the role of polarity protein angiomotin (amot) in the ...€¦ · abby patricia farrell master of...

171
The Role of Polarity Protein Angiomotin (AMOT) in the Human Placenta by Abby Patricia Farrell A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Science University of Toronto © Copyright by Abby Farrell 2018

Upload: others

Post on 18-Jul-2020

4 views

Category:

Documents


0 download

TRANSCRIPT

  • The Role of Polarity Protein Angiomotin (AMOT) in the

    Human Placenta

    by

    Abby Patricia Farrell

    A thesis submitted in conformity with the requirements

    for the degree of Master of Science

    Institute of Medical Science

    University of Toronto

    © Copyright by Abby Farrell 2018

  • ii

    The Role of Polarity Protein Angiomotin (AMOT) in the Human

    Placenta

    Abby Patricia Farrell

    Master of Science

    Institute of Medical Science

    University of Toronto

    2018

    Abstract

    Angiomotin (AMOT) is a scaffolding protein involved in cell polarity regulation, cell migration

    and early embryo lineage differentiation, yet its biological significance in the human placenta

    remains unknown. I hypothesized that AMOT controls trophoblast cell polarity and migration, and

    is further regulated by transforming growth factor beta (TGF) signalling and upstream oxygen

    tension. AMOT localization to extravillous trophoblast (EVT) cells, corroborated by AMOT 80

    overexpression increasing JEG3 cell migration rate, supports a role for AMOT in EVT migration.

    TGF1/3 treatment decreased AMOT protein levels and redistributed AMOT from the tight-

    junction to cytoplasmic F-actin in JEG3 cells. TGF1/3 also prompted a novel association between

    AMOT 80 and Partitioning Defective Protein-6. Similarly, low oxygen exposure negatively

    regulated AMOT levels and localization. Furthermore, Jumonji C Domain Containing Protein-6

    (JMJD6), an oxygen sensor, was discovered to positively regulate AMOT via lysyl hydroxylation.

    Finally, AMOT levels were found markedly reduced in preeclampsia, a disease characterized by

    aberrant TGF signalling and chronic hypoxia. In conclusion, this study reveals AMOT is a

    mediator of TGF and oxygen signalling to regulate trophoblast migration in the human placenta.

  • iii

    Table of Contents

    Table of Contents ......................................................................................................................... iii

    Acknowledgments ........................................................................................................................ vi

    List of Abbreviations .................................................................................................................. vii

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

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

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

    Introduction .................................................................................................................................1

    1.1 Angiomotin (AMOT) .........................................................................................................2

    1.1.1 Discovery and Structure ...........................................................................................2

    1.1.2 Role of AMOT in Hippo Pathway Signalling..........................................................6

    1.1.3 Role of AMOT in Cell Migration ............................................................................8

    1.2 Cell Polarity Regulation ....................................................................................................9

    1.2.1 Tight Junctions .........................................................................................................9

    1.2.2 Polarity Protein Complexes ...................................................................................10

    1.2.3 Role of Cell Polarity in Proliferation, Migration and Invasion .............................11

    1.2.4 AMOT as a Novel Regulator of Cell Polarity .......................................................12

    1.3 Human Placenta Development........................................................................................16

    1.3.1 Trophoblast Differentiation ...................................................................................17

    1.3.2 TGF signalling pathways .....................................................................................22

    1.3.3 Role of TGFβ Signalling in Trophoblast Differentiation ......................................26

    1.4 Preeclampsia .....................................................................................................................29

    1.4.1 Altered Trophoblast Differentiation in preeclampsia ............................................30

    1.4.2 Impairments in oxygen sensing in preeclampsia ...................................................32

    1.4.3 JMJD6: a novel oxygen sensor and regulator in the placenta ................................33

    1.5 Rationale, Hypothesis and Objectives ...............................................................................37

    Chapter 2 Materials and Methods..............................................................................................39

    Materials and Methods ..............................................................................................................39

    2.1 Human Placenta Tissue Collection ....................................................................................39

    2.2 JEG3 Human Choriocarcinoma Cell Culture ....................................................................41

    2.3 In vitro treatments in JEG3 cells ........................................................................................42

    2.3.1 Transforming Growth Factor- (TGF) Treatment ...............................................42

    2.3.2 SB-431542 Treatment in JEG3 cells......................................................................42

  • iv

    2.3.3 Minoxidil Treatment ..............................................................................................43

    2.3.4 Low Oxygen (3% O2) Treatment in JEG3 cells.....................................................43

    2.4 Plasmid DNA Constructs for Overexpression Studies ......................................................43

    2.4.1 Plasmid DNA Transfection ....................................................................................45

    2.5 siRNA Transfections ..........................................................................................................45

    2.6 Wound Healing Assay .......................................................................................................46

    2.7 Time-Lapse Live Cell Imaging ..........................................................................................46

    2.8 Western Blot Analysis .......................................................................................................47

    2.9 Antibodies ..........................................................................................................................49

    2.10 Immunoprecipitation (IP)...................................................................................................50

    2.11 Immunohistochemistry (IHC) ............................................................................................51

    2.12 Immunofluorescence (IF) ...................................................................................................53

    2.13 Proximity Ligation Assay ..................................................................................................56

    2.14 RNA Isolation, cDNA conversion and Quantitative-PCR .................................................57

    2.15 In vitro JMJD6 Hydroxylation Reaction............................................................................58

    2.16 MALDI-TOF Mass Spectrometry......................................................................................59

    2.17 Statistical analysis ..............................................................................................................60

    Chapter 3 Results .........................................................................................................................61

    Results .......................................................................................................................................61

    3.1 AMOT exhibits distinct temporal and spatial expression patterns during human placenta development.........................................................................................................61

    3.2 AMOT is regulated by TGF signalling pathway .............................................................67

    3.2.1 AMOT resides at tight junction, cytoplasm and protruding edge of JEG3 cells ...67

    3.2.2 TGF1/3 ligand treatment reduces AMOT 130 and 80 protein levels ..................68

    3.2.3 TGF1/3 treatment promotes subcellular redistribution of AMOT ......................68

    3.3 AMOT 130 is regulated by Smad-dependent TGF pathway ...........................................74

    3.4 TGFβ promotes AMOT redistribution in migrating cells ..................................................78

    3.5 AMOT 80 promotes JEG3 cell migration ..........................................................................79

    3.6 Novel AMOT/Par6 interaction and its regulation by TGFβ ..............................................84

    3.7 PDZ and coiled-coil binding domains are important for AMOT and Par6 interaction .....89

    3.8 AMOT promotes dissolution of RhoA at the tight junction ..............................................93

    3.9 AMOT protein levels and distribution is disrupted in preeclampsia .................................96

    3.9.1 AMOT and Par6 interaction is impaired in preeclampsia .....................................97

  • v

    3.10 AMOT protein levels and localization is disrupted in low oxygen .................................101

    3.11 Oxygen sensor JMJD6 positively regulates AMOT ........................................................104

    3.11.1 AMOT is subject to lysyl hydroxylation by JMJD6 ............................................104

    Chapter 4 Discussion .................................................................................................................112

    Discussion and Conclusions ....................................................................................................112

    4.1 General Discussion ..........................................................................................................112

    4.2 Conclusions ......................................................................................................................126

    Chapter 5 Future Directions .....................................................................................................129

    Future Directions .....................................................................................................................129

    5.1 Using in vivo villous explants to assess AMOT’s role in trophoblast cell differentiation ...................................................................................................................130

    5.2 Deciphering the role of AMOT in placental mesenchymal cells .....................................132

    5.3 Establishing a role for the AMOT/TAZ axis in trophoblast cell differentiation .............137

    5.4 Investigating lysosomal degradation of AMOT ..............................................................141

    References ...................................................................................................................................143

    Appendix - Statement of Contributions ...................................................................................158

  • vi

    Acknowledgments

    The academic and personal growth I have experienced while working towards my Master’s degree

    is beyond what I could have imagined, and there are many people I would like to thank for their

    support along the way. First and foremost, I want to express my sincerest gratitude to my

    supervisor, Dr. Isabella Caniggia, for her steadfast commitment and unwavering guidance over

    these past two years. Thank you for providing me with an innovative project and incredible

    opportunities that have inspired me as a young scientist and aspiring clinician. Your passion for

    science, life and adventure is admirable and I feel incredibly fortunate to have been your student.

    I would also like to thank my PAC members Dr. Kellie Murphy, Dr. Brian Cox and Dr. Theodore

    Brown for their attention to detail and guidance throughout the duration of my project.

    In my short time with the Caniggia Lab, I have had the absolute pleasure of working alongside

    driven, brilliant and energetic people. Joelcio, Julien, Tyler, Leo, Andrea and Tingting- you all, in

    one form or another, have made invaluable contributions to my project and enriched my graduate

    student experience. Thank you for always supporting and caring about me, and of course for the

    many, many laughs we all shared together. To Taylor, thank you for being my best friend this past

    year, and empathizing with me every step of this journey. I am grateful to have found a life-long

    friend in you. To Sruthi, I can confidently say that my project would not have been as successful

    if it weren’t for you. Your daily academic and personal support has been irreplaceable and I can’t

    thank you enough for being such a great friend and mentor to me.

    To my extraordinary friends, Jessie and Nicole, thank you for always checking up on me, asking

    me about my research and jumping through hoops to find time to see me.

    To my boyfriend Brandon, thank you for your unconditional love and patience day in and day out.

