control of cardiomyocyte proliferation by …...ii control of cardiomyocyte proliferation by...

187
Control of Cardiomyocyte Proliferation by p53/MDM2- Regulated microRNAs by Shanna Stanley-Hasnain A thesis submitted in conformity with the requirements for the degree of Masters of Science Medical Science Institute of Medical Science, Cardiovascular Collaborative Program University of Toronto © Copyright by Shanna Stanley-Hasnain 2016

Upload: others

Post on 29-Aug-2020

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

Control of Cardiomyocyte Proliferation by p53/MDM2-Regulated microRNAs

by

Shanna Stanley-Hasnain

A thesis submitted in conformity with the requirements for the degree of Masters of Science – Medical Science

Institute of Medical Science, Cardiovascular Collaborative Program University of Toronto

© Copyright by Shanna Stanley-Hasnain 2016

Page 2: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

ii

Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated

microRNAs

Shanna Stanley-Hasnain

Masters of Science – Medical Science

Institute of Medical Science, Cardiovascular Collaborative Program University of Toronto

2016

Abstract

Defining the roadblocks responsible for adult cardiomyocyte cell cycle arrest lies at the core of

developing cardiac regenerative therapies. Inactivation of the p53/MDM2 tumor suppressor

circuitry in the heart caused a significant increase in cardiomyocyte proliferation, through an

upregulation of factors involved in cell cycle re-entry. These factors may be regulated by

microRNAs (miRNAs), in quiescent cardiomyocytes. Therefore, we hypothesized that

inactivation of p53/MDM2-regulated miRNAs could promote the expression of cell cycle

activators and induce proliferation of cardiomyocytes. Comparison of miRNA expression

profiles from cardiac specific p53/MDM2 double knockout (DKO) mouse hearts and wild type

controls revealed 11 miRNAs that were downregulated in the "proliferative" DKO hearts and

enriched for mRNA targets involved in cell cycle regulation. Knockdown of these 11 miRNAs in

neonatal rat cardiomyocytes significantly increased the occurrence of cytokinesis, revealing a

novel subset of p53/MDM2-regulated miRNAs responsible for maintaining cell cycle arrest in

the heart.

Page 3: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

iii

Acknowledgments

Thank you to my beautiful friends and family for their unending support throughout my academic endeavors. Your confidence and belief in me has been my driving force throughout the last 6 years of my life when I began my undergraduate degree at the University of Toronto. I

have a few specific acknowledgments to individuals who have really helped shape the person I am over the course of my life and years studying.

Firstly. I’d like to thank Effie Argyropoulos, my roommate of 5 years, and best friend. She has been there for me every step of the way, through every all-nighter, and every stressful moment, since I began studying in Toronto. I am thankful to her for listening to me practice countless

presentations, and guide me through many important life decisions. She has been my rock, and everything I do in life, I do in hopes of making her proud.

Next, I want to acknowledge Jonah Chevrier, because without him, I would not be the person I am today, or have the love for science that I currently hold. We have been side by side throughout almost our entire post-secondary careers, and have managed to teach each other many

things, support each other and importantly, make some of the best memories I’ll ever have throughout this time. Thank you for the ~1,000s of hours we spent together at Gerstein, you

made them fun. I would not be the person I am today if I had not met you.

Next, I have to recognize Keith Dadson, the post-doc in the Billia Lab, for making every day an absolute treat to come into the lab. Keith was always there to talk and kept me motivated, happy

and in a great mood throughout my days in the lab. I am thankful that I was able to learn a great deal from him through his many years of wisdom and understanding of the field and of research

as a whole. On top of this, I am grateful for his kindness, generosity, and patience. He was always there to lend a helping hand, and to listen. His attitude towards research and life is something I hope to carry with me through the rest of my time. I’m also thankful that he was

around to make sure that I took the time to eat lunch, and for all our trips to Fast Fresh.

Thank you to all my girlfriends back in Ottawa, for always being there for me. I am blessed to know that I will always have them on my side, despite any distance between us. Their successes

push me to be a better person every day, and I am so lucky that we all ended up as friends at De La Salle, because everyone of us made each other a better person. When I look at us now, I am

proud that we have all made it to where we are, together.

I also need to thank Taras Lesiuk, for being the most wonderful distractio n imaginable. Next, are the summer students Amelia Fung and Shelly Chauhan, for always brightening up the lab!

Most importantly, thank you to Joanne Stanley and Sadiq Hasnain, my parents. They are the

reason that I was able to follow my dreams, and the number one reason for where I am today. Nothing would be possible without them. Other important individuals are my step father Klaas Van Weringh and my father’s partner Sasha Sadilova. Thank you for taking care of my family

while I have been away, and for raising me along side of my parents. Your support and love means the world to me.

Lastly, thank you for taking the time to read my thesis thus far.

Page 4: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

iv

Contributions

Dr. Phyllis Billia, my mentor, and supervisor contributed holistically to this project through her undying support, insight and wisdom in my research endeavors and in my life as a whole. She helped to carefully craft a unique and exciting research project that I was passionate and excited

about, and was there to guide me every step of the way throughout my thesis. She supported me throughout all the experiment planning, trouble shooting, talk giving, poster presenting and

scholarship applications, as well as in going to conferences and participating in extracurricular activities. Furthermore, she guided me through the thesis writing process and providing me extremely valuable insight and thorough edits. It is thanks to her that I was able to experience a

well-rounded, inspiring and successful two year Master’s degree. I am eternally grateful that she believed in me, and gave me a shot in her lab, which turned out to be two of the best years of my

life. Phyllis is a role model to me, and much of my efforts in the future will be to make her proud of the scientist that blossomed under her supervision, guidance and mentorship.

Daniela Grothe, our incredible lab manager, taught me everything I needed to know about

working in the lab. She trained me exhaustively in all the techniques that I would require for the experiments in my thesis in an incomparably in-depth manner. Working along her side was a

once in a life time opportunity to learn the practice of meticulous lab bench work at a level that I have never seen before. She taught me the importance of trouble shooting and always using logic to make sure that what you are doing actually makes sense, as opposed to blindly following

protocols. She also taught me that it is always of benefit to try new things, and never to remain stuck in old methods and techniques, because the field of science is always advancing. Most

importantly, she was there for me every step of the way in all the experiments I conducted throughout my thesis, to either help me directly in the lab, answer my questions (there were a lot), or talk out and rationalize the upcoming experiments until I was 100%n confident that I

knew what I was doing. From RNA isolation, to spending hours at the microscope, she was always there to lend a helping hand. Together, we established and worked on many protocols that the lab now uses on a regular basis, including microRNA RT-qPCR, immunoprecipitations

and cell transfections. I am grateful for the weekends she sacrificed to come in and help me with countless cardiomyocyte isolations, especially on the ones where I was away and completely

relying on her for cells to keep my project moving. It is with her support and wisdom that a large majority of my experiments were successful, and that any complications I encountered, we were able to easily fix and surmount. I am forever grateful to everything she has taught me and done

for me over the past two years, and know that there is no way that my thesis could have turned out the way it did without her by my side. Finally, and maybe most importantly, she has been

there for me and a friend and confident through two very important years of my life. I will never forget all the hours we spent together in the lab, singing, laughing, dancing and talking. Her positive attitude and energy made it an absolute joy to work along her side, and made my two

years in the Billia lab fly by (almost too quickly).

Ludger Hauck, the Research Associate in the Billia lab has also been a major support throughout

my Master’s degree. He helped me countless times to plan out experiments, trouble shoot, and understand how to critically analyze my data. His brilliant mind helped guide and expand my project, and I could always count on him when I was stuck and didn’t know in what direction to

turn. His passion for high quality research and thinking outside of the status quo of a given field has shaped the kind of researcher that I now am. I am extremely grateful to him for his wisdom,

and the many hours he sacrificed to help me work through my project and understand my results,

Page 5: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

v

as well as the weekends he gave up to help with my cardiomyocyte isolations. His patience and ability to trouble shoot with the help of Dani were the factors that allowed us to finally perfect

our cardiomyocyte isolation technique. On top of this, I am extremely thankful for the time he took to read my thesis from front to back, and provide me with extensive suggestions and edits

that largely improved my work. Finally, I am thankful that he believed in me, and in my abilit ies to execute this project. I am overjoyed to see how proud he is in the scientist that I have become over the past two years, thanks to him and the other members of the Billia lab.

My program advisory committee members Dr. Anthony Gramolini and Dr. Vivek Rao, for the support, wisdom and knowledge they have provided with me throughout my Master’s. Together,

they pushed me to be a better critic of my own work and guided my project, ensuring that my goals were feasible. I am forever grateful for the time they have sacrificed to push me as a young scientist and for all their advice and insight that they provided following my PAC meetings in

terms of future experiments, as well as for my thesis.

Finally, thank you to Cedric Manlhiot, for your help with statistics. I greatly appreciate you

taking the time out of your day to set me in the right direction regarding in my statistic calculations for RT-qPCR.

Page 6: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

vi

Table of Contents

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

Contributions................................................................................................................................... iv

Table of Contents ............................................................................................................................ vi

List of Tables ................................................................................................................................... x

List of Figures ................................................................................................................................. xi

List of Abbreviations .....................................................................................................................xiii

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

1 Literature Review ........................................................................................................................1

1.1 Anatomy...............................................................................................................................1

1.2 Diastolic and Systolic Performance .....................................................................................1

1.3 Cardiomyocyte Structure and Function ...............................................................................2

1.4 Cardiovascular Disease ........................................................................................................4

1.4.1 General Overview ....................................................................................................4

1.4.2 Ischemic Heart Disease ............................................................................................5

1.4.3 Myocardial Infarction ..............................................................................................5

1.4.4 Heart Failure ............................................................................................................7

1.4.5 Ventricular Remodeling ...........................................................................................8

1.4.5.1 Cardiomyocyte Hypertrophy .....................................................................9

1.4.5.2 Scar Formation ..........................................................................................9

1.4.6 Current Treatments for Ischemic Heart Disease and Heart Failure .........................9

1.5 Cell Cycle Regulation ........................................................................................................10

1.5.1 The Cell Cycle .......................................................................................................10

1.5.2 G1 Phase ................................................................................................................12

1.5.3 S Phase ...................................................................................................................13

1.5.4 G2 Phase ................................................................................................................15

Page 7: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

vii

1.5.5 M Phase..................................................................................................................16

1.6 Cardiogenesis and Cardiomyocyte Cell Cycle Regulation ................................................20

1.6.1 Cardiogenesis .........................................................................................................20

1.6.2 Cardiomyocyte Regeneration.................................................................................21

1.7 MicroRNAs ........................................................................................................................22

1.7.1 miRNA Nomenclature ...........................................................................................23

1.7.2 miRNA Biogenesis ................................................................................................23

1.7.3 miRNA Mechanism and Function .........................................................................25

1.7.4 MicroRNAs and the Heart .....................................................................................27

1.7.4.1 MicroRNAs, Cardiac Development and Homeostasis ............................27

1.7.4.2 MicroRNAs and cardiovascular disease..................................................29

1.7.5 MicroRNA Therapeutics........................................................................................31

1.7.5.1 Delivery Methods for miRNA Based Therapies .....................................31

1.7.5.2 miRNA Mimics .......................................................................................32

1.7.5.3 miRNA Antagomirs.................................................................................33

1.7.6 MicroRNAs and the cell cycle ...............................................................................34

1.8 Tumor Suppressors – A Focus on p53 ...............................................................................36

1.8.1 The p53/Mdm2 Tumor Suppressor Circuitry ........................................................38

1.8.2 Canonical p53 Pathways ........................................................................................39

1.8.3 Non-Canonical p53 Pathways ................................................................................40

1.8.4 Post-Translational Regulation of p53 by MDM2 ..................................................41

1.8.5 p53/MDM2 and the Heart ......................................................................................42

1.8.6 p53 and miRNAs....................................................................................................45

Chapter 2 Rational Hypothesis ......................................................................................................50

2 Hypothesis and Thesis Aims .....................................................................................................50

2.1 Hypothesis..........................................................................................................................51

Page 8: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

viii

2.2 Thesis Aims........................................................................................................................51

Chapter 3 Data Chapter..................................................................................................................52

3 Control of Cardiomyocyte Proliferation by p53/MDM2-regulated microRNAs......................52

3.1 Introduction ........................................................................................................................53

3.2 Methods..............................................................................................................................57

3.2.1 MDM2 and p53 conditional mutant mice ..............................................................57

3.2.2 RNA Isolation ........................................................................................................58

3.2.3 Nanostring nCounter miRNA assay.......................................................................59

3.2.4 miRNA RT-qPCR ..................................................................................................59

3.2.5 Western Blot ..........................................................................................................60

3.2.6 Neonatal rat cardiomyocyte and fibroblast isolation and culture ..........................60

3.2.7 miRNA antagomir development ............................................................................60

3.2.8 Neonatal rat cardiomyocyte transfection ...............................................................61

3.2.9 Cell Cycle RT-qPCR Array ...................................................................................61

3.2.10 Immunofluorescence ..............................................................................................61

3.2.11 Statistical Analyses ................................................................................................62

3.3 Results ................................................................................................................................63

3.3.1 The tumor suppressor circuitry p53/MDM2 regulate expression of a unique subset of miRNAs within the heart. .......................................................................63

3.3.2 p53/MDM2-regulated miRNAs are enriched for mRNA gene targets involved in cell cycle progression.........................................................................................67

3.3.3 p53/MDM2-regulated miRNAs are novel cardiomyocyte cell cycle inhibitors ....71

3.3.4 p53/MDM2-regulated miRNAs inhibit cardiomyocyte proliferation by downregulating target genes involved in the progression of all 4 phases of the

cell cycle. ...............................................................................................................78

3.4 Discussion ..........................................................................................................................81

3.4.1 p53 and MDM2 regulate the expression of muscle specific miRNAs in the

heart........................................................................................................................81

Page 9: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

ix

3.4.2 p53 and MDM2 maintain cardiomyocyte quiescence through the regulation of a unique group “anti-proliferative” miRNAs. ........................................................82

3.4.3 p53/MDM2-regulated miRNAs maintain cardiomyocyte cell cycle arrest through the inhibition of multiple cell cycle regulators. ........................................84

3.5 Supplementary Figures and Tables ....................................................................................87

Chapter 4 Conclusion.....................................................................................................................96

4 Conclusion and Future Directions.............................................................................................96

4.1 General Discussion ............................................................................................................96

4.1.1 Thesis summary .....................................................................................................96

4.1.2 The 11 p53/MDM2-regulated miRNAs are well characterized tumor suppressors .............................................................................................................98

4.1.2.1 The miR-30 family ..................................................................................98

4.1.2.2 The let-7 family .......................................................................................98

4.1.2.3 miR-181a .................................................................................................99

4.1.2.4 miR-26b ...................................................................................................99

4.1.2.5 miR-204 ...................................................................................................99

4.1.2.6 miR-149 .................................................................................................100

4.1.2.7 miR-194 .................................................................................................100

4.1.3 Tumor suppressive p53/MDM2-regulated miRNAs may function

synergistically to promote cardiac cell cycle arrest .............................................101

4.1.4 Mechanisms of cardiac regeneration with a therapeutic goal in mind .................102

4.1.4.1 Cell-based cardiac regenerative therapies .............................................102

4.2 Limitations and Future Directions ...................................................................................109

4.3 Overall Conclusion ..........................................................................................................114

5 References ...............................................................................................................................116

Copyright Acknowledgements.....................................................................................................136

Permissions granted for use of external figures: ..............................................................136

Page 10: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

x

List of Tables

Table 3.1 - miRNAs significantly downregulated in DKO vs. vehicle control…………………66

Table 3.2 - Final list of p53/MDM2-regulated miRNAs……….……..………………………...70

Supplementary Table 3.1 - Normalized RT-qPCR Ct values for miRNA expression levels

following 11 antagomir transfection……………………………………………………………..95

Page 11: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

xi

List of Figures

Figure 1.1 - Anatomy of the cardiac sarcomere…………………………………………………..3

Figure 1.2 - The main molecular proponents of myocardial ischemia reperfusion injury………..7

Figure 1.3 - The major events of the cell cycle are regulated by transitions between CDK/cyclin

complexes…………………………………………………………………………………..……11

Figure 1.4 - Assembly of the pre-replicative and pre-loading complex………………...………14

Figure 1.5 - The main stages of mitosis………………………………………………………....16

Figure 1.6 – Aurora B Localization During Mitosis and Cytokinesis…………………………..19

Figure 1.7 - The Canonical Pathway of miRNA Biogenesis……………………………………25

Figure 1.8 - An overview to cell cycle control by microRNAs………………………………....35

Figure 1.9 - Regulation of the cell cycle by p53-induced miRNAs……………………………..47

Figure 3.1 - Significant miRNA transcriptional changes in the absence of p53 and/or MDM2

create unique miRNA profiles…………………………………………………………………...64

Figure 3.2 - miRNAs downregulated in the DKO are enriched for target genes within the cell

cycle pathway…………………………………………………………………………………....67

Figure 3.3 - Genes within the cell cycle pathway are redundantly targeted by the miRNAs

downregulated in the DKO……………………………………………………………………....69

Figure 3.4 - Antagomirs are specific between miRNA families but redundant within miRNA

families…………………………………………………………………………………………...72

Figure 3.5 - Inhibition of 11 target miRNAs by antagomir cocktail upregulates cardiomyocyte

cytokinesis………………………………………………………………………………………..74

Figure 3.6 - Treatment of cardiomyocytes with cocktail of 11 antagomirs promotes

cardiomyocyte cytokinesis……………………………………………………………………….77

Figure 3.7 - Inhibition of 11 target miRNAs causes upregulation of many miRNA target genes

within the cell cycle pathway…………………………………………………………………....80

Supplemental Figure 3.1 - Validation of cardiac specific MDM2 and p53 knock-down

following Tamoxifen injection …………………………………………………………………..87

Supplemental Figure 3.2. Biological pathway analysis for the 11 p53/MDM2-regulated

miRNAs………………………………………………………………………………………….88

Supplemental Figure 3.3 - 11 miRNAs hits are downregulated in cardiac fibroblasts………...89

Supplemental Figure 3.4 - Efficient transfection of neonatal rat cardiomyocytes with red

Page 12: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

xii

fluorescent positive control……………………………………………………………………...90

Supplemental Figure 3.5 - Raw count data for cytokinesis events following antagomir

treatment………………………………………………………………………………………...91

Supplemental Figure 3.6 - Average Cytokinetic events and average RFU show similar %

change trends in transfected cardiomyocytes…………………………………………………...92

Supplemental Figure 3.7 - RT-qPCR for cardiomyocyte housekeeping genes………………..93

Supplemental Figure 3.8 - MirSystem miRNA target gene biological pathway ranking score.94

Page 13: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

xiii

List of Abbreviations

ACE Angiotensin-converting enzyme

AGO Argonaute

AIP1 Actin-interacting protein 1

AKT AKT serine/threonine kinase

AMPK 5' AMP-activated protein kinase

ANOVA Analysis of variance

APAF1 Apoptotic peptidase activating factor 1

APC Anaphase promoting complex

APC Adenomatous polyposis coli

ARF Alternate reading frame protein

ATM Ataxia telangiectasia mutated

ATP Adenosine triphosphate

APAF1 Apoptotic peptidase activating factor 1

BCL2 B-cell lymphoma 2

BIN1 Bridging integrator 1

BRCA1 Breast cancer 1

BRMS Breast cancer metastasis suppressor 1

BAX BCL2 Associated X Protein

CDK Cyclin-dependent kinase

CKI Cyclin-dependent kinase inhibitor

C-kit KIT Proto-Oncogene Receptor Tyrosine Kinase

CMG Cdc45-MCM-GINS

Ca2+ Calcium

CDC Cell division cycle

Page 14: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

xiv

CDH1 Cadherin 1

CRISPR Clustered regularly interspaced short palindromic repeats

DAPI 4',6-diamidino-2-phenylindole

DDC2 DNA damage checkpoint protein 2

DDK Dbf4-dependent Cdc7 kinase

DDX5 DEAD-Box helicase 5

DGCR8 DiGeorge Syndrome critical region gene 8

DKO Double knockout

DR5 Death receptor 5

DP-1 E2f dimerization partner 1 / Transcription factor Dp-1

Drosha Drosha ribonuclease III

MED23 Mediator complex subunit 23

MEF2A Myocyte enhancer factor 2A

MEF2C Myocyte enhancer factor 2C

DBF4 Dbf4 Zinc finger

DPB11 DNA replication regulator Dpb11

eIf4E Eukaryotic translation initiation factor 4E

eIF6 Eukaryotic translation initiation factor 6

E2F E2 Factor

EF Ejection fraction

ERBB2/4 Erb-b2 receptor tyrosine kinase 2/4

ERK Extracellular signal-regulated kinase

EDE1 EH domain-containing and endocytosis protein 1

EI24 Etoposide induced 2.4/Autophagy associated transmembrane protein

FA Franconi anemia related tumor suppressor

FGF Fibroblast growth factor

Page 15: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

xv

G1 Gap phase 1

G2 Gap phase 2

GINS Go Ichi Nii and San complex

GADD45A Growth arrest and DNA damage inducible alpha

GATA4 GATA Binding Protein 4

GlUT1/4 Glucose transporter 1/4

GTP Guanosine-5'-triphosphate

HIF-1 Hypoxia-inducible factor 1-alpha

INN1 Ingression protein 1

iPSC Induced pluripotent stem cell

ISL1 ISL LIM Homeobox 1

IRS2 Insulin receptor substrate 2

KCND2 Potassium voltage-gated channel subfamily D member 2

Ki-67 Marker of proliferation Ki-67

KRAS KRAS proto-oncogene GTPase

LNA Locked nucleic acid

LV Left ventricle

MAD2L1 MAD2 mitotic arrest deficient- like 1

MAPK Mitogen-activated protein kinase

MLH1 MutL Homolog 1

MPTP Mitochondrial permeability transition pore

MSH2 MutS Homolog 2

Mcm Tamoxifen inducible Cre recombinase (MerCreMer)

MCM2-7 Minichromosome maintenance proteins 2-7

MDM2 Murine double minute-2

Mdm2KO Murine double minute-2 knockout

Page 16: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

xvi

MDM4 Mouse double minute 4 / Mdm2-like p53-binding protein

miRNA microRNA

mTOR Mammalian/mechanistic target of rapamycin

MULE MCL-1 ubiquitin ligase E3

MYC V-Myc avian myelocytomatosis viral oncogene homolog

Na+ Sodium

NBS1 Nisbrin

NFAT Nuclear factor of activated T-cells

NPT Nuclear protein, ataxia-telangiectasia locus

NET1 Neuroepithelial cell transforming 1

NKX2.5 NK2 homeobox 5

NOXA Phorbol-12-Myristate-13-Acetate-Induced protein 1

NRG1 Neuregulin 1

OACIS Osaka Acute Coronary Insufficiency Study

PCNA Proliferating cell nuclear antigen

PGC1β PPARG coactivator 1 beta

PIK3CB Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit beta

PI3K Phosphatidylinositide 3-kinase

PTEN Phosphatase and tensin homolog

PERP TP53 apoptosis effector

PIDD1 P53-Induced death domain protein 1

PiH3 Phosphorylated histone H3

PTPRV Protein tyrosine phosphatase, receptor type V, Pseudogene

PUMA P53 up-regulated modulator of apoptosis

p107 Retinoblastoma-like protein 1

p130 Retinoblastoma-like protein 2

Page 17: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

xvii

p21Cip1 Cyclin-dependent kinase inhibitor 1A

p27Kip1 Cyclin-dependent kinase inhibitor 1B

p38 MAPK p38 mitogen-activated protein kinase

p53AIP1 Tumor protein P53 regulated apoptosis inducing protein 1

p53KO p53 knockout

PCR Polymerase chain reaction

RAB22A RAB22A, member RAS oncogene family

RAD21 RAD21 cohesin complex component

RalA RALA Ras like proto-oncogene A

RB Retinoblastoma

RELA RELA proto-oncogene, NF-kB subunit

RISC RNA-induced silencing complex

RIZ1 Retinoblastoma protein-interacting zinc finger protein

RFU Relative fluorescence units

RhoA Ras homolog family member A

ROCK Rho associated coiled-coil containing protein kinase 1

SA1/STAG1 Stromal antigen 1

SA2/STAG2 Stromal antigen 2

SCA1 Ataxin-1

SERCA2 ATPase sarcoplasmic/endoplasmic reticulum Ca2+ transporting 2

SIRT1 Sirtuin 1

SSSK2 S-phase kinase-associated protein 2, E3 ubiquitin protein ligase

SNAI1 Snail family transcriptional repressor 1

SOX4 SRY-box 4

SR Sarcoplasmic reticulum

SLD2/3/7 DNA replication regulator Sld2/3/7

Page 18: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

xviii

SMC1/SMC3 Structural maintenance of chromosomes 1/3

TAC Trans-aortic constriction

TBX5 T-box protein 5

TIGAR TP53 Induced glycolysis regulatory phosphatase

TIMP1 Tissue inhibitor of metalloproteinases 1

TRAF6 Tumor necrosis factor receptor associated factor 6

Tam 4-Hydroxytamoxifen

UTR Un-translated region

WNT Wingless-type MMTV integration site family

XPC XPC complex subunit, DNA damage recognition and repair factor

YAP1 Yes associated protein 1

YWHAS tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein sigma

/ Stratifin

Page 19: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

1

Chapter 1

Introduction

1 Literature Review

1.1 Anatomy

The structure and function of the heart, like in any organ, are inseparable, both in health and

disease. Accordingly, Kresh and Armour advocated that the “heart should be considered as self-

regulating functional system, which is greater than the sum of its constitutive parts”1. This infers

that the loss of structural integrity in one area of the heart can have devastating effects on heart

function as a whole.

The heart pumps on average 5 L of blood a minute2, requiring tight synergy of its electrical and

mechanical function. It is composed of four chambers; the smaller left and right atria, which

receive the incoming blood, and the larger left and right ventricles, which send the blood to be

oxygenated by the lungs, and to the rest of the body, respective ly. The ventricular myocardial

fibers are organized in a helicoid structure that form two spiral turns, known as the ventricular

myocardial band3. It is this configuration that allows the heart to perform the dual function of

both ejection and suctioning blood3. In other words, the intricate organization of the heart is

fundamental for its proper role as a muscular pump that functions to supply oxygen and

metabolites to the peripheral tissues of the body. This tightly regulated process of contraction

and relaxation that drive blood flow throughout the body is called the cardiac cycle. Diastole is

the portion of the cardiac cycle where the heart refills with blood following systole, the portion

of the cycle primarily involved in contraction.

1.2 Diastolic and Systolic Performance

Systolic performance is the ability of the ventricles to empty. This is influenced by myocardial

contractile function, ventricular load, and ventricular configuration4. As the systolic performance

of the left ventricle (LV) is required for supplementing the entire body with blood, it is generally

of main focus. The ability of the LV to empty can be calculated as the LV emptying fraction or

ejection fraction (stroke volume divided by end-diastolic volume).4 Consequently, impaired LV

emptying, indicative of systolic dysfunction, can be measured by a decrease in ejection fraction

Page 20: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

2

(<50%)5. When studying murine cardiac contractility however, fractional shortening, which

determines the change in ventricular diameter between the contracted and relaxed state (%

Fractional Shortening = LV end-diastolic diameter – LV end-systolic diameter / LV end-diastolic

diameter x 100) is measured instead of ejection fraction6. A fractional shortening less than 30%

is considered impaired diastolic performance6.

As an effective pump, the ventricles must not only eject but also fill with blood. Effectively,

normal LV diastolic function is defined as its sufficient filling to produce a cardiac output

proportionate to the body’s requirements4. Thus, to maintain an adequate cardiac output when

LV ejection fraction is decreased, a larger end-diastolic volume is required, indicating that the

amount of filling required is dependent on LV systolic performance5. The LV fills in diastole in

response to the pressure gradient from the left atrium to the LV. This occurs two times during the

cardiac cycle: early in diastole after mitral valve opening and late in diastole during atrial systole.

Most of LV filling occurs early in diastole, and less than 25% of the LV stroke volume enters the

LV during atrial systole. The rate of early LV filling pressure is determined by two factors: the

rate of LV relaxation and left atrial pressure at the time of mitral valve opening4.

1.3 Cardiomyocyte Structure and Function

Cardiomyocytes, the contracting functional cellular unit of the heart, occupy an average of 70%

of the ventricular wall under normal physiological conditions, making them the major structural

component of the myocardium7. In the adult, 80% of these long ellipsoidal cells are binucleated,

and are interconnected by gap junctions to allow for coordinated contractile activity8,9,10.

Cardiomyocytes also form close interactions with the extracellular matrix, primarily composed

of fibroblasts, endothelial cells and vascular smooth muscle cells to form a complex 3D network

of cells9.

Intracellularly, the sarcomere forms the basic contractile unit of the cardiomyocyte, whereby

repeating molecules of actin (thin filaments) and myosin (thick filaments) are the building blocks

of this structure (Figure 1.1)9. The sarcomere itself consists of ~20 proteins, but forms

connections with many others linking the myocytes and their extracellular matrix to regulate

contractility9. One sarcomere unit is defined as a segment between two Z-discs (anchoring plane

that cross-links thin filaments from opposing sarcomere halves through a lattice formed by

Page 21: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

3

α-actinin and many other proteins)9. The thick filaments sit in between two thin filaments, in the

middle of the sarcomere, and are linked to the Z-discs by the protein titin11. The Z-disc is thought

to be crucial in facilitating cardiomyocyte response to mechanical stress by acting as the liaison

between the sarcomeres and the extracellular matrix.

At the level of the individual cardiomyocyte, contraction is coordinated by the flux of calcium

(Ca2+) into and out of the cell and the sarcoplasmic reticulum through spatially defined ion

channels and exchangers12. Systole will occur when an action potential is fired from pacemaker

cells within sinoatrial node of the heart12. These specialized non-contractile cardiomyocytes

generate repetitive action potentials and dictate the heart rate12. The action potential will first

spread throughout the atria and then to the ventricles leading to depolarization of the

cardiomyocytes12. Depolarization causes voltage gated Ca2+ channels within the sarcolemma to

open and allow influx of Ca2+ into the cytoplasm12. The build-up of Ca2+ causes ryanodine

receptors within the sarcoplasmic reticulum (SR) to release more Ca2+ from the SR into the

cytoplasm (Ca2+ induced Ca2+ release)11. Cytoplasmic Ca2+ will interact with troponin C of the

troponin complex (major thin filament proteins), resulting in a conformational change in the

Figure 1.1 Anatomy of the cardiac sarcomere9. (A) Diagram of the basic organization of the sarcomere.

(B) Representation of the major proteins of the sarcomere. Originally published by Harvey, P. A. &

Leinwand, L. A. J. Cell Biol. 194, 355–65 (2011)9. Used with permission of the author and publisher.

Page 22: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

4

position of tropomyosin on the thin filament12. This conformational change reveals sites where

actin and myosin filaments can interact to form cross-bridges, and results in the shortening of the

myofilaments and force generation12. Diastole occurs when Ca2+ is removed from the cytosol

through SR re-uptake by the Ca+ transporter (SERCA2) and extrusion through the sarcolemma

by the Na+/Ca2+ exchanger12. Removal of Ca2+ causes tropomyosin to resume its original

configuration resulting in cross bridge detachment11. Coordination of this response between

individual cardiomyocytes to allow for synchronized contraction is dependent on the cardiac

conduction system and direct electrical contact between the myocytes11. Highlighting the

importance of the precise regulation of the cardiac cycle, dysfunction or mutation of almost any

protein involved in this process has been shown to induce or increase the risk of heart failure.

1.4 Cardiovascular Disease

1.4.1 General Overview

Cardiovascular diseases are the leading cause of death worldwide despite many advances in

pharmaceutical treatments and surgical procedures13. Cardiovascular disease is an overarching

term used to describe groups of ailments of the heart or blood vessels. The most common

cardiovascular diseases are: hypertensive heart disease caused by increased blood pressure13,

ischemic heart disease elicited by the narrowing of the coronary arteries14 and cerebrovascular

disease - pertaining to blockages of the blood vessels in the brain15. Cardiovascular disease poses

a significant burden on health care systems globally as they are the primary cause of death.

Importantly, it is known that adopting healthy life style changes such as smoking cessation,

maintaining a balanced diet and increasing physical activity, all of which help to lower blood

pressure (the primary risk factor for cardiovascular disease) on top of their own individual

benefits, can postpone the onset of heart disease by up to 14 years16. It was estimated that in

2011 one person died every 7 minutes from heart disease or stroke (66,000 Canadians) and that

1.3 million were living with heart disease16. This costs the Canadian economy more than $20.9

billion a year, accounting for a significant burden on health care services, lost wages and

decreased productivity16.

Page 23: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

5

1.4.2 Ischemic Heart Disease

Ischemic heart disease (otherwise known as coronary artery disease) is a major public health

problem, as it is the most common cause of death in most western countries17. Ischemic heart

disease manifests itself as ischemia as a result of restricted blood supply to the heart tissue

followed by angina (substernal pains/discomfort caused by reduced blood flow to the heart). This

can lead to myocardial infarction, arrhythmias, heart failure, and sudden death if left untreated17.

The most common cause of ischemic heart disease is atherosclerosis, a condition in which

plaques of fatty material are deposited on the inner walls of arteries. Build-up of atherosclerotic

plaques in the coronary artery system can lead to reduced blood flow and, consequently, reduced

oxygen transportation to areas of the heart. The mismatch between supply and demand for

oxygen results in myocardial ischemia18. Chronic oxygen supply-demand imbalance will have

important myocardial consequences over time. At the cellular level, cardiac remodeling

(hypertrophy), and cardiomyocyte autophagy (catabolic degradation of organelles and

macromolecules within the cell) are known to occur during chronic ischemic heart disease19,20.

Acutely, if an atherosclerotic plaque ruptures, exposure of its fatty core provides a potent

substrate for activation of the coagulation cascade21. Coagulation cascade activation leads to the

formation of a platelet-rich thrombus (blood clot) which can grow to the point of vessel

occlusion21. Alternatively, a thromboembolism may occur, whereby the blood clot breaks loose

from the vessel and travels through the circulatory system, risking occlusion of a distal blood

vessel21. The result of complete coronary artery occlusion is acute myocardial infarction21.

1.4.3 Myocardial Infarction

Myocardial infarction afflicts 7.6 million Americans each year, increasing an individual’s

susceptibility to a subsequent myocardial infarction, and often precedes adverse cardiac

remodeling, hypertrophy and heart failure22. A myocardial infarction occurs when blood flow to

an area of the heart is reduced to a critical level whereby normal cellular function can no longer

occur due to lack of oxygen23. A series of biochemical changes in the myocardium ensue with

lack of oxygen; halted oxidative phosphorylation leading to mitochondrial membrane

depolarization, ATP depletion, and subsequently, inhibition of cardiomyocyte contractile

function23. Cellular metabolism switches to anaerobic glycolysis, resulting in a buildup of lactate

Page 24: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

6

and a reduction in intracellular pH23. The acidic intracellular environment eventually leads to

Na+ and Ca2+ overload within the cell23. If blood flow is not restored (reperfusion), and

ischemia persists for a prolonged period of time, irreparable myocyte damage and/or cell death

can occur23. Coronary artery occlusion due to a thrombus superimposed on a ruptured or unstable

atherosclerotic plaque, is the most common cause for myocardial infarction23. Six primary risk

factors for myocardial infarction and coronary artery disease have been identified:

hyperlipidemia, diabetes mellitus, hypertension, tobacco use, male gender, and family history of

atherosclerosis.

Cell death primarily occurs in the area of the heart most distal to the blocked arterial blood

supply, the endocardium24. Over the duration of the occlusion, the area of cell death will enlarge

into the myocardium and finally the epicardium24. The magnitude of a myocardial infarction is

defined by the extent of myocardial cell death24. Removing the occlusion and restoring blood

flow to the ischemic area of the heart as quickly as possible is essential to salvage the viable

myocardium24. However, reperfusion itself can also induce cardiomyocyte death, known as

myocardial-reperfusion injury23. This significantly reduces the efficiency of reperfusion as a

treatment for myocardial infarction, making myocardial reperfusion injury an important

therapeutic target. The major mediators of reperfusion injury are oxidative stress, intracellular

Ca2+ overload, rapid pH restoration causing hypercontraction (reperfusion with an acidic buffer

in an animal model of ischemia reperfusion injury can reduce myocardial infarction size25),

mitochondrial permeability transition pore (MPTP) opening (blocking MPTP opening during

reperfusion is associated with greater post ischemic recovery26) and inflammation23 (Figure 1.2).

All of these stimulators of myocardial reperfusion injury seem to occur within the first few

minutes following reperfusion23. However, processes such as apoptosis and inflammation

initiated at this time, as well as during ischemia, will continue to occur over several hours or

even days following reperfusion23. Halting these processes to minimize the extent of

cardiomyocyte cell death is another important therapeutic option to be considered, as the extent

of cardiomyocyte cell death during ischemia and reperfusion is an important risk factor for the

development of heart failure following myocardial infarction. The severity of cardiomyocyte

loss, and lack of regenerative capacity of the heart is highlighted by the fact that 33% of

myocardial infarction survivors will develop heart failure over time24.

Page 25: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

7

.

1.4.4 Heart Failure

Heart failure, a disease affecting over 5 million North Americans, occurs when the heart can no

longer generate adequate contractile force to sustain the required cardiac output of an individual

at normal filling pressures8. It is the culminating pathway for numerous diseases that affect the

heart. Irreversible damage to the heart muscle caused by myocardial infarction is the most

common risk factor for heart failure, followed by hypertension27. Furthermore, the Framingham

Heart Study suggests that coronary artery disease is the most common cause of heart failure,

albeit coronary artery disease comes often hand in hand with hypertension and elevated lipids27.

pC

Figure 1.2 The main molecular proponents of myocardial ischemia reperfusion injury23. During acute

myocardial infarction, the cell metabolism switches to anaerobic respiration, leading to the buildup of lactate and

a drop in intracellular pH. The Na+-H+ exchanger will consequently extrude H+, resulting in intracellular

Na+ overload. This activates the Na+-Ca2+ exchanger to function in reverse to extrude Na+ and leads to intracellular

Ca2+ overload. During reperfusion, the electron transport chain is reactivated, generating ROS, which mediates

myocardial reperfusion injury by inducing the opening of the MPTP, acting as a neutrophil chemoattractant, and

mediating dysfunction of the sarcoplasmic reticulum. Reperfusion and reactivation of the Na+-H+ exchanger result

in washout of lactic acid, resulting in the rapid restoration of physiological pH, which releases the inhibitory effect

on MPTP opening and cardiomyocyte contraction. The restoration of the mitochondrial membrane potential drives

calcium into the mitochondria, which can also induce MPTP opening. Originally published by Hausenloy, D. J. &

Yellon, D. M. J. Clin. Invest. 123, 92–100 (2013)23. Used with permission of the author and publisher.

Page 26: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

8

Heart failure is a clinical syndrome that can arise from diverse causes, and is a results of a

complex interplay of structural and functional abnormalities. As such, heart failure has been

described as a cardio-renal disorder causing excessive salt and water retention, a hemodynamic

disorder with peripheral vasoconstriction and reduced cardiac output, a hormonal disorder with

the activation of the renin-angiotensin-aldosterone system and adrenergic nervous system, an

inflammatory syndrome characterized by increased circulating and local inflammatory cytokines,

or a direct myocardial disorder instigated by injury to the heart followed by pathological

ventricular remodeling24. Yet, these factors are not mutually exclusive and often many of them

combined cause the onset and progression of heart failure28. The causative injury can either be

acute and obvious (e.g. acute myocardial infarction), or stealthy and asymptomatic (e.g. chronic

hypertension)27. Following this, a series of compensatory mechanisms are initiated, but

eventually these mechanisms will be detrimental to the heart, resulting in heart failiure28.

1.4.5 Ventricular Remodeling

Left ventricular remodeling is a process by which neurohormonal, mechanical and genetic

factors alter ventricular function, shape and size29. This process occurs following numerous types

of injury and can continue with the onset and progression of heart failure. It is an adaptive

process during development, but becomes pathological following injuries such as myocardial

infarction, cardiomyopathy, hypertension or valvular disease30. As myocardial infarction is the

injury of focus in this thesis, ventricular remodeling will be overviewed in this context.

The acute loss of myocardium following myocardial infarction causes a sudden increase in

cardiac loading conditions which produce a unique pattern of cardiac remodeling in the infarct

border zone as well as in the distant non-infarcted myocardium30. This increase in cardiac

loading triggers a cascade of intracellular signaling pathways that initiate reparative changes

such as hypertrophy, dilatation, and collagen deposition to form a scar30. Injury caused by

myocardial infarction and reperfusion also result in the migration of immune cells such as

macrophages and neutrophils into the infarct zone30. These cells will instigate intracellular

inflammatory signaling and neurohormonal activation, both of which play an important role in

remodeling30. The remodeling process has been divided into an early (within 72 hours) phase

Page 27: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

9

characterized by infarct expansion, and late (beyond 72 hours) phase associated with more global

ventricular dilatation and hypertrophy29,30.

1.4.5.1 Cardiomyocyte Hypertrophy

Cardiomyocyte hypertrophy is initiated during early remodeling and continues throughout the

late remodeling process. It can be visualized by microscopy, whereby up to a 70% increase in

myocyte size has been noted30. It is instigated by the neurohormonal activation and

cardiomyocyte stretching that occurs during the remodeling process. Primarily, it functions as an

adaptive process to offset increased load, and compensate for the lost cardiomyocytes, but

ultimately cardiomyocytes hypertrophy can become detrimental if not halted30. The mitogen-

activated protein kinase (MAPK) cascade activation via diverse growth factors (insulin- like

growth factor, fibroblast growth factor, epidermal growth factor, insulin, platelet derived growth

factor, etc.) is the major molecular executor of transcriptional and morphological changes that

occur during cardiomyocytes hypertrophy30.

1.4.5.2 Scar Formation

Myofibroblasts are responsible for reconstruction of the collagen network and fibrotic scar post-

myocardial infarction30. This reparative process is triggered by many stimuli including cytokines

released from necrotic/apoptotic or injured cardiomyocytes (such as transforming growth factor-

β1), hormonal activity (Angiotensin II), fibroblast growth factor (FGF), nitric oxide release etc30.

Angiotensin II released by myofibroblasts stimulates aldosterone production which subsequently

stimulates the transcription of collagen type I and III. Several weeks following myocardial

infarction, a stable fibrotic structure, composed primarily of collagen type I and type III, will

have formed in the infarcted zone31. Accordingly, this process can be blocked by angiotensin II

receptor blockade32.

1.4.6 Current Treatments for Ischemic Heart Disease and Heart Failure

Due to the complex mechanisms and myriad of causes underlying heart failure, and

cardiovascular disease as a whole, the development of effective treatments has been challenging.

Currently, pharmaceutical and surgical options are available to manage symptoms of ischemic

heart disease and heart failure, in addition to lifestyle modifications. However, no cures are

Page 28: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

10

available. Management of ischemic heart disease is largely through symptom relief (angina), and

lifestyle modifications such as increased physical activity, improved diet and quitting smoking29.

Surgical interventions such as angioplasty/stents or coronary artery bypass grafts are common if

plaque buildup begins to significantly restrict coronary blood flow, heightening risk for acute

myocardial infarction29. As for heart failure, angiotensin converting enzyme (ACE)-inhibitor

treatment to inhibit the renin-angiotensin-aldosterone pathway and reduce the work load of the

heart is one of the first steps in patients with heart failure29. The major goal is to minimize the

process of maladaptive remodeling. As heart failure progresses, β-blocker treatment, aldosterone

antagonists, and ionotropes can be added to the treatment regimen29. Patients with poor

prognosis despite optimal medical therapy will likely be considered for candidacy for heart

transplantation33. However, it is important to note that even heart transplantation is not a cure.

One year survival rates following heart transplantation are 90%, and the median length of

survival following transplantation is 11 years34. For selected patients who are too ill to wait for a

heart donor, ventricular assist devices are often utilized as a life-saving intermediate. These

devices are a mechanical option that serve to unload the failing heart and help maintain

perfusion to critical organs35. Heart transplantation is also inaccessible to many patients due to

donor organ unavailability, long wait lists and strict patient candidacy requirements.

Consequently, with the improvement in ventricular assist device technology over the past

decade, this therapy has shifted from merely a bridge to transplant, to providing lifetime support

for certain patients with end-stage heart failure35, as destination therapy. Despite major advances

in the field of transplantation and implanted mechanical devices, regenerative options to mitigate

cardiomyocyte loss or repair damaged cardiac tissue remains a sought after goal in cardiology.

Such options would strive to prevent the development of heart failure, and reduce the current

excessive demand for heart transplant.

1.5 Cell Cycle Regulation

1.5.1 The Cell Cycle

The cell cycle is the universal process by which cells reproduce, and lies at the core of growth,

development and tissue regeneration of all living organisms36. It consists of two main stages,

DNA synthesis (S phase), and chromosomal segregation/mitosis (M phase), interspersed by

quality control gap phases (G1 and G2 phase)37. These events culminate at the physical division

Page 29: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

11

of the cell into two daughter cells (cytokinesis) in the order G1, S, G2, M, cytokinesis37. Mitosis

can further be subdivided into five discrete phases: prophase, prometaphase, metaphase,

anaphase and telophase37.

