the effects of oxygen glucose deprivation and trpm7 ... · the effects of oxygen glucose...

130
The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity by Ervis Kola A thesis submitted in conformity with the requirements for the degree of Master of Science Graduate Department of Cell and Systems Biology University of Toronto © Copyright by Ervis Kola (2013)

Upload: others

Post on 30-May-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase

Activity

by

Ervis Kola

A thesis submitted in conformity with the requirements

for the degree of Master of Science

Graduate Department of Cell and Systems Biology

University of Toronto

© Copyright by Ervis Kola (2013)

Page 2: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

ii  

The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

Ervis Kola

Master of Science

Department of Cell and Systems Biology

University of Toronto

2013

Abstract

Transient Receptor Potential Melastatin 7 (TRPM7) is a ubiquitously expressed divalent cation

channel implicated as a key regulator of neuronal cell death in stroke. Our research group has

previously shown that TRPM7 dependent cytoskeletal regulation particularly via cofilin mediates

neuronal death in oxygen glucose deprivation (in vitro stroke model). LIMK1 phosphorylation

was also shown to decrease downstream of TRPM7 activation during anoxia. In the present

study we investigated the effects of TRPM7 activation during anoxia, on three regulators of

LIMK and cofilin; Rho-associated kinase 2 (ROCK2), P-21 activated kinase 3 (PAK3) and

Slingshot family phosphatase 1 (SSH1). Our findings suggest that PAK3 activity is

downregulated during OGD through TRPM7 independent mechanisms. However, SSH1 activity

appears to be regulated downstream of TRPM7 in a manner that is consistent with LIMK and

cofilin regulation. Overall, our work suggests that SSH1 is a new link between anoxia-induced

TRPM7activity and cofilin hyperactivation.

Page 3: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

iii  

Acknowledgments

I would firstly like to thank my graduate supervisor Dr. Michelle Aarts for giving me the

opportunity to pursue a Master’s Degree at the University of Toronto Scarborough. Dr. Aarts

provided a comfortable and supportive environment and for this I am very grateful. I would also

like to thank my committee members Dr. Zhengping Jia and Dr. Rongmin Zhao for their

constructive criticism and advice throughout my graduate degree.

I am very grateful for current and past members of the Aarts lab for their support and technical

advice including Russel Bent, Colin Seepersad, Suhail Asrar, Jonathan Chan, Nagmeh Lesani,

Anjali Anne Ajit, and Kareem Younis. Special thanks go to Melanie Ratnam, who has been an

amazing friend and source of advice and encouragement especially during challenging times, I

will miss you dearly. I am also indebt to everyone in the surrounding research groups for their

support, conversation and technical expertise. Thanks go to Kristin Lizal, Darren Gigliozzi, Chris

Young-Kee, Sherri Thiele, the entire Brown, Terebiznik and Harrison lab. Finally I would like to

thank my family and my wonderful wife, Orsida Luzati-Kola, for the love and support.

Page 4: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

iv  

Table of Contents

Acknowledgments..........................................................................................................................iii

Table of contents.............................................................................................................................iv

Lists of tables................................................................................................................................viii

List of figures................................................................................................................................viii

List of Abbreviations...................................................................................................................ix-x

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

1.1 Transient Receptor Potential Channels.....................................................................1 - 5

1.2 TRPM subfamily......................................................................................................6 - 7

1.2.1 TRPM1...............................................................................................................8

1.2.2 TRPM2.........................................................................................................8 - 9

1.2.3 TRPM3.......................................................................................................9 - 10

1.2.4 TRPM4/TRPM5.........................................................................................10-11

1.2.5 TRPM6.............................................................................................................11

1.2.6 TRPM8.............................................................................................................12

1.3 Transient Receptor Melastatin 7 (TRPM7)...........................................................12- 13

1.3.1 TRPM7 structure......................................................................................13 - 15

1.3.2 TRPM7 in divalent cation homeostasis....................................................17 - 19

1.3.3 TRPM7 in ischemic stroke.......................................................................19 - 21

1.3.4 TRPM7 and cytoskeleton.........................................................................22 - 23

1.4 Cofilin – the actin depolymerizing factor.............................................................25 - 26

Page 5: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

v  

1.4.1 Cofilin structure and expression...............................................................26 - 27

1.4.2 Cofilin and the regulation of actin dynamics............................................28 - 30

1.4.3 Regulation of cofilin activity....................................................................30 - 31

1.4.4 Cofilin in neurodegeneration....................................................................31 - 33

1.5 Rho associated kinase (ROCK)............................................................................34 - 35

1.5.1 ROCK protein signalling .........................................................................35 - 36

1.5.2 ROCK protein function............................................................................36 - 37

1.6 P-21 activated kinase (PAK)........................................................................................37

1.6.1 Structure and expression of P-21 activated kinases (PAKs)....................37 - 38

1.6.2 Regulation of PAK activity......................................................................38 - 39

1.6.3 PAK protein function...............................................................................39 – 40

1.7 Slingshot phosphatase (SSH).......................................................................................41

1.7.1 Slingshot phosphatase structure...............................................................41 – 42

1.7.2 Regulation of SSH1 activity.....................................................................42 - 43

1.7.3 SSH1 function...........................................................................................43 - 44

Chapter 2 – Objectives and Hypothesis..................................................................................45 - 46

Chapter 3 – Materials and Methods...............................................................................................47

3.1 Antibodies....................................................................................................................47

3.2 Rat Primary Neuronal Culture..............................................................................48 - 49

3.3 In vitro Stroke Model –Oxygen Glucose Deprivation (OGD).............................49 - 50

3.4 Protein extraction.........................................................................................................50

3.5 DC-Lowry Protein Assay.....................................................................................50 - 51

Page 6: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

vi  

3.6 SDS- Polyacrylamide Gel Electrophoresis (SDS-PAGE)...........................................51

3.7 Western Blotting...................................................................................................51 - 52

3.8 Immunofluorescent Analysis and Imaging...........................................................52 - 53

3.9 Cell Death, Viability and Apoptosis Assay..........................................................53 - 54

3.10 Statistical Analysis.....................................................................................................54

Chapter 4 – Results........................................................................................................................55

4.1 Neuronal survival following OGD: cytotoxicity and viability.............................55 - 58

4.2 Necrosis is the dominant form of cell death following OGD exposure while

apoptosis is more prevalent following NMDAR and TRPM7 inhibition...................64 - 65

4.3 The effects of oxygen glucose deprivation on ROCK2 activity..................................67

4.4 Oxygen glucose deprivation attenuates P-21 activated kinase 3 activity.............69 - 70

4.5 Modulation of SSH1 activity by TRPM7 following oxygen glucose

deprivation.........................................................................................................................73

4.6 Immunofluorescent analysis of intracellular protein localization........................76 - 77

Chapter 5 – Discussion...........................................................................................................81 - 82

5.1 Anti-excitotoxic treatment protects neuronal cultures against 2 hours of

oxygen glucose deprivation......................................................................................82 - 84

5.2 Different cell death mechanisms appear to dominate during OGD versus

NMDAR and TRPM7 inhibition..............................................................................84 - 86

5.3 Investigating the effects of ROCK signalling in TRPM7 mediated neuronal cell

death.........................................................................................................................86 - 88

5.4 Ischemic depression of PAK3 activity...............................................................89 - 90

Page 7: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

vii  

5.5 A role for SSH1 downstream of TRPM7 mediated activation in oxygen

glucose deprivation..................................................................................................90 - 92

5.6 Model of TRPM7 mediated neuronal cell death................................................93 - 94

5.7 Future studies of TRPM7 mediated cell death...................................................95 - 97

Chapter 6 – Summary............................................................................................................98 - 99

Reference list .....................................................................................................................100 - 120

Page 8: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

viii  

List of Tables

Table 1. TRP subfamilies...........................................................................................................5 - 6

Table 2. Proposed regulators and substrates of the mammalian TRPM7 channel..................24- 25

Table 3.Antibodies ........................................................................................................................47

List of Figures

Figure 1. TRPM7 Channel Structure.............................................................................................16

Figure 2. Cytotoxicity following oxygen glucose deprivation...............................................58 - 61

Figure 3.Confirmation of MCN rescue of neuronal death using Hoechst and propidium iodide staining..........................................................................................................................61- 63

Figure 4. Apoptosis is not the predominant cell death mechanism observed in neurons following OGD exposure.......................................................................................................65 - 66

Figure 5. OGD, MCN and Gd3+ do not affect phosphorylation of ROCK within the activation loop........................................................................................................................68 - 69

Figure 6. OGD attenuates PAK3 phosphorylation ................................................................71 - 72

Figure 7. TRPM7 modulates SSH1 activity............................................................................74- 75

Figure 8. LIMK1 and P-PAK3 co-immunostaining in cortical neurons................................77 - 78

Figure 9. LIMK1 and P-SSH1 co-immunostaining in cortical neurons..................................79- 80

Figure 10. New proposed model of TRPM7 mediated cofilin regulation during OGD................94

Page 9: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

ix  

List of Abbreviations

ABP – Actin binding protein

AD – Alzheimer’s disease

ADF – Actin depolymerizing factor

AET – Anti-excitotoxic therapy

AID – Auto inhibitory domain

AIP1 – Actin interacting protein 1

BDNF – Brain derived neurotrophic factor

CAP – Cyclase associated protein

CIN – Chronophin phosphatase

DMPK – Myotonic dystrophy protein kinase

eEF2 – Eukaryotic elongation factor 2

eEF2-K - Eukaryotic elongation factor 2 kinase

LIMK – Lin-11, Isl 1, Mec-3 kinase

MHR - TRPM homology region

NLS – Nuclear localization sequence

NUDT9H – TRPM2 nudix hydroxylase domain

PAK – P21 activated kinase

PBD – P21 binding domain

PLC – Phospholipase C

ROCK – Rho coiled-coil kinase or Rho associated kinase

TRP – Transient Receptor Potential

TRPA – Transient Receptor Potential Ankyrin

TRPC – Transient Receptor Potential Canonical

TRPM – Transient Receptor Potential Melastatin

Page 10: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

x  

TRPML – Transient Receptor Potential Mucolipin

TRPN – Transient Receptor Potential Nitric Oxide Mechanopotential

TRPP – Transient Receptor Potential Polycystin

TRPV – Transient Receptor Potential Vanilloid

OGD – Oxygen Glucose Deprivation

PH – Pleckstrin homology

PIP2 – Phosphatidylinositol 4,5 bisphosphate

PDGF – Platelet derived growth factor

RBD – Rho binding domain

ROS – Reactive Oxygen Species

SSH – Slingshot family phosphatase

Page 11: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

1  

Chapter 1

Introduction

Transient receptor potential melastatin 7 (TRPM7), a divalent cation permeable ion

channel, has been implicated as a key regulator of neuronal cell death in stroke (Aarts, 2003;

Aarts 2005). TRPM7 is unique in that it contains both a channel pore domain and a functional

enzyme (alpha-kinase), though the contribution of each domain to TRPM7 signalling is unclear.

According to recent studies TRPM7 is thought to play a role in regulating Mg2+ homeostasis, cell

growth, proliferation, death and cytoskeletal regulation (Ryazanova et al 2010; Aarts 2003;

Zhang et al 2010). Our most recent data shows that TRPM7 dependent cytoskeletal regulation,

particularly via the actin binding protein cofilin, mediates neuronal death in oxygen glucose

deprivation (a model of stroke) (Bent 2011). Cofilin is known to be regulated by LIM kinase

(Amano et al. 2002) and Slingshot family phosphatase 1 (Kurita et al. 2008). Recent findings in

our lab have show that LIMK phosphorylation is also regulated downstream of TRPM7

activation in anoxia. The goal of this thesis project is to extend our current findings by

elucidating potential LIMK and cofilin regulators that operate downstream of TRPM7 activation

during oxygen glucose deprivation. In this thesis I will describe Slingshot phosphatase 1 as a

new potential player that could contributes to TRPM7 mediated ischemic pathology via

regulation of LIMK and cofilin activity.

1.1 Transient Receptor Potential Channels

Page 12: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

2  

TRP (Transient Receptor Potential) channels are a distinct superfamily of cation selective

ion channels. TRP channels were first identified in Drosophila, where photoreceptors carrying

trp gene mutations exhibit a transient voltage response to continuous light stimulation. There are

now 28 identified TRP genes, some of which are expressed in a diverse range of organisms from

yeast, to worm, rodents and humans (Venkatachalam and Montell 2007). In addition to their

widespread prevalence in multicellular organisms, TRP channels display enormous diversity in

ion selectivity and regulatory mechanisms. Unlike most ion channels, which are classified

according to ion selectivity or function, TRP channels are classified based sequence homology

and structural characteristics into Group I and Group II TRPs. (Clapham et al. 2005;

Venkatachalam and Montell, 2007). Group I TRPs consist of 5 subfamilies that bear the greatest

sequence homology to the founding member of the superfamily, the Drosophila trp, while Group

II TRPs consist of two subfamilies that are distantly related to group I (Venkatachalam and

Montell 2007). Group I channels include TRPA(ankyrin), TRPC (canonical/ classical), TRPM

(melastatin), TRPN (Nitric oxide mechanopotential) and TRPV (vanilloid), while Group II

consists of TRPP (polycystin) and TRPML (mucolipin) ( refer to Table1) (Clapham et al.

2005). Of the 7 subfamilies, the classical TRPs (TRPCs) share the greatest sequence homology

with the Drosophila trp, while the polycystin TRPs appear to be the most evolutionarily ancient

as they are expressed in the widest range of organisms from yeast to humans (Vankatachalam

and Montell, 2007).

Despite the large amino acid sequence variation among TRP members, TRP channel

subunits share several structural features. Each TRP channel subunit consists of six

transmembrane segments with a pore forming loop (P-loop) between the fifth and sixth domains,

and an intracellular N- and C-termini (Clapham et al. 2005). TRP subunits assemble into

Page 13: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

3  

tetrameric complexes forming the functional TRP channels (Clapham et al. 2005). The N- and C-

terminal ends vary considerably amongst TRP members both in length and amino acid sequence.

The N- and C- termini contain domains, motifs, and enzymatic regions; these domains are in turn

involved in channel assembly, activity, and downstream signalling. Some of the N- and C-

terminal domains and motifs are broadly expressed amongst the different TRP subfamilies while

others are restricted to a particular few (see Table 1). For instance, TRPA, TRPC, and TRPV

contain N-terminal ankyrin repeats which are involved in a variety of functions from activating

ligands to gating by voltage and temperature (Venkatachalam and Montell, 2007). In contrast,

TRPM2, TRPM6 and TRPM7 are the only TRP channels to contain a C-terminal enzymatic

domain, thereby allowing them to respond to stimuli in a bifunctional manner. To add to the

complexity, closely related TRP channel proteins, such as TRP1 and TRP3, form

heteromultimeric complexes with regulatory and biophysical properties different from their

respective monomers (Lintschinger et al. 2000)

TRP channel activity is modulated by a variety of stimuli such as temperature, pH,

osmolarity, mechanical stretch, ion concentration (e.g. Ca2+ and Mg2+), PIP2, sphingolipids, and

plant compounds (Clapham et al. 2005; Venkatachalam and Montell, 2007). Given that TRP

channel proteins are classified according to sequence similarity, it is difficult to predict the

regulatory mechanisms of a particular TRP channel based on subfamily classification. Indeed

variations in channel regulation are observed within members of the same subfamily. For

instance, within the TRPM subfamily TRPM4 and TRPM5 are activated by intracellular Ca2+

depletion; TRPM2 is activated by reactive oxygen and nitrogen species; whereas, TRPM8 is

activated by cool temperatures (8-28°C) (Venkatachalam and Montell, 2007). However, some

similarities in channel regulation do exist within and outside of particular TRP subfamilies as is

Page 14: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

4  

the case with the phospholipase C (PLC) signalling pathway. All members of the TRPC

subfamily are regulated by pathways coupled to PLC stimulation; furthermore, TRP proteins

from other subfamilies such as TRPM and TRPP are also regulated by PLC pathway signalling

(Venkatachalam and Montell, 2007). TRP channels also exhibit enormous diversity in cation

selectivity with the majority of TRP channels being permeable to divalent cations including Ca2+

and Mg2+ (Clapham et al. 2005; Venkatachalam and Montell. 2007). However, exceptions do

exist as is the case with TRPM4 and TRPM5, the only TRP channels that are permeable

exclusively to monovalent cations (Venkatachalam and Montell. 2007).

Physiologically, TRP channels are involved in sensory reception and transduction of

environmental stimuli including light, touch, temperature, taste, olfaction and pain. TRPV1 and

TRPV2 contribute to warm and hot sensation respectively, while TRPM8 and TRPA1 contribute

to cool and cold sensation respectively (Venkatachalam and Montell. 2007, McKemy et al.

2002). TRP channels also function as cellular sensors of their local environment. TRPC3, for

example, regulates axonal guidance by brain-derived neurotrophic factor (Li et al. 1999). Some

of the physiological roles of TRP proteins have been discerned through genetic mutants and

knockout models. For instance, TRPP channels are important for kidney function as autosomal

dominant polycystic kidney disease is caused by TRPP1 mutations (Wu et al. 1998). Mutations

in the TRPML1 gene are responsible for mucolipidosis, a lysosomal storage disorder

characterized by severe neurodegeneration, mental retardation, achlorhydria and retinal

degeneration (Bargal et al. 2000). Additional pathological roles of TRP channels are being

uncovered and they range from hypomagnesemia and hympocalcemia, to progressive familial

heart block, amyotrophic lateral sclerosis, Parkinsonism, Alzheimer’s, and spinal muscular

atrophy (Wu et al. 2010). However, there is still a lot of work to be done, it is unclear how many

Page 15: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

5  

TRP channels are activated in vivo, as very few known endogenous TRP stimulators and

inhibitors have been discovered, thereby making their study even more challenging.

Table 1. TRP subfamilies

There are six TRP subfamilies organized according to sequence homology. Humans do not

express TRPN; all other subfamilies express at least one member. Below are listed some of the

common physiological roles of the TRP subfamily members (data from Clapham et al. 2005;

Venkatachalam and Montell, 2007; Wu et al. 2010)

Subfamily Members found in Humans

Physical Characteristics

Physiological Roles

TRPA (Ankyrin) 1 N-terminal ankyrin repeats

-Sensitive to plant derived irritants (mustard oils) and possibly to mechanical stretch

TRPC (Canonical) 6 N-terminal ankyrin repeats C-terminal TRP domain C-terminal calmodulin/IP3R binding (CRIB) domain N-terminal and C-terminal coiled coil domain

-Receptor mediated Ca2+-dependent secretion and contraction (TRPC1) - Resistance artery myogenic tone, airway regulation (TRPC3) -Agonist dependent vasorelaxation in vascular-endothelium (TRPC4) - Cation influx in response to PLC activation (TRPC6) and Gq activation (TRPC7)

TRPM (Melastatin)

8 N-terminal TRPM homology region C-terminal TRP domain C-terminal coiled coil domain C-terminal alpha kinase (TRPM6 & TRPM7) C-terminal ADP-ribose hydroxylase domain

-Light sensation ? (TRPM1) -Cellular redox sensor (TRPM2, TRPM7) -Ca2+ absorption in renal collecting tubule (TRPM3) -Renal and gastrointestinal Mg2+ uptake (TRPM6) -Myogenic cerebral artery constriction (TRPM4)

Page 16: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

6  

(TRPM2) -Sweet, bitter, umami taste transduction (TRPM5) - Cellular Mg2+ homeostasis; OGD induced Ca2+ influx and neuronal death (TRPM7) -Cold sensation (TRPM8)

TRPN (Nitric Oxide Mechanopotential)

0 N-terminal ankyrin repeats C-terminal TRP domain

TRPV (Vanilloid) 6 N-term ankyrin repeats C-terminal coiled coil domain C-term TRP domain C-terminal Calmodulin/IP3R binding domain

-Ca2+ entry in response to heat, capsaicin; pain sensation (TRPV1) -Ca2+ entry in thermal pain sensation (TRPV2) -warm sensation (TRPV3) -osmolarity sensing (TRV4)

TRPML (Mucolipin)

3 Long extracellular loop between S1 and S2 Short N- and C-terminus C-terminal EF hand

-Ca2+ dependent lysosomal exocytosis (?) (TRPML1)

TRPP (Polycystin) 3 Long extracellular loop between S1 and S2 C-terminal EF hand C-terminal coiled coil domain

-Kidney and liver cyst formation; mechanosensation in kidney cilia (TRPP1) - kidney and retinal development (TRPP2) - Involved in sorting or transport in late endocytic pathway (TRPP3)

1.2 TRPM subfamily

The TRPM subfamily is named after the TRPM1 protein also known as ‘melastatin’

whose expression has been found to correlate inversely with melanoma tumor progression

(Duncan et al. 1998). TRPM channel proteins, like other TRP proteins, contain 6 transmembrane

segments with a pore forming loop between S5-S6, and intracellular N-terminal and C-terminal

ends (Clapham et al. 2005; Venkatachalam and Montell, 2007). Similar to TRPC, TRPV and

Page 17: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

7  

TRPP channels, TRPM proteins contain a C-terminal coiled-coil domain, which is important for

channel assembly, multimerization, and function (Latorre et al. 2009). The presence of an N-

terminal TRPM homology region (MHR) is unique to the TRPM channel subfamily; however, its

functional role is largely unknown (Wu et al, 2010). Another distinct feature of TRPM family

members is the presence of a C-terminal enzymatic domain in TRPM2, TRPM6 and TRPM7

(Clapham et al. 2005; Venkatachalam and Montell, 2007). Tissue distribution varies among

TRPM subfamily member since some TRPMs, such as TRPM2, TRPM4 and TRPM7, exhibit

broad expression throughout multiple tissues whereas others, including TRPM1 and TRPM6 and

TRPM8, are restricted to a small subset of tissues (Fonfria et al. 2006).

TRPM subfamily members have the most divergent biophysical properties and regulation

compared to the other subfamilies such as TRPCs and TRPVs. For instance, TRPM2 channels

have a high permeability for Ca2+, analogous to other TRPs, whereas TRPM7 channels are more

permeable to trace metals and Mg2+ than Ca2+ (Clapham et al.2005; Monteilh-Zoller et al. 2002).

Furthermore, other TRPs, such as TRPM4 and TRPM5, are exclusively permeable to

monovalent cations (Guinamard et al. 2011). TRPM subfamily members also vary in the modes

of regulation, as some are modulated by reactive oxygen species, low pH, intracellular Mg2+ and

Ca2+ levels, and others are modulated by external stimuli such as light and temperature. These

differences in structural organization and regulation of channel activity, in conjunction with the

varying tissue distribution contribute to a diverse range of physiological functions including:

neural tube and embryonic development; maintenance of Ca2+ and Mg2+ homeostasis; sensation

of light, temperature, and taste (Venkatachalam and Montell, 2007).

Page 18: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

8  

1.2.1 TRPM1

TRPM1, the founding member of the TRPM subfamily, serves as a prognostic marker of

melanoma metastasis (Duncan et al. 1998). Despite its discovery15 years ago, very little progress

was made in identifying the molecular properties and physiological functions of TRPM1.

TRPM1 expression is observed in the brain, heart, melanocytes and retina with a predominant

localization in intracellular vesicles (Fonfria et al. 2006; Oancea et al. 2011). This intracellular

localization appears to be regulated by a direct interaction between TRPM1S, a short splice

variant, and TRPM1 full length protein, which inhibits TRPM1 translocation to the plasma

membrane (Xu et al. 2001). Physiologically, TRPM1 is involved in the light sensation; the

TRPM1 mediated cationic current is critical in mediating ON bipolar cell depolarization

following exposure to light (Oancea et al. 2011). In addition, TRPM1 may be involved in

melanin production since TRPM1 expression positively correlates with melanin content in

human melanocytes, while TRPM1 knockdown significantly reduced melanin content in these

cells (Oacea et al. 2009). Despite some recent progress much work needs to be done in

identifying the role TRPM1 plays in melanocytes and in melanoma tumor progression.

1.2.2 TRPM2

Unlike TRPM1, TRPM2 channels are ubiquitously expressed throughout many of the

body tissues with the exception of cartilage and bone (Fonfria et al. 2006). TRPM2 is a non-

selective cation channel that is permeable to both monovalent and divalent cations including

Ca2+, Mg2+ and Na+ (Wu et al. 2010). A unique feature of TRPM2 channels is the presence of a

C-terminal nudix hydroxylase domain (NUDT9-H), which is homologous to the NUDT9 of

Page 19: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

9  

ADP-ribose hydroxylase (Perraud et al. 2001). Cyclic ADP-ribose, arachidonic acid, and

intracellular Ca2+ positively modulate TRPM2 activity (Hara et al. 2002). This is particularly

important since TRPM2 functions as a cellular sensor of oxidative stress. Reactive oxygen and

reactive nitrogen species activate the TRPM2 channel through the generation of ADP-ribose

from mitochondria; the consequent Ca2+ influx ultimately leads to cell death (Perraud et al. 2005;

Kaneko et al. 2006). In addition, inhibition of the ROS mediated TRPM2 current is protective in

both heterologous expression systems and primary neurons (Perraud et al, 2005; Kaneko et al.