    I am fortunate enough to celebrate milestones, such as this one, with someone as kind-hearted and

    sweet as you. Thank you for being so wonderful to me, always.

    Lastly, I would like to thank my family, who collectively have shaped the young woman I am

    today. To my sister Jenna, living in Toronto with you this past year has been so much fun. Being

    able to actively support each other’s goals has been profound, and I will always look back fondly

    on this time we have spent together. To my guardian angel, Dad- you always told me I could

    achieve anything I set my mind to. Thank you for instilling perseverance in me to follow my

    dreams, even when it gets tough. I miss you, always. Finally, to my Mom- there will never be

    enough words to describe what you mean to me. Thank you for doing everything in your power to

    ensure I am healthy, happy and successful. But most importantly, thank you for constantly

    grounding me and reminding me what is truly important in life.

  • vii

    List of Abbreviations

    AMOT Angiomotin

    ABC complex Activin-biotin complex

    ACOG American College of Obstetricians and Gynecologists

    Alk1-7 Activin receptor-like kinase 1-7

    AMOT 130 Angiomotin 130kDa isoform

    AMOT 80 Angiomotin 80kDa isoform

    AMOTL1 Angiomotin-Like 1

    AMOTL2 Angiomotin-Like 2

    aPKC atypical protein kinase

    BSA Bovine serum albumin

    Cdc42 Cell division control protein 42

    cDNA complementary DNA

    ChIP Chromatin Immunoprecipitation

    cm centimeter

    Co-IP Co-immunoprecipitation

    Coll-1 Collagen type 1

    Crb Crumbs

    CT Cytotrophoblast

    DAB Diaminobenzidine tetraaminobiphenyl

    DAPI 4',6 Diamidino-2-phenylindole

    ddH2O Double distilled water

    DEPC Diethyl Pyrocarbonate

    Dlg1 Drosophila disc large tumour suppressor

    Dlg1 Discs large protein

    DMEM Dulbecco’s Modified Essential Medium

    DMSO Dimethyl sulfoxide

    DNA Deoxyribonucleic Acid

    E-PE Early-onset preeclampsia

    ECL Enhanced chemiluminescence

    ECM Extracellular matrix

    EMEM Eagle's Minimal Essential Medium

    EMT Epithelial-mesenchymal transition

    ERVW-1 Endogenous retrovirous group W member 1

    EVT Extravillous trophoblast

    EV Empty Vector

    FBS Fetal bovine serum

    Fe2+ Ferrous iron

    FIH Factor inhibiting HIF1

    Flt1 VEGF receptor 1 (aka VEGFR1)

    GAP GTPase activating protein

    GCM-1 Glial cells missing homolog 1

    GTP Guanosine triphosphate

    H&E Hematoxylin & Eosin

    H2O2 Hydrogen peroxide

  • viii

    HCl Hydrochloric acid

    HIF-1α Hypoxia inducible factor-1 alpha

    HRP Horseradish peroxidase

    Hz Hertz

    IF Immunofluorescence

    IHC Immunohistochemistry

    JAM Junctional adhesion molecules

    JmjC Jumonji C

    JMJD6 Jumonji C domain containing protein 6

    KCl Potassium chloride

    L-PE Late-onset preeclampsia

    LAMP-1 Lysosomal associated membrane protein 1

    LATS1/2 Large tumour suppressor kinase 1/2

    Lgl1/2 Lethal giant larvae protein 1/2

    MAE Mouse aortic endothelial cells

    MALDI-TOF Matrix-assisted laser desorption ionization time of flight

    MAP kinase Mitogen-activated protein kinase

    MDCK Madin-Darby Canine Kidney

    MgCl2 Magnesium chloride

    mL Millilitre

    mM Millimolar

    mm Hg Millimetres of mercury

    MMP Matrix metalloproteinases

    mRNA Messenger ribonucleic acid

    NH4Cl Ammonium Chloride

    O2 Molecular oxygen

    OE Overexpression

    Pals1 Protein associated with Lin-7 1

    Par3 Partitioning defective protein-3

    Par6 Partitioning defective protein-6

    Patj Pals1 associated tight junction protein homolog

    PBS phosphate buffer saline

    PDZ PSD95, Dlg1, ZO-1

    PE Preeclampsia

    PFA Paraformaldehyde

    PHD1-3 Prolyl hydroxylase domain (1-3)

    PLA Proximity ligation assay

    PLOD1 Procollagen-lysine 5-dioxygenase 1

    pMSC Placental mesenchymal cells

    pO2 Partial pressure of oxygen

    PSD95 Post synaptic density protein

    pSMAD2-3 Phosphorylated SMAD 2-3

    PTC Pre-term control

    qPCR Quantitative polymerase chain reaction

    Rac1 Ras related C3 Botulinum Toxin Substrate 1

    RhoA Ras homolog gene family member A

    Rich1 Rho-type GTPase activating protein 17

  • ix

    RIPA buffer Radioimmunoprecipitation buffer

    RNA Ribonucleic acid

    Scrib Scribble

    SDS Sodium dodecyl sulfate

    SDS-PAGE SDS polyacrylamide gel electrophoresis

    SEM Standard error of the mean

    siRNA Small interfering RNA

    Smad2,3,7 Small-Mothers Against Decapentaplegic 2,3,7

    Smurf1 Smad ubiquitination regulatory factor 1

    ss Scrambled sequence

    ST Syncytiotrophoblast

    Syx Rho guanine exchange factor

    TAZ Transcriptional coactivator with PDZ binding motif

    TBST Tris buffered saline +Tween

    TEAD1 TEA domain family member 1

    TGFβRI-II Transforming growth factor beta receptor I-II

    TGFβ1-3 Transforming growth factor beta 1-3

    TIMPS Tissue inhibitors of MMPs

    TJ Tight junction

    uM Micromolar

    VEGF Vascular endothelial growth factor

    VHL von Hippel-Lindau tumour suppressor protein

    WB Western blotting/blot

    YAP Yes-associated protein

    ZEB2 Zinc finger E-box binding homeobox 2

    ZO-1 Zona occludens-1

    ZONAB ZO- associated nucleic acid binding protein

    α-tubulin alpha tubulin

    β- actin Beta-actin

    µg Microgram

    µL Microliter

  • x

    List of Tables

    Table 2.1 Clinical features of patient population .......................................................................... 40

  • xi

    List of Figures

    Figure 1.1 Protein structures of ‘motin’ family members ............................................................... 5

    Figure 1.2 Polarity complexes in migrating epithelial cells ......................................................... 14

    Figure 1.3 Human placenta development and trophoblast differentiation .................................... 20

    Figure 1.4 Role of Smad-dependent and Par6-mediated TGFβ signalling in trophoblast cell

    differentiation ................................................................................................................................ 28

    Figure 1.5 Molecular and phenotypic characteristics of preeclampsia ......................................... 35

    Figure 1.6 Model of Hypothesis ................................................................................................... 38

    Figure 3.1 AMOT protein levels and mRNA expression during early placenta development ..... 64

    Figure 3.2 Spatial localization of AMOT in floating villi in early placenta development ........... 65

    Figure 3.3 Spatial localization of AMOT in anchoring villi in early placenta development ........ 66

    Figure 3.4 Co-localization of AMOT with tight junction protein ZO-1 in JEG3 choriocarcinoma

    cells ............................................................................................................................................... 70

    Figure 3.5 Effect of TGFβ1/3 on AMOT protein levels in JEG3 cells ........................................ 71

    Figure 3.6 Effect of TGFβ1/3 on AMOT and ZO-1 co-localization in JEG3 cells ...................... 72

    Figure 3.7 Effect of TGFβ1/3 on AMOT and F-actin co-localization in JEG3 cells ................... 73

    Figure 3.8 Contribution of Smad-dependent TGFβ signalling on AMOT localization ................ 76

    Figure 3.9 Contribution of Smad-dependent TGFβ signalling on AMOT protein levels ............. 77

  • xii

    Figure 3.10 Effect of TGFβ on AMOT localization in migrating edge of JEG3 cells ................. 80

    Figure 3.11 AMOT localization in live cell imaging of migrating JEG3 cells............................. 82

    Figure 3.12 Effect of AMOT 80 overexpression on migration rate of JEG3 cells during wound

    healing. .......................................................................................................................................... 83

    Figure 3.13 Effect of TGFβ1/3 on AMOT and Par6 co-localization in JEG3 cells ..................... 86

    Figure 3.14 Effect of TGFβ1/3 on AMOT and Par6 interaction in JEG3 cells ............................ 87

    Figure 3.15 AMOT 80 and Par6 interaction in human placenta tissue ......................................... 88

    Figure 3.16 Description and validation of AMOT 130 and AMOT 80 plasmid constructs ......... 91

    Figure 3.17 Importance of PDZ and coiled-coil binding domains to AMOT-Par6 interaction .... 92

    Figure 3.18 Effect of AMOT overexpression on protein levels of RhoA .................................... 94

    Figure 3.19 Effect of AMOT overexpression on RhoA localization ............................................ 95

    Figure 3.20 AMOT protein levels in preeclamptic and normotensive pre-term control placentae

    ....................................................................................................................................................... 98

    Figure 3.21 AMOT localization in preeclamptic and pre-term control placenta tissue sections .. 99

    Figure 3.22 AMOT 80 and Par6 association in preeclamptic and pre-term control placenta .... 100

    Figure 3.23 Effect of low oxygen on AMOT protein levels in JEG3 cells ................................ 102

