the subcellular localisation, tissue expression, substrate ... · pkn protein kinase n pmsf phenyl...

257
The subcellular localisation, tissue expression, substrate specificity and binding partners of Stress-Activated Protein Kinase-3 By Naomi Wynne Court Bachelor of Science (Hons) This thesis is submitted for the degree of Doctor of Philosophy University of Western Australia School of Biomedical & Chemical Sciences Discipline of Biochemistry & Molecular Biology 2004

Upload: dangnhan

Post on 08-Mar-2019

223 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

The subcellular localisation, tissue expression,

substrate specificity and binding partners of

Stress-Activated Protein Kinase-3

By

Naomi Wynne Court

Bachelor of Science (Hons)

This thesis is submitted for the degree of Doctor of Philosophy

University of Western Australia

School of Biomedical & Chemical Sciences

Discipline of Biochemistry & Molecular Biology

2004

Page 2: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

DECLARATION

The research presented in this thesis is my own work, with the exception of staining of

E16.5 and E13.5 mice with the SAPK3 antibody (Figure 4.8 and Figure 4.9), which was

performed by Emerald Perlas. This work was carried out at the School of Biomedical

and Chemical Sciences in the Discipline of Biochemistry and Molecular Biology at the

University of Western Australia. This thesis has not been submitted for any other degree

Naomi Wynne Court

18-06-2004

i

Page 3: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

ACKNOWLEDGEMENTS There are a number of people who I would like to extend my sincere appreciation to. First and foremost, my supervisor Marie, the kinase queen, for all her patience, enthusiasm and wisdom. Thank you for always encouraging me and challenging me, for your advice, for listening to my presentations, proof-reading my work, and writing references. It has been a real privilege to learn from you and I greatly appreciate everything you have done for me. Secondly, to the Bogoyevitch lab members (past and present) for all the fun times – I will leave with many fond memories! I think I will be hard-pressed to find such a great group of people to work with in the future. Special thanks to Renae, Dom (aka “Twinny”), Bahareh, Cecilia, Ingrid and Marilyn who made the lab a happy place to be and who have been great friends in and out of the lab. I will miss all the cake! Thank you to Oonagh and Ivana, for helping me to make the Sab mutant GST-fusion proteins during their third-year projects. Special thanks to Oonagh for carrying on the SAPK3 project! I would also like to thank “non-Bogoyevitch lab” people for making the Biochemistry department such a friendly place to be. Particularly Roopwant Judge, Corrine Porter, Lauren Goldie, Eiling Tan, Sean Lim and Sandra Tanz. I would like to thank Peter Klinken for letting me perform the yeast-two hybrid part of my project in his lab at the Western Australian Institute for Medical Research. Special thanks to Evan Ingley, who helped me to perform the library screen, and for his patience with my endless questions. Thank you to Miranda Grounds for allowing me to steal people from her lab to help me with mouse work. Special thanks to Marilyn Davies for euthanasing mice for me and always being happy to help me out. Thank you to Paul Rigby, for his friendly help with the use of the confocal microscope. Thank you to Nadia Rosenthal and her lab members for making me feel so welcome when I was there to learn about mouse embryology lab techniques. Special thanks to Maria Paola, Esfir, and Emerald for helping me out with mouse work. Thank you to Simone Ross for breeding mice for me. I would like to thank the following people for supplying constructs, samples and antibodies during the course of my project: Patrick Humbert, for the Scribble antibody; Jim Whelan, for the HSP60 antibody; Pietro De Camilli, for the HA-OMP25 construct; Carolyn Wiltshire, for the Sab constructs (wild-type and KIM mutants); Ana Cuenda, for

ii

Page 4: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

the GST-α1-Syntrophin construct; Angel Nebreda, for the MalE-MKK6(DD) construct; and Cris dos Remedios for the human heart samples. Thank you to George Yeoh for the use of his camera to photograph the embryo sections. Thank you to Jenny, Greg, Jude and Eddy for your help with all things administrative and computers! I would like to sincerely thank the Heart Foundation and Australian Federation of University Women for providing me with scholarships during the course of my PhD. Finally (almost), I would like to thank the people outside of university who have kept me sane during my PhD. This includes all of my friends (you know who you are), and family: Dad, Kate, Sam, Harry, Bernie, Wez and particularly Mum, who has always been supportive of me in whatever I do. And last but certainly not least, Dave, for being the kind and warm-hearted person that you are – thank you for putting up with all my bad moods and helping me to see the light at the end of the tunnel. I love you!

iii

Page 5: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

PUBLICATIONS

Data presented in this thesis has been published or submitted for publication as follows:

Court NW, Ingley E, Klinken SP, and Bogoyevitch MA. Outer Membrane Protein 25 – a

mitochondrial anchor and inhibitor of Stress-Activated Protein Kinase-3. Submitted.

Court NW, Kuo I, Quigley O, and Bogoyevitch MA. (2004). Phosphorylation of the

mitochondrial protein Sab by stress-activated protein kinase 3. Biochem Biophys Res

Commun. 319, 130-137.

Court NW, dos Remedios CG, Cordell J, and Bogoyevitch MA. (2002). Cardiac

expression and subcellular localization of the p38 mitogen-activated protein kinase

member, stress-activated protein kinase-3 (SAPK3). J Mol Cell Cardiol. 34, 413-426.

Additional contributions during the candidature of this degree included:

Bogoyevitch MA and Court NW. (2004). Counting on mitogen-activated protein kinases - ERKs 3, 4, 5, 6, 7 and 8. Cell Signal. In Press.

Ng DC, Court NW, dos Remedios CG, Bogoyevitch MA. (2003). Activation of signal

transducer and activator of transcription (STAT) pathways in failing human hearts.

Cardiovasc Res. 57, 333-346.

Bogoyevitch MA, Ng DC, Court NW, Draper KA, Dhillon A, and Abas L. (2000). Intact

mitochondrial electron transport function is essential for signalling by hydrogen

peroxide in cardiac myocytes. J Mol Cell Cardiol. 32,1469-1480.

iv

Page 6: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

ABBREVIATIONS

APAAP Alkaline Phosphatase and Anti-Alkaline

Phosphatase

ASK1 Apoptosis signal-regulating kinase 1

ATP Adenosine triphosphate

BCIP 5-bromo-4-chloro-3-indolyl-phosphate

BSA Bovine serum albumin

DEPC Diethyl pyrocarbonate

Dlg Discs-large

DLK Dual leucine zipper-bearing kinase

DMSO Dimethyl sulfoxide

DNA Deoxyribonucleic acid

DTT Dithiothreitol

EDTA Ethylenediaminetetraacetic acid

EGF Epidermal growth factor

ERK Extracellular signal-regulated kinase

FCS Foetal calf serum

FGF Fibroblast growth factor

GDP Guanine diphosphate

GEF Guanine nucleotide exchange factor

GST Glutathione-S-transferase

GTP Guanine triphosphate

HA Hemagglutinin

v

Page 7: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

HEK Human embryonic kidney

HtrA2 High temperature requirement A 2

IPTG Isopropyl-beta-D-thio galactopyranoside

IL-1 Interleukin-1

JIP1 JNK interacting protein 1

JNK c-Jun N-terminal kinase

KIM Kinase interaction motif

LB Luria-Bertani

LIF Leukemia inhibitory factor

MalE Maltose binding protein

MDCK Madin-Darby canine kidney

MAPK Mitogen-activated protein kinase

MEF Myocyte enhancing factor

MEK Mitogen-activated protein kinase kinase/ERK

kinase

MEKK Mitogen-activated protein kinase kinase

kinase/ERK kinase kinase

MKK Mitogen-activated protein kinase kinase

MKK Mitogen-activated protein kinase kinase kinase

MLK Mixed-lineage kinase

MORG1 Mitogen-activated protein kinase organizer 1

MP1 MEK partner 1

MRK Mixed lineage kinase-related kinase

NBT Nitroblue tetrazolium chloride

vi

Page 8: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

NCBI National Center for Biotechnology Information

NCS Newborn calf serum

NES Nuclear export signal

NGF Nerve growth factor

OMP25 Outer membrane protein 25

PAK1 p21-activated kinase 1

PAPK Polyploidy-associated protein kinase

PBS Phosphate-buffered saline

PCR Polymerase chain reaction

PDZ PSD-95/Discs-Large/ZO-1

PEG Polyethylene glycol

PH Pleckstrin homology

PICK1 Protein that interacts with C-kinase 1

PKA Protein kinase A

PKC Protein kinase C

PKN Protein kinase N

PMSF Phenyl methyl sulfonyl fluoride

PSD-95 Post-synaptic density-95

PTB Phospho-tyrosine binding

RNA Ribonucleic acid

RT Room temperature

Sab SH3 domain-binding protein that preferentially

associates with Btk

SAPK Stress-activated protein kinase

SB Smithkline Beecham

SDS-PAGE Sodium dodecyl sulphate polyacrylamide gel

electrophoresis

vii

Page 9: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

SH2 Src homology 2

SH3 Src homology 3

Sos Son of sevenless

SAP90 Synapse-associated protein 90

TAB Transforming growth factor-β-activated protein

kinase 1-binding protein 1

TAK1 Transforming growth factor-β-activated kinase 1

TBST Tris-Buffered Saline with Tween

TNF Tumour necrosis factor

ZO-1 Zona occludens-1

viii

Page 10: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

TABLE OF CONTENTS

Declaration i

Acknowledgements ii

Publications iv

Abbreviations v

Table of contents ix

List of figures xv

List of tables xix

Abstract xx

Chapter 1: Introduction and Literature Review 1

1.1 Intracellular signalling and protein kinases 1

1.2 The Mitogen-Activated Protein Kinases: Extracellular Signal- 2

Regulated Kinases as the paradigm

1.3 Parallel MAPK signalling pathways 5

1.3.1 The JNK MAPK family 6

1.3.2 The p38 MAPK family 6

1.3.3 Regulation of interactions in the MAPK pathways 8

1.4 Stress-Activated Protein Kinase-3 10

1.4.1 Evolutionary conservation and structure of SAPK3 10

1.4.2 SAPK3 expression and activation 11

1.4.3 SAPK3 substrate specificity and potential roles in the cell 13

1.4.4 SAPK3 intracellular localisation 14

1.4.5 SAPK3 contains a unique C-terminal tail 14

1.5 Study aims 16

Chapter 2: Materials and Methods 18

2.1 Reagents and chemicals 18

2.2 Antibodies 19

2.3 Methods 19

ix

Page 11: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.1 Culture of cell lines 19

2.3.2 Isolation of rat neonatal cardiac myocytes 20

2.3.3 Preparation of coverslips for immunostaining 21

2.3.4 Cell transfection 22

2.3.4(a) Lipofectamine only transfection protocol 23

2.3.4(b) Lipofectamine and Lipofectamine Plus transfection

protocol 23

2.3.5 Cell treatments 24

2.3.6 Isolation of embryonic mice and embryonic mouse hearts 25

2.3.7 Processing of embryos for sectioning 26

2.3.8 Immunostaining 27

2.3.8(a) Immunostaining of cultured cells 27

2.3.8(b) Immunostaining of mouse embryo sections 28

2.3.8(c) Wholemount staining of mouse embryos 29

2.3.9 Haematoxylin and eosin staining 30

2.3.10 Scanning confocal microscopy 30

2.3.11 Preparation of cell extracts 31

2.3.12 Preparation of extracts from tissue samples 32

2.3.13 Protein estimation 32

2.3.13(a) Spectrophotometric estimation 32

2.3.13(b) Estimation using SDS-PAGE separated proteins 32

2.3.14 Immunoprecipitation for subsequent Western blotting 33

2.3.15 Immunoprecipitation for subsequent kinase assays 33

2.3.16 Activation of GST-SAPK3, GST-JNK2 and GST-ERK2 34

2.3.17 Kinase assays 34

2.3.18 SDS-PAGE 35

2.3.19 Western blotting 36

2.3.20 Stripping Western blots 37

2.3.21 Preparation of competent E. coli 38

2.3.22 Transformation of XL1-Blue and BL21 DE3 plyss E.coli

strains with plasmid DNA 39

x

Page 12: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.23 Preparation of plasmid DNA using the QIAGEN miniprep and

maxiprep kits 39

2.3.24 Isolation of RNA from embryonic, neonatal and adult mouse

hearts 40

2.3.25 Quantitation of RNA and DNA samples 40

2.3.26 Agarose gel electrophoresis 40

2.3.27 DNA sequencing 41

2.3.28 Expression and purification of recombinant proteins 42

2.3.29 Recombinant DNA techniques 43

2.3.30 Restriction enzyme digest 44

2.3.31 Polymerase chain reaction (PCR) 44

2.3.32 Reverse transcription PCR 45

2.3.33 DNA ligation 46

2.3.34 Site-directed mutagenesis 46

2.3.35 Yeast two-hybrid screening 47

2.3.36 Preparation of selective media for yeast two-hybrid screening 48

2.3.37 Large-scale yeast transformation 49

2.3.38 β-galactosidase assay 51

2.3.39 Preparation of glycerol stocks of yeast two-hybrid screening

positive colonies 51

2.3.40 Isolation of DNA from the S. cerevisiae strain L40 52

2.3.41 Preparation of electrocompetent KC8 cells 52

2.3.42 Bacterial electroporation 53

2.3.43 Isolation of plasmid DNA from KC8 bacterial cells 54

2.3.44 Small-scale yeast transformation 54

2.3.45 Screening of a cDNA library with the SAPK3 antibody 55

2.3.46 Initial preparation for the library screening procedure 55

2.3.47 Establishing the library titre 55

2.3.48 Screening procedure 56

xi

Page 13: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Chapter 3: Evaluation of the expression and localisation of SAPK3 in

cell culture models 58

3.1 Introduction 58

3.2 Results 61

3.2.1 A specific anti-SAPK3 monoclonal antibody suitable for

immunoblotting and immunolocalisation 61

3.2.2 SAPK3 shows a punctate localisation in HEK293 fibroblasts

and cardiac myocytes 62

3.2.3 Leptomycin B treatment confirms SAPK3 cytoplasmic

localisation 64

3.2.4 SAPK3 co-localises with mitochondrial markers 64

3.2.5 SAPK3 overexpression does not appear to alter the morphology

or survival of cardiac myocytes 65

3.2.6 SAPK3 phosphorylation and expression in response to agonist

treatment 66

3.3 Discussion 66

Chapter 4: Evaluation of the expression and activation of SAPK3 in the

intact heart 71

4.1 Introduction 71

4.2 Results 73

4.2.1 SAPK3 is expressed in the heart 73

4.2.2 SAPK3 expression levels do not change in human heart disease 75

4.2.3 SAPK3 is expressed throughout embryonic and postnatal

development of the heart 76

4.3 Discussion 79

Chapter 5: Investigation of the substrate specificity of SAPK3 83

5.1 Introduction 83

5.2 Results 86

5.2.1 The activation of SAPK3 in vitro 86

xii

Page 14: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

5.2.2 Investigating the phosphorylation of mitochondrial proteins

by SAPK3 87

5.2.3 Investigating the phosphorylation of the mitochondrial protein

Sab by SAPK3 89

5.2.4 Loss of the first Kinase Interaction Motif (KIM) of Sab prevents

its phosphorylation by SAPK3 90

5.2.5 Serine 321 in Sab is the major site of phosphorylation by SAPK3

and JNK2 92

5.2.6 Surrounding sequence composition and location dictate the

favouring of Ser321 in Sab 93

5.3 Discussion 95

Chapter 6: Identification of proteins that bind to SAPK3 by a PDZ

domain-mediated interaction 103

6.1 Introduction 103

6.2 Results 105

6.2.1 Screening a yeast two-hybrid library with full-length SAPK3 105

6.2.2 SAPK3 interacts with Scribble and Lin-7C by yeast two-hybrid

analysis 107

6.2.3 SAPK3 interacts with the mitochondrial outer membrane protein,

OMP25 110

6.2.4 OMP25 can act as an inhibitor of SAPK3 in vitro 113

6.3 Discussion 114

Chapter 7: General Discussion 120

7.1 General Discussion 120

7.1.1 Evaluating the expression and subcellular localisation of SAPK3

in cultured cells 120

7.1.2 SAPK3 expression in the intact heart and evaluation of its role

in cardiac development and disease 122

7.1.3 Determining the substrate specificity of SAPK3 123

xiii

Page 15: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

7.1.4 Searching for binding partners of SAPK3 127

7.2 Conclusions 128

Chapter 8: References 130

Appendices 157

xiv

Page 16: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Appendix II could not be included in this digital thesis for copyright reasons. Please refer to the print copy of the thesis, held in the University Library.

Page 17: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

LIST OF FIGURES

Figure 1.1 Protein kinases 1a

Figure 1.2 Schematic diagram of the organisation of the ERK MAPK

pathway 3a

Figure 1.3 The structure of inactive and active (phosphorylated) ERK2 4a

Figure 1.4 Parallel MAPK phosphorylation cascades 6a

Figure 1.5 The structure of activated SAPK3 complexed with ATP 10a

Figure 1.6 Potential signalling intermediates leading to the activation

of SAPK3 12a

Figure 1.7 A Class I PDZ domain complexed with a peptide substrate 15a

Figure 2.1 Insertion of full-length rat SAPK3 into pBTM116 for yeast

two-hybrid screening 43a

Figure 2.2 The design of primers to insert a fragment of OMP25 into

the pVP16 vector 43b

Figure 2.3 Insertion of amino acids 37-177 of rat OMP25 into pVP16 43c

Figure 3.1 SAPK3 can be detected in HEK293 and neonatal rat cardiac

myocyte cell lysates 62a

Figure 3.2 Endogenous SAPK3 shows a punctate cytoplasmic distribution

in HEK293 fibroblasts 63a

Figure 3.3 Endogenous SAPK3 shows a punctate cytoplasmic distribution

in neonatal rat cardiac myocytes 63b

Figure 3.4 SAPK3 has a punctate cytoplasmic localisation and this does not

alter with Leptomycin B treatment of neonatal rat cardiac

myocytes 64a

Figure 3.5 SAPK3 does not co-localise with Paxillin in cardiac myocytes 64b

Figure 3.6 SAPK3 co-localises with mitochondrial markers 65a

Figure 3.7 Overexpression of SAPK3 in cardiac myocytes does not appear

to induce morphological changes or cell death 65b

Figure 3.8 Overexpression of SAPK3 in cardiac myocytes does not appear

to induce morphological changes or cell death 65c

xv

Page 18: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Figure 3.9 SAPK3 expression levels do not change in response to agonist

treatment of cultured cardiac myocytes 66a

Figure 3.10 Agonist treatment stimulates phosphorylation of SAPK2a, but

not SAPK3, in cultured cardiac myocytes 66b

Figure 4.1 SAPK3 is expressed in the whole heart 73a

Figure 4.2 SAPK3 expression decreases in neonatal cardiac myocytes with

increased culture time 74a

Figure 4.3 SAPK3 is expressed in the hearts of various species 75a

Figure 4.4 SAPK3 shows a relatively restricted tissue distribution in the

mouse 75b

Figure 4.5 SAPK3 expression levels do not change in failing human hearts 76a

Figure 4.6 SAPK3 is constantly expressed during cardiovascular

development 77a

Figure 4.7 Haematoxylin and eosin staining of E16.5, E13.5 and E12.5 mice 78a

Figure 4.8 SAPK3 is expressed in various tissues of the E16.5 mouse 78b

Figure 4.9 SAPK3 is expressed in various tissues of E13.5 and E12.5 mice 78c

Figure 4.10 SAPK3 expression in E8.5 and E10.5 embryos using wholemount

staining 78d

Figure 5.1 Optimising a protocol for GST-SAPK3 activation 86a

Figure 5.2 GST-SAPK3 pre-incubated with MalE-MKK6(DD) shows

increased activity towards GST-ATF2 87a

Figure 5.3 Expression of overexpressed mitochondrial proteins in HEK293

fibroblasts 87b

Figure 5.4 Immunoprecipitation of mitochondrial proteins from HEK293

fibroblast lysates 88a

Figure 5.5 SAPK3 can phosphorylate mitochondrial proteins involved in

apoptosis 89a

Figure 5.6 Sab is a substrate of SAPK3 in vitro 90a

Figure 5.7 GST-JNK2 shows different substrate preferences to GST-SAPK3

in vitro 90b

Figure 5.8 GST-ERK2 does not phosphorylate Sab in vitro 90c

xvi

Page 19: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Figure 5.9 A C-terminal SAPK3 peptide inhibits the phosphorylation of

α1-Syntrophin but not Sab, by SAPK3 91a

Figure 5.10 Loss of the first Kinase Interaction Motif of Sab prevents its

phosphorylation by SAPK3 91b

Figure 5.11 A Ser321Ala mutant of Sab is not phosphorylated by SAPK3 93a

Figure 5.12 A Ser321Ala mutant of Sab is not phosphorylated by JNK2 93b

Figure 5.13 The phosphorylation of Sab by SAPK3 is dependent on the

surrounding sequence and the position of the site within Sab 94a

Figure 5.14 Analysis of sequence determinants for phosphorylation at the

+2 and +3 positions of Ser321 95a

Figure 6.1 pBTM116-SAPK3 is unable to activate transcription alone in

the yeast two-hybrid system 106a

Figure 6.2 Screening the yeast two-hybrid EML Clone.1 cDNA library

using full-length SAPK3 106b

Figure 6.3 The identification of Scribble and Lin7C as SAPK3-interacting

proteins by yeast two-hybrid screening 107b

Figure 6.4 Scribble and Lin7C clones isolated in yeast two-hybrid screening 107c

Figure 6.5 The expression of Scribble in adult mouse tissues 109a

Figure 6.6 Determining the localisation of SAPK3 and Scribble in MDCK

cells 109b

Figure 6.7 Schematic diagram of Outer Membrane Protein 25 (OMP25) 110a

Figure 6.8 Overexpression of HA-OMP25 in HEK293 fibroblasts 110b

Figure 6.9 Confirmation of OMP25 and SAPK3 co-localisation 111a

Figure 6.10 Interaction of SAPK3 with OMP25 detected by yeast two-hybrid

analysis 111b

Figure 6.11 SAPK3 and OMP25 are co-immunoprecipitated from cells 112a

Figure 6.12 Using HA-OMP25(NLS) to alter the subcellular localisation of

SAPK3 113a

Figure 6.13 The expression of GST-OMP25 in bacteria 113b

Figure 6.14 OMP25 is not a substrate of SAPK3 in vitro 114a

xvii

Page 20: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Figure 6.15 GST-OMP25 inhibits the phosphorylation of GST-Sab and

GST-α1-Syntrophin by SAPK3 in vitro 114b

xviii

Page 21: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

LIST OF TABLES

Table 1.1 Cells and tissues with detected expression of SAPK3

mRNA or protein. 11a

Table 2.1 List of Primary Antibodies Used During the Course

of this PhD Project 19a

Table 2.2 List of Secondary Antibodies Used During the Course

of this PhD Project 19b

Table 2.3 Eukaryotic Expression Vectors and Inserts. 22a

Table 2.4 GST- and MalE-Fusion Proteins. 42a

Table 2.5 List of Mutations Made to Proteins During the Course

of this PhD Project 47a

Table 6.1 Summary of SAPK3 interacting clones isolated

using yeast two-hybrid analysis 107a

xix

Page 22: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

ABSTRACT

Cells need to be able to detect changes in their surrounding environment

and transduce these signals into the appropriate cellular compartments. One of the

major ways that the cell achieves this signal transduction is through the process of

phosphorylation. Protein kinases are the enzymes responsible for catalysing this transfer

of phosphate groups from ATP to amino acid residues of their specific substrates. A

subfamily of serine/threonine kinases known as the Mitogen-Activated Protein Kinases

(MAPKs) is essential in a diverse range of cell processes including growth, metabolism,

differentiation and death.

The first identified MAPKs, the Extracellular Signal-Regulated Kinases

(ERKs), were found to be activated in response to mitogenic stimuli such as growth

factors. However, since the discovery of the ERKs, other pathways leading to the

activation of related kinases have been recognised. These kinases are preferentially

activated in response to stress, and are thus termed “Stress-Activated Protein Kinases” or

SAPKs. They consist of the c-Jun N-terminal kinase isoforms 1, 2 and 3 (also called

SAPK1γ, SAPK1α and SAPKβ respectively) and the p38 MAPKs, p38α, p38β, p38γ

and p38δ (also called SAPK2a, SAPK2b, SAPK3 and SAPK4 respectively). A major

challenge in this field has been to identify the substrates and functions of the SAPKs.

This has been partly achieved by the development of inhibitors for the JNK MAPKs and

SAPK2a/b. However, no inhibitors currently exist that specifically inhibit SAPK3 and

SAPK4. Therefore, elucidating the function of these SAPKs has proved more difficult.

Recent studies suggest that SAPK3 may play a unique role in the cell

compared to other members of the p38 MAPK family. For example, several signalling

proteins appear to specifically activate SAPK3 in certain circumstances while not

activating other members of the p38 MAPK family. In addition, SAPK3 contains a

unique sequence motif that allows it to bind to specialised domains known as PDZ

domains. The interaction of SAPK3 with proteins containing these domains may

regulate its subcellular localisation and interactions with other proteins in the cell.

xx

Page 23: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

This project was undertaken to expand the knowledge on the expression,

localisation, substrate specificity and binding partners of SAPK3. In Chapter 3 of this

thesis, a SAPK3 monoclonal antibody was evaluated for its ability to specifically

recognise endogenous SAPK3 protein. SAPK3 was found to be expressed in

immortalised cell lines and primary cultures of neonatal rat myocytes, and to be co-

localised with the mitochondria of these cells. This co-localisation remained unaltered

in response to treatment with the nuclear export inhibitor Leptomycin B, and with

exposure to osmotic shock, suggesting that SAPK3 substrates may be localised at the

mitochondria.

In Chapter 4, the expression of SAPK3 in different tissue types was

examined. SAPK3 was found to be expressed at its highest levels in skeletal muscle

tissue, with lower levels in other tissues such as lung, testes and heart. SAPK3

expression was detected in heart samples of various mammalian species, suggesting its

evolutionary conservation. SAPK3 was also identified in samples of left ventricular

tissue taken from patients with heart failure and non-failing donor hearts, and during

pre- and post-natal development of the rat and mouse heart. These results suggest a

potential role for SAPK3 in heart disease and development.

In Chapter 5 SAPK3 was shown to phosphorylate various mitochondrial

substrates, including various members of the Bcl2 protein family, and Sab, an in vitro

substrate previously attributed to JNK. Mutational analysis of this substrate identified

sequence determinants required for SAPK3 phosphorylation. Finally, Chapter 6

employed a Yeast 2-hybrid screening approach to identify binding partners that may

interact with SAPK3 in a cellular context. This screen identified the PDZ proteins Lin-

7C and Scribble, suggesting that SAPK3 interactions are governed by its PDZ domain

binding motif. SAPK3 was also shown to interact with the mitochondrial PDZ protein

OMP25. Subsequently, it was demonstrated that OMP25 could inhibit SAPK3 activity,

suggesting a novel mechanism for negative regulation of this kinase within the cell.

xxi

Page 24: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

In conclusion, the results presented in this thesis expand the knowledge on

a poorly-characterised member of the p38 MAPK family. The tissue distribution,

mitochondrial localisation, substrate specificity and binding partner selection place

SAPK3 as a unique member of the p38 MAPK family. In combination with future in

vivo studies employing the use of specific inhibitors and knockout models of this kinase,

these results should allow the importance of SAPK3 in cell signalling systems to be

realised.

xxii

Page 25: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Chapter 1

Introduction and Literature Review

Page 26: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

1.1 Intracellular signalling and protein kinases

Alterations in the extracellular environment are communicated into and

throughout a cell via complex intracellular signalling pathways. These signalling

pathways contribute to the regulation of a variety of different cellular responses,

including growth, morphogenesis, differentiation, and death (Assoian, 2002). A variety

of different molecules act in these pathways, and these include the transmembrane

receptor proteins themselves as well as adaptor proteins, scaffolding proteins, enzymes

and inhibitors (Marshall, 2000). Of the enzyme catalysed reactions, protein

phosphorylation has been extensively studied as a critical control mechanism in

intracellular signalling.

Protein kinases are the enzymes responsible for catalysing the transfer of

the γ-phosphate of adenosine triphosphate (ATP) to a specific amino acid residue on a

protein acceptor molecule in a process commonly referred to as phosphorylation (Hanks

and Hunter, 1995). This is summarised in Figure 1.1A. The addition of phosphate

groups to protein substrates alters their structure, and this usually equates to a change in

function or activity. Protein kinases are classified according to the amino acid which

they phosphorylate. Thus, there are the serine/threonine kinases, which can

phosphorylate either serine or threonine residues (Graves et al., 1995), the tyrosine

kinases, which can only phosphorylate tyrosine residues (Tsygankov, 2003), and the

dual specificity kinases, which are capable of phosphorylating serine, threonine or

tyrosine residues in a protein substrate (Dhanasekaran and Premkumar, 1998). It has

also become apparent that other groups of protein kinases exist which do not

phosphorylate these typical amino acid residues, such as the histidine kinases, however

in many of the reported cases of histidine kinases, the γ-phosphate is transferred via an

intermediate amino acid (such as aspartate) before its transfer to histidine of the

substrate (Steeg et al., 2003).

In addition to the catalysis of the phosphorylation reaction, serine/threonine

or tyrosine protein kinases can also be identified by their conserved amino acid

1

Page 27: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

O P O-

O-

O

Protein

Protein Kinase

Protein

Protein Phosphatase H2OPi

OH

Mg2+ ATP Mg2+ ADP

B

GXGXXGK E

DXXXXNDFG

APEDXXXXG R

I II V VIA VIB VII VIII IX X XIIII IV

Amino-terminal lobe (ATP binding)

Carboxy-terminal lobe(peptide binding and phosphotransfer)

Figure 1.1: Protein kinases.(A) The covalent attachment of phosphate to the side-chain hydroxyl group of an amino acid residue (serine, threonine or tyrosine) is carried out by protein kinases. Protein phosphatases catalyse removal of the phosphate by hydrolysis.(B) Protein kinase sequences have been divided into eleven subdomains, which contain regions of conserved sequence. These sequence motifs are critical for processes such as ATP binding and substrate recognition by the kinase. Taken from: Hanks, Genome Biology 4: 111 (2003).

1a

Page 28: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

sequences, which have been divided into a series of subdomains. This subdomain

structure is represented in Figure 1.1B, with conserved amino acids critical for protein

kinase activity indicated. Protein kinases have been conserved throughout evolution,

and are found in all mammalian species as well as in a variety of lower eukaryotes, such

as the worm, fly and yeast, as well as in prokaryotes (Manning et al., 2002a; Steeg et al.,

2003). Eukaryotic protein kinases are one of the largest groups of protein families,

making up 1.5% - 2.5% of all eukaryotic genes (Manning et al., 2002a; Manning et al.,

2002b). The conservation of protein kinases and their high levels of representation in

the genome reinforce the likely importance of protein phosphorylation in intracellular

signalling events.

1.2 The Mitogen-Activated Protein Kinases: Extracellular Signal-Regulated

Kinases as the paradigm

A superfamily of serine/threonine protein kinases known as the Mitogen-

Activated Protein Kinases (MAPKs), is activated in response to a variety of stimuli.

These stimuli include mitogens such as growth factors or hormones, but also cytokines

such as interleukin-1, tumour necrosis factor (TNF) and leukemia inhibitory factor

(LIF), and stresses such us ultra-violet and gamma irradiation, shear stress, osmotic

shock and chemical poisons (Widmann et al., 1999). The activation of MAPKs results

in the phosphorylation of substrates such as transcription factors, cytoskeletal proteins,

membrane proteins and other protein kinases (Widmann et al., 1999). Thus, MAPKs

can serve to link cytoplasmic and nuclear events in response to specific stimuli.

The archetypical MAPK members, the Extracellular signal-Regulated

Kinases (ERKs) 1 and 2, were originally found to be activated in response to insulin and

showed activity towards microtubule-associated protein-2 (Boulton et al., 1990). Since

this initial discovery, ERKs have been implicated in a variety of different cellular

functions, most commonly being associated with cell survival and proliferation (Okuda

et al., 1992; Cobb et al., 1994; Guyton et al., 1996). However ERK signalling has also

been associated with cell death (Stanciu et al., 2000; Stanciu and DeFranco, 2002;

2

Page 29: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Murray et al., 1998; Levinthal and DeFranco, 2004; Alessandrini et al., 1999). These

differences in signalling outcomes may arise from the differences in the kinetics and/or

duration of ERK signalling, the specific cell type in which the signalling occurs, and the

localisation of the ERK signalling events within the cell (Stanciu et al., 2000; Stanciu

and DeFranco, 2002). These additional considerations result in signalling events and

outcomes of even greater complexity.

The ERKs and other members of the MAPK family are activated in

response to mitogens and stress stimuli via a cascade of protein kinases in which a

protein kinase phosphorylates and activates the subsequent downstream protein kinase.

One result of this cascade of events is the amplification of signals (Seger and Krebs,

1995; Tibbles and Woodgett, 1999). The ERK cascade is the best-characterised of the

MAPK pathways and has acted as a paradigm for the study of other MAPK pathways

(Denhardt, 1996; Tibbles and Woodgett, 1999). This kinase cascade is typically

activated via transmembrane receptors such as the Epidermal Growth Factor (EGF)

receptor, which upon engaging their stimulus, autophosphorylate on tyrosine residues

(Margolis and Skolnik, 1994) (Figure 1.2). The following series of events then take

place. The tyrosine phosphorylated EGF receptor recruits proteins containing Src

Homology 2 (SH2) domains such as the protein Grb2, which recognise phosphorylated

tyrosine residues within specific sequence contexts (Kuriyan and Cowburn, 1997). Grb2

also contains a SH3 domain, which allows it to recruit the proline-rich motif of the

nucleotide exchange factor, Son of Sevenless (Sos) (Buday and Downward, 1993;

Chardin et al., 1993; Rozakis-Adcock et al., 1993). Thus, Grb2 acts as an adaptor

protein in the ERK signalling pathway, bringing upstream and downstream components

of the pathway together. Sos is a guanine nucleotide exchange factor (GEF) for the

small GTP-binding protein Ras, allowing for the exchange of Ras-bound guanine

diphosphate (GDP) for guanine triphosphate (GTP) (Chardin et al., 1993; Rozakis-

Adcock et al., 1993). Thus, Ras acts as a molecular switch, being inactive when bound

to GDP, and becoming active when bound to GTP (Shih et al., 1986).

3

Page 30: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Epidermal Growth Factor

EGF Receptor(Receptor Tyrosine Kinase)

PGrb2

Sos Ras

Raf PP

MKK1/2PP

ERK1/2PP

MKK1/2

ERK1/2

CytoskeletalproteinsTranscription

FactorsProtein Kinases

Cell Membrane

Extracellular Region

Intracellular Region

Farnesylmodification

Figure 1.2: Schematic diagram of the organisation of the ERK MAPK pathway.When growth factors (in this case, epidermal growth factor) activate their specific transmembrane receptors, these receptors undergo tyrosine phosphorylation. This creates binding sites for adaptor proteins with SH2 domains, such as Grb2. The SH3 domain of Grb2 binds proline-rich motifs of the guanine nucleotide exchange factor, Sos. This protein mediates the activation of the small GTP-binding protein, Ras, which is held at the cell membrane by a farnesylmodification. Once activated, Ras recruits the MAPKKK Raf to the membrane. Raf is then phosphorylated and adopts an active conformation, allowing it to catalyse the transfer of phosphate from ATP to MKK1/2. Once activated, MKK1/2 can then phosphorylate the MAPKs, ERK1/2. ERK1/2 are then able to phosphorylate a variety of different substrates, including transcription factors, cytoskeletal proteins and other protein kinases. The phosphorylation of these substrates alters their structure and thus their activity, leading to a specific cell response to the original stimulus.

3a

Page 31: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Ras remains localised at the cell membrane due to its C-terminal farnesyl

modification, catalysed by farnesyl transferase (Casey et al., 1989; Maltese et al., 1990;

Reiss et al., 1990). The activation of Ras results in the recruitment of the

serine/threonine kinase Raf to the cell membrane. This recruitment and subsequent

phosphorylation of Raf results in its activation, thus marking the beginning of the

MAPK phosphorylation cascade, with the subsequent phosphorylation of the dual

specificity kinase, MAPK kinase (MKK) 1/2. Once phosphorylated by Raf, MKK1/2

becomes active and is able to phosphorylate the MAPKs, ERK1 and ERK2 (Widmann et

al., 1999).

MAPKs such as ERK1 and ERK2 become activated by dual

phosphorylation on a Thr-Xaa-Tyr motif in a surface loop known as the

“phosphorylation loop”, located in subdomain VIII of the kinase (Figure 1.1) (Payne et

al., 1991; Ahn et al., 1991). In the case of ERK1/2, this phosphorylation occurs on a

Thr-Glu-Tyr motif, and phosphorylation of threonine and tyrosine leads to over 1000-

fold activation of ERK1 and ERK2 (Robbins et al., 1993). Phosphorylation of Thr183

allows ionic contacts to be made with residues in the N-terminal domain of the kinase.

This causes the N- and C-terminal domains to rotate toward each other, organising

catalytic residues within the active site and therefore exposing the substrate binding site

(Canagarajah et al., 1997) (Figure 1.3). The phosphorylation of Tyr185 positions this

residue on the surface of the enzyme where it can participate in substrate recognition. It

is thought that this forms the basis for MAPK specific substrate recognition of serine or

threonine residues that are directly followed by a proline residue (Canagarajah et al.,

1997; Alvarez et al., 1991; Clark-Lewis et al., 1991).

The phosphorylation of Thr183 and Tyr185 appears essential for the

activation of ERK2, and mutations of these residues to glutamate (which can mimic

phosphorylation and thus activity for some protein kinases (Mansour et al., 1994)) does

not confer constitutive activation (Robbins et al., 1993; Canagarajah et al., 1997).

Therefore, studies evaluating the effects of overexpressing activated MAPKs within

cells cannot simply be achieved by the mutation of the MAPK itself. Instead these

4

Page 32: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Figure 1.3: The structure of inactive and active (phosphorylated) ERK2.(A) The phosphorylation loop (Lip) of inactive ERK2 occludes the active site of the kinase. (B) In the active form of ERK2, phosphorylated Thr183 forms ionic contacts with residues in the N-terminal domain, causing domain rotation and a re-organisation of catalytic residues within the active site, exposing the substrate binding site. Phosphorylated Tyr185 is positioned on the surface of the enzyme where it can participate in substrate recognition. This is thought to form the basis for ERK2 recognition of substrate phosphorylation sites that are directly followed by a proline residue. The catalytic loop (“C-loop”) in subdomain VI contains the conserved Asp147 and Asn152 residues which are important in catalysis. Taken From: Canagarajah et al., Cell 90: 859-869 (1997).

B

A

4a

Page 33: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

studies require the co-expression of the MAPK with its upstream activating kinases or

the addition of a MAPK-activating stimulus to the cells.

Once phosphorylated, ERKs can phosphorylate substrates such as other

protein kinases (Chen et al., 1992), cytoskeletal proteins (Lefebvre et al., 2002; Terret et

al., 2003), or membrane proteins (Wu et al., 2003). Alternatively, ERKs have been

shown to translocate to the nucleus, where they phosphorylate transcription factors

(Chen et al., 1992; Boulton TG et al., 1991).

The phosphorylation of substrates by MAPKs is proline-directed in that

MAPKs phosphorylate serine or threonine residues directly followed by a proline.

Specifically, the consensus site for MAPK phosphorylation has been determined as Pro-

Xaa-Ser/Thr-Pro or Xaa-Xaa-Ser/Thr-Pro, where Xaa is any amino acid (Alvarez et al.,

1991; Clark-Lewis et al., 1991). This was determined by sequence alignment of

different MAPK substrates, as well as by the use of synthetic peptides to define the

optimal consensus phosphorylation site (Alvarez et al., 1991; Clark-Lewis et al., 1991).

However not all serine or threonine residues followed by proline residues are

phosphorylated in intracellular proteins and clearly additional substrate recognition

motifs must exist for the MAPKs.

1.3 Parallel MAPK signalling pathways

Since the initial identification of the ERK MAPK signalling pathway, other

similar pathways have been identified (Bogoyevitch and Court, 2004; Barr and

Bogoyevitch, 2001; Kyriakis and Avruch, 2001). These pathways are most commonly

activated in response to stress stimuli rather than mitogenic stimuli. The two pathways

that have been studied the most to date are those leading to the activation of the c-Jun N-

terminal Kinases (JNKs) and the p38 MAPKs (Kyriakis and Avruch, 2001).

5

Page 34: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

1.3.1 The JNK MAPK family

The JNK MAPKs were first identified as kinases activated in response to

UV irradiation that could phosphorylate amino acids Ser63 and Ser73 of the

transcription factor c-Jun and result in increases in its transcriptional activity (Derijard et

al., 1994; Hibi et al., 1993; Westwick et al., 1994). The JNK MAPKs are

phosphorylated by the upstream activators, MKK4 and MKK7 (Derijard et al., 1995;

Yao et al., 1997) (Figure 1.4). Since their initial identification, the JNKs have been

implicated in a diverse number of processes including cell survival, proliferation,

differentiation, development and apoptosis (Lamb et al., 2003; Du et al., 2004; Nagata et

al., 1998; Kim and Freeman, 2003; Kuan et al., 1999; Deng et al., 2003). Due to their

activation in response to stress stimuli, the JNKs have also been referred to as “Stress-

Activated Protein Kinases” (SAPKs) and the three JNK isoforms (JNK1, JNK2 and

JNK3) are also termed SAPKγ, SAPKα and SAPKβ respectively (Cohen, 1997).

1.3.2 The p38 MAPK family

The p38 MAPKs are also activated mainly in response to environmental

stress, as well as cytokines such as interleukin-1 (IL-1) and tumour necrosis factor

(TNF)-α (Han et al., 1994; Raingeaud et al., 1995). The archetypical member of the p38

MAPK family, p38α, was first identified when activated in response to

lipopolysaccharide (Han et al., 1994). Since this initial discovery, a further three p38

MAPK family members have been identified, namely p38β, p38γ, and p38δ (Jiang et al.,

1996; Li Z et al., 1996; Jiang et al., 1997). Due to their activation in response to stress,

the p38 MAPKs are also referred to as SAPK2a, SAPK2b, SAPK3 and SAPK4,

respectively (Cohen, 1997).

SAPK2a and SAPK2b share 74% sequence identity and comparable tissue

distribution patterns with regards to mRNA expression (Jiang et al., 1996).

Furthermore, these two kinases share similar substrate preferences and are both

susceptible to inhibition by the ATP-competitive pyridinyl imidazole SmithKline

6

Page 35: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Raf, Mos

MEKK1-4, MLK2/3,

TPL2, DLKTAK1,

PAK, ASK1MKKK

MEKK1

MKK MKK1, MKK2

MKK4, MKK7

MKK3,MKK6

MKK4

MAPK ERK JNK p38/SAPK2a

Figure 1.4: Parallel MAPK phosphorylation cascades.MAPKs are activated by a cascade of protein kinases in which a MKKK phosphorylates and activates a MKK which then phosphorylates and activates a MAPK. Although cross-talk occurs at the level of the MKKK, the interactions of MKKs with their MAPKs is very specific. This is supported by MKKs having the lowest number of members in the MAPK cascade. In contrast, a relatively large number of MKKKs have been identified. These proteins often contain motifs suchas Pleckstrin Homology (PH) domains, proline-rich regions for binding SH3 domains, as well as phosphorylation sites for serine/threonine and tyrosine kinases. The presence of these different domains allows for a diverse set of interactions within each cascade (Garrington and Johnson, 1999).

6a

Page 36: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Beecham inhibitors SB202190 and SB203580 (Jiang et al., 1996; Enslen et al., 1998).

In contrast, the more recently identified members of the p38 MAPK subfamily, SAPK3

and SAPK4, are insensitive to inhibition with these compounds (Kumar et al., 1997;

Cuenda et al., 1997; Goedert et al., 1997a). It has been determined that the alteration of

a single amino acid in the ATP binding region of JNK1, SAPK3 or SAPK4 confers

sensitivity to SB203580, highlighting the structural similarities of the MAPK

superfamily (Eyers et al., 1998). Due to the lack of inhibitors for SAPK3 and SAPK4,

elucidating the substrate specificity and function of SAPK3 and SAPK4 has proved to be

a more challenging task (Cohen, 1997).

The p38 MAPK family members are phosphorylated by the direct upstream

activators, MKK3 and MKK6 (Enslen et al., 1998) (Figure 1.4). Phosphorylation of the

p38 MAPK family members occurs on a Thr-Gly-Tyr motif in their activation loop

(Raingeaud et al., 1995), in contrast to the ERKs where phosphorylation occurs on a

Thr-Glu-Tyr motif (Payne et al., 1991; Ahn et al., 1991), and JNKs, on a Thr-Pro-Tyr

motif (Derijard et al., 1994). SAPK2a, SAPK3 and SAPK4 can be phosphorylated by

either MKK3 or MKK6, whilst SAPK2b can be phosphorylated by MKK6 only (Enslen

et al., 1998; Fleming et al., 2000). MKK4, and to a lesser extent, MKK7, are also

capable of phosphorylating SAPK2a (Lin et al., 1995; Derijard et al., 1995; Moriguchi et

al., 1995; Fleming et al., 2000). This suggests that some level of cross-talk may occur

between the different MAPK pathways at the level of MKKs in certain circumstances.

In a unique mechanism, SAPK2a has also been found to associate with transforming

growth factor-β-activated protein kinase 1-binding protein 1 (TAB1), and this interaction

allows SAPK2a to autophosphorylate, conferring SAPK2a activity that is comparable to

incubation with MKK6 (Ge et al., 2002). Thus, it appears that other mechanisms

besides the typical MKKK-MKK-MAPK cascade may exist to activate members of the

MAPK family (Ge et al., 2002).

7

Page 37: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

1.3.3 Regulation of interactions in the MAPK pathways

MAPK signalling events appear to be controlled by a variety of factors,

including binding domain interactions, scaffolding interactions, and spatial and temporal

organisation of pathway components (Schaeffer and Weber, 1999). A substrate docking

site, comprised of a series of acidic amino acids, has been identified in the C-terminal

domain of MAPKs (Tanoue et al., 2000; Kallunki et al., 1994). This site is important for

the specificity of interactions between MAPKs and their upstream activators,

phosphatases and substrates (Tanoue et al., 2000). For example, this substrate-binding

groove is necessary for the binding and subsequent phosphorylation of c-Jun by JNK2

(Kallunki et al., 1994). The replacement of this region of sequence in JNK1 with the

corresponding sequence in JNK2 improved the JNK1-c-Jun binding interaction, to the

strength of the JNK2-c-Jun interaction (Kallunki et al., 1994). Recent studies have

determined that MAPK substrates, activators and phosphatases contain “Docking

domains” or “Kinase Interaction Motifs” (KIMs) (Gavin and Nebreda, 1999; Jacobs et

al., 1999; Smith et al., 1999; Tanoue et al., 2000). These KIMs are comprised of short

clusters of positively charged amino acids, thus explaining the requirement for acidic

residues in the MAPK docking regions (Tanoue et al., 2000).

Several different “scaffolding” proteins have been identified in the MAPK

pathways, which are thought to be responsible for bringing the individual components of

the phosphorylation cascades into the correct location and orientation for signal

transduction (Morrison and Davis, 2003). For example, JNK-interacting protein 1 (JIP1)

selectively binds to JNK, the upstream JNK activator MKK7, and the MKK7 activators

mixed-lineage kinase 3 (MLK3) and dual leucine zipper-bearing kinase (DLK), and

facilitates signalling through this pathway (Whitmarsh et al., 1998). Furthermore,

MKK/ERK kinase (MEK) partner 1 (MP1) selectively binds to ERK1 and MKK1,

enhancing MKK1 activation by Raf in vitro and ERK activation when overexpressed in

cells (Schaeffer et al., 1998). Furthermore, despite the sequence similarity between

ERK1 and ERK2, MP1 does not bind to ERK2 and MKK2, suggesting that scaffolding

proteins may represent a regulatory mechanism for selectively activating specific protein

8

Page 38: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

kinase members within a single MAPK pathway (Schaeffer and Weber, 1999).

Recently, other potential scaffolds for MAPK pathways have also been identified,

including mitogen-activated protein kinase organizer 1 (MORG1) (Vomastek et al.,

2004) and dystroglycan (Spence et al., 2004). It is likely that other scaffolding proteins

will be identified that are responsible for organising other MAPK signalling cascades in

response to specific extracellular stimuli.

The subcellular localisation of MAPKs allows the correct transmission of

signals to their substrates. This localisation may be regulated by the interaction of

MAPKs with as yet unidentified anchoring proteins, similar to the Protein Kinase A

anchoring molecules that target that kinase to its specific subcellular locations (Klauck

et al., 1996; Michel and Scott, 2002). Alternatively, this localisation may be mediated

by MAPK substrates or upstream regulators. For example, the ERK1/2 upstream

activators, MKK1/2, contain a leucine-rich stretch of sequence which acts as a nuclear

export signal (NES) (Fukuda et al., 1996). In unstimulated cells, ERK1/2 remains

localised in the cytoplasm complexed with MKK1/2 (Fukuda et al., 1997a). The

activation of ERK leads to the dissociation of the MKK1/2-ERK1/2 complex and the

translocation of ERK1/2 to the nucleus, whilst MKK1/2 remains in the cytoplasm

(Fukuda et al., 1997a). Mutation of the MKK1/2 NES exaggerates the activity of

ERK1/2 (Fukuda et al., 1997b). This suggests that the MKK1/2 NES acts as a

regulatory mechanism to ensure the correct localisation of ERK1/2 and prevent

unwanted sustained signalling events (Fukuda et al., 1997b).

It has also been shown that ERKs can remain cytoplasmic or translocate to

the nucleus following their activation. It has become increasingly clear that this

translocation is dependent upon the original stimulus and the level of ERK activation.

Thus, stimulation of PC12 cells with Nerve Growth Factor (NGF) leads to the sustained

activation of ERKs, their translocation to the nucleus, and cell differentiation, whereas

EGF stimulation results in transient ERK activation, cytoplasmic localisation, and cell

growth (Nguyen et al., 1993; Qui and Green, 1992; Boglari et al., 1998). However, this

response appears to also be dependent on the cellular context, since sustained activation

9

Page 39: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

of ERKs in NIH3T3 fibroblasts leads to cellular growth and transformation (Cowley et

al., 1994). The sustained versus transient activation of MAPK family members may also

be dependent on negative feedback loops and the action of specific phosphatases

(Schaeffer and Weber, 1999; Camps et al., 2000; Keyse, 2000; Theodosiou and

Ashworth, 2002).

1.4 Stress-Activated Protein Kinase-3

The focus of the studies presented in this thesis has been the p38 MAPK

family member, Stress-Activated Protein Kinase-3, or SAPK3. In the following

sections, a more detailed description is given on the similarities shared between MAPK

and SAPK3 signalling. Major differences are also highlighted, emphasising that this

protein kinase may play unique roles in intracellular signalling.

1.4.1 Evolutionary conservation and structure of SAPK3

The p38 MAPK family member SAPK3, with a calculated molecular

weight of 42 kDa, was initially isolated from human (Li et al., 1996), mouse (Lechner et

al., 1996) and rat (Mertens et al., 1996) tissue, and was called p38γ MAPK, ERK6, and

SAPK3 respectively. Due to the sole use of the rat sequence for this project, SAPK3

will be the name used throughout this thesis. The structure of phosphorylated SAPK3 is

shown in Figure 1.5 (Bellon et al., 1999). Activation of SAPK3 is conferred by

phosphorylation on Thr183 and Tyr185 within a Thr-Gly-Tyr motif within the

phosphorylation loop sequence within subdomain VIII (Mertens et al., 1996).

Similarities can be drawn when comparing the structure of phosphorylated SAPK3 and

phosphorylated ERK2. For example, phosphorylation of Thr183 in SAPK3 promotes

the rotation of the N- and C-terminal domains toward each other, organising catalytic

residues within the active site and exposing the substrate binding site (Bellon et al.,

1999). Furthermore, in the active SAPK3 structure, phosphorylated Tyr185 shows an

almost identical conformation to that seen in active ERK2, suggesting that both of these

10

Page 40: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

N-terminal lobe

P-Thr 183; P-Tyr 185

C-terminal lobe

ATP

Figure 1.5: The structure of activated SAPK3 complexed with ATP.The structure of activated SAPK3 is similar to that of activated ERK2. Phosphorylated Thr183 and Tyr185 are shaded in green. When compared to the structures of inactive ERK2 and SAPK2a, phosphorylation of Thr183 in subdomain VIII of SAPK3 promotes the rotation of the N- and C-terminal domains toward each other, organising catalytic residues within the active site and exposing the substrate binding site. Phosphorylated Tyr185 is positioned on the surface of the kinase where it can participate in specific substrate recognition. Taken from Bellon et al., Structure 7: 1057-1065 (1999).

10a

Page 41: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

MAPKs use the same mechanism for specific recognition of proline in their substrates

(Bellon et al., 1999).

SAPK3 shows 60% sequence homology to mouse SAPK2a, 45% to the

SAPK2a yeast homologue, Hog1, 47% to human JNK1, and 42% to rat ERK2 (Mertens

S et al., 1996). In addition to being cloned from human, mouse and rat, SAPK3 has also

been cloned from zebrafish, and more recently, Xenopus (Fabre S et al., 2000;

Perdiguero et al., 2003). The identification of SAPK3 expression in a range of species

suggests the conservation of SAPK3 throughout evolution, and thus supports an

important role for this stress-activated protein kinase.

1.4.2 SAPK3 expression and activation

SAPK3 was initially found to be expressed in a variety of different tissues

at the mRNA level, including heart and lung, with highest levels in skeletal muscle

(Mertens et al., 1996; Lechner et al., 1996; Li et al., 1996). Since its initial cloning,

SAPK3 has also been found to be expressed in a variety of other cells and tissues, and

these are summarised in Table 1.1.

An initial study found that SAPK3 was activated by stresses such as

osmotic shock, anisomycin (a protein synthesis inhibitor) and UV irradiation as well as

pro-inflammatory cytokines such as IL-1 and TNF-α when overexpressed in HEK293

cells, supporting its role as a “stress-activated” kinase (Cuenda et al., 1997). In other

reports, overexpressed SAPK3 has also been activated in response to the DNA damaging

agent cisplatin (Pillaire et al., 2000; Losa et al., 2003).

Further studies have since examined the activation of endogenous SAPK3,

and most of these have been performed in immortalised cell lines. Using antibodies

directed against the phosphorylated form of p38 MAPKs, it was determined that SAPK3

and SAPK2a could be activated in response to hypoxia in PC12 cells (Conrad et al.,

1999). Furthermore, SAPK3 was shown to be activated by γ-irradiation in U2OS cells

11

Page 42: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Table 1.1: Cells and tissues with detected expression of SAPK3 mRNA or protein.

