the impact of ros scavenging on nmda and ampa receptor … · 2013-11-22 · i would like to thank...

121
The impact of ROS scavenging on NMDA and AMPA receptor whole cell currents in pyramidal neurons of the anoxia tolerant western painted turtle by David Dukoff A thesis submitted in conformity with the requirements for the degree of Master of Science Cell & Systems Biology University of Toronto © Copyright by David Dukoff (2013)

Upload: others

Post on 28-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

The impact of ROS scavenging on NMDA and AMPA receptor whole cell currents in pyramidal neurons of the

anoxia tolerant western painted turtle

by

David Dukoff

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

Cell & Systems Biology University of Toronto

© Copyright by David Dukoff (2013)

Page 2: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

ii

The impact of ROS scavenging on NMDA and AMPA receptor

whole cell currents in pyramidal neurons of the anoxia tolerant

western painted turtle

David Dukoff

Master of Science

Cell and Systems Biology

University of Toronto

2013

ABSTRACT

Extended periods of oxygen deprivation cause brain death in mammals but the western painted

turtle overwinters in anoxic mud for months without damage. Neural protection is achieved

through decreases in the whole cell currents of N-methyl-D-aspartate and alpha-amino-3-

hydroxy-5-methyl-4-isoxazolepropionic acid receptors (NMDAR and AMPAR) that are

dependent on a mild increase in intracellular calcium from the mitochondria. The goal of this

research was to determine if natural anoxic decreases in reactive oxidative species (ROS) serve

as the signal to bring about these changes. Reductions in cellular ROS levels were demonstrated

to have no effect on AMPAR currents or intracellular calcium and produced massive increases in

NMDAR currents, indicating that ROS depression does not directly mediate anoxic alterations.

Interestingly, mammalian neural tissue also experiences a similar increase in NMDAR whole cell

current in response to reducing agents suggesting a possible conserved mechanism for normoxic

receptor control.

Page 3: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

iii

ACKNOWLEDGMENTS

“Complete and finished.” That is what I have been told that the writing and submission of

a master’s thesis feels like. Within a hundred or so pages students summarize two years of

laboratory work, complete the requirements of their degree and move to the next phase of their

careers. However, as I approach the end of this project, this feeling of closure has yet to sink in.

During my first year living at the University of Toronto’s Graduate Residence, I was told that the

reason so much of the building seemed raw and unfinished was due to the architect’s belief that

“a graduate student’s work is never done”. Although I came to believe that this explanation was

an excuse for ill-placed support struts and walls so thin you could hear your roommates shift

their blankets, the phrase stuck a chord. Science is a potent addiction and no matter where my

life goes after my degree I don’t think I will ever stop thinking like a researcher or cease

studying the world around me. What is more, the people that have helped and supported me

during my time here are similarly addicting. This section is for all of them.

I would like to begin by thanking the professors and administrative staff whose efforts

have made my project and degree possible. I would like to thank Dr. Les Buck for everything he

has to done to support me during my time in the lab and make research fun. Aside from being a

constant presence to help me with nearly every aspect of my project, he has also been there to

trade jokes and offer advice on all manner of non-science related issues. He has kept me on track

but never been afraid to let me make my own mistakes (of which there have been many) so that I

can learn how to recover and be a better scientist. I would also like to thank my committee

members for getting me this far. The advice provided by Dr. Martin Wojtowicz and Dr. David

Lovejoy has helped shape the directions and aims of my experiments. More importantly, their

questions and comments have kept me versed in information outside the scope of my project and

reminded me of the value of the big picture. Finally, I would thank the administrative staff who

make this department work and who put up with the strange creatures that are graduate students.

A special thanks to Ian Buglass, Tamar Mamourian, Peggy Salmon, James Dix, and Trung Luu.

I would next like to thank my fellow students in our incredible department for the help

and support they have shown me over the last two years. I would like to first thank David Hogg

who, through unstinting assistance, opinionated discussions (often involving words I can’t

include in a thesis) and stories about his family, has kept me going and reminded me that there is

Page 4: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

iv

more to life than what is under the microscope. Next I would like to thank Peter Hawrysh for

being a seemingly infinite resource of technical advice, scientific explanations, dirty stories,

nerdy references, awesome video games, and cold beer (not usually during work hours). I would

like to thank Aaron Chowdhury for his electronic expertise, great sense of humor, inquisitive

questions, and devious pranks that have kept me sharp, motivated, and protective of my laptop. I

would like to thank Hilary Bond for her stories and comments which have helped shape my

decisions about who I want to be. I would like to thank Lydia Farnell for giving me someone to

trade country stories and country music with. I would also like to thank all of my fellow

colleagues in the department whose friendships have kept me sane. A special thanks to Jessica

Pressey, Louise de Lannoy, Zoltan Torontali, Dave Wasserman, Sabina Romanescu, Felix

Gunawan and all the members of CSBGU. I would not have made it without all of you.

I would like to thank my friends and family outside the lab for always being there. My

parents have provided unconditional love and support for every choice I have made. Through

visits to take me out to lunch and time spent editing my writing, they have helped me in any way

possible. I hope I have made them proud. I would like to thank my little brother Andrew for

sending me music on lonely nights at the lab. I would like to thank David Darling and my

godchild Kaylie for Sudbury vacations that recharged my batteries. Finally, I would like to thank

my Toronto/Kitchener friends for creating occasions to get out of the lab and into the sunlight.

Special thanks to Brittany, Serge, Robert and Vanessa for their efforts in this regard.

The last acknowledgement I would like to make is to science and nature (not to be

confused with the professional journals). I went into biology for the same reason I spent my

childhood taking things apart in my Dad’s workshop: I wanted to understand how things work.

There are no greater puzzles or more complex mechanisms than those found in the natural world

and this may be why I find myself, like so many others, completely obsessed with studying it.

The famous biologist Lewis Thomas once wrote about scientific research, “I don't know of any

other human occupation, even including what I have seen of art, in which the people engaged in

it are so caught up, so totally pre-occupied, so driven beyond their strength and resources…

There is nothing to touch the spectacle.” Thank you science for being so interesting.

Page 5: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

v

TABLE OF CONTENTS

ABSTRACT………………………………………………………………………………………ii

ACKNOWLEDGEMENTS……………………………………………….……………………iii

TABLE OF CONTENTS…………………..……………………………………………………v

LIST OF FIGURES……………………………………………………………………………..ix

ABBREVIATIONS……………………………………………………………………………...xi

CHAPTER 1: INTRODUCTION………..……………………………………………………..1

1.1 The importance of oxygen and low oxygen tolerance………………………………...1

1.2 Ice ages, glacial periods and their contributions to modern day biodiversity…………2

1.3 Life after glaciation and adaptations to low oxygen…………………………………..3

1.3.1 Chrysemys picta belli………………………………………………………..4

1.3.2 Spermophilus parryii parryii………………………………...……………...6

1.3.3 Carassius carassius…………………………………………..……………..8

1.4 Requirements for anoxia/hypoxia tolerance…………………………………………..9

1.5 The importance of neuronal down regulation………………………………………..10

1.6 Models for potential human application……………………………………………..11

1.7 Turtle synaptic organization………………………………………………………….12

1.8 Anoxia mediated neuroprotective mechanisms……………………………………...13

1.8.1 Spike arrest…………………………………………………………………13

1.8.2 Channel arrest……………………………………………………………...14

1.9 Mechanisms controlling “channel arrest”……………………………………………16

1.9.1 Increases in adenosine……………………………………………………...16

1.9.2 Activation of ATP-sensitive K+ (KATP) channels………………………..17

1.9.3 Increases in intracellular calcium…………………………………………..19

Page 6: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

vi

1.9.4 Enhancements of calmodulin and phosphatase activity……………………19

1.10 Reactive oxidative species (ROS)…………………………………………………..20

1.11 The role of ROS in anoxia-intolerant vertebrates…………………………………..22

1.11.1 Redox changes affect NMDAR but not AMPAR activity………………..22

1.11.2 Alterations in ROS affect NMDAR activity……………………………...23

1.11.3 NMDAR redox regulation is blocked by kinase inhibition………………24

1.11.4 mKATP mediated ROS increases affect ischemic preconditioning……...26

1.12 The antioxidant defence system…………………………………………………….27

1.13 ROS regulation of GABA in anoxic turtle cortex…………………………………..28

1.14 Rationale for study………………………………………………………………….29

CHAPTER 2: MATERIALS AND METHODS……………………………………………...30

2.1 Animals………………………………………………………………………………31

2.2 Dissection and whole-cell patch-clamp recording protocol………………………….30

2.3 Neuron identification………………………………………………………………...34

2.4 Evoked NMDAR and AMPAR current amplitude recordings………………………34

2.5 Fluorescence investigation of cytosolic Ca2+ changes……………………………….36

2.6 Whole cell spontaneous neuronal activity recordings………………………………..38

2.7 Pharmacology and drug application………………………………………………….39

2.8 Measurement of oxygen tension……………………………………………………..40

2.9 Chemicals…………………………………………………………………………….41

2.10 Statistical analysis…………………………………………………………………..41

CHAPTER 3: RESULTS………...…………………………………………………………….42

3.1 Pre-current recording evaluations……………………………………………………42

3.1.1 Anoxic conditions are produced within the bathing chamber……………...42

Page 7: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

vii

3.1.2 All recordings were conducted within pyramidal cells…………………….43

3.2 Whole-cell NMDAR current experiments…………………………………………...44

3.2.1 Normoxic NMDAR currents are stable throughout the time course………44

3.2.2 Normoxic NMDAR current amplitudes are reduced during anoxia……….45

3.2.3 NMDAR current amplitudes are increased by ROS scavenging…………..47

3.2.4 NMDAR current amplitudes are decreased by H2O2 addition……………..49

3.2.5 Blocking mitochondrial ROS production increases NMDAR currents……49

3.2.6 Anoxia attenuated NMDAR currents are not reversed by ROS addition….51

3.3 Whole-cell AMPAR current experiments……………………………………………53

3.3.1 Normoxic AMPAR currents are stable throughout the time course……….53

3.2.2 Normoxic AMPAR current amplitudes are reduced during anoxia………..53

3.3.3 AMPAR current amplitudes are unaffected by ROS scavenging………….55

3.3.4 AMPAR current amplitudes are unaffected by H2O2 addition…………….55

3.4 Oregon green intracellular calcium assessments…………………………………….57

3.4.1 Excitation scans confirmed dye loading in turtle brain sheets……………..57

3.4.2 Normoxic calcium concentrations are stable throughout the time course…57

3.4.3 Intracellular calcium concentrations are increased by anoxia……………..58

3.4.4 Intracellular calcium concentrations are unaffected by ROS scavenging….58

3.4.5 Intracellular calcium concentrations are unaffected by H2O2 addition…….59

3.5 Action potential frequency assessments in spontaneously active pyramidal cells…..62

3.5.1 Action potential frequency is reduced by ROS scavenging………………..62

CHAPTER 4: DISCUSSION…………………………………………...……………………...64

4.1 Control normoxic and anoxic NMDA and AMPA currents…………………………64

4.2 Pharmacological manipulation of ROS………………………………………………64

Page 8: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

viii

4.2.1 The NMDAR modulatory redox site in turtle cortical slices………………64

4.2.2 Mitochondrial produced ROS in NMDAR redox modulation……………..67

4.2.3 A potential mechanism for ROS mediated NMDAR control……………...69

4.2.4 AMPAR redox insensitivity………………………………………………..74

4.2.5 Anoxia-mediated block of NMDAR redox control………………………..75

4.3 A role for anoxia-mediated calcium increases in blocking redox control…………...76

4.4 Roles for redox sensitive GABA release…………………………………………….79

4.5 Potential mechanism of NMDAR/AMPAR control under anoxic conditions……….80

4.6 Concluding remarks and directions for future research……………………………...83

REFERENCES………………………………………………………………………………….85

Page 9: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

ix

LIST OF FIGURES

CHAPTER 1: INTRODUCTION

Figure 1.1: The ranges of the four currently recognized subspecies of Chrysemys picta……...…6

Figure 1.2: NMDAR and AMPAR structures……………………………………………………16

Figure 1.3: Schematic illustration of anoxia-mediated changes contributing to channel arrest…20

Figure 1.4: The mechanisms of ROS production and cysteine oxidation………………………..25

CHAPTER 2: MATERIALS AND METHODS

Figure 2.1: Diagrammatic representation of cortical sheet excision and experimental setup…...33

Figure 2.1: NMDAR and AMPAR whole-cell patch clamp recording timeline………………...35

Figure 2.2: Oregon Green fluorescent recording timeline……………………………………….38

Figure 2.3: Whole cell spontaneous activity recording timeline………………………………...39

CHAPTER 3: RESULTS

Figure 3.1: Change in oxygen tension during normoxic to anoxic transition……………………43

Figure 3.2: Spike frequency adaptation in pyramidal and stellate neurons……………………...44

Figure 3.3: Effect of anoxia on whole cell NMDAR currents…………...………….…………...46

Figure 3.4: Effect of ROS scavenging on whole cell NMDAR currents………………………...48

Figure 3.5: Effect of H2O2 manipulation on whole cell NMDAR currents……………………...50

Figure 3.6: Effect of H2O2 addition on anoxia-attenuated whole cell NMDAR currents………52

Figure 3.7: Effect of anoxia on whole cell AMPAR currents……………………………………54

Figure 3.8: Effect of ROS manipulation on whole cell AMPAR currents………………………56

Figure 3.9: Oregon Green excitation scans………………………………………………………57

Figure 3.10: Effect of oxygen and ROS manipulation on Oregon Green fluorescence……...….60

Figure 3.11: Sample Oregon Green fluorescence traces…………………………………………61

Figure 3.12: Changes in spontaneous activity in response to ROS scavenging…………………63

CHAPTER 4: DISCUSSION

Page 10: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

x

Figure 4.1: Diagrammatic representation of the proposed mechanism for ROS mediated

NMDAR control under normoxic conditions……………………………………………………73

Figure 4.1: Diagrammatic representation of the proposed mechanism for anoxia mediated

NMDAR/AMPAR downregulation……………………………………………………………...82

Page 11: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

xi

ABBREVIATIONS

C degrees Celsius

Ψm mitochondrial membrane potential

[Ca2+]i intracellular calcium concentration

8-PT 8-pentyltheophyilline

5-HD 5-hydroxydecanoic acid

12h:12h twelve hours of light and twelve hours of dark light cycle

aCSF artificial cerebrospinal fluid

AFU arbitrary fluorescence unit

AMP adenosine monophosphate

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

AMPAR α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor

AP action potential

APf action potential frequency

ATP adenosine triphosphate

BAPTA 1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid

BDM butanedione-monoxime

Ca2+ calcium ion

CaCl2 calcium chloride

CaM calmodulin

cGMP cyclic guanosine monophosphate

Cl- chloride ion

CO2 carbon dioxide

Page 12: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

xii

CM-H2DCFDA 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate, acetyl ester

C.p. Chrysemys picta

Cys cysteine

DTNB 5,5'-dithiobis-(2-nitrobenzoic acid)

DTT dithiothreitol

EPSP excitatory postsynaptic potential

EEG electroencephalogram

ER endoplasmic reticulum

Fig Figure

Fmax maximal fluorescence signal

Fmin background fluorescence

GΩ gigaohm

GABA gamma-Aminobutyric acid

GPx glutathione peroxidase

GSH glutathione

GSSG glutathione disulfide

H+ proton

H2O2 hydrogen peroxide

HCO3- bicarbonate

IMM inner mitochondrial membrane

I/V current/voltage

IV intravenous

K+ potassium ion

KATP channel ATP-sensitive potassium channel

Page 13: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

xiii

KCl potassium chloride

Kd dissociation constant

kDA kilo dalton

kHz kilohertz

L litre

MΩ megaohm

MCU mitochondrial calcium uniporter

Mg2+ magnesium ion

MgCl2 magnesium chloride

min minutes

mKATP channel mitochondrial ATP-sensitive potassium channel

mM millimolar

mmHg millimeters of mercury

Mn2+ manganese ion

MnSOD manganese superoxide dismutase

mOsM milliosmole

MPG N-2-mercaptopropionylglycine

mPTP mitochondrial permeability transition pore

ms millisecond

mV millivolt

n sample size

N2 nitrogen gas

Na+ sodium ion

NaCl sodium chloride

Page 14: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

xiv

NAC n-acetylcysteine

NaGTP sodium guanosine triphosphate

NaATP sodium adenosine triphosphate

NaH2PO4 sodium dihydrogen phosphate

NaHCO3 sodium bicarbonate

Na-HEPES sodium 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid

nm nanometre

nM nanomolar

NMDA N-Methyl-D-Aspartate

NMDAR N-Methyl-D-Aspartate receptor

nNOS nitric oxide synthase

NO nitric oxide

O2 molecular oxygen

O2 superoxide anion

ONOO peroxynitrite

pKATP channel plasmalemmal ATP-sensitive potassium channel

PP protein phosphatase

PP2B calcineurin

PSD95 postsynaptic density protein-95

PSC postsynaptic current

Q10 temperature coefficient

ROS reactive oxygen species

[ROS]i intracellular ROS concentration

s seconds

Page 15: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

xv

S. Spermophilus

SA spike arrest

S.E.M. standard error of the mean

sGC soluble guanylate cyclase

SOD superoxide dismutase

S.p. Spermophilus parryii

t time

TTX tetrodotoxin

μM micromolar

WTR water transport route

X/XO xanthine/xanthine oxidase

Page 16: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

1

CHAPTER 1: INTRODUCTION

1.1 The importance of oxygen and low oxygen tolerance

Humans and most other vertebrates require a constant supply of oxygen to survive. Brief

hypoxic exposure causes hyperventilation and tachycardia that temporarily compensate for

decreased systemic oxygen but sustained anoxia for more than a few minutes results in

neurological damage and eventually death [1, 2]. Aerobic life primarily depends on oxidative

phosphorylation for the production of the cellular energy substrate - adenosine triphosphate

(ATP), and if oxygen transport to tissues becomes compromised energy demands cannot be met

and cellular death occurs [3, 4]. Despite these limitations, some vertebrates have developed the

ability to survive under hypoxic or anoxic conditions for extended periods of time.

Understanding the mechanisms through which tolerance is accomplished has applications within

fields as broad as stroke prevention to space travel [5, 6].

Research has been conducted for decades on hypoxia and anoxia tolerant species to try

and better understand their physiological and molecular adaptations. Running in parallel with

this work has been research to try and understand the conditions that produced these adaptations.

Related species to those with anoxia/hypoxia tolerance rarely possess the same capabilities,

indicating a historical pressure that selected for the trait and brought about a speciation event.

Understanding why some species have adapted to survive long term anoxia may help us better

understand how they do it.

Within the northern latitudes a number of hypoxia tolerant mammals and anoxia tolerant

reptiles and fish are found in environments that demonstrate seasonal temperature decreases and

the development of ice cover. In order to survive these seasonal changes species have adapted

various hibernation and overwintering strategies that involve tolerating extended periods of

anoxia/hypoxia. It has been proposed that these survival strategies were selected for during

periods of globally decreased temperature and increased ice cover. The most recent such event

was the last glacial period.

Page 17: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

2

1.2 Ice ages, glacial periods and their contributions to modern day biodiversity

When extensive ice sheets are present in the northern and southern hemispheres the

period is designated as an ice age. Our planet has passed through four major ice ages since its

formation and is currently experiencing the fifth that began 2.6 million years ago marking the

beginning of the Quaternary Period [7]. Environmental conditions during ice ages alternate over

tens of thousands of years between two opposing conditions. “Interglacial” periods are

characterized by warmer global temperatures and glacial recession. “Glacial” periods are marked

by colder global temperatures and ice sheet advances. Movement between these two periods is

brought on by large scale changes in the amount of thermal energy present in the Earth’s

atmosphere and oceans caused by alterations in the energy generated by the sun, changes in the

distance of the Earth’s orbit, and adjustments in atmospheric composition. We are currently

experiencing an interglacial period that began 11,400 years ago [7, 8].

The last glacial period began 120,000 years ago and at that time the regional temperatures

and climates were nearly identical to those seen today. Glaciers expanded across the globe

reaching a maximum between 26,500 and 20,000 years ago when glaciers and permafrost

covered more than a quarter of the Earth’s surface [7, 8]. Approximately 28,000 years ago the

surface temperatures of the oceans began to warm leading to slow rises in ocean levels and by

12,000 years ago most regions returned to climates close to what they had been seen previously.

This marked the transition from glacial to interglacial [7, 8].

Worldwide biodiversity was not significantly affected by the initial interglacial-glacial

transition or the sustained 100,000 yearlong glacial period itself. As ice sheets expanded most

species were able to migrate to areas not covered by ice, commonly referred to as refugia. These

areas were found sparsely throughout glaciated regions but primarily existed closer to the

equator [9]. Changing climate and the addition of new plant and animal life caused these regions

to begin to mimic the environments that had previously been found further north and south and

as a result, migrating animals did not face extensive pressures to adapt or change [7]. Most

species were forced into a third of the territory they occupied pre-glaciation, but mild population

decreases and the establishment of new territorial limits prevented vast losses of biodiversity [7].

Increased glaciation decreased sea levels by 350 feet generating land bridges like from Alaska to

Page 18: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

3

Siberia that allowed for migrations between refugia, further preserving diversity [7, 10].

However as the glacial cycle came to an end, a major global extinction took place.

This extinction event took place between 16,000 and 10,000 years ago, and unlike

previous major global extinction events took place only on land. Nearly all large terrestrial

mammals over 44 kg were wiped out [7, 10]. Similar extinctions did not occur during exit from

previous glacial periods and the cause remains contested. The most supported hypotheses

involve rapid environmental changes that caused the loss of much of the world’s mega flora,

overhunting by early humans as they migrated into new regions following ice sheets recession,

or a combination of the two [7, 10]. Whatever the cause, all of the continents experienced losses

in large terrestrial animal life: South America (83%), North America (72%), Australia (88%),

Eurasia (35%), and Africa (21%) [10]. The mass extinction left a number of environmental

niches empty and provided surviving species with an opportunity to adapt and expand.

1.3 Life after glaciation and adaptations to low oxygen

About 10,000 years ago plant and animal life began to expand outwards from refugia as

the ice sheets receded [7, 9]. Some of these species/subspecies were new, produced from the

isolation of members of the same species in different refugia throughout the glacial cycle where

genetic drift and varying selective pressures caused speciation events [11, 12]. Expansion of

species from refugia following the last glacial cycle altered the biota from what existed pre-

glaciation to a greater degree than seen during exit from previous glacial cycles [13, 14]. The

new territory opened up by the mass extinction event was quickly colonized by smaller faster

breeding species leading to changes in plant and animal life. Adaptations were developed to

survive under these novel conditions increasing differences between new species/subspecies and

establishing new territorial ranges. These adaptations were generally minor and most

species/subspecies continued to live in similar habitats, experiencing only small changes in

physical features, ranges or behaviours [15, 16]. However, following glacial retreat some species

in North America and Europe expanded into regions that still experienced seasonal ice cover,

reduced temperatures and decreasing available food fostering the development of distinct

adaptations. Species with flight capabilities survived by developing adaptations to seasonally

migrate south. Other small and less mobile species adapted to outlast the winters. Three

Page 19: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

4

examples of such species are Chrysemys picta belli, Spermophilus parryii parryii, and Carassius

carassius.

1.3.1 Chrysemys picta belli

The North American turtle Chrysemys picta (C.p.) is the most widely distributed turtle

species in North America with a range extending from the Maritime Provinces west to British

Columbia and south to Oregon, Louisiana, and Georgia [17]. Fossils have been found for the

species indicating that they have been widely distributed for at least two, and possibly more than

five million years [17-19]. Previous to the last glacial period C.p. existed as two forms divided

along the Atlantic coastal and west of the Appalachian Mountains [20]. The expansion of glacial

ice and the loss of vegetation forced both forms southward and isolated the turtles into three

distinct populations that became distinct species with C.p. picta in the southeast Atlantic coast,

C.p. dorsalis in the lower Mississippi and C.p. belli in the southwest [21-23]. During glacial

recession all three of the C.p. species migrated out of refugia north and through interbreeding

between C.p. belli and C.p. dorsalis it is believed a hybridized fourth intermediated taxon C.p.

marginata came about [20]. Species eventually settled into their modern day locales with C.p.

picta in the Atlantic Coastal region, C.p. belli on the Great Plains, C.p. marginata around the

Great Lakes, and C. p. dorsalis within the southeastern United States [Figure 1.1] [20]. Recent

genetic investigation has proposed that interbreeding between species has begun to break down

the classic differentiations between groups and they may be more accurately regarded as a

northern subspecies (C. p. picta, C. p. belli, C.p. marginata) and a southern subspecies (C. p.

dorsalis) [24]. However within the scientific literature the classic separations still remain.

