interactome analysis identifies novel targets of

109
Interactome Analysis Identifies Novel Targets of Phosphoinositide 3-kinase (PI3K) that Mediate Astrocyte Neuroprotection by Samih Ahmad Alqawlaq A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Laboratory Medicine and Pathobiology University of Toronto © Copyright by Samih Ahmad Alqawlaq 2018

Upload: others

Post on 29-Jan-2022

12 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Interactome Analysis Identifies Novel Targets of

Interactome Analysis Identifies Novel Targets of Phosphoinositide 3-kinase (PI3K) that Mediate

Astrocyte Neuroprotection

by

Samih Ahmad Alqawlaq

A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy

Laboratory Medicine and Pathobiology University of Toronto

© Copyright by Samih Ahmad Alqawlaq 2018

Page 2: Interactome Analysis Identifies Novel Targets of

ii

Interactome Analysis Identifies RNA Binding Protein ZC3H14 as a Novel Interactor of Phosphoinositide 3-

kinase (PI3K) and a Mediator of Astrocyte Neuroprotection

Samih Ahmad Alqawlaq

Doctor of Philosophy

Laboratory Medicine and Pathobiology University of Toronto

2019

Abstract

In a homeostatic state, astrocytes can support neuronal survival and function through a

range of secreted signals that protect against neurotoxicity, oxidative stress, and

apoptotic cascades. Thus, the analysis of astrocyte conditioned media (ACM) may

provide valuable insight into the nature of these protective mechanisms, and how they

might be promoted. Previously, we characterized a potent neuroprotective activity

mediated by ACM in neurons and the retina in metabolic stress models. However, the

molecular entity and mechanism underlying this activity remained unclear. Here, a

chemical genetics screen revealed phosphoinositide 3-kinase (PI3K) as a central player

transducing ACM-mediated neuroprotection. To identify additional proteins contributing

Page 3: Interactome Analysis Identifies Novel Targets of

iii

to the protective activity, endogenous neuronal PI3K was immunoprecipitated from

astrocytes exposed to ACM or control media, and MS/MS analyses were undertaken.

MS data analyses pointed toward only five additional proteins that co-

immunoprecipitated with PI3K and were regulated by the ACM signal. These hits

included expected PI3K interactors, such as the platelet-derived growth factor receptor

A (PDGFRA), and novel interactors, such as the zinc finger CCCH-type containing 14

(ZC3H14). ZC3H14 has recently emerged as an important RNA binding protein that

modifies poly-adenosine tail lengths on nascent mRNA transcripts. In downstream

validation studies we show that Platelet Derived Growth Factor-BB (PDGF-BB) strongly

activates PI3K signaling to protect neuronal cells. Finally, PDGF-BB treatment induced

recruitment of ZC3H14 to PI3K, and inhibiting this interaction eliminated ACM-mediated

neuroprotection. Thus, we identified a novel ACM- and PDGF-induced neuroprotective

signaling cascade mediated through PI3K that involves recruitment of ZC3H14.

Enhancing this pathway may present a promising strategy to promote astrocyte-

secreted neuroprotective signals.

Page 4: Interactome Analysis Identifies Novel Targets of

iv

Dedication

So many of my loved ones are deserving of this dedication that I feel I am responsible

for only a minute fraction of this thesis. My grandmothers and grandfathers have

dedicated their entire being to educate my parents against all the social, economic, and

political odds. In a time where many parents sent their children to work at young age to

make a living in Palestine and Egypt, my grandparents fought with their hearts to send

my parents to university. My parents, Samia Ibrahim and Ahmad Al Qawlaq carried the

torch forward and lit my life with love, wisdom, and patience. Without them, this thesis

would not be. In all truth however, my sacrifices, and those of my parents and

grandparents were to carry our vision and identity into the future, in the hands of my

daughter Leen. Habibti Leen, on behalf of your grandparents, your mom, and whole

family, I dedicate this thesis to you. Carry it forward.

Page 5: Interactome Analysis Identifies Novel Targets of

v

Acknowledgments

Throughout this five-year journey, I was inspired by so many brilliant minds and hearts.

Way before that, so many have laid down the foundation of this work, brick by brick. I

am eternally grateful for my academic team, my family, and my friends who kept their

doors open for me whenever I needed them. Without a doubt, I will fail to acknowledge

some of you, but I want you to know that I am eternally grateful for your contribution,

despite my limited memory. In the next few pages, I hope I will pay a small tribute to

those who made a difference in this work, directly and indirectly.

This work was primarily driven by the patient and kind mentoring of my project

supervisor Dr. Jeremy Sivak, who provided endless support at all stages of the project.

Dr. Sivak, there were many moments along the way where the challenges of the project

seemed overwhelming; I often shared with my colleagues that if I was supervised by

any other supervisor in the department I would not be successful. Thank you for being

so understanding and supportive, while being insightful and helpful.

I would like to acknowledge the contributions of my graduate committee advisors, Dr.

James Eubanks and Dr. Gerold Schmitt-Ulms, who provided essential feedback and

direction throughout the program. Dr. Schmitt-Ulms, you taught me excellent standards

of scientific research with your precise and thorough approach to experimental design.

Dr. Eubanks, your vast knowledge in Neuroscience was very assuring to have onboard

the committee, and it challenged me to explore the deep intricacies of my project.

Declan Williams of the Schmitt-Ulms group, I am grateful for your expert input in the

interactome study, the main pillar of this project. My friend and colleague Mohadeseh

Mehrabian was also incredibly helpful in troubleshooting the interactome study, offering

Page 6: Interactome Analysis Identifies Novel Targets of

vi

ingenious tips and recommendations throughout the study. Dr. Anita Corbett and her

associate Sara Leung generously provided reagents and expertise on this project that

pushed us through a challenging bottleneck. Furthermore, Dr. Jeff Wrana and Dr.

Alessandro Datti of the The High-Throughput Screening Division provided the expertise

and facilities to carry out the chemical genetics screen, which kicked off this project. I

am grateful to my colleagues at the Sivak lab: Dr. Izhar Livne-Bar, Darren Chan, Cindy

Guo, Alessandra Tuccito, and Nevena Vicic who provided essential technical and

morale support throughout the years. Thank you for chats, laughs, and the venting

sessions, could not have made it without those.

My dear family, you are the foundation and the roots of this success, thank you for

teaching me to love life and to respect science. My dad, Ahmad Al Qawlaq and mom

Samia Ibrahim, thank you for always seeing me in a better way than I can see myself.

Mom, I can write forever about how a magnificent soul you are; I can only say that your

spirit and generosity can never be matched by another being. Dad, you are the best

fighter I know, you taught me that giving up is never an option. Thank you for giving me

the bows and arrows to survive the jungle of life. My dearest wife Ayat, I often came

home from the lab with multiple failed experiments and a broken heart. At times, I could

not see hope. Your love and faith in me were the only things that made me get out of

bed the next day. I always told you this is your PhD, and I say it here again. Leen, thank

you for keeping me up all night and making me forgive you the moment I see you in the

morning. Enas, Ayman, Ehab, Eyad and Yazan Alqawalq, I always sensed how much

faith you had in me- that kept me going. You supported me during my ailments and joys

and I cannot ask for better friends.

Page 7: Interactome Analysis Identifies Novel Targets of

vii

My Krembil friends Yuriy Baglaenko, Nardos Tassew, Ahmed Labban, and Elena

Darmos, thank you for the many therapeutic chats, laughs, and fun outings. You have

certainly brightened my days at Krembil. Chilbert Dong, my old friend, thank you for all

of the visits, your sense of humor and kindness empowered me to carry on. Hsain Al

Shihabi and Bashar Jabbour, thank you for your love and support over the years and for

believing in me. I have learned so much from your compassion and courage. Hsain, I

will never forget that night you let me stay over at your place while Leen was at the

hospital; without you, I would not have been able to finish my PhD. Eric Lee, thank you

for all the fancy dinners and brunches; you have certainly elevated my taste in

restaurants and coffee shops.

My dear friends at the Haigue: Mark, Mel, Scott Zoltok, Scott Baker, Lexi, Melyssa, and

Jamie, thank you from the bottom of my heart. You have made a beautiful home for me

to land in in Toronto in my first year, my toughest year. You have taught me so much

about living in a community and about friendship, I am truly grateful you came into my

path. It was so hard to leave the Haigue, but I am so fortunate we stayed in touch.

And for anyone else I missed: those who smiled, those who kept the door open behind

them, and those who offered a gesture of encouragement of any sort, thank you. Your

name is on this work.

Page 8: Interactome Analysis Identifies Novel Targets of

viii

Table of Contents

Dedication ...................................................................................................................................... iv

Acknowledgments ........................................................................................................................... v

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

List of Tables .................................................................................................................................. x

List of Figures ................................................................................................................................ xi

Introduction ................................................................................................................................ 2

1.1 Glaucoma ............................................................................................................................ 2

1.2 Mechanisms of glaucomatous neurodegeneration .............................................................. 3

1.2.1 Astrocyte dysfunction ............................................................................................. 4

1.2.2 NF deprivation ...................................................................................................... 12

1.2.3 Metabolic and Oxidative stress ............................................................................. 14

1.3 Phosphatidylinositol 3-kinase ........................................................................................... 18

1.3.1 PI3K role in promoting cell survival .................................................................... 19

1.3.2 PI3K interaction with RNA binding proteins ....................................................... 20

1.3.3 Poly-Adenosine RNA binding RBP ...................................................................... 23

Rationale, Hypothesis and Aims .............................................................................................. 38

2.1 Rationale ........................................................................................................................... 38

2.2 Global Hypothesis ............................................................................................................. 43

2.3 Purpose and aims .............................................................................................................. 43

Interactome Analysis Identifies RNA Binding Protein ZC3H14 as a Novel Interactor of

Phosphoinositide 3-kinase (PI3K) and a Mediator of Astrocyte Neuroprotection .................. 47

3.1 Abstract ............................................................................................................................. 47

3.2 Introduction ....................................................................................................................... 48

Page 9: Interactome Analysis Identifies Novel Targets of

ix

3.3 Methods ............................................................................................................................. 49

3.3.1 Cell cultures .......................................................................................................... 49

3.3.2 PI3K inhibitor and PDGF ..................................................................................... 50

3.3.3 Kinase inhibitor library screening ......................................................................... 51

3.3.4 Cell Viability Assay .............................................................................................. 52

3.3.5 Immunoprecipitations ........................................................................................... 52

3.3.6 Mass spectroscopy ................................................................................................ 53

3.3.7 Protein identification and analyses ....................................................................... 54

3.3.8 Immunoblotting ..................................................................................................... 55

3.3.9 SiRNA transfection ............................................................................................... 56

3.3.10 Statistics ................................................................................................................ 56

3.4 Results ............................................................................................................................... 57

3.4.1 ACM neuroprotection is mediated through PI3K ................................................. 57

3.4.2 PI3K immunoprecipitation .................................................................................... 60

3.4.3 ACM induced interactome identification .............................................................. 62

3.4.4 ZC3H14 complexes with PI3k .............................................................................. 68

3.4.5 PDGF induces neuroprotective PI3K recruitment of ZC3H14 ............................. 70

3.5 Discussion ......................................................................................................................... 72

3.6 ACKNOWLEDGEMENTS .............................................................................................. 75

Conclusions and future directions ............................................................................................ 88

4.1 RBPs in the context of neurodegeneration ........................................................................ 88

4.2 ZC3H14 is essential to the stability of metabolic transcripts ........................................... 89

4.3 PDGFr as a key receptor in ACM-mediated neuroprotection .......................................... 91

4.4 Considerations for future direction ................................................................................... 92

Page 10: Interactome Analysis Identifies Novel Targets of

x

List of Tables

Table 1. Top 20 PI3K interactome hits, sorted by spectral counts in 6 samples ............. 63

Table 2 List of ACM-induced PI3K interactors and corresponding CFM:ACM fold

changes ....................................................................................................................................... 67

Supplementary Table 1. A list of PI3K antibodies used in immunoprecipitation screen,

along with molecular weight of target peptide, and corresponding IgG isotype

…………………………………………………………………………………………………...80

Supplementary Table 2. Full list of proteins captured in the PI3K interactome ranked by

PSM……………………………………………………………………………………………..81

Page 11: Interactome Analysis Identifies Novel Targets of

xi

List of Figures

Figure 1-1. Astrocytes characterization .................................................................................... 6

Figure 1-2. ACM or CFM was injected intravitreally 1 day prior to KA challenge. ............. 8

Figure 1-3. Characterization of LXA and LXB activity in vitro ............................................. 10

Figure 1-4 A Schematic of PI3K/AKT pathway and key targets. ........................................ 20

Figure 1-5. A schematic of the mechanism of action of the RBP KSRP in response to

NF-initiated AKT phosphorylation. ........................................................................................... 22

Figure 2-1. Characterization of primary retinal astrocytes ................................................... 39

Figure 2-2. Assessing ACM neuroprotection against metabolic stress in Ht22 cells ...... 40

Figure 3-1. PI3K is necessary for ACM-mediated neuroprotection. .................................. 59

Figure 3-2. A PI3K interactome yields highly specific capture of ACM induced proteins.

....................................................................................................................................................... 61

Figure 3-3. A summary of ACM-regulated PI3K interactions. ............................................. 65

Figure 3-4. ZC3H14 is a novel PI3K interactor. .................................................................... 69

Figure 3-5. PDGF is enriched in ACM and induces PI3K recruitment of ZC3H14. ........ 71

Figure 3-6. Summary of the mechanisms of ACM neuroprotection against metabolic

stress. ........................................................................................................................................... 74

Figure 4-1. Immunostaining of ZC3H14, and AKT in retinal sections treated with vehicle and

Kianic acid (1hr)……………………………………………………………………………………… 94

Page 12: Interactome Analysis Identifies Novel Targets of

1

Chapter 1: Literature Review

(Note that this chapter incorporates material published in the following articles [1, 2])

Page 13: Interactome Analysis Identifies Novel Targets of

2

Introduction

Neurodegenerative diseases of the central nervous system and the retina share a range

of pathological features and disease mechanisms [3, 4]. One example that is central

among these diseases is altered astrocyte-neuron interactions [5-8]. Thus, identifying

models that enable the study of astrocyte-neuron interactions is essential to

understanding neurodegenerative processes and developing efficacious treatment

approaches. The retina is an excellent experimental model to study astrocyte-neuron

interactions for several reasons. Firstly, the retina has a simple anatomy and is highly

accessible compared to the brain. Secondly, astrocytes and neurons reside side-by-side

in the nerve fibre layer (NFL) of the inner retina, allowing direct investigation into their

communication. Thus, studying retinal diseases is an advantageous approach to

understanding astrocyte-neuron interactions in the neurodegenerative context. The

current chapter provides an overview of Glaucoma as a model neurodegenerative

model of astrocyte-neuron interactions in the context of neurodegeneration.

1.1 Glaucoma

Glaucoma is a blinding optic nerve neuropathy that is expected to affect 76 million

people worldwide by 2020 [9, 10]. In this sense, glaucoma is the most common

neurodegenerative disease, sharing pathological features throughout the central

nervous system [4]. An integrated network of pathological mechanisms has been

attributed to glaucomatous neurodegeneration, including astrocyte reactivity, loss of

neurotrophic support, and metabolic/oxidative stress. Importantly, these common

Page 14: Interactome Analysis Identifies Novel Targets of

3

pathological cascades can be initiated by individual, or combinations of, trigger factors.

The current framework argues that these cascades are a good target of therapeutic

intervention, since they are common to a number of triggers, as will be described in the

next part of this review. Some key cascades, investigated in detail by our laboratories’

and others, are also discussed in the following sections.

Glaucoma predispositions include age (>55 years), ethnicity, lifestyle, and family history

[11-13]. These predispositions place patients at higher risk of developing other

glaucoma risk factors including poor ocular perfusion, deleterious biomechanical

properties of the sclera and cornea, and low central corneal thickness [14-22]. Further,

glaucoma is often observed alongside a number of comorbidities, such as low mean

arterial blood pressure, diabetes, immune disease, and age-related vascular

degeneration [23-27]. While the etiology of glaucoma subtypes may vary greatly, they

share a common characteristic degeneration of retinal ganglion cells (RGCs) and their

axons, accompanied by remodeling of the lamina cribrosa of the optic nerve head

(ONH) [28-30]. This so-called cupping of the ONH is a hallmark of glaucoma, and it

occurs due to a combination of nerve fibre degeneration [31-33], along with their

support network of glial cells, and significant remodeling of extracellular matrix (ECM)

[34, 35].

1.2 Mechanisms of glaucomatous neurodegeneration

Research into the intricate mechanisms involved in triggering the pathological chain,

and the resulting molecular cascades is laying the foundation for future effective

Page 15: Interactome Analysis Identifies Novel Targets of

4

treatments for glaucoma. In particular, targeting ‘cascade’ events might provide

conserved targets for therapeutic intervention of neurodegenerative mechanisms that

are not limited to specific risk factors. While these cascades progress to a common

apoptotic pathway, they provide a larger window for intervention than anti-apoptotic

approaches, which may only target those cells in the process of dying.

An integrated network of pathological mechanisms has been attributed to glaucomatous

neurodegeneration, including astrocyte reactivity, loss of neurotrophic support, and

metabolic/oxidative stress. Importantly, these common pathological cascades can be

initiated by individual, or combinations of, trigger factors. Some key cascades,

investigated in detail by our laboratories’ and others, are also discussed in the following

sections.

1.2.1 Astrocyte dysfunction

1.2.1.1 Homeostatic roles of Astrocytes

Astrocytes make up approximately 50% of the non-mylenated human ONH, and interact

closely with RGCs in the NFL (Figure 1-1A) and axonal bundles (Figure 1-1B) in the LC.

Their abundance and proximity to RGCs make their dysfunction in glaucoma of

particular importance [36]. Under physiological conditions, astrocytes and related Müller

glia support RGCs and their axons, mainly by providing metabolic and mechanical

support, vascular regulation, neurotrophic support, and ECM maintenance, in addition to

regulating ionic and neurochemical homeostasis, particularly against glutamate and

Page 16: Interactome Analysis Identifies Novel Targets of

5

reaction oxygen species (ROS) [37-39]. In addition, they secrete a range of

neurotrophic factors (NFs) [40, 41].

Page 17: Interactome Analysis Identifies Novel Targets of

6

A B

Figure 1-1. Astrocytes characterization

(A) Astrocyte fibers (green, stained for GFAP) closely interact with neighboring RGC bodies (red, stained

for RPBMS) in the inner retina. (B) Astrocyte fibers (green, stained for GFAP) outline axonal bundles in

the ONH (red, stained for -tub). Adopted with permission from Alqawlaq,et al, 2018 [1].

Recently, our group has identified the dysregulation of astrocyte-secreted pro-resolving

lipid mediators, the lipoxins, as a contributor to inner retinal injury. Lipoxins, including

LXA and LXB, were shown to be potently neuroprotective of RGCs in vitro, and in

chronic and acute inner retinal injury and glaucoma models [2]. The protective activity

produced by transplanted retinal astrocytes could be driven by endogenous detoxifying

mechanisms or secreted signals. To distinguish these possibilities, we collected

Astrocyte conditioned media (ACM) and tested whether it was sufficient to provide

protection. Either ACM or cell-free control media incubated under identical conditions

was injected intravitreally into C57BL/6 mice 24 hours prior to Kainic Acid (KA)

challenge. RGC survival was specifically assessed by probing for the marker RBPMS,

along with complementary TUNEL staining and quantification. Significant rescue of

RGCs was observed in eyes injected with ACM compared with control media (Figure 1-

2A and B), while TUNEL staining showed that ACM injection strongly reduced GCL

Page 18: Interactome Analysis Identifies Novel Targets of

7

death (Figure 1-2C and D). To facilitate further study, we established an in vitro assay in

which ACM was applied to the neuronal cell line HT22. Consistent with the in vivo

results, ACM treatment produced significant HT22 cell protection from glutamate

challenge (Figure 1-2E). These data established a platform for future studies of the

ACM activity. Surprisingly, in preliminary fractionation experiments, we discovered that

a substantial portion of protective activity was contained in filtrate smaller than 3 kDa,

indicating the involvement of a small molecule (Figure 1-2F). In order to identify small

molecules in the ACM that might account for the protective activity, we performed

metabolomic screening, using liquid chromatography–tandem mass spectrometry (LC-

MS/MS). Lipid mediators were quantified to generate a lipidomic profile. Lipidomic

analyses revealed significant enrichment of the lipoxins LXA4 and LXB4 in ACM

compared with control media (Figure 1-2G).

Page 19: Interactome Analysis Identifies Novel Targets of

8

Figure 1-2. ACM or CFM was injected intravitreally 1 day prior to KA challenge.

(A) ACM treatment reduced KA-induced RGC loss compared with cell-free control media (RBPMS: green,

arrows). (B) Quantification of RBPMS results showing significant RGC survival with ACM injection (n = 5).

(C) Complimentary results showing ACM-mediated reduction in TUNEL-labeled cells (green, arrows)

compared with control media. (D) Quantification of TUNEL results showing significant decrease in

ganglion cell layer apoptosis from ACM media (n = 5). (E) ACM protection was reproduced in vitro with

HT22 neuronal cells challenged with 5 mM glutamate (n = 3). (F) Substantial protective activity in ACM is

contained in a 3-kDa filtrate (n = 3). (G) High concentrations of LXA4 and LXB4 were detected in ACM

compared with control media by LC-MS/MS (n = 3). Scale bar: 50 μm. *P < 0.05; **P < 0.01; ***P < 0.005.

Bars represent SEM. Statistical analyses were performed by 1-way ANOVA with TUKEY post-hoc test.

Adopted with permission from Livne-Bar et al, 2017 [2].

Page 20: Interactome Analysis Identifies Novel Targets of

9

To determine whether the protective activity we observed was direct, we treated

neuronal cells with LXA4 or LXB4 in our established in vitro assay. Increasing

concentrations of LXA4 or LXB4 produced increased viability of glutamate-challenged

HT22 cells (Figure 1-3A). Consistent with our in vivo results, LXB4 appeared more

potent than LXA4, with significant activity at 50 nM versus 500 nM, respectively (Figure

1-3A). In contrast, neither the lipoxin precursor 15-HETE nor the structurally related

trihydroxy SPM RvD2 had any protective activity up to 1 μM (Figure 1-3B). There is no

established receptor for LXB4. However, LXA4 and RvD2 signal via the ALX/FPR2 and

GPR18 receptors [42], respectively. Therefore, we assessed whether ALX/FPR2 or

GPR18 antagonists could block LXB4-mediated protection. Increasing concentrations of

WRW4 (ALX/FPR2 antagonist; IC50 = 0.23 μM) or O-1918 (GPR18 antagonist; IC50 =

5.3 μM) did not block LXB4-protective activity (Figure 1-3C). To further clarify the

LXB4 effect on cell viability, we also assessed mitochondrial membrane potential.

