effects of trpm2 inhibition in neuroprotection following

89
Effects of TRPM2 Inhibition in Neuroprotection following Neonatal Hypoxic-Ischemic Brain Injury by Feiya Li A thesis submitted in conformity with the requirements for the degree of Master of Science Department of Physiology University of Toronto © Copyright by Feiya Li 2017

Upload: others

Post on 19-Nov-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Effects of TRPM2 Inhibition in Neuroprotection following

Effects of TRPM2 Inhibition in Neuroprotection following Neonatal Hypoxic-Ischemic Brain Injury

by

Feiya Li

A thesis submitted in conformity with the requirements for the degree of Master of Science

Department of Physiology University of Toronto

© Copyright by Feiya Li 2017

Page 2: Effects of TRPM2 Inhibition in Neuroprotection following

ii

Effects of TRPM2 Inhibition in Neuroprotection following

Neonatal Hypoxic-Ischemic Brain Injury

Feiya Li

Master of Science

Department of Physiology

University of Toronto

2017

Abstract

Neonatal hypoxic-ischemic (HI) brain injury is a major cause of acute mortality and

chronic neurological morbidity in infants and children. Studies indicate that transient

receptor potential melastatin 2 or TRPM2 (a non-selective cation channel with high

permeability to calcium that can be activated by intracellular adenosine diphosphoribose

[ADPR] and H2O2) can mediate neuronal death following acute ischemic insults in adult

mice as well as HI brain injury in neonatal mice. My study tested the effect of a newly

described TRPM2 channel inhibitor AG490 by using a H2O2-induced neuronal cell death

in vitro model and mouse HI brain injury in vivo model. I found that the inhibition of

TRPM2 channels by AG490 demonstrates a neuroprotective effect both in vitro and in

vivo. The neuroprotective effect of AG490 following post-injury treatment suggests the

potential clinical implications of this drug, including the possible prevention of HI related

neurological complications such as hypoxic-ischemic encephalopathy.

Page 3: Effects of TRPM2 Inhibition in Neuroprotection following

iii

Acknowledgements

I would first like to express my profound gratitude to my supervisor, Dr. Hong-Shuo Sun,

for taking me as a student in September 2015 and giving me, an international student, this

opportunity to work in such a wonderful and collaborative lab. Over the past 2 years, he

has given me so much advice and trained me to improve my research capabilities as well

as my language skills. I would also like to express my gratitude to my mentor, Dr. Zhong-

Ping Feng, for her critical advice and guidance throughout the program. Dr. Feng knows

students well and reads my minds, and her strong scientific sense and logical thinking gave

me so much good advice in my project design as well as scientific presentation skills.

During the past 2 years of graduate study, I grew up fast and learnt a lot. When I looked

back to my first draft of proposal, which almost got fully re-written by Dr. Sun, and thought

back to my first presentation practice on the lab meeting, I clearly noticed that how much

time and effort my supervisor Dr. Sun and my mentor Dr. Feng had put on me. I would

like to sincerely express my gratefulness for their patience and supports. I would also like

to thank my supervisory committee members, Dr. Tianru Jin and Dr. Shuzo Sugita, for

their good comments and insightful advice and being supportive throughout my training

process.

I would like to thank all of my lab mates: Vivian Szeto, Ekaterina Turlova, Haitao Wang,

Raymond Wong, Nancy Dong, Sammen Huang, Ahmed Abussaud, Ji-Sun Kim, Shuzhen

Zhu, Meihua Bao, Qing Li, Joseph Leung, for their help whenever I needed during my

graduate study process. Especially thank to Haitao, who was the first one I met in lab and

taught me everything including techniques and background knowledges and gave

Page 4: Effects of TRPM2 Inhibition in Neuroprotection following

iv

suggestions in my project design. Also, specially thank to Vivian, who was always patient

and gave me courage whenever I was feeling either a bit lost because of the culture

differences, or struggling with my projects.

I also want to thank all of my close friends I met in Knox College (Fan Xia, Linyang Yu,

Dongzi Li, Siqi Liu, Qing Yuan…) and everyone in FIP committee (Kiru, Ankur,

Hanna…), for making my life in University of Toronto more colorful and full of laughter.

Special thank to Dongzi Li and Qing Yuan, for their understanding, accompany and

encouragements throughout my preparation of this thesis and the thesis defense.

Finally, I would like to express my gratitude to my family: my parents, grandparents, my

uncle and aunt, for providing me with unfailing supports and continuous encouragements

throughout my years of study. This accomplishment would not have been possible without

them.

Page 5: Effects of TRPM2 Inhibition in Neuroprotection following

v

Table of Contents

Abstract ............................................................................................................................... ii

Acknowledgements ............................................................................................................ iii

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

List of Abbreviations ....................................................................................................... viii

List of Figures ................................................................................................................... xii

Chapter 1 Introduction ....................................................................................................... 1

1 Neonatal Hypoxic-Ischemic Brain Injury .................................................................... 1

1.1 Incidence and global impact ................................................................................. 1

1.2 Standard diagnostic criteria for neonatal HI ......................................................... 2

1.3 Current therapy ..................................................................................................... 3

2 Failure in Targeting Glutamate Receptors as Pharmacological Targets ...................... 4

3 Targeting Non-Glutamate Channels ............................................................................ 7

3.1 Sodium-calcium exchangers ................................................................................. 7

3.2 Hemichannels ........................................................................................................ 8

3.3 Volume-regulated anion channels......................................................................... 9

3.4 Acid-sensing ion channels (ASICs) ...................................................................... 9

3.5 Transient receptor potential melastatin (TRPM) subfamily ............................... 10

4 Transient Receptor Potential Channels (TRPs) ......................................................... 12

5 TRPM2 Channel ........................................................................................................ 12

5.1 TRPM2 protein structure, transmembrane topology and distribution ................ 12

5.2 TRPM2 biophysical properties and gating mechanism ...................................... 16

5.3 The physiological and pathophysiological role of TRPM2 channels ................. 17

6 Pharmacological Interactions ..................................................................................... 20

6.1 Flufenamic acid (FFA) ........................................................................................ 20

6.2 Anti-fungal agents (clotrimazole and econazole) ............................................... 20

6.3 2-APB ................................................................................................................. 21

6.4 Divalent heavy metal cations .............................................................................. 21

6.5 AG490 ................................................................................................................. 21

Chapter 2 Rationale and Hypothesis ................................................................................ 23

Page 6: Effects of TRPM2 Inhibition in Neuroprotection following

vi

Rationale ....................................................................................................................... 23

Hypothesis..................................................................................................................... 23

Chapter 3 Aims and Experimental Design....................................................................... 24

Aims: ............................................................................................................................. 24

Experimental Design Outline:....................................................................................... 24

Chapter 4 Materials and Methods .................................................................................... 25

1 Ethics Approval ......................................................................................................... 25

2 Animals ...................................................................................................................... 25

3 Reagents ..................................................................................................................... 25

4 Cell Culture ............................................................................................................ 25

5 In vitro H2O2-induced Neuronal Cell Death Model ................................................... 26

6 Cell Viability Assay ................................................................................................... 26

7 Electrophysiology (Whole Cell Patch Clamp) ........................................................... 27

8 Drug Administration .................................................................................................. 27

9 In vivo Hypoxic-Ischemic Mouse Model .................................................................. 28

10 Infarct Volume Measurement, Whole Brain Imaging and Histological Assessments

....................................................................................................................................... 29

10.1 TTC staining/Infarct volume measurement ...................................................... 29

10.2 Whole brain imaging/Nissl staining.................................................................. 29

11 Neurobehavioral Assessments ................................................................................. 30

11.1 Geotaxis reflex .................................................................................................. 30

11.2 Cliff avoidance test ........................................................................................... 30

11.3 Grip test ............................................................................................................. 31

11.4 Passive avoidance test ....................................................................................... 31

12 Immunohistochemistry and Confocal Imaging ........................................................ 32

13 Western Blot ............................................................................................................ 32

14 Statistics and Data Analysis ..................................................................................... 33

Chapter 5 Results ............................................................................................................. 34

1. The level of TRPM2 mRNA expression in the hippocampus and cortex increases

consistently with the developmental age ...................................................................... 34

2 AG490 as a pharmacological inhibitor of the TRPM2 channel ................................. 36

3 AG490 protects neurons from H2O2-induced cell injury in vitro .............................. 38

Page 7: Effects of TRPM2 Inhibition in Neuroprotection following

vii

4 The Effect of AG490 Pre-treatment on Hypoxic-Ischemic Brain Injury in vivo. ..... 40

4.1 Pre-treatment with TRPM2 inhibitor AG490 reduced the brain infarct volume

of hypoxic-ischemic brain injury in vivo. ................................................................. 41

4.2 Pre-treatment with TRPM2 inhibitor AG490 reduced brain damage following

hypoxic-ischemic brain injury. ................................................................................. 44

4.3 Pre-treatment with TRPM2 inhibitor AG490 promotes recovery after HI

challenge ................................................................................................................... 46

4.4 Pre-treatment with TRPM2 inhibitor AG490 improves short-term

neurobehavioral performance after HI ...................................................................... 47

4.5 Pre-treatment with TRPM2 inhibitor AG490 also improves long-term

neurobehavioral performance after HI ...................................................................... 49

4.6 Pre-treatment with TRPM2 inhibitor AG490 reduces reactive astrocyte

activation ................................................................................................................... 51

4.7 Pre-treatment with TRPM2 inhibitor AG490 may reduce HI brain damage

through Akt mediated signaling pathways ................................................................ 53

5 The Effect of AG490 Post-treatment on Hypoxic-Ischemic Brain Injury in vivo. .... 54

5.1 AG490 *post-treatment 1 (30 mg/kg, i.p.) reduced brain infarct volume of

hypoxic-ischemic brain injury in vivo. ..................................................................... 54

5.2 AG490 *post-treatment 1 (30 mg/kg, i.p.) reduced brain damage following

hypoxic-ischemic brain injury .................................................................................. 54

5.3 AG490 *post-treatment 1 (30 mg/kg, i.p.) improves neurobehavioral

performance and general recovery after HI .............................................................. 57

6 AG490 *post-treatment 2 (30mg/kg, i.p., immediately after HI induction)

demonstrate a trend towards neuroprotection following HI brain injury ..................... 59

Discussion ......................................................................................................................... 60

1. Connection between clinics and the current study .................................................... 60

2. Summary of major findings ...................................................................................... 61

3. Significance of the current study .............................................................................. 62

4. Differences between neonatal HI brain injury and adult stroke ............................... 62

5. Proposed mechanism of neonatal HI brain injury..................................................... 63

6. Pitfalls in the current study and proposed future directions ..................................... 65

REFERENCES ................................................................................................................. 68

Page 8: Effects of TRPM2 Inhibition in Neuroprotection following

viii

List of Abbreviations

ADPR Adenosine disphosphate ribose

Akt Protein kinase B

AMP Adenosine monophosphate

AMPA α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid

receptor

ASICs Acid-sensing ion channels

ATP Adenosine triphosphate

BDNF Brain-derived neurotrophic factor

cAMP Cyclic adenosine monophosphate

cADPR Cyclic adenosine diphosphate ribose

Ca2+ Calcium ion

CaM Calmodulin

Cl- Chloride ion

CNS Central nervous system

CTL Control

Cx43 Connexin 43

DMSO Dimethyl sulfoxide

EEG Electroencephalography

E16 Embryonic day 16

FFA Flufenamic acid

GAPDH Glyceraldehyde 3-phosphata

GFAP Glial fibrillary acidic protein

Page 9: Effects of TRPM2 Inhibition in Neuroprotection following

ix

GPCRs G-protein-coupled receptors

GSK-3α Glycogen synthase kinase 3 alpha

GSK-3β Glycogen synthase kinase 3 beta

HEK293 Human embryonic kidney 293

HI Hypoxic-Ischemic

HIE Hypoxic-Ischemic Encephalopathy

H2O2 Hydrogen Peroxide

ICC Immunocytochemistry

IHC Immunohistochemistry

i.p. Intraperitoneal

IP3 Inositol 1,4,5-trisphosphate

i.v. Intravenous

IV Current-voltage

JAK2 Janus kinase 2

KATP ATP-sensitive potassium channel

K+ Potassium ion

KO Knockout

MAPK Mitogen-activated protein kinases

MCAO Middle cerebral artery occlusion

MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium

bromide)

MRI Magnetic resonance imaging

Na+ Sodium ion

NAD Nicotinamide adenine dinucleotide

Page 10: Effects of TRPM2 Inhibition in Neuroprotection following

x

NCX Na(+)/Ca(2+) exchanger/Sodium-calcium exchanger

NMDA N-methyl-D-aspartate receptor

NSAIDs Non-steroidal anti-inflammatory drugs

NUDT9-H Nudix (Nucleoside Diphosphate Linked Moiety X)-Type

Motif 9

OGD Oxygen glucose deprivation

PARG Poly (ADP-ribose) glycohydrolase

PARP Poly (ADP-ribose) polymerase

PCR Polymerase chain reaction

pH Potential of Hydrogen

PI3K Phosphoinositide 3-kinase

PIP2 Phosphatidylinositol 4,5-bisphosphate

P7/P14 Postnatal day 7/14

RCT Randomized controlled trials

ROS Reactive oxygen species

siRNA Small interfering RNA

S5/S6 Segment 5/ Segment 6

TRPs Transient receptor potential ion channel superfamily

TRPA Transient receptor potential ankyrin

TRPC Transient receptor potential canonical

TRPM Transient receptor potential melastatin

TRPV Transient receptor potential vanilloid

TRPP Transient receptor potential polycystin

TRPML Transient receptor potential mucolipin

Page 11: Effects of TRPM2 Inhibition in Neuroprotection following

xi

TRPM2 Transient receptor potential melastatin 2

TTC Tetrazolium chloride

VRACs Volume-regulated anion channels

WT Wildtype

Page 12: Effects of TRPM2 Inhibition in Neuroprotection following

xii

List of Figures

Figure 1. Classical glutamate receptors (NMDA and AMPA receptor) model of neuronal

cell death …………………………………………………………………………….........5

Figure 2. Potential mechanisms involved in excitotoxicity following ischemic stress.......9

Figure 3. Family tree of TRP channel superfamily………………………………………11

Figure 4. TRPM2 protein structure and variants…………………………………….......13

Figure 5. Proposed mechanisms of TRPM2 channel activation by H2O2 and involvement

TRPM2 channel activity in physiological and pathophysiological processes...................16

Figure 6. TRPM2 channel inhibitors.................................................................................19

Figure 7. An outline of the project experimental design...................................................21

Figure 8. TRPM2 expression in the cortex and hippocampus increases with

development.......................................................................................................................31

Figure 9. AG490 efficiently inhibited H2O2-induced TRPM2 current on TRPM2

overexpression HEK293 cells............................................................................................33

Figure 10. TRPM2 inhibitor AG490 pre-treatment reduced neuronal cell death following

H2O2-induced cell injury....................................................................................................35

Figure 11. Timeline of neonatal hypoxic-ischemic injury and experimental

procedures..........................................................................................................................36

Figure 12. Lower dose of AG490 (15 mg/kg) did not show effect on

neuroprotection..................................................................................................................38

Figure 13. Pretreatment of TRPM2 currents inhibitor AG490 (30 mg/kg) reduced brain

infarct volume of hypoxic-ischemic brain injury in vivo...................................................39

Figure 14. TRPM2 inhibitor AG490 reduced brain damage following hypoxic-ischemic

brain injury.........................................................................................................................41

Page 13: Effects of TRPM2 Inhibition in Neuroprotection following

xiii

Figure 15. AG490 pre-treatment (20 min before HI injury) improves general health

recovery after HI challenge...............................................................................................42

Figure 16. AG490 pre-treatment (20 min before HI injury) improves neurobehavioral

performance after HI challenge.........................................................................................44

Figure 17. Long – term behavioral assessment of functional recovery in the pre-treatment

paradigm following hypoxic-ischemic injury....................................................................46

Figure 18. AG490 pre-treatment restores neuronal cell numbers and reduces reactive

astrocyte activation............................................................................................................53

Figure 19. Biochemical assessment of signaling pathways affected by hypoxic-ischemic

insult on neonatal brain in a pre-treatment paradigm........................................................54

Figure 20. *Post-treatment 1 (30 mg/kg, i.p.) reduced brain infarct volume of hypoxic-

ischemic brain injury in vivo..............................................................................................56

Figure 21. TRPM2 inhibitor AG490 reduced brain damage following hypoxic-ischemic

brain injury.........................................................................................................................57

Figure 22. AG490 post-treatment 1 (immediately after ischemic injury) improves general

health and neurobehavioral performance after HI challenge.............................................59

Figure 23. *Post-treatment 2 (30 mg/kg, i.p.) shows a trend of neuroprotective effect

following hypoxic-ischemic brain injury...........................................................................60

Figure 24. Propose mechanism for inhibitory effect of AG490........................................65

Page 14: Effects of TRPM2 Inhibition in Neuroprotection following

1

Chapter 1 Introduction

1 Neonatal Hypoxic-Ischemic Brain Injury

When a term neonate’s (defined as 36 gestational weeks or later1-3) brain does not receive sufficient

oxygen (hypoxia) and blood (ischemia), neonatal hypoxic-ischemic encephalopathy may result (or

HIE for short4). Neonatal encephalopathy (HIE) can result from diverse conditions, with hypoxic-

ischemic brain injury (HI brain injury) being the most common. Therefore, the terms HI brain

injury and HIE are usually used synonymously. HI brain injury is a condition which can cause

significant mortality and long-term morbidity. The injury can be a clinical consequence of

perinatal, birth and/or neonatal asphyxia4.