    Figure 3.24 Effect of low oxygen on AMOT localization in JEG3 cells ................................... 103

  • xiii

    Figure 3.25 Effect of silencing and overexpressing JMJD6 on AMOT protein in JEG3 cells... 107

    Figure 3.26 Effect of overexpressing JMJD6 on AMOT localization in JEG3 cells ................. 108

    Figure 3.27 MALDI-Mass Spectrometry analysis of AMOT peptide mass profile following in

    vitro JMJD6 enzyme reaction ..................................................................................................... 109

    Figure 3.28 Effect of minoxidil induced inhibition of lysyl hydroxylation on AMOT protein

    levels in JEG3 cells ..................................................................................................................... 110

    Figure 3.29 Effect of minoxidil induced inhibition of lysyl hydroxylation on AMOT localization

    in JEG3 cells ............................................................................................................................... 111

    Figure 4.1 Putative model depicting the role and regulation of AMOT in normal placentation and

    in preeclampsia. .......................................................................................................................... 128

    Figure 5.1 Investigating AMOT in placenta mesenchymal cells (pMSC) isolated from term

    placentae. .................................................................................................................................... 135

    Figure 5.2 Investigating AMOT localization term and preeclamptic placental mesenchymal cells

    (pMSC) ....................................................................................................................................... 136

    Figure 5.3 Investigating AMOT and TAZ spatial association in the human placenta ............... 139

    Figure 5.4 Effect of TGFβ1/3 on AMOT and TAZ localization in JEG3 cells .......................... 140

  • 1

    Chapter 1

    Introduction

    Introduction

    Trophoblast proliferation, migration and invasion are critical events during human placenta

    development that contribute to the establishment of the feto-maternal interface in pregnancy.

    Changes in oxygen tension and downstream TGF signalling experienced by the developing

    placenta have been demonstrated to regulate these events. However, in preeclampsia (PE), a

    devastating pregnancy disorder associated with increased maternal and perinatal mortality

    worldwide, persistent chronic hypoxia and aberrant TGF signalling has been shown to impair

    trophoblast differentiation and function, ultimately leading to defects in spiral artery remodelling

    and vascularization of the fetus. A fundamental element in eukaryotic cells regulated by TGF

    signalling, that is also integral to cell migration processes, is apical-basolateral cell polarity. Yet,

    the contribution TGF to the regulation of a novel cell polarity protein, termed Angiomotin

    (AMOT), remains to be established. Moreover, AMOTs role in the human placenta and in

    trophoblast cell events remains unknown. In this chapter, I will first introduce the current literature

    surrounding AMOT’s discovery and function, next elucidate the complex regulation of placenta

    development by upstream oxygen and TGF signalling, and finally how it all goes awry in PE.

  • 2

    1.1 Angiomotin (AMOT)

    1.1.1 Discovery and Structure

    Angiomotin (AMOT) was first discovered in 2001 for its ability to bind to and mediate the effects

    of angiostatin, a circulating inhibitor of endothelial cell migration and tube formation during

    angiogenesis (Troyanovsky et al., 2001). Comprised of 675 amino acid residues and with a

    molecular mass of 80kDa, AMOT was the founding member of the ‘motin’ family of proteins

    (Bratt et al., 2002; Troyanovsky et al., 2001). Succeeding investigations identified a 130kDa

    AMOT product, structurally identical to the 80 kDa AMOT protein with an additional 409 amino

    acid N-terminal extension that arises from alternative splicing of the AMOT gene between exons

    2 and 3 (Ernkvist et al., 2006). Thus, it was determined that the AMOT gene encodes for two

    isoforms: AMOT 80 and AMOT 130. In pursuit of identifying conserved domains important for

    AMOT function, two other motin family members with significant sequence homology to AMOT

    were also identified: angiomotin-like 1 (AMOTL-1), and angiomotin-like 2 (AMOTL-2) (Bratt et

    al., 2002). The motin family of proteins exhibit two conserved protein motifs, namely a coiled-coil

    and PDZ binding domain (PSD95 (post synaptic density protein), Dlg1 (drosophila disc large

    tumour suppressor), ZO-1 (zona occludens-1)) (Figure 1.1).

    Coiled-coil domains, structurally characterized by two alpha helices woven around each other, are

    a principal protein motif permitting the oligomerization of proteins and mediating protein-protein

    interactions (Burkhard et al., 2001). PDZ domains are abundant, globular interaction motifs that

    facilitate protein-protein interactions during signal transduction events, and organize proteins to

    specific sites or membrane structures (Lee and Zheng, 2010). In particular, PDZ domains are

    common components of proteins that localize to tight junctions (Tsukita et al., 2001). In fact,

    studies in epithelial and endothelial cells have revealed all motin members to localize at the tight

  • 3

    junctions, where it is suggested they elicit an important role in apical-basolateral cell polarity and

    stability of the cytoskeleton (Moleirinho et al., 2014). The presence of these characterized protein-

    interaction domains underscores the capability of the motin family of proteins to be involved in

    cellular scaffolding and signal transduction events.

    The structural similarities amongst AMOT (80kDa and 130kDa isoforms), AMOTL-1 and

    AMOTL-2 foster a degree of functional redundancy; however, there is functional variability within

    the family. These functional differences are highlighted by the variable spatial and temporal

    expression patterns amongst members, and the tissue-and cell-dependent expression patterns

    (Moleirinho et al., 2014). Further, a number of functional and mechanistic studies have been

    conducted in pursuit of distinguishing the relevance of individual motin family members in

    different organ systems and signalling pathways.

    Early studies in mouse aortic endothelial (MAE) cells revealed that AMOT 80 expression increases

    endothelial cell migration and contributes to vessel tube formation (Troyanovsky et al., 2001).

    Moreover, studies examining the functional necessity of PDZ binding domain on AMOT 80

    revealed that mutations in its PDZ domain resulted in complete loss of migratory activity in MAE

    cells (Levchenko et al., 2003). Further, transgenic mice expressing AMOT with mutated PDZ lose

    response to growth factors and are embryonic lethal due to impaired vascularization. On the other

    hand, AMOT 130 expression did not promote migration or mediate a response to angiostatin but

    was found to induce changes in cell shape through binding and stabilization of the F-actin

    cytoskeleton (Ernkvist et al., 2006). This AMOT 130/ F-actin interaction was determined to be

    mediated by the N-terminal domain on AMOT 130 (Ernkvist et al., 2008). Thus, the distinction

    was made that AMOT 80 functions primarily in endothelial cell migration, whereas AMOT 130

    primarily controls changes in cell shape and regulates cytoskeleton reorganization. With regards

  • 4

    to AMOTL1 and AMOTL2, both proteins have been shown to interact with tight junction residing

    transmembrane proteins (Nishimura et al., 2002a; Patrie, 2005; Sugihara-Mizuno et al., 2007).

    However, AMOTL1 and AMOTL2 lack the hydrophobic angiostatin binding domain present in

    AMOT 130 and 80 and thereby fail to mediate its effects during angiogenesis (Bratt et al., 2002).

    Additionally, transcript expression analysis of motin family members at the time of AMOT

    discovery revealed that AMOT had highest mRNA expression in the human placenta, compared

    to AMOTL1 and AMOTL2 which had highest expression in lung and skeletal muscle (Bratt et al.,

    2002). Although this study identified AMOT expression in the human placenta, there has been no

    further investigation into its role. Hence, the focus of the present study remained on AMOT

    protein, both 130kDa and 80kDa isoforms, and its undiscovered role in the human placenta.

    These aforementioned studies act as an introduction to the breadth of work which has been

    subsequently conducted to distinguish the function of AMOT isoforms. These include

    investigations into regulation of the Hippo pathway, as well as studies in cell polarity regulation,

    which will be described in this chapter.

  • 5

    Figure 1.1 Protein structures of ‘motin’ family members

    The members of the motin family of proteins include: (1) Angiomotin (AMOT), (2) Angiomotin

    Like-1 (AMOTL1), and (3) Angiomotin Like-2 (AMOTL-2). Notably, AMOT is expressed as two

    isoforms due to alternative splicing, AMOT 130 and AMOT 80, which corresponds to their

    respective molecular weights. The motin protein members share distinct structural characteristics.

    AMOT 130 and AMOT 80 contain coiled coil regions and PDZ binding domains, motifs conserved

    across all motin members, as well as an angiostatin binding domains. However, AMOT 130

    contains a 409 amino acid N-terminal extension comprising distinct PPxY binding motifs that

    promotes its interaction with Hippo Pathway effectors, YAP (Yes-associated protein) and TAZ

    (Transcriptional coactivator with PDZ binding motif), in addition to mediating the interaction with

    the F-actin cytoskeleton (Chan et al., 2011; Ernkvist et al., 2006). AMOTL-1 and AMOTL-2

    exhibit similar sequence identity with AMOT 130, however they lack the angiostatin binding

    domain. AMOTL-1 was initially identified in a screen for novel tight-junction associated proteins

    (Nishimura et al., 2002b) and named JEAP (junction-enriched and-associated protein), but later

    discovered to share sequence homology with AMOT 130 and was subsequently named AMOTL-

    1. AMOTL-2 is also referred to as MASCOT (MAGI-1-associated coiled-coil tight junction

    protein) due to its colocalization with MAGI-1 at epithelial tight junctions (Patrie, 2005). Despite

    these structural similarities, the individual motin proteins are functionally distinct, made evident

    by their differential expression in various tissues and cell types. PDZ- PSD95 (post synaptic

    density protein), Dlg1 (drosophila disc large tumour suppressor), ZO-1 (zona occludens-1).