Tissues:Skeletal musle (Mertens et al., 1996; Lechner et al., 1996; Li et al., 1996)Lung, heart, testes (Mertens et al., 1996)

Blood cells:early hematopoietic cells (Uddin et al., 2004)primary erythroid progenitors (Uddin et al., 2004)endothelial cells (Hale et al., 1999)

Immortalised cell lines:C2C12 myoblasts (Lechner et al., 1996; Tortorella et al., 2003)Sol8 and L6 myoblasts (Tortorella et al., 2003)OVCAR3 ovarian carcinoma cell line (Abdollahi et al., 2003)

Tissues:Skeletal muscle (Boppart et al., 2000)

Cells of neural origin:hippocampal neurons (Kimonides et al., 1999; Sabio et al., 2004)cerebellar granular cells (Sabio et al., 1999)

Blood cells:lymphocytes (Fabre et al., 2000)

Immortalised cell lines:C2C12 myoblasts (Cuenda and Cohen, 1999)PC12 cells (Conrad et al., 1999; Sabio et al., 2004) THP1-wtCD14 and U937 human monocytic cell lines (Fearns et al., 2000)OVCAR3 ovarian carcinoma cell line (Abdollahi et al., 2003)SH-SY5Y cells (Singh et al., 2003)OCM-1 human melanoma cell line (Pillaire et al., 2000)U2OS human osteosarcoma cell line (Wang et al., 2000)

mRNA

Protein

11a

Page 43: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

(Wang et al., 2000). Another study examined the activation of JNK and p38 isoforms in

skeletal muscle samples taken from marathon runners, and showed preferential

activation of JNK1 and SAPK3 (Boppart et al., 2000). Thus, these studies with

endogenous SAPK3 support a role for SAPK3 as a “stress-activated” kinase.

Recent studies have shed some light on the upstream signalling events that

lead to SAPK3 activation. The different proteins implicated in the activation of SAPK3

are represented in Figure 1.6. One study has suggested that the GTP binding protein Rit

is involved in SAPK3 activation leading to cellular transformation (Sakabe et al., 2002).

Other studies have examined the role of the GTP binding protein RhoA in SAPK3-

specific activation (Marinissen et al., 2001; Singh et al., 2003). In these reports, there

appeared to be specific activation of SAPK3 via these signalling intermediates.

Therefore other MAPKs are not always activated in parallel with SAPK3, suggesting a

unique role for this kinase.

Several kinases have also been implicated in signalling upstream of

SAPK3. Polyploidy-Associated Protein Kinase (PAPK) and Protein Kinase N (PKN)

have been shown to be required for SAPK3 activation, being implicated as MKKKKs in

the pathway leading to SAPK3 phosphorylation (Marinissen et al., 2001; Takahashi et

al., 2003; Nishigaki et al., 2003). Furthermore, the role of PKN as a potential scaffold

protein in this pathway has been demonstrated (Takahashi et al., 2003). Both Mixed

Lineage Kinase-Related Kinase (MRK) and the oncoprotein Cot have been implicated as

MKKKs in the phosphorylation cascade leading to SAPK3 activation (Gross et al.,

2002; Chiariello et al., 2000). Finally, both MKK6 and MKK3 are able to phosphorylate

SAPK3 on its TGY motif, resulting in activation (Cuenda et al., 1997; Enslen et al.,

1998). It has been suggested that the level of MKK6 activity may dictate its preference

for p38 MAPK family member phosphorylation (Alonso et al., 2000). Thus, low levels

of MKK6 activity have been found to only activate SAPK2a, whereas highly activated

MKK6 can phosphorylate SAPK2a and SAPK3, both in vitro and in cells (Alonso et al.,

2000). This suggests that the level of MKK6 activity from a given stimulus may

determine the pattern of downstream p38 MAPK signalling events (Alonso et al., 2000).

12

Page 44: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Hypoxia, inflammatory cytokines, osmotic stress, UV and γ-irradiation

Stimulus

Rit, RhoAG-Proteins

MKKKK PKN

MKKK MRK, PAPK, Cot

MKK MKK3, MKK6

SAPK3MAPK

Figure 1.6: Potential signalling intermediates leading to the activation of SAPK3.The general arrangement of the SAPK3 signalling pathway follows a similar series of steps shown in Figure 1.4 for the ERK, JNK and p38/SAPK2a pathways. The SAPK3 signalling pathway is activated by a variety of different stresses. The activation of small G-proteins leads to the activation of upstream kinases and the subsequent phosphorylation of specific signalling intermediates ultimately leads to the phosphorylation and activation of SAPK3.

12a

Page 45: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

1.4.3 SAPK3 substrate specificity and potential roles in the cell

SAPK3 can phosphorylate a variety of different substrates in vitro. These

include transcription factors such as Activating Transcription Factor 2 (ATF2), Elk-1,

Myocyte Enhancing Factor (MEF) 2A, MEF2B and MEF2D; cytoskeletal proteins such

as the microtubule-associated protein Tau; and proteins localised at the cell membrane

such as α1-Syntrophin and Synapsin-Associated Protein 90 (SAP90) (Kumar et al.,

1997; Cuenda et al., 1997; Goedert et al., 1997a; Marinissen et al., 1999; Goedert et al.,

1997b; Hasegawa et al., 1999; Sabio et al., 2004). Only two of these proteins have been

confirmed as SAPK3 substrates in intact cells, Tau (Buee-Scherrer and Goedert M.,

2002) and SAP90 (Sabio et al., 2004). The identification of a wide array of SAPK3

substrates suggests diverse roles for this kinase.

Recent reports support this hypothesis. SAPK3 has been implicated in a

variety of disease states, including muscular dystrophy, where microarray analysis

showed that SAPK3 levels were downregulated (Chen et al., 2000). SAPK3 has also

been found to be upregulated in hepatocellular carcinoma (Liu et al., 2003), and

implicated in the activation of the c-jun promoter leading to cellular transformation

(Marinissen et al., 2001). SAPK3 was also found to be downregulated following

interleukin-8 treatment, and upregulated following TNF-related apoptosis-inducing

ligand treatment of the ovarian carcinoma cell line OVCAR3 (Abdollahi et al., 2003). It

has recently been identified that SAPK3 is upregulated in postnatal development of

skeletal muscle, suggesting that SAPK3 may have a role in the development of this

tissue (Tortorella et al., 2003). SAPK3 has also recently been proposed to regulate

glucose transport in skeletal muscle (Ho et al., 2004).

Several studies have implicated SAPK3 in cell cycle arrest and

differentiation. For example, it was initially shown that overexpression of SAPK3 in the

C2C12 myoblast cell line enhanced the differentiation of these cells into myotubes

13

Page 46: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

(Lechner et al., 1996). The identification of RhoA, a mediator of muscle differentiation

(Takano et al., 1998), as an upstream activator of SAPK3 (Marinissen et al., 2001; Singh

et al., 2003) further supports this hypothesis. A recent study implicated SAPK3 in γ-

irradiation-induced G2 arrest (Wang et al., 2000). Furthermore, SAPK3 mediates the

downregulation of cyclin D1 in response to hypoxia, in a calcium-dependent manner

(Conrad et al., 1999; Conrad et al., 2000). Taken together, these results suggest that

SAPK3 may promote cell cycle arrest and differentiation (Seta and Sadoshima, 2002).

1.4.4 SAPK3 intracellular localisation

The localisation of MAPKs within the cell determines the substrates that

they can phosphorylate. For example, the nuclear localisation of active ERK is required

for its phosphorylation of cAMP response element-binding protein (CREB) and the

ternary complex factor Elk-1 (Davis et al., 2000). Overexpressed, phosphorylated

SAPK3 has been shown to be expressed in the nucleus when co-transfected into NIH3T3

cells with MKK6 and RhoA (Marinissen et al., 2001). Endogenous SAPK3 has also

been reported to translocate into the nucleus from the cytosol of hippocampal cells in

response to corticosterone treatment (Kimonides et al., 1999). Other reports have shown

endogenous SAPK3 to be localised at the synaptic junctions in hippocampal neurons

(Sabio et al., 2004) and the neuromuscular junction in skeletal muscle (Hasegawa et al.,

1999). Thus, it appears that SAPK3 may exhibit different localisation patterns in

different cell types, and this may affect its substrate preferences.

1.4.5 SAPK3 contains a unique C-terminal tail

An interesting feature of SAPK3 is its conserved C-terminal, which has the

sequence of Lys-Glu-Thr-Xaa-Leu, where Xaa is Ala in rat, mouse, Xenopus and

zebrafish, or Pro in human (Fabre et al., 2000). This C-terminal tail enables SAPK3 to

bind to proteins containing specialised protein interaction domains called PDZ domains,

named after the first three proteins this domain was identified in: Post-Synaptic Density-

95 (PSD-95), Discs-Large (Dlg), and Zona Occludens-1 (ZO-1) (Hasegawa et al., 1999;

14

Page 47: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Cho et al., 1992; Woods and Bryant, 1989; Anderson et al., 1988). PDZ domains are

often found in scaffolding proteins that also contain a number of other domains. Thus,

PDZ domain-containing proteins often function to organize components of signalling

pathways, or to anchor other proteins to their correct subcellular location (Sheng, 1996;

Hung and Sheng, 2002; Pawson and Scott, 1997). The vast majority of PDZ domain-

containing proteins are localised to the cell membrane (Fanning and Anderson, 1999).

PDZ domains have been classified into 3 distinct classes, based on the C-

terminal or internal hairpin-forming sequence of their protein ligands (Hung and Sheng,

2002). Within an individual class, sequences of representative domains show

approximately 50 – 70% homology (Daniels et al., 1998). Class I PDZ domains bind to

the consensus sequence of Ser/Thr-Xaa-Ø-COOH, where Ø is any hydrophobic amino

acid (Hung and Sheng, 2002). The dissociation constants for PDZ-ligand interactions

range from approximately 50 nM to 10 µM (Songyang et al., 1997).

An example of a Class I PDZ domain bound to its ligand is shown in

Figure 1.7 (Doyle et al., 1996). PDZ domains consist of six β strands and two α helices

(Doyle et al., 1996; Daniels et al., 1998). The protein ligand binds to the PDZ domain at

a 1:1 ratio in a groove of the PDZ domain, between the βB strand and the αB helix,

antiparallel to the βB strand (Doyle et al., 1996). A connecting loop (termed the

carboxylate binding loop) between the βA and βB strands contains the conserved

sequence Gly-Leu-Gly-Phe. These amino acids play an important role in binding the

ligand, forming a hydrophobic pocket in which the most C-terminal amino acid binds,

explaining the requirement for a hydrophobic amino acid at the carboxylate terminal of

the ligand (Doyle et al., 1996).

The SAPK3 C-terminal tail has been found to be essential for the

interaction of SAPK3 with the Dystrophin complex protein, α1-Syntrophin (Hasegawa

et al., 1999). Furthermore, this tail is required for the phosphorylation of α1-Syntrophin

by SAPK3 (Hasegawa et al., 1999). Recently, it has been determined that SAPK3 can

also use this tail to interact with the PDZ domain of another substrate protein, SAP90

15

Page 48: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Figure 1.7: A Class I PDZ domain complexed with a peptide substrate.Class I PDZ domains bind to protein ligands with the consensus sequence of Ser/Thr-Xaa-Ø-COOH. The PDZ domain is comprised of eight elements of secondary structure, with six β-strands and two α-helices. The protein ligand (denoted in orange) binds in a groove between the β-B-strand and the α-B-helix. Amino acids within the β-B-strand, the α-B-helix, and the carboxylate binding loop form contacts with the protein ligand.Taken from Doyle et al., Cell 85: 1067-1076 (1996).

15a

Page 49: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

(Sabio et al., 2004). The identification of further PDZ domain-binding partners for

SAPK3 may help to elucidate its function in different tissues.

1.5 Study aims

At the commencement of this study, little was known about the function

that SAPK3 plays in any tissue; few substrates had been identified for this kinase, and

there were no reports addressing the localisation of endogenous SAPK3 within any cell

type. Furthermore, although the expression of SAPK3 had been previously identified to

be highest at the mRNA level in skeletal muscle, there were no reports on the expression

of endogenous SAPK3 protein in any tissues. Therefore, this study was designed to

address some of these unknown features of SAPK3, with the ultimate aim to more

clearly understand the possible biological role of SAPK3.

Aim 1 of this study was to characterise the use of a SAPK3 monoclonal

antibody to determine its expression and localisation in cells. In Chapter 3, a SAPK3

monoclonal antibody was evaluated for its ability to recognise the endogenous protein in

both Western blotting and immunocytochemical protocols.

Aim 2 of this study was to determine the expression of SAPK3 within

whole tissues rather than isolated cell models. This was considered an important step in

determining the role that SAPK3 may play in a living organism. In Chapter 4, SAPK3

expression was evaluated within different tissues, with emphasis on its expression in the

heart in normal and diseased states. In addition, this Chapter also evaluated the

expression of SAPK3 during development. Specifically, expression levels of this kinase

were compared in the adult and neonatal heart, as well as during embryonic

development from embryonic days 8.5 – 18.5. This provided information on the spatial

expression of SAPK3 (the pattern of tissue expression) and the temporal expression of

SAPK3 (the tissues with earliest expression or high expression) during development.

16

Page 50: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Aim 3 of this study was to determine the substrate specificity of SAPK3.

Studies in Chapter 5 involved the development of an in vitro protocol to activate SAPK3

for subsequent use in the identification of potential substrates of this kinase. In addition,

by using site-directed mutagenesis, sequence determinants responsible for the favouring

of certain phosphorylation sites by SAPK3 were evaluated.

Finally, Aim 4 of this study was to use screening approaches to identify

possible substrates and/or binding partners of SAPK3. In Chapter 6, a yeast two-hybrid

screening approach was used to identify potential SAPK3 interacting proteins. Two

PDZ domain-containing binding partners were identified in this screen and evaluated for

their possible role as physiological SAPK3 interactors. The results of this screen

prompted the study into other potential PDZ domain-containing binding partners of

SAPK3.

In summary, by determining the expression patterns, subcellular

localisation, substrate specificity and binding partners of SAPK3, advances have been

made in elucidating the role that SAPK3 may play in mammalian cells.

17

Page 51: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Chapter 2

Materials and Methods

Page 52: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.1 Reagents and chemicals

The general reagents used during the course of this PhD project were of the

highest purity available, and purchased from the following companies:

AJAX Chemicals (NSW, Australia), Ambion Incorporated (Texas, USA), Amersham

Biosciences (England, UK), Amresco (Ohio, USA), Astral (NSW, Australia), BDH

Laboratory Supplies (England, UK), Bio-Rad (California, USA), Calbiochem

(California, USA), Clontech (California, USA), DAKO Laboratories (NSW, Australia),

Fisher Biotec (WA, Australia), Pharmacia Biotech (New Jersey, USA), PIERCE

(Illinois, USA), ProSci Tech (Queensland, Australia), Roche Applied Science (Indiana,

USA), Sigma Chemical Company (Missouri, USA) and Vector Laboratories (California,

USA).

Reagents for mammalian cell line culture, including media, foetal calf serum, and

trypsin-EDTA, were purchased from Invitrogen Life Technologies (California, USA),

Sigma Chemical Company (Missouri, USA) and Thermo Trace (Melbourne, Australia).

Components of bacterial media, including Bacto-Yeast extract and Bacto-Tryptone, and

agar, were purchased from DIFCO Laboratories (Michigan, USA).

Molecular biological reagents were purchased from Invitrogen Life Technologies

(California, USA), Promega, Inc. (Wisconsin, USA), QIAGEN Sciences (Maryland,

USA) and Stratagene (California, USA).

The SAPK3 C-terminal peptide based on the C-terminal PDZ domain-binding motif

sequence of rat SAPK3 (sequence: Biotin-Aminohexanoic acid-Arg-Gln-Leu-Gly-Ala-

Arg-Val-Pro-Lys-Glu-Thr-Ala-Leu-OH) was synthesized by Proteomics International

(Perth, Australia).

[γ-32P]-adenosine triphosphate ([γ-32P]-ATP) was purchased from Perkin Elmer

(Massachusetts, USA).

18

Page 53: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.2 Antibodies

Table 2.1 and Table 2.2 list the primary and secondary antibodies,

respectively, used during the course of this PhD project, as well as the suppliers of these

antibodies.

2.3 Methods

2.3.1 Culture of cell lines

Cell lines (human embryonic kidney (HEK) 293 fibroblasts and Madin-

Darby canine kidney (MDCK) epithelial cells) were maintained in a controlled

humidified environment of 5% (v/v) CO2 at 37°C. All cell lines used were grown in 175

cm2 tissue culture flasks in Dulbecco’s Modified Eagle Medium (DMEM) supplemented

with 10% (v/v) heat-inactivated foetal calf serum (FCS) and 1% (v/v) penicillin-

streptomycin (hereafter referred to as "growth DMEM").

To passage confluent cells, culture flasks were rinsed with 1x Dulbecco’s Phosphate

Buffered Saline (PBS) (without Mg2+ or Ca2+) and then exposed to trypsin-

ethylenediaminetetraacetic acid (EDTA) (0.05 % (w/v) trypsin; 0.53 mM EDTA) for 5

min to dissociate cells from the flask. Growth DMEM (10 ml) was then added to this

cell suspension to inactivate the trypsin. The cell suspension was then centrifuged

(1,000 x g, 5 min, room temperature; RT). The supernatant was discarded and the cell

pellet resuspended in 10 ml fresh growth DMEM. A small volume (1 ml) of this cell

suspension was then added to a 175 cm2 culture flask and made to a total volume of 25

ml with growth DMEM.

A cell count was then derived from a sample of the remaining cell

suspension. This involved taking a small aliquot of cells (20 µl) and diluting with

medium to a final volume of 200 µl. The counting chambers of a haemocytometer were

then filled with this mixture and cells counted using a phase contrast microscope (10x

19

Page 54: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

objective magnification). The number of cells per ml of the original cell culture was

calculated by obtaining an average count per chamber (which holds 0.1 µl), multiplying

this by 104 (to obtain number of cells per ml) and multiplying this number by the

dilution factor (10). Once a cell count was obtained, cells were diluted in growth

DMEM and aliquoted into 60 mm cell culture dishes (6 x 105 cells per dish) for cell

treatments. Cells to be used for microscopy were aliquoted into 35 mm cell culture

dishes (2 x 105 cells per dish) containing glass coverslips (22 x 22 mm).

2.3.2 Isolation of rat neonatal cardiac myocytes

Neonatal Sprague-Dawley rats were sacrificed by decapitation and the

hearts removed into ice-cold filter-sterilised ADS Buffer (116 mM NaCl, 20 mM

HEPES, 1 mM NaH2PO4, 5.5 mM glucose, 5.4 mM KCl and 0.8 mM MgSO4; pH 7.3).

Atria and connective tissue were removed and the remaining ventricles cut and spread

out to expose more surface area for the action of the dissociation enzymes. The hearts

were then transferred to a sterile 100 ml Schott bottle and 6 ml of sterile Dissociation

Mix (0.4 mg/ml collagenase and 0.6 mg/ml pancreatin in ADS buffer) was added. The

Schott bottle was then placed in a 37°C waterbath for 3 min with shaking at 160 rpm.

The supernatant was then discarded, as this first digestion mainly consists of non-viable

cells and red blood cells (MA Bogoyevitch, unpublished observation). Another 6 ml of

sterile Dissociation Mix was added to the hearts and the Schott Bottle placed in the

waterbath for 20 min with shaking at 200 rpm. Following this digestion, the supernatant

was removed into a separate 15 ml Falcon tube and 6 ml Dissociation Mix was added to

the hearts to commence the next digestion. The supernatant containing myocytes was

centrifuged (120 x g, 5 min, RT). The supernatant was then removed and discarded, and

the cell pellet gently resuspended in 5 ml newborn calf serum (NCS) and maintained at

37°C in a tissue culture incubator (5% (v/v) CO2). This digestion procedure was

repeated a further 4 times, and following this, the supernatants containing myocytes

were pooled into 30 ml of NCS.

20

Page 55: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

The cell suspension was then centrifuged (120 x g, 5 min, RT) and the

supernatant discarded. The cell pellet was then gently resuspended in 20 ml of Plating

Media (68% (v/v) M199, 17% (v/v) DMEM, 10% (v/v) horse serum, 5% (v/v) FCS and

0.68% (v/v) penicillin/streptomycin) and re-centrifuged (120 x g, 5 min, RT). The cells

were then again gently resuspended in Plating Media and transferred to a 75 cm2 flask

and maintained at 37°C with 5% (v/v) CO2 for 45 min. This incubation time allowed

“pre-plating” of the cells, where the fibroblasts present within the culture adhered to the

bottom of the flask, but the myocytes remained in suspension. Following this pre-

plating step, the media containing the suspended cardiac myocytes was removed from

the flask into a 50 ml Falcon tube. Plating Media (2 x 5 ml) was then added to the flask

to wash off any remaining myocytes, and these washes added to the Falcon tube to give

a total of 30 ml.

The concentration of myocytes in the cell suspension was determined by

counting of the cells using phase contrast microscopy. An aliquot (100 µl) of cells was

diluted with the addition of 400 µl PBS and 500 µl trypan blue. This cell suspension

was then loaded into a haemocytometer and counted. Viable cells were identified by

their ability to exclude trypan blue that crosses the membrane of dead cells. A typical

yield of cardiac myocytes from the isolation procedure was approximately 4 x 106 cells

per heart.

After counting, cells were seeded into gelatin-coated 60 mm dishes or 35

mm dishes containing laminin-coated glass coverslips, at a density of 4 x 106 or 1 x 106

cells, respectively. Cells were then maintained at 37°C in a humidified atmosphere

containing 5% (v/v) CO2.

2.3.3 Preparation of coverslips for immunostaining

When cells were immunostained, glass coverslips (22 x 22 mm) were used.

It was important to clean the coverslips to allow cell adhesion and optimal visualisation

of cells. Therefore, before use, coverslips were washed in a dilute (120 mM) HCl

21

Page 56: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

solution, rinsed in double-deionised H2O (ddH2O), and stored in 70% (v/v) ethanol until

further use. Immediately prior to use, coverslips were briefly passed through the flame

of a bunsen burner. This evaporated the ethanol and sterilised the coverslips.

To coat glass coverslips with laminin (required for the adhesion of cardiac

myocytes), 10 µg/ml laminin in PBS was pipetted on to the surface of the coverslips.

After 45 – 60 min in a sterile laminar flow cabinet to allow the laminin to coat the

coverslips, excess laminin was removed and the coverslips left to air-dry for

approximately 45 min before the cells were added.

2.3.4 Cell transfection

Plasmids used for transfecting mammalian cells are listed in Table 2.3.

To prepare these plasmids for transfection into mammalian cells, overnight cultures of

the Escherichia coli strain XL1-blue containing the plasmid of interest were subjected to

a plasmid purification protocol, as described in the QIAGEN® Plasmid Maxikit (see

Section 2.3.23). Following the preparation of plasmid DNA, the purity of the DNA was

confirmed by DNA gel electrophoresis, typically using 1% agarose gels containing 200

µg/ml ethidium bromide.

MDCK or HEK293 cells aliquoted into 60 mm tissue culture dishes

(approximately 6 x 105 MDCK or HEK293 cells in 3 ml growth DMEM) or into 35 mm

tissue culture dishes (approximately 2 x 105 MDCK or HEK293 cells in 2 ml growth

DMEM) with glass coverslips were left to adhere to dishes/coverslips overnight (37°C, 5

% CO2). Cardiac myocytes were isolated and aliquoted into 35 mm dishes containing

coverslips and left to adhere to coverslips overnight (37°C, 5 % CO2).

Cells were transfected with the appropriate plasmid DNA using two

different methods, employing Lipofectamine® and Lipofectamine Plus® Reagents, or

using Lipofectamine Reagent® only. Typically, Lipofectamine only was used when no

more than 1.2 µg of DNA was to be transfected into cells. Lipofectamine and

22

Page 57: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Lipofectamine Plus Reagent were used in combination when 1.2 µg – 2 µg of DNA was

to be transfected into cells. These two methods are described below.

2.3.4(a) Lipofectamine only transfection protocol

Preparation of transfection mixtures was performed in sterile bacterial petri

dishes. In sterile Eppendorf tubes, each plasmid DNA was diluted to a concentration of

0.044 mg/ml in sterile PBS. Diluted DNA was then added to each labelled area on a

petri dish to a total amount of 1.2 µg in 27 µl PBS for 60 mm tissue culture dishes, or

0.7 µg in 16 µl PBS for 35 mm tissue culture dishes. PBS (17µl for 60 mm dishes and

10 µl for 35 mm dishes) was then pipetted next to each DNA mixture. Lipofectamine

Reagent (10 µl for 60 mm dishes or 6 µl for 35 mm dishes) was added to the PBS. The

DNA and PBS/Lipofectamine mixtures were then mixed together. The lid was then

replaced on the petri dish for 15 min and the DNA:Lipofectamine complex allowed to

form.

During this incubation time, the cells were washed once in DMEM without

serum (hereafter referred to as "serum-free DMEM"). Serum-free DMEM (1.4 ml for 60

mm dishes and 0.8 ml for 35 mm dishes) was then added to each dish of cells. A small

volume (340 µl for 60 mm dishes; 200 µl for 35 mm dishes) of serum-free DMEM was

added to each DNA:lipofectamine transfection complex and each entire mixture slowly

and gently pipetted into each dish of cells. The cells were then returned to the incubator

(37°C, 5% (v/v) CO2) for 6 hours. Following this incubation, the cells were washed in

growth DMEM and incubated in growth DMEM for 40 hours, or as otherwise specified,

before fixation or lysis.

2.3.4(b) Lipofectamine and Lipofectamine Plus transfection protocol

In sterile Eppendorf tubes, each plasmid DNA was diluted to a

concentration of 1 mg/ml in sterile PBS. Sterile tubes were then labeled for each

individual dish to be transfected. Diluted DNA (up to 2 µg) was then added to each tube

as needed, and the volume made up to 150 µl with 1 x sterile PBS. To this DNA-PBS

mixture, 10 µl of Lipofectamine Plus reagent was added, mixed and left at RT for 15

23

Page 58: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

min. During this time a dilution of the Lipofectamine Reagent was prepared, consisting

of 6 µl Lipofectamine with 150 µl PBS per transfection.

Following the 15 min Lipofectamine Plus incubation, 150 µl of the diluted

Lipofectamine was added to each transfection, mixed and left to incubate at RT for a

further 15 min. This allowed the DNA:Lipofectamine complex to form. During this

incubation time, the cells were washed once in serum-free DMEM, and then serum-free

DMEM was added to each dish (1.2 ml for 60 mm dishes; 0.55 ml for 35 mm dishes).

Following this incubation time, 200 µl of serum-free DMEM was added to the

transfection mixes and pipetted up and down, before being carefully distributed over the

cells in each dish. The cells were then returned to the incubator (37°C, 5% (v/v) CO2)

for 6 hours. Following this incubation, the cells were washed in growth DMEM and

incubated in growth DMEM for 40 hours, or as otherwise specified, before fixation or

lysis.

2.3.5 Cell treatments

Following the plating of cells into dishes, cells were cultured for

approximately 20 hours in growth DMEM, followed by approximately 20 hours in

serum-free DMEM (37°C, 5% (v/v) CO2). Cells were then treated with the appropriate

agonist by the addition of agonists directly to the culture medium or by the replacement

of the culture medium with serum-free medium containing the agonist for specified

periods of time (37°C, 5% (v/v) CO2). Specifically, for sorbitol-treatment of cells,

sorbitol was dissolved directly in serum-free DMEM, and this used to replace the serum-

free DMEM in which the cells had been cultured. As a control, media on cells was

replaced with fresh serum-free DMEM containing no sorbitol. In contrast, for

endothelin-1, isoproterenol and phenylephrine, stock solutions of these agonists were

diluted in serum-free DMEM and then small volumes of these solutions added directly

to the media already covering the cells to give the final desired concentration. The

controls for these treatments were cell cultures with the addition of extra media to each

dish. For interleukin-1, leukemia inhibitory factor and lysophosphatidic acid, stocks

24

Page 59: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

were diluted in PBS containing 0.1% (v/v) bovine serum albumin (BSA), and these

diluted reagents added directly to the media already covering the cells. The control for

these treatments was to add PBS containing 0.1% (v/v) BSA to the dish. Following

treatments, the media was then removed and the cells washed with ice-cold PBS. Cells

were then immunostained (Section 2.3.8) and visualised by Confocal Scanning

Microscopy (Section 2.3.10) or lysed (Section 2.3.11).

2.3.6 Isolation of embryonic mice and embryonic mouse hearts

Wild-type Friends virus B (FVB) mice, aged 2-5 months, were bred by

staff at the L Block Animal Care Unit, Queen Elizabeth II Medical Centre, Nedlands

(Perth). Female mice were driven into oestrus by the administration of gonadotrophin

and chorionic gonadotrophin prior to mating with males. Copulation was determined by

the presence of a vaginal plug the following morning, and this was considered day 0.5 of

pregnancy, and the equivalent of embryonic day (E) 0.5.

Pregnant mice were euthanased by cervical dislocation. The abdominal

cavity was then opened, exposing the uterine horns. These were removed into ice-cold

PBS and each embryo sac was removed carefully to expose the embryo within. If

embryos were to be used for wholemount staining, they were placed into ice-cold Dent’s

fixative solution (20% (v/v) dimethyl sulfoxide (DMSO), 80% (v/v) methanol) and

gently rocked at 4°C overnight. If embryos were to be used for sectioning, they were

placed into ice-cold sectioning fixative (4% (w/v) paraformaldehyde in PBS) and gently

rocked at 4°C overnight. If only the embryonic hearts were required for RNA or protein

isolated, these were quickly removed from the embryos and snap-frozen in liquid

nitrogen.

25

Page 60: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.7 Processing of embryos for sectioning

Following overnight fixation (see Section 2.3.6), embryos were transferred

to 70% (v/v) ethanol and stored at 4°C until processing. The processing procedure was

performed in a Shandon Citadel 1000 processor with a 10-hour cycle, as listed:

90% (v/v) ethanol 90 min

100% ethanol 90 min

100% ethanol 90 min

100% ethanol 90 min

Toluene 60 min

Toluene 60 min

Wax (56°C melting point) 60 min

Wax (56°C melting point) 60 min

Wax vacuum 30 min

Therefore, this procedure comprised of four stages – ethanol, to remove all water from

the embryos; removal of ethanol from the embryos and the addition of a clearing agent

(toluene); addition of wax as a supporting medium for the tissue; and vacuum to remove

all air from the sample. Following processing, embryos were then embedded in wax in

cassettes, ready for sectioning.

Sectioning of embryos was performed using a Leica Rotary Microtome,

model #2135. Sections were cut at a thickness of 5 µm, and floated on to slides coated

with a 175:2 ratio of acetone:3-aminopropyltriethoxysilane. Sections were then placed in

a 45°C oven overnight to dry, then stored at room temperature until use.

26

Page 61: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.8 Immunostaining

2.3.8(a) Immunostaining of cultured cells

Cells were cultured on coverslips in 35 mm tissue culture dishes in

preparation for immunostaining, and were either transfected with plasmid DNA and

incubated in growth DMEM for up to 40 hours following transfection, or plated out

without transfection and cultured for up to approximately 40 hours. If cells were to be

treated with a particular agonist, they were incubated in growth DMEM for

approximately 20 hours after plating, followed by an additional 20 hours in serum-free

DMEM. Cells were then treated with the appropriate agonist for the specified times.

In preparation for staining, media was removed and cells were rinsed with

1x PBS. When Leptomycin B was used, cells were incubated in growth DMEM

containing Leptomycin B (10 ng/ml) for 3.5 hours (37°C, 5% (v/v) CO2), then washed

with 1 x PBS prior to fixation. When Mitotracker Red was used, cells were incubated in

growth DMEM containing 300 µM Mitotracker Red for 45 min (37°C, 5% (v/v) CO2),

then washed with 1 x PBS prior to fixation. Cells were fixed with 4% (w/v)

paraformaldehyde in sodium cacodylate buffer (0.1 M sodium cacodylate trihydrate; pH

7.9) for 20 min at room temperature. Cells were then rinsed thoroughly with PBS, and

permeabilised with 0.2% (v/v) Triton X-100 in PBS for 15 min at room temperature.

Cells were again rinsed in PBS, and then blocked by incubating the coverslips with 5%

(v/v) FCS in Wash Buffer (0.5% Bovine Serum Albumin (BSA); 0.02% glycine in 1 x

PBS) for 60 min at room temperature. Primary antibody (see Table 2.1 for

concentrations) was added to the coverslips and left to incubate for 60 min at 37ºC.

Following washing of cells with Wash Buffer, secondary antibody (see Table 2.2 for

concentrations) was added and incubated with cells for 60 min at 37ºC. Typically, this

secondary antibody was conjugated to a fluorophore for visualisation of cells using

confocal microscopy.

Following the secondary antibody incubation, the cells were again washed

in Wash Buffer, and Hoescht 33258 stain (2 µg/ml in 5 mM Tris-HCl, 1 mM EDTA; pH

27

Page 62: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

7.4) was added for 5 min to stain nuclear DNA. Therefore, the nuclei of all cells on the

coverslip could be visualised and counted under fluorescent illumination. Calculating

the ratio of the number of stained cells to the total number of cells (Hoescht 33258

stained nuclei) allowed the estimation of transfection efficiency. After washing the cells

several times in PBS, the coverslips were air-dried (although not permitted to over-dry),

and a small drop of Vectashield Mounting Medium was added to each coverslip. The

coverslips were then inverted on to glass microscopy slides, and nail polish was used to

seal the edges of the coverslips. Stained cells were then stored at 4°C for not longer than

1 week before being viewed on the confocal microscope.

2.3.8(b) Immunostaining of mouse embryo sections

Sections were de-waxed in 2 x 10 min incubations of xylene, then washed

in 100% ethanol (2 x 5 min) followed by 100% methanol (10 min). Sections were then

fixed overnight in Dent’s fixative solution (4°C).

The following day, sections were rinsed in 100% methanol (10 min, RT),

and then re-hydrated in graded methanol solutions, diluted in ddH2O (75% (v/v); 50%

(v/v); 25% (v/v); 10 min each solution). Sections were then washed in embryo TBST

(140 mM NaCl, 2.7 mM KCl, 25 mM Tris-HCl pH 7.5, 0.1% (v/v) Tween-20) (3 x 5

min), then blocked in embryo blocking solution (2% (w/v) BSA in embryo TBST) for 2

hours at RT. Slides were then incubated with SAPK3 antibody diluted in blocking

solution, or blocking solution only, overnight at 4°C.

The following day, sections were washed in embryo TBST (3 x 5 min),

then incubated in DAKO rabbit anti-rat immunoglobulin diluted in blocking solution for

1 hour (RT). Sections were then washed in embryo TBST (3 x 5 min) before incubation

in rat monoclonal APAAP (soluble complexes of calf intestinal Alkaline Phosphatase

and Anti-Alkaline Phosphatase) for 1 hour (RT), diluted in blocking solution. Sections

were then washed in embryo Tris-Buffered Saline with Tween (TBST) (3 x 5 min), then

washed in NaCl/Tris/MgCl2/Tween (NTMT) buffer (100 mM NaCl, 100 mM Tris-HCl

pH 9.5, 50 mM MgCl2, 0.1% (v/v) Tween-20) (2 x 10 min). This was then removed

28

Page 63: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

from the sections and replaced with substrates (45 µg/ml Nitroblue tetrazolium chloride

(NBT) and 175 µg/ml 5-bromo-4-chloro-3-indolyl-phosphate, 4-toluidine salt (BCIP))

diluted in NTMT buffer. The reaction was allowed to proceed for approximately 1 hour,

until colouration occurred. The reaction was then stopped by washing slides in stop

solution (2 mM EDTA in PBS) (2 x 10 min). Slides were then briefly rinsed in ddH2O,

then dehydrated through graded ethanols diluted in ddH2O (50% (v/v), 70% (v/v), 95%

(v/v), 100%; 2 min per solution). Slides were then placed into xylene (at least 2 min)

then mounted on to coverslips with DEPEX mounting medium, and left to dry overnight

at RT. Embryo sections could then be viewed using a dissecting microscope or inverted

microscope and photographed.

2.3.8(c) Wholemount staining of mouse embryos

Following overnight fixation in Dent’s fixative solution, embryos were

washed with cold 100% methanol (2 x 10 min). The embryos could then be stored at

-20°C for several months, or processed for wholemount staining as follows:

Embryos were rehydrated in graded methanol solutions diluted in embryo

TBST (75% (v/v); 50% (v/v); 25% (v/v); 15 min per solution). Embryos were then

washed in embryo TBST (1 x 15 min), then blocked for 4 hours in embryo blocking

solution (RT). Embryos were then transferred to 2 ml Eppendorf tubes and immersed in

SAPK3 antibody diluted in embryo blocking solution, or embryo blocking solution only,

overnight at 4°C.

The following morning, embryos were washed in embryo blocking solution

(4 x 15 min), then incubated with DAKO rabbit anti-rat immunoglobulin, diluted in

embryo blocking solution, for 2 hours. Embryos were then washed again (4 x 15 min) in

embryo blocking solution, then incubated with rat APAAP solution, diluted in embryo

blocking solution, for 1 hour. Embryos were then washed again (4 x 15 min) in embryo

blocking solution, before washing in NTMT buffer (2 x 10 min). This was then

removed from the sections and replaced with NBT and BCIP diluted in NTMT buffer.

The reaction was allowed to proceed for 15 – 45 min, until colouration occurred. The

29

Page 64: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

reaction was then stopped by washing embryos in stop solution (2 mM EDTA in PBS)

(2 x 10 min).

Following the staining detection, embryos were transferred to cold 4%

(w/v) paraformaldehyde in PBS and fixed overnight at 4°C. The following day,

embryos were washed in PBS (3 x 15 min; RT), then equilibrated in 50% (v/v) glycerol

in PBS for at least 1 hour, before transfer to 70% (v/v) glycerol in PBS. Once embryos

had sunk to the bottom of the tubes they could be visualised using the dissecting

microscope and photographed.

2.3.9 Haematoxylin and eosin staining

To visualise the histological features of embryo sections, they were stained

with haematoxylin and eosin. Embryo sections were placed into a 60°C oven for 30

min, then dewaxed in 3 x 3 min changes of xylene. Sections were then rehydrated by

incubation in 100% ethanol (2 x 3 min), 70% (v/v) ethanol (1 x 3 min) and water (1 x 3

min). Sections were then incubated in haematoxylin stain to stain cell nuclei (1 x 1

min), then washed thoroughly in 2 changes of water (until the water ran clear). Sections

were then washed in 70% (v/v) ethanol (1 x 3 min) before incubation in eosin stain to

stain the cell cytoplasm (1 x 15 sec). Sections were then washed in 3 x 3 min changes of

70% (v/v) ethanol, then dehydrated through 2 x 3 min changes of 100% ethanol.

Sections were then incubated in xylene (3 x 3 min), before mounting on coverslips with

DEPEX mounting medium and allowing to dry overnight at RT. Embryo sections could

then be viewed using a dissecting microscope or inverted microscope and photographed.

2.3.10 Scanning confocal microscopy

Following the immuostaining procedure using fluorescently labelled

secondary antibodies as described in Section 2.3.8a, cells were viewed using the Bio-

Rad MRC 1000/1024 UV Scanning Confocal Microscope (Bio-Rad Microscience,

California, USA) at the Biomedical Confocal Microscopy Research Centre, located in

30

Page 65: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

the Department of Pharmacology, Queen Elizabeth II Medical Centre, Nedlands (Perth).

To visualise red fluorescence (excitation 540 nm; emission 580 nm), a green HeNe

mixed gas laser was used. Green fluorescence (excitation 488 nm; emission 530 nm)

was visualised with a blue Argon ion UV laser. To visualise Hoescht staining, an Argon

ion UV laser was used, selecting the 351 nm laser line. Acquired images were subjected

to Kalman filtering to reduce noise. For all samples, a 40x oil immersion objective lens

and a 1 - 1.5x zoom factor were used.

2.3.11 Preparation of cell extracts

Cell extracts were prepared from untreated cells, or cells that had been treated with

agonists. After treatment of cells with the appropriate agonist, they were washed in ice-

cold PBS. If cells were to be left untreated, they were simply cultured for approximately

40 hours then washed in ice-cold PBS. Following removal of excess PBS, cells were

lysed by addition of 100 µl ice-cold Lysis Buffer (20 mM HEPES, 2.5 mM MgCl2, 0.1

mM EDTA, 20 mM β-glycerophosphate, 100 mM NaCl; pH 7.7) supplemented

immediately prior to use with 0.05% (v/v) Triton X-100, 500 µM dithiothreitol (DTT),

100 µM Na3VO4, 20 µg/ml leupeptin, 100 µg/ml phenyl methyl sulfonyl fluoride

(PMSF) and 20 µg/ml aprotinin. Cell debris was removed by centrifugation (20,800 x g,

10 min, 4°C), and the supernatant collected as the cell lysate. For protein estimation, 20

µl of the lysate was used in the protocol described in Section 2.3.13. The remainder of

the cell lysates were used either to perform immunoprecipitations or sodium dodecyl

sulphate polyacrylamide gel electrophoresis (SDS-PAGE) followed by Western blotting.

For SDS-PAGE samples, the lysate was diluted with 40 µl of 3x concentrated SDS

sample buffer (187.5 mM Tris-HCl, 6% (w/v) SDS, 30% (v/v) glycerol, 15% (v/v) β-

mercaptoethanol, 0.015% (w/v) bromophenol blue; pH 6.8). Samples were then stored

at -20°C prior to use in the protocols described in Section 2.3.18. For

immunoprecipitation, lysate was directly used in the protocols described in Sections

2.3.14 or 2.3.15.

31

Page 66: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.12 Preparation of extracts from tissue samples

Tissue samples from various sources (as referred to throughout this thesis)

were ground to a fine powder under liquid nitrogen in a mortar and pestle, and then

weighed. These samples were then lysed with the addition of Lysis Buffer (5:1; lysis

buffer:tissue) and vortexing. Samples were then placed immediately on ice for 10 min

and then centrifuged (20,800 x g, 10 min, 4°C). The supernatant was then removed and

20µl of this was set aside for protein estimation. To the remainder of the supernatant, 3

x concentrated SDS sample buffer was added in preparation for SDS-PAGE. Samples

were then stored at -20°C prior to use in the protocols described in Section 2.3.18 and

2.3.19.

2.3.13 Protein estimation

Quantitation of protein concentrations was performed using one of two

methods. Both of these methods required the use of a protein standard. This was a

commercial preparation of bovine serum albumin (BSA) with an initial stock

concentration of 2 mg/ml.

2.3.13(a) Spectrophotometric estimation

The first of these methods involved a spectrophotometric assay where a

linear range of known BSA concentrations was prepared (0 - 0.5 µg protein). Dilutions

of the protein sample were also prepared. An acidic dye (Bradford Reagent) was then

added to each protein solution and the absorbance subsequently measured at a

wavelength of 595nm with a microplate reader. The standard curve that was produced

allowed for a relative measurement of protein concentrations in the protein sample of

interest.

2.3.13(b) Estimation using SDS-PAGE separated proteins

The second method was used to confirm both the protein concentration

and purity of recombinant proteins (Section 2.3.28). This method involved preparation

32

Page 67: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

of a linear range of concentrations of BSA (2.5 – 10 µg protein) and a series of dilutions

of the protein of interest in SDS sample buffer. These protein solutions were then

separated by SDS-PAGE. The resulting gel was then stained with Coomassie Brilliant

Blue (Coomassie stain; 45% (v/v) methanol, 45% (v/v) glacial acetic acid, 0.05% (w/v)

Coomassie Brilliant Blue) and destained (7.5% (v/v) methanol, 7.5% (v/v) glacial acetic

acid), therefore allowing the protein of interest as well as any contaminating proteins to

be visualised on the stained gel. Following destaining, the intensity of the bands was

quantitated by photographing the gel and using Image Gauge computer program. The

concentration of the sample protein could then be measured relative to the BSA standard

curve.

2.3.14 Immunoprecipitation for subsequent Western blotting

HEK293 fibroblasts were transfected with mammalian expression

constructs (Table 2.3) as described in Section 2.3.4 and incubated for approximately 24

hours in growth DMEM to allow protein expression. Cells were then lysed and lysates

containing 100 µg protein were incubated with mixing at 4 °C overnight with the

appropriate antibody (amounts listed in Table 2.1). The following morning, 30 µl of

Protein G-sepharose slurry (1:1 Protein G-sepharose:Lysis Buffer) was added and this

mixed at 4°C for a further 3 hours to capture the antibody-antigen complex. Samples

were then briefly centrifuged and the pellet washed three times in Lysis Buffer. Excess

buffer was removed from the pellet and 25 µl of 3 x concentrated SDS-sample buffer

added in preparation for SDS-PAGE according to protocols described in Section 2.3.18.

2.3.15 Immunoprecipitation for subsequent kinase assays

HEK293 fibroblasts were transfected with mammalian expression

constructs (Table 2.3) as described in Section 2.3.4 and the cells then incubated for

approximately 40 hours in growth DMEM to allow protein expression. The cells were

then lysed, and lysates containing 300 µg protein incubated with Flag or Hemagluttinin

(HA) antibodies overnight to capture the expressed proteins (see Table 2.1). The

33

Page 68: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

following day, Protein G sepharose was added to the lysates and mixed at 4°C for 3

hours to capture the antibody-antigen complex. The sepharose resin was then separated

from the lysates by centrifugation and washed twice with Lysis Buffer and twice with

Activation Buffer (50 mM Tris-HCl, 10 mM MgCl2, 20 mM β-glycerophosphate; pH

7.5) to remove any contaminating proteins. Excess buffer was then removed and

immunoprecipitates used in kinase assays (Section 2.3.17).

2.3.16 Activation of GST-SAPK3, GST-JNK2 and GST-ERK2

GST-SAPK3, GST-ERK2 and GST-JNK2 (80 µg/ml of each) were

incubated with MalE-MKK6(DD), GST-MKK1(EE) or GST-MKK4(ED) (160 µg/ml of

each) respectively, in Activation Buffer containing 330 µM ATP, overnight at 30°C (see

Section 2.3.28 for recombinant protein production). The following day, glutathione-

sepharose (GSH-sepharose) resuspended in EDTA-stop solution (10mM EDTA pH 8.0,

50mM Tris-HCl pH 7.5) was added at an equal volume to the reaction mix to terminate

the reaction, and this was mixed end-over-end at 4°C to capture the GST-fusion proteins.

The GSH-sepharose, bound to GST-fusion proteins, was then washed several times in

Activation Buffer to remove the other reaction components, and the GST-fusion proteins

eluted with the addition of 5 mM reduced glutathione in 50 mM Tris-HCl (pH 8.0). This

was repeated a further three times and the supernatants were pooled and dialysed against

Activation Buffer. The proteins could then be directly added to kinase assays (Section

2.3.17), or stored at 4°C for later use.

2.3.17 Kinase assays

Activated kinase (1 µg) was added to immunoprecipitates from HEK293

fibroblasts or 10 µg of recombinant protein substrate (see Section 2.3.28), in Activation

Buffer containing 10 µM ATP and 1µCi-3µCi of [γ-32P]-ATP (as specified in Figure

Legends) then incubated at 30°C for 35 min. When the SAPK3 C-terminal peptide was

used (see Section 2.1 and Chapter 5) it was included at a final concentration of 10µM or

100µM and pre-incubated with the substrate for 10 min at 30°C prior to the protein

34

Page 69: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

kinase assay. This allowed the C-terminal peptide time to bind to GST-α1-Syntrophin

or GST-Sab substrates being used in the assay of SAPK3 activity. When GST-

OMP25(1-177) was used as an inhibitor, 1 – 10 µg of the protein was pre-incubated

with activated GST-SAPK3 for 10 min at 30°C prior to the protein kinase assay. This

allowed GST-OMP25 to bind to GST-SAPK3. The reactions were terminated by the

addition of 3 x concentrated SDS sample buffer to give a 1 x final concentration. The

samples were then boiled and the reaction components separated by SDS-PAGE (10 or

12% acrylamide gels, as specified in Figure Legends). The resulting gels were then

Coomassie stained for 20 min and destained overnight as described in Section 2.3.13b.

Gels were then dried (Bio-Rad model 583 gel dryer), photographed, and exposed to

autoradiographic film. The protein bands of interest were then excised and 32P

incorporation quantitated using Cerenkov counting.

2.3.18 SDS-PAGE

Samples consisting of cell lysates or kinase assay components that had

been diluted in 3 x SDS sample buffer were heated for 5 – 10 min at 95°C. To perform

SDS-PAGE, the small-format mini-PROTEAN II or mini-PROTEAN III gel

electrophoresis system (Bio-Rad) was routinely used. Unless otherwise stated, 10%

resolving gels (10% (w/v) acrylamide, 0.25% (w/v) bis-acrylamide, 0.1% (w/v) SDS,

0.375M Tris-HCl pH 8.8) and 6% stacking gels (6% (w/v) acrylamide, 0.15% (w/v) bis-

acrylamide, 0.1% (w/v) SDS, 0.125 M Tris-HCl pH 8.8) with 10 wells were cast. Gels

of 0.75 mm thickness were used when loading sample volumes not exceeding 25 µl.

Gels of 1.5 mm thickness were used when loading sample volumes not exceeding 50 µl.

These gels were polymerized by the addition of 0.1% (v/v) TEMED and 0.875 M

ammonium persulphate. Gel electrophoresis was performed at 200V (constant voltage)

for approximately 45 min so that the dye front was approximately 0.5 cm from the

bottom of the glass plates. For large-format gels, the Hoefer Vertical Slab Gel SE400

was used in a similar manner, except that electrophoresis was at 100V for approximately

1 hour (until the samples had run out of the loading wells), followed by approximately 4

hours at 160V.

35

Page 70: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.19 Western blotting

Following SDS-PAGE, the stacking gel was discarded and the resolving

gel was soaked in transfer buffer (48 mM Tris-HCl, 39 mM glycine, 1.2 mM SDS, 20%

(v/v) methanol; pH 8.0) for approximately 15 min. Meanwhile, 6 pieces of Whatmann

3MM filter paper and a nitrocellulose membrane, all cut to the size of the SDS-PAGE

separation gels, were soaked separately in transfer buffer. If PVDF membranes were to

be used for transfer, these were first soaked in 100% methanol, followed by soaking in

DDH2O, then in transfer buffer. The transfer was then set up as follows:

Top Cathode (-ve)

3 pieces of presoaked filter paper

polyacrylamide gel

nitrocellulose membrane

3 pieces of presoaked filter paper

Bottom Anode (+ve)

The electrophoretic transfer of proteins to nitrocellulose membranes was

routinely performed with the Trans-Blot Semi-Dry electrophoretic transfer cell (Bio-

Rad). This transfer was performed at 12V (constant voltage) for 45 min for 0.75 mm

gels and 90 min for 1.5 mm gels. In some cases for the transfer of large (>200 kDa)

proteins (see Chapter 6 and experiments evaluating the SAPK3 interacting partner

Scribble), the mini trans-blot module (Bio-Rad), utilizing a wet transfer system, was

used. Here, the transfer tank was filled with transfer buffer, and the polyacrylamide gel

sandwiched next to the nitrocellulose membrane and filter papers held in the transfer

cassette. This wet transfer was performed at 100V (constant voltage) for 60 min and

used only for the transfer of proteins from 0.75 mm gels.

Following transfer, the nitrocellulose membrane was blocked with 5%

(w/v) non-fat milk powder in TBST (10 mM Tris-HCl, 150 mM NaCl, 0.1% v/v Tween

36

Page 71: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

20; pH 7.5) for 60 min with gentle agitation. Membranes were then incubated with

primary antibody (diluted in 5% (w/v) non-fat milk powder or 5% (w/v) BSA in TBST;

see Table 2.1) overnight at 4°C with agitation. When 5% (w/v) BSA was used as the

diluent for the primary antibody, membranes were rinsed in several changes of TBST

after incubation in the blocking prior to the incubation in the presence of the appropriate

primary antibody.

The following morning, the nitrocellulose membrane was washed (3x5 min

in ~50 ml TBST for each wash) to remove any excess primary antibody. The membrane

was then incubated with secondary antibody conjugated to horseradish peroxidase for at

least 2 hours at RT. This secondary antibody was diluted in 1% (w/v) non-fat milk

powder in TBST (see Table 2.2 for antibody dilutions). Following further washing (3x5

min + 1x10 min in ~50 ml TBST for each wash; RT), membranes were treated with

Supersignal Chemiluminescent Substrates for 5 min. Western blots were then exposed

to Hyperfilm for <5 min.

To determine whether transfer of proteins was even across the membrane,

the membranes were briefly re-hydrated in TBS, then incubated in Ponceau S stain

(0.1% (w/v) Ponceau SS Acid Red 150 in 2% (v/v) glacial acetic acid) for approximately

2 min, then destained in 2% (v/v) glacial acetic acid.

2.3.20 Stripping Western blots

Western blot membranes were re-hydrated in TBS for 5 min with gentle

agitation. Strip solution (62.5 mM Tris-HCl pH 6.8, 2% (w/v) SDS, 0.1% (v/v) β-

mercaptoethanol), pre-heated to approximately 60°C, was then added to immunoblots

and incubated in a 60°C waterbath for 20 min. This strip solution was then changed and

membranes incubated for a further 25 min. The strip solution was then removed, and

immunoblots washed extensively in TBS, followed by extensive washing in TBST. The

immunoblots were then blocked in 5% (w/v) non-fat milk powder in TBST, and

processed as for Western Blotting (section 2.3.19).

37

Page 72: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.21 Preparation of competent E. coli

All solutions to be used for making competent cells were prepared sterile

and cooled to 4°C overnight. Two different E.coli strains were used for transformation

with plasmid DNA – the XL1-Blue and BL21 DE3 pLyss cells. For XLI-Blue E. coli,

glycerol stocks were streaked on to a Luria-Bertani (LB; 1% (w/v) bacto-tryptone, 0.5%

(w/v) bacto-yeast extract, 170 mM NaCl; pH 7.0)-agar (15 g/L) plate and incubated

overnight at 37°C. The same protocol was followed for BL21 DE3 pLyss cells except

that the LB plates were additionally supplemented with 100 µg/ml chloramphenicol to

ensure the presence of the pLyss plasmid. The following day, 50 ml of autoclaved LB

(or LB/chloramphenicol) was inoculated with 1 colony and grown to saturation

overnight at 37°C, 200 rpm.

The following day, 2 x 25 ml of the overnight culture was added to 2 x 250

ml of LB (or LB/chloramphenicol for E. coli containing pLyss), equilibrated to 37°C.

