177
© 2013 Juanmahel Davila

© 2013 Juanmahel Davila - IDEALS

  • Upload
    others

  • View
    6

  • Download
    0

Embed Size (px)

Citation preview

Page 1: © 2013 Juanmahel Davila - IDEALS

© 2013 Juanmahel Davila

Page 2: © 2013 Juanmahel Davila - IDEALS

THE ROLE OF RAS-RELATED C3 BOTULINUM TOXIN SUBSTRATE 1 (RAC1)-

DEPENDENT PATHWAY IN THE REGULATION OF UTERINE FUNCTION DURING

EARLY PREGNANCY

BY

JUANMAHEL DAVILA

DISSERTATION

Submitted in partial fulfillment of the requirements

for the degree of Doctor of Philosophy in VMS - Comparative Biosciences

in the Graduate College of the

University of Illinois at Urbana-Champaign, 2013

Urbana, Illinois

Doctoral Committee:

Professor Indrani C. Bagchi, Chair and Research Director

Professor Emerita Janice M. Bahr

Professor Jodi A. Flaws

Professor Paul S. Cooke, University of Florida-Gainesville

Research Assistant Professor Quanxi Li

Page 3: © 2013 Juanmahel Davila - IDEALS

ii

ABSTRACT

During implantation, uterine stromal cells differentiate into specialized decidual cells, which

become secretory and support the growth of the embryo before placentation. At the same time,

new blood vessels begin to form in the maternal decidua from pre-existing vasculature. This

process termed angiogenesis is critical for the establishment of the placenta and the maintenance

of pregnancy. Although it is clear that decidualization and angiogenesis play a crucial role during

early pregnancy, the complex molecular pathways underlying these processes remain largely

unknown. Our recent studies revealed that RAC1 (Ras-related C3 botulinum toxin substrate 1), a

pleiotropic signaling factor belonging to the Rho family of GTPases, is induced in the uterine

stroma during decidualization and angiogenesis, suggesting a role for RAC1 in these critical

processes. To address the role of RAC1 during pregnancy, we created a conditional knockout of

the Rac1 gene in the uterus by crossing mice carrying floxed Rac1 with PgrCre mice, thereby

generating the conditional mutation termed as Rac1d/d. A six-month breeding study indicated a

severe defect in fertility of the Rac1d/d females. Further analysis revealed that while embryo

attachment appeared to be unaffected, differentiation and secretory functions of decidual cells were

impaired in Rac1d/d uteri. This abnormality of decidual cells led to multifactorial consequences,

including compromised uterine vascular remodeling and angiogenesis, maternal blood vessel

hemorrhage and placental abnormalities resulting in embryo loss around days 10 to 12 of

pregnancy. Fetuses that survived to day 15 of pregnancy showed indications of intrauterine growth

restriction. Based on the results obtained in this dissertation, we propose that RAC1 regulates

stromal differentiation and secretory functions, which produce and secrete critical local factors

that regulate vascular remodeling at the maternal-fetal interface and promote proper formation of

the placental disk. Therefore, RAC1 induced in the uterine stroma during decidualization plays a

critical role in maintaining proper function of the placental vascular bed and supporting embryo

growth and development during early and mid-stages of pregnancy.

Page 4: © 2013 Juanmahel Davila - IDEALS

iii

To Aurora, Mom, Dad, and Brother

Page 5: © 2013 Juanmahel Davila - IDEALS

iv

ACKNOWLEDGEMENTS

This dissertation would have never been possible without the generous help and support of many

people that I have encounter through my time in Illinois.

I will like to start by expressing my most sincere and deepest appreciation to my current and former

research advisors and mentors, Drs. Indrani Bagchi, Milan Bagchi and Paul Cooke. I owe great

debt of gratitude to these people because thanks to their patient guidance, continuous support,

constant challenge and encouragement, and mentorship, I have reached this far. All three have

been extremely instrumental in guiding me along the many challenges that were presented to me

during my academic journey, and have been key elements in my personal and scientific growth,

as well as achievements. I am also appreciative for the support of my former (Drs. William

Helferich and O. David Sherwood) and current (Drs. Janice M. Bahr, Jodi A. Flaws, and Quanxi

Li) members of my doctoral committee for their continuous support, motivation, and guidance

throughout these years. I have been very fortunate to have these three wonderful human beings as

pillars of my academic learning.

I am extremely thankful to all the people with whom I had interactions at the University of Illinois.

The former members of the Cooke laboratory (Drs. Liz Simon, Melissa Cimafranca,

Santhipriyadarsini Sridharan, Gaurav Tyagi, Heather Schlesser, Jaspreet Kaur, Mr. Daryl Meling,

and Ms. Gail Ekman), which were instrumental in my early scientific carrier providing me with

technical support and training, as well personal support and friendship. The former and current

members of the Bagchi laboratories, especially Hatice Kaya, Janelle Mapes, Sandeep Pawar, Dr.

Yuechao Zhao, Dr. Shanmugasundaram Nallasamy, and Alison Hantak, which have been also

supportive in my latest stages of scientific growth, as well as providing me with their friendship. I

will like to express a special appreciation to Drs. Mary J. Laws, Quanxi Li, and Athilakshmi

Kannan, my colleagues and dearest friends, who provided me with extensive support, mentorship,

assistance, and guidance during my transition and establishment to the Bagchi laboratory. Also, a

special thanks to all the wonderful past and current colleagues of the Department of Comparative

Biosciences, especially Dr. Denise Archambeault, Dr. Amrita Das, Thushara Chakkath, Snehita

Sri Varma, Joseph Cacioppo, Dr. Megan Mahoney, Dr. Leslie McNeil, Dr. CheMyong “Jay” Ko,

Page 6: © 2013 Juanmahel Davila - IDEALS

v

Dr. Jing Yang, Dr. Duncan Fergusson, Dr. David Bunick, Dr. Rex Hess, Dr. Marie-Claude

Hofmann, Dr. Melissa Clark, Dr. Zelieann Craig, Dr. Ju Lan Chun, Dr. Thomas Garcia, Patrick

Hannon, and Dr. Jackye Peretz, for a supportive, fun, and educational work environment. Finally,

I most express my gratitude to the administrative support of the former and current members of

the main office of the Department of Comparative Biosciences, with special attention to Carrol

Bunick, Carla Manuel, Todd Blazaitis, Cindy Dillman, and Mona Nordbrock.

I will like to extend my appreciation to the principal investigator and members of the laboratories

of Drs. Milan Bagchi, Duncan Ferguson, David Bunick, Marie-Claude Hofmann and Rex Hess

(former CB investigators), Jodi Flaws, and Jing Yang, for technical assistant and for allowing me

to use critical instrument that made part of my work possible; specifically acknowledging Dr.

Zelieann Craig for assistance with ELISA (Flaws’ Lab); Hatice Kaya for assistance with western

blots and microarray analysis (M. Bagchi’s Lab); Drs. Zhigang Jin, Jin Wei "Kenneth" Chung, R.

Tyler Schwend for assistance with microscope and western blots (Yang’s Lab); Dr. Thomas Garcia

for assistance in microscopy and fluorescent imaging (Hofmann’s Lab); and David Bunick for

assistance with quantitative real-time PCR. Also, I will like to thank the histology laboratory,

especially Karen Doty, for technical assistance with immunostaining.

Last but not least, I will like to thank my family and closest friends (Jeff Brown, Sylvia Choi, and

Ranjani Murali)! Which without their continuous support, encouragement, and confidence in me,

I would not be able to accomplish my career goals and achievements. I am extremely gratefully to

my brother Isamac Dávila, my mother Zelithez Lorenzo, my father Juan Dávila, my best friend

Emanuel Rivera, as well as my closest relatives, who inspired and pushed me to pursue my dreams

and to never give up without trying. I am in greatest debt to my loving fiancé Aurora M. Cruz,

who motivated, inspired, and supported me throughout all these years, as well as been one of my

strongest pillars in the pursuit of my doctoral degree. I am also, thankful to my new family

members, Wanda Torres, Andres Cruz (deceased), Andy Cruz, and Zoriann Cruz, for their

continuous encouragement and motivation.

Page 7: © 2013 Juanmahel Davila - IDEALS

vi

TABLE OF CONTENT

LIST OF FIGURES AND TABLES ........................................................................................... ix

LIST OF ABBREVIATIONS .................................................................................................... xii

CHAPTER 1: INTRODUCTION ................................................................................................ 1

CHAPTER 2: REVIEW OF RELEVANT LITERATURE ...................................................... 4

2.1 DEFINING THE PROBLEM: CLINICAL EVIDENCE FOR RECURRENT MISCARRIAGE AND PREGNANCY

LOSS ................................................................................................................................................. 5

2.1.1 FACTORS THAT MAY CONTRIBUTE TO MISCARRIAGE .............................................................. 5

2.1.1.1 CONGENITAL ABNORMALITIES ....................................................................................... 6

2.1.1.2 VASCULAR PROBLEMS AND BLEEDING DISORDERS ........................................................ 6

2.1.2 INTRAUTERINE GROWTH RESTRICTION ................................................................................... 7

2.1.3 ASSISTED REPRODUCTIVE TECHNOLOGIES .............................................................................. 9

2.2 UTERINE RECEPTIVITY, THE WINDOW OF IMPLANTATION, AND EARLY PREGNANCY IN THE MOUSE

......................................................................................................................................................... 9

2.2.1 CRITICAL EVENTS DURING PREGNANCY ................................................................................ 10

2.2.1.1 IMPLANTATION ............................................................................................................. 10

2.2.1.2 DECIDUALIZATION: A KEY EVENT FOR SUCCESSFUL ESTABLISHMENT OF PREGNANCY ..

...................................................................................................................................... 11

2.2.1.3 ANGIOGENESIS AND VASCULAR REMODELING ARE ESSENTIAL FOR MAINTENANCE OF

PREGNANCY ................................................................................................................................... 13

2.2.1.4 PLACENTATION ............................................................................................................. 17

2.2.1.4.1 PLACENTATION IN HUMANS AND MICE .................................................................... 17

2.2.1.4.2 REGULATION OF TROPHOBLAST INVASION AND PLACENTAL PATHOLOGIES ........... 18

2.2.1.4.3 THE MID-STAGES OF PLACENTATION IN THE MOUSE ................................................ 19

2.3 RHO GTPASES ................................................................................................................................ 19

2.3.1 THE RHO GTPASES ............................................................................................................... 20

2.3.2 ROLE OF RHO GTPASES IN ACTIN DYNAMICS ....................................................................... 20

2.3.2.1 FUNCTIONS BEYOND ACTIN DYNAMICS ........................................................................ 21

2.3.3 REGULATORS OF THE RHO GTPASES .................................................................................... 21

2.3.4 RHO GTPASES AND DISEASE ................................................................................................. 23

2.3.5 RAS-RELATED C3 BOTULINUM TOXIN SUBSTRATE 1 (RAC1) ............................................... 23

2.3.6 AVAILABLE MOUSE MODELS TO STUDY RAC1 FUNCTION .................................................... 26

Page 8: © 2013 Juanmahel Davila - IDEALS

vii

2.4 THE AIMS OF THE DISSERTATION ................................................................................................... 27

2.5 CHAPTER FIGURES ......................................................................................................................... 28

CHAPTER 3: GENERATION AND PHENOTYPIC ANALYSIS OF RAC1D/D MUTANT

FEMALE MICE.......................................................................................................................... 38

ABSTRACT............................................................................................................................................... 39

INTRODUCTION ....................................................................................................................................... 40

MATERIALS AND METHODS .................................................................................................................... 42

RESULTS AND DISCUSSION ..................................................................................................................... 48

RAC1 IS EXPRESSED IN UTERINE STROMA DURING DECIDUALIZATION, BUT ITS EXPRESSION IS

INDEPENDENT OF STEROID HORMONES .............................................................................................. 48

THE RAC1D/D MOUSE MODEL ............................................................................................................... 49

CONDITIONAL DELETION OF UTERINE RAC1 LEADS TO SEVERE SUBFERTILITY ................................. 49

CONDITIONAL DELETION OF RAC1 HAS NO EFFECT ON OVARIAN FUNCTION ..................................... 50

THE SEVERE SUBFERTILITY IN RAC1D/D IS DUE TO MATERNAL BUT NOT EMBRYONIC DEFECT ........... 52

CHAPTER FIGURES AND TABLES ............................................................................................................ 54

CHAPTER 4: RAC1 IS A CRITICAL REGULATOR OF UTERINE ANGIOGENESIS

DURING EARLY PREGNANCY IN THE MOUSE .............................................................. 78

ABSTRACT............................................................................................................................................... 79

INTRODUCTION ....................................................................................................................................... 80

MATERIALS AND METHODS .................................................................................................................... 82

RESULTS AND DISCUSSION ..................................................................................................................... 87

MICROARRAY ANALYSIS REVEALS IMPAIRED ANGIOGENIC NETWORK IN RAC1-ABLATED UTERI ..... 87

ABLATION OF RAC1 IN THE UTERUS LEADS TO A DECREASED SECRETION OF PRO-ANGIOGENIC

FACTORS ............................................................................................................................................. 89

CONDITIONAL DELETION OF RAC1 IN THE UTERI LEADS TO SEVERE HEMORRHAGE .......................... 90

CHAPTER FIGURES .................................................................................................................................. 92

CHAPTER 5: CONCLUSIONS AND WORKING MODEL ............................................... 101

CHAPTER FIGURE ..................................................................................................................... 105

CHAPTER 6: FUTURE DIRECTIONS ................................................................................. 106

REFERENCES .......................................................................................................................... 109

Page 9: © 2013 Juanmahel Davila - IDEALS

viii

APPENDIXES ........................................................................................................................... 134

APPENDIX A: BREEDING STRATEGIES FOR THE FOUNDER AND EXPERIMENTAL COLONY

..................................................................................................................................................... 135

APPENDIX B: GENE EXPRESSION VALIDATION OF OTHERS MOLECULES INVOLVED IN

STROMAL CELL DECIDUALIZATION AND ANGIOGENESIS ........................................... 141

APPENDIX C: PRELIMINARY DATA FOR FUTURE DIRECTIONS ............................................ 143

C1. BACKGROUND INFORMATION AND THE RATIONALE................................................................. 143

C2. MATERIALS AND METHODS ...................................................................................................... 145

C3. PRELIMINARY DATA .................................................................................................................. 148

APPENDIX D: GENOTYPING PRIMERS AND PROTOCOL .......................................................... 156

APPENDIX E: ANTIBODY DILUTIONS .......................................................................................... 158

APPENDIX F: PRIMER SEQUENCES FOR GENE EXPRESSION ASSAYS .................................. 160

Page 10: © 2013 Juanmahel Davila - IDEALS

ix

LIST OF FIGURES AND TABLES

FIGURE 2.1: HORMONE PROFILE DURING PREGNANCY IN MICE. ...................................................... 28

FIGURE 2.2: COMPARISON OF THE EARLY EVENTS OF PREGNANCY IN HUMANS AND MICE.............. 29

FIGURE 2.3: MORPHOLOGICAL CHANGES DURING STROMAL CELL DIFFERENTIATION..................... 30

FIGURE 2.4: MAJOR EVENTS DURING IMPLANTATION AND DECIDUALIZATION. .............................. 32

FIGURE 2.5: DEVELOPMENTAL STAGES OF THE MOUSE PLACENTA. ................................................ 33

FIGURE 2.6: STRUCTURAL COMPARISON BETWEEN MEMBERS OF THE HUMAN RHO FAMILY OF

GTPASES. ............................................................................................................................... 34

FIGURE 2.7: REGULATORY MECHANISMS OF THE RHO FAMILY OF GTPASES. ................................ 35

FIGURE 2.8: PROTEIN SEQUENCE COMPARISON BETWEEN HUMAN AND MOUSE RAC1. .................. 36

FIGURE 3.1: BREEDING SCHEME TO GENERATE THE CONDITIONAL MUTANT FOR RAC1 USING CRE-

LOXP STRATEGY. .................................................................................................................... 54

FIGURE 3.2: EXPRESSION PROFILE FOR RAC1 GENE AND PROTEIN IN MICE. .................................... 56

FIGURE 3.3: RAC1 IS EFFICIENTLY DELETED FROM THE UTERUS USING PGRCRE. ............................ 58

TABLE 3.1: IMPACT OF RAC1 DELETION ON FEMALE FERTILITY ...................................................... 60

FIGURE 3.4: CONDITIONAL DELETION OF RAC1 DOES NOT AFFECT OVARIAN FUNCTION. ................ 61

FIGURE 3.5: CONDITIONAL DELETION OF RAC1 HAS DOES NOT PREVENT EMBRYO ATTACHMENT AND

INVASION INTO THE ENDOMETRIUM. ....................................................................................... 63

FIGURE 3.6: CONDITIONAL DELETION OF UTERINE RAC1 DOES NOT AFFECT THE FORMATION OF THE

DECIDUOMA, BUT DISPLAYS DYSREGULATION IN ALPL ACTIVITY. ........................................ 65

FIGURE 3.7: CONDITIONAL DELETION OF RAC1 LEADS TO A DYSREGULATED STROMAL CELL

DIFFERENTIATION. .................................................................................................................. 66

FIGURE 3.8: ACTIVATION OF THE DECIDUAL CELL RAC1 PATHWAY IS NECESSARY FOR STROMAL

CELL MORPHOLOGY AND NORMAL DIFFERENTIATION. ............................................................ 70

FIGURE 3.9: REPRESENTATIVE UTERI SAMPLES AT DIFFERENT STAGES OF PREGNANCY. ................ 71

FIGURE 3.10: SEVERE HEMORRHAGE IS OBSERVED IN IMPLANTATION NODULES FROM RAC1D/D

UTERI. ..................................................................................................................................... 72

TABLE 3.2: SINGLE OR EPITHELIAL-SPECIFIC RAC1 ALLELE DELETION DOES NOT IMPACT FERTILITY

............................................................................................................................................... 75

FIGURE 3.11: PREGNANCY DEFECT OBSERVED IN RAC1 CONDITIONAL-NULL UTERI IS NOT DUE TO

AN EMBRYONIC DEFECT. ......................................................................................................... 76

Page 11: © 2013 Juanmahel Davila - IDEALS

x

FIGURE 4.1: BIOLOGICAL PROCESS LIST OF GENES THAT WERE DOWN-REGULATED LESS THAN 1.5

FOLD ON MICROARRAY. .......................................................................................................... 92

FIGURE 4.2: CONDITIONAL DELETION OF RAC1 IN THE UTERUS LEADS TO DOWN-REGULATION OF

SEVERAL GENES THAT ARE CRITICAL FOR UTERINE ANGIOGENESIS. ........................................ 93

FIGURE 4.3: EXPRESSION OF TWO WELL-KNOWN GENE TARGETS OF RAC1 ARE UNAFFECTED. ...... 94

FIGURE 4.4: SUPPRESSION OF THE RAC1 PATHWAY COMPROMISES THE FORMATION OF THE

DECIDUAL VASCULAR PLEXUS. ............................................................................................... 95

FIGURE 4.5: CONDITIONAL DELETION OF UTERINE RAC1 IMPAIRS ANGIOGENESIS. ......................... 97

FIGURE 4.6: CONDITIONAL DELETION OF RAC1 IMPAIRS VEGFA TRANSPORT AND SECRETION OF

UTERINE ENDOMETRIAL CELLS. .............................................................................................. 98

FIGURE 4.7: RAC1D/D UTERI DISPLAY EXTRAVASATION OF RED BLOOD CELLS. ................................ 99

FIGURE 4.8: DECIDUA FROM RAC1D/D MICE SHOW SEVERE BLOOD HEMORRHAGE IN THE LATERAL

BLOOD SINUSES. ................................................................................................................... 100

FIGURE 5.1: WORKING MODEL FOR THE ROLE OF RAC1 IN THE UTERUS DURING EARLY EVENTS OF

PREGNANCY. ......................................................................................................................... 105

FIGURE A.1: BREEDING STRATEGY FOR GENERATING THE RAC BREEDING COLONY. ................... 135

FIGURE A.2: EXPECTANCY FREQUENCIES OF ANIMALS/GENOTYPE BASED ON THE BREEDING

STRATEGY FOR GENERATING THE FOUNDING ANIMAL COLONY. ............................................ 136

FIGURE A.3: BREEDING STRATEGY FOR GENERATING THE EXPERIMENTAL ANIMALS ................... 137

FIGURE A.4: EXPECTANCY FREQUENCIES OF ANIMALS/GENOTYPE BASED ON THE BREEDING

STRATEGY FOR GENERATING THE BREEDING COLONY. .......................................................... 138

FIGURE A.5: BREEDING STRATEGY FOR THE SIX-MONTH BREEDING STUDY. ................................ 139

FIGURE A.6: EXPECTANCY FREQUENCIES OF ANIMALS/GENOTYPE BASED ON THE SIX-MONTH

BREEDING STUDY. ................................................................................................................. 140

FIGURE B.1: GENE EXPRESSION OF OTHER CRITICAL MOLECULES, IMPLICATED IN STROMAL CELL

DIFFERENTIATION, WERE NOT AFFECTED BY THE CONDITIONAL RAC1 MUTATION. ............... 141

FIGURE B.2: GENE EXPRESSION OF OTHER MOLECULES IMPLICATED IN UTERINE ANGIOGENESIS. 142

FIGURE C3.1: CONDITIONAL DELETION OF RAC1 IN UTERINE DECIDUAL CELLS, DOES NOT PREVENT

EMBRYO IMPLANTATION, BUT LEADS TO TROPHOBLAST OVERGROWTH IN VIVO. ................... 148

FIGURE C3.2: GENE EXPRESSION PROFILE OF REGULATORS OF TROPHOBLAST EXPANSION. ......... 150

Page 12: © 2013 Juanmahel Davila - IDEALS

xi

FIGURE C3.3: CONDITIONAL DELETION OF RAC1 IN UTERINE TISSUES LEADS TO PLACENTAL

ABNORMALITIES AND INDICATIONS OF INTRAUTERINE GROWTH RESTRICTION. .................... 151

FIGURE C3.4: CONDITIONAL DELETION OF RAC1 IN UTERINE TISSUES LEADS TO PLACENTAL

ABNORMALITIES THAT DISPLAY IMPAIRED VASCULAR REMODELING. ................................... 153

Page 13: © 2013 Juanmahel Davila - IDEALS

xii

LIST OF ABBREVIATIONS

ADM Adrenomedullin

ALPL Liver/kidney/bone alkaline phosphatase

AM Anti-mesometrial decidua

ANGPT1 Angiopoietin 1

ANGPT2 Angiopoietin 2

ANGPT4 Angiopoietin 4

ANGPTs Angiopoietins

ANOVA Analysis of variance

ART Assisted reproductive technologies

ATP Adenosine triphosphate

BLAST Basic local alignment search tool

BMP2 Bone morphogenetic protein 2

BZ Basal zone

CDC42 Cell division control protein 42 homolog

CLDN3 Claudin 3

CS Circular smooth muscle

CZ Compact zone

DAVID Database for annotation visualization and integrated discovery

DC Decidual zone

E Embryo

E2 Estrogen (17β-estradiol )

ECM Extracellular matrix

ELISA Enzyme-linked immunosorbent assay

EPAS1/HIF2α hypoxia inducible factor 2α

ESR1/ERα Estrogen receptor α

F13A1 Coagulation factor XIII subtype A1

GAP GTPase-activating proteins

GDI Guanine nucleotide-dissociation inhibitors

GDP Guanosine diphosphate

GEF Guanine nucleotide exchange factors

GJP1/CX43 Connexin 43

GTP Guanosine triphosphate

GTPases Guanosine triphosphatases

HBSS Hank's balanced salt solution

Page 14: © 2013 Juanmahel Davila - IDEALS

xiii

HIF1α hypoxia inducible factor 1α

HIFs hypoxia inducible factors

HRP Horseradish peroxidase

IGF1 Insulin-like growth factor 1

IGF2 Insulin-like growth factor 2

IGFBP1 IGF binding protein 1

IGFBP4 IGF binding protein 4

IGFBPs IGF binding proteins

IGFs Insulin-like growth factors

IgG Immunoglobulin G

IUGR Intrauterine growth restriction

MKI67 Proliferation-related MKi-67 antigen

L Lumen

LDZ Lateral decidual zone

LS Lateral blood sinusoids (also known as vascular web)

LSMeans Least square means

M Mesometrial decidua

MG Metrial gland

MMP9 Matrix metalloproteinase 9

MMPs Matrix metalloproteinases

MT Mesometrial triangle

MY Myometrium

NBF Neutral-buffered formalin

NCBI National center for biotechnology information

NRP1 Neuropilin 1

P4 Progesterone

PBR Polybasic region

PBS Phosphate buffered saline

PECAM1/CD31 Platelet endothelial cell adhesion molecule 1

PGR Progesterone receptor

PI3K Phosphoinositide 3-kinase

PIP5K Phosphatidyl-dependentinositol-4-phosphate5-kinase

PREX1 PI3K-dependent Rac exchange factor

PRL3C1/PLPJ Prolactin family 3, subfamily c, member 1; PLP-J

PRL8A2/dPRP Prolactin family 8, subfamily a, member 2; decidual prolactin-related

protein

Page 15: © 2013 Juanmahel Davila - IDEALS

xiv

qPCR Quantitative real-time PCR

RAC1 Ras-related C3 botulinum toxin substrate 1

RAC1d/d Mice containing a conditional deletion for Rac1 gene at specific sites

where PGR is expressed

RAC1f/f Mice containing Rac1 gene flanked by LoxP sites

RAC2 Ras-related C3 botulinum toxin substrate 2

RAC3 Ras-related C3 botulinum toxin substrate 3

rER Rough endoplasmic reticulum

RHO Ras homologs

RHOA Ras homolog gene family, member A

RUNX1 Runt-related transcription factor 1

S.E.M Standard error of the mean

siRNA Small interfering RNA

SLC2A4/GLUT4 Glucose transporter type 4

SPHK1 Sphingosine kinase 1

TEK/TIE2 ANGPT 1/2 receptor

TIMPs Tissue inhibitors of MMPs

VEGF(A) Vascular-endothelial growth factor (A), also referred to as VEGF

VEGFR1/FLT1 VEGF receptor 1

VEGFR2/FLK1 VEGF receptor 2

WNT4 Wingless-related murine mammary tumor virus integration site 4

WNT5A Wingless-related murine mammary tumor virus integration site 5a

WNT7A Wingless-related murine mammary tumor virus integration site 7a

Page 16: © 2013 Juanmahel Davila - IDEALS

1

CHAPTER 1: INTRODUCTION

Page 17: © 2013 Juanmahel Davila - IDEALS

2

Implantation is a fundamental biological process during which the embryo attaches to the uterine

epithelium, invades the underlying stroma and promotes differentiation of the stromal cells to

decidual cells, which support embryonic growth and survival. It is a critical step for successful

establishment of pregnancy [1-11]. In humans, infertility is one of the most common disturbances

of reproductive health, with 10-15% of couples finding it difficult or impossible to conceive. [4,

5, 8, 12-16]. Despite significant advances in assisted reproductive technologies (ART), many

couples experience infertility as a result of failed implantation of the fertilized embryos into the

uterus and subsequent loss of these embryos. The implantation rates in ART remain low, even with

high-quality embryos, pointing to the importance of this process as a major cause of pregnancy

failure and infertility [1-3, 15, 17-21]. Understanding the signaling mechanisms central to embryo

implantation has the potential to alleviate many problems associated with infertility and improve

the clinical success of increasingly used ART.

This dissertation focuses on the newly identified role of RAC1 (Ras-related C3 botulinum toxin

substrate 1), a pleiotropic signaling molecule, in endometrial function during embryo implantation.

Elucidating the role of RAC1 signaling in the uterus may identify molecular targets for treating

infertility and improving the outcome of ART.

Implantation is a fundamental biological process during which the embryo comes into intimate

contact with the uterine epithelium, invades the stroma and establishes the placenta [1-5, 8-10, 22-

28]. This complex multistage process is pivotal for successful establishment of pregnancy. In

rodents and humans, as the embryo breaches the luminal epithelium following attachment, the

underlying stromal cells undergo a dramatic transformation to form a specialized tissue, known as

the decidua [5, 7-9, 22-25]. During this process, known as decidualization, fibroblastic stromal

cells proliferate and then differentiate into the secretory decidual cells that support embryo growth

and survival until placentation ensues. The decidual tissue also undergoes extensive remodeling to

control embryo invasion and accommodate the growing embryo. As the decidua forms around the

implanted embryo, the uterine endothelial cells proliferate to create an extensive vascular network,

which is embedded in the decidua and critical for embryo development [23, 29-33]. Even though

it is clear that decidualization is a key event in establishment of early pregnancy, the signaling

molecules that participate in the formation and function of this tissue remain poorly understood.

Page 18: © 2013 Juanmahel Davila - IDEALS

3

To gain an insight into the signaling pathways that are involved in the decidualization process, we

performed gene expression profiling of RNA collected from mouse uteri before and after decidual

stimulation. Our studies revealed that the gene encoding the Rac1, a pleiotropic signaling

molecule, is induced in uterine stroma at the time of decidualization. RAC1 is a member of the

Rho family of GTPases that regulate a wide range of cellular processes, including cell

proliferation, differentiation, cell-cell adhesion, endothelial cell function, and cell motility via actin

cytoskeleton remodeling. Because decidualization encompasses many of these physiological

events, it is possible that RAC1 regulates some of these processes during early pregnancy. In

support of this notion, a recent study has shown that invasion of the human embryo into

endometrial stroma in vitro involves motility of human endometrial stromal cells and that this

motility is dependent on the activity of the Rho GTPase RAC1 [7, 26].

To address the role of RAC1 in vivo during decidualization, we employed a loss-of-function

approach using genetically engineered mice. In this project, we generated a mutant mouse model

in which Rac1 gene is ablated from uterine stroma, characterized the mouse model and identified

the pathways that mediate RAC1 function in the uterus during decidualization. An in depth

analysis of this new mouse model is predicted to improve our understanding of the cause and

consequence of pregnancy complications involving impaired implantation, dysregulated

decidualization and placental complications in the human.

Page 19: © 2013 Juanmahel Davila - IDEALS

4

CHAPTER 2: REVIEW OF RELEVANT LITERATURE

Page 20: © 2013 Juanmahel Davila - IDEALS

5

2.1 Defining the problem: Clinical evidence for recurrent miscarriage and pregnancy loss

In recent years, worldwide, there has been a marked increase in human infertility, as well as the

use of assisted reproductive technologies (ART) [34-37]. The increased use of ART and the trend

towards low fertility has been attributed to the socioeconomic status, career demands in women,

improvement in contraception, or education, however biological factors that might contribute to

this decline need consideration [34]. In fact, clinically it is recognized that miscarriage and early

implantation failure are amongst the most common complications in pregnancy [6, 7, 26, 38, 39].

In many cases, this goes unnoticed because most miscarriages occur within the first 10 to 20 weeks

of gestation. Although pre-implantation pregnancy loss is considered to be normal in mammals as

an evolutionary advantage to select the best possible embryos, dysregulation of any of the events

before, during, or after implantation will lead to termination of pregnancy [1]. This concept has

created a challenge to researchers and clinicians who are trying to uncover ways to circumvent

human infertility, and at the same time, selecting for healthy offspring.

Recurrent miscarriage, which is defined clinically as pregnancy loss in more than three

reproductive cycles, is becoming more frequent in our society and in many cases (approximately

50%) the reasons leading to pregnancy loss remains unknown [40, 41]. The available data of a

large prospective register linkage study have shown that maternal age at conception is a strong risk

factor for miscarriage [42], and the risk of fetal loss increases steeply after the age of 35 years,

rising from 9% at 20–24 years to nearly 75% at 45 years and older [42-44]. Certainly there is an

urgent need to gain an understanding of the genetic- and molecular-basis of miscarriage, with

particular emphasis on the maternal environment that leads to pregnancy failure. A literature

review was undertaken to obtain current knowledge about certain maternal factors that may

contribute to miscarriage, as well as the influence of the uterine environment, and current

treatments for recurrent miscarriage.

2.1.1 Factors that may contribute to miscarriage

Recurrent miscarriage is clinically defined, by many clinicians, as the loss of three or more

consecutive pregnancies, and depending on how conservative the clinicians are when diagnosing

Page 21: © 2013 Juanmahel Davila - IDEALS

6

a patient with recurrent miscarriage, the incidence can affect anywhere from 1 to 5 % of couples

trying to conceive. It has been attributed to either genetic, structural, infections, endocrine,

immune, or unexplained causes [40, 44, 45], and in many cases, miscarriage is also associated with

psychological morbidity and stress for the patients. Very often women, more than men, attending

fertility clinics are clinically depressed, and about 20% of these women have anxiety levels that

are similar to those in psychiatric outpatient populations [17-20, 46, 47].