During migrations out of the refugia the northern turtle subspecies slowly adapted to

survive conditions of seasonal ice cover, decreased temperatures and reduced food availability

allowing for the expansion of their ranges [20]. They developed physiological and cellular

adaptations to survive the winter submerged in insulating mud at the bottoms of frozen fresh

water bodies in an unresponsive metabolically reduced state [25, 26]. Although the primary

function of these adaptations was probably freezing avoidance and energy conservation it also

produced capabilities for extreme anoxia tolerance as the overwintering environment required

turtles to survive for months without oxygen. Under laboratory conditions C.p. belli, picta, and

Page 20: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

5

marginata at 3°C have all been demonstrated to survive over 120 days of anoxia, compared to

the southern subspecies (C.p. dorsalis) that does not overwinter and perishes after only 50 days

under identical experimental conditions [27-29]. Anoxia tolerance is greatest within members of

the subspecies found further north and the Western painted turtle (C.p. belli) that inhabits the

most northern ranges has been demonstrated to withstand the longest periods of anoxia [27, 30,

31].

The overwintering capabilities of C.p. belli are thought to have arisen from enhancements

of pre-existing diving abilities common to most turtle species [32, 33]. In comparative analysis

between C.p. dorsalis and C.p. belli it was concluded that increased tolerance is the result of

better capabilities to lower metabolic rate and limit lactate accumulation [27]. During

overwintering energy is conserved via decreases in brain activity, heart contraction, protein

synthesis, urea production and the maintenance of a comatose like state [34-38]. This leads to a

70-80% reduction in ATP turnover [34]. The remaining energy demands are met via glycolytic

fermentation supported by enhanced glycogen stores within the liver (15% of the organs mass)

[39]. The two units of lactate and free hydrogen ions produced for every unit of glucose

anaerobically consumed are buffered by a high plasma bicarbonate concentrations and ion

exchange systems between the shell and bones (comprising 37.5% of their total mass) [40].

Induction of the overwintering state occurs in response to low oxygen and does not appear to be

controlled seasonally like within mammal hibernators [41].

Page 21: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

6

Figure 1.1: The ranges of the four currently recognized Chrysemys picta subspecies.

Figure adapted from online source [42].

1.3.2 Spermophilus parryii parryii

The arctic ground squirrel (Spermophilus parryii parryii) is the largest of the American

ground squirrel genus and the most northern dwelling of the squirrel family. They are found

throughout the Canadian Arctic, subsist socially in large burrows, and during the coldest 7 to 8

months of the year they hibernate in their dens at temperatures sometimes below 0C [43, 44].

S. parryii originally arose in Asia from a common ancestor to the central Siberian S.

undulatus approximately 100 000 years [45, 46]. The speciation event took place during the last

Page 22: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

7

glaciation within a large refugium consisting of portions of eastern Siberia, Alaska, and the

western Yukon Territories connected by the Bering Land Bridge [8, 11, 47-51]. The newly

speciated S. parryii migrated to North America and further subdivided into a large arctic (S. p.

parryii) and subarctic subspecies (S. p. plesius). Following glacial recession the two subspecies

dispersed north into the regions they currently occupy today with S.p. parryii found north of the

Brooks Range in Alaska and arctic Canada and S.p. plesius found south in interior Alaska and

subarctic Canada [45, 46].

Like the western painted turtle the arctic ground squirrel survives the winter through

physiological and cellular changes that conserve energy and allow them to tolerate low

temperatures. During hibernation resting body temperature is reduced to between 2-10 °C, heart

rate decreases (25 fold), neurological protein synthesis is arrested (0.04% of active levels) and

blood flow to various brain regions is reduced (80-90%). These changes cumulatively result in

glucose utilization rates that are 1-2% of resting levels [3, 52-55]. The resting respiration rate is

also decreased (25 fold) and O2 consumption is reduced (90-96%) allowing the squirrels to

survive prolonged durations in burrows that can become hypoxic during the winter season [3,

56]. Metabolic needs are met through the breakdown of fat and lean muscle reserves. Increased

levels of steroids/androgens and lipases three weeks prior to hibernation allows the squirrels to

increase their mass by 42% (87% fat and 3% protein) ensuring an adequate fuel supply [57, 58].

Also like the western painted turtle, the adaptations utilized by the arctic ground squirrel

to survive the winter season are believed to have been produced through modifications of

existing capabilities common to ground squirrels as a whole. Several other grounds squirrel

species hibernate although for shorter periods at not as low temperatures [59]. Most ground

squirrel species also experience oxygen fluctuations within their burrows (6-20%) at different

points of the year [60-62].

Ground squirrels do not remain quiescent throughout the hibernation period and at

regular periods briefly warm and reverse depressions in brain activity and heart rate without

awakening. These arousal periods are part of the torpor arousal cycle and although energetically

costly, are thought to be necessary for the maintenance of neural tissue and to undergo sleep

cycles that cannot be accomplished during hibernation [63]. They also allow squirrels to filter

Page 23: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

8

out and excrete metabolic waste products that accumulate during the inactive state. The torpor

arousal cycle and entrance/exit from the hibernation state are based on a circannual cycle and

persists under constant photoperiods with food provided ad libitum [43, 64-66].

1.3.3 Carassius carassius

The crucian carp (Carassius carassius) is widely distributed throughout northern Europe

with a range spanning from England to Russia [67]. The species lives within lakes and ponds

that for several months every winter become anoxic as the surface freezes over preventing

aquatic photosynthesis and atmospheric oxygen diffusion [68]. During this period only the

crucian carp survives, remaining active in water at 2°C, without food, for up to 5.5 months [69].

Previous to the last glacial period, the crucian carp was widely dispersed across Eurasia.

During glaciation, the species survived within bodies of water in southern and eastern refugia

and re-dispersed to central and north European waters following ice sheet recession [70].

Because the carp are aquatic organisms it is unclear if their current range or adaptations to

seasonal ice cover are the result of the post glacial extinction event or are products of previous

glacial periods.

The physiological adaptations that allow the carp to survive under the ice involve

decreases in metabolic rates and body temperature, similar to those seen in the species discussed

above. Body heat production is reduced by 37%, whole organism protein synthesis is down

down-regulated 50-95%, and heart rates is lowered from 88 to 10 beats per minute [71-73].

Although crucian carp remain active throughout the anoxic bout, they do experience a major

reduction in activity within optical centres of the brain (90% decrease) that brings about

blindness without deficits to survival [73, 74]. The cumulative down regulation of these

processes decrease metabolic energy requirements to levels that can be met through anaerobic

glycolysis. To accomplish this, the crucian carp also possess a large glycogen reserve

accounting for 30% of the liver’s wet mass and 4.5% of the fish’s weight that allows them to

survive anaerobically and without feeding through the winter [75]. In order to deal with lactate

accumulation, the carp convert lactate to ethanol and release it through their gills into the

surrounding environment [72]. Similar to the western painted turtle the crucian carp brings about

Page 24: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

9

cellular and physiological changes in response to decreases in available oxygen and not as a

result of circannual rhythms [76].

Similar to the other species discussed above the crucian carp’s adaptations for winter

survival are believed to be the result of adjustments to pre-existing capabilities. The goldfish

(Carassius auratus) is a cousin of the crucian carp that is found farther south in regions

throughout Eastern Asia and sheltered within the same refugia during the last glacial period [70,

77]. In order to tolerate decreases in oxygen brought on by brief ice cover or algae blooms

goldfish have adapted to tolerate several weeks of hypoxia or hours of anoxia [78].

1.4 Requirements for anoxia/hypoxia tolerance

It has been stated that in order for prolonged anoxia/hypoxia tolerance to be

accomplished adaptations must exist that allow organisms to conserve energetic substrates and

prevent self-pollution by end-product accumulation [79]. All of the species discussed above

possess strategies to accomplish these goals, many of which are common between species.

Reduction of metabolic activities is the primary method through which both of these

goals are met, termed “metabolic arrest”. Down regulations of ATP consuming processes

decrease rates of substrate utilization and by-product production. During overwintering all of the

species discussed above experience depressions in heart rate/blood flow, protein synthesis and

brain activity. Decreasing environmental temperatures also allow the species to reduce resting

body temperature and heat production. The temperature effects aid in the reduction of metabolic

activities but are not solely responsible. Most organisms exhibit 2-3 fold depressions per 10 °C

reduction (Q10= 2-3) but the western painted turtle (Q10= 8.4 at 3-10 °C), the arctic ground

squirrel (Q10=14.1 at 4-20 °C), and the crucian carp (Q10=5.1 at 10–15°C) experience greater

decreases indicting other mechanisms at work [37, 80, 81].

Reduced metabolisms are supported for prolonged periods by increased internal fuel

stores. Both the crucian carp and the western painted turtles possess large livers with enhanced

glycogen concentrations in order to support extended anaerobic metabolism [39, 75]. Squirrels

increase fat stores and lean muscle prior to hibernation and aerobically metabolize all of it during

Page 25: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

10

the winter season [57]. Waste products are still produced, although slowly and in small quantities

that can accumulate over the anoxic period. The strategies to deal with this accumulation are

diverse and less universal between the species discussed involving increases in buffering

capabilities, exchange processes between shell/bone, arousal periods to filter blood/excrete

wastes and the conversion of lactate into ethanol for expulsion into the environment [40, 72, 81,

82].

1.5 The importance of neuronal down regulation

The mammalian brain accounts for around 2% of total body mass but is responsible for

20% of the total body oxygen consumption making it the most energy demanding tissue [83, 84].

It is similarly responsible for a lower but significant percentage of the aerobic capacity within

reptiles and fish [3, 37, 85]. All of the species discussed above undergo decreases in neuronal

function during their overwintering periods believed to be vital for energy conservation and

anoxia/hypoxia tolerance. In the absence of these neuroprotective changes, oxygen deprivation

within the brain triggers a lethal cascade.

Approximately 50-60% of total brain ATP is used by cellular Na+/K+ ATPase in order to

maintain and re-establish cellular ion gradients [83, 84]. The gradients are perturbed through the

activities of post-synaptic currents, action potentials and neurotransmitter uptake [86]. The high

proportion of energy devoted to simply maintaining homeostasis makes neurons extremely

vulnerable to excitability during times of ATP depletion like hypoxia/anoxia. The loss of the

membrane ion gradients results in a net outward movement of K+ ions preventing cells from

repolarizing after firing. Na+ and Ca2+ channels become over activated and the whole cell is

depolarized. The resulting Ca2+ influx into the cell triggers vesicular release of excitatory

neurotransmitters such as glutamate into the synaptic cleft activating glutamatergic receptors on

associated cells [87, 88]. Neurotransmitter excision is compromised by ATP depletion and

glutamate lingers in the synapse causing receptor over activation and excessive Ca2+ influx.

Ca2+ plays important roles in second messenger pathways and metabolic regulation but in

high quantities is toxic to the cell. Under normoxic conditions extracellular Ca2+ levels are

maintained 10,000 times greater than intracellular levels through the activities of plasma

Page 26: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

11

membrane transporters. Inside the cell calcium is stored in the mitochondria and endoplasmic

reticulum or sequestered by calcium binding proteins [92-94]. These processes are ATP

dependent and compromised by anoxia/hypoxia [95]. Excessive glutamatergic activation and

compromised Ca2+ management capabilities result in excessive intracellular concentrations of

free Ca2+ that cause cell swelling and trigger enzymatic cascades that eventually bring about cell

death [85][96]. This is termed excitotoxic cell death (ECD).

1.6 Models for potential human application

Stroke represents the third leading cause of death in North America killing 14,000

Canadians each year and hospitalizing a further 46,000 that may suffer neurological impairments

for the rest of their lives [89-91]. This represents a cost of $3.6 billion a year in physician

services, hospital fees, lost wages, and decreased productivity [92]. About 80% of all strokes are

caused by the interruption of blood flow to the brain due to a blood clot. The affected brain

regions are deprived of glucose and oxygen (ischemia), triggering the neurotoxic set of events

discussed above.

Tissue death caused by an obstruction of blood supply is termed infarction and cell death

in the tissues localized to the blockage is an infarct. Until blood flow is returned the infarct

grows in size affecting a greater proportion of the surrounding tissue. Within mammals, the size

of this infarct can be reduced and cell death prevented through “ischemic preconditioning” that

involves subjecting whole organisms to brief periods of ischemia prior to a prolonged lethal

ischemic insults in order to reduce infarct sizes. Ischemic preconditioning was first

demonstrated in canine myocardial tissue and has since been identified to offer protective roles

in a multitude of mammalian models within organ systems including brain, heart, liver, small

intestine, skeletal muscle, kidney and lung [93, 94] . In neurological evaluations in rats and

gerbils the development of global ischemia through temporary surgical blockage of the carotid

artery for 2-3 minutes reduced infarct sizes in response to lethal ischemia (5-10 minutes) by 53-

98%. Neuroprotection from lethal ischemia required 24-72 hours to develop after the initial

ischemic exposure and was maintained for 1-7 days [95-98].

Page 27: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

12

The time span required for ischemic preconditioned neuroprotection to be achieved and

the short amount of time it is maintained for limit practical applications within the field of

medicine. As well, repeatedly exposing patients to brief ischemic periods is dangerous and

impractical. Attempts have been made to pharmacologically mimic the protective response but

have met with limited success in human application [94, 99]. Finally, ischemic preconditioning

has been demonstrated to reduce the size of infarcts but not prevent them all together resulting in

minor to moderate brain damage.

The hypoxia/anoxia tolerant organisms discussed above do not suffer neurological

impairments during their hibernation periods. Studying the mechanisms through which their

tolerances are achieved may offer an alternative route to dealing with conditions brought on by

blood flow reduction or may aide in our understanding and successful application of the

mechanisms controlling ischemic preconditioning. Preconditioning reveals that even within

mammals there is capacity to increase the hypoxia/anoxia tolerance of a tissue and the

underlying mechanisms may be similar to those used by anoxia-tolerant species. Of the three

species discussed above, the western painted turtle is the ideal model for study. As a terrestrial

vertebrate they are a better comparative model for potential human application than the crucian

carp. The neurological mechanisms permitting anoxia tolerance are triggered rapidly in response

to oxygen deprivation and not controlled circanually like the arctic ground squirrel. Lastly the

western painted turtle brain is tolerant to ischemia as well as anoxia [100].

1.7 Turtle synaptic organization

The anatomy and synaptic organization of the turtle brain has been investigated within

various turtle species and some of the structures have been compared against those found within

other organisms [101-104]. The turtle dorsal cortex shares several important anatomical

similarities with the mammalian neocortex including afferent and efferent connections to various

regions of the brain and electrically innervated organs [104-106]. However, although the

extrinsic connectivity of the turtle dorsal cortex resembles neocortex, the intracortical structure

of the turtle is greatly simplified compared to that of the mammal. Instead of a six-layered

isocortex, the turtle has only three cortical layers (molecular, cell and subcellular) that are

composed of two general classes of neurons: pyramidal cells that form the main output of the

Page 28: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

13

cortex, and stellate interneurons [104, 107, 108]. The turtle brain has similarities to the

mammalian brain allowing for a degree of comparability in the context of anoxic/ischemic

investigation. However, it should be stressed that the resemblance between organs is limited.

Electrophysiological analyses have been performed within the dorsal cortex of the turtle

Pseudemys scripta elegans in order to better characterize the pyramidal and stellate cell types.

The pyramidal cells have their somata in the cell layer, extend several densely spined apical

dendrites through the molecular layer to the pia, and have spiny basilar dendrites directed

through the subcellular layer toward the ependymal border. Physiologically, pyramidal cells have

relatively prolonged action potentials that show marked frequency adaptation during a sustained

suprathreshold current pulse. In contrast, stellate cells have their somata in the molecular and

subcellular zones, and their dendrites are horizontally oriented and spine-free. Stellate cells have

relatively brief action potentials and show little or no spike frequency adaptation [101].

1.8 Anoxia mediated neuroprotective mechanisms

As discussed above, entrance into the overwintering state by the western painted turtle is

associated with major decreases in metabolic processes. Although the effects of decreasing

temperature aid in reducing metabolism (Q10 effects) oxygen deprivation is the primary trigger

for metabolic down regulation. Turtles submerged in tanks at 20 C experience a four-fold

depression in the frequency of heart contractions, and after 8 hours exhibit an 80 % reduction in

metabolic rate [35]. Similarly, submergence at 25 C decreases protein synthesis and urea

production by 92% and 72% respectively after 12 hours [36]. Anoxic down regulation of

metabolic activity leads to reductions in neuronal activity and the induction of a comatose-like

state as a result of spike and channel arrest.

1.8.1 Spike arrest

Gamma-aminobutyric acid (GABA) is the primary inhibitory neurotransmitter within the

mature central nervous system of vertebrates and mediates wide spread electrical depression in

most neurons [109-111]. Activation of postsynaptic GABAA receptors increases chloride (Cl-)

conductance through the channel’s pore region. Activation of postsynaptic GABAB receptors

increases K+ conductance via G-protein coupling to K+ channels. The reversal potentials for Cl-

Page 29: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

14

and K+ are near neuronal resting potential and opening of GABA receptors generates an

inhibitory shunting current near the GABA reversal potential (EGABA) that impedes action

potential (AP) generation [112]. Within the brain of the western painted turtle, extracellular

GABA concentrations are elevated to 90 times normoxic levels after 4 hours of anoxia. As well

GABAA-receptor density increases by 22% and 29% after 12 and 24 hours of anoxia,

respectively [113, 114]. This produces decrease in whole brain activity demonstrated by

reductions in action potential frequency (APf), field potentials and electroencephalogram (EEG)

activity in the anoxic turtle [115-117]. Depressed electrical activity is termed “spike arrest” and

serves to conserve ATP throughout the anoxic period [118].

1.8.2 Channel arrest

Activation of post-synaptic N-methyl-D-aspartate receptors (NMDAR) is the primary

route for extracellular Ca2+ entry. NMDARs are hetero-tetramers formed by two GluN1 and two

GluN2 subunits (previously denoted as NR1 and NR2) of which there are four subtypes in

vertebrates (GluN2A-D) [Figure 1.2]. In mammals GluN2A and GluN2B are expressed

throughout the forebrain including the hippocampus, GluN2C is restricted to the cerebellum and

GluN2D is found predominantly in the midbrain [119]. Binding of glutamate to the GluN1 and

the co-agonist glycine to the GluN2 is necessary to trigger Na+/Ca2+ influx and K+ outflow [120].

NMDAR activation is inhibited at resting membrane potentials by a magnesium ion (Mg2+)

block within the pore region. The blockade is maintained in a voltage-dependent manner and

removed by cellular depolarization following postsynaptic α-amino-3-hydroxy-5-methyl-4-

isoxazolepropionic acid receptor (AMPAR) activation. AMPARs are hetero-tetramers composed

of different combinations of the four GluA (previously denoted as GluR) subtypes (GlurA1-4)

each possessing a binding site for glutamate [Figure 1.2]. AMPARs are permeable to Na+, K+

and also Ca2+ in configurations without GluR2. AMPARs are opened when two of the glutamate

binding sites become occupied producing a depolarizing current, the removal of the Mg2+ block

from NMDARs, and Ca2+ influx [121]. Activation of these receptors and the resulting Ca2+

influx plays important roles in learning, memory, synaptic plasticity and cognitive function

[122].

Page 30: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

15

As previously discussed, glutamate concentrations are rapidly elevated following oxygen

deprivation within anoxia intolerant nervous systems. The resulting excessive Ca2+ influx is

produced primarily by NMDAR and AMPAR over activation. In the anoxic brain of the western

painted turtle, excessive glutamate release is partially prevented through GABA mediated

decreases in cell firing that keep glutamate levels relatively constant throughout (∼1 μM) [113,

123]. Adaptations are also present that decrease the degree to which NMDAR and AMPAR are

activated by glutamate during anoxia. Following 40 minutes of anoxia NMDAR and AMPAR

currents are decreased by 45-65% and 50-60%, respectively [124-128]. Rapid changes are

believed to be the result of reversible modifications made to the receptor subunits that affect

open time [129]. Over long term anoxia (3-21 days) NMDARs are removed from the cellular

membrane demonstrated through a 40% reduction in GluN1 subunit abundance and a depression

of Ca2+ permeability after 6 weeks [5]. It is unclear if a similar mechanism exists for AMPARs.

Rapid and long term changes in NMDARs and AMPARs are collectively termed “channel

arrest” and are utilized to conserve energy and prevent ECD [79].

Page 31: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

16

Figure 1.2: NMDAR and AMPAR structures.

A, schematic diagram of the NMDA and AMPAR receptor complexes. B, schematic diagram of

the NMDAR and AMPAR subunit arrangements. Figures adapted from online sources [130, 131]

1.9 Mechanisms controlling “channel arrest”

Initial decreases in NMDAR and AMPAR whole cell currents are triggered rapidly in

response to oxygen deprivation. The mechanisms controlling these changes are believed to be

signalled by changes associated with anoxia. The effects of anoxia mediated changes in

adenosine, ATP-sensitive K+ (KATP) channels and intracellular calcium have all been

investigated.

1.9.1 Increases in adenosine

A temporary but significant decrease (20%) in ATP concentrations occurs in the brain of

the western painted during transition into anoxia. This decrease lasts for approximately an hour

before metabolic processes are reduced sufficiently for energetic costs to be met by anaerobic

Page 32: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

17

glycolysis [34]. When ATP breakdown exceeds ATP production, AMP begins to accumulate in

the cell and 5’-nucleotidase activity increases. 5’-nucleotidases are a family of enzymes found in

the vertebrate CNS and inhibited by ATP. When activated they break down AMP into adenosine

[3, 132].

Adenosine is a “retaliatory metabolite” produced in response to oxidative stress in a

number of vertebrates [133]. Extracellular adenosine increases trigger protective changes

through interactions with adenosine receptors in different tissues (A1, A2A, A2B, and A3) [134].

Changes involved in neuroprotection include: suppression of pre-synaptic excitatory

neurotransmitter release (ex: glutmate), stimulation of anaerobic glycolysis, increased cerebral

blood flow and reduction of neuronal excitability via inhibition of Ca2+ channels/activation of G

protein-coupled inwardly-rectifying K+ channels [3, 134-138]. Neuronal extracellular adenosine

levels after 100 minutes of anoxia are 12 fold of normoxic concentrations [139]. This increase is

temporary and as metabolic processes are depressed in the turtle as discussed above, energy

needs are met by anaerobic glycolysis and normal ATP concentrations are re-established. After

300 minutes adenosine concentrations return to baseline levels [139].

Increased extracellular adenosine levels in the western painted turtle brain increase

resistance to K+ efflux in order to preserve cellular ion gradients. This is mediated by binding of

the A1 receptor [140]. Adenosine and A1 receptor activation has also has also been implicated as

a mediator of NMDAR channel arrest. Within neural tissue from the western painted turtle

normoxic adenosine addition reduces NMDA-mediated Ca2+ entry by 62 %. This decrease is

abolished by perfusion of the A1 antagonist 8-pentyltheophyilline (8-PT) [141]. However during

anoxia, A1 receptor antagonism does not block decreases in NMDAR open time or NMDA-

mediated Ca2+ entry [142]. This suggests that adenosine is capable of reducing NMDAR activity

but is not a necessary component of anoxia-mediated NMDAR channel arrest.

1.9.2 Activation of ATP-sensitive K+ (KATP) channels

Like adenosine production, KATP channel activity is controlled by ATP concentrations.

These channels, found in the plasma membrane and internal organelles like the mitochondria, are

gated by high concentrations of ATP and activated when levels are low like during oxidative

Page 33: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

18

stress. These channels are found within the brain of vertebrates, particularly in the hippocampus

[97]. KATP channels are comprised of pore-forming Kir subunits and ATP-binding sulfonyl urea

subunits (SUR) [143]. The configuration and combination of the subunits varies based on the

type of membrane they are found within. The plasmalemmal KATP (pKATP) channel is composed

of two Kir6.1 and Kir6.2 bound to two SUR1 and SUR2 [144]. The mitochondrial KATP (mKATP)

is also thought to be a heteromultimer consisting of mitoKir and mitoSUR and containing neither

Kir6.1 or Kir6.2 [143, 145].

The freshwater turtle (Trachemys scripta) is another anoxia tolerant turtle species able to

survive at least 48 h of anoxia at 25°C [146]. Within this species increased pKATP channel

activity is necessary during anoxia for the maintenance of cellular ion gradients. K+ efflux from

neurons is reduced by 50% within one hour of anoxia, but abolished with administration of the

KATP channel blocker glibenclamide [147]. Increases in mKATP channel activity work in concert

with adenosine to block glutamate release within the first 3 hours of anoxia. Addition of the KATP

channel blocker BDM or the adenosine receptor antagonist theophylline produced increase in

glutamate release of approximately 70% above anoxic levels. At 5 hours, adenosine inhibition

produces an approximate 100% increase in glutamate release but KATP channel blocking has no

effect [136].