1.1 Incidence and global impact

Clinically, most HI brain injury cases are due to birth asphyxia which causes 840,000 or 23 % of

all neonatal deaths worldwide5, 6. The severity and length of oxygen and blood deprivation affects

whether HI brain injury occurs and how severe it is. Due to different levels of severity, patients

suffer from symptoms ranging from transient behavioral abnormalities, occasional periods of

apnea and seizure-related symptoms to even death from cardiorespiratory failure5. 50-80 % of the

survivors will suffer from severe developmental or cognitive delays, motor impairments and

learning disabilities5, 7. Symptoms may worsen as the child continues to develop. The lifetime costs

to the health care system have been estimated to be as high as $1.5 million per person in Canada5.

Page 15: Effects of TRPM2 Inhibition in Neuroprotection following

2

1.2 Standard diagnostic criteria for neonatal HI

Based on biomedical markers that correlate to clinical outcomes, there is an established set of

predictors for neonatal HI brain injury1, 7, 8:

i) Apgar Score

At birth, doctors and nurses carefully examine the newborn's condition and give a

number rating from 0 to 10. This number is called an Apgar score. The Apgar rates skin

color, heart rate, muscle tone, reflexes and breathing effort. An Apgar score of less than

or equal to 5 (at 5 and 10 minutes after birth) clearly confers an increased relative risk

for HI brain injury9-11.

ii) Fetal Umbilical Artery pH

Fetal Umbilical Artery pH less than 7.0 and/or a base deficit worse than or equal to

minus 12 mmol/L from cord/arterial/venous/capillary blood gas obtained within 60

minutes of birth increase the probability of neonatal HI brain injury2, 10, 12.

iii) MRI

Magnetic resonance imaging (MRI) is the neuroimaging modality that best defines the

nature and extent of neonatal HI brain injury. Distinct patterns of MRI aberrations are

recognized in term neonates due to HI brain injury. Recently, electroencephalography

(EEG) has also shown helpful information for predicting the clinical outcome of HI

brain injury, though some studies reported that EEG is not as reliable as MRI13.

iv) Presence of Multisystem Organ Failure and Abnormal Neurological Signs

Dysfunction of multiple organs also results in a higher risk for neonatal HI brain

injury9. The dysfunctional organ systems include hematologic abnormalities, cardiac

dysfunction, metabolic derangements and gastrointestinal injury, or a combination of

Page 16: Effects of TRPM2 Inhibition in Neuroprotection following

3

any of above. Abnormal neurological signs are supplementary predictors of HI brain

injury. For example, hypotonic muscles or lack of a sucking reflex may also indicate a

probability of HI brain injury8.

1.3 Current therapy

There is currently only one available licensed treatment for HI: hypothermia14-16.

Hypothermia should be offered to term infants with moderate or severe HI brain injury within 6

hours of birth. 6 large randomized controlled trials (RCT) of hypothermia for neonatal HI brain

injury have been published, with all involved neonates being ≥36 weeks of gestation (termed

infants). The target temperature was 33°C to 34°C, with the cooling duration being 72 hrs14, 15, 17.

Rewarming was processed slowly, with an increase of 0.5°C per hour14, 15. Neonates underwent a

set of neurological examinations during the process as well as at the end of cooling. Some clinical

trials have demonstrated that either head cooling or whole body cooling reduces mortality or

disability either in all the infants or within a certain group of infants between 18 to 24 months of

age14-16, 18.

Even though hypothermia has shown impressive clinical outcomes and is currently well

established as a standard treatment for neonates suffering from moderate to severe HI brain injury,

it is stated to be “partially effective”19, 20. Therefore, there is an urgent need for novel therapeutic

opportunities beyond this current form of treatment.

Page 17: Effects of TRPM2 Inhibition in Neuroprotection following

4

2 Failure in Targeting Glutamate Receptors as Pharmacological

Targets

Although the exact pathophysiology of HI brain injury is not completely understood, it is

commonly accepted that a lack of sufficient blood flow in conjunction with decreased blood

oxygen content leads to loss of normal cerebral autoregulation and diffuse brain injury21, 22.

Glutamate is an important excitatory neurotransmitter at excitatory synapses in the CNS22-24. Based

on the theory of excitotoxicity, it has long been accepted that a lack of blood flow can lead to high

concentrations of glutamate release and eventually HI brain injury25. Therefore, glutamate

receptors have been extensively investigated as potential therapeutic targets for neuroprotection.

The biochemical cascade of the theory of excitotoxicity is summarized as follows22: Lack of

cerebral blood flow triggered energy failure and neuronal depolarization which then released large

amounts of glutamate into the extracellular space. Excessive glutamate in the extracellular space

overactivated NMDA (N-methyl-d-aspartic acid) and AMPA (dl-α-amino-3-hydroxy-5-methyl-4-

isoxazole propionic acid) glutamate receptors, leading to an increased influx of calcium.

Subsequently, intracellular calcium overload can be observed within neurons., which is thought to

induce neuronal cell death. Popular strategies against excitotoxicity have targeted the

pharmacological blockage of NMDA receptors. Whereas several compounds including

dezocilipine maleate (MK-801), aptiganel hydrochloride (Cerestat), dexthrometorphan (DMX)

and CGS 19755 (Selfotel) demonstrated promising neuroprotective effects in rodent models21, 26,

all clinical trials aimed at using NMDA and AMPA glutamate receptors as pharmacological targets

failed to yield the expected protective outcomes22, 27.

One potential explanation for this failure is that the injured brain may attempt to recruit

endogenous recovery mechanisms22, 28. While glutamate signals may be truly neurotoxic to

Page 18: Effects of TRPM2 Inhibition in Neuroprotection following

5

neurons, this may suggest that some aspects of the signaling process may still have beneficial

effects. It is already known that synaptic NMDA receptors and extrasynaptic NMDA receptors

may have opposite effects following activation21, 29. The activation of synaptic receptors could

promote cell survival while the activation of extrasynaptic receptors can downregulate BDNF and

ultimately lead to cell death22, 30. Another potential limitation of this standard NMDA-AMPA

model is that it does not consider the roles of cells other than neurons, for example, astrocytes and

oligodendrocytes23, 31, 32. As astrocytes and oligodendrocytes also express NMDA and AMPA

receptors23, 33, they too are vulnerable to excessive glutamate and play important roles in glutamate

regulation. Additionally, glutamate is an essential neurotransmitter that is necessary for important

physiological processes. Therefore, the blockage of glutamate receptors during the treatment of

ischemic injury may lead to unwanted side effects. Studies have shown that the hypofunction of

NMDA receptors may be partially responsible for the memory loss associated with

aging34. Schizophrenia has also been reported in association with NMDA receptor dysfunction35.

Page 19: Effects of TRPM2 Inhibition in Neuroprotection following

6

Figure 1. Classical glutamate receptor (NMDA and AMPA receptor) model of neuronal cell

death. This classical glutamate receptor driven model indicates the potential roles of NMDA and

AMPA receptors involved in inducing neuronal cell death through excessive extracellular

glutamate. Overactivation of these two channels leads to intracellular calcium imbalance,

consequently inducing several pathways that eventually lead to neuronal cell death. This figure

was modified from Elaine Besancon et al., 2008.

Page 20: Effects of TRPM2 Inhibition in Neuroprotection following

7

Taken together, the traditional model of excitotoxicity emphasized treatment at the level of

glutamate receptor channels to curtail such events. However, the failure of targeting glutamate

receptors in all clinical trials indicated that new therapeutic targets needed to be identified for HI

brain injury.

3 Targeting Non-Glutamate Channels

Several non-glutamate ion channels, including transient receptor potential channels, acid-sensing

channels22, hemichannels36, volume-regulated anion channels37 and sodium-calcium exchangers

(NCX)38, 39 etc. have been implicated and thus identified as potential novel therapeutic targets for

ischemic brain injury.

3.1 Sodium-calcium exchangers

The Na(+)/Ca(2+) exchangers (NCX) are bi-directional transmembrane proteins that express

widely in the brain38. Under normal physiological conditions, the NCXs exchange one calcium ion

out of the cell with three sodium ions going into the cell38. Under pathophysiological conditions

like ischemia, the activity of NCXs can be reversed38, 40-42. Instead of transporting calcium ions

out of cells, they may alternatively transport them into cells. Since dysregulation of sodium and

calcium homeostasis is a fundamental hallmark following ischemic brain injury, the role of NCXs

under ischemia has been studied both in vitro and in vivo41-44. However, the conclusions remain

controversial. Some studies have shown that using NCXs blockers may reduce brain infarction in

in vivo stroke models43-45, while other reports brought up conflicting results that inhibition of

NCXs can lead to even worse infarction outcomes22, 42. These variations may in part be due to the

Page 21: Effects of TRPM2 Inhibition in Neuroprotection following

8

diverse responses of NCXs to differences in the severity of ischemic brain injury22. Under mild

ischemic brain injury conditions, the NCXs operate in transporting calcium ions out of cells.

Therefore, blockage of NCXs reduces calcium extrusion and ends up worsening calcium-mediated

cell injury45. On the other hand, severe ischemic brain injury conditions involving an overload of

intracellular sodium leads to the reverse where NCXs conduct calcium into the cells41, 46. Hence,

the blockage of NCXs under these severe ischemic brain injury conditions may potentially be

neuroprotective. Therefore, the targeting of these differential responses and the resulting beneficial

effects needs to be further elucidated.

3.2 Hemichannels

Hemichannels are proteins that are involved in forming gap junctions. One gap junction channel

is composed of two hemichannels, and each hemichannel consists of a hexamer from the connexins

transmembrane protein family47. The gating of gap junction channels is regulated by the

phosphorylation status of connexin proteins47, 48. Under normal conditions, the two hemichannel

components stay in a closed state while forming an open state for gap junction channels. Under

ischemic conditions, the active opening of hemichannels leads to a reduction in the opening of gap

junction channels which consequently results in decreased cell-cell communication36, 48, 49.

Research on hemichannels has elucidated their clear involvement in the response to ischemia brain

injury. However, their precise role remains elusive and controversial. Some studies have shown

that knocking out connexin 43 (Cx43) in mice results in an increased level of brain infarct volume

and apoptosis following stroke47, 50-54. In contrast, other studies report that under specific

conditions, gap junctions exacerbate ischemic brain injury by spreading cytotoxic substances into

cells22, 50, 55. Further validation on the role of hemichannels is warranted before we can truly assess

them as therapeutic targets for stroke and brain injury.

Page 22: Effects of TRPM2 Inhibition in Neuroprotection following

9

3.3 Volume-regulated anion channels

Chloride (Cl-) permeates the cell membrane through several types of Cl– channels. An important

class of Cl– channels is the volume-regulated anion channel (VRAC). VRACs are responsible for

mediating the swelling-induced Cl– current37, 56, which also plays essential role in the regulatory

mechanism in cells for balancing cell volume during osmotic perturbations57-59. Under normal

physiological conditions, VRACs stay in a closed state58-60. Under pathophysiological conditions

like ischemia, VRACs are abnormally overactivated58. Once overactivated, subsequent

pathological mechanisms can be triggered, including the inhibition of NCXs due to ATP

depletion22. VRACs inhibitors have shown to provide a neuroprotective effect following stroke in

rats61-64, which supports the hypothesis that the activation of VRACs can be neurotoxic. DCPIB,

a VRACs specific inhibitor, was recently shown to have a neuroprotective effect in several rodent

models of hypoxic-ischemic brain injury62.

3.4 Acid-sensing ion channels (ASICs)

Acidosis, which worsens neurotoxicity, is a featured outcome that always follows ischemia65, 66.

Acid-sensing ion channel 1a (ASIC1a) is highly expressed in the brain and is believed to be

involved in acidosis induced brain injury65, 67. During ischemic conditions, the accumulation of

lactic acid rapidly decreases the pH level of the brain to 6.2 or even lower, and subsequently

activates ASICs68, 69. In vitro and in vivo studies have shown that the blockage of ASICs results in

neuroprotective effects. In particular, an in vitro study showed that ASIC currents and ASIC

desensitization could be amplified following oxygen-glucose deprivation (OGD), which increased

the length of calcium influx70. Other in vivo studies have shown that knockouts of ASIC1 in mice

protected animals from acidosis-reduced brain injury, and the administration of ASIC blockers

Page 23: Effects of TRPM2 Inhibition in Neuroprotection following

10

reduced brain infarct volume following MCAO brain injury68, 71. Hence, evidence suggests ASICs

as potential therapeutic targets for the inhibition of ischemic neuronal death.

3.5 Transient receptor potential melastatin (TRPM) subfamily

The transient receptor potential melastatin (TRPM) protein family is one of 6 subfamilies among

the TRP channels superfamily72. Two members of the family, TRPM7 and TRPM2, have been

implicated in mediating neuronal cell death73-78. In vitro pharmacological blockage79 and in vivo

siRNA suppression of TRPM7 both resulted in neuroprotective effects80. Recently, in vivo studies

have investigated the role of TRPM2 channels in hypoxic-ischemic brain injury and the results

showed that knockouts of TRPM2 channels in rodent models have neuroprotective effects81-83. The

underlying mechanism of TRPM family-mediated ischemic brain injury still remains unknown.

Since TRPM proteins are calcium permeable ion channels, dysregulation of calcium levels and

overload of intracellular calcium levels are the most acceptable mechanisms78, 83, 84. However,

TRPM ion channels as TRPM7 are also permeable to other ions such as zinc85, which has also

been illustrated to play a role during the hypoxic ischemic cascade. In this study, our focus is on

the role of TRPM2 channels during neonatal hypoxic ischemic brain injury.

Page 24: Effects of TRPM2 Inhibition in Neuroprotection following

11

Figure 2. Potential mechanisms involved in excitotoxicity following ischemic stress. The left

half of the figure shows the traditional glutamate driven model of excitotoxicity. The right half of

the figure shows that more and more evidence nowadays suggests that despite the traditional

model, non-glutamate driven channels including NCXs, hemichannels, VRACs, ASICs, TRPs

may play important roles in mediating the excitotoxicity following ischemic stress. This figure

was modified from Elaine Besancon et al., 2008.

Page 25: Effects of TRPM2 Inhibition in Neuroprotection following

12

4 Transient Receptor Potential Channels (TRPs)

As mentioned above, the TRPM channel subfamily is one of 6 subfamilies among the TRP channel

superfamily. The TRP channel was first identified as a protein in Drosophila melanogaster86. TRP

channels are non-selective cation channels that can be grouped into 6 families named TRPC

(canonical), TRPM (melastatin), TRPV (vanilloid), TRPP (polycystin), TRPML (mucolipin) and

TRPA (ankyrin) 72, 86, 87. There are 28 mammalian TRP channels that have been identified to date88.

All TRP channels consist of six transmembrane domains arranged in a tetrameric structure, and

they are widely expressed in various cell types in the body including neurons87, 88.

5 TRPM2 Channel

5.1 TRPM2 protein structure, transmembrane topology and

distribution

TRPM2 is a calcium-mediated nonselective cation channel. TRPM2 channels are expressed in

many tissues including the brain (high expression), lung, liver and heart89, 90. TRPM2 also

expresses in various cell types, including neurons, microglial cells, immune cells and pancreatic

β-cells78, 89.

TRPM2 proteins are encoded by TRPM2 genes. In rodents, the TRPM2 gene consists of 34 exons

and spans around 61 kb. In human, the TRPM2 gene consists of 32 exons and spans about 90 kb,

with the location of the gene being on chromosome 21q22.390. There is an additional exon located

at the 5’ terminus with a CgG island in the human TRPM gene. The full length transcript of

TRPM2 is approximately 6.5 kb and encodes TRPM2 protein comprising of 1503 amino acids

Page 26: Effects of TRPM2 Inhibition in Neuroprotection following

13

Figure 3. The TRP channel superfamily. The TRP channel superfamily comprises of 6

subfamilies including TRPC (canonical), TRPM (melastatin), TRPV (vanilloid), TRPP

(polycystin), TRPML (mucolipin) and TRPA (ankyrin).

Page 27: Effects of TRPM2 Inhibition in Neuroprotection following

14

with a molecular weight of 170 kDa90. In addition to full length TRPM2 transcripts, four splice

variants of TRPM2 have been identified: TRPM2-ΔN, TRPM2-ΔC, TRPM2 –S and TRPM2-

SSF89-91. Consistent with their names, TRPM2-ΔN is loss of amino acids 538–557 in the N-

terminus; TRPM2-ΔC is loss of amino acids 1292–1325 in the C-terminus, particularly the CAP

domain of the NUDT9-H domain; TRPM2-S (short) is loss of the entire C terminus including the

channel pore; TRPM2-SSF (striatum short form) is loss of the first 214 amino acids of the N-

terminal and has been found to uniquely express within the striatum89-91.