2006). Besides acting as a cellular redox sensor, TRPM2 is involved in the regulation of insulin

secretion by pancreatic β-cells as well as in ROS induced chemokine production by immune cells

(Togashi et al. 2006; Yamamoto et al. 2008). Thus, the development of TRPM2 specific

inhibitors could help alleviate ROS mediated pathologies.

1.2.3 TRPM3

TRPM3 is one of the least characterized members of the TRPM subfamily. In humans

TRPM3 is primarily expressed in the kidney, with lower expression also found in brain, testis

and spinal cord (Lee et al. 2003). Heterologously expressed TRPM3 channels mediate a

constitutive Ca2+ influx, which is enhanced by a hypotonic environment and blocked by Gd3+

(100µM) treatment (Grimm et al. 2003; Lee et al. 2003). Treatment with D-erythro-sphingosine

selectively enhances the TRPM3 mediated Ca2+ current; however, whether this interaction is

physiologically relevant remains unknown (Grimm et al. 2005). A characteristic feature of

TRPM3 channels is the presence of multiple splice variants, some of which have been shown to

exhibit clearly distinct biophysical properties. Using two splice variants that only differed at a

Page 20: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

10  

splice site forming the pore region of the TRPM3 channel, Oberwinkler and colleagues (2005)

revealed that one isoform was exclusively permeable to monovalent cations while the other was

highly permeable to divalent cations. This finding further complicates the interpretation of

TRPM3 biophysical data as investigators have to take into consideration the splice variants

expressed in their experimental paradigm.

1.2.4 TRPM4/TRPM5

TRPM4 and TRPM5 share many structural and functional features, and thus will be

described together. In humans TRPM4 is ubiquitously expressed throughout all tissues

examined, with highest expression found in the intestine and prostate (Fonfria et al. 2006).

TRPM5 however, is predominately found in taste cells, with some expression in the pituitary,

kidney, intestine, pancreas, and prostate (Perez et al. 2002; Fonfria et al. 2006). TRPM4 and

TRPM5 channels are permeable to monovalent cations, a feature unique to these two among the

entire TRP superfamily (Guinamard et al. 2011). The presence of specific amino acid residues

within the channel loop region, located between transmembrane segment 5 and 6, are deemed

responsible for the monovalent cation selectivity (Nilius et al. 2005). TRPM4 and TRPM5 are

activated by a rise in intracellular Ca2+; their activity is also enhanced by membrane

depolarization (voltage sensitive), and PIP2treatment (Nilius et al. 2005; Guinamard et al. 2011).

Interestingly, intracellular ATP inhibits TRPM4 but has no effect on TRPM5 activity (Nilius et

al. 2005). The presence of an ATP binding domain on TRPM4 appears to be critical for TRPM4

inhibition, as mutations in the ATP binding domain abolish this effect (Nilius et al. 2005).

Another compound that takes advantage of the ATP binding site is decavanadate, which has been

Page 21: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

11  

shown to enhance TRPM4 activity without affecting TRPM5 (Nilius et al. 2004). In this

situation decavanadate is likely competing with ATP for the ATP binding site (Nilius et al.

2004). These structural and functional differences between TRPM4 and TRPM5 channels could

prove useful in isolating the currents of each channel. Physiologically TRPM5 is involved in

taste transduction whereas TRPM4, given its widespread tissue distribution, has been implicated

in numerous processes including: modulation of insulin secretion, immune response, constriction

of cerebral arteries, inspiratory neuron firing rate, and cardiac activity (Guinamard et al. 2011).

1.2.5 TRPM6

TRPM6 is a non-selective divalent cation permeable channel that is expressed in the

brain, kidney, and intestine; with the latter two showing the greatest expression (Fonfria et al.

2006). TRPM6 and its closest homologue TRPM7 have similar protein structure and biophysical

properties. Both proteins have a C-terminal alpha kinase and a channel pore permeable to

divalent cations (Wu et al. 2010). TRPM6 mediates a small Ca2+ and Mg2+ inward current under

physiological conditions (Wu et al. 2010). This current in inhibited by intracellular Mg2+ and

potentiated by acidic extracellular pH (Voets et al. 2004; Jiang et al. 2005). TRPM6 mutants

have been linked to hypomagnesaemia and secondary hypocalcaemia, suggestive of a role for

TRPM6 in Mg2+ and Ca2+ uptake in the intestine and kidney (Schlingmann et al. 2002; Voets et

al. 2004). Furthermore, TRPM6 may play are role in neural tube closure in development since

TRPM6 knockout animals have neural tube defects and die by embryonic day 12.5 (Walder et al.

2009).

Page 22: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

12  

1.2.6 TRPM8

The TRPM8 channel is best characterized for its involvement in cold sensation. Initially

identified in the prostate, TRPM8 expression has been observed in many other tissues including

liver, dorsal root ganglia, trigeminal ganglia, bladder, male genital tract, tongue, gastric fundus,

vascular smooth muscle, lung, spinal cord and brain (Fonfria et al. 2006; Liu and Qin, 2011).

TRPM8 can be activated by multiple types of stimuli from innocuous cool to noxious cold

temperatures (McKemy et al. 2002). In addition, cooling agents such as menthol and icilin

induce a response similar to thermal cold by activating TRPM8 (McKemy et al. 2002). TRPM8

activity is modulated by a variety of factors including PIP2, α2- adrenoreceptor signalling,

polyphosphates and lipid rafts (Liu and Qin, 2011). Recently TRPM8 has been suggested as a

potential therapeutic target for chronic pain relief and prostate cancer (Liu and Qin, 2011).

1.3 Transient Receptor Melastatin 7 (TRPM7)

Transient Receptor Melastatin 7 (TRPM7) is a ubiquitously expressed non-selective

divalent cation channel with murine expression greatest in the heart, kidney and cerebrum (Keil

et al. 2006). In humans the greatest expression is seen in the heart, adipose tissue and bone

(Fonfria et al. 2006). TRPM7 channels are permeable to divalent cations such as Ca2+ and Mg2+

as well as trace metals (Monteilh-Zoller et al. 2002). Normally, under physiological conditions

the TRPM7 currents are inhibited, that is they exhibit a very small inward current. However,

TRPM7 channels have the potential to drive a large inward Ca2+ current during conditions of

anoxia (OGD), oxidative stress, and low extracellular Ca2+ (Aarts et al. 2003; Xiong et al. 2002).

TRPM7 channel activity can be blocked through a variety of non-specific inhibitors including:

Page 23: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

13  

Gd3+ (Aarts et al. 2003); 2-aminoethoxydiphenyl borate (2-APB) (Peppiatt et al. 2003); 5-

lipoxygenase inhibitors namely, NDGA, MK886 and AA861 (Chen et al. 2010); as well as

intracellular Mg2+ ([Mg2+]i) and Mg2+-ATP ([Mg2+-ATP]i) (Nadler et al. 2001). Investigating the

function and regulation TRPM7 channels, in vivo, has been difficult particularly due to the

embryonic lethality of TRPM7 mutants and the absence of specific TRPM7 activators and

inhibitors. However, some advancement has been made through targeted knockdown using viral

vectors.

1.3.1 TRPM7 structure

The structure of TRPM7 is similar to other TRPM subfamily members but it does contain

some distinct features (see Figure 1). Similar to TRPM members, TRPM7 contains 6

transmembrane domains that are flanked by an intracellular N-terminal and C-terminal end

(Clapham et al. 2005; Wu et al. 2010). A loop region between segment 5 and segment 6 makes

up the channel pore of TRPM7 (Clapham et al. 2005; Wu et al. 2010). The N-terminus contains

a large TRPM homology region (MHR) that is conserved amongst TRPM subfamily members. A

study by Phelps and Gaudet (2007) shows that MHR is essential for TRPM8 channel localization

since deletions within the MHR region impair TRPM8 localization to the plasma membrane.

Whether, the MHR region plays a similar role for TRPM7 and other TRPM family members

remains to be determined. TRPM7 also contains an N-terminal snapin binding domain important

for vesicle fusion and neurotransmitter release (Krapivinsky et al. 2006; Brauchi et al. 2007).

The proximal C-terminal end of TRPM7 contains a TRP domain, which interacts with

phosphatidylinositol 4,5 bisphosphate (PIP2) a positive regulator of some TRP channels (Rohacs

Page 24: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

14  

et al. 2005; Bae et al. 2011). The TRP domain is followed by a coiled-coil domain, which is

essential for TRP channel protein assembly and tetramerization (Tsuruda et al. 2006).

What sets TRPM7 apart from most TRP channels is the presence of an α-kinase in the

distal end of the C-terminus, a feature that it only shares with TRPM6 (Clapham et al. 2005; Wu

et al. 2010). Alpha kinases are relatively rare enzymes that share no sequence homology with

conventional eukaryotic Ser/Thr kinases (Ryazanov et al. 2002). Unlike conventional kinases

which phosphorylate their substrates within loops, turns or regions with irregular structure; alpha

kinases phosphorylate their substrates within alpha-helices, hence their name (Ryazanov et al.

1999). Interestingly, the structure of the TRPM7 catalytic domain is similar to that of

conventional protein kinases despite the amino acid sequence differences (Ryazanov et al. 2002),

however; different sets of residues are engaged in contacts with the base and sugar moieties of

ATP making the TRPM7 alpha-kinase an ideal target for drug development (Li et al. 2011).

Since the discovery of the TRPM7 alpha-kinase domain, there has been much interest and

controversy in identifying the regulatory mechanisms and binding partners of this channel with

kinase activity (chanzyme). Runnels and colleagues (2002) were among the first to report a

model of TRPM7 regulation via the PLC signalling pathway. The authors suggested that PIP2 is a

key modulator of TRPM7 function, as receptor mediated activation of PLC and the consequent

hydrolysis of PIP2 inhibited TRPM7 currents (Runnels et al. 2002). In contrast, a study by

Takezawa et al (2004) showed that receptor mediated activation of PLC had no effect on

TRPM7 current, thereby suggesting that PIP2 depletion is not the mechanism contributing to

TRPM7 inhibition. Rather, they argue that receptor mediated regulation of TRPM7 activity

occurs at the level of adenylyl cyclase, as enhanced cyclic AMP levels enhance TRPM7 channel

activity (Takezawa et al. 2004). Another study by Langeslag et al (2007) was in complete

Page 25: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

15  

disagreement with Runnels and colleagues (2002) showing that receptor mediated activation of

PLC and the consequent PIP2 depletion led to enhanced TRPM7 channel activity(Langeslag et

al. 2007). Furthermore, the authors showed that cAMP levels did not affect the TRPM7 currents

(Langeslag et al. 2007). Some of these differences could be accounted for by variations in the

expression models used, the PLC receptor agonists employed, as well the electrophysiological

recording methods. However, the findings of Langeslag et al (2007) appear to be consistent with

a physiological regulation of endogenously expressed TRPM7.

Controversy also extends to investigations into the role of the TRPM7 alpha kinase

domain. It was originally suggested that the TRPM7 alpha kinase was required for channel

activity (Runnels et al. 2001). However, it was later shown that kinase dead mutants still form

functional channels and that the kinase domain is not essential for channel activation (Schmitz et

al. 2003). Instead, the alpha-kinase appears to play modulatory role by reducing the sensitivity of

TRPM7 channel to [Mg2+]i mediated suppression, which in turn will feedback to modulate kinase

activity via Mg2+ influx (Schmitz et al. 2003). The TRPM7 alpha-kinase phosphorylates a

number of proteins including Annexin I (Dorovkov and Ryazanov 2004), Myosin IIA (Clark et

al. 2006) and eEF2-K (Perraud et al. 2011) implicating TRPM7 in Ca2+ signal transduction,

cytoskeletal organization and regulation of translation respectively. The significance of the above

mentioned interactions and the various regulatory roles of TRPM7 will be discussed in the

sections below.

Page 26: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

16  

Figure 1. TRPM7 Channel Structure.

The TRPM7 channel is unique in that it contains both a channel domain and an alpha-kinase

domain hence it’s often called a ‘chanzyme’. TRPM7 channels consist of six transmembrane

subunits flanked by an intracellular N-terminus and C-terminus. The channel pore resides

between S5-S6, and is permeable to various divalent cations including Ca2+ and Mg2+, whereas

the kinase domain resides in the C-terminal end. Both the channel domain and the alpha-kinase

domain have been implicated in numerous signalling pathways, and have shown to influence

each other’s activity.

Snapin = snapin binding protein; TRP = conserved TRP box domain; CC = coiled coil domain;

Ser/Thr kinase = apha-kinase domain

Page 27: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

17  

1.3.2 TRPM7 in divalent cation homeostasis

TRPM7 channels are inherently permeable to a wide range of divalent cations with the

largest permeability for Zn2+ and Ni2+ followed in order by Ba2+, Co2+, Mg2+, Mn2+, Sr2+, Cd2+

and Ca2+ (Monteilh-Zoller et al. 2002). The above observation also points out that, in contrast to

other TRPs, TRPM7 is more permeable for Mg2+ than Ca2+, consistent with its involvement in

Mg2+ homeostasis (Monteilh-Zoller et al. 2002). In divalent cation free extracellular solution

large K+ and Na+ currents develop (Runnels et al. 2001; Schmitz et al. 2003), whereas in the

presence of trivalent cations, such as La3+ and Gd3+, TRPM7 currents are blocked (Runnels et al.

2001; Monteilh-Zoller et al. 2002; Aarts et al. 2003). Heterologous expression systems have

shown that TRPM7 channels are constitutively active and that their activity is regulated by

intracellular Mg2+ and Mg2+-ATP. Patch clamp experiments reveal a rapid induction of TRPM7

currents within minutes following depletion of intracellular Mg2+; addition of intracellular Mg2+

or Mg2+-ATP ( ≥1mM) strongly inhibited the TRPM7 channel currents (Nadler et al. 2001;

Schmitz et al. 2003). Initially it was believed that Mg2+-ATP directly contributed to inhibition of

TRPM7 channel activity through an ATP-mediated mechanism (Nadler et al. 2001); however,

follow up studies have showed that Mg2+-ATP inhibits TRPM7 currents by functioning as a

source of Mg2+ (Kozak and Cahalan 2003). Given that under physiological conditions TRPM7

channels exhibit only 10% of their maximal activity (Nadler et al. 2001), the above findings are

indicative of a feedback mechanism involved in maintaining cellular Mg2+ homeostasis.

The role of TRPM7 in ion homeostasis is ultimately linked with cell death since deletion

of TRPM7 from B-lymphocytes leads to proliferative arrest, cell senescence and eventually cell

death (Schmitz et al. 2003). Supplementation with excess Mg2+ or expression of a TRPM7

mutant lacking kinase activity corrected the Mg2+ deficiency and led to functional recovery of

Page 28: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

18  

the TRPM7 knockout cells suggesting that TRPM7 channel function alone is sufficient for Mg2+

homeostasis (Schmitz et al. 2003). This effect was specific to Mg2+, since only Mg2+ containing

salts complemented the TRPM7 deficiency (Schmitz et al. 2003). In addition, TRPM7 function

is essential in development as TRPM7 knockout mice exhibit embryonic lethality by gestation

day 7.5 (Jin et al. 2008). Surprisingly, the alpha-kinase domain of TRPM7 appears to play an

essential role since deletion of the TRPM7 kinase domain was sufficient to generate early

embryonic lethality in mice (Ryazanova et al. 2010). Furthermore, mice heterozygous for the

kinase deletion were viable but had defects in intestinal Mg2+ absorption; leading the authors to

suggest that TRPM7 is essential for both cellular and whole body Mg2+ homeostasis (Ryazanova

et al. 2010).

Calcium is a ubiquitous signalling molecule involved in numerous cellular processes

including growth, differentiation, migration, synaptic activity, and cell death. Given its

contribution to numerous signaling pathways intracellular Ca2+ levels are tightly regulated

through a combination of controlling Ca2+ influx, efflux and storage. Dysregulation of Ca2+

homeostasis results in the activation of signalling cascades that are otherwise silent or operating

at a low level ultimately leading to a pathological state (Arundine and Tymianski, 2003). In

addition to its role in Mg2+ homeostasis, TRPM7 has been implicated in a number of Ca2+

mediated physiological processes such as cytoskeletal regulation, cell adhesion and cell

migration (Su et al. 2006; Wei et al. 2009). Furthermore, TRPM7 Ca2+ currents have been shown

to have the most profound effects in various pathologies including ischemia and cancer (Aarts et

al. 2003; Chen et al. 2010; Middelbeek et al. 2012). The TRPM7 channel has been recently

identified as a mechanical sensor, conducting localized Ca2+ currents in the leading edge of

migrating cells (Wei et al. 2009). This localized Ca2+ entry from TRPM7 is amplified by Ca2+

Page 29: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

19  

release from the ER through IP3R thereby generating the calcium microdomains that are required

for guidance and directional migration (Wei et al. 2009). TRPM7 Ca2+ currents have also shown

to regulate cell adhesion by controlling the activity of m-calpain, a Ca2+ dependent protease (Su

et al. 2006). A pro-migratory role for TRPM7 has been identified in human nasopharyngeal

carcinoma whereby inhibition of the TRPM7 Ca2+ current, or TRPM7 knockdown, impaired

migratory potential (Chen et al. 2010). On the other hand, enhancement of TRPM7 current, or

TRPM7 overexpression, increased the migratory potential of human nasopharyngeal carcinoma

cells (Chen et al. 2010). In addition to its role in nasopharyngeal carcinoma, TRPM7 channel

function has been shown to be essential in breast tumor cell metastasis, and can be used as a

prognostic marker of poor patient outcome (Middelbeek et al. 2012). TRPM7 Ca2+ currents also

contribute to anoxic neuronal cell death, the specifics of which will be discussed in the following

section.

1.3.3 TRPM7 in ischemic stroke

Ischemic stroke is a consequence of transient or permanent reduction in cerebral blood

flow in a restricted vascular region due to vessel occlusion or hemorrhage. Neuronal damage

results from a combination of excitotoxicity, calcium overload, and inflammation that ultimately

results in cell death (Aarts and Tymianski 2005). Generally, focal impairment prevents the

transport of oxygen, glucose and other substrates to the affected tissue. This state of oxygen and

glucose deprivation (OGD) interrupts oxidative phosphorylation by the mitochondria, which

reduces ATP production resulting in a rapid depletion of ATP levels (Katsura et al. 1994). The

profound loss of ion gradients and membrane depolarization that follow (Katsura et al. 1994)

Page 30: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

20  

cause an excess of excitatory amino acids (EAAs) to be released in extracellular space (Arundine

and Tymianski, 2003). These supraphysiological glutamate levels stimulate surrounding neurons

thereby initiating a positive feedback loop that results in even more glutamate release. The

prolonged exposure to EAA stimulation and the consequent dysregulation of Ca2+ homeostasis

can lead to the activation of signalling events that ultimately result in cell death, hence the term

excitotoxicity (Arundine and Tymianski, 2003).

It has been established that virtually all glutamate receptor subtypes contribute to

excitotoxicity through a Ca2+ mediated process including ionotropic N-methyl-D-aspartate

receptor(NMDAR), 2-amino-3-(3-hydroxy-5-methylisoxazol-4-yl) receptor(AMPAR), and

kainite receptors (Choi 1985,1987; Arundine and Tymianski, 2003). Treatment of primary

cortical neurons with glutamate receptor antagonists is neuroprotective for up to 1 hour of OGD

(Aarts et al. 2003). Blockage of NMDA subtype glutamate channels in animal models of stroke

has also shown to be neuroprotective and prevent excitotoxicity but only when administered

prior to or immediately following stroke onset(Aarts and Tymianski 2005). Despite the strong

role of for excitotoxicity in ischemic brain damage human clinical trials using anti-excitotoxic

therapies (AET) failed to show any significant neuroprotection, and were associated with many

adverse side effects (Cheng et al. 2004). In retrospect is not that surprising that global inhibition

of glutamate signalling would have profound physiological effects given glutamate’s

involvement in many cognitive functions including learning and memory.

The failure of AET indicated that although glutamate receptor signalling contributes to

neurotoxicity, other non-excitotoxic mechanisms are also involved. A study by Aarts and

colleagues (2003) identified a non-excitotoxic mechanism of neurotoxicity involving TRPM7

mediated cation currents. The authors used OGD or NaCN treatment as their in vitro model of

Page 31: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

21  

ischemia on primary cortical neurons. AET treatment was able to prevent the ensuing Ca2+

influx normally observed following OGD, for up to 1 hour; however, by 2 hours of OGD

intracellular Ca3+ levels were similar to the untreated OGD cells, suggesting that AET is not

sufficient to prevent Ca2+ dysregulation following prolonged ischemia (Aarts et al. 2003).

Furthermore, AET unmasked a TRPM7 mediated cation current with high permeability for Ca2+.

Heterologous TRPM7 expression enhanced the current whereas Gd3+ treatment or TRPM7

knockdown blocked it (Aarts et al. 2003). In addition, TRPM7 inhibition was neuroprotective for

up to 3 hours of OGD, even in the absence of AET, indicating thatTRPM7 activity contributes to

neuronal death independently of glutamate signalling (Aarts et al. 2003). Reactive oxygen

species (ROS) potentiated both the TRPM7 current and neuronal cell death, whereas antioxidant

treatment inhibited both the TRPM7 current and neuronal death (Aarts and Tymianski; 2003).

The above findings led the authors to propose a mechanism whereby ROS production during

ischemia activates TRPM7 channels leading to Ca2+ influx, which in turn generates more ROS

thereby starting a positive feedback loop that leads to Ca2+ overload and neuronal death (Aarts et

al, 2003). A follow up study by Sun and colleagues (2009), using an in vivo rat model of global

ischemia, revealed the involvement of TRPM7 channels in hippocampal neuronal injury.

Suppression of TRPM7 expression in CA1 neurons via intrahippocampal injection of viral

vectors containing shRNA specific to TRPM7 had no adverse effects on animal survival,

neuronal morphology, neuronal excitability or synaptic plasticity (Sun et al, 2009). However,

TRPM7 knockdown was neuroprotective against ischemia and preserved neuronal morphology

and function. Behavioural assays also revealed that TRPM7 suppression preserved hippocampal

dependent memory following ischemia (Sun et al. 2009).

Page 32: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

22  

1.3.4 TRPM7 and cytoskeleton

The cytoskeleton forms the structural framework of a cell and regulates many aspects of

cellular physiology including cell shape, migration, signalling, division, differentiation, and

death. TRPM7 is a bifunctional protein that influences the cytoskeleton through channel

conductance and/or the alpha-kinase domain. A study by Nadler and colleagues (2001) was the

first to provide evidence regarding the involvement of TRPM7 in cytoskeletal regulation. The

authors reported that overexpression of TRPM7 in HEK293 cells resulted in a loss of cell

adhesion, cell rounding, and ultimately cell death (Nadler et al. 2001). Su et al (2006) extended

these findings by showing that localized TRPM7 currents in peripheral adhesion complexes

contribute to loss of cell adhesion via activation of a Ca2+-dependent protease, m-calpain. It was

later discovered that TRPM7 channel activity causes nitrosative and oxidative stress, which

induces cell rounding through p38 MAPK/JNK dependent activation of m-calpain (Su et al.

2010). TRPM7 currents also contribute to directional cell migration through the generation of

high calcium microdomains called ‘Ca2+-flickers’ (Wei et al. 2009). Platelet derived growth

factor (PDGF) treatment increased the frequency of the Ca2+-flickers and induced directional cell

migration of fibroblasts; whereas suppression of TRPM7 expression decreased the frequency of

Ca2+-flickers and impaired the turning of fibroblasts in response to PDGF (Wei et al. 2009). The

above studies reveal a mechanism of actomyosin cytoskeleton regulation through a TRPM7

mediated Ca2+ current. However, it was recently shown that a TRPM7 mediated Mg2+ current

also regulates the actomyosin cytoskeleton in fibroblasts (Su et al. 2011). TRPM7 knockdown

disrupted the actomyosin cytoskeleton, focal adhesions, cell polarity and directional migration all

of which could be rescued with the expression of a kinase dead TRPM7 channel (Su et al 2011).

Page 33: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

23  

The TRPM7 alpha-kinase is also involved in cytoskeleton regulation. Annexin I is the

first identified substrate of the TRPM7 alpha kinase (see Table 2)(Dorovkov and Ryazanov

2004). Annexin 1 is a Ca2+ and phospholipid binding protein that can promote Ca2+ dependent

membrane fusion, as well as regulating cell growth, differentiation and apoptosis

(Monastyrskaya et al. 2009). TRPM7 phosphorylates annexin 1 at a conserved Ser5 residue in the

N-terminal region, in a Ca2+ dependent manner (Dorvkov and Ryazanov, 2004). Heterologous

TRPM7 expression in HEK-293 cells enhances annexin I phosphorylation. Given that the N-

terminal region of annexin proteins governs their interactions, Ser5 phosphorylation via TRPM7

may modulate annexin I function (Dorovkov and Ryazanov, 2004).Myosin IIA is another

substrate of the TRPM7 alpha-kinase (see Table 2). Clark et al (2006) have shown that the

TRPM7 alpha kinase directly interacts with myosin IIA heavy chain in a Ca2+ dependent manner.