    Angiomotin (AMOT)

    130 kDa

    80 kDa

    1084 a.a

    675 a.a

    1 20 280 312 342 672 675

    AMOT 130

    AMOT 80

    Angiostatin binding

    PDZ binding

    Coiled coil binding

    106 LPTY

    109

    1 429 689 721 751 1081 1084239PPEY

    242

    284 PPEY287

    867 1005

    458 596

    AMOT Like-1 (AMOTL1)

    188 LPTY

    191

    1 438 694 729 953 956310PPEY

    313

    367 PPEY370

    762 106 kDa956 a.a

    AMOT Like-2 (AMOTL2)

    86 kDa780 a.a

    104 LPTY

    107

    1 308 581 777 780210 PPQY213

    252 PPVF255

  • 6

    1.1.2 Role of AMOT in Hippo Pathway Signalling

    Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are

    transcriptional co-activators of the Hippo Pathway, a signalling pathway which has over the years

    been regarded to have important regulatory control in several cellular processes including cell

    proliferation and differentiation, and in cancer cell progression (Moroishi et al., 2015). YAP and

    TAZ are structural homologs and exhibit functional redundancy. Specifically, YAP and TAZ

    transcription factors shuttle between cytoplasm and the nucleus, where they are permitted to

    interact with TEA domain family members (TEAD) transcription factors to induce the

    transcription of genes controlling cell proliferation and epithelial-mesenchymal transition (Lei et

    al., 2008; Zhao et al., 2008). Tight regulation of YAP/TAZ activity is essential as hyperactivity of

    YAP/TAZ has been implicated in a variety of malignant and metastatic cancers and associated

    with poor outcomes (Piccolo et al., 2014). YAP and TAZ activity is regulated by inhibitory Hippo

    kinases large tumour repressor 1 and 2 (LATS1 and LATS2), which directly phosphorylate YAP

    and TAZ at several serine residues, promoting the sequestration of YAP and TAZ in the cytoplasm

    (Oh and Irvine, 2010), or alternatively targeted for ubiquitination and subsequent proteasomal

    degradation (Zhao et al., 2010). However, a third fate exists, which is the direct binding and

    regulation of YAP and TAZ by AMOT 130 (Chan et al., 2011; Zhao et al., 2011). AMOT 130

    interaction with YAP/TAZ occurs between tryptophan binding domains (WW) on YAP/TAZ, and

    the PPxY binding motif within the N-terminal extension found on AMOT 130 (Chan et al., 2011).

    However, the nature of AMOT’s regulation over YAP/TAZ remains a subject of controversy

    within the field of the Hippo pathway, as conflicting reports have outlined AMOT as both a

    negative and a positive regulator of YAP/TAZ activity.

  • 7

    AMOT 130 as a Negative Regulator of YAP/TAZ in the Hippo Pathway

    The first evidence of AMOT 130 as a negative regulator of Hippo Pathway was demonstrated in a

    study by Zhao et al showing that AMOT 130 inhibits YAP activity in Madin-Darby Canine Kidney

    (MDCK) cells by sequestering YAP to intracellular tight junctions, or to the F-actin cytoskeleton

    in the cytoplasm, thus effectively preventing YAP nuclear translocation and gene transcription

    (Zhao et al., 2011). This mechanism is independent of LATS1/2 mediated phosphorylation.

    Additionally, AMOT 130 can negatively regulate YAP/TAZ by promoting inhibitory

    phosphorylation of YAP and TAZ (Zhao et al., 2011). This apparent tumour suppressor role of

    AMOT 130 on YAP/TAZ has been elucidated in a number of cancer studies (Moleirinho et al.,

    2017; Moroishi et al., 2015). For example, expression of AMOT 130 is significantly decreased in

    clinical lung cancer specimens, and furthermore knockdown of AMOT 130 in lung

    adenocarcinoma cancer cell line was shown to initiate cancer proliferation, migration and invasion

    by promoting YAP/TAZ nuclear translocation (Hsu et al., 2015).

    AMOT 130 as a Positive Regulator of YAP/TAZ in the Hippo Pathway

    In stark contrast to these findings attributing an inhibitory role for AMOT 130 in Hippo signalling,

    reports in biliary epithelial cells have demonstrated AMOT 130 to function as a positive-regulator

    of YAP/TAZ (Hong, 2013). AMOT 130 can compete with LATS1 for binding to YAP, thereby

    preventing YAP phosphorylation, which facilitates YAP/TAZ nuclear translocation. Further,

    studies in hepatic epithelial cells have demonstrated AMOT 130 can also localize to the nucleus,

    where it forms a complex with YAP and TEAD1, thus contributing to transcription of YAP/TAZ

    target genes associated with tumorigenesis (Yi et al., 2013a). This positive interplay between

    AMOT and YAP/TAZ has been observed in models of hepatic carcinoma, renal epithelial cells

    and renal cell cancers (Lv et al., 2016; Yi et al., 2013a).

  • 8

    Recent studies in embryonic kidney cells, endothelial cells, and mouse and zebrafish embryos

    suggest that the phosphorylation status of AMOT is important in distinguishing whether AMOT

    functions as a positive or negative regulator of YAP/TAZ mediated transcription in the Hippo

    pathway (Chan et al., 2013; Dai et al., 2013; Hirate and Sasaki, 2014). Phosphorylation of AMOT

    by LATS1/2 promotes AMOT mediated redistribution of YAP/TAZ from either the nucleus or

    cytoplasm, to the plasma membrane. As such, the ability of YAP/TAZ to promote cell proliferation

    and tumorigenesis is abrogated. On the other hand, when AMOT is in its unphosphorylated state,

    AMOT is able to translocate into the nucleus and function as a positive cofactor in the transcription

    of YAP target genes.

    1.1.3 Role of AMOT in Cell Migration

    Independent of its role in mediating downstream Hippo pathway gene transcription, AMOT has

    also been shown to influence endothelial and epithelial cell migration. This was apparent in

    zebrafish studies which showed Amot knockdown reduced the number of filopodia in endothelial

    cells, severely impairing the migration of intersegmental vessels during embryogenesis (Aase et

    al., 2007). Total knockdown of Amot in mice resulted in embryonic lethality between E11 and

    E11.5, as a result of severe vascular insufficiency in the intersomitic region and dilated vessels in

    the brain (Aase et al., 2007). Notably, the placenta was not thoroughly investigated. Nonetheless,

    this study suggested AMOTs role in cell migration could be attributed to novel function of AMOT

    in the regulation of cell polarity (Aase et al., 2007). Thereafter, studies have underscored how the

    intracellular scaffolding abilities of AMOT can promote the binding and shuttling of various cell

    polarity components, including tight junction components, polarity complex proteins and small G-

    proteins, in order to promote alterations in cell polarity.

  • 9

    1.2 Cell Polarity Regulation

    1.2.1 Tight Junctions

    Cell polarity is a fundamental element in all eukaryotic cells that controls changes in cell-shape,

    cell migration, cell fusion and epithelial-to-mesenchymal transition (Martin-Belmonte and

    Mostov, 2008). Apical-basolateral cell polarity arises in epithelial cells as a result of asymmetric

    distribution of lipids and proteins to the cell surface on the apical or basolateral ends of the cell

    (Assemat et al., 2008). Alongside the adherens junction, which provide structural support to the

    cells via linkage to cytoskeletal elements, the tight junctions (TJ) is a key intracellular junctional

    complex involved in the regulation of cell polarity (Shin et al., 2006). The TJ mediates adhesion

    between neighboring cells by forming a selectively permeable barrier to diffusion through

    intercellular space, a property referred to as the “barrier” function. However, in regards to its role

    in cell polarity, the TJ is able to restrict the intracellular localization of proteins and

    macromolecules between the apical and basolateral regions of the cell by delineating the boundary

    between these two domains, a characteristic referred to as the “fence function” (Shin et al., 2006;

    Zihni et al., 2016). The TJ is comprised of transmembrane protein components; occludin, claudin

    and junctional adhesion molecules, all of which interact with underlying peripheral membrane

    proteins to form a complex protein network (Shin et al., 2006). Importantly, these underlying

    peripheral membrane proteins serve as a link to the actin cytoskeleton (Zihni et al., 2016). These

    underlying proteins, which typically contain PDZ and tryptophan (WW) binding domains, include

    scaffolding proteins, kinases, phosphatases, small GTPases and their activating proteins, actin

    binding proteins and F-actin itself (Quiros and Nusrat, 2014). For example, Zonula occludens-1

    (ZO-1) is a peripheral scaffolding protein belonging to the membrane-associated guanylate kinase

    (MAGUK) family of proteins, which connects tight junction proteins (occludin and claudin) to the

    actin cytoskeleton (Shin et al., 2006). Further, Ras homolog gene family member A (RhoA), a

  • 10

    small GTPase protein, is another peripheral membrane protein that has dual regulatory roles in

    tight junction assembly and actin cytoskeleton arrangement (Quiros and Nusrat, 2014). Thus,

    molecules involved in tight junction stability, are also involved in cytoskeleton reorganization and

    altogether contribute to the establishment of epithelial polarization.

