Top Banner
Good, better, best. Never let it rest. Until your good is better and your better is best Tim Duncan
114

Good, better, best. Never let it rest. Until your good is ...

Jan 24, 2022

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Good, better, best. Never let it rest. Until your good is ...

Good, better, best.

Never let it rest.

Until your good is better and your better is best

Tim Duncan

Page 2: Good, better, best. Never let it rest. Until your good is ...

Intracellular localization and regulation of

Gelatinase-A in zebrafish skeletal muscle

by

Amina Mohammed Ahmed Fallata

Bachelor of Science, King Abdul-Aziz University, 2009

A Thesis Submitted in Partial Fulfillment of the Requirements for the Degree of

Master of Science

in the Graduate Academic Unit of Biology

Supervisor: Bryan D. Crawford, Ph. D., Department of Biology

Examining Board: Les Cwynar, Ph. D., Department of Biology, Chair Shawn R. MacLellan, Ph. D., Department of Biology David Lentz, Ph. D., Department of Geology

This thesis is accepted by the Dean of Graduate Studies

THE UNIVERSITY OF NEW BRUNSWICK

November, 2015

©Amina M. Fallata, 2016

Page 3: Good, better, best. Never let it rest. Until your good is ...

ii

ABSTRACT

Matrix metalloproteinase (MMPs) are class-I secreted proteins known to function in

extracellular matrix remodeling. However, studies in the last decade and a half revealed

the unexpected presence of MMP-2 (a.k.a. gelatinase-A) intracellularly, within

cardiomyocytes and implicated them in the pathology of ischemia/reperfusion injury

(IRI). Furthermore, the activity of this protease in mammals is controlled by

phosphorylation implicating the existence of unknown kinases and phosphatases, and

possibly a signalling system that modulate MMP-2 activity inside cells. Two questions

that emerge from these discoveries are (1) is the intracellular localization of gelatinase-A

is something common in striated muscle, and (2) is its regulation by phosphorylation of

physiological significance? Answering these questions is the objective of this thesis.

Using immunofluorescence, confocal microscopy, and ultrathin sectioning, I have

confirmed the intracellular localization of Mmp2 in zebrafish skeletal muscle. However, I

observed zebrafish Mmp2 accumulating on M-bands within sarcomeres, rather than in the

Z-discs as has been reported for mammalian MMP-2 within cardiomyocytes. I also note

that the signal sequence that directs this protease into the secretory pathway is

consistently poorly recognized, indicating a selective pressure for maintaining a

significant intracellular portion of this enzyme. While I was unable to determine the

phosphorylation status of Mmp2 purified from zebrafish muscle, there are high

probability phosphorylation sites in the Mmp2 sequence that are well- conserved among

homologues of this protease for which sequence is available. Thus I show that the

intracellular localization of gelatinase-A proteases within the sarcomere of striated

Page 4: Good, better, best. Never let it rest. Until your good is ...

iii

muscle is not unique to mammalian cardiomyocytes, and that its regulation by

phosphorylation is likely an evolutionarily conserved characteristic of physiological

significance. I speculate that this protease is a previously unrecognized component of the

mechanism that regulates protein turnover within the contractile apparatus of striated

muscle.

Page 5: Good, better, best. Never let it rest. Until your good is ...

iv

DEDICATION

To my father and to my mom

Thanks for everything

Page 6: Good, better, best. Never let it rest. Until your good is ...

v

ACKNOWLEDGEMENTS

First of all, I would like to give my sincere appreciation to my supervisor Dr. Bryan

Crawford for his advice to help me complete my thesis. Without him, I would not have

been able to finish my work. Also I would like to thank my committee members Dr.

Tillmann Benfey and Dr. Katherine Barclay for their help to complete my thesis. A super

thanks goes to my friend Sheila Thompson, for her support and suggestions in writing my

thesis. I would also like to thank Christopher Small, Kelsey Katherine Mann, Aaron

Frenette, and all my friends in Bryan’s lab for their help, as well as Robyn O’Keefe and

Robyn Shortt for their care of the fish.

I would like to thank King Abdullah Bin Abdul-Aziz of the Kingdom of Saudi

Arabia for giving me the chance to achieve my dreams and enrol in his Scholarship

program. Appreciation is also due to the Saudi Cultural Bureau in Canada for everything

they helped me with during my study in Canada. A special thanks is in order for my

supervisor in the Embassy Dr. Maha Abou-Elghit for her help and advice during my

study time.

Finally, I would like to give big thanks to my family: my mother (Fatima), brothers

(Nizar and Raed), sisters (Dr. Eman and Ebtehal), and grandpas (Dr. Abdul Razzaq

Fallatah and Abdul Rahim Fallatah), as well as to my friend Eman Mohsen Basaheeh and

to other friends and relatives back home, for their encouragement, love, support and

prayers.

Page 7: Good, better, best. Never let it rest. Until your good is ...

vi

!

شك! & تق#"!

<تق!' بج;1: 2لشك$ & 2لإمتنا( %لى 2لمش$6 2ل!$2سي 2ل!كت&$ ب$12( ك$&ف&$! لما ق!م- لي م( !ع' &%$شا!

! الله ب. عب+*لع&'& (%حم" الله), 0ج;! م: 9ج7 ن-7 !,جة )لماجست-,. ك3ل$ )لشك, 0 )لتق!-, لبعثة )لمل$ عب!

. #لملحق9ة #لثقاف9ة #لسع5%9ة بكن%# لما ق%م45 لي م1 %ع/ .ثناء فت$' #ل%$#سة!0'/. 'لتعل+* 'لعالي 'لسع!#", !

لأمي *لس-': فا8مة # لإخ#*تي #ك2ل1 لجم-ع *لأ+! # *لأص'قاء #ك! )لس%" محم" ! لأبي .لشك* م'ص'& %$ضا

م2 ساع(ني ' ت/ك-ني ب(ع'& صالحة.

!

Page 8: Good, better, best. Never let it rest. Until your good is ...

vii

TABLE OF CONTENTS

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

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

Acknowledgements ........................................................................................................... v

Table of contents ............................................................................................................. vii

List of figures ..................................................................................................................... x

List of abbreviations ........................................................................................................ xi

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

1.1 Matrix Metalloproteinases (MMPs) .......................................................................... 1

1.1.1 Matrix Metalloproteinase structure and biochemistry ....................................... 3

1.1.2 The function of Matrix Metalloproteinases ....................................................... 7

1.1.2.1 Extracellular matrix remodelling ................................................................ 7

1.1.2.2 Matrix Metalloproteinase in development and disease ............................... 8

1.2 Intracellular localization of MMPs ........................................................................... 8

1.3 Muscle structure and function ................................................................................. 10

1.4 MMPs in ischemia-reperfusion injury .................................................................... 14

1.5 Zebrafish as model system for the study of muscle ................................................ 15

1.6 Objectives ............................................................................................................... 16

1.7 Overall hypothesis .................................................................................................. 17

1.8 References ............................................................................................................... 19

Page 9: Good, better, best. Never let it rest. Until your good is ...

viii

Chapter 2: Intracellular localization of gelatinase-A (Mmp2) in zebrafish skeletal

muscle ............................................................................................................................... 24

2.1 Introduction ............................................................................................................. 24

2.2 Materials and methods ............................................................................................ 29

2.2.1 Spawning zebrafish and collecting embryos .................................................... 29

2.2.2 Immunostaining and confocal microscopy ...................................................... 29

2.2.3 Cryo-sectioning ................................................................................................ 30

2.2.4 Prediction the signal sequence cleavage sites in Gelatinase-A, Gelatinase-B

and BiP (Binding immunoglobulin protein) using SignalP ...................................... 30

2.2.5 Statistical analysis ............................................................................................ 31

2.3 Results ..................................................................................................................... 32

2.4 Discussion ............................................................................................................... 38

2.5 References ............................................................................................................... 40

Chapter 3: Phosphorylation status of Mmp2 in zebrafish myocytes ......................... 42

3.1 Introduction ............................................................................................................. 42

3.2 Materials and methods ............................................................................................ 46

3.2.1 Tissue preparation ............................................................................................ 46

3.2.2 Isolation of Gelatinases using gelatin-affinity chromatography ...................... 46

3.2.3 SDS-polyacrylaminde gel electrophoresis and phospho-protein identification48

3.2.4 Immunoblots .................................................................................................... 48

3.2.5 Gelatin zymography ......................................................................................... 49

3.2.6 Silver staining .................................................................................................. 50

Page 10: Good, better, best. Never let it rest. Until your good is ...

ix

3.2.7 Prediction of phosphorylation sites within vertebrate Gelatinase-A homologues

................................................................................................................................... 51

3.3 Results ..................................................................................................................... 52

3.4 Discussion ............................................................................................................... 62

3.5 References ............................................................................................................... 64

Chapter 4: General Discussion and Conclusions ......................................................... 66

4.1 Mmp2 is unequivocally localized intracellularly within skeletal muscle in zebrafish

....................................................................................................................................... 67

4.2 Evolutionary argument for an important physiological function of intracellular

gelatinase-A .................................................................................................................. 70

4.3 What role(s) does gelatinase-A play in the sarcomere? .......................................... 74

4.4 Conclusion .............................................................................................................. 78

4.5 References ............................................................................................................... 80

Appendix A ....................................................................................................................... 84

Appendix B ....................................................................................................................... 91

Curriculum Vitae

Page 11: Good, better, best. Never let it rest. Until your good is ...

x

LIST OF FIGURES

Chapter 1 figures

Figure 1.1 .............................................................................................................. 4!

Figure 1.2 ............................................................................................................ 12!

Chapter 2 figures

Figure 2.1 ............................................................................................................ 27!

Figure 2.2 ............................................................................................................ 35!

Figure 2.3 ............................................................................................................ 35!

Figure 2.4 ............................................................................................................ 36!

Figure 2.5 ............................................................................................................ 37!

Chapter 3 figures

Figure 3.1 ............................................................................................................ 55!

Figure 3.2 ............................................................................................................ 56!

Figure 3.3 ............................................................................................................ 57!

Figure 3.4 ............................................................................................................ 58!

Figure 3.5 ............................................................................................................ 59!

Figure 3.6 ............................................................................................................ 60!

Figure 3.7 ............................................................................................................ 60!

Figure 3.8 ............................................................................................................ 61!

Page 12: Good, better, best. Never let it rest. Until your good is ...

xi

LIST OF ABBREVIATIONS

α-actinin Alpha-actinin

ATP Adenosine 5’-triphosphate

ADP Adenosine 5’-diphosphate

BiP Binding immunoglobulin protein

BSA Bovine serum albumin

DMSO Dimethyl sulfoxide

DNA Deoxyribonucleic acid

dNTP Deoxyribonucleotide triphosphate dT Deoxy thymine nucleotides

ECL Enhanced Chemiluminescence

ECM Extracellular matrix

ERM Embryo rearing medium

ER Endoplasmic reticulum

g Gravity force

GPI Glycosylphosphatidylinositol

hpf Hours post-fertilization

HRP Horse-radish peroxidase

H2O2 Hydrogen peroxide

IRI Ischemia and reperfusion injury

MMP Matrix metalloproteinase

MT-MMP Membrane type matrix metalloproteinase

Mmp2 Matrix metalloproteinase 2 protein from zebrafish

mmp2 Matrix metalloproteinase 2 gene in zebrafish

MMP-2 Matrix metalloproteinase-2 protein from human (Homo sapiens)

Page 13: Good, better, best. Never let it rest. Until your good is ...

xii

mmp-2 Matrix metalloproteinase-2 gene in human

MMP2 Matrix metalloproteinase 2 protein from rat, or mouse

MuRF Muscle-specific RING finger

NO· Nitric oxide

PBS Phosphate Buffered Saline

PBSTx Phosphate buffered saline solution with tritonX-100

PBSTw Phosphate buffered saline with Tween-20

ONOO- Peroxynitrite

PVDF Polyvinylidene difluoride

ROS Reactive oxygen species RECK Reversion-inducing-cysteine-rich protein with kazal motifs RT-PCR Reverse transcriptase polymerase chain reaction

RNA Ribonucleic acid

SSs Signal sequences

SRP Signal recognition particle

SRPR Signal recognition particle receptor

SDS-PAGE Sodium dodecyl sulfate- polyacrylamide gel electrophoresis

SERCA Sarco(endo)plasmic reticulum Ca+2- ATPase

·O2 - Superoxide

TAILS Terminal Amine Isotopic Labeling of Substrates

TEMED Tetramethylethylenediamine

TEM Transmission Electron Microscope

TIMP Tissue Inhibitor of metalloproteinase

!!

Page 14: Good, better, best. Never let it rest. Until your good is ...

xiii

!*Note on nomenclature: In this thesis, I have used the standard nomenclature of matrix metalloproteinases. When referring to zebrafish proteins, normal font is used with the first letter capitalized; for their genes or transcripts lowercase italics are used with no hyphen between the protein/gene name and its number (e.g., the zebrafish mmp2 gene and the zebrafish Mmp2 protein).When referring to mouse/rat and human proteins, the names are written in capital letters, with a hyphen used only for human proteins (e.g., the mouse/rat MMP2 and the human MMP-2). In the transcripts for both mouse/rat and human, lowercase italics are used (e.g., the mouse/rat mmp2 and the human mmp-2).

Page 15: Good, better, best. Never let it rest. Until your good is ...

1

Chapter 1: Introduction

1.1 Matrix Metalloproteinases (MMPs)

Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases whose

importance to living organisms is profound. MMPs play significant roles in diverse

physiological and pathological processes, many of which have been discovered in the

five decades since the discovery of the first MMP (Gross and Lapiere 1962; reviewed in

Lapière 2005; Page-McCaw et al. 2007; Kessenbrock et al. 2010). ‘Matrix

metalloproteinases’, ‘matrixins’ or ‘matrix degrading metalloenzymes’ are all terms used

to describe the same family of enzymes, although matrix metalloproteinases is the most

popular (reviewed in Amălinei et al. 2007). These proteases were first recognized in

experiments investigating the degradation of collagen fibres during tadpole

metamorphosis (Gross and Lapière, 1962). Since then, dozens of MMPs have been

characterized in animals ranging from Cnidarians to Vertebrates, and genes encoding

related metalloproteinases have been identified in plants, viruses and prokaryotes

(reviewed in Gomis-Ruth 2009). All MMPs are synthesized in a latent 'pro-enzyme' form

and need to be activated post-translationally in order to function. In addition to post-

translational activation, MMPs are subject to reversible and irreversible post-translational

inhibition, as the result of forming complexes with endogenous inhibitors such as the

tissue inhibitors of metalloproteinases (TIMPs) or reversion-inducing cysteine-rich

protein with Kazal motifs (RECK), and ultimately proteolytic degradation of the enzyme

(reviewed in Tallant et al. 2010). The complement of MMPs encoded by chordate

Page 16: Good, better, best. Never let it rest. Until your good is ...

2

genomes is quite diverse. The ascidian Ciona intestinalis has only 7 MMPs (Huxley-

Jones et al. 2007), whereas vertebrates generally have two-dozen or more. Zebrafish

(Danio rerio), for example, have 25 MMPs (Wyatt et al. 2009), humans have 24 and

mice have 25 (reviewed in Jackson et al. 2010), Xenopus leavis has 29 and X. tropicalis

has 27 (Fu et al. 2009). The larger number of MMP genes in the genomes of vertebrates

may be the result of duplication and expansion of the ancestral chordate MMP genes

during vertebrate evolution, or the loss of ancestral deuterostome MMP genes during

ascidian evolution, as the sea urchin Strongylocentrotus purpuratus has 26 MMPs

(reviewed in Fanjul-Fernández et al. 2010), or a combination of both.

Mammalian MMPs have been classified on the basis of their substrate affinities,

including gelatinases (MMP-2 and MMP-9), collagenases (MMP-1, 8 and 13) and

stromelysins (MMP-3, 10 and 11), matrilysins (MMP-7 and MMP-26), as well as on the

basis of their subcellular localization, as secreted (MMP-1, 2, 7, 8, 9, 10, 11, 12, 13, 18,

19, 20, 21, 23, 27, 28), membrane type (MMP-14 (MT1-MMP), MMP-15 (MT2-MMP),

MMP-16 (MT3-MMP) and MMP-24 (MT5-MMP)), or the glycosyl-phosphatidyl-

inositol (GPI) membrane tethered MMPs (MMP-17 (MT4-MMP) and MMP-25 (MT6-

MMP)) (reviewed in Visse and Nagase 2003). The gelatinases degrade the basement

membrane (laminin), denatured collagen (a.k.a. gelatin), and collagen types IV, V and XI

(reviewed in Murphy and Nagase 2008). The collagenases degrade fibrillar collagens

such as collagen type I, II and III, as well as other extracellular matrix proteins (reviewed

in Murphy and Nagase 2008). Stromelysins break down many extracellular matrix

proteins except collagens (reviewed in Klein and Bischoff 2011). The membrane type

MMPs digest type I, II, and III collagens, and most of them degrade fibronectin and

Page 17: Good, better, best. Never let it rest. Until your good is ...

3

laminin as well (reviewed in Amălinei et al. 2007). While these overlapping substrate

specificities are interesting and consistent with the evolutionary divergence of the MMPs

as a family, it is worth noting that most of what is known about MMP-substrate

interactions is based on biochemical analysis in vitro, and may not provide an accurate

picture of their biologically relevant proteolytic activities.

1.1.1 Matrix Metalloproteinase structure and biochemistry

Most MMPs share four identifiable sequence hallmarks, and can be distinguished

according to the presence or absence of specific structures. From amino to carboxyl,

these consist of a secretory signal peptide motif, a pro-peptide domain, a catalytic

domain, a hinge region, and finally a hemopexin-like domain (reviewed in Nagase et al.

2006) (Figure 1.1). All MMPs have an amino-terminal signal peptide domain, which is

responsible for delivering the nascent protein to the endoplasmic reticulum (ER) during

translation for export (or anchoring into the plasma membrane through the glycosyl-

phosphatidyl-inositol (GPI) linkage or transmembrane domain) (reviewed in Murphy and

Nagase 2010).

The pro-peptide domain, which consists of about 80 to 100 moderately-conserved

amino acids (reviewed in Nagase et al. 2006), contains a cystine switch motif!

(PRCGxPD) (where ‘x’ indicates a non-conserved residue). The pro-peptide plays an

important role in controlling the activity of MMPs by folding into the catalytic site,

positioning the thiol of the cysteine switch in such a way as to interact with the catalytic

zinc ion, thereby preventing it from activating water molecules and thus inhibiting

proteolytic activity (Van Wart and Birkedal-Hansen 1990; reviewed in Cauwe and

Page 18: Good, better, best. Never let it rest. Until your good is ...

4

Opdenakker 2010). Activation of MMPs therefore requires disruption of the interaction

between the pro-peptide and the catalytic site; this is typically accomplished by

proteolytic removal of the N-terminal pro-peptide domain, but can also result from

Figure 1.1: MMP Structure. All MMPs share an architecture consisting of a signal peptide (S), pro-peptide (Pro) and catalytic domain. Some other structures are present in some MMPs but not all; for example type II fibronectin repeats (Fn), cysteine-rich domains (Ca), immunoglobulin-like domains (Ig), type I or II transmembrane domains (I or II), vitronectin inserts (V), glycophosphatidylinistitol linkages (G), and cytoplasmic tails (Cp) (from Visse and Nagase 2003).

chemical modification of the cysteine in the switch motif (Okamoto et al. 2001).

The catalytic domain, common to all MMPs, binds two zinc ions, one for structure

and the other as a catalyst, and three calcium ions that provide stability to the structure.

Page 19: Good, better, best. Never let it rest. Until your good is ...

5

This zinc-binding motif of the catalytic domain contains the consensus sequence

HExGHxxGxxH, in which the three histidines coordinate the catalytic Zn+2 ion and the

glutamic acid side chain serves as the general base, which buffers the protons released by

the activated water molecules (reviewed in Visse and Nagase 2003). Additionally, most

MMPs exhibit a conserved methionine residue (Met-turn) in the catalytic domain for

maintaining the structure of the S1-specificity pocket (reviewed in Visse and Nagase

2003). Carboxyl to the catalytic domain, a hinge region works as a link between the

catalytic domain and the hemopexin-like domain. The hemopexin-like domain is

responsible for interaction between the MMP enzymes and their substrates as well as

with endogenous tissue inhibitors of metalloproteinases (TIMPs) (reviewed in Amălinei

et al. 2007). However, some MMPs lack the hinge region and hemopexin-like domains,

for example MMP-7 (matrilysin-1), MMP-26 (matrilysin-2) and MMP-23 (reviewed in

Cauwe and Opdenakker 2010). In addition, there are three fibronectin type II repeats

within the catalytic domains of MMP-2 (a.k.a. Gelatinase-A) and MMP-9 (a.k.a.

Gelatinase-B), which also facilitate interaction with their substrates (reviewed in

Amălinei et al. 2007).