Furthermore, this interaction results in myosin IIA heavy chain phosphorylation consistent with a

role for TRPM7 in regulating actomyosin contractility (Clark et al. 2006). TRPM7 can also bind

to actin; however this does not result in actin phosphorylation (Clark et al. 2006). The

bifunctional nature of TRPM7 allows it to regulate the actin cytoskeleton predominately through

divalent cation influx but also through the direct phosphorylation of cytoskeletal proteins. Given

that TRPM7 channel currents modulate kinase activity and the kinase domain in turn modulates

channel activity is indicative of an additional layer of regulation that could serve to ensure an

appropriate response by TRPM7.

Page 34: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

24  

Table 2: Proposed regulators and substrates of the mammalian TRPM7 channel

Region of TRPM7 interaction

Proposed interacting protein

Proposed function Notes Reference

C-terminal kinase domain

Phospholipase C (PLC) isoforms

Receptor mediated activation of PLC leads to inactivation of TRPM7 current

Inactivation appears to be due to a direct interaction with PIP2 and not its downstream signalling molecules

Runnels et al. 2002

TRP consensus domain

PLC, PIP2 Receptor mediated activation of PLC causes PIP2 breakdown leading to TRPM7 activation

TRPM7-dependent Ca2+ increase correlated with PLC activation. Similar results as above were observed when [Mg2+]i was below physiological levels (1-2mM)

Langeslag et al. 2007

C-terminal kinase domain

cAMP via PKA

TRPM7 activity is regulated through its endogenous kinase domain responding to changes in cAMP levels brought forth by Gi/Gs regulation of adenylyl cyclase.

PLC activation has no effect on TRPM7 current

Takezawa et al. 2004

Channel pore Mg2+, Mg2+-ATP

TRPM7 regulates Mg2+ homeostasis and divalent cation fluxes based on metabolic state of the cell

Increased intracellular [Mg2+] strongly suppress the TRPM7 mediated current

Nadler et al. 2001

Direct/indirect ?

Beta actin TRPM7 binds to actomyosin cytoskeleton via beta actin

IP: beta-actin was present in a complex with TRPM7

Clark et al. 2006

Alpha kinase domain

Myosin IIA TRPM7 α-kinase phosphorylates Myosin IIA in a Ca2+

dependent manner. This interaction affects actomyosin cytoskeleton.

TRPM7 mediated Ca2+ influx enhances TRPM7/Myosin IIA interaction via active kinase domain. Active kinase is required as in kinase dead TRPM7

Clark et al. 2006

Page 35: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

25  

mutants do not interact with Myosin IIA.

Alpha kinase Activity

Annexin I TRPM7 phosphorylation of annexin I may have a role in cell death

TRPM7 phosphorylates annexin I at a conserved serine-5 residue within the amphipathic alpha-helix region.

Dorovkov and Ryazanaov, 2004

N-terminal region

Snapin, synapsin I, synaptotagmin I

TRPM7 vesicular channel activity is essential for neurotransmitter (Ach) release in sympathetic neurons

TRPM7 forms molecular complexes with synaptic vesicle proteins synapsin I, synaptotagmin I and snapin (direct) and mediates acetylcholine release

Krapivinsky et al. 2006

Alpha kinase domain

eEF2 kinase TRPM7 mediates eEF2 phosphorylation through eEF2-K under low Mg2+ availability

Indirect influence via phosphorylation of eEF2-K at Ser77. Phosphorylated eEF2-K in turn phosphorylates eEFT2 at Thr-56

Perraud et al. 2011

1.4 Cofilin – the actin depolymerizing factor

The actin cytoskeleton is a dynamic structure that forms the structural framework of a

cell; it mediates cell attachment, migration, division, and death. In addition, the cellular

cytoskeleton allows cells to respond to their local environment by migrating toward, or away

from chemotactic cues. The cytoskeleton organizes the localization of cellular organelles; it also

contributes to intracellular transport, and cell polarity. Thus, regulation of the actin cytoskeleton

is a fundamental component of cellular physiology. The actin depolymerizing factor

(ADF)/cofilin family of proteins are essential regulators of the actin dynamics. During actin

Page 36: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

26  

treadmilling ATP bound G-actin monomers are added to the barbed end of actin filaments while

ADP-bound G-actin monomers dissociate from the pointed end thereby enabling the

unidirectional movement of actin filaments. ADF/cofilin family members enhance actin filament

dynamics by contributing to the maintenance of the actin monomer pool through

depolymerisation and dissociation of F-actin (Carlier et al. 1997). However, cofilin can also

promote actin polymerization and nucleation depending on the amount of available active cofilin

(Ichetovkin et al. 2002). Cofilin phosphorylation, by one of its upstream regulators, results in an

inactive cofilin, whereas dephosphorylation results in cofilin activation (Andrianantoandro and

Pollard, 2006). Cofilin activity is essential in development (Gurniak et al. 2005) and has been

implicated in numerous neuronal pathologies including Alzheimer’s disease (Maciver and

Harrington 1995) and stroke (Minamide et al. 2000; Bent 2011).

1.4.1 Cofilin structure and expression

The ADF/cofilin family of proteins are ubiquitously expressed throughout eukaryotes

(Bamburg, 1999). These are relatively small actin binding proteins (19kDa) that play essential

roles in regulating actin dynamics. ADF/cofilin proteins are characterized by the presence of

highly conserved actin binding domains and a single regulatory phosphorylation site on Ser-3

(Bamburg, 1999). In addition, ADF/cofilin proteins contain two PIP2 binding domains and a

nuclear localization sequence (NLS) (Bamburg, 1999). The mammalian ADF/cofilin family can

be divided into three homologous members: cofilin-1, also known as non-muscle cofilin; cofilin-

2, also known as muscle specific cofilin; and ADF (van Troys et al, 2008). The expression

patterns of these proteins vary across development in a tissue specific manner. For instance,

Page 37: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

27  

cofilin 1 is the predominant isoform during development and it remains ubiquitously expressed

throughout adulthood (Vartiainen et al. 2002, Gurniak et al. 2005). During late embryogenesis

and after birth cofilin 2 replaces cofilin 1 in striated muscle thereby forming the only isoform to

be expressed in skeletal muscle and the predominant isoform in cardiac muscle (Nakashima et al.

2005). Whereas, ADF expression increases post-natally, particularly in epithelial and endothelial

cells (Vartiainen et al. 2002). Despite the fact that multiple cofilin isoforms can be found in

certain cells/tissues, they appear to have distinct functional roles, particularly during

development. For instance, cofilin 1 is critical in development, especially for development of the

nervous system as cofilin 1 knockout mice exhibit defects in neural tube closure, impaired neural

crest cell migration and are embryonically lethal (Gurniak et al. 2005). These Cofilin 1

knockouts had no detectable levels of cofilin2 however they did show a 3 to 4 fold increase in

ADF expression (Gurniak et al. 2005). Given that the enhanced ADF expression did not recover

the nervous system dysfunction nor the embryonic lethality suggests that these actin binding

proteins are not functionally redundant and that they play distinct roles in development (Gurniak

et al. 2005). Further evidence comes from ADF knockout studies, where mice lacking ADF

expression are viable and undergo normal development. However, shortly after birth these ADF

knockouts exhibit hyper-proliferation of the corneal epithelium leading to corneal thickening and

blindness (Ikeda et al. 2003). Despite the presence of 3 ADF/cofilin isoforms in mice and

humans, we will focus on the predominant isoform cofilin 1 which we will simply refer to as

‘cofilin’.

Page 38: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

28  

1.4.2 Cofilin and the regulation of actin dynamics

Actin is an integral member of the cytoskeleton and it is normally found in two cellular

forms: the unbound globular actin (G-actin) monomer, and the bound or filamentous actin (F-

actin) polymer form. Actin filaments are double helical polymers composed of globular subunits

arranged head-to-tail to give the filament molecular polarity. Under steady state conditions F-

actin undergoes rapid polymerization on the barbed end and depolymerisation on the other end,

called the pointed end (Pollard and Borisy, 2003). ATP bound actin monomers (ATP-G-actin)

are added on the barbed end. Once incorporated into F-actin the monomers undergo hydrolysis

of the bound ATP to ATD and Pi (intermediate state). Dissociation of Pi initiates disassembly

reactions by inducing filament debranching and the binding of cofilin to ADP-loaded actin,

which in turn promotes filament dissociation at the pointed end (Pollard and Borisy, 2003). The

ADP-bound actin monomers can re-enter the polymerization cycle once they are ATP bound

(Pollard and Borisy, 2003). This polarized growth and turnover of actin filaments is an integral

component of cell division, migration and the formation of cellular protrusions including

lamellipodia, filopodia, podosomes, and growth cones (Pollard and Borisy, 2003). In these

dynamic structures actin filaments are organized in polarized arrays that exhibit treadmilling

with the barbed ends facing outward towards the edge of the cell pushing the cell membrane

forward as they undergo polymerization (Pollard and Borisy, 2003). Actin filament turnover can

be enhanced through de novo synthesis, increasing the number of filament ends and/or by

increasing the available monomer pool (Pollard and Borisy, 2003).

Actin filaments can turn over by themselves, but they do so at a much slower rate than

needed for cell movement indicating that other factors must contribute to actin dynamics

(Andrianantoandro and Thomas, 2008).Cofilin, profilin, Arp2/3, tropomyosin, and capping

Page 39: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

29  

proteins are some of the actin binding proteins (ABPs), which contribute to the regulation of

actin dynamics. Cofilin can bind both filamentous actin and actin monomers. In filamentous

actin, each cofilin molecule binds two actin monomers at the cleft between them (van Troys et

al. 2008). Cofilin preferentially binds to ADP/Pi and ADP bound actin over ATP-bound: cofilin

promotes filament severing and dissociation on the pointed end of F-actin (van Troys et al.

2008). In addition, cofilin binds actin cooperatively; as a result cofilin bound actin subunits are

clustered on actin filaments (Bamburg et al. 1999). In both severing and depolymerisation,

cofilin binding is believed to induce a twist in the actin filament, which through long range

effects destabilizes the actin-actin interactions thereby leading to filament fragmentation

(McGough et al. 1997; Bobkov et al. 2006).

Although the actin severing and depolymerizing function of cofilin are well accepted,

much debate exists regarding the functional role of cofilin in regulating actin dynamics. Initially

it was suggested that cofilin activity, during steady state actin polymerization, contributes to the

generation of the actin monomer pool through depolymerisation of actin filaments on the pointed

end (Carlier et al. 1997). However, follow up studies revealed that cofilin activity contributes to

actin polymerisation by severing actin filaments and exposing new barbed ends that could be

subsequently polymerized (Ichetovkin et al. 2002). Both models are not mutually exclusive,

rather they are thought to operate under different conditions. One factor that determines the

effects of cofilin on actin filaments is the proportion of the available pool of active cofilin

monomers given that a low cofilin pool favours actin filament severing whereas a high cofilin

pool favours actin filament nucleation (Andrianantoandro and Pollard, 2006). When a few

cofilin molecules bind to actin (cofilin:actin ≤ 1:2) the number of torsionally strained interfaces

between twisted filament regions is large thereby leading to filament severing to relieve the

Page 40: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

30  

torsional stress (Bobkov et al. 2006). However, when F-actin is decorated by cofilin

(cofilin:actin ≈ 1:1 ) the number of torsionally strained interfaces decreases leading to overall

enhanced filament stability (Bobkov et al. 2006; Andrianantoandro and Pollard, 2006). In this

case, filament severing is not observed but disassembly occurs at the pointed end at rates

equivalent to those observed by undecorated F-actin filaments (van Troys et al. 2008). At very

high cofilin concentrations (cofilin: actin > 2:1), G-actin binding and de novo nucleation are

observed leading to filament assembly (Andrianantoandro and Pollard, 2006).

1.4.3 Regulation of cofilin activity

Cofilin activity can be regulated through phosphorylation, and interaction with

intracellular proteins such as PIP2 and other ABPs. Cofilin phosphorylation interferes with its

ability to bind actin (Wriggers et al. 1998). Thus, phosphorylation of the N-terminal Ser-3 leads

to cofilin inactivation, whereas dephosphorylation of Ser-3 residue leads to activation (Bamburg,

1999). This phospho-regulation is finely tuned through a number of signalling pathways that

control the activity state of several key regulatory kinases and phosphatases. These key

regulatory proteins include LIM (Lin-11, Isl1, and Mec-3) kinases (LIMK) and testicular kinases

(TESK), which inhibit cofilin through phosphorylation of Ser-3 ( Edwards et al 1999; Toshima

et al. 2001) and slingshot family phosphatases (SSH) and chronophin phosphatase (CIN) that

reactivate cofilin through dephosphorylation of Ser-3 (Ohta et al. 2003;Mizuno et al. 2013).

Membrane phosphoinositides, particularly PIP2 and PIP3 are well known for their ability to bind

cofilin; this interaction interferes with cofilin’s ability to bind actin due to an overlap between

the G/F-actin and phosphoinositide binding sites (Gorbatyuk et al. 2006). Given that PIP2 binds

Page 41: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

31  

cofilin with similar affinity irrespective of its phosphorylation state, this represents a regulatory

mechanism that is distinct from that of cofilin phospho-regulation (Gorbatyuk et al. 2006). The

formation of PIP2 rich microdomains in the inner leaflet of the plasma membrane could function

to recruit cofilin to the plasma membrane; stimulus induced PIP2 hydrolysis would contribute to

a high amount of cofilin near the membrane to exert its effects on the actin cytoskeleton

(Gorbatyuk et al. 2006; Van Troys et al. 2008). Cofilin activity is also modulated by other actin

binding proteins. For instance, Arp2/3 complex and cofilin function in a synergistic fashion to

promote F-actin polymerization (DesMarais et al. 2005), whereas the binding of tropomyosin to

actin prevents cofilin mediated actin severing (Ono 2007). Most of the ADF/cofilin proteins

exhibit enhanced actin depolymerisation activity under basic pH than they do at neutral or acidic

pH values (Bamburg1999; Bernstein et al. 2000). However, in vertebrates ADF activity is much

more sensitive to changes in pH in comparison to cofilin. ADF cellular distribution is also

sensitive to intracellular pH, since in the presence of an acidic environment (pH<7) ADF

associates with F-actin in the cytoskeleton whereas cofilin localization remains largely

unaffected (Bernstein et al. 2000).

1.4.4 Cofilin in neurodegeneration

Ischemia, excitotoxicity and oxidative stress are key initiators of neurodegenerative

disease and acute CNS injury (Maloney and Bamburg 2007; Aarts et al, 2003). These initiators

can arise from stroke, trauma, or mitochondrial dysfunction and they can be induced or enhanced

by genetic abnormalities (Maloney and Bamburg 2007). A role for cofilin in neurodegeneration

comes from its identification in Hirano bodies, intracellular rod-shaped eosinophilic inclusions

Page 42: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

32  

found in numerous neurodegenerative disease including Alzheimer’s, dementia, and kuru

(Maciver and Harrington 1995). Ischemia and mitochondrial dysfunction have both been shown

to result in cofilin and ADF hyper-activation and the subsequent formation of cofilin-actin rods

(Bamburg 1999, Minamide et al. 2000). For instance, blockage of mitochondrial and glycolytic

ATP production in hippocampal and cortical neurons led to a rapid ATP depletion, in a matter of

minutes, and equally rapid cofilin activation through dephosphorylation (Minamide et al. 2000,

Huang et al, 2008). These changes in cofilin activity were accompanied by rapid actin-cofilin rod

formation in the majority of neurons (>80%) within a similar time frame. Expression of a

constitutively active cofilin mutant clearly showed that rod formation was due to cofilin hyper-

activation (Minamide et al. 2000). Interestingly, repletion of neuronal cultures with ATP resulted

in rapid dissolution of cofilin rods (Minamide et al. 2000; Huang et al. 2008). Given that neurons

consume nearly 50% of their energy through actin turnover (Bernstein et al. 2003), the

generation of the actin-cofilin rods could represent a neuroprotective mechanism that responds to

fluctuations in ATP levels during stress. Indeed Bernstein and colleagues (2006) have shown that

the generation of actin-cofilin rods in rat hippocampal neurons can slow ATP decline during

anoxic stress. Prolonged exposure to neuronal stress (i.e. ROS, glutamate excitotoxicity, anoxia)

results in the presence of persistent cofilin-actin rods, which can grow long enough to span the

diameter of neurites (Minamide et al. 2000). Despite their initial protective effects, the prolonged

presence of the actin-cofilin rods is proposed to disrupt cytoskeletal structure, intracellular

transport and contribute to the degeneration of neurites beyond the rods (Minamide et al. 2000).

In addition to their formation in the presence of energetic stress (ATP depletion), actin-

cofilin rods can form in the presence of ATP. For instance, glutamate or peroxide treatment have

no significant effects on cellular ATP levels, however the formation of actin-cofilin rods is still

Page 43: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

33  

observed (Minamide et al. 2000). Furthermore, unlike the transient rods that form upon ATP

depletion, peroxide and glutamate treatment lead to the formation of persistent rods. The

persistence of cofilin-actin rods could be due to enhanced cross-linking between cofilin

molecules. Cofilin-actin rods generated in response to excitotoxic stress have higher oxidation

levels than those cofilin-actin rods generated by cofilin overexpression (Bernstein et al 2012).

This enhanced oxidation is thought to promote cofilin’s incorporation into rods through

intermolecular disulfide bridges which in turn enhance rod stability (Bernstein et al. 2012).

Given that glutamate excitotoxicity, oxidative stress, mitochondrial dysfunction and ATP

depletion are all consequences of anoxia (Aarts et al. 2003; Maloney and Bamburg 2007) the

generation of persistent actin-cofilin rods could represent a pathological mechanism observed in

the ischemic brain.

In addition to the formation of actin-cofilin rods, cofilin function is also implicated in cell

death. In response to oxidative stress, cofilin has been shown to translocate to mitochondria

where it mediates the opening of the permeability transition pore resulting in cytochrome C

release and ultimately apoptosis (Klamt et al. 2009). Klamt et al. (2009) suggested that oxidation

of cofilin causes it to lose its affinity for actin and translocate to the mitochondria, moreover,

dephosphorylation of the regulatory Ser-3 is essential for this cofilin mediated translocation

(Klamt et al. 2009). The above finding suggests that sequestration of cofilin into rods in response

to transient stress could serve as a neuroprotective measure to circumvent the induction of

apoptosis. Consistent with this notion, Bernstein et al (2006) revealed that the formation of

cofilin-actin rods hindered mitochondrial membrane potential decline. However, the regulatory

mechanisms that induce actin-cofilin rod formation and disassembly still need to be determined.

Page 44: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

34  

1.5 Rho associated kinases (ROCKs)

Rho-associated kinases (ROCKs) are the first and best characterized effector proteins of

Rho-GTPases. ROCKs are serine/threonine kinases which consist of an amino-terminal kinase

domain followed by a coiled-coil region, and a pleckstrin homology (PH) domain at the

carboxyl-terminus (Nakagawa et al. 1996). The coiled-coil region contains a Rho-binding

domain (RBD) which is thought to interact with other α-helical proteins, whereas the PH domain

in the carboxyl terminus is thought to be involved in protein localization (Riento and Ridley,

2003). The PH domain and the RBD form the autoinhibitory loop that folds back onto the amino

terminus where it inhibits ROCK activity by interacting directly with the kinase domain (Amano

et al, 1999). Cleavage of the C-terminal end results in a constitutively active ROCK, consistent

with the inhibitory role of the C-terminal end of ROCK (Leung et al. 1996). Their closest

homologue is myotonic dystrophy protein kinase (DMPK) with which they share the above

mentioned structural features.

ROCK proteins exist as two closely related isoforms: ROCK1 and ROCK2 which show

the highest homology in their kinase domains and lowest homology in the coiled-coil domain

(Nakagawa et al, 1996). The diversity within the coiled-coil domain could serve in differential

regulation of ROCKs as well as subcellular localization through interactions with other

regulatory proteins. Both ROCK1 and ROCK2 are ubiquitously expressed in mouse and rat

tissues; ROCK2 is largely expressed in muscle and brain whereas ROCK1 expression is highest

in the liver, spleen, kidney and testis (Nakagawa et al. 1996). These isoforms also exhibit

differential expression within the CNS, with ROCK1 expression observed in astrocytes whereas

ROCK2 expression observed in neurons, especially in presynaptic and postsynaptic elements

Page 45: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

35  

(Iizuka et al. 2012). The variation in tissue distribution and subcellular localization suggest that

ROCK1 and ROCK2 may play distinct functional roles (Riento and Ridley 2003).

1.5.1 ROCK protein signalling

The Rho subfamily proteins (RhoA, RhoB, RhoC) are the only known GTPases that can

activate ROCK proteins in vivo (Yoneda et al. 2005). These Rho proteins bind to the RBD of

ROCK only in the activated (GTP-bound) form. This interaction between Rho and the RBD is

believed to induce conformational changes that diminish ROCK autoinhibition by exposing the

kinase domain (Riento and Ridley, 2003). In addition to its regulation by upstream Rho-

GTPases, ROCK protein activity is also regulated through Rho independent mechanisms. For

instance, arachidonic acid and phosphoinositides PIP2 and PIP3 have been reported to activate

ROCK2, suggestive that interactions with lipids induce conformational changes that expose the

kinase domain of ROCK2 (Feng et al. 1999; Yoneda et al. 2005). These findings also reveal that

ROCK1 and ROCK2 can be regulated through distinct mechanisms. Further support for this

differential regulation comes from other studies that have shown that Rho E specifically inhibits

ROCK1 (Riento et al. 2003). In addition, during apoptosis caspase-3 mediated cleavage of the

ROCK1 C-terminal end results in a constitutively active kinase (Coleman et al. 2001).

ROCK proteins phosphorylate numerous downstream targets that regulate the cellular

cytoskeleton. Consequently ROCKs are implicated in regulating cell adhesion, motility, division

and cell death (Riento and Ridley 2003). Myosin light chain phosphatase (MLCP), LIMK,

Ezrin/Radaxin/Moesin (ERM) and intermediate filament proteins are well known ROCK

substrates. For instance, site directed mutagenesis studies have shown that ROCK1 and ROCK2

Page 46: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

36  

phosphorylate LIMK1 and LIMK2 at Thr-508 and Thr-505 residues, respectively, and that these

phosphorylation events are essential for LIMK activity (Ohashi et al. 2000; Sumi et al. 2001).

For many of these substrates only one ROCK isoform has been tested, however given that the

ROCK1 and ROCK2 kinase domains are 92% identical (Nakagawa et al. 1999), both are

believed to phosphorylate the same substrates (Riento and Ridley 2003).

1.5.2 ROCK protein function

ROCK is an important regulator of the actin cytoskeleton through its influence on cell

division, adhesion, motility and death. For instance, ROCK is essential in cell division since

inhibition of ROCK activity prevents cytokinesis in mammalian cells (Yasui et al. 1998). In

fibroblasts, ROCK activity promotes actomyosin contractility given that ROCK activation results

in the formation of stress fibres and focal adhesions through the phosphorylation of MLCP

(Leung et al. 1996; Riento and Ridley 2003). ROCK activity also contributes to stress fiber

formation through phosphorylation of LIMK, which in turn phosphorylates and inactivates

cofilin thereby increasing acting filaments (Amano et al. 2001). In addition to regulating cell-

substratum adhesion, ROCKs influence cell-cell contacts since enhanced ROCK activity

compromises the integrity of tight junctions and adherens junctions (Riento and Ridley, 2003).

ROCKs also regulate numerous apoptotic events. Plasma membrane blebbing, a signature feature

of apoptotic cells, is regulated by myosin light chain phosphorylation and actomyosin

contraction (Mills et al. 1998). Caspase-3 mediated activation of ROCK1 has been shown to be

sufficient and necessary in promoting blebbing through enhancing myosin light chain

phosphorylation and actomyosin contractility (Shi and Wei, 2007). On the other hand, a decrease

Page 47: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

37  

in ROCK activity appears to mediate phagocytic uptake of apoptotic cells (Tosello-Trampont et

al. 2003).