In the western painted turtle, mitochondrial specific KATP (mKATP) channels are involved

in anoxia-mediated down regulation of NMDAR and AMPAR currents. Addition of the mKATP

channel activator diazoxide under normoxic conditions reduces both whole-cell AMPAR and

NMDAR currents to levels comparable to anoxia. Inversely mKATP blockade with 5-

hydroxydecanoate (5-HD) abolishes anoxia-mediated reduction of NMDAR and AMPAR

currents [125, 126]. Activation of mKATP channels is also associated with an uncoupling of turtle

mitochondria by 10–20% and reduced Ca2+ uptake into the mitochondria [126] [148]. The

resulting rise in intracellular Ca2+ levels can bring about downstream effects in NMDAR and

AMPAR activity.

Page 34: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

19

1.9.3 Increases in intracellular calcium

Following the onset of anoxia in turtle neural tissue, intracellular calcium ([Ca2+]i )

concentrations increase [126, 128, 149]. The [Ca2+]i increase is necessary for the development of

anoxia mediated AMPAR/NMDAR channel arrest. The inclusion of the Ca2+ chelator 1,2-bis(o-

aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA) in the pipette solutions of whole-cell

patch clamp recording electrodes abolishes anoxia-mediated reductions in AMPAR/NMDAR

currents [125, 129]. Increases in [Ca2+]i occur in the absence of extracellular calcium, indicating

that Ca2+ is released from an intracellular source [126, 128, 149]. Within neurons Ca2+ is stored

in the mitochondria and the endoplasmic reticulum (ER). Inhibition of ER specific calcium

release via ryanodine does not abolish anoxia mediated NMDAR current reductions, indicating

mitochondrial specific Ca2+ release is responsible for anoxia mediated changes [126]. As

discussed above activation of mKATP channels is associated with mitochondrial uncoupling and

reduced Ca2+ uptake. Activation of mKATP channels with diazoxide or levcromakalim produces

increases in [Ca2+]i and decreases in NMDAR/AMPAR currents of similar magnitude to those

seen during anoxia. The addition of the mKATP antagonists 5-HD or glibenclamide prevented

both of these changes [125, 126]. Increases in [Ca2+]i trigger the activation of a variety of

intracellular regulating proteins that can serve as the intermediate step in signalling

NMDAR/AMPAR current down regulation in response to mitochondrial Ca2+ release.

1.9.4 Enhancements of calmodulin and phosphatase activity

Calmodulin is a calcium-binding messenger protein expressed in all eukaryotic cells.

Calmodulin can modulate NMDAR directly by binding to the COOH terminus of the NMDAR

GluN1 subunit and inactivating the channel in a Ca2+-dependent manner [150]. Within the

western painted turtle inhibition of calmodulin with calmidazolium attenuates anoxia mediated

decreases of NMDAR currents within neural tissue [129]. Inhibition has yet to be evaluated on

anoxia mediated AMPAR current reductions.

NMDAR activity can also be reversibly modulated by intracellular kinases and protein

phosphatases (PPs), some of which are controlled by intracellular calcium concentrations.

Dephosphorylation via PP1, PP2A and the Ca2+/calmodulin-dependent phosphatase PP2B

Page 35: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

20

(calcineurin) all attenuate NMDAR activity [151-154]. Within the anoxic turtle brain, addition of

the PP1 inhibitor okadaic acid or the PP1/PP2A inhibitor calyculin A prevent anoxia-mediated

attenuation of NMDAR currents [129]. The inhibition of calcinuerin via cypermethrin similarly

attenuates NMDAR currents over the short term (20 minutes) but not the long term suggesting

that calcinuerin might only play a role in early attenuation [129]. The effect of phosphatases on

anoxia mediated AMPAR current reductions have yet to be tested.

Figure 1.3: Schematic illustration of anoxia-mediated changes contributing to channel arrest.

1.10 Reactive oxidative species (ROS)

Other cellular changes aside from decreases in intracellular ATP concentrations occur

during anoxia that may contribute to “channel arrest” and/or “spike arrest”. One such change is

reductions in reactive oxidative species (ROS). All reactive molecules and free radicals produced

as unwanted by-products from the incomplete reduction of diatomic oxygen (O2) during

oxidative metabolism are termed as ROS [155]. All aerobic organisms produce ROS and have

developed cellular anti-oxidant defence systems that prevent concentrations from reaching

harmful levels. Under normoxic conditions, intracellular ROS levels are maintained at relatively

Page 36: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

21

low concentrations but never completely removed. During oxygen deprivation in anoxia tolerant

organisms, oxidative phosphorylation halts and ROS levels decline to immeasurable levels [155].

Within the brain of the western painted turtle CM-H2DCFDA fluorescence evaluations have

demonstrated that ROS concentrations decrease by 25% within 10 minutes of anoxia [156].

ROS are primarily produced within the mitochondria between complexes and I and III

where 2-3% of all O2 consumed is left partially reduced as the highly reactive superoxide anion

(O2) [Figure 1.3] [157, 158]. This molecule is short lived and in most cases binds with water

either spontaneously or through induction by mitochondrial superoxide dismutase (SOD) to

produce hydrogen peroxide (H2O2) [159-161]. H2O2 is one of the longest lived cellular ROS but

is eventually converted to water and O2 by catalase or glutathione peroxidase (GPx) [93, 162].

Levels of H2O2 are directly linked to metabolic activities and increases or decreases can serve to

signal cellular functions such as apoptosis, gene expression, and the activation of cell signaling

cascades via reversible oxidation of cysteine (Cys) thiol residues on signalling proteins and

membrane ion channels [163-167]. H2O2 is cell permeable and capable of moving through the

lipid bilayer by free diffusion or through aquaporins (AQP)/water channels and oxidizing

extracellular as well as intracellular Cys residues. In this manner, H2O2 acts as an inter and intra

cellular messenger [168-171]. The oxidized Cys residues can be reduced by elements of the

antioxidant defence system or can react with other sulfide compounds to produce disulfide

bridges [Figure 1.3] [165, 168-171].

Although there remains controversy over whether CNS neurons contain aquaporins

neurons do exhibit low water permeability under resting conditions and are capable of moving

water through alternative means such as membrane cotransporters (ex: KCC1) [172]. H2O2 is

very water soluble and should be capable of following the movement of water through available

channels similar to movement through APQs [168, 173, 174]. For the purposes of this thesis, the

routes for facilitated water transport (aquaporins, cotransporters, or other) will be grouped and

designated as water transport routes (WTRs) for the sake of simplicity.

Another source of cellular ROS is nitric oxide (NO) produced by neuronal NO synthase

(nNOS). nNOS is tethered to the NMDAR by the postsynaptic density protein-95 (PSD95) [175,

176]. nNOS produces NO from arginine and O2 and is triggered by the activated Ca2+-binding

Page 37: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

22

protein calmodulin (CaM) [Figure 1.3] [177-179]. Tethering of nNOS to PSD-95 and Ca2+

dependent activation couple the production of NO to NMDAR activation. Like H2O2 NO is short

lived, capable of moving through the lipid bilayer by free diffusion or through WTRs, and can

reversibly oxidize cysteine residues through a processes termed S-nitrosylation. NO can also

react with O2 and form peroxynitrite (ONOO) that is another effective oxidizing agent. It can

also activate soluble guanylate cyclase (sGC) and produce cyclic guanosine monophosphate

(cGMP) capable of triggering downstream changes that include further ROS generation [168,

179-182]. [Figure 1.3].

1.11 The role of ROS in anoxia-intolerant vertebrates

Under normoxic conditions, the role of ROS in cellular signalling has been well

documented in a number of anoxia-intolerant organisms. Changes in ROS increase and decrease

the activities of a number of ion channels and intracellular proteins [183, 184]. Changes in the

NMDAR redox state and intracellular ROS levels affect NMDAR activity. Like anoxia-mediated

regulation, these changes appear to be affected by the phosphorylation state of the NMDAR.

1.11.1 Redox changes affect NMDAR but not AMPAR activity

The structure and function of several membrane bound receptors containing Cys residues

are sensitive to redox conditions [185]. Specific Cys residues in the NMDAR form redox

modulatory sites whose oxidation/reduction state affects the activity of the receptor. Redox

modulatory sites have been demonstrated to exist within GluN1, GluN2A and GluN2B subunits

[186-188]. Experiments using site-directed mutagenesis in rat and frog oocytes have identified

residues within the GluN1 (Cys744, Cys798, Cys 79 and Cys 308) and GluN2A (Cys 87, Cys

320 and Cys 399) subunits whose alteration affects redox modulation but the site in the GluN2B

has yet to be localized. All of the sites identified have been localized to extracellular regions of

the receptor subunits within their respective species. However, there is some controversy as to

whether intracellular or transmembrane redox sites also exist but have yet to be identified [185,

187-189].

Page 38: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

23

The effect of redox modulation on NMDA receptors has been observed in a wide variety

of tissue preparations including intact chick retina, frog oocytes, rat retinal ganglion,

hippocampal, cortical and striatal slices and post-mortem human cortex [188, 190-198].

Evaluations of NMDAR activity within these preparations using methods including intracellular,

extracellular, and patch-clamp recording have demonstrated that addition of oxidizing agents

(DTNB and GSSG) decrease receptor activity while reducing agents (DTT) increase receptor

activity. The increases produced through reduction are greater than decreases through oxidation

(6-11 times), suggesting that redox sites are maintained in a more oxidized state [190] [196,

199].

Unlike the NMDAR, no modulatory redox sites have been found in the structures of the

AMPAR GluR subunits. Within many of the same tissue preparations used to demonstrate the

effects of NMDAR redox modulation, AMPA addition has been shown to be unaffected by

oxidizing or reducing agents [190, 196-198].

1.11.2 Alterations in ROS affect NMDAR activity

Like reducing/oxidizing agents, changes in ROS have been shown to alter NMDAR

activity. Within rat cortical neurons oxygen free radicals generated by xanthine and xanthine

oxidase (X/XO) are capable of reversing increases in NMDAR activity brought on by reducing

agents and decreasing activity similar to oxidizing agents [191, 196]. The inhibition of

mitochondrial ROS production has also been demonstrated to effect NMDAR activity. Rotenone

is a complex I inhibitor and decreases mitochondrial ROS production in mitochondria from rat

brain, heart, liver, dog brain and human cell cultures [157, 200-202]. This is accomplished

through inhibition of electron transfer from iron-sulfur centers in complex I to ubiquinone

[Figure 1.3]. In whole cell patches of dopamine neurons from slices of rat midbrain the addition

of rotenone increases the average amplitude of evoked NMDAR currents by 162% without

affecting AMPAR activity [203].

The effect of NO manipulation has also been evaluated in cultured rat cortical neurons

and frog oocytes through Ca2+ imaging and whole cell patch clamp recordings. Like oxidizing

Page 39: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

24

agents and ROS generators, the application of NO generators produced marked decreases in

NMDAR activity [189, 204, 205].

1.11.3 NMDAR redox regulation is blocked by kinase inhibition

As discussed above, increased phosphatase activity is associated with decreases in

NMDAR activity during anoxia in the turtle brain [151-154]. There is some evidence to suggest

that phosphorylation of the NMDAR is necessary for oxidation/reduction of the redox

modulatory site. Electrophysiological recordings in hippocampal rat slices demonstrated

increases in NMDAR-mediated synaptic responses with DTT application. Increases were

blocked by inhibition of Ca2+/calmodulin-dependent protein kinase II but not by inhibition of the

protein phosphatases PP1 or calcineurin [196]. Similarly in horizontal slices from rat ventral

midbrain rotenone augmentation of N-methyl-D-aspartate (NMDA)-evoked currents were

inhibited by the inclusion of a tyrosine-kinase in intracellular pipettes [206]. These findings may

indicate that the NMDAR redox site is only accessible when the NMDAR is phosphorylated.

Page 40: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

25

Figure 1.4: The mechanisms of ROS production and cysteine oxidation.

A, depiction of the mitochondrial electron transport chain showing the sites of reactive oxygen

species production and rotenone inhibition. B, depiction of NMDAR coupled nNOS activation

and nitric oxide synthesis. C, depiction of the effect of H2O2 and NO oxidation on cysteine

residues. Figures adapted from (A) Pamenter 2007 and (B) Nakamura 2007 [156, 179].

Page 41: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

26

1.11.4 mKATP mediated ROS increases affect ischemic preconditioning

Changes in ROS have recently been linked to the development of protection produced

through ischemic preconditioning. Like anoxia mediated NMDAR/AMPAR downregulation in

the brain of the western painted turtle, activation of the mKATP channel has been identified to be

involved in ischemic preconditioning. Within cardiac tissue from rats and rabbits the addition of

the selective mKATP antagonist 5-HD prevents the development of protective effects from

ischemic preconditioning [207-209]. In neural tissue from rats and pigs the addition of the

mKATP agonist diazoxide protects the brain against ischemic and anoxic stresses similar to

ischemic preconditioning [210-213].

Activation of mKATP channels in the western painted turtle is believed to trigger a mild

increase in intracellular calcium necessary for the development of downstream changes. In

ischemic preconditioning however the activation of mKATP channels and downstream changes

are associated with increased production of mitochondrial ROS. In rat hippocampal slices,

isolated rabbit hearts, and human atrial-derived cell lines the addition of the ROS scavenger N-2-

mercaptopropionylglycine (MPG) reversed diazoxide mediated protection similar to 5-HD

addition [214-216]. Fluorescent investigation in human atrial derived cell line, rabbit myocardial

tissue, and rat retinal tissue has demonstrated that mKATP channel activation increases oxidation

of the detective probes indicative of increases in cellular ROS levels [216-218]. This seems to

indicate that mKATP channel opening increases ROS generation that then triggers the

development of the preconditioned state.

There is also some evidence to suggest that mKATP channels are more favourably

activated in the presence of mitochondrial ROS or NO. Within rat myocytes, reconstituted

bovine ventricular myocardium mitochondria, embryonic chick ventricular myocytes, rabbit

ventricular myocytes, rat dorsal root ganglion neurons, and rat cardiac submitochondrial particles

the addition of NO donators, O2•- generators, ROS scavengers and exogenous H2O2 have all

been demonstrated to affect mKATP activity [219-224]. Increased ROS caused mKATP channels to

become less inhibited by ATP and more readily activated. This increase has been attributed to

Page 42: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

27

increased oxidation of cysteine residues in the SUR subunit, similar to what occurs in the

NMDAR redox site [223]. In this way, mKATP activation during ischemic precondition may

cause channel oxidation and promote further activity in a positive feedback loop [221].

1.12 The antioxidant defence system

Antioxidants are substances that when present at low concentrations, compared to that of

an oxidizable substrate, significantly delays or inhibits oxidation [225]. The antioxidant defence

system protects the cell from unwanted reactions by maintaining levels of ROS generated

through processes like nNOS activation and mitochondrial oxidative phosphorylation at low

concentrations [226].

The three intracellular antioxidants most relevant to nNOS and mitochondrial ROS

generation are superoxide dismutase (SOD), catalase and glutathione (GSH)/glutathione

peroxidase (GPx). SOD is found within the mitochondria of all eukaryotes and differentiated

from SOD found in chloroplasts and prokaryotic organisms by the type of metal cofactor

(MnSOD) [227]. SOD catalyses the dismutation of O2 , produced from oxidative

phosphorylation, into H2O2 at a reaction rate that is 10,000-fold faster rate than spontaneous

dismutation (O2 + O2

+ 2H+ => 2H2O2 + O2) [228]. SODs are nuclear encoded and there is

some evidence to suggest that synthesis is sensitive to environmental stressors and can be altered

in response to changes in ROS production [227]. Intracellular concentrations of H2O2 are

managed through a wide range of enzymes, including catalase and GPx. Catalase forms a

intermediate complex with H2O2 and generates water and O2 (2H2O2 => O2 + 2H2O). GPx

catalyzes the reaction of GSH with H2O2 to produce oxidized glutathione (GSSG) the helps to

maintain a redox balance in the cellular compartments (2GSH + H2O2 => GSSG + 2H2O). A

central nucleophilic cysteine residue is responsible for the high reductive potential of GSH,

allowing it to scavenge cytotoxic H2O2 and reacts non-enzymatically with other ROS like

singlet oxygen, superoxide radicals, hydroxyl radicals, and NO [226, 227]. The cumulative

actions of these and other antioxidants maintain the intracellular environment within a reduced

state [168].

Page 43: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

28

As discussed above both H2O2 and NO are capable of leaving the cell through free

diffusion or WTRs and interacting in the extracellular environment. The resulting extracellular

ROS concentrations are managed by antioxidants produced within the cell and transported to the

extracellular environment. GSH, catalase and GPx act in the extracellular environment but

generally exist at lower concentrations than found within the cell [229]. The major extracellular

thiol is free L-Cys that exists in both a reduced (Cys) and oxidized (Cyss) form, although in the

plasma the oxidized form is predominant. Influx and efflux of reduced Cys between the

intracellular environment may contribute to extracellular redox status and help to maintain a

Cys/Cyss balance [168]. Due to lower antioxidant concentrations and the movement of ROS out

of the cell, the extracellular environment exists within a predominantly oxidized state [168].

Antioxidants in both the extracellular and intracellular environments can act to reverse

ROS reactions, such as by reducing thiol residues in proteins oxidized through ROS interactions.

Aside from scavenging roles, GSH and free L-Cys have been implicated to act as the primary

intracellular and extracellular protein reductants, respectively [230]. Intracellular and

extracellular levels are maintained relatively constant in most cells and act to establish a baseline

balance with ROS products between oxidation and reduction states of cellular proteins [168,

230].. During anoxia in the western painted turtle when ROS concentrations fall the balance can

be disturbed leading to a shift towards protein reduction in both the intracellular and extracellular

environment.

1.13 ROS regulation of GABA in anoxic turtle cortex

As previously discussed one of the mechanisms through which turtles reduce ATP

consumption within the brain during the over wintering period is through increases in GABA

receptor postsynaptic currents (PSCs) that serve to decrease whole brain activity via spike arrest

[113, 114]. Recent investigation within our lab has demonstrated that in cortical slices from the

western painted turtle under normoxic conditions, ROS scavenging increased GABAergic PSC

amplitudes similar to anoxic exposure. Application of the mitochondrial complex IV inhibitor

cyanide decreased intracellular ROS to the same extent as anoxia and also resulted in a similar

increase in GABAergic PSCs. Finally GABAergic PSC increases from ROS scavenging and

Page 44: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

29

anoxia were both reversed by exogenous application of H2O2 [231]. These findings demonstrate

that anoxic decreases in mitochondrial ROS production signal increases in GABA release.

1.14 Rationale for study

Decreases in reactive oxidative species following entrance into anoxia within the western

painted turtle have been documented [156] [231]. Pharmacologically scavenging ROS or

inhibiting mitochondrial ROS synthesis under normoxic conditions increases GABAergic PSCs

similar to anoxia [231]. Theses finding demonstrate that ROS decreases play a role in signalling

anoxia-mediated changes. Aside from increases in GABA release, during anoxia neuroprotective

decreases in NMDARs and AMPARs currents also occur and are associated with mild increases

in intracellular calcium. The primary aim of this study was to determine whether

pharmacological reduction of cellular ROS under normoxic conditions produced decreases in

whole-cell evoked NMDAR/AMPAR currents similar to those seen during transition from

normoxia to anoxia. The secondary aim was to evaluate whether the mild increase in Ca2+

associated with receptor down regulation is also affected by changes in ROS.

Within anoxia-intolerant vertebrates decreases in ROS and reduction of the NMDAR

redox site are associated with increased NMDAR activity. Because the western painted turtle

experiences decreases in ROS during entrance into anoxia, you would expect to see a similar

increase in activity if a similar mechanism were present. Instead NMDAR activity is

downregulated suggesting that either the structure of the NMDARs in the turtle brain are

different from those seen in other documented vertebrates and respond to ROS changes in an

opposing manner or an anoxia mediated mechanism is present that overrides and reverses normal

redox modulation. The mechanism will also need to down regulate AMPAR activity, that is

documented to be unaffected by ROS modulation.

Based on the material discussed above on ion channel control during normoxia and

anoxia, there are a couple of viable routes through which anoxia-mediated channel arrest could

be accomplished. Redox control could be overridden through increases in the activity of cellular

phosphatases. As discussed above, there is evidence that NMDARs may need to be

phosphorylated in order for redox modulation to take place. Increases in phosphatase activity

Page 45: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

30

suggested to occur during anoxia in the western painted turtle may be sufficient to override and

reverse redox modulation. The increases in phosphatase activity may be also triggered by ROS

changes. As discussed above the activity of the mKATP channel can be affected by its

oxidation/reduction state and in anoxia-intolerant organisms is preferentially activated in the

presence of ROS. The mKATP subunit that exists in the turtle brain may be modified such that it

reacts to decreases in ROS/ increased reduction by becoming more easily activated and

triggering anoxia-mediated increases in intracellular calcium and phosphatase activity. Based

upon this reasoning I propose the following:

Hypothesis: Decrease in reactive oxidative species during anoxia triggers a down regulation of

NMDAR and AMPAR whole cell currents via an increase in the activity of mKATP and a

subsequent increase in intracellular calcium.

Aims:

1. Pharmacological reduction of cellular ROS will reduce normoxic NMDAR/AMPAR currents

and addition of H2O2 under anoxia conditions will increase NMDAR/AMPAR currents.

Using whole-cell patch-clamp techniques NMDAR/AMPAR whole cell currents will be

measured following pharmacological manipulation of ROS under normoxic and anoxic

conditions.

2. Decreases in ROS will trigger increases in cytosolic calcium levels comparable to those

measured during anoxia.

The fluorescent Ca2+ indicator dye, Oregon Green, will be used to measure changes in cytosolic

calcium levels following anoxic exposure and/or pharmacological manipulation of ROS.

Page 46: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

31

CHAPTER 2: MATERIALS AND METHODS

2.1 Animals

This study was approved by the University of Toronto Animal Care committee and

conforms to the relevant guidelines for the care and handling of experimental animals as outlined

in the Guide to the care and Use of Experimental animals as determined by the Canadian Council

on Animal Care.

Adult male and female western painted turtles were purchased from Niles Biological Inc.

(Sacramento, CA, USA). Animals were housed in an indoor aquarium equipped with a flow-

through de-chlorinated freshwater system, a heating lamp, and a basking platform. The water

temperature was maintained at 20°C. The turtles were given continuous access to food, and were

kept on a 12h:12h light-dark photoperiod.

2.2 Dissection and whole-cell patch-clamp recording protocol

Turtles were selected at random and decapitated. Whole brains were rapidly excised from

the cranium of western painted turtles and cortical sheets were isolated and placed in artificial

cerebral spinal fluid (aCSF; mm: 107 NaCl, 2.6 KCl, 1.2 CaCl2, 1 MgCl2, 2 NaH2PO4, 26.5

NaHCO3, 10 glucose, 5 imidazole, pH 7.4; osmolality 280–290 mosmol l−1). Individual cortical

sheets (3-4) were cut medially from the visual cortex of each cerebral hemisphere [Figure 2.1].

Page 47: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

32

Cortical sheets were placed in an RC-26 chamber with a P1 platform (Warner

Instruments, CT, USA). During recordings of NMDAR activity, cortical brain tissues were

bathed in standard aCSF. During recordings of AMPAR activity, cortical tissues were bathed in a

modified high Mg2+ aCSF solution [4 MgCl2] in order to block NMDAR activity, and isolate

AMPAR currents specifically.

aCSF solutions were gravity perfused into the chamber via 1-L glass bottles attached to

intravenous (IV) drippers. A three way valve allowed for transitions to be made between initial

and experimental bottles during recordings. Conditions of normoxia were developed by bubbling

with 95% O2/5% CO2. Investigations performed under anoxic conditions were accomplished by

switching to a secondary bottle of aCSF bubbled with 95% N2/5% CO2. The perfusion tubing

was double jacketed and gassed with 95% N2/5% CO2 to maintain anoxic conditions. As well, a

plastic rim with anoxia tubing connected was placed around the saline bath to gently gas 95%

N2/5% CO2 during anoxic conditions. All experiments were conducted at room temperature (22

°C).

Whole-cell recordings were performed using 5-8 MΩ micropipettes produced from

borosilicate glass capillary tubes with a P-97 micropipette puller model (Sutter Instruments).

Micropipettes were fire-polished using a MF-830 Microforge (Narashige Group). Pipette

solution contained the following (in mM): 8 NaCl, 0.0001 CaCl2, 10 Na-HEPES, 130 potassium

gluconate, 1 MgCl2, 0.3 NaGTP, and 2 NaATP (adjusted to pH 7.4 and osmolarity 290-300

mOsM). Cell-attached 1-20 GΩ seals were obtained using the blind-patch technique of Blanton

et al. (1989). To break into the cell, the recording electrode potential was voltage-clamped to -80

mV and a sharp pulse of negative pressure was applied. Once the whole-cell configuration was

Page 48: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

33

established, cells were given at least 2 minutes to acclimate before series resistance was

measured, which normally ranged from 20-100MΩ. Patches were discarded if series resistance

varied by >25% over the course of an experiment. Data was collected at 5-10 kHz using an

Axopatch-1D amplifier, a CV-4 head stage, and a Digidata 1200 interface (Axon Instruments)

and stored on computer using Clampex 10 software (Axon Instruments). A fast-step perfusion

system (VC-6 perfusion valve controller and SF-77B fast-step perfusion system; Warner

Instruments) was used to deliver pharmacological agents to the tissue.