There is a significant level of complexity in the regulation of MMPs and the control

of their activity, including at the level of transcription, translation, and post-translational

processes including secretion, activation of the pro-MMPs (zymogen), and inhibition the

MMPs' activity by TIMPs (reviewed in Page-McCaw et al. 2007; Mannello and Medda

2012). Most MMPs are transcriptionally regulated by hormones, cytokines, growth

factors, and cell-cell and cell-matrix interactions (reviewed in Amălinei et al. 2007).

Page 20: Good, better, best. Never let it rest. Until your good is ...

6

Post-translationally, MMP-11 and MT-MMPs can be activated intracellularly by furin

pro-protein convertase, as these MMPs bear conserved Rx(R/K)R paired basic amino

acid cleavage sites for this serine protease between their pro-peptides and catalytic

domains (Pei and Weiss 1995; reviewed in Nagase et al. 2006). Other MMPs are

activated extracellularly by collaborating proteases including other MMPs (reviewed in

Klein and Bischoff 2011).!

Non-proteolytic chemical agents, especially in pathological contexts, can cause

activation of full-length MMPs. For instance peroxynitrite (ONOO-), which occurs in

cells under oxidative stress, can nitrosylate the thiol of the cysteine switch rendering it

ineffective, and thereby giving rise to an activated full-length MMP (reviewed in Schulz

2007). Once MMPs are activated, TIMPs and other endogenous inhibitors are responsible

for regulating their activity and protecting substrates from uncontrolled degradation and

associated diseases (reviewed in Loffek et al. 2011). Of particular interest is the recent

discovery that phosphorylation also modulates MMP activity (Sariahmetoglu et al. 2007).

This is notable because, while phosphorylation is a common post-translational

modification that is known to regulate the activity of many enzymes, it occurs as the

result of the activity of kinases and relatively high local concentrations of adenosine 5’-

triphosphate (ATP), and is therefore generally characteristic of intracellular, rather than

extracellular proteins. However it is worth noting that the biological importance of

‘exokinases’ has recently begun to emerge (Yalak et al. 2014) (discussed in Chapter 3),

making it inaccurate to consider phosphorylation as a strictly intracellular phenomenon.

Page 21: Good, better, best. Never let it rest. Until your good is ...

7

1.1.2 The function of Matrix Metalloproteinases

1.1.2.1 Extracellular matrix remodelling

The extracellular matrix (ECM) is a cross-linked matrix of insoluble glycoproteins

and protoglycans secreted by the cells of all multicellular organisms, which give tissues

structural integrity and many of their biological functions. In animals, the ECM is

composed of collagen, elastin, fibronectin, laminin, nidogen and other large insoluble

glycoproteins that are ubiquitous constituents of almost all tissues (reviewed in Frantz et

al. 2010). Although considered by some as ‘packing material’ and uninteresting, the

many dynamic and pleiotropic roles of the ECM are continuing to emerge and are

becoming central in understanding the development, function and evolution of animal

tissues. The matrix influences cell differentiation, proliferation, and tissue reorganization

by binding directly to receptors such as integrins (reviewed in Harburger and Calderwood

2009), and by modulating the presentation, stability and distribution of signalling

molecules such as growth factors and cytokines (reviewed in Venkatasubramanian 2012).

Thus the ECM plays many essential roles in cellular processes and embryonic

development; providing permissive and non-permissive substrates for cell migration,

permitting and resisting changes in cell-shape, and promoting or inhibiting cell

proliferation and survival. Further, the composition and even mechanical load on the

matrix influences cell fate decisions and differentiation, and works as a bio-scaffold for

rebuilding injured cells and tissues (reviewed in Lu et al. 2011).

Remodelling of the ECM during or after embryonic development is clearly crucial

to the formation new tissue and structures in vivo. However, it is worth noting that under

Page 22: Good, better, best. Never let it rest. Until your good is ...

8

normal conditions, the level of MMP expression is generally low in adult tissues, and its

induction depends on several factors, many of which are associated with inflammatory or

other pathological processes (reviewed in Phatharajaree et al. 2007).

1.1.2.2 Matrix Metalloproteinase in development and disease

MMPs are strongly implicated in many physiological and pathological processes.

The physiological processes include those associated with embryonic development, such

as angiogenesis, normal tissue remodeling, and in adults MMPs are required for normal

inflammatory response, wound healing and tissue homeostasis (reviewed in Visse and

Nagase 2003). MMPs are also implicated in a wide range of devastating diseases.

Examples include rheumatoid arthritis and osteoarthritis, in which collagenases (MMP-1,

-8, -13, and -18) are responsible for breaking down connective tissues (reviewed in

Vartak et al. 2007). MMP-3 expression in adults is associated with neurodegenerative

diseases such as Parkinson’s disease (reviewed in Kim and Hwang 2011) and many more.

The most relevant with respect to the work discussed in this thesis are the roles of MMP-

2 in muscle disorders. MMP-2 has a significant impact on muscle atrophy (Liu 2011)

and, intracellular MMP-2 is implicated in the degradation of intracellular sarcomeric

proteins in cardiomyocytes under oxidative stress (reviewed in Schulz 2007).

1.2 Intracellular localization of MMPs

For most of the past four decades, research on MMPs concentrated on their

canonical biological functions proteolyzing and remodeling extracellular proteins.

However, more recently evidence has demonstrated that MMPs can degrade non-

extracellular matrix proteins inside and outside the cells (reviewed in Cauwe and

Page 23: Good, better, best. Never let it rest. Until your good is ...

9

Opdenakker 2010). Several MMPs are detected at significant concentrations inside cells,

such as in the nucleus (MMP-1, -2, -3, -9, -13, -26 and -14) (Limb et al. 2005; Kwan et

al. 2004; Eguchi et al. 2008; Yang et al. 2010; Cuadrado et al. 2009; Zhang et al. 2002;

Yang et al. 2010), mitochondria (MMP-1, -2 and -9) (Limb et al. 2005; Wang et al. 2002;

Moshal et al. 2008; reviewed in Cauwe and Opdenakker 2010), and within sarcomeres in

cardiac myocytes (Sawicki et al. 2005). In the context of cardiac sarcomeres,

pathologically activated MMP-2 has been shown to degrade troponin I, myosin light

chain, α-actinin and titin (Wang et al. 2002; Sawicki et al. 2005; Sung et al. 2007; Ali et

al. 2010; reviewed in Ali et al. 2011a), all of which are essential sarcomeric proteins,

making the pathological activation of intracellular MMP-2 a direct cause of the loss of

muscle cell contractility in ischemia/reperfusion injury.

MMP-2 is the most abundant member of the MMP family. It is constitutively

expressed in most tissues and it is ubiquitous in cardiomyocytes (reviewed in Schulz

2007). The intracellular localization of MMP-2 and its pathological role in cardiac

muscle was reported a decade ago (Sawicki et al. 2005). Recently, Ali and colleagues

(2011b) reported the mechanisms that result in this unexpected distribution of this

ostensibly secreted enzyme. Firstly, the N-terminal signal sequence of the canonical

MMP-2- MEALMARGALTGPLRALCLLGCLLSHAAA - (the sequence of amino acids

that direct the newly synthesized peptide to the ER via signal recognition particles (SRP))

appears to interact only weakly with the SRP. The SRP binds secretory signals in nascent

polypeptides as they emerge from the large subunit of the ribosome, and simultaneously

binds the incoming tRNA binding site (the A-site in the large ribosomal subunit) causing

Page 24: Good, better, best. Never let it rest. Until your good is ...

10

a pause in translation. The paused translation complex is then bound by protein

translocator complexes embedded in the endoplasmic reticulum membrane. The SRP is

then released and translation continues, with the nascent protein being elongated into the

lumen of the endoplasmic reticulum (Cooper and Hausman 2009). From the endoplasmic

reticulum, secreted proteins proceed through the Golgi apparatus and into the constitutive

secretory pathway. Therefore, the efficiency with which a protein’s secretory signal is

recognized by the SRP determines what proportion of that protein is translocated into the

ER and thence into the secretory pathway. At least in the case of human MMP-2, a

significant proportion of the translated protein is not recognized by the SRP, and remains

cytosolic (Ali et al. 2011b). As will be discussed further in Chapter 2, this appears to be

an evolutionarily conserved feature of Gelatinase-A. Furthermore, alternatively spliced

mmp-2 transcripts expressed in human cardiomyocytes encode an MMP-2 protein

completely lacking an N-terminal secretory signal, contributing more of this protease to

the cytosolic pool (Ali et al. 2011b). To understand potential consequences of

metalloproteinases within muscle cells we must recall some basics of sarcomere structure

and function.

1.3 Muscle structure and function

Muscle cells are a highly specialized, essential cell type in most metazoans. In

addition to allowing voluntary movement under the control of the nervous system, the

contractions of muscles participate in maintaining body homeostasis by moving blood

through vessels, air or oxygenated water across gas-exchange surfaces, and food through

the digestive system (Arms & Camp 1997). All muscle cells contain specialized

Page 25: Good, better, best. Never let it rest. Until your good is ...

11

complexes of cytoskeletal proteins (primarily actin and myosin, with an array of

accessory proteins discussed below), which exert contractile force fuelled by the

regulated hydrolysis of ATP.

Vertebrates have both striated and smooth muscle. Much of the involuntary

musculature associated with the gut and vascular system is smooth muscle, in which the

contractile apparatus of the cells is not organized into repeating units. In contrast, the

contractile apparatus of cardiac and skeletal muscle is arranged in repeating units called

sarcomeres, which give these cells their characteristic striated appearance.

Cardiac and smooth muscle forms about 10% of the muscle mass of typical

vertebrates, in contrast to skeletal muscle, which is large and highly complex tissue

forming about 40% of the body weight in humans, and around 60 % of the total body

weight in fish (Sänger and Stoiber 2001). Muscle consists one or more fibres, each of

which consist of groups of myofibrils covered by sarcolemma (a.k.a. cell membrane).

The myofibrils are made up of linearly repeated sarcomeres. The sarcomere is a highly

organized structural unit of cytoskeletal proteins (see Figure 1.2). Thus, the muscle

consists of hierarchically organized groups of contractile fibres.

The contractile function of the sarcomere results from the interaction of two kinds

of myofilament proteins; actin that forms the main component of thin filaments, and

myosin that forms the thick filaments. Thin and thick filaments overlap and the myosin

protein converts the chemical energy of ATP into mechanical energy to drive the

filaments past one another, which causes muscular contraction. Inside the sarcomere, the

ends of the actin filaments are attached to the terminal Z-discs by α-actinin which extends

from the Z-disc into the light I-band.

Page 26: Good, better, best. Never let it rest. Until your good is ...

12

!!

!

Figure 1.2: A) Schematic diagram and B) electron micrograph of a longitudinal section through zebrafish muscle showing the main components of the sarcomere. Both A) and B) show the intra-sarcomeric A-band comprised of thick filament (myosin) spanning the M-band (green). The thin filaments (actin) are connected to the Z-discs and overlap with the myosin filament. α–actinin, located on the Z-discs (red), transmits the force from the interaction between actin and myosin. Titin protein extends from the Z-discs to the M-band.

Sarcomere

M-band Z-disc

A-band

I-band I-band

Z-disc

Actin filament

Titin

α–actinin

Myosin heads &

necks Myosin tails

A)

Tn#I

Troponin I Tn#I

B)

A&band

M&band

Z&disc

Page 27: Good, better, best. Never let it rest. Until your good is ...

13

At their free ends, actin filaments are stabilized by trompomodulin caps. Along their

length, which is determined by nebulin, the actin filaments are bound by a complex of

proteins including tropomyosin (Tm), troponin T (tropomyosin-binding), troponin C

(Ca+2 binding), and troponin I (inhibitory proteins), which regulate the exposure of actin

binding sites in a calcium-dependent manner.

Myosin thick filaments are composed of tail, neck and P-loop ATP-ase head

domains; the tails of multiple myosin heavy chains interact to form the thick filament,

and are oriented towards the middle of the sarcomere (M-band), while the heads are

oriented away from the M-band, such that they can interact with the actin filaments

(Sarantitis et al. 2012). In addition, there are titin and nebulin macromolecule proteins

that form a part of the sarcomere structure. Titin is the largest protein in nature and

extends from Z-band of the sarcomere to the M-band in the middle (reviewed in Kruger

and Linke 2011). It works as a molecular spring and scaffold protein that keeps the

myosin filaments in the middle of the sarcomere. Finally, nebulin is connected to the Z-

discs and regulates the growth of actin filaments, acting as a molecular ruler to adjust the

length of thin filaments in sarcomeres (reviewed in Fowler et al. 2006).

The sliding filament model of contraction in striated muscle was first proposed in

1954 by two independent groups (Huxley & Niedergerke 1954; Huxley & Hanson 1954).

Contraction is triggered by the release of acetylcholine by motor neuron synapses at the

neuromuscular junction, causing depolarization of the muscle cell membrane. This gives

rise to the liberation of calcium from the sarcoplasmic reticulum into the cytoplasm of the

muscle cell. Ca+2 binds troponin C causing a conformational change which exposes

myosin binding sites on the actin filaments. In each cycle of contraction, the myosin

Page 28: Good, better, best. Never let it rest. Until your good is ...

14

heads bind ATP, hydrolyze the ATP to ADP+Pi which triggers a dramatic change in

conformation typical of P-loop ATPases, and while in an ADP-bound state they gain

affinity for actin and bind exposed sites on the thin filaments. This binding triggers the

relaxation of the lever arm of the head group which exerts contractile force on the actin

filament, and the expulsion of the ADP. The myosin molecule is then free to bind ATP

which triggers the release of the thin filament and the cycle repeats, causing the myosin

thick filament to pull its way along the actin filaments in ratchet-like steps.

During relaxation of the muscle, the sarcoplasmic reticulum Ca+2 - ATPase

(SERCA)- dependent calcium transporters present in the membrane of the sarcoplasmic

reticulum pump calcium ions back into storage. This reverses the conformational change

in tropomyosin, eliminating the interaction between myosin head groups and actin thin

filaments, and allowing the thick and thin filaments to slide past each other back to their

original configuration.

1.4 MMPs in ischemia-reperfusion injury

Ischemia occurs when the flow of blood to tissues is temporarily blocked. Ischemia

and reperfusion injury (IRI) is a pathological situation arising when oxygenated blood

flow is restored after a period of ischemia. This leads to the production of reactive

oxygen species (ROS) as built-up reducing power from electron transport is transferred to

oxygen faster than endogenous mechanisms can manage (reviewed in Chow et al. 2007).

Prolonged ischemia causes an alteration in cellular function and metabolism by reducing

oxidative phosphorylation and depleting high-energy phosphate molecules, such as ATP.

Furthermore, ATP catabolism during ischemia-altered sodium/potassium ATPase activity

Page 29: Good, better, best. Never let it rest. Until your good is ...

15

leads to the influx of sodium, calcium and water molecules into cells.

Reperfusion after ischemia leads to inflammation of cells due to ionic imbalance

and production of ROS. The inability of the cell to eliminate ROS such as superoxide

(·O2-), and nitric oxide (NO·) results in the production of highly reactive molecules such

as peroxynitrite (ONOO-). The latter in particular stimulates the activity of MMPs due to

the nitrosylation and/or glutathiolation of the auto-inhibitory cystine switch (Viappiani et

al. 2009). As a result, the intracellular MMP-2 becomes activated and destroys the

contractile proteins within the sarcomere (reviewed in Ali et al. 2011a). Making matters

worse, peroxynitrite additionally inhibits the action of TIMPs in vitro (Donnini et al.

2008), which could exacerbate the activity of MMPs in ischemia-reperfusion injury.

1.5 Zebrafish as model system for the study of muscle

For decades now, the zebrafish has been described as a tractable vertebrate model

system for research of many types. It has been extensively utilized for studying genetics

and human diseases (reviewed in Lieschke and Currie 2007). The advantage of zebrafish

over insect or other invertebrate model systems is primarily that, as a vertebrate, it is less

evolutionarily divergent from mammals, making it easier to generalize findings of

biomedical significance. The advantages zebrafish have over mammalian models such as

mice include the optical clarity of the externally fertilized embryos that simplify

observation of biological processes and their rapid, external development at room

temperature, requiring only 72 hours to become feeding larvae, compared to almost a

month for rats or mice. Furthermore, the cost to obtain and raise zebrafish is less that a

thousandth of the cost associated with mice. Many of the attractive features of

Page 30: Good, better, best. Never let it rest. Until your good is ...

16

mammalian systems, such as the availability of many mutant and transgenic lines, a

sequenced genome, amenability to genome editing, and reverse genetic approaches, also

apply to zebrafish. An additional advantage is the availability and amenability of the

zebrafish to high throughput and transgenic approaches; thousands of embryos can be

arrayed in plates for treatment with libraries of compounds or other combinatorial

experiments, and we can order various types of transgenic fish from stock centres or

engineer our own transgenic fish as necessary. What is more crucial is their fundamental

similarity to human and other vertebrates, particularly with respect to muscular

development and function (reviewed in Squire et al. 2008). Thus, using zebrafish gives us

a good opportunity to study the action and activity of MMPs (Wyatt et al. 2009) and

conduct other biological research.

1.6 Objectives

The common ancestor of humans and zebrafish lived approximately 460 to 360

Mya (Campbell et al. 2008). Although many of the characteristics of this organism are

now obscure, it is clear that it had a gelatinase-A gene. What is less clear is whether the

protease encoded by that gene was inefficiently secreted (as is the case with modern

mammalian orthologues), whether it accumulated within the sarcomeres of ancestral

striated muscle, whether its activity was regulated by phosphorylation, what proteins it

degraded, and what, if any, roles it might have played in normal muscle cell physiology.

In this thesis I hope to shed some light on these questions by determining if zebrafish

Mmp2 is present within skeletal muscle sarcomeres and if it is phosphorylated.

Page 31: Good, better, best. Never let it rest. Until your good is ...

17

1.7 Overall hypothesis

My core hypothesis is that gelatinase-A has ancient conserved functions within the

sarcomere of striated muscle. My predictions are therefore: 1) gelatinase-A will be

observed to localize intracellularly in most, if not all, striated muscle cells rather than

this being an idiosyncratic characteristic of mammalian cardiac myocytes. 2) The

secretory signals of gelatinase-A proteins from phylogenetically diverse vertebrates will

be consistently poorly recognized as such, and therefore indicative of evolutionary

selection in favour of intracellular accumulation of this protease. Finally, 3) the

phosphorylation phenomenon and its role in modulating the activity of gelatinase-A will

be conserved across vertebrate species. The specific approaches I undertook to test these

predictions, which are presented in this thesis are as follows: 1) I unequivocally localized

zebrafish Mmp2 within the sarcomeres of zebrafish skeletal muscle using

immunofluorescence in both optical and physical sections of embryonic and adult

muscle. 2) I examined the predicted amino acid sequences encoding gelatinase-A genes

and compared their secretory signals to those of two efficiently ER-targeted proteins

(gelatinase-B and BiP) for all species for which good sequence data are available for all

three genes. I found that the N-terminal sequence of gelatinase-A proteins is significantly

less well recognized as such than the secretory signals of either gelatinase-Bs or BiPs.

Finally, 3) while I was unable to complete the analysis of its phosphorylation status

biochemically, I have developed a protocol to purify zebrafish Mmp2 protein from

skeletal muscle using gelatin-affinity chromatography, and the determination of its

phosphorylation status will only require small changes in the technique. In addition, I

analyzed the conservation of phosphorylation sites in available gelatinase-A sequence

Page 32: Good, better, best. Never let it rest. Until your good is ...

18

data, and found that several phosphorylation sites identified in human MMP-2 are also

conserved in zebrafish as well as in other species.

These findings support my core hypothesis, and I conclude this thesis with some

speculation regarding the conserved physiological function gelatinase-A may have within

the sarcomeres of striated muscle.

Page 33: Good, better, best. Never let it rest. Until your good is ...

19

1.8 References

Ali MA, Cho WJ, Hudson B, Kassiri Z, Granzier H, Schulz R. 2010. Titin is a target of matrix metalloproteinase-2: implications in myocardial ischemia/reperfusion injury. Circulation. 122(20): 2039-47.

Arms K, Camp PS. 1997. Biology. Fourth edition. Orlando (Fl): Saunders College Publishing. p. 844-862.

Ali MA, Fan X, Schulz R. 2011a. Cardiac sarcomeric proteins: novel intracellular targets of matrixmetalloproteinase-2 in heart disease. Trends Cardiovasc Med. 21(4): 112-118. Ali MA, Chow AK, Kandasamy AD, Fan X, West LJ, Crawford BD, Simmen T, Schulz R. 2011b. Mechanisms of cytosolic targeting of matrix metalloproteinase-2. J Cell Physiol. 227: 3397-3404. Amalinei C, Caruntu ID, Balan RA. 2007. Biology of metalloproteinases. Rom J Morphol Embryol. 48(4): 323–334. Campbell NA, Reece JB, Urry LA, Cain ML, Wasserman SA, Minorsky PV, Jackson RB. 2008. Biology. Eighth edition. San Francisco (CA): Pearson Education Inc. p. 654-665. Cauwe B, Opdenakker G. 2010. Intracellular substrate cleavage: a novel dimension in the biochemistry, biology and pathology of matrix metalloproteinases. Crit Rev Biochem Mol Biol. 45(5): 351–423.

