction, enhanced ROS production and NLRP3 inflammasome activation

(Menu et al., 2012a). Lastly, excessive activation of voltage dependent anion channels (VDCC)

which are major source of communication between between endoplasmic reticulum and

mitochondria results in increased ROS production and inflammasome activation in microglial

cells (R. Zhou et al., 2011).

NLRC4

Gram negative Bacteria like salmonella typhimurium, Pseudomonas Aeruginosa,

Kelbsiella pneumonia, shigella and Bukholderia pseudomammei triggers NLCR4 dependent

caspase-1 activation and pyroptosis as an innate immune response (Duncan & Canna, 2018;

Hafner-Bratkovic, 2017). NLCR4 is related to clinical syndromes such as enterocolitis, auto-

inflammation and macrophage activation syndrome (Duncan et al., 2018). NLCR4 plays an

important role for mounting an immune response against systemic and enteric pathogens (Duncan

et al., 2018). Release of bacterial proteins (flagellin and type III secreted system) into cytosol

occurs as a result of fusion of bacteria-containing phagosomes with lysosomes (Latz et al., 2013;

Vijay A. K. Rathinam et al.). Both intracellular and extra cellular bacterial flagellin and T3SS

trigger immune response via NLCR4 inflammasome pathway (Zhao & Shao, 2015). Needle and

rod proteins of flagellin and Type III secreted system (T3SS) released into cytosol will be detected

by NAIP proteins (NBD domain containing inhibitor of apoptosis proteins) (Hafner-Bratkovic,

2017). Phosphorylation of the NLCR4 proteins leads to activation and enables it to interact with

NAIP proteins (Duncan et al., 2018). These bacterial ligands along with NAIP proteins bind to

NLRC4 proteins and attract other dormant NLRC4 molecules ultimately leading to formation of

NLRC4 inflammasome (Haitao Guo et al., 2015). NLRC4 inflammasome leads to formation strong

40

and robust immune response against bacterial pathogens (Hafner-Bratkovic, 2017). Another

unique feature of NLCR4 is that it can interact with pro-caspase-1 even in the absence of ASC

protein (Apoptosis speck containing a CARD) (Duncan et al., 2018). NLRP4 inflammasome

differs from NLRP3 inflammasome in that it does not require priming for activation (Denes et al.,

2015; Luigi Franchi et al., 2006). The role of ASC speck in NLCR4 inflammasome includes, pro-

caspase-1 cleavage, IL-1 beta and IL-18 maturation and neutrophil migration (Hafner-Bratkovic,

2017). Additionally, NLCR4 inflammasome activation is capable of producing pro-inflammatory

prostaglandins and leukotrienes via activation of arachidonic acid metabolism (Duncan et al.,

2018; von Moltke et al., 2012). The end target effects of NLCR4 inflammasome activation includes

pyroptosis, apoptosis, expulsion, eicosanoid secretion and pro-inflammatory cytokine secretion

(Duncan et al., 2018; Hafner-Bratkovic, 2017). The main functional significance of NLCR4

includes mounting an immune response against enteric, oral mucosal and lung pathogens (Cai et

al., 2012; L. Franchi et al., 2012; Tomalka et al., 2011). A growing body of evidence suggests that,

NLCR4 induced systemic inflammatory response following enteric pathogens can lead to ischemic

brain injury in mice (Denes et al., 2015). Recent studies suggest that, NLCR4 inflammasome is

associated with accentuating breast cancer progression and attenuating melanoma progression

(Janowski et al., 2016; Kolb et al., 2016). The other functions of NLRC4 includes actin

reorganization and infected epithelial cell expansion (Hafner-Bratkovic, 2017).

Microglia and Neuro-inflammation in PD

Under physiological conditions, normal microglia are present in resting or quiescent state

in healthy human brain (S. U. Kim & de Vellis, 2005; Q. Li & Barres, 2017; Wolf et al., 2017).

Microglial cells are regarded as first line of defense in response to injury, infection or toxin

41

exposure (S. U. Kim et al., 2005; Q. Li et al., 2017; Wolf et al., 2017). Slight disturbance in CNS

homeostasis will alert the resting microglial cells, which will proliferate and migrate to the site of

injury to destroy and remove the pathogens or damage associated molecules (S. U. Kim et al.,

2005; Q. Li et al., 2017; Wolf et al., 2017). They secrete chemokines and cytokines as well as

nitric oxide and reactive oxygen species to mount an immune response to remove the damaged

cell (S. U. Kim et al., 2005; Q. Li et al., 2017; Wolf et al., 2017). Uncontrolled and excessive

microglial activation can be deleterious to neighboring neurons and can lead to pathologic

consequences (S. U. Kim et al., 2005; Q. Li et al., 2017; Wolf et al., 2017). Oxidative stress,

proteosomal dysfunction and mitochondrial dysfunction have been implicated in the pathogenesis

of PD (J. L. Eriksen et al., 2005b). Activation of microglia is regarded as a histological hallmark

of brain pathology (Dheen et al., 2007). Microglial activation, production of pro-inflammatory

cytokines and neuro-inflammation are regarded as most common predisposing factors in

pathogenesis of neurodegenerative diseases like PD (Y. S. Kim & Joh, 2006; Sudhakar R.

Subramaniam & Howard J. Federoff, 2017). Prolonged deleterious crosstalk between reactive

microglia and neurons can ultimately lead to neuronal damage through release of pro-

inflammatory cytokines and reactive oxygen intermediates (Dheen et al., 2007; Wolf et al., 2017).

Activated microglia exhibit morphological changes consistent with transition from

neuroprotective to neurotoxic subsets and ultimately progression of dopaminergic

neurodegeneration in Parkinson’s disease (Sawada et al., 2006). In neuropsychiatric lupus, Tweak

stimulation of microglia resulted in increased expression of pro-inflammatory cytokines like TNF,

IP-10, MMP-9 and iNOS 4-6 fold as compared to isotype control (Stock & Putterman, 2014).

There is substantial evidence that, pharmacological blocking of Tweak / Fn14 pathway has

afforded protection in multiple models of neuro-inflammation (T. Dohi & L. Burkly, 2012). The

42

potential causative factors those are responsible for microglial neuro-inflammation in PD are not

very known. Activated microglia, T-lymphocyes and pro-inflammatory markers are present in the

substantia nigra of PD patients (Brochard et al., 2009; Hirsch et al., 2012; Sudhakar R.

Subramaniam et al., 2017). Additionally, endogenous pathogenic antigens like alpha synuclein

can lead to activation of adaptive immunity as evidenced by the presence of CD8+ and CD4+

lymphocytes in the brain of PD patients (Brochard et al., 2009; Hirsch et al., 2012). Chronic

microglial activation leads to the production of pro-inflammatory cytokines like TNF-alpha, IL-1

beta, IL-6 and IFN gamma which eventually leads to dopaminergic neurodegeneration in PD (J.

L. Eriksen et al., 2005b; Carina Cintia Ferrari et al., 2006; McCoy et al., 2006). The intracellular

signaling pathways that are responsible for microglial activation and pro-inflammatory cytokine

production includes MAPK (Mitogen activated protein kinase) pathways (Kiernan et al., 2016). It

is interesting to note that, polymorphisms of genes like TNF-alpha, IL-1 beta and IFN gamma were

associated with increased risk of developing PD (Hirsch et al., 2012). Accumulating evidence

suggests that, autosomal recessive PD associated genes like LRRK2, Parkin, Pink, DJ-1 are also

associated with microglial activation and neuro-inflammation in PD (J. L. Eriksen et al., 2005b).

Neurotoxic factors like alpha-synuclein, MMP-3 and ATP released from degenerating neurons can

amplify the neuro-inflammatory response and thus can continuously accentuate the cycle of

dopaminergic neurodegeneration in PD (George et al., 2015; Y.-S. Kim et al., 2007; W. Zhang,

Wang, et al., 2005). Accumulating evidence suggests that there are two microglial phenotypes that

can influence the process of neuro-inflammation in PD (Sudhakar R. Subramaniam et al., 2017).

Microglial phenotype (M1) is pro-inflammatory or disease causing phenotype and their activation

leads to production of pro-inflammatory cytokines like TNF-α, IL-1β, IL-6, IL-12, NOS and

reactive oxygen species which can accelerate the pro-inflammatory response in PD (Pisanu et al.,

43

2014; Sudhakar R. Subramaniam et al., 2017). Microglial phenotype (M2) is anti-inflammatory or

neuroprotective phenotype and their activation leads to production of anti-inflammatory cytokines

like IL-4, IL-13, IL-10, and TGF-β which can offer neuroprotection in PD (Pisanu et al., 2014;

Sudhakar R. Subramaniam et al., 2017). In addition, peripheral immune cells like macrophages

can migrate to the brain parenchyma via a breach in BBB and can lead to the generation of

cytokines like IL-1β, TNF-α and IL-6 (Blatteis, 1992). Blood brain barrier disruption and

endothelial dysfunction have been implicated in PD (Kortekaas et al., 2005). As a result, peripheral

macrophages and lymphocytes can enter the brain parenchyma and initiate neuro-inflammatory

response thus contributing to the pathogenesis of dopaminergic neurodegeneration in PD

(Kortekaas et al., 2005). In this context, immune response mounted by peripheral infections has

been regarded as a risk factor for development and progression of PD (Bu et al., 2015).

The various mechanisms through which microglia induced neuro-inflammation causes

oxidative stress in neurons includes production of Reactive oxygen species (ROS),

cyclooxygenase (COX) and lipoxygenase pathways (Tansey et al., 2007). Prostaglandins produced

by the microglial COX-2 enzymes induce oxidative stress in neurons by perturbation of

intracellular signaling mechanisms like decrease in mitochondrial membrane potential, decrease

in GSH activity, excessive generation of lipid peroxidation products and regulation of pro-

inflammatory cytokine production (Blom et al., 1997; Tansey et al., 2007). Eicosanoids produced

by the microglial lipo-oxygenase enzymes produce oxidative stress in neurons through production

of peroxide radicals and activation of programmed cell death pathways (Maccarrone et al., 2001).

Neuron-microglia interactions have been shown to regulate in microglial activation and neuro-

inflammation in the brain. In this regard, some of protein fragments released from degenerating

neurons like neuromelanin (NM) will enhance the microglial phagocytic activity against dying

44

dopaminergic neurons facilitating their own removal. Some of the immunomodulators secreted by

the neuronal cells like CX3CL1, CD200, CD22, CD47, CD95 and neural cell adhesion molecule

(NCAM), bind to and help to maintain microglia in their resting state (Hoek et al., 2000; Mott et

al., 2004; Numakawa et al., 2004; Sheridan & Murphy, 2013). Deficiency or dysfunction of these

immunomodulators results in conversion of resting microglia to activated microglia thus leading

to neuro-inflammatory response and dopaminergic neurodegeneration in PD.

Since microglial activation is considered one of the important contributing factor for

dopaminergic neuronal death, a wide variety of pharmacological interventions have been assessed

to rescue the dopaminergic neurons from cell death. Accumulating evidence suggests that, PPAR

gamma (peroxisome proliferator activated receptor) agonists and glucocorticoids receptor agonists

can protect against microglial activation, neuro-inflammation and dopaminergic

neurodegeneration in animal models of PD (Nolan et al., 2013). Additionally, minocycline (Broad-

spectrum antibiotic) has been effective in offering protection against dopaminergic neuronal cell

death owing to its ability to reduce blood brain barrier leakiness (BBB), microglial activation,

leucocyte migration and TNF upregulation (Ryu & McLarnon, 2006; Tansey et al., 2007;

Wasserman & Schlichter, 2007; Yong et al., 2004). Furthermore, NSAIDS (Non-Steroidal Anti-

inflammatory Drugs) like ibuprofen can potentially decrease the PD risk in the population by 20%-

30% probably due to their various anti-inflammatory effects (N. P. Rocha et al., 2015). The various

potential mechanisms through which NSAIDS act are through blocking PGE2 production (Blom

et al., 1997), suppressing glutamate excitotoxicity (Pasinetti, 1998), reduction of free radical

production (Lehmann et al., 1997), activation of PPAR gamma receptors (Lehmann et al., 1997;

Ricote et al., 1998) and attenuating oxidative stress (Kimura, 1997). Finally, antibiotic drug like

rifampicin reduces rotenone induced inflammation through suppressing NLRP3 inflammasome

45

activation, microglial activation and neuro-inflammation (Y. Liang et al., 2015). Further research

should be done to characterize the detrimental effects of microglia on neuronal survival in animal

models to facilitate the identification of new molecular targets.

Astrocytes

Astrocytes are glial cells and are related to physiological and pathological functions in the

central nervous system. They perform many functions like neuronal development, CNS blood

flow, transmitter homeostasis, synapse formation, metabolism and blood brain barrier maintenance

(Sofroniew & Vinters, 2010). In case of CNS injury, astrocytes are involved in neuroprotection,

blood brain barrier repair, reducing vasogenic edema and limiting the spread of infection to

surrounding areas in the central nervous system (Sofroniew et al., 2010). Astrocytes are associated

activation of Parkinsonian neurotoxin MPTP into active form MPP+ through mono-amino

oxidases (Przedborski et al., 2000). Astrocyte dysfunction due to mutated familial PD genes can

lead to impairment in inflammatory response, lipid handling, mitochondrial health and lysosomal

function (Booth et al., 2017). DJ-1 and parkin mutations can lead to astrocyte dysfunction and

neuronal death via mitochondrial stress and ER stress respectively (Phatnani & Maniatis, 2015).

Reactive astrogliosis has been evidenced in many regions like substantia nigra, putamen and

hippocampus in LPS and MPTP animal model of PD (Cabezas et al., 2014). Additionally, excess

alpha-synuclein accumulation in astrocytes can lead to impairment of their physiological functions

thereby accentuating dopaminergic neurotoxicity in PD (Gu et al., 2010). Activated astroglial cells

can secrete pro-inflammatory cytokines like TNF-alpha, IL-1 beta and IL-6 which can potentiate

dopaminergic neuronal death (Rappold & Tieu, 2010). Activated astrocytes can also cause

microglial activation which can ultimately result in pro-inflammatory cytokine production and

dopaminergic neurodegeneration in mid brain (Gu et al., 2010). Overexpression of Nrf2 in

46

astrocytes has been shown to afford protection against MPTP induced neurodegeneration in animal

model of PD (P. C. Chen et al., 2009).

Animal Models of PD

LPS Animal Model

Microglial activation and neuro-inflammation are recognized as key contributing factors

for dopaminergic neurodegeneration in PD. Therefore, development of appropriate models to

study the mechanism of neuro-inflammation will lead to development of effective therapeutic

interventions. LPS (Lipopolysaccride) is the endotoxin found in the outer membrane of gram

negative bacteria that can cause wide variety of systemic responses ranging from pyrexia to septic

shock (M. Liu & Bing, 2011; Schletter et al., 1996). LPS indirectly influences dopaminergic

neuronal survival via its effects on glial activation. LPS injection into brain results in upregulation

of CD14 receptor and acts through Toll like receptor (TL4) in the microglia (Hoshino et al., 1999;

Lacroix et al., 1998). It is interesting that, there are regional difference in LPS induced

neurotoxicity in the rat brain. Using in vitro cell culture models such as mixed neuronal cultures

of substantia nigra, LPS was found to induce the production of pro-inflammatory cytokines and

death of dopaminergic neurons whereas hippocampus and cortex mixed cultures were relatively

unresponsive to LPS treatment (W. G. Kim et al., 2000). LPS induced microglial activation results

in pro-inflammatory mediators like TNF alpha, IL-1 beta, nitric oxide species and reactive oxygen

species which ultimately results in dopaminergic neurodegeneration and neuronal cell death in PD

models (Arai et al., 2004; Boje & Arora; M. Liu et al., 2011). A growing body of evidence supports

the involvement of MAPK pathway in the mechanism of microglial activation response (Bhatia et

al., 2016; Bhatia et al., 2017; S. H. Kim, Smith, et al., 2004; Santa-Cecilia et al., 2016; Xing et al.,

2011). Additionally, downstream signaling mediator such as NADPH oxidase has been implicated

in LPS induced microglial reactive oxygen species (ROS) production and pro-inflammatory gene

47

expression which is linked to dopaminergic neurotoxicity (Liya Qin et al., 2004; Liya Qin et al.,

2007). Moreover, LPS and neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine(MPTP) can

act synergistically to elicit NADPH oxidase activation and NOS production leading to

dopaminergic neurodegeneration (H.-M. Gao et al., 2003).

Systemic LPS administration results in production of peripheral TNF alpha which crosses

the blood brain barrier and results in microglial mediated pro-inflammatory cytokine production

and delayed loss of dopaminergic neurons in substantia nigra in PD (Liya Qin et al., 2007). Along

with systemic administration of LPS, other invivo models were also developed to study the effect

of LPS in nigrostriatal dopaminergic system. In intra-nigral LPS model, LPS was stereotaxically

injected into substantia nigra (SN) of the mid brain and its effects were studied. In this context,

direct injection of LPS into SN of the mid brain resulted in microglial activation, reduction of

dopamine levels and tyrosine hydroxylase activity and ultimately resulting in dopaminergic cell

damage (Castaño et al., 1998). Furthermore, direct injection of LPS into SN of the mid brain

resulted in behavioral abnormalities, mitochondrial respiratory abnormalities, intracytoplasmic

inclusions and loss of dopaminergic neurons (D.-Y. Choi et al., 2009).

Another model that was used to study the LPS induced neuro-inflammatory response is

stereotaxic injection of LPS into striatum of midbrain (Intra-striatal LPS animal model).

Substantia nigra with relatively higher population of microglia is more susceptible than striatum

to stereotaxic injection of LPS as its main mode of action is via induction of strong microglial

activation and neuro-inflammation (Herrera et al., 2000). Stereotaxic LPS injection into substantia

nigra or striatum resulted in progressive dopaminergic degeneration and dopamine depletion via

production of production of proinflammatory cytokines like TNF-alpha, IL-6, IL-1 alpha and NO

both in substantia nigra and striatum (Castaño et al., 1998; M. Liu et al., 2011). Nigral stereotaxic

48

injection of LPS resulted in microglial activation, IL-1 beta release, caspase-11 activation and

decreased TH positive cells in substantia nigra of midbrain (Arai et al., 2004). In another study,

intrastrial stereotaxic LPS injection resulted in dopaminergic cell death mainly via NO induced

neuro-inflammation (L Hunter et al., 2009).

In another model, intranasal administration of LPS for 5 months resulted in behavioral

abnormalities, cytoplasmic inclusions, striatal dopamine loss and loss of dopaminergic neurons

(Q. He et al., 2013). In order to develop novel therapeutic interventions against LPS-induced

microglial activation response and associated dopaminergic neurodegeneration, studies were

performed in cell culture and animal models of PD. For example, revasterol (nonflavonoid

polyphenol) has been shown to be protective against LPS induced neurotoxicity by inhibiting

MAPK pathway, Nuclear factor-kB activating pathway and NADPH oxidase (F. Zhang et al.,

2010). Catalpol (an iridoid glycoside,contained richly in the roots of Rehmannia glutinosa) was

found to offer neuroprotection against LPS induced microglial activation by significantly reducing

the secretion pro-inflammatory mediators like TNF alpha, nitric oxide species (NOS) and reactive

oxygen species (ROS) (Tian et al., 2006). Other neuroprotective agents that were found to offer

protective against LPS induced neurotoxicity includes fluoxetine, 3-hydroxymorphinan, Gö6976

(selective protein kinase C (PKC) inhibitor and IL-10 through various mechanisms involving

inhibition of P38, MAPK pathway, NADPH oxidase and pro-inflammatory cytokine production

(Jeohn et al., 2002; Li Qian et al., 2006; F. Zhang et al., 2012; W. Zhang, Qin, et al., 2005).

MPTP Mouse Models

The most widely used experimental model of PD include the murine MPTP model. MPTP

(1-methyl-4-Phenyl-1, 2, 3, 6 tetrahydro-pyridine) is lipophilic and selectively destroys the

nigrostriatal neurons (G. Meredith & D. Rademacher, 2011). Despite the popularity of the model,

49

variable loss of nigral neurons, striatal DA depletion and neurobehavioral deficits have hampered

the utility of the model. Importantly, the MPTP models fails to reproduce the motor disturbance

evidenced in PD. Despite that it has been routinely used to better understand the pathological basis

of PD and cellular mechanisms that underlie the increased vulnerability of dopaminergic neurons

to the neutotoxic effects of MPTP. In this context, MPTP mouse models can display variable

degrees of nigral cell death, striatal dopamine loss and behavioral deficits depending upon the

dosage and timing of the MPTP regimen. The most commonly used MPTP models include acute,

subacute and chronic. MPTP can be co-administrated with probenecid which retards renal

clearance of MPTP thereby potentially increasing its neurotoxicity (Petroske et al., 2001). In

theacute MPTP model, C57black mice were injected with 4x 18 mg/kg of MPTP and sacrificed at

3h 6h 12h 24h and 72h after the last MPTP injection (N. Panicker, H. Saminathan, H. Jin, M. Neal,

D. S. Harischandra, et al., 2015). In subacute model, 15-30 mg/kg MPTP is given on a daily basis

for 5-10 days and animals are sacrificed 3 days after last injection (Anamitra Ghosh et al., 2012).

In chronic MPTP mouse model, 25 mg/kg MPTP is injected every 3.5 days for 5 weeks and

animals were sacrificed 1-3 weeks’ after injection (Meredith et al., 2008). A key aspect that is

evidenced in different MPTP mouse models include a reduction in dopamine uptake which

positively correlates with DA cell loss in substantia nigra (Meredith et al., 2008; Novikova et al.,

2006).

In Acute and Subacute mouse model, less than 50% of the nigral dopaminergic neurons

are destroyed whereas in chronic MPTP model there is wide spread destruction of nigral

dopaminergic neurons (Meredith et al., 2008). Acute MPTP model produces 40%

dopaminergic (DA) cell loss in substantia nigra (SN) and 90% loss of striatal DA 7 days after

50

treatment (Andrea Serra et al., 2008; G. Meredith et al., 2011). In Subacute MPTP model, death

of dopaminergic neurons occurs in apoptotic form 6 days after MPTP treatment and 59% striatal

DA depletion at 1day after MPTP treatment (Andrea Serra et al., 2008). Death of nigral neurons

occurs by non-apoptotic form in Acute MPTP model whereas it is apoptotic in subacute MPTP

model (G. Meredith et al., 2011).

Acute MPTP model regimen causes increase in DA release and its extracellular metabolites

like 3-methoxytyramine (3-MT) and decrease in dopamine metabolites like DOPAC (3, 4-

dihydroxyphenyl acetic acid) and HVA (homovanillic acid) in the dialysates of striatum (Andrea

Serra et al., 2008). In Sub-acute MPTP model, there is progressive day to day reduction in DA and

3-MT levels in the dialysates of striatum (Andrea Serra et al., 2008). It is interesting to know that

loss of striatal DA and reduction in dopamine uptake tend to spontaneously reverse over a period

of time in Acute and Subacute MPTP model (G. Meredith et al., 2011). In contrast, loss of striatal

DA and reduction in dopamine uptake tend to persist for 6 months in chronic MPTP model (G.

Meredith et al., 2011). Acute and Subacute MPTP models cause rapid transient neurotoxic insult

to the nigrostriatal system that does not exceed threshold for producing long lasting motor

abnormalities (Petroske et al., 2001). Whereas, in chronic MPTP model there is long lasting

neurotoxic injury to the nigrostriatal system which positively correlates with significant motor

abnormalities over a period of time (Petroske et al., 2001). In chronic MPTP model, mice show

significant disability as early as 3 days after MPTP treatment and impaired performance persists

for over 6 months (Novikova et al., 2006). Chronic MPTP model produces 50 -70% dopaminergic

(DA) cell loss in substantia nigra and 95-98% loss of striatal DA 3 weeks after treatment (Meredith

et al., 2008). Strong inflammatory responses with reactive microglia were visualized in SN as early

51

as 3 weeks after MPTP treatment and they persist for months in chronic MPTP model (Meredith

et al., 2008).

MPTP easily crosses the blood brain barrier due to its lipid solubility (Singer & Ramsay,

1990; Tieu, 2011). Once it enters the central nervous system, it is converted into its active MPP+

form by mono-amino oxidases (MAO-B) (Singer et al., 1990; Tieu, 2011). MPP+ is actively taken

up into nigral dopaminergic neurons by dopamine (DA) transporters (Singer et al., 1990; Tieu,

2011). Various mechanisms through which MPTP causes dopaminergic cell neurotoxicity includes

complex I inhibition, dopamine metabolism, disruption of calcium homeostasis, oxidative stress,

alteration of iron metabolism and GSH depletion. (Singer, T.P. et al 1990 & Sian, J. et al 1999).

Two most commonly proposed hypotheses for MPTP induced dopaminergic neurodegeneration

includes oxidative stress and mitochondrial impairment. According to the oxidative stress

hypothesis, there is generation of superoxide radicals and GSH depletion whereas in mitochondrial

impairment there is inhibition of NADH dehydrogenase, complex I and oxidative phosphorylation

(Singer, T.P. et al 1990).

Microglial activation is another most important feature that is present in both acute and

chronic MPTP models (G. Meredith et al., 2011). In MPTP treated mice there is significant

neuroinflammation characterized by upregulation of mRNA and proteins levels of TNF-alpha, IL-

1 beta and IL-6 cytokine and their receptor levels in substantia nigra and caudate-putamen regions

(Lofrumento et al., 2010). The neuroinflammation that develops in MPTP treated mice is not

limited to substantia nigra but can occur in other regions like olfactory bulb (Vroon et al., 2007).

Previous studies indicate that use of orally active apocynin and rosiglitazone loaded microspheres

(continuous dopaminergic stimulation) offered protection against MPTP induced neuro-

inflammation in mouse models of PD (Anamitra Ghosh et al., 2012; X. Yu et al., 2015).

52

There can be difference in the susceptibility of mouse strains to MPTP induced

dopaminergic cell loss, striatal DA depletion and behavioral abnormalities (G. Meredith et al.,

2011). The various underlying mechanisms underlying differential vulnerability of mouse strains

to MPTP induced neurotoxicity includes, uptake of MPTP into brain MAO-B expression, DA

transporter expression, metabolism of MPTP, number of TH positive neurons in the SN, and

neuroprotective mechanisms (Gainetdinov et al., 1997; Imai et al., 2001; Muthane et al., 1994;

Sedelis et al., 2000). Two most commonly used behavioral test that were used to assess the

dopaminergic cell loss and dopamine depletion in MPTP mouse models of PD include open rod

test, rotarod test (G. Meredith et al., 2011). In open rod test, the amount of time spent in moving

is measured whereas as rota rod test measures the duration spent by mice moving over the rod

(Rozas et al., 1997; Rozas et al., 1998; Sedelis et al., 2000). Another study used mice swim test

for measuring striatal dopamine depletion in MPTP model of Parkinson Disease (Haobam et al.,

2005).

Since MPTP treated mice develop dopaminergic neurodegeneration reminiscent of PD,

various interventions were studied and therapeutic benefit was monitored by measuring the

parameters like dopaminergic cell loss, DA depletion and behavioral abnormalities (G. Meredith

et al., 2011). Therapies that were targeting various mechanism of actions underlying MPTP

induced neurodegeneration has been developed and tested in the past. Therapeutic interventions

aimed at oxidative stress (Antioxidant agents), microglial activation (Apocynin, Minocycline,

PPAR gamma agonists), neuronal nitric acid synthase pathway (7-Nitroindazole), dopamine

deletion (Bromocriptine), caspase activation (Bcl-2), NADH linked oxidation have shown modest

therapeutic success (Breidert et al., 2002; S. Lin et al., 2003; Muralikrishnan & Mohanakumar,

1998; Nicklas et al., 1985; Perry et al., 1985; L. Yang et al., 1998).

53

6-OHDA model

The next most commonly used model for studying molecular pathogenesis in Parkinson

Disease is 6-OHDA (Hydroxy Dopamine) model. 6-OHDA model is used for testing the efficacy

of novel therapeutic interventions and for studying the mechanism of neurotransmission in

midbrain (Bové et al., 2005; Tieu, 2011). The most frequently used method of delivery of 6-OHDA

for developing animal model is intracerebral injection either into substantia nigra or striatum as it

cannot cross the blood brain barrier (Batassini et al., 2015; Bové et al., 2005; Tieu, 2011).

Intracerebral injection of 6-OHDA in substantia nigra produces dopaminergic cell death occurring

within 24h whereas delivery to striatum results in more protracted neurodegeneration with cell

death occurring within 1-3 weeks (Bové et al., 2005; Tieu, 2011). 6-OHDA striatum model

(progressive neuro degeneration) is regarded as a better model to study because it resembles

clinically progressive PD and is associated with non-motor symptoms (Tieu, 2011). It is important

to note here that, the success rate of intracerebral injection of 6-OHDA into smaller brain structure

like substantia nigra is only 30% whereas injection into bigger brain structure such as striatum has

a success rate is close to 90% (Simola et al., 2007; Tieu, 2011; Torres & Dunnett, 2012). In most

cases, 6-OHDA unilateral model is used and bilateral model is rarely used because it can lead to

severe neurological disabilities and ultimately death of the animal (Blesa et al., 2012). Another

advantage of unilateral 6-OHDA model is that, lesioned hemisphere can be compared with

contralateral non-lesioned hemisphere using behavioral tests (Blesa et al., 2012; Bové et al., 2005;

Tieu, 2011). 6-OHDA bears a structural resemblance with dopamine and thereby enters the

dopaminergic neurons through dopamine transporters (DAT) (Blesa et al., 2012; Bové et al., 2005;

Hanrott et al., 2006; Tieu, 2011). Due to its structural resemblance of 6-OHDA to catecholamine,

it can also pass through nor-epinephrine transporters and cause degeneration of central sympathetic

54

nervous systems (Bové et al., 2005; Hirsch et al., 2003; Tieu, 2011). The mechanism of 6-OHDA

induced neurotoxicity is debatable, but various mechanisms has been proposed such as oxidative

stress, mitochondrial dysfunction, caspase activation and ROS production (Bové et al., 2005;

Hanrott et al., 2006; Y. Saito et al., 2007). It is interesting to know that the 6-OHDA induced cell

death of dopaminergic neurons exhibit the features of both apoptosis and necrosis (Hanrott et al.,

2006). It has been proposed that, 6-OHDA induced apoptosis of dopaminergic neurons is due to

caspase-3 dependent cleavage of PKC-delta (Hanrott et al., 2006). Another mechanism by which

6-OHDA mediates dopaminergic cell death involves ERK activation which can be rescued by

parkin mediated P62 stabilization and upregulation (X. O. Hou et al., 2015; Kulich et al., 2007).

Astroglial cell involvement is also regarded as an important contributing factor for dopaminergic

degeneration in 6-OHDA model as evidenced by increase in CSF levels of GFAP and S100B

(Batassini et al., 2015; Bové et al., 2005). Lewy bodies, the hallmark pathological feature of PD

are not always present in 6-OHDA models (Bové et al., 2005). The two most commonly performed

behavioral tests for assessing 6-OHDA models include circular motor behavior and forelimb

akinesia whose severity is directly proportional to the degree of nigrostriatal degeneration (Blesa

et al., 2012; Deumens et al., 2002; Hanrott et al., 2006; Metz et al., 2005; Tieu, 2011). Additional

studies that can be done for studying 6-OHDA model include measuring dopamine levels in the

brain and examining the TH-positive neurons in the mid brain (De Jesús-Cortés et al., 2015; Stayte

et al., 2015). Nicotinamide adenine dinucleotide analogues (P7C3 compounds) are shown to be

neuroprotective against behavioral abnormalities, dopamine level changes and dopaminergic cell

death in 6-OHDA PD model in rats (De Jesús-Cortés et al., 2015).

55

Alpha-Synuclein Brain Infusion Model

Alpha-synuclein is found in Lewy bodies which are histological hallmark of Parkinson

Disease (Braak et al., 1999; Spillantini et al., 1998). Misfolded alpha-synuclein tends to aggregate

and is proposed to be involved in the dopaminergic neurodegeneration in PD (Braak et al., 1999;

Spillantini et al., 1998). Animal models were developed by delivering alpha-synuclein

intracerebrally either by injecting viral vectors or direct injection of mutant alpha-synuclein fused

to protein transduction domain (Bazzu et al., 2010; Kirik et al., 2002; Lo Bianco et al., 2004;

Recchia et al., 2008)

Lentiviral vectors are used for studying the effect of alpha-synuclein in triggering rapid

dopaminergic neurodegeneration and Lewy body pathology in substantia nigra in animal models

of PD (Chesselet, 2008). Rats are preferred over mice for lentiviral overexpression studies as they

have a larger brain size thus making it relatively easier to target substantia nigra for delivery of

alpha-aynuclein (Chesselet, 2008). Lentiviral overexpression models usually produce partial loss

of dopaminergic neurons as compared to toxin models which usually produce massive loss of

dopaminergic neurons due to differential expression of viral vectors witnin dopaminergic neuronal

cells (Kirik et al., 2002). The advantage of using lentiviral vectors overexpressing alpha-synuclein

is that, they can be administered unilaterally and contralateral untreated side can be used as a

control for comparison (Kirik et al., 2002).

Lentivirus overexpressing human alpha-synuclein injected stereotaxically into substantia

nigra resulted in 30-80% loss of dopaminergic neurons, 40-50% loss of striatal dopamine levels,

40-50% loss of tissue TH activity, significant behavioral abnormalities along with wide

distribution of alpha-synuclein positive inclusions within the dopaminergic neurons and

neurodegeneration (Kirik et al., 2002; Lo Bianco et al., 2002). In another model, direct stereotaxic

56

injection of alpha-synuclein fused to protein transduction domain (TAT-- AlphasynA30P) into

substantia nigra resulted in 26% loss of dopaminergic neurons and 78% loss of striatal dopamine

levels and 79% loss of DOPAC (Dihydroxyphenylacetic acid) (Recchia et al., 2008). In another

study Chung et al demonstrated that, AAV overexpressing alpha-synuclein injection into rat

substantia nigra resulted in changes in synaptic transmission, axonal transport and microtubules

along with neuroinflammation (Chung et al., 2009). Likewise, lentivirus over expressing mutant

alpha-synuclein injected into the striatum and substantia nigra of mice resulted in alpha-synuclein

aggregation within neurons, Lewy body pathology, alpha-synuclein positive neuronal varicosities

subsequently resulting progressive degeneration of alpha-synuclein positive neurons within one

year (Lauwers et al., 2003). Moreover, adenoviral overexpression of alpha-synuclein into rat

substantia nigra resulted in progressive dopaminergic neurodegeneration in SN over 13 weeks,

serine 129 phosphorylation of alpha-synuclein and caspase-9 activation with smilar changes in

cortical region (Mochizuki et al., 2006). In a similar fashion, adenovirus overexpression of alpha-

synuclein into rat substantia nigra resulted in progressive dopaminergic neurodegeneration,

activation of caspase-9 and upregulation of heat shock proteins at 24 weeks (St Martin et al., 2007).

Intrastriatal injection of pathogenic alpha-synuclein fibrils resulted in cell to cell

transmission, Lewy body pathology, loss of dopaminergic neurons, dopamine reduction in

substantia nigra pars compacta associated with behavioral deficits (K. C. Luk et al., 2012; Paumier

et al., 2015). Intrastriatal injection of Lewy bodies into monkeys and mice results in neuronal

uptake of alpha-synuclein, transmission of alpha-synuclein anterogradely and retrogradely

ultimately resulting in progressive dopaminergic neurodegeneration (Recasens, S. et al).

57

Spleen Tyrosine Kinase (SYK)

Structure and Expression

Syk is a non-receptor tyrosine kinase, which has two Src homology domains (SH2) and

kinase domain (Futterer et al., 1998; Mócsai et al., 2010; Taniguchi et al., 1991). These domains

are interconnected by inter-domains: A and B (Geahlen, 2014; Sada et al., 2001). Its Molecular

weight is 74 kDa (Mócsai et al., 2010). Alternative splicing can yield another form namely Syk-B

that lacks 29 AA from inter-domain B (Turner et al., 2000; L. Wang et al., 2003). There are some

structural resemblances between human and mouse Syk (Andres et al., 1995). Mammalian Syk or

Zeta chain associated protein kinase (ZAP70) bears close structural resemblance with Syk

although less homology with Syk amino acid sequences (Andres et al., 1995). Mostly, Syk is

located near the plasma membrane in the cytoplasm but, sometimes it is localized in the nucleus

too (Kulathu et al., 2009; H. Ma et al., 2001; S. Yanagi et al., 2001). Syk is expressed in wide

variety of cell types. Hematopoietic cells, macrophages, neuronal cells, fibroblasts, epithelial cells

and vascular endothelial cells express Syk in varying proportions (S. Yanagi et al., 2001; Shigeru

Yanagi et al., 1995).

Mechanism of Activation

The factors that can activate Syk include Low-Density Lipoprotein (LDL)(S. H. Choi et

al., 2009) Lipopolysaccharide (Miller et al., 2012), Pathogen-Associated Molecular Patterns

(PAMPs) (Rogers et al., 2005) and Damage-Associated Molecular Patterns (DAMPs) (Huysamen

et al., 2008; Sancho et al., 2009; Yamasaki et al., 2008). In the resting cells, syk is in inactive auto-

inhibited state due to linking of kinase domain with inter-domain A & B (Geahlen, 2009; Kulathu

et al., 2009). Phosphorylation of tyrosine residues in the inter domains A and B will result in release

of syk from auto-inhibited state to its active state (Geahlen, 2014; Mócsai et al., 2010). The most

58

crucial event for activation of Syk include phosphorylation of ITAMs (Immuno-receptor tyrosine-

based activation motifs) in immune cells (Hirose et al., 2004; C. A. Lowell, 2011). Phosphorylation

of ITAM tyrosine is through Src family kinases like Lyn (H. Ma et al., 2001). Next, syk binds to

the phosphorylated tyrosines of ITAM receptors which will result in phosphorylation of additional

tyrosine residues on syk kinase leading to activated spleen tyrosine kinase (Geahlen, 2009, 2014;

Mócsai et al., 2010; Tsang et al., 2008). Activated syk kinase will activate downstream molecules

to mediate various signaling events within the immune cells (Geahlen, 2009; Mócsai et al., 2010;

Tsang et al., 2008). Alternatively, another report mentions that Syk can also be activated by

binding to cytoplasmic tail of 3 integrin without interaction with phosphorylated ITAMs

(Woodside et al., 2001). Active Syk with its phosphorylated tyrosines can interact with

downstream signaling molecules like VAV, PLC-gamma, PI3K and SLP-76/65 (Geahlen, 2014;

C. A. Lowell, 2011; Mócsai et al., 2010; Ying et al., 2011). Furthermore, Syk regulates

physiological and pathological processes through interaction with intermediate signaling

molecules like NF-kB, CARD-BCL10-MALT complex, AKT, Ca+, MAP kinases, PKC and

protein kinase2 (Geahlen, 2014; C. A. Lowell, 2011; Mócsai et al., 2010).

Functions

Syk mediates cytoskeleton rearrangement, Reactive Oxygen Species (ROS) generation,

pro-inflammatory cytokine production and phagocytosis (Berton et al., 2005; S. H. Choi et al.,

2012; Jaumouille et al., 2014; M. J. Kim et al., 2012; Rogers et al., 2005; Y.-S. Yi et al., 2014a;

Yoon et al., 2013). Furthermore, it is involved in diverse biological processes like cell adhesion,

innate pathogen recognition, inflammatory responses and antibody dependent cytotoxicity (Endale

et al., 2013; Jeong et al., 2014; Mócsai et al., 2010; W. S. Yang et al., 2015).

59

Pathogen Recognition and Immune Response

Syk is required for P- Selectin Glycoprotein Ligand 1 (PSGL1) mediated leukocyte rolling

on the endothelium which is essential for transport of immune cells to the site of injury for

mounting an immune response (Mócsai et al., 2010; Newton & Dixit, 2012). Foreign pathogens

trigger Syk mediated phagocytosis, ROS and chemokine production in immune cells thus helping

in mounting an immune response in host cells in response to pathogens (Brungs et al., 2015;

Greenberg, 1999; Mócsai et al., 2010; Newton et al., 2012). Lastly, syk is responsible for Epstein-

barr virus transformation and lytic replication of Kaposi Sarcome virus within the infected B-cells

(Grywalska & Rolinski, 2015; Tomlinson & Damania, 2004).

NLRP3 Inflammasome

Spleen tyrosine kinase (Syk) is regarded as a major upstream regulating kinase responsible

for inflammasome activation in immune cells (Federica Laudisi et al., 2014). Nigericin, Mono

sodium urate and Alum has been known to cause Syk mediated inflammasome activation in LPS

primed peritoneal macrophages (Y. C. Lin et al., 2015). Syk has been shown to cause

phosphorylation of apoptosis associated speck-like protein containing a CARD (ASC) thus

promoting caspase-1 activation thereby, resulting in NLRP3 inflammasome mediated production

of pro-inflammatory cytokines like IL-1 beta and IL-18 (Gross et al., 2009; H. Hara, K. Tsuchiya,

I. Kawamura, R. Fang, E. Hernandez-Cuellar, Y. Shen, J. Mizuguchi, E. Schweighoffer, V.

Tybulewicz, et al., 2013). Syk triggers NLRP3 inflammasome mediated caspase-1 activation

through ASC polymerization and oligomerization in immune cells (Y. C. Lin et al., 2015).

Infection with fungal pathogens usually elicits NLRP3 inflammasome activation through Syk

regulated ROS production, K+ efflux and NF-kB signaling events (Gross et al., 2009). Syk

60

mediated signaling through CARD9-BCL10-MALT1 is essential in pathogen and tissue damage

recognition (Marakalala et al., 2010; Newton et al., 2012).

Platelet, Lymphocyte and Osteoclast Development

Syk mediated ITAM signaling is very important for osteoclast development and function

in bone metabolism through PLC-gamma2 (Phospho Lipase-C Gamma) and NFAT1 (Nuclear

Factor Activator of T-cells) (Mocsai et al., 2004; W. Zou et al., 2008). In platelets, Syk based

signaling is essential for development, function and arterial thrombus formation through PLC-

gamma2 and SLP76 (SH2 domain containing Leukocyte Domain) (Rivera et al., 2009). During

vascular development, Syk -ITAM signaling is useful for separation of blood and lymphatic

vessels via PLC-gamma2 and SLP76 (Abtahian et al., 2003). Syk is required differentiation of

pro-B cells to pre-B cells acting through intermediate signaling molecules like SLP76 (Herzog et

al., 2009).

Clinical Diseases

Spleen tyrosine kinase is associated with various inflammatory diseases like Rheumatic

arthritis, Allergic Asthma, Allergic Rhinitis, Gut ischemia reperfusion injury, Systemic Lupus

Erythematosis (SLE) and idiopathic thrombocytopenic purpura (ITP) (Bahjat et al., 2008; Bajpai

et al., 2008; Blease, 2005; Masuda & Schmitz, 2008; Podolanczuk et al., 2009; Wong et al., 2004).

Neurological Diseases

Numerous studies were done previously to implicate syk in neuro-inflammation after

subarachnoid hemorrhage in rats (Yue He et al., 2015). Previous findings indicate that,

phosphorylated-syk expression is increased in neurodegenerative diseases like Alzheimer disease

and Nasu-Hakola Disease (Satoh et al., 2012; Schweig et al., 2017). Increased Syk expression is

61

associated with neuroinflammation, gliosis, tau phosphorylation, amyloid beta accumulation and

pathological lesions of Alzheimer disease (Daniel Paris et al., 2014; Satoh et al., 2012; Schweig et

al., 2017). Fc receptor mediated Syk activation is also responsible for downstream signaling events

like cytokine secretion, phagocytosis and antigen presentation in Alzheimer Disease (Fuller et al.,

2014). In microglial cells, spleen tyrosine kinase is recruited to the stress granules leading to

phagocytic dysfunction and ROS & NOS production ultimately resulting in neuronal cell death (S.

Ghosh & Geahlen, 2015; Wolozin & Ikezu, 2015). Accumulating evidence suggests that, Syk

induced tyrosine phosphorylation of alpha-synuclein can be neuroprotective by decreasing its

oligomerization and aggregation (Lebouvier et al., 2008; A. Negro et al., 2002). In cerebral

ischemia, there is increased expression of Syk and phospho-syk 2h, 4h, 10h and 24h after

reperfusion (Yukiya Suzuki et al., 2013). Accordingly, Piceatannol (Syk inhibitor) protected

against cerebral via downregulation of phospho-syk, ICAM-1, MMP-9 and upregulation of

claudin-5 (Yukiya Suzuki et al., 2013). Furthermore, Syk inhibition resulted in resulted reduced

infarct size and offered neuroprotection in mice through decreased platelet activation (McKenzie,

2016). Additionally, mice treated with oral bioavailable syk inhibitor had reduced infarct size and

better neurological outcome 24h after transient middle cerebral artery occlusion (van Eeuwijk et

al., 2016).

Cancers

It can play a dual role in cancers. In hematological malignancies, it acts as a tumor promoter

whereas in cancers of non-immune cells it mediates tumor suppression (Krisenko & Geahlen,

2015). Spleen tyrosine kinase exhibits tumor suppressor activity by reducing metastasis and

promoting apoptosis in cancers such as breast cancer, hepatocellular carcinoma, melanoma and

pancreatic adenocarcinoma (Geahlen, 2014). In hematological malignancies like acute

62

lymphoblastic leukemia (ALL), chronic lymphocytic leukemia (CLL), Epstein Barr virus

associated lymphoma and non-hodgkins lymphoma, syk acts as a pro-survival factor through its

role in B-cell development and tonic signaling through B-cell receptor (Geahlen, 2014). In these

malignancies, it promotes tumor cell survival via activation of AKT, mTOR, PKC-delta, Mcl-1

and downregulation of GSK-3 beta (Geahlen, 2014). There is overexpression of Syk and tyrosine

Y525/526 phosphorylation in T-cell lymphomas (Feldman et al., 2008). In resistant diffuse B-cell

lymphomas, synthetic syk inhibitors resulted in G1-S cell cycle arrest via PLC-gamma and AKT

(Cheng et al., 2011). Accordingly, Syk inhibition would be an attractive therapeutic target for

therapeutic interventions in T-cell and B-cell lymphomas. So various Syk inhibitors like

fostamatinib, entospletinib and cerdulatinib are currently under development for various

hematological malignancies (D. Liu & Mamorska-Dyga, 2017). Retinoblastoma has high Syk

expression and it promotes survival via inactivation of GSK-3 beta and stabilization of anti-

apoptotic Mcl-1 (Myeloid cell leukemia sequence) (Geahlen, 2014; J. Zhang et al., 2012). In

human prostate and ovarian cancer, increased syk expression is associated with malignant

progression, recurrence and treatment resistance (Ghotra et al., 2015; Y. Yu et al., 2015).

Furthermore, it is associated with cancers like B-cell induced leukemia’s, Kaposi Sarcoma and

Epstein-Barr virus induced cancers (Mócsai et al., 2010).

Rheumatoid Arthritis

Rheumatoid arthritis (RA) is an autoimmune disease characterized by wide spread joint

destruction and bone inflammation due to hypersensitivity to self-antigens (Deng et al., 2016a;

Geahlen, 2014). It is an immune complex phenomenon mediated by type II and type III

hypersensitivity associated with IgG immune-complexes formed with self-antigens (Deng et al.,

2016a; Geahlen, 2014). Binding of IgG immune-complexes with Fc-gamma receptors on

63

neutrophils and macrophages leads to activation of Syk-ITAM signaling resulting in production of

reactive oxygen species, nitric oxide and pro-inflammatory cytokines (Deng et al., 2016a; Geahlen,

2014; Mócsai et al., 2010). Thus deposition of these immune complexes on joints and tissues leads

joint destruction and other symptoms characteristic of RA (Boers, 2011; Deng et al., 2016a;

Geahlen, 2014). Accordingly, blocking of syk-ITAM signaling with syk inhibitors leads to

attenuating joint destruction in animal models and phase II clinical trials in humans (Boers, 2011;

Deng et al., 2016a; Flight, 2012; Geahlen, 2014; Nijjar et al., 2013; D. L. Scott, 2011; I. C. Scott

& Scott, 2014). Previous reports indicate that, there are higher levels of Phospho-Syk in peripheral

blood of rheumatic arthritis patients as compared to control patients (Deng et al., 2016b).

Treatment with Syk inhibitor resulted in clinical benefits in patients with Rheumatic arthritis

receiving methotrexate therapy (Weinblatt et al., 2008). Combined inhibition of Syk and JNK

kinases results greater clinical success than syk alone inhibition in chronic destructive arthritis of

mice (Llop-Guevara et al., 2015). Syk is essential for RANK-FcR-gamma signaling required for

osteoclast activation and bone damage in mouse models of RA (Geahlen, 2014). Moreover, syk is

also required for TNF-alpha induced production of proinflammatory cytokines and other degrading

enzymes responsible for bone damage in RA (Geahlen, 2014). Furthermore, Syk inhibition

resulted in downregulation of JNK induced expression of pro-inflammatory IL-6 and MMP-3 in

synoviocytes (Cha et al., 2006). Lastly, syk is also required for migration, rolling and adherence

of macrophages, neutrophils and monocytes to inflamed endothelium thus indirectly playing an

important role on pathogenesis of atherosclerosis in RA (Geahlen, 2014).

64

Ischemic Reperfusion Injury

Syk activation has been linked to intestinal damage in intestinal ischemia reperfusion injury

model via Fcgamma receptor and complement signaling. In this context, blocking Syk mediated

Fcgamma receptor signaling in platelets resulted in reduced lung injury in intestinal mesentric

ischemia (Lapchak et al., 2012; Pamuk et al., 2010).

Tumor Necrosis Factor Weak stimulator of Apoptosis (TWEAK)

Introduction

Most of the members of the TNF family consist of ligands and receptors that activate

various signaling pathways to cause various effects ranging from cell survival, cell death and

development (Bodmer et al., 2002). Tweak is a member of TNF superfamily of cytokines. These

cytokines are synthesized as a type II trans membrane protein with a conserved C- terminal domain

(Bodmer et al., 2002). All the membrane bound forms can be cleaved by the certain CMK proteases

to a soluble form that can exert its actions in distant targets (Bodmer et al., 2002; BROWN et al.,

2003). The predisposing factors that regulate the processing of full length membrane tweak to

soluble tweak needs further investigation. The relative ratio of membrane tweak and soluble tweak

depends on the physiological and pathological state on the target tissues. The two forms of tweak

are functionally distinct and activate different downstream signaling pathways (Roos et al., 2010).

Tweak cDNA was accidently identified from mouse peritoneal macrophages during search

for erythropoietin messenger RNA. Later this cytokine was identified as Tweak due to its

proximity to TNF superfamily and its ability to weakly induce apoptosis (Chicheportiche et al.,

1997). After that, they identified complete human Tweak cDNA by performing PCR of human

tonsil and fetal liver tissues. Masters et al, also identified tweak cDNA from fetal kidney and

named it as APO3 ligand as it has affinity for death domain binding receptor Apo3 or DR3

65

(Marsters et al., 1998). Apo3 ligand can cause caspase activation, Nf-kB activation and apoptosis

(Marsters et al., 1998). Further analysis showed that human Tweak gene and Fn14 gene is localized

to chromosome 17 (Chicheportiche et al., 1997; Marsters et al., 1998; Meighan-Mantha et al.,

1999). There is close structural resemblance between human tweak sequences and members of

TNF superfamily which explains which might explain the commonality with the characteristics of

TNF superfamily. Furthermore, Tweak is a multipotent cytokine that is associated with various

biological functions like growth, differentiation, migration, apoptosis, angiogenesis and cytokine

production. These responses tend to vary with cell type, receptor activated and pathway involved.

The wide variety of cellular responses that are mediated by Tweak / Fn14 pathway in different cell

types might be responsible for maintaining normal health status and mediating various

pathological conditions. Tweak binds to its receptor Fn14 and exerts many of its actions (Burkly

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

Fibroblast growth factor inducible gene (Fn14) gene is regulated by growth factors and is

a type I trans membrane protein (Locksley et al., 2001). It does not share sequence similarity with

any other protein (Locksley et al., 2001). Fn14 is the major signaling receptor for Tweak. CD163

receptor appears to be scavenger receptor for tweak (Bover et al., 2007). CD163 receptor is mainly

present in the macrophages and monocytes and is mainly responsible for internalization and

degradation of soluble tweak (Bover et al., 2007). Thus, over expression of CD163 receptor can

sequester the soluble tweak and prevent it from exerting its biological actions. TNF super family

usually mediates downstream pathways through TRAF and Death Domain molecules (Fesik, 2000;

Inoue et al., 2000). Through these molecules, they activate pathways like NF-kB (nuclear kB) and

MAPK (mitogen activated protein kinase pathways) (Bover et al., 2007; BROWN et al., 2003;

Burkly et al., 2011; Locksley et al., 2001). Additionally, various kinases such as Glycogen

66

Synthase kinase-3 and sphingomyelinase are also activated by TNF family members (Naismith et

al., 1995; Novoyatleva et al., 2010; Wajant et al., 1999). Tweak shares some of the above-

mentioned features with the TNF family.

One of the most important signaling pathways activated by Tweak / Fn14 interactions is

Nf-KB pathways which results in expression of target genes. Over expression of Fn14 receptor in

malignant glioma leads to increased migration rate through Rac1 / NF-kB dependent mechanism

which ultimately leads to poor patient survival rates (Tran et al., 2006). Tweak treatment or over

expression of fn14 in malignant glioma results in increased glioma cell survival and resistance to

cytotoxic therapy through NF-kB dependent upregulation of Bcl-xl / Bcl-w expression (Tran et

al., 2006). Evidence of activation of Nf-kB pathway by Tweak can be seen in glioblastoma cancer

(Tran et al., 2006), keratinocytes (L. Jin et al., 2004), renal tubular cells (A. B. Sanz et al., 2010),

and neurons (Polavarapu et al., 2005).

Tweak can activate Mitogen activated protein kinase (MAPK) pathway in some cell types.

Transforming growth factor activated kinase has been shown to be required for activation of

MAPK/ JNK pathway by Tweak / Fn14 interactions (Bhattacharjee et al., 2012; M. Kumar et al.,

2009). Tweak increases the expression of sclerotin through JNK and ERK pathways in osteoblasts

and thus reduces bone formation (Vincent et al., 2009). In addition, it down regulated bone

morphogenic protein expression (Alkaline phosphatase) and up regulated RANKL expression in

osteoblasts through activation of MAPK ERK pathway (Ando et al., 2006).

Tweak / Fn 14 pathway can be responsible for regulating normal health and physiological

status. There is evidence that, tweak causes hepatocyte proliferation which can cause liver

regeneration after injury (Burkly et al., 2011; Girgenrath et al., 2006; Jakubowski et al., 2005). It

can also regulate mesenchymal stem cell proliferation (Girgenrath et al., 2006). Moreover, it also

67

acts on Fn14 receptor in mesenchymal stem cells and leads to increase in pro survival and cell

cycle responsive genes (Burkly et al., 2011; Girgenrath et al., 2006). Additionally, it can cause

blockade of myogenesis (Burkly et al., 2011; Dogra et al., 2006) adipogenesis (Burkly et al., 2011)

chrondrogenesis and osteogenesis (Perper et al., 2006). So abnormal function of this Tweak-Fn14

pathway in tissues might result in pathological tissue remodeling (Burkly et al., 2011; Zheng &

Burkly, 2008). This pathogenic tissue remodeling is important in regulating many autoimmune

and systemic disorders (W.-D. Xu et al., 2016; Zheng et al., 2008).

TNF family members are involved in wide variety of diseases like atherosclerosis,

osteoporosis, autoimmune diseases, allograft rejection and cancer (Locksley et al., 2001). Tweak

-Fn14 interactions were implicated in pathogenesis of many autoimmune disorders, systemic

diseases, cancers and neurological diseases (Burkly et al., 2011; Jeffrey A. Winkles, 2008). Tweak-

Fn14 signaling pathway in particular is associated with systemic lupus erythematosis (SLE), Lupus

nephritis, Rheumatoid arthritis, malignancies and skeletal muscle pathologies (Bodmer et al.,

2002; Burkly et al., 2011; Jeffrey A. Winkles, 2008). It also appears to be involved in various

neurological conditions like experimental autoimmune encephalitis and acute ischemic stroke and

multiple sclerosis (Burkly et al., 2011). It is quite possible that, Tweak / Fn14 pathway can be a

potential target for developing therapeutic interventions in the various systemic, autoimmune and

neurological disorders in the near future.

Tweak and Neurological Diseases

Tweak and its Fn14 receptor are expressed in tissues of central nervous system and are

found to be involved in pathogenesis of wide variety of neurological diseases. Tweak is also

expressed in neurons, astrocytes, microglia and endothelial cells (Desplat-Jego et al., 2009; Yepes,

2007; Yepes et al., 2005). Microglia but not astrocytes expressed membrane Tweak on them after

68

stimulation with interferon gamma (Yepes et al., 2005). The functional roles of membrane and

secreted Tweak in glial cells are yet to be determined. Tweak acting on the endothelial cells seems

to control the permeability of blood brain barrier (Yepes, 2007). Tweak stimulates the endothelial

cells to secrete cytokines like IL-6 that can contribute to leakage of blood brain barrier (Lynch et

al., 1999). According to Polavarupu et al, Tweak contributes to disruption of BBB by NF-kB

activation and MMP-9 upregulation in brain (Polavarapu et al., 2005). Another study mentions

that, under cerebral ischemia, Tweak upregulation leads to increased expression of monocyte

chemo-attractant (MCP-1) in astrocytes, increased passage of neutrophils into ischemic area thus

causing BBB disintegration through MMP-9 production (Woldeab B. Haile et al., 2010) Tweak

can cause proliferation and gliosis which is a hallmark of many degenerative and malignant

neoplasms in central nervous system (Yepes et al., 2005). The higher expression of Fn14 in

neurons suggests that it might be having some functional / physiological role in them. According

to Tanabe et al, over expression of Fn14 receptor occurs in Dorsal root ganglions after their injury

to promote growth cone lamelipodial formation and neuronal over growth (Tanabe et al., 2003).

After neural injury, Fn14 mediates neuronal regeneration through RAC1 GTPase dependent

mechanism via its ligand Tweak (Tanabe, K. et al 2003). Apart from Fn14, its ligand Tweak also

seems to have physiological function in neurons. Tweak causes inhibition of proliferation of neural

progenitor cells from sub-ventricular region of adult mice through activation of Nf-kB pathway

and lowered expression of Hes1 transcription factor (Scholzke et al., 2011). It is interesting to note

that, Tweak induced blockage of neuronal cell proliferation is due to suppression of neuronal

differentiation but not due to neuronal cell death.

69

Tweak and Cerebral Ischemia

Little is known about the predisposing factors that would enhance the expression of Tweak

and Fn14 receptor in neurons and glial cells. According to Haile et al, oxygen / glucose deprivation

was found to cause increased expression of Tweak and Fn14 in wild type neurons and wild type

astrocyte cultures (W. B. Haile, R. Echeverry, F. Wu, et al., 2010; Woldeab B. Haile et al., 2010;

Yepes et al., 2005). Oxygen / glucose deprived conditions can also cause increased Tweak / Fn14

expression in endothelial cell basement membrane – astrocyte interface (Woldeab B. Haile et al.,

2010). Tweak / Fn14 pathway in astrocytes – endothelial interface can lead to up regulation of

MCP-1 protein through activation of Nf-kB which is a strong chemotactic signal for macrophages

/ monocytes thus contributing to neuro inflammatory component in ischemic stroke (Woldeab B.

Haile et al., 2010). Tweak can stimulate endothelial cells, microglial cells and astrocytes to secrete

wide variety of pro-inflammatory cytokines that can also contribute to neuro inflammation in acute

stroke model (Bowerman et al., 2015; S. H. Kim, Kang, et al., 2004; Polavarapu et al., 2005;

Yepes, 2007; Manuel Yepes, 2013). Tweak can induce cell death by multiple mechanisms like

caspase activation, cathepsin-B mediated necrosis, TNF cooperation, MAPK, NF-kB activation

and JAK-STAT signaling (Chapman et al., 2013; Nakayama et al., 2002; Varfolomeev et al., 2012;

Vince et al., 2008; Harald Wajant, 2013). Acute Middle Cerebral Artery occlusion in mice leads

to up regulation of Tweak, Fn14, active caspase-3 with accumulation poly ADP ribose polymers

in ischemic areas as compared to contralateral normal side (Woldeab B. Haile et al., 2010). In

another experimental middle cerebral artery occulsion study, Tweak / Fn14 induced neuronal cell

death is due to activation of NF-kB pathway, caspase-3 activation and IKB alpha kinase activation

but is completely independent of TNF alpha cooperation (Woldeab B. Haile et al., 2010; Potrovita

et al., 2004). Previous studies indicate that, monitoring serum Tweak levels might be beneficial

70

and their levels can correlate with prognosis and severity of neurological diseases like cerebral

ischemia. According to Inta et al, serum Tweak levels were elevated above normal within 24 h

within stroke onset (Inta et al., 2008). Within 24 h of cerebral ischemia, Tweak / Fn14 levels were

up regulated in primary cortical neurons (Potrovita et al., 2004). The elevated serum Tweak levels

within 24 h of stroke onset might represent release of excess Tweak from infarcted brain tissues

(Inta et al., 2008). Thus monitoring serum Tweak levels within 24h of stroke might indicate

severity of ischemia thereby warranting immediate therapy to rescue infarcted brain tissue.

Developing therapeutic interventions against Tweak / Fn14 receptor might afford protection

against and prevent worsening of cerebral ischemia. According to Yepes et al, administration of

soluble Fn14-Fc decoy receptor can be helpful in preventing neuronal cell death, microglial

activation and reducing infarct volume in middle cerebral artery occlusion (Yepes, 2007; Manuel

Yepes, 2013; Yepes et al., 2005). Likewise, Zhang et al, showed that administration of similar

Fn14-Fc decoy receptor within 1 hr of stroke onset can decrease NF-kB activation, MMP-9

production and blood brain barrier degradation (X. Zhang et al., 2007). Conversely other studies

propose that, Tweak / Fn14 pathway activation during cerebral ischemia might be beneficial and

offers ischemic tolerance to surviving neurons in the ischemic penumbra. According to Echeverry

et al, up regulation of Tweak / Fn14 during cerebral ischemia helps in promoting ischemic

tolerance thereby promoting survival via ERK activation, TNF alpha upregulation and inactivation

of B-cell lymphoma associated death promoter protein (Echeverry et al., 2012). Taken together,

the neuro inflammatory, neurotoxic and neuro protective role of Tweak / Fn14 pathway in acute

middle cerebral artery occlusion and cerebral ischemia still need to be investigated in the near

future.

71

Tweak and Multiple Sclerosis

Multiple sclerosis is a demyelinating disease that usually has relapsing and remitting

pattern of clinical course. The main pathological hallmarks of multiple sclerosis include myelin

loss, oligodendrocyte destruction, plaque formation and accumulation of reactive astrocytes /

microglia (Iocca et al., 2008; Lucchinetti et al., 2000). Tweak is also expressed in spinal cord and

seems to be involved in the pathogenesis of diseases. Tweak and Fn14 expression is up regulated

in the spinal cord but not in encephalon in EAE and thus it can be potentially involved in

pathogenesis of experimental autoimmune encephalitis (Desplat-Jego et al., 2005; Iocca et al.,

2008; Mueller, Pedré, et al., 2005). Lymphocytes, macrophages and glial cells might be

responsible for the increased Tweak expression in spinal cord during experimental autoimmune

encephalitis (Mueller, Pedre, et al., 2005; B. Serafini et al., 2008). The exact mechanism by which

Tweak causes demyelination of the spinal cord neurons is not well known until now. In cuprizone

induced model of EAE, Tweak null mice tend to have decreased microglia accumulation and

significant delay in demyelination thus providing the evidence for involvement of Tweak in

pathophysiology of EAE (Iocca et al., 2008). Tweak can cause accumulation of reactive microglia

and decrease in oligodendrocytes in corpus callosum thus contributing to the pathogenesis of

experimental autoimmune encephalitis (EAE). Tweak / Fn14 pathway can activate multiple

signaling pathways to produce end target effects in EAE. The downstream signaling pathways that

are activated by Tweak / Fn14 interactions in EAE include NF-kB and MAPK pathways (A. Nazeri

et al., 2014). According to Nazeri et al, the various mechanisms by which Tweak mediates EAE

includes neuro degeneration, tissue remodeling, BBB disruption and astrogliosis (A. Nazeri et al.,

2014). So based on the above mentioned facts, Tweak and Fn14 would be potential targets in

reducing the severity of clinical and histological parameters in experimental autoimmune

72

encephalitis. According to Desplat–Jego et al, administration of anti-Tweak monoclonal antibodies

resulted in decreased CNS inflammation, angiogenesis, neuronal loss and clinical severity on EAE

(Desplat-Jego et al., 2005). Tweak can cause endothelial cells and astrocytes to release CCL2 /

MCP-1 which is a potent chemo-attractant for inflammatory cells into CNS / spinal cord thus

amplifying neuro inflammation (Woldeab B. Haile et al., 2010; Mueller, Pedre, et al., 2005).