    1.2.2 Polarity Protein Complexes

    There are three main protein complexes involved in the maintenance and loss of apical-basolateral

    polarity: Par/aPKC complex, composed of Par3, Par6, and atypical protein kinase C (aPKC);

    Crumbs complex, composed of crumbs, Pals1 (Protein associated with Lin-7), Patj (Pals1

    associated tight junction protein homolog); and Scribble complex, comprised of scribble, discs

    large (Dlg) and lethal giant larvae (Lgl) (Assemat et al 2007). Protein subunits within these

    complexes are able to interact with one another through shared PDZ-binding domain motifs and

    localize certain cellular components to poles of the cell, promoting apical-basolateral membrane

    identity and cell polarization (Assemat et al., 2008; Martin-Belmonte and Mostov, 2008).

    The Par complex defines the apical region of the cell, and is involved in the early events of cell-

    cell adhesion and tight junction formation (Henrique and Schweisguth, 2003). Specifically, Par3

    binds to transmembrane junctional adhesion molecules (JAM) at the site of cell-cell contact, while

    Par6 and aPKC initially reside with Lgl component of the scribble complex in the cytoplasm. Next,

    cell division control protein 42 homolog (Cdc42), a master regulator of actin cytoskeleton

    rearrangements explained in detail below, becomes activated and promotes Par6 and aPKC to

    associate with Par3 at the tight junction to complete the Par complex at the apical/basolateral

    membrane (Yamanaka et al., 2001). Simultaneously, the Lgl component of the scribble complex

    is displaced to the basolateral membrane where it is found associate to other scribble proteins and

    defines the basolateral domain. The crumbs complex resides in the apical region alongside the Par

  • 11

    complex, and collectively they identify the apical-membrane (Figure 1.2A). Loss of any one of

    these polarity complex proteins can lead to the disruption of cell polarity in mammalian epithelial

    cells (Assemat et al., 2008; Macara, 2004; Martin-Belmonte and Mostov, 2008).

    1.2.3 Role of Cell Polarity in Proliferation, Migration and Invasion

    It is evident that tight junction proteins are not only permeability barriers between cells, but are

    sensors involved in a variety of cellular processes, such as cell proliferation. In conditions of

    increasing cell density within epithelial sheets, tight junction associated mechanisms sense this,

    and inhibit further cell proliferation by impeding transcription of proliferative related genes.

    Specifically, tight junction protein ZO-1 increases expression in conditions of high cell density

    (Balda and Matter, 2000), binds to ZO-1-associated nucleic acid binding protein (ZONAB), and

    sequesters it to the cytoplasm to prevent its mediated gene transcription and ultimately reduce

    proliferation (Balda et al., 2003). Another example is the previously outlined role of AMOT 130

    in recruiting Hippo pathway kinases to phosphorylate and inactivate transcription activators

    YAP/TAZ, or sequestering YAP/TAZ out of the nucleus (Zhao et al., 2011).

    Tight junction proteins are also important when applied in the context of epithelial cell migration.

    Importantly, a series of tightly coordinated steps are required for proper directional cell migration.

    Cell migration begins in response to external stimuli such as growth factors or extracellular matrix

    (ECM) molecules, where the driving force is the polarized extension of a leading edge protrusion,

    or “lamellipodium” in response to direction of movement. Following the formation of the

    lamellipodium, new adhesion sites are established at the leading edge, the cell undergoes

    actin/myosin contraction, and previous adhesion sites located at the tail of the cell are detached to

    permit cell movement. One family of proteins that plays a pivotal role in these steps of cell

    migration is the tight junction residing Rho family of GTPases, namely Cdc42, Rac1 (Ras related

  • 12

    C3 Botulinum Toxin Substrate-1) and RhoA (Ras homolog gene family member A). Cdc42

    responds to external stimuli and regulates the direction of cell movement, Rac1 stimulates actin

    polymerization and integrin adhesion complexes to the lamellipodium, and RhoA promotes actin:

    myosin contraction to complete the cycle (Raftopoulou and Hall, 2004). Studies aimed at

    investigating the mechanism in which polarity proteins assemble at the leading edge of migrating

    epithelial cells revealed that tight junction component occludin localized to the leading edge of

    migrating cells, and regulated directional cell migration by binding to and localizing polarity

    complex aPKC-Par3 and Patj to the leading edge. In turn, this promoted cell protrusions by

    activating Rac1 to direct lamellipodia formation (Du et al., 2010) (Figure 1.2B).

    1.2.4 AMOT as a Novel Regulator of Cell Polarity

    Studies conducted in endothelial and epithelial cells have characterized AMOT localization at the

    tight junction and determined a role for AMOT in apical-basolateral cell polarity and cytoskeletal

    stabilization (Lv et al., 2017). A study investigating polarity complexes in MDCK cells determined

    that the coiled-coil domain on AMOT 80 binds to Rich1, a GTPase activating protein necessary to

    tight junction formation and stabilization, and collectively the two proteins localize to the tight

    junction (Wells et al., 2006). Here, AMOT 80 was demonstrated to directly interact with Par and

    Crumbs Complex proteins Pals1, Patj/Mupp1, and Par3 via their individual PDZ-binding motifs

    (Wells et al., 2006). However, MDCK cells stably overexpressing AMOT 80 exhibited tight

    junction dissolution. A potential explanation is that AMOT 80 overexpression above a certain

    threshold can promote the selective redistribution of Par and Crumbs complex components

    alongside scaffolding protein AMOT into punctuate structures within the cytoplasm, and away

    from the tight junction (Wells et al., 2006). Resulting from the sequestration of essential tight

    junction proteins away from cell boundaries to the cytoplasm include disruptions in tight junction

  • 13

    integrity, alterations in apical-basolateral cell polarity and promotion of cell movement. The roles

    of AMOT 80 in cell migration and cell polarity were linked in a study by Aase et al, where isolated

    and immortalized embryonic stem cells from an Amot deficient mouse model (Amot - EC) exhibited

    defects in cell migration, as wild type EC migrated five times longer than Amot – EC in two

    independent migration assays (Boyden chamber and time-lapse wound healing) (Aase et al., 2007).

    AMOT – EC also displayed defects in cell polarization, as shown by failure of the Golgi apparatus

    and GTPase Rich1 to localize to the lamellipodia. Further, AMOT – EC exhibited defects in actin

    cytoskeleton organization and changes in cell shape, as elucidated by disorganized pattern of actin

    fibers and shorter focal adhesions in Amot – EC compared to wildtype EC (Aase et al., 2007). In

    another study, Ernkvist et al found that AMOT 80 binds to RhoA GTPase exchange factor ‘Syx’

    via its PDZ binding motif, which forms a ternary protein complex with Patj (AMOT/Patj/Syx) and

    regulates activity of RhoA GTPase in lamellipodium of migrating cells (Ernkvist et al., 2009).

    These findings provided further evidence that AMOT’s role as a cell polarity regulator is intricately

    connected to its role in promoting cell migration.

    The role of cell polarity, and the signalling pathways that regulate it, has been a subject of intense

    investigation in the field of cancer due to the strong correlation between malignancy of epithelial

    cancer and loss of epithelial organization attributed to loss of polarity (Bilder, 2004). Further, loss

    or deregulation of epithelial cell polarity processes has been characterized as a hallmark of cancer,

    as it plays a role in the initiation of tumorigenesis and later stages of tumour development (Royer

    and Lu, 2011). Interestingly, comparisons have been drawn between cancer cell differentiation in

    the tumour microenvironment, and the differentiation of trophoblast cells at the fetomaternal

    interface (Holtan et al., 2009). More specifically, the parallel lies in the proliferation, migration

    and invasion of tumour cells during cancer, and trophoblast cells during placental development.

  • 14

    Figure 1.2 Polarity complexes in migrating epithelial cells

    (A) The Crumbs, Par and Scribble protein complexes tightly regulate cell polarity in epithelial

    cells. The localization of these complexes and their complex protein-protein interactions define

    the apical and basolateral regions within epithelial cells. The Par and Crumbs complex define the

    apical domain of epithelial cells, whereas the Scribble complex defines the basolateral domain.

    During the early events of tight junction formation, Par6 and aPKC components of the Par complex

    are found bound to Lgl component of the scribble complex within the cytoplasmic region of the

    A

    B

  • 15

    cell. Upon sensing of extracellular cues to establish apical basolateral cell polarity, Cdc42 is

    activated and promotes Par6 and aPKC to redistribute to the tight junction to join Par3 component

    which is bound to transmembrane protein JAM, and ultimately complete the Par complex.

    Simultaneously, Lgl component is displaced to basolateral membrane and joins to other scribble

    complex proteins(Chatterjee and McCaffrey, 2014). The complex interactions amongst these

    protein complexes, as well as interactions between individual proteins and cellular scaffolding

    proteins, occur via shared putative PDZ binding domains. The known interactions between polarity

    proteins and scaffolding protein AMOT are noted by the blue asterisk (Sugihara-Mizuno et al.,

    2007; Wells et al., 2006). (B) The distinct, step-wise events during epithelial cell migration require

    precise regulation by Rho family members (Cdc42, Rac1 and RhoA), as well as require changes

    in polarity complex localization and their complex interactions (1) Cdc42, a master regulator of

    actin cytoskeletal rearrangements, controls the direction of cell migration in response to external

    growth factors or ECM signals by promoting the protrusion of the lamellipodia (cells leading

    edge). Rac1 stimulating actin polymerization at the cell’s leading edge, as well as the localization

    of apical polarity complex proteins to the leading edge mediate the formation of this protrusion.