Chow AK, Cena J, Schulz R. 2007. Acute actions and novel targets of matrix metalloproteinases in the heart and vasculature. Br J Pharmacol. 152(2): 189–205.

Cooper GM, Hausman RE. 2009. The cell: a molecular approach. Fifth edition. Washington (DC): The American Society of Microbiology Press. p. 383-432.

Cuadrado E, Rosell A, Borrell-Pages M, Garcia-Bonilla L, Hernandez- Guillamon M, Ortega-Aznar A, Montaner J. 2009. Matrix metalloproteinase-13 is activated and is found in the nucleus of neural cells after cerebral ischemia. J Cereb Blood Flow Metab. 29(2): 398–410.

Donnini S, Monti M, Roncon R, Morbidelli L, Rocchigiani M, Oliviero S, Casella L, Giachetti A, Schulz R, Ziche M. 2008. Peroxynitrite inactivates human-tissue inhibitor of metalloproteinase-4. FEBS Lett. 582(7): 1135–1140.

Eguchi T, Kubota S, Kawata K, Mukudai Y, Uehara J, Ohgawara T, Ibaragi S, Sasaki A, Kuboki T, Takigawa M. 2008. Novel transcription-factor-like function of human matrix metalloproteinase 3 regulating the CTGF/CCN2 gene. Mol Cell Biol. 28(7): 2391–413.

Page 34: Good, better, best. Never let it rest. Until your good is ...

20

Fanjul-Fernández M, Folgueras AR, Cabrera S, López-Otín C. 2010. Matrix metalloproteinases: evolution, gene regulation and functional analysis in mouse models. Biochim Biophys Acta.1803(1): 3-19.

Fowler VM, McKeown CR, Fischer RS. 2006. Nebulin: does it measure up as a ruler? Curr Biol. 16(1): R18–20.

Frantz C, Stewart KM, Weaver VM. 2010. The extracellular matrix at a glance. J Cell Sci. 123(24): 4195-4200.

Fu L, Das B, Mathew S, Shi YB. 2009. Genome-wide identification of Xenopus matrix metalloproteinases: conservation and unique duplications in amphibians. BMC Genomics. 10:81.

Gomis-Ruth FX. 2009. Catalytic domain architecture of metzincin metalloproteases. J Biol Chem. 284(23): 15353-7.

Gross J, Lapiere CM. 1962. Collagenolytic activity in amphibian tissues: a tissue culture assay. Proc Natl Acad Sci U S A. 48(6): 1014-1022.

Harburger DS, Calderwood DA. 2009. Integrin signalling at a glance. J Cell Sci. 122(2): 159-163.

Huxley-Jones J, Clarke TK, Beck C, Toubaris G, Robertson DL, Boot-Handford RP. 2007. The evolution of the vertebrate metzincins; insights from Ciona intestinalis and Danio rerio. BMC Evol Biol. 7:63.

Huxley AF, Niedergerke R. 1954. Structural changes in muscle during contraction; interference microscopy of living muscle fibres. Nature.173(4412): 971-973.

Huxley H, Hanson J. 1954. Changes in the cross-striations of muscle during contraction and stretch and their structural interpretation. Nature.173(4412): 973-976.

Jackson BC, Nebert DW, Vasiliou V. 2010. Update of human and mouse matrix metalloproteinase families. Hum Genomics. 4(3): 194–201.

Kessenbrock K, Plaks V, Werb Z. 2010. Matrix metalloproteinases: regulators of the tumor microenvironment. Cell. 141(1): 52-67.

Kim EM, Hwang O. 2011. Role of matrix metalloproteinase-3 in neurodegeneration. J Neurochem. 116(1): 22-32.

Klein T, Bischoff R. 2011. Physiology and pathophysiology of matrix metalloproteases. Amino Acids. 41(2): 271-290.

Kruger M, Linke WA. 2011. The Giant Protein Titin: a regulatory node that integrates myocyte signaling pathways. J Biol Chem. 286(12): 9905-9912.

Page 35: Good, better, best. Never let it rest. Until your good is ...

21

Kwan JA, Schulze CJ, Wang W, Leon H, Sariahmetoglu M, Sung M, Sawicka J, Sims DE, Sawicki G, Schulz R. 2004. Matrix metalloproteinase-2 (MMP-2) is present in the nucleus of cardiac myocytes and is capable of cleaving poly (ADP-ribose) polymerase (PARP) in vitro. FASEB J. 18(6): 690-2.

Lapière ChM. 2005. Tadpole collagenase, the single parent of such a large family. Biochimie. 87(3-4): 243–247.

Lieschke GJ, Currie PD. 2007. Animal models of human disease: zebrafish swim into view. Nat Rev Genet. 8(5): 353-67.

Limb GA, Matter K, Murphy G, Cambrey AD, Bishop PN, Morris GE, Khaw PT. 2005. Matrix metalloproteinase-1 associates with intracellular organelles and confers resistance to lamin A/C deg- radation during apoptosis. Am J Pathol. 166(5): 1555–1563.

Liu X. 2011. Emerging ideas: Matrix Metalloproteinase-2 in muscle atrophy. Clin Orthop Relat Res. 469(6): 1797–1799.

Loffek S, Schilling O, Franzke CW. 2011. Biological role of matrix metalloproteinases: a critical balance. Eur Respir J. 38(1): 191–208.

Lu P, Takai K, Weaver VM, Werb Z. 2011. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb Perspect Biol. 3(12): a005058.

Mannello F, Medda V. 2012. Nuclear localization of Matrix metalloproteinases. Prog Histochem Cytochem. 47(1): 27–58.

Moshal KS, Tipparaju SM, Vacek TP, Kumar M, Singh M, Frank IE, Patibandla PK, Tyagi N, Rai J, Metreveli N, et al. 2008. Mitochondrial matrix metalloproteinase activation decreases myocyte contractility in hyperhomocysteinemia. Am J Physiol Heart Circ Physiol. 295(2): 890–897.

Murphy G, Nagase H. 2008. Progress in matrix metalloproteinase research. Mol Aspects Med. 29(5): 290–308.

Murphy G, Nagase H. 2010. Localizing matrix metalloproteinase activities in the pericellular environment. FEBS J. 278(1): 2 –15.

Nagase H, Visse R, Murphy G. 2006. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res. 69(3): 562–573.

Okamoto T, Akaike T, Sawa T, Miyamoto Y, Van der Vliet A, Maeda H. 2001. Activation of matrix metalloproteinases by peroxynitrite-induced protein S-glutathiolation via disulfide S-oxide formation. J Biol Chem. 276 (31): 29596–602.

Page-McCaw A, Ewald AJ, Werb Z. 2007. Matrix metalloproteinases and the regulation

Page 36: Good, better, best. Never let it rest. Until your good is ...

22

of tissue remodeling. Nat Rev Mol Cell Biol. 8(3): 221-233.

Pei D, Weiss SJ. 1995. Furin-dependent intracellular activation of the human stromelysin-3 zymogen. Nature. 375(6528): 244-247.

Phatharajaree W, Phrommintikul A, Chattipakorn N. 2007. Matrix metalloproteinases and myocardial infarction. Can J Cardiol. 23(9): 727-733.

Sänger AM, Stoiber W. Muscle fibre diversity and plasticity. In: Johnston IA, editor. Muscle development and growth. San Diego (CA): Academic Press; 2001. p. 187-250. Sarantitis I, Papanastasopoulos P, Manousi M, Baikoussis NG, Apostolakis E. 2012. The cytoskeleton of the cardiac muscle cell. Hellenic J Cardiol. 53(5): 367-79.

Sariahmctoglu M, Crawford BD, Leon H, Sawicka J, Li L, Ballermann BJ, Holmes C, Berthiaume LG, Holt A, Sawicki G, et al. 2007. Regulation of matrix metalloproteinase-2 (MMP-2) activity by phosphorylation. FASEB J. 21(10): 2486-2495.

Sawicki G, Leon H, Sawicka J, Sariahmctoglu M, Schulze CJ, Scott PG, Szczesna-Cordary D, Schulz R. 2005. Degradation of myosin light chain in isolated rat hearts subjected to ischemia-reperfusion injury: a new intracellular target for matrix metalloproteinase-2. Circulation. 112(4): 544-552.

Schulz R. 2007. Intracellular targets of matrix metalloproteinase-2 in cardiac disease: rationale and therapeutic approaches. Annu Rev Pharmacol Toxicol. 47: 211–242.

Squire JM, Knupp C, Luther PK. 2008. Zebrafish topical, transparent, and tractable for ultrastructural studies. J Gen Physiol. 131(5): 439–443.

Sung MM, Schulz CG, Wang W, Sawicki G, Bautista-Lopez NL, Schulz R. 2007. Matrix metalloproteinase-2 degrades the cytoskeletal protein α- actinin in peroxynitrite mediated myocardial injury. J Mol Cell Cardiol. 43(4): 429-436.

Tallant C, Marrero A, Gomis-Rüth FX. 2010. Matrix metalloproteinases: fold and function of their catalytic domains. Biochim Biophys Acta. 1803(1): 20-8.

Vartak DG, Gemeinhart RA. 2007. Matrix metalloproteases: underutilized targets for drug delivery. J Drug Targeting. 15(1): 1-20.

Van-Wart HE, Birkedal-Hansen H. 1990. The cysteine switch: a principle of regulation of metalloproteinase activity with potential applicability to the entire matrix metalloproteinase gene family. Proc Natl Acad Sci USA. 87(14): 5578-5582.

Venkatasubramanian PN. Imaging the pancreatic ECM. In: Grippo PJ, Munshi HG, editors. Pancreatic cancer and tumor microenvironment. Trivandrum (India): Transworld Research Network; 2012. p. 11-27.

Page 37: Good, better, best. Never let it rest. Until your good is ...

23

Viappiani S, Nicolescu AC, Holt A, Sawicki G, Crawford BD, León H, van Mulligen T, Schulz R. 2009. Activation and modulation of 72kDa matrix metalloproteinase-2 by peroxynitrite and glutathione. Biochem Pharmacol. 77(5): 826–34.

Visse R, Nagase H. 2003. Matrix metalloproteinases and tissue inhibitors of metalloproteinases structure, function, and biochemistry. Circ Res. 92(8): 827-839.

Wang W, Schulze CJ, Suarez-Pinzon WL, Dyck JR, Sawicki G, Schulz R. 2002. Intracellular action of matrix metalloproteinase-2 accounts for acute myocardial ischemia and reperfusion injury. Circulation.106(12): 1543-1549.

Wyatt RA, Keow JY, Harris ND, Hache CA, Li DH, Crawford BD. 2009. The zebrafish embryo: a powerful model system for investigating matrix remodeling. Zebrafish. 6(4): 347-354.

Yalak G, Ehrlich YH, Olsen BR. 2014. Ecto-protein kinases and phosphatases: an emerging field for translational medicine. J Transl Med. 12:165.

Yang Y, Candelario-Jalil E, Thompson JF, Cuadrado E, Estrada EY, Rosell A, Montaner J, Rosenberg GA. 2010. Increased intranuclear matrix metalloproteinase activity in neurons interferes with oxidative DNA repair in focal cerebral ischemia. J Neurochem 112(1): 134–149.

Zhang J, Cao YJ, Zhao YG, Sang QX, Duan EK. 2002. Expression of matrix metalloproteinase-26 and tissue inhibitor of metalloproteinase-4 in human normal cytotrophoblast cells and a choriocarcinoma cell line, JEG-3. Mol Hum Reprod. 8(7): 659–666.

Page 38: Good, better, best. Never let it rest. Until your good is ...

24

Chapter 2: Intracellular localization of gelatinase-A (Mmp2) in zebrafish

skeletal muscle

2.1 Introduction

As discussed in the previous chapter, MMPs are known as secreted proteases, and

collectively, they degrade all of the proteinacious components of the ECM. However,

recent evidence has emerged that indicates some MMPs are not secreted from the cell,

and that these proteases may have significant roles in the pathology of

ischemia/reperfusion injury (reviewed in Chow et al. 2007). For instance, MMP-2 has

been detected intracellularly in rat cardiac myocytes, and is implicated in the degradation

of sarcomeric proteins under conditions of oxidative stress injury (Wang et al. 2002;

Sawicki et al. 2005; Sung et al. 2007; Ali et al. 2010; reviewed in Schulz 2007 and!Ali et

al. 2011a). One obvious question that arises from these observations is ‘how does this

secreted protein accumulate in the cytoplasm of myocytes?’ Gelatinase-A is a class-I

secreted protein, which means that its N-terminus consists of a signal sequence that is

bound by the signal recognition particle (SRP) as the nascent polypeptide emerges from

the ribosome. The SRP facilitates binding to the endoplasmic reticulum (ER)

translocation complex, such that the protein is translocated into the ER, and from there to

the Golgi apparatus and secretory pathway (Cooper and Hausman 2009, and discussed

further in Chapter 4). However, it has now become clear that the signal sequence of the

human MMP-2 protein - MEALMARGALTGPLRALCLLGCLLSHAAA – is not bound

efficiently by the SRP, and consequently approximately 40% of the MMP-2 protein

produced in cells remains cytosolic (Ali et al. 2011b). Furthermore, in human cardiac

Page 39: Good, better, best. Never let it rest. Until your good is ...

25

myocytes, a splice variant of mmp-2 is expressed in which an alternative first exon

encodes a slightly truncated form of the protein lacking any secretory signal (Ali et al.

2011b).

Intuitively, it would seem unlikely that cardiac myocytes accumulate intracellular

stores of dangerous proteases simply to make themselves more vulnerable to oxidative

stress. If this pathological activity of gelatinase-A were not balanced by some valuable

physiological role, one would expect natural selection to have driven the evolution of a

more efficient secretory signal for this protease, as is found in other secreted proteins.

Furthermore, MMP-2 is phosphorylated in vivo, and its proteolytic activity is modulated

by this post-translational modification (Sariahmetoglu et al. 2007). This implies the

existence of kinases and phosphatases that regulate the activity of this intracellular pool

of MMP-2, and furthermore that such regulatory pathways likely evolved to modulate a

normal, physiological role for this protease. What might be the function of this

intracellular gelatinase-A?

In order to address these questions, I used zebrafish to determine if the intracellular

localization of gelatinase-A is unique to mammalian cardiac myocytes, or alternatively, if

it may be a common feature of striated muscle, implying a more ancient and

evolutionarily-conserved function for this protease in muscle physiology. The zebrafish is

an excellent vertebrate model for the study of ultrastructure and muscle physiology

(Squire et al. 2008), and this is enhanced by the availability of anti-zebrafish Mmp2

antibody (Harris 2010). In zebrafish mmp2 expression at the mRNA level is essentially

constant and homogeneous in all tissues throughout development (Zhang et al. 2003).

Page 40: Good, better, best. Never let it rest. Until your good is ...

26

However, immunoblots of whole embryo homogenates reveal Mmp2 protein

accumulation beginning during gastrulation, and immunostaining reveals the protein

accumulates heterogeneously, with significant concentrations occurring along motor

axons, in the developing central nervous system, at myotome boundaries, within the

epidermal folds of developing fins, and within the muscle tissue itself (Lévesque 2010).

Detailed examination of the distribution of Mmp2 protein in the skeletal muscle of the

trunk reveals a distinctly striated pattern, consistent with sarcomeric localization of the

protease (Figure 2.1) (Lévesque 2010).

In order to determine if this protein is actually located within the sarcomere of

zebrafish muscle, I used double immunostaining of tissues from embryos and adult fish,

using anti-Mmp2 to characterize its distribution relative to α–actinin, a sarcomeric

protein known to be a component of the Z-disc (reviewed in Luther 2009). These data

suggest that Mmp2 accumulates in M-bands within the sarcomere of zebrafish skeletal

muscle. However, because the axial resolution of confocal microscopy (Murray 2006)

and the thickness of myofibrils (reviewed in Recher et al. 2009) are nearly the same (~1

µm) it is impossible to distinguish periodic cell surface staining from periodic

intracellular staining within the sarcomeres. Thus my interpretation of immunostaining

results could be contested. By using cryo-sectioning to cut the muscle samples at around

a half micron in thickness, I was able to prove the intracellular localization of Mmp2 in

zebrafish skeletal muscle. Furthermore, using a bioinformatic approach, I show that the

poorly-recognized secretory signal described for human MMP-2 is typical of gelatinase-

A orthologues across a phylogenetically diverse sample of vertebrates. This is consistent

Page 41: Good, better, best. Never let it rest. Until your good is ...

27

Figure 2.1: Mmp2 is recognized on the Z-discs of zebrafish sarcomeres. A) Mmp2 protein is heterogeneously distributed in the 72 hpf embryos. It accumulates in brain, motor neurons, striated muscle, and neural crest-derived matrix rich structures. B) The expression of Mmp2 protein in the striated muscle. C) The sarcomeric localization of Mmp2 protein in skeletal muscle. (From Lévesque 2010).

Muscle

Brain

Motor neurons Neural crest

A)

Page 42: Good, better, best. Never let it rest. Until your good is ...

28

with the existence of an unknown but selectively advantageous function for gelatinase-A

within the sarcomeres of striated muscle.

Despite the fact that the expression of Mmp2 is very high in skeletal muscle, the

role(s) of intracellular Mmp2 in zebrafish skeletal muscle remains unknown. I speculated

that Mmp2 may play a role in sarcomere development, which occurs in an anterior-to-

posterior wave in the 24 hpf embryo (Kimmel et al. 1995), but immunostaining shows

Mmp2 does not accumulate in developing myofibrils until after sarcomere establishment

is complete, suggesting a role in maintenance, rather than formation of the contractile

apparatus.

Page 43: Good, better, best. Never let it rest. Until your good is ...

29

2.2 Materials and methods

2.2.1 Spawning zebrafish and collecting embryos

Wild-type adult zebrafish were maintained on a 14-hour light and 10-hour dark

cycle as described in Westerfield (1995) and fed brine shrimp twice a day and fish pellets

(Adult zebrafish diet, Zeigler) three to four times a day. The embryos were collected after

natural spawning over trays filled with marbles. Embryos were grown in embryo rearing

medium (ERM) (13 mM NaCl, 0.5 mM KCl, 0.02 mM Na2HPO4, 0.04 mM KH2PO4, 1.3

mM CaCl2, 1.0 mM MgSO4, and 4.2 mM NaHCO3, pH 7.4) at 28.5 °C. 0.001-0.002%

methylene blue was added to minimize fungal growth. For pre-hatching stages, embryos

were manually dechronionated using fine forceps under the microscope prior to fixation.

2.2.2 Immunostaining and confocal microscopy

To prepare for immunostaining, 10 embryos at 24 and 72 hour post fertilization

(hpf) were collected, dechorionated, and chilled. Three adult zebrafish were terminally

anaesthetized in 0.3 mg/ml tricaine methanesulfonate (Sigma) in ERM, after which the

fish tail muscles were dissected from the fish using forceps. Subsequently, embryos and

adult muscle tissue samples were fixed in Dent’s solution (20% dimethyl sulfoxide

(DMSO), 80% methanol) overnight at 4°C. Samples were washed with PBSTx (0.1%

Triton X-100 in PBS (20 mM phosphate pH 7.3, 137 mM NaCl, 2.7 mM KCl)) five times

for five minutes to remove fixative, blocked in blocking buffer (5% bovine serum

albumin (BSA) in PBSTx) overnight at 4°C, and incubated with primary antibodies,

rabbit anti-MMP2 (Anaspec catalogue #55111) and mouse anti α-actinin (Sigma,

catalogue #A7811), diluted (1:1000) in blocking buffer overnight at 4°C. Samples were

Page 44: Good, better, best. Never let it rest. Until your good is ...

30

washed another five times for five minutes with PBSTx and then incubated with

fluorescent conjugated secondary antibodies, goat anti-rabbit Alexa-488 and goat anti-

mouse Alexa-633 (Invitrogen) diluted (1:1000) in blocking buffer overnight at 4°C. After

the final incubation, they were again washed with PBSTx, five times for five minutes

each, and imaged using a Leica SP2 laser scanning confocal microscope with 63x 1.4 NA

lens.

2.2.3 Cryo-sectioning

A dozen 72 hpf zebrafish embryos were immunostained as described above and

then washed in PBSTx and embedded in 2.3 M sucrose dissolved in PBS overnight at 4

°C. The following day, the embedded embryos were frozen with liquid nitrogen and cut

into 500 nm ultrathin sections using a Leica Ultracut T ultramicrotome. Sections were

mounted on poly-L-Lysine coated glass slides and imaged as described above.