2.1.1.1 Congenital abnormalities

Although several known factors put some women at risk for miscarriage including malformation

of the uterus, diabetes, obesity, and alcohol use, the relationships between these conditions and

recurrent miscarriage have not been extensively studied and remain uncertain. Congenital uterine

abnormalities are commonly reported in women with recurrent miscarriage [48-51]. However,

some have argued that congenital uterine abnormalities might not represent a barrier anymore,

because there are women with structural abnormalities that have conceived children without

complications. The fact is that early miscarriages, especially the sporadic ones, are typically

associated with chromosome abnormalities of the embryo, which result in implantation failure or

growth arrest of the embryos leading to termination of pregnancy. Miscarriages without fetal

chromosomal abnormality or malformation are often thought to be a result of abnormal trophoblast

invasion and development [45, 52-54]. There is also evidence to suggest that impaired trophoblast

growth and invasion may be caused by an increased local or systemic immunological reaction to

the fetus or trophoblast [6]. In contrast to sporadic miscarriage, recurrent miscarriage tends to

occur even when the fetus has a normal chromosome complement, suggesting that the uterine

environment plays a role in determining pregnancy outcome. Therefore, recurrent miscarriage

undoubtedly is as complex disease, which may or may not have an underlying genetic component.

2.1.1.2 Vascular problems and bleeding disorders

It is well established that proper vascularization of decidua is critical for the success of pregnancy,

and it has been suggested that vascular disorders may play a role in pregnancies with complications

[55-58]. More recently, however, hypercoagulation disorders have been implicated in recurrent

Page 22: © 2013 Juanmahel Davila - IDEALS

7

pregnancy loss, which widens the scope of investigations and potential management to treat

recurrent miscarriage. However, data on the frequency of genetic thrombophilia are limited by the

small number of individual studies [44].

In some cases of early pregnancy failure, miscarriage has been attributed to the onset of a

precocious maternal blood flow leading to increased oxidative stress [52, 59]; and this is

independent of the karyotype of the conceptus [60]. Moreover, the early onset of maternal

circulation, which results in increased oxygen levels, can lead to widespread trophoblast damage

and placental degeneration [52, 59]. Furthermore, oxidative stress may modulate the architecture

of vasculature and the expression of angiogenic factors resulting in a dysregulated vascularization

[61]. Consistent with this observation, it has been reported that in women with miscarriages, there

is a reduced expression of vascular endothelial growth factor (VEGF) and its receptors [62].

Similarly, the decidual tissues of women with recurrent miscarriages show imbalance of the

expression ratio of angiopoietins (Angpt2/Angpt1), in favoring Angpt1, compared with the

predominance of Angpt2 in normal pregnancies [63]. While angiopoietin 2 is maximally expressed

during the early stages of pregnancy (i.e. first trimester) and declines afterwards, Angpt1

expression increases towards the third trimester, suggesting blood vessel stability [64]. Overall,

the data suggest that the role ANGPT2 during the first trimester is in branching angiogenesis,

leading to elongating and sprouting vessels and complex vascular network, whereas ANGPT1

predominates in the third trimester enhancing non-branching angiogenesis, maturation and

stabilization of the vascular network [58, 63, 64]. Therefore, a strong association between vessel

density and miscarriages is not farfetched.

2.1.2 Intrauterine growth restriction

Intrauterine growth restriction (IUGR) occurs with an incidence of 4 to 7% of all births, resulting

in low birth weight and preterm delivery. IUGR is associated with increased neonatal morbidity

and mortality because of placental problems leading to nutritional deficiencies [65, 66]. Several

risk factors have been associated with the pathogenesis of IUGR, which include chromosomal

abnormalities, infectious diseases, and defective vascularization of the uteroplacental unit.

Consistent with the latter, placental samples from women with IUGR showed a significantly lower

Page 23: © 2013 Juanmahel Davila - IDEALS

8

number of capillaries and a decreased capillary area in terminal villi of the placenta compared to

those with normal term pregnancies [67, 68]. Interestingly, previous studies have shown that in

pregnancies with IUGR, there is an apparent reduction in cytotrophoblast proliferation, poor

placental blood vessel development and an apparent increase in placental oxygen [52, 69, 70] [60].

Given that hypoxia promotes VEGF production, and thus angiogenesis, it has been suggested that

the relatively high oxygen levels in placentas with IUGR, will limit angiogenesis [69]. Consistent

with this notion, Khaliq et al. [71] reported a decreased expression of VEGF and an altered

expression of angiopoietins in IUGR placentas. The opposite is observed during the first trimester

of pregnancy where hypoxia upregulates VEGF secretion in the placenta. Given that trophoblasts

express the receptors for both VEGF and angiopoietins, which are known to influence behavior in

these cells, it is possible that a dysregulation of angiogenic factors can lead to perturbations in

proliferation and invasion of the trophoblast cells, as well as abnormal trophoblast migration and

differentiation (leading to placental abnormalities). Collectively, these result in an insufficient

exchange of oxygen and nutrients [72] in an otherwise normal embryo.

During the development of the placenta, trophoblast cells invade into the uterine tissues to gain

access to the maternal blood supply [73]. This process requires a balanced interplay between the

factors that promote and restrain trophoblast invasion. Consistent with this notion, early work has

suggested that the uterine microenvironment controls invasion of trophoblast, supporting the role

of the maternal decidual tissue as a restrain to trophoblast invasion [57, 74]. These studies also

imply that factors that promote trophoblast invasion are produced and secreted by the trophoblast

cells. Supportive of this model, it has been shown that the embryo secrete insulin-like growth

factor (IGF) 2, which drives trophoblast migration [1, 5, 9, 10, 29, 32, 75-77], as well as matrix

metalloproteinases (MMPs), which degrade the decidual extracellular matrix (ECM) [5, 78, 79].

To counteract these factors, the maternal decidua secretes tissue inhibitors of MMPs (TIMPs) [80,

81], as well as several IGF-binding proteins (IGFBP), including IGFBP1 [82-85] and IGFBP4 [65,

86-92]; molecules that are known to inhibit trophoblast invasion. Similarly, an interplay between

IGF and IGFBP exists, where IGF2 inhibits maternal decidual IGFBP1 and TIMP-3 expression,

supporting paracrine actions of trophoblast-derived IGF2 on decidual regulators of invasion [93].

Indeed, mounting evidence suggests that IGF and IGFBPs play critical roles during early

pregnancy, and it has been suggested that alterations of the IGF/IGFBP system can have serious

Page 24: © 2013 Juanmahel Davila - IDEALS

9

complications leading to IUGR [84, 85, 94]. In concordance with the importance of the IGF/IGFBP

system, overexpression of IGF1 [95] or IGFBP1 [94] in animal models leads to fetal loss or altered

placental development, respectively. In the clinics, women that are destined to deliver a growth-

restricted infant, display elevated levels of serum IGFBP4 compared to women with normal

pregnancies [65]. It is clear that the IGF family of proteins play an important role during pregnancy

overlapping the window of implantation and placentation [82, 85, 86, 90, 93, 96, 97]. Although

there are species differences, studies on transgenic mice have provided insights into the

IGF/IGFBP system during pregnancy [94, 95, 98-101].

2.1.3 Assisted reproductive technologies

The use of assisted reproductive technologies (ART) has increased dramatically in the last decade

[37, 39]. Many couples that have problems conceiving turn to ART to aide in this process. To

increase the likelihood of successful pregnancy, pre-implantation genetic screening of biopsied

blastomeres during in vitro fertilization has been recommended in order to prevent the transfer of

karyotypically abnormal embryos in couples with normal karyotypes. However, this practice may

not be effective in couples with recurrent idiopathic miscarriage due to limitations of genetic

testing [102]. Nonetheless, there is a need for research to investigate potential factors that could

predispose an individual to recurrent pregnancy loss.

2.2 Uterine receptivity, the window of implantation, and early pregnancy in the mouse

Steroid hormones, 17β-estradiol (E2) and progesterone (P4), regulate cell proliferation,

differentiation, and vascular remodeling of the endometrium by binding to their respective nuclear

receptors estrogen, ESR1/ERα (estrogen receptor α) and PGR (progesterone receptor). In each

reproductive cycle, under the influence of these hormones, the endometrium becomes competent

to receive an embryo during a short duration of time known as “window of receptivity” [11, 103-

106] (Figure 2.1). Intimate interactions between the uterus and fetus occur during this period that

promotes implantation leading to establishment of pregnancy [11, 107, 108]. About one third of

human pregnancies end in spontaneous abortion. Indeed, implantation failure still remains an

Page 25: © 2013 Juanmahel Davila - IDEALS

10

unsolved problem in reproductive medicine. It is a major cause of infertility [6, 37, 39, 45] and

represents a major challenge in ART.

2.2.1 Critical events during pregnancy

The concomitant actions of steroid hormones prepare the endometrium for the arrival of an embryo

and allow it to invade so that pregnancy is established. Whereas E2 initially regulates uterine

growth by stimulating epithelial cell proliferation and edema, P4 is mostly required for the

differentiation of stromal cells during pregnancy. Indeed genomic profiling of mouse and human

endometrium has uncovered a complex, yet highly conserved network of steroid-regulated genes

that supports stromal cell differentiation. In order to advance our understanding of the mechanisms

regulating critical aspects of pregnancy and better address the clinical challenges of infertility, it

is important to integrate the information gained from the human and mouse models [2, 109, 110].

Undeniably functional studies using transgenic mouse models over the past several years have

yielded valuable information regarding the molecular basis of pregnancy complications in humans

[25, 111]. Below we summarize some of the critical events required for the successful

establishment of pregnancy.

2.2.1.1 Implantation

Embryo implantation and placentation are critical events that support embryonic growth and

ultimately delivery of a live offspring in the mammalian system. [1, 25]. Failure of the

endometrium to provide a permissive growth environment can severely compromise the outcome

of pregnancy, leading to an impaired embryonic development or death. Despite some species

differences, in general, the principles of implantation and placentation are well conserved among

mammalian species [1, 5, 10, 14, 27, 28, 112, 113]. In species with invasive placentation,

implantation into the uterine endometrium is an absolute requirement and involves a series of

complex interactions between the implanting embryo and the endometrium [5, 41, 114-116].

In mice following fertilization, which typically occurs on the day the copulatory plug is found, is

termed as day 1 of pregnancy (Figure 2.2). The newly formed embryo then travels through the

Page 26: © 2013 Juanmahel Davila - IDEALS

11

oviduct and enters the uterine cavity on day 4 of pregnancy [41, 107, 117]. The embryo will then

begin an intimate crosstalk with the endometrium to initiate the implantation process (Figures 1.2

and 1.3) [1, 5, 116]. A transitory rise in E2, which in mice occurs on noon of day 4 pregnancy,

known as “nidatory estrogen”, is responsible for the initiation of implantation [1, 5, 8, 14, 118].

The necessity of the pre-implantation estrogen was established in the delayed implantation mouse

model, where ovariectomy before the nidatory estrogen surge prevents implantation. Replacement

of estrogen in these ovariectomized mice rescues implantation [103, 106]. Although is clear that

steroid hormones regulate embryo implantation in humans, it is still debatable whether a similar

E2 surge initiates embryo implantation in these species [14].

In a series of coordinated steps, i.e. apposition, adhesion, and invasion, the cells of the

trophectoderm penetrate into the stromal compartment of the endometrium and invasion into the

antimesometrial side of the uterus, i.e. the uterine side opposite to the mesometrial ligament starts

pregnancy [41, 117]. During this period, the uterus under the influence of P4 and the E2 becomes

receptive, a critical step for the initiation of this process (Figures 1.1 and 1.2). However, the uterus

will not remain permanently in a receptive phase and between 24 to 36 hours after the nidatory E2,

the uterus becomes refractory to implantation [11, 104, 105].

2.2.1.2 Decidualization: A key event for successful establishment of pregnancy

Maternal-fetal dialogue involving an intimate interaction between the specialized trophectodermal

cells of the embryo and the receptive uterine lining of the mother starts pregnancy [5, 10, 25]. The

attachment and breaching of the implanting embryo through epithelial barrier causes the

underlying fibroblastic stromal cells to undergo further proliferation and subsequent differentiation

into polyploid secretory decidual cells (Figure 2.3), in a process termed as decidualization [10,

108, 116, 119]. During decidualization, stromal cells transform into epitheloid cells that have

unique biosynthetic and secretory properties [23-25, 29, 33, 80, 84, 90, 91, 120-123]. These cells

are responsible for producing and secreting hormones, growth factors, and cytokines, which are

essential for tissue and vascular remodeling to support the growth and development of the embryo

until the establishment of the chorioallantoic (definitive) placenta. This typically happens on day

9 of pregnancy in rodents [1, 5, 9, 10, 23, 28, 29, 31, 78, 112, 124-133]. Another important feature

Page 27: © 2013 Juanmahel Davila - IDEALS

12

of the decidua is to resist the invasive capacity of trophoblasts by secreting products (such as

IGFBPs, TIMPs) that bind to trophoblast and modulate their migration and invasion [5, 80, 81, 83,

84, 86, 87, 122, 123, 134]. The invading trophoblasts secrete MMPs that degrade the decidual

ECM [5, 23, 78] and the decidual cells express high levels of TIMPs [135, 136], which control

trophoblast invasion. The maternal decidua allows trophoblast invasion far enough to access the

maternal blood supply, but not too far that the mother becomes endangered [23, 25, 29, 94, 130,

137, 138]. As embryo invades, it initiates a connection with the maternal vasculature and

ultimately, placentation ensues. Clearly, the formation of a functional placenta requires extensive,

but tightly regulated, invasion of the trophoblast through the endometrium [23, 25, 29, 94, 137,

138]. Insufficient trophoblast invasion can lead to number of placental complications such as

preeclampsia [139-143], whereas excessive invasion is associated with disorders such as placenta

accreta [125, 144-148].

The decidual cells are also thought to provide an immune barrier to the mother from the allogenic

embryo [23, 29, 149-151]. Mature decidual cells produce and secrete cytokines and chemokines

that maintain the trophoblast in its allogenic phenotype to avoid being targeted by the maternal

immune system. To this end, it has been thought that the modulation of the maternal immune

system is mediated by the recruitment of peripheral and resident leukocytes in the maternal

decidua, which provide local immunomodulation and support placental development [131, 143,

152, 153]. Indeed an immune environment is established in the endometrium, which is permissive

to the developing embryo [5, 130, 143, 152, 153].

The initiation of the stromal differentiation process can be monitored by the gene expression or

enzymatic activity of liver/kidney/bone alkaline phosphatase (ALPL, Figure 2.4) [154]. Around

day 4 midnight of pregnancy (or early day 5 morning), after the embryo has embedded itself into

endometrium, it is surrounded by the newly formed decidual cells of the primary decidual zone

[29, 155]. By day 6 and 7 of pregnancy, the stromal cells next to the primary decidual zone begin

to proliferate and then differentiate to form the secondary decidual zone (Figure 2.3B), forming

the decidua capsularis [1, 155]. As these cells differentiate, the cells of the antimesometrial

decidua, which differentiate first, will exit the cell cycle and begin expressing decidual prolactin-

related protein (PRL8A2/dPRP), which is typically used to assess the differentiation potential of

Page 28: © 2013 Juanmahel Davila - IDEALS

13

these cells [22, 156-158]. By day 8 of pregnancy, the differentiation program continues in the

mesometrial side of the uterus, i.e. the side that is closest to the principal maternal blood vessels,

and later forms the decidual basalis. These two decidual cell populations differ not only in their

morphology, but also by the genes they express and the putative roles they play in pregnancy [150].

The antimesometrial decidua is formed primarily by giant, polyploid, and closely packed decidual

cells, whereas the mesometrial decidua is formed by much smaller, loosely packed decidual cells.

It is relatively easy to separate these two sub-populations by the differences in their size and density

[150, 155]. Also, it is assumed that the mesometrial decidual cells regulate many important aspects

of vascular remodeling (Figure 2.3) [23, 29-32], energy storage and metabolism [159-161], as well

as placentation (Figure 2.5) [28, 31, 112]. By day 10 of pregnancy, however, the terminally

differentiated stromal cells undergo apoptosis to accommodate the growing embryo and

placentation ensues to maintain the conceptus until term [23, 150, 155]. Therefore, perturbations

in the decidual program will lead to pregnancy complications that may manifest in early pregnancy

loss. In humans, however, it should be noted that the stromal cell differentiation program is

initiated in the absence of embryo, but the extent of decidualization as well as vascular remodeling

increases significantly with the onset of implantation and establishment of pregnancy [33].

Experimentally, stromal cell differentiation into decidual cells can be studied independently of the

implanting embryo in an in vivo [154, 162, 163] as well as an in vitro model systems [164, 165].

In vivo, the experimentally-induced stromal differentiation is performed by ovariectomizing non-

pregnant mice, then replacing estrogen and progesterone, in a timely manner. Once the uterus is

primed, an external stimulus (most often oil injection) is given to one uterine horn, whereas the

other horn remains unstimulated, and serves as an internal control [163]. The in vivo

experimentally-induced differentiation model has been used extensively, by our laboratory, as well

as others, to establish the importance of steroid hormones in the regulation of stromal

differentiation during decidualization.

2.2.1.3 Angiogenesis and vascular remodeling are essential for maintenance of pregnancy

The establishment of an extensive angiogenic network during decidual transformation [23, 31, 57,

116, 166-168] is a key physiological event that supports early pregnancy and embryo development

Page 29: © 2013 Juanmahel Davila - IDEALS

14

(Figure 2.3). In mice, the initiation of vascular remodeling occurs soon after implantation and

manifests as an increase in microvascular permeability; only seen at the sites of embryo apposition

[116, 169-171]. Chicago blue dye injected via tail vein can be used to determine the sites of embryo

attachment on day 5 and 6 of pregnancy [169, 171]. This increase in vascular permeability is

thought to be a pre-requisite for embryo implantation [169, 172]. Under the influence of estrogen,

vascular-endothelial growth factor (VEGF) is produced and secreted as early as day 5 of pregnancy

[173-175]. This potent mitogen stimulates endothelial cells to proliferate and is thought to play a

major role in uterine angiogenesis during the decidualization process [176-178]. Because Vegf is

induced and maintained during the stromal cell differentiation process, it is considered as a marker

of decidualization [178, 179]. VEGF is a potent driver of angiogenesis, however, other cytokines

and growth factors are also recognized as angiogenic promoters. Therefore, one or more factor(s)

may be acting in concert with VEGF to mediate endometrial angiogenesis.

Vascular endothelial growth factor is a key regulator of angiogenesis [180, 181], and targeted

deletion of gene encoding Vegf results in embryonic death due to abnormal blood vessel formation

[181, 182]. VEGF has been shown to be one of the earliest genes activated during pre-implantation

embryo development [183]. Extensive analyses on the expression of VEGF, its steroid hormone

regulation, and involvement of specific receptors during pregnancy have suggested a critical role

for the VEGF system in pregnant uterus [175, 183]. VEGF is a heparin binding homodimeric

glycoprotein that mediates its action through tyrosine kinase receptors, namely VEGFR1/FLT1,

VEGFR2/FLK1, as well as the glycoprotein co-receptor neuropilin 1 (NRP1) [168, 178, 184, 185].

VEGFR2/FLK1 is the predominant VEGF receptor in the uterus [185], and binding of VEGF to

this receptor activates the VEGF signaling cascade. VEGFR1/FLT1 is also expressed in the uterus,

but is thought to have minimal to no effect during uterine angiogenesis [185]. Previous studies

have also shown that the human blastocysts are able to stimulate angiogenesis via the production

of significant amounts of VEGF [186]. Several studies have reported the expression of VEGF and

its receptors, in first-trimester human decidua basalis and parietalis, as well as in several cell types,

including endothelial cells, epithelial cells, macrophages and trophoblasts [187, 188]. VEGFA

(also indicated as VEGF), the predominant vascular endothelial growth factor that drives blood

vascular angiogenesis, has also been shown to stimulate trophoblast proliferation and invasion

Page 30: © 2013 Juanmahel Davila - IDEALS

15

[189]. Therefore, VEGFA may play an active role in trophoblast invasion and angiogenesis [187,

188].

Although it is clear that estrogen and progesterone play a major role during endometrial

angiogenesis [175, 178, 190], their specific roles in this process remain unsolved and controversial.

Whereas vascular permeability is under the regulation of estrogen [191], endothelial cells seem to

proliferate in response to progesterone [192, 193]. The importance of estrogen during angiogenesis

was recently studied in our laboratory [32, 194]. Laws et al. [32] clearly showed that ablation of

connexin 43 (Gjp1/Cx43), an estrogen-regulated gene from the decidual cells, caused a defect in

stromal cell differentiation and impaired vascular remodeling, primarily due to a marked reduction

in hypoxia inducible factors (Hif1α and Hif2α), Vegfa, angiopoietins (Angpt2 and Angpt4).

Similarly, Das et al. [194] reported that inhibition of local uterine estrogen can effectively impair

the production of pro-angiogenic factors, such as angiopoietins, which are well-known regulators

of angiogenesis [113].

During pregnancy, members of the angiopoietin family including Angpt1 and Angpt2, along with

Tek/Tie2, are expressed in the endometrium during stromal cell differentiation and modulate

angiogenesis [195, 196]. Angiopoietins are a class of angiogenic regulators that complement

VEGF signaling [180, 184, 197]. Angiopoietins mediate their actions by binding to the tyrosine

kinase receptor TEK/TIE2, and in the presence of VEGF, angiopoietins, especially ANGPT2,

promotes vascular remodeling in front of the invading vascular sprouts [184, 197, 198]. Although

it has been reported that steroids regulate the expression of these genes in the endometrium [192,

193], the exact mechanism and signaling pathways by which these hormones regulate angiogenesis

remain unclear.

Hypoxia is a detectable feature in the uterine environment during embryonic development [199-

201]. HIFs are major transcriptional regulators of Vegf. They play a critical role in maintaining the

physiological balance of oxygen tension and are closely associated with angiogenesis [1, 199, 202-

205]. Oxygen homeostasis is critical for cell survival and in response to hypoxic conditions, cells

activate the transcription of Hif1α to restore homeostasis [1, 206, 207]. Activation of HIF

stimulates Vegf expression, which promotes angiogenesis, thereby restoring oxygen homeostasis.

Page 31: © 2013 Juanmahel Davila - IDEALS

16

Both Hif1α and Hif2α transcripts are upregulated during the early stages of implantation and their

expressions are maintained during decidualization [1, 199, 202-204]. It has been reported that

EPAS1/HIF2α complements the action of HIF1α to regulate VEGF expression [205, 208].

Previous studies from our laboratory have demonstrated that both Angpts and Hif2α are regulated

by local endometrial E2 production during decidualization, thus contributing to VEGF regulation

and angiogenesis [194].

Whereas many of the early events leading to angiogenesis and vascular remodeling are regulated

by estrogen during pregnancy, progesterone also plays a major role in these events. Several in vivo

studies have suggested a pro-angiogenic role for progesterone [192, 193]. Human endometrial

endothelial cell cultures show an enhanced capacity to form tube-like structures and sprouts when

progesterone is added to the culture media [209]. It has been suggested that P4-induced

proliferation is most likely caused by a direct, but non-genomic effect of progesterone. Consistent

with this notion, Ma et al. [192] reported progesterone-mediated induction of endothelial cell

proliferation in ovariectomized mice and suggested the involvement of VEGF and its receptor

VEGFR2/FLK1 in conjunction with NRP1 in this process. Furthermore, progesterone-induced

endometrial endothelial cell proliferation is thought to be mediated by multiple angiogenic

mechanisms [32, 194, 210-212] and regulatory factors, acting in a paracrine manner between cells

within the endometrium [210-212]. These paracrine effects are absent in vitro, which could explain

the disparity between some in vitro and in vivo observations.

With the progression of decidualization and pregnancy, new blood vessels begin to form a vascular

plexus in the mesometrial side of the uterus (Figure 2.3) [23, 31, 57, 166, 167]. Moreover, the

specific pattern this plexus acquires is thought to be influenced by signals that originate from the

embryo [30, 130, 198]. In the experimentally-induced differentiation model the specific pattern of

vascular remodeling that is typically observed in pregnancy, is not acquired [30, 198]. It is believed

that the concerted actions of steroids, in addition to the signals originating from the embryo, are

thought to regulate the final outcome of uterine angiogenesis during pregnancy [23, 29, 30, 32, 57,

112, 130, 176, 194, 213].

Page 32: © 2013 Juanmahel Davila - IDEALS

17

2.2.1.4 Placentation

Whereas placentation is highly variable and species-dependent, the primary and major goals of the

placenta are to 1) anchor the conceptus in the maternal uterine wall, 2) establish a vascular supply

that enable optimal growth and development of the conceptus, 3) and protect the fetal allograft

from rejection [5, 9, 23, 28, 29, 31, 112, 124-128, 149-151, 214]. To achieve these goals, there are

complex molecular dialogues that take place between the maternal decidua and the developing

conceptus, as well as the placental structures [1, 5, 10, 23, 29, 78, 112, 127-133, 215].

During the early stages of gestation, it is thought that the developing embryo is supported by the

secretions of the luminal epithelium of the oviductal cells and those from the uterus [116, 216].

However, as the embryo grows in size and the trophectoderm begins to differentiate, the nutritional

requirements of the embryo change. Initially, the primary sources of energy for the early embryo

are pyruvate, lactate, and amino acids. But after compaction of the morula, glucose becomes the

primary source of energy for the embryo [28, 116, 217, 218]. At this point, the developing embryo

involving a dynamic crosstalk between trophoblasts and the maternal decidua begins attachment

and invasion through the luminal epithelium into the maternal endometrium [1, 5, 10, 29, 171,

219].

2.2.1.4.1 Placentation in humans and mice

Mice and humans exhibit hemochorial-type of placenta, and although there are some differences

in the architectural details of these placentas, their overall structures and the molecular

mechanisms underlying placental development are thought to be quite similar [28, 126-128, 132,

220]. As a consequence, the rodent system, especially the mouse, has been extensively used as a

model for studying the fundamentals of placental development. In mice, placental development

begins on day 4 of pregnancy when the blastocyst is formed (Figure 2.5) [5, 28, 126, 128]. Around

the time of invasion into the endometrium on day 5 of pregnancy, the mural trophectoderm cells,

i.e. those not in contact with the inner cell mass, proliferate and begin to give rise to two diploid

cell types: the extraembryonic ectoderm – presumably those which give rise to the chorionic

trophoblast forming the placental labyrinth and are analogous to cytotrophoblast of the human

Page 33: © 2013 Juanmahel Davila - IDEALS

18

placenta – and ectoplacental cone – presumably those which give rise to the spongiotrophoblast,

and are thought to support the developing labyrinth [28, 126, 128, 132]. The trophoblast cells that

are farthest from the inner cell mass stop dividing, but continue endoreduplication of the genome

without mitosis, and become the trophoblast giant cells (analogous to human extravillous

cytotrophoblast cells). The trophoblast giant cells form the interface with maternal decidua and are

capable of invasion into the endometrium [28, 31, 124, 132]. The cells from the ectoplacental cone

and extraembryonic ectoderm lineages thus establish the three layers of murine placenta: 1) The

labyrinth zone, which is the zone closest to the chorionic plate; 2) the juctional zone, which

supports and surrounds the labyrinth and is composed spongiotrophoblasts and trophoblast

glycogen cells; and the final layer 3) the trophoblast giant cells, which are at the maternal-fetal

interphase and a subgroup of these cells invade the maternal blood vessels [5, 10, 28, 31, 126, 127,

132, 214]. Although it is clear that there are interactions and crosstalk between the maternal and

embryonic cells, however, due to the complex nature of these interactions, the mechanism

underlying the placental formation is not clear.

2.2.1.4.2 Regulation of trophoblast invasion and placental pathologies

An important function of the decidua is to resist the invasive capacity of trophoblasts by secreting

factors that bind to trophoblasts and modulate their migration and invasion [23-25, 29, 33, 80, 84,

90, 91, 120-123, 127, 221]. Beyond the in vitro data, which strongly support the concept of a

“decidual barrier”, the results from classical embryo transfer experiments support the role of the

decidua in regulating trophoblast invasion. When mouse blastocysts are transferred to subepithelial

ectopic sites (such as kidney, testis, or anterior chamber of the eye), the embryo is able to penetrate

into the non-decidualized connective tissue much more deeply than into uterine decidua [222-225].

Similar examples of this phenomenon have been reported in human pregnancies established at

ectopic pregnancy sites. In oviductal pregnancies, for example, the trophoblast cells tend to be

more invasive [226]. It has been speculated that a potential reason for this excessive invasion is

because the pseudo-decidualized oviductal cells [129, 227] do not produce IGFBP1 [129].

Furthermore, it has been reported that excessive trophoblast invasion or ectopic pregnancy tends

to occur in tissues that exhibit impaired decidualization [129, 227]. Thus the unique feature of the

decidua to suppress uncontrolled trophoblast invasion, provides a possible explanation for why, in

Page 34: © 2013 Juanmahel Davila - IDEALS

19

pathological conditions where the embryo invades ectopic sites, trophoblast invasion tends to be

more invasive [127, 129, 226]. In more severe cases, where the decidual barrier is defective, little

or too much invasion can result in further complication to the mother. [39, 53, 54, 58-60]. One of

the hallmarks of preeclampsia is shallow trophoblast invasion, thought to cause an inadequate

spiral artery remodeling, leading to placental hypoxia. It is widely accepted that placental hypoxia

initiates the cascade of events that ultimately results in the maternal manifestations of high blood

pressure and proteinuria [61-64]. Similarly, in placenta accreta it has been proposed that either

primary deficiency of decidua or abnormal maternal vascular remodeling leads to excessive

trophoblastic invasion, resulting in massive postpartum hemorrhage and commonly leads to

emergency hysterectomy [65-70]. Nonetheless, the formation of the placenta is a critical process

during embryogenesis, and without a healthy placenta, the embryo will not survive and this

malformation can trigger a spontaneous abortion.

2.2.1.4.3 The mid-stages of placentation in the mouse

Day 10 of pregnancy is a critical stage in mouse placenta. At this point, the chorioallantoic

circulation becomes the primary support for the growing embryo. The placenta forms an essential

interface between the maternal and fetal layers that allow the exchange of oxygen, nutrients, and

waste products to provide a nourishing environment to the growing fetus [5, 9, 28, 31, 112, 176].

By day 12 of pregnancy, the fetal trophoblast cells start to invade into the maternal decidua to

reach spiral arteries and induce modifications with make high capacitance, low resistant blood

vessels [167, 228-232]. As a consequence of trophoblast invasion, the endometrial spiral arteries

undergo dilation, which results from the loss of smooth muscle cells surrounding the arterioles

[233]. Therefore, defective or improper invasion of fetal trophoblast cells into maternal decidua

can result in various clinical problems such as, spontaneous abortion, preeclampsia, and/or

intrauterine growth restriction [173, 174, 234-237].

2.3 Rho GTPases

The Ras Homology (Rho) family of guanosine triphosphatases (GTPases) are small signaling G

proteins (~21 kDa), and belong to the subfamily of the Ras superfamily of proteins (Figure 2.6).

Page 35: © 2013 Juanmahel Davila - IDEALS

20

Members of the Rho GTPase family regulate many aspects of actin dynamics and are found in

organisms raging from plants and Caenorhabditis elegans, to mice and humans. While their first

discovered role was regulation of actin dynamics in fibroblast cells [238], recent evidence has

suggested that the members of this family play important roles in signal transduction, cell

proliferation and differentiation, vesicle transport, cancer progression and metastasis, angiogenesis

and gene expression [238-248]. Clearly, these proteins play critical roles in many cellular

processes.

2.3.1 The Rho GTPases

Since the discovery of the first Rho member Rhoa in 1985, from a low stringency cDNA screening

[249], other members including Ras-related C3 botulinum toxin substrate 1 (Rac1) and Rac2 in

1989 [250, 251], and cell division control protein 42 homolog (Cdc42) in 1990 were discovered

[252]. To date, more than 18 members in eight distinct subfamilies have been reported [244], and

of these three members of the family, RHOA, RAC1, and CDC42 [238, 249, 250, 253], have been

extensively studied. The Rho proteins are virtually found in all eukaryotic species examined so

far, and are highly conserved [241, 244, 247, 254-259]. In addition, given their close homology to

Ras GTPases, the Rho GTPases act as molecular switches to regulate signal transduction pathways

by alternating between an inactive GDP-bound state and an active GTP-bound conformational

states. The activities of these proteins are primarily controlled by growth factors or integrins, and

seven-transmembrane receptors [238, 239, 243, 244, 246, 254, 255, 258, 260-265].