1.6 P-21 Activated Kinases (PAKs)

1.6.1 The structure and expression of P-21 activated kinases (PAKs)

P21 activated kinases (PAKs) are a family of serine/threonine protein kinases consisting

of 6 different isoforms. These isoforms are subdivided into two groups based on sequence

similarities, as well as structural and biochemical properties; Group I consists of PAK 1-3 and

Group II consists of PAK 4-6 (Eswaran et al. 2008). All PAK isoforms contain an N-terminal

regulatory domain and a C-terminal kinase domain. The regulatory domain of group I PAKs

consists of a highly conserved p21- binding domain (PBD) that overlaps with an auto-inhibitory

domain (AID) (Kreis and Barnier, 2008). PAKs are the main kinase effectors for the Rac1 and

Cdc42 family of GTPases. Thus, Rac1 or Cdc42 binding to the PBD causes a conformational

shift that dissociates the AID from the catalytic domain and allows for its autophosphorylation

which is required for full kinase activity (Benner et al. 1995, Lei et al. 2000). In contrast to

Group I, Group II PAKs do not have an AID domain and thus undergo different regulation.

Indeed, Rac or Cdc42 binding does not substantially enhance kinase activity of Group II PAKs,

instead Rac/Cdc42 interaction is important for the translocation of Group II PAKs to specific

intracellular compartments (Arias-Romero and Chernoff, 2008). These findings in conjunction

with sequence variation between Group I and Group II PAKs indicate that these two groups are

differentially regulated (Arias-Romero and Chernoff 2008). Our focus will be on group I PAKs

with a particular emphasis on PAK3.

Page 48: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

38  

Expression analysis reveals distinct tissue and cellular distribution patterns for Group I

PAKs. For instance, PAK 1 is highly expressed in the brain and spleen, PAK3 is mainly

expressed in the brain, and PAK 2 shows ubiquitous expression (Kreis and Barnier 2009).

Although all three Group 1 PAKs are expressed in the brain, they show differential expression

with PAK1 expression found in neurons, astrocytes and oligodendrocytes, and PAK3 expression

mainly found in neurons (Kreis et al 2008, Kreis and Barnier 2009). Thus, despite the high

sequence similarity between Group I PAKs, these proteins appear to be involved in different

biological processes. This is especially evident from loss-of function mutants in which PAK1

mutants display immune system impairments while PAK3 mutants display abnormal synaptic

plasticity and learning deficiencies (Hofmann et al. 2004, Meng et al. 2005).

1.6.2 Regulation of PAK activity

Group I PAKs are the main effector proteins of the Rac and Cdc42 family of GTPases.

Although they have been shown to be activated by numerous Rac/Cdc42 members, Group 1

PAKs were recently shown to exhibit differential selectivity for their activators. For instance,

PAK3 was recently shown to have higher affinity for Cdc42 than for Rac1 (Kreis et al. 2007).

Given that the PBD is highly conserved among Group 1 PAKs, the substrate specificity might

arise from regions surrounding PBD that might participate with GTPase interactions (Reeder et

al. 2001), and/or through GTPase interactions with PAK binding partners such as guanine

exchange factor cool/pix (Kreis and Barnier 2009). Group 1 PAKs are fully active once

phosphorylated. The interaction of Rac/Cdc42 with PAK induces a conformational change in the

AID which activates the catalytic domain that performs the autophosphorylation of key residues

Page 49: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

39  

in the regulatory region important for enzymatic activity (Chong et al. 2001). In this model, the

key event involves phosphorylation of a Threonine residue in the activation loop of Group 1

PAKs which in turn mediates the autophosphorylation (Chong et al. 2001). Chong and

colleagues (2001) identified S-139 and Thr-421 as the phosphorylation sites essential for PAK3

activity. In addition to the Rac/Cdc42 mediated activation of Group 1 PAKs, they can also be

activated through GTPase independent mechanisms. For instance, Group 1 PAKs have been

shown to be activated by the lipid sphingosine (Bokoch 2003; Chong et al. 2001).

1.6.3 PAK protein function

Myosin light chain kinase (MLCK), Mitogen activated kinase (MAPK), Bcl-2/Bcl-XL-

antagonist causing cell death (BAD) and LIM kinase (LIMK) are a few of the well characterized

downstream targets of Group I PAKs (Arias-Romero and Chernoff, 2008). Given the diverse

range of downstream targets, Group 1 PAKs have been implicated in a wide range of biological

processes including cell cycle progression, neuronal cell fate determination, differentiation,

neurite outgrowth, axonal guidance, synaptic plasticity, survival and apoptosis (Kreis and

Barnier, 2009). Despite the high sequence similarity among Group 1 PAKs, numerous studies

have described the involvement of specific Group I PAKs in distinct cellular processes. For

instance, PAK1 activation promotes cell survival whereas PAK3 activity is implicated in cell

death (McPhie et al. 2003). Using an Alzheimer’s disease mutant model, McPhie et al (2003)

reported the specific involvement of PAK3 in amyloid precursor protein (APP) mediated

apoptosis, an effect that was abolished using a dominant negative kinase mutant of PAK3 or by

deletion of the APP binding domain of PAK3. In neurons the establishment of polarity is

Page 50: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

40  

essential for correct migration and the specification of dendrites and axons. A role for PAK1 in

neuronal polarity has been proposed by Rosso et al. (2004), they suggested PAK1 activation and

subcellular localization promote neurite outgrowth by regulating the actin cytoskeleton via

phosphorylation of LIMK1. On the other hand Cobos et al. (2007) reported a role for PAK3 in

inhibiting neurite growth and promoting the migration of immature GABAergic interneurons as

they migrate tangentially to the cortex.

PAK3 appears to be involved in regulating synapse structure and function. The

phosphorylated (active) form of PAK3 has been observed in dendritic spines (Chen et al. 2007).

Theta burst stimulation markedly increased the numbers of spines containing phosphorylated

PAK3 and phosphorylated cofilin (Chen et al. 2007). More importantly this enhanced

phosphorylation of PAK3 and cofilin corresponded with enlarged synapses thereby implicating

PAK3 activity with synaptic plasticity (Chen et al. 2007). Alterations in PAK3 activity have

also been linked to the pathogenesis of Alzheimer’s disease (AD) and mental retardation. For

instance, a mouse model of Alzheimer’s disease revealed a significant reduction in PAK1 and

PAK3 expression and activity (Zhao et al. 2006). Pharmacological inhibition of PAK activity in

normal mice was sufficient to induce cofilin pathology and memory impairment similar to

Alzheimer’s disease thereby suggesting that PAK signalling is involved in the synaptic

dysfunction and cognitive impairments (Zhao et al. 2006). In addition to AD pathology, PAK3 is

linked to non-syndromic mental retardation with five identified genetic mutants affecting

effector binding and kinase activity (Kreis and Barnier 2009). PAK3 knockout mice reveal

impairments in synaptic plasticity, as well as in learning and memory consistent with the notion

that PAK3 plays an essential role in regulating synapse function and cognition (Meng et al.

2005)

Page 51: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

41  

1.7 The Slingshot phosphatase (SSH) family

1.7.1 Slingshot phosphatase structure

Slingshot phosphatases (SSHs) are novel family of protein phosphatases that can

specifically dephosphorylate and activate cofilin. In humans and mice the SSH family consists of

three members SSH1, SSH2 and SSH3, each of which has multiple isoforms with the long

isoforms named SSH-1L, SSH-2L and SSH-3L, respectively (Ohta et al. 2003). Unless making

distinctions between the different SSH isoforms I will use the terms SSH1, SSH2, SSH3, to refer

to SSH1L, SSH2L and SSH3L respectively. All 3 SSH members exhibit ubiquitous expression

through many of the tissues examined, with distinct tissue expression patterns between members.

For instance, SSH-1L is highly expressed in the brain, kidney and thymus whereas SSH-2L is

highly expressed in the thymus, testis, heart and brain, and SSH-3L is only expressed in the brain

(Ohta et al. 2000). SSH proteins contain three highly conserved regions starting from the N-

terminal non-catalytic (SSH-N) domain, consisting of subdomain A and B, followed by the

catalytic P subdomain (Kurita et al. 2008). The P-subdomain itself has basal phosphatase activity

but no actin binding ability (Kurita et al. 2008, Yamamoto et al. 2008). However, the B and P

subdomains are essential for phospho-cofilin binding by SSH1 and the cofilin phosphatase

activity of SSH1 (Kurita et al. 2008). The A subdomain, on the other hand is required for F-actin

mediated activation of SSH1 but not for phospho-cofilin binding (Kurita et al. 2008). Unlike the

highly conserved N-terminal region, the C-terminal end of SSH family members is fairly

variable with the exception of the S-domain, a Serine rich region conserved between mammalian

SSH1 and SSH2 but not found in SSH3 (Mizuno et al. 2013). The S-domain of SSH1 and SSH2

contains an actin binding region as well as conserved residues that regulate phosphatase activity

Page 52: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

42  

(Yamamoto et al. 2006; Soosairahaj et al. 2005). To date SSH1 has 3 defined F-actin binding

regions including an LKR motif within the S-domain, an LHK motif within the B-subdomain,

and Trp-458 flanking the P-subdomain (Yamamoto et al. 2006). Deletion of either the B-

subdomain or S-domain reduces acting binding activity by nearly 50% whereas mutation of Trp-

458 completely abolishes actin binding activity suggesting that the latter residue is essential for

actin binding (Yamamoto et al. 2006).

1.7.2 Regulation of SSH1 activity

Although all three SSH members dephosphorylate phospho-cofilin at the Ser-3 position,

SSH3 is less effective than the other two (Ohta et al. 2003). In addition, SSH3 shows no actin

binding capacity, unlike SSH1 and SSH2 who have been reported to tightly bind to actin (Ohta et

al. 2003). Kurita et al. (2008) previously reported that F-actin binding enhances the cofilin

phosphatase activity of SSH1 by 1200 fold in comparison to a 2.2 fold increase in basal

phosphatase activity towards pNPP, an artificial phosphatase substrate typically used in

phosphatase assays. In addition to the A-subdomain, Trp-458 has also been reported to be

essential for the F-actin mediated SSH1 activation (Nishita et al. 2005). Given the reported actin

mediated activation of SSH1, it is likely that the enhanced cofilin phosphatase activity of SSH2,

in comparison to SSH3, is due to the actin binding activity of the former and not the latter.

In addition to the F-actin binding mediated activation of SSH1, SSH1 activity is also

regulated by phosphorylation and interaction with 14-3-3, a cytosolic scaffolding protein

(Yamamoto et al. 2006; Soosairajah et al. 2005). Incubation of PAK4 with a C-terminally

truncated SSH1 mutant (residues 1-535 out of 1049) has been shown to increase the

Page 53: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

43  

phosphorylation of this SSH1 mutant (Soosairajah et al. 2005). Although the identity of the

phosphorylated residues is unknown, this phosphorylation of SSH1 decreases its ability to

dephosphorylate LIMK and cofilin, two known substrates of SSH1 (Soosairajah et al. 2005). In

addition, phosphorylation of Ser-937 and Ser-978 within the S-domain of SSH1 promotes 14-3-3

binding (Nagata-Ohashi et al. 2004). This interaction of 14-3-3 with SSH1 is thought to interfere

with the ability of SSH1 to bind to actin. Indeed, it has been shown that pre-incubation of SSH1

with 14-3-3ζ results in a decrease in F-actin binding for SSH1 (Soosairajah et al. 2005) and its

cofilin phosphatase activity (Nagata-Ohashi et al. 2004). Calcineurin (CaN) and protein kinase D

(PKD) have been proposed as potential regulators of SSH1 activity. PKD has been shown to

result in Ser-937 and Ser-978 phosphorylation of SSH1 (Peterbus et al. 2009) whereas CaN has

been reported to dephosphorylate and activate SSH1 although it’s unclear whether Ser-937 and

Ser-978 are among the dephosphorylated residues (Wang et al. 2005). In contrast to 14-3-3

interaction, SSH binding with acting binding proteins, coronin-1B or coronin-2A, is thought to

promote SSH1 activity and regulate lamellipodia dynamics at the leading edge of migrating cells

(Mizuno et al. 2013).

1.7.3 SSH1 function

As previously mentioned cofilin plays an important role in regulating the actin

cytoskeletal dynamics. The functional modulation of cofilin activity is performed by kinases

such as LIMK and TESK which inhibit cofilin via Ser-3 phosphorylation, and by phosphatases

such as SSH and CIN, which active cofilin. Given the various cofilin activators and inhibitors,

the overall effect on cofilin activity is a consequence of the spatiotemporal regulation of the

Page 54: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

44  

numerous activators and inhibitors. Indeed, the regulation of SSH1 and LIMK has shown to

influence numerous cofilin mediated cellular processes such as cell migration, chemotaxis, and

cell division (Mizuno et al. 2013). Nishita et al. (2005) provide an exquisite model of SSH1 and

LIMK1 spatiotemporal regulation in directional cell migration. Based on data from knockdown

studies of LIMK1 and SSH1 in chemokine stimulated Jurkat T cells, the authors propose that

LIMK1 is initially activated to promote lamellipodia formation followed by SSH1 activation to

prevent membrane extensions in one direction while allowing local cofilin activity to the leading

edge of the migrating cell (Nishita et al. 2005). SSH1 and LIMK1 have also been shown to

function in a complementary fashion during cell division. Early in the cell cycle LIMK1 is

hyper-activated while SSH1 is inhibited, but as cells enter telophase and cytokinesis LIMK1 is

inhibited while SSH1 activity is increased (Amano et al. 2002; Kaji et al. 2003). Both of these

changes in LIMK1 and SSH1 activity correspond well with the enhanced cofilin phosphorylation

observed early in the cell cycle and its progressive decrease in phosphorylation as it enters

telophase and cytokinesis (Amano et al. 2002; Kaji et al. 2003).

Page 55: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

45  

Chapter 2

Objectives and Hypothesis

TRPM7 channels have been linked to neuronal death both in vitro and in vivo models of

ischemia (Aarts et al. 2003; Sun et al. 2009). The founder study by Aarts and colleagues (2003)

showing that TRPM7 channels play a more important role in ischemic cell death, particularly

during prolonged ischemia (>1.5 hours) led our research group into the identification of potential

signalling relationships operating downstream of TRPM7 activation. This search led to the

identification of cofilin-1, which was shown to be hyperactivated downstream of TRPM7

activity during anoxia; furthermore this enhanced cofilin activity contributed to neuronal death.

Inhibition of TRPM7 activity through Gd3+ or suppression of TRPM7 expression via TRPM7

specific siRNA attenuated cofilin hyperactivation during OGD (Bent 2011). Furthermore,

TRPM7 activity during anoxia corresponded with LIMK inhibition, an effect that was relieved

following TRPM7 inhibition (Bent 2011). Given the observation that LIMK – cofilin signalling

pathway operated downstream of TRPM7 the primary focus of this study is to identify potential

regulators of LIMK and cofilin activity that operate downstream of TRPM7. Rho-kinases are

known regulators of LIMKs (Amano et al. 2001) and recent findings implicate Rho-associated

kinases as contributors of ischemic neuronal cell death (Yamashita et al. 2007; Gisselson et al.

2009; Jeon et al. 2013) Thus, I hypothesize that TRPM7 activity during OGD influences ROCK2

protein activity. However, p-21 activated kinases are also known regulators LIMK function

(Arias-Romero and Chernoff 2008). Given that dysregulation of PAK3 is linked to synaptic

impairments I hypothesize that TRPM7 activity influences PAK3 activity during OGD. In

Page 56: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

46  

addition, Slingshot-family phosphatase 1 (SSH1) is known to regulate cofilin and LIMK (Kurita

et al. 2008; Soosairajah et al. 2005). Furthermore, oxidative stress and increased intracellular

Ca2+ have been shown to promote the cofilin phosphatase activity of SSH1, suggestive that

SSH1 activity is enhanced by an ischemic microenvironment (Wang et al. 2004; Kim et al.

2009). Based on the previous reports I hypothesize that TRPM7 activity during OGD enhances

SSH1 activity.

The experiments of this study will be split into three aims. First I will establish a neuronal

cell death model that confirms the previous findings of Aarts et al. (2003). This model will then

be used to evaluate the neuroprotective effects of ROCK2 during TRPM7 channel activity in

OGD. Furthermore, this cell death model will also be used to evaluate the cell death signalling

mechanisms that operate during OGD. Secondly, I will evaluate the effects of TRPM7 activity

on ROCK2, PAK3 and SSH1 using phospho-specific antibodies targeting sites that influence

protein activity. Lastly, to determine whether phospho-PAK3 and phospho-SSH1 localization is

influenced by TRPM7 activity during OGD, immunofluorescent images labelling the above two

proteins will be taken during normoxic conditions and compared to those of OGD and TRPM7

inhibition. Co-labeling with LIMK1, should expose any differential localization between LIMK

and its upstream regulators.

Page 57: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

47  

Chapter 3

3. Materials and Methods

3.1 Antibodies

Table 3: The antibodies used for western blotting and immunofluorescence analysis are listed at

their respective dilutions.

Product description

Specificity Clonality Dilutions used

Species reactivity

Manufacturer

Rabbit Anti-PAK3 (phospho S154) Antibody

Detects endogenous levels of PAK3 when phosphorylated at Ser-154 in humans (Ser139 orthologue in rats)

Polyclonal WB (1:1000) ICC (1:200)

Human Rat

Abcam

Rabbit Anti-PAK3 Antibody

Detects endogenous levels of total PAK3 protein

Monoclonal WB (1:1000) ICC (1:250)

Human Rat Mouse

Millipore

Rabbit Anti-ROCK2 (phospho T249) Antibody

Detects endogenous levels of ROCK2 when phosphorylated at Thr-249

Polyclonal WB(1:1000) ICC(1:200)

Human Rat Mouse

Abcam

Rabbit Anti-ROCK2 Antibody

Detects endogenous levels of ROCK2

Polyclonal WB(1:1000) IF(1:200)

Human Rat Mouse

Abcam

Rabbit Anti-Slingshot-1L (phospho S978)

Detects endogenous levels of Slingshot-1L phosphorylated at Ser-978.

Polyclonal WB (1:1000) IF (1:200)

Human Rat Mouse Chicken

ECM Biosciences

Rabbit Anti-Slingshot-1L (C-terminal region)

Detects endogenous levels of total Slingshot 1L

Polyclonal WB(1:1000) IF(1:200)

Human Rat Mouse

ECM Biosciences

Mouse Anti-LIMK1 Antibody

Detects endogenous levels of total LIMK1

Monoclonal IF (1:200) Human Rat Mouse

Abcam

WB = western blot; IF = Immunofluorescene

Page 58: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

48  

3.2 Rat Primary Neuronal Culture

Cortical neurons were isolated from embryonic day 18 (E18) Spargue-Dawley rats as

described by Choi et al. (1987), but with minor modifications. In short neurons were removed

from the uterus and placed in chilled DMEM (Invitrogen). Embryos were then removed from

their amniotic sac and placed in a petri dish containing chilled DMEM (Invitrogen). Using iris

microscissors, a horizontal incision was made along the dorsal-midline at the brain stem – spinal

cord border. The same incision was then used to make a vertical cut upwards the nose along the

midline skull suture. The iris microscissors were used to apply pressure along the line between

the eye and the ear to push the brain out of the skull. The olfactory bulbs were removed and the

two hemispheres were spread apart along the midline. An incision was made to cut each cortical

hemisphere from the striatum. The meninges were then removed and cortices collected in a six-

well plate containing chilled DMEM. Following collection of all the cortices, the DMEM was

removed, and cortices were incubated at 37˚C for 7 minutes with pre-heated (37˚C) Trypsin-

EDTA (0.05%, Invitrogen). After incubation, the Trypsin-EDTA solution was aspirated and the

cortices were washed twice in plating media (MEM supplemented with 10% fetal bovine serum,

10% heat-inactivated horse serum, and 2M dextrose). This was followed by tituration with a

flamed glass Pasteur pipette to dissociate the cells. The resultant homogeneous mixture of cells

was then filtered through a 0.75µm cell strainer, into fresh media. Tryphan blue staining and a

haemocytometer were used to determine the concentration of live cells. The cells were diluted in

media consisting of 50% plating media and 50% growth media (Neurobasal supplemented with

B-27 supplement, 1% fetal bovine serum, and 1% glutamine) and were then plated in Poly-D-

Lysine coated Corning CellBind plates (VWR) at a density of 6 x 105cells/cm2 or on Poly-D-

Lysine coated, acid-washed, coverslips at a density of 3.75 x 104cells/cm2. The cells were left

Page 59: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

49  

overnight in a humidified, 5% CO2 incubator at 37˚C. The following day (day in vitro 1 or

DIV1) a half media change was done with growth media, and on DIV4 a half media change was

done with growth media containing 10µM 5-fluoro-deoxyuridine (Sigma) to inhibit glial growth.

Half-media changes were then performed every 3 days until DIV12 when they were used for

experiments.

3.3 In vitro stroke model – Oxygen Glucose Deprivation

Oxygen Glucose Deprivation (OGD) was used as the in vitro model of ischemia. DIV12

neurons were washed three times in degassed, glucose-free Hanks Balanced Salt Solution

(HBSS) and maintained at 37˚C in an anoxic chamber (Thermo EC) with a 5% CO2, and 95% N2

(<0.01% O2) atmosphere. HBSS containing MCN was used to isolate for the ‘TRPM7 like’

currents. MCN consists of MK-801 (10µM, Sigma-Aldrich), CNQX (25µM, Sigma-Aldrich),

and Nimodipine (2µM, Sigma-Aldrich) was used to block glutamate and voltage gated calcium

channels and isolate the ‘TRPM7-like’ currents. Gd3+(10µM) treatment was used to inhibit

TRPM currents and Y-27632(10 µM) was used to block ROCK2 proteins. In control

experiments neurons treated with HBSS alone or with HBSS in combination with MCN, MCN

Gd3+ or MCN Y-27632, were incubated for two hours in a humidified 5% CO2 incubator.

Following the 2 hour treatment (OGD or HBSS) the cells were either placed in fresh growth

media (for cytotoxicity, viability, and apoptosis assay), or fixed (for immunofluorescent

analysis), or proteins were extracted (for western blot analysis). HBSS was comprised of

5.33mM KCl, 0.441mM KH2PO4, 4.17 mM NaHCO3, 137.93 mM NaCl, 0.338mM Na2HPO4,

1.7mM CaCl2, 2.3mM MgCl2 and 5.56mM D-Glucose (all Sigma-Aldrich), with a pH 7.4.

Page 60: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

50  

Glucose and MgCl2 were completely removed from the glucose-free solution which was

subsequently degassed overnight to form the OGD media.

3.4 Protein Extraction

Following the experimental treatment, cells were washed twice in PBS and collected in

pre-chilled microtubes with a cell scraper. The cells were centrifuged at 1000 rpm for 5 minutes.

The supernatant was then removed and the pellet was resuspended in an equal volume of ice-

cold Cell Lytic Mammalian Cell Lysis Reagent (Sigma-Aldrich) containing phosphatase and

protease inhibitors (phosphatase inhibitor cocktail II [Sigma-Aldrich] and protease inhibitor

cocktail [Roche]). The cells were incubated for 15 minutes on ice with mixing at the halfway

point and then centrifuged for 10 minutes at 10,000xg to pellet the unlysed cells and debris. The

supernatant was transferred to a fresh microtube which was then stored at -80˚C for later use.

3.5 DC-Lowry Protein Assay

Cell extracts were quantified in 96 well plates using the DC protein assay (Bio-Rad). Cell

extracts of proteins samples were made in 1/10th dilution. Protein standards ranging from 0.0625

to 1mg/ml were made using bovine serum albumin (Bio-Rad). Five µL of each standard and

sample was loaded into a separate well; followed by the addition of 25µL of reagent A’ and

200µL of reagent B. After 15 minutes incubation at room temperature, absorbance was measured

at 750nm using Synergy-HT (Bio-Tek) microplate reader. If protein concentrations were outside

Page 61: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

51  

the standard curve, the assay was repeated with new protein dilutions to ensure correct

determination of protein concentration.

3.6 SDS – Polyacrylamide Gel Electrophoresis (SDS – PAGE)

Standardized protein extracts of the DIV12 experiments were separated by size using the

Bio-Rad Mini-Protein SDS-PAGE system. The proteins were run on discontinuous gels with a

4% stacking layer and a 7% or 10% separating layer depending on the size of the proteins. The

stacking gel consists of 0.5M Tris-HCl, pH 6.8 and 4% SDS; the separating gel consists of 1.5M

Tris-HCl, pH8.8, and 0.4% SDS. Gels were run at 90V for 20 minutes, until the proteins reached

the separating layer, and then were run at 100V for 90 – 150 minutes. Once running was

complete the proteins were transferred onto 0.4 µm pore-sized nitrocellulose membranes (Bio-

Rad) in 1X Transfer Buffer (292mM Tris, 192mM Glycine, 20% methanol) at 100V for 70

minutes. To verify proper transfer, the nitrocellulose membranes were temporarily stained with

Ponceau S (0.1% w/v Ponceau in 3% v/v acetic acid). The stain was then removed by washing 3

times in TBST for 5 minutes each.