DNA replication and mitosis are tightly regulated by, and dependent on, an interplay of many

cell cycle proteins, some of the most important being cyclin dependent kinases (CDKs) and

cyclins38. CDKs are serine-threonine-specific kinases and their activity and substrate specificity

is controlled by diverse cyclins, which serve as the regulatory subunit for CDKs38. Binding of

cyclins to CDKs promotes a conformational change in the kinase, allowing it to bind to ATP and

to their phosphorylation substrate. This interaction occurs in a specific orientation that facilitates

the transfer of the terminal phosphate group of ATP to the target serine or threonine residue of

the bound substrate37. The involvement of these proteins in the cell cycle was first illustrated in

yeast, in which a single CDK could promote the transition between different phases of the cell

cycle, based on which cyclin it was associated with38,39. The analogy was accordingly made that

CDKs act as the engine which drives cell cycle progression, and cyclins serve as the gears

changed between each phase to assist in the transition. As in any vehicle, brakes are essential to

maintain control. In this case, cyclin dependent inhibitors (CKIs) are the brakes of the cell cycle,

which regulate the kinase activity of CDK/cyclin complexes, and can halt cell cycle progression

under unfavorable conditions38. Tightly regulated oscillations between levels and activities of

CDK/cyclin complexes within the cell are the foundation upon which cell cycle progression is

built (Figure 1.3).

Figure 1.3. The major events of the cell cycle are regulated by transitions between CDK/cyclin complexes 37.

G1-cyclins (D type cyclins) complexed with CDKs are required to initiate the cell cycle and activate B-type-

cyclin-CDK activity. Low levels of B-type-cyclin-Cdk2 activity are sufficient to trigger S phase. CDK1 activation

and high levels of B-type-cyclin-CDK1 triggers mitosis and activates the anaphase promoting complex, triggering

anaphase and forming a negative feedback loop that in turn inhibits CDK1 activity. CDK1 inactivation allows

exit from mitosis and re-entry into G1 phase. Originally published by Rhind, N. & Russell, P. Cold Spring Harb.

Perspect. Biol. 4, a005942 (2012)37. Used with permission of the author and publisher.

Page 30: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

12

1.5.2 G1 Phase

In the first gap phase (G1 phase), the longest phase of the cell cycle, cells grow and prepare for

the onset of DNA synthesis40. Importantly, it is during this phase that the cell commits to

entering into a new cell cycle41. Specifically, it is a checkpoint late in the G1 phase called the

restriction point that the cell must pass in the presence of the appropriate growth factors, to

proceed into the next stages of the cycle42,43. Conversely, if the appropriate growth factors are

not present, the cell cycle will arrest at the restriction point, and entre the quiescent G0 phase42.

The tumor suppressor protein retinoblastoma (RB) is a potent inhibitor of the transition from G1

to S phase, preventing the initiation of DNA replication43. RB is found within a family of tumor

suppressor proteins including retinoblastoma-like protein 1 (p107) and retinoblastoma-like

protein 2 (p130), all of which are fundamental in the regulation of cell cycle and proliferat ion41.

RB is a nuclear phosphoprotein that can be phosphorylated by diverse CDK/cyclin complexes,

and is active when found in a hypophosphorylated state. In this state, early in the G1 phase, RB

can bind and inhibit the function of E2F transcription activators (E2F1, E2F2, E2F3A).

Furthermore, it recruits histone deacetylases to E2F regulated promoters which epigenetically

alter their conformation, rendering them inaccessible to the transcriptional machinery41,43.

Conversely, E2F transcriptional repressors (E2F4 and E2F5) are bound by p130 and p107, which

allows their nuclear localization where they can inhibit transcription of S phase related genes43.

RB confines the transcriptional activity E2F1-3 until the later stages of the G1 phase because this

family of proteins activates the transcription of genes that promote the transition from the G1

phase to the S phase. In the mid G1 phase, RB becomes hyperphosphorylated by CDK/cyclin

complexes (CDK4/6 and cyclin D1, D2 and D3), rendering it inactive until M phase41. This

allows activator E2Fs to induce transcription of many genes required for DNA replication such

as the E2f dimerization partner 1 (DP-1), DNA polymerase-α, proliferating cell nuclear antigen

(PCNA, a DNA clamp that functions with DNA polymerase), the DNA cutting enzyme

topoisomerase type 1, as well as genes essential for cell cycle progression like the proto-

oncogene avian myelocytomatosis viral oncogene homolog (c-MYC), cyclin D1, cyclin A, cyclin

E, CDK2, CDK1 (also known as CDC2), and CDC2541. D-type cyclins will reach their peak

expression level by late G1 phase. The degradation of cyclin D1 before this time point disables

the cell from progressing into S phase41, which highlights the requirement of E2F transcriptional

Page 31: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

13

activity for cell cycle progression, enabled by CDK-cyclin D1 dependent inhibition of RB. While

D-type cyclins are important for the progression of the G1 phase (marked by the switch from RB

activation to inactivation) cyclin Es and cyclin As are thought to be required at the G1 to S

transition40,41. Cyclin E’s expression peaks at this transition, and is subsequently degraded during

S phase40,41. Cyclin A at this time, will bind and activate CDK2, triggering S phase initiation40,41.

Its expression begins at late G1 phase following as previously mentioned and accumulates

throughout S and G2 phase, then disappears during M phase40,41.

1.5.3 S Phase

In eukaryotic cells, replication propagates bidirectionally from thousands of sites along the

genome (origins of replication)44. These origins of replication are first recognized by the origin

of replication complex (ORC1-6) during G1 phase44. Recognition of these sites by the ORCs will

localize the loading of the replicative DNA helicase minichromosome maintenance proteins 2-7

Complex (MCM2-7) to the origins44. Loading of the MCM2-7 also requires the proteins cell

division cycle 6 (CDC6) and chromatin licensing and DNA replication factor 1 (CDT1), which

are associated to the ORC complex and MCM2-7 respectively44. This event is often referred to as

“Origin Licensing”, and together the assembled proteins form the pre-replicative complex44.

During S phase, the MCM2-7 DNA helicase must become activated, which is followed by the

assembly of the replisome. The replisome consists of this helicase, which unwinds DNA, DNA

polymerases, which synthesize the leading and lagging strands of DNA, and a primase, which

facilitates the initiation of DNA synthesis44. Loading proteins, DNA replication regulator

SLD2/3/7 (SLD2, SLD3, and SLD7) are then recruited to the re-replicative complex44.

Phosphorylation of the pre-replicative complex (inactive DNA helicase) by Dbf4-dependent

Cdc7 kinase (DDK), and the loading proteins SLD3 and SLD7, will stimulate the recruitment of

cell division cycle 45 (CDC45), an essential helicase activator44. S-phase-CDKs such as CDK2

will subsequently phosphorylate SLD3 and SLD245. Phosphorylated Sld2 results in its interaction

with the DNA replication regulator DPB11 (DPB11)45. It is thought that this interaction

stimulates the recruitment of DNA polymerase-ε (polymerase that extends the leading strand of

DNA during replication) and GINS (a multi-protein complex), leading to the formation of what

is known as the pre-loading/initiation complex4546. The pre-loading complex will dock onto

phosphorylated SLD3 via DPB11 and deliver GINS and DNA polymerase-ε to the nascent

Page 32: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

14

replication complexes45. GINS, the second necessary helicase activator, will compete with SLD3

for binding to CDC45 and MCM2-747, resulting in the unloading of SLD3 from this complex and

the formation of the functional, replicative DNA helicase, CDC45/MCM2-7/GINS, otherwise

known as the CMG complex44. DNA polymerase-ε will bind directly to the CMG48.

Furthermore, although its time of assembly to the replisome is debated45, MCM10 is required for

recruiting the remaining DNA-polymerase-α/primase and DNA-polylmerase-δ44. As DNA

polymerases require a primer on which to begin DNA synthesis, the recruitment of DNA-

polymerase-α/primase, following the unwinding of the DNA strands by the CMG complex,

accomplishes this task49. The RNA primase will synthesize a ~10-nucleotide RNA primer

followed by 10 to 20 DNA bases. DNA polymerase-ε and δ will subsequently begin their tasks

of replicating the leading and lagging strands, respectively49,50 (Figure 1.4).

Figure 1.4 Assembly of the pre-replicative and pre-loading complex50. The origin of recognition complex

(ORC1-6) serves as a platform for the assembly of the pre-replication complex. CDC6 and CDT1 bind to the

ORC (origin licensing) which facilitates the loading of the inactive helicase complex proteins MCM 2-7. This is

known as the pre-replicative complex (PreRC). Helicase activator CDC45 is then recruited by SLD3, SDLD7 and

DDK. Phosphorylation of SLD2 and SLD3 by CDK2 promotes their interaction with DPB11 and the recruitment

of GINS to form the re-loading/initiation complex (PreIC). Unloading of SLD3 from the complex results in the

formation of the mature, functional DNA helicase, CDC45/Mcm2-7/GINS. Recruitment of Mcm10 and DNA

polymerases subsequently leads to the commencement of DNA replication. Modified from the original

publication by Aladjem, M. I. Nat. Rev. Genet. 8, 588–600 (2007)50. Used with permission of the author and

publisher.

SLD7

CDK2

P P

DDK

Page 33: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

15

G1 and S phase cyclins and CDKs are crucial for the sophisticated choreography of DNA

replication. Together they help to assemble and activate the previously mentioned prereplication

complex at the origins of replication of the genome51. Cyclin E-CDK2 and cyclin A-CDK2

complexes are of the most crucial for S phase initiation and progression51. Cyclin E-CDK2

phosphorylates many components of the prereplication complex which regulates initiation of

DNA replication, as well as proteins like nuclear protein ataxia-telangiectasia locus (NPAT)

which are required to activate the transcription of histones51. In turn, cyclin A-CDK2 is found

with replicating foci of DNA, and phosphorylates many components of the replication machinery

such as DNA polymerase α,δ, and PCNA51. Further evidence of cyclin A’s importance in DNA

replication is demonstrated by the fact that its ectopic overexpression is sufficient to promote S

phase progression52 and concordantly, injection of anti-sense DNA constructs for cyclin A

inhibits S phase entry53.

1.5.4 G2 Phase

The G2-M phase checkpoint is essential to ensure replication of the genome has occurred

faithfully and to its entirety, thereby preventing the division of mutated cells, cells lacking

genetic material, or cells with DNA damage. During this phase, cell growth continues, and

proteins are synthesized for the execution of mitosis. Again, this crucial checkpoint is tightly

regulated by CDKs42, where the transition from G2-M phase and entry into prophase is initiated

by the activation of CDK1 complexed to B-type cyclins (cyclin B1 and B2)54. Knockdown of

either of these components can cause G2 arrest54,55,56. Therefore during G2, CDK1 activity is

dampened by various mechanisms until the cell is completely prepared for M phase. Primarily,

this occurs through retaining CDK1-cyclin B in the cytoplasm, where its activity can be

negatively regulated by the WEE1 G2 checkpoint kinase (WEE1)37. WEE1 phosphorylates

CDK1 on the tyrosine and threonine residues, Y15 and T14 respectively, which inhibits its

catalytic activity by disrupting its substrate binding ability37. During the G2/M transition,

following CDK-activating kinase (CAK) phosphorylation, the CDK1-cyclin B complex will

translocate into the nucleus where inhibitory phosphate residues can be removed by the nuclear

bound phosphatase cell division cycle 25 (CDC25) such that it can proceed to phosphorylate its

many nuclear targets37. Dephosphorylation of Y15 and T14 by Cdc25 is the rate limiting step for

progression into mitosis37.

Page 34: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

16

1.5.5 M Phase

CDK1 has hundreds of downstream phosphorylation targets including many different effector

kinases necessary for the progression of mitosis37. For example, the initial chromosomal

condensation in prophase is dependent on the phosphorylation of the condensin II protein by

CDK157. This process can be visualized microscopically as string-like structures within the

nuclei, and is a hallmark of M phase initiation57.

M phase can be broken down into two large events; nuclear division (mitosis), followed by

cellular division (cytokinesis). During mitosis, the newly replicated sister chromatids are

separated equally for the formation of two daughter cells58,59. It is the most dramatic phase of the

cell cycle, as nearly all the inner contents of the cell become reorganized. Mitosis is

conventionally separated into 5 stages: prophase, prometaphase, metaphase, anaphase, and

telophase59 (Figure 1.5).

Figure 1.5 The main stages of mitosis 37. Prophase: CDK1 activation followed by CDK1-cylin B trafficking

into the nucleus leads to chromosomal condensation. Prometaphase: Breakdown of the nuclear envelope allows

formation of the mature spindle with centrosomes on either side of the cell. Kinetochore microtubules interact

with kinetochore protein complexes formed at the centromere, moving chromosomes to the spindle midzone.

Metaphase: Formation of the metaphase plate at the middle of the spindle, chromosomes become “bi-oriented”

through attachment to kinetochore microtubules. Unattached chromosomes produce a checkpoint signal that

prevents metaphase/anaphase transition. Anaphase: Once all chromosomes are attached and oriented properly,

the anaphase promoting complex ubiquitin ligase is activated, leading to ubiquitin -dependent proteolysis of

inhibitory protein, securin, allowing activation of a protease called separase which cleaves the cohesion proteins

that maintain cohesion between sister chromatids, allowing the chromosomes to separate to opposite poles of the

cell. Telophase: Following chromosome segregation, CDK1 becomes inhibited through anaphase promoting

complex mediated degradation cyclin B, and activation of the CDK1-antagonizing phosphatase CDC14. Dramat ic

decrease in CDK1 activity leads to reconstruction of the nuclear envelop, chromosomal decondensation and entry

into G1 following cytokinesis. Originally published by Rhind, N. & Russell, P. Cold Spring Harb. Perspect. Biol.

4, a005942 (2012)37. Used with permission of the author and publisher.

Page 35: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

17

During prophase, chromosomal condensation begins and the centrosomes (microtubule

organizing organelles) move to opposite sides of the nucleus. There, the centrosomes will serve

as the two poles of the mitotic spindle (microtubule structure responsible for separating the

chromosomes to the two opposite ends of the cell)37, which begins its construction during late

prophase. The condensed sister chromatids are held together at the centromere, the DNA region

where proteins will later attach to form the kinetochore, a structure that mediates attachment of

the chromosomes to the mitotic spindle37. The striking breakdown of the nuclear envelope marks

the end of prophase and the commencement of prometaphase, freeing the sister chromatids from

the nucleus37. In prometaphase, the mature mitotic spindle is formed with poles residing at the

two opposite sides of the cell37. Kinetochore microtubules will emanate from the spindle poles

and interact with the assembled kinetochores of the sister chromosomes37. This process triggers

the brief onset of metaphase, where the chromatids are aligned at the mid zone of the mitotic

spindle, known as the metaphase plate37,59. Transition into anaphase is triggered by activation of

the anaphase promoting complex (APC)37. The APC is a multi-subunit E3 ubiquitin ligase

complex that targets many mitotic proteins for proteolytic degradation. Its activation at the

metaphase-anaphase transition is mediated by the binding of CDC20, and APC-CDC20 will

proceed to degrade key proteins that function to prevent the separation of the sister chromatids,

thus permitting anaphase to occur37. A key substrate of APC-CDC20 at this time is the protein

securin37. Securin is a protein that inhibits separase, which is the protease responsible for

cleaving the proteins within the cohesin complex37 (a group of proteins including structural

maintenance of chromosomes 1 (SMC1), structural maintenance of chromosomes 3 (SMC3),

RAD21 cohesin complex component (RAD21), stromal antigen 1 (SA1/STAG1) and stromal

antigen 2 (SA2/STAG2), which maintain cohesion between sister chromatids following

replication)60. Loss of chromosomal cohesion by separase-dependent cleavage of this complex is

necessary to permit chromatid segregation, allowing the mitotic spindle to pull the sister

chromatids to opposite poles37. The end of mitosis is marked by telophase, the stage in which the

chromosomes decondense and the nuclei reform around the sister chromatids37. Nuclear envelop

reconstruction is permitted by the inactivation of CDK, as a result of cyclin B ubiquitination and

degradation through APC bound to cadherin 1 (CDH1)37.

Page 36: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

18

Cytokinesis, the mechanical process that cleaves the newly replicated mother cell into two

separate daughter cells, is the final step of mitosis59. During cytokinesis, the cell goes through

significant shape changes that must be tightly controlled, spatially and temporally, to ensure

proper segregation of genetic and cellular material61. The events of cytokinesis initiate during

anaphase, and are mediated by the formation of a contractile ring of actin and myosin filaments

beneath the plasma membrane, known as the cleavage furrow59. The ring is formed

perpendicularly to the mitotic spindle. Its contraction results in cleavage of the cell in the plane

that passes through the metaphase plate, pulling the plasma membrane inward and eventually

pinching the cell in half59.

Much like the rest of the cell cycle, cytokinesis is regulated by waves of cyclin and CDK

expression62. In budding yeast, degradation of mitotic cyclins by the APC, inactivation of CDK1

and upregulation of the major CDK antagonizing phosphatase CDC14, promotes each sequential

step of cytokinesis, beginning with furrow ingression, followed by plasma membrane resolution

and ultimately cell separation62. Experiments executed in HeLa cells expressing non degradable

cyclin B263, or temperature sensitive CDC14 mutants in yeast64,65, resulted in cells arresting in

late anaphase, without the completion of cytokinesis. CDC14 is sequestered into the nucleolus by

the protein neuroepithelial cell transforming 1 (NET1), throughout the majority of the cell cycle,

keeping it inactive66. However, in anaphase an abrupt increase in phosphorylation of NET1 by

the mitotic cyclin B1-CDK1 complex releases CDC14 it into the nucleus and cytoplasm where it

can now access its substrates67,68.

Recently, Kuilman et al. (2015), conducted phosphoproteome analyses to determine which

mitotic CDK substrates required dephosphorylation by CDC14 for the proper execution of

cytokinesis62. Dephosphorylation of actin interacting protein 1 (AIP1), EH domain-containing

and endocytosis protein 1 (EDE1), and Ingression protein 1 (INN1) were deemed necessary62.

They proposed that phosphorylated AIP1 acts as a negative regulator of acto-myosin ring

contraction, (dephosphorylation of AIP1 inhibits its function). Conversely, dephosphorylated

EDE1 acts as a positive regulator of cytokinesis (deletion of EDE1 but not AIP1 results in

increased cytokinetic defects), and dephosphorylation of INN1 promotes INN1’s timely

recruitment to the bud neck region of the dividing cell where it is required for ingression of the

plasma membrane62.

Page 37: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

19

Following furrow ingression, dividing cells are partitioned into two distinct daughters, but

remain connected by a thin intracellular bridge-like structure called the midbody69. This

structure arises from the central mitotic spindle (the microtubule array assembled between the

separating sister chromatids during anaphase), known as the midzone69. Conversion of the

midzone into the midbody corresponds with furrow ingression, as it is thought that this

contraction process compacts the microtubule bundles of the midzone into a single large

structure, creating the core of the midbody69. The serine-threonine kinase Aurora B, localizes to

the midzone and midbody structures, and also plays a role in chromosomal condensation through

histone H3 phosphorylation (PiH3)70. Due to its colocalization with the highly distinctive

midzone and midbody structures, immunofluorescence with antibodies against this protein is a

robust method to visualize cells undergoing mitosis and cytokinesis71. RNAi ablation of this

kinase results in a wide range of defects in prometaphase chromosomal condensation72,73,

anaphase chromosomal segregation73 and cytokinesis74 (Figure 1.6).

Figure 1.6 Aurora B Localization During Mitosis and Cytokinesis 71. HeLa cells were fixed and stained to

reveal Aurora B (red), tubulin (green), and DNA (blue). Aurora B translocates from the mitotic chromosomes in

metaphase to the central spindle early in anaphase. In early telophase when the cleavage furrow begins to form,

the cell can be seen pinching inwards in the same plane as the metaphase plate. At this stage Aurora B highlights

the spindle midzone. As telophase progresses, cleavage furrow ingression proceeds and the midzone is converted

into the midbody structure which is clearly marked by Aurora B that accumulates at the midbody arms in mid and

late telophase. Scale bars: 10 μm. Originally published by D’Avino, P. P. & Capalbo, L. Front. Oncol. 5, 221

(2015)71. Used with permission of the author and publisher.

Page 38: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

20

It is clear by the strict regulation of this complex process occurring at the all levels within the

cell, that the precise execution of the cell cycle is crucial for the proper function of every organ.

It is through the multiple signaling pathways and checkpoints described that critical events of

cell division occur in the proper order, and that quality control prevents cells from dividing with

damaged DNA or misaligned chromosomes. Certain cell types in the human body can re-enter

the cell cycle when stimulated by specific cues, to regenerate lost tissue, such as the liver, or are

in continuous turnover, such as endothelial cells. Other cells, such as adult cardiomyocytes, do

not have this luxury, and remain under tight cell cycle arrest from early in development. The

differences in regenerative potential between cells of different organs have important

implications for the types of diseases that can afflict them.

1.6 Cardiogenesis and Cardiomyocyte Cell Cycle Regulation

Adult cardiomyocytes are terminally differentiated cells that have exited the cell cycle, and

exhibit extremely poor regenerative potential. As previously discussed, different insults to the

heart such as ischemia (myocardial infarction), hypertension, atherosclerosis, and many others

can lead to heart disease, dysfunction or loss of cardiac muscle cells, and eventually death. The

problem lies in part, in the fact that the adult mammalian heart cannot efficiently generate new

cardiomyocytes in response to injury. Understanding early cardiogenesis, and the mechanisms of

cardiomyocyte cell cycle exit are crucial for the development of future therapies that involve

cardiac repair, as opposed to the current treatments that only transiently improve the function of

pre-existing cells.

1.6.1 Cardiogenesis

During embryogenesis in mice, two populations of cardiac progenitors derived from a common

precursor in the mesoderm contribute to the formation of the heart. Cardiac fate of these cells is

induced by bone morphogenic protein produced by the endodermal layer, whereas signals from

the wingless-type MMTV integration site family (WNT proteins, large family of secreted

glycoproteins which regulate crucial aspects of cell fate determination) from the neural tube and

notochord suppress cardiomyocyte specification75,76. At embryonic day 7.5, the primary heart

field, or cardiac crescent, is derived from the anterior mesoderm75,76. The secondary heart field is

derived from the pharyngeal mesoderm, located anterior and medial to the primary field75. Cells

Page 39: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

21

from the primary heart field migrate to form a linear heart tube at embryonic day 8, which acts as

the initial scaffold for further cardiac growth, including posterior and anterior expansion from

cells migrating from the secondary heart field75. The heart tube subsequently undergoes

rightward looping, which is the basis for the formation of the ventricles and atria and eventually

leads to the proper development of the four heart chambers75. Heart maturation includes septum

and valve formation. GATA Binding Protein 4 (GATA4) and NK2 homeobox 5 (NKX2.5) are

fundamental transcription factors in both heart fields, and two of the earliest cardiac lineage

markers, whereas T-box protein 5 (TBX5) and ISL LIM Homeobox 1 (ISL1) are uniquely

expressed in the primary and secondary heart fields, respectively75. Insight into these lineage

markers has been essential for the production of cardiomyocytes derived from induced

pluripotent stem cells (iPSCs) as well as for promoting cardiac repair in injured adult hearts.

1.6.2 Cardiomyocyte Regeneration

Throughout embryogenesis, mammalian cardiomyocytes undergo cell division such that the

heart grows through cardiomyocyte proliferation77. After birth however, this drastically changes.

Cardiomyocytes undergo a final round of DNA synthesis (karyokinesis), but do not complete

cytokinesis, rendering the vast majority of cells binucleated, which marks their terminal

differentiation77. Morphologically, terminal differentiation of cardiomyocytes is characterized by

binucleated cells with increased size, myofibril density, and mature intercalated discs77. At this

time, there is a transition from hyperplastic (cell proliferation) to hypertrophic (cell enlargement)

cardiomyocyte growth. This was highlighted in a study which showed that cardiomyocytes

isolated from the hearts of 1 day old rats increased in number by 68% over the first three days in

vitro and subsequently remained constant. In contrast, from day 3 to day 12, cardiomyocytes

increased in volume by 2.5 fold, as opposed to in number78. In addition to an increased

proliferative capacity, neonatal hearts also retain a heightened regenerative capacity in response

to injury, similar to amphibians and zebrafish79. To demonstrate this, murine ventricular apexes

were resected 1 day after birth and at 21 days following resection, the entire apical defect

incurred by these mice had been replaced by new cardiomyocytes, accompanied by increased

markers of proliferation, restored systolic function, with the absence of hypertrophy or fibrosis79.

Interestingly, resection at 7 days after birth, a time point which aligns with cardiomyocyte cell

cycle withdrawal, does not promote cardiac regeneration79. Following surgery, these hearts did

Page 40: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

22

not show any indication of increased cardiomyocyte proliferation, and instead experienced large

amounts of fibrosis79. These results indicate that there is a drastic switch in the proliferative and

regenerative capacity of cardiomyocytes within the first week after birth. This loss of the

proliferative capacity is associated with an exit from the cell cycle. The cell cycle exit observed

has been shown to correspond with a stark upregulation of CDK inhibitors leading to a dramatic

downregulation of many cyclins and their CDKs to almost undetectable levels (their expression

is known to be high in embryonic, proliferative hearts)77. Tane et al. (2015) demonstrated that

the upregulation of two CDK inhibitors, cyclin-dependent kinase inhibitor 1A and 1B

(CDKN1A/p21Cip1 and CDKN1B/p27Kip1) mediate cell cycle exit in postnatal cardiomyocytes,

and the inhibition of two cell cycle activators, cyclin D1 and CDK1, mainta ins cell cycle arrest

throughout life80,81. Firstly, they demonstrated that p21Cip1 and p27kip1 levels peak at post-natal

day 581. Upregulation of these inhibitors coincides with when cyclin D1, cyclin E and cyclin A

activities begin to decrease, which are essential for G1 entry, progression and transition into S

phase81. Next they showed that over expression of cyclin D1 in mice using a Cre-loxP system

allowed adult cardiomyocytes to re-enter into the cell cycle, but these cells became arrested in M

phase due to lack of CDK1 activation80. They therefore proposed that two blockades of cell cycle

progression exist in adult cardiomyocytes, a G1 blockade, due to cyclin D1 downregulation, and

an M phase blockade, due to inhibition of CDK1 activation80. It is clear from this work that a

multi-pronged approach that could induce the upregulation of a battery of cell cycle activating

genes, and/or reduce key cell cycle inhibitors is necessary to promote cardiomyocyte

proliferation, but targeting solely one cell cycle regulator is not sufficient, as road-blocks at

multiple levels are in place.

1.7 MicroRNAs

MicroRNAs (miRNAs) are a group of small non-coding RNAs with the capability of expansive

genetic regulation. This class of endogenous 21-25 nucleotide long single-stranded RNA

molecules function as guide molecules in RNA silencing through base pairing to their

complementary mRNA targets82,83,84. By targeting specific mRNAs for degradation or

translational repression, miRNAs acts as inhibitors of gene expression, and are involved in

almost all developmental and pathological processes in animals. Since the discovery of the first

miRNA in Caenorhabditis elegans, lin-4, in 199385, cloning and size-fractionated RNA

Page 41: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

23

techniques have allowed for the identification of thousands of miRNAs, many of which have

been shown to have regulatory roles in cell cycle regulation, proliferation, cell death, cardiac

development, metabolism, neuronal patterning, hematopoietic differentiation and the immune

system82. Recently, high throughput sequencing, computational techniques and bioinformatics

algorithms have allowed for more in depth studies of the diverse target genes of miRNAs, and it

is known that one given miRNA can bind hundreds of target mRNAs. Based on the potential for

vast impact on gene expression, miRNA biogenesis is tightly regulated at multiple levels within

the cell.

1.7.1 miRNA Nomenclature

Over time, nomenclature of miRNAs has been inconsistent. Early discovered miRNAs were

named after their phenotypes (e.g. lin-4 or let-7), whereas miRNAs discovered through cloning

or sequencing were given numerical names (e.g. the lin-4 homologues in other species are called

miR-125)84. miRNAs generated from different loci that have identica l “seed” sequences from

nucleotides 2-7 are considered a family of miRNAs, and generally arose through gene

duplication84. For example, in humans, there are 14 paralogous loci (encoding “miRNA sisters”

that belong to the let-7 family, and are indicated by lettered suffixes (e.g. let-7a, let-7f)84. Each

locus produces two mature miRNAs; one from the 5’ strand and one from the 3’ strand of the

pre-miRNA (e.g. miR-181a-5p and miR-181a-3p), but one arm is then chosen as the guide

strand, and is usually 96-99% more prevalent and more biologically active than the other,

passenger strand76.

1.7.2 miRNA Biogenesis

miRNA genes are transcribed by RNA polymerase II or III as a long (> 1 kilobase pairs) primary

transcript (pri-miRNA) containing a hairpin structure in which the miRNA sequence is

embedded83. Still within the nucleus, the pri-miRNA is recognized by the double stranded RNA

binding domain regions of the DiGeorge Syndrome critical region gene 8 protein (DGCR8),

which will guide the class 2 RNAse III enzyme Drosha ribonuclease III (Drosha) to cleave the

RNA molecule83. Cleavage by Drosha liberates a 60-70 nucleotide long stem structure known as

the pre-miRNA. Proper cleavage by Drosha is crucial as it defines the terminus of an miRNA,

and thus, its specificity83. Drosha will cleave the pri-miRNA ~11 base pairs from the basal

Page 42: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

24

junction, which serves as the major reference point for cleavage site determination, and then

again ~22 base pairs from the apical junction83,86. The cleaved pre-miRNA has a staggered end

with a ~2 nucleotide single stranded overhang at its 3’ end. This overhang allows it to be

recognized by exportin-5 which will mediate its transport into the cytoplasm for further

maturation83. Here, Dicer, a second RNAse III endonuclease, will process the pre-miRNA into

its mature form through specific cleavage ~22 nucleotides from its preexisting 5’ terminus (in

mammals)83. The approximately 22 nucleotide miRNA duplex is subsequently loaded onto the

Argonaute (AGO) protein complex, and one strand of the duplex (passenger strand) is degraded

by the endo-nucleolytic enzyme activity of AGO87, whereas the remaining single stranded RNA

molecule becomes the mature guide strand/miRNA. The mature miRNA bound to the AGO

complex is known as the RNA-induced silencing complex (RISC). Guide versus passenger

strand selection is mediated partially by the thermodynamic stability of the two ends of the

duplex, whereby the strand with lower internal stability at the 5’ terminus is preferentially bound

by AGO and retained in the RISC complex88. Additionally, AGO proteins are biased to selecting

guide strands that contain a uridine at their 5’ terminus. However, it is known that these rules of

strand selection are not completely strict, as passenger strand selection (arm switching) has been

observed with varying frequencies between species89,90. The mature miRNA serves to guide the

RISC complex to its target mRNA, and complementary binding of the miRNA to its target

gene’s mRNA transcript will lead to silencing of the mRNA through its direct degradation, by

translational repression, or by promoting mRNA decay84,91 (Figure 1.7).

Mature miRNAs are thought to be quite stable within the cell, as they can still be detected 48

hours after depletion of miRNA processing factors92. Binding of mature miRNAs to AGO

proteins enhances their stability even further93. Different miRNAs are seen to be degraded at

different rates within the cell, and degradation occurs at the precursor level as well as of mature

miRNAs, indicating that this may be yet another layer of regulation in the processing pathway92.

However, nucleases that degrade human miRNAs remain to be uncovered92.

Page 43: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

25

1.7.3 miRNA Mechanism and Function

The RISC complex, guided by a mature miRNA to recognize specific mRNAs, post-

transcriptionally downregulates gene expression by one of three mechanisms: 1) inhibition of

mRNA translation, 2) mRNA cleavage, or 3) acceleration of miRNA decay84. miRNAs bind to

their targets through Watson-Crick base pairing, most frequently in the 3’ un-translated region

Figure 1.7 The Canonical Pathway of miRNA Biogenesis.91 (A) RNA polymerase II or III mediated

transcription of the primary miRNA transcript (pri-miRNA) and cleavage of the pri-miRNA by the

microprocessor complex Drosha-DGCR8 within the nucleus begins miRNA biogenesis. This results in the

formation of the pre-miRNA which is exported into the cytoplasm by exportin-5. In the cytoplasm, the RNase

Dicer complexed with TRBP will cleave the pre-miRNA into its mature 21-25 nucleotide length. The guide

miRNA strand is loaded onto the Argonaute proteins (AGO) to form the RNA -induced silencing complex

(RISC). The RISC complex, guided by the mature miRNA will silence target mRNAs through mRNA cleavage,

translational repression or mRNA deadenylation83,91. (B) The microprocessor complex measures ~11 bp from

the basal junction and ~ 22 bp from the apical junction and will cleave the RNA strand at this position to generate

the pre-miRNA83,100. Originally published by Winter, J et al. Nat. Cell Biol. 11, 228–234 (2009)91. Used with

permission of the author and publisher.

A B

Page 44: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

26

(UTR) of target mRNAs84. Binding is not always 100% complementary, and mismatches and

bulges can be observed84. The degree of complementarity between miRNA and mRNA is the

major determinant of whether the mRNA becomes cleaved or if translation is simply inhibited84.

A high degree of complementarity is necessary to facilitate AGO-catalyzed mRNA cleavage,

whereas increased mismatching leads to translational repression or mRNA decay84.

The exact mechanism of translational inhibition by RISC has not been completely elucidated.

However, density gradient centrifugation has been used to show that translation can be inhibited

by miRNAs at either the translation initiation step (mRNA found complexed with RISC and

translational initiation factors) or at the elongation step (larger, incomplete polysomes

detected)84. Peterson et al. 2006 proposed that RISC binding can promote early ribosome

dissociation from mRNAs, thus interfering with the elongation process94.

Two different models have been proposed for miRNA inhibition of translational initiation. First,

RISCs have been shown to compete with eukaryotic translation initiation factor 4E (eIF4E) for

binding to the 5’ cap of the mRNA, resulting in failure to initiate translation90,94. The second

model proposes that binding of the RISC complex to the mRNA and 40S ribosomal subunit

preinitiation complex recruits the eukaryotic translation initiation factor 6 (eIF6), a ribosome

inhibitory protein. eIF6 binds to the 60S ribosomal subunit and prevents productive assembly of

80S ribosomes, and it was shown that depletion of Eif6 abrogates the Caenorhabditis elegans lin-

4 mediated translational repression of endogenous LIN-4 mRNA.

mRNA decay is promoted by RISC destabilization of the mRNA transcript. This occurs when

RISC binding to certain targets leads to rapid deadenylation of the 3’ Poly(A) tail of the

mRNA95,96. 3’ Deadenylation destabilizes the mRNA and facilitates its decay in the cytoplasm.

The let-7 family has been shown to function through this mechanism95,96.

Overall, the exact mechanism of silencing, be it translational repression, mRNA decay through

destabilization of its structure, or AGO mediated mRNA cleavage, remains to be elucidated for

the large majority of mammalian miRNAs. It is clear, however, that their influence on gene

expression is potent and widespread.

Page 45: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

27

1.7.4 MicroRNAs and the Heart

1.7.4.1 MicroRNAs, Cardiac Development and Homeostasis

Zhao et al. (2007)97 and Chen et al. (2008)98 were the first to reveal the crucial role of miRNAs

in heart development and function through the conditional, tissue-specific knockout of Dicer, the

RNase III enzyme essential for the biogenesis of mature miRNAs. Dicer knockout led to early

embryonic97 or post-natal98 lethality (depending on time of knockout), due to severe

developmental defects in the cardiovascular system. Using Cre recombinase technology under

the control of the endogenous NKX2.5 promoter, knockout of Dicer, specifically in cardiac

progenitors by embryonic day 8.5 (E8.5), caused pericardial edema, poorly developed ventricles

myocardium and death by E12.597. Although early cardiac markers such as TBX5 remained

unchanged, microarray analysis of hearts at E11.5 revealed upregulation and downregulation of

numerous genes97. For example, the endoderm marker α-fetoprotein and the skeletal muscle

troponin, were upregulated, whereas myoglobin and the potassium voltage-gated channel

subfamily D member 2 (KCND2) were strongly downregulated97. Interestingly, ablation of Dicer

using Cre recombinase under the control of the α-myosin heavy chain promoter (α-MHC), which

directs cardiac specific expression, did not lead to embryonic lethality. Instead, all mice died

shortly after birth, by day 498. The discrepancy between these two studies likely lies in the timing

of Dicer knockout, as use of the α-MHC Cre recombinase system leads to knockout by E14.5 as

opposed to E8.5 when using the NKX2.5 promoter. This indicates that miRNAs play a role in

early and in late cardiac development. Dicer knockout by E14.5 led to the development of

substantially larger hearts compared to non-mutated littermates, due to dramatic left ventricle

dilation with intracardiac thrombus98. Decreased myocardial organization, decreased

cardiomyocyte integrity, and disarrayed myofibrils and sarcomeres, due to a significant decrease

in expression of contractile proteins were also observed, indicating that miRNAs are essential for

the development of proper heart structure and contractile function98. Disarrayed sarcomeres were

also dramatically shorter and decreased in abundance in mutant mice98. Specifically, there was a

strong decrease in myosin heavy chain proteins (except for β-myosin heavy chain) and cardiac

troponin T, which likely accounts for the structural phenotypes observed98. Furthermore, there

was a drop in heart rate also observed, suggestive of conductive defects98. Concordant with these

molecular changes, Dicer mutant mice also suffered from severe left ventricular dilatation and

Page 46: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

28

decreased fractional shortening98. In all, it was concluded that loss of miRNA production in

cardiomyocytes by E14.5 results in a phenotype suggestive of dilated cardiomyopathy with the

concomitant development of heart failure, shortly following birth. This demonstrates that proper

expression of miRNAs is absolutely essential for the intricate development of the cardiomyocyte

contractile apparatus, and thus heart function98.

To determine the role miRNAs play in the maturing and adult heart, da Costa Martins et al.

(2008) subsequently knocked out Dicer in mice at 3 and 8 weeks of age99. Cardiac-specific

deletion of Dicer 3 weeks following birth results in sudden death, with only mild cardiac

remodeling, whereas deletion 8 weeks after birth caused extensive hypertrophy and the

accumulation of fibrotic lesions99. In addition, deletion of Dicer at 3 weeks of age led to mild

inflammatory cell infiltration, but no myofibril disarray, hypertrophy or fibrosis99. A

downregulation of the KCND2 calcium channel was also observed (similarly observed in Dicer

knockout at E8.597), along with enhanced arrhythmias, which likely accounts for the sudden

death experienced by these mice99. This revealed a necessity for miRNA function not only in the

embryonic heart, but also during cardiac maturation99.

Knockout of Dicer in 8 week old adult mouse hearts however led to a severely different

phenotype, with mutant mice developing heart weights twice the mass of their non-mutant

littermates, severely hypertrophic and disarrayed cardiomyocytes, strong inflammatory

infiltration and interstitial fibrosis99. Four weeks following Dicer depletion, mice experienced a

50% decrease in fractional shortening and significant dilation of the left ventricle, indicating that

Dicer depletion in the adult myocardium led to rapid pathological remodeling, and a phenotype

resembling heart failure99. In both mutants, a strong upregulation of pro-hypertrophic and fetal

genes occurred99. Together, these three studies indicate that subsets of miRNAs play different

but crucial roles in cardiac function at all stages of development as well as in cardiac

maintenance/homeostasis. Based on the requirement of miRNAs to maintain mature cardiac

homeostasis, Rao et al. (2009) sought to profile the population of miRNAs expressed in the adult

murine heart using Illumina/Solexa deep sequencing of a small RNA library100. Unexpectedly,

analysis of over 7 million reads from each sample of 6-8 week old hearts revealed that 18

miRNAs make up more than 90% of all expressed small RNAs, and the miR-1/206 family

accounted for almost 40% of all known miRNA reads in the heart100. The miR-30, let-7, miR-29

Page 47: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

29

and miR-26 families were the next most abundant miRNA families expressed in the adult murine

heart100. The full list of top 20 most enriched miRNAs, and miRNA families can be found in

Figure 1. of Rao et al. (2009)100. miRNAs expressed within the introns of important cardiac

genes (e.g. miR-378 encoded within an intron of the PPARG coactivator 1 beta (PGC1-β) gene, a

crucial regulator of mitochondrial biogenesis) were also found to have a relatively high

expression in the heart, likely reflecting the high levels of transcription of these genes100. These

results indicate that either a limited number of highly expressed miRNAs regulate cardiac

maintenance/homeostasis, or a large number of low-abundance miRNAs with overlapping

functions, as well as the few highly abundant ones, are involved in these processes101. In line

with this study, Porrello et al. (2011) showed that from day 1 after birth to day 7, there is a

drastic switch in the miRNA profile in mice102. By day 7, the miR-15, let-7 and miR-30 families

were all significantly upregulated, correlating with the time of cardiomyocyte maturation and

cell cycle exit102. Overall, it is now well documented that the timely expression of specific

miRNAs is crucial for cardiac development and homeostasis.

1.7.4.2 MicroRNAs and cardiovascular disease

Given the importance of miRNAs in development, it is evident that deregulation of miRNA

expression could be implicated in human disease. Recently, miRNAs have been found

circulating in blood, and have been emerging as important biomarkers in numerous

cardiomyopathies103–105. Understanding the role of these miRNAs in the pathogenesis of

cardiovascular disease may has opened a new avenue for the development of novel therapies and

diagnostic tools.

The ischemic process during and following myocardial infarction is the major cause of global

transcriptomic changes within the heart, and triggers an inflammatory response22. Subsequently,

maladaptive cardiac remodeling plays an important pathologic role in the development of heart

failure post-myocardial infarction. The role of miRNAs in cardiac remodeling post-myocardial

infarction came to light when da Costa Martins et al. (2008) demonstrated that the miRNA

biogenesis pathway was essential for overall cardiac function, and that its loss appeared to

contribute to maladaptive cardiac remodeling and heart failure99. Several studies subsequently

demonstrated a crucial role for miRNAs in regulating angiogenesis, fibrosis and hypertrophy,

Page 48: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

30

following myocardial infarction, linking altered miRNA expression to cardiomyocyte

ischemia106,107,108. The deregulation of miRNAs following ischemic insult highlights their

involvement in the regulation of normal heart function, suggesting that detecting alterations in

miRNA expression could be a sensitive prognostic marker in early the stages of heart failure.

The use of high-sensitive real-time PCR (RT-qPCR) or small RNA array techniques facilitate

miRNA detection in blood samples, even at very low concentrations in the circulation22. For

example, miR-208b was found to be increased by ~1600 fold in the plasma of patients with acute

myocardial infarction, compared to patients presenting with atypical chest-pain without

myocardial infarction104. Furthermore, miR-499-5p levels were found to be elevated in acute

myocardial infarction patients and in animal models of myocardial infarction109. Increased levels

of miR-499-5p were also positively correlated with 12-month mortality in a group of elderly

myocardial infarction subjects, highlighting its potential as a prognostic biomarker109. Moreover,

patients in the Osaka Acute Coronary Insufficiency Study (OACIS), who died due to a cardiac

cause within a year of discharge, had a ~3-4 fold upregulation of miR-155 and miR-380*,

suggesting measuring their levels in patients following acute myocardial infarction may have

prognostic value105. It was also shown that OACIS patients surviving acute myocardial

infarction, who developed de novo heart failure within one year, had significantly upregulated

levels of the p53-responsive miRNAs-192, -194 and -34a (see section 1.8.6 p53 and miRNAs) in

their sera103.

Not only have miRNAs been implicated in the onset of heart failure, post-ischemic injury, but a

plethora of miRNAs are also dysregulated during the progression of heart failure22. Multiple

studies have demonstrated that subsets of miRNAs that become upregulated in heart failure

promote the formation of interstitial fibrosis110,111,112 , hypertrophic extracellular signal-regulated

kinase (ERK)/MAPK110, and nuclear factor of activated T-cells (NFAT)/calcineurin signaling113,

the fetal gene program114, glucose metabolism115,116, apoptosis116, cytoskeletal remodeling117 and

arrythmogenesis116 (often by miRNA-induced inhibition of negative regulators of these

pathways). Conversely, miRNAs which maintain normal low physiological expression levels of

target genes that promote cytoskeletal and myofibrillar rearrangements (hallmarks of

hypertrophy), such as the RAS homolog gene family member A (RhoA)118 and CDC42119,

Page 49: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

31

become downregulated in hypertrophy and heart failure (such that these processes become

upregulated)120.

Given these observations, miRNAs are involved in a myriad signaling and biological processes

critical for the regulation of cardiac tissue homeostasis and function. Thus, exploiting these

functions through miRNA-based therapies is poised to lead to novel approaches to treat patients

following myocardial infarction or with heart failure.

1.7.5 MicroRNA Therapeutics

Over the years, as it has become more apparent that miRNAs function as master regulators of the

genome, and are differentially regulated in multiple disease states, many groups have sought to

take advantage of their function for the development of miRNA based therapeutics against

different types of diseases. Through over-expression by miRNA mimics or by knock down with

the use of antagomirs, using either viral vectors or lipid based techniques as delivery methods,

the therapeutic potential of miRNAs is extensive.