    (2) New cell adhesions at the lamellipodia are generated via Rac1 promoting the formation integrin

    adhesion complexes. (3) Rho A promotes actin-myosin contraction of the epithelial cell body, (4)

    simultaneously promoting the dissolution of old, rear adhesions and retraction of the trailing ‘tail’

    of the epithelial to promote cell migration in the respective direction (Raftopoulou and Hall, 2004).

    TJ (Tight junction) AJ (Adherens junction), Par6 (Partitioning defective protein 6), Par3

    (Partitioning defective protein 3), Patj (Pals1 associated tight junction protein), Pals1 (Protein

    associated with Lin-7 1), Crb, (Crumbs) Cdc42 (Cell division control protein 42), aPKC (atypical

    protein kinase 1), Scrib (Scribble), Lgl1/2 (Lethal giant larvae protein homolog1/2), Dlg (Discs

    large protein), RhoA (Ras homolog gene family member A), Rac1 (Ras related C3 Botulinum

    Toxin Substrate 1).

  • 16

    1.3 Human Placenta Development

    Successful pregnancy is fundamentally dependent on placental function, and thereby hinges on

    proper placenta development. The human placenta is vital to the proper growth and development

    of the fetus, as elucidated by its multi-functionality in a variety of critical functions including gas

    exchange, transportation of nutrients, elimination of waste, and production of hormones (Costa,

    2016). Development of the human placenta begins immediately following fertilization and is a

    continuous process that occurs alongside development of fetus. Specifically, the formation of the

    blastocyst on the fifth day after fertilization delineates two critical structures: (1) the inner cell

    mass, which subsequently forms the embryo and fluid-filled blastocoel, and (2) the outer layer of

    cells referred to as trophectoderm, which continue on to form the placenta and fetal membranes.

    Initial nourishment of the developing embryo is provided through uterine secretions containing

    oxygen and metabolic substrates, and subsequent uptake by the trophoblast layer (Burton et al.,

    2001). However, to provide sustainable nutrient and oxygen support necessary for further

    development, access to maternal decidua is required. Specifically, the blastocyst implants into the

    uterine lining (referred to as the ‘decidua’) via invasion of the outer trophoblast cells into the

    endometrial layer of the uterus. Initial trophoblast invasion into the decidua leads to erosion of

    uterine tissue, and consequently generates a network of lacunae which become filled with maternal

    blood for nutrition of the fetus. This blood-filled lacuna network is referred to as the ‘intervillous

    space’. Following implantation of the blastocyst, progenitor trophoblast cells differentiate into

    distinct trophoblast subtypes which are vital to the development of a fully functioning placenta.

    These trophoblast differentiation events, and their regulation, will be outlined in this next section.

  • 17

    1.3.1 Trophoblast Differentiation

    Progenitor trophoblast cells, referred to as cytotrophoblast (CT), are proliferative cells with two

    fates during development. One is fusion of CT to form the multi-nucleated syncytiotrophoblast

    cell layer (ST), which functions as the site of gas and nutrient exchange between the mother and

    fetus in the intervillous space. These two cellular layers, along with a mesenchymal core of

    connective tissues and blood vessels, comprise a villous structure directly bathed in maternal blood

    referred to as the floating villus. Concurrently, CT also undergo transformation to become

    extravillous trophoblast cells (EVT). Here, the EVT’s erupt through the overlaying syncytium at

    4-5 weeks of gestation to form cellular columns, which anchor the villi to the maternal decidua,

    and are known as the anchoring villi. This transformation of CT to EVT and subsequent

    attachment to uterine tissue involves cellular differentiation of CT from a proliferative, to a

    migratory, and finally invasive phenotype moving from proximal to distal ends of the anchoring

    villi. Once EVTs acquire invasive capabilities, they first undergo interstitial invasion and infiltrate

    the decidua. At the decidua, the EVTs begin endovascular invasion of the maternal spiral arteries

    by displacing vascular smooth muscle and endothelial cells. This effectively transforms the spiral

    arteries from narrow, high-resistance vessels, into wide, low resistance conduits, facilitating the

    increase of oxygenated blood flow into the lacunar vascular system and establishment of

    uteroplacental circulation. It is at this point in development where important physiologic changes

    in oxygen tension occur (Figure 1.3).

    Distinct molecular changes occur within epithelial CT as they transform into EVTs and begin their

    quest towards invading the maternal spiral arteries. One such change includes the increased

    secretion of metalloproteinases, which breaks down extracellular matrix proteins to facilitate EVT

    migration and invasion through the endometrium (Fisher et al., 1989). Another important

  • 18

    molecular alteration involves the upregulation of 5 and 1 integrins in differentiating and

    invasive EVTs, which has proven to be critical for EVT invasion (Damsky et al., 1994).

    Interestingly, this process of EVT differentiation in the human placenta has been compared to the

    process of epithelial-mesenchymal transition (EMT) often seen during embryonic development or

    cancer metastasis (Kalluri and Weinberg, 2009). During EMT, epithelial cells lose expression of

    cell-cell junction molecules (i.e. E-cadherin, ZO-1, and desmoplakin), adhere to ECM via integrin

    binding, and shift cell polarity from apical-basolateral to “front-back” orientation to facilitate

    migratory and invasive potential (Hay, 1995). Recently, gene expression analysis by qPCR

    revealed that EVT cells isolated from first trimester placentae exhibited downregulated expression

    of epithelial markers such as E-cadherin and occludin, and upregulated expression of mesenchymal

    markers such as vimentin, fibronectin and extracellular matrix integrins 5 and 1, when

    compared to first-trimester isolated CT (DaSilva-Arnold et al., 2015). Additionally, EVT cells had

    upregulated matrix metalloproteinases MMP2 and MMP9 that are necessary for invasion of ECM,

    as well as a robust increase in EMT regulator ZEB2 (Zinc finger E-box binding homeobox 2)

    (Gheldof et al., 2012). EMT processes require strict regulation by growth factors and other

    molecules, or else pathologies may arise in which cells have aberrant abilities to grow, proliferate,

    migrate or invade (i.e. carcinoma progression). EMT-like trophoblast differentiation, and resultant

    invasion, is tightly regulated during human placenta development by a variety of growth factors,

    cytokines and most evidently, changes in oxygen tension (Knofler, 2010).

    Prior to endovascular EVT invasion of the spiral arteries, the early human placenta must develop

    in a hypoxic environment to protect the embryo against oxygen free radical mediated damage

    (Burton et al., 2003). The low oxygen environment is established as a result of EVTs forming

    endovascular ‘plugs’ along the lumen of spiral arteries, which restricts maternal blood flow into

  • 19

    the intervillous space. Development in low oxygen is required not only to protect the developing

    embryo, but also functions to maintain progenitor trophoblast cells in a proliferative, non-invasive

    phenotype (Genbacev et al., 1996). Later, coinciding with the timing of spiral artery remodeling,

    steep changes in placental oxygen tension occur; oxygen levels measured to be 50mmHg by 12- weeks of gestation (Jauniaux et al., 2000). Recent

    studies have, however, suggested increases in oxygen tension may occur even earlier; contrast

    enhanced ultrasound imaging showed detectable increases in blood flow into intervillous space as

    early as 6-weeks of gestation due to progressive disintegration of uteroplacental ‘plugs’ (Roberts

    et al., 2017). Nonetheless, these alterations in oxygen tension experienced by the placenta have

    been shown to regulate the aforementioned trophoblast differentiation events via transcription

    factor hypoxia-inducible factor 1 (HIF-1) (Caniggia et al., 2000). Specifically, in low oxygen

    conditions such as those seen in early placenta development, HIF-1 positively regulates levels of

    transforming growth factor 3 (TGF3) to maintain trophoblast cells in a proliferative, non-

    invasive phenotype. Increased exposure to oxygen reduces HIF-1 and levels of TGF3, allowing

    EVT differentiation and invasion into the decidua to continue (Caniggia et al., 1999; Caniggia et

    al., 2000; Ietta et al., 2006).

    TGFs are a subgroup of growth factors within the TGF superfamily that regulate multiple

    biological processes including cell proliferation, differentiation, migration, EMT and apoptosis

    (Massague, 2008). Interestingly, the human placenta is a major tissue source of TGF (Jones et

    al., 2006), where it has been reported to regulate trophoblast cells during development. The

    signalling events activated by the TGF family of growth factors, and how this impacts trophoblast

    cell function is outlined next.

  • 20

    Figure 1.3 Human placenta development and trophoblast differentiation

    (A) General architecture and vascularization of the mature human placenta. The fetal side of the

    placenta (chorion) is comprised of umbilical veins and arteries, chorionic villi (cytotrophoblast

    (CT) and syncytiotrophoblast (ST) layers), and amnion. The maternal side of the placenta attached

    to the uterine wall, is comprised of uterine vessels and decidua basalis. Upon remodeling of the

    maternal spiral arteries by distal, endovascular extravillous trophoblast cells (EVT), the maternal

    A

    B

    Oxygen Gradient ~20 mmHg ~2-3% O2

    ~55 mmHg ~6-8 % O2

    5-9 weeks 10-12 weeks

    Fetal Side Maternal Side Decidua Myometrium

    Anchoring villi column

    Floating villi

    ST CT

    Proximal Intermediate Distal

    pMSC

    Spiral artery

    Migratory EVT

    Invasive EVT

    Fetal Blood Vessel Stroma

    Umbilical arteries

    Umbilical veins

    Chorionic Villi

    Maternal spiral artery

    Intervillous space

    CT ST

    EVT

  • 21

    blood pools into the intervillous space and comes in direct contact with the chorionic villi structure,

    achieving uteroplacental vascularization. (B) Structure of floating villi and anchoring villi at the

    fetomaternal interface. In the floating villi, underlying proliferative CT fuse to form the overlying,

    multinucleated ST that facilitates gas and nutrient exchange. Placenta mesenchymal cells (pMSC)

    and fetal blood vessels and capillaries are found within the stromal core of the chorionic villi.