2.3.2 Role of Rho GTPases in actin dynamics

It is clear that the actin cytoskeleton drives many dynamic processes in cells, including migration,

phagocytosis, endocytosis, vesicle trafficking to the plasma membrane, morphogenesis, and

cytokinesis [238, 241-244, 247, 248, 250, 260, 266-270]. Further, depending on their biological

function and location within the body, cells can acquire many shapes and forms. However, before

a cell can undergo cellular processes like mitosis or locomotion, a degree of polarity is required

[238, 244], which requires the cell to adopt a specific shape or direction rather than existing as an

amorphous cell. Each member of the Rho family, i.e. RHO, RAC, and CDC42, is responsible for

Page 36: © 2013 Juanmahel Davila - IDEALS

21

specific regulation of actin dynamics [238, 244]. One of the most well-known morphological

structure that is controlled by Rho proteins is the formation of lamellipodia and filopodia, which

look like “sheets” or “fingers”, respectively, and aid in the propulsion and sensing across surfaces

[238]. Rho GTPases are also responsible for the formation of stress fibers (originally thought to

arise from the effects of tension) [241, 270], important in cell motility, contractility, and adhesion

[243].

2.3.2.1 Functions beyond actin dynamics

Researchers working with Rho GTPases in the mid-1980s quickly realized their importance in

actin dynamics. However, more recent work has demonstrated that Rho GTPases do much more

than regulation of actin assembly, cell shape and migration. Over the last 20 years using a

combination of techniques, such as: 1) use of dominant-negative mutants of Rho, 2) use of RAC

and CDC42 toxins such as C3 (Rho inhibitor) or toxin B (RHO, RAC and CDC42 inhibitor), 3)

analysis of transgenic model organisms, and 4) use of siRNA have demonstrated how critical each

of these molecules are for the proper functioning of many cellular processes within a cell. Several

lines of evidence seem to suggest that members of the Rho family can regulate a wide range of

cellular functions including, cell-cell communication, gene expression, activation of the NADPH

oxidase complex, maintenance of cell polarity, cell proliferation/differentiation, angiogenesis, and

cancer progression and metastasis [113, 242, 246-248, 250, 254, 261, 262, 264, 269-282]. Signal

transduction pathways from the Rho proteins have been also implicated in the regulation of gene

transcription. For example, RHO is known to activate serum-response factor, whereas RAC1 and

CDC42 activate the c-Jun N-terminal kinase and p38 mitogen-activated protein kinase pathways

[244].

2.3.3 Regulators of the Rho GTPases

Under normal physiologic conditions, GTPases, including Ras and Ras-like proteins, are

predominantly regulated at the post-translational level. Promoter analysis of the human Ras

oncogene and Rac1 has revealed that the gene architecture resembles that of a housekeeping gene.

The promoters lack regulatory elements, like the TATA box and CAAT box, have multiple

Page 37: © 2013 Juanmahel Davila - IDEALS

22

initiation start sites, and a rich CG content, with CpG islands surrounding the transcription

initiation sites [283, 284]. However, use of in silico assays to study the promoter of the human

Rac1 gene indicated consensus binding sites for several transcription factors, including E2F-4, as

well as several zinc finger transcription factors, such as SP1, KLF15, MAZ, MZF1, PLAG1, and

WT1 [285]. The discrepancies in these results could be due to the use of different human Rac1

gene sequences, but clearly, further studies are needed.

The post-transitional modifications that control intracellular signaling pathways regulated by Rho

GTPases, act by affecting the state of the molecular switch. Alternating between GDP-bound and

GTP-bound forms, affect the conformation of the GTP-binding switch 1 and switch 2 regions

(Figure 2.7) [250, 254, 261, 264, 273, 274, 280, 286, 287]. The GDP-bound form is considered as

the inactive state, whereas the GTP-bound state activates downstream pathways by binding to

effectors. The cycling of Rho GTPases between the active/inactive states is regulated by three

families of proteins, guanine nucleotide-exchange factors (GEFs), GTPase-activating proteins

(GAPs) and guanine nucleotide-dissociation inhibitors (GDIs) [250, 254, 255, 261, 264, 273, 274,

286]. GEFs, however, activate Rho GTPases, GAPs and GDIs act as inhibitors of the protein

activity [244, 248, 255, 258, 260, 264, 287].

Because GDP binds tightly and the GTPase capacity of the molecule is slow, small GTPases

require GEFs to facilitate dissociation of GDP in exchange for a GTP molecule. Activated Rho

GTPases are known to interact with close to 80 effector molecules (i.e. kinase, phospholipases,

and scaffold proteins), which regulate downstream signaling events, including cell proliferation

(by contributing to spindle orientation), differentiation (by regulating gene expression, and perhaps

cell geometry and shape [288, 289]), cell migration (by affecting actin dynamics), and gene

expression (via downstream protein-protein activation). Soon after the GTPase becomes active,

GAPs associate with the protein and promote the GTPase activity leading to the hydrolysis of GTP

to GDP [287]. Hence, the transition (or the molecular switch) between one state and the other is

carefully balanced by the activation or suppression of GEFs and GAPs. Finally, the activity of Rho

proteins can be further attenuated by their interaction with GDIs, which shields the hydrophobic

tail that is incorporated in the C-terminus, and prevents the interaction with downstream effectors

and confines the GTPase to the cytosol (Figure 2.7) [287]. The hydrophobic tail allows the

Page 38: © 2013 Juanmahel Davila - IDEALS

23

anchoring and incorporation of the GTPase into the cell membrane when active (Figure 2.7) and

permits the interactions with effector proteins. Interestingly, not all known GTPases have the

capacity to hydrolyze GTP [255, 260]. Other mechanisms regulating Rho GTPases also exist, i.e.

phosphorylation, ubiquitination, and degradation, but these are minor, compared to those

mentioned above.

2.3.4 Rho GTPases and disease

The founding member of the Ras superfamily was intensely studied during the 1980s, since it was

found to be mutated in 30% of all human cancers. Further studies showed that a single amino acid

substitution was responsible for the protein’s insensitivity to GAPs and therefore remained

constitutively active [244]. Sequencing information derived from human cancers has uncovered

no similar mutations in Rho GTPases. However, genetic alterations in the Rho GTPase signaling

pathways have been reported in several human diseases. For instance, alterations in Rac2 gene

sequence have been associated with immunological disorders such as leukocyte adhesion

deficiency [245, 267, 290]. Similarly, other mutations in GAPs and GEFs have been reported [240,

244, 291]. To date, Rac1 has been shown to be abnormally expressed in several tumor samples

[203, 240, 265, 291]. Although there is evidence for polymorphism in the Rac1 gene sequence,

most are intronic, and thus may not contribute to any genetic dysregulation [285]. However, a

recent report has shown that a possible functional mutation due to a polymorphism in the Rac1

gene could result in drug resistance [285]. In addition, an alternatively spliced variant of Rac1,

Rac1b, containing an additional exon (exon 3b), and encoding a protein with 19 additional amino

acid residues, has been described. Rac1b gene, primarily found in cancers [292-296], produces a

functional GTPase that displays an increased intrinsic guanine nucleotide exchange rate [294] and

a decreased intrinsic GTPase activity [294, 297] compared with those of RAC1.

2.3.5 Ras-related C3 botulinum toxin substrate 1 (RAC1)

Among the Rho GTPases, the subfamily of proteins named Rac (Ras-related C3 botulinum toxin

substrate) has been most studied. There are four known Rac proteins (RAC1, 2, 3, and 1b), which

display ~ 90% identity in their coding sequences and overall structure [250, 261, 268, 280], but

Page 39: © 2013 Juanmahel Davila - IDEALS

24

have different expression patterns. Whereas Rac1 is widely distributed in many cells [250, 251],

Rac2 expression is mostly restricted to cells of the hematopoietic lineage [250, 290, 298], and

Rac3 expression is in the brain, liver, and lung [268, 299]. Rac1b, the splice variant of exon 3 of

Rac1, is detected primarily in cancerous cells [292-296]. Available protein sequences indicate that

RAC1 is highly conserved among eukaryotes [241, 259], with high homology between mouse and

human (Figure 2.8).

RAC1 is known to play a fundamental role in a wide variety of cellular processes, which include

actin cytoskeletal reorganization, cell transformation, angiogenesis, axonal guidance, and cell

migration. More recently, researchers have begun to uncover new roles for RAC1 in a variety of

physiological settings [7, 26, 255, 261, 266, 300-302]. Some of the physiologic functions that have

been attributed to RAC1, include regulation of cell growth, vesicle trafficking, and generation of

reactive oxygen species.

Like other members of the Rho family of GTPases, RAC1 transduces extracellular signals from

seven-transmembrane G-coupled protein receptors, integrins, and growth factor receptors to

effector molecules that modulate multiple signaling pathways [260]. RAC1 can promote the

activation of MAP kinase pathways and gene activation, however, its most important functions

include the regulation of cytoskeletal structure and shape, resulting in important cell functions

related to cell attachment, movement, and vesicle trafficking [248, 257, 260, 303-308]. Indeed,

one of the primary functions of RAC1 is to promote the formation of the lamellopodium, which

are sheet-like projections on the leading edge of a motile cell that propel the cell across a substrate

[247, 248]. Furthermore, the action of RAC1 on actin fibers plays critical roles in vesicle transport.

In dendritic cells, RAC1 induces actin-rich membrane protrusions during pinocytosis [309], which

allows for antigen presentation, nutrient uptake, and sampling of the environment. RAC1 is also

involved in phagocytosis by mediating localized polymerization of actin at the membrane to

promote the internalization of attached particles and microorganisms [260]. In other cell types,

RAC1 has been shown to regulate endocytic and exocytic trafficking pathways [247, 308]. Unlike

CDC42, RAC1 is not involved in the early secretory pathway or membrane trafficking from the

rER to the Golgi [308, 310]. However it does play a role in the later stages of secretion. RAC1

regulates clathrin-dependent endocytosis [311] by modulating phosphatidylinositol-3 kinase

Page 40: © 2013 Juanmahel Davila - IDEALS

25

(PI3K) and synaptojanin 2, a novel RAC1 effector [312]. Phosphatidylinositol 4,5-bisphosphate is

phosphorylated by PI3K to generate inositol triphosphate, which induces the activation of RAC1-

GEFs to regulate actin rearrangement and vesicular trafficking in various cell types [313].

Recently, PREX1, a PI3K-dependent Rac exchange factor, has been identified as a novel regulator

of the glucose transporter 4 (SLC2A4/GLUT4) trafficking in adipocytes. PREX1 can activate

RAC1, and this in turn stimulates actin polymerization and facilitates SLC2A4/GLUT4 transport

to the plasma membrane [314]. Moreover, RAC1 can modulate clathrin-independent endocytosis

by interacting with phosphatidyl-dependentinositol-4-phosphate5-kinase (PIP5K), via the

polybasic region (PBR binding domain of RAC1). Calmodulin can also interact with RAC1 via

the PBR and disrupt the interaction between RAC1 and PIP5K through steric hindrance [315].

RAC1 has also been reported to regulate calcium-dependent exocytosis in some cells. In neurons,

a late step of Ca2+-dependent neurotransmitter release is regulated by RAC1 [316]. Activation of

the RAC1 pathway in bovine chromaffin cells, transfected with human chorionic gonadotropin,

led to an increase in secretion [317]. Similarly, in acinar cells of the pancreas, RAC1 can increase

the secretion of amylase in response to cholecystokinin [318]. However, the precise mechanisms

by which RAC1 pathways regulates exocytosis remain unknown.

Another function of RAC1 that has received attention is its involvement in angiogenesis [203, 275,

277, 319-326]. Hypoxia occurs in many cellular environments that are undergoing growth or

differentiation. Oxygen homeostasis is critical for cell survival and in response to hypoxic

conditions, cells activate the transcription of Hif1α to restore homeostasis [327-334]. Activation

of HIF proteins stimulates the expression and production of Vegf, which drives angiogenesis and

restores oxygen homeostasis [199, 205, 327, 329, 332, 333, 335]. Several lines of evidence have

suggested that RAC1 regulates endothelial cell function by responding to VEGF [277, 323, 326].

Indeed, activation of the RAC1 pathway has been shown to enhance VEGF-induced

neovascularization, whereas inhibition of this pathway prevents neovascularization in vitro [326].

Furthermore, activation of the RAC1 pathway in embryonic endothelial cells is critical for

embryonic angiogenesis, and failure to do so, results in embryo death [277]. Additionally, in other

cell systems such as pulmonary artery muscle cells, RAC1 can also stimulate Hif transcription

[202-204, 280, 325] in a PI3K-dependent pathway [202-204, 280], and indeed Rac1 promoter

Page 41: © 2013 Juanmahel Davila - IDEALS

26

appears to contain HIF1α response element [325]. Collectively these studies raise the interesting

possibility that RAC1 may play a role in uterine angiogenesis during early pregnancy by regulating

the expression of VEGF, via a HIF-mediated manner.

Although the cellular functions of RAC1 have been described in the literature, very little is known

about their function in the endometrium, especially during pregnancy. Recent studies have

suggested that RAC1 pathway is activated in human endometrial cells and may play a role in

embryo attachment and invasion into the endometrium in the human [7, 26]. Furthermore, in the

above studies, the authors concluded that the activation of the RAC1 pathway in the maternal

decidual cells is necessary to allow invasion of the trophoblasts [7, 26]. However, due to the

obvious ethical limitations, these studies were conducted in vitro and to the best of our knowledge,

the role of RAC1 has not been explored in vivo.

2.3.6 Available mouse models to study RAC1 function

Despite the availability of mice carrying a global knockout mutation of Rac1 gene, these mice are

not suitable to study the functional role of RAC1 in the uterus during early pregnancy. Global

deletion of Rac1 leads to embryonic lethality around the time of during gastrulation [271].

Therefore, to overcome this limitation, we have developed a mouse model system that has

provided us with the unique opportunity to study the functional and physiological role of RAC1

pathway in the uterus. We have made use of the Cre-LoxP technology, to specifically target the

deletion of Rac1 to specific tissues, thereby circumventing the embryonic lethality associated with

the global knockout mutations of Rac1 [271]. This lethality is not seen in global knockouts for

Rac2 [290] and Rac3 [299]. It should be noted that RAC2 and RAC3 mutants are fertile, and have

not shown any indications of infertility or any other reproductive impairments [290, 299]. Our

studies indicate that in contrast to RAC2 and RAC3, RAC1 plays a critical role in maintenance of

pregnancy.

Page 42: © 2013 Juanmahel Davila - IDEALS

27

2.4 The aims of the dissertation

To establish the functional role of uterine RAC1 during early events of pregnancy, I have used the

PgrCre/+ Rac1f/f mouse model to specifically target Rac1 deletion in any tissue expressing Pgr,

which includes the uterus. The following aims were established and are further discussed in this

dissertation:

Aim 1. Analyze the expression of RAC1 in the uterus during early pregnancy.

1a. Determine the uterine cellular compartment that expresses RAC1 during early

pregnancy.

1b. Determine whether Rac2 and Rac3, the other members of the Rac subfamily, are

expressed in the murine uterus during early pregnancy.

Aim 2. Characterize the defects in fertility and identify the biological processes that prevent

pregnancy maintenance in Rac1-null female mice.

2a. Determine the timing of embryo demise in Rac1-ablated uteri during early pregnancy.

2b. Perform an in depth analysis to determine whether conditional deletion of Rac1 in the

murine uterus leads to improper stromal cell differentiation.

2c. Investigate whether the deletion of uterine Rac1 impairs neovascularization in the

endometrium during early pregnancy.

2d. Determine whether conditional deletion of Rac1 prevents trophoblast migration and

formation of placenta.

Aim 3. Identify the pathways that mediate RAC1 function during decidualization.

3. Perform gene expression profiling using endometrial RNA from control and Rac1-null

uteri on day 8 of gestation to identify the RAC1-regulated pathways involved in

angiogenesis during decidualization

Page 43: © 2013 Juanmahel Davila - IDEALS

28

2.5 Chapter Figures

FIGURE 2.1: HORMONE PROFILE DURING PREGNANCY IN MICE. Schematic representation of the critical events of pregnancy in mice and the

hormone profile that accompany and regulate these events. The red line shows the hormone level for E2, whereas the blue line shows the hormone

levels of P4. The inset picture, shows an embryo during the time of implantation attached to the luminal epithelium of the uterus.

Page 44: © 2013 Juanmahel Davila - IDEALS

29

FIGURE 2.2: COMPARISON OF THE EARLY EVENTS OF PREGNANCY IN HUMANS AND MICE. After fertilization, the zygote makes its way to the

uterus, which takes about four days in mice and five to seven days in humans, prior to embryo implantation. For pregnancy timing in mice, our

laboratory defines day 1 of pregnancy as the day of fertilization (others refer to it as E0.5). Around day 5 of pregnancy (in mice), and day 8 of

pregnancy (in humans), the embryo begins the implantation process into the endometrium. Image adapted with permission from © 2001 Terese

Winslow [336].

Page 45: © 2013 Juanmahel Davila - IDEALS

30

FIGURE 2.3: MORPHOLOGICAL CHANGES DURING STROMAL CELL DIFFERENTIATION. (A) Diagrammatic representation of the early and mid-

stages of pregnancy in mice. Image adapted with permission from [337]. AM = antimesometrial side; M = mesometrial side; E = embryo; PDZ =

primary decidual zone (highlighted by dash-line); secondary decidual zone; DS = deep stromal cells. Images are at the same magnification.

Page 46: © 2013 Juanmahel Davila - IDEALS

31

FIGURE 2.3: MORPHOLOGICAL CHANGES DURING STROMAL CELL DIFFERENTIATION (CONTINUED). (B) Shown here are cross-sections of

control uteri stained with hematoxylin and eosin at the beginning of embryo implantation and early stromal cells differentiation (day 4 midnight)

and during the later stages of stromal cell differentiation (day 7 of pregnancy). In mice, embryo attachment and invasion occurs in the antimesometrial

side of the uterus, i.e. the side opposite to the mesometrial ligament of the uterus, triggers the fibroblastic stromal cells to differentiate into an

epitheloid and highly secretory decidual cells. Of note, decidualization does not occur at inter-implantation sites. AM = antimesometrial side; M =

mesometrial side; E = embryo; PDZ = primary decidual zone (highlighted by dash-line); secondary decidual zone; DS = deep stromal cells. Images

are at the same magnification.

Page 47: © 2013 Juanmahel Davila - IDEALS

32

FIGURE 2.4: MAJOR EVENTS DURING IMPLANTATION AND DECIDUALIZATION. Shown here are cross-sections of control uteri, on day 8 of

pregnancy, stained for ALPL activity (blue staining), PRL8A2/dPRP (red staining enclosed by dash-line), and PECAM1/CD31 (red staining), which

are well-known markers of the process of decidualization and vascular remodeling, respectively. AM = antimesometrial decidua; M = mesometrial

decidua; E = embryo; LS = lateral blood sinusoid. Images are at the same magnification.

Page 48: © 2013 Juanmahel Davila - IDEALS

33

FIGURE 2.5: DEVELOPMENTAL STAGES OF THE MOUSE PLACENTA. Lineage specification and differentiation of the trophectodermal

(extraembryonic) cells begin at on day 4 of pregnancy [embryonic day (E) 3.5] with the formation of the blastocyst. Around day 9 (E8.5) of

pregnancy, chorion and allantois attach and this is followed by formation of the placental labyrinth, via branching morphogenesis. The mature

(definitive) placenta is formed by day 15 (E14.5) of pregnancy, and it consists of three layers: the labyrinth, the spongiotrophoblast, and the maternal

decidua. E0.5 = day 1 of pregnancy. AM = antimesometrial decidua; M = mesometrial decidua, Image adapted and modified with permission from

[28].

Page 49: © 2013 Juanmahel Davila - IDEALS

34

FIGURE 2.6: STRUCTURAL COMPARISON BETWEEN MEMBERS OF THE HUMAN RHO FAMILY OF GTPASES. The Rho family of GTPases (RHOA

[338], RAC1 [339], and CDC42 [340]) are structurally similar to the prototype GTPase, RAS GTPase [341]. Three-dimensional images based on

X-ray diffraction crystallography for these molecules were extracted, modified, and adapted from the NCBI Molecular Modeling Database [342].

Page 50: © 2013 Juanmahel Davila - IDEALS

35

FIGURE 2.7: REGULATORY MECHANISMS OF THE RHO FAMILY OF GTPASES. 1) In the resting state cells, the non-activated GTPase is complexed

with a guanine nucleotide dissociation inhibitor (GDI), which maintains the protein inactive in the cytosol by preventing the translocation to the

plasma membrane. 2) Upon a signal, the GTPase is released from GTPase-GDI complex and the membrane association of the protein is promoted

due to the hydrophobic tail in C-terminus. 3) At the same time, cycling between an inactive GDP-bound and an active GTP-bound mediated by

interactions with a guanine nucleotide exchange factors (GEF). The activated membrane-associated proteins interact with downstream effectors to

activate signal transduction pathways that lead to diverse cellular functions. 4) Association with GTPase activating proteins (GAPs) accelerates the

hydrolysis of GTP, attenuating the activity of the GTPase. Once the cycle is complete and the protein is no longer needed, it will again form a

complex with a GDI.

Page 51: © 2013 Juanmahel Davila - IDEALS

36

FIGURE 2.8: PROTEIN SEQUENCE COMPARISON BETWEEN HUMAN AND MOUSE RAC1. (A) Shown here is the comparison between the 192 amino

acid protein sequence for human (GI 8574038) and mouse (GI 403115559) RAC1. As shown all 192 amino acid in the sequence match.

Page 52: © 2013 Juanmahel Davila - IDEALS

37

FIGURE 2.8: PROTEIN SEQUENCE COMPARISON BETWEEN HUMAN AND MOUSE RAC1 (CONTINUED). (B) The dot plot shows the graphical

comparison between the two amino acid sequences. The “x”-axis represent each amino acids of the human RAC1 sequence and the “y”-axis shows

those of the mouse RAC1 sequence. The protein sequences for human and mouse RAC1 were obtained from NCBI Protein and aligned using NCBI

BLAST. The alignment and analysis were done using the onboard tool in BLAST [343, 344].

Page 53: © 2013 Juanmahel Davila - IDEALS

38

CHAPTER 3: GENERATION AND PHENOTYPIC

ANALYSIS OF RAC1D/D MUTANT

FEMALE MICE

Page 54: © 2013 Juanmahel Davila - IDEALS

39

Abstract

Members of the Rho GTPase family are pleiotropic signaling molecules that regulate a wide range

of cellular processes, including cell cycle, cell differentiation, cell motility (through actin

cytoskeleton remodeling), and angiogenesis. Recent in vitro studies suggested that one of the

members of this superfamily such as Ras-related C3 botulinum toxin substrate 1 (RAC1),

expressed by decidual cells, might be capable of influencing embryo implantation in the human.

Although these studies suggested that RAC1 activity is critical for trophoblast invasion in vitro, it

is not yet known whether RAC1 exhibits similar functions in vivo. Using a conditional deletion

approach (Cre-loxP system), we generated transgenic mice that lack Rac1 gene expression and

protein in uterine cells that are positive for progesterone receptor (PgrCre/+ Rac1f/f or Rac1d/d). Our

results showed that conditional deletion of uterine Rac1 led to a severe subfertility phenotype. We

observed more than 90% reduction in birth rate in Rac1d/d mice when compared to control Pgr+/+

Rac1f/f (Rac1f/f) female mice. Initial assessment of implantation nodules during early pregnancy

(day 6 to 8) revealed that embryo attachment and gross morphology of implantation nodules were

similar in the two genotypes. However, in depth histological analysis of uterine sections on day 8

of gestation revealed dysregulation of stromal differentiation in the uteri of Rac1d/d mice. In

response to aberrant stromal differentiation, we observed that between day 8 and 10 of pregnancy,

a significant number of the implanted embryos became hemorrhagic and had begun to undergo

resorption. By day 15 of pregnancy, most embryos had been lost and the few that survived,

developed abnormal placentas and were surrounded by hemorrhagic maternal tissue. It should be

noted that the fertility defect in Rac1d/d females is due to maternal shortcomings rather than an

embryonic defect. Crossing of PgrCre/Cre Rac1f/f males x Pgr+/+ Rac1f/f females, which produced

Rac1d/d embryos, resulted in viable offspring without pregnancy complications. Whereas a number

of previous studies have shown that a defect in the embryo severely affects placentation and

pregnancy outcome, we are reporting for the first time that a perturbation in the maternal

differentiation program of Rac1d/d uteri adversely affects trophoblast function and maintenance of

pregnancy. Future studies will address an in depth analysis of this new mouse model to improve

our understanding of the cause and consequence of pregnancy complications in the human.

Page 55: © 2013 Juanmahel Davila - IDEALS

40

Introduction

Implantation of the embryo to the uterine wall followed by placentation are pivotal events for

establishment and maintenance of pregnancy [1-3, 110]. These processes involve steroid-

dependent complex crosstalk between the embryo and the maternal endometrium, which in turn

supports the growing conceptus, and ensure reproductive success [2, 4, 5, 8, 107, 115]. Failure in

any of the events can compromise maintenance of pregnancy and lead to miscarriage [5-7, 26, 32,

143, 345]. In fact, clinically, implantation failure and spontaneous abortion following implantation

are among the leading causes of infertility in women of reproductive age and reveals a major

challenge for assisted reproductive technology [6, 7, 38, 39]. In many instances the reasons for

implantation failure are unknown.

In rodents, as the embryo breaches the luminal epithelium following attachment, the underlying

stromal cells undergo a dramatic transformation to form a specialized tissue, known as the decidua

[5, 23, 25, 29, 117, 154, 346]. During decidualization, the fibroblastic stromal cells proliferate and

then differentiate into mature secretory decidual cells that support embryo growth and survival

until placentation [23-25, 29, 121-123]. In addition, the decidual tissue undergoes extensive

remodeling to accommodate the growing embryo. Concomitant with the differentiation process,

as the decidua forms around the implanted embryo, endothelial cells in the uterine stroma

proliferate to create an extensive vascular network, which is embedded in the decidua and is critical

for embryo development [5, 23, 25, 29, 32, 117, 154, 346]. It is clear that decidualization is a key

event in establishment of early pregnancy, however, the signaling molecules that participate in the

formation and function of this tissue remain poorly understood.

Microarrays are powerful molecular tools that allow rapid screening of gene expression changes

under different experimental conditions. Using this tool, we compared differentially expressed

genes in the uterus during decidualization and identified Ras-related C3 botulinum toxin substrate

1 (RAC1) as one of the genes whose expression is markedly elevated at the time decidualization.

While a previous study performed in vitro, implicated a potential role of RAC1 in embryo invasion,

its role in vivo during implantation remains unknown [7, 26]. We, therefore, decided to explore

the in vivo function of RAC1 in the uterus during early pregnancy.

Page 56: © 2013 Juanmahel Davila - IDEALS

41

RAC1, a pleiotropic signaling molecule and a member of the Rho family of GTPases, has been

shown to regulate many cellular processes. These include cell cycle, cell-cell adhesion, cell

differentiation, cell motility through actin cytoskeleton remodeling, endothelial cell function, as

well as angiogenesis [113, 242, 250, 254, 261, 262, 269, 271-280, 301]. Because decidualization

encompasses many of these physiological events, it is possible that RAC1 regulates one or more

of these processes during early pregnancy. In support of this notion, Grewal et al. [7, 26] has shown

recently that invasion of the human embryo into endometrial stroma, in vitro, involves the motility

of endometrial stromal cells and this movement is dependent on the activity of Rho GTPases,

especially RAC1. Here, we show that RAC1 is necessary for early pregnancy, as uterine loss of

this molecule led to severe subfertility. However, contrary to the previous in vitro observations,

we now show that RAC1 activity is not necessary for embryo attachment or invasion into the

uterine stroma to initiate embryo implantation.

Page 57: © 2013 Juanmahel Davila - IDEALS

42

Materials and Methods

Animals and Tissue Collection

All animal breeding, housing, procedures, experiments, and euthanasia were approved and

conducted according to the guidelines of the Institutional Animal Care and Use Committee of

University of Illinois at Urbana-Champaign. The health and care of the animals was provided by

the veterinarians and staff of Division of Animal Resources. Rac1 conditional null mice were

generated using Cre-LoxP strategy [347] (Figure 3.1A). Mice expressing homozygous copies of

the Rac1 gene flanked by loxP sites [Pgr+/+ Rac1f/f (official gene symbol Rac1tm1Djk/J), or Rac1f/f

from now onwards] [267] were obtained from Dr. Ann Sutherland (University of Georgia) in a

C57BL/6-129SV background. Mice carrying the Cre Recombinase under the control of the

progesterone receptor (Pgr) promoter [348] [PgrCre/+ Rac1+/+ (official gene symbol Pgrtm2(cre)Lyd/+),

or PgrCre from now onward], were obtained from Drs. John Lyndon and Francesco De Mayo

(Baylor College of Medicine) in a C57BL/6 background. To establish the mouse colony for this

study, initial breeding between the two mouse lines was done to obtain the F1 progeny, which are

heterozygous for Pgr and Rac1 (Appendix A). Female mice used for this study were generated by

breeding male mice of specific genotypes from the F2 progeny (see Figure 3.1B) with Rac1f/f

female mice. Genotyping of the animals was performed following the protocols established by the

respective laboratories that generated the transgenic mice [267, 348] (Appendix A). Conditional

knockout female mice (PgrCre/+ Rac1f/f) will be referred to as Rac1d/d from now onwards.

Rac1f/f or Rac1d/d females were euthanized by CO2 asphyxiation, followed by a blood draw via

cardiac puncture, and cervical dislocation. Uteri and ovaries from pregnant mice or from mice

subjected to experimentally-induced decidualization (see below) were collected at different time

points during pregnancy or following the differentiation stimulus. The harvested organs were

either fixed for histological evaluation [immersion-fixed in 10% neutral-buffered formalin (NBF)],

flash frozen in liquid N2 for RNA or frozen sectioning, or the decidual mass was harvested for

gene expression studies. For samples collected at different time points during pregnancy,

identification of a copulatory plug following mating was used as a reference and for our

experimental purposes, to indicate day 1 of pregnancy.

Page 58: © 2013 Juanmahel Davila - IDEALS

43

Some of the frozen implantation nodules, collected on day 8 of pregnancy, were thawed by

submerging them in 1x phosphate-buffer saline (PBS) at room temperature for morphological

evaluation of the decidua. The decidual mass was exposed by mechanical removal of the

myometrium. Pictures were taken using a Leica M165 FC dissection scope connected to a Leica

450 C camera with c-mount interface containing a 5 Megapixel CCD sensor. Similarly, for

morphological evaluation of embryos on day 15 of pregnancy, mechanical removal of the

myometrium was done on samples that had been fixed in 10% NBF under the dissecting scope, as

mentioned above.

Fertility testing

To determine whether conditional mutation of Rac1 in uterine tissues had any effect in female

fertility, Rac1f/f and Rac1d/d mice of reproductive age (8 weeks or more) were paired with proven

wild-type males for a period of six months (Appendix A). The total number of pups born in each

litter as well as the number of pregnancies during this period were recorded.

Experimentally-induced decidualization

To test whether RAC1 plays any role during decidualization, 8 week old female mice were

subjected to experimentally induced decidualization protocol as described previously [32, 154,

349-352]. Briefly, adult, non-pregnant, Rac1f/f and Rac1d/d female mice were ovariectomized under

general anesthesia and rested for two weeks following ovariectomy to remove any circulating

hormones. After two weeks of rest, the animals were injected subcutaneously (s.c.) with 100 ng of

17β-estradiol (E2) in 100 µL corn oil daily for three days, then rested for two days. Following

resting period, mice were injected with a solution containing 6.7 ng E2 and 1 mg P4 per 100 µL

(E2 + P4) corn oil s.c. for three days. On the third day of E2 + P4 injection, decidualization was

initiated, under general anesthesia, in one horn by injection of 50 μL oil, whereas the other horn

was left unstimulated to serve as an internal control. Daily injections of E2 + P4 were continued

for up to an additional 96 h post-stimulation. Mice were euthanized and uterine horns were

collected and weighed at different time points following the decidualization stimulus.

Page 59: © 2013 Juanmahel Davila - IDEALS

44

Hormonal treatment

To test whether Rac1 gene expression is affected by hormonal treatment, Rac1f/f mice were

subjected to different doses of steroid hormones as described previously [353]. Briefly, female

mice were subjected to bilateral ovariectomy. Two weeks later, the mice were injected

subcutaneously with 100 µL of vehicle (oil), E2 (100 ng), P4 (1 mg) or combination of E2 + P4.

The animals were then euthanized at 6 h and 24 h following E2 alone; after 2 doses daily of P4; or,

in case of the combination, 2 daily doses were given, followed by E2, then tissue was collected 6h

and 24 h after the last injection.

Alkaline Phosphatase Activity Assay

The protocol for detecting alkaline phosphatase (ALPL) activity was followed as previously

published [352, 354], with modifications. Briefly, frozen uterine sections were incubated in 10%

NBF for 10 min at room temperature then washed in 1x PBS three times for 5 min each (3x 5 min.)