Vaccination with extracellular domain of Tweak / Fn14 decreased the mononuclear infiltration and

clinical severity in rat and murine variant of EAE (Mueller, Pedre, et al., 2005). T- lymphocytes

also seem to play important role in pathogenesis of MS and EAE. Administration of Fn14 – TRAIL

fusion protein reduced the clinical severity in EAE by dampening T- lymphocyte induced

inflammatory response in spinal cord and brain (Prinz-Hadad et al., 2013). Taken together, we can

assume that Tweak / Fn14 pathway can cause neuro degeneration, neuro inflammation, neuronal

loss and astrogliosis thereby perpetuating the extent of neurological injury in the mouse / rat model

of experimental autoimmune enchepalomyelitis. Further research should take place to explore the

regulation of Tweak expression and downstream signaling pathways activated to develop novel

therapeutic strategies to reduce clinical severity in MS and EAE.

Tweak and Cerebral Neoplasms

Tweak / Fn14 pathway is involved in regulation of many cancers including systemic and

brain neoplasms. Tweak and Fn14 gene expression is seen in brain tumors like Glioblastoma

Multiforme (Prinz-Hadad et al., 2013). Tweak can regulate physiological function of tumor cells.

Tweak is involved in cancer cell survival, proliferation, migration and apoptosis (Winkles et al.,

2006). Exploring the Tweak pathway can yield much more information regarding its role in cancer

cell physiology. Fn14 receptor expression tends to vary between cancerous and normal brain

tissue. Fn14 receptor is over expressed in high grade brain tumors like pilocytic astrocytoma,

73

anaplastic astrocytoma, oligodendroglioma and Glioblastoma multiformae (Winkles et al., 2006).

Surprisingly, overexpression of Fn14 receptor is associated negative health outcomes. Over

expression of Fn14 receptor in malignant glioma leads to increased migration rate through Rac1 /

NF-kB dependent mechanism which ultimately leads to poor patient survival rates (Tran et al.,

2006). The capacity of cancer to metastasize and invade the neighboring tissues is usually

associated with poor clinical outcome. Angiogenesis or formation of new blood vessels is one the

most important predisposing factor for cancer metastasis (Winkles et al., 2006). Tweak can cause

endothelial cell proliferation and angiogenesis thus promoting tumor metastasis (Winkles et al.,

2006). Tumor cells produce fibroblast growth factor (FGF-2) and vascular endothelial cell growth

factor (VEGF) which can up regulate Fn14 gene expression (Donohue et al., 2003; Winkles et al.,

2006). Tweak, FGF-2 and VEGF can cooperate with each other in mediating vascular proliferation

thus causing tumor cell metastasis. The downstream signaling pathways that will be activated by

Tweak / Fn14 interactions to promote tumor migration will be a potential target for interventions

in the future. It was shown that, Tweak / Fn14 pathway in Glioblastoma causes tumor cell

migration by activating RhoG- specific guanine nucleotide specific exchange factor which is

overexpressed in these neoplasms (Fortin Ensign et al., 2013). Cancer progression is also

dependent on the ability of the tumor cells to survive and withstand cytotoxic therapy. Tweak /

Fn14 pathway tends to regulate the survival of cancerous cells by activating specific signaling

pathways. Tweak treatment or over expression of fn14 in malignant glioma results in increased

glioma cell survival and resistance to cytotoxic therapy through NF-kB dependent upregulation of

Bcl-xl / Bcl-w expression (Tran et al., 2006). Another mechanism that helps in cancer cell survival

would be local invasion of cancer cells to surrounding brain tissue that would reduce the success

rate of surgical resections. There is evidence that, Tweak can function as chemotactic factor that

74

promotes Glioblastoma tumor cell migrate into surrounding brain tissue by activating Lyn Kinase

(Dhruv et al., 2014). Third mechanism by which Tweak / Fn14 pathway can increase Glioma cell

survival would be activation of Akt2 pathway (Fortin Ensign et al., 2013). Taken together, we can

assume that designing interventions against Tweak / Fn14 pathway can lead to better clinical

outcomes in high grade solid brain tumors by decreasing cancer cell survival, migration,

angiogenesis and metastasis. In one study, agonistic antiFn14 antibody is likely to be highly

efficient in hindering tumor burden in multiple xenograft models (Michaelson et al., 2011).

Another option would be to target downstream signaling pathways like NF-kB and Akt2 activated

with Tweak / Fn14 interactions that would increase clinical survival rate in these brain neoplasms.

Tweak and Blood Brain Barrier

The blood brain barrier limits the entry of large molecules from systemic circulation into

brain. It is formed collectively by microvascular endothelial cells, astrocytes, pericytes and

neurons (L. Liu et al., 2013; Rubin & Staddon, 1999). The capillary endothelial cells are bound

together tightly by tight junctions and zonula adherens which form a protective covering limiting

the entry of toxic substances into the brain circulation (Rubin et al., 1999). Tweak expressed in

endothelial cells is involved in endothelial cell proliferation and angiogenesis independent of

vascular endothelial growth factor (VEGF) (Lynch et al., 1999). Endothelial cells and astrocytes

present in BBB can secrete Tweak during inflammatory conditions thus leading to decreased

integrity of blood brain barrier (Yepes, 2007; Yepes et al., 2005). Tweak acting on the endothelial

cells controls the permeability of blood brain barrier (Yepes, 2007). Tweak stimulates the

endothelial cells to secrete cytokines like IL-6 that can contribute to leakage of blood brain barrier

(Lynch et al., 1999). According to Polavarupu et al, Tweak contributes to disruption of BBB by

NF-kB activation and MMP-9 up regulation in brain (Polavarapu et al., 2005). Another study

75

mentions that, under cerebral ischemia, increased Tweak leads to increased expression of

monocyte chemo-attractant protein (MCP-1) in astrocytes, increased passage of neutrophils into

ischemic area and increased MMP-9 production thereby mediating blood brain barrier

disintegration (Woldeab B. Haile et al., 2010). According to Stephan et al, Tweak can stimulate

capillary endothelial cells to secrete pro-inflammatory cytokines and cell adhesion molecules

through MAPK pathway thereby increasing blood brain barrier permeability (Stephan et al., 2013).

Any compromise in the integrity of the blood brain barrier can lead to entry of toxic substances,

immune cells and inflammatory cells from systemic circulation into brain circulation thereby

causing deleterious effects. According to Wen et al, Tweak induced leakage in blood brain barrier

can lead to entry of autoantibodies, activated lymphocytes and cytokines into brain thus causing

neuropsychiatric manifestations in lupus nephritis patients (Wen et al., 2013). Based on the above

mentioned facts we can conclude that, Tweak / Fn14 pathway plays an important role in regulating

the integrity of BBB. Developing therapeutic agents to block the Tweak / Fn14 pathway can

prevent leakage via NF-kB activation, MAPK activation, MMP-9 production, MCP-1 production,

basement membrane laminin degradation and neuropsychiatric manifestations in pathological

conditions of the brain (Woldeab B. Haile et al., 2010; Polavarapu et al., 2005; Stephan et al.,

2013; Wen et al., 2013; X. Zhang et al., 2007).

Tweak and Parkinson’s Disease

There are very few studies done to study the role of Tweak /Fn14 pathway in dopaminergic

neurodegeneration of PD. According to a recent study, Tweak protein expression is increased in

striatum but not in substantia nigra in MPTP treated mice (Mustafa et al., 2016a). Furthermore,

neutralizing the Tweak /Fn14 interactions with the help of anti-Tweak antibody resulted in

substantial reduction in MPTP induced neurotoxicity in Sub acute MPTP model (Mustafa et al.,

76

2016a). The neutralizing effects of Tweak-FN14 blockade in subacute acute MPTP model used in

this study might be due to protective effects against apoptotic cell death or microglial activation.

In the absence of Tweak, MPTP induced dopaminergic neurodegeneration occurs via TNF alpha

induced apoptotic cell signaling events (Mustafa et al., 2016b; Ray et al., 2015; Sriram et al., 2002).

These studies indicate that, Tweak / Fn14 pathway plays a minor role in the pathogenesis of

dopaminergic neurodegeneration in PD. Further research should be done to study whether Tweak

level fluctuates in serum and CSF of patients of various stages of Parkinson disease.

Overall, interruption of Tweak / Fn14 pathway seems to have favorable outcomes in

neurological diseases like intervertebral disc degeneration, ischemic stroke, cerebral edema and

multiple sclerosis (Arash Nazeri et al., 2013; H. Wajant, 2013; W. D. Xu et al., 2016; Yepes et al.,

2005; Yepes & Winkles, 2006).

Bibliography

A Lynch-Day, M., et al. (2012). The Role of Autophagy in Parkinson's Disease (Vol. 2).

Abais-Battad, J., et al. (2014). Redox Regulation of NLRP3 inflammasomes: ROS as trigger or

effector? (Vol. 22).

Abtahian, F., et al. (2003). Regulation of blood and lymphatic vascular separation by signaling

proteins SLP-76 and Syk. Science, 299(5604), 247-251. doi:10.1126/science.1079477

Allyson Jones, C., et al. (2012). Incidence and mortality of Parkinson’s disease in older

Canadians. Parkinsonism & Related Disorders, 18(4), 327-331.

doi:https://doi.org/10.1016/j.parkreldis.2011.11.018

Alvarez-Arellano, L., et al. (2018). Autophagy impairment by caspase-1-dependent inflammation

mediates memory loss in response to beta-Amyloid peptide accumulation. J Neurosci

Res, 96(2), 234-246. doi:10.1002/jnr.24130

77

Alvarez-Erviti, L., et al. (2010). Chaperone-mediated autophagy markers in parkinson disease

brains. Archives of Neurology, 67(12), 1464-1472. doi:10.1001/archneurol.2010.198

Alvarez-Erviti, L., et al. (2010). Chaperone-mediated autophagy markers in Parkinson disease

brains. Arch Neurol, 67(12), 1464-1472. doi:10.1001/archneurol.2010.198

Ando, T., et al. (2006). TWEAK/Fn14 interaction regulates RANTES production, BMP-2-

induced differentiation, and RANKL expression in mouse osteoblastic MC3T3-E1 cells.

Arthritis Res Ther, 8(5), R146. doi:10.1186/ar2038

Andrea Serra, P., et al. (2008). The MPTP mouse model: Cues on DA release and neural stem

cell restorative role (Vol. 14 Suppl 2).

Andres, A. C., et al. (1995). Protein tyrosine kinase expression during the estrous cycle and

carcinogenesis of the mammary gland. Int J Cancer, 63(2), 288-296.

Anglade, P., et al. (1997). Apoptosis and autophagy in nigral neurons of patients with

Parkinson's disease. Histol Histopathol, 12(1), 25-31.

Antony, P. M., et al. (2013). The hallmarks of Parkinson's disease. Febs j, 280(23), 5981-5993.

doi:10.1111/febs.12335

Arai, H., et al. (2004). Neurotoxic effects of lipopolysaccharide on nigral dopaminergic neurons

are mediated by microglial activation, interleukin-1beta, and expression of caspase-11 in

mice. J Biol Chem, 279(49), 51647-51653. doi:10.1074/jbc.M407328200

Arduíno, D. M., et al. (2009). Endoplasmic reticulum and mitochondria interplay mediates

apoptotic cell death: Relevance to Parkinson's disease. Neurochemistry International,

55(5), 341-348. doi:https://doi.org/10.1016/j.neuint.2009.04.004

Arif, I., et al. (2010). Environmental toxins and Parkinson's disease: Putative roles of impaired

electron transport chain and oxidative stress (Vol. 26).

Ariga, H., et al. (2013). Neuroprotective Function of DJ-1 in Parkinson’s Disease.

Oxidative Medicine and Cellular Longevity, 2013, 9. doi:10.1155/2013/683920

78

Baba, M., et al. (1998). Aggregation of alpha-synuclein in Lewy bodies of sporadic Parkinson's

disease and dementia with Lewy bodies. The American Journal of Pathology, 152(4),

879-884.

Bahjat, F. R., et al. (2008). An orally bioavailable spleen tyrosine kinase inhibitor delays disease

progression and prolongs survival in murine lupus. Arthritis Rheum, 58(5), 1433-1444.

doi:10.1002/art.23428

Bajpai, M., et al. (2008). Spleen tyrosine kinase: a novel target for therapeutic intervention of

rheumatoid arthritis. Expert Opin Investig Drugs, 17(5), 641-659.

doi:10.1517/13543784.17.5.641

Batassini, C., et al. (2015). Striatal Injury with 6-OHDA Transiently Increases Cerebrospinal

GFAP and S100B. Neural Plasticity, 2015, 9. doi:10.1155/2015/387028

Bazzu, G., et al. (2010). alpha-Synuclein- and MPTP-generated rodent models of Parkinson's

disease and the study of extracellular striatal dopamine dynamics: a microdialysis

approach. CNS Neurol Disord Drug Targets, 9(4), 482-490.

Béraud, D., et al. (2011). α-Synuclein Alters Toll-Like Receptor Expression (Vol. 5).

Berton, G., et al. (2005). Src and Syk kinases: key regulators of phagocytic cell activation.

Trends Immunol, 26(4), 208-214. doi:10.1016/j.it.2005.02.002

Bhatia, H. S., et al. (2016). Rice bran derivatives alleviate microglia activation: possible

involvement of MAPK pathway. Journal of Neuroinflammation, 13, 148.

doi:10.1186/s12974-016-0615-6

Bhatia, H. S., et al. (2017). Alleviation of Microglial Activation Induced by p38

MAPK/MK2/PGE2 Axis by Capsaicin: Potential Involvement of other than TRPV1

Mechanism/s. Scientific Reports, 7(1), 116. doi:10.1038/s41598-017-00225-5

Bhattacharjee, M., et al. (2012). A Bioinformatics Resource for TWEAK-Fn14 Signaling

Pathway. Journal of Signal Transduction, 2012, 10. doi:10.1155/2012/376470

Bjorklund, A., et al. (2007). Dopamine neuron systems in the brain: an update. Trends Neurosci,

30(5), 194-202. doi:10.1016/j.tins.2007.03.006

79

Bjorkoy, G., et al. (2009). Monitoring autophagic degradation of p62/SQSTM1. Methods

Enzymol, 452, 181-197. doi:10.1016/s0076-6879(08)03612-4

Blatteis, C. M. (1992). Role of the OVLT in the febrile response to circulating pyrogens. Prog

Brain Res, 91, 409-412.

Blease, K. (2005). Targeting kinases in asthma. Expert Opinion on Investigational Drugs,

14(10), 1213-1220. doi:10.1517/13543784.14.10.1213

Blesa, J., et al. (2012). Classic and new animal models of Parkinson's disease. J Biomed

Biotechnol, 2012, 845618. doi:10.1155/2012/845618

Blesa, J., et al. (2015). Oxidative stress and Parkinson's disease. Front Neuroanat, 9, 91.

doi:10.3389/fnana.2015.00091

Blom, M. A., et al. (1997). NSAIDS inhibit the IL-1 beta-induced IL-6 release from human post-

mortem astrocytes: the involvement of prostaglandin E2. Brain Res, 777(1-2), 210-218.

Bodmer, J. L., et al. (2002). The molecular architecture of the TNF superfamily. Trends Biochem

Sci, 27(1), 19-26.

Boers, M. (2011). Syk kinase inhibitors for rheumatoid arthritis: trials and tribulations. Arthritis

Rheum, 63(2), 329-330. doi:10.1002/art.30109

Bohingamu Mudiyanselage, S., et al. (2017). Cost of Living with Parkinson’s Disease

over 12 Months in Australia: A Prospective Cohort Study. Parkinson’s Disease,

2017, 13. doi:10.1155/2017/5932675

Boje, K. M., et al. Microglial-produced nitric oxide and reactive nitrogen oxides mediate

neuronal cell death. Brain Research, 587(2), 250-256. doi:https://doi.org/10.1016/0006-

8993(92)91004-X

Bonifati, V., et al. (2003). Mutations in the DJ-1 gene associated with autosomal recessive early-

onset parkinsonism. Science, 299(5604), 256-259. doi:10.1126/science.1077209

Booth, H. D. E., et al. (2017). The Role of Astrocyte Dysfunction in Parkinson’s Disease

Pathogenesis. Trends in Neurosciences, 40(6), 358-370. doi:10.1016/j.tins.2017.04.001

80

Bové, J., et al. (2005). Toxin-Induced Models of Parkinson's Disease. NeuroRx, 2(3), 484-494.

Bover, L. C., et al. (2007). A previously unrecognized protein-protein interaction between

TWEAK and CD163: potential biological implications. J Immunol, 178(12), 8183-8194.

Bowerman, M., et al. (2015). Tweak regulates astrogliosis, microgliosis and skeletal muscle

atrophy in a mouse model of amyotrophic lateral sclerosis. Hum Mol Genet, 24(12),

3440-3456. doi:10.1093/hmg/ddv094

Boya, P., et al. (2013). Emerging regulation and functions of autophagy. Nature Cell Biology, 15,

713. doi:10.1038/ncb2788

Braak, H., et al. (1998). Evolution of neuronal changes in the course of Alzheimer's disease. J

Neural Transm Suppl, 53, 127-140.

Braak, H., et al. (2003). Staging of brain pathology related to sporadic Parkinson's disease.

Neurobiol Aging, 24(2), 197-211.

Braak, H., et al. (1999). Extensive axonal Lewy neurites in Parkinson's disease: a novel

pathological feature revealed by alpha-synuclein immunocytochemistry. Neurosci Lett,

265(1), 67-69.

Bravo, R., et al. (2013). Chapter Five - Endoplasmic Reticulum and the Unfolded Protein

Response: Dynamics and Metabolic Integration. In K. W. Jeon (Ed.), International

Review of Cell and Molecular Biology (Vol. 301, pp. 215-290): Academic Press.

Breckenridge, D. G., et al. (2003). Regulation of apoptosis by endoplasmic reticulum pathways.

Oncogene, 22(53), 8608-8618. doi:10.1038/sj.onc.1207108

Bredesen, D. E., et al. (2006). Cell death in the nervous system. Nature, 443(7113), 796-802.

doi:10.1038/nature05293

Breidert, T., et al. (2002). Protective action of the peroxisome proliferator-activated receptor-

gamma agonist pioglitazone in a mouse model of Parkinson's disease. J Neurochem,

82(3), 615-624.

Bresgen, N., et al. (2015). Oxidative Stress and the Homeodynamics of Iron Metabolism.

Biomolecules, 5(2), 808-847. doi:10.3390/biom5020808

81

Brochard, V., et al. (2009). Infiltration of CD4+ lymphocytes into the brain contributes to

neurodegeneration in a mouse model of Parkinson disease. The Journal of Clinical

Investigation, 119(1), 182-192. doi:10.1172/JCI36470

Bronner, D. N., et al. (2015). Endoplasmic Reticulum Stress Activates the Inflammasome via

NLRP3- and Caspase-2-Driven Mitochondrial Damage. Immunity, 43(3), 451-462.

doi:10.1016/j.immuni.2015.08.008

Brooks, D. J. (2010). Examining Braak's hypothesis by imaging Parkinson's disease. Mov

Disord, 25 Suppl 1, S83-88. doi:10.1002/mds.22720

BROWN, S. A. N., et al. (2003). The Fn14 cytoplasmic tail binds tumour-necrosis-factor-

receptor-associated factors 1, 2, 3 and 5 and mediates nuclear factor-kappaB activation.

Biochemical Journal, 371(2), 395-403. doi:10.1042/bj20021730

Broz, P., et al. (2016). Inflammasomes: mechanism of assembly, regulation and signalling.

Nature Reviews Immunology, 16, 407. doi:10.1038/nri.2016.58

Broz, P., et al. (2013). Noncanonical inflammasomes: caspase-11 activation and effector

mechanisms. PLoS Pathog, 9(2), e1003144. doi:10.1371/journal.ppat.1003144

Brungs, S., et al. (2015). Syk phosphorylation – a gravisensitive step in macrophage signalling.

Cell Communication and Signaling : CCS, 13, 9. doi:10.1186/s12964-015-0088-8

Bu, X.-L., et al. (2015). The association between infectious burden and Parkinson's disease:

A case-control study. Parkinsonism & Related Disorders, 21(8), 877-881.

doi:https://doi.org/10.1016/j.parkreldis.2015.05.015

Burguillos, M. A., et al. (2011). Apoptosis-inducing factor mediates dopaminergic cell death in

response to LPS-induced inflammatory stimulus: evidence in Parkinson's disease

patients. Neurobiol Dis, 41(1), 177-188. doi:10.1016/j.nbd.2010.09.005

Burke, R. E. (2008). Programmed cell death and new discoveries in the genetics of

parkinsonism. J Neurochem, 104(4), 875-890. doi:10.1111/j.1471-4159.2007.05106.x

Burke, R. E., et al. (2008). A critical evaluation of the Braak staging scheme for Parkinson's

disease. Ann Neurol, 64(5), 485-491. doi:10.1002/ana.21541

82

Burke, R. E., et al. (2008). A Critical Evaluation of The Braak Staging Scheme for Parkinson’s

Disease. Annals of neurology, 64(5), 485-491. doi:10.1002/ana.21541

Burke, W. J. (2003). 3,4-dihydroxyphenylacetaldehyde: a potential target for neuroprotective

therapy in Parkinson's disease. Curr Drug Targets CNS Neurol Disord, 2(2), 143-148.

Burkly, L. C., et al. (2011). TWEAK/Fn14 pathway: an immunological switch for shaping tissue

responses. Immunol Rev, 244(1), 99-114. doi:10.1111/j.1600-065X.2011.01054.x

Button, R. W., et al. (2017). Accumulation of autophagosomes confers cytotoxicity. J Biol

Chem, 292(33), 13599-13614. doi:10.1074/jbc.M117.782276

Cabezas, R., et al. (2014). Astrocytic modulation of blood brain barrier: perspectives on

Parkinson's disease. Front Cell Neurosci, 8, 211. doi:10.3389/fncel.2014.00211

Cai, P., et al. (2016). Inhibition of Endoplasmic Reticulum Stress is Involved in the

Neuroprotective Effect of bFGF in the 6-OHDA-Induced Parkinson’s Disease Model

(Vol. 7).

Cai, S., et al. (2012). NLRC4 inflammasome-mediated production of IL-1beta modulates

mucosal immunity in the lung against gram-negative bacterial infection. J Immunol,

188(11), 5623-5635. doi:10.4049/jimmunol.1200195

Cai, Y., et al. (2016). Interplay of endoplasmic reticulum stress and autophagy in

neurodegenerative disorders. Autophagy, 12(2), 225-244.

doi:10.1080/15548627.2015.1121360

Calabresi, P., et al. (2014). Direct and indirect pathways of basal ganglia: a critical reappraisal.

Nat Neurosci, 17(8), 1022-1030. doi:10.1038/nn.3743

Calì, T., et al. (2011). Mitochondria, calcium, and endoplasmic reticulum stress in Parkinson's

disease (Vol. 37).

Canet-Avilés, R. M., et al. (2004). The Parkinson's disease protein DJ-1 is neuroprotective due to

cysteine-sulfinic acid-driven mitochondrial localization. Proceedings of the National

Academy of Sciences of the United States of America, 101(24), 9103-9108.

doi:10.1073/pnas.0402959101

83

Cao, S. S., et al. (2014). Endoplasmic reticulum stress and oxidative stress in cell fate decision

and human disease. Antioxid Redox Signal, 21(3), 396-413. doi:10.1089/ars.2014.5851

Carvalho, A., et al. (2012). Ubiquitin–Proteasome System Impairment and MPTP-Induced

Oxidative Stress in the Brain of C57BL/6 Wild-type and GSTP Knockout Mice (Vol. 47).

Castaño, A., et al. (1998). Lipopolysaccharide Intranigral Injection Induces Inflammatory

Reaction and Damage in Nigrostriatal Dopaminergic System (Vol. 70).

Cha, H. S., et al. (2006). A novel spleen tyrosine kinase inhibitor blocks c-Jun N-terminal

kinase-mediated gene expression in synoviocytes. J Pharmacol Exp Ther, 317(2), 571-

578. doi:10.1124/jpet.105.097436

Chandra, R., et al. (2017). α-Synuclein in gut endocrine cells and its implications for Parkinson’s

disease. JCI Insight, 2(12), e92295. doi:10.1172/jci.insight.92295

Chang, W., et al. (2013). Pyroptosis: An inflammatory cell death implicates in atherosclerosis.

Medical Hypotheses, 81(3), 484-486. doi:https://doi.org/10.1016/j.mehy.2013.06.016

Chang, Y. P., et al. (2015). Resveratrol inhibits NLRP3 inflammasome activation by preserving

mitochondrial integrity and augmenting autophagy. J Cell Physiol, 230(7), 1567-1579.

doi:10.1002/jcp.24903

Chapman, M. S., et al. (2013). TWEAK signals through JAK-STAT to induce tumor cell

apoptosis. Cytokine, 61(1), 210-217. doi:10.1016/j.cyto.2012.09.020

Chaudhari, N., et al. (2014). A molecular web: endoplasmic reticulum stress, inflammation, and

oxidative stress. Front Cell Neurosci, 8, 213. doi:10.3389/fncel.2014.00213

Chen, L., et al. (2009). Tyrosine and serine phosphorylation of alpha-synuclein have opposing

effects on neurotoxicity and soluble oligomer formation. J Clin Invest, 119(11), 3257-

3265. doi:10.1172/jci39088

Chen, P. C., et al. (2009). Nrf2-mediated neuroprotection in the MPTP mouse model of

Parkinson's disease: Critical role for the astrocyte. Proc Natl Acad Sci U S A, 106(8),

2933-2938. doi:10.1073/pnas.0813361106

84

Cheng, S., et al. (2011). SYK inhibition and response prediction in diffuse large B-cell

lymphoma. Blood, 118(24), 6342-6352. doi:10.1182/blood-2011-02-333773

Chesselet, M. F. (2008). In vivo alpha-synuclein overexpression in rodents: a useful model of

Parkinson's disease? Exp Neurol, 209(1), 22-27. doi:10.1016/j.expneurol.2007.08.006

Cheung, E. C., et al. (2005). Apoptosis-inducing factor is a key factor in neuronal cell death

propagated by BAX-dependent and BAX-independent mechanisms. J Neurosci, 25(6),

1324-1334. doi:10.1523/jneurosci.4261-04.2005

Cheung, Z. H., et al. (2009). The emerging role of autophagy in Parkinson's disease. Molecular

Brain, 2(1), 29. doi:10.1186/1756-6606-2-29

Chicheportiche, Y., et al. (1997). TWEAK, a new secreted ligand in the tumor necrosis factor

family that weakly induces apoptosis. J Biol Chem, 272(51), 32401-32410.

Choi, D.-Y., et al. (2009). Striatal Neuroinflammation Promotes Parkinsonism in Rats (Vol. 4).

Choi, D. H., et al. (2011). Role of matrix metalloproteinase 3-mediated alpha-synuclein cleavage

in dopaminergic cell death. J Biol Chem, 286(16), 14168-14177.

doi:10.1074/jbc.M111.222430

Choi, S. H., et al. (2009). Lipoprotein accumulation in macrophages via toll-like receptor-4-

dependent fluid phase uptake. Circ Res, 104(12), 1355-1363.

doi:10.1161/circresaha.108.192880

Choi, S. H., et al. (2012). Spleen tyrosine kinase regulates AP-1 dependent transcriptional

response to minimally oxidized LDL. PLOS ONE, 7(2), e32378.

doi:10.1371/journal.pone.0032378

Chong, W., et al. (2017). alpha-Synuclein Enhances Cadmium Uptake and Neurotoxicity via

Oxidative Stress and Caspase Activated Cell Death Mechanisms in a Dopaminergic Cell

Model of Parkinson's Disease. Neurotox Res, 32(2), 231-246. doi:10.1007/s12640-017-

9725-x

Chu, C. T., et al. (2007). Beclin 1-independent pathway of damage-induced mitophagy and

autophagic stress: implications for neurodegeneration and cell death. Autophagy, 3(6),

663-666.

85

Chu, C. T., et al. (2005). Apoptosis inducing factor mediates caspase-independent 1-methyl-4-

phenylpyridinium toxicity in dopaminergic cells. J Neurochem, 94(6), 1685-1695.

doi:10.1111/j.1471-4159.2005.03329.x

Chu, Y., et al. (2009). Alterations in lysosomal and proteasomal markers in Parkinson's disease:

relationship to alpha-synuclein inclusions. Neurobiol Dis, 35(3), 385-398.

doi:10.1016/j.nbd.2009.05.023

Chung, C. Y., et al. (2009). Dynamic changes in presynaptic and axonal transport proteins

combined with striatal neuroinflammation precede dopaminergic neuronal loss in a rat

model of AAV alpha-synucleinopathy. J Neurosci, 29(11), 3365-3373.

doi:10.1523/jneurosci.5427-08.2009

Codolo, G., et al. (2013). Triggering of inflammasome by aggregated alpha-synuclein, an

inflammatory response in synucleinopathies. PLOS ONE, 8(1), e55375.

doi:10.1371/journal.pone.0055375

Colla, E., et al. (2012a). Endoplasmic Reticulum Stress Is Important for the Manifestations of α-

Synucleinopathy <em>In Vivo</em>. The Journal of Neuroscience, 32(10), 3306-3320.

doi:10.1523/jneurosci.5367-11.2012

Colla, E., et al. (2012b). Endoplasmic reticulum stress is important for the manifestations of α-

synucleinopathy in vivo. J Neurosci, 32(10), 3306-3320. doi:10.1523/JNEUROSCI.5367-

11.2012

Conway, K. A., et al. (2001). Kinetic stabilization of the alpha-synuclein protofibril by a

dopamine-alpha-synuclein adduct. Science, 294(5545), 1346-1349.

doi:10.1126/science.1063522

Cookson, M. R. (2009). α-Synuclein and neuronal cell death. Molecular Neurodegeneration, 4,

9-9. doi:10.1186/1750-1326-4-9

Cookson, M. R. (2012). Parkinsonism due to mutations in PINK1, parkin, and DJ-1 and

oxidative stress and mitochondrial pathways. Cold Spring Harb Perspect Med, 2(9),

a009415. doi:10.1101/cshperspect.a009415

Cooper, A. A., et al. (2006). Alpha-synuclein blocks ER-Golgi traffic and Rab1 rescues neuron

loss in Parkinson's models. Science, 313(5785), 324-328. doi:10.1126/science.1129462

86

Credle, J. J., et al. (2015). alpha-Synuclein-mediated inhibition of ATF6 processing into COPII

vesicles disrupts UPR signaling in Parkinson's disease. Neurobiol Dis, 76, 112-125.

doi:10.1016/j.nbd.2015.02.005

Cregan, S. P., et al. (2002). Apoptosis-inducing factor is involved in the regulation of caspase-

independent neuronal cell death. J Cell Biol, 158(3), 507-517.

doi:10.1083/jcb.200202130

Crocker, S. J., et al. (2003). Inhibition of calpains prevents neuronal and behavioral deficits in an

MPTP mouse model of Parkinson's disease. Journal of Neuroscience, 23(10), 4081-4091.

Cruz, C. M., et al. (2007a). ATP Activates a Reactive Oxygen Species-dependent Oxidative

Stress Response and Secretion of Proinflammatory Cytokines in Macrophages. J Biol

Chem, 282(5), 2871-2879. doi:10.1074/jbc.M608083200

Cruz, C. M., et al. (2007b). ATP Activates a Reactive Oxygen Species-dependent Oxidative

Stress Response and Secretion of Proinflammatory Cytokines in Macrophages. Journal of

Biological Chemistry, 282(5), 2871-2879. doi:10.1074/jbc.M608083200

Cuervo, A. M., et al. (2004). Impaired degradation of mutant alpha-synuclein by chaperone-

mediated autophagy. Science, 305(5688), 1292-1295. doi:10.1126/science.1101738

Cullinan, S. B., et al. (2004). PERK-dependent Activation of Nrf2 Contributes to Redox

Homeostasis and Cell Survival following Endoplasmic Reticulum Stress. Journal of

Biological Chemistry, 279(19), 20108-20117. doi:10.1074/jbc.M314219200

Dadakhujaev, S., et al. (2010). Autophagy protects the rotenone-induced cell death in alpha-

synuclein overexpressing SH-SY5Y cells. Neurosci Lett, 472(1), 47-52.

doi:10.1016/j.neulet.2010.01.053

Dasuri, K., et al. (2013). Oxidative stress, neurodegeneration, and the balance of protein

degradation and protein synthesis. Free Radical Biology and Medicine, 62, 170-185.

doi:https://doi.org/10.1016/j.freeradbiomed.2012.09.016

Davie, C. A. (2008). A review of Parkinson's disease. Br Med Bull, 86, 109-127.

doi:10.1093/bmb/ldn013

87

De Jesús-Cortés, H., et al. (2015). Protective efficacy of P7C3-S243 in the 6-hydroxydopamine

model of Parkinson&#39;s disease. Npj Parkinson&#39;S Disease, 1, 15010.

doi:10.1038/npjparkd.2015.10

https://www.nature.com/articles/npjparkd201510#supplementary-information

de Lau, L. M., et al. (2006). Epidemiology of Parkinson's disease. Lancet Neurol, 5(6), 525-535.

doi:10.1016/s1474-4422(06)70471-9

Del Tredici, K., et al. (2002). Where does parkinson disease pathology begin in the brain? J

Neuropathol Exp Neurol, 61(5), 413-426.

DelleDonne, A., et al. (2008). Incidental Lewy body disease and preclinical Parkinson disease.

Arch Neurol, 65(8), 1074-1080. doi:10.1001/archneur.65.8.1074

DeLong, M. R., et al. (2015). Basal Ganglia Circuits as Targets for Neuromodulation in

Parkinson Disease. JAMA Neurol, 72(11), 1354-1360. doi:10.1001/jamaneurol.2015.2397

Denes, A., et al. (2015). AIM2 and NLRC4 inflammasomes contribute with ASC to acute brain

injury independently of NLRP3. Proceedings of the National Academy of Sciences,

112(13), 4050-4055. doi:10.1073/pnas.1419090112

Deng, G.-M., et al. (2016a). Targeting Syk in Autoimmune Rheumatic Diseases. Frontiers in

Immunology, 7, 78. doi:10.3389/fimmu.2016.00078

Deng, G.-M., et al. (2016b). Targeting Syk in Autoimmune Rheumatic Diseases. Frontiers in

Immunology, 7(78). doi:10.3389/fimmu.2016.00078

Desplat-Jego, S., et al. (2005). Anti-TWEAK monoclonal antibodies reduce immune cell

infiltration in the central nervous system and severity of experimental autoimmune

encephalomyelitis. Clin Immunol, 117(1), 15-23. doi:10.1016/j.clim.2005.06.005

Desplat-Jego, S., et al. (2009). TWEAK is expressed at the cell surface of monocytes during

multiple sclerosis. J Leukoc Biol, 85(1), 132-135. doi:10.1189/jlb.0608347

Desplats, P., et al. (2009). Inclusion formation and neuronal cell death through neuron-to-neuron

transmission of alpha-synuclein. Proc Natl Acad Sci U S A, 106(31), 13010-13015.

doi:10.1073/pnas.0903691106

88

Deumens, R., et al. (2002). Modeling Parkinson's disease in rats: an evaluation of 6-OHDA

lesions of the nigrostriatal pathway. Exp Neurol, 175(2), 303-317.

doi:10.1006/exnr.2002.7891

Devi, L., et al. (2008). Mitochondrial import and accumulation of alpha-synuclein impair

complex I in human dopaminergic neuronal cultures and Parkinson disease brain. J Biol

Chem, 283(14), 9089-9100. doi:10.1074/jbc.M710012200

Dheen, S. T., et al. (2007). Microglial activation and its implications in the brain diseases. Curr

Med Chem, 14(11), 1189-1197.

Dhruv, H. D., et al. (2014). Tumor necrosis factor-like weak inducer of apoptosis (TWEAK)

promotes glioblastoma cell chemotaxis via Lyn activation. Carcinogenesis, 35(1), 218-

226. doi:10.1093/carcin/bgt289

Di Domenico, F., et al. (2014). Oxidative Stress and Proteostasis Network: Culprit and Casualty

of Alzheimer's-Like Neurodegeneration (Vol. 2014).

Diamond, C. E., et al. (2015). Novel perspectives on non-canonical inflammasome activation.

Immunotargets and Therapy, 4, 131-141. doi:10.2147/ITT.S57976

Dias, V., et al. (2013). The role of oxidative stress in Parkinson's disease. J Parkinsons Dis, 3(4),

461-491. doi:10.3233/jpd-130230

Dickson, D. W. (2012). Parkinson’s Disease and Parkinsonism: Neuropathology. Cold Spring

Harbor Perspectives in Medicine, 2(8), a009258. doi:10.1101/cshperspect.a009258

Dickson, D. W., et al. (2010). Evidence in favor of Braak staging of Parkinson's disease. Mov

Disord, 25 Suppl 1, S78-82. doi:10.1002/mds.22637

Ding, J., et al. (2017). SnapShot: The Noncanonical Inflammasome. Cell, 168(3), 544-544.e541.

doi:10.1016/j.cell.2017.01.008

Dogra, C., et al. (2006). Tumor necrosis factor-like weak inducer of apoptosis inhibits skeletal

myogenesis through sustained activation of nuclear factor-kappaB and degradation of

MyoD protein. J Biol Chem, 281(15), 10327-10336. doi:10.1074/jbc.M511131200

89

Dohi, T., et al. (2012). The TWEAK/Fn14 pathway as an aggravating and perpetuating factor in

inflammatory diseases: Focus on inflammatory bowel diseases (Vol. 92).

Donohue, P. J., et al. (2003). TWEAK is an endothelial cell growth and chemotactic factor that

also potentiates FGF-2 and VEGF-A mitogenic activity. Arterioscler Thromb Vasc Biol,

23(4), 594-600. doi:10.1161/01.atv.0000062883.93715.37

Dooneief, G., et al. (1992). An estimate of the incidence of depression in idiopathic Parkinson's

disease. Arch Neurol, 49(3), 305-307.

Dostert, C., et al. (2008a). Innate Immune Activation Through Nalp3 Inflammasome Sensing of

Asbestos and Silica (Vol. 320).

Dostert, C., et al. (2008b). Innate Immune Activation Through Nalp3 Inflammasome Sensing of

Asbestos and Silica. Science, 320(5876), 674-677. doi:10.1126/science.1156995

Douglas, D. L., et al. (2014). PARP1-mediated necrosis is dependent on parallel JNK and

Ca(2)(+)/calpain pathways. J Cell Sci, 127(Pt 19), 4134-4145. doi:10.1242/jcs.128009

Duda, J. E., et al. (2000). Neuropathology of synuclein aggregates. J Neurosci Res, 61(2), 121-

127. doi:10.1002/1097-4547(20000715)61:2<121::aid-jnr1>3.0.co;2-4

Duncan, J. A., et al. (2018). The NLRC4 Inflammasome. Immunological Reviews, 281(1), 115-

123. doi:10.1111/imr.12607

DuRose, J. B., et al. (2006). Intrinsic capacities of molecular sensors of the unfolded protein

response to sense alternate forms of endoplasmic reticulum stress. Mol Biol Cell, 17(7),

3095-3107. doi:10.1091/mbc.E06-01-0055

Echeverry, R., et al. (2012). The cytokine tumor necrosis factor-like weak inducer of apoptosis

and its receptor fibroblast growth factor-inducible 14 have a neuroprotective effect in the

central nervous system. J Neuroinflammation, 9, 45. doi:10.1186/1742-2094-9-45

Egawa, N., et al. (2010). The Endoplasmic Reticulum Stress Sensor, ATF6 , Protects against

Neurotoxin-induced Dopaminergic Neuronal Death (Vol. 286).

Eisenhofer, G., et al. (1999). Dopamine sulphate: an enigma resolved. Clin Exp Pharmacol

Physiol Suppl, 26, S41-53.

90

Elbaz, A., et al. (2016). Epidemiology of Parkinson's disease. Rev Neurol (Paris), 172(1), 14-26.

doi:10.1016/j.neurol.2015.09.012

Elliott, E., et al. (2015). Initiation and perpetuation of NLRP3 inflammasome activation and

assembly (Vol. 265).

Elliott, E. I., et al. (2015). Initiation and perpetuation of NLRP3 inflammasome activation and

assembly. Immunol Rev, 265(1), 35-52. doi:10.1111/imr.12286

Endale, M., et al. (2013). Quercetin disrupts tyrosine-phosphorylated phosphatidylinositol 3-

kinase and myeloid differentiation factor-88 association, and inhibits MAPK/AP-1 and

IKK/NF-κB-induced inflammatory mediators production in RAW 264.7 cells.

Immunobiology, 218(12), 1452-1467. doi:https://doi.org/10.1016/j.imbio.2013.04.019

Eriksen, J., et al. (2010). Regulation of dopamine transporter function by protein-protein

interactions: new discoveries and methodological challenges. J Neurochem, 113(1), 27-

41. doi:10.1111/j.1471-4159.2010.06599.x

Eriksen, J. L., et al. (2005a). Molecular pathogenesis of Parkinson disease. Arch Neurol, 62(3),

353-357. doi:10.1001/archneur.62.3.353

Eriksen, J. L., et al. (2005b). Molecular pathogenesis of parkinson disease. Archives of

Neurology, 62(3), 353-357. doi:10.1001/archneur.62.3.353

Ermak, G., et al. (2002). Calcium and oxidative stress: from cell signaling to cell death.

Molecular Immunology, 38(10), 713-721. doi:https://doi.org/10.1016/S0161-

5890(01)00108-0

Ermak, G., et al. (2002). Calcium and oxidative stress: from cell signaling to cell death (Vol.

38).

Eskelinen, E. L., et al. (2009). Autophagy: a lysosomal degradation pathway with a central role

in health and disease. Biochim Biophys Acta, 1793(4), 664-673.

doi:10.1016/j.bbamcr.2008.07.014

Exner, N., et al. (2007). Loss-of-function of human PINK1 results in mitochondrial pathology

and can be rescued by parkin. J Neurosci, 27(45), 12413-12418.

doi:10.1523/jneurosci.0719-07.2007

91

Farrer, M., et al. (2001). Lewy bodies and parkinsonism in families with parkin mutations. Ann

Neurol, 50(3), 293-300.

Feldman, A. L., et al. (2008). Clonally related follicular lymphomas and histiocytic/dendritic cell

sarcomas: evidence for transdifferentiation of the follicular lymphoma clone. Blood,

111(12), 5433-5439. doi:10.1182/blood-2007-11-124792

Fernandes, Hugo J., et al. (2016). ER Stress and Autophagic Perturbations Lead to Elevated

Extracellular α-Synuclein in GBA-N370S Parkinson's iPSC-Derived Dopamine Neurons.

Stem Cell Reports, 6(3), 342-356. doi:10.1016/j.stemcr.2016.01.013

Fernandes, Hugo J. R., et al. ER Stress and Autophagic Perturbations Lead to Elevated

Extracellular &#x3b1;-Synuclein in <em>GBA-N370S</em> Parkinson's iPSC-Derived

Dopamine Neurons. Stem Cell Reports, 6(3), 342-356. doi:10.1016/j.stemcr.2016.01.013

Ferrari, C. C., et al. (2006). Progressive neurodegeneration and motor disabilities induced by

chronic expression of IL-1β in the substantia nigra. Neurobiology of Disease, 24(1), 183-

193. doi:https://doi.org/10.1016/j.nbd.2006.06.013

Ferrer, I. (2011). Neuropathology and Neurochemistry of Nonmotor Symptoms in Parkinson's

Disease. Parkinson's Disease, 2011, 708404. doi:10.4061/2011/708404

Fesik, S. W. (2000). Insights into programmed cell death through structural biology. Cell,

103(2), 273-282.

Flight, M. H. (2012). High hopes for oral SYK inhibitor in rheumatoid arthritis. Nature Reviews

Drug Discovery, 11, 10. doi:10.1038/nrd3631

Fortin Ensign, S. P., et al. (2013). The Src Homology 3 Domain-containing Guanine Nucleotide

Exchange Factor Is Overexpressed in High-grade Gliomas and Promotes Tumor Necrosis

Factor-like Weak Inducer of Apoptosis-Fibroblast Growth Factor-inducible 14-induced

Cell Migration and Invasion via Tumor Necrosis Factor Receptor-associated Factor 2.

The Journal of Biological Chemistry, 288(30), 21887-21897.

doi:10.1074/jbc.M113.468686

Franchi, L., et al. (2006). Cytosolic flagellin requires Ipaf for activation of caspase-1 and

interleukin 1β in salmonella-infected macrophages. Nat Immunol, 7, 576.

doi:10.1038/ni1346

https://www.nature.com/articles/ni1346#supplementary-information

92

Franchi, L., et al. (2012). NLRC4-driven production of IL-1beta discriminates between

pathogenic and commensal bacteria and promotes host intestinal defense. Nat Immunol,

13(5), 449-456. doi:10.1038/ni.2263

Frank, C., et al. (2006). Approach to diagnosis of Parkinson disease. Canadian Family

Physician, 52(7), 862-868.

Freeman, L., et al. (2015). The pathogenic role of the inflammasome in neurodegenerative

diseases (Vol. 136).

Fuertes, G., et al. (2003). Role of proteasomes in the degradation of short-lived proteins in

human fibroblasts under various growth conditions. Int J Biochem Cell Biol, 35(5), 651-

664.

Fujiwara, H., et al. (2002). alpha-Synuclein is phosphorylated in synucleinopathy lesions. Nat

Cell Biol, 4(2), 160-164. doi:10.1038/ncb748

Fuller, J. P., et al. (2014). New roles for Fc receptors in neurodegeneration-the impact on

Immunotherapy for Alzheimer's Disease. Frontiers in Neuroscience, 8, 235.

doi:10.3389/fnins.2014.00235

Futterer, K., et al. (1998). Structural basis for Syk tyrosine kinase ubiquity in signal transduction

pathways revealed by the crystal structure of its regulatory SH2 domains bound to a

dually phosphorylated ITAM peptide. J Mol Biol, 281(3), 523-537.

doi:10.1006/jmbi.1998.1964

Gainetdinov, R. R., et al. (1997). Dopamine transporter is required for in vivo MPTP

neurotoxicity: evidence from mice lacking the transporter. J Neurochem, 69(3), 1322-

1325.

Galvan, L., et al. (2012). Functional Differences Between Direct and Indirect Striatal Output

Pathways in Huntington's Disease. J Huntingtons Dis, 1(1), 17-25. doi:10.3233/jhd-2012-

120009

Gao, H.-M., et al. (2003). Synergistic Dopaminergic Neurotoxicity of the Pesticide Rotenone and

Inflammogen Lipopolysaccharide: Relevance to the Etiology of Parkinson's Disease. The

Journal of Neuroscience, 23(4), 1228-1236.

93

Geahlen, R. L. (2009). Syk and pTyr'd: Signaling through the B cell antigen receptor. Biochim

Biophys Acta, 1793(7), 1115-1127. doi:10.1016/j.bbamcr.2009.03.004

Geahlen, R. L. (2014). Getting Syk: spleen tyrosine kinase as a therapeutic target. Trends

Pharmacol Sci, 35(8), 414-422. doi:10.1016/j.tips.2014.05.007

George, J., et al. (2015). Different danger signals differently impact on microglial proliferation

through alterations of ATP release and extracellular metabolism: ATP Metabolism Tunes

Microglia Proliferation (Vol. 63).

Ghosh, A., et al. (2012). Anti-inflammatory and neuroprotective effects of an orally active

apocynin derivative in pre-clinical models of Parkinson’s disease (Vol. 9).

Ghosh, S., et al. (2015). Stress Granules Modulate SYK to Cause Microglial Cell Dysfunction in

Alzheimer's Disease. EBioMedicine, 2(11), 1785-1798. doi:10.1016/j.ebiom.2015.09.053

Ghotra, V. P., et al. (2015). SYK is a candidate kinase target for the treatment of advanced

prostate cancer. Cancer Res, 75(1), 230-240. doi:10.1158/0008-5472.can-14-0629

Gibb, W. R., et al. (1988). The relevance of the Lewy body to the pathogenesis of idiopathic

Parkinson's disease. J Neurol Neurosurg Psychiatry, 51(6), 745-752.

Girgenrath, M., et al. (2006). TWEAK, via its receptor Fn14, is a novel regulator of

mesenchymal progenitor cells and skeletal muscle regeneration. Embo j, 25(24), 5826-

5839. doi:10.1038/sj.emboj.7601441

Goldberg, M. S., et al. (2003). Parkin-deficient mice exhibit nigrostriatal deficits but not loss of

dopaminergic neurons. J Biol Chem, 278(44), 43628-43635.

doi:10.1074/jbc.M308947200

Gomez-Suaga, P., et al. (2012). Leucine-rich repeat kinase 2 regulates autophagy through a

calcium-dependent pathway involving NAADP. Hum Mol Genet, 21(3), 511-525.

doi:10.1093/hmg/ddr481

Gorbatyuk, M., et al. (2012). Glucose Regulated Protein 78 Diminishes -Synuclein Neurotoxicity

in a Rat Model of Parkinson Disease (Vol. 20).

94

Gosavi, N., et al. (2002). Golgi fragmentation occurs in the cells with prefibrillar alpha-synuclein

aggregates and precedes the formation of fibrillar inclusion. J Biol Chem, 277(50),

48984-48992. doi:10.1074/jbc.M208194200

Greenberg, S. (1999). Modular components of phagocytosis. J Leukoc Biol, 66(5), 712-717.

Greene, A. W., et al. (2012). Mitochondrial processing peptidase regulates PINK1 processing,

import and Parkin recruitment. EMBO Rep, 13(4), 378-385. doi:10.1038/embor.2012.14

Groenewegen, H. J. (2003). The basal ganglia and motor control. Neural Plast, 10(1-2), 107-120.

doi:10.1155/np.2003.107

Gross, O., et al. (2009). Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal

host defence. Nature, 459(7245), 433-436. doi:10.1038/nature07965

Grywalska, E., et al. (2015). Epstein-Barr Virus–Associated Lymphomas. Seminars in Oncology,

42(2), 291-303. doi:https://doi.org/10.1053/j.seminoncol.2014.12.030

Gu, X. L., et al. (2010). Astrocytic expression of Parkinson's disease-related A53T alpha-

synuclein causes neurodegeneration in mice. Mol Brain, 3, 12. doi:10.1186/1756-6606-3-

12

Guo, H., et al. (2015). Inflammasomes: mechanism of action, role in disease, and therapeutics.

Nature Medicine, 21, 677. doi:10.1038/nm.3893

Hafner-Bratkovic, I. (2017). The NLRC4 inflammasome: The pieces of the puzzle are falling into

place (Vol. 3).

Haile, W. B., et al. (2010). Tumor necrosis factor-like weak inducer of apoptosis and fibroblast

growth factor-inducible 14 mediate cerebral ischemia-induced poly(ADP-ribose)

polymerase-1 activation and neuronal death. Neuroscience, 171(4), 1256-1264.

doi:10.1016/j.neuroscience.2010.10.029

Haile, W. B., et al. (2010). The interaction between tumor necrosis factor-like weak inducer of

apoptosis and its receptor fibroblast growth factor-inducible 14 promotes the recruitment

of neutrophils into the ischemic brain. Journal of Cerebral Blood Flow and Metabolism:

Official Journal of the International Society of Cerebral Blood Flow and Metabolism,

30(6), 1147-1156. doi:10.1038/jcbfm.2009.280

95

Halle, A., et al. (2008). The NALP3 inflammasome is involved in the innate immune response to

amyloid-beta. Nat Immunol, 9(8), 857-865. doi:10.1038/ni.1636

Halliday, G., et al. (2012). Evaluation of the Braak hypothesis: how far can it explain the

pathogenesis of Parkinson's disease? Expert Rev Neurother, 12(6), 673-686.

doi:10.1586/ern.12.47

Halliday, G. M., et al. (2010). The progression of pathology in Parkinson's disease. Ann N Y

Acad Sci, 1184, 188-195. doi:10.1111/j.1749-6632.2009.05118.x

Hanrott, K., et al. (2006). 6-hydroxydopamine-induced apoptosis is mediated via extracellular

auto-oxidation and caspase 3-dependent activation of protein kinase Cdelta. J Biol Chem,

281(9), 5373-5382. doi:10.1074/jbc.M511560200

Hansen, T. E., et al. (2011). Following autophagy step by step. BMC Biol, 9, 39.

doi:10.1186/1741-7007-9-39

Hao, L.-Y., et al. (2010). DJ-1 is critical for mitochondrial function and rescues PINK1 loss of

function. Proceedings of the National Academy of Sciences of the United States of

America, 107(21), 9747-9752. doi:10.1073/pnas.0911175107

Haobam, R., et al. (2005). Swim-test as a function of motor impairment in MPTP model of

Parkinson's disease: A comparative study in two mouse strains. Behavioural Brain

Research, 163(2), 159-167. doi:https://doi.org/10.1016/j.bbr.2005.04.011

Hara, H., et al. (2013). Phosphorylation of the adaptor ASC acts as a molecular switch that

controls the formation of speck-like aggregates and inflammasome activity. Nat Immunol,

14(12), 1247-1255. doi:10.1038/ni.2749

Hardy, J., et al. (2003). Genes and parkinsonism. Lancet Neurol, 2(4), 221-228.

Harris, J., et al. (2017). Autophagy and inflammasomes. Molecular Immunology, 86, 10-15.

doi:https://doi.org/10.1016/j.molimm.2017.02.013

Hartmann, A., et al. (2000). Caspase-3: A vulnerability factor and final effector in apoptotic

death of dopaminergic neurons in Parkinson's disease. Proc Natl Acad Sci U S A, 97(6),

2875-2880. doi:10.1073/pnas.040556597

96

Hartmann, A., et al. (2001). Is Bax a mitochondrial mediator in apoptotic death of dopaminergic

neurons in Parkinson's disease? J Neurochem, 76(6), 1785-1793.

Hartmann, A., et al. (2001). Caspase-8 is an effector in apoptotic death of dopaminergic neurons

in Parkinson's disease, but pathway inhibition results in neuronal necrosis. J Neurosci,

21(7), 2247-2255.

Hassouna, I., et al. (1996). Increase in bax expression in substantia nigra following 1-methyl-4-

phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment of mice. Neurosci Lett, 204(1-2),

85-88.

Hauser, D. N., et al. (2013). Mitochondrial dysfunction and oxidative stress in Parkinson’s

disease and monogenic parkinsonism. Neurobiology of disease, 51, 35-42.

doi:10.1016/j.nbd.2012.10.011

Hayashi, T., et al. (2009). DJ-1 binds to mitochondrial complex I and maintains its activity.

Biochem Biophys Res Commun, 390(3), 667-672. doi:10.1016/j.bbrc.2009.10.025

He, Q., et al. (2013). Intranasal LPS-Mediated Parkinson’s Model Challenges the Pathogenesis

of Nasal Cavity and Environmental Toxins. PLOS ONE, 8(11), e78418.

doi:10.1371/journal.pone.0078418

He, Y., et al. (2016). Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends

Biochem Sci, 41(12), 1012-1021. doi:10.1016/j.tibs.2016.09.002

He, Y., et al. Mechanism and Regulation of NLRP3 Inflammasome Activation. Trends in

Biochemical Sciences, 41(12), 1012-1021. doi:10.1016/j.tibs.2016.09.002

He, Y., et al. (2015). Mincle/Syk Signaling Pathway Contributes to Neuroinflammation after

Subarachnoid Hemorrhage in Rats. Stroke, 46(8), 2277-2286.

doi:10.1161/STROKEAHA.115.010088

Healy, D. G., et al. (2004). DJ-1 mutations in Parkinson's disease. J Neurol Neurosurg

Psychiatry, 75(1), 144-145.

Heid, M. E., et al. (2013). Mitochondrial Reactive Oxygen Species Induces NLRP3-Dependent

Lysosomal Damage and Inflammasome Activation. The Journal of Immunology, 191(10),

5230-5238. doi:10.4049/jimmunol.1301490

97

Henchcliffe, C., et al. (2008). Mitochondrial biology and oxidative stress in Parkinson disease

pathogenesis. Nat Clin Pract Neurol, 4(11), 600-609. doi:10.1038/ncpneuro0924

Herrera, A. J., et al. (2000). The single intranigral injection of LPS as a new model for studying

the selective effects of inflammatory reactions on dopaminergic system. Neurobiol Dis,

7(4), 429-447. doi:10.1006/nbdi.2000.0289

Herzog, S., et al. (2009). Regulation of B-cell proliferation and differentiation by pre-B-cell

receptor signalling. Nat Rev Immunol, 9(3), 195-205. doi:10.1038/nri2491

Hikida, T., et al. (2010). Distinct roles of synaptic transmission in direct and indirect striatal

pathways to reward and aversive behavior. Neuron, 66(6), 896-907.

doi:10.1016/j.neuron.2010.05.011

Hirose, M., et al. (2004). Phosphorylation and recruitment of Syk by immunoreceptor tyrosine-

based activation motif-based phosphorylation of tamalin. J Biol Chem, 279(31), 32308-

32315. doi:10.1074/jbc.M400547200

Hirsch, E. C., et al. (1993). Does Oxidative Stress Participate in Nerve Cell Death in Parkinson’s

Disease? European Neurology, 33(suppl 1)(Suppl. 1), 52-59.

Hirsch, E. C., et al. (2003). Animal models of Parkinson's disease in rodents induced by toxins:

an update. J Neural Transm Suppl(65), 89-100.

Hirsch, E. C., et al. (2012). Neuroinflammation in Parkinson's disease. Parkinsonism Relat

Disord, 18 Suppl 1, S210-212. doi:10.1016/s1353-8020(11)70065-7

Hoek, R. M., et al. (2000). Down-regulation of the macrophage lineage through interaction with

OX2 (CD200). Science, 290(5497), 1768-1771.

Holtz, W. A., et al. (2003). Parkinsonian Mimetics Induce Aspects of Unfolded Protein Response

in Death of Dopaminergic Neurons. Journal of Biological Chemistry, 278(21), 19367-

19377. doi:10.1074/jbc.M211821200

Homer, C. R., et al. (2012). A dual role for receptor-interacting protein kinase 2 (RIP2) kinase

activity in nucleotide-binding oligomerization domain 2 (NOD2)-dependent autophagy. J

Biol Chem, 287(30), 25565-25576. doi:10.1074/jbc.M111.326835

98

Hong, S. J., et al. (2004). Nuclear and mitochondrial conversations in cell death: PARP-1 and

AIF signaling. Trends Pharmacol Sci, 25(5), 259-264. doi:10.1016/j.tips.2004.03.005

Hoozemans, J. J. M., et al. (2007). Activation of the Unfolded Protein Response in Parkinson’s

Disease (Vol. 354).

Hornung, V., et al. (2009). AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating

inflammasome with ASC. Nature, 458(7237), 514-518. doi:10.1038/nature07725

Hoshino, K., et al. (1999). Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are

hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product. J

Immunol, 162(7), 3749-3752.

Hou, X. O., et al. (2015). Parkin represses 6-hydroxydopamine-induced apoptosis via stabilizing

scaffold protein p62 in PC12 cells. Acta Pharmacol Sin, 36(11), 1300-1307.

doi:10.1038/aps.2015.54

Huang, C., et al. (2011). Preconditioning involves selective mitophagy mediated by Parkin and

p62/SQSTM1. PLOS ONE, 6(6), e20975. doi:10.1371/journal.pone.0020975

Huysamen, C., et al. (2008). CLEC9A Is a Novel Activation C-type Lectin-like Receptor

Expressed on BDCA3(+) Dendritic Cells and a Subset of Monocytes. The Journal of

Biological Chemistry, 283(24), 16693-16701. doi:10.1074/jbc.M709923200

Hwang, O. (2013). Role of Oxidative Stress in Parkinson's Disease. Experimental Neurobiology,

22(1), 11-17. doi:10.5607/en.2013.22.1.11

Ibanez, P., et al. (2006). Mutational analysis of the PINK1 gene in early-onset parkinsonism in

Europe and North Africa. Brain, 129(Pt 3), 686-694. doi:10.1093/brain/awl005

Imai, Y., et al. (2001). An unfolded putative transmembrane polypeptide, which can lead to

endoplasmic reticulum stress, is a substrate of Parkin. Cell, 105(7), 891-902.

Inoue, J., et al. (2000). Tumor necrosis factor receptor-associated factor (TRAF) family: adapter

proteins that mediate cytokine signaling. Exp Cell Res, 254(1), 14-24.

doi:10.1006/excr.1999.4733

99

Inta, I., et al. (2008). Induction of the cytokine TWEAK and its receptor Fn14 in ischemic stroke.

J Neurol Sci, 275(1-2), 117-120. doi:10.1016/j.jns.2008.08.005

Iocca, H. A., et al. (2008). TNF superfamily member TWEAK exacerbates inflammation and

demyelination in the cuprizone-induced model. J Neuroimmunol, 194(1-2), 97-106.

doi:10.1016/j.jneuroim.2007.12.003

Itier, J. M., et al. (2003). Parkin gene inactivation alters behaviour and dopamine

neurotransmission in the mouse. Hum Mol Genet, 12(18), 2277-2291.

doi:10.1093/hmg/ddg239

Iwata, A., et al. (2005). HDAC6 and microtubules are required for autophagic degradation of

aggregated huntingtin. J Biol Chem, 280(48), 40282-40292.

doi:10.1074/jbc.M508786200

Jakubowski, A., et al. (2005). TWEAK induces liver progenitor cell proliferation. J Clin Invest,

115(9), 2330-2340. doi:10.1172/jci23486

Jankovic, J. (2008). Parkinson’s disease: clinical features and diagnosis. Journal of Neurology,

Neurosurgery &amp; Psychiatry, 79(4), 368-376. doi:10.1136/jnnp.2007.131045

Janowski, A. M., et al. (2016). NLRC4 suppresses melanoma tumor progression independently

of inflammasome activation. The Journal of Clinical Investigation, 126(10), 3917-3928.

doi:10.1172/JCI86953

Jaumouille, V., et al. (2014). Actin cytoskeleton reorganization by Syk regulates Fcgamma

receptor responsiveness by increasing its lateral mobility and clustering. Dev Cell, 29(5),

534-546. doi:10.1016/j.devcel.2014.04.031

JC, G. J. C., et al. (2016). (Vol. 627).

Jellinger, K. A. (2000). Cell death mechanisms in Parkinson's disease. J Neural Transm

(Vienna), 107(1), 1-29. doi:10.1007/s007020050001

Jellinger, K. A. (2009). A critical evaluation of current staging of α-synuclein pathology in Lewy

body disorders. Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease,

1792(7), 730-740. doi:https://doi.org/10.1016/j.bbadis.2008.07.006

100

Jellinger, K. A. (2010). Critical evaluation of the Braak staging scheme for Parkinson's disease.

Ann Neurol, 67(4), 550. doi:10.1002/ana.21638

Jenner, P. (2003). Oxidative stress in Parkinson's disease. Annals of Neurology, 53(S3), S26-S38.

doi:10.1002/ana.10483

Jenner, P., et al. (1992). Oxidative stress as a cause of nigral cell death in Parkinson's disease and

incidental Lewy body disease. The Royal Kings and Queens Parkinson's Disease

Research Group. Ann Neurol, 32 Suppl, S82-87.