2.2.4 Prediction the signal sequence cleavage sites in Gelatinase-A, Gelatinase-B and

BiP (Binding immunoglobulin protein) using SignalP

SignalP is a program that uses a neural network that has been trained using known

secreted and non-secreted proteins. Analysis of eukaryotic protein sequences using

SignalP generates a high discrimination score (D-score) (i.e., above 0.450) for proteins

that are secreted, allowing SignalP to identify secreted proteins with 90-91% accuracy

(Klee and Ellis 2005).

The sequences of gelatinase-A, gelatinase-B, and BiP orthologues from 73

vertebrates were selected from the PubMed protein database

(http:/www.ncbi.nlm.nih.gov/protein/), on the basis of the existence of high quality

Page 45: Good, better, best. Never let it rest. Until your good is ...

31

sequence data for all three proteins for each organism. SignalP 4.1 software (Nielsen et

al., 1997) (http://www.cbs.dtu.dk/services/SignalP/) was used to score the probability of

secretion for each sequence (See appendix A).

2.2.5 Statistical analysis

The Mann-Whitney test (for data that are not normally distributed) was performed

using http://www.socscistatistics.com/tests/mannwhitney/, to determine if there were

significant differences among the D scores generated by SignalP for each of the proteins.

Significance was determined at the p < 0.01 level. Data are expressed as mean ± standard

error of the mean (S.E.M).

Page 46: Good, better, best. Never let it rest. Until your good is ...

32

2.3 Results

2.3.1 Mmp2 in the skeletal muscle of adult and embryonic zebrafish

Mmp2 appears to be present in the M-bands of skeletal muscle sarcomeres (i.e.,

between α-actinin containing the Z-discs) of 72 hpf embryos (n = 10) as well as in the

adult zebrafish (n = 3) (Figure 2.2). However, because the axial resolution of the optical

system used to obtain these images (800 nm) is very close to the diameter of a myofibril

(~1 µm), it is possible that the apparent localization of Mmp2 between α-actinin positive

Z-discs is an illusion arising from Mmp2 localized circumferentially at the surface of the

cells, surrounding the M-bands. To eliminate this possibility, I examined physical

sections in addition to optical sections.

2.3.2 Intracellular localization of Mmp2 in the sarcomeres of zebrafish skeletal muscle

I initially attempted to address this question using transmission electron microscopy

and immunogold labeling of thin sections of muscle embedded in London Resin, but

unfortunately the epitope recognized by the anti-Mmp2 antibody used here is not

preserved by aldehyde fixatives (see Appendix B). However, ultrathin cryo-sections of 72

hpf zebrafish muscle fixed in Dent’s (a denaturing fixative) probed with the same

primary and secondary antibodies revealed a strong and distinctive concentration of

Mmp2 on the M-bands of sarcomeres in skeletal muscle (Figure 2.3) Because the

physical thickness of the cryosections (500 nm) imaged in Figure 2.3 is significantly less

than the thickness of myofibrils (~1 µm), if the Mmp2 immunoreactivity were restricted

to the surface of the cells, it would only appear between α-actinin positive Z-discs when

the section captured both the surface of the cells and the underlying sarcomeric structure.

Furthermore, periodic surface staining would be expected to frequently produce partial

Page 47: Good, better, best. Never let it rest. Until your good is ...

33

bands of Mmp2 labeling, in sections that took a chord through the roughly cylindrical

fibril, and thereby included oblique sections of the sarcolemma. Neither of these effects

are observed; robust Mmp2-immunoreactive bands are consistently complete and

positioned precisely between α-actinin positive Z-discs in these sections, showing

unequivocally that the Mmp2 is inside the cells, and positioned within the M-bands of the

sarcomeres.

2.3.3 Ontogeny of MMP-2 in zebrafish sarcomeres

The skeletal muscle of zebrafish embryos differentiates from mesodermally derived

somites that are specified and differentiate sequentially along the anterior-posterior axis

(reviewed in Goody et al. 2015). Thus, in a 24 hpf embryo, anterior somites have

differentiated as functional, contractile skeletal muscle, while the most posterior somites

have yet to be specified. Thus, examining presumptive skeletal muscle cells in a posterior

to anterior direction in a single embryo is like looking at a time lapse series illustrating

muscle cell specification and differentiation. I took advantage of this feature of zebrafish

development to determine which comes first, α-actinin-positive Z-discs or Mmp2-

positive M-bands. In every embryo examined (n = 10), α-actinin-positive Z-discs were

established before (i.e., posteriorly to) Mmp2-positive M-bands (Figure 2.4).

2.3.4 Gelatinase-A secretory signals are poorly recognized

Although SignalP software was not developed explicitly to determine the efficiency

with which a protein will be secreted, the discrimination score (D-score) SignalP

generates for a protein is expected to correlate with this (H. Nielsen, personal

Page 48: Good, better, best. Never let it rest. Until your good is ...

34

communication).

I assembled a database of all gelatinase-A, gelatinase-B and BiP homologues from

vertebrate species for which all three were available as complete sequences, and

subjected them to analysis using SignalP (Appendix A). In this analysis, BiP is a positive

control, in that it is a well-known ER-resident protein with a strong signal sequence.

Gelatinase-B (MMP-9) was also included in this analysis, as a closely related MMP to

gelatinase-A.

Gelatinase-A homologues have a mean D-score of 0.651 (± 0.273) (Figure 2.5).

The default SignalP threshold above which a protein is considered ‘secreted’ is 0.450,

making Mmp2 homologues clearly identifiable as secreted proteins as expected.

However, both gelatinase-B and BiP had much higher mean D-scores (0.801 ± 0.170 and

0.854 ± 0.164, respectively; Figure 2.5), consistent with the hypothesis that the inefficient

secretion of gelatinase-A homologues observed in mammals and zebrafish may be a

conserved feature of this protein. Because the D-scores generated for these sequences

were not normally distributed, I used a Mann-Whitney U-test to determine if the

differences in the scores were statistically significant, and found that the D-scores of

gelatinase-A was significantly lower than the D-scores of gelatinase-B and BiP (p <

0.01).

Page 49: Good, better, best. Never let it rest. Until your good is ...

35

Figure 2.2: Apparent Mmp2 localization at M-bands in zebrafish muscles. The longitudinal images of 72 hpf embryo (left) and adult (right) zebrafish skeletal musculature showing the distribution of Mmp2 protein (green) with respect to α–actinin (red) imaged by confocal microscopy.

Figure 2.3: Evidence of M-band localization of Mmp2 within 72 hpf zebrafish sarcomeres. Representative image of ultra-thin (500 nm) cryosection of 72 hpf zebrafish skeletal muscle with Mmp2 (green) clearly localized precisely between the Z-discs that contain α-actinin (red).

Page 50: Good, better, best. Never let it rest. Until your good is ...

36

Figure 2.4: The ontogeny of Mmp2 in zebrafish myogenesis. Mmp2 immunostaining in newly formed myofibrils in the trunk of 24 hpf embryos reveals that sarcomeric localization of Mmp2 occurs only after Z-discs have become established. Distinct α-actinin-positive Z-disks (red) invariably appear before (i.e. posteriorly to) Mmp2-positive (green) M-bands.

Page 51: Good, better, best. Never let it rest. Until your good is ...

37

Figure 2.5: Comparison of SignalP predictions in gelatinase-A (MMP-2), gelatinase-B (MMP-9), and Binding immunoglobulin protein (BiP) protein secretions. Summary data for all homologues available for each sequence showing the average "Discrimination score" indicating the likelihood that each protein is secreted. Gelatinase-A homologues have a significantly lower D-scores compared to gelatinase-B and BiP proteins. Error bars represent standard deviations of the means. In this comparison, ‘a’ indicates statistical significance at p < 0.01 vs. ‘b’ (MMP-9 and BiP).

0

0.2

0.4

0.6

0.8

1

1.2

BiP MMP-9 MMP-2

b b a

Mea

n Si

gnal

P Sc

ores

Page 52: Good, better, best. Never let it rest. Until your good is ...

38

2.4 Discussion

In this chapter I show by using immunofluorescence, cryosectioning, and confocal

microscopy that Mmp2 has distinct intracellular localization in skeletal muscle in

zebrafish. Furthermore, the poorly recognized secretory signal characterized in human

MMP-2 appears typical of gelatinase-A orthologues, suggesting a conserved function of

this protease within the sarcomeres of vertebrate striated muscle.

I found Mmp2 accumulated on the M-bands of zebrafish skeletal muscle sarcomeres.

Interestingly, Mmp2 is not detected in Z-discs in zebrafish muscle, in contrast to what has

been reported regarding mammalian MMP-2 (Wang et al. 2002; Sawicki et al. 2005; Ali

et al. 2010). By using immunohistochemistry, Ali and co workers showed strong co-

localization of MMP2 with the T12 anti-titin antibody, which binds close to the Z-disc in

human and rat cardiomyocytes (Ali et al. 2010). On the other hand, the immunoreactivity

of MMP2 did not co-localize with the M8 anti-titin antibody, which targets the M-bands

(reviewed in Müller et al. 2012). Thus, while it is clear that in zebrafish Mmp2 is present

within the sarcomere of skeletal muscle, its distribution within the contractile apparatus

appears to be different than that in mammalian cardiomyocytes. This difference may have

important functional implications, and is worthy of further investigation.

The sarcomeric localization of Mmp2 in zebrafish skeletal muscle occurs

subsequently to the establishment of Z-discs, and therefore, while I cannot rule out a

developmental function for this protease in the development of the contractile apparatus,

it seems more likely that Mmp2 functions in the maintenance of the sarcomere.

Page 53: Good, better, best. Never let it rest. Until your good is ...

39

Others have shown empirically that mammalian MMP-2 is inefficiently secreted (Ali

et al. 2011b), and my analysis of signal sequences in all vertebrate gelatinase-A

homologues for which sequence data are available is consistent with this. This

conservation of inefficient signal sequences suggests a selective pressure to maintain an

intracellular pool of this protease. This intracellular pool is clearly involved in

pathological activities under conditions of oxidative stress (reviewed in Kandasamy et al.

2010; Ali et al. 2011a), but its physiological function remains obscure.

Page 54: Good, better, best. Never let it rest. Until your good is ...

40

2.5 References

Ali MA, Cho WJ, Hudson B, Kassiri Z, Granzier H, Schulz R. 2010. Titin is a target of matrix metalloproteinase-2: implications in myocardial ischemia/reperfusion injury. Circulation. 122(20): 2039-47.

Ali MA, Fan X, Schulz R. 2011a. Cardiac sarcomeric proteins: novel intracellular targets of matrixmetalloproteinase-2 in heart disease. Trends Cardiovasc Med. 21(4): 112-118.

Ali MA, Chow AK, Kandasamy AD, Fan X, West LJ, Crawford BD, Simmen T, Schulz R. 2011b. Mechanisms of cytosolic targeting of matrix metalloproteinase-2. J Cell Physiol. 227: 3397-3404. !

Chow AK, Cena J, Schulz R. 2007. Acute actions and novel targets of matrix metalloproteinases in the heart and vasculature. Br J Pharmacol. 152(2): 189–205.

Cooper GM, Hausman RE. 2009. The cell: a molecular approach. Fifth edition. Washington (DC): The American Society of Microbiology Press. p. 383-432.

Goody MF, Sher RB, Henry CA. 2015. Hanging on for the ride: adhesion to the extracellular matrix mediates cellular responses in skeletal muscle morphogenesis and disease. Dev Biol.401(1): 75–91.

Harris ND. 2010. Molecular ontogeny of extracellular matrix remodeling proteases during development of the zebrafish, and the effects of simulated micro-gravity on their expression. MSc thesis. University of New Brunswick [Fredericton (NB)].

Kandasamy AD, Chow AK, Ali MA, Schulz R. 2010. Matrix metalloproteinase-2 and myocardial oxidative stress injury: beyond the matrix. Cardiovasc Res. 85(3): 413–423.

Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, Schilling TF. 1995. Stages of embryonic development of the zebrafish. Dev Dyn. 203(3): 253-310.

Klee EW, Ellis LB. 2005. Evaluating eukaryotic secreted protein prediction. BMC Bioinf. 14(6): 256.

Lévesque JM. 2010. The localization of MMP2 in zebrafish embryos implies unexpected roles in neurogenesis and muscle function. Honours thesis. University of New Brunswick [Fredericton (NB)].

Luther PK. 2009. The vertebrate muscle Z-disc: sarcomere anchor for structure and signalling. J Muscle Res Cell Motil. 30(5-6): 171-85.

Müller AL, Hryshko LV, Dhalla NS. 2012. Extracellular and intracellular proteases in cardiac dysfunction due to ischemia–reperfusion injury. Int J Cardiol. 164(1): 39-47.

Murray JM. Confocal microscopy, deconvolution, and structured illumination methods.

Page 55: Good, better, best. Never let it rest. Until your good is ...

41

In: Spector DL, Goldman RD, editors. Basic methods in microscopy, protocols and concepts from cell: a laboratory manual. New York (NY): Cold Spring Harbor Laboratory Press; 2006. p. 43-81.

Nielsen H, Engelbrecht J, Brunak S, von Heijne G.1997. Identification of prokaryotic and eukaryotic signal peptides and prediction of their cleavage sites. Protein Eng. 10(1): 1-6.

Recher G, Rouede D, Richard P, Simon A, Bellanger JJ, Tiaho F. 2009. Three distinct sarcomeric patterns of skeletal muscle revealed by SHG and TPEF microscopy. Opt Express.17(22): 19763-77.

Sariahmetoglu M, Crawford BD, Leon H, Sawicka J, Li L, Ballermann BJ, Holmes C, Berthiaume LG, Holt A, Sawicki G, et al. 2007. Regulation of matrix metalloproteinase-2 (MMP-2) activity by phosphorylation. FASEB J. 21(10): 2486-2495.

Sawicki G, Leon H, Sawicka J, Sariahmctoglu M, Schulze CJ, Scott PG, Szczesna-Cordary D, Schulz R. 2005. Degradation of myosin light chain in isolated rat hearts subjected to ischemia-reperfusion injury: a new intracellular target for matrix metalloproteinase-2. Circulation. 112(4): 544-552.

Schulz R. 2007. Intracellular targets of matrix metalloproteinase-2 in cardiac disease: rationale and therapeutic approaches. Annu Rev Pharmacol Toxicol. 47: 211–242.!

Squire JM, Knupp C, Luther PK. 2008. Zebrafish topical, transparent, and tractable for ultrastructural studies. J Gen Physiol. 131(5): 439–443.

Sung MM, Schulz CG, Wang W, Sawicki G, Bautista-Lopez NL, Schulz R. 2007. Matrix metalloproteinase-2 degrades the cytoskeletal protein α- actinin in peroxynitrite mediated myocardial injury. J Mol Cell Cardiol. 43(4): 429-436.

Wang W, Schulze CJ, Suarez-Pinzon WL, Dyck JR, Sawicki G, Schulz R. 2002. Intracellular action of matrix metalloproteinase-2 accounts for acute myocardial ischemia and reperfusion injury. Circulation.106(12): 1543-1549.

Westerfield M. 1995. The Zebrafish Book: A guide for the laboratory use of zebrafish (Danio rerio). Eugene (OR): University of Oregon Press.

Zhang J, Bai S, Zhang X, Nagase H, Sarras MP. 2003. The expression of gelatinase A (MMP-2) is required for normal development of zebrafish embryos. Dev Genes Evol. 231(9): 456–463.

Page 56: Good, better, best. Never let it rest. Until your good is ...

42

Chapter 3: Phosphorylation status of Mmp2 in zebrafish myocytes

3.1 Introduction

After 40 years of study MMPs were viewed as functioning exclusively as extracellular

proteases (reviewed in Brinckerhoff and Matrisian 2002). However, in the last decade

and a half, work primarily from the Schulz lab at the University of Alberta revealed the

unexpected presence of MMP2 within cardiomyocytes of mammals (Wang et al. 2002;

Sawicki et al. 2005), and implicated it in the pathology of ischemia/reperfusion injury

(IRI) (reviewed in Schulz 2007). Other labs have now demonstrated the presence of

MMP2 within the skeletal muscle fibres of mouse (Hadler-Olsen et al. 2014) as well as

within neurons (Yang et al. 2010) and hepatocytes (Kwan et al. 2004). In 2007,

Sariahmetoglu and her colleagues reported that human MMP-2 is phosphorylated at

several serine and threonine residues in vivo, and that this post-translational modification

dramatically changes its enzymatic activity (Sariahmetoglu et al. 2007; reviewed in

Schulz 2007). Regulation by phosphorylation is generally a characteristic of intracellular

proteins, although there are exceptions (Yalak et al. 2014). Moreover, the specificity of

protein kinases suggests that this may be an evolutionarily conserved regulatory

mechanism.

Subsequently, several studies addressed the roles of this post-transitional modification

on the structure and activity of gelatinase-A in the presence of peroxynitrite (ONOO-).

Sariahmetoglu and her colleagues (2012) reported the importance of phosphorylation

status of MMP2 in myocardial injury. They noticed that increasing the proportion of

Page 57: Good, better, best. Never let it rest. Until your good is ...

43

phosphorylated (inactive) MMP2 using phosphatase inhibitors helps protect the

contractile proteins from degradation through the deleterious action of MMP2 activated

by ONOO- (Sariahmetoglu et al. 2012; reviewed in Sawicki 2013). Moreover, the

combined effect of both ONOO- and dephosphorylation of MMP2 generated a greater

increase in proteolytic activity than ONOO- alone (Jacob-Ferreira et al. 2013), suggesting

that mis-regulation of MMP2 phosphorylation may be more important in the pathology of

IRI than the direct effect of activation by reactive oxygen species (ROS).

Protein kinases are one of the biggest families of eukaryotic genes (reviewed in

Ubersax and Ferrell 2007). They can be roughly divided into two major classes: tyrosine

kinases and serine/threonine kinases. Tyrosine, serine and threonine are the only amino

acids that are normally phosphorylated by kinases in proteins. Kinases catalyze the

nucleophilic attack of the deprotonated hydroxyl group in the amino acid side chain on

the gamma phosphate of an adenosine triphosphate (ATP) molecule, yielding adenosine

diphosphate and a phosphorylated protein. Conversely, phosphatases remove a phosphate

group from phosphoproteins, yielding inorganic phosphate (Pi) and a dephosphorylated

protein (reviewed in Cheng et al. 2011).

Although the post-translational modification by phosphorylation is generally

associated with regulation of intracellular proteins, a variety of proteins in the

extracellular milieu appeared to be phosphorylated by exokinases (Yalak et al. 2014). For

example, Fam20C is a kinase that resides in the Golgi apparatus and is responsible for the

phosphorylation of proteins destined for secretion, including caseins and bone proteins

that are important in biomineralization. Fam20C phosphorylates the serine residue within

Page 58: Good, better, best. Never let it rest. Until your good is ...

44

S-x-E motifs (where x is any amino acid) (Tagliabracci et al. 2012).

The finding that human gelatinase-A (MMP-2) is phosphorylated in living cells, and

that this phosphorylation changes its enzymatic activity, made me want to explore

whether this mechanism applies outside of mammalian cells, and therefore may represent

an ancient regulatory mechanism. The presence and activity of such a system may shed

some light on the normal physiological roles of MMP-2 inside cells, and may also

indicate possible strategies for reducing undesirable MMP-2 activity associated with

pathology.

Here I present an approach for the detection of phospho-Mmp2 in tissue samples using

homogenized muscle cells of adult zebrafish. This approach involves affinity

chromatography, in which gelatin sepharose is bound by the fibronectin type II repeats of

the collagen binding domain of Mmp2 (zebrafish gelatinase-A) and Mmp9 (zebrafish

gelatinase-B). I used Pro-Q Diamond stain that is specific for phosphoproteins on SDS-

PAGE gels. By comparing the fluorescent intensity of protein bands stained with Pro-Q-

Diamond to the intensity obtained using SyproRuby, which is a general protein stain, and

then comparing that to a series of known standards, it is possible to quantitatively

determine the proportion of any given protein that is phosphorylated in a standard SDS-

PAGE gel (reviewed in Delom and Chevet 2006). Unfortunately, technical limitations

prevented me from purifying sufficient Mmp2 from zebrafish muscle to use this

approach. I therefore turned to a bioinformatic analysis of the conservation of predicted

phosphorylation sites in the gelatinase-A sequences across as many species as possible, in

order to look for evidence that phosphorylation of this protease may be significant in its

Page 59: Good, better, best. Never let it rest. Until your good is ...

45

physiological regulation.