The slides were then incubated with a solution containing 0.1 M Tris-HCl pH 8.5, 0.5mM naphthol

AS-MX phosphate disodium salt (ALPL substrate), and 1.5 mM fast blue RR (4-benzoylamino-

2,5-dimethoxyaniline diazonium) salt at 37 oC for 30 min. The uterine alkaline phosphatase

activity releases orthophosphate and naphthol derivatives from the substrate. The naphthol

derivatives are simultaneously coupled with diazonium salt present in the incubating medium to

form a dark blue dye, which marks the site of enzyme action. To terminate the enzymatic reaction,

the tissue sections were rinsed in tap water, mounted using crystal mount medium, and visualized

using an Olympus BX51 microscope equipped for light microscopy connected to a Jenoptik

ProgRes C14 digital camera with c-mount interface containing a 1.4 Megapixel CCD sensor.

Immunohistochemistry and Immunofluorescence

Paraffin-embedded or frozen uterine sections were subjected to immunohistochemistry as

described previously [32, 353, 355, 356]. Briefly, paraffin-embedded sections were deparaffinized

in xylene and rehydrated in a series of graded alcohols followed by rinsing in tap water. When

frozen tissues were used, the sections were thawed at room temperature for 5 min, then fixed for

Page 60: © 2013 Juanmahel Davila - IDEALS

45

another 5 min in 10% NBF. Sections were then rinsed in 1x PBS, followed by blocking in 10%

non-immunized serum for 1 h at room temperature. The slides were incubated with a solution

containing the diluted antibody (see Appendix E for details) and 1% serum in 1x PBS overnight at

4 oC in a humidified chamber. Following overnight incubation, slides were washed 3x 5 min with

1x PBS, then incubated with diluted secondary antibody (see Appendix E for details) for 1 h at

room temperature in a humidified chamber. When a chromogenic labeling was done, sections were

incubated in a solution containing 3% H2O2 in methanol to quench endogenous peroxidase. Slides

were then rinsed in 1x PBS, followed by a 30-45 min incubation with horseradish peroxidase-

conjugated streptavidin (Invitrogen). Slides were then exposed to chromogens (3, 3’-

diaminobenzidine or 3-amino-9-ethylcarbazole) until optimal signal was obtained. Following the

chromogenic labeling, the slides were counterstained with hematoxylin, and mounted. Slides that

were incubated with a fluorescent secondary antibody were mounted with a medium containing

DAPI. Pictures were taken using the Olympus BX51 microscope equipped for light and fluorescent

imaging and connected to a Jenoptik ProgRes C14 digital camera with c-mount interface

containing a 1.4 Megapixel CCD sensor. Fluorescent images were merged and processed using

CS4 Adobe Photoshop Extend (Adobe Systems).

RNA Isolation and Real-time quantitative PCR

Total RNA was isolated from tissues or cells using TRIzol reagent (Invitrogen) according to the

manufacturer’s instructions. Oligonucleotides specific for genes of interest were developed (see

Appendix F for details) and real-time quantitative PCR (qPCR) reactions were carried out using

SYBR-green master mix (Applied Biosystems) on a 7500 Applied Biosystems Real-time PCR

machine. Gene expression data analysis were done using the 2-ΔΔCt method [357] and 36b4 gene –

which is conserved among many species and encodes an acidic ribosomal phosphoprotein P0 that

interacts with 40S ribosomal subunit – was used as a housekeeping gene and loading control [358].

Endometrial Stromal Cell Isolation

Endometrial stromal cells were isolated from day 1 and 4 pregnancy samples as previously

described [164, 352]. Uteri were removed from mice after euthanasia with CO2. The uteri were cut

Page 61: © 2013 Juanmahel Davila - IDEALS

46

open and digested with a solution containing 6 g/L dispase (Gibco) and 25 g/L pancreatin (Sigma)

in 1x HBSS (Gibco) for 45min at room temperature, followed by 15 min at 37 oC, with occasional

mixing. Cells were washed and then incubated in 0.5 g/L collagenase (Sigma) in HBSS for 1 h at

37 oC. After incubation, the tubes were vortexed for 10-12 seconds until the supernatant became

turbid with dispersed cells. The contents of the tube were then passed through an 80-μm gauze

filter (Millipore) into a new tube. Cells were resuspended in Dulbeccos modified Eagle's medium-

F12 medium (Gibco) supplemented with 2% heat-inactivated fetal calf serum, 100 units/L

penicillin, 0.1 g/L streptomycin (Gibco), 1.25 mg/L fungizone, 10 nM E2, and 1 µM P4. The

numbers of live cells were assessed by trypan blue staining using a hemocytometer.

Western Blots

Mouse endometrial stromal cells or decidual masses were isolated from female mice at different

days of pregnancy. The cells from day 1 and 4 pregnant uteri were lysed in ice-cold lysis buffer

containing 1% NP40, 50 mM Tris-HCl pH 7.6, 125 mM NaCl, 1 mM EDTA, 1x protease inhibitor

cocktail. Decidual mass on D8 of pregnancy was lysed with lysis buffer aided by maceration using

a mortar and pestle. The cell debris was removed by centrifugation, and the protein concentration

of the lysate was determined by bicinchoninic acid protein assay according the manufacturer's

instruction (Pierce/Thermo/Fisher Scientific). The cell lysates were analyzed by standard Western

blotting, using antibodies against RAC1 (1:1000) and β-TUBULIN (1:5000, as loading control,

see Appendix E for antibody details).

Hormone assay

Serum P4 concentrations were measured for at least 3 animals per time point by radioimmunoassay

at the core facility of University of Virginia at Charlottesville.

Statistical analysis

Experimental data were collected from a minimum of three independent animals (not from the

same litter). Uterine weights, serum hormones, embryo perimeter, placental weight, and qPCR

Page 62: © 2013 Juanmahel Davila - IDEALS

47

results were expressed as mean ± S.E.M. Litter size were expressed as means. Statistical analysis

was done using Student’s t-test (for single comparison), one-way analysis of variance with a

Dunnett’s post-test (for multiple comparison between treatments or time points) or two-way

ANOVA with a Bonferroni post-test (for multiple comparison between different genotypes and

time points). An analysis of equal variances was done on all numerical data to determine whether

a parametric or non-parametric hypothesis test was appropriate. Data were considered statistically

significant at p ≤ 0.05, and is indicated by an asterisk in the figures. All data was analyzed and

plotted using GraphPad Prism 4.0 (GraphPad Software).

Page 63: © 2013 Juanmahel Davila - IDEALS

48

Results and Discussion

Rac1 is expressed in uterine stroma during decidualization, but its expression is independent of

steroid hormones

To gain insight into the molecular pathways underlying decidualization, we performed gene

expression profiling using RNA from uterine horns at different times after exposure to decidual

stimulation. Our studies revealed upregulation of Rac1 in response to decidual stimulation. To

confirm the results of the microarray analysis, we performed qPCR using RNA isolated from the

non-stimulated and stimulated horns at different times after decidual stimulation. As shown in

Figure 3.2A, Rac1 expression increased in response to decidual stimulation. A marked increase in

Rac1 expression was observed at 72 h post decidual stimulation. Consistent with this expression

profile, we observed a significant upregulation in Rac1 expression during normal pregnancy on

day 7 and day 8 of gestation (Figure 3.2B).

Because steroid hormones, estrogen (E2) and progesterone (P4) are known to influence

decidualization, we investigated whether Rac1 gene expression is regulated by steroid hormones.

Administration of E2, P4, or E2 + P4 to ovariectomized mice failed to induce Rac1 expression in

the uterus, indicating that it is not regulated by steroid hormones (Figure 3.2C). Interestingly,

previous studies have indicated the presence of response elements for several transcription factors,

including HIFs, in the Rac1 promoter [285]. It is possible that HIFs, which are induced during

stromal differentiation, are involved in the regulation of Rac1 in the uterus during early pregnancy.

However, further analysis of the Rac1 promoter during stromal cell differentiation will be needed

to validate this possibility.

We next monitored by western blot analysis the expression profile of RAC1 protein in the uterus

at different days of gestation. We observed a band corresponding to RAC1 protein on day 4 of

gestation. But consistent with the RNA profile, we observed a marked enhancement in RAC1

protein expression in uterine stromal cells on day 8 of pregnancy, overlapping the window of

decidualization (Figure 3.2D). Interestingly on day 8, we also noted the presence of a band

Page 64: © 2013 Juanmahel Davila - IDEALS

49

corresponding to phosphorylated RAC1 (pRAC1) in the western blot. The functional significance

of pRAC1 in the uterus remains unknown.

The Rac1d/d mouse model

To address the role of RAC1 in the uterus, we generated a mouse line where the Rac1 gene is

conditionally deleted using a Cre recombinase that is under the control of one copy of the

progesterone receptor promoter (termed PgrCre/+), whereas the other copy expresses functional

Pgr, which is sufficient to preserve fertility of these animals [348]. The conditional deletion

approach was used because the Rac1 global knockout is embryonic lethal [271]. Further, PgrCre

mouse line has been extensively used to ablate genes in specific cells lineages within the body [32,

345, 348, 353, 359-361]. Rac1 gene is conditionally excised in only those tissues that are positive

for Pgr, which includes the uterus. The term Rac1f/f and Rac1d/d will be used to refer to the control

and conditional null mice, respectively.

Ablation of Rac1 in the Rac1d/d mice was assayed by qPCR and our studies indicate efficient

ablation of Rac1 in the uteri on day 4 to day 10 of pregnancy (Figure 3.3A). Consistent with the

RNA profile, we observed a marked decline in the levels of RAC1 protein in Rac1d/d uteri on day

4 or day 8 of gestation (Figure 3.3B). Our results also indicate that the gene expression of other

members of the Rho family of GTPases, such as Rac2, Rac3, Rhoa, and Cdc42, were unaffected

upon conditional deletion of Rac1 (Figure 3.3C).

Conditional deletion of uterine Rac1 leads to severe subfertility

To determine the role of RAC1 in the regulation of fertility, a six-month breeding study was

performed by crossing Rac1d/d or Rac1f/f mice with proven wild-type males (see Appendix A for

details). At the completion of the study, we noted a severe subfertility phenotype in Rac1d/d female

mice compared to Rac1f/f controls. More than 90% of the pups were lost during pregnancy

indicating a drastic reduction in the litter size (Table 2.1). In addition, most Rac1d/d females

exhibited a drastic reduction in their fertility rate after the first pregnancy (data not shown), and

many females did not produce any litters after the second or third pregnancy. Although these

Page 65: © 2013 Juanmahel Davila - IDEALS

50

results are of interest to us, we have not investigated this issue any further as part of this dissertation

project.

Conditional deletion of Rac1 has no effect on ovarian function

Because progesterone receptor is transiently expressed in the granulosa cells of the ovary around

the time of ovulation [362-366], and given that RAC1 is ubiquitously expressed in many cell types,

it is conceivable Rac1 may be deleted from the ovary, resulting in insufficient ovarian function.

Therefore, to ensure that the severe subfertility phenotype, and pregnancy loss, observed in Rac1d/d

mice was not due to an ovarian defect, ovaries from days 4, 8, 10 (data not shown), 12, and 15 of

pregnancy were collected and evaluated histologically for signs of anovulation or lack of corpora

lutea (CL) (Figure 3.4). Ovaries collected from Rac1d/d females displayed follicles at all stages of

development and CL with normal appearance (Figure 3.4A). Moreover, we monitored Rac1 gene

expression in the ovary on day 4 of pregnancy, and consistent with the ovarian histology data,

there was no significant difference in Rac1 mRNA expression in the ovaries from Rac1f/f and

Rac1d/d mice (Figure 3.4B). We also determined the serum progesterone concentrations in the two

genotypes to ensure that the fertility defect was not due to a deficiency in circulating P4. Indeed,

no significant difference was found in serum P4 concentration between Rac1d/d and Rac1f/f mice

(Figure 3.4C). Moreover, we did not detect any significant difference in the average number of

blastocysts recovered from the uteri of Rac1f/f and Rac1d/d mice on day 4 of pregnancy (data not

shown).

We next investigated the effect of deletion of uterine Rac1 on embryo implantation. Previous

studies by Grewal et al. [7, 26] suggested that activation of the RAC1 pathway in the decidual cells

is required for implantation and trophoblast invasion and expansion. Interestingly, our studies

revealed that uterine deletion of Rac1 gene did not prevent attachment of the embryos into the

maternal endometrium (Figure 3.5A). We noted the presence of implanted embryos in the uterus

beginning on day 4 of pregnancy and, up to day 8 of pregnancy, we did not detect any overt

abnormality of the implantation nodules in the two genotypes. Interestingly, a closer histological

examination of uterine sections on different days of gestation revealed that starting on day 7, the

embryos in Rac1d/d mice started to display abnormality at the implantation site and this became

Page 66: © 2013 Juanmahel Davila - IDEALS

51

more pronounced on day 8 of pregnancy. Staining of uterine sections on day 8 with cytokeratin,

which marks the trophoblast cells showed aberrant overexpansion of trophoblast cells in maternal

stroma (Figure 3.5B and Appendix C). At this time, we have not fully characterized the

overexpansion phenotype of the embryos. However, it is possible that the dysregulated trophoblast

expansion could be a result of improper differentiation of uterine stroma due to ablation of RAC1.

A number of previous studies have implicated RAC1 as a regulator of critical cellular processes,

including cell proliferation and differentiation [242, 261, 269, 273, 276, 280, 301, 367]. Because

stromal cell differentiation is a critical event for establishment of pregnancy, we determined

whether conditional deletion of uterine Rac1 affected this process by employing a well-established

differentiation model that is independent of the embryo [154]. We observed that the gross decidual

response in Rac1d/d uteri was comparable to that of control Rac1f/f uteri (Figure 3.6, lower panel).

During decidualization, stromal cells proliferate, exit the cell cycle, and terminally differentiate

into mature decidual cells [346, 368-372]. The progression of this differentiation program can be

traced by monitoring the expression of biochemical markers in the uterus. As cells enter the

differentiation program, they express liver/bone/kidney alkaline phosphatase (ALPL) (Figure 2.3).

We further investigated the decidual response in Rac1f/f and Rac1d/d uteri by monitoring the

expression of ALPL. As expected, uterine sections from Rac1f/f mice displayed prominent ALPL

activity at 48 h after the administration of decidual stimulus (Figure 3.6B, upper panel).

Interestingly, we noted a dramatic induction of ALPL activity in Rac1d/d uteri at similar time point,

indicating dysregulated stromal differentiation program in the absence of uterine RAC1 (Figure

3.6B, lower panel).

We next examined the role of RAC1 in stromal decidualization during normal pregnancy. We did

not observe any difference in cell proliferation on day 8 of pregnancy, as indicated by MKI67

staining (Figure 3.7C). However, consistent with the observation that the stromal differentiation

program is dysregulated by the loss of uterine RAC1, we noted a marked upregulation in Alpl gene

expression (Figure 3.7A) as well as activity (Figure 3.7B) in Rac1d/d uteri in comparison to Rac1f/f

controls. We also examined the expression of other markers of differentiation including Wnt4 and

Prl8a2/dPrp during decidualization (Figures 3.7 and Appendix B). While the expression of Wnt4

Page 67: © 2013 Juanmahel Davila - IDEALS

52

was upregulated (Figure 3.7A) in Rac1d/d uteri, we observed a slight downregulation in the

expression of PRL8A2/dPRP protein (Figure 3.7D) in uterine sections of Rac1d/d mice compared

to Rac1f/f controls on day 8 of pregnancy. A reduction in Prl3c1/Plpj gene expression was also

noted (Figure 3.7A). PRL3C1/PLPJ has been reported to be produced by the decidual cells

immediately surrounding the embryo [373] and may be implicated in decidual cell development

and the endometrial vasculature [374] around day 8 of pregnancy. We also monitored the

expression of Alpl and Wnt4 in cultured stromal cells collected from Rac1f/f and Rac1d/d uteri on

day 4 of pregnancy and subjecting them to in vitro decidualization. Again, similar to the pregnancy

samples, our results revealed a marked enhancement in Alpl and Wnt4 expression in stromal cells

from Rac1d/d uteri (Figures 3.8). Collectively, these results indicate that in the absence of uterine

RAC1, the stromal differentiation program during pregnancy goes awry.

The severe subfertility in Rac1d/d is due to maternal but not embryonic defect

How do we rule out the possibility that the subfertility in Rac1d/d mice is not due to an inherent

defect in the embryo? A number of previous studies have shown that a defect in the embryo

severely affects trophoblast migration and placentation [9, 59, 129, 130, 132, 300]. However, to

the best of our knowledge, only a limited number of mouse models exist in the literature in which

a perturbation in the maternal decidua, and not the embryo, adversely affects trophoblast function

and placentation [32, 359, 360, 375-378]. In that regard, the Rac1d/d mice are unique. Thus, to

avoid any complication of potential non-specific deletion of Rac1 gene in the embryo, we bred the

Rac1d/d and Rac1f/f females to wild-type male mice of proven fertility. This breeding scheme

generated heterozygous embryos for Rac1 gene (see Appendix A for details), therefore any

problems associated with pregnancy are likely due to a defect in the maternal decidua, and not in

the embryo or its membranes. Our breeding studies further indicated that embryo loss occurred

around day 10 to 12 of pregnancy, and appear as hemorrhagic implantation nodules (Figures 3.9

and 3.10A). It should be noted, however, that not all embryos died at this time point. A small

percentage of the embryos managed to survive to later stages of gestation (Figure 3.10C), and even

to be born (Table 3.1). It is intriguing that some embryos managed to survive, whereas others die.

One observation we have consistently made is that there is a cleft that remains in the uteri of

Rac1d/d mice (Figure 3.10B), possibly from defective migration of decidual cells. In Rac1d/d uteri,

Page 68: © 2013 Juanmahel Davila - IDEALS

53

this area is highly hemorrhagic and many neutrophils infiltrate the luminal space of the cleft, while

a thin decidua surrounds the embryo in the mesometrial side (data not shown). An interesting

possibility is that in Rac1d/d uteri, the decidua fails to form an immune barrier from the allogenic

embryo [23, 29, 149-151], resulting in embryo resorption and termination of pregnancy.

Studies performed in the early 1990s showed that the mouse blastocyst expresses detectable Pgr

mRNA expression [379]. However, we ruled out any possibility that a potential deletion of Rac1

in the embryonic tissues adversely influenced the outcome of pregnancy because of the following

reasons. First, heterozygous females for both Rac1 and PgrCre (PgrCre/+ Rac1f/+) are fertile (Table

3.2), and when bred to either PgrCre/+ Rac1f/f or PgrCre/Cre Rac1f/f males they 1) produced litters

with the expected genotypes (based on genetic expectancy of the crossing; see Appendix A for

details); 2) they produced viable offspring, which were later used for breeding purposes; and 3)

pups with a PgrCre/Cre Rac1f/f genotype were generated in this crossing. In addition, crossing of

Rac1f/f females with either PgrCre/+ Rac1f/f, PgrCre/Cre Rac1f/f, or Pgr+/+ Rac1+/+ males of proven

fertility, showed that Rac1f/f females carrying Rac1d/d embryos were fertile and produced viable

litters (Figure 3.11), strongly suggesting that the fertility defect observed in the mutant females is

due to a maternal rather than an embryonic defect. Collectively, these results indicate that the

maternal environment due to dysregulated stromal differentiation in Rac1d/d uteri is not conducive

to embryo growth. In the next chapter, we have investigated how impaired stromal differentiation

is affecting maternal environment leading to embryo demise in uteri lacking RAC1.

Page 69: © 2013 Juanmahel Davila - IDEALS

54

Chapter Figures and Tables

FIGURE 3.1: BREEDING SCHEME TO GENERATE THE CONDITIONAL MUTANT FOR RAC1 USING CRE-LOXP STRATEGY. (A) The gene construct

for Rac1 gene and the primer position for genotyping strategy are shown. Floxed and wild-type bands are detected with the primer combination

P91/P33, whereas the conditional knockout band is detected with the primer combination P45/P33 [347]. Triangles indicate the LoxP sites.

Page 70: © 2013 Juanmahel Davila - IDEALS

55

FIGURE 3.1: BREEDING SCHEME TO GENERATE THE CONDITIONAL MUTANT FOR RAC1 USING CRE-LOXP STRATEGY (CONTINUED). (B) The

breeding scheme for the generation of the experimental animals is shown. Briefly, male mice, with specific genotypes, were bred to Rac1f/f females

to generate the conditional mutant. In these animals, exon 1, which contains the translation initiation site and part of the GTPase activity of RAC1,

will be conditionally excised in all cells that express Pgr gene.

Page 71: © 2013 Juanmahel Davila - IDEALS

56

FIGURE 3.2: EXPRESSION PROFILE FOR RAC1 GENE AND PROTEIN IN MICE. (A – C) Whole uterus gene expression profile for Rac1 after

experimentally-induced decidualization (panel A), hormonal stimulation (panel B), and during pregnancy (panel C). (D) RAC1 protein expression

profile from isolated endometrial stromal cells on day 4 of pregnancy or decidual cells on day 8 of pregnancy; day 1 shows a similar profile as day

4 of pregnancy (data not shown). As expected, both gene and protein increase in expression during cell differentiation, but are unaffected by hormonal

treatment alone. n ≥ 3 per time point. Asterisks indicate significant differences from 0 h on panel A or day 1 of pregnancy in panel B (p < 0.01).

Page 72: © 2013 Juanmahel Davila - IDEALS

57

FIGURE 3.2: EXPRESSION PROFILE FOR RAC1 GENE AND PROTEIN IN MICE (CONTINUED). (A – C) Whole uterus gene expression profile for Rac1

after experimentally-induced decidualization (panel A), hormonal stimulation (panel B), and during pregnancy (panel C). (D) RAC1 protein

expression profile from isolated endometrial stromal cells on day 4 of pregnancy or decidual cells on day 8 of pregnancy; day 1 shows a similar

profile as day 4 of pregnancy (data not shown). As expected, both gene and protein increase in expression during cell differentiation, but are

unaffected by hormonal treatment alone. n ≥ 3 per time point. Asterisks indicate significant differences from 0 h on panel A or day 1 of pregnancy

in panel B (p < 0.01).

Page 73: © 2013 Juanmahel Davila - IDEALS

58

FIGURE 3.3: RAC1 IS EFFICIENTLY DELETED FROM THE UTERUS USING PGRCRE. (A) Whole uterus gene expression profile demonstrates an

efficient knockdown of Rac1 gene in Rac1d/d mice in samples obtained at different times during pregnancy. (B) RAC1 is efficiently ablated in

endometrial stromal cells on day 4 of pregnancy, as well as on decidual cells on day 8 of pregnancy. Residual RAC1 protein may come from

contamination from other cell populations (like uterine natural killer cells), which migrate into the uterus around day 8 of pregnancy.

Page 74: © 2013 Juanmahel Davila - IDEALS

59

FIGURE 3.3: RAC1 IS EFFICIENTLY DELETED FROM THE UTERUS USING PGRCRE (CONTINUED). (C) Whole uterus gene expression profile on day

8 of pregnancy shows that Rac1 is the only member that is affected by the conditional deletion; other members of the Rho family of GTPases are

not affected by the conditional deletion. n ≥ 3 per time point. Asterisks indicate significant differences from controls (**, p < 0.01, ***p < 0.001).

Page 75: © 2013 Juanmahel Davila - IDEALS

60

TABLE 3.1: IMPACT OF RAC1 DELETION ON FEMALE FERTILITY

Genotype # Females † # Pups # Litters

Average

Pups/Litter ‡

Pgr+/+

Rac1f/f

7 315 31 10

PgrCre/+

Rac1f/f

7 19 11 2

† One Rac1d/d

female did not produce any litters during or after the breeding study ‡ p < 0.0001

Page 76: © 2013 Juanmahel Davila - IDEALS

61

FIGURE 3.4: CONDITIONAL DELETION OF RAC1 DOES NOT AFFECT OVARIAN FUNCTION. (A) Representative histological samples of ovaries from

Rac1f/f and Rac1d/d on days of pregnancy (D) 4, 8, 12, and 15 stained with H&E. No differences in the ovarian morphology were observed at these

time points. Bars = 200 µm.

Page 77: © 2013 Juanmahel Davila - IDEALS

62

FIGURE 3.4: CONDITIONAL DELETION OF RAC1 DOES NOT AFFECT OVARIAN FUNCTION (CONTINUED). (B) Consistent with the observation in

Panel A, no significant difference ovarian was observed in Rac1 gene expression by D4. (C) Serum progesterone (P4) concentration was unaffected

by the conditional deletion of Rac1. n = 3 per time point. (p > 0.05).

Page 78: © 2013 Juanmahel Davila - IDEALS

63

FIGURE 3.5: CONDITIONAL DELETION OF RAC1 HAS DOES NOT PREVENT EMBRYO ATTACHMENT AND INVASION INTO THE ENDOMETRIUM. (A

and B) Shows representative samples from Rac1f/f and Rac1d/d on day 8 of pregnancy. No gross morphological difference was observed between

conditional null and control mice (panel A).

Page 79: © 2013 Juanmahel Davila - IDEALS

64

FIGURE 3.5: CONDITIONAL DELETION OF RAC1 HAS DOES NOT PREVENT EMBRYO ATTACHMENT AND INVASION INTO THE ENDOMETRIUM

(CONTINUED). (A and B) Also, uterine deletion of Rac1 gene does not prevent embryo implantation (panel B). CYTOKERATIN 8 was used as a

general marker of trophoblast cells to highlight the implanted embryo. Bars = 500 µm. n ≥ 5.

Page 80: © 2013 Juanmahel Davila - IDEALS

65

FIGURE 3.6: CONDITIONAL DELETION OF UTERINE RAC1 DOES NOT AFFECT THE FORMATION OF THE DECIDUOMA, BUT DISPLAYS

DYSREGULATION IN ALPL ACTIVITY. (A) No difference was detected in the wet weight of the deciduoma induced experimentally in conditional

Rac1-null mice compared to controls (p > 0.05). Shown here are representative sample after 96 h post stimulus uteri. The right horn was stimulated,

whereas the left horn served as an internal control and was not stimulated. (B) Conditional deletion of uterine RAC1 led to a dysregulated increase

in the activity of the early marker of decidualization, kidney/liver/bone alkaline phosphatase (ALPL) activity at all the observed time points. Shown

here is uterine ALPL activity 48 h post stimulus. n = 3 per time point.

Page 81: © 2013 Juanmahel Davila - IDEALS

66

FIGURE 3.7: CONDITIONAL DELETION OF RAC1 LEADS TO A DYSREGULATED STROMAL CELL DIFFERENTIATION. (A) Gene expression assay

was done on day 8 of pregnancy to access the expression of well-known makers that are differentially expressed during stromal cell differentiation.

Critical genes that are known to be expressed during the early stages of differentiation, i.e. Alpl and Wnt4, show a dysregulated gene expression.

Prl3c1/Plpj a putative marker of late stromal cell differentiation was reduced (p = 0.14), as compared to control uteri. Other genes involved in the

differentiation of stromal cells, were not significantly changed at this time point of pregnancy. Asterisks indicate significant differences from controls

(**, p < 0.01, ***p < 0.001).

Page 82: © 2013 Juanmahel Davila - IDEALS

67

FIGURE 3.7: CONDITIONAL DELETION OF RAC1 LEADS TO A DYSREGULATED STROMAL CELL DIFFERENTIATION (CONTINUED). (B) Alkaline

phosphatase (ALPL) activity is a well-known indication of early stromal cell differentiation, and persist during the differentiation of stromal cells.

Our data show that there is noticeable elevation in both Alpl gene expression (panel A) and activity in day 8 pregnancy samples. Similar observations

were made on day 7 of pregnancy (data not shown). Bars = 500 µm.

Page 83: © 2013 Juanmahel Davila - IDEALS

68

FIGURE 3.7: CONDITIONAL DELETION OF RAC1 LEADS TO A DYSREGULATED STROMAL CELL DIFFERENTIATION (CONTINUED). (C & D) It is

well established that during the differentiation process, decidualizing stromal cells rapidly proliferate and quickly enter the differentiation program

[346, 368-372]. Our data indicate that stromal cell from Rac1d/d uteri are capable of initiating the differentiation into decidual cells, as no difference

in MKI67 (panel C), a maker of cell proliferation. However, despite no change in gene expression, uteri deficient for Rac1 display a reduced

expression of PRL8A2/dPRP (panel D), a marker of late stromal cell differentiation, when compared to control uteri. The area below the dash mark

indicate the area negative for MKI67, which positive cells have a reddish/brown nuclear staining. Bars = 200 µm (panel C and D).

Page 84: © 2013 Juanmahel Davila - IDEALS

69

FIGURE 3.7: CONDITIONAL DELETION OF RAC1 LEADS TO A DYSREGULATED STROMAL CELL DIFFERENTIATION (CONTINUED). (C & D) It is

well established that during the differentiation process, decidualizing stromal cells rapidly proliferate and quickly enter the differentiation program

[346, 368-372]. Our data indicate that stromal cell from Rac1d/d uteri are capable of initiating the differentiation into decidual cells, as no difference

in MKI67 (panel C), a maker of cell proliferation. However, despite no change in gene expression, uteri deficient for Rac1 display a reduced

expression of PRL8A2/dPRP (panel D), a marker of late stromal cell differentiation, when compared to control uteri. PRL8A2/dPRP is highlighted

by the cells with a red cytoplasmic stain under the dash mark. Bars = 200 µm (panel C and D). n ≥ 4.

Page 85: © 2013 Juanmahel Davila - IDEALS

70

FIGURE 3.8: ACTIVATION OF THE DECIDUAL CELL RAC1 PATHWAY IS NECESSARY FOR STROMAL CELL MORPHOLOGY AND NORMAL

DIFFERENTIATION. Shown here are cultured uterine decidual cells, under differentiation conditions for 72 h. As shown here, cells, where Rac1 gene

has been deleted (Rac1d/d), show abnormal cell morphology, compared to control cells (Rac1f/f). Similarly, during the differentiation of decidual

cells, gene expression of factors, likes alkaline phosphatase (Alpl) and Wnt4 are known to be expressed, but shown here is a dysregulated gene

expression of these factors in the mutant decidual cells; other factors, like decidual prolactin-related protein (Prl8a2/dPRP) gene expression remain

unchanged. Blue = DAPI, which stains the nucleus, and green = Phalloidin, which stains actin filaments. n = 3. Asterisks indicate significant

differences from controls (p < 0.001).

Page 86: © 2013 Juanmahel Davila - IDEALS

71

FIGURE 3.9: REPRESENTATIVE UTERI SAMPLES AT DIFFERENT STAGES OF PREGNANCY. Uteri samples at different times of pregnancy were

collected from Rac1f/f and Rac1d/d on days 8 through 15 of pregnancy. Black arrows point hemorrhagic implantation nodules, whereas blue arrow

head highlight lateral hemorrhage, which commonly seen in hemochorial placentation. n ≥ 3.

Page 87: © 2013 Juanmahel Davila - IDEALS

72

FIGURE 3.10: SEVERE HEMORRHAGE IS OBSERVED IN IMPLANTATION NODULES FROM RAC1D/D UTERI. (A) The graph shows a survival curve

analysis of the onset of hemorrhagic uteri collected from day 4 of pregnancy. Hemorrhagic implantation nodules are typically noticed as early as

day 7 of pregnancy.

Page 88: © 2013 Juanmahel Davila - IDEALS

73

FIGURE 3.10: SEVERE HEMORRHAGE IS OBSERVED IN IMPLANTATION NODULES FROM RAC1D/D UTERI (CONTINUED). (B) Shown here are

representative samples of pregnancy nodules on day 8 of pregnancy from control, conditional knockout, and heterozygous uteri after the nodules

were liberated from the myometrium to expose the decidua. Severe hemorrhage is typically observed in the conditional knockout decidua, whereas

the control and heterozygous uteri appear do not display this severe bleeding. Bars = 10 mm.

Page 89: © 2013 Juanmahel Davila - IDEALS

74

FIGURE 3.10: SEVERE HEMORRHAGE IS OBSERVED IN IMPLANTATION NODULES FROM RAC1D/D UTERI (CONTINUED). (C) Representative samples

implantation nodules on day 15 of pregnancy from control and conditional knockout females after the nodules were liberated from the rest of the

uterine horn to expose each embryo and the placenta (P). Severe hemorrhage are observed in nodules obtained from conditional knockout uteri,

whereas the control uteri appear do not show signs of hemorrhaging. Also, note that the placenta of the conditional knockout female have many

focal hematoma (H), and this is typically observed in these females. Of note, the hemorrhage observed in the pregnancy nodule originates from small

hemorrhages in the maternal tissues (M) surrounding the placenta and chorioallantoic membranes (CA) of the embryo. Once the maternal layers are

removed and the fetal chorioallantoic membranes are exposed, no hemorrhage is observed. Bars = 5 mm. n ≥ 3.