The TRPM2 protein structure consists of six transmembrane segments (S1-S6) with a pore loop

region located between S5 and S690. To form a channel, TRPM proteins typically assemble into

homotetramers with both N- and C- termini flanking the intracellular sides90. At the N-terminus,

there are 4 homologous regions and a calmodulin (CaM) binding domain, which is a region that

plays an important role in regulating the channel activation property90. At the C-terminus, there is

a TRP box as well as a coiled-coil domain, both of which are assumed to be essential for TRPM2

homogenous tetrameric assembly90.

Page 28: Effects of TRPM2 Inhibition in Neuroprotection following

15

Figure 4. TRPM2 protein structure and variants. The upper panel of the figure shows a

representative structure of the TRPM2 channel and its topology. The lower panel of the figure

shows different forms of TRPM2, including the full-length long form TRPM2 (TRPM2-FL),

TRPM2 cleavage of N terminus K538-Q557 (TRPM2-ΔN), TRPM2 cleavage of C terminus T-

1292-L1325 (TRPM2-ΔC), TRPM2 short striatum variant that has 214 residues missing from the

C terminus (TRPM2-SSF) and TRPM2 cleavage of the entire C terminus (TRPM2-S). This figure

was modified from Lin-Hua Jiang et al., 2010.

Page 29: Effects of TRPM2 Inhibition in Neuroprotection following

16

5.2 TRPM2 biophysical properties and gating mechanism

The TRPM2 channel exhibits a linear I/V curve, which suggests that channel activity is

independent from voltage-gating87, 92. Instead, TRPM2 is a ligand-gated channel that can be

activated by several intracellular and extracellular features, among which ADPr and hydrogen

peroxide are the most potent activators89, 90.

Hydrogen peroxide can directly and indirectly activate TRPM2 channels. The ability of H2O2 to

activate TRPM2 channels has attracted significant scientific interest. Studies have used the H2O2

driven mechanism for explaining the pathological processes that are mediated by elevation of the

oxidative microenvironment79, 93. Such pathological processes include hypoxic ischemic (HI) brain

injury, diabetes, inflammation and other neurodegenerative disorders like bipolar diseases.94, 95

Endogenously, H2O2 is initially generated from mitochondria following oxidative

phosphorylation96. Exogenously, generation of H2O2 is induced as a result of responding to

external factors such as certain drugs, heavy metals, visible light or even heat, in consistence with

other reactive oxygen species (ROS) like hydroxyl radicals (OH.)97-99. Overgeneration of ROS

results in dramatic damage to biological molecules such as DNA and proteins, or any molecule

that is involved in the chain reaction cascade producing cellular damage and disease.

In terms of activating the TRPM2 channel indirectly, hydrogen peroxide activates the TRPM2

channel through regulation of the metabolic pathway that produces ADPR100. ADPR can bind to

TRPM2 at the active site in the NUDT9-H region at the C-terminus90, 100. The extracellular

stimulation of hydrogen peroxide leads to an intracellular increase in hydrolase activity, which

thereby hydrolyzes more NAD+ and cADPR to produce additional ADPR90. Another source of

ADPR is the action combination of poly (ADPR) polymerases (PARPs, PARP enzymes) and poly

(ADPR) glycohydrolases (PARG enzymes)89, 90, 101. This source indirectly generates ADPR

Page 30: Effects of TRPM2 Inhibition in Neuroprotection following

17

through the formation and hydrolysis of poly-ADPR when it is overactivated in response to DNA

damage89, 90, 100, 101. At the TRPM2 N-terminus, calcium can bind to the CaM-binding motif, which

is another mechanism of gating that is independent of ADPR and hydrogen peroxide89, 90, 102.

5.3 The physiological and pathophysiological role of TRPM2

channels

As mentioned above, TRPM2 has been identified in several different cell types including neurons,

immune cells and pancreatic β-cells. Therefore, it is not surprising that TRPM2 is associated with

ailments such as CNS diseases and type II diabetes78, 103, 104. However, the precise mechanisms of

these pathologies still require further investigation. In this case, oxidative stress78, 105-107 and

amyloid beta108-110 mediated pathological activation of TRPM2 channels seem the most likely

mechanisms. TRPM2 is highly permeable to calcium, and can mobilize calcium ions from both

the extracellular and intracellular spaces. Hence, its biological significance is strongly associated

with the intracellular calcium level. Under normal physiological conditions, the calcium-mediated

activity of the TRPM2 channel has been reported to be involved in several physiological processes,

including inflammation111, synaptic transmission112, microglial activation113 and insulin

secretion114. Under pathophysiological conditions, the abnormal overactivation of the TRPM2

channel may lead to intracellular calcium overload, which can subsequently lead to various

diseases.

In the CNS, TRPM2 is most abundantly expressed in the brain where it has been implicated in

triggering numerous physiological and pathophysiological processes. For example, TRPM2 is

involved in mediating neuronal cell death that can consequently lead to CNS diseases including

stroke, Alzheimer’s disease and bipolar disorder. One study has shown that patients with bipolar

Page 31: Effects of TRPM2 Inhibition in Neuroprotection following

18

disorders have relatively higher levels of basal intracellular calcium ions and that the TRPM2 gene

sites are located within chromosome region 21q22.3, conferring increased susceptibility to this

pathology95. Another study showed that knocking out TRPM2 reduced ischemic brain damage in

MCAO model of adult mice83. Our lab also recently demonstrated that TRPM2 knockouts provide

a neuronal protective effect following HI brain injury in neonatal mice115. Together, these previous

studies indicate that TRPM2 is a promising therapeutic target for the treatment of HI brain injury.

Therefore, my project will examine the effects of TRPM2 inhibition on neuroprotection that may

lead to potential drug development for neonatal hypoxic-ischemic brain injury.

In addition to the CNS, TRPM2 has also been identified in pancreatic β-cells114. Activation of

TRPM2 channels has also been linked to insulin secretion and H2O2-induced apoptosis of insulin-

secreting cells, implicating a potential role of TRPM2 in diabetes. A recent study using the TRPM2

knockout mouse model revealed the involvement of the channel in insulin secretion from

pancreatic β-cells. In this case, TRPM2 knockout mice demonstrated relatively higher basal blood

glucose levels in comparison to WT mice, while plasma insulin levels remained similar114.

TRPM2 has also been identified in some cell types in the immune system, including macrophages,

neutrophils and lymphocytes, suggesting a possible association with inflammatory diseases90, 116.

Page 32: Effects of TRPM2 Inhibition in Neuroprotection following

19

Figure 5. Proposed mechanisms of TRPM2 channel activation by H2O2 and involvement

TRPM2 channel activity in physiological and pathophysiological processes. H2O2 can activate

the opening of TRPM2 channels directly and indirectly. Activation of the TRPM2 channel leads

to extracellular calcium influx, which may further facilitate the channel opening. Influx of calcium

leads to intracellular calcium imbalance, which suggests the involvement of TRPM2 channels in

physiological processes such as insulin release, cytokine production, increased endothelial

permeability and cell death. Therefore, the actions of TRPM2 may contribute to pathologies or

disease conditions. +: activation. This figure was modified from Lin-Hua Jiang et al., 2010.

Page 33: Effects of TRPM2 Inhibition in Neuroprotection following

20

6 Pharmacological Interactions

As there is significant interest in the exploration of TRPM2 as a potential target for

neurodegenerative diseases, the pharmacology of TRPM2 has also started to receive considerable

attention in the field of research. Through patch clamp electrophysiological techniques and the

availability of HEK293 cells, different TRPM2 channel inhibitors have been studied. Thus far,

none have demonstrated a desirable specificity with compounds that have been reported to have

an inhibitory effect on the TRPM2 channel also affecting other TRP channels89.

6.1 Flufenamic acid (FFA)

Flufenamic acid (FFA) was the first TRPM2 blocker identified117. It belongs to class of non-

steroidal anti-inflammatory drugs (NSAIDs). Such fenamates are capable of producing anti-

inflammatory effects in the CNS. Studies have been conducted on TRPM2-overexpression

HEK293 cells, where FFA evoked a pH-dependent inhibition of ADPR- or H2O2-induced cation

currents117. However, the inhibitory effect of FFA is not limited to the TRPM2 channel; it can

similarly affect other channels in the TRP family including TRPM4, TRPM5, TRPC3 and

TRPC590, 117. Furthermore, it also has an activation effect on TRPC6 and transient receptor

potential ankyrin 1 (TRPA1)90, 117. Therefore, fenamates such as FFA are hardly satisfactory tools

in clarifying the role of the TRPM2 channel.

6.2 Anti-fungal agents (clotrimazole and econazole)

Anti-fungal compounds block TRPM2 channels activated by ADPR in TRPM2-overexpressing

HEK293 cells, but the inhibitory effect is irreversible89, 118.

Page 34: Effects of TRPM2 Inhibition in Neuroprotection following

21

6.3 2-APB

2-APB was first identified as an inositol 1,4,5-trisphosphate(IP3) receptor antagonist. It has also

been reported to exert an inhibitory effect on certain TRPC and TRPM channels while having an

activation effect on some TRPV channels24, 117, 119.

6.4 Divalent heavy metal cations

Some heavy metal ions, La3+ and Gd3+ for instance, are known to have an inhibitory effect on most

TRP channels, though TRPM2 seems to be an exception. A recent study shows that divalent copper

(Cu2 + ) may be a potent TRPM2 channel blocker98.

6.5 AG490

Recently, AG490 was identified to have an inhibitory effect on the TRPM2 channel120. AG490

was shown to almost completely block H2O2-induced intracellular Ca2+ increase and significantly

reduce H2O2-induced TRPM2 currents120. While H2O2 can also activate the TRPA1 channel,

AG490 has no significant effect on the H2O2-induced Ca2+ influx mediated by TRPA1 channels.

ADPR is another endogenous activator for TRPM2. However, in a patch clamp study on TRPM2-

overexpressing HEK293 cells, AG490 only inhibited H2O2-induced but not ADPR- induced

TRPM2 inward current120. Structurally, AG490 belongs to the tyrphostin family and was

synthesized in the early 1990s. Since AG490 was initially found to be a JAK2 inhibitor, the same

study examined whether the inhibitory effect of AG490 on TRPM2 activity was dependent on the

inhibition of JAK2. The study tested the effects of other JAK2 inhibitors, none of which had an

effect on H2O2-induced Ca2+ increase by TRPM2 channels. Thus, the results suggested that the

inhibitory effect of AG490 on TRPM2 activation is independent from JAK2 inhibition120. Hence,

AG490 may manifest its inhibitory effect by scavenging intracellular hydroxyl radicals.

Page 35: Effects of TRPM2 Inhibition in Neuroprotection following

22

Figure 6. TRPM2 channel inhibitors. Shown above are the chemical structures of compounds

that have been indicated to have an inhibitory effect on TRPM2 channels. FFA, anti-fungal agents

(clotrimazole and econazole) and 2-APB are the classical TRPM2 inhibitors. However, studies

have shown their inhibitory effects to be unsatisfactory. AG490 is a newly discovered TRPM2

inhibitor which inhibits activation by efficiently blocking TRPM2 currents.

Page 36: Effects of TRPM2 Inhibition in Neuroprotection following

23

Chapter 2 Rationale and Hypothesis

Rationale: HI brain injury is a severe public health issue with no effective pharmacological

method of prevention thus far. It has been previously confirmed that TRPM2 plays a

neuroprotective role in adult cerebral ischemia using TRPM2 KO mice83. Our lab has also recently

shown that knocking out TRPM2 provided a neuroprotective effect following HI brain injury in

neonates115. Therefore, I proposed to test the effects of a TRPM2 channel inhibitor in HI brain

damage in relation to potential drug development for HI brain injury.

AG490 is a recently identified TRPM2 current inhibitor that efficiently blocks H2O2 induced

TRPM2 current in HEK cells120. AG490 was initially discovered as a JAK2 inhibitor but could

inhibit TRPM2 in a JAK2 independent manner120. In this case, AG490 may act as a ROS scavenger

and reduce the concentration of hydrogen peroxide: . It was previously

reported that silencing TRPM2 using siRNA reduced H2O2-induced neuronal cell death in vitro79.

It is also reported that following hypoxic-ischemic injury, there is an accumulation of H2O2 in the

neonatal brain at postnatal day 7 (neonatal mice brains) which is not been seen in the postnatal day

42 mouse brain (adult mice brains)121. Thus, it is expected that TRPM2 may play a greater role in

neonates compared to adult, and AG490 could be a suitable pharmacological tool for my project

Hypothesis: I hypothesize that inhibition of TRPM2 channels by TRPM2 inhibitor AG490

provides neuroprotection following hypoxic-ischemic brain injury in neonates.

Page 37: Effects of TRPM2 Inhibition in Neuroprotection following

24

Chapter 3 Aims and Experimental Design

Aims:

AIM1: Investigate the effect of TRPM2 inhibition on H2O2-induced neuronal cell death in vitro.

AIM2: Investigate the effect of TRPM2 inhibition on neonatal hypoxic-ischemic brain injury in

vivo.

Experimental Design Outline:

Figure 7. An outline of the project experimental design.

Page 38: Effects of TRPM2 Inhibition in Neuroprotection following

25

Chapter 4 Materials and Methods

1 Ethics Approval

All protocols were carried out strictly accordant to Canadian Council on Animal Care (CCAC

protocol) guidelines. Protocols were approved by the local Animal Care Committee (Office of

Research Ethics, University of Toronto). All of the experiments have been reported using the

ARRIVE guidelines.

2 Animals

In considering the day of birth as postnatal day 0 (P0), pups used in this study were P7. Timed-

pregnant CD-1 mice were purchased from Charles River Laboratories (Sherbrooke, QB, Canada).

Mice were housed under temperature condition of 20 ± 1°C and a 12 hrs light/dark cycle with free

access to a standard laboratory chow diet and water.

3 Reagents

TRPM2 inhibitor AG490 (CAS #82749-70-0) was purchased from Tocris (BRS, UK). 30 %

Hydrogen Peroxide (HC4060-500ML) was purchased from Biobasic (Amherst, NY, USA). Cresyl

violet, 2,3,5- triphenyl- 2H- tetrazolium chloride (T8877, TTC) and Dimethyl sulfoxide (D2650,

DMSO) were purchased from Sigma- Aldrich (St. Louis, MO, USA).

4 Cell Culture

TRPM2 overexpression HEK293 were cultured as follows: doxycycline-inducible HEK293 cells

with stable expression of TRPM2/ pCDNA4 were cultured with DMEM supplemented with 10 %

FBS, 1 % antibiotic-antimyocotic, blasticidin (5μg/ml, Sigma-Aldrich, MO, USA) and zeocin

Page 39: Effects of TRPM2 Inhibition in Neuroprotection following

26

(0.4mg/ml, Invitrogen, USA). TRPM2 expression was induced by adding 1 μg/ml doxycycline

(1μg/ml, Sigma-Aldrich, USA) to the culture at least 24 hrs before experiments.

Embryonic primary cortex neurons were cultured from E16 CD-1 mice. Dissected cortexes were

digested with 0.025 % trypsin/EDTA at 37°C for 15 min. Cell density was determined using an

Improved Neubauer hemocytometer, and 1.0× 104 cells were plated on poly-D-lysine coated glass

coverslips (12 mm no. 1 German Glass, Bellco cat. no. 1943-10012, Sigma-Aldrich, USA). The

cells were kept in 5 % CO2 at 37°C in culture medium (Neurobasal medium supplemented with

1.8 % B27, 0.25 % Glutamax, and 1 % antibiotic-antimyocotic).

5 In vitro H2O2-induced Neuronal Cell Death Model

The H2O2-induced cell death in vitro model was carried out using CD-1 E16 embryos. Dilutions

of H2O2 were made from a 30 % stock solution into culture medium prior to each experiment.

Exposures to H2O2 were performed by simple addition of a specific volume of H2O2 diluted in

culture medium at x100 directly to each well in a 96-well plate. After exposure to H2O2, the plate

was kept in 5 % CO2 at 37°C for 24 hrs before biochemical measurements were performed.

6 Cell Viability Assay

Cell viability was assessed by MTT assay. MTT, a yellow tetrazole, will be reduced by NAD(P)H-

dependent cellular oxidoreductase enzymes to insoluble formazan with a purple color when

incubating with culture medium. The ratio of yellow MTT to purple formazan indicates the amount

of viable cells. Cells were plated on 96-well culture plates with density of 5×104 cells/ml. Cells

were treated by adding various concentrations of AG490 and incubated with AG490 for 24 hrs.

MTT (0.5 mg/ml MTT in PBS) was diluted with culture medium with a dilution ratio of 1:10 and

added to each well. After 3 hrs of incubation, the medium was removed from each well and 100

Page 40: Effects of TRPM2 Inhibition in Neuroprotection following

27

μl DMSO was added. The absorbance at 490 nm was measured in a microplate reader (Syngery

H1, Biotek, USA). Cell viability was expressed as a percentage of the control.

Cell death assessment was carried out by using propidium iodide (PI) staining. Propidium iodide

is a fluorescent intercalating agent that can only cross the membrane when the cells are necrotic,

therefore, it can be used to stain cells and differentiate cells with different living status (either

necrotic or healthy). Propidium iodide (PI, 1 μg/ml) was added in living cultures. After 24 hrs, the

fluorescent intensity was measured by the Synergy HT Multi-Mode Micro Plate Reader.