Jenner, P., et al. (1998). Understanding cell death in parkinson's disease. Annals of Neurology,

44(S1), S72-S84. doi:10.1002/ana.410440712

Jeohn, G.-H., et al. (2002). Gö6976 Protects Mesencephalic Neurons from Lipopolysaccharide-

Elicited Death by Inhibiting p38 MAP Kinase Phosphorylation. Annals of the New York

Academy of Sciences, 962(1), 347-359. doi:10.1111/j.1749-6632.2002.tb04079.x

Jeong, D., et al. (2014). Anti-inflammatory activities and mechanisms of Artemisia asiatica

ethanol extract. J Ethnopharmacol, 152(3), 487-496. doi:10.1016/j.jep.2014.01.030

Jha, N., et al. (2014). Role of Oxidative Stress, ER Stress and Ubiquitin Proteasome System in

Neurodegeneration (Vol. 01).

Jiang, P., et al. (2010). ER stress response plays an important role in aggregation of alpha-

synuclein. Mol Neurodegener, 5, 56. doi:10.1186/1750-1326-5-56

Jin, L., et al. (2004). Induction of RANTES by TWEAK/Fn14 interaction in human

keratinocytes. J Invest Dermatol, 122(5), 1175-1179. doi:10.1111/j.0022-

202X.2004.22419.x

John Betteridge, D. (2000). Betteridge, D. J. What is oxidative stress? Metabolism 49, 3-8 (Vol.

49).

Junn, E., et al. (2009). Mitochondrial localization of DJ-1 leads to enhanced neuroprotection. J

Neurosci Res, 87(1), 123-129. doi:10.1002/jnr.21831

Kalaitzakis, M. E., et al. (2009). The morbid anatomy of dementia in Parkinson's disease. Acta

Neuropathol, 118(5), 587-598. doi:10.1007/s00401-009-0597-x

101

Kanneganti, T.-D., et al. (2006). Critical Role for Cryopyrin/Nalp3 in Activation of Caspase-1 in

Response to Viral Infection and Double-stranded RNA. Journal of Biological Chemistry,

281(48), 36560-36568. doi:10.1074/jbc.M607594200

Kasten, M., et al. (2007). Epidemiology of Parkinson's disease. Handb Clin Neurol, 83, 129-151.

doi:10.1016/s0072-9752(07)83006-5

Kaur, J., et al. (2015). Autophagy at the crossroads of catabolism and anabolism. Nat Rev Mol

Cell Biol, 16(8), 461-472. doi:10.1038/nrm4024

Kaushik, S., et al. (2011). Chaperone-mediated autophagy at a glance. Journal of Cell Science,

124(4), 495-499. doi:10.1242/jcs.073874

Kayagaki, N., et al. (2015). Caspase-11 cleaves gasdermin D for non-canonical inflammasome

signalling. Nature, 526(7575), 666-671. doi:10.1038/nature15541

Kiernan, E. A., et al. (2016). Mechanisms of microglial activation in models of inflammation and

hypoxia: Implications for chronic intermittent hypoxia. The Journal of Physiology,

594(6), 1563-1577. doi:10.1113/JP271502

Kim, M. J., et al. (2012). Spleen tyrosine kinase is important in the production of

proinflammatory cytokines and cell proliferation in human mesangial cells following

stimulation with IgA1 isolated from IgA nephropathy patients. J Immunol, 189(7), 3751-

3758. doi:10.4049/jimmunol.1102603

Kim, M. J., et al. (2016). Mitophagy: a balance regulator of NLRP3 inflammasome activation.

BMB Rep, 49(10), 529-535.

Kim, R. H., et al. (2005). Hypersensitivity of DJ-1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyrindine (MPTP) and oxidative stress. Proc Natl Acad Sci U S A, 102(14),

5215-5220. doi:10.1073/pnas.0501282102

Kim, S. H., et al. (2004). TWEAK can induce pro-inflammatory cytokines and matrix

metalloproteinase-9 in macrophages. Circ J, 68(4), 396-399.

Kim, S. H., et al. (2004). Importance of MAPK pathways for microglial pro-inflammatory

cytokine IL-1 beta production. Neurobiol Aging, 25(4), 431-439. doi:10.1016/s0197-

4580(03)00126-x

102

Kim, S. U., et al. (2005). Microglia in health and disease. J Neurosci Res, 81(3), 302-313.

doi:10.1002/jnr.20562

Kim, W. G., et al. (2000). Regional difference in susceptibility to lipopolysaccharide-induced

neurotoxicity in the rat brain: role of microglia. J Neurosci, 20(16), 6309-6316.

Kim, Y.-S., et al. (2007). A pivotal role of matrix metalloproteinase-3 activity in dopaminergic

neuronal degeneration via microglial activation (Vol. 21).

Kim, Y. S., et al. (2006). Microglia, major player in the brain inflammation: their roles in the

pathogenesis of Parkinson's disease. Exp Mol Med, 38(4), 333-347.

doi:10.1038/emm.2006.40

Kimura, K. (1997). Mechanisms of active oxygen species reduction by non-steroidal anti-

inflammatory drugs. Int J Biochem Cell Biol, 29(3), 437-446.

Kirik, D., et al. (2002). Parkinson-like neurodegeneration induced by targeted overexpression of

alpha-synuclein in the nigrostriatal system. J Neurosci, 22(7), 2780-2791. doi:20026246

Klein, C., et al. (2000). Parkin deletions in a family with adult-onset, tremor-dominant

parkinsonism: expanding the phenotype. Ann Neurol, 48(1), 65-71.

Klein, C., et al. (2012). Genetics of Parkinson’s Disease. Cold Spring Harbor Perspectives in

Medicine, 2(1), a008888. doi:10.1101/cshperspect.a008888

Kolb, R., et al. (2016). Obesity-associated NLRC4 inflammasome activation drives breast cancer

progression. Nature Communications, 7, 13007. doi:10.1038/ncomms13007

Koprich, J. B., et al. (2017). Animal models of alpha-synucleinopathy for Parkinson disease drug

development. Nat Rev Neurosci, 18(9), 515-529. doi:10.1038/nrn.2017.75

Korczyn, A. D. (2004). Drug treatment of Parkinson's disease. Dialogues in Clinical

Neuroscience, 6(3), 315-322.

Kortekaas, R., et al. (2005). Blood-brain barrier dysfunction in Parkinsonian midbrain in vivo

(Vol. 57).

103

Kostrzewa, R. M. (2000). Review of apoptosis vs. necrosis of substantia nigra pars compacta in

Parkinson's disease. Neurotox Res, 2(2-3), 239-250.

Kowal, S. L., et al. (2013). The current and projected economic burden of Parkinson's disease in

the United States. Mov Disord, 28(3), 311-318. doi:10.1002/mds.25292

Kraft, C., et al. (2010). Selective autophagy: ubiquitin-mediated recognition and beyond. Nat

Cell Biol, 12(9), 836-841. doi:10.1038/ncb0910-836

Kravitz, A. V., et al. (2012). Distinct roles for direct and indirect pathway striatal neurons in

reinforcement. Nature Neuroscience, 15, 816. doi:10.1038/nn.3100

https://www.nature.com/articles/nn.3100#supplementary-information

Krisenko, M. O., et al. (2015). Calling in SYK: SYK's dual role as a tumor promoter and tumor

suppressor in cancer. Biochim Biophys Acta, 1853(1), 254-263.

doi:10.1016/j.bbamcr.2014.10.022

Kroemer, G., et al. (2008). Autophagic cell death: the story of a misnomer. Nat Rev Mol Cell

Biol, 9(12), 1004-1010. doi:10.1038/nrm2529

Kulathu, Y., et al. (2009). Autoinhibition and adapter function of Syk. Immunol Rev, 232(1),

286-299. doi:10.1111/j.1600-065X.2009.00837.x

Kulich, S. M., et al. (2007). 6-Hydroxydopamine induces mitochondrial ERK activation. Free

Radic Biol Med, 43(3), 372-383. doi:10.1016/j.freeradbiomed.2007.04.028

Kumar, M., et al. (2009). TNF-like weak inducer of apoptosis (TWEAK) activates

proinflammatory signaling pathways and gene expression through the activation of TGF-

beta-activated kinase 1. J Immunol, 182(4), 2439-2448. doi:10.4049/jimmunol.0803357

Kuusisto, E., et al. (2003). Morphogenesis of Lewy bodies: dissimilar incorporation of alpha-

synuclein, ubiquitin, and p62. J Neuropathol Exp Neurol, 62(12), 1241-1253.

L Cassel, S., et al. (2008). The Nalp3 inflammasome is essential for the development of silicosis

(Vol. 105).

104

L Hunter, R., et al. (2009). Intrastriatal Lipopolysaccharide Injection Induces Parkinsonism in

C57/B6 Mice (Vol. 87).

Lacroix, S., et al. (1998). The bacterial endotoxin lipopolysaccharide has the ability to target the

brain in upregulating its membrane CD14 receptor within specific cellular populations.

Brain Pathol, 8(4), 625-640.

Lamkanfi, M., et al. (2010). Inflammasome-dependent release of the alarmin HMGB1 in

endotoxemia. J Immunol, 185(7), 4385-4392. doi:10.4049/jimmunol.1000803

Lapchak, P. H., et al. (2012). Inhibition of Syk activity by R788 in platelets prevents remote lung

tissue damage after mesenteric ischemia-reperfusion injury. Am J Physiol Gastrointest

Liver Physiol, 302(12), G1416-1422. doi:10.1152/ajpgi.00026.2012

Latz, E., et al. (2013). Activation and regulation of the inflammasomes. Nat Rev Immunol, 13(6),

397-411. doi:10.1038/nri3452

Laudisi, F., et al. (2014). Tyrosine kinases: the molecular switch for inflammasome activation.

Cellular and Molecular Immunology, 11(2), 129-131. doi:10.1038/cmi.2014.4

Lauwers, E., et al. (2003). Neuropathology and neurodegeneration in rodent brain induced by

lentiviral vector-mediated overexpression of alpha-synuclein. Brain Pathol, 13(3), 364-

372.

Lazarou, M., et al. (2012). Role of PINK1 binding to the TOM complex and alternate

intracellular membranes in recruitment and activation of the E3 ligase Parkin. Dev Cell,

22(2), 320-333. doi:10.1016/j.devcel.2011.12.014

Lebeaupin, C., et al. (2015). ER stress induces NLRP3 inflammasome activation and hepatocyte

death. Cell Death &Amp; Disease, 6, e1879. doi:10.1038/cddis.2015.248

https://www.nature.com/articles/cddis2015248#supplementary-information

Lebouvier, T., et al. (2011). Colonic neuropathology is independent of olfactory dysfunction in

Parkinson's disease. J Parkinsons Dis, 1(4), 389-394. doi:10.3233/jpd-2011-11061

Lebouvier, T., et al. (2008). The microtubule-associated protein tau is phosphorylated by Syk.

Biochim Biophys Acta, 1783(2), 188-192. doi:10.1016/j.bbamcr.2007.11.005

105

Lee, G. S., et al. (2012). The calcium-sensing receptor regulates the NLRP3 inflammasome

through Ca2+ and cAMP. Nature, 492(7427), 123-127. doi:10.1038/nature11588

Lee, H. J., et al. (2010). Direct transfer of alpha-synuclein from neuron to astroglia causes

inflammatory responses in synucleinopathies. J Biol Chem, 285(12), 9262-9272.

doi:10.1074/jbc.M109.081125

Lehmann, J. M., et al. (1997). Peroxisome proliferator-activated receptors alpha and gamma are

activated by indomethacin and other non-steroidal anti-inflammatory drugs. J Biol Chem,

272(6), 3406-3410.

Lei, Y., et al. (2012). The mitochondrial proteins NLRX1 and TUFM form a complex that

regulates type I interferon and autophagy. Immunity, 36(6), 933-946.

doi:10.1016/j.immuni.2012.03.025

Lerner, A. G., et al. (2012). IRE1alpha induces thioredoxin-interacting protein to activate the

NLRP3 inflammasome and promote programmed cell death under irremediable ER

stress. Cell Metab, 16(2), 250-264. doi:10.1016/j.cmet.2012.07.007

Levine, B., et al. (2011). Autophagy in immunity and inflammation. Nature, 469(7330), 323-

335. doi:10.1038/nature09782

Li, D. (2006). Selective degradation of the IkappaB kinase (IKK) by autophagy. Cell Res,

16(11), 855-856. doi:10.1038/sj.cr.7310110

Li, J.-Q., et al. (2014). The role of the LRRK2 gene in Parkinsonism. Molecular

Neurodegeneration, 9(1), 47. doi:10.1186/1750-1326-9-47

Li, Q., et al. (2017). Microglia and macrophages in brain homeostasis and disease. Nature

Reviews Immunology. doi:10.1038/nri.2017.125

Li, W. W., et al. (2012). Microautophagy: lesser-known self-eating. Cell Mol Life Sci, 69(7),

1125-1136. doi:10.1007/s00018-011-0865-5

Li, Y., et al. (2009). Mutant LRRK2(R1441G) BAC transgenic mice recapitulate cardinal

features of Parkinson's disease. Nat Neurosci, 12(7), 826-828. doi:10.1038/nn.2349

106

Liang, Y., et al. (2015). Rifampicin attenuates rotenone-induced inflammation via suppressing

NLRP3 inflammasome activation in microglia. Brain Res, 1622, 43-50.

doi:10.1016/j.brainres.2015.06.008

Lilienbaum, A. (2013). Relationship between the proteasomal system and autophagy.

International Journal of Biochemistry and Molecular Biology, 4(1), 1-26.

Lim, K.-L., et al. (2007). Role of the ubiquitin proteasome system in Parkinson's disease. BMC

Biochemistry, 8(Suppl 1), S13-S13. doi:10.1186/1471-2091-8-S1-S13

Lin, S., et al. (2003). Minocycline blocks 6-hydroxydopamine-induced neurotoxicity and free

radical production in rat cerebellar granule neurons. Life Sci, 72(14), 1635-1641.

Lin, Y. C., et al. (2015). Syk is involved in NLRP3 inflammasome-mediated caspase-1 activation

through adaptor ASC phosphorylation and enhanced oligomerization. J Leukoc Biol,

97(5), 825-835. doi:10.1189/jlb.3HI0814-371RR

Lindholm, D., et al. (2006). Lindholm D, Wootz H, Korhonen LER stress and neurodegenerative

diseases. Cell Death Differ 13:385-392 (Vol. 13).

Liu, D., et al. (2017). Syk inhibitors in clinical development for hematological malignancies.

Journal of Hematology & Oncology, 10, 145. doi:10.1186/s13045-017-0512-1

Liu, L., et al. (2013). From Blood to the Brain: Can Systemically Transplanted Mesenchymal

Stem Cells Cross the Blood-Brain Barrier? Stem Cells International, 2013, 7.

doi:10.1155/2013/435093

Liu, M., et al. (2011). Lipopolysaccharide Animal Models for Parkinson&#39;s Disease.

Parkinson&#39;s Disease, 2011. doi:10.4061/2011/327089

Liu, Z., et al. (2008). A Drosophila model for LRRK2-linked parkinsonism. Proceedings of the

National Academy of Sciences of the United States of America, 105(7), 2693-2698.

doi:10.1073/pnas.0708452105

Llop-Guevara, A., et al. (2015). Simultaneous inhibition of JAK and SYK kinases ameliorates

chronic and destructive arthritis in mice. Arthritis Res Ther, 17, 356. doi:10.1186/s13075-

015-0866-0

107

Lo Bianco, C., et al. (2002). alpha -Synucleinopathy and selective dopaminergic neuron loss in a

rat lentiviral-based model of Parkinson's disease. Proc Natl Acad Sci U S A, 99(16),

10813-10818. doi:10.1073/pnas.152339799

Lo Bianco, C., et al. (2004). Lentiviral vector delivery of parkin prevents dopaminergic

degeneration in an alpha-synuclein rat model of Parkinson's disease. Proc Natl Acad Sci

U S A, 101(50), 17510-17515. doi:10.1073/pnas.0405313101

Locksley, R. M., et al. (2001). The TNF and TNF receptor superfamilies: integrating mammalian

biology. Cell, 104(4), 487-501.

Lofrumento, D., et al. (2010). MPTP-Induced Neuroinflammation Increases the Expression of

Pro-Inflammatory Cytokines and Their Receptors in Mouse Brain (Vol. 18).

Loos, B., et al. (2014). Defining and measuring autophagosome flux-concept and reality.

Autophagy, 10(11), 2087-2096. doi:10.4161/15548627.2014.973338

Lorenz, G., et al. (2014). Canonical and non-canonical effects of the NLRP3 inflammasome in

kidney inflammation and fibrosis. Nephrology Dialysis Transplantation, 29(1), 41-48.

doi:10.1093/ndt/gft332

Lowell, C. A. (2011). Src-family and Syk kinases in activating and inhibitory pathways in innate

immune cells: signaling cross talk. Cold Spring Harb Perspect Biol, 3(3).

doi:10.1101/cshperspect.a002352

Lu, M., et al. (2014). Uncoupling protein 2 deficiency aggravates astrocytic endoplasmic

reticulum stress and nod-like receptor protein 3 inflammasome activation. Neurobiol

Aging, 35(2), 421-430. doi:10.1016/j.neurobiolaging.2013.08.015

Lucchinetti, C., et al. (2000). Heterogeneity of multiple sclerosis lesions: implications for the

pathogenesis of demyelination. Ann Neurol, 47(6), 707-717.

Lucking, C. B., et al. (2000). Association between early-onset Parkinson's disease and mutations

in the parkin gene. N Engl J Med, 342(21), 1560-1567.

doi:10.1056/nejm200005253422103

Luk, K., et al. (2012). Pathological -Synuclein Transmission Initiates Parkinson-like

Neurodegeneration in Nontransgenic Mice (Vol. 338).

108

Luk, K. C., et al. (2012). Pathological alpha-synuclein transmission initiates Parkinson-like

neurodegeneration in nontransgenic mice. Science, 338(6109), 949-953.

doi:10.1126/science.1227157

Lushchak, V. I. (2012). Glutathione Homeostasis and Functions: Potential Targets for Medical

Interventions. Journal of Amino Acids, 2012, 26. doi:10.1155/2012/736837

Lynch-Day, M. A., et al. (2012). The role of autophagy in Parkinson's disease. Cold Spring Harb

Perspect Med, 2(4), a009357. doi:10.1101/cshperspect.a009357

Lynch-Day, M. A., et al. (2012). The Role of Autophagy in Parkinson’s Disease. Cold Spring

Harbor Perspectives in Medicine, 2(4), a009357. doi:10.1101/cshperspect.a009357

Lynch, C. N., et al. (1999). TWEAK induces angiogenesis and proliferation of endothelial cells.

J Biol Chem, 274(13), 8455-8459.

Ma, C., et al. (2016). Pre-administration of BAX-inhibiting peptides decrease the loss of the

nigral dopaminergic neurons in rats. Life Sci, 144, 113-120. doi:10.1016/j.lfs.2015.11.019

Ma, C. L., et al. (2014). The prevalence and incidence of Parkinson's disease in China: a

systematic review and meta-analysis. J Neural Transm (Vienna), 121(2), 123-134.

doi:10.1007/s00702-013-1092-z

Ma, H., et al. (2001). Visualization of Syk-antigen receptor interactions using green fluorescent

protein: differential roles for Syk and Lyn in the regulation of receptor capping and

internalization. J Immunol, 166(3), 1507-1516.

Maccarrone, M., et al. (2001). Lipoxygenases and their involvement in programmed cell death.

Cell Death Differ, 8(8), 776-784. doi:10.1038/sj.cdd.4400908

Maharjan, S., et al. (2014). Mitochondrial impairment triggers cytosolic oxidative stress and cell

death following proteasome inhibition. Scientific Reports, 4, 5896.

doi:10.1038/srep05896

Maiuri, M. C., et al. (2007). Self-eating and self-killing: crosstalk between autophagy and

apoptosis. Nat Rev Mol Cell Biol, 8(9), 741-752. doi:10.1038/nrm2239

109

Mandir, A. S., et al. (1999). Poly(ADP-ribose) polymerase activation mediates 1-methyl-4-

phenyl-1, 2,3,6-tetrahydropyridine (MPTP)-induced parkinsonism. Proc Natl Acad Sci U

S A, 96(10), 5774-5779.

Mao, Z., et al. (2017). The NLRP3 Inflammasome is Involved in the Pathogenesis of Parkinson's

Disease in Rats. Neurochem Res, 42(4), 1104-1115. doi:10.1007/s11064-017-2185-0

Marakalala, M. J., et al. (2010). The role of Syk/CARD9-coupled C-type lectin receptors in

immunity to Mycobacterium tuberculosis infections. Clin Dev Immunol, 2010, 567571.

doi:10.1155/2010/567571

Marella, M., et al. (2009). Parkinson’s disease and mitochondrial complex I: a perspective on the

Ndi1 therapy. Journal of bioenergetics and biomembranes, 41(6), 493-497.

doi:10.1007/s10863-009-9249-z

Mariathasan, S., et al. (2006). Cryopyrin activates the inflammasome in response to toxins and

ATP (Vol. 440).

Marques, O., et al. (2012). Alpha-synuclein: From secretion to dysfunction and death (Vol. 3).

Marsters, S. A., et al. (1998). Identification of a ligand for the death-domain-containing receptor

Apo3. Curr Biol, 8(9), 525-528.

Martin, L. J., et al. (2006). Parkinson's disease alpha-synuclein transgenic mice develop neuronal

mitochondrial degeneration and cell death. J Neurosci, 26(1), 41-50.

doi:10.1523/jneurosci.4308-05.2006

Martinon, F., et al. (2006). Gout-associated uric acid crystals activate the NALP3 inflammasome.

Nature, 440(7081), 237-241. doi:10.1038/nature04516

Martins, J. D., et al. (2015). Autophagy and inflammasome interplay. DNA Cell Biol, 34(4), 274-

281. doi:10.1089/dna.2014.2752

Masliah, E., et al. (2000). Dopaminergic loss and inclusion body formation in alpha-synuclein

mice: implications for neurodegenerative disorders. Science, 287(5456), 1265-1269.

110

Masuda, E. S., et al. (2008). Syk inhibitors as treatment for allergic rhinitis. Pulmonary

Pharmacology & Therapeutics, 21(3), 461-467.

doi:https://doi.org/10.1016/j.pupt.2007.06.002

Matsuda, N., et al. (2010). PINK1 stabilized by mitochondrial depolarization recruits Parkin to

damaged mitochondria and activates latent Parkin for mitophagy. J Cell Biol, 189(2),

211-221. doi:10.1083/jcb.200910140

McCoy, M. K., et al. (2011). DJ-1 regulation of mitochondrial function and autophagy through

oxidative stress. Autophagy, 7(5), 531-532.

McCoy, M. K., et al. (2006). Blocking soluble tumor necrosis factor signaling with dominant-

negative tumor necrosis factor inhibitor attenuates loss of dopaminergic neurons in

models of Parkinson's disease. J Neurosci, 26(37), 9365-9375.

doi:10.1523/jneurosci.1504-06.2006

McKenzie, S. E. (2016). Syk Inhibition in Ischemic Stroke. Arterioscler Thromb Vasc Biol,

36(6), 1054-1055. doi:10.1161/atvbaha.116.307709

McNaught, K. S., et al. (2001). Failure of the ubiquitin-proteasome system in Parkinson's

disease. Nat Rev Neurosci, 2(8), 589-594. doi:10.1038/35086067

Medeiros, M. S., et al. (2016). Iron and Oxidative Stress in Parkinson’s Disease: An

Observational Study of Injury Biomarkers. PLOS ONE, 11(1), e0146129.

doi:10.1371/journal.pone.0146129

Meighan-Mantha, R. L., et al. (1999). The mitogen-inducible Fn14 gene encodes a type I

transmembrane protein that modulates fibroblast adhesion and migration. J Biol Chem,

274(46), 33166-33176.

Meiser, J., et al. (2013). Complexity of dopamine metabolism. Cell Commun Signal, 11(1), 34.

doi:10.1186/1478-811x-11-34

Meiser, J., et al. (2013a). Complexity of dopamine metabolism. Cell Communication and

Signaling, 11(1), 34. doi:10.1186/1478-811x-11-34

Meiser, J., et al. (2013b). Complexity of dopamine metabolism. Cell Communication and

Signaling : CCS, 11, 34-34. doi:10.1186/1478-811X-11-34

111

Menu, P., et al. (2012). ER stress activates the NLRP3 inflammasome via an UPR-independent

pathway (Vol. 3).

Mercado, G., et al. (2015). ER proteostasis disturbances in Parkinson's disease: novel insights.

Frontiers in Aging Neuroscience, 7(39). doi:10.3389/fnagi.2015.00039

Meredith, G., et al. (2011). MPTP mouse models of Parkinson's disease: An update (Vol. 1).

Meredith, G. E., et al. (2008). Modeling PD pathogenesis in mice: Advantages of a chronic

MPTP protocol. Parkinsonism & Related Disorders, 14, S112-S115.

doi:https://doi.org/10.1016/j.parkreldis.2008.04.012

Metz, G. A., et al. (2005). The unilateral 6-OHDA rat model of Parkinson's disease revisited: an

electromyographic and behavioural analysis. Eur J Neurosci, 22(3), 735-744.

doi:10.1111/j.1460-9568.2005.04238.x

Meulener, M., et al. (2005). Drosophila DJ-1 mutants are selectively sensitive to environmental

toxins associated with Parkinson's disease. Curr Biol, 15(17), 1572-1577.

doi:10.1016/j.cub.2005.07.064

Michaelson, J. S., et al. (2011). Development of an Fn14 agonistic antibody as an anti-tumor

agent. MAbs, 3(4), 362-375.

Michel, P. P., et al. (2016). Understanding Dopaminergic Cell Death Pathways in Parkinson

Disease. Neuron, 90(4), 675-691. doi:10.1016/j.neuron.2016.03.038

Milber, J. M., et al. (2012). Lewy pathology is not the first sign of degeneration in vulnerable

neurons in Parkinson disease. Neurology, 79(24), 2307-2314.

doi:10.1212/WNL.0b013e318278fe32

Miller, Y. I., et al. (2012). The SYK side of TLR4: signalling mechanisms in response to LPS

and minimally oxidized LDL. Br J Pharmacol, 167(5), 990-999. doi:10.1111/j.1476-

5381.2012.02097.x

Mochizuki, H., et al. (1996). Histochemical detection of apoptosis in Parkinson's disease. J

Neurol Sci, 137(2), 120-123.

112

Mochizuki, H., et al. (2006). Alpha-synuclein overexpression model. J Neural Transm

Suppl(70), 281-284.

Mocsai, A., et al. (2004). The immunomodulatory adapter proteins DAP12 and Fc receptor

gamma-chain (FcRgamma) regulate development of functional osteoclasts through the

Syk tyrosine kinase. Proc Natl Acad Sci U S A, 101(16), 6158-6163.

doi:10.1073/pnas.0401602101

Mócsai, A., et al. (2010). The SYK tyrosine kinase: a crucial player in diverse biological

functions. Nature reviews. Immunology, 10(6), 387-402. doi:10.1038/nri2765

Mogi, M., et al. (2000). Caspase activities and tumor necrosis factor receptor R1 (p55) level are

elevated in the substantia nigra from parkinsonian brain. J Neural Transm (Vienna),

107(3), 335-341. doi:10.1007/s007020050028

Moore, D. J., et al. (2005). Molecular pathophysiology of Parkinson's disease. Annu Rev

Neurosci, 28, 57-87. doi:10.1146/annurev.neuro.28.061604.135718

Mori, H., et al. (1998). Pathologic and biochemical studies of juvenile parkinsonism linked to

chromosome 6q. Neurology, 51(3), 890-892.

Mott, R. T., et al. (2004). Neuronal expression of CD22: novel mechanism for inhibiting

microglial proinflammatory cytokine production. Glia, 46(4), 369-379.

doi:10.1002/glia.20009

Moubarak, R. S., et al. (2007). Sequential Activation of Poly(ADP-Ribose) Polymerase 1,

Calpains, and Bax Is Essential in Apoptosis-Inducing Factor-Mediated Programmed

Necrosis. Mol Cell Biol, 27(13), 4844-4862. doi:10.1128/mcb.02141-06

Mueller, A. M., et al. (2005). Targeting fibroblast growth factor-inducible-14 signaling protects

from chronic relapsing experimental autoimmune encephalomyelitis. J Neuroimmunol,

159(1-2), 55-65. doi:10.1016/j.jneuroim.2004.10.001

Mueller, A. M., et al. (2005). Targeting fibroblast growth factor-inducible-14 signaling protects

from chronic relapsing experimental autoimmune encephalomyelitis. Journal of

Neuroimmunology, 159(1), 55-65. doi:https://doi.org/10.1016/j.jneuroim.2004.10.001

Mulak, A., et al. (2015). Brain-gut-microbiota axis in Parkinson's disease. World Journal of

Gastroenterology : WJG, 21(37), 10609-10620. doi:10.3748/wjg.v21.i37.10609

113

Muralikrishnan, D., et al. (1998). Neuroprotection by bromocriptine against 1-methyl-4-phenyl-

1,2,3,6-tetrahydropyridine-induced neurotoxicity in mice. Faseb j, 12(10), 905-912.

Mustafa, S., et al. (2016a). The role of TWEAK/Fn14 signaling in the MPTP-model of

Parkinson's disease. Neuroscience, 319, 116-122.

doi:10.1016/j.neuroscience.2016.01.034

Mustafa, S., et al. (2016b). The role of TWEAK/Fn14 signaling in the MPTP-model of

Parkinson’s disease. Neuroscience, 319, 116-122.

doi:10.1016/j.neuroscience.2016.01.034

Muthane, U., et al. (1994). Differences in nigral neuron number and sensitivity to 1-methyl-4-

phenyl-1,2,3,6-tetrahydropyridine in C57/bl and CD-1 mice. Exp Neurol, 126(2), 195-

204. doi:10.1006/exnr.1994.1058

Mytilineou, C., et al. (2002). Glutathione depletion and oxidative stress. Parkinsonism & Related

Disorders, 8(6), 385-387. doi:https://doi.org/10.1016/S1353-8020(02)00018-4

Nah, J., et al. (2015). Autophagy in Neurodegenerative Diseases: From Mechanism to

Therapeutic Approach. Molecules and Cells, 38(5), 381-389.

doi:10.14348/molcells.2015.0034

Naimark, D., et al. (1996). Psychotic symptoms in Parkinson's disease patients with dementia. J

Am Geriatr Soc, 44(3), 296-299.

Naismith, J. H., et al. (1995). Crystallographic evidence for dimerization of unliganded tumor

necrosis factor receptor. J Biol Chem, 270(22), 13303-13307.

Nakagawa, T., et al. (2000). Cross-Talk between Two Cysteine Protease Families: Activation of

Caspase-12 by Calpain in Apoptosis. The Journal of Cell Biology, 150(4), 887-894.

Nakahira, K., et al. (2011). Autophagy proteins regulate innate immune responses by inhibiting

the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol,

12(3), 222-230. doi:10.1038/ni.1980

Nakayama, M., et al. (2002). Multiple pathways of TWEAK-induced cell death. J Immunol,

168(2), 734-743.

114

Narendra, D., et al. (2010). p62/SQSTM1 is required for Parkin-induced mitochondrial clustering

but not mitophagy; VDAC1 is dispensable for both. Autophagy, 6(8), 1090-1106.

Narendra, D., et al. (2008). Parkin is recruited selectively to impaired mitochondria and promotes

their autophagy. J Cell Biol, 183(5), 795-803. doi:10.1083/jcb.200809125

Nazeri, A., et al. (2013). A Further TWEAK to Multiple Sclerosis Pathophysiology (Vol. 49).

Nazeri, A., et al. (2014). A further TWEAK to multiple sclerosis pathophysiology. Mol

Neurobiol, 49(1), 78-87. doi:10.1007/s12035-013-8490-y

Nedelsky, N. B., et al. (2008). Autophagy and the ubiquitin-proteasome system: collaborators in

neuroprotection. Biochim Biophys Acta, 1782(12), 691-699.

doi:10.1016/j.bbadis.2008.10.002

Negro, A., et al. (2002). Multiple phosphorylation of alpha-synuclein by protein tyrosine kinase

Syk prevents eosin-induced aggregation. Faseb j, 16(2), 210-212. doi:10.1096/fj.01-

0517fje

Newton, K., et al. (2012). Signaling in innate immunity and inflammation. Cold Spring Harb

Perspect Biol, 4(3). doi:10.1101/cshperspect.a006049

Nicklas, W. J., et al. (1985). Inhibition of NADH-linked oxidation in brain mitochondria by 1-

methyl-4-phenyl-pyridine, a metabolite of the neurotoxin, 1-methyl-4-phenyl-1,2,5,6-

tetrahydropyridine. Life Sci, 36(26), 2503-2508.

Nijjar, J. S., et al. (2013). Inhibition of spleen tyrosine kinase in the treatment of rheumatoid

arthritis. Rheumatology (Oxford), 52(9), 1556-1562. doi:10.1093/rheumatology/ket225

Niso-Santano, M., et al. (2011). ASK1 overexpression accelerates paraquat-induced autophagy

via endoplasmic reticulum stress. Toxicol Sci, 119(1), 156-168.

doi:10.1093/toxsci/kfq313

Nixon, R. A. (2013). The role of autophagy in neurodegenerative disease. Nat Med, 19(8), 983-

997. doi:10.1038/nm.3232

115

Nixon, R. A., et al. (2012). Autophagy and Neuronal Cell Death in Neurological Disorders. Cold

Spring Harbor Perspectives in Biology, 4(10), a008839.

doi:10.1101/cshperspect.a008839

Nolan, Y. M., et al. (2013). Parkinson's disease in the nuclear age of neuroinflammation. Trends

Mol Med, 19(3), 187-196. doi:10.1016/j.molmed.2012.12.003

Nopparat, C., et al. (2014). 1-Methyl-4-phenylpyridinium-induced cell death via autophagy

through a Bcl-2/Beclin 1 complex-dependent pathway. Neurochem Res, 39(2), 225-232.

doi:10.1007/s11064-013-1208-8

Novikova, L., et al. (2006). Early signs of neuronal apoptosis in the substantia nigra pars

compacta of the progressive neurodegenerative mouse 1-methyl-4-phenyl-1,2,3,6-

tetrahydropyridine/probenecid model of Parkinson’s disease. Neuroscience, 140(1), 67-

76. doi:https://doi.org/10.1016/j.neuroscience.2006.02.007

Novoyatleva, T., et al. (2010). TWEAK is a positive regulator of cardiomyocyte proliferation.

Cardiovasc Res, 85(4), 681-690. doi:10.1093/cvr/cvp360

Numakawa, T., et al. (2004). Neuronal Roles of the Integrin-associated Protein (IAP/CD47) in

Developing Cortical Neurons. Journal of Biological Chemistry, 279(41), 43245-43253.

Offen, D., et al. (2000). Mice overexpressing Bcl-2 in their neurons are resistant to myelin

oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune

encephalomyelitis (EAE). J Mol Neurosci, 15(3), 167-176. doi:10.1385/jmn:15:3:167

Ogata, M., et al. (2006). Autophagy is activated for cell survival after endoplasmic reticulum

stress. Mol Cell Biol, 26(24), 9220-9231. doi:10.1128/mcb.01453-06

Olanow, C. W., et al. (2006). Ubiquitin–proteasome system and Parkinson's disease. Movement

Disorders, 21(11), 1806-1823. doi:10.1002/mds.21013

Oyadomari, S., et al. (2004). Roles of CHOP&sol;GADD153 in endoplasmic reticulum stress

(Vol. 11).

Pamuk, O. N., et al. (2010). Spleen tyrosine kinase inhibition prevents tissue damage after

ischemia-reperfusion. Am J Physiol Gastrointest Liver Physiol, 299(2), G391-399.

doi:10.1152/ajpgi.00198.2010

116

Pan, T., et al. (2008). The role of autophagy-lysosome pathway in neurodegeneration associated

with Parkinson's disease. Brain, 131(Pt 8), 1969-1978. doi:10.1093/brain/awm318

Panicker, N., et al. (2015). Fyn Kinase Regulates Microglial Neuroinflammatory Responses in

Cell Culture and Animal Models of Parkinson's Disease. J Neurosci, 35(27), 10058-

10077. doi:10.1523/jneurosci.0302-15.2015

Pankiv, S., et al. (2007). p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of

ubiquitinated protein aggregates by autophagy. J Biol Chem, 282(33), 24131-24145.

doi:10.1074/jbc.M702824200

Paris, D., et al. (2014). The Spleen Tyrosine Kinase (Syk) Regulates Alzheimer Amyloid-β

Production and Tau Hyperphosphorylation. Journal of Biological Chemistry, 289(49),

33927-33944. doi:10.1074/jbc.M114.608091

Paschen, W., et al. (2001). Endoplasmic reticulum dysfunction – a common denominator for cell

injury in acute and degenerative diseases of the brain? Journal of Neurochemistry, 79(4),

719-725. doi:10.1046/j.1471-4159.2001.00623.x

Pasinetti, G. M. (1998). Cyclooxygenase and inflammation in Alzheimer's disease: experimental

approaches and clinical interventions. J Neurosci Res, 54(1), 1-6. doi:10.1002/(sici)1097-

4547(19981001)54:1<1::aid-jnr1>3.0.co;2-m

Paumier, K. L., et al. (2015). Intrastriatal injection of pre-formed mouse alpha-synuclein fibrils

into rats triggers alpha-synuclein pathology and bilateral nigrostriatal degeneration.

Neurobiol Dis, 82, 185-199. doi:10.1016/j.nbd.2015.06.003

Pelegrin, P., et al. (2006). Pannexin-1 mediates large pore formation and interleukin-1β release

by the ATP-gated P2X(7) receptor. The EMBO Journal, 25(21), 5071-5082.

doi:10.1038/sj.emboj.7601378

Pellegrini, C., et al. (2017). Canonical and Non-Canonical Activation of NLRP3 Inflammasome

at the Crossroad between Immune Tolerance and Intestinal Inflammation. Frontiers in

Immunology, 8, 36. doi:10.3389/fimmu.2017.00036

Peng, T. I., et al. (2010). Oxidative stress caused by mitochondrial calcium overload. Ann N Y

Acad Sci, 1201, 183-188. doi:10.1111/j.1749-6632.2010.05634.x

117

Perez, R. G., et al. (2002). A role for alpha-synuclein in the regulation of dopamine biosynthesis.

J Neurosci, 22(8), 3090-3099. doi:20026307

Perier, C., et al. (2012). Mitochondrial Biology and Parkinson's Disease. Cold Spring Harbor

Perspectives in Medicine, 2(2), a009332. doi:10.1101/cshperspect.a009332

Periquet, M., et al. (2007). Aggregated alpha-synuclein mediates dopaminergic neurotoxicity in

vivo. J Neurosci, 27(12), 3338-3346. doi:10.1523/jneurosci.0285-07.2007

Perper, S. J., et al. (2006). TWEAK is a novel arthritogenic mediator. J Immunol, 177(4), 2610-

2620.

Perry, T. L., et al. (1985). Partial protection from the dopaminergic neurotoxin N-methyl-4-

phenyl-1,2,3,6-tetrahydropyridine by four different antioxidants in the mouse. Neurosci

Lett, 60(2), 109-114.

Petroske, E., et al. (2001). Mouse model of Parkinsonism: A comparison between subacute

MPTP and chronic MPTP/probenecid treatment (Vol. 106).

Phatnani, H., et al. (2015). Astrocytes in neurodegenerative disease. Cold Spring Harb Perspect

Biol, 7(6). doi:10.1101/cshperspect.a020628

Pirkevi, C., et al. (2009). From genes to proteins in mendelian Parkinson's disease: an overview.

Anat Rec (Hoboken), 292(12), 1893-1901. doi:10.1002/ar.20968

Pisanu, A., et al. (2014). Dynamic changes in pro- and anti-inflammatory cytokines in microglia

after PPAR-gamma agonist neuroprotective treatment in the MPTPp mouse model of

progressive Parkinson's disease. Neurobiol Dis, 71, 280-291.

doi:10.1016/j.nbd.2014.08.011

Podolanczuk, A., et al. (2009). Of mice and men: an open-label pilot study for treatment of

immune thrombocytopenic purpura by an inhibitor of Syk. Blood, 113(14), 3154-3160.

doi:10.1182/blood-2008-07-166439

Polavarapu, R., et al. (2005). Tumor necrosis factor-like weak inducer of apoptosis increases the

permeability of the neurovascular unit through nuclear factor-kappa B pathway

activation. J Neurosci, 25(44), 10094-10100. doi:10.1523/jneurosci.3382-05.2005

118

Polster, B. M., et al. (2005). Calpain I induces cleavage and release of apoptosis-inducing factor

from isolated mitochondria. J Biol Chem, 280(8), 6447-6454.

doi:10.1074/jbc.M413269200

Potrovita, I., et al. (2004). Tumor necrosis factor-like weak inducer of apoptosis-induced

neurodegeneration. J Neurosci, 24(38), 8237-8244. doi:10.1523/jneurosci.1089-04.2004

Prinz-Hadad, H., et al. (2013). Amelioration of autoimmune neuroinflammation by the fusion

molecule Fn14·TRAIL. J Neuroinflammation, 10, 36-36. doi:10.1186/1742-2094-10-36

Przedborski, S. (2005). Pathogenesis of nigral cell death in Parkinson's disease. Parkinsonism

Relat Disord, 11 Suppl 1, S3-7. doi:10.1016/j.parkreldis.2004.10.012

Przedborski, S., et al. (2000). The parkinsonian toxin MPTP: action and mechanism. Restor

Neurol Neurosci, 16(2), 135-142.

Puntarulo, S. (2005). Iron, oxidative stress and human health. Molecular Aspects of Medicine,

26(4), 299-312. doi:https://doi.org/10.1016/j.mam.2005.07.001

Py, B. F., et al. (2013). Deubiquitination of NLRP3 by BRCC3 critically regulates

inflammasome activity. Mol Cell, 49(2), 331-338. doi:10.1016/j.molcel.2012.11.009

Qian, L., et al. (2006). Interleukin-10 Protects Lipopolysaccharide-Induced Neurotoxicity in

Primary Midbrain Cultures by Inhibiting the Function of NADPH Oxidase. Journal of

Pharmacology and Experimental Therapeutics, 319(1), 44-52.

doi:10.1124/jpet.106.106351

Qian, S., et al. (2017). Inflammasome and Autophagy Regulation: A Two-way Street. Molecular

Medicine, 23, 188-195. doi:10.2119/molmed.2017.00077

Qin, L., et al. (2004). NADPH Oxidase Mediates Lipopolysaccharide-induced Neurotoxicity and

Proinflammatory Gene Expression in Activated Microglia. Journal of Biological

Chemistry, 279(2), 1415-1421. doi:10.1074/jbc.M307657200

Qin, L., et al. (2007). Systemic LPS causes chronic neuroinflammation and progressive

neurodegeneration. Glia, 55(5), 453-462. doi:10.1002/glia.20467

119

Rao, R. V., et al. (2004). Coupling endoplasmic reticulum stress to the cell death program. Cell

Death And Differentiation, 11, 372. doi:10.1038/sj.cdd.4401378

Rappold, P. M., et al. (2010). Astrocytes and therapeutics for Parkinson’s disease.

Neurotherapeutics, 7(4), 413-423. doi:10.1016/j.nurt.2010.07.001

Rathinam, V. A., et al. (2016). Inflammasome Complexes: Emerging Mechanisms and Effector

Functions. Cell, 165(4), 792-800. doi:10.1016/j.cell.2016.03.046

Rathinam, Vijay A. K., et al. TRIF Licenses Caspase-11-Dependent NLRP3 Inflammasome

Activation by Gram-Negative Bacteria. Cell, 150(3), 606-619.

doi:10.1016/j.cell.2012.07.007

Ray, A., et al. (2015). MPTP activates ASK1–p38 MAPK signaling pathway through TNF-

dependent Trx1 oxidation in parkinsonism mouse model. Free Radical Biology and

Medicine, 87, 312-325. doi:https://doi.org/10.1016/j.freeradbiomed.2015.06.041

Recasens, A., et al. (2014). Alpha-synuclein spreading in Parkinson’s disease. Frontiers in

Neuroanatomy, 8(159). doi:10.3389/fnana.2014.00159

Recchia, A., et al. (2008). Generation of a alpha-synuclein-based rat model of Parkinson's

disease. Neurobiol Dis, 30(1), 8-18. doi:10.1016/j.nbd.2007.11.002

Ricote, M., et al. (1998). The peroxisome proliferator-activated receptor-gamma is a negative

regulator of macrophage activation. Nature, 391(6662), 79-82. doi:10.1038/34178

Rideout, H. J., et al. (2014). The neurobiology of LRRK2 and its role in the pathogenesis of

Parkinson's disease. Neurochem Res, 39(3), 576-592. doi:10.1007/s11064-013-1073-5

Riess, O., et al. (1999). Parkinson's disease--a multifactorial neurodegenerative disorder. J

Neural Transm Suppl, 56, 113-125.

Rietdijk, C. D., et al. (2017). Exploring Braak’s Hypothesis of Parkinson’s Disease. Frontiers in

Neurology, 8, 37. doi:10.3389/fneur.2017.00037

Rivera, J., et al. (2009). Platelet receptors and signaling in the dynamics of thrombus formation.

Haematologica, 94(5), 700-711. doi:10.3324/haematol.2008.003178

120

Rocha, N. P., et al. (2015). Insights into Neuroinflammation in Parkinson's Disease: From

Biomarkers to Anti-Inflammatory Based Therapies. Biomed Res Int, 2015, 628192.

doi:10.1155/2015/628192

Rocha, S., et al. (2014). Long-Term Mortality Analysis in Parkinson&#x2019;s Disease Treated

with Deep Brain Stimulation. Parkinson&#x2019;s Disease, 2014, 5.

doi:10.1155/2014/717041

Rodgers, M. A., et al. (2014). Regulation where autophagy intersects the inflammasome.

Antioxid Redox Signal, 20(3), 495-506. doi:10.1089/ars.2013.5347

Rogers, N. C., et al. (2005). Syk-dependent cytokine induction by Dectin-1 reveals a novel

pattern recognition pathway for C type lectins. Immunity, 22(4), 507-517.

doi:10.1016/j.immuni.2005.03.004

Ron, D., et al. (2008). How IRE1 Reacts to ER Stress. Cell, 132(1), 24-26.

doi:https://doi.org/10.1016/j.cell.2007.12.017

Roos, C., et al. (2010). Soluble and transmembrane TNF-like weak inducer of apoptosis

differentially activate the classical and noncanonical NF-kappa B pathway. J Immunol,

185(3), 1593-1605. doi:10.4049/jimmunol.0903555

Rossi, A., et al. (2006). Sarcoplasmic reticulum: The dynamic calcium governor of muscle (Vol.

33).

Rozas, G., et al. (1997). An automated rotarod method for quantitative drug-free evaluation of

overall motor deficits in rat models of parkinsonism. Brain Res Brain Res Protoc, 2(1),

75-84.

Rozas, G., et al. (1998). The overall rod performance test in the MPTP-treated-mouse model of

Parkinsonism. J Neurosci Methods, 83(2), 165-175.

Rubin, L. L., et al. (1999). The cell biology of the blood-brain barrier. Annu Rev Neurosci, 22,

11-28. doi:10.1146/annurev.neuro.22.1.11

Ryu, E. J., et al. (2002). Endoplasmic Reticulum Stress and the Unfolded Protein Response in

Cellular Models of Parkinson's Disease. The Journal of Neuroscience, 22(24), 10690-

10698.

121

Ryu, E. J., et al. (2002). Endoplasmic reticulum stress and the unfolded protein response in

cellular models of Parkinson's disease. J Neurosci, 22(24), 10690-10698.

Ryu, J. K., et al. (2006). Minocycline or iNOS inhibition block 3-nitrotyrosine increases and

blood-brain barrier leakiness in amyloid beta-peptide-injected rat hippocampus. Exp

Neurol, 198(2), 552-557. doi:10.1016/j.expneurol.2005.12.016

S, S. S., et al. (2012). (Vol. 122).

Sada, K., et al. (2001). Structure and function of Syk protein-tyrosine kinase. J Biochem, 130(2),

177-186.

Saha, A. R., et al. (2000). Induction of neuronal death by alpha-synuclein. Eur J Neurosci, 12(8),

3073-3077.

Saito, Y., et al. (2012). [Braak's hypothesis and non-motor symptoms of Parkinson disease].

Brain Nerve, 64(4), 444-452.

Saito, Y., et al. (2007). Molecular mechanisms of 6-hydroxydopamine-induced cytotoxicity in

PC12 cells: involvement of hydrogen peroxide-dependent and -independent action. Free

Radic Biol Med, 42(5), 675-685. doi:10.1016/j.freeradbiomed.2006.12.004

Saitoh, T., et al. (2016). Regulation of inflammasomes by autophagy. Journal of Allergy and

Clinical Immunology, 138(1), 28-36. doi:https://doi.org/10.1016/j.jaci.2016.05.009

Sancho, D., et al. (2009). Identification of a dendritic cell receptor that couples sensing of

necrosis to immunity. Nature, 458(7240), 899-903. doi:10.1038/nature07750

Sano, R., et al. (2013). ER stress-induced cell death mechanisms. Biochimica et Biophysica Acta

(BBA) - Molecular Cell Research, 1833(12), 3460-3470.

doi:https://doi.org/10.1016/j.bbamcr.2013.06.028

Sano, R., et al. (2013). ER stress-induced cell death mechanisms. Biochim Biophys Acta,

1833(12), 3460-3470. doi:10.1016/j.bbamcr.2013.06.028

Santa-Cecilia, F. V., et al. (2016). Doxycycline Suppresses Microglial Activation by Inhibiting

the p38 MAPK and NF-kB Signaling Pathways. Neurotox Res, 29(4), 447-459.

doi:10.1007/s12640-015-9592-2

122

Santpere, G., et al. (2009). LRRK2 and neurodegeneration. Acta Neuropathol, 117(3), 227-246.

doi:10.1007/s00401-008-0478-8

Sanz, A. B., et al. (2010). TWEAK activates the non-canonical NFkappaB pathway in murine

renal tubular cells: modulation of CCL21. PLOS ONE, 5(1), e8955.

doi:10.1371/journal.pone.0008955

Satoh, J., et al. (2012). Phosphorylated Syk expression is enhanced in Nasu-Hakola disease

brains. Neuropathology, 32(2), 149-157. doi:10.1111/j.1440-1789.2011.01256.x

Sawada, M., et al. (2006). Role of cytokines in inflammatory process in Parkinson's disease. J

Neural Transm Suppl(70), 373-381.

Scarffe, L. A., et al. (2014). Parkin and PINK1: much more than mitophagy. Trends Neurosci,

37(6), 315-324. doi:10.1016/j.tins.2014.03.004

Schletter, J., et al. (1996). Molecular mechanisms of endotoxin activity (Vol. 164).

Scholzke, M. N., et al. (2011). TWEAK regulates proliferation and differentiation of adult neural

progenitor cells. Mol Cell Neurosci, 46(1), 325-332. doi:10.1016/j.mcn.2010.10.004

Schroder, K., et al. (2010). The Inflammasomes. Cell, 140(6), 821-832.

doi:https://doi.org/10.1016/j.cell.2010.01.040

Schroder, K., et al. (2010). The NLRP3 inflammasome: a sensor for metabolic danger? Science,

327(5963), 296-300. doi:10.1126/science.1184003

Schwarz, D. S., et al. (2016). The endoplasmic reticulum: structure, function and response to

cellular signaling. Cell Mol Life Sci, 73(1), 79-94. doi:10.1007/s00018-015-2052-6

Schweig, J. E., et al. (2017). Alzheimer’s disease pathological lesions activate the spleen tyrosine

kinase. Acta Neuropathologica Communications, 5, 69. doi:10.1186/s40478-017-0472-2

Scott, D. L. (2011). Role of spleen tyrosine kinase inhibitors in the management of rheumatoid

arthritis. Drugs, 71(9), 1121-1132. doi:10.2165/11591480-000000000-00000

123

Scott, I. C., et al. (2014). Spleen tyrosine kinase inhibitors for rheumatoid arthritis: where are we

now? Drugs, 74(4), 415-422. doi:10.1007/s40265-014-0193-9

Scott, R. C., et al. (2007). Direct induction of autophagy by Atg1 inhibits cell growth and

induces apoptotic cell death. Curr Biol, 17(1), 1-11. doi:10.1016/j.cub.2006.10.053

Sedelis, M., et al. (2000). MPTP susceptibility in the mouse: behavioral, neurochemical, and

histological analysis of gender and strain differences. Behav Genet, 30(3), 171-182.

Selvaraj, S., et al. (2012). Neurotoxin-induced ER stress in mouse dopaminergic neurons

involves downregulation of TRPC1 and inhibition of AKT/mTOR signaling. J Clin

Invest, 122(4), 1354-1367. doi:10.1172/jci61332

Serafini, B., et al. (2008). Expression of TWEAK and its receptor Fn14 in the multiple sclerosis

brain: implications for inflammatory tissue injury. J Neuropathol Exp Neurol, 67(12),

1137-1148. doi:10.1097/NEN.0b013e31818dab90

Shen, S., et al. (2011). Association and dissociation of autophagy, apoptosis and necrosis by

systematic chemical study. Oncogene, 30(45), 4544-4556. doi:10.1038/onc.2011.168

Sheridan, G. K., et al. (2013). Neuron–glia crosstalk in health and disease: fractalkine and

CX(3)CR1 take centre stage. Open Biology, 3(12), 130181. doi:10.1098/rsob.130181

Shi, C.-S., et al. (2012). Activation of Autophagy by Inflammatory Signals Limits IL-1β

Production by Targeting Ubiquitinated Inflammasomes for Destruction (Vol. 13).

Shi, C. S., et al. (2012). Activation of autophagy by inflammatory signals limits IL-1beta

production by targeting ubiquitinated inflammasomes for destruction. Nat Immunol,

13(3), 255-263. doi:10.1038/ni.2215

Shi, J., et al. (2015). Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell

death. Nature, 526(7575), 660-665. doi:10.1038/nature15514

Shimura, H., et al. (2000). Familial Parkinson disease gene product, parkin, is a ubiquitin-protein

ligase. Nat Genet, 25(3), 302-305. doi:10.1038/77060

124

Sian, J., et al. (1994). Alterations in glutathione levels in Parkinson's disease and other

neurodegenerative disorders affecting basal ganglia. Annals of Neurology, 36(3), 348-

355. doi:10.1002/ana.410360305

Sies, H. (2015). Oxidative stress: a concept in redox biology and medicine. Redox Biology, 4,

180-183. doi:https://doi.org/10.1016/j.redox.2015.01.002

Simola, N., et al. (2007). The 6-hydroxydopamine model of Parkinson's disease. Neurotox Res,

11(3-4), 151-167.

Singer, T. P., et al. (1990). Mechanism of the neurotoxicity of MPTP: An update. FEBS Letters,

274(1), 1-8. doi:https://doi.org/10.1016/0014-5793(90)81315-F

Smith, W. W., et al. (2005). Endoplasmic reticulum stress and mitochondrial cell death pathways

mediate A53T mutant alpha-synuclein-induced toxicity. Hum Mol Genet, 14(24), 3801-

3811. doi:10.1093/hmg/ddi396

Snyder, H., et al. (2003). Aggregated and monomeric alpha-synuclein bind to the S6'

proteasomal protein and inhibit proteasomal function. J Biol Chem, 278(14), 11753-

11759. doi:10.1074/jbc.M208641200

Sofroniew, M. V., et al. (2010). Astrocytes: biology and pathology. Acta Neuropathol, 119(1), 7-

35. doi:10.1007/s00401-009-0619-8

Song, J., et al. (2015). Mangiferin inhibits endoplasmic reticulum stress-associated thioredoxin-

interacting protein/NLRP3 inflammasome activation with regulation of AMPK in

endothelial cells. Metabolism, 64(3), 428-437. doi:10.1016/j.metabol.2014.11.008

Song, P., et al. (2016). Parkin promotes proteasomal degradation of p62: implication of selective

vulnerability of neuronal cells in the pathogenesis of Parkinson's disease. Protein Cell,

7(2), 114-129. doi:10.1007/s13238-015-0230-9

Spillantini, M. G., et al. (1998). α-Synuclein in filamentous inclusions of Lewy bodies from

Parkinson’s disease and dementia with Lewy bodies. Proceedings of the National

Academy of Sciences of the United States of America, 95(11), 6469-6473.

Spinelli, K. J., et al. (2014). Presynaptic Alpha-Synuclein Aggregation in a Mouse Model of

Parkinson's Disease. The Journal of Neuroscience, 34(6), 2037-2050.

doi:10.1523/jneurosci.2581-13.2014

125

Sriram, K., et al. (2002). Mice deficient in TNF receptors are protected against dopaminergic

neurotoxicity: implications for Parkinson's disease. Faseb j, 16(11), 1474-1476.

doi:10.1096/fj.02-0216fje

St Martin, J. L., et al. (2007). Dopaminergic neuron loss and up-regulation of chaperone protein

mRNA induced by targeted over-expression of alpha-synuclein in mouse substantia

nigra. J Neurochem, 100(6), 1449-1457. doi:10.1111/j.1471-4159.2006.04310.x

Stayte, S., et al. (2015). Activin A protects midbrain neurons in the 6-hydroxydopamine mouse

model of Parkinson's disease. PLOS ONE, 10(4), e0124325.

doi:10.1371/journal.pone.0124325

Stefanis, L., et al. (2001). Expression of A53T mutant but not wild-type alpha-synuclein in PC12

cells induces alterations of the ubiquitin-dependent degradation system, loss of dopamine

release, and autophagic cell death. J Neurosci, 21(24), 9549-9560.

Stephan, D., et al. (2013). TWEAK/Fn14 pathway modulates properties of a human

microvascular endothelial cell model of blood brain barrier. J Neuroinflammation, 10, 9-

9. doi:10.1186/1742-2094-10-9

Stock, A., et al. (2014). TWEAK Stimulation of Microglia and Implications for Neuropsychiatric

Lupus (P5.176). Neurology, 82(10 Supplement).

Subramaniam, S. R., et al. (2017). Targeting Microglial Activation States as a Therapeutic

Avenue in Parkinson’s Disease. Frontiers in Aging Neuroscience, 9, 176.

doi:10.3389/fnagi.2017.00176

Suen, D. F., et al. (2008). Mitochondrial dynamics and apoptosis. Genes Dev, 22(12), 1577-

1590. doi:10.1101/gad.1658508

Suzuki, Y., et al. (2013). Involvement of Mincle and Syk in the changes to innate immunity after

ischemic stroke. Scientific Reports, 3, 3177. doi:10.1038/srep03177

https://www.nature.com/articles/srep03177#supplementary-information

Swahn, C. G., et al. (1976). Determination of conjugated monoamine metabolites in brain tissue.

Journal of Neural Transmission, 39(4), 281-290. doi:10.1007/BF01266304

126

Szegezdi, E., et al. (2006). Mediators of endoplasmic reticulum stress-induced apoptosis. EMBO

Reports, 7(9), 880-885. doi:10.1038/sj.embor.7400779

Taira, T., et al. (2004). DJ-1 has a role in antioxidative stress to prevent cell death. EMBO Rep,

5(2), 213-218. doi:10.1038/sj.embor.7400074

Takano, K., et al. (2007). Methoxyflavones protect cells against endoplasmic reticulum stress

and neurotoxin (Vol. 292).

Tan, J. M., et al. (2009). Protein misfolding and aggregation in Parkinson's disease. Antioxid

Redox Signal, 11(9), 2119-2134. doi:10.1089/ars.2009.2490

Tanabe, K., et al. (2003). Fibroblast growth factor-inducible-14 is induced in axotomized

neurons and promotes neurite outgrowth. J Neurosci, 23(29), 9675-9686.

Taniguchi, T., et al. (1991). Molecular cloning of a porcine gene syk that encodes a 72-kDa

protein-tyrosine kinase showing high susceptibility to proteolysis. J Biol Chem, 266(24),

15790-15796.

Tanner, C. M., et al. (1996). Epidemiology of Parkinson's disease. Neurol Clin, 14(2), 317-335.

Tansey, M. G., et al. (2007). Neuroinflammatory mechanisms in Parkinson's disease: potential

environmental triggers, pathways, and targets for early therapeutic intervention. Exp

Neurol, 208(1), 1-25. doi:10.1016/j.expneurol.2007.07.004

Tatton, N. A. (2000). Increased caspase 3 and Bax immunoreactivity accompany nuclear

GAPDH translocation and neuronal apoptosis in Parkinson's disease. Exp Neurol, 166(1),

29-43. doi:10.1006/exnr.2000.7489

Tatton, N. A., et al. (1997). In situ detection of apoptotic nuclei in the substantia nigra compacta

of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice using terminal

deoxynucleotidyl transferase labelling and acridine orange staining. Neuroscience, 77(4),

1037-1048.

Tatton, N. A., et al. (1998). A fluorescent double-labeling method to detect and confirm

apoptotic nuclei in Parkinson's disease. Ann Neurol, 44(3 Suppl 1), S142-148.

127

Tatton, W. G., et al. (2003). Apoptosis in Parkinson's disease: signals for neuronal degradation.

Ann Neurol, 53 Suppl 3, S61-70; discussion S70-62. doi:10.1002/ana.10489

Taylor, J. P., et al. (2003). Aggresomes protect cells by enhancing the degradation of toxic

polyglutamine-containing protein. Hum Mol Genet, 12(7), 749-757.

Tian, Y. Y., et al. (2006). Catalpol protects dopaminergic neurons from LPS-induced

neurotoxicity in mesencephalic neuron-glia cultures. Life Sci, 80(3), 193-199.

doi:10.1016/j.lfs.2006.09.010

Tieu, K. (2011). A guide to neurotoxic animal models of Parkinson's disease. Cold Spring Harb

Perspect Med, 1(1), a009316. doi:10.1101/cshperspect.a009316

Tomalka, J., et al. (2011). A Novel Role for the NLRC4 Inflammasome in Mucosal Defenses

against the Fungal Pathogen Candida albicans. PLoS Pathog, 7(12), e1002379.

doi:10.1371/journal.ppat.1002379

Tomlinson, C. C., et al. (2004). The K1 protein of Kaposi's sarcoma-associated herpesvirus

activates the Akt signaling pathway. J Virol, 78(4), 1918-1927.

Tong, Q., et al. (2016). Inhibition of endoplasmic reticulum stress-activated IRE1α-TRAF2-

caspase-12 apoptotic pathway is involved in the neuroprotective effects of telmisartan in

the rotenone rat model of Parkinson's disease (Vol. 776).

Torres, E. M., et al. (2012). 6-OHDA Lesion Models of Parkinson’s Disease in the Rat. In E. L.

Lane & S. B. Dunnett (Eds.), Animal Models of Movement Disorders: Volume I (pp. 267-

279). Totowa, NJ: Humana Press.

Tran, N. L., et al. (2006). Increased fibroblast growth factor-inducible 14 expression levels

promote glioma cell invasion via Rac1 and nuclear factor-kappaB and correlate with poor

patient outcome. Cancer Res, 66(19), 9535-9542. doi:10.1158/0008-5472.can-06-0418

Tronnier, V. M., et al. (2002). High frequency stimulation of the basal ganglia for the treatment

of movement disorders: current status and clinical results. Minim Invasive Neurosurg,

45(2), 91-96. doi:10.1055/s-2002-32495

Tsang, E., et al. (2008). Molecular mechanism of the Syk activation switch. J Biol Chem,

283(47), 32650-32659. doi:10.1074/jbc.M806340200

128

Tsujii, S., et al. (2015). Modulation of endoplasmic reticulum stress in Parkinson's disease (Vol.

765).

Turmel, H., et al. (2001). Caspase-3 activation in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine

(MPTP)-treated mice. Mov Disord, 16(2), 185-189.

Turner, M., et al. (2000). Tyrosine kinase SYK: essential functions for immunoreceptor

signalling. Immunol Today, 21(3), 148-154.

Tysnes, O. B., et al. (2017). Epidemiology of Parkinson's disease. J Neural Transm (Vienna),

124(8), 901-905. doi:10.1007/s00702-017-1686-y

Urano, F., et al. (2000). Coupling of Stress in the ER to Activation of JNK Protein Kinases by

Transmembrane Protein Kinase IRE1 (Vol. 287).

Uutela, P., et al. (2009). Discovery of Dopamine Glucuronide in Rat and Mouse Brain

Microdialysis Samples Using Liquid Chromatography Tandem Mass Spectrometry.