These cultures were then grown to an optical density at 650 nm (OD650nm) of 0.2 (37°C,

200 rpm; approximately 30 min). Meanwhile, another 150 ml of culture medium was

equilibrated to 37°C. After the cultures had reached the correct density, 75 ml of

equilibrated medium was added to each culture and grown for a further 30 min, at 37°C,

200 rpm. Cells were then harvested by centrifugation (3000 x g, 20 min, 4°C). The

supernatant was decanted, and cells washed in 90 ml of 100 mM MgCl2. Cells were

then incubated on ice for 5 min before centrifugation (1500 x g, 10 min, 4°C). The

supernatant was decanted, and cells washed with 15-20 ml of ice-cold 100 mM CaCl2.

Cells were then incubated on ice for 20 min, and then harvested by centrifugation at

(1300 x g, 10 min, 4°C). Cells were then resuspended in 3.5 ml of storage solution (85

mM CaCl2, 15% (v/v) glycerol), aliquoted, and slowly frozen in a polystyrene box at

-80°C.

38

Page 73: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.22 Transformation of XL1-Blue and BL21 DE3 plyss E. coli strains with

plasmid DNA

A 100µl aliquot of XL1-Blue or BL21 cells was incubated on ice with

entire ligation mix (in the case of subcloning procedures) or 400 – 800 ng of plasmid

DNA for 40 – 60 min. Cells were then heat-shocked by incubation in a 42°C water bath

for 90 sec, before being plunged into ice for 10 min, and then incubated at RT for 5 min.

Cells were then diluted into 0.5 ml LB media previously warmed to 37°C and allowed to

recover at 37°C for 1 hour, 200 rpm. The E. coli were then plated out on LB-agar plates

containing the required antibiotic/s, and incubated at 37°C overnight.

2.3.23 Preparation of plasmid DNA using the QIAGEN miniprep and maxiprep

kits

E. coli containing the plasmid DNA of interest were streaked out on an LB-

agar plate containing the appropriate antibiotics for selection, and grown overnight at

37°C. When using the miniprep kit, a single colony was selected from the plate the

following day and grown overnight at 37°C, 200 rpm in 5 ml LB media containing the

appropriate antibiotics for selection. When using the maxiprep kit, a single colony was

selected from the plate and grown for 8 hours in 2 ml LB media containing the

appropriate antibiotics. An aliquot of this starter culture (400 µl) was then used to

inoculate 2 L of LB media containing the appropriate antibiotics, and grown overnight

(37°C, 200 rpm). The following day, 700 µl of the overnight bacterial culture was

diluted with the addition of 700 µl 40% glycerol and stored at -80°C in a cryotube as a

glycerol stock. The remaining culture was used to isolate plasmid DNA from the

bacterial cells using the miniprep or maxiprep kit, according to the manufacturer’s

instructions.

39

Page 74: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.24 Isolation of RNA from embryonic, neonatal and adult mouse hearts

All reagents to be used for RNA isolation were diluted where appropriate in

diethyl pyrocarbonate (DEPC)-treated water. Immediately prior to use, all equipment to

be used for RNA isolation was washed in “RNase ZAP solution” (Ambion). Trizol

reagent (500 µl) was added to each group of hearts (3-5 hearts for each embryo stage, 2

hearts for neonatal, 1 heart for adult) and these were then homogenised using a hand-

held mortar and pestle. Homogenates were then centrifuged (12,000 x g, 10 min, 4°C),

and the supernatant retained. The supernatant was incubated for 5 min (RT) before the

addition of 100 µl chloroform. The tube was then mixed vigorously for 15 sec, then

incubated at RT for 3 min. This was then centrifuged again (12,000 x g, 10 min, 4°C),

and the upper aqueous phase transferred into a new tube. The RNA was then

precipitated with the addition of 250 µl isopropanol followed by mixing and then

incubating at RT for 10 min. This solution was then centrifuged (12,000 x g, 10 min,

4°C) and the supernatant discarded. The pellet was then washed in 550 µl 75% (v/v)

ethanol, before centrifugation (7,500 x g, 5 min, 4°C). The supernatant was then

discarded and the pellet air-dried for 10 min, before resuspending in 30 µl sterile ddH2O.

2.3.25 Quantitation of RNA and DNA samples

RNA and DNA, diluted in ddH2O, were quantitated using the Eppendorf

Biophotometer on the “RNA” or “double-stranded DNA” settings, respectively. ddH2O

was used as a blank reading. Calculations of RNA and DNA concentration were

determined based on an A260 of 1 = 40 µg/ml, or A260 of 1 = 50 µg/ml, respectively.

2.3.26 Agarose gel electrophoresis

Prior to electrophoresis, DNA samples were resuspended in 6 x

concentrated DNA loading buffer (0.25% (w/v) Bromophenol blue, 0.25% (w/v) xylene

cyanol, 30% (v/v) glycerol). DNA samples were routinely electrophoresed on 1% (w/v)

agarose gels (1% (w/v) agarose, 40 mM Tris-acetate pH 8.0, 1 mM EDTA, 200 µg/ml

40

Page 75: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

ethidium bromide) in TAE Buffer (40 mM Tris-acetate pH 8.0, 1 mM EDTA) at 90 V

for 50 min in a MiniSub horizontal gel electrophoresis tank (Bio-Rad, California, USA).

DNA bands were then visualised and photographed under UV illumination.

2.3.27 DNA sequencing

Plasmid DNA was sequenced using dye-labelled dideoxynucleotides, and

the method was based on the Sanger dideoxy sequencing method (Sanger, 1981).

Sequencing reactions were carried out in 0.2ml Polymerase Chain Reaction (PCR) tubes.

These reactions typically consisted of 300 – 600 ng DNA template and 1.6 pmol primer,

diluted in Big Dye Terminator Mix (Applied Biosystems; version 3.1) up to a volume of

10 µl. Reagents were mixed together and overlaid with PCR wax to prevent evaporation

of reaction components during the PCR reaction. Reaction components were then

subjected to a PCR reaction with the following conditions:

96°C 10 seconds

25 x 50°C 5 seconds

60°C 4 minutes

4°C hold

Following the sequencing reaction, the PCR products were precipitated by

the addition of 25 µl 95% (v/v) ethanol and 1 µl of 3 M sodium acetate pH 5.0 followed

by incubation for 30 min (RT). The samples were then centrifuged (20 min, 20,800 x g,

RT) and the supernatants removed. The pellets were washed with 250 µl of 70% (v/v)

ethanol, and the samples were then centrifuged again (20,800 x g, 10 min, RT). The

supernatants were decanted and the pellets air dried. The DNA pellets were then

sequenced by the DNA sequencing service at the Australian Neuromuscular Research

Institute (Queen Elizabeth II Medical Centre, Nedlands, Perth).

41

Page 76: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.28 Expression and purification of recombinant proteins

The E. coli strain BL21 DE3 plyss was transformed with expression vectors

encoding either the proteins of interest fused to glutathione-S-transferase (GST) or MalE

(Maltose Binding Protein). Glycerol stocks of each of these E.coli lines were stored at -

80°C. Autoclaved LB Medium (50 ml) containing the required antibiotics (50µg/ml

ampicillin, 10µg/ml chloramphenicol) were inoculated with the glycerol stock of

interest. Cultures were incubated for 12-16 hours at 37°C, 200 rpm. Cultures were then

diluted up to 1 L with LB medium containing the required antibiotics and grown (37 °C,

200rpm) to an optical density of OD600 = 0.6. Following this incubation, the expression

of the GST-fusion protein was induced with 0.1 - 1mM isopropyl-beta-D-thio

galactopyranoside (IPTG) (see Table 2.4) and in the majority of cases the cultures were

left to grow at 37°C, 200 rpm for a further 4 hours. For certain proteins, this induction

was performed at 23°C at 200 rpm for 6 hours (see Table 2.4). The cell pellets were

then centrifuged (3,500 x g, 10 min, 4°C) and resuspended in 20 - 40 ml Resuspension

Buffer (50 mM Tris-HCl, 100 mM NaCl, 1 mM EDTA, 1 mM benzamidine, 10 mM β-

mercaptoethanol, 10 µg/ml leupeptin; pH 8.0). These resuspended pellets were then

snap-frozen in liquid nitrogen and stored at -80°C.

The recombinant proteins were extracted and purified by sonicating the

thawed resuspended E.coli pellets (4 x 45 second bursts) with the Sonifier B-12

(Branson, Danbury, Conneticut, USA). The detergent BRIJ35 was then added to a final

concentration of 0.02% (v/v) and the suspension centrifuged (10,000 x g, 35 min, 4°C)

to remove the cell debris. Glutathione-sepharose or amylose resin 1:1 gel slurry (1 ml)

pre-washed in Wash Buffer (50 mM Tris-HCl, 100 mM NaCl, 1 mM EDTA, 1 mM

benzamidine, 10 mM β-mercaptoethanol, 0.02% v/v BRIJ35; pH 8.0) was added to the

supernatant and incubated with mixing for 1 hour at 4°C. The glutathione-sepharose or

amylose resin was then collected (3,000 x g, 5 min, RT) and washed in Wash Buffer

(1x25 ml and 2x15 ml).

42

Page 77: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

The recombinant proteins were eluted from glutathione-sepharose by

adding 800 µl volumes of 5 mM reduced glutathione in 50 mM Tris-HCl (pH 8.0) or

from amylose resin by adding 800 µl volumes of maltose elution buffer (50mM NaCl,

1mM dithiothreitol (DTT), 0.1mM EDTA, 12mM maltose, 50mM Tris-HCl pH7.5),

mixing briefly, centrifuging, and collecting the supernatant. This was repeated a further

three times and the supernatants were pooled and dialysed against Dialysis Buffer (50

mM Tris-HCl, 100 mM NaCl, 1 mM EDTA, 1 mM benzamidine, 10 mM β-

mercaptoethanol, 0.02% (v/v) BRIJ 35, 5% (v/v) glycerol; pH 8.0). MalE-MKK6(DD),

which was to be used in activating GST-SAPK3, was dialysed into supplemented

Activation Buffer containing no MgCl2 (20mM β-glycerophosphate, 1mM benzamidine,

0.5mM DTT, 20mM HEPES; pH 7.6). The resulting solution containing the

recombinant protein of interest was then quantitated using the Bradford protein assay

and gel electrophoresis (Section 2.3.13), aliquoted, and stored at -80°C until further use.

2.3.29 Recombinant DNA techniques

During the course of this PhD project, recombinant DNA techniques were

used to introduce specific DNA sequences into particular vectors for applications such

as the production of recombinant proteins (Section 2.3.28) and screening in the yeast

two-hybrid system (Sections 2.3.35 – 2.3.44). Two different methods were used for

subcloning –

(i) Restriction enzyme digestion and religation. In this method the vector containing the

insert of interest (“donor vector”) was digested with the appropriate restriction

endonucleases. This insert was then ligated with the new (“acceptor”) vector. An

example of this procedure, used to insert full-length SAPK3 into the pBTM116 vector

for Yeast 2-hybrid screening, is shown in Figure 2.1.

(ii) PCR amplification, restriction enzyme digestion and religation. In this method PCR

primers were designed to flank the insert in the donor vector. Amplification of the insert

with these primers in PCR reactions allowed the introduction of new restriction

endonuclease sites. The PCR fragment and acceptor vector were then digested with the

appropriate restriction endonucleases and subsequently ligated (Figure 2.2 and 2.3).

43

Page 78: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

AmpR

EcoRI site:GAATTC

EcoRI site:GAATTC

EcoRISmaI

BamHISalIPstI

pBTM116~ 5.5 kb

AmpR

LexApEFm-plink

~ 6.5 kb

Full-length rat SAPK3

Polycloningsite

Digest with EcoRI

Run digested vector and insert on gel

pBTM116

SAPK3 insert

“Donor Vector” “Acceptor Vector”

LexA

AmpR

pBTM116(EcoRI digested)

~5.5 kb

Full-length rat SAPK3 insert

Agarose Gel

pEFm-plink

Extract DNA, ligate overnight

Transform DNA into XLI-Blue cells; select for

Ampicillin-resistant colonies

AmpR

LexA

pBTM116~6.6 kb

Full-length rat SAPK3

Figure 2.1. Insertion of full-length rat SAPK3 into pBTM116 for yeast two-hybrid screening.The “donor vector” pEFm-plink (containing full-length rat SAPK3) and the “acceptor vector” empty pBTM116 were digested with EcoRI and the digested products separated on an agarosegel. The resulting bands of DNA were then excised from the gel and the DNA extracted using a QIAGEN Gel Extraction Kit. The vector was treated with alkaline phosphatase, and subsequently incubated with the SAPK3 insert overnight at 16°C, in the presence of T4 DNA Ligase. The following day, the ligation mixture was transformed into competent XLI-Blue cells. Ampicillin-resistant colonies were selected and sequenced to confirm the insertion of full-length SAPK3 into the pBTM116 vector, as an in-frame fusion with LexA.

43a

Page 79: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

AmpR

pcDNA3(HA)~ 6 kb

Full-length rat OMP25

HA tag

rat OMP25 PCR fragment

pVP16~ 8 kb

NLS

VP16

PCR

AmpR

“OMP PDZ Forward Primer”: 5’ CGGAGAGCAGTCAGATGGATCCTGATGAACGG 3’

“OMP PDZ Reverse Primer”:5’ TACAGGAACTCCGCGGCCGCCTAACTCG 3’

Full-lengthrat OMP25

BAmphipathic helix PDZ domain TM

1 73 160 178 200 206

37 160 177PDZ domain

Residues 37-177rat OMP25

Figure 2.2: The design of primers to insert a fragment of OMP25 into the pVP16 vector.(A) pcDNA3(HA) was obtained from P. DeCamilli containing the full-length rat OMP25 sequence. To insert nucleotides encoding amino acids 37-177 of OMP25 into pVP16, primers were created to flank the nucleotides in pcDNA3(HA) encoding amino acids 37-177. The “OMP PDZ Forward” primer sequence included a BamHI restriction site (underlined) to enable insertion of a BamHI site upstream of the required fragment, and extra nucleotides (shown in italics) to ensure an in-frame fusion of the OMP25 fragment with VP16. The “OMP PDZ Reverse” primer included a STOP codon (shown in red), followed by a NotI restriction site (underlined) to insert downstream of the required fragment, by PCR reaction. Digestion of the PCR product and pVP16 with BamHI and NotI could then be carried out, and the PCR product and pVP16 ligated. (B) A schematic of the full-length rat OMP25 protein is shown, and residues 37-177 which were inserted into the pVP16 vector.

43b

Page 80: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

AmpR

pcDNA3(HA)~ 6 kb

Full-length rat OMP25

HA tagBamHIKpnINotI

EcoRI

pVP16~ 7.5 kb

AmpR

VP16

Polycloningsite

NLS

Perform PCR with “OMP25 PDZ Forward” and “OMP25

PDZ Reverse” primers

PCR product

Digestion of PCR product and pVP16 with BamHI and NotI

Agarose Gel

vector

PCR product

Run digested vector and PCR product on Agarose gel

Extract DNA from gel and ligate pVP16 vector and PCR product overnight

pVP16~ 8 kb

AmpR

VP16

NLS

OMP25 PCR fragment

Transform DNA into XLI-Blue cells; select for

Ampicillin-resistant colonies

Figure 2.3. Insertion of amino acids 37-177 of rat OMP25 into pVP16.Primers were designed around nucleotides encoding amino acids 37-177 of rat OMP25 (see Fig 2.2). PCR was performed to amplify this region of OMP25 and the PCR product and pVP16 were digested with BamHI and NotI restriction endonucleases. The digested products were separated on an agarose gel, and the vector and PCR products were excised from the gel. The DNA was extracted using a QIAGEN Gel Extraction Kit. The vector and insert DNA was then incubated together overnight at 16°C, in the presence of T4 DNA Ligase. The following day, the ligation mixture was transformed into competent XLI-Blue cells. Ampicillin-resistant colonies were selected and sequenced to confirm the insertion of the OMP25 fragment into the pVP16 vector.

43c

Page 81: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

The various methods used during these subcloning procedures are described individually

in Sections 2.3.30, 2.3.31 and 2.3.32.

2.3.30 Restriction enzyme digest

Analytical restriction enzyme digests were carried out for 1 -3 hours at

37°C. For a typical restriction digest, approximately 1 µg of DNA template was

incubated with approximately 5 U1 of the required restriction endonuclease (Promega),

with the appropriate buffer and 2µg acetylated BSA, made up to a final volume of 20 µl

in ddH2O.

If restriction digest products were to be used for subsequent ligation

reactions, the digests were typically scaled up to a final volume of 50 µl containing 4 µg

of DNA template, 10 U of the required restriction enzyme (Promega), with the

appropriate buffer and 5 µg acetylated BSA. The digested DNA was then separated

from other reaction components with the use of PCR purification columns (QIAGEN),

according to the manufacturer’s instructions.

2.3.31 Polymerase Chain Reaction (PCR)

A typical PCR reaction consisted of 50 ng plasmid DNA as a template, 0.5

µM forward primer, 0.5 µM reverse primer, 0.2 mM dNTP mixture, 1.5 mM MgCl2 and

2.5 U2 Platinum® Taq Polymerase, made up to a final volume of 50 µl in PCR Buffer

(50 mM KCl, 20 mM Tris-HCl pH 8.4). PCR reaction mixtures in sterile 0.2 ml tubes

were overlaid with PCR wax to prevent evaporation of the reaction components during

the PCR reaction. PCR reactions were carried out using a MiniCycler™ (MJ Research,

California, USA). Typical conditions for a PCR reaction were:

1 One unit of restriction endonuclease activity is defined as the amount of enzyme required to completely digest 1 µg of substrate DNA in one hour at 37°C. 2 One unit of Platinum® Taq Polymerase incorporates 10 nmol of deoxyribonucleotide into acid-precipitable material in 30 min at 74°C.

44

Page 82: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

94°C 4 min

94°C 1 min

30 x 40°C - 60°C 1 min (annealing temperature varied according to

the primers used – see Appendix I)

72°C 1 min

72°C 10 min

4°C hold

Colony screen PCR was used to identify bacterial colonies from cloning and yeast

colonies from yeast two-hybrid analysis (see Section 2.3.35) that contained the plasmid

insert of interest. In this procedure, a normal PCR reaction mix was assembled, with a

scaled-down final volume of 12 µl. A small amount of colony was then added to the

tube as the template for the PCR reaction. Normally, a gene-specific primer was used in

combination with a vector-specific primer to amplify the insert. This increased the

specificity of the reaction.

2.3.32 Reverse Transcription PCR

Reverse transcription PCR (RT-PCR) was used to amplify SAPK3 mRNA

from heart and skeletal muscle samples. The Access RT-PCR system (Promega) was

used for this protocol. The reaction consisted of 1 µg total RNA as a template, 1 µM

forward primer, 1 µM reverse primer, 0.2 mM dNTP mixture, 1 mM MgCl2, 5 U3 AMV

Reverse Transcriptase and 5 U Tfl DNA Polymerase, made up to a final volume of 50 µl

in AMV/Tfl reaction buffer (components unspecified by manufacturer). PCR reactions

were assembled in sterile 0.2 ml tubes using nuclease-free water as a diluent.

Conditions for the RT-PCR reaction were:

3 1 unit of AMV reverse transcriptase is defined as the amount of enzyme required to catalyse the incorporation of 1nmole of dTTP into acid insoluble form in 10 minutes at 37°C using poly(A)-oligo(dT) as a template.

45

Page 83: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

48°C 45 min

94°C 2 min

94°C 30 sec

40 x 60°C 1 min

72°C 2 min

72°C 7 min

4°C hold

2.3.33 DNA ligation

Ligations were prepared using 100 ng of vector DNA. The amount of

insert DNA added was then varied to give an insert:vector concentration ratio of 3:1, 5:1

and 10:1. T4 DNA Ligase (1.5 units4) and T4 DNA Ligase Buffer (50 mM Tris-HCl pH

7.5, 10 mM MgCl2, 10 mM DTT, 1 mM ATP, 25 µg/ml BSA) were then added to the

reaction, and the final volume made to 10 µl with ddH2O. The reactions were then

placed in a thermocycler at 16°C overnight.

2.3.34 Site-directed mutagenesis

Primers were designed to incorporate the desired mutation and covered

approximately 15-20 bases on either side of the mutated area (see Appendix I for primer

sequences). Site-directed mutagenesis was carried out using the Stratagene

QuikChange® Site-Directed Mutagenesis Kit according to the manufacturer’s

instructions. Briefly, a PCR reaction was set up to contain 50 ng DNA template, 125 ng

forward primer, 125 ng reverse primer, 200 µM dNTP mix and 3 U5 Pfu DNA

polymerase in Pfu Buffer (20 mM Tris-HCl pH 8.8, 10 mM KCl, 10 mM (NH4)2SO4, 2

mM MgSO4, 0.1% (v/v) Triton X-100 and 0.1 mg/ml nuclease-free BSA).

This reaction mixture was then overlaid with PCR wax and subjected to the

4 0.01 unit (Weiss unit) of T4 DNA ligase is defined as the amount of enzyme required to catalyse the ligation of greater than 95% of the Hind III fragments of 1 µg of Lambda DNA at 16°C in 20 min.

46

Page 84: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

following conditions:

95°C 30 sec

95°C 30 sec

12-18 times6 55°C 1 min

68°C 2 min/kb of plasmid length

Following the reaction, the PCR wax was removed. The reactions were

then placed on ice to cool for at least 5 min, and 1 µl of DpnI restriction enzyme added.

The reaction mixture was then mixed and placed in a 37°C waterbath for 1 hour. The

purpose of this incubation time was to allow the DpnI enzyme to digest the parental

(non-mutated) DNA. Since plasmid DNA isolated from most E.coli strains is dam

methylated, it is susceptible to DpnI digestion, which is specific for methylated and

hemimethylated DNA. Following this incubation, 5 µl of the mixture was transformed

into XL1-Blue competent cells (see section 2.3.22).

During the course of this project, site-directed mutagenesis was used to

introduce mutations into SAPK3, Sab and OMP25. The exact mutations made are listed

in Table 2.5.

2.3.35 Yeast two-hybrid screening

To isolate binding partners of SAPK3, yeast two-hybrid screening was

carried out. This involved cloning the full-length rat SAPK3 protein into the LexA

fusion vector pBTM116 (Figure 2.1), and screening a pVP16 cDNA library made from

mRNA prepared from the lymphohemopoietic progenitor cell line, EML Clone.1. The

pBTM116 vector contains the TRP1 gene and therefore yeast cells containing this

plasmid grew in the absence of the amino acid tryptophan. The pVP16 vector contains

5One unit of Pfu DNA Polymerase incorporates 10 nmol of deoxyribonucleotide into acid-precipitable material in 30 min at 75°C. 6The number of cycles required, according to Stratagene is: nucleotide point mutations, 12 cycles; single amino acid changes, 16 cycles; multiple amino acid deletions or insertions, 18 cycles.

47

Page 85: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

the LEU2 gene and therefore yeast cells containing this plasmid grew in the absence of

leucine.

The general principles underlying yeast two-hybrid screening are outlined

in Figure 6.2. Briefly, the Saccharomyces cerevisiae L40 strain (MATα, his3∆200, trp1-

901, leu2-3, 112, ade2, LYS2::(lexAop)4-HIS3, URA3::(lexAop)8-lacZ, GAL4) was co-

transformed with LexA-SAPK3 and the pVP16 cDNA library. If an interaction occurred

between SAPK3 and a protein present within the cDNA library, the DNA binding

domain of the LexA protein and the transactivation domain of the VP16 protein would

be brought into close proximity to reconstitute an active transcription factor. Therefore,

the reporter genes HIS3 and lacZ would be activated, resulting in the growth of yeast in

the absence of the amino acid histidine, and the changing of yeast colonies to a blue

colour when 5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside (X-gal) was added

(Section 2.3.37, 2.3.38).

To determine whether these interacting proteins were “true positives”,

genomic and plasmid DNA was isolated from L40 yeast cells (Section 2.3.40), and

introduced into competent KC8 bacterial cells (2.3.41, 2.3.42). pVP16 DNA was then

selectively isolated from the bacterial cells (Section 2.3.43) and re-introduced into L40

yeast cells (2.3.44), where the library proteins were again tested for their ability to

interact with pBTM116-SAPK3, alongside other proteins, sub-cloned into pBTM116 to

act as negative controls (see Chapter 6). If the expressed library proteins were again

shown to be positive for interaction with SAPK3, the DNA encoding these proteins was

sequenced (Section 2.3.27) and these matched with sequences on the National Center for

Biotechnology Information (NCBI) database.

2.3.36 Preparation of selective media for yeast two-hybrid screening

To select yeast transformed with either pBTM116 or pVP16 or both

plasmids, or to select for an interaction between pBTM116-SAPK3 and proteins cloned

into pVP16, selective media was required. Selective media was made up in filter-

48

Page 86: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

sterilised Synthetic Defined (SD) media (0.17% (w/v) yeast nitrogen base, 4 mM

ammonium sulfate, 100 mM D-glucose) with the appropriate antibiotics omitted. For

example, if yeast were to be selected containing the pBTM116 plasmid (which contains

the TRP1 gene), tryptophan was omitted from the selective media. If selection was to be

carried out using a plating method rather than in liquid media, agar was used at a

concentration of 15 g/L of media. Amino acids were made up at the following

concentrations (to produce 50x concentrated stock solutions): adenine, 2 mg/ml;

histidine, 1 mg/ml; leucine, 3 mg/ml; tryptophan 2 mg/ml. These amino acids were then

used in selective media at 1 x concentration as required.

2.3.37 Large-scale yeast transformation

A glycerol stock of the S. cerevisiae L40 strain, previously transformed

with pBTM116 containing full-length SAPK3 (see Section 2.3.44), was streaked on to a

Trp- selective plate and allowed to grow for 2 days at 30°C. A yeast colony was then

selected from this plate and grown overnight in 200 ml Trp- selective media, at 30°C,

180 rpm. The following morning, the overnight culture was added to 1 L Yeast extract-

peptone-dextrose medium + adenine (YPAD) (1% (w/v) yeast extract, 2% (w/v)

peptone, 0.1% (w/v) adenine, 100 mM D-glucose) and grown for approximately 3.5

hours to reach an optical density of OD600 = 0.6 - 0.8. The cells were then collected by

centrifugation (1050 x g, 5 min, RT). Immediately after centrifugation, the supernatant

was decanted and the pellet resuspended in 500 ml sterile ddH2O. The cells were then

centrifuged (1050 x g, 5 min, RT) and the pellet resuspended in 20 ml Tris-EDTA

(TE)/lithium acetate (LiAc) solution (50 mM Tris, 5 mM EDTA, 100 mM LiAc pH 7.5).

Denatured, sheared salmon sperm DNA (10 mg) and pVP16 cDNA library

made from the lymphohemopoietic progenitor cell line, EML Clone.1 (500 µg) were

mixed together and added to the resuspended cells. After thorough mixing, 140 ml

Polyethylene Glycol (PEG)/TE/LiAc mixture (40% (w/v) PEG-4000, 100 mM Tris, 10

mM EDTA, 100 mM LiAc pH 7.5) was added and incubated at 30°C, 90 rpm for 30

min. Following this incubation, 18 ml DMSO was added and the flask swirled to mix.

49

Page 87: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

The flask was then placed in a 42°C waterbath for 6 min, to heat shock the yeast cells

and allow uptake of the cDNA library. This flask was swirled every 20 sec to allow heat

transfer. The flask contents were then transferred to a sterile 1 L Schott bottle

containing 400 ml Yeast extract-peptone medium + adenine (YPA) (YPAD without

glucose) at RT. This was mixed and transferred to centrifuge tubes, then centrifuged

(1050 x g, 5 min, RT). The supernatant was removed immediately and the cells

resuspended in 500 ml YPA. The cells were then re-centrifuged as before and the

supernatant removed immediately. The pellets were then resuspended in 1 L YPAD,

pre-warmed to 30°C, and incubated at 30°C, 100 rpm, for 1 hour to allow recovery of

the cells.

After this incubation, cells were centrifuged (1050 x g, 5 min, RT) and the

pellet resuspended in 500 ml Trp- Leu- selective media. Cells were then re-centrifuged

(1050 x g, 5 min, RT), resuspended in 1 L Trp- Leu- selective media pre-warmed to

30°C, then incubated at 30°C, 100 rpm, for 4 hours. Cells were then centrifuged (1050 x

g, 5 min, RT), supernatant decanted, and pellets resuspended in 250 ml tryptophan-,

histidine-, uracil-, leucine-, and lysine-deficient (THULL-) media (40 µg/ml adenine in

SD media). This washing in THULL- media removed all traces of histidine that would

interfere with the subsequent selection of SAPK3-interacting partners by growth in the

absence of histidine (HIS3 reporter). Cells were then re-centrifuged as before (1050 x g,

5 min, RT) and resuspended in 250 ml THULL- media. After another centrifugation

step, the supernatant was decanted and the cells resuspended in 10 ml THULL- media.

To check transformation effieciency, 10 µl of this cell suspension was spread on a 90

mm T- L- selective plate. The remainder of the cell suspension was spread on to 20

THULL- agar plates (each 137 mm in diameter). All plates were then left at RT

overnight to dry, and placed in a 30°C incubator for 3 days to allow growth of the yeast

colonies.

50

Page 88: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.38 β-Galactosidase assay

Following the growth of transformed yeast colonies on THULL- (or Trp-

Leu- plates in the case of a small-scale transformation), yeast were re-streaked to a grid

on fresh THULL- (or Trp- Leu-) plates and left to grow at 30°C for 2 days. A

nitrocellulose membrane and Whatman 3MM filter paper was cut to fit the size of the

plate, and the nitrocellulose membrane was laid over the yeast colonies, ensuring that no

air bubbles appeared. The membrane was then lifted from the plate and allowed to dry

for approximately 5 min on the bench (colony side up). To fix colonies to the

membrane, the membrane was placed colony side up on a pre-cooled aluminium boat

floating in liquid nitrogen for 30 sec, then immersed in liquid nitrogen for 5 sec. The

membrane was then removed from the boat and placed at RT until thawed. Meanwhile,

a Petri dish was prepared for the reaction. In the lid of a Petri dish, 3 ml of X-gal

solution (0.27% (v/v) β-mercaptoethanol, 0.33 mg/ml X-gal in Z buffer (60 mM

Na2HPO4, 40 mM NaH2PO4, 10 mM KCl, 1 mM MgSO4, pH7.0)) was pipetted and the

circle of filter paper placed in this. The membrane was then laid over the filter paper,

colony side up, avoiding air bubbles. This was then covered with the bottom of the dish,

and placed at 30°C, for up to 4 hours. Strong interactions generally resulted in a colour

change within 30 min.

2.3.39 Preparation of glycerol stocks of yeast two-hybrid screening positive

colonies

Following testing of library interactors for lacZ reporter activation, glycerol

stocks of the positive colonies were made for extraction of plasmid DNA and re-

transformation into yeast cells. Glycerol stock mix (His- Trp- Leu- media containing

15% (v/v) glycerol) was pipetted into several 96-well plates (50 µl per well), and a

single colony was placed into each well. The lids of the plates were then replaced and

sealed tightly with Parafilm, before storing at -80°C.

51

Page 89: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.40 Isolation of DNA from the S. cerevisiae strain L40

Glycerol stocks of yeast colonies were streaked out on Leu- plates to select

for pVP16 plasmids, and left to grow at 30°C for 2 days. Liquid cultures (5 ml) in Leu-

selective media were then made from single colonies, and grown for 2 days at 30°C, 180

rpm. Cultures were then centrifuged (1050 x g, 5 min, RT) and the supernatants

decanted. The cell pellets were then resuspended in 200 µl Yeast Lysis Solution (2%

(v/v) Triton X-100, 1% (w/v) SDS, 100 mM NaCl, 10 mM EDTA, 10 mM Tris-HCl pH

8.0). Acid-washed glass beads (425-600 microns) were aliquoted into Eppendorf tubes

up to the 100 µl graduation, and the resuspended cells added. One volume of

phenol:chloroform (1:1) was added and each sample vortexed for 2 min. Samples were

then centrifuged (13,000 x g, 10 min, RT) and the supernatants transferred to clean

Eppendorf tubes. The plasmid DNA was then precipitated with the addition of 20 µl 3

M sodium acetate (pH 5.0), mixing, and adding 500 µl 100% ethanol with mixing. This

was then placed in a -20°C freezer for 20 min. The samples were then centrifuged

(13,000 x g, 20 min, RT) and the supernatant decanted. To wash the pellets, 500 µl 70%

ethanol was added and the tubes re-centrifuged (13,000 x g, 10 min, RT). The

supernatant was then decanted and the pellets allowed to air-dry for approximately 10

min. Pellets were then resuspended in 25 µl TE Buffer (100 mM Tris, 10 mM EDTA)

and stored at -20°C until use.

2.3.41 Preparation of electrocompetent KC8 cells

A glycerol stock of the E. coli strain KC8 was streaked on to a LB-agar

plate containing 30 µg/ml kanamycin, and grown overnight at 37°C. The following day,

colonies were added to LB media containing 30 µg/ml kanamycin and the cultures

grown at 37°C, 250 rpm, until an A600 of 0.6-0.8 was reached. The cells were chilled on

ice for 20 min, then centrifuged (3000 x g, 7 min, 2°C). The supernatant was decanted

and the pellets resuspended in 1 litre of sterile ice-cold ddH2O. The cells were then

centrifuged again (3500 x g, 10 min, 2°C). The pellets were then resuspended in 500 ml

of sterile ice-cold ddH2O and re-centrifuged (4000 x g, 10 min, 2°C). The supernatant

52

Page 90: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

was decanted and the pellet resuspended in 20ml of ice-cold 10% (v/v) glycerol in

ddH2O. This was then centrifuged (2300 x g, 20 min, 2°C). The supernatant was

removed and the pellet was resuspended in 1 ml of ice-cold 10% (v/v) glycerol in

ddH2O. The cells were then electroporated or stored at -80 °C for later use.

2.3.42 Bacterial electroporation

Freshly prepared electrocompetent cells, or frozen electrocompetent cells

thawed on ice were used for electroporation. Approximately 100 ng (2 µl) of DNA

isolated from yeast (Section 2.3.40) was mixed with 40 µl of competent KC8 cells in a

cold 1.5 ml Eppendorf tube and kept on ice for 1 – 10 min. This mixture was then

transferred to a cold 0.2 cm electroporation cuvette (Bio-Rad), and the cuvette tapped to

ensure the mixture evenly covered the base. The Bio-Rad Gene Pulser was set at 25 µF,

2.5 kV and the pulse controller set at 200 Ω. The cuvette was placed into a safety

chamber slide and the slide pushed into the chamber of the Gene Pulser until the cuvette

was seated between the contacts at the base of the chamber. One pulse was then

delivered, resulting in a time constant of approximately 4.5 msec. Immediately after

pulse delivery, 1 ml of sterile SOC Buffer (2% (w/v) Bacto-tryptone, 0.5% (w/v) Yeast

extract, 8.6 mM NaCl, 2.5 mM KCl, 10 mM MgCl2; pH 7.0) was added and the cells

resuspended. The resuspended cells were then transferred to 1.5 ml Eppendorf tubes and

incubated at 37°C for 1 hour. After this incubation time, cells were centrifuged (13,000

x g, 1 min, RT) and the supernatant removed. The cell pellet was then resuspended in

100 µl M9 salts solution (22 mM KH2PO4, 48 mM Na2HPO4, 8.6 mM NaCl2, 18.6 mM

NH4Cl; pH 7.2), and centrifuged again (13,000 x g, 1 min, RT). The supernatant was

decanted and the pellet resuspended in 100 µl M9 salts solution, before being spread on

to Minimal Media plates (20% (v/v) M9 salts solution, 2 mM MgSO4, 20 mM D-

glucose, 100 µM CaCl2, 40 µg/ml L-tryptophan, 20 µg/ml L-histidine, 100 µg/ml

ampicillin, 30 µg/ml kanamycin, 1 mM thiamine-HCl, 1.5% (w/v) agar). Plates were

incubated for 2 days at 37°C to allow growth of bacterial colonies.

53

Page 91: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.43 Isolation of plasmid DNA from KC8 bacterial cells

Following the growth of KC8 bacterial colonies on Minimal Media plates,

single colonies were picked, inoculated into 2.5 ml Minimal Media, and grown

overnight (37°C, 200 rpm). The following morning, minipreps were performed to

isolate plasmid DNA using the QIAGEN miniprep kit, according to the manufacturer’s

instructions.

2.3.44 Small-scale yeast transformation

A glycerol stock of the S. cerevisiae L40 strain was streaked on to a

YPAD-agar plate (YPAD containing 1.5% (w/v) agar) and left to grow overnight at

30°C. The next day, 40 ml of YPAD media was inoculated with a single colony of L40

yeast and grown overnight at 30°C, 180 rpm.

The following day, the culture was centrifuged (670 x g, 5 min, RT) and

the pellet resuspended in 40 ml ddH2O. The resuspended cells were then re-centrifuged

(670 x g, 5 min, RT) and the pellet resuspended in 5 ml of LiAc/TE solution. The cells

were then incubated at RT for 10 min. Meanwhile, DNA to be transformed into yeast

cells was aliquoted into 1.5 ml Eppendorf tubes. This consisted of 1 µl of pBTM116

plasmid DNA, containing the insert of interest, 1 µl of pVP16 plasmid DNA containing

the insert of interest, and 10 µl (100 µg) of denatured, sheared salmon sperm DNA. The

DNA was mixed together thoroughly before addition of 100 µl of the yeast cell mix.

After thorough mixing, 700 µl LiAc/PEG/TE mixture was added, the contents

thoroughly mixed, and the tubes incubated at 30°C for 30 min. Following this

incubation time, 88 µl DMSO was added to each tube, mixed, and incubated at 42°C for

7 min. Tubes were then centrifuged at 13,000 x g for 10 sec and the supernatants

removed. The pellets were then resuspended in 100 µl TE Buffer and spread on to

plates containing the required selective media. Plates were left to dry at RT for

approximately 1 hour before being placed in a 30°C incubator for 3 days to allow growth

of transformed yeast colonies.

54

Page 92: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

2.3.45 Screening of a cDNA library with the SAPK3 antibody

A λ phage heart cDNA library was screened with the SAPK3 antibody to

isolate SAPK3 clones and confirm both the specificity of SAPK3 antibody specificity

and the cardiac expression of SAPK3. This method involved plating out a λ phage heart

cDNA library on a bacterial (Y1090r-) lawn and allowing the phage to lyse the bacterial

cells. Plates were then overlaid with IPTG-impregnated nitrocellulose filters to induce

protein expression of the library. Filters were then removed from the plates, blocked,

and subjected to immunoblotting with the SAPK3 antibody. Positive plaques were then

identified by a colourimetric detection system, isolated and sequenced.

2.3.46 Initial preparation for the library screening procedure

At least two days prior to screening, LB agar plates containing MgSO4 (LB

medium, 10 mM MgSO4, 15g/L agar; pH 7.0; autoclaved) were poured and stored at

4°C. Top agarose (LB medium, 10 mM MgSO4, 7.2 g/L agarose; pH 7.0; autoclaved)

was also made and stored at 4°C. Two days before screening, the E.coli host strain

Y1090r- was streaked on to an LB agar plate containing 50 µg/ml ampicillin and left to

grow overnight at 37 °C. The following day, 50 ml of LB medium was inoculated with

a single colony isolated from this plate and left to grow overnight (37 °C, 200 rpm).

2.3.47 Establishing the library titre

Before the commencement of library screening, it was necessary to titre the

library to confirm the number of plaques formed per ml of library. This would then

allow for the control of the number of plaques per plate in the screening procedure.

Therefore, serial dilutions of the library were made in Sterile Media (SM) (100 mM

NaCl, 8 mM MgSO4, 0.1 % (w/v) gelatin, 50 mM Tris-HCl, pH 8.0; autoclaved) and

mixed with the host bacterial strain and top agarose, as described in Section 2.3.44.

These were left to grow for 3 hours at 37 °C and then transferred to 42 °C for

55

Page 93: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

approximately 5 hours. The resulting plaques were then counted to obtain the plaques

formed per ml of library. From this, the amount of stock phage required to give

approximately 15,000 plaques per plate for primary library screening was calculated (a

dilution of 1 in 105).

2.3.48 Screening procedure

An aliquot of the Rat Heart 5'-STRETCH PLUS cDNA Library (Clontech)

was diluted in SM (1 in 105) and stored at 4°C. Diluted phage (300 µl) were mixed with

600 µl of the overnight bacterial culture and incubated for 15 min at 37°C. Top agarose

warmed to 45°C was added to a final volume of 10 ml and this entire mixture quickly

poured over a warm agar plate (150 mm) on a level platform. After the top agarose had

solidified, plates were then left inverted at 42°C to allow bacterial growth and lysis by

phage for approximately three hours. During this period, nitrocellulose filters were

soaked in sterile 10 mM IPTG in ddH2O and dried in a sterile laminar flow hood. The

plates were then overlaid with these dried, IPTG-impregnated filters to induce protein

expression. Reference points were marked on the filters by piercing through the filters

and agar with a sharp instrument. The plates were then incubated for approximately 4

hours at 37 °C (until plaques were approximately 1 mm in diameter) to induce protein

expression. The filters were removed from the LB agar plates and incubated with 10 ml

Blocking Solution (5% (w/v) skim milk powder in TBST; RT), then incubated overnight

at 4°C with the SAPK3 antibody diluted in blocking solution.

The following day, the filters were washed in TBST (3 x 5 min; RT), and

then incubated with anti-rat antibody linked to horseradish peroxidase. The filters were

then washed again in TBST (3 x 5 min; 1 x 10 min; RT). Positive plaques were detected

with 2.8 mM 4-chloro-1-napthol in 10 mM imidazole buffer (pH 8.1) containing 17%

methanol and 3.7 mM hydrogen peroxide for approximately 30 min (RT). The reaction

was stopped with 3 x 5 min washes in ddH2O.

56

Page 94: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Well-isolated homogeneously-positive colonies for sequencing required a

2-3 further screening rounds. Therefore, filters were aligned with their corresponding

agar plates, and tentatively positive colonies were excised from the agar and placed into

1 ml SM containing 20 µl chloroform. These were then left at 4 °C overnight to allow

lysis of bacterial cells. The diluted phage were then titered as described in Section

2.3.47 to determine the volume required to give approximately 100-200 plaques on a 90

mm petri dish. This low plaque density was to enable the plaques to grow to a larger

size than in the primary screen, giving stronger signals and permitting individual

positives to be more easily identified and selected. Following titering, the diluted phage

were screened as described for primary screening, with the exception that plates were

incubated overnight following induction of protein expression with IPTG-impregnated

filters, to allow increased growth of plaques.

57

Page 95: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Chapter 3

Evaluation of the expression and localisation of SAPK3 in cell culture models

Page 96: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

3.1 Introduction

The activation of protein kinase signalling pathways such as the MAPK

pathways has been implicated in a wide variety of diseases, including neurodegenerative

diseases, arthritis, muscular dystrophy, cancer and cardiovascular disease (Hunot et al.,

2004; Anguita et al., 2002; Nakamura et al., 2001; Kolodziejczyk et al., 2001;

MacDonald et al., 2001; Cook et al., 1999). For this reason, protein kinases are now

considered by some researchers to be the second most important group of drug targets,

after the G-protein-coupled receptors (Cohen, 2002; Vlahos et al., 2003).

Cardiovascular disease is the leading cause of death in Australia,

accounting for 39% of all deaths in 2000 (National Heart Foundation of Australia). This

incidence is similar in other developed countries and is likely to approach these levels in

developing countries as they adopt more sedentary lifestyles, a higher fat intake,

relatively lower intakes of plant foods, and lower levels of everyday physical activity

(Yusuf, 1998; Walker et al., 2004). The molecular mechanisms responsible for the

development of cardiovascular disease are poorly understood, but are thought to involve

intracellular signalling pathways (Chien, 1999). Many MAPK family members are

expressed in the heart (Kim et al., 1998), and their expression as well as their activation

state has been shown to change both during normal heart development and during heart

disease, implicating them for a role in these processes (Corson et al., 2003; Cook et al.,

1999; Lemke et al., 2001).

It was first proposed that the ERK MAPKs could mediate the development

of cardiac hypertrophy (Bogoyevitch et al., 1993), the disease process in which the

cardiac myocytes undergo enlargement without cell division to adapt to a demand in

increased workload (Chien et al., 1991). This has been independently confirmed by

studies using selective ERK MAPK pathway inhibitors to prevent cardiac hypertrophy

(Yue et al., 2000), or by the cardiac-restricted overexpression of active MKK1 (the

upstream activator of ERK MAPKs) to cause concentric hypertrophy in vivo (Bueno et

al., 2000). ERK signalling may also play a cardioprotective role. For example, ERK2

58

Page 97: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

signalling protects the myocardium from ischemia-reperfusion injury in vivo (Lips et al.,

2004).

The role of JNK and p38 MAPK family members in cardiac tissue remains

controversial, with many studies supporting the role of these MAPK family members in

the progression to heart failure, and others in a cardioprotective function. For example,

the use of transgenic mouse models with targeted expression of active MKK7 in cardiac

myocytes led to heart failure, suggesting that activated JNK MAPKs facilitate the

processes leading to heart failure (Petrich et al., 2004). Conversely, another study has

shown that the MEKK1-JNK pathway plays a protective role in pressure overload in

vivo through the use of MEKK1 knockout mice (Sadoshima et al., 2002).

JNK and SAPK2a activation have been found to be increased in heart

samples from patients with heart failure secondary to ischemic heart disease (Cook et al.,

1999). A pro-apoptotic role for SAPK2a during ischemia is suggested by the protection

of cardiac myocytes from ischemic damage by the specific p38 isoform inhibitor,

SB203580 (Mackay and Mochly-Rosen, 1999). Indeed, a role for SAPK2a in stress

signalling in the heart is implicated by its activation in response to a variety of different

agonists in cardiac myocytes, including ischemia and reperfusion (Bogoyevitch et al.,

1996), cytokines (Clerk et al., 1999), hypertrophic agonists (Clerk and Sugden, 1999)

and mechanical stretch (Kudoh et al., 1998). However, despite the implication of

SAPK2a in cardiac cell death, this kinase has also been implicated in beneficial

signalling in the heart. For example, SAPK2a signalling has been implicated in

mediating ischemic preconditioning of the heart (Nakano et al., 2000).

Despite continued interest in the p38 pathway, no studies to date have

addressed the role of p38 isoforms other than SAPK2a and SAPK2b in the heart. The

role of the two p38 MAPK isoforms SAPK2a and SAPK2b has been described

following their overexpression in vitro (Wang et al., 1998a). Thus, SAPK2b mediated

cardiac hypertrophy but SAPK2a mediated apoptotic cell death when overexpressed in

cardiac myocytes (Wang et al., 1998a). This highlights that different members within a

59

Page 98: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

single SAPK family can play distinct roles in the heart. One study has examined the in

vivo effects of constitutive p38 MAPK family signalling in the heart. In this study,

transgenic mice with ventricular-specific expression of the constitutively active p38

MAPK upstream activators MKK3 and MKK6 showed differential effects on heart

chamber remodelling and premature death (Liao et al., 2001). In contrast to early

studies which showed that SAPK2b mRNA was expressed in the heart (Stein et al.,

1997; Jiang et al., 1996), SAPK2b protein was not detected in these hearts, suggesting

that the differences in cardiac phenotypes could be attributed to differential activation of

other p38 isoforms (Liao et al., 2001). The absence of SAPK2b mRNA and protein in

the heart is supported by Lemke et al., who reported that SAPK2a is the predominant

isoform expressed in human heart, with lower levels of SAPK3 and SAPK4 detected

(Lemke et al., 2001). Thus, it remains possible that other members of the p38 MAPK

family could play a role in these signalling events in the heart.

SAPK3, a distinct p38 MAPK isoform, has been shown to be expressed (at

the mRNA level) in a variety of tissues, with the highest levels of expression in skeletal

muscle (Mertens S et al., 1996). SAPK3 is also phosphorylated and activated by the

upstream activators, MKK3 and MKK6 (Enslen et al., 1998; Cuenda et al., 1997). The

identification of binding of SAPK3 to the α1-syntrophin PDZ domain suggests that

SAPK3 may have distinct subcellular localisation in muscle (Hasegawa et al., 1999).

SAPK3 and other p38 isoforms are therefore unlikely to be functionally redundant.

The work in this Chapter was undertaken to determine the expression and

localisation of SAPK3 in the human embryonic kidney (HEK) 293 cell line and primary

cultures of isolated neonatal cardiac myocytes. To perform these analyses, a specific

SAPK3 antibody was required. As described in Chapter 1, the first aim of this project

was to characterise the use of a specific SAPK3 monoclonal antibody for detecting

endogenous SAPK3. In this Chapter, HEK293 fibroblasts were used for initial

characterisation of this antibody due to the ease of culturing and transfecting these cells.

Once the expression and localisation of SAPK3 could be determined in the HEK293

60

Page 99: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

fibroblast models, this would then allow similar analyses to be performed on the

neonatal cardiac myocytes.

3.2 Results

3.2.1 A specific anti-SAPK3 monoclonal antibody suitable for immunoblotting and

immunolocalisation

Previous studies have reported the expression of SAPK3 mRNA in a

variety of tissues, with particularly high levels in skeletal muscle (Mertens et al., 1996;

Lechner et al., 1996; Li et al., 1996). The characteristics of exogenously overexpressed

SAPK3 (as introduced by transient transfection) have been studied at the protein level

(Lechner et al., 1996; Cuenda et al., 1997), however at the commencement of these

studies the expression and activation of endogenous SAPK3 protein was poorly

characterised with only a limited number of studies being performed in immortalised cell

lines (Conrad et al., 1999; Wang et al., 2000) and only one study in skeletal muscle

(Boppart et al., 2000). To begin to characterise this kinase, a monoclonal antibody

against full-length recombinant SAPK3 was used. This had previously been generated

by Jacky Cordell at the Institute for Cancer Research Hybridoma Unit (London, UK).

The specificity of the SAPK3 antibody had been previously confirmed in

overexpression systems that allowed inclusion of appropriate controls not possible when

examining endogenously-expressed proteins. Because these results have been presented

previously in a Bachelor of Science Honours thesis they are not presented here. Instead,

the reader is referred to Appendix II of this thesis (Court et al., 2002). Briefly, in those

studies HEK293 fibroblasts were transiently transfected with a myc epitope-tagged

SAPK3 construct. SAPK3 expression was then confirmed by immunoblotting with a

myc epitope tag antibody and the SAPK3 antibody. Both detected a protein of ~45 kDa

in cells transfected with > 0.1 µg SAPK3 plasmid DNA (Appendix II, Figure 1).

Allowing for the size of the myc epitope tag, this agreed with the reported size of murine

SAPK3 (42 kDa) (Mertens et al., 1996) which is larger than SAPK2a (41 kDa) (Han et

61

Page 100: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

al., 1994). To determine whether the SAPK3 antibody was capable of detecting

endogenous levels of SAPK3, HEK293 cells were seeded into dishes, lysed, and

subjected to Western blotting using the SAPK3 antibody. As seen in Figure 3.1A, the

SAPK3 antibody detected a single band of approximately 44 kDa in size.

MAPKs and SAPKs have been suggested to play important roles in the

heart, being implicated in the growth, development, differentiation, disease and death of

this tissue (Braz et al., 2002; Ueyama et al., 2000; Corson et al., 2003; Regan et al.,

2002; Davidson and Morange, 2000; Liao et al., 2001; Cook et al., 1999; Kaiser et al.,

2004; Aoki et al., 2002). Due to the implication of MAPKs and SAPKs in these

processes in cardiac tissue, it was of interest to determine whether SAPK3 was detected

in the heart. Primary cultures of neonatal rat cardiac myocytes have previously been

used to show expression of MAPK family members and their activation in response to

specific stimuli (Bogoyevitch et al., 1993; Bogoyevitch et al., 1994). Therefore, cultured

neonatal rat cardiac myocytes were lysed and subjected to Western blotting using the

SAPK3 antibody. The SAPK3 antibody detected a single band at approximately 42 kDa

in size (Fig 3.1B). Two fainter bands were also detected. Thus, the SAPK3 antibody

appeared to be capable of recognising both overexpressed (Appendix II) and endogenous

(Figure 3.1) SAPK3.

3.2.2 SAPK3 shows a punctate localisation in HEK293 fibroblasts and cardiac

myocytes

Previous work has shown that SAPK3 localises throughout the cell

cytoplasm and is largely excluded from the nucleus when overexpressed in Cos and

HEK293 fibroblasts (N. Court, BSc Hons). Again, the reader is referred to Appendix II.

However, at the time of commencing these studies, the localisation of endogenous

SAPK3 had not been reported in any tissue or cell type. Having confirmed the

specificity of the SAPK3 antibody by detecting overexpressed SAPK3 by Western

blotting and staining, and endogenous SAPK3 by Western blotting, this antibody was

used to examine the localisation of endogenous SAPK3 in cultured HEK293 fibroblasts

62

Page 101: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Aα-SAPK3

45 kDa

30 kDa

66 kDa97 kDa

220 kDa

20.1 kDa

SAPK3

Bα-SAPK3

45 kDa

30 kDa

66 kDa97 kDa

220 kDa

20.1 kDa

SAPK3

Figure 3.1: SAPK3 can be detected in HEK293 and neonatal rat cardiac myocyte cell lysates.(A) HEK293 fibroblasts were seeded into dishes and cultured for 40 hours before lysis. Cell lysates (65 µg) were then subjected to Western blotting with the SAPK3 monoclonal antibody. (B) Neonatal rat cardiac myocytes were isolated from 1-day old Sprague-Dawley rats and seeded into dishes. Cells were cultured for 40 hours before lysis. Cell lysates (40 µg) were then subjected to Western blotting with the SAPK3 monoclonal antibody. Open arrows indicate lower molecular weight bands that are possible degradation products of the SAPK3 protein. The slight decrease in apparent molecular weight of rat SAPK3 compared to human (HEK293 fibroblasts) SAPK3 was confirmed when comparing the expression of SAPK3 in tissue samples from the two species (Chapter 4).

62a

Page 102: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

(Fig 3.2). Staining of HEK293 fibroblasts using this antibody showed SAPK3 to have a

punctate, cytoplasmic distribution (Fig 3.2A). This staining was reduced to control

levels (where the primary antibody was omitted; Fig 3.2C) when the SAPK3 antibody

had been previously incubated with a 100-fold excess (by weight) of GST-SAPK3 (Fig

3.2B).

Having determined that the SAPK3 antibody was appropriate for

immunostaining of HEK293 fibroblasts, this antibody was then used to determine the

localisation of SAPK3 in cultured cardiac myocytes. The localisation of SAPK3 in

cultured cardiac myocytes was compared to that of SAPK2a/b using the SAPK3

antibody and a commercial antibody which was raised against the human SAPK2a

sequence (Fig 3.3). SAPK2a and SAPK2b share 74% sequence identity (Jiang et al.,

1996) and therefore the commercial SAPK2a antibody recognises both proteins.