1.7.5.1 Delivery Methods for miRNA Based Therapies

Viral-based delivery systems usually exploit retroviruses, lentiviruses, adenoviruses or adeno-

associated viruses as delivery vectors to overexpress a given gene of interest121. These systems

can also be employed for the overexpression of miRNAs, often in a tissue-specific manner. Viral

vectors have three major advantages: 1) high infection efficiency, 2) high and constant

expression of the miRNA mimic or antagomir of interest and 3) certain viruses can be

engineered to have a particular tissue specificity depending on the receptors they use to enter

into cells121. Furthermore, certain genomic modifications which inhibit their ability to replicate

have improved the safety of this delivery system121.

Transfection of miRNA inhibitors or mimics using lipid based systems is one of the most

common ways to study miRNA overexpression or downregulation121. In this approach, a cell-

membrane-like lipid surface will encapsulate the nucleic acids which need to be delivered into

the cell, and the membrane- like qualities of the capsule allow the liposome to pass through the

cell’s membrane, into the cytoplasm121. Using a lipid-based approach circumvents many of the

problems of viral vectors, but lipid based techniques still cause a certain amount of toxicity to the

Page 50: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

32

cell, the transfection efficiency of this method is often less potent, and the liposome particles

have a short half-life121. Yet, liposomes remain as the more clinically relevant method to alter

miRNA expression. Commonly, miRNA mimics, which act to overexpress a given miRNA, or

miRNA antagomirs (anti-miRs), which act to downregulate the expression of a given miRNA,

are transfected into cells using this approach121.

1.7.5.2 miRNA Mimics

miRNA mimic (miR-mimic) technology is an innovative approach to gene silencing, through

exploitation of miRNA function122,123. This is achieved by designing a ~22 nucleotide RNA

fragment with its 5’ end partially complementary to a selected unique sequence in the 3’ UTR of

the target gene of choice123. Once transfected into the cell, this RNA molecule, mimicking an

endogenous miRNA, will be picked up by the RISC complex, bind specifically to its target gene,

and induce post-transcriptional repression of the gene123. Alternatively, a miRNA mimic can be

designed to have the same sequence as a known miRNA, which allows you to study the effect of

having increased amounts of a given miRNA within a cell122. miRNA mimics have been used

extensively to study the consequences of miRNA overexpression in the heart, and have been

utilized in a therapeutic framework to substitute protective miRNAs that become depleted in a

diseased state. For example, Huang et al. (2016) transfected neonatal rat cardiomyocytes with a

cocktail of miR-21-mimic and miR-146a-mimic, two miRNAs previously known to have anti-

apoptotic and anti-inflammatory properties, respectively124, following hypoxia in vitro, and

myocardial infarction in vivo. Co-transfection of these two miRNA mimics reduced the degree of

cardiomyocyte apoptosis124. Furthermore, this treatment reduced infarct size and improved

cardiac function post-myocardial infarction124. Looking at gene expression in the heart following

treatment, they found that upregulation of these miRNAs significantly reduced the expression of

the phosphatase and tensin homolog (PTEN) and tumor necrosis factor receptor associated factor

6 (TRAF6), important apoptotic regulators, leading to the increased phosphorylation of p38

MAPK and a consequential downregulation of caspase 3 activity (involved in the signal

transduction cascade responsible for apoptosis execution)124. This study highlights the

therapeutic value and feasibility of upregulating cardioprotective miRNAs using miRNA

mimics.

Page 51: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

33

1.7.5.3 miRNA Antagomirs

miRNA antagomirs employ the opposite approach to mimics, and function by intercepting

overexpressed, detrimental miRNAs122. As their name suggests, antagomirs antagonize the

function of miRNAs, and have served as a promising strategy to target oncogenic miRNAs for

the treatment of cancer, or upregulated pathogenic miRNAs in other diseases122. This technology

was pioneered by Krutzfeldt et al. 2005, who designed chemically modified, cholesterol-

conjugated single-stranded RNA analogues complementary to miRNAs, and gave them the name

antagomirs125,126. These molecules can be transfected or transduced into cells in vitro or in vivo,

and function by intercepting and degrading mature miRNAs, preventing the functional miRNA

effect, that is silencing of their respective target genes125,126. Antagomir nucleotide modification

was also optimized by this group, including 2’O-methyl nucleotides for enhanced miRNA-

mRNA duplex stability, a phosphorothioate backbone to prevent RNase-mediated antagomir

degradation and conjugation to a cholesterol molecule for improved delivery across cell

membranes in vivo without the requirement of liposomes125. Further improvements to antagomir

technology have been made over the years, such as utilizing fully locked nucleic acid (LNA)-

modified oligonucleotides122. LNAs are RNA nucleotides that are modified to contain an extra

bridge on the ribose moiety, locking the ribose in a conformation which significantly improved

hybridization properties, therefore improving the potency of antagomirs122.

The therapeutic potential of miRNA antagomirs in the heart has been highlighted by many

studies, showing that knockdown of specific miRNAs can improve heart function in a battery of

cardiovascular disease models. For example, Wahlquist et al. (2014) demonstrated that miR-25

becomes significantly upregulated in human myocardial samples from patients with severe heart

failure. This miRNA strongly targets and downregulates expression of the Ca2+ pump SERCA2

(Ca2+ uptake is impaired due to a downregulation in SERCA2 expression in heart failure)127.

They next showed that transduction of anti-miR-25 in vivo into trans-aortic constriction (TAC)-

induced failing mouse hearts led to a significant increase in SERCA2 levels, restored fractional

shortening to normal levels (~20% increase), stabilized the heart weight to body weight ratio and

drastically improved survival (7/8 of anti-miR-25+TAC mice surviving vs. 7/22 TAC mice

surviving)127. Due to their ability to enter cells without the requirement of liposomes or viral

vectors, antagomirs may be a safe and viable avenue for drug development in cardiovascular

Page 52: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

34

disease as in other diseases. Current research is now beginning to focus on non-oligonucleotide

based methods to modulate miRNA expression levels, such as through small molecules.

1.7.6 MicroRNAs and the cell cycle

Similar to embryogenesis and development, the carefully timed expression of cell cycle

activators and inhibitors is essential for the proper regulation of the cell cycle. Furthermore, as

cells of different organs proliferate at different rates, or are completely removed from the cell

cycle, it is clear that the expression level of these cell cycle regulators is highly tissue specific.

Thus, it is not surprising that miRNAs, with the capability to fine tune the expression level of

hundreds of genes, are key players in cell cycle regulation. The differential expression of

miRNAs in the different tissues of the body likely plays an important role on regulating that

tissue’s proliferative capacity. Consequently, over the years, miRNAs have been emerging as

novel regulators of tumor suppression and oncogenesis.

Each stage of the cell cycle is regulated by a battery of miRNAs, targeting mRNA transcripts of

both cell cycle activating and inhibiting genes128. One of the first links to implicate miRNAs with

the cell cycle was made by Calin et al. (2002) who uncovering the anti-proliferative effects of

the miR-15a-16-1 cluster (which expresses mature miRNAs miR-15a and miR-16), and their

implication in cancer129. This cluster was identified as the target of specific chromosome

aberrations in chronic lymphocytic leukemia patients, and is deleted or downregulated in ~70%

of these patients129. Further studies then found that it was also deleted or downregulated in

pituitary adenomas130, gastric cancer131,132 and prostate cancer133, providing supporting evidence

for an important role in tumor suppression. It is thought that these two miRNAs maintain cell

cycle arrest at the G1 phase by targeting CDK1, CDK2, CDK as well as cyclins D1, D3 and

E1132,134,135. Following this, a plethora of other miRNAs were found to target these major cell

cycle kinase complexes. It is now clear that at each phase of the cell cycle, many important cell

cycle regulators are targeted by multiple miRNAs in a largely redundant manner. This highlights

the importance of keeping a tight regulation over these key genes (Figure 1.8).

Page 53: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

35

Based on the knowledge that miRNAs have a strong influence on the cell cycle, Eulalio et al.

(2012) sought to determine which miRNAs play a role in maintaining cell cycle arrest in

cardiomyocytes136. To do this, they performed a fluorescence-microscopy-based screen of 875

miRNA mimics in neonatal rat cardiomyocytes to systematically identify miRNAs that trigger

cardiomyocyte cell cycle re-entry136. Staining for the proliferation antigen Ki-67 and for 5-

Figure 1.8 An overview to cell cycle control by microRNAs 128. let-7 (let-7a-f) and miR-15 (miR-15, miR-16

and miR-195) indicate several members of the corresponding miRNA family. miRNAs with proliferative potential

are shown in red whereas antiproliferative miRNAs are in blue. S = S-phase, M = Mitosis, G1 and G2 indicate the

gap phases of the cell cycle and G0 indicates quiescent cells . Originally published by Bueno, M. J. & Malumbres,

M. Biochim. Biophys. Acta - Mol. Basis Dis. 1812, 592–601 (2011)128. Used with permission of the author and

publisher .

Page 54: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

36

ethynyl-29-deoxyuridine (EdU, a uridine analogue that is incorporated into newly synthesized

DNA), they found that 204 miRNA mimics significantly increased neonatal cardiomyocyte

proliferation by more than two-fold, and 331 mimics decreased Edu incorporation and Ki-67

positivity from the baseline of 12.5% to 0%136. Interestingly, the top miRNAs promoting

cardiomyocyte proliferation were not among those affecting cardiomyocyte size or

hypertrophy136. In this study, the top 4 pro-proliferative miRNAs which significantly increased

both EdU incorporation and Ki-67 positivity were miR-590-3p, miR-199a-3p, miR-1825 and

hsa-miR-33b*136. Conversely, the top 4 miRNAs with tumor suppressor properties, that

significantly decreased cardiomyocyte proliferation were miR-1287, miR-34c, miR-885 and

miR-449136. The authors subsequently showed that treatment of mice with mimics to these

potentially pro-regenerative miRNAs (miR-590-3p and miR-199a) significantly boosted the

repair of adult cardiomyocytes post-infarct, and that the increase in cardiomyocyte proliferation

was correlated with improved functional outcomes136. The overall implication of these results are

twofold. Firstly, that a vast number of miRNAs must be differentially regulated in adult

cardiomyocytes to maintain cell cycle arrest, whereby the majority of pro-proliferative miRNAs

are downregulated and miRNAs promoting cell cycle arrest are more prominently expressed.

Secondly, that upregulating cardiomyocyte proliferation following injury to stimulate heart

regeneration is a viable therapeutic option that merits further investigation.

1.8 Tumor Suppressors – A Focus on p53

Neoplasia and tumorigenesis represent a loss of control in the processes of cell proliferation and

differentiation137. Tumor suppressor genes were first identified because they were inactivated in

mammalian tumors, and subsequent experimentation demonstrated their ability to protect against

neoplastic transformation137,138. These crucial regulatory genes are now known to influence

fundamental cellular processes such as development, growth and cell cycle, genome

maintenance, differentiation, cell death and tissue regeneration137,138. There are four main

mechanisms by which tumor suppressors may act to protect cells from neoplastic transformation:

suppression of cell division, DNA damage repair, induction of apoptosis or senescence, and

inhibition of metastasis139

Page 55: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

37

Tumor suppressors that suppress cell cycle and division include RB, adenomatosis polyposis coli

(APC), alternate reading frame (ARF), retinoblastoma protein-interacting zinc finger protein

(RIZ1), cyclin-dependent kinase inhibitor 2B (CDKN2B/p15ink4b), cyclin-dependent kinase

inhibitor 2A (CDKN2A/p16ink4a), cyclin-dependent kinase inhibitor 2C (CDKN2C/p18ink4c),

p21Cip1, p27Kip1, and Trp53 (p53)140–150. Inhibition of aberrant cell division is the main

mechanism for most tumor suppressors, and many of these genes are frequently mutated in all

types of cancer. RB was the first discovered tumor suppressor. It inhibits the transcription of

factors required for cell cycle progression through repressing the activity transcription factors

like the E2Fs140,141. p15INK4B, p16INK4A, p18INK4C, p21Cip1 and p27Kip1 function by

inhibiting CDKs, which in turn inhibit RB and promote cell cycle progression144–148. Thus,

inhibition of CDKs permits RB activity, allowing it to maintain G1 phase arrest. The tumor

suppressor p53, is a transcription factor that stimulates the expression of many cell cycle

inhibitors and pro-apoptotic genes, among many other roles which will be discussed further150.

ARF in turn functions as tumor suppressor by stimulating the p53 pathway through neutralizing

the effects of p53’s two negative regulators murine double minute 2 (MDM2) and MCL-1

ubiquitin ligase E3 (MULE)149. Furthermore, independently of p53, ARF functions to antagonize

the function of major cell cycle activators such as c-MYC and E2F1149.

The proper regulation of apoptosis maintains normal homeostasis and suppresses cancer.

Consequently, cancerous cells are often resistant to apoptosis through a downregulation or

deletion of key pro-apoptotic tumor suppressors151. Tumor suppressors that promote apoptosis of

aberrant cells are p53, APC, cluster of differentiation 95 (CD95), bridging integrator 1 (BIN1),

and PTEN152–156.

The ability to fix DNA damages is known as the DNA damage response. Although the

replication of mammalian cells is a high-fidelity process with multiple checkpoints in place, the

genome is at constant risk of mutation by many sources such as ultraviolet light, ionizing

radiation and reactive oxygen species151. The ability of genes within the DNA damage response

pathway to monitor the integrity of their genome and repair damaged DNA is crucial to prevent

the transmission of mutations to daughter cells during replication, which can contribute to

malignant transformation and tumorigenesis if the given mutation is found within a tumor

suppressor or oncogene. Tumor suppressors that maintain genome integrity are mutS homolog 2

Page 56: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

38

(MSH2), mutL homolog 1 (MLH1), Ataxia-telangiectasia-mutated gene product (ATM), breast

cancer protein (BRCA), Nijmegen breakage syndrome 1 (NBS1), Fanconi-Anemia-related tumor

suppressor (FA) and p53157–161.

Although tumorigenesis is promoted by loss of regulation of the first three mechanisms of tumor

suppression discussed above, the majority of cancer deaths are caused by metastasis (the spread

of a cancer to a different organ)151. During metastasis, cancerous cells interact with endothelial

cells, signaling to initiate angiogenesis and break down vascular walls which facilitates their

spread throughout the body151. Genes like p53, metastin, breast cancer metastasis suppressor 1

(BRMS1), tissue inhibitor of metalloproteinases (TIMP1) mediator complex subunit 23

(MED23) and CD82 promote endothelial integrity, inhibit angiogenesis and the epithelial to

mesenchymal transition, and exert many other functions to suppress metastasis162–167.

It is hypothesized that in a context-specific manner, some tumor suppressors may also have an

effect on suppression of regeneration in cells that lack strong regenerative capacity137. Studying

tumor suppressors in this context could allow us to promote regeneration in organs such as the

heart that largely lack this ability137. The tumor suppressor p53 will be of focus for the rest of

this literature review as it functions in all 4 main tumor suppressor mechanisms, is mutated and

or inactivated in nearly half of all sporadic human cancers, and has been found to be implicated

in tissue regeneration and the injury response in numerous organisms.

1.8.1 The p53/Mdm2 Tumor Suppressor Circuitry

Trp53 (p53) is a tumor suppressor gene encoding for a transcription factor that is the most

frequent target for mutations in tumors168. Due to this fact, the increased susceptibility for cancer

in patients with Li-Fraumeni syndrome (individuals who inherit a mutant p53 allele, and the

spontaneous development of tumors in p53 null mice169, the importance of p53’s role in tumor

suppression is unequivocal168. It is also known that in addition to its tumor suppressive function,

p53 plays an important role in homeostasis as a cellular stress sensor. It can trigger transient or

permanent cell cycle arrest, apoptosis or DNA repair in response to many stimuli including

hypoxia, DNA damage, ribonucleotide depletion and nutrient starvation, and is involved in the

regulation of metabolism and autophagy168. Depending on the stress experienced by the cell, the

phenotypic end result following p53 activation, due to displacement of its negative regulators

Page 57: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

39

MDM2/MDM4 or MULE, is either cell recovery and survival, or death137. Additionally, p53

belongs to a family of transcription factors including p63 and p73 which have overlapping and

distinct roles168.

Much of our initial understanding of p53’s tumor suppressive roles came from the generation of

p53-knockout mice. These mice, although developmentally normal, experience early onset,

spontaneous tumors (primarily CD4+CD8+ T cell lymphomas) at 100% penetrance170–172.

Moreover, reminiscent of patients with Li-Fraumeni syndrome, p53-null heterozygous mice have

a heightened predisposition to cancer compared to wild type mice, and primarily develop

sarcomas, as well as some lymphomas and carcinomas170–172. Subsequently, more refined studies

of tissue-specific p53 knockouts have helped to highlight p53’s role in individual tissues of the

body.

1.8.2 Canonical p53 Pathways

p53’s most thoroughly characterized biochemical role within the cell is as a transcription

activator, however it also is known to repress transcription of certain genes (estimated ~15% of

target genes), as well as to help promote mitochondrial membrane permeabilization to trigger

apoptosis168. More than 80% of p53 mutations in human tumors occur within the DNA binding

domains of this protein, compromising its ability for sequence-specific DNA binding and its

function as a transcription factor168. p53 forms tetramers through a carboxy-terminus

tetramerization domain which will subsequently bind to p53 response elements within the

genome to activate transcription of p53-dependent genes168. The consensus DNA binding

sequence for p53 consists of two repeats of the 10bp motif 5’-PuPuPuC(A/T)(A/T)GPyPyPy-3’

(Pu=purine, Py=pyrimidine) typically separated by a spacer of 0-13 nucleotides168,173. These

p53-dependent genes have largely been elucidated through genetic studies and chromatin

immunoprecipitation experiments, to help determine p53’s precise role within the cell168.

p53’s contribution to tumor suppression through regulation of cell cycle arrest lies in its

transcriptional activation of important cell cycle inhibitors168. Activation of p21Cip1 by p53

occurs in response to DNA damage, which leads to G1 phase cell cycle arrest, through p21Cip1

mediated inhibition of CDK1, CDK2 and PCNA174,175. p53 also regulates other cell cycle genes

such as protein tyrosine phosphatase receptor type V (PTPRV) which is also involved in G1

Page 58: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

40

arrest176, growth arrest and DNA-damage inducible 45 (GADD45A)177 and stratifin (YWHAS)178

which control the G2/M transition175.

To promote apoptosis, p53 activates the transcription of pro-apoptotic genes such as BCL2

associated X protein (BAX)179, CD95180, death receptor 5 (DR5)173, etoposide induced 2.4

(EI24)181, p53-Induced death domain protein 1 (PIDD1)173, tumor protein P53 regulated

apoptosis inducing protein 1(p53AIP1)182, p53 up-regulated modulator of apoptosis (PUMA)183,

phorbol-12-Myristate-13-Acetate-Induced protein 1 (NOXA)184 and p53 apoptosis effector

(PERP)185,173. These genes are the downstream effectors of p53 mediated apoptosis, which is

dependent on the apoptotic peptidase activating factor 1 (APAF1)/Capsase-9 pathway and

cytochrome c release from the mitochondria173.

On top of p53’s ability to trigger cell cycle arrest or apoptosis in response to DNA damage, it

further earns its name as “guardian of the genome” through its critical role in nucleotide excision

repair186. p53 mediates nucleotide excision repair in response to DNA damage through

transcriptional activation of two genes in this pathway, DNA damage checkpoint protein 2

(DDC2)187 and XPC complex subunit, DNA damage recognition and repair factor (XPC)188,

which recognize and process UV photoproducts in the genome.

1.8.3 Non-Canonical p53 Pathways

Non-canonical roles of p53 such as its involvement in metastasis, metabolism and autophagy

have been emerging as essential for tumor suppression and homeostasis168. It is now clear that

loss of p53 not only leads to tumor development, but the ability of tumors to spread throughout

the body168,162. Loss of p53 causes an increase in expression of GTP-bound (active) RhoA, a

small GTPase that regulates cell migration, and Rho associated coiled-coil containing protein

kinase 1 (ROCK1), its main effector protein162. These proteins control actin dynamics which

promote tumor cell invasiveness. Furthermore, loss of p53 also stimulates the epithelial-to-

mesenchymal transition of tumor cells, the activation of genes that are part of the adhesive

machinery required for motility and invasion, and degradation of the extracellular matrix162.

p53 also has a widespread role over metabolism within the cell189. Several studies have shown

that p53 can regulate both glycolysis and oxidative phosphorylation189. Reportedly, p53 inhibits

Page 59: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

41

glycolysis (tumor cells have a high metabolic demand and require heightened sugar uptake,

which is facilitated loss of p53) by inhibiting the expression of the glucose transporters GLUT1

and GLUT4190, while activating the expression of the TP53 induced glycolysis regulatory

phosphatase (TIGAR, a negative regulator of glycolysis)191. p53’s restraint on glycolysis comes

with an opposing stimulation of oxidative phosphorylation, by activating transcription of many

genes involved in mitochondrial maintenance189.

Owing to reports of autophagy playing a role in tumorigenesis and in the control of metabolic

stress in healthy cells, it is not surprising that p53 has been found to directly activate this

process191. p53 can induce transcription of the 5' AMP-activated protein kinase (AMPK) and

inhibit the expression of the mechanistic target of rapamycin (mTOR), thereby promoting the

process of autophagy192. Furthermore, canonical p53 target genes such as BAX and PUMA can

also act as positive regulators of autophagy, in addition to their role as mediators of p53-

dependent apoptosis193. As metabolic dysregulation and autophagy have been implicated in

cardiovascular disease and heart failure194,195, these relatively novel functions of p53, along with

p53’s regulation of canonical signaling pathways in the heart, are interesting therapeutic avenues

to be pursued.

1.8.4 Post-Translational Regulation of p53 by MDM2

Due to the vast effect that p53 can exert on the cell, it is imperative that its activity is tightly

regulated. p53 is subject to multiple modifications including phosphorylation, ubiquitination,

methylation, acetylation, sumoylation, and neddylation, all of which intricately regulate p53

function at an epigenetic level196,197. Among these, ubiquitination, phosphorylation and

acetylation are the most extensively studied, and are involved in the regulation of all 3 steps

required for p53 activation: stabilization of the p53 protein, p53 binding to regulatory DNA

recognition elements and transcriptional activation of target gene expression196,197.

Ubiquitination is the main contributor to p53 stability, and this job is largely assumed by the

oncoprotein, MDM2.

p53’s function is predominantly regulated by its protein stability, and has a short half-life of 5-30

minutes in normal unstressed cells196. This knowledge led to the search for a protein that could

Page 60: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

42

degrade p53, thereby explaining this very short intracellular half-life196. Immunoprecipitation

experiments identified this protein as MDM2, which forms a 90kDa complex with p53198.

Mdm2 is an E3 ubiquitin ligase that targets p53 for proteolytic degradation196,197,199. MDM2 was

first discovered as a gene that was overexpressed by more than 50-fold in a spontaneously

transformed mouse cell line (3T3-DM)200. The reason for the transformation potential of

overexpressed MDM2 was discovered, when it was shown to bind p53 and inhibit its

transcriptional transactivation capacity198. Similarly, it was found that MDM2 gene amplification

occurs in over 1/3 of human sarcomas that maintain wild type p53201.

Ubiquitination of p53 by MDM2 occurs in a series of steps that involves 3 enzymes196,197. The

E1 enzyme binds and activates the ubiquitin protein, E2 then accepts the activated ubiquitin and

transfers it to the E3 enzyme (MDM2 in this case), which is a substrate-specific ligase that

covalently bonds ubiquitin to its substrate (p53)196,197,199. MDM2 monoubiquitinates p53 at

multiple lysine residues, which is the critical step in mediating its degradation by nuclear and

cytoplasmic proteasomes196,197,199.

Mdm2 itself is the product of a p53 inducible gene197. Therefore, the two proteins form an

autoregulatory negative feedback loop that tightly maintains p53 levels within the cell197.

Moreover, this relationship limits the duration and severity of p53-dependent biological

responses following a non-lethal stressor to the cell199. In turn, low levels of p53 activity result in

constitutively low MDM2 levels197. To escape the negative regulation of MDM2 upon cellular

stress, p53 and MDM2 both become phosphorylated by diverse kinases, preventing MDM2’s

ability to bind its substrate202,203. Unlike the p53 knockouts, MDM2-null mice experience a very

early embryonic lethality, due to an overactive p53, impairing organ development204,205.

Consequently, concurrent knockout of p53 and MDM2 rescues the embryonic lethality of

MDM2-null mice204. MDM2 therefore plays an essential role in tumorigenesis as well as

homeostasis, by antagonizing the function of p53.

1.8.5 p53/MDM2 and the Heart

The development of primary cardiac tumors is extremely rare, with a prevalence of 0.001–0.03%

in autopsy series206. Based on this low prevalence of tumorigenesis, and the hearts low

Page 61: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

43

regenerative capacity, tumor suppressors may be exerting an important growth-limiting role

within this organ. The exact function of the p53/MDM2 tumor suppressor circuitry in the heart

has remained elusive. Some involvement of p53 and cardiovascular disease however, has been

inferred. It has been shown that cardiomyocyte-specific deletion of p53 in the heart is capable of

protecting against the transition of hypertrophy to heart failure207. p53 ablation resulted in an

upregulation of the pro-angiogenic hypoxia-inducible-factor-1 (HIF-1) which increased the

number of microvessels in the heart, and improved systolic function compared to wild type mice

subjected to the same insult207. Furthermore, the authors showed that following pressure

overload, p53 and phosphorylated p53 levels accumulate in the heart, and concluded that this

upregulation may be partially responsible for the detrimental effects on heart function

experienced by these mice207. Four studies have also demonstrated that p53 becomes

upregulated in response to cardiac ischemia, and that inhibition of p53 conveyed a protective

effect through reduction of apoptosis, via a decrease in BAX expression, and ameliorated

ventricular remodeling208–211. More recently, Yoshida et al. (2015) demonstrated that the

accumulation of p53 during cardiac pressure overload occurs also in cardiac endothelial cells212.

Upregulation of p53 in endothelial cells resulted in an upregulation of the intercellular adhesion

molecule-1, which is essential for the infiltration of inflammatory cells into the heart upon

pressure overload212. Therefore, upregulation of p53 following pressure overload contributes to

the inflammatory response induced by this insult, and exacerbates cardiac dysfunction212.

Although some research has been conducted on the effect of p53-dependent apoptosis in the

heart following pressure overload or hypoxia induced stress, gaps of knowledge exist in our

understanding of other functions that this tumor suppressor circuitry exerts on the heart, such as

cell cycle, regeneration and metabolism. Furthermore, few studies have examined the

combinatorial effect of p53 and MDM2 in the heart. The Billia lab hypothesized that based on

p53 and MDM2’s expansive control over cell cycle in many other organs, an explanation for the

heart’s low regenerative potential must lie, at least in part, within this pathway213. Research in

our laboratory sought to determine if the p53/MDM2 complex acts as an intrinsic inhibitor of

adult cardiomyocyte proliferation. To do this, p53 and MDM2 were inactivated specifically in

the adult mouse heart employing the Cre-loxP recombination system213. Using an inducible

cardiomyocyte-specific transgenic mouse, mcm, in which the cardiac muscle α-myosin heavy

Page 62: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

44

chain 6 promoter drives the expression of the Cre recombinase fused to two mutant estrogen-

receptor ligand binding domains, when exposed to 4-Hydroxytamoxofen (Tam) injections,

MDM2 (Mdm2KO), p53 (p53KO), and both proteins (double knockout, DKO), were knocked

out, specifically in the adult mouse heart213.

These three different mice had strikingly different phenotypes214. As expected, the p53KO did

not immediately show any adverse effects, but exhibited mortality by 6 months post-Tam. By

this time point, these mice exhibited hypertrophy and increases in fetal gene expression. Loss of

p53 also led to a decrease in two cardiac transcription factors, GATA4 and NKX2.5, identifying

these genes as previously unknown targets of p53 in the heart. As previously mentioned, the role

of p53 in controlling energy production and mitochondrial function has recently been brought to

light. Since the heart is a biomechanical pump with an enormous demand for ATP, this requires a

high mitochondrial content within the cardiomyocyte. It was therefore hypothesized that p53’s

role in energy regulation would be crucial within the heart. Indeed, by the 6 month time period,

ATP content significantly dropped. Thus, p53 is clearly essential within the heart to protect

cardiomyocytes against aberrant growth and metabolic transformation. Next, p53’s canonical

role of promoting cell cycle arrest was investigated in the heart. Interestingly, knockout of p53

alone was not sufficient to induce cardiomyocyte proliferation.

Conversely, the Mdm2KO mice had high early onset mortality by 14 days214. As expected, due

to high levels of p53 in the absence of MDM2, these mutant hearts exhibited significantly

increased cardiomyocyte apoptosis, accompanied by a reduced fractional shortening.

Furthermore, unlike the p53KO mice, energy balance defects were less pronounced.

Additionally, loss of MDM2 did not induce cardiomyocyte proliferation, as expected, due to

elevated levels of p53214.

Tam-induced homozygous ablation of both p53 and MDM2 caused dramatic early onset death of

all mice by day 10214. These results indisputably reveal that p53 and MDM2 are integral players

in a pathway that maintains normal cardiac performance. DKO mice developed dilated

cardiomyopathy with wall thinning, increased chamber sizes and substantial fibrosis, but had no

indications of hypertrophy. Furthermore, the cardiomyocytes exhibited sarcomeric structure

abnormalities, corresponding to a markedly impaired fractional shortening. Many genes within

Page 63: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

45

the fetal gene program were reactivated, with a concurrent decline in the expression of essential

cardiac genes. Similar to the p53KO, the DKO mice experienced a drop in energy production,

but at a much more dramatic rate. By 8 days post-Tam, ATP levels dropped significantly, and

this corresponded with a drastic reduction in mitochondrial copy number, mitochondrial gene

transcription, and a significant downregulation in mitochondrial Complex I-V genes.

Furthermore, as previously mentioned, p53 inhibits glycolysis and promotes fatty acid oxidation

(the main mechanism of energy production in the heart), and loss of p53 led to a significant

decrease in canonical p53 target genes that maintain fatty acid oxidation including AMPKα, and

TIGAR, concordant with an increase in glycolytic markers such as the RELA proto-oncogene,

NF-kB subunit (RELA, a crucial activator of the glycolytic program). These results revealed that

p53 and MDM2 are crucial for the maintenance of mitochondrial bioenergetics and energy

metabolism. Investigation of cardiomyocyte proliferation in the DKO mice revealed another

striking and unexpected phenotype. These mice had a significant upregulation of

cardiomyocytes in S phase, M phase and undergoing cytokinesis, in all four chambers of the

heart. Thus, it appears reasonable to assume that on top of p53, MDM2 regulates important cell

cycle activators within the heart, and that removal of both the negative regulator (p53) and the

inhibitor of positive regulators (MDM2) is required to induce cardiomyocyte proliferation. It

seems as though in the heart, multiple roadblocks are in place that must be removed

simultaneously to stimulate cell cycle re-entry214.

In all, there is an abundant amount of literature that demonstrates the role of p53 as an essential

contributor to growth arrest and apoptosis in response to various types of stress. Through

analysis of three heart specific mutant mice (p53KO, Mdm2KO and DKO) Billia et al. (2016)

established p53 and MDM2 to also be necessary under physiological conditions of low stress in

the heart, where they act to control heart function through the maintenance of cardiomyocyte

differentiation, hypertrophy, energy production/balance, and cell cycle arrest214.

1.8.6 p53 and miRNAs

It has been hypothesized that miRNAs evolved, to allow organisms to deal with stress, as

miRNAs have been shown to play integral roles in cell cycle, apoptosis, tumor

suppression/oncogenesis, metabolism, and much more215,216. In keeping with this, miRNAs have

Page 64: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

46

emerged as important executors of p53 dependent tumor suppression and stress responses217,218.

miRNAs are heavily interconnected in the p53 stress response pathway, not only because they

can be induced by p53, but because p53 is a downstream target of many miRNAs217.

The mechanism by which p53 mediates repression of target genes has been unclear, but it is now

thought that miRNAs contribute to a large majority of the mRNA and protein expression

downregulation observed following p53 activation217. Besides regulating the expression of

specific miRNA-coding genes, p53 also affects the processing of miRNAs directly through

interaction with DDX5 to enhance its interaction with the Drosha complex, promoting miRNA

biogenesis219. The discovery of many p53-dependent miRNAs has therefore expanded p53’s role

within the cell allowing it to indirectly regulate the expression of a multitude of genes. In

response to stress, p53-regulated miRNAs become expressed, engage in many ‘feedforward’ and

‘feedback’ loops that control amplification, robustness, fine-tuning and buffering of signals, all

of which contribute to the appropriate cellular response that occurs.

Several groups initially identified miR-34a and miR-34b/c as the first direct miRNA

transcriptional targets of p53, and have demonstrated that they help mediate many of functions of

p53 including induction of apoptosis, senescence and cell cycle arrest220–226. Since then, many

other p53-dependent miRNAs have been discovered including miR-15a/16-1, miR-145, miR-

200, miR-192, miR-104, miR-215, the let-7 family, miR-107, miR-149227, miR-29, miR-605217.

Recently, Hunten et al. (2015) undertook a high throughput approach, utilizing pulsed stable

isotope labeling, next generation sequencing, and chromatin immunoprecipitation, to determine

the genome wide effect of p53 upregulation in a colorectal cancer cell line228. They found that

p53 induction (using a vector based technique to overexpress p53) led to widespread differential

regulation of proteins (542 up, 569 down), mRNAs (1258 up, 415 down), miRNAs (111 up, 95

down), and long non-coding RNAs (270 up, 123 down)228. Transcriptionally induced genes

displayed more occupied p53 binding sites than repressed genes, suggesting indirect mechanisms

of repression, such as through miRNAs, are at play. Moreover, 50% of the transcriptionally

repressed genes contained seed-matching sequences to p53-induced miRNAs in their 3’-

UTRs228. This study further supports the notion that p53-dependent gene repression is mediated

Page 65: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

47

indirectly through p53-induced miRNAs. A full table of p53-regulated miRNAs can be found in

Supplemental Figure 11 of Hunten et al. (2015)228.

Many p53-regulated miRNAs have been shown to have robust effects on cell cycle, and

themselves act as tumor suppressors217. Numerous studies have shown distinct p53-regulated

miRNAs to be downregulated by genetic or epigenetic mechanisms in diverse human tumors,

and that their re-introduction through miRNA mimic technology can mitigate tumor cell

proliferation and improve cancer outcomes217. Cell cycle-related factors have bioinformatically

been shown to be over-represented in the profiles of mRNAs targeted by p53-regulated miRNAs

such as miR-34a220–222. For example, CDK4, CDK6, cyclin E2, E2F3, c-MYC and n-MYC are

all downregulated by miR-34a220–222. p53 also promotes the expression of miR-145 such that it

can downregulate MYC, CDK4 and CDK5229. As c-MYC is also repressed by p53 independently

of miRNAs230, miR-145 and miR-34a engage in a feedforward loop that ensures the robust

suppression of c-MYC expression. It therefore follows that their downregulation in cancer would

lead to the relief of inhibition over a large number of cell cycle regulators that can promote

tumorigenesis (Figure 1.9).

In addition to proliferation, the miR-34 family has been shown to induce apoptosis, when

upregulated, in multiple cell lines, likely by repressing the expression of many mRNA targets

Figure 1.9 Regulation of the cell cycle by p53-induced miRNAs217. Cell cycle regulators and effectors

repressed by p53-induced miRNAs are grouped according to the cell cycle phase in which they are mainly

expressed or most functionally relevant. p53-induced miRNA mediated inhibition of these factors may cause or

contribute to cell cycle arrest in the respective phase of the cell cycle in which they are required. Downregulation

of these miRNAs may therefore promote cell cycle progression. Originally published by Hermeking, H. Nat. Rev.

Cancer 12, 613–626 (2012)217. Used with permission of the author and publisher.

Page 66: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

48

including B-cell lymphoma 2 (BCL2)222, survivin231, sirtuin 1 (SIRT1)232 and snail family

transcriptional repressor 1 (SNAI1)233. As apoptosis is a fundamental feature of ageing

cardiomyocytes, Boon et al. (2013) investigated if the p53-dependent miR-34 family played a

role in cardiac ageing234. They discovered that the miR-34 family is significantly upregulated in

aged mouse hearts, and that miR-34 expression, but not muscle specific miR-1 or miR-133

expression, was correlated with age in human cardiac biopsies235. With this knowledge, they

inhibited the miR-34 family using antagomirs in 18 month old mice, and saw that this treatment

significantly reduced age-associated cell death in the heart235. Young mice treated with anti-miR-

34 were phenotypically similar to their littermates, whereas old anti-miR-34 treated mice

exhibited less hypertrophic cardiomyocytes, reduced cardiomyocyte apoptosis and preserved

cardiac function, compared to non-treated littermates235. They concluded that the upregulation of

miR-34 by p53 in aged hearts contributes to the progressive decline in heart function throughout

the aging process235. Since this study, it was shown that p53-dependent miRNAs are not only

involved in cardiac ageing, but that circulating p53 responsive miRNAs become upregulated in

the serum of patients post-AMI who subsequently develop heart failure103. Matsumoto et al.

(2013) found that miR-192, miR-194 and miR-34a all become upregulated in the serum of

patients from the OACIS study who developed heart failure following myocardial infarction236.

As the development of heart failure following myocardial infarction is positively correlated with

infarct size, it follows that a larger stressor may more strongly activate the p53 stress response,

leading to an upregulation of p53-responsive miRNAs in these patients. They concluded that

these miRNAs may be of value for predicting the risk of heart failure development following

myocardial infarction103.

Based on these studies, understanding the role of p53-regulated miRNAs can provide insight into

the mechanisms of aging and cardiovascular disease in the heart. Furthermore, it may be possible

to exploit the function of p53-regulated miRNAs in the heart for the development of cardiac

related therapies. Knowledge gaps reside in our understanding of whether MDM2 plays a role in

miRNA regulation, and which miRNAs are regulated by the p53/MDM2 tumor suppressor

complex specifically in the heart under physiological conditions of low stress. Identifying the

specific miRNAs which are regulated by p53/MDM2 in the heart may provide more insight into

Page 67: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

49

how these proteins exert their crucial homeostatic roles of preserving cardiac function,

bioenergetics and cardiomyocyte cell cycle exit.

Page 68: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

50

Chapter 2

Rational Hypothesis

2 Hypothesis and Thesis Aims

Adult cardiomyocytes are terminally differentiated cells that exist in a non-proliferative, post-

mitotic state237,238. Cell cycle exit occurs shortly after birth, and coincides with a concomitant

decrease in the expression of cell cycle activators and increase in the expression of cell cycle

inhibitors, resulting in adult cardiomyocytes that are refractory to proliferative stimuli81. Billia et

al. (2016) have demonstrated that inactivating both the tumor suppressor p53 and its negative

regulator MDM2, specifically in the heart (double knockout mouse, DKO), can allow

differentiated cardiomyocytes to regain their proliferative capacity, through an upregulation of

factors involved in cell cycle re-entry (cyclins and cyclin-dependent kinases) and a

downregulation of cell cycle inhibitors. Specifically, DKO mice exhibited a significant

upregulation in CDK2 kinase activity, as well as an upregulation in the expression of cyclin E,

both of which are crucial for cell cycle reactivation. Concurrently, the expression of p21Cip1 and

p27Kip1, two major inhibitors of cell cycle progression, was downregulated by up to 90%. These

changes within the cell were accompanied by a significant increase in cardiomyocytes in all

phases of the cell cycle, including cytokinesis. Importantly, knockout of either gene individually

(p53KO and Mdm2KO) failed to promote cardiomyocyte cell cycle re-entry. Further

transcriptome analysis revealed that loss of p53 and MDM2 caused a widespread upregulation in

cell cycle regulators, indicating that p53/MDM2 repress their expression in wild type

cardiomyocytes, promoting their quiescence. The ability of p53 to directly repress gene

expression, however, is debated. What is known is that p53 can mediate transcriptional

repression indirectly, through the activation of microRNAs (miRNAs). As many miRNAs have

been shown to function as potent tumor suppressors, we hypothesized that in the heart, there are

a subset of miRNAs regulated by the dual presence of both p53 and MDM2, which function to

downregulate important cell cycle activators and maintain adult cardiomyocyte quiescence.

Upon knockout of p53 and MDM2 in the heart, the expression of this subset of p53/MDM2-

regulated miRNAs is lost, which alleviates the inhibition on multiple cell cycle regulators and

permits cardiomyocytes to regain proliferative capacity.

Page 69: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

51

2.1 Hypothesis

In the heart, p53 and MDM2 regulate a set of “anti-proliferative” microRNAs that maintain

cardiomyocyte cell cycle arrest. Inactivation of these p53/MDM2-regulated miRNAs will

alleviate their inhibition of crucial cell cycle regulators and thus, promote cardiomyocyte cell

cycle re-entry and proliferation.

2.2 Thesis Aims

Aim 1: Identification of p53/MDM2-regulated miRNAs involved in the regulation of cell cycle.

a) Perform a miRNA microarray analysis comparing hearts from p53KO,

Mdm2KO and DKO mice to identify miRNAs regulated by both p53 and

MDM2.

b) Perform target gene analysis of identified p53/MDM2 regulated miRNAs to

determine their involvement in cell cycle regulation.

c) Develop antagomirs that can inhibit identified “anti-proliferative” p53/MDM2-

regulated miRNAs.

Aim 2: Inhibition of identified miRNAs in vitro in rat neonatal cardiomyocytes using

antagomirs.

a) Transfect cardiomyocytes with a cocktail of antagomirs targeting the identified

p53/MDM2-regulated miRNAs and determine if this treatment promotes

cardiomyocyte proliferation.

b) Determine if putative miRNA target genes involved in cell cycle regulation

become upregulated following antagomir treatment to elucidate which factors

support cardiomyocyte cell cycle re-entry.

Page 70: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

52

Chapter 3

Data Chapter

3 Control of Cardiomyocyte Proliferation by p53/MDM2-regulated microRNAs

Authors: Shanna Stanley-Hasnain1,2, Ludger Hauck2, Daniela Grothe2, and Filio Billia1,2,3,4*

Affiliations: 1Institute of Medical Science, University of Toronto, 1 King's College Circle, Toronto, Ontario Canada M5G 1A8 2Toronto General Research Institute, Toronto, Ontario Canada, 100 College St., M5G 1L7 3 Division of Cardiology, University Health Network (UHN), Toronto, Ontario, Canada, 200

Elizabeth St., Toronto, Ontario Canada, M5G 2C4 4Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto

Formatted for submission to the journal Molecular Therapy – Nucleic Acids

Page 71: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

53

3.1 Introduction

Trp53 (p53) is a tumor suppressor gene encoding a transcription factor that is the most frequent

target for mutations in tumors168. In addition to its tumor suppressive abilities, p53 acts as a

cellular stress sensor239–241, and plays an important role in homeostasis213 and senescence242–244.

It can trigger transient or permanent cell cycle arrest, apoptosis or DNA repair in response to

many stimuli including hypoxia, DNA damage, ribonucleotide depletion and nutrient starvation,

and is involved in the regulation of metabolism and autophagy168,245. Based on the observations

that p53-knockout mice develop early onset spontaneous tumors with 100% penetrance171, that

individuals with Li-Fraumeni syndrome who inherit a mutant p53 allele have heightened

susceptibility for tumorigenesis169, and that cells with dysfunctional p53 exhibit aberrantly high

levels of proliferation246, the importance of p53’s role in growth control is unequivocal.

Due to the vast effect that p53 can exert within the cell, it is imperative that its activity is tightly

regulated. Post-translational ubiquitination is one of the means by which to control p53 stability.

This is largely mediated by the oncoprotein murine double minute 2 (MDM2)247. MDM2 is an

E3 ubiquitin ligase that monoubiquitinates p53 at multiple lysine residues, a critical step in

promoting its degradation by nuclear and cytoplasmic proteasomes196,197,199. Interestingly,

MDM2 itself is the product of a p53 inducible gene, creating a negative feedback loop that

tightly maintains p53 levels within the cell197. In turn, low levels of p53 activity result in

constitutively low MDM2 levels197. This feedback loop allows the cell to limit the duration and

severity of p53-dependent biological responses199. Unlike p53 knockouts, MDM2-null mice

experience a very early embryonic lethality, due to high levels of p53, impairing organ

development through an excessive induction of apoptosis and diminished cell proliferation204,205.

The most thoroughly characterized biochemical role of p53 within the cell is as a transcriptional

activator, however it also is known to repress transcription of certain genes (estimated ~15% of

target genes). As a transcription factor, p53 mediates cell cycle arrest through activation of

important cell cycle inhibitors168. Cell cycle target genes transcriptionally regulated by p53

include cyclin-dependent kinase inhibitor 1A (p21Cip1), and upregulation of p21Cip1 expression by

p53 leads to G1 phase cell cycle arrest, through p21Cip1-mediated inhibition of cyclin-dependent

kinase 1 (CDK1), cyclin-dependent kinase 2 (CDK2) and proliferating cell nuclear antigen

Page 72: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

54

(PCNA)174,175. Further genes activated by p53 which have cell cycle regulatory functions include

the protein tyrosine phosphatase receptor type V (PTPRV) which is also involved in G1 arrest176,

growth arrest and DNA-damage inducible 45 (GADD45a)177 and Stratifin (YWHAS)178 which

control the G2/M transition175.