Figure 2.1: Diagrammatic representation of cortical sheet excision and experimental setup.

Figure adapted from an online source [232].

Page 49: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

34

2.3 Neuron identification

All investigations were carried out within pyramidal cells. Neurons were identified as

pyramidal or stellate based on the expressed electrophysiological properties. Previous to all

whole cell patch clamp experiments cells were current clamped and a current was injected for

450 ms to cause a 30 mV depolarizing change in membrane potential and action potential firing.

Characterization was accomplished by examining the adaptation pattern and amplitudes of

action-potential firing and comparing these to the standards previously developed for

differentiating between stellate and pyramidal neurons [101].

2.4 Evoked NMDAR and AMPAR current amplitude recordings

After patches stabilized neurons were voltage-clamped to a holding potential of -80 mV

for the entirety of the recording period. All recordings were carried out within the same time

frame in order to ensure comparability. Recordings were made at 0 min, 10 min, 30 min, 50 min,

70 min, and 90 min [Figure 2.2]. Access resistance was measured at the beginning and end of

each recording and if resistance fluctuated by >20% from the initial value patches were

discarded. Initial recordings at 0 and 10 were made under normoxic conditions to establish a

baseline for comparison and confirm currents were stable. The initial amplitude recording at t= 0

min was set to 100% and all subsequent recordings were normalized to that control value for

comparison purposes. Patches were discarded if t=10 min varied by more than 20% from t= 0

min. Experimental conditions were induced immediately after the establishment of a stable

baseline and currents were evaluated after 20 and 40 minutes. Normoxic conditions were then

reintroduced in order to reverse experimental changes and evaluated after 20 and 40 minutes.

Page 50: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

35

The peak current values were taken from the recordings and used for the purposes of comparison

and statistical analysis.

Tetrodotoxin (TTX) was administered via drip perfusion 5 minutes previous to all

recordings in order to prevent the opening of voltage gated sodium channels and ensure currents

elicited were only from NMDAR/AMPAR activation. The only exception was during H2O2

experiments where 5 minute TTX addition was carried out 10 minutes prior to all recordings.

This allowed for H2O2 to be drip perfused 5 minutes prior to recordings to optimize its effects.

The effects of TTX have been shown to extend beyond 15 minutes ensuring that

NMDAR/AMPAR currents were isolated throughout the extended recording period. After TTX

administration cortical sheets were drip perfused with NMDA or AMPA for 3-10 seconds to

induce a current response. The same application time was then used for every subsequent

recording performed on the same neuron within a single experiment.

Figure 2.2: NMDAR and AMPAR whole-cell patch clamp recording timeline.

NMDAR and AMPAR current amplitude measurements were all taken at specified time points

(top line) during varying treatments (bottom).

Page 51: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

36

2.5 Fluorescence investigation of cytosolic Ca2+ changes

To assess changes in cytosolic Ca2+ cortical slices were loaded with the Ca2+-sensitive

dye Oregon-Green 488 BAPTA-1 AM (Invitrogen). Oregon-Green 488 BAPTA-1 was chosen

due to its high Ca2+ affinity (Kd ≈170 nM). Tissue sheets were incubated individually in an

opaque vial containing 2 mL of aCSF with 40 uM Oregon-Green (from a 1 mM Oregon-Green

stock solution in dimethylsulfoxide and 20% pluronic acid). Cortical slices were loaded within

the vials for 3 hours at 3-5 °C and subsequently washed in unmodified aCSF for 15 minutes.

Following dye loading slices were placed in a flow-through recording chamber as previously

described for whole-cell patch clamp experiments. During anoxic experiments a plastic cuff was

placed around the objective in order to gently gas 95% N2/5% CO2 across the bath.

Oregon-Green was excited at 488 nm using a DeltaRam X high-speed random access

monochromator and a LPS220B light source (Photon Technology International) at a bandwidth

of 4.5 nm. Fluorescence measurements were acquired at 1 second intervals using an Olympus

BX51W1 microscope and U-CMAD3 camera. Fluorescence emission measurements at 488 nm

were acquired at 10 second intervals under 400x magnification using an Olympus BX51W1

microscope (Olympus Canada Inc.) and Rolera-MGi Digital EMCCD camera (QImaging).

Camera gain and exposure time values were 4.0 and 3 s, respectively. Previous to all recordings

excitation scans were performed from 450 to 550 nm in order to confirm peak excitation at 488

nm and successful cellular loading of Oregon Green.

Fluorescent recordings were all carried out within the same time frame to ensure

comparability. The time frame was reduced compared to that utilized within whole cell patch

Page 52: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

37

clamping experiments to compensate for the rapid rate of photo bleaching and ensure that the

fluorescence signal was not exhausted. Cells were recorded under normoxic conditions for 10

minutes to obtain a stable baseline, followed by a treatment period for a further 10 minutes, and

concluded with normoxic reperfusion for a final 10 minutes [Figure 2.3]. All fluorescent

experiments were conducted with bulk perfusion as drip perfusion proved unreliable and affected

image stability. Solutions were gas bubbled in the same manner as solutions used for whole cell

patching. After the reperfusion period, aCSF flow was halted and tissues were incubated with the

Ca2+ ionophore ionomycin (2 μmol) for 5 minutes to increase extracellular Ca2+ uptake and

obtain the maximal fluorescence signal (Fmax). This was followed by addition of MnCl2 (2 mM)

to preferentially bind the Oregon green dye and quench fluorescence so that the minimal

fluorescence signal could be obtained (Fmin). The protocol and concentrations used were based

on previous investigations [149, 233].

A minimum of 10 neurons were selected for analysis in each experimental recording

period. In order to ensure good dye uptake and cellular health neurons exhibiting low to

moderate basal calcium fluorescence were chosen. The change in fluorescence was determined

from a line of slope drawn over the treatment area and compared against a parallel line of slope

across the pre and post treatment area (resting fluorescence signal) [see Figure 3.7 for

illustration]. The difference between these lines at the midpoint of the experimental period (F)

was taken and divided by the established range of fluorescence (Fmax-Fmin) to establish the

change in Ca2+ fluorescence. Expressed as the function: Ca2+ Change= (F/(Fmax-Fmin)) x 100.

The percent change value for all neurons selected within a trace was averaged and used as a

single n-value for final statistical analysis.

Page 53: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

38

Figure 2.3: Oregon Green fluorescent recording timeline.

Fluorescent recordings were all carried out within the timeframe with the treatment applications

(bottom) all at the same time points (top).

2.6 Whole cell spontaneous neuronal activity recordings

Spontaneously firing pyramidal neurons were utilized to assess changes in neuronal

activity. Whole cell patches were obtained using the same procedure as described above.

Neurons displaying spontaneous activity were confirmed to be pyramidal cells in the same

manner as used in evoked NMDAR/AMPAR recordings. Following successful identification

neurons were recorded over a 40 minute time frame. Cells were recorded under normoxic

conditions for 5 minutes to establish a measurement of baseline activity, followed by a treatment

period for 20 minutes, and concluded with normoxic reperfusion for a final 15 minutes [Figure

2.4]. All experimental solutions were administered via bulk perfusion. AP frequency (APf) was

evaluated for 2 minutes after the cell was allowed to stabilized for 1 minute (t=1-3 min). This

value was set to 1 and all subsequent values were normalized to the value for comparison

purposes. APf values for pre-treatment/normoxia (t=3-5 min), treatment (t=23-25 min), and

washout/recovery periods (t=38-40 min) were evaluated within the last 2 minutes of each

treatment period.

Page 54: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

39

Figure 2.4: Whole cell spontaneous activity recording timeline.

Spontaneous activity recordings were all carried out within the timeframe with the treatment

applications (red) all at the same time points (black). APf was evaluated for 2 min within the

various treatment periods (blue).

2.7 Pharmacology and drug application

During NMDAR/AMPAR current measurements drugs were applied either by drip perfusion

alone or in conjunction with bulk perfusion. Drip perfusion was utilized for agents whose mode

of action required little exposure time or whose extended presence could prove toxic to the cell.

1 uM TTX, 300 uM NMDA and 50 uM AMPA were concentrations based on previous

publications within our laboratory and were all exclusively drip applied as all agents could be

toxic during longer exposures [125-127, 234]. 50 uM H2O2 represents the primary mitochondrial

ROS product and was used to test the effects of increased ROS presence on receptor activity.

Concentrations were identical to those used in previous experiments with turtle cortical neurons,

where 50 uM was the lowest concentration in which a significant change was detected in CM-

H2DCF ROS fluorescence [231]. H2O2 was drip perfused for 5 minutes only in order to try and

Page 55: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

40

limit excessive reactions. Drip solutions were appropriately bubbled with 95% O2/5% CO2 or

95% N2/5% CO2 to ensure they were normoxic/anoxic.

Drugs were bulk perfused in the same manner as the normoxic and anoxic solutions for

both electrophysiological and fluorescence experiments. Separate 1 litre IV bottles were used

and filled with the respective drug solutions and transition between conditions was made using a

three way valve. All drug agents were bubbled with 95% O2/5% CO2 in order to ensure they

were normoxic. Cell permeable ROS scavengers N-2-mercaptopropionylglycine (MPG, 0.5 mM)

and n-acetylcysteine (NAC, 0.5 mM) were used to artificially decrease normoxic ROS and test

the effects of whole cell ROS decreases on NMDAR/AMPAR currents. Concentrations were

identical to those used in previous experiments with turtle cortical neurons where 30 minutes of

0.5 mM MPG/NAC decreased CM-H2DCF ROS fluorescence comparable to what was

documented for anoxia within the same time frame [231]. Rotenone (25 uM) was utilized to

inhibit mitochondrial complex I, prevent mitochondrial specific ROS production and determine

the role of mitochondrial ROS in NMDAR/AMPAR control. A rotenone concentration of 25 uM

was previously validated as safe in turtle cortical slices [156].

2.8 Measurement of oxygen tension

Oxygen concentrations within the recording chamber during induced anoxic periods were

tested using an E101 oxygen electrode (Cameron Instruments Co.) containing 1M KCl and 20

μM sodium azide connected through a Model 1900 polarographic amplifier (A-M Systems) to a

LabPro interface (Vernier Software and Technology) for computer data collection. Logger Pro

3.8.4.2 was used for data collection at a sampling rate of 2 seconds. Oxygen tension was

Page 56: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

41

calibrated to approximately 160 mmHg by placing the electrode in a glass bottle bubbled with

21% O2/79% N2 and to 0 mmHg in a glass bottle bubbled with 95% N2/5% CO2. The electrode

was submerged in aCSF within a chamber identical to that used during fluorescent and whole-

cell patch-clamp recordings.

2.9 Chemicals

Tetrodotoxin was purchased from Tocris Bioscience (Ellisville, MO, USA) and Oregon

Green 488 was purchased from Invitrogen Life Technologies (Burlington, ON, Canada). All

other chemicals were obtained from Sigma-Aldrich Chemical Co. (Oakville, ON, Canada).

2.10 Statistical analysis

Electrophysiological and fluorescent data was analyzed using SigmaPlot software

package version 11.0 (Systat Software, Inc., San Jose, CA). NMDAR/AMPAR whole-cell peak

current data and Oregon-Green fluorescence data was analyzed following division by a factor of

1000 and root arcsine transformation with a one-way RM ANOVA (Tukey test) to compare the

means of normoxic controls and treatments within treatment groups. Results are expressed as

means and standard error of the means (S.E.M.). Significance was determined at p < 0.05.

Page 57: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

42

CHAPTER 3: RESULTS

3.1 Pre-current recording evaluations

3.1.1 Anoxic conditions are produced within the bathing chamber

To confirm the recording chamber was anoxic during perfusion with N2/CO2 - bubbled

saline, an O2 electrode was used to measure the change in the partial pressure of oxygen during

the transition from normoxic to anoxic conditions. The solution was bubbled with room air at an

approximate partial pressure of 160 (~21% O2) mmHg, and after transition to aCSF bubbled

with 95% N2/5% CO2 the pressure fell to 0.6 mmHg (~0% O2) after 10 minutes [Figure 3.1].

This value was at the limits of detection of the O2 electrode and was not different from the level

measured in the N2/CO2 - bubbled bottle.

Page 58: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

43

Figure 3.1: Change in oxygen tension during normoxic to anoxic transition.

Sample trace representing oxygen tension over the course of 16 minutes in the recording

chamber. The switch to anoxic conditions was made at t = 5 min and the return to normoxia was

made at t= 11 min. Complete anoxic conditions were met after approximately 10 minutes.

3.1.2 All recordings were conducted within pyramidal cells

Pyramidal neurons were ideal for this investigation due to their abundance within the

turtle cortex and their use of glutamate as their primary excitatory neurotransmitter. Previous

publications within the turtle cortex have primarily conducted experiments within these cell

types. To confirm that all neurons tested were pyramidal cells the adaptation pattern and

amplitudes of action-potential firing of all cells were tested previous to NMDAR/AMPAR

current recordings. In order to generate the necessary electrophysiological traces for comparison

cells were current clamped and then current injected for 450 ms to generate a 30 mV

depolarizing change in membrane potential and action potential firing. Cells that displayed

action potentials exhibiting marked frequency adaptation during a sustained injection of current

Page 59: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

44

about firing threshold, as well as firing two discrete sizes of action potential were classified as

pyramidal cells. Cells that exhibited no spike frequency adaptation, uniform spike amplitudes,

and a short but large undershoot of membrane potential following action potentials were

classified as stellate cells. The difference between cell types was distinct when traces were

compared [Figure 3.2].

Figure 3.2: Spike frequency adaptation in pyramidal and stellate neurons.

A) Response of a representative pyramidal neuron to a suprathreshold pulse of current. B)

Response of a representative stellate neuron to a suprathreshold pulse of current.

3.2 Whole-cell NMDAR current experiments

3.2.1 Normoxic NMDAR currents are stable throughout the time course

The 90 minute experimental time frame utilized for all NMDAR pharmacological

experiments was based on previous experiments by our laboratory within the turtle cortex. To

confirm that currents remained constant and changes brought on by pharmacological

Page 60: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

45

manipulation were not artifacts, normoxic NMDAR current controls were conducted for the

entirety of the experimental time course. To ensure recordings were stable throughout the time

course, cells that experienced a greater than 20mV change in membrane potential from t=0 min

were discarded. Normalized NMDAR peak currents did not change after 10, 30, 50, 70 and 90

minutes of normoxic saline perfusion (93.83 ± 8.00% [n=8], 97.55 ± 9.40% [n=8], 98.77 ±8.56

[n=8], 95.52 ± 9.67% [n=8] and 98.16 ± 10.23% [n=4], respectively) [Figure 3.3].

3.2.2 Normoxic NMDAR current amplitudes are reduced during anoxia

NMDAR whole cell currents in turtle cortical pyramidal neurons undergo a large

approximately 50% reduction in amplitude during anoxia [127, 129, 149]. In order to confirm

that these decreases could be replicated and generate comparisons for pharmacological

manipulations anoxic conditions were induced and evaluated. Following 20 and 40 minutes of

anoxia NMDAR peak current amplitudes were reduced comparable to what has been previously

documented (61.93 ± 8.57% [n=7, p= 0.004] and 58.58 ± 8.97% [n=7, p= 0.008], respectively).

The effects were mostly reversed after 20 minutes of normoxic reperfusion (82.72 ± 12.16%

[n=6]) and completely reversed after 40 minutes of normoxic reperfusion (98.17 ± 4.21% [n=5])

[Figure 3.3].

Page 61: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

46

Figure 3.3: Effect of anoxia on whole cell NMDAR currents.

A, Percentage normalized NMDAR peak current amplitudes after 20 min (t=30, blue bars) and

40 min (t=50, red bars) of treatment followed by 20 min (t=70, green bars) and 40 min (t= 90,

purple bars) of washout. Dashed lines represent normoxic baseline controls taken at t=10 min

(red) and 40 min anoxic treatment value (blue). Data are expressed as means ± S.E.M. Asterisks

(*) indicate data significantly different from the paired normoxic control (p < 0.05). B-C, Paired

sample NMDAR currents of the initial baseline recording (t= 10 min) and following 20 min of

the indicated experimental treatment (t= 30 min).

Page 62: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

47

3.2.3 NMDAR current amplitudes are increased by ROS scavenging

In order to test the hypothesis that decreases in intracellular ROS concentrations ([ROS]i)

during anoxia serve to trigger decreases in whole cell NMDAR currents, the ROS scavengers

MPG and NAC were administered separately under normoxic conditions. Following 20 and 40

minutes of scavenging, large increases in NMDAR peak current amplitudes were observed for

MPG (201.42 ± 8.25% [n=6, p< 0.001] and 195.06 ± 16.31% [n=4, p< 0.001], respectively) and

NAC (192.46 ± 19.86 % [n=6, p= 0.046] and 208.93 ± 37.32% [n=4 p= 0.035], respectively).

The effects of these increases often resulted in hyperactivity, depolarization and the loss of the

patch. Within the recordings that were maintained, normoxic reperfusion for 20 and 40 minutes

failed to reverse drug effects from MPG (182.84 ± 16.32% [n=3, p< 0.001] and 186.67 ± 5.33%

[n=3, p< 0.001], respectively) and only mildly reversed the effects of NAC (168.35 ± 26.05%

[n=3] and 139.55 ± 24.74% [n=3], respectively) [Figure 3.4]. In only one recording for both

experiments were the effects completely reversed after a period of 110 minutes for MPG and 120

minutes for NAC [Data Not Shown].

Page 63: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

48

Figure 3.4: Effect of ROS scavenging on whole cell NMDAR currents.

A, Percentage normalized NMDAR peak current amplitudes after 20 min (t=30, blue bars) and

40 min (t=50, red bars) of treatment followed by 20 min (t=70, green bars) and 40 min (t= 90,

purple bars) of washout. Dashed lines represent normoxic baseline controls taken at t=10 min

(red) and 40 min anoxic treatment value (blue). Data are expressed as means ± S.E.M. Asterisks

(*) indicate data significantly different from the paired normoxic control (p < 0.05). B-C, Paired

sample NMDAR currents of the initial baseline recording (t= 10 min) and following 20 min of

the indicated experimental treatment (t= 30 min). Abbreviations: N-2-mercaptopropionyl glycine

(MPG), n-acetylcysteine (NAC).

Page 64: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

49

3.2.4 NMDAR current amplitudes are decreased by H2O2 addition

In order to evaluate the effect of increases in [ROS]i on receptor activity the primary

mitochondrial ROS product H2O2 was administered under normoxic conditions. H2O2

administration was limited to 5 minutes prior to recordings in order to prevent/limit H2O2

toxicity. At 20 and 40 minutes of the treatment regimen significant decreases in NMDAR peak

current amplitudes were observed (80.24 ± 3.11% [n=9, p= 0.023] and 78.42 ± 4.43% [n=5, p=

0.036], respectively). The effects were completely reversed after reperfusion for 20 (93.23 ±

7.11% [n=4]) and 40 minutes (99.26 ± 7.78% [n=4]) [Figure 3.5].

3.2.5 Blocking mitochondrial ROS production increases NMDAR currents

In order to determine if the increases in NMDAR activity due to decreases in [ROS]i are

from a mitochondrial or non-mitochondrial source the mitochondrial complex I inhibitor

rotenone was used to block mitochondrial ROS production under normoxic conditions. After 20

and 40 minutes of rotenone treatment increases in NMDAR peak current amplitudes were

observed (216.57 ± 30.14% [n=5, p= 0.017] and 232.26 ± 28.47% [n=4, p= 0.01], respectively).

The effects were reversed after 20 and 40 minutes of reperfusion (100.08 ± 2.89% [n=3] and

104.56 ± 3.44% [n=2], respectively) [Figure 3.5].

Page 65: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

50

Figure 3.5: Effect of H2O2 manipulation on whole cell NMDAR currents.

A, Percentage normalized NMDAR peak current amplitudes after 20 min (t=30, blue bars) and

40 min (t=50, red bars) of treatment followed by 20 min (t=70, green bars) and 40 min (t= 90,

purple bars) of washout. Dashed lines represent normoxic baseline controls taken at t=10 min

(red) and 40 min anoxic treatment value (blue). Data are expressed as means ± S.E.M. Asterisks

(*) indicate data significantly different from the paired normoxic control (p < 0.05). B-C, Paired

sample NMDAR currents of the initial baseline recording (t= 10 min) and following 20 min of

the indicated experimental treatment (t= 30 min). Abbreviations: hydrogen peroxide (H2O2).

Page 66: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

51

3.2.6 Anoxia attenuated NMDAR currents are not reversed by ROS addition

In order to test if the effects of anoxia are reversed by the re-introduction of ROS, H2O2

was added after anoxic reductions of NMDAR were measured. As a result the time frame was

modified. Decreases in NMDAR peak current amplitudes were confirmed after 20 and 40

minutes of anoxia (69.62± 3.45% [n=5, p= 0.031] and 66.56 ± 5.96% [n=5, p= 0.013],

respectively). Anoxic conditions were maintained and H2O2 was administered over a further 40

minute period. H2O2 administration did not reverse anoxia induced changes after 20 or 40

minutes (58.60 ± 6.73% [n=5, p= 0.001] and 54.36 ± 5.57% [n=5, p< 0.001], respectively). The

decreases produced from H2O2 administration were not statistically significant from those

produced by anoxia alone [p >0.05]. Normoxic reperfusion only reversed the effects of anoxia

and H2O2 mildly after 20 minutes (59.00 ± 11.48% [n=3, p= 0.006]) and moderately after 40

minutes (75.36 ± 10.10% [n=3]) [Figure 3.6].

Page 67: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

52

Figure 3.6: Effect of H2O2 addition on anoxia-attenuated whole cell NMDAR currents.

A, Normalized NMDAR peak current amplitudes after 20 and 40 min of anoxia followed by 20

and 40 min of H2O2 treatment and concluded with 20 and 40 min of washout (n=6). Data are

expressed as means ± S.E.M. Asterisks (*) indicate data significantly different from the paired

normoxic control (p < 0.05). B, paired sample NMDAR currents of a 10 min control and 20 min

of anoxia (t= 30 min), and 20 min of H2O2 + anoxia (t= 70 min). Abbreviations: hydrogen

peroxide (H2O2).

Page 68: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

53

3.3 Whole-cell AMPAR current experiments

3.3.1 Normoxic AMPAR currents are stable throughout the time course

The time course utilized for AMPAR experiments was the same as that used for NMDAR

experiments. AMPAR currents under normoxic conditions were examined throughout the

experimental time course to confirm that current amplitudes remained stable. Normalized

AMPAR peak current amplitudes did not change after 10, 30, 50, 70 and 90 minutes of normoxic

saline perfusion (99.85 ± 4.40% [n=6], 96.92 ± 7.10% [n=6], 97.15 ± 7.01 [n=6], 104.94 ±

2.72% [n=4], 105.95 ± 0.78% [n=4] respectively) [Figure 3.7].

3.2.2 Normoxic AMPAR current amplitudes are reduced during anoxia

Like NMDARs, AMPARs also undergo large decreases in current amplitudes under

anoxic conditions. In order to generate anoxic decreases for comparisons to pharmacological

conditions anoxia was induced and currents were induced and evaluated within the established

time frame. Following 20 and 40 minutes of anoxia AMPAR peak current amplitudes were

reduced to a comparable degree of to those previously published (69.95 ± 5.69% [n=5, p= 0.006]

and 55.16 ± 6.04% [n=5, p< 0.001], respectively) [125, 234]. The effects were reversed after 20

and 40 minutes of normoxic reperfusion (98.76 ± 4.86% [n=4] and 101.32 ± 5.19% [n=4]

respectively) [Figure 3.7].

Page 69: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

54

Figure 3.7: Effect of anoxia on whole cell AMPAR currents.

A, Percentage normalized AMPAR peak current amplitudes after 20 min (t=30, blue bars) and 40

min (t=50, red bars) of treatment followed by 20 min (t=70, green bars) and 40 min (t= 90,

purple bars) of washout. Dashed lines represent normoxic baseline controls taken at t=10 min

(red) and 40 min anoxic treatment value (blue). Data are expressed as means ± S.E.M. Asterisks

(*) indicate data significantly different from the paired normoxic control (p < 0.05). B-C, Paired

sample AMPAR currents of the initial baseline recording (t= 10 min) and following 20 min of

the indicated experimental treatment (t= 30 min).

Page 70: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

55

3.3.3 AMPAR current amplitudes are unaffected by ROS scavenging

In order to test the hypothesis that decreased [ROS]i has an inhibitory effect on AMPAR

current amplitudes the ROS scavengers MPG and NAC were administered separately under

normoxic conditions. No changes in AMPAR peak current amplitude were found after 20 or 40

minutes for MPG (105.40 ± 4.61% [n=6] and 94.71 ± 7.30% [n=4], respectively) or NAC

(100.48 ± 3.21 % [n=7] and 103.48 ± 2.80 % [n=6], respectively). Currents remained unchanged

through 20 and 40 minutes of normoxic reperfusion for MPG (95.26 ± 3.29 % [n=4] and 97.81 ±

12.60 % [n=3]) and NAC (98.62 ± 2.81% [n= 4] and 97.04 ± 3.86 % [n=3]) [Figure 3.8].