Analytical Chemistry, 81(1), 427-434. doi:10.1021/ac801846w

Valdés, P., et al. (2014). Control of dopaminergic neuron survival by the unfolded protein

response transcription factor XBP1 (Vol. 111).

van Eeuwijk, J. M., et al. (2016). The Novel Oral Syk Inhibitor, Bl1002494, Protects Mice From

Arterial Thrombosis and Thromboinflammatory Brain Infarction. Arterioscler Thromb

Vasc Biol, 36(6), 1247-1253. doi:10.1161/atvbaha.115.306883

Varfolomeev, E., et al. (2012). Cellular inhibitors of apoptosis are global regulators of NF-

kappaB and MAPK activation by members of the TNF family of receptors. Sci Signal,

5(216), ra22. doi:10.1126/scisignal.2001878

Venda, L. L., et al. (2010). α-Synuclein and dopamine at the crossroads of Parkinson’s disease.

Trends in neurosciences, 33(12), 559-568. doi:10.1016/j.tins.2010.09.004

Venderova, K., et al. (2012). Programmed cell death in Parkinson's disease. Cold Spring Harb

Perspect Med, 2(8). doi:10.1101/cshperspect.a009365

129

Vila, M., et al. (2001). Bax ablation prevents dopaminergic neurodegeneration in the 1-methyl-

4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease. Proc Natl Acad

Sci U S A, 98(5), 2837-2842. doi:10.1073/pnas.051633998

Vilar, M., et al. (2008). The fold of α-synuclein fibrils. Proceedings of the National Academy of

Sciences of the United States of America, 105(25), 8637-8642.

doi:10.1073/pnas.0712179105

Vince, J. E., et al. (2008). TWEAK-FN14 signaling induces lysosomal degradation of a cIAP1-

TRAF2 complex to sensitize tumor cells to TNFalpha. J Cell Biol, 182(1), 171-184.

doi:10.1083/jcb.200801010

Vincent, C., et al. (2009). Pro-inflammatory cytokines TNF-related weak inducer of apoptosis

(TWEAK) and TNFalpha induce the mitogen-activated protein kinase (MAPK)-

dependent expression of sclerostin in human osteoblasts. J Bone Miner Res, 24(8), 1434-

1449. doi:10.1359/jbmr.090305

Visanji, N. P., et al. (2013). The prion hypothesis in Parkinson's disease: Braak to the future.

Acta Neuropathol Commun, 1, 2. doi:10.1186/2051-5960-1-2

von Coelln, R., et al. (2004). Parkin-associated Parkinson's disease. Cell Tissue Res, 318(1), 175-

184. doi:10.1007/s00441-004-0924-4

von Moltke, J., et al. (2012). Rapid induction of inflammatory lipid mediators by the

inflammasome in vivo. Nature, 490(7418), 107-111. doi:10.1038/nature11351

Vonsattel, J. P., et al. (1985). Neuropathological classification of Huntington's disease. J

Neuropathol Exp Neurol, 44(6), 559-577.

Voon, V., et al. (2009). Chronic dopaminergic stimulation in Parkinson's disease: from

dyskinesias to impulse control disorders. Lancet Neurol, 8(12), 1140-1149.

doi:10.1016/s1474-4422(09)70287-x

Vosler, P. S., et al. (2009). Calcium dysregulation induces apoptosis-inducing factor release:

Cross-talk between PARP-1- and calpain- signaling pathways. Experimental Neurology,

218(2), 213-220. doi:https://doi.org/10.1016/j.expneurol.2009.04.032

Vroon, A., et al. (2007). Neuroinflammation in Parkinson’s patients and MPTP-treated mice is

not restricted to the nigrostriatal system: Microgliosis and differential expression of

130

interleukin-1 receptors in the olfactory bulb. Experimental Gerontology, 42(8), 762-771.

doi:https://doi.org/10.1016/j.exger.2007.04.010

W Smith, W., et al. (2006). Endoplasmic reticulum stress and mitochondrial cell death pathways

mediate A53T mutant alpha-synuclein-induced toxicity (Vol. 14).

Wajant, H. (2013). The TWEAK-Fn14 system as a potential drug target. Br J Pharmacol,

170(4), 748-764. doi:10.1111/bph.12337

Wajant, H. (2013). The TWEAK-Fn14 system as a potential drug target. Br J Pharmacol,

170(4), 748-764. doi:10.1111/bph.12337

Wajant, H., et al. (1999). TNF receptor associated factors in cytokine signaling. Cytokine Growth

Factor Rev, 10(1), 15-26.

Wakabayashi, K., et al. (1997). Neuropathology of autonomic nervous system in Parkinson's

disease. Eur Neurol, 38 Suppl 2, 2-7.

Wakabayashi, K., et al. (2007). The Lewy body in Parkinson's disease: molecules implicated in

the formation and degradation of alpha-synuclein aggregates. Neuropathology, 27(5),

494-506.

Walsh, D. M., et al. (2016). A critical appraisal of the pathogenic protein spread hypothesis of

neurodegeneration. Nat Rev Neurosci, 17(4), 251-260. doi:10.1038/nrn.2016.13

Wang, B., et al. (2016). Dysregulation of autophagy and mitochondrial function in Parkinson’s

disease. Translational Neurodegeneration, 5(1), 19. doi:10.1186/s40035-016-0065-1

Wang, D., et al. (2017). The role of NLRP3-CASP1 in inflammasome-mediated

neuroinflammation and autophagy dysfunction in manganese-induced, hippocampal-

dependent impairment of learning and memory ability. Autophagy, 13(5), 914-927.

doi:10.1080/15548627.2017.1293766

Wang, H., et al. (2003). Apoptosis inducing factor and PARP-mediated injury in the MPTP

mouse model of Parkinson's disease. Ann N Y Acad Sci, 991, 132-139.

131

Wang, L., et al. (2003). Alternative splicing disrupts a nuclear localization signal in spleen

tyrosine kinase that is required for invasion suppression in breast cancer. Cancer Res,

63(15), 4724-4730.

Wang, Y., et al. (2009). Poly(ADP-ribose) Signals to Mitochondrial AIF: A Key Event in

Parthanatos. Experimental Neurology, 218(2), 193-202.

doi:10.1016/j.expneurol.2009.03.020

Wasserman, J. K., et al. (2007). Minocycline protects the blood-brain barrier and reduces edema

following intracerebral hemorrhage in the rat. Exp Neurol, 207(2), 227-237.

doi:10.1016/j.expneurol.2007.06.025

Watanabe, Y., et al. (2013). p62/SQSTM1-Dependent Autophagy of Lewy Body-Like α-

Synuclein Inclusions. PLOS ONE, 7(12), e52868. doi:10.1371/journal.pone.0052868

Weber, K., et al. (2014). Lysosomes Integrate Metabolic-Inflammatory Cross-talk in Primary

Macrophage Inflammasome Activation. Journal of Biological Chemistry, 289(13), 9158-

9171. doi:10.1074/jbc.M113.531202

Weinblatt, M. E., et al. (2008). Treatment of rheumatoid arthritis with a Syk kinase inhibitor: a

twelve-week, randomized, placebo-controlled trial. Arthritis Rheum, 58(11), 3309-3318.

doi:10.1002/art.23992

Wen, J., et al. (2013). Neuropsychiatric disease in murine lupus is dependent on the

TWEAK/Fn14 pathway. J Autoimmun, 43, 44-54. doi:10.1016/j.jaut.2013.03.002

Werkman, T. R., et al. (2006). Dopamine receptor pharmacology: interactions with serotonin

receptors and significance for the aetiology and treatment of schizophrenia. CNS Neurol

Disord Drug Targets, 5(1), 3-23.

Wichmann, T., et al. (2011). Deep-Brain Stimulation for Basal Ganglia Disorders. Basal ganglia,

1(2), 65-77. doi:10.1016/j.baga.2011.05.001

Wilms, H., et al. (2003). Activation of microglia by human neuromelanin is NF-kappaB

dependent and involves p38 mitogen-activated protein kinase: implications for

Parkinson's disease. Faseb j, 17(3), 500-502. doi:10.1096/fj.02-0314fje

132

Winkles, J. A. (2008). The TWEAK–Fn14 cytokine–receptor axis: discovery, biology and

therapeutic targeting. Nature reviews. Drug discovery, 7(5), 411-425.

doi:10.1038/nrd2488

Winkles, J. A., et al. (2006). TWEAK and Fn14: new molecular targets for cancer therapy?

Cancer Lett, 235(1), 11-17. doi:10.1016/j.canlet.2005.03.048

Winklhofer, K. F., et al. (2010). Mitochondrial dysfunction in Parkinson's disease. Biochimica et

Biophysica Acta (BBA) - Molecular Basis of Disease, 1802(1), 29-44.

doi:https://doi.org/10.1016/j.bbadis.2009.08.013

Winslow, A. R., et al. (2011). The Parkinson disease protein alpha-synuclein inhibits autophagy.

Autophagy, 7(4), 429-431.

Wolf, S. A., et al. (2017). Microglia in Physiology and Disease. Annual Review of Physiology,

79(1), 619-643. doi:10.1146/annurev-physiol-022516-034406

Wolozin, B., et al. (2015). Syk and Yea Shall Find. EBioMedicine, 2(11), 1590-1591.

doi:10.1016/j.ebiom.2015.11.012

Won, J. H., et al. (2015). Rotenone-induced Impairment of Mitochondrial Electron Transport

Chain Confers a Selective Priming Signal for NLRP3 Inflammasome Activation. J Biol

Chem, 290(45), 27425-27437. doi:10.1074/jbc.M115.667063

Wong, B. R., et al. (2004). Targeting Syk as a treatment for allergic and autoimmune disorders.

Expert Opin Investig Drugs, 13(7), 743-762. doi:10.1517/13543784.13.7.743

Woodside, D. G., et al. (2001). Activation of Syk protein tyrosine kinase through interaction with

integrin beta cytoplasmic domains. Curr Biol, 11(22), 1799-1804.

Wright Willis, A., et al. (2010). Geographic and ethnic variation in Parkinson disease: a

population-based study of US Medicare beneficiaries. Neuroepidemiology, 34(3), 143-

151. doi:10.1159/000275491

Wu, J., et al. (2007). ATF6α Optimizes Long-Term Endoplasmic Reticulum Function to Protect

Cells from Chronic Stress (Vol. 13).

133

Wu, L., et al. (2013). Inhibition of endoplasmic reticulum stress is involved in the

neuroprotective effects of candesartan cilexitil in the rotenone rat model of Parkinson's

disease. Neuroscience Letters, 548, 50-55.

doi:https://doi.org/10.1016/j.neulet.2013.06.008

Xilouri, M., et al. (2013). Boosting chaperone-mediated autophagy in vivo mitigates alpha-

synuclein-induced neurodegeneration. Brain, 136(Pt 7), 2130-2146.

doi:10.1093/brain/awt131

Xing, B., et al. (2011). Microglial p38α MAPK is critical for LPS-induced neuron degeneration,

through a mechanism involving TNFα. Molecular Neurodegeneration, 6(1), 84.

doi:10.1186/1750-1326-6-84

Xu, W.-D., et al. (2016). Role of the TWEAK/Fn14 pathway in autoimmune diseases.

Immunologic Research, 64(1), 44-50. doi:10.1007/s12026-015-8761-y

Xu, W. D., et al. (2016). Role of the TWEAK/Fn14 pathway in autoimmune diseases. Immunol

Res, 64(1), 44-50. doi:10.1007/s12026-015-8761-y

Xu, Y., et al. (2006). Poly(ADP-ribose) Polymerase-1 Signaling to Mitochondria in Necrotic

Cell Death Requires RIP1/TRAF2-mediated JNK1 Activation. Journal of Biological

Chemistry, 281(13), 8788-8795. doi:10.1074/jbc.M508135200

Yamada, M., et al. (2004). Overexpression of alpha-synuclein in rat substantia nigra results in

loss of dopaminergic neurons, phosphorylation of alpha-synuclein and activation of

caspase-9: resemblance to pathogenetic changes in Parkinson's disease. J Neurochem,

91(2), 451-461. doi:10.1111/j.1471-4159.2004.02728.x

Yamaguchi, H., et al. (2004). CHOP Is Involved in Endoplasmic Reticulum Stress-induced

Apoptosis by Enhancing DR5 Expression in Human Carcinoma Cells. Journal of

Biological Chemistry, 279(44), 45495-45502. doi:10.1074/jbc.M406933200

Yamamoto, A., et al. (2006). Autophagy-mediated clearance of huntingtin aggregates triggered

by the insulin-signaling pathway. J Cell Biol, 172(5), 719-731.

doi:10.1083/jcb.200510065

Yamamura, Y., et al. (1993). [Early-onset parkinsonism with diurnal fluctuation--clinical and

pathological studies]. Rinsho Shinkeigaku, 33(5), 491-496.

134

Yamasaki, S., et al. (2008). Mincle is an ITAM-coupled activating receptor that senses damaged

cells. Nat Immunol, 9(10), 1179-1188. doi:10.1038/ni.1651

Yanagi, S., et al. (2001). Syk expression and novel function in a wide variety of tissues. Biochem

Biophys Res Commun, 288(3), 495-498. doi:10.1006/bbrc.2001.5788

Yanagi, S., et al. (1995). The structure and function of nonreceptor tyrosine kinase p72syk

expressed in hematopoietic cells. Cellular Signalling, 7(3), 185-193.

doi:https://doi.org/10.1016/0898-6568(94)00088-S

Yanagida, T., et al. (2009). Oxidative stress induction of DJ-1 protein in reactive astrocytes

scavenges free radicals and reduces cell injury. Oxid Med Cell Longev, 2(1), 36-42.

Yang, L., et al. (1998). 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyride neurotoxicity is attenuated in

mice overexpressing Bcl-2. J Neurosci, 18(20), 8145-8152.

Yang, W., et al. (2009). Paraquat activates the IRE1/ASK1/JNK cascade associated with

apoptosis in human neuroblastoma SH-SY5Y cells (Vol. 191).

Yang, W. S., et al. (2015). Toll-like receptor 4/spleen tyrosine kinase complex in high glucose

signal transduction of proximal tubular epithelial cells. Cell Physiol Biochem, 35(6),

2309-2319. doi:10.1159/000374034

Yasuda, T., et al. (2010). The regulatory role of alpha-synuclein and parkin in neuronal cell

apoptosis; possible implications for the pathogenesis of Parkinson's disease. Apoptosis,

15(11), 1312-1321. doi:10.1007/s10495-010-0486-8

Yasuda, T., et al. (2013). alpha-Synuclein and neuronal cell death. Mol Neurobiol, 47(2), 466-

483. doi:10.1007/s12035-012-8327-0

Yepes, M. (2007). TWEAK and the central nervous system. Mol Neurobiol, 35(3), 255-265.

Yepes, M. (2013). TWEAK and Fn14 in the Neurovascular Unit. Frontiers in Immunology, 4,

367. doi:10.3389/fimmu.2013.00367

Yepes, M., et al. (2005). A soluble Fn14-Fc decoy receptor reduces infarct volume in a murine

model of cerebral ischemia. Am J Pathol, 166(2), 511-520. doi:10.1016/s0002-

9440(10)62273-0

135

Yepes, M., et al. (2006). Inhibition of TWEAK activity as a new treatment for inflammatory and

degenerative diseases. Drug News Perspect, 19(10), 589-595.

doi:10.1358/dnp.2006.19.10.1068005

Yi, Y.-S., et al. (2014). Functional Roles of Syk in Macrophage-Mediated Inflammatory

Responses. Mediators of Inflammation, 2014, 270302. doi:10.1155/2014/270302

Ying, H., et al. (2011). Syk Mediates BCR- and CD40-Signaling Intergration during B Cell

Activation. Immunobiology, 216(5), 566-570. doi:10.1016/j.imbio.2010.09.016

Yokota, T., et al. (2003). Down regulation of DJ-1 enhances cell death by oxidative stress, ER

stress, and proteasome inhibition. Biochem Biophys Res Commun, 312(4), 1342-1348.

Yokota, T., et al. (2004). Down regulation of DJ-1 enhances cell death by oxidative stress, ER

stress, and proteasome inhibition (Vol. 312).

Yokoyama, H., et al. (2010). Poly(ADP-ribose)polymerase inhibitor can attenuate the neuronal

death after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity in mice.

J Neurosci Res, 88(7), 1522-1536. doi:10.1002/jnr.22310

Yong, V. W., et al. (2004). The promise of minocycline in neurology. Lancet Neurol, 3(12), 744-

751. doi:10.1016/s1474-4422(04)00937-8

Yoon, J. Y., et al. (2013). Methanol extract of Evodia lepta displays Syk/Src-targeted anti-

inflammatory activity. J Ethnopharmacol, 148(3), 999-1007.

doi:10.1016/j.jep.2013.05.030

Yoshihara, E., et al. (2014). Thioredoxin/Txnip: Redoxisome, as a Redox Switch for the

Pathogenesis of Diseases. Frontiers in Immunology, 4(514).

doi:10.3389/fimmu.2013.00514

Yu, J., et al. (2014). Inflammasome activation leads to Caspase-1-dependent mitochondrial

damage and block of mitophagy. Proc Natl Acad Sci U S A, 111(43), 15514-15519.

doi:10.1073/pnas.1414859111

Yu, S. W., et al. (2006). Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-

induced cell death. Proc Natl Acad Sci U S A, 103(48), 18314-18319.

doi:10.1073/pnas.0606528103

136

Yu, X., et al. (2015). Protection of MPTP-induced neuroinflammation and neurodegeneration by

rotigotine-loaded microspheres. Life Sciences, 124, 136-143.

doi:https://doi.org/10.1016/j.lfs.2015.01.014

Yu, Y., et al. (2015). Inhibition of Spleen Tyrosine Kinase Potentiates Paclitaxel-Induced

Cytotoxicity in Ovarian Cancer Cells by Stabilizing Microtubules. Cancer Cell, 28(1),

82-96. doi:10.1016/j.ccell.2015.05.009

Zatloukal, K., et al. (2002). p62 Is a common component of cytoplasmic inclusions in protein

aggregation diseases. Am J Pathol, 160(1), 255-263. doi:10.1016/s0002-9440(10)64369-6

Zecca, L., et al. (2006). A proposed dual role of neuromelanin in the pathogenesis of Parkinson's

disease. Neurology, 67(7 Suppl 2), S8-11.

Zhang, F., et al. (2010). Resveratrol Protects Dopamine Neurons Against Lipopolysaccharide-

Induced Neurotoxicity through Its Anti-Inflammatory Actions. Molecular Pharmacology,

78(3), 466-477. doi:10.1124/mol.110.064535

Zhang, F., et al. (2012). Fluoxetine protects neurons against microglial activation-mediated

neurotoxicity (Vol. 18 Suppl 1).

Zhang, G., et al. (2014). Impulsive and Compulsive Behaviors in Parkinson’s Disease. Frontiers

in Aging Neuroscience, 6(318). doi:10.3389/fnagi.2014.00318

Zhang, H., et al. (2012). Regulation of striatal dopamine release by presynaptic auto- and

heteroreceptors. Basal ganglia, 2(1), 5-13. doi:10.1016/j.baga.2011.11.004

Zhang, J., et al. (2012). A novel retinoblastoma therapy from genomic and epigenetic analyses.

Nature, 481(7381), 329-334. doi:10.1038/nature10733

Zhang, W., et al. (2005). 3-Hydroxymorphinan is neurotrophic to dopaminergic neurons and is

also neuroprotective against LPS-induced neurotoxicity (Vol. 19).

Zhang, W., et al. (2005). Aggregated alpha-synuclein activates microglia: a process leading to

disease progression in Parkinson's disease (Vol. 19).

137

Zhang, X., et al. (2007). TWEAK-Fn14 pathway inhibition protects the integrity of the

neurovascular unit during cerebral ischemia. J Cereb Blood Flow Metab, 27(3), 534-544.

doi:10.1038/sj.jcbfm.9600368

Zhang, X. J., et al. (2013). Why should autophagic flux be assessed? Acta Pharmacol Sin, 34(5),

595-599. doi:10.1038/aps.2012.184

Zhang, Y., et al. (2014). Endothelial PINK1 mediates the protective effects of NLRP3 deficiency

during lethal oxidant injury. J Immunol, 192(11), 5296-5304.

doi:10.4049/jimmunol.1400653

Zhao, Y., et al. (2015). The NAIP–NLRC4 inflammasome in innate immune detection of

bacterial flagellin and type III secretion apparatus. Immunological Reviews, 265(1), 85-

102. doi:10.1111/imr.12293

Zheng, T. S., et al. (2008). No end in site: TWEAK/Fn14 activation and autoimmunity

associated- end-organ pathologies. J Leukoc Biol, 84(2), 338-347.

doi:10.1189/jlb.0308165

Zhong, Z., et al. (2016). Autophagy, NLRP3 inflammasome and auto-inflammatory/immune

diseases (Vol. 34).

Zhou, C., et al. (2008). Oxidative Stress in Parkinson's Disease: A Mechanism of Pathogenic and

Therapeutic Significance. Annals of the New York Academy of Sciences, 1147, 93-104.

doi:10.1196/annals.1427.023

Zhou, R., et al. (2011). A role for mitochondria in NLRP3 inflammasome activation. Nature,

469(7329), 221-225. doi:10.1038/nature09663

Zhu, J. H., et al. (2007). Regulation of autophagy by extracellular signal-regulated protein

kinases during 1-methyl-4-phenylpyridinium-induced cell death. Am J Pathol, 170(1),

75-86. doi:10.2353/ajpath.2007.060524

Zitka, O., et al. (2012). Redox status expressed as GSH:GSSG ratio as a marker for oxidative

stress in paediatric tumour patients (Vol. 4).

Zou, C. G., et al. (2009). The molecular mechanism of endoplasmic reticulum stress-induced

apoptosis in PC-12 neuronal cells: the protective effect of insulin-like growth factor I.

Endocrinology, 150(1), 277-285. doi:10.1210/en.2008-0794

138

Zou, W., et al. (2008). DAP12 couples c-Fms activation to the osteoclast cytoskeleton by

recruitment of Syk. Mol Cell, 31(3), 422-431. doi:10.1016/j.molcel.2008.06.023

Zuch, C. L., et al. (2000). Time course of degenerative alterations in nigral dopaminergic

neurons following a 6-hydroxydopamine lesion. J Comp Neurol, 427(3), 440-454.

139

CHAPTER 2. INVOLVEMENT OF MITOCHONDRIA MEDIATED OXIDATIVE

STRESS DEPENDENT CELL SIGNALING EVENTS AND SYK TYROSINE KINASE

ACTIVATION IN TUMOR NECROSIS LIKE WEAK STIMULATOR OF APOPTOSIS

(TWEAK) INDUCED DOPAMINERGIC CELL DEATH.

To be Submitted to Journal of Biological Chemistry

Sri Kanuri1,2, Vivek Lawana1, Huajun Jin1, Vellareddy Anantharam1, Anumantha Kanthasamy1 and

Arthi Kanthasamy 1 *

Parkinson's Disorder Research Laboratory, Iowa Center for Advanced Neurotoxicology, Dept. of

Biomedical Sciences, Iowa State University, Ames, IA.

Abstract

Neuro-inflammation, mitochondrial dysfunction, and impaired clearance of aggregate

prone proteins are implicated in Parkinson's Disease (PD) pathogenesis; however, the mechanism

by which inflammatory mediators increase the vulnerability of dopaminergic neurons to oxidative

stress-induced apoptotic cell death remain poorly characterized. Therefore, the goal of the present

study was to investigate the cell signaling events that underlie TWEAK induced apoptotic cell

death using an in vitro dopaminergic cell culture model, N27 cells. Herein, we show that N27

cells express both TWEAK and its receptor Fn14 and that TWEAK-elicited dose dependent

apoptotic cell death in N27 cells. Exposure of N27 cells to TWEAK evoked dissipation of

mitochondrial membrane potential (MMP), suppression of GSH levels, activation of caspase-8 and

3 and concomitant up regulation of Phospho-Tau and Phospho-NF-kB P65 levels. Moreover, these

changes were accompanied by the down regulation of Phospho-AKT, P-GSK3Beta (Ser9) and

LC3 levels in TWEAK treated dopaminergic neuronal cells. Intriguingly, upregulation of TWEAK

1 Department of Biomedical Sciences, Iowa State University, Ames, IA 50010

2 Primary researcher and author

*To whom correspondence should be addressed. E-mail: [email protected]

140

was evidenced in MPP+ treated dopaminergic neuronal cells. Likewise TWEAK was upregulated

in the SNpc of MPTP treated mice. Consistent with the role of TWEAK in the induction of

oxidative stress response, pretreatment of N27 cells with varying concentration of quercetin, a

bioflavanoid abrogated TWEAK-induced apoptotic cell death. In a similar fashion NF-kB

inhibitor, SN50 ameliorated TWEAK-induced loss of dopaminergic cell viability. Together,

these data suggest that TWEAK exerts deleterious effects on dopaminergic neuronal survival via

aberrant activation of NF-kB and impaired mitochondrial function in an oxidative stress dependent

manner.

Keywords Tweak; Neurotoxicity; Mitochondrial dysfunction; GSK-3 beta; Tau phosphorylation;

Protesasome; Autophagy; NF-kB activation; Parkinsonism Disease.

Abbreviations: PD, Parkinson Disease; AD, Alzheimer Disease; Tweak, Tumor Necrosis Factor

Weak stimulator of apoptosis; TNF, Tumor Necrosis Factor; ALS, Autophagy Lysosomal

system; UPS, Ubiquitin Proteasomal system; SLE, Systemic Lupus Erythematosis; BBB, Blood

brain barrier; MMP, Mitochondrial membrane potential; LC3, Light Chain 3.

Introduction

PD is one of the most common neurodegenerative disease characterized by chronic

progressive dopaminergic neurodegeneration in substantia nigra. The underlying mechanisms that

predispose to demise of dopaminergic neurons are complex, but oxidative stress is hypothesized

to one important predisposing factor that accentuates dopaminergic neuronal death (Javier Blesa

et al., 2015; Vera Dias et al., 2013; Hwang, 2013; C. Zhou et al., 2008). The commonly molecular

mechanisms that perpetuate oxidative stress in PD include, dopamine metabolism, mitochondrial

dysfunction, iron metabolism, GSH depletion, ubiquitin-proteosome dysfunction and autophagy

141

dysfunction (Greenamyre et al., 2001; Hwang, 2013; Juarez Olguin et al., 2016; J. Lee et al., 2012;

K. L. Lim, 2007; M. S. Medeiros et al., 2016; Smeyne & Smeyne, 2013). The markers of oxidative

stress that are present in substantia nigra of post mortem PD cases includes decrease in

polyunsaturated fatty acids (PUFA), increase in malondialdehyde, 4-hydroxy-2-nonenal and

upregulation of nitrotyrosine (C. Zhou et al., 2008). DA metabolism can induce production of

superoxide and hydrogen peroxide radicals thereby contributing to oxidative stress and cell death

(Juarez Olguin et al., 2016; Miyazaki & Asanuma, 2008). Impairment of mitochondrial complex I

of ETC (Electron Transport Chain) that is commonly associated with PD results in upregulation

of ROS production, oxidative stress and neuronal death(Greenamyre et al., 2001; Keeney et al.,

2006). Ubiquitin proteasome system dysfunction can cause accumulation of misfolded,

aggregated, damaged proteins ultimately resulting in oxidative stress and contributing to neuronal

demise (K. L. Lim, 2007). GSH depletion is noticed in 40% of postmortem substantia nigra (SN)

tissues of PD patients (Smeyne et al., 2013). Excessive iron accumulation in PD can lead to

production of oxygen radicals through fenton reaction which ultimately leads to lipid peroxidation,

oxidative stress and dopaminergic cell death (Poetini et al.). Accordingly, GSH deficiency results

in excessive ROS production, oxidative stress and dopaminergic neurodegeneration in PD

(Smeyne et al., 2013). Dysfunctional autophagy process will hasten the accumulation of damaged,

oxidized mitochondria thereby leading to excess ROS production and oxidative stress induced

demise of dopaminergic neurons in PD (J. Lee et al., 2012).

PKC-delta is a novel PKC isoform which is widely expressed in various cells and tissues

(Kikkawa et al., 2002). It is involved in numerous physiological cell functions like growth,

differentiation and apoptosis (Kikkawa et al., 2002). Members of PKC family usually have an N-

terminal regulatory region and a C-terminal catalytic region (Newton, A.C. et al 1995). It is

142

activated via generation of catalytic active fragment or through tyrosine phosphorylation (Kaul et

al., 2005; Kikkawa et al., 2002). Dopaminergic neurotoxins like MPP+ and TNF-alpha can activate

the PKC-delta by proteolytic cleavage ultimately resulting in dopaminergic neurodegeneration in

cell culture and animal models of PD (Richard Gordon et al., 2012; Kaul et al., 2003). Caspase-3

induced cleavage of proapoptotic protein PKC-delta into its active form ultimately results in

dopaminergic neurodegeneration in MPP+ and Dieldrin treated N27 cells (Kanthasamy et al.,

2006; Kanthasamy et al., 2008; Y. Yang et al., 2004). Furthermore, H2O2 induced tyrosine-311

phosphorylation of PKC-delta leads to its activation of its catalytic fragment and results in

dopaminergic neurodegeneration in cell culture model of PD (Kaul et al., 2005). Another report

implicates the role of PKC-delta in NOX-1 expression, oxidative stress and dopaminergic

neurodegeneration with paraquat treatment in cell culture model of PD (Cristovao et al., 2013).

Additional studies done indicate that, PKC-delta can lead to downregulation of tyrosine

hydroxylase activity and dopamine synthesis by upregulation of protein phosphatase-2A in

dopaminergic neurons in cell culture and animal models of PD (D. Zhang et al., 2007). LPS

induced stimulation of primary microglial cells resulted in PKC delta upregulation which

subsequently resulted in dopaminergic neurodegeneration in experimental models of PD (R.

Gordon et al., 2016). Accordingly, previous studies done with resveratrol had demonstrated the

dopaminergic neuroprotection by altering PKC-delta signaling apoptotic signaling pathway and

downregulating microglial activation (Richard Gordon et al., 2010).

Tumor necrosis factor like weak stimulator of apoptosis (Tweak) is a recently

discovered cytokine belonging to TNF superfamily. Tweak is 262 amino acid type II

transmembrane protein, whereas its major receptor, Fn14 gene is located on chromosome17p13,

which is very close to P53 tumor suppressor gene (Burkly et al., 2007; Chicheportiche et al., 1997;

143

Marsters et al., 1998). Cell associated Tweak exists in membrane bound form (30-35 kDa) and

soluble form (18kDa) (Chicheportiche et al., 1997; De Ketelaere et al., 2004). All the membrane

bound forms can be cleaved by specific proteases to a soluble form that can exert its actions in the

neighboring cells and distant targets (Trebing et al., 2014). Tweak expression is highest in heart,

pancreas, colon, small gut, lung, ovary, prostate, spleen, lymph nodes, and appendix

(Chicheportiche et al., 1997). Tweak exerts many of its actions via Fn14 activation (Burkly et al.,

2011; Inoue et al., 2000). Tweak / Fn14 interaction appears to be involved in angiogenesis,

inflammation, proliferation, migration, cytokine production, cytotoxicity and apoptosis (Burkly et

al., 2007; Inoue et al., 2000). Tweak / Fn14 interactions were implicated in pathogenesis of

Rheumatic arthritis, SLE and skeletal muscle disorders (Burkly et al., 2007; Trebing et al., 2014).

Tweak also appears to be involved in various neurological conditions like stroke, neuro-

inflammatory diseases and multiple sclerosis (Burkly et al., 2007). It is quite possible that, Tweak

/ Fn14 pathway can be a potential target for developing therapeutic interventions in systemic

diseases, autoimmune conditions and cancers.

Spleen tyrosine kinase (Syk) in involved in neuronal growth and differentiation

(Tsujimura et al., 2001). Numerous studies were done previously to implicate syk in neuro-

inflammation after subarachnoid hemorrhage in rats (Yue He et al., 2015; Y. He, L. Xu, B. Li, Z.

N. Guo, et al., 2015). Defective TREM2 receptor induced Syk mediated down signaling events

leads to defective microglial phagocytosis, accumulation of amyloid plaques and neuronal cell

death in Alzheimer disease (Dempsey, 2015). Syk mediated signaling events are responsible for

neuro-inflammation, gliosis, amyloid beta accumulation and tau phosphorylation in Alzheimer

disease (D. Paris et al., 2014). In microglial cells, spleen tyrosine kinase is recruited to the stress

granules leading to phagocytic dysfunction and ROS & NOS production ultimately resulting in

144

neuronal cell death (S. Ghosh et al., 2015; Wolozin et al., 2015). Previous findings indicate that,

phosphorylated-syk expression is increased in neurodegenerative diseases like Alzheimer disease

and Nasu-Hakola Disease (Satoh et al., 2012; Schweig et al., 2017). Recent report suggests that

tyrosine phosphorylated Syk is found in neurofibrillary tangles of hippocampal neurons in mouse

model of Alzheimer disease that providing evidence for linking to spleen tyrosine kinase to

neurodegeneration (Köhler et al., 2017). Increased Syk expression is associated with tau

phosphorylation, amyloid beta accumulation and pathological lesions of Alzheimer disease (Satoh

et al., 2012; Schweig et al., 2017). Fc receptor mediated Syk activation is also responsible for

downstream signaling events like cytokine secretion, phagocytosis and antigen presentation in

Alzheimer Disease (Fuller et al., 2014). Recently, it was reported that, alpha-synuclein can be

phosphorylated by spleen tyrosine kinase thus implicating its role in the pathogenesis of Parkinson

Disease (Lebouvier et al., 2008). Syk induced tyrosine phosphorylation of alpha-synuclein can be

neuroprotective by decrease oligomerization and aggregation (A. Negro et al., 2002).

Although there were several studies relating Tweak / Fn14 pathway with neuro-

inflammatory diseases, stroke and neurological malignancies, thus far its role in neurodegenerative

diseases has not been vigorously investigated. However, thus far there are very few studies thus

far examining the involvement of Tweak /Fn14 pathway in in neurodegenerative disorders like

PD. Our goal is to identify the role of this Tweak / Fn14 pathway in the pathogenesis of Parkinson’s

disease. Therefore, in the present study we sought to investigate the role of TWEAK in

dopaminergic neurotoxicity a using in vitro cell culture models of PD and to further confirm and

extend our in vitro data in an MPTP mouse model of PD to identify the signaling mechanisms of

dopaminergic neurodegeneration. Here we report that TWEAK treatment-induce oxidative stress,

mitochondrial dysfunction and NF-kB activation and that inhibition of NF-kB activation and

145

abrogation of mitochondrial dysfunction is associated with neuroprotection. Our results suggest

that TWEAK activation might be a critical intermediate in PD associated dopaminergic neuronal

demise and that alteration of downstream signaling mechanisms may be conducive for the

successful dismantling of the dopaminergic neuronal cells. Therefore, precise understanding of

pathogenic mechanisms and identification of new molecular targets pertaining to Tweak / Fn14

pathway would be helpful in designing novel therapeutic strategies that might attenuate or reverse

the disease progression and afford symptomatic relief in PD patients.

Materials and Methods

Chemicals and Reagents

Cell culture supplies including RPMI 1640 media, penicillin/streptomycin, L-glutamine,

fetal bovine serum were purchased from Invitrogen. Recombinant Tweak was obtained from

Pepro-Tech technologies. Caspase-3, 8, 9 & 2 substrates and MTT assay kit were purchased from

Sigma technologies, St. Louis, MO. NF-kB inhibitor (SN-50) and Quercitin were purchased from

Life Technologies. Mitochondrial membrane potential JC-1 dye was obtained from Molecular

Probes, Eugene, OR. GSH assay kit was purchased from Cayman chemicals. Tweak antibody was

purchased from Santa Cruz Biotechnology, Dallas, TX. β-actin antibody was obtained from Sigma

technologies, St. Louis, MO. LC3A/B, Beclin , Fn14, Phospho-AKT, GSK-3 Beta total, GSK-3

Beta Tyrosine 216, GSK-3 Beta Serine 9 and Phospho-Tau were purchased from Cell Signaling

Technology, MA, USA.

Cell culture and Treatment

Rat mesencephalic dopaminergic cell line, generally referred to as N27 dopaminergic

cells, was a generous donation from Dr. Kedar N. Prasad (University of Colorado Health Sciences

146

Center, Denver, CO). N27 cells has been used for studying the neurotoxic mechanisms related to

Parkinson Disease in our laboratory previously (Kanthasamy et al., 2006; Kaul et al., 2003). N27

cells were grown in RPMI 1640 medium supplemented with 10% fetal bovine serum, 2mM L-

glutamine, 50 units penicillin and 50μg/ml streptomycin. Cell cultures were maintained under 5%

CO2 at 37°C as previously described (Y. Yang et al., 2004). MN9D cell line (Fusion of ventral

mesencephalic and neuroblastoma cells) was a kind gift from Dr. Syed Ali (National Center for

Toxicological Research/FDA, Jefferson, AR). MN9D cells were grown in Dulbecco’s modified

Eagle’s medium supplemented with 10% fetal bovine serum, 2 mM L-glutamine, 50 U penicillin,

and 50 units streptomycin. N27 cells were stimulated with Tweak 100 ng for 3h, 6h, 12h and 24h

respectively. Post treatment, N27 cells were collected for caspase assay, mitochondrial membrane

potential, proteosomal assay and western blot experiments respectively.

Animal Studies.

Wild type C57 black mice were house under standard conditions at constant temperature

(22 C), 30% relative humidity with 12h light cycle as per IACUC protocol with ad libitum access

to food and water. 6-8 old week mice were used in this study. The well characterized acute MPTP

mouse model is used in this study (G. E. Meredith & D. J. Rademacher, 2011; Przedborski et al.,

2004). The mice in the MPTP treatment group received 4x 18 mg/kg of MPTP every 2h and the

control mice received equivalent saline injections. The mice were sacrificed at 3h 6h 12h 24h and

72h after the last MPTP injection. Later substantia nigra tissues were dissected out and tissues

lysates were immunoblotted against protein of interest.

MTT Assay

Cell death was determined by MTT (3-(4,5-dimethylthiazol-3-yl)-2,5-diphenyl tetrazolium

bromide) assay, in which the activity of mitochondrial dehydrogenase enzymes in cleaving

147

tetrazolium ring to formazan crystals was assessed (Afeseh Ngwa et al., 2009; Kitazawa et al.,

2001; Sun et al., 2006; Sun et al., 2009). After Tweak treatment, cells were incubated in serum-

free medium containing 0.25 mg/ml MTT for 1 hr at 37°C. Supernatant was removed and MTT

formazan crystals were solubilized with 200 ul dimethyl sulfoxide (DMSO). Formation of

formazan from tetrazolium was measured at 570 nm with a reference wavelength at 630 nm using

a Spectra-Max microplate reader (Molecular Devices, Sunnyvale California).

Sytox Assay

The assessment of cell viability is done through Sytox Green dye (membrane impermeable

dye) that enters the dead cells and intercalates with DNA and produces fluorescence (Afeseh Ngwa

et al., 2009; Latchoumycandane et al., 2005). After Tweak treatment, N27 cells grown in 24 well

plate were incubated with 1 uM Sytox Green dye. DNA bound Sytox green dye produces green

fluorescence that can be detected at an excitation wavelength of 485 nm and emission wavelength

of 538 nm with the help of fluorescent plate reader (Bio-tek microplate reader)(H. Jin et al., 2011;

Sherer et al., 2007). Sytox stained cells can be observed using Nikon TE 200 fluorescence

microscope equipped with SPOT digital camera.

Western Blot Analysis of Whole Cells Lysates

Treated cells were collected and lysed with RIPA buffer (Sigma) in ice and sonicated for

15 seconds on ice for each sample. Samples were centrifuged at 14,000 g for 15 min at 4°C.

Supernatants were collected and stored at −80°C until use. Protein estimation was done using

Bradford reagent and samples with equal amount of protein were separated on 8–15% SDS-PAGE,

depending on the molecular weight of the target protein. Later they were transferred onto

nitrocellulose membranes by electro-blotting at 110V for 90 min at 4 C (Ice-cold transfer). Next,

membranes were blocked briefly for 1h by incubating them in blocking buffer and later incubated

148

with primary antibodies overnight at 4°C. To ensure equal protein loading, β-actin (1:10,000) was

used as loading control. Membranes were washed several times and incubated with specific

fluorescent labelled secondary antibody for 1 h at room temperature. Membranes were scanned

using the Odyssey IR Imaging system (LICOR) and data were analyzed with Odyssey 2.0 software

(Kanthasamy et al., 2006; Kitazawa et al., 2001; Sun et al., 2006; Sun et al., 2009).

Measurement of Mitochondrial Membrane Potential (MMP).

Changes in the mitochondrial membrane potential was detected by 5,5’, 6.6’- tetrachloro-

1,1’, 3,3’-tetraethyl-benzimidazolcarbocyanineidide (JC-1 dye) (Molecular Probes, Thermo Fisher

Scientific) a cataionic fluorescent lipophilic dye. After treatment, cells were exposed with 2 ug/ml

of JC-1 dye for 20 min at 37C in a 24 well plate. Later, cells were incubated with 200 uL of 10

mM Tris-HCL containing 0.1% Triton X-100 for 30 min on an eppendorf tube shaker. Samples

were then transferred to 96-well plates and measured with fluorescence plate reader. Healthy

mitochondria form J-aggregates which displays strong red fluorescence at excitation and emission

wavelengths at 560 and 595 nm, respectively. In depolarized mitochondria, JC-1 exists as J

monomers which shows strong green fluorescent intensity with excitation and emission

wavelength at 485 and 535 nm respectively. The shift in ratio of fluorescent intensity of JC-1

aggregates (Red fluorescence) to fluorescent intensity of monomers (green fluorescence) is used

as an indicator of depolarization of mitochondrial membrane potential (M. Lin et al., 2012).

Proteasomal Assay

The UPS (Ubiquitin Proteosomal System) degradation was measured by monitoring the

proteasome activity as described in our previous publications (Sun et al., 2006; Sun et al., 2009).

Briefly, after treatment cells were re-suspended in buffer containing 50mM Tris–HCl, 1mM EDTA

and 10mM EGTA, and incubated at 37°C for 20 min. After incubation, lysates were centrifuged

149

at 16,000 × g for 10 min. Supernatants were collected and incubated with fluoregenic substrate,

50μM Suc-LLVY-AMC, for determination of proteasomal activities. After 1 hour incubation with

the substrate, samples were analyzed with fluorescence plate reader (excitation 380 nm and

emission 460 nm) (Gemini Plate Reader, Molecular Devices Corporation) (M. Lin et al., 2012).

GSH assay

The mono-chlorobimane fluorometric method was used to determine the cellular GSH

levels. After treatment, cells were collected, washed with PBS, and sonicated in a lysis buffer

(50 mM Tris, pH 7.4, 5 mM EDTA and 0.001% 3, 5-di-tert-butyl-4-hydroxytoulene (BHT).

Samples were centrifuged at 14,000 g for 10 minutes at 4°C. 1 mM of mono-chlorobimane and

10 U/mL of glutathione S-transferase in 50 mM Tris, pH 7.4, were dissolved and added to the

resulting supernatant. 200 μL of the mixture was transferred to a black 96-well plate and incubated

at 24°C for 30 minutes. The fluorescence of samples was measured using a spectramax microplate

reader (Molecular Devices Corp., Sunnyvale, CA) with excitation at 485 nm and emission at

645 nm (Chandramani Shivalingappa et al., 2012)

Caspase Enzymatic Activity Assay

Caspases activity was measured using specific fluorescent substrates namely, caspase-2

(Z-VDVAD-AFC), caspase-3 (Ac-DEVD-AFC), caspase-8 (Ac-VETD-AMC) and caspase-9 (Ac-

LEHD-AMC). Briefly, after treatment, cells were lysed with lysis buffer (50mM Tris-HCl, 1mM

EDTA, and 10mM EGTA) containing 10mM digitonin for 30 mins at 37 °C. Lysates were

centrifuged at 10,000 × g for 5 mins and cell-free supernatants were collected. Cell free

supernatants were incubated with a 50 uM specific fluorescent substrate at 37 °C for 1 h. Caspases-

2, 3, 8 and 9 activity was then measured using SpectraMax Gemini XS Microplate Reader

(Molecular Devices, Sunnyvale, CA) with excitation at 400 nm and emission at 400 nm. The

150

caspase-3, 8 and 9 activities were calculated as fluorescence units per milligram of protein (M. Lin

et al., 2012).

Immunohistochemistry

Double Immuno-labelling studies in the Substantia Nigra sections were performed as per

previously published protocols (Richard Gordon et al., 2012; M. Lin et al., 2012) Briefly, sections

were grown on poly-d-lysine-coated coverslips and treated 24-48 h later. At the end of treatments,

sections were fixed with 4% PFA, washed in PBS for three times. Later sections were incubated

in blocking buffer (PBS containing 2% BSA, 0.5% Triton X-100 and 0.05% Tween 20) for 1 h at

room temperature. Next, the sections were incubated with respective primary antibodies (Tweak)

diluted in 5% serum solution overnight at 4°C. Samples were then washed several times in PBS

and incubated with Alexa-488 and -555 dye-conjugated secondary antibodies for 1h at room

temperature. After several washes, sections were dehydrated by rising concentration of ethanol

followed by xylenes and coverslips were mounted using the Prolong Gold antifade reagent

(Molecular Probes. The formation of immune-complexes were visualized using Nikon inverted

fluorescence microscope equipped with an X100 oil immersion objective and images were

captured with SPOT digital camera (R. Gordon et al., 2016).

Data Analysis

Results are presented (PRISM software, Graph Pad, San Diego) as fold induction as

compared with control group. Results represent mean ± S.E.M. Statistical analysis was

performed by using one-way ANOVA followed by Student Newman Keuls post hoc test

(PRISM software) in order to compare between groups. P values < 0.05 were considered

significant.

151

Results

Expression Pattern of Tweak in the Mouse Brain.

Expression of both TWEAK and Fn14 expression has been detected in both neurons and

astrocytes; however astrocytes displayed higher levels of TWEAK expression while Fn14

expression was found to be more prominent in neurons (Yepes et al., 2005). Previous studies

have demonstrated the role of Tweak / Fn14 pathway in experimental models of various

neurological disorders including cerebral ischemia, multiple sclerosis, Encephalitis and Parkinson

Disease (Burkly et al., 2007; Mustafa et al., 2016a; Jeffrey A. Winkles, 2008), therefore, we

characterized the expression profile of membrane and soluble Tweak in various brain regions.

Tissue expression profiling as assessed via WB analysis revealed constitutive expression of

membrane bound full-length TWEAK in the hippocampus and olfactory bulb whereas prominent

expression of cleaved soluble TWEAK was evidenced in the striatum and substantia nigra.

Intriguingly cerebellum expressed very low levels of both soluble and native TWEAK. Taken

together pronounced expression of soluble TWEAK levels in the striatum and substantia nigra

suggest that TWEAK may be a key regulator of dopaminergic neuronal survival in these brain

regions. Indeed, TWEAK neutralizing body was found to rescue dopaminergic neurons in a sub-

acute MPTP mouse model further supporting the deleterious effects of TWEAK on dopaminergic

neuronal integrity (Mustafa et al., 2016a).

Tweak and Fn14 are Expressed in Dopaminergic Neurons in Culture

Tweak has been shown to be expressed in diverse cells including neurons, astrocytes and

microglia (Inoue et al., 2000). Given that rat dopaminergic neuronal (N27) cells and ventral

mesencephalic and neuroblastoma (Mn9D) cells are routinely used for the investigation of cell

signaling events underlying PD-toxin induced dopaminergic neurotoxicity we initially determined

152

the Tweak protein expression levels in rat dopaminergic neuronal (N27) cells and ventral

mesencephalic and neuroblastoma (Mn9D) cells. Immunoblot analysis revealed marked

expression of soluble and relatively low levels of expression of membrane bound full-length

Tweak in N27 cells (Fig 2A). On the contrary, we found that there is increased membrane bound

full-length and relatively low Soluble levels of Tweak expression in Mn9D cells.

Tweak and its receptor Fn14 are abundantly expressed in Neurovascular Unit comprising

of perivascular astrocytes, pericytes and neurons (M. Yepes, 2013). Since Tweak’s action is

mediated via Fn14 receptor, we decided to determine its expression levels in dopaminergic

neuronal cells using immunoblot analysis. Our data revealed an inverse correlation between Fn14

expression levels and concentrations of TWEAK treatment. Specifically, increased expression of

Fn14 was observed at lower doses of Tweak (30ng) whereas decreased Fn14 expression was

observed at higher doses of Tweak (50ng and 100 ng) (Fig 2B) suggesting that TWEAK may be a

key regulator of the signaling mechanisms that are activated under injury conditions.

Upregulation of Tweak Levels in the Serum and CSF Samples of PD Patients.

Targeting Tweak/Fn14 pathway can be helpful for developing therapeutic interventions in

systemic and neurological disorders (Desplat-Jego et al., 2002; Yepes et al., 2005). Monitoring

CSF and serum Tweak levels may provide further information on the diagnostic and prognostic

potential of this inflammatory chemokine. In this context serum Tweak levels were found to be

elevated within 24 hrs following stroke onset (Inta et al., 2008). It is plausible that serum Tweak

levels can fluctuate with severity of neurodegenerative diseases and characterizing their levels

might be beneficial in monitoring disease severity and determining therapeutic efficacy of

antiparkinsonian drugs. For this purpose, we determined serum Tweak levels in human PD

patients and compared them with age- matched control subjects. We obtained human serum

153

samples from Human Brain and Spinal Resource Center (VA Greater Los Angeles Health care

system). Serum samples were collected from PD patients (n=4) and healthy controls (n=4). The

levels of TWEAK were assessed via WB analysis of patients’ samples. Western blotting analysis

revealed a 2-3-fold increase in TWEAK levels in the serum of PD patients compared to age-

matched controls (Fig 3A). Likewise, a marked increase in the expression of TWEAK (controls-

4; PD samples-8) was evidenced in the CSF of PD patients as compared to age matched control

subjects (Fig3B). Further longitudinal studies should be conducted to determine whether

expression levels of TWEAK can be used to predict motor dysfunction or cognitive decline in PD

patients.

Impact of a Parkinsonian Toxin on TWEAK Expression in Dopaminergic Neuronal Cells

MPP+ is the active metabolite of MPTP that crosses the blood brain barrier and causes

death of dopaminergic neurons (Sawada et al., 2006). MPP+ is a Parkinsonian toxin that causes

distinct pattern of cell death characterized by apoptosis and necrosis in PC12 cells, cerebellar

granule cells & mesencephalic dopaminergic neurons (Sawada et al., 2006). Therefore we

evaluated whether the MPP+ altered the levels of TWEAK in N27 dopaminergic cells. N27cells

were exposed to MPP+ (300 uM) for varying duration (6, 12 & 24h). At the end of the

corresponding time points cell lysates were prepared and Tweak expression was determined using

WB analysis. Our results revealed that MPP+ induces a time dependent upregulation of Tweak in

N27 cells suggesting that TWEAK induction might be linked to dopaminergic neurotoxicity (Fig

4A).

Next, we validated our findings in an in vivo MPTP mouse model of PD. Administration

of MPTP causes selective degeneration of nigrostriatal dopaminergic neurons and leads to

pathologic changes similar to Parkinson’s disease (Sawada et al., 2006). Acute MPTP model is

154

widely used to study neurodegeneration in nigrostriatal dopaminergic system in PD (N. Panicker,

H. Saminathan, H. Jin, M. Neal, & D. S. Harischandra, 2015). Recently, it was reported that

blocking Tweak / Fn14 pathway can attenuate the dopaminergic neurodegeneration in subacute

MPTP model (Mustafa et al., 2016a). With previous studies implicating the role of Tweak/Fn14

pathway in dopaminergic neurodegeneration, we next investigated whether there is an

upregulation of Tweak expression in the substantia nigra of mice subjected to acute MPTP

treatment (4x 18 mg/kg of MPTP every 2h and sacrificed at 3h 6h 12h 24h and 72h after the last

MPTP injection). Using immunoblotting studies, we found out that TWEAK levels in the mouse

substantia nigra were significantly upregulated following MPTP treatment. A time course study

revealed that the maximal TWEAK expression occurs at 3-6 h following MPTP treatment

suggesting that TWEAK may serve as an early biomarker when no motor deficits are observed

(Fig 4B).

Furthermore, we confirmed our WB data using IHC studies using an MPTP mouse model

of PD. We performed IHC studies to determine the levels of TWEAK expression in tyrosine

hydroxylase (TH) positive neurons in substantia nigra using a sub-acute MPTP model of PD. In

this model, C57black mice were injected with 25 mg/kg of MPTP once daily for 5 consecutive

days and sacrificed at 3 days after the last MPTP injection. Brains were harvested and successive

30 μm ventral midbrain sections were processed for Tweak expression. Consistent with WB

findings, immune-histochemical analysis further validated pronounced expression of TWEAK in

the TH positive neurons in the substantia nigra in MPTP treated mice suggesting that TWEAK

may serve as a biomarker for surviving dopaminergic neurons in the PD brain (Fig 4C).

155

Tweak-induces Concentration-Dependent Cell Death in N27 Dopaminergic Neuronal Cells

Tweak has been shown to elicit cytotoxic effects in human peripheral blood monocytes,

human natural killer cells, murine cortical neurons, murine renal cortical cell lines, murine

mesangial cell lines and murine mammary epithelial cell lines (Burkly et al., 2011). We initially

determined whether TWEAK impacts dopaminergic neuronal viability using the well

characterized N27 dopaminergic neuronal cell culture model. N27 cells were exposed to increasing

concentration of Tweak and TNF 30 ng was used as a positive control. At the end of 24h following

drug treatment (Tweak or TNF), cell viability was assessed by MTT analysis. Our results indicate

that, Tweak at 50, 75, and 100 ng induced 10%, 30% 25% apoptotic cell death in N27 cells

indicating that it induces concentration dependent cytotoxicity in N27 dopaminergic neuronal cells

(Fig 5A). We further validated TWEAK induced dopaminergic neurotoxicity using the Sytox

staining. Sytox green dye uptake occurs following disruption of plasma membrane, usually in

apoptotic dying cells. As anticipated both MPP + (300 uM) and TWEAK (150 ng) treated N27

cells exhibited remarkable dye uptake as compared to vehicle treated cells further confirming the

cell death inducing effects of TWEAK on dopaminergic neuronal cells (Fig 5B).

Effect of Tweak on Mitochondrial Membrane Potential (MMP) and GSH

evels in N27 Cells

Tweak has been shown to alter mitochondrial membrane potential in endometrial cells and

HT29 adenocarcinoma cell lines (Sabour Alaoui et al., 2012; Wilson & Browning, 2002). It is well

established that mitochondrial dysfunction and ROS production initiates a cascade of downstream

signaling events that eventually leads to in caspase dependent apoptotic neuronal cell death (S.

Maharjan et al., 2014). Since TWEAK-induced apoptotic cell death in N27 cells we hypothesized

that mitochondrial dysfunction might contribute to dopaminergic neurotoxicity. Therefore, we

first we sought to determine whether mitochondrial dysfunction is an important upstream signaling

156

event in Tweak induced apoptotic cell death in N27 dopaminergic neuronal cells. We treated N27

dopaminergic neuronal cells with Tweak for increasing duration and measured the mitochondrial

membrane potential (MMP) using the cationic lipophilic JC-1 dye. As shown in Fig 6A, a time

dependent increase in the collapse of MMP (depolarization of ΔΨm) was observed in Tweak

treated N27 dopaminergic neuronal cells. Next, we examined whether the Quercitin (Flavonoid

antioxidant) can protect dopaminergic cells from Tweak-induced mitochondrial depolarization.

N27 dopaminergic cells were pretreated for 1 hour with Quercitin (3 uM) and then subsequently

exposed to Tweak. Tweak-induced dissipation of MMP in N27 cells was attenuated in the presence

of Quercitin at later time points (12h and 24h) (Fig 6B) suggesting that mitochondrial dysfunction

may serve a critical determinant of TWEAK-induced apoptotic cell death.

Several neurological disorders are characterized by dramatic reductions in GSH levels

thereby sensitizing neurons to mitochondria associated oxidative stress insults (Mari et al., 2009).

Therefore, assessing total cellular GSH levels will provide us a partial glimpse of the antioxidant

status in TWEAK treated cells. N27 dopaminergic neuronal cells were exposed to Tweak or TNF

for 24h. At the end of the incubation period total cellular GSH levels were determined by the

mono-chlorobimane fluorometric method. As shown Fig 6 C, intracellular stores of GSH levels

were depleted by 25% at the end of 24 hours following Tweak or TNF alpha treatment. Taken

together, our studies demonstrate that mitochondrial dysfunction coupled with GSH depletion may

initiate the deleterious cascade of downstream signaling events that eventually leads to apoptotic

cell death in Tweak stimulated N27 dopaminergic neuronal cells.

Tweak-Mediated Activation of Caspase-2,3, 8 and Caspase-9 in N27 Cells

In human endometrial cells, Tweak causes apoptotic cell death through activation of

caspases 3, 7 and 9 (D. Wang et al., 2010). Caspase 2 has been linked to apoptotic cascade

157

mechanism in several culture cells (S. Kumar, 2009; L. Wang et al., 1994). In fact knock out of

caspase-2 gene was found to confere resistance against MPTP-induced dopaminergic

neurotoxicity (Tiwari et al., 2011) Mitochondrial dysfunction is commonly associated with

caspase dependent neuronal death (Maharjan, S. et al 2014). Proteosomal impairment leads to

caspase-3 dependent programmed cell death in the cultured cortical neurons (Tuffy et al., 2010).

Therefore, we sought to investigate whether caspase activation occurs prior to apoptotic cell death

in Tweak stimulated N27 cells. N27 neuronal cells were exposed to Tweak for increasing duration

(3-24h). At the end of the treatment period caspase activity was measured with the help of

Fluorogenic substrates for caspase 2 (Ac-VDVAD-AFC) caspase 3 (Ac-DEVD-AMC), 8 (Ac-

LEHD-AFC) and caspase 9 (Ac-LEHD-AMC). Exposure to 100 ng Tweak resulted in a time

dependent increase in caspase 2,3,8 and 9 enzymic activity in N27 cells (Fig 7A-D). In fact,

maximal induction of caspase activity was observed at 24h post Tweak treatment in N27

dopaminergic neuronal cells. Together our studies suggest that caspase activation might be linked

to TWEAK-induced dopaminergic neurotoxicity.

Impaired Ubiquitin Proteasome system (UPS) and Auto-lysosomal system (ALS) in Tweak

treated N27 cells.

In neurodegenerative diseases, autophagy impairment leads to accumulation of damaged

organelles and pathogenic proteins thereby leading to neuronal cell death. In PD, impairment of

mitophagy leads to accumulation of damaged mitochondria and caspase mediated apoptotic cell

death in neurons (R. A. Nixon & D. S. Yang, 2012). Alpha synuclein induced blockage of

chaperone mediated autophagy (CMA) prevents its uptake and clearance by lysosomes ultimately

resulting in pathological consequences in PD (R. A. Nixon et al., 2012). Measuring the levels of

auto-phagosomal membrane associated LC3 II using western blot analysis is a reliable method of

determining autophagic activation in the cell (Tanida et al., 2008). Therefore, we sought to

158

investigate whether autophagy alteration is associated with Tweak induced cell death in N27

dopaminergic neuronal cells by immunoblot analysis. N27 cells were exposed to TWEAK for

varying duration (3h, 6h, 12h and 24h), cell lysates were prepared at the end of the indicated time

period and LC3I and II levels were determined using WB analysis. We observed time dependent

decrease in LC3 II levels in N27 dopaminergic cells following exposure to Tweak (Fig 8A).

Beclin1 dysfunction is associated with neurodegenerative disorders and cancers (Kang et al.,

2011). Since beclin1 is regarded as an important factor for regulating autophagy, we next

proceeded to determine the beclin1 levels in Tweak treated N27 cells by immunoblot analysis.

N27 cells were exposed to Tweak for varying time points (3h, 6h, 12h and 24h). Post incubation,

cell lysates were prepared and beclin1levels were determined using WB analysis. Similar to LC3

II levels we observed a time dependent decrease in beclin1 expression in N27 dopaminergic cells

exposed to Tweak (Fig 8B). Thus, our studies raise the possibility that impairment in autophagic

mechanism might partially contribute to TWEAK-induced apoptotic cell death partly via

accumulation of aggregate prone proteins and damaged organelles.

Previously we demonstrated using N27 cells that proteosomal inhibition leads to

inadequate clearance and accumulation of ubiquitin positive protein aggregates within the cytosol

(Sun et al., 2005; Sun et al., 2006; Sun et al., 2007). Proteasomal dysfunction also activates caspase

dependent and caspase independent apoptosis inducing pathways in neurons (Tuffy et al., 2010).

Recent studies suggest that proteasomal inhibition promotes programmed cell death in

proliferating endothelial cells (Drexler et al., 2000). Since ubiquitin-proteasome system is

primarily involved in cellular protein degradation, assessing proteosomal activity will provide us

information regarding cellular proteostasis. Therefore, we investigated the proteosomal activity in

Tweak treated N27 dopaminergic neuronal cells. N27 cells were exposed to Tweak for increasing

159

time period. At the end of the indicated time points (3h, 6h, 12h and 24h), cell lysates were

prepared for the assessment of proteosomal function. As shown in Fig 8 C, a time-dependent

decrease in proteasomal activity was observed in N27 dopaminergic neuronal cells exposed to

Tweak (100 ng). In this context, proteasomal activity was reduced as early as 3 h following Tweak

treatment, and it steadily declined in a time dependent fashion. Taken together, impairment in the

cellular degradation machinery can be regarded as a key event that predisposes dopaminergic

neurons to TWEAK-induced apoptotic cell death.

Tweak Induced NF-kb Activation in N27 Cells

Tweak has been shown to activate both canonical and non- canonical NF-kB signaling

pathways (Saitoh et al., 2003). Activation of NF-kB pathway by Tweak has been demonstrated in

glioblastoma cells (Tran et al., 2005), keratinocytes (L. Jin et al., 2004), and renal tubular cells (A.

B. Sanz et al., 2010), and neurons (Polavarapu et al., 2005). Persistent activation of NF-kB

signaling pathway has been identified as a critical signaling event in chronic inflammatory diseases

(Hoesel & Schmid, 2013). Exposure of PC12 cells to 6-OHDA has been shown to induce NF Kb

activation via IkB alpha degradation and subsequent translocation of NF-kB dimers into nucleus

thereby leading to the induction of pro-apoptotic effects (Tarabin & Schwaninger, 2004). Given

the importance of NF-kB pathway in cell death mechanisms we investigated the role of NF-kB

signaling pathway in Tweak induced neuronal cell death in N27 cells. For this purpose, initially

we measured Tweak induced IkB-alpha degradation in N27 dopaminergic neuronal cells by

immunoblot analysis. N27 cells were exposed to TWEAK for increasing time period (6h, 12h and

24h), and cell lysates were prepared at the end of the indicated time points and immunoblotted for

IKB-alpha. As anticipated a time dependent increase in IkB-alpha degradation was evidenced in

Tweak treated N27 cells indicative of NF-kB activation (Fig 9A).

160

To further investigate the mechanism of NF-kB signaling pathway, we next examined

phosphorylation of P65 in Tweak treated N27 cells by immunoblot analysis. N27 cells were

subjected to the same experimental conditions as detailed above and the magnitude of p65

phosphorylation was assessed via WB analysis. Our data indicated a time dependent increase in

P65 phosphorylation in Tweak treated N27 cells (Fig 9B). As shown above SN50, NF-kB inhibitor

attenuated TWEAK-induced apoptotic cell death further supporting the role of NF-kB activation

in the mechanism of TWEAK-induced dopaminergic neurodegeneration (Fig 9C).

Tweak Induced Downregulation of Phospho-AKT is Associated With GSK-3 Beta Activation

and Tau Phosphorylation.

Tweak exerts diverts effects via the activation of P13K/ AKT pathway in a cell type

specific manner. For example, Tweak can cause skeletal muscle wasting through inhibition of

P13K/ AKT pathway (C. Dogra et al., 2007). On the contrary, it promotes tumor cell survival

through activation of AKT pathway (L. Wu et al., 2013). Given the importance of P13K/ AKT

pathway in regulation of cell survival, we determined the activation status of Akt. So, we first

assessed phospho-AKT levels in Tweak treated N27 cells using immunoblot analysis. N27

dopaminergic neuronal cells were exposed to Tweak for varying duration (6-24h) and at the end

of the incubation period cell lysates were immunoblotted for Phospho-AKT. We found that Tweak

induced the downregulation of Phospho-AKT in dopaminergic cells (Fig 10A). These results

suggest that Tweak induced downregulation of Phospho-AKT levels may increase the

vulnerability of dopaminergic neuronal cells to pro-apoptotic insults.