3.7 Western Blotting

Nitrocellulose membranes containing proteins were blocked for 1 hour at room

temperature in TBST containing 5% BSA with gentle agitation. Membranes were then incubated

with primary antibody solution overnight, at 4˚C with gentle agitation. The primary antibody

solutions were made in TBST with 2% BSA (see 3.1 for specific antibody dilutions). The

Page 62: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

52  

following day blots were washed 3 times in TBST (5 minutes each). After washing, the blots

were incubated for 1 hour at room temperature under gentle agitation with secondary antibody

solutions consisting of TBST with 1% BSA and HRP-conjugated secondary antibody goat anti-

mouse (Sigma-Aldrich) or mouse anti-rabbit (ECM biosciences). Blots were washed 5 times in

TBST (5 minutes each), developed with chemiluminescent substrate (Thermo Scientific) and

immediately visualized in ChemiDoc XRS imager (Bio-Rad) using Image Lab software (Bio-

Rad). Densiometric analysis of the bands of interest was performed using NIH ImageJ analysis

software.

3.8 Immunofluorescent Analysis and Imaging

Primary cortical neurons were plated on poly-D-Lysine coated glass coverslips (1µg/µl)

and then subjected to the experimental treatment on DIV12 as outlined in section 3.3. Following

treatment the cells were washed twice in PBS 1X, (Sigma-Aldrich) and then fixed with ice cold

1X PBS containing 4% paraformaldehyde for 15 minutes at room temperature. After fixing, cells

were washed 3 times (5 minutes each) with PBS and permeabilized using PBS containing 0.04%

Tween-20 (Sigma-Aldrich) for 15 minutes at room temperature. Cells were then washed 3 times

(5minutes each) with PBS 1X and blocked in blocking solution (consisting of PBS with 5%

BSA) for 1 hour at room temperature. After blocking, cells were incubated overnight at 4˚C with

primary antibody solution (see 3.1 for specific antibody dilutions) containing the antibody of

interest in PBS with 2% BSA. The following day the cells were washed 3 times (5 minutes each)

with PBS and incubated, for 1 hour at room temperature, with secondary antibody solutions

consisting of PBS with 2% BSA and secondary antibodies (Invitrogen): Alexa Fluor 488 donkey

Page 63: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

53  

anti- rabbit (1:1000) and Alexa Fluor 555 donkey anti-mouse (1:1000). The cells were then

washed 3 times with PBS and coverslips mounted on glass slides using ProLong® Gold anti-fade

reagent with DAPI (Life technologies). Epifluorescent images were captured with an Olympus

IX81 microscope imaging system with a Q-imaging Fast 1394 Rolera-MGi Plus camera using

Metamorph software.

3.9 Cell death, viability, and apoptosis assay

Primary rat cortical neurons were plated at 3 x 105cells/cm2 in 96 well plates and

subjected to 2 hours of HBSS (HBSS, HBSS MCN, HBSS MCN Gd3+) or OGD (OGD, OGD

MCN, OGD MCN Gd3+) treatment on DIV12. Then the treatment solution (HBSS or OGD) was

aspirated and fresh growth media was added to the cells. Cytotoxicity, viability and apoptosis

were measured at 24 hours following treatment, using the ApoTox-Glo Triplex Assay

(Promega). In short, at the 24 hour time point, viability/cytotoxicity reagent was added to each

well followed by 40-50 minute incubation in humidified 5% CO2 incubator at 37˚C. After

incubation cell viability fluorescence was measured at 360Ex/485Em and cytotoxicity fluorescence

was measured at 485Ex/520Em using Synergy-HT (Bio-Tek) microplate reader. Caspase-Glo 3/7

reagent was then added to each well and the cells were again incubated for 40-50 minutes in a

humidified 5% CO2 incubator at 37˚C. Apoptosis was then quantified by measuring

luminescence with Synergy-HT (Bio-Tek) microplate reader.

Cytotoxicity was also measured using Hoechst and Propridium iodide staining in primary

rat cortical neurons plated in 12 well plates. After the experimental procedure on DIV12,

treatment solution (HBSS or OGD) was aspirated and fresh growth media was added to cells,

Page 64: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

54  

which were then placed in humidified, 5% CO2 incubator. After 24 hours, Hoechst and

propidium iodide (PI) was added to the cells incubated for 30 minutes in a humidified, 5%CO2

incubator. Following incubation epifluorescent images (20X) were taken with an Olympus IX81

microscope imaging system with a Q-imaging Fast 1394 Rolera MGi Plus camera using

Metamorph software. Cytotoxicity was quantified by counting the number of dead (PI stained)

cells over the total number of cells (Hoechst stained) in each captured field.

3.10 Statistical Analysis

The data are presented as the mean ± SEM. Statistical comparisons were performed by

either Student’s T-test or one way analysis of variance (ANOVA). Dunnett’s multiple

comparisons test was performed for multiple comparisons in situations where significant

statistical differences were observed. All experiments were repeated at least three times (N=3)

unless otherwise noted.

Page 65: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

55  

Chapter 4

4 Results

4.1 Neuronal survival following OGD: cytotoxicity and viability

Rodent cortical neurons exposed to oxygen glucose deprivation (OGD) for periods up to

1 hour undergo neuronal cell death that is predominantly mediated by excitotoxicity (Goldberg

and Choi, 1993). Excitotoxicity is also the predominant mechanism responsible for ischemic

neuronal death in vivo (Goldberg and Choi 1993). More recently non-excitotoxic mechanisms of

neuronal death, mediated by cell surface and intracellular cation channels have been implicated

in mediating ischemic and anoxic neurotoxicity. Aarts and colleagues (2003) first showed that

although excitotoxicity is an early contributor of ischemic neuronal death, TRPM7 channels are

likely to be more important in neuronal death during prolonged exposure (>1.5 hours) to anoxia.

TRPM7 knockdown or Gd3+ treatment can prevent cell death in primary neurons exposed to up

to 3 hours of anoxia (Aarts et al. 2003). In addition, it has recently been proposed that ROCK

acts as a mediator of excitotoxic cell death (Jeon et al. 2013; Wang and Liao 20103). ROCK

protein inhibition protects against ischemic damage both in vitro and in vivo (Yamashita et al.

2007; Gisselsson et al. 2010). Recent data from our research group shows that TRPM7 activation

impairs neuronal survival through hyper-activation of the actin depolymerizing protein, cofilin-1

(Bent 2011). Since ROCK proteins can regulate cofilin activity through LIM kinase, it is possible

that ROCK signalling is responsible for the cofilin mediated neuronal pathology observed during

TRPM7 activation.

Page 66: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

56  

In order to study the potential downstream mediators of TRPM7 in anoxia I first needed

to become proficient in primary neuronal culture techniques and reproduce cell death

experiments using prolonged OGD. I exposed primary cortical neurons to 2 hours of OGD in the

presence of MCN (to block excitotoxicity and isolate for TRPM7 currents), MCN Gd3+ (to

inhibit TRPM7-channels) and MCN Y-27632 (to inhibit ROCK2). Control experiments were

performed by incubating neurons for 2 hours in HBSS, HBSS MCN, HBSS MCN Gd3+ and

HBSS MCN Y-27632 (see Figure 2B, 2D, and 2B). Cytotoxicity, viability and apoptosis

(discussed in 3.9) were assessed 24 hours following treatment by using ApoTox-Glo Triplex

assay (see Figure 2A - D), a new assay to our lab, which allows for the evaluation of

cytotoxicity, viability and apoptosis in the same cell-based assay well.

Live cell protease activity is measured using GF-AFC, a membrane permeable peptide

substrate, which enters cells and is cleaved by the live-cell protease activity to generate a

fluorescent signal proportional to the number of living cells. These live-cell proteases become

inactive in cells that have lost their membrane integrity thereby allowing GF-AFC to label live

cells exclusively. Dead cell protease activity is measured using bis-AAF-R110 (membrane

impermeable substrate) which is cleaved by dead cell proteases released from cells that have lost

their membrane integrity. The fluorogenic products AFC and R110 have different excitation and

emission spectra (AFC = 400Ex/505Em; R110= 485Ex/520Em) thereby allowing viability and

cytotoxicity to be measured simultaneously. The plate reader used in this assays did not have a

filter to match the recommended excitation and emission values for the viability assay thus the

next closest filter was used for viability (360Ex/485Em); cytotoxicity was measured at the

recommended values (485Ex/520Em). The 2 hour OGD treatment resulted in a 79 ± 6 % increase

in cytotoxicity over HBSS control (see Figure 2A), and this difference between HBSS and

Page 67: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

57  

OGD was significant (p<0.05). This is consistent with previous reports showing enhanced

neuronal cell death following exposure to OGD (Goldberg and Choi, 1993, Aarts et al. 2003).

However, in the MCN treated neurons exposed to OGD, cell death values significantly fell back

to control levels (p<0.01). Equivalent significant decreases in cell death were also observed for

the MCN Gd3+ and the MCN Y-27632 treated neurons exposed to OGD (p<0.01 for both). The

cytotoxicity assay results were complemented by the viability assay (see Figure 2C), which

revealed a 30 ± 3 % decrease (p<0.01) in viability for the OGD treated groups over HBSS

control, whereas the OGD MCN and OGD MCN Gd3+ treated cultures exhibit viability values

equivalent to the HBSS controls (P<0.01 for both). Control experiments reveal that the 2 hour

incubation with HBSS, HBSS MCN, HBSS MCN Gd3+ and HBSS MCN Y-27632 had no

observable cytotoxic effects (see Figure 2B, 2D).

Surprisingly, the OGD MCN, OGD MCN Gd3+, and OGD MCN Y-27632 treated groups

exhibited equivalent levels of neuroprotection; no significant differences in cytotoxicity or

viability were observed between OGD MCN, OGD MCN Gd3+and OGD MCN Y-27632 treated

cultures suggesting that MCN treatment alone was sufficient at rescuing the OGD phenotype

(see Figure 2A, 2B). To confirm the validity of the ApoTox Triplex assay data, another

cytotoxicity assay was performed using Hoechst (which labels the nuclei of all cells) and

propidium iodide (PI; labels only dead cells) double staining. Hoechst and PI positive cells were

counted in HBSS Ctrl, OGD, OGD MCN, OGD MCN Gd3+, and OGD MCN Y-27632 treated

neuronal cultures. Exposure to OGD resulted in a 246 ± 86 % increase in cell death relative to

the HBSS control (p<0.001) as indicated by the increased number of PI positive neurons (see

Figure 3). OGD MCN, OGD MCN Gd3+, and OGD MCN Y-27632 treatments cause equivalent

reductions in cell death that were highly significantly different (p<0.001) in comparison to OGD

Page 68: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

58  

exposure alone. Similar to the ApoTox Triplex assay data, MCN alone appeared sufficient to

prevent OGD-mediated cell death. The cell death studies show that I was successful at growing

neurons that are susceptible to OGD and can be protected by MCN. However, these findings are

inconsistent with the Aarts et al (2003) which showed significant neuronal cell death during

prolonged anoxia (>1.5 hours) despite MCN treatment. Variability in the cell culture models (rat

vs mouse) as well as contamination by glia might account for the increased effectiveness of the

MCN treatment.

Figure 2. Cytotoxicity following oxygen glucose deprivation.

Embryonic cortical neurons were exposed to 2 hours of oxygen glucose deprivation (OGD) in

the presence of MCN, MCN Gd3+ and MCN Y-27632. Control treatments were exposed for 2

hours with HBSS, HBSS MCN, HBSS MCN Gd3+, and HBSS MCN Y-27632. Cytotoxicity and

viability were quantified according to the emission spectra of the cytotoxic product (R110) and

viability product (AFC) (n=3). (A) OGD treatment alone displays a significant 79 ± 6 %

increase in cytotoxicity relative to control, while OGD treatment in combination with MCN,

MCN Gd3+ or MCN Y-27632 displayed significantly lower cytotoxicity values in comparison to

OGD alone. (B) The HBSS MCN, HBSS MCN Gd3+, and HBSS MCN Y-7632 control

treatments had no apparent effect on neuronal cytotoxicity. (C) Viability significantly decreased

by 30 ± 3 % for the OGD treatment in comparison to HBSS control, whereas OGD MCN and

OGD MCN Gd3+ treatments showed significantly enhanced viability in comparison to OGD

alone. (D) The HBSS MCN, HBSS MCN Gd3+ and HBSS MCN Y-7632 control treatments had

no apparent effect on neuronal viability (n=3). All values are normalized to HBSS control, each

Page 69: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

59  

column represents mean ± SD. Results were analyzed via one-way ANOVA followed by

Dunnett’s multiple comparison test; * and ** indicate P<0.05 and P<0.01, respectively, in

comparison to control (HBSS Ctrl); # and # # indicate P<0.05 and P< 0.01, respectively, in

comparison to OGD treatment. MCN = MK801 (10µM), CNQX (25 µM) and Nimodipine (2

µM) (used to isolate for TRPM7 currents); Gd3+ = Gadolinium (10 µM); Y-27632 = ROCK

inhibitor (10µM).

HBSS Ctrl

OG

D

OG

D MCN

OG

D MCN G

d3+

OGD MCN Y

-276

320

50

100

150

200

250

% C

yto

tox

icity

A

*

# ## #

# #

Page 70: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

60  

HBSS Ctrl

HBSS MCN C

trl

HBSS MCN G

d3+ C

trl

HBSS MCN Y

-276

32 C

trl0

50

100

150

% C

yto

tox

icity

B

HBSS Ctrl

OG

D

OGD M

CN

OG

D MCN G

d3+

OGD MCN Y

-276

320

50

100

150

% V

iab

ility

**

# # # #

C

Page 71: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

61  

Figure 3: Evaluation of OGD mediated cytotoxicity using Hoechst and

propidium iodide staining.

Embryonic cortical neurons were exposed to 2 hours of oxygen glucose deprivation (OGD) in

the presence of MCN, MCN Gd3+ and MCN Y-27632. Control treatments were exposed for 2

hours with HBSS. Cytotoxicity was quantified by comparing the number of dead cells (PI

positive) to total cells (Hoechst positive) (A) Hoechst and PI double staining reveal a 246 ± 86 %

increase in cell death (p<0.001) for the OGD treated samples relative to the HBSS control (n=4).

(B) Epifluorescent images (20X) of the Hoechst and PI double labelling assay; PI = labels dead

HBSS Ctrl

HBSS MCN C

trl

HBSS MCN G

d3+ C

trl

HBSS MCN Y

-276

32 C

trl0

50

100

150

% V

iab

ility

D

Page 72: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

62  

cells only; Hoechst = labels all cell nuclei. All values are normalized to HBSS control, each

column represents mean ± SD. Results were analyzed via one-way ANOVA followed by

Dunnett’s multiple comparison test, *** indicate P<0.001 in comparison to control (HBSS Ctrl);

# # # indicates P<0.001, in comparison to OGD treatment. MCN = MK801 (10µM), CNQX (25

µM) and Nimodipine (2 µM) (used to isolate for TRPM7 currents); Gd3+ = Gadolinium (10 µM);

Y-27632 = ROCK inhibitor (10µM).

HBSS Ctrl

OG

D

OGD M

CN

OG

D MCN G

d3+

OGD MCN Y

-276

320

100

200

300

400

500

% C

yto

tox

icity

(PI/H

oe

ch

st)

Re

lativ

e to

HB

SS

Ctr

l ***

# # # # # # # # #

A

Page 73: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

63  

Page 74: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

64  

4.2 Necrosis is the dominant form of cell death following OGD exposure while

apoptosis is more prevalent following NMDAR and TRPM7 inhibition

To gain some insight into the post ischemic cell death mechanisms, 24 hours after

exposure to OGD neuronal cultures were probed for caspase activity using the Caspase-Glo3/7

reagent (of ApoTox Triplex Assay). The caspase-Glo® 3/7 reagent contains a luminogenic

DEVD-peptide substrate for caspase3/7 and a thermostable luciferase. Caspase3/7 cleavage of

DEVD-peptide substrate releases luciferin, a substrate for luciferase that generates light. The

light generated is quantified using a luminometer. The 2 hour exposure to OGD resulted in a 31

± 4 % decrease (p<0.05) in apoptosis in comparison to HBSS control group suggesting that non-

apoptotic mechanisms contribute to the OGD mediated neuronal cell death (see Figure 4A). The

OGD MCN, OGD MCN Gd3+ and OGD MCN Y-27632 treatment groups displayed a 28±8%,

35±8% and 32±9% increase in apoptosis in comparison to HBSS Ctrl, respectively. In

comparison to OGD treatment, OGD MCN, OGD MCN Gd3+ and OGD MCN Y-27632

displayed a 59 ± 24%, 66 ± 22% and 63 ± 25% increase respectively and the latter two increases

were significant in comparison to OGD treatment (P<0.05). Some neuronal apoptosis was

observed among all control groups, likely due to stress associated with the experimental

procedure (i.e. removal of cultures out of incubator, adding and removing treatment solution).

However, apoptosis values did not differ significantly between HBSS, HBSS MCN, HBSS MCN

Gd3+, and HBSS MCN Y-27632 control groups suggesting that MCN, Gd3+, and Y-27632

treatments do not affect cell death fate under control conditions (see Figure 4B). The decreased

apoptosis during OGD indicates that the majority of neuronal cell death reported by the

cytotoxicity assays is necrotic, consistent with previous models of ischemic cell death (Martin et

al. 1998). Interestingly, inhibition of both excitotoxicity (MCN) and TRPM7 (Gd3+) was

Page 75: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

65  

sufficient at changing neuronal cell death fate towards apoptosis. Similarly inhibition of

excitotoxicity and ROCK activity was equally effective at changing neuronal cell death fate

towards apoptosis suggesting that TRPM7 mediated and ROCK mediated signalling influences

neuronal cell death fate in OGD.

Figure 4. Apoptosis is not the predominant cell death mechanism observed in

neurons following OGD exposure.

Embryonic cortical neurons were exposed to OGD for 2 hours. Apoptosis was assayed 24 hours

post-OGD treatment using a caspase 3/7 dependent luciferase assay. (A) OGD treated cultures

display a 31 ± 4 % reduction in apoptosis relative to HBSS controls. On the other hand, OGD

MCN, OGD MCN Gd3+ and OGD MCN Y-27632 treated neurons display a 28±8%, 35±8% and

32±9% increase in apoptosis, respectively, in comparison to HBSS Ctrl, and 59 ± 24%, 66 ±

22% and 63 ± 25% increase in apoptosis respectively, relative to OGD. (B) HBSS, HBSS MCN,

HBSS MCN Gd3+, and HBSS MCN Y-27632 treated controls cultures display equivalent levels

of apoptosis. All values are normalized to HBSS control, each column represents mean ± SD.

HBSS control and OGD values were analyzed via paired T-test; * indicates P<0.05 relative to

HBSS Ctrl; whereas all other comparisons with OGD were performed via unpaired T-test; #

indicates P<0.05 relative to OGD. MCN = MK801 (10µM), CNQX (25 µM) and Nimodipine (2

µM) (used to isolate for TRPM7 currents); Gd3+ = Gadolinium (10 µM); Y-27632 = ROCK2

inhibitor (10µM).

Page 76: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

66  

HBSS Ctrl

OG

D

OGD M

CN

OG

D MCN G

d3+

OGD MCN Y

-276

320

50

100

150

200

% A

po

pto

sis

*

A

# #

HBSS Ctrl

HBSS MCN C

trl

HBSS MCN G

d3+ C

trl

HBSS MCN Y

-276

32 C

trl0

50

100

150

200

% A

po

pto

sis

B

Page 77: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

67  

4.3 The effects of oxygen glucose deprivation on ROCK 2 activity

Recent findings from our research group reveal a decrease in LIMK and cofilin

phosphorylation following a two hour exposure to OGD (Bent 2011). In addition, TRPM7

inhibition during OGD significantly enhanced LIMK and cofilin phosphorylation relative to

OGD alone (Bent 2011). Given that LIMKs are downstream targets of ROCKs, ROCK2 activity

was measured to investigate a potential involvement in the OGD and TRPM7 mediated effects of

LIMK previously reported by our research group. It has previously been suggested that trans-

autophosphorylation of the ROCK activation loop is essential for protein activity (Chen et al.

2002). Thus, to determine whether ROCK2 activation is involved in OGD or TRPM7 mediated

cell death, phosphorylation of ROCK2 levels within its activation loop was measured using an

anti-ROCK2 (phospho – T249) antibody. DIV12 primary cortical neurons were exposed to 2

hours of OGD with or without MCN, MCN Gd3+, and MCN Y-2732. Results reveal that ROCK2

phosphorylation was only statistically different from HBSS control in the presence of the ROCK

specific inhibitor Y-27632 (see Figure 5). Y-27632 treatment resulted in a 34 ± 3% decrease

ROCK2 phosphorylation (p<0.05) in comparison to HBSS control. Neither OGD MCN nor Gd3+

treatment had any observable effect on ROCK2 phosphorylation. Although previous studies

suggest ROCK family proteins play a role in ischemic pathology (Yamashita et al. 2007;

Gisselsson et al. 2010) the current data suggests that the activating phosphorylation at Threonine

249 is not involved in ROCK2 activity following in vitro exposure to anoxia.

Page 78: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

68  

Figure 5. OGD, MCN, and Gd3+ do not affect phosphorylation of ROCK within the

activation loop.

Proteins were extracted from DIV12 neurons following a 2 hour treatment with HBSS, HBSS

MCN, HBSS MCN Gd3+, OGD, OGD MCN, OGD MCN Gd3+, and OGD MCN Y27632. To

determine whether phosphorylation within the activation loop of ROCK2 changes following the

various treatments, cell extracts were separated via SDS-PAGE and probed for anti-ROCK2

(phospho-T249) antibody and mouse anti-rabbit-HRP secondary. Total ROCK2 was measured

using anti-ROCK2 antibody and mouse anti-rabbit-HRP secondary. Results show a significant

decrease (34± 3%) in ROCK2 phosphorylation only for the OGD MCN Y-27632 treated

cultures. All values are normalized to HBSS control, each column represents mean ± SD. Results

were analyzed via one-way ANOVA followed by Dunnett’s multiple comparison test, as well as

via an unpaired T-test, ** signifies P<0.01 relative to HBSS control. HBSS= Hanks Buffered

Salt Solution; MCN = MK801 (10µM), CNQX (25 µM) and Nimodipine (2 µM) (used to isolate

for TRPM7 currents); Gd3+ = Gadolinium (10 µM); Y-27632 = ROCK inhibitor (10µM).

Page 79: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

69  

4.4 Oxygen Glucose Deprivation attenuates P-21 activated kinase 3 activity

Recent findings from our research group report a decrease in LIMK1 activity

(dephosphorylation) following OGD and this decrease correlates with enhanced cofilin activity

(dephosphorylation) (Bent 2011). Furthermore, inhibition of TRPM7 currents with Gd3+ or

suppression of TRPM7 expression, using siRNA specific for TRPM7, impaired the cofilin

hyperactivation observed in the OGD treatment (Bent 2011). Group I PAKs are known to

activate LIMK proteins via phosphorylation of critical threonine residues (Arias-Romero and

Chernoff 2008). In addition, Group I PAK protein activity is regulated by phosphorylation of

Page 80: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

70  

critical residues within and outside the kinase domain (Edwards and Gill, 1999). Thus, PAK3

phosphorylation at Ser-144, a site known to contribute to PAK3 kinase activation (Chong et al.

2001), was measured to determine whether PAK3 activation is altered following anoxia and

whether TRPM7 lies upstream of PAK3 regulation. TRPM7 activity was isolated during OGD

using MCN and subsequently blocked with Gd3+; changes in PAK3 activity were then

determined as a measure of PAK3 phosphorylation (see Figure 6). The HBSS MCN control

group displayed similar levels of PAK3 phosphorylation as HBSS control, indicating NMDAR,

AMPA/kainite receptor, and voltage-gated Ca2+ channels have no baseline effect on PAK3

activity. Despite an apparent 18 ± 2% increase in PAK3 phosphorylation for the HBSS MCN

Gd3+ control group this difference was not significant relative to the other two controls further

suggesting that inhibition of TRPM7 channels has no baseline effect on PAK3 activity. A

significant reduction in PAK3 phosphorylation was observed for the OGD (38±5 %;p<0.001),

OGD MCN (26±3 %;p<0.05), and OGD MCN Gd3+ (25± 0.5% ;p<0.05) cultures compared to

control. Given that PAK3 activity is regulated by phosphorylation; these results indicate that

PAK3 activity is reduced following oxygen glucose deprivation and may therefore lie upstream

of reduced LIMK activity in anoxia. Furthermore, this reduction in PAK3 activity during OGD,

in comparison to HBSS control, appears to be independent of glutamate excitotoxicity and

TRPM7 inhibition as neither MCN nor MCN Gd3+ treatment were able to rescue the decrease in

PAK3 activity during OGD.