NetPhos is an artificial neural network that predicts potential phosphorylation sites by

examining the sequences flanking empirically demonstrated sites of phosphorylation, and

using a heuristic based on these, determines the probability that a given serine, threonine

or tyrosine in an unknown sequence will be phosphorylated (Blom et al. 1999). I

collected and made an alignment of all available gelatinase-A sequences, and analyzed

the conservation of motifs that were identified as high-probability sites of

phosphorylation by using NetPhos. While I was unable to empirically verify Mmp2

phosphorylation in zebrafish, I determined that it is highly probable that zebrafish Mmp2

is subject to phosphorylation and that this post-translational modification is important in

regulating gelatinase-A activity throughout the animal kingdom.

Page 60: Good, better, best. Never let it rest. Until your good is ...

46

3.2 Materials and methods

3.2.1 Tissue preparation

In the pilot attempt of the gelatin affinity chromatography preparation, six adult

zebrafish were terminally anesthetized in 0.3 mg/ml tricaine methanesulfonate (Sigma)

solution for 10 minutes. The tail muscles were dissected from the fish, frozen in liquid

nitrogen and crushed with a cell crusher (Stratech Scientific Limited). Following several

attempts working with this method, I increased the number of fish in order to increase the

amount of muscle homogenate, using 57 adult zebrafish in the final chromatography

experiment.

3.2.2 Isolation of Gelatinases using gelatin-affinity chromatography

Protein was purified by pulverizing tail muscle dissected from adult zebrafish after

freezing in liquid nitrogen. The frozen pulverized muscle was transferred to cold

homogenization buffer (150 mM NaCl, 50 mM Tris pH 7.5, 0.1% Brij-35, 1% NP-40,

0.02 M sodium deoxycholate, 5 mM EDTA, 1x protease inhibitor cocktail), vortexed and

placed on ice. Homogenized muscle was centrifuged at 16,000x g for 25 minutes at 4°C

to pellet the insoluble debris. The resulting supernatant was collected and stored at -20°C

for later use. Gelatin affinity columns were prepared using either commercially available

gelatin sepharose 4B (GE Healthcare Bio-Sciences AB, Bjorkgatan, Sweden) or gelatin

affinity media prepared using cyanogen bromide activated sepharose 4B (Sigma), with a

final bed volume of 25 ml. For the cyanogen bromide preparation, 1 g of dry resin was

washed with 100 ml of 1 mM HCl at room temperature for 10 minutes four times and left

Page 61: Good, better, best. Never let it rest. Until your good is ...

47

for five minutes to allow the resin to settle down in order to pour off the supernatant. The

resin was washed with distilled H2O and then with two volumes of coupling buffer (0.5

M NaCl, 0.1 M NaHCO3, pH 8.5). While preparing the resin, 10 mg/ml of ligand gelatin

protein (Fisher) was dissolved in coupling buffer, with gentle warming. The washing

coupling buffer was poured off and ligand solution was added immediately, and

incubated overnight at 4°C with gentle agitation. Supernatant was removed, the resin

washed with 100 ml of coupling buffer for 10 minutes and left to settle. Buffer was

poured off and the unreacted sites were blocked by incubating with 0.2 M glycine in

coupling buffer overnight at 4°C with gentle agitation. Finally, the prepared resin was

poured into the column and washed extensively, alternating with two column volumes of

acetate buffer (0.5 M NaCl, 0.1 M sodium acetate pH 4), and two column volumes of

coupling buffer, five times. The resin was equilibrated with binding buffer before use, as

described below.

Prior to loading in the column, the thawed muscle homogenate was centrifuged at

16,000x g for 25 minutes at 4°C in order to pellet any precipitate. Gelatin sepharose

medium was equilibrated with at least 5 column volumes of binding buffer (150 mM

NaCl, 50 mM Tris pH 7.5, 0.1% Brij-35, 5 mM EDTA), and roughly 170 ml of muscle

homogenate was allowed to run into the column at 2 ml per minute. Unbound protein was

washed from the column using 10 column volumes of binding buffer at 2 ml per minute.

Finally, bound protein was eluted with elution buffer (4M urea, 50 mM Tris pH 7.5, 5

mM EDTA, and 0.1% Brij-35). Roughly 30 ml of eluted fractions were concentrated

down to 200 µl using an Amicon Ultra 4 mL Filter (Millipore) centrifuged at 894.4x g at

4°C.

Page 62: Good, better, best. Never let it rest. Until your good is ...

48

3.2.3 SDS-polyacrylaminde gel electrophoresis and phospho-protein identification

Samples of total homogenate and gelatin-affinity purified proteins were diluted 1:1 in

2x reducing sample buffer (250 mM Tris-HCl pH 6.8, 4% sodium dodecyl sulfate, 0.2%

bromophenol blue, 20% glycerol, 200 mM β-mercaptoethanol). 70 µl of each sample, as

well as 6 µl of EZ-run (Thermo) protein molecular weight standards, were resolved on a

10% SDS-PAGE gel at 50V for four hours. The gel was fixed in 100 ml of 50% methanol

and 10% acetic acid for 30 minutes, twice, and washed three times with 100 ml of

deionized water for 10 minutes. The gel was incubated in 60 ml of Pro-Q Diamond

Phosphoprotein Gel Stain solution (Invitrogen) for 60-90 minutes according to the

manufacturer’s instructions. After incubation, the gel was destained three times in 100 ml

of 20% acetonitrile and 50 mM sodium acetate, pH 4, for 30 minutes. Finally, the gel was

washed twice with 100 ml of deionized water for five minutes, and imaged using the

ChemiDoc MP imaging system.

After documenting the abundance of phosphoproteins, the gel was re-stained in 60 ml

of Sypro Ruby (Invitrogen) gel stain overnight for total protein detection. After washing

in 10% methanol and 7% acetic acid for 30 minutes the gel was re-imaged with the

ChemiDoc MP imaging system, and intensity quantified using Image Lab version 4.0.1

(Bio-Rad).

3.2.4 Immunoblots

Purified protein samples (from 3.2.2) were diluted 1:1 in 2x reducing sample buffer

(250 mM Tris-HCl pH6.8, 4% sodium dodecyl sulfate, 0.2% bromophenol blue, 20%

glycerol, 200 mM β-mercaptoethanol) and separated using SDS-PAGE on 10% gels, as

Page 63: Good, better, best. Never let it rest. Until your good is ...

49

described in 3.2.3. The proteins were transferred to polyvinylidene difluoride (PVDF)

membranes at 50 volts for four hours in Tobin buffer (25 mM Tris base, 192 mM glycine

and 20% methanol, pH 8.3). The membrane’s remaining protein-binding capacity was

saturated by incubating overnight in blocking buffer (5% skim milk powder in PBSTw

(0.1% Tween-20 in PBS (20 mM phosphate pH 7.3, 137 mM NaCl, 2.7 mM KCl)) at 4°C

with gentle agitation. The blocked membrane was incubated with primary antibody

(rabbit anti-Mmp2) (Anaspec catalogue #55111) diluted (1:1000) in blocking buffer and

gently agitated overnight at 4°C. The membrane was then washed with PBSTw three

times for five minutes each time. The membrane was re-incubated in horse-radish

peroxidase (HRP) conjugated goat anti-rabbit secondary antibody diluted in blocking

buffer (1:1000) overnight at 4°C with constant agitation. After washing the membrane

three times for five minutes each time with PBSTw, 2 ml of enhanced

chemiluminescence (ECL plus) (GE Healthcare Life Sciences) was applied to the

membrane and luminescent bands imaged using the ChemiDoc MP imaging system

3.2.5 Gelatin zymography

Samples of muscle homogenate and gelatin affinity purified proteins were diluted 1:1

in 2x non-reducing SDS-PAGE sample buffer (250 mM Tris-HCl pH 6.8, 4% sodium

dodecyl sulfate, 0.2% bromophenol blue, 20% glycerol) and resolved on a 10%

acrylamide zymogram containing 4 µg/ml gelatin (Fisher) at 50 V for four hours. The

zymogram was washed twice for 30 minutes at room temperature in 1x zymogram

renaturing buffer (2.5% Triton X-100 in 50 mM TrisHCl pH 7.5). The renaturing buffer

was removed and replaced with 1x zymogram developing buffer (50 mM TrisHCl pH

Page 64: Good, better, best. Never let it rest. Until your good is ...

50

7.5, 5 mM CaCl2, 1 mM ZnSO4, 2.5% Triton X-100) and gently agitated for 30 minutes

at room temperature. This buffer was replaced with a fresh solution of 1x zymogram

developing buffer and incubated for 48 h at 28°C. The gel was then stained in 0.5%

Coomassie brilliant blue dye G-250, dissolved in 25% methanol and 10% acetic acid at

room temperature overnight. Destaining was conducted the following day using a

solution of methanol, acetic acid, and water (40:10:50). Images of the gelatinolytic

activity detected as clear bands against the Coomassie brilliant blue stained background

were taken using the ChemiDoc MP imaging system.

3.2.6 Silver staining

Samples of whole muscle homogenate and affinity purified eluate in 2x reducing

sample buffer, as well 6 µl of Precision Plus Protein All Blue standards (BioRad), were

resolved on a 10% SDS-PAGE gel as described above. The gel was fixed in a solution of

50% methanol, 12% glacial acetic acid, 38% water with 200 µl of 37% formaldehyde,

with heating for one minute in the microwave. The gel was washed in 100 ml of 1:1 95%

ethanol: water in the microwave for one minute and rehydrated in distilled water for 2-3

minutes at room temperature. The proteins in the gel were then reduced in a solution of

0.04% sodium thiosulphate in the microwave for one minute. This was followed by

washing in distilled water for 30 seconds at room temperature and staining in 100 ml of

water, 200 mg of silver nitrate, and 150 µl 37% formaldehyde for one minute in the

microwave. Finally, the gel was washed for 30 seconds in distilled water, and developed

in a solution of 100 ml water, 6 g sodium carbonate, 10 µl 4% sodium thiosulfate, and

100 µl 37% formaldehyde at room temperature. 3% acetic acid was used to stop the

Page 65: Good, better, best. Never let it rest. Until your good is ...

51

developing reaction, and the results photographed using the ChemiDoc MP imaging

system.

3.2.7 Prediction of phosphorylation sites within vertebrate Gelatinase-A homologues

The sequences of vertebrate MMP-2 genes (see Appendix A) were obtained from

PubMed protein database (http://www.ncbi.nlm.nih.gov/protein/?term=MMP-2). NetPhos

2.0 software (http://www.cbs.dtu.dk) was used to predict the phosphorylation sites. This

program is based on a neural network method that predicts tyrosine, threonine and serine

phosphorylations domains located in eukaryote proteins (Blom et al. 1999).

Page 66: Good, better, best. Never let it rest. Until your good is ...

52

3.3 Results

3.3.1 The purification of Mmp2 using gelatin affinity chromatography

In order to determine the phosphorylation status of Mmp2 in zebrafish muscle, I

attempted to purify this protease from skeletal muscle using gelatin affinity

chromatography. I was able to purify small amounts of Mmp2 from adult zebrafish

muscle and the identity of the eluted protein was confirmed by probing western blots of

eluted fractions with anti-Mmp2 (Figure 3.1). Further, when I modified the protocols and

used urea in the elution buffer in some experiments, I was able to detect gelatinolytic

activity in the eluate (Figure 3.2), although the residual urea in these samples caused

extensive distortion of the bands during electrophoresis. I conclude that gelatin affinity

chromatography can be an effective method for extracting Mmp2 from fish muscle tissue.

However, replicating these results proved very challenging, presumably due to low yields

and/or degradation of the protease during or after chromatographic purification.

In my final set of experiments, I dramatically increased the amount of starting

material, using the muscle dissected from 57 adult fish. Unfortunately, this resulted in the

highly contaminated eluate shown in Figure 3.3. I was not able to detect any gelatinolytic

activity in zymograms of this material (Figure 3.4), nor was there any Mmp2

immunoreactivity (Figure 3.5). It seems likely that the Mmp2 broke down during

homogenate preparation or during storage, as both homogenate and eluate were stored for

one week while phosphorylation analysis was conducted, before immunobloting and

zymography was done in order to verify the presence of Mmp2.

Page 67: Good, better, best. Never let it rest. Until your good is ...

53

3.3.2 Mmp2 phosphorylation status remains undetermined

Analysis of phosphorylation was conducted prior to confirmation of the presence of

Mmp2 in the eluate. In hindsight, this was an error. However, a small amount of 62 kD

protein appears to be present in the eluate, which may represent Mmp2 (Figure 3.3).

Moreover, abundant proteins in the homogenate muscle sample were clearly detectable in

both SYPRO Ruby and Pro-Q Diamond stains, indicating the workability of this

technique to analyze the phosphorylation status of zebrafish proteins (Figure 3.6 A).

Using SYPRO Ruby stain, the 62 kD protein present in the eluate is just detectable

(Figure 3.6 A and B), but it is not detectable using Pro-Q Diamond (Figure 3.7),

suggesting that Mmp2 either is not phosphorylated, or its abundance is below the limit of

detection for Pro-Q Diamond. The lower limit of detection for Pro-Q Diamond is in the

range of 1-16 ng of phosphoprotein (Invitrogen product information sheet), depending on

the extent of phosphorylation, while the limit of detection for SYPRO Ruby is

approximately 1 ng of protein (Invitrogen product information sheet). Thus, if the circled

band in Figure 3.6 represents a few nanograms of Mmp2, it may be that the

phosphorylation is below the limit of Pro-Q Diamond sensitivity. Therefore, it is

impossible to determine what the phosphorylation status of Mmp2 is from this result.

3.3.3 Prediction of putative MMP-2 phosphorylation sites

Using NetPhos software, human MMP-2 phosphorylation sites have been predicted,

and several of these sites have been empirically verified by mass spectrometry

(Sariahmetoglu et al. 2007; Jacob-Ferreira et al. 2013). One of the major questions in my

Page 68: Good, better, best. Never let it rest. Until your good is ...

54

thesis is whether this post-translational regulatory mechanism might apply outside of

humans, and may therefore represent an evolutionarily ancient characteristic of

gelatinases. Because I was unable to establish the phosphorylation status of Mmp2

empirically, I used NetPhos to look for putative phosphorylation sites in the sequences of

gelatinase-A orthologues from as many vertebrates for which sequence data are available.

The domain structure of gelatinase-A orthologues is highly conserved, with a

stereotypical pattern of cysteines that form disulfide bridges that constrain protein

folding. The position of these cysteine residues makes the aligning of MMP-2 sequences

from different species relatively straightforward even when there are short insertions or

deletions. This also makes the conservation, or lack of conservation, of putative

phosphorylation sites quite obvious. NetPhos predicted that there are 14 high-probability

phosphorylation sites common to all or nearly all gelatinase-A sequences (out of an

average of 32 high-probability phosphorylation sites per protein) (Figure 3.8). Three of

these sites are conserved in all 73 sequences available, seven of them are conserved in

72/73, and four are conserved in 71/73 sequences. I did not consider sites that showed

less conservation.

Seven of the conserved high-probability phosphorylation sites are located in the

collagen-binding domain, and six predicted sites in the collagnase-like domain. In the C-

terminal hemopexin-like domain, there is one predicted site, on a tyrosine residue.

Interestingly, there are no conserved phosphorylation sites predicted in the pro-peptide

domain.

Page 69: Good, better, best. Never let it rest. Until your good is ...

55

Figure 3.1: Western blotting confirms purification of Mmp2 using gelatin affinity chromatography. A single Mmp2 immnuoreactive band is detected in the eluate of gelatin affinity columns at the predicted 72 kD molecular weight of Mmp2.

72 KD

56 KD

43 KD

26 KD

170 KD

130 KD

Blot Anti-Mmp2

Column Eluate

Page 70: Good, better, best. Never let it rest. Until your good is ...

56

Figure 3.2: Gelatin zymography verified the presence of Mmp2 in the eluate. Gelatinolytic activity is detected at ~72 kD molecular weight of Mmp2 (black circle) in the eluate, supporting the identification of this material as Mmp2. Bands in these samples were distorted due to high concentrations of urea. Lane1: protein marker; Lane 2: gelatin column eluate.

2 2

72

50

25

10

15

100

150

250

KD

1

Page 71: Good, better, best. Never let it rest. Until your good is ...

57

Figure 3.3: Silver staining of homogenate and gelatin affinity eluate. The bands detected in lane two represent the homogenate proteins while the bands in lane three represent the gelatin column eluate proteins after the purification. The appearance of non-Mmp2 proteins in lane 3 (between 75 and 50 KD) indicate inadequate column washing resulted in contamination. Lane 1: protein marker; Lane 2: homogenate; Lane 3: gelatin column eluate.

1 2 3

10 15

20 25

50

75

100

150

250

KD

Page 72: Good, better, best. Never let it rest. Until your good is ...

58

Figure 3.4: Gelatin zymography of eluate obtained from the large-scale chromatography. Homogenate and eluent from 57 adult zebrafish subject to gelatin zymography did not show any gelatinolytic activity around gelatinase-A or -B molecular weights (72 and 95 kD). Lane1: protein marker; Lane 2: gelatin column eluate.

170 130

95

72

56

11 17

26

KDa

1 2

Page 73: Good, better, best. Never let it rest. Until your good is ...

59

Figure 3.5: Western blotting of homogenate and eluted proteins obtained from the large-scale chromatography. The affinity purified samples and the homogenate were run on a western blot and probed using anti-Mmp2. No Mmp2 immunoreactivity was recognized in its molecular weight of 72 kD suggesting protein degradation.

170 130

95

56

72!

43

17 11

KD

34

MW Homogenate Eluent

Page 74: Good, better, best. Never let it rest. Until your good is ...

60

A) B)

Figure 3.6: SYPRO Ruby staining of homogenate and eluate. A) SYPRO Ruby stained gel. Lane1: protein marker; Lane 2: adult homogenate; Lane 3: gelatin chromatography eluate. B) 3D densitometric analysis of SYPRO Ruby stained gel. The blue circle in both images indicate on a protein consistent with Mmp2 molecular weights (72 and 62 KD) is barely detectable in the eluate lane.

Figure 3.7: Pro-Q Diamond gel staining of muscle homogenate and eluate. Lane 1: protein marker; Lane 2: adult homogenate; Lane 3: gelatin chromatography eluate. The eluate protein (Lane 3) consistent with Mmp2 molecular weights (72 and 62 KD) did not stain using Pro-Q Diamond, indicating protein degradation or below the limit of detection.

Page 75: Good, better, best. Never let it rest. Until your good is ...

61

Figure 3.8: Potential phosphorylation sites conserved in 73 vertebrate gelatinase-A sequences. Schematic structure of gelatinase-A domains with disulfide linkages between conserved cysteines (brown) illustrated with fine black lines. High probability threonine (T), serine (S) and tyrosine (Y) phosphorylation sites that are conserved in at least 71/73 sequences analyzed are shown in green, blue and red, respectively. The thick horizontal black line represents the NetPhos cut-off for identifying a putative phosphorylation site, and the height of the blue, green and red lines represents the 'phosphorylation potential' NetPhos has attributed to these sites. High probability phosphorylation sites that are conserved in all gelatinase-A sequences (T155, Y425 and Y581) are rendered with 100% opacity. Sites that are conserved in 72/73 sequences (T307, T319, T460, S205, S246, Y182 and Y314) are rendered with 50% opacity. Sites conserved in 71/73 sequences (S365, S434, Y302 and Y329) are rendered with 10% opacity.

Page 76: Good, better, best. Never let it rest. Until your good is ...

62

3.4 Discussion

The complex regulation of MMPs, which is affected by several factors including

regulation by post-translational processes, has been the centre of attention for some time.

The action of phosphorylation in controlling MMP activity had not been considered until

2007. Sariahmetoglu et al. (2007) reported for the first time that human MMP-2 is

phosphorylated on several serine and thronine residues, and that it dramatically regulates

the enzymatic activity of this protease. Elucidating whether this phenomenon may apply

in a broad spectrum of vertebrates including zebrafish is the focus of this chapter of my

thesis. This effort may reveal an evolutionarily ancient characteristic of gelatinases.

In this exploration, several efforts were made, both empirically and in silico, to

determine if Mmp2 from zebrafish muscle may be phosphorylated in vivo. Despite some

success using gelatin chromatography to purify gelatinase-A from zebrafish muscle, I

was not able to determine whether Mmp2 is phosphorylated in zebrafish. This may be

due to the limit of detection of Pro-Q Diamond in combination with poor protein yields

and/or degradation of the protein during or after purification.

In order to continue the work, I tested my hypothesis in silico, using software to look

for phosphorylation sites in the sequences of gelatinases from taxa for which sequence

data are available. I used NetPhos, a bioinformatic analysis method, to predict specific

motifs within protein sequences that are liable to phosphorylation. Although

phosphorylation sites have been predicted and empirically verified at several serine and

threonine residues in human and rat MMP-2 (Sariahmetoglu et al. 2007; Sariahmetoglu et

al. 2012), nothing has been done to verify if these sites are evolutionarily conserved. If

Page 77: Good, better, best. Never let it rest. Until your good is ...