LXB4 treatment had no effect on membrane potential alone, but strongly protected

against glutamate-induced activity compared with vehicle (Figure 1-3D). These data

suggest that LXB4 neuroprotection is mediated through a distinct signaling mechanism

influencing mitochondrial activity independently of LXA4 or RvD2.

Page 21: Interactome Analysis Identifies Novel Targets of

10

Figure 1-3. Characterization of LXA and LXB activity in vitro

(A) Treatment of HT22 neuronal cells with LXA4 or LXB4 significantly protected them from metabolic

insult in a dose-dependent manner up to 500 nM (n = 3). (B) No protective activity was observed by

treatment with up to 1 μM of the related molecules 15-HETE or RvD2 (n = 3). (C) LXB4 protective activity

at 500 nM was not blocked by treatment with increasing μM concentrations of WRW4 or the GPR18

antagonist O-1918 (n = 3). Adopted with permission from Livne-Bar et al, 2017 [2].

1.2.1.2 Characteristics of astrocyte reactivity

Following glaucomatous injury, a parainflammatory switch occurs in astrocyte function

leading to complex positive and negative non cell-autonomous effects on neighboring

RGCs and their axons. Astrocyte reactivity is characterized by a number of

morphological and functional changes that mainly include 1) an increase in type III

intermediate filament expression, namely glial fibrillary acidic protein (GFAP) and

Page 22: Interactome Analysis Identifies Novel Targets of

11

vimentin 2) hypertrophy, migration and invasion of axonal bundles, 3) changes in

expression of cell surface and ECM-related protein [43] 4) altered buffering capacity of

key neurochemicals, namely glutamate, as a result of reduced glutamine synthetase

(GS), 5) excess production of pro-inflammatory cytokines [44, 45], and increased

production of antioxidant enzymes [46, 47], particularly in the production of essential

precursors for the CNS peptide glutathione [48]. However, these changes do not occur

simultaneously; instead, each stage of the reactive cascade has its unique features [49].

For example, evidence suggests that early gene expression changes in the ONH mainly

affect inflammation and immune regulation, while late stage changes involve scar

formation and debris cleaning [50]. Further, astrocyte reactivity varies in duration; in

mice, it can be completely reversed 6 weeks post mild intraocular pressure (IOP)

elevation (30mmHg) for one hour, with no evidence of neurodegeneration [51].

While many examples of reactivity appear to mediate deleterious effects, emerging

evidence suggests positive consequences can also be induced [52]. These positive

responses provide another treatment opportunity, whereby astrocyte protective roles

can be harnessed and promoted to protect neurons. For example, the formation of glial

scar tissue following injury had been thought to prevent neuronal regeneration, but can

also promote regeneration given the presence of the appropriate growth factors [53].

Similarly, attenuating astrocyte hypertrophy and reactive remodeling, by knocking out

the transcription factor STAT3 led to increased RGC loss in an experimental glaucoma

and nerve crush [54]. From a more global perspective, genomic analyses of CNS

astrocyte responses by Barres et al have led to the identification of distinct induced

neurotoxic and neuroprotective astrocyte populations, respectively [8, 49]. These

Page 23: Interactome Analysis Identifies Novel Targets of

12

studies also showed that activated microglia influence astrocyte reactive cascades,

indicating a complex interactive environment directs the reactive process that has only

begun to be elucidated. Thus, depending on the degree of neuronal injury, the

vulnerability of the ONH to glaucomatous damage, and the caliber of the immune

system, glial cell roles range from protective to neurodegenerative [55-57].

Taken together, these data suggest that glial reactivity is not inherently harmful, but

might be managed to promote beneficial effects. Thus, there are growing reports of so-

called ‘glioprotective’ strategies that have meaningful neuroprotective outcomes [58-60].

Also, several standard IOP lowering drugs, including Latanoprost, have been shown to

lower GFAP immunoreactivity in an IOP rat model, although the causal relationship

behind this effect remains unclear [61]. A recent study published by our group utilized

the small molecule Withaferin A to attenuate astrocyte and Müller glial reactivity in vivo,

by inhibiting GFAP and vimentin polymerization. This inhibition blocked p38 mitogen

activated protein kinase (MAPK)-dependent secretion of TNFα, resulting in dramatically

reduced neuronal apoptosis [59]. Similarly, Minocycline was shown to reduce microglial

activation and improve RGC axonal transport and survival in a chronic IOP model [62].

Additional dissection of the mechanisms regulating positive and negative aspects of the

reactive cascade may generate new therapeutic opportunities.

1.2.2 NF deprivation

Neurotrophic deprivation is a well-established mechanism of glaucomatous

degeneration [63, 64]. In experimental glaucoma models RGCs and their axons lose

Page 24: Interactome Analysis Identifies Novel Targets of

13

access to key pro-survival NFs, and in contrast, show prolonged survival in response to

supplemental NFs such as brain drived neurotrophic factor (BDNF) [65-67], Nerve

growth factors (NGF) [68], neurotrophin-4/5 (NT-4/5) [69], Ciliary Neurotrophic factor

(CNTF) [70], and Glial-cell line derived neurotropic factor (GDNF) [71]. Combining NFs

generally mediates better neuroprotection outcomes in experimental models, compared

to the use of a single NF [72, 73]. However, one common finding in these studies is that

individuzal NF therapy alone has not been supported as a long-term treatment

approach for glaucoma. This is mainly due to the downregulation of tyrosine receptor

kinases (TRKB) following injury, which desensitized neurons to NFs [74]. Further, NF

therapy does not compensate for the shortage of a range of other pro-survival factors

needed for normal neuronal function.

RGCs receive NF support from two main sources: 1) axonal targets, such as the

superior colliculus, whereby NFs are transported through axons by retrograde transport,

and 2) from local glia, primarily microglia, astrocytes and Müller cells [75, 76]. In

glaucoma, the interruption of NF supply to neurons can occur from both of these

sources. First, interrupted anterograde and retrograde transport of NFs between RGCs

and their targets has been demonstrated in experimental models, by elevated IOP or

lowered CSFP [77, 78]. The pressure gradient has been shown to constrict the axons in

the LC, interrupting RGC access to key target-derived NFs. Further, studies have

shown that NFs, particularly BDNF, appear trapped in the unmyelinated ONH,

accumulating along with its TRKB receptor and motor dynein proteins, following chronic

and acute IOP elevation [79-81]. Secondly, NF productions can be altered as microglia

and astrocytic Müller cells become reactive in response to various triggers. Microglial

Page 25: Interactome Analysis Identifies Novel Targets of

14

NF secretion shifts under stress, promoting pro-apoptotic or pro-survival signaling,

depending on the context [82, 83]. A recent study showed that this reduction in NF

production is reflected in lower BDNF concentrations in POAG patient aqueous humor,

lacrimal fluid and serum levels [84]. We have also demonstrated that NFL and ONH

astrocyte reactivity is associated with reduced neuroprotective lipid mediators, following

metabolic stress, leading to increased RGC vulnerability [2].

NFs promote neuronal survival through inhibition of apoptotic pathways [85]. More

specifically, NF deprivation induces the intrinsic apoptotic pathway, causing reduced

ATP production, ROS generation, cytochrome C release, and subsequent caspase

activation [86]. This occurs through the actions of MAPKs, such as c-Jun N-terminal

kinases (JNKs) and p38 [87] to mediatex mitochondrial dysfunction, and apoptosis

through the pro-apoptotic BCL-2 protein BAX [88, 89]. Thus, different trigger events,

namely elevated IOP and low CSFP, can converge to cause NF deprivation in the retina

through mechanical blockage and reduced glial secretion, resulting in increased RGC

vulnerability.

1.2.3 Metabolic and Oxidative stress

Oxidative stress, a key mechanism in glaucomatous degeneration, stems from two

major soruces: mitochondrial dysfunction, and the consequences of glial reactivity [90].

Mitochondrial dysfunction is observed following both vascular and/or mechanical injury

[91, 92], and also as part of the apoptotic cascade (both intrinsic and extrinsic) [86].

Mitochondria are abundantly present around and within the ONH, and particularly within

unmyelinated RGC axons in the retina, in order to meet the energetic demands of

Page 26: Interactome Analysis Identifies Novel Targets of

15

constant neuronal communication and axoplasmic transport [93]. Their high

concentration in the ONH makes any defect in function highly amplified for the ONH’s

physiological integrity. Up to 90% of the ATP produced by mitochondria is used to

maintain action potentials and neuronal survival [94]. In addition to being energy

generating organelles, they also serve as an integration centre for the various cellular

signals, such as inflammatory cytokines, ultimately deciding the fate of RGCs.

Oxidative stress, in the form of excessive ROS production, results from impaired

mitochondrial electron chain transfer. ROS, including superoxide anions (O2-) and

hydrogen peroxide (H2O2) are highly unstable chemical species that cause DNA

damage and protein oxidation, and may also impact axonal membrane polarity [95] . In

normal physiology, ROS are produced at low levels in the cytosol and play the role of a

second messenger in several cellular transduction pathways and regulate apoptosis.

Evidence of mitochondrial stress in glaucomatous retinas is reflected by marked

upregulation of hypoxia-sensitive pathways. For example, the hypoxia-inducible factor-

1alpha (HIF-1α) is found at higher concentrations in glaucomatous retinas, compared to

normal eyes [96]. HIF-1α is a transcriptional activator that induces the expression of

several proteins whose main function is to increase O2 availability in hypoxic tissue [97].

Additionally, HIF-1α localization within the retina corresponds to positions of visual

defects in examined eyes.

Age is the number one risk factor associated with glaucoma, and this may be linked to

underlying mitochondrial dysfunction [98]. High levels of mitochondrial DNA (mtDNA)

mutations in the transgenic polymerase γ (PolG) mouse model produced decreased

Page 27: Interactome Analysis Identifies Novel Targets of

16

expression of mitochondrial oxidative phosphorylation enzymes, and a higher

susceptibility to various RGC injuries [99, 100]. The coenzyme Q10 (CoQ10) was shown

to ameliorate oxidative stress–mediated RGC degeneration by preventing the

mitochondrial alterations in mice [101]. Further, Osbourne et al have suggested that

mitochondrial defects related to aging can be attenuated using non-invasive light

therapy [102, 103].

Oxidative stress occurs as a result of malfunctions in one or more of the mitochondria’s

four main functions: generation of energy in the form of ATP, regulation of ROS

production, regulation of cytosolic calcium levels, and apoptosis modulation via

mitochondrial permeability [104, 105]. An imbalance in the proportions of mitochondrial

proteins encoded by cellular and mitochondrial genes are believed to underlie these

changes [106]. Williams et al recently reported a range of differentially expressed genes

in the mitochondrial dysfunction and oxidative phosphorylation pathways in the DBA2J

glaucoma mouse model [107]. They further demonstrated that the resulting

mitochondrial vulnerability can be attenuated with Vitamin B3 supplement, providing a

candidate complementary treatment to IOP management. Oxidative stress can also

inhibit the vital mammalian target of rapamycin (mTOR) pathway, a master regulator of

nutrient sensing and mitochondrial function [108]. This inhibition acts through the

tuberous sclerosis complex (TSC1/2) and the regulated in development and DNA

damage response (REDD) proteins, leading to dendrite retraction and RGC death [109].

Page 28: Interactome Analysis Identifies Novel Targets of

17

1.2.3.1 Integration of mitochondria with other cascades.

Mitochondria dysfunction can also be caused by interplay with other cascades. For

example, failure in astrocyte anti-oxidant buffering capacity plays a major role in

increasing oxidative stress and cytokine release in the retina [47]. Similarly, TNFα has

been shown to induce mitochondrial dysfunction through caspase-mediated BH3

interacting domain death agonist (BID) cleavage [110]. Our group has also

demonstrated that this loss is linked to the transcriptional co-activator peroxisome

proliferator-activated receptor γ coactivator-1α (PGC-1α), which regulates cellular

adaptive energy [46]. Rescue of this signal through agonism of the upstream AMP-

activated protein kinase (AMPK) with the AMP mimetic AICAR (5-aminoimidazole-4-

carboxamide ribonucleotide), provided therapeutic increases in astrocytic antioxidants,

particularly essential precursors for the key detoxifying CNS peptide glutathione (GSH).

This mechanism led to increased survival of RGCs and reduced glial reactivity in an

acute rat IOP-dependent ischemia model [48]. In support of this concept, a recent large-

cohort analyses showed that the AMPK-inducing drug, metformin, is associated with a

dose-dependent reduced risk of POAG in diabetic patients [111]. Further, NF withdrawal

in glaucoma also indirectly contributes to oxidative stress by activating the mitochondrial

intrinsic apoptosis pathway [85, 89].

There are three general points that emerge when discussing these cascades and

proposed interventions: 1. cascade events are not mutually exclusive, but are

overlapping and contingent on one another. For instance, while glial reactivity and

oxidative stress are often described as two distinct events, each can contribute to the

Page 29: Interactome Analysis Identifies Novel Targets of

18

incidence of the other [47, 48, 90]. Thus, when targeting one glaucoma mechanism, it is

expected that other dependent mechanisms would in turn be affected. 2. Cascade

events may not be inherently deleterious, but can simply be outside of their homeostatic

range. For instance, excess nitric oxide (NO) production by reactive glia is derived from

constitutive production necessary for vascular regulation [112, 113]. This observation

may be important in determining the degree and aggressiveness of therapeutic

intervention. 3. Regardless of the induced cascades, the end result is typically RGC

apoptosis [85, 114], as described in studies utilizing animal models, such as elevated

IOP [115-118], ON crush and axotomy [117, 119], excitotoxicity [59, 120, 121], and in

human disease [122-125]. Still, some common themes emerge that are driven by

particular triggers.

1.3 Phosphatidylinositol 3-kinase

One of the key signaling pathways activated by NFs is the phosphatidylinositol 3-kinase

(PI3K) pathway. PI3ks are lipid kinases, which mediate intracellular signals regulating a

wide range of biological processes, including cell proliferation, metabolism, migration,

and protection against metabolic stress [126]. The PI3K family has three different

classes (Class I, Class II, and Class III) based on the structure, target specificity, and

cell type [127]. For the current discussion, class I will be discussed, since it is primarily

involved in transducing growth factor signaling [128]. Class IA PI3Ks, including PI3K α,

β , δ are heterodimers composed by one of the p110 catalytic subunits (p110 α , p110 β

, and p110 δ), coupled to a regulatory subunit of the p85 family, including p85α, P85β,

and p55y. (see review [129]). PI3K is activated by binding of its p85 regulatory subunit

Page 30: Interactome Analysis Identifies Novel Targets of

19

directly to phosphorylated RTK, or indirectly to an adapter protein such as insulin

receptor substrate-1,2 (IRS-1,2)[130]. Following IRS activation, the signaling cascade

boosts glucose uptake through a series of phosphorylation events [131]. Alternatively,

PI3k is activated or by interaction of its p110 catalytic subunit with Ras-GTP. PI3K

activation leads to the catalyzation the second messenger polyphosphoinositide

phosphatidylinositol 3,4,5-trisphosphate (PIP3) at the cell membrane [132] and

subsequent phosphorylation of AKT [129].

1.3.1 PI3K role in promoting cell survival

Following its phosphorylation, Akt translocates in the cytosol and to the nucleus, where

it activates a number of metabolic regulators, pro-survival factors, and transcription

factors [129]. For example, phosphorylalation of AKT leads to; 1) inactivation of

apoptotic transcription factors such as the nuclear factor kappa-light-chain-enhancer of

activated B cells (NF-kB), BAD, procaspase-9 and Forkhead (FKHR) transcription

factors (Figure 1-4) [129, 133], 2) activation of metabolic regulators such as endothelial

nitric oxide synthase (eNOS) and glycogen synthase kinase (GSK)-3β, leading to

boosting of cellular bioenergetics [134, 135], and 3) activation of mammalian target of

rapamycin (mTOR), which leads to increased translation of growth proteins[135].

Furthermore, mTOR phosphorylates components of the protein synthesis machinery,

including the ribosomal protein S6 kinases (p70S6K) and 4E-binding protein (4E-BP),

resulting in anti-apoptotic activities [136]. Conversely, mTOR is inhibited in response to

energy depletion, allowing energy conservation through LKB1 (STK11)-mediated

activation of AMP-activated protein kinase (AMPK), a sensor of the cellular ATP/ADP

Page 31: Interactome Analysis Identifies Novel Targets of

20

ratio. Globally, AKT regulates its targets through generating binding sites for the adaptor

protein 14-3-3 proteins, modifying their localization, activity, and stability in cytosol

[137].

Figure 1-4 A Schematic of PI3K/AKT pathway and key targets.

The PI3K/AKT pathway culminates into pro-survival, anti-apoptotic, and metabolic boosting effects.

Adopted with permission from Hennessy et al, 2005 [136].

1.3.2 PI3K interaction with RNA binding proteins

RNA binding proteins (RBP) play an important role in post-transcriptional gene

regulation. RBPs’ functions include splicing, nuclear export, and polyadenylation, which

Page 32: Interactome Analysis Identifies Novel Targets of

21

are carried out through physical interactions with adenine-(or adenosine) and uridine-

rich elements of RNA [138]. A range of kinases have been shown to interact and

activate RBPs, often through adapter proteins, ultimately altering the stability or half-life

of mRNA transcripts[139-141]. Thus, RBP phosphorylation by kinases and

dephosphorylation by phosphatases functions as an on/off switch to control mRNA

decay in response to extracellular stimuli [142].

PI3K exerts part of its activity through interacting with a range of RBPs, with the adapter

protein 14-3-3 functioning as a mediator for the interaction. Among the RBP substrates

of AKT are KSRP, BRF1 (or TIS11b), NF90, CELF1, and YB1 [142]. In the case of

TIS11b, PI3K promotes the stability of Iterlukin-3 (IL-3) mRNA through AKT

phosphorylation of TIS11b, an interaction mediated by 14-3-3[143]. Following

phosphorylation of TIS11, it is sequestered in the cytoplasm, disabling it from promoting

mRNA decay[144]. The role of the PI3K pathway in controlling mRNA stability was

further illustrated by its involvement in stabilizing 20 other transcripts, through

interactions with the RBPs TIS11b and KSRP [145]. In C2C12 myoblasts, KSRP

phosphorylation at the Ser193 site by AKT detaches myogenin mRNA from KSRP in the

cytoplasm, reversing KSRP-mediated decay of myogenin mRNA (Figure 1-5) [142]. This

process occurs in conjunction with 14-3-3ζ, interaction with KSRP, translocating it from

the cytoplasm to the nucleus, and mediating the maturation of myogenin mRNA

transcript.

Page 33: Interactome Analysis Identifies Novel Targets of

22

Figure 1-5. A schematic of the mechanism of action of the RBP KSRP in response to NF-initiated

AKT phosphorylation.

Following NF binding, KSRP is phosphorylated by AKT, which leads to the dissociation

of KSRP from myogenin mRNA. This dissociation leads to the stabilization of the mRNA

in the cytosol, as KSRP is inhibited from degrading it. Subsequently, KSRP associates

Page 34: Interactome Analysis Identifies Novel Targets of

23

with 14-3-3 and translocates into the nucleus. Adopted with permission Thepar, R and

Denmon, A. P., 2013 [142].

1.3.3 Poly-Adenosine RNA binding RBP

Polyadenosine RNA binding proteins (Pabs) are involved in regulating several stages of

gene expression. Among the key Pabs are PABPN1 and PABPC1, as well as the

recently characterized Pab, Zinc Finger CCCH-type containing #14, or ZC3H14 [146].

Biochemical studies demonstrate that ZC3H14 utilizes five evolutionarily conserved

tandem cysteine3histidine (CCCH) zinc fingers, which binds to polyadenosine (polyA)

RNA with high affinity and specificity[147]. There are at least four known alternatively

spliced variants of ZC3H14, which give rise to four distinct protein isoforms [148].

Isoforms 1-3 have a N-terminal proline tryptophan isoleucine (PWI), which mediates

entry through the nuclear pore; thus, these isoforms are expected to localized within the

cytoplasm as well as the nucleus [149]. Isoform 4 lacks key domains needed to interact

with the nuclear pore and is therefore localized mainly within the cytoplasm [148].

Through binding with the polyA tails, ZC3H14 carries out a range of post-transcriptional

modifications on premature transcripts [147, 150]. These include control of PolyA tails

length [151], nuclear export [149], and mRNA splicing [152]. Mutation of the ZC3H14

impairs neural function in Drosophila and has been associated with cognitive deficits in

humans [153]. ZC3H14 knockout mouse models show anatomical changes in brain

ventricles as well as working memory deficits [154]. Thus far, no studies have reported

a ZC3H14 interaction with PI3K, or other kinases.

Page 35: Interactome Analysis Identifies Novel Targets of

24

The THO complex 1 (Thoc1) encodes a nuclear matrix protein that also binds to PolyA

tails of mRNA [155]. As part of the THO ribonucleoprotein complex, Thoc1 protein is

recruited to premature RNA transcripts, which allows access to various RNA processing

and export machinery [156, 157]. Impaired Thoc1 function negatively impacts

transcription elongation and nuclear export, which also affects mRNA stability in the

cytoplasm [157]. A 2018 study reported that ZC3H14 interacts with THO complex to

coordinately control RNA processing, poly(A) tail length, and consequently mRNA

stability [155]. The same study showed that mRNA targets of ZC3H14 and THOC1

include the postsynaptic density protein 95 (Psd95), ATP synthase lipid-binding protein

(Atp5g1) and microtubule-associated protein tau (MAPT). Conversely, knockdown of

ZC3H14 or THO components lead to decreased levels of mature transcripts and

accumulation of Atp5g1 and Psd95 pre-mRNA in the cytoplasm. The gene encodes a

subunit of mitochondrial ATP synthase, which catalyzes ATP synthesis [158]. Thus,

ZC3H14 and THOC1’s functions may have a direct impact on cell energy synthesis and

survival.

Page 36: Interactome Analysis Identifies Novel Targets of

25

Chapter 1 references

1. Alqawlaq, S., J.G. Flanagan, and J.M. Sivak, All roads lead to glaucoma: Induced retinal injury cascades contribute to a common neurodegenerative outcome. Exp Eye Res, 2018.

2. Livne-Bar, I., et al., Astrocyte-derived lipoxins A4 and B4 promote neuroprotection from acute and chronic injury. J Clin Invest, 2017.

3. Xie, A., et al., Shared mechanisms of neurodegeneration in Alzheimer's disease and Parkinson's disease. Biomed Res Int, 2014. 2014: p. 648740.

4. Sivak, J.M., The aging eye: common degenerative mechanisms between the Alzheimer's brain and retinal disease. Invest Ophthalmol Vis Sci, 2013. 54(1): p. 871-80.

5. Booth, H.D.E., W.D. Hirst, and R. Wade-Martins, The Role of Astrocyte Dysfunction in Parkinson's Disease Pathogenesis. Trends Neurosci, 2017. 40(6): p. 358-370.