Dysregulation of PI3K/Akt signaling event has been linked to PD neuropathology Studies

conducted in post mortem PD brains revealed a reduction in the ratio of p-Akt/t-Akt (Timmons et

al., 2009). Several drugs that are prescribed for PD treatment exert their therapeutic effects via

Akt activation (Aleyasin et al., 2010; Fernandez-Gomez et al., 2006; L. A. Greene et al., 2011; J.

161

H. Lim et al., 2008; Nakaso et al., 2008; Salinas et al., 2001; Tasaki et al., 2010). For example

rasagiline has been shown to protect nigral dopaminergic neurons via activation of PI3K/Akt

signaling mechanism (Weinreb et al., 2006). Therefore, in the present study we sought to

determine Akt activation in TWEAK treated cells. Tweak treatment evoked a marked down

regulation of p-Akt levels at all-time points tested. Given that PI3K/Akt is a key upstream

regulator of GSK-3b, the down regulation of Akt might be associated with GSK-3b activation and

resultant cell death (H. Zhang et al., 2011). Together our studies suggest that impairment in the

PI3K/Akt pathway may underlie TWEAK-induced dopaminergic neurotoxicity owing to

compromised cell survival machinery.

Aberrant GSK-3 activity has been linked to numerous neurodegenerative disorders

including AD and PD (Credle, George, et al., 2015). In fact GSK 3 activity has been shown to

induce neuronal apoptosis and conversely GSK inhibitors have been shown to elicit

neuroprotective and antiapoptotic effects in several cellular and mouse models (Beurel & Jope,

2006; Pap & Cooper, 1998). Furthermore, PI3/Akt signaling pathway has been linked to the

inactivation of GSK-3b activity via phosphorylation at the Ser9 site (M.-H. Liang & Chuang,

2006). Having demonstrated that TWEAK-induced Akt down regulation, we hypothesized that

the inhibitory state of GSK 3b will be down regulated (GSK-3b ser9) while the activated state of

GSK-3b will be upregulated thereby facilitating the mechanism of dopaminergic cell death in

TWEAK treated cells. First, we assessed the response of GSK-3b signaling events in TWEAK

treated N27 dopaminergic cells at various time points. An increase in phosphorylation of GSK 3b

(pGSK-3 Beta Tyrosine 216) was evidenced as early as 6h and remained elevated for the reminder

of the treatment period (Fig 10C). Conversely down regulation of pGSK-3b Serine 9 was observed

in TWEAK treated cells, consistent with neurodegeneration (Fig 10D). In the Tet-inducible GSK-

162

3b models postnatal expression of tet-inducible expression of GSK-3b-S9A (kinase active)

transgene led to neurodegeneration in the cortex striatum and hippocampus (Credle, George, et al.,

2015) further confirming the neuroprotective role of this kinase inactive state of GSK-3b. GSK-

3b has been shown to cause abnormal Tau phosphorylation. In fact his kinase has been linked to

a-synuclein-induced tau phosphorylation. Consistent with this, inhibition of GSK-3b was found

to inhibit accumulation of a-synuclein and p-tau formation in an MPTP mouse model of PD (Duka

et al., 2006). Given that Tau is a substrate of GSK-3b we assessed the levels of pTau in N27 cells

treated with TWEAK. An increase in p-tau was evidenced as early as 6h post drug treatment and

maximal induction was observed at 12h post TWEAK treatment (Fig 10E). Consistent with

previous reports a positive correlation between GSK-3 b activation and hyperphosphorylation of

p-Tau further suggest that functional interaction between GSk-3b and Tau coupled with

proteasomal dysfunction may lead to excessive accumulation of Tau ultimately leading to

dopaminergic neurodegeneration.

Discussion

Tumor necrosis factor like weak stimulator of apoptosis (Tweak) is a recently discovered

cytokine belonging to TNF superfamily. TWEAK has been implicated in numerous neurological

conditions, including Stroke, Neuro-inflammatory diseases, Glioblastoma Multiforme and

Multiple sclerosis(Bodmer et al., 2002; Burkly et al., 2007). We identified that Tumor Necrosis

Factor like Weak inhibitor of apoptosis (TWEAK) through targeted qPCR array which is

significantly up regulated in immortalized dopaminergic neuronal cells (N27 cells) following

inflammatory insult. In our study, initially we demonstrated differential Tweak expression in

dopaminergic neuronal cells and distinct brain regions. Next, we showed that, Tweak induces dose

dependent neurotoxicity in rat dopaminergic neuronal cells. Furthermore, we demonstrated that,

163

there is marked upregulation of TWEAK levels in the mouse substantia in MPTP treated mice.

Additionally, investigation into downstream signaling mechanisms revealed that, TWEAK-

induced dissipation of MMP and consequent reduction in GSH levels might linked to TWEAK-

induced dopaminergic neurotoxicity. Furthermore, upregulation of redox sensitive kinases namely

SYK and PKC delta was accompanied by a concomitant down regulation of UPS and autophagic

mechanisms further highlighting the importance of cellular redox machinery in the modulation of

cellular protein quality control machinery in TWEAK treated cells. In this context, our studies

raise the possibility that dysregulated Akt/GSK/Tau signaling events might atleast in part

contribute to dopaminergic cell death mechanism owing to accumulation of aggregate prone

proteins. Intriguingly SN-50, NF-kB inhibitor and Quercetin (Flavonoid) attenuated cell death

and improved mitochondrial function thereby resulting in increased survival of dopaminergic

neuronal cells. Together, these findings suggest that TWEAK induced dopaminergic

neuropathology may be linked to PD pathogenesis. A schematic of Tweak-induced dopaminergic

cell death is depicted in Fig 3.6.

Neurons, astrocytes and microglia exhibit differential TWEAK expression pattern

(Desplat-Jego et al., 2002). For example, Tweak is expressed in primary murine neurons and

upregulated following OGD (oxygen glucose deprivation) (J. A. Winkles, 2008). Consistent with

this report our study is the first to demonstrate higher basal levels of membrane bound full-length

TWEAK in the hippocampus and olfactory bulb while higher basal levels of cleaved soluble

TWEAK was evidenced in the striatum and substantia nigra of C57 black mice. This differential

expression of Tweak in various brain regions and different cell types might imply that Tweak /

Fn14 pathway might elicit differential effects under pathological and physiological conditions.

Fn14 receptor is upregulated in PC12 cells by nerve growth factor treatment and promotes neuritic

164

overgrowth and lamellipodia formation (Tanabe et al., 2003). Upregulation of Fn14 is associated

many inflammatory, neurological diseases and malignancies (J. A. Winkles, 2008). We found that,

Tweak upregulates Fn14 expression at lower concentration (30ng) with subsequent

downregulation at higher doses. It is plausible that TWEAK might induce receptor desensitization

at higher concentrations. Thus, further studies are warranted to study the effects of TWEAK on

Fn14 levels. Serum Tweak levels positively correlate with disease activity in Psoriatic arthritis /

Rheumatic arthritis and confirms its pathogenic role in the production of pro-inflammatory

cytokines (M. C. Park et al., 2008; Xia et al., 2011). Interestingly, our studies revealed a 2-3-fold

increase in TWEAK levels in the serum of PD patients compared to age-matched control patients

suggesting that TWEAK may serve as a biomarker for disease progression.

Administration of MPTP to mice causes selective degeneration of nigrostriatal

dopaminergic neurons and leads to pathologic changes similar to Parkinson’s disease (W. S. Choi

et al., 1999). Changes in neuro-inflammatory markers in PD patients showed increased levels of

TNF-alpha and IL-6, which may be responsible for mediating neuronal death (Dexter & Jenner,

2013; Dobbs et al., 1999). Previously we demonstrated that TNF-alpha-induced dopaminergic

apoptotic cell death through PKC delta proteolytic cleavage mechanism (Richard Gordon et al.,

2012) further supporting the pro apoptotic effects of TNF-alpha on dopaminergic neurons

(Gahring et al., 1996). Moreover, in a recent report blocking Tweak / Fn14 pathway was found to

attenuate dopaminergic neurodegeneration in a subacute MPTP model (Mustafa et al., 2016a). In

contrast to this report, Western blot analysis revealed marked upregulation of Tweak levels in the

mouse substantia nigra occuring at 3-6 h following MPTP treatment. Likewise, upregulation of

Tweak expression was observed in the TH positive neurons in the substantia nigra as determined

by IHC. Together our studies support the notion that TWEAK may serve as an oxidative stress

165

sensitive pro-apoptotic factor that increases the vulnerability of dopaminergic neurons to PD

pathogenesis.

NF-kB has been shown to be upregulated in post mortem PD brains (A. Ghosh et

al., 2007). For example selective inhibition of the activation of NF-kB was found to protect

dopaminergic neurons in MPTP-intoxicated mice (A. Ghosh et al., 2007). In addition it has been

demonstrated that PD neurotoxin such as 6_OHDA- induced NF-kB activation elicited a

proapoptotic effect in PC12 cells (Tarabin et al., 2004). NF-kB are activated in response to

cytokines, pathogens, and other cell injuries (Camandola & Mattson, 2007). NF-kB exists as a

homodimer and heterodimer and these dimers are retained in the cytoplasm in their inactive state

via IkB repressor protein. Signal transduction via the p50:p65 heterodimer is involved in the

canonical pathway. Following stimulation by diverse ligands including TNFa, a signaling complex

is formed which leads to the activation of IkB kinase (IKK). IKK catalyze the phosphorylation

IkB and subsequent degradation via the UPS which leads to the nuclear translocation of p50:p65

and transcriptional activation of downstream NF-kB target genes (Kopitar-Jerala, 2015). Tweak

has been shown to activate the canonical and non-canonical NF-kB pathways (T. Dohi & L. C.

Burkly, 2012; Polavarapu et al., 2005; Saitoh et al., 2003). Both pathways have been shown to

require cIAP1 which is a critical component of downstream signaling events mediated by TWEAK

(Enwere et al., 2014). TWEAK-Fn14 has been demonstrated to activate canonical NF-kB

signaling in numerous cell types (L. Jin et al., 2004; Polavarapu et al., 2005; A. B. Sanz et al.,

2010; Tran et al., 2005). For example, TWEAK was found to induce apoptosis of cortical neurons

subjected to OGD atleast in part via NF-kB activation (Potrovita et al., 2004) and has been

postulated to play a role in the post ischemic inflammatory response. In another study, activation

of the canonical NFkB signaling pathway was linked to TWEAK-induced muscle protein

166

degradation (Al-Sawaf et al., 2014; Charu Dogra et al., 2007). Consistent with these reports

marked induction of IkB degradation and p65 phosphorylation was evidenced in TWEAK

stimulated cells and as anticipated inhibition of NFkB activation via SN50 resulted in abrogation

of TWEAK-induced cell death. Indeed in a recent report Nrf2 mediated repression of NfkB was

found to protect against TWEAK-induced cell death following ischemia reperfusion in Nrf2-KO

mice (Al-Sawaf et al., 2014). Taken together these data indicate a role for NFkB in TWEAK-

induced induced dopaminergic cell death.

In the intrinsic apoptotic pathway loss of mitochondrial membrane permeability

leads to cytochrome c release and formation of the apoptosome complex encompassing

cytochrome c, APAF, ATP, and procaspase 9 in the cytoplasm which finally leads to cell death in

a caspase-3 dependent manner (Ghavami et al., 2014). Alternatively, the extrinsic apoptotic

pathway involves the activation of death receptors located in the plasma membrane such as TNF-

R. Stimulation of this receptor leads to receptor trimerization to which FADD and procaspase-8

has been shown to bind. Subsequently, activation of caspase-8 leads to the activation of caspase-

3 thereby completing the process of apoptotic cell death. Among the caspases, caspase-2, -8, -9

and -10 constitute the initiator caspases whereas caspase-3, -6, and -7 represent the effector

(Gómez-Sintes et al., 2011). Initiator caspase participate in the cleavage of the effector caspase

and cleaved effector caspase, in turn, cleave other protein substrates that are involved in apoptotic

cell death such as PARP (Gómez-Sintes et al., 2011). In the present study we found a positive

association between that the activation of Caspase-2,-8,-9, and cell death which suggest that

caspase-3 mediated activation of PARP cleavage is probably involved in TWEAK-induced

dopaminergic cell death. In addition GSK-3 has been shown to activate the intrinsic apoptotic

167

pathway via its effects on Bax (Linseman et al., 2004), Bim (Hongisto et al., 2003), VDAC

(Pastorino et al., 2005) thereby contributing to mitochondrial dysfunction. In another study

thapsigargin, and ER stress inducer not only induced caspase-3 activation but also GSK-3 activity

supporting the positive impact of GSK-3 on caspase-3 mediated intrinsic cell death signaling

events (L. Song et al., 2002). Consistent with these reports, in the present study loss of

mitochondrial membrane potential preceded both GSK-3b and caspase-3 activation suggesting

functional interaction between mitochondrial dysfunction and GSK-3b activation via a caspase-3

dependent mechanism in TWEAK-induced apoptotic cell death.

The PI3K/Akt is one among the well characterized cell survival signaling

mechanisms. Ketamine induced downregulation of phospho-AKT and upregulation of GSK-3

beta phosphorylation leads to apoptosis of cultured rat cortical neurons (Shang et al., 2007). On

the other hand, erythropoietin induced activation of AKT levels and associated downregulation of

GSK-3 beta phosphorylation enhances cellular survival in rat cortical neurons (Shang et al., 2007).

Consistent with previous findings, downregulation of Phospho-AKT is positively associated with

GSK-3 beta activation and Tau Phosphorylation in Tweak treated N27 dopaminergic neuronal

cells suggesting that down regulation of Akt activation coupled with activation of GSK-3b may

underlie the increased vulnerability of dopaminergic neurons to TWEAK-induced apoptotic cell

death. In line with our finding a previous report showed that TWEAK inhibited phosphorylation

of Akt and its downstream target GSK-3b, suggesting that inhibition of PI3K/Akt signaling

mechanism might contribute to skeletal muscle atrophy in response to TWEAK (Charu Dogra et

al., 2007). These results suggest the existence of a possible negative feedback loop between Akt

and GSK3b and that the lack of Akt dependent inactivation of GSK-3b (GSK-3bY265) might have

168

contributed to the neurotoxic mechanisms and associated dopaminergic cell death in TWEAK

treated cells.

It is well established that impairment in GSK3b contribute to the induction of cell

death signaling events that ultimately promote neuropathology in numerous neurodegenerative

diseases including PD (Petit-Paitel, 2010). In this context, a previous study revealed that GSK-3

beta is upregulated in the substantia nigra and that GSK-3b is localized within the halo of Lewy

bodies in post mortem PD brains suggesting that GSK-3b plays a role in PD pathogenesis (Nagao

& Hayashi, 2009). Furthermore in a recent report impairment in GSK-3 beta function was found

to contribute to the accumulation of tau and a-synuclein which in turn was found to be associated

with Parkinsonian neuropathology in a brain region specific manner (Credle, George, et al., 2015).

Consistent with this report a positive association between GSK3b activation and Tau

hyperphosphorylation was evidenced in TWEAK treated cells. To what extent GSK3b mediated

Tau hyperphosphorylation contributes to TWEAK-induced neuronal cell death remains to be

investigated.

It is well accepted that disruption of cellular protein quality control

characterized by accumulation of damaged organelles and misfolded proteins in the dopaminergic

neurons is a critical component of PD pathogenesis (Daniel J. Klionsky, 2007). Accumulation of

aggregate prone proteins can result from impaired protein clearance mechanisms owing to

sequential dysfunction of the ubiquitin proteasome system (UPS) and the autophagy lysosomal

system (ALS). Oxidative stress has been identified as key factor facilitating impaired proteostasis

(Janda et al., 2012). Using disease models and PD brains it has been implicated that dysfunction

in the autophagic process and UPS may be linked to PD pathogenesis (Dehay et al., 2010;

Ebrahimi-Fakhari et al., 2012; Janda et al., 2012). Additionally, enhancement of autophagy has

169

been shown to be beneficial in experimental models of PD (Spencer et al., 2009). Previously, we

reported in N27 dopaminergic neuronal cells, that methamphetamine (MA)-induced oxidative

stress via PKC delta activation contributes to impairment in the UPS and autophagy (M. Lin et al.,

2012). More recently we demonstrated that impairment in the autophagic mechanisms via c-Abl,

a redox sensitive tyrosine kinase, may contribute to enhanced microglial activation response

characterized the activation of NLRP3 related markers in dopamine enriched brain regions

(Lawana et al., 2017). Consistent with these reports in the present study we demonstrated that

depletion of autophagic markers and down regulation of UPS coincided with proteolytic cleavage

of redox sensitive kinases such as PKC delta and SYK activation supporting the role of elevated

oxidative stress upon impairment of cellular proteostasis. In this context, both MMP collapse and

ROS generation preceded the disruption of cellular protein quality control and activation of redox

sensitive kinases further highlighting the potential involvement of oxidative stress in impaired

cellular proteostasis upon TWEAK stimulation of N27 cells. This is in accordance with a previous

report from our lab demonstrating that proteosomal inhibition via MG132 in N27 cells is

associated with oxidative stress response and ultimately cell death. (Sun et al., 2006). Further,

uncovering the role of autophagy and UPS in TWEAK-induced neurodegeneration may lead to

improved understanding of cellular mechanisms underlying dopaminergic neuropathology in PD.

Immunoreceptors have been shown to act as either an activator when present in

immunoreceptor tyrosine-based activation motifs (ITAMs) or as an inhibitor when present with

immunoreceptor tyrosine based inhibition motifs (ITIMs). Coordinated activation of Src and SYK

kinase families have been implicated in ITAM based signaling cascade (C. A. Lowell, 2011). Syk

tyrosine kinase is expressed in numerous cells including hematopoietic cells (Shigeru Yanagi et

170

al., 2001). In a previous report Syk expression has been demonstrated in the neuronal cytoplasm

and SYK was found to phosphorylate synuclein (Alessandro Negro et al., 2001). In another report

Syk was found to phosphorylate Tau on Y18, and the interaction between kinase and the substrate

was demonstrated (Lebouvier et al., 2008). Intriguingly, neurofibrillary tangle (NFT) has been

shown to contain a phosphorylated Y18 residue, suggesting that SYK may be linked to AD

pathophysiology (G. J. Ho et al., 2005; Williamson et al., 2002). Consistent with this report WB

analysis performed in cell lysates from TWEAK treated cells displayed increased SYK activation

that positively correlated with proteolytic cleavage of PKC delta at 24h. Moreover, Tyr 525/526-

phosphorylated SYK and pTyr18 was present in granulovacuolar inclusions and in neurons with

neuro fibrillary tau pathology (Köhler et al., 2017) further supporting the neurodegenerative role

of SYK. Conversely, in another report Paris et al., (D. Paris et al., 2014) demonstrated that SYK

inhibition reduces Aβ production and increases the clearance of Ab across the blood brain barrier

(BBB). Together our data highlight that SYK activation may be linked to disruption of protein

clearance machinery thereby leading to the accumulation of aggregate prone proteins and

ultimately dopaminergic neurodegeneration presumably via an oxidative stress dependent

mechanism.

In conclusion, we provide the first evidence that TWEAK-induced dopaminergic

neurodegeneration via mitochondria mediated and caspase-3 mediated mechanism as well as a

SYK dependent mechanism. TWEAK could serve as a potential biomarker for PD based on the

observation that TWEAK levels were elevated in the serum of PD patients. Additionally, SYK

could serve as a novel target for the development of novel therapeutics for PD.

171

Bibliography

Afeseh Ngwa, H., et al. (2009). Vanadium Induces Dopaminergic Neurotoxicity Via Protein

Kinase C-Delta Dependent Oxidative Signaling Mechanisms: Relevance to

Etiopathogenesis of Parkinson's Disease. Toxicology and applied pharmacology, 240(2),

273-285. doi:10.1016/j.taap.2009.07.025

Al-Sawaf, O., et al. (2014). Nrf2 Protects Against TWEAK-mediated Skeletal Muscle Wasting.

Sci Rep, 4, 3625. doi:10.1038/srep03625

Aleyasin, H., et al. (2010). DJ-1 protects the nigrostriatal axis from the neurotoxin MPTP by

modulation of the AKT pathway. Proc Natl Acad Sci U S A, 107(7), 3186-3191.

doi:10.1073/pnas.0914876107

Beurel, E., et al. (2006). The paradoxical pro- and anti-apoptotic actions of GSK3 in the intrinsic

and extrinsic apoptosis signaling pathways. Prog Neurobiol, 79(4), 173-189.

doi:10.1016/j.pneurobio.2006.07.006

Blesa, J., et al. (2015). Oxidative stress and Parkinson’s disease. Frontiers in Neuroanatomy, 9,

91. doi:10.3389/fnana.2015.00091

Bodmer, J. L., et al. (2002). The molecular architecture of the TNF superfamily. Trends Biochem

Sci, 27(1), 19-26.

Burkly, L. C., et al. (2007). TWEAKing tissue remodeling by a multifunctional cytokine: role of

TWEAK/Fn14 pathway in health and disease. Cytokine, 40(1), 1-16.

doi:10.1016/j.cyto.2007.09.007

Burkly, L. C., et al. (2011). TWEAK/Fn14 pathway: an immunological switch for shaping tissue

responses. Immunol Rev, 244(1), 99-114. doi:10.1111/j.1600-065X.2011.01054.x

Camandola, S., et al. (2007). NF-kappa B as a therapeutic target in neurodegenerative diseases.

Expert Opin Ther Targets, 11(2), 123-132. doi:10.1517/14728222.11.2.123

Chandramani Shivalingappa, P., et al. (2012). N-Acetyl Cysteine Protects against

Methamphetamine-Induced Dopaminergic Neurodegeneration via Modulation of Redox

Status and Autophagy in Dopaminergic Cells. Parkinson's Disease, 2012, 424285.

doi:10.1155/2012/424285

172

Chicheportiche, Y., et al. (1997). TWEAK, a new secreted ligand in the tumor necrosis factor

family that weakly induces apoptosis. J Biol Chem, 272(51), 32401-32410.

Choi, W. S., et al. (1999). Characterization of MPP(+)-induced cell death in a dopaminergic

neuronal cell line: role of macromolecule synthesis, cytosolic calcium, caspase, and Bcl-

2-related proteins. Exp Neurol, 159(1), 274-282. doi:10.1006/exnr.1999.7133

Credle, J. J., et al. (2015). GSK-3beta dysregulation contributes to parkinson's-like

pathophysiology with associated region-specific phosphorylation and accumulation of tau

and alpha-synuclein. Cell Death Differ, 22(5), 838-851. doi:10.1038/cdd.2014.179

Cristovao, A. C., et al. (2013). PKCdelta mediates paraquat-induced Nox1 expression in

dopaminergic neurons. Biochem Biophys Res Commun, 437(3), 380-385.

doi:10.1016/j.bbrc.2013.06.085

De Ketelaere, A., et al. (2004). Involvement of GSK-3beta in TWEAK-mediated NF-kappaB

activation. FEBS Lett, 566(1-3), 60-64. doi:10.1016/j.febslet.2004.04.041

Dehay, B., et al. (2010). Pathogenic lysosomal depletion in Parkinson's disease. J Neurosci,

30(37), 12535-12544. doi:10.1523/jneurosci.1920-10.2010

Dempsey, L. A. (2015). TREM2 function in microglia. Nat Immunol, 16, 447.

doi:10.1038/ni.3166

Desplat-Jego, S., et al. (2002). TWEAK is expressed by glial cells, induces astrocyte

proliferation and increases EAE severity. J Neuroimmunol, 133(1-2), 116-123.

Dexter, D. T., et al. (2013). Parkinson disease: from pathology to molecular disease mechanisms.

Free Radic Biol Med, 62, 132-144. doi:10.1016/j.freeradbiomed.2013.01.018

Dias, V., et al. (2013). The Role of Oxidative Stress in Parkinson’s Disease. Journal of

Parkinson's disease, 3(4), 461-491. doi:10.3233/JPD-130230

Dobbs, R. J., et al. (1999). Association of circulating TNF-α and IL-6 with ageing and

parkinsonism. Acta Neurologica Scandinavica, 100(1), 34-41. doi:10.1111/j.1600-

0404.1999.tb00721.x

173

Dogra, C., et al. (2007). TNF-related weak inducer of apoptosis (TWEAK) is a potent skeletal

muscle-wasting cytokine. Faseb j, 21(8), 1857-1869. doi:10.1096/fj.06-7537com

Dogra, C., et al. (2007). TNF-related weak inducer of apoptosis (TWEAK) is a potent skeletal

muscle-wasting cytokine. The FASEB Journal, 21(8), 1857-1869. doi:10.1096/fj.06-

7537com

Dohi, T., et al. (2012). The TWEAK/Fn14 pathway as an aggravating and perpetuating factor in

inflammatory diseases: focus on inflammatory bowel diseases. J Leukoc Biol, 92(2), 265-

279. doi:10.1189/jlb.0112042

Drexler, H. C., et al. (2000). Inhibition of proteasome function induces programmed cell death in

proliferating endothelial cells. Faseb j, 14(1), 65-77.

Duka, T., et al. (2006). Alpha-synuclein induces hyperphosphorylation of Tau in the MPTP

model of parkinsonism. Faseb j, 20(13), 2302-2312. doi:10.1096/fj.06-6092com

Ebrahimi-Fakhari, D., et al. (2012). Protein degradation pathways in Parkinson's disease: curse

or blessing. Acta Neuropathol, 124(2), 153-172. doi:10.1007/s00401-012-1004-6

Enwere, E. K., et al. (2014). Role of the TWEAK-Fn14-cIAP1-NF-κB Signaling Axis in the

Regulation of Myogenesis and Muscle Homeostasis. Frontiers in Immunology, 5, 34.

doi:10.3389/fimmu.2014.00034

Fernandez-Gomez, F. J., et al. (2006). Pyruvate protects cerebellar granular cells from 6-

hydroxydopamine-induced cytotoxicity by activating the Akt signaling pathway and

increasing glutathione peroxidase expression. Neurobiol Dis, 24(2), 296-307.

doi:10.1016/j.nbd.2006.07.005

Fuller, J. P., et al. (2014). New roles for Fc receptors in neurodegeneration-the impact on

Immunotherapy for Alzheimer's Disease. Frontiers in Neuroscience, 8, 235.

doi:10.3389/fnins.2014.00235

Gahring, L. C., et al. (1996). Neuronal expression of tumor necrosis factor alpha in the murine

brain. Neuroimmunomodulation, 3(5), 289-303.

Ghavami, S., et al. (2014). Autophagy and apoptosis dysfunction in neurodegenerative disorders.

Prog Neurobiol, 112, 24-49. doi:10.1016/j.pneurobio.2013.10.004

174

Ghosh, A., et al. (2007). Selective inhibition of NF-kappaB activation prevents dopaminergic

neuronal loss in a mouse model of Parkinson's disease. Proc Natl Acad Sci U S A,

104(47), 18754-18759. doi:10.1073/pnas.0704908104

Ghosh, S., et al. (2015). Stress Granules Modulate SYK to Cause Microglial Cell Dysfunction in

Alzheimer's Disease. EBioMedicine, 2(11), 1785-1798. doi:10.1016/j.ebiom.2015.09.053

Gómez-Sintes, R., et al. (2011). GSK-3 Mouse Models to Study Neuronal Apoptosis and

Neurodegeneration. Frontiers in Molecular Neuroscience, 4, 45.

doi:10.3389/fnmol.2011.00045

Gordon, R., et al. (2012). Proteolytic activation of proapoptotic kinase protein kinase Cδ by

tumor necrosis factor α death receptor signaling in dopaminergic neurons during

neuroinflammation. J Neuroinflammation, 9, 82-82. doi:10.1186/1742-2094-9-82

Gordon, R., et al. (2010). Resveratrol Protects Dopaminergic Neurons in Parkinson’s Disease

Models by Modulating the PKC-delta Apoptotic Signaling Pathway & Microglial

Activation. The FASEB Journal, 24(1 Supplement), 763.765-763.765.

Gordon, R., et al. (2016). Protein kinase Cdelta upregulation in microglia drives

neuroinflammatory responses and dopaminergic neurodegeneration in experimental

models of Parkinson's disease. Neurobiol Dis, 93, 96-114. doi:10.1016/j.nbd.2016.04.008

Greenamyre, J. T., et al. (2001). Complex I and Parkinson's Disease. IUBMB Life, 52(3-5), 135-

141. doi:10.1080/15216540152845939

Greene, L. A., et al. (2011). Akt as a victim, villain and potential hero in Parkinson’s disease

pathophysiology and treatment. Cellular and molecular neurobiology, 31(7), 969-978.

doi:10.1007/s10571-011-9671-8

He, Y., et al. (2015). Mincle/Syk Signaling Pathway Contributes to Neuroinflammation after

Subarachnoid Hemorrhage in Rats. Stroke, 46(8), 2277-2286.

doi:10.1161/STROKEAHA.115.010088

He, Y., et al. (2015). Macrophage-Inducible C-Type Lectin/Spleen Tyrosine Kinase Signaling

Pathway Contributes to Neuroinflammation After Subarachnoid Hemorrhage in Rats.

Stroke, 46(8), 2277-2286. doi:10.1161/strokeaha.115.010088

175

Ho, G. J., et al. (2005). Altered p59Fyn kinase expression accompanies disease progression in

Alzheimer's disease: implications for its functional role. Neurobiol Aging, 26(5), 625-

635. doi:10.1016/j.neurobiolaging.2004.06.016

Hoesel, B., et al. (2013). The complexity of NF-kappaB signaling in inflammation and cancer.

Mol Cancer, 12, 86. doi:10.1186/1476-4598-12-86

Hongisto, V., et al. (2003). Lithium blocks the c-Jun stress response and protects neurons via its

action on glycogen synthase kinase 3. Mol Cell Biol, 23(17), 6027-6036.

Hwang, O. (2013). Role of Oxidative Stress in Parkinson's Disease. Experimental Neurobiology,

22(1), 11-17. doi:10.5607/en.2013.22.1.11

Inoue, J., et al. (2000). Tumor necrosis factor receptor-associated factor (TRAF) family: adapter

proteins that mediate cytokine signaling. Exp Cell Res, 254(1), 14-24.

doi:10.1006/excr.1999.4733

Inta, I., et al. (2008). Induction of the cytokine TWEAK and its receptor Fn14 in ischemic stroke.

J Neurol Sci, 275(1-2), 117-120. doi:10.1016/j.jns.2008.08.005

Janda, E., et al. (2012). Defective autophagy in Parkinson's disease: role of oxidative stress. Mol

Neurobiol, 46(3), 639-661. doi:10.1007/s12035-012-8318-1

Jin, H., et al. (2011). Transcriptional Regulation of Pro-apoptotic Protein Kinase Cδ:

IMPLICATIONS FOR OXIDATIVE STRESS-INDUCED NEURONAL CELL

DEATH. J Biol Chem, 286(22), 19840-19859. doi:10.1074/jbc.M110.203687

Jin, L., et al. (2004). Induction of RANTES by TWEAK/Fn14 interaction in human

keratinocytes. J Invest Dermatol, 122(5), 1175-1179. doi:10.1111/j.0022-

202X.2004.22419.x

Juarez Olguin, H., et al. (2016). The Role of Dopamine and Its Dysfunction as a Consequence of

Oxidative Stress. Oxid Med Cell Longev, 2016, 9730467. doi:10.1155/2016/9730467

Kang, R., et al. (2011). The Beclin 1 network regulates autophagy and apoptosis. Cell Death

Differ, 18(4), 571-580. doi:10.1038/cdd.2010.191

176

Kanthasamy, A. G., et al. (2006). A novel peptide inhibitor targeted to caspase-3 cleavage site of

a proapoptotic kinase protein kinase C delta (PKCdelta) protects against dopaminergic

neuronal degeneration in Parkinson's disease models. Free Radic Biol Med, 41(10), 1578-

1589. doi:10.1016/j.freeradbiomed.2006.08.016

Kanthasamy, A. G., et al. (2008). Environmental neurotoxin dieldrin induces apoptosis via

caspase-3-dependent proteolytic activation of protein kinase C delta (PKCdelta):

Implications for neurodegeneration in Parkinson's disease. Mol Brain, 1, 12.

doi:10.1186/1756-6606-1-12

Kaul, S., et al. (2005). Tyrosine phosphorylation regulates the proteolytic activation of protein

kinase Cdelta in dopaminergic neuronal cells. J Biol Chem, 280(31), 28721-28730.

doi:10.1074/jbc.M501092200

Kaul, S., et al. (2003). Caspase-3 dependent proteolytic activation of protein kinase C delta

mediates and regulates 1-methyl-4-phenylpyridinium (MPP+)-induced apoptotic cell

death in dopaminergic cells: relevance to oxidative stress in dopaminergic degeneration.

Eur J Neurosci, 18(6), 1387-1401.

Keeney, P. M., et al. (2006). Parkinson's disease brain mitochondrial complex I has oxidatively

damaged subunits and is functionally impaired and misassembled. J Neurosci, 26(19),

5256-5264. doi:10.1523/jneurosci.0984-06.2006

Kikkawa, U., et al. (2002). Protein Kinase C&delta; (PKC&delta;): Activaton Mechanisms and

Functions. The Journal of Biochemistry, 132(6), 831-839.

Kitazawa, M., et al. (2001). Dieldrin-induced oxidative stress and neurochemical changes

contribute to apoptopic cell death in dopaminergic cells. Free Radic Biol Med, 31(11),

1473-1485.

Klionsky, D. J. (2007). Autophagy: from phenomenology to molecular understanding in less than

a decade. Nature Reviews Molecular Cell Biology, 8, 931. doi:10.1038/nrm2245

https://www.nature.com/articles/nrm2245#supplementary-information

Köhler, C., et al. (2017). Distribution of spleen tyrosine kinase and tau phosphorylated at

tyrosine 18 in a mouse model of tauopathy and in the human hippocampus. Brain

Research, 1677(Supplement C), 1-13. doi:https://doi.org/10.1016/j.brainres.2017.08.029

177

Kopitar-Jerala, N. (2015). Innate Immune Response in Brain, NF-Kappa B Signaling and

Cystatins. Front Mol Neurosci, 8, 73. doi:10.3389/fnmol.2015.00073

Kumar, S. (2009). Caspase 2 in apoptosis, the DNA damage response and tumour suppression:

enigma no more? Nat Rev Cancer, 9(12), 897-903. doi:10.1038/nrc2745

Latchoumycandane, C., et al. (2005). Protein kinase Cdelta is a key downstream mediator of

manganese-induced apoptosis in dopaminergic neuronal cells. J Pharmacol Exp Ther,

313(1), 46-55. doi:10.1124/jpet.104.078469

Lawana, V., et al. (2017). Involvement of c-Abl Kinase in Microglial Activation of NLRP3

Inflammasome and Impairment in Autolysosomal System. J Neuroimmune Pharmacol,

12(4), 624-660. doi:10.1007/s11481-017-9746-5

Lebouvier, T., et al. (2008). The microtubule-associated protein tau is phosphorylated by Syk.

Biochim Biophys Acta, 1783(2), 188-192. doi:10.1016/j.bbamcr.2007.11.005

Lee, J., et al. (2012). Autophagy, mitochondria and oxidative stress: cross-talk and redox

signalling. Biochemical Journal, 441(Pt 2), 523-540. doi:10.1042/BJ20111451

Liang, M.-H., et al. (2006). Differential Roles of Glycogen Synthase Kinase-3 Isoforms in the

Regulation of Transcriptional Activation. Journal of Biological Chemistry, 281(41),

30479-30484. doi:10.1074/jbc.M607468200

Lim, J. H., et al. (2008). Bromocriptine activates NQO1 via Nrf2-PI3K/Akt signaling: novel

cytoprotective mechanism against oxidative damage. Pharmacol Res, 57(5), 325-331.

doi:10.1016/j.phrs.2008.03.004

Lim, K. L. (2007). Ubiquitin-proteasome system dysfunction in Parkinson's disease: current

evidence and controversies. Expert Rev Proteomics, 4(6), 769-781.

doi:10.1586/14789450.4.6.769

Lin, M., et al. (2012). Methamphetamine-induced neurotoxicity linked to ubiquitin-proteasome

system dysfunction and autophagy-related changes that can be modulated by protein

kinase C delta in dopaminergic neuronal cells. Neuroscience, 210, 308-332.

doi:10.1016/j.neuroscience.2012.03.004

178

Linseman, D. A., et al. (2004). Glycogen synthase kinase-3beta phosphorylates Bax and

promotes its mitochondrial localization during neuronal apoptosis. J Neurosci, 24(44),

9993-10002. doi:10.1523/jneurosci.2057-04.2004

Lowell, C. A. (2011). Src-family and Syk kinases in activating and inhibitory pathways in innate

immune cells: signaling cross talk. Cold Spring Harb Perspect Biol, 3(3).

doi:10.1101/cshperspect.a002352

Maharjan, S., et al. (2014). Mitochondrial impairment triggers cytosolic oxidative stress and cell

death following proteasome inhibition. Sci Rep, 4, 5896. doi:10.1038/srep05896

Mari, M., et al. (2009). Mitochondrial glutathione, a key survival antioxidant. Antioxid Redox

Signal, 11(11), 2685-2700. doi:10.1089/ars.2009.2695

Marsters, S. A., et al. (1998). Identification of a ligand for the death-domain-containing receptor

Apo3. Curr Biol, 8(9), 525-528.

Medeiros, M. S., et al. (2016). Iron and Oxidative Stress in Parkinson's Disease: An

Observational Study of Injury Biomarkers. PLoS One, 11(1), e0146129.

doi:10.1371/journal.pone.0146129

Meredith, G. E., et al. (2011). MPTP Mouse Models of Parkinson’s Disease: An Update. Journal

of Parkinson's disease, 1(1), 19-33. doi:10.3233/JPD-2011-11023

Miyazaki, I., et al. (2008). Dopaminergic neuron-specific oxidative stress caused by dopamine

itself. Acta Med Okayama, 62(3), 141-150. doi:10.18926/amo/30942

Mustafa, S., et al. (2016). The role of TWEAK/Fn14 signaling in the MPTP-model of

Parkinson's disease. Neuroscience, 319, 116-122.

doi:10.1016/j.neuroscience.2016.01.034

Nagao, M., et al. (2009). Glycogen synthase kinase-3beta is associated with Parkinson's disease.

Neurosci Lett, 449(2), 103-107. doi:10.1016/j.neulet.2008.10.104

Nakaso, K., et al. (2008). Caffeine activates the PI3K/Akt pathway and prevents apoptotic cell

death in a Parkinson's disease model of SH-SY5Y cells. Neurosci Lett, 432(2), 146-150.

doi:10.1016/j.neulet.2007.12.034

179

Negro, A., et al. (2001). Multiple phosphorylation of a-synuclein by protein tyrosine kinase Syk

prevents eosin-induced aggregation. The FASEB Journal. doi:10.1096/fj.01-0517fje

Negro, A., et al. (2002). Multiple phosphorylation of alpha-synuclein by protein tyrosine kinase

Syk prevents eosin-induced aggregation. Faseb j, 16(2), 210-212. doi:10.1096/fj.01-

0517fje

Nixon, R. A., et al. (2012). Autophagy and neuronal cell death in neurological disorders. Cold

Spring Harb Perspect Biol, 4(10). doi:10.1101/cshperspect.a008839

Panicker, N., et al. (2015). Fyn Kinase Regulates Microglial Neuroinflammatory Responses in

Cell Culture and Animal Models of Parkinson's Disease. 35(27), 10058-10077.

doi:10.1523/jneurosci.0302-15.2015

Pap, M., et al. (1998). Role of glycogen synthase kinase-3 in the phosphatidylinositol 3-

Kinase/Akt cell survival pathway. J Biol Chem, 273(32), 19929-19932.

Paris, D., et al. (2014). The spleen tyrosine kinase (Syk) regulates Alzheimer amyloid-beta

production and Tau hyperphosphorylation. J Biol Chem, 289(49), 33927-33944.

doi:10.1074/jbc.M114.608091

Park, M. C., et al. (2008). Relationship of serum TWEAK level to cytokine level, disease

activity, and response to anti-TNF treatment in patients with rheumatoid arthritis. Scand J

Rheumatol, 37(3), 173-178. doi:10.1080/03009740801898608

Pastorino, J. G., et al. (2005). Activation of glycogen synthase kinase 3beta disrupts the binding

of hexokinase II to mitochondria by phosphorylating voltage-dependent anion channel

and potentiates chemotherapy-induced cytotoxicity. Cancer Res, 65(22), 10545-10554.

doi:10.1158/0008-5472.can-05-1925

Petit-Paitel, A. (2010). [GSK-3beta: a central kinase for neurodegenerative diseases?]. Med Sci

(Paris), 26(5), 516-521. doi:10.1051/medsci/2010265516

Poetini, M. R., et al. Hesperidin attenuates iron-induced oxidative damage and dopamine

depletion in Drosophila melanogaster model of Parkinson's disease. Chemico-Biological

Interactions. doi:https://doi.org/10.1016/j.cbi.2017.11.018

180

Polavarapu, R., et al. (2005). Tumor necrosis factor-like weak inducer of apoptosis increases the

permeability of the neurovascular unit through nuclear factor-kappa B pathway

activation. J Neurosci, 25(44), 10094-10100. doi:10.1523/jneurosci.3382-05.2005

Potrovita, I., et al. (2004). Tumor necrosis factor-like weak inducer of apoptosis-induced

neurodegeneration. J Neurosci, 24(38), 8237-8244. doi:10.1523/jneurosci.1089-04.2004

Przedborski, S., et al. (2004). MPTP as a mitochondrial neurotoxic model of Parkinson's disease.

J Bioenerg Biomembr, 36(4), 375-379. doi:10.1023/B:JOBB.0000041771.66775.d5

Sabour Alaoui, S., et al. (2012). TWEAK affects keratinocyte G2/M growth arrest and induces

apoptosis through the translocation of the AIF protein to the nucleus. PLoS One, 7(3),

e33609. doi:10.1371/journal.pone.0033609

Saitoh, T., et al. (2003). TWEAK Induces NF-κB2 p100 Processing and Long Lasting NF-κB

Activation. Journal of Biological Chemistry, 278(38), 36005-36012.

doi:10.1074/jbc.M304266200

Salinas, M., et al. (2001). Akt1/PKBalpha protects PC12 cells against the parkinsonism-inducing

neurotoxin 1-methyl-4-phenylpyridinium and reduces the levels of oxygen-free radicals.

Mol Cell Neurosci, 17(1), 67-77. doi:10.1006/mcne.2000.0921

Sanz, A. B., et al. (2010). TWEAK activates the non-canonical NFkappaB pathway in murine

renal tubular cells: modulation of CCL21. PLOS ONE, 5(1), e8955.

doi:10.1371/journal.pone.0008955

Sato, S., et al. (2014). TWEAK/Fn14 Signaling Axis Mediates Skeletal Muscle Atrophy and

Metabolic Dysfunction. Frontiers in Immunology, 5, 18. doi:10.3389/fimmu.2014.00018

Satoh, J., et al. (2012). Phosphorylated Syk expression is enhanced in Nasu-Hakola disease

brains. Neuropathology, 32(2), 149-157. doi:10.1111/j.1440-1789.2011.01256.x

Sawada, M., et al. (2006). Role of cytokines in inflammatory process in Parkinson's disease. J

Neural Transm Suppl(70), 373-381.

Schweig, J. E., et al. (2017). Alzheimer’s disease pathological lesions activate the spleen tyrosine

kinase. Acta Neuropathologica Communications, 5, 69. doi:10.1186/s40478-017-0472-2

181

Shang, Y., et al. (2007). Protective effect of erythropoietin against ketamine-induced apoptosis in

cultured rat cortical neurons: involvement of PI3K/Akt and GSK-3 beta pathway.

Apoptosis, 12(12), 2187-2195. doi:10.1007/s10495-007-0141-1

Sherer, T. B., et al. (2007). Mechanism of toxicity of pesticides acting at complex I: relevance to

environmental etiologies of Parkinson's disease. J Neurochem, 100(6), 1469-1479.

doi:10.1111/j.1471-4159.2006.04333.x

Smeyne, M., et al. (2013). Glutathione Metabolism and Parkinson’s Disease. Free Radic Biol

Med, 62, 13-25. doi:10.1016/j.freeradbiomed.2013.05.001

Song, L., et al. (2002). Central role of glycogen synthase kinase-3beta in endoplasmic reticulum

stress-induced caspase-3 activation. J Biol Chem, 277(47), 44701-44708.

doi:10.1074/jbc.M206047200

Spencer, B., et al. (2009). Beclin 1 gene transfer activates autophagy and ameliorates the

neurodegenerative pathology in alpha-synuclein models of Parkinson's and Lewy body

diseases. J Neurosci, 29(43), 13578-13588. doi:10.1523/jneurosci.4390-09.2009

Sun, F., et al. (2005). Dieldrin induces ubiquitin-proteasome dysfunction in alpha-synuclein

overexpressing dopaminergic neuronal cells and enhances susceptibility to apoptotic cell

death. J Pharmacol Exp Ther, 315(1), 69-79. doi:10.1124/jpet.105.084632

Sun, F., et al. (2006). Proteasome inhibitor MG-132 induces dopaminergic degeneration in cell

culture and animal models. Neurotoxicology, 27(5), 807-815.

doi:10.1016/j.neuro.2006.06.006

Sun, F., et al. (2007). Environmental neurotoxic chemicals-induced ubiquitin proteasome system

dysfunction in the pathogenesis and progression of Parkinson's disease. Pharmacol Ther,

114(3), 327-344. doi:10.1016/j.pharmthera.2007.04.001

Sun, F., et al. (2009). Mitochondrial accumulation of polyubiquitinated proteins and differential

regulation of apoptosis by polyubiquitination sites Lys-48 and -63. J Cell Mol Med,

13(8b), 1632-1643. doi:10.1111/j.1582-4934.2009.00775.x

Tanabe, K., et al. (2003). Fibroblast growth factor-inducible-14 is induced in axotomized

neurons and promotes neurite outgrowth. J Neurosci, 23(29), 9675-9686.

182

Tanida, I., et al. (2008). LC3 and Autophagy. Methods Mol Biol, 445, 77-88. doi:10.1007/978-1-

59745-157-4_4

Tarabin, V., et al. (2004). The role of NF-kappaB in 6-hydroxydopamine- and TNFalpha-

induced apoptosis of PC12 cells. Naunyn Schmiedebergs Arch Pharmacol, 369(6), 563-

569. doi:10.1007/s00210-004-0938-1

Tasaki, Y., et al. (2010). Meloxicam protects cell damage from 1-methyl-4-phenyl pyridinium

toxicity via the phosphatidylinositol 3-kinase/Akt pathway in human dopaminergic

neuroblastoma SH-SY5Y cells. Brain Res, 1344, 25-33.

doi:10.1016/j.brainres.2010.04.085

Timmons, S., et al. (2009). Akt signal transduction dysfunction in Parkinson's disease. Neurosci

Lett, 467(1), 30-35. doi:10.1016/j.neulet.2009.09.055

Tiwari, M., et al. (2011). A knockout of the caspase 2 gene produces increased resistance of the

nigrostriatal dopaminergic pathway to MPTP-induced toxicity. Exp Neurol, 229(2), 421-

428. doi:10.1016/j.expneurol.2011.03.009

Tran, N. L., et al. (2005). The tumor necrosis factor-like weak inducer of apoptosis (TWEAK)-

fibroblast growth factor-inducible 14 (Fn14) signaling system regulates glioma cell

survival via NFkappaB pathway activation and BCL-XL/BCL-W expression. J Biol

Chem, 280(5), 3483-3492. doi:10.1074/jbc.M409906200

Trebing, J., et al. (2014). Analyzing the signaling capabilities of soluble and membrane

TWEAK. Methods Mol Biol, 1155, 31-45. doi:10.1007/978-1-4939-0669-7_4

Tsujimura, T., et al. (2001). Syk protein-tyrosine kinase is involved in neuron-like differentiation

of embryonal carcinoma P19 cells. FEBS Lett, 489(2-3), 129-133.

Tuffy, L. P., et al. (2010). Characterization of Puma-dependent and Puma-independent neuronal

cell death pathways following prolonged proteasomal inhibition. Mol Cell Biol, 30(23),

5484-5501. doi:10.1128/mcb.00575-10

Wang, D., et al. (2010). TWEAK/Fn14 promotes apoptosis of human endometrial cancer cells

via caspase pathway. Cancer Lett, 294(1), 91-100. doi:10.1016/j.canlet.2010.01.027

183

Wang, L., et al. (1994). Ich-1, an Ice/ced-3-related gene, encodes both positive and negative

regulators of programmed cell death. Cell, 78(5), 739-750.

doi:https://doi.org/10.1016/S0092-8674(94)90422-7

Weinreb, O., et al. (2006). Involvement of multiple survival signal transduction pathways in the

neuroprotective, neurorescue and APP processing activity of rasagiline and its propargyl

moiety. J Neural Transm Suppl(70), 457-465.

Williamson, R., et al. (2002). Rapid tyrosine phosphorylation of neuronal proteins including tau

and focal adhesion kinase in response to amyloid-beta peptide exposure: involvement of

Src family protein kinases. J Neurosci, 22(1), 10-20.

Wilson, C. A., et al. (2002). Death of HT29 adenocarcinoma cells induced by TNF family

receptor activation is caspase-independent and displays features of both apoptosis and

necrosis. Cell Death Differ, 9(12), 1321-1333. doi:10.1038/sj.cdd.4401107

Winkles, J. A. (2008). The TWEAK-Fn14 cytokine-receptor axis: discovery, biology and

therapeutic targeting. Nat Rev Drug Discov, 7(5), 411-425. doi:10.1038/nrd2488

Winkles, J. A. (2008). The TWEAK–Fn14 cytokine–receptor axis: discovery, biology and

therapeutic targeting. Nature reviews. Drug discovery, 7(5), 411-425.

doi:10.1038/nrd2488

Wolozin, B., et al. (2015). Syk and Yea Shall Find. EBioMedicine, 2(11), 1590-1591.

doi:10.1016/j.ebiom.2015.11.012

Wu, L., et al. (2013). Inhibition of endoplasmic reticulum stress is involved in the

neuroprotective effects of candesartan cilexitil in the rotenone rat model of Parkinson's

disease. Neuroscience Letters, 548, 50-55.

doi:https://doi.org/10.1016/j.neulet.2013.06.008

Xia, L., et al. (2011). Increased serum TWEAK levels in Psoriatic arthritis: relationship with

disease activity and matrix metalloproteinase-3 serum levels. Cytokine, 53(3), 289-291.

doi:10.1016/j.cyto.2010.12.003

Yanagi, S., et al. (2001). Syk Expression and Novel Function in a Wide Variety of Tissues.

Biochem Biophys Res Commun, 288(3), 495-498.

doi:https://doi.org/10.1006/bbrc.2001.5788

184

Yang, Y., et al. (2004). Suppression of caspase-3-dependent proteolytic activation of protein

kinase C delta by small interfering RNA prevents MPP+-induced dopaminergic

degeneration. Mol Cell Neurosci, 25(3), 406-421. doi:10.1016/j.mcn.2003.11.011

Yepes, M. (2013). TWEAK and Fn14 in the Neurovascular Unit. Front Immunol, 4, 367.

doi:10.3389/fimmu.2013.00367

Yepes, M., et al. (2005). A soluble Fn14-Fc decoy receptor reduces infarct volume in a murine

model of cerebral ischemia. Am J Pathol, 166(2), 511-520. doi:10.1016/s0002-

9440(10)62273-0

Zhang, D., et al. (2007). Protein kinase C delta negatively regulates tyrosine hydroxylase activity

and dopamine synthesis by enhancing protein phosphatase-2A activity in dopaminergic

neurons. J Neurosci, 27(20), 5349-5362. doi:10.1523/jneurosci.4107-06.2007

Zhang, H., et al. (2011). Tacrine(2)-ferulic acid, a novel multifunctional dimer, attenuates 6-

hydroxydopamine-induced apoptosis in PC12 cells by activating Akt pathway.

Neurochem Int, 59(7), 981-988. doi:10.1016/j.neuint.2011.09.001

Zhou, C., et al. (2008). Oxidative Stress in Parkinson's Disease: A Mechanism of Pathogenic and

Therapeutic Significance. Annals of the New York Academy of Sciences, 1147, 93-104.

doi:10.1196/annals.1427.023

185

Figures

Fig 1: Expression pattern of Tweak in the mouse brain. C57 black mice were selected for determining

Tweak expression in various brain regions. Untreated C57 black mice were sacrificed and various brain

regions such as substantia nigra, striatum, hippocampus, olfactory bulb, cortex and cerebellum were

dissected out. Brain tissue lysates were prepared and probed using a rabbit polyclonal antibody to Tweak

that detects membrane and active cleaved form. Optical densities of protein bands were analyzed with the

Odyssey imaging system (LI-COR) and -actin was used a loading control. Band intensities of membrane

and active cleaved Tweak were quantified with densitometric analysis and are normalized to -actin.

Representative immunoblot of at least 3 independent experiments was shown. The densitometric analysis

of normalized band intensity for each treatment group expressed as percentage of control group was plotted

on histogram. Statistical analysis was carried out using Anova followed by Student Newman Keul’s post

test. Asterisk (* p < 0.05,** p < 0.01) represents significant differences between Tweak expression between

various brain regions.

186

Fig 2. Tweak and Fn14 are expressed in dopaminergic neurons in culture. A. Differential Tweak

expression in N27 cells and Mn9D cells. N27 cells (Rat dopaminergic neuronal cell line) and Mn9D cells

(fusion of embryonic ventral mesencephalic and neuroblastoma cells) were used to determine Tweak

expression in dopaminergic neuronal cells. N27 cells and Mn9D cells were grown in culture separately. Later,

untreated N27 cells and Mn9D cells were collected and lysates were prepared. Next, whole cell lysates were

probed with rabbit polyclonal antibody to Tweak that detects membrane and cleaved soluble Tweak. Odyssey

imaging system (LI-COR) was used to analyze the protein bands. Band intensities were normalized with -

actin for equal loading and quantified with densitometric analysis. Representative immunoblot of three

independent experiment was shown. Histogram represents normalized band intensity for each treatment group

expressed as percentage of control group. Statistical analysis was carried out using Anova followed by Student

Newman Keul’s post test. Asterisks (* p < 0.05 and** p < 0.01) represent significant differences between

membrane and soluble Tweak expression in N27 cells and Mn9D cells. B. Fn14 (Tweak receptor) upregulation

in Tweak stimulated N27 cells. Rat dopaminergic N27 cells treated with 30 ng/ml, 50 ng/ml and 100 ng/ml

Tweak for 24h. Post incubation whole cell lysates were prepared and resolved with 15% SDS-PAGE. Later

they were subjected to immunoblot analysis using antibodies specific to Fn14 receptor. Membranes were re-

probed with -actin to ensure equal protein loading. Band intensity was analyzed with the Odyssey imaging

system (LI-COR). Representative immunoblot of at least 3 independent experiments was shown. Histogram

represents normalized band intensity for each treatment group expressed as percentage of control group.

Statistical analysis was carried out using Anova followed by Student Newman Keul’s post test. Asterisk (** p

< 0.01) represents significant differences in Fn14 expression between control and Tweak treated groups.

187

Fig 3 Upregulation of Tweak levels in the serum and CSF samples of Human PD patients. A-

B. Serum and CSF samples of PD patients and age matched controls were obtained from Human

Brain and Spinal Resource Center (VA Greater Los Angeles Health care system). Representative

western blot for Tweak expression in serum (Fig A) and CSF (Fig B) from human PD patients and

age matched controls probed using rabbit polyclonal antibody directed against Tweak. Increased

Tweak levels were evident in PD patient samples as compared to age matched control patients.

Densitometric band intensities for Tweak determined from control and PD samples expressed as

fold of control. Statistical analysis was carried out using student T test.

188

189

Fig 4 Impact of Parkinsonian toxin on Tweak expression in dopaminergic neuronal cells A.

Time dependent increase in Tweak expression in MPP+ treated N27 dopaminergic neuronal cells.

N27 cells were treated MPP 300 uM for 6h,12h and 24h. Post treatment, whole cell lysates were

prepared. 40 ug of protein was loaded and resolved with 15% SDS-PAGE. Later they were

subjected to immunoblot analysis with rabbit polyclonal Tweak antibody that detects membrane

and active cleaved Tweak. Membranes were also probed with -actin to ensure equal protein

loading. A time dependent increase in Tweak expression was evidenced in MPP+ treated

dopaminergic neuronal cells which peaked at 12h and 24h. Experiments were repeated three times

and a representative western-blot was shown. The bar graph represents band intensities for the

cleaved Tweak from three blots quantified using densitometric analysis and normalized to -actin.

Asterisk (* p < 0.05) represents significant differences in between MPP+ treated groups and

controls using one-way ANOVA with bonferroni post test comparision. B. Tweak upregulation in

nigral dopaminergic neurons in Acute MPTP animal model of Parkinson Disease. In this model,

C57 black mice were injected with four MPTP doses (18 mg/kg) intraperitoneally and were

euthanized at 3h, 6h, 12h, 24h and 72h post MPTP injection. Substantia nigra was dissected out

and tissue lysates were prepared from mice treated with either MPTP or saline. Later, they were

subjected to immunoblot analysis with Tweak antibody that detects membrane and soluble Tweak.

-actin was used a loading control for equal loading of samples. Cleaved Tweak band intensity

was quantified by densitometric analysis after normalization to β-actin. Experiment was repeated

for three times with 4 mice per group. Representative immuno-blot indicates maximum Tweak

expression occurs at 3h and 6h in Substantia Nigra (Fig B) of MPTP treated mice as compared to

control mice. Asterisk (*p<0.05) significant differences in between MPTP treated groups and

190

control group using one-way ANOVA with bonferroni post test comparison. C. Double

immunolabelling IHC for Tweak and TH in mouse nigral sections in subacute MPTP animal

model of Parkinson Disease. In this model, C57black mice were injected with 25 mg/kg of MPTP

once daily for 5 consecutive days and sacrificed at 3 days after the last MPTP injection. Later

successive 30 μm ventral midbrain sections will be cut with a cryostat and processed for immuno-

histochemical analysis of Tweak and TH expression with double immuno labeling studies.

Experiments were repeated three times and representative images were shown. Marked Tweak

expression in the TH+ neurons along the nigral tract of the MPTP treated mice (Fig C) .

Fig 5. Tweak induces concentration dependent cell death in N27 dopaminergic neuronal

cells. A. Assessment of dopaminergic neuronal cell viability with Tweak and TNF with MTT

analysis. N27 cells were treated with Tweak 30 ng, Tweak 50 ng, Tweak 75 ng, Tweak 100 ng and

TNF 30 ng for 24h. Post incubation, cell viability was assessed by measuring the formation of

formazan crystals from tetrazolium inside the active mitochondria. The mitochondrial activity

decreased significantly after exposure Tweak and TNF for 24 h . Cell survival was presented as

percent of control. Bar graph values are representative of mean ± S.E.M. for each experiment

performed with n = 8. Asterisk (p***<0.001) represents significant differences between Tweak

treated groups and control groups as determined by ANOVA followed by Student Newman Keul’s

test. B. Visualization of MPP+ and Tweak toxicity in dopaminergic neurons by Sytox assay. N27

cells were treated with MPP 300 uM, Tweak 300 ng and Tweak 150 ng for 24h. Post treatment,

cells were incubated with with 1uM Sytox green dye and cell death was determined by Sytox green

imaging. Visualization of Sytox positive cells was done under fluorescence microscopy with

excitation of 485 nm and emission of 538 nm. Increased green florescence is evident with Tweak

and MPP+ treatment indicating significant neurotoxicity in dopaminergic neuronal cells.

191

Fig 6. Effect of Tweak on Mitochondrial Membrane potential (MMP) and GSH levels in N27

cells A. Time course studies of the impact of Tweak with and without Quercitin on MMP using

JC-1 fluorometry. N27 dopaminergic neuronal cells were treated with Tweak 100 ng for 3h,6h,12h

and 24h with and without Quercitin 3 uM. Post treatment, lysates were collected and incubated

with JC-1 dye for 30 min. Post incubation, changes in mitochondrial membrane potential were

measured spectrophotometrically using redox sensitive JC-1 dye with the help of Gemini

Fluorescence plate reader. Reduction in red: green absorbance ratio of JC-1 dye indicate the loss

of mitochondrial membrane potential (Δψm

). Each experiment is repeated three times with n=8.

Significant time dependent dissipation of mitochondrial membrane potential was evidenced in

dopaminergic neuronal cells upon exposure to Tweak which was partially reversed with quercitin.

Asterisk (**p<0.01, p***<0.001) represents significant differences between Tweak treated groups

and control group as determined by ANOVA followed by Student Newman Keul’s test. B.

Quercitin (Flavonoid antioxidant) offers partial protection against Tweak induced apoptotic cell

death in N27 cells. N27 cells were treated with Tweak 100ng with or without Quercitin for 3h, 6h,

12h and 24h. Post incubation, cell viability was assessed using MTT analysis which is based on

formation of formazan crystals. Tweak induced cell death in dopaminergic neuronal cells is

partially reversed by Quercitin. Each experiment is repeated for three times with n = 8. Asterisks

6A

6B

6C

192

(**p<0.01, p***<0.001) represent significant differences between Tweak / Quercitin treated

groups and control group as determined by ANOVA followed by Student Newman Keul’s test. C.

GSH depletion with Tweak and TNF treatment in N27 cells. Rat dopaminergic N27 cells treated

with 100 ng/ml Tweak and TNF 30 ng for 24h. Post-treatment, Tweak induced reduction in GSH

levels was measured by with Monochlorobimane fluometric method. Formation of MCB-GSH

complex was measured with spectrophotometer at Emission 460 nm and Excitation 360 nm to

measure the GSH levels in Tweak and TNF treated N27 cells. Each experiment is repeated three

times with n= 4. Values are expressed as percentage of GSH compared with untreated control.

Significant depletion of GSH levels (25%) were observed in dopaminergic neuronal cells upon

exposure with Tweak and TNF for 24h. Asterisk (*p<0.05) represents significant differences

between Tweak / TNF treated groups and control group as determined by ANOVA followed by

Student Newman Keul’s test.

Fig 7. Tweak induces extrinsic and intrinsic caspase activation in N27 cells. A-D. N27 cells were

treated with Tweak 100ng treatment for 3h, 6h, 12h and 24h. Post treatment, aliquots of cell extracts

were collected and the activities of caspase-8 (A), caspase-2 (B), caspase-9 (C) and caspase-3 (D)

were determined using fluorogenic substrates Ac-LEHD-AFC Ac-VDVAD-AFC Ac-LEHD-AMC

and Ac-DEVD-AMC, respectively with excitation of 380 nm and emission of 460 nm. Significant

increase in caspase-8 (n=6; p***<0.001), caspase-2 (n=6; p***<0.001), caspase-9 (n=6;

p***<0.001) and caspase-3 (n=6; p***<0.001) were observed after exposure to Tweak for 24h in

dopaminergic neuronal cells. Asterisk (p***<0.001) represents significant differences between

Tweak treated groups and control group as determined by ANOVA followed by Student Newman

Keul’s test.

193

Fig 8. Impaired Auto-lysosomal system (ALS) and Proteosomal system (UPS) in Tweak treated N27

cells. A-B. Tweak causes Autophagy dysfunction in N27 cells. Immunoblot analysis of LC3 A/B and Beclin

in whole cell lysates of N27 dopaminergic neuronal cells treated with 100 ng/ml Tweak for 3h, 6h, 12h and

24h. Equal loading of protein in each lane is confirmed by probing the membrane with -actin. Densitometric

analysis of LC3 A/B and Beclin levels is represented next to western blot. Each experiment is repeated three

times and a representative immuno-blot representing LC3 A/B and beclin expression in Tweak treated N27

cells was shown. LC3 II and Beclin bands were quantified and expressed as percentage of control. Significant

downregulation of autophagy markers LC3 and beclin were evidenced in Tweak treated N27 cells. Asterisks

(* p < 0.05, *** p < 0.001) represents significant differences between Tweak treated control groups and

control group as indicated by Anova followed by Student Newman Keul’s post test. C. Proteosomal system

(UPS) in Tweak stimulated N27 rat dopaminergic neuronal cells. Rat dopaminergic N27 cells treated with

100 ng/ml Tweak for 3h,6h,12h to 24h. Post treatment, proteosomal activity was assayed by with the help

of substrate Suc-Leu-Val-Tyr-AMC (chymotrypsin like activity) cleavage in the Tweak stimulated cell

culture lysates. Enzymatic activity was normalized to protein concentration and expressed as percentage of

vehicle treated cells. Each experiment is performed three times with n=8. Significant time dependent

downregulation of proteosomal activity was noticed in dopaminergic neuronal cells upon Tweak treatment

as compared to controls cells. Asterisk (*p<0.05) represents significant differences between Tweak treated

groups and control group as determined by ANOVA followed by Student Newman Keul’s test.