63

they are of functional significance, they should be conserved in many species. This would

support the hypothesis that gelatinases have important role(s) within the cells, and

suggest that these functions may be regulated by phosphorylation. I found that there are a

number of predicted phosphorylation sites, in particular within the collagen-binding

domain, conserved in zebrafish as well as in all other vertebrates for which sequence data

are available. One of these (S365) has already been empirically verified by mass

spectrometry as being phosphorylated in humans (Sariahmetoglu et al. 2007), suggesting

that phosphorylation within this domain affects substrate binding.

As a result of this work, I conclude that gelatinase-A proteins are likely to be

phosphorylated in vertebrates. This implies the existence of one or more kinases and one

or more phosphatases that act on gelatinase-A orthologues, either extracellularly or

intracellularly. Because the putative phosphorylation sites identified do not conform to

the SxE motif recognized by known exokinases (Yalak et al. 2014), I favour the former

hypothesis. Thus it seems likely that some intracellular mechanism involving a specific

kinase has evolved to regulate the activity of this protease within the sarcomere.

Due to the technical difficulties experienced in purifying Mmp2, work must be done

to refine this process in order to analyze the phosphorylation status of Mmp2 empirically.

Although zebrafish is an ideal model to analyze the regulation of MMP activity, the

difficulties in obtaining sufficient quantities of purified Mmp2 from zebrafish make it

worthwhile to attempt this approach using muscle tissue from larger fish, such as salmon

or trout.

Page 78: Good, better, best. Never let it rest. Until your good is ...

64

3.5 References

Blom N, Gammeltoft S, Brunak S. 1999. Sequence and structure-based prediction of eukaryotic protein phosphorylation sites. J Mol Biol. 294(5): 1351- 62.

Brinckerhoff CE, Matrisian LM. 2002. Matrix metalloproteinases: a tail of a frog that became a prince. Nat Rev Mol Cell Biol. 3(3): 207-14.

Cheng HC, Qi RZ, Paudel H, Zhu HJ. 2011. Regulation and function of protein kinases and phosphatases. Enzyme Res. 2011: 1-3. Delom F, Chevet E. 2006. Phosphoprotein analysis: from proteins to proteomes. Proteome Sci. 4: 15.

Hadler-Olsen E, Solli AI, Hafstad A, Winberg JO, Uhlin- Hansen L. 2014. Intracellular MMP-2 activity in skeletal muscle is associated with type II fibers. J Cell Physiol. 230(1): 160-9.

Jacob-Ferreira AL, Kondo MY, Baral PK, James MN, Holt A, Fan X, Schulz R. 2013. Phosphorylation status of 72 kDa MMP-2 determines its structure and activity in response to peroxynitrite. PLoS One. 8(8): e71794.

Kwan JA, Schulze CJ, Wang W, Leon H, Sariahmetoglu M, Sung M, Sawicka J, Sims DE, Sawicki G, Schulz R. 2004. Matrix met- alloproteinase-2 (MMP-2) is present in the nucleus of cardiac myocytes and is capable of cleaving poly (ADP-ribose) polymerase (PARP) in vitro. FASEB J. 18(6): 690–692.

Sariahmetoglu M, Crawford BD, Leon H, Sawicka J, Li L, Ballermann BJ, Holmes C, Berthiaume LG, Holt A, Sawicki G, et al. 2007. Regulation of matrix metalloproteinase-2 (MMP-2) activity by phosphorylation. FASEB J. 21(10): 2486-2495.

Sariahmetoglu MI, Skrzypiec-Spring M, Yossef N, Jacob-Ferreira AL, Sawicka J, Holmes C, Sawicki G, Schulz R. 2012. Phosphorylation status of matrix metalloproteinase 2 in myocardial ischaemia-reperfusion injury. Heart. 98(8): 656-62.

Sawicki G, Leon H, Sawicka J, Sariahmctoglu M, Schulze CJ, Scott PG, Szczesna-Cordary D, Schulz R. 2005. Degradation of myosin light chain in isolated rat hearts subjected to ischemia-reperfusion injury: a new intracellular target for matrix metalloproteinase-2. Circulation. 112(4): 544-552.

Sawicki G. 2013. Intracellular regulation of matrix metalloproteinase-2 activity: new strategies in treatment and protection of heart subjected to oxidative stress. Scientifica. 2013:1-12. doi:10.1155/2013/130451.

Schulz R. 2007. Intracellular targets of matrix metalloproteinase-2 in cardiac disease: rationale and therapeutic approaches. Annu Rev Pharmacol Toxicol. 47: 211–242.

Page 79: Good, better, best. Never let it rest. Until your good is ...

65

Tagliabracci VS,Engel JL, Wen J, Wiley SE, Worby CA, Kinch LN, Xiao J, Grishin NV, Dixon JE. 2012. Secreted kinase phosphorylates extracellular proteins that regulate biomineralization. Science. 336 (6085): 1150-1153.

Ubersax JA, Ferrell JE Jr. 2007. Mechanisms of specificity in protein phosphorylation. Nat Rev Mol Cell Biol. 8(7): 530-41.

Wang W, Schulze CJ, Suarez-Pinzon WL, Dyck JR, Sawicki G, Schulz R. 2002. Intracellular action of matrix metalloproteinase-2 accounts for acute myocardial ischemia and reperfusion injury. Circulation.106(12):1543-1549.

Yalak G, Ehrlich YH, Olsen BR. 2014. Ectoprotein kinases and phosphatases: an emerging field for translational medicine. J Transl Med. 12:165.

Yang Y, Candelario-Jalil E, Thompson JF, Cuadrado E, Estrada EY, Rosell A, Montaner J, Rosenberg GA. 2010. Increased intranuclear matrix metalloproteinase activity in neurons interferes with oxidative DNA repair in focal cerebral ischemia. J Neurochem. 112(1): 134–149.

Page 80: Good, better, best. Never let it rest. Until your good is ...

66

Chapter 4: General Discussion and Conclusions

The discovery of matrix metalloproteinases (MMPs) in the 1960s has had enormous

impact on our understanding of organismal development, evolution, and physiology, as

well as many pathologies, such as the metastasis of cancer cells and cardiac disease. New

biological roles of these enzymes are still being uncovered. Originally they were

characterized as class-1 secreted enzymes that work to hydrolyze and remodel a variety

of extracellular matrix proteins under both pathological and physiological conditions

(reviewed in Lenz et al. 2000; Bellayr et al. 2009). However, this assessment, and even

the name "matrix metalloproteinases”, does not encompass their biological roles. In

addition to matrix proteins, MMPs degrade many non-matrix substrates, both

extracellularly and intracellularly. When researchers first detected MMPs within cells,

little attention was given to their potential roles there. However, the pathological and/or

physiological roles that intracellular MMPs may play are now being explored as

researchers recognize the significance of their activities (reviewed in Schulz 2007;

Cauwe and Opdenakker 2010). The vast majority of this work has been conducted using

in vitro studies and, to my knowledge, all of it has been focused on mammalian cells

rather than non-mammalian vertebrate or invertebrate organisms. This, and the emerging

popularity of the zebrafish as a model system for the study of MMPs (Wyatt et al. 2009),

led me to investigate whether gelatinase-A accumulates within the sarcomeres of non-

mammalian striated muscle, and whether the activity of Mmp2 in zebrafish is regulated

by phosphorylation, as has been shown in mammals. This investigation adds to the pool

of current knowledge by looking at the question from a different evolutionary

Page 81: Good, better, best. Never let it rest. Until your good is ...

67

perspective, and by assessing the localization and phosphorylation status of Mmp2 in

skeletal muscle rather than in tissue culture or cardiac muscle. All of my data support the

theory that gelatinase-A is present within the sarcomeres of striated (i.e., both cardiac and

skeletal) muscle and likely has physiological role(s) that may be regulated by

phosphorylation.

4.1 Mmp2 is unequivocally localized intracellularly within skeletal muscle in zebrafish

As outlined in Chapter 2, I demonstrate that Mmp2 is unequivocally localized

intracellularly in the skeletal myofibrils of zebrafish. My result is partially consistent with

the findings of other researchers who looked at MMP-2 inside cardiac myocytes (Wang

et al. 2002; Sawicki et al. 2005; Sung et al. 2007; Ali et al. 2010). However, I found that

Mmp2 protein accumulates exclusively in the M-band of the sarcomeres in zebrafish

skeletal muscle, in contrast to the localization of MMP2 in the Z-disc region in

mammalian cardiac myocytes. However, even in the context of mammalian cardiac

myocytes, the data are not entirely consistent with Z-disc localization of MMP-2. Using

immunogold electron microscopy, MMP2 immunoreactivity has been seen scattered

throughout sarcomeres of rat cardiac myocytes (Wang et al. 2002). Sawicki et al. (2005)

discovered that in rat hearts subjected to ischemia/reperfusion injury (IRI), myosin light

chain-1 (MLC-1), a component of thick filaments found in the A-band of sarcomeres, is

an intracellular substrate that is degraded by MMP2. In addition, immunogold labeling

shows that MMP2 is localized along the A band/thick filaments (reviewed in Chow et al.

2007; Cauwe and Opdenakker 2010).

More consistent with the purported Z-disc localization, in human and rat

Page 82: Good, better, best. Never let it rest. Until your good is ...

68

cardiomyocytes MMP2 is found close to the T12 epitope of titin, which is bound to the

Z-disc, while it is not co-localized with the M8 epitope that is associated with the M-band

(Ali et al. 2010; reviewed in Müller et al. 2012). Furthermore, Sung et al. (2007) report

the co-localization of MMP2 with the cytoskeletal protein α-actinin at the Z-disc and that

α-actinin and another cytoskeletal protein, desmin, are subject to breakdown by MMP2

following oxidative stress stimulus in isolated rat hearts (reviewed in Kandasamy et al.

2010; Ali et al. 2011a).

Thus, although some data are consistent with MMP2 being present at the Z-discs of

mammalian cardiac myocytes, different studies localize it scattered throughout the

sarcomere and acting on M-band proteins. In my experimental work, Mmp2 has been

clearly localized in the M-band. This raises the possibility that the localization of this

protease within the sarcomere may not be static; its location may be modulated by the

physiological/pathological state of the cells. Alternatively, this discrepancy between my

data and those of previous researchers may reflect differences between cardiac and

skeletal, or mammalian and teleost muscle, or both.

Recently, dystrophin, which links the actin cytoskeleton of the muscle cell to the

membrane complexes that bind laminin in the extracellular matrix, has been identified as

a novel substrate for MMP2 in rabbit hearts with ischemic injury (Buchholz et al. 2014).

Ischemic preconditioning prevents breakdown of dystrophin by inhibiting the activity of

MMP2 (reviewed in Bozzi et al. 2015). Interestingly, MMP2 is found not only in cardiac

myocytes, but is also localized in skeletal muscles of mouse (Hadler-Olsen et al. 2014).

In situ gelatin zymography on sections of vastus lateralis and bicep muscles stained with

Page 83: Good, better, best. Never let it rest. Until your good is ...

69

ATPase reveals that type II fast-twitch muscle fibres have more gelatinolytic activity

compared to the type I slow-twitch fibres, and immunostaining reveals MMP2 in the Z-

discs of fast-twitch type II fibres, their mitochondria, and associated with their nuclear

membranes.

Skeletal and cardiac muscle are similar in their sarcomereic protein components but

they have differences, mainly regarding the mechanism triggering contraction. However,

we now know that gelatinase-A is located within the sarcomeres of both of these types of

muscles. The intracellular localization of Mmp2 in zebrafish skeletal muscle suggests this

is an ancestral characteristic in striated muscle types.

Metalloproteinases are an ancient family of proteases, with representatives in the

genomes of both prokaryotes and eukaryotes (reviewed in Massova et al. 1998; Das et al.

2003). The subset of these enzymes classified as matrix metalloproteinases (MMPs) are

generally thought to have arisen as a result of gene duplication events associated with the

emergence of chordates, although there are examples of MMP-like proteases in

invertebrates, plants and even some bacteria, suggesting that this group may even have

been around longer than multicellular organisms (reviewed in Massova et al. 1998).

Consistent with this, MMP-like genes in plants and invertebrates are more similar to each

other than they are to vertebrate MMPs (reviewed in Fanjul-Fernandez et al. 2010).

Gelatinases, which are different from other MMPs due to the presence of fibronectin-

type II (FnII) repeats in their catalytic domains (reviewed in Nagase et al. 2006), are

absent among the MMPs of Cnidarians (reviewed in Sarras et al. 2002) and were thought

Page 84: Good, better, best. Never let it rest. Until your good is ...

70

to be unique to vertebrates (Das et al. 2003). However, the urochordate Ciona intestinlis

MMPe sequence (Huxley-Jones et al. 2007) clearly has FnII repeats within the catalytic

domain, showing that the gelatinases arose earlier in the deuterostome lineage than

previously thought. It would seem the first gelatinases appeared during the expansion of

the ancestral chordata MMP genes during evolution.

With regard to the relationship of gelatinases to striated muscle, based on the

observation that Cnidarians such as hydra and jellyfish have both smooth and striated

muscles (reviewed in Seipel and Schmid 2005), it appears that muscle developed initially,

and then gelatinases either took on their unknown function within the muscle, or

gelatinase-A evolved from an unknown MMP (which was initially lacking FnII repeats)

that functioned within the muscle prior to the divergence of chordates. It would therefore

be interesting to determine whether any of the MMP-like proteases found in the hydra

genome accumulate within the sarcomeres of these animals.

4.2 Evolutionary argument for an important physiological function of intracellular

gelatinase-A

While some alternatively spliced forms of MMP-2 genes do not share the presence of

an N-terminal signal peptide with other MMPs, canonical MMP-2 sequence domains

clearly do (reviewed in Kandasamy et al. 2010; Sawicki 2013). Proteins with N-terminal

secretory signals are subjected to a cascade of interactions that are generally thought to

culminate in their export from the cell. These processes involve moving the nascent

secretory proteins into the lumen of endoplasmic reticulum (ER), then to the Golgi

apparatus, and finally targeting them to vesicles bound for fusion with the plasma

Page 85: Good, better, best. Never let it rest. Until your good is ...

71

membrane for secretion. The first step in this process is the recognition of the secretory

signal (SS) near the amino (N) terminus of the nascent polypeptide as it emerges from the

ribosome by the signal recognition particle (SRP). The SS consists of a stretch of

approximately 20 primarily hydrophobic amino acids with a core of at least 10

hydrophobics in a row. Binding of the SRP complex blocks entry of transfer ribonucleic

acids (tRNAs) to the A-site on the large ribosomal subunit, causing a pause in translation,

until the whole complex is bound to the rough ER through binding to the signal

recognition particle receptor (SRPR) on the ER membrane. The SRPR releases the SRP

and translation resumes. Subsequently, the SS is positioned to enter the ER through a

pore in the protein translocation complex, called Sec61. As translation proceeds, the

nascent protein moves through the translocon into the ER, and the signal peptidase cuts

off the SS. This is an important point to mention, because it means that mutations in the

SS will not have any effect on the function of the mature protein; it will only affect its

localization. Completion of translation releases the polypeptide into the lumen, where an

enormous group of molecular chaperones assists in folding; one of these chaperones is

called Binding immunoglobulin Protein (BiP), which comes to reside in the ER by virtue

of its own very effective SS (Cooper and Hausman 2009).

In 2011b, Ali and coworkers elucidated two mechanisms underlying the surprising

intracellular localization of MMP-2 in mammalian cardiac myocytes. An alternative

splice variant of MMP-2 lacking the SS motif is expressed in human cardiomyocytes,

giving a simple mechanism that results in some cytoplasmic MMP-2. There is no

evidence for the expression of such a splice variant in zebrafish. However, by applying a

Page 86: Good, better, best. Never let it rest. Until your good is ...

72

chimeric protein approach Ali and his colleagues showed that the SS of the canonical

MMP-2 does not result in efficient sorting of the nascent MMP-2 into the secretory

pathway, leading to a significant portion of this protease being retained in the cytoplasm.

The SS of BiP is known to be efficiently recognized by the SRP, and replacing the SS of

MMP-2 with that of BiP significantly increases the amount of MMP-2 protein secretion

compared to the canonical MMP-2. On the other hand, replacing the BiP SS with that of

MMP-2 dramatically reduces the proportion of BiP entering into the ER. Thus, it would

seem the SRP does not recognize the SS of canonical MMP-2 efficiently, and this gives

rise to a significant intracellular pool of this protease. Expression of the splice variant

form that completely lacks any SS would therefore simply increase the abundance of

MMP-2 in the cytoplasmic pool (reviewed in Jacob-Ferreira and Schulz 2013a; DeCoux

et al. 2014).

This is consistent with my observation that zebrafish Mmp2 has a signal sequence

that is poorly recognized as such by software designed to detect secreted proteins.

Empirically, others in our lab have observed that expressing epitope tagged proteins

bearing the zebrafish Mmp2 SS often remain cytoplasmic (E. Chaston, personal

communication). Moreover, all the gelatinase-A orthologues for which I was able to get

good sequence data generally share this poorly recognized SS. Mutations in the SS do not

normally have deleterious consequences on the function of mature proteins. Therefore, if

efficient secretion of gelatinase-A proteins were selectively advantageous, mutations that

improve the efficiency of the SS would become fixed in the population and inefficient

SSs would be uncommon, as is the case for most secreted proteins. The fact that I observe

Page 87: Good, better, best. Never let it rest. Until your good is ...

73

the opposite implies that selective pressure favours inefficient secretion of gelatinase-A;

this suggests an advantageous, i.e., physiological rather than pathological, intracellular

function for this protease.

The conservation of the phosphorylation sites in gelatinase-A orthologues provides

another basis from which to follow the same line of thinking. Although extracellular

kinases have recently been discovered (reviewed in Yalak and Vogel 2012), the action of

kinases on their protein substrates requires a relatively high concentration of ATP, a

common feature within an intracellular milieu, but not common extracellularly. As

mentioned previously, human MMP-2 is phosphorylated, and this phosphorylation

dramatically alters its enzymatic activity, altering both KM and kcat (Sariahmetoglu et al.

2007). I show in Chapter 3 that several high-probability phosphorylation sites are

conserved among the gelatinase-A orthologues for which sequence data are available.

Moreover, the majority of these phosphorylation sites are situated in the collagen-binding

domain of the MMP-2 structure. Unlike phosphatases, which are notably promiscuous

regarding their substrates, kinases are generally very specific. The conservation of

putative kinase recognition motifs across such a broad phylogenetic sample of gelatinase-

A orthologues suggests functional constraints of these sites, implying the physiological

importance of the action of these kinases on gelatinase-A. This is consistent with reports

regarding the effect of phosphorylation on the activity of MMP-2 under the pathological

condition of IRI. Sariahmetoglu et al. (2012) found that isolated rat hearts were protected

from IRI, and degradation of troponin I was reduced by treatment with phosphatase

inhibitors, suggesting that keeping MMP2 in a phosphorylated (inactive) state, may be an

Page 88: Good, better, best. Never let it rest. Until your good is ...

74

effective approach to protecting heart muscle from the damage caused by ischemic

events. Another study, by Jacob-Ferreira et al. (2013b), reports the influence of the

phosphorylation on the conformation of MMP-2, as well as its reaction with ONOO-. The

combined effect of both ONOO- and dephosphorylation of MMP-2 dramatically increases

its proteolytic activity over the effect of ONOO- alone, suggesting that mis-regulation of

MMP-2 phosphorylation may be more important in the pathology of IRI than the direct

effect of activation by reactive oxygen species (ROS) (all reviewed in Sawicki 2013;

Jacob-Ferreira and Schulz 2013a).

It is also possible that phosphorylation modulates the interaction of gelatinase-A with

other protein partners, and thereby alters the distribution of the protein within the

sarcomere, which might explain the discrepancy between my observations of Mmp2 in

the M-band of zebrafish muscle and other reports of MMP2 at the Z-discs.

4.3 What role(s) does gelatinase-A play in the sarcomere?

What are the possible novel action(s) of Mmp2 in the sarcomere? Might there be

beneficial role(s) that makes keeping an intracellular pool of this protease worth the risk

of its pathological effects? All the facts that I have discussed in this thesis support the

theory that this protease has some conserved function(s) within the contractile apparatus,

but these function(s), and how they may be modulated by phosphorylation, remain

obscure. To date, there have been no reports of muscle myopathies in MMP2 knock-out

in mice, but like the phenotypes of other MMP knock-outs, these effects may not be

obvious in mammals in which there are numerous redundant members of this family of

proteases. Significant negative effects of the loss of MMP2 genes are not obvious in

Page 89: Good, better, best. Never let it rest. Until your good is ...

75

animals tested under lab conditions. This may not be the case for those animals living in

normal, wild conditions, in which survival and daily activities such as finding food and

reproduction may require more demanding physical activity than regularly required of

laboratory mice.