In contrast, p53-mediated transcriptional repression has been a contentious area of research, and

direct mechanisms of p53 repression have recently been refuted in a computational meta-analysis

of 6 genome-wide analyses of p53 dependent gene expression, published by Fischer et al.

(2014)248. Regardless, it has been noted that activation of p53 leads to the downregulation of cell

cycle activators, beyond those targeted by p21Cip1 activation228.

Recently, microRNAs (miRNAs) have been suggested as the main mediators of p53-regulated

gene repression228. miRNAs are a group of small non-coding RNAs with the capability of

expansive genetic regulation. This class of endogenous 21-25 nucleotide long, single-stranded

RNA molecules function as guide molecules in RNA silencing through base pairing to their

complementary mRNA targets82–84. By targeting specific mRNAs for degradation or translational

repression, miRNAs act as inhibitors of gene expression, and are involved in almost all

developmental and pathological processes in animals, including multiple pathways regulated by

the tumor suppressor p5382. Hunten et al. (2015) demonstrated that upon p53 activation, 111

miRNAs become upregulated, indicating that there is subset of miRNAs transcriptionally

activated by p53 (p53-regulated miRNAs)228. Furthermore, in this study, of the 415 genes

downregulated following p53 activation, 50% contained complementary seed-matching

sequences to p53-regulated miRNAs in their 3’-UTRs228. This supports the notion that p53-

dependent gene repression can be mediated indirectly through p53-regulated miRNAs.

Many p53-regulated miRNAs have been shown to have robust effects on cell cycle, and

themselves act as tumor suppressors217. Numerous studies have demonstrated the downregulation

of distinct p53-regulated miRNAs by genetic or epigenetic mechanisms in diverse human

tumors, and that their re-introduction through miRNA mimic technology can mitigate tumor cell

proliferation and improve cancer outcomes249–252. Cell cycle-related factors have

bioinformatically been shown to be over-represented in the profiles of mRNAs targeted by p53-

regulated miRNAs such as miR-34a220–222. For example, cyclin-dependent kinase 4 (CDK4),

Page 73: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

55

cyclin-dependent kinase 6 (CDK6), cyclin E2, E2 factor 3 (E2F3), V-Myc avian

myelocytomatosis viral oncogene homologs (c-MYC and n-MYC) are all downregulated by

miR-34a220–222. p53 also promotes the expression of miR-145 such that it can downregulate c-

MYC, CDK4 and cyclin-dependent kinase 5 (CDK5)229. Furthermore, certain p53-regulated

miRNAs have been shown not only to play a role in cancer, but in other diseases such as

cardiovascular disease103,253. As miRNAs are involved in a myriad of signaling and biological

processes which regulate growth and regeneration, cardiac function, homeostasis, and the

progression of cardiac remodeling, hypertrophy and heart failure, exploiting their function

through miRNA based therapies have been previously studied in cardiovascular disease and heart

failure254.

Heart failure is a leading cause of morbidity and mortality in Canada. Adult cardiomyocytes are

terminally differentiated cells that exist in a non-proliferative, post-mitotic state237,238. Thus, the

regenerative capacity of the heart is poor, and cardiac tissue is particularly vulnerable to injury.

This is emphasized by the fact that cardiomyocyte loss due to insults such as myocardial

infarction is a major contributing factor to the development of heart failure255,256. As the quality

of life and prognosis for these patients is poor, therapies to prevent and treat heart failure are

urgently needed28. Therefore, determining the roadblocks responsible for maintaining

cardiomyocyte cell cycle exit lies at an important crossroad in the development of potential

regenerative therapies to promote cardiac repair following injury. Given p53’s crucial role in

promoting growth arrest, Billia et al (2016) sought to determine the function of the p53/MDM2

tumor suppressor circuitry in the heart213,214. They demonstrated that inactivation of both p53

and MDM2 specifically in the heart (double knockout mouse, DKO) can allow differentiated

cardiomyocytes to regain proliferative capacity, through an upregulation of factors involved in

cell cycle re-entry (cyclins and cyclin-dependent kinases) and a downregulation of cell cycle

inhibitors. Importantly, knockout of either two genes individually (p53KO and Mdm2KO) was

not sufficient to promote cardiomyocyte cell cycle re-entry. Based on p53’s known ability to

indirectly repress gene expression through the activation of miRNAs, and the ability of miRNAs

to function as potent tumor suppressors, we hypothesized that in the heart, there are a subset of

miRNAs regulated by the dual presence of both p53 and MDM2, that function to downregulate

important cell cycle activators and maintain adult cardiomyocyte quiescence. Upon knockout of

Page 74: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

56

p53 and MDM2 in the heart, the expression of this subset of p53/MDM2-regulated miRNAs is

lost, which alleviates the ihibition on multiple cell cycle regulators and permits cardiomyocytes

to regain proliferative capacity.

The aim of this study was to characterize the effect of p53 and MDM2 in the heart through their

ability to regulate the expression of miRNAs. Herein, we identified a unique subset of miRNAs

that become downregulated upon the loss of both p53 and MDM2, in the heart. This subset of

miRNAs were enriched for target genes involved in cell cycle regulation, and their inhibition

using antagomirs significantly promoted wild type neonatal rat cardiomyocyte proliferation.

These data support the existence of a novel role for p53 and MDM2 in the heart, in maintaining

cardiomyocyte cell cycle arrest through regulating a subset of anti-proliferative miRNAs.

Page 75: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

57

3.2 Methods

3.2.1 MDM2 and p53 conditional mutant mice

All animal usage in this study was in accordance with approved institutional animal care

guidelines of the UHN (AUP 1815/1379, Canadian Council in Animal Care). The mcm

transgenic mice on a C57BL/6J background were obtained from Jackson (Bar Harbor,

ME04609 USA; strain name: B6.FVB(129)-Tg(Myh6-cre/Esr1*)1Jmk/J; stock number:

005657). The Mdm2f/f mice (strain number 01XH9) and p53f/f mice (strain number 01XC2)

were obtained from the mouse repository of the National Cancer Institute/National

Institutes of Health at Frederick (Rockville, MD 20852 USA). The Mdm2f/f and p53f/f strains

were backcrossed onto C57BL/6J inbred mice (Jackson; stock number 000664) for at least eight

generations. Age-matched syngeneic adult male mice (12-13-week-old; 22-27 g body weight)

were used in this study. All experiments utilized controls of matched age and sex. Genotyping

was carried out by PCR employing alkaline hydrolysis of genomic DNA isolated from tail tips

using the Terra PCR Direct Kit (mcm, Mdm2f/f, p53f/f; Clontech) or AccuStart II GelTrack PCR

Supermix and the following oligonucleotides:

mcm forward 5’-AGGTGGACCTGATCATGGAG-3’

mcm reverse 5’-ATACCGGAGATCATGCAAGC-3’

Mdm2f/f forward 5’-CTGTGTGAGCTGAGGGAGATGTG-3’

Mdm2f/f reverse 5’-CCTGGATTTAATCTGCAGCACTC-3’

p53f/f forward 5’-CACAAAAACAGGTTAAACCCAG-3’

p53f/f reverse 5’-AGCACATAGGAGGCAGAGAC-3’

To achieve inactivation of MDM2 and p53 in the adult mouse heart, we used the Cre-loxP

recombination system of bacteriophage P1. We employed an inducible cardiomyocyte-specific

transgenic mouse in which the cardiac muscle D-myosin heavy chain 6 (Myh6) promoter

drives the expression of tamoxifen (Tam)-inducible Cre recombinase protein fused to two

mutant estrogen- receptor ligand-binding domains (mcm). In this strain, the mcm fusion

protein is expressed only in cardiomyocytes, not in non-cardiomyocytes, and is retained in the

Page 76: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

58

cytoplasm. Administration of Tam induces mcm nuclear translocation thereby permitting Cre-

mediated recombination. This system allowed us to explore conditional mutations of

p53/Mdm2 that might differ from the germ line absence of both of these factors in the adult

heart. Next, we crossed mcm transgenic mice with mice carrying conditional Mdm2f/f and

p53f/f alleles to obtain Mdm2f/f; p53f/f;mcm (DKO), Mdm2f/f;mcm (Mdm2KO) and p53f/f;mcm

(p53KO) mice on a C57BL/6J background. In Mdm2KO mice, Mdm2 exons 7 to 9 are flanked

with loxP sites to facilitate inactivation of the RING finger domain which exerts ubiquitin

ligase activity towards p53. p53KO mice carry loxP sites in p53 introns 1 to 10 to ensure Cre-

mediated elimination of the majority of the coding sequence. In the absence of Tam, animals of

all these strains develop normally and lack an obvious phenotype.

We injected an ethanol/peanut oil (P2144, Sigma-Aldrich) emulsion of 4-Hydroxytamoxifen

(H6278, Sigma-Aldrich) intraperitoneally. Tam-treated adult Mdm2f/f, p53f/f, Mdm2f/f; p53f/f and

mcm mice were analyzed by immunohistology and echocardiography rendering no evidence of

fibrosis or cardiac dysfunction. To achieve conditional genetic ablation of MDM2 and p53

in adult cardiomyocytes in vivo, Tam was intraperitoneally injected for four consecutive days.

The day of the fourth Tam injection was arbitrarily set as day zero.

3.2.2 RNA Isolation

Total RNA, including the small RNA fraction, was isolated from mouse hearts (n= 3 hearts per

strain) or neonatal rat cardiomyocytes using the standard Trizol method (Invitrogen). To isolate

RNA from cultured neonatal rat cardiomyocytes, Trizol was also used. 300 µl of trypsin was

added per well of a 6 well plate and incubated for 5 minutes at 37 degrees Celsius. The reaction

was halted by 700 µl of media containing 10% horse and fetal bovine serum. Cells were

subsequently scraped and pelleted by centrifugation. 500ml of Trizol was added to each cell

pellet, and a 28 gauge syringe was utilized to rupture the cell membranes and release the nucleic

acids. Phase Lock Gels (5PRIME) were utilized to ensure a pure and distinct organic and

aqueous phase separation (gel creates a physical barrier between the two phases), leading to the

isolation high quality non-contaminated RNA. Cardiomyocyte and fibroblast RNA concentration

and purity was measured both by the NanoDrop 2000 spectrophotometer (Thermo Scientific)

and the Qubit 2.0 fluorometer (Thermo Fisher Scientific) prior to utilization for RT-qPCR. The

Page 77: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

59

RNA isolated from mouse hearts which was used for the Nanostring miRNA assay was passed

through a Bioanalyzer (Agilent) by the Princess Margaret Genomics Centre core facility to more

accurately assess the quality of the RNA. Only RNA with an RNA Integrity Number (RIN) of

≥9.0 (out of 10), was utilized for the miRNA assay.

3.2.3 Nanostring nCounter miRNA assay

33 ng of total RNA from each heart was used as input for the nCounter mouse miRNA assay and

was and processed according to the manufacturer’s protocols. For each sample, a high-density

scan (1155 fields of view) was performed. Raw array data was processed using the Nanostring

nSolver Analysis Software 2.5. The mean of the negative controls plus one standard deviation

was subtracted for background correction. Normalization of the data for sample/RNA content

was performed using the geometric mean of the top 100 most highly expressed genes.

Normalized miRNA expression levels were transformed from ratios (treatment vs. vehicle

control) to fold change values and analyzed using a t-test to identify significantly differentially

expressed miRNA between samples. Heatmaps were generated with the fold change values using

Euclidean distance as the distance metric and the average as the linkage method. Pathway

analysis was executed with the online platform miRSystem ver. 20150312.

3.2.4 miRNA RT-qPCR

Total RNA isolated from total hearts, cardiomyocytes or fibroblasts was reverse transcribed

using the qScript microRNA cDNA Synthesis Kit (Quanta BioSciences, Inc.) using an initial

input of 500ng of RA. The RNA first undergoes a polyadenylation step, followed by reverse

transcription with an oligo-dT adapter primer which has a unique sequence at its 5’ end allowing

amplification of the cDNAs in quantitative PCR reactions (qPCR). qPCR is carried out using the

Roche Light Cycler 480, a PerfeCTa miRNA assay unique for each individual miRNA along

with the PerfeCTa Universal PCR Primer (specific to the unique sequence of the oligo-dT

adapter primer) and PerfeCTA SYBR Green SuperMix (5ng cDNA/25µl reaction). Relative

miRNA expression and fold change was calculated by the standard ΔΔCt method, and was

normalized to the endogenous control RNU6 (small RNA component of spliceosome) using the

LC480 SW 1.5.1 analysis software.

Page 78: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

60

3.2.5 Western Blot

Protein was isolated from mouse hearts and neonatal rat cardiomyocytes using the NE-PER

Nuclear and Cytoplasmic Extraction Kit (ThermoFisher) according to the manufacturer’s

protocol. For Western Blot, we used SDS/PAGE gels (4–12% Bolt Bis-Tris Plus; Invitrogen),

PVDF membranes (iBlot; Invitrogen), primary antibodies against p53 (Enzo) and Mdm2

(Sigma), horseradishperoxidase (HRP) conjugated secondary antibodies (Cell Signaling), and the

Luminata Crescendo HRP substrate for chemiluminescence detection (EMD Millipore). Blots

were imaged with the BioRad ChemiDoc XRS+ and analyzed with Image Lab 5.0 software.

3.2.6 Neonatal rat cardiomyocyte and fibroblast isolation and culture

Left ventricular cardiomyocytes from 2 to 3 day old wild type Wistar rats (Charles River) were

isolated using collagenase II (0.5 mg/mL, Invitrogen) and pancreatin (1 mg/mL, Sigma). To

obtain highly pure cardiomyocyte cultures, one round or “pre-plating” was utilized. The cardiac

digestion mixture was plated onto 10cm dishes and incubated for 50 minutes. This is sufficient

time to allow cardiac fibroblasts to adhere to the plates, but not cardiomyocytes, as described

previously257. The supernatant containing a purified population of cardiomyocytes was removed

from the 10 cm plates and plated onto Collagen I coated cell culture dishes (1x106 cells/well in 6

well plates and150,000 cells/well in 48 well plates) (Corning). Cardiomyocytes were cultured in

DMEM/F12 medium containing 3 mM Na-pyruvate, 2 mM glutamine, antibiotics (Gibco), 0.2%

bovine serum albumin (Sigma), 0.1 mM ascorbic acid, 10% fetal bovine serum (Sigma), 10%

horse serum (Sigma) and 25 µM arabinosylcytosine (AraC; Sigma) to inhibit remaining non-

cardiomyocyte proliferation for 48 hours prior to treatment or analysis. 500,000 fibroblasts were

seeded/well in 6 well plates, and cultured in DMEM/F12 medium containing 10% fetal bovine

serum (Sigma), 10% horse serum (Sigma) and antibiotics. Cells were cultured to confluency

prior to isolation of RNA for RT-qPCR.

3.2.7 miRNA antagomir development

miRIDIAN microRNA Hairpin Inhibitors were ordered from Dhamarcon, Thermo Scientific

(5nmol). The Anti-cel-67 targeting a Caenhabdoartisi elegans miRNA not expressed in

mammals was employed as a negative control.

Page 79: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

61

3.2.8 Neonatal rat cardiomyocyte transfection

Cardiomyocytes were transfected using the TransIT TKO Transfection Reagent (Mirus Bio) and

50 nM of BLOCK-iT Alexa Fluor 555 Red Fluorescent Oligo (Positive Control) to determine

transfection efficiency. Transfection efficiency was assessed fluorescent microscopy, calculating

the number of MEF2A positive, Alexa Fluor 555 positive cells/well. Transfection of antagomirs

(25nM each, Dhamarcon) was carried out using the standard transfection protocol provided by

Mirus Bio., 48 hours following plating of cells, in cardiomyocyte media (DMEM/F12)

containing 1% fetal bovine serum, 1% horse serum and antibiotics. 24 hours after transfection,

media was replaced, and 48 hours following transfection, cells were fixed for

immunofluorescence or RNA was isolated.

3.2.9 Cell Cycle RT-qPCR Array

RNA was purified using the method described above from cardiomyocytes transfected with

the cocktail of all 11 miRNA antagomirs (25nM each), the optimized final 9 antagomir

cocktail (25nM each), or a negative control (cel-67 100 nM) RNA concentrations were

determined using a NanoDrop 2000 spectrophotometer (Thermo Scientific). Equal amounts of

RNA (400ng/sample) were converted into cDNA using the First Strand cDNA Synthesis

(Qiagen) kit. The PCR Array profiled the expression of 87 cell cycle genes, 5 housekeeping

genes, controls for genomic DNA contamination, and efficiency of both the (RT-PCR: PCR)

reactions

3.2.10 Immunofluorescence

Cardiomyocytes were fixed with 4% PBS-buffered formalin for 7 minutes, permeabilized with

0.5% Triton-X-100 in 1X-Tris-buffered saline. Cells were stained for 2 hours with primary

antibodies diluted in 0.5% Triton-X-100 in 1X-Tris-buffered saline: Aurora B (Sigma), Gata4

(BD), Phalloidin (Thermo Fisher Scientific). Cells were then washed and incubated for 45

minutes in their respective secondary antibodies conjugated to Alexa Fluor- and Alexa Fluor-555

(Life Technologies) and Genomic DNA was stained with 4',6-diamidino-2-phenylindole

dihydrochloride (DAPI; Life Sciences). Finally, cells were fixed again with 4% PBS-buffered

formalin for 7 minutes. Cells were imaged using the Zeiss Axio Observer inverted fluorescent

microscope. Images were acquired with the MetaMorph Microscopy Automation & Image

Page 80: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

62

Analysis Software and analyzed using ImageJ. The cells were also run through the Hidex Sense

microplate reader to detect Alexa Fluor-488 and 555 fluorescence levels (measured in Relative

Fluorescent Units; RFU).

3.2.11 Statistical Analyses

Numerical data are expressed as either the ratio change or % change of the means ± the standard

error of the mean (SEM). The Student’s t-test was utilized to determine significance between

experiments with only 2 variables. One-way analysis of variance (ANOVA) with Tukey’s

Multiple Comparison Post-Test was employed to analyze data between multiple independent

(unrelated) groups using the GraphPad Prism 5.01 software. Level of significance was first

determined by ANOVA analyses (p < 0.05), and subsequently significance between every

pairwise group in the experiment was determined by Tukey’s Multiple Comparison Test.

Page 81: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

63

3.3 Results

3.3.1 The tumor suppressor circuitry p53/MDM2 regulate expression of a unique subset of miRNAs within the heart.

To evaluate the role of p53 and MDM2 as regulators of miRNA expression within the heart, total

miRNA expression was profiled from cardiac-specific conditional p53/MDM2 DKO mouse

hearts, and was compared to single knockouts (p53KO and Mdm2KO), and vehicle injected

controls, using the Nanostring nCounter miRNA platform (n=3 hearts per group). p53 and

MDM2 knockout following tamoxifen injections was confirmed for all three strains by Western

blot (Supplemental Figure 3.1).

The nCounter miRNA array profiled over 600 mouse miRNAs. Heat map construction by

agglomerative clustering (the "bottom up" hierarchical clustering approach where each miRNA

starts in its own cluster, and pairs of clusters are merged when moving up the hierarchy), using

the nSolver analysis software, revealed that the Mdm2KO and DKO miRNA profiles were

closely related, whereas the p53KO’s miRNA profile was distinct from the other two knockouts

(Figure 3.1 A). Furthermore, the p53KO miRNA profile was more closely related to the miRNA

prolife of vehicle- injected control hearts, whereas the profiles in the Mdm2KO and DKO hearts

were more distantly related (Figure 3.1 G). Setting the miRNA expression fold change cut off as

greater than 1.2 or less than -1.2, a total of 89 significantly changed miRNA transcripts out of

600 (~15%) were identified in the three mutant hearts compared to the vehicle- injected controls,

with a P-value of ≤ 0.01 (Figure 3.1 B) (p53KO: 3 up, 12 down, Mdm2KO: 29 up, 19 down,

DKO: 36 up, 29 down).

The top 20 differentially regulated miRNAs were determined for each strain. Members of the

miR-15 family, miR-15a and miR-16, well-characterized p53-regulated miRNAs258, were in the

top 12 downregulated miRNAs of the p53KO. These miRNAs were also significantly

downregulated in the DKO, but were upregulated in the Mdm2KO, likely due to the known

higher levels of p53. This highlights the importance of MDM2 in maintaining normal levels of

Page 82: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

64

Page 83: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

65

p53-regulated genes within the cell by dampening p53’s activity. Interestingly, miR-1 and miR-

133b, classic muscle specific miRNAs which play a critical role in myogenic differentiation259,

were in the top 10 downregulated miRNAs of DKO mouse hearts. miR-1 was also in the top 10

downregulated miRNAs in Mdm2KO hearts. Furthermore, loss of p53 seemed to have little

effect on miRNA upregulation, as the expression of only a total of 3 miRNAs was significantly

increased in the p53KO. Conversely, in the Mdm2KO and DKO, a total of 29 and 36 miRNAs

respectively, were significantly upregulated compared to control hearts (Figure 3.1 D-F).

Although the change in expression of many miRNAs overlapped between the three different

strains, each knockout also had a subset of uniquely up and downregulated miRNAs (Figure 3.1

C). For this study, the subset of interest were the miRNAs uniquely downregulated in the DKO

strain, because we sought to determine if inhibition of these miRNAs in wild type

cardiomyocytes could promote proliferation. In DKO mouse hearts, 15 uniquely downregulated

miRNAs were identified with a significant fold change of less than -1.2, that were not

significantly changed either of the single knockout mice. These became the target miRNAs of

interest, and were defined as p53/MDM2-regulated miRNAs (Table 3.1).

Figure 3.1. Significant miRNA transcriptional changes in the absence of p53 and/or MDM2

create unique miRNA profiles. (A) Agglomerative clustering was preformed to generate a heat

map demonstrating miRNA fold change in the heart between the three different strains post Tamoxifen injection vs. the vehicle- injected control. 600 miRNAs were profiled using the nCounter mouse miRNA panel (Nanostring). Fold change values are depicted by color intens ity,

where blue = repressed and red = induced. Mice were analyzed at 6 months post-Tam (p53KO), 14d post Tam (Mdm2KO) and 8d post-Tam (DKO), n=3 mice per strain. (B) Heat map

representing significant (P≤0.01) miRNA fold changes between the three different strains post Tamoxifen injection vs. control with a fold change ≥ 1.2 (±) . 81 miRNA transcripts (rows) out of 600 (13.5%) were significantly changed in at least one of the three strains (columns). (C) Venn

diagram analysis illustrating the overlap of significantly up and downregulated miRNAs between the 3 KOs vs. control. The 15 miRNAs uniquely downregulated in the DKO are the target

miRNAs of interest. (D-F) Top 20 differentially regulated genes in (D) p53KO vs. control, (E)

Mdm2KO vs. control and (F) DKO vs. control. (P≤0.01) (G) Scatter plot matrix representing normalized Log2 counts for each miRNA profiled of p53KO (green), Mdm2KO (pink) and DKO

(blue) (Y axis) compared to Log2 counts of vehicle injected control (X axis). Each point represents one miRNA. R2 values indicate that the p53KO miRNA profile is more closely related

to that of the control than of the Mdm2KO or DKO profiles.

Page 84: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

66

Table 3.1. miRNAs significantly downregulated in DKO vs. vehicle control. Using the nCounter Analysis Software, miRNAs with a fold change ≥ -1.2 and p value of ≤ 0.01 that were

uniquely downregulated in the DKO vs. vehicle control were determined. Mmu-miR-1931 had no predicted target genes (determined using the miRSystem online software) and was thus eliminated from further analysis. mmu-miR-133a and mmu-miR-133b did not target genes within the cell

cycle pathway (determined using the miRSystem software) and were also eliminated from further analysis. mmu-miR-212 expression change determined by Nanostring array was not validated by

RT-qPCR (See Figure 2) and was therefore also removed from further analysis. The 11 remaining miRNAs are the final “hits” from the miRNA screen.

Page 85: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

67

3.3.2 p53/MDM2-regulated miRNAs are enriched for mRNA gene targets involved in cell cycle progression.

To characterize enriched pathways and functions regulated by the target genes of the 15

p53/MDM2-regulated miRNAs identified by the Nanostring array, the online miRNA pathway

analysis platform, miRSystem, was employed (Figure 3.2 B-C). miRSystem integrates 7

Page 86: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

68

miRNA target gene prediction algorithms (DIANA, miRanda, miRBridge, PicTar, PITA, rna22

and TargetScan) as well as 2 experimental validation databases to predict miRNA target genes.

A valid miRNA target gene is required to be identified by 4 out of the 7 prediction programs. A

P-value is then assigned to each biological pathway (KEGG and Biocarta pathways) represented

by the target genes of the input group of miRNAs. This P-value is based on how many miRNA

target genes are found within a given pathway and the overall number of genes within that

pathway. Enriched biological pathways are subsequently ranked on a score calculated by

multiplying the P-value of a given pathway by the weighted expression values of the miRNAs

that target genes within this pathway. (Supplemental Figure 3.8)260.

In this analysis, miR-1931 did not yield any predicted target genes and was eliminated from

analysis. Only 2 out of the 14 remaining miRNAs (muscle specific miR-133a and miR-133b) did

not have target genes within the cell cycle pathway and were also removed from further analysis,

whereas 12/14 miRNAs targeted at least one gene within this pathway. To validate that the 12

miRNA hits were truly downregulated in the DKO compared to control hearts as seen by the

Nanostring array, RT-qPCR was performed using the same RNA sampled for the array. RT-

qPCR revealed that 11/12 miRNAs were significantly downregulated in the DKO compared to

the vehicle control. Only 1 miRNA, miR-212, showed an opposite expression pattern than what

Figure 3.2. miRNAs downregulated in the DKO are enriched for target genes within the

cell cycle pathway. (A) Reverse transcription and quantitative PCR (RT-qPCR) was performed

to validate miRNA expression values measured by the Nanostring nCounter mouse miRNA

panel. All 12 miRNA hits of interest were measured. Data are represented as the miRNA

expression ratio change in DKO mouse hearts compared to controls for each primer using the

ΔΔCt method ± s.e.m, n=3. A significant difference between the mean normalized delta Ct

values for the DKO vs. control is indicated by * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001 determined

by the Student’s t-test. The 11 validated miRNAs correspond to the final miRNA hits extracted

from the Nanostring microarray, whereas miR-212 was removed from further analysis. (B-C)

miRSystem Software was utilized to determine the predicted target genes of the 11 miRNA hits

within the KEGG cell cycle pathway (125 total genes) (B) Percent of each miRNA’s total target

genes found within the cell cycle pathway (*P≤0.05, **P≤ 0.01). (C) The number of cell cycle

activating and inhibiting genes targeted by the 11 miRNA hits.

Page 87: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

69

was determined by the array, and was upregulated compared to vehicle control (Figure 3.2 A).

As the expression level of this miRNA could not be confirmed, it was eliminated from further

study. Cell cycle target genes of the remaining 11 miRNAs were subsequently investigated in

further depth. The most enriched biological pathways that the target genes of these miRNAs

Figure 3.3 Genes within the cell cycle pathway are redundantly targeted by the miRNAs

downregulated in the DKO. (A) Schematic of the 26 cell cycle activating genes targeted by the

12 miRNA hits as determined by the miRSystem software. 81% of cell cycle genes targeted play a role in cell cycle activation. (B) Schematic of the 6 cell cycle inhibiting genes targeted by the

12 miRNA hits as determined by the miRSystem software. 19% of cell cycle genes targeted play a role in cell cycle inhibition.

Page 88: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

70

belong to are illustrated in Supplemental Figure 3.2. The cell cycle pathway was within the top

30th percentile of biological pathways for this group of miRNAs. Target genes of these miRNAs

also belonged to many other biological pathways that regulate the cell cycle, including mitogenic

signaling pathways, the wingless-type MMTV integration site family (WNT) signaling pathway,

and pathways involved in regulating the actin cytoskeleton. These 11 miRNAs were enriched

for target genes that function directly within the cell cycle. For example, approximately 10% of

miR-30e’s target genes were found within this pathway (Figure 3.2 B). Furthermore, there was a

high amount of redundancy between the 11 miRNAs and their targets within the cell cycle,

where some genes were targeted by up to 6 of the identified miRNAs (Figure 3.2 C). This

redundancy was apparent for target genes involved in cell cycle activation, whereas genes

involved cell cycle inhibition were only targeted by a maximum of 4 miRNAs. Furthermore, 26

out of the 32 miRNA target genes found within the cell cycle pathway were involved in cell

cycle activation (81%), whereas only 6 out of 32 were involved in cell cycle inhibition (Figure

3.2 C, Figure 3.3 A, B), suggesting that upon downregulation of these 11 miRNAs, the

inhibition over a battery of key cell cycle activating genes may be alleviated, promoting their re-

expression within the cell.

Therefore, the 11 validated miRNAs, uniquely downregulated in DKO mouse hearts vs. controls,

who target at least one gene within the cell cycle pathway were defined as the final “hits” from

this miRNA screen, and are listed in Table 3.2. These miRNAs were hypothesized to function as

cell cardiac cell cycle inhibitors.

Table 3.2. Final list of p53/MDM2-regulated miRNAs. List of the 11 miRNAs downregulated uniquely in DKO mouse hearts vs. controls with a fold change of ≥ -1.2 and a P value of ≤ 0.01

determined by the Nanostring miRNA array, that were validated by RT-qPCR, and target at least one gene within the cell cycle. These are the final 11 miRNAs selected to be inhibited by antagomirs, and

are defined as p53/Mdm2-regulated miRNAs.

Page 89: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

71

3.3.3 p53/MDM2-regulated miRNAs are novel cardiomyocyte cell cycle inhibitors

Primary rat neonatal cardiomyocytes were utilized as an in vitro model to explore the role of the

11 p53/MDM2-regulated miRNAs in modulating cardiac cell cycle arrest. To support the

hypothesized “anti-proliferative” function of these miRNAs, their expression levels were

examined in both wild type cardiomyocytes, which do not proliferate, and cardiac fibroblasts,

which are known to proliferate extensively in response to cardiac injury261. It was thus

hypothesized that the expression of these 11 cell “cycle-inhibitory” miRNAs would be

downregulated in cardiac fibroblasts compared to non-proliferative cardiomyocytes. RT-qPCR

revealed that the expression of all 11 miRNAs was significantly decreased in fibroblasts

compared to cardiomyocytes by up to 80% (Supplemental Figure 3.3). This strengthens the

notion that inhibition of these miRNAs in wild type cardiomyocytes could promote cell cycle re-

entry and proliferation, as they are found to be downregulated in cardiomyocytes that have re-

entered the cell cycle (in DKO mouse hearts), as well as in wild type fibroblasts which

proliferate naturally.

Antagomirs (chemically modified, cholesterol-conjugated, single-stranded RNA analogues

complementary to miRNAs that antagonize the function of miRNAs125,126), targeting the 11

p53/MDM2-regulated miRNAs were subsequently utilized to investigate if the identified

miRNAs regulate cell cycle in wild type cardiomyocytes..

To study the specificity and inhibitory effect of antagomirs in cardiomyocytes, we transfected

cells with 2 antagomirs (anti-miR-30b and anti-let-7f) separately (cardiomyocyte transfection

efficiency of 93% ± 6%, see Supplemental Figure 3.4). This was to determine if antagomirs are

cross-reactive between and within miRNA families, as 4 of the miRNA hits are part of the miR-

30 family, and 2 belong to the let-7 family. Antagomir inhibition of their target miRNA was

highly potent: anti-miR-30b and anti-let-7f downregulated the expression of miR-30b by 97%

(P≤0.0001, n=4) and let-7f by 99% (P≤0.0001, n=4) respectively, as measured by RT-qPCR.

Conversely, transfection of anti-miR-30b had no significant effect on the expression of let-7f.

Similarly, anti-let-7f did not significantly alter the expression of miR-30b in rat neonatal

cardiomyocytes (Figure 3.4 C). This suggests that these two antagomirs do not inhibit off target,

unrelated miRNAs. However, assessing the expression levels of other miR-30 and let-7 family

Page 90: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

72

members in cardiomyocytes transfected with anti-mir-30b and anti-let-7f, respectively, revealed

that that antagomirs downregulate off target miRNAs from within the same miRNA family as

their target miRNA to varying degrees of potency. For example, cardiomyocytes transfected with

anti-miR-30b also exhibited a significant downregulation of miR-30c (P≤0.0001, n=4), but not

of miR-30e, and anti-let-7f induced a potent downregulation in the expression of let-7f’s sister

miRNA, let-7a (P≤0.0001, n=4) (Figure 3.4 A, B).

Figure 3.4. Antagomirs are specific between miRNA families but redundant within

miRNA families (A) RT-qPCR for cardiomyocytes transfected with 50 nM of anti-miR30b or anti-cel-67 (negative control that targets Caenorhabditis elegans specific miRNA). Anti-miR-

30b decreases expression of its target miRNAs (miR-30b) significantly, and the expression of miRNAs from within the miR-30 family to varying degrees of potency. Data are represented as

miRNA expression ratio change of the antagomir treated group compared to the anti-cel-67 negative control group for each primer ± s.e.m, **P≤0.01, ***P≤0.001, n=4, and analyzed by Student’s t-test. (B) RT-qPCR for cardiomyocytes transfected with 50nM of anti-let-7f or anti-

cel-67. Anti-let-7f decreases expression of its target, let-7f, as well as let-7a (off target within the same family) significantly (ratio change values are ± s.e.m, **P≤0.01, ***P≤0.001, n=4,

analyzed by Student’s t-test). (C) RT-qPCR for cardiomyocytes transfected with either 50 nM of anti-let-7f (checkered bars), anti-miR-30b (solid grey bars) or anti-cel-67 Antagomirs significantly decrease expression of their target miRNA, but do not significantly decrease

expression of off target miRNAs of different miRNA families. Data was analyzed by one-way ANOVA (P≤0.0001) and Tukey’s Multiple Comparison post hoc test to identify differences

between groups and are represented as miRNA expression ratio change of the antagomir treated

group compared to the cel-67 negative control group for each primer± s.e.m, ***P≤0.001, ****P≤0.0001, n=4.

Page 91: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

73

To determine if antagomirs can effectively inhibit their target miRNA’s expression levels when

delivered as a cocktail, cardiomyocytes were next transfected with a combination of all 11 anti-

miRs. Inhibition of the 11 target miRNAs remained highly effective (Figure 3.5 A). Different

antagomirs exhibit varying degrees of inhibitory potency. Antagomirs targeting the miR-30

family were the most potent, and this could possibly be due to combined cross reactivity between

the different antagomirs within this family, as noted in Figure 3.4 A. These results confirmed

that transfecting cardiomyocytes with a cocktail of antagomirs was a feasible approach to knock

down multiple miRNAs at one time.

Knowing that the selected antagomirs effectively inhibit their target miRNAs, we proceeded to

determine whether knockdown of all 11 p53/MDM2-regulated miRNAs in cardiomyocytes could

promote cell cycle re-entry and proliferation. Cardiomyocyte proliferation was assessed by

immunofluorescence, staining for Aurora B (cytokinesis marker), co-stained with α-sarcomeric-

actin (cardiomyocyte-specific sarcomere marker), and DAPI (nuclear DNA marker) (Figure 3.6

A-H). Indeed, transfecting cardiomyocytes with a cocktail of antagomirs targeting all 11

p53/Mdm2-regulated miRNAs led to a striking increase in the occurrence of cytokinetic events

(Aurora B positive mid-body structures) by over 90%, compared to the negative control

transfected wells (~50,000 cells per well) (P≤0.01, n=3) (Figure 3.5 B, Supplemental Figure

3.5 A, B). This result underscores the role of these 11 miRNAs as cell cycle inhibitors in

cardiomyocytes.

Cardiomyocytes were also transfected with varying combinations of antagomirs to determine

which of the 11 were necessary to induce proliferation. To determine the minimal number of

antagomirs required to produce this proliferative phenotype, a “process of elimination” similar to

the concept pioneered by Takahashi & Yamanaka (2006) for determining the factors necessary in

pluripotent stem cell induction was utilized. Each antagomir was removed individually from the

cocktail and proliferation was assessed by immunofluorescence (Figure 3.5 B, Supplemental

Figure 3.5 A, B). If removing a specific antagomir reduced proliferation significantly compared

to the cocktail containing all 11 antagomirs (P≤ 0.05), it was deemed an essential member of the

antagomir cocktail. In contrast, if removing a specific antagomir had no effect on proliferation

Page 92: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

74

(i.e. a statistically significant number of cytokinetic events was maintained compared to negative

control transfected wells), this antagomir was noted as non-essential for the induction of

cardiomyocyte proliferation, and removed from the cocktail. Following this screen, it was

determined that antagomirs targeting miR-30c and let-7a were non-essential to promote

cardiomyocyte proliferation, whereas anti-miR-30b, anti-let-7f, anti-miR-181a, anti-miR-149

and anti-miR-194 were necessary components of the cocktail. To increase the throughput of the

screening process, we tested if the use of a microplate reader (picking up overall red 555

fluorescent signal in each well, corresponding to the Aurora B antibody) would parallel the

microscopy findings. As indicated by relative fluorescent units (RFU), the microplate reader also

Page 93: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

75

determined that cells transfected with all 11 antagomirs had increased fluorescence compared to

the negative control transfected wells, however the machine was not sensitive enough to detect

significant changes (Supplemental Figure 3.6 A, B). One-way ANOVA and Tukey’s Multiple

Comparison Post-Test were the statistical manipulations utilized to determine significance for

these experiments.

To confirm that anti-miR-30c and anti-let-7a were non-essential to promote cardiomyocyte

Figure 3.5. Inhibition of 11 target miRNAs by antagomir cocktail upregulates

cardiomyocyte cytokinesis. (A) Transfection with 11 antagomir cocktail efficiently knocks

down all target miRNAs. RT-qPCR performed for cardiomyocytes transfected with either a cocktail of all 11 antagomirs (25nM each) or anti cel-67 (negative control, 100 nM). Data are represented as represented as miRNA expression ratio change of the antagomir treated group

compared to the anti-cel-67 negative control group for each primer ± s.e.m, ** p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001, n=3 and analyzed by Student’s t-test. (B) Cardiomyocytes were

transfected with either anti-cel-67, a cocktail of all 11 antagomirs (25nM each, red bar), a cocktail containing 10/11 antagomirs (label under bar indicates antagomir excluded from cocktail) or not transfected (black bar). Cardiomyocytes were subsequently stained with

antibodies against Aurora B, α-actinin and DAPI. The total number of cardiomyocyte cytokinetic events (indicated by Aurora B positive mid-body structures between two cells with

α-actinin positive sarcomeric structures) were counted in each well (~50,000 cells/well). Blue bars indicate the cocktails that retained a statistically significant number of cytokinetic events compared to the negative control, despite removal of one antagomir. These antagomirs are

hypothesized to be unnecessary for induction of cardiomyocyte proliferation. Light grey bars indicate cocktails where removal of the given antagomir significantly reduced cardiomyocyte

proliferation compared to the cocktail containing all 11 antagomirs. These antagomirs are hypothesized to be essential within the cocktail for induction of cardiomyocyte proliferat ion. Data represented as % change of Aurora B positive mid-body structures in treated cells

compared to negative control. Data analysis by one-way ANOVA (p=0.0035) and Tukey’s Multiple Comparison Test to identify differences between groups. ** indicates p ≤ 0.01, *** p

≤0.001 for antagomir treatment compared to negative control treatment. # indicates P≤0.05, ##P≤0.01, ### P≤0.001 for antagomir treatment compared to all 11 antagomir cocktail treatment, ± s.e.m, n=3. (C) Cardiomyocytes were treated according to the + and – symbols

under the x axis. Transfection with only anti-miR-30c+anti- let-7a significantly reduced cardiomyocytes proliferation compared to the cocktail containing all 11 antagomirs.

Cardiomyocytes transfected with all antagomirs except anti-miR-30c+anti- let-7a retained significant upregulation in proliferation compared to negative control. Data analysis by one-way ANOVA (P≤0.0001) and Tukey’s Multiple Comparison Test. *** indicates p ≤0.001 for

antagomir treatment compared to negative control treatment, ### indicates P≤0.001 for antagomir treatment compared to all 11 antagomir cocktail treatment, ± s.e.m, n=3. (D) List of

9 antagomirs in the finalized antagomir cocktail. anti-miR-30c and anti-let-7a were elimina ted.

Page 94: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

76

proliferation, a follow-up experiment was conducted where both antagomirs were removed from

the cocktail simultaneously. In this experiment, cardiomyocytes transfected with this cocktail of

only 9 antagomirs (containing all antagomirs except for anti-miR-30c and anti-let-7a) and

cardiomyocytes transfected with the cocktail containing all 11 antagomirs experienced over a

100% increase in cytokinetic events, compared to negative control treated cells. Accordingly,

transfection with a cocktail containing only anti-let-7a and anti-miR-30c significantly decreased

cardiomyocyte proliferation compared to cells transfected with all 11 antagomirs, and led to an

8% decrease in proliferation compared to the negative control (not significant). Similarly,

treatment of cardiomyocytes with either of those antagomirs alone caused a significant decrease

in proliferation compared to cardiomyocytes transfected with all 11 antagomirs. In fact,

transfection of cardiomyocytes with anti-miR-30c alone caused a significant ~23% decrease in

cytokinetic events compared to negative control treated cells, indicating that downregulating

miR-30c may be hindering cardiomyocyte proliferation in the cocktail containing all 11

antagomirs (Figure 3.5 C, Supplemental Figure 3.4 C, D). Thus, anti-miR-30c and anti-let-7a

were considered unnecessary members of the optimal antagomir cocktail required to induce

cardiomyocyte proliferation.

Next, we tested whether transfecting cardiomyocytes with only the five antagomirs deemed

“necessary” (anti-miR-30b, anti-let-7f, anti-miR-181a, anti-miR-149 and anti-miR-194) was

sufficient to promote cell cycle re-entry. Treatment of cardiomyocytes with this cocktail induced

a significant upregulation in proliferation of ~60%, however this was also significantly less

potent than either the cocktail containing 11 or 9 antagomirs. This indicated that antagomirs

other than those deemed necessary (dark grey bars, Figure 3.5 A: anti-miR-30a, anti-miR-30e,

anti-miR-204 or anti-miR-26b) were also adding to the ability of cardiomyocytes to proliferate.

Page 95: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

77

Figure 3.6. Treatment of cardiomyocytes with cocktail of 11 antagomirs promotes cardiomyocyte cytokinesis.

(A-H) Two 40x confocal immunofluorescence images of cardiomyocytes transfected with a cocktail of all 11

antagomirs (25nM each) undergoing cytokinesis. Cells were fixed and stained for (A,E) α-sarcomeric actin (red, cardiomyocyte specific sarcomere marker), (B,F) DAPI (blue), and (C,G) Aurora B (green, cytokinesis marker). (D,H) Merged image of all 3 channels. White arrows indicate Aurora B positive mid-body structures between two

cells undergoing cytokinesis

Page 96: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

78

We therefore sequentially removed each of these antagomirs from the cocktail of 9 antagomirs

(lacking the two unnecessary anti-miRs; anti-let-7a and anti-miR-30c) to determine if any were

revealed to be essential within this cocktail. Interestingly, removal of any of the 4 antagomirs

from the cocktail of 9 led to a significant decrease in cardiomyocyte proliferation compared to

cells treated with all 11 or 9 inhibitors. From this we concluded that treatment of cardiomyocytes

with 9/11 antagomirs (without anti-let-7a and anti-miR-30c) was the lowest number of

antagomirs required in the cocktail to promote proliferation (Figure 3.5 C, Supplemental

Figure 3.4 D). Thus, inhibition of p53/MDM2 regulated miR-30a, miR-30b, miR-30e, let-7f,

miR-204, miR-26b, miR-149, and miR-194 simultaneously was necessary for cardiomyocytes to

regain proliferative ability. A cocktail of 9 antagomirs targeting these specific miRNAs was

identified as the optimized final cocktail sufficient to promote cell cycle reactivation in

cardiomyocytes (Figure 3.5 D).

3.3.4 p53/MDM2-regulated miRNAs inhibit cardiomyocyte proliferation by downregulating target genes involved in the progression of all 4 phases of the cell cycle.

To elucidate how treatment of cardiomyocytes with either the cocktail of all 11 antagomirs, or

the final optimized cocktail of 9 antagomirs mechanistically promotes cell cycle reactivation, a

cell cycle pathway specific RT-qPCR array (Qiagen) which profiles the expression of 86 cell

cycle related genes was utilized. We were interested to determine if inhibition of these miRNAs

could promote re-expression of their targets genes within the cell cycle (listed in Figure 3.2 C

and 3.3 A, B). This would assist to confirm their role as cell cycle inhibitory and/or tumor

suppressive miRNAs.

Of the 32 genes within the cell cycle pathway targeted by the 11 p53/MDM2-regulated miRNAs,

20 were contained in the RT-qPCR cell cycle array. This array would therefore allow us to

confirm certain predicted miRNA target genes, but also help us to identify novel genes regulated

by these miRNAs that have not been previously characterized.