194

Fig 9. Tweak induced NF-kB activation precedes cell death in N27 rat dopaminergic neuronal

cells. A-B. NF-kB pathway activation in Tweak treated N27 cells. N27 dopaminergic neuronal

cells treated with Tweak for 6h, 12h and 24h. Post incubation, lysates were probed with NF-kB

phospho-P65 and IkB-alpha by western blotting. Significant time dependent IkB-alpha

degradation and NF-kB phospho-P65 activation was evidenced in dopaminergic neuronal cells

upon Tweak stimulation. Band intensities of NF-kB phsospho-P65 and IkB-alpha from three blots

were quantified using densitometric analysis after normalizing with -actin. Each experiment is

repeated three times and a representative western blot is shown. Asterisk (** p < 0.01) represents

significant differences between Tweak treated groups and control group as analyzed by ANOVA

followed by Student Newman Keul’s post test. C. SN-50 (NF-kB inhibitor) offers partial

neuroprotection against Tweak induced neurotoxicity in N27 cells. N27 cells were treated with

Tweak 100ng and 30 ng TNF with or without NF-kB inhibitor SN-50 for 24h. Post incubation, cell

viability was assessed using MTT analysis. Each experiment performed three times with n = 8.

Tweak induced dopaminergic neuronal cell death is partially reversed by SN-50 (NF-kB inhibitor).

Asterisk (p***<0.001) represent significant differences between Tweak/TNF treated groups and

control group as determined by ANOVA followed by Student Newman Keul’s test.

195

Fig 10. Phospho-AKT, GSK-3 beta and Phospho-tau immunoblot analysis following Tweak

treatment. A-E. Rat dopaminergic N27 cells were treated with 100 ng/ml Tweak for 3h, 6h, 12h

and 24h. Post treatment, aliquots of cell extracts were collected and lysates were prepared. Equal

amounts of protein from treated cell lysates were separated on SDS-PAGE and transferred onto

nitrocellulose membrane for immunoblot analysis using antibodies for Phospho-AKT, GSK-3

parent, GSK-3 beta Serine9 and Phospho-tau. Equal loading in all the lanes is ascertained by re-

probing the membrane with -actin. All the experiments were repeated three times and a

representative western blot is shown. Bar graphs represent densitometric scanning analysis of

protein bands after normalization with -actin corresponding to the western blot. Downregulation

of Phospho-AKT is associated with GSK-3 beta activation and phospho-tau accumulation in

Tweak stimulated N27 rat dopaminergic neuronal cells. Asterisks (* p < 0.05,** p < 0.05)

represents significant differences between Tweak treated groups and control group as determined

by Anova followed by Student Newman Keul’s post test.

196

CHAPTER 3. SYK CONTRIBUTES TO TWEAK-INDUCED INFLAMMATORY

RESPONSE THROUGH ACTIVATION OF NLRP3 INFLAMMASOME ACTIVATION,

ER STRESS RESPONSE and ALS DYSFUNCTION IN MICROGLIA

To be Submitted to Journal of Neuroscience

Sri Kanuri1,2, Neeraj Singh1, Vivek Lawana1, Huajun Jin1, Vellareddy Anantharam1, Anumantha

Kanthasamy1 and Arthi Kanthasamy1 *

Abstract

Accumulating evidence suggest a positive association between microglial activation

response and DAgic neurodegenerationin in the pathogenesis of Parkinson’s disease (PD). But

the precise innate immune mechanism that the microglia recruit to trigger or promote inflammatory

response during PD progression remain poorly understood. The present study explored the roles

of syk, a pro-inflamamtory kinase in the regulation of Tweak-induced microglial activation

response using BV2 microglial cell line and primary microglia in culture. In BV2 microglial cells,

TWEAK challenge for increasing duration resulted in a coincident increase in SYK activation and

stress granule formation as assessed via WB and enzymatic analysis. We further found that

TWEAK enhanced NLRP3 inflammasome activation, as reflected by increased generation of IL-

1b, IL-18 in Bv2 cells. Intriguingly, TWEAK-induced NLRP3 inflammasome activation coincided

with an upregulation of NOX, ER stress response and JNK as well as impairment of auto-

phagosomal lysosomal system (ALS) function in BV2 microglial cells. Pretreatment with RX788,

a selective SYK inhibitor or siRNA mediated gene silencing of SYK in BV2 microglial cells

attenuated TWEAK-induced NLRP3 inflammasome activation, ER stress response, NOX and JNK

1 Department of Biomedical Sciences, Iowa State University, Ames, IA 50010

2 Primary researcher and author

*To whom correspondence should be addressed. E-mail: [email protected]

197

activation, as well as ALS dysfunction. Intriguingly, our in vivo studies using a TWEAK neuro-

inflammation mouse model (TWEAK infusion into the striatum) failed to show an alteration in the

expression levels of SYK or NLRP3 related markers in the striatum. Likewise, piceatannol, a Syk

inhibitor failed to elicit an anti-inflammatory response in the TWEAK neuro-inflammation model.

Taken together our studies suggest that at least in TWEAK stimulated BV2 microglial cells, SYK

mediated oxidative stress response, ER stress response and ALS dysfunction may contribute to

NLRP3 inflammasome activation. Further studies are needed to optimize the TWEAK neuro-

inflammation mouse model.

Introduction

Parkinson’s disease (PD) is most common neurodegenerative disorder characterized

by microglial activation and neuroinflammation and dopaminergic neurodegeneration in

substantia nigra (Machado et al., 2016; L. Qian & Flood, 2008). PD is characterized by motor

symptoms like tremor, rigidity, bradykinesia, gait disturbances, impaired handwriting, speech

deficits and non-motor symptoms like apathy, depression, cognitive dysfunction, hallucinosis,

orthostatic hypotension, urogenital dysfunction, constipation and rapid sleep movement disorder

(Moustafa et al., 2016; Poewe, 2008). Neuro-inflammation is regarded as an important

contributing factor for dopaminergic cell death in PD (Machado et al., 2016; Q. Wang et al., 2015).

Microglial mediated neuro-inflammatory is being increasingly recognized as a critical link in PD

pathology (Machado et al., 2016; L. Qian et al., 2008). Activated microglia has been demonstrated

in substantia nigra of post mortem brain tissues and positron emission tomography scan (PET) has

provided substantia evidence of microglial activation in mid brain tissues (Bartels et al., 2010;

McGeer et al., 1988). Sustained and chronic microglial activation in mid brain results in production

of pro-inflammatory cytokines like IL-1 beta, TNF-alpha, IL-6 and IFN-gamma which

198

subsequently leads to dopaminergic neurodegeneration (C. C. Ferrari et al., 2006; K. Kaur et al.,

2017; Nagatsu & Sawada, 2007; Q. Wang et al., 2015)

Innate immune response functions as a defense mechanism against invading pathogens and

damage associated molecules. Inflammasomes are multiprotein platforms that has been linked to

the pathogenesis of numerous diseases including AD (H. Guo et al., 2015; Lamkanfi & Dixit,

2012; M. K. Saito, 2011). The most commonly studied inflammasome nucleotide-binding

oligomerization domain, leucine rich repeat and pyrin domain containing inflammasome (NLRP3)

is an intracellular multi-meric protein complex that becomes activated by wide variety of

pathogens, environmental irritants, toxins and PAMPs (Pathogen associated molecular patterns)

(Kate Schroder et al., 2010). It consists of NLRP3 scaffold, ASC adaptor and caspase-1(Kate

Schroder et al., 2010). A wide variety of exogenous factors can activate the NLRP3 inflammasome

including ATP, Asbestos, Silica, uric acid and amyloid (Cassel et al., 2008; S. Mariathasan et al.,

2006; Martinon et al., 2006).

NLRP3 serves as a platform for the maturation and secretion of IL-1B and IL-18 in a

caspase-1 dependent manner (Gurung & Kanneganti, 2016; Shaw et al., 2011). Intriguingly, in a

recent report fibrillary alpha synuclein was found to activate IL-1B production in an NLRP3

dependent manner in monocytes (Codolo et al., 2013) Likewise the expression of NLRP3 related

markers were found to be upregulated around the site of injection in the LPS and 6-OHDA

intracerebral rat model. Notably, caspase-1 inhibitor, Ac-YVAD-CMK inhibited the mRNA and

protein expression of NLRP3 inflammasome related markers. The inhibition of NLRP3

inflammasome was found to positively correlate with the preservation of dopamine neurons in the

substantia nigra (Mao et al., 2017). Thus, these studies provide further support for the positive

immunomodulatory role of NLRP3 inflammasome in neurodegenerative diseases including PD.

199

Microglial cells mediate many inflammatory responses through activation of various

intracellular signaling molecules such as tyrosine kinases (Y. S. Yi et al., 2014a). Syk is a newly

identified non-receptor tyrosine kinase that belongs to SYK family that participates in various

physiological and pathological processes (Berton et al., 2005; S. H. Choi et al., 2009; Rogers et

al., 2005). Syk is a non-receptor tyrosine kinase, which has two Src homology domains (SH2) and

a kinase domain (Futterer et al., 1998; Mocsai et al., 2010; Taniguchi et al., 1991). These domains

are inter-connected by inter-domains: A and B (Futterer et al., 1998; Mocsai et al., 2010; Taniguchi

et al., 1991). Hematopoietic cells, macrophages, neuronal cells, fibroblasts, epithelial cells and

vascular endothelial cells express Syk in varying proportions (S. Yanagi et al., 2001; S. Yanagi et

al., 1995). It has been shown to play an important role in immune signaling, autoimmune diseases,

microglial activation, osteoclast maturation, platelet development and hematological malignancies

(Mocsai et al., 2010). Syk is regarded as a major upstream regulating kinase responsible for NLRP3

inflammasome activation in immune cells (F. Laudisi et al., 2014). Syk has been believed to cause

phosphorylation of apoptosis associated speck-like protein containing a CARD (ASC) thus

promoting caspase-1 activation thereby, resulting in NLRP3 inflammasome mediated production

of pro-inflammatory cytokines like IL-1 beta and IL-18 (Gross et al., 2009; H. Hara, K. Tsuchiya,

I. Kawamura, R. Fang, E. Hernandez-Cuellar, Y. Shen, J. Mizuguchi, E. Schweighoffer, & V.

Tybulewicz, 2013; Y. C. Lin et al., 2015). Nigericin, Mono sodium urate and Alum has been

known to cause Syk mediated inflammasome activation in LPS primed peritoneal macrophages

(F. Laudisi et al., 2014).

The recently characterized TNF superfamily member include the TNF-like weak inducer

of apoptosis (TWEAK) and its receptor fibroblast growth factor inducible 14 (Fn14) receptor

(Chicheportiche et al., 1997; Wiley et al., 2001).TWEAK is a cytokine that is widely expressed in

200

astrocytes, neurons, and macrophages and is upregulated in many neurological diseases, namely,

Stroke, Multiple Sclerosis Encephalomyelitis and Parkinson Disease (J. A. Winkles, 2008; Yepes,

2007). Moreover it has been implicated in variety of biological responses like inflammation,

migration, angiogenesis and apoptosis (Burkly et al., 2007). TWEAK-Fn14 signaling axis has been

linked to NF-kB activation via both canonical and non-canonical pathways involving cIAP1 and

increased transcription of the downstream mediator cyclooxygenase-2 which has been shown to

be upregulated in PD brains and in experimental models of PD (Teismann et al., 2003). In fact in

a recent report expression of TWEAK was found to be upregulated transiently in the striatum of

MPTP treated mice suggesting that TWEAK might an important component of MPTP-induced

neurotoxicity and thereby PD neuropathology (Mustafa et al., 2016a). Moreover, it is plausible

that TWEAK-induced inflammatory response may increase the vulnerability of dopaminergic

neurons to oxidative stress-induced neuronal injury. Thus far, TWEAK mediated downstream

signaling response upon microglial activation remain poorly characterized.

In the present study, we investigated the molecular basis of TWEAK-induced

inflammatory response. Our central hypothesis is that, Tweak causes NLRP3 inflammasome

activation via the activation of SYK kinase in an oxidative stress-dependent manner in the

microglia. Our results revealed that, Tweak causes microglia activation through Syk/NLRP3 axis.

Furthermore, our study highlights the anti-inflammatory effect of R788 in the TWEAK in vitro

cell culture model. Our results suggest that TWEAK induced Spleen Tyrosine Kinase activation

might be a critical upstream signaling event that elicits NLRP3 inflammasome activation

presumably via an ER stress (ERS) and autophagolysosomal system (ALS) dependent manner.

201

Materials and Methods

Chemicals and Reagents

Lipopolysaccharide (LPS) (Escherichia coli 0111: B4, endotoxin content 6.6000000

EU/mg) and β-actin were obtained from Sigma-Aldrich (St Louis, Mo, USA). R788 Syk inhibitor

was purchased from Santa Cruz Biotechnology, Dallas, TXBiotechnology). Recombinant Tweak

was obtained from Pepro-Tech (Rocke Hill, NJ, USA). IL-1 beta, IL-18 and TNF alpha ELISA

were purchased from eBiosciences (San Diego, California). PCR primers for IL-1 beta, IL-18 and

TNF alpha, DCFDA-H2 kit for ROS assay, RPMI, FBS, l-glutamine, penicillin, streptomycin,

were purchased from Invitrogen (Gaitherburg, MD). JC-1 Dye was purchased from Thermo-Fisher

Scientific (New Hampshire, USA). Bradford protein assay kit was purchased from Bio-Rad

Laboratories (Hercules, CA, USA). NLRP3, Caspase-1, IL-1 beta, ER stress markers (CHOP,

Phospho-PERK, Phospho-elf and GRP-94), Autophagy Markers (LC3, Beclin, P62, P-AMPK and

ATG5-12), Phospho-JNK and Phospho-P38 were purchased from Cell Signaling Technology,

MA, USA.

BV2 Cell Culture

The BV-2 immortalized cell line was obtained from Dr. Sanjay Maggirwar (University of

Rochester Medical Center, Rochester, NY, USA) and was grown and routinely maintained in 10%

RPMI at 37°C and 5% CO2. BV2 cells has been used for studying the neurotoxic

mechanisms related to Parkinson Disease in our laboratory. BV2 cells were grown in RPMI 1640

medium supplemented with 10% fetal bovine serum, 2mM L-glutamine, 50U penicillin and

50μg/ml streptomycin. Cell cultures were maintained under 5% CO2 at 37°C as previously

described. The media is changed every few days. After treatment, cells were collected by

202

trypsinization, spun down at 200 g for 5 min, and washed with ice-cold phosphate-buffered saline

(PBS). The lysates from the cell pellets were appropriately used for various assays.

Western Blot Analysis

Treated cells were collected and lysed with RIPA buffer (Sigma) in ice and sonicated for

15 seconds on ice for each sample. Samples were centrifuged at 14,000 g for 15 min at 4°C.

Supernatants were collected and stored at −80°C until use. Protein estimation was done using

Bradford reagent and samples with equal amount of protein were separated on 8–15% SDS-PAGE,

depending on the molecular weight of the target protein. Later they were transferred onto

nitrocellulose membranes by electro-blotting at 110V for 90 min at 4 C (Ice-cold transfer). Next,

membranes were blocked briefly for 1h by incubating them in blocking buffer and later incubated

with primary antibodies overnight at 4°C. To ensure equal protein loading, β-actin (1:10,000) was

used as loading control. Membranes were washed several times and incubated with specific

fluorescent labelled secondary antibody for 1 h at room temperature. Membranes were scanned

using the Odyssey IR Imaging system (LICOR) and data were analyzed with Odyssey 2.0

software.

Mitochondrial Membrane Potential (MMP)

Changes in the mitochondrial membrane potential was detected by 5,5’, 6.6’- tetrachloro-

1,1’, 3,3’-tetraethyl-benzimidazolcarbocyanineidide (JC-1 dye) (Molecular Probes, Thermo Fisher

Scientific) a cataionic fluorescent lipophilic dye. In depolarized mitochondria, monomeric form

shows strong green fluorescence with excitation and emission wavelength at 485 and 535 nm

respectively. In healthy mitochondria, aggregated monomers usually display strong red

fluorescence at excitation and emission wavelengths at 560 and 595 nm, respectively. The shift in

ratio of fluorescent intensity of JC-1 aggregates (Red fluorescence) to fluorescent intensity of

203

monomers (green fluorescence) is used as an indicator of depolarization of mitochondrial

membrane potential.

ELISA Assay

Microglia were plated in a 96-well plate. Later supernatant was collected and was used for

determination of IL-1β, IL-18 and TNF alpha concentrations using ELISA kit (ebiosciences) as

per manufacturer’s instructions.

ROS Assay

Changes in intracellular ROS was determined using CM-H2DCFDA, a fluorescent

probe, by following the protocols as suggested by manufacturer instructions (Molecular Probes,

Thermo Fisher Scientific). BV2 microglial cells were plated in 96-well plates, post treatment,

treatment media was removed and cells were washed with PBS and incubated with 1uM CM-

H2DCFDA dye dissolved in HBSS for 1 hr. Later, cells were washed with PBS and ROS

production was assessed using a microplate reader with excitation 485 nm and emission 530 nm

respectively.

Griess Assay

Nitric oxide production was assessed by quantification of nitrate in the supertanant with

the help of Griess assay. Cells were plated in a 96 well plate and treated with Tweak 100 ng, TNF

30 ng and LPS 1ug/ml. Post treatment, 100 ul of supernatant was collected and mixed with equal

volume of Griess agent and incubated in a plate shaker for color development for 15 min. The

absorbance at 450 nm was measured using microplate reader and nitrate concentration was

assessed from sodium nitrite standard curve (N. Panicker, H. Saminathan, H. Jin, M. Neal, & D.

S. Harischandra, 2015).

204

qRT-PCR

RNA isolation from treated BV2 microglial cells was performed using the Absolutey RNA

Miniprep kit (Trizol Reagent). Later, extracted RNA was used for cDNA synthesis by reverse

transcription with the AffinityScript qPCR cDNA synthesis system (Agilent Technologies) as per

the manufacturer's instructions. Quantitative SYBR Green PCR assays for gene expression were

performed using the RT2 SYBR Green Master Mix with pre-validated primers (SA Biosciences).

Master Mix was used to perform qRT-PCR analysis with verified primers TNF-α (catalog

#PPM57735E), IL-1β (catalog #PPM57735E) and IL-18 (catalog #PPM57735E). The mouse 18S

rRNA gene (catalog #PPM57735E) was used as the housekeeping gene for normalization. The

fold change in gene expression was determined by the ΔΔCt method using the threshold cycle (Ct)

value for the housekeeping gene and the respective target gene of interest in each sample.

Lysosomal PH

Changes in Lysosomal pH was characterized using the ratiometric lysosomal pH probe

LysoSensor Yellow/Blue dextran (Invitrogen L22460). At the end of the treatment period, cells

were loaded with 1 mg/ml LysoSensor and incubated overnight. Cells were then washed,

trypsinized and re-suspended in PBS. The fluorescence was monitored using the fluorescent

microplate reader with emission wavelengths at 535 nm and 430 nm with excitation at 340 nm.

The ratio of emission at 535 nm/430 nm was used for assessing lysosomal pH for each sample.

Syk Kinase Assay

Post treatment, treated cell lysates were added to ice cold master-mix consisting of: 5 μl

10X kinase buffer (200 mM HEPES, pH 7.4, 10 mM MnCl2, 10 mM MgCl2, 10 mMDTT, 10 mM

Na3VO4, 10 mM NaATP), 5ug Syk Kinase peptide substrate biotin labelled (Ana-Spec) and final

volume adjusted with ddH2O to 50 μl. Recombinant Syk enzyme (1 μg/reaction) was used as a

205

positive control. Reactions were incubated at 30°C for 30 min. Reaction was stopped by addition

of 950 μl 2% ice-cold BSA. Next, 100 ul of supertanant was added to streptavidin coated 96 well

plate and incubated for 3h and later plates were washed for 3 times. Furthermore, 4G10 anti-

phosphotyrosine antibody (Millipore) diluted 1:1000 in 5% milk/TBS was added to each well,

allowed to incubate for 1 h at room temperature on a shaker. Plates were washed for 3 times with

wash buffer, and anti-IgG2b HRP-conjugated secondary (Southern Biotech) diluted 1: 1000 in5%

milk/TBS was added to each well and incubated for 1 h at room temperature followed by washing

with wash buffer for 3 times. Lastly, plates were developed with Horseradish Peroxidase Substrate

Kit (Biorad) and color was allowed to develop for 15 min. Plates were read with an Infinite m200

plate reader (Tecan) at 405 nm (Schlatterer et al., 2011).

Animal Study

6-8-week-old C57 black mice were obtained from Charles River and housed under

standard conditions at 22 C with 30% relative humidity with 12h light cycle as per IACUC

protocol. Mice were randomly assigned into three different groups (Control, Tweak and Tweak

and Syk inhibitor). Piceatannol (100 mg/kg) was administered twice 12h before and 1h before

Tweak treatment (Y. He, L. Xu, B. Li, Z. N. Guo, et al., 2015; Yukiya Suzuki et al., 2013). Acute

intra-striatal Tweak induced neuroinflammation model of PD was used for this study (Polavarapu

et al., 2005). Mice were given intra-striatal injections of 2 ul Tweak (1 ug/ul) using stereotaxic

surgery (W. B. Haile, R. Echeverry, F. Wu, et al., 2010; Polavarapu et al., 2005). 24 h post

treatment, mice were sacrificed and striatum were collected and stored at 80 C. Later tissue lysates

were immunoblotted for the protein of interest.

206

Syk SiRNA Transfection

Transfection of BV2 cells was done using Amaxa Nucleofector Kit (Lonza). Briefly, Bv2

cells were mixed with 100 ul transfection buffer (400 uM ATP-disodium, 600 uM magnesium

chloride, 100 uM potassium hypophosphate, 20 uM sodium bicarbonate and 5 uM glucose. Later

Syk SiRNA and control SiRNA (Santa Crux BioTechnology, CAT # SC-44328 and CAT # SC-

44230) were added to the transfection mixture. Next, transfection of cells was done with

electroporation using A23 program of Lonza Nucleofector 2b Devise. Finally, cell were incubated

for 48-72 h followed by treatment with Tweak for another 24h.

Data Analysis

Results are analyzed with PRISM 6.0 software (Graph Pad, San Diego). Results are

presented as mean ± S.E.M. P values were assessed using one-way ANOVA followed by post hoc

Tukey test (PRISM software) in order to compare between treatment groups. P values < 0.05 were

considered significant.

Results

Temporal Pattern of Spleen Tyrosine Kinase Activation and Stress Granule Formation in

Microglial Cells in Response to Inflammogens

A growing body of evidence supporta a role for SYK in the activation of inflammatory

cells including microglial cells. For example Spleen Tyrosine Kinase is involved in LPS induced

intracellular signaling pathways and pro-inflammatory responses in macrophages (Ulanova et al.,

2007). Additionally, in cell culture models of Alzheimer Disease amyloid fibrils activate spleen

tyrosine kinase in primary microglial cells to induce pro-inflammatory responses (Combs et al.,

1999; S. Ghosh et al., 2015).Therefore, we utilized the well characterized BV2 microglia model,

which has been routinely used for the study of PD associated neuro-inflammation (R. Gordon et

al., 2016). In the present study, we sought to investigate whether SYK is activated in LPS or

207

TWEAK treated microglial cells. BV2 microglial cells were either exposed to LPS or TWEAK

for increasing duration (3h, 6h, 12h, 18h and 24h) and expression of spleen tyrosine kinase was

determined using WB analysis.

As shown in (Fig 1A), we found that both LPS and Tweak combination treatment

induced a time dependent increase in Spleen Tyrosine Kinase expression in Bv2 microglial cells.

A similar trend was evidenced in Tweak alone treated cells (Fig 1B). We further determined

whether the increase in SYK protein levels is accompanied by a concomitant increase in SYK

kinase activity. To this end we performed enzymatic kinase activity assays in BV2 cells stimulated

with TWEAK. For this purpose, R788, Syk inhibitor pretreated cells were stimulated with Tweak.

As anticipated TWEAK treated BV2 microglial cells showed a time dependent increase in the

activation of SYK kinase at the end of 12 and 24h post drug treatment. In fact, a pronounced

upregulation was observed at 24h post TWEAK treatment. Furthermore R788, a SYK kinase

inhibitor markedly attenuated SYK kinase activity (63 % reduction) in TWEAK treated cells

further confirming the role of SYK in inflammagen-induced microglial activation response (Fig

1C). Taken together, these results indicate that, Tweak-induced microglial activation response may

be linked to SYK activation.

In a previous report SYK was found to localize to stress granules (SGs) in primary

microglial cells stimulated with A beta or sodium arsenite (S. Ghosh et al., 2015). To examine

whether stress granules are formed in response to TWEAK, we treated BV2 microglia with

TWEAK for the indicated duration (0-24h). At the end of the drug treatment period cell lysates

were prepared and immunoblotted for the SG marker G3BP1. A marked time dependent increase

in G3BP1 was evidenced in TWEAK treated cells (Fig 1D). Notably a positive correlation between

SYK activation and G3BP1 upregulation was evidenced in TWEAK treated cells as compared to

208

vehicle treated cells suggesting that SYK activation may promote the formation of SGs to

sequester aggregate prone proteins.

Temporal Pattern of Activation of NLRP3 Related Markers in LPS Primed TWEAK

Stimulated Microglial Cells

NLRP3 (Nucleotide-binding domain and leucine rich repeat containing family, pyrin

domain containing 3) has been shown to be a key regulator of innate immunity and activated in

response to diverse stimuli including microbial infection or exposure to sterile inflammatory

stimuli (H. Guo et al., 2015; Gurung et al., 2016; Lamkanfi et al., 2012; Kate Schroder et al.,

2010; Shaw et al., 2011). In this context caspase-1 mediated proteolytic processing of pro-IL-1b

and pro-IL 18 to their active forms has been linked to NLRP3 inflammasome activation. LPS is a

widely studied inflammagen that has been shown induce the expression of pro IL-1B via an NF-

kB dependent mechanism in microglial cells (C. Dostert et al., 2008). Therefore, microglial cells

were primed with LPS to promote NF-kB activation which has been shown to serve as signal-1for

NLRP3 inflammasome activation. Additionally, it recapitulates excessive microglial activation

response observed in PD brains (Machado et al., 2016; S. R. Subramaniam & H. J. Federoff, 2017;

Q. Wang et al., 2015). In the initial set of experiments, we determined the temporal pattern of

activation of NLRP3 related markers in either TWEAK or LPS/TWEAK treated cells. BV2 cells

were exposed to TWEAK (100 ng/ml) for increasing duration (3h, 6h, 12h and 24h) in the presence

or absence of LPS (1 ug/ml, 3h priming). As shown in Fig 2A-D, NLRP3, caspase-1, IL-1 beta

and IL-18 expression was upregulated in LPS and Tweak stimulated BV2 microglial cells.

Interestingly, we also noticed a similar trend in BV2 cells exposed to Tweak alone (Fig 2E-G).

Given that TWEAK alone elicited the activation of NRLP3 related markers in the absence of a

priming step we next studied the influence of TWEAK on pro-inflammatory factor secretion in

microglial cells. For this reason, we measured pro-inflammatory cytokine secretion after treatment

209

of primary microglial cells with Tweak at various time points (8h and 24h) using ELISA analysis.

We noticed a significant increase in the secretion of pro-inflammatory cytokines namely IL-1 beta

and TNF-alpha (Fig 2H) in Tweak stimulated BV2 microglia at 8h and 24h, respectively. To

further assess whether increased pro-inflammatory cytokine secretion in Tweak treated BV2

microglial cells was due to increased gene transcription, we performed gene expression analysis

of pro-inflammatory cytokines using Real time PCR. We found that, Tweak significantly up-

regulated mRNA levels of IL-1 beta, IL-18 and TNF-alpha in a time dependent manner (Fig 2I) in

BV2 microglial cells. Taken together, our studies suggest that TWEAK by itself as well as LPS

and TWEAK combination results in a heightened inflammatory response characterized by marked

upregulation of pro-inflammatory markers including NLRP3 inflammasome related markers.

Activation of SYK and NLRP3 Related Markers in Primary Microglia Upon TWEAK

Stimulation

Next, we validated TWEAK-induced pro inflammatory response in primary microglial

cells. For these studies, primary microglia were isolated from brains of mice either post-natal day

(PND) 2 or 3. Primary microglial cells were treated with TWEAK for the indicated duration (3,

6, 12 &24h) and immunoblotted for SYK, NLRP3 related markers such as caspase-1, IL-1b,

NLRP3 as well as redox sensitive kinase PKC delta (PKCd). A time dependent upregulation of

SYK and accompanying enhancement of NLRP3 related markers and PKC delta was evidenced in

TWEAK treated cells (Fig 3A-E). Interestingly, a marked increase in PKC delta and NLRP3

inflammasome activation as well SYK was observed at 24h post drug treatment. Together, these

results demonstrate that redox sensitive kinases are upregulated upon microglial activation with

an inflammatory stimulus.

210

Tweak Promotes ROS Generation and Dissipation of Mitochondrial Membrane Potential

(MMP) in BV2 Microglial Cells.

Mitochondria mediated ROS (Reactive Oxygen species) production and has been shown

to function as signal-2 for NLRP3 inflammasome activation in immune cells (E. K. Jo et al., 2016;

J. Yu et al., 2014). We next determined whether NLRP3 inflammasome activation is associated

with mitochondrial ROS generation in TWEAK treated cells. For this purpose, BV2 microglial

cells were treated with Tweak at the indicated time points and ROS production was examined

using redox sensitive CM-H2DCFDA dye. A time dependent increase in ROS generation was

evidenced whereby ROS generation was observed as early as 3h post TWEAK treatment and

remained elevated during the entire treatment duration (Fig- 4 A).

Previous studies have shown that, mitochondrial impairment is commonly associated

with NLRP3 inflammasome activation in immune cells. In this context, Caspase-1 induced

dissipation of mitochondrial membrane potential and associated mitochondrial damage and

dysregulation of mitophagy has been linked to the activation of the NLRP3 inflammasome (J. Yu

et al., 2014). In order to determine whether TWEAK induced ROS production is associated with

loss of mitochondrial membrane potential we assessed mitochondrial membrane potential using

redox sensitive JC-1 dye, a catatonic fluorescent lipophilic dye. As shown in the Fig 4B, Tweak

and LPS in combination induced a sustained reduction in MMP quantification of mitochondrial

membrane potential (MMP) as assessed via spectrometric plate reader analysis. Together these

data suggest that TWEAK-induced excessive ROS generation presumably via loss of MMP and

associated mitochondrial dysfunction may lead to the activation of NLRP3 inflammasome.

Previous reports have demonstrated that SYK dependent activation of NADPH oxidase is

critical for autophagosome formation which in turn enhances fungicidal activity in macrophages

(Tam et al., 2014). In another study SYK was found to regulate OxLDL-induced ROS generation

211

in macrophages (S. H. Choi et al., 2015). We therefore examined whether NOX-2 activation

requires SYK. We depleted endogenous levels of SYK via siRNA mediated gene KD. NOX-2

activation in TWEAK stimulated BV2 microglial cells was reduced in SYK SiRNA transfected

cells as compared to scramble transfected cells (Fig 4C). Together these data provide new insights

into the mechanisms by which TWEAK regulates ROS generation in the microglia. Thus, hyper

activation of NOX via SYK may partially contribute to TWEAK-induced oxidative stress

response.

Tweak Induces ER Stress Response (ERS) in BV2 Microglial Cells

After having established the expression of SYK and increased ROS generation in the BV2

microglia, the next step was to test the ability of TWEAK to induce ER stress response (ERS)

Recent studies have demonstrated that ER stress can-induce NLRP3 inflammasome activation (D.

N. Bronner et al., 2015; Lerner et al., 2012). For example ER stress has been shown to cause the

release of mitochondria derived release of damage associated molecular patterns such as ROS

which in turn has been linked to NLRP3 inflammasome activation (Iyer et al., 2013; R. Zhou et

al., 2011). Unfolded protein response sensors such as IREa, ATF6 and PERK have been identified

as major sensors of ER stress response, an adaptive program that has been shown to regulate cell

fate in cells subjected to cellular stress. To determine whether ER stress response is induced during

TWEAK-induced NLRP3 inflammasome activation we investigated the levels of ER stress

markers in TWEAK treated microglial cells. Robust activation of ER stress markers including

CHOP, GRP-94, Phospho-Elf alpha, Phospho-PERK and ATF-4 expression were seen in TWEAK

treated BV2 microglial cells in a delayed manner (Fig 5 A-E). Thus Tweak-induced the activation

of ER stress markers in microglial cells as previously observed in cells subjected to microbial

212

infection (Denise N. Bronner et al., 2015; Martinon, 2012). Overall, the results suggest that

TWEAK induces ERS in BV2 microglial cells.

Autophagosomal Lysosomal System (ALS) Modulation in TWEAK Stimulated BV2

Microglial Cells.

Autophagy represent a catabolic mechanism that sequesters portions of cytoplasm

including damaged organelles and macromolecules into double membrane vesicles known as

autophagic vacuoles and eventually delivered to the lysosome for degradation leading to the

recycling of macromolecules (D. J. Klionsky et al., 2016). Studies indicate that, autophagy

negatively regulates NLRP3 inflammasome activation and pro-inflammatory cytokine production

in immune cells (Tatsuya Saitoh et al., 2016; Yuk & Jo, 2013; Z. Zhong et al., 2016). It has been

proposed that impairment in autophagic mechanisms may lead sustained activation of NLRP3

inflammasome in immune cells (Tatsuya Saitoh et al., 2016; Yuk et al., 2013; Z. Zhong et al.,

2016).

To determine the relationship between TWEAK-induced inflammatory response and

autophagic mechanism, the BV2 microglial cell line was treated with TWEAK for increasing

duration and the induction of autophagic markers were assessed via WB analysis. The extent of

accumulation of autophagic markers including LC3 II, p62, but not beclin 1 (upregulation observed

in a delayed manner-24h) increased in a time dependent manner (FIG 6A-E) indicating an impaired

autophagic flux in microglial cells treated with TWEAK. Alternatively, the accumulation of AV

related markers may be attributed to lysosomal dysfunction. These studies indicate a positive

association between impairment in autophagic function and NLRP3 inflammasome activation in

microglia treated with TWEAK.

213

Next, we determined whether lysosomal damage is linked to NLRP3 inflammasome

activation in Tweak treated microglial cells. Earlier studies indicate that, NLRP3 inflammasome

activation and IL-1 beta secretion in LPS treated macrophages occurs through the release of

cytoplasmic lysosomal Cathepsin B originating from altered lysosomal membrane permeability

(K. Weber & J. D. Schilling, 2014). Moreover lysosomal membrane damage and resultant release

of cathepsin-B into the cytosol has been implicated in NLRP3 inflammasome activation in

endothelial cells and immune cells exposed to an inflammatory stimuli (Y. Chen et al., 2015; E.

K. Jo et al., 2016). Therefore, we assessed the expression of lysosomal cathepsins in Tweak treated

BV2 microglial cells. For this purpose, BV2 microglia was incubated with Tweak for the indicated

duration (3h, 6h, 12h and 24h). Post incubation, expression of lysosomal cathepsins was

determined using WB analysis. As shown in the Fig 6 F-G we found out that Cathepsin-B,

Cathepsin-D is upregulated at 24h following Tweak treatment in Bv2 microglia. In fact, increase

in lysosomal PH results in release of cathepsin-B and subsequent activation of NLRP3

inflammasome in pneumococcal meningitis (Hoegen et al., 2011). Since lysosomal PH disruption

is associated with NLRP3 inflammasome activation in immune cells, we next characterized

lysosomal pH in Tweak treated microglial cells. BV2 microglial cells were treated with Tweak at

various time points (6h, 12h and 24h) and lysosomal pH was measured by spectro-photometric

plate reader analysis using a redox sensitive Lyso-Sensor dye. As shown in the Fig 6H, lysosomal

pH is increased in Tweak treated microglial cells in time dependent manner indicating that

lysosomal acidity is altered. Taken together, our studies suggest that impairment in lysosome

mediated clearance of autophagic vacuoles owing to loss of lysosomal acidity and associated loss

of membrane permeability might partly contribute to the activation of NLRP3 inflammasome in

microglial cells challenged with TWEAK.

214

SYK Regulates The Activation of NLRP3 Inflammasome in Tweak Treated Microglial Cells

Studies had shown that Spleen tyrosine kinase is key player in NLRP3 inflammasome

activation in immune cells. Spleen tyrosine kinase is involved in NLRP3 inflammasome activation

in yeast cells, macrophages and neutrophils in response to microbial infection (Mocsai et al.,

2010). We therefore sought to investigate the role of SYK in NLRP3 regulation in TWEAK treated

macrophages. We investigated whether pharmacological inhibition via R788 could suppress

NLRP3 inflammasome activation in response to pro-inflammatory stimuli such as TWEAK.

Genetic deletion of SYK results in early embryonic lethality, precluding the inclusion of SYK -/-

mice in the current study (Jakus et al., 2010; Mocsai et al., 2010). Hence, we tested the anti-

inflammatory effects of R788, fostamatinib sodium, a small molecule of Syk inhibitor that has

shown efficacy in animal models of Asthma, Lymphoma, Glomerulonephritis and Diabetes

(McAdoo, 2011). To determine the effects of SYK inhibition on TWEAK-induced NLRP3

inflammasome activation, BV2 microglial cells were pretreated with R788 (Syk inhibitor) for 1h

followed by treatment with Tweak for 24h. Post treatment, NLRP3 inflammasome related markers

expression was determined using western blot analysis. As shown in Fig. 6A, R788 pretreatment

resulted in decreased activation of NLPR3 (Fig 7 A) and Caspase-1 (Fig 7 B) as well as IL-1 beta

(Fig 7 C) in TWEAK stimulated microglial cells. To further ascertain the regulatory role of syk in

inflammasome activation in microglial cells, we used RNAi approach. For these studies, BV2

microglial cells were initially transfected with scrambled or Syk siRNA. At the end of 72h

following transfection the SYK expression was evaluated using WB analysis. Greater than 70%

reduction in SYK levels was observed in SYK siRNA transfected cells as compared with

scrambled siRNA transfected microglial cells validating down regulation of SYK expression (Fig.

215

7 D). BV2 microglial cells were treated with Tweak for another 24h following transfection with

the respective siRNA. Post treatment, NLRP3 inflammasome related markers expression were

determined using western blot analysis. Immunoblot analysis revealed that TWEAK-induced

induction of NLRP3 related markers as well as pro-inflammatory cytokine levels were diminished

by siRNA mediated knock down of SYK (Fig 7 E-F). Taken together, these results confirmed that

TWEAK-induced SYK regulated activation of NLRP3 related markers in BV2 cells.

Involvement of SYK in TWEAK-induced MAPK Activation

MAPK family include the well-studied ERK1/2, JNK, and p38 MAPK which has been

linked to the induction of numerous cellular functions including inflammatory response (Johnson

& Lapadat, 2002). In several cell types activation of MAPK family members have been linked to

the activation of iNOS, COX-2 and proinflammatory cytokines (Dai et al., 2011; Frazier et al.,

2009; Hevia et al., 2004; J. Y. Kim et al., 2006). We next examined whether TWEAK-induced

the activation of MAPK signaling pathway in BV2 microglial cells. As anticipated Tweak induced

the upregulation of all three MAPKs as early as 30 min in BV2 microglial cells (Fig. 8A-C). In

fact inhibition of MAPK via antioxidants such as resveratrol has been shown to inhibit NO, PGE2,

iNOS, COX-2 and pro-inflammatory cytokine (Zhong et al., 2012). Moreover, SYK-induced

MAPK8/JNK2 activation in response to OxLDL has been shown to play an important role in

MHC-II expression in macrophages (S. H. Choi et al., 2015). Furthermore, JNK and SYK have

been shown to be essential for NLRP3 inflammasome activation in an in vivo model of MSU and

alum-induced peritonitis (H. Hara, K. Tsuchiya, I. Kawamura, R. Fang, E. Hernandez-Cuellar, Y.

Shen, J. Mizuguchi, E. Schweighoffer, & V. Tybulewicz, 2013). To test whether the suppressive

effects of R788 on NLRP3 inflammasome activation occurred via MAPK signaling pathway in

TWEAK treated microglia, BV2 microglial cells were pretreated with the SYK inhibitor R788 and

216

subsequently incubated with TWEAK for another 24h. As shown in (Fig. 8D-E), MAPK

activation markers pJNK, and p38MAPK in response to TWEAK were decreased in R788

pretreated cells. Thus, our data suggest that SYK mediated activation of MAPK elicits a pro-

inflammatory response via increased NLRP3 inflammasome activation in TWEAK treated

microglial cells.

SYK Regulates TWEAK-induced Impairment In Autophago Lysosomal Function and GSK

3b Activation

Because SYK has been shown to regulate autophagy in macrophages following microbial

infection (S. H. Choi et al., 2015), we next investigated whether it is involved in the regulation of

autophago lysosomal function in TWEAK treated BV2 microglial cells. For this purpose, BV2

microglial cells were transfection with the scrambled and Syk siRNA and subsequently incubated

with Tweak. Post treatment, ALS markers expression were determined using western blot analysis.

As shown in (Fig 9 A-B) siRNA mediated knock down of SYK attenuated TWEAK-induced

impairment in ALS function in BV2 microglial cells. These data indicate that SYK may contribute

to the regulation of ALS function in response to TWEAK in BV2 microglial cells.

Next, we explored whether Tweak induces lysosomal damage in Bv2 microglial cells

through a Syk dependent mechanism. SYK mediated lysosomal dysfunction has been implicated

in the response to infection in innate immune cells (J. He et al., 2005; Y. Q. Lu et al., 2017). For

this purpose, BV2 microglial cells were pretreated with R788 (Syk inhibitor) for 1h followed

treatment with Tweak for 24h. Post treatment, lysosomal cathepsin-B expression was determined

using western blot analysis. As shown in (Fig 9 C) R788 pretreatment resulted in decreased

activation of cathepsin-B in Tweak stimulated BV2 microglia. To further ascertain the regulatory

effects of Syk on lysosomal damage in inflammogen treated microglial cells, we used RNAi

approach. Immunoblot analysis revealed downregulation of cathepsin-B (Fig 9D) in TWEAK

217

stimulated Syk Si RNA transfected cells as compared to scrambled siRNA transfected cells. Taken

together, our studies indicate that, Spleen tyrosine kinase is an important regulator of ALS function

in Tweak stimulated BV2 microglial cells.

Glycogen synthase kinase (GSK)-3 is a multifunctional kinase that is differentially

regulated by the phosphorylation of serine or Tyr. For example, phosphorylation of serine 9 in

GSK-3B has been shown to inhibit its activation whereas the enzymatic activity of GSK-3 beta is

enhanced by phosphorylation of TYR 216 (Jope et al., 2007). Furthermore numerous studies have

identified that ER stress is a positive regulator of GSK-3B which in turn can lead to an elevated

inflammatory response (Resende et al., 2008). Moreover, SYK has been shown to negatively

regulate TLR4-mediated IL-10 production while promoting the generation of pro-inflammatory

cytokines (TNFa and IL-6) via modulation of GSK 3b (H. Yin et al., 2016). In the present study,

we sought to determine whether SYK can modulate GSK-3B activation in TWEAK treated

microglial cells. As expected immunoblotting studies confirmed that TWEAK induced Tyr216

GSK-3B phosphorylation. Consistent with a previous study demonstrating that SYK deficiency

promotes anti-inflammatory response in dendritic cells (D. Paris et al., 2014; H. Yin et al., 2016).

SYK inhibition via R788 markedly down regulated TWEAK-induced GSK-3b activation (Fig 9E).

Likewise, siRNA mediated knockdown of SYK in Bv2 microglial cells resulted in reduced

expression of Tyr216 GSK-3B in response to TWEAK in Bv2 microglial cells (Fig 9F). Taken

together, our studies suggest that SYK is a critical determinant of GSK 3 beta activation and ALS

dysfunction in BV2 cells upon TWEAK stimulation.

218

Evaluation Of The Therapeutic Efficacy of Syk Inhibitor (Piceatannol) In a TWEAK

Neuroinflammation Mouse Model.

To further investigate whether the data generated using in vitro models of TWEAK can be

validated in an in vivo scenario we utilized the intra striatal infusion neuro-inflammation model of

TWEAK. Additionally, we investigated the effects of SYK inhibitor Piceatannol in the afore

mentioned neuro-inflammation model. Previous studies have demonstrated the validity of using

the neuro-inflammation model of TWEAK to study TWEAK-induced inflammatory response (W.

B. Haile, R. Echeverry, J. Wu, et al., 2010; Polavarapu et al., 2005). Moreover, at the doses used

in the present study Piceatannol (Syk inhibitor) has been shown to downregulate Syk mediated

neuro-inflammation in mouse models of subarachnoid hemorrhage and ischemic stroke (Y. He, L.

Xu, B. Li, Z. Guo, et al., 2015; Y. Suzuki et al., 2013). To determine the effects of Piceatannol on

TWEAK-induced inflammatory response, mice were pretreated with Piceatannol (100 mg/kg

intraperitoneally) 15h and 1h before Tweak (2ul, 1 ug/ul) was stereotaxically injected unilaterally

into the striatum. Mice were sacrificed 24h after TWEAK administration and striatal tissues were

examined for the expression of neuro-inflammation related markers using Western blot analysis.

As shown in Fig 10 A-H, no significant difference in the striatal inflammatory markers between

control and TWEAK infused mice was evidenced. In a similar fashion Piceatannol pretreatment

treatment had no effect on TWEAK-induced modulation of Syk, Caspase-1, CHOP, LC3, Beclin,

GSK-3 beta Y216 and cathepsin-B expression in the striatum (Fig 10 A-H). At this time

optimization of the TWEAK neuro-inflammation model is needed and therefore we propose to

perform additional dose response studies to dissect TWEAK associated inflammatory response in

the striatum. Additionally, the dose of Piceatannol needs to be optimized to understand the

therapeutic effects of this drug in the in vivo TWEAK neuro-inflammation model.

219

Discussion

In the present study, we demonstrate the potential regulatory effects of SYK in TWEAK-

induced microglial activation response. Upon treatment of microglia with Tweak, a dramatic

upregulation of spleen tyrosine kinase coincided with NLRP3 inflammasome activation, pro-

inflammatory cytokine production, ER stress response (ERS), mitochondrial dysfunction, GSK-3

beta and ALS dysfunction. Interestingly, Syk inhibition downregulated NLRP3 inflammasome

activation and pro-inflammatory cytokine production in Tweak treated microglial cells. To further

identify the molecular mechanism of anti-inflammatory effects of R788 we investigated whether

R788 affected TWEAK-induced ERS and ALS dysfunction. Our studies revealed a positive

association between NLRP3 inflammasome inhibition and attenuation of ERS, ALS impairment

and mitochondrial dysfunction in cells that received a combination of TWEAK and R788. In our

in vivo studies, Piceatannol, a Syk inhibitor displayed a trend toward attenuation of TWEAK-

induced ALS modulation in an intracerebral Tweak neuro-inflammation mouse model although it

failed to reach statistical significance. Further studies are warranted to quantify the in vivo effects

of Piceatannol in the TWEAK Neuro-inflammation model. Our results highlight the pro-

inflammatory role of SYK in mediating TWEAK-induced NLRP3 inflammasome activation. A

potential mechanism by which TWEAK may induce NLRP3 inflammasome activation via SYK

by initiating downstream signaling events is illustrated in Fig 4.6.

Spleen Tyrosine Kinase is is 72 kDa non-receptor tyrosine kinase that plays an important

role in microglial activation and neuro-inflammation in immune cells (Y. S. Yi et al., 2014b). The

main trigger for activation of spleen tyrosine kinase is phosphorylation of ITAMs

(Immunoreceptor Tyrosine based Activation Motifs) (H. Hara, K. Tsuchiya, I. Kawamura, R.

Fang, E. Hernandez-Cuellar, Y. Shen, J. Mizuguchi, E. Schweighoffer, & V. Tybulewicz, 2013;

220

Turner et al., 2000; Y. S. Yi et al., 2014b). Its main functions include Innate pathogen recognition,

signaling tissue damage, inflammasome activation, bone metabolism, arterial thrombus formation,

vascular development, B-cell development, phagocytosis and gonadal development (Mocsai et al.,

2010; Tohyama & Yamamura, 2009; Y. S. Yi et al., 2014b). Consistent with a pro-inflammatory

role for SYK, TWEAK-induced robust activation of this enzyme in Bv2 and primary microglial

cells.

NLRP3 inflammasome is an intracellular multi-meric protein complex that becomes

activated by wide variety of pathogens, environmental irritants, toxins and PAMPs (Pathogen

associated molecular patterns) (Kate Schroder et al., 2010). It consists of NLRP3 scaffold, ASC

adaptor and caspase-1 (Kate Schroder et al., 2010). A wide variety of factors can activate the

NLRP3 inflammasome including ATP, Asbestos, Silica, uric acid and amyloid (Cassel et al., 2008;

Halle et al., 2008; S. Mariathasan et al., 2006; Martinon et al., 2006). NLRP3 plays an important

role in the generation of mature pro-inflammatory cytokines namely IL-1B and IL-18 in a caspase-

1 dependent manner in order to mount an innate immune response (Kate Schroder et al., 2010).

Accordingly, we demonstrated the upregulation of NLRP3, caspase-1, IL-18 and IL-1 beta via WB

analysis in Tweak and LPS treated BV2 microglial cells. A growing body of evidence supports a

role for SYK in the regulation of NLRP3 inflammasome upon exposure of inflammatory cells to

infectious agents (Gross et al., 2009; H. M. Lee et al., 2012; Shio et al., 2009). For example SYK

dependent ASC phosphorylation has been shown to promote NLRP3 inflammasome assembly and

activation (H. Hara, K. Tsuchiya, I. Kawamura, R. Fang, E. Hernandez-Cuellar, Y. Shen, J.

Mizuguchi, E. Schweighoffer, & V. Tybulewicz, 2013). Accordingly, Syk inhibitor (R788)

treatment resulted in attenuated expression of NLRP3 and caspase-1 in Tweak treated microglial

cells. Like-wise, siRNA mediated knockdown of Syk downregulated the expression of NLRP3

221

and caspase-1 in Tweak treated BV2 microglial cells. Syk induced NLRP3 activation is associated

with pro-inflammatory cytokine production including IL-1B and IL-18 in neutrophils and

macrophages (Feng et al., 2015; R. Lu et al., 2012; M. W. Ma et al., 2017; Miller et al., 2012).

Moreover, Syk inhibitors downregulated the pro-inflammatory cytokine production and

inflammatory responses in dendritic cells and immune cells (Aouar et al., 2016; Llop-Guevara et

al., 2015; Patterson et al., 2014). Accordingly, in the present study R788 induced the down

regulation of mRNA and protein levels of pro-inflammatory cytokines including IL-1B, IL-18,

and TNFa in Tweak treated primary microglial cells. To the best of our knowledge this is the first

study to demonstrate that SYK is a key regulator of NLRP3 inflammasome activation in microglial

cells in response to TWEAK.

The most common modes of NLRP3 activation includes K+ efflux, lysosomal damage and

ROS production (Cassel et al., 2008; C. Dostert et al., 2008; Halle et al., 2008; Hornung et al.,

2009; T. D. Kanneganti et al., 2006). Thus, in the present study, we investigated whether SYK

modulates the activation of ER stress, mitochondrial dysfunction, ALS dysfunction in Tweak

treated microglial cells. In innate immune cells such as microglia, SYK has been shown to play a

key role in receptor mediated generation of ROS and RNS (Y. S. Yi et al., 2014b). Moreover,

functional interaction of A beta with microglial cells has been shown to induce the phosphorylation

and activation of SYK thereby leading to ROS generation and neurotoxicity (Combs et al., 1999;

S. Ghosh et al., 2015; McDonald et al., 1997). Microglia stimulated with LPS trigger cytokine

secretion and ROS generation in a NOX2 dependent manner (S. Hong & Yu, 2017; L. Hou et al.,

2017; Kwon et al., 2017; L. Qin et al., 2004). Consistent with a role for SYK in oxidative stress

mechanisms, SYK inhibitors blocked the ability of chronically activated microglia to generate

ROS and RNS thereby blocking its neurotoxic effects (S. Ghosh et al., 2015). Moreover, ROS

222

triggered NLRP3 inflammasome activation have been identified as a proximal trigger although the

role of ROS remains controversial (Cassel et al., 2008; C. M. Cruz et al., 2007). Our results also

support a role for SYK in the regulation of NOX2 which is implicated in NLRP3 inflammasome

activation (Feng et al., 2015; M. W. Ma et al., 2017). Likewise, siRNA mediated KD of SYK

attenuated TWEAK-induced NOX2 activation suggesting that elevated NOX2 derived ROS may

be linked to NLRP3 inflammasome activation. Alternatively, we and others have demonstrated

that mitochondrial dysfunction leads to ROS production, release of mitochondrial DNA into the

cytosol eventually leading to NLRP3 inflammasome activation (L. Chen et al., 2015; Lawana et

al., 2017; Sarkar et al., 2017). Although, we demonstrated dissipation of mitochondrial membrane

potential, NOX2 activation and ROS production paralleled NLRP3 inflammasome activation in

Tweak treated BV2 microglial cells the contribution of mitochondrially derived ROS cannot be

entirely excluded. Thus, it is likely that concomitant activation of NOX2 and mitochondrial

dysfunction may lead to NLRP3 inflammasome activation in TWEAK treated microglial cells.

The expression levels of chaperone proteins including GRP78 and GRP 94 as well

as p-eiF2a are increased in response to unfolded protein response (UPR) and therefore have been

used as markers for ER stress response (Jain et al.). BV2 microglial cells treated with TWEAK

displayed an increase in the levels of the afore mentioned proteins, indicating that TWEAK induces

ER stress response (ERS). Owing to a positive correlation between increased SYK activity and

activation of ERS we hypothesized that TWEAK stimulation led to ERS via SYK activation.

Accordingly, our data indicate that TWEAK-induce ERS via SYK activation based on the fact

that, R788 and siRNA mediated knock down of SYK, both of which inhibited the induction of

ERS. ERS serves as an important triggering factor for NLRP3 inflammasome (Menu et al., 2012b;

J. Wang et al., 2015). ER stress sensor IRE1-alpha causes mitochondrial damage through caspase-

223

2 and Bid thereby leading to NLRP3 inflammasome activation and IL-1 beta production (D. N.

Bronner et al., 2015). Furthermore, in the UPR dependent pathway, ER stress related proteins IRE1

alpha and PERK leads to upregulation of Thioredoxin interacting protein (TXNIP) & CHOP;

thereby leading to NLRP3 inflammasome activation (D. N. Bronner et al., 2015; Lebeaupin et al.,

2015a; Lerner et al., 2012). A marked reduction in NLRP3 inflammasome markers positively

correlated with diminished ERS in a SYK dependent manner suggesting that TWEAK-induced

ER stress mechanism via SYK mediated mechanism may at least in part mediate NLRP3

inflammasome activation. Further study is required to clarify the impact of inhibition of ER stress

response on NLRP3 inflammasome activation under our experimental conditions.

Glycogen synthase kinase 3B (GSK-3b) has been shown to play a critical role in response

to bacterial infection. Depending on the cellular context, type of pathogen in which host bacteria

interaction occurs, GSK3B may elicits an inhibitory or stimulatory effect(Cortes-Vieyra et al.,

2012). For example, in LPS stimulated human monocytes GSK 3b was found to negatively

regulate the level of IL-1Ra, an anti-inflammatory cytokine while simultaneously increasing the

levels of IL-1B which was attributed to the modulation of extracellular signal related kinase 1/2

(ERK1/2) (Rehani et al., 2009). Furthermore, previous studies have demonstrated that inhibition

of GSK beta increased the production of anti-inflammatory cytokines while reducing the levels of

pro-inflammatory cytokines consistent with a model proposed by Martin et al (M. Martin et al.,

2005) whereby GSK-3b was found to promote pro inflammatory response (H. Wang et al., 2011;

Z. Wang et al., 2008). In line with these findings, TWEAK treatment elicited a marked increase

in the expression levels of tyrosine phosphorylated GSK B (Y2016 GSK-3B) while down

regulation of SYK activation via pharmacological inhibition or SYK siRNA resulted in profound

abrogation of pGSK 3B (Y2016) expression suggesting that SYK is a key regulator of GSK 3b

224

activation. Consistent with our finding SYK deficiency in dendritic cells was found to enhance

LPS-induced IFNb and IL-10 expression but suppressed pro-inflammatory cytokine levels, namely

TNF-a and IL-6 via PI3K-Akt and NF-kB signaling pathways suggesting a divergent influence of

SYK on pro-and anti-inflamamtory in TLR-mediated response (Hui Yin et al., 2016).

Complex interactions between cytoplasmic kinases including MAP kinase and generation

of proinflammatory cytokines via NF-kB dependent mechanisms have been suggested (Chiou et

al., 2011). In our previous study using LPS stimulated microglia we demonstrated that Fyn-PKCd

signaling axis is a critical regulator of MAP kinase activation in microglial cells (N. Panicker, H.

Saminathan, H. Jin, M. Neal, & D. S. Harischandra, 2015) suggesting that Fyn kinase is a major

upstream regulator of pro-inflammatory signaling events. The present results demonstrate a time

dependent increase in the activation of MAP kinase signaling events as demonstrated by increased

phosphorylations of p38, ERK1/2, and JNK following TWEAK stimulation. Moreover, R788

pretreatment in TWEAK stimulated microglial cells markedly down regulated MAPK activation.

Our results are consistent with a previous observation that pharmacological inhibition of SYK

attenuate MAPK, including P38MAPK and JNK signaling events (Clifford A. Lowell, 2011).

Contrary to our finding, a recent report demonstrated that Syk deficiency fails to alter the activation

of MAPK signaling in LPS stimulated dendritic cells (Hui Yin et al., 2016). Thus, it appears that

SYK may evoke cell type and context specific effects in regards to MAPK activation upon

inflammagen stimulation of myeloid cells. Further studies are warranted to study the impact of

MAPK inhibition on NLRP3 inflammasome activation in BV2 microglial cells subjected to

TWEAK stimulation.

Autophagic mechanisms involves the engulfment of damaged organelles and cellular into

double membrane vacuoles called autophagosomes. The autophagosomes ultimately fuse with the

225

lysosomes thereby facilitating recycling of cellular macromolecules. In fact, lysosomal

dysfunction is associated with the abnormal accumulation of autophagosomes in familial PD cases

(Dehay et al., 2010). Moreover, autophagy negatively regulates NLRP3 inflammasome activation

and pro-inflammatory cytokine production in immune cells (V. A. Rathinam et al., 2012; T. Saitoh

& S. Akira, 2016; Yuk et al., 2013; Z. Zhong et al., 2016). Conversely, genetic ablation of

autophagy related markers including LC3-II and BECN-1 was found to aggravate NLRP3

inflammasome activation in ATP stimulated macrophages (P. Gurung et al., 2015; Salminen et al.,

2012). In our study, we demonstrated increased expression of autophagy markers in Tweak treated

microglial cells. Given that mitochondrial dysfunction and ROS generation precedes NLRP3

inflammasome activation in TWEAK treated microglial cells our results raise the possibility that

excessive ROS generation via NOX2 activation alongside defective mitophagic mechanism owing

to impaired clearance of ROS generating damaged mitochondria might have partly contributed to

NLRP3 inflammasome activation. Indeed SYK has been shown to mediate ROS generation and

activation of MAPK which in turn activates autophagy via disassociation of BECN1 from Bcl2

(S. H. Choi et al., 2015; Hideki Hara et al., 2013; Kang et al., 2011). Our results underscore the

importance of SYK mediated autophagy upregulation in the induction of microglial pro

inflammatory response. Another NLRP3 inflammasome activating mechanism involves the

lysosomal membrane destabilization resulting in the cytosolic release of lysosomal hydrolases

including cathepsin B (R. Lu et al., 2012; Orlowski et al., 2015; Tseng et al., 2013; L. Wang et al.,

2016; K. Weber et al., 2014). Microbial toxins that trigger lysosomal membrane destabilization

leads to release of lysosomal cathepsin B into cytosol thereby causing NLRP3 inflammasome

activation (Tseng et al., 2013). Moreover, in LPS treated macrophages lysosomal Cathepsin B

release into cytosol results in NLRP3 inflammasome activation and IL-1 beta secretion (K. Weber

226

et al., 2014). Consistent with these reports, TWEAK-induced NLRP3 inflammasome activation

positively correlated with upregulation of cathespin B suggesting altered lysosomal function might

be partly responsible for NLRP3 inflammasome activation. Conversely, abrogation of cathepsin

B reduced caspase-1 mediated IL-1B release in response to MSU crystals (Kingsbury et al., 2011;

Välimäki et al., 2013). Intriguingly, Syk has been linked to autophagolysosomal system (ALS)

dysfunction upon microbial infection in macrophages (Krisenko, Higgins, et al., 2015).

Consistently, both R788 and Syk siRNA ameliorated TWEAK induced ALS impairment as

evidenced by reduced accumulation of LC3-II, Becn-1 and Cathepsin-B levels in BV2 microglial

cells. Together our studies suggest that SYK is a key upstream regulator of ALS function in

microglia in response to TWEAK. Further studies are warranted to determine whether ASC

phosphorylation is an important step in the mechanism of ALS dysfunction in TWEAK treated

cells.

Syk inhibitors are currently used for treatment of allergic rhinitis, autoimmune

thrombocytopenia,and B-cell malignancies (Mocsai et al., 2010). Fostamatinib sodium is a small

molecule of Syk inhibitor that has shown efficacy in animal models of Asthma, Lymphoma,

Glomerulonephritis and Diabetes (McAdoo, 2011). Based on the observation that R788 and siRNA

Syk known-down attenuated NLRP3 inflammasome activation, Caspase-1, CHOP, Cathepsin-B

and GSK-3 Beta activation in Tweak treated BV2 microglial cells we next sought to corroborate

our in vitro findings in an intra-striatal infusion mouse model of TWEAK that were pretreated with

the TWEAK inhibitor Piceatannol. In the in vivo TWEAK model although several of the

inflammation related markers displayed a trend toward an increase it failed to reach significance

as compared to vehicle treated mice. In a similar fashion Piceatannol failed to modulate TWEAK-

induced effects. Therefore, further studies are needed to confirm the inflammatory response

227

elicited following TWEAK infusion into the striatum, and the optimal dose of Piceatannol needed

to elicit an anti-inflammatory response.

Our present study emphasizes the pro-inflammatory role of SYK as assessed via the

activation of NLRP3 inflammasome in microglial cells in response to TWEAK in microglial cells.

Moreover, this is the first study demonstrating that inhibition of ROS generation, mitochondrial

dysfunction, ERS, and ALS dysfunction may partially account for the suppressive effect of SYK

inhibition on NLRP3 inflammasome activation. In conclusion, the study provides novel insights

into the anti-inflammatory role of R788 and in this context highlighted its ability to inhibit NLRP3

inflammasome via suppression of oxidative stress dependent mechanisms. Thus, selective

targeting of SYK may serve as a novel therapeutic intervention in the modulation of inflammatory

immune responses in several neuro-inflammation related disorders including PD.

Bibliography

Aouar, B., et al. (2016). Dual Role of the Tyrosine Kinase Syk in Regulation of Toll-Like

Receptor Signaling in Plasmacytoid Dendritic Cells. PLoS One, 11(6), e0156063.

doi:10.1371/journal.pone.0156063

Bartels, A. L., et al. (2010). [11C]-PK11195 PET: quantification of neuroinflammation and a

monitor of anti-inflammatory treatment in Parkinson's disease? Parkinsonism Relat

Disord, 16(1), 57-59. doi:10.1016/j.parkreldis.2009.05.005

Berton, G., et al. (2005). Src and Syk kinases: key regulators of phagocytic cell activation.