6. Brambilla, L., F. Martorana, and D. Rossi, Astrocyte signaling and neurodegeneration: new insights into CNS disorders. Prion, 2013. 7(1): p. 28-36.

7. Zamanian, J.L., et al., Genomic Analysis of Reactive Astrogliosis. Journal of Neuroscience, 2012. 32(18): p. 6391-6410.

8. Liddelow, S.A., et al., Neurotoxic reactive astrocytes are induced by activated microglia. Nature, 2017. 541(7638).

9. Tham, Y.C. and C.Y. Cheng, Author reply: To PMID 24974815. Ophthalmology, 2015. 122(7): p. e41-2.

10. Cook, C. and P. Foster, Epidemiology of glaucoma: what's new? Can J Ophthalmol, 2012. 47(3): p. 223-6.

11. Friedman, D.S., et al., Prevalence of open-angle glaucoma among adults in the United States. Arch Ophthalmol, 2004. 122(4): p. 532-8.

12. Samarawickrama, C., et al., Ethnic differences in optic nerve head and retinal nerve fibre layer thickness parameters in children. Br J Ophthalmol, 2010. 94(7): p. 871-6.

13. Tielsch, J.M., et al., Racial variations in the prevalence of primary open-angle glaucoma. The Baltimore Eye Survey. JAMA, 1991. 266(3): p. 369-74.

14. Kass, M.A., et al., The Ocular Hypertension Treatment Study: a randomized trial determines that topical ocular hypotensive medication delays or prevents the onset of primary open-angle glaucoma. Arch Ophthalmol, 2002. 120(6): p. 701-13; discussion 829-30.

Page 37: Interactome Analysis Identifies Novel Targets of

26

15. Leske, M.C., et al., Incidence of open-angle glaucoma: the Barbados Eye Studies. The Barbados Eye Studies Group. Arch Ophthalmol, 2001. 119(1): p. 89-95.

16. Sommer, A., et al., Relationship between intraocular pressure and primary open angle glaucoma among white and black Americans. The Baltimore Eye Survey. Arch Ophthalmol, 1991. 109(8): p. 1090-5.

17. Leske, M.C., et al., Predictors of long-term progression in the early manifest glaucoma trial. Ophthalmology, 2007. 114(11): p. 1965-72.

18. Liu, L., et al., Optical Coherence Tomography Angiography of the Peripapillary Retina in Glaucoma. JAMA Ophthalmol, 2015. 133(9): p. 1045-52.

19. Satilmis, M., et al., Rate of progression of glaucoma correlates with retrobulbar circulation and intraocular pressure. Am J Ophthalmol., 2003. 135(5): p. 664-9.

20. Sigal, I.A., et al., - Investigation of the biomechanical environment within the optic nerve head by finite element modelling, in Computational Fluid and Solid Mechanics 2003, K.J. Bathe, Editor. 2003, Elsevier Science Ltd: Oxford. p. 1810-1813.

21. Nguyen, T.D. and C.R. Ethier, Biomechanical assessment in models of glaucomatous optic neuropathy. Exp Eye Res, 2015. 141: p. 125-38.

22. Jonas, J.B., et al., Facts and myths of cerebrospinal fluid pressure for the physiology of the eye. Prog Retin Eye Res, 2015. 46: p. 67-83.

23. Cheng, J.-W., et al., The Prevalence of Primary Glaucoma in Mainland China: A Systematic Review and Meta-analysis. Journal of Glaucoma, 2013. 22(4): p. 301-306 10.1097/IJG.0b013e31824083ca.

24. Zhao, D., et al., The Association of Blood Pressure and Primary Open-Angle Glaucoma: A Meta-analysis. American Journal of Ophthalmology, 2014. 158(3): p. 615-627.e9.

25. Charlson, M.E., et al., Nocturnal Systemic Hypotension Increases the Risk of Glaucoma Progression. Ophthalmology, 2014. 121(10): p. 2004-2012.

26. Wong, V.H.Y., B.V. Bui, and A.J. Vingrys, Clinical and experimental links between diabetes and glaucoma. Clinical and Experimental Optometry, 2011. 94(1): p. 4-23.

27. Zhou, M., et al., Diabetes mellitus as a risk factor for open-angle glaucoma: a systematic review and meta-analysis. PLoS One, 2014. 9(8): p. e102972.

28. Quigley, H.A., Open-angle glaucoma. N Engl J Med, 1993. 328(15): p. 1097-106.

Page 38: Interactome Analysis Identifies Novel Targets of

27

29. Weinreb, R.N., et al., Primary open-angle glaucoma. Nat Rev Dis Primers, 2016. 2: p. 16067.

30. Tham, Y.C., et al., Global prevalence of glaucoma and projections of glaucoma burden through 2040: a systematic review and meta-analysis. Ophthalmology, 2014. 121(11): p. 2081-90.

31. Nickells, R.W., Apoptosis of retinal ganglion cells in glaucoma: an update of the molecular pathways involved in cell death. Surv Ophthalmol, 1999. 43 Suppl 1: p. S151-61.

32. Quigley, H.A., N.R. Miller, and T. George, Clinical evaluation of nerve fiber layer atrophy as an indicator of glaucomatous optic nerve damage. Arch Ophthalmol, 1980. 98(9): p. 1564-71.

33. Miki, A., et al., Rates of retinal nerve fiber layer thinning in glaucoma suspect eyes. Ophthalmology, 2014. 121(7): p. 1350-8.

34. Jones, H.J., et al., Quantitative analysis of three-dimensional fibrillar collagen microstructure within the normal, aged and glaucomatous human optic nerve head. J R Soc Interface, 2015. 12(106).

35. Exler, R.E., et al., Biomechanical insult switches PEA-15 activity to uncouple its anti-apoptotic function and promote erk mediated tissue remodeling. Experimental Cell Research, 2016. 340(2): p. 283-294.

36. Calkins, D.J., et al., The challenge of regenerative therapies for the optic nerve in glaucoma. Experimental Eye Research, 2017. 157: p. 28-33.

37. Hernandez, M.R., H. Miao, and T. Lukas, Astrocytes in glaucomatous optic neuropathy. Prog Brain Res, 2008. 173: p. 353-73.

38. Pekny, M. and M. Pekna, ASTROCYTE REACTIVITY AND REACTIVE ASTROGLIOSIS: COSTS AND BENEFITS. Physiological Reviews, 2014. 94(4): p. 1077-1098.

39. Vohra, R., et al., Essential Roles of Lactate in Müller Cell Survival and Function. Mol Neurobiol, 2018.

40. Chang, M.Y., et al., Neurons and astrocytes secrete factors that cause stem cells to differentiate into neurons and astrocytes, respectively. Mol Cell Neurosci, 2003. 23(3): p. 414-26.

41. Patel, A.J. and C.W. Gray, Neurotrophic Factors Produced by Astrocytes involved in the Regulation of Cholinergic Neurons in the Central Nervous System, in Biology and Pathology of Astrocyte-Neuron Interactions, S. Fedoroff, B.H.J. Juurlink, and R. Doucette, Editors. 1993, Springer US: Boston, MA. p. 103-115.

Page 39: Interactome Analysis Identifies Novel Targets of

28

42. Chiang, N., et al., Identification of resolvin D2 receptor mediating resolution of infections and organ protection. J Exp Med, 2015. 212(8): p. 1203-17.

43. Rogers, R.S., et al., Proteomics Analyses of Human Optic Nerve Head Astrocytes Following Biomechanical Strain. Molecular & Cellular Proteomics, 2012. 11(2).

44. Tezel, G. and F.A.P.O.R.I.C.W. Group, The role of glia, mitochondria, and the immune system in glaucoma. Invest Ophthalmol Vis Sci, 2009. 50(3): p. 1001-12.

45. Chong, R.S. and K.R. Martin, Glial cell interactions and glaucoma. Current Opinion in Ophthalmology, 2015. 26(2): p. 73-77.

46. Guo, X., et al., PGC-1 alpha Signaling Coordinates Susceptibility to Metabolic and Oxidative Injury in the Inner Retina. American Journal of Pathology, 2014. 184(4): p. 1017-1029.

47. Nahirnyj, A., et al., ROS Detoxification and Proinflammatory Cytokines Are Linked by p38 MAPK Signaling in a Model of Mature Astrocyte Activation. Plos One, 2013. 8(12).

48. Guo, X., et al., The AMPK-PGC-1alpha signaling axis regulates the astrocyte glutathione system to protect against oxidative and metabolic injury. Neurobiology of disease, 2018. 113: p. 59-69.

49. Zamanian, J.L., et al., Genomic Analysis of Reactive Astrogliosis. The Journal of neuroscience : the official journal of the Society for Neuroscience, 2012. 32(18): p. 6391-6410.

50. Qu, J. and T.C. Jakobs, The Time Course of Gene Expression during Reactive Gliosis in the Optic Nerve. PLoS One, 2013. 8(6): p. e67094.

51. Sun, D., J. Qu, and T.C. Jakobs, Reversible reactivity by optic nerve astrocytes. Glia, 2013. 61(8): p. 1218-35.

52. Pekny, M., U. Wilhelmsson, and M. Pekna, The dual role of astrocyte activation and reactive gliosis. Neuroscience Letters, 2014. 565: p. 30-38.

53. Anderson, M.A., et al., Astrocyte scar formation aids central nervous system axon regeneration. Nature, 2016. 532(7598): p. 195-200.

54. Sun, D., S. Moore, and T.C. Jakobs, Optic nerve astrocyte reactivity protects function in experimental glaucoma and other nerve injuries. J Exp Med, 2017. 214(5): p. 1411-1430.

55. Ebneter, A., et al., Microglial activation in the visual pathway in experimental glaucoma: spatiotemporal characterization and correlation with axonal injury. Invest Ophthalmol Vis Sci, 2010. 51(12): p. 6448-60.

Page 40: Interactome Analysis Identifies Novel Targets of

29

56. Wax, M.B. and G. Tezel, Immunoregulation of retinal ganglion cell fate in glaucoma. Exp Eye Res, 2009. 88(4): p. 825-30.

57. Johnson, E.C. and J.C. Morrison, Friend or Foe? Resolving the Impact of Glial Responses in Glaucoma. Journal of Glaucoma, 2009. 18(5): p. 341-353.

58. Colangelo, A.M., L. Alberghina, and M. Papa, Astrogliosis as a therapeutic target for neurodegenerative diseases. Neurosci Lett, 2014. 565: p. 59-64.

59. Livne-Bar, I., et al., Pharmacologic inhibition of reactive gliosis blocks TNF-alpha-mediated neuronal apoptosis. Cell Death Dis, 2016. 7(9): p. e2386.

60. de Pablo, Y., et al., Drugs targeting intermediate filaments can improve neurosupportive properties of astrocytes. Brain Research Bulletin, 2018. 136: p. 130-138.

61. Vidal, L., et al., Reaction of Muller cells in an experimental rat model of increased intraocular pressure following timolol, latanoprost and brimonidine. Brain Res Bull, 2010. 82(1-2): p. 18-24.

62. Bosco, A., et al., Reduced retina microglial activation and improved optic nerve integrity with minocycline treatment in the DBA/2J mouse model of glaucoma. Investigative Ophthalmology & Visual Science, 2008. 49(4): p. 1437-1446.

63. Johnson, E.C., et al., Neurotrophin roles in retinal ganglion cell survival: Lessons from rat glaucoma models. Experimental Eye Research, 2009. 88(4): p. 808-815.

64. Kimura, A., et al., Neuroprotection, Growth Factors and BDNF-TrkB Signalling in Retinal Degeneration. International Journal of Molecular Sciences, 2016. 17(9).

65. Mansourrobaey, S., et al., EFFECTS OF OCULAR INJURY AND ADMINISTRATION OF BRAIN-DERIVED NEUROTROPHIC FACTOR ON SURVIVAL AND REGROWTH OF AXOTOMIZED RETINAL GANGLION-CELLS. Proceedings of the National Academy of Sciences of the United States of America, 1994. 91(5): p. 1632-1636.

66. Di Polo, A., et al., Prolonged delivery of brain-derived neurotrophic factor by adenovirus-infected Muller cells temporarily rescues injured retinal ganglion cells. Proceedings of the National Academy of Sciences of the United States of America, 1998. 95(7): p. 3978-3983.

67. Valiente-Soriano, F.J., et al., BDNF Rescues RGCs But Not Intrinsically Photosensitive RGCs in Ocular Hypertensive Albino Rat Retinas. Investigative Ophthalmology & Visual Science, 2015. 56(3): p. 1924-1936.

68. Lambiase, A., et al., Experimental and clinical evidence of neuroprotection by nerve growth factor eye drops: Implications for glaucoma. Proceedings of the National Academy of Sciences of the United States of America, 2009. 106(32): p. 13469-13474.

Page 41: Interactome Analysis Identifies Novel Targets of

30

69. Sawai, H., et al., Brain-derived neurotrophic factor and neurotrophin-4/5 stimulate growth of axonal branches from regenerating retinal ganglion cells. Journal of Neuroscience, 1996. 16(12): p. 3887-3894.

70. Ji, J.Z., et al., CNTF promotes survival of retinal ganglion cells after induction of ocular hypertension in rats: the possible involvement of STAT3 pathway. European Journal of Neuroscience, 2004. 19(2): p. 265-272.

71. Schmeer, C., et al., Dose-dependent rescue of axotomized rat retinal ganglion cells by adenovirus-mediated expression of glial cell-line derived neurotrophic factor in vivo. European Journal of Neuroscience, 2002. 15(4): p. 637-643.

72. Deister, C. and C.E. Schmidt, Optimizing neurotrophic factor combinations for neurite outgrowth. J Neural Eng, 2006. 3(2): p. 172-9.

73. Cao, X. and M.S. Shoichet, Investigating the synergistic effect of combined neurotrophic factor concentration gradients to guide axonal growth. Neuroscience, 2003. 122(2): p. 381-9.

74. Cui, Q., et al., Expression of trkA, trkB, and trkC in injured and regenerating retinal ganglion cells of adult rats. Invest Ophthalmol Vis Sci, 2002. 43(6): p. 1954-64.

75. Seki, M., et al., Muller cells as a source of brain-derived neurotrophic factor in the retina: Noradrenaline upregulates brain-derived neurotrophic factor levels in cultured rat Muller cells. Neurochemical Research, 2005. 30(9): p. 1163-1170.

76. Spalding, K.L., R.A. Rush, and A.R. Harvey, Target-derived and locally derived neurotrophins support retinal ganglion cell survival in the neonatal rat retina. Journal of Neurobiology, 2004. 60(3): p. 319-327.

77. Quigley, H.A., et al., Retrograde axonal transport of BDNF in retinal ganglion cells is blocked by acute IOP elevation in rats. Invest Ophthalmol Vis Sci, 2000. 41(11): p. 3460-6.

78. Zhang, Z., et al., Axonal Transport in the Rat Optic Nerve Following Short-Term Reduction in Cerebrospinal Fluid Pressure or Elevation in Intraocular Pressure. Invest Ophthalmol Vis Sci, 2015. 56(8): p. 4257-66.

79. Pease, M.E., et al., Obstructed axonal transport of BDNF and its receptor TrkB in experimental glaucoma. Invest Ophthalmol Vis Sci, 2000. 41(3): p. 764-74.

80. Iwabe, S., et al., Retrograde axonal transport obstruction of brain-derived neurotrophic factor (BDNF) and its TrkB receptor in the retina and optic nerve of American Cocker Spaniel dogs with spontaneous glaucoma. Vet Ophthalmol, 2007. 10 Suppl 1: p. 12-9.

Page 42: Interactome Analysis Identifies Novel Targets of

31

81. Martin, K.R., et al., Optic nerve dynein motor protein distribution changes with intraocular pressure elevation in a rat model of glaucoma. Exp Eye Res, 2006. 83(2): p. 255-62.

82. Harada, T., et al., Microglia-Muller glia cell interactions control neurotrophic factor production during light-induced retinal degeneration. Journal of Neuroscience, 2002. 22(21): p. 9228-9236.

83. Filous, A.R. and J. Silver, "Targeting astrocytes in CNS injury and disease: A translational research approach". Progress in Neurobiology, 2016. 144: p. 173-187.

84. Shpak, A.A., et al., Brain-Derived Neurotrophic Factor in Patients with Primary Open-Angle Glaucoma and Age-related Cataract. Current eye research, 2018. 43(2): p. 224-231.

85. Almasieh, M., et al., The molecular basis of retinal ganglion cell death in glaucoma. Progress in Retinal and Eye Research, 2012. 31(2): p. 152-181.

86. Green, D.R., L. Galluzzi, and G. Kroemer, Metabolic control of cell death. Science, 2014. 345(6203): p. 1466-+.

87. Yang, Z., et al., Changes in gene expression in experimental glaucoma and optic nerve transection: The equilibrium between protective and detrimental mechanisms. Investigative Ophthalmology & Visual Science, 2007. 48(12): p. 5539-5548.

88. Moldoveanu, T., et al., Many players in BCL-2 family affairs. Trends in Biochemical Sciences, 2014. 39(3): p. 101-111.

89. Maes, M.E., C.L. Schlamp, and R.W. Nickells, BAX to basics: How the BCL2 gene family controls the death of retinal ganglion cells. Prog Retin Eye Res, 2017. 57: p. 1-25.

90. Tezel, G., Oxidative stress in glaucomatous neurodegeneration: mechanisms and consequences. Prog Retin Eye Res, 2006. 25(5): p. 490-513.

91. Kaur, C., W.S. Foulds, and E.A. Ling, Hypoxia-ischemia and retinal ganglion cell damage. Clin Ophthalmol, 2008. 2(4): p. 879-89.

92. Kaufman, D.M., et al., Ageing and hypoxia cause protein aggregation in mitochondria. Cell Death and Differentiation, 2017. 24(10): p. 1730-1738.

93. Bristow, E.A., et al., The distribution of mitochondrial activity in relation to optic nerve structure. Arch Ophthalmol, 2002. 120(6): p. 791-6.

94. Osborne, N.N., et al., Glaucoma: Focus on mitochondria in relation to pathogenesis and neuroprotection. European Journal of Pharmacology, 2016. 787: p. 127-133.

Page 43: Interactome Analysis Identifies Novel Targets of

32

95. Almasieh, M., et al., Axonal Degeneration in Retinal Ganglion Cells Is Associated with a Membrane Polarity-Sensitive Redox Process. J Neurosci, 2017. 37(14): p. 3824-3839.

96. Tezel, G. and M.B. Wax, Hypoxia-inducible factor 1alpha in the glaucomatous retina and optic nerve head. Arch Ophthalmol, 2004. 122(9): p. 1348-56.

97. Zagorska, A. and J. Dulak, HIF-1: the knowns and unknowns of hypoxia sensing. Acta Biochim Pol, 2004. 51(3): p. 563-85.

98. Bhatia-Dey, N., et al., Cellular Senescence as the Causal Nexus of Aging. Frontiers in Genetics, 2016. 7.

99. Kong, Y.X.G., et al., Increase in mitochondrial DNA mutations impairs retinal function and renders the retina vulnerable to injury. Aging Cell, 2011. 10(4): p. 572-583.

100. Wang, A.L., et al., Age-related increase in mitochondrial DNA damage and loss of DNA repair capacity in the neural retina. Neurobiology of Aging, 2010. 31(11): p. 2002-2010.

101. Lee, D., et al., Coenzyme Q10 Inhibits Glutamate Excitotoxicity and Oxidative Stress-Mediated Mitochondrial Alteration in a Mouse Model of Glaucoma. Investigative Ophthalmology & Visual Science, 2014. 55(2): p. 993-1005.

102. Osborne, N.N., C. Nunez Alvarez, and S. del Olmo Aguado, Targeting mitochondrial dysfunction as in aging and glaucoma. Drug Discovery Today, 2014. 19(10): p. 1613-1622.

103. Osborne, N.N., et al., Visual light effects on mitochondria: The potential implications in relation to glaucoma. Mitochondrion, 2017. 36: p. 29-35.

104. Zorov, D.B., M. Juhaszova, and S.J. Sollott, MITOCHONDRIAL REACTIVE OXYGEN SPECIES (ROS) AND ROS-INDUCED ROS RELEASE. Physiological Reviews, 2014. 94(3): p. 909-950.

105. Kanamori, A., et al., Superoxide is an associated signal for apoptosis in axonal injury. Brain, 2010. 133(9): p. 2612-25.

106. Kotiadis, V.N., M.R. Duchen, and L.D. Osellame, Mitochondrial quality control and communications with the nucleus are important in maintaining mitochondrial function and cell health. Biochimica Et Biophysica Acta-General Subjects, 2014. 1840(4): p. 1254-1265.

107. Williams, P.A., et al., Vitamin B-3 modulates mitochondrial vulnerability and prevents glaucoma in aged mice. Science, 2017. 355(6326): p. 756-760.

108. Johnson, S.C., P.S. Rabinovitch, and M. Kaeberlein, mTOR is a key modulator of ageing and age-related disease. Nature, 2013. 493(7432): p. 338-345.

Page 44: Interactome Analysis Identifies Novel Targets of

33

109. Morquette, B., et al., REDD2-mediated inhibition of mTOR promotes dendrite retraction induced by axonal injury. Cell Death and Differentiation, 2015. 22(4): p. 612-625.

110. Tezel, G., TNF-alpha signaling in glaucomatous neurodegeneration. Glaucoma: an Open Window to Neurodegeneration and Neuroprotection, 2008. 173: p. 409-421.

111. Lin, H.-C., et al., Association of Geroprotective Effects of Metformin and Risk of Open-Angle Glaucoma in Persons With Diabetes Mellitus. Jama Ophthalmology, 2015. 133(8): p. 915-923.

112. Mishra, A., et al., Astrocytes mediate neurovascular signaling to capillary pericytes but not to arterioles. Nature Neuroscience, 2016. 19(12): p. 1619-1627.

113. Liu, B. and A.H. Neufeld, Expression of nitric oxide synthase-2 (NOS-2) in reactive astrocytes of the human glaucomatous optic nerve head. Glia, 2000. 30(2): p. 178-186.

114. Maes, M.E., C.L. Schlarnp, and R.W. Nickells, BAX to basics: How the BCL2 gene family controls the death of retinal ganglion cells. Progress in Retinal and Eye Research, 2017. 57: p. 1-25.

115. Guo, L., et al., Retinal ganglion cell apoptosis in glaucoma is related to intraocular pressure and IOP-induced effects on extracellular matrix. Invest Ophthalmol Vis Sci, 2005. 46(1): p. 175-82.

116. McKinnon, S.J., et al., Caspase activation and amyloid precursor protein cleavage in rat ocular hypertension. Invest Ophthalmol Vis Sci, 2002. 43(4): p. 1077-87.