Four samples from each treatment group (11 antagomir cocktail, final 9 antagomir cocktail and

negative anti-cel-67 control) were profiled by the cell cycle array. This array revealed that

Page 97: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

79

inhibition of p53/MDM2-regulated miRNAs in both treatment cocktails causes a wide spread

upregulation in factors involved in cell cycle re-entry and progression. Specifically, inhibition of

all 11 miRNAs caused a significant (P≥0.05) upregulation of 32 genes involved in cell cycle

regulation (~36% of genes on the array). Genes necessary at each of the four phases of the cell

cycle became significantly upregulated, including cyclin D2 which promotes G1 phase

progression, minichromosome maintenance proteins 2/4 (MCM2/4) and origin of replication

complex subunit 2 (ORC2) which are necessary for DNA replication in S phase, cell division

cycle 25 (CDC25) which controls the rate limiting step of CDK1 activation in late G2 phase, and

importantly, CDK1, the absolutely essential kinase that induces M phase initiation and

progression37. Only one gene from this array, cyclin D1, was found to be significantly

downregulated by both treatments. This highlights the canonical role of miRNAs as inhibitors of

gene expression, such that their inhibition by antagomirs allows for a widespread upregulation of

their target genes. Notably, in cardiomyocytes transfected with the cocktail of all 11 antagomirs,

40% (8 out of 20) of the predicted miRNAs’ cell cycle target genes profiled by the array were

upregulated (Figure 3.7A).

Cells treated with the final antagomir cocktail also exhibited a significant upregulation in 25 out

of the 86 cell cycle genes profiled by this array, 7 of which were predicted target genes of the

miRNAs inhibited by the treatment (Figure 3.7 B). This treatment also induced the upregulation

of genes required for cell cycle progression at G1, S, G2 and M phase that are turned off in non-

treated cardiomyocytes, likely accounting for their quiescence. Importantly, treatment with both

cocktails produced a significant overlap in the change in gene expression elicited. 15 genes out

of the total number of genes significantly changed by both treatments were the same, indicating

that these two cocktails likely upregulate cell cycle re-entry and proliferation through a similar

mechanism. The extensive effects that inhibition of p53/MDM2-regulated miRNAs have on the

expression of a plethora of crucial cell cycle genes supports their role as strategic mediators of

cell cycle arrest in cardiomyocytes.

Page 98: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

80

Figure 3.7. Inhibition of 11 target miRNAs causes upregulation of many miRNA target genes within the cell

cycle pathway. Significantly (≤0.05) changed cell cycle related genes (ratio change of >1.2 or < 0.5) following treatment of cardiomyocytes with a cocktail of all 11 antagomirs or the final cocktail of 9 antagomirs (without unnecessary anti-miR-30c and anti-let-7a). The Qiagen Cell Cycle RT2 Profiler PCR Array was used to profile 86 cell

cycle relates genes following antagomir treatment compared to negative control treatment (anti-cel-67) by RT-qPCR. (A) Significantly changed cell cycle related genes following treatment of cardiomyocytes with all 11 antagomirs vs.

control treatment. (B) Significantly changed cell cycle related genes following treatment of cardiomyocytes with the final cocktail of 9 antagomirs vs. control treatment. White bars in (a) and (b) reflect genes that were significantly changed by both treatments. Values are represented as the ratio change of treatment vs. control ± s.e.m, n=4 and

analyzed by the Student’s t-test.

Page 99: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

81

3.4 Discussion

Our data show that a subset of 9 to 11 miRNAs regulated by p53/MDM2 function within

cardiomyocytes to maintain cell cycle arrest. The implications of these findings are two-fold.

Firstly, they reveal a novel role for p53 and MDM2 within the heart, of maintaining

cardiomyocyte cell cycle control through the regulation of miRNAs. Secondly they uncover

group of anti-proliferative miRNAs actively regulating cardiomyocyte quiescence.

3.4.1 p53 and MDM2 regulate the expression of muscle specific miRNAs in the heart

Following the conditional, cardiac-specific knockout of the tumor suppressor circuitry p53 and

MDM2, we were able to identify distinct populations of miRNAs that are regulated by each gene

individually, as well as in tandem. Loss of p53 led to the downregulation of 12 miRNAs whereas

19 and 29 miRNAs were downregulated in the Mdm2KO and DKO, respectively. Furthermore,

knockout of p53 produced an upregulation of only 3 miRNAs, whereas the Mdm2KO had 29

upregulated genes, and DKO had 36, many of which overlapped between the two strains,

indicating that the over-activation of p53 in the Mdm2KO is not the sole cause of the gene

expression changes observed. This highlights a potentially new role for MDM2, in regulating the

stability of other miRNA transcriptional activators yet to be identified. Among the

downregulated miRNAs in DKO hearts were the muscle specific miR-1 (also downregulated in

Mdm2KO) and miR-133 families. These miRNAs are expressed in adult skeletal and cardiac

muscle, but not in other tissues, and have been shown to play key roles in repressing the fetal

gene program in the adult heart120,262. Knockout of these two miRNAs in the adult heart was

shown to induce a re-expression of many fetal genes including Glycogenin 1, Glucan Branching

Enzyme 1, smooth muscle actin-α2, myosin heavy chain-β and myosin light chain-β120,262.

Interestingly, our lab had demonstrated that the double knockout of p53 and MDM2 specifically

in the heart also leads to the re-expression of the fetal gene program, and entails a metabolic shift

from fatty acid synthesis to glycolysis214 and our current study brings to light a possible

mechanism through which this may be occurring. Thus, the elucidation of p53 and MDM2

regulated miRNAs has helped to construct a specific role for these two genes within the heart of

maintaining cardiac differentiation and metabolism, apart from their canonical roles in tumor

Page 100: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

82

suppression. Future studies revealing the distinct interplay between p53, MDM2 and these

muscle specific miRNAs may reveal novel insights into the dedifferentiation and metabolic shift

exhibited by the heart during the progression of heart failure.

3.4.2 p53 and MDM2 maintain cardiomyocyte quiescence through the regulation of a unique group “anti-proliferative” miRNAs.

It has been widely accepted that a large number of miRNAs function as tumor suppressors within

the cell128. The cell cycle is a molecular cascade that entails numerous feedback loops, involving

an expansive number of regulators with overlapping functions. The robustness of this network

highlights the sheer evolutionary importance of this pathway, as the cell does not rely solely on a

small number of proteins for this process to occur, but instead utilizes the tight interplay of a

large number of proteins with degrees of functional redundancy, such that the loss of a single

factor does not result in a mitotic catastrophe. For example, knockout of CDK2, CDK4 or CDK6

alone does not produce a significant effect on cell proliferation263–265. CDK1 has been shown to

compensate for the loss of CDK2 and is capable to bind cyclin E in S phase266, and CDK2 was

shown to assume the functions of CDK4/6 upon their loss. It therefore follows that miRNAs play

an important role in maintaining quiescence of differentiated cells, as their capacity to fine tune

or dampen the expression of an expansive number of genes gives them the ability to

simultaneously downregulate the expression of a multitude of cell cycle regulators, overcoming

the internal redundancy of this pathway. Unpublished research from our lab suggests that by 7

days after birth, when cardiomyocytes are known to exit the cell cycle, there is a blanket

downregulation in almost all factors regulating the cell cycle pathway, including activators and

inhibitors, making the cells refractory to growth stimuli. This comprehensive shutdown of the

cell cycle pathway coincides with a switch in miRNA expression profiles between day 1 and day

10 post-natal mouse hearts, as demonstrated by Porrello et al (2011)102. It is possible that during

this switch in miRNA profiles, there is a bias towards the upregulating of anti-proliferative

miRNAs which contributes to the widespread downregulation of cell cycle regulators noted.

Based on previous research in our laboratory demonstrating a reactivation of the cardiomyocyte

cell cycle upon specific deletion of both p53 and MDM2 in the heart, and p53’s known ability to

regulate the expression of tumor suppressive miRNAs, we sought to determine if in the heart,

p53 and MDM2 may be promoting cell cycle arrest through the activation of “anti-proliferative

Page 101: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

83

miRNAs”. We found that the loss of both p53 and MDM2 led to the downregulation of a unique

subset of 15 miRNAs which were not significantly changed in the hearts of either single KOs.

We therefore hypothesized that these miRNAs may be crucial inhibitors of cell cycle in wild

type mouse hearts, and their downregulation following the loss of p53/MDM2 facilitates the cell

cycle re-entry we observe in these mutant mice (not observed in either of the single KOs).

Indeed, through bioinformatic target gene analysis, we found that 11 of these miRNAs (miR-30a,

miR-30b, miR-30c, let-7a, let-7f, miR-181a, miR-26b, miR-204, miR-149 and miR-194) possess

target genes within the cell cycle, and that a large majority of these genes (81%), are involved in

cell cycle activation. The implication of this finding is that downregulation of these miRNAs,

such as what is seen in DKO hearts, could remove the inhibition over a large group of key cell

cycle factors necessary for reactivation of this pathway and proliferation of cardiomyocytes. We

therefore inhibited these 11 miRNAs all together in wild type cardiomyocytes using a cocktail of

11 antagomirs, which elicited over a 100% increase in cytokinesis (determined by the presence

of Aurora B positive mid-body structures as opposed to indirect measurements of proliferation

such as Ki-67 or PiH3). This indicates that these miRNAs have a broad effect over cell cycle

control, and that their inhibition not only induces re-entry, but the completion of this pathway to

its entirety, resulting in the formation of new cardiomyocytes. This is important because it is

known that cardiomyocytes have cell cycle roadblocks at multiple phases in the pathway, and

although the upregulation of a single factor can promote cell cycle re-entry, it is extremely rare

that they are shown to complete the process and physically divide into two separate cells81. This

data also fits in well with the research presented by Porrello et al., as they demonstrated that the

miR-30 and let-7 families were prominently upregulated in P10 hearts coinciding with cell cycle

arrest, and antagomirs targeting 5 miRNAs from these families make up a large portion of our

antagomir cocktail102.

The approach we have taken to promote cardiomyocyte proliferation demonstrates that targeting

miRNAs, who have an overarching, all-inclusive control over biological pathways with the

capability to inhibit the expression of hundreds of genes, may be an efficient method to promote

a desired phenotype. Furthermore, the redundancy in target genes between different miRNAs

(e.g. the stromal antigen 1 (STAG1) is a target of 6/11 miRNAs) supports the cocktail method

that we utilized. Targeting simply one miRNA may not be sufficient to upregulate all the cell

Page 102: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

84

cycle factors required to promote cardiomyocyte proliferation, because other anti-proliferative

miRNAs remain, who have overlapping target genes, and can maintain their inhibition despite

the loss of one miRNA. miRNAs likely do not function in isolation124. Rather, the expression of

multiple miRNAs that target genes with a certain degree of overlap promotes a synergistic

function, enhancing their potency within the cell to shut off biological pathways, while

enhancing others124. With this notion in mind, we were interested in defining the potential for a

smaller cocktail of antagomirs. Through sequential removal of each antagomir from the cocktail

individually, we concluded that the presence of 9 antagomirs was sufficient to upregulate

cardiomyocyte proliferation to a level comparable with the cocktail of all 11 antagomirs. It is

interesting that the two non-essential antagomirs (anti-miR-30c and anti-let-7a) inhibit miRNAs

who have other family members also being targeted in the cocktail, whereas inhibition of each

unique miRNA was necessary. Two plausible explanations can be deduced: (1) within the

cocktail there are antagomirs targeting three other miR-30 family members and one other let-7

family member, and inhibition of these other family members causes a sufficiently potent

upregulation of their target genes, such that miR-30c and let-7a need not be inhibited (miR-30c

target genes 100% overlap with those of miR-30a, let-7a target genes 100% overlap with those of

let-7f), (2) antagomirs targeting miR-30a, miR-30b or miR-30e non-specifically inhibit miR-30c,

and the antagomir targeting let-7f non-specifically inhibits let-7a such that anti-miR-30c and

anti-let-7a are not necessary.

3.4.3 p53/MDM2-regulated miRNAs maintain cardiomyocyte cell cycle arrest through the inhibition of multiple cell cycle regulators.

To determine a mechanism by which our antagomir cocktails upregulated cardiomyocyte

proliferation, we investigated the expression of 86 key cell cycle regulators following treatments,

which included 20 out of the 32 of the predicted cell cycle target genes of the 11 miRNAs. Both

treatments (all 11 antagomirs or optimized final cocktail with 9 antagomirs) significantly

upregulated the expression of a multitude of cell cycle regulators required at each phase of the

cycle, including a significant percentage of miRNA cell cycle target genes predicted by the target

gene prediction software that we utilized. Treatment of cardiomyocytes with all 11 antagomirs

upregulated the expression of 32 cell cycle regulators, and the optimized 9 antagomir cocktail

promoted re-expression of 24 genes, whereas only cyclin D1 was significantly downregulated by

Page 103: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

85

both treatments. This demonstrates that the knockdown of these miRNAs alleviates their

inhibition over a large number of genes essential for cell cycle entry (early G1 phase),

progression (G1/S transition, S phase, G2/M transition, M phase) and completion (cytokinesis).

The majority of the genes upregulated were cell cycle activators, but key cell cycle inhibitors

such as retinoblastoma (RB), retinoblastoma-like protein 1 (RBL1/p107) and retinoblastoma-like

protein 2 (RBL2/p130) were also induced. This finding indicates that the treatments promote

cardiomyocytes to begin cycling through the cell cycle, whereby a large majority of the cells are

likely in G1 phase (the longest phase of the cell cycle) in which the expression of these cell cycle

inhibitors are important for the timely coordination of E2F mediated gene expression267. In line

with this, the inhibitory E2F5 gene is also upregulated by both treatments, and therefore can be

bound by p130 allowing it to traffic to the nucleus and inhibit important S phase genes while the

cell is preparing itself for cell cycle commitment in early G1 phase267. Factors that promote G1/S

phase transition such as CDK6 and the S-phase kinase-associated protein 2 E3 ubiquitin protein

ligase (SKP2) were also upregulated. CDK6 phosphorylates RB, which inhibits its function and

allows the cell to progress into S phase. The E3 ubiquitin ligase SKP2 plays a similar role, and

degrades p27Kip1, a protein which promotes G1 phase arrest by inhibiting the catalytic activity of

Cyclin-CDK complexes268. Crucial S phase executors such as MCM2 and MCM4 are induced.

These proteins are components of the DNA helicase and form the foundation for the construction

of active replisomes, allowing DNA replication to proceed44. The essential G2/M transition

mediator, cell division cycle 25B (CDC25B) is also upregulated. CDC25B removes the

inhibitory phosphate groups from CDK1, which is the rate limiting step for the initiation of

mitosis269. Importantly, this step would be redundant if CDK1 was not also expressed (it is

downregulated in adult hearts), as M phase cannot progress without this key kinase57. Indeed,

both treatments significantly upregulated the expression of CDK1 in cardiomyocytes, indicating

that not only can the cells enter into the cell cycle, but can also proceed through mitosis. Other

regulators of M phase such as the MAD2 mitotic arrest deficient-like 1 protein (MAD2L1), were

increased as well. MAD2L1 is a component of the mitotic checkpoint complex, which monitors

chromosome alignment and is necessary to halt transition into anaphase if chromosomes become

misaligned37. Moreover, as we observed an increased number of cells undergoing cytokinesis,

we presumed key regulators of this process would be upregulated. Indeed stathmin (STMN1), an

important mediator of microtubule dynamics, whose activity is necessitated during cytokinesis to

Page 104: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

86

modulate the drastic cytoskeletal changes that occur during this process, was significantly

upregulated following both antagomir treatments270. In all, cell cycle progression requires an

intricate orchestration of cell cycle activators, inhibitors and checkpoint regulators, many of

which we have shown to be inhibited in cardiomyocytes by a group of “anti-proliferative”

miRNAs regulated by p53/MDM2. The inhibition of these identified miRNAs permits the

upregulation of a widespread group of cell cycle regulators that as a whole, allow the re-entry

and controlled cycling of cardiomyocytes through each phase of the cell cycle.

The ability of miRNAs to inhibit many genes offers a potential therapeutic advantage over other

agents that act only on a single gene, especially when striving to regulate a process that requires

the reactivation of multiple genes. Furthermore, targeting a group of miRNAs who function in a

similar pathway may be important in effectively modulating complex physiological processes.

We have identified a group of miRNAs who function in the heart to promote cardiomyocyte

quiescence, under the regulation of the tumor suppressor circuitry p53 and MDM2. These data

provide a new level of understanding to how cell cycle arrest is regulated in the heart, and how

p53 and MDM2 participate in the mediation of this process. In addition, this approach has the

possibility to be exploited for the development of novel regenerative therapies to replenish lost

heart cells following injurious events like myocardial infarction.

Page 105: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

87

3.5 Supplementary Figures and Tables

Supplemental Figure 3.1. Validation of cardiac specific MDM2 and p53 knock-down

following Tamoxifen injection. To specifically knock-down p53 and Mdm2 in the adult mouse heart, a tamoxifen (Tam) inducible Cre-loxP system where MerCreMer recombinase

(mcm) expression is regulated by the cardiomyocyte specific myosin heavy chain 6 promoter was used to generate the following knockout (KO) strains: p53KO p53f/f;mcm; Mdm2KO,

Mdm2f/f;mcm; DKO, Mdm2f/f;p53f/f;mcm. Tam was injected intraperitoneally once daily for 4 consecutive days into 8 week old mice. Day 0 = day of last Tam injection. Total protein isolated from the left ventricle of the 3 KO strains was probed with primary antibodies against

p53 and Mdm2 as indicated (left). An antibody against nucleophosmin (Npm1) was reprobed for on all membranes to verify equal protein loading into all lanes.

Page 106: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

88

Supplemental Figure 3.2. Biological pathway analysis for the 11 p53/MDM2-regulated miRNAs. miRNA

target genes were grouped into GSEA and KEGG biological pathways. Represented are the top 100 enriched pathways that the 11 miRNAs regulate, ranked based on their target genes and expression levels (fold change) (miRSystem Pathway ranking Score – see Supplemental Figure 6 for score equation). The KEGG “Cell Cycle”

pathway is in the top 30 biological pathways that the p53/MDM2-regulated miRNAs regulate

Page 107: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

89

Supplemental Figure 3.3. 11 miRNAs hits are downregulated in

cardiac fibroblasts. RT-qPCR of cardiomyocytes and fibroblas ts probing for the 11 miRNA hits. Data are represented as the miRNA

expression ratio change in cardiac fibroblasts compared to wild type cardiomyocytes for each primer ± s.e.m, n=3. A significant difference

between the mean normalized delta Ct values for fibroblasts vs. cardiomyocytes is indicated by * p ≤ 0.05, ** p ≤ 0.01, *** p ≤ 0.001, **** p ≤ 0.0001, n=3. Data analyzed by Student’s t-test.

Page 108: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

90

Supplemental Figure 3.4. Efficient transfection of neonatal rat cardiomyocytes with red fluorescent positive

control. (A-D) 10x immunofluorescence images of cardiomyocytes transfected with a positive control (Alexa Fluor®555 Red Fluorescent Control siRNA, 100 nM). Transfection efficiency = 93% ± 6%. (A,E) Nuclear DAPI

stain, (B,D) cardiomyocyte specific nuclear stain, MEF2A, (C.G) Alexa Fluor®555 Red Fluorescent Control siRNA within the cytoplasm of the cardiomyocytes, and (D,H) a merged image of all 3 channels. (E-H) Zoomed

in view of one section of micrographs A-D. The fluorescent positive control can be seen within the cytoplasm of

each cell.

Page 109: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

91

Supplemental Figure 3.5. Raw count data for cytokinesis events following antagomir

treatment. (A) Total number of cytokinetic events for graphs shown in Figure 4 B, (B) Total number of cytokinetic events for graphs shown in Figure 4B, (C) Total number of cytokinet ic events for graphs shown in Figure 4C, and (d) Total number of cytokinetic events for graphs shown

in Figure 4C. Each graph indicates a separate experiment. Data analysis by one-way ANOVA (A)

p=0.0035, (B) p<0.0001, (C) p<0.0001 (D) p<0.0001 and Tukey’s Multiple Comparison Posttest to

identify differences between groups. ** indicates p ≤ 0.01, *** p ≤0.001 for antagomir treatment compared to negative control treatment. # indicates P≤0.05, ## P≤0.01, ### P≤0.001 for antagomir

treatment compared to all 11 antagomir cocktail treatment, ± s.e.m, n=3

Page 110: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

92

Supplemental Figure 3.6. Average Cytokinetic events and average RFU show similar % change trends in

transfected cardiomyocytes. For each transfected cocktail, the % change of mean cytokinetic events and RFU was

calculated compared to the negative control. (A) The same cardiomyocytes transfected and stained in Figure 4 b were passed through a fluorescent microplate reader. The Relative Fluorescent Units (RFU) were measured to determine the fluorescence intensity of Aurora B in each well. Aurora B fluorescence was highest in the

cardiomyocytes transfected with all 11 antagomirs. One-way ANOVA analysis (p=0.1) revealed that this method was not sufficiently sensitive to pick up significant changes in RFU between treatments. (B) The % change

calculated by each method shows similar trends between cocktails despite lack of significance in microplate reader

data.

Page 111: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

93

Supplemental Figure 3.7. RT-qPCR for cardiomyocyte housekeeping genes. (A)

Cardiomyocytes were transfected with a cocktail of all 11 antagomirs (25nM, n=2), anti-cel-67 negative control (100nM, n=2) or not transfected (n=2), and probed for RNU6 (small nuclear RNA component of the spliceosome) by RT-qPCR. RNU6 expression was not strongly affected

by antagomir treatments and was thus chosen as the housekeeping gene to be used for normalization for all subsequent miRNA RT-qPCR experiments. (B) Cardiomyocytes were

transfected with a cocktail of all 11 antagomirs (25nM, n=4), the final cocktail of 9 antagomirs (25nM, n=4) or anti-cel-67 negative control (100nM, n=4) or not transfected (n=3). Out of 5 housekeeping genes contained in the Qiagen RT2 profiler array (B2M, ACTB, LDHA, HPRT1

and RPLP1), the expression of LDHA and HPRT1 remained the most consistent between all three treatments and were thus chosen as the housekeeping genes to be used for normalization of

all subsequent genes within the array.

Page 112: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

94

Supplemental Figure 3.8. MirSystem miRNA target gene biological

pathway ranking score. For a given set of miRNA, expression levels are

used as the weight (ѿi). The weight of one miRNA is calculated by dividing its absolute expression value by the absolute sum of the expression values of all input miRNAs. Next, after identifying the target genes by the prediction

algorithms, for each functional category (biological pathway), the ranking score is obtained by summation of miRNAs times its enrichment –log (P-

value) from the predicted target genes

Page 113: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

95

Supplemental Table 3.1. Normalized RT-qPCR Ct values for miRNA expression levels

following 11 antagomir transfection. RT-qPCR for cardiomyocytes transfected with either

a cocktail of all 11 antagomirs (25nM each) or anti cel-67 as a negative control (100 nM). Data are represented as mean Ct values normalized to small RNA housekeeping gene RNU6.

Page 114: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

96

Chapter 4

Conclusion

4 Conclusion and Future Directions

4.1 General Discussion

4.1.1 Thesis summary

The heart is an organ with limited regenerative capacity. Shortly after birth, proliferation in the

mammalian heart almost completely comes to a halt271. The implication of this is that following

injury, any lost cardiac muscle cells will not be replaced. Rather, the heart will attempt to heal by

cardiac hypertrophy and scar formation. The precise evolutionary mechanisms that set us apart

from amphibians such as newts, that can regenerate their cardiac tissue, remains to be defined271.

Various principal investigators have postulated that it is either due to an evolutionary trade off

preventing the development of cancer272, which is an extremely rare occurrence within the heart.

Others suggest that an evolutionary prioritization of hemostasis, hypertrophy and fibrosis over

regeneration is the cause of the adult heart’s inherent inability to activate this process271.

Remarkably, we have been able to demonstrate that the cardiac-specific knockout of p53 and

MDM2 can cause cardiomyocytes to re-enter into the cell cycle, in stark contrast to wild type

hearts214. However, loss of both p53 and MDM2 in the heart also caused early onset lethality,

due to many perturbations in cardiac metabolism and structural integrity214. We therefore sought

to determine the mechanism responsible for this proliferative response. We reasoned that if we

could modulate a single part of the downstream system, as opposed to the entire circuitry

regulated by p53/MDM2, this could provide a more directed approach to promote cardiomyocyte

proliferation while avoiding the other negative outcomes. p53 is known to activate the expression

of miRNAs that act as tumor suppressors in other organs217. Intriguingly, a canonical p53-

regulated miRNA family, miR-15, was shown to be capable of regulating the post-natal mitotic

arrest of cardiomyocytes, among other important miRNAs102. Thus, the specific aim of this thesis

was to identify the miRNAs regulated by p53 and MDM2 that maintain cardiomyocyte cell cycle

arrest in the adult mammalian heart.

Page 115: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

97

Through comparison of miRNA expression profiles, we found that a distinct set of deregulated

miRNAs could be identified in each of the three cardiac-specific mutant strains of p53 and

MDM2. As expected, the cardiac miRNA profile of DKO mice had some overlap with the two

single knockouts, but also a subset of uniquely deregulated miRNAs were present. More

specifically, 11 miRNAs from this group become downregulated in the DKO hearts, and are

enriched for target genes within the cell cycle pathway. As miRNAs exert their function post-

transcriptionally through inhibition of their target genes’ mRNA transcripts, we hypothesized

that these miRNAs may function as “anti-proliferative” miRNAs, by inhibiting multiple target

genes within the cell cycle pathway. This would help to explain the cell cycle re-entry exhibited

by DKO mice, where these miRNAs are downregulated, and therefore no longer exerting their

anti-proliferative effects within the cell.

To further characterize these miRNAs as regulators of cell cycle arrest, we determined their

expression levels in a proliferative cell type in the heart, that is, cardiac fibroblasts. Cardiac

fibroblasts have a strong proliferative capacity in vitro, and we expected that this group of

miRNAs might be downregulated in these cells as compared to the non-proliferative

cardiomyocytes. Indeed, each miRNA was significantly downregulated in fibroblasts, supporting

the idea that they form a roadblock for cell cycle re-entry in cardiomyocytes. We therefore

hypothesized that inhibiting these 11 miRNAs in isolated cardiomyocytes might promote a

phenotype similar to what was observed in the DKO, and allow for cell cycle re-entry. To test

this hypothesis, we knocked down the expression of the 11 miRNAs using a cocktail of

antagomirs in cultured neonatal rat cardiomyocytes. We found that antagomir treatment did lead

to significantly increased numbers of cardiomyocytes undergoing cytokinesis, through the

upregulation of a widespread number of cell cycle regulators. Therefore, we were able to clearly

demonstrate that the identified 11 p53/MDM2-regulated miRNAs negatively regulated the

expression of crucial cell cycle mediators required by cardiomyocytes to undergo cell division.

We also demonstrated that two antagomirs, anti-let-7a and anti-miR-30c, were not necessary

within the initial cocktail to promote cardiomyocyte proliferation, although the reason behind

this remains to be elucidated, as discussed in the Discussion section 3.4.2 of Chapter 3.

These p53/MDM2-regulated miRNAs seem to act in concert to maintain cardiomyocyte cell

cycle arrest, as at least 9 out of the 11 must be inhibited to promote proliferation. Notably, each

Page 116: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

98

of these miRNAs have been shown to function as tumor suppressors in cell types more amenable

to ectopic cell cycle re-entry, whereby their upregulation can halt the progression of diverse

types of cancers.

4.1.2 The 11 p53/MDM2-regulated miRNAs are well characterized tumor suppressors

4.1.2.1 The miR-30 family

miR-30a, miR-30b, miR-30c and miR-30e all act as tumor suppressors in breast cancer,

colorectal cancer, chondrosarcoma, and multiple myeloma, through their inhibition over

oncogenic factors such as metadherin, the insulin receptor substrate 2 (Irs2), SRY-box 4 (SOX4),

and the WNT/β-catenin pathway. In each of these distinct cancer types, the expression of one or

more miR-30 family members was decreased, and the ectopic administration of mimics for these

miRNAs halted cancer progression through induction of cell cycle arrest and inhibition of

metastasis273–279.

4.1.2.2 The let-7 family

The let-7 family is also widely characterized as a group of individual tumor suppressor miRNAs.

Primarily, members of this family including let-7a and let-7f, were shown to be downregulated in

lung cancer which was correlated with an upregulation of the RAS oncogene280. Subsequently, it

was demonstrated that these miRNAs were also downregulated in neuroblastomas, breast cancer,

renal cell carcinoma and many others, and that they inhibit numerous oncogenes, the most

notable being c-MYC281–286. Interestingly, Ohshima et al (2010) showed exosomic secretion of

the let-7 family in a metastatic gastric cancer cell line, presumably for protection of the

cancerous cells against their tumor suppressive effects287. This highlights that the let-7 family

likely exerts a potent anti-tumor effect, that cancer cells must evolve mechanisms to circumvent.

Re-expression of the let-7 family in cancers has thus been a highly researched topic in cancer

therapeutics, and synthetic let-7 administration in multiple cancer cell lines has been shown to

promote cancer cell quiescence and an accumulation of cells in the G0 phase288. Furthermore, in

agreement with the research presented in this master’s thesis, upregulation of the let-7 family in

liver and in lung cancer cell lines promoted either directly or indirectly a downregulation of

many cell cycle regulating factors including cyclin A2, E2F5, CDC25 and SKP2288. Following

Page 117: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

99

treatment of rat neonatal cardiomyocytes with our cocktail of antagomirs, including two that

inhibit members of the let-7 family, we also saw a significant re-expression of all the genes listed

above.

4.1.2.3 miR-181a

miR-181a also has notable tumor suppressor properties. This miRNA is downregulated in

gliomas, lung cancer, and chronic myelogenous leukemia289–291. In lung cancer cells (A549

cells), and leukemic cells (K562 cells), re-expression of miR-181a was shown to directly

downregulate two oncogenes, KRAS proto-oncogene GTPase (KRAS)290 and RALA Ras like

proto-oncogene A (RalA)291 respectively, thereby inhibiting proliferation of these cells. miR-

181a overexpression of RalA in leukemic cells caused G2 phase arrest and induction of

apoptosis291. Thus, inhibiting this miRNA in cardiomyocytes may have therapeutic value, not

only in promoting cell cycle reactivation, but also by mitigating apoptosis, which is key in the

pathogenesis of many cardiac diseases.

4.1.2.4 miR-26b

miR-26b similarly exerts a strong tumor suppressive effect over multiple types of cancer. This

miRNA has been found significantly downregulated in colon cancer, breast cancer, melanoma,

and bladder cancer, and has been documented as a potent inhibitor of cell proliferation in many

of these diseases292–295. Other studies have shown that miR-26b potently inhibits the cell cycle at

the G1/S phase transition by repression of cyclin D2, cyclin E1 and CDK6 mRNA transcripts in

fibroblasts and spermatocyte derived cells296,297. In support of these findings, the data presented

in this thesis show a significant upregulation of both cyclin D2 and CDK6 following antagomir

cocktail treatment, which includes an inhibitor against miR-26b.

4.1.2.5 miR-204

Reduced expression of miR-204 in gastric cancer, breast cancer and acute myeloid leukemia

predicts poor patient prognosis, based on three different studies which indicate that this miRNA

clearly plays an important role in preventing cancer progression298–300. It has also been identified

to be strongly downregulated in bladder and colorectal cancers, where its re-expression promotes

the inhibition of the anti-apoptotic BCL2 gene301,302, and a member of the RAS oncogene family

Page 118: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

100

RAB22A301, sensitizing these cancer cells to chemotherapy by upregulating apoptosis and

inducing cell cycle arrest.

4.1.2.6 miR-149

Overwhelming amounts of research support miR-149 as a tumor suppressor that inhibits

proliferation and mediates cell cycle arrest when re-expressed in multiple cancers. It therefore

follows that it is downregulated in numerous cancers including gliomas, gastric cancers, breast

cancers, colorectal cancers, ovarian cancers among many others301,303–307. Furthermore, miR-149

was shown to be downregulated in cancer-associated fibroblasts, presumably to promote the

creation of a tumor microenvironment that is more amenable for tumor growth and metastasis. In

line with this view, miR-149 re-expression in cancer-associated fibroblasts inhibited the pro-

inflammatory cytokine, IL-6, that functions to promote the cross talk between tumor cells and

fibroblasts308. Thus, downregulation of this miRNA in cardiomyocytes following cardiac injury

may promote not only the beneficial effect of upregulating proliferation and repair, but also by

promoting an anti-inflammatory environment.

4.1.2.7 miR-194

Finally, miR-194 has also been well characterized as a tumor suppressor in various cancers

including oral squamous cell carcinoma, colorectal cancer and hepatocellular carcinoma309–311.

On top of this, low levels of miR-194 are now known as an independent risk factor for the

reoccurrence of colorectal adenomas, with a high sensitivity and specificity of >70% as a

predictor312. It is interesting to note that this miRNA has also been found to work in a feedback

loop with p53. Wang et al. (2015) demonstrated that this miRNA can directly target the mitogen-

activated protein kinase kinase kinase kinase 4 (MAP4K4)309. Intriguingly, MDM2 is a

downstream target of MAP4K4, and inhibition of this kinase by miR-194 downregulated the

expression of MDM2, in turn, leading to p53 stabilization. This finding serves to explain the

broad downregulation of miR-194 in many cancers.

Page 119: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

101

4.1.3 Tumor suppressive p53/MDM2-regulated miRNAs may function synergistically to promote cardiac cell cycle arrest

With the discussion above, there is a large amount of evidence from the cancer field that the 11

p53/MDM2-regulated miRNAs identified in this thesis are crucial inhibitory molecules

regulating the pathways of growth and proliferation. This is supported by the miRNA target gene

analysis we conducted that listed “pathways in cancer” to be the second most enriched biological

pathway regulated by this group of miRNAs, preceded only by the MAPK pathway. If the

postulated evolutionary reasoning behind the lack of regenerative potential in the heart is due to

a trade off in the prevention of cancer, it follows well that these potent tumor suppressor

miRNAs would be upregulated in the heart. The downregulation of just one of these miRNAs

has the ability to heighten an individual’s risk for the development of cancer in tissues that are

more amenable to cell cycle re-entry. It is therefore plausible that the downregulation of all these

inhibitors together is required in cardiomyocytes to remove the rigorous block in the cell cycle

that they are subject to. Furthermore, the expression levels of these miRNAs within a cell may

play a role in defining their regenerative capacity, e.g. upregulated in cells which are strongly

refractory to proliferative stimuli such as cardiomyocytes, lower levels of expression in cells

which retain the natural ability to proliferate such as fibroblasts and lowest levels of expression

in cells which undergo aberrant growth/proliferation such as tumor cells. These findings also

highlight that the potency of p53 as a tumor suppressor may be strengthened through the

activation of multiple downstream tumor suppressor miRNAs. The upregulated expression of

these miRNAs in cardiomyocytes likely allows them to work synergistically to enhance p53’s

function as a tumor suppressor, all together creating a potent inhibitory network surveying

aberrant cell cycle re-entry through multiple different avenues.

The synergistic function of miRNAs has been previously described by Zu et al (2013), who show

that the overlapping target genes of miRNAs potentiates their ability to repress expression of

these genes313. Studies regarding the synergistic function of miRNAs have since focused on the

delivery of two miRNA mimics to produce a more potent phenotype over the delivery of either

one individually. For example, to promote tumor suppression, combinations of miRNA mimics

of miR-34+let-7314, miR-143+miR-145315 and miR-10b+miR-21316 synergistically inhibited the

proliferation of tumor cells. In contrast, the combination of miR-133+miR-499 potentiates

Page 120: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

102

cardiomyocyte differentiation317, and the synergistic combination of miR-21+miR-146a

attenuates cardiac apoptosis following ischemia reperfusion124. However, in the present study,

we exploit this mechanism in reverse, whereby the inhibition of a large group of miRNAs

removes their synergistic repression over numerous cell cycle regulators, thereby promoting

cardiomyocytes proliferation, whereas inhibition of these miRNAs in isolation is not sufficiently

potent to promote this effect.

4.1.4 Mechanisms of cardiac regeneration with a therapeutic goal in mind

The findings described in this thesis shed light on a group of tumor suppressive miRNAs

previously unknown to function within the heart, that have the potential to be utilized in the

development of novel regenerative cardiovascular therapies. Following cardiac injury, such as

myocardial infarction, millions of cardiomyocytes can be lost during the ischemic process as

well as during reperfusion318. Such dramatic losses significantly disrupt the structural and

functional integrity of the heart. Thus, future cardiovascular research should be aimed at the

replacement of lost cardiomyocytes as opposed to preventing further damage from occurring by

mitigating the work-load of the heart. This would prevent the negative repercussions that ensue

when the heart attempts to compensate for these losses by fibrosis and hypertrophy. Ultimately,

the goal would be to prevent or reverse the development of heart failure by intrinsic regeneration.

The focus of regenerative cardiovascular therapies has historically revolved around two main

themes: promotion of endogenous repair through reactivation of the cardiomyocyte cell cycle or,

promotion of exogenous repair through delivery of cell therapies.

4.1.4.1 Cell-based cardiac regenerative therapies

The transplantation of multiple different cell types into the heart has been proposed as a means to

promote myocardial repair and regeneration. Although research has yielded promising results, it

has also revealed many caveats that come along with introducing exogenous cells into the heart.

Much research is still focused on determination of the optimal cell type to be used in

transplantation, promotion of proper cell integration, and understanding the mechanisms by

which benefit is being provided.

Some of the earliest work in this field focused on transplanting skeletal myoblasts (progenitor

cells found in skeletal muscle) into the heart, with the hope that they would differentiate into

Page 121: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

103

cardiomyocytes when provided the change in microenvironment319. Rather, these cells

differentiated into myotubes (early developmental stage of muscle fiber formed by the fusion of

multiple myoblasts to create a syncytium), and were unsuccessful at integrating electrically with

the host myocardium319. Nevertheless, multiple clinical trials were undertaken due to the

observations of functional improvements in pre-clinical trials. However, results suggested that

these cells had limited integration efficiency into human hearts and thus, put patients at risk for

arrhythmias319. These studies highlighted that due to the tightly organized electrical conductive

system in the heart, not present in other organs, a major issue for cardiac cell-based therapies

would be to optimize graft integration to avoid the onset of arrhythmogenesis,

Following this, most work has been focused on transplanting cardiac derived cell types, obtained

from either embryonic stem cells, induced pluripotent stem cells, or arising from resident or non-

resident cardiac progenitor cells320.

Due to their unlimited capacity for self-renewal, and their ability to be efficiently differentiated

into cardiomyocytes, embryonic stem cells were an appealing candidate for cardiac stem cell

based therapies320. Undifferentiated embryonic stem cells cannot be injected into tissues, due to

the development of teratomas (tumors derived from cells not normally present in host organ).

However, injection of embryonic stem cell derived cardiomyocytes into immunocompatible

hearts were able to form intracardiac grafts that improved heart function in mice, rats and other

small animal models320. These cells were also able to achieve electrical integration, and co-

delivery with paracrine factors or bio-engineered scaffolds improved engraftment and survival of

these cells321,322. Numerous drawbacks were also identified for the use of embryonic stem cell-

derived cardiomyocytes, including the ethical issues of cell procurement, determining

immunocompatibility and the likely requirement for immunosuppression, and the immaturity of

the cardiomyocytes derived from embryonic stem cells.323 This last drawback likely prevents

optimal cardiac integration and function. Furthermore, the safety of this therapeutic option is in

question, due to the unknown potential for the development of teratomas, the longevity of the

grafts and the possibility of de-integration leading to arryhthmogenesis323.

Induced pluripotent stem cells have more recently come to the forefront as a more advantageous

source of cells from which to derive cardiomyocytes for transplanting purposes323. The protocol

for cardiomyocyte derivation from induced pluripotent stem cells has been largely fine-tuned

Page 122: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

104

over the last few years, such that the cardiomyocytes derived from these cells more closely

resemble adult cardiomyocytes in terms of their calcium handling and contractile ability323.

Furthermore, deriving cardiomyocytes by this method side steps the ethical issues surrounding

embryonic stem cells and the requirement for immunosuppression (as induced pluripotent stem

cells can be generated autologously). Despite these improvements, logistical issues arise for

developing therapies based on the transplantation of cardiomyocytes derived from induced

pluripotent stem cells. Firstly, generating cardiomyocytes from autologous induced pluripotent

stem cells would require numerous months of preparation. For this reason, their use to treat the

immediate damage that occurs in acute illnesses such as myocardial infarction would not be

possible. Secondly, a uniform manufacturing protocol to generate induced pluripotent stem cell

derived cardiomyocytes is a large regulatory hurdle for their use in the clinic.

The search for a population of resident cardiac progenitor cells has been contentious. Although

these cells have been shown to exist, their function in the heart is poorly understood, as they are

clearly not efficiently activated following cardiac injury to regenerate the heart, unlike skeletal

muscle cell progenitors. Many different populations of cardiac progenitor cells have been

reported, expressing diverse stem cell markers such as KIT Proto-Oncogene Receptor Tyrosine

Kinase (C-kit) + cardiac progenitor cells324, ataxin-1 (SCA1) + cardiac progenitor cells325, ISL1+

cardiac progenitor cells326, and cardiosphere-derived cells327. The cell type of main focus has

been C-kit+ cells found within the heart. C-kit is a receptor tyrosine kinase that is highly

expressed on hematopoietic stem cells, mature circulating cells, mast cells, some endothelial

cells, and immature cardiomyocytes323. Therefore, this marker has the issue of lack of specificity

to cardiac lineage cells. However, differentiation of these cells into the three major cell types of

the heart, cardiomyocytes, smooth muscle cells and endothelial cells, has been reported328. As

such, when injected into the heart, they were found to form not only new cardiomyocytes, but

also new blood vessels, enhancing vascularization of the damaged areas328. However, the

cardiomyogenic activity of these C-kit+ cells, which arise from the bone marrow, has been

contested by many groups323. Despite this, a prominent clinical trial was carried out, whereby

autologous C-kit+ cells expanded from myocardial tissue harvested from patients who underwent

a coronary artery bypass graft, were transplanted back into these patients to determine if this

could improve the outcomes of ischemic heart failure237. Minor improvements of left ventricular

ejection fraction were observed (7% increase), but it was hypothesized that this improvement

Page 123: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

105

was due to the secretion of paracrine factors from the transplanted cells, as opposed to a direct

cardiomyogenic contribution328.

Non-resident cardiac progenitor cells have also been studied for their potential to promote

myocardial repair323. C-kit+, hematopoietic lineage negative, bone marrow cells were isolated

and transplanted into the border zone of myocardial infarcts in mice, and were shown to generate

new myocardium in the infarct zone329. Unfortunately, the differentiation of c-kit+ bone marrow

cells into cardiomyocytes could not be reproduced in other studies330,331. It has been

hypothesized that the transplanted cells may fuse to resident cardiomyocytes, and this could

account for the “differentiation” that was observed323. Due to the ease of access of these cells,

despite the discrepant findings in small animal models, clinical trials were carried out that

injected different types of bone marrow derived cells into the heart, including mesenchymal

stromal cells, hematopoietic cells, and endothelial progenitor cells332–334. The outcomes of these

trials were modest at best in terms of cardiac functional improvement332–334.

Another interesting regenerative concept has been the reprograming of cardiac fibroblasts into

cardiomyocytes. It raises the appealing possibility that the same cells which produce deleterious

scarring following myocardial infarction, could be utilized to replenish the lost cardiac cells323.

Retroviral delivery of three factors crucial in cardiomyocyte differentiation, TBX5, myocyte

enhancer factor 2C (MEF2C), and GATA4, has been shown to successfully reprogram murine

fibroblasts into “cardiomyocyte-like cells”, suggesting that this could also occur in vivo335.

Hurdles to overcome still remain for this novel regenerative option, such as delivery of these

factors through other mechanisms than integrating viruses which pose issues for clinical

translation.

The mechanisms behind how different cell-based therapies have led to modest improvements in

heart function remain to be elucidated. The initial goal of these methods were to replenish lost

cardiomyocytes through the differentiation of the injected cells into cardiomyocytes, or directly

injecting differentiated cardiomyocytes into the tissue. However, all of this work has

demonstrated that only a low, permanent cellular engraftment level is achievable. For example,

25% of injected C-kit + cardiac progenitor cells present at 5 minutes following transplantation

remained after 24 hours, and only 2.8% remained after 35 days336. Furthermore, engraftment

efficiency is even lower when cells are delivered via an intracoronary injection as opposed to

Page 124: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

106

directly into the myocardium, and only a limited number of cells can be injected directly into the

myocardium at one time323. Based on the low level of cellular engraftment, it is hypothesized

that the number of cardiomyocytes formed directly from these cells is too low to be conveying

the functional benefits reported. These observations led to the concept that the injected cells are

secreting paracrine factors that act on the endogenous myocardium by either affecting the non-

cardiomyocytes to promote angiogenesis, or function on the cardiomyocytes themselves to

promote survival or cell division323.

If one of the major hypothesized mechanisms by which cell-based therapies improve cardiac

function is through stimulating endogenous cardiomyocyte proliferation by the secretion of

paracrine factors, then upregulating this process by methods that circumvent the logistical issues

surrounding cell transplantation may prove to be a more efficient method for promoting

cardiomyocyte regeneration.

Numerous studies have attempted to drive cardiomyocytes into cell division, through the

manipulation of cell cycle activators or inhibitors or pro-mitogenic signaling pathways.