3.3.4 AMPAR current amplitudes are unaffected by H2O2 addition

In order to evaluate if increases in [ROS] bring about changes in AMPAR currents the

primary mitochondrial ROS product H2O2 was administered under normoxic conditions.

Normoxic AMPAR peak current amplitudes were not altered by 20 or 40 minutes of H2O2

addition (100.00 ± 3.35% [n=11] and 99.19 ± 2.44% [n=7], respectively). AMPAR current

amplitudes remained unchanged through 20 and 40 minutes of normoxic reperfusion (106.33 ±

3.91% [n=5] and 100.00 ± 2.95% [n=3], respectively) [Figure 3.8].

Page 71: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

56

Figure 3.8: Effect of ROS manipulation on whole cell AMPAR currents.

A, Percentage normalized AMPAR peak current amplitudes after 20 min (t=30, blue bars) and 40

min (t=50, red bars) of treatment followed by 20 min (t=70, green bars) and 40 min (t= 90,

purple bars) of washout. Dashed lines represent normoxic baseline controls taken at t=10 min

(red) and 40 min anoxic treatment value (blue). Data are expressed as means ± S.E.M. Asterisks

(*) indicate data significantly different from the paired normoxic control (p < 0.05). B-C, Paired

sample AMPAR currents of the initial baseline recording (t= 10 min) and following 20 min of

the indicated experimental treatment (t= 30 min). Abbreviations: n-acetylcysteine (NAC), N-2-

mercaptopropionyl glycine (MPG), hydrogen peroxide (H2O2).

Page 72: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

57

3.4 Oregon green intracellular calcium assessments

3.4.1 Excitation scans confirmed dye loading in turtle brain sheets

Dye-loading within the cells was confirmed by running excitation scans from 450 nm to

550 nm prior to all recordings. Peak excitation was observed around 488 nm in all dye loaded

cell but was absent in non-dye loaded cells [Figure 3.8].

Figure 3.9: Oregon Green excitation scans.

A-B, sample excitation scans from 450 nm to 550 nm in Oregon Green dye-loaded (A) and non-

dye-loaded (B) neurons.

3.4.2 Normoxic calcium concentrations are stable throughout the time course

The time course for fluorescent experiments was different from that utilized for whole

cell recordings. Photo bleaching occurs after extended periods of exposure that can cause a loss

of the fluorescent signal. The experimental time frame consisted of a 10 minute normoxic time

Page 73: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

58

period that allowed for the fluorescent signal to stabilize, followed by 10 minutes of

pharmacological treatment, and 10 minutes of normoxic recovery. To confirm that the Oregon

Green fluorescence was stable throughout the experimental time course traces were collected

under normoxic conditions through a 30 minute time period. Under normoxic conditions Oregon

Green fluorescence did not change significantly with respect to baseline (0.61 ± 0.77% [n= 6])

and decreased at a steady rate due to photo-bleaching and/or dye exocytosis [Figure 3.10].

3.4.3 Intracellular calcium concentrations are increased by anoxia

The entrance into anoxia is associated with a mild increase in intracellular calcium [126,

128, 149]. These experiments were replicated to confirm previous findings and generate rates of

change for comparison to pharmacological treatments. Anoxic administration mildly increased

Oregon Green fluorescence (16.94 ± 3.29% [n= 6, p< 0.001]) [Figure 3.10].

3.4.4 Intracellular calcium concentrations are unaffected by ROS scavenging

As previously discussed intracellular calcium is increased as a result of anoxia. In order

to determine if decreases in ROS served to bring about the mild increases in calcium associated

with anoxia the ROS scavengers MPG and NAC were separately applied during an experimental

time course. Oregon green fluorescence was not significantly altered with respect to the baseline

by MPG or NAC addition (0.56 ± 0.64% [n= 6] and 0.96 ± 1.28% [n= 6]) [Figure 3.10].

Page 74: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

59

3.4.5 Intracellular calcium concentrations are unaffected by H2O2 addition

H2O2 was bulk perfused onto the slice as drip perfusion proved unreliable and affected

image stability. H2O2 bulk concentrations were altered to be comparable to those brought on by

drip perfusion into the recording bath during electrophysiological recordings (drip= 50 uM,

bulk= 5 uM). H2O2 addition did not significantly alter fluorescence in respect to baseline (0.84 ±

1.39% [n= 6]) [Figure 3.10].

Page 75: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

60

Figure 3.10: Effect of oxygen and ROS manipulation on Oregon Green fluorescence.

Percentage normalized changes in Oregon Green fluorescence post treatment. Data are expressed

as means ± S.E.M. Asterisks (*) indicate data significantly different from the paired normoxic

control (p < 0.05). Abbreviations: Arbitrary Fluorescence Units (AFU), N-2-mercaptopropionyl

glycine (MPG), n-acetylcysteine (NAC), Hydrogen peroxide (H2O2).

Page 76: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

61

Figure 3.11: Sample Oregon Green fluorescence traces.

Sample traces of Oregon Green fluorescence from neurons treated as indicated. Changes in

fluorescence were calculated between two parallel tangents, as illustrated in B. C, Illustration of

the method by which Fmin and Fmax were determined. Abbreviations: Arbitrary Fluorescence

Units (AFU), N-2-mercaptopropionyl glycine (MPG), n-acetylcysteine (NAC), Hydrogen

peroxide (H2O2).

Page 77: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

62

3.5 Action potential frequency assessments in spontaneously active pyramidal cells

3.5.1 Action potential frequency is reduced by ROS scavenging

As previously discussed entrance into anoxia is associated with an increase in

GABAergic PSCs that serve to bring about SA and inhibit/decrease neuronal firing [115-117].

This is most clearly seen within neurons that exhibit spontaneous activity and fire continuously

at regular intervals. Recordings from these cells within turtle cortical sheets demonstrate a 70%

reduction in action potential frequency (APf) with the transition to anoxia [112]. ROS scavenging

with MPG and NAC has also been shown to bring about increases in GABAergic PSCs similar

to anoxia [231]. Therefore, in order to determine if decreases in ROS result in SA similar to that

seen during anoxia, spontaneously active pyramidal cells were treated with MPG during APf

recordings. APf decreased by approximately 97% after 18 minutes of MPG treatment (0.04 ±

0.04 [n=4, p< 0.001]) that was reversed and increased after 13 minutes of normoxic reperfusion

(2.33 ± 0.44 [n=4, p= 0.016]) [Figure 3.11].

Page 78: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

63

Figure 3.12: Changes in spontaneous activity in response to ROS scavenging.

A, Summary of APf expressed as fold change relative to pretreatment baseline. Data are

expressed as means ± S.E.M. Dashed line represents normoxic baseline APf. Asterisks (*)

indicate data significantly different from the paired normoxic control (p < 0.05). B, Raw

spontaneous activity sample trace. Abbreviations: N-2-mercaptopropionyl glycine (MPG).

Page 79: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

64

CHAPTER 4: DISCUSSION

4.1 Control normoxic and anoxic NMDA and AMPA currents

In agreement with previous findings normoxic NMDA and AMPA administration

produced consistent repeatable currents throughout the 90 minute time course of the experiment.

The introduction of anoxia produced a decrease of approximately 50% in both NMDA and

AMPA currents within 20 minutes consistent with previous reports [126, 149, 234]. This served

to further confirm neurological stability and pharmacological validity.

4.2 Pharmacological manipulation of ROS

4.2.1 The NMDAR modulatory redox site in turtle cortical slices

Cellular ROS scavenging with both MPG and NAC produced massive increases in

NMDAR whole cell currents contrary to the anoxia mediated decrease typically observed in the

western painted turtle cortex. Inversely, the addition of H2O2 under normoxic conditions

decreased NMDAR whole cell currents. Although this indicates that decreases in ROS are not

responsible for NMDAR attenuation during anoxia, the findings are consistent with what has

been documented within neural tissue from other vertebrates and may reveal a conserved

mechanism of NMDAR control during normoxia [190-198].

As discussed above, pharmacological increases in ROS or the addition of oxidizing

agents has been documented to decrease NMDAR activity within tissue preparations from

Page 80: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

65

mammals, amphibians, and birds. Pharmacological decreases in ROS or the addition of reducing

agents has also been shown to produce increases in NMDAR activity within these same groups.

These changes have been previously attributed to alterations in the oxidation/reduction state of

extracellular cysteine residues found within the NMDAR GluN1 (Cys744, Cys798, Cys 79 and

Cys 308), GluN2A (Cys 87, Cys 320 and Cys 399), and/or GluN2B subunits (Cys not yet

localized) that collectively make up NMDAR redox sites [187-189]. The changes documented

within cortical slices from the turtle during ROS manipulation may also be attributed to

alterations in the NMDAR redox site. Specific regions within NMDAR subunits have been

documented to be highly conserved between vertebrate species. Comparisons of the Brown ghost

knifefish GluN1 amino acid structure found an 88% sequence similarity to the amino acid

structures in frog, duck, rat and human [235]. Similarly, the amino acid structures of the four

NR2 subtypes possess high levels of conservation between species within sequences coding the

lining of the ion pore, glutamate binding pockets, and C-terminal region [119, 236]. The redox

modulatory sites on the GluN1 and GluN2 subunits may be similarly conserved between species.

Increases and decreases in NMDAR current amplitude between ROS scavenging and

addition were not uniform. The decreases in receptor currents with H2O2 addition were only

~20% compared to the ~100% increase with ROS scavenging. This is consistent with what has

been seen in other vertebrates where the addition of reducing agents produced increases that

were 6-11 times greater than decreases produced through oxidation [190, 196, 199]. Our findings

suggest that the turtle NMDAR redox modulatory site is maintained closer to a fully oxidized

than fully reduced form under normoxic conditions, in agreement with what has been

documented in other vertebrate species. This provides support for the Cys residues being found

on the extracellular portion of the receptors, where the environment is predominantly oxidized

Page 81: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

66

[168]. It may also indicate a maintained ROS presence in close proximity to the receptors and

redox sites.

The level of recovery from treatment varied drastically between ROS scavenging and

addition. Although recovery from H2O2 was achieved within the 40 minute reperfusion period,

MPG/NAC recovery was not and cells often depolarized and did not recover preventing the

exploration of a longer recovery period. This is consistent with what has been documented in

other vertebrates where decreases in NMDAR activity induced through the application of

oxidizing agents were consistently reversed with washout, but increases in NMDAR activity

induced through reductive agents were maintained. The effects of reducing agents have only

been documented to be reliably recovered to baseline through the addition of oxidizing agents

[196, 237]. The reasoning behind the lack of recovery from scavenging/reducing agents may be

explained by slow cellular ROS production and slow degradation of scavenging/reducing agents.

As discussed above, ROS are produced slowly in the cell (only 2-3% of all mitochondrial O2

consumed) maintained at low levels and may move slowly across the extracellular membrane

[168]. It may take a long time for baseline levels to be recovered following ROS scavenging

and/or re-oxidize cysteine residues affected by scavenging agents. Furthermore, scavenging and

reducing agents may take a long time to be removed from the cell or to be degraded through

oxidation, further delaying the re-establishment of baseline ROS levels and NMDAR activity.

The addition of oxidizing agents would help in re-oxidization of redox sites and the degradation

of scavenging/reducing agents, potentially explaining why this tactic was successful in reversing

the effects of reducing agents on NMDAR activity. Cells are likely to be better equipped to deal

with ROS increases then their absence as the complete removal of ROS is unlikely for most

aerobic life.

Page 82: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

67

The risks of NMDAR over activation and resulting ECD are common to all vertebrates

and NMDAR activity must be tightly regulated to prevent it. Ca2+ entry must be maintained

within a range where intake concentrations are great enough to signal downstream changes but

small enough to be taken up by the mitochondria and ER so that they don’t bring on toxic

effects. Most cellular activities are maintained within a desired range via negative feedback

systems allowing products generated by specific processes to regulate the actions of the

processes themselves. NMDAR Ca2+ dependent ROS production via nNOS or mitochondrial

activation may be a mechanism through which NMDAR activity is regulated among vertebrates.

4.2.2 Mitochondrial produced ROS in NMDAR redox modulation

The mitochondrial complex I inhibitor rotenone decreases ROS production in various

vertebrate tissues including neuronal tissue [157, 200-202]. Rotenone addition in turtle cortical

slices produced increases in NMDAR currents comparable to what was seen with MPG/NAC

ROS scavenging. This suggests that changes in mitochondrial ROS production affect NMDAR

activity. Full recovery from rotenone treatment was achieved within the reperfusion period

unlike what was seen with scavenging agents. This supports the concept that the recovery delay

seen during scavenging is the result of agents taking a long period to be degraded and removed

from the cell.

The finding that mitochondrial ROS production affects NMDAR activity does not rule

out a role for NO generated from nNOS. During whole cell patch experiments, TTX application

and voltage clamping would have prevented natural glutamate release and blocked NO

Page 83: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

68

production except during NMDA addition. Because nNOS activity directly linked to NMDAR

activation and NO can oxidize cysteine residues in a similar manner to H2O2, it is very likely that

both NO and mitochondrial ROS play a role in NMDAR control. In studies within other

vertebrates the addition of nNOS inhibitors increases baseline NMDAR currents while artificial

NO generators bring about receptor attenuation [241, 242]. However, nNOS inhibitors do not

prevent mitochondrial ROS increases triggered by NMDAR activation, suggesting that the

mechanisms function independently of each other [238, 239]. Further investigation is necessary

to determine the role and contribution of NO in NMDAR redox control.

Mitochondrial ROS generation is increased by NMDAR activation within mammalian

neural cultures. NMDA mediated ROS increases were not prevented by nNOS inhibitors but

were blocked by the removal of extracellular calcium, rotenone/ other mitochondrial uncouplers,

and Ca2+ uniporter inhibitors [238, 239]. This suggests that increased mitochondrial ROS is

generated as a result of NMDAR specific Ca2+ influx and increased mitochondrial Ca2+ uptake. If

Ca2+ influx were to exceed the capabilities of the storage systems like mitochondrial uptake, then

Ca2+ would begin to spread through the cell causing toxicity. It is therefore not unreasonable that

mitochondrial uptake might influence NMDAR activity.

Mitochondrial calcium uptake is primarily accomplished through the actions of the

mitochondrial calcium uniporter (MCU) located on the inner mitochondrial membrane (IMM).

The outer mitochondrial membrane is permeable to most large ions (<6000 DA) allowing for

unaided Ca2+ entry. The IMM is not as permeable and transporters are required to allow for Ca2+

movement [240]. The MCU becomes active in response to large concentrations of localized Ca2+

known as microdomains that in excitable cells need to be 500-600 nM in order to generate a

Page 84: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

69

response [241]. Activity is driven by the large negative potential of the mitochondrial matrix

established by the actions of the respiratory chain. Electrophoretic uptake of Ca2+ into the

mitochondrial matrix through the uniporter partially dissipates the electrochemical gradient [242,

243]. Compensation is accomplished via Ca2+ induced increases in electron transport chain

activity that pump H+ out of the matrix helping to re-establish the electrochemical gradient and

also bringing about increases in ATP production [242]. Enhancements in ROS production can be

accomplished through the increases in ETC activity, generating ROS as a by-product. There is

also some evidence to suggest that matrix Ca2+ can partially dislocate cytochrome c from the

IMM thereby slowing down the electron transfer from complex III to complex IV and enhancing

ROS generation at the Q cycle [239, 244]. Ca2+ within the matrix is transient and the actions of

multiple Ca2+ efflux mechanisms, the primary one in excitable tissues being the Na+/Ca2+

exchanger, work in concert to move Ca2+ back into the cytosol [242, 245]. Storage within the

mitochondrial matrix can be quite high and has been documented in excess of 100 uM [246].

However, the capacity is not unlimited and if Ca2+ overload occurs and intake exceeds efflux

mitochondrial membrane potentials can collapse leading to excessive Ca2+ efflux and the

triggering of apoptosis [247, 248].

4.2.3 A potential mechanism for ROS mediated NMDAR control

For NMDAR activity to be regulated via oxidation/reduction ROS synthesis must not

only be tied directly to receptor activity, ROS production must be accomplished within close

proximity to the channel. The redox site will be maintained in a preferentially oxidized state by

likely existing within the extracellular environment, but changes in intracellular ROS changes

will only be effective signals if ROS production occurs within the vicinity of the NMDAR. As

Page 85: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

70

discussed above, this may involve NO produced by nNOS that is tethered to NMDAR via PSD-

95. However, for mitochondrial ROS generation to be effective in modulating activity as shown

the rotenone experiments, mitochondrial localization must also take place.

ROS are produced from the mitochondria in relatively small quantities (2-3% of O2

consumed) that are maintained at low levels through the actions of cellular antioxidant defence

systems. This coupled with high reactivity leads to small and inconsistent mitochondrial ROS

levels that exist throughout the cytoplasm limiting their signalling ability to short distances [163,

249]. Furthermore, in order for ROS to affect extracellular redox sites ROS must pass into the

extracellular environment through diffusion or WTRs where it can then be further degraded by

extracellular antioxidants [168-171]. Therefore in order for ROS control to be effective and

consistent, target cysteine residues must be in close proximity to the mitochondria. NMDARs are

primarily located within post synaptic densities of the excitatory synapse and mitochondria are

similarly packed into these synapses in order to provide the ATP necessary for synaptic activities

[250-252]. These synaptic regions are high in membrane ion channels that transport water

(WTRs) potentially allowing for an efficient flux of generated ROS across the membrane [172].

Recent investigation in mammalian neurons found NMDAR specific Ca2+ entry was far more

rapidly taken up by the mitochondria than Ca2+ entry through other channels [253]. This tight

coupling of mitochondrial Ca2+ uptake to NMDAR activation is suggested to be the result of the

organelles and receptors being maintained within close proximity to each other in order to better

control cytosolic Ca2+ levels [253, 254]. There is a considerable body of work in non-excitable

cells that suggests that rapid mitochondrial Ca2+ uptake requires high levels of Ca2+ to be present

in close proximity (~2-5 uM) to the mitochondria and that the endoplasmic reticulum and

mitochondria are positioned so that excess calcium leaving the ER is high enough to signal this

Page 86: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

71

action and be quickly sequestered [255-259]. Within excitable cells like neurons this high Ca2+

concentration has been shown not to be required to activated rapid uptake, but localized cytosolic

Ca2+ levels of above 500–600nM are still required to stimulate influx supporting the tight

coupling concept [241].

Given my experimental findings and those of similar experiments conducted in other

organisms oxidation/reduction of the NMDAR redox site appears to have a similar effect across

multiple vertebrate species suggesting a common mechanism of control. Using information

collected from other studies I submit the following potential mechanism for mitochondrial ROS

mediated NMDAR control under normoxic conditions. I do this in order to try and better explain

how ROS control might be accomplished so that that I can better explain later how anoxia

mediated mechanisms may override these processes. The proposed mechanism for is as follows:

NMDAR activation causes an influx of extracellular Ca2+ into the cell that serves to

signal changes downstream changes via activation of receptor localized Ca2+-dependent

enzymes. Excessive calcium that does not become bound is rapidly taken up by localized

mitochondria to prevent it from spreading out through the cell and undergoing unwanted

reactions. Ca2+ freely flows across the permeable mitochondrial outer membrane and is then

taken up into the matrix via the action of the MCU. Ca2+ entrance stimulates increases in ETC

activity to compensate for the inward ion flow, generating increased levels of ROS as result. The

ROS produced will rapidly move out of the mitochondria and into the cytosol. ROS may then

move across the extracellular membrane and into the extracellular environment by simple

diffusion or through WTRs where it can interact with NMDAR redox sites in close proximity,

reversibly oxidizing the modulatory redox site and decreasing the receptors open time in

Page 87: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

72

response to stimulus. The cumulative action of multiple mitochondria undergoing these

processes will serve to generate a cloud or microdomain of ROS that will help maintain

NMDARs primarily in an oxidized state and resist the actions of antioxidant enzymes. Increases

in NMDAR activation will bring on increases in Ca2+ influx and subsequent ROS generation that

will increase receptor oxidation and decrease sensitivity. Similarly decreases in NMDAR

activation will lead to decreases in ROS allowing for enzymes like GSH/L-Cys to reduce a

greater portion of receptors and increase sensitivity. Through this negative feedback mechanism

Ca2+ influx can be maintained within a range that brings about necessary changes without

becoming high enough to overtake mitochondrial uptake mechanisms and trigger overload

[Figure 4.1].

Although this mechanism will maintain NMDAR activity within a normal range during

normoxia it will not function under anoxic conditions as mitochondrial ROS generation requires

O2. The loss of cellular oxygen will halt oxidative phosphorylation and allow for cellular ROS

levels to be quickly depleted. The method of maintained NMDAR inhibition will quickly be lost

as the receptors are rapidly reduced leading to hyper excitability. Activation of the receptors will

trigger a massive influx of Ca2+ bringing about excitotoxic cell death. In order for the western

painted turtle to survive prolonged anoxia another mechanisms must serve to override ROS

control and trigger the decreases in NMDAR activity documented to occur.

Page 88: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

73

Figure 4.1: Diagrammatic representation of the proposed mechanism for ROS mediated

NMDAR control under normoxic conditions.

Page 89: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

74

4.2.4 AMPAR redox insensitivity

AMPAR currents were unaffected by MPG, NAC or H2O2 consistent with what is seen in

mammalian tissue with oxidizing/reducing agents suggesting that the mechanisms for AMPAR

control in respect to ROS changes are conserved between species [196-198, 203].

The reasoning behind why there exists a mechanism for ROS mediated NMDAR control

but not AMPAR may be a direct result of how oxidation of the NMDAR leads to inhibition of its

activity. AMPARs activation produces excitatory postsynaptic potentials (EPSPs) that cause

neuronal depolarization and the removal of Mg2+ from the pore region of the NMDAR allowing

activation and Ca2+ influx [260-263]. It is through this indirect mechanism of action that

AMPARs exhibit control over intracellular Ca2+ influx. Changes to the modulatory redox site

may override AMPAR control by affecting the efficiency to which NMDARs can maintain Mg2+

within the pore region. Treatment of mammalian neural tissue with rotenone resulted in increases

in NMDAR activity as previously discussed, but also documented that NMDAR currents lost the

characteristic region of negative-slope conductance that is normally produced by voltage-

dependent block by Mg2+. NMDAR increases were mimicked by low extracellular Mg2+ and

rotenone induced increases were antagonized by a high levels of Mg2+. Rotenone addition had no

effect on AMPAR currents [203, 206]. This suggests that oxidation/reduction alters the ability of

the NMDAR to maintain Mg2+ within the pore region and overrides/circumvents AMPAR

control.

Page 90: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

75

The lack of AMPAR down regulation in response to decreased [ROS]i further supports

the concept that ROS changes are not responsible for NMDAR/AMPAR current decreases in the

anoxic Western painted turtle brain and that a secondary mechanism must be at work.

4.2.5 Anoxia-mediated block of NMDAR redox control

Anoxic decreases in NMDAR currents were not reversed by the addition of H2O2

confirming that ROS changes are not responsible for anoxia mediated changes. H2O2 addition to

anoxia-attenuated NMDAR currents appeared to further reduce whole cell currents (~10%), but

it is not clear if this decrease was simply the result of extended anoxia. Average currents after 90

minutes of anoxia with H2O2 reached ~54% of normoxic levels that was not significantly

different from that seen after 30 minutes of anoxia in control experiments (~58%) or previous

publications (~52%) [149]. H2O2 addition alone produced decreases after 40 min of

approximately 22% from normoxic baseline compared to addition after anoxia that produced

after 40 min only an approximate 12% change. This would seem to support the concept that the

secondary mechanism for triggering anoxic NMDAR activity decreases exists that overrides the

mechanisms of ROS control. However without anoxic controls for a comparable period of time

the assumption cannot be verified. Regardless, the results confirm that another mechanism aside

from ROS decreases is responsible for the induction of NMDAR down regulation. The most

likely candidate for these changes is changes in cellular calcium from an intracellular source.

Page 91: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

76

4.3 A role for anoxia-mediated calcium increases in blocking redox control

A mild increase in cytosolic calcium from the mitochondria is seen during anoxia in the

western painted turtle and is necessary to bring about decreases in NMDAR and AMPAR whole

cell currents [125, 264]. Investigations using Oregon Green Ca2+ fluorescence confirmed an

anoxic increase in intracellular calcium similar to what has been previously documented (~15%)

[149]. However, the addition of MPG, NAC and H2O2 failed to elicit any change in the

fluorescent Ca2+ signal demonstrating that ROS changes do not play a role in the anoxia-

mediated increase in cytosolic Ca2+. This serves to confirm that a secondary mechanism for

anoxia-mediated changes is at work and suggests that mitochondrial release of Ca2+ is central to

its activity.