117. Quigley, H.A., et al., Retinal ganglion cell death in experimental glaucoma and after axotomy occurs by apoptosis. Invest Ophthalmol Vis Sci, 1995. 36(5): p. 774-86.

118. Liu, H.H. and J.G. Flanagan, A Mouse Model of Chronic Ocular Hypertension Induced by Circumlimbal Suture. Invest Ophthalmol Vis Sci, 2017. 58(1): p. 353-361.

119. Sanchez-Migallon, M.C., et al., Apoptotic Retinal Ganglion Cell Death After Optic Nerve Transection or Crush in Mice: Delayed RGC Loss With BDNF or a Caspase 3 Inhibitor. Invest Ophthalmol Vis Sci, 2016. 57(1): p. 81-93.

120. Seitz, R. and E.R. Tamm, N-methyl-D-aspartate (NMDA)-mediated excitotoxic damage: a mouse model of acute retinal ganglion cell damage. Methods Mol Biol, 2013. 935: p. 99-109.

121. Li, Y., et al., p53 regulates apoptotic retinal ganglion cell death induced by N-methyl-D-aspartate. Mol Vis, 2002. 8: p. 341-50.

Page 45: Interactome Analysis Identifies Novel Targets of

34

122. Kerrigan, L.A., et al., TUNEL-positive ganglion cells in human primary open-angle glaucoma. Arch Ophthalmol, 1997. 115(8): p. 1031-5.

123. Okisaka, S., et al., Apoptosis in retinal ganglion cell decrease in human glaucomatous eyes. Jpn J Ophthalmol, 1997. 41(2): p. 84-8.

124. Tatton, N.A., et al., In situ detection of apoptosis in normal pressure glaucoma. a preliminary examination. Surv Ophthalmol, 2001. 45 Suppl 3: p. S268-72; discussion S273-6.

125. Cordeiro, M.F., et al., Real-time imaging of single neuronal cell apoptosis in patients with glaucoma. Brain, 2017. 140: p. 1757-1767.

126. Koh, S.H. and E.H. Lo, The Role of the PI3K Pathway in the Regeneration of the Damaged Brain by Neural Stem Cells after Cerebral Infarction. J Clin Neurol, 2015. 11(4): p. 297-304.

127. Leevers, S.J., B. Vanhaesebroeck, and M.D. Waterfield, Signalling through phosphoinositide 3-kinases: the lipids take centre stage. Curr Opin Cell Biol, 1999. 11(2): p. 219-25.

128. Jean, S. and A.A. Kiger, Classes of phosphoinositide 3-kinases at a glance. J Cell Sci, 2014. 127(Pt 5): p. 923-8.

129. Hers, I., E.E. Vincent, and J.M. Tavare, Akt signalling in health and disease. Cellular Signalling, 2011. 23(10): p. 1515-1527.

130. Vanhaesebroeck, B., et al., The emerging mechanisms of isoform-specific PI3K signalling. Nat Rev Mol Cell Biol, 2010. 11(5): p. 329-41.

131. Koh, S.H. and E.H. Lo, The Role of the PI3K Pathway in the Regeneration of the Damaged Brain by Neural Stem Cells after Cerebral Infarction. Journal of clinical neurology (Seoul, Korea), 2015. 11(4): p. 297-304.

132. Vanhaesebroeck, B. and M.D. Waterfield, Signaling by distinct classes of phosphoinositide 3-kinases. Exp Cell Res, 1999. 253(1): p. 239-54.

133. Carnero, A., et al., The PTEN/PI3K/AKT signalling pathway in cancer, therapeutic implications. Current Cancer Drug Targets, 2008. 8(3): p. 187-198.

134. Kitagishi, Y., et al., Dietary regulation of PI3K/AKT/GSK-3β pathway in Alzheimer's disease. Alzheimer's research & therapy, 2014. 6(3): p. 35-35.

135. Altomare, D.A. and A.R. Khaled, Homeostasis and the importance for a balance between AKT/mTOR activity and intracellular signaling. Current medicinal chemistry, 2012. 19(22): p. 3748-3762.

136. Hennessy, B.T., et al., Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov, 2005. 4(12): p. 988-1004.

Page 46: Interactome Analysis Identifies Novel Targets of

35

137. Gomez-Suarez, M., et al., 14-3-3 Proteins regulate Akt Thr308 phosphorylation in intestinal epithelial cells. Cell Death Differ, 2016. 23(6): p. 1060-72.

138. Venigalla, R.K. and M. Turner, RNA-binding proteins as a point of convergence of the PI3K and p38 MAPK pathways. Front Immunol, 2012. 3: p. 398.

139. Sadler, A.J., Orchestration of the activation of protein kinase R by the RNA-binding motif. J Interferon Cytokine Res, 2010. 30(4): p. 195-204.

140. Park, H., et al., TAR RNA-binding protein is an inhibitor of the interferon-induced protein kinase PKR. Proc Natl Acad Sci U S A, 1994. 91(11): p. 4713-7.

141. Li, J., et al., Modulation of an RNA-binding protein by abscisic-acid-activated protein kinase. Nature, 2002. 418: p. 793.

142. Thapar, R. and A.P. Denmon, Signaling pathways that control mRNA turnover. Cell Signal, 2013. 25(8): p. 1699-710.

143. Ming, X.F., et al., Parallel and independent regulation of interleukin-3 mRNA turnover by phosphatidylinositol 3-kinase and p38 mitogen-activated protein kinase. Mol Cell Biol, 2001. 21(17): p. 5778-89.

144. Benjamin, D., et al., BRF1 protein turnover and mRNA decay activity are regulated by protein kinase B at the same phosphorylation sites. Mol Cell Biol, 2006. 26(24): p. 9497-507.

145. Graham, J.R., et al., mRNA degradation plays a significant role in the program of gene expression regulated by phosphatidylinositol 3-kinase signaling. Mol Cell Biol, 2010. 30(22): p. 5295-305.

146. Wigington, C.P., et al., The Polyadenosine RNA-binding Protein, Zinc Finger Cys3His Protein 14 (ZC3H14), Regulates the Pre-mRNA Processing of a Key ATP Synthase Subunit mRNA. J Biol Chem, 2016. 291(43): p. 22442-22459.

147. Kelly, S.M., et al., Recognition of polyadenosine RNA by zinc finger proteins. Proc Natl Acad Sci U S A, 2007. 104(30): p. 12306-11.

148. Leung, S.W., et al., Splice variants of the human ZC3H14 gene generate multiple isoforms of a zinc finger polyadenosine RNA binding protein. Gene, 2009. 439(1-2): p. 71-8.

149. Marfatia, K.A., et al., Domain analysis of the Saccharomyces cerevisiae heterogeneous nuclear ribonucleoprotein, Nab2p. Dissecting the requirements for Nab2p-facilitated poly(A) RNA export. J Biol Chem, 2003. 278(9): p. 6731-40.

150. Kelly, S.M., et al., Recognition of polyadenosine RNA by the zinc finger domain of nuclear poly(A) RNA-binding protein 2 (Nab2) is required for correct mRNA 3'-end formation. J Biol Chem, 2010. 285(34): p. 26022-32.

Page 47: Interactome Analysis Identifies Novel Targets of

36

151. Kelly, S.M., et al., A conserved role for the zinc finger polyadenosine RNA binding protein, ZC3H14, in control of poly(A) tail length. RNA, 2014. 20(5): p. 681-8.

152. Soucek, S., et al., The Evolutionarily-conserved Polyadenosine RNA Binding Protein, Nab2, Cooperates with Splicing Machinery to Regulate the Fate of pre-mRNA. Mol Cell Biol, 2016.

153. Pak, C., et al., Mutation of the conserved polyadenosine RNA binding protein, ZC3H14/dNab2, impairs neural function in Drosophila and humans. Proc Natl Acad Sci U S A, 2011. 108(30): p. 12390-5.

154. Rha, J., et al., The RNA-binding protein, ZC3H14, is required for proper poly(A) tail length control, expression of synaptic proteins, and brain function in mice. Hum Mol Genet, 2017. 26(19): p. 3663-3681.

155. Morris, K.J. and A.H. Corbett, The polyadenosine RNA-binding protein ZC3H14 interacts with the THO complex and coordinately regulates the processing of neuronal transcripts. Nucleic Acids Res, 2018. 46(13): p. 6561-6575.

156. Luna, R., A.G. Rondón, and A. Aguilera, New clues to understand the role of THO and other functionally related factors in mRNP biogenesis. Biochim Biophys Acta, 2012. 1819(6): p. 514-20.

157. Rehwinkel, J., et al., Genome-wide analysis of mRNAs regulated by the THO complex in Drosophila melanogaster. Nat Struct Mol Biol, 2004. 11(6): p. 558-66.

158. Bonora, M., et al., Role of the c subunit of the FO ATP synthase in mitochondrial permeability transition. Cell Cycle, 2013. 12(4): p. 674-83.

Page 48: Interactome Analysis Identifies Novel Targets of

37

Chapter 2: Rationale, purpose, Hypothesis and Aims

Page 49: Interactome Analysis Identifies Novel Targets of

38

Rationale, Hypothesis and Aims

2.1 Rationale

Until the early 1980’s, glial cells had been thought of as simply structural, binding

together neuronal tissue, including the retina, and hence their name (glia is derived from

the Greek word for glue). This concept became challenged by growing evidence

demonstrating that glial cells, and especially astrocytes, sustain neuronal functions and

homeostasis in diverse and complex ways [1]. One important facet among these

functions is the astrocyte capacity to alleviate the impact of metabolic stress in neurons

through secreted pro-survival factors. This observation is of particular importance in the

context of neurodegenerative diseases, since excessive stimulation by neurochemicals,

such as glutamate, leads to death of neurons in a number of diseases including

Alzheimer’s disease, and glaucoma. However, we currently lack an understanding of

the basic neuronal pathways involved in transducing these astrocyte signals, which

prevents us from leveraging them for future treatment or diagnostic strategies.

Studying ACM provides a window into the inner workings of astrocyte-neuron

communication and associated protective outcomes in neurons. ACM contains a

complex mixture of proteins, lipids, neurochemicals and nucleic-acid containing

exosomes [2-5]. To explore ACM components, we isolated and cultured primary retinal

astrocytes as previously described [6, 7], and subsequently collected ACM for analysis.

Page 50: Interactome Analysis Identifies Novel Targets of

39

Astrocytes cultured with this protocol exhibit characteristic astrocyte morphology and a

range of specific markers, including GFAP and GS (Figure 2-1).

Figure 2-1. Characterization of primary retinal astrocytes

Astrocyte markers (I) GFAP (green), (II) GS (green) are shown in separate immunofluorescence trials in

cultured astrocytes; DAPI is used as a co-stain (blue). (III) Immuno blots of GS and GFAP in cultured

astrocytes.

We used cultured primary retinal astrocytes to collect ACM and assess its

neuroprotective effects against metabolic stress in an in vitro model, enabling high

throughput screening of involving signaling mechanisms (methods described in Chapter

Page 51: Interactome Analysis Identifies Novel Targets of

40

3). ACM was shown to be highly protective against glutamate-induced metabolic stress

in transformed hippocampal neurons (Ht22), and the effect was demonstrated to have a

concentration-dependent effect (Figure 2-2).

Figure 2-2. Assessing ACM neuroprotection against metabolic stress in Ht22 cells

(I) ACM protection in Ht22 cells against 5 mM glutamate injury is demonstrated by increased metabolic

activity of neurons pre-conditioned with ACM, compared to control media. (II) ACM activity is maintained

when diluted to 25% of its volume, beyond which the neuroprotective activity is virtually lost (n=3, **

p<0.05).

However, the combined neuroprotective effect of ACM is complex, due to the wealth of

other biologics in ACM, including growth factors. Identifying the key ligands that mediate

ACM activity can be tackled through numerous approaches, including proteomic,

genomic, and pharmacological technologies. However, due to the complexity of the

ACM mixture, ‘omic’ type analyses carry with them the potential to generate large

Page 52: Interactome Analysis Identifies Novel Targets of

41

biological datasets that are challenging to analyze. For example, preliminary data

generated by our group showed that boiling or trypsinizing ACM eliminates its

neuroprotective activity, strongly suggesting a protein component to the activity. This

thinking led us to initially carry out global mass spectroscopy and RNAseq analysis on

ACM and astrocytes, respectively. The analysis was unsuccessful due to two reasons:

1) the necessary presence of serum in ACM to maintain the protective activity, which

masked astrocyte-secreted proteins with its abundance, and 2) lack of a strong control

strategy for ranking sufficiently specific candidates. Eliminating serum from ACM,

through a series of optimization studies, failed to generate a suitable defined media

replacement.

An alternative approach is the use of chemical genetics screens using tool compounds

to identify specific targets induced by ACM. This approach has several advantages: 1)

in contrast with global screens, tool compounds, have defined targets that can elucidate

specific signaling pathways and ligands of interest. 2) Combining tool compound

libraries with robotic capabilities allow high throughput analyses of specific targets

simultaneously. 3) Tool compounds screens are modular in the sense that they can be

scaled up or down, based on experimental goals, to profile a wide range of tool

compound library sizes. 4) Finally, these screens can provide functional information, in

the sense that the primary endpoint is ACM induced protection. Thus, the use of tool

compound screens can identify necessary signaling pathways through which the

neuroprotection is transduced. Therefore, this strategy was ultimately chosen to identify

key neuroprotective signaling pathways activated by ACM.

Page 53: Interactome Analysis Identifies Novel Targets of

42

Among the screened targets are kinases, which transduce extracellular signals

efficiently through phosphorylation of downstream targets. Kinases mediate

phosphorylation reactions by transferring the gamma phosphate of Adenosine

Triphosphate (ATP) onto hydroxyl groups of various lipids, sugars or amino acids[8].

Kinases are essential players in signaling pathways and are responsible for maintaining

cellular homeostasis, survival, and growth [9]. The current project focuses on a key pro-

survival kinase, PI3K, a key mediator of astrocyte-neuron communication. Further,

identifying the binding partners of kinases such as PI3K can provide insight into the

neuroprotective signaling pathway and its activators. Analyzing these interactions in

neurons following ACM exposure can provide valuable information into how the

neuroprotection is transduced along a specific signaling pathway, such as PI3K. Thus,

proteomic analysis was employed in this project to identify how PI3K binding patterns

change in neurons following ACM exposure.

Mass spectrometry (MS) enables high throughput identification and functional

annotation of proteins in a cell, tissue, or organisms. It is an analytical technique that

measures the mass-to-charge ratio (m/z) of an ion, generated following fragmentation of

a protein sample with high energy. It is used to identify known as well as novel

macromolecules, including proteins, and provide detailed information about their

structures. Quantitative mass spectrometry (MS) provides an effective approach to

concurrent identification and determination of concentration ratios of proteins from

different samples, eliminating inter-run variability[10]. One of these labeling methods is

featured in the current work: the isobaric tag for relative and absolute

Page 54: Interactome Analysis Identifies Novel Targets of

43

quantitation (iTRAQ) technology. The technique employs isotope labeled tags that can

be covalently bonded to the N-termini and side chain amines of proteins. [11]. Due to

the isobaric design employed in iTRAQ labeling, differentially labeled peptides appear

as single peaks in MS-MS scans, which reduces the likelihood of peak

overlapping. This allows the relative quantification of proteins pooled from up to 8

different samples.

2.2 Global Hypothesis

I hypothesize that ACM mediated neuroprotection against metabolic stress is driven by

astrocyte-secreted factors. These factors activate pro-survival signaling pathways, such

as PI3K, which can be dissected and targeted to enhance neuronal survival under

stress conditions.

2.3 Purpose and aims

The purpose of the current research is to identify key signals induced by astrocyte-

secreted factors involved in protecting neurons from metabolic stress. To achieve this

goal, I will address three main aims:

1. Identify ACM-induced neuronal mechanisms, such as kinases, which can be

utilized to expand key protective signaling pathways.

a. Carry out a tool compound screen to identify key kinases and receptors

along ACM-mediated neuroprotection pathway in the glutamate sensitive

cell line Ht22.

Page 55: Interactome Analysis Identifies Novel Targets of

44

b. Validate screen hits independently in Ht22 cells in primary neurons.

c. Confirm involvement of key kinases using immunoblotting.

2. Identify ACM-regulated target interactors in neurons, including downstream

effectors and upstream receptors.

a. Generate interactome data on a candidate signaling hub protein in Ht22

cells treated with ACM using mass spectroscopy.

b. Guide bioinformatic analysis to identify ACM-regulated interactors.

c. Confirm interactions using co-immunoprecipitation and immunoblotting.

3. Validate the neuroprotective activity of candidate kinases and interactors in in

vitro and in vivo metabolic stress models.

a. Assess the effect of stimulating identified protein-protein interactions on

ACM neuroprotection.

b. Assess the impact of knocking down identified interactors on ACM

neuroprotection.

Together these aims will provide new insights into how astrocytes protect neurons from

metabolic stress through secreted factors and induced neuronal pathways.

Page 56: Interactome Analysis Identifies Novel Targets of

45

Chapter 2 References

1. Alqawlaq, S., J.G. Flanagan, and J.M. Sivak, All roads lead to glaucoma: Induced retinal injury cascades contribute to a common neurodegenerative outcome. Exp Eye Res, 2018.

2. Baldwin, K.T. and C. Eroglu, Molecular mechanisms of astrocyte-induced synaptogenesis. Current opinion in neurobiology, 2017. 45: p. 113-120.

3. Livne-Bar, I., et al., Astrocyte-derived lipoxins A4 and B4 promote neuroprotection from acute and chronic injury. J Clin Invest, 2017.

4. Hughes, E.G., S.B. Elmariah, and R.J. Balice-Gordon, Astrocyte secreted proteins selectively increase hippocampal GABAergic axon length, branching, and synaptogenesis. Molecular and cellular neurosciences, 2010. 43(1): p. 136-145.

5. Wang, G., et al., Astrocytes secrete exosomes enriched with proapoptotic ceramide and prostate apoptosis response 4 (PAR-4): potential mechanism of apoptosis induction in Alzheimer disease (AD). J Biol Chem, 2012. 287(25): p. 21384-95.

6. Livne-Bar, I., et al., Pharmacologic inhibition of reactive gliosis blocks TNF-α-mediated neuronal apoptosis. Cell Death Dis, 2016. 7(9): p. e2386.

7. Nahirnyj, A., et al., ROS Detoxification and Proinflammatory Cytokines Are Linked by p38 MAPK Signaling in a Model of Mature Astrocyte Activation. Plos One, 2013. 8(12).

8. Fabbro, D., S.W. Cowan-Jacob, and H. Moebitz, Ten things you should know about protein kinases: IUPHAR Review 14. Br J Pharmacol, 2015. 172(11): p. 2675-700.

9. Duncan, J.S., et al., Regulation of cell proliferation and survival: convergence of protein kinases and caspases. Biochim Biophys Acta, 2010. 1804(3): p. 505-10.

10. Wiese, S., et al., Protein labeling by iTRAQ: a new tool for quantitative mass spectrometry in proteome research. Proteomics, 2007. 7(3): p. 340-50.

11. Ross, P.L., et al., Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol Cell Proteomics, 2004. 3(12): p. 1154-69.

Page 57: Interactome Analysis Identifies Novel Targets of

46

Chapter 3: Interactome Analysis Identifies RNA

Binding Protein ZC3H14 as a Novel Interactor of

Phosphoinositide 3-kinase (PI3K) and a Mediator of

Astrocyte Neuroprotection

(Please note that this chapter incorporates material from a manuscript in preparation for

publication by the authors below)

Page 58: Interactome Analysis Identifies Novel Targets of

47

Interactome Analysis Identifies RNA Binding Protein ZC3H14 as a Novel Interactor of Phosphoinositide 3-kinase (PI3K) and a Mediator of Astrocyte Neuroprotection

Samih Alqawlaq, Izhar Livne-Bar, Declan Williams, Sara W Leung, Darren Chan, Anita

H Corbett, Gerold Schmitt-Ulms, Jeremy M Sivak

Candidate’s role: contributed to creating, designing and performing experiments, data

collection and analysis, as well as manuscript assembly and editing

3.1 Abstract

In a homeostatic state, astrocytes can support neuronal survival and function through a

range of secreted signals that protect against neurotoxicity, oxidative stress, and

apoptotic cascades. Thus, the analysis of astrocyte conditioned media (ACM) may

provide valuable insight into the nature of these protective mechanisms, and how they

might be promoted. Previously, we characterized a potent neuroprotective activity

mediated by ACM in neurons and the retina in metabolic stress models. However, the

molecular entity and mechanism underlying this activity remained unclear. Here, a

chemical genetics screen revealed phosphoinositide 3-kinase (PI3K) as a central player

transducing ACM-mediated neuroprotection. To identify additional proteins contributing

to the protective activity, endogenous neuronal PI3K was immunoprecipitated from

astrocytes exposed to ACM or control media, and MS/MS analyses were undertaken.

MS data analyses pointed toward only five additional proteins that co-

immunoprecipitated with PI3K and were regulated by the ACM signal. These hits

included expected PI3K interactors, such as the platelet-derived growth factor receptor

Page 59: Interactome Analysis Identifies Novel Targets of

48

A (PDGFRA), and novel interactors, such as the zinc finger CCCH-type containing 14

(ZC3H14). ZC3H14 has recently emerged as an important RNA binding protein that

modifies poly-adenosine tail lengths on nascent mRNA transcripts. In downstream

validation studies we show that Platelet Derived Growth Factor-BB (PDGF-BB) strongly

activates PI3K signaling to protect neuronal cells. Finally, PDGF-BB treatment induced

recruitment of ZC3H14 to PI3K, and inhibiting this interaction eliminated ACM-mediated

neuroprotection. Thus, we identified a novel ACM- and PDGF-induced neuroprotective

signaling cascade mediated through PI3K that involves recruitment of ZC3H14.

Enhancing this pathway may present a promising strategy to promote astrocyte-

secreted neuroprotective signals.

3.2 Introduction

Astrocytes support neuronal survival through a range of homeostatic functions,

including secretions of pro-survival factors and metabolic stress mediators. Astrocyte

conditioned media (ACM) provides a window into astrocyte secretory activities and may

introduce novel biologics for the treatment of various neurodegenerative diseases.

Using a model of primary retinal astrocytes we recently demonstrated potent

neuroprotective effects of a secreted astrocyte activity in acute and chronic metabolic

injury models, both in vitro and in vivo [1]. We defined a method for collecting ACM, and

showed that it contains a complex mixture of protein and lipid neuroprotective factors.

Yet, the protective mechanism integrating these neuroprotective signals remains

unclear. Thus, the purpose of the current study is to identify and dissect the key

signaling pathways that transduce ACM-mediated protection in neurons.