Overexpression of cell cycle activators such as cyclin D1, D2 or D3 in cardiac specific

transgenic mice successfully resulted in the upregulation of cardiomyocyte DNA synthesis and

potentially, cell cycle re-entry337. Transgenic delivery of cyclin D2 mice was the only model (out

of the three) where persistent DNA synthesis following myocardial infarction was observed,

albeit, at a low efficiency337. However, it is important to note that this study did not provide any

evidence that cyclin D2 overexpression upregulated the actual incidence of cardiomyocyte cell

division, and only DNA synthesis assays were performed. A more recent study demonstrated that

cyclin D1 overexpression in cardiomyocyte resulted in increased DNA synthesis but that these

cells were blocked from entering M phase due to a lack in activation of CDK181. Thus, it is

possible that hearts of cyclin D2 transgenic mice had cardiomyocytes increasing in cell size

through re-entry into G1 and S phase, but had no increase in the number of newly formed cells,

due to the block at M phase. In fact, the demonstration of cardiomyocytes actually undergoing

cytokinesis has been rare in this field of research. More commonly surrogate markers of

proliferation such as phosphorylated histone H3, as a marker of M phase, and the proliferation

antigen Ki-67 are utilized. This highlights one of the strengths of our study, in that we utilized

Aurora B-positive midbody structures as our primary marker for cardiomyocyte proliferation,

Page 125: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

107

which is to date some of the strongest evidence to suggest that cardiomyocytes are undergoing

the process of cytokinesis leading to the formation of new cells, as the mid-body is the last

structure linking the two daughter cells together70.

The manipulation of mitogenic signaling pathways that regulate proliferation is proposed to be a

more efficient method to promote cardiomyocyte cell division, as opposed to targeting one

specific cell cycle regulating factor323. This is a similar concept to what we have proposed:

targeting multiple miRNAs that have a broad effect on regulating cell cycle at different levels,

leading to the upregulation of multiple cell cycle factors, as opposed to one in particular. Studies

have shown that targeting the Hippo pathway, a highly evolutionarily conserved signaling

pathway that regulates proliferation, organ size, and cell survival, can have a strong effect on

cardiomyocyte proliferation338,339. Specifically, it has been shown that knockdown of the

upstream effector proteins in the Hippo pathway, or over expression of the downstream target

protein, yes associated protein 1 (YAP1), can promote cardiomyocyte proliferation and

ventricular wall thickening339. Conversely, inhibition of YAP1 during the course of cardiac

development led to cardiomyocyte hypoplasia and lethality340. Another study also showed that

the downstream target of YAP1, phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit

beta (PIK3CB) can upregulate cardiomyocyte proliferation when overexpressed338. They utilized

Aurora B as a marker for cardiomyocyte cytokinesis in vivo to demonstrate that cardiac specific

PIK3CB gain of function could promote the occurrence of cytokinesis in 0.01% of

cardiomyocytes, indicating how rare this process is, whereas the presence of PiH3 positive and

BrdU positive nuclei were increased 0.3% and 20% respectively338. The rate of cytokinesis

observed in this study is comparable with our observations. It would be interesting to determine

if combining both these approaches could have an additive effect on cardiomyocyte proliferation.

Administration of signaling molecules to the myocardium has also promoted cardiomyocyte cell

cycle re-entry. For example, treatment of cardiomyocytes with the cytokine fibroblast growth

factor-1, along with an inhibitor to the proapoptotic p38 MAPK pathway, promotes

cardiomyocyte proliferation, through a mechanism that promoted initial sarcomere

dedifferentiation, followed by proliferation341. It is interesting to note that a certain number of the

cells undergoing cytokinesis in our experiments had disorganized sarcomeric structures, whereas

cells that appeared to be in the very late stages of this process tended to have more maturely

Page 126: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

108

constructed sarcomeres. This observation is likely due to the massive cytoskeletal

rearrangements that arise throughout cytokinesis. In another approach, infusion of the molecule

neuregulin (NRG1) which signals through the ERBB2 receptor tyrosine kinase 2/4

(ERBBB2/ERBB4) pathway, was also shown to promote fetal and adult cardiomyocyte

proliferation, and these studies warranted a clinical trial342. NRG1 was subsequently infused into

patients with heart failure and promoted an acute improvement of cardiac function that lasted for

up to three months343. However, the mechanism of this functional improvement remains to be

elucidated.

As previously discussed, miRNAs also have emerged as important regulators of cardiac

development and the mediation of cell cycle arrest. Overexpression of miR-590 and miR-199a

was capable of promoting cardiomyocyte proliferation136. Others have demonstrated that

downregulating the expression of certain miRNAs such as the miR-15 family can promote

cardiomyocyte cell cycle re-entry and upregulate cytokinesis102. miRNAs are attractive

therapeutic targets because the miRNA mimic and antagomir technologies that have been

developed can be delivered as synthetic small molecules that have the capability of entering into

cells and exerting their function in vivo127. Another benefit to utilizing antagomirs or mimics is

that their effect is only transient, as opposed to utilizing viral vectors or other genome editing

techniques such as CRISPR. This is important because maintaining controlled levels of

proliferation is necessary to retain the structural integrity of the heart, and sustained proliferation

over extensive periods of time could have detrimental effects on heart function.

In all, the technique of stimulating endogenous cardiomyocyte cell cycle re-entry and

proliferation is very attractive in the field of cardiac regeneration, because it is through this

mechanism that regeneration occurs in lower vertebrates, as well as in neonatal mammals271.

This method promotes the generation of new, autologous cardiomyocytes, which implies that

they will be mechanically, electrically and vascularly integrated into the myocardium, because

they arise from cells that already have these properties, as opposed to exogenous cells that have

to surpass the hurdles posed by engraftment. Future work remains to determine whether the level

of cardiomyocyte proliferation that has been successfully induced in our lab and in others, is

sufficient to expand the cardiomyocyte population in a clinically meaningful manner.

Importantly, the optimal method for cardiomyocyte cell cycle reactivation is still up for debate. It

Page 127: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

109

is possible that taking a multi-pronged approach by combining multiple methods outlined in this

thesis e.g. targeting miRNAs, in tandem with the administration of certain cytokines such as

fibroblast growth factor-1, might be more effective than any one method on their own. The work

that is presented herein provides insight into the potential mechanisms by which cardiomyocytes

maintain cell cycle arrest, and offers novel targets that could be manipulated in a clinical setting

to promote cardiac regeneration. Ultimately, our final goal would be to initiate a phase III

clinical trial with the use of an optimized cocktail of antagomirs, delivered via an intra-coronary

injection at the time of reperfusion following presentation of patients with acute myocardial

infarction. By this approach, the antagomirs could reach the area of damage and transiently

reactivate the cell cycle in cardiomyocytes to help replenish the lost cardiomyocyte population,

while possibly also promoting other beneficial effects such as inhibiting apoptosis and

controlling inflammation.

4.2 Limitations and Future Directions

Despite the exciting findings revealed throughout this thesis, there are a number of limitations

that warrant further discussion, to pave the way for future experimentation and directions.

Firstly, to determine the initial miRNA expression profiles between the three knockout mice

(p53KO, Mdm2KO and DKO) in our study, we utilized RNA isolated from whole hearts, as

opposed to cardiomyocyte specific RNA. Although knockout of the three genes was carried out

using a cardiac-specific promoter, there is a high degree of cross talk between fibroblasts and

cardiomyocytes in the myocardium. For this reason, it is possible that miRNA gene expression

changes were also occurring in the fibroblasts, and by our method, there was no way to

determine if downregulation of the 11 identified “anti-proliferative” miRNAs was occurring

solely in cardiomyocytes. In an attempt to partially address this issue, we determined the

expression profile of these miRNAs in wild type cardiac-derived fibroblasts, and observed that

the miRNAs were already significantly downregulated, as compared to neonatal cardiomyocytes.

Therefore we hypothesized that the downregulation of these miRNAs that we observed between

DKO and wild type hearts was occurring in the cardiomyocytes, which allowed them to regain

their proliferative capacity. Another method to circumvent this limitation would be to

specifically isolate DKO cardiomyocytes by either magnetic assisted sorting or negative

Page 128: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

110

selection columns (which bind fibroblasts and endothelial cells but allow cardiomyocytes to pass

through). Performing the Nanostring miRNA array using RNA from these purified adult DKO

cardiomyocytes and wild type cardiomyocytes would provide us with a more precise depiction of

the miRNA gene expression changes occurring in cardiomyocytes specifically, in response to

loss of p53 and MDM2.

Secondly, to identify miRNA target genes, we utilized a target gene prediction software called

miRSystem. Computational prediction tools to determine miRNA target genes scan for Watson-

Crick base pairing nucleotides between the seed regions of miRNAs (nucleotides 2-7) and

3′UTR of genomic sequences, to identify putative miRNA binding sites344,345. Most prediction

tools now also make use of thermodynamics to predict the likelihood of these interactions to

occurs, as well as evolutionary conservation at the binding sites identified, binding site structural

accessibility and nucleotide composition of the regions flanking the binding sites to minimize

false positives, thereby increasing the specificity of target gene prediction344. Although use of

computational techniques is the most rapid and cost efficient method to determine putative

miRNA target genes, to date, miRNA target prediction tools continue to produce a high rate of

false positive hits, and an unknown number of false negatives344. Additionally, the predicted

target genes of a given miRNA derived from different prediction algorithms often do not yield

overlapping results. Due to these limitations, algorithm based miRNA target prediction tools

likely fall substantially short of capturing in complete detail the physical, temporal and spatial

requirements of miRNAs and their target mRNAs344. In effort to mitigate this limitation, we

utilized a software that combines data from 7 miRNA target gene prediction algorithms as well

as 2 experimental validation databases, whereby the target gene was required to be predicted by

4 out of the 7 algorithms to be considered a putative target. This likely produces a more accurate

representation of miRNA target genes compared to using only one of the algorithms. However,

the putative list of cell cycle target genes that we obtained from this program likely does not

reflect the exact mRNA transcripts targeted by the 11 identified miRNAs in vivo. In order to

improve the accuracy of the target genes predicted for the miRNAs of focus in this thesis, certain

experimental procedures are available. Traditionally, the identification of precise miRNA target

genes has been executed with the use of luciferase assays345. In this method, the target gene of

interest is linked to a luciferase reporter gene, and following overexpression of the miRNA of

Page 129: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

111

interest in vitro, luciferase activity is monitored (reduction in luciferase activity indicates

miRNA binding to target gene)345. While this method is highly specific, it is labour intensive, has

low throughput and requires pre-existing knowledge of the miRNA target gene. Alternatively,

many groups have utilized microarray platforms or next generation sequencing following the

immunoprecipitation of AGO2 or other proteins of the RISC complex, to capture all the mRNA

transcripts bound by RISC complexes within a cell (RIP-Chip, or RIP-Seq)345. Although this

technique is high-throughput, can illuminate cell type specific transcripts being silenced, and can

expose changes in gene silencing following diverse treatments, it still relies on computational

techniques to determine which miRNAs are targeting the mRNAs that were sequenced345.

Recently, a moderate throughput method has been described which allows the detection of

specific miRNA target genes by the use of synthetic miRNA-duplexes346. In this method, cells

are transfected with biotinylated miRNA duplexes, then, streptavidin coated beads are utilized to

pull down these duplexes, and the mRNAs bound to the duplexes are sequenced to determine the

direct mRNA targets for a given miRNA346. In our study, we carried out a cell cycle RT-qPCR

array following our antagomir treatments, and were capable of detecting an acceptable amount of

overlap between the target genes predicted by the miRSystem software, and the gene expression

changes detected by RT-qPCR. In the future, to determine the precise target genes of the 11

p53/MDM2-regulated miRNAs in cardiomyocytes, the technique described above could be

utilized. This would allow us to construct a more detailed model which accurately illustrates the

gene silencing effects that these miRNAs exert within the heart.

Thirdly, the detection method that we utilized to determine cardiomyocyte proliferation could be

optimized. Our method was based on the presence of Aurora B kinase positive mid-body

structures located between two cardiomyocytes. By fluorescence microscopy, the number of

these structures were counted in each well containing treated or non-treated cells (48 well plates

were utilized). Unfortunately, this method is inherently biased due to the fact that an individual

must determine what corresponds to an “Aurora B positive mid-body structure”. This can

sometimes be a difficult task due to the fact that the structures change in appearance depending

on the stage of cytokinesis that is occurring. Furthermore, cell counting was not executed in a

blinded manner, and the treatments in each well were known, which further increased bias. The

screening was also only carried out in one plane, and thus any proliferating cardiomyocytes

Page 130: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

112

found in a different plane could be missed, possibly resulting in an underestimation in the total

number of proliferating cells per well. Although the method utilized to detect proliferation is

more accurate than utilizing surrogate markers such as phosphorylated histone H3 or Ki-67,

alternative methods could be employed to minimize bias within the results obtained. For

example, the use of an automated microscope that takes a Z-stack of each well in its entirety, and

a computer program that mathematically computes the number of Aurora B positive signals

found in each well (based on a threshold defined manually), would be the ideal way to calculate

the number of proliferating cells in each well. For future experimentation, we will utilize the

Opera Phenix High-Content Screening System (PerkinElmer) that is now available. This system

contains a fluorescent confocal microscope that allows the screening of cells in many different

formats of plates (including 48 well), as opposed to only glass slides, and can be automated to

take in focus Z-stack images in each well. Using this system would largely increase the

reliability of our results.

Finally, there are limitations in the in vitro model system that we employed. Based on ease of

isolation and culture, we utilized neonatal rat cardiomyocytes to assess the ability of the diverse

antagomir cocktails to promote proliferation. Although this in vitro system has been extensively

utilized to characterize cardiomyocyte proliferation and cell cycle arrest, it is not optimal based

on the fact that neonatal cardiomyocytes have a higher proliferative capacity compared to adult

cardiomyocytes. Thus, upregulation of proliferation would be inherently easier to induce in these

cells, compared to cells isolated from adult hearts that have been arrested from the cell cycle for

many weeks or months. We transfect our cells at 4-5 days following birth, and it has been

suggested that by this time point cardiomyocytes have already exited the cell cycle, however it is

likely that over time, they become increasingly refractory to proliferative stimuli. Thus, to

provide strong evidence that the 11 identified “anti-proliferative” miRNAs are in fact regulating

cell cycle arrest in adult hearts, it is of utmost importance that the antagomir treatments we have

formulated be tested in either adult cardiomyocyte cultures, or in vivo, in adult mouse or rat

hearts.

Based on the limitations defined, the most important future experiment to be carried out would

be to determine the effect of the antagomir treatments in vivo. If positive results were obtained

following in vivo studies, whereby the antagomir treatments significantly upregulated adult

Page 131: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

113

cardiomyocyte proliferation, this would validate the results obtained from our in vitro screen.

LNA-modified antagomirs have been developed by the company Exiqon347. These molecules

potently downregulate miRNA targets when injected in vivo without the requirement of

transfection reagents, which can be highly toxic to cells. We would inject both the final

optimized cocktail of 9 antagomirs, and the 11 antagomir cocktail into mice to determine the

effect of downregulating p53/MDM2-regulated miRNAs in vivo. These injections could be

executed intramyocardially, to determine the precise effect of the treatment on the heart, as well

as intravenously, to determine if other tissues are also affected. For example, it will be

interesting to determine if cardiac fibroblast proliferation is upregulated by the antagomir

cocktails. We showed that the expression of the 11 identified miRNAs is already downregulated

in fibroblasts, and therefore their further downregulation may not have a potent effect.

Alternatively, this could significantly upregulate fibroblast proliferation to a deleterious amount

in the heart. It would also be of interest to determine if the antagomirs can cross the blood brain

barrier and promote proliferation in the nervous system, where certain cell types such as neurons

are also notorious for their lack of regenerative capability348. In vivo experiments would also help

elucidate the effect that inhibition of p53/MDM2-regulated miRNAs has on overall heart

function and survival, and would allow us to investigate other phenotypes such as hypertrophy,

and apoptosis that may be up or downregulated by the treatments. During these experiments, we

could investigate other pathways that these miRNAs have been predicted to regulate, such as the

MAPK pathway, the ERBB pathway, FGF signaling, and the phosphatidylinositide 3-kinase/

AKT serine/threonine kinase (PI3K/AKT) pathway, all of which have been shown to regulate

cardiomyocyte proliferation, as described above. The miRNAs identified in this thesis also have

multiple putative targets within each of these pathways, including the PIK3CB protein which

was shown to be an important regulator of YAP1 induced cardiomyocyte proliferation338

Certainly, the p53/MDM2-regulated miRNAs identified in the work presented herein have a

much more broad regulation over the cell cycle than just the precisely defined cell cycle pathway

itself. Delving into other signaling pathways which ultimately influence the cell cycle pathway

would provide us with a more holistic understanding of how these miRNAs regulate cell cycle

arrest within cardiomyocytes.

Page 132: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

114

Ultimately, if the antagomir treatments we have devised successfully promote adult

cardiomyocyte proliferation, we could begin to determine their therapeutic value. We could

investigate the use of these treatments in models of ischemia/reperfusion or myocardial

infarction, through intracoronary injection (in larger animal models), or by peri-infarct injections.

Subsequently, proliferation of cardiomyocytes would be assessed to determine if the cocktails

can upregulate cell cycle re-entry following injury, and if this has therapeutic value based on

improved cardiac functional outcomes.

Identifying miRNAs regulated by other tumor suppressors or oncogenes could provide us with

further insight into cardiac cell cycle regulation. Our lab has multiple other cardiac specific

knockout strains for crucial tumor suppressors such as ARF and RB. Eventually, we would like

to utilize the same approach taken throughout this study to elucidate novel miRNA subsets

regulated by these proteins. It will be intriguing to determine if the miRNA profiling in these

mutant mice will have overlaps with the miRNA profiles from p53/MDM2 knockout mice, as the

ARF/RB and p53/MDM2 signaling pathways are highly interconnected349. Alternatively,

inhibiting miRNAs regulated by both pathways uniquely could produce an additive effect on

cardiomyocyte proliferation. These future avenues of research highlight that the intricately

interconnected pathways governing the cell cycle in cardiomyocytes remain largely unknown,

and warrant substantial additional research.

4.3 Overall Conclusion

To conclude, the research presented in this thesis demonstrated that the cardiac cell cycle is

tightly regulated by complex pathways including tumor suppressors and their downstream small

RNA effectors. We have identified 11 miRNAs regulated by the presence of the crucial tumor

suppressor circuitry of p53 and MDM2 in cardiomyocytes, which function to promote cell cycle

arrest within these cells. Importantly, we highlight the concept of synergism within the cell,

whereby multiple regulators of a pathway function together to promote a specific phenotype.

This research breaks away from the mold of “one gene, one phenotype” or the classical miRNA

studies that conclude one given miRNA target gene is the effector of the intracellular changes

elicited following deregulation of the miRNA. Instead, it shows that regulation of the cell cycle

within cardiomyocytes involves a complex network of genes mediating this process with varying

Page 133: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

115

degrees of redundancy, explaining the limited success in cell cycle reactivation through

modulating a single activator or inhibitor. Importantly, this thesis has provided an increased

understanding on how cardiomyocytes maintain cell cycle arrest, and paves the way for the

development of novel cardiovascular therapies by inhibiting groups of miRNAs who

synergistically dampen the regenerative potential in the heart.

Page 134: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

116

5 References

1. Kresh, J. Y. & Armour, J. A. The heart as a self‐regulating system: integration of homeodynamic mechanisms. Technology and Health Care 5, 159–169 (1997).

2. Nawrat, Z. Review of Research in Cardiovascular Devices. Handbook of Polymer

Applications in Medicine and Medical Devices (2013). doi:10.1016/B978-0-323-22805-3.00008-6

3. Torrent-Guasp, F. et al. Towards new understanding of the heart structure and function. Eur. J. Cardio-thoracic Surg. 27, 191–201 (2005).

4. Fukuta, H. & Little, W. C. The Cardiac Cycle and the Physiologic Basis of Left

Ventricular Contraction, Ejection, Relaxation, and Filling. Heart Fail. Clin. 4, 1–11 (2008).

5. Little, W. C. & Applegate, R. J. Congestive heart failure: systolic and diastolic function. J. Cardiothorac. Vasc. Anesth. 7, 2–5 (1993).

6. Gao, S., Ho, D., Vatner, D. E. & Vatner, S. F. Echocardiography in Mice. Curr. Protoc.

Mouse Biol. 1, 71–83 (2011).

7. Interstitial Fibrosis in Heart Failure | Francisco Villarreal | Springer. at

<http://www.springer.com/us/book/9780387228242>

8. Woodcock, E. a. & Matkovich, S. J. Cardiomyocytes structure, function and associated pathologies. Int. J. Biochem. Cell Biol. 37, 1746–1751 (2005).

9. Harvey, P. A. & Leinwand, L. A. The cell biology of disease: cellular mechanisms of cardiomyopathy. J. Cell Biol. 194, 355–65 (2011).

10. Liu, Z., Yue, S., Chen, X., Kubin, T. & Braun, T. Regulation of cardiomyocyte polyploidy and multinucleation by cyclinG1. Circ. Res. 106, 1498–1506 (2010).

11. Hoshijima, M. Mechanical stress-strain sensors embedded in cardiac cytoskeleton: Z disk,

titin, and associated structures. Am. J. Physiol. Heart Circ. Physiol. 290, H1313–25 (2006).

12. Bootman, M. D., Smyrnias, I., Thul, R., Coombes, S. & Roderick, H. L. Atrial cardiomyocyte calcium signalling. Biochim. Biophys. Acta - Mol. Cell Res. 1813, 922–934 (2011).

13. Review, S. A Literature Review of Cardiovascular Disease Management Programs in Managed Care Populations. 10, (2004).

14. Pantely, G. A. & Bristow, J. D. Ischemic cardiomyopathy. Prog. Cardiovasc. Dis. 27, 95–114

15. Current Review of Cerebrovascular Disease. (Springer Science & Business Media, 2000).

at <https://books.google.com/books?id=wWCSBAAAQBAJ&pgis=1>

16. Statistics. at

<http://www.heartandstroke.com/site/c.ikIQLcMWJtE/b.3483991/k.34A8/Statistics.htm>

17. Grech, E. D. Pathophysiology and investigation of coronary artery disease. BMJ 326, 1027–30 (2003).

Page 135: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

117

18. Cassar, A., Holmes, D. R., Rihal, C. S. & Gersh, B. J. Chronic coronary artery disease: diagnosis and management. Mayo Clin. Proc. 84, 1130–46 (2009).

19. Pangonyte, D. et al. Cardiomyocyte remodeling in ischemic heart disease. Medicina (Kaunas). 44, 848–54 (2008).

20. Gustafsson, A. B. & Gottlieb, R. A. Autophagy in ischemic heart disease. Circ. Res. 104, 150–8 (2009).

21. Zaman, A. G. The role of plaque rupture and thrombosis in coronary artery disease.

Atherosclerosis 149, 251 – 266

22. Neppl, R. L. & Wang, D.-Z. The myriad essential roles of microRNAs in cardiovascular

homeostasis and disease. Genes Dis. 1, 18–39 (2014).

23. Hausenloy, D. J. & Yellon, D. M. Myocardial ischemia-reperfusion injury: a neglected therapeutic target. J. Clin. Invest. 123, 92–100 (2013).

24. French, B. A. & Kramer, C. M. Mechanisms of Post-Infarct Left Ventricular Remodeling. Drug Discov. Today. Dis. Mech. 4, 185–196 (2007).

25. Qian, T., Nieminen, A. L., Herman, B. & Lemasters, J. J. Mitochondrial permeability transition in pH-dependent reperfusion injury to rat hepatocytes. Am. J. Physiol. 273, C1783–92 (1997).

26. Javadov, S., Karmazyn, M. & Escobales, N. Mitochondrial Permeability Transition Pore Opening as a Promising Therapeutic Target in Cardiac Diseases. J. Pharmacol. Exp. Ther.

330, 670–678 (2009).

27. Lloyd-Jones, D. M. Lifetime Risk for Developing Congestive Heart Failure: The Framingham Heart Study. Circulation 106, 3068–3072 (2002).

28. Krum, H. & Abraham, W. T. Heart failure. Lancet 373, 941–955 (2009).

29. Jessup, M. & Brozena, S. Heart Failure. http://dx.doi.org/10.1056/NEJMra021498 (2009).

30. Sutton, M. G. S. J. & Sharpe, N. Left Ventricular Remodeling After Myocardial Infarction : Pathophysiology and Therapy. Circulation 101, 2981–2988 (2000).

31. Ertl, G. & Frantz, S. Healing after myocardial infarction. Cardiovasc. Res. 66, 22–32

(2005).

32. Silvestre, J.-S. et al. Activation of Cardiac Aldosterone Production in Rat Myocardial

Infarction : Effect of Angiotensin II Receptor Blockade and Role in Cardiac Fibrosis. Circulation 99, 2694–2701 (1999).

33. Costanzo, M. R. et al. Selection and Treatment of Candidates for Heart Transplantation :

A Statement for Health Professionals From the Committee on Heart Failure and Cardiac Transplantation of the Council on Clinical Cardiology, American Heart Association.

Circulation 92, 3593–3612 (1995).

34. Lund, L. H. et al. The Registry of the International Society for Heart and Lung Transplantation: 32nd Official Adult Heart Transplantation Report - 2015. J. Hear. Lung

Transplant. 34, online document (2015).

35. Givertz, M. M. Cardiology patient pages: ventricular assist devices: important information

Page 136: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

118

for patients and families. Circulation 124, e305–11 (2011).

36. Nurse, P. A long twentieth century of the cell cycle and beyond. Cell 100, 71–78 (2000).

37. Rhind, N. & Russell, P. Signaling pathways that regulate cell division. Cold Spring Harb. Perspect. Biol. 4, a005942– (2012).

38. Lim, S. & Kaldis, P. Cdks, cyclins and CKIs: roles beyond cell cycle regulation. Development 140, 3079–93 (2013).

39. Nurse, P. & Thuriaux, P. Regulatory genes controlling mitosis in the fission yeast

Schizosaccharomyces pombe. Genetics 96, 627–37 (1980).

40. Pardee, A. B. G1 events and regulation of cell proliferation. Science (80-. ). 246, 603–609

(1989).

41. Donjerkovic, D. & Scott, D. W. Regulation of the G1 phase of the mammalian cell cycle. Cell Res. 10, 1–16 (2000).

42. Cooper, G. M. The Eukaryotic Cell Cycle. (2000). at <http://www.ncbi.nlm.nih.gov/books/NBK9876/>

43. Bertoli, C., Skotheim, J. M. & de Bruin, R. A. M. Control of cell cycle transcription during G1 and S phases. Nat. Rev. Mol. Cell Biol. 14, 518–28 (2013).

44. Kang, S., Warner, M. D. & Bell, S. P. Multiple functions for Mcm2-7 ATPase motifs

during replication initiation. Mol. Cell 55, 655–65 (2014).

45. Thu, Y. M. & Bielinsky, A.-K. Enigmatic roles of Mcm10 in DNA replication. Trends

Biochem. Sci. 38, 184–94 (2013).

46. Langston, L. D. et al. CMG helicase and DNA polymerase form a functional 15-subunit holoenzyme for eukaryotic leading-strand DNA replication. Proc. Natl. Acad. Sci. 111,

15390–15395 (2014).

47. Bruck, I. & Kaplan, D. L. GINS and Sld3 compete with one another for Mcm2-7 and

Cdc45 binding. J. Biol. Chem. 286, 14157–67 (2011).

48. Sun, J. et al. The architecture of a eukaryotic replisome. Nat. Struct. Mol. Biol. 22, 1–9 (2015).

49. Leman, A. R. & Noguchi, E. The replication fork: understanding the eukaryotic replication machinery and the challenges to genome duplication. Genes (Basel). 4, 1–32

(2013).

50. Aladjem, M. I. Replication in context: dynamic regulation of DNA replication patterns in metazoans. Nat. Rev. Genet. 8, 588–600 (2007).

51. Woo, R. a & Poon, R. Y. C. Cyclin-dependent kinases and S phase control in mammalian cells. Cell Cycle 2, 316–324 (2003).

52. Rosenberg, A. R. et al. Overexpression of human cyclin A advances entry into S phase. Oncogene 10, 1501–9 (1995).

53. Zindy, F. et al. Cyclin A is required in S phase in normal epithelial cells. Biochem.

Biophys. Res. Commun. 182, 1144–54 (1992).

Page 137: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

119

54. Huang, Y. et al. The Kinetics of G2 and M Transitions Regulated by B Cyclins. PLoS One 8, e80861 (2013).

55. van den Heuvel, S. & Harlow, E. Distinct roles for cyclin-dependent kinases in cell cycle control. Science 262, 2050–4 (1993).

56. Riabowol, K. et al. The cdc2 kinase is a nuclear protein that is essential for mitosis in mammalian cells. Cell 57, 393–401 (1989).

57. Abe, S. et al. The initial phase of chromosome condensation requires Cdk1-mediated

phosphorylation of the CAP-D3 subunit of condensin II. Genes Dev. 25, 863–874 (2011).

58. Alberts, B. et al. Intracellular Control of Cell-Cycle Events. (2002).

59. Cooper, G. M. The Events of M Phase. (2000).

60. Peters, J.-M., Tedeschi, A. & Schmitz, J. The cohesin complex and its roles in chromosome biology. Genes Dev. 22, 3089–3114 (2008).

61. Srivastava, V., Iglesias, P. A. & Robinson, D. N. Cytokinesis: Robust cell shape regulation. Semin. Cell Dev. Biol. 53, 39–44 (2016).

62. Kuilman, T. et al. Identification of Cdk targets that control cytokinesis. EMBO J. 34, 81–96 (2015).

63. Gallant, P. & Nigg, E. A. Cyclin B2 undergoes cell cycle-dependent nuclear translocation

and, when expressed as a non-destructible mutant, causes mitotic arrest in HeLa cells. J. Cell Biol. 117, 213–24 (1992).

64. Visintin, R. et al. The Phosphatase Cdc14 Triggers Mitotic Exit by Reversal of Cdk-Dependent Phosphorylation. Mol. Cell 2, 709–718 (1998).

65. Wäsch, R. & Cross, F. R. APC-dependent proteolysis of the mitotic cyclin Clb2 is

essential for mitotic exit. Nature 418, 556–62 (2002).

66. Visintin, R., Hwang, E. S. & Amon, A. Cfi1 prevents premature exit from mitosis by

anchoring Cdc14 phosphatase in the nucleolus. Nature 398, 818–23 (1999).

67. Sullivan, M. & Morgan, D. O. Finishing mitosis, one step at a time. Nat. Rev. Mol. Cell Biol. 8, 894–903 (2007).

68. Azzam, R. et al. Phosphorylation by cyclin B-Cdk underlies release of mitotic exit activator Cdc14 from the nucleolus. Science 305, 516–9 (2004).

69. Hu, C.-K., Coughlin, M. & Mitchison, T. J. Midbody assembly and its regulation during cytokinesis. Mol. Biol. Cell 23, 1024–34 (2012).

70. Murata-Hori, M., Tatsuka, M. & Wang, Y.-L. Probing the dynamics and functions of

aurora B kinase in living cells during mitosis and cytokinesis. Mol. Biol. Cell 13, 1099–108 (2002).

71. D’Avino, P. P. & Capalbo, L. New Auroras on the Roles of the Chromosomal Passenger Complex in Cytokinesis: Implications for Cancer Therapies. Front. Oncol. 5, 221 (2015).

72. Giet, R. & Glover, D. M. Drosophila aurora B kinase is required for histone H3

phosphorylation and condensin recruitment during chromosome condensation and to

Page 138: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

120

organize the central spindle during cytokinesis. J. Cell Biol. 152, 669–82 (2001).

73. Adams, R. R., Maiato, H., Earnshaw, W. C. & Carmena, M. Essential roles of Drosophila

inner centromere protein (INCENP) and aurora B in histone H3 phosphorylation, metaphase chromosome alignment, kinetochore disjunction, and chromosome segregation.

J. Cell Biol. 153, 865–80 (2001).

74. Kaitna, S., Mendoza, M., Jantsch-Plunger, V. & Glotzer, M. Incenp and an aurora-like kinase form a complex essential for chromosome segregation and efficient completion of

cytokinesis. Curr. Biol. 10, 1172–81 (2000).

75. Xin, M., Olson, E. N. & Bassel-Duby, R. Mending broken hearts: cardiac development as

a basis for adult heart regeneration and repair. Nat. Rev. Mol. Cell Biol. 14, 529–41 (2013).

76. Evans, S. M., Yelon, D., Conlon, F. L. & Kirby, M. L. Myocardial lineage development.

Circ. Res. 107, 1428–44 (2010).

77. Pasumarthi, K. B. S. & Field, L. J. Cardiomyocyte Cell Cycle Regulation. Circ. Res. 90,

1044–1054 (2002).

78. Li, F., Wang, X., Capasso, J. M. & Gerdes, A. M. Rapid transition of cardiac myocytes from hyperplasia to hypertrophy during postnatal development. J. Mol. Cell. Cardiol. 28,

1737–46 (1996).

79. Porrello, E. R. et al. Transient Regenerative Potential of the Neonatal Mouse Heart.

Science (80-. ). 331, 1078–1080 (2011).

80. Tane, S. et al. CDK inhibitors, p21Cip1 and p27Kip1, participate in cell cycle exit of mammalian cardiomyocytes. Biochem. Biophys. Res. Commun. 443, 1105–1109 (2014).

81. Tane, S. et al. Two inhibitory systems and CKIs regulate cell cycle exit of mammalian cardiomyocytes after birth. Biochem. Biophys. Res. Commun. 466, 147–154 (2015).

82. He, L. & Hannon, G. J. MicroRNAs: small RNAs with a big role in gene regulation. Nat. Rev. Genet. 5, 522–531 (2004).

83. Ha, M. & Kim, V. N. Regulation of microRNA biogenesis. Nat. Rev. Mol. Cell Biol. 15,

509–524 (2014).

84. Wahid, F., Shehzad, A., Khan, T. & Kim, Y. Y. MicroRNAs: Synthesis, mechanism,

function, and recent clinical trials. Biochim. Biophys. Acta - Mol. Cell Res. 1803, 1231–1243 (2010).

85. Lee, R. C., Feinbaum, R. L. & Ambros, V. The C. elegans heterochronic gene lin-4

encodes small RNAs with antisense complementarity to lin-14. Cell 75, 843–854 (1993).

86. Han, J. et al. Molecular basis for the recognition of primary microRNAs by the Drosha-

DGCR8 complex. Cell 125, 887–901 (2006).

87. Diederichs, S. & Haber, D. A. Dual Role for Argonautes in MicroRNA Processing and Posttranscriptional Regulation of MicroRNA Expression. Cell 131, 1097–1108 (2007).

88. Khvorova, A., Reynolds, A. & Jayasena, S. D. Functional siRNAs and miRNAs Exhibit Strand Bias. Cell 115, 209–216 (2003).

Page 139: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

121

89. Okamura, K., Liu, N. & Lai, E. C. Distinct Mechanisms for MicroRNA Strand Selection by Drosophila Argonautes. Mol. Cell 36, 431–444 (2009).

90. Czech, B. et al. An endogenous small interfering RNA pathway in Drosophila. Nature 453, 798–802 (2008).

91. Winter, J., Jung, S., Keller, S., Gregory, R. I. & Diederichs, S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat. Cell Biol. 11, 228–234 (2009).

92. Winter, J. & Diederichs, S. in 3–22 (2011). doi:10.1007/978-1-60761-863-8_1

93. Winter, J. & Diederichs, S. Argonaute proteins regulate microRNA stability: Increased microRNA abundance by Argonaute proteins is due to microRNA stabilization. RNA Biol.

8, 1149–57

94. Petersen, C. P., Bordeleau, M.-E., Pelletier, J. & Sharp, P. A. Short RNAs Repress Translation after Initiation in Mammalian Cells. Mol. Cell 21, 533–542 (2006).

95. Wakiyama, M., Takimoto, K., Ohara, O. & Yokoyama, S. Let-7 microRNA-mediated mRNA deadenylation and translational repression in a mammalian cell-free system. Genes

Dev. 21, 1857–62 (2007).

96. Wu, L., Fan, J. & Belasco, J. G. MicroRNAs direct rapid deadenylation of mRNA. Proc. Natl. Acad. Sci. U. S. A. 103, 4034–9 (2006).

97. Zhao, Y. et al. Dysregulation of Cardiogenesis, Cardiac Conduction, and Cell Cycle in Mice Lacking miRNA-1-2. Cell 129, 303–317 (2007).

98. Chen, J.-F. et al. Targeted deletion of Dicer in the heart leads to dilated cardiomyopathy and heart failure. Proc. Natl. Acad. Sci. 105, 2111–2116 (2008).

99. da Costa Martins, P. A. et al. Conditional Dicer Gene Deletion in the Postnatal

Myocardium Provokes Spontaneous Cardiac Remodeling. Circulation 118, 1567–1576 (2008).

100. Rao, P. K. et al. Loss of cardiac microRNA-mediated regulation leads to dilated cardiomyopathy and heart failure. Circ. Res. 105, 585–94 (2009).

101. Hata, A. Functions of MicroRNAs in Cardiovascular Biology and Disease.

http://dx.doi.org/10.1146/annurev-physiol-030212-183737 (2013).

102. Porrello, E. R. et al. MiR-15 family regulates postnatal mitotic arrest of cardiomyocytes.

Circ. Res. 109, 670–679 (2011).

103. Matsumoto, S. et al. Circulating p53-responsive microRNAs are predictive indicators of heart failure after acute myocardial infarction. Circ. Res. 113, 322–6 (2013).

104. Corsten, M. F. et al. Circulating MicroRNA-208b and MicroRNA-499 Reflect Myocardial Damage in Cardiovascular Disease. Circ. Cardiovasc. Genet. 3, 499–506 (2010).

105. Matsumoto, S. et al. A subset of circulating microRNAs are predictive for cardiac death after discharge for acute myocardial infarction. Biochemical and Biophysical Research Communications 427, (2012).

106. Fan, Z.-X. & Yang, J. The role of microRNAs in regulating myocardial ischemia reperfusion injury. Saudi Med. J. 36, 787–93 (2015).

Page 140: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

122

107. Bostjancic, E., Zidar, N. & Glavac, D. MicroRNA microarray expression profiling in human myocardial infarction. Dis. Markers 27, 255–68 (2009).

108. Port, J. D. et al. Temporal expression of miRNAs and mRNAs in a mouse model of myocardial infarction. Physiol. Genomics 43, 1087–95 (2011).

109. Olivieri, F. et al. Admission levels of circulating miR-499-5p and risk of death in elderly patients after acute non-ST elevation myocardial infarction. International Journal of Cardiology 172, (2014).

110. Thum, T. et al. MicroRNA-21 contributes to myocardial disease by stimulating MAP kinase signalling in fibroblasts. Nature 456, 980–4 (2008).

111. Roy, S. et al. MicroRNA expression in response to murine myocardial infarction: miR-21 regulates fibroblast metalloprotease-2 via phosphatase and tensin homologue. Cardiovasc. Res. 82, 21–9 (2009).

112. Kato, M. et al. TGF-beta activates Akt kinase through a microRNA-dependent amplifying circuit targeting PTEN. Nat. Cell Biol. 11, 881–9 (2009).

113. da Costa Martins, P. A. et al. MicroRNA-199b targets the nuclear kinase Dyrk1a in an auto-amplification loop promoting calcineurin/NFAT signalling. Nat. Cell Biol. 12, 1220–7 (2010).

114. Montgomery, R. L. et al. Therapeutic inhibition of miR-208a improves cardiac function and survival during heart failure. Circulation 124, 1537–47 (2011).

115. El Azzouzi, H., Leptidis, S., Doevendans, P. A. & De Windt, L. J. HypoxamiRs: regulators of cardiac hypoxia and energy metabolism. Trends Endocrinol. Metab. (2015). doi:10.1016/j.tem.2015.06.008

116. Aurora, A. B. et al. MicroRNA-214 protects the mouse heart from ischemic injury by controlling Ca2+ overload and cell death. J. Clin. Invest. 122, 1222–32 (2012).

117. Boon, R. A. et al. MicroRNA-34a regulates cardiac ageing and function. Nature 495, 107–10 (2013).

118. Brown, J. H., Del Re, D. P. & Sussman, M. A. The Rac and Rho hall of fame: a decade of

hypertrophic signaling hits. Circ. Res. 98, 730–42 (2006).

119. Nagai, T. et al. Cdc42 plays a critical role in assembly of sarcomere units in series of

cardiac myocytes. Biochem. Biophys. Res. Commun. 305, 806–810 (2003).

120. Carè, A. et al. MicroRNA-133 controls cardiac hypertrophy. Nat. Med. 13, 613–8 (2007).

121. Yang, N. An overview of viral and nonviral delivery systems for microRNA. Int. J.

Pharm. Investig. 5, 179–81 (2015).

122. Baumann, V. & Winkler, J. miRNA-based therapies: strategies and delivery platforms for

oligonucleotide and non-oligonucleotide agents. Future Med. Chem. 6, 1967–84 (2014).

123. Wang, Z. in 211–223 (2011). doi:10.1007/978-1-60761-863-8_15

124. Huang, W. et al. Combination of microRNA-21 and microRNA-146a Attenuates Cardiac

Dysfunction and Apoptosis During Acute Myocardial Infarction in Mice. Mol. Ther. Acids 5, e296 (2016).

Page 141: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

123

125. Krützfeldt, J. et al. Silencing of microRNAs in vivo with ‘antagomirs’. Nature 438, 685–689 (2005).

126. Krützfeldt, J. et al. Specificity, duplex degradation and subcellular localization of antagomirs. Nucleic Acids Res. 35, 2885–2892 (2007).

127. Wahlquist, C. et al. Inhibition of miR-25 improves cardiac contractility in the failing heart. Nature 508, 531–5 (2014).

128. Bueno, M. J. & Malumbres, M. MicroRNAs and the cell cycle. Biochim. Biophys. Acta -

Mol. Basis Dis. 1812, 592–601 (2011).

129. Calin, G. A. et al. Frequent deletions and down-regulation of micro- RNA genes miR15

and miR16 at 13q14 in chronic lymphocytic leukemia. Proc. Natl. Acad. Sci. U. S. A. 99, 15524–9 (2002).

130. Bottoni, A. et al. miR-15a and miR-16-1 down-regulation in pituitary adenomas. J. Cell.

Physiol. 204, 280–5 (2005).

131. Bonci, D. et al. The miR-15a-miR-16-1 cluster controls prostate cancer by targeting

multiple oncogenic activities. Nat. Med. 14, 1271–7 (2008).

132. Takeshita, F. et al. Systemic delivery of synthetic microRNA-16 inhibits the growth of metastatic prostate tumors via downregulation of multiple cell-cycle genes. Mol. Ther. 18,

181–7 (2010).

133. Xia, L. et al. miR-15b and miR-16 modulate multidrug resistance by targeting BCL2 in

human gastric cancer cells. Int. J. cancer 123, 372–9 (2008).

134. Liu, Q. et al. miR-16 family induces cell cycle arrest by regulating multiple cell cycle genes. Nucleic Acids Res. 36, 5391–404 (2008).

135. Linsley, P. S. et al. Transcripts targeted by the microRNA-16 family cooperatively regulate cell cycle progression. Mol. Cell. Biol. 27, 2240–52 (2007).

136. Eulalio, A. et al. Functional screening identifies miRNAs inducing cardiac regeneration. Nature 492, 376–381 (2012).

137. Pomerantz, J. H. & Blau, H. M. Tumor suppressors: enhancers or suppressors of

regeneration? Development 140, 2502–12 (2013).

138. Sherr, C. J. et al. Principles of Tumor Suppression. Cell 116, 235–246 (2004).

139. Sun, W. & Yang, J. Functional mechanisms for human tumor suppressors. J. Cancer 1, 136–140 (2010).

140. Müller, H. & Helin, K. The E2F transcription factors: key regulators of cell proliferation.

Biochim. Biophys. Acta - Rev. Cancer 1470, M1–M12 (2000).

141. Vandel, L. et al. Transcriptional repression by the retinoblastoma protein through the

recruitment of a histone methyltransferase. Mol. Cell. Biol. 21, 6484–94 (2001).

142. Green, R. A., Wollman, R. & Kaplan, K. B. APC and EB1 function together in mitosis to regulate spindle dynamics and chromosome alignment. Mol. Biol. Cell 16, 4609–22

(2005).

Page 142: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

124

143. He, L. et al. RIZ1, but not the alternative RIZ2 product of the same gene, is underexpressed in breast cancer, and forced RIZ1 expression causes G2-M cell cycle

arrest and/or apoptosis. Cancer Res. 58, 4238–44 (1998).

144. Hannon, G. J. & Beach, D. p15INK4B is a potential effector of TGF-beta-induced cell

cycle arrest. Nature 371, 257–61 (1994).

145. Wang, T.-J. et al. Comparisons of Tumor Suppressor p53, p21, and p16 Gene Therapy Effects on Glioblastoma Tumorigenicity in Situ. Biochem. Biophys. Res. Commun. 287,

173–180 (2001).

146. Hirai, H., Roussel, M. F., Kato, J. Y., Ashmun, R. A. & Sherr, C. J. Novel INK4 proteins,

p19 and p18, are specific inhibitors of the cyclin D-dependent kinases CDK4 and CDK6. Mol. Cell. Biol. 15, 2672–2681 (1995).