However, previous studies have shown that SAPK2b was not expressed in the heart at

the mRNA or protein level (Lemke et al., 2001). The staining of cardiac myocytes using

this antibody therefore should be mainly, if not solely, attributed to the SAPK2a protein.

The localisation of SAPK2a has been reported previously, and shown to be

cytoplasmic and nuclear in 293T cells (Ben Levy et al., 1998; Lee et al., 2000).

Overlaying the images of SAPK3 (Fig 3.3A) and SAPK2a (Fig 3.3C) showed distinct

patterns of protein localisation (Fig 3.3E). SAPK2a was expressed throughout the cell

with high levels in the nucleus, as demonstrated when co-staining with Hoechst 33258

(Fig 3.3F). In contrast, SAPK3 was excluded from the nucleus and showed a punctate

distribution in the cell cytoplasm, suggesting organelle-specific localization (Fig 3.3A,

3.3F). This closely resembled the pattern seen in HEK293 fibroblasts (Figure 3.2) and

therefore was not likely the result of non-specific cross-reaction with proteins of smaller

molecular weight seen in Figure 3.1. No immunofluorescence was detected when

primary antibodies were omitted (Fig 3.3B, 3.3D).

63

Page 103: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

SAPK3

A

Pre-absorbed SAPK3

B

α-rat-Alexa 488 only(no SAPK3 antibody)

C

Figure 3.2: Endogenous SAPK3 shows a punctate cytoplasmic distribution in HEK293 fibroblasts.HEK293 cells were cultured for 40 hours, then fixed, permeabilised and stained with SAPK3 antibody (A) or SAPK3 antibody that had been incubated overnight with 100-fold excess (by weight) recombinant GST-SAPK3 protein (B). No staining was visualised when the primary antibody was omitted (C). Cells were also stained with Hoescht 33258 to visualise the nuclei (A, B, C). Results shown are representative of 2 independent experiments. Scale bar = 20 µm.

63a

Page 104: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

SAPK3

SAPK3 +

SAPK2a

SAPK2a

A B

DC

E F

α-rat-Alexa 488 only (no SAPK3 antibody)

α-rabbit-Alexa 546 only (no SAPK2a/b antibody)

SAPK3+

SAPK2a+

Hoechst 33258

Figure 3.3: Endogenous SAPK3 shows a punctate cytoplasmic distribution in neonatal rat cardiac myocytes.Neonatal rat cardiac myocytes on laminin-coated glass coverslips were cultured for 2 days, then fixed in 4% paraformaldehyde and stained with anti-SAPK3 antibody (A), SAPK2a/b antibody (C), or both antibodies together (E). No immunofluorescence was detected when the primary antibodies were omitted (B, D). Hoechst 33258 stain identified the nuclei (F). Results shown are representative of 3 independent experiments. Scale Bar = 36 µm.

63b

Page 105: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

3.2.3 Leptomycin B treatment confirms SAPK3 cytoplasmic localisation

Inactive SAPK2a is a nuclear protein complexed with one of its substrates,

MAPKAPK2 (Ben Levy et al., 1998). The results presented here support the nuclear

localisation of SAPK2a (Fig 3.3C; 3.3F), but show nuclear exclusion of endogenous

SAPK3 (Fig 3.3A; 3.3F). Therefore the inhibitor of nuclear export signal-dependent

transport, Leptomycin B, was used (Wolff et al., 1997). This approach has been used

previously to inhibit the MKK1/2 nuclear export signal-dependent shuttling of ERK

between the nucleus and cytoplasm (Adachi et al., 2000). This study showed that the

shuttling of ERK between the nucleus and cytoplasm was dependent on ERK interacting

with its upstream activators, MKK1/2 (Adachi et al., 2000).

SAPK3 localisation in cardiac myocytes was unaltered following treatment

with Leptomycin B (compare Fig 3.4A with Fig 3.4C). In contrast, more SAPK2a was

detected in the nucleus following Leptomycin B treatment (compare Fig 3.4E with Fig

3.4G). Controls showed no staining when primary antibodies were omitted (Fig 3.4B,

3.4D, 3.4F, 3.4H). This confirmed that inactive SAPK3 was cytoplasmic whereas

inactive SAPK2a was nuclear in agreement with earlier reports (Ben Levy et al., 1998).

3.2.4 SAPK3 co-localises with mitochondrial markers

SAPK3 staining appeared to show a punctate pattern in both HEK293

fibroblasts and cardiac myocytes. This punctate staining showed some similarities to

that expected for focal adhesion points in the cell (P. Rigby, personal communication).

Therefore, cardiac myocytes were stained with a specific antibody against the protein

Paxillin, which localises to focal adhesions (Turner et al., 1990). As seen in Figure 3.5,

SAPK3 staining did not co-localise with Paxillin. The SAPK3 staining appeared to

represent punctate structures (Fig 3.5A). These were further inside the cell, rather than

at the cell periphery where Paxillin was localised (Fig 3.5B).

64

Page 106: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Figure 3.4: SAPK3 has a punctate cytoplasmic localisation and this does not alter with Leptomycin B treatment of neonatal rat cardiac myocytes.Neonatal rat cardiac myocytes were pretreated with Leptomycin B (a nuclear export inhibitor) for 3.5 hours, then fixed in 4% paraformaldehyde and stained with antibodies specific for SAPK3 or a related family member, SAPK2a. When cells are treated with Leptomycin B, SAPK2a is retained in the nucleus, and therefore brighter staining is observed (G), compared to control (E). However, LeptomycinB treatment did not alter SAPK3 localisation (compare panels A and C), suggesting that it is primarily a cytoplasmic protein. Scale Bar = 40 µm.

α-SAPK3

α -SAPK3 + Leptomycin B

α-SAPK2a/2b+ Leptomycin B

α-rat Alexa 488(no SAPK3 antibody)

α-rabbit Alexa 546(no SAPK2a/b antibody)

α-rat Alexa 488(no SAPK3 antibody)

α-rabbit Alexa 546(no SAPK2a/b antibody)

A B

C D

E F

G H

α -SAPK2a/2b

64a

Page 107: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Overlay+ Hoescht 33258SAPK3 Paxillin

A CB

Hoescht 33258 +α-rat-Alexa 488 only(no SAPK3 antibody)

Hoescht 33258 +α-rabbit-Alexa 546 only(no Paxillin antibody)

ED

Figure 3.5: SAPK3 does not co-localise with Paxillin in cardiac myocytes.Cardiac myocytes were isolated from 1 day old Sprague-Dawley rats and maintained in culture for 2 days. Cells were then fixed, permeabilised and stained with anti-SAPK3 (A) or anti-Paxillin (B). Panel C shows an overlay of panels A and B. Green staining indicates position of SAPK3 in cells; red staining indicates Paxillin. Hoescht 33258 staining (blue) is also included to indicate the position of nuclei in the cells. No staining was visualised when the primary antibodies were omitted (Panels D, E). Scale Bar = 20 µm.

64b

Page 108: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

To determine whether SAPK3 staining represented co-localisation with the

mitochondria, cardiac myocytes and HEK293 fibroblasts were treated with the dye

Mitotracker Red that accumulates in actively respiring mitochondria, or the cells were

stained with an antibody against the mitochondrial protein HSP60. The cells were then

co-stained with the SAPK3 antibody. As shown in Figure 3.6, cells stained with the

SAPK3 antibody demonstrated a punctate pattern of staining (Fig 3.6A, D, G, and J),

and this staining pattern was also observed with Mitotracker (Fig 3.6B and H) or the

anti-HSP60 antibody (Fig 3.6E and K). Overlaying these images showed SAPK3

staining co-localising with Mitotracker or HSP60 staining in both cardiac myocytes and

HEK293 fibroblasts (Fig 3.6C, F, I and L).

3.2.5 SAPK3 overexpression does not appear to alter the morphology or survival of

cardiac myocytes

The overexpression of MAPKs and their upstream activators can induce

morphological changes in neonatal rat cardiac myocytes, indicating roles for these

proteins in cardiac hypertrophy and cell death (Ueyama et al., 2000; Wang et al., 1998a).

Thus, myc-tagged SAPK3 was overexpressed in neonatal rat cardiac myocytes to

examine its effects. Cells were also co-stained with phalloidin-TRITC to assess cell

morphology and the organization of actin filaments. In addition, Hoechst 33258 staining

was used to examine nuclei for indications of cell death. This stain has previously been

used to differentiate between nuclei from normal, necrotic and apoptotic cells (Chihab et

al., 1998). Figures 3.7 and 3.8 show cardiac myocytes overexpressing SAPK3 (Fig

3.7A, 3.7B, 3.8A, 3.8B). The pattern of actin filaments and size of these cells was

similar to non-transfected cells (Fig 3.7C, 3.7D, 3.8C, 3.8D), suggesting that no

morphological changes followed SAPK3 overexpression. Furthermore, the staining of

nuclei with Hoechst 33258 indicated that cells were not undergoing cell death, a

situation that would be evident by the appearance of brightly staining small nuclei (Fig

3.7E, 3.7F, 3.8E, 3.8F) (Chihab et al., 1998). Overlaying all three images shows SAPK3

overexpression in the cytoplasm and nucleus (Fig 3.7G, 3.7H, 3.8G, 3.8H), in contrast to

the endogenous protein which is expressed in the cytoplasm only (Fig 3.3A).

65

Page 109: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Mitotracker Overlay + Hoescht 33258SAPK3

A B C

SAPK3 Overlay + Hoescht 33258HSP60

D E F

Overlay + Hoescht 33258MitotrackerSAPK3

H IG

HSP60SAPK3 Overlay + Hoescht 33258

K LJ

Figure 3.6: SAPK3 co-localises with mitochondrial markers.Panels A-F: Cardiac myocytes were isolated from 1 day old Sprague-Dawley rats and maintained in culture for 2 days. Cells were then treated with Mitotracker Red for 40 min (panel B) or left untreated, then fixed, permeabilised and stained with anti-SAPK3 (A, D). Cells not treated with Mitotracker were then stained with anti-HSP60 (panel E). Panels C and F show the overlay of panels A and B, or D and E, respectively. Yellow staining represents where SAPK3 (green) and mitochondrial markers (red) co-localise. Hoescht 33258 staining (blue) is also included to indicate the position of nuclei in the cells. Scale Bar = 36 µm.Panels G-L: HEK 293 fibroblasts were seeded into dishes and cultured for 2 days. Cells were treated with Mitotracker Red for 40 min (panel H) or left untreated, then fixed, permeabilised and stained with anti-SAPK3 (G, J). Cells not treated with Mitotracker were then stained with anti-HSP60 (panel K). Panels I and L show the overlay of panels G and H, or J and K, respectively. Yellow staining represents where SAPK3 (green) and mitochondrial markers (red) co-localise. Hoescht 33258 staining (blue) is also included to indicate the position of nuclei in the cells. Scale Bar = 18 µm.

65a

Page 110: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

Hoechst33258

Phalloidin-TRITC

Myc

B

E F

DC

G H

Overlay

Figure 3.7: Overexpression of SAPK3 in cardiac myocytes does not appear to induce morphological changes or cell death.Cardiac myocytes were cultured for 18 hours, transfected with 0.4 µg plasmid encoding myc-SAPK3, then fixed 40 hours post-transfection. Cells were stained with anti-Myc antibody (A, B). Phalloidin-TRITC identified actin filaments (C, D) and was therefore used to examine cell morphology. Hoechst 33258 stain identified the nuclei (E, F), and this was used to evaluate whether transfected cells were undergoing apoptosis or necrosis. Overlaying images shows both nuclear and cytoplasmic localisation of overexpressed SAPK3 (G, H). Arrows indicate transfected cells. Results shown are representative of 2 independent experiments. Scale Bar = 40µm.

65b

Page 111: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A B

SAPK3

C D

Phalloidin-TRITC

FE

Hoechst33258

HG

Overlay

Figure 3.8: Overexpression of SAPK3 in cardiac myocytes does not appear to induce morphological changes or cell death.Cardiac myocytes were cultured for 18 hours, transfected with 0.4 µg plasmid encoding myc-SAPK3, then fixed 40 hours post-transfection. Cells were stained with anti-SAPK3 antibody (I, J). Phalloidin-TRITC identified actin filaments (K, L) and was therefore used to examine cell morphology. Hoechst 33258 stain identified the nuclei (M, N), and this was used to evaluate whether transfected cells were undergoing apoptosis or necrosis. Overlaying images shows both nuclear and cytoplasmic localisation of overexpressed SAPK3 (O, P). Arrows indicate transfected cells. Results shown are representative of 2 independent experiments. Scale Bar = 40µm.

65c

Page 112: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

3.2.6 SAPK3 phosphorylation and expression in response to agonist treatment

Primary cultures of neonatal rat cardiac myocytes serve as a model system

for studying hypertrophic changes in the pathological myocardium (van Bilsen and

Chien, 1993). Therefore, neonatal cardiac myocytes cells were treated with a variety of

different hypertrophic agonists and examined for changes in SAPK3 expression levels.

As seen in Figure 3.9, SAPK3 protein expression levels did not change with agonist

treatment. When these same samples were analysed for the presence of phospho-p38,

activation of SAPK2a could be detected with Interleukin-1, Leukemia Inhibitory Factor,

and Lysophosphatidic Acid treatment (Figure 3.10). However, no bands were seen at

the expected size for phospho-SAPK3 (Figure 3.10). Furthermore, when these cells

were treated with other agonists of the p38 MAPK pathways such as sorbitol, sodium

arsenite or anisomycin (Hoover et al., 2000; Wang and Proud, 1997; Barros et al., 1997)

for periods ranging from 5 to 60 min, no phospho-SAPK3 bands could be detected

(results not shown). These results suggest that SAPK3 is not phosphorylated and

therefore not activated in cardiac myocytes upon acute or chronic stimulation with the

agonists tested.

3.3 Discussion

Several groups have reported the expression of SAPK3 at the mRNA level

in a variety of tissues (Mertens et al., 1996; Lechner et al., 1996; Li et al., 1996).

However, the expression or activation of endogenous SAPK3, other than in

immortalised cell lines, has been poorly studied. The generation of a specific

monoclonal antibody against SAPK3 has allowed the expression and subcellular

localisation of endogenous SAPK3 to be evaluated in cultured cells, including HEK293

fibroblasts and neonatal rat cardiac myocytes.

The accessibility of a protein kinase to a substrate is one level of control in

the phosphorylation reactions that occur in each cell. Thus, the subcellular localisation of

66

Page 113: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

SAPK346 kDa

Vehicle Control ET-1 (100 nM) Iso(100µM) PE (100µM)Time (h): 24 48 72 24 48 72 24 48 72 24 48 72

Vehicle Control IL-1 (1ng/mL) LIF (10ng/mL) LPA (50 µM)Time (h): 24 48 72 24 48 72 24 48 72 24 48 72

46 kDaSAPK3

Figure 3.9: SAPK3 expression levels do not change in response toagonist treatment of cultured cardiac myocytes. Cardiac myocytes were isolated from 1 day old Sprague-Dawley rats and cultured for 24 hours. Cells were then serum-starved for a further 24 hours, then incubated with various hypertrophic agents as indicated for 24, 48 or 72 hours. Cell lysates were then immunoblotted with the monoclonal SAPK3 antibody. Arrows indicate the position of SAPK3. Results shown are representative of 2 independent experiments.Abbreviations: ET-1 = Endothelin-1; Iso = Isoproterenol; PE = Phenylephrine; IL-1 = Interleukin-1; LIF = Leukemia Inhibitory Factor; LPA = Lysophosphatidic acid.

66a

Page 114: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Phospho-SAPK2a

Vehicle Control IL-1 (1ng/mL) LIF (10ng/mL) LPA (50 µM)Time (h): 24 48 72 24 48 72 24 48 72 24 48 72

46 kDa

SAPK3SAPK2a

46 kDa

Figure 3.10: Agonist treatment stimulates phosphorylation of SAPK2a, but not SAPK3, in cultured cardiac myocytes. Cardiac myocytes were isolated from 1 day old Sprague-Dawley rats and cultured for 24 hours. Cells were then serum-starved for a further 24 hours, then incubated with various hypertrophic agents as indicated for 24, 48 or 72 hours. Cell lysates were then immunoblotted with the phospho-p38 MAPK antibody (top panel), or SAPK3 rat monoclonal antibody and SAPK2a rabbit polyclonal antibody in parallel (bottom panel). Results shown are representative of 2 independent experiments. Abbreviations: IL-1 = Interleukin-1; LIF = Leukemia Inhibitory Factor; LPA = Lysophosphatidic acid.

66b

Page 115: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

a protein kinase can provide useful information on the possible substrate proteins for

each specific kinase. For example, SAPK2a has been identified as a nuclear protein in

its inactive state, bound to its substrate, MAPKAPK2 (Ben Levy et al., 1998).

Following activation, SAPK2a phosphorylates MAPKAPK2, and this phosphorylation

masks a nuclear localization signal in MAPKAPK2, resulting in the export of both

proteins into the cytoplasm (Ben Levy et al., 1998). This mechanism would therefore

allow SAPK2a and MAPKAPK2 to phosphorylate cytoplasmic substrates. It was

investigated whether SAPK3 was localised to the nucleus of cardiac myocytes.

However, whilst SAPK2a was localised to the nucleus and cytoplasm of these cells,

SAPK3 was exclusively in the cell cytoplasm and showed a punctate distribution (Figure

3.3). This staining was shown to co-localise with mitochondrial markers (Figure 3.6).

The distinct intracellular localisation of SAPK3 supports a model in which this p38

MAPK isoform has the potential to play distinct roles in the heart.

The co-localisation of SAPK3 with the mitochondria suggests that its

substrates will be identified at this organelle. The lack of a mitochondrial targeting

sequence in the SAPK3 protein (unpublished observations), and the fact that only low

levels of non-ionic detergents are required to isolate it from cell lysates (Figure 3.1

lysates were prepared using 0.5% Triton X-100) suggests that it is localised on the outer

mitochondrial membrane rather than the inner mitochondrial membrane, intermembrane

space or mitochondrial matrix. Treatment of HEK293 fibroblasts with 0.5 M sorbitol for

time periods ranging from 5 min to 60 min did not result in SAPK3 translocation to

other areas of the cell (unpublished observations). This hyperosmotic stress has

previously been shown to activate overexpressed SAPK3 in HEK293 cells with 30 min

treatment (Cuenda et al., 1997); N. Court, BSc Hons thesis). This suggests that SAPK3

may be activated in response to a different subset of cellular stresses.

It cannot be ruled out that SAPK3 translocates to other areas of the cell in

response to other stimuli which remain to be tested. For example, stimuli resulting in

the sustained activation of SAPK3 may also cause its translocation to other

compartments of a cell. This would be similar to the situation observed for ERK when

67

Page 116: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

PC12 cells are treated with NGF, causing the sustained activation of ERK and its

translocation to the nucleus (Nguyen et al., 1993; Boglari et al., 1998). It should be

noted however, that unlike other MAPKs such as ERK, SAPK3 is monomeric in

structure (Bellon et al., 1999). It has been found that the homo-dimerisation of

phosphorylated ERK2 is essential for its translocation to the nucleus (Khokhlatchev et

al., 1998). Thus, it would appear unlikely that SAPK3 is subject to the same regulation

mechanisms that result in nuclear translocation of ERK MAPKs. At this stage, however,

other mechanisms cannot be ruled out.

When cells overexpressing SAPK3 were examined for any changes in

morphology or cell death, no changes were evident. However, further studies will be

required to determine whether SAPK3 when overexpressed and activated by appropriate

stimuli could exert other short- or long-term biological effects, such as metabolic

changes or alterations in gene expression patterns. For example, the activation of

overexpressed SAPK2a in CCL39 Chinese hamster fibroblasts has previously been

shown to significantly inhibit mitogen-induced cyclin D1 expression, whereas inhibition

of SAPK2a activity with SB203580 enhanced cyclin D1 transcription and protein level

(Lavoie et al., 1996). Both SAPK2a and SAPK3 were found to increase the

transcriptional activity of MEF2A in NIH 3T3 fibroblasts when they were overexpressed

in conjunction with MEF2A and MKK6 (Marinissen et al., 1999).

The use of transient transfection by cationic liposomes such as

Lipofectamine in cardiac myocytes shows approximately 1-2 % transfection efficiency

(unpublished observations). To undertake a more complete study of SAPK3

overexpression in these cells, adenoviral expression therefore should be used in future

work because with this approach over 90% of primary cell cultures of neonatal rat

mycocytes can be made to overexpress the protein of interest (Liao et al., 2002). For

example, this approach has been used to overexpress constitutively active mutants of the

upstream JNK and p38 MAPK family members, MKK3 and MKK7 in cardiac myocytes

(Liao et al., 2002; Wang et al., 1998b). These studies implicated JNK and SAPK2a in

cardiac hypertrophy and the inhibition of cardiac contractility, respectively (Wang et al.,

68

Page 117: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

1998b; Liao et al., 2002). To investigate the role of SAPK3 activation in these cells,

future studies would involve adenoviral overexpression of SAPK3 with a constitutively

active mutant of MKK6. However these studies would also require the additional

control of the SAPK2a/b inhibitor SB203580 so that effects could be attributed to

SAPK3.

No alterations in SAPK3 expression levels or phosphorylation were

detected in cardiac myocytes treated with a variety of different agonists either in long-

term exposure (Figure 3.9 and Figure 3.10) or acute exposure (unpublished

observations). It remains unknown why phospho-SAPK3 bands could not be detected in

these protocols. The phospho-antibody that has been used (Figure 3.10) has been used

previously to detect phosphorylated forms of SAPK2a and SAPK3 (Conrad et al., 1999;

Boppart et al., 2000). However, one possible explanation for the inability to detect

phospho-SAPK3 in cardiac myocytes might be reduced sensitivity of this antibody

towards phospho-SAPK3. This is possible because this antibody is raised against the

phosphorylated human SAPK2a protein rather than phosphorylated SAPK3.

Specifically there is 20% sequence difference in the ten amino acids either side of the

Thr-Gly-Tyr motif in the phosphorylation loop of the two proteins and this could be

responsible for reduced sensitivity to phospho-SAPK3. Alternatively, it could be due to

the relatively high levels of SAPK2a compared to SAPK3 in the heart (Lemke et al.,

2001). Thus when evaluating phosphorylation levels in cardiac myocyte lysates,

immunoblots are dominated by a phospho-SAPK2a band. This explanation seems likely

to be the case, since when the C2C12 skeletal myoblast cell line was treated with

arsenite and sorbitol for time periods ranging from 15 to 60 min and immunoblotted for

the presence of phospho-SAPK3, phospho-SAPK3 could be detected (unpublished

observations).

The development of an effective assay for measuring the phosphorylation

status or activity of SAPK3 in the heart should provide further clues on its role in this

tissue. For example, it may be possible to identify changes in the phosphorylation state

of SAPK3 by iso-electric focusing. The iso-electric point of SAPK3 should be altered

69

Page 118: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

when it becomes phosphorylated, and therefore the inactive protein would theoretically

show a different migration to the active, phosphorylated protein based on this property.

This approach has previously been used to separate phosphorylated and non-

phosphorylated forms of HSP25 and HSP27 (Clerk et al., 1998a). Alternatively, the

development of a SAPK3 antibody that efficiently and specifically immunoprecipitates

endogenous SAPK3 from cells would be extremely useful. However, the SAPK3

antibody used in this Chapter, although showing high specificity and avidity for SAPK3

in Western blotting and immunocytochemistry applications, fails to immunoprecipitate

SAPK3 efficiently (results not shown). Furthermore, although another SAPK3 antibody,

raised to the C-terminal tail sequence of this protein, has been shown to

immunoprecipitate SAPK3, it interferes with some subsequent assays of activity

(Hasegawa et al., 1999). An antibody recognising another domain of SAPK3 and that

does not interfere with its kinase activity is therefore desirable.

In summary, the results presented in this Chapter show the expression of

SAPK3 in HEK293 fibroblasts and cultured neonatal cardiac myocytes, and a unique

subcellular localisation of endogenous SAPK3 compared to that of SAPK2a. Through

the development of inhibitors for SAPK3 and methods to assay its activity, the role of

SAPK3 in these cells may be better understood. The expression of SAPK3 in isolated

cardiac myocytes suggests a role for this kinase in cardiac tissue. In the following

Chapter this will therefore be addressed by investigating the expression of SAPK3 in

whole heart tissue taken from embryonic, neonatal and adult animals.

70

Page 119: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Chapter 4

Evaluation of the expression and activation of SAPK3 in the intact heart

Page 120: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

4.1 Introduction

Despite the usefulness of cardiac myocytes as a model for the processes

occurring in the whole heart, discrepancies arise when comparing the results from

different studies using these cells. Specifically, while most reports tend to agree on a role

for ERK in mediating cardiac myocyte hypertrophy and survival (Bueno and Molkentin,

2002), there is disagreement on the role of JNK and p38 MAPK family members in this

process. For example, several studies have found that activation of JNK and p38 MAPK

family members leads to hypertrophy in cardiac myocytes (Ramirez et al., 1997; Wang et

al., 1998b; Clerk et al., 1998b; Zechner et al., 1997), while others have suggested that

these SAPKs play a role in cardiac myocyte apoptosis (Wang et al., 1998a; Adams et al.,

1998; Aoki et al., 2002; Remondino et al., 2003). It has been suggested that factors

including cardiac myocyte culture density and the extent of culture contamination with

fibroblasts may be responsible for such discrepancies (Hines et al., 1999; Liang and

Molkentin, 2003).

One example which highlights the danger of using overexpression models to

evaluate the cardiac function of MAPKs is a study which investigated the role of p38

MAPK family members in cardiac myocytes. This study used overexpression of wild-

type and dominant-negative mutants of SAPK2a and SAPK2b in cardiac myocytes to

implicate these proteins in pro-apoptotic and hypertrophic roles, respectively (Wang et

al., 1998a). Although other reports have supported a pro-apoptotic role for SAPK2a/b in

the heart (Iwai-Kanai et al., 2001; Iwakura et al., 2001; Mackay and Mochly-Rosen,

1999), other studies have claimed that SAPK2b mRNA and protein cannot be detected in

the heart, therefore appearing to dismiss any role for this protein in the wild-type heart

(Lemke et al., 2001; Liao et al., 2001).

More recent studies aimed at evaluating the role of MAPKs in the failing

heart have turned to transgenic mouse models, in which individual MAPKs or their

upstream activators are overexpressed specifically in the heart, or knockout models, in

which the expression of individual MAPKs or their upstream activators is lost. Although

71

Page 121: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

the results from these studies appear to be more uniform than those in cardiac myocytes,

this approach has still provided some contradicting results on the role of JNKs and p38

MAPKs in the heart. For example, some studies suggest that JNKs and p38 MAPKs are

involved in the hypertrophic response (Pellieux et al., 2000; De Windt et al., 2000; Dash

et al., 2003), whereas others have dismissed the role of these kinases in stimulating

cardiac hypertrophy (Sadoshima et al., 2002; Braz et al., 2003; Petrich et al., 2002). One

explanation for these contradictory results could be that the upregulation of MAPK

activities in these pathways can lead to spurious effects (Liang and Molkentin, 2003).

Alternatively, deletion of the MAPK of interest may alter the activities of other similar

protein kinases to compensate for the loss (Liang and Molkentin, 2003). Furthermore,

the cardiac phenotypes displayed in these studies may (where applicable) be dependent

on the type of stimulus that is used to stress the heart (Izumiya et al., 2003). Therefore,

whilst cardiac myocyte and transgenic mouse models can help to define the roles of

MAPK signalling proteins in the heart, ultimately studies that evaluate the expression and

activation of endogenous MAPK signalling molecules in the heart will be required.

As well as their proposed role in heart disease, MAPKs have been

implicated in mediating signalling events in the developing heart. At least two groups

have detected ERK activity in the embryonic cardiovascular system, as well as in other

regions of the developing embryo (Corson et al., 2003; Lai and Pawson, 2000).

Furthermore, ShcA, which acts as an adaptor protein upstream of the ERK signalling

cascade, is required for the development of the heart and the establishment of mature

blood vessels (Lai and Pawson, 2000). Specifically, ShcA-deficient embryos were found

to have impaired ERK activation and defects in the cardiovascular system, and died by

day 12 of gestation (Lai and Pawson, 2000).

SAPK3 mRNA has been previously detected in the heart (Mertens S et al.,

1996; Lemke et al., 2001), however no studies have examined the expression of SAPK3

protein in cardiac muscle tissue. In Chapter 3 it was shown that SAPK3 is expressed in

primary cultures of neonatal cardiac myocytes. This Chapter therefore addresses the

72

Page 122: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

second aim of this project, which is to examine the expression of SAPK3 in different

tissues with specific emphasis on the heart.

4.2 Results

4.2.1 SAPK3 is expressed in the heart

The role of p38 MAPKs in the heart has been suggested in a number of

studies examining the activation of SAPK2a/b (Bogoyevitch et al., 1996; Behr et al.,

2001), the effects of overexpressing SAPK2a/b (Wang et al., 1998a) and the use of the

SAPK2a/b inhibitor SB203580 (Mackay and Mochly-Rosen, 1999; Martin et al., 2001).

Based on earlier studies describing the skeletal muscle-restricted expression of SAPK3,

the role of SAPK3 in the heart has been discounted (Lechner et al., 1996; Li et al., 1996).

However, these previous studies did not examine SAPK3 expression at the protein level,

and it is not clear that the protocols used in these studies were optimised for detection of

lower levels of SAPK3 expression in tissues other than skeletal muscle.

The results presented in Chapter 3 showed that SAPK3 is expressed in

HEK293 fibroblasts, as well as in primary cultures of neonatal rat cardiomyocytes. It

was therefore of interest to evaluate the expression of SAPK3 in the whole heart. Lysates

from adult rat heart were immunoblotted with antibodies for either SAPK2a/b, SAPK3,

or both antibodies together (Fig 4.1A). A protein was detected with the SAPK3 antibody

(Fig 4.1A, lane 2), and its size (~42 kDa) agreed with that expected for SAPK3 (Mertens

et al., 1996). An antibody specific for SAPK2a/b detected a protein at ~41 kDa (Fig

4.1A, lane 1). A protein doublet was detected when immunoblotting with both antibodies

simultaneously (Fig 4.1A, lane 3). This confirmed the expression of SAPK3 in whole

isolated heart tissue and the difference in size of the p38 MAPK isoforms, and further

showed that the SAPK3 antibody did not cross-react with other closely-related p38

MAPKs.

73

Page 123: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

α-S

APK

2a/b

46 kDa

SAPK3

α-S

APK

3

α-S

APK

2a/b

-SA

PK3

SAPK2a

A

Bα-rat alkaline phosphatase only

(no SAPK3 antibody)

Figure 4.1: SAPK3 is expressed in the whole heart.(A) Rat hearts lysates were subjected to SDS-PAGE on large-format gels, before immunoblotting using the anti-SAPK3 antibody, anti-SAPK2a/b antibody, or both antibodies. (B) Whole hearts were isolated from adult mice, fixed, paraffin embedded and sectioned, before being subjected to staining with the SAPK3 antibody (left panel) or anti-rat secondary antibody only (right panel).

α-SAPK3left ventricleright ventricle

60X 60X

73a

Page 124: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

To determine whether SAPK3 staining could be visualised in the isolated

whole heart, sections of heart ventricles from wild-type FVB mice were stained with the

SAPK3 antibody, followed by detection with an anti-rat antibody linked to alkaline

phosphatase. As seen in Figure 4.1B, SAPK3 staining could be detected throughout the

left and right ventricles of the heart and no staining was detected when the primary

antibody was omitted. This agreed with the results of the Western blot analysis by

showing SAPK3 expression in the heart.

Having found SAPK3 to be expressed in rat and mouse heart, a rat heart

cDNA library was screened with the SAPK3 antibody. All 3 positive clones identified

from 5 x 105 plaques screened were confirmed to be SAPK3 following phage purification

and sequencing. The membranes from a sample screening procedure are shown in

Appendix III together with the corresponding sequencing results that identified the

positive plaques as SAPK3. The sequence was identical to that in the publicly available

databases (accession number NM_021746). This further confirmed the antibody

specificity and cardiac SAPK3 expression.

To address the possible differences that may occur between SAPK3

expression levels in cultured cardiac myocytes versus the whole heart, lysates were

obtained from myocytes at different culturing times. Thus, hearts freshly obtained from

neonatal rats were lysed and the expression of SAPK3 compared to lysates of isolated

myocytes at 16, 24 and 40 hours of culture (Figure 4.2A). In parallel, these lysates were

also immunoblotted with the SAPK2a/b antibody (Figure 4.2B). As shown in Figure

4.2A, SAPK3 expression appeared to decrease with increasing time in culture in cardiac

myocytes following their isolation from the heart, whereas no changes in the level of

SAPK2a expression were observed (Figure 4.2B).

Because the expression of SAPK3 in skeletal muscle has been well-

documented (Mertens S et al., 1996; Lechner C et al., 1996; Li Z et al., 1996), the relative

levels of SAPK3 in heart and skeletal muscle of adult mice were compared. Results

indicated similar levels of SAPK3 expression in fast-twitch (extra digitorium longus,

74

Page 125: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

Hours of culture: 0 16 24 40

45 kDaα-SAPK3 SAPK3

45 kDaPonceau

BHours of culture: 0 16 24 40

45 kDaα-SAPK2a/b SAPK2a/b

45 kDaPonceau

Figure 4.2: SAPK3 expression decreases in neonatal cardiac myocytes with increased culture time. Hearts were dissected from 1 day-old Sprague-Dawley rats and lysed, or the cardiac myocytes isolated and cultured for periods ranging from 16 to 40 hours before lysis. Lysates were then subjected to Western blotting with the SAPK3 antibody (A, upper panel) or the SAPK2a/b antibody (B, upper panel). Results shown are representative of 2 independent experiments. To show even loading of samples, nitrocellulose membranes were stained with Ponceau Acid Red following transfer of protein to the membranes.

74a

Page 126: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

EDL), slow-twitch (soleus, Sol) or mixed-fibre (tibialis anterior, TA and quadriceps,

Quad) skeletal muscle (Fig 4.3A, lanes 1-4). SAPK3 expression was also detected in

heart (Figure 4.3A, lane 5). SAPK3 expression was then compared in heart samples

obtained from human, mouse, rat, dog and pig. For each species, there was

immunoreactivity with a single band migrating between 43 and 46 kDa (Fig 4.3B, upper

panel). Other bands detected in rat and pig samples resulted from cross-reactivity with

secondary antibody (Fig 4.3B, middle panel). Therefore the cardiac expression of

SAPK3 appears to be conserved across this range of mammalian species. When heart

lysates were immunoblotted in parallel with SAPK2a/b antibody, a single band (~41

kDa) was detected for each species (Fig 4.3B, lower panel).

To determine the expression of SAPK3 in different tissues, lysates from

mouse heart, skeletal muscle, brain, liver, lung, thymus, testes, spleen and kidney were

immunoblotted with the SAPK3 antibody. In parallel, these tissues were also

immunoblotted with the SAPK2a/b antibody. SAPK2a/b expression was detected at

similar levels in all tissues examined (Fig 4.4, lower panel). In contrast, SAPK3

expression was extremely low in brain, liver, spleen and kidney (Fig 4.4, upper panel).

Higher expression levels were detected in heart, skeletal muscle, lung, thymus and testes

(Fig 4.4, upper panel). The expression of SAPK3 protein therefore appears to be

relatively restricted.

4.2.2 SAPK3 expression levels do not change in human heart disease

Previous studies have shown that the expression as well as the activation

states of MAPKs can be altered in disease states. For example, reports have suggested

that JNK MAPK expression decreases in cardiac failure (Cook et al., 1999), and that

SAPK3 mRNA decreases in the skeletal muscle of Duchenne Muscular Dystrophy

patients (Chen et al., 2000). To determine whether SAPK3 expression levels change

during heart disease, samples of left ventricular tissue were collected from human

patients from ischemic heart disease (IHD) or dilated cardiomyopathy (DCM) at the time

of transplantation surgery. A group of patients with normal or non-diseased hearts who

75

Page 127: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A Skeletal MuscleQuad Sol EDL TA H

SAPK346 kDa

BH

uman

Mou

se

Dog

Rat

Pig

46 kDa

2° Abcross-reaction

α-SAPK3SAPK3

46 kDa2° Abcross-reactionα-rat-HRP

only

46 kDaSAPK2a

α-SAPK2a/b

Figure 4.3: SAPK3 is expressed in the hearts of various species.(A) Mouse skeletal muscle and heart tissue were crushed under liquid nitrogen. Protein lysates were prepared and then subjected to Western blotting using the anti-SAPK3 antibody.Quad = Quadriceps; Sol = Soleus; EDL = Extra Digitorium Longus; TA = Tibialis Anterior.(B) Hearts from different animal species were crushed under liquid nitrogen. Protein lysates were prepared and then subjected to Western blotting using the anti-SAPK3 antibody (upper panel). Non-specific bands due to secondary antibody cross-reactivity are indicated (middle panel). Tissue lysates were also subjected to immunoblotting with anti-SAPK2a/b antibody (lower panel).

75a

Page 128: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

H Skm B Li Lu Th Te S K

SAPK2a/b

SAPK346 kDa

46 kDa

Figure 4.4: SAPK3 shows a relatively restricted tissue distribution in the mouse.Mouse tissues were crushed under liquid nitrogen. Protein lysates were prepared and then subjected to Western blotting using the anti-SAPK3 antibody (upper panel) or anti-SAPK2a/b antibody (lower panel). Abbreviations: H = Heart; Skm = Skeletal Muscle; B = Brain; Li = Liver; Lu = Lung; Th = Thymus; Te = Testes; S = Spleen; K = Kidney.

75b

Page 129: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

were found to be unsuitable for donor hearts due to other complications (for example,

renal tumours) were used as a control group. A total of 25 human samples were

examined for expression levels of SAPK3, SAPK2a/b and phosphorylated forms of p38

MAPK family members (Figure 4.5).

No consistent differences in SAPK3 expression were detected when

comparing failing and non-failing samples of human hearts (Figure 4.5). This was

further confirmed by subjecting selected donor, IHD and DCM samples to SDS-PAGE

and Western blotting on the same gels (results not shown). Furthermore, no change was

observed in the expression of SAPK2a/b in human hearts, which is consistent with

previous results (Fig 4.5) (Cook et al., 1999). However, when immunoblotted with a

phospho-p38 MAPK antibody, which detects the phosphorylated form of all p38

isoforms, only a single band was detected and this was consistent with the size of

SAPK2a, not SAPK3. When comparing the intensity of these bands between samples,

phospho-SAPK2a levels appeared to be increased in IHD and DCM samples (Figure 4.5).

A full statistical analysis of these samples with internal controls for each individual gel

found that although SAPK2a phosphorylation was increased in IHD and DCM, only the

increase in SAPK2a phosphorylation in IHD was significant compared to donor hearts

(Ng et al., 2003). The significant increase in SAPK2a phosphorylation in ischemic heart

disease agrees with previous results (Cook et al., 1999). No phospho-SAPK3 bands

appeared to be visualised when using the phospho-p38 MAPK antibody (Figure 4.5).

This may reflect the lower levels of SAPK3 activation or that changes are below the level

of detection with this technique.

4.2.3 SAPK3 is expressed throughout embryonic and postnatal development of

the heart

MAPK levels have previously been shown to change during heart postnatal

development (Kim et al., 1998). To determine whether any changes in cardiac expression

of SAPK3 occur in postnatal development, ventricular heart samples from neonatal and

76

Page 130: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A: Donorα-SAPK3

45 kDa SAPK3

45 kDaSAPK2a

α-SAPK2a

45 kDa

45 kDa

45 kDa

66 kDaα-Phospho-p38 MAPK

P-SAPK2a

B: Ischemic Heart Disease

66 kDa

45 kDa

α-SAPK3

α-SAPK2a

α-Phospho-p38 MAPK

SAPK3

SAPK2a

P-SAPK2a45 kDa

C: Dilated Cardiomyopathy

45 kDa

α-SAPK3

α-SAPK2a

α-Phospho-p38 MAPK66 kDa

45 kDa

SAPK3

SAPK2a

P-SAPK2a

Figure 4.5: SAPK3 expression levels do not change in failing human hearts.Human heart samples (donor, A; ischemic heart disease, B; or dilated cardiomyopathy, C) were crushed under liquid nitrogen. Protein lysates were prepared and then subjected to Western blotting using the anti-SAPK3 antibody (upper panels), anti-SAPK2a/b antibody (middle panels) or anti-phospho p38 MAPK antibody (lower panels). Open-ended arrows indicate non-specific bands.

76a

Page 131: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

adult rats were examined. However, no difference was seen in SAPK3 expression levels

when comparing these neonatal and adult hearts by Western blotting analysis (Fig 4.6A).

It was therefore assessed whether any changes may occur in SAPK3 protein

expression during embryonic development of the heart. Hearts were collected from

mouse embryos, ranging in age from embryonic day (E) 12.5 until birth. To initially

determine whether SAPK3 mRNA could be detected, embryonic hearts were

homogenised and total RNA was isolated. This RNA was then subjected to reverse-

transcription PCR (RT-PCR) with SAPK3 specific primers, to amplify SAPK3 mRNA.

As seen in Figure 4.6B, SAPK3 mRNA could be detected from as early as E12.5, and

continued to be detected throughout embryogenesis and in the newborn and adult heart.

To determine whether the SAPK3 protein levels in the heart may change throughout

embryonic development, hearts were homogenised and total protein isolated. These

protein lysates were then subjected to Western blotting with the SAPK3 antibody. As

shown in Fig 4.6C, SAPK3 protein could be detected throughout embryonic development

of the heart, with no changes seen in the expression levels throughout embryonic

development or compared to the adult heart. Western blots using the phospho-p38

MAPK antibody did not detect any phosphorylated SAPK2a or SAPK3 in these samples

(results not shown).

By Western blot analysis, it was shown that SAPK3 was expressed from at

least E12.5 in the heart. To further investigate the expression of SAPK3 during

development of other tissues, staining of embryos for SAPK3 expression was carried out.

Two different methods were used for staining embryos. The first method was paraffin

embedding, sectioning and staining of embryos. This method was required for embryos

at stage E12.5 or later due to the thicker outside layers of the embryo which are not able

to be penetrated by reagents. Therefore, E16.5, E13.5 and E12.5 embryos were stained

using this procedure. Staining of later-staged embryos was performed to determine the

spatial pattern of SAPK3 expression throughout the whole embryo during development.

77

Page 132: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

46 kDa

neon

atal

adul

t

SAPK3α-SAPK3

B

Age: E12.5 E13.5 E14.5 E15.5 E16.5 E17.5 E18.5 New Ad Skm Neg

500bp400bp300bp

SAPK3

C

Embryonic Day: 12.5 13.5 14.5 15.5 16.5 17.5 18.5 New Ad

45 kDa SAPK3α-SAPK3

45 kDa

220 kDa

66 kDa

97 kDaCoomassie

Figure 4.6: SAPK3 is constantly expressed during cardiovascular development.(A) Whole hearts were isolated from neonatal and adult rats and crushed under liquid nitrogen. Protein lysates were prepared and then subjected to Western blotting using the anti-SAPK3 antibody.(B) Hearts were isolated from embryonic (E12.5 – E18.5), newborn (New) and adult (Ad) mice, and total RNA extracted. SAPK3-specific primers were then used to amplify a 414bp fragment by RT-PCR. RNA isolated from skeletal muscle (skm) was used as a positive control, and an identical reaction with template omitted was used as a negative control (Neg).(C) Hearts were isolated from embryonic (E12.5 – E18.5), newborn and adult mice,and crushed under liquid nitrogen. Protein lysates were prepared and then subjected to Western blotting using the anti-SAPK3 antibody (upper panel). Coomassie Blue staining confirmed comparable levels of protein loaded in all lanes (lower panel).

77a

Page 133: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Figure 4.7 shows the staining of E16.5, E13.5 and E12.5 mice with

haematoxylin and eosin to illustrate the anatomical organisation of the embryos. Figures

4.8 and 4.9 show the results of staining E16.5, E13.5 and E12.5 mice with the SAPK3

antibody. Western blotting analysis had already shown that SAPK3 was expressed in the

hearts of embryos at these stages (Figure 4.6C). In the E16.5 mouse, SAPK3 levels were

highest in the heart, diaphragm, bladder and patches of skeletal muscle including in the

limbs. Lower levels of staining could be detected in the lung, and very light staining

could also be detected in the brain and liver (Figure 4.8A). A closer examination of the

heart in these embryos showed staining throughout the ventricular and atrial walls, as

well as in the trabeculae (Figure 4.8B). No staining was detected in the control embryo,

where the primary SAPK3 antibody was omitted (Figure 4.8A, left panel). In the E13.5

mouse, staining was highest in the heart, brain, spinal cord, tongue, intestine and lung

(Figure 4.9A). In the E12.5 mouse, SAPK3 staining appeared to be particularly high in

the heart, lung, tongue, brain and spinal cord (Figure 4.9B). No staining was detected in

control embryos, where the primary SAPK3 antibody was omitted (Figure 4.9, left

panels).

Western blot analysis indicated that expression of SAPK3 in the heart was

constant from at least E12.5 (Figure 4.6C). Earlier embryos were too small to

specifically isolate their hearts for Western blot analysis. Therefore, E8.5 and E10.5

embryos were chosen for staining analysis to determine the time point at which SAPK3

expression was switched on. Wholemount staining could be used for these embryos

without sectioning. This staining showed SAPK3 to be expressed at its highest levels in

the telencephalon (forebrain) and along the dorsal edge of the E8.5 mouse, where the

somites are located (Fig 4.10A). Only very faint staining for SAPK3 was seen under

these conditions in the developing heart. However, SAPK3 expression could be detected

throughout the embryo if the staining protocol was lengthened (Fig 4.10A). Even with

this increased detection time, no staining was visualised in the control embryo, in which

the primary antibody had been omitted from the staining procedure (Fig 4.10A). This

suggested that SAPK3 was expressed in most regions of the embryo at this early stage,

albeit at lower levels. In the E10.5 mouse, SAPK3 staining was specifically detected in

78

Page 134: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

diaphragm

E16.5

30X

heart

lung

liverbladder

vertebraeribs

forelimb

E13.5

40X

telocoelmyelencephalon

tongue

heart

lungliververtebrae

mesencephalon

myelencephalon

tongue

heart

lung

liver

spinal cord

tail

E12.5

55X

B

C

Figure 4.7: Haematoxylin and eosin staining of E16.5, E13.5 and E12.5 mice.Embryos were dissected from FVB wild-type mice at E16.5 (A), E13.5 (B) and E12.5 (C). Following fixation in paraformaldehyde overnight, embryos were embedded in paraffin wax and sectioned. Paraffin sections were then stained with haematoxylin and eosin to show the general histological features of the embryos.

78a

Page 135: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

α-rat-alkaline phosphatase only (no SAPK3 antibody) α-SAPK3

30X

E16.5

heartlung

bladder

diaphragmvertebrae

muscle

30X

E16.5A

forelimb

hindlimb

B

120X

trabeculae

ventricular wall

atrial wall

Figure 4.8: SAPK3 is expressed in various tissues of the E16.5 mouse.(A) Embryos were dissected from FVB wild-type mice at E16.5. Following fixation in paraformaldehyde overnight, embryos were embedded in paraffin wax and sectioned. Paraffin sections were then stained with the SAPK3 antibody, followed by an anti-rat secondary antibody linked to alkaline phosphatase. Signal detection with alkaline phosphatase substrates was for 60 min to show patterns of SAPK3 expression.(B) Further examination of the hearts of these embryos showed specific staining for SAPK3 in the ventricular and atrial walls, and in the trabeculae of the heart.

78b

Page 136: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

α-rat-alkaline phosphatase only (no SAPK3 antibody) α-SAPK3

A

40X

E13.5mesencephalonmyelencephalon

heart

tongue

lung

spinal cord

nasal chamber

intestine

40X

E13.5

E12.5

45X

B E12.5

45X

heart

tongue

mesencephalon

myelencephalon

spinal cord

lung

Figure 4.9: SAPK3 is expressed in various tissues of E13.5 and E12.5 mice.Embryos were dissected from FVB wild-type mice at E13.5 (A) and E12.5 (B). Following fixation in paraformaldehyde overnight, embryos were embedded in paraffin wax and sectioned. Paraffin sections were then stained with the SAPK3 antibody, followed by an anti-rat secondary antibody linked to alkaline phosphatase. Signal detection with alkaline phosphatase substrates was for 60 min to show patterns of SAPK3 expression.

78c

Page 137: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

E 8.5 E 8.5

157x157x

hearttelencephalon

tailsomites

E 8.5

157x

ControlSignal detection: 45 min

Anti-SAPK3Signal detection: 15 min

Anti-SAPK3Signal detection: 45 min

B

heart

mesencephalon

telencephalon

hindlimb bud

E 10.5 E 10.5Non-specific stainingtail

70x 70x

forelimb bud

somitesAnti-SAPK3

Signal Detection: 15 minControl

Signal detection: 15 min

E 10.5

76x

E 10.5

76x

ControlSignal detection: 45 min

Anti-SAPK3Signal detection: 45 min

Figure 4.10: SAPK3 expression in E8.5 and E10.5 embryos using wholemount staining.Whole embryos were dissected from FVB wild-type mice at 8.5 (A) and 10.5 (B) days gestation. Following fixation in Dent’s fixative overnight, embryos were processed for staining with the SAPK3 antibody, and an anti-rat secondary antibody linked to alkaline phosphatase was used to detect staining. Signal detection with alkaline phosphatase substrates was for 15 min or 45 min to show areas of highest and lower SAPK3 expression, respectively.

78d

Page 138: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

the heart, limb buds, telencephalon, somites and tail tip (Fig 4.10B). When colour

detection was allowed to continue for 45 min, the embryo changed to an intense

blue/purple colour when compared to the control embryo where the primary SAPK3

antibody had been omitted (Fig 4.10C). This suggested that SAPK3 is also expressed in

other tissues at lower levels in the E10.5 mouse.

4.3 Discussion

Several reports have suggested that MAPKs play important roles in the

development and disease of the heart (Lai and Pawson, 2000; Corson et al., 2003; Cook

et al., 1999). Due to the detection of SAPK3 in primary cultures of neonatal cardiac

myocytes (Chapter 3), it was of interest to determine the expression of SAPK3 in the

intact heart. The discovery of lower SAPK3 expression levels with increased culture

time of cardiac myocytes (Figure 4.2) highlighted the importance of performing these

studies in the whole heart as well as in isolated primary cardiac myocyte cultures. This

illustration of SAPK3 cardiac expression has important consequences when evaluating

studies that implicate the actions of SAPK2a in the heart. For example, the effects of the

p38 MAPK family upstream activators MKK3 and MKK6 in the heart cannot be solely

attributed to the downstream activation of SAPK2a (Liao et al., 2001) because it is now

possible that SAPK3 actions may also contribute to the final outcome.

It was shown in this Chapter that SAPK3 expression in the heart was

relatively high when compared with other tissues, and this cardiac expression was

conserved throughout a variety of mammalian species (Figure 4.3B). This, along with

the detection of SAPK3 in Xenopus and Zebrafish (Perdiguero et al., 2003; Fabre S et al.,

2000), suggests the evolutionary conservation and thus an important function of SAPK3

across this diversity of species. Indeed, it seems unlikely that the functions of SAPK3 are

redundant with those of SAPK2a due to its unique tissue distribution (Figure 4.4) and its

unique subcellular localisation (Figure 3.3). It is unknown why SAPK3 shows this

unique tissue distribution. Due to the localisation of SAPK3 at the mitochondria in

cardiac myocytes (Chapter 3), it was hypothesised that the tissues showing the highest

79

Page 139: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

levels of SAPK3 expression may also contain higher numbers of mitochondria and thus

higher levels of mitochondrial proteins. However, parallel Western blotting with

antibodies against the mitochondrial proteins HSP60 and HSP70 did not show similar

patterns of tissue distribution to SAPK3 (results not shown). Clearly this correlative link

will require further evaluation. Avenues for further investigation include an assessment

of the promoter regions controlling SAPK3 expression.

Staining of transverse sections of adult mouse heart used alkaline

phosphatase-labelled secondary antibodies to detect the distribution of SAPK3 in the

heart (Figure 4.1B). The fluorescently-labelled secondary antibodies that were used in

Chapter 3 could not be used in these studies due to high cross-reactivity with

immunoglobulins in mouse tissue sections (results not shown). The co-localisation of

SAPK3 with the mitochondria could therefore not be confirmed in the whole heart

sections, since the SAPK3 staining in this case required viewing with a conventional light

microscope, which does not show the same level of resolution as the confocal microscope

(White et al., 1987). Although protein localisation in isolated cardiac myocytes can be

the same in the whole heart, as is the case for A-kinase anchoring protein 100

(AKAP100) (Yang et al., 1998a), further work will be required to confirm this for

SAPK3. In some cases proteins in isolated cardiac myocytes are not necessarily the same

in the whole heart. For example, while MKK6 and SAPK2a are diffusely localised

through the cytoplasm in cultured cardiac myocytes, they co-localise with t-tubular/z-disc

structures in the adult heart (Liang and Molkentin, 2003; Lemke et al., 2001).

This Chapter examined the expression and activation state of SAPK3 in the

failing heart to determine its possible role in this process. Whilst alterations were seen in

the phosphorylation state of SAPK2a in failing hearts, the analysis of human heart

samples did not show any changes in SAPK3 expression or phosphorylation state (Figure

4.5). However, as discussed in Chapter 3, this could be explained by the lower sensitivity

of the antibody for phosphorylated SAPK3. Alternatively, the lower expression of

SAPK3 in the heart compared to SAPK2a (Lemke et al., 2001) could limit detection.

The development of specific antibodies for phosphorylated SAPK3 should therefore

80

Page 140: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

assist in the detection of activated SAPK3 in the heart. Furthermore, antibodies suitable

for immunoprecipitation of SAPK3 would facilitate studies to directly assess its activity

by in vitro kinase assays.

An analysis of SAPK3 expression in developing heart samples suggested

that SAPK3 protein expression remained unaltered from at least E12.5 into adulthood

(Figure 4.6). Staining of embryos at E12.5, E13.5 and E16.5 also showed that compared

to other tissues, SAPK3 expression was relatively high in the heart (Figures 4.8 and 4.9).

Similar to the later staged embryos, relatively high cardiac expression of SAPK3 was

detected at E10.5. However, when analysing the E8.5 mouse, SAPK3 appeared to be

expressed at its highest levels in the developing brain (the telencephalon) rather than the

heart (Figure 4.10). The lower expression of SAPK3 in the heart at this stage may be a

reflection of the earlier developmental stage of this tissue. For example, at E8.5, the

heart is at its initial stages of development, undergoing looping from a simple linear tube

and beginning to contract, whereas at E10.5, paired heart chambers have been formed and

the interventricular septum is being formed (Fishman and Chien, 1997; Rugh, 1968).