Page 60: Interactome Analysis Identifies Novel Targets of

49

ACM contains an array of neurotrophic factors (NF), which primarily activate the

Phosphoinositide 3-kinase (PI3K) signaling pathway [2-4]. PI3K mediates intracellular

signals regulating a wide range of neuronal processes, prominently including

bioenergetic modulation and protection against metabolic stress [5-7]. PI3K is activated

by binding of its p85 regulatory subunit directly to a phosphorylated receptor tyrosine

kinase (RTK), or indirectly to an adapter protein such as insulin receptor substrate-1,2

(IRS-1,2)[7]. However, the specific interactions, which modulate PI3K signaling in a

neurodegenerative context are still unclear.

Here we present a unique neuronal interactome that explores the shift in PI3K binding

preferences following ACM exposure. The analysis provides new insight into the

functions of PI3K through identification of a novel interaction with an RNA binding

protein (RBP).

3.3 Methods

3.3.1 Cell cultures

HT22 neuronal cells were cultured in DMEM-high glucose (Sigma, D6546)

supplemented with 10% FBS/1% penicillin/streptomycin [8]. Murine primary cortical

neurons were cultured as outlined [9]. Briefly, cortices from E16 mouse embryos were

isolated and cleaned in Hank's Balanced Salt Solution (HBSS) on ice. The tissue was

mechanically homogenized and dissociated with a Papain dissociation system

Page 61: Interactome Analysis Identifies Novel Targets of

50

(Worthington, LK003150). Cortical neurons were grown in Neurobasal-A media with L-

glutamine and B27 supplement without antioxidants (Thermo, 10889038).

Primary retinal astrocytes were isolated and cultured as previously described [10, 11].

Astrocytes cultured with this protocol exhibit characteristic astrocyte morphology and a

range of specific markers, including the green fibrillary acidic protein (GFAP), vimentin,

and Glutamine Synthetase (GS), and S100A. Briefly, retinas were dissected from adult

Wistar rat eyes and placed in ice-cold EMEM (Wisent,320-005-CL) supplemented with

10% FBS/1% penicillin/streptomycin. Retinas were dissociated with a Papain

dissociation system, after which they were triturated, counted and seeded in astrocyte

growth media (Lonza, CC-4123) on the first day. Media was changed the following day

to EMEM supplemented with 5% FBS, 5% Horse serum, 1% penicillin/streptomycin. At

80% confluency, cells were placed on a rotating shaker for 6 to 8 hours to remove

microglia and then re-plated in 6-well plates at 1.5 x 105 cells/well. ACM or cell-free

control medium (CFM) was harvested after 24 hours incubation and stored at –80°C in

serum-containing media.

3.3.2 PI3K inhibitor and PDGF

To induce PI3K activity, Ht22 cells were plated in 10 cm dishes in 5% FBS DMEM.

Reduced serum conditions were used to reduce signaling baseline and enhance

detection of induced phosphorylation signals [12]. The following day, Recombinant

Murine PDGF-BB (Peprotech, 315-18) was added to cells in serum-free media at 3

Page 62: Interactome Analysis Identifies Novel Targets of

51

different concentrations: 0.5, 5, 50 ng/mL. One hour later, cell lysates were collected in

NP40 lysis buffer, containing 150 mM Tris, 150 mM NaCl, in addition to

protease/phosphatase inhibitor cocktail. Various kinase inhibitors were added to cells at

over a range of concentrations, using IC50 values as guideline.

3.3.3 Kinase inhibitor library screening

In order to identify key neuronal signaling components of this protective cascade, we

used our in vitro platform to perform a chemical genetics kinase inhibitor screen

following treatment with ACM or CFM. More specifically, a library of kinase inhibitors

was used to inhibit the activity of ACM and measure the subsequent change in

neuroprotection in Ht22 cells. The study was carried out at The High-Throughput

Screening Division, University Health Network, Toronto, Ontario. A 480 tool compound

kinase inhibitor library (BIOMOL) was screened in 384 well plates at 1M, seeded with

1x103 per well, using either ACM or CFM incubated under identical conditions. Cells

received a 5mM glutamate challenge, and their survival was assessed 20 hours later

using the XTT Cell Proliferation Assay Kit (Roche, 11465015001). Selection of hit

compounds was based on two criteria: 1) the compound reduced ACM neuroprotection

by at least 10%, and 2) did not have an inherent toxic effects (less than 10% reduction

in survival) on cells.

Page 63: Interactome Analysis Identifies Novel Targets of

52

3.3.4 Cell Viability Assay

Validating top hits from the screen was carried out using a similar viability assy. Briefly,

5 x 10 3 Ht22 cells, or 2 x 104 primary cortical neurons were seeded in 96-well plates;

the following day, cells were pre-treated with ACM or CFM for 3 hours, inhibitors were

then added to the cells an hour before receiving a 5mM glutamate challenge. 20 hours

later, viability was assessed using the XTT proliferation kit.

3.3.5 Immunoprecipitations

Ht22 cells were plated in 15 CM2 dishes at 70% confluency. Following a 1 hr incubation

with either ACM or CFM, cells were first fixed with 0.5% formalin solution in PBS for 10

minutes to preserve labile and transient protein-protein interactions. Formalin

neutralization was completed with 150 mM Tris base, 150 mM Glycine aolution for 10

mins. Cell lysates were collected in 0.2% NP40 lysis buffer, containing 150 mM Tris,

150 mM NaCl, in addition to protease/phosphatase inhibitor cocktail.

A library of PI3k antibodies (CST, 9655) targeting different subunits were screened to

choose the antibody with the most efficient capture efficiency (Supplementary Table 1).

25 μL of each antibody was incubated with 300 μL of fixed lysates at 4C overnight, after

which 25 μL of Protein A Sepharose slurry (GE Healthcare, CL-4B) was added to the

mixture. Following a 3-hour incubation at 4C, the IP reaction was centrifuged for 3

minutes at 1000 RPM, followed by one wash with 500 mM NaCL and two washes with

lysis buffer. Beads were eluted using 0.2% Trifluoroacetic acid (TFA) and 20%

Page 64: Interactome Analysis Identifies Novel Targets of

53

Acetonitrile solution. An aliquot of lysate input, IP supernatant fraction (unbound), and

eluate was saved for confirmation by western blotting. To generate a negative control

for the affinity capture, the optimal antibody was pre-incubated with its blocking peptide

at 1:1 v/v ratio for 2 hours at room temperature, and an IP was carried out as described

above. A complimentary ZC3H14 IP was carried out using ab169061.

3.3.6 Mass spectroscopy

The preparation and analysis of immunoaffinity preparations followed previously

published methods (Wang et al. eLife 2017;6:e28401, Gunawardana et al. Mol Cell

Proteomics 2015; 14(11):3000-14). Affinity capture eluates were dried in a centrifugal

evaporator to remove the acetonitrile and trifluoroacetic acid. Next, protein samples

were denatured with 9M urea at room temperature. Subsequently, 1 M

tetraethylammonium bicarbonate (TEAB) was added to adjust the pH to 8.0, followed by

cysteine reduction for 30 min at 60°C at 5 mM tris(2-carboxyethyl) phosphine. Protein

sulfhydryl groups were then alkylated for 1 hour at room temperature in the presence of

10 mM 4-vinylpyiridine. The urea concentration was decreased below 1.5 M by dilution

with 500 mM TEAB then protein digestion was affected with porcine trypsin (Thermo

Fisher Scientific, Burlington, ON, Canada) at 37°C overnight. Isobaric labeling of the

tryptic digests with an iTRAQ 8plex reagent set (Sciex, Concord, ON, Canada) was

performed according to the manufacturer’s protocol. The iTRAQ modified digests were

pooled and the mixture purified in parallel on C18 Bond Elut OMIX columns (Agilent

Technologies, Santa Clara, CA, USA) at pH 2 and with a high pH reversed-phase

peptide fractionation kit (Thermo Fisher Scientific, Waltham, MA, USA) from which

Page 65: Interactome Analysis Identifies Novel Targets of

54

fractions were collected at 12.5%, 17.5%, 22.5% and 50% acetonitrile. All purified

samples were dried in a centrifugal evaporator then reconstituted in water containing

5% acetonitrile and 0.1% formic acid then analyzed by (Thermo Fisher Scientific).

Peptide sequence information was generated from MS and MS2 scans while

quantitative iTRAQ reporter ion information was generated from MS3 scans of the ten

most abundant product ions in each MS2 spectrum. The MS, MS2 and MS3 scans

were acquired in the Orbitrap, linear ion trap and Orbitrap mass analyzers respectively.

The time allowed between MS scans was fixed to a maximum of 3 seconds, during

which the maximum number of MS2 and MS3 spectra were collected. Mass spectra

were collected during 260 minute liquid chromatography runs having acetonitrile content

increasing from 0 to 30% in 180 minutes then to 100% in 60 minutes then remaining at

100% for 20 minutes. The 25 cm long, 75 micrometer inner diameter analytical column

contained Acclaim PepMap RSLC C18 particles of 2 micrometer diameter with 100 Å

pores, and was operated at 300 nl/minute.

3.3.7 Protein identification and analyses

Protein identification and quantification was done by Mascot (Version 2.4; Matrix

Science Ltd, London, UK) and Sequest HT search engines within Proteome Discoverer

software (Version 1.4; Thermo Fisher Scientific) as well as PEAKS studio 8.5

(Bioinformatics Solutions Incorporated, Waterloo, ON, Canada) using the human

international protein index (IPI) database (Version 3.87). Protein levels were normalized

to one ACM replicate (ACM3), which was used as an internal reference.

Page 66: Interactome Analysis Identifies Novel Targets of

55

To determine the extent to which PI3k interactions changed as a results of ACM

treatment, the data was filtered over three main stages: 1. Interactors with a

negative/ACM3 ratio>0.35 were filtered out to eliminate non-specific interactions. 2.

Interactors were then included on the basis of having at least a 0.8 ACM:ACM3 ratio,

which was set as an indicator of high internal reliability between ACM triplicates. 3.

Among high reliability candidates, an ACM-induced interactor was defined as a protein

with a CFM: ACM ratio of either >1.35 (downregulated) or > 0.65 (upregulated).

Identifying known interactors was done using the STRING database (http://string-

db.org), which provides an assessment and integration of protein-protein interactions,

including direct (physical) as well as indirect (functional) associations [13].

3.3.8 Immunoblotting

Cell lysates for western blots were collected in either RIPA buffer for denaturing

applications (Cell Signaling), or NP-40 (described above) for non-denaturing

applications, supplemented with protease/phosphatase inhibitor cocktail. Protein

concentrations were quantified using Bicinchoninic acid assay (BCA) (Thermo, 23225)

and equal amounts were loaded in SDS-PAGE. Proteins were transferred to

polyvinylidene fluoride (PVDF) membrane and blocked with 5% bovine serum albumin

in Tris-buffered saline with 0.1% Tween 20 (TBST), and incubated overnight at 4 °C with

the primary antibody. probed with antibodies raised PI3 Kinase p85 (CST, 4257), pan

AKT (CST,2920), Phospho-Akt (Ser473) (CST, 4060), GAPDH (Calbiochem), ZC3H14

[14], and THOC1 (Abcam, ab487). Membranes were then washed three times with

Page 67: Interactome Analysis Identifies Novel Targets of

56

TBST and probed with appropriate IRDye secondary antibody (Li-Cor Biosciences,

Lincoln, NE). Blots were imaged and analyzed with an Odyssey infrared imaging system

(Li-Cor Biosciences), with each band being normalized to internal control.

3.3.9 SiRNA transfection

Stealth RNAi (Thermo) was used to deplete ZC3H14 in cultured Ht22 cells. Cells were

plated at a density of 1.5 × 105 cells per well in six-well plates in 10% FBS DMEM. Each

well received 4 µl of Lipofectamine 2000 (Thermo, 11668027), diluted in 150 µl of

Optimem media (Thermo); siRNA was also diluted in 150 µL of Optimem media.

Lipofectamine 2000-SiRNA complexes were combined with 1200 µl of serum free

DMEM, and left on for 6 hours. Next, medium containing transfection complexes was

replaced by fresh 10% FBS DMEM. ZC3H14 knockdown was evaluated by

immunoblotting hours following transfection. Ht22 cells were also transfected with

scrambled negative control – medium GC content (Thermo) or ZC3H14-1,

TGACTGACCTGAGTGTGGCACAGAA . The sequence of the ZC3H14-1 siRNA was

adopted from [15]; a predesigned siRNA was tested in parallel for optimization.

3.3.10 Statistics

For all experiments, n refers to the number of animals or biological replicates. Statistical

analyses were performed by t test or 1-way ANOVA with Tukey’s post hoc analyses.

Page 68: Interactome Analysis Identifies Novel Targets of

57

3.4 Results

3.4.1 ACM neuroprotection is mediated through PI3K

In order to identify kinase signaling that is necessary to transduce ACM-mediated

neuroprotection signals, we designed a chemical genetics robotics screen (Figure 3-

1A). The kinase screen cell results show a clear separation in viability between ACM

and CFM treated samples (Figure 3-1B), demonstrating that most of the 480 kinase

inhibitors had no effect on ACM induced protection at 1M. Remarkably, out of the total

library only 10 kinase inhibitors met the selection criteria, previously defined. Six out of

the top 10 compound hits targeted the PI3k/AKT pathway, strongly implicating a role for

PI3k in transducing the protective ACM effect (Figure 3-1C). Other single-hit kinase

inhibitors targeted receptor tyrosine kinases, cyclin-dependent kinases (CDKs),

Ca2+/calmodulin-dependent protein kinase (CaMk), and the Mitosis inhibitor protein

kinase Wee1.

The effect of PI3K inhibition, and downstream targets, was verified independently in

Ht22 cells and in primary cortical neurons. PX866, (IC50: 0.1-88 nM) [16]) a potent open

ring analogue of Wortmannin [17], reduced ACM-mediated neuroprotection by more

than 80% in Ht22 cells, without causing additional cytotoxicity (Figure 3-1D). We also

reproduced the ACM neuroprotective effect in primary cortical neurons, and verified that

the PI3K inhibitor, ZSTK474 (IC50: 0.7 nM-210 nM)[18, 19], and the AKT inhibitor

GSK690693(IC50: 20 nM-890 nM) [20], similarly blocked the activity (Figure 3-1E).

Next, we assessed whether ACM treatment is sufficient to activate the PI3K pathway by

immunoblotting for AKT phosphorylation following ACM addition in Ht22 cells. ACM

Page 69: Interactome Analysis Identifies Novel Targets of

58

rapidly mediated AKT phosphorylation in neurons as reflected by a significantly

increased AKT phosphorylation signal by 0.5 hr following ACM addition, and still

detectable up to 4 hours later (Figure 3-1F,G).

Page 70: Interactome Analysis Identifies Novel Targets of

59

Figure 3-1. PI3K is necessary for ACM-mediated neuroprotection.

(A) Overview of chemical genetics screen. (B) Scatter blot of XTT absorbance values for 480

kinase inhibitors screened under ACM (red) and CFM (black) conditions in Ht22 cells. A clear

separation is observed between treatment conditions for most compounds, indicating general

ACM protection compared to CFM. (C) Hits were considered inhibitors that reduced ACM

protective activity, but did not affect CFM conditions. Of the top 10 hits, 7 targeted the PI3K/AKT

pathway. (D) As validation, PX866, an open ring analogue of the PI3K inhibitor Wortmannin,

was used to inhibit ACM activity in HT22 cells (n=3). (E) Similarly, the PI3K inhibitor ZSTK474,

and AKT inhibitor GSK690693 each effectively block ACM activity in murine primary cortical

neurons (n=3). (F,G) ACM treatment induces rapid phosphorylation of AKT as early as 0.5

hours, a signal that can be observed up to 4 hours after later (n=3)(**p < 0.01; bars are S.E.M).

Page 71: Interactome Analysis Identifies Novel Targets of

60

3.4.2 PI3K immunoprecipitation

PI3K activation can initiate pleotropic signaling through a variety of described pathways

[21, 22]. However, identifying downstream signaling nodes provides insight into the

precise functional outcome of ACM-mediated neuroprotection. In order begin unraveling

this signaling cascade we initiated a study to immunoprecipitated PI3K for the purpose

of generating an interactome of the ACM-induced signaling complex compared to

control CFM. This was achieved by designing a strategy to immunoprecipitate

endogenous PI3K in crosslinked Ht22 cell lysates, following treatment with ACM or

CFM, and analyzing co-precipitates using mass spectroscopy (Figure 3-2A). As

endogenous PI3K interactomes have not been well described in neuronal cells, an

antibody panel was first probed for their ability to effectively deplete and elute PI3K from

crosslinked Ht22 cell lysates (Supplementary Figure 1A). Ab2, which targets the p85

subunit of PI3K, was chosen as it produced the strongest PI3K immunodepletion in the

unbound fraction, combined with the strongest elution (Supplementary Figure 1B). In

order to differentiate true PI3k interactors from non-specific binders, a negative control

was generated by carrying out a parallel immunoprecipitation by pre-incubating Ab2

with its blocking peptide. Under this control condition there was minimal depletion of

PI3k in the unbound fraction, and a reduced PI3k elution band (Figure 3-2B).

Subsequent coomassie staining of blots shows highly specific capture of PI3k, as

reflected by generally clean eluate lanes compared to the corresponding input and

unbound lanes (Figure 3-2B).

Page 72: Interactome Analysis Identifies Novel Targets of

61

Figure 3-2. A PI3K interactome yields highly specific capture of ACM induced proteins.

(A) A schematic of the interactome experimental design: Cell lysates were isolated from Ht22 cells treated

with ACM (n=3), CFM (n=3). Lysates were lightly fixed and submitted to immunoprecipitation of

endogenous PI3K, along with an additional negative control with a PI3K blocking peptide. Covalent

modifications of primary amines in all 7 samples were done with iTRAQ labeling, after which samples

were pooled to be analyzed concurrently by MS/MS. (B) Following a PI3K IP screen, Ab2 generated

depletion of PI3K in the unbound fraction, and produced a strong band in the eluted fraction. An Ab2

blocking peptide inhibited the capture of PI3K as reflected by no depletion in the unbound fraction, and a

reduced PI3K band in the eluted fraction. (C) A coomassie stain of the blot indicates PI3K capture was

highly specific as shown by a relatively clear eluate lane compared to the input and unbound lanes. (D)

Box plots showing Log2 fold change of PI3K regulatory subunit count intensities in all conditions,

normalized to ACM3, which was chosen as internal control. The peptide was abundant in all ACM and

CFM conditions, but very low in the negative control, as expected, confirming specificity of affinity capture

and negative control performance.

Page 73: Interactome Analysis Identifies Novel Targets of

62

3.4.3 ACM induced interactome identification

Proteome Discoverer sequencing of the LC-MS data from PIK3R1 immunoprecipitates

identified 293 proteins with 12117 peptide-to-spectrum matches (PSMs) at or exceeding

95% confidence. An inclusion criterion of at least 2 spectral counts was established to

select quantified proteins, while proteins with less than 2 assigned spectral counts were

considered unquantified. The protein list was sorted based on abundance, as

determined by PSM (Table 1). Two major observations were made that highlight the

selectivity of the affinity capture: 1) PI3k subunits, both catalytic and regulatory, were

the most abundant in the interactome with more than 2000 PSMs for each (Table 1),

indicating highly specific immunoprecipitation of PI3K, and 2) the abundance level of

PI3K subunits in the negative control was at least 6 folds lower than that of other

experimental conditions (Figure 3-2C). PIK3CA and PIK3CB were the second and third

most abundant proteins based on spectral counting respectively, with over 2000 PSMs

each, indicating that the PI3K complex was highly purified (Table 1).

Page 74: Interactome Analysis Identifies Novel Targets of

63

Table 1. Top 20 PI3K interactome hits, sorted by spectral counts in 6 samples. The ratio

columns represent average iTRAQ intensity ratios observed for a given protein. Hits

were filtered to include proteins with a minimum 0.8 ratio to the internal reference

ACM3, to ensure high reliability, and 0.4 negative control/ACM ratio to eliminate non-

specific binders. Note that the most abundant hits are the regulatory (p85) and catalytic

(p110) PI3K subunits.

Accession Description PSM CFM1/ ACM3

ACM1/ ACM3

CFM2/ ACM3

ACM2/ ACM3

CFM3/ ACM3

negative ctrl/ACM3

IPI00263878.2 Phosphatidylinositol 3-kinase regulatory subunit alpha

2821 0.826 0.896 0.995 0.891 0.938 0.106

IPI00136110.4 Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta isoform

2108 0.83 0.885 1.078 0.804 0.926 0.114

IPI00331708.3 Isoform 1 of MKL/myocardin-like protein 1

1551 0.9 0.858 1.074 0.906 1.016 0.069

IPI00323357.3 Heat shock cognate 71 kDa protein

735 0.99 0.853 1.126 0.822 0.904 0.133

IPI00119627.1 Insulin receptor substrate 1 488 0.69 0.866 0.846 0.861 0.734 0.124

IPI00379844.5 Insulin receptor substrate 2 359 0.753 0.885 0.934 0.851 0.744 0.278

IPI00117159.2 Phosphatidylinositol 3-kinase regulatory subunit beta

171 0.681 0.84 0.925 0.98 0.926 0.059

IPI00230632.2 Isoform Cas-A of Breast cancer anti-estrogen resistance protein 1

148 1.152 0.954 1.204 0.964 1.04 0.289

IPI00406794.2 GRB2-associated-binding protein 1

106 0.718 0.847 0.785 0.892 0.603 0.097

IPI00319992.1 78 kDa glucose-regulated protein

102 0.858 0.804 1.064 0.806 0.788 0.075

IPI00554929.3 Heat shock protein HSP 90-beta

91 0.865 0.843 0.942 0.868 0.733 0.18

IPI00223902.2 Isoform 2 of Uncharacterized protein KIAA1310

66 0.84 1.052 0.965 1.139 0.874 0.013

IPI00987441.1 Uncharacterized protein (Fragment)

60 1.497 1.028 1.233 1.085 1.724 0.292

IPI00139780.1 60S ribosomal protein L23 42 0.696 0.832 0.883 0.866 0.788 0.072

IPI00323806.4 Putative uncharacterized protein

33 0.935 0.937 1.156 0.865 0.94 0.23

IPI00653962.1 Uncharacterized protein 30 1.246 0.906 1.809 0.816 1.94 0.089

IPI00762542.2 40S ribosomal protein S11 29 0.897 0.917 0.881 0.842 0.753 0.108

IPI00465880.4 40S ribosomal protein S17 27 0.706 0.922 0.963 0.949 0.558 0.164

IPI00466258.2 Isoform 1 of SH3 domain-containing kinase-binding protein 1

25 0.786 0.989 1.009 0.934 0.556 0.126

IPI00986371.1 60S ribosomal protein L27a-like

20 1.679 0.8 1.379 0.83 0.846 0.258

Page 75: Interactome Analysis Identifies Novel Targets of

64

Additional bioinformatic analyses further narrowed down the list of interactors to achieve

three main criteria: 1) capturing quantifiable hits with at least 2 PSMs, 2) filtering out

non-specific binders using the negative control as a reference, and 3) including

reproducible hits across the three triplicates. A total of 122 proteins were quantified from

three or more MS3 spectra having reporter ion signals representing all six PIK3R1

immunoprecipitates (all three CFM treated replicates and all three ACM treated

replicates). Of these, 90 proteins had negative control/ACM3 iTRAQ ratios of less than

0.35, indicating that they were co-enriched with PI3KR1 specifically. Further, only 26

candidate PI3KR1 interactors fit the reproducibility criteria of having median reporter ion

signals varying less than 20% among all three ACM replicates (Figure 3-3A).