147. Wade Harper, J. et al. The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1

cyclin-dependent kinases. Cell 75, 805–816 (1993).

148. Tomoda, K., Kubota, Y. & Kato, J. Degradation of the cyclin-dependent-kinase inhibitor

p27Kip1 is instigated by Jab1. Nature 398, 160–5 (1999).

149. Bertwistle, D., Sugimoto, M. & Sherr, C. J. Physical and functional interactions of the Arf tumor suppressor protein with nucleophosmin/B23. Mol. Cell. Biol. 24, 985–96 (2004).

150. Levine, A. J., Finlay, C. A. & Hinds, P. W. P53 is a tumor suppressor gene. Cell 116, S67–S70 (2004).

151. Sun, W. Functional Mechanisms for Human Tumor Suppressors. J. Cancer 136 (2010). doi:10.7150/jca.1.136

152. Benchimol, S. p53-dependent pathways of apoptosis. Cell Death Differ. 8, 1049–1051

(2001).

153. Steigerwald, K., Behbehani, G. K., Combs, K. A., Barton, M. C. & Groden, J. The APC

tumor suppressor promotes transcription- independent apoptosis in vitro. Mol. Cancer Res. 3, 78–89 (2005).

154. Müschen, M., Warskulat, U. & Beckmann, M. W. Defining CD95 as a tumor suppressor

gene. J. Mol. Med. (Berl). 78, 312–25 (2000).

155. DuHadaway, J. B., Sakamuro, D., Ewert, D. L. & Prendergast, G. C. Bin1 mediates

apoptosis by c-Myc in transformed primary cells. Cancer Res. 61, 3151–6 (2001).

156. Simpson, L. & Parsons, R. PTEN: Life as a Tumor Suppressor. Exp. Cell Res. 264, 29–41 (2001).

157. Seifert, M. & Reichrath, J. The role of the human DNA mismatch repair gene hMSH2 in DNA repair, cell cycle control and apoptosis: implications for pathogenesis, progression

and therapy of cancer. J. Mol. Histol. 37, 301–7 (2006).

158. Prolla, T. A., Christie, D. M. & Liskay, R. M. Dual requirement in yeast DNA mismatch repair for MLH1 and PMS1, two homologs of the bacterial mutL gene. Mol. Cell. Biol.

14, 407–15 (1994).

159. Shiloh, Y. ATM (ataxia telangiectasia mutated): expanding roles in the DNA damage

Page 143: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

125

response and cellular homeostasis. Biochem. Soc. Trans. 29, 661–6 (2001).

160. Hartman, A.-R. & Ford, J. M. BRCA1 and p53: compensatory roles in DNA repair. J.

Mol. Med. (Berl). 81, 700–7 (2003).

161. Nakanishi, K. et al. Interaction of FANCD2 and NBS1 in the DNA damage response. Nat.

Cell Biol. 4, 913–20 (2002).

162. Powell, E., Piwnica-Worms, D. & Piwnica-Worms, H. Contribution of p53 to Metastasis. Cancer Discov. 4, 405–414 (2014).

163. Hori, A. et al. Metastin Suppresses the Motility and Growth of CHO Cells Transfected with Its Receptor. Biochem. Biophys. Res. Commun. 286, 958–963 (2001).

164. Meehan, W. J. et al. Breast cancer metastasis suppressor 1 (BRMS1) forms complexes with retinoblastoma-binding protein 1 (RBP1) and the mSin3 histone deacetylase complex and represses transcription. J. Biol. Chem. 279, 1562–9 (2004).

165. Gomis-Rüth, F. X. et al. Mechanism of inhibition of the human matrix metalloproteinase stromelysin-1 by TIMP-1. Nature 389, 77–81 (1997).

166. Ohtaki, T. et al. Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor. Nature 411, 613–7 (2001).

167. Dong, J. T. et al. KAI1, a metastasis suppressor gene for prostate cancer on human

chromosome 11p11.2. Science 268, 884–6 (1995).

168. Bieging, K. T., Mello, S. S. & Attardi, L. D. Unravelling mechanisms of p53-mediated

tumour suppression. Nat. Rev. Cancer 14, 359–70 (2014).

169. Malkin, D. Li-fraumeni syndrome. Genes Cancer 2, 475–84 (2011).

170. Donehower, L. A. et al. Mice deficient for p53 are developmentally normal but

susceptible to spontaneous tumours. Nature 356, 215–21 (1992).

171. Jacks, T. et al. Tumor spectrum analysis in p53-mutant mice. Curr. Biol. 4, 1–7 (1994).

172. Purdie, C. A. et al. Tumour incidence, spectrum and ploidy in mice with a large deletion in the p53 gene. Oncogene 9, 603–9 (1994).

173. Benchimol, S. p53-dependent pathways of apoptosis. Cell Death Differ. 8, 1049–1051

(2001).

174. El-Deiry, W. et al. WAF1, a potential mediator of p53 tumor suppression. Cell 75, 817–

825 (1993).

175. Agarwal, M. L., Agarwal, A., Taylor, W. R. & Stark, G. R. p53 controls both the G2/M and the G1 cell cycle checkpoints and mediates reversible growth arrest in human

fibroblasts (p21/WAF1/Li-Fraumeni cells/tetracycline/mimosine/cyclin-cyclin-dependent kinase). Cell Biol. 92, 8493–8497 (1995).

176. Doumont, G. et al. G1 checkpoint failure and increased tumor susceptibility in mice lacking the novel p53 target Ptprv. EMBO J. 24, 3093–103 (2005).

177. Kastan, M. B. et al. A mammalian cell cycle checkpoint pathway utilizing p53 and

GADD45 is defective in ataxia-telangiectasia. Cell 71, 587–597 (1992).

Page 144: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

126

178. Hermeking, H. et al. 14-3-3σ Is a p53-Regulated Inhibitor of G2/M Progression. Mol. Cell 1, 3–11 (1997).

179. Toshiyuki, M. et al. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 80, 293–299 (1995).

180. Owen-Schaub, L. B. et al. Wild-type human p53 and a temperature-sensitive mutant induce Fas/APO-1 expression. Mol. Cell. Biol. 15, 3032–40 (1995).

181. Lehar, S. M. et al. Identification and cloning of EI24, a gene induced by p53 in etoposide-

treated cells. Oncogene 12, 1181–7 (1996).

182. Oda, K. et al. p53AIP1, a Potential Mediator of p53-Dependent Apoptosis, and Its

Regulation by Ser-46-Phosphorylated p53. Cell 102, 849–862 (2000).

183. Yu, J. et al. PUMA Induces the Rapid Apoptosis of Colorectal Cancer Cells. Mol. Cell 7, 673–682 (2001).

184. Oda, E. et al. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 288, 1053–8 (2000).

185. Attardi, L. D. et al. PERP, an apoptosis-associated target of p53, is a novel member of the PMP-22/gas3 family. Genes Dev. 14, 704–18 (2000).

186. Smith, M. L. & Seo, Y. R. p53 regulation of DNA excision repair pathways. Mutagenesis

17, 149–156 (2002).

187. Fitch, M. E. et al. The DDB2 nucleotide excision repair gene product p48 enhances global

genomic repair in p53 deficient human fibroblasts. DNA Repair (Amst). 2, 819–826 (2003).

188. Hwang, B. J., Ford, J. M., Hanawalt, P. C. & Chu, G. Expression of the p48 xeroderma

pigmentosum gene is p53-dependent and is involved in global genomic repair. Proc. Natl. Acad. Sci. U. S. A. 96, 424–8 (1999).

189. Vousden, K. H. & Ryan, K. M. p53 and metabolism. Nat. Rev. Cancer 9, 691–700 (2009).

190. Schwartzenberg-Bar-Yoseph, F., Armoni, M. & Karnieli, E. The tumor suppressor p53 down-regulates glucose transporters GLUT1 and GLUT4 gene expression. Cancer Res.

64, 2627–33 (2004).

191. Bensaad, K. et al. TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 126,

107–20 (2006).

192. Feng, Z., Zhang, H., Levine, A. J. & Jin, S. The coordinate regulation of the p53 and mTOR pathways in cells. Proc. Natl. Acad. Sci. U. S. A. 102, 8204–9 (2005).

193. Yee, K. S., Wilkinson, S., James, J., Ryan, K. M. & Vousden, K. H. PUMA- and Bax-induced autophagy contributes to apoptosis. Cell Death Differ. 16, 1135–45 (2009).

194. Wang, Z. V, Li, D. L. & Hill, J. A. Heart failure and loss of metabolic control. J. Cardiovasc. Pharmacol. 63, 302–13 (2014).

195. Martinet, W. et al. Autophagy in cardiovascular disease. Trends Mol. Med. 13, 482–91

(2007).

Page 145: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

127

196. Shi, D. & Gu, W. Dual Roles of MDM2 in the Regulation of p53: Ubiquitination Dependent and Ubiquitination Independent Mechanisms of MDM2 Repression of p53

Activity. Genes Cancer 3, 240–8 (2012).

197. Iwakuma, T. & Lozano, G. MDM2 , An Introduction. 1, 993–1000 (2003).

198. Momand, J. et al. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell 69, 1237–1245 (1992).

199. Moll, U. M. & Petrenko, O. The MDM2-p53 interaction. Mol. Cancer Res. 1, 1001–8

(2003).

200. Fakharzadeh, S. S., Trusko, S. P. & George, D. L. Tumorigenic potential associated with

enhanced expression of a gene that is amplified in a mouse tumor cell line. EMBO J. 10, 1565–9 (1991).

201. Oliner, J. D., Kinzler, K. W., Meltzer, P. S., George, D. L. & Vogelstein, B. Amplification

of a gene encoding a p53-associated protein in human sarcomas. Nature 358, 80–3 (1992).

202. Meek, D. W. Mechanisms of switching on p53: a role for covalent modification?

Oncogene 18, 7666–7675 (1999).

203. Meek, D. W. & Knippschild, U. Posttranslational modification of MDM2. Mol. Cancer Res. 1, 1017–26 (2003).

204. de Oca Luna, R. M., Wagner, D. S. & Lozano, G. Rescue of early embryonic lethality in mdm2-deficient mice by deletion of p53. Nature 378, 203–206 (1995).

205. Jones, S. N., Roe, A. E., Donehower, L. A. & Bradley, A. Rescue of embryonic lethality in Mdm2-deficient mice by absence of p53. Nature 378, 206–208 (1995).

206. Sütsch, G., Jenni, R., von Segesser, L. & Schneider, J. [Heart tumors: incidence,

distribution, diagnosis. Exemplified by 20,305 echocardiographies]. Schweiz. Med. Wochenschr. 121, 621–9 (1991).

207. Sano, M. et al. p53-induced inhibition of Hif-1 causes cardiac dysfunction during pressure overload. Nature 446, 444–448 (2007).

208. Ikeda, S., Hamada, M. & Hiwada, K. Cardiomyocyte apoptosis with enhanced expression

of P53 and bax in right ventricle after pulmonary arterial banding. Life Sci. 65, 925–933 (1999).

209. Long, X. et al. p53 and the hypoxia-induced apoptosis of cultured neonatal rat cardiac myocytes. J. Clin. Invest. 99, 2635–43 (1997).

210. Liu, P., Xu, B., Cavalieri, T. A. & Hock, C. E. Pifithrin-alpha attenuates p53-mediated

apoptosis and improves cardiac function in response to myocardial ischemia/reperfusion in aged rats. Shock 26, 608–14 (2006).

211. Naito, A. T. et al. Promotion of CHIP-Mediated p53 Degradation Protects the Heart From Ischemic Injury. Circ. Res. 106, 1692–1702 (2010).

212. Yoshida, Y. et al. p53-Induced inflammation exacerbates cardiac dysfunction during

pressure overload. J. Mol. Cell. Cardiol. 85, 183–198 (2015).

213. Billia, F. Control of cardiomyocyte proliferation by Mdm2 and p53. Funded Oper. grant

Page 146: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

128

to Can. Institutes Heal. Res. (2013).

214. Filio Billia, Ludger Hauck, Daniela Grothe, Amelia Fung, Shanna Stanley-Hasnain,

Roozbeh Aschar-Sobbi, Sanja Beca, Jagdish Butany, Peter H. Backx, T. W. M. Absence of p53/Mdm2 signaling triggers the development of heart failure and sudden cardiac death

in mice. (2016).

215. Leung, A. K. L. et al. MicroRNA functions in stress responses. Mol. Cell 40, 205–15 (2010).

216. Leung, A. K. L. et al. microRNAs: a safeguard against turmoil? Cell 130, 581–5 (2007).

217. Hermeking, H. MicroRNAs in the p53 network: micromanagement of tumour

suppression. Nat. Rev. Cancer 12, 613–626 (2012).

218. Hermeking, H. et al. p53 enters the microRNA world. Cancer Cell 12, 414–8 (2007).

219. Suzuki, H. I. et al. Modulation of microRNA processing by p53. Nature 460, 529–33

(2009).

220. Raver-Shapira, N. et al. Transcriptional activation of miR-34a contributes to p53-

mediated apoptosis. Mol. Cell 26, 731–43 (2007).

221. Chang, T.-C. et al. Transactivation of miR-34a by p53 broadly influences gene expression and promotes apoptosis. Mol. Cell 26, 745–52 (2007).

222. Bommer, G. T. et al. p53-mediated activation of miRNA34 candidate tumor-suppressor genes. Curr. Biol. 17, 1298–307 (2007).

223. Tazawa, H., Tsuchiya, N., Izumiya, M. & Nakagama, H. Tumor-suppressive miR-34a induces senescence-like growth arrest through modulation of the E2F pathway in human colon cancer cells. Proc. Natl. Acad. Sci. U. S. A. 104, 15472–7 (2007).

224. Tarasov, V. et al. Differential regulation of microRNAs by p53 revealed by massively parallel sequencing: miR-34a is a p53 target that induces apoptosis and G1-arrest. Cell

Cycle 6, 1586–93 (2007).

225. He, L. et al. A microRNA component of the p53 tumour suppressor network. Nature 447, 1130–4 (2007).

226. Corney, D. C., Flesken-Nikitin, A., Godwin, A. K., Wang, W. & Nikitin, A. Y. MicroRNA-34b and MicroRNA-34c are targets of p53 and cooperate in control of cell

proliferation and adhesion-independent growth. Cancer Res. 67, 8433–8 (2007).

227. Jin, L. et al. MicroRNA-149*, a p53-responsive microRNA, functions as an oncogenic regulator in human melanoma. Proc. Natl. Acad. Sci. 108, 15840–15845 (2011).

228. Hünten, S. et al. p53-Regulated Networks of Protein, mRNA, miRNA, and lncRNA Expression Revealed by Integrated Pulsed Stable Isotope Labeling With Amino Acids in

Cell Culture (pSILAC) and Next Generation Sequencing (NGS) Analyses. Mol. Cell. Proteomics 14, 2609–2629 (2015).

229. Sachdeva, M. et al. p53 represses c-Myc through induction of the tumor suppressor miR-

145. Proc. Natl. Acad. Sci. U. S. A. 106, 3207–12 (2009).

230. Ho, J. S. L., Ma, W., Mao, D. Y. L. & Benchimol, S. p53-Dependent Transcriptional

Page 147: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

129

Repression of c-myc Is Required for G1 Cell Cycle Arrest. Mol. Cell. Biol. 25, 7423–7431 (2005).

231. Kaller, M. et al. Genome-wide characterization of miR-34a induced changes in protein and mRNA expression by a combined pulsed SILAC and microarray analysis. Mol. Cell.

Proteomics 10, M111.010462 (2011).

232. Yamakuchi, M., Ferlito, M. & Lowenstein, C. J. miR-34a repression of SIRT1 regulates apoptosis. Proc. Natl. Acad. Sci. U. S. A. 105, 13421–6 (2008).

233. Siemens, H. et al. miR-34 and SNAIL form a double-negative feedback loop to regulate epithelial-mesenchymal transitions. Cell Cycle 10, 4256–71 (2011).

234. Boon, R. A. et al. MicroRNA-34a regulates cardiac ageing and function. Nature 495, 107–10 (2013).

235. Boon, R. A. et al. MicroRNA-34a regulates cardiac ageing and function. Nature 495,

107–10 (2013).

236. Matsumoto, S. et al. Circulating p53-responsive MicroRNAs are predictive indicators of

heart failure after acute myocardial infarction. Circ. Res. 113, 322–326 (2013).

237. Bolli, R. et al. Cardiac stem cells in patients with ischaemic cardiomyopathy (SCIPIO): Initial results of a randomised phase 1 trial. Lancet 378, 1847–1857 (2011).

238. Senyo, S. E., Lee, R. T. & Kühn, B. Cardiac regeneration based on mechanisms of cardiomyocyte proliferation and differentiation. Stem Cell Res. 13, 532–541 (2014).

239. Daniely, Y., Dimitrova, D. D. & Borowiec, J. A. Stress-Dependent Nucleolin Mobilization Mediated by p53-Nucleolin Complex Formation. Mol. Cell. Biol. 22, 6014–6022 (2002).

240. Olson, M. O. J. Sensing cellular stress: another new function for the nucleolus? Sci. STKE 2004, pe10 (2004).

241. Bouafia, A. et al. p53 Requires the Stress Sensor USF1 to Direct Appropriate Cell Fate Decision. PLoS Genet. 10, e1004309 (2014).

242. Itahana, K., Dimri, G. & Campisi, J. Regulation of cellular senescence by p53. Eur. J.

Biochem. 268, 2784–91 (2001).

243. Qian, Y. & Chen, X. Senescence regulation by the p53 protein family. Methods Mol. Biol.

965, 37–61 (2013).

244. Chen, Z. et al. Crucial role of p53-dependent cellular senescence in suppression of Pten-deficient tumorigenesis. Nature 436, 725–730 (2005).

245. Giaccia, A. J. & Kastan, M. B. The complexity of p53 modulation: emerging patterns from divergent signals. Genes Dev. 12, 2973–83 (1998).

246. Zhao, Z. et al. p53 loss promotes acute myeloid leukemia by enabling aberrant self-renewal. Genes Dev. 24, 1389–1402 (2010).

247. Hu, W., Feng, Z. & Levine, A. J. The Regulation of Multiple p53 Stress Responses is

Mediated through MDM2. Genes Cancer 3, 199–208 (2012).

Page 148: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

130

248. Fischer, M., Steiner, L. & Engeland, K. The transcription factor p53: not a repressor, solely an activator. Cell Cycle 13, 3037–58 (2014).

249. Hino, K., Fukao, T. & Watanabe, M. Regulatory Interaction of HNF1 to microRNA194 Gene During Intestinal Epithelial Cell Differentiation. Nucleic Acids Symp. Ser. 51, 415–

416 (2007).

250. Bader, A. G. miR-34 – a microRNA replacement therapy is headed to the clinic. Front. Genet. 3, 120 (2012).

251. Kang, W. et al. Targeting of YAP1 by microRNA-15a and microRNA-16-1 exerts tumor suppressor function in gastric adenocarcinoma. Mol. Cancer 14, 52 (2015).

252. Trang, P. et al. Systemic Delivery of Tumor Suppressor microRNA Mimics Using a Neutral Lipid Emulsion Inhibits Lung Tumors in Mice. Mol. Ther. 19, 1116–1122 (2011).

253. Emanueli, C. & Thum, T. miRNAGE-34 induces cardiac damAGE. Cell Res. 23, 866–867

(2013).

254. Hinkel, R., Ng, J. K. M. & Kupatt, C. Targeting microRNAs for cardiovascular

therapeutics in coronary artery disease. Curr. Opin. Cardiol. 29, 586–94 (2014).

255. Gill, C., Mestril, R. & Samali, A. Losing heart: the role of apoptosis in heart disease--a novel therapeutic target? FASEB J. 16, 135–146 (2002).

256. Sabbah, H. N., Sharov, V. G. & Goldstein, S. Cell death, tissue hypoxia and the progression of heart failure. Heart Fail. Rev. 5, 131–8 (2000).

257. Ehler, E., Moore-Morris, T. & Lange, S. Isolation and Culture of Neonatal Mouse Cardiomyocytes. J. Vis. Exp. e50154–e50154 (2013). doi:10.3791/50154

258. Shi, L. et al. p53-induced miR-15a/16-1 and AP4 form a double-negative feedback loop to

regulate epithelial-mesenchymal transition and metastasis in colorectal cancer. Cancer Res. 74, 532–42 (2014).

259. Koutsoulidou, A. et al. Expression of miR-1, miR-133a, miR-133b and miR-206 increases during development of human skeletal muscle. BMC Dev. Biol. 11, 34 (2011).

260. Lu, T.-P. et al. miRSystem: an integrated system for characterizing enriched functions and

pathways of microRNA targets. PLoS One 7, e42390 (2012).

261. Chen, W. & Frangogiannis, N. G. Fibroblasts in post-infarction inflammation and cardiac

repair. Biochim. Biophys. Acta 1833, 945–53 (2013).

262. Wei, Y. et al. Multifaceted roles of miR-1s in repressing the fetal gene program in the heart. Cell Res. 24, 278–92 (2014).

263. Berthet, C. & Kaldis, P. Cdk2 and Cdk4 cooperatively control the expression of Cdc2. Cell Div. 1, 10 (2006).

264. Malumbres, M. et al. Mammalian Cells Cycle without the D-Type Cyclin-Dependent Kinases Cdk4 and Cdk6. Cell 118, 493–504 (2004).

265. Berthet, C. et al. Cdk2 Knockout Mice Are Viable. Curr. Biol. 13, 1775–1785 (2003).

266. Aleem, E., Kiyokawa, H. & Kaldis, P. Cdc2–cyclin E complexes regulate the G1/S phase

Page 149: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

131

transition. Nat. Cell Biol. 7, 831–836 (2005).

267. Hurford, R. K., Cobrinik, D., Lee, M.-H. & Dyson, N. pRB and p107/p130 are required

for the regulated expression of different sets of E2F responsive genes.

268. Coqueret, O. et al. New roles for p21 and p27 cell-cycle inhibitors: a function for each cell

compartment? Trends Cell Biol. 13, 65–70 (2003).

269. Karlsson, C., Katich, S., Hagting, A., Hoffmann, I. & Pines, J. Cdc25B and Cdc25C differ markedly in their properties as initiators of mitosis. J. Cell Biol. 146, 573–84 (1999).

270. Baquero, M. T. et al. Stathmin expression and its relationship to microtubule-associated protein tau and outcome in breast cancer. Cancer 118, 4660–9 (2012).

271. Graham, R. M. & Harvey, R. P. The Ontogeny of Cardiac Regeneration. Circ. Res. 108, 1304–1305 (2011).

272. Blau, H. M. Re‘evolutionary’ Regenerative Medicine. Jama 305, 87 (2011).

273. Liao, W.-T. et al. MicroRNA-30b functions as a tumour suppressor in human colorectal cancer by targeting KRAS, PIK3CD and BCL2. J. Pathol. 232, 415–27 (2014).

274. Ouzounova, M. et al. MicroRNA miR-30 family regulates non-attachment growth of breast cancer cells. BMC Genomics 14, 139 (2013).

275. Zhu, E.-D. et al. miR-30b, Down-Regulated in Gastric Cancer, Promotes Apoptosis and

Suppresses Tumor Growth by Targeting Plasminogen Activator Inhibitor-1. PLoS One 9, e106049 (2014).

276. Zhao, J.-J. et al. miR-30-5p functions as a tumor suppressor and novel therapeutic tool by targeting the oncogenic Wnt/β-catenin/BCL9 pathway. Cancer Res. 74, 1801–13 (2014).

277. Lu, N. et al. Association of SOX4 regulated by tumor suppressor miR-30a with poor

prognosis in low-grade chondrosarcoma. Tumour Biol. 36, 3843–52 (2015).

278. Zhang, N. et al. MicroRNA-30a suppresses breast tumor growth and metastasis by

targeting metadherin. Oncogene 33, 3119–28 (2014).

279. Zhang, Q. et al. Role of MicroRNA 30a Targeting Insulin Receptor Substrate 2 in Colorectal Tumorigenesis. Mol. Cell. Biol. 35, 988–1000 (2015).

280. Johnson, S. M. et al. RAS is regulated by the let-7 microRNA family. Cell 120, 635–47 (2005).

281. Lee, Y. S. & Dutta, A. The tumor suppressor microRNA let-7 represses the HMGA2 oncogene. doi:10.1101/gad.1540407

282. Molenaar, J. J. et al. LIN28B induces neuroblastoma and enhances MYCN levels via let-7

suppression. Nat. Genet. 44, 1199–1206 (2012).

283. Kobayashi, M. et al. Ovarian cancer cell invasiveness is associated with discordant

exosomal sequestration of Let-7 miRNA and miR-200. J. Transl. Med. 12, 4 (2014).

284. Li4, W. Let-7a as a Tumor Suppressor Modulates Invasion via Smad2 Signal in Breast Cancer. 2,

285. Liu, Y., Yin, B., Zhang, C., Zhou, L. & Fan, J. Hsa-let-7a functions as a tumor suppressor

Page 150: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

132

in renal cell carcinoma cell lines by targeting c-myc. Biochem. Biophys. Res. Commun. 417, 371–375 (2012).

286. Chiu, S.-C. et al. Therapeutic potential of microRNA let-7: tumor suppression or impeding normal stemness. Cell Transplant. 23, 459–69 (2014).

287. Ohshima, K. et al. Let-7 microRNA family is selectively secreted into the extracellular environment via exosomes in a metastatic gastric cancer cell line. PLoS One 5, e13247 (2010).

288. Johnson, C. D. et al. The let-7 microRNA represses cell proliferation pathways in human cells. Cancer Res. 67, 7713–22 (2007).

289. Shi, L. et al. hsa-mir-181a and hsa-mir-181b function as tumor suppressors in human glioma cells. Brain Res. 1236, 185–193 (2008).

290. Ma, Z. et al. MiR-181a-5p inhibits cell proliferation and migration by targeting Kras in

non-small cell lung cancer A549 cells. doi:10.1093/abbs/gmv054

291. Fei, J. et al. miR-181a Post-Transcriptionally Downregulates Oncogenic RalA and

Contributes to Growth Inhibition and Apoptosis in Chronic Myelogenous Leukemia (CML). PLoS One 7, e32834 (2012).

292. Li, J. et al. MiRNA-26b inhibits proliferation by targeting PTGS2 in breast cancer.

Cancer Cell Int. 13, 7 (2013).

293. Zhang, Z. et al. Cancer Biology and Signal Transduction MicroRNA-26b Represses Colon

Cancer Cell Proliferation by Inhibiting Lymphoid Enhancer Factor 1 Expression. doi:10.1158/1535-7163.MCT-13-1000

294. Li, M., Long, C., Yang, G., Luo, Y. & Du, H. MiR-26b inhibits melanoma cell

proliferation and enhances apoptosis by suppressing TRAF5-mediated MAPK activation. Biochem. Biophys. Res. Commun. 471, 361–7 (2016).

295. Miyamoto, K. et al. Tumour-suppressive miRNA-26a-5p and miR-26b-5p inhibit cell aggressiveness by regulating PLOD2 in bladder cancer. Br. J. Cancer (2016). doi:10.1038/bjc.2016.179

296. Liu, Y. et al. Overexpression of miR-26b-5p regulates the cell cycle by targeting CCND2 in GC-2 cells under exposure to extremely low frequency electromagnetic fields. Cell

Cycle 15, 357–67 (2016).

297. Zhu, Y. et al. MicroRNA-26a/b and their host genes cooperate to inhibit the G1/S transition by activating the pRb protein. Nucleic Acids Res. 40, 4615–25 (2012).

298. Butrym, A. et al. Low expression of microRNA-204 (miR-204) is associated with poor clinical outcome of acute myeloid leukemia (AML) patients. (2012). doi:10.1186/s13046-

015-0184-z

299. Li, W. et al. Decreased expression of miR-204 is associated with poor prognosis in patients with breast cancer. Int J Clin Exp Pathol 7, 3287–3292 (2014).

300. Chen, X., Liu, X., Liu, H., Lu, Y. & Li, Y. Reduced expression of serum miR-204 predicts poor prognosis of gastric cancer. Mol. Res 15, (2016).

Page 151: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

133

301. Hwang, T. I.-S. et al. miR-204 acts as a tumor suppressor in human bladder cancer cell T24 by targeting antiapoptotic BCL2. Urol. Sci. (2016). doi:10.1016/j.urols.2016.02.004

302. Yin, Y. et al. miR-204-5p inhibits proliferation and invasion and enhances chemotherapeutic sensitivity of colorectal cancer cells by downregulating RAB22A. Clin.

Cancer Res. 20, 6187–99 (2014).

303. Zhan, Y. et al. MiRNA-149 modulates chemosensitivity of ovarian cancer A2780 cells to paclitaxel by targeting MyD88. J. Ovarian Res. 8, 48 (2015).

304. Xu, K. et al. MicroRNA-149 Suppresses Colorectal Cancer Cell Migration and Invasion by Directly Targeting Forkhead Box Transcription Factor FOXM1. Cell. Physiol.

Biochem. 35, 499–515 (2015).

305. Pan, S.-J. et al. MicroRNA-149 inhibits proliferation and invasion of glioma cells via blockade of AKT1 signaling. Int. J. Immunopathol. Pharmacol. 25, 871–81

306. Bischoff, A. et al. miR149 functions as a tumor suppressor by controlling breast epithelial cell migration and invasion. Cancer Res. 74, 5256–65 (2014).

307. Wang, Y. et al. MicroRNA-149 Inhibits Proliferation and Cell Cycle Progression through the Targeting of ZBTB2 in Human Gastric Cancer. PLoS One 7, e41693 (2012).

308. Li, P. et al. Epigenetic silencing of microRNA-149 in cancer-associated fibroblasts

mediates prostaglandin E2/interleukin-6 signaling in the tumor microenvironment. Cell Res. 25, 588–603 (2015).

309. Wang, B. et al. MiR-194, commonly repressed in colorectal cancer, suppresses tumor growth by regulating the MAP4K4/c-Jun/MDM2 signaling pathway. Cell Cycle 14, 1046–58 (2015).

310. Chi, H. miR-194 regulated AGK and inhibited cell proliferation of oral squamous cell carcinoma by reducing PI3K-Akt-FoxO3a signaling. Biomed. Pharmacother. 71, 53–7

(2015).

311. Bao, C. et al. NF-κB signaling relieves negative regulation by miR-194 in hepatocellular carcinoma by suppressing the transcription factor HNF-1α. Sci. Signal. 8, ra75 (2015).

312. Wang, Z.-H. et al. miR-194 as a predictor for adenoma recurrence in patients with advanced colorectal adenoma after polypectomy. Cancer Prev. Res. (Phila). 7, 607–16

(2014).

313. Zhu, W. et al. Dissection of Protein Interactomics Highlights MicroRNA Synergy. PLoS One 8, e63342 (2013).

314. Kasinski, A. L. et al. A combinatorial microRNA therapeutics approach to suppressing non-small cell lung cancer. Oncogene 34, 3547–3555 (2015).

315. Noguchi, S. et al. Replacement treatment with microRNA-143 and -145 induces synergistic inhibition of the growth of human bladder cancer cells by regulating PI3K/Akt and MAPK signaling pathways. Cancer Lett. 328, 353–61 (2013).

316. Dong, C. G. et al. Co-inhibition of microRNA-10b and microRNA-21 exerts synergistic inhibition on the proliferation and invasion of human glioma cells. Int. J. Oncol. 41,

1005–12 (2012).

Page 152: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

134

317. Pisano, F. et al. Combination of miRNA499 and miRNA133 Exerts a Synergic Effect on Cardiac Differentiation. Stem Cells 33, 1187–1199 (2015).

318. Laflamme, M. A. & Murry, C. E. Heart regeneration. Nature 473, 326–35 (2011).

319. Sanganalmath, S. K. & Bolli, R. Cell therapy for heart failure: a comprehensive overview

of experimental and clinical studies, current challenges, and future directions. Circ. Res. 113, 810–34 (2013).

320. Lin, Z. & Pu, W. T. Strategies for Cardiac Regeneration and Repair. Sci. Transl. Med. 6,

239rv1 (2014).

321. Laflamme, M. A. et al. Cardiomyocytes derived from human embryonic stem cells in pro-

survival factors enhance function of infarcted rat hearts. Nat. Biotechnol. 25, 1015–24 (2007).

322. Madden, L. R. et al. Proangiogenic scaffolds as functional templates for cardiac tissue

engineering. Proc. Natl. Acad. Sci. U. S. A. 107, 15211–6 (2010).

323. Lin, Z. & Pu, W. T. Strategies for cardiac regeneration and repair. Sci. Transl. Med. 6,

239rv1 (2014).

324. Orlic, D. et al. Bone marrow cells regenerate infarcted myocardium. Nature 410, 701–705 (2001).

325. Matsuura, K. et al. Adult Cardiac Sca-1-positive Cells Differentiate into Beating Cardiomyocytes. J. Biol. Chem. 279, 11384–11391 (2004).

326. Cai, C.-L. et al. Isl1 Identifies a Cardiac Progenitor Population that Proliferates Prior to Differentiation and Contributes a Majority of Cells to the Heart. Dev. Cell 5, 877–889 (2003).

327. Messina, E. et al. Isolation and Expansion of Adult Cardiac Stem Cells From Human and Murine Heart. Circ. Res. 95, 911–921 (2004).

328. Magenta, A., Avitabile, D., Pompilio, G. & Capogrossi, M. C. c-kit-Positive Cardiac Progenitor Cells: The Heart of Stemness. Circ. Res. 112, 1202–1204 (2013).

329. Orlic, D. et al. Bone marrow cells regenerate infarcted myocardium. Nature 410, 701–5

(2001).

330. Balsam, L. B. et al. Haematopoietic stem cells adopt mature haematopoietic fates in

ischaemic myocardium. Nature 428, 668–73 (2004).

331. Nygren, J. M. et al. Bone marrow-derived hematopoietic cells generate cardiomyocytes at a low frequency through cell fusion, but not transdifferentiation. Nat. Med. 10, 494–501

(2004).

332. Janssens, S. et al. Autologous bone marrow-derived stem-cell transfer in patients with ST-

segment elevation myocardial infarction: double-blind, randomised controlled trial. Lancet (London, England) 367, 113–21 (2006).

333. Assmus, B. et al. Clinical outcome 2 years after intracoronary administration of bone

marrow-derived progenitor cells in acute myocardial infarction. Circ. Heart Fail. 3, 89–96 (2010).

Page 153: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

135

334. Vrtovec, B. et al. Effects of intracoronary CD34+ stem cell transplantation in nonischemic dilated cardiomyopathy patients: 5-year follow-up. Circ. Res. 112, 165–73 (2013).

335. Ieda, M. et al. Direct reprogramming of fibroblasts into functional cardiomyocytes by defined factors. Cell 142, 375–86 (2010).

336. Hong, K. U. et al. A highly sensitive and accurate method to quantify absolute numbers of c-kit+ cardiac stem cells following transplantation in mice. Basic Res. Cardiol. 108, 346 (2013).

337. Pasumarthi, K. B. S., Nakajima, H., Nakajima, H. O., Soonpaa, M. H. & Field, L. J. Targeted Expression of Cyclin D2 Results in Cardiomyocyte DNA Synthesis and Infarct

Regression in Transgenic Mice. Circ. Res. 96, 110–118 (2004).

338. Lin, Z. et al. Pi3kcb links Hippo-YAP and PI3K-AKT signaling pathways to promote cardiomyocyte proliferation and survival. Circ. Res. 116, 35–45 (2015).

339. von Gise, A. et al. YAP1, the nuclear target of Hippo signaling, stimulates heart growth through cardiomyocyte proliferation but not hypertrophy. Proc. Natl. Acad. Sci. 109,

2394–2399 (2012).

340. Zhou, J. An emerging role for Hippo-YAP signaling in cardiovascular development. J. Biomed. Res. 28, 251–4 (2014).

341. Engel, F. B. et al. p38 MAP kinase inhibition enables proliferation of adult mammalian cardiomyocytes. 1175–1187 (2005). doi:10.1101/gad.1306705.chaemic

342. Bersell, K. et al. Neuregulin1/ErbB4 signaling induces cardiomyocyte proliferation and repair of heart injury. Cell 138, 257–70 (2009).

343. Jabbour, A. et al. Parenteral administration of recombinant human neuregulin-1 to patients

with stable chronic heart failure produces favourable acute and chronic haemodynamic responses. Eur. J. Heart Fail. 13, 83–92 (2011).

344. Oulas, A. et al. A new microRNA target prediction tool identifies a novel interaction of a putative miRNA with CCND2. RNA Biol. 9, 1196–207 (2012).

345. Martinez-Sanchez, A. & Murphy, C. MicroRNA Target Identification—Experimental

Approaches. Biology (Basel). 2, 189–205 (2013).

346. Wani, S. & Cloonan, N. Profiling direct mRNA-microRNA interactions using synthetic

biotinylated microRNA-duplexes. bioRxiv 0–11 (2014). doi:10.1101/005439

347. Di Martino, M. T. et al. In Vitro and In Vivo Activity of a Novel Locked Nucleic Acid (LNA)-Inhibitor-miR-221 against Multiple Myeloma Cells. PLoS One 9, e89659 (2014).

348. Fleisch, V. C., Fraser, B. & Allison, W. T. Investigating regeneration and functional integration of CNS neurons: Lessons from zebrafish genetics and other fish species.

Biochim. Biophys. Acta - Mol. Basis Dis. 1812, 364–380 (2011).

349. Sherr, C. J. & McCormick, F. The RB and p53 pathways in cancer. Cancer Cell 2, 103–112 (2002).

Page 154: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

136

Copyright Acknowledgements

Permissions granted for use of external figures:

Figure 1.1 - Anatomy of the cardiac sarcomere. Harvey, P. A. & Leinwand, L. A. The cell biology of disease: cellular mechanisms of

cardiomyopathy. J. Cell Biol. 194, 355–65 (2011).

Permission to Use Copyrighted Material in a Master’s Thesis

Shanna Stanley-Hasnain

Today 2:13 PM

[email protected]

Sent Items

August 4, 2016 Dear Dr. Leinwand,

I am a University of Toronto graduate student at the Institute of Medical Science completing my Master’s thesis entitled “Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs”.

My thesis will be available in full text on the internet for reference, study and / or copy. Except in situations where a thesis is under embargo or restriction, the electronic version will be accessible through the U of T Libraries web pages, the Library’s web catalogue, and also through web search engines. I will also be granting Library and Archives Canada and ProQuest/UMI a non-exclusive license to reproduce, loan, distribute, or sell single copies of my thesis by any means and in any form or format.

I am requesting your permission to allow inclusion of the following material in my thesis: Figure 1. Anatomy of the cardiac sarcomere, from the published article Harvey, P. A. & Leinwand, L. A. The cell biology of disease: cellular mechanisms of cardiomyopathy. J. Cell Biol. 194, 355–65 (2011).

The material will be attributed through a citation. Please confirm in writing or by email that these arrangements meet with your approval.

Thank you very much for your time.

Sincerely, -- Shanna Stanley-Hasnain

Master's Candidate | Institute of Medical Science, Cardiovascular Sciences Collaborative Program University of Toronto

The University Health Network

Toronto General Research Institute

Billia Lab, MaRS Centre, Max Bell Research Center

(647)-985-0770

Address: 232 Gerrard Street East, Apartment D Toronto, ON, Canada M5A 2E8

Page 155: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

137

Reply all| Thu 4:31 PM

Shanna Stanley-Hasnain

<[email protected]>

I believe we assigned copyright to the Journal of Cell Biology. I always ask journal when I want to reproduce a figure from a published article, so I think you have to go through JCB as we no long own the rights.

Journal of Cell Biology / Rockefeller University Press Copyright Policy:

Noncommercial third-party reuse:

Third parties may reuse our content for noncommercial purposes without specific permission as long as they provide proper attribution (see citation preferences provided above). Within the first 6 months after publication, the creation of mirror sites is prohibited.

http://www.rupress.org/site/misc/permissions.xhtml

Page 156: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

138

Figure 1.2 - The main molecular proponents of myocardial ischemia reperfusion injury. Hausenloy, D. J. & Yellon, D. M. Myocardial ischemia-reperfusion injury: a neglected

therapeutic target. J. Clin. Invest. 123, 92–100 (2013).

Permission to Use Copyrighted Material in a Master’s Thesis Shanna Stanley-Hasnain

Today 2:23 PM

[email protected]

Sent Items

August 4, 2016

Dear Dr. Yellon, I am a University of Toronto graduate student at the Institute of Medical Science completing my Master’s thesis entitled “Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs”.

My thesis will be available in full text on the internet for reference, study and / or copy. Except in situations where a thesis is under embargo or restriction, the electronic version will be accessible through the U of T Libraries web pages, the Library’s web catalogue, and also through web search engines. I will also be granting Library and Archives Canada and ProQuest/UMI a non-exclusive license to reproduce, loan, distribute, or sell single copies of my thesis by any means and in any form or format.

I am requesting your permission to allow inclusion of the following material in my thesis: Figure 1. Schematic illustrating the main proponents of acute myocardial IRI, from the published article Hausenloy, D. J. & Yellon, D. M. Myocardial ischemia-reperfusion injury: a neglected therapeutic target. J. Clin. Invest. 123, 92–100 (2013).

The material will be attributed through a citation. Please confirm in writing or by email that these arrangements meet with your approval.

Thank you very much for your time. Sincerely, -- Shanna Stanley-Hasnain

Master's Candidate | Institute of Medical Science, Cardiovascular Sciences Collaborative Program

University of Toronto

The University Health Network

Toronto General Research Institute Billia Lab, MaRS Centre, Max Bell Research Center

(647)-985-0770

Address: 232 Gerrard Street East, Apartment D

Toronto, ON, Canada M5A 2E8

Page 157: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

139

Reply all| Today 2:02 AM

Shanna Stanley-Hasnain

Yellon, Derek <[email protected]>

Dear Shana Thank you for your email. Let me confirm that you have my full permission to include Figure 1 from our J.

Clin. Invest. 123, 92–100 (2013) paper for your thesis. I hope it all goes well. Best wishes Derek Professor Derek Yellon Professor of Molecular & Cellular Cardiology Director of the Hatter Cardiovascular Institute UCL, Programme Director (Cardiometabolic), NIHR UCLH Biomedical Research Centre. University College London & UCLH 67 Chenies Mews, London WC1E 6HX Tel: +44 203 447 9591 Fax: +44 203 447 9818 PA:[email protected]

Page 158: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

140

Page 159: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

141

Figure 1.3 - The major events of the cell cycle are regulated by transitions between CDK/cyclin complexes.

Rhind, N. & Russell, P. Signaling pathways that regulate cell division. Cold Spring Harb. Perspect. Biol. 4, a005942 (2012).

Permission to Use Copyrighted Material in a Master’s Thesis Shanna Stanley-Hansain

Today 2:31 PM

[email protected]

Sent Items

August 4, 2016

Dear Dr. Rhind,

I am a University of Toronto graduate student at the Institute of Medical Science completing my Master’s thesis entitled “Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs”.

My thesis will be available in full text on the internet for reference, study and / or copy. Except in situations where a thesis is under embargo or restriction, the electronic version will be accessible through the U of T Libraries web pages, the Library’s web catalogue, and also through web search engines. I will also be granting Library and Archives Canada and ProQuest/UMI a non-exclusive license to reproduce, loan, distribute, or sell single copies of my thesis by any means and in any form or format.

I am requesting your permission to allow inclusion of the following material in my thesis: Box 1. The main stages of mitosis, and Figure 1. The major events of the cell cycle, from the published article Rhind, N. & Russell, P. Signaling pathways that regulate cell division. Cold Spring Harb. Perspect. Biol. 4, a005942 (2012).

The material will be attributed through a citation. Please confirm in writing or by email that these arrangements meet with your approval.

Thank you very much for your time. Sincerely, -- Shanna Stanley-Hasnain

Master's Candidate | Institute of Medical Science, Cardiovascular Sciences Collaborative Program University of Toronto

The University Health Network

Toronto General Research Institute

Billia Lab, MaRS Centre, Max Bell Research Center

(647)-985-0770

Address:

232 Gerrard Street East, Apartment D Toronto, ON, Canada M5A 2E8

Page 160: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

142

Reply all| Today 3:02 PM

Shanna Stanley-Hasnain;

<[email protected]>

Dear Shanna,

You have my permission to use the requested figure.

Good luck with your thesis,

Nick

Proprietary Rights Notice for Cold Spring Harbor Perspectives in Biology Copyright © 2016, Cold Spring Harbor Perspectives in Biology by Cold Spring Harbor

1. Authorized users of Cold Spring Harbor Perspectives in Biology may view, reproduce, or store

copies of articles for the purposes of scholarly, research, educational, and individual use only. 2. Any copies reproduced from articles from Cold Spring Harbor Perspectives in Biology, whether in

whole or in part, must bear the original copyright notice. 3. The distribution of article(s) in whole or in part from Cold Spring Harbor Perspectives in

Biology is prohibited except with the prior written permission of Cold Spring Harbor Laboratory Press.

4. All authorized use of article(s) from Cold Spring Harbor Perspectives in Biology, in whole or in part, must include a full reference which shall include the article‘s bibliographic citation, including author attribution, date, article title, journal title, volume, and DOI.

5. For permission to copy or use any material published in Cold Spring Harbor Perspectives in Biology beyond the uses permitted by Section 107 and/or 108 of the U.S. Copyright Law and for reprints, click here .