7 Electrophysiology (Whole Cell Patch Clamp)

Whole-cell patch-clamp experiments were carried out using an Axopatch 700B (Axon

Instruments, Inc.) to examine the effect of AG490 on TRPM2 current in TRPM2 over-expression

HEK293 cells. TRPM2 over-expression HEK293 cells were induced with 1 μg/ml doxycycline at

least 24 hrs before whole cell patch clamping. Currents were recorded using a 400 ms voltage ramp

protocol (-100 to +100 mV) with an interval of 5 s at 2 kHz and digitized at 5 kHz. Pipette solution

containedv(in mM): 145 cesium methanesulfonate, 8 NaCl, 10 EGTA, and 10 HEPES, pH adjusted

to 7.2 with CsOH. Bath solution contained (in mM) 140 NaCl, 5 KCl, 2 CaCl2, 20 HEPES, and 10

glucose (pH adjusted to 7.4 with NaOH). After filling with pipette solution, patch pipette resistance

was between 5-9 megaohms. pClamp 9.2 software was used for data generation and Clampfit 9.2

was used for data analysis. All experiments were carried out at room temperature.

8 Drug Administration

In vitro administration: Various concentrations of AG490 were prepared by dissolving the

compound in B27-free culture medium. AG490 was added to the culture media prior to exposure

to H2O2.

Page 41: Effects of TRPM2 Inhibition in Neuroprotection following

28

In vivo administration: Pups with body weights of 5 g were randomly assigned into: sham control

group (Sham), HI + vehicle (Vehicle, 5 % DMSO and 5 % Tween-80 in 0.9 % saline) or HI +

AG490 (AG490, 30 mg/kg). AG490 was dissolved in 5 % DMSO and 5 % Tween-80 (P-8074) in

0.9 % saline for the final concentration of 30 mg/kg. AG490 or vehicle control was administered

to the pups 20 min prior to ischemia induction for pre-treatment as well as right after ischemia

induction for post-treatment 1 and immediately after hypoxia for post-treatment 2. The compound

was administered intraperitoneally (i.p.) in a volume of 20 μl/g (injection ratio to body weight).

9 In vivo Hypoxic-Ischemic Mouse Model

Mouse hypoxic-ischemic (HI) model was performed according to a well described protocol with

modifications77, 80, 122. Postnatal day 7 (P7) mice were anesthetized with isoflurane (3.0 % for

induction and 1.5 % for maintenance). The whole process contained two main parts: ischemia and

hypoxia. Ischemia was carried out by isolation of the right common carotid artery and then ligation

with a bipolar electrocoagulation device (Vetroson V-10 Bi-polar electrosurgical unit, Summit Hill

Laboratories, Tinton Falls, NJ, USA). The remaining ligated artery was cut using microscissors.

Pups were then returned to their dam and allowed to recover for 1.5 hrs. After recovery, the

hypoxia process was then achieved by placing the pups in a 37°C chamber (A-Chamber A-15274

with ProOx 110 Oxygen Controller/E-720 Sensor, Biospherix, NY, USA) perfused with a gas

mixture of 7.5 % oxygen and 92.5 % nitrogen for 60 min. A homoeothermic blanket control unit

(K-017484 Harvard Apparatus, MA, USA) was used to monitor the chamber temperature. Animals

in sham groups only undergo exposure of common carotid artery under anesthesia but not ligation

and hypoxia.

Page 42: Effects of TRPM2 Inhibition in Neuroprotection following

29

10 Infarct Volume Measurement, Whole Brain Imaging and

Histological Assessments

10.1 TTC staining/Infarct volume measurement

24 hrs after the HI, brain tissues were collected and coronally sectioned into four ~1 mm slices.

These slices were stained with 2,3,5-triphenyltetrazolium chloride (TTC), a redox indicator

(indicate cellular respiration for differentiating between metabolically active and inactive tissues),

to visualize the infarct area. Slices were stained with 1.5 % TTC and placed in a dark incubator

maintained at 37 oC for 20 min. The infarct areas were traced using image analysis software

(ImageJ). Infarct volume will be calculated by summing the representative areas in all brain

sections and multiplying by the slice thickness. After correcting for edema, the infarct volumes

will be calculated as follows: Corrected infarct volume (CIV), (%) = [contralateral hemisphere

volume - (ipsilateral hemisphere volume - infarct volume)] /contralateral hemisphere volume *

100 %.

10.2 Whole brain imaging/Nissl staining

7 days after the HI, whole brain tissues were collected and imaged to reveal morphological changes

between the groups. At this stage, the infarct areas in the brains underwent liquefactive necrosis

and the severity of the brain damage would be quantified. These brains were subsequently sliced

into ~100 µm coronal sections and stained with 1 % Cresyl violet (Nissl) to indicate histological

brain damage. The infarct areas were traced using image analysis software (ImageJ). Infarct

volume will be calculated as follows: infarct volume (IV), (%) = infarct volume/contralateral

hemisphere volume * 100 %.

Page 43: Effects of TRPM2 Inhibition in Neuroprotection following

30

11 Neurobehavioral Assessments

Short term neurobehavioral tests (geotaxic reflex, cliff aversion and grip test) were carried out to

assess the recovery outcomes of the HI on P1, P3 and P7 days after HI. These reflexes were chosen

because they represent the earliest stages of development in mice and are good indicators of

sensorimotor function. Specifically, 1) geotaxis reflex studies for vestibular and proprioceptive

function123; 2) cliff aversion reflex tests the maladaptive impulse behavior123, 3) grip test assesses

force and fatigability123. These neurobehavioral tests for the determination of the functional

recovery of animals have been well-documented in previous studies from my own lab77, 124, 125.

Long term neurobehavioral test, passive avoidance test126, 127 was also used to assess contextual

fear learning, memory deficits, and also long term motor functional improvements. These abilities

are not well-developed until later in life, thus, they will be tested in post-surgery week 3, indicating

long term neurobehavioral recovery.

11.1 Geotaxis reflex

Geotaxis reflex is an automatic, stimulus-bound orientation movement. Pups were placed head

down in the middle of a board inclined with an angle of 45°. The latency for the pup to rotate 180°

was recorded.

11.2 Cliff avoidance test

Pups were placed on the edge of a platform, and the latency for the pup to remove both paws from

the edge, by turning away from the cliff was recorded.

Page 44: Effects of TRPM2 Inhibition in Neuroprotection following

31

11.3 Grip test

Pups were suspended by their forepaws on a wire stretched over a cotton pad in a cage. The latency

for the pup to fall was recorded.

11.4 Passive avoidance test

Passive Avoidance Test is a 3 day, 1 trailed long-term behavioral test for testing both the motor

functional recovery as well as learning and memory recoveries. 3 weeks after the HI injury, mice

in all three groups (sham, HI + vehicle, HI + AG490) underwent the Passive Avoidance Test. The

protocol consists of day 1 habituation, day 2 acquisition/conditioning and day 3 testing. The

apparatus consists of two parts: a large (250 (W) x 250 (D) x 240 (H) mm) illuminated

compartment and a small (195 (W) x 108 (D) x 120 (H) mm) dark compartment with electrified

grid floor (LE872, Panlab, Harvard Apparatus, BCN, Spain). The two compartments are separated

by a guillotine gate. Mice have innate preference toward dark. During the habituation session, the

mouse was placed into the illuminated compartment and allowed to explore for 1 min. After 1 min,

the door for entering the dark compartment was opened, the latency of entering the dark room was

recorded and the door was closed for 30 s before the mouse was returned to its home cage. During

the acquisition/conditioning session, the mouse was allowed to explore the illuminated

compartment for 30 s and had access to the dark compartment. The mouse received a foot shock

(0.4 mA, 2 s) 3 s after entering the dark compartment. During the testing session, which took place

24 hrs after the acquisition/conditioning session, the mouse was placed into the illuminated

compartment, the door to the dark compartment was opened after 5 s and the latency to enter the

dark compartment was recorded (step-through maximal latency: 300 s). The latency to enter the

dark compartment during the retention session was taken as an index of memory performance.

Page 45: Effects of TRPM2 Inhibition in Neuroprotection following

32

12 Immunohistochemistry and Confocal Imaging

Brain tissues were collected 7 days after HI (P14) and fixed in 4 % paraformaldehyde/30 % sucrose

solution at 4°C overnight. Brains samples were sectioned coronally into ~50 μm slices using a

vibratome (Tissue Sectioning System Microtome Vibratome, HuiYou, China) which underwent

immunohistochemical staining. Samples were probed with mouse anti-neuronal nuclei (NeuN)

antibody (MAB377, 1:500; Chemicon, Temecula, USA) and anti-glial fibrillary acidic protein

(GFAP) (ab7260, 1:1000; Abcam, Cambridge, UK) antibodies overnight at 4°C. Next, the sections

were incubated with secondary antibodies Alexa 488 and 568 (#835724, #632115, 1:200; Cell

Signaling Technology) for 1 hr at room temperature and mounted on glass coverslips with ProLong

Gold antifade reagent (P36930; Thermo Fisher Scientific, Burlington, CA). Confocal laser

microscope (LSM700 Zeiss; Oberkochen, Germany) was used to image the immunostained brain

slices. Three brains per treatment group were collected, and 3 to 5 coronal slices per brain were

imaged. All the treatment groups were imaged at the same laser settings with a 40× lens. Cortical

areas directly adjacent to the injury site were imaged. At least 5 randomly chosen fields were

imaged and the number of cells per field was quantified using the Cell Counter plugin for ImageJ

software (National institute of Health, Bethesda, MD, USA).

13 Western Blot

24 hrs after HI, the ipsilateral and contralateral hemispheres of the mice brains were collected and

frozen in dry ice. To study the underlying mechanism of HI, protein was extracted from the cortex.

The brain samples were homogenized in RIPA buffer with a cocktail of proteinase and phosphatase

inhibitors, then incubated at 4 ºC for 1 h and centrifuged for 15 min at 13,000 rpm. The protein

concentrations were measured using the Bio-Rad Protein Assay reagent (Bio-Rad, Hercules, CA).

Samples of the mouse brain (30 μg) were separated on a 10 % SDS-PAGE gel that was transferred

Page 46: Effects of TRPM2 Inhibition in Neuroprotection following

33

to a nitrocellulose membrane (350 mA, 90 min). Blots blocking was carried out using 5 % non-fat

milk in Tris-buffered saline (TBS). Blots were incubated with primary antibodies at 4ºC overnight

and secondary antibodies at room temperature, respectively anti-phospho-Akt (#9271S, Ser473,

1:1000); anti-Akt (#9272S, 1:1000); anti-GAPDH (#2118S, 1: 10,000). Protein signals of interest

were tested using enhanced chemiluminescent reagents (PerkinElmer, Mass, USA) and analyzed

through exposure to film (Helot CL, NJ, USA).

14 Statistics and Data Analysis

Data were presented as means ± SEM. Student’s t-test was performed to assess the statistical

significance of the difference in 2 groups between vehicle group and the AG490 treated group. In

multiple groups, one-way ANOVA following with the Bonferroni test were used. Significance was

defined by the probability level of lower than 0.05 (P<0.05).

Page 47: Effects of TRPM2 Inhibition in Neuroprotection following

34

Chapter 5 Results

1. The level of TRPM2 mRNA expression in the hippocampus and

cortex increases consistently with the developmental age

In order to choose the optimal materials and specific time points for my study, I extracted in situ

hybridization data from the Allen Brain Atlas and searched for the developmental pattern of

TRPM2 in the brain. Due to the lack of availability of developing mouse brain TRPM2 data, I

calculated corresponding mouse developmental age as described in Workman et al., 2013 and used

data from BrainSpan atlas151 of the Developing Human Brain.

From the extracted data, I first confirmed TRPM2 expression in the brain with high expression in

the hippocampus and cortex. The mRNA expression in these two specific regions reach a peak

point at 1 year of age (Figure 8). In my study, I chose the cortex for cell culture in my in vitro

experiments. TRPM2 reaches highest expression level at 37 weeks after conception, which is a

time period equivalent to P7 in a mouse. Therefore, I used P7 mice pups for my in vivo

experiments. There are abundant TRPM2 channel proteins for activation in the event of H2O2

accumulation in the neonatal brain121, and therefore the TRPM2 inhibitor AG490, which acts as a

hydroxyl radical, could be suitable for addressing my scientific questions.

Page 48: Effects of TRPM2 Inhibition in Neuroprotection following

35

Figure 8. TRPM2 expression in the cortex and hippocampus increases with development.

According to data extracted from the BrainSpan Atlas of Developing Human Brain for the TRPM2

gene, TRPM2 is highly expressed in the developing human brain. In the hippocampus and cortex

TRPM2 expression increases with age and peaks at 1 year of age. Each human developmental age

is correlated with the associated mouse age with similar whole brain development. Data are

expressed as mean log2 of reads per kilobase ± SEM. For cortical measurements n = 27, 11, 22,

11, 11. For hippocampal measurements n = 2, 1, 2, 1, 1. RPKM: reads per kilobase per million.

pcw: post conception week.

Page 49: Effects of TRPM2 Inhibition in Neuroprotection following

36

2 AG490 as a pharmacological inhibitor of the TRPM2 channel

AG490 structurally belongs to the tyrphostin family and was initially found to be a JAK2

inhibitor128. It has been recently reported by Mori and colleagues that AG490 significantly reduced

H2O2-induced TRPM2 activation, but not ADPR-induced TRPM2 current120. Therefore, AG490

is a suitable pharmacological tool to test the effect of inhibition of H2O2-activated TRPM2 channel

activity following neonatal HI brain injury.

To verify the efficiency of the inhibitory effect of AG490 on TRPM2 current, I first carried out

whole-cell patch clamp recording on TRPM2 overexpression HEK293 cells. 1 μg/ml doxycycline

was used to induce TRPM2 overexpression in HEK293 cells, with the cells being induced at least

24 hrs before whole cell patch clamping. As shown in Figure 9C, H2O2 (200 μM) elicited a large

and inwardly rectifying current in TRPM2 overexpression HEK293 cells with the current density

of 1497.97 ± 452.03 mA (n=3), whereas pretreatment of HEK293 cells with AG490 reduced the

current to 67.33 ± 31.63 mA (n=3; p=0.0343). Spontaneous TRPM2 current without exposure to

H2O2 was small (Figure 9A).

These results suggest that TRPM2 channel activity is sensitive to inhibition by AG490 and that

AG490 is a valid pharmacological tool for further in vitro and in vivo studies.

Page 50: Effects of TRPM2 Inhibition in Neuroprotection following

37

Figure 9. AG490 efficiently inhibited H2O2-induced TRPM2 current in TRPM2

overexpression HEK293 cells. A. Spontaneous TRPM2 currents in doxycycline-induced TRPM2

overexpression HEK293 cells. B. H2O2-induced TRPM2 currents in doxycycline-induced TRPM2

overexpression HEK293 cells. C. AG490 inhibited H2O2-induced TRPM2 currents in

doxycycline-induced TRPM2 overexpression HEK293 cells. D. Representative I-V trace (black

line is trace of bath solution; red line is trace of perfusion with 200 µΜ H2O2; blue line is trace of

pretreatment with 50 µΜ for 2 hrs and perfusion with 200 µΜ H2O2). E. Summary bar chart

comparing the H2O2-induced TRPM2 currents at +90 mV with and without application of AG490.

*represents p < 0.01 (Student's t-test, n = 3/group).

Page 51: Effects of TRPM2 Inhibition in Neuroprotection following

38

3 AG490 protects neurons from H2O2-induced cell injury in vitro

After verifying that AG490 efficiently blocks TRPM2 current activity in TRPM2 overexpression

HEK293 cells, I investigated the effects of AG490 on the viability and proliferation of cortical

neurons. Cortical neurons are vulnerable to H2O2. To test whether AG490 can protect neurons

from H2O2-induced cell death in vitro, cortical neurons were cultured and treated as described in

the methods section. The MTT assay was carried out to assess cell viability and propidium iodide

(PI) staining was performed to assess cell death. Hydrogen peroxide produced a progressive

apoptotic effect on cortical neurons in a dose-dependent manner from 6-100 μM (Figure 10A, p <

0.05, n=12). Figure 10B shows that AG490 pre-treatment (40 min prior to exposure to H2O2)

improved cell viability of cortical neurons at the optimum concentration of 50 μM. Propidium

iodide (PI) fluorescence intensity in cortical neurons was found significantly greater after exposure

to H2O2 in a dose-dependent manner from 6-50 μM (Figure 10C, p < 0.05, n=9). Figure 10D shows

that AG490 pre-incubation for 40 min significantly reduced PI fluorescence intensity at 50μM

(Figure 10D, p < 0.05, n = 6). This in vitro data indicates that AG490 could protect cultured

neurons from insult caused by H2O2 exposure.

Page 52: Effects of TRPM2 Inhibition in Neuroprotection following

39

Figure 10. TRPM2 inhibitor AG490 pre-treatment reduced neuronal cell death following

H2O2-induced cell injury. A & C. Hydrogen peroxide produced a progressive apoptotic effect on

cortical neurons in a dose-dependent manner from 6-100 μM after 24 hrs of incubation with

AG490. B & D. 50 μM AG490 significantly protected neurons from H2O2-induced injury (CTL,

control; Results are mean ± SEM; *, versus CTL group, #, versus non-treated group, p<0.05; One-

way ANOVA with subsequent Bonferroni test, n as indicated on the bars).