Mechanical and chemical stresses have an impact on proteins within and outside of

all cells, and this is particularly true of the contractile apparatus. Therefore, it is not

surprising that proteolytic recycling of damaged proteins is essential to maintain muscle

function (reviewed in Lyon et al. 2013). For example, calpain 3 is a muscle-specific

cystein protease thought to degrade specific isoforms of titin, and mutations in this

protease result in limb girdle muscular dystrophy (reviewed in Clark et al. 2002).

As it appears that Mmp2 accumulates in the M-bands only after the sarcomeres have

become established (Chapter 2), it does not seem likely that this protease plays an

important role in the initial establishment of the contractile apparatus. According to this I

speculate that it may participate in providing homeostasis to the sarcomere, either during

normal muscle activity or during recovery from intense exercise, through proteolysis of

unneeded or damaged proteins inside the contractile system, thereby encouraging further

regeneration of new protein molecules.

We know that there are a significant number of proteins associated with the M-band

of the sarcomeres. Several of these, such as myomesin and M-protein, are thought to

function primarily in maintaining the structure of the thick filaments by cross linking the

the anti-parallel tails of the myosin heavy chains and the carboxyl termini of the giant

Page 90: Good, better, best. Never let it rest. Until your good is ...

76

titin proteins that keep the thick filaments centred in the M-band (reviewed in Hu et al.

2015). More notably in the context of this thesis are the large collection of kinases and

phosphatases present in the M-band.

Protein Kinase A (PKA) and Protein Kinase C (PKC) are both present and active

within the M-band (Ackermann and Kontrogianni-Konstantopoulos 2011). Protein kinase

C is particularly noteworthy because it has been shown to phosphorylate MMP-2 in vitro

(Sariahmetoglu et al. 2007). Unc-83, a serine/threonine protein kinase associated with the

M-band of striated muscle in C. elegans, plays a role in thick filament assembly and

maintenance (Hoppe et al. 2010). Obscurin is an essential protein in the margin of the

sarcomeric M-band that contributes to myofibrillogenesis, cytoskeleton organization, and

cell adhesion. It has catalytically active kinase domains at its C-terminus (Hu and

Kontrogianni-Konstantopoulos 2013), making this region of the sarcomere inherently a

site of protein phosphorylation. Finally, titin itself possesses an active kinase domain,

which interestingly is present near its carboxyl terminus which is the end that is found in

the M-band. Of particular note, the kinase activity of titin is regulated by mechanical load

(Puchner et al. 2008), and is thought to regulate muscle-specific RING finger domain

protein (MuRF)-dependent protein turnover in response to exercise (Lange et al. 2005).

With respect to phosphatases, calcineurin (a.k.a. protein phosphatase 2B, (PP2B)), a

calmodulin-dependent serine/threonine phosphatase localized in the M-band of skeletal

muscle sarcomeres, is thought to regulate the function of striated muscles and their

development (Torgan and Daniels 2006). Protein phosphatase 2A and small C-terminal

domain (CTD) phosphatase-like 1 (SCPL-1) are also found in the M-band, but their

Page 91: Good, better, best. Never let it rest. Until your good is ...

77

functions remain uncharacterized (reviewed in Hu et al. 2015).

One prominent role of the kinases and phosphatases in the M-band is the regulation

of proteasomal recycling of sarcomeric proteins (reviewed in Hu et al. 2015). This is

entirely consistent with the role that I suggest gelatinase-A plays in the homeostasis of

the contractile apparatus. The continued contraction of muscles under mechanical stress

leads to damage to the protein contractile apparatus, which in turn is recognized and

degraded through proteasomal degradation systems to maintain homeostasis in the

muscles. The MuRFs are members of the E3 ubiquitin ligase family, localized in striated

muscles, which attach ubiquitin to damaged proteins and target them to the 26S

proteasomal degradation system for recycling (Attaix et al. 2005). Any protease that

hydrolyzes denatured or otherwise damaged proteins in the sarcomere, making them

substrates for ubiquitinylation by MuRFs and subsequent recycling, could function as a

component of this system. Pathologically, this occurs in the muscles of patients suffering

from chronic kidney disease; activation of caspase-3 results in cleavage of sarcomeric

proteins and their subsequent ubiqutinylation by MuRF-1 (Thomas and Mitch 2013).

Thus, the enzymatic machinery necessary to activate and inactivate gelatinase-A is

known to be present in the M-band, and the activity of these kinases and phosphatases

has already been linked to the regulated protein turnover of sarcomeric components. I

speculate that Mmp2 is a previously unrecognized component of this system.

In addition to the work presented here, there are some unpublished data to support

this hypothesis. Terminal Amine Isotopic Labeling of Substrates (TAILS) has allowed

the identification of some novel Mmp2 substrates by comparing the 'degradome' of

Page 92: Good, better, best. Never let it rest. Until your good is ...

78

control zebrafish embryos to that of siblings developing in the presence of an irreversible

gelatinase-specific inhibitor. Cleavage of a substrate protein by a given protease

obviously generates a novel amino (as well as carboxyl) terminus. A TAILS experiment

involves making two proteomes, one from tissue in which a protease of interest is active

and one from tissue in which that protease is inactive. The amino termini present in each

proteome are then labeled by reacting with isotopically distinct formaldehyde, and the

proteomes are then analyzed by mass spectrometry. N-termini present, either exclusively

or enriched, in the proteome in which the protease was active can then be identified and

validated as bona fide targets of the protease in question (Kleifeld et al. 2010). A TAILS

assay comparing the proteomes made from 1000 48 hpf zebrafish embryos exposed to

either a gelatinase-specific inhibitor (SB-3CT (Lee et al. 2005)) or vehicle control

identified skeletal muscle specific myosin heavy chain as a highly significant target of

gelatinases (B. Crawford, personal communication). Given that at this stage of

development the other gelatinase (Mmp9) is not significantly expressed (Harris, 2010),

this suggests that Mmp2 is likely degrading myosin heavy chain molecules in the thick

filaments of skeletal muscle. It would be very interesting to replicate this experiment in

the presence and absence of phosphatase inhibitors to see if it has the predicted effect on

the presence of this putative Mmp2 degradation target.

4.4 Conclusion

In this thesis, I set out initially to establish if Mmp2 accumulates within skeletal

muscle cells of zebrafish, and if so, where could it be? Is it similar to the myocytes of

mammals? Secondly, I wanted to determine if Mmp2 is phosphorylated in zebrafish, as it

Page 93: Good, better, best. Never let it rest. Until your good is ...

79

is in mammals, thereby supporting the hypothesis that its activity is subject to regulation

by kinases and phosphatases.

I was completely successful in the first objective by demonstrating the intracellular

localization of Mmp2 within the M-bands of sarcomeres of zebrafish skeletal muscle.

Moreover, I determined that inefficient secretion appears to be an evolutionarily

conserved feature of gelatinase-A orthologues among vertebrates. Regarding the second

objective, I was able to partially purify some of the protease using gelatin affinity

chromatography and use western blots and zymography to verify its identity. However, I

was unable to determine its phosphorylation status biochemically. On the other hand,

analysis of predicted phosphorylation sites in silico reveals there are several conserved

sites in known gelatinase-A sequences, implying this post-translational modification is of

physiological importance. To empirically verify that Mmp2 is phosphorylated in fish, as

it is in mammals, I am confident that using different materials, such as muscle tissue from

larger fish (e.g., salmon or trout) will provide enough starting material to facilitate the

experimental approach I have demonstrated here.

In the end, the intracellular localization of Mmp2, in combination with the predicted

conservation of phosphorylation sites in gelatinase-A orthologues, along with the

emerging role of M-band kinases in regulating protein turnover in the sarcomeres, expose

exciting possibilities for uncovering previously unknown roles of gelatinase-A in the

homeostasis of the contractile apparatus.

Page 94: Good, better, best. Never let it rest. Until your good is ...

80

4.5 References

Ackermann MA, Kontrogianni-Konstantopoulos A. 2011. Myosin binding protein-C slow is a novel substrate for protein kinase A (PKA) and C (PKC) in skeletal muscle. J Proteome Res.10(10): 4547-4555.

Ali MA, Cho WJ, Hudson B, Kassiri Z, Granzier H, Schulz R. 2010. Titin is a target of matrix metalloproteinase-2: implications in myocardial ischemia/reperfusion injury. Circulation. 122(20): 2039-47.

Ali MA, Fan X, Schulz R. 2011a. Cardiac sarcomeric proteins: novel intracellular targets of matrixmetalloproteinase-2 in heart disease. Trends Cardiovasc Med. 21(4): 112-118.

Ali MA, Chow AK, Kandasamy AD, Fan X, West LJ, Crawford BD, Simmen T, Schulz R. 2011b. Mechanisms of cytosolic targeting of matrix metalloproteinase-2. J Cell Physiol. 227: 3397-3404.

Attaix D, Ventadour S, Codran A, Béchet D, Taillandier D, Combaret L. 2005. The ubiquitin-proteasome system and skeletal muscle wasting. Essays Biochem. 41: 173–186.

Bellayr IH, Mu X, Li Y. 2009. Biochemical insights into the role of matrix metalloproteinases in regeneration: challenges and recent developments. Future Med Chem. 1(6): 1095-1111.

Bozzi M, Sciandra F, Brancaccio A. 2015. Role of gelatinases in pathological and physiological processes involving the dystrophin–glycoprotein complex. Matrix Biol. http://dx.doi.org/10.1016/j.matbio.2015.02.005.

Buchholz B, Perez V, Siachoque N, Miksztowicz V, Berg G, Rodríguez M, Donato M, Gelpi RJ. 2014. Dystrophin proteolysis: a potential target for MMP-2 and its prevention by ischemic preconditioning. Am J Physiol Heart Circ Physiol. 307(1): H88-96.

Cauwe B, Opdenakker G. 2010. Intracellular substrate cleavage: a novel dimension in the biochemistry, biology and pathology of matrix metalloproteinases. Crit Rev Biochem Mol Biol. 45(5): 351–423.

Chow AK, Cena J, Schulz R. 2007. Acute actions and novel targets of matrix metalloproteinases in the heart and vasculature. Br J Pharmacol. 152(2): 189–205.

Clark KA, McElhinny AS, Beckerle MC, Gregorio CC. 2002. Striated muscle cytoarchitecture: an intricate web of form and function. Annu Rev Cell Dev Biol. 18:637-706.

Cooper GM, Hausman RE. 2009. The cell: a molecular approach. Fifth edition. Washington (DC): The American Society of Microbiology Press. p. 383-432.

Das S, Mandal M, Chakraborti T, Mandal A, Chakraborti S. 2003. Structure and

Page 95: Good, better, best. Never let it rest. Until your good is ...

81

evolutionary aspects of matrix metalloproteinases: a brief overview. Mol Cell Biochem. 253(1-2): 31-40.

DeCoux A, Lindsey ML,Villarreal F, Garcia RA, Schulz R. 2014. Myocardial matrix metalloproteinase-2: inside out and upside down. J Mol Cell Cardiol. 77: 64-72.

Fanjul-Fernández M, Folgueras AR, Cabrera S, López-Otín C. 2010. Matrix metalloproteinases: evolution, gene regulation and functional analysis in mouse models. Biochim Biophys Acta. 1803(1): 3-19.

Hadler-Olsen E, Solli AI, Hafstad A, Winberg JO, Uhlin- Hansen L. 2014. Intracellular MMP-2 activity in skeletal muscle is associated with type II fibers. J Cell Physiol. 230(1): 160-9.

Harris ND. 2010. Molecular ontogeny of extracellular matrix remodeling proteases during development of the zebrafish, and the effects of simulated micro-gravity on their expression [master’s thesis]. [Fredericton (NB)]: University of New Brunswick.

Hoppe PE, Chau J, Flanagan KA, Reedy AR, Schriefer LA. 2010. Caenorhabditis elegans unc-82 encodes a Serine/Threonine kinase important for myosin filament organization in muscle during growth. Genetics. 184(1): 79-90. Hu LY, Ackermann MA, Kontrogianni-Konstantopoulos A. 2015. The sarcomeric M-region: a molecular command center for diverse cellular processes. BioMed Res Int. vol. 2015, Article ID 714197, 25 pages. doi:10.1155/2015/714197.

Hu LY, Kontrogianni-Konstantopoulos A. 2013. The kinase domains of obscurin interact with intercellular adhesion proteins. FASEB J. 27(5): 2001-2012.

Huxley-Jones J, Clarke TK, Beck C, Toubaris G, Robertson DL, Boot-Hanford RP. 2007.The evolution of the vetebrates metzinicins; insights from Ciona intestinalis and Danio rerio. BMC Evol Biol. 7: 63-83.

Jacob-Ferreira AL, Schulz R. 2013a. Activation of intracellular matrix metalloproteinase-2 by reactive oxygen–nitrogen species: Consequences and therapeutic strategies in the heart. Arch Biochem Biophys. 540(1-2): 82-93.

Jacob-Ferreira AL, Kondo MY, Baral PK, James MN, Holt A, Fan X, Schulz R. 2013b. Phosphorylation status of 72 kDa MMP-2 determines its structure and activity in response to peroxynitrite. PLoS One. 8(8): e71794.

Kandasamy AD, Chow AK, Ali MA, Schulz R. 2010. Matrix metalloproteinase-2 and myocardial oxidative stress injury: beyond the matrix. Cardiovasc Res. 85(3): 413–423.

Kleifeld O, Doucet A, auf dem Keller U, Prudova A, Schilling O, Kainthan RK, Starr AE, Foster LJ, Kizhakkedathu JN, Overall CM. 2010. Isotopic labeling of terminal

Page 96: Good, better, best. Never let it rest. Until your good is ...

82

amines in complex samples identifies protein N-termini and protease cleavage products. Nat Biotechnol. 28(3): 281-288.

Lange S, Xiang F, Yakovenko A, Vihola A, Hackman P, Rostkova E, Kristensen J, Brandmeier B, Franzen G, Hedberg B, et al. 2005. The kinase domain of Titin controls muscle gene expression and protein turnover. Science. 308(5728): 1599-603.

Lee M, Bernardo MM, Meroueh SO, Brown S, Fridman R, Mobashery S. 2005. Synthesis of Chiral 2-(4-Phenoxyphenylsulfonylmethyl) thiiranes as selective gelatinase inhibitors. Org Lett. 7(20): 4463-4465.

Lenz O, Elliot SJ, Stetler-Stevenson WG. 2000. Matrix metalloproteinases in renal development and disease. J Am Soc Nephrol. 11(3): 574-81.

Lyon RC, Lange S, Sheikh F. 2013. Breaking down protein degradation mechanisms in cardiac muscle. Trends Mol Med. 19(4): 239-249.

Massova I, Kotra LP, Fridman R, Mobashery S. 1998. Matrix metalloproteinases: structures, evolution, and diversification. FASEB J. 12(12): 1075-1095.

Müller AL, Hryshko LV, Dhalla NS. 2012. Extracellular and intracellular proteases in cardiac dysfunction due to ischemia–reperfusion injury. Int J Cardiol. 164(1): 39-47.

Nagase H, Visse R, Murphy G. 2006. Structure and function of matrix metalloproteinases and TIMPs. Cardiovasc Res. 69(3): 562 – 573.

Puchner EM, Alexandrovich A, Kho AL, Hensen U, Scha¨fer LV, Brandmeier B, Grater F, Grubmuller H, Gaub HE, Gautel M. 2008. Mechanoenzymatics of Titin kinase. PNAS. 105(36):13385–13390.

Sarras MP JR, Yan L, Leontovich A, Zhang JS. 2002. Structure, expression, and developmental function of early divergent forms of metalloproteinases in hydra. Cell Res.12(3-4): 163-176.

Sariahmetoglu M, Crawford BD, Leon H, Sawicka J, Li L, Ballermann BJ, Holmes C, Berthiaume LG, Holt A, Sawicki G, et al. 2007. Regulation of matrix metalloproteinase-2 (MMP-2) activity by phosphorylation. FASEB J. 21(10): 2486-2495.

Sariahmetoglu MI, Skrzypiec-Spring M, Yossef N, Jacob-Ferreira AL, Sawicka J, Holmes C, Sawicki G, Schulz R. 2012. Phosphorylation status of matrix metalloproteinase 2 in myocardial ischaemia-reperfusion injury. Heart. 98(8): 656-62.

Sawicki G, Leon H, Sawicka J, Sariahmctoglu M, Schulze CJ, Scott PG, Szczesna-Cordary D, Schulz R. 2005. Degradation of myosin light chain in isolated rat hearts subjected to ischemia-reperfusion injury: a new intracellular target for matrix metalloproteinase-2. Circulation. 112(4): 544-552.

Page 97: Good, better, best. Never let it rest. Until your good is ...

83

Sawicki G.2013. Intracellular regulation of Matrix Metalloproteinase-2 activity: New strategies in treatment and protection of heart subjected to oxidative stress. Scientifica. 2013:1-12. doi:10.1155/2013/130451.

Schulz R. 2007. Intracellular targets of matrix metalloproteinase-2 in cardiac disease: rationale and therapeutic approaches. Annu Rev Pharmacol Toxicol. 47: 211–242.

Seipel K, Schmid V. 2005. Evolution of striated muscle: Jellyfish and the origin of triploblasty. Dev Biol. 282(1): 14-26.

Sung MM, Schulz CG, Wang W, Sawicki G, Bautista-Lopez NL, Schulz R. 2007. Matrix metalloproteinase-2 degrades the cytoskeletal protein α- actinin in peroxynitrite mediated myocardial injury. J Mol Cell Cardiol. 43(4): 429-436. Thomas SS, Mitch WE. 2013. Mechanisms stimulating muscle wasting in chronic kidney disease: the roles of the Ubiquitin-Proteasome system and myostatin. Clin Exp Nephrol. 17(2): 174-182.

Torgan CE, Daniels MP. 2006. Calcineurin localization in skeletal muscle offers insights into potential new targets. J Histochem Cytochem. 54(1): 119–128.

Wang W, Schulze CJ, Suarez-Pinzon WL, Dyck JR, Sawicki G, Schulz R. 2002. Intracellular action of matrix metalloproteinase-2 accounts for acute myocardial ischemia and reperfusion injury. Circulation.106(12): 1543-1549.

Wyatt RA, Keow JY, Harris ND, Hache CA, Li DH, Crawford BD. 2009. The zebrafish embryo: a powerful model system for investigating matrix remodeling. Zebrafish. 6(4): 347-354.

Yalak G, Vogel V. 2012. Extracellular phosphorylation and phosphorylated proteins: not just curiosities but physiologically important. Sci Signaling. 5(225): re7.

Page 98: Good, better, best. Never let it rest. Until your good is ...

84

Appendix A

Table A.1: SignalP discrimination scores of identifiable gelatinase-A, -B, and BiP sequences of vertebrate species.

Organisms !

Gelatinase-A Accession

Gelatinase -A

D-scores

Gelatinase-B

Accession

Gelatinase -B

D-scores

BiP

Accession BiP

D-scores

Ictalurus punctatus (channel catfish) AHH43049.1

0.752 NP_001187157.1

0.749 AHH41738.1

0.8

Danio rerio (zebrafish) NP_932333.1

0.67 NP_998288.1

0.778 NP_998223.1

0.891

Pseudopodoces humilis (Tibetan

ground-tit) XP_005526411.1

0.71 XP_005524762.1

0.903 XP_005529214.1 0.913

Gallus gallus (chicken) NP_989751.1

0.814 NP_989998.1

0.883 NP_990822.1

0.881

Alligator mississippiensis

(American alligator) XP_006273079.1

0.553 XP_006260206.1

0.874 XP_006271368.1

0.869

Mus musculus (house mouse) NP_032636.1

0.884 NP_038627.1

0.877 NP_001156906.1

0.909

Rattus norvegicus (Norway rat) NP_112316.2

0.884 NP_112317.1

0.888 NP_037215.1

0.888

Peromyscus maniculatus bairdii (prairie deer mouse)

XP_006984109.1

0.845 XP_006971033.1 0.858 XP_006988789.1 0.892

Homo sapiens (human) NP_004521.1

0.812 NP_004985.2

0.916 NP_005338.1

0.898

Camelus ferus (Wild Bactrian camel) XP_006186033.1

0.103 XP_006187699.1

0.913 XP_006187781.1

0.92

Canis lupus familiaris (dog) XP_535300.4

0.213 NP_001003219.1

0.768 XP_863385.2

0.901

Panthera tigris altaica (Amur tiger) XP_007090237.1

0.104 XP_007077889.1

0.758 XP_007094457.1

0.904

Felis catus (domestic cat) XP_003998091.1

0.104 XP_003983461.1

0.741 XP_006939520.1

0.904

Capra hircus (goat) XP_005692042.1

0.104 XP_005688780.1

0.797 XP_005687195.1

0.934

Page 99: Good, better, best. Never let it rest. Until your good is ...