Quantitative analyses will be required to determine the levels of SAPK3 expression at the

early stages of heart development and whether SAPK3 is expressed in the heart region as

early as E7.0, when there is assembly of the myocardial plate (Fishman and Chien, 1997).

It is interesting to note that the general pattern of SAPK3 expression

changed in several tissues. For example, in the brain, high expression levels of SAPK3

were detected at E8.5 – E13.5, but only low levels of staining were detected in the brain

at E16.5. The pattern of staining seen in the E16.5 mouse (Figure 4.8) was similar to that

seen in the adult mouse, with higher levels of staining seen in skeletal muscle, heart and

lung, and lower levels in the liver and brain (Figure 4.4). This suggests that SAPK3

expression levels are altered in embryonic development of tissues such as the brain

before remaining constantly low in postnatal development. Quantitative analyses using

brain samples from mice at different stages of postnatal development would be required

to confirm these findings.

81

Page 141: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Further work will be required to determine the role of SAPK3 in the heart

and other tissues during development. It may be possible that SAPK3 plays a role in the

differentiation of heart progenitor cells to become terminally differentiated cardiac

myocytes. For example, the overexpression of SAPK3 has previously been shown to

drive differentiation of C2C12 myoblasts into myotubes (Lechner et al., 1996), and

although there are striking differences in the development of cardiac muscle cells and

skeletal muscle cells it remains possible that SAPK3 could play a similar role in the

heart. SAPK3 has also previously been shown to phosphorylate the Myocyte Enhancing

Factor (MEF) family of transcription factors in vitro (Marinissen et al., 1999). These

transcription factors have been shown to play critical roles in the development of the

heart, particularly in late-stage differentiation of this tissue (Lilly et al., 1995; Ross et al.,

1996; Goswami et al., 1994; Bour et al., 1995). Although SAPK3 has not been shown to

translocate to the nucleus in cardiac myocytes (Chapter 3), MEFs have been shown to

shuttle between the cytoplasm and the nucleus (De Angelis et al., 1998). Alternatively, a

specific stimulus may cause SAPK3 to translocate into the nuclear compartment. The use

of conditional knockouts of SAPK3 in the heart may help to resolve some of these issues.

A major challenge remains in the identification of the extracellular stimuli

that lead to the activation of SAPK3 as well as its downstream substrates. To address

these challenges requires further analysis of SAPK3 in a cellular and tissue context

together with screening strategies to identify SAPK3 substrates and interacting partners.

In the next chapter, the ability of SAPK3 to phosphorylate mitochondrial target proteins

is further explored.

82

Page 142: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Chapter 5

Investigation of the substrate specificity of SAPK3

Page 143: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

5.1 Introduction

The activation of MAPKs following the exposure of cells to specific

extracellular stimuli leads to the phosphorylation of specific intracellular substrates.

This addition of phosphate groups to specific serine and/or threonine residues within

these substrate proteins can alter their conformation, and thus their activity or function.

These changes contribute to the cellular response and it therefore follows that

identifying the substrates of each MAPK can provide clues on the possible roles that a

MAPK plays within the cell (Cohen, 1997; Berwick and Tavare, 2004).

A variety of protein domains, such as the Src-homology domains 2 or 3

(SH3 or SH2 domains), Pleckstrin Homology domains (PH domains), Phosphotyrosine

Binding domains (PTB domains) and PSD-95/Discs-large/ZO-I domains (PDZ

domains), are important for the specificity of interactions between the proteins involved

in intracellular signaling pathways (Pawson and Nash, 2003). Furthermore, conserved

regions of several MAPK phosphatases, MAPKKs, scaffolds and substrates have been

identified and called “docking domains” or “Kinase Interaction Motifs” (KIMs). These

domains or motifs mediate interactions with specific MAPKs (Tanoue et al., 2000;

Sharrocks et al., 2000; Wiltshire et al., 2002). In the case of MAPK substrates, the

interaction via these docking domains or KIMs appears critical for their phosphorylation

(Yang et al., 1999; Livingstone et al., 1995; Yang et al., 1998b; Wiltshire et al., 2002).

Another determinant of substrate phosphorylation is the sequence

immediately surrounding the Ser/Thr to be phosphorylated. The consensus amino acid

sequence for substrate phosphorylation by the archetypical MAPK, Extracellular Signal-

Regulated Kinase (ERK), has been defined by the four amino acid linear sequence

(Pro/X)-X-(Ser/Thr)-Pro, where X is any amino acid and the amino acid phosphorylated

is indicated in italics (Clark-Lewis et al., 1991). As discussed in Chapter 1, the

phosphorylated tyrosine residue in the activation loop of MAPKs is thought to be

responsible for MAPK specific substrate recognition of serine or threonine residues that

are directly followed by a proline residue (Canagarajah et al., 1997; Alvarez et al., 1991;

83

Page 144: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Clark-Lewis et al., 1991). More recent work has evaluated how amino acids within the

active sites of serine/threonine kinases make contact with the substrate phosphorylation

site. This has allowed the definition of consensus sequences that are more specific for

each protein kinase (Brinkworth et al., 2003).

Although MAPKs were initially shown to mediate nuclear, transcriptional

events through the phosphorylation of their specific transcription factor substrates

(Boulton et al., 1991), increasingly MAPK substrates have been found in other cellular

compartments such as the cytoskeleton (Goedert et al., 1997b; Neidhart et al., 2001;

Lefebvre et al., 2002; Huang et al., 2003; Terret et al., 2003), the plasma membrane

(Hasegawa et al., 1999; Wu et al., 2003) or the mitochondria (Donovan et al., 2002;

Schroeter et al., 2003; Wiltshire et al., 2002). As discussed in Chapter 1, little is known

about the substrate specificity of SAPK3 within the cell or in vitro. Studies using

overexpressed SAPK3 have shown that this kinase is stress-activated and that it can

phosphorylate proteins such as Activating Transcription Factor 2 (ATF2) and Myelin

Basic Protein in vitro (Cuenda et al., 1997). Data presented in Chapter 4 showed the

expression of SAPK3 in a variety of tissues, with highest levels of expression in skeletal

muscle, heart, lung, thymus and testes. This suggested a role for SAPK3 in a number of

tissues. Possible substrates for SAPK3 include the dystrophin complex protein α1-

Syntrophin (Hasegawa et al., 1999) which was isolated in a two-hybrid screen for

SAPK3-interacting proteins, and the microtubule-associated protein Tau (Goedert et al.,

1997b) identified as a potential substrate using in vitro kinase assays. However, given

the larger number of substrates determined for other MAPKs, it remains likely that there

are many substrates of SAPK3 that are not yet defined.

In Chapter 3, SAPK3 was shown to colocalise with mitochondrial markers.

The exposure of cells to hyperosmolarity, a stress previously shown to activate SAPK3

when overexpressed in cells (Cuenda et al., 1997), did not alter this localisation (results

not shown). It therefore seems likely that substrates for SAPK3 will be identified at the

mitochondria. Proteins involved in the regulation of cell death, such as Bcl-2, Bim, and

Bad, can be found at the mitochondria, where they have been implicated in a number of

84

Page 145: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

signalling pathways (Danial et al., 2003; Klumpp and Krieglstein, 2002). Furthermore,

several of these proteins are phosphorylated by members of the MAPK family, including

SAPKs such as JNK (Deng et al., 2001; Ley et al., 2003; Donovan et al., 2002). This

phosphorylation has been found to alter cell survival. For example, the phosphorylation

of Bad on Ser 112 and Ser 136 by survival-promoting kinases sequesters Bad in the

cytosol by its complex formation with 14-3-3 proteins, thus preventing Bad from

antagonising the activity of the anti-apoptotic Bcl-XL (Zha et al., 1996). However,

phosphorylation of Bad on Ser 128 by JNK (Donovan et al., 2002) or Cdc2 (Konishi et

al., 2002) inhibits the ability of 14-3-3 proteins to sequester Bad, and stimulates the pro-

apoptotic activity of Bad (Konishi et al., 2002). Thus, the phosphorylation of pro- and

anti-apoptotic proteins is a complex mechanism of regulating cell survival. It is

therefore likely that a number of different protein kinases are responsible for this

regulation.

It has been recently shown that JNK phosphorylates the protein Sab

(Wiltshire et al., 2002). This protein was previously identified as a binding partner and

negative regulator of Bruton’s Tyrosine Kinase (Btk) (Matsushita et al., 1998; Yamadori

et al., 1999). Active JNK and Sab were shown to co-localise at the mitochondria

(Wiltshire et al., 2002). Due to the localisation of SAPK3 at the mitochondria (Chapter

3) it was of interest to determine whether Sab could act as a substrate of SAPK3. The

aim of this Chapter was therefore to determine whether SAPK3 could phosphorylate

mitochondrial protein substrates such as Sab or Bcl-2 family members including Bcl-2

and Bad. This first involved optimising a protocol for activating recombinant SAPK3 in

vitro using a constitutively-active MKK6 mutant. A constitutively active mutant of

MKK6 has been previously reported, where the amino acids Ser207 and Thr211 within the

activation loop of this protein kinase have been mutated to Asp (D), to mimic

phosphorylation of these residues (Alonso et al., 2000). This constititutively active

MKK6 mutant, MKK6(DD), can be used in the activation of SAPK3 in vitro (Alonso et

al., 2000). Following activation of GST-SAPK3, protein kinase assays were performed

to determine its substrate preferences.

85

Page 146: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

5.2 Results

5.2.1 The activation of SAPK3 in vitro

To identify SAPK3 substrates, activated SAPK3 was required for in vitro

kinase assays. As discussed in Chapter 1, the mutation of threonine and tyrosine

residues in the activation loop of MAPKs to glutamate does not confer activity as it does

for other kinases such as the MKKs (Mansour et al., 1994). A constitutively-active

mutant of the SAPK3 upstream activator, MKK6 (MKK6-DD) was therefore obtained.

Full-length MKK6(DD) had previously been cloned into the pMalc2 vector, creating a

fusion with the MalE gene. This allowed the expression in E. coli of protein of MKK6

and the Maltose Binding Protein (Alonso et al., 2000).

To confirm that MalE-MKK6(DD) was capable of phosphorylating GST-

SAPK3, kinase assays were performed with differing amounts of MalE-MKK6(DD) and

GST-SAPK3 in the presence of [γ-32P]-labelled ATP (Figure 5.1A). In this initial

experiment, it was determined that 10 µg of MalE-MKK6(DD) and 5 µg of GST-

SAPK3 was sufficient for MalE-MKK6(DD) phosphorylation of SAPK3, as determined

by the incorporation of 32P into SAPK3. To determine whether the phosphorylation of

GST-SAPK3 by MalE-MKK6(DD) increased with respect to time, 5 µg of GST-SAPK3

was incubated with [γ-32P]-labelled ATP alone, or with 10 µg MalE-MKK6(DD) and [γ-32P]-labelled ATP, for times ranging from 30 min to 8 hours. The reactions were then

stopped and reaction components separated by SDS-PAGE, as described in Chapter 2.

As shown in Figure 5.1B, the incorporation of 32P into the GST-SAPK3 protein

increased as GST-SAPK3 was incubated with MalE-MKK6(DD) for longer periods of

time. This phosphorylation was not due to autophosphorylation, since only very low

levels of phosphorylation were detected when GST-SAPK3 was incubated with [γ-32P]-

labelled ATP alone.

A protocol was required to test whether the phosphorylation of SAPK3

resulted in its activation. Thus, GST-SAPK3 that had previously been incubated with

86

Page 147: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

MalE-MKK6(DD) (µg):GST-SAPK3 (µg):

0 0.5 1 2 10 10 10 0 0.5 0.5 0.5 0.5 0.5 0 5 5

45 kDa

66 kDa

97 kDaGST-SAPK3MalE-MKK6(DD)

Autoradiograph

GST-SAPK3MalE-MKK6(DD)

45 kDa

66 kDa

97 kDa

Coomassie

B

GST-SAPK3

GST-SAPK3MalE-MKK6(DD)

30 min 1 hour 4 hours 8 hours

Autoradiograph

Coomassie

66 kDa97 kDa

66 kDa97 kDa

MalE-MKK6(DD): - + - + - + - +GST-SAPK3: + + + + + + + +

Figure 5.1: Optimising a protocol for GST-SAPK3 activation.(A) GST-SAPK3 (0.5 µg or 5 µg as indicated) was incubated with different amounts (0.5, 1, 2 or 10 µg) of a constitutively active MKK6 mutant (MalE-MKK6DD) and 2µCi of [γ-32P]-ATP, or radiolabelled ATP only, at 30°C for 30 min. The reactions were then stopped with SDS sample buffer and proteins separated by SDS-PAGE. An autoradiograph of the gel is shown after 1 hour exposure time. The lower panel shows the Coomassie stained gel to illustrate even protein loading. (B) GST-SAPK3 (5 µg) was incubated with MalE-MKK6(DD) (10 µg) and 2µCi of [γ-32P]-ATP, or radiolabelled ATP only, at 30°C for 30 min to 8 hours. The reactions were then stopped with SDS sample buffer and proteins separated by SDS-PAGE. An autoradiograph of the gel is shown after 1 hour exposure time. The lower panel shows the Coomassie stained gel to illustrate even protein loading.

86a

Page 148: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

MalE-MKK6(DD) or incubated alone, was immobilised on GSH-sepharose, washed

with Activation Buffer, and subjected to kinase assays with a known in vitro substrate

GST-ATF2 (Cuenda et al., 1997) and [γ-32P]-labelled ATP. As seen in Figure 5.2, when

GST-SAPK3 was incubated with MalE-MKK6(DD), this increased SAPK3 activity by

approximately 40-fold towards GST-ATF2, when compared to control reactions in

which SAPK3 was incubated in the absence of MKK6(DD).

5.2.2 Investigating the phosphorylation of mitochondrial proteins by SAPK3

To implicate SAPK3 in a role at the mitochondria, it was of interest to

determine whether SAPK3 could phosphorylate mitochondrial proteins. Members of the

MAPK family such as ERK and JNK have previously been found to phosphorylate

mitochondrial proteins involved in apoptosis (Ishikawa et al., 2003; Donovan et al.,

2002; Ley et al., 2003; Deng et al., 2001). Thus, mammalian expression constructs

encoding the anti-apoptotic proteins Bcl-2 and Bag-1, and the pro-apoptotic proteins

BimEL, Bad, Blk and Bid, were obtained. Bacterial expression constructs were not

chosen for these experiments due to the complications such as instability, insolubility,

and low protein yields when expressing these proteins in bacteria (D. Huang, personal

communication). Each mammalian expression construct allowed expression of the

protein of interest with the N-terminal fusion to either a Flag or HA epitope tag.

Specifically, the constructs encoding Bcl-2 and Bag-1 contained a Flag tag, whilst the

BimEL, Bad, Blk and Bid constructs contained a HA tag. In addition, a mammalian

expression construct encoding the caspase inhibitor p35 was obtained. This was co-

transfected into cells with the pro-apoptotic protein constructs, to prevent these cells

from undergoing apoptosis. As a control, anti-apoptotic proteins were co-transfected

with empty Bluescript vector DNA.

To confirm expression of these mitochondrial proteins following

transfection, lysates were made from the transfected cells and subjected to Western

blotting with HA or Flag antibodies. As seen in Figure 5.3, all proteins appeared to be

87

Page 149: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

MalE-MKK6(DD) - + GST-SAPK3 + + GST-ATF2 + +

45 kDa

30 kDaGST-ATF2

GST-ATF2

45 kDa

30 kDa

66 kDa

66 kDa

GST-SAPK3

GST-SAPK3

Autoradiograph

Coomassie

MalE-MKK6(DD) - + GST-SAPK3 + + GST-ATF2 + +

05

1015202530354045

Fold

Act

ivat

ion

(Pho

spho

ryla

tion

of G

ST-A

TF2)

Figure 5.2: GST-SAPK3 pre-incubated with MalE-MKK6(DD) shows increased activity towards GST-ATF2.GST-SAPK3 (5 µg) was either incubated with MalE-MKK6(DD) (10 µg) and ATP, or ATP only, overnight at 30°C. GST-SAPK3 was separated from the other reaction components by incubation with GSH-sepharose, then incubated with GST-ATF2(19-96) and 2µCi of [γ-32P]-ATP at 30°C for 35 min. The reaction was stopped with the addition of SDS-PAGE sample buffer, and proteins separated by SDS-PAGE. An autoradiograph of the gel is shown after 7 hours exposure time. The middle panel shows the Coomassie stained gel to illustrate even protein loading, and the lower panel shows the quantitation of 3 independent assays as determined by Cerenkov counting of the GST-ATF2 bands. Results are expressed as fold activation of SAPK3 relative to assays conducted in the absence of MalE-MKK6(DD). The results are shown as means ± standard error (SE).

87a

Page 150: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

Bcl-2 Bag-1 -Transfection:

30 kDa

20.1 kDa

45 kDa

14.3 kDa

Bcl-2Bag-1

****

α-Flag

*

B

Transfection: BimEL Bad Blk Bid -

30 kDa

20.1 kDa

45 kDa

14.3 kDa

Bid

Blk

BimEL/Bad

**α-HA

Figure 5.3: Expression of overexpressed mitochondrial proteins in HEK293 fibroblasts.HEK293 fibroblasts were transfected with 0.6 µg of the expression constructs Flag-Bcl-2 or Flag-Bag-1 (both in conjunction with 0.6 µg empty Bluescript vector) or 0.6 µg HA-Bim, HA-Bad, HA-Blk or HA-Bid (all in conjunction with 0.6 µg of the caspase inhibitor p35), or the controls (1.2 µg Bluescript only or p35 only; denoted “-”) and left for 40 hours to express the proteins. The cells were then lysed, sample buffer added, and Western blotting performed using Flag (A) or HA (B) antibodies. Arrows indicate the detected proteins. Asterisks indicate cross-reacting bands (identity unknown).

87b

Page 151: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

expressed following transfection, since immunoreactive bands of approximately the

expected molecular weight were apparent when HEK293 fibroblasts were transfected

with the mitochondrial proteins but not when they were transfected with Bluescript or

p35 alone. Using protein insert sequence information, the molecular weights of the

mitochondrial protein products were calculated to be: Bcl-2, 21.9 kDa; Bag-1, 21.9 kDa;

BimEL, 19.2 kDa; Bad, 16.5 kDa; Blk, 15.5 kDa; Bid, 22 kDa. Comparing these

calculated molecular weights to the main band seen in each case on the Western blots,

most of the proteins appeared to migrate at approximately 5-10 kDa above the expected

size, with the exception of Blk and BimEL which appeared to migrate only slightly (1-2

kDa) above the expected size. When immunoblotting for Bcl-2 and Bag-1 several other

bands besides those representing Bcl-2 and Bag-1 were also present (Figure 5.3A, lanes

1 and 2). However, these were most likely due to the Flag antibody cross-reacting non-

specifically with other proteins in the lysates, since they were also present when

HEK293 fibroblasts were transfected with Bluescript alone (Figure 5.3A, lane 3).

Although Western blots with the HA antibody appeared to be cleaner (Figure 5.3B), an

extra band was detected in the case of Bad. This extra band, since it is only slightly

larger than the calculated molecular weight of Bad, and it did not appear in the p35-only

transfection, is most likely to be due to post-translational modifications of Bad. In

addition, an extra band was detected in the case of Blk transfection. Since this band

migrated at approximately double the calculated molecular weight of Blk, this was

unlikely to be due to post-translational modifications of Blk and the identity of this band

is unknown.

To confirm that the expressed proteins could be immunoprecipitated from

the cells, immunoprecipitations were performed from the HEK293 cell lysates using HA

or Flag antibodies. Western blotting was then performed on the pellets and supernatants

of these immunoprecipitations, as described in Chapter 2. As shown in Figure 5.4 by

asterisks, proteins were detected by Western blotting in the pellet fractions of the

immunoprecipitations for Bcl-2, Bag-1, BimEL and Blk. In addition Bag-1 and Bim

were also detected in the supernatant fractions of the immunoprecipitations, suggesting

that the immunoprecipitation was not 100% efficient in these cases. Protein bands for

88

Page 152: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

AImmunoprecipitate Flag; Western Blot Flag

45 kDa

30 kDa

66 kDa

20.1 kDa

14.3 kDa

Blk Bid BimEL -Bad

IgG HC

IgG LC

IgG HC

IgG LC

P S P S

Bcl-2 Bag-1 -P S

45 kDa

30 kDa

66 kDa

20.1 kDa

**

BImmunoprecipitate HA; Western Blot HA

P S P SP S P S

****

P S

Figure 5.4: Immunoprecipitation of mitochondrial proteins from HEK293 fibroblast lysates.HEK293 fibroblasts (3 dishes of 1 x 105 cells per construct) were transfected with 1 µg of the expression constructs Flag-Bcl-2 or Flag-Bag-1 (both in conjunction with 1 µg empty Bluescript vector) or 1 µg HA-Bim, HA-Bad, HA-Blk or HA-Bid (all in conjunction with 1 µg of the caspase inhibitor p35), or the controls (2 µg Bluescript only or p35 only; denoted as “-”) and allowed to express the proteins for 40 hours. The cells were then lysed, and Flag (A) or HA (B) antibodies added. Protein G-Sepharose was then added to capture the antigen-antibody complex, and immunoprecipitates washed extensively. These immunoprecipitates and lysate supernatants were then diluted in SDS sample buffer and subjected to SDS-PAGE and Western blotting, probing with Flag (A) or HA (B) antibodies. Asterisks indicate proteins in pellet or supernatant fractions of immunoprecipitation as expected from the calculated molecular weight.P = Immunoprecipitation pellet fraction; S = Immunoprecipitation supernatant fraction; IgG HC = Immunoglobulin Heavy Chain; IgG LC = Immunoglobulin Light Chain.

88a

Page 153: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Bid and Bad could not be detected in the pellet fractions of the immunoprecipitations.

However, extra bands were present in the supernatant fractions of these

immunoprecipitations that were not present in the “p35 only” control. This suggested

that Bid and Bad were expressed, but were not immunoprecipitated efficiently.

To determine whether GST-SAPK3 was capable of phosphorylating these

pro- and anti-apoptotic proteins, HEK293 fibroblasts were transfected with the protein

constructs and allowed to express the proteins. The cells were then lysed, and

immunoprecipitations performed as described in Chapter 2. The immunoprecipitated

proteins were then subjected to kinase assays with activated GST-SAPK3 and 3µCi [γ-32P]-ATP. Figure 5.5 shows a representative autoradiograph from this experiment with

phosphorylated protein bands in the reactions where Bcl-2, Bag-1, BimEL and Bad were

immunoprecipitated. Only weak phosphorylation of Blk and no phosphorylation of Bid

was detected. No bands in the size range of 15 – 35 kDa as expected for the sizes of the

Bcl-2 family members were detected in the control reactions, where only empty

Bluescript vector or p35 DNA were transfected into cells. These results suggest that a

number of Bcl-2 family members can be phosphorylated by SAPK3, but clearly these

protocols are limited by the problems associated with expressing sufficient amounts of

these proteins to act as substrates. Therefore, as outlined in the following sections,

additional mitochondrial substrates for SAPK3 were evaluated.

5.2.3 Investigating the phosphorylation of the mitochondrial protein Sab by

SAPK3

JNK, a well-characterised SAPK, has been recently shown to localise to the

mitochondria and phosphorylate a mitochondrial protein, Sab (Wiltshire et al., 2002).

Due to the localisation of SAPK3 also at the mitochondria (Chapter 3), the ability of

SAPK3 to phosphorylate Sab was assessed. Using activated GST-SAPK3, in vitro

kinase assays were performed to compare the phosphorylation of GST-Sab with the

known in vitro SAPK3 substrates, GST-ATF2 and GST-α1-Syntrophin. GST-ATF2 has

been shown to be an in vitro substrate for both JNK and SAPK3 (Gupta et al., 1995;

89

Page 154: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

BS Bcl-2 Bag-1 Bim Bad Blk Bid p35Transfection:

30 kDa

20.1 kDa

220 kDa

97.5 kDa66 kDa

45 kDa

Figure 5.5: SAPK3 can phosphorylate mitochondrial proteins involved in apoptosis.HEK293 fibroblasts (3 dishes of 1 x 105 cells per construct) were transfected with 1 µg of the expression constructs Flag-Bcl-2 or Flag-Bag-1 (both in conjunction with 1 µg empty Bluescript vector) or 1 µg HA-Bim, HA-Bad, HA-Blk or HA-Bid (all in conjunction with 1 µg of the caspase inhibitor p35) or controls (2 µg p35 or Bluescript DNA only) and allowed to express the proteins for 40 hours. The cells were then lysed, and HA or Flag tag antibodies added. Protein G-Sepharose was then added to capture the antigen-antibody complex, and immunoprecipitates washed extensively. These immunoprecipitates were then subjected to kinase assays with activated GST-SAPK3 and 3µCi [γ-32P]-ATP. The kinase assay components were then separated using SDS-PAGE and the gel exposed to film. A representative autoradiograph is shown from one of 3 identical experiments. The white arrows show the phosphorylated protein bands; little Blk and no Bid phosphorylation was detected.

89a

Page 155: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Cuenda et al., 1997). In addition, the phosphorylation of the protein c-Jun (a good JNK

substrate (Hibi et al., 1993)), by SAPK3 was determined.

As shown in Figure 5.6, Sab produced as a recombinant fusion protein in E.

coli was phosphorylated by SAPK3 in vitro. The incorporation of 32P was higher than

that of the incorporation of 32P into ATF2 and neared that of the incorporation into α1-

Syntrophin. It is possible that the higher level of 32P incorporation detected for α1-

Syntrophin was due to phosphorylation occurring on two sites (Ser193 and Ser201)

(Hasegawa et al., 1999). In contrast, c-Jun with at least two sites available for

phosphorylation by MAPKs or SAPKs (Ser63 and Ser73; (Smeal et al., 1991; Pulverer

et al., 1993; Hibi et al., 1993)), was a poor substrate for SAPK3.

To determine whether members of the JNK subfamily might share similar

substrate preferences, a second series of in vitro kinase assays was performed with

activated JNK2. In contrast to SAPK3, JNK2 phosphorylated c-Jun to the greatest

extent, followed by Sab. ATF2 was less favoured, and α1-Syntrophin was a poor

substrate (Figure 5.7). Again, due to the phosphorylation of c-Jun on two sites by JNK,

this could account for the higher incorporation of 32P (Hibi et al., 1993). These results

show that although SAPK3 and JNK share Sab as a substrate, they have differing

preferences for other substrates. In contrast, ERK2, a MAPK preferentially activated by

growth factors and other mitogenic stimuli, did not phosphorylate Sab, but demonstrated

high activity towards a previously characterised substrate, Elk-1 (Gille et al., 1995)

(Figure 5.8).

5.2.4 Loss of the first Kinase Interaction Motif (KIM) of Sab prevents its

phosphorylation by SAPK3

To determine the mechanism for interaction of SAPK3 with Sab, a peptide

representing the last 13 amino acids of the rat SAPK3 sequence, Arg-Gln-Leu-Gly-Ala-

Arg-Val-Pro-Lys-Glu-Thr-Ala-Leu-OH was used. This C-terminal tail of SAPK3 has

90

Page 156: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

MalE-MKK6(DD) - + - + - + - + GST-SAPK3 + + + + + + + + GST-Substrate: ATF2 α1-Syn Sab c-Jun

Autoradiograph

97 kDa66 kDa

45 kDa

GST-ATF2

GST-Sab

GST-c-Jun

GST-α1-Syntrophin

GST-ATF2

GST-Sab

GST-c-Jun

GST-α1-Syntrophin97 kDa66 kDa

45 kDaCoomassie

- + - + - + - + + + + + + + + +

MalE-MKK6(DD):GST-SAPK3:

0

20

40

60

80

100

120

ATF2 α1-Syn Sab c-Jun

Subs

trat

e Ph

osph

oryl

atio

n by

SA

PK3

(% c

ompa

red

to α

1-Sy

ntro

phin

)

Figure 5.6: Sab is a substrate of SAPK3 in vitro.GST-SAPK3 was either incubated with MalE-MKK6(DD) and ATP, or ATP only, at 30°C overnight. GST-SAPK3 was separated from the other reaction components, and subjected to kinase assays with GST-ATF2(19-96), GST-α1-syntrophin, GST-Sab(219-425) or GST-c-Jun(1-135). An autoradiograph of the gel is shown after 2.5 hours exposure. The middle panel shows the Coomassie stained gel to illustrate even protein loading, and the lower panel shows the quantitation of 3 independent assays as determined by Cerenkov counting of the protein substrate bands. In the case of GST-α1-syntrophin, only the upper band showing 32P incorporation was quantitated. The results are shown as means ± SE.

90a

Page 157: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Autoradiograph

Coomassie

GST-ATF2

GST-Sab

GST-c-Jun

GST-α1-Syntrophin

GST-α1-Syntrophin

GST-MKK4(ED) + + + + GST-JNK2 + + + + GST-Substrate: ATF2 α1-Syn Sab c-Jun

97 kDa

66 kDa

45 kDa

97 kDa

66 kDa

45 kDaGST-Sab

GST-c-Jun

GST-ATF2

60

80

100

Phos

phor

ylat

ion

of s

ubst

rate

by

JN

K2

(% o

f c-J

un)

ATF2 α1-Syntrophin c-JunSab0

20

40

Figure 5.7: GST-JNK2 shows different substrate preferences to GST-SAPK3 in vitro.GST-JNK was activated by MKK4(ED) and subjected to kinase assays with GST-ATF2(19-96), GST-α1-Syntrophin, GST-Sab(219-425) or GST-c-Jun(1-135). An autoradiograph of the gel is shown after 3.75 hours exposure. The middle panel shows the Coomassie stained gel to illustrate even protein loading, and the lower panel shows the quantitation of 4 independent assays as determined by Cerenkov counting of the protein substrate bands. The results are shown as means ± SE.

90b

Page 158: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

GST-Sab

GST-Elk

GST-Elk

45 kDa

GST-Sab

GST-MKK1(EE) + +GST-ERK2 + +GST-Sab + -GST-Elk - +

Autoradiograph

Coomassie

45 kDa

Figure 5.8: GST-ERK2 does not phosphorylate Sab in vitro.GST-ERK2 was activated by MKK1(EE) and subjected to kinase assays with GST-Sab(219-425) or GST-Elk1(307-428). An autoradiograph of the gel is shown after 3.75 hours exposure. The lower panel shows the Coomassie stained gel to illustrate even protein loading.

90c

Page 159: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

previously been shown to be crucial for the interaction of SAPK3 with the PDZ domain

of the in vitro substrate, α1-Syntrophin (Hasegawa et al., 1999). Specifically, a C-

terminal truncation mutant of SAPK3 could not phosphorylate α1-Syntrophin

(Hasegawa et al., 1999). As shown in Figure 5.9, results consistent with this model were

seen when the C-terminal SAPK3 peptide blocked the phosphorylation of α1-Syntrophin

by SAPK3. However this peptide had no effect on the phosphorylation of Sab by

SAPK3. This suggests that SAPK3 uses different mechanisms to recognise Sab and α1-

Syntrophin as substrates, such that Sab recognition is independent of the C-terminal

motif of SAPK3. Although similar experiments were not performed with Bcl-2 family

members, it would be predicted that the phosphorylation of these proteins (Bcl-2, Bag-1,

Bim and Bad) would also be unaffected by this peptide because these proteins have no

recognisable PDZ domain sequence.

An alternative interaction motif, the KIM, can mediate interactions in the

MAPK pathway. The consensus sequence for a KIM is R/K(X)3-5LXL (Yang et al.,

1998b; Chang et al., 2002; Sharrocks et al., 2000). There are two KIMs in the human

Sab sequence, located at positions 313-319 (KIM1; sequence RPGSLDL), and 400-406

(KIM2; sequence RMKQLSL) (Wiltshire et al., 2002), where the residues in bold

conform to the KIM consensus sequence. Each KIM has been selectively mutated to

remove the hydrophobic residues that mediate the interaction with MAPKs (Wiltshire et

al., 2002). This strategy has shown that JNK interacts with Sab using the first of these

KIMs (i.e. KIM1 at positions 313-319) identified in the Sab sequence, so that the loss of

KIM1 prevents the phosphorylation of Sab by JNK (Wiltshire et al., 2002). These KIM

mutants are referred to as the –KIM1 mutant (where KIM1 is mutated), -KIM2 mutant

(where KIM2 is mutated) and the –KIM1/2 mutant (where both KIMs are mutated), and

are illustrated in Figure 5.10A.

To identify whether SAPK3 interacts with Sab using either of these KIMs,

the -KIM1, -KIM2 and -KIM1/2 mutants of Sab were subjected to in vitro kinase assays

with activated SAPK3. As shown in Figure 5.10B, phosphorylation of Sab by SAPK3

was greatly reduced when KIM1 was mutated, however there was a minimal effect on

91

Page 160: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

GST-α1-SyntrophinGST-Sab

Peptide (µM): - 10 100 - 10 100

Autoradiograph

97 kDa

66 kDa

97 kDa

66 kDaGST-Sab

GST-α1-Syntrophin

GST-Sab

GST-α1-SyntrophinCoomassie

No Peptide 100µM10µM

(* p<0.01 compared to No Peptide)

0

20

40

60

80

100

120

Subs

trat

e Ph

osph

oryl

atio

n by

SA

PK3

(%)

Sabα1-Syntrophin

*

*

Figure 5.9: A C-terminal SAPK3 peptide inhibits the phosphorylation of α1-Syntrophin but not Sab, by SAPK3.GST-Sab(219-425) and GST-α1-Syntrophin were pre-incubated with various concentrations of SAPK3 C-terminal peptide at 30°C for 10 min, then subjected to kinase assays with activated GST-SAPK3 and radiolabelled ATP. An autoradiograph of the gel is shown after 2.5 hours exposure time. The middle panel shows the Coomassie stained gel to illustrate even protein loading, and the lower panel shows the quantitation of 3 independent assays as determined by Cerenkov counting of the GST-Sab and GST-α1-Syntrophin bands. The results are shown as means ± SE. Asterisks denote significant differences (p<0.01) compared to reactions where no peptide was added.

91a

Page 161: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

AS391S346S301 S321

KIM1(313-319)

KIM2(400-406)

1 425

RPGSLDL RMKQLSL

RPGSADA RMKQASA

Modified from Wiltshire et al., Biochem J. 367: 577-585 (2002)

B MalE-MKK6(DD): + + + + + + + +GST-SAPK3:

-KIM2WT -KIM1 -KIM1/2GST-Sab:66 kDa

45 kDa

66 kDa

45 kDa

Coomassie

Autoradiograph GST-Sab

GST-Sab

- --0

20

40

60

80

100

120

wild-type KIM1 KIM2 KIM1/2

Phos

phor

ylat

ion

of S

ab

(% o

f wild

-type

)

**

*

(* p<0.01 compared to wild-type)

Figure 5.10: Loss of the first Kinase Interaction Motif of Sab prevents its phosphorylation by SAPK3.(A) A schematic diagram of the human Sab protein, showing the positions of the MAPK consensus sites and KIM-like sequences. The mutations of the essential hydrophobic residues in the KIM sequences to alanine residues created the –KIM1, -KIM2, and –KIM1/2 mutants. (B) GST-SAPK3 was activated by MalE-MKK6(DD), and subjected to kinase assays with wild-type GST-Sab(219-425), GST-Sab (-KIM1 mutant), GST-Sab (-KIM2 mutant) or GST-Sab (-KIM1/2 mutant). An autoradiograph of a representative gel is shown after 1.5 hours exposure. The middle panel shows the Coomassie stained gel to illustrate even protein loading, and the lower panel shows the quantitation of 3 independent assays as determined by Cerenkov counting of the GST-Sab bands. The results are shown as means ± SE. Asterisks denote significant difference (p<0.01) compared to wild-type GST-Sab.

91b

Page 162: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

SAPK3 phosphorylation of Sab when KIM2 was mutated (approximately 5% decrease

compared to wild-type), indicating the requirement for KIM1 when Sab is

phosphorylated by SAPK3. Therefore KIM1 appears to be required when Sab is

phosphorylated by JNK or SAPK3.

5.2.5 Serine 321 in Sab is the major site of phosphorylation by SAPK3 and JNK2

Wild-type Sab has four possible MAPK phosphorylation sites as shown:

Site 1: Ser301 (298GPTSPSE304)

Site 2: Ser321 (318DLPSPVS324)

Site 3: Ser346 (343GASSPEC349)

Site 4: Ser391 (388SSTSPEG394).

The Predikin program (Brinkworth et al., 2003) was used to predict which

of these sites within the Sab sequence could be phosphorylated by SAPK3. Predikin uses

structural information on kinase-substrate interactions to more precisely define a

consensus site of phosphorylation by a serine/threonine protein kinase (Brinkworth et

al., 2003). According to Predikin analysis, the optimal consensus sequence for

phosphorylation of a substrate by SAPK3 is:

(X)-3 (P/V/A/L/S) -2 (F/M/L) -1 (S/T)0 P+1 (F/Xa) +2 (R/K/Q/S/L) +3,

where X is any amino acid and Xa is any aliphatic amino acid. The italicised amino

acids denote the phosphorylated amino acids, and all amino acids are thus numbered

from –3 to +3 relative to that position.

Using this approach, it was predicted that serine 321 in the sequence

318DLPSPVS324 was the most likely site for phosphorylation of Sab by SAPK3 with six

of the seven amino acids in the sequence agreeing with the Predikin prediction. The

serine residue at position 321 was therefore mutated to an alanine residue. In addition,

as a “control” mutation, serine 391 in the sequence 388SSTSPEG394 was mutated because

this was predicted to be a more unlikely site for phosphorylation with only four of the

seven amino acids agreeing with the Predikin prediction. It should be noted that the

92

Page 163: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

sequences surrounding the other two possible MAPK sites (298GPTSPSE304 and

343GASSPEC349) also do not closely follow the Predikin consensus sequence. Most

notably these sites, like that of the serine 391 site, have negatively charged residues at

the +2 or +3 positions in the site when Predikin predicts that preferred SAPK3 substrates

will have aliphatic or basic residues in these positions.

As seen in Figure 5.11, when serine 321 was mutated to alanine (S321A),

this greatly reduced the phosphorylation of Sab by SAPK3, to approximately 15% of the

wild-type level. This would suggest that serine 321 is the major site of phosphorylation

of Sab by SAPK3. When serine 391 was mutated to alanine (S391A), there was only a

marginal (although significant) decrease in phosphorylation of Sab to approximately

85% of wild-type level. This would suggest that serine 391 can be phosphorylated by

SAPK3, but to a much lesser extent. When these Sab mutants were incubated with

activated JNK (Figure 5.12), similar results were seen however when Ser391 was

mutated to alanine the overall level of phosphorylation did not change, suggesting that

JNK2 did not phosphorylate Sab on this site. Therefore serine 321, which is only 1 of 4

putative MAPK consensus sites, appears to be the major phosphorylation site in Sab for

both SAPK3 and JNK.

5.2.6 Surrounding sequence composition and location dictate the favoring of Ser

321 in Sab

To determine how Ser321 phosphorylation was favoured by SAPK3, a

series of mutations was conducted around this site. First, to assess whether the position

of Ser321 within the Sab protein, or the sequence surrounding serine 321, was favoured

by SAPK3, mutations were introduced into wild-type Sab or the S321A Sab mutant, to

“swap” the sequences around Ser321 and Ser391. This created the 321/391 mutant (with

a site surrounding Ser321 changed to the sequence of the original Ser391) or the

391/321 mutant (with a site surrounding Ser391 changed to a sequence like that

surrounding the original Ser321). In the latter case of the 391/321 mutant, this was

created in a Sab S321A mutant so that SAPK3 would no longer be able to phosphorylate

93

Page 164: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

MalE-MKK6(DD): + + ++ + +GST-SAPK3:

GST-Sab

GST-Sab

45 kDa

66 kDa

45 kDa

66 kDa

GST-Sab: S391AWT S321A

Autoradiograph

Coomassie

(* p<0.01 compared to wild-type)

*

*

0

20

40

60

80

100

120

WT S321A S391ASab

Phos

phor

ylat

ion

by S

APK

3 (%

)

Figure 5.11: A Ser321Ala mutant of Sab is not phosphorylated by SAPK3.GST-SAPK3 was activated by MalE-MKK6(DD) and subjected to kinase assays with wild-type GST-Sab(219-425) or mutants where serine has been changed to alanine at position 321 (S321A) or 391 (S391A). An autoradiograph of a representative gel is shown after 1.5 hours exposure. The middle panel shows the Coomassie stained gel to illustrate even protein loading, and the lower panel shows the quantitation of 4 independent assays as determined by Cerenkov counting of the GST-Sab bands. The results are shown as means ± SE. Asterisks denote a significant difference (p<0.01) compared to the phosphorylation of wild-type GST-Sab.

93a

Page 165: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

GST-MKK4(ED): + + ++ + +GST-JNK2:

GST-Sab: S391AWT S321A

GST-Sab

GST-Sab

45 kDa

66 kDa

45 kDa

66 kDa

(* p<0.01 compared to wild-type)

*

0

20

40

60

80

100

120

WT S321A S391ASab

Phos

phor

ylat

ion

by J

NK

2 (%

)

Figure 5.12: A Ser321Ala mutant of Sab is not phosphorylated by JNK2.GST-JNK2 was activated by GST-MKK4(ED) and subjected to kinase assays with wild-type GST-Sab(219-425) or mutants where serine has been changed to alanine at position 321 (S321A) or 391 (S391A). An autoradiograph of a representative gel is shown after 4 hours exposure. The middle panel shows the Coomassie stained gel to illustrate even protein loading, and the lower panel shows the quantitation of 4 independent assays as determined by Cerenkov counting of the GST-Sab bands. The results are shown as means ± SE. Asterisks denote a significant difference (p<0.01) from the phosphorylation of wild-type GST-Sab.

93b

Page 166: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

the S321 in its wild-type position, and therefore would not interfere with the

interpretation of the results of this analysis. These sequences have been described in

Appendix I of this thesis.

As shown in Figure 5.13, the 321/391 mutant (i.e. with the sequence around

Ser321 swapped to that usually surrounding Ser391, in the wild-type background)

reduced the phosphorylation of Ser321 to approximately 40% of the level of wild-type

phosphorylation. The 391/321 mutant (i.e. the sequence around Ser391 swapped to that

usually surrounding Ser321, in the Sab S321A mutant background) showed a similar

decrease although its phosphorylation did remain significantly higher than that of the

S321A Sab mutant. This result with the 391/321 mutant suggests that the position of the

phosphorylation site within the Sab sequence was a dominant contributor to its ability to

be phosphorylated by SAPK3. This conclusion was reached since altering the sequence

surrounding Ser391 did not bring the level of Ser391 phosphorylation to that normally

seen for Ser321. However, because altering the sequence composition around Ser391

still slightly improved Sab phosphorylation, the surrounding sequence was likely still of

importance at this site.

Whilst one interpretation of the data on the phosphorylation of the 321/391

mutant is that the Ser391 sequence context is not able to be phosphorylated even when

in the appropriate position in the Sab sequence, this is confounded by the close

proximity of the Ser321 phosphorylation site to KIM1 in the Sab sequence. Examination

of the sequence of Sab shows that the residue at the –2 position of the Ser321 site is

actually the second essential hydrophobic residue in KIM1 as shown in bold in the

following sequence 313RPGSLDLPSPVS324. Thus, the 321/391 mutant may no longer be

phosphorylated only because the integrity of KIM1 has been disrupted by the loss of 1 of

the 2 essential hydrophobic residues.

To more carefully examine the effects of changing the Ser321 site without

altering KIM1, mutations were introduced to only change amino acids 323 (i.e. +2

position in the phosphorylation sequence) and 324 (i.e. +3 position in the

94

Page 167: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

45 kDa

66 kDa

45 kDa

66 kDa

GST-Sab

WT 321/ S321A 391/391 321

+ + + + + + + +

MalE-MKK6(DD):GST-SAPK3:GST-Sab:

GST-SabAutoradiograph

Coomassie

*

(* p<0.001 compared to wild-type; # p<0.001 compared to S321A)

#

0

20

40

60

80

100

120

WT 321/391 S321A 391/321

Phos

phor

ylat

ion

of S

ab

by S

APK

3 (%

of W

ild-T

ype)

Figure 5.13: The phosphorylation of Sab by SAPK3 is dependent onthe surrounding sequence and the position of the site within Sab.GST-SAPK3 was activated by MalE-MKK6(DD) and subjected to kinase assays with wild-type GST-Sab(219-425), Sab (321/391) mutant, S321A mutant, or Sab (391/321) mutant. An autoradiograph of a representative gel is shown after 1.5 hours exposure. The middle panel shows the Coomassie stained gel to illustrate even protein loading, and the lower panel shows the quantitation of 3 independent assays as determined by Cerenkov counting of the GST-Sab bands. The results are shown as means ± SE.

94a

Page 168: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

phosphorylation sequence) in the Sab sequence. Normally, a Val residue is present at

position 323 in the Sab sequence, and a Ser residue at position 324. Site-directed

mutagenesis was used to change Val323 to Glu or Ala, or Ser324 to Glu or Ala. These

sequences have been described in Appendix I. Changing these amino acids to Glu more

closely resembled the sequences surrounding the other putative MAPK consensus sites

in the Sab sequence. The replacement with Ala further defined the sequence

requirements at the +2 and +3 position of this site in Sab.

As seen in Figure 5.14, the V323E mutant in which Glu was introduced

into the +2 position of the Ser321 site decreased phosphorylation of Sab by SAPK3 by

approximately 75%. Introduction of Ala at this position (V323A) also affected

phosphorylation, albeit to a lesser extent. The S324E mutant in which Glu was

introduced into the +3 position of the Ser321 site only decreased the phosphorylation of

Sab by SAPK3 by approximately 15%, and the introduction of Ala here (S324A) had no

detrimental effect on the ability of Sab to be phosphorylated by SAPK3, but rather

improved it by approximately 10%. Collectively, these results suggest that both the

position of the phosphorylation site within the target protein and the exact sequence

surrounding the phosphorylation site are important for the recognition of the Sab

phosphorylation site by SAPK3. It appears that the KIM sequence and the amino acid at

the +2 position of the phosphorylation site are crucial for phosphorylation of Sab at Ser

321 by SAPK3.

5.3 Discussion

When compared to the archetypical p38 MAPKs, SAPK2a and SAPK2b,

SAPK3 is a relatively poorly studied member of the p38 MAPK family. Little is known

about its activation, localisation, or substrate specificity, apart from studies in which

SAPK3 has been overexpressed in mammalian cells (Buee-Scherrer and Goedert, 2002;

Takahashi et al., 2003; Sakabe et al., 2002; Chiariello et al., 2000; Cuenda et al., 1997;

Kumar et al., 1997). In Chapter 3 SAPK3 was shown to co-localise with mitochondria.

95

Page 169: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

45 kDa

66 kDa

45 kDa

66 kDa

MalE-MKK6(DD):GST-SAPK3:

GST-Sab:

+ + + + + + + + + +

Autoradiograph

GST-Sab

WTV32

3EV32

3AS32

4ES32

4A

GST-Sab

Coomassie

*

**

*

(* p<0.01 compared to wild-type)

0

20

40

60

80

100

120

140

WT V323E V323A S324E S324APhos

phor

ylat

ion

of S

ubst

rate

by

SA

PK3

(% o

f WT)

Figure 5.14: Analysis of sequence determinants for phosphorylation at the +2 and +3 positions of Ser321. GST-SAPK3 was activated by MalE-MKK6(DD) and subjected to kinase assays with wild-type GST-Sab (219-425), GST-Sab V323E mutant, GST-Sab V323A mutant, GST-Sab S324E mutant, or GST-Sab S324A mutant. An autoradiograph of a representative gel is shown after 3.5 hours exposure time. The middle panel shows the Coomassiestained gel to illustrate even protein loading, and the lower panel shows the quantitation of 3 independent assays as determined by Cerenkov counting of the GST-Sab bands. The results are shown as means ± SE.

95a

Page 170: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Importantly, mitochondrial co-localisation suggests that there will be mitochondrial

substrates for SAPK3. This Chapter therefore addressed the phosphorylation of

mitochondrial proteins by SAPK3.

Following the activation of recombinant SAPK3 in vitro, it was shown that

SAPK3 phosphorylated Bcl-2, Bag-1, BimEL and Bad (Figure 5.5). This potentially

implicates SAPK3 in a role at the mitochondria alongside other members of the MAPK

family such as ERK and JNK, which have previously been shown to phosphorylate

proteins localised at this organelle (Ishikawa et al., 2003; Donovan et al., 2002; Ley et

al., 2003; Deng et al., 2001). For example, JNK phosphorylates the anti-apoptotic

proteins Bcl-2 and Bclx(L) and the pro-apoptotic proteins Bad and Bim (Fan et al.,

2000; Basu and Haldar, 2003; Donovan et al., 2002; Lei and Davis, 2003). These

actions of JNK remain somewhat controversial, and JNK has been suggested to act in a

pro-and anti-apoptotic fashion in many cell types via its actions on proteins at the

mitochondria (Bhakar et al., 2003; Donovan et al., 2002; Basu and Haldar, 2003; Lei and

Davis, 2003; Lei et al., 2002; Yu et al., 2004). It remains possible that SAPK3 could

play similar roles in the regulation of mitochondrial proteins. One report has suggested

that SAPK3 could play a pro-apoptotic role in the ovarian carcinoma cell line OVCAR3

(Abdollahi et al., 2003). However, most studies to date using overexpressed SAPK3

have not implicated this kinase in apoptosis. Rather SAPK3 has been suggested to play

diverse roles such as regulation of gene transcription (Marinissen et al., 1999), γ-

radiation-induced cell cycle arrest (Wang et al., 2000), cell differentiation (Lechner et

al., 1996), meiotic G(2)/M progression of Xenopus oocytes (Perdiguero et al., 2003), and

the regulation of glucose uptake (Ho et al., 2004).

In this Chapter, SAPK3 was shown to phosphorylate the pro- and anti-

apoptotic mitochondrial proteins to different extents. This may be due to differences in

substrate preference, differences in expression levels of the substrate proteins, or

different numbers of phosphorylation sites within each substrate protein. It is also

possible that each of the proteins was immunoprecipitated to different extents. This

seems likely when examining the Western blot results, which showed that Bid and Bad

96

Page 171: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

did not show high immunoreactivity in the pellet fraction of the immunoprecipitations

(Figure 5.4). It may be possible that the HA tag is obscured by the folding of these

particular proteins, resulting in lower recognition by the HA antibody. Furthermore, it

seems likely that some of the immunoprecipitated Bid and Bad would be difficult to

visualise since they migrate to approximately the same point on SDS-PAGE as light

chain immunoglobulin, and so would be obscured by this band on the Western blot

(Figure 5.4).

The migration of the mitochondrial proteins on SDS-PAGE largely did not

seem to agree with the calculated molecular weight of these proteins (Figures 5.3 and

5.4). It is unknown why this discrepancy occurs, but possible reasons include the

presence of a HA or Flag tag (both of which are approximately 1 kDa), or post-

translational modifications such as myristylation, palmitylation, farnesylation,

glycosylation or phosphorylation, although apart from phosphorylation these

modifications of this family of proteins has not been reported. The difficulties in

expression and analysis of these proteins in mammalian cells highlights that alternative

methods for substrate preparation are desirable. Expression of Bcl-2 family members in

bacterial systems has not been successful due to instability, insolubility, and low yields

of these proteins (D. Huang, personal communication). Future protocols could therefore

explore their expression in insect cell systems, yeast, or cell-free systems.

In addition to showing that SAPK3 could phosphorylate a variety of pro-

and anti-apoptotic proteins at the mitochondria, it was shown that SAPK3

phosphorylates the mitochondrial protein Sab (Figure 5.6). This protein first described as

a binding partner of Bruton’s Tyrosine Kinase (Btk) (Matsushita et al., 1998; Yamadori

et al., 1999), has more recently been shown to be an in vitro substrate of the JNK family

of SAPKs (Wiltshire et al., 2002). The advantage in the study of the phosphorylation of

Sab was the ability to express sufficient quantities of the recombinant protein as a GST-

fusion protein in E.coli. Furthermore, wild-type and mutants of GST-Sab could be

evaluated for their phosphorylation by SAPK3 in subsequent in vitro assays. In this

Chapter it has been shown that Sab is an in vitro substrate shared by SAPK3 and JNK2

97

Page 172: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

(Figure 5.7). A survey of the literature shows that other substrates such as Elk-1 are also

phosphorylated by SAPK3 and JNK in vitro (Cavigelli et al., 1995; Cuenda et al., 1997),

but notably JNK has been found to phosphorylate substrates such as c-Jun (Hibi et al.,

1993) that are not phosphorylated by SAPK3 (Cuenda et al., 1997). Conversely the

substrate Tau is phosphorylated by SAPK3 but acts as a poor substrate for JNK (Buee-

Scherrer and Goedert, 2002).

The sharing of substrates by multiple closely-related but not identical

protein kinases such as the different SAPKs could provide a number of important levels

of control in these intracellular signaling pathways. When different stimuli preferentially

activate either SAPK3 or JNK2, it is possible that the phosphorylation of the same

substrates can result. This allows diverse stimuli to initiate the same intracellular action.

This is similar to the phosphorylation of Mnk1 and Mnk2 by both ERK and SAPK2a,

which stimulates the kinase activity of Mnk1/2 and results in the phosphorylation of the

eukaryotic initiation factor-4E (eIF-4E) (Waskiewicz et al., 1997). Alternatively,

substrate activation may require the phosphorylation of one site by SAPK3 and another

site by JNK2. A similar situation appears to occur in the phosphorylation of JNK by

MKK4 and MKK7, where JNK is preferentially phosphorylated on Thr183 by MKK7,

and on Tyr185 by MKK4, and thus the two MKKs can operate synergistically to activate

JNK (Lawler et al., 1998; Fleming et al., 2000). It also remains possible that SAPK3

and JNK2 each phosphorylate some common substrates but independently and at

different sites. This may have different functional consequences for the actions of the

phosphorylated protein and could lead to different cellular responses. For example, it has

been reported that JNK phosphorylation of c-Jun occurs on the N-terminal sites of Ser63

and Ser73, promoting the activity of c-Jun, whereas ERK phosphorylation of c-Jun

occurs at an inhibitory C-terminal site (Minden et al., 1994).

In the case of Sab, serine 321 was identified as the major site of

phosphorylation by SAPK3 and JNK2 (Figure 5.11 and 5.12). The identification of the

same phosphorylation site for both kinases was not surprising, given that SAPK3 and

JNK interact with Sab using the same KIM (Figure 5.10), and thus would be expected to

98

Page 173: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

come into contact with a similar region of the Sab protein for phosphorylation.