Increases in mitochondrial Ca2+ release are associated with activation of the calcium-

binding messenger protein calmodulin (Ca2+/calmodulin). Ca2+/calmodulin is a small Ca2+

binding protein that is highly conserved within eukaryotes, present in high concentrations in the

brain and capable of modulating a number of different enzymes and cellular processes [265,

266]. Within the western painted turtle inhibition of Ca2+/calmodulin has been demonstrated to

prevent decreases in NMDAR activity. Ca2+/calmodulin may also activate the Ca2+/calmodulin-

dependent phosphatase PP2B (calcineurin), whose inhibition has also been shown to inhibit early

anoxia mediated NMDAR downregulation. Interestingly inhibition of PP1 and PP2A was also

shown to inhibit NMDAR decreases suggesting a dependency between both dephosphorylation

and calmodulin interaction [129]. The sites of modification and binding offer means for

NMDAR/AMPAR control that may also serve to override the mechanism of ROS mediated

NMDAR regulation.

Page 92: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

77

Calmodulin has been shown to exert effects via binding to two identified sites (CBS1 and

CBS2) the C0 terminus of the GluN1 via destabilization of the NMDARs from the cytoskeleton

[267-269]. It has also been demonstrated that phosphorylation of the GluN1 serine residues

decreases affinity for calmodulin, an effect that can be reversed by the actions of PP1 and PP2A

[150]. This has led to the hypothesis that during anoxia, PP1 and 2A dephosphorylate the serine

residue on the C-terminus of the GluN1 subunit allowing for the binding of Ca2+/calmodulin and

an attenuation of NMDAR function [264]. As discussed above NMDAR redox modulatory sites

have been identified on the GluN1, (Cys744, Cys798, Cys 79 and Cys 308) GluN2A (Cys 87,

Cys 320 and Cys 399) and GluN2B (not yet localized) [187-189]. GluN1 dephosphorylation or

calmodulin binding both offer a potential means to block modulatory redox sites, preventing

oxidation/reduction and overriding ROS control. Furthermore, calcineurin dephosphorylates the

GluN2A subunit (not GluN1) that besides having been shown to contribute to glycine

desensitisation and NMDAR current reduction, offers a means through which modulatory redox

sites on the GluN2A subunit may be blocked [267, 270]. It is also possible that calmodulin

induced NMDAR cytoskeletal dissociation may bring about destabilization the modulatory redox

site leading to a loss of ROS control. AMPAR activity has not been demonstrated to be regulated

through calmouldin binding but has been identified to be controlled by dephosphorylation.

Increases in calcineurin activity dephosphorylate Ser845 on the GluA1 subunit triggering a

decrease in AMPAR function [271-273].

In support of this mechanism, inhibition of intracellular calcium release and subsequent

phosphatase activity has been associated with increases in NMDAR activity under anoxic

conditions. Addition of the calcium chelator BAPTA increased NMDAR whole cell currents by

Page 93: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

78

35% after 20 minutes of anoxia. Similarly addition of the PP1/PP2A inhibitor calyculin A

increased NMDAR whole cell currents by 45% after 40 minutes of anoxia. Finally, addition of

the calmodulin inhibitor calmidazolium increased NMDAR whole cell currents by 20% after 40

minutes of anoxia [129].

The mechanism by which calcium is released from the mitochondria has yet to be

established. However recent investigation has suggested that it is the result of mKATP activation

and subsequent mitochondrial permeability transition pore (mPTP) formation. The decreases in

AMPAR and NMDAR activity seen during anoxia are mimicked by mKATP activators and

blocked by mKATP inhibitors [125, 126]. Furthermore, inhibition of mKATP prevents the mild

increase in intracellular Ca2+ characteristic of anoxia, while activation produces Ca2+ increases of

a similar magnitude[126]. mKATP channels are traditionally gated by high levels of ATP under

normoxia. During anoxia ATP production at the level of the mitochondria decreases causing

local ATP levels to be reduced potentially leading to the opening of mKATP channels. Opening

then leads to an influx of K+ that can trigger mPTP formation as a result of mitochondrial

membrane depolarization. The mPTP is a temporary channel that can be formed in the inner

mitochondrial membrane and permit Ca2+ release. The formation of a singular large mPTP often

bring on excessive Ca2+ release and toxic results, but the formation of several small mPTPs has

been shown to bring about safe levels of Ca2+ efflux [274]. Within the western painted turtle, the

addition of an mPTP activator produced decreases in NMDAR currents similar to those seen

during anoxia that were abolished by the addition of a Ca2+ chelator. The addition of the mPTP

activator also produced an increase in Ca2+ fluorescence identical to that seen during anoxia

[149]. The mPTP is naturally formed under conditions of mitochondrial stress such as K+

mediated uncoupling that can trigger mild to moderate membrane depolarization (10-20%) [275].

Page 94: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

79

Furthermore, the pore is traditionally unstable and short lived causing it to quickly collapsing

and potentially only produce a mild increase in intracellular calcium [276].

4.4 Roles for redox sensitive GABA release

Entrance into anoxia is associated with a decrease in spontaneous AP frequency of

approximately 70%, which is attributed to increases in GABAR activity [112]. Recently

decreases in ROS were shown to produce increases in GABAergic PSC activity suggesting that

they play a role in GABAergic anoxic signalling [231]. In support of these findings MPG

addition reduced spontaneous AP frequency similar to anoxia although to a greater degree

(~98%). These results serve to confirm that although decreases in ROS during anoxia are not

responsible for NMDAR and AMPAR down regulation they do appear to be involved in the

processes controlling GABAergic activity increases.

Recovery from MPG treatment resulted in a major increase in firing above previous

baseline levels (~74%). This may be the result of NMDAR over excitation and Ca2+ influx,

triggered by ROS decreases as previously discussed. As discussed previously, MPG may take a

long time to be removed from the cell leading to NMDAR currents increases that were not

reversed after 40 minutes of normoxic reperfusion. Therefore, even though GABA mediated

ROS effects were quickly reversed the NMDAR augmentation may have remained and resulted

in excessive Ca2+ influx, increased neuronal firing, and generated the AP frequency increases

seen.

Page 95: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

80

Interestingly, increases in GABAergic PSCs will serve to decrease/inhibit postsynaptic

glutamate release preventing NMDAR and AMPAR activation. Through this mode of action

GABA changes in response to low oxygen may serve as a preliminary level of protection,

preventing NMDAR from activating in a reduced/overexcited and triggering ECD long enough

for the mechanisms controlled via mitochondrial Ca2+ release to take effect.

4.5 Potential mechanism of NMDAR/AMPAR control under anoxic conditions

The mechanisms proposed above for control of NMDAR activity through redox changes

will maintain intracellular Ca2+ influx within a normal range under normoxic conditions.

However, during anoxia O2 and ROS depletion will produce a state of NMDAR hyper

excitability that has the potential to produce massive increases in Ca2+ intake. Within the brain

of the western painted turtle ECD is not seen to occur and NMDAR/AMPAR receptors

experience major down regulations in activity. This suggests that a secondary mechanism of

control is activated by the anoxic state that can override the mechanism of ROS/redox control.

Given my and others findings in the brain of the western painted turtle during oxygen

deprivation I propose the following mechanism of anoxia-mediated NMDAR/AMPAR control:

as oxygen concentrations are depleted within neuronal tissue oxidative phosphorylation will

begin to cease. This will result in mitochondrial localized decreases in ATP triggering the

opening of mKATP channels. Influx of K+ through these channels will trigger mild/moderate loss

of the mitochondrial membrane potential and bring about the formation of multiple small

mPTPs. Ca2+ will move rapidly through these channels that do to their instability quickly

collapse. The resulting mild cytosolic Ca2+ increase will bind to calmodulin activating it.

Page 96: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

81

Ca2+/calmodulin can then bind directly to the NMDAR GluN1 subunit either with or without the

aid of dephosphorylation via PP1 and PP2A. This binding will serve to inhibit receptor activity

as well as block the GluN1 redox modulatory sites overriding the mechanisms of ROS control.

The activation of calcineurin via calmodulin binding will serve to dephosphorylate the GluN2A

subunit contributing to receptor down regulation and GluN2A modulatory redox site

destabilization. Calcineurin will also dephosphorylate the Ser845 on the GluA1 subunit of the

AMPAR down regulating activity. While these changes are occurring increases in GABAergic

activity triggered by decreasing ROS levels will serve to prevent glutamate release and

subsequent NMDAR/AMPAR activation [Figure 4.2].

Page 97: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

82

Figure 4.2: Diagrammatic representation of the proposed mechanism for anoxia mediated

NMDAR/AMPAR downregulation.

Page 98: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

83

4.6 Concluding remarks and directions for future research

These results suggest that changes in oxidation/reduction state of the NMDAR

modulatory redox site lead to increases and decreases in activity respectively, without effect on

AMPAR. This is in agreement with what has been demonstrated within investigations in other

vertebrates. To our knowledge this is the first case where evidence for this mechanism has been

demonstrated in reptiles. Our findings also support the hypothesis that these redox changes are

regulated by natural ROS levels, and in this way receptor function can be conservatively

regulated between vertebrates under normoxic conditions. Our research demonstrated that this

mechanism is not responsible for NMDAR/AMPAR downregulations seen during anoxia within

the western painted turtle, but found evidence to support an alternative mechanism involving

mitochondrial Ca2+ release.

In order for a better determination of the validity of the mechanisms proposed, a number

of questions still need to be answered and different experiments should be conducted. (1) It has

yet to be conclusively established how anoxia-mediated changes override ROS control, though

there is evidence to suggest it occurs through mitochondrial Ca2+ release. The mKATP activator

diazoxide has been previously documented to produce anoxic decreases in NMDAR currents

similar to those seen during anoxia. Diazoxide mediated decreases under normoxia should be

evaluated in conjunction with MPG/NAC in order to determine if scavenging effects are

mitigated. (2) The effects of phosphatases or calmodulin on AMPAR activity have yet to be

evaluated in the western painted turtle brain. Anoxia mediated AMPAR currents should be

evaluated in the presence of inhibitors for PP1, PP2A, calmodulin, and calcineurin in order to

determine if which if any of these groups is responsible for anoxia mediated AMPAR

Page 99: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

84

downregulation. (3) The role of NO in NMDAR control has yet to be firmly established. nNOS

inhibitors should be used in the same manner as roteone to determine if their blockade produces

comparable increases to ROS scavenging.

Understanding neurological anoxia tolerance is critical to many of the medical problems

faced within our society today. Stroke represents the third leading cause of death in North

America and with both childhood and adult obesity rates increasing across North America the

number of incidents is predicted to increase [277]. The development of preventative and fast

acting measures, like seen in the overwintering capabilities of the western painted turtle, may

offer a means to save thousands of lives.

Page 100: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

85

REFERENCES

1. Bärtsch P. & Gibbs J.S.R. Effect of altitude on the heart and the lungs. (2007).

Circulation, 116(19): p. 2191-2202.

2. Bosley T.M., Woodhams P.L., Gordon R.D. & Balazs R. Effects of anoxia on the

stimulated release of amino-acid neurotransmitters in the cerebellum invitro. (1983).

Journal of Neurochemistry, 40(1): p. 189-201.

3. Lutz P.L., Nilsson G.E. & Prentice H.M. The brain without oxygen: causes of failure -

physiological and molecular mechanisms for survival. 3rd ed. (2003), The Netherlands:

Kluwer Academic Publishers. 260.

4. Lipton P. Ischemic cell death in brain neurons. (1999). Physiological Reviews, 79(4): p.

1431-1568.

5. Bickler P.E. & Buck L.T. Adaptations of vertebrate neurons to hypoxia and anoxia:

maintaining critical Ca2+ concentrations. (1998). Journal of Experimental Biology,

201(8): p. 1141-1152.

6. Dave K.R., Prado R., Raval A.P., Drew K.L. & Perez-Pinzon M.A. The Arctic ground

squirrel brain is resistant to injury from cardiac arrest during euthermia. (2006). Stroke,

37(5): p. 1261-1265.

7. Snook J. Ice age extinction. (2008), New York: Algora Publishing.

8. Pielou E.C. After the ice age: the return of life to glaciated North America. (1991):

University of Chicago Press.

9. Rogers R.A., Rogers L.A., Hoffmann R.S. & Martin L.D. Native-american biological

diversity and the biogeographic influence of ice-age refugia. (1991). Journal of

Biogeography, 18(6): p. 623-630.

10. Koch P.L. & Barnosky A.D. Late quaternary extinctions: state of the debate. (2006).

Annual Review of Ecology Evolution and Systematics, 37: p. 215-250.

11. Macpherson A.H. The origin of diversity in mammals of the Canadian arctic tundra.

(1965). Systematic Zoology, 14(3): p. 153-173.

12. Hewitt G.M. Post-glacial re-colonization of European biota. (1999). Biological Journal

of the Linnean Society, 68(1-2): p. 87-112.

13. Hewitt G.M. Some genetic consequences of ice ages, and their role in divergence and

speciation. (1996). Biological Journal of the Linnean Society, 58(3): p. 247-276.

14. Hewitt G.M. Speciation, hybrid zones and phylogeography - or seeing genes in space

and time. (2001). Molecular Ecology, 10(3): p. 537-549.

Page 101: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

86

15. Stone K.D., Flynn R.W. & Cook J.A. Post-glacial colonization of northwestern North

America by the forest-associated American marten (Martes americana, Mammalia:

Carnivora: Mustelidae). (2002). Mol Ecol, 11(10): p. 2049-2063.

16. Hoffmann R.S., Koeppl J.W. & Nadler C.F. The relationships of the amphiberingian

marmots (Mammalia, Sciuridae). Vol. 83. (1979), University of Kansas: Museum of

Natural History 1-56.

17. Wilbur H.M. The Evolutionary and Mathematical Demography of the Turtle Chrysemys

picta. (1975). Ecology, 56(1): p. 64-77.

18. Gaffney E.S. & Jenkins F.A. The cranial morphology of Kayentachelys, an Early

Jurassic cryptodire, and the early history of turtles. (2010). Acta Zoologica, 91(3): p. 335-

368.

19. Ernst C.H. & Lovich J.E. Turtles of the United States and Canada. (2009): The John

Hopkins University Press.

20. Bleakney S. Postglacial dispersal of the turtle Chrysemys picta. (1958). Herpetologica,

14(2): p. 101-104.

21. Adams C.C. Southeastern United States as a center of geographical distribution of flora

and fauna. (1902). Biological Bulletin, 3(3): p. 115-131.

22. Brown A.E. Post-Glacial Nearctic Centres of Dispersal for Reptiles. (1904). Proceedings

of the Academy of Natural Sciences of Philadelphia, 56(2): p. 464-474.

23. Smith P.W. An Analysis of Post-Wisconsin Biogeography of the Prairie Peninsula

Region Based on Distributional Phenomena among Terrestrial Vertebrate Populations.

(1957). Ecology, 38(2): p. 205-218.

24. Starkey D.E., Shaffer H.B., Burke R.L., Forstner M.R.J., Iverson J.B., Janzen F.J.,

Rhodin A.G.J. & Ultsch G.R. Molecular systematics, phylogeography, and the effects

of Pleistocene glaciation in the painted turtle (Chrysemys picta) complex. (2003).

Evolution, 57(1): p. 119-128.

25. Crawford K.M. The winter environment of painted turtles, Chrysemys Picta -

temperature, dissolved-oxygen, and potential cues for emergence. (1991). Canadian

Journal of Zoology-Revue Canadienne De Zoologie, 69(9): p. 2493-2498.

26. Jackson D.C. & Ultsch G.R. Long-Term Submergence at 3-Degrees-C of the Turtle,

Chrysemys-Picta-Bellii, in Normoxic and Severely Hypoxic Water .2. Extracellular Ionic

Responses to Extreme Lactic-Acidosis. (1982). Journal of Experimental Biology,

96(Feb): p. 29-43.

27. Ultsch G.R., Hanley R.W. & Bauman T.R. Responses to Anoxia during Simulated

Hibernation in Northern and Southern Painted Turtles. (1985). Ecology, 66(2): p. 388-

395.

Page 102: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

87

28. Ultsch G.R., Carwile M.E., Crocker C.E. & Jackson D.C. The physiology of

hibernation among painted turtles: The eastern painted turtle Chrysemys picta picta.

(1999). Physiological and Biochemical Zoology, 72(4): p. 493-501.

29. Reese S.A., Crocker C.E., Jackson D.C. & Ultsch G.R. The physiology of hibernation

among painted turtles: the midland painted turtle (Chrysemys picta marginata). (2000).

Respiration Physiology, 124(1): p. 43-50.

30. Bickler P.E. & Buck L.T. Hypoxia tolerance in reptiles, amphibians, and fishes: life

with variable oxygen availability. (2007). Annual Review of Physiology, 69(1): p. 145-

170.

31. Reese S.A., Stewart E.R., Crocker C.E., Jackson D.C. & Ultsch G.R. Geographic

variation of the physiological response to overwintering in the painted turtle (Chrysemys

picta). (2004). Physiological and Biochemical Zoology, 77(4): p. 619-630.

32. Berkson H. Physiological adjustments to deep diving in the pacific green turtle

(Chelonia mydas agassizzii). (1967). Comparative Biochemistry and Physiology 21(3): p.

507–524.

33. Gatten R.E. Aerial and Aquatic Oxygen-Uptake by Freely-Diving Snapping Turtles

(Chelydra-Serpentina). (1980). Oecologia, 46(2): p. 266-271.

34. Lutz P.L., McMahon P., Rosenthal M. & Sick T.J. Relationships between aerobic and

anaerobic energy production in turtle brain in situ. (1984). American Journal of

Physiology - Regulatory, Integrative and Comparative Physiology, 247(4): p. R740-

R744.

35. Herbert C.V. & Jackson D.C. Temperature effects on the responses to prolonged

submergence in the turtle Chrysemys picta bellii. II. metabolic rate, blood acid-base and

ionic changes, and cardiovascular function in aerated and anoxic water. (1985).

Physiological Zoology, 58(6): p. 670-681.

36. Land S.C., Buck L.T. & Hochachka P.W. Response of protein synthesis to anoxia and

recovery in anoxia-tolerant hepatocytes. (1993). American Journal of Physiology -

Regulatory, Integrative and Comparative Physiology, 265(1): p. R41-R48.

37. Jackson D.C. Hibernating without oxygen: physiological adaptations of the painted

turtle. (2002). The Journal of Physiology, 543(3): p. 731-737.

38. Jackson D.C. Living without oxygen: lessons from the freshwater turtle. (2000).

Comparative Biochemistry and Physiology Part A: Molecular & Integrative Physiology,

125(3): p. 299-315.

39. Clark V.M. & Miller A.T. Studies on anaerobic metabolism in the fresh-water turtle

(Pseudemys scripta elegans). (1973). Comparative Biochemistry and Physiology Part A:

Physiology, 44(1): p. 55-62.

Page 103: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

88

40. Jackson D.C., Crocker C.E. & Ultsch G.R. Bone and shell contribution to lactic acid

buffering of submerged turtles Chrysemys picta bellii at 3°C. (2000). American Journal

of Physiology - Regulatory, Integrative and Comparative Physiology, 278(6): p. R1564-

R1571.

41. Kondo N., Sekijima T., Kondo J., Takamatsu N., Tohya K. & Ohtsu T. Circannual

Control of Hibernation by HP Complex in the Brain. (2006). Cell, 125(1): p. 161-172.

42. Wikipedia. Painted turtle. (2013) [cited July 27 2013]; Available from:

http://en.wikipedia.org/wiki/Painted_turtle.

43. Buck C.L. & Barnes B.M. Annual cycle of body composition and hibernation in free-

living arctic ground squirrels. (1999). Journal of Mammalogy, 80(2): p. 430-442.

44. Barnes B.M. Freeze avoidance in a mammal: body temperatures below 0 degree C in an

Arctic hibernator. (1989). Science, 244(4912): p. 1593-1595.

45. Eddingsaas A.A., Jacobsen B.K., Lessa E.P. & Cook J.A. Evolutionary history of the

arctic ground squirrel (Spermophilus parryii) in Nearctic Beringia. (2004). Journal of

Mammalogy, 85(4): p. 601-610.

46. Nadler C.F. & Hoffmann R.S. Patterns of evolution and migration in the arctic ground

squirrel, Spermophilus parryii (Richardson). (1977). Canadian Journal of Zoology.,

55(4): p. 748–758.

47. Hoffmann R.S. Different voles for different holes: environmental restrictions on refugial

survival of mammals. in: Evolution Today. (1981), Carnegie-Mellon University,

Pittsburgh, Pennsylvania.: Proceedings of the Second International Congress of

Systematic and Evolutionary Biology.

48. Hoffmann R.S. An ecological and zoogeographical analysis of animal migration across

the Bering Land Bridge during the Quaternary period. in: Beringia in the Cenozoic Era.

(1986), Rotterdam, The Netherlands.: A. A. Balkema.

49. Hopkins D.M. The Cenozoic history of Beringia—a synthesis. Pp. 451–484 in The

Bering Land Bridge (D. M. Hopkins, ed.). (1967), Stanford, California: Stanford

University Press.

50. Hopkins D.M., Matthews J.V., Schweger C.E. & B. Y.S. Paleoecology of Beringia:

Aspects of the paleogeography of Beringia during the late Pleistocene. (1982), New

York: Academic Press. 3–28.

51. Hulten E. Outline of the history of arctic and boreal biota during the quaternary period.

(1937), New York: Cramer.

52. Frerichs K.U., Dienel G.A., Cruz N.F., Sokoloff L. & Hallenbeck J.M. Rates of

glucose utilization in brain of active and hibernating ground squirrels. (1995). American

Journal of Physiology - Regulatory, Integrative and Comparative Physiology, 268(2): p.

R445-R453.

Page 104: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

89

53. Frerichs K.U., Smith C.B., Brenner M., DeGracia D.J., Krause G.S., Marrone L.,

Dever T.E. & Hallenbeck J.M. Suppression of protein synthesis in brain during

hibernation involves inhibition of protein initiation and elongation. (1998). Proceedings

of the National Academy of Sciences, 95(24): p. 14511-14516.

54. Frerichs K.U., Kennedy C., Sokoloff L. & Hallenbeck J.M. Local Cerebral Blood

Flow During Hibernation, a Model of Natural Tolerance to [ldquo]Cerebral

Ischemia[rdquo]. (1994). J Cereb Blood Flow Metab, 14(2): p. 193-205.

55. Willis J.S. Cold tolerance in mammalian cells. (1987). Symposia of the Society for

Experimental Biology, 41: p. 285-309.

56. Carey H.V., Andrews M.T. & Martin S.L. Mammalian Hibernation: Cellular and

Molecular Responses to Depressed Metabolism and Low Temperature. (2003).

Physiological Reviews, 83(4): p. 1153-1181.

57. Boonstra R., Bradley A. & Delehanty B. Not by Fat Alone: how to hibernate and

survive the deep freeze of an Arctic winter. (2011). Functional Ecology.

58. Galster W. & Morrison P. Seasonal changes in body composition of the arctic ground

squirrel, Citellus undulatus. (1976). Canadian Journal of Zoology, 54(1): p. 74-78.

59. Tenney S. & Boggs D. Comprehensive Physiology: Comparative Mammalian

Respiratory Control, 2011, John Wiley & Sons, Inc.

60. Andrews M.T., Squire T.L., Bowen C.M. & Rollins M.B. Low-temperature carbon

utilization is regulated by novel gene activity in the heart of a hibernating mammal.

(1998). Proceedings of the National Academy of Sciences of the United States of

America, 95(14): p. 8392-8397.

61. Mcarthur M.D. & Milsom W.K. Ventilation and Respiratory Sensitivity of Euthermic

Columbian and Golden-Mantled Ground-Squirrels (Spermophilus-Columbianus and

Spermophilus-Lateralis) during the Summer and Winter. (1991). Physiological Zoology,

64(4): p. 921-939.

62. Kuhnen G. O2 and CO2 concentrations in burrows of euthermic and hibernating golden

hamsters. (1986.). Com Biochem Physiol A, 84: p. 517–522.

63. Daan S., Barnes B.M. & Strijkstra A.M. Warming up for sleep? — Ground squirrels

sleep during arousals from hibernation. (1991). Neuroscience Letters, 128(2): p. 265-268.

64. Craig Heller H. & Poulson T.L. Circanian rhythms—II. Endogenous and exogenous

factors controlling reproduction and hibernation in chipmunks (Eutamias) and ground

squirrels (Spermophilus). (1970). Comparative Biochemistry and Physiology, 33(2): p.

357-383.

65. Pengelley E.T., Asmundson S.J., Barnes B. & Aloia R.C. Relationship of light

intensity and photoperiod to circannual rhythmicity in the hibernating ground squirrel,

Page 105: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

90

Citellus lateralis. (1976). Comparative Biochemistry and Physiology Part A: Physiology,

53(3): p. 273-277.