Page 161: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

143

Figure 1.4 - Assembly of the pre-replicative and pre-loading complex. Aladjem, M. I. Replication in context: dynamic regulation of DNA replication patterns in

metazoans. Nat. Rev. Genet. 8, 588–600 (2007).

Permission to Use Copyrighted Material in a Master’s Thesis Shanna Stanley-Hasnain

Today 2:49 PM

[email protected]

Sent Items

August 4, 2016

Dear Dr. Aladjem

I am a University of Toronto graduate student at the Institute of Medical Science completing my Master’s thesis entitled “Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs”.

My thesis will be available in full text on the internet for reference, study and / or copy. Except in situations where a thesis is under embargo or restriction, the electronic version will be accessible through the U of T Libraries web pages, the Library’s web catalogue, and also through web search engines. I will also be granting Library and Archives Canada and ProQuest/UMI a non-exclusive license to reproduce, loan, distribute, or sell single copies of my thesis by any means and in any form or format.

I am requesting your permission to allow inclusion of the following material in my thesis: Figure 1. Assembly of pre-replication and pre-initiation protein complexes, from the published article Aladjem, M. I. Replication in context: dynamic regulation of DNA replication patterns in metazoans. Nat. Rev. Genet. 8, 588–600 (2007).

The material will be attributed through a citation. Please confirm in writing or by email that these arrangements meet with your approval.

Thank you very much for your time. Sincerely, -- Shanna Stanley-Hasnain

Master's Candidate | Institute of Medical Science, Cardiovascular Sciences Collaborative Program

University of Toronto

The University Health Network

Toronto General Research Institute Billia Lab, MaRS Centre, Max Bell Research Center

(647)-985-0770

Address: 232 Gerrard Street East, Apartment D

Toronto, ON, Canada M5A 2E8

Page 162: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

144

Reply all| Today 3:00 PM

Shanna Stanley-Hasnain;

Aladjem, Mirit (NIH/NCI) [E] <[email protected]>

Dear Ms. Stanley-Hasnain Thank you for contacting me. I am happy that you are using this figure, and I will be happy to grant you permission, however have you checked that you do not need permission from Nature Reviews Genetics? If the publisher states that you need permission from me, you have my permission. Best of luck wit your thesis. Sincerely, Mirit I. Aladjem, Ph.D. Senior Investigator Head, DNA replication Group, Developmental Therapeutics Branch Center for Cancer Research, NCI, NIH Bldg. 37, Rm 5056 37 Convent Dr. Bethesda, MD 20892-4255 Tel. 301-435-2848 Fax 301-402-0752

NATURE PUBLISHING GROUP LICENSE

TERMS AND CONDITIONS

Aug 04, 2016

This Agreement between The University of Toronto -- Shanna Stanley-Hasnain ("You") and Nature Publishing Group ("Nature Publishing Group") consists of your license details and the terms and conditions provided by Nature Publishing Group and Copyright

Clearance Center.

License Number 3922140503042

License date Aug 04, 2016

Licensed Content Publisher Nature Publishing Group

Licensed Content Publication Nature Reviews Genetics

Licensed Content Title Replication in context: dynamic regulation of DNA

replication patterns in metazoans

Licensed Content Author Mirit I. Aladjem

Licensed Content Date Aug 1, 2007

Page 163: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

145

Licensed Content Volume Number 8

Licensed Content Issue Number 8

Type of Use reuse in a dissertation / thesis

Requestor type academic/educational

Format print and electronic

Portion figures/tables/illustrations

Number of figures/tables/illustrations

1

High-res required no

Figures Figure 1. Assembly of pre-replication and pre-initiation

protein complexes Aladjem, M. I. Replication in context: dynamic regulation of DNA replication patterns in

metazoans. Nat. Rev. Genet. 8, 588–600 (2007).

Author of this NPG article no

Your reference number

Title of your thesis / dissertation Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs

Expected completion date Sep 2016

Estimated size (number of pages) 140

Requestor Location The University of Toronto

232 Gerrard Street East

Toronto, ON M5A 2E8

Canada

Attn: The University of Toronto

Billing Type Invoice

Billing Address The University of Toronto 232 Gerrard Street East

Toronto, ON M5A 2E8

Canada Attn: Shanna Stanley-Hasnain

Total 0.00 CAD

Terms and Conditions

Terms and Conditions for Permissions

Nature Publishing Group hereby grants you a non-exclusive license to reproduce this

material for this purpose, and for no other use,subject to the conditions below:

1. NPG warrants that it has, to the best of its knowledge, the rights to license reuse of this material. However, you should ensure that the material you are requesting is original to

Nature Publishing Group and does not carry the copyright of another entity (as credited in

Page 164: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

146

the published version). If the credit line on any part of the material you have requested

indicates that it was reprinted or adapted by NPG with permission from another source, then you should also seek permission from that source to reuse the material.

2. Permission granted free of charge for material in print is also usually granted for any

electronic version of that work, provided that the material is incidental to the work as a

whole and that the electronic version is essentially equivalent to, or substitutes for, the print version.Where print permission has been granted for a fee, separate permission

must be obtained for any additional, electronic re-use (unless, as in the case of a full paper, this has already been accounted for during your initial request in the calculation of

a print run).NB: In all cases, web-based use of full-text articles must be authorized separately through the 'Use on a Web Site' option when requesting permission.

3. Permission granted for a first edition does not apply to second and subsequent editions and for editions in other languages (except for signatories to the STM Permissions

Guidelines, or where the first edition permission was granted for free).

4. Nature Publishing Group's permission must be acknowledged next to the figure, table or

abstract in print. In electronic form, this acknowledgement must be visible at the same time as the figure/table/abstract, and must be hyperlinked to the journal's homepage.

5. The credit line should read:

Reprinted by permission from Macmillan Publishers Ltd: [JOURNAL NAME] (reference citation), copyright (year of publication)

For AOP papers, the credit line should read:

Reprinted by permission from Macmillan Publishers Ltd: [JOURNAL NAME], advance online publication, day month year (doi: 10.1038/sj.[JOURNAL ACRONYM].XXXXX)

Note: For republication from the British Journal of Cancer, the following credit

lines apply.

Reprinted by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK: [JOURNAL NAME] (reference citation), copyright (year of publication)For AOP papers, the

credit line should read: Reprinted by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK:

[JOURNAL NAME], advance online publication, day month year (doi: 10.1038/sj.[JOURNAL ACRONYM].XXXXX)

6. Adaptations of single figures do not require NPG approval. However, the adaptation should be credited as follows:

Adapted by permission from Macmillan Publishers Ltd: [JOURNAL NAME] (reference

citation), copyright (year of publication)

Note: For adaptation from the British Journal of Cancer, the following credit line

applies. Adapted by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK:

[JOURNAL NAME] (reference citation), copyright (year of publication)

7. Translations of 401 words up to a whole article require NPG approval. Please

visithttp://www.macmillanmedicalcommunications.com for more information.Translations of up to a 400 words do not require NPG approval. The translation should be credited as

follows:

Translated by permission from Macmillan Publishers Ltd: [JOURNAL NAME] (reference

citation), copyright (year of publication).

Note: For translation from the British Journal of Cancer, the following credit line applies.

Page 165: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

147

Translated by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK:

[JOURNAL NAME] (reference citation), copyright (year of publication)

We are certain that all parties will benefit from this agreement and wish you the best in the

use of this material. Thank you.

Special Terms:

v1.1

Questions? [email protected] or +1-855-239-3415 (toll free in the US) or

+1-978-646-2777.

Page 166: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

148

Figure 1.5 - The main stages of mitosis. Rhind, N. & Russell, P. Signaling pathways that regulate cell division. Cold Spring Harb.

Perspect. Biol. 4, a005942 (2012).

Permission to Use Copyrighted Material in a Master’s Thesis Shanna Stanley-Hansain

Today 2:31 PM

[email protected]

Sent Items

August 4, 2016

Dear Dr. Rhind,

I am a University of Toronto graduate student at the Institute of Medical Science completing my Master’s thesis entitled “Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs”.

My thesis will be available in full text on the internet for reference, study and / or copy. Except in situations where a thesis is under embargo or restriction, the electronic version will be accessible through the U of T Libraries web pages, the Library’s web catalogue, and also through web search engines. I will also be granting Library and Archives Canada and ProQuest/UMI a non-exclusive license to reproduce, loan, distribute, or sell single copies of my thesis by any means and in any form or format.

I am requesting your permission to allow inclusion of the following material in my thesis: Box 1. The main stages of mitosis, and Figure 1. The major events of the cell cycle, from the published article Rhind, N. & Russell, P. Signaling pathways that regulate cell division. Cold Spring Harb. Perspect. Biol. 4, a005942 (2012).

The material will be attributed through a citation. Please confirm in writing or by email that these arrangements meet with your approval.

Thank you very much for your time. Sincerely, -- Shanna Stanley-Hasnain

Master's Candidate | Institute of Medical Science, Cardiovascular Sciences Collaborative Program

University of Toronto The University Health Network

Toronto General Research Institute

Billia Lab, MaRS Centre, Max Bell Research Center

(647)-985-0770

Address:

232 Gerrard Street East, Apartment D Toronto, ON, Canada

M5A 2E8

Page 167: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

149

Reply all| Today 3:02 PM

Shanna Stanley-Hasnain;

<[email protected]>

Dear Shanna,

You have my permission to use the requested figure.

Good luck with your thesis,

Nick

Proprietary Rights Notice for Cold Spring Harbor Perspectives in Biology

Copyright © 2016, Cold Spring Harbor Perspectives in Biology by Cold Spring Harbor

6. Authorized users of Cold Spring Harbor Perspectives in Biology may view, reproduce, or store

copies of articles for the purposes of scholarly, research, educational, and individual use only. 7. Any copies reproduced from articles from Cold Spring Harbor Perspectives in Biology, whether in

whole or in part, must bear the original copyright notice. 8. The distribution of article(s) in whole or in part from Cold Spring Harbor Perspectives in

Biology is prohibited except with the prior written permission of Cold Spring Harbor Laboratory Press.

9. All authorized use of article(s) from Cold Spring Harbor Perspectives in Biology, in whole or in part, must include a full reference which shall include the article‘s bibliographic citation, including author attribution, date, article title, journal title, volume, and DOI.

10. For permission to copy or use any material published in Cold Spring Harbor Perspectives in Biology beyond the uses permitted by Section 107 and/or 108 of the U.S. Copyright Law and for reprints, click here .

Page 168: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

150

Figure 1.6 – Aurora B Localization During Mitosis and Cytokinesis. D’Avino, P. P. & Capalbo, L. New Auroras on the Roles of the Chromosomal Passenger

Complex in Cytokinesis: Implications for Cancer Therapies. Front. Oncol. 5, 221 (2015).

Permission to Use Copyrighted Material in a Master’s Thesis Shanna Stanley-Hasnain

Today 3:01 PM

[email protected]; [email protected]

Sent Items

August 4, 2016

Dear Dr. D'Avino and Dr. Capalbo

I am a University of Toronto graduate student at the Institute of Medical Science completing my Master’s thesis entitled “Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs”.

My thesis will be available in full text on the internet for reference, study and / or copy. Except in situations where a thesis is under embargo or restriction, the electronic version will be accessible through the U of T Libraries web pages, the Library’s web catalogue, and also through web search engines. I will also be granting Library and Archives Canada and ProQuest/UMI a non-exclusive license to reproduce, loan, distribute, or sell single copies of my thesis by any means and in any form or format.

I am requesting your permission to allow inclusion of the following material in my thesis: Figure 1. The CPC shows dynamic localization during mitosis and cytokinesis, from the published article D’Avino, P. P. & Capalbo, L. New Auroras on the Roles of the Chromosomal Passenger Complex in Cytokinesis: Implications for Cancer Therapies. Front. Oncol. 5, 221 (2015). The material will be attributed through a citation. Please confirm in writing or by email that these arrangements meet with your approval.

Thank you very much for your time. Sincerely, -- Shanna Stanley-Hasnain

Master's Candidate | Institute of Medical Science, Cardiovascular Sciences Collaborative Program

University of Toronto

The University Health Network

Toronto General Research Institute Billia Lab, MaRS Centre, Max Bell Research Center

(647)-985-0770

Address: 232 Gerrard Street East, Apartment D

Toronto, ON, Canada M5A 2E8

Page 169: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

151

Permission to Use Copyrighted Material in a Master’s Thesis Thu 11:57 PM

Shanna Stanley-Hasnain;

[email protected];

[email protected]

Sent Items

August 4, 2016

Dear Frontiers of Oncology Journal, I am a University of Toronto graduate student at the Institute of Medical Science completing my Master’s thesis entitled “Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs”. My thesis will be available in full text on the internet for reference, study and / or copy. Except in situations where a thesis is under embargo or restriction, the electronic version will be accessible through the U of T Libraries web pages, the Library’s web catalogue, and also through web search engines. I will also be granting Library and Archives Canada and ProQuest/UMI a non-exclusive license to reproduce, loan, distribute, or sell single copies of my thesis by any means and in any form or format. I am requesting your permission to allow inclusion of the following material in my thesis: Figure 1. The CPC shows dynamic localization during mitosis and cytokinesis, from the published article D’Avino, P. P. & Capalbo, L. New Auroras on the Roles of the Chromosomal Passenger Complex in Cytokinesis: Implications for Cancer Therapies. Front. Oncol. 5, 221 (2015). The material will be attributed through a citation. I was not able to request permission through the Copyright Clearance Center because the option for requestion to use a single figure in an academic thesis was not available. On the site it said "Copyright Clearance Center has upgraded its republication service. Unfortunately, the rightsholder (copyright owner) is not currently enrolled. Consider contacting the rightsholder directly for assistance." Please confirm in writing or by email that these arrangements meet with your approval. Alternatively, it would be greatly appreciated if you could direct me to the appropriate individual to approach for obtaining permission to use this figure in my thesis. Thank you very much for your time. Sincerely, -- Shanna Stanley-Hasnain Master's Candidate | Institute of Medical Science, Cardiovascular Sciences Collaborative Program

University of Toronto

The University Health Network

Toronto General Research Institute

Billia Lab, MaRS Centre, Max Bell Research Center

(416)-581-8476

Page 170: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

152

Reply all| Today 1:56 PM

Shanna Stanley-Hasnain

Pier Paolo D'Avino <[email protected]>

Dear Shanna,

Sorry for the delay.

We are happy for you to use our Figure in your thesis, providing that our paper is cited in

the Figure itself or in its legend.

Regards,

Paolo D'Avino and Luisa Capalbo

Reply all| Today 1:48 AM

Shanna Stanley-Hasnain

Frontiers Editorial Office <[email protected]>

Dear Mrs Stanley-Hasnain,

Thank you for your inquiry. All frontiers articles are published under the CC-BY 4.0 license,

which allows re-use provided that the original creators are correctly attributed.

As a courtesy, I would recommend that you inform the originators of the work, Pier Paolo

D’Avino, ([email protected];) and Luisa Capalbo, ([email protected]).

Best regards,

Faraz Alam, PhD

Ethics & Integrity Specialist

---

Gearóid Ó Faoleán, PhD Ethics & Integrity Manager

Frontiers

www.frontiersin.org EPFL Innovation Square, Building I Lausanne, Switzerland

Office T +44 79 34 46 47 49

Page 171: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

153

Figure 1.7 - The Canonical Pathway of miRNA Biogenesis. Winter, J., Jung, S., Keller, S., Gregory, R. I. & Diederichs, S. Many roads to maturity:

microRNA biogenesis pathways and their regulation. Nat. Cell Biol. 11, 228–234 (2009).

Permission to Use Copyrighted Material in a Master’s Thesis Shanna Stanley-Hasnain

Today 3:10 PM

[email protected]

Sent Items

August 4, 2016

Dear Dr. Diederichs

I am a University of Toronto graduate student at the Institute of Medical Science completing my Master’s thesis entitled “Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs”.

My thesis will be available in full text on the internet for reference, study and / or copy. Except in situations where a thesis is under embargo or restriction, the electronic version will be accessible through the U of T Libraries web pages, the Library’s web catalogue, and also through web search engines. I will also be granting Library and Archives Canada and ProQuest/UMI a non-exclusive license to reproduce, loan, distribute, or sell single copies of my thesis by any means and in any form or format.

I am requesting your permission to allow inclusion of the following material in my thesis: Figure 1. The ‘linear’ canonical pathway of microRNA processing, from the published article Winter, J., Jung, S., Keller, S., Gregory, R. I. & Diederichs, S. Many roads to maturity: microRNA biogenesis pathways and their regulation. Nat. Cell Biol. 11, 228–234 (2009). The material will be attributed through a citation. Please confirm in writing or by email that these arrangements meet with your approval.

Thank you very much for your time. Sincerely, -- Shanna Stanley-Hasnain

Master’s Candidate | Institute of Medical Science, Cardiovascular Sciences Collaborative Program

University of Toronto

The University Health Network

Toronto General Research Institute Billia Lab, MaRS Centre, Max Bell Research Center

(647)-985-0770

Address: 232 Gerrard Street East, Apartment D

Toronto, ON, Canada M5A 2E8

Page 172: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

154

Reply all| Today 3:49 AM

Shanna Stanley-Hasnain

Diederichs, Sven <[email protected]>

Dear Ms. Stanley-Hasnain, thank you for your email and your interest in our research. If you cite the source accordingly as well as for non-commercial purposes, I grant you permission to use our figure. Best regards, Sven Diederichs

________________________________________ Prof. Dr. Sven Diederichs University Professor - Cancer Research (University Hospital Freiburg) Division Head - RNA Biology & Cancer (German Cancer Research Center Heidelberg) Albert-Ludwigs-University Freiburg Medical Center - Department of Thoracic Surgery Division of Cancer Research Breisacher Str. 115, 79106 Freiburg, Germany German Cancer Research Center (DKFZ) Foundation under Public Law Division of RNA Biology & Cancer Im Neuenheimer Feld 280 (B150), 69120 Heidelberg, Germany Management Board: Prof. Dr. Michael Boutros (interim), Prof. Dr. Josef Puchta, VAT-ID No.: DE143293537 [email protected] www.diederichslab.org

NATURE PUBLISHING GROUP LICENSE

TERMS AND CONDITIONS

Aug 05, 2016

This Agreement between The University of Toronto -- Shanna Stanley-Hasnain ("You")

and Nature Publishing Group ("Nature Publishing Group") consists of your license details

Page 173: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

155

and the terms and conditions provided by Nature Publishing Group and Copyright

Clearance Center.

License Number 3922301450856

License date Aug 05, 2016

Licensed Content Publisher Nature Publishing Group

Licensed Content Publication Nature Cell Biology

Licensed Content Title Many roads to maturity: microRNA biogenesis pathways and their regulation

Licensed Content Author Julia Winter,Stephanie Jung,Sarina Keller,Richard I. GregoryandSven Diederichs

Licensed Content Date Mar 1, 2009

Licensed Content Volume Number 11

Licensed Content Issue Number 3

Type of Use reuse in a dissertation / thesis

Requestor type academic/educational

Format print and electronic

Portion figures/tables/illustrations

Number of figures/tables/illustrations 1

High-res required no

Figures Figure 1. The ‘linear’ canonical pathway of microRNA

processing.

Author of this NPG article no

Your reference number

Title of your thesis / dissertation Control of Cardiomyocyte Proliferation by

p53/Mdm2-Regulated microRNAs

Expected completion date Sep 2016

Estimated size (number of pages) 140

Requestor Location The University of Toronto 232 Gerrard Street East

Toronto, ON M5A 2E8

Canada Attn: The University of Toronto

Billing Type Invoice

Billing Address The University of Toronto

232 Gerrard Street East

Toronto, ON M5A 2E8

Page 174: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

156

Canada

Attn: Shanna Stanley-Hasnain

Total 0.00 CAD

Terms and Conditions

Terms and Conditions for Permissions

Nature Publishing Group hereby grants you a non-exclusive license to reproduce this

material for this purpose, and for no other use,subject to the conditions below:

8. NPG warrants that it has, to the best of its knowledge, the rights to license reuse of this material. However, you should ensure that the material you are requesting is original to

Nature Publishing Group and does not carry the copyright of another entity (as credited

in the published version). If the credit line on any part of the material you have requested indicates that it was reprinted or adapted by NPG with permission from

another source, then you should also seek permission from that source to reuse the material.

9. Permission granted free of charge for material in print is also usually granted for any

electronic version of that work, provided that the material is incidental to the work as a

whole and that the electronic version is essentially equivalent to, or substitutes for, the print version.Where print permission has been granted for a fee, separate permission

must be obtained for any additional, electronic re-use (unless, as in the case of a full paper, this has already been accounted for during your initial request in the calculation

of a print run).NB: In all cases, web-based use of full-text articles must be authorized

separately through the 'Use on a Web Site' option when requesting permission.

10. Permission granted for a first edition does not apply to second and subsequent editions and for editions in other languages (except for signatories to the STM Permissions

Guidelines, or where the first edition permission was granted for free).

11. Nature Publishing Group's permission must be acknowledged next to the figure, table or

abstract in print. In electronic form, this acknowledgement must be visible at the same time as the figure/table/abstract, and must be hyperlinked to the journal's homepage.

12. The credit line should read:

Reprinted by permission from Macmillan Publishers Ltd: [JOURNAL NAME] (reference

citation), copyright (year of publication) For AOP papers, the credit line should read:

Reprinted by permission from Macmillan Publishers Ltd: [JOURNAL NAME], advance online publication, day month year (doi: 10.1038/sj.[JOURNAL ACRONYM].XXXXX)

Note: For republication from the British Journal of Cancer, the following credit

lines apply.

Reprinted by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK: [JOURNAL NAME] (reference citation), copyright (year of publication)For AOP papers, the

credit line should read: Reprinted by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK:

[JOURNAL NAME], advance online publication, day month year (doi:

10.1038/sj.[JOURNAL ACRONYM].XXXXX)

13. Adaptations of single figures do not require NPG approval. However, the adaptation

should be credited as follows:

Page 175: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

157

Adapted by permission from Macmillan Publishers Ltd: [JOURNAL NAME] (reference

citation), copyright (year of publication)

Note: For adaptation from the British Journal of Cancer, the following credit line applies.

Adapted by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK:

[JOURNAL NAME] (reference citation), copyright (year of publication)

14. Translations of 401 words up to a whole article require NPG approval. Please visithttp://www.macmillanmedicalcommunications.com for more information.Translations

of up to a 400 words do not require NPG approval. The translation should be credited as follows:

Translated by permission from Macmillan Publishers Ltd: [JOURNAL NAME] (reference citation), copyright (year of publication).

Note: For translation from the British Journal of Cancer, the following credit line

applies.

Translated by permission from Macmillan Publishers Ltd on behalf o f Cancer Research UK: [JOURNAL NAME] (reference citation), copyright (year of publication)

We are certain that all parties will benefit from this agreement and wish you the best in the

use of this material. Thank you.

Special Terms:

v1.1

Questions? [email protected] or +1-855-239-3415 (toll free in the US) or +1-978-646-2777.

Page 176: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

158

Figure 1.8 - An overview to cell cycle control by microRNAs. Bueno, M. J. & Malumbres, M. MicroRNAs and the cell cycle. Biochim. Biophys. Acta - Mol.

Basis Dis. 1812, 592–601 (2011).

Permission to Use Copyrighted Material in a Master’s Thesis Shanna Stanley-Hasnain

Today 3:16 PM

[email protected]

Sent Items

August 4, 2016 Dear Dr. Malumbres,

I am a University of Toronto graduate student at the Institute of Medical Science completing my Master’s thesis entitled “Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs”.

My thesis will be available in full text on the internet for reference, study and / or copy. Except in situations where a thesis is under embargo or restriction, the electronic version will be accessible through the U of T Libraries web pages, the Library’s web catalogue, and also through web search engines. I will also be granting Library and Archives Canada and ProQuest/UMI a non-exclusive license to reproduce, loan, distribute, or sell single copies of my thesis by any means and in any form or format.

I am requesting your permission to allow inclusion of the following material in my thesis: Figure 1. An overview to cell cycle control by microRNAs, from the published article Bueno, M. J. & Malumbres, M. MicroRNAs and the cell cycle.Biochim. Biophys. Acta – Mol. Basis Dis. 1812, 592–601 (2011). The material will be attributed through a citation. Please confirm in writing or by email that these arrangements meet with your approval. Thank you very much for your time. Sincerely, -- Shanna Stanley-Hasnain

Master’s Candidate | Institute of Medical Science, Cardiovascular Sciences Collaborative Program

University of Toronto The University Health Network

Toronto General Research Institute

Billia Lab, MaRS Centre, Max Bell Research Center

(647)-985-0770

Address: 232 Gerrard Street East, Apartment D Toronto, ON, Canada

M5A 2E8

Page 177: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

159

Reply all| Today 3:17 PM

Shanna Stanley-Hasnain;

[email protected]

Dear Shanna, Sure, I am glad you are interested in this figure. Please use it at will! Best Marcos

ELSEVIER LICENSE

TERMS AND CONDITIONS

Aug 05, 2016

This Agreement between The University of Toronto -- Shanna Stanley-Hasnain ("You")

and Elsevier ("Elsevier") consists of your license details and the terms and conditions

provided by Elsevier and Copyright Clearance Center.

License Number 3922311100306

License date Aug 05, 2016

Licensed Content Publisher Elsevier

Licensed Content Publication Biochimica et Biophysica Acta (BBA) - Molecular Basis of

Disease

Licensed Content Title MicroRNAs and the cell cycle

Licensed Content Author María José Bueno,Marcos Malumbres

Licensed Content Date May 2011

Licensed Content Volume Number 1812

Licensed Content Issue Number 5

Licensed Content Pages 10

Start Page 592

End Page 601

Type of Use reuse in a thesis/dissertation

Intended publisher of new work other

Page 178: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

160

Portion figures/tables/illustrations

Number of figures/tables/illustrations 1

Format both print and electronic

Are you the author of this Elsevier article?

No

Will you be translating? No

Order reference number

Original figure numbers Figure 1. An overview to cell cycle control by microRNAs

Title of your thesis/dissertation Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs

Expected completion date Sep 2016

Estimated size (number of pages) 140

Elsevier VAT number GB 494 6272 12

Requestor Location The University of Toronto 232 Gerrard Street East

Toronto, ON M5A 2E8

Canada Attn: The University of Toronto

Total 0.00 CAD

Terms and Conditions

INTRODUCTION 1. The publisher for this copyrighted material is Elsevier. By clicking "accept" in connection with completing this licensing transaction, you agree that the following terms and conditions apply to this transaction (along with the Billing and Payment terms and conditions established by Copyright Clearance Center, Inc. ("CCC"), at the time that you opened your Rightslink account and that are available at any time at http://myaccount.copyright.com).

GENERAL TERMS 2. Elsevier hereby grants you permission to reproduce the aforementioned material subject to the terms and conditions indicated. 3. Acknowledgement: If any part of the material to be used (for example, figures) has appeared in our publication with credit or acknowledgement to another source, permission must also be sought from that source. If such permission is not obtained then that material may not be included in your publication/copies. Suitable acknowledgement to the source must be made, either as a footnote or in a reference list at the end of your publication, as follows: "Reprinted from Publication title, Vol /edition number, Author(s), Title of article / title of chapter, Pages No., Copyright (Year), with permission from Elsevier [OR APPLICABLE SOCIETY COPYRIGHT OWNER]." Also Lancet special credit - "Reprinted from The Lancet, Vol. number, Author(s), Title of article, Pages No., Copyright (Year), with permission from Elsevier." 4. Reproduction of this material is confined to the purpose and/or media for which permission is hereby given. 5. Altering/Modifying Material: Not Permitted. However figures and illustrations may be altered/adapted minimally to serve your work. Any other abbreviations, additions, deletions

Page 179: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

161

and/or any other alterations shall be made only with prior written authorization of Elsevier Ltd. (Please contact Elsevier at [email protected]) 6. If the permission fee for the requested use of our material is waived in this instance, please be advised that your future requests for Elsevier materials may attract a fee. 7. Reservation of Rights: Publisher reserves all rights not specifically granted in the combination of (i) the license details provided by you and accepted in the course of this licensing transaction, (ii) these terms and conditions and (iii) CCC's Billing and Payment terms and conditions. 8. License Contingent Upon Payment: While you may exercise the rights licensed immediately upon issuance of the license at the end of the licensing process for the transaction, provided that you have disclosed complete and accurate details of your proposed use, no license is finally effective unless and until full payment is received from you (either by publisher or by CCC) as provided in CCC's Billing and Payment terms and conditions. If full payment is not received on a timely basis, then any license preliminarily granted shall be deemed automatically revoked and shall be void as if never granted. Further, in the event that you breach any of these terms and conditions or any of CCC's Billing and Payment terms and conditions, the license is automatically revoked and shall be void as if never granted. Use of materials as described in a revoked license, as well as any use of the materials beyond the scope of an unrevoked license, may constitute copyright infringement and publisher reserves the right to take any and all action to protect its copyright in the materials. 9. Warranties: Publisher makes no representations or warranties with respect to the licensed material. 10. Indemnity: You hereby indemnify and agree to hold harmless publisher and CCC, and their respective officers, directors, employees and agents, from and against any and all claims arising out of your use of the licensed material other than as specifically authorized pursuant to this license. 11. No Transfer of License: This license is personal to you and may not be sublicensed, assigned, or transferred by you to any other person without publisher's written permission. 12. No Amendment Except in Writing: This license may not be amended except in a writing signed by both parties (or, in the case of publisher, by CCC on publisher's behalf). 13. Objection to Contrary Terms: Publisher hereby objects to any terms contained in any purchase order, acknowledgment, check endorsement or other writing prepared by you, which terms are inconsistent with these terms and conditions or CCC's Billing and Payment terms and conditions. These terms and conditions, together with CCC's Billing and Payment terms and conditions (which are incorporated herein), comprise the entire agreement between you and publisher (and CCC) concerning this licensing transaction. In the event of any conflict between your obligations established by these terms and conditions and those established by CCC's Billing and Payment terms and conditions, these terms and conditions shall control. 14. Revocation: Elsevier or Copyright Clearance Center may deny the permissions described in this License at their sole discretion, for any reason or no reason, with a full refund payable to you. Notice of such denial will be made using the contact information provided by you. Failure to receive such notice will not alter or invalidate the denial. In no event will Elsevier or Copyright Clearance Center be responsible or liable for any costs, expenses or damage incurred by you as a result of a denial of your permission request, other than a refund of the amount(s) paid by you to Elsevier and/or Copyright Clearance Center for denied permissions.

LIMITED LICENSE The following terms and conditions apply only to specific license types: 15. Translation: This permission is granted for non-exclusive world English rights only unless your license was granted for translation rights. If you licensed translation rights you may only translate this content into the languages you requested. A professional translator must perform all translations and reproduce the content word for word preserving the integrity of the article.

Page 180: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

162

16. Posting licensed content on any Website: The following terms and conditions apply as follows: Licensing material from an Elsevier journal: All content posted to the web site must maintain the copyright information line on the bottom of each image; A hyper-text must be included to the Homepage of the journal from which you are licensing athttp://www.sciencedirect.com/science/journal/xxxxx or the Elsevier homepage for books athttp://www.elsevier.com; Central Storage: This license does not include permission for a scanned version of the material to be stored in a central repository such as that provided by Heron/XanEdu. Licensing material from an Elsevier book: A hyper-text link must be included to the Elsevier homepage at http://www.elsevier.com . All content posted to the web site must maintain the copyright information line on the bottom of each image. Posting licensed content on Electronic reserve: In addition to the above the following clauses are applicable: The web site must be password-protected and made available only to bona fide students registered on a relevant course. This permission is granted for 1 year only. You may obtain a new license for future website posting. 17. For journal authors: the following clauses are applicable in addition to the above:

Preprints: A preprint is an author's own write-up of research results and analysis, it has not been peer-reviewed, nor has it had any other value added to it by a publisher (such as formatting, copyright, technical enhancement etc.). Authors can share their preprints anywhere at any time. Preprints should not be added to or enhanced in any way in order to appear more like, or to substitute for, the final versions of articles however authors can update their preprints on arXiv or RePEc with their Accepted Author Manuscript (see below). If accepted for publication, we encourage authors to link from the preprint to their formal publication via its DOI. Millions of researchers have access to the formal publications on ScienceDirect, and so links will help users to find, access, cite and use the best available version. Please note that Cell Press, The Lancet and some society-owned have different preprint policies. Information on these policies is available on the journal homepage. Accepted Author Manuscripts: An accepted author manuscript is the manuscript of an article that has been accepted for publication and which typically includes author-incorporated changes suggested during submission, peer review and editor-author communications. Authors can share their accepted author manuscript:

- immediately o via their non-commercial person homepage or blog o by updating a preprint in arXiv or RePEc with the accepted manuscript o via their research institute or institutional repository for internal

institutional uses or as part of an invitation-only research collaboration work-group

o directly by providing copies to their students or to research collaborators for their personal use

o for private scholarly sharing as part of an invitation-only work group on commercial sites with which Elsevier has an agreement

- after the embargo period o via non-commercial hosting platforms such as their institutional

repository o via commercial sites with which Elsevier has an agreement

In all cases accepted manuscripts should: - link to the formal publication via its DOI

Page 181: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

163

- bear a CC-BY-NC-ND license - this is easy to do - if aggregated with other manuscripts, for example in a repository or other site, be

shared in alignment with our hosting policy not be added to or enhanced in any way to appear more like, or to substitute for, the published journal article.

Published journal article (JPA): A published journal article (PJA) is the definitive final record of published research that appears or will appear in the journal and embodies all value-adding publishing activities including peer review co-ordination, copy-editing, formatting, (if relevant) pagination and online enrichment. Policies for sharing publishing journal articles differ for subscription and gold open access articles: Subscription Articles: If you are an author, please share a link to your article rather than the full-text. Millions of researchers have access to the formal publications on ScienceDirect, and so links will help your users to find, access, cite, and use the best available version. Theses and dissertations which contain embedded PJAs as part of the formal submission can be posted publicly by the awarding institution with DOI links back to the formal publications on ScienceDirect. If you are affiliated with a library that subscribes to ScienceDirect you have additional private sharing rights for others' research accessed under that agreement. This includes use for classroom teaching and internal training at the institution (including use in course packs and courseware programs), and inclusion of the article for grant funding purposes. Gold Open Access Articles: May be shared according to the author-selected end-user license and should contain a CrossMark logo, the end user license, and a DOI link to the formal publication on ScienceDirect. Please refer to Elsevier's posting policy for further information. 18. For book authors the following clauses are applicable in addition to the above: Authors are permitted to place a brief summary of their work online only. You are not allowed to download and post the published electronic version of your chapter, nor may you scan the printed edition to create an electronic version. Posting to a repository: Authors are permitted to post a summary of their chapter only in their institution's repository. 19. Thesis/Dissertation: If your license is for use in a thesis/dissertation your thesis may be submitted to your institution in either print or electronic form. Should your thesis be published commercially, please reapply for permission. These requirements include permission for the Library and Archives of Canada to supply single copies, on demand, of the complete thesis and include permission for Proquest/UMI to supply single copies, on demand, of the complete thesis. Should your thesis be published commercially, please reapply for permission. Theses and dissertations which contain embedded PJAs as part of the formal submission can be posted publicly by the awarding institution with DOI links back to the formal publications on ScienceDirect.

Elsevier Open Access Terms and Conditions You can publish open access with Elsevier in hundreds of open access journals or in nearly 2000 established subscription journals that support open access publishing. Permitted third party re-use of these open access articles is defined by the author's choice of Creative Commons user license. See our open access license policy for more information.

Terms & Conditions applicable to all Open Access articles published with Elsevier: Any reuse of the article must not represent the author as endorsing the adaptation of the article nor should the article be modified in such a way as to damage the author's honour or reputation. If any changes have been made, such changes must be clearly indicated. The author(s) must be appropriately credited and we ask that you include the end user license and a DOI link to the formal publication on ScienceDirect.

Page 182: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

164

If any part of the material to be used (for example, figures) has appeared in our publication with credit or acknowledgement to another source it is the responsibility of the user to ensure their reuse complies with the terms and conditions determined by the rights holder.

Additional Terms & Conditions applicable to each Creative Commons user license: CC BY: The CC-BY license allows users to copy, to create extracts, abstracts and new works from the Article, to alter and revise the Article and to make commercial use of the Article (including reuse and/or resale of the Article by commercial entities), provided the user gives appropriate credit (with a link to the formal publication through the relevant DOI), provides a link to the license, indicates if changes were made and the licensor is not represented as endorsing the use made of the work. The full details of the license are available athttp://creativecommons.org/licenses/by/4.0. CC BY NC SA: The CC BY-NC-SA license allows users to copy, to create extracts, abstracts and new works from the Article, to alter and revise the Article, provided this is not done for commercial purposes, and that the user gives appropriate credit (with a link to the formal publication through the relevant DOI), provides a link to the license, indicates if changes were made and the licensor is not represented as endorsing the use made of the work. Further, any new works must be made available on the same conditions. The full details of the license are available at http://creativecommons.org/licenses/by-nc-sa/4.0. CC BY NC ND: The CC BY-NC-ND license allows users to copy and distribute the Article, provided this is not done for commercial purposes and further does not permit distribution of the Article if it is changed or edited in any way, and provided the user gives appropriate credit (with a link to the formal publication through the relevant DOI), provides a link to the license, and that the licensor is not represented as endorsing the use made of the work. The full details of the license are available at http://creativecommons.org/licenses/by-nc-nd/4.0. Any commercial reuse of Open Access articles published with a CC BY NC SA or CC BY NC ND license requires permission from Elsevier and will be subject to a fee. Commercial reuse includes:

- Associating advertising with the full text of the Article - Charging fees for document delivery or access - Article aggregation - Systematic distribution via e-mail lists or share buttons

Posting or linking by commercial companies for use by customers of those companies. 20. Other Conditions : v1.8

Questions? [email protected] or +1-855-239-3415 (toll free in the US) or +1-978-

646-2777.

Page 183: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

165

Figure 1.9 - Regulation of the cell cycle by p53-induced miRNAs. Hermeking, H. MicroRNAs in the p53 network: micromanagement of tumour suppression. Nat.

Rev. Cancer 12, 613–626 (2012).

Permission to Use Copyrighted Material in a Master’s Thesis Shanna Stanley-Hasnain

Today 3:33 PM

[email protected]

Sent Items

August 4, 2016 Dear Dr. Hermeking,

I am a University of Toronto graduate student at the Institute of Medical Science completing my Master’s thesis entitled “Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs”.

My thesis will be available in full text on the internet for reference, study and / or copy. Except in situations where a thesis is under embargo or restriction, the electronic version will be accessible through the U of T Libraries web pages, the Library’s web catalogue, and also through web search engines. I will also be granting Library and Archives Canada and ProQuest/UMI a non-exclusive license to reproduce, loan, distribute, or sell single copies of my thesis by any means and in any form or format.

I am requesting your permission to allow inclusion of the following material in my thesis: Figure 2. Regulation of the cell cycle, metastasis and stemness by p53-induced microRNAs, from the published article Hermeking, H. MicroRNAs in the p53 network: micromanagement of tumour suppression. Nat. Rev. Cancer 12, 613–626 (2012). The material will be attributed through a citation. Please confirm in writing or by email that these arrangements meet with your approval. Thank you very much for your time. Sincerely, -- Shanna Stanley-Hasnain

Master’s Candidate | Institute of Medical Science, Cardiovascular Sciences Collaborative Program University of Toronto

The University Health Network

Toronto General Research Institute

Billia Lab, MaRS Centre, Max Bell Research Center

(647)-985-0770

Address: 232 Gerrard Street East, Apartment D Toronto, ON, Canada M5A 2E8

Page 184: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

166

Reply all|

Today 4:56 AM

Shanna Stanley-Hasnain

Hermeking, Heiko Prof.Dr. <[email protected]>

Ok with me.

Prof. Heiko Hermeking

Experimental and Molecular Pathology

Institute of Pathology Ludwig-Maximilians-Universität München

Thalkirchner Str. 36

D-80337 Munich, Germany

Tel.: ++ 49-89-2180-73685

Fax: ++ 49-89-2180-73697

Mobil: 0151-22377825

e-mail: [email protected]

http://www.pathologie.med.uni-muenchen.de/020wissenschaft/009ag_hermeking/engl_ag_hermeking/index.html

NATURE PUBLISHING GROUP LICENSE

TERMS AND CONDITIONS

Aug 05, 2016

This Agreement between The University of Toronto -- Shanna Stanley-Hasnain ("You")

and Nature Publishing Group ("Nature Publishing Group") consists of your license details

and the terms and conditions provided by Nature Publishing Group and Copyright

Clearance Center.

License Number 3922311500892

License date Aug 05, 2016

Licensed Content Publisher Nature Publishing Group

Licensed Content Publication Nature Reviews Cancer

Page 185: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

167

Licensed Content Title MicroRNAs in the p53 network: micromanagement

of tumour suppression

Licensed Content Author Heiko Hermeking

Licensed Content Date Aug 17, 2012

Licensed Content Volume Number 12

Licensed Content Issue Number 9

Type of Use reuse in a dissertation / thesis

Requestor type academic/educational

Format print and electronic

Portion figures/tables/illustrations

Number of figures/tables/illustrations 1

High-res required no

Figures Figure 2. Regulation of the cell cycle, metastasis and

stemness by p53 induced microRNAs

Author of this NPG article no

Your reference number

Title of your thesis / dissertation Control of Cardiomyocyte Proliferation by

p53/Mdm2-Regulated microRNAs

Expected completion date Sep 2016

Estimated size (number of pages) 140

Requestor Location The University of Toronto 232 Gerrard Street East

Toronto, ON M5A 2E8

Canada Attn: The University of Toronto

Billing Type Invoice

Billing Address The University of Toronto

232 Gerrard Street East

Toronto, ON M5A 2E8 Canada

Attn: Shanna Stanley-Hasnain

Total 0.00 CAD

Terms and Conditions

Terms and Conditions for Permissions

Nature Publishing Group hereby grants you a non-exclusive license to reproduce this

material for this purpose, and for no other use,subject to the conditions below:

Page 186: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

168

1. NPG warrants that it has, to the best of its knowledge, the rights to license reuse of this material. However, you should ensure that the material you are requesting is original to

Nature Publishing Group and does not carry the copyright of another entity (as credited in the published version). If the credit line on any part of the material you have

requested indicates that it was reprinted or adapted by NPG with permission from

another source, then you should also seek permission from that source to reuse the material.

2. Permission granted free of charge for material in print is also usually granted for any

electronic version of that work, provided that the material is incidental to the work as a

whole and that the electronic version is essentially equivalent to, or substitutes for, the print version.Where print permission has been granted for a fee, separate permission

must be obtained for any additional, electronic re-use (unless, as in the case of a full paper, this has already been accounted for during your initial request in the calculation of

a print run).NB: In all cases, web-based use of full-text articles must be authorized separately through the 'Use on a Web Site' option when requesting permission.

3. Permission granted for a first edition does not apply to second and subsequent editions and for editions in other languages (except for signatories to the STM Permissions

Guidelines, or where the first edition permission was granted for free).

4. Nature Publishing Group's permission must be acknowledged next to the figure, table or

abstract in print. In electronic form, this acknowledgement must be visible at the same time as the figure/table/abstract, and must be hyperlinked to the journal's homepage.

5. The credit line should read:

Reprinted by permission from Macmillan Publishers Ltd: [JOURNAL NAME] (reference citation), copyright (year of publication)

For AOP papers, the credit line should read:

Reprinted by permission from Macmillan Publishers Ltd: [JOURNAL NAME], advance online publication, day month year (doi: 10.1038/sj.[JOURNAL ACRONYM].XXXXX)

Note: For republication from the British Journal of Cancer, the following credit

lines apply.

Reprinted by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK: [JOURNAL NAME] (reference citation), copyright (year of publication)For AOP papers, the

credit line should read: Reprinted by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK:

[JOURNAL NAME], advance online publication, day month year (doi: 10.1038/sj.[JOURNAL ACRONYM].XXXXX)

6. Adaptations of single figures do not require NPG approval. However, the adaptation

should be credited as follows:

Adapted by permission from Macmillan Publishers Ltd: [JOURNAL NAME] (reference

citation), copyright (year of publication)

Note: For adaptation from the British Journal of Cancer, the following credit line applies.

Adapted by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK: [JOURNAL NAME] (reference citation), copyright (year of publication)

7. Translations of 401 words up to a whole article require NPG approval. Please visithttp://www.macmillanmedicalcommunications.com for more information.Translations

of up to a 400 words do not require NPG approval. The translation should be credited as follows:

Page 187: Control of Cardiomyocyte Proliferation by …...ii Control of Cardiomyocyte Proliferation by p53/Mdm2-Regulated microRNAs Shanna Stanley-Hasnain Masters of Science – Medical Science

169

Translated by permission from Macmillan Publishers Ltd: [JOURNAL NAME] (reference citation), copyright (year of publication).

Note: For translation from the British Journal of Cancer, the following credit line

applies.

Translated by permission from Macmillan Publishers Ltd on behalf of Cancer Research UK: [JOURNAL NAME] (reference citation), copyright (year of publication)

We are certain that all parties will benefit from this agreement and wish you the best in the

use of this material. Thank you.

Special Terms:

v1.1

Questions? [email protected] or +1-855-239-3415 (toll free in the US) or +1-978-646-2777.