Page 90: © 2013 Juanmahel Davila - IDEALS

75

TABLE 3.2: SINGLE OR EPITHELIAL-SPECIFIC RAC1 ALLELE DELETION DOES NOT IMPACT FERTILITY

Genotype # Females # Pups # Litters Average

Pups/Litter ‡

Pgr+/+

Rac1f/f

7 307 31 10

PgrCre/+

Rac1f/f

7 19 11 2

PgrCre/+

Rac1f/+

2†

76 12 6

Wnt7aCre/+

Rac1f/f

3 141 15 9

† The data shown represent the breeding performance over a six-month period. Different from the Rac1f/f

, Rac1

d/d, and Wnt7a

Cre/+ Rac1

f/f females, the

data collected for PgrCre/+

Rac1f/f

(heterozygous) females was from the current breeding colony, where these females are paired to PgrCre/Cre

Rac1f/f

or

PgrCre/Cre

Rac1f/+

males, not wild-type males.

This table is for comparison purposes only. Because of the limitation in samples of heterozygous, appropriate statistical analysis could not be

performed. Data collected from Wnt7aCre females were collected at a time PgrCre and control females. Data from heterozygous females was

collected from the available data from the breeding colony, which at the time was experiencing breeding problems. Nonetheless, the data here show

that heterozygous females are fertile and capable of producing viable litters.

Page 91: © 2013 Juanmahel Davila - IDEALS

76

FIGURE 3.11: PREGNANCY DEFECT OBSERVED IN RAC1 CONDITIONAL-NULL UTERI IS NOT DUE TO AN EMBRYONIC DEFECT. (A) To determine

if the potential expression of Pgr in the blastocyst is sufficient to produce any of the defects that are seen in Rac1d/d females, uteri were collected

from control (Pgr+/+ Rac1f/f) female mice on day 8 of pregnancy after they were mated with either homozygous Cre (PgrCre/Cre Rac1f/f), floxed (Pgr+/+

Rac1f/f), or wild-type (Pgr+/+ Rac1+/+) male mice. Homozygous Cre males bred to control females produce offspring that have the same genotype as

the conditional null mice (PgrCre/+ Rac1f/f), while wild-type males bred to control females, produce offspring that are similar to the experimental

conditions of the mating for this study (Pgr+/+ Rac1f/+). In all, the gestation nodules collected were normal, without any alteration to the expected

uterine gross morphology.

Page 92: © 2013 Juanmahel Davila - IDEALS

77

FIGURE 3.11: PREGNANCY DEFECT OBSERVED IN RAC1 CONDITIONAL-NULL UTERI IS NOT DUE TO AN EMBRYONIC DEFECT (CONTINUED). (B)

Uteri in panel A were further dissected and representative implantation nodules, and the correspondent decidual masses and embryos are shown. No

morphological perturbations were observed. Bars = 1 mm. n ≥ 2 per genotype

Page 93: © 2013 Juanmahel Davila - IDEALS

78

CHAPTER 4: RAC1 IS A CRITICAL REGULATOR OF

UTERINE ANGIOGENESIS DURING

EARLY PREGNANCY IN THE MOUSE

Page 94: © 2013 Juanmahel Davila - IDEALS

79

Abstract

In mice, following implantation, the uterine stromal cells differentiate into specialized decidual

cells, which support the growth of the embryo before placentation. Concomitantly, new blood

vessels begin to form in the maternal decidua from pre-existing vasculature through angiogenesis,

an absolutely critical event for the establishment of placenta and maintenance of pregnancy. It is

clear that decidualization and angiogenesis play crucial roles during early gestation, however, the

complex molecular pathways underlying these processes remain unknown. Our recent studies

revealed that Rac1 (Ras-related C3 botulinum toxin substrate 1), a pleiotropic signaling factor, is

induced in uterine stroma during decidualization and may play a role in regulating this process. To

address the role of RAC1 during pregnancy, we generated a conditional knockout of the Rac1 gene

in the uterus by crossing mice carrying floxed Rac1 with PgrCre mice. A six-month breeding study

indicated a severe defect in fertility of the Rac1-ablated (Rac1d/d) females. Further analysis showed

hemorrhagic uteri starting on day 7 of pregnancy and by day 10 of gestation, resorption of embryos

was clearly evident. An in depth analysis showed dysregulation of the stromal differentiation

program during decidualization in Rac1d/d uteri. Interestingly, the loss of Rac1 expression in

uterine stromal cells also resulted in a marked impairment in the development of maternal blood

vessels as indicated by the expression of PECAM1/CD31, Hif2α, Angpt2, and NRP1. This uterine

angiogenic defect is, presumably, one of the main contributors to pregnancy loss seen in Rac1d/d

mice at mid-gestation. Based on these results, we propose that RAC1 controls the differentiation

of stromal cells to decidual cells, which produce critical local factors that impact on blood vessel

formation at the maternal-fetal interface during early pregnancy.

Page 95: © 2013 Juanmahel Davila - IDEALS

80

Introduction

Decidual neovascularization, the process by which new blood vessels originate from pre-existing

vessels, during the post-implantation period is an absolute requirement for pregnancy maintenance

in humans and rodents [73]. Indeed, it is thought that the newly formed uterine vasculature during

decidualization functions as an early site of nutrient and gas exchange for the developing embryo,

until the placenta becomes functionally competent [57, 74]. Although the details underlying the

process of decidualization and angiogenesis are slightly different between humans and rodents, the

basic sequence of events and many of the molecular pathways that regulate these processes are

similar between the two species.

In the pregnant uterus, angiogenesis is a critical process that precedes placentation, and requires

signals that originate from the embryo and the endometrium [1, 5, 9, 10, 29, 32, 75-77, 360].

Among the many signaling molecules, hypoxia-inducible transcription factor (HIF), vascular

endothelial growth factor (VEGF), and angiopoietins (ANGPTs) play major roles in endometrial

vascular remodeling during decidualization. Interestingly, hypoxia is a detectable feature in the

uterine environment during embryonic development [199-201]. Oxygen homeostasis is critical for

cell survival and in response to hypoxic conditions, cells activate the transcription of Hif1α to

restore oxygen homeostasis [206, 207, 327-334]. Activation of HIF stimulates Vegf expression,

which promotes angiogenesis [203, 205, 327, 329, 332, 333, 335]. Several lines of evidence

suggest that under hypoxic conditions, RAC1 regulates endothelial cell function by responding to

VEGF signaling [323]. Activation of the RAC1 pathway, using a dominant active form of RAC1,

has been shown to enhance VEGF-induced neovascularization, whereas inhibition of RAC1

activity prevents neovascularization in vitro [326]. Further, activation of RAC1 in embryonic

endothelial cells is critical for embryonic angiogenesis, and global mutations of RAC1 lead to

defective vascularization and embryonic death [277]. Interestingly, in other cell systems such as

pulmonary artery muscle cells, RAC1 has been shown to stimulate Hif transcription [202-204, 280,

325] via the PI3K-dependent pathway [202-204, 280]. Collectively, these studies raise the

interesting possibility that RAC1 may play a role in angiogenesis in decidual tissues by regulating

VEGF expression.

Page 96: © 2013 Juanmahel Davila - IDEALS

81

To gain an insight into the pathways that are affected by the ablation of Rac1 in uterine stroma and

lead to termination of pregnancy, we performed gene expression profiling of decidual RNA

collected from Rac1-intact and Rac1-null uteri on day 8 of gestation. We focused on day 8 because

severe hemorrhage and embryo loss occur by day 10 of pregnancy, suggesting that the processes

that are leading to embryo demise are most likely arising on day 8 of pregnancy. Consistent with

RAC1’s role in angiogenesis, our studies revealed that RAC1 is a critical regulator of this process

in the uterus. Ablation of uterine RAC1 led to a dysregulation in vascular network formation in

the maternal decidual bed due to defective expression and secretion of pro-angiogenic factors.

Page 97: © 2013 Juanmahel Davila - IDEALS

82

Materials and Methods

Animals and Tissue Collection

All animal breeding, housing, procedures, experiments, and euthanasia were approved and

conducted according to the guidelines of the Institutional Animal Care and Use Committee of

University of Illinois at Urbana-Champaign. The health and care of the animals was provided by

the veterinarians and staff of Division of Animal Resources. Rac1 conditional null mice were

generated using Cre-LoxP strategy [347] (Figure 3.1A). Mice expressing homozygous copies of

the Rac1 gene flanked by loxP sites [Pgr+/+ Rac1f/f (official gene symbol Rac1tm1Djk/J), or Rac1f/f

from now onwards] [267] were obtained from Dr. Ann Sutherland (University of Georgia) in a

C57BL/6-129SV background. Mice carrying the Cre Recombinase under the control of the

progesterone receptor (Pgr) promoter [348] [PgrCre/+ Rac1+/+ (official gene symbol Pgrtm2(cre)Lyd/+),

or PgrCre from now onward], were obtained from Drs. John Lyndon and Francesco De Mayo

(Baylor College of Medicine) in a C57BL/6 background. To establish the mouse colony for this

study, initial breeding between the two mouse lines was done to obtain the F1 progeny, which are

heterozygous for Pgr and Rac1 (Appendix A). Female mice used for this study were generated by

breeding male mice of specific genotypes from the F2 progeny (see Figure 3.1B) with Rac1f/f

female mice. Genotyping of the animals was performed following the protocols established by the

respective laboratories that generated the transgenic mice [267, 348] (Appendix A). Conditional

knockout female mice (PgrCre/+ Rac1f/f) will be referred to as Rac1d/d from now onwards.

Rac1f/f or Rac1d/d females were euthanized by CO2 asphyxiation, followed by a blood draw via

cardiac puncture, and cervical dislocation. Uteri from pregnant mice were collected at different

time points during pregnancy. The harvested organs were either fixed for histological evaluation

[immersion-fixed in 10% neutral-buffered formalin (NBF)], flash frozen in liquid N2 for RNA or

frozen sectioning, stromal cell isolation, or decidual mass was harvested for gene expression

studies. For samples collected at different time points during pregnancy, identification of a

copulatory plug following mating was used as a reference and for our experimental purposes, to

indicate day 1 of pregnancy.

Page 98: © 2013 Juanmahel Davila - IDEALS

83

For microarray analysis, decidual nodules from day 8 of pregnancy were isolated following blood

perfusion, as previously described [380-382]. Briefly, day 8 pregnant mice were anesthetized with

ketamine-xylazine cocktail (87 mg ketamine; 15 mg xylazine / kg body weight). After the mice

were unconscious, a V-shape incision expanding radially to the chest was made so that the uterus,

liver, and ribs were visible. The chest cavity was opened and the heart was exposed. Immediately,

the mouse was perfused through the left ventricle of the heart, while the blood drained through the

right atrium, with at approximately 20 mL of Hank’s balanced salt solution containing heparin

(HBSS+H , 30 USP Units / mL) until the liver and uterine implantation nodules were visually

cleared of blood. Following perfusion, the uterine horns were dissected and placed in HBSS+H

for separation of the decidual mass. With the use of a dissection microscope (Leica M165 FC), the

uterine myometrium was mechanically removed with jeweler forceps and the decidual mass

containing the embryo was excised. Once all the masses were separated from the muscle, they

were transferred into a new 35 mm dish with fresh HBSS+H to remove any leftover blood and the

embryo. To remove the embryo, the isolated decidual mass was rotated so that one of the two

mesometrial indentations was visible. Then, using the indentation as a guide, the decidua was

carefully opened and the embryo was removed from antimesometrial side by carefully lifting it

out of the decidual mass. The decidual masses, freed from the embryos, were transferred to a new

35 mm dish with fresh HBSS+H for a final rinse before collected and flash frozen for further RNA

isolation. Four decidual masses/animal were collected for microarray analysis.

Microarray and expression analysis

Total RNA was extracted from the isolated and perfused decidual masses with TRIzol (Invitrogen)

following the manufacturer's protocol. The RNA preparations were then purified with the RNeasy

kit (Qiagen). Samples were stored in RNase-free water until further use. To evaluate the purity and

quality of the isolated RNA, the samples were submitted to the Roy J. Carver Biotechnology

Center (at the University of Illinois) for assessment of RNA quality and chip hybridization for

microarray analysis. Quality of the RNA samples were assessed using an Agilent Bioanalyzer

System and samples with a RNA integrity number of 10 were used for microarray analysis using

a GeneChip Mouse Genome 430A 2.0 Array (Affymetrix), containing approximately 14,000

annotated gene sequences. Subsequently, the chips were scanned and the data were extracted using

Page 99: © 2013 Juanmahel Davila - IDEALS

84

Gene-Chip Operating Software version 1.3 (Affymetrix), which later were exported to Microsoft

Excel 2013 (Microsoft Corporation). Expression values for each gene below 50 was considered as

background. Relative gene expression fold value was determined by the ratio of gene expression

of Rac1d/d decidua to Rac1f/f decidua. A list of genes that had a relative fold change of +/- 1.5, were

further analyzed for gene ontology and functional classification using the Database for Annotation,

Visualization and Integrated Discovery (DAVID, National Institute of Allergy and Infectious

Diseases, National Institutes of Health) [383].

Quantitative PCR gene validation and RNA isolation

Total RNA was isolated from whole uterine samples, as reported above. RNA was converted to

cDNA using a kit (Applied Biosystems), according to the manufacturer’s instructions.

Oligonucleotides specific for genes of interest were developed (see Appendix F for details) and

real-time quantitative PCR (qPCR) reactions were carried out using SYBR-green master mix

(Applied Biosystems) on a 7500 Applied Biosystems Real-time PCR machine (Applied

Biosystems). Gene expression data analysis were done using the 2-ΔΔCt method [357] and 36b4

gene – which is conserved among many species and encodes an acidic ribosomal phosphoprotein

P0 that interacts with 40S ribosomal subunit – was used as a housekeeping gene and loading

control [358].

Immunohistochemistry, Immunocytochemistry, and Immunofluorescence

Cultured cells, paraffin-embedded or frozen-sectioned uterine sections were subjected to

immunohistochemistry or immunocytochemistry as described previously [32, 353, 355, 356].

Briefly, paraffin-embedded sections were deparaffinized in xylene and rehydrated in a series of

graded alcohols followed by rinsing in tap water. When frozen tissues were used, the sections were

thawed at room temperature for 5 min, then fixed for 5 min in 10% NBF at room temperature. If

cells were used, they were fixed for 5 min in 10% NBF at room temperature. Sections or cells were

then rinsed in 1x phosphate-buffer saline (PBS), followed by serum blocking using 10% non-

immunized serum for 1 h at room temperature. Following serum block, slides were incubated with

a solution containing the diluted antibody (see Appendix E for details) and 1% serum in 1x PBS

Page 100: © 2013 Juanmahel Davila - IDEALS

85

overnight at 4 oC in a humidified chamber (for slides) or in the culture plate (for cells). Following

overnight incubation, slides were washed 3 times, 5 min each, with 1x PBS, then incubated with

diluted secondary antibody (see Appendix E for details) for 1 h at room temperature. When

chromogenic labeling was done, the samples were incubated in a solution containing 3% H2O2 in

methanol to quench endogenous peroxidase. Samples were then rinsed in 1x PBS, followed by a

30-45 min incubation with horseradish peroxidase-conjugated streptavidin (Invitrogen). The

samples were then exposed to chromogens (3, 3’-diaminobenzidine or 3-amino-9-ethylcarbazole)

until optimal signal was obtained. Following the chromogenic labeling, the samples were

counterstained with hematoxylin and mounted. When immunofluorescence was done, the samples

were incubated with a fluorescent secondary antibody were mounted with a medium containing

DAPI. Pictures were taken using the Olympus BX51 microscope equipped for light and fluorescent

imaging and was adapted to a Jenoptik ProgRes C14 digital camera with c-mount interface

containing a 1.4 Megapixel CCD sensor or an Olympus Ix70 inverted microscope adapted to

Diagnostic Instrument digital camera containing a 2.0 Megapixel CCD sensor. Fluorescent images

were merged and processed using CS4 Adobe Photoshop Extend (Adobe Systems).

Primary endometrial stromal cell isolation and culture

Endometrial stromal cells were isolated, from three independent mice per genotype on day 4 of

pregnancy as previously described [164, 352]. Briefly, uteri were removed from mice after

euthanasia with CO2. The uteri were flushed with 200 – 300 µL of HBSS to remove embryos and

to confirm pregnancy. Then, the uteri were cut open and digested with a solution containing 6 g/L

dispase (Gibco) and 25 g/L pancreatin (Sigma) in 1x HBSS (Gibco) for 45min at room

temperature, followed by 15 min at 37 oC, with occasional mixing. The enzyme activities were

quenched by adding equal volumes of 10% FBS solution, then cells were washed three times with

1x HBSS (Gibco). Cells were then incubated in 0.5 g/L collagenase (Sigma) in HBSS for 1 h at

37 oC. After incubation, the tubes were vortexed for 10-12 seconds until the supernatant became

turbid with dispersed cells. The contents of the tube were then passed through an 80-μm gauze

filter (Millipore) into a new tube. Cells were resuspended and plated for cell culture in Dulbeccos

modified Eagle's medium-F12 medium (Gibco) supplemented with 2% heat-inactivated fetal calf

serum, 100 units/L penicillin, 0.1 g/L streptomycin (Gibco), 1.25 mg/L fungizone, 10 nM E2, and

Page 101: © 2013 Juanmahel Davila - IDEALS

86

1 µM P4. The numbers of live cells were assessed by trypan blue staining using a hemocytometer.

Cells were plated for 2 hours at a density of 5 x 105 cell/well in 6-well culture plates to allow

stromal cells, but not epithelial cells, to attach. After the 2 hour incubation (0 h of culture), the

medium was aspirated and the cells were rinsed with two to three changes of fresh media, until

most of the unattached cells were removed. Media were again changed at 24 h (and stored for a

baseline reading) and 48 h after the initial change (0 h). Following the 48 h media change, the cells

were cultured for up to 96 h from the initial plating (0 h), and media were collected in 24 h

intervals. The media from three-wells were pooled and collected at 24, 72, and 96 h time point,

and the cellular debris was removed via centrifugation, and stored for ELISA. In addition, the cells

were collected by rinsing with HBSS followed by incubation with 0.25% trypsin at 37 oC until the

cells detached. The cells and media were stored at -80 oC until needed. The numbers of live cells

were determined using trypan blue staining and a hemocytometer, then, the cells were placed in

TRIzol reagent (Invitrogen) for further RNA isolation.

Measurement of vascular-endothelial growth factor A (VEGFA) in culture medium

Measurements of VEGFA production in vitro were done from pooled media from three wells (from

a 6-well plate, Corning) from at least three independent cell cultures (one culture = one animal),

plated at an initial density of 5x105 cell/well. Enzyme-linked immuno sorbent assay (ELISA) for

mouse VEGF were done according to the manufacturer recommendations (R&D Systems).

Statistical analysis

Experimental data were collected from a minimum of three independent animals (not from the

same litter), which were subjected to the same experimental conditions. ELISA and qPCR results

were expressed as mean ± S.E.M. Statistical analysis was done using Student’s t-test. An analysis

of equal variances was done in all numerical data to determine whether a parametric or non-

parametric hypothesis test was appropriate. Data were considered statistically significant if p ≤

0.05, and is indicated by an asterisks in the figures. All data were analyzed and plotted using

GraphPad Prism 4.0 (GraphPad Software) or Microsoft Excel 2013 (Microsoft Corporation).

Page 102: © 2013 Juanmahel Davila - IDEALS

87

Results and Discussion

Microarray analysis reveals impaired angiogenic network in Rac1-ablated uteri

We previously established that the severe subfertility phenotype observed in the Rac1d/d female

mice was due to a defect in maternal environment. Our studies also revealed that embryo loss

occurred between days 10 to 12 of pregnancy in Rac1d/d females, suggesting that the disruption of

events preceding these dates are critical for maintenance of gestation. During this window, several

biological processes are known to occur in the uterus. These include ongoing differentiation of the

decidual cells, extensive vascular remodeling and angiogenesis [23, 31, 57, 166, 167], trophoblast

proliferation and invasion into the endometrium, as well as formation of the placenta [5, 9, 28, 78,

112, 126, 128, 129, 131-133] (Figures 1.4 and 1.5). We employed the microarray approach to

identify the genes and the regulatory pathways that might be affected by Rac1 deletion in the

endometrium. The microarray analysis was performed by isolating RNA from decidua of Rac1-

intact and Rac1-null uteri on day 8 of pregnancy.

Out of the approximately 14,000 well-characterized genes that were analyzed in the 430A

microarray chip, 1345 genes were differentially regulated (± 1.5 fold). Out of the differentially

regulated genes, 796 were down-regulated in Rac1-null decidua compared to Rac1-intact controls.

Gene ontology was performed by using NIH DAVID for analysis [383]. We identified multiple

biological processes, including angiogenesis, which appeared to be regulated by RAC1 in the

decidua during pregnancy (Figure 4.1).

The expression of several genes that are known to play major roles in neovascularization in the

endometrium during early pregnancy were affected by the loss of RAC1 [32, 194, 384]. Validation

of the microarray data showed that expression of these genes including hypoxia-inducible factor 2

alpha (Epas1/Hif2α) [207, 385], angiopoietin 2 (Angpt2), and Angpt4 were markedly reduced in

the absence of uterine expression of Rac1 (Figure 4.2). The expression of other genes involved in

angiogenesis, including sphingosine kinase 1 (Sphk1), neuropilin 1 (Nrp1), and the clotting factor

XIII subtype A1 (F13a1) were also attenuated by the loss of uterine Rac1 (Figure 4.2).

Intriguingly, however, the expression of hypoxia-inducible factor 1 alpha (Hif1α) and vascular-

Page 103: © 2013 Juanmahel Davila - IDEALS

88

endothelial growth factor A (Vegfa), which are well known regulators of angiogenesis (Figure 4.3),

remained unchanged. Nonetheless, the significant alterations in transcription of genes associated

with uterine angiogenesis led to a marked reduction in vascular network formation, as noted by

the reduced expression of platelet endothelial cell adhesion molecule 1 (PECAM1/CD31) on day

8 (Figure 4.4A) or day 10 of pregnancy (Figure 4.4B).

Whereas the expression of Vegf was unaltered, we observed a marked downregulation of

angiopoietin 2 in the absence of RAC1 in the uterine stroma. Angiopoietins, acting on the

angiopoietin receptor, TEK/TIE2, work along with VEGF to promote new blood vessel formation

from pre-existing ones [30, 58, 63, 64, 72, 180, 184, 197]. Interestingly, decidual tissues of women

with recurrent miscarriages show imbalance in the expression ratio of Angpt2 and Angpt1, to favor

Angpt1 rather than Angpt2 [63], which is predominant during uterine angiogenesis. ANGPT2 plays

a role in branching angiogenesis, which may be involved in the development of complex vascular

network seen in the uterus during pregnancy [58, 63, 64]. Although the precise role of

angiopoietins in uterine angiogenesis remains unknown, previous studies have shown that genetic

deletion of Angpt2 causes capillary dysmorphogenesis in the kidney [386], as well as abnormal

patterning of the blood vessels of the retina in mice [387].

The actions of VEGF are mediated through several receptors, primarily VEGFR1/FLT1 and

VEGFR2/ FLK1. VEGFs can also activate neuropilin receptors including NRP1 [168, 178, 184,

185]. Our studies revealed that along with Angpt2, neuropilin 1 gene and protein expression was

severely compromised in the absence of uterine Rac1 (Figures 4.2 and 4.5). Previous studies have

shown that in response to VEGF stimulation, NRP1 plays a role in guidance of neuronal and

endothelial cells during the process of neurogenesis, which is usually accompanied by

angiogenesis [388, 389]. Studies have also shown that mutant embryos, deficient for Nrp1, display

disorganization of the vasculature, without affecting the formation of primary vascular network or

differentiation of endothelial cells [390]. The fact that Nrp1 is significantly downregulated in the

decidua (Figure 4.5), suggests that migration and proliferation of endothelial cells might be

affected in the uteri of Rac1d/d mice. It is possible that during decidualization, RAC1-dependent

pathways in the maternal stroma are involved in the regulation of endothelial cell reorganization.

Page 104: © 2013 Juanmahel Davila - IDEALS

89

Ablation of Rac1 in the uterus leads to a decreased secretion of pro-angiogenic factors

To gain insights into the underlying mechanisms of the angiogenic defect observed in Rac1d/d

females, we used a well-established in vitro cell culture system of decidualization [164, 352].

Stromal cells isolated from Rac1-intact and Rac1-null uteri cultured in vitro showed distinct

differences in morphology and architecture as indicated by staining for F-actin using phalloidin

(Figure 3.7). It is known that actin cytoskeletons regulate certain aspects of vesicle trafficking and

protein secretion [248, 255, 257, 260, 308, 314, 316, 391, 392]. Indeed, one of the described

functions of RAC1 is to promote the formation polymerization of actin fibers to regulate vesicle

transport [247, 255, 263, 308, 314, 391]. We, therefore, explored the possibility that RAC1 may

be involved in secretion of proteins such as VEGFA in decidual cells. We analyzed the protein

expression of VEGFA by immunocytochemistry in cultured mouse endometrial stromal cells that had

been subjected to decidualization in vitro. Our results show that Rac1-null cells display an impaired

vesicular transport of VEGFA. Stromal cells from conditional knockout uteri displayed an

accumulation of VEGFA in the cellular compartment where Golgi/rER are normally located within the

cell (Figure 4.6A). Consistent with this observation, measurement of VEGFA in the supernatant of

mature decidual cells revealed a significant reduction in VEGFA production and secretion in Rac1-

null cells (Figure 4.6). Vegfa gene expression, however, was similar in Rac1-intact and Rac1-null

stromal cells. The precise stage at which RAC1 regulates the secretory activity of decidual cells

warrants further investigation. However, previous data has implicated RAC1 in trafficking secretory

vesicles from the Golgi to the plasma membrane [308, 310, 314]. Thus, it is highly possible that

RAC1’s actions are at the final stages of vesicular transport.

It is believed that RAC1 is not involved in the early stages of secretory pathway (i.e. rER to Golgi)

[310], nor has it been localized to the rER membrane. However, RAC does play a role in the later

stages of secretion. Recently, PREX1, a PI3K-dependent Rac exchange factor, has been identified

as a novel regulator of the glucose transporter 4 (SLC2A4/GLUT4) trafficking in adipocytes.

PREX1 can activate RAC1, and this in turn stimulate actin polymerization and facilitate

SLC2A4/GLUT4 transport to the plasma membrane [314]. Additionally, RAC1 has also been

reported to regulate exocytosis in a calcium-dependent manner [316, 318, 392]. The precise

mechanisms and the cellular compartments in which RAC1 elicits its effect to regulate exocytosis

Page 105: © 2013 Juanmahel Davila - IDEALS

90

is still controversial. However, it is clear that actin polymerization plays a critical role in this

process.

Conditional deletion of Rac1 in the uteri leads to severe hemorrhage

Despite a severe reduction of the vascular network on day 8 of pregnancy, we consistently observed

an increase in blood hemorrhage in Rac1-null decidua, especially around the lateral sinusoids (Figures

2.6 and 3.7). Using eosin-Y with phloxine to labels red blood cells, we noticed that the cells were

extravascular, indicative of vascular leakage and hemorrhage (Figure 4.7). Whereas Rac1f/f decidual

blood was mostly cleared following blood perfusion, Rac1d/d decidua consistently failed to clear,

indicative of hemorrhage (Figure 4.8). Interestingly, sphingolipid signaling has been shown to prevent

vascular leakage during uterine angiogenesis [377]. The fact that members of the sphingolipid pathway

such as Sphk1 are down regulated in the Rac1d/d uteri, suggests that attenuation of sphingolipid

pathways functioning downstream of RAC1 may be involved in the vascular leakiness observed in

Rac1d/d uteri (Figure 4.2). In addition to factors in the sphingolipid pathway, tight junction proteins in

endothelial cells are known to regulate vascular permeability [277]. Interestingly, our microarray

analysis identified a tight junction molecule such as claudin 3, whose expression was significantly

downregulated in the absence of uterine Rac1 (Figure 4.2). Claudin 3 (CLDN3) has been shown to be

present in the blood-brain barrier and loss of Cldn3 in the endothelial cells compromises the integrity

of the blood-brain barrier [393]. Exactly how stromal RAC1 regulates claudins in the endothelial cells

remains to be determined, but it is known that decidual cells secrete factors that affect endothelial cell

function [30, 57, 170, 177, 178, 194, 198, 207].

It is also possible that the severe hemorrhage observed in Rac1-null uteri is because of the reduction

in the expression of F13a1 – the gene coding for coagulation factor XIII subtype A1, which is the last

zymogen to become activated in the blood coagulation cascade and is the factor responsible for

crosslinking fibrin molecules that stabilize the blood clot. F13a1 global knockout female mice are

available and are able to get pregnant. However, breeding studies of these mice show that a severe

uterine hemorrhage develops around day 10 to 12 of pregnancy, which often leads to maternal death

[384]. Interestingly, there are also loss-of-function mutations in the human isoforms of F13 (A and B

subunits) [394]. It has been reported that certain women with recurrent miscarriage possess one or both

of these mutations. The fact that in Rac1d/d mice, there is a significant reduction in this enzyme,

Page 106: © 2013 Juanmahel Davila - IDEALS

91

suggests that RAC1 signaling may be crucial for F13A1 production and/or secretion in the uterus

during early pregnancy.

Page 107: © 2013 Juanmahel Davila - IDEALS

92

Chapter Figures

FIGURE 4.1: BIOLOGICAL PROCESS LIST OF GENES THAT WERE DOWN-REGULATED LESS THAN 1.5 FOLD ON MICROARRAY. Gene list shown

here is a condensed list of genes that were clustered by biological by NIH DAVID. The gene list is based on 576 unique gene probes out of which

259 gene probes were charted based on their known function within a biological process. The size of the pie slices are representative of the number

genes within each biological process. Some gene probes are contain within more than one biological process. Not all biological process are listed in

this pie chart.

Page 108: © 2013 Juanmahel Davila - IDEALS

93

FIGURE 4.2: CONDITIONAL DELETION OF RAC1 IN THE UTERUS LEADS TO DOWN-REGULATION OF SEVERAL GENES THAT ARE CRITICAL FOR

UTERINE ANGIOGENESIS. Gene expression analysis of day 8 of pregnancy uterine samples confirms the microarray analysis, which showed that

critical genes involved in uterine vascularization and angiogenesis during pregnancy are critically reduced in Rac1d/d compared to Rac1f/f uteri. Genes

such as Epas1/Hif2α, Angtp2, and Nrp1 are typically expressed during vascular remodeling in the pregnant uterus. n ≥ 3. Asterisks indicate significant

differences from controls (* p < 0.05, ** p < 0.01, *** p < 0.001).

Vegfr1/Flt1

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Vegfr2/Flk1

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Pecam1/Cd31

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Epas1/Hif2

Rac1f/f

Rac1d/d

0

1

2

*

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Angpt1

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Angpt2

Rac1f/f

Rac1d/d

0

1

2

**R

ela

tiv

e G

ene

Ex

pre

ssio

n

(2-

Ct )

Angpt4

Rac1f/f

Rac1d/d

0

1

2

***

Rela

tive G

en

e E

xp

ress

ion

(2-

Ct )

Sphk1

Rac1f/f

Rac1d/d

0

1

2

*

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Nrp1

Rac1f/f

Rac1d/d

0

1

2

*

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Cldn3

Rac1f/f

Rac1d/d

0

1

2

*

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

F13a1

Rac1f/f

Rac1d/d

0

1

2

*R

ela

tiv

e G

ene

Ex

pre

ssio

n

(2-

Ct )

Mmp9

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Page 109: © 2013 Juanmahel Davila - IDEALS

94

FIGURE 4.3: EXPRESSION OF TWO WELL-KNOWN GENE TARGETS OF RAC1 ARE UNAFFECTED. Under hypoxic conditions, RAC1 has been shown

previously to regulate the expression of Hif1α, indirectly leading to an increased production of Vegfa gene expression, via HIF1α. Interestingly, in

the uterus, conditional deletion of Rac1 does not affect the expression of these two genes on day 8 of pregnancy. n ≥ 3. (p > 0.05).

Hif1

Rac1f/f

Rac1d/d

0

1

2

Rela

tiv

e G

en

e E

xp

ress

ion

(2-

Ct )

Vegfa

Rac1f/f

Rac1d/d

0

1

2

Rela

tiv

e G

en

e E

xp

ress

ion

(2-

Ct )

Page 110: © 2013 Juanmahel Davila - IDEALS

95

FIGURE 4.4: SUPPRESSION OF THE RAC1 PATHWAY COMPROMISES THE FORMATION OF THE DECIDUAL VASCULAR PLEXUS. (A and B) The

extent of the angiogenic defect was evaluated using PECAM1/CD31, which is a general marker of endothelial cells and is labeled by the red staining.