A B

C D

Page 53: Effects of TRPM2 Inhibition in Neuroprotection following

40

4 The Effect of AG490 Pre-treatment on Hypoxic-Ischemic Brain

Injury in vivo.

The neonatal hypoxic-ischemic injury model was implemented on postnatal 7-day old (P7) CD-1

mice. AG490 (30 mg/kg) or vehicle were administered as a single intraperitoneal injection to P7

pups according to the timeline in Figure 11.

Figure 11. Timeline of neonatal hypoxic-ischemic injury and experimental procedures.

Postnatal 7-day old pups (P7) were randomly grouped into 3 groups (sham, vehicle, AG490) and

injected with either vehicle or AG490 (no injection for sham group) 20 min prior to ischemia

induction for pre-treatment and immediately after ischemia induction for post-treatment 1 and

immediately after hypoxia for post-treatment 2. This was followed by 90 min of recovery and 60

min of hypoxia with 7.5 % O2 and 92.5 % N2. TTC staining and western blot were performed 24

hrs after HI (P8). Whole brain imaging, Nissl staining and immunohistochemistry was performed

7 days after the HI (P14). Short term neurobehavioral assessment was performed 1 day, 3 days and

7 days after HI (P8, P10, P14). Long term neurobehavioral assessment was performed 3 weeks

after the HI (P35).

Page 54: Effects of TRPM2 Inhibition in Neuroprotection following

41

4.1 Pre-treatment with TRPM2 inhibitor AG490 reduced the brain

infarct volume of hypoxic-ischemic brain injury in vivo.

After confirming that AG490 delivered neuroprotective effect following H2O2-induced cell death,

I next investigated whether AG490 generated neuroprotective effects in vivo. By using a mouse

neonatal hypoxic-ischemic brain injury model followed by TTC staining, I found that a lower dose

of AG490 (15 mg/kg, i.p., 20 min before HI) did not afford neuroprotection (Figure 13). However,

AG490 pre-treatment with a higher dose (30 mg/kg i.p., 20 min before HI) significantly reduced

brain infarct volume in comparison to the vehicle treated group (Figure 13). TTC is a redox

indicator and it stains for metabolically active tissues. The white area represents the infarct area

and illustrates the damaged tissue which is not metabolically active. TTC staining was carried out

on coronal sections of mouse brains 24 hrs after HI. Representative images of TTC staining are

shown in Figure 13C, where the white un-stained areas indicated the infarct volume. Infarct

volume in the vehicle-treated HI group (Vehicle) was 58.00 ± 5.11 % (n = 13 pups). AG490 pre-

treatment (30 mg/kg) significantly reduced the infarct volume to 22.76 ± 3.11 % (n = 17 pups) in

comparison to the vehicle-treated group (*, p < 0.05). There was no detectable infarction in the

sham group (data not shown). There was no detectable protective effect at 15 mg/kg AG490 pre-

treatment (Figure 12, infarct volume in the Vehicle group was 50.79 ± 6.53 %, n=8; infarct volume

in the 15 mg/kg AG490 treated group was 53.40 ± 9.03 %, n=11).

Page 55: Effects of TRPM2 Inhibition in Neuroprotection following

42

Figure 12. Lower dose of AG490 (15 mg/kg) did not provide neuroprotection. The dose of 15

mg/kg was chosen based on literature review129-132. With three repeated sets of experiments, there

was no observable neuroprotective effect. A. TTC result summary chart from the 1st experiment;

B. TTC result summary chart from 2nd experiment; C. TTC result summary chart from 3rd

experiment; D. Summary chart of all combined TTC results from 3 sets of experiments; E.

Representative brain slices for TTC staining. All data presented as mean ± SEM. Statistical

analysis was done by student’s t-test. There was no significant difference between vehicle group

and the AG490 group.

A B C

D E

Page 56: Effects of TRPM2 Inhibition in Neuroprotection following

43

Figure 13. Pre-treatment with TRPM2 currents inhibitor AG490 (30 mg/kg) reduced the

brain infarct volume of hypoxic-ischemic brain injury in vivo. A.B. Representative triphenyl

tetrazolium chloride (TTC) staining of brains treated with vehicle (5 % DMSO + 5 % Tween 80

in 0.9 % saline) and AG490 (30 mg/kg) 20 min before the onset of injury (pre-treatment). The

brains were harvested and stained 24 hrs following injury. Corrected infarct volume of vehicle-

treated (58.00 ± 5.109, n=13) and AG490-treated (22.76 ± 3.106, n=17) groups respectively 24 hrs

following injury. All data presented as mean ± SEM. Statistical analysis was done by student’s t-

test (*p<0.05).

1st Timeline TTC

Veh

icle

AG49

0

0

20

40

60

80

13 17

*

Infa

rcti

on

Vo

lum

e (

%)

A B

Page 57: Effects of TRPM2 Inhibition in Neuroprotection following

44

4.2 Pre-treatment with TRPM2 inhibitor AG490 reduced brain

damage following hypoxic-ischemic brain injury.

Next, I tested whether the neuroprotective action of AG490 was effective 7 days after HI brain

injury. Whole brains were collected, fixed, imaged, and then sectioned for Nissl staining 7 days

after HI (P14). On the 7th day after HI insult, the brain infarction had already undergone

liquefactive necrosis resulting in loss of brain weight (Figure 14A). Whole brain weight was used

as an indicator of the liquefaction level and measured in all groups. The sham and AG490-treated

groups had greater brain weights in comparison to the vehicle-treated group (Figure 14C, sham

group 0.43 ± 0.01 g, vehicle-treated + HI group 0.33 ± 0.01 g and AG490-treated + HI group 0.37

± 0.01 g). Consistent with this data, the AG490 pre-treatment (30 mg/kg) group demonstrated

significantly less brain damage (both in whole brains and coronal sections, n = 15 pups, Figure

14B) in comparison to vehicle treatment group (n = 21 pups). Whole brains were coronally sliced

into ~100 μm sections for Nissl (cresyl violet) staining to reveal that the AG490 treated group

sustained less brain damage in comparison to the vehicle treated group. There was no detectable

brain damage in sham group. With respect to the neuroprotective effects of AG490, whole brain

imaging at 7 days further verified the TTC staining results at 24 hrs after HI.

Page 58: Effects of TRPM2 Inhibition in Neuroprotection following

45

Figure 14. TRPM2 inhibitor AG490 reduced brain damage following hypoxic-ischemic brain

injury. A. Overall brain morphology was preserved in the pre-treatment paradigm 7 days after HI

injury. Nissl staining showed reduced liquefaction volume in the AG490-treated group in

comparison to the vehicle-treated group during the pre-treatment paradigm. B. Ipsilateral

liquefaction volume was significantly reduced in the AG490-treated group in comparison to the

vehicle group during the pre-treatment paradigm (sham: 0, n=16; vehicle: 63.72 ± 2.938 %, n=21;

AG490: 31.15 ± 4.410 %, n=15). C. Brain weight was significantly higher in the AG490-treated

group in comparison to the vehicle-treated group in the pre-treatment paradigm (vehicle: 0.33 ±

0.006 g, n=21; AG490: 0.38 ± 0.01g, n=15). All data presented as mean ± SEM. Statistical analysis

was done by one-way ANOVA followed by the Bonferroni post-hoc (*p<0.05). * comparison of

vehicle versus sham group; # comparison of AG490 versus vehicle group.

Page 59: Effects of TRPM2 Inhibition in Neuroprotection following

46

4.3 Pre-treatment with TRPM2 inhibitor AG490 promotes recovery

after HI challenge

Body weight is one of the most frequently used indicators of the general health of a mouse pup77,

133. Pups were randomly assigned to different experimental groups with no significant difference

in body weights between groups (sham group 5.05 ± 0.08 g, vehicle-treated + HI group 5.08 ±

0.06 g and AG490-treated + HI group 4.93 ± 0.09 g). The body weights of each group were

measured at 4 timelines: prior to the onset of HI as well as 1, 3, and 7 days after HI (Figure 15).

On day 1 after HI, the mean body weight of the pups that underwent HI surgery was significantly

reduced in comparison to the sham group. 7 days after HI, mice in sham (9.98 ± 0.49 g) and

AG490-treated groups (9.81 ± 0.50 g) gained significantly more weight than vehicle-treated HI

mice (8.32 ± 0.37 g, p < 0.05). These results indicated that AG490 treatment promoted general

health recovery after the HI procedure.

Figure 15. AG490 pre-treatment (20 min before HI injury) improves general health recovery

after HI challenge. Body weight, as an indicator of recovery after HI, was found to be

significantly higher in sham and AG490-treated groups than in vehicle-treated group on the 1st day

and 7 days after HI. All data presented as mean ± SEM. Statistical analysis: one-way ANOVA

followed by Boferroni post-hoc, *p<0.05. * comparison of vehicle versus sham group; #

comparison AG490 versus vehicle group.

Page 60: Effects of TRPM2 Inhibition in Neuroprotection following

47

4.4 Pre-treatment with TRPM2 inhibitor AG490 improves short-

term neurobehavioral performance after HI

To further examine the neuroprotective effect generated by AG490 pre-treatment, I further

assessed the functional outcomes in sham animals (n = 16 pups), vehicle-treated animals (n = 21

pups) and AG490 pre-treated animals (n = 15 pups). Short-term neurobehavioral tests evaluating

the geotaxis reflex, cliff avoidance and grip tests were performed on 1, 3, and 7 days after HI in

the 3 groups respectively. In comparison to pups in the sham group, the neurobehavioral

functioning of pups in the vehicle-treated HI group was significantly impaired 1, 3 and 7 days after

HI (Figure 16, *, p < 0.05). The AG490-treated group performed significantly better in the geotaxis

test 7 days after HI (Figure 16A) compared to the vehicle-treated group (2.33 ± 0.25 s in AG490-

treated HI group versus 8.74 ± 1.63 s in the vehicle-treated HI group; p < 0.05). The cliff avoidance

test performance was also significantly better in the AG490-treated group 3 and 7 days after HI

(Figure 16B) compared to the vehicle-treated group (2.58 ± 0.22 s in the AG490-treated group

versus 4.36 ± 1.33 s in the vehicle-treated group 3 days after HI; 3.86 ± 0.85 s in the AG490-

treated group versus 6.94 ± 1.15 s in the vehicle-treated group 7 days after HI; p < 0.05). Grip test

performance was also significantly better 1, 3 and 7 days after HI in the AG490-treated group

compared to the vehicle-treated group (Figure 16C) (p < 0.05). Therefore, AG490 treatment not

only reduced brain damage but also improved neurobehavioral outcomes after HI brain injury.

Page 61: Effects of TRPM2 Inhibition in Neuroprotection following

48

Figure 16. AG490 pre-treatment (20 min before HI injury) improved neurobehavioral

performance after HI challenge. A.B.C. Neurobehavioral evaluation was performed as described

in the methods section. Geotaxis reflex (A), cliff avoidance test (B) and grip test (C) of sham (n =

16), vehicle (HI + vehicle, n = 21) and AG490 (30 mg/kg) pretreatment (HI + AG490, n = 15)

groups were measured 1 day, 3 days and 7 days after HI (*, p < 0.05 versus sham group; #, p <

0.05 versus vehicle group). All data presented as mean ± SEM. Statistical analysis: one-way

ANOVA followed by the Boferroni post-hoc, *p<0.05.

Page 62: Effects of TRPM2 Inhibition in Neuroprotection following

49

4.5 Pre-treatment with TRPM2 inhibitor AG490 also improves long-

term neurobehavioral performance after HI

To determine whether AG490 pre-treatment would also improve long-term functional recovery, I

subjected mice to the passive avoidance test 3 weeks after HI brain injury and assessed whether

AG490 treatment attenuated memory impairment after HI.

I found that AG490-treated mice groups showed significantly better memory function in

comparison to the vehicle-treated mice, via evidence of significantly longer latency to enter the

dark room 24 hrs after foot shock (Figure 17A). Whole brains extracted 30 days after HI procedure

were used for assessing morphology changes Nissl staining was carried out to indicate the

histology changes. In Figure 17B left panel, representative images of whole brains and Nissl

staining all shown reduced liquefaction volume in AG490-treated group compared to vehicle-

treated group. Summary bar chart in Figure 17B right panel shown that AG490-treated group has

significant less ipsilateral liquefaction comparing to vehicle-treated group (sham: 0, n=11; vehicle:

49.65 ± 10.24 %, n=8; AG490: 24.22 ± 6.039 %, n=10).

By this, my data show that despite short term motor functional recovery improvement, AG490

treatment helps alleviate long term neurobehavioural deficits such as memory deficits following

neonatal HI brain injury in pre-treatment paradigm.

Page 63: Effects of TRPM2 Inhibition in Neuroprotection following

50

Figure 17. Long – term behavioral assessment of functional recovery in the pre-treatment

paradigm following hypoxic-ischemic injury. A. In the Passive Avoidance test, AG490-treated

mice showed better memory function compared to vehicle-treated mice, as was evident from

significantly longer latency to enter the dark room 24 hrs after the conditioning (foot shock). B.

Representative overall brain morphology image and Nissl staining images all shown reduced

liquefaction volume in AG490-treated group compared to vehicle-treated group. Summary bar

chart comparing AG490-treated group and vehicle-treated group (sham: 0, n=11; vehicle: 49.65 ±

10.24 %, n=8; AG490: 24.22 ± 6.039 %, n=10). All data presented as mean ± SEM. Statistical

analysis: one-way ANOVA with Boferroni post-hoc, *p<0.05.

P a s s iv e A v o id a n c e T e s t

0

5 0

1 0 0

1 5 0

2 0 0

2 5 0 *

La

ten

cy

to

En

ter (

s)

S h a m V e h ic le A G 4 9 0

a fte r fo o t s h o c k

b e fo re fo o t s h o c k

*

16 8 1 016 8 1 0

A

B

Page 64: Effects of TRPM2 Inhibition in Neuroprotection following

51

4.6 Pre-treatment with TRPM2 inhibitor AG490 reduces reactive

astrocyte activation

To assess the effects of AG490 on apoptotic signaling and neuronal survival, I performed

immunohistochemical staining and analysis of the penumbra area of the brain slices in sham,

vehicle- and AG490-treated groups 7 days after HI injury. Representative confocal images are

shown in Figure 18A. NeuN is the neuronal nuclear antigen, which is commonly used as a

biomarker for neurons77, 134. GFAP expression in astrocytes represent astroglial activation and

reactive gliosis77, 134, which are hallmarks following neurodegenerative conditions. As seen in

Figure 18B, AG490 pre-treatment significantly reduced the loss of NeuN-positive cells when

compared to the vehicle-treated group. I also found an upregulation of GFAP expression in

astrocytes, which indicates reactive gliosis and astroglial activation during neurodegeneration in

the vehicle-treated group. The expression of GFAP was significantly less in the AG490-treated

group in comparison to the vehicle group.

Page 65: Effects of TRPM2 Inhibition in Neuroprotection following

52

Figure 18. AG490 pretreatment restores neuronal cell numbers and reduces reactive

astrocyte activation. A. Representative confocal images of immunohistochemical staining. NeuN

stains for neurons, GFAP stains for reactive astrocytes. B. Summary bar charts of NeuN- and

GFAP-positive cells per 40x field.

N e u N -p o s it iv e c e lls p e r 4 0 x f ie ld

Sh

am

Veh

icle

AG

490

0

5 0

1 0 0

1 5 0

2 0 0

3 3 3

*

#

Ne

uN

- p

os

itiv

e c

ell

s

G F A P -p o s it iv e c e lls p e r 4 0 x f ie ld

Sh

am

Veh

icle

AG

490

0

5 0

1 0 0

1 5 0

2 0 0

2 5 0*

#

3 3 3

GF

AP

- p

os

itiv

e c

ell

s

A B NeuN GFAP

Page 66: Effects of TRPM2 Inhibition in Neuroprotection following

53

4.7 Pre-treatment with TRPM2 inhibitor AG490 may reduce HI

brain damage through Akt mediated signaling pathways

To determine the potential underlying signalling cascades affected by AG490 treatment, I

examined several signalling cascades that are known to be affected during neonatal HI injury.

During development, the p-Akt/t-Akt level is high at P7 in mice135. My data found that HI injury

significantly reduced Akt phosphorylation levels in the ipsilateral hemisphere. However, AG490

pre-treatment restored the normal levels of these proteins, indicating the protective effect of this

inhibitor.