Page 76: Interactome Analysis Identifies Novel Targets of

65

Figure 3-3. A summary of ACM-regulated PI3K interactions.

(A) Overview of PI3K interactors and subsequent bioinformatic analysis. (B) A total of five PI3K

protein-protein interactions were upregulated following ACM treatment. The receptor tyrosine

kinase Pdgfra, and adaptor proteins Ywhaz and Ywhae have been previously established as

PI3K interactors through a variety of sources. In addition two novel ACM induced interactors

were detected; the poly-A RBPs ZC3H14 and THOC1.

Beyond identifying specific PI3K interactors, we also investigated how PI3K binding

preferences shifted following ACM treatment. Thus, we investigated interactors that, in

addition to having high reproducibility and specificity ratios (previously defined), have a

CFM/ACM ratio between 0.65 and 1.35, indicating ACM regulated binding with PI3K.

Remarkably, a total of only 5 interactors met these criteria, all showing increased

binding with PI3K following ACM treatment (Table 2). Three ACM-induced interactors

Page 77: Interactome Analysis Identifies Novel Targets of

66

are known based on established literature, including the platelet-derived growth factor

receptor alpha (PDGFRA), and two 14-3-3 adaptor proteins [23, 24]. However, two of

the identified ACM-induced binders are novel, previously unknown PI3k interactors

(Figure 3-3B). These novel interactors include the Isoform 1 of Zinc finger CCCH

domain-containing protein 14 (Zc3h14) and Isoform 1 of THO complex subunit 4

(Thoc1). Intriguingly, both of these novel interactors are involved in mRNA

polyadenylation and processing.

Page 78: Interactome Analysis Identifies Novel Targets of

67

Table 2 List of ACM-induced PI3K interactors and corresponding CFM:ACM fold

changes

Accession Description PSM

Sum of Peptides

CFM1/ ACM3

CFM2/ ACM3

CFM3/ ACM3

Average fold

change Negative/ACM

IPI00461416.4 Isoform 1 of Zinc finger CCCH

domain-containing protein 14 (Zc3h14)

29 99 0.48 0.56 0.54 0.53 0.186

IPI00844650.1 Isoform 1 of Alpha-

type platelet-derived growth factor

receptor (Pdgfra)

24 70 0.62 0.52 0.45 0.53 0.392

IPI00114407.2 Isoform 1 of THO

complex subunit 4 (Thoc1)

15 60 0.31 0.44 0.38 0.38 0.03

IPI00116498.1 14-3-3 protein

zeta/delta (Ywhaz)

7 52 0.42 0.51 0.42 0.45 0.129

IPI00118384.1 14-3-3 protein epsilon (Ywhae)

6 41 0.42 0.51 0.43 0.45 0.143

Page 79: Interactome Analysis Identifies Novel Targets of

68

3.4.4 ZC3H14 complexes with PI3k

Among the ACM-regulated interactors is the RBP ZC3H14, which we report here as a

PI3K interactor for the first time. ZC3H14 is an RBP that has been shown to stabilize

messenger RNA(mRNA) transcripts through modifying their poly-Adenosine (polyA) tail

length [25-27]. Thus, observing PI3K-ZC3H14 interaction fits with the role of PI3K/AKT

pathway in promoting cell survival through boosting protein synthesis [2, 28]. To confirm

the interaction between PI3k and ZC3H14 biochemically, Co-Ip were carried out by

immunoprecipitating PI3K and probing for ZC3H14 (Figure 3-4A). ZC3H14 has three

known isoforms; isoform 1, which was identified in the interactome, as well as isoform 2

and 3 all co-immunoprecipitated with PI3k. In order to confirm the identity of the eluted

band, ZC3H14 was knocked down (Figure 3-4B). A separate PI3k IP was carried out in

ZC3H14 knockdown Ht22 cell lysates compared to scrambled control, and followed by

probing for ZC3H14. Depletion of ZC3H14 led to loss of the eluted band (Figure 3-4C).

Finally, a reverse Co-Ip of the two proteins was carried out by immunoprecipitating

ZC3H14, followed by probing for PI3K p85, which further supported the interaction

(Figure 3-4D).

Page 80: Interactome Analysis Identifies Novel Targets of

69

Figure 3-4. ZC3H14 is a novel PI3K interactor.

(A) Co-immunoprecipitation (Co-Ip) of PI3K was carried out in Ht22 cells and probed with an

antibody to ZC3H14 to verify interaction between the two proteins (I). Successful IP of PI3K was

confirmed by blotting for PI3K (II). (B) The efficiency of knockdown was verified by

immunoblotting for ZC3H14 in corresponding lysates, compared to control. (C) ZC3H14 was

knocked down in Ht22 cells. Eluates from the IP in knockdown lysates show a missing ZC3H14

band, compared to control. (D) As a further validation for the interaction, a reverse Co-IP was

carried out by immunoprecipitating ZC3H14 and probing for PI3K. Successful ZC3H14 IP is

demonstrated by immunoblotting for ZC3H14.

Page 81: Interactome Analysis Identifies Novel Targets of

70

3.4.5 PDGF induces neuroprotective PI3K recruitment of ZC3H14

The MS/MS interactome results suggested that PDGFRA binding to PI3K was

increased following ACM addition. To validate this finding, we first investigated whether

exogenous PDGF activates the PI3K pathway in HT22 cells. Addition of recombinant

PDGF-BB rapidly induced robust AKT phosphorylation at 5 and 50ng/mL, indicating

activation of the PI3k pathway (Figure 3-5A). This is consistent with the ACM-induced

activation previously shown in Figure 1E, and is consistent with PDGF-BB as a

neuroprotective component of ACM. Next, we investigated whether recombinant PDGF-

BB can account for the neuroprotection of ACM in Ht22 cells. Thus, we returned to the

glutamate injury model previously described for the chemical genetics screen. The

experiment demonstrated a robust protection effect mediated by recombinant PDGF-BB

at 50 ng/mL (Figure 3-5B).

Since PDGFRA and ZC3H14 were both components of the PI3K complex in the ACM

interactome, we investigated whether PDGF treatment affects PI3k-ZC3H14 binding. A

PI3K Co-IP was carried out, as previously described, following 20 ng/mL PDGF-BB and

vehicle treatment. For each IP, equal concentrations of cell lysate, antibody, and beads

were used. The resulting blots demonstrate a dramatic increase in ZC3H14 elution. This

observation is supported by complimentary ZC3H14 depletion in the unbound fraction of

PDGF treated cells compared to the vehicle control (Figure 3-5C). In comparison, the

PI3K depletion and elution were consistent for each condition (Figure 3-5C). As a

functional validation, knocking down ZC3H14 rendered Ht22 cells insensitive to PDGF-

Page 82: Interactome Analysis Identifies Novel Targets of

71

mediated neuroprotection (Figure 3-5D). Together, these data suggest PDGF treatment

increases PI3K recruitment of ZC3H14 to mediate the neuroprotective signal.

Figure 3-5. PDGF is enriched in ACM and induces PI3K recruitment of ZC3H14.

(A) Recombinant PDGF treatment produces a strong p-akt signal in Ht22 cells at 5 ng/mL and

50 ng/mL, indicating PI3K pathway activation. (B) A robust protection effect is mediated by

recombinant PDGF-BB at 50 ng/mL against glutamate-induced metabolic stress (n=3). (C)

PDGF treatment leads to increased association between PI3K and ZC3H14 in coIP eluates.

Following PI3K IP the ZC3H14 band is increased in PDGF treated cells compared to control.

Immunoblotting of PI3K was also carried out to confirm equal amounts of captured PI3K in both

conditions. (D) ZC3H14 knockdown eliminates PDGF-mediated neuroprotection against

glutamate injury in Ht22 cells (n=3)(**p < 0.01; bars are S.E.M).

Page 83: Interactome Analysis Identifies Novel Targets of

72

3.5 Discussion

Analysis of ACM is an established approach to investigating astrocyte-neuron

interactions and identifying novel astrocyte-derived neuroprotective factors against

metabolic injuries [1, 29-31]. This study used a combination of chemical genetics

screening and mass spectroscopy to identify key signaling pathways and ligands

involved in ACM-mediated neuroprotection against metabolic stress. The chemical

genetics screen identified PI3K as the main signaling hub for ACM activity; we further

validated this finding in primary cortical neurons. Following a highly specific PI3K

immunoprecipitation in crosslinked Ht22 lysates, we used mass spectroscopy to

generate a PI3K interactome with the goal of identifying upstream effectors and

downstream targets. The interactome lead to the identification of a novel PI3K-ZC3H14

interaction, which was upregulated by ACM addition in Ht22 cells. Further, the

interactome showed that PI3K-PDGFr interaction was also upregulated by ACM,

strongly implicating PDGF as a key ligand in ACM-mediated neuroprotection. Follow up

validation studies showed that PDGF protected neurons from glutamate metabolic

stress in vitro, and increased PI3K recruitment of ZC3H14. Finally, we showed that

ZC3H14 knockdown eliminated PDGF-mediated neuroprotection, which highlights the

importance of ZC3H14 in coordinating neuroprotective signals against metabolic stress.

PDGF is secreted primarily by macroglia, including astrocytes and retinal Muller cells,

as well as by neurons, mediating short-range paracraine communication between

astrocytes and neurons [32]. It has also been shown to protect neurons against

oxidative stress and NMDA-induced metabolic stress in primary neurons and retinal

Page 84: Interactome Analysis Identifies Novel Targets of

73

ganglion cells [33-37]. Activation of the PI3K-AKT pathway with NFs such as PDGF is

known to promote cell survival through inactivation of apoptotic factors [6, 38], activation

of metabolic regulators [39, 40], and activation of mammalian target of rapamycin

(mTOR), which leads to increased translation of growth proteins and further anti-

apoptotic signaling [38, 41]. Less known is PI3K’s protective role through interacting

with a range of RBPs, with the adapter protein 14-3-3 functioning as a mediator,

ultimately leading to stabilizing of mRNA transcripts [42, 43].

RBPs play an important role in post-transcriptional gene regulation through physical

interactions with adenine-(or adenosine) and uridine-rich elements of RNA [44, 45]. An

example of these RBPs is ZC3H14, which binds polyadenosine (polyA) tails and carries

out a range of post-transcriptional modifications on premature transcripts [46, 47].

These include control of PolyA tail length [27], nuclear export [48], and mRNA splicing

[49]. A 2018 study reported that ZC3H14 interacts with THO complex 1 (THOC1) to

coordinately control RNA processing, poly(A) tail length, and consequently mRNA

stability [25]. THOC1 was among the ACM-regulated novel PI3K interactors in our

screen. Morris and Corbett also showed that mRNA targets of ZC3H14 and THOC1

include the postsynaptic density protein 95 (Psd95), ATP synthase lipid-binding protein

(Atp5g1) and microtubule-associated protein tau (MAPT), which are integral to neuronal

function and metabolism. Further, altering ZC3H14 expression impairs neural function in

Drosophila and has been associated with cognitive deficits in humans [50], anatomical

changes in mouse brain ventricles as well as working memory deficits [26]. Thus, our

Page 85: Interactome Analysis Identifies Novel Targets of

74

findings provide a novel insight into the role of PI3K and ZC3H14 in regulating neuronal

function and survival (Figure3- 6).

Figure 3-6. Summary of the mechanisms of ACM neuroprotection against

metabolic stress.

(1) PDGF, a component of ACM, binds to PDGFr in neurons, (2) phosphorylating PI3K,

which in turn binds to the RBP ZC3H14. (3) Activated ZC3H14 then binds with target

mRNAs’ polyA tails, (4) leading to increased stability of pro-survival and metabolic

mRNAs.

Future studies will investigate whether ACM-mediated neuroprotection can be

linked to the stability of mRNA targets of ZC3H14 and THOC1. Exploring this novel

interaction further will provide a better understanding of how astrocytes communicate

with neurons to maintain their survival. Further, it may ultimately lead to the

development of novel therapeutic approaches for the treatment of neurodegenerative

diseases.

Page 86: Interactome Analysis Identifies Novel Targets of

75

3.6 ACKNOWLEDGEMENTS

CIHR, NSERC, TWGH Foundation Glaucoma Research Chair (JS), VSRP Program

(SA).

Page 87: Interactome Analysis Identifies Novel Targets of

76

Chapter 3 references

1. Livne-Bar, I., et al., Astrocyte-derived lipoxins A4 and B4 promote neuroprotection from acute and chronic injury. J Clin Invest, 2017.

2. Ward, P.S. and C.B. Thompson, Signaling in control of cell growth and metabolism. Cold Spring Harb Perspect Biol, 2012. 4(7): p. a006783.

3. Laurino, L., et al., PI3K activation by IGF-1 is essential for the regulation of membrane expansion at the nerve growth cone. J Cell Sci, 2005. 118(Pt 16): p. 3653-62.

4. Zhang, H., et al., PDGFRs are critical for PI3K/Akt activation and negatively regulated by mTOR. The Journal of clinical investigation, 2007. 117(3): p. 730-738.

5. Koh, S.H. and E.H. Lo, The Role of the PI3K Pathway in the Regeneration of the Damaged Brain by Neural Stem Cells after Cerebral Infarction. J Clin Neurol, 2015. 11(4): p. 297-304.

6. Carnero, A., et al., The PTEN/PI3K/AKT signalling pathway in cancer, therapeutic implications. Current Cancer Drug Targets, 2008. 8(3): p. 187-198.

7. Vanhaesebroeck, B., et al., The emerging mechanisms of isoform-specific PI3K signalling. Nat Rev Mol Cell Biol, 2010. 11(5): p. 329-41.

8. Fukui, M., et al., Mechanism of glutamate-induced neurotoxicity in HT22 mouse hippocampal cells. Eur J Pharmacol, 2009. 617(1-3): p. 1-11.

9. Hilgenberg, L.G. and M.A. Smith, Preparation of dissociated mouse cortical neuron cultures. J Vis Exp, 2007(10): p. 562.

10. Livne-Bar, I., et al., Pharmacologic inhibition of reactive gliosis blocks TNF-α-mediated neuronal apoptosis. Cell Death Dis, 2016. 7(9): p. e2386.

11. Nahirnyj, A., et al., ROS Detoxification and Proinflammatory Cytokines Are Linked by p38 MAPK Signaling in a Model of Mature Astrocyte Activation. Plos One, 2013. 8(12).

12. Kaplan-Albuquerque, N., et al., Patterns of gene expression differentially regulated by platelet-derived growth factor and hypertrophic stimuli in vascular smooth muscle cells: markers for phenotypic modulation and response to injury. J Biol Chem, 2005. 280(20): p. 19966-76.

13. Szklarczyk, D., et al., STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res, 2015. 43(Database issue): p. D447-52.

Page 88: Interactome Analysis Identifies Novel Targets of

77

14. Leung, S.W., et al., Splice variants of the human ZC3H14 gene generate multiple isoforms of a zinc finger polyadenosine RNA binding protein. Gene, 2009. 439(1-2): p. 71-8.

15. Apponi, L.H., et al., Loss of nuclear poly(A)-binding protein 1 causes defects in myogenesis and mRNA biogenesis. Hum Mol Genet, 2010. 19(6): p. 1058-65.

16. Ihle, N.T., et al., Molecular pharmacology and antitumor activity of PX-866, a novel inhibitor of phosphoinositide-3-kinase signaling. Mol Cancer Ther, 2004. 3(7): p. 763-72.

17. Le Cras, T.D., et al., Inhibition of PI3K by PX-866 prevents transforming growth factor-alpha-induced pulmonary fibrosis. Am J Pathol, 2010. 176(2): p. 679-86.

18. Zask, A., et al., Discovery of 2-ureidophenyltriazines bearing bridged morpholines as potent and selective ATP-competitive mTOR inhibitors. Bioorg Med Chem Lett, 2010. 20(8): p. 2644-7.

19. Rewcastle, G.W., et al., Synthesis and biological evaluation of novel analogues of the pan class I phosphatidylinositol 3-kinase (PI3K) inhibitor 2-(difluoromethyl)-1-[4,6-di(4-morpholinyl)-1,3,5-triazin-2-yl]-1H-benzimidazole (ZSTK474). J Med Chem, 2011. 54(20): p. 7105-26.

20. Heerding, D.A., et al., Identification of 4-(2-(4-amino-1,2,5-oxadiazol-3-yl)-1-ethyl-7-{[(3S)-3-piperidinylmethyl]oxy}-1H- imidazo[4,5-c]pyridin-4-yl)-2-methyl-3-butyn-2-ol (GSK690693), a novel inhibitor of AKT kinase. J Med Chem, 2008. 51(18): p. 5663-79.

21. Leevers, S.J., B. Vanhaesebroeck, and M.D. Waterfield, Signalling through phosphoinositide 3-kinases: the lipids take centre stage. Curr Opin Cell Biol, 1999. 11(2): p. 219-25.

22. Vanhaesebroeck, B. and M.D. Waterfield, Signaling by distinct classes of phosphoinositide 3-kinases. Exp Cell Res, 1999. 253(1): p. 239-54.

23. Maheswaranathan, M., et al., Platelet-derived growth factor (PDGF) regulates Slingshot phosphatase activity via Nox1-dependent auto-dephosphorylation of serine 834 in vascular smooth muscle cells. J Biol Chem, 2011. 286(41): p. 35430-7.

24. Jones, S.M., et al., PDGF induces an early and a late wave of PI 3-kinase activity, and only the late wave is required for progression through G1. Curr Biol, 1999. 9(10): p. 512-21.

25. Morris, K.J. and A.H. Corbett, The polyadenosine RNA-binding protein ZC3H14 interacts with the THO complex and coordinately regulates the processing of neuronal transcripts. Nucleic Acids Res, 2018. 46(13): p. 6561-6575.

Page 89: Interactome Analysis Identifies Novel Targets of

78

26. Rha, J., et al., The RNA-binding protein, ZC3H14, is required for proper poly(A) tail length control, expression of synaptic proteins, and brain function in mice. Hum Mol Genet, 2017. 26(19): p. 3663-3681.

27. Kelly, S.M., et al., A conserved role for the zinc finger polyadenosine RNA binding protein, ZC3H14, in control of poly(A) tail length. RNA, 2014. 20(5): p. 681-8.

28. Thapar, R. and A.P. Denmon, Signaling pathways that control mRNA turnover. Cell Signal, 2013. 25(8): p. 1699-710.

29. Madrigal, J.L., et al., Astrocyte-derived MCP-1 mediates neuroprotective effects of noradrenaline. J Neurosci, 2009. 29(1): p. 263-7.

30. Sandhu, J.K., et al., Astrocyte-secreted GDNF and glutathione antioxidant system protect neurons against 6OHDA cytotoxicity. Neurobiol Dis, 2009. 33(3): p. 405-14.

31. Hayakawa, K., et al., Transfer of mitochondria from astrocytes to neurons after stroke. Nature, 2016. 535(7613): p. 551-5.

32. Mudhar, H.S., et al., PDGF and its receptors in the developing rodent retina and optic nerve. Development, 1993. 118(2): p. 539-52.

33. Valenzuela, C.F., et al., Platelet-derived growth factor induces a long-term inhibition of N-methyl-D-aspartate receptor function. J Biol Chem, 1996. 271(27): p. 16151-9.

34. Tseng, H.C. and M.A. Dichter, Platelet-derived growth factor-BB pretreatment attenuates excitotoxic death in cultured hippocampal neurons. Neurobiol Dis, 2005. 19(1-2): p. 77-83.

35. Beazely, M.A., et al., Platelet-derived growth factor selectively inhibits NR2B-containing N-methyl-D-aspartate receptors in CA1 hippocampal neurons. J Biol Chem, 2009. 284(12): p. 8054-63.

36. Osborne, A., J. Sanderson, and K.R. Martin, Neuroprotective Effects of Human Mesenchymal Stem Cells and Platelet-Derived Growth Factor on Human Retinal Ganglion Cells. Stem Cells, 2018. 36(1): p. 65-78.

37. Johnson, T.V., et al., Identification of retinal ganglion cell neuroprotection conferred by platelet-derived growth factor through analysis of the mesenchymal stem cell secretome. Brain, 2014. 137(Pt 2): p. 503-19.

38. Numakawa, T., et al., BDNF function and intracellular signaling in neurons. Histology and Histopathology, 2010. 25(2): p. 237-258.

39. Kitagishi, Y., et al., Dietary regulation of PI3K/AKT/GSK-3β pathway in Alzheimer's disease. Alzheimer's research & therapy, 2014. 6(3): p. 35-35.

Page 90: Interactome Analysis Identifies Novel Targets of

79

40. Altomare, D.A. and A.R. Khaled, Homeostasis and the importance for a balance between AKT/mTOR activity and intracellular signaling. Current medicinal chemistry, 2012. 19(22): p. 3748-3762.

41. Hers, I., E.E. Vincent, and J.M. Tavare, Akt signalling in health and disease. Cellular Signalling, 2011. 23(10): p. 1515-1527.

42. Graham, J.R., et al., mRNA degradation plays a significant role in the program of gene expression regulated by phosphatidylinositol 3-kinase signaling. Mol Cell Biol, 2010. 30(22): p. 5295-305.

43. Venigalla, R.K. and M. Turner, RNA-binding proteins as a point of convergence of the PI3K and p38 MAPK pathways. Front Immunol, 2012. 3: p. 398.

44. Hentze, M.W., et al., A brave new world of RNA-binding proteins. Nat Rev Mol Cell Biol, 2018. 19(5): p. 327-341.

45. Dominguez, D., et al., Sequence, Structure, and Context Preferences of Human RNA Binding Proteins. Mol Cell, 2018. 70(5): p. 854-867 e9.

46. Kelly, S.M., et al., Recognition of polyadenosine RNA by the zinc finger domain of nuclear poly(A) RNA-binding protein 2 (Nab2) is required for correct mRNA 3'-end formation. J Biol Chem, 2010. 285(34): p. 26022-32.

47. Kelly, S.M., et al., Recognition of polyadenosine RNA by zinc finger proteins. Proc Natl Acad Sci U S A, 2007. 104(30): p. 12306-11.