Our results show a dramatic reduction on blood vessel density in Rac1d/d uteri on days of pregnancy 8 (panel A) and 10 (panel B). A diagrammatic

representation of the vascular network of the implantation nodules is shown to aid in the visualization (adapted and modified with permission form

[155]). E = embryo, AM = anti-mesometrial decidua, M = mesometrial decidua, LS = lateral blood sinusoids, BZ = basal zone, CS = circular smooth

muscle zone, CZ = compact zone, DC = decidua capsularis, P = Placenta, L = luminal epithelium, LDZ = lateral decidual zone, MY = myometrium,

MG = metrial gland, MT = mesometrial triangle.

Page 111: © 2013 Juanmahel Davila - IDEALS

96

FIGURE 4.4: SUPPRESSION OF THE RAC1 PATHWAY COMPROMISES THE FORMATION OF THE DECIDUAL VASCULAR PLEXUS (CONTINUED). (A

and B) The extent of the angiogenic defect was evaluated using PECAM1/CD31, which is a general marker of endothelial cells and is labeled by

the red staining. Our results show a dramatic reduction on blood vessel density in Rac1d/d uteri on days of pregnancy 8 (panel A) and 10 (panel B).

A diagrammatic representation of the vascular network of the implantation nodules is shown to aid in the visualization (adapted and modified with

permission form [155]). E = embryo, AM = anti-mesometrial decidua, M = mesometrial decidua, LS = lateral blood sinusoids, BZ = basal zone, CS

= circular smooth muscle zone, CZ = compact zone, DC = decidua capsularis, P = Placenta, L = luminal epithelium, LDZ = lateral decidual zone,

MY = myometrium, MG = metrial gland, MT = mesometrial triangle. n ≥ 3.

Page 112: © 2013 Juanmahel Davila - IDEALS

97

FIGURE 4.5: CONDITIONAL DELETION OF UTERINE RAC1 IMPAIRS ANGIOGENESIS. Expression of neuropilins in the uterus has been implicated in

the regulation of angiogenesis. Here, we show that neuropilin 1 (NRP1) expression is dramatically reduced in Rac1d/d decidua on day 8 of pregnancy.

PECAM1, a marker for endothelial cells, is used to highlight the vascular tree. Lower left inset on the panel shows no primary antibody control. In

panel B, high magnification images of the vascular plexus (boxed areas 1 and 2) are shown to the right of each image. E = embryo, AM = anti-

mesometrial decidua, M = mesometrial decidua, LS = lateral blood sinuses (also known as vascular web). Bars = 500 µm. n ≥ 3.

Page 113: © 2013 Juanmahel Davila - IDEALS

98

FIGURE 4.6: CONDITIONAL DELETION OF RAC1 IMPAIRS VEGFA TRANSPORT AND SECRETION OF UTERINE ENDOMETRIAL CELLS. VEGFA

immunocytochemistry done in decidual cells that were cultured for 72 h in differentiation media are shown here. In all instances, we stained 72 h

cultured cells, VEGF accumulated in the perinuclear region of the cell, whereas in the control cells VEGF vesicles were either uniformly distributed

in the cytoplasm or the number was reduced, presumably, due to secretion. Gene expression analysis of the differentiation marker Prl8a2/dPrp, as

well as Vegfa, shows no change in expression levels between the experimental and control groups. n = 3. Asterisks indicate significant differences

from controls (*** p < 0.001).

Page 114: © 2013 Juanmahel Davila - IDEALS

99

FIGURE 4.7: RAC1D/D UTERI DISPLAY EXTRAVASATION OF RED BLOOD CELLS. Extravasation and vascular leakage was confirmed in Rac1d/d mice

by staining uterine sections on day 8 of pregnancy with eosin-Y containing phloxine. Here, we show that despite a severe reduction in blood

vasculature in the conditional null uteri, blood hemorrhage, and blood vessel leakiness is observed (inset box). Dotted lines demarcate the blood

sinuses. Upper left inset on panel shows high magnification images of the boxed area near the lateral sinuses. E = embryo, AM = anti-mesometrial

decidua, M = mesometrial decidua, LS = lateral blood sinuses (also known as vascular web). Bars = 500 µm. n = 4.

Page 115: © 2013 Juanmahel Davila - IDEALS

100

FIGURE 4.8: DECIDUA FROM RAC1D/D MICE SHOW SEVERE BLOOD HEMORRHAGE IN THE LATERAL BLOOD SINUSES. Representative images of

uterine samples following blood perfusion, as compared to a non-perfused Rac1f/f control uterus. The upper images in the panel show three isolated

implantation nodules, including myometrium. The lower images in the panel show the isolated decidua (rotated 90o from the position observed in

the upper images of the panel). Here, we show that Rac1-null decidua fail to clear the vascular sinuses, as control decidua does, following vascular

perfusion with saline containing heparin. All samples shown here were evaluated on day 8 of pregnancy. n = 3.

.

Page 116: © 2013 Juanmahel Davila - IDEALS

101

CHAPTER 5: CONCLUSIONS AND WORKING

MODEL

Page 117: © 2013 Juanmahel Davila - IDEALS

102

The work presented as part of this dissertation reveals novel roles for RAC1 in the uterus during

pregnancy (Figure 5.1). Our studies have shown that in uteri lacking RAC1, the embryos are able

to attach to the epithelium to initiate implantation. However, a six-month breeding study showed

that female mice with a null mutation of Rac1 in the uterine tissues became severely subfertile,

and towards the end of the study became sterile. Although we have not unraveled the complete

repertoire of the pathways affected by this mutation, we were able to identify certain critical events

in pregnancy that were severely compromised in response to this mutation. For example, we found

that mutation of uterine Rac1 led to dysregulation of genes that are involved in endometrial stromal

cell differentiation. Because decidualization involves stromal cell proliferation followed by

differentiation and RAC1 has been reported previously to regulate proliferation in other systems

[367], it is of interest to determine the impact of RAC1 ablation on proliferation of stromal cells.

Our studies revealed no significant difference in cell proliferation of stromal cells in Rac1f/f and

Rac1d/d uteri. However, a marked upregulation of Alpl and Wnt4 and a downregulation of

Prl3c1/Plpj gene expression were observed in Rac1-ablated uteri, indicating dysregulated

differentiation program in Rac1d/d mice during decidualization. The expression of certain other

genes known to be critical for stromal differentiation such as Bmp2, Gjp1/Cx43, or Vegfa were not

affected by the deletion of Rac1. It is possible that these genes are functioning independent of

RAC1 pathway in the uterus. It is also possible that RAC1 regulates the post-transcriptional events

and is involved in the secretion of some of these factors, including VEGFA.

A consequence of impaired stromal differentiation in Rac1-null uteri is a defect in uterine

angiogenesis and blood vessel stability. Gene expression profiling of the decidual tissues showed

that expression of many genes that are necessary for uterine angiogenesis, such as Hif2α, Anpt2,

Angpt4, and Nrp1, were significantly downregulated in Rac1-ablated uteri. Interestingly, however,

Hif1α, a known target of RAC1 during hypoxic conditions [319-322, 324, 395-399], was not

affected upon uterine deletion of Rac1. It is possible that in decidual uteri, Hif2α rather than Hif1α

is functioning downstream of the RAC1 pathway to regulate angiogenesis. Consistent with these

observations, we noted impaired vascular plexus formation in Rac1d/d uteri.

Immunostaining with PECAM1/CD31 showed a noticeable reduction in the vascular tree in

Rac1d/d uteri. Absence of blood clearance following perfusion of the blood vasculature clearly

indicated unstable and leaky blood vessels in uterine tissues of Rac1d/d females. Interestingly,

Page 118: © 2013 Juanmahel Davila - IDEALS

103

disruption of endothelial cell network, vascular breakage, and hemorrhage during early pregnancy

has been reported previously in female mice deficient for Sphk1 gene. Consistent with this

observation, we noted a marked reduction in the expression of Sphk1 in the Rac1d/d uteri. Because

the suppression of the sphingolipid pathway was sufficient to cause significant alterations in the

blood vasculature in the Sphk1 mutant mice [377], it is possible that similar events could be

occurring in the Rac1d/d uteri. Furthermore, we noted with interest that the expression of F13a1, a

gene involved in clotting and hemorrhage, was reduced in the Rac1d/d uteri, further supporting the

notion of a compromised vascular tree in these mutant animals.

Interestingly, the expression of Vegfa was not altered in the uteri of Rac1d/d females. VEGFA,

acting through VEGF receptors (VEGFR1, VEGFR2, and NRP1) and in conjunction with the

ANGPT system, coordinates many important processes that ultimately lead to vascular remodeling

[178, 184, 390, 400]. Gene expression analysis on day 8 of pregnancy showed a marked reduction

in the expression of Angpt2 and Nrp1 in the uterine tissue. Both ANGPT2 and NRP1 have been

implicated in uterine angiogenesis [30, 57, 72, 178, 195, 198, 400, 401]. The fact that these two

molecules are downregulated in the Rac1d/d uteri further supports the angiogenic defect observed

in these animals. Additionally, VEGF, in the absence of the ANGPT system can lead to abnormal

vascular leakage and blood vessel instability [184]. It should be noted, however, that we made one

critical observation with regard to VEGFA secretion in Rac1-deficient decidual cells. These cells

displayed a marked reduction in VEGFA secretion compared to Rac1-intact stromal cells.

However, besides decidual cells, trophoblasts [57, 62, 69, 186, 402] and uterine natural killer cells

[360, 403, 404] can also contribute to the production and secretion of VEGFA, which can lead to

dysregulated VEGFA signaling. Taken together, we are of the view that severe vascularization

defect observed in the Rac1d/d female mice is the primary cause of embryo loss by mid-gestation.

It has been recognized for a long time that the differentiated decidual cells secrete a variety of

factors that are important for the growth and development of the implanted embryo [23-25, 29, 91,

121-123]. However, the molecular mechanisms by which the decidual cells undergo secretory

activity remain largely unknown. In this study, we show for the first time that primary decidual

cells from Rac1d/d uteri display reduced VEGFA secretion, despite no impairment in VEGFA

synthesis and initial packaging into secretory vesicles. It is possible that decidual cells utilize

RAC1-dependent actin polymerization for vesicle transport. CDC42, like RAC1, can induce actin

Page 119: © 2013 Juanmahel Davila - IDEALS

104

polymerization and has been involved in vesicle secretion. However, CDC42 is known to regulate

the earlier events of vesicle trafficking (i.e. rER to Golgi), whereas RAC1 seems to regulate the

later stages of trafficking (i.e. Golgi to plasma membrane) [310]. Nonetheless, it is clear that a

RAC1-dependent secretory function of decidual cells is important for embryo survival and

maintenance of pregnancy.

Studies aiming to understand the underlying reasons for failures in implantation and placental

development are clinically important. Indeed, about one-third of normal human pregnancies end

in spontaneous abortion, and a fair percentage of these pregnancies will be lost before they are

detected clinically [12-14]. Similarly, failure in embryonic development, due to defects in the

embryo or the maternal environment, account for almost 80% of the embryonic loss that occurs in

farm animal species [405-407]. In humans, abnormalities in the vascular connections result in

preeclampsia and/or IUGR, diseases that account for significant morbidity and mortality to both

the mother and fetus [12, 44, 51, 59, 62]. Therefore, research aimed to understand the physiological

and pathological events that lead to pregnancy complications are required to further our

understanding of this complicated process. Studies described in this thesis suggest that

disturbances in RAC1-dependent pathways may be relevant to humans with recurrent pregnancy

failures.

Page 120: © 2013 Juanmahel Davila - IDEALS

105

Chapter Figure

FIGURE 5.1: WORKING MODEL FOR THE ROLE OF RAC1 IN THE UTERUS DURING EARLY EVENTS OF PREGNANCY. Activation of RAC1 in the

endometrium during early events of pregnancy results in a series of signaling cascades that ultimately: 1) regulate gene expression of factors

responsible for stromal cell differentiation, as well as angiogenesis and vascular stability; 2) induce changes in actin dynamics to promote vesicle

transport from the rough endoplasmic reticulum (rER) to the plasma membrane, as well as the secretion of the protein content to the extracellular

matrix. Finally changes in actin dynamics also promote 3) cell migration to establish an intimate relationship with the embryo. All these events are

necessary to ensure embryo survival and pregnancy maintenance.

Page 121: © 2013 Juanmahel Davila - IDEALS

106

CHAPTER 6: FUTURE DIRECTIONS

Page 122: © 2013 Juanmahel Davila - IDEALS

107

The work described in this dissertation uncovered new roles for RAC1 in uterine tissue and raised

interesting questions related to maternal-fetal interactions during pregnancy. RAC1, a pleotropic

signaling molecule, is known to be involved in various biological processes. We have observed

some of the critical functions displayed by RAC1 in the uterus during early pregnancy.

During the exploration of the Rac1d/d mouse model, we made a few critical observations that might

be of clinical importance. We noted that implanted embryos displayed a dysregulated trophoblast

expansion by day 8 of pregnancy (Appendix C, Figure C3.1). Although we do not know what

drives trophoblast cells to over expand in the uterus of the conditional null mice, it is our hypothesis

that the overexpansion of the trophoblast cells is most likely linked to the inherent impairment of

Rac1-deficent stromal cells. Our preliminary wound-healing assay (data not shown) showed that

Rac1-deficient stromal cells exhibit migratory impairments. Furthermore, our studies have

revealed that the Rac1-null decidual cells have an overall secretion defect. Therefore, it is possible

to assume that the dysregulated trophoblast expansion could be the result of a combinatorial effect

arising from maternal deficiencies in migration and secretion.

In an attempt to gain insights into the potential pathways that might be dysregulated in the decidua,

we performed gene expression analysis of known molecules that have been implicated in

regulating trophoblast migration (Appendix C, Figure C3.2). Whereas IGF2 is secreted from the

blastocyst to promote trophoblast migration, IGFBPs, secreted from the decidual cells, are thought

to interfere with this process. Our preliminary data show an upregulated expression of Igbp4, while

gene expression of adrenomedullin (Adm) and Igfbp3 is not changed. Both Igfbp4 and Adm have

been suggested to regulate trophoblast migration. In particular, high expression of IGBP4 has been

found in women with recurrent miscarriage. Because decidual cells deficient for RAC1 exhibit a

defect in secretory activity, it is possible that certain factors critical for proper functioning of the

mother and the fetus are absent in Rac1-null uteri. Thus, it would be important to determine if

other factors besides VEGFA exhibit impaired secretory activity. It is clear that the secretory

functions of the decidual cells are crucial to maintain the embryo under control in the proper

cellular compartment. Any perturbation in this process and a dysregulation in the migratory

potential of the trophoblast cells can have serious consequences to the outcome of pregnancy.

Page 123: © 2013 Juanmahel Davila - IDEALS

108

Thus, elucidating how decidual cells precisely regulate trophoblast invasion has great clinical

significance.

Intrauterine fetal growth restriction (IUGR) is another area of great clinical importance. It is now

widely accepted that the exposure of developing embryo to the environment can have severe

consequences on the metabolic phenotype of an organism without alterations in the genomic

sequence. Epigenetic modifications caused by the nutritional environment of the developing

embryo have gained great interest recently because of the obesity epidemic that is affecting the

USA. Interestingly, our preliminary observations of implantation nodules obtained on day 15 of

pregnancy, suggest that the Rac1-deficient uterine environment may not be fully conducive to

proper embryonic growth. When implantation nodules were dissected, we noticed growth

retardation in the embryos we were able to collect (Appendix C, Figure C3.3B). Although we have

a small sample size, we did notice a strong trend towards IUGR, as assessed by the weight of the

embryos from Rac1f/f and Rac1d/d mice. Additionally, the placentas obtained from these growth-

retarded embryos displayed abnormal shapes, and were significantly heavier than the control

placenta (Appendix C, Figures C3.3B to C3.3D). Consistent with the phenotype associated with

IUGR, implantation nodules collected on day 12 of pregnancy displayed abnormalities in blood

vasculature. Further, uterine samples from Rac1d/d mice showed intermediate to no modifications

of spiral artery, which is detrimental to maintenance of pregnancy (Appendix C, Figure C3.4).

Further studies should establish whether the lack of spiral artery modification combined with the

vascular defect is driving embryonic loss at mid-gestation in the Rac1d/d mouse. We anticipate that

further studies in each of these areas will provide an in depth knowledge of function of RAC1 in

the uterus at distinct stages of early pregnancy. Ultimately this knowledge will be useful in

developing treatment options for women with pathologies associated with pregnancy, including

recurrent miscarriage and IUGR.

Page 124: © 2013 Juanmahel Davila - IDEALS

109

REFERENCES

Page 125: © 2013 Juanmahel Davila - IDEALS

110

1. Dey SK, Lim H. Implantation. In: Jimmy DN, Ph.D, Tony MP, Ph.D, Donald WP, Ph.D,

John RGC, D.Sc, F.R.S.C, David MdK, M.D, A.O, et al. (eds.), Knobil and Neill's

Physiology of Reproduction (Third Edition). St Louis: Academic Press; 2006: 147-188.

2. Lee KY, DeMayo FJ. Animal models of implantation. Reproduction 2004; 128:679-695.

3. Makrigiannakis A, Minas V. Mechanisms of implantation. Reprod Biomed Online 2007;

14:102-109.

4. Rogers PA. Current studies on human implantation: a brief overview. Reprod Fertil Dev

1995; 7:1395-1399.

5. Cross JC, Werb Z, Fisher SJ. Implantation and the placenta: key pieces of the development

puzzle. Science 1994; 266:1508-1518.

6. Edwards RG. Human implantation: the last barrier in assisted reproduction technologies?

Reprod Biomed Online 2007; 14 Spec No 1:5-22.

7. Grewal S, Carver J, Ridley AJ, Mardon HJ. Human endometrial stromal cell rho GTPases

have opposing roles in regulating focal adhesion turnover and embryo invasion in vitro.

Biol Reprod 2010; 83:75-82.

8. Paria BC, Reese J, Das SK, Dey SK. Deciphering the cross-talk of implantation: advances

and challenges. Science 2002; 296:2185-2188.

9. Cross JC, Hemberger M, Lu Y, Nozaki T, Whiteley K, Masutani M, Adamson SL.

Trophoblast functions, angiogenesis and remodeling of the maternal vasculature in the

placenta. Mol Cell Endocrinol 2002; 187:207-212.

10. Carson DD, Bagchi I, Dey SK, Enders AC, Fazleabas AT, Lessey BA, Yoshinaga K.

Embryo implantation. Developmental Biology 2000; 223:217-237.

11. Psychoyos A. Endocrine control of egg implantation. In: Astwood ROGEG (ed.)

Handbook of Physiology. Washington, DC: American Physiological Society; 1973: 187-

215.

12. Bricker L, Farquharson RG. Types of pregnancy loss in recurrent miscarriage: implications

for research and clinical practice. Human Reproduction 2002; 17:1345-1350.

13. Wilcox AJ, Weinberg CR, O'Connor JF, Baird DD, Schlatterer JP, Canfield RE, Armstrong

EG, Nisula BC. Incidence of early loss of pregnancy. N Engl J Med 1988; 319:189-194.

14. Wang H, Dey SK. Roadmap to embryo implantation: clues from mouse models. Nat Rev

Genet 2006; 7:185-199.

15. Evers JL. Female subfertility. Lancet 2002; 360:151-159.

16. Garcia-Enguidanos A, Calle ME, Valero J, Luna S, Dominguez-Rojas V. Risk factors in

miscarriage: a review. European Journal of Obstetrics Gynecology and Reproductive

Biology 2002; 102:111-119.

17. Cousineau TM, Domar AD. Psychological impact of infertility. Best Practice & Research

in Clinical Obstetrics & Gynaecology 2007; 21:293-308.

18. Lykeridou K, Gourounti K, Deltsidou A, Loutradis D, Vaslamatzis G. The impact of

infertility diagnosis on psychological status of women undergoing fertility treatment.

Journal of Reproductive and Infant Psychology 2009; 27:223-237.

19. Boivin J, Appleton TC, Baetens P, Baron J, Bitzer J, Corrigan E, Daniels KR, Darwish J,

Guerra-Diaz D, Hammar M, McWhinnie A, Strauss B, et al. Guidelines for counselling in

infertility: outline version. Human Reproduction 2001; 16:1301-1304.

20. Read J. ABC of sexual health: sexual problems associated with infertility, pregnancy, and

ageing. BMJ 1999; 318:587-589.

Page 126: © 2013 Juanmahel Davila - IDEALS

111

21. Mascarenhas MN, Flaxman SR, Boerma T, Vanderpoel S, Stevens GA. National, Regional,

and Global Trends in Infertility Prevalence Since 1990: A Systematic Analysis of 277

Health Surveys. Plos Medicine 2012; 9.

22. Gu Y, Soares MJ, Srivastava RK, Gibori G. Expression of decidual prolactin-related

protein in the rat decidua. Endocrinology 1994; 135:1422-1427.

23. Irwin JC, Giudice LC. Decidua. In: Knobil E, Neill JD (eds.), Encyclopedia of

Reproduction. San Diego: Academic Press; 1998: 822-835.

24. Kennedy TG. Decidualization. In: Editors-in-Chief: Helen LH, Anthony WN (eds.),

Encyclopedia of Hormones. New York: Academic Press; 2003: 379-385.

25. Ramathal CY, Bagchi IC, Taylor RN, Bagchi MK. Endometrial decidualization: of mice

and men. Semin Reprod Med 2010; 28:17-26.

26. Grewal S, Carver JG, Ridley AJ, Mardon HJ. Implantation of the human embryo requires

Rac1-dependent endometrial stromal cell migration. Proc Natl Acad Sci U S A 2008;

105:16189-16194.

27. Rinkenberger JL, Cross JC, Werb Z. Molecular genetics of implantation in the mouse.

Developmental genetics 1997; 21:6-20.

28. Watson ED, Cross JC. Development of structures and transport functions in the mouse

placenta. Physiology (Bethesda) 2005; 20:180-193.

29. Abrahamsohn PA, Zorn TM. Implantation and decidualization in rodents. J Exp Zool 1993;

266:603-628.

30. Bany BM, Cross JC. Post-implantation mouse conceptuses produce paracrine signals that

regulate the uterine endometrium undergoing decidualization. Dev Biol 2006; 294:445-

456.

31. Cross JC. How to make a placenta: mechanisms of trophoblast cell differentiation in mice-

-a review. Placenta 2005; 26 Suppl A:S3-9.

32. Laws MJ, Taylor RN, Sidell N, DeMayo FJ, Lydon JP, Gutstein DE, Bagchi MK, Bagchi

IC. Gap junction communication between uterine stromal cells plays a critical role in

pregnancy-associated neovascularization and embryo survival. Development 2008;

135:2659-2668.

33. Gellersen B, Brosens IA, Brosens JJ. Decidualization of the human endometrium:

mechanisms, functions, and clinical perspectives. Semin Reprod Med 2007; 25:445-453.

34. Skakkebaek NE, Jorgensen N, Main KM, Rajpert-De Meyts E, Leffers H, Andersson AM,

Juul A, Carlsen E, Mortensen GK, Jensen TK, Toppari J. Is human fecundity declining?

International Journal of Andrology 2006; 29:2-11.

35. Velde ET, Burdorf A, Nieschlag E, Eijkemans R, Kremer JAM, Roeleveld N, Habbema D.

Is human fecundity declining in Western countries? Human Reproduction 2010; 25:1348-

1353.

36. Lutz W, O'Neill BC, Scherbov S. Demographics: Europe's population at a turning point.

Science 2003; 299:1991-1992.

37. 2010 Assisted Reproductive Technology National Summary Report. In: Center for Disease

Control and Prevention ASfRM, and Society for Assisted Reproductive Technology (ed.).

Atlanta, GA: US Department of Health and Human Services; 2010.

38. Mardon HJ, Bagchi MK, Bagchi IC, Peng C, Karpovich N, Wang Y. Hormonal and

paracrine regulation of embryonic implantation: a workshop report. Placenta 2007; 28

Suppl A:S82-84.

Page 127: © 2013 Juanmahel Davila - IDEALS

112

39. Assisted reproductive technology in the United States: 1996 results generated from the

American Society for Reproductive Medicine/Society for Assisted Reproductive

Technology Registry. Fertil Steril 1999; 71:798-807.

40. Ford HB, Schust DJ. Recurrent pregnancy loss: etiology, diagnosis, and therapy. Rev

Obstet Gynecol 2009; 2:76-83.

41. Dey SK, Lim H, Das SK, Reese J, Paria BC, Daikoku T, Wang H. Molecular cues to

implantation. Endocr Rev 2004; 25:341-373.

42. Nybo Andersen AM, Wohlfahrt J, Christens P, Olsen J, Melbye M. Maternal age and fetal

loss: population based register linkage study. BMJ 2000; 320:1708-1712.

43. Knudsen UB, Hansen V, Juul S, Secher NJ. Prognosis of a new pregnancy following

previous spontaneous abortions. Eur J Obstet Gynecol Reprod Biol 1991; 39:31-36.

44. Rai R, Regan L. Recurrent miscarriage. Lancet 2006; 368:601-611.

45. Practice Committee of the American Society for Reproductive M. Evaluation and treatment

of recurrent pregnancy loss: a committee opinion. Fertil Steril 2012; 98:1103-1111.

46. Volgsten H, Skoog Svanberg A, Ekselius L, Lundkvist O, Sundstrom Poromaa I.

Prevalence of psychiatric disorders in infertile women and men undergoing in vitro

fertilization treatment. Hum Reprod 2008; 23:2056-2063.

47. Boivin J, Griffiths E, Venetis CA. Emotional distress in infertile women and failure of

assisted reproductive technologies: meta-analysis of prospective psychosocial studies.

BMJ 2011; 342:d223.

48. Grimbizis GF, Camus M, Tarlatzis BC, Bontis JN, Devroey P. Clinical implications of

uterine malformations and hysteroscopic treatment results. Hum Reprod Update 2001;

7:161-174.

49. Woelfer B, Salim R, Banerjee S, Elson J, Regan L, Jurkovic D. Reproductive outcomes in

women with congenital uterine anomalies detected by three-dimensional ultrasound

screening. Obstetrics and gynecology 2001; 98:1099-1103.

50. Salim R, Regan L, Woelfer B, Backos M, Jurkovic D. A comparative study of the

morphology of congenital uterine anomalies in women with and without a history of

recurrent first trimester miscarriage. Human Reproduction 2003; 18:162-166.

51. Branch DW, Heuser C. Recurrent Miscarriage. Reproductive Endocrinology and

Infertility: Integrating Modern Clinical and Laboratory Practice 2010:281-296.

52. Jauniaux E, Burton GJ. Pathophysiology of histological changes in early pregnancy loss.

Placenta 2005; 26:114-123.

53. Jauniaux E, Poston L, Burton GJ. Placental-related diseases of pregnancy: Involvement of

oxidative stress and implications in human evolution. Hum Reprod Update 2006; 12:747-

755.

54. Morikawa M, Yamada H, Kato EH, Shimada S, Yamada T, Minakami H. Embryo loss

pattern is predominant in miscarriages with normal chromosome karyotype among women

with repeated miscarriage. Hum Reprod 2004; 19:2644-2647.

55. Roberts JM, Redman CWG. Preeclampsia - More Than Pregnancy-Induced Hypertension.

Lancet 1993; 341:1447-1451.

56. Vailhe B, Dietl J, Kapp M, Toth B, Arck P. Increased blood vessel density in decidua

parietalis is associated with spontaneous human first trimester abortion. Human

Reproduction 1999; 14:1628-1634.

57. Zygmunt M, Herr F, Munstedt K, Lang U, Liang OD. Angiogenesis and vasculogenesis in

pregnancy. Eur J Obstet Gynecol Reprod Biol 2003; 110 Suppl 1:S10-18.

Page 128: © 2013 Juanmahel Davila - IDEALS

113

58. Plaisier M, Dennert I, Rost E, Koolwijk P, van Hinsbergh VW, Helmerhorst FM. Decidual

vascularization and the expression of angiogenic growth factors and proteases in first

trimester spontaneous abortions. Hum Reprod 2009; 24:185-197.

59. Burton GJ, Jauniaux E. Placental oxidative stress: from miscarriage to preeclampsia. J Soc

Gynecol Investig 2004; 11:342-352.

60. Greenwold N, Jauniaux E, Gulbis B, Hempstock J, Gervy C, Burton GJ. Relationship

among maternal serum endocrinology, placental karyotype, and intervillous circulation in

early pregnancy failure. Fertility and sterility 2003; 79:1373-1379.

61. Kingdom JC, Kaufmann P. Oxygen and placental villous development: origins of fetal

hypoxia. Placenta 1997; 18:613-621; discussion 623-616.

62. Vuorela P, Carpen O, Tulppala M, Halmesmaki E. VEGF, its receptors and the tie receptors

in recurrent miscarriage. Mol Hum Reprod 2000; 6:276-282.

63. Plaisier M, Rodrigues S, Willems F, Koolwijk P, van Hinsbergh VWM, Helmerhorst FM.

Different degrees of vascularization and their relationship to the expression of vascular

endothelial growth factor, placental growth factor, angiopoietins, and their receptors in

first-trimester decidual tissues. Fertility and sterility 2007; 88:176-187.

64. Geva E, Ginzinger DG, Zaloudek CJ, Moore DH, Byrne A, Jaffe RB. Human placental

vascular development: Vasculogenic and angiogenic (branching and nonbranching)

transformation is regulated by vascular endothelial growth factor-A, angiopoietin-1, and

angiopoietin-2. Journal of Clinical Endocrinology & Metabolism 2002; 87:4213-4224.

65. Qiu Q, Bell M, Lu X, Yan X, Rodger M, Walker M, Wen SW, Bainbridge S, Wang H,

Gruslin A. Significance of IGFBP-4 in the development of fetal growth restriction. J Clin

Endocrinol Metab 2012; 97:E1429-1439.

66. Watson CS, Bialek P, Anzo M, Khosravi J, Yee SP, Han VK. Elevated circulating insulin-

like growth factor binding protein-1 is sufficient to cause fetal growth restriction.

Endocrinology 2006; 147:1175-1186.

67. Krebs C, Macara LM, Leiser R, Bowman AW, Greer IA, Kingdom JCP. Intrauterine

growth restriction with absent end-diastolic flow velocity in the umbilical artery is

associated with maldevelopment of the placental terminal villous tree. American Journal

of Obstetrics and Gynecology 1996; 175:1534-1542.

68. Jauniaux E, Greenwold N, Hempstock J, Burton GJ. Comparison of ultrasonographic and

Doppler mapping of the intervillous circulation in normal and abnormal early pregnancies.

Fertility and sterility 2003; 79:100-106.

69. Ahmed A, Dunk C, Ahmad S, Khaliq A. Regulation of placental vascular endothelial

growth factor (VEGF) and placenta growth factor (PIGF) and soluble Flt-1 by oxygen--a

review. Placenta 2000; 21 Suppl A:S16-24.

70. Plaisier M. Decidualisation and angiogenesis. Best Pract Res Clin Obstet Gynaecol 2011;

25:259-271.

71. Khaliq A, Dunk C, Jiang J, Shams M, Li XF, Acevedo C, Weich H, Whittle M, Ahmed A.

Hypoxia down-regulates placenta growth factor, whereas fetal growth restriction up-

regulates placenta growth factor expression: molecular evidence for "placental hyperoxia"

in intrauterine growth restriction. Lab Invest 1999; 79:151-170.

72. Ahmed A, Perkins J. Angiogenesis and intrauterine growth restriction. Baillieres Best Pract

Res Clin Obstet Gynaecol 2000; 14:981-998.

Page 129: © 2013 Juanmahel Davila - IDEALS

114

73. Walter LM, Rogers PA, Girling JE. Differential expression of vascular endothelial growth

factor-A isoforms in the mouse uterus during early pregnancy. Reprod Biomed Online

2010; 21:803-811.

74. Kashiwagi A, DiGirolamo CM, Kanda Y, Niikura Y, Esmon CT, Hansen TR, Shioda T,

Pru JK. The postimplantation embryo differentially regulates endometrial gene expression

and decidualization. Endocrinology 2007; 148:4173-4184.

75. Bany BM, Cross JC. Post-implantation mouse conceptuses produce paracrine signals that

regulate the uterine endometrium undergoing decidualization. Developmental Biology

2006; 294:445-456.

76. Herington JL, Bany BM. Do molecular signals from the conceptus influence endometrium

decidualization in rodents? J Exp Zool B Mol Dev Evol 2009; 312:797-816.

77. Torry DS, Leavenworth J, Chang M, Maheshwari V, Groesch K, Ball ER, Torry RJ.

Angiogenesis in implantation. J Assist Reprod Genet 2007; 24:303-315.

78. Aplin JD. Implantation, trophoblast differentiation and haemochorial placentation:

mechanistic evidence in vivo and in vitro. J Cell Sci 1991; 99 ( Pt 4):681-692.

79. Fisher SJ, Damsky CH. Human cytotrophoblast invasion. Semin Cell Biol 1993; 4:183-

188.

80. Bany BM, Schultz GA. Tissue inhibitor of matrix metalloproteinase-3 expression in the

mouse uterus during implantation and artificially induced decidualization. Mol Reprod

Dev 2001; 59:159-167.