Figure 19. Biochemical assessment of signalling pathways affected by hypoxic-ischemic

insult on the neonatal brain in a pre-treatment paradigm. A. Akt activation was reduced in

the ipsilateral hemispheres of the vehicle-treated group in comparison to sham group; AG490

pre-treatment restored Akt signalling (vehicle: 0.33 ± 0.09, n=3; AG490: 0.99 ± 0.02, n=3; data

normalized to sham). B. Representative western blots of proteins extracted from the ipsilateral

hemispheres of sham, vehicle-treated and AG490 pre-treated groups 24 hrs after HI. All data

presented as mean ± SEM. Statistical analysis: one-way ANOVA followed by the Boferroni

post-hoc, *p<0.05.

p-A

kt/G

AP

DH

(%

to

sh

am

)

sham

vehic

le

AG

490

0 .0

0 .5

1 .0

1 .5

*

3 3 3

t-A

kt/G

AP

DH

(%

to

sh

am

)

sham

vehic

le

AG

490

0 .0

0 .5

1 .0

1 .5

3 3 3

p-A

kt/t

-A

kt

(%

to

sh

am

)

sham

vehic

le

AG

490

0 .0

0 .5

1 .0

1 .5

3 3 3

*

p-Akt

t-Akt

GAPDH

Sham Vehicle AG490

A

B

Page 67: Effects of TRPM2 Inhibition in Neuroprotection following

54

5 The Effect of AG490 Post-treatment on Hypoxic-Ischemic Brain

Injury in vivo.

5.1 AG490 *post-treatment 1 (30 mg/kg, i.p.) reduced brain infarct

volume of hypoxic-ischemic brain injury in vivo.

*post-treatment 1: immediately after ischemia induction

Next, instead of injecting AG490 20 min prior to the onset of HI brain injury, I administrated

AG490 immediately after ischemia induction in order to test whether AG490 can also provide a

neuroprotective effect in a post-treatment paradigm. By using the same methodology as during

pre-treatment, TTC staining following the post-treatment paradigm showed that AG490

administration right after ischemia induction also reduced brain damage in the neonatal hypoxic-

ischemic brain injury model (Figure 20). AG490 post-treatment (30 mg/kg, i.p., immediately after

ischemia induction) significantly reduced the brain infarct volume to 28.84 ± 3.609 (n=10) in

comparison to the vehicle treated group (50.51 ± 5.100, n=12, *, p < 0.05).

5.2 AG490 *post-treatment 1 (30 mg/kg, i.p.) reduced brain

damage following hypoxic-ischemic brain injury

*post-treatment 1: immediately after ischemia induction

Using the same methodology as during pretreatment, whole brains for post-treatment 1 were

collected, fixed and imaged 7 days after HI with the whole brain weights being measured. The

AG490 post-treatment (30 mg/kg) group demonstrated significantly less brain damage in

comparison to the vehicle treatment group (Figure 21, n = 21 pups). The sham group and AG490-

treated groups had greater brain weights in comparison to the vehicle-treated group (sham: 0, n=5;

vehicle: 57.81 ± 2.434 %, n=19; AG490: 34.24 ± 3.914 %, n=11). Therefore, I conclude that post-

treatment of AG490 also reduced brain damage following HI brain injury.

Page 68: Effects of TRPM2 Inhibition in Neuroprotection following

55

Figure 20. Post-treatment 1 (30 mg/kg, i.p.) reduced brain infarct volume and brain damage

following hypoxic-ischemic brain injury in vivo. Upper panel shows the timeline for TTC

staining and whole brain imaging in AG490 post-treatment. Lower panel A and B show

representative images for TTC staining and a summary chart for corrected brain infarction volume

following AG490 post-treatment. All data presented as mean ± SEM. Statistical analysis was done

by student’s t-test (*p<0.05).

A B

Page 69: Effects of TRPM2 Inhibition in Neuroprotection following

56

Figure 21. TRPM2 inhibitor AG490 reduced morphological and histological damage

following hypoxic-ischemic brain injury. A. Representative images for whole brain and Nissl

staining. B. The ipsilateral liquefaction volume was significantly reduced in the AG490-treated

group compared to vehicle group in post-treatment paradigm (sham: 0, n=16; vehicle: 57.81 ±

2.434 %, n=17; AG490: 34.24 ± 3.914 %, n=11). B. Brain weight was significantly higher in the

AG490-treated group compared to the vehicle-treated group in post-treatment paradigm (sham:

0.15 ± 0.006 g, n=16; vehicle: 0.35 ± 0.014 g, n=17; AG490: 0.40 ± 0.007 g, n=11). All data

presented as mean ± SEM. Statistical analysis was done by one-way ANOVA following by the

Bonferroni post-hoc (*p<0.05). * comparison of vehicle versus sham group; # comparison of

AG490 versus vehicle group.

B

Sh

am

Veh

icle

AG

490

0 .0

0 .1

0 .2

0 .3

0 .4

0 .5

1 6 1 7 1 1

*

#

bra

in w

eig

ht

(g)

A C

he

mis

ph

ere

liq

ue

fac

tio

n v

olu

me

(%

)

Sh

am

Veh

icle

AG

490

0

2 0

4 0

6 0

8 0

1 6 1 7 1 1

*

#

Page 70: Effects of TRPM2 Inhibition in Neuroprotection following

57

5.3 AG490 *post-treatment 1 (30 mg/kg, i.p.) improves

neurobehavioral performance and general recovery after HI

*post-treatment 1: immediately after ischemia induction

Body weight in each group was measured and used as indicators for the general health of mice.

Similar to pre-treatment, pups were randomly assigned to different experimental groups with no

significant difference in body weight between groups (sham group 5.05 ± 0.08 g, vehicle-treated

+ HI group 5.08 ± 0.06 g and AG490-treated + HI group 4.93 ± 0.09 g). The body weights of each

group were measured at 4 timelines: prior to the onset of HI as well as 1, 3, and 7 days after HI

(Figure 22A). On day 3 and day 7 after HI, the mean body weight of pups in the vehicle treated

group was significantly lower than those in the sham and AG490 treated groups. Therefore, AG490

post-treatment 1 also promoted body weight. Consistent with these results, AG490 post-treatment

1 also demonstrated improvement in short-term neuronal behavioral tests (see Figure 22B, C, D).

These results indicated that AG490 can also improve general health recovery as well as neuronal

behavioral outcomes following HI injury using a post-treatment paradigm.

Page 71: Effects of TRPM2 Inhibition in Neuroprotection following

58

Figure 22. AG490 post-treatment 1 (immediately after ischemic injury) improves general

health and neurobehavioral performance after HI challenge. A. Body weights were measured

1, 3 and 7 days after HI. B.C. D. Neurobehavioral evaluation was performed as described in the

methods section. Geotaxis reflex (B), cliff avoidance test (C) and grip test (D) in the sham (n =

16), vehicle (HI + vehicle, n = 19) and AG490 (30 mg/kg) pretreatment (HI + AG490, n = 11)

groups were measured 1, 3 and 7 days after HI (*, p < 0.05 versus sham group; #, p < 0.05 versus

vehicle group). All data presented as mean ± SEM. Statistical analysis: one-way ANOVA followed

by the Boferroni post-hoc, *p<0.05.

C D

B A

Page 72: Effects of TRPM2 Inhibition in Neuroprotection following

59

6 AG490 *post-treatment 2 (30mg/kg, i.p., immediately after HI

induction) demonstrate a trend towards neuroprotection following

HI brain injury

*post-treatment 2: immediately after hypoxia ischemia induction

To further evaluate the therapeutic potential of AG490, I evaluated another time point of

administration. In this case, AG490 was administrated immediately after whole HI induction

(including ischemia and hypoxia processes). The same TTC method as before was used to evaluate

the brain infarct volume between groups.

According to my results, the administration of AG490 immediately after HI brain injury

demonstrated a trend towards neuroprotection, though this effect was not yet significant.

Figure 23. *Post-treatment 2 (30 mg/kg, i.p.) shows a trend towards neuroprotection

following hypoxic-ischemic brain injury. Upper panel shows the timeline for TTC staining for

post-treatment in AG490 post-treatment. Lower panel shows representative images for TTC

staining and a summary chart for corrected brain infarction volume following AG490 post-

treatment. All data presented as mean ± SEM. Statistical analysis was done by student’s t-test.

Page 73: Effects of TRPM2 Inhibition in Neuroprotection following

60

Discussion

1. Connection between clinics and the current study

Despite major advances in understanding fetal and neonatal pathologies, hypoxic-ischemic (HI)

brain injury remains a serious issue that causes significant mortality and long-term morbidity in

neonates. Due to lack of efficient treatment for neonatal HI brain injury, survivors and their

families suffer from the burden of lifetime health care costs. The current licensed and most efficient

treatment for neonatal HI brain injury is hypothermia14. However, clinical studies have shown that

this therapy is not effective for all infants affected by HI. Therefore, in considering the global

prevalence of this ailment and the poor long-term outcomes, novel neuroprotective treatments that

may be used in conjunction with hypothermia are urgently required in the treatment of this

disorder.

Traditional glutamate driven mechanisms were long thought to be the major pathways involved in

the ischemic cascade. Hence, glutamate receptors including NMDA and AMPA receptors were

considered as promising therapeutic targets22. However, all clinical trials using compounds

targeting glutamate receptors failed to generate the expected neuroprotective outcomes22. TRPM2

channels, among the major proteins whose activities were investigated under non-glutamate driven

mechanisms, have been since implicated to be involved in numerous physiological and

pathological processes including HI brain injury. The absence of TRPM2 channel activity by either

using siRNA to silence TRPM2 in vitro 79or knocking out TRPM2 channels in vivo83, 115 was

observed to provide neuroprotective effects. Therefore, TRPM2 channels are important therapeutic

targets for further study. In my study, I used a newly identified TRPM2 channel current inhibitor

Page 74: Effects of TRPM2 Inhibition in Neuroprotection following

61

AG490 to verify the role of this structure in neonatal HI brain injury as well as tested the potential

clinical use of this compound.

2. Summary of major findings

Here, I evaluated a novel inhibitor of the TRPM2 channel (AG490) during treatment in vitro

following H2O2-induced neuronal cell death as well as in vivo in a mouse model of neonatal

hypoxic-ischemic brain injury. AG490 was initially studied as a JAK2 inhibitor. AG490 blocks

TRPM2 activity by acting as a hydroxyl radical scavenger, indicating that AG490 significantly

reduces H2O2-induced TRPM2 activation through scavenging hydroxyl radicals rather than Jak2-

dependent mechanisms. By using a H2O2-induced neuronal cell death model, I found that AG490

protected neurons from apoptosis. I then showed the strong preventative and therapeutic potential

of AG490 in our mouse model of neonatal HI brain injury. I found that pre-treatment with AG490

immediately after hypoxic ischemic exposure significantly reduced brain infarction volumes

compared to vehicle controls 24 hrs after HI. I also found that AG490 administration reduced brain

mass loss and preserved overall brain morphology up to 7 days after HI injury in comparison to

the vehicle group. General health, short-term and long-term functional recovery was significantly

improved with AG490 pretreatment 20 min prior to the onset of HI brain injury as well as

following AG490 posttreatment immediately after ischemic induction. The underlying

mechanisms of this neuroprotective effect requires further evaluation. My data suggests that HI

injury significantly reduced Akt phosphorylation levels in the ipsilateral hemisphere, and that

AG490 pre-treatment restored this deficit. Therefore, the neuroprotective effect of AG490 may be

related to the Akt cascade. Taken together, my study conclusively demonstrates that AG490 has

neuroprotective and therapeutic properties in a mouse model of neonatal HIE.

Page 75: Effects of TRPM2 Inhibition in Neuroprotection following

62

3. Significance of the current study

My study has revealed that the inhibition of TRPM2 channel activity provides neuroprotection

against neonatal hypoxic-ischemic brain injury, and that TRPM2 channel inhibitors may be

potential pharmacological treatments for HI brain injury. This knowledge may help in providing

improved healthcare options for hypoxic-ischemic brain injury in children. It may also divulge

potential preventive measures for HI related neurological complications such as hypoxic-ischemic

encephalopathy and cerebral palsy. The principle findings may also be applied to the study of other

related neurological disorders as well.

4. Differences between neonatal HI brain injury and adult stroke

Neonatal HI brain injury has important differences in comparison to adult ischemic stroke5. For

example, liquifactive disintegration can be the result of severe HI events in the infant brain, but is

not seen following adult ischemic stroke4. Additionally, newly formed blood vessels in neonates

are prone to rupture. The blood brain barrier (BBB) is also compromised following neonatal HI

brain injury4, 5. The autoregulation of the cerebrovasculature is another factor of concern in

infants136. Pre-term neonates demonstrate a “pressure passive” cerebral circulation while sick term

infants show impairment in autoregulation3, 4. Moreover, the concentrations and actions of various

signaling molecules including caspase-3 are different in the developing brain4.

Neonatal HI injury may also evolve over time while adult ischemic stroke does not15. By using

MRI scanning system, it has been found that injuries within the first few hours following HI were

subtle and restrictively diffused only in putamen and thalami, then the injuries progress over within

the next 3-4 days and diffused to other areas in the brain15.

Page 76: Effects of TRPM2 Inhibition in Neuroprotection following

63

To add to this complexity, the NMDA receptor is relatively over-expressed in the developing

brain140, 141. In P6 rats, the NMDA receptor is expressed at 150–200 % of the adult levels142. The

combination of NMDA receptor subunits in the perinatal period are thought to be favored of

prolonged calcium influx for a given excitation143. Increased level of glutamate has been found in

the cerebrospinal fluid (CSF) of infants who have suffered from severe HI injury137, 144. The

neonatal brain is more sensitive to seizure activity compared to the mature brain, suggesting a

prominent role for neuronal hyperexcitability and excitotoxicity145, 146. Neonatal brains are also

more vulnerable to hydrogen peroxide elevation, which is one of the characteristics following HI

brain injury121. The accumulation of hydrogen peroxide coupled with low antioxidant activity in

neonates results in sensitivity to oxidative stress induced by HI.

5. Proposed mechanism of neonatal HI brain injury

GSK-3 was first identified as a regulatory protein for glycogen metabolism. It has two isoforms

named GSK-3α and GSK-3β respectively147. GSK-3 is involved in numerous cellular processes in

the brain, including the Wnt-1/β–catenin and phosphoinositide 3-kinase/protein kinase

B(PI3K/Akt) signaling pathways147, 148. The kinase activity of GSK-3 may be inhibited through

phosphorylation of GSK-3α (pGSK-3α) at the Ser21 site and GSK-3β (p-GSK-3β) at the Ser9

site147, 149.

Studies have shown that phosphorylation of Akt could inhibit GSK-3β kinase activity and

consequently result in the downregulation of caspase-3 mediated apoptotic signaling135. This

suggests that the regulation of GSK-3β kinase activity may be linked to cell death as observed in

our model. Our previous study has shown that using TDZD-8135, which is a specific blocker of

Page 77: Effects of TRPM2 Inhibition in Neuroprotection following

64

Figure 24. Propose mechanism for inhibitory effect of AG490. The mechanism of inhibitory

effect of AG490 needs further evaluation. It may inhibit TRPM2 activities through i) direct block

of TRPM2 channels; ii) inhibit TRPM2 activity through ADPR-independent pathways and reduce

the level of TRPM2 activator H2O2; iii) inhibit TRPM2 activity through ADPR-dependent

pathways and reduce the level of H2O2 first then block the ADPR binding site. The inhibitory

effect may get involved in p-Akt – GSK-3β – caspase 3 signaling pathways.

Page 78: Effects of TRPM2 Inhibition in Neuroprotection following

65

GSK-3β, can also generate a neuroprotective effect. Therefore, AG490’s neuroprotective and

therapeutic effects in our mouse model may act through the Akt/GSK-3β/Caspase-3 pathway.

Further experimentation needs to be carried out in order to verify the link between AG490 to GSK-

3β and Caspase-3.

6. Pitfalls in the current study and proposed future directions

My study tested the inhibitory effect of AG490 on TRPM2 channel activity and found that this

drug demonstrated a promising neuroprotective effect both in vitro and in vivo. The in vivo

neuroprotective effect of AG490 could be observed at different time points. In order to limits gaps

between the mouse model and a future clinical study, the following improvements may be made

to the current study:

i) Instead of injecting AG490 only once, the drug could be administrated multiple times.

Multiple administrations of the compound may result in stronger and even more

prolonged neuroprotective effects. As shown in my TTC data, the neuroprotective

effect of AG490 was strong within the 1st and 2nd timelines. However, the data showed

a trend of decreasing neuroprotective effect according to the time period of AG490

administration.

ii) To better evaluate the effects of AG490, different doses of the compound could be

tested on our HI mouse model. The lower dose of AG490 (15 mg/kg) initially utilized

in the study was chosen based on literature review. As we did not see a difference

between the vehicle treated and AG490 treated groups, we increased the dose to 30

Page 79: Effects of TRPM2 Inhibition in Neuroprotection following

66

mg/kg to observe a neuroprotective effect. To further test the effects of AG490, 2 more

doses (1 dose lower than 30 mg/kg but higher than 15mg/kg; the other dose higher than

30 mg/kg) could be chosen for additional experiments in our HI mouse model. By this,

we could verify whether AG490 generates a neuroprotective effect in a dose-dependent

manner. Additionally, higher doses of AG490 may lead to prolonged neuroprotective

effects.

iii) AG490 was initially studied as a JAK2 inhibitor, and is therefore not a specific inhibitor

for TRPM2 channels. An in vitro study indicated that AG490 may inhibit H2O2-induced

TRPM2 activation in a JAK2-independent manner. In this case, Mori and colleagues

used the whole cell patch clamp technique to test the inhibitory effect of AG490 on

TRPM2 HEK293 cells. Interestingly, AG490 efficiently blocked H2O2-induced

TRPM2 channel currents while Jak inhibitor 1 and staurosporine, which are commonly

used as JAK2 inhibitors (Lawrie et al., 1997; Nakagawa et al., 2011), did not affect

TRPM2 channel activity at all. These observations suggest that inhibition of H2O2-

induced TRPM2 channel activation by AG490 is independent of JAK2 mechanisms.