    During the formation of the anchoring villi, trophoblast cells break through the syncytium and

    develop into EVT to form an anchoring column. In the anchoring column, EVTs differentiate

    through proliferative, migratory and invasive phenotypes as they move from proximal to distal

    ends of the column. Distal EVTs establish fetomaternal blood flow by invading through the

    maternal decidua (interstitial EVTs) to ultimately reach and remodel the spiral arteries within the

    myometrium layer (endovascular EVTs). Levels of oxygen regulate these trophoblast

    differentiation events. During early gestation (5-9 weeks), low oxygen levels retain trophoblast

    cells in a proliferative, undifferentiated state; however, following a rise in oxygen tension around

    10-12 weeks, CT differentiate into migratory and invasive EVTs required for spiral artery

    remodeling and increase in maternal blood flow to the intervillous space and ultimately the fetus

    (Simon and Keith, 2008).

  • 22

    1.3.2 TGF signalling pathways

    The TGF superfamily is the largest family of secreted morphogens and its members are highly

    conserved across animals (Wrana, 2013). In addition to the three TGF isoforms (TGF1, TGF2

    and TGF3), this superfamily of growth factors also includes activins, bone morphogenic proteins

    (BMP), and growth differentiating factors (GDF). While all three TGF isoforms are expressed in

    the human placenta, TGF3 is the only isoform demonstrated to exhibit a temporal expression

    pattern, peaking at 7/8 weeks of gestation, and dropping thereafter. This is unlike TGF1 and

    TGF2, which remained consistently expressed across gestation (Caniggia et al., 1999). As

    mentioned, this temporal peak in TGF3 is attributed to the hypoxic environment during early

    placenta development, where HIF-1 levels are abundant and positively regulate levels of TGF3

    (Caniggia et al., 2000; Ietta et al., 2006). The drop in TGF3 levels coincide with an increase in

    oxygen tension and a reduction of HIF-1 stability at 10-12 weeks of gestation. Aside from its

    differential expression pattern, TGF3 has proven to be an inhibitor of cytotrophoblast outgrowth

    and invasion. Specifically, in vivo studies of placental explants showed that inhibition of TGF3,

    but not TGF1 or TGF2, restored the invasive capabilities of trophoblast cells, and increased

    both MMP production and increase fibronectin deposition (Caniggia et al., 1999). Moreover, only

    the TGF3 isoform was found elevated in placentae complicated with preeclampsia. Altogether

    these data suggest a role for TGF3 in regulating trophoblast differentiation events during human

    placenta development. However, investigation into the soluble factors within the decidua that

    regulate trophoblast invasion revealed that decidual derived TGF1 plays a role in the inhibition

    of trophoblast outgrowth and invasion (Graham and Lala, 1991; Lala and Graham, 1990).

    Mechanistically, in vitro studies in first trimester derived trophoblast cells demonstrated TGF1

    to supress trophoblast invasion by: (1) upregulating tissue inhibitors of metalloproteases (TIMPs),

  • 23

    which inhibit trophoblast-derived matrix proteases required for invasion (Graham and Lala, 1991);

    (2) increasing the fusion of trophoblast cells into non-invasive multinucleated cells (Graham et al.,

    1992); (3) upregulating cell surface integrins that result in elevated trophoblast adherence to ECM

    that impedes cell migration (Irving and Lala, 1995); (4) downregulation of urokinase type

    plasminogen activator (uPA), which is also required for invasion (Graham and Lala, 1992).

    Investigation into the expression of TGF in maternal decidua and placenta revealed TGF2 to be

    particularly expressed in the ECM of first-trimester decidua and cytoplasm of term decidua cells

    (Graham et al., 1992; Lysiak et al., 1995).

    TGF signalling begins with generation of mature homo-or heterodimer TGF ligands. Following

    proteolytic cleavage of dormant, precursor TGF proteins within the extracellular matrix (ECM),

    the mature cleaved segments actively dimerize through disulfide links (Budi et al., 2017). The

    resultant dimerized TGF ligands are now primed to bind to a heteromeric transmembrane

    complex of serine/threonine receptors and activate downstream TGF signalling. This heteromeric

    complex is composed of two major types of transmembrane receptors: TGF receptor type I

    (TGFRI), also referred to as activin receptor-like kinase (alk); and TGF receptor type II

    (TGFRII), both of which possess intrinsic serine-threonine kinase activity (Attisano and Wrana,

    2002; Wrana, 2013). While seven types of TGFRI, commonly referred to as alk1-7, have been

    identified, TGF signalling occurs primarily via alk5. In the instance of endothelial cells, however,

    signalling also occurs through alk1 (Piek et al., 1999). Following TGF ligand binding,

    downstream signalling occurs via Smad-dependent and Smad-independent pathways.

    In Smad-dependent TGF signalling, also referred to as canonical TGF signalling, the TGF

    ligand binds to the constitutively active TGFRII, and promotes trans-phosphorylation and

  • 24

    activation of TGFRI (alk5) at a glycine-serine (GS) rich region. Activation of alk5 kinase activity,

    in turn, promotes the phosphorylation of Smad2 and Smad3 at C-terminal serine residues, which

    form a trimeric complex with Smad4 that translocates into to the nucleus (Wrana, 2013). Resident

    in the nucleus, this Smad complex interacts with other transcription factors to either activate or

    repress the expression of select genes, such as those involved in cell proliferation, migration and

    invasion. For example, in oral squamous cancer cells, TGFβ signalling via Smad3 upregulated the

    expression of microRNA miR-455-5p which promoted cancer cell proliferation (Cheng et al.,

    2016). Inhibitory Smad proteins including Smad7 can repress TGF signalling by directly binding

    to phosphorylated TGFRI (Wrana, 2013). This effectively prevents the phosphorylation of

    Smad2/3 while simultaneously targeting the receptor for degradation. Although Smad proteins are

    recognized as the main mediators of TGF signalling, TGF signalling can also occur independent

    of Smad activation, commonly referred to as Smad-independent or non-canonical TGF signalling

    (Moustakas and Heldin, 2005). These include activation of MAP kinase pathway, as well as

    phosphatidylinositol-3-kinase/protein kinase B pathway (AKT) (Zhang, 2009). Notably, another

    non-canonical TGF pathway that has been implicated in TGF-mediated loss of tight junctions

    and loss of cell polarity during EMT, is the Par6/Smurf1 polarity pathway (Ozdamar et al., 2005).

    In the TGF-Par6 polarity pathway, TGF ligand binding to the type II receptor induces the direct

    association and subsequent phosphorylation of polarity protein Par6 at the intracellular tight

    junctions (Bose and Wrana, 2006; Ozdamar et al., 2005). As mentioned previously, Par6 is a

    regulator of epithelial cell polarity and tight junction integrity (Assemat et al., 2008). Following

    the direct phosphorylation of Par6 by TGFRII, Smad ubiquitination regulatory factor 1 (Smurf1),

    an E3 ubiquitin ligase, is recruited to interact with Par6. In turn, Smurf1 ubiquitinates the tight

    junction stabilizer protein GTPase RhoA, resulting in its targeted proteasomal degradation.

  • 25

    Considering RhoA plays a fundamental role in tight junction stabilization and formation, its

    degradation leads to disassembly of the actin cytoskeleton, dissolution of tight junctions and loss

    of apical-basolateral cell polarity, all events which typify EMT (Ozdamar et al., 2005). In addition

    to EMT, phosphorylation of Par6 via non-canonical TGF signalling has proven to be essential for

    cell migration and invasion, key characteristics of cancer progression. Particular studies in prostate

    cancer showed phosphorylated Par6 to form a complex with aPKC polarity component at the

    leading edge of membrane ruffles. Further, the use of PKC specific inhibitors interfered with the

    formation of this polarity complex, and prevented prostate cancer cell invasion (Mu et al., 2015).

    Congruently, studies in breast cancer showed the TGFβ-Par6 polarity pathway to regulate cancer

    metastasis; interference with Par6 signalling prevented TGFβ induced loss of polarity in mammary

    cells grown in 3D structures (Viloria-Petit et al., 2009). In addition, suppression of Par6 in an in

    vivo orthotopic mouse model induced formation of ZO-1 positive epithelium within the tumour

    whilst supressing lung metastasis (Viloria-Petit et al., 2009). This further highlights the

    involvement of non-canonical TGF signalling via polarity protein Par6, and its importance not

    only to EMT but to other cellular processes regulated by TGF including cell migration and

    invasion.

    It is evident that both canonical and non-canonical TGFβ signalling pathways are involved in

    TGFβ regulation over multiple cellular processes. In some cases, activation of two different TGFβ

    signalling arms can result in the same effect. For instance, induction of EMT that is achieved by

    the non-canonical TGFβ/Par6 polarity pathway via RhoA degradation, can also be the result of

    canonical TGFβ signalling where Smad2/3 promotes the transcription of genes involved in EMT,

    such as Snail (Peinado et al., 2003). On the other hand, TGFβ ligands have the ability to induce

    different TGFβ signalling arms, and thus variable downstream pathways, which can account for

  • 26

    the vast multifunctionality of these growth factors, as well as explain the conflicting reports which

    show TGFβ to promote opposing effects in the same tissues/cells. This is particularly relevant to

    the human placenta, where different TGFβ isoforms have been implicated as negative regulators

    of trophoblast cell migration and invasion (Caniggia et al., 1999; Graham and Lala, 1991, 1992;

    Karmakar and Das, 2002; Tse et al., 2002), as well as positive regulators of trophoblast migration

    and proliferation (Xu et al., 2016). This latter role of TGF is in line with the positive effect of

    TGFβ on cell migration and invasion observed in many cancers.