Interestingly, SAPK3 was also shown to phosphorylate serine 391, albeit to a much

lesser extent than serine 321. However, mutation of serine 391 in Sab did not result in

any loss of phosphorylation by JNK2, showing that whilst SAPK3 can phosphorylate

both Ser321 and Ser391 in Sab, JNK2 appears unable to phosphorylate Ser391. Since

mutation of Ser321 to alanine did not completely abolish phosphorylation of Sab by

JNK2 however, it seems that besides Ser321, JNK2 can also phosphorylate another site

within Sab apart from Ser321 and Ser391. Therefore, JNK2 and SAPK3 appear to share

subtly different site preferences within the Sab protein. The phosphorylation of Sab by

SAPK3 or JNK2 may therefore result in different outcomes.

When analysing the possible phosphorylation sites within any one

substrate, consensus sequences for phosphorylation have been useful to help define

which serine or threonine residues might be phosphorylated by kinases such as the

MAPKs or SAPKs. As noted earlier, the consensus sequence for MAPKs has been

defined as (Pro/X)-X-(Ser/Thr)-Pro, where X is any amino acid and the amino acid

phosphorylated is indicated in italics (Clark-Lewis et al., 1991). The identification of

this consensus relied on the design of synthetic peptides based on the MAPK

phosphorylation site of myelin basic protein, with mutations at various positions (Clark-

Lewis et al., 1991). Kinetic studies were then used to determine the sequence which had

the optimal rate of phosphorylation (Clark-Lewis et al., 1991). Evaluating the sequences

surrounding the sites of phosphorylation by SAPK3 (eg α1-Syntrophin 190PPASPLQ196

and 198QPSSPGP204; tau 393VYKSPVV399 and 401GDTSPRH407) has suggested this loose

consensus also holds true for SAPK3. However this simple comparison with only a

limited number of SAPK3 substrates does not allow the more precise prediction of

SAPK3 substrate preferences. A recent advance in the prediction of substrate

phosphorylation sites comes with release of the Predikin program (Brinkworth et al.,

2003). This uses structural information derived from the interactions of Ser/Thr protein

kinases with their substrates to define more closely the preferred sites for

phosphorylation for any Ser/Thr protein kinase.

99

Page 174: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

For SAPK3 the optimal consensus sequence is predicted to be:

X-3 (P/V/A/L/S) -2 (F/M/L) -1 (S/T)0 P+1 (F/Xa) +2 (R/K/Q/S/L) +3

where X is any amino acid, Xa is an aliphatic amino acid, and the italicised amino acid

(Ser or Thr) can be phosphorylated.

Similar analysis for JNK2 predicts its consensus sequence to be:

(L/I/M/F)-3 (E/D/S/R)-2 (F/M/L)-1 (S/T)0 P+1 (P/F/L/I/D/E)+2 (R/K/Q/S/L)+3.

This confirms the suggestion that these two SAPKs may not always share similar

preferences for substrate phosphorylation sites due to major differences in the residue

preferred at the –2 position, some differences in the +2 position, and a greater

requirement for specific residues in the –3 position when phosphorylation by JNK2 is

considered.

It is interesting to note that under some circumstances active ERK2 has

been found to localise to the mitochondria (Zhu et al., 2003). The Predikin consensus

site for phosphorylation by ERK2 is predicted to be:

(R/K/F)-3 (P/V/A/L/S)-2 (F/M/L)-1 (S/T)0 P+1 (F/XA)+2 (R/K/Q/S/L)+3.

This is very similar to the consensus site for SAPK3, with the only difference being a

more defined prediction for the –3 residue in the phosphorylation site. According to this

analysis, it was predicted that there would be more likelihood that a substrate would be

shared by ERK2 and SAPK3 than would be shared by JNK2 and SAPK3. In contrast it

was shown that Sab is phosphorylated by SAPK3 and JNK2, and that ERK2 was unable

to phosphorylate Sab (Figure 5.8). Therefore, it is likely that other differences in the

mechanisms of substrate recognition by ERK2 and SAPK3/JNK2 account for these

differences.

Other domain interactions in the recognition and phosphorylation of

substrates by SAPK3 will likely add these additional layers of complexity. It has

previously been shown than the C-terminal sequence of SAPK3, KETXL, is essential for

the interaction of SAPK3 with the PDZ domain of α1-Syntrophin, and the subsequent

phosphorylation of this protein (Hasegawa et al., 1999). In Chapter 6 it is shown that

SAPK3 can use this C-terminal tail to interact with the PDZ domains of other proteins

100

Page 175: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

such as Scribble and OMP25. A cell-permeable version of this C-terminal peptide has

recently been used as an intracellular SAPK3 inhibitor (Sabio et al., 2004). However, in

this Chapter it was shown that SAPK3 phosphorylates Sab via a mechanism that uses

KIM1, located at position 313-319 in Sab (Figure 5.10). For Sab, the C-terminal peptide

does not inhibit phosphorylation by SAPK3 (Figure 5.9). Therefore, the use of the cell-

permeable C-terminal peptide will only inhibit a subset of SAPK3 substrate

phosphorylation. An analysis of the literature suggests that this is the first reported

confirmation of the phosphorylation of a substrate by SAPK3 using a KIM motif. As

described in more detail in the following paragraph, this implies that SAPK3 can, like

other kinases, use this mechanism for binding to its substrates.

When comparing the sequences of Sab to other substrates of SAPK3, such

as ATF2 (Cuenda et al., 1997) and Tau (Goedert et al., 1997b) KIM-like sequences

(R/K(X)3-5LXL) can be identified in both of these other substrates. Specifically,

47KKMPLDL53 in ATF2 has been previously implicated as a KIM (Livingstone et al.,

1995; Gupta et al., 1995) and 221REPKKVAV228 in Tau appears to resemble the KIM

sequence. When examining the sequences of some of the mitochondrial proteins found

to be phosphorylated by SAPK3 in Figure 5.5, Bcl-2, Bag-1 and Bim all have sequences

resembling KIMs (Bcl-2 218KTLLSLAL225; Bag-1 345RLQSTNLAL353; Bim

180RMVILRL186). α1-Syntrophin also contains two KIM-like sequences (9RTGLLEL15

and 497KVTRLGL503). As mentioned earlier, previous studies have found that SAPK3

interacts with α1-Syntrophin using its PDZ domain-binding motif (Hasegawa et al.,

1999). The data showing that the C-terminal SAPK3 peptide blocks the phosphorylation

of α1-Syntrophin supports the idea that the most significant interaction between SAPK3

and α1-Syntrophin is via the PDZ domain, and that this domain is required for

phosphorylation.

In conclusion, the results obtained in this Chapter suggest that SAPK3 is

able to phosphorylate mitochondrial substrates such as Bcl-2, Bag-1, BimEL, Bad and

Sab. Thus, SAPK3 and JNK may share common targets at the mitochondria. In

addition, some of the determinants responsible for phosphorylation site preference by

101

Page 176: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

SAPK3 have been identified. The results presented here show that the sequence

surrounding the phosphorylation site of a substrate, as well as the position of that site

within the entire substrate protein is important for recognition by SAPK3. This

information should prove helpful in the future identification of other potential SAPK3

substrate proteins. The localisation of SAPK3 at the mitochondria, and its

phosphorylation of mitochondrial substrates suggest that SAPK3 may engage in a

variety of different interactions at this organelle. Therefore, the following Chapter uses

a yeast two-hybrid screening approach to identify some of the SAPK3 binding proteins

that may be responsible for anchoring it at the mitochondria and regulating its

interaction with substrate proteins.

102

Page 177: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Chapter 6

Identification of proteins that bind to SAPK3 by a PDZ domain-mediated interaction

Page 178: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

6.1 Introduction

SAPK3 possesses a unique C-terminal tail with the sequence of Lys-Glu-

Thr-Xaa-Leu-COOH, which allows it to bind to PDZ domains (Hasegawa et al., 1999).

SAPK3 has previously been found to interact with the PDZ domains of α1-Syntrophin

and Post Synaptic Density-95 (PSD-95) (Hasegawa et al., 1999; Sabio et al., 2004). In

each case, the subsequent phosphorylation of these proteins was dependent on the

interaction of the C-terminal tail of SAPK3 with each PDZ domain (Hasegawa et al.,

1999; Sabio et al., 2004).

Although many PDZ-mediated interactions involve proteins at the plasma

membrane (Srivastava et al., 1998; Niethammer et al., 1996), proteins at intracellular

organelles such as the Golgi also can contain PDZ domains (Stricker and Huganir, 2003;

Liu et al., 2001; Kuo et al., 2000). A limited number of PDZ domain-containing

proteins have also been identified at the mitochondria. These include Protein that

Interacts with C-Kinase 1 (PICK1) (Wang et al., 2003a), High Temperature

Requirement A 2 (HtrA2/Omi) (Suzuki et al., 2001)and Outer Membrane Protein 25

(OMP25) (Nemoto and De Camilli, 1999). Other PDZ domain-containing proteins have

also been found in recent proteomic analysis of human heart mitochondria (Taylor et al.,

2003).

In Chapter 3, SAPK3 was shown to be co-localised with the mitochondria,

and this localisation did not change with hyperosmotic stress (results not shown).

Furthermore, in Chapter 4 several mitochondrial proteins were identified as in vitro

substrates of SAPK3. It is not yet clear how SAPK3 is localised at the mitochondria.

For example, an analysis of the SAPK3 sequence using prediction programs such as

PSORT (http://psort.nibb.ac.jp/) does not reveal a mitochondrial targeting sequence

(unpublished observations). Therefore a candidate mechanism for SAPK3 involves the

interaction of the C-terminal tail of SAPK3 with PDZ-containing anchor proteins in the

outer mitochondrial membrane. An approach to identify possible SAPK3 binding

partners that “anchor” SAPK3 at the mitochondria could therefore involve the screening

103

Page 179: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

of a library using SAPK3 or the direct testing of SAPK3 interaction with mitochondrial

PDZ domain-containing proteins.

Yeast two-hybrid analysis is a screening technique now widely used to

identify protein-protein interactions within a eukaryotic cell context (Fields and Song,

1989). This method exploits the functional delineation of transcription factor proteins as

two separable and functionally essential domains: the DNA binding domain and

transactivation domain (Keegan et al., 1986; Hope and Struhl, 1986; Fields and Song,

1989). The DNA binding domain of a transcription factor which binds to specific DNA

sequences (“ABC”) can be fused to a protein, “X”, and the transactivation domain to

another protein, “Y”. Any interaction between X and Y brings the DNA binding domain

and the transactivation domain into close proximity and reconstitutes an active

transcription factor, thereby stimulating the transcription of a gene regulated by the ABC

sequence in the promoter (Fields and Song, 1989).

One of the major aims of this project (as outlined in Chapter 1) was to

identify SAPK3 binding partners that may be responsible for holding it to a particular

location within the cell. A yeast two-hybrid screening strategy employed in this Chapter

used full-length SAPK3 as a “bait” to search for binding partners in a cDNA library

derived from the mouse lymphohematopoietic progenitor cell line EML Clone.1 (Tsai et

al., 1994). Using this screening approach, SAPK3 binding proteins were identified

which contained PDZ domains. The identification of these proteins implies that SAPK3

interactions within the cell will be dominated by these domains. Furthermore this

suggests a mechanism for the anchoring of SAPK3 at the mitochondria. In the second

approach, the yeast two-hybrid system was used to evaluate the interaction between

SAPK3 and a mitochondrial PDZ domain-containing partner OMP25. This protein was

found not to be a substrate for SAPK3, and unlike previously identified PDZ domain-

containing partners, it inhibited the activity of SAPK3. These results suggest the

possibility of additional complexity in the control of MAPK pathways.

104

Page 180: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

6.2 Results

6.2.1 Screening a yeast two-hybrid library with full-length SAPK3

Little is known about the binding partners of SAPK3 that allow its specific

localisation within the cell. A previous report, using yeast two-hybrid screening of

skeletal muscle, identified the PDZ domain-containing protein α1-Syntrophin as a

possible binding partner of SAPK3 (Hasegawa et al., 1999). It was shown that SAPK3

can bind to α1-Syntrophin using a C-terminal PDZ domain binding motif, and that this

allows the subsequent phosphorylation of α1-Syntrophin (Hasegawa et al., 1999). This

published screen for SAPK3 substrates was carried out in a library from skeletal muscle

presumably because this tissue has a high expression of SAPK3 (Mertens S et al., 1996;

Lechner C et al., 1996; Li Z et al., 1996) (Chapter 4). Several studies have examined the

role of SAPK3 in skeletal muscle tissue and in skeletal muscle myoblast and myotube

cells (Ho et al., 2004; Boppart et al., 2000; Tortorella et al., 2003; Hasegawa et al., 1999;

Lechner C et al., 1996). However, SAPK3 is also expressed in a range of other tissues

(Chapter 4), and little is known about the substrates and/or binding partners of SAPK3 in

any of these tissues.

Yeast two-hybrid analysis is generally useful as a screening and/or binding

assay for proteins that do not have any transcriptional activity themselves. Proteins that

do have transcriptional activity when fused to a DNA binding domain, (i.e. they are able

to activate transcription of the reporter genes HIS3 and lacZ without a binding partner

linked to a transactivation domain) are “autoactivators” and thus are inappropriate to use

for a yeast two-hybrid screen. To determine whether SAPK3 was an autoactivator of

transcription, full-length rat SAPK3 was sub-cloned into the LexA DNA binding protein

fusion vector pBTM116 (Vojtek et al., 1993), as an in-frame fusion to LexA. The

Saccharomyces cerevisiae L40 strain (MATa, his3∆200, trp1-901, leu2-3, 112, ade2,

LYS2::(lexAop)4-HIS3, URA3::(lexAop)8-lacZ, GAL4) was then transformed with this

construct. The yeast colonies resulting from this transformation were then assayed for

β-galactosidase activity, as described in Chapter 2. In a separate transformation, another

105

Page 181: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

LexA DNA binding domain fusion protein, LexA-ARL6, was expressed from the

plasmid pBTM116. This was introduced into yeast in conjunction with the protein A21

that had been sub-cloned into the VP16 transactivation domain fusion vector pVP16,

thus producing a fusion to the VP16 transactivation domain. A21 and ARL6 have

previously been identified as interacting partners, and thus the co-transformation of

these two proteins into yeast would act as a positive control for lacZ and HIS3 reporter

activation (E. Ingley, personal communication). Following the transformations, the

resulting yeast colonies were assayed for β-galactosidase activity. Using this method, it

was shown that whilst the positive control activated the lacZ reporter, SAPK3 could not

activate transcription of this reporter gene alone, or in combination with pVP16-A21

(Figure 6.1). Furthermore, apart from the positive control, no growth of the yeast was

seen on media lacking histidine, suggesting no activation of the HIS3 reporter gene

(results not shown). Therefore, SAPK3 was not an autoactivator and could be used in

subsequent yeast two-hybrid screening protocols.

SAPK3 has previously been shown to be expressed in early hematopoietic

progenitor cells (Uddin et al., 2004), and thus it was thought that a library derived from

these cells would be appropriate for screening. Yeast two-hybrid screening of a cDNA

library derived from the mouse lymphohematopoietic progenitor cell line EML Clone.1

(Tsai et al., 1994) has been optimised by Dr Evan Ingley and colleagues at the Western

Australian Institute for Medical Research. Furthermore, several published studies have

arisen from the screening of this library, identifying binding partners of proteins such as

the oncoprotein Myeloid Leukemia Factor-1 (Lim et al., 2002) and Lyn Tyrosine Kinase

(Ingley et al., 2001; Ingley et al., 2000). Therefore, this library was chosen to screen for

potential SAPK3 binding partners.

The general scheme for screening the EML Clone.1 cDNA library is

outlined in Figure 6.2. The S. cerevisiae L40 strain, transformed with the LexA-SAPK3

pBTM116 construct, was subsequently transformed with the EML Clone.1 cDNA

library in which the library proteins were expressed as a VP16 transactivation domain

fusion. Positive interaction of SAPK3 with library proteins was then determined by the

106

Page 182: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

ARL6 SAPK3pBTM116:pVP16: - -

Selective Media:Trp-

ARL6 SAPK3pBTM116:pVP16: A21 A21

Selective Media:Leu-, Trp-

Figure 6.1: pBTM116-SAPK3 is unable to activate transcription alone in the yeast two-hybrid system.The S. cerevisiae L40 strain was transformed with the pBTM116 vector encoding SAPK3 or ARL6, in combination with the protein A21 (cloned into pVP16), or alone. Yeast colonies were then streaked to a grid on the appropriate selective media and assayed for β-galactosidase activity.

106a

Page 183: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

lacZ HIS3

lacZ HIS3

lacZ HIS3

A

B

C

LexA DNA Binding DomainFull-length SAPK3

Proteins represented in the cDNA libraryVP16 Transactivation Domain

lacZ and HIS3 reporter geneslacZ HIS3

Figure 6.2: Screening the yeast two-hybrid EML Clone.1 cDNA library using full-length SAPK3.(A) The S. cerevisiae L40 strain, containing the lacZ and HIS3 reporter genes, was transformed with the LexA fusion vector pBTM116, encoding full-length SAPK3 fused to the LexA DNA binding protein. (B) Yeast cells were subsequently transformed with the EML Clone.1 cDNA library, encoded in the pVP16 vector and fused to the transactivation domain of the VP16 protein.(C) When SAPK3 interacted with a protein from the library, the LexA DNA binding domain and VP16 transactivation domain were brought into close proximity to reconstitute an active transcription factor. This resulted in the activation of the lacZ and HIS3 reporter genes, thus the yeast turned blue with the addition of X-gal, and grew in the absence of the amino acid histidine.

106b

Page 184: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

activation of the HIS3 and lacZ reporter genes. From this large-scale screen, 420 clones

were isolated which activated the lacZ and HIS3 reporter genes (Table 6.1). Yeast

colonies containing positively interacting library plasmids were rescued into Escherichia

coli and co-transformed into the L40 yeast strain with LexA-SAPK3. HIS3 and β-

galactosidase assays were then performed to confirm a positive interaction. Colonies

that showed a positive interaction with SAPK3 in this secondary confirmation procedure

were then sequenced. Using this secondary confirmation procedure, 148 clones were

found to be “true” positives, in that they activated the lacZ and HIS3 reporters in the

presence of a LexA-SAPK3, but not LexA-Lamin or LexA-ARL6 hybrid fusion protein

(Figure 6.3).

6.2.2 SAPK3 interacts with Scribble and Lin-7C by yeast two-hybrid analysis

By sequencing the library clones from the secondary screening, it was

determined that the SAPK3 interactors were the PDZ domain-containing proteins,

Scribble (NCBI Accession Number NP_598850) and Lin-7C (NCBI Accession Number

NP_035829). To determine whether the interaction of Lin-7C and Scribble was

dependent on the C-terminal tail of SAPK3, yeast were co-transformed with Lin-7C or

Scribble, and a truncation mutant of SAPK3, where the 5 most C-terminal amino acids

had been deleted (SAPK3∆5C). No interaction was detected between SAPK3∆5C and

Lin-7C or Scribble, suggesting that the interaction of SAPK3 with these proteins was

dependent on the C-terminal tail (Figure 6.3).

As verified by sequencing, at least two independent clones of each of the

PDZ domain-containing proteins, Scribble and Lin-7C were identified, and these are

illustrated in Figure 6.4. After sequencing 40 positives in the secondary screen, it was

clear that Scribble was being most frequently isolated. Due to this high percentage of

Scribble clones isolated in the screen, a colony screen PCR procedure was optimised

using Scribble-specific primers (Appendix I). Colony screen PCR was then carried out

on all clones isolated from yeast cells to identify Scribble clones and eliminate these

from subsequent lacZ and HIS3 reporter assays. This reduced the number of clones that

107

Page 185: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Table 6.1: Summary of SAPK3 interacting clones isolated using yeast two-hybrid analysis

Number of Primary Positive Clones Isolated (HIS3 and lacZ reporter activation)

420

Number of Secondary Positive Clones Isolated(HIS3 and lacZ reporter activation)

148

Number of Secondary Positive Clones Identified as Scribble (NP_598850)

131

Number of Secondary Positive Clones Identified as Lin-7C (NP_035829)

17

107a

Page 186: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

HIS 3

Scribble

Lin 7C

“Bait”: SAPK3 ARL6 Lamin A SAPK3(∆5C)

B

“Bait”: SAPK3 ARL6 Lamin A SAPK3(∆5C)

Scribble

Lin 7C

Lac Z

Figure 6.3: The identification of Scribble and Lin7C as SAPK3-interacting proteins by yeast two-hybrid screening.Tentatively positive clones from large scale yeast two-hybrid screening were rescued into E.coli and transformed into the S. cerevisiae L40 strain. These were then tested for interaction with LexA-SAPK3, by assaying the activity of the lacZ (A) and HIS3 (B) reporter genes. As a control, the interaction of tentatively positive library clones with LexA-ARL6 and LexA-Lamin A was assessed. Proteins that showed activation of the reporter genes when tested against LexA-SAPK3, but not LexA-ARL6 and LexA-Lamin A, were considered to be “true positives”. To determine whether the interaction of SAPK3 with Lin7C and Scribble was dependent on the C-terminal tail of SAPK3, interaction was also measured against LexA-SAPK3 that was missing the last 5 C-terminal amino acids (SAPK3∆5C).

107b

Page 187: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

(i)

(ii)

(iii)

B

(i)

800740

LRR

28 450 845 935 988 1061 117710841 1665

944267 LRR

712 898

1 197L27

13 68 91 173

PDZ1

PDZ1

PDZ1

PDZ2

PDZ2

PDZ2

PDZ3 PDZ4

107c

PDZ

1 194L27

13 68 91 173

PDZ(ii)

L277 1PDZ(iii) 86Figure 6.4: Scribble and Lin7C clones isolated in yeast two-hybrid screening.Schematic diagrams are shown to illustrate the full-length Scribble (A i) and Lin-7C (B i) proteins, as well as the different Scribble (A ii, iii) and Lin7C (B ii, iii) clones isolated from yeast two-hybrid screening. The identity of these clones was determined by DNA sequencing.

Page 188: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

needed to be analysed for lacZ and HIS3 reporter gene activity, and allowed the potential

identification of other SAPK3 interacting proteins. Ultimately, Scribble clones isolated

comprised 89% of the total number of true positives, and Lin-7C made up the other

11%. No other proteins were identified.

Analysis of the full-length Scribble and Lin-7C protein sequences showed

that they both contained Class I PDZ domains. Scribble contained 4 PDZ domains and

Lin-7C contained only 1. The clones identified in the yeast two-hybrid screen each

contained at least one of these domains. This is consistent with a previous report

showing that SAPK3 interacts with Class I PDZ domains (Hasegawa et al., 1999). Class

I PDZ domains interact with the consensus sequence of (Ser/Thr)-Xaa-

(Val/Iso/Leu/Ala)-COOH at the C-terminus of their protein ligands (Songyang et al.,

1997; Kornau et al., 1995), and this is consistent with the C-terminal sequence of the rat

SAPK3 protein used: Thr-Ala-Leu-COOH.

Previous reports have shown Lin-7C to localise at cell-cell junctions (Irie et

al., 1999; Yamamoto et al., 2002). One study has also examined the expression of Lin-

7C within different tissues, finding that the mRNA is ubiquitously expressed, however

the protein is only expressed in a select number of tissues (Irie et al., 1999).

Specifically, Lin-7C was expressed at high levels in the brain, kidney and liver, low

levels of the protein were detected in spleen and lung, and no Lin-7C was detected in

heart, testis or skeletal muscle (Irie et al., 1999). Comparing the characteristics of Lin-

7C to that of SAPK3 expression and localisation presented in Chapters 3 and 4, it

appeared unlikely that these two proteins could be physiological binding partners.

A large proportion of clones isolated in the yeast two-hybrid screen were

identified as Scribble (Table 6.1). The Drosophila homologue of Scribble has been

relatively well-characterised in comparison to the mammalian protein. At the time of

commencing work on this part of the project, it was known that Scribble co-localised at

epithelial cell junctions in Drosophila (Bilder and Perrimon, 2000), however little was

known about the subcellular localisation or tissue distribution of mammalian Scribble.

108

Page 189: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

The access to a specific anti-Scribble monoclonal antibody, as well as the relatively

limited knowledge on this protein meant that further investigation was required to

determine whether Scribble was a possible SAPK3 binding partner.

To determine whether Scribble was a potential physiological binding

partner of SAPK3, the expression of Scribble was examined in a variety of different

mouse tissues using a specific Scribble monoclonal antibody (Figure 6.5A). It was

shown in Chapter 4 that SAPK3 was expressed in a number of different tissues, with

highest expression levels in skeletal muscle, heart, lung and testes. This Western blot is

shown again for reference in Figure 6.5B. By immunoblotting these tissues with the

Scribble antibody, Scribble was found to be expressed at its highest levels in thymus.

Immunoreactivity was also found in spleen, kidney, liver and brain. However, very little

immunoreactivity was detectable in heart, skeletal muscle, or testes. Therefore, the

tissue distribution pattern of Scribble appeared to differ from that of SAPK3.

Scribble is expressed highly in epithelial cells, where it is involved in the

regulation of cell polarity (Bilder and Perrimon, 2000). Due to the high level of Scribble

expression in Madin-Darby canine kidney (MDCK) epithelial cells (P. Humbert,

personal communication), the subcellular localisation of Scribble in these cells was

determined. First, expression of Scribble in MDCK cells was confirmed with the

Scribble antibody, by immunoblotting lysates from MDCK cells prepared under the

conditions used previously for cell and tissue samples (Fig 6.6A). When these cells

were stained with the Scribble antibody and analysed with confocal microscopy,

Scribble staining appeared at the edges of these cells and was consistent with a plasma

membrane localisation (Figure 6.6Bii). This did not co-localise with SAPK3 staining

(Figure 6.6Biii). Indeed, it was shown in Chapter 3 that SAPK3 co-localised with

mitochondria in cardiac myocytes and in HEK293 fibroblasts. As seen in Figure 6.6B,

when using the SAPK3 antibody and Mitotracker, SAPK3 also demonstrated a

mitochondrial localisation pattern in MDCK cells (Figure 6.6Bvi). Thus, it appears that

Scribble and SAPK3 do not share the same tissue distribution (Figure 6.5) or localisation

in cells (Figure 6.6) and are unlikely to be physiological binding partners.

109

Page 190: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A α-Scribble

H Skm B Li Lu Th Te S K

Scribble220 kDa

B α-SAPK3

H Skm B Li Lu Th Te S K

SAPK346 kDa

CCoomassie

H Skm B Li Lu Th Te S K

220 kDa

97.5 kDa66 kDa

45 kDa

30 kDa

Figure 6.5: The expression of Scribble in adult mouse tissues.Mouse tissues were crushed under liquid nitrogen and lysed. Protein lysates (30 µg) were then subjected to Western blotting using the anti-Scribble antibody (A) or anti-SAPK3 antibody (B). A Coomassie stained gel is shown to illustrate relatively even loading of samples (C). Western blotting results are representative of at least 2 independent experiments.Abbreviations: H = Heart; Skm = Skeletal Muscle; B = Brain; Li = Liver; Lu = Lung; Th = Thymus; Te = Testes; S = Spleen; K = Kidney.

109a

Page 191: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

220 kDa Scribbleα-Scribble

B

Anti-SAPK3 Anti-Scribble Overlayi ii iii

OverlayAnti-SAPK3 Mitotracker

Figure 6.6: Determining the localisation of SAPK3 and Scribble in MDCK cells.(A) MDCK cells were seeded into dishes and grown for 2 days before lysis. Protein (30 µg) was then separated by SDS-PAGE and subjected to Western blotting using a specific Scribble monoclonal antibody. (B) MDCK cells were seeded into dishes on coverslips and treated with Mitotrackerfor 45 min (v) or left untreated, then stained with the SAPK3 antibody (i, iv), or Scribble antibody (ii). Panels (iii) and (vi) show an overlay of panels (i) and (ii) or (iv) and (v), respectively, and Hoescht 33258 staining is also shown to visualise the cell nuclei. Arrows show Scribble staining at the cell edges. Yellow staining indicates where SAPK3 and Mitotracker co-localise. Results shown are representative of 2 independent experiments. Scale bar = 20 µm.

iv v vi

109b

Page 192: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

6.2.3 SAPK3 interacts with the mitochondrial outer membrane protein, OMP25

An alternative use of the yeast two-hybrid system is to test directly the

interaction between two known proteins rather than use it as a library screening tool.

Due to the mitochondrial localisation of SAPK3, the literature was surveyed for PDZ

domain-containing proteins localised at the mitochondria. A protein was identified

called “Outer Mitochondrial Protein 25” (OMP25) (Nemoto and De Camilli, 1999);

Figure 6.7). OMP25 mRNA is expressed in a variety of tissues, with highest levels of

expression reported for skeletal muscle (Nemoto and De Camilli, 1999). The OMP25

protein is inserted into the outer mitochondrial membrane, with its PDZ domain facing

the intracellular environment, allowing its interaction with cytosolic proteins containing

PDZ domain-binding motifs (Nemoto and De Camilli, 1999). This suggests that

OMP25 can perform an “anchoring” function by holding specific proteins at the

mitochondria. Consistent with this role, OMP25 has previously been found to associate

with the inositol phosphatase Synaptojanin 2A, localising this protein to the

mitochondria (Nemoto and De Camilli, 1999).

A mammalian expression vector, encoding full-length OMP25 fused to a

HA tag at the N-terminus, was obtained (Nemoto and De Camilli, 1999) and used to

transfect into HEK293 fibroblasts. Cells were lysed following varying times of

transfection, and the lysates subjected to Western blotting with an antibody against the

HA tag. As seen in Figure 6.8A, the expression of HA-OMP25 could be detected 22, 30

or 46 hours following transfection. To determine the optimal amount of HA-OMP25

DNA for transfection, 0 – 1.2 µg DNA was transfected into HEK293 fibroblasts, and the

cells lysed after 40 hours. These lysates were then subjected to Western blotting with

the HA antibody. As shown in Figure 6.8B, the expression of HA-OMP25 could be

detected when as little as 0.4 µg DNA was introduced into the cells, and the expression

of HA-OMP25 increased following transfection with more DNA (0.6 or 1.2 µg).

110

Page 193: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Amphipathic helix PDZ domain TM1 73 160 178 200 206

Amino acid residue

2.64

-3.04

0

20 40 60 80 100 120 140 160 180 200

hydr

ophi

licity

From Nemoto and De Camilli; EMBO J; 18: 2991-3006 (1999)

Figure 6.7: Schematic diagram of Outer Membrane Protein 25 (OMP25).A schematic representation of the rat OMP25 domain structure, and a hydrophilicity plot are shown. OMP25 contains a transmembrane domain (aa 178-200) which allows the protein to be inserted into the outer mitochondrial membrane. OMP25 is orientated with the PDZ domain facing the intracellular environment (Nemoto and De Camilli, 1999).

110a

Page 194: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A

45 kDa

30 kDa

66 kDa97.5 kDa

20.1 kDa

HA-OMP25

HA-OMP25 Transfection: - + + + + -Time of expression (hours): 8 8 22 30 46 46

α-HA

B

Amount of HA-OMP25 DNA (µg): 0 0.1 0.2 0.4 0.6 1.2

45 kDa

30 kDa

66 kDa97.5 kDa

20.1 kDa

HA-OMP25

α-HA

Figure 6.8: Overexpression of HA-OMP25 in HEK293 fibroblasts.(A) HEK293 fibroblasts were transfected with 0.6 µg HA-OMP25 DNA and 0.6 µg Bluescript DNA, and allowed to express the protein for 8 – 46 hours. The cells were then lysed, and protein separated by SDS-PAGE. Western blotting was then performed using a HA antibody. (B) HEK293 fibroblasts were transfected with 0 – 1.2 µg HA-OMP25 DNA, in conjunction with Bluescript DNA or alone, and allowed to express the protein for 40 hours. The cells were then lysed, and protein separated by SDS-PAGE. Western blotting was then performed using a HA antibody.In both panels, high molecular weight proteins (indicated by open arrows) cross-reacted with the HA antibody.

110b

Page 195: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

OMP25 has previously been shown to be localised at the outer

mitochondrial membrane, by immunofluorescence, proteinase K treatment and alkali

carbonate treatment (Nemoto and De Camilli, 1999). To confirm the co-localisation of

OMP25 with SAPK3 at the mitochondria, HEK293 fibroblasts were transfected with

HA-OMP25 and allowed to express the protein for 40 hours. Cells were then fixed,

permeabilised and stained with HA and SAPK3 antibodies. As shown in Figure 6.9,

non-transfected cells showed typical punctate staining for endogenous SAPK3 and no

staining for HA-OMP25. However, in cells overexpressing HA-OMP25, intense

staining with the HA antibody and coalescence of the mitochondria was observed, which

is consistent with a previous report (Nemoto and De Camilli, 1999). In these cells, co-

localisation of endogenous SAPK3 with the transfected, overexpressed HA-OMP25 was

visualised (Figure 6.9C).

To determine whether SAPK3 interacted with OMP25, yeast two-hybrid

analysis was used. The binding of these two proteins was assessed using a truncation

mutant of OMP25, consisting of amino acids 37 – 177 (OMP25(37-177)). This

truncation mutant allowed the assessment of SAPK3 binding to the OMP25 PDZ

domain, and also removed the outer mitochondrial transmembrane domain of OMP25, at

the C-terminal end of the protein. This has been previously shown to be required for

targeting of OMP25 to the mitochondria (Nemoto and De Camilli, 1999). Therefore the

presence of this targeting domain in OMP25 may compromise the yeast two-hybrid

system, where proteins must be nuclear for an interaction to be detected because the

reporter system is based on transcription which can only occur in the nucleus. It was

found that OMP25(37-177) interacted with SAPK3, by the activation of the HIS3 and

lacZ reporters, but did not interact with the negative control proteins ARL6 or Lamin A

(Figure 6.10). Deletion of the last five amino acids of SAPK3 in the SAPK3∆5C mutant

abolished the interaction between OMP25 and SAPK3 (Figure 6.10). This suggested

that the interaction was dependent on the C-terminal tail of SAPK3, consistent with an

interaction of this sequence with the PDZ domain of OMP25.

111

Page 196: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Anti-HA (OMP25)

A

Anti-SAPK3

B

Overlay

C

Figure 6.9: Confirmation of OMP25 and SAPK3 co-localisation.293 cells were transfected with HA-OMP25 and allowed to express the protein for 40 hours. Cells were then fixed, permeabilised and stained with HA (A) and SAPK3 (B) antibodies. Panel C shows an overlay of Panels A and B, and Hoescht 33258 staining is included to visualise the cell nuclei. Results shown are representative of 2 identical experiments. Scale bar = 18 µm.

111a

Page 197: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A“Bait”: SAPK3 ARL6 Lamin A SAPK3∆5C

OMP25(37-177)

HIS 3

B“Bait”: SAPK3 ARL6 Lamin A SAPK3∆5C

OMP25(37-177) lacZ

Figure 6.10: Interaction of SAPK3 with OMP25 detected by yeast two-hybrid analysis.VP16-OMP25 DNA was transformed into yeast and tested for interaction with LexA-SAPK3. This was assessed using the activation of the HIS3 (A) or lacZ (B) reporters as indicators. As negative controls, the interaction of VP16-OMP25 was determined towards LexA-ARL6 and LexA-lamin. VP16-OMP25 was also tested for interaction with the C-terminal truncation mutant of SAPK3 (SAPK3∆5C).

111b

Page 198: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

To further investigate the interaction between these proteins, a co-

immunoprecipitation of HA-OMP25 and Myc-SAPK3 was performed. HEK293

fibroblasts were transfected with HA-OMP25 and Myc-SAPK3. The HA monoclonal

antibody was then used to immunoprecipitate HA-OMP25 from cell lysates, and

Western blotting was performed using the specific SAPK3 monoclonal antibody. As

seen in Figure 6.11, when HA antibody was added to the lysate, SAPK3 was isolated in

the immunoprecipitation. No SAPK3 was detected when HA antibody was omitted

from the procedure. This suggested that OMP25 and SAPK3 could interact in cells.

Due to the migration of OMP25 on SDS-PAGE at the same size as light chain

immunoglobulin, it was not possible to perform an immuoprecipitation with Myc

antibody and probe for HA-OMP25 using the HA antibody.

It was found that SAPK3 and OMP25 co-localise at the mitochondria using

immunofluorescence, and that these proteins can interact by co-immunoprecipitation and

yeast two-hybrid analysis. To determine whether OMP25 could alter the subcellular

localisation of SAPK3, a truncation mutant of HA-OMP25 was made, introducing a stop

codon immediately upstream of amino acid 178. OMP25 lacking the transmembrane

segment (amino acids 178-200) has previously been shown to be distributed throughout

the cytosol, rather than at the mitochondria (Nemoto and De Camilli, 1999). Following

this, using PCR, a nuclear localisation signal (Sequence: Pro-Pro-Lys-Lys-Lys-Arg-Lys-

Val-Ala) (Junqueira et al., 2003) was introduced immediately upstream of the OMP25

sequence (following the HA epitope tag), to direct the protein into the nucleus (HA-

OMP25(NLS)). It was proposed that HA-OMP25(NLS) would bind to SAPK3 and that

this protein complex could then localise to the nucleus.

To determine whether HA-OMP25(NLS) could alter the localisation of

overexpressed SAPK3, cells were transfected with HA-OMP25(NLS) and Myc-tagged

SAPK3, and allowed to express the proteins for 40 hours. Cells were then fixed,

permeabilised and stained with SAPK3 and HA antibodies. Myc antibody could not be

used to detect SAPK3-transfected cells because both the HA and Myc antibodies were

raised in mice, and so could not be detected simultaneously with immunostaining.

112

Page 199: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

+α-HA

-α-HA

HA Abonly

45 kDa

30 kDa

Myc-SAPK3IgG HC

IgG LC

Figure 6.11: SAPK3 and OMP25 are co-immunoprecipitated from cells.HEK293 fibroblasts were transfected with myc-tagged SAPK3 and HA-tagged OMP25 and allowed to express the proteins for 24 hours. The cells were then lysed, and protein lysates incubated with HA antibody (right panel), or no antibody (left panel) overnight. The following day, immunoprecipitated proteins were captured on Protein G sepharose and subjected to Western blotting with the SAPK3 antibody. HA antibody (5 µg) was also subjected to Western blotting (middle panel) to show the position of the immunoglobulin heavy and light chains. Results shown are representative of at least 4 independent experiments.IgG HC = Immunoglobulin Heavy Chain; IgG LC = Immunoglobulin Light Chain.

112a

Page 200: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

However, the limit of detection for overexpressed SAPK3 was much lower than that for

endogenous SAPK3 using the confocal microscope, and thus the staining represents the

overexpressed protein only. As shown in Figure 6.12A, when compared to cells

transfected with Myc-SAPK3 only, some cells expressing HA-OMP25 and Myc-SAPK3

showed increased nuclear accumulation of SAPK3. This suggested that HA-

OMP25(NLS) could be binding to SAPK3 and altering its subcellular localisation in

some instances. However, not all SAPK3 in HA-OMP25(NLS) overexpressing cells

was moved to the nucleus, with cytoplasmic staining of this overexpressed protein

remaining visible (Figure 6.12Aiii).

To determine whether OMP25(NLS) could alter the subcellular

localisation of endogenous SAPK3, cells were transfected with HA-OMP25(NLS),

allowed to express the protein for 40 hours, then fixed, permeablised and stained with

SAPK3 and HA antibodies. As shown in Figure 6.12B, no difference was seen in

endogenous SAPK3 distribution when cells were transfected with HA-OMP25(NLS)

(white arrows), compared to non-transfected cells. Nuclear localisation of HA-

OMP25(NLS) was confirmed by staining the cells with Hoescht 33258. This suggested

that HA-OMP25(NLS) could not alter the subcellular localisation of endogenous

SAPK3.

6.2.4 OMP25 can act as an inhibitor of SAPK3 in vitro

To further characterise the interaction of these proteins, OMP25 was cloned

into the pGEX-6P vector, to allow expression of the protein in bacteria. Full-length

OMP25 was poorly expressed in bacteria, as determined by Bradford protein assay and

SDS-PAGE (Figure 6.13). This was presumably due to the hydrophobicity of the

transmembrane segment, and therefore a truncation mutant of the OMP25 protein was

made by introducing a stop codon immediately upstream of amino acid 178 (GST-

OMP25(1-177)). This truncation mutant was expressed at a higher level in bacteria

(Figure 6.13). Examination of the OMP25 sequence showed that this protein contains

no serines or threonines that are directly followed by a proline (the minimum consensus

113

Page 201: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

AOverlayAnti-SAPK3 Anti-HA

Anti-SAPK3 Anti-HA Overlay

Anti-HAHoescht 33258 Overlay

Figure 6.12: Using HA-OMP25(NLS) to alter the subcellular localisation of SAPK3.(A) HEK293 fibroblasts were transfected with 0.05 µg Myc-SAPK3 and 2 µg HA-OMP25(NLS) and allowed to express the proteins for 40 hours. Cells were then fixed, permeabilised and stained with SAPK3 (i) or HA (ii, v) antibodies. Cells were also stained with Hoescht 33258 to visualise the nuclei (iv). Panels (iii) and (vi) show an overlay of panels (i) and (ii) or (iv) and (v), respectively. White arrows indicate cells that are overexpressing SAPK3. Results shown are representative of 2 independent experiments. Scale bar = 20 µm.(B) HEK293 fibroblasts were transfected with 2 µg HA-OMP25(NLS) and allowed to express the protein for 40 hours. Cells were then fixed, permeabilised and stained with SAPK3 (i) or HA (ii, v) antibodies. Cells were also stained with Hoescht 33258 to visualise the nuclei (iv). Panels (iii) and (vi) show an overlay of panels (i) and (ii) or (iv) and (v), respectively. Results shown are representative of 2 independent experiments. Scale bar = 20 µm.

i

iv

B

v

ii

vi

iii

iiii ii

Hoescht 33258 Anti-HA Overlayviiv v

113a

Page 202: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

A“OMP SDM Reverse”: 3’ CTG CCT CTC GGC ACT CCT CAA GGA CAT CGA C 5’

5’….CGA GGC GAC GGA GAG CCG AGT GGA GTT CCT GTA GCT GTG GTG……3’3’….GCT CCG CTG CCT CTC GGC TCA CCT CAA GGA CAT CGA CAC CAC……5’

“OMP SDM Forward”: 5’ GAC GGA GAG CCG TGA GGA GTT CCT GTA GCT G 3’

BGST-OMP25 GST-OMP25

(1-177) (wild-type)

GST-OMP25 (wild-type)GST-OMP25(1-177)45 kDa

66 kDa

Figure 6.13: The expression of GST-OMP25 in bacteria.(A) “OMP SDM Forward” and “OMP SDM Reverse” primers were used tointroduce a mutation into the OMP25 full-length sequence at the position coding for amino acid 178. This mutation (area shown in red) altered the wild-type amino acid sequence at position 178 from a Ser residue (AGT) to a “STOP” codon (TGA), therefore resulting in a truncated OMP25 product at amino acid 177.(B) Full-length OMP25 was subcloned into the pGEX-6P vector and expressed in BL21plyss cells, as described in Chapter 2. In addition, site-directed mutagenesis was used to introduce a stop codon at position 178 in the OMP25 protein (OMP25(1-177)). This truncated protein was also expressed in BL21plyss cells. The purified proteins were then run on SDS-PAGE, and the gel stained with Coomassie Brilliant Blue.

113b

Page 203: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

sequence required for phosphorylation of a substrate by a member of the MAPK family

(Clark-Lewis et al., 1991)). However, to ascertain that OMP25 was not a substrate of

SAPK3, in vitro kinase assays were performed with activated GST-SAPK3 and GST-

OMP25. As seen in Figure 6.14, GST-OMP25 was not phosphorylated by active GST-

SAPK3, whereas GST-α1-Syntrophin was phosphorylated to a high extent under the

same conditions.

Since OMP25 contains a PDZ domain which appears to bind SAPK3 with

high affinity but that it cannot itself be phosphorylated, it was proposed that GST-

OMP25 may interact with SAPK3 and inhibit the interaction of GST-SAPK3 with its

PDZ domain-containing substrates. To investigate this, active GST-SAPK3 was pre-

incubated with GST-OMP25(1-177), then subjected to in vitro kinase assays with GST-

Sab and GST-α1-Syntrophin (Hasegawa et al., 1999). As shown in Figure 6.15, 1 µg

GST-OMP25 inhibited the phosphorylation of 5 µg GST-α1-Syntrophin and GST-Sab

by GST-SAPK3, by approximately 40%. This increased to approximately 60%

inhibition when 5 µg GST-OMP25 was used, and 90% when 10 µg was used. This

suggests that OMP25 can act as an inhibitor of SAPK3. Furthermore, this inhibition

appears to be independent of whether or not the SAPK3 substrate contains a PDZ

domain.

6.3 Discussion

Many kinase signalling pathways contain proteins that do not directly

signal themselves, but that have anchoring or scaffolding roles, thereby placing the

kinase or substrate in the correct location to enhance signalling events. These anchoring

or scaffolding interactions involve a variety of different protein domains, such as the

SH2 or SH3 domains, PDZ domains, or Phosphotyrosine domains (Pawson and Nash,

2003). An example of an anchoring role is seen in the effective signalling by Protein

Kinase C-alpha (PKCα) being dependent on an interaction with the PDZ domain of

PICK1 (Wang et al., 2003b). PKCα is targeted to the mitochondria in response to serum

stimulation and is anchored at this organelle through a PDZ domain-mediated interaction

114

Page 204: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

45 kDa

66 kDa

97 kDa

30 kDa

MalE-MKK6(DD): + + + GST-SAPK3: + + + GST substrate: - α1-Syn OMP25

(1-177)

Autoradiograph

GST-α1-Syntrophin

GST-OMP25(1-177)

45 kDa

66 kDa

97 kDa

30 kDa

GST-α1-Syntrophin

GST-OMP25(1-177)

Coomassie

Figure 6.14: OMP25 is not a substrate of SAPK3 in vitro.GST-SAPK3 was activated by MalE-MKK6(DD) and subjected to kinase assays with GST-α1-Syntrophin or GST-OMP25(1-177). An autoradiograph of a representative gel from one of 2 identical experiments is shown after 1 hour exposure. The lower panel shows the Coomassie-stained gel to illustrate even loading of GST substrates.

114a

Page 205: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

MalE-MKK6(DD): + + + + + + + +GST-SAPK3: + + + + + + + +GST substrate: Sab α1-SyntrophinGST-OMP25(1-177)(µg): 0 1 5 10 0 1 5 10

GST-α1-Syntrophin

GST-Sab

GST-OMP25(1-177)

GST-α1-Syntrophin

GST-Sab

45 kDa

66 kDa

97 kDa

45 kDa

66 kDa

97 kDa

Autoradiograph

Coomassie

GST-SabGST-α1-Syntrophin

Phos

phor

ylat

ion

of G

ST s

ubst

rate

(%

com

pare

d to

no

GST

-OM

P25)

(* p<0.01 compared to no GST-OMP25)

**

** * *

0

20

40

60

80

100

120

0 µg 1 µg 5 µg 10 µgGST-OMP25:

Figure 6.15: GST-OMP25 inhibits the phosphorylation of GST-Sab and GST-α1-Syntrophin by SAPK3 in vitro.GST-SAPK3 was activated by MalE-MKK6(DD) and pre-incubated with GST-OMP25(1-177) (0 – 10 µg) for 15 min, before being subjected to kinase assays with GST-Sab or GST-α1-Syntrophin. An autoradiograph of a representative gel is shown in the top panel, and the middle panel shows the Coomassie-stained gel to illustrate even loading of GST-Sab and GST-α1-Syntrophin, as well as the increasing amount of GST-OMP25 in each assay. The levels of GST-SAPK3 and MalE-MKK6(DD) are below the level of detection for Coomassie staining. The bottom panel shows the quantitation of 3 assays as determined by Cerenkov counting of the GST-Sab or GST α1-syntrophin bands. The open arrows indicate GST-α1-Syntrophin degradation products. The results are shown as means ± SE. * p<0.01 compared to samples where no GST-OMP25 was added.

114b

Page 206: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

with PICK1 (Wang et al., 2003b). An example of a scaffolding protein involved in

signalling pathways is JNK Interacting Protein-1 (JIP-1), which specifically binds to

components of the JNK MAPK signalling cascade including JNK, MKK7, and mixed-

lineage kinase 3 (MLK3) (Whitmarsh et al., 1998). It has been suggested that JIP-1

mediates activation of the JNK pathway by bringing these proteins into close proximity

to each other (Whitmarsh et al., 1998).

The identification of SAPK3 mitochondrial localisation, and the ability of

SAPK3 to phosphorylate mitochondrial targets, prompted the search for binding partners

that may be responsible for “anchoring” SAPK3 at the mitochondria. In this Chapter,

the yeast two-hybrid system was used as a method to identify SAPK3 binding partners.

The PDZ domain proteins Lin-7C and Scribble were isolated as binding partners of

SAPK3 using this screen. The isolation of two PDZ domain-containing proteins as

binding partners in this screen suggests that SAPK3 interactions in the cell may be

dominated by these domains.

The high percentage of Scribble clones isolated in the yeast two-hybrid

screen (Table 6.1) implied that Scribble could be a physiologically relevant SAPK3

binding partner. However, it cannot be ruled out that Scribble is highly represented

within this particular cDNA library prepared from the lymphohematopoietic progenitor

cell line EML Clone.1, which would bias this screen. The apparently large proportion of

plasma membrane PDZ proteins compared to those located at intracellular organelles

(Fanning and Anderson, 1999) would also create a library bias. Further screening

attempts using different sources of libraries such as heart or skeletal muscle may be

required to identify other SAPK3 binding partners. However at this stage it cannot be

completely discounted that SAPK3 and Scribble interact with each other in certain

circumstances, such as following the exposure of cells to certain stimuli, or in cell types

in which SAPK3 has not yet been studied. It should be noted for example, that punctate

Scribble staining has been observed in Drosophila, which was thought to be reminiscent

of vesicles (Bilder and Perrimon, 2000). However, the localisation or translocation of

115

Page 207: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

overexpressed or endogenous SAPK3 to the plasma membrane has not yet been

reported.

Because the library screen failed to isolate mitochondrial PDZ domain-

containing proteins, a direct assessment of binding between SAPK3 and OMP25 was

undertaken (Figure 6.10 and 6.11). Since OMP25 was shown to bind SAPK3, the

question arises as to why OMP25 was not isolated in the original yeast two-hybrid

screen. There are several possible reasons for this. Firstly, the expression of OMP25 in

the EML Clone.1 library has not been determined. Further screening attempts with a

skeletal muscle library, the tissue in which SAPK3 and OMP25 are most highly

expressed (Chapter 4) (Mertens et al., 1996; Nemoto and De Camilli, 1999), could

possibly isolate OMP25 as a SAPK3 binding protein. Another possible reason for not

originally isolating OMP25 in this screen could be that as mentioned earlier, most PDZ

domain-containing proteins are localised to the cell membrane (Fanning and Anderson,

1999), and therefore those localised to intracellular organelles such as the mitochondria

would be less prominent in the library. Thirdly, the mitochondrial targeting sequence of

OMP25 could prevent it from being transported into the nucleus to activate the

transcription of the reporter genes. Although the pVP16 vector contains a nuclear

localisation signal, it is not known whether this NLS could “out-compete” the

mitochondrial localisation signal of OMP25.

It may be useful to perform a yeast two-hybrid screen using mutants of

SAPK3 to isolate different binding partners or substrates. For example, SAPK3(1-363)

could be used to screen a library, in order to identify binding partners that do not contain

PDZ domains. In order to bias a screen towards finding binding partners of SAPK3

rather than substrates, a kinase-dead mutant could be used. Yeast two-hybrid screens

using kinase-dead mutants have previously been reported to search for binding partners

that are not dependent on the kinase domain (Ingley et al., 2000). For example, a yeast

two-hybrid screen performed with Lyn tyrosine kinase, in parallel with LynY397F,

identified hematopoietic lineage cell-specific protein (HS1) as binding the SH3 domain

116

Page 208: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

of Lyn (Ingley et al., 2000). HS1 bound with higher affinity to LynY397F than to wild-

type Lyn (Ingley et al., 2000).

To identify weaker and possibly transient binding partners for SAPK3 such

as those with particular substrate proteins that do not rely on PDZ domain-mediated

interactions, it may be necessary to co-transform SAPK3 into a library with

constitutively active MKK6 (Alonso et al., 2000). This would allow SAPK3 to be

phosphorylated and thus adopt an active conformation. It has been proposed that

SAPK3, in its unphosphorylated form, binds ATP poorly (Fox et al., 1999).

Furthermore, the structure of phosphorylated SAPK3 has been solved, and it has been

proposed that phosphorylated Tyr185 interacts with Arg189 and Arg192 in the

activation loop of the kinase, which unblocks the substrate binding site (Bellon et al.,

1999). Therefore, it is possible that the use of active SAPK3 to screen yeast two-hybrid

libraries may increase the likelihood of finding substrate proteins.

The results presented in this Chapter imply that OMP25 may anchor

SAPK3 at the mitochondria, and suggest a mechanism by which SAPK3 could be

brought in close proximity to potential substrates in this location. Examination of the

data presented in Figure 6.12B suggests that HA-OMP25(NLS) was unable to alter the

subcellular localisation of endogenous SAPK3. It is possible that endogenous SAPK3,

already involved in a PDZ-mediated interaction with endogenous OMP25, was unable to

be released and thus remained at the mitochondria. Furthermore, when OMP25 is

overexpressed in conjunction with SAPK3 (Fig 6.12A), this may allow the newly-

translated SAPK3 to bind to OMP25.

Further studies will be required to investigate the possible interaction of

SAPK3 and OMP25 within the cell. For example, if these two proteins interact, an

OMP25 knockout would be expected to show disruptions in SAPK3 mitochondrial

localisation. For example, the proteins nNOS, CAPON and Synapsin I have previously

been found to co-localise in synaptic vesicles in the mouse brain, where they are thought

to form a ternary complex (Jaffrey et al., 2002). In Synapsin I/II double knockout mice,

117

Page 209: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

alterations in nNOS and CAPON subcellular localisation are seen (Jaffrey et al., 2002).

As an alternative to gene knockout studies, RNA interference to knock down OMP25

protein production could also be useful.