Trends Immunol, 26(4), 208-214. doi:10.1016/j.it.2005.02.002

Bronner, D. N., et al. (2015). Endoplasmic Reticulum stress activates the inflammasome via

NLRP3-caspase-2 driven mitochondrial damage. Immunity, 43(3), 451-462.

doi:10.1016/j.immuni.2015.08.008

Bronner, D. N., et al. (2015). Endoplasmic Reticulum Stress Activates the Inflammasome via

NLRP3- and Caspase-2-Driven Mitochondrial Damage. Immunity, 43(3), 451-462.

doi:10.1016/j.immuni.2015.08.008

228

Burkly, L. C., et al. (2007). TWEAKing tissue remodeling by a multifunctional cytokine: role of

TWEAK/Fn14 pathway in health and disease. Cytokine, 40(1), 1-16.

doi:10.1016/j.cyto.2007.09.007

Cassel, S. L., et al. (2008). The Nalp3 inflammasome is essential for the development of

silicosis. Proc Natl Acad Sci U S A, 105(26), 9035-9040. doi:10.1073/pnas.0803933105

Chen, L., et al. (2015). NLRP3 inflammasome activation by mitochondrial reactive oxygen

species plays a key role in long-term cognitive impairment induced by paraquat exposure.

Neurobiol Aging, 36(9), 2533-2543. doi:10.1016/j.neurobiolaging.2015.05.018

Chen, Y., et al. (2015). Endothelial Nlrp3 inflammasome activation associated with lysosomal

destabilization during coronary arteritis. Biochim Biophys Acta, 1853(2), 396-408.

doi:10.1016/j.bbamcr.2014.11.012

Chicheportiche, Y., et al. (1997). TWEAK, a new secreted ligand in the tumor necrosis factor

family that weakly induces apoptosis. J Biol Chem, 272(51), 32401-32410.

Chiou, W. F., et al. (2011). Psoralidin inhibits LPS-induced iNOS expression via repressing Syk-

mediated activation of PI3K-IKK-IkappaB signaling pathways. Eur J Pharmacol, 650(1),

102-109. doi:10.1016/j.ejphar.2010.10.004

Choi, S. H., et al. (2015). SYK regulates macrophage MHC-II expression via activation of

autophagy in response to oxidized LDL. Autophagy, 11(5), 785-795.

doi:10.1080/15548627.2015.1037061

Choi, S. H., et al. (2009). Lipoprotein accumulation in macrophages via toll-like receptor-4-

dependent fluid phase uptake. Circ Res, 104(12), 1355-1363.

doi:10.1161/circresaha.108.192880

Codolo, G., et al. (2013). Triggering of inflammasome by aggregated alpha-synuclein, an

inflammatory response in synucleinopathies. PLoS One, 8(1), e55375.

doi:10.1371/journal.pone.0055375

Combs, C. K., et al. (1999). Identification of microglial signal transduction pathways mediating a

neurotoxic response to amyloidogenic fragments of beta-amyloid and prion proteins. J

Neurosci, 19(3), 928-939.

229

Cortes-Vieyra, R., et al. (2012). Role of glycogen synthase kinase-3 beta in the inflammatory

response caused by bacterial pathogens. J Inflamm (Lond), 9(1), 23. doi:10.1186/1476-

9255-9-23

Cruz, C. M., et al. (2007). ATP activates a reactive oxygen species-dependent oxidative stress

response and secretion of proinflammatory cytokines in macrophages. J Biol Chem,

282(5), 2871-2879. doi:10.1074/jbc.M608083200

Dai, J. N., et al. (2011). Gastrodin inhibits expression of inducible NO synthase,

cyclooxygenase-2 and proinflammatory cytokines in cultured LPS-stimulated microglia

via MAPK pathways. PLoS One, 6(7), e21891. doi:10.1371/journal.pone.0021891

Dehay, B., et al. (2010). Pathogenic lysosomal depletion in Parkinson's disease. J Neurosci,

30(37), 12535-12544. doi:10.1523/jneurosci.1920-10.2010

Dostert, C., et al. (2008). Innate immune activation through Nalp3 inflammasome sensing of

asbestos and silica. Science, 320(5876), 674-677. doi:10.1126/science.1156995

Feng, L., et al. (2015). P2X7R blockade prevents NLRP3 inflammasome activation and brain

injury in a rat model of intracerebral hemorrhage: involvement of peroxynitrite. J

Neuroinflammation, 12, 190. doi:10.1186/s12974-015-0409-2

Ferrari, C. C., et al. (2006). Progressive neurodegeneration and motor disabilities induced by

chronic expression of IL-1beta in the substantia nigra. Neurobiol Dis, 24(1), 183-193.

doi:10.1016/j.nbd.2006.06.013

Frazier, W. J., et al. (2009). Increased inflammation, impaired bacterial clearance, and metabolic

disruption after gram-negative sepsis in Mkp-1-deficient mice. J Immunol, 183(11),

7411-7419. doi:10.4049/jimmunol.0804343

Futterer, K., et al. (1998). Structural basis for Syk tyrosine kinase ubiquity in signal transduction

pathways revealed by the crystal structure of its regulatory SH2 domains bound to a

dually phosphorylated ITAM peptide. J Mol Biol, 281(3), 523-537.

doi:10.1006/jmbi.1998.1964

Ghosh, S., et al. (2015). Stress Granules Modulate SYK to Cause Microglial Cell Dysfunction in

Alzheimer's Disease. EBioMedicine, 2(11), 1785-1798. doi:10.1016/j.ebiom.2015.09.053

230

Gordon, R., et al. (2016). Protein kinase Cdelta upregulation in microglia drives

neuroinflammatory responses and dopaminergic neurodegeneration in experimental

models of Parkinson's disease. Neurobiol Dis, 93, 96-114. doi:10.1016/j.nbd.2016.04.008

Gross, O., et al. (2009). Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal

host defence. Nature, 459(7245), 433-436. doi:10.1038/nature07965

Guo, H., et al. (2015). Inflammasomes: mechanism of action, role in disease, and therapeutics.

Nat Med, 21(7), 677-687. doi:10.1038/nm.3893

Gurung, P., et al. (2016). Autoinflammatory Skin Disorders: The Inflammasomme in Focus.

Trends Mol Med, 22(7), 545-564. doi:10.1016/j.molmed.2016.05.003

Gurung, P., et al. (2015). Mitochondria: diversity in the regulation of the NLRP3 inflammasome.

Trends Mol Med, 21(3), 193-201. doi:10.1016/j.molmed.2014.11.008

Haile, W. B., et al. (2010). Tumor necrosis factor-like weak inducer of apoptosis and fibroblast

growth factor-inducible 14 mediate cerebral ischemia-induced poly(ADP-ribose)

polymerase-1 activation and neuronal death. Neuroscience, 171(4), 1256-1264.

doi:10.1016/j.neuroscience.2010.10.029

Haile, W. B., et al. (2010). The interaction between tumor necrosis factor-like weak inducer of

apoptosis and its receptor fibroblast growth factor-inducible 14 promotes the recruitment

of neutrophils into the ischemic brain. J Cereb Blood Flow Metab, 30(6), 1147-1156.

doi:10.1038/jcbfm.2009.280

Halle, A., et al. (2008). The NALP3 inflammasome is involved in the innate immune response to

amyloid-beta. Nat Immunol, 9(8), 857-865. doi:10.1038/ni.1636

Hara, H., et al. (2013). Phosphorylation of the adaptor ASC acts as a molecular switch that

controls the formation of speck-like aggregates and inflammasome activity. 14(12), 1247-

1255. doi:10.1038/ni.2749

He, J., et al. (2005). Lysosome is a primary organelle in B cell receptor-mediated apoptosis: an

indispensable role of Syk in lysosomal function. Genes Cells, 10(1), 23-35.

doi:10.1111/j.1365-2443.2004.00811.x

231

He, Y., et al. (2015a). Mincle/Syk Signaling Pathway Contributes to Neuroinflammation after

Subarachnoid Hemorrhage in Rats. Stroke, 46(8), 2277-2286.

doi:10.1161/strokeaha.115.010088

He, Y., et al. (2015b). Macrophage-Inducible C-Type Lectin/Spleen Tyrosine Kinase Signaling

Pathway Contributes to Neuroinflammation After Subarachnoid Hemorrhage in Rats.

Stroke, 46(8), 2277-2286. doi:10.1161/strokeaha.115.010088

Hevia, H., et al. (2004). 5'-methylthioadenosine modulates the inflammatory response to

endotoxin in mice and in rat hepatocytes. Hepatology, 39(4), 1088-1098.

doi:10.1002/hep.20154

Hoegen, T., et al. (2011). The NLRP3 inflammasome contributes to brain injury in

pneumococcal meningitis and is activated through ATP-dependent lysosomal cathepsin B

release. J Immunol, 187(10), 5440-5451. doi:10.4049/jimmunol.1100790

Hong, S., et al. (2017). Prolonged exposure of lipopolysaccharide induces NLRP3-independent

maturation and secretion of interleukin (IL)-1beta in macrophages. J Microbiol

Biotechnol. doi:10.4014/jmb.1709.09017

Hornung, V., et al. (2009). AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating

inflammasome with ASC. Nature, 458(7237), 514-518. doi:10.1038/nature07725

Hou, L., et al. (2017). NADPH oxidase-derived H(2)O(2) mediates the regulatory effects of

microglia on astrogliosis in experimental models of Parkinson's disease. Redox Biology,

12, 162-170. doi:10.1016/j.redox.2017.02.016

Iyer, S. S., et al. (2013). Mitochondrial cardiolipin is required for Nlrp3 inflammasome

activation. Immunity, 39(2), 311-323. doi:10.1016/j.immuni.2013.08.001

Jain, A., et al. Endothelin-1 Induces Endoplasmic Reticulum Stress by Activating the PLC-

IP<sub>3</sub> Pathway. The American Journal of Pathology, 180(6), 2309-2320.

doi:10.1016/j.ajpath.2012.03.005

Jakus, Z., et al. (2010). Genetic deficiency of Syk protects mice from autoantibody-induced

arthritis. Arthritis Rheum, 62(7), 1899-1910. doi:10.1002/art.27438

Jo, E. K., et al. (2016). Molecular mechanisms regulating NLRP3 inflammasome activation. Cell

Mol Immunol, 13(2), 148-159. doi:10.1038/cmi.2015.95

232

Johnson, G. L., et al. (2002). Mitogen-activated protein kinase pathways mediated by ERK, JNK,

and p38 protein kinases. Science, 298(5600), 1911-1912. doi:10.1126/science.1072682

Jope, R. S., et al. (2007). Glycogen synthase kinase-3 (GSK3): inflammation, diseases, and

therapeutics. Neurochem Res, 32(4-5), 577-595. doi:10.1007/s11064-006-9128-5

Kang, R., et al. (2011). The Beclin 1 network regulates autophagy and apoptosis. Cell Death

Differ, 18(4), 571-580. doi:10.1038/cdd.2010.191

Kanneganti, T. D., et al. (2006). Critical role for Cryopyrin/Nalp3 in activation of caspase-1 in

response to viral infection and double-stranded RNA. J Biol Chem, 281(48), 36560-

36568. doi:10.1074/jbc.M607594200

Kaur, K., et al. (2017). Neuroinflammation - A major cause for striatal dopaminergic

degeneration in Parkinson's disease. J Neurol Sci, 381, 308-314.

doi:10.1016/j.jns.2017.08.3251

Kim, J. Y., et al. (2006). Involvement of MAPK activation in chemokine or COX-2 productions

by Toxoplasma gondii. Korean J Parasitol, 44(3), 197-207.

Kingsbury, S. R., et al. (2011). The role of the NLRP3 inflammasome in gout. Journal of

Inflammation Research, 4, 39-49. doi:10.2147/JIR.S11330

Klionsky, D. J., et al. (2016). Guidelines for the use and interpretation of assays for monitoring

autophagy (3rd edition). Autophagy, 12(1), 1-222. doi:10.1080/15548627.2015.1100356

Krisenko, M. O., et al. (2015). Syk Is Recruited to Stress Granules and Promotes Their Clearance

through Autophagy. J Biol Chem, 290(46), 27803-27815. doi:10.1074/jbc.M115.642900

Kwon, D. H., et al. (2017). Inhibitory effects on the production of inflammatory mediators and

reactive oxygen species by Mori folium in lipopolysaccharide-stimulated macrophages

and zebrafish. An Acad Bras Cienc, 89(1 Suppl 0), 661-674. doi:10.1590/0001-

3765201720160836

Lamkanfi, M., et al. (2012). Inflammasomes and their roles in health and disease. Annu Rev Cell

Dev Biol, 28, 137-161. doi:10.1146/annurev-cellbio-101011-155745

233

Laudisi, F., et al. (2014). Tyrosine kinases: the molecular switch for inflammasome activation.

Cell Mol Immunol, 11(2), 129-131. doi:10.1038/cmi.2014.4

Lawana, V., et al. (2017). Involvement of c-Abl Kinase in Microglial Activation of NLRP3

Inflammasome and Impairment in Autolysosomal System. J Neuroimmune Pharmacol,

12(4), 624-660. doi:10.1007/s11481-017-9746-5

Lebeaupin, C., et al. (2015). ER stress induces NLRP3 inflammasome activation and hepatocyte

death. Cell Death Dis, 6, e1879. doi:10.1038/cddis.2015.248

Lee, H. M., et al. (2012). Mycobacterium abscessus activates the NLRP3 inflammasome via

Dectin-1-Syk and p62/SQSTM1. Immunol Cell Biol, 90(6), 601-610.

doi:10.1038/icb.2011.72

Lerner, A. G., et al. (2012). IRE1alpha induces thioredoxin-interacting protein to activate the

NLRP3 inflammasome and promote programmed cell death under irremediable ER

stress. Cell Metab, 16(2), 250-264. doi:10.1016/j.cmet.2012.07.007

Lin, Y. C., et al. (2015). Syk is involved in NLRP3 inflammasome-mediated caspase-1 activation

through adaptor ASC phosphorylation and enhanced oligomerization. J Leukoc Biol,

97(5), 825-835. doi:10.1189/jlb.3HI0814-371RR

Llop-Guevara, A., et al. (2015). Simultaneous inhibition of JAK and SYK kinases ameliorates

chronic and destructive arthritis in mice. Arthritis Res Ther, 17, 356. doi:10.1186/s13075-

015-0866-0

Lowell, C. A. (2011). Src-family and Syk Kinases in Activating and Inhibitory Pathways in

Innate Immune Cells – Signaling Crosstalk. Cold Spring Harbor perspectives in biology,

3(3), 10.1101/cshperspect.a002352 a002352. doi:10.1101/cshperspect.a002352

Lu, R., et al. (2012). CEACAM1 negatively regulates IL-1beta production in LPS activated

neutrophils by recruiting SHP-1 to a SYK-TLR4-CEACAM1 complex. PLoS Pathog,

8(4), e1002597. doi:10.1371/journal.ppat.1002597

Lu, Y. Q., et al. (2017). NLRP3 inflammasome activation results in liver inflammation and

fibrosis in mice infected with Schistosoma japonicum in a Syk-dependent manner. Sci

Rep, 7(1), 8120. doi:10.1038/s41598-017-08689-1

234

Ma, M. W., et al. (2017). NADPH Oxidase 2 Regulates NLRP3 Inflammasome Activation in the

Brain after Traumatic Brain Injury. 2017, 6057609. doi:10.1155/2017/6057609

Machado, V., et al. (2016). Microglia-Mediated Neuroinflammation and Neurotrophic Factor-

Induced Protection in the MPTP Mouse Model of Parkinson's Disease-Lessons from

Transgenic Mice. Int J Mol Sci, 17(2). doi:10.3390/ijms17020151

Mao, Z., et al. (2017). The NLRP3 Inflammasome is Involved in the Pathogenesis of Parkinson's

Disease in Rats. Neurochem Res, 42(4), 1104-1115. doi:10.1007/s11064-017-2185-0

Mariathasan, S., et al. (2006). Cryopyrin activates the inflammasome in response to toxins and

ATP. Nature, 440(7081), 228-232. doi:10.1038/nature04515

Martin, M., et al. (2005). Toll-like receptor-mediated cytokine production is differentially

regulated by glycogen synthase kinase 3. Nat Immunol, 6(8), 777-784.

doi:10.1038/ni1221

Martinon, F. (2012). The endoplasmic reticulum: a sensor of cellular stress that modulates

immune responses. Microbes Infect, 14(14), 1293-1300.

doi:10.1016/j.micinf.2012.07.005

Martinon, F., et al. (2006). Gout-associated uric acid crystals activate the NALP3 inflammasome.

Nature, 440(7081), 237-241. doi:10.1038/nature04516

McAdoo, S. P. (2011). Fostamatinib Disodium. 36(4), 273-.

McDonald, D. R., et al. (1997). Amyloid fibrils activate tyrosine kinase-dependent signaling and

superoxide production in microglia. J Neurosci, 17(7), 2284-2294.

McGeer, P. L., et al. (1988). Reactive microglia are positive for HLA-DR in the substantia nigra

of Parkinson's and Alzheimer's disease brains. Neurology, 38(8), 1285-1291.

Menu, P., et al. (2012). ER stress activates the NLRP3 inflammasome via an UPR-independent

pathway. Cell Death Dis, 3, e261. doi:10.1038/cddis.2011.132

Miller, Y. I., et al. (2012). The SYK side of TLR4: signalling mechanisms in response to LPS

and minimally oxidized LDL. Br J Pharmacol, 167(5), 990-999. doi:10.1111/j.1476-

5381.2012.02097.x

235

Mocsai, A., et al. (2010). The SYK tyrosine kinase: a crucial player in diverse biological

functions. Nat Rev Immunol, 10(6), 387-402. doi:10.1038/nri2765

Moustafa, A. A., et al. (2016). Motor symptoms in Parkinson's disease: A unified framework.

Neurosci Biobehav Rev, 68, 727-740. doi:10.1016/j.neubiorev.2016.07.010

Mustafa, S., et al. (2016). The role of TWEAK/Fn14 signaling in the MPTP-model of

Parkinson's disease. Neuroscience, 319, 116-122.

doi:10.1016/j.neuroscience.2016.01.034

Nagatsu, T., et al. (2007). Biochemistry of postmortem brains in Parkinson's disease: historical

overview and future prospects. J Neural Transm Suppl(72), 113-120.

Orlowski, G. M., et al. (2015). Multiple Cathepsins Promote Pro-IL-1beta Synthesis and NLRP3-

Mediated IL-1beta Activation. J Immunol, 195(4), 1685-1697.

doi:10.4049/jimmunol.1500509

Panicker, N., et al. (2015). Fyn Kinase Regulates Microglial Neuroinflammatory Responses in

Cell Culture and Animal Models of Parkinson's Disease. 35(27), 10058-10077.

doi:10.1523/jneurosci.0302-15.2015

Paris, D., et al. (2014). The spleen tyrosine kinase (Syk) regulates Alzheimer amyloid-beta

production and Tau hyperphosphorylation. J Biol Chem, 289(49), 33927-33944.

doi:10.1074/jbc.M114.608091

Patterson, H., et al. (2014). Protein kinase inhibitors in the treatment of inflammatory and

autoimmune diseases. Clin Exp Immunol, 176(1), 1-10. doi:10.1111/cei.12248

Poewe, W. (2008). Non-motor symptoms in Parkinson's disease. Eur J Neurol, 15 Suppl 1, 14-

20. doi:10.1111/j.1468-1331.2008.02056.x

Polavarapu, R., et al. (2005). Tumor necrosis factor-like weak inducer of apoptosis increases the

permeability of the neurovascular unit through nuclear factor-kappa B pathway

activation. J Neurosci, 25(44), 10094-10100. doi:10.1523/jneurosci.3382-05.2005

Qian, L., et al. (2008). Microglial cells and Parkinson's disease. Immunol Res, 41(3), 155-164.

doi:10.1007/s12026-008-8018-0

236

Qin, L., et al. (2004). NADPH oxidase mediates lipopolysaccharide-induced neurotoxicity and

proinflammatory gene expression in activated microglia. J Biol Chem, 279(2), 1415-

1421. doi:10.1074/jbc.M307657200

Rathinam, V. A., et al. (2012). Regulation of inflammasome signaling. Nat Immunol, 13(4), 333-

342. doi:10.1038/ni.2237

Rehani, K., et al. (2009). Toll-like receptor-mediated production of IL-1Ra is negatively

regulated by GSK3 via the MAPK ERK1/2. J Immunol, 182(1), 547-553.

Resende, R., et al. (2008). ER stress is involved in Abeta-induced GSK-3beta activation and tau

phosphorylation. J Neurosci Res, 86(9), 2091-2099. doi:10.1002/jnr.21648

Rogers, N. C., et al. (2005). Syk-dependent cytokine induction by Dectin-1 reveals a novel

pattern recognition pathway for C type lectins. Immunity, 22(4), 507-517.

doi:10.1016/j.immuni.2005.03.004

Saito, M. K. (2011). [Inflammasomes and related diseases]. Nihon Rinsho Meneki Gakkai Kaishi,

34(1), 20-28.

Saitoh, T., et al. (2016). Regulation of inflammasomes by autophagy. J Allergy Clin Immunol,

138(1), 28-36. doi:10.1016/j.jaci.2016.05.009

Saitoh, T., et al. (2016). Regulation of inflammasomes by autophagy. Journal of Allergy and

Clinical Immunology, 138(1), 28-36. doi:https://doi.org/10.1016/j.jaci.2016.05.009

Salminen, A., et al. (2012). Inflammaging: disturbed interplay between autophagy and

inflammasomes. Aging (Albany NY), 4(3), 166-175. doi:10.18632/aging.100444

Sarkar, S., et al. (2017). Mitochondrial impairment in microglia amplifies NLRP3 inflammasome

proinflammatory signaling in cell culture and animal models of Parkinson's disease. 3,

30. doi:10.1038/s41531-017-0032-2

Schlatterer, S. D., et al. (2011). Neuronal c-Abl overexpression leads to neuronal loss and

neuroinflammation in the mouse forebrain. J Alzheimers Dis, 25(1), 119-133.

doi:10.3233/jad-2011-102025

237

Schroder, K., et al. (2010). The Inflammasomes. Cell, 140(6), 821-832.

doi:https://doi.org/10.1016/j.cell.2010.01.040

Shaw, P. J., et al. (2011). Inflammasomes and autoimmunity. Trends Mol Med, 17(2), 57-64.

doi:10.1016/j.molmed.2010.11.001

Shio, M. T., et al. (2009). Malarial hemozoin activates the NLRP3 inflammasome through Lyn

and Syk kinases. PLoS Pathog, 5(8), e1000559. doi:10.1371/journal.ppat.1000559

Subramaniam, S. R., et al. (2017). Targeting Microglial Activation States as a Therapeutic

Avenue in Parkinson's Disease. Front Aging Neurosci, 9, 176.

doi:10.3389/fnagi.2017.00176

Suzuki, Y., et al. (2013). Involvement of Mincle and Syk in the changes to innate immunity after

ischemic stroke. Scientific Reports, 3, 3177. doi:10.1038/srep03177

https://www.nature.com/articles/srep03177#supplementary-information

Suzuki, Y., et al. (2013). Involvement of Mincle and Syk in the changes to innate immunity after

ischemic stroke. Sci Rep, 3, 3177. doi:10.1038/srep03177

Tam, J. M., et al. (2014). Dectin-1–Dependent LC3 Recruitment to Phagosomes Enhances

Fungicidal Activity in Macrophages. The Journal of Infectious Diseases, 210(11), 1844-

1854. doi:10.1093/infdis/jiu290

Taniguchi, T., et al. (1991). Molecular cloning of a porcine gene syk that encodes a 72-kDa

protein-tyrosine kinase showing high susceptibility to proteolysis. J Biol Chem, 266(24),

15790-15796.

Teismann, P., et al. (2003). Cyclooxygenase-2 is instrumental in Parkinson's disease

neurodegeneration. Proc Natl Acad Sci U S A, 100(9), 5473-5478.

doi:10.1073/pnas.0837397100

Tohyama, Y., et al. (2009). Protein tyrosine kinase, syk: a key player in phagocytic cells. J

Biochem, 145(3), 267-273. doi:10.1093/jb/mvp001

Tseng, W. A., et al. (2013). NLRP3 inflammasome activation in retinal pigment epithelial cells

by lysosomal destabilization: implications for age-related macular degeneration. Invest

Ophthalmol Vis Sci, 54(1), 110-120. doi:10.1167/iovs.12-10655

238

Turner, M., et al. (2000). Tyrosine kinase SYK: essential functions for immunoreceptor

signalling. Immunol Today, 21(3), 148-154.

Ulanova, M., et al. (2007). Involvement of Syk protein tyrosine kinase in LPS-induced responses

in macrophages. J Endotoxin Res, 13(2), 117-125. doi:10.1177/0968051907079125

Välimäki, E., et al. (2013). Monosodium Urate Activates Src/Pyk2/PI3 Kinase and Cathepsin

Dependent Unconventional Protein Secretion From Human Primary Macrophages.

Molecular & Cellular Proteomics : MCP, 12(3), 749-763.

doi:10.1074/mcp.M112.024661

Wang, H., et al. (2011). The role of glycogen synthase kinase 3 in regulating IFN-beta-mediated

IL-10 production. J Immunol, 186(2), 675-684. doi:10.4049/jimmunol.1001473

Wang, J., et al. (2015). The NLRP3 inflammasome is dispensable for ER stress-induced

pancreatic β-cell damage in Akita mice. Biochem Biophys Res Commun, 466(3), 300-305.

doi:https://doi.org/10.1016/j.bbrc.2015.09.009

Wang, L., et al. (2016). Enhancement of endothelial permeability by free fatty acid through

lysosomal cathepsin B-mediated Nlrp3 inflammasome activation. Oncotarget, 7(45),

73229-73241. doi:10.18632/oncotarget.12302

Wang, Q., et al. (2015). Neuroinflammation in Parkinson's disease and its potential as

therapeutic target. Transl Neurodegener, 4, 19. doi:10.1186/s40035-015-0042-0

Wang, Z., et al. (2008). Glycogen synthase kinase 3 in MLL leukaemia maintenance and targeted

therapy. Nature, 455(7217), 1205-1209. doi:10.1038/nature07284

Weber, K., et al. (2014). Lysosomes integrate metabolic-inflammatory cross-talk in primary

macrophage inflammasome activation. J Biol Chem, 289(13), 9158-9171.

doi:10.1074/jbc.M113.531202

Wiley, S. R., et al. (2001). A novel TNF receptor family member binds TWEAK and is

implicated in angiogenesis. Immunity, 15(5), 837-846.

Winkles, J. A. (2008). The TWEAK-Fn14 cytokine-receptor axis: discovery, biology and

therapeutic targeting. Nat Rev Drug Discov, 7(5), 411-425. doi:10.1038/nrd2488

239

Yanagi, S., et al. (2001). Syk expression and novel function in a wide variety of tissues. Biochem

Biophys Res Commun, 288(3), 495-498. doi:10.1006/bbrc.2001.5788

Yanagi, S., et al. (1995). The structure and function of nonreceptor tyrosine kinase p72syk

expressed in hematopoietic cells. Cell Signal, 7(3), 185-193.

Yepes, M. (2007). TWEAK and the central nervous system. Mol Neurobiol, 35(3), 255-265.

Yi, Y. S., et al. (2014a). Functional roles of Syk in macrophage-mediated inflammatory

responses. Mediators Inflamm, 2014, 270302. doi:10.1155/2014/270302

Yi, Y. S., et al. (2014b). Functional Roles of Syk in Macrophage-Mediated Inflammatory

Responses. Mediators Inflamm, 2014. doi:10.1155/2014/270302

Yin, H., et al. (2016). Syk Negatively Regulates TLR4-mediated IFNβ and IL-10 production and

Promotes Inflammatory Responses in Dendritic Cells. Biochim Biophys Acta, 1860(3),

588-598. doi:10.1016/j.bbagen.2015.12.012

Yin, H., et al. (2016). Syk negatively regulates TLR4-mediated IFNbeta and IL-10 production

and promotes inflammatory responses in dendritic cells. Biochim Biophys Acta, 1860(3),

588-598. doi:10.1016/j.bbagen.2015.12.012

Yu, J., et al. (2014). Inflammasome activation leads to Caspase-1-dependent mitochondrial

damage and block of mitophagy. Proc Natl Acad Sci U S A, 111(43), 15514-15519.

doi:10.1073/pnas.1414859111

Yuk, J. M., et al. (2013). Crosstalk between Autophagy and Inflammasomes. Mol Cells, 36(5),

393-399. doi:10.1007/s10059-013-0298-0

Zhong, L. M., et al. (2012). Resveratrol inhibits inflammatory responses via the mammalian

target of rapamycin signaling pathway in cultured LPS-stimulated microglial cells. PLoS

One, 7(2), e32195. doi:10.1371/journal.pone.0032195

Zhong, Z., et al. (2016). NF-kappaB Restricts Inflammasome Activation via Elimination of

Damaged Mitochondria. Cell, 164(5), 896-910. doi:10.1016/j.cell.2015.12.057

Zhou, R., et al. (2011). A role for mitochondria in NLRP3 inflammasome activation. Nature,

469(7329), 221-225. doi:10.1038/nature09663

240

Figures

241

Fig 1. Increased Spleen Tyrosine Kinase and stress granules expression in Tweak treated BV2

microglia. A-B. Increased Syk expression in Tweak and LPS treated BV2 microglia. BV2 microglial cells

was treated with LPS 1ug/ml and Tweak 100 ng/ml for 3h, 6h, 12h, 24h. The control group was treated with

vehicle (1x PBS) used to prepare LPS and Tweak prior to treatment. Post treatment, 40 ug of protein that

was loaded was separated with 10% SDS-PAGE and probed with antibodies specific for Spleen Tyrosine

Kinase. To confirm equal protein in each lane, membranes were re-probed with Actin. The experiment

was repeated 2 times and a representative western blot was shown. Significant upregulation of spleen

tyrosine kinase expression was demonstrated in BV2 microglial cells upon stimulation with Tweak and LPS

using densitometric analysis after normalizing with -actin. Asterisk (**p < 0.01 or ***p < 0.001) represents

significant difference between Tweak / LPS treated groups and control group as determined by Anova

followed by Student Newman Keul’s post test. (C) Increased SYK Kinase activity in Tweak treated primary

microglial cells. The kinase assay was determined using a standard immunoprecipitation kinase assay.

Primary microglia were treated with Tweak 100 ng/ml for 12h and 24h with and without R788 Syk inhibitor

pretreatment. The SYK protein was immuno-precipitated from 500 ug of total protein and processed for in

vitro kinase assay with 5 ug biotin labelled SYK kinase substrate and ATP. Tyrosine phosphorylation of

SYK substrate was identified by monoclonal anti-phosphotyrosine antibody. The kinase activity is

quantified by densitometric analysis of phosphorylated SYK kinase substrate and expressed as percent

control. Recombinant SYK enzyme was used as a positive control in this experiment. SYK kinase assay is

repeated two times with N=6 and a representative graph is shown. The increase in SYK protein is

accompanied by increase in SYK kinase activity at 24h which was partially attentuated by pretreatment with

R788 (SYK inhibitor). Asterisk (**p < 0.01) represents significant differences between Tweak / R788

treated groups and control groups as determined by Anova followed by Student Newman Keul’s post test.

D) Increased stress granules expression in Tweak treated BV2 microglial cells. Immortalized BV2

microglial cells was treated with Tweak 100 ng/ml for 3h, 6h, 12h, 24h and then G3BP1 stress granules

levels were determined with western blotting. Actin was used as loading control for equal protein loading

in all the lanes. Representative western blots were shown and experiments were repeated two times.

Densitometric scanning analysis revealed increased G3BP1 expression in Tweak treated microglial cells at

12h and 24h. Asterisk (***p < 0.001) indicates significant differences between Tweak treated groups and

control group as calculated by Anova followed by Student Newman Keul’s post test.

242

243

Fig.2 Increased NLRP3 related markers and pro-inflammatory cytokine secretion in Tweak and

LPS treated BV2 microglial cells. A-D. BV2 microglial cells were primed with LPS (1ug/ml) for 3h

followed by stimulation with Tweak (100 ng/ml) for various time points (6h,12h & 24h). Post incubation,

lysates were separated with 12% SDS-PAGE and immunoblotted with antibodies specific for NLRP3

(A), Caspase-1 (B), IL-1 beta (C) and IL-18 (D). Membranes are re-probed with actin to ensure equal

protein loading in all the lanes. Densitometric scanning analysis revealed increased time dependent

increase in NLRP3 related markers in LPS and Tweak treated BV2 microglial cells. The experiment is

repeated two times and representative western blot is shown. Each data set is expressed as percentage of

control group and represents Mean ± S.E.M. Statistical differences were assessed with ANOVA followed

by Student Newman Keul’s post test. Asterisks (**p < 0.01 or ***p < 0.001) indicates significant

differences between Tweak treated groups and control groups. E–G Increased NLRP3 inflammasome

markers in Tweak stimulated BV2 microglia. BV2 microglia was treated with Tweak 100 ng/ml for 3h,

6h, 12h and 24h and lysates were probed with antibodies specific for NLRP3, Caspase-1 and IL-1 beta.

Band intensities of NLRP3, caspase-1 and IL-1 beta were quantified by densitometric analysis and

normalized to actin. Immunoblot analysis revealed that upregulation of NLRP3 (E), Caspase-1 (F) and

IL-1 beta (G) in Tweak treated BV2 microglial cells.The experiment was repeated two times and a

representative blot is shown. Asterisk (*p < 0.05) represents significant differences between Tweak

treated groups and control groups as assessed by ANOVA followed by Student Newman Keul’s post test.

H. Increased pro-inflammatory cytokine secretion in Tweak treated BV2 microglia. BV2 microglia were

treated with Tweak 100 ng for 8h and 24h. Cytokine levels in the supernatant were quantified at various

time points (8h and 24h) with the help of Luminex immunoassay system. Bar graph represents secreted

IL-1β and TNF-α in supernatant of Tweak treated microglial cells at various time points as determined

by ELISA immunoassay. The experiment is repeated two times and is represented as mean ± S.E.M with

N=6 in each treatment group. Statistical differences were assessed with ANOVA followed by Student

Newman Keul’s post test. Asterisks (**p < 0.01 or ***p < 0.001) indicates significant differences

between Tweak treated groups and control groups. I Increased pro-inflammatory cytokine mRNA

expression in Tweak treated BV2 microglia. SYBR Green real time quantitative PCR analysis of IL-1

beta, IL-18 and TNF mRNA in BV2 microglia treated with Tweak 100 ng/ml for 3h and 6h. After

treatment, RNA was extracted and RT-PCR was performed with 18s used as internal standard for

normalization. The normalized fold of increase in pro-inflammatory cytokine gene expression over the

control group was calculated by the ΔΔCt method. Each experiment is performed in triplicates at least

3 times independently. Statistical differences were assessed with ANOVA followed by Student Newman

Keul’s post test. Asterisks (**p < 0.01 or ***p < 0.001) indicates significant differences between Tweak

treated groups and control groups.

244

245

Fig 3. Increased NLRP3 inflammasome expression and Kinase expression in Tweak treated

BV2 microglial cells. A–C. Primary microglia was treated with Tweak 100 ng/ml for 3h, 6h,12h

and 24h. Lysates were probed for NLRP3, Caspase-1, IL-1 beta and -actin by western blotting.

Band intensities were quantified by densitometric analysis revealed upregulation of NLRP3 (A),

Caspase-1 (B), IL-1 beta (C) in Tweak treated primary microglial cells. Experiments were repeated

two times and a representative western blot is shown. Each data set is expressed as percentage of

control group. Asterisk (*p < 0.05 or **p < 0.01) represent significant differences between Tweak

treated groups and control groups as determined by ANOVA followed by Student Newman Keul’s

post test. D-E. Upregulation of SYK and PKC-delta kinase expression in Tweak treated primary

microglial cells. Following treatment of primary microglia with Tweak 100 ng/ml for 3h, 6h,12h

and 24h, redox sensitive kinases expression was evaluated when lysates were separated with SDS-

PAGE and subjected to western blot analysis with antibodies specific for SYK and PKC-delta.

Equal amount of protein was loaded in all the lanes with the help of -actin. Densitometric analysis

revealed upregulation of SYK (D) and PKC-delta (E) in Tweak treated primary microglial cells.

Experiment is repeated two times with representative blot shown. Asterisk (*p < 0.05 or **p < 0.01)

indicates significant differences between Tweak treated groups and control group as determined by

ANOVA followed by Student Newman Keul’s post test.

4A

4B

246

Fig.4 Tweak induced dissipation of Mitochondrial Membrane potential (MMP), ROS and

NOS production in BV2 microglial cells. A. Increased ROS production in Tweak treated BV2

microglia. Reactive oxygen species production was quantified in BV2 microglia treated with LPS

1ug/ml 6h, Tweak 50 ng, 250 ng and 500 ng for 3h and 6h. Total iROS generation was measured

by quantifying the redox sensitive CM-H2DCFDA spectrophotometrically using microplate

reader. Significant upregulation of ROS production was demonstrated in Tweak treated BV2

microglia that peaks at 3h and 6h. Experiment is repeated three times with N=6 in each treatment

group. Asterisk (p***<0.001) indicates significant differences between Tweak treated groups and

control group as determined by ANOVA followed by Student Newman Keul’s test. B. JC-1

fluorometry revealed significant time dependent dissipation of mitochondrial membrane potential

(MMP) in Tweak treated BV2 cells. BV2 microglial cells primed with LPS 1ug/ml for 3h were

treated with Tweak 100 ng for 24h prior to incubation with JC-1 dye for 30 min. Changes in

membrane potential (ΔΨm) were determined spectrophotometrically using Gemini Fluorescence

microplate reader. Reduction in red: green absorbance ratio of JC-1 dye indicate the loss of

mitochondrial membrane potential (Δψm). Significant time dependent dissipation of mitochondrial

membrane potential (MMP) was noticed in BV2 microglial cells upon Tweak and LPS treatment.

The experiment was repeated for three times with N=8 and a representative graph is shown.

Asterisk (p***<0.001) represents significant differences between Tweak / LPS treated groups and

control group as determined by ANOVA followed by Student Newman Keul’s test. C. SiRNA

mediated SYK knockdown suppressed in Tweak induced NOX2 production in BV2 microglia.

BV2 microglial cells were transfected with scrambled or SYK specific SiRNA for 72h and then

treated with or without Tweak (100 ng/ml) treatment for 24h. Whole cell lysates were probed with

NOX2 antibody using western blot with equal loading ascertained by -actin. Densitometric

analysis revealed that Syk knockdown downregulated NOX2 production Tweak stimulated BV2

microglial cells. Experiments were repeated four times and a representative sample was shown.

Asterisk (P***<0.001) represents significant differences between Tweak treated groups and

Tweak treated Syk SiRNA transfected as determined by ANOVA followed by Student Newman

Keul’s test.

4C

247

Fig.5 Increased ER stress marker expression in Tweak treated BV2microglia. A-E. Western

blot analysis of cell lysates from BV2 microglial cells treated with Tweak (100 ng/ml) for 3h, 6h,

12h, 18h and 24h and probed for antibodies specific to CHOP, GRP-94, Phospho-elf, ATF-4 and

Phospho-PERK. -actin was used in ascertain equal protein loading in all the lanes. Band

intensities of ER stress markers were analyzed after normalization with -actin and were presented

next to western blot. The experiment is repeated three times and a representative blot was shown.

Each treatment group is represented as Mean ± S.E.M. Statistical differences between Tweak

treated groups and controls are analyzed using ANOVA followed by Student Newman Keul’s post

test. Asterisk (p < 0.01, *** p < 0.001) represents significant differences between Tweak treated

groups and control group.

248

249

250

Fig.6 Autophagosomal lysosomal marker modulation in Tweak stimulated Bv2 microglial

cells. A-E. Increased Autophagy marker expression in Tweak treated BV2 microglia. Western blot

analysis of cell lysates treated from BV2 microglial cells were treated with Tweak (100 ng/ml) for

3-24h using antibodies specific to LC3, Beclin, P62 , ATG 5-12 and P-AMPK. All the lanes had

equal protein loading that were confirmed with -actin that was used as a internal loading control.

Bar graphs represent mean ± S.E.M with results are representative of at least 2 independent

experiments. Significant upregulation of autophagy markers was demonstrated in Tweak treated

BV2 microglia. Statistical differences between Tweak treated groups and controls was carried out

using ANOVA followed by Student Newman Keul’s post test. Asterisk (* p < 0.05,** p < 0.01,

*** p < 0.001) represents significant differences between Tweak treated groups as compared with

control. F-G. Lysosomal dysfunction with release of Cathepsin-B and Cathepsin-D in Tweak

treated BV2microglia. BV2 microglial cells were treated with 100 ng/ml Tweak for increasing

time periods (3-24h) and proteins are resolved by SDS-PAGE and immunoblotted for Cathepsin-

B and Cathepsin-D to detect Immature and mature active forms. Optical densities of protein bands

were analyzed with the Odyssey imaging system (LI-COR) and -actin was used a internal loading

control. Immunoblot analysis shows time dependent increase in secretion of active form of

cathepsin B and D in Tweak treated BV2 microglial cells. Results are representative of at least 2

independent experiments and a representative western blot is shown. Statistical analysis was

carried out using ANOVA followed by Student Newman Keul’s post test. Asterisk (*** p < 0.001)

represents significant differences between Tweak treated groups and control group. H. Increased

Lysosomal PH in Tweak treated BV2 cells. BV2 microglial cells were treated with 100 ng/ml

Tweak for 3h, 6h, 12h, 18h and 24h. Post treatment, lysosomal pH quantification was further

performed using the ratiometric lysosomal pH probe LysoSensor Yellow/Blue dextran (Invitrogen

L22460). Changes in lysosomal PH was determined by using the fluorescent microplate reader

with emission wavelengths at 535 nm and 430 nm with excitation at 340 nm. The experiment is

repeated three times with N=6 and expressed as percent of control. The data represent mean ±

S.E.M. Statistical analysis is performed with ANOVA followed by Student Newman Keul’s post

test. Asterisk (*** p < 0.001) represents significant differences between Tweak treated groups and

control groups

251

252

253

Figure 7. Spleen Tyrosine Kinase regulates NLRP3 related markers in Tweak treated

microglial cells. A-C. R788 attenuates NLRP3 inflammasome related markers in the Tweak

treated BV2 microglial cells. BV2 microglial cells were pretreated with Syk inhibitor R788 (3uM

1hr) followed by treatment with Tweak (100ng/ml) stimulated BV2 microglial for 24h. Western

blot analysis of whole cell lysates from above treated microglial cells with NLRP3 related markers

showed that R788 pretreatment attenuated NLRP3, caspase-1 and IL-1 beta activation in Tweak

treated BV2 microglial cells. The band intensities of each treatment group were normalized with

-actin and presented as Mean ± S.E.M. The experiment is repeated for 2 times and a representative

blot is shown. Asterisk (* p < 0.05) represents significant differences between Tweak treated

groups and control groups as determined by ANOVA followed by Student Newman Keul’s post

test. D. SiRNA mediated SYK knockdown resulted in efficient knockdown of SYK expression in

BV2 cells. BV2 microglial cells were transfected with either scrambled or SYK siRNA. 72 hrs

post transfection, BV2 microglial cells were subjected to immunoblot analysis for SYK to confirm

efficient downregulation of SYK expression. Representative immunoblot image confirming the

SiRNA knockdown of SYK but not in scrambled SiRNA, reduces the SYK protein to 20% of

control levels in BV2 microglia (D). Asterisk (* p < 0.05) demonstrates significant differences

between Si RNA SYK transfected groups and scrambled SiRNA transfected groups as determined

by ANOVA followed by Student Newman Keul’s post test. E-F. Syk knockdown attenuates

NLRP3 inflammasome markers in Tweak treated microglial cells. 72h post-siRNA transfection

with either scrambled or the Syk siRNA, BV2 microglial cells were subjected to the Tweak (100

ng/ml) stimulation for 24h. Whole cell lysates were later probed with antibodies specific to NLRP3

and caspase-1 with equal loading ascertained with re-probing the membranes with -actin. Results

are representative of at least 3 independent experiments. Syk knockdown attenuated Tweak

mediated upregulated NLRP3 and Caspase-1 in BV2 microglia. Statistical differences between

treatment groups were determined by ANOVA followed by Student Newman Keul’s post test.

Asterisk (** p < 0.01 & *** p < 0.001) represents significant differences between as Tweak / SYK

SiRNA treated groups and Tweak treated groups.

254

255

Figure 8. Involvement of Syk in MAPK activation in microglial cells. A-C. Increased MAP

kinases expression in Tweak treated BV2 microglia. BV2 microglial cells were treated with 100

ng/ml Tweak for 30 min, 60 min, 120 min and 180 min. Post treatment, whole cell lysates were

harvested and separated with SDS-PAGE. MAPK phosphorylation status was probed by western

blotting using antibodies specific to Phospho-P38, Phospho-JNK and Phospho-ERK. Values are

means ± S.E.M obtained from three independent experiments. Representative western blot

revealed significant upregulation of MAPKs Phospho- P38 (A), Phospho-C Jun ( B) and Phospho-

ERK (C) in Tweak treated BV2 microglial cells. Statistical differences between Tweak treated

groups and control was carried out using Anova followed by Student Newman Keul’s post test.

Asterisk (* p < 0.05,** p < 0.01) represents significant differences as compared with control group.

256

D-E. R788 attenuates MAPK activation in the Tweak treated BV2 microglial cells. BV2 microglial

cells were pretreated with Syk inhibitor R788 (3uM 1hr) followed by treatment with Tweak

(100ng/ml) stimulated BV2 microglial for 1h. Western blot analysis of whole cell lysates from

above treated microglial cells showed that R788 pretreatment attenuated MAPK activation in

Tweak treated BV2 microglial cells. The band intensities of each treatment group were normalized

with -actin and presented as Mean ± S.E.M. The experiment is repeated for 2 times and a

representative blot is shown. Asterisk (* p < 0.05) represents significant differences between

Tweak treated groups and control groups as determined by Student Newman Keul’s post test.

257

258

Figure 9. Syk regulates Tweak induced impairment of auto-lysosomal system and GSK-3

beta in Bv2 microglial cells. A-B. Syk knockdown attenuates Tweak induced impairment of auto-

lysosomal system (ALS) in Bv2 microglial cells. BV2 Cells were transfected with either scrambled

or the Syk siRNAs for 72 h. The transfected microglia were subsequently treated with and without

Tweak (100ng/ml) for 24 h. Post treatment, whole cell lysates were collected, separated with SDS-

PAGE and probed with antibodies specific for LC3 and Beclin. Equal protein loading in all the

lanes is ascertained with -actin. Results are representative of at least 4 independent experiments.

Syk knockdown attenuated Tweak mediated upregulated LC3 and Beclin (A-B) in BV2 microglia.

Asterisk (*** p < 0.001) represents significant differences between as Tweak / SYK SiRNA treated

groups and Tweak treated groups. C-D. SiRNA mediated SYK knockdown and SYK inhibitor

(R788) attenuates Tweak induced Cathepsin-B expression in BV2 microglial cells. For

knockdown studies, BV2 microglia were transfected with scrambed and Syk scrambled SiRNA

for 72h and then treated with or without Tweak (100 ng/ml) for 24h. For SYK inhibitor studies,

BV2 microglia were pre-incubated with SYK inhibitor R788 (3uM) or DMSO for 1hr and then

stimulated with or without Tweak (100 ng/ml) for 24h. Post treatment, whole cell lysates were

separated with SDS-PAGE and probed for Cathepsin-B using western blot with equal loading

ascertained by actin. The experiments were repeated for four times and a representative western

blot was shown. Si RNA mediated SYK knockdown or SYK inhibition by R-788 suppressed

Tweak mediated Cathepsin-B upregulation (C-D) in BV2 microglial cells. Asterisk (* p < 0.05,

*** p < 0.001) indicates significant differences between Tweak / SYK Si RNA / R788 treated

groups and Tweak treated groups as determined by Anova followed by Student Newman Keul’s

post test. E-F. SiRNA mediated SYK knockdown and SYK inhibitor (R788) attenuates Tweak

induced GSK-3 beta Y216 expression in BV2 microglial cells. For knockdown studies, BV2

microglia were transfected with scrambed and Syk scrambled SiRNA for 72h and then treated with

or without Tweak (100 ng/ml) for 24h. For SYK inhibitor studies, BV2 microglia were pre-

incubated with SYK inhibitor R788 (3uM) or DMSO for 1hr and then stimulated with or without

Tweak (100 ng/ml) for 24h. Post treatment, whole cell lysates were probed for GSK-3 beta Y216

using western blot with equal loading ascertained by actin. The experiments were repeated for

four times and a representative western blot was shown. Densitometric scanning analysis revealed

that, Si RNA mediated SYK knockdown or SYK inhibition by R-788 attentuated Tweak mediated

GSK-3 Beta Y 216 upregulation (E-F) in BV2 microglial cells. Asterisk (* p < 0.05, *** p <

0.001) indicates significant differences between Tweak / SYK Si RNA / R788 treated groups and

Tweak treated groups as determined by Anova followed by Student Newman Keul’s post test.

259

260

261

262

Figure 10. Effect of Syk inhibitor Piceatannol on Tweak treated striatal brain tissues. A-G.

Mice were treated with 2 ul of PBS or Tweak (1ug/ml) injected stereotaxically into striatum with

and without 12h and 1h Piceatannol pretreatment. Mice were sacrificed after 24h Tweak treatment.

Striatal brain tissues were dissected out from PBS or Tweak with and without Piceatannol

pretreated mice 24h after Tweak treatment. Striatal tissues lysates were prepared, separated with

SDS-PAGE and probed by western blotting for Syk, GSK-3 beta Y216, Beclin, LC3 II, CHOP,

Cathepsin-B and Caspase-1. -actin is used to ensure equal protein loading in the lanes. A

representative western blot is shown and experiment is repeated two times. The normalized band

intensity for each treatment group expressed as percentage of control group was plotted on

histogram. No significant difference in the striatal inflammatory markers between control and

TWEAK infused mice was evidenced. In a similar fashion Piceatannol pretreatment treatment had

no effect on TWEAK-induced modulation of inflammation related marker expression in the

striatum Statistical analysis was carried out using Anova followed by Student Newman Keul’s post

test.

263

CHAPTER 4. SPLEEN TYROSINE KINASE MEDIATES NLRP3 INFLAMMASOME

ACTIVATION IN LPS STIMULATED MICROGLIA VIA NF-KB ACTIVATION, MAP

KINASE AND ROS GENERATION

To be Submitted to Journal of Biological Chemistry

Sri Kanuri1,2, Wesley Schoo1, Huajun Jin1, Vellareddy Anantharam1, Anumantha Kanthasamy1 and

Arthi Kanthasamy1*

Abstract

Parkinson’s disease (PD) is a multifactorial neurodegenerative disorder that is

characterized by pronounced microglial activation response. Accumulating evidence indicates that

dopaminergic neurotoxicity may results from the cross talk between oxidative stress and the

activation of stress sensitive kinases in the microglia. The present study investigated the anti-

inflammatory effects of the spleen tyrosine kinase (Syk) inhibitor R788 in an in vitro N9 microglial

cell line challenged with LPS. Exposure of N9 microglial cells led to a time dependent increase in

Syk activation, ROS generation, MAPK and NF-kB activation. Further LPS induced NF-kB

activation preceded the activation of NLRP3 inflammasome as evidenced by the increased

expression levels of NLRP3 as well as mature IL-1β, and Cas-1. Intriguingly, inhibition of Syk via

R788 suppressed the activation of microglia via the suppression of iROS and nitric oxide

production, phosphorylation of NF-kB, mitogen activated protein kinases (MAPKs), extracellular

signal regulated kinases 1 and 2 (ERK1/2), p38 MAPK and glycogen synthase kinase-3β (GSK3β).

Notably the activation of the NLRP3 inflammasome was markedly inhibited by R788. In

conclusion, these data suggest that R788 exerts its anti-inflammatory response upon LPS

1 Department of Biomedical Sciences, Iowa State University, Ames, IA 50010

2 Primary researcher and author

*To whom correspondence should be addressed. E-mail: [email protected]

264

stimulation by inhibiting NLRP3 inflammasome activation which might be partly attributed to

inhibition of NF-kB activation and MAPK activation as well as down regulation of ROS

production. Thus, R788 may serve as a useful therapeutic intervention for the treatment of

neuroinflammatory disorders including PD.

Introduction

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

approximately 1% of the population at age 55 and nearly 5% of the age of 85 (Block, Zecca, et al.,

2007; Hirsch & Hunot, 2009). It is characterized by the degeneration of dopaminergic neurons in

the substantia nigra resulting in the motor deficits recognized as the cardinal signs of the disease:

rigidity, bradykinesia, and tremor (Hirsch et al., 2012). Though the direct cause of this neuronal

loss is not fully understood, there has been evidence suggesting that neuroinflammation could be

involved in the progressive degeneration of these neurons (Block, Zecca, et al., 2007; L. Qin et al.,

2007).

Microglia, the resident innate immune cell, are present in varying concentrations

throughout the brain with the highest concentration in the substantia nigra (Block, Zecca, et al.,

2007). Normally, these cells exist in a resting, inactivated state but can be activated by

immunological stimuli (Block, Zecca, et al., 2007). When activated, microglia exhibit increased

expression of cellular receptors and molecules including pattern recognition receptors (PRR), Toll-

like receptors (TLR), and NADPH oxidase (Y. S. Kim et al., 2006; Rock et al., 2004); however,

upon chronic activation, these enzymes can potentially produce toxic amounts of their cytotoxic

products such as reactive oxygen species (ROS) and nitrous oxide (NO) (Hirsch et al., 2009)

thereby leading to neuronal death. Neuronal death or the release of damaging molecules is one

stimulus that can induce this over activated state in microglia, resulting in the phenomena known

265

as reactive microgliosis (Block & Hong, 2007; Block, Zecca, et al., 2007). Lipopolysaccharide

(LPS) activation of microglia via the transmembrane receptor TLR4 is frequently used as a

neuroinflammatory model system and has been found to cause progressive dopaminergic

neurodegeneration reminiscent of PD (Block, Zecca, et al., 2007).

Spleen tyrosine kinase (Syk) is a cytoplasmic non-receptor tyrosine kinase that has been

shown to be involved in the receptor signaling pathways of a number of diverse cell types (Mocsai

et al., 2010). Structurally, Syk has two SRC homology 2 domains and a C-terminus kinase domain

with a molecular weight of 72 kDa (Mocsai et al., 2010; Y. S. Yi et al., 2014a). It has a well-

documented and crucial role in the intracellular signaling pathways of the classical

immunoreceptors; B cell receptors, T cell receptors and Fc receptors (Mocsai et al., 2010). Syk

has been shown to be important for proper development of both T and B cells, with varying

involvement at different stages in each cell type’s development (Mocsai et al., 2010). This adaptive

immune cell signaling acts via immunoreceptor tyrosine-based activation motifs (ITAMs) in which

the phosphorylation of the cytoplasmic tyrosine residues results in the recruitment of Syk (Mocsai

et al., 2010). Syk changes conformation when it binds to the phosphorylated ITAMs, allowing for

activation, autophosphorylation, and dissociation from the ITAM (Y. S. Yi et al., 2014a). It is

then able to activate downstream proteins in the signaling pathway via phosphorylation or direct

association (Y. S. Yi et al., 2014a). Syk has more recently been found to have a role in several

other cellular functions through activation by different upstream molecules and receptors (Mocsai

et al., 2010). Transmembrane integrins utilize ITAM-containing transmembrane adaptor

molecules to facilitate phosphorylation of Syk’s SH2 domain tyrosines, resulting in adhesion and

migration in leukocytes, activation of platelets, and promotes development of osteoclasts (Mocsai

et al., 2010). Syk has also been implicated in signaling pathways activated by P-selectin

266

glycoprotein ligand-1 resulting in transcriptional activation via the ITAM related sequences of the

ezrin-radixin-moesin proteins (Berton et al., 2005).

Previous studies indicate that LPS is associated with chronic neuro-inflammation and

progressive neuro-degenration. Systemic administration of LPS in mice resulted in microglial

NLRP3 inflammasome activation, increased pro-inflammatory cytokines (TNF-alpha, IL-1 beta

and MCP-1) and reduction of dopaminergic neurons in substantia nigra (L. Qin et al., 2007; Z.-T.

Zhang et al., 2016; W. Zhu et al., 2017). Accumulating evidence suggests that, LPS induces neuro-

inflammatory responses through spleen tyrosine kinase in immune cells. In immune cells, LPS

induced activation of spleen tyrosine kinase resulted in NF-kB activation, NLRP3 inflammasome

formation, ROS generation and pro-inflammatory cytokines production (Miller et al., 2012).

Recent study reported that, Syk tyrosine kinase is essential for LPS mediated inflammation in

macrophages (Ulanova et al., 2007). Another study mentions that, spleen tyrosine kinase is

important for LPS induced regulation of inflammatory responses and endotoxin tolerance through

degradation of RIP140 in macrophages (P. C. Ho et al., 2012).

The multiple roles of Syk in leukocyte activation, function and inflammatory response has

increased interest in its possible role in the pathogenesis and progression of many immune-related

disorders and diseases (Y. S. Yi et al., 2014a). In this study, we propose to characterize the role

of Syk and its downstream signaling pathways in LPS induced proinflammatory signaling cascade,

including NLRP3 inflammasome activation in N9 microglia. Further characterization of these

signaling pathways would provide a better understanding of the therapeutic efficacy of Syk

inhibitors in modulating excessive immunomodulatory response as evidenced in Parkinson’s

disease.

267

Materials and methods

Chemicals and Reagents

IMDM, fetal bovine serum (FBS), L-glutamine, IRDye®-tagged 2°antibodies, penicillin,

and streptomycin were purchased from Invitrogen (Gaithersburg, MD). 10X Tris glycine SDS

buffer, 10X Tris glycine, 10% Tween-20, and nitrocellulose membrane were purchased from Bio-

Rad (Hercules, CA). APS, TEMED, lauryl sulfate and Bromophenol Blue were purchased from

Sigma Chemicals (St. Louis, MO). Phospho-p38 and Phospho-ERK antibodies were purchased

from Cell Signaling Technology, MA. Syk, Phospho-p65 and Nox-2 antibodies were obtained

from Santa Cruz Biotechnology. NLRP3 antibody was obtained from Novus Biologicals. Caspase-

1 antibody was purchased from Adipogen. GSK3β Tyrosine 216 antibody was obtained from

Abcam.

N9 Microglial Cultures

The immortalized murine N9 microglial cells were maintained in Iscove's Modified

Dulbecco's Medium (IMDM) containing 10% FBS, 50 U/ml penicillin, 50µg/ml streptomycin, and

2mM L-glutamine and incubated in a humidified atmosphere of 5% CO2 at 37°C. Prior to

experiments cells were plated and allowed to settle for at least 24 hours at 37°C in 5% CO2. Cells

were treated with 1µg/ml LPS in IMDM with supplements, containing 0% FBS, for the duration

indicated in the experiments.

Immunoblotting analysis

N9 cell lysates were prepared using modified RIPA buffer before normalization for equal

amounts using the Bradford Protein Assay Kit. Equal amounts of protein were loaded and samples

268

were separated on 12% SDS PAGE gels. Proteins were then transferred to a nitrocellulose

membrane and probed overnight at 4°C with their respective 1° antibodies; Caspase-1, Syk,

Phospho-p65, iNOS, Phospho-p38, Phospho-ERK & GSK3β Tyrosine 216. Membranes were then

washed three times with PBS containing 0.1% Tween-20, then IRDye®-tagged 2°antibodies (IR

Dye 800-conjugated anti-rabbit IgG (1:5000) or Alexa Flour 680-conjugated anti-mouse IgG

(1:10000; Molecular Probes, Invitrogen)) for 1hr at room temperature were added. Membranes

were visualized on the Odyssey infrared imaging system. β-actin (1:5,000) was used as a loading

control.

ROS Assay

Intracellular ROS was determined using 2′,7′-dichlorodihydrofluorescein diacetate assay

(DCFH-DA), a fluorescent probe, per previous published reports with minor alterations (L. Qian

et al., 2007). N9 microglial cells were plated in 96-well plates and treated with LPS 1ug/ml for

12h and 24h and 3 uM R788 for 1h. Post incubation, treatment media was removed and cells were

washed with PBS and incubated with 1uM CM-H2DCFDA dye dissolved in HBSS for 1 hr. Later,

cells were washed with PBS and ROS production was assessed using a microplate reader with

excitation 485 nm and emission 530 nm respectively using a Synergy2 multimode microplate

reader. Control culture fluorescence was subtracted as background and was interpreted as

increased iROS (L. Qian et al., 2007).

269

Nitric oxide detection.

Nitric oxide production was determined by indirect quantification of nitrite, an end product

of NO oxidation, in the supernatant using the Griess reagent (sigma Aldrich). N9 microglial cells

were plated at a density of 105 cells per well in a 96-well plate before treating with 1µg/ml LPS

for 12hr and 24hr; or were pretreated with 3µM R788 for 1h. After the incubation, 50µl of the

supernatant was mixed with 50µl of Griess reagent in a new 96-well plate and incubated on a plate

shaker at room temperature for 15 minutes. Absorbance was measured at 450nm using a Synergy2

multimode microplate reader. Sample concentrations of nitrite were determined by comparison to

sodium nitrite standard curve.

Syk Inhibitor R788

Syk inhibitor (R788) was purchased from Santa Cruz Biotechnology. The Syk inhibitor is

dissolved in DMSO and stored at -20C for future use. We used the 3uM R788 pretreatment for 1h

in N9 microglia and later treated with LPS 1ug/ml for respective time points (Y. C. Lin et al.,

2010).

Data analysis

Data analysis was calculated using Prism 7.02 (GraphPad Software, San Diego, CA). Data

was analyzed using one-way ANOVA, expressed as mean ± SEM. Differences of p<0.05 were

considered statistically significant.

270

Results

Upregulation of Syk and Activation of NLRP3 Inflammasome in N9 Microglial Cells

Exposed to LPS

It has been previously shown that exposure of neutrophils to LPS induced the

phosphorylation of Syk leading to the complex formation encompassing TLR4, p-Syk and p-

CEACAM1 (R. Lu et al., 2012). Furthermore LPS treatment was found to induce ROS generation,

caspase-1 induction, and IL-1β secretion in neutrophils (R. Lu et al., 2012) therefore we sought to

investigate the expression of Syk and inflammasome markers in N9 microglial cells exposed to

LPS by immunoblotting analysis. N9 microglial cells are a well characterized immortalized

microglial cell line that has been routinely used for examining the therapeutic efficacy of anti-

inflammatory agents. Briefly, N9 microglial cells were exposed to LPS for 3, 6, 12, and 24h in the

presence or absence of Syk inhibitor, R788, and the levels of Syk and NLRP3 related

proinflammatory markers were determined by Western blot analysis. As shown in Figure 1A Syk

expression increased at 6hr after LPS challenge, peaked at 12h, and remained elevated for the

reminder of the treatment period. An upregulation of Syk was accompanied by the upregulation

of proinflammatory markers as evidenced by the induction of NLRP3 related markers including

procaspase-1 and caspase-1respectively. The results demonstrated a time dependent increase in

the levels of NLRP3 markers whereby a maximal increase in the levels of procaspase-1, cleaved

caspase were evidenced at 12 and 24h respectively upon LPS challenge (Fig. 1A). To further

evaluate the influence of Syk on LPS-induced upregulation of NLRP3 markers we tested the anti-

inflammatory effects of Syk inhibitor R788 on LPS-induced upregulation of NLRP3 markers.

Remarkably, R788 suppressed LPS-induced upregulation of NLRP3 markers as measured by a

dramatic reduction in the levels of NLRP3, caspase-1, pro-IL-1β, and IL-1β respectively (Fig. 1B).

Therefore, our results suggest that Syk is a key upstream regulator of NLRP3 inflammasome and

271

that R788 elicits its anti-inflammatory effects via suppression of NLRP3 inflammasome activation

in LPS stimulated microglia.