    1.3.3 Role of TGFβ Signalling in Trophoblast Differentiation

    TGFβ regulation of trophoblast cell differentiation in the human placenta occurs via both the

    canonical (Smad mediated) and non-canonical signalling (Par6/Smurf1 mediated) (Xu J et al 2016,

    Sivasubramaniyam et al 2013) (Figure 1.4). Furthermore, this regulation during placentation

    occurs in a temporal and spatial manner. In regards to the canonical TGFβ signalling, receptor

    activated Smad2 is found particularly expressed in ST, where studies using explants and BeWo

    cells showed it to negatively regulate trophoblast cell fusion via downregulation of fusion

    regulators GCM-1 and ERVW-1 (Xu et al., 2016). Receptor activated Smad2 is also found in

    proliferating EVTs, and in vitro studies using JEG3 cells showed it to positively regulate

    trophoblast cell proliferation as elucidated by the upregulated expression of cell cycle regulators

    CCNE1 and CDK4 (Xu et al., 2016). As gestation progressed, ST levels of pSmad2 decreased, and

    levels of inhibitory Smad7 increased, consistent with increased rates of cell fusion observed with

    advancing gestation. Concurrently, decreasing pSMAD2 and increasing SMAD7 levels were seen

    toward the distal end of the EVT column (Xu et al., 2016). This is consistent with the

    differentiation of EVTs from a proliferative to a migratory and invasive phenotype with increasing

  • 27

    gestation. The differential Smad expression patterns seen here highlight the fact that canonical

    TGFβ signalling plays a distinct role in different trophoblast subtypes.

    Concerning the non-canonical TGFβ pathway, Par6/Smurf1 signalling was observed to be

    activated at 10-12 weeks of gestation in EVTs located in the intermediate region of anchoring

    column (Xu et al., 2016). Studies using JEG3 cells revealed that TGFβ treatment increases the

    association of Par6 and Smurf1 and promotes trophoblast cell migration in vitro via dissolution of

    tight junctions (Xu et al., 2016). This prompted the conclusion that trophoblast cell migration is

    regulated by the TGFβ-Par6 polarity pathway. Additionally, this study also observed Par6/Smurf1

    association in CT at 10-12 weeks of gestation, a time when CT are known to undergo fusion into

    ST, suggesting that TGFβ-Par6 signalling also mediates trophoblast fusion. These findings

    underscore the fundamental role that cell polarity plays in the human placenta, particularly in the

    proper functioning and differentiation of trophoblast cells. However, aside from the

    aforementioned findings on polarity protein Par6 from our group, no studies have investigated the

    role of other cell polarity proteins and signalling events on trophoblast cell differentiation. Further,

    no studies have looked at the impact of TGFβ on the functionality of other polarity proteins, or

    scaffolding proteins, in the placenta.

    This section has elucidated to the complexity of trophoblast differentiation during human placenta

    development, and thus has underscored the grave importance of its precise regulation. In fact,

    dysregulation of these differentiation processes is implicated in the pathogenesis of placenta

    related diseases such as preeclampsia.

  • 28

    Figure 1.4 Role of Smad-dependent and Par6-mediated TGFβ signalling in trophoblast cell

    differentiation

    TGFβ regulates trophoblast cell fate via canonical (Smad-dependent) and non-canonical (Smad-

    independent) signalling pathways. (A) In the Smad-dependent TGFβ pathway, TGFβ ligand

    binding to its tetrameric receptor complex promotes the phosphorylation and activation of Smad2,

    which complexes together with Smad4 to collectively translocate into the nucleus and may

    activate/repress target gene transcription that favor trophoblast cell proliferation over cell fusion.

    (B) In the Par6 mediated TGFβ pathway, TGFβRII can directly promote the phosphorylation of

    polarity protein Par6, which recruits Smad ubiquitination regulator factor (Smurf1) to selectively

    ubiquitinate (u) small GTPase protein RhoA for targeted proteasomal degradation. RhoA

    degradation results in tight junction dissolution leading to a loss in apical-basolateral cell polarity

    and promotion of cell motility events, such as trophoblast cell migration (Bose and Wrana, 2006;

    Ozdamar et al., 2005; Wrana, 2013).

  • 29

    1.4 Preeclampsia

    Preeclampsia (PE) is a serious hypertensive disorder that affects 5-8% of all pregnancies, and is

    associated with increased maternal and perinatal mortality worldwide (American College of et al.,

    2013). According to the current American College of Obstetricians and Gynecologists (ACOG)

    guidelines, a diagnosis of PE is given when the blood pressure of a previously normotensive

    mother is greater than or equal to 140 mmHg (systolic) and 90mmHg (diastolic) and there is new

    onset of one of the following events: proteinuria, thrombocytopenia, renal insufficiency, impaired

    liver function, pulmonary edema or visual disturbances (American College of et al., 2013). Any of

    these conditions pose a great deal of stress on both the mother and offspring, and due to the lack

    of treatment for PE, the solution is typically pre-term delivery. Although the clinical symptoms of

    PE typically resolve upon delivery, there are often significant long-term health consequences for

    both mother and offspring. Women who develop PE are at double the risk of developing

    cardiovascular or cerebrovascular diseases, and three times the risk of chronic hypertension

    (Goffin et al., 2018). Offspring of preeclamptic mothers also possess an increased risk for stroke

    later in life (Kajantie et al., 2009), and show significantly increased blood pressure in childhood

    and adulthood (Davis et al., 2012). In serious cases, immediate fetal complications include

    cognitive and physical impairments such as cerebral palsy, epilepsy and blindness. PE is

    subclassified into two distinct disorders depending on when clinical symptoms manifest during

    pregnancy: early-onset preeclampsia (E-PE), manifested prior to 34 weeks of gestation; and late-

    onset (L-PE), manifested after 34 weeks (Raymond and Peterson, 2011). Evidence suggests that

    the E-PE and L-PE have unique biological profiles and pathogenesis: E-PE originates from the

    placenta, and exhibits more serious clinical symptoms, whereas L-PE is maternal in origin, and

    results from an enhanced susceptibility of the maternal endothelium to react to pro-inflammatory

    factors and elicit an abnormal maternal response (Huppertz, 2008; Redman and Sargent, 2005). In

  • 30

    the present study, PE and E-PE are used interchangeably, unless otherwise stated. The placenta is

    considered an integral figure in the etiology of PE since in most cases, removal of the placenta is

    required for symptoms to regress. In pursuit of understanding how the placenta contributes to

    development of PE, several studies have showed that impaired trophoblast differentiation events

    during early development are central to the pathogenesis of this disease.

    1.4.1 Altered Trophoblast Differentiation in preeclampsia

    In PE placentae, extravillous trophoblast cells are arrested in an immature, proliferative phenotype

    (Redline and Patterson, 1995), and further exhibit shallow cell migration and invasion into the

    maternal decidua (Naicker et al., 2003; Robertson et al., 1985). Proliferative cytotrophoblast cells

    at the base of the anchoring column within the uterine wall retain their original “epithelial” like

    phenotype, and neglect to undergo the differentiation process which is required to invade the

    decidua (Fisher, 2015). This phenotype is demonstrated in one early study, where EVT from PE

    placentae failed to undergo integrin switching to express the invasive 51 integrin that is involved

    in EVT invasion in normal pregnancy (Zhou et al., 1993). Further, impaired ECM degradation by

    lowered levels and activity of MMP-9 in PE trophoblasts contributes to defective invasion (Lim et

    al., 1997). As a result, there is insufficient remodelling of the maternal spiral arteries, leading to a

    marked reduction in uteroplacental blood flow to the fetus. This reduction in placental blood flow,

    and increase in uteroplacental vascular resistance, in PE pregnancies can be observed via doppler

    ultrasound of the uterine arties (Harrington et al., 1996). Higher sensitivity analysis using MRI

    imaging has since confirmed this reduction in placental perfusion in E-PE placentae compared to

    normal pregnancies, which did not experience decreases in placental perfusion until later in

    gestation (Sohlberg et al., 2014). Reduced placental perfusion into the intervillous space ultimately

    leads to persistent uteroplacental hypoxia, a defining feature of PE (Soleymanlou et al., 2005).

  • 31

    Work from our lab implicated chronic hypoxia as a trademark of PE through microarray analyses,

    which revealed gene expression profiles of placental villous explants exposed to 3% O2, placentae

    from high-altitude residence, and PE placentae were strikingly similar (Soleymanlou et al., 2005).

    In turn, this resultant uteroplacental hypoxia can further exacerbate impaired trophoblast invasion

    and contribute to PE pathogenesis (Caniggia and Winter, 2002; Roberts and Cooper, 2001).

    To date, it is still not entirely clear what the molecular aberration is that leads to this impaired

    trophoblast differentiation and invasion seen in PE and thus investigations aimed at understanding

    this are ongoing. However, studies from our lab have attributed el