OMP25 has previously been shown to anchor Synaptojanin 2A at the

mitochondria (Nemoto and De Camilli, 1999). Due to the presence of only one PDZ

domain in the OMP25 protein, binding of proteins to the OMP25 PDZ domain would be

mutually exclusive, since only one C-terminal tail can bind to one PDZ domain at any

time (Doyle et al., 1996). Therefore, if one isoform of OMP25 binds to Synaptojanin 2A

and SAPK3 in vivo, a 1:1 ratio of OMP25 to the cumulative total of binding partners

could exist in the cell. This would involve regulated expression of OMP25 and the

anchored proteins. Alternatively, it is possible that different isoforms of OMP25 anchor

different proteins. It has been suggested that several different isoforms of OMP25 exist

(Nemoto and De Camilli, 1999). The SAPK3 C-terminal tail has the sequence of: Lys-

Glu-Thr-Xaa-Leu-COOH, whereas Synaptojanin 2A has the C-terminal tail of: Ser-Gly-

Ser-Ser-Val-COOH (Nemoto and De Camilli, 1999). Only one OMP25 isoform has

been tested for binding to Synaptojanin 2A and SAPK3. However, it is possible that

different isoforms of OMP25 may bind with different affinities to Synaptojanin 2A and

SAPK3 based on their C-terminal tails.

Several different classes of PDZ domains have been defined, based on the

C-terminal or internal hairpin-forming PDZ domain recognition motifs of their binding

partners (Songyang et al., 1997; Hung and Sheng, 2002). Several reports have examined

the binding specificities of PDZ domains (Fuh et al., 2000; Doyle et al., 1996; Skelton et

al., 2003; Songyang et al., 1997). It has been found that adjustment of single amino

acids in PDZ domain binding motifs can greatly alter binding affinities (Gee et al., 2000;

Skelton et al., 2003). Further characterisation of the binding specificity of PDZ domains

for their ligands may help to distinguish physiological versus artefactual binding

partners in the cell.

118

Page 210: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

In this Chapter, it was found that OMP25 can act as an inhibitor of SAPK3

in vitro (Figure 6.15). This suggests that within the cell, if OMP25 anchors SAPK3 to

the cell mitochondria, it may act as a mechanism for keeping SAPK3 “inactive”, thus

preventing unwanted signalling events. Further studies will be required to identify

whether these proteins associate in vivo, and if so, to determine the mechanism by which

SAPK3 is released from the PDZ domain-mediated interaction. The identification of a

PDZ domain having a negative effect on substrate phosphorylation by SAPK3 suggests

that the structure of these domains could be used as a basis to design inhibitors

specifically targeting this kinase. The development of such inhibitors would go a long

way in assisting the search for in vivo SAPK3 substrates.

119

Page 211: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Chapter 7

General Discussion

Page 212: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

7.1 General Discussion

The p38 MAPK family has four members – SAPK2a, SAPK2b, SAPK3

and SAPK4. Although studies evaluating the function of SAPK2a and SAPK2b have

been facilitated by the use of ATP-competitive inhibitors such as SB203580 (Cuenda et

al., 1995), no specific small molecule inhibitors of SAPK3 and SAPK4 have been

described. Thus, elucidating the functions of SAPK3 and SAPK4 has proved to be a

more challenging task (Cohen, 1997). At the commencement of this project, little was

known about the role of SAPK3 in any tissues. Specifically, most studies had used

overexpression of the protein kinase in cultured cell lines to evaluate its activation,

substrate specificity and subcellular localisation (Cuenda et al., 1997; Marinissen et al.,

2001). However, the overexpression of protein kinases can lead to spurious effects

(Liang and Molkentin, 2003; Cohen, 1997), and therefore it remains important to

investigate the features of endogenously-expressed protein kinases. The aims of this

study therefore, were to investigate the subcellular localisation, expression patterns,

substrate specificity and binding partners of SAPK3. In this way a better understanding

of the function that this protein kinase plays within the cell could be achieved.

7.1.1 Evaluating the expression and subcellular localisation of SAPK3 in cultured

cells

In Chapter 3, a SAPK3 monoclonal antibody was used to show that SAPK3

was expressed in the HEK293 cell line, as well as primary cultures of rat cardiac

myocytes. Using this antibody in immunostaining showed SAPK3 colocalisation with

the mitochondria (Chapter 3). Although the specificity of this antibody was assessed by

several means including Western blotting, screening a cDNA library for SAPK3 clones,

and pre-adsorption with recombinant SAPK3 before immunostaining to show a

reduction in staining, further studies will be required to show unequivocally that this

antibody only recognises SAPK3 in all protocols. For example, tissues from a SAPK3

knockout mouse could be analysed by Western blotting and immunostaining to show

that immunoreactivity is no longer seen. It may be possible to perform these

120

Page 213: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

experiments in the near future, since a SAPK3 knockout mouse has been developed (P.

Cohen and A. Cuenda, personal communication). Alternatively, RNA interference

studies to knock down SAPK3 expression in cultured cells such as those used in Chapter

3 could be employed to confirm the antibody specificity.

The co-localisation of SAPK3 with the mitochondria suggested that its

substrates may reside at this organelle. This was further supported by the observation

that the treatment of HEK293 fibroblasts with hyperosmolarity in the form of high

concentrations of sorbitol, which has previously been shown to activate overexpressed

SAPK3 in these cells (Cuenda et al., 1997), did not alter the localisation of this protein

kinase (results not shown). In future work, the expression of a constitutively-active

MKK6 mutant (Alonso et al., 2000) could be used as an alternative method to activate

SAPK3. It would then be possible to evaluate the localisation patterns of activated

SAPK3 in this situation. Alternatively, the localisation of SAPK3 substrates may be

altered in response to specific stimuli. Thus SAPK3 substrates that are predominantly in

locations such as the cell cytosol could be re-localised to the mitochondria. Once at the

mitochondria, these proteins might then be phosphorylated by SAPK3, also at this

location. An example of this movement of a protein kinase substrate rather than a

protein kinase itself comes with sorbitol treatment and the localisation of Tau that

changes from cytoskeletal to the soluble fractions of the cell (Jenkins et al., 2000). If

such changes occurred in the SAPK3 signalling pathway, these would need to be

confirmed with antibodies directed against protein substrates of SAPK3 rather than

SAPK3 itself.

The treatment of cells with the nuclear export inhibitor Leptomycin B did

not alter the localisation of SAPK3, unlike SAPK2a that was retained in the nucleus

(Figure 3.4). However, Leptomycin B only inhibits Crm-1 receptor-dependent nuclear

transport (Fukuda et al., 1997c; Fornerod et al., 1997; Ossareh-Nazari et al., 1997) and

therefore it is possible that SAPK3 could translocate between the nucleus and cytosol by

a Crm-1 receptor-independent mechanism. For example, the 45kDa form of T-cell

Protein Tyrosine Phosphatase (TCPTP), TC45, appears to be exported from the nucleus

121

Page 214: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

via passive diffusion (Lam et al., 2001). However further evidence supports that it is

unlikely that SAPK3 translocates to the nucleus. Specifically, active SAPK3 does not

homodimerise (Bellon et al., 1999), but dimerisation is thought to be essential for

nuclear translocation of ERK (Khokhlatchev et al., 1998). If SAPK3 is to move to the

nucleus under stress conditions, it would therefore require a mechanism distinct from

that used by ERK MAPKs and likely to be Crm-1 independent.

7.1.2 SAPK3 expression in the intact heart and evaluation of its role in cardiac

development and disease

In Chapter 4, SAPK3 was shown to be expressed in the heart. This

expression was not altered during ischemic heart disease or dilated cardiomyopathy, or

during pre- and post-natal cardiac development. In addition, the activation of SAPK3 in

these samples could not be detected (Figure 4.5 and results not shown). As discussed in

previous Chapters, there are a number of possible explanations for these observations.

First this lack of detection of phospho-SAPK3 could be due to the lower expression of

SAPK3 in the heart compared to SAPK2a (Lemke et al., 2001). Therefore

phosphorylation of SAPK2a would be most easily detected under the conditions

employed in Western blotting. However it is also possible that sequence differences

between SAPK2a and SAPK3 could result in the phospho-p38 MAPK antibody showing

higher immunoreactivity with SAPK2a. The activation loop sequences of human

SAPK2a and SAPK3 are given below:

SAPK2a: GLARHTDDEMTGYVATRWYRAPE |||| | |||||| |||||||| SAPK3: GLARQADSEMTGYVVTRWYRAPE

Therefore in the 20 amino acids shown here surrounding the TGY motif in subdomain

VIII, 20% of these amino acids are not identical in both SAPK2a and SAPK3. It would

therefore be an advantage to develop an antibody that specifically recognises the

phosphorylated form of SAPK3 whilst not reacting with other p38 MAPKs. This would

entail the use of a dual-phosphorylated 23-mer peptide based specifically on the SAPK3

122

Page 215: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

sequence with subsequent affinity purification to select for the phospho-SAPK3

sequence and to also select against the phospho-SAPK2a sequence. This antibody could

then be potentially used, for example, in samples of stressed cultured cells or heart

failure samples, or to analyse patterns of SAPK3 phosphorylation in the embryonic

mouse. These experiments would thus pinpoint the in vitro stimuli that could activate

SAPK3, whether SAPK3 activation accompanies the development of cardiovascular

disease and progression to failure, and in the case of the embryonic samples, tissues in

which SAPK3 may play a crucial signalling role in development. This latter approach

has recently been used with phospho-ERK antibodies to determine the patterns of

fibroblast growth factor (FGF)-dependent and FGF-non-dependent ERK signalling

during embryonic development (Corson et al., 2003). These immunostaining

experiments for active SAPK3 would ideally also include the use of a secondary

antibody conjugated to a fluorophore that does not show cross-reactivity with

immunoglobulins in the mouse tissues. Although colourimetric staining reactions like

that used in Figures 4.8, 4.9 and 4.10 with alkaline phosphatase are useful for

determining the overall pattern of SAPK3 distribution, they are not appropriate for

determining the subcellular localisation of proteins. This is because the coloured

precipitate formed during signal detection can diffuse away from the position of the

primary antibody and thus does not give an accurate representation of the position of the

antigen (Corson et al., 2003).

7.1.3 Determining the substrate specificity of SAPK3

The identification of protein kinase substrates remains one of the biggest

challenges in the field of cell signalling. It is essential to determine the role of each

substrate because these will be the mediators of the actions of each protein kinase within

the cell (Berwick and Tavare, 2004; Cohen, 1997). In Chapter 5, the ability of SAPK3

to phosphorylate a series of mitochondrial proteins was assessed. By using activated

SAPK3 in in vitro kinase assays, it was found that SAPK3 could phosphorylate the

mitochondrial proteins Bcl-2, Bag-1, Bim, Bad and Sab. Interestingly, all of these

proteins have been implicated in the process of apoptosis. Specifically, Bcl-2 and Bag-1

123

Page 216: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

are anti-apoptotic proteins, whilst Bim and Bad are pro-apoptotic (Hockenbery et al.,

1990; Takayama et al., 1995; O'Connor et al., 1998; Yang et al., 1995). One report has

suggested that Sab plays an anti-apoptotic role, since the overexpression of this protein

in the DT40 chicken B lymphoma cell line slowed B cell antigen receptor apoptosis

(Yamadori et al., 1999). It remains to be determined whether this anti-apoptotic role of

Sab is modulated by its phosphorylation. Furthermore it will be important to map the

phosphorylation sites on proteins such as Bcl-2, Bag-1, Bim and Bad, and ascertain how

phosphorylation at these sites alters their functions when these proteins are

endogenously expressed.

It is interesting to note that both SAPK3 and JNK phosphorylate Sab, as

well as members of the Bcl-2 family (Chapter 5) (Wiltshire et al., 2002; Donovan et al.,

2002; Deng et al., 2001; Lei and Davis, 2003). The mitochondrial localisation of both of

these kinases suggests that they may share common targets in vivo. Indeed, several

reports have shown specific activation of SAPK3 and JNK without activation of other

MAPKs. For example, the MKKK, MRK, preferentially activates SAPK3 and JNK via

MKK3/6 and MKK4/7 respectively (Gross et al., 2002). Also, the transfection of cells

with the MKKKK, PAPK, in conjunction with members of the MAPK family leads to

the activation of SAPK3 and JNK, but not other MAPKs (Nishigaki et al., 2003).

Furthermore, marathon running transiently increased JNK and SAPK3 activity in human

skeletal muscle (Boppart et al., 2000). This suggests that JNK and SAPK3 may be

coordinately activated despite their apparent lack of common upstream activators.

Specifically JNK activation relies on activation of MKK4 and MKK7 (Derijard et al.,

1995; Yao et al., 1997), whereas SAPK3 activation relies on MKK6 and to a lesser

extent MKK3 (Cuenda et al., 1997; Enslen et al., 1998).

One future challenge will be to determine whether any of the proteins

identified in Chapter 5 are physiological substrates of SAPK3 and if so, the consequence

of this phosphorylation. As discussed in Chapter 5, the phosphorylation of Bcl-2 family

members such as Bad is complex, with phosphorylation on some sites promoting

apoptotis (Donovan et al., 2002; Konishi et al., 2002), and on others leading to an anti-

124

Page 217: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

apoptotic response (Zha et al., 1996). Therefore, the identification of these proteins as

substrates of SAPK3 is not sufficient to implicate SAPK3 in a role for cell survival or

cell apoptosis and needs to be expanded in the future with functional studies using

phosphorylation site mutants of these proteins.

To determine whether SAPK3 is able to phosphorylate any of these

substrates in the intact cell, one approach would be to transfect cells with MKK6(DD)

and determine the effects from SAPK3 activation. The use of the SB203580 inhibitor

could then be used to prevent the subsequent activities of SAPK2a/b. However, it is still

possible that the effects seen are mediated by MKK6-activated kinases other than

SAPK3, such as SAPK4. One way to circumvent this would be to develop a specific

SAPK3 inhibitor. Alternatively, there is the possible development of a constitutively

active SAPK3 mutant. It has previously been found that mutation of Thr183 to alanine

or Tyr185 to phenylalanine in the activation loop of ERK2 abolishes kinase activity

(Robbins et al., 1993). Furthermore, mutation of these amino acids to glutamate, which

can mimic phosphorylation and thus activates some kinases, does not confer activity

(Robbins et al., 1993; Canagarajah et al., 1997; Mansour et al., 1994). However, a

genetic screen of S. cerevisiae mutants has identified a number of hyperactive mutants of

the SAPK2a homologue, Hog1 (Bell et al., 2001). Most of the mutations were found in

a domain at the C-terminal extension of the kinase known as L16 (see Figure 1.3) (Bell

et al., 2001). Furthermore, combinations of these mutations resulted in even higher

catalytic activity of Hog1 (Yaakov et al., 2003). Therefore, it is possible that similar

mutations may be able to activate other members of the MAPK family such as SAPK3.

This will require the creation of SAPK3 mutants based on these results with Hog1,

followed by extensive evaluation of their activity. Initial studies would require

assessment of their activity in vitro towards substrates such as Sab and α1-Syntrophin.

These constitutively active mutants could then be used in experiments in cells or in vivo.

An additional way to identify SAPK3 substrates would be to further define

its phosphorylation site preferences. The results presented in Chapter 5 showed that

amino acids at the +2 and +3 position of the Ser321 phosphorylation site in Sab were

125

Page 218: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

important for recognition by SAPK3. The reduction of the Sab protein to a short peptide

encompassing this site and sequential replacement of each amino acid in this peptide

may help to further define SAPK3 substrate specificity. In several cases, peptide

substrates of MAPKs have not shown to be effective (Jacobs et al., 1999; Gavin and

Nebreda, 1999). This is thought to be a result of the lack of appropriate targeting

domains (Holland and Cooper, 1999). The sequences in Sab however may overcome

this with a short linear sequence containing both a targeting domain (KIM1) and the

phosphorylation sequence (Ser321).

Alternatively, oriented peptide libraries may be used to identify the optimal

consensus site for SAPK3 phosphorylation. This approach has been used extensively to

define the substrate specificity of kinases such as Protein Kinase A and Akt (Songyang

et al., 1994; Obata et al., 2000). However, these results must always be put into the

context of the whole protein. For example, as shown in Chapter 5, the mutation of

amino acids surrounding Ser391 in the Sab protein to those normally surrounding

Ser321 did not result in the same degree of phosphorylation as that seen with Ser321,

showing that the position of Ser321 within the Sab protein was important (Figure 5.13).

In addition, there was a requirement for the KIM1 of Sab for phosphorylation by SAPK3

(Figure 5.10).

In Chapter 5, activated SAPK3 was used in in vitro kinase assays to

directly test its phosphorylation of potential substrate proteins. Other possible in vitro

approaches also exist to identify substrates of SAPK3. For example, one group

incubated subfractionated cell extracts with [γ-32P]-ATP of high specific radioactivity,

together with activated, GST-tagged versions of SAPK2a, SAPK3 or SAPK4 (Knebel et

al., 2001). The resultant phosphorylated proteins were then identified by purification

and tryptic mass fingerprinting. This method was used to identify eukaryotic elongation

factor 2 kinase as a substrate of SAPK4 (Knebel et al., 2001). Although approaches

such as this can be used to identify MAPK substrates, eventually in vivo approaches

need to be used to confirm that these are physiological substrates. For example, within

the cell, subcellular compartmentalisation, protein phosphatases and binding proteins

126

Page 219: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

may normally prevent certain kinase-substrate interactions from occurring (Berwick and

Tavare, 2004). One recent approach which partially overcomes this has involved

mutating a conserved bulky residue in the ATP-binding site of ERK2 to small amino

acids such as glycine and alanine, to enlarge the ATP binding pocket (Eblen et al.,

2003). This mutant was then transfected into cells, activated by EGF-stimulation, and

immunoprecipitated under non-stringent conditions to isolate any associated binding

proteins. These immunoprecipitates were then incubated with an ATP analogue with a

bulky side chain attached that could only be used by the mutant protein kinase (Eblen et

al., 2003). Proteins specifically phosphorylated by the mutant kinase could then be

identified by mass spectrometry (Eblen et al., 2003).

7.1.4 Searching for binding partners of SAPK3

In Chapter 6, it was shown that SAPK3 could bind to the PDZ domain-

containing protein OMP25, and that OMP25 could act as an inhibitor of SAPK3 activity.

Future experiments will be required with OMP25 antibodies and knockout models of

OMP25 and SAPK3 to determine whether these proteins interact in vivo. For example,

as discussed in Chapter 6, the knockdown of OMP25 would be predicted to result in the

loss of mitochondrial co-localisation of SAPK3 if OMP25 is a dominant binding partner

for SAPK3. To gain a better understanding of the interaction that occurs between

OMP25 and SAPK3, it would also be informative to co-crystallise these proteins.

Previous reports have only shown crystallisation of PDZ domains with peptide ligands,

rather than full-length proteins (Figure 1.7) (Doyle et al., 1996; Daniels et al., 1998).

This will help to decipher whether the inhibition of SAPK3 activity arises from OMP25

not only occupying the C-terminal PDZ domain binding motif (to inhibit SAPK3

phosphorylation of PDZ domain-containing substrates), but also potentially covering the

C-terminal substrate binding groove that is crucial for the binding of MAPKs to the

KIMs of their substrates (Tanoue et al., 2000).

The interaction of SAPK3 with OMP25 does raise the question: how does

SAPK3 signal within the cell if it is being kept inactive by OMP25? It would appear

127

Page 220: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

that the release of SAPK3 from this interaction would be needed. A possible release

mechanism might include the phosphorylation of the SAPK3 C-terminal tail by another

protein kinase, to release the PDZ interaction. For example, the phosphorylation of the

-2 serine of the inward rectifier K+ channel Kir 2.3 by Protein Kinase A (PKA) prevents

its association with the PDZ domain of PSD-95 (Cohen et al., 1996). In another

example, phosphorylation of the -2 serine of the β2-adrenergic receptor by G-protein

coupled receptor kinase-5 disrupts interaction with the PDZ domain of NHERF (Cao et

al., 1999). The C-terminal sequence of SAPK3 is Lys-Glu-Thr-Xaa-Leu-COOH, where

Xaa is Ala in mouse and rat, or Pro in human. It is possible that the threonine residue at

the -2 position (Thr365) is phosphorylated and that this could release SAPK3 from a

PDZ-mediated interaction. Therefore, future work could involve mutation of this

threonine to a glutamic or aspartic acid residue, to mimic phosphorylation. The SAPK3

mutant would then not be expected to bind OMP25. Although this mutation would also

likely affect the interaction of SAPK3 with PDZ domain-containing proteins such as α1-

Syntrophin, it would still be expected to show activity towards non-PDZ domain-

containing substrate proteins such as Sab. Examination of the sequence surrounding

Thr365 shows similarity to the cyclic GMP-dependent protein kinase consensus site for

phosphorylation (Pearson and Kemp, 1991). Further studies will be required to

determine whether this site is phosphorylated by cyclic GMP-dependent protein kinase

or another protein kinase.

7.2 Conclusions

At the commencement of these studies, little was known about the role that

SAPK3 may play in the cell. The aim of this project, therefore, was to characterise some

of the features of SAPK3, including its tissue expression, localisation, substrate

specificity, and binding partners. Collectively, the results presented in this thesis

together with the published literature in the last 4 years suggest that SAPK3 is a unique

member of the p38 MAPK family. This is supported by its unique tissue distribution,

subcellular localisation, and binding to PDZ domain-containing proteins. These results

in combination with further studies utilising SAPK3 knockout models and specific

128

Page 221: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

SAPK3 inhibitors should allow the ultimate function of this protein kinase to be

determined.

129

Page 222: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Chapter 8

References

Page 223: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

References

Abdollahi,T., Robertson,N.M., Abdollahi,A., and Litwack,G. (2003). Identification of interleukin 8 as an inhibitor of tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis in the ovarian carcinoma cell line OVCAR3. Cancer Res 63, 4521-4526.

Adachi,M., Fukuda,M., and Nishida,E. (2000). Nuclear export of MAP kinase (ERK) involves a MAP kinase kinase (MEK)-dependent active transport mechanism. J Cell Biol 148, 849-856.

Adams,J.W., Sakata,Y., Davis,M.G., Sah,V.P., Wang,Y., Liggett,S.B., Chien,K.R., Brown,J.H., and Dorn,G.W. (1998). Enhanced Galphaq signaling: a common pathway mediates cardiac hypertrophy and apoptotic heart failure. Proc Natl Acad Sci U S A 95, 10140-10145.

Ahn,N.G., Seger,R., Bratlien,R.L., Diltz,C.D., Tonks,N.K., and Krebs,E.G. (1991). Multiple components in an epidermal growth factor-stimulated protein kinase cascade. In vitro activation of a myelin basic protein/microtubule-associated protein 2 kinase. J Biol Chem. 266, 4220-4227.

Alessandrini,A., Namura,S., Moskowitz,M.A., and Bonventre,J.V. (1999). MEK1 protein kinase inhibition protects against damage resulting from focal cerebral ischemia. Proc Natl Acad Sci U S A 96, 12866-12869.

Alonso,G., Ambrosino,C., Jones,M., and Nebreda,A.R. (2000). Differential activation of p38 mitogen-activated protein kinase isoforms depending on signal strength. J Biol Chem. 275, 40641-40648.

Alvarez,E., Northwood,I.C., Gonzalez,F.A., Latour,D.A., Seth,A., Abate,C., Curran,T., and Davis,R.J. (1991). Pro-Leu-Ser/Thr-Pro is a consensus primary sequence for substrate protein phosphorylation. Characterization of the phosphorylation of c-myc and c-jun proteins by an epidermal growth factor receptor threonine 669 protein kinase. J Biol Chem. 266, 15277-15285.

Anderson,J.M., Stevenson,B.R., Jesaitis,L.A., Goodenough,D.A., and Mooseker,M.S. (1988). Characterization of ZO-1, a protein component of the tight junction from mouse liver and Madin-Darby canine kidney cells. J Cell Biol 106, 1141-1149.

Anguita,J., Barthold,S.W., Persinski,R., Hedrick,M.N., Huy,C.A., Davis,R.J., Flavell,R.A., and Fikrig,E. (2002). Murine Lyme arthritis development mediated by p38 mitogen-activated protein kinase activity. J Immunol. 168, 6352-6357.

130

Page 224: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Aoki,H., Kang,P.M., Hampe,J., Yoshimura,K., Noma,T., Matsuzaki,M., and Izumo,S. (2002). Direct activation of mitochondrial apoptosis machinery by c-Jun N-terminal kinase in adult cardiac myocytes. J Biol Chem. 277, 10244-10250.

Assoian,R.K. (2002). Common sense signalling. Nat. Cell Biol 4, E187-E188.

Barr,R.K. and Bogoyevitch,M.A. (2001). The c-Jun N-terminal protein kinase family of mitogen-activated protein kinases (JNK MAPKs). Int. J Biochem Cell Biol 33, 1047-1063.

Barros,L.F., Young,M., Saklatvala,J., and Baldwin,S.A. (1997). Evidence of two mechanisms for the activation of the glucose transporter GLUT1 by anisomycin: p38(MAP kinase) activation and protein synthesis inhibition in mammalian cells. J Physiol 504 ( Pt 3), 517-525.

Basu,A. and Haldar,S. (2003). Identification of a novel Bcl-xL phosphorylation site regulating the sensitivity of taxol- or 2-methoxyestradiol-induced apoptosis. FEBS Lett 538, 41-47.

Behr,T.M., Nerurkar,S.S., Nelson,A.H., Coatney,R.W., Woods,T.N., Sulpizio,A., Chandra,S., Brooks,D.P., Kumar,S., Lee,J.C., Ohlstein,E.H., Angermann,C.E., Adams,J.L., Sisko,J., Sackner-Bernstein,J.D., and Willette,R.N. (2001). Hypertensive end-organ damage and premature mortality are p38 mitogen-activated protein kinase-dependent in a rat model of cardiac hypertrophy and dysfunction. Circulation 104, 1292-1298.

Bell,M., Capone,R., Pashtan,I., Levitzki,A., and Engelberg,D. (2001). Isolation of hyperactive mutants of the MAPK p38/Hog1 that are independent of MAPK kinase activation. J Biol Chem. 276, 25351-25358.

Bellon,S., Fitzgibbon,M.J., Fox,T., Hsiao,H.M., and Wilson,K.P. (1999). The structure of phosphorylated p38gamma is monomeric and reveals a conserved activation-loop conformation. Structure. Fold. Des 7, 1057-1065.

Ben Levy,R., Hooper,S., Wilson,R., Paterson,H.F., and Marshall,C.J. (1998). Nuclear export of the stress-activated protein kinase p38 mediated by its substrate MAPKAP kinase-2. Curr. Biol 8, 1049-1057.

Berwick,D.C. and Tavare,J.M. (2004). Identifying protein kinase substrates: hunting for the organ-grinder's monkeys. Trends Biochem Sci 29, 227-232.

Bhakar,A.L., Howell,J.L., Paul,C.E., Salehi,A.H., Becker,E.B., Said,F., Bonni,A., and Barker,P.A. (2003). Apoptosis induced by p75NTR overexpression requires Jun kinase-dependent phosphorylation of Bad. J Neurosci. 23, 11373-11381.

Bilder,D. and Perrimon,N. (2000). Localization of apical epithelial determinants by the basolateral PDZ protein Scribble. Nature 403, 676-680.

131

Page 225: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Boglari,G., Erhardt,P., Cooper,G.M., and Szeberenyi,J. (1998). Intact Ras function is required for sustained activation and nuclear translocation of extracellular signal-regulated kinases in nerve growth factor-stimulated PC12 cells. Eur. J Cell Biol 75, 54-58.

Bogoyevitch MA and Court NW (2004). Counting on Mitogen-activated protein kinases - ERKs 3, 4, 5, 6, 7 and 8. Cell Signal. In Press.

Bogoyevitch,M.A., Gillespie-Brown,J., Ketterman,A.J., Fuller,S.J., Ben Levy,R., Ashworth,A., Marshall,C.J., and Sugden,P.H. (1996). Stimulation of the stress-activated mitogen-activated protein kinase subfamilies in perfused heart. p38/RK mitogen-activated protein kinases and c-Jun N-terminal kinases are activated by ischemia/reperfusion. Circ. Res 79, 162-173.

Bogoyevitch,M.A., Glennon,P.E., Andersson,M.B., Clerk,A., Lazou,A., Marshall,C.J., Parker,P.J., and Sugden,P.H. (1994). Endothelin-1 and fibroblast growth factors stimulate the mitogen-activated protein kinase signaling cascade in cardiac myocytes. The potential role of the cascade in the integration of two signaling pathways leading to myocyte hypertrophy. J Biol Chem. 269, 1110-1119.

Bogoyevitch,M.A., Glennon,P.E., and Sugden,P.H. (1993). Endothelin-1, phorbol esters and phenylephrine stimulate MAP kinase activities in ventricular cardiomyocytes. FEBS Lett 317, 271-275.

Boppart,M.D., Asp,S., Wojtaszewski,J.F., Fielding,R.A., Mohr,T., and Goodyear,L.J. (2000). Marathon running transiently increases c-Jun NH2-terminal kinase and p38 activities in human skeletal muscle. J Physiol 526 Pt 3, 663-669.

Boulton TG, Nye SH, Robbins DJ, Ip NY, Radziejewska E, Morgenbesser SD, DePinho RA, Panayotatos N, Cobb MH, and Yancopoulos GD. (1991). ERKs: a family of protein-serine/threonine kinases that are activated and tyrosine phosphorylated in response to insulin and NGF. Cell 65, 663-675.

Boulton,T.G., Yancopoulos,G.D., Gregory,J.S., Slaughter,C., Moomaw,C., Hsu,J., and Cobb,M.H. (1990). An insulin-stimulated protein kinase similar to yeast kinases involved in cell cycle control. Science 249, 64-67.

Bour,B.A., O'Brien,M.A., Lockwood,W.L., Goldstein,E.S., Bodmer,R., Taghert,P.H., Abmayr,S.M., and Nguyen,H.T. (1995). Drosophila MEF2, a transcription factor that is essential for myogenesis. Genes Dev. 9 , 730-741.

Braz,J.C., Bueno,O.F., De Windt,L.J., and Molkentin,J.D. (2002). PKC alpha regulates the hypertrophic growth of cardiomyocytes through extracellular signal-regulated kinase1/2 (ERK1/2). J Cell Biol 156, 905-919.

Braz,J.C., Bueno,O.F., Liang,Q., Wilkins,B.J., Dai,Y.S., Parsons,S., Braunwart,J., Glascock,B.J., Klevitsky,R., Kimball,T.F., Hewett,T.E., and Molkentin,J.D. (2003).

132

Page 226: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Targeted inhibition of p38 MAPK promotes hypertrophic cardiomyopathy through upregulation of calcineurin-NFAT signaling. J Clin. Invest 111, 1475-1486.

Brinkworth,R.I., Breinl,R.A., and Kobe,B. (2003). Structural basis and prediction of substrate specificity in protein serine/threonine kinases. Proc Natl Acad Sci U S A 100, 74-79.

Buday,L. and Downward,J. (1993). Epidermal growth factor regulates p21ras through the formation of a complex of receptor, Grb2 adapter protein, and Sos nucleotide exchange factor. Cell 73, 611-620.

Buee-Scherrer and Goedert M. (2002). Phosphorylation of microtubule-associated protein tau by stress-activated protein kinases in intact cells. FEBS Lett. 515, 151-154.

Bueno,O.F., De Windt,L.J., Tymitz,K.M., Witt,S.A., Kimball,T.R., Klevitsky,R., Hewett,T.E., Jones,S.P., Lefer,D.J., Peng,C.F., Kitsis,R.N., and Molkentin,J.D. (2000). The MEK1-ERK1/2 signaling pathway promotes compensated cardiac hypertrophy in transgenic mice. EMBO J 19, 6341-6350.

Bueno,O.F. and Molkentin,J.D. (2002). Involvement of extracellular signal-regulated kinases 1/2 in cardiac hypertrophy and cell death. Circ. Res 91, 776-781.

Camps,M., Nichols,A., and Arkinstall,S. (2000). Dual specificity phosphatases: a gene family for control of MAP kinase function. FASEB J 14, 6-16.

Canagarajah,B.J., Khokhlatchev,A., Cobb,M.H., and Goldsmith,E.J. (1997). Activation mechanism of the MAP kinase ERK2 by dual phosphorylation. Cell 90, 859-869.

Cao,T.T., Deacon,H.W., Reczek,D., Bretscher,A., and von Zastrow,M. (1999). A kinase-regulated PDZ-domain interaction controls endocytic sorting of the beta2-adrenergic receptor. Nature 401, 286-290.

Casey,P.J., Solski,P.A., Der,C.J., and Buss,J.E. (1989). p21ras is modified by a farnesyl isoprenoid. Proc Natl Acad Sci U S A 86, 8323-8327.

Cavigelli,M., Dolfi,F., Claret,F.X., and Karin,M. (1995). Induction of c-fos expression through JNK-mediated TCF/Elk-1 phosphorylation. EMBO J 14, 5957-5964.

Chang,C.I., Xu,B.E., Akella,R., Cobb,M.H., and Goldsmith,E.J. (2002). Crystal structures of MAP kinase p38 complexed to the docking sites on its nuclear substrate MEF2A and activator MKK3b. Mol Cell 9, 1241-1249.

Chardin,P., Camonis,J.H., Gale,N.W., van Aelst,L., Schlessinger,J., Wigler,M.H., and Bar-Sagi,D. (1993). Human Sos1: a guanine nucleotide exchange factor for Ras that binds to GRB2. Science 260, 1338-1343.

Chen,R.H., Sarnecki,C., and Blenis,J. (1992). Nuclear localization and regulation of erk- and rsk-encoded protein kinases. Mol Cell Biol 12, 915-927.

133

Page 227: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Chen,Y.W., Zhao,P., Borup,R., and Hoffman,E.P. (2000). Expression profiling in the muscular dystrophies: identification of novel aspects of molecular pathophysiology. J Cell Biol 151, 1321-1336.

Chiariello,M., Marinissen,M.J., and Gutkind,J.S. (2000). Multiple mitogen-activated protein kinase signaling pathways connect the cot oncoprotein to the c-jun promoter and to cellular transformation. Mol Cell Biol 20, 1747-1758.

Chien,K.R. (1999). Stress pathways and heart failure. Cell 98, 555-558.

Chien,K.R., Knowlton,K.U., Zhu,H., and Chien,S. (1991). Regulation of cardiac gene expression during myocardial growth and hypertrophy: molecular studies of an adaptive physiologic response. FASEB J 5, 3037-3046.

Chihab,R., Bossenmeyer,C., Oillet,J., and Daval,J.L. (1998). Lack of correlation between the effects of transient exposure to glutamate and those of hypoxia/reoxygenation in immature neurons in vitro. J Neurochem. 71, 1177-1186.

Cho,K.O., Hunt,C.A., and Kennedy,M.B. (1992). The rat brain postsynaptic density fraction contains a homolog of the Drosophila discs-large tumor suppressor protein. Neuron 9, 929-942.

Clark-Lewis,I., Sanghera,J.S., and Pelech,S.L. (1991). Definition of a consensus sequence for peptide substrate recognition by p44mpk, the meiosis-activated myelin basic protein kinase. J Biol Chem. 266, 15180-15184.

Clerk,A., Harrison,J.G., Long,C.S., and Sugden,P.H. (1999). Pro-inflammatory cytokines stimulate mitogen-activated protein kinase subfamilies, increase phosphorylation of c-Jun and ATF2 and upregulate c-Jun protein in neonatal rat ventricular myocytes. J Mol Cell Cardiol. 31, 2087-2099.

Clerk,A., Michael,A., and Sugden,P.H. (1998a). Stimulation of multiple mitogen-activated protein kinase sub-families by oxidative stress and phosphorylation of the small heat shock protein, HSP25/27, in neonatal ventricular myocytes. Biochem J 333 (Pt 3), 581-589.

Clerk,A., Michael,A., and Sugden,P.H. (1998b). Stimulation of the p38 mitogen-activated protein kinase pathway in neonatal rat ventricular myocytes by the G protein-coupled receptor agonists, endothelin-1 and phenylephrine: a role in cardiac myocyte hypertrophy? J Cell Biol 142, 523-535.

Clerk,A. and Sugden,P.H. (1999). Activation of protein kinase cascades in the heart by hypertrophic G protein-coupled receptor agonists. Am. J Cardiol. 83, 64H-69H.

Cobb,M.H., Hepler,J.E., Cheng,M., and Robbins,D. (1994). The mitogen-activated protein kinases, ERK1 and ERK2. Semin. Cancer Biol 5, 261-268.

134

Page 228: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Cohen,N.A., Brenman,J.E., Snyder,S.H., and Bredt,D.S. (1996). Binding of the inward rectifier K+ channel Kir 2.3 to PSD-95 is regulated by protein kinase A phosphorylation. Neuron 17, 759-767.

Cohen, P. (1997). The search for physiological substrates of MAP and SAP kinases in mammalian cells. Trends Cell Biol. 7, 353-361.

Cohen,P. (2002). Protein kinases--the major drug targets of the twenty-first century? Nat. Rev. Drug Discov. 1, 309-315.

Conrad,P.W., Millhorn,D.E., and Beitner-Johnson,D. (2000). Novel regulation of p38gamma by dopamine D2 receptors during hypoxia. Cell Signal. 12, 463-467.

Conrad,P.W., Rust,R.T., Han,J., Millhorn,D.E., and Beitner-Johnson,D. (1999). Selective activation of p38alpha and p38gamma by hypoxia. Role in regulation of cyclin D1 by hypoxia in PC12 cells. J Biol Chem. 274, 23570-23576.

Cook,S.A., Sugden,P.H., and Clerk,A. (1999). Activation of c-Jun N-terminal kinases and p38-mitogen-activated protein kinases in human heart failure secondary to ischaemic heart disease. J Mol Cell Cardiol. 31, 1429-1434.

Corson,L.B., Yamanaka,Y., Lai,K.M., and Rossant,J. (2003). Spatial and temporal patterns of ERK signaling during mouse embryogenesis. Development 130, 4527-4537.

Court NW, dos Remedios CG, Cordell J, and Bogoyevitch MA. (2002). Cardiac expression and subcellular localization of the p38 mitogen-activated protein kinase member, stress-activated protein kinase-3 (SAPK3). J Mol Cell Cardiol. 34, 413-426.

Cowley,S., Paterson,H., Kemp,P., and Marshall,C.J. (1994). Activation of MAP kinase kinase is necessary and sufficient for PC12 differentiation and for transformation of NIH 3T3 cells. Cell 77, 841-852.

Cuenda,A., Cohen,P. (1999). Stress-activated protein kinase-2/p38 and a rapamycin-sensitive pathway are required for C2C12 myogenesis. J Biol Chem 274, 4341-4346.

Cuenda,A., Cohen,P., Buee-Scherrer,V., and Goedert,M. (1997). Activation of stress-activated protein kinase-3 (SAPK3) by cytokines and cellular stresses is mediated via SAPKK3 (MKK6); comparison of the specificities of SAPK3 and SAPK2 (RK/p38). EMBO J 16, 295-305.

Cuenda,A., Rouse,J., Doza,Y.N., Meier,R., Cohen,P., Gallagher,T.F., Young,P.R., and Lee,J.C. (1995). SB 203580 is a specific inhibitor of a MAP kinase homologue which is stimulated by cellular stresses and interleukin-1. FEBS Lett 364, 229-233.

Danial,N.N., Gramm,C.F., Scorrano,L., Zhang,C.Y., Krauss,S., Ranger,A.M., Datta,S.R., Greenberg,M.E., Licklider,L.J., Lowell,B.B., Gygi,S.P., and Korsmeyer,S.J. (2003). BAD and glucokinase reside in a mitochondrial complex that integrates glycolysis and apoptosis. Nature 424, 952-956.

135

Page 229: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Daniels,D.L., Cohen,A.R., Anderson,J.M., and Brunger,A.T. (1998). Crystal structure of the hCASK PDZ domain reveals the structural basis of class II PDZ domain target recognition. Nat. Struct. Biol 5, 317-325.

Dash,R., Schmidt,A.G., Pathak,A., Gerst,M.J., Biniakiewicz,D., Kadambi,V.J., Hoit,B.D., Abraham,W.T., and Kranias,E.G. (2003). Differential regulation of p38 mitogen-activated protein kinase mediates gender-dependent catecholamine-induced hypertrophy. Cardiovasc. Res 57, 704-714.

Davidson,S.M. and Morange,M. (2000). Hsp25 and the p38 MAPK pathway are involved in differentiation of cardiomyocytes. Dev. Biol 218, 146-160.

Davis,S., Vanhoutte,P., Pages,C., Caboche,J., and Laroche,S. (2000). The MAPK/ERK cascade targets both Elk-1 and cAMP response element-binding protein to control long-term potentiation-dependent gene expression in the dentate gyrus in vivo. J Neurosci. 20, 4563-4572.

De Angelis,L., Borghi,S., Melchionna,R., Berghella,L., Baccarani-Contri,M., Parise,F., Ferrari,S., and Cossu,G. (1998). Inhibition of myogenesis by transforming growth factor beta is density-dependent and related to the translocation of transcription factor MEF2 to the cytoplasm. Proc Natl Acad Sci U S A 95, 12358-12363.

De Windt,L.J., Lim,H.W., Haq,S., Force,T., and Molkentin,J.D. (2000). Calcineurin promotes protein kinase C and c-Jun NH2-terminal kinase activation in the heart. Cross-talk between cardiac hypertrophic signaling pathways. J Biol Chem. 275, 13571-13579.

Deng,X., Xiao,L., Lang,W., Gao,F., Ruvolo,P., and May,W.S., Jr. (2001). Novel role for JNK as a stress-activated Bcl2 kinase. J Biol Chem. 276, 23681-23688.

Deng,Y., Ren,X., Yang,L., Lin,Y., and Wu,X. (2003). A JNK-dependent pathway is required for TNFalpha-induced apoptosis. Cell 115, 61-70.

Denhardt,D.T. (1996). Signal-transducing protein phosphorylation cascades mediated by Ras/Rho proteins in the mammalian cell: the potential for multiplex signalling. Biochem J 318 (Pt 3), 729-747.

Derijard,B., Hibi,M., Wu,I.H., Barrett,T., Su,B., Deng,T., Karin,M., and Davis,R.J. (1994). JNK1: a protein kinase stimulated by UV light and Ha-Ras that binds and phosphorylates the c-Jun activation domain. Cell 76, 1025-1037.

Derijard,B., Raingeaud,J., Barrett,T., Wu,I.H., Han,J., Ulevitch,R.J., and Davis,R.J. (1995). Independent human MAP-kinase signal transduction pathways defined by MEK and MKK isoforms. Science 267, 682-685.

Dhanasekaran,N. and Premkumar,R.E. (1998). Signaling by dual specificity kinases. Oncogene 17, 1447-1455.

136

Page 230: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Donovan,N., Becker,E.B., Konishi,Y., and Bonni,A. (2002). JNK phosphorylation and activation of BAD couples the stress-activated signaling pathway to the cell death machinery. J Biol Chem. 277, 40944-40949.

Doyle,D.A., Lee,A., Lewis,J., Kim,E., Sheng,M., and MacKinnon,R. (1996). Crystal structures of a complexed and peptide-free membrane protein-binding domain: molecular basis of peptide recognition by PDZ. Cell 85, 1067-1076.

Du,L., Lyle,C.S., Obey,T.B., Gaarde,W.A., Muir,J.A., Bennett,B.L., and Chambers,T.C. (2004). Inhibition of cell proliferation and cell cycle progression by specific inhibition of basal JNK activity: evidence that mitotic Bcl-2 phosphorylation is JNK-independent. J Biol Chem. 279, 11957-11966.

Eblen,S.T., Kumar,N.V., Shah,K., Henderson,M.J., Watts,C.K., Shokat,K.M., and Weber,M.J. (2003). Identification of novel ERK2 substrates through use of an engineered kinase and ATP analogs. J Biol Chem. 278, 14926-14935.

Enslen,H., Raingeaud,J., and Davis,R.J. (1998). Selective activation of p38 mitogen-activated protein (MAP) kinase isoforms by the MAP kinase kinases MKK3 and MKK6. J Biol Chem. 273, 1741-1748.

Eyers,P.A., Craxton,M., Morrice,N., Cohen,P., and Goedert,M. (1998). Conversion of SB 203580-insensitive MAP kinase family members to drug-sensitive forms by a single amino-acid substitution. Chem. Biol 5 , 321-328.

Fabre S, Reynaud C, and Jalinot P. (2000). Identification of functional PDZ domain binding sites in several human proteins. Mol Biol Rep. 27, 217-224.

Fan,M., Goodwin,M., Vu,T., Brantley-Finley,C., Gaarde,W.A., and Chambers,T.C. (2000). Vinblastine-induced phosphorylation of Bcl-2 and Bcl-XL is mediated by JNK and occurs in parallel with inactivation of the Raf-1/MEK/ERK cascade. J Biol Chem. 275, 29980-29985.

Fanning,A.S. and Anderson,J.M. (1999). PDZ domains: fundamental building blocks in the organization of protein complexes at the plasma membrane. J Clin. Invest 103, 767-772.

Fearns,C., Kline,L., Gram,H., Di Padova,F., Zurini,M., Han,J., and Ulevitch,R.J. (2000). Coordinate activation of endogenous p38alpha, beta, gamma, and delta by inflammatory stimuli. J Leukoc. Biol 67, 705-711.

Fields,S. and Song,O. (1989). A novel genetic system to detect protein-protein interactions. Nature 340, 245-246.

Fishman,M.C. and Chien,K.R. (1997). Fashioning the vertebrate heart: earliest embryonic decisions. Development 124, 2099-2117.

137

Page 231: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Fleming,Y., Armstrong,C.G., Morrice,N., Paterson,A., Goedert,M., and Cohen,P. (2000). Synergistic activation of stress-activated protein kinase 1/c-Jun N-terminal kinase (SAPK1/JNK) isoforms by mitogen-activated protein kinase kinase 4 (MKK4) and MKK7. Biochem J 352 Pt 1, 145-154.

Fornerod,M., Ohno,M., Yoshida,M., and Mattaj,I.W. (1997). CRM1 is an export receptor for leucine-rich nuclear export signals. Cell 90, 1051-1060.

Fox,T., Fitzgibbon,M.J., Fleming,M.A., Hsiao,H.M., Brummel,C.L., and Su,M.S. (1999). Kinetic mechanism and ATP-binding site reactivity of p38gamma MAP kinase. FEBS Lett 461, 323-328.

Fuh,G., Pisabarro,M.T., Li,Y., Quan,C., Lasky,L.A., and Sidhu,S.S. (2000). Analysis of PDZ domain-ligand interactions using carboxyl-terminal phage display. J Biol Chem. 275, 21486-21491.

Fukuda,M., Gotoh,Y., and Nishida,E. (1997a). Interaction of MAP kinase with MAP kinase kinase: its possible role in the control of nucleocytoplasmic transport of MAP kinase. EMBO J 16, 1901-1908.

Fukuda,M., Gotoh,I., Adachi,M., Gotoh,Y., and Nishida,E. (1997b). A novel regulatory mechanism in the mitogen-activated protein (MAP) kinase cascade. Role of nuclear export signal of MAP kinase kinase. J Biol Chem. 272, 32642-32648.

Fukuda,M., Asano,S., Nakamura,T., Adachi,M., Yoshida,M., Yanagida,M., and Nishida,E. (1997c). CRM1 is responsible for intracellular transport mediated by the nuclear export signal. Nature 390, 308-311.

Fukuda,M., Gotoh,I., Gotoh,Y., and Nishida,E. (1996). Cytoplasmic localization of mitogen-activated protein kinase kinase directed by its NH2-terminal, leucine-rich short amino acid sequence, which acts as a nuclear export signal. J Biol Chem. 271, 20024-20028.

Gavin,A.C. and Nebreda,A.R. (1999). A MAP kinase docking site is required for phosphorylation and activation of p90(rsk)/MAPKAP kinase-1. Curr. Biol 9, 281-284.

Ge,B., Gram,H., Di Padova,F., Huang,B., New,L., Ulevitch,R.J., Luo,Y., and Han,J. (2002). MAPKK-independent activation of p38alpha mediated by TAB1-dependent autophosphorylation of p38alpha. Science 295, 1291-1294.

Gee,S.H., Quenneville,S., Lombardo,C.R., and Chabot,J. (2000). Single-amino acid substitutions alter the specificity and affinity of PDZ domains for their ligands. Biochemistry 39, 14638-14646.

Gille,H., Kortenjann,M., Thomae,O., Moomaw,C., Slaughter,C., Cobb,M.H., and Shaw,P.E. (1995). ERK phosphorylation potentiates Elk-1-mediated ternary complex formation and transactivation. EMBO J 14, 951-962.

138

Page 232: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Goedert,M., Cuenda,A., Craxton,M., Jakes,R., and Cohen,P. (1997a). Activation of the novel stress-activated protein kinase SAPK4 by cytokines and cellular stresses is mediated by SKK3 (MKK6); comparison of its substrate specificity with that of other SAP kinases. EMBO J 16, 3563-3571.

Goedert,M., Hasegawa,M., Jakes,R., Lawler,S., Cuenda,A., and Cohen,P. (1997b). Phosphorylation of microtubule-associated protein tau by stress-activated protein kinases. FEBS Lett 409, 57-62.

Goswami,S., Qasba,P., Ghatpande,S., Carleton,S., Deshpande,A.K., Baig,M., and Siddiqui,M.A. (1994). Differential expression of the myocyte enhancer factor 2 family of transcription factors in development: the cardiac factor BBF-1 is an early marker for cardiogenesis. Mol Cell Biol 14 , 5130-5138.

Graves,J.D., Campbell,J.S., and Krebs,E.G. (1995). Protein serine/threonine kinases of the MAPK cascade. Ann N Y Acad Sci 766, 320-343.

Gross E.A., Callow M.G., Waldbaum L., Thomas S., and Ruggieri R. (2002). MRK, a mixed lineage kinase-related molecule that plays a role in gamma-radiation-induced cell cycle arrest. J Biol Chem. 277, 13873-13882.

Gupta,S., Campbell,D., Derijard,B., and Davis,R.J. (1995). Transcription factor ATF2 regulation by the JNK signal transduction pathway. Science 267, 389-393.

Guyton,K.Z., Liu,Y., Gorospe,M., Xu,Q., and Holbrook,N.J. (1996). Activation of mitogen-activated protein kinase by H2O2. Role in cell survival following oxidant injury. J Biol Chem. 271, 4138-4142.

Hale,K.K., Trollinger,D., Rihanek,M., and Manthey,C.L. Differential expression and activation of p38 mitogen-activated protein kinase alpha, beta, gamma, and delta in inflammatory cell lineages. J Immunol 162, 4246-4252.

Han J, Lee JD, Bibbs L, and Ulevitch RJ. (1994). A MAP kinase targeted by endotoxin and hyperosmolarity in mammalian cells. Science 265, 808-811.

Hanks,S.K. (2003). Genomic analysis of the eukaryotic protein kinase superfamily: a perspective. Genome Biol 4, 111.

Hanks,S.K. and Hunter,T. (1995). Protein kinases 6. The eukaryotic protein kinase superfamily: kinase (catalytic) domain structure and classification. FASEB J 9, 576-596.

Hasegawa,M., Cuenda,A., Spillantini,M.G., Thomas,G.M., Buee-Scherrer,V., Cohen,P., and Goedert,M. (1999). Stress-activated protein kinase-3 interacts with the PDZ domain of alpha1-syntrophin. A mechanism for specific substrate recognition. J Biol Chem. 274, 12626-12631.

139

Page 233: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Hibi,M., Lin,A., Smeal,T., Minden,A., and Karin,M. (1993). Identification of an oncoprotein- and UV-responsive protein kinase that binds and potentiates the c-Jun activation domain. Genes Dev. 7, 2135-2148.

Hines W.A., Thorburn J., and Thorburn A. (1999). Cell density and contraction regulate p38 MAP kinase-dependent responses in neonatal rat cardiac myocytes. Am J Physiol. 277, H331-H341.

Ho,R.C., Alcazar,O., Fujii,N., Hirshman,M.F., and Goodyear,L.J. (2004). p38gamma MAPK regulation of glucose transporter expression and glucose uptake in L6 myotubes and mouse skeletal muscle. Am. J Physiol Regul. Integr. Comp Physiol 286, R342-R349.

Hockenbery,D., Nunez,G., Milliman,C., Schreiber,R.D., and Korsmeyer,S.J. (1990). Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature 348, 334-336.

Holland,P.M. and Cooper,J.A. (1999). Protein modification: docking sites for kinases. Curr. Biol 9, R329-R331.

Hoover,H.E., Thuerauf,D.J., Martindale,J.J., and Glembotski,C.C. (2000). alpha B-crystallin gene induction and phosphorylation by MKK6-activated p38. A potential role for alpha B-crystallin as a target of the p38 branch of the cardiac stress response. J Biol Chem. 275, 23825-23833.

Hope,I.A. and Struhl,K. (1986). Functional dissection of a eukaryotic transcriptional activator protein, GCN4 of yeast. Cell 46, 885-894.

Huang,C., Rajfur,Z., Borchers,C., Schaller,M.D., and Jacobson,K. (2003). JNK phosphorylates paxillin and regulates cell migration. Nature 424, 219-223.

Hung,A.Y. and Sheng,M. (2002). PDZ domains: structural modules for protein complex assembly. J Biol Chem. 277, 5699-5702.

Hunot,S., Vila,M., Teismann,P., Davis,R.J., Hirsch,E.C., Przedborski,S., Rakic,P., and Flavell,R.A. (2004). JNK-mediated induction of cyclooxygenase 2 is required for neurodegeneration in a mouse model of Parkinson's disease. Proc Natl Acad Sci U S A 101, 665-670.

Ingley,E., Chappell,D., Poon,S.Y., Sarna,M.K., Beaumont,J.G., Williams,J.H., Stillitano,J.P., Tsai,S., Leedman,P.J., Tilbrook,P.A., and Klinken,S.P. (2001). Thyroid hormone receptor-interacting protein 1 modulates cytokine and nuclear hormone signaling in erythroid cells. J Biol Chem. 276, 43428-43434.

Ingley,E., Sarna,M.K., Beaumont,J.G., Tilbrook,P.A., Tsai,S., Takemoto,Y., Williams,J.H., and Klinken,S.P. (2000). HS1 interacts with Lyn and is critical for erythropoietin-induced differentiation of erythroid cells. J Biol Chem. 275, 7887-7893.

140

Page 234: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Irie,M., Hata,Y., Deguchi,M., Ide,N., Hirao,K., Yao,I., Nishioka,H., and Takai,Y. (1999). Isolation and characterization of mammalian homologues of Caenorhabditis elegans lin-7: localization at cell-cell junctions. Oncogene 18, 2811-2817.

Ishikawa,Y., Kusaka,E., Enokido,Y., Ikeuchi,T., and Hatanaka,H. (2003). Regulation of Bax translocation through phosphorylation at Ser-70 of Bcl-2 by MAP kinase in NO-induced neuronal apoptosis. Mol Cell Neurosci. 24, 451-459.

Iwai-Kanai,E., Hasegawa,K., Sawamura,T., Fujita,M., Yanazume,T., Toyokuni,S., Adachi,S., Kihara,Y., and Sasayama,S. (2001). Activation of lectin-like oxidized low-density lipoprotein receptor-1 induces apoptosis in cultured neonatal rat cardiac myocytes. Circulation 104, 2948-2954.