Page 81: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

71  

Figure 6. OGD attenuates PAK3 phosphorylation

Proteins were extracted from DIV12 neurons following a 2 hour treatment with HBSS, HBSS

MCN, HBSS MCN Gd3+, OGD, OGD MCN and OGD MCN Gd3+. To determine the effects of

OGD and TRPM7 activity on PAK3 phosphorylation cell extracts were separated via SDS-

PAGE and probed for anti-phospho PAK3 antibody and mouse anti-rabbit-HRP secondary. Total

PAK3 was measured using anti-PAK3 antibody and mouse anti-rabbit-HRP secondary. All

values are normalized to HBSS control, each column represents mean ± SD. Results were

analyzed via one-way ANOVA followed by Dunnett’s multiple comparison test, * and ***

represent P < 0.05 and P< 0.001,respectively, in comparison to HBSS control. HBSS= Hanks

Buffered Salt Solution; MCN = MK801 (10µM), CNQX (25 µM) and Nimodipine (2 µM) (used

to isolate for TRPM7 currents); Gd3+ = Gadolinium (10 µM).

Page 82: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

72  

Page 83: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

73  

4.5 Modulation of SSH1 activity by TRPM7 following oxygen glucose

deprivation.

As previously mentioned LIMK activation is mediated via phosphorylation by both Rho-

activated kinases and p-21 activated kinases; whereas dephosphorylation of LIMK via

phosphatases results in inhibition of kinase activity. Slingshot phosphatase 1 is known to

dephosphorylate both LIMK and cofilin (Ohta et al. 2003, Soosairajah et al. 2005).

Phosphorylation of C-terminal residues Ser-937 and Ser-978 of SSH1 promotes 14-3-3 binding

which inhibits SSH1 phosphatase activity (Nagata-Ohashi et al. 2004). To investigate whether

SSH1 plays a role in the LIMK and cofilin dephosphorylation following OGD, cortical neurons

were exposed to 2 hours of OGD. The role of TRPM7 in regulating SSH1 activity was also

investigated by isolating for TRPM7 currents through MCN, and then inhibiting those currents

with Gd3+. The HBSS, HBSS MCN, and HBSS MCN Gd3+ controls did not show statistically

significant differences in SSH1 phosphorylation (see Figure 7). A small but statistically

significant decrease in SSH1 phosphorylation (13± .7%), was observed for the OGD treatment

group in comparison to HBSS control (paired T-test; p= 0.02). OGD MCN treatment caused a

25± 9% increase in SSH1 phosphorylation relative to OGD; however, this difference was not

statistically significant. A 76 ± 20% increase in SSH1 phosphorylation was observed for the

OGD MCN Gd3+ cultures, relative to OGD alone; this result was statistically significant (p <

0.01). Overall these results suggest that TRPM7 may be involved in SSH1 regulation during

oxygen glucose deprivation.

Page 84: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

74  

Figure 7. TRPM7 modulates SSH1 activity

Proteins were extracted from DIV12 neurons following a 2 hour treatment with HBSS, HBSS

MCN, HBSS MCN Gd3+, OGD, OGD MCN and OGD MCN Gd3+. To determine whether SSH1

activity is modulated by OGD and TRPM7, cell extracts were separated via SDS-PAGE and

probed for anti-phospho S978 SSH1 antibody and mouse anti-rabbit-HRP secondary. Total

SSH1 was measured using anti-SSH1 antibody and mouse anti-rabbit-HRP secondary. All values

are normalized to HBSS control, each column represents mean ± SD. Results were analyzed via

one-way ANOVA followed by Dunnett’s multiple comparison test, as well as paired T-test. *

represents P < 0.05 relative to HBSS control, and ## represents P<0.01 relative to OGD. HBSS=

Hanks Buffered Salt Solution; MCN = MK801 (10µM), CNQX (25 µM) and Nimodipine (2 µM)

(used to isolate for TRPM7 currents); Gd3+ = Gadolinium (10 µM).

Page 85: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

75  

Page 86: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

76  

4.6 Immunofluorescent analysis of intracellular protein localization

Work from our research group has focused on identifying the proteins which interact with

TRPM7, and proteins that form a signalling relationship with TRPM7 in mediating neuronal cell

death. This work led to the identification of LIMK1 and cofilin-1 signalling transduction partners

downstream of TRPM7 in OGD. Although a direct interaction between TRPM7 and LIMK1 or

cofilin has not been shown yet, biochemical analysis of the signalling pathways behind TRPM7

activation during OGD and its consequent inhibition following Gd3+ treatment has implicated

TRPM7 in mediating cofilin hyperactivation (dephosphorylation) during OGD (Bent, 2011). To

determine whether the observed changes in PAK3 and SSH1 phosphorylation influenced protein

localization, immunofluorescent analysis of cortical neurons exposed to 2 hours of HBSS, OGD

and OGD MCN Gd3+ was performed. Successful immunostaining was achieved for LIMK1, P-

SSH1 and P-PAK3 (see Figures 8 and 9). LIMK1 labelling was largely concentrated in the

cytoplasm of the cell with strong staining along the perinuclear area, and no staining in the

nucleus and along the cell processes (see Figure 8 and Figure 9). P-PAK3 shows strong

fluorescence in the cell soma particularly around the perinuclear area; weak staining is observed

along cellular processes with no staining in the nucleus (see Figure 8). P-SSH1 labelling reveals

similar results as P-PAK3, with strong staining in the cell body and along the perinuclear area,

weak labelling in the cellular process, and no labelling in the cell nucleus (see Figure 9). Merged

images reveal colocalization of P-PAK3 and LIMK1 labelling (see Figure 8), similar

colocalization is observed for P-SSH1 and LIMK1 (see Figure 9). When comparing the

immunofluorescent staining patterns of LIMK1 no observable differences in cellular localization

can be seen between the three different treatment groups (HBSS, OGD, and OGD MCN Gd3+).

The same is true for P-PAK3 and P-SSH1 cellular localization. The data suggests that the

Page 87: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

77  

intracellular localization of LIMK1 and its upstream regulators P-PAK3 and P-SSH1 is not

affected by OGD treatment or OGD MCN Gd3+ treatment.

Figure 8. LIMK1 and P-PAK3 co-immunostaining in cortical neurons

Following a two hour exposure of DIV12 cortical neuronal cultures to HBSS, OGD, or OGD

MCN Gd3+, co-immunostaining was performed with mouse-anti-LIMK1 antibody (abcam) and

rabbit-anti-PAK3 (phospho S154) antibody (abcam) followed by goat-anti-mouse Alexa Fluor

555 secondary and goat-anti-rabbit Alexa Fluor 488 secondary, respectively. Epifluorescent

images of cells were taken with a conventional microscope with a 60X objective. Nuclei were

counterstained with DAPI (present in the Prolong Gold® Antifade mounting reagent).

Page 88: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

78  

Page 89: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

79  

Figure 9. LIMK1 and P-SSH1 co-immunostaining in cortical neurons

Following a two hour exposure of DIV12 cortical neuronal cultures to HBSS, OGD, or OGD

MCN Gd3+, co-immunostaining was performed with mouse-anti-LIMK1 antibody (abcam) and

rabbit-anti-SSH1 (phospho S978) antibody (ECM biosciences) followed by goat-anti-mouse

Alexa Fluor 555 secondary and goat-anti-rabbit Alexa Fluor 488 secondary, respectively.

Epifluorescent images of cells were taken with a conventional microscope with a 60X objective.

Nuclei were counterstained with DAPI (present in the Prolong Gold® Antifade mounting

reagent).

Page 90: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

80  

Page 91: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

81  

Chapter 5

5 Discussion

In this study primary neuronal cultures, isolated from E18 Sprague Dawley rats, were

used to investigate neuronal survival and apoptosis following exposure to oxygen glucose

deprivation. Inhibition of the excitotoxic cascade via MCN (which also isolates for TRPM7

currents); and TRPM7 currents with Gd3+ were used to evaluate the neuroprotective effects of

TRPM7 channels. Consistent with previous findings from our research group, a two hour

exposure to oxygen glucose deprivation significantly enhanced neuronal death. Surprisingly,

MCN treatment alone was sufficient at restoring neuronal survival to control levels. As a

consequence of the complete recovery of neuronal cell death by the OGD MCN treatment

conclusions regarding TRPM7’s contribution to neuronal pathology during OGD could not be

made potentially due to the MCN treatment masking any neuroprotective effects from TRPM7

inhibition in the OGD MCN Gd3+ treatment. Similarly, due to the complete rescue of neuronal

cell death by MCN during 2 hours of OGD, it is still unclear whether inhibition of ROCK

activity contributes to any additive neuroprotection through non-excitotoxic mechanisms.

Recent findings from our research group linking TRPM7 activation to LIMK and cofilin

dephosphorylation led to the investigation of three upstream regulators of LIMK kinase activity,

ROCK2, PAK3 and SSH1, the latter of which is also a direct regulator of cofilin activity (Amano

et al. 2001,Ohta et al. 2003). Western blot analysis suggests that activating phosphorylation of

Thr-249 is not involved in ROCK2 activity following in vitro ischemia (OGD). Given numerous

studies linking ROCK2 to ischemic neuropathology (Yamashita et al. 2007;Jeon et al. 2013;

Wang and Liao 2012;Gisselson et al. 2009) it is tempting to speculate that ROCK2

Page 92: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

82  

autophosphorylation is not essential for regulating ROCK2 activity during in vitro ischemia,

however, more work needs to be done before any conclusions can be made. PAK3

phosphorylation decreased following OGD and did not recover with either MCN or MCN Gd3+

treatment suggesting that glutamate and TRPM7 independent signalling mechanisms are

responsible for the suppression of PAK3 activity during OGD. A small decrease in SSH1

phosphorylation was observed during OGD treatment, and TRPM7 inhibition dramatically

enhanced SSH1 phosphorylation suggestive of a potential TRPM7 mediated regulation of SSH1

during ischemia.

To investigate whether the observed changes in SSH1 and PAK3 phosphorylation would

affect their subcellular localization and that of their downstream target LIMK1, E18 cortical

neurons exposed to two hours of HBSS, OGD or OBD MCN Gd3+ were co-stained with

antibodies against phospho-SSH1 and LIMK1 , and phospho-PAK3 and LIMK1.

Immunofluorescent analysis reveals strong labelling in the cell soma particularly in the

pericellular region for all three proteins LIMK1, P-PAK3 and P-SSH1. Surprisingly, differential

subcellular localization was not observed among the HBSS, OGD, and OGD MCN Gd3+ treated

groups for all three proteins.

5.1 Anti-excitotoxic treatment protects neuronal cultures against 2 hours of

oxygen glucose deprivation

The findings by Aarts and colleagues (2003) showing that TRPM7 channels may play a

more important role in ischemic cell death particularly during prolonged ischemia (>1.5 hours)

led to the evaluation of this model in the present study. OGD exposure led to significant

Page 93: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

83  

increases in neuronal cell death (see Figure 2, 3) consistent with previous reports (Goldberg and

Choi 1993; Aarts et al. (2003). In contrast to the findings by Aarts et al. (2003), MCN treatment

alone was sufficient at restoring neuronal survival to control levels based on both cytotoxicity

and viability values of the ApoTox Glo Triplex assay (see Figure 2A and 2C). PI and Hoechst

co-staining has been previously used to measure excitotoxic cell death and was used here (see

Figure 3) to validate the results of the newer ApoTox Glo Triplex assay. The results reveal that

TRPM7 inhibition with Gd3+ did not yield any additive neuroprotective effect over MCN alone;

however, in my experiments MCN restored cell survival to control levels with no additional

room for the Gd3+ effects. It is likely that TRPM7 channels were activated as both OGD

treatment and the absence of Mg2+in the extracellular solution would contribute to enhanced

TRPM7 channel activity (Kozak and Cahalan, 2003; Aarts et al. 2003). Consistent with previous

research TRPM7 inhibition was achieved with 10µM Gd3+, a concentration shown to inhibit the

TRPM7 current as well as promote neuronal survival (Aarts et al. 2003; Bent 2011). Evaluation

of neuronal viability with longer OGD exposure (i.e. 3 hours) may yield differences between

MCN and Gd3+ treatments however, this would not fit with our previous research evaluating

TRPM7 signal transduction at 2 hours of OGD injury. There is an inherent variability in primary

neuronal cultures which need to differentiate in vitro, consist of mixed population of cells, and

come from different animals with each set of experiments and thus, my cultures may have been

more readily rescued by MCN than previous studies.

In addition to cytotoxicity, viability was also measured with the ApoTox-Glo Triplex

assay (promega). Interestingly, the viability measurements did not show the same magnitude of

changes across as observed for cytotoxicity. The cytotoxicity assay values revealed a 79 ± 6 %

increase in neuronal cell death (see Figure 2A); however, the viability assay reported a 30 ± 3 %

Page 94: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

84  

decrease in neuronal survival (see Figure 2C) when an a difference of equivalent magnitude

between the cytotoxicity and viability assay values was expected. Due to the limitations of the

filters present in the Synergy-HT (Bio-Tek) microplate reader, viability was measured using an

excitation emission spectrum (360Ex/485Em) that was slightly different from the manufacturers’

(promega) recommended values (400Ex505Em). This use of a suboptimal excitation and emission

spectrum could explain the difference in magnitude between the cytotoxicity and viability values

of the ApoTox-Glo Triplex assay (promega). Despite the variations in cytotoxicity and viability

assay results, the overall data trends reveal that two hours of OGD severely impair neuronal

survival and that inhibition of the excitotoxic cascade (via MCN) is sufficient at rescuing

neuronal viability.

5.2 Different cell death mechanisms appear to dominate during OGD versus

TRPM7 inhibition.

The cell death mechanisms operating during ischemia are largely dependent on the

severity and duration the ischemic insult; consequently both necrotic and apoptotic phenotypes

have been observed in vitro as well as in vivo models (Martin et al. 1998, Lipton 1999; Unal-

Cevik et al. 2004). Apoptotic mechanisms are initiated following exposure to an adverse

stimulus such as ischemia (Lipton 1999); however, whether these mechanisms results in

apoptotic cell death is largely determined on the amount of available intracellular ATP (Leist et

al. 1997; Nicotera et al. 1999). Apoptosis is an energy dependent process (Leist et al. 1997;

Single et al. 2001), thus the dramatic depletion of ATP observed during a severe ischemic insult

would favor necrotic cell death. Cell death fate has profound implications for stroke

Page 95: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

85  

pathophysiology because unlike the removal of cells with minimal disruption of tissue

architecture that occurs in apoptosis, necrotic cell death is pro-inflammatory causing damage to

surrounding regions that result in scarring (Ren and Savill 1998), which can profoundly

influence brain structure and ultimately impair critical brain functions and cognitive processes.

To gain some insight into the cell death mechanisms operating in our in vitro model of

ischemia (OGD) caspase activity was measured to quantify apoptosis. The apoptosis assay data

reveals a significant decrease in apoptosis for OGD treated cultures (Figure 4 A, B) in

comparison to HBSS control. Given the enhanced neuronal cell death observed for OGD (Figure

2, 3), the apoptosis assay data are suggestive that the majority of ischemic neuronal cell death is

non-apoptotic. These results are consistent with findings from Almeida et al. (2002) showing

that 1 hour of OGD results in necrotic neuronal cell death. Surprisingly, of the enhanced

apoptosis observed for the OGD MCN and OGD MCN Gd3+ cultures, increases for only the

latter group were significant indicating that inhibition of TRPM7 activity may influence neuronal

cell death fate. If inhibition of TRPM7 activity promotes apoptotic rather than necrotic cell death

in vivo, it can help explain the decreased tissue damage and preservation of memory tasks

observed in TRPM7 suppressed rodent hemispheres following global ischemia (Sun et al. 2009).

Furthermore, it allows for the potential recovery of neurons through anti-apoptotic interventions.

However, conclusions regarding the effects of TRPM7 activity on neuronal fate cannot

be made based on the apoptosis assay results. Since the OGD MCN and OGD MCN Gd3+ treated

groups show similar increases in neuronal apoptosis (see Figure 4), we cannot distinguish the

AET mediated effects on cell death from those of TRPM7 inhibition. Variation within samples is

a possible contributor to the non-significant enhancement of apoptosis for the OGD MCN

cultures; inhibition of NMDA and AMPA/kainite channels has been previously reported to

Page 96: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

86  

expose apoptotic cell death in OGD treated neurons (Gwag et al. 1995). Furthermore, only

caspase 3 and 7 activity was recorded in the present study, providing a limited view of neuronal

apoptosis. Investigation of other pro-apoptotic factors (i.e. Bak, Bax) and nuclear markers (i.e.

DNA ladder pattern, TUNEL staining) would allow for a more comprehensive study of the AET

and TRPM7 mediated effects on apoptosis. In addition, the use of RNAi to knockdown TRPM7

expression would provide more direct evidence of the TRPM7 mediated effects in comparison to

the Gd3+ treatment.

5.3 Investigation of ROCK signalling in OGD mediated neuronal cell death

Recent reports from our research group show that during OGD, TRPM7 mediated

inhibition of LIMK and subsequent hyperactivation of cofilin result in the formation of cofilin-

actin rods (Bent 2011), aberrant structures associated with neurodegenerative disease (Maciver

and Harrington). Suppression of cofilin expression via siRNA led to a 20% reduction in neuronal

cell death in comparison to OGD, suggesting that the TRPM7 mediated cofilin hyper-activation

contributes neuronal cell death during OGD. Given that Rho associated kinases are known

regulators of LIMK activity (Amano et al. 2001; Ohashi et al. 2000) I decided to investigate

whether the TRPM7 mediated effects during OGD extend to the regulation of ROCK2. Based on

previous reports suggesting that trans-autophosphorylation within the activation loop of ROCK2

is essential for protein activity (Chen et al. 2002), phosphorylation of Thr-249 (located within

activation loop of ROCK2) was used to measure ROCK2 activity (Figure 5). Surprisingly, no

significant difference in ROCK2 phosphorylation was observed following OGD in comparison

to control and no differences in phosphorylation were observed across treatment groups with the

Page 97: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

87  

exception of the cultures treated with ROCK2 inhibitor (OGD MCN Y-27632 treatment), which

showed a significant decrease in phosphorylation. Although this data suggests that ROCK2

activity is not affected during in vitro ischemia (OGD), numerous studies linking ROCK to

ischemia pathology suggest otherwise (Yamashita et al. 2007;Jeon et al. 2013; Wang and Liao

2012;Gisselson et al. 2009). It is possible that ROCK2 signalling may operate during the initial

stages of OGD, which would cause us to miss this signalling event by the 2 hour time point.

Indeed, ROCK activity has been shown to significantly increase in the striatum at 5 min

following in vivo ischemia and then progressively fall to control levels at 2 hours of ischemia

(Yamashita et al. 2007). This enhanced ROCK activity during initial stages of ischemia is

supported by studies linking ROCK protein signalling to glutamate mediated excitotoxicity

which operates within the same time course (Jeon et al. 2002; Gisselson et al. 2009). For

instance, induction of glutamate excitotoxicity in H19-7/IGF-IR (immortalized hippocampal cell

line) results in enhanced ROCK activity as early as 2 minutes and was sustained to 30 minutes

consistent with ROCK signalling events operating during early stages of the excitotoxic cascade

(Jeon et al. 2002). Thus, measuring ROCK2 activity at different time points during OGD

exposure would provide a more complete picture of its temporal regulation.

An alternative possibility is that ROCK2 activity is not regulated by Thr-249

phosphorylation, or that phosphorylation within the activation loop of ROCK2 is not necessary

for enzyme activity. Indeed analysis of the X-ray crystal structure of ROCK suggests the protein

kinase domain has a catalytically competent conformation in the absence of phosphorylation

(Jacobs et al. 2006). Thus, it still remains to be determined whether TRPM7 regulates ROCK

function during OGD, and whether the potential changes in ROCK activity contribute to the

LIMK and cofilin mediated effects observed by our research group.

Page 98: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

88  

Given that upregulation of ROCK2 activity during ischemia onset contributes to neuronal

cell death (Yamashita et al. 2007), I wanted to determine whether disruption of ROCK signalling

in the presence of TRPM7 activity (OGC MCN Y-27632 treatment) would contribute to

enhanced neuroprotection. This way, the presence of additive neuroprotective effects over OGD

MCN treatment would link TRPM7 activity to ROCK. Previous in vitro and in vivo studies have

reported that inhibition of ROCK activity via fasudil or Y-277632 confers similar

neuroprotective effects as does NMDA channel inhibition (Yamashita et al. 2007; Gisselson et

al. 2009); however, the neuroprotective effects of ROCK inhibition in conjunction with anti-

excitotoxic therapy (MCN treatment) have not yet been investigated.

The cytotoxicity and viability assay results reveal no differences in neuroprotection

between OGD MCN, OGD MCN Gd3+ and OGD MCN Y-27632 treated groups (Figure 2,3). As

previously stated the observation that MCN alone completely recovers neuronal viability,

precludes any potential additive neuroprotective effects from being observed. Exposure of

neuronal cultures to 3 hours of OGD might expose differences in neuronal viability between

OGD MCN, OGD MCN Gd3+ and OGD MCN Y-27632 treatment groups. However, the

possibility that ROCK inhibition in the presence of MCN treatment does not result in additional

neuroprotection cannot be excluded as the cytotoxic effects of ROCK2 have been described to

operate downstream of NMDA channel activation (Jeon et al. 2002; Gisselson et al. 2009). Thus,

whether TRPM7 activity contributes to ROCK mediated cytotoxicity during OGD still remains

to be determined.

Page 99: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

89  

5.4 Ischemic depression of PAK3 activity

Like ROCKs, P-21 activated kinases are another family of Ser/Thr protein kinases known

to phosphorylate LIMK (Edwards et al. 1999) and this LIMK phosphorylation is essential for its

kinase activity (Edwards and Gill 1999). Rac and Cdc42 are two members of the Rho family of

GTPases known to regulate PAK activity (Arias-Romero and Chernoff 2008) and recent studies

have implicated both Rac and Cdc42 in ischemic neuronal cell death (Zhao et al. 2007;

Lazarovici et al. 2012; Gutierrez-Vargas et al. 2010). For instance, a decrease in Rac activity has

been observed both in vivo and in vitro following a 2 hour exposure to ischemia (Lazarovici et

al. 2012). This depression in Rac activity has been shown to persist for up to 1 month post

ischemia (Gutierrez-Vargas et al. 2010). In contrast, Cdc42 activity is enhanced following

ischemia/reperfusion, and this Cdc42 upregulation is shown to contribute to delayed neuronal

cell death (Zhao et al. 2007).

Despite the recent interest in the role of Rho GTPases in ischemic pathology, there are

virtually no studies investigating the involvement of PAK proteins in ischemia. Given the

decrease in LIMK and cofilin phosphorylation following OGD observed by our research group,

an investigation into the role of PAK proteins was necessary. Evaluation of P-PAK3 cellular

organization during OGD and TRPM7 inhibition, reveal no differences in P- PAK3 localization

between treatments (Figure 8), suggestive that OGD treatment does not influence PAK3

localization. It is possible that P-PAK3 may exert its signalling effects in the cell soma. PAK3

phosphorylation levels were also measured to evaluate PAK3 activity in response OGD, AET

and TRPM7 inhibition. Results (Figure 6) reveal a significant reduction in PAK3 activity during

in vitro ischemia (OGD). However, these levels did not show significant recovery with either

anti-excitotoxic therapy (AET) or TRPM7 inhibition suggesting that neither the excitotoxic

Page 100: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

90  

cascade nor TRPM7 activity contribute to the reduction in PAK3 activity during in vitro

ischemia. It is possible that the previously reported depression in Rac activity following ischemia

(Gutierrez-Vargas et al. 2010; Lazarovici et al. 2012) contributes to the observed decrease in

PAK3 activity. However, Rac activity levels following OGD exposure need to be evaluated to

confirm a potential involvement in PAK3 regulation. Never the less, the observed decrease in

PAK3 activity during OGD is suggestive of a potential contribution to the reduced

phosphorylation of LIMK and cofilin previously reported by our research group, albeit through

Glutamate independent and TRPM7 independent mechanisms.

PAK1, PAK2 and PAK3 share ~ 90% sequence homology in their regulatory PBD-AID

domain and over 93% homology in their kinase domain (Arias-Romero and Chernoff, 2008),

hence it is of interest to investigate whether PAK1 and PAK2 exhibit similar regulation during

OGD as does PAK3. Although some functional overlap between Group I PAKs exists, they also

exhibit distinct roles, as in regulating cell death. For instance, PAK1 activity is associated with

neuronal survival whereas PAK3 activation is linked to neuronal cell death (Kreis et al. 2009);

thus suppression of PAK3 during OGD could represent an endogenous neuroprotective

mechanism.

5.5 A role for SSH1 downstream of TRPM7 mediated activation in oxygen

glucose deprivation

In addition to evaluating the effects of known up-regulators of LIMK activity (i.e. ROCK

and PAK) during OGD and TRPM7 inhibition, attention was diverted to potential down-

regulators of LIMK activity. Slingshot phosphatase 1 (SSH1) has been described as a cofilin

Page 101: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

91  

phosphatase, which removes the inhibitory phosphate group on Ser-3 and activates cofilin

(Kurita et al. 2008 ). In addition to cofilin dephosphorylation, SSH1 inhibits LIMK via

dephosphorylation of Thr-508, a critical phosphorylation site essential for LIMK activity

(Soosairajah et al 2005). Given that the phosphatase activity of SSH1 is restricted to LIMK and

cofilin, it is an ideal candidate for mediating the changes in LIMK and cofilin activity reported

by our research group. Thus, SSH1 activity was determined by the level of Ser-978

phosphorylation which suppresses the phosphatase activity of SSH1 (Nagata-Ohashi et al. 2004).