48. Marfatia, K.A., et al., Domain analysis of the Saccharomyces cerevisiae heterogeneous nuclear ribonucleoprotein, Nab2p. Dissecting the requirements for Nab2p-facilitated poly(A) RNA export. J Biol Chem, 2003. 278(9): p. 6731-40.

49. Soucek, S., et al., The Evolutionarily-conserved Polyadenosine RNA Binding Protein, Nab2, Cooperates with Splicing Machinery to Regulate the Fate of pre-mRNA. Mol Cell Biol, 2016.

50. Pak, C., et al., Mutation of the conserved polyadenosine RNA binding protein, ZC3H14/dNab2, impairs neural function in Drosophila and humans. Proc Natl Acad Sci U S A, 2011. 108(30): p. 12390-5.

Page 91: Interactome Analysis Identifies Novel Targets of

80

Supplementary Tables

Supplementary Table 3. A list of PI3K antibodies used in immunoprecipitation

screen, along with molecular weight of target peptide, and corresponding IgG

isotype

Ab # Antibody description and catalogue number Size (kD)

IgG isotype

Ab 1 Phospho-PI3 Kinase p85 (Tyr458)/p55 (Tyr199) Antibody 4228

60 and 85

Rabbit IgG

Ab 2 PI3 Kinase p85 (19H8) Rabbit mAb 4257 85 Rabbit IgG

Ab 3 PI3 Kinase p110α (C73F8) Rabbit mAb 4249 110 Rabbit IgG

Ab 4 PI3 Kinase p110β (C33D4) Rabbit mAb 3011 110 Rabbit IgG

Ab 5 PI3 Kinase Class III (D4E2) Rabbit mAb 3358 100 Rabbit IgG

Ab 6 PI3 Kinase p110γ (D55D5) Rabbit mAb 5405 110 Rabbit IgG

Ab 7 PI3 Kinase p85 Antibody #4292 85 Rabbit IgG

Page 92: Interactome Analysis Identifies Novel Targets of

81

81

Supplementary Table 4. Full list of proteins captured in the PI3K interactome ranked by PSM

Accession Description

Sum(Protein

Sequence Coverage)

Sum(#

Proteins)

Sum of

Unique Peptides

Sum of

Peptides

Sum of Peptide-

to-

spectrum matches (# PSMs)

Cf1/ACM3

(113/118)

Cf1/ACM3

Count

ACM1/ACM3

(114/118)

ACM1/ACM3

Count

Cf2/ACM3

(115/118)

Cf2/ACM3

Count

ACM2/ACM3

(116/118)

ACM2/ACM3

Count

Cf3/ACM3

(117/118)

Cf3/ACM3

Count

negative

ctrl/ACM3 (119/118)

negative

ctrl/ACM3 Count

IPI00263878.2 Phosphatidylinositol 3-kinase regulatory subunit alpha 88.54% 9 40 63 2821 0.826 899 0.896 905 0.995 907 0.891 898 0.938 901 0.106 344

IPI00309224.5

Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha isoform 77.62% 8 59 81 2267 0.669 763 0.85 772 0.914 772 0.792 767 0.777 766 0.084 360

IPI00136110.4

Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta isoform 66.64% 3 52 77 2108 0.83 676 0.885 674 1.078 681 0.804 667 0.926 677 0.114 308

IPI00331708.3 Isoform 1 of MKL/myocardin-like protein 1 54.56% 12 29 41 1551 0.9 382 0.858 389 1.074 388 0.906 392 1.016 390 0.069 152

IPI00117352.1 Tubulin beta-5 chain 65.77% 2 4 25 866 0.747 295 0.814 294 0.915 294 0.78 294 0.757 291 0.058 196

IPI00169463.1 Tubulin beta-2C chain 57.08% 3 1 21 790 0.802 246 0.823 245 0.951 246 0.767 245 0.781 242 0.061 170

IPI00323357.3 Heat shock cognate 71 kDa protein 74.30% 8 30 42 735 0.99 265 0.853 264 1.126 271 0.822 265 0.904 264 0.133 173

IPI00109073.5 Tubulin beta-4 chain 45.95% 2 1 16 717 0.803 215 0.821 214 0.958 215 0.771 214 0.79 211 0.061 148

IPI00338039.1 Tubulin beta-2A chain 62.02% 4 1 21 701 0.797 216 0.795 215 0.957 214 0.743 214 0.783 213 0.056 145

IPI00117348.4 Tubulin alpha-1B chain 69.18% 4 1 22 538 0.666 242 0.758 245 0.893 246 0.803 247 0.708 244 0.065 150

IPI00110753.1 Tubulin alpha-1A chain 69.18% 3 1 22 528 0.655 241 0.753 244 0.878 245 0.791 245 0.688 242 0.064 154

IPI00403810.2 Tubulin alpha-1C chain 63.70% 2 1 20 517 0.655 234 0.759 237 0.89 238 0.796 238 0.683 235 0.062 151

IPI00119627.1 Insulin receptor substrate 1 51.42% 2 24 51 488 0.69 155 0.866 157 0.846 156 0.861 157 0.734 156 0.124 95

IPI00885697.2

phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit delta isoform isoform c 46.46% 8 25 40 467 0.824 143 0.898 143 1.037 146 0.783 137 0.875 141 0.126 74

IPI00112251.1 Tubulin beta-3 chain 44.44% 4 1 18 458 0.817 152 0.814 150 0.955 152 0.768 152 0.79 151 0.054 115

IPI00379844.5 Insulin receptor substrate 2 37.55% 1 19 36 359 0.753 94 0.885 96 0.934 99 0.851 97 0.744 95 0.278 54

IPI00130391.1 protease, serine, 1 19.92% 4 2 5 278 0.746 102 0.741 108 1.155 109 0.739 107 1.122 109 1.433 111

IPI00122928.1 Tubulin beta-6 chain 46.09% 2 1 17 235 0.878 74 0.841 73 0.962 75 0.745 74 0.785 74 0.062 57

IPI00331628.5 Peroxisomal multifunctional enzyme type 2 43.95% 1 15 28 182 1.121 58 0.594 49 0.875 58 0.743 55 0.676 55 0.659 57

IPI00117159.2 Phosphatidylinositol 3-kinase regulatory subunit beta 35.32% 1 2 22 171 0.681 60 0.84 61 0.925 60 0.98 61 0.926 60 0.059 30

IPI00338309.5 Cardiac Ca2+ release channel 21.40% 2 3 69 161 0.293 5 0.578 4 1.654 6 0.272 3 0.121 4 0.357 3

IPI00755181.1 keratin, type I cytoskeletal 10 37.79% 4 6 19 153 0.373 38 0.206 38 0.43 41 0.305 37 0.519 42 0.209 38

Page 93: Interactome Analysis Identifies Novel Targets of

82

IPI00230632.2

Isoform Cas-A of Breast cancer anti-estrogen resistance protein 1 39.59% 3 10 26 148 1.152 43 0.954 43 1.204 43 0.964 43 1.04 42 0.289 24

IPI00970652.1

protein-L-isoaspartate(D-aspartate) O-methyltransferase 76.84% 5 8 16 143 1.139 47 0.673 45 1.209 45 0.697 45 0.852 46 0.936 47

IPI00850337.2 glyceraldehyde-3-phosphate dehydrogenase-like isoform 1 48.90% 29 6 12 126 0.499 36 0.782 36 0.735 36 0.644 36 0.573 36 0.096 30

IPI00759953.1 trypsin 5 19.92% 7 2 5 123 1.322 13 0.747 13 1.308 12 0.807 12 1.335 12 1.462 13

IPI00139795.2 60S acidic ribosomal protein P2 78.26% 2 5 7 118 0.85 56 0.579 56 0.648 56 0.719 56 0.417 54 0.287 53

IPI00649335.3 Uncharacterized protein 32.56% 2 1 8 116 0.07 11 0.104 12 2.667 12 0.052 8 0.064 11 0.052 5

IPI00988790.1 60S ribosomal protein L13-like 49.29% 7 5 12 116 1.072 39 0.87 37 1.42 40 0.788 37 1.425 37 0.334 36

IPI00553798.2 AHNAK nucleoprotein isoform 1 18.58% 2 2 39 111 0.391 14 0.51 14 0.608 14 0.545 14 0.3 14 0.049 12

IPI00406794.2 GRB2-associated-binding protein 1 36.69% 1 9 17 106 0.718 37 0.847 36 0.785 39 0.892 37 0.603 35 0.097 28

IPI00348328.4 keratin Kb40 39.33% 3 2 25 105 0.477 34 0.192 34 0.521 34 0.315 34 0.808 34 0.229 33

IPI00319992.1 78 kDa glucose-regulated protein 36.18% 1 3 20 102 0.858 30 0.804 30 1.064 30 0.806 30 0.788 30 0.075 19

IPI00461416.4 Isoform 1 of Zinc finger CCCH domain-containing protein 14 46.12% 4 11 29 99 0.48 21 0.88 20 0.559 22 0.707 20 0.536 21 0.186 12

IPI00221797.1 Keratin, type II cytoskeletal 75 31.40% 8 2 14 98 0.417 35 0.174 32 0.558 35 0.396 35 1.006 35 0.231 29

IPI00222228.5 hypothetical protein LOC239673 25.84% 5 1 13 93 0.596 12 0.173 12 0.76 12 0.288 12 0.657 12 0.192 10

IPI00554929.3 Heat shock protein HSP 90-beta 37.43% 4 4 19 91 0.865 24 0.843 24 0.942 24 0.868 24 0.733 24 0.18 16

IPI00889253.1

Isoform 4 of GTPase-activating protein and VPS9 domain-containing protein 1 37.43% 6 1 31 90 0.051 3 0.188 3 2.722 3 0.024 3 0.168 3 0.038 2

IPI00622240.4 Keratin, type II cytoskeletal 2 epidermal 33.66% 2 3 17 87 0.84 16 0.307 13 0.929 17 0.349 15 1.343 16 0.353 16

IPI00230365.5 Keratin, type I cytoskeletal 17 42.26% 4 2 18 86 0.419 21 0.178 19 0.475 22 0.337 19 0.7 21 0.243 20

IPI00307837.6 Elongation factor 1-alpha 1 47.40% 4 10 15 85 0.623 34 0.797 34 1.082 34 0.749 34 0.694 34 0.137 27

IPI00354151.3 RIMS-binding protein 3 11.52% 2 1 16 82 0.117 5 0.252 7 3.035 7 0.043 3 0.237 6 0.093 3

IPI01027267.1 disco-interacting protein 2 homolog C 17.03% 5 1 17 80 0.258 7 0.856 8 1.187 8 0.3 6 0.39 7 0.772 8

IPI00625729.2 Keratin, type II cytoskeletal 1 37.52% 3 2 22 73 0.612 25 0.294 22 0.693 25 0.405 25 0.908 25 0.431 22

IPI00473320.2 Putative uncharacterized protein 42.25% 27 5 15 72 0.415 25 0.574 26 0.65 26 0.569 26 0.385 26 0.074 21

IPI00346834.1 Keratin, type II cytoskeletal 2 oral 30.64% 2 1 16 71 0.861 17 0.308 16 1.458 18 0.597 16 1.293 18 0.243 12

IPI00124287.2 Polyadenylate-binding protein 1 46.23% 10 5 25 71 0.75 16 0.81 16 0.963 17 0.782 16 0.657 17 0.326 13

IPI00844650.1

Isoform 1 of Alpha-type platelet-derived growth factor receptor 28.47% 3 9 24 70 0.616 15 0.676 16 0.524 15 0.823 18 0.455 16 0.392 6

IPI00131138.11 Isoform 1 of Filamin-A 24.86% 8 7 42 68 0.76 9 0.814 10 0.791 10 0.654 10 0.509 9 0.477 9

IPI00223902.2 Isoform 2 of Uncharacterized protein KIAA1310 49.83% 3 3 25 66 0.84 3 1.052 3 0.965 3 1.139 3 0.874 3 0.013 1

IPI00114407.2 Isoform 1 of THO complex subunit 4 58.82% 5 1 15 60 0.308 11 0.661 12 0.442 12 0.855 12 0.384 11 0.03 6

IPI00987441.1 Uncharacterized protein (Fragment) 24.68% 2 1 15 60 1.497 4 1.028 4 1.233 4 1.085 4 1.724 4 0.292 4

Page 94: Interactome Analysis Identifies Novel Targets of

83

IPI00313222.5 60S ribosomal protein L6 37.84% 5 3 13 58 0.756 12 0.684 12 1.28 13 0.646 13 0.805 13 0.302 13

IPI00123181.4 Myosin-9 21.22% 4 1 32 56 0.699 4 0.724 4 0.645 4 0.71 4 0.356 3 0.079 3

IPI00111412.3 60S ribosomal protein L4 42.96% 1 5 20 56 0.609 12 0.737 13 1.212 13 0.693 13 0.597 11 0.074 8

IPI00122577.8 Isoform 2 of Cytospin-B 22.28% 5 1 15 56 0.121 8 0.24 8 2.844 9 0.187 6 0.211 7 0.157 6

IPI00468696.3 Keratin, type I cytoskeletal 42 46.68% 2 2 16 55 0.357 12 0.138 9 0.731 13 0.178 10 0.42 12 0.132 11

IPI00988271.1 Uncharacterized protein 50.86% 16 2 6 54 0.486 16 0.573 16 0.622 17 0.717 16 0.489 17 0.176 16

IPI00462140.1 Keratin, type II cytoskeletal 1b 32.87% 3 1 16 53 0.772 17 0.193 15 0.704 17 0.365 17 1.206 17 0.302 15

IPI00116498.1 14-3-3 protein zeta/delta 35.51% 14 4 7 52 0.42 21 0.704 22 0.509 22 0.741 19 0.423 18 0.129 12

IPI00756198.2 Kinetochore-associated protein 1 26.64% 1 4 33 52 1.239 6 0.855 7 1.058 6 0.973 7 0.819 8 1.676 3

IPI00331092.7 40S ribosomal protein S4, X isoform 38.78% 3 5 10 51 0.667 14 0.813 14 0.983 15 0.758 15 0.766 15 0.208 14

IPI00458765.2

KH domain-containing, RNA-binding, signal transduction-associated protein 1 39.05% 1 3 20 48 0.841 7 0.642 7 1.182 7 0.724 7 1.043 7 0.198 2

IPI00604967.4 Uncharacterized protein 63.35% 18 6 13 48 0.561 17 0.706 16 0.923 17 0.775 17 0.68 17 0.081 15

IPI00124499.2 Keratin, type II cytoskeletal 79 30.13% 1 1 11 44 0.76 23 0.393 22 0.855 23 0.932 23 1.789 23 0.344 21

IPI00113377.1 60S acidic ribosomal protein P1 57.02% 1 2 3 44 0.738 29 0.546 29 0.637 29 0.609 29 0.479 29 0.169 29

IPI00775915.1 23 kDa protein 33.83% 6 2 8 44 0.796 19 0.891 19 1.01 19 0.712 19 0.614 19 0.068 9

IPI00139780.1 60S ribosomal protein L23 71.43% 3 5 8 42 0.696 20 0.832 20 0.883 20 0.866 20 0.788 20 0.072 20

IPI00118384.1 14-3-3 protein epsilon 21.57% 13 2 6 41 0.418 16 0.698 17 0.509 16 0.76 14 0.429 13 0.143 10

IPI00320267.1 Microspherule protein 1 41.99% 3 3 15 41 0.368 4 0.518 4 0.897 4 0.499 4 0.837 3

IPI00227522.2 Uncharacterized protein 10.19% 2 1 17 41 0.841 3 0.229 3 0.777 3 0.807 3 1.542 3 0.363 3

IPI00461946.2 Isoform 1 of Probable histone acetyltransferase MYST1 41.92% 1 1 13 40 1.78 8 1.303 9 2.079 8 0.975 6 1.757 6 1.488 4

IPI00111957.3 Histone H2B type 1-A 51.18% 15 1 8 36 0.529 4 0.438 4 0.626 4 0.649 4 0.481 4 0.529 4

IPI00131674.3 trypsinogen 7 23.08% 1 1 5 36 0.743 14 0.628 14 1.416 14 0.814 14 1.173 14 1.801 14

IPI00895079.1 Uncharacterized protein 16.17% 3 1 11 36 0.114 3 0.199 3 2.797 3 0.09 3 0.168 3 0.051 3

IPI00330363.8 60S ribosomal protein L7a 39.85% 14 4 9 34 0.749 8 0.823 8 1.3 8 0.729 8 0.655 8 0.117 8

IPI00457585.1 Isoform 2 of Probable histone acetyltransferase MYST1 65.46% 1 1 9 34 1.77 8 1.313 8 2.079 8 0.986 5 1.397 6 1.667 3

IPI00971244.1 40S ribosomal protein S8-like 26.09% 9 2 6 34 0.599 15 0.771 15 1.134 15 0.591 15 0.601 15 0.138 13

IPI00323806.4 Putative uncharacterized protein 44.38% 4 3 11 33 0.935 12 0.937 12 1.156 12 0.865 12 0.94 11 0.23 11

IPI00137787.3 60S ribosomal protein L8 29.96% 1 2 10 32 1.063 4 1.823 4 1.33 4 0.685 4 0.652 4 0.765 4

IPI00410858.2 Isoform 2 of Polyadenylate-binding protein 2 33.90% 6 1 8 32 0.82 6 0.746 6 0.933 6 0.703 6 0.544 6 0.174 2

IPI00775948.1 Uncharacterized protein 28.32% 4 3 8 31 0.705 8 0.892 8 1.737 9 0.525 9 0.685 8 0.179 7

IPI00307907.1 HAUS augmin-like complex subunit 4 15.70% 1 1 7 30 0.106 7 0.202 8 2.687 8 0.043 5 0.168 8 0.039 6

Page 95: Interactome Analysis Identifies Novel Targets of

84

IPI00653962.1 Uncharacterized protein 30.65% 3 1 4 30 1.246 8 0.906 6 1.809 7 0.816 6 1.94 8 0.089 1

IPI00762542.2 40S ribosomal protein S11 37.97% 3 4 6 29 0.897 11 0.917 11 0.881 11 0.842 11 0.753 11 0.108 11

IPI00222547.6 60S ribosomal protein L28 39.42% 2 1 6 28 0.804 3 0.86 3 1.231 3 0.678 3 1.202 3 0.207 3

IPI00465880.4 40S ribosomal protein S17 54.81% 5 3 5 27 0.706 14 0.922 15 0.963 14 0.949 15 0.558 14 0.164 14

IPI00229542.1 Histone H2A 51.94% 15 2 8 27 0.581 6 0.674 6 0.737 6 0.819 6 0.577 6 0.67 6

IPI00955744.1 ribonuclease inhibitor isoform b 23.58% 2 2 9 27 0.732 4 0.808 4 1.14 4 0.663 4 0.668 4 0.524 1

IPI00555113.2 60S ribosomal protein L18 41.49% 2 1 11 25 0.409 7 0.564 6 1.692 7 0.487 7 0.554 7 0.118 7

IPI00322562.5 40S ribosomal protein S14 46.36% 3 3 7 25 0.869 6 0.73 6 1.12 6 0.702 6 0.498 6 0.116 6

IPI00466258.2

Isoform 1 of SH3 domain-containing kinase-binding protein 1 30.04% 9 2 13 25 0.786 3 0.989 3 1.009 3 0.934 3 0.556 3 0.126 2

IPI00626366.3 Uncharacterized protein 62.78% 3 4 11 25 0.517 7 0.596 7 1.257 7 0.597 7 0.646 6 0.403 4

IPI00115992.1 Uncharacterized protein 42.40% 3 3 7 24 1.7 5 0.818 5 1.265 5 0.783 5 0.805 5 0.196 4

IPI00136251.1 DnaJ homolog subfamily A member 2 31.80% 1 1 6 23 0.883 7 0.738 8 0.836 8 0.939 8 0.786 8 0.438 4

IPI00128904.1 Poly(rC)-binding protein 1 51.69% 1 2 9 23 0.714 3 0.756 3 0.851 3 0.666 3 0.574 3 0.117 3

IPI00331597.6 Histone H1.3 27.60% 4 1 8 22 0.796 8 0.621 8 1.033 8 0.834 8 0.72 8 0.237 8

IPI00122421.5 60S ribosomal protein L27 22.06% 4 2 4 21 1.455 7 0.664 6 1.254 7 0.969 6 0.77 6 0.262 7

IPI00990114.1 Uncharacterized protein 25.99% 2 2 6 21 0.594 11 0.681 11 1.231 11 0.589 10 0.546 11 0.066 5

IPI00986371.1 60S ribosomal protein L27a-like 30.67% 8 2 4 20 1.679 11 0.8 11 1.379 11 0.83 10 0.846 11 0.258 6

IPI00116281.3 T-complex protein 1 subunit zeta 28.06% 1 3 10 19 0.599 8 0.794 8 0.775 8 0.848 8 0.609 8 0.107 7

IPI00116279.3 T-complex protein 1 subunit epsilon 27.36% 2 2 11 18 0.696 3 0.618 3 0.876 3 0.659 3 1.028 3 0.348 1

IPI00469327.5 hypothetical protein LOC277089 30.36% 2 1 6 18 0.897 7 0.775 6 0.976 7 0.338 6 0.342 6 0.25 2

IPI00605025.2 Uncharacterized protein 30.77% 4 1 5 17 0.55 5 0.734 5 1.153 5 0.731 5 0.758 5 0.106 5

IPI00989404.1 60S ribosomal protein L12-like 60.61% 7 3 8 17 0.749 4 0.64 4 1.212 4 0.537 4 0.633 4 0.177 4

IPI00121311.1 DbpA murine homologue 29.22% 9 1 7 16 0.511 4 0.631 4 0.767 5 0.784 5 0.623 4 0.084 3

IPI00621625.1 Uncharacterized protein 41.03% 5 3 6 15 1.516 8 0.813 8 1.237 8 0.653 8 0.875 8 0.178 7

IPI00676904.3 19 kDa protein 28.40% 1 1 3 15 1.021 3 0.953 3 1.633 3 1.06 3 0.35 3 1.523 3

IPI01007744.1 Uncharacterized protein (Fragment) 34.20% 13 1 7 15 1.73 5 0.626 5 1.27 5 0.618 5 0.829 5 0.167 5

IPI00606550.3 Isoform 1 of Ig gamma-2B chain C region 20.05% 5 1 6 14 1.028 6 3.592 6 5.078 6 0.552 5 0.431 6 0.961 4

IPI00226993.5 Thioredoxin 47.62% 1 1 4 14 0.627 11 0.623 11 2.132 11 0.504 11 0.531 11 0.411 10

IPI00228616.5 Histone H1.1 27.70% 1 1 6 12 0.537 3 0.489 3 0.934 3 0.62 3 0.613 3 0.205 2

IPI00118775.2 Uncharacterized protein 31.30% 6 2 3 12 0.482 7 0.493 7 1.183 7 0.51 7 0.635 7 0.129 6

IPI00377441.3 40S ribosomal protein S26 29.57% 2 2 3 11 0.777 3 0.842 3 1.094 3 0.703 3 0.746 3 0.078 3

Page 96: Interactome Analysis Identifies Novel Targets of

85

IPI00648105.1 Uncharacterized protein 16.47% 6 3 3 11 0.656 9 0.604 9 0.875 9 0.586 9 0.427 9 0.206 9

IPI00137736.1 40S ribosomal protein S28 43.48% 2 1 4 10 0.992 4 0.894 3 1.919 5 1.026 3 0.978 3 0.868 3

IPI00849847.1 60S ribosomal protein L23a-like 17.31% 1 1 3 9 0.587 4 0.495 4 1.524 4 0.737 4 0.842 4 0.146 4

IPI00473170.1 Proliferating cell nuclear antigen 31.68% 1 1 5 8 0.432 3 0.886 3 0.769 3 0.845 3 0.379 3 0.039 2

IPI00626239.4 Uncharacterized protein 21.51% 3 1 3 8 1.102 4 0.714 4 0.939 4 0.758 4 0.667 4 0.086 4

IPI00111959.2 CTP synthase 1 10.32% 1 1 5 7 1.04 3 0.547 3 0.703 3 0.579 3 0.595 3 0.277 3

IPI00117288.3 Heterogeneous nuclear ribonucleoprotein A/B 20.00% 3 2 5 7 0.239 3 0.652 3 0.707 3 0.595 3 0.709 3 0.17 1

IPI00798511.1 Isoform 2 of Heterogeneous nuclear ribonucleoprotein F 15.19% 4 2 4 6 0.502 3 0.649 3 0.589 3 0.579 3 0.448 3 0.165 3

Page 97: Interactome Analysis Identifies Novel Targets of

86

86

Supplementary Figures

Supplementary Figure 1. PI3K Immunoprecipitation antibody screen. A seven-monoclonal PI3K

antibody panel was screened to identify an optimal reagent antibody for affinity capture. (A) Ab2 depleted

most of the target protein as shown by the minimal reduced PI3K band in the unbound fraction. (B) Four

antibodies captured PI3K effectively (A2, Ab3, Ab4, and Ab7) with Ab2 demonstrating mediating the

strongest best elution capture band.