Iwakura,A., Fujita,M., Hasegawa,K., Toyokuni,S., Sawamura,T., Nohara,R., Sasayama,S., and Komeda,M. (2001). Pericardial fluid from patients with ischemic heart disease induces myocardial cell apoptotis via an oxidant stress-sensitive p38 mitogen-activated protein kinase pathway. J Mol Cell Cardiol. 33, 419-430.

Izumiya,Y., Kim,S., Izumi,Y., Yoshida,K., Yoshiyama,M., Matsuzawa,A., Ichijo,H., and Iwao,H. (2003). Apoptosis signal-regulating kinase 1 plays a pivotal role in angiotensin II-induced cardiac hypertrophy and remodeling. Circ. Res 93, 874-883.

Jacobs,D., Glossip,D., Xing,H., Muslin,A.J., and Kornfeld,K. (1999). Multiple docking sites on substrate proteins form a modular system that mediates recognition by ERK MAP kinase. Genes Dev. 13, 163-175.

Jaffrey,S.R., Benfenati,F., Snowman,A.M., Czernik,A.J., and Snyder,S.H. (2002). Neuronal nitric-oxide synthase localization mediated by a ternary complex with synapsin and CAPON. Proc Natl Acad Sci U S A 99, 3199-3204.

Jenkins,S.M., Zinnerman,M., Garner,C., and Johnson,G.V. (2000). Modulation of tau phosphorylation and intracellular localization by cellular stress. Biochem J 345 Pt 2, 263-270.

Jiang,Y., Chen,C., Li,Z., Guo,W., Gegner,J.A., Lin,S., and Han,J. (1996). Characterization of the structure and function of a new mitogen-activated protein kinase (p38beta). J Biol Chem. 271, 17920-17926.

Jiang,Y., Gram,H., Zhao,M., New,L., Gu,J., Feng,L., Di Padova,F., Ulevitch,R.J., and Han,J. (1997). Characterization of the structure and function of the fourth member of p38 group mitogen-activated protein kinases, p38delta. J Biol Chem. 272, 30122-30128.

Junqueira,D., Cilenti,L., Musumeci,L., Sedivy,J.M., and Zervos,A.S. (2003). Random mutagenesis of PDZ(Omi) domain and selection of mutants that specifically bind the Myc proto-oncogene and induce apoptosis. Oncogene 22, 2772-2781.

Kaiser,R.A., Bueno,O.F., Lips,D.J., Doevendans,P.A., Jones,F., Kimball,T.F., and Molkentin,J.D. (2004). Targeted inhibition of p38 mitogen-activated protein kinase

141

Page 235: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

antagonizes cardiac injury and cell death following ischemia-reperfusion in vivo. J Biol Chem. 279, 15524-15530.

Kallunki,T., Su,B., Tsigelny,I., Sluss,H.K., Derijard,B., Moore,G., Davis,R., and Karin,M. (1994). JNK2 contains a specificity-determining region responsible for efficient c-Jun binding and phosphorylation. Genes Dev. 8, 2996-3007.

Keegan,L., Gill,G., and Ptashne,M. (1986). Separation of DNA binding from the transcription-activating function of a eukaryotic regulatory protein. Science 231, 699-704.

Keyse,S.M. (2000). Protein phosphatases and the regulation of mitogen-activated protein kinase signalling. Curr. Opin. Cell Biol 12, 186-192.

Khokhlatchev,A.V., Canagarajah,B., Wilsbacher,J., Robinson,M., Atkinson,M., Goldsmith,E., and Cobb,M.H. (1998). Phosphorylation of the MAP kinase ERK2 promotes its homodimerization and nuclear translocation. Cell 93, 605-615.

Kim,J. and Freeman,M.R. (2003). JNK/SAPK mediates doxorubicin-induced differentiation and apoptosis in MCF-7 breast cancer cells. Breast Cancer Res Treat. 79, 321-328.

Kim,S.O., Irwin,P., Katz,S., and Pelech,S.L. (1998). Expression of mitogen-activated protein kinase pathways during postnatal development of rat heart. J Cell Biochem 71, 286-301.

Kimonides,V.G., Spillantini,M.G., Sofroniew,M.V., Fawcett,J.W., and Herbert,J. (1999). Dehydroepiandrosterone antagonizes the neurotoxic effects of corticosterone and translocation of stress-activated protein kinase 3 in hippocampal primary cultures. Neuroscience 89, 429-436.

Klauck,T.M., Faux,M.C., Labudda,K., Langeberg,L.K., Jaken,S., and Scott,J.D. (1996). Coordination of three signaling enzymes by AKAP79, a mammalian scaffold protein. Science 271, 1589-1592.

Klumpp,S. and Krieglstein,J. (2002). Serine/threonine protein phosphatases in apoptosis. Curr. Opin. Pharmacol. 2, 458-462.

Knebel A, Morrice N, and Cohen P. (2001). A novel method to identify protein kinase substrates: eEF2 kinase is phosphorylated and inhibited by SAPK4/p38delta. EMBO J. 20, 4360-4369.

Kolodziejczyk,S.M., Walsh,G.S., Balazsi,K., Seale,P., Sandoz,J., Hierlihy,A.M., Rudnicki,M.A., Chamberlain,J.S., Miller,F.D., and Megeney,L.A. (2001). Activation of JNK1 contributes to dystrophic muscle pathogenesis. Curr. Biol 11, 1278-1282.

Konishi,Y., Lehtinen,M., Donovan,N., and Bonni,A. (2002). Cdc2 phosphorylation of BAD links the cell cycle to the cell death machinery. Mol Cell 9, 1005-1016.

142

Page 236: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Kornau,H.C., Schenker,L.T., Kennedy,M.B., and Seeburg,P.H. (1995). Domain interaction between NMDA receptor subunits and the postsynaptic density protein PSD-95. Science 269, 1737-1740.

Kuan,C.Y., Yang,D.D., Samanta Roy,D.R., Davis,R.J., Rakic,P., and Flavell,R.A. (1999). The Jnk1 and Jnk2 protein kinases are required for regional specific apoptosis during early brain development. Neuron 22, 667-676.

Kudoh,S., Komuro,I., Hiroi,Y., Zou,Y., Harada,K., Sugaya,T., Takekoshi,N., Murakami,K., Kadowaki,T., and Yazaki,Y. (1998). Mechanical stretch induces hypertrophic responses in cardiac myocytes of angiotensin II type 1a receptor knockout mice. J Biol Chem. 273, 24037-24043.

Kumar,S., McDonnell,P.C., Gum,R.J., Hand,A.T., Lee,J.C., and Young,P.R. (1997). Novel homologues of CSBP/p38 MAP kinase: activation, substrate specificity and sensitivity to inhibition by pyridinyl imidazoles. Biochem Biophys Res Commun. 235, 533-538.

Kuo,A., Zhong,C., Lane,W.S., and Derynck,R. (2000). Transmembrane transforming growth factor-alpha tethers to the PDZ domain-containing, Golgi membrane-associated protein p59/GRASP55. EMBO J 19, 6427-6439.

Kuriyan,J. and Cowburn,D. (1997). Modular peptide recognition domains in eukaryotic signaling. Annu. Rev. Biophys Biomol. Struct. 26, 259-288.

Kyriakis,J.M. and Avruch,J. (2001). Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev. 81, 807-869.

Lai,K.M. and Pawson,T. (2000). The ShcA phosphotyrosine docking protein sensitizes cardiovascular signaling in the mouse embryo. Genes Dev. 14, 1132-1145.

Lam,M.H., Michell,B.J., Fodero-Tavoletti,M.T., Kemp,B.E., Tonks,N.K., and Tiganis,T. (2001). Cellular stress regulates the nucleocytoplasmic distribution of the protein-tyrosine phosphatase TCPTP. J Biol Chem. 276, 37700-37707.

Lamb,J.A., Ventura,J.J., Hess,P., Flavell,R.A., and Davis,R.J. (2003). JunD mediates survival signaling by the JNK signal transduction pathway. Mol Cell 11, 1479-1489.

Lavoie,J.N., L'Allemain,G., Brunet,A., Muller,R., and Pouyssegur,J. (1996). Cyclin D1 expression is regulated positively by the p42/p44MAPK and negatively by the p38/HOGMAPK pathway. J Biol Chem. 271, 20608-20616.

Lawler,S., Fleming,Y., Goedert,M., and Cohen,P. (1998). Synergistic activation of SAPK1/JNK1 by two MAP kinase kinases in vitro. Curr. Biol 8, 1387-1390.

Lechner C, Zahalka MA, Giot JF, Moller NP, and Ullrich A. (1996). ERK6, a mitogen-activated protein kinase involved in C2C12 myoblast differentiation. Proc Natl Acad Sci U S A. 93, 4355-4359.

143

Page 237: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Lee,S.H., Park,J., Che,Y., Han,P.L., and Lee,J.K. (2000). Constitutive activity and differential localization of p38alpha and p38beta MAPKs in adult mouse brain. J Neurosci. Res 60, 623-631.

Lefebvre,C., Terret,M.E., Djiane,A., Rassinier,P., Maro,B., and Verlhac,M.H. (2002). Meiotic spindle stability depends on MAPK-interacting and spindle-stabilizing protein (MISS), a new MAPK substrate. J Cell Biol 157, 603-613.

Lei,K. and Davis,R.J. (2003). JNK phosphorylation of Bim-related members of the Bcl2 family induces Bax-dependent apoptosis. Proc Natl Acad Sci U S A 100, 2432-2437.

Lei,K., Nimnual,A., Zong,W.X., Kennedy,N.J., Flavell,R.A., Thompson,C.B., Bar-Sagi,D., and Davis,R.J. (2002). The Bax subfamily of Bcl2-related proteins is essential for apoptotic signal transduction by c-Jun NH(2)-terminal kinase. Mol Cell Biol 22, 4929-4942.

Lemke,L.E., Bloem,L.J., Fouts,R., Esterman,M., Sandusky,G., and Vlahos,C.J. (2001). Decreased p38 MAPK activity in end-stage failing human myocardium: p38 MAPK alpha is the predominant isoform expressed in human heart. J Mol Cell Cardiol. 33, 1527-1540.

Levinthal,D.J. and DeFranco,D.B. (2004). Transient phosphatidylinositol 3-kinase inhibition protects immature primary cortical neurons from oxidative toxicity via suppression of extracellular signal-regulated kinase activation. J Biol Chem. 279, 11206-11213.

Ley,R., Balmanno,K., Hadfield,K., Weston,C., and Cook,S.J. (2003). Activation of the ERK1/2 signaling pathway promotes phosphorylation and proteasome-dependent degradation of the BH3-only protein, Bim. J Biol Chem. 278, 18811-18816.

Li Z, Jiang Y, Ulevitch RJ, and Han J. (1996). The primary structure of p38 gamma: a new member of p38 group of MAP kinases. Biochem Biophys Res Commun. 228, 334-340.

Liang,Q. and Molkentin,J.D. (2003). Redefining the roles of p38 and JNK signaling in cardiac hypertrophy: dichotomy between cultured myocytes and animal models. J Mol Cell Cardiol. 35, 1385-1394.

Liao,P., Georgakopoulos,D., Kovacs,A., Zheng,M., Lerner,D., Pu,H., Saffitz,J., Chien,K., Xiao,R.P., Kass,D.A., and Wang,Y. (2001). The in vivo role of p38 MAP kinases in cardiac remodeling and restrictive cardiomyopathy. Proc Natl Acad Sci U S A 98, 12283-12288.

Liao,P., Wang,S.Q., Wang,S., Zheng,M., Zheng,M., Zhang,S.J., Cheng,H., Wang,Y., and Xiao,R.P. (2002). p38 Mitogen-activated protein kinase mediates a negative inotropic effect in cardiac myocytes. Circ. Res 90, 190-196.

144

Page 238: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Lilly,B., Zhao,B., Ranganayakulu,G., Paterson,B.M., Schulz,R.A., and Olson,E.N. (1995). Requirement of MADS domain transcription factor D-MEF2 for muscle formation in Drosophila. Science 267, 688-693.

Lim,R., Winteringham,L.N., Williams,J.H., McCulloch,R.K., Ingley,E., Tiao,J.Y., Lalonde,J.P., Tsai,S., Tilbrook,P.A., Sun,Y., Wu,X., Morris,S.W., and Klinken,S.P. (2002). MADM, a novel adaptor protein that mediates phosphorylation of the 14-3-3 binding site of myeloid leukemia factor 1. J Biol Chem. 277, 40997-41008.

Lin,A., Minden,A., Martinetto,H., Claret,F.X., Lange-Carter,C., Mercurio,F., Johnson,G.L., and Karin,M. (1995). Identification of a dual specificity kinase that activates the Jun kinases and p38-Mpk2. Science 268, 286-290.

Lips,D.J., Bueno,O.F., Wilkins,B.J., Purcell,N.H., Kaiser,R.A., Lorenz,J.N., Voisin,L., Saba-El-Leil,M.K., Meloche,S., Pouyssegur,J., Pages,G., De Windt,L.J., Doevendans,P.A., and Molkentin,J.D. (2004). MEK1-ERK2 Signaling Pathway Protects Myocardium From Ischemic Injury In Vivo. Circulation.

Liu,L.X., Liu,Z.H., Jiang,H.C., Zhang,W.H., Qi,S.Y., Hu,J., Wang,X.Q., and Wu,M. (2003). Gene expression profiles of hepatoma cell line HLE. World J Gastroenterol. 9, 683-687.

Liu,T.F., Kandala,G., and Setaluri,V. (2001). PDZ domain protein GIPC interacts with the cytoplasmic tail of melanosomal membrane protein gp75 (tyrosinase-related protein-1). J Biol Chem. 276, 35768-35777.

Livingstone,C., Patel,G., and Jones,N. (1995). ATF-2 contains a phosphorylation-dependent transcriptional activation domain. EMBO J 14, 1785-1797.

Losa,J.H., Cobo,C.P., Viniegra,J.G., Sanchez-Arevalo,L., V, Cajal,S., and Sanchez-Prieto,R. (2003). Role of the p38 MAPK pathway in cisplatin-based therapy. Oncogene 22, 3998-4006.

MacDonald,T.J., Brown,K.M., LaFleur,B., Peterson,K., Lawlor,C., Chen,Y., Packer,R.J., Cogen,P., and Stephan,D.A. (2001). Expression profiling of medulloblastoma: PDGFRA and the RAS/MAPK pathway as therapeutic targets for metastatic disease. Nat. Genet. 29, 143-152.

Mackay,K. and Mochly-Rosen,D. (1999). An inhibitor of p38 mitogen-activated protein kinase protects neonatal cardiac myocytes from ischemia. J Biol Chem. 274, 6272-6279.

Maltese,W.A., Sheridan,K.M., Repko,E.M., and Erdman,R.A. (1990). Post-translational modification of low molecular mass GTP-binding proteins by isoprenoid. J Biol Chem. 265, 2148-2155.

Manning,G., Plowman,G.D., Hunter,T., and Sudarsanam,S. (2002a). Evolution of protein kinase signaling from yeast to man. Trends Biochem Sci 27, 514-520.

145

Page 239: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Manning,G., Whyte,D.B., Martinez,R., Hunter,T., and Sudarsanam,S. (2002b). The protein kinase complement of the human genome. Science 298, 1912-1934.

Mansour,S.J., Matten,W.T., Hermann,A.S., Candia,J.M., Rong,S., Fukasawa,K., Vande Woude,G.F., and Ahn,N.G. (1994). Transformation of mammalian cells by constitutively active MAP kinase kinase. Science 265 , 966-970.

Margolis,B. and Skolnik,E.Y. (1994). Activation of Ras by receptor tyrosine kinases. J Am. Soc. Nephrol. 5, 1288-1299.

Marinissen MJ, Chiariello M, and Gutkind JS. (2001). Regulation of gene expression by the small GTPase Rho through the ERK6 (p38 gamma) MAP kinase pathway. Genes Dev. 15, 535-553.

Marinissen,M.J., Chiariello,M., Pallante,M., and Gutkind,J.S. (1999). A network of mitogen-activated protein kinases links G protein-coupled receptors to the c-jun promoter: a role for c-Jun NH2-terminal kinase, p38s, and extracellular signal-regulated kinase 5. Mol Cell Biol 19, 4289-4301.

Marshall,C. (2000). Signal transduction takes centre stage. Nat. Cell Biol 2, E185-E187.

Martin,J.L., Avkiran,M., Quinlan,R.A., Cohen,P., and Marber,M.S. (2001). Antiischemic effects of SB203580 are mediated through the inhibition of p38alpha mitogen-activated protein kinase: Evidence from ectopic expression of an inhibition-resistant kinase. Circ. Res 89, 750-752.

Matsushita,M., Yamadori,T., Kato,S., Takemoto,Y., Inazawa,J., Baba,Y., Hashimoto,S., Sekine,S., Arai,S., Kunikata,T., Kurimoto,M., Kishimoto,T., and Tsukada,S. (1998). Identification and characterization of a novel SH3-domain binding protein, Sab, which preferentially associates with Bruton's tyrosine kinase (BtK). Biochem Biophys Res Commun. 245, 337-343.

Mertens S, Craxton M, and Goedert M. (1996). SAP kinase-3, a new member of the family of mammalian stress-activated protein kinases. FEBS Lett. 383, 273-276.

Michel,J.J. and Scott,J.D. (2002). AKAP mediated signal transduction. Annu. Rev. Pharmacol. Toxicol. 42, 235-257.

Minden,A., Lin,A., Smeal,T., Derijard,B., Cobb,M., Davis,R., and Karin,M. (1994). c-Jun N-terminal phosphorylation correlates with activation of the JNK subgroup but not the ERK subgroup of mitogen-activated protein kinases. Mol Cell Biol 14, 6683-6688.

Moriguchi,T., Kawasaki,H., Matsuda,S., Gotoh,Y., and Nishida,E. (1995). Evidence for multiple activators for stress-activated protein kinase/c-Jun amino-terminal kinases. Existence of novel activators. J Biol Chem. 270, 12969-12972.

Morrison,D.K. and Davis,R.J. (2003). Regulation of MAP kinase signaling modules by scaffold proteins in mammals. Annu. Rev. Cell Dev. Biol 19, 91-118.

146

Page 240: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Murray,B., Alessandrini,A., Cole,A.J., Yee,A.G., and Furshpan,E.J. (1998). Inhibition of the p44/42 MAP kinase pathway protects hippocampal neurons in a cell-culture model of seizure activity. Proc Natl Acad Sci U S A 95, 11975-11980.

Nagata,Y., Takahashi,N., Davis,R.J., and Todokoro,K. (1998). Activation of p38 MAP kinase and JNK but not ERK is required for erythropoietin-induced erythroid differentiation. Blood 92, 1859-1869.

Nakamura,A., Harrod,G.V., and Davies,K.E. (2001). Activation of calcineurin and stress activated protein kinase/p38-mitogen activated protein kinase in hearts of utrophin-dystrophin knockout mice. Neuromuscul. Disord. 11, 251-259.

Nakano,A., Baines,C.P., Kim,S.O., Pelech,S.L., Downey,J.M., Cohen,M.V., and Critz,S.D. (2000). Ischemic preconditioning activates MAPKAPK2 in the isolated rabbit heart: evidence for involvement of p38 MAPK. Circ. Res 86, 144-151.

National Heart Foundation of Australia. (2004). Statistics. Canberra, Australia. www.heartfoundation.com.au.

Neidhart,S., Antonsson,B., Gillieron,C., Vilbois,F., Grenningloh,G., and Arkinstall,S. (2001). c-Jun N-terminal kinase-3 (JNK3)/stress-activated protein kinase-beta (SAPKbeta) binds and phosphorylates the neuronal microtubule regulator SCG10. FEBS Lett 508, 259-264.

Nemoto,Y. and De Camilli,P. (1999). Recruitment of an alternatively spliced form of synaptojanin 2 to mitochondria by the interaction with the PDZ domain of a mitochondrial outer membrane protein. EMBO J 18, 2991-3006.

Ng,D.C., Court,N.W., dos Remedios,C.G., and Bogoyevitch,M.A. (2003). Activation of signal transducer and activator of transcription (STAT) pathways in failing human hearts. Cardiovasc. Res 57, 333-346.

Nguyen,T.T., Scimeca,J.C., Filloux,C., Peraldi,P., Carpentier,J.L., and Van Obberghen,E. (1993). Co-regulation of the mitogen-activated protein kinase, extracellular signal-regulated kinase 1, and the 90-kDa ribosomal S6 kinase in PC12 cells. Distinct effects of the neurotrophic factor, nerve growth factor, and the mitogenic factor, epidermal growth factor. J Biol Chem. 268, 9803-9810.

Niethammer,M., Kim,E., and Sheng,M. (1996). Interaction between the C terminus of NMDA receptor subunits and multiple members of the PSD-95 family of membrane-associated guanylate kinases. J Neurosci. 16, 2157-2163.

Nishigaki,K., Thompson,D., Yugawa,T., Rulli,K., Hanson,C., Cmarik,J., Gutkind,J.S., Teramoto,H., and Ruscetti,S. (2003). Identification and characterization of a novel Ste20/germinal center kinase-related kinase, polyploidy-associated protein kinase. J Biol Chem. 278, 13520-13530.

147

Page 241: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

O'Connor,L., Strasser,A., O'Reilly,L.A., Hausmann,G., Adams,J.M., Cory,S., and Huang,D.C. (1998). Bim: a novel member of the Bcl-2 family that promotes apoptosis. EMBO J 17, 384-395.

Obata,T., Yaffe,M.B., Leparc,G.G., Piro,E.T., Maegawa,H., Kashiwagi,A., Kikkawa,R., and Cantley,L.C. (2000). Peptide and protein library screening defines optimal substrate motifs for AKT/PKB. J Biol Chem. 275, 36108-36115.

Okuda,K., Sanghera,J.S., Pelech,S.L., Kanakura,Y., Hallek,M., Griffin,J.D., and Druker,B.J. (1992). Granulocyte-macrophage colony-stimulating factor, interleukin-3, and steel factor induce rapid tyrosine phosphorylation of p42 and p44 MAP kinase. Blood 79, 2880-2887.

Ossareh-Nazari,B., Bachelerie,F., and Dargemont,C. (1997). Evidence for a role of CRM1 in signal-mediated nuclear protein export. Science 278, 141-144.

Pawson,T. and Nash,P. (2003). Assembly of cell regulatory systems through protein interaction domains. Science 300, 445-452.

Pawson,T. and Scott,J.D. (1997). Signaling through scaffold, anchoring, and adaptor proteins. Science 278, 2075-2080.

Payne,D.M., Rossomando,A.J., Martino,P., Erickson,A.K., Her,J.H., Shabanowitz,J., Hunt,D.F., Weber,M.J., and Sturgill,T.W. (1991). Identification of the regulatory phosphorylation sites in pp42/mitogen-activated protein kinase (MAP kinase). EMBO J 10, 885-892.

Pearson,R.B. and Kemp,B.E. (1991). Protein kinase phosphorylation site sequences and consensus specificity motifs: tabulations. Methods Enzymol. 200, 62-81.

Pellieux,C., Sauthier,T., Aubert,J.F., Brunner,H.R., and Pedrazzini,T. (2000). Angiotensin II-induced cardiac hypertrophy is associated with different mitogen-activated protein kinase activation in normotensive and hypertensive mice. J Hypertens. 18, 1307-1317.

Perdiguero,E., Pillaire,M.J., Bodart,J.F., Hennersdorf,F., Frodin,M., Duesbery,N.S., Alonso,G., and Nebreda,A.R. (2003). Xp38gamma/SAPK3 promotes meiotic G(2)/M transition in Xenopus oocytes and activates Cdc25C. EMBO J 22, 5746-5756.

Petrich,B.G., Eloff,B.C., Lerner,D.L., Kovacs,A., Saffitz,J.E., Rosenbaum,D.S., and Wang,Y. (2004). Targeted activation of c-Jun N-terminal kinase in vivo induces restrictive cardiomyopathy and conduction defects. J Biol Chem. 279, 15330-15338.

Petrich,B.G., Liao,P., and Wang,Y. (2002). Using a gene-switch transgenic approach to dissect distinct roles of MAP kinases in heart failure. Cold Spring Harb. Symp. Quant. Biol 67, 429-437.

148

Page 242: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Pillaire MJ, Nebreda AR, and Darbon JM. (2000). Cisplatin and UV radiation induce activation of the stress-activated protein kinase p38gamma in human melanoma cells. Biochem Biophys Res Commun. 278, 724-728.

Pulverer,B.J., Hughes,K., Franklin,C.C., Kraft,A.S., Leevers,S.J., and Woodgett,J.R. (1993). Co-purification of mitogen-activated protein kinases with phorbol ester-induced c-Jun kinase activity in U937 leukaemic cells. Oncogene 8, 407-415.

Qui,M.S. and Green,S.H. (1992). PC12 cell neuronal differentiation is associated with prolonged p21ras activity and consequent prolonged ERK activity. Neuron 9, 705-717.

Raingeaud,J., Gupta,S., Rogers,J.S., Dickens,M., Han,J., Ulevitch,R.J., and Davis,R.J. (1995). Pro-inflammatory cytokines and environmental stress cause p38 mitogen-activated protein kinase activation by dual phosphorylation on tyrosine and threonine. J Biol Chem. 270, 7420-7426.

Ramirez,M.T., Sah,V.P., Zhao,X.L., Hunter,J.J., Chien,K.R., and Brown,J.H. (1997). The MEKK-JNK pathway is stimulated by alpha1-adrenergic receptor and ras activation and is associated with in vitro and in vivo cardiac hypertrophy. J Biol Chem. 272, 14057-14061.

Regan,C.P., Li,W., Boucher,D.M., Spatz,S., Su,M.S., and Kuida,K. (2002). Erk5 null mice display multiple extraembryonic vascular and embryonic cardiovascular defects. Proc Natl Acad Sci U S A 99, 9248-9253.

Reiss,Y., Goldstein,J.L., Seabra,M.C., Casey,P.J., and Brown,M.S. (1990). Inhibition of purified p21ras farnesyl:protein transferase by Cys-AAX tetrapeptides. Cell 62, 81-88.

Remondino,A., Kwon,S.H., Communal,C., Pimentel,D.R., Sawyer,D.B., Singh,K., and Colucci,W.S. (2003). Beta-adrenergic receptor-stimulated apoptosis in cardiac myocytes is mediated by reactive oxygen species/c-Jun NH2-terminal kinase-dependent activation of the mitochondrial pathway. Circ. Res 92, 136-138.

Robbins,D.J., Zhen,E., Owaki,H., Vanderbilt,C.A., Ebert,D., Geppert,T.D., and Cobb,M.H. (1993). Regulation and properties of extracellular signal-regulated protein kinases 1 and 2 in vitro. J Biol Chem. 268, 5097-5106.

Ross,R.S., Navankasattusas,S., Harvey,R.P., and Chien,K.R. (1996). An HF-1a/HF-1b/MEF-2 combinatorial element confers cardiac ventricular specificity and established an anterior-posterior gradient of expression. Development 122, 1799-1809.

Rozakis-Adcock,M., Fernley,R., Wade,J., Pawson,T., and Bowtell,D. (1993). The SH2 and SH3 domains of mammalian Grb2 couple the EGF receptor to the Ras activator mSos1. Nature 363, 83-85.

Rugh, R. (1968). The Mouse: Its Reproduction and Development. (Minneapolis: Burgess Publishing Company).

149

Page 243: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Sabio,G., Reuver,S., Feijoo,C., Hasegawa,M., Thomas,G.M., Centeno,F., Kuhlendahl,S., Leal-Ortiz,S., Goedert,M., Garner,C., and Cuenda,A. (2004). Stress- and mitogen-induced phosphorylation of the synapse-associated protein SAP90/PSD95 by activation of SAPK3/p38gamma and ERK1/ERK2. Biochem J 380 (Pt1), 19-30.

Sadoshima,J., Montagne,O., Wang,Q., Yang,G., Warden,J., Liu,J., Takagi,G., Karoor,V., Hong,C., Johnson,G.L., Vatner,D.E., and Vatner,S.F. (2002). The MEKK1-JNK pathway plays a protective role in pressure overload but does not mediate cardiac hypertrophy. J Clin. Invest 110, 271-279.

Sakabe,K., Teramoto,H., Zohar,M., Behbahani,B., Miyazaki,H., Chikumi,H., and Gutkind,J.S. (2002). Potent transforming activity of the small GTP-binding protein Rit in NIH 3T3 cells: evidence for a role of a p38gamma-dependent signaling pathway. FEBS Lett 511, 15-20.

Sanger,F. (1981). Determination of nucleotide sequences in DNA. Science 214, 1205-1210.

Schaeffer,H.J., Catling,A.D., Eblen,S.T., Collier,L.S., Krauss,A., and Weber,M.J. (1998). MP1: a MEK binding partner that enhances enzymatic activation of the MAP kinase cascade. Science 281, 1668-1671.

Schaeffer,H.J. and Weber,M.J. (1999). Mitogen-activated protein kinases: specific messages from ubiquitous messengers. Mol Cell Biol 19, 2435-2444.

Schroeter,H., Boyd,C.S., Ahmed,R., Spencer,J.P., Duncan,R.F., Rice-Evans,C., and Cadenas,E. (2003). c-Jun N-terminal kinase (JNK)-mediated modulation of brain mitochondria function: new target proteins for JNK signalling in mitochondrion-dependent apoptosis. Biochem J 372, 359-369.

Seger,R. and Krebs,E.G. (1995). The MAPK signaling cascade. FASEB J 9, 726-735.

Seta K and Sadoshima J. (2002). What is the unique function of SAPK3/p38gamma in cardiac myocytes? J Mol Cell Cardiol. 34, 597-600.

Sharrocks,A.D., Yang,S.H., and Galanis,A. (2000). Docking domains and substrate-specificity determination for MAP kinases. Trends Biochem Sci 25, 448-453.

Sheng,M. (1996). PDZs and receptor/channel clustering: rounding up the latest suspects. Neuron 17, 575-578.

Shih,T.Y., Hattori,S., Clanton,D.J., Ulsh,L.S., Chen,Z.Q., Lautenberger,J.A., and Papas,T.S. (1986). Structure and function of p21 ras proteins. Gene Amplif. Anal. 4, 53-72.

Singh,U.S., Pan,J., Kao,Y.L., Joshi,S., Young,K.L., and Baker,K.M. (2003). Tissue transglutaminase mediates activation of RhoA and MAP kinase pathways during retinoic acid-induced neuronal differentiation of SH-SY5Y cells. J Biol Chem. 278, 391-399.

150

Page 244: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Skelton,N.J., Koehler,M.F., Zobel,K., Wong,W.L., Yeh,S., Pisabarro,M.T., Yin,J.P., Lasky,L.A., and Sidhu,S.S. (2003). Origins of PDZ domain ligand specificity. Structure determination and mutagenesis of the Erbin PDZ domain. J Biol Chem. 278, 7645-7654.

Smeal,T., Binetruy,B., Mercola,D.A., Birrer,M., and Karin,M. (1991). Oncogenic and transcriptional cooperation with Ha-Ras requires phosphorylation of c-Jun on serines 63 and 73. Nature 354, 494-496.

Smith,J.A., Poteet-Smith,C.E., Malarkey,K., and Sturgill,T.W. (1999). Identification of an extracellular signal-regulated kinase (ERK) docking site in ribosomal S6 kinase, a sequence critical for activation by ERK in vivo. J Biol Chem. 274, 2893-2898.

Songyang,Z., Blechner,S., Hoagland,N., Hoekstra,M.F., Piwnica-Worms,H., and Cantley,L.C. (1994). Use of an oriented peptide library to determine the optimal substrates of protein kinases. Curr. Biol 4, 973-982.

Songyang,Z., Fanning,A.S., Fu,C., Xu,J., Marfatia,S.M., Chishti,A.H., Crompton,A., Chan,A.C., Anderson,J.M., and Cantley,L.C. (1997). Recognition of unique carboxyl-terminal motifs by distinct PDZ domains. Science 275, 73-77.

Spence,H.J., Dhillon,A.S., James,M., and Winder,S.J. (2004). Dystroglycan, a scaffold for the ERK-MAP kinase cascade. EMBO Rep. 5, 484-489.

Srivastava,S., Osten,P., Vilim,F.S., Khatri,L., Inman,G., States,B., Daly,C., DeSouza,S., Abagyan,R., Valtschanoff,J.G., Weinberg,R.J., and Ziff,E.B. (1998). Novel anchorage of GluR2/3 to the postsynaptic density by the AMPA receptor-binding protein ABP. Neuron 21, 581-591.

Stanciu,M. and DeFranco,D.B. (2002). Prolonged nuclear retention of activated extracellular signal-regulated protein kinase promotes cell death generated by oxidative toxicity or proteasome inhibition in a neuronal cell line. J Biol Chem. 277, 4010-4017.

Stanciu,M., Wang,Y., Kentor,R., Burke,N., Watkins,S., Kress,G., Reynolds,I., Klann,E., Angiolieri,M.R., Johnson,J.W., and DeFranco,D.B. (2000). Persistent activation of ERK contributes to glutamate-induced oxidative toxicity in a neuronal cell line and primary cortical neuron cultures. J Biol Chem. 275, 12200-12206.

Steeg,P.S., Palmieri,D., Ouatas,T., and Salerno,M. (2003). Histidine kinases and histidine phosphorylated proteins in mammalian cell biology, signal transduction and cancer. Cancer Lett 190, 1-12.

Stein,B., Yang,M.X., Young,D.B., Janknecht,R., Hunter,T., Murray,B.W., and Barbosa,M.S. (1997). p38-2, a novel mitogen-activated protein kinase with distinct properties. J Biol Chem. 272, 19509-19517.

Stricker,N.L. and Huganir,R.L. (2003). The PDZ domains of mLin-10 regulate its trans-Golgi network targeting and the surface expression of AMPA receptors. Neuropharmacology 45, 837-848.

151

Page 245: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Suzuki,Y., Imai,Y., Nakayama,H., Takahashi,K., Takio,K., and Takahashi,R. (2001). A serine protease, HtrA2, is released from the mitochondria and interacts with XIAP, inducing cell death. Mol Cell 8, 613-621.

Takahashi,M., Gotoh,Y., Isagawa,T., Nishimura,T., Goyama,E., Kim,H.S., Mukai,H., and Ono,Y. (2003). Regulation of a mitogen-activated protein kinase kinase kinase, MLTK by PKN. J Biochem (Tokyo) 133, 181-187.

Takano,H., Komuro,I., Oka,T., Shiojima,I., Hiroi,Y., Mizuno,T., and Yazaki,Y. (1998). The Rho family G proteins play a critical role in muscle differentiation. Mol Cell Biol 18, 1580-1589.

Takayama,S., Sato,T., Krajewski,S., Kochel,K., Irie,S., Millan,J.A., and Reed,J.C. (1995). Cloning and functional analysis of BAG-1: a novel Bcl-2-binding protein with anti-cell death activity. Cell 80, 279-284.

Tanoue,T., Adachi,M., Moriguchi,T., and Nishida,E. (2000). A conserved docking motif in MAP kinases common to substrates, activators and regulators. Nat. Cell Biol 2, 110-116.

Taylor SW, Fahy E, Zhang B, Glenn GM, Warnock DE, Wiley S, Murphy AN, Gaucher SP, Capaldi RA, Gibson BW, and Ghosh SS. (2003). Characterization of the human heart mitochondrial proteome. Nat. Biotechnol. 21, 281-286.

Terret,M.E., Lefebvre,C., Djiane,A., Rassinier,P., Moreau,J., Maro,B., and Verlhac,M.H. (2003). DOC1R: a MAP kinase substrate that control microtubule organization of metaphase II mouse oocytes. Development 130, 5169-5177.

Theodosiou,A. and Ashworth,A. (2002). MAP kinase phosphatases. Genome Biol 3, REVIEWS3009.

Tibbles,L.A. and Woodgett,J.R. (1999). The stress-activated protein kinase pathways. Cell Mol Life Sci 55, 1230-1254.

Tortorella,L.L., Lin,C.B., and Pilch,P.F. (2003). ERK6 is expressed in a developmentally regulated manner in rodent skeletal muscle. Biochem Biophys Res Commun. 306, 163-168.

Tsai,S., Bartelmez,S., Sitnicka,E., and Collins,S. (1994). Lymphohematopoietic progenitors immortalized by a retroviral vector harboring a dominant-negative retinoic acid receptor can recapitulate lymphoid, myeloid, and erythroid development. Genes Dev. 8, 2831-2841.

Tsygankov,A.Y. (2003). Non-receptor protein tyrosine kinases. Front Biosci. 8, s595-s635.

Turner,C.E., Glenney,J.R., Jr., and Burridge,K. (1990). Paxillin: a new vinculin-binding protein present in focal adhesions. J Cell Biol 111, 1059-1068.

152

Page 246: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Uddin,S., Ah-Kang,J., Ulaszek,J., Mahmud,D., and Wickrema,A. (2004). Differentiation stage-specific activation of p38 mitogen-activated protein kinase isoforms in primary human erythroid cells. Proc Natl Acad Sci U S A 101, 147-152.

Ueyama,T., Kawashima,S., Sakoda,T., Rikitake,Y., Ishida,T., Kawai,M., Yamashita,T., Ishido,S., Hotta,H., and Yokoyama,M. (2000). Requirement of activation of the extracellular signal-regulated kinase cascade in myocardial cell hypertrophy. J Mol Cell Cardiol. 32, 947-960.

van Bilsen,M. and Chien,K.R. (1993). Growth and hypertrophy of the heart: towards an understanding of cardiac specific and inducible gene expression. Cardiovasc. Res 27, 1140-1149.

Vlahos,C.J., McDowell,S.A., and Clerk,A. (2003). Kinases as therapeutic targets for heart failure. Nat. Rev. Drug Discov. 2, 99-113.

Vojtek,A.B., Hollenberg,S.M., and Cooper,J.A. (1993). Mammalian Ras interacts directly with the serine/threonine kinase Raf. Cell 74, 205-214.

Vomastek,T., Schaeffer,H.J., Tarcsafalvi,A., Smolkin,M.E., Bissonette,E.A., and Weber,M.J. (2004). Modular construction of a signaling scaffold: MORG1 interacts with components of the ERK cascade and links ERK signaling to specific agonists. Proc Natl Acad Sci U S A 101, 6981-6986.

Walker,A.R., Walker,B.F., and Segal,I. (2004). Some puzzling situations in the onset, occurrence and future of coronary heart disease in developed and developing populations, particularly such in sub-Saharan Africa. J R. Soc. Health 124, 40-46.

Wang,W.L., Yeh,S.F., Chang,Y.I., Hsiao,S.F., Lian,W.N., Lin,C.H., Huang,C.Y., and Lin,W.J. (2003a). PICK1, an anchoring protein that specifically targets protein kinase Calpha to mitochondria selectively upon serum stimulation in NIH 3T3 cells. J Biol Chem. 278, 37705-37712.

Wang,W.L., Yeh,S.F., Chang,Y.I., Hsiao,S.F., Lian,W.N., Lin,C.H., Huang,C.Y., and Lin,W.J. (2003b). PICK1, an anchoring protein that specifically targets protein kinase Calpha to mitochondria selectively upon serum stimulation in NIH 3T3 cells. J Biol Chem. 278, 37705-37712.

Wang,X., McGowan,C.H., Zhao,M., He,L., Downey,J.S., Fearns,C., Wang,Y., Huang,S., and Han,J. (2000). Involvement of the MKK6-p38gamma cascade in gamma-radiation-induced cell cycle arrest. Mol Cell Biol 20 , 4543-4552.

Wang,X. and Proud,C.G. (1997). p70 S6 kinase is activated by sodium arsenite in adult rat cardiomyocytes: roles for phosphatidylinositol 3-kinase and p38 MAP kinase. Biochem Biophys Res Commun. 238, 207-212.

153

Page 247: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Wang,Y., Huang,S., Sah,V.P., Ross,J., Jr., Brown,J.H., Han,J., and Chien,K.R. (1998a). Cardiac muscle cell hypertrophy and apoptosis induced by distinct members of the p38 mitogen-activated protein kinase family. J Biol Chem. 273, 2161-2168.

Wang,Y., Su,B., Sah,V.P., Brown,J.H., Han,J., and Chien,K.R. (1998b). Cardiac hypertrophy induced by mitogen-activated protein kinase kinase 7, a specific activator for c-Jun NH2-terminal kinase in ventricular muscle cells. J Biol Chem. 273, 5423-5426.

Waskiewicz,A.J., Flynn,A., Proud,C.G., and Cooper,J.A. (1997). Mitogen-activated protein kinases activate the serine/threonine kinases Mnk1 and Mnk2. EMBO J 16, 1909-1920.

Westwick,J.K., Cox,A.D., Der,C.J., Cobb,M.H., Hibi,M., Karin,M., and Brenner,D.A. (1994). Oncogenic Ras activates c-Jun via a separate pathway from the activation of extracellular signal-regulated kinases. Proc Natl Acad Sci U S A 91, 6030-6034.

White,J.G., Amos,W.B., and Fordham,M. (1987). An evaluation of confocal versus conventional imaging of biological structures by fluorescence light microscopy. J Cell Biol 105, 41-48.

Whitmarsh,A.J., Cavanagh,J., Tournier,C., Yasuda,J., and Davis,R.J. (1998). A mammalian scaffold complex that selectively mediates MAP kinase activation. Science 281, 1671-1674.

Widmann,C., Gibson,S., Jarpe,M.B., and Johnson,G.L. (1999). Mitogen-activated protein kinase: conservation of a three-kinase module from yeast to human. Physiol Rev. 79, 143-180.

Wiltshire,C., Matsushita,M., Tsukada,S., Gillespie,D.A., and May,G.H. (2002). A new c-Jun N-terminal kinase (JNK)-interacting protein, Sab (SH3BP5), associates with mitochondria. Biochem J 367, 577-585.

Wolff,B., Sanglier,J.J., and Wang,Y. (1997). Leptomycin B is an inhibitor of nuclear export: inhibition of nucleo-cytoplasmic translocation of the human immunodeficiency virus type 1 (HIV-1) Rev protein and Rev-dependent mRNA. Chem. Biol 4, 139-147.

Woods,D.F. and Bryant,P.J. (1989). Molecular cloning of the lethal(1)discs large-1 oncogene of Drosophila. Dev. Biol 134, 222-235.

Wu,Y., Chen,Z., and Ullrich,A. (2003). EGFR and FGFR signaling through FRS2 is subject to negative feedback control by ERK1/2. Biol Chem. 384, 1215-1226.

Yaakov,G., Bell,M., Hohmann,S., and Engelberg,D. (2003). Combination of two activating mutations in one HOG1 gene forms hyperactive enzymes that induce growth arrest. Mol Cell Biol 23, 4826-4840.

Yamadori,T., Baba,Y., Matsushita,M., Hashimoto,S., Kurosaki,M., Kurosaki,T., Kishimoto,T., and Tsukada,S. (1999). Bruton's tyrosine kinase activity is negatively

154

Page 248: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

regulated by Sab, the Btk-SH3 domain-binding protein. Proc Natl Acad Sci U S A 96, 6341-6346.

Yamamoto,Y., Mandai,K., Okabe,N., Hoshino,T., Nakanishi,H., and Takai,Y. (2002). Localization of mLin-7 at nectin-based cell-cell junctions. Oncogene 21, 2545-2554.

Yang,E., Zha,J., Jockel,J., Boise,L.H., Thompson,C.B., and Korsmeyer,S.J. (1995). Bad, a heterodimeric partner for Bcl-XL and Bcl-2, displaces Bax and promotes cell death. Cell 80, 285-291.

Yang,J., Drazba,J.A., Ferguson,D.G., and Bond,M. (1998a). A-kinase anchoring protein 100 (AKAP100) is localized in multiple subcellular compartments in the adult rat heart. J Cell Biol 142, 511-522.

Yang,S.H., Galanis,A., and Sharrocks,A.D. (1999). Targeting of p38 mitogen-activated protein kinases to MEF2 transcription factors. Mol Cell Biol 19, 4028-4038.

Yang,S.H., Yates,P.R., Whitmarsh,A.J., Davis,R.J., and Sharrocks,A.D. (1998b). The Elk-1 ETS-domain transcription factor contains a mitogen-activated protein kinase targeting motif. Mol Cell Biol 18, 710-720.

Yao,Z., Diener,K., Wang,X.S., Zukowski,M., Matsumoto,G., Zhou,G., Mo,R., Sasaki,T., Nishina,H., Hui,C.C., Tan,T.H., Woodgett,J.P., and Penninger,J.M. (1997). Activation of stress-activated protein kinases/c-Jun N-terminal protein kinases (SAPKs/JNKs) by a novel mitogen-activated protein kinase kinase. J Biol Chem. 272, 32378-32383.

Yu,C., Minemoto,Y., Zhang,J., Liu,J., Tang,F., Bui,T.N., Xiang,J., and Lin,A. (2004). JNK suppresses apoptosis via phosphorylation of the proapoptotic Bcl-2 family protein BAD. Mol Cell 13, 329-340.

Yue,T.L., Gu,J.L., Wang,C., Reith,A.D., Lee,J.C., Mirabile,R.C., Kreutz,R., Wang,Y., Maleeff,B., Parsons,A.A., and Ohlstein,E.H. (2000). Extracellular signal-regulated kinase plays an essential role in hypertrophic agonists, endothelin-1 and phenylephrine-induced cardiomyocyte hypertrophy. J Biol Chem. 275, 37895-37901.

Yusuf,S. (1998). The global problem of cardiovascular disease. Int. J Clin. Pract. Suppl 94, 3-6.

Zechner,D., Thuerauf,D.J., Hanford,D.S., McDonough,P.M., and Glembotski,C.C. (1997). A role for the p38 mitogen-activated protein kinase pathway in myocardial cell growth, sarcomeric organization, and cardiac-specific gene expression. J Cell Biol 139, 115-127.

Zha,J., Harada,H., Yang,E., Jockel,J., and Korsmeyer,S.J. (1996). Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L). Cell 87, 619-628.

155

Page 249: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Zhu,J.H., Guo,F., Shelburne,J., Watkins,S., and Chu,C.T. (2003). Localization of phosphorylated ERK/MAP kinases to mitochondria and autophagosomes in Lewy body diseases. Brain Pathol. 13, 473-481.

156

Page 250: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Appendix I

Page 251: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Primer Name Sequence (5’ to 3’) Annealing Temp Used

Purpose

Lambda gt11 forward sequencing primer

GGT GGC GAC GAC TCC TGG AGC C 50°C Sequencing of library clones from SAPK3 antibody library screening

Lambda gt11 reverse sequencing primer

GAC ACC AGA CCA ACT GGT AAT G 50°C Sequencing of library clones from SAPK3 antibody library screening

SAPK3 Begin GCG CTT GGC AAA CAG CTC CGA 50°C Confirmation of SAPK3 insertion into the pBTM116 yeast vector by sequencing

SAPK3 End GCC CAC CCA TAC TTT GAG TCC 50°C Confirmation of SAPK3 insertion into the pBTM116 yeast vector by sequencing

SAPK3 SDM Forward GCC AGA GTT CCA TAG GAG ACA GCT CTG TGA AG

55°C Inserting a STOP codon into the SAPK3 insert to truncate the protein at amino acid 363 in the pBTM116 vector

pBTM116 Forward CAG CAG AGC TTC ACC ATT G 50°C Confirmation of SAPK3 site-directed mutagenesis in pBTM116 by sequencing

SAPK3 SDM Reverse CTT CAC AGA GCT GTC TCC TAT GGA ACT CTG GC

55°C Inserting a STOP codon into the SAPK3 insert to truncate the protein at amino acid 363 in the pBTM116 vector

Appendix I: List of Primers Used During the Course of this PhD Project

Page 252: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Primer Name Sequence (5’ to 3’) Annealing Temp Used

Purpose

pBTM116 Reverse ATC ATA AGA AAT TCG CCC G 50°C Confirmation of SAPK3 site-directed mutagenesis in pBTM116 by sequencing

M13-F Primer GTA AAA CGA CGG CCA GT 50°C Sequencing of pVP16 library clones from Yeast 2-hybrid screening

VP16-2 Primer GAG TTT GAG CAG ATG TTT ACC G 50°C Sequencing of pVP16 library clones from Yeast 2-hybrid screening

SAPK3 Forward GAA GAT CAC AGG GAC GCC CCC TCC

60°C RT-PCR of SAPK3 from mouse embryonic hearts

SAPK3 Reverse ACC CCC CAA ACC ACC CAG AGG TCG

60°C RT-PCR of SAPK3 from mouse embryonic hearts

OMPT7 Forward ATA CGA CTC ACT ATA GGG AGA CC 50°C Confirmation of the rat OMP25 sequence in pcDNA3(HA)

ScribF GTG CTG GAG TCC GAG TTG GTG ACA 56°C Colony screens of suspected Scribble clones from yeast two-hybrid screening

ScribR TCA GCA GGG AGA CAG CAT GGT CGT

56°C Colony screens of suspected Scribble clones from yeast two-hybrid screening

Appendix I continued: List of Primers Used During the Course of this PhD Project

Page 253: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Primer Name Sequence (5’ to 3’) Annealing Temp Used

Purpose

OMPSP6 Reverse GCT CTA GCA TTT AGG TGA CAC 50°C Confirmation of the rat OMP25 sequence in pcDNA3(HA)

OMP SDM Forward GAC GGA GAG CCG TGA GGA GTT CCT GTA GCT G

55°C Inserting a STOP codon into the OMP25 insert to truncate the protein at amino acid 177 in the pcDNA3 and pGEX-6P vector

OMP SDM Reverse CAG CTA CAG GAA CTC CTC ACG GCT CTC CGT C

55°C Inserting a STOP codon into the OMP25 insert to truncate the protein at amino acid 177 in the pcDNA3 and pGEX-6P vector

OMP GST Forward CTA GGA TCC ATG TTC CTG AGG GGG TAT AAG

50°C Subcloning full-length OMP25 from pcDNA3 vector into pGEX-6P vector

OMP GST Reverse GAG GAA TTC TCA GAG CTG CTT TCG GTA TCT CAC

50°C Subcloning full-length OMP25 from pcDNA3 vector into pGEX-6P vector

OMP NLS Forward CTC GGA TCC CCT CCA AAA AAG AAG AGA AAG GTA GCT ATG TTC CTG AGG GGG TAT AAG AGA GAG

40°C Inserting a Nuclear Localisation Signal (NLS) into the truncated OMP25 insert (1-177) at the N-terminus of the protein

OMP NLS Reverse GAA TCT AGA TCA CGG CTC TCC GTC 40°C Inserting a Nuclear Localisation Signal (NLS) into the truncated OMP25 insert (1-177) at the N-terminus of the protein

Appendix I continued: List of Primers Used During the Course of this PhD Project

Page 254: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Primer Name Sequence (5’ to 3’) Annealing Temp Used

Purpose

Sab 1 Forward SDM GCC TGG ATC TGC CCG CCC CTG TGT CCC TGT C

55°C To mutate serine 321 in the Sab protein to an alanine residue

Sab 2 Reverse SDM CCC TCA GCC GCG GTG CTG CTT TGG CTC TTA C

55°C To mutate serine 391 in the Sab protein to an alanine residue

Sab Site 1 Mut Forward GAG GCC TGG CAG CCT GAG TTC GAC CAG CCC TGA GGG CCT GTC AGA G

55°C To mutate the sequence surrounding Ser321 to that surrounding Ser391 (321/391 mutant)

Sab Site 1 Mut Reverse CTC TGA CAG GCC CTC AGG GCT GGT CGA ACT CAG GCT GCC AGG CCT C

55°C To mutate the sequence surrounding Ser321 to that surrounding Ser391 (321/391 mutant)

Sab Site 2 Mut Forward GCA GTA AGA GCC AAG ACC TCC CCT CCC CTG TGA GCC AGG CCT TGG

55°C To mutate the sequence surrounding Ser391 to that surrounding Ser321 (391/321 mutant)

Sab Site 2 Mut Reverse CCA AGG CCT GGC TCA CAG GGG AGG GGA GGT CTT GGC TCT TAC TGC

55°C To mutate the sequence surrounding Ser391 to that surrounding Ser321 (391/321 mutant)

Sab 1 Reverse SDM GAC AGG GAC ACA GGG GCG GGC AGA TCC AGG C

55°C To mutate serine 321 in the Sab protein to an alanine residue

Sab 2 Forward SDM GTA AGA GCC AAA GCA GCA CCG CCC CTG AGG G

55°C To mutate serine 391 in the Sab protein to an alanine residue

Appendix I continued: List of Primers Used During the Course of this PhD Project

Page 255: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Appendix I continued: List of Primers Used During the Course of this PhD Project

Primer Name Sequence (5’ to 3’) Annealing Temp Used

Purpose

323 Glu Forward GGA TCT GCC CAG CCC TGA GTC CCT GTC AGA G

55°C To mutate amino acid residue Val323 in the human Sab protein to a glutamic acid residue

323 Glu Reverse CTC TGA CAG GGA CTC AGG GCT GGG CAG ATC C

55°C To mutate amino acid residue Val323 in the human Sab protein to a glutamic acid residue

323 Ala Forward GGA TCT GCC CAG CCC TGC GTC CCT GTC AGA G

55°C To mutate amino acid residue Val323 in the human Sab protein to an alanine residue

323 Ala Reverse CTC TGA CAG GGA CGC AGG GCT GGG CAG ATC C

55°C To mutate amino acid residue Val323 in the human Sab protein to an alanine residue

324 Glu Forward GCC TGG ATC TGC CCA GCC CTG TGG AAC TGT CAG AG

55°C To mutate amino acid residue Ser324 in the human Sab protein to a glutamic acid residue

324 Glu Reverse CTC TGA CAG TTC CAC AGG GCT GGG CAG ATC CAG GC

55°C To mutate amino acid residue Ser324 in the human Sab protein to a glutamic acid residue

324 Ala Forward GGA TCT GCC CAG CCC TGT GGC CCT GTC AGA G

55°C To mutate amino acid residue Ser324 in the human Sab protein to an alanine residue

324 Ala Reverse CTC TGA CAG GGC CAC AGG GCT GGG CAG ATC C

55°C To mutate amino acid residue Ser324 in the human Sab protein to an alanine residue

Page 256: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Appendix III

Page 257: The subcellular localisation, tissue expression, substrate ... · PKN Protein kinase N PMSF Phenyl methyl sulfonyl fluoride PSD-95 Post-synaptic density-95 PTB Phospho-tyrosine binding

Appendix III: Screening a rat heart cDNA library for SAPK3 with a SAPK3-specific monoclonal antibody

1° Screen 2° Screen-a screen of~1.6 x 105 clones producedone positive

-the positive clone was re-screened to further purify it to 5% of the population

3° Screen4° Screen-a final round of screening ensured the positive clonemade up 100% of the population

-re-screening again brought the percentage of positives to ~25%

Isolated DNA,sequencing results showed positive clone to be SAPK3