66. Lee T.M. & Zucker I. Suprachiasmatic Nucleus and Photic Entrainment of Circannual

Rhythms in Ground Squirrels. (1991). Journal of Biological Rhythms, 6(4): p. 315-330.

67. Raicu P., Taisescu E. & Banarescu P. Carassius carassius and C. auratus, a pair of

diploid and tetraploid rep- resentative species (Pisces, Cyprinidae). . (1981). Cytologia

46: p. 233–240.

68. Nilsson G.E. & Renshaw G.M.C. Hypoxic survival strategies in two fishes: extreme

anoxia tolerance in the North European crucian carp and natural hypoxic preconditioning

in a coral-reef shark. (2004). Journal of Experimental Biology, 207(18): p. 3131-3139.

69. Blazka P. The anaerobic metabolism of fish. (1958). Physiol Zool, 31((2)): p. 117-128.

70. Holopainen I.J., Tonn W.M. & Paszkowski C.A. Tales of two fish: The dichotomous

biology of crucian carp (Carassium carassius) (L.) in northern Europe. (1997). Annales

Zoologici Fennici, 34: p. 1-22.

71. Smith R.W., Houlihan D.F., Nilsson G.E. & Brechin J.G. Tissue-specific changes in

protein synthesis rates in vivo during anoxia in crucian carp. (1996). American Journal of

Physiology - Regulatory, Integrative and Comparative Physiology, 271(4): p. R897-

R904.

72. Johansson D., Nilsson G.E. & Tornnlom E. Effects of anoxia on energy metabolism in

crucian carp brain slices studied with microcalorimetry. (1995). Journal of Experimental

Biology, 198(3): p. 853-9.

73. Matikainen N. & Vornanen M. Effect of Season and Temperature Acclimation on the

Function of Crucian Carp (Carassius Carassius) Heart. (1992). Journal of Experimental

Biology, 167(1): p. 203-220.

74. Johansson D., Nilsson G.E. & Døving K.B. Anoxic depression of light-evoked

potentials in retina and optic tectum of crucian carp. (1997). Neuroscience Letters,

237(2–3): p. 73-76.

75. Hyvärinen H., Holopainen I.J. & Piironen J. Anaerobic wintering of crucian carp

(Carassius carassius L.)-I. Annual dynamics of glycogen reserves in nature. (1985).

Comparative Biochemistry and Physiology Part A: Physiology, 82(4): p. 797-803.

76. Sollid J., De Angelis P., Gundersen K. & Nilsson G.E. Hypoxia induces adaptive and

reversible gross morphological changes in crucian carp gills. (2003). Journal of

Experimental Biology, 206(20): p. 3667-3673.

77. Penáz M., Ráb P. & Prokes M. Cytological analysis, gynogenesis and early

development of Carassius auratus gibelio. (1979). Acta Sci. Nat. Brno 13(7): p. 1-33.

Page 106: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

91

78. van den Thillart G., Kesbeke F. & van Waarde A. Influence of anoxia on the energy

metabolism of goldfish Carassius auratus (L.). (1976). Comparative Biochemistry and

Physiology Part A: Physiology, 55(4): p. 329-336.

79. Hochachka P.W. Defense strategies against hypoxia and hypothermia. (1986). Science,

231(4735): p. 234-241.

80. Sollid J., Weber R.E. & Nilsson G.E. Temperature alters the respiratory surface area of

crucian carp Carassius carassius and goldfish Carassius auratus. (2005). Journal of

Experimental Biology, 208(6): p. 1109-1116.

81. Buck C.L. & Barnes B.M. Effects of ambient temperature on metabolic rate, respiratory

quotient, and torpor in an arctic hibernator. (2000). American Journal of Physiology -

Regulatory, Integrative and Comparative Physiology, 279(1): p. R255-R262.

82. Dowd W.W., Renshaw G.M.C., Cech J.J. & Kültz D. Compensatory proteome

adjustments imply tissue-specific structural and metabolic reorganization following

episodic hypoxia or anoxia in the epaulette shark (Hemiscyllium ocellatum). (2010).

Physiological Genomics, 42(1): p. 93-114.

83. Hansen A.J. Effect of anoxia on ion distribution in the brain. (1985). Physiological

Reviews, 65(1): p. 101-148.

84. Erecinska M. & Silver I.A. ATP and brain function. (1989). J. Cereb. Blood Flow 84: p.

745– 755.

85. Lutz P.L. Mechanisms for anoxic survival in the vertebrate brain. (1992). Annual Review

of Physiology, 54: p. 601-618.

86. Staples J.F. & Buck L.T. Matching cellular metabolic supply and demand in energy-

stressed animals. (2009). Comparative Biochemistry and Physiology Part A: Molecular

& Integrative Physiology, 153(2): p. 95-105.

87. Choi D.W. Excitotoxic cell death. (1992). Journal of Neurobiology, 23(9): p. 1261-1276.

88. Choi D.W. Calcium and excitotoxic neuronal injury. (1994). Annals of the New York

Academy of Sciences, 747(1): p. 162-171.

89. Miniño A.M., Murphy S.L., Xu J. & Kochanek K.D. Deaths: Final data for 2008.

(2011). National Vital Statistics Reports, 59(10).

90. Canada S. Canada Health Measures Survey – Cholesterol levels of Canadians, 2009-

2011. 2012.

91. Hakim A.M., Silver F. & Hodgson C. Organized stroke care: a new era in stroke

prevention and treatment. (1998). Canadian Medical Association Journal 159(6): p. S1.

92. Canada P.H.A.o. Tracking Heart Disease and Stroke in Canada, 2009. (2009).

Page 107: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

92

93. Murry C.E., Jennings R.B. & Reimer K.A. Preconditioning with Ischemia - a Delay of

Lethal Cell Injury in Ischemic Myocardium. (1986). Circulation, 74(5): p. 1124-1136.

94. Dawson V.L. & Dawson T.M. Neuronal ischaemic preconditioning. (2000). Trends in

pharmacological sciences, 21(11): p. 423-424.

95. Kitagawa K., Matsumoto M., Tagaya M., Hata R., Ueda H., Niinobe M., Handa N.,

Fukunaga R., Kimura K., Mikoshiba K. & Kamada T. ‘Ischemic tolerance’

phenomenon found in the brain. (1990). Brain Research, 528(1): p. 21-24.

96. Kobayashi S., Harris V.A. & Welsh F.A. Spreading Depression Induces Tolerance of

Cortical Neurons to Ischemia in Rat Brain. (1995). J Cereb Blood Flow Metab, 15(5): p.

721-727.

97. Heurteaux C., Lauritzen I., Widmann C. & Lazdunski M. Essential role of adenosine,

adenosine A1 receptors, and ATP-sensitive K+ channels in cerebral ischemic

preconditioning. (1995). Proceedings of the National Academy of Sciences of the United

States of America, 92(10): p. 4666-4670.

98. Dowden J. & Corbett D. Ischemic Preconditioning in 18- to 20-Month-Old Gerbils:

Long-Term Survival With Functional Outcome Measures. (1999). Stroke, 30(6): p. 1240-

1246.

99. Schaller B. & Graf R. Cerebral ischemic preconditioning. (2002). Journal of Neurology,

249(11): p. 1503-1511.

100. Pamenter M.E., Hogg D.W., Gu X.Q., Buck L.T. & Haddad G.G. Painted turtle

cortex is resistant to an in vitro mimic of the ischemic mammalian penumbra. (2012). J

Cereb Blood Flow Metab, 32(11): p. 2033-2043.

101. Connors B.W. & Kriegstein A.R. Cellular physiology of the turtle visual cortex:

distinctive properties of pyramidal and stellate neurons. (1986). The Journal of

Neuroscience, 6(1): p. 164-177.

102. Midtgaard J. Stellate cell inhibition of Purkinje cells in the turtle cerebellum in vitro.

(1992). The Journal of Physiology, 457(1): p. 355-367.

103. Humphrey O.D. On the brain of the snapping turtle, (Chelydra Serpentina). (1894).

Journal of Comparative Neurology, 4(2): p. 73-116.

104. Desan P.H. The organization of the cerebral cortex of the pond turtle, Pseudemys scripta

elegans. Ph.D. dissertation, ed. H. University. (1984), Cambridge, MA.

105. Hall J.A., Foster R.E., Ebner F.F. & Hall W.C. Visual cortex in a reptile, the turtle

(Pseudem ys scripta and Chrysem ys picta). (1977). Brain Research, 130(2): p. 197-216.

106. Smith L.M., Ebner F.F. & Colonnier M. The thalamocortical projection in Pseudemys

turtles: A quantitative electron microscopic study. (1980). The Journal of Comparative

Neurology, 190(3): p. 445-461.

Page 108: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

93

107. Northcutt R.G. The telencephalon of the western painted turtle (Chrysemys picta belli).

Illinois Biology Monographs. Vol. 43. (1970), Urbana, IL. 1-113.

108. Balaban C.D. Structure of the pallial thickening in turtles (Pseudemvs scripta eleaans).

(1978). Anat. Rec., 190: p. 330-33 1.

109. Lutz P.L. & Milton S.L. Negotiating brain anoxia survival in the turtle. (2004). Journal

of Experimental Biology, 207(18): p. 3141-3147.

110. Martyniuk C.J., Crawford A.B., Hogan N.S. & Trudeau V.L. GABAergic modulation

of the expression of genes involved in GABA synaptic transmission and stress in the

hypothalamus and telencephalon of the female goldfish (Carassius auratus). (2005).

Journal of Neuroendocrinology, 17(5): p. 269-275.

111. Turner A.J. & Whittle S.R. Biochemical dissection of the gamma-aminobutyrate

synapse. (1983). Biochem J., 209(1): p. 29-41.

112. Pamenter M.E., Hogg D.W., Ormond J., Shin D.S., Woodin M.A. & Buck L.T.

Endogenous GABAA and GABAB receptor-mediated electrical suppression is critical to

neuronal anoxia tolerance. (2011). Proceedings of the National Academy of Sciences.

113. Nilsson G.E. & Lutz P.L. Release of inhibitory neurotransmitters in response to anoxia

in turtle brain. (1991). American Journal of Physiology - Regulatory, Integrative and

Comparative Physiology, 261(1): p. R32-R37.

114. Lutz P.L. & Leone-Kabler S.L. Upregulation of the GABAA/benzodiazepine receptor

during anoxia in the freshwater turtle brain. (1995). American Journal of Physiology -

Regulatory, Integrative and Comparative Physiology, 268(5): p. R1332-R1335.

115. Fernandes J.A., Lutz P.L., Tannenbaum A., Todorov A.T., Liebovitch L. & Vertes

R. Electroencephalogram activity in the anoxic turtle brain. (1997). American Journal of

Physiology - Regulatory, Integrative and Comparative Physiology, 273(3): p. R911-

R919.

116. Pérez-Pinzón M.A., Rosenthal M., Lutz P.L. & Sick T.J. Anoxic survival of the

isolated cerebellum of the turtle Pseudemis scripta elegans. (1992). Journal of

Comparative Physiology B, 162(1): p. 68-73.

117. Perez-Pinzon M.A., Chan C.Y., Rosenthal M. & Sick T.J. Membrane and synaptic

activity during anoxia in the isolated turtle cerebellum. (1992). American Journal of

Physiology - Regulatory, Integrative and Comparative Physiology, 263(5): p. R1057-

R1063.

118. Sick T.J., Perez-Pinzon M.A., Lutz P.L. & Rosenthal M. Maintaining coupled

metabolism and membrane function in anoxic brain: A comparison between the turtle and

the rat. In P. W. Hochachka, P. L. Lutz, T. J. Sick, M. Rosenthal & G. Van den Thillart

(Eds.), Surviving Hypoxia: Mechanisms of Control and Adaptation. (1993), Boca Raton,

FL: CRC Press.

Page 109: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

94

119. Ryan T., Emes R., Grant S. & Komiyama N. Evolution of NMDA receptor

cytoplasmic interaction domains: implications for organisation of synaptic signalling

complexes. (2008). BMC Neuroscience, 9(1): p. 6.

120. Dalkara T., Erdemli G., Barun S. & Onur R. Glycine is required for NMDA receptor

activation: electrophysiological evidence from intact rat hippocampus. (1992). Brain

Research, 576(2): p. 197-202.

121. Arundine M. & Tymianski M. Molecular mechanisms of calcium-dependent

neurodegeneration in excitotoxicity. (2003). Cell Calcium, 34(4–5): p. 325-337.

122. Malenka R.C. & Nicoll R.A. NMDA-receptor-dependent synaptic plasticity: multiple

forms and mechanisms. (1993). Trends in Neurosciences, 16(12): p. 521-527.

123. Nilsson G.E. & Lutz P.L. Anoxia tolerant brains. (2004). Journal of Cerebral Blood

Flow and Metabolism, 24: p. 475-486.

124. Pamenter M.E., Shin D.S.-H. & Buck L.T. AMPA receptors undergo channel arrest in

the anoxic turtle cortex. (2008). American Journal of Physiology - Regulatory,

Integrative and Comparative Physiology, 294(2): p. R606-R613.

125. Zivkovic G. & Buck L.T. Regulation of AMPA receptor currents by mitochondrial

ATP-sensitive K+ channels in anoxic turtle neurons. (2010). Journal of Neurophysiology,

104(4): p. 1913-1922.

126. Pamenter M.E., Shin D.S.-H., Cooray M. & Buck L.T. Mitochondrial ATP-sensitive

K+ channels regulate NMDAR activity in the cortex of the anoxic western painted turtle.

(2008). The Journal of Physiology, 586(4): p. 1043-1058.

127. Shin D.S.H. & Buck L.T. Effect of Anoxia and Pharmacological Anoxia on Whole‐Cell

NMDA Receptor Currents in Cortical Neurons from the Western Painted Turtle. (2003).

Physiological and Biochemical Zoology, 76(1): p. 41-51.

128. Bickler P.E., Donohoe P.H. & Buck L.T. Hypoxia-induced silencing of NMDA

receptors in turtle neurons. (2000). The Journal of Neuroscience, 20(10): p. 3522-3528.

129. Shin D.S.-H., Wilkie M.P., Pamenter M.E. & Buck L.T. Calcium and protein

phosphatase 1/2A attenuate N-methyl-d-aspartate receptor activity in the anoxic turtle

cortex. (2005). Comparative Biochemistry and Physiology Part A: Molecular &

Integrative Physiology, 142(1): p. 50-57.

130. AnaesthesiaUK. NMDA receptor. (2005) [cited 2013 July 27]; Available from:

http://www.frca.co.uk/article.aspx?articleid=100515.

131. Chen P. Dr Philip Chen Research Profile. [cited 2013 July 27]; Available from:

http://pure.rhul.ac.uk/portal/en/persons/philip-chen%286d179072-30d1-4429-af9f-

d71de690f8f0%29.html.

Page 110: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

95

132. Latini S. & Pedata F. Adenosine in the central nervous system: release mechanisms and

extracellular concentrations. (2001). Journal of Neurochemistry, 79(3): p. 463-484.

133. Newby A.C. Adenosine and the concept of ‘retaliatory metabolites’. (1984). Trends in

Biochemical Sciences, 9(2): p. 42-44.

134. Dunwiddie T.V. & Masino S.A. The role and regulation of adenosine in the central

nervous system. (2001). Annual Review of Neuroscience, 24(1): p. 31-55.

135. Prince D.A. & Stevens C.F. Adenosine decreases neurotransmitter release at central

synapses. (1992). Proceedings of the National Academy of Sciences of the United States

of America, 89(18): p. 8586-8590.

136. Thompson J.W., Prentice H.M. & Lutz P.L. Regulation of extracellular glutamate

levels in the long-term anoxic turtle striatum: coordinated activity of glutamate

transporters, adenosine, K ATP + channels and GABA. (2007). Journal of Biomedical

Science, 14(6): p. 809-817.

137. Collis M.G. The vasodilator role of adenosine. (1989). Pharmacology and Therapeutics,

41: p. 143-162.

138. Buck L.T. Adenosine as a signal for ion channel arrest in anoxia-tolerant organisms.

(2004). Comparative Biochemistry and Physiology Part B: Biochemistry and Molecular

Biology, 139(3): p. 401-414.

139. Nilsson G.E. & Lutz P.L. Adenosine release in the anoxic turtle: a possible mechanism

for anoxic survival. (1992). Journal of Experimental Biology, 162: p. 345-351.

140. Nilsson G.E., Alfaro A.A. & Lutz P.L. Changes in turtle brain neurotransmitters and

related substances during anoxia. (1990). American Journal of Physiology - Regulatory,

Integrative and Comparative Physiology, 259(2): p. R376-R384.

141. Buck L.T. & Bickler P.E. Role of adenosine in NMDA receptor modulation in the

cerebral cortex of an anoxia-tolerant turtle (Chrysemys picta belli). (1995). Journal of

Experimental Biology, 198(7): p. 1621-8.

142. Buck L.T. & Bickler P.E. Adenosine and anoxia reduce N-methyl-D-aspartate receptor

open probability in turtle cerebrocortex. (1998). Journal of Experimental Biology, 201(2):

p. 289-97.

143. Seino S. ATP-sensitivie potassium channels: a model of heteromultimeric potassium

channel/receptor assemblies. (1999). Annual Review of Physiology, 61(1): p. 337-362.

144. Aguilar-Bryan L. & Bryan J. Molecular Biology of Adenosine Triphosphate-Sensitive

Potassium Channels. (1999). Endocrine Reviews, 20(2): p. 101-135.

145. Seharaseyon J., Ohler A., Sasaki N., Fraser H., Sato T., Johns D.C., O'Rourke B. &

Marbán E. Molecular composition of mitochondrial ATP-sensitive potassium channels

Page 111: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

96

probed by viral Kir gene transfer. (2000). Journal of Molecular and Cellular Cardiology,

32(11): p. 1923-1930.

146. Lutz P.L., Rosenthal M. & Sick T. Living without oxygen: turtle brain as a model of

anaerobic metabolism. (1985). Mol. Physiol, 8: p. 411–425.

147. Pek-Scott M. & Lutz P.L. ATP-sensitive K+ channel activation provides transient

protection to the anoxic turtle brain. (1998). American Journal of Physiology -

Regulatory, Integrative and Comparative Physiology, 275: p. 2023-2027.

148. Holmuhamedov E.L., Wang L. & Terzic A. ATP-sensitive K+ channel openers prevent

Ca2+ overload in rat cardiac mitochondria. (1999). The Journal of Physiology, 519(2): p.

347-360.

149. Hawrysh P. NMDA receptor silencing is mediated by calcium release from the

mitochondria via the permeability transition pore in anoxia-tolerant turtle neurons.

(2012). University Of Toronto Research Repository.

150. Hisatsune C., Umemori H., Inoue T., Michikawa T., Kohda K., Mikoshiba K. &

Yamamoto T. Phosphorylation-dependent regulation of N-Methyl-d-aspartate receptors

by calmodulin. (1997). Journal of Biological Chemistry, 272(33): p. 20805-20810.

151. Wang L.-Y., Orser B.A., Brautigan D.L. & MacDonald J.F. Regulation of NMDA

receptors in cultured hippocampal neurons by protein phosphatases 1 and 2A. (1994).

Nature, 369(6477): p. 230-232.

152. Mulkey R.M., Endo S., Shenolikar S. & Malenka R.C. Involvement of a calcineurin/

inhibitor-1 phosphatase cascade in hippocampal long-term depression. (1994). Nature,

369(6480): p. 486-488.

153. Wang J.H. & Kelly P.T. The balance between postsynaptic Ca(2+)-dependent protein

kinase and phosphatase activities controlling synaptic strength. (1996). Learning &

Memory, 3(2-3): p. 170-181.

154. Wang J.-H. & Kelly P.T. Postsynaptic calcineurin activity downregulates synaptic

transmission by weakening intracellular Ca2+ signaling mechanisms in hippocampal

CA1 neurons. (1997). The Journal of Neuroscience, 17(12): p. 4600-4611.

155. Chandel N.S. & Schumacker P.T. Cellular oxygen sensing by mitochondria: old

questions, new insight. (2000). Journal of Applied Physiology, 88(5): p. 1880-1889.

156. Pamenter M.E., Richards M.D. & Buck L.T. Anoxia-induced changes in reactive

oxygen species and cyclic nucleotides in the painted turtle. (2007). Journal of

Comparative Physiology B, 177(4): p. 473-481.

157. Liu Y., Fiskum G. & Schubert D. Generation of reactive oxygen species by the

mitochondrial electron transport chain. (2002). Journal of Neurochemistry, 80(5): p. 780-

787.

Page 112: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

97

158. Chen Q., Vazquez E.J., Moghaddas S., Hoppel C.L. & Lesnefsky E.J. Production of

reactive oxygen species by mitochondria: central role of complex III. (2003). Journal of

Biological Chemistry, 278(38): p. 36027-36031.

159. Boveris A. & Chance B. The mitochondrial generation of hydrogen peroxide. General

properties and the effect of hyperbaric oxygen. (1973). Biochemistry, 134(3): p. 707–716.

160. Chance B., Sies H. & Boveris A. Hydroperoxide Metabolism in Mammalian Organs.

(1979). Physiological Reviews, 59(3): p. 527-605.

161. Fahn S., Davies J.N. & Rowland L.P. Advances in Neurology: Cerebral hypoxia and its

consequences. Vol. 26. (1979), New York: Raven Press.

162. Gerich F.J., Funke F., Hildebrandt B., Fasshauer M. & Muller M. H2O2-mediated

modulation of cytosolic signaling and organelle function in rat hippocampus. (2009).

Pflugers Archiv-European Journal of Physiology, 458(5): p. 937-952.

163. D'Autreaux B. & Toledano M.B. ROS as signalling molecules: mechanisms that

generate specificity in ROS homeostasis. (2007). Nature Reviews Molecular Cell

Biology, 8(10): p. 813-824.

164. Turrens J.F., Beconi M., Barilla J., Chavez U.B. & Mccord J.M. Mitochondrial

Generation of Oxygen Radicals during Reoxygenation of Ischemic Tissues. (1991). Free

Radical Research Communications, 12-3: p. 681-689.

165. Szeto H.H. Cell-Permeable, Mitochondrial-Targeted, Peptide Antioxidants, in Drug

Addiction, R. Rapaka and W. Sadée, Editors. 2008, Springer New York. p. 535-546.

166. Cross J.V. & Templeton D.J. Regulation of signal transduction through protein cysteine

oxidation. (2006). Antioxid Redox Signal 8: p. 1819–1827.

167. Rhee S.G., Chang T.-S., Bae Y.S., Lee S.-R. & Kang S.W. Cellular regulation by

hydrogen peroxide. (2003). Journal of the American Society of Nephrology, 14(suppl 3):

p. S211-S215.

168. Ottaviano F.G., Handy D.E. & Loscalzo J. Redox Regulation in the Extracellular

Environment. (2008). Circulation Journal, 72(1): p. 1-16.

169. Bienert G.P., Møller A.L.B., Kristiansen K.A., Schulz A., Møller I.M., Schjoerring

J.K. & Jahn T.P. Specific Aquaporins Facilitate the Diffusion of Hydrogen Peroxide

across Membranes. (2007). Journal of Biological Chemistry, 282(2): p. 1183-1192.

170. Henzler T. & Steudle E. Transport and metabolic degradation of hydrogen peroxide in

Chara corallina: model calculations and measurements with the pressure probe suggest

transport of H2O2 across water channels. (2000). Journal of Experimental Botany,

51(353): p. 2053-2066.

Page 113: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

98

171. Hooijmaijers C., Rhee J.Y., Kwak K.J., Chung G.C., Horie T., Katsuhara M. &

Kang H. Hydrogen peroxide permeability of plasma membrane aquaporins of

Arabidopsis thaliana. (2012). Journal of Plant Research, 125(1): p. 147-153.

172. MacAulay N., Hamann S. & Zeuthen T. Water transport in the brain: Role of

cotransporters. (2004). Neuroscience, 129(4): p. 1029-1042.

173. Andrew R.A., Mark W. Labron M.W., Boehnke S.E., Carnduff L. & Kirov S.A.

Physiological evidence that pyramidal neurons lack functional water channels. (2007).

Cerebral Cortex, 17: p. 787--802.

174. Mollajew R., Zocher F., Horner A., Wiesner B., Klussmann E. & Pohl P. Routes of

Epithelial Water Flow: Aquaporins versus Cotransporters. (2010). Biophysical Journal,

99(11): p. 3647-3656.

175. Christopherson K.S., Hillier B.J., Lim W.A. & Bredt D.S. PSD-95 assembles a ternary

complex with the N-Methyl-D-aspartic acid receptor and a bivalent neuronal NO

synthase PDZ domain. (1999). Journal of Biological Chemistry, 274(39): p. 27467-

27473.

176. Garthwaite J. Concepts of neural nitric oxide-mediated transmission. (2008). European

Journal of Neuroscience, 27(11): p. 2783-2802.

177. Chandler L.J., Guzman N.J., Sumners C. & Crews F.T. Magnesium and zinc

potentiate ethanol inhibition of N-methyl-D-aspartate-stimulated nitric oxide synthase in

cortical neurons. (1994). Journal of Pharmacology and Experimental Therapeutics,

271(1): p. 67-75.