Page 98: Interactome Analysis Identifies Novel Targets of

87

Chapter 4: Conclusions and future directions

Page 99: Interactome Analysis Identifies Novel Targets of

88

Conclusions and future directions

The current project identified the PI3K pathway as a key player for ACM mediated

neuroprotection in a metabolic stress model. PI3K directs a range of key cellular

processes relating to survival, growth, and oxidative stress modulation [1-3]. Aside from

phosphorylating pro-survival kinases and suppressing apoptotic signals, PI3K and AKT

also interact with a range of RBPs, through the adapter protein 14-3-3, which serves as

a mediator [4]. PI3K’s interaction with RBPs ultimately promotes the stability of a range

of target mRNAs such as IL-3, myogenin, and 20 other identified transcripts[5-7] In the

current study, a highly specific interactome identified novel PI3K binders , including

ZC3H14 and THOC1, through 14-3-3 adaptors. This is the first study to identify these

PI3K binders, shedding new light on the kinase’s in activating Poly-A binding RBPs in

the context of neuroprotection.

4.1 RBPs in the context of neurodegeneration

Neurodegenerative diseases are increasingly recognized as having an RNA regulatory

dysfunction component, through abnormal RBP activity[8-10]. Mutations in RBPs have

been associated with several neurodegenerative diseases, including Amyotrophic

Lateral Sclerosis (ALS) [11], epilepsy [12], and Parkinson’s disease [13]. In order to

orchestrate the complex neuronal functions, nascent mRNAs must be modified

extensively through dynamic processes carried out by specialized RBPs. A single RBP

is typically involved in the processing of hundreds of different mRNAs. [14] Thus,

mutations in a single RBP can lead to widespread errors in post-transcriptional

modification of target transcripts in neurons. Furthermore, RBPs’ processes are precise

Page 100: Interactome Analysis Identifies Novel Targets of

89

and elaborate, involving multiple steps, and requiring a constant supply of energy in the

energetically taxed neuronal environment [15]. Thus, changes in metabolic rates,

typically seen with aging, leads to a reduction in mature mRNA turnover rates and the

accuracy with which transcripts are processed [16, 17]. Given the involvement of aging

in neurodegenerative diseases, it is reasonable to expect that cumulative mutations in

RBPs, combined with age-related metabolic decline? make RBPs’ functions highly

relevant to neuronal degeneration. Thus, RBPs can be a highly relevant area of focus

when considering novel treatments for neurodegenerative diseases,

4.2 ZC3H14 is essential to the stability of metabolic transcripts

Through binding with mRNA transcripts’ polyA tails, ZC3H14 carries out a range of post-

transcriptional modifications including modifying PolyA tails length [18], nuclear export

[19], and mRNA splicing [20]. Several lines of evidence affirm ZC3H14’s role in

regulating neuronal functional and survival. Firstly, mutation of the ZC3H14 impairs

neural function in Drosophila and has been associated with cognitive deficits in humans

[21]. An increase in bulk RNA poly(A) tail length of target mRNAs is seen in these

models; however, it remains unclear how hyperadenylated mRNAs lead to neuronal

dysfunction. Further work is needed to investigate how hyperadenylation in this context

affects mRNA stability and subsequent expression patterns. Alternatively, one can

explore a possible correlation between hyperadenylation and prevalence in

neurodegenerative disease populations. Secondly, ZC3H14 knockout mouse models

show anatomical changes in brain ventricles as well as working memory deficits [22].

This evidence further emphasizes the role of ZC3H14 in maintaining normal neuronal

functioning, as seen in its impact on anatomy of neuronal tissue, not just their functions.

Page 101: Interactome Analysis Identifies Novel Targets of

90

Thirdly, ZC3H14 has been shown to control mRNA transcripts critical to metabolism,

such as the ATP5g1, which is essential to mitochondrial output [23, 24]. Altered stability

of ATP5g1 may therefore hinder mitochondrial functions, which has negative

implications on neuronal function and survival. Mitochondria are abundantly present in

neuronal tissue; in the eye, mitochondria are particularly dense within unmyelinated

RGC axons in the retina, in order to sustain energetic needs of neurons [25]. To provide

context, up to 90% of the ATP produced by mitochondria is used to maintain action

potentials and neuronal survival [26]. In addition to being the cell’s powerhouse,

mitochodnria also serve as an integration centre for inflammatory and apoptotic signals

in neurons. Thus, one future direction would be to investigate a possible link between

altered ZC3H14 expression and mitochondrial health. An example of this direction

would be to correlate the rate of ZC3H14 mutations with mitochondrial dysfunction in

neurodgenerative diseases, such as glaucoma and Alzheimer’s disease. Another

direction would be to explore the effect of upregulating ZC3H14 expression on neuronal

rescue in the context of metabolic stress.

Another PI3K binder that emerged from the interactome is the RBP THOC1. THOC1

encodes a nuclear matrix protein and is recruited to premature RNA transcripts to

perform various post-transcriptional processing functions [27, 28]. THOC1 also forms a

complex with ZC3H14 coordinately control RNA processing, poly(A) tail length, and

consequently mRNA stability, including ATP5g1 [23]. The scope of the current project

did not include the characterization of THOC1-PI3K interaction; however, a proposed

future direction would be to explore this interaction. Because it is known that THOC1

Page 102: Interactome Analysis Identifies Novel Targets of

91

forms a complex with ZC3H14, it would be insightful to demonstrate whether PI3K

interacts directly with THOC1.

4.3 PDGFr as a key receptor in ACM-mediated neuroprotection

The current study also shows that PI3K-ZC3H14 association was upregulated through

activation of PDGFr with PDGF-BB in Ht22 cells. Exogenous PDGF-BB was also

sufficient to reproduce both ACM-induced AKT phosphorylation as well as ACM-

mediated neuroprotection in Ht22 cells. A number of other studies showed that

astrocyte-mediated neuroprotection relieson growth factors, including vascular

endothelial growth factors (VEGF) and fibroblast growth factor (FGF), and the insulin-

like growth factor-1 (IGF-1) neurons [29-32]. Neurons of the central nervous system

(CNS) and sensory tissues like the retina, rely on NF support from neighboring glia and

target sites to modulate their function from developmental stages through maturity [33,

34]. NFs maintain neuronal survival through inhibition of apoptotic pathways, promoting

anti-oxidant activities, and boosting cellular bioenergetics [35]. Conversely, NF

deprivation induces the intrinsic apoptotic pathway, causing reduced ATP production,

reactive oxygen species generation, cytochrome C release, and subsequent caspase

activation [36]. However, to our knowledge this is the first study describing a

neuroprotective role of astrocyte-induced PDGFr activation. PDGF is a dimer of A- and

B-chains (AA, BB, or AB) which signals through tyrosine kinase receptors (RTKs)

(PDGFRα and PDGFRβ [37, 38].

Page 103: Interactome Analysis Identifies Novel Targets of

92

In a neuronal context PDGF is secreted primarily by macroglia, including astrocytes and

retinal Muller cells, as well as by neurons, mediating short-range paracrine

communication between astrocytes and neurons [39]. It has also been shown to protect

neurons against oxidative stress and NMDA-induced metabolic stress in primary

neurons and retinal ganglion cells [40-44]. In human, PDGF is highly expressed in white

matter in humans following stroke, suggesting that PDGF may be involved in either

regeneration or protection of damaged neurons [45]. Thus, PDGF involvement in

astrocyte-mediated neuroprotection in the current study adds further evidence to the

role PDGF in neuroprotection against metabolic stress. However, there are some

considerations for the use of PDGF as a neuroprotective biologic in aerogeneration.

One of PDGF’s roles is to promote new blood vessel maturation; thus, the impact of

PDGF on retinal vasculature needs to be considered carefully, given the risk of aberrant

vascularization[46] . Furthermore, unlike the homogenous cell line used to asses PDGF

neuroprotective potential, neuronal tissue is highly complex and heterogenous. As a

result, administering PDGF will affect surrounding glia and connective tissue, initiating

signaling cascades in a number of cell populations. The collective outcome of this

signaling wave needs to be addressed carefully in future studies to assess PDGF’s

safety in neuronal tissue.

4.4 Considerations for future direction

Taken together, this study provides a case for the use of interactomes in

exploring signalling pathways involved in biological activities of interest, such as

neuroprotection or regeneration. This approach can be especially powerful if a specific

Page 104: Interactome Analysis Identifies Novel Targets of

93

protein is known to be critical to an activity; in this case, the protein can serve as a bait

with which the rest of the pathway is captured. However, a few points are important to

point out with respect to the interactome data. While the interaction between ZC3H14

and PI3K has been implied using evidence mass spectroscopy and IP studies, it is

unclear whether the interaction occurs directly or as part of a larger complex. PI3K

interacts with a number of targets along the AKT-mTOR pathway (see Chapter 1).

Therefore, it is important to investigate the involvement of other targets downstream of

PI3K in transducing ACM signalling in neurons through ZC3H14 binding. A likely

downstream target is mTOR, which has been shown to be involved in a range of

neuroprotection and regeneration models. Secondly, while using ACM derived from

primary astrocytes, for consistency the PI3K interactome performed in this work was

carried out using the transformed hippocampal cell line Ht22, which is a well-established

model in neurodegenerative research [47-49]. While useful and convenient,

immortalized cell lines are genetically altered, and are prone to cumulative mutations

with passaging, which may introduce functional and phenotypic differences compared to

their primary counterparts. Therefore, one future direction of this project is to confirm the

PI3K-ZC3H14 interaction in vivo using brain or retinal models. We have generated

immunofluorescence images of retina sections showing localization of ZC3H14 in the

GCL layer; we also demonstrate co-localization with PI3K following Kainic acid injury

(Figure 4-1). Further, it is essential to confirm whether PDGF administration increases

the PI3K association with ZC3H14, as observed in Ht22 cells. Similarly, it would be

useful to assess the neuroprotective effect of PDGF administration in ZC3H14 knockout

models under metabolic stress conditions.

Page 105: Interactome Analysis Identifies Novel Targets of

94

Figure 4- 1 Immunostaining of ZC3H14, and AKT in retinal sections treated with

vehicle and Kianic acid (1hr).

In conclusion, neurodegeneration and neuroprotection are extremely complex

processes that involve hundreds of biological factors and signalling pathways in delicate

orchestration. Thus, it is more than likely that ZC3H14’s role in protecting neurons is

complemented with a range of other factors and pathways. Future work can expand on

the current work using proteomic analyses that can identify other binding partners of

ZC3H14, to generate a snapshot of this RBP in its natural complex. Using this

approach, one can then compare global functional patterns of this ZC3H14 and its

partners in the normal and disease settings. This ‘bird eye’ view takes into account the

complexity of neurodegenerative disease, and may ultimately bring us closer to

incorporating ZC3H14 targeting in a neuroprotective therapy.

Page 106: Interactome Analysis Identifies Novel Targets of

95

Chapter 4 references

1. Vanhaesebroeck, B. and M.D. Waterfield, Signaling by distinct classes of phosphoinositide 3-kinases. Exp Cell Res, 1999. 253(1): p. 239-54.

2. Vanhaesebroeck, B., et al., The emerging mechanisms of isoform-specific PI3K signalling. Nat Rev Mol Cell Biol, 2010. 11(5): p. 329-41.

3. Hennessy, B.T., et al., Exploiting the PI3K/AKT pathway for cancer drug discovery. Nat Rev Drug Discov, 2005. 4(12): p. 988-1004.

4. Thapar, R. and A.P. Denmon, Signaling pathways that control mRNA turnover. Cell Signal, 2013. 25(8): p. 1699-710.

5. Ming, X.F., et al., Parallel and independent regulation of interleukin-3 mRNA turnover by phosphatidylinositol 3-kinase and p38 mitogen-activated protein kinase. Mol Cell Biol, 2001. 21(17): p. 5778-89.

6. Benjamin, D., et al., BRF1 protein turnover and mRNA decay activity are regulated by protein kinase B at the same phosphorylation sites. Mol Cell Biol, 2006. 26(24): p. 9497-507.

7. Graham, J.R., et al., mRNA degradation plays a significant role in the program of gene expression regulated by phosphatidylinositol 3-kinase signaling. Mol Cell Biol, 2010. 30(22): p. 5295-305.

8. Nussbacher, J.K., et al., RNA-binding proteins in neurodegeneration: Seq and you shall receive. Trends Neurosci, 2015. 38(4): p. 226-36.

9. Conlon, E.G. and J.L. Manley, RNA-binding proteins in neurodegeneration: mechanisms in aggregate. Genes Dev, 2017. 31(15): p. 1509-1528.

10. Doxakis, E., RNA binding proteins: a common denominator of neuronal function and dysfunction. Neurosci Bull, 2014. 30(4): p. 610-26.

11. Johnson, J.O., et al., Mutations in the Matrin 3 gene cause familial amyotrophic lateral sclerosis. Nat Neurosci, 2014. 17(5): p. 664-666.

12. Wagnon, J.L., et al., CELF4 regulates translation and local abundance of a vast set of mRNAs, including genes associated with regulation of synaptic function. PLoS Genet, 2012. 8(11): p. e1003067.

13. Bryant, C.D. and N. Yazdani, RNA-binding proteins, neural development and the addictions. Genes Brain Behav, 2016. 15(1): p. 169-86.

14. Keene, J.D. and S.A. Tenenbaum, Eukaryotic mRNPs may represent posttranscriptional operons. Mol Cell, 2002. 9(6): p. 1161-7.

Page 107: Interactome Analysis Identifies Novel Targets of

96

15. Herculano-Houzel, S., The remarkable, yet not extraordinary, human brain as a scaled-up primate brain and its associated cost. Proceedings of the National Academy of Sciences, 2012. 109(Supplement 1): p. 10661.

16. Ma, J.F., D.T. Hall, and I.E. Gallouzi, The impact of mRNA turnover and translation on age-related muscle loss. Ageing Res Rev, 2012. 11(4): p. 432-41.

17. Gorospe, M., RNA-binding proteins: post-transcriptional control of aging traits: an introduction to a series of review articles. Ageing Res Rev, 2012. 11(4): p. 421-2.

18. Kelly, S.M., et al., A conserved role for the zinc finger polyadenosine RNA binding protein, ZC3H14, in control of poly(A) tail length. RNA, 2014. 20(5): p. 681-8.

19. Marfatia, K.A., et al., Domain analysis of the Saccharomyces cerevisiae heterogeneous nuclear ribonucleoprotein, Nab2p. Dissecting the requirements for Nab2p-facilitated poly(A) RNA export. J Biol Chem, 2003. 278(9): p. 6731-40.

20. Soucek, S., et al., The Evolutionarily-conserved Polyadenosine RNA Binding Protein, Nab2, Cooperates with Splicing Machinery to Regulate the Fate of pre-mRNA. Mol Cell Biol, 2016.

21. Pak, C., et al., Mutation of the conserved polyadenosine RNA binding protein, ZC3H14/dNab2, impairs neural function in Drosophila and humans. Proc Natl Acad Sci U S A, 2011. 108(30): p. 12390-5.

22. Rha, J., et al., The RNA-binding protein, ZC3H14, is required for proper poly(A) tail length control, expression of synaptic proteins, and brain function in mice. Hum Mol Genet, 2017. 26(19): p. 3663-3681.

23. Morris, K.J. and A.H. Corbett, The polyadenosine RNA-binding protein ZC3H14 interacts with the THO complex and coordinately regulates the processing of neuronal transcripts. Nucleic Acids Res, 2018. 46(13): p. 6561-6575.

24. Bonora, M., et al., Role of the c subunit of the FO ATP synthase in mitochondrial permeability transition. Cell Cycle, 2013. 12(4): p. 674-83.

25. Bristow, E.A., et al., The distribution of mitochondrial activity in relation to optic nerve structure. Arch Ophthalmol, 2002. 120(6): p. 791-6.

26. Osborne, N.N., et al., Glaucoma: Focus on mitochondria in relation to pathogenesis and neuroprotection. European Journal of Pharmacology, 2016. 787: p. 127-133.

27. Luna, R., A.G. Rondón, and A. Aguilera, New clues to understand the role of THO and other functionally related factors in mRNP biogenesis. Biochim Biophys Acta, 2012. 1819(6): p. 514-20.

Page 108: Interactome Analysis Identifies Novel Targets of

97

28. Rehwinkel, J., et al., Genome-wide analysis of mRNAs regulated by the THO complex in Drosophila melanogaster. Nat Struct Mol Biol, 2004. 11(6): p. 558-66.

29. Arai, K. and E.H. Lo, Astrocytes protect oligodendrocyte precursor cells via MEK/ERK and PI3K/Akt signaling. J Neurosci Res, 2010. 88(4): p. 758-63.

30. Sun, Y.Y., et al., Cell type-specific dependency on the PI3K/Akt signaling pathway for the endogenous Epo and VEGF induction by baicalein in neurons versus astrocytes. PLoS One, 2013. 8(7): p. e69019.

31. Ito, J., et al., Astrocytes produce and secrete FGF-1, which promotes the production of apoE-HDL in a manner of autocrine action. J Lipid Res, 2005. 46(4): p. 679-86.

32. Pitt, J., et al., Neuroprotective astrocyte-derived insulin/insulin-like growth factor 1 stimulates endocytic processing and extracellular release of neuron-bound Abeta oligomers. Mol Biol Cell, 2017. 28(20): p. 2623-2636.

33. Seki, M., et al., Muller cells as a source of brain-derived neurotrophic factor in the retina: Noradrenaline upregulates brain-derived neurotrophic factor levels in cultured rat Muller cells. Neurochemical Research, 2005. 30(9): p. 1163-1170.

34. Spalding, K.L., R.A. Rush, and A.R. Harvey, Target-derived and locally derived neurotrophins support retinal ganglion cell survival in the neonatal rat retina. Journal of Neurobiology, 2004. 60(3): p. 319-327.

35. Almasieh, M., et al., The molecular basis of retinal ganglion cell death in glaucoma. Progress in Retinal and Eye Research, 2012. 31(2): p. 152-181.

36. Green, D.R., L. Galluzzi, and G. Kroemer, Metabolic control of cell death. Science, 2014. 345(6203): p. 1466-+.

37. Heldin, C.H. and B. Westermark, Platelet-derived growth factor: three isoforms and two receptor types. Trends Genet, 1989. 5(4): p. 108-11.

38. Schlessinger, J., Cell signaling by receptor tyrosine kinases. Cell, 2000. 103(2): p. 211-25.

39. Mudhar, H.S., et al., PDGF and its receptors in the developing rodent retina and optic nerve. Development, 1993. 118(2): p. 539-52.

40. Valenzuela, C.F., et al., Platelet-derived growth factor induces a long-term inhibition of N-methyl-D-aspartate receptor function. J Biol Chem, 1996. 271(27): p. 16151-9.

41. Tseng, H.C. and M.A. Dichter, Platelet-derived growth factor-BB pretreatment attenuates excitotoxic death in cultured hippocampal neurons. Neurobiol Dis, 2005. 19(1-2): p. 77-83.

Page 109: Interactome Analysis Identifies Novel Targets of

98

42. Beazely, M.A., et al., Platelet-derived growth factor selectively inhibits NR2B-containing N-methyl-D-aspartate receptors in CA1 hippocampal neurons. J Biol Chem, 2009. 284(12): p. 8054-63.

43. Osborne, A., J. Sanderson, and K.R. Martin, Neuroprotective Effects of Human Mesenchymal Stem Cells and Platelet-Derived Growth Factor on Human Retinal Ganglion Cells. Stem Cells, 2018. 36(1): p. 65-78.

44. Johnson, T.V., et al., Identification of retinal ganglion cell neuroprotection conferred by platelet-derived growth factor through analysis of the mesenchymal stem cell secretome. Brain, 2014. 137(Pt 2): p. 503-19.

45. Krupinski, J., et al., A putative role for platelet-derived growth factor in angiogenesis and neuroprotection after ischemic stroke in humans. Stroke, 1997. 28(3): p. 564-73.

46. Dong, A., et al., Antagonism of PDGF-BB suppresses subretinal neovascularization and enhances the effects of blocking VEGF-A. Angiogenesis, 2014. 17(3): p. 553-62.

47. Jacewicz, D., et al., Method for detection of hydrogen peroxide in HT22 cells. Sci Rep, 2017. 7: p. 45673.

48. Choi, J.H., et al., Baicalein protects HT22 murine hippocampal neuronal cells against endoplasmic reticulum stress-induced apoptosis through inhibition of reactive oxygen species production and CHOP induction. Experimental &Amp; Molecular Medicine, 2010. 42: p. 811.

49. Rao, W., et al., Homer1a attenuates glutamate-induced oxidative injury in HT-22 cells through regulation of store-operated calcium entry. Scientific Reports, 2016. 6: p. 33975.