As a future direction for my study, the effects of JAK2 specific inhibitors like Jak2

inhibitor 1 and staurosporine could be tested in vivo. If this in vivo data is not consistent

with or even opposite to the effects observed under AG490, we could then verify that

the inhibitory effect of AG490 on TRPM2 channel activity in vivo is also independent

from JAK2 mechanisms. If the in vivo data generated using other JAK2 specific

inhibitors shows a similar effect as AG490, then more experiments need to be carried

out to further evaluate the action of AG490 in either JAK2 dependent or independent

Page 80: Effects of TRPM2 Inhibition in Neuroprotection following

67

mechanisms. For example, the combined administration of AG490 and a JAK2 specific

activator/inhibitor into the HI mouse model could be one possible experiment.

iv) Since AG490 acts as a hydroxyl radical scavenger, it is important to test whether the

neuroprotective effect generated by this compound is specifically mediated via

inhibition of TRPM2 channel activity. Due to the availability of TRPM2 knockout

mice, a proposed next step could be the administration of AG490 into this model to

view changes in neuroprotective effects in comparison to the controls.

v) Another future direction could include the observance of additional long-term

behavioral tests to fully assess the functional recovery outcomes of TRPM2 inhibition

by AG490. For instance, a novel objective test could be conducted150.

Page 81: Effects of TRPM2 Inhibition in Neuroprotection following

68

REFERENCES

1. Perlman, J.M. Intrapartum hypoxic-ischemic cerebral injury and subsequent cerebral

palsy: medicolegal issues. Pediatrics 99, 851-859 (1997).

2. Perlman, J.M. & Risser, R. Severe fetal acidemia: neonatal neurologic features and short-

term outcome. Pediatric neurology 9, 277-282 (1993).

3. Shah, P.S., Beyene, J., To, T., Ohlsson, A. & Perlman, M. Postasphyxial hypoxic-

ischemic encephalopathy in neonates: outcome prediction rule within 4 hours of birth. Archives

of pediatrics & adolescent medicine 160, 729-736 (2006).

4. Millar, L.J., Shi, L., Hoerder-Suabedissen, A. & Molnar, Z. Neonatal Hypoxia Ischaemia:

Mechanisms, Models, and Therapeutic Challenges. Frontiers in cellular neuroscience 11, 78

(2017).

5. Ferriero, D.M. Neonatal brain injury. The New England journal of medicine 351, 1985-

1995 (2004).

6. Lawn, J.E., Cousens, S., Zupan, J. & Lancet Neonatal Survival Steering, T. 4 million

neonatal deaths: when? Where? Why? Lancet 365, 891-900 (2005).

7. Finer, N.N., Robertson, C.M., Richards, R.T., Pinnell, L.E. & Peters, K.L. Hypoxic-

ischemic encephalopathy in term neonates: perinatal factors and outcome. The Journal of

pediatrics 98, 112-117 (1981).

8. Richer, L.P., Shevell, M.I. & Miller, S.P. Diagnostic profile of neonatal hypotonia: an 11-

year study. Pediatric neurology 25, 32-37 (2001).

9. Levene, M.I., Sands, C., Grindulis, H. & Moore, J.R. Comparison of two methods of

predicting outcome in perinatal asphyxia. Lancet 1, 67-69 (1986).

10. Ruth, V.J. & Raivio, K.O. Perinatal brain damage: predictive value of metabolic acidosis

and the Apgar score. Bmj 297, 24-27 (1988).

11. Laptook, A.R., et al. Outcome of term infants using apgar scores at 10 minutes following

hypoxic-ischemic encephalopathy. Pediatrics 124, 1619-1626 (2009).

12. Robertson, N.J., Cowan, F.M., Cox, I.J. & Edwards, A.D. Brain alkaline intracellular pH

after neonatal encephalopathy. Annals of neurology 52, 732-742 (2002).

13. Weeke, L.C., et al. Role of EEG background activity, seizure burden and MRI in

predicting neurodevelopmental outcome in full-term infants with hypoxic-ischaemic

encephalopathy in the era of therapeutic hypothermia. European journal of paediatric neurology

: EJPN : official journal of the European Paediatric Neurology Society 20, 855-864 (2016).

14. Choi, H.A., Badjatia, N. & Mayer, S.A. Hypothermia for acute brain injury--mechanisms

and practical aspects. Nature reviews. Neurology 8, 214-222 (2012).

15. Fatemi, A., Wilson, M.A. & Johnston, M.V. Hypoxic-ischemic encephalopathy in the

term infant. Clinics in perinatology 36, 835-858, vii (2009).

16. Gunn, A.J., Battin, M., Gluckman, P.D., Gunn, T.R. & Bennet, L. Therapeutic

hypothermia: from lab to NICU. Journal of perinatal medicine 33, 340-346 (2005).

17. Gunn, A.J. & Gunn, T.R. The 'pharmacology' of neuronal rescue with cerebral

hypothermia. Early human development 53, 19-35 (1998).

18. Kracer, B., Hintz, S.R., Van Meurs, K.P. & Lee, H.C. Hypothermia therapy for neonatal

hypoxic ischemic encephalopathy in the state of California. The Journal of pediatrics 165, 267-

273 (2014).

Page 82: Effects of TRPM2 Inhibition in Neuroprotection following

69

19. Pauliah, S.S., Shankaran, S., Wade, A., Cady, E.B. & Thayyil, S. Therapeutic

hypothermia for neonatal encephalopathy in low- and middle-income countries: a systematic

review and meta-analysis. PloS one 8, e58834 (2013).

20. Savman, K. & Brown, K.L. Treating neonatal brain injury - promise and inherent

research challenges. Recent patents on inflammation & allergy drug discovery 4, 16-24 (2010).

21. Arundine, M. & Tymianski, M. Molecular mechanisms of glutamate-dependent

neurodegeneration in ischemia and traumatic brain injury. Cellular and molecular life sciences :

CMLS 61, 657-668 (2004).

22. Besancon, E., Guo, S., Lok, J., Tymianski, M. & Lo, E.H. Beyond NMDA and AMPA

glutamate receptors: emerging mechanisms for ionic imbalance and cell death in stroke. Trends

in pharmacological sciences 29, 268-275 (2008).

23. Anderson, C.M. & Swanson, R.A. Astrocyte glutamate transport: review of properties,

regulation, and physiological functions. Glia 32, 1-14 (2000).

24. Chinopoulos, C., Gerencser, A.A., Doczi, J., Fiskum, G. & Adam-Vizi, V. Inhibition of

glutamate-induced delayed calcium deregulation by 2-APB and La3+ in cultured cortical

neurones. Journal of neurochemistry 91, 471-483 (2004).

25. Hetman, M. & Kharebava, G. Survival signaling pathways activated by NMDA

receptors. Current topics in medicinal chemistry 6, 787-799 (2006).

26. Liu, Y., et al. NMDA receptor subunits have differential roles in mediating excitotoxic

neuronal death both in vitro and in vivo. The Journal of neuroscience : the official journal of the

Society for Neuroscience 27, 2846-2857 (2007).

27. Gladstone, D.J., Black, S.E., Hakim, A.M., Heart & Stroke Foundation of Ontario Centre

of Excellence in Stroke, R. Toward wisdom from failure: lessons from neuroprotective stroke

trials and new therapeutic directions. Stroke; a journal of cerebral circulation 33, 2123-2136

(2002).

28. Ikonomidou, C. & Turski, L. Why did NMDA receptor antagonists fail clinical trials for

stroke and traumatic brain injury? The Lancet. Neurology 1, 383-386 (2002).

29. Krebs, C., Fernandes, H.B., Sheldon, C., Raymond, L.A. & Baimbridge, K.G. Functional

NMDA receptor subtype 2B is expressed in astrocytes after ischemia in vivo and anoxia in vitro.

The Journal of neuroscience : the official journal of the Society for Neuroscience 23, 3364-3372

(2003).

30. Bonfoco, E., Krainc, D., Ankarcrona, M., Nicotera, P. & Lipton, S.A. Apoptosis and

necrosis: two distinct events induced, respectively, by mild and intense insults with N-methyl-D-

aspartate or nitric oxide/superoxide in cortical cell cultures. Proceedings of the National

Academy of Sciences of the United States of America 92, 7162-7166 (1995).

31. Matute, C., Alberdi, E., Ibarretxe, G. & Sanchez-Gomez, M.V. Excitotoxicity in glial

cells. European journal of pharmacology 447, 239-246 (2002).

32. Wong, R. NMDA receptors expressed in oligodendrocytes. BioEssays : news and reviews

in molecular, cellular and developmental biology 28, 460-464 (2006).

33. Karadottir, R., Cavelier, P., Bergersen, L.H. & Attwell, D. NMDA receptors are

expressed in oligodendrocytes and activated in ischaemia. Nature 438, 1162-1166 (2005).

34. Newcomer, J.W., Farber, N.B. & Olney, J.W. NMDA receptor function, memory, and

brain aging. Dialogues in clinical neuroscience 2, 219-232 (2000).

35. Coyle, J.T., Basu, A., Benneyworth, M., Balu, D. & Konopaske, G. Glutamatergic

synaptic dysregulation in schizophrenia: therapeutic implications. Handbook of experimental

pharmacology, 267-295 (2012).

36. Bargiotas, P., Monyer, H. & Schwaninger, M. Hemichannels in cerebral ischemia.

Current molecular medicine 9, 186-194 (2009).

Page 83: Effects of TRPM2 Inhibition in Neuroprotection following

70

37. Alibrahim, A., et al. Neuroprotective effects of volume-regulated anion channel blocker

DCPIB on neonatal hypoxic-ischemic injury. Acta pharmacologica Sinica 34, 113-118 (2013).

38. Blaustein, M.P. & Lederer, W.J. Sodium/calcium exchange: its physiological

implications. Physiological reviews 79, 763-854 (1999).

39. Bojarski, C., Meloni, B.P., Moore, S.R., Majda, B.T. & Knuckey, N.W. Na+/Ca2+

exchanger subtype (NCX1, NCX2, NCX3) protein expression in the rat hippocampus following

3 min and 8 min durations of global cerebral ischemia. Brain research 1189, 198-202 (2008).

40. Khananshvili, D. Sodium-calcium exchangers (NCX): molecular hallmarks underlying

the tissue-specific and systemic functions. Pflugers Archiv : European journal of physiology 466,

43-60 (2014).

41. Gomez-Villafuertes, R., Mellstrom, B. & Naranjo, J.R. Searching for a role of

NCX/NCKX exchangers in neurodegeneration. Molecular neurobiology 35, 195-202 (2007).

42. Meng, F., To, W.K. & Gu, Y. Role of TRP channels and NCX in mediating hypoxia-

induced [Ca(2+)](i) elevation in PC12 cells. Respiratory physiology & neurobiology 164, 386-

393 (2008).

43. Matsuda, T., et al. SEA0400, a novel and selective inhibitor of the Na+-Ca2+ exchanger,

attenuates reperfusion injury in the in vitro and in vivo cerebral ischemic models. The Journal of

pharmacology and experimental therapeutics 298, 249-256 (2001).

44. Kormos, A., et al. Efficacy of selective NCX inhibition by ORM-10103 during simulated

ischemia/reperfusion. European journal of pharmacology 740, 539-551 (2014).

45. Pignataro, G., et al. Evidence for a protective role played by the Na+/Ca2+ exchanger in

cerebral ischemia induced by middle cerebral artery occlusion in male rats. Neuropharmacology

46, 439-448 (2004).

46. Boscia, F., et al. Permanent focal brain ischemia induces isoform-dependent changes in

the pattern of Na+/Ca2+ exchanger gene expression in the ischemic core, periinfarct area, and

intact brain regions. Journal of cerebral blood flow and metabolism : official journal of the

International Society of Cerebral Blood Flow and Metabolism 26, 502-517 (2006).

47. Belousov, A.B., Fontes, J.D., Freitas-Andrade, M. & Naus, C.C. Gap junctions and

hemichannels: communicating cell death in neurodevelopment and disease. BMC cell biology 18,

4 (2017).

48. Thompson, R.J., Zhou, N. & MacVicar, B.A. Ischemia opens neuronal gap junction

hemichannels. Science 312, 924-927 (2006).

49. Johansen, D., Cruciani, V., Sundset, R., Ytrehus, K. & Mikalsen, S.O. Ischemia induces

closure of gap junctional channels and opening of hemichannels in heart-derived cells and tissue.

Cellular physiology and biochemistry : international journal of experimental cellular

physiology, biochemistry, and pharmacology 28, 103-114 (2011).

50. Contreras, J.E., et al. Role of connexin-based gap junction channels and hemichannels in

ischemia-induced cell death in nervous tissue. Brain research. Brain research reviews 47, 290-

303 (2004).

51. Davidson, J.O., et al. Connexin hemichannel blockade is neuroprotective after asphyxia

in preterm fetal sheep. PloS one 9, e96558 (2014).

52. Davidson, J.O., et al. Connexin hemichannel blockade improves outcomes in a model of

fetal ischemia. Annals of neurology 71, 121-132 (2012).

53. Kim, Y., et al. Tonabersat Prevents Inflammatory Damage in the Central Nervous System

by Blocking Connexin43 Hemichannels. Neurotherapeutics : the journal of the American Society

for Experimental NeuroTherapeutics (2017).

54. Xu, J., Chen, L. & Li, L. Pannexin hemichannels: A novel promising therapy target for

oxidative stress related diseases. Journal of cellular physiology (2017).

Page 84: Effects of TRPM2 Inhibition in Neuroprotection following

71

55. Nielsen, B.S., Hansen, D.B., Ransom, B.R., Nielsen, M.S. & MacAulay, N. Connexin

Hemichannels in Astrocytes: An Assessment of Controversies Regarding Their Functional

Characteristics. Neurochemical research (2017).

56. Best, L., Brown, P.D., Sener, A. & Malaisse, W.J. Electrical activity in pancreatic islet

cells: The VRAC hypothesis. Islets 2, 59-64 (2010).

57. Voss, F.K., et al. Identification of LRRC8 heteromers as an essential component of the

volume-regulated anion channel VRAC. Science 344, 634-638 (2014).

58. Pedersen, S.F., Okada, Y. & Nilius, B. Biophysics and Physiology of the Volume-

Regulated Anion Channel (VRAC)/Volume-Sensitive Outwardly Rectifying Anion Channel

(VSOR). Pflugers Archiv : European journal of physiology 468, 371-383 (2016).

59. Jentsch, T.J., Lutter, D., Planells-Cases, R., Ullrich, F. & Voss, F.K. VRAC: molecular

identification as LRRC8 heteromers with differential functions. Pflugers Archiv : European

journal of physiology 468, 385-393 (2016).

60. Bowens, N.H., Dohare, P., Kuo, Y.H. & Mongin, A.A. DCPIB, the proposed selective

blocker of volume-regulated anion channels, inhibits several glutamate transport pathways in

glial cells. Molecular pharmacology 83, 22-32 (2013).

61. Fujii, T., et al. Inhibition of gastric H+,K+-ATPase by 4-(2-butyl-6,7-dichloro-2-

cyclopentylindan-1-on-5-yl)oxybutyric acid (DCPIB), an inhibitor of volume-regulated anion

channel. European journal of pharmacology 765, 34-41 (2015).

62. Han, Q., et al. DCPIB, a potent volume-regulated anion channel antagonist, attenuates

microglia-mediated inflammatory response and neuronal injury following focal cerebral

ischemia. Brain research 1542, 176-185 (2014).

63. Minieri, L., et al. The inhibitor of volume-regulated anion channels DCPIB activates

TREK potassium channels in cultured astrocytes. British journal of pharmacology 168, 1240-

1254 (2013).

64. Trouet, D., et al. Inhibition of VRAC by c-Src tyrosine kinase targeted to caveolae is

mediated by the Src homology domains. American journal of physiology. Cell physiology 281,

C248-256 (2001).

65. Wang, J.J. & Xu, T.L. [Acid-sensing ion channels as a target for neuroprotection:

acidotoxicity revisited]. Sheng li xue bao : [Acta physiologica Sinica] 68, 403-413 (2016).

66. Yermolaieva, O., Leonard, A.S., Schnizler, M.K., Abboud, F.M. & Welsh, M.J.

Extracellular acidosis increases neuronal cell calcium by activating acid-sensing ion channel 1a.

Proceedings of the National Academy of Sciences of the United States of America 101, 6752-

6757 (2004).

67. Voilley, N. Acid-sensing ion channels (ASICs): new targets for the analgesic effects of

non-steroid anti-inflammatory drugs (NSAIDs). Current drug targets. Inflammation and allergy

3, 71-79 (2004).

68. Xiong, Z.G., et al. Neuroprotection in ischemia: blocking calcium-permeable acid-

sensing ion channels. Cell 118, 687-698 (2004).

69. Allen, N.J. & Attwell, D. Modulation of ASIC channels in rat cerebellar Purkinje neurons

by ischaemia-related signals. The Journal of physiology 543, 521-529 (2002).