Fig 1. LPS-induces NLRP3 inflammasome activation via a Syk dependent mechanism. (A)

SYK and NLRP3 related markers expression in LPS treated N9 microglia. N9 microglia were

treated with 1µg/ml LPS for 3hr, 6hr, 12hr, and 24hr. The control group is treated with vehicle (1x

PBS) used to prepare LPS prior to treatment. Post incubation, whole cell lysates were separated

with SDS-PAGE, and probed by western blotting using antibody against SYK, NLRP3, caspase-1

(pro and cleaved) and β actin. -actin was used a internal loading control for equal loading. The

1A

1B

272

experiment is repeated two times and a representative western blot is presented. Western blot

analysis demonstrated that is dramatic upregulation of SYK and NLRP3 related markers in LPS

treated N9 microglia. (B) R788 attenuates the LPS induced NLRP3 related markers in N9

microglia. N9 microglia were treated with 1µg/ml LPS with or without one hour pre-treatment

with 2.5 uM and 3.5 µM R788 for 24h and probed with antibodies specific to NLRP3, caspase-1,

IL-1 beta with western blot analysis. To confirm equal protein loading in all the lanes, membranes

were re-probed with -actin. Results are representative of at least 2 independent experiments.

Western blot analysis revealed that, R788 downregulated LPS induced upregulation of NLRP3,

caspase-1, IL-1 beta in N9 microglia.

R788 partially Inhibited The Generation of ROS And Nitrite Species via Syk in LPS-

Stimulated N9 Microglial Cells

To investigate the potential relationship between Syk and ROS generation we used Western

blot analysis (Fig 2A). N9 microglia were treated with 1µg/ml LPS for 12h and 24h in the presence

or absence of 3µM Syk inhibitor R788. The stimulation of N9 microglial cells with LPS resulted

in significant (p<0.001) increase in the levels of iROS at both the 12h and 24h time points

respectively. However pretreatment of N9 microglial cells with R788 significantly inhibited LPS

induced iROS generation (Fig 2A). This suggest that Syk is a critical regulator of intracellular

ROS generation.

Next, in order to assess how Syk activation is linked to nitrite generation we performed the

Griess assay. N9 cells were subjected to the same treatment conditions as detailed above. The

stimulation of N9 microglial cells with LPS led to a significant increase in the release of nitrite

species in the conditioned media; however pretreatment with R788 significantly suppressed nitrite

generation (Fig 2B). Next to investigate the mechanistic basis of R788 inhibitory effects on NO

generation we determined the levels of iNOS using Western blot analysis. LPS induced a rapid

and transient induction of iNOS (Fig 2C); however, R788 markedly reduced iNOS protein

expression in LPS stimulated N9 microglial cells (Fig. 2D). Taken together these results suggest

that Syk is involved in the generation of iROS and NO through the upregulation of iNOS in LPS

273

stimulated microglia. This suggest that R788 might elicit its anti-inflammatory effects via its

antioxidant effects in LPS stimulated microglia.

2A

2B

2C

274

Fig 2 Effect of R788 on LPS-induced iROS and NO generation in N9 microglial cells. (A).

R788 partially attenuates ROS production in LPS treated microglia. N9 cells were treated with

LPS (1µg/ml) for 12hr and 24hr with or without pretreatment with 3µM R788 for 1hr. Post-

treatment, reactive oxygen species production was quantified spectrophotometrically using redox

sensitive CM-H2DCFDA. The data is represented as percent of control and the experiment is

repeated 2 times with n=8 in each treatment group. Statistical differences were determined by

ANOVA followed by Student Newman Keul’s test. Asterisk (p***<0.001) represents significant

differences between LPS treated groups and control group. (B). R788 partially attenuates NOS

production in LPS treated N9 microglia. N9 cells were treated with LPS (1µg/ml) for 12hr and

24hr with or without pretreatment with 3µM R788 for 1hr. At the end of treatment times, 100 μl

of supernatant is mixed with equal volume of griess reagent. Later, the absorbance is measured at

540 nm using multimode microplate reader and nitrate concentration was determined from sodium

nitrite standard curve. The data is represented as percent of control and the experiment is repeated

2 times with n=8 in each treatment group. Statistical differences were determined by ANOVA

2D

275

followed by Student Newman Keul’s test. Asterisk (p***<0.001) represents significant differences

between LPS treated groups and control group. (C) iNOS expression in LPS treated N9 microglia.

N9 microglia were treated with LPS (1µg/ml) for 30 min, 1h and 2h. Whole cell lysates were

separated with SDS-PAGE and subjected to western blot analysis by probing with iNOS antibody.

Equal protein is loaded in all the lanes with the help of -actin. The experiments are repeated two

times and a representative western blot is presented. Immunoblot analysis revealed that there is

dramatic upregulation of iNOS production in LPS treated N9 microglia.

Inhibition of ERK and p38 MAPK Activation By R788 in LPS Stimulated N9 Microglial

Cells.

It is well established that MAPK signaling events are linked to cytokine and LPS-simulated

upregulation of iNOS in microglial cells (Fiebich et al., 2002). Next we examined whether the

suppressive effect of R788 on the generation of proinflammatory mediators occurs via MAPK

signaling pathway. N9 cells were treated as described above and the levels of the phospho forms

of ERK and p38 MAPK were determined via Western blot analysis. Our studies revealed that LPS

induced a dramatic time dependent increase in the phosphorylation of p38 MAPK and ERK. Upon

LPS stimulation for increasing duration (30-6h) p38 MAPK phosphorylation was evidenced as

early as 30 min, peaked at 60’, and nearly diminishes at 2h following LPS challenge (Fig. 3A).

However the phosphorylation of ERK is evidenced at 30’ of LPS stimulation and remains elevated

for 60’ and a marked reduction was observed at 2hr (Fig. 3A). Furthermore, R788 remarkably

reduced the LPS-induced phosphorylation of p38 MAPK and ERK at 1h (Fig. 3B). These findings

suggest that R788 is an effective inhibitor of p38 MAPK and ERK in LPS-stimulated microglia

and that Syk may serve as a critical regulator of the activation of MAPK signaling pathways.

276

Fig 3. Involvement of Syk in LPS-induced upregulation of MAPK signaling pathways in N9

microglial cells. (A) Immunoblot analysis of LPS (1µg/ml) treated N9 microglia for 30min, 1h

and 2h and MAPK phosphorylation status was probed using antibodies specific for Phospho-ERK

and Phospho-P38. Membranes were re-probed with -actin which was used a loading control. The

experiments were repeated for 2 times and a representative immunoblot is shown. Immunoblot

analysis shows that is there is rapid and transient upregulation of MAP kinases Phospho-ERK and

Phospho-P38 in LPS treated microglia. (B) R788 attenuates the LPS induced MAPK activation in

N9 microglia. N9 microglia were treated with 1µg/ml LPS with or without one hour pre-treatment

2.5 uM and 3.5 µM R788 for 1h and subjected to immunoblot analysis using antibodies Phospho-

P38 and Phospho-ERK. To confirm equal protein loading in all the lanes, membranes were re-

probed with -actin. Results are representative of at least 2 independent experiments. Western blot

analysis revealed that, R788 downregulated LPS induced upregulation of Phospho-P38 and

Phospho-ERK in N9 microglia.

3B

3A

277

Suppression of LPS-Induced Activation of the Transcription Regulator NF-kB via R788 in

N9 Microglial Cells

It is well established that NF-kB serves as a critical intermediate in promoting

proinflammatory gene expression (Ulanova et al., 2007). Moreover, NF-kB has been identified as

a key contributor to the priming step during the activation of NLRP3 (Y. C. Lin et al., 2015).

Therefore, we asked whether R788-mediated inhibition of NF-kB activation may underlie its anti-

inflammatory responses. We used Western blot analysis to determine the effects of the compound

on LPS-induced phosphorylation of the p65 subunit of NF-kB in N9 microglial cells. As shown

in Fig. 4A treatment of N9 microglia with 1µg/ml LPS for 30 min, 1h, and 2h resulted in a rapid

and transient upregulation of pp65. This process was markedly inhibited by R788 (Fig. 4B). This

suggests that R788 via its inhibitory effects on NF-kB activation might not only attenuate NLRP3

inflammasome activation but also proinflammatory cytokine production in a Syk dependent

manner.

4A

278

Fig 4. LPS-induced activation of NF-kB inhibited by treatment with R788. A. Immunoblot

analysis of LPS (1µg/ml) treated N9 microglia for 30min, 1h and 2h and activation of NF-kB

signaling pathway was probed using antibodies against NF-kB P65. Membranes were re-probed

with -actin which was used a loading control. The experiments were repeated for 2 times and a

representative immunoblot is shown. Immunoblot analysis shows that is there is early activation

of NF-kB P65 pathway in LPS treated N9 microglia. B. R788 attenuates the LPS induced NF-kB

signaling pathways in N9 microglia. N9 microglia were treated with 1µg/ml LPS with or without

one hour pre-treatment 2.5 uM and 3.5 µM R788 for 1h and NF-kB immunoblot analysis was

performed with NF-kB P65. To confirm equal protein loading in all the lanes, membranes were re-

probed with -actin. Results are representative of at least 2 independent experiments. Western blot

analysis revealed that, R788 downregulated LPS induced upregulation of Phospho-P65 in N9

microglia.

4B

279

R788 Attenuates LPS-Induced GSK3β Activation

The role of glycogen synthase kinase 3β (GSK3β) in peripheral and CNS disorders are well

established (Bhat et al., 2004). However, the influence of Syk on GSK3β activation during LPS-

induced microglial activation remains poorly characterized therefore N9 microglial cells were

exposed to LPS for varying duration. At the end of the incubation period cell lysates were prepared

and probed for the phosphorylation status of GSK3β. As shown in Fig 5A, exposure of N9 cells

to LPS resulted in a time dependent increase in the phosphorylation status of GSK3β. GSK3β was

activated as early 2h peaked at 12h and remained elevated for the reminder of the treatment period.

We next determined whether inhibition of GSK3β activation may be linked to the anti-

inflammatory effects of R788 at 24h. R788 pretreatment in LPS treated cells caused a near

complete abrogation of GSK3β phosphorylation as compared to LPS only treated cells (Fig 5B).

Therefore our results suggest that R788 mediated inhibition of GSK3β activation might at least in

part contribute to its anti-inflammatory effects.

5A

280

Fig 5. R788 attentuates LPS induced GSK-3 beta activation in N9 microglial cells. A. GSK-3

beta Y 216 upregulated expression in LPS treated N9 microglia. N9 microglia were treated with

1µg/ml LPS for 30min, 1h, 2h, 6h, 12h and 24h. Whole cell lysates were separated with SDS-

PAGE and GSK-3 beta phosphorylation status was probed by using antibodies specific to for GSK-

3 beta Y216. Equal protein is loaded in all the lanes with the help of -actin. Band intensities

normalized to -actin reveal that there is dramatic time dependent increase in GSK-3 beta Y216

in LPS treated N9 microglia. The experiments are repeated two times and a representative western

blot is presented. Statistical differences between LPS treated groups and control groups were

assessed with ANOVA with followed by Student Newman Keul’s post test. B. R788 attenuates

the LPS induced GSK-3 beta Y 216 upregulation in N9 microglia. N9 microglia were treated with

1µg/ml LPS with or without one hour pre-treatment 2.5 uM and 3.5 µM R788 for 24h and GSK-3

beta immunoblot is performed as described. To confirm equal protein loading in all the lanes,

membranes were re-probed with -actin. Results are representative of at least 2 independent

experiments. Western blot analysis revealed that, R788 downregulated LPS induced upregulation

of GSK-3 beta Y216 in N9 microglia.

Discussion

LPS has previously been shown to activate microglia and lead to chronic activation similar

to that seen in neurodegenerative disorders like Parkinson’s disease (Block, Zecca, et al., 2007; H.

M. Gao et al., 2002). The excessive activation of the NLRP3 inflammasome has been suggested

to be linked to the induction of enhanced inflammatory response and has been identified as a

5B

281

critical component in the disease pathology of numerous disorders, including autoinflammatory

and neurodegenerative disorders (Gustin et al., 2015; Halle et al., 2008). In this study, we

investigated the involvement of Syk in the induction of the proinflammatory downstream signaling

pathway especially the activation of NLRP3 inflammasome upon stimulation of N9 microglial

cells with LPS. Herein we provide evidence that LPS enhanced Syk phosphorylation with a

concomitant activation of several positive mediators of the inflammasome, including ROS, ERK

and p38 MAP kinases, and NF-kB. Pharmacological inhibition of Syk by R788 suppressed the

activation of the afore mentioned proteins, suggesting an important role for Syk in NLRP3

inflammasome activation and downstream proinflammatory signaling mediators. This study, for

the first time, demonstrates the proinflammatory role of Syk during microglial activation response

following LPS treatment.

When immune cells are activated by treatment with lipopolysaccharide (LPS), a toll-like

receptor 4 (TLR4) ligand, Syk shows greatly increased activation in under 2 minutes, leading to

the activation of several downstream ligands, including the nod-like receptor family pyrin domain-

containing 3 (NLRP3) inflammasome and of nuclear factor-κB (NF-kB) (Miller et al., 2012; Y. S.

Yi et al., 2014a). This results in a range of cellular adaptations including the production of ROS,

cell differentiation and survival, and the increased expression of proinflammatory genes and

mediators (Miller et al., 2012; Y. S. Yi et al., 2014a). Consistent with these results we show a

similar pathway whereby microglia activation by LPS induces a time dependent increase in Syk

signaling events which is associated with the induction of proinflammatory response including

NLRP3 inflammasome activation pathway through ROS, MAPK and GSK3β signaling

mechanisms. Intriguingly pharmacological inhibition of Syk via R788 attenuated Syk dependent

downstream signaling events. Our findings are consistent with a recent report that demonstrated

282

that inhibition of Syk suppresses the inflammatory response in macrophages exposed to an

inflammatory stimuli (Y. S. Yi et al., 2014a).

The NLRP3 inflammasome is a multiprotein complex, composed of NLRP3, ASC,

and caspase-1, which are found in some cell types of the innate immune system and is responsible

for the maturation of cytokines including IL-1β and IL-18 (Guarda et al., 2011). NLRP3

inflammasome activation is a two-step process, requiring a first signal to induce the transcription

of NLRP3 and pro-IL-1β followed by a second signal leading to the activation of NLRP3

inflammasome and caspase-1 and the resultant maturation and secretion of IL-1β and IL-18 (Gross

et al., 2009). In this context a recent report demonstrated that microglial cells express the activated

NLRP3 inflammasome upon exposure to NLRP3 activating stimuli (Gustin et al., 2015). LPS is

frequently utilized as the priming signal for inflammasome activation, and some studies have

indicated that Syk plays a direct role in the formation of the inflammasome (Gross et al., 2009; Y.

C. Lin et al., 2015). Consistent with this, we found that LPS treatment of N9 microglia resulted in

the time dependent increased expression of Syk, NLRP3, pro-caspase-1, pro-IL-1β, cleaved

caspase-1 and IL-1β. When we blocked Syk, the increased expression of NLRP3, caspase-1

cleavage, and the maturation of IL-1β was not observed. This suggests a role for Syk in NLRP3

inflammasome activation. These findings are consistent with previous research in which LPS was

found to be sufficient for pro-IL-1β maturation and secretion by neutrophils (R. Lu et al., 2012).

Chronically activated microglia are a major source of ROS, chemokines and cytokines in

many neurodegenerative disorders (Block, Zecca, et al., 2007). The activation of NADPH oxidase

and iNOS in these microglia results in the excess production of free radicals that have been

implicated in the pathogenesis of neurodegenerative diseases (R. Gordon et al., 2016; Y. S. Yi et

al., 2014a). Syk has been shown to mediate NOX2 induction in macrophages after minimally

283

oxidized LDL binding to TLR4 and has been implicated in ROS production in human monocytes

(Bae et al., 2009; Miller et al., 2012). Furthermore, iNOS induction was found to be mediated in

a Syk dependent manner in LPS stimulated peritoneal macrophages and murine macrophages via

activation of MAP kinases (Chiou et al., 2011; S. H. Kim et al., 2015). Consistent with these

reports LPS stimulation of N9 microglia resulted in a marked increase in iNOS protein expression

with a concomitant increase in NO generation. Blockade of Syk resulted in an attenuated

expression iNOS, with an accompanying decrease in the generation of ROS and greatly diminished

NO release. Our studies demonstrate that Syk is an important regulator of ROS and NO release

which might be at least in part mediated via an NF-kB dependent mechanism (Chiou et al., 2011;

Y. S. Yi et al., 2014a).

MAP kinases play a crucial role as upstream regulators in the activation of several

transcription factors including NF-kB and several proinflammatory mediators, including iNOS,

IL-1β, and ROS production (Chaudhary et al., 2007; Y. S. Yi et al., 2014a). These proinflammatory

mediators have been linked to the proliferation of immune cells observed in neuroinflammatory

disorders. In addition, they have also been shown to be involved in the induction of NLRP3 and

pro-IL-1β, inflammasome components in LPS stimulated macrophages (Moon et al., 2015).

Moreover, Syk has previously been implicated as a positive mediator in both ERK and p38 MAP

kinase phosphorylation and activation (M. G. Ghonime et al., 2014; Miller et al., 2012). In the

present study, we found that LPS stimulation of microglia resulted in the rapid activation of both

ERK and p38 MAP kinase, suggesting Syk as a possible upstream regulator of MAPK activation.

As anticipated R788 inhibited LPS induced MAPK activation mechanisms further confirming that

Syk is a key upstream regulator of MAPK signaling events. Our results are in accordance with

previous findings in bone marrow derived macrophages exposed to Syk inhibitors (Eliopoulos et

284

al., 2006). Together our results indicate that Syk is a positive regulator of the MAP kinase

pathways which in turn presumably leads to the activation of NF-kB and resultant NLRP3

inflammasome activation, at least in LPS stimulated microglia cells.

GSK3β is a kinase involved in the regulation of very diverse functions, including cell cycle

control, apoptosis, cell differentiation and motility, and inflammation (Cortes-Vieyra et al., 2012).

GSK3β is regulated via autophosphorylation at tyrosine 216 resulting in kinase activation or

inhibited by phosphorylation of serine 9 (Cortes-Vieyra et al., 2012) resulting in a kinase-inactive

state. Furthermore, GSK3β has been shown to regulate the magnitude and duration of

inflammatory conditions as well as the progression of neurological disorders related to

neuroinflammation (Chan et al., 2009; Maixner & Weng, 2013; M. Martin et al., 2005; D. W. Park

et al., 2014). In this context, a recent report demonstrated that the dysregulation of GSK3β was

linked to Parkinsonian like pathophysiology with an accompanying brain region specific

phosphorylation and accumulation of tau and a-syn (Credle, George, et al., 2015). On the contrary,

siRNA mediated knock down of GSK3β or pharmacological inhibition of GSK3β was found to

diminish proinflammatory cytokine production in LPS stimulated monocytes (M. Martin et al.,

2005). In the same study, treatment of mice with GSK inhibitor SB216763 was found to

significantly attenuate LPS-induced mortality. Furthermore GSK3β has also been linked to the

suppression of anti-inflammatory cytokine generation, including IL-10 (Hu et al., 2006; M. Martin

et al., 2005). Consistent with these reports inhibition of GSK3β phosphorylation via R788

positively correlated with diminished proinflammatory mediator(s) generation suggesting a

positive modulatory role for GSK3β in the induction of proinflammatory signaling cascade. In

line with our findings, gene silencing of Syk was found to promote the generation of anti-

inflammatory cytokine IL-10 but suppress the levels of proinflammatory cytokines, TNF-a, IL-6

285

in a GSK3β dependent manner in dendritic cells (H. Yin et al., 2016). Together, our results suggest

that Syk and GSK3β cooperated with one and another via NF-kB activation to induce the

generation of proinflammatory mediators in LPS stimulated microglial cells.

Activation of NF-kB is a critical step in the transcription of many proinflammatory

mediators and microglial activation in response to LPS stimulation and Syk activation (Ulanova

et al., 2007; Y. S. Yi et al., 2014a). This activation is characterized by the phosphorylation and

activation of Inhibitory κB kinase (IKK) by MAP kinases, which then phosphorylates the

inhibitory protein inhibitory κB-α (IκBα) marking it for proteasomal degradation (N. Panicker, H.

Saminathan, H. Jin, M. Neal, D. S. Harischandra, et al., 2015). NF-kB subunit p65 subsequently

moves into the nucleus where it induces the transcription of proinflammatory genes and increased

NLRP3 and pro-IL-1β expression (Y. C. Lin et al., 2015). We showed increased Syk

phosphorylation was accompanied by phosphorylation of the p65 subunit of NF-kB during

stimulation of microglial cells with LPS in line with previous studies implicating Syk activation

in the induction of the NF-kB pathway in neutrophils and macrophages (Y. G. Lee et al., 2009;

Miller et al., 2012). Furthermore, blocking Syk activity in LPS treated cells also results in the

decreased activation of the NFkB reflected by decreased phosphorylation of p65 subunit of NF-

kB. Another study demonstrated decreased activation of NF-kB upon Syk inhibition in LPS

activated murine macrophages and peritoneal macrophages (Jeong et al., 2013). Consistent with

these studies we show that in R788 treated LPS challenged microglial cells a marked attenuation

of NFkB activation as indicated by decreased phosphorylation of the p65 subunit of NF-kB was

evidenced. These results suggest that Syk elicits its proinflammatory effect via an NF-kB

dependent cell signaling mechanisms at least in LPS stimulated microglial cells.

286

Taken together our results demonstrates how LPS stimulation of N9 microglia leads to the

activation of the NLRP3 inflammasome, GSK3β, MAP kinases and NF-kB, and ROS production

in a Syk dependent manner. Furthermore, the pharmacological inhibition of Syk by R788 results

in the inhibition of the afore mentioned proinflammatory mediators in LPS stimulated N9

microglial cells highlighting its anti-inflammatory effects. Taken together our results indicate that

R788 pretreatment attenuates LPS-induced NLRP3 inflammasome activation via suppression of

the maturation and release of IL-1β. Therefore, Syk may represent a novel therapeutic avenue for

he amelioration of neuroinflammation associated neurodegenerative disorders including PD.

Bibliography

Bae, Y. S., et al. (2009). Macrophages generate reactive oxygen species in response to minimally

oxidized low-density lipoprotein: toll-like receptor 4- and spleen tyrosine kinase-

dependent activation of NADPH oxidase 2. Circ Res, 104(2), 210-218, 221p following

218. doi:10.1161/CIRCRESAHA.108.181040

Berton, G., et al. (2005). Src and Syk kinases: key regulators of phagocytic cell activation.

Trends Immunol, 26(4), 208-214. doi:10.1016/j.it.2005.02.002

Bhat, R. V., et al. (2004). Glycogen synthase kinase 3: a drug target for CNS therapies. J

Neurochem, 89(6), 1313-1317. doi:10.1111/j.1471-4159.2004.02422.x

Block, M. L., et al. (2007). Chronic microglial activation and progressive dopaminergic

neurotoxicity. Biochem Soc Trans, 35(Pt 5), 1127-1132. doi:10.1042/BST0351127

Block, M. L., et al. (2007). Microglia-mediated neurotoxicity: uncovering the molecular

mechanisms. Nat Rev Neurosci, 8(1), 57-69. doi:10.1038/nrn2038

Chan, M. M., et al. (2009). A role for glycogen synthase kinase-3 in antagonizing mycobacterial

immune evasion by negatively regulating IL-10 induction. J Leukoc Biol, 86(2), 283-291.

doi:10.1189/jlb.0708442

287

Chaudhary, A., et al. (2007). Tyrosine kinase Syk associates with toll-like receptor 4 and

regulates signaling in human monocytic cells. Immunol Cell Biol, 85(3), 249-256.

doi:10.1038/sj.icb7100030

Chiou, W. F., et al. (2011). Psoralidin inhibits LPS-induced iNOS expression via repressing Syk-

mediated activation of PI3K-IKK-IkappaB signaling pathways. Eur J Pharmacol, 650(1),

102-109. doi:10.1016/j.ejphar.2010.10.004

Cortes-Vieyra, R., et al. (2012). Role of glycogen synthase kinase-3 beta in the inflammatory

response caused by bacterial pathogens. J Inflamm (Lond), 9(1), 23. doi:10.1186/1476-

9255-9-23

Credle, J. J., et al. (2015). GSK-3beta dysregulation contributes to parkinson's-like

pathophysiology with associated region-specific phosphorylation and accumulation of tau

and alpha-synuclein. Cell Death Differ, 22(5), 838-851. doi:10.1038/cdd.2014.179

Eliopoulos, A. G., et al. (2006). The tyrosine kinase Syk regulates TPL2 activation signals. J Biol

Chem, 281(3), 1371-1380. doi:10.1074/jbc.M506790200

Fiebich, B. L., et al. (2002). Inhibition of LPS-induced p42/44 MAP kinase activation and

iNOS/NO synthesis by parthenolide in rat primary microglial cells. J Neuroimmunol,

132(1-2), 18-24.

Gao, H. M., et al. (2002). Microglial activation-mediated delayed and progressive degeneration

of rat nigral dopaminergic neurons: relevance to Parkinson's disease. J Neurochem, 81(6),

1285-1297.

Ghonime, M. G., et al. (2014). Inflammasome Priming by Lipopolysaccharide Is Dependent

upon ERK Signaling and Proteasome Function. Journal of Immunology, 192(8), 3881-

3888. doi:10.4049/jimmunol.1301974

Gordon, R., et al. (2016). Protein kinase Cdelta upregulation in microglia drives

neuroinflammatory responses and dopaminergic neurodegeneration in experimental

models of Parkinson's disease. Neurobiol Dis, 93, 96-114. doi:10.1016/j.nbd.2016.04.008

Gross, O., et al. (2009). Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal

host defence. Nature, 459(7245), 433-436. doi:10.1038/nature07965

288

Guarda, G., et al. (2011). Differential expression of NLRP3 among hematopoietic cells. J

Immunol, 186(4), 2529-2534. doi:10.4049/jimmunol.1002720

Gustin, A., et al. (2015). NLRP3 Inflammasome Is Expressed and Functional in Mouse Brain

Microglia but Not in Astrocytes. PLoS One, 10(6), e0130624.

doi:10.1371/journal.pone.0130624

Halle, A., et al. (2008). The NALP3 inflammasome is involved in the innate immune response to

amyloid-beta. Nat Immunol, 9(8), 857-865. doi:10.1038/ni.1636

Hirsch, E. C., et al. (2009). Neuroinflammation in Parkinson's disease: a target for

neuroprotection? Lancet Neurol, 8(4), 382-397. doi:10.1016/S1474-4422(09)70062-6

Hirsch, E. C., et al. (2012). Neuroinflammation in Parkinson's disease. Parkinsonism Relat

Disord, 18 Suppl 1, S210-212. doi:10.1016/s1353-8020(11)70065-7

Ho, P. C., et al. (2012). NF-kappaB-mediated degradation of the coactivator RIP140 regulates

inflammatory responses and contributes to endotoxin tolerance. Nat Immunol, 13(4), 379-

386. doi:10.1038/ni.2238

Hu, X., et al. (2006). IFN-gamma suppresses IL-10 production and synergizes with TLR2 by

regulating GSK3 and CREB/AP-1 proteins. Immunity, 24(5), 563-574.

doi:10.1016/j.immuni.2006.02.014

Jeong, D., et al. (2013). In vitro and in vivo anti-inflammatory effect of Rhodomyrtus tomentosa

methanol extract. J Ethnopharmacol, 146(1), 205-213. doi:10.1016/j.jep.2012.12.034

Kim, S. H., et al. (2015). The dietary flavonoid Kaempferol mediates anti-inflammatory

responses via the Src, Syk, IRAK1, and IRAK4 molecular targets. Mediators Inflamm,

2015, 904142. doi:10.1155/2015/904142

Kim, Y. S., et al. (2006). Microglia, major player in the brain inflammation: their roles in the

pathogenesis of Parkinson's disease. Exp Mol Med, 38(4), 333-347.

doi:10.1038/emm.2006.40

Lee, Y. G., et al. (2009). Distinct role of spleen tyrosine kinase in the early phosphorylation of

inhibitor of kappaB alpha via activation of the phosphoinositide-3-kinase and Akt

pathways. Int J Biochem Cell Biol, 41(4), 811-821. doi:10.1016/j.biocel.2008.08.011

289

Lin, Y. C., et al. (2010). Anti-inflammatory actions of Syk inhibitors in macrophages involve

non-specific inhibition of toll-like receptors-mediated JNK signaling pathway. Mol

Immunol, 47(7-8), 1569-1578. doi:10.1016/j.molimm.2010.01.008

Lin, Y. C., et al. (2015). Syk is involved in NLRP3 inflammasome-mediated caspase-1 activation

through adaptor ASC phosphorylation and enhanced oligomerization. J Leukoc Biol,

97(5), 825-835. doi:10.1189/jlb.3HI0814-371RR

Lu, R., et al. (2012). CEACAM1 negatively regulates IL-1beta production in LPS activated

neutrophils by recruiting SHP-1 to a SYK-TLR4-CEACAM1 complex. PLoS Pathog,

8(4), e1002597. doi:10.1371/journal.ppat.1002597

Maixner, D. W., et al. (2013). The Role of Glycogen Synthase Kinase 3 Beta in

Neuroinflammation and Pain. J Pharm Pharmacol (Los Angel), 1(1), 001.

doi:10.13188/2327-204X.1000001

Martin, M., et al. (2005). Toll-like receptor-mediated cytokine production is differentially

regulated by glycogen synthase kinase 3. Nat Immunol, 6(8), 777-784.

doi:10.1038/ni1221

Miller, Y. I., et al. (2012). The SYK side of TLR4: signalling mechanisms in response to LPS

and minimally oxidized LDL. Br J Pharmacol, 167(5), 990-999. doi:10.1111/j.1476-

5381.2012.02097.x

Mocsai, A., et al. (2010). The SYK tyrosine kinase: a crucial player in diverse biological

functions. Nat Rev Immunol, 10(6), 387-402. doi:10.1038/nri2765

Moon, J. S., et al. (2015). Fatty acid synthesis and NLRP3-inflammasome. Oncotarget, 6(26),

21765-21766. doi:10.18632/oncotarget.4781

Panicker, N., et al. (2015). Fyn Kinase Regulates Microglial Neuroinflammatory Responses in

Cell Culture and Animal Models of Parkinson's Disease. J Neurosci, 35(27), 10058-

10077. doi:10.1523/jneurosci.0302-15.2015

Park, D. W., et al. (2014). GSK3beta-dependent inhibition of AMPK potentiates activation of

neutrophils and macrophages and enhances severity of acute lung injury. Am J Physiol

Lung Cell Mol Physiol, 307(10), L735-745. doi:10.1152/ajplung.00165.2014

290

Qian, L., et al. (2007). Microglia-mediated neurotoxicity is inhibited by morphine through an

opioid receptor-independent reduction of NADPH oxidase activity. J Immunol, 179(2),

1198-1209.

Qin, L., et al. (2007). Systemic LPS causes chronic neuroinflammation and progressive

neurodegeneration. Glia, 55(5), 453-462. doi:10.1002/glia.20467

Rock, R. B., et al. (2004). Role of microglia in central nervous system infections. Clin Microbiol

Rev, 17(4), 942-964, table of contents. doi:10.1128/CMR.17.4.942-964.2004

Ulanova, M., et al. (2007). Involvement of Syk protein tyrosine kinase in LPS-induced responses

in macrophages. J Endotoxin Res, 13(2), 117-125. doi:10.1177/0968051907079125

Yi, Y. S., et al. (2014). Functional roles of Syk in macrophage-mediated inflammatory responses.

Mediators Inflamm, 2014, 270302. doi:10.1155/2014/270302

Yin, H., et al. (2016). Syk negatively regulates TLR4-mediated IFNbeta and IL-10 production

and promotes inflammatory responses in dendritic cells. Biochim Biophys Acta, 1860(3),

588-598. doi:10.1016/j.bbagen.2015.12.012

Zhang, Z.-T., et al. (2016). Activation of the NLRP3 inflammasome in lipopolysaccharide-

induced mouse fatigue and its relevance to chronic fatigue syndrome. J

Neuroinflammation, 13, 71. doi:10.1186/s12974-016-0539-1

Zhu, W., et al. (2017). NLRP3 inflammasome activation contributes to long-term behavioral

alterations in mice injected with lipopolysaccharide. Neuroscience, 343, 77-84.

doi:10.1016/j.neuroscience.2016.11.037

291

CHAPTER 5. CONCLUSIONS

This chapter summarizes the results presented in the individual chapters of this dissertation

with special emphasis on the role of Spleen Tyrosine Kinase in mediating dopaminergic

neurodegeneration and microglial activation following Tweak treatment and its relevance to

pathogenesis of Parkinson Disease. The major findings of each research chapter have been detailed

in the discussion section of the pertinent chapters.

Involvement of the Mitochondria Mediated Oxidative Stress Dependent Cell Signaling

Events and Syk Tyrosine Kinase Activation in Tumor Necrosis Factor Like Weak Stimulator

of Apoptosis (TWEAK) Induced Dopaminergic Cell Death

The primary finding from the chapter 3 of the thesis is that the Spleen Tyrsoine Kinase

(SYK) and mitochondrial oxidative stress events play a significant role in TWEAK induced

dopaminergic neurodegeneration in PD. Inflammation and oxidative damage have been implicated

in the pathogenesis of Parkinson’s disease (PD); however, key signaling events that potentiate the

oxidative stress response and resultant dopaminergic neurodegeneration remains to be established.

In this study, we sought to investigate the role of TWEAK and its downstream signaling events in

dopaminergic neurotoxicity using both in vitro and in vivo experimental model of PD. Initially

using a targeted qPCR array, we identified that TWEAK (Tweak Necrosis Factor Weak Inducer

of apoptosis) is significantly up regulated in dopaminergic neuronal cells (N27 cells) in response

to an inflammatory stressor. Later, we found that there is a 2-3-fold increase in TWEAK levels in

the serum of PD patients compared to age-matched control patients as assessed via western blot

analysis. Intriguingly, serum Tweak levels have been shown to fluctuate with disease severity and

that monitoring its levels have been shown to be beneficial in managing Rheumatoid Arthritis

(RA), Multiple Sclerosis (MS) and Peritoneal inflammation (Bertin et al., 2013; Ana Belen Sanz

et al., 2014). Furthermore, time course studies performed in an MPTP mouse model of PD

292

revealed that TWEAK is upregulated as early as 3-6h post drug treatment prior to the expression

of motor deficits suggesting that TWEAK may serve as an early biomarker for PD. A recent study

showed that TWEAK protein level was increased in the striatal tissue of MPTP treated mice

implicating TWEAK/FN14 pathway in dopaminergic neurodegeneration (Mustafa et al., 2016a).

In line with this finding, our results revealed that Tweak is differentially expressed in various brain

regions including substantia nigra, cortex, striatum, hippocampus, cerebellum and olfactory bulb

in control mice which might alteast in part explain the pathological significance of TWEAK

expression and its association to PD neuropathology. These studies highlight the utility of

TWEAK as a biomarker for the early detection of PD.

To dissect the mechanism of TWEAK induced dopaminergic neurodegeneration, we

utilized the well characterized rat dopaminergic neuronal cell line (N27 cells) that has been

routinely used to study PD associated neuropathological changes (G Kanthasamy et al., 2008).

Initially, we demonstrated the expression of Tweak and its receptor Fn14 expression in N27

dopaminergic cells. In agreement with our finding, TWEAK was previously shown to be expressed

in neurons, microglia and astrocytes in Multiple Sclerosis brain (Barbara Serafini et al., 2008). To

the best of our knowledge, we for the first time demonstrate that TWEAK and its receptor is

robustly expressed in N27 cells. Furthermore, we found that Tweak induced dose-dependent

increase in apoptotic cell death in N27 dopaminergic cells. Consistent with our finding, Tweak has

been shown to elicit toxic effects in cortical neurons, renal cortical cell lines, mesangial cell lines

and mammary epithelial cell lines (Burkly et al., 2007). To further characterize the mechanisms

underlying TWEAK induced dopaminergic neurotoxicity, we systematically examined

mitochondrial apoptotic signaling events, protein degradation machinery and NF-kB pathway. We

found a significant dissipation of mitochondrial membrane potential (MMP), suppression of GSH

293

levels, caspase activation (2, 3, 8 & 9), impairment of protein degradation machinery, upregulation

of phospho-Tau, increased phospho-NF-kB P65 levels, depletion of phospho-AKT and GSK-3

beta activation in Tweak treated N27 dopaminergic neuronal cells. Moreover, our results also

revealed that these effects were accompanied by upregulation of redox sensitive kinases like

Spleen Tyrosine Kinase and PKC-delta in N27 dopaminergic neuronal cells treated with TWEAK.

Intriguingly, SN-50 (NF-kB inhibitor), Quercitin (Bioactive plant flavonoid) and R-788 (Syk

inhibitor) conferred resistance against Tweak induced neuronal cell death highlighting the

importance of NF-kB, mitochondrial stress and Syk signaling mechanisms in dopaminergic

neurodegeneration. Taken together, our results support a model whereby TWEAK exerts

dopaminergic neurotoxicity via SYK / NF-kB signaling mechanisms in a mitochondria dependent

manner.

SYK Contributes to TWEAK-Induced Inflammatory Response Through Activation of

NLRP3 Inflammasome Activation, ER Stress Response, and ALS Dysfunction in Microglia.

Emerging evidence suggests that microglial activation associated with NLRP3 activation

may be critically linked to PD associated neurodegeneration (Mao et al., 2017). Recent studies

support a role for Spleen Tyrosine Kinase (SYK) in NLRP3 inflammasome activation in immune

cells (Federica Laudisi et al., 2014; K. Neumann & Ruland, 2013). Additionally, Tweak is

implicated in the regulation of neuro-inflammatory responses in Multiple sclerosis, Stroke,

Neuropsychiatric Lupus, Glioma, and Schizophrenia (Boulamery & Desplat-Jégo, 2017).

However the key signaling events underlying Tweak induced microglial NLRP3 activation in

Parkinson’s disease remains speculative. Therefore in chapter 4, we sought to understand the role

of SYK in Tweak induced microglial activation response using an in vitro cell culture model and

neuroinflammation mouse model of PD. To dissect the mechanism of TWEAK induced microglial

NLRP3 activation, we utilized the well characterized microglial cell line (BV2 cells) that has been

294

routinely used by our lab and others for the examination of neuro-inflammatory response (Nikhil

Panicker et al., 2015). Moreover, BV2 microglial cells can be considered as an efficient substitute

to primary microglial cells as they secrete substantial amounts of cytokines and NO upon

stimulation with LPS (Stansley et al., 2012). First, we demonstrated that there is increased SYK

expression and kinase activity in BV2 microglial cells in response to TWEAK stimulation. To

further elucidate the mechanistic basis of TWEAK induced inflammatory response we performed

time course studies. BV2 microglial cells were treated with TWEAK (100 ng/ml) for increasing

duration (3-24h) and NLRP3 inflammasome related markers were assessed by Western blot and

ELISA analysis. Our immunoblot analysis revealed that there is significant upregulation of NLRP3

related markers in BV2 microglial cells upon TWEAK treatment. ELISA analysis of BV2

microglial cells treated with TWEAK revealed the upregulation of pro-inflammatory cytokines

such as IL-1 beta and TNF-alpha. To further characterize the mechanisms underlying TWEAK-

induced microglial NLRP3 inflammasome activation we examined mitochondrial function, ER

stress response and autophagolysosomal system (ALS) function. Immunoblot analysis, revealed

ER stress response, mitochondrial dysfunction, lysosomal damage, MAPK activation, GSK-3 beta

activation in TWEAK stimulated BV2 microglial cells. Accumulating evidence suggests that, SYK

induced ROS production and cathepsin-B production is responsible for NLRP3 inflammasome

activation in LPS treated neutrophils (Rongze Lu et al., 2012). Therefore, in the next set of

experiments, we proceeded to examine the impact of SYK on oxidative stress, ALS dysfunction

and NLRP3 and associated signaling events in TWEAK treated microglial cells. Upon blockade

of SYK using a pharmacological inhibitor of SYK, RX788 or siRNA mediated gene silencing, we

found that attenuation of ALS dysfunction coincided with suppression of NLRP3 inflammasome

activation, ER stress response, NOX and JNK activation in TWEAK treated cells further

295

supporting the role of SYK in the regulation of redox mechanisms. To further validate our in vitro

findings we utilized the TWEAK neuroinflammation mouse model. For this purpose, TWEAK

was stereotaxically injected into the mouse striata in the presence or absence of Piceatannol (SYK

inhibitor) to further affirm the role of SYK in NLRP3 inflammasome activation. Intriguingly, our

vivo studies demonstrated that there was no significant difference in the expression levels of SYK

or NLRP3 related markers between Tweak infused mice and control mice. Likewise, piceatannol,

a Syk inhibitor had no effect in the TWEAK neuroinflammation model. At this stage TWEAK

related dose response studies are necessary to evaluate TWEAK associated neuroinflammatory

response in the mouse brain. Collectively, our findings highlight the importance of Syk mediated

oxidative stress, ER stress, ALS dysfunction and GSK-3 beta activation in Tweak induced

microglial activation response involving NLRP3 inflammasome activation. Taken together, our

results highlight the pivotal role of SYK in mediating heightened microglial activation response

upon TWEAK treatment.

Spleen Tyrosine Kinase Mediates NLRP3 Inflammasome Activation in LPS Treated N9

Microglial Cells via NF-kB, MAP Kinase and ROS Generation.

Previous studies indicate that, functional NLRP3 inflammasome is expressed in the

microglia of mouse brain (Gustin et al., 2015). NLRP3 inflammasome activation has been

recognized as a critical contributor to dopaminergic neurodegeneration in experimental models of

Parkinson’s disease (Mao et al., 2017). ERK signaling and proteasome function have been

implicated in the priming of NLRP3 inflammasome in LPS treated monocytes (Mohammed G.

Ghonime et al., 2014). However the underlying downstream signaling events that are responsible

for the activation of NLPR3 inflammasome activation remains to be established. Therefore in

chapter 5, we sought to examine the role of SYK in NLRP3 inflammasome activation in LPS

296

stimulated N9 microglial cells. To dissect the mechanism of LPS induced microglial NLRP3

activation, we utilized well established retroviral immortalized microglial cell line (N9 cells). N9

microglial cells can be considered as an efficient substitute to primary microglial cells to study

neuro-inflammatory mechanisms as they produce cytokines like TNF-alpha, IL-1 and IL-6 upon

LPS stimulation (Righi et al., 1989; Stansley et al., 2012). First using N9 microglial cell culture

model, we demonstrated that, Spleen Tyrosine Kinase (SYK) expression is increased in LPS

treated N9 microglial cells. Additionally, LPS has been previously shown to induce NLRP3

inflamamsome activation in neutrophils and macrophages (Y.-C. Lin et al., 2015; Rongze Lu et

al., 2012). To assess whether LPS is capable of inducing NLRP3 inflammasome activation in N9

microglial cells, we stimulated N9 microglial cells with LPS and quantified the neuro-

inflammatory responses using WB analysis and Luminex analysis. Our immunoblot analysis and

luminex cytokine analysis revealed upregulation of NLRP3 inflammasome and its markers. LPS

treated macrophages has been shown to generate pro-inflammatory cytokines via activation of NF-

kB, AKT and PDK1 (Y.-S. Yi et al., 2014b). Consistent with this report immunoblot analysis

revealed ROS generation, NOS production GSK-3 beta, MAPK and NF-kB activation in LPS

stimulated microglial cells. Additionally, pharmacological inhibition of SYK was found to

attenuate microglial activation response via inhibition of JNK and NF-kB signaling pathway (Y.

C. Lin et al., 2010). Therefore, in the next set of experiments, we examined whether blockade of

SYK attenuated NLRP3 inflammasome activation and proinflammatory marker (s) expression in

LPS treated microglial cells using RX788, a SYK inhibitor. Interestingly, SYK inhibition with

RX788 suppressed LPS induced microglial NLRP3 activation coincident with suppression of ROS

and nitric oxide production, phosphorylation of NF-kB, and activation of mitogen activated protein

kinases (MAPKs), extracellular signal regulated kinases 1 and 2 (ERK1/2), p38 MAPK and

297

glycogen synthase kinase-3β (GSK3β). In conclusion, our results support a model, whereby SYK

mediates microglial activation response in LPS stimulated cells via induction of NLRP3

inflammasome activation and ROS generation as well as NF-kB, MAPK and GSk-3 beta

activation. Taken together, our results highlight the therapeutic significance of targeting SYK for

the amelioration of neuroinflammation related disorders including PD.

Bibliography

Bertin, D., et al. (2013). Is TWEAK a Biomarker for Autoimmune/Chronic Inflammatory

Diseases? Frontiers in Immunology, 4, 489. doi:10.3389/fimmu.2013.00489

Boulamery, A., et al. (2017). Regulation of Neuroinflammation: What Role for the Tumor

Necrosis Factor-Like Weak Inducer of Apoptosis/Fn14 Pathway? Frontiers in

Immunology, 8, 1534.

Burkly, L. C., et al. (2007). TWEAKing tissue remodeling by a multifunctional cytokine: Role of

TWEAK/Fn14 pathway in health and disease. Cytokine, 40(1), 1-16.

doi:https://doi.org/10.1016/j.cyto.2007.09.007

G Kanthasamy, A., et al. (2008). Environmental neurotoxin dieldrin induces apoptosis via

caspase-3-dependent proteolytic activation of protein kinase C delta (PKCdelta):

Implications for neurodegeneration in Parkinson's disease (Vol. 1).

Ghonime, M. G., et al. (2014). Inflammasome priming by LPS is dependent upon ERK signaling

and proteasome function(). Journal of immunology (Baltimore, Md. : 1950), 192(8),

3881-3888. doi:10.4049/jimmunol.1301974

Gustin, A., et al. (2015). NLRP3 Inflammasome Is Expressed and Functional in Mouse Brain

Microglia but Not in Astrocytes. PLOS ONE, 10(6), e0130624.

doi:10.1371/journal.pone.0130624

Laudisi, F., et al. (2014). Tyrosine kinases: the molecular switch for inflammasome activation.

Cellular and Molecular Immunology, 11(2), 129-131. doi:10.1038/cmi.2014.4

Lin, Y.-C., et al. (2015). Syk is involved in NLRP3 inflammasome-mediated caspase-1

activation through adaptor ASC phosphorylation and enhanced oligomerization. Journal

of Leukocyte Biology, 97(5), 825-835. doi:10.1189/jlb.3HI0814-371RR

298

Lin, Y. C., et al. (2010). Anti-inflammatory actions of Syk inhibitors in macrophages involve

non-specific inhibition of toll-like receptors-mediated JNK signaling pathway. Mol

Immunol, 47(7-8), 1569-1578. doi:10.1016/j.molimm.2010.01.008

Lu, R., et al. (2012). CEACAM1 Negatively Regulates IL-1β Production in LPS Activated

Neutrophils by Recruiting SHP-1 to a SYK-TLR4-CEACAM1 Complex. PLoS

Pathogens, 8(4), e1002597. doi:10.1371/journal.ppat.1002597

Mao, Z., et al. (2017). The NLRP3 Inflammasome is Involved in the Pathogenesis of Parkinson's

Disease in Rats. Neurochem Res, 42(4), 1104-1115. doi:10.1007/s11064-017-2185-0

Mustafa, S., et al. (2016). The role of TWEAK/Fn14 signaling in the MPTP-model of

Parkinson's disease. Neuroscience, 319, 116-122.

doi:10.1016/j.neuroscience.2016.01.034

Neumann, K., et al. (2013). Kinases conquer the inflammasomes. Nature Immunology, 14, 1207.

doi:10.1038/ni.2763

Panicker, N., et al. (2015). Fyn Kinase Regulates Microglial Neuroinflammatory Responses in

Cell Culture and Animal Models of Parkinson's Disease. The Journal of Neuroscience,

35(27), 10058-10077. doi:10.1523/jneurosci.0302-15.2015

Righi, M., et al. (1989). Monokine production by microglial cell clones. Eur J Immunol, 19(8),

1443-1448. doi:10.1002/eji.1830190815

Sanz, A. B., et al. (2014). TWEAK Promotes Peritoneal Inflammation. PLOS ONE, 9(3),

e90399. doi:10.1371/journal.pone.0090399

Serafini, B., et al. (2008). Expression of TWEAK and Its Receptor Fn14 in the Multiple Sclerosis

Brain (Vol. 67).

Stansley, B., et al. (2012). A comparative review of cell culture systems for the study of

microglial biology in Alzheimer's disease. J Neuroinflammation, 9, 115.

doi:10.1186/1742-2094-9-115

Yi, Y.-S., et al. (2014). Functional Roles of Syk in Macrophage-Mediated Inflammatory

Responses. Mediators of Inflammation, 2014, 12. doi:10.1155/2014/270302

299

CHAPTER 6. FUTURE AREAS OF INVESTIGATION

What is The Role of ER Stress Mediated CXCL10/CXCR3 Signaling Axis In The Induction

of a Proinflamamtory Response and Neuronal Injury in Experimental Models of PD?

It is well established that activated microglial can elicit beneficial effects during the early

phase of neurodegeneration in PD (Norden et al., 2015) but excessive activation of microglia can

lead to neuronal damage owing to the buildup of neurotoxic mediators including TNF-a, IL-1b,

IL-6 (Y. S. Kim et al., 2006) in the vicinity of dopaminergic neurons. Excessive glial cell

activation is a critical feature in post mortem PD brains (McGeer et al., 1988) and animal models

of PD (Qinqin Wang et al., 2015). Moreover, in a recent study using neuroinflammation rat models

of PD (LPS and 6-OHDA), activation of inflammatory mediators including NLRP3 inflammasome

was found to accompany dopaminergic neurodegeneration in the substantia nigra compacta (Mao

et al., 2017) and inhibition of caspase-1 via Ac-YVAD-CMK was found to confer resistance

against neurotoxin-induced dopaminergic neurodegenerative effect. Nevertheless, the key

mediators that regulate microglial recruitment and activation remains elusive.

Chemokines represent inflammatory mediators that regulate microglial migration and

activation subsequent to inflammatory response by binding to specific receptors expressed on

target cells. In this context, CXCR3 serves as a receptor for CXC subclass members such as CXCL

9, 10, and 11 (Lacotte et al., 2009). CXCR3 participates in the recruitment of T cells and microglia

(de Jong et al., 2008). Moreover, CXCL10 expression has been shown to be increased in several

neurodegenerative diseases including AD (Galimberti et al., 2006). Furthermore functional cross

talk between CXCL10 and CXCR3 has been shown to induce microglial activation and induce

neuronal death in several models of neurodegeneration (N. Jo et al., 2003). Having demonstrated

that TWEAK stimulation of microglial cells elicits ERS and that it is positively associated with a

heightened microglial inflammatory response it would be important to understand to what extent

300

ER stress mediates CXCL10/CXCR3 signaling axis in TWEAK treated cells. In a previous study

the expression of CXCL10 and CXCR3 was found to be increased by two fold in response to ER

stress (ERS) in a retinal injury model (Ha et al., 2015). Furthermore, retinal cell apoptosis was

found to be elevated in WT mice as compared to CXCR3-deficient mice upon ERS further

supporting the idea that CXCR3 has a critical role in ER stress induced retinal neuronal injury.

We propose to assess the activation of CXCL10/CXCR3 signaling axis in response to diverse

inflammagens including TNF alpha, LPS and TWEAK and whether its upregulation is linked to

ER stress response in BV2 cells, primary microglial cells and in an in vivo neuroinflammation

model of PD. We hypothesize that ER stress mediated activation of CXCL10/CXCR3 signaling

axis contributes to microglial migration and resultant neuronal injury in neuroinflammation models

of PD. Also, to what extent mitochondria mediated oxidative stress contributes to the activation

of CXCL10/CXCR3 signaling axis and its impact on NLRP3 inflammasome, SYK and ALS

activation will provide novel insights into the role of mitochondria in the activation of chemokine

mediated signaling events and its influence on dopaminergic neuronal wellbeing.

What is The Contribution of Mitophagy Pathway During TWEAK-Induced Microglial

Activation Response?

Mitophagy refers to the selective removal of damaged mitochondria via the

autophagolysosomal degradative machinery. Numerous studies have demonstrated a pivotal role

for mitochondrial damage and subsequent release of mitochondrial DNA and mitochondrial ROS

in the induction of NLRP3 inflammasome activation (Prajwal Gurung et al., 2015). In this context

PARKIN, a E3 ubiquitin ligase is the best known player in the mitophagy process. The PINK1 and

PARKIN pathway contributes atleast in part to the clearance of damaged mitochondria via

mitophagy. Upon mitochondrial depolarization PINK1 accumulates in the outer mitochondrial

membrane (OMM) and recruits PARKIN and activates it by phosphorylation. Activated PARKIN

301

ubiquitinates OMM which can be recognized by p62 an adapter protein which in turn recruits LC3

present on autophagosomes thereby facilitating the degradation of damaged mitochondria via

mitophagy. In a recent study Zhong et al found that p62 is increased by NF-kB signaling in

response to an inflammatory stimuli and that the p62 is recruited to the damaged mitochondria

which in turn is ubiquitinated by PARKIN thereby facilitating mitophagy. In another study

caspase-1 which is activated upon NLRP3 inflammasome activation was found to cause the

cleavage of PARKIN thereby leading to the accumulation of damaged mitochondria and

inflammasome activation via a vicious feed forward cycle (Jiujiu Yu et al., 2014). In the chapter

3 we demonstrated that TWEAK induced impairment in the autophagolysosomal system (ALS)

was associated with NLRP3 system. Therefore it would be interesting to investigate the cross talk

between PINK-PARKIN pathway and NLRP3 inflammasome activation in TWEAK-induced

heightened microglial activation response. Loss of PINK1 and PARKIN has been shown to

increase dopamine neurons susceptibility to stressors. Our studies raise the interesting possibility

as to whether dysfunctional mitophagy results in the generation of neurotoxic mediators thereby

facilitating the demise of dopaminergic neurons which are in a compromised bioenergetic state.

Therefore it would be beneficial to investigate the factors that regulate PINK-PARKIN to develop

novel therapeutics to mitigate aberrant inflammation associated disorders including PD.

Investigations Into The Role of SYK In Astrocytic Response Upon TWEAK Stimulation And

In Experimental Models of PD

Astrocytes are the most abundant type of glial cells in the brain. To date only few studies

have studied the role of astrocytes in neurodegenerative diseases including PD. Intriguingly,

several genes that have been shown to play a causative role in PD pathogenesis have been

expressed in astrocytes (Booth et al., 2017). Therefore improved understanding on their influence

in PD neuropathology might lead to the development of novel therapeutics for PD.

302

Astrocytes provide structural and metabolic support and participates in the regulation of

synaptic transmission and blood flow within the brain. Moreover, astrocytes contribute to the

maintenance of blood brain barrier (BBB) which has been shown to be impaired in PD brains

(Gray & Woulfe, 2015). In fact survival of dopaminergic neurons are dependent on astrocyte

derived neurotrophic factor namely glial derived neurotrophic factor (GDNF) (F. H. Lin et al.,

1993; Saavedra et al., 2006; X. Wu et al., 2008). Emerging evidence indicate that dopaminergic

neurodegeneration in PD may be intimately linked to astrocyte dysfunction. In this context in post

mortem PD brains DJ-1 was found to be enhanced in the astrocytes (Bandopadhyay et al., 2004;

M. Neumann et al., 2004). Moreover DJ-1 has been linked to mitochondrial function and oxidative

stress in astrocytes exposed to rotenone (Mullett & Hinkle, 2009). Therefore we would like to test

whether there is a cross talk between SYK and DJ-1 in TWEAK stimulated cells. We would be

interested in addressing the following questions: 1) Is there a functional interaction between SYK

and DJ-1 in TWEAK stimulated astroglial cells? What is its relationship to the induction of an

inflammatory response? 2) What is the impact of SYK on astroglial mitochondrial function

especially mitochondrial dynamics in DJ-1 KO astrocytes? What is the role of SYK in

Parkinsonian toxin (rotenone)-induced mitochondrial bioenergetics deficits in astrocytes and how

does this compare with TWEAK-induced astrocytosis? If, so does blockade of SYK protect

astrocytes against bioenergetics deficits induced by Parkinsonian toxins. Successful completion

of the afore mentioned studies would provide novel insights into the role of SYK in the regulation

of astrocytic function in response to diverse stimuli. Furthermore it would improve our

understanding on the contribution of non-cell autonomous signaling mechanisms in the PD

associated dopaminergic neurodegeneration, which, in turn, may lead to improved therapeutic

outcome.

303

Bibliography

Bandopadhyay, R., Kingsbury, A. E., Cookson, M. R., Reid, A. R., Evans, I. M., Hope, A. D., . .

. Lees, A. J. (2004). The expression of DJ-1 (PARK7) in normal human CNS and

idiopathic Parkinson's disease. Brain, 127(Pt 2), 420-430. doi:10.1093/brain/awh054

Booth, H. D. E., Hirst, W. D., & Wade-Martins, R. (2017). The Role of Astrocyte Dysfunction in

Parkinson’s Disease Pathogenesis. Trends in Neurosciences, 40(6), 358-370.

doi:10.1016/j.tins.2017.04.001

de Jong, E. K., de Haas, A. H., Brouwer, N., van Weering, H. R., Hensens, M., Bechmann, I., . . .

Biber, K. (2008). Expression of CXCL4 in microglia in vitro and in vivo and its possible

signaling through CXCR3. J Neurochem, 105(5), 1726-1736. doi:10.1111/j.1471-

4159.2008.05267.x

F. H. Lin, L., Doherty, D. H., Lile, J., and Bektesh, S., & Collins, F. (1993). GDNF: a Glial cell

line-derived Neurotrophic Factor for midbrain dopaminergic neurons (Vol. 260).

Galimberti, D., Schoonenboom, N., Scheltens, P., & et al. (2006). Intrathecal chemokine

synthesis in mild cognitive impairment and alzheimer disease. Archives of Neurology,

63(4), 538-543. doi:10.1001/archneur.63.4.538

Gray, M. T., & Woulfe, J. M. (2015). Striatal Blood–Brain Barrier Permeability in Parkinson'S

Disease. Journal of Cerebral Blood Flow & Metabolism, 35(5), 747-750.

doi:10.1038/jcbfm.2015.32

Gurung, P., Lukens, J. R., & Kanneganti, T.-D. (2015). Mitochondria: Diversity in the regulation

of NLRP3 inflammasome. Trends in molecular medicine, 21(3), 193-201.

doi:10.1016/j.molmed.2014.11.008

Ha, Y., Liu, H., Xu, Z., Yokota, H., Narayanan, S. P., Lemtalsi, T., . . . Zhang, W. (2015).

Endoplasmic reticulum stress-regulated CXCR3 pathway mediates inflammation and

neuronal injury in acute glaucoma. Cell Death &Amp; Disease, 6, e1900.

doi:10.1038/cddis.2015.281

https://www.nature.com/articles/cddis2015281#supplementary-information

304

Jo, N., Wu, G.-S., & Rao, N. A. (2003). Upregulation of Chemokine Expression in the Retinal

Vasculature in Ischemia–Reperfusion Injury. Investigative Ophthalmology & Visual

Science, 44(9), 4054-4060. doi:10.1167/iovs.02-1308

Kim, Y. S., & Joh, T. H. (2006). Microglia, major player in the brain inflammation: their roles in

the pathogenesis of Parkinson's disease. Exp Mol Med, 38(4), 333-347.

doi:10.1038/emm.2006.40

Lacotte, S., Brun, S., Muller, S., & Dumortier, H. (2009). CXCR3, inflammation, and

autoimmune diseases. Ann N Y Acad Sci, 1173, 310-317. doi:10.1111/j.1749-

6632.2009.04813.x

Mao, Z., Liu, C., Ji, S., Yang, Q., Ye, H., Han, H., & Xue, Z. (2017). The NLRP3 Inflammasome

is Involved in the Pathogenesis of Parkinson's Disease in Rats. Neurochem Res, 42(4),

1104-1115. doi:10.1007/s11064-017-2185-0

McGeer, P. L., Itagaki, S., Boyes, B. E., & McGeer, E. G. (1988). Reactive microglia are

positive for HLA-DR in the substantia nigra of Parkinson's and Alzheimer's disease

brains. Neurology, 38(8), 1285-1291.

Mullett, S. J., & Hinkle, D. A. (2009). DJ-1 knock-down in astrocytes impairs astrocyte-

mediated neuroprotection against rotenone. Neurobiol Dis, 33(1), 28-36.

doi:10.1016/j.nbd.2008.09.013

Neumann, M., Muller, V., Gorner, K., Kretzschmar, H. A., Haass, C., & Kahle, P. J. (2004).

Pathological properties of the Parkinson's disease-associated protein DJ-1 in alpha-

synucleinopathies and tauopathies: relevance for multiple system atrophy and Pick's

disease. Acta Neuropathol, 107(6), 489-496. doi:10.1007/s00401-004-0834-2

Norden, D. M., Muccigrosso, M. M., & Godbout, J. P. (2015). Microglial priming and enhanced

reactivity to secondary insult in aging, and traumatic CNS injury, and neurodegenerative

disease. Neuropharmacology, 96(Pt A), 29-41. doi:10.1016/j.neuropharm.2014.10.028

Saavedra, A., Baltazar, G., Santos, P., Carvalho, C. M., & Duarte, E. P. (2006). Selective injury

to dopaminergic neurons up-regulates GDNF in substantia nigra postnatal cell cultures:

role of neuron-glia crosstalk. Neurobiol Dis, 23(3), 533-542.

doi:10.1016/j.nbd.2006.04.008

305

Wang, Q., Liu, Y., & Zhou, J. (2015). Neuroinflammation in Parkinson’s disease and its

potential as therapeutic target. Translational Neurodegeneration, 4, 19.

doi:10.1186/s40035-015-0042-0

Wu, X., Chen, P. S., Dallas, S., Wilson, B., Block, M. L., Wang, C.-C., . . . Hong, J.-S. (2008).

Histone deacetylase inhibitors up-regulate astrocyte GDNF and BDNF gene transcription

and protect dopaminergic neurons. The international journal of

neuropsychopharmacology / official scientific journal of the Collegium Internationale

Neuropsychopharmacologicum (CINP), 11(8), 1123-1134.

doi:10.1017/S1461145708009024

Yu, J., Nagasu, H., Murakami, T., Hoang, H., Broderick, L., Hoffman, H. M., & Horng, T.

(2014). Inflammasome activation leads to Caspase-1–dependent mitochondrial damage

and block of mitophagy. Proceedings of the National Academy of Sciences, 111(43),

15514-15519. doi:10.1073/pnas.1414859111

306

ACKNOWLEDGMENTS

I would like to whole-heartedly thank my major Professor Dr. Arthi

Kanthasamy and Co-Major Professor Dr Anumantha Kanthasamy, for providing me of this

opportunity for pursuing my course work and research projects in their labs and supporting me

entirely during the Biomedical Sciences PHD graduate program. Their persistant encouragement,

assistance, guidance and optimism is the main reason to my successful completion of graduate

program and research projects. During the whole period of my graduate school in Iowa state, I had

developed skills so design research projects, perform experimental procedures, apply scientific

knowledge and investigate important research questions regarding pathophysiology of

neurological diseases. I would also like to appreciate and be very grateful of my committee

members Dr. Bellaire Bryan, Dr. Manju Reddy, Dr. Wilson Rumbeiha and Dr. Jesse Goff for their

constant support, encouragement, guidance regarding my research projects of the graduate PHD

program. Furthermore, I would like to extend my gratitude and acknowledgement to Dr

Anantharam Vellareddy for helping in ordering resources and animals for my research studies in

the lab. Additionally, I would like to thank the current lab members, Vivek Lawana and Neeraj

Singh for providing me great help in my experiments and I really enjoyed working with them as a

team. Also, I would like to thankful for my previous lab members- Drs. Huajun Jin, Richard

Gordon, Afeseh Ngwa Hilary and Anamitra Ghosh who helped me learning the research

techniques and helped me to adjusted to the lab. Also I would like to extend my gratitude to the

Department of Biomedical Sciences Staff – Loonan Rochelle, Kim Adams, William B. Robertson,

Amy Brucker and Hashman Emma for helping in the administrative tasks and duties during my

period of the PHD program.

307

Finally, I would like to thank my loved ones - my grandparents, my parents, my wife, my

family and my friends for their persistant unconditional support and constant encouragement,

without which it would have been near impossible to finish this arduous task of complete PHD

graduate studies in Iowa state.