81. Higuchi T, Kanzaki H, Nakayama H, Fujimoto M, Hatayama H, Kojima K, Iwai M, Mori

T, Fujita J. Induction of tissue inhibitor of metalloproteinase 3 gene expression during in

vitro decidualization of human endometrial stromal cells. Endocrinology 1995; 136:4973-

4981.

82. Giudice LC. Multifaceted roles for IGFBP-1 in human endometrium during implantation

and pregnancy. Ann N Y Acad Sci 1997; 828:146-156.

83. Giudice LC, Mark SP, Irwin JC. Paracrine actions of insulin-like growth factors and IGF

binding protein-1 in non-pregnant human endometrium and at the decidual-trophoblast

interface. J Reprod Immunol 1998; 39:133-148.

84. Irwin JC, Giudice LC. Insulin-like growth factor binding protein-1 binds to placental

cytotrophoblast alpha5beta1 integrin and inhibits cytotrophoblast invasion into

decidualized endometrial stromal cultures. Growth Horm IGF Res 1998; 8:21-31.

85. Irwin JC, Suen LF, Martina NA, Mark SP, Giudice LC. Role of the IGF system in

trophoblast invasion and pre-eclampsia. Hum Reprod 1999; 14 Suppl 2:90-96.

86. Giudice LC, Conover CA, Bale L, Faessen GH, Ilg K, Sun I, Imani B, Suen LF, Irwin JC,

Christiansen M, Overgaard MT, Oxvig C. Identification and regulation of the IGFBP-4

protease and its physiological inhibitor in human trophoblasts and endometrial stroma:

evidence for paracrine regulation of IGF-II bioavailability in the placental bed during

human implantation. J Clin Endocrinol Metab 2002; 87:2359-2366.

87. Ganeff C, Chatel G, Munaut C, Frankenne F, Foidart JM, Winkler R. The IGF system in

in-vitro human decidualization. Mol Hum Reprod 2009; 15:27-38.

88. Zygmunt M Fau - Mazzuca DM, Mazzuca Dm Fau - Walton J, Walton J Fau - Han VK,

Han VK. Local fetal signal is not required for maintaining IGFBP gene expression in the

human decidua: evidence from extrauterine pregnancies. 2000.

Page 130: © 2013 Juanmahel Davila - IDEALS

115

89. Ning Y, Schuller AG, Conover CA, Pintar JE. Insulin-like growth factor (IGF) binding

protein-4 is both a positive and negative regulator of IGF activity in vivo. Mol Endocrinol

2008; 22:1213-1225.

90. Carter AM, Nygard K, Mazzuca DM, Han VK. The expression of insulin-like growth factor

and insulin-like growth factor binding protein mRNAs in mouse placenta. Placenta 2006;

27:278-290.

91. Henemyre C, Markoff E. Decidualization and expression of insulin-like growth factor-I

and insulin-like growth factor binding protein-4 in the periimplantation mouse uterus. Biol

Reprod 1998; 58:801-806.

92. Markoff E, Henemyre C, Fellows J, Pennington E, Zeitler PS, Cedars MI. Localization of

insulin-like growth factor binding protein-4 expression in the mouse uterus during the peri-

implantation period. Biol Reprod 1995; 53:1103-1109.

93. Irwin JC, Suen LF, Faessen GH, Popovici RM, Giudice LC. Insulin-like growth factor

(IGF)-II inhibition of endometrial stromal cell tissue inhibitor of metalloproteinase-3 and

IGF-binding protein-1 suggests paracrine interactions at the decidua:trophoblast interface

during human implantation. J Clin Endocrinol Metab 2001; 86:2060-2064.

94. Crossey PA, Pillai CC, Miell JP. Altered placental development and intrauterine growth

restriction in IGF binding protein-1 transgenic mice. J Clin Invest 2002; 110:411-418.

95. Nakamura O, Hondo E, Namba Y, Kiso Y. IGF-I Overexpression causes fetal loss during

placentation in mice. J Reprod Dev 2004; 50:375-380.

96. Giudice LC, Irwin JC, Dsupin BA, Pannier EM, Jin IH, Vu TH, Hoffman AR. Insulin-like

growth factor (IGF), IGF binding protein (IGFBP), and IGF receptor gene expression and

IGFBP synthesis in human uterine leiomyomata. Hum Reprod 1993; 8:1796-1806.

97. Giudice LC, Lamson G, Rosenfeld RG, Irwin JC. Insulin-like growth factor-II (IGF-II) and

IGF binding proteins in human endometrium. Ann N Y Acad Sci 1991; 626:295-307.

98. Schneider MR, Lahm H, Wu M, Hoeflich A, Wolf E. Transgenic mouse models for

studying the functions of insulin-like growth factor-binding proteins. FASEB J 2000;

14:629-640.

99. Coan PM, Fowden AL, Constancia M, Ferguson-Smith AC, Burton GJ, Sibley CP.

Disproportional effects of Igf2 knockout on placental morphology and diffusional

exchange characteristics in the mouse. J Physiol 2008; 586:5023-5032.

100. Eggenschwiler J, Ludwig T, Fisher P, Leighton PA, Tilghman SM, Efstratiadis A. Mouse

mutant embryos overexpressing IGF-II exhibit phenotypic features of the Beckwith-

Wiedemann and Simpson-Golabi-Behmel syndromes. Genes Dev 1997; 11:3128-3142.

101. Conover CA, Bale LK, Overgaard MT, Johnstone EW, Laursen UH, Fuchtbauer EM,

Oxvig C, van Deursen J. Metalloproteinase pregnancy-associated plasma protein A is a

critical growth regulatory factor during fetal development. Development 2004; 131:1187-

1194.

102. Practice Committee of Society for Assisted Reproductive T, Practice Committee of

American Society for Reproductive M. Preimplantation genetic testing: a Practice

Committee opinion. Fertil Steril 2008; 90:S136-143.

103. Conneely OM, Mulac-Jericevic B, DeMayo F, Lydon JP, O'Malley BW. Reproductive

functions of progesterone receptors. Recent Prog Horm Res 2002; 57:339-355.

104. Huet YM, Dey SK. Role of early and late oestrogenic effects on implantation in the mouse.

J Reprod Fertil 1987; 81:453-458.

105. Psychoyos A (ed.). Nidation window. New York: Springer; 1995.

Page 131: © 2013 Juanmahel Davila - IDEALS

116

106. Yoshinaga K, Adams CE. Delayed implantation in the spayed, progesterone treated adult

mouse. J Reprod Fertil 1966; 12:593-595.

107. Finn CA, Martin L. The control of implantation. J Reprod Fertil 1974; 39:195-206.

108. Yoshinaga K. Uterine receptivity for blastocyst implantation. Ann N Y Acad Sci 1988;

541:424-431.

109. Carson DD, Bagchi I, Dey SK, Enders AC, Fazleabas AT, Lessey BA, Yoshinaga K.

Embryo implantation. Dev Biol 2000; 223:217-237.

110. Lee KY, Jeong JW, Tsai SY, Lydon JP, DeMayo FJ. Mouse models of implantation.

Trends Endocrinol Metab 2007; 18:234-239.

111. Sapin V, Blanchon L, Serre AF, Lemery D, Dastugue B, Ward SJ. Use of transgenic mice

model for understanding the placentation: towards clinical applications in human

obstetrical pathologies? Transgenic Res 2001; 10:377-398.

112. Adamson SL, Lu Y, Whiteley KJ, Holmyard D, Hemberger M, Pfarrer C, Cross JC.

Interactions between trophoblast cells and the maternal and fetal circulation in the mouse

placenta. Developmental Biology 2002; 250:358-373.

113. D'Amico G, Jones DT, Nye E, Sapienza K, Ramjuan AR, Reynolds LE, Robinson SD,

Kostourou V, Martinez D, Aubyn D, Grose R, Thomas GJ, et al. Regulation of lymphatic-

blood vessel separation by endothelial Rac1. Development 2009; 136:4043-4053.

114. Bagchi IC, Li Q, Cheon YP. Role of steroid hormone-regulated genes in implantation. Ann

N Y Acad Sci 2001; 943:68-76.

115. Psychoyos A. Uterine receptivity for nidation. Ann N Y Acad Sci 1986; 476:36-42.

116. Wooding P, Burton G. Implantation, Maternofetal Exchange and Vascular Relationships.

In: Comparative Placentation: Structures, Functions and Evolution. Berlin: Springer; 2008:

47-82.

117. Finn CA. The Implantation Reaction. In: Wynn RM (ed.) Biology of the Uterus. New York:

Plenum Press; 1977: 246-308.

118. Bowen JA, Hunt JS. The Role of Integrins in Reproduction. Proceedings of the Society for

Experimental Biology and Medicine 2008; 223:331-343.

119. Paria BC, Huet-Hudson YM, Dey SK. Blastocyst's state of activity determines the

"window" of implantation in the receptive mouse uterus. Proc Natl Acad Sci U S A 1993;

90:10159-10162.

120. Finn CA. The initiation of the decidual cell reaction in the uterus of the aged mouse. J

Reprod Fertil 1966; 11:423-428.

121. Finn CA. The biology of decidual cells. Adv Reprod Physiol 1971; 5:1-26.

122. Giudice LC, Irwin JC. Roles of the insulinlike growth factor family in nonpregnant human

endometrium and at the decidual: trophoblast interface. Semin Reprod Endocrinol 1999;

17:13-21.

123. Irwin JC, de las Fuentes L, Giudice LC. Growth factors and decidualization in vitro. Ann

N Y Acad Sci 1994; 734:7-18.

124. Cross JC, Baczyk D, Dobric N, Hemberger M, Hughes M, Simmons DG, Yamamoto H,

Kingdom JCP. Genes, Development and Evolution of the Placenta. Placenta 2003; 24:123-

130.

125. Wang J, Mayernik L, Armant DR. Trophoblast adhesion of the peri-implantation mouse

blastocyst is regulated by integrin signaling that targets phospholipase C. Dev Biol 2007;

302:143-153.

Page 132: © 2013 Juanmahel Davila - IDEALS

117

126. Wooding P, Burton G. Haemochorial Placentation: Mouse, Rabbit, Man, Apes, Monkeys.

In: Comparative Placentation: Structures, Functions and Evolution. Berlin: Springer; 2008:

185-230.

127. Wooding P, Burton G. Placentation Fundamentals. In: Comparative Placentation:

Structures, Functions and Evolution. Berlin: Springer; 2008: 1-45.

128. Rinkenberger J, Werb Z. The labyrinthine placenta. Nat Genet 2000; 25:248-250.

129. Goffin F, Munaut C, Malassine A, Evain-Brion D, Frankenne F, Fridman V, Dubois M,

Uzan S, Merviel P, Foidart JM. Evidence of a limited contribution of feto-maternal

interactions to trophoblast differentiation along the invasive pathway. Tissue Antigens

2003; 62:104-116.

130. Hess AP, Hamilton AE, Talbi S, Dosiou C, Nyegaard M, Nayak N, Genbecev-Krtolica O,

Mavrogianis P, Ferrer K, Kruessel J, Fazleabas AT, Fisher SJ, et al. Decidual stromal cell

response to paracrine signals from the trophoblast: amplification of immune and

angiogenic modulators. Biol Reprod 2007; 76:102-117.

131. Red-Horse K, Zhou Y, Genbacev O, Prakobphol A, Foulk R, McMaster M, Fisher SJ.

Trophoblast differentiation during embryo implantation and formation of the maternal-

fetal interface. J Clin Invest 2004; 114:744-754.

132. Rossant J, Cross JC. Placental development: lessons from mouse mutants. Nat Rev Genet

2001; 2:538-548.

133. Zhang S, Lin H, Kong S, Wang S, Wang H, Wang H, Armant DR. Physiological and

molecular determinants of embryo implantation. Mol Aspects Med 2013.

134. Giudice LC. IGF binding protein-3 protease regulation: how sweet it is! J Clin Endocrinol

Metab 1995; 80:2279-2281.

135. Waterhouse P, Denhardt DT, Khokha R. Temporal expression of tissue inhibitors of

metalloproteinases in mouse reproductive tissues during gestation. Mol Reprod Dev 1993;

35:219-226.

136. Hampton AL, Salamonsen LA. Expression of messenger ribonucleic acid encoding matrix

metalloproteinases and their tissue inhibitors is related to menstruation. J Endocrinol 1994;

141:R1-3.

137. Bischof P, Meisser A, Campana A. Biochemistry and molecular biology of trophoblast

invasion. Ann N Y Acad Sci 2001; 943:157-162.

138. Giudice LC. Elucidating endometrial function in the post-genomic era. Hum Reprod

Update 2003; 9:223-235.

139. Cox B, Sharma P, Evangelou AI, Whiteley K, Ignatchenko V, Ignatchenko A, Baczyk D,

Czikk M, Kingdom J, Rossant J, Gramolini AO, Adamson SL, et al. Translational analysis

of mouse and human placental protein and mRNA reveals distinct molecular pathologies

in human preeclampsia. Mol Cell Proteomics 2011; 10:M111 012526.

140. Fisher SJ. The placental problem: linking abnormal cytotrophoblast differentiation to the

maternal symptoms of preeclampsia. Reprod Biol Endocrinol 2004; 2:53.

141. Sibai BM. Diagnosis and management of gestational hypertension and preeclampsia.

Obstetrics and gynecology 2003; 102:181-192.

142. Redman CW, Sargent IL. Latest advances in understanding preeclampsia. Science 2005;

308:1592-1594.

143. Croy BA, Ashkar AA, Minhas K, Greenwood JD. Can murine uterine natural killer cells

give insights into the pathogenesis of preeclampsia? J Soc Gynecol Investig 2000; 7:12-

20.

Page 133: © 2013 Juanmahel Davila - IDEALS

118

144. Tantbirojn P, Crum CP, Parast MM. Pathophysiology of placenta creta: the role of decidua

and extravillous trophoblast. Placenta 2008; 29:639-645.

145. Earl U, Bulmer JN, Briones A. Placenta accreta: an immunohistological study of

trophoblast populations. Placenta 1987; 8:273-282.

146. Hutton L, Yang SS, Bernstein J. Placenta accreta. A 26-year clinicopathologic review

(1956--1981). New York state journal of medicine 1983; 83:857-866.

147. Khong TY. The pathology of placenta accreta, a worldwide epidemic. J Clin Pathol 2008;

61:1243-1246.

148. Khong TY, Robertson WB. Placenta creta and placenta praevia creta. Placenta 1987;

8:399-409.

149. Parr MBELP (ed.). The implantation reaction. New York: Plenum Press; 1989.

150. Gu Y, Gibori G. Deciduoma. In: Knobil E, Neill JD (eds.), Encyclopedia of Reproduction.

San Diego: Academic Press; 1998: 836-842.

151. Dosiou C, Giudice LC. Natural killer cells in pregnancy and recurrent pregnancy loss:

endocrine and immunologic perspectives. Endocr Rev 2005; 26:44-62.

152. Red-Horse K, Drake PM, Gunn MD, Fisher SJ. Chemokine ligand and receptor expression

in the pregnant uterus: reciprocal patterns in complementary cell subsets suggest functional

roles. Am J Pathol 2001; 159:2199-2213.

153. Ashkar AA, Croy BA. Functions of uterine natural killer cells are mediated by interferon

gamma production during murine pregnancy. Semin Immunol 2001; 13:235-241.

154. Finn CA, Hinchliffe JR. Reaction of the Mouse Uterus during Implantation and Deciduoma

Formation as Demonstrated by Changes in the Distribution of Alkaline Phosphatase. J

Reprod Fertil 1964; 8:331-338.

155. Stewart I, Peel S. The differentiation of the decidua and the distribution of metrial gland

cells in the pregnant mouse uterus. Cell Tissue Res 1978; 187:167-179.

156. Orwig KE, Dai G, Rasmussen CA, Soares MJ. Decidual/trophoblast prolactin-related

protein: characterization of gene structure and cell-specific expression. Endocrinology

1997; 138:2491-2500.

157. Rasmussen CA, Orwig KE, Vellucci S, Soares MJ. Dual expression of prolactin-related

protein in decidua and trophoblast tissues during pregnancy in rats. Biol Reprod 1997;

56:647-654.

158. Soares MJ, Muller H, Orwig KE, Peters TJ, Dai G. The uteroplacental prolactin family and

pregnancy. Biol Reprod 1998; 58:273-284.

159. Adastra KL, Frolova AI, Chi MM, Cusumano D, Bade M, Carayannopoulos MO, Moley

KH. Slc2a8 deficiency in mice results in reproductive and growth impairments. Biol

Reprod 2012; 87:49.

160. Frolova A, Flessner L, Chi M, Kim ST, Foyouzi-Yousefi N, Moley KH. Facilitative

glucose transporter type 1 is differentially regulated by progesterone and estrogen in

murine and human endometrial stromal cells. Endocrinology 2009; 150:1512-1520.

161. Frolova AI, Moley KH. Quantitative analysis of glucose transporter mRNAs in endometrial

stromal cells reveals critical role of GLUT1 in uterine receptivity. Endocrinology 2011;

152:2123-2128.

162. Finn CA, Keen PM. The Induction of Deciduomata in the Rat. J Embryol Exp Morphol

1963; 11:673-682.

Page 134: © 2013 Juanmahel Davila - IDEALS

119

163. Ledford BE, Rankin JC, Markwald RR, Baggett B. Biochemical and morphological

changes following artificially stimulated decidualization in the mouse uterus. Biol Reprod

1976; 15:529-535.

164. Li Q, Kannan A, Wang W, Demayo FJ, Taylor RN, Bagchi MK, Bagchi IC. Bone

morphogenetic protein 2 functions via a conserved signaling pathway involving Wnt4 to

regulate uterine decidualization in the mouse and the human. J Biol Chem 2007;

282:31725-31732.

165. Yee GM, Kennedy TG. Role of cyclic adenosine 3',5'-monophosphate in mediating the

effect of prostaglandin E2 on decidualization in vitro. Biol Reprod 1991; 45:163-171.

166. Charnock-Jones DS, Kaufmann P, Mayhew TM. Aspects of human fetoplacental

vasculogenesis and angiogenesis. I. Molecular regulation. Placenta 2004; 25:103-113.

167. Brosens I, Robertson WB, Dixon HG. The physiological response of the vessels of the

placental bed to normal pregnancy. J Pathol Bacteriol 1967; 93:569-579.

168. Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nature

medicine 1995; 1:27-31.

169. Finn CA, McLaren A. A study of the early stages of implantation in mice. J Reprod Fertil

1967; 13:259-267.

170. Bany BM, Hamilton GS. Assessment of permeability barriers to macromolecules in the

rodent endometrium at the onset of implantation. Methods Mol Biol 2011; 763:83-94.

171. Deb K, Reese J, Paria BC. Methodologies to study implantation in mice. Methods Mol

Med 2006; 121:9-34.

172. Kennedy TG, Gillio-Meina C, Phang SH. Prostaglandins and the initiation of blastocyst

implantation and decidualization. Reproduction 2007; 134:635-643.

173. Geusens N, Hering L, Verlohren S, Luyten C, Drijkoningen K, Taube M, Vercruysse L,

Hanssens M, Dechend R, Pijnenborg R. Changes in endovascular trophoblast invasion and

spiral artery remodelling at term in a transgenic preeclamptic rat model. Placenta 2010;

31:320-326.

174. Kaufmann P, Black S, Huppertz B. Endovascular trophoblast invasion: implications for the

pathogenesis of intrauterine growth retardation and preeclampsia. Biol Reprod 2003; 69:1-

7.

175. Chakraborty I, Das SK, Dey SK. Differential expression of vascular endothelial growth

factor and its receptor mRNAs in the mouse uterus around the time of implantation. J

Endocrinol 1995; 147:339-352.

176. Albrecht ED, Babischkin JS, Lidor Y, Anderson LD, Udoff LC, Pepe GJ. Effect of estrogen

on angiogenesis in co-cultures of human endometrial cells and microvascular endothelial

cells. Hum Reprod 2003; 18:2039-2047.

177. Matsumoto H, Ma WG, Daikoku T, Zhao X, Paria BC, Das SK, Trzaskos JM, Dey SK.

Cyclooxygenase-2 differentially directs uterine angiogenesis during implantation in mice.

J Biol Chem 2002; 277:29260-29267.

178. Halder JB, Zhao X, Soker S, Paria BC, Klagsbrun M, Das SK, Dey SK. Differential

expression of VEGF isoforms and VEGF(164)-specific receptor neuropilin-1 in the mouse

uterus suggests a role for VEGF(164) in vascular permeability and angiogenesis during

implantation. Genesis 2000; 26:213-224.

179. Sugino N, Kashida S, Karube-Harada A, Takiguchi S, Kato H. Expression of vascular

endothelial growth factor (VEGF) and its receptors in human endometrium throughout the

menstrual cycle and in early pregnancy. Reproduction 2002; 123:379-387.

Page 135: © 2013 Juanmahel Davila - IDEALS

120

180. Ferrara N. Vascular endothelial growth factor and the regulation of angiogenesis. Recent

Prog Horm Res 2000; 55:15-35; discussion 35-16.

181. Ferrara N, Carver-Moore K, Chen H, Dowd M, Lu L, O'Shea KS, Powell-Braxton L, Hillan

KJ, Moore MW. Heterozygous embryonic lethality induced by targeted inactivation of the

VEGF gene. Nature 1996; 380:439-442.

182. Carmeliet P, Ferreira V, Breier G, Pollefeyt S, Kieckens L, Gertsenstein M, Fahrig M,

Vandenhoeck A, Harpal K, Eberhardt C, Declercq C, Pawling J, et al. Abnormal blood

vessel development and lethality in embryos lacking a single VEGF allele. Nature 1996;

380:435-439.

183. Krussel JS, Bielfeld P, Polan ML, Simon C. Regulation of embryonic implantation. Eur J

Obstet Gynecol Reprod Biol 2003; 110 Suppl 1:S2-9.

184. Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J. Vascular-specific

growth factors and blood vessel formation. Nature 2000; 407:242-248.

185. Douglas NC, Tang H, Gomez R, Pytowski B, Hicklin DJ, Sauer CM, Kitajewski J, Sauer

MV, Zimmermann RC. Vascular endothelial growth factor receptor 2 (VEGFR-2)

functions to promote uterine decidual angiogenesis during early pregnancy in the mouse.

Endocrinology 2009; 150:3845-3854.

186. Kapiteijn K, Koolwijk P, van der Weiden RM, van Nieuw Amerongen G, Plaisier M, van

Hinsbergh VW, Helmerhorst FM. Human embryo-conditioned medium stimulates in vitro

endometrial angiogenesis. Fertility and sterility 2006; 85 Suppl 1:1232-1239.

187. Plaisier M, Rodrigues S, Willems F, Koolwijk P, van Hinsbergh VW, Helmerhorst FM.

Different degrees of vascularization and their relationship to the expression of vascular

endothelial growth factor, placental growth factor, angiopoietins, and their receptors in

first-trimester decidual tissues. Fertility and sterility 2007; 88:176-187.

188. Cooper JC, Sharkey AM, McLaren J, Charnock-Jones DS, Smith SK. Localization of

vascular endothelial growth factor and its receptor, flt, in human placenta and decidua by

immunohistochemistry. J Reprod Fertil 1995; 105:205-213.

189. Charnock-Jones DS, Sharkey AM, Boocock CA, Ahmed A, Plevin R, Ferrara N, Smith

SK. Vascular endothelial growth factor receptor localization and activation in human

trophoblast and choriocarcinoma cells. Biol Reprod 1994; 51:524-530.

190. Hyder SM, Stancel GM. Regulation of angiogenic growth factors in the female

reproductive tract by estrogens and progestins. Mol Endocrinol 1999; 13:806-811.

191. Hastings JM, Licence DR, Burton GJ, Charnock-Jones DS, Smith SK. Soluble vascular

endothelial growth factor receptor 1 inhibits edema and epithelial proliferation induced by

17beta-estradiol in the mouse uterus. Endocrinology 2003; 144:326-334.

192. Ma W, Tan J, Matsumoto H, Robert B, Abrahamson DR, Das SK, Dey SK. Adult tissue

angiogenesis: evidence for negative regulation by estrogen in the uterus. Mol Endocrinol

2001; 15:1983-1992.

193. Walter LM, Rogers PA, Girling JE. The role of progesterone in endometrial angiogenesis

in pregnant and ovariectomised mice. Reproduction 2005; 129:765-777.

194. Das A, Mantena SR, Kannan A, Evans DB, Bagchi MK, Bagchi IC. De novo synthesis of

estrogen in pregnant uterus is critical for stromal decidualization and angiogenesis. Proc

Natl Acad Sci U S A 2009; 106:12542-12547.

195. Wulff C, Wilson H, Dickson SE, Wiegand SJ, Fraser HM. Hemochorial placentation in the

primate: expression of vascular endothelial growth factor, angiopoietins, and their

receptors throughout pregnancy. Biol Reprod 2002; 66:802-812.

Page 136: © 2013 Juanmahel Davila - IDEALS

121

196. Yu Q, Stamenkovic I. Angiopoietin-2 is implicated in the regulation of tumor angiogenesis.

Am J Pathol 2001; 158:563-570.

197. Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C, Compton

D, McClain J, Aldrich TH, Papadopoulos N, Daly TJ, Davis S, et al. Angiopoietin-2, a

natural antagonist for Tie2 that disrupts in vivo angiogenesis. Science 1997; 277:55-60.

198. Hess AP, Hirchenhain J, Schanz A, Talbi S, Hamilton AE, Giudice LC, Krussel JS.

Angiopoietin-1 and -2 mRNA and protein expression in mouse preimplantation embryos

and uteri suggests a role in angiogenesis during implantation. Reprod Fertil Dev 2006;

18:509-516.

199. Ryan HE, Lo J, Johnson RS. HIF-1alpha is required for solid tumor formation and

embryonic vascularization. The EMBO journal 1998; 17:3005-3015.

200. Rodesch F, Simon P, Donner C, Jauniaux E. Oxygen measurements in endometrial and

trophoblastic tissues during early pregnancy. Obstetrics and gynecology 1992; 80:283-285.

201. Simon MC, Keith B. The role of oxygen availability in embryonic development and stem

cell function. Nature reviews. Molecular cell biology 2008; 9:285-296.

202. Xue Y, Li NL, Yang JY, Chen Y, Yang LL, Liu WC. Phosphatidylinositol 3'-kinase

signaling pathway is essential for Rac1-induced hypoxia-inducible factor-1(alpha) and

vascular endothelial growth factor expression. Am J Physiol Heart Circ Physiol 2011;

300:H2169-2176.

203. Du J, Xu R, Hu Z, Tian Y, Zhu Y, Gu L, Zhou L. PI3K and ERK-induced Rac1 activation

mediates hypoxia-induced HIF-1alpha expression in MCF-7 breast cancer cells. PloS one

2011; 6:e25213.

204. Xue Y, Bi F, Zhang X, Zhang S, Pan Y, Liu N, Shi Y, Yao X, Zheng Y, Fan D. Role of

Rac1 and Cdc42 in hypoxia induced p53 and von Hippel-Lindau suppression and

HIF1alpha activation. Int J Cancer 2006; 118:2965-2972.

205. Majmundar AJ, Wong WJ, Simon MC. Hypoxia-inducible factors and the response to

hypoxic stress. Mol Cell 2010; 40:294-309.

206. Matsumoto H, Sato E. Uterine angiogenesis during implantation and decidualization in

mice. Reproductive Medicine and Biology 2006; 5:81-86.

207. Daikoku T, Matsumoto H, Gupta RA, Das SK, Gassmann M, DuBois RN, Dey SK.

Expression of hypoxia-inducible factors in the peri-implantation mouse uterus is regulated

in a cell-specific and ovarian steroid hormone-dependent manner. Evidence for differential

function of HIFs during early pregnancy. J Biol Chem 2003; 278:7683-7691.

208. Ema M, Taya S, Yokotani N, Sogawa K, Matsuda Y, Fujii-Kuriyama Y. A novel bHLH-

PAS factor with close sequence similarity to hypoxia-inducible factor 1alpha regulates the

VEGF expression and is potentially involved in lung and vascular development. Proc Natl

Acad Sci U S A 1997; 94:4273-4278.

209. Kayisli UA, Luk J, Guzeloglu-Kayisli O, Seval Y, Demir R, Arici A. Regulation of

angiogenic activity of human endometrial endothelial cells in culture by ovarian steroids.

J Clin Endocrinol Metab 2004; 89:5794-5802.

210. Greb RR, Heikinheimo O, Williams RF, Hodgen GD, Goodman AL. Vascular endothelial

growth factor in primate endometrium is regulated by oestrogen-receptor and

progesterone-receptor ligands in vivo. Hum Reprod 1997; 12:1280-1292.

211. Perrot-Applanat M, Ancelin M, Buteau-Lozano H, Meduri G, Bausero P. Ovarian steroids

in endometrial angiogenesis. Steroids 2000; 65:599-603.

Page 137: © 2013 Juanmahel Davila - IDEALS

122

212. Tille JC, Pepper MS. Mesenchymal cells potentiate vascular endothelial growth factor-

induced angiogenesis in vitro. Exp Cell Res 2002; 280:179-191.

213. Yu J, Wu J, Bagchi IC, Bagchi MK, Sidell N, Taylor RN. Disruption of gap junctions

reduces biomarkers of decidualization and angiogenesis and increases inflammatory

mediators in human endometrial stromal cell cultures. Mol Cell Endocrinol 2011; 344:25-

34.

214. Cross JC. Trophoblast cell fate specification. In: Mosffet A, Loke C, McLaren A (eds.),

Biology and Pathology of Trophoblast. New York: Cambridge Press; 2006: 3-14.

215. Ivaskevicius V, Biswas A, Thomas A, Lyonga S, Rott H, Halimeh S, Kappert G,

Klammroth R, Scholz U, Eberl W, Harbrecht U, Gnida C, et al. A common F13A1 intron

1 variant IVS1+12(A) is associated with mild FXIII deficiency in Caucasian population.

Ann Hematol 2013.

216. Burton GJ, Hempstock J, Jauniaux E. Nutrition of the human fetus during the first

trimester--a review. Placenta 2001; 22 Suppl A:S70-77.

217. Pantaleon M, Kaye PL. Glucose transporters in preimplantation development. Reviews of

Reproduction 1998; 3:77-81.

218. Gardner HG, Kaye PL. Characterization of Glucose-Transport in Preimplantation Mouse

Embryos. Reproduction Fertility and Development 1995; 7:41-50.

219. Cheon YP, Li Q, Xu X, DeMayo FJ, Bagchi IC, Bagchi MK. A genomic approach to

identify novel progesterone receptor regulated pathways in the uterus during implantation.

Mol Endocrinol 2002; 16:2853-2871.

220. Georgiades P, Ferguson-Smith AC, Burton GJ. Comparative developmental anatomy of

the murine and human definitive placentae. Placenta 2002; 23:3-19.

221. Menkhorst EM, Lane N, Winship AL, Li P, Yap J, Meehan K, Rainczuk A, Stephens A,

Dimitriadis E. Decidual-secreted factors alter invasive trophoblast membrane and secreted

proteins implying a role for decidual cell regulation of placentation. PLoS One 2012;

7:e31418.

222. Kirby DR. The development of mouse blastocysts transplanted to the scrotal and

cryptorchid testis. J Anat 1963; 97:119-130.

223. Kirby DR. Development of mouse eggs beneath the kidney capsule. Nature 1960; 187:707-

708.

224. Kirby DRS. Reciprocal Transplantation of Blastocysts between Rats and Mice. Nature

1962; 194:785-&.

225. Porter DG. Observations on the development of mouse blastocysts transferred to the testis

and kidney. American Journal of Anatomy 1967; 121:73-85.

226. Kemp B, Kertschanska S, Kadyrov M, Rath W, Kaufmann P, Huppertz B. Invasive depth

of extravillous trophoblast correlates with cellular phenotype: a comparison of intra- and

extrauterine implantation sites. Histochemistry and Cell Biology 2002; 117:401-414.

227. Spornitz UM. Pseudo-Decidualization at the Site of Implantation in Tubal Pregnancy.

Archives of Gynecology and Obstetrics 1993; 253:85-95.

228. Fonseca BM, Correia-da-Silva G, Teixeira NA. The rat as an animal model for

fetoplacental development: a reappraisal of the post-implantation period. Reprod Biol

2012; 12:97-118.

229. Craven CM, Morgan T, Ward K. Decidual spiral artery remodelling begins before cellular

interaction with cytotrophoblasts. Placenta 1998; 19:241-252.

Page 138: © 2013 Juanmahel Davila - IDEALS

123

230. Hirano H, Imai Y, Ito H. Spiral artery of placenta: development and pathology-

immunohistochemical, microscopical, and electron-microscopic study. Kobe J Med Sci

2002; 48:13-23.