70. Immke, D.C. & McCleskey, E.W. Lactate enhances the acid-sensing Na+ channel on

ischemia-sensing neurons. Nature neuroscience 4, 869-870 (2001).

71. Baron, A. & Lingueglia, E. Pharmacology of acid-sensing ion channels - Physiological

and therapeutical perspectives. Neuropharmacology 94, 19-35 (2015).

72. Li, H. TRP Channel Classification. Advances in experimental medicine and biology 976,

1-8 (2017).

Page 85: Effects of TRPM2 Inhibition in Neuroprotection following

72

73. Gees, M., Colsoul, B. & Nilius, B. The role of transient receptor potential cation channels

in Ca2+ signaling. Cold Spring Harbor perspectives in biology 2, a003962 (2010).

74. Morelli, M.B., Amantini, C., Liberati, S., Santoni, M. & Nabissi, M. TRP channels: new

potential therapeutic approaches in CNS neuropathies. CNS & neurological disorders drug

targets 12, 274-293 (2013).

75. Nilius, B., Owsianik, G., Voets, T. & Peters, J.A. Transient receptor potential cation

channels in disease. Physiological reviews 87, 165-217 (2007).

76. Aarts, M., et al. A key role for TRPM7 channels in anoxic neuronal death. Cell 115, 863-

877 (2003).

77. Chen, W., et al. TRPM7 inhibitor carvacrol protects brain from neonatal hypoxic-

ischemic injury. Molecular brain 8, 11 (2015).

78. Aarts, M.M. & Tymianski, M. TRPMs and neuronal cell death. Pflugers Archiv :

European journal of physiology 451, 243-249 (2005).

79. Kaneko, S., et al. A critical role of TRPM2 in neuronal cell death by hydrogen peroxide.

Journal of pharmacological sciences 101, 66-76 (2006).

80. Sun, H.S., et al. Suppression of hippocampal TRPM7 protein prevents delayed neuronal

death in brain ischemia. Nature neuroscience 12, 1300-1307 (2009).

81. Miller, B.A. & Cheung, J.Y. TRPM2 protects against tissue damage following oxidative

stress and ischemia-reperfusion. The Journal of physiology (2015).

82. Shimizu, T., et al. Extended therapeutic window of a novel peptide inhibitor of TRPM2

channels following focal cerebral ischemia. Experimental neurology 283, 151-156 (2016).

83. Alim, I., Teves, L., Li, R., Mori, Y. & Tymianski, M. Modulation of NMDAR subunit

expression by TRPM2 channels regulates neuronal vulnerability to ischemic cell death. The

Journal of neuroscience : the official journal of the Society for Neuroscience 33, 17264-17277

(2013).

84. Olah, M.E., et al. Ca2+-dependent induction of TRPM2 currents in hippocampal neurons.

The Journal of physiology 587, 965-979 (2009).

85. Ye, M., et al. TRPM2 channel deficiency prevents delayed cytosolic Zn2+ accumulation

and CA1 pyramidal neuronal death after transient global ischemia. Cell death & disease 5, e1541

(2014).

86. Venkatachalam, K. & Montell, C. TRP channels. Annual review of biochemistry 76, 387-

417 (2007).

87. Ramsey, I.S., Delling, M. & Clapham, D.E. An introduction to TRP channels. Annual

review of physiology 68, 619-647 (2006).

88. Hellmich, U.A. & Gaudet, R. Structural biology of TRP channels. Handbook of

experimental pharmacology 223, 963-990 (2014).

89. Eisfeld, J. & Luckhoff, A. Trpm2. Handbook of experimental pharmacology, 237-252

(2007).

90. Jiang, L.H., Yang, W., Zou, J. & Beech, D.J. TRPM2 channel properties, functions and

therapeutic potentials. Expert opinion on therapeutic targets 14, 973-988 (2010).

91. Kraft, R. & Harteneck, C. The mammalian melastatin-related transient receptor potential

cation channels: an overview. Pflugers Archiv : European journal of physiology 451, 204-211

(2005).

92. Clapham, D.E., Runnels, L.W. & Strubing, C. The TRP ion channel family. Nature

reviews. Neuroscience 2, 387-396 (2001).

93. Shimizu, S., et al. Sensitization of H2O2-induced TRPM2 activation and subsequent

interleukin-8 (CXCL8) production by intracellular Fe(2+) in human monocytic U937 cells. The

international journal of biochemistry & cell biology 68, 119-127 (2015).

Page 86: Effects of TRPM2 Inhibition in Neuroprotection following

73

94. Jang, Y., et al. TRPM2, a Susceptibility Gene for Bipolar Disorder, Regulates Glycogen

Synthase Kinase-3 Activity in the Brain. The Journal of neuroscience : the official journal of the

Society for Neuroscience 35, 11811-11823 (2015).

95. Sumoza-Toledo, A. & Penner, R. TRPM2: a multifunctional ion channel for calcium

signalling. The Journal of physiology 589, 1515-1525 (2011).

96. Jiang, L., et al. Mitochondria dependent pathway is involved in the protective effect of

bestrophin-3 on hydrogen peroxide-induced apoptosis in basilar artery smooth muscle cells.

Apoptosis : an international journal on programmed cell death 18, 556-565 (2013).

97. Ogawa, N., Kurokawa, T. & Mori, Y. Sensing of redox status by TRP channels. Cell

calcium (2016).

98. Zeng, B., Chen, G.L. & Xu, S.Z. Divalent copper is a potent extracellular blocker for

TRPM2 channel. Biochemical and biophysical research communications 424, 279-284 (2012).

99. Simon, F., Varela, D. & Cabello-Verrugio, C. Oxidative stress-modulated TRPM ion

channels in cell dysfunction and pathological conditions in humans. Cellular signalling 25,

1614-1624 (2013).

100. Kolisek, M., Beck, A., Fleig, A. & Penner, R. Cyclic ADP-ribose and hydrogen peroxide

synergize with ADP-ribose in the activation of TRPM2 channels. Molecular cell 18, 61-69

(2005).

101. Naziroglu, M. & Luckhoff, A. A calcium influx pathway regulated separately by

oxidative stress and ADP-Ribose in TRPM2 channels: single channel events. Neurochemical

research 33, 1256-1262 (2008).

102. Starkus, J., Beck, A., Fleig, A. & Penner, R. Regulation of TRPM2 by extra- and

intracellular calcium. The Journal of general physiology 130, 427-440 (2007).

103. Gelderblom, M., et al. Transient receptor potential melastatin subfamily member 2 cation

channel regulates detrimental immune cell invasion in ischemic stroke. Stroke; a journal of

cerebral circulation 45, 3395-3402 (2014).

104. Kahya, M.C., Naziroglu, M. & Ovey, I.S. Modulation of Diabetes-Induced Oxidative

Stress, Apoptosis, and Ca2+ Entry Through TRPM2 and TRPV1 Channels in Dorsal Root

Ganglion and Hippocampus of Diabetic Rats by Melatonin and Selenium. Molecular

neurobiology 54, 2345-2360 (2017).

105. Takahashi, N., Kozai, D., Kobayashi, R., Ebert, M. & Mori, Y. Roles of TRPM2 in

oxidative stress. Cell calcium 50, 279-287 (2011).

106. Akpinar, H., Naziroglu, M., Ovey, I.S., Cig, B. & Akpinar, O. The neuroprotective action

of dexmedetomidine on apoptosis, calcium entry and oxidative stress in cerebral ischemia-

induced rats: Contribution of TRPM2 and TRPV1 channels. Scientific reports 6, 37196 (2016).

107. Chong, Z.Z., Li, F. & Maiese, K. Oxidative stress in the brain: novel cellular targets that

govern survival during neurodegenerative disease. Progress in neurobiology 75, 207-246 (2005).

108. Fonfria, E., et al. Amyloid beta-peptide(1-42) and hydrogen peroxide-induced toxicity

are mediated by TRPM2 in rat primary striatal cultures. Journal of neurochemistry 95, 715-723

(2005).

109. Mattson, M.P., et al. beta-Amyloid peptides destabilize calcium homeostasis and render

human cortical neurons vulnerable to excitotoxicity. The Journal of neuroscience : the official

journal of the Society for Neuroscience 12, 376-389 (1992).

110. Ostapchenko, V.G., et al. The Transient Receptor Potential Melastatin 2 (TRPM2)

Channel Contributes to beta-Amyloid Oligomer-Related Neurotoxicity and Memory Impairment.

The Journal of neuroscience : the official journal of the Society for Neuroscience 35, 15157-

15169 (2015).

Page 87: Effects of TRPM2 Inhibition in Neuroprotection following

74

111. Haraguchi, K., et al. TRPM2 contributes to inflammatory and neuropathic pain through

the aggravation of pronociceptive inflammatory responses in mice. The Journal of neuroscience

: the official journal of the Society for Neuroscience 32, 3931-3941 (2012).

112. Xie, Y.F., et al. Dependence of NMDA/GSK-3beta mediated metaplasticity on TRPM2

channels at hippocampal CA3-CA1 synapses. Molecular brain 4, 44 (2011).

113. Kraft, R., et al. Hydrogen peroxide and ADP-ribose induce TRPM2-mediated calcium

influx and cation currents in microglia. American journal of physiology. Cell physiology 286,

C129-137 (2004).

114. Uchida, K., et al. Lack of TRPM2 impaired insulin secretion and glucose metabolisms in

mice. Diabetes 60, 119-126 (2011).

115. Huang, S., et al. Transient receptor potential melastatin 2 channels (TRPM2) mediate

neonatal hypoxic-ischemic brain injury in mice. Experimental neurology 296, 32-40 (2017).

116. Zhong, Z., et al. TRPM2 links oxidative stress to NLRP3 inflammasome activation.

Nature communications 4, 1611 (2013).

117. Chen, G.L., et al. Pharmacological comparison of novel synthetic fenamate analogues

with econazole and 2-APB on the inhibition of TRPM2 channels. British journal of

pharmacology 167, 1232-1243 (2012).

118. Hill, K., McNulty, S. & Randall, A.D. Inhibition of TRPM2 channels by the antifungal

agents clotrimazole and econazole. Naunyn-Schmiedeberg's archives of pharmacology 370, 227-

237 (2004).

119. Togashi, K., Inada, H. & Tominaga, M. Inhibition of the transient receptor potential

cation channel TRPM2 by 2-aminoethoxydiphenyl borate (2-APB). British journal of

pharmacology 153, 1324-1330 (2008).

120. Shimizu, S., et al. Inhibitory effects of AG490 on H2O2-induced TRPM2-mediated

Ca(2+) entry. European journal of pharmacology 742, 22-30 (2014).

121. Lafemina, M.J., Sheldon, R.A. & Ferriero, D.M. Acute hypoxia-ischemia results in

hydrogen peroxide accumulation in neonatal but not adult mouse brain. Pediatric research 59,

680-683 (2006).

122. Rice, J.E., 3rd, Vannucci, R.C. & Brierley, J.B. The influence of immaturity on hypoxic-

ischemic brain damage in the rat. Annals of neurology 9, 131-141 (1981).

123. Lubics, A., et al. Neurological reflexes and early motor behavior in rats subjected to

neonatal hypoxic-ischemic injury. Behavioural brain research 157, 157-165 (2005).

124. Xiao, A.J., et al. Marine compound xyloketal B reduces neonatal hypoxic-ischemic brain

injury. Marine drugs 13, 29-47 (2015).

125. Xu, B., et al. Neuroprotective Effects of a PSD-95 Inhibitor in Neonatal Hypoxic-

Ischemic Brain Injury. Molecular neurobiology (2015).

126. Jarvik, M.E. & Kopp, R. An improved one-trial passive avoidance learning situation.

Psychological reports 21, 221-224 (1967).

127. Castellano, C. & Pavone, F. Effects of ethanol on passive avoidance behavior in the

mouse: involvement of GABAergic mechanisms. Pharmacology, biochemistry, and behavior 29,

321-324 (1988).

128. Seo, I.A., et al. Janus Kinase 2 Inhibitor AG490 Inhibits the STAT3 Signaling Pathway

by Suppressing Protein Translation of gp130. The Korean journal of physiology &

pharmacology : official journal of the Korean Physiological Society and the Korean Society of

Pharmacology 13, 131-138 (2009).

129. Chai, H.T., Yip, H.K., Sun, C.K., Hsu, S.Y. & Leu, S. AG490 suppresses EPO-mediated

activation of JAK2-STAT but enhances blood flow recovery in rats with critical limb ischemia.

Journal of inflammation 13, 18 (2016).

Page 88: Effects of TRPM2 Inhibition in Neuroprotection following

75

130. Davoodi-Semiromi, A., Hassanzadeh, A., Wasserfall, C.H., Droney, A. & Atkinson, M.

Tyrphostin AG490 agent modestly but significantly prevents onset of type 1 in NOD mouse;

implication of immunologic and metabolic effects of a Jak-Stat pathway inhibitor. Journal of

clinical immunology 32, 1038-1047 (2012).

131. Higuchi, T., et al. Prevention of acute lung allograft rejection in rat by the janus kinase 3

inhibitor, tyrphostin AG490. The Journal of heart and lung transplantation : the official

publication of the International Society for Heart Transplantation 24, 1557-1564 (2005).

132. Toda, T., Yamamoto, S., Yonezawa, R., Mori, Y. & Shimizu, S. Inhibitory effects of

Tyrphostin AG-related compounds on oxidative stress-sensitive transient receptor potential

channel activation. European journal of pharmacology 786, 19-28 (2016).

133. Chen, W.L., et al. Xyloketal B suppresses glioblastoma cell proliferation and migration

in vitro through inhibiting TRPM7-regulated PI3K/Akt and MEK/ERK signaling pathways.

Marine drugs 13, 2505-2525 (2015).

134. Turlova, E., et al. TRPM7 Regulates Axonal Outgrowth and Maturation of Primary

Hippocampal Neurons. Molecular neurobiology 53, 595-610 (2016).

135. Wang, H., et al. Tideglusib, a chemical inhibitor of GSK3beta, attenuates hypoxic-

ischemic brain injury in neonatal mice. Biochimica et biophysica acta 1860, 2076-2085 (2016).

136. Volpe, J.J. Neonatal encephalopathy: an inadequate term for hypoxic-ischemic

encephalopathy. Annals of neurology 72, 156-166 (2012).

137. Hagberg, H., Andersson, P., Kjellmer, I., Thiringer, K. & Thordstein, M. Extracellular

overflow of glutamate, aspartate, GABA and taurine in the cortex and basal ganglia of fetal

lambs during hypoxia-ischemia. Neuroscience letters 78, 311-317 (1987).

138. Choi, D.W. Calcium-mediated neurotoxicity: relationship to specific channel types and

role in ischemic damage. Trends in neurosciences 11, 465-469 (1988).

139. Choi, D.W. Excitotoxic cell death. Journal of neurobiology 23, 1261-1276 (1992).

140. McDonald, J.W., Roeser, N.F., Silverstein, F.S. & Johnston, M.V. Quantitative

assessment of neuroprotection against NMDA-induced brain injury. Experimental neurology

106, 289-296 (1989).

141. Represa, A., Tremblay, E. & Ben-Ari, Y. Transient increase of NMDA-binding sites in

human hippocampus during development. Neuroscience letters 99, 61-66 (1989).

142. Tremblay, E., Roisin, M.P., Represa, A., Charriaut-Marlangue, C. & Ben-Ari, Y.

Transient increased density of NMDA binding sites in the developing rat hippocampus. Brain

research 461, 393-396 (1988).

143. Danysz, W. & Parsons, C.G. Glycine and N-methyl-D-aspartate receptors: physiological

significance and possible therapeutic applications. Pharmacological reviews 50, 597-664 (1998).

144. Riikonen, R.S., Kero, P.O. & Simell, O.G. Excitatory amino acids in cerebrospinal fluid

in neonatal asphyxia. Pediatric neurology 8, 37-40 (1992).

145. Holmes, G.L. The long-term effects of seizures on the developing brain: clinical and

laboratory issues. Brain & development 13, 393-409 (1991).

146. Holmes, G.L. & Ben-Ari, Y. The neurobiology and consequences of epilepsy in the

developing brain. Pediatric research 49, 320-325 (2001).

147. Beurel, E., Grieco, S.F. & Jope, R.S. Glycogen synthase kinase-3 (GSK3): regulation,

actions, and diseases. Pharmacology & therapeutics 148, 114-131 (2015).

148. Jope, R.S., Yuskaitis, C.J. & Beurel, E. Glycogen synthase kinase-3 (GSK3):

inflammation, diseases, and therapeutics. Neurochemical research 32, 577-595 (2007).

149. Shim, S.S. & Stutzmann, G.E. Inhibition of Glycogen Synthase Kinase-3: An Emerging

Target in the Treatment of Traumatic Brain Injury. Journal of neurotrauma (2016).

Page 89: Effects of TRPM2 Inhibition in Neuroprotection following

76

150. Nadler, J.J., et al. Automated apparatus for quantitation of social approach behaviors in

mice. Genes, brain, and behavior 3, 303-314 (2004).

151. Website: © 2015 Allen Institute for Brain Science. Allen Developing Mouse Brain Atlas

[Internet]. Available from: http://developingmouse.brain-map.org. 2017.