178. Kawamata T. & Omote K. Activation of spinal N-methyl-D-aspartate receptors

stimulates a nitric oxide/cyclic guanosine 3,5-monophosphate/glutamate release cascade

in nociceptive signaling. (1999). Anesthesiology, 91(5): p. 1415-1424.

179. Nakamura T. & Lipton S.A. S-Nitrosylation and uncompetitive/fast off-rate (UFO)

drug therapy in neurodegenerative disorders of protein misfolding. (2007). Cell Death

Differ, 14(7): p. 1305-1314.

180. Herrera M., Hong N.J. & Garvin J.L. Aquaporin-1 Transports NO Across Cell

Membranes. (2006). Hypertension, 48(1): p. 157-164.

181. Girouard H., Wang G., Gallo E.F., Anrather J., Zhou P., Pickel V.M. & Iadecola C.

NMDA receptor activation increases free radical production through nitric oxide and

NOX2. (2009). The Journal of Neuroscience, 29(8): p. 2545-2552.

182. Kishida K.T. & Klann E. Sources and targets of reactive oxygen species in synaptic

plasticity and memory. (2007). Antioxid Redox Signal 9: p. 233-244.

183. Thannickal V.J. & Fanburg B.L. Reactive oxygen species in cell signaling. (2000).

American Journal of Physiology - Lung Cellular and Molecular Physiology, 279(6): p.

L1005-L1028.

Page 114: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

99

184. Hancock J.T., Desikan R. & Neill S.J. Role of reactive oxygen species in cell signalling

pathways. (2001). Biochem Soc Trans., Pt 2:: p. 345-350.

185. Gozlan H. & Ben-Ari Y. NMDA receptor redox sites: are they targets for selective

neuronal protection? (1995). Trends in Pharmacological Sciences, 16(11): p. 368-374.

186. Brimecombe J.C., Potthoff W.K. & Aizenman E. A Critical Role of theN-Methyl-d-

aspartate (NMDA) Receptor Subunit (NR) 2A in the Expression of Redox Sensitivity of

NR1/NR2A Recombinant NMDA Receptors. (1999). Journal of Pharmacology and

Experimental Therapeutics, 291(2): p. 785-792.

187. Sullivan J.M., Traynelis S.F., Chen H.-S.V., Escobar W., Heinemann S.F. & Lipton

S.A. Identification of two cysteine residues that are required for redox modulation of the

NMDA subtype of glutamate receptor. (1994). Neuron, 13(4): p. 929-936.

188. Choi Y.B., Chen H.S.V. & Lipton S.A. Three pairs of cysteine residues mediate both

redox and Zn2+ modulation of the NMDA receptor. (2001). Journal of Neuroscience,

21(6): p. 392.

189. Kim W.-K., Choi Y.-B., Rayudu P.V., Das P., Asaad W., Arnelle D.R., Stamler J.S.

& Lipton S.A. Attenuation of NMDA Receptor Activity and Neurotoxicity by Nitroxyl

Anion, NO−. (1999). Neuron, 24(2): p. 461-469.

190. Aizenman E., Lipton S.A. & Loring R.H. Selective modulation of NMDA responses by

reduction and oxidation. (1989). Neuron, 2(3): p. 1257-1263.

191. Aizenman E., Hartnett K.A. & Reynoldst I.J. Oxygen free radicals regulate NMDA

receptor function via a redox modulatory site. (1990). Neuron, 5(6): p. 841-846.

192. Tauck D.L. Redox Modulation of Nmda Receptor-Mediated Synaptic Activity in the

Hippocampus. (1992). Neuroreport, 3(9): p. 781-784.

193. Palmer A.M. & Burns M.A. Preservation of redox, polyamine, and glycine domains of

the N-Methyl-D-aspartate in alzheimer's disease. (1994). Journal of Neurochemistry,

62(1): p. 187-196.

194. Woodward J.J. & Blair R. Redox Modulation of N-Methyl-D-Aspartate-Stimulated

Neurotransmitter Release from Rat Brain Slices. (1991). Journal of Neurochemistry,

57(6): p. 2059-2064.

195. Janáky R., Varga V., Saransaari P. & Oja S.S. Glutathione modulates the N-methyl-d-

aspartate receptor-activated calcium influx into cultured rat cerebellar granule cells.

(1993). Neuroscience Letters, 156(1–2): p. 153-157.

196. Bodhinathan K., Kumar A. & Foster T.C. Intracellular Redox State Alters NMDA

Receptor Response during Aging through Ca2+/Calmodulin-Dependent Protein Kinase

II. (2010). Journal of Neuroscience, 30(5): p. 1914-1924.

Page 115: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

100

197. Gozlan H., Khazipov R., Diabira D. & Benari Y. In CA1 hippocampal-neurons, the

redox state of NMDA receptors determines LTP expressed by NMDA but not by AMPA

receptors. (1995). Journal of Neurophysiology, 73(6): p. 2612-2617.

198. Gozlan H., Chinestra P., Diabira D. & Ben-Ari Y. NMDA redox site modulates long-

term potentiation of NMDA but not of AMPA receptors. (1994). European Journal of

Pharmacology, 262(1–2): p. R3-R4.

199. Choi Y.B. & Lipton S.A. Redox modulation of the NMDA receptor. (2000). Cellular

and Molecular Life Sciences, 57(11): p. 1535-1541.

200. Cino M. & Del Maestro R.F. Generation of hydrogen peroxide by brain mitochondria:

the effect of reoxygenation following postdecapitative ischemia. (1989). Archives of

Biochemistry and Biophysics, 269(2): p. 623-638.

201. Liu Y., Rosenthal R.E. & Fiskum G. H2O2 release by isolated brain mitochondria in a

canine cerebral ischemia/reperfusion model. (1993). Faseb Journal, 7: p. A424.

202. Li Y. & Trush M.A. Diphenyleneiodonium, an NAD(P)H Oxidase Inhibitor, also

Potently Inhibits Mitochondrial Reactive Oxygen Species Production. (1998).

Biochemical and Biophysical Research Communications, 253(2): p. 295-299.

203. Wu Y.N. & Johnson S.W. Rotenone potentiates NMDA currents in substantia nigra

dopamine neurons. (2007). Neuroscience Letters, 421(2): p. 96-100.

204. Lei S.Z., Pan Z.-H., Aggarwal S.K., Chen H.-S.V., Hartman J., Sucher N.J. &

Lipton S.A. Effect of nitric oxide production on the redox modulatory site of the NMDA

receptor-channel complex. (1992). Neuron, 8(6): p. 1087-1099.

205. Lipton S.A., Choi Y.-B., Pan Z.-H., Lei S.Z., Chen H.-S.V., Sucher N.J., Loscalzo J.,

Singel D.J. & Stamler J.S. A redox-based mechanism for the neuroprotective and

neurodestructive effects of nitric oxide and related nitroso-compounds. (1993). Nature,

364(6438): p. 626-632.

206. Wu Y.N. & Johnson S.W. Rotenone reduces Mg2+-dependent block of NMDA currents

in substantia nigra dopamine neurons. (2009). Neurotoxicology, 30(2): p. 320-325.

207. Fryer R.M., Eells J.T., Hsu A.K., Henry M.M. & Gross G.J. Ischemic preconditioning

in rats: role of mitochondrial KATP channel in preservation of mitochondrial function.

(2000). American Journal of Physiology - Heart and Circulatory Physiology, 278(1): p.

H305-H312.

208. Gross G.J. & Fryer R.M. Sarcolemmal versus mitochondrial ATP-sensitive K+

channels and myocardial preconditioning. (1999). Circulation Research, 84(9): p. 973-

979.

209. Ockaili R., Emani V.R., Okubo S., Brown M., Krottapalli K. & Kukreja R.C.

Opening of mitochondrial KATPchannel induces early and delayed cardioprotective

Page 116: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

101

effect: role of nitric oxide. (1999). American Journal of Physiology - Heart and

Circulatory Physiology, 277(6): p. H2425-H2434.

210. Shimizu K., Lacza Z., Rajapakse N., Horiguchi T., Snipes J. & Busija D.W.

MitoKATP opener, diazoxide, reduces neuronal damage after middle cerebral artery

occlusion in the rat. (2002). American Journal of Physiology - Heart and Circulatory

Physiology, 283(3): p. H1005-H1011.

211. Domoki F., Perciaccante J.V., Veltkamp R., Bari F. & Busija D.W. Mitochondrial

Potassium Channel Opener Diazoxide Preserves Neuronal-Vascular Function After

Cerebral Ischemia in Newborn Pigs. (1999). Stroke, 30(12): p. 2713-2719.

212. Garcia de Arriba S., Franke H., Pissarek M., Nieber K. & Illes P. Neuroprotection by

ATP-dependent potassium channels in rat neocortical brain slices during hypoxia. (1999).

Neuroscience Letters, 273(1): p. 13-16.

213. Wang L., Zhu Q.-L., Wang G.-Z., Deng T.-Z., Chen R., Liu M.-H. & Wang S.-W.

The protective roles of mitochondrial ATP-sensitive potassium channels during hypoxia–

ischemia–reperfusion in brain. (2011). Neuroscience Letters, 491(1): p. 63-67.

214. Liang H.-W., Xia Q. & Bruce I.C. Reactive oxygen species mediate the neuroprotection

conferred by a mitochondrial ATP-sensitive potassium channel opener during ischemia in

the rat hippocampal slice. (2005). Brain Research, 1042(2): p. 169-175.

215. Pain T., Yang X.-M., Critz S.D., Yue Y., Nakano A., Liu G.S., Heusch G., Cohen

M.V. & Downey J.M. Opening of mitochondrial KATP channels triggers the

preconditioned state by generating free radicals. (2000). Circulation Research, 87(6): p.

460-466.

216. Carroll R., Gant V.A. & Yellon D.M. Mitochondrial KATP channel opening protects a

human atrial-derived cell line by a mechanism involving free radical generation. (2001).

Cardiovascular Research, 51(4): p. 691-700.

217. Tanaka K., Weihrauch D., Ludwig L.M., Kersten J.R., Pagel P.S. & Warltier D.C.

Mitochondrial Adenosine Triphosphate–regulated Potassium Channel Opening Acts as a

Trigger for Isoflurane-induced Preconditioning by Generating Reactive Oxygen Species.

(2003). Anesthesiology, 98(4): p. 935-943.

218. Roth S., Dreixler J.C., Shaikh A.R., Lee K.H. & Bindokas V. Mitochondrial

potassium ATP channels and retinal ischemic preconditioning. (2006). Investigative

Ophthalmology & Visual Science, 47(5): p. 2114-2124.

219. Badziuk O.B., Mazur I. & Kosterin S.O. Regulation of the mitochondrial ATP-

sensitive potassium channel in rat uterus cells by ROS. (2011). Ukr Biokhim Zh., 83(3):

p. 48-57.

220. Zhang D.X., Chen Y.-F., Campbell W.B., Zou A.-P., Gross G.J. & Li P.-L.

Characteristics and Superoxide-Induced Activation of Reconstituted Myocardial

Page 117: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

102

Mitochondrial ATP-Sensitive Potassium Channels. (2001). Circulation Research, 89(12):

p. 1177-1183.

221. Lebuffe G., Schumacker P.T., Shao Z.-H., Anderson T., Iwase H. & Vanden Hoek

T.L. ROS and NO trigger early preconditioning: relationship to mitochondrial KATP

channel. (2003). American Journal of Physiology - Heart and Circulatory Physiology,

284(1): p. H299-H308.

222. Sasaki N., Sato T., Ohler A., O’Rourke B. & Marbán E. Activation of mitochondrial

ATP-dependent potassium channels by nitric oxide. (2000). Circulation, 101(4): p. 439-

445.

223. Kawano T., Zoga V., Kimura M., Liang M.-Y., Wu H.-E., Gemes G., McCallum

J.B., Kwok W.-M., Hogan Q. & Sarantopoulos C. Nitric oxide activates ATP-sensitive

potassium channels in mammalian sensory neurons: action by direct S-nitrosylation.

(2009). Molecular Pain, 5(1): p. 12.

224. Ljubkovic M., Shi Y., Cheng Q., Bosnjak Z. & Jiang M.T. Cardiac mitochondrial

ATP-sensitive potassium channel is activated by nitric oxide in vitro. (2007). FEBS

Letters, 581(22): p. 4255-4259.

225. Halliwell B. & Gutteridge J.M.C. Free radicals in biology and medicine (2nd edn)

(1989), Oxford: Clarendon Press.

226. Sies H. Strategies of antioxidant defense. (1993). European Journal of Biochemistry,

215(2): p. 213-219.

227. Blokhina O., Virolainen E. & Fagerstedt K.V. Antioxidants, Oxidative Damage and

Oxygen Deprivation Stress: a Review. (2003). Annals of Botany, 91(2): p. 179-194.

228. Bowler C., Montagu M.V. & Inze D. Superoxide Dismutase and Stress Tolerance.

(1992). Annual Review of Plant Physiology and Plant Molecular Biology, 43(1): p. 83-

116.

229. Jones D.P., Carlson J.L., Mody Jr V.C., Cai J., Lynn M.J. & Sternberg Jr P. Redox

state of glutathione in human plasma. (2000). Free Radical Biology and Medicine, 28(4):

p. 625-635.

230. Ziegler D.M. ROLE OF REVERSIBLE OXIDATION-REDUCTION OF ENZYME

THIOLS-DISULFIDES IN METABOLIC-REGULATION. (1985). Annual Review of

Biochemistry, 54: p. 305-329.

231. Hogg D., Pamenter M.E., Dukoff D. & Buck L. Anoxic decreases in mitochondrial

reactive oxygen species generation initiates GABA receptor-mediated electrical

suppression in painted turtle cerebral cortex. (2013). Journal of Neuroscience (submitted

pending acceptance).

232. ThinkQuest. Turtle Anatomy. [cited 2013. August 14.]; Available from:

http://library.thinkquest.org/J0112124/turtle_anatomy.htm.

Page 118: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

103

233. Wittmann M., Queisser G., Eder A., Wiegert J.S., Bengtson C.P., Hellwig A.,

Wittum G. & Bading H. Synaptic Activity Induces Dramatic Changes in the Geometry

of the Cell Nucleus: Interplay between Nuclear Structure, Histone H3 Phosphorylation,

and Nuclear Calcium Signaling. (2009). The Journal of Neuroscience, 29(47): p. 14687-

14700.

234. Pamenter M.E., Shin D.S.H. & Buck L.T. AMPA receptors undergo channel arrest in

the anoxic turtle cortex. (2008). Am. J. Physiol. Regul. Integr. Comp. Physiol., 294(2): p.

R606-R613.

235. Dunn R.J., Bottai D. & Maler L. Molecular biology of the apteronotus NMDA receptor

NR1 subunit. (1999). Journal of Experimental Biology, 202(10): p. 1319-1326.

236. Paoletti P. & Neyton J. NMDA receptor subunits: function and pharmacology. (2007).

Current Opinion in Pharmacology, 7(1): p. 39-47.

237. Köhr G., Eckardt S., Lüddens H., Monyer H. & Seeburg P.H. NMDA receptor

channels: Subunit-specific potentiation by reducing agents. (1994). Neuron, 12(5): p.

1031-1040.

238. Dugan L.L., Sensi S.L., Canzoniero L.M., Handran S.D., Rothman S.M., Lin T.S.,

Goldberg M.P. & Choi D.W. Mitochondrial production of reactive oxygen species in

cortical neurons following exposure to N-methyl-D-aspartate. (1995). The Journal of

Neuroscience, 15(10): p. 6377-6388.

239. Duan Y., Gross R.A. & Sheu S.-S. Ca2+-dependent generation of mitochondrial

reactive oxygen species serves as a signal for poly(ADP-ribose) polymerase-1 activation

during glutamate excitotoxicity. (2007). The Journal of Physiology, 585(3): p. 741-758.

240. Cooper G.M. & Sunderland M.A. The Cell: A Molecular Approach. (2000): Sinauer

Associates.

241. Young K.W., Bampton E.T.W., Pinòn L., Bano D. & Nicotera P. Mitochondrial Ca2+

signalling in hippocampal neurons. (2008). Cell Calcium, 43(3): p. 296-306.

242. O-Uchi J., Pan S. & Sheu S.-S. Molecular identities of mitochondrial Ca2+ influx

mechanism: Updated passwords for accessing mitochondrial Ca2+-linked health and

disease. (2012). The Journal of General Physiology, 139(6): p. 435-443.

243. Kirichok Y., Krapivinsky G. & Clapham D.E. The mitochondrial calcium uniporter is

a highly selective ion channel. (2004). Nature, 427(6972): p. 360-364.

244. Ott M., Robertson J.D., Gogvadze V., Zhivotovsky B. & Orrenius S. Cytochrome c

release from mitochondria proceeds by a two-step process. (2002). Proceedings of the

National Academy of Sciences, 99(3): p. 1259-1263.

245. Palty R., Silverman W.F., Hershfinkel M., Caporale T., Sensi S.L., Parnis J., Nolte

C., Fishman D., Shoshan-Barmatz V., Herrmann S., Khananshvili D. & Sekler I.

Page 119: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

104

NCLX is an essential component of mitochondrial Na+/Ca2+ exchange. (2010).

Proceedings of the National Academy of Sciences, 107(1): p. 436-441.

246. Montero M., Alonso M.T., Carnicero E., Cuchillo-Ibanez I., Albillos A., Garcia

A.G., Garcia-Sancho J. & Alvarez J. Chromaffin-cell stimulation triggers fast

millimolar mitochondrial Ca2+ transients that modulate secretion. (2000). Nat Cell Biol,

2(2): p. 57-61.

247. Lemasters J.J., Theruvath T.P., Zhong Z. & Nieminen A.-L. Mitochondrial calcium

and the permeability transition in cell death. (2009). Biochimica et Biophysica Acta

(BBA) - Bioenergetics, 1787(11): p. 1395-1401.

248. Peng T.-I. & Jou M.-J. Oxidative stress caused by mitochondrial calcium overload.

(2010). Annals of the New York Academy of Sciences, 1201(1): p. 183-188.

249. Cohen G. Enzymatic/nonenzymatic sources of oxyradicals and regulation of antioxidant

defenses. (1994). Annals of the New York Academy of Sciences, 738(1): p. 8-14.

250. Danysz W. & Parsons C.G. Glycine and N-Methyl-d-Aspartate Receptors:

Physiological Significance and Possible Therapeutic Applications. (1998).

Pharmacological Reviews, 50(4): p. 597-664.

251. Das S., Sasaki Y.F., Rothe T., Premkumar L.S., Takasu M., Crandall J.E., Dikkes

P., Conner D.A., Rayudu P.V., Cheung W., Chen H.S.V., Lipton S.A. & Nakanishi

N. Increased NMDA current and spine density in mice lacking the NMDA receptor

subunit NR3A. (1998). Nature, 393(6683): p. 377-381.

252. Ly C.V. & Verstreken P. Mitochondria at the Synapse. (2006). The Neuroscientist,

12(4): p. 291-299.

253. Peng T.-I. & Greenamyre J.T. Privileged access to mitochondria of calcium influx

through N-Methyl-d-aspartate receptors. (1998). Molecular Pharmacology, 53(6): p. 974-

980.

254. Baron K.T., Wang G.J., Padua R.A., Campbell C. & Thayer S.A. NMDA-evoked

consumption and recovery of mitochondrially targeted aequorin suggests increased Ca2+

uptake by a subset of mitochondria in hippocampal neurons. (2003). Brain Research,

993(1–2): p. 124-132.

255. Rizzuto R., Brini M., Murgia M. & Pozzan T. Microdomains with high Ca2+ close to

IP3-sensitive channels that are sensed by neighboring mitochondria. (1993). Science,

262(5134): p. 744-747.

256. Filippin L., Magalhães P.J., Di Benedetto G., Colella M. & Pozzan T. Stable

Interactions between Mitochondria and Endoplasmic Reticulum Allow Rapid

Accumulation of Calcium in a Subpopulation of Mitochondria. (2003). Journal of

Biological Chemistry, 278(40): p. 39224-39234.

Page 120: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

105

257. Rizzuto R., Pinton P., Carrington W., Fay F.S., Fogarty K.E., Lifshitz L.M., Tuft

R.A. & Pozzan T. Close contacts with the endoplasmic reticulum as determinants of

mitochondrial Ca2+ responses. (1998). Science, 280(5370): p. 1763-1766.

258. Perocchi F., Gohil V.M., Girgis H.S., Bao X.R., McCombs J.E., Palmer A.E. &

Mootha V.K. MICU1 encodes a mitochondrial EF hand protein required for Ca2+

uptake. (2010). Nature, 467(7313): p. 291-296.

259. Collins S. & Meyer T. Cell biology: a sensor for calcium uptake. (2010). Nature,

467(7313): p. 283-283.

260. Cull-Candy S., Brickley S. & Farrant M. NMDA receptor subunits: diversity,

development and disease. (2001). Current Opinion in Neurobiology, 11(3): p. 327-335.

261. Dingledine R., Borges K., Bowie D. & Traynelis S.F. The Glutamate Receptor Ion

Channels. (1999). Pharmacological Reviews, 51(1): p. 7-62.

262. Conti F. & Weinberg R.J. Shaping excitation at glutamatergic synapses. (1999). Trends

in Neurosciences, 22(10): p. 451-458.

263. Nowak L., Bregestovski P., Ascher P., Herbet A. & Prochiantz A. Magnesium gates

glutamate-activated channels in mouse central neurones. (1984). Nature, 307(5950): p.

462-465.

264. Shin D.S., Wilkie M.P., Pamenter M.E. & Buck L.T. Calcium and protein phosphatase

1/2A attentuate N-methyl-D-aspartate receptor activity in the anoxic turtle cortex. (2005).

Comparative Biochemistry and Physiology, 142: p. 50-57.

265. Lin Y.M. Calmodulin. (1982). Molecular and Cellular Biochemistry, 45(2): p. 101-112.

266. Johnson D.J. & Mills J.S. Calmodulin. (1986). Medicinal Research Reviews, 6(3): p.

341-363.

267. Krupp J.J., Vissel B., Thomas C.G., Heinemann S.F. & Westbrook G.L. Calcineurin

acts via the C-terminus of NR2A to modulate desensitization of NMDA receptors.

(2002). Neuropharmacology, 42(5): p. 593-602.

268. Ehlers M.D., Zhang S., Bernhardt J.P. & Huganir R.L. Inactivation of NMDA

Receptors by Direct Interaction of Calmodulin with the NR1 Subunit. (1996). Cell, 84(5):

p. 745-755.

269. Zhang S., Ehlers M.D., Bernhardt J.P., Su C.-T. & Huganir R.L. Calmodulin

Mediates Calcium-Dependent Inactivation of N-Methyl-D-Aspartate Receptors. (1998).

Neuron, 21(2): p. 443-453.

270. Tong G., Shepherd D. & Jahr C.E. Synaptic desensitization of NMDA receptors by

calcineurin. (1995). Science, 267(5203): p. 1510-1512.

Page 121: The impact of ROS scavenging on NMDA and AMPA receptor … · 2013-11-22 · I would like to thank my little brother Andrew for sending me music on lonely nights at the lab. I would

106

271. He K., Song L., Cummings L.W., Goldman J., Huganir R.L. & Lee H.-K.

Stabilization of Ca2+-permeable AMPA receptors at perisynaptic sites by GluR1-S845

phosphorylation. (2009). Proceedings of the National Academy of Sciences, 106(47): p.

20033-20038.

272. Esteban J.A., Shi S.-H., Wilson C., Nuriya M., Huganir R.L. & Malinow R. PKA

phosphorylation of AMPA receptor subunits controls synaptic trafficking underlying

plasticity. (2003). Nat Neurosci, 6(2): p. 136-143.

273. Snyder G.L., Galdi S., Fienberg A.A., Allen P., Nairn A.C. & Greengard P.

Regulation of AMPA receptor dephosphorylation by glutamate receptor agonists. (2003).

Neuropharmacology, 45(6): p. 703-713.

274. Murchison D. & Griffith W.H. Mitochondria buffer non-toxic calcium loads and

release calcium through the mitochondrial permeability transition pore and

sodium/calcium exchanger in rat basal forebrain neurons. (2000). Brain Research, 854(1–

2): p. 139-151.

275. Ichas F. & Mazat J.-P. From calcium signaling to cell death: two conformations for the

mitochondrial permeability transition pore. Switching from low- to high-conductance

state. (1998). Biochimica et Biophysica Acta (BBA) - Bioenergetics, 1366(1–2): p. 33-50.

276. Hüser J. & Blatter L.A. Fluctuations in mitochondrial membrane potential caused by

repetitive gating of the permeability transition pore. (1999). Biochem J., 343(2): p. 311-

317.

277. Shields M., Carroll M.D. & Ogden C.L. Adult obesity prevalence in Canada and the

United States. (2011). NCHS Data Brief, 56: p. 1-8.