14-3-3 binding to phosphorylated C-terminal serine residues (Ser-937, Ser-978) performs this

SSH1 inhibition by interfering with the ability of SSH1 to bind to F-actin, which in turn prevents

the F-actin mediated activation of SSH1 (Nagata-Ohashi et al. 2004).

The enhanced SSH1 activity observed during OGD exposure (see Figure 7) is consistent

with previously reported reductions in phosphorylation of LIMK1 and cofilin during OGD (Bent

2011). In addition, inhibition of TRPM7 activity during OGD resulted in dramatic decrease in

SSH1 activity, which is in support with previous reports from our research group showing

enhanced LIMK and cofilin phosphorylation in the TRPM7 inhibited cultures exposed to OGD.

To determine whether these changes in SSH1 activity influenced its cellular organization

immunofluorescent images SSH1 were taken during OGD and TRPM7 inhibition. The

immunofluorescent analysis (Figure 9) revealed that the inactive pool of SSH1 (P-SSH1)

remained perinuclear and did not change between treatments suggesting that the ischemia does

not affect the localization of inactive SSH1. To date no studies have been performed to evaluate

the effects of anoxia/ischemia on SSH1 activity. The results of this study are the first to describe

changes in SSH1 activity in response to in vitro ischemia (OGD). Furthermore, these results are

the first to implicate TRPM7 channel signalling in the regulation of SSH1 activity. Thus, the

Page 102: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

92  

western analysis data support a potential TRPM7 mediated signalling relationship with SSH1

which can be added to our working model of cofilin regulation during OGD.

Numerous studies provide support for a potential SSH1 mediated effect on cofilin activity

during anoxia. For instance, exposure of HeLa cells to H2O2 results in cofilin activation through

SSH1 phosphatase; oxidation of 14-3-3ζ promotes its dissociation from SSH1 allowing the

dissociated SSH1 to become activated by F-actin binding and consequently activate

(dephosphorylate) cofilin (Kim et al. 2009). In addition, SSH1 has been shown to mediate Ca2+

induced cofilin dephosphorylation via calcineurin in 293T and HeLa cells (Wang et al. 2004).

The authors describe a pathway whereby Ca2+ influx results in calcineurin activation; the active

calcineurin then dephosphorylates SSH1, increasing its cofilin-phosphatase activity (Wang et al.

2004). Given that TRPM7 activation during OGD has been shown to contribute to calcium influx

and oxidative stress (Aarts et al. 2003) it is possible that a calcineurin-SSH1-cofilin signalling

pathway could operate downstream of TRPM7 activity. In addition, TRPM7 may also regulate

SSH1 activity directly through 14-3-3 since co-immunoprecipitation experiments from our

research group show that 14-3-3 pulls down along with TRPM7. Additional studies are

necessary to confirm the western analysis data of SSH1 including in vitro phosphatase assays of

SSH1 to evaluate its cofilin-phosphatase activity; SSH1 knockdown and expression studies to

confirm that changes in SSH1 activity are sufficient at influencing cofilin phosphorylation; and

Ca2+ chelation and anti-oxidant treatments to evaluate the mechanisms behind SSH1 modulation.

Page 103: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

93  

5.6 Model of TRPM7 mediated cell death.

The present study provides a new potential link into the TRPM7 mediated cofilin

hyperactivation recently described by our research group. The model outlined in Figure 10

reflects what may occur when a cell is under stress through OGD. Initially TRPM7 channels are

activated by the ensuing ATP depletion (Katsura et al. 1994), oxidative stress and Ca2+influx

from upstream channel activity (i.e. NMDA, AMPA) (Aarts et al. 2003; Arundine and

Tymianski 2003). TRPM7 channel activation results in further Ca2+ influx which contributes to

the generation of nitric oxide mediated ROS production (Aarts et al. 2003). The ensuing Ca2+

influx promotes further TRPM7 channel activity and ROS production through positive feedback

(Aarts et al. 2003). The Ca2+ overload can activate calcineurin which in turn activates SSH1

(Wang et al. 2005). The oxidative stress may further enhance SSH1 activity by promoting its

dissociation from 14-3-3ζ (Kim et al. 2009). The cofilin-phosphatase activity of active SSH1

would result in cofilin hyperactivation (dephosphorylation) and LIMK1 inhibition

(dephosphorylation). Decreased LIMK activity could also result from suppression of PAK3

activity, albeit through TRPM7 and NMDAR independent signalling events. Ultimately, cofilin

hyperactivation results in the formation of cofilin-actin rods (Bamrug 1999, Minamide et al.

2000, Bent 2011).

Given that neurons consume nearly 50% of their energy through actin turnover, the

generation of actin-cofilin rods is believed to be neuroprotective at first by turning off actin-

treadmilling (Bernstein et al. 2003).However, during prolonged exposure to neuronal stress, as is

the case with OGD, the actin-cofilin rods can grow long enough to span the diameter of neurites

which in turn disrupts cytoskeletal structure and intracellular transport ultimately leading to

degeneration of neurites beyond the rods (Minamide et al. 2000). By disrupting intracellular

Page 104: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

94  

transport and neuronal communication, the actin-cofilin rods ultimately result in neuronal death

(Maloney and Bamburg). Indeed, suppression of cofilin activity during OGD via siRNA

contributed to a 20% reduction in neuronal cell death, affirming a cofilin mediated effect in

ischemia pathology (Bent 2011).

Figure 10. Proposed model of TRPM7 mediated cofilin regulation during OGD

Page 105: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

95  

5.7 Future studies

Evaluation of ROCK2 activity downstream of TRPM7 and its contribution to stroke

pathology was inconclusive. Additional studies are necessary to evaluate ROCK2 activity

downstream of NMDA signalling and TRPM7 signalling. Purification of ROCK2 from cell

lysates and its subsequent use in vitro kinase assays would provide a more direct evaluation of

protein activity. Measuring phosphorylation levels of known ROCK2 substrates (i.e. LIMK,

myosin light chain phosphatase) as well as the activity of ROCK2 upstream regulators (i.e.

RhoA) would provide additional insight into ROCK2 activity levels during the various treatment

employed in the present study. Furthermore, stimulation as well as inhibition of ROCK2 activity

could be useful at evaluating the ROCK2 mediated signalling events and its contribution to

stroke pathology.

Although the present results are in agreement with previous reports linking SSH1 activity

to ischemic pathology, the western analysis data is only correlative and additional studies are

necessary to confirm that SSH1 activity is indeed influenced by TRPM7. Initially, SSH1 activity

should be investigated more thoroughly via in vitro phosphatase assays, as well as through

measuring the phosphorylation levels of LIMK and cofilin. The use of a constitutively active

form of SSH1 and phosphatase dead mutants in the various treatments used in this study would

further reaffirm the capacity of SSH1 to regulate LIMK and cofilin activity. In addition,

knockdown of SSH1 expression would unveil other potential regulators of cofilin activity such

as chronophin phosphatase. Chronophin is known to enhance cofilin activity via

dephosphorylation (Mizuno et al. 2013). The potential effects of chronophin on cofilin activity

during OGD cannot be excluded from the present study. Thus, future studies should elucidate

Page 106: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

96  

the effects of TRPM7 channels on chronophin and evaluate choronophin’s ability to influence

cofilin during OGD.

In addition, the recent observation from our research group showing 14-3-3 to pull down

along with TRPM7 are suggestive of a potential link between TRPM7 and SSH1 through 14-3-3.

Future studies should be aimed at identifying the TRPM7 regions responsible for 14-3-3 binding

(i.e. via TRPM7 point mutations and Co-IP), and then use this knowledge to determine the

functional outcome of disrupting the 14-3-3 interaction with TRPM7. In addition, the affinity of

TRPM7 for 14-3-3 should be evaluated when the channel is active as well as when suppressed.

It is possible that, the recruitment of 14-3-3 along with its binding partners such as SSH1, to

TRPM7 channels could present a potential mechanism of SSH1 activation during OGD by

exposing 14-3-3 bound SSH1 to a microenvironment that favours its activation. The TRPM7

interaction with 14-3-3 extends beyond SSH1 signalling; 14-3-3 proteins have been described to

have anti-apoptotic functions (Steinacker et al. 2011). Thus, examination of 14-3-3 activity along

with TRPM7 could expose additional mechanisms of TRPM7 mediated cell death.

The TRPM7 mediated regulation of cofilin is only part of the story, since knockdown of

cofilin expression during OGD suppressed neuronal cell death by only 20%. Future studies

should focus on the identification of potential TRPM7 binding partners through the use of co-

immunoprecipitation. In addition to identifying potential downstream mechanisms through

which TRPM7 channels are involved, co-immunoprecipitation can also identify potential

upstream regulators of TRPM7 activity. Such knowledge could help the development of TRPM7

specific inhibitors as to date none have been developed. Conservation of the TRPM7 alpha-

kinase for millennia suggests that its functional role is important. Co-immunoprecipitation

experiments could be coupled with in vitro kinase assays to identify endogenous TRPM7

Page 107: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

97  

substrates. Indeed myosin IIA, annexin I, and eEF2 kinase have been shown to be

phosphorylated by TRPM7 alpha kinase activity (Clark et al. 2006; Dorvkov and Ryazanov,

2004; Perraud et al. 2011) however the functional significance of such events is still unclear.

Future studies should evaluate TRPM7 substrate relationships both under homeostatic conditions

as well as during pathological states such as ischemia in order to identify direct contributors of

ischemic pathology. Furthermore, the bi-modal nature of TRPM7 makes it difficult to distinguish

the channel mediated effects from those of the alpha kinase. The expression of kinase dead and

channel dead TRPM7 proteins could help isolate the channel mediated signalling events from

those of the alpha kinase.

Page 108: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

98  

Chapter 6

6. Summary

The primary aim of this project was to identify new molecular interactors that operate

downstream of TRPM7 during OGD. Through western analysis, TRPM7 activity was shown to

influence SSH1 activity during OGD. These changes in SSH1 activity correspond with

previously reported TRPM7 mediated changes in LIMK and cofilin activity. These results

suggest that SSH1 is regulated downstream of TRPM7 and could serve as a potential contributor

to the cofilin mediated neurodegeneration previously reported by our research group. However,

additional studies are necessary to evaluate SSH1 activity during OGD and whether it’s

sufficient at regulating LIMK and cofilin. TRPM7 and NMDA-AMPPA/Kainate signalling did

not appear to contribute to the observed reductions in PAK3 activity. Other studies suggest that

Rac may contribute to PAK3 depression; whether this is indeed the case for OGD still needs to

be determined. Unfortunately, cell death assays and western analyses were inconclusive at

linking ROCK2 mediated neurodegeneration downstream of TRPM7. The use of methods that

directly measure ROCK2 activity would provide more conclusive data. In an attempt to visualize

P-SSH1 and P-PAK3 localization during OGD or TRPM7 inhibition revealed that P-SSH1 and

P-PAK3 localization does not change between treatments. Lastly I was able to develop a cell

death model that resulted in neuronal cell death during OGD, and was completely rescued by

MCN treatment. The use of a 3 hour exposure period (instead of 2 hours) might have caused

differences between OGD MCN and OGD MCN Gd3+ and OGD MCN Y-37632 to emerge. It

will be interesting to see whether ROCK2 mediated neurodegeneration is implicated with

TRPM7 signalling and whether the reported changes in SSH1 influence cofilin activity during

Page 109: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

99  

OGD. Although, it has been a decade since TRPM7 channels have been implicated in neuronal

pathology, much is unknown about the mechanisms that operate downstream of TRPM7

channels during ischemia. The identification of direct TRPM7 interacting partners and the

development of TRPM7 specific pharmacological inhibitors are essential to enhancing the

progress of TRPM7 research.

Page 110: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

100  

Reference List:

Aarts M., Iihara K., Wei W. L., Xiong Z. G., Arundine M., Cerwinski W., MacDonald J. F., and

Tymianski M. (2003) A Key role for TRPM7 Channels in anoxic neuronal death. Cell 115, 863-

877.

Aarts M. and Tymianski M. (2005) TRPMs and neuronal cell death. Eur. J. Physiol. 451,243-

249.

Almeida A., Delgado-Esteban M., Bolanos J. P. and Medina J. M. (2002). Oxygen glucose

deprivation induces mitochondrial dysfunction and oxidative stress in neurons but not in

astrocytes in primary culture. Journal of Neurochemistry 81,207-217.

Amano M., Chihara K., Nakamura N., Kaneko T., Matsuura Y. and Kaibuchi K. (1999) The

COOH terminus of Rho-kinase negatively regulates Rho-kinase activity. J. Biol. Chem. 247,

32418-32424.

Amano M., Ito M., Kimura K., Fukata Y., Chihara K., Nakano T., Matsuura Y. and Kaibuchi K.

(1996) Phosphorylation and activation of myosin by rho-associated kinase (Rho-kinase).J. Biol.

Chem. 271, 20246-2-249.

Amano T., Kaji N., Ohashi K. and Mizuno K. (2002) Mitosis-specific activation of LIM motif-

containing protein kinase and roles of cofilin phosphorylation and dephosphorylation .J. Biol.

Chem. 277,22093-22102.

Page 111: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

101  

Amano T., Tanabe K., Eto T., Narumiya S. and Mizuno K. (2001) LIM-kinase 2 induces

formation of stress fibres, focal adhesions and membrane blebs, dependent on its activation by

Rho-associated kinase-catalyzed phosphorylation at threonine-505. Biochem. J. 354, 149-159.

Andrianantoandro E. and Pollard T. D. (2006) Mechanism of actin filament turnover by severing

and nucleation at different concentrations of ADF/cofilin. Molecular Cell 24, 13-23.

Arias-Romero L. E. and Chernoff J. (2008) A tale of two Paks. Biol. Cell 100,97-108.

Arundine M. and Tymianski M. (2003) Molecular mechanisms of calcium-dependent

neurodegeneration in excitotoxicity. Cell Calcium 34, 325-337.

Bamburg J. R. (1999) Proteins of the ADF/cofilin family: essential regulators of actin dynamics.

Annu. Rev. Cell Dev. Biol. 15,185-230.

Bargal R., Avidan N., Ben-Asher E., Olender Z., Zeigler M., Frumkin A., Raas-Rothschild A.,

Glusman G., Lancet D., Bach G.(2000) Identification of the gene causing mucolipidosis type IV.

Nat. Genet. 26,118-123.

Benner G. E., Dennis P. B. and Masaracchia R. A. (1995) Activation of an S6/H4 kinase

(PAK65) from human placenta by intramolecular and intermolecular autophosphorylation. J.

Biol. Chem. 270, 21121-21128.

Bent R. (2011) Transient Receptor Potential ‘Melastatin’ 7 Channels Regulate Neuronal

Cytoskeletal Dynamics.Unpublished Master’s Thesis, University of Toronto, Ontario, Canada

Bernstein B. W. and Bamburg J. R. (2003) Actin-ATP hydrolysis is a major energy drain for

neurons. J. Neurosci. 23, 1-6.

Page 112: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

102  

Bernstein B. W., Chen H., Boyle J. A. and Bamburg J. R. (2006) Formation of acitn-ADF/cofilin

rods transiently retards decline of mitochondrial potential and ATP in stressed neurons.

American Journal of physiology 291, C828-C839.

Bernstein B. W., Painter W. B., Chen H., Minamide L. S., Abe H. and Bamburg J. R. (2000)

Intracellular pH modulation of ADF/cofilin proteins. Cell Motility and Cytoskeleton 47,319-336.

Bernstein B. W., Shaw A. E., Minamide L. S., Pak C. W. and Bamburg J. R. (2012)

Incorporation of cofilin into rods depends on disulfide intermolecular bonds: implications for

actin regulation and neurodegenerative disease. J. Neurosci. 32, 6670-6681.

Bokoch G. M. (2003) Biology of the p21-activated kinases. Annu. Rev. Biochem. 72,743-781.

Bobkov A A., Muhlrad A., Pavlov D. A., Kokabi K., Yilmaz A. and Reisler E. (2005)

Cooperative effects of cofilin (ADF) on actin structure suggest allosteric mechanism of cofilin

function. J. Mol. Biol. 356,325-334.

Brauchi S., Krapivinsky G., Krapivinsky L., Clapham D. E. (2007) TRPM7 facilitates

cholinergic vesicle fusion with plasma membrane. PNAS 105, 8304-8308.

Carlier M. F., Laurent V., Santolini J., Melki R., Didry D., Xia G. X., Hong Y., Chua N. H. and

Pantaloni D. (1997) Actin depolymerisation factor (ADF/cofilin) enhances the rate of filament

turnover: implication in actin-based motility. J. Cell Biol. 136, 1307-1322.

Chen J. P., Luan Y., You C. X., Chen X. H., Luo R. C. and Li. R. (2010) TRPM7 regulates the

migration of human nasopharyngeal carcinoma cell by mediating Ca influx. Cell Calcium 47,

425-432.

Page 113: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

103  

Chen L. Y., Rex C. S., Casale M. S., Gall C. M. and Lynch G. (2007). Changes in synaptic

morphology accompany actin signalling during LTP. J. Neurosci. 27, 5363-5372.

Chen X., Tan I., Ng C. H., H C., Lim L and Leung T. (2002) Characterization of RhoA-binding

kinase ROKα function using region-specific antibodies. J. Biol. Chem. 277, 1260-12688.

Cheng Y. D., Al-Khoury L. and Zivin J. A. (2004) Neuroprotection for ischemic stroke: two

decades of success and failure. NeuroRx 1,36-45.

Choi D. W. (1985) Glutamate neurotoxicity in cortical cell culture is calcium dependent.

Neuroscience Letters 58, 293-297.

Choi D. W. (1987) Ionic dependence of glutamate neurotoxicity. J Neurosci. 7,369-375.

Choi D. W., Maulucci-Gedde M. and Kriegstein A. R. (1987) Glutamate neurotoxicity in cortical

cell culture. J. Neurosci. 7,357-368.

Chong C., Tan L., Lim L. and Manser E. (2001) The mechanism of PAK activation:

autophosphorylation events in both regulatory and kinase domains control activity. J. Biol.

Chem. 276,17347-17353.

Clapham D.E., Julius D., Montell C., Schultz G. (2005) International Union of Pharmacology.

XLIX. Nomenclature and Structure-Function Relationships of Transient Receptor Potential

Channels. Pharmacol. Rev. 57, 427-450.

Clark K., Langeslag M., van Leeuwen B., Ran L., Ryazanov A. G., Figdor C. G., Moolenaar W.

H., Jalink K. and van Leeuwen F. (2006). TRPM7, a novel regulator of actomyosin contractility

and cell adhesion. The EMBO Journal 25,290-231.

Page 114: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

104  

Cobos J., Borello U. and Rubenstein J. L. (2007) Dlx transcription factors promote migration

through repression of axon and dendrite growth. Neuron 54,873-888.

Coleman M. L., Sahai E. A., Yeo M., Bosch M., Dewar A. and Olson M. F. (2001) Membrane

blebbing during apoptosis results from caspase-mediated activation of ROCK1. Nat. Cell Biol.

3,339-345.

Duncan L. M., Deeds J., Hunter J., Shao J., Holmgren L. M., Woolf E. A., Tepper R. I., Shyjan

A. W. (1998) Down-Regulation of the Novel Gene Melastatin Correlates with Potential for

Melanoma Metastasis. Cancer Research 58, 1515-1520.

Edwards D. C. and Gill G. N. (1999) Structural features of LIM kinase that control effects on the

actin cytoskeleton. J. Biol. Chem. 247, 11352-11361.

Edwards D. C., Sanders L. C., Bokoch G. M. and Gill G. N. (1999) Activation of LIM-kinase by

Pak1 couples Rac/Cdc42 GTPase signalling to actin cytoskeletal dynamics. Nature Cell Biology

1, 253-259.

Eswaran J., Soundararajan M., Kumar R. and Knapp S. (2008) UnPAKing the class differences

among p21-activated kinases. Trends Biochem. Sci. 33,394-403.

Feng J., Ito M., Kureishi Y., Ichikawa K., Amano M., Isaka N., Okawa K., Iwamatsu A.,

Kaibuchi K., Hartshorne D. J. and Nakano T. (1999) Rho-associated kinase of chicken gizzard

smooth muscle. J. Biol. Chem. 274, 3744-3752.

Fonfria E., Murdock P. R., Cusdin F. S., Benham C. D., Kelsell R. E., McNulty S. (2006) Tissue

Distribution Profiles of the Human TRPM Cation Channel Family. Journal of Receptors and

Signal Transduction 26, 157-178.

Page 115: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

105  

Gisselsson L., Toresson H., Ruscher K., Wieloch T. (2009) Rho kinase inhibition protects CA1

cells in organotypic hippocampal slices during in vitro ischemia. Brain Research 1316, 92- 100.

Goldberg M. P. and Choi D. W. (1993) Combined oxygen glucose deprivation in cortical cell

culture: calcium-dependent and calcium –independent mechanisms of neuronal injury. J.

Neurosci. 13,3510-3524.

Grimm C., Kraft R., Sauerbruch S., Schultz G., Harteneck C. (2003) Molecular and Functional

Characterization of the Melastatin-related Cation Channel TRPM3. J. Biol. Chem. 278,21493-

21501.

Grimm C., Kraft R., Schultz G., Harteneck C.(2005) Activation of the Melastatin-Related Cation

Channel TRPM3 by D-erythro-Sphingosine. Mol Pharmacol. 67, 798-906.

Guinamard R., Salle L., Simard C. (2011) The Non-selective Monovalent Cationic Channels

TRPM4 and TRPM5. Advances in Experimental Medicine and Biology 704, 147-171.

Gurniak C. B., Perlas E. and Witke W. (2005) The actin depolymerizing factor n-cofilin is

essential for neural tube morphogenesis and neural crest cell migration. Developmental Biology

278, 231-241.

Gutierrez-Vargas J., Castro-Alvarez J. F., Velasquez-Carvajal D., Montanez-Velasquez M. N.,

Cespedes-Rubio A. and Cardona-Gomez G. P. (2010) Rac1 activity changes are associated with

neuronal pathology and spatial memory long-term recovery after global cerebral ischemia.

Neurochemistry International 57, 762-733.

Hara Y., Wakamori M., Ishii M., Maeno E., Nishida M., Yoshida T., Yamada H., Shimizu S.,

Mori E., Kudoh J., Shimizu N., Kurose H., Okada Y., Imoto K., and Mori Y. LTRPC2 Ca2+ -

Page 116: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

106  

permeable channel activated by changes in redox status confers susceptibility to cell death.

Molecular Cell 9, 163-173.

Hofmann C., Shepelev M. and Chernoff J. (2004) The genetics of Pak. J. Cell Science 117,4343-

4354.

Huang T. Y., Minamide L. S., Bamburg J. S., and Bokoch G. M. (2008) Chronophin mediates

and ATP-sensing mechanism for cofilin dephosphorylation and neuronal cofilin-actin rod

formation. Dev. Cell. 15, 691-703.

Ichetovkin I., Grant W. and Condeelis J. (2002) Cofilin produces newly polymerized actin

filaments that are preferred for dendritic nucleation by the Arp2/3 complex. Current Biology 12,

79-84.

Iizuka M., Kimura K., Wang S., Kato K., Amano M., Kaibuchi K. and Mizoguchi A. (2012)

Distinct distribution and localization of Rho-kinase in mouse epithelial, muscle and neural

tissues. Cell Struct. Funct. 37, 155-175.

Ikeda S., Cunningham L. A., Boggess D., Hawes N., Hobson C. D., Sundberg J. P., Naggert J.

K., Smith R. S. and Nishina P. M. (2003) Aberrant actin cytoskeleton leads to accelerated

proliferation of corneal epithelial cells in mice deficient for destrin (actin depolymerizing factor).

Hum. Mol. Genet. 12,1029-1037.

Jacobs M., Hayakawa K., Swenson L., Bellon S., Fleming M., Taslimi P and Doran J. (2006)

The structure of dimeric ROCK1 reveals the mechanism for ligand selectivity. J. Biol. Chem.

281, 260-268.

Page 117: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

107  

Jeon B. T., Jeong E. A., Park S.Y., Son H., Shin H. J., Lee D. H., Kim H. J., Kang S. S., Cho G.

J., Choi W. S. and Roh G. S. (2012) The Rho-kinase (ROCK) inhibitor Y-27632 protects against

excitotoxicity-induced neuronal death in vivo and in vitro. Neurotox Res. 23,238-248.

Jeon S., Kim S., Park J.B., Suh P. G., Kim Y. S., Bae C.D. and Park J. (2002) RhoA and Rho-

kinase dependent phosphorylation of moesin at Thr-558 in hippocampal neuronal cells by

glutamate. J. Biol. Chem. 277, 16576-16584.