85

Sus scrofa (pig) NP_999357.1

0.779 NP_001033093.1

0.856 XP_001927830.3

0.77

Elephantulus edwardii (Cape elephant shrew) XP_006882587.1

0.889 XP_006881803.1

0.928 XP_006892626.1

0.921

Chrysochloris asiatica (Cape golden mole) XP_006863586.1

0.691 XP_006839297.1

0.88 XP_006834828.1

0.912

Latimeria chalumnae (coelacanth) XP_006000312.1

0.79 XP_006002447.1

0.838 XP_005987377.1

0.904

Eptesicus fuscus (big brown bat) XP_008150589.1

0.768 XP_008157169.1

0.942 XP_008153515.1

0.904

Bos taurus (cattle) NP_777170.1

0.851 NP_777169.1

0.836 NP_001068616.1

0.931

Oryctolagus cuniculus (rabbit) NP_001075678.1 0.734

NP_001075672.1

0.726 XP_008271523.1

0.904

Calypte anna (Anna's hummingbird)

XP_008499514.1

0.837 XP_008495975.1

0.801 XP_008489796.1

0.101

Cynoglossus semilaevis (tongue

sole)

XP_008308597.1

0.703 XP_008316390.1

0.72 XP_008324204.1

0.893

Stegastes partitus (bicolor damselfish)

XP_008280164.1

0.759 XP_008294599.1

0.839 XP_008299833.1

0.863

Chrysemys picta bellii (western painted turtle) XP_005289836.1 0.901

XP_005304889.1 0.878

XP_005279469.1 0.769

Tarsius syrichta (Philippine tarsier)

XP_008068148.1

0.788 XP_008056104.1

0.769 XP_008058393.1

0.917

Chlorocebus sabaeus (green monkey)

XP_007991519.1

0.811 XP_008013649.1

0.892 XP_008004447.1

0.898

Orycteropus afer afer XP_007945330.1

0.709 XP_007936550.1

0.929 XP_007954566.1

0.909

Callorhinchus milii (elephant shark)

AEW46994.1

0.628 XP_007910512.1

0.662 XP_007907713.1 0.825

Cricetulus griseus (Chinese hamster)

XP_007619400.1

0.882 XP_007639491.1 0.899

NP_001233668.1 0.834

Erinaceus europaeus (western European

hedgehog)

XP_007532773.1

0.817 XP_007533116.1

0.825 XP_007540052.1

0.904

Monodelphis domestica (gray short-

tailed opossum)

XP_001373030.2

0.791 XP_007474988.1

0.937 XP_001365714.1

0.895

Page 100: Good, better, best. Never let it rest. Until your good is ...

86

Lipotes vexillifer (Yangtze River

dolphin)

XP_007459512.1

0.103 XP_007451691.1

0.864 XP_007471548.1

0.826

Astyanax mexicanus (Mexican tetra)

XP_007246942.1

0.724 XP_007231993.1

0.759 XP_007252148.1 0.868

Physeter catodon (sperm whale)

XP_007110336.1

0.104 XP_007115590.1

0.775 XP_007117702.1

0.849

Pteropus alecto (black flying fox)

XP_006908907.1

0.165 XP_006921942.1

0.878 ELK07198.1

0.904

Myotis davidii XP_006777132.1

0.103 XP_006773601.1

0.943 XP_006768631.1 0.901

Vicugna pacos (alpaca) XP_006207898.1

0.103 XP_006202537.1

0.911 XP_006205575.1 0.92

Bos mutus XP_005890996.1

0.883 XP_005897616.1

0.836 XP_005893110.1

0.931

Myotis brandtii (Brandt's bat)

XP_005865346.1

0.1 XP_005862214.1

0.943 XP_005881560.1

0.901

Geospiza fortis (medium ground-

finch)

XP_005422778.1

0.684 XP_005422371.1

0.164 XP_005421393.1

0.527

Oreochromis niloticus (Nile tilapia)

XP_003437582.2

0.819 XP_003448187.1

0.802 XP_005470418.1

0.865

Falco cherrug (Saker falcon)

XP_005444651.1

0.8 XP_005444884.1

0.809 XP_005437288.1 0.121

Chinchilla lanigera (long-tailed chinchilla)

XP_005388374.1

0.839 XP_005392450.1

0.759 XP_005382523.1

0.917

Xenopus (Silurana) tropicalis (western

clawed frog)

NP_001015789.1

0.653 AAH76927.1

0.535 XP_002941690.1

0.894

Callithrix jacchus (white-tufted-ear

marmoset) XP_002761063.1

0.598 JAB41082.1

0.908 JAB09769.1 0.923

Microtus ochrogaster (prairie vole)

XP_005369650.1

0.847 XP_005363192.1

0.885 XP_005346064.1

0.888

Falco peregrinus (peregrine falcon)

XP_005238944.1

0.8 XP_005239973.1

0.809 XP_005231480.1

0.115

Melopsittacus undulatus (budgerigar)

XP_005152279.1

0.683 XP_005140762.1

0.877 XP_005146335.1

0.881

Page 101: Good, better, best. Never let it rest. Until your good is ...

87

Ficedula albicollis (collared flycatcher)

XP_005052630.1 0.765

XP_005057082.1

0.109 XP_005055797.1

0.913

Mesocricetus auratus (golden hamster)

XP_005079205.1

0.894 XP_005085041.1

0.882 XP_005082124.1

0.924

Cavia porcellus (domestic guinea pig)

XP_003477589.1

0.838 XP_003467841.1

0.757 XP_003470822.1

0.917

Heterocephalus glaber (naked mole-rat)

XP_004847968.1

0.905 XP_004858177.1

0.806 XP_004849435.1

0.917

Mustela putorius furo (domestic ferret)

XP_004744256.1

0.851 XP_004746360.1

0.793 XP_004757385.1

0.904

Condylura cristata (star-nosed mole)

XP_004693625.1

0.103 XP_004687492.1 0.837

XP_004677392.1

0.904

Octodon degus (degu) XP_004637072.1

0.717 XP_004636179.1

0.816 XP_004641630.1

0.908

Echinops telfairi (small Madagascar

hedgehog)

XP_004704722.1

0.781 XP_004698361.1

0.873 XP_004716507.1

0.91

Jaculus jaculus (lesser Egyptian jerboa)

XP_004664921.1

0.789 XP_004663952.1

0.887 XP_004662863.1

0.941

Sorex araneus (European shrew)

XP_012787573.1

0.103 XP_004618833.1

0.697 XP_004613383.1

0.884

Ochotona princeps (American pika)

XP_004583913.1

0.717 XP_004585927.1

0.724 XP_004593628.1

0.889

Salmo salar (Atlantic salmon)

NP_001133404.1

0.803 NP_001133929.1

0.861 NP_001135114.1

0.844

Ceratotherium simum simum (southern white

rhinoceros)

XP_004431664.1

0.744 XP_004430375.1 0.885

XP_004423507.1

0.904

Odobenus rosmarus divergens (Pacific

walrus)

XP_004395877.1

0.779 XP_004405315.1 0.803

XP_004392650.1

0.904

Trichechus manatus latirostris (Florida

manatee)

XP_004371635.1

0.816 XP_004370610.1 0.929

XP_004375722.1

0.91

Orcinus orca (killer whale)

XP_004264952.1

0.695 XP_004272975.1 0.869

XP_004269273.1

0.826

Macaca mulatta (Rhesus monkey)

AFJ70978.1

0.812 NP_001253834.1

0.892 NP_001253525.1 0.898

Page 102: Good, better, best. Never let it rest. Until your good is ...

88

Gorilla gorilla gorilla (western lowland

gorilla)

XP_004057705.1 0.812 XP_004062370.1 0.916

XP_004048650.1

0.898

Pan troglodytes (chimpanzee)

XP_009429086.1

0.812 XP_003317021.1

0.916 BAK63666.1

0.898

Saimiri boliviensis boliviensis (Bolivian

squirrel monkey)

XP_003937326.1

0.585 XP_003936522.1

0.899 XP_003940675.1 0.916

Pan paniscus (pygmy chimpanzee)

XP_008962127.1

0.103 XP_008949630.1 0.33

XP_003808092.1

0.898

Otolemur garnettii (small-eared galago)

XP_003792593.1

0.895 XP_003787712.1

0.709 XP_003785350.1

0.904

Sarcophilus harrisii (Tasmanian devil)

XP_003757305.1

0.724 XP_012409327.1

0.889 XP_012409173.1

0.915

Pongo abelii (Sumatran orangutan) XP_002826479.1 0.841

XP_002830421.1 0.896 NP_001126927.1 0.898

Page 103: Good, better, best. Never let it rest. Until your good is ...

89

Figure A.1: Histograms of MMP-2 (A), MMP-9 (B), and BiP (C) signalP D- scores.

MMP-2 A)

MMP-9 B)

BiP C)

Page 104: Good, better, best. Never let it rest. Until your good is ...

90

Table A.2: The NetPhos prediction scores of gelatinase-A sequences of vertebrate species.

(in order to save space in this thesis, the raw data from the NetPhos analysis of

gelatinase-A sequences is available online at this URL)

https://unbscholar.lib.unb.ca/islandora/object/unbscholar%3A7073

Page 105: Good, better, best. Never let it rest. Until your good is ...

91

Appendix B

I. Transmission Electron Microscopy (TEM)

The purpose of this experiment was to demonstrate the intracellular localization of

Mmp2 in zebrafish skeletal muscle. Although the immunostaining data imaged by

confocal microscopy strongly suggested that Mmp2 was present within the sarcomeres

(chapter 2), due to the limited axial resolution of this optical microscope, it was possible

that this important result could be contested. Before resorting to cryosectioning, I tried

immunogold detection of Mmp2 by transmission electron microscopy, hoping that I

would be able to unequivocally localize Mmp2 within the sarcomere has had been done

with mammalian cardiomyocytes (Wang et al. 2002; Sawicki et al. 2005).

Although I was able to get good sections of skeletal muscle tissue, the results were

disappointing because the immunogold labeling of Mmp2 was not successful (Figure A.1).

This is likely due to the necessity of using aldehyde-fixatives in the preparation of tissue

for TEM, which we now know destroy the epitope recognized by the rabbit anti-Mmp2

antibody I have used throughout this thesis.

Material and methods

Transmission Electron Microscopy (TEM)

Seventy-two hour post fertilization embryos were incubated in a primary fixative that

contained 3% paraformaldehyde in 0.1 M sodium cacodylate buffer, pH 7.4, overnight at

4°C. Samples were rinsed in sodium cacodylate buffer, with at least three changes of

Page 106: Good, better, best. Never let it rest. Until your good is ...

92

buffer, for two hours; no secondary fixative was used. The embryos were dehydrated via

increasing concentrations of ethanol (30, 50, 70, 80 to 90%) for 30 minutes each,

culminating in two 30 minute incubations in anhydrous ethanol. Ethanol was replaced

with a 1:2 ratio of LR (London Resin) white resin to ethanol and set for four hours, then

replaced with a 2:1ratio of LR white resin to ethanol and left overnight. The final change

with 100% fresh resin was made and the sample left for four hours.

Twenty-one embryos were then embedded in gelatin capsules with fresh resin and

cured in an oven at 50-55°C for 24 hours. The following day, the samples were cut into

80-100 nm ultrathin sections using a diamond knife (Leica Ultracut T ultramicrotome),

and collected on 400 mesh nickel grids with formvar support films. The grids with

ultrathin sections were floated on blocking buffer (5% Bovine serum albumin (BSA) in

PBS (20 mM phosphate pH 7.3, 137 mM NaCl, 2.7 mM KCl)) overnight in a humid

chamber at room temperature. The sections were then incubated overnight with either

anti-Mmp2 or anti-α-actinin, or blocking buffer alone (as a negative control) at room

temperature in humid chamber. They were then rinsed with PBS for15 minutes, and

incubated with protein A/G conjugated to10-nm gold particles (British Biocell

International) (diluted 1:20 in PBS) for 30 minutes at room temperature in humid

chamber. The sections were washed with a PBS 3x5 minutes, rinsed with dH2O, dried,

stained with 2% uranyl acetate for five minutes, rinsed with dH2O, stained with lead

citrate for one minute and finally rinsed in dH2O. The stained sections on grids were

dried in desiccator before being examined using a JEOL-2011 (Scanning) TEM (at 200

kV) with a Gatan Ultrascan digital camera and Digital Micrograph software.

Page 107: Good, better, best. Never let it rest. Until your good is ...

93

!

Figure I.1. Transmission Electron Micrograph of longitudinal section of zebrafish muscle processed for immunogold detection of Mmp2. A) Representative micrograph of negative control. B) The sections of sarcomeres were probed with anti-Mmp2 and while a few 10 nm gold particles were retained in some sections, these were rare (comparable to negative controls treated with no primary antibody) and exhibited no spatially distinct pattern. !

A)

Page 108: Good, better, best. Never let it rest. Until your good is ...

94

II. Investigation of Mmp2’s potential role in muscle hypertrophy

The aim of this experiment was to address whether the intracellular Mmp2 is

involved in muscle cell growth. To conduct this experiment I exercised zebrafish and

treated them with testosterone to induce muscle hypertrophy, as has previously been

reported (Piferrer and Donaldson, 1994; McClelland, et al. 2006). I hoped to detect

changes in Mmp2 distribution, phosphorylation and/or expression that might correlate

with hypertrophy.

Materials and methods

Testosterone hormone treatments

A total of 62 age-matched male and female adult zebrafish were distributed into

three litre containers (five individuals in each of 10 containers, and six in two containers).

The containers had continuous aeration as well as artificial plants for cover. The fish

were fed pellets and brine shrimp 4-5 times per day. 10% of the water was changed per

day, and the temperature was maintained between 26 and 28°C with a 14/10 hour

light/dark cycle (as described in Westerfield, 1995). Each treatment group was

maintained for 4 weeks.

Treatments consisted of exercise (see below) (n=16); hormone (400 µg/l

testosterone) plus exercise (n=15); hormone alone (n=16); or no treatment (n=15).

Exercise treatment was modelled after McClelland, et al. (2006); the fish were

exercised for three hours twice daily, for six days each week for four weeks, with a two-

hour rest period when the fish were fed. Fish were required to swim at 2 body lengths per

Page 109: Good, better, best. Never let it rest. Until your good is ...

95

second (BL/s), equivalent to 6.0 cm/s, and each week the speed was increased 1 BL/s to

the final speed of 5 BL/s (15.0 cm/s). After the fourth week the speed of the water was

maintained at 3 cm/s (∼1 BL/s), including overnight and in non-exercising conditions.

At the end of the experiment the fish were removed from the buckets and anesthetized

(see 3.2.2) as previously described, with the length and weight being recorded. After the

tail muscles were dissected from the fish, the hypertrophy of muscle tissue was

determined using quantitative PCR, with MyoD and Myf5 transcription factors as

positive controls.

RNA purification, cDNA synthesis, and qPCR

RNA was extracted using a Norgen Total RNA purification kit following the

manufacturer’s instructions. cDNA was synthesized using Enhanced Avian Reverse

Transcriptase (Sigma) according to the manufacturer’s protocol. cDNAs from each

treatment group were then subjected to qPCR in order to determine if there were

significant changes in myogenic (MyoD and Myf5) genes (with respect to housekeeping

controls GAPDH and EF1), and also to characterize any changes in the expression of

mmp2. Primers are listed in Table II.1 below. All reactions were done in triplicate for

each primer set. The PCR efficiencies for all primers and the values of critical threshold

(Ct) were verified using LinRegPCR software.

Page 110: Good, better, best. Never let it rest. Until your good is ...

96

Table II.1. The sequences of myogenic primers specific to the mRNA of the zebrafish muscle, GAPDH, EF1, and mmp2.

Primers Sequencing

MyoD Forward 5’-CACCAAATGCTGACGCAC-3’

MyoD Reverse 5’-CTTGATAAATGGTTTCCTGAGCCT-3’

Myf5 Forward 5’-AGGCTGAAGAAGGTCAATCAC-3’

Myf5 Reverse 5’-TTGCAGTCAACCATGCTCTC-3’

GAPDH Forward 5’-AATGTCTCTGTTGTGGATCTG-3’

GAPDH Reverse 5’-TCATTGTCATACCATGTGACC-3’

Ef1!α Forward 5’-AATTGGAGGTATTGGAACTG-3’

Ef1!α Reverse 5’-ATCTCAACAGACTTGACCT-3’

mmp2 Forward 5’-GTTCTGGAGATACAATGAAGC-3’

mmp2 Reverse 5’-TGAGTCTTTGAAGAAGTAGC-3’

Page 111: Good, better, best. Never let it rest. Until your good is ...

97

Results

The hormone/exercise zebrafish treatment did not result in hypertrophy

Disappointingly, none of the treatments resulted in significant increases in mass

(Figure II.1), body size, expression of myogenic genes, or expression of mmp2 (data not

shown). Indeed, although these changes were not significant, none of the treated fish

(exercise alone, hormone alone, or combination of exercise and hormone) grew as well as

the untreated controls. This suggests that the experimental conditions were somewhat

stressful to the fish, reducing their growth rates.

Because of the failure of any treatments to yield evidence of muscle hypertrophy,

interpretation of any changes in the distribution or phosphorylation of Mmp2 protein

would not have been possible, so the experiment was abandoned.

Page 112: Good, better, best. Never let it rest. Until your good is ...

98

Figure II.1: The average of adult zebrafish muscle mass treated with testosterone hormone compared to the control. The muscle mass of fish that treated with hormone as well as with exercise dose not change in comparing to the control fish. H+E= treated with hormone in combination with exercise; H-E= receiving hormone alone without exercise; E= only exercise.!

!0.4%

!0.3%

!0.2%

!0.1%

0%

0.1%

0.2%

0.3%

0.4%

0.5%

0.6%

Control H+E H E

Mea

n in

crea

se in

mas

s (g)

Page 113: Good, better, best. Never let it rest. Until your good is ...

99

References

McClelland GB, Craig PM, Dhekney K, Dipardo S. 2006. Temperature- and exercise-induced gene expression and metabolic enzyme changes in skeletal muscle of adult zebrafish (Danio rerio). J Physiol. 577(2): 739-51.

Piferrer F, Donaldson EM. 1994. Uptake and clearance of exogenous estradiol-1713 and testosterone during the early development of coho salmon (Oncorhynchus kisutch), including eggs, alevins and fry. Fish Physiol Biochem. 13(3): 219-232.

Sawicki G, Leon H, Sawicka J, Sariahmctoglu M, Schulze CJ, Scott PG, Szczesna-Cordary D, Schulz R. 2005. Degradation of myosin light chain in isolated rat hearts subjected to ischemia-reperfusion injury: a new intracellular target for matrix metalloproteinase-2. Circulation. 112(4): 544-552.

Wang W, Schulze CJ, Suarez-Pinzon WL, Dyck JR, Sawicki G, Schulz R. 2002. Intracellular action of matrix metalloproteinase-2 accounts for acute myocardial ischemia and reperfusion injury. Circulation.106(12): 1543-1549.

Westerfield M. 1995. The zebrafish book. A guide for the laboratory use of zebrafish (Danio rerio). 3rd edition. Eugene (OR): University of Oregon Press. 385 p.

Page 114: Good, better, best. Never let it rest. Until your good is ...

Curriculum Vitae

Candidate’s full name: Amina Mohammed Ahmed fallata

Universities attended (with dates and degrees obtained):

King Abdul-Aziz University (2009, B.Sc).

Conference Presentations:

1) Amina M. Fallata and Bryan D. Crawford, April 2014. Intracellular localization and regulation of Gelatinase A in zebrafish skeletal muscle. Graduate Research Conference (GRC), University of New Brunswick, Fredericton, NB, Canada. (Oral presentation).

2) Amina M. Fallata and Bryan D. Crawford, March 2014. Intracellular localization and regulation of Gelatinase A in zebrafish skeletal muscle. Canadian Zebrafish Conference, Mont-Temblant, QC, Canada. (Poster presentation).

3) Amina M. Fallata and Bryan D. Crawford, March 2014. Intracellular localization and regulation of Gelatinase A in zebrafish skeletal muscle. The 7th Canadian Developmental Biology, Mont-Temblant, QC, Canada. (Poster presentation).

4) Amina M. Fallata and Bryan D. Crawford, June 2013. Intracellular localization and regulation of Gelatinase A in zebrafish skeletal muscle. The 3rd North Atlantic Zebrafish Research Symposium, University of Maine, Orono, Maine, USA. (Poster presentation).

5) Amina M. Fallata and Bryan D. Crawford, July 2012. Intracellular localization and regulation of Gelatinase A in zebrafish skeletal muscle. Society for Developmental Biology 71st annual meeting, Montreal, QC, Canada. (Poster presentation).

6) Amina M. Fallata and Bryan D. Crawford, May 2012. Intracellular localization and regulation of Gelatinase A in zebrafish skeletal muscle. 51st Canadian Society of Zoology meeting, Sackville, NB, Canada. (Poster presentation).