231. Pijnenborg R, Bland JM, Robertson WB, Brosens I. Uteroplacental arterial changes related

to interstitial trophoblast migration in early human pregnancy. Placenta 1983; 4:397-413.

232. Kam EP, Gardner L, Loke YW, King A. The role of trophoblast in the physiological change

in decidual spiral arteries. Hum Reprod 1999; 14:2131-2138.

233. Benirschke K KP (ed.). Pathology of the Human Placenta. New York: Springer; 2000.

234. Aubuchon M, Schulz LC, Schust DJ. Preeclampsia: animal models for a human cure. Proc

Natl Acad Sci U S A 2011; 108:1197-1198.

235. Dokras A, Hoffmann DS, Eastvold JS, Kienzle MF, Gruman LM, Kirby PA, Weiss RM,

Davisson RL. Severe feto-placental abnormalities precede the onset of hypertension and

proteinuria in a mouse model of preeclampsia. Biol Reprod 2006; 75:899-907.

236. Rasmussen S, Irgens LM. Fetal growth and body proportion in preeclampsia. Obstet

Gynecol 2003; 101:575-583.

237. Long PA, Abell DA, Beischer NA. Fetal growth retardation and pre-eclampsia. Br J Obstet

Gynaecol 1980; 87:13-18.

238. Hall A. Rho GTPases and the actin cytoskeleton. Science 1998; 279:509-514.

239. Bishop AL, Hall A. Rho GTPases and their effector proteins. Biochemical Journal 2000;

348:241-255.

240. Boettner B, Van Aelst L. The role of Rho GTPases in disease development. Gene 2002;

286:155-174.

241. Boureux A, Vignal E, Faure S, Fort P. Evolution of the Rho family of ras-like GTPases in

eukaryotes. Mol Biol Evol 2007; 24:203-216.

242. Brown JH, Del Re DP, Sussman MA. The Rac and Rho hall of fame: a decade of

hypertrophic signaling hits. Circulation Research 2006; 98:730-742.

243. Bustelo XR, Sauzeau V, Berenjeno IM. GTP-binding proteins of the Rho/Rac family:

regulation, effectors and functions in vivo. Bioessays 2007; 29:356-370.

244. Hall A. Rho family GTPases. Biochem Soc Trans 2012; 40:1378-1382.

245. Pai SY, Kim C, Williams DA. Rac GTPases in human diseases. Dis Markers 2010; 29:177-

187.

246. Ridley AJ. Signaling networks, Rho GTPases and cell migration. Molecular Biology of the

Cell 1999; 10:117a-117a.

247. Ridley AJ. Rho GTPases and actin dynamics in membrane protrusions and vesicle

trafficking. Trends Cell Biol 2006; 16:522-529.

248. Ridley AJ, Schwartz MA, Burridge K, Firtel RA, Ginsberg MH, Borisy G, Parsons JT,

Horwitz AR. Cell migration: integrating signals from front to back. Science 2003;

302:1704-1709.

249. Madaule P, Axel R. A Novel Ras-Related Gene Family. Cell 1985; 41:31-40.

250. Didsbury J, Weber RF, Bokoch GM, Evans T, Snyderman R. rac, a Novel Ras-Related

Family of Proteins That Are Botulinum Toxin Substrates. Journal of Biological Chemistry

1989; 264:16378-16382.

251. Moll J, Sansig G, Fattori E, van der Putten H. The murine rac1 gene: cDNA cloning, tissue

distribution and regulated expression of rac1 mRNA by disassembly of actin

microfilaments. Oncogene 1991; 6:863-866.

Page 139: © 2013 Juanmahel Davila - IDEALS

124

252. Munemitsu S, Innis MA, Clark R, Mccormick F, Ullrich A, Polakis P. Molecular-Cloning

and Expression of a G25k Cdna, the Human Homolog of the Yeast-Cell Cycle Gene Cdc42.

Molecular and Cellular Biology 1990; 10:5977-5982.

253. Tapon N, Hall A. Rho, Rac and Cdc42 GTPases regulate the organization of the actin

cytoskeleton. Current Opinion in Cell Biology 1997; 9:86-92.

254. Hall A, Nobes CD. Rho GTPases: molecular switches that control the organization and

dynamics of the actin cytoskeleton. Philosophical transactions of the Royal Society of

London. Series B, Biological sciences 2000; 355:965-970.

255. Jaffe AB, Hall A. Rho GTPases: biochemistry and biology. Annu Rev Cell Dev Biol 2005;

21:247-269.

256. Pedersen E, Brakebusch C. Rho GTPase Function in Cell Morphogenesis. In: Encyclopedia

of Life Sciences (ELS): John Wiley & Sons, Ltd: Chichester; 2001.

257. Raftopoulou M, Hall A. Cell migration: Rho GTPases lead the way. Developmental

Biology 2004; 265:23-32.

258. VanAelst L, DSouzaSchorey C. Rho GTPases and signaling networks. Genes &

Development 1997; 11:2295-2322.

259. Wherlock M, Mellor H. The Rho GTPase family: a Racs to Wrchs story. J Cell Sci 2002;

115:239-240.

260. Etienne-Manneville S, Hall A. Rho GTPases in cell biology. Nature 2002; 420:629-635.

261. Heasman SJ, Ridley AJ. Mammalian Rho GTPases: new insights into their functions from

in vivo studies. Nature Reviews Molecular Cell Biology 2008; 9:690-701.

262. Nobes CD, Hall A. Rho GTPases control polarity, protrusion, and adhesion during cell

movement. J Cell Biol 1999; 144:1235-1244.

263. Symons M, Rusk N. Control of vesicular trafficking by Rho GTPases. Curr Biol 2003;

13:R409-418.

264. Takai Y, Sasaki T, Matozaki T. Small GTP-binding proteins. Physiol Rev 2001; 81:153-

208.

265. Vega FM, Ridley AJ. Rho GTPases in cancer cell biology. Febs Letters 2008; 582:2093-

2101.

266. Bosco EE, Mulloy JC, Zheng Y. Rac1 GTPase: a "Rac" of all trades. Cell Mol Life Sci

2009; 66:370-374.

267. Glogauer M, Marchal CC, Zhu F, Worku A, Clausen BE, Foerster I, Marks P, Downey GP,

Dinauer M, Kwiatkowski DJ. Rac1 deletion in mouse neutrophils has selective effects on

neutrophil functions. Journal of immunology 2003; 170:5652-5657.

268. Haataja L, Groffen J, Heisterkamp N. Characterization of RAC3, a novel member of the

Rho family. J Biol Chem 1997; 272:20384-20388.

269. Hall A. Small GTP-binding proteins--a new family of biologic regulators. American

journal of respiratory cell and molecular biology 1992; 6:245-246.

270. Ridley AJ, Hall A. The Small Gtp-Binding Protein Rho Regulates the Assembly of Focal

Adhesions and Actin Stress Fibers in Response to Growth-Factors. Cell 1992; 70:389-399.

271. Sugihara K, Nakatsuji N, Nakamura K, Nakao K, Hashimoto R, Otani H, Sakagami H,

Kondo H, Nozawa S, Aiba A, Katsuki M. Rac1 is required for the formation of three germ

layers during gastrulation. Oncogene 1998; 17:3427-3433.

272. Hall A. Ras-related GTPases and the cytoskeleton. Molecular biology of the cell 1992;

3:475-479.

Page 140: © 2013 Juanmahel Davila - IDEALS

125

273. Nobes C, Hall A. Regulation and function of the Rho subfamily of small GTPases. Current

opinion in genetics & development 1994; 4:77-81.

274. Nobes CD, Hall A. Rho, rac and cdc42 GTPases: regulators of actin structures, cell

adhesion and motility. Biochemical Society transactions 1995; 23:456-459.

275. Sawada N, Salomone S, Kim HH, Kwiatkowski DJ, Liao JK. Regulation of endothelial

nitric oxide synthase and postnatal angiogenesis by Rac1. Circ Res 2008; 103:360-368.

276. Silbert O, Wang Y, Maciejewski BS, Lee HS, Shaw SK, Sanchez-Esteban J. Roles of RhoA

and Rac1 on actin remodeling and cell alignment and differentiation in fetal type II

epithelial cells exposed to cyclic mechanical stretch. Exp Lung Res 2008; 34:663-680.

277. Tan W, Palmby TR, Gavard J, Amornphimoltham P, Zheng Y, Gutkind JS. An essential

role for Rac1 in endothelial cell function and vascular development. FASEB J 2008;

22:1829-1838.

278. Wells CM, Walmsley M, Ooi S, Tybulewicz V, Ridley AJ. Rac1-deficient macrophages

exhibit defects in cell spreading and membrane ruffling but not migration. J Cell Sci 2004;

117:1259-1268.

279. Wheeler AP, Wells CM, Smith SD, Vega FM, Henderson RB, Tybulewicz VL, Ridley AJ.

Rac1 and Rac2 regulate macrophage morphology but are not essential for migration. J Cell

Sci 2006; 119:2749-2757.

280. Sun D, Xu D, Zhang BL. Rac signaling in tumorigenesis and as target for anticancer drug

development. Drug Resistance Updates 2006; 9:274-287.

281. Di-Poi N, Faure J, Grizot S, Molnar G, Pick E, Dagher MC. Mechanism of NADPH

oxidase activation by the Rac/Rho-GDI complex. Biochemistry 2001; 40:10014-10022.

282. Abo A, Pick E, Hall A, Totty N, Teahan CG, Segal AW. Activation of the NADPH oxidase

involves the small GTP-binding protein p21rac1. Nature 1991; 353:668-670.

283. Matos P, Skaug J, Marques B, Beck S, Verissimo F, Gespach C, Boavida MG, Scherer

SW, Jordan P. Small GTPase Rac1: structure, localization, and expression of the human

gene. Biochem Biophys Res Commun 2000; 277:741-751.

284. Ishii S, Merlino GT, Pastan I. Promoter region of the human Harvey ras proto-oncogene:

similarity to the EGF receptor proto-oncogene promoter. Science 1985; 230:1378-1381.

285. Bourgine J, Garat A, Allorge D, Crunelle-Thibaut A, Lo-Guidice JM, Colombel JF, Broly

F, Billaut-Laden I. Evidence for a functional genetic polymorphism of the Rho-GTPase

Rac1. Implication in azathioprine response? Pharmacogenet Genomics 2011; 21:313-324.

286. Scheffzek K, Stephan I, Jensen ON, Illenberger D, Gierschik P. The Rac-RhoGDI complex

and the structural basis for the regulation of Rho proteins by RhoGDI. Nature structural

biology 2000; 7:122-126.

287. Cherfils J, Zeghouf M. REGULATION OF SMALL GTPases BY GEFs, GAPs, AND

GDIs. Physiological Reviews 2013; 93:269-309.

288. Kilian KA, Bugarija B, Lahn BT, Mrksich M. Geometric cues for directing the

differentiation of mesenchymal stem cells. Proc Natl Acad Sci U S A 2010; 107:4872-

4877.

289. Chen CS, Mrksich M, Huang S, Whitesides GM, Ingber DE. Geometric control of cell life

and death. Science 1997; 276:1425-1428.

290. Roberts AW, Kim C, Zhen L, Lowe JB, Kapur R, Petryniak B, Spaetti A, Pollock JD,

Borneo JB, Bradford GB, Atkinson SJ, Dinauer MC, et al. Deficiency of the hematopoietic

cell-specific Rho family GTPase Rac2 is characterized by abnormalities in neutrophil

function and host defense. Immunity 1999; 10:183-196.

Page 141: © 2013 Juanmahel Davila - IDEALS

126

291. Jaffe AB, Hall A. Rho GTPases in transformation and metastasis. Adv Cancer Res 2002;

84:57-80.

292. Jordan P, Brazao R, Boavida MG, Gespach C, Chastre E. Cloning of a novel human Rac1b

splice variant with increased expression in colorectal tumors. Oncogene 1999; 18:6835-

6839.

293. Matos P, Collard JG, Jordan P. Tumor-related alternatively spliced Rac1b is not regulated

by Rho-GDP dissociation inhibitors and exhibits selective downstream signaling. J Biol

Chem 2003; 278:50442-50448.

294. Schnelzer A, Prechtel D, Knaus U, Dehne K, Gerhard M, Graeff H, Harbeck N, Schmitt

M, Lengyel E. Rac1 in human breast cancer: overexpression, mutation analysis, and

characterization of a new isoform, Rac1b. Oncogene 2000; 19:3013-3020.

295. Singh A, Karnoub AE, Palmby TR, Lengyel E, Sondek J, Der CJ. Rac1b, a tumor

associated, constitutively active Rac1 splice variant, promotes cellular transformation.

Oncogene 2004; 23:9369-9380.

296. Zhou C, Licciulli S, Avila JL, Cho M, Troutman S, Jiang P, Kossenkov AV, Showe LC,

Liu Q, Vachani A, Albelda SM, Kissil JL. The Rac1 splice form Rac1b promotes K-ras-

induced lung tumorigenesis. Oncogene 2013; 32:903-909.

297. Fiegen D, Haeusler LC, Blumenstein L, Herbrand U, Dvorsky R, Vetter IR, Ahmadian

MR. Alternative splicing of Rac1 generates Rac1b, a self-activating GTPase. J Biol Chem

2004; 279:4743-4749.

298. Shirsat NV, Pibnolo RJ, Kreider BL, Rovera G. A Member of the Ras Gene Superfamily

Is Expressed Specifically in T B and Myeloid Hemopoietic Cells. Oncogene 1990; 5:769-

772.

299. Corbetta S, Gualdoni S, Albertinazzi C, Paris S, Croci L, Consalez GG, de Curtis I.

Generation and characterization of Rac3 knockout mice. Mol Cell Biol 2005; 25:5763-

5776.

300. Nicola C, Lala PK, Chakraborty C. Prostaglandin E2-mediated migration of human

trophoblast requires RAC1 and CDC42. Biol Reprod 2008; 78:976-982.

301. Stappenbeck TS, Gordon JI. Rac1 mutations produce aberrant epithelial differentiation in

the developing and adult mouse small intestine. Development 2000; 127:2629-2642.

302. Bopp A, Wartlick F, Henninger C, Kaina B, Fritz G. Rac1 modulates acute and subacute

genotoxin-induced hepatic stress responses, fibrosis and liver aging. Cell Death Dis 2013;

4:e558.

303. Del Valle-Perez B, Martinez VG, Lacasa-Salavert C, Figueras A, Shapiro SS, Takafuta T,

Casanovas O, Capella G, Ventura F, Vinals F. Filamin B plays a key role in vascular

endothelial growth factor-induced endothelial cell motility through its interaction with

Rac-1 and Vav-2. J Biol Chem 2010; 285:10748-10760.

304. Wang F, Yamauchi M, Muramatsu M, Osawa T, Tsuchida R, Shibuya M. RACK1

regulates VEGF/Flt1-mediated cell migration via activation of a PI3K/Akt pathway. J Biol

Chem 2011; 286:9097-9106.

305. Crespo P, Bustelo XR, Aaronson DS, Coso OA, Lopez-Barahona M, Barbacid M, Gutkind

JS. Rac-1 dependent stimulation of the JNK/SAPK signaling pathway by Vav. Oncogene

1996; 13:455-460.

306. Salojin KV, Zhang J, Delovitch TL. TCR and CD28 are coupled via ZAP-70 to the

activation of the Vav/Rac-1-/PAK-1/p38 MAPK signaling pathway. Journal of

immunology 1999; 163:844-853.

Page 142: © 2013 Juanmahel Davila - IDEALS

127

307. Zhang J, Salojin KV, Delovitch TL. CD28 co-stimulation restores T cell responsiveness in

NOD mice by overcoming deficiencies in Rac-1/p38 mitogen-activated protein kinase

signaling and IL-2 and IL-4 gene transcription. International immunology 2001; 13:377-

384.

308. Chi X, Wang S, Huang Y, Stamnes M, Chen JL. Roles of rho GTPases in intracellular

transport and cellular transformation. Int J Mol Sci 2013; 14:7089-7108.

309. Nobes C, Marsh M. Dendritic cells: new roles for Cdc42 and Rac in antigen uptake? Curr

Biol 2000; 10:R739-741.

310. Matas OB, Fritz S, Luna A, Egea G. Membrane trafficking at the ER/Golgi interface:

Functional implications of RhoA and Rac1. European Journal of Cell Biology 2005;

84:699-707.

311. Lamaze C, Chuang TH, Terlecky LJ, Bokoch GM, Schmid SL. Regulation of receptor-

mediated endocytosis by Rho and Rac. Nature 1996; 382:177-179.

312. Malecz N, McCabe PC, Spaargaren C, Qiu RG, Chuang Y, Symons M. Synaptojanin 2, a

novel Rac1 effector that regulates clathrin-mediated endocytosis. Molecular Biology of the

Cell 2000; 11:326a-326a.

313. Oikawa T Fau - Yamaguchi H, Yamaguchi H Fau - Itoh T, Itoh T Fau - Kato M, Kato M

Fau - Ijuin T, Ijuin T Fau - Yamazaki D, Yamazaki D Fau - Suetsugu S, Suetsugu S Fau -

Takenawa T, Takenawa T. PtdIns(3,4,5)P3 binding is necessary for WAVE2-induced

formation of lamellipodia.

314. Balamatsias D, Kong AM, Waters JE, Sriratana A, Gurung R, Bailey CG, Rasko JEJ,

Tiganis T, Macaulay SL, Mitchell CA. Identification of P-Rex1 as a Novel Rac1-Guanine

Nucleotide Exchange Factor (GEF) That Promotes Actin Remodeling and GLUT4 Protein

Trafficking in Adipocytes. Journal of Biological Chemistry 2011; 286:43229-43240.

315. Vidal-Quadras M, Gelabert-Baldrich M, Soriano-Castell D, Llado A, Rentero C, Calvo M,

Pol A, Enrich C, Tebar F. Rac1 and Calmodulin Interactions Modulate Dynamics of ARF6-

Dependent Endocytosis. Traffic 2011; 12:1879-1896.

316. Doussau F, Gasman S, Humeau Y, Vitiello F, Popoff M, Boquet P, Bader MF, Poulain B.

A Rho-related GTPase is involved in Ca2+-dependent neurotransmitter exocytosis. Journal

of Biological Chemistry 2000; 275:7764-7770.

317. Li Q Fau - Ho CS, Ho Cs Fau - Marinescu V, Marinescu V Fau - Bhatti H, Bhatti H Fau -

Bokoch GM, Bokoch Gm Fau - Ernst SA, Ernst Sa Fau - Holz RW, Holz Rw Fau - Stuenkel

EL, Stuenkel EL. Facilitation of Ca(2+)-dependent exocytosis by Rac1-GTPase in bovine

chromaffin cells.

318. Bi Y, Williams JA. A role for Rho and Rac in secretagogue-induced amylase release by

pancreatic acini. Am J Physiol Cell Physiol 2005; 289:C22-32.

319. Hirota K, Semenza GL. Rac1 activity is required for the activation of hypoxia-inducible

factor 1. J Biol Chem 2001; 276:21166-21172.

320. Turcotte S, Desrosiers RR, Beliveau R. HIF-1alpha mRNA and protein upregulation

involves Rho GTPase expression during hypoxia in renal cell carcinoma. J Cell Sci 2003;

116:2247-2260.

321. Zhan H, Liang H, Liu X, Deng J, Wang B, Hao X. Expression of Rac1, HIF-1alpha, and

VEGF in gastric carcinoma: correlation with angiogenesis and prognosis. Onkologie 2013;

36:102-107.

Page 143: © 2013 Juanmahel Davila - IDEALS

128

322. Zhang XZ, Huang X, Qiao JH, Zhang JJ, Zhang MX. Inhibition of hypoxia-induced retinal

neovascularization in mice with short hairpin RNA targeting Rac1, possibly via blockading

redox signaling. Exp Eye Res 2011; 92:473-481.

323. Li SM, Zeng LW, Feng L, Chen DB. Rac1-dependent intracellular superoxide formation

mediates vascular endothelial growth factor-induced placental angiogenesis in vitro.

Endocrinology 2010; 151:5315-5325.

324. Diebold I, Djordjevic T, Hess J, Gorlach A. Rac-1 promotes pulmonary artery smooth

muscle cell proliferation by upregulation of plasminogen activator inhibitor-1: role of

NFkappaB-dependent hypoxia-inducible factor-1alpha transcription. Thromb Haemost

2008; 100:1021-1028.

325. Diebold I, Petry A, Djordjevic T, Belaiba RS, Fineman J, Black S, Schreiber C, Fratz S,

Hess J, Kietzmann T, Gorlach A. Reciprocal regulation of Rac1 and PAK-1 by HIF-1alpha:

a positive-feedback loop promoting pulmonary vascular remodeling. Antioxid Redox

Signal 2010; 13:399-412.

326. Hoang MV, Nagy JA, Senger DR. Active Rac1 improves pathologic VEGF neovessel

architecture and reduces vascular leak: mechanistic similarities with angiopoietin-1. Blood

2011; 117:1751-1760.

327. Covello Kl Fau - Simon MC, Simon MC. HIFs, hypoxia, and vascular development.

328. Cowden Dahl KD, Fryer BH, Mack FA, Compernolle V, Maltepe E, Adelman DM,

Carmeliet P, Simon MC. Hypoxia-inducible factors 1α and 2α regulate trophoblast

differentiation. Mol Cell Biol 2005; 25:10479-10491.

329. Hickey MM, Simon MC. Regulation of angiogenesis by hypoxia and hypoxia-inducible

factors. Curr Top Dev Biol 2006; 76:217-257.

330. Maes C Fau - Carmeliet G, Carmeliet G Fau - Schipani E, Schipani E. Hypoxia-driven

pathways in bone development, regeneration and disease.

331. Semenza GL. Hypoxia-inducible factors in physiology and medicine.

332. Liu L, Simon MC. Regulation of transcription and translation by hypoxia. Cancer Biol

Ther 2004; 3:492-497.

333. Patel SA, Simon MC. Biology of hypoxia-inducible factor-2alpha in development and

disease. Cell Death Differ 2008; 15:628-634.

334. Adelman DM, Gertsenstein M, Nagy A, Simon MC, Maltepe E. Placental cell fates are

regulated in vivo by HIF-mediated hypoxia responses. Genes & Development 2000;

14:3191-3203.

335. Skuli N, Simon MC. HIF-1alpha versus HIF-2alpha in endothelial cells and vascular

functions: is there a master in angiogenesis regulation? Cell Cycle 2009; 8:3252-3253.

336. Stem Cells: Scientific Progress and Future Research Directions. In. Bethesda, MD: US

Department of Health and Human Services; 2001.

337. Lim HJ, Wang H. Uterine disorders and pregnancy complications: insights from mouse

models. J Clin Invest 2010; 120:1004-1015.

338. Ihara K, Muraguchi S, Kato M, Shimizu T, Shirakawa M, Kuroda S, Kaibuchi K,

Hakoshima T. Crystal structure of human RhoA in a dominantly active form complexed

with a GTP analogue. J Biol Chem 1998; 273:9656-9666.

339. Krauthammer M, Kong Y, Ha BH, Evans P, Bacchiocchi A, McCusker JP, Cheng E, Davis

MJ, Goh G, Choi M, Ariyan S, Narayan D, et al. Exome sequencing identifies recurrent

somatic RAC1 mutations in melanoma. Nat Genet 2012; 44:1006-1014.

Page 144: © 2013 Juanmahel Davila - IDEALS

129

340. Phillips MJ, Calero G, Chan B, Ramachandran S, Cerione RA. Effector proteins exert an

important influence on the signaling-active state of the small GTPase Cdc42. J Biol Chem

2008; 283:14153-14164.

341. Brunger AT, Milburn MV, Tong L, deVos AM, Jancarik J, Yamaizumi Z, Nishimura S,

Ohtsuka E, Kim SH. Crystal structure of an active form of RAS protein, a complex of a

GTP analog and the HRAS p21 catalytic domain. Proc Natl Acad Sci U S A 1990; 87:4849-

4853.

342. Madej T, Addess KJ, Fong JH, Geer LY, Geer RC, Lanczycki CJ, Liu C, Lu S, Marchler-

Bauer A, Panchenko AR, Chen J, Thiessen PA, et al. MMDB: 3D structures and

macromolecular interactions. Nucleic Acids Res 2012; 40:D461-464.

343. Altschul SF, Wootton JC, Gertz EM, Agarwala R, Morgulis A, Schaffer AA, Yu YK.

Protein database searches using compositionally adjusted substitution matrices. Febs

Journal 2005; 272:5101-5109.

344. Altschul SF, Madden TL, Schaffer AA, Zhang J, Zhang Z, Miller W, Lipman DJ. Gapped

BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic

Acids Res 1997; 25:3389-3402.

345. Franco HL, Lee KY, Broaddus RR, White LD, Lanske B, Lydon JP, Jeong JW, DeMayo

FJ. Ablation of Indian hedgehog in the murine uterus results in decreased cell cycle

progression, aberrant epidermal growth factor signaling, and increased estrogen signaling.

Biol Reprod 2010; 82:783-790.

346. Finn CA. Oestrogen and the Decidual Cell Reaction of Implantation in Mice. J Endocrinol

1965; 32:223-229.

347. Gu H, Marth JD, Orban PC, Mossmann H, Rajewsky K. Deletion of a DNA-Polymerase-

Beta Gene Segment in T-Cells Using Cell-Type-Specific Gene Targeting. Science 1994;

265:103-106.

348. Soyal SM, Mukherjee A, Lee KY, Li J, Li H, DeMayo FJ, Lydon JP. Cre-mediated

recombination in cell lineages that express the progesterone receptor. genesis 2005; 41:58-

66.

349. Lydon JP, DeMayo FJ, Funk CR, Mani SK, Hughes AR, Montgomery CA, Jr., Shyamala

G, Conneely OM, O'Malley BW. Mice lacking progesterone receptor exhibit pleiotropic

reproductive abnormalities. Genes Dev 1995; 9:2266-2278.

350. Cheon YP, DeMayo FJ, Bagchi MK, Bagchi IC. Induction of cytotoxic T-lymphocyte

antigen-2 beta, a cysteine protease inhibitor in decidua - A potential regulator of embryo

implantation. Journal of Biological Chemistry 2004; 279:10357-10363.

351. Lee KY, Jeong JW, Wang J, Ma L, Martin JF, Tsai SY, Lydon JP, DeMayo FJ. Bmp2 is

critical for the murine uterine decidual response. Mol Cell Biol 2007; 27:5468-5478.

352. Das A, Li Q, Laws MJ, Kaya H, Bagchi MK, Bagchi IC. Estrogen-induced expression of

Fos-related antigen 1 (FRA-1) regulates uterine stromal differentiation and remodeling. J

Biol Chem 2012; 287:19622-19630.

353. Li Q, Kannan A, DeMayo FJ, Lydon JP, Cooke PS, Yamagishi H, Srivastava D, Bagchi

MK, Bagchi IC. The antiproliferative action of progesterone in uterine epithelium is

mediated by Hand2. Science 2011; 331:912-916.

354. Merchantlarios H, Mendlovic F, Alvarezbuylla A. Characterization of Alkaline-

Phosphatase from Primordial Germ-Cells and Ontogenesis of This Enzyme in the Mouse.

Differentiation 1985; 29:145-151.

Page 145: © 2013 Juanmahel Davila - IDEALS

130

355. Li QX, Cheon YP, Kannan A, Shanker S, Bagchi IC, Bagchi MK. A novel pathway

involving progesterone receptor, 12/15-lipoxygenase-derived eicosanoids, and peroxisome

proliferator-activated receptor gamma regulates implantation in mice. Journal of Biological

Chemistry 2004; 279:11570-11581.

356. Kannan A, Fazleabas AT, Bagchi IC, Bagchi MK. The transcription factor C/EBPbeta is a

marker of uterine receptivity and expressed at the implantation site in the primate. Reprod

Sci 2010; 17:434-443.

357. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time

quantitative PCR and the 2(T)(-Delta Delta C) method. Methods 2001; 25:402-408.

358. Laborda J. 36B4 cDNA used as an estradiol-independent mRNA control is the cDNA for

human acidic ribosomal phosphoprotein PO. Nucleic Acids Res 1991; 19:3998.

359. Athilakshmi K, Bagchi MK, Bagchi IC. Role of transcription factor Runx1 in uterine

stromal cell differentiation and maternal-fetal interaction during mouse pregnancy.

University of Illinois at Urbana-Champaign; 2013.

360. Nagashima T, Li Q, Clementi C, Lydon JP, Demayo FJ, Matzuk MM. BMPR2 is required

for postimplantation uterine function and pregnancy maintenance. J Clin Invest 2013;

123:2539-2550.

361. Nallasamy S, Li Q, Bagchi MK, Bagchi IC. Msx homeobox genes critically regulate

embryo implantation by controlling paracrine signaling between uterine stroma and

epithelium. PLoS Genet 2012; 8:e1002500.

362. Robker RL, Akison LK, Russell DL. Control of oocyte release by progesterone receptor-

regulated gene expression. Nucl Recept Signal 2009; 7:e012.

363. Park OK, Mayo KE. Transient expression of progesterone receptor messenger RNA in

ovarian granulosa cells after the preovulatory luteinizing hormone surge. Mol Endocrinol

1991; 5:967-978.

364. Kim J, Bagchi IC, Bagchi MK. Control of ovulation in mice by progesterone receptor-

regulated gene networks. Mol Hum Reprod 2009; 15:821-828.

365. Palanisamy GS, Cheon YP, Kim J, Kannan A, Li QX, Sato M, Mantena SR, Sitruk-Ware

RL, Bagchi MK, Bagchi IC. A novel pathway involving progesterone receptor, endothelin-

2, and endothelin receptor B controls ovulation in mice. Molecular Endocrinology 2006;

20:2784-2795.

366. Palanisamy GS, Cheon YP, Li QX, Sitruk-Ware R, Bagchi MK, Bagchi IC. Endothelin-2,

a novel target of progesterone receptor regulation in the mouse ovary, plays a critical role

in ovulation. Biology of Reproduction 2004:161-161.

367. Michaelson D, Abidi W, Guardavaccaro D, Zhou M, Ahearn I, Pagano M, Philips MR.

Rac1 accumulates in the nucleus during the G2 phase of the cell cycle and promotes cell

division. J Cell Biol 2008; 181:485-496.

368. Finn CA, Martin L. Patterns of cell division in the mouse uterus during early pregnancy. J

Endocrinol 1967; 39:593-597.

369. Martin L, Finn CA. Hormonal regulation of cell division in epithelial and connective

tissues of the mouse uterus. J Endocrinol 1968; 41:363-371.

370. Finn CA, Martin L. The cellular response of the uterus of the aged mouse to oestrogen and

progesterone. J Reprod Fertil 1969; 20:545-547.

371. Finn CA, Martin L. The role of the oestrogen secreted before oestrus in the preparation of

the uterus for implantation in the mouse. J Endocrinol 1970; 47:431-438.

Page 146: © 2013 Juanmahel Davila - IDEALS

131

372. Martin L, Finn CA. Interactions of oestradiol and progestins in the mouse uterus. J

Endocrinol 1970; 48:109-115.

373. Toft DJ, Linzer DI. Prolactin (PRL)-like protein J, a novel member of the PRL/growth

hormone family, is exclusively expressed in maternal decidua. Endocrinology 1999;

140:5095-5101.

374. Alam SM, Konno T, Sahgal N, Lu L, Soares MJ. Decidual cells produce a heparin-binding

prolactin family cytokine with putative intrauterine regulatory actions. J Biol Chem 2008;

283:18957-18968.

375. Esadeg S, He H, Pijnenborg R, Van Leuven F, Croy BA. Alpha-2 macroglobulin controls

trophoblast positioning in mouse implantation sites. Placenta 2003; 24:912-921.

376. Lague MN, Detmar J, Paquet M, Boyer A, Richards JS, Adamson SL, Boerboom D.

Decidual PTEN expression is required for trophoblast invasion in the mouse. Am J Physiol

Endocrinol Metab 2010; 299:E936-946.

377. Mizugishi K, Li C, Olivera A, Bielawski J, Bielawska A, Deng CX, Proia RL. Maternal

disturbance in activated sphingolipid metabolism causes pregnancy loss in mice. J Clin

Invest 2007; 117:2993-3006.

378. Rosario GX, Konno T, Soares MJ. Maternal hypoxia activates endovascular trophoblast

cell invasion. Dev Biol 2008; 314:362-375.

379. Hou Q, Gorski J. Estrogen receptor and progesterone receptor genes are expressed

differentially in mouse embryos during preimplantation development. Proc Natl Acad Sci

U S A 1993; 90:9460-9464.

380. Petiet A, Delatour B, Dhenain M. Models of neurodegenerative disease - Alzheimer's

anatomical and amyloid plaque imaging. Methods Mol Biol 2011; 771:293-308.

381. Shea K, Geijsen N. Dissection of 6.5 dpc mouse embryos. J Vis Exp 2007:160.

382. Natale DR, Starovic M, Cross JC. Phenotypic analysis of the