Jiang J., Li M., Yue L. (2005) Potentiation of TRPM7 inward currents by protons. J. Gen.

Physiol. 126, 137-150.

Jin J., Desai B. N., Navarro B., Donovan A., Andrews N. C. and Clapham D. E. (2008) Deletion

of TRPM7 disrupts embryonic development and thymopoiesis without altering Mg2+

homeostasis. Science 322,756-760.

Kaji N., Ohashi K., Shuin M., Niwa R., Uemura T. and Mizuno K. (2003) Cell cycle-associated

changes in slingshot phosphatase activity and roles in cytokinesis in animal cells. J. Biol. Chem.

278, 33450-33455.

Kamt F., Zdanov S., Levine R. L., Pariser A., Zhang Y., Zhang B., Yu L. R., Veenstra T. D. And

Shacter E. (2009) Oxidant-induced apoptosis is mediated by oxidation of the actin-regulatory

protein cofilin. Nature Cell Biology 11,1241- 1258.

Kaneko S., Kawakami S., Hara Y., Wakamori M., Itoh E., Minami T., Takada Y., Kume T.,

Katsuki H., Mori Y., Akaike A. (2006) A Critical Role of TRPM2 in Neuronal Cell Death by

Hydrogen Peroxide. J. Pharmacol. Sci. 101, 66-76.

Page 118: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

108  

Katsura K. I., Kristian T. and Siesjo B. K. (1994) Energy metabolism, ion homeostasis, and cell

damage in the brain. Biochemical society transactions 22, 991-996.

Krapivinsky G., Mochida S., Krapivinsky L., Cibulsky S. M., Clapham D. (2006) The TRPM7

ion channel functions in cholinergic synaptic vesicles and affects transmitter release. Neuron 52,

485-495.

Kreis P. and Barnier J. V. (2009) PAK signalling in neuronal physiology. Cellular Signalling

21,384-393.

Kreis P., Rousseau V., Thevenot E., Combeau G. and Barnier J. V. (2008) The four mammalian

splice variants encoded by the p21-activated kinase 3 gene have different biological properties. J.

Neurochem. 108, 1184-1197.

Kreis P., Thevenot E., Rousseau V., Boda B., Muller D. and Barnier J. V. (2007) The p21-

activated kinase 3 implicated in mental retardation regulates spine morphogenesis through a

Cdc42-dependent pathway. J. Biol. Chem. 282, 21497-21506.

Kurita S., Watanabe Y., Gunji E., Ohashi K. and Mizuno K. (2008) Molecular dissection of the

mechanisms of substrate recognition and F-actin-mediated activation of cofilin-phosphatase

Slingshot-1. J. Biol. Chem. 283,32542-32552.

Kurnet-Keil C., Bisping F., Kruger J., Brinkmeier H. (2006) Tissue-specific expression of TRP

channel genes in the mouse and its variation in three different mouse strains. BMC Genomics

7,159.

Langeslag M., Clark K., Moolenaar W. H., van Leeuwen F. N. and Jalink K. (2007) Activation

of TRPM7 channels by phospholipase C-coupled receptor agonists. J. Biol. Chem. 282, 232-239.

Page 119: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

109  

Latorre R., Zaelzer C., Brauchi S. (2009) Structure-functional intimacies of transient receptor

potential channels. Quarterly Reviews of Biophysics 42, 201-246.

Lazarovici P., Cohen G., Arien-Zakay H., Chen J., Zhang C., Chopp M. and Jiang H. (2012)

Multimodal Neuroprotection induced by PACAP38 in oxygen-glucose deprivation and middle

cerebral artery occlusion stroke models. J. Mol. Neurosci. 48, 526-540.

Lee N., Chen J., Sun L., Wu S., Gray K. R., Rich A., Huang M., Lin J. H., Feder J. N., Janovitz

E. B., Levesque P. C., Blanar M. A. (2003) Expression and Characterization of Human Transient

Receptor Potential Melastatin 3 (hTRPM3). J. Biol. Chem. 278, 20890-20897.

Lei M., Lu W., Meng W., Parrini M.C., Eck M. J., Mayer B. J. and Harrison S. C. (2000)

Structure of PAK1 in an autoinhibited conformation reveals a multistage activation switch. Cell

102, 387-397.

Leist M., Single B., Naumann H., Fava E., Simon B., Kuhnle S. and Nicotera P. (1999) Nitric

Oxide Inhibits execution of apoptosis at two distinct ATP-dependent steps upstream and

downstream of mitochondrial cytochromes C release. Biochem. Biophys. Res. Commun. 258,

215-221.

Leung T., Chen X. Q., Manser E. and Lim L. (1996) The p160 RhoA-binding kinase ROK alpha

is a member of a kinase family and is involved in the reorganization of the cytoskeleton. Mol.

Cell Biol. 16, 5313-5327.

Li H. S., Xu X. Z. and Montell C. (1999) Activation of a TRPC3-dependent cation current

through the neurotrophin BDNF. Neuron 1, 261-273.

Page 120: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

110  

Li M., Yu Y., Yang J. (2011) Structural biology of TRP channels. Advances in Experimental

Medicine and Biology 704, 1-23.

Lintschinger B., Balzer-Geldsetzer M., Baskaran T., Graier W. F., Romanin C., Zhu M. X.,

Groschner K. (2000) Coassembly of Trp1 and Trp3 Proteins Generates Diacylglycerol- and

Ca2+- sensitive Cation Channels. Journal of Biological Chemistry 275, 27799-27805.

Lipton P. (1999) Ischemic cell death in brain neurons. Pharmacological reviews 79,1431-1568.

Liu Y. and Qin N. (2011) TRPM8 in health and disease: cold sensing and beyond. Advances in

Experimental Medicine and Biology 704, 185-208.

Maciver S. K. and Harrington C. R. (1995) Two actin binding proteins, actin depolymerizing

factor and cofilin, are associated with Hirano bodies. Neuroreport 6, 1985-1988.

Martin L. J., Al-Abdulla N. A., Brambrink A. M., Kirsch J. R., Sieber F. E. And Portera-Cailliau

C. (1997) Neurodegeneration in excitotoxicity, global cerebral ischemia, and target deprivation:

A perspective on the contributions of apoptosis and necrosis. Brain Research Bulletin 46, 281-

309.

McGough A., Pope Brian., Chiu W. and Weeds A. (1997) Cofilin changes the twist of F-actin:

implications for actin filament dynamics and cellular function. J. Cell Biol. 138, 771-781.

McKemy D D., Neuhausser W. M., Julius D. (2002) Identification of a cold receptor reveals a

general role for TRP channels in thermosensation. Nature 416, 52-58.

McPhie D. L., Coopersmith R., Hines-Peralta A., Chen Y., Ivins K. J., Manly S. P., Kozlowski

M. R., Neve K. A. and Neve R. L. (2003) DNA synthesis and neuronal apoptosis caused by

Page 121: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

111  

familial Alzheimer disease mutants of the amyloid precursor protein are mediated by the p21

activated kinase PAK3. J. Neurosci. 23,6914-6927.

Meng J., Meng Y., Hanna A., Janus C. and Jia Z. (2005) Abnormal long-lasting synaptic

plasticity and cognition in mice lacking the mental retardation gene Pak3. J. Neuroscience 25,

6641-6650.

Middelbeek J., Kuipers A. J., Henneman L., Visser D., Eidhof I., Horssen R. v., Wieringa B.,

Canisius S. V., Zwart W., Wessels L. F., Sweep F. C. G. J., Bult P., Span P. N., Leeuwen F. N.v.

and Jalink K. (2012) TRPM7 is required for breast tumor cell metastasis. Cancer Research 72,

4250-4261.

Mills J. C., Stone N. L., Erhardt J. and Pittman R. N. (1998) Apoptotic membrane blebbing is

regulated by myosin light chain phosphorylation. J. Cell Biol. 140,627-636.

Minamide L. S., Striegl A. M., Boyle J. A., Meberg P. J. and Bamburg J. R. (2000)

Neurodegenerative stimuli induce persistent ADF/cofilin-actin rods that disrupt distal neurite

function. Nature Cell Biology 2, 628-636.

Mizuno K. (2013) Signalling mechanisms and functional roles of cofilin phosphorylation and

dephosphorylation. Cellular Signalling 25,457-469.

Monastyrskaya K., Babiychuk E. B., Draeger (2009) The annexins: spatial and temporal

coordination of signalling events during cellular stress. Cell. Mol. Life Sci. 66,2623-2642.

Monteilh-Zoller M., Hermosura M. C., Nadler M. J. S., Scharenberg A. M., Penner R. and Fleig

A. (2002) TRPM7 provides an ion channel mechanism for cellular entry of trace metal ions. J.

Gen. Physiol. 121, 49-60.

Page 122: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

112  

Nadler M. J. S., Hermosura M. C., Inabe K., Perraud A. L., Zhu Q., Stokes A. J., Kurosald T.,

Klnet J. P., Penner R., Scharenberg A. M. and Fleig A. (2001) LTRPC7 is a Mg•ATP-regulated

divalent cation channel required for cell viability. Nature 411,590-595.

Nagata-Ohashi K., Ohta Y., Goto K., Chiba S., Mori R., Nishita M., Ohashi K., Kousaka K.,

Iwamatsu A., Niwa R., Uemura T. and Mizuno K. (2004). A pathway of neuregulin-induced

activation of cofilin-phosphatase Slingshot and cofilin in lamellipodia. J. Biol. Chem. 165, 465-

471.

Nakagawa O., Fujisawa K., Ishizaki T., Saito Y., Nakao K. and Narumiya S. (1996) ROCK-I and

ROCK-II, two isoforms of Rho-associated coiled-coil forming protein serinte/threonine kinase in

mice. FEBS letters 392,189-193.

Nakashima K., Sato N., Nakagaki T., Abe H., Ono S. and Obinata T. (2005) Two mouse cofilin

isoforms, muscle-type (MCF) and non-muscle type (NMCF), interact with F-actin with different

efficiencies. J. Biochem. 138, 519-526.

Nicotera P., Leist M., Single B. and Volbracht C. (1999) Execution of apoptosis: converging of

diverging pathways? Biol. Chem. 380, 1035-1040.

Nilius B., Prenen J., Janssens A., Voets T., Droogmans G. (2004) Decavanadate modulates

gating of TRPM4 cation channels. J. Physiol. 560, 753-765.

Nilius B., Prenen J., Janssens A., Owisianik G., Wang C., Zhu M. X., Voets T. (2005) The

selectivity filter of the cation channel TRPM4. J. Biol. Chem. 280, 22899-22906.

Page 123: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

113  

Nilius B., Prenen J., Tang J., Wang C., Owisianik G., Janssens A., Voets T., Zhu M. X. (2005)

Regulation of the Ca2+sensitivity of the nonselective cation channel TRPM4. J Biol. Chem. 280,

6423-6433.

Nishita M., Tomizawa C., Yamamoto M., Horita Y., Ohashi K. and Mizuno K. (2005) Spatial

and temporal regulation of cofilin activity by LIM kinase and Slingshot is critical for directional

cell migration. J. Biol. Chem. 171,349-359.

Oancea E., Vriens J., Brauchi S., Jun J., Splawski I., Clapham D E. (2009) TRPM1 Forms Ion

Channels Associated with Melanin Content in Melanocytes. Sci Signal 2, ra21.

Oancea E., Wicks N. L., (2011) TRPM1: New Trends for an Old TRP. Adv. Exp. Med. Biol. 704,

135-145.

Oberwinkler J., Lis A., Giehl K. M., Flockerzi V., Philipp S. E. (2005) Alternative Splicing

switches the divalent cation selectivity of TRPM3 channels. J. Biol. Chem. 280, 22540-22548.

Ohashi K., Nagata K., Maekawa M., Ishizaki T., Narumiya S. and Mizuno K. (2000) Rho-

associated kinase ROCK activates LIM-kinase 1 by phosphorylation at Threonine 508 within the

activation loop. J. Biol. Chem. 275, 3577-3582.

Ohta Y., Kousaka K., Nagata-Ohashi K., Ohashi K., Muramoto A., Shima Y., Niwa R., Uemura

T. and Mizuno K. (2003) Differential activities, subcellular distribution and tissue expression

patterns of three members of slingshot family phosphatases that dephosphorylate cofilin. Genes

to Cells 8, 811-824.

Ono S.(2007) Mechanism of depolymerisation and severing of actin filaments and its

significance in cytoskeletal dynamics. Int. Rev. Cytol. 258,1-82.

Page 124: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

114  

Perraud A. L., Fleig A., Dunn C. A., Bagley L. A., Launay P., Schmitz C., Stokes A J., Zhu Q.,

Bessman M. J., Penner R., Kinet J. P., Scharenberg A. M. (2001) ADP-ribose gating of the

calcium-permeable LTRPC2 channel revealed by Nudix motif homology. Nature 411, 595-599.

Perraud A. L., Takanishi C. L., Shen B., Kang S., Smith M. K., Schmitz C., Knowles H. M.,

Ferraris D., Li Weixing., Zhang J., Stoddard B. L., Scharenber A. M. (2005) Accumulation of

Free ADP-ribose from Mitochondria Mediates Oxidative Stress-Induced Gating of TRPM2

Cation Channels. J. Biol. Chem. 280, 6128-6148.

Perraud A. L., Zhao X., Rayazanov A. G., Schmitz C. (2011) The channel-kinase TRPM7

regulates phosphorylation of the translational factor eEF2 via eEF2-k. Cell Signal. 23, 586-593.

Peterburs P., Heering J. Link G., Pfizenmaier K., Olayioye M. A. and Hausser A. (2009) Protein

kinase D regulates cell migration by direct phosphorylation of the cofilin phosphatase slingshot 1

like. Cancer Research 69, 5634-5638.

Phelphs C B. and Gaudet R. (2007) The role of the N-terminus and transmembrane domain of

TRPM8 in channel localization and tetramerization. J. Biol. Chem. 282, 36474-36480.

Pollard T. and Borisy G. G. (2003) Cellular motility driven by assembly and disassembly of actin

filaments. Cell 112, 453-465.

Reeder M. K., Serebriiskii I. G., Golemis E. A. and Chernoff J. (2001) Analysis of small GTPase

signalling pathways using p21-activated kinase mutants that selectively couple to cdc42. J. Biol.

Chem. 276, 40606-40613.

Ren Y. and Savill J. (1998) Apoptosis: the importance of being eaten. Cell Death and

Differentiation 5, 563-568.

Page 125: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

115  

Riento K., Guasch R. M., Garg R., Jin B. and Ridley A. J. (2003) RhoE binds to ROCK1 and

inhibits downstream signalling. Mol. Cell Biol. 23,4219-4229.

Riento K. and Ridley A. J. (2003) Rocks: Multifunctional kinases in cell behaviour.

Nat.Rev.Mol.Cell.Biol 4, 446-456.

Rohacs T., Lopes C. M. B., Michailidis I., Logothetis D. E. (2005) PI(4,5)P2 regulates the

activation and desensitization of TRPM8 channels through the TRP domain. Nature

Neuroscience 8, 626-634.

Rosso S., Bollati F., Bisbal M., Peretti D., Sumi T., Nakamura T., Quiroga S., Ferreira A. and

Caceres A. (2004) LIMK1 regulates Golgi dynamics, traffic of Golgi-derived vesicles, and

process extension in primary cultured neurons. Mol. Biol. Cell 15,3433-3449.

Runnels L. W., Yue L. and Clapham D. E. (2001) TRP-PLIK, a bifunctional protein with kinase

and ion channel activities. Science 291, 1043-1047.

Runnels L. W., Yue L. and Clapham D. E. (2002) The TRPM7 channel is inactivated by PIP2

hydrolysis. Nature Cell Biology 4,329-336.

Ryazanov A G. (2002) Elongation factor-2 kinase and its newly discovered relatives. FEBS

Letters 514, 26-29.

Ryazanov A G., Ward M. D., Mendola C. E., Pavur K. S., Dorovkov M. V., Wiedmann M.,

Erdjument-Bromage H., Tempst P., Parmer T. G., Prostko C. R., Germino F. J. and Hait W. N.

(1997) Identification of a new class of protein kinases represented by eukaryotic elongation

factor-2 kinase. Proc. Natl. Acad. Sci. U.S.A. 94,4884-4889.

Page 126: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

116  

Schmitz C., Perraud A. L., Johnson C. O., Inabe K., Smith M. K., Penner R., Kurosaki T., Fleig

A., and Scharenberg A. M. (2003) Regulation of Vertebrate Cellular Mg2+ Homeostasis by

TRPM7. Cell 114. 191-200.

Sclingmann K. P., Weber S., Peters M., Nejsum L. N., Vitzthum H., Klingel K., Kratz M.,

Haddad E., Ristoff E., Dinour D., Syrrou M., Neilsen S., Sassen M., Waldegger S., Seyberth H

W., Konrad M. (2002) Hypomagnesemia and secondary hypocalcemia is caused by mutations in

TRPM6, a new member of the TRPM gene family. Nature Genetics 31, 166-170.

Shi J. and Wei L. (2007) Rho kinase in the regulation of cell death and survival. Arch. Immunol.

Ther. Exp. (Warsz) 55,61-75.

Soosairajah J., Maiti S., Wiggan O. Sarmiere P., Moussi N., Sarcevic B., Sampath R., Bamburg

J. R. and Bernard O. (2005). The EMBO Journal 24,473-486.

Su L. T., Agapito M. A., Li M., Simonson W. T. N., Huttenlocher A., Habas R., Yue L. and

Runnels L. W. (2006) TRPM7 regulates cell adhesion by controlling the calcium-dependent

protease calpain. Journal of Biological Chemistry 281,11260-11270.

Sumi T., Matsumoto K. and Nakamura T. (2001) Specific activation of LIM kinase 2 via

phosphorylation of threonine 505 by ROCK, a Rho-dependent protein kinase. J. Biol. Chem.

276,670-676.

Sun H. S., Jackson M. F., Martin L. J., Jansen K., Teves L., Cui H., Kiyonaka S., Mori Y., Jones

M., Forder J. P., Glode T. E., Orser B. A., MacDonald J. F. and Tymianski M. (2009)

Suppression of hippocampal TRPM7 protein prevents delayed neuronal death in brain ischemia.

Nature Neuroscience, 12, 1300-1307.

Page 127: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

117  

Takezawa R., Schmitz C., Demeuse P., Scharenber A. M., Penner R. and Fleig A. (2004)

Receptor-mediated regulation of the TRPM7 channel through its endogenous protein kinase

domain. PNAS 101, 6009-6014.

Togashi K., Hara Y., Tominaga T., Higashi T., Konishi Y., Mori Y., Tominaga M. (2006)

TRPM2 activation by cyclic ADP-ribose at body temperature is involved in insulin secretion.

EMBO J. 25, 1804-1815.

Tosello-Trampont A. C., Nakada-Tsukui K., Ravichandran K. S. (2003) Engulfment of apoptotic

cells is negatively regulated by Rho-mediated signalling. J. Biol. Chem. 278,49911-49919.

Toshima J., Toshima J. Y., Amano T., Yang N., Narumiya S. and Mizuno K. (2001) Cofilin

phosphorylation by protein kinase testicular protein kinase 1 and its role in integrin-mediated

actin reorganization and focal adhesion formation. Mol. Biol. Cell. 12, 1131-1145.

Tsuruda P. R., Julius D., and Minor Jr D. L. (2006) Coiled coils direct assembly of a cold-

activated TRP channel. Neuron 51, 201-212.

Unal-Cevik I., Kilinc M., Can A., Gursoy-Ozdemir Y. and Dalkara T. (2004) Apoptotic and

Necrotic mechanisms are concomitantly activated in the same cell after cerebral ischemia. Stroke

35, 2189-2194.

Van Troys M., Huyck L., Leyman S., Dhaese S., Vandekerkhove J. and Ampe C. (2008) Ins and

outs of ADF/cofilin activity and regulation. European Journal of Cell Biology 87, 649-667.

Vartiainen M. K., Mustonen T., Mattila P. K., Ojala P. J., Thesleff I., Partanen J. and

Lappalainen P. (2002) The three mouse actin-depolymerizing factor/cofilins evolved to fulfill

cell-type-specific requirements for actin dynamics. Molecular Biology of the Cell 13, 183-194.

Page 128: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

118  

Venkatachalam K. and Montell C. (2007) TRP Channels. Annu. Rev. Biochem. 76, 387-417.

Voets T., Nilius B., Hoefs S., Kemp A. W. C. M., Droogmans G., Bindels B. J. M., Hoenderop J.

G. J. (2004) TRPM6 Forms the Mg2+influx Channel involved in intestinal and renal Mg2+

absorption. J. Biol. Chem. 279,19-25.

Walder R. Y., Landau D., Meyer P., Shalev H., Tsolia M., Brochowitz Z., Boettger M. B., Beck

G. E., Engelhardt R. K., Carmi R., Sheffield V. C., (2002) Mutation of TRPM6 causes familial

hypomagnesemia with secondary hypocalcemia. Nat. Genet. 31,171-174.

Wang Q. M. and Liao J. K. (2012) ROCKs as immunomodulators of stroke. Expert Opin. Ther.

Targets 16, 1013 – 1025.

Wang Y., Shibasaki F. and Kensaku M. (2005) Calcium signal-induced cofilin

dephosphorylation is mediated by slingshot via calcineurin. J. Biol. Chem. 280,12683-12689.

Wei C., Wang X., Chen M., Ouyang K., S L. S., and Cheng H. (2009) Calcium Flickers Steer

Cell Migration. Nature 457,901-905.

Wriggers W., Tang J. X., Azuma T., Marks P. W. And Janmey P. A. (1998). Cofilin and gelsolin

segment-1: molecular dynamics stimulation and biochemical analysis predict a similar actin

binding mode. J. Mol. Biol. 282, 921-932.

Wu G., D’Agati V., Cai Y., Markowitz G., Park J. H., Reynolds D. M., Maeda Y., Le T. C., Hou

H. Jr. Kucherlapati R., Edelmann W. and Somlo S. (1998) Somatic inactivation of Pkd2 results

in polycystic kidney disease. Cell 93, 177-188.

Page 129: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

119  

Wu L. J., Sweet T. B., Clapham D. E. (2010) International Union of Basic and Clinical

Pharmacology. LXXVI. Current Progress in the Mammalian TRP Ion Channel Family.

Pharmacol. Rev. 62,381-404.

Xu S., Moebius F., Gill D. L., Montell C. (2001) Regulation of melastatin, a TRP-related protein,

through interaction with a cytoplasmic isoform. Proc. Natl. Acad. Sci. 98, 10692-10697.

Yamamoto M., Nagata-Ohashi K., Ohta Y., Ohashi K. and Mizuno K. (2006) Identification of

multiple actin-binding sites in cofilin-phosphatase Slingshot-1L. FEBS Letters 580,1789-1794.

Yamamoto S., Shimizu S., Kiyonaka S., Takahashi N., Wajima T., Hara Y., Negoro T., Hiroi T.,

Kiuchi Y., Okada T., Kaneko S., Lange I., Fleig A., Penner R., Nishi M., Takeshima H., Mori Y.

(2008) TRPM2-mediated Ca2+ influx induces chemokine production in monocytes that

aggravates inflammatory neutrophil infiltration. Nat. Med. 14,738-747.

Yamashita K., Kotani Y., Nakajima Y., Shimazawa M., Yoshimura S., Nakashima S., Iwamama

T. and Hara H. (2007) Fasudil, a Rho kinase (ROCK) inhibitor, protects against ischemic

neuronal damage in vitro and in vivo by acting directly on neurons. Brain Research 1154,215-

224.

Yasui Y., Amano M., Nagata K., Inagaki N., Nakamura H., Saya H., Kaibuchi K. and Inagaki M.

(1998). J. Cell Biol. 143, 1249-1258.

Yoneda A., Multhaupt H. A. B. and Couchman J. R. (2005) The Rho kinases I and II regulate

different aspects of myosin II activity. J. Cell Biol. 170,443-453.

Zhao L., Ma Q. L., Calon F., Harris-White M. E., Yang F., Lim G. P., Morihara T., Ubeda O. J.,

Ambegaokar S., Hansen J. E., Weisbart R. H., Teter B., Frautschy S. A. and Cole G. M. (2006)

Page 130: The Effects of Oxygen Glucose Deprivation and TRPM7 ... · The Effects of Oxygen Glucose Deprivation and TRPM7 Activity on Slingshot Phosphatase and P-21 Activated Kinase Activity

120  

The role of p-21 activated kinase in pathway defects in the cognitive deficits of Alzheimer’s

disease. Nature Neurosci. 9, 234-242.

Zhao J., Pei D. S., Zhang Q.G. and Zhang G. Y. (2007) Down-regulation of Cdc42 attenuates

neuronal apoptosis through inhibition of MLK3/JNK3 cascade during ischemic reperfusion in rat

hippocampus. Cell Signal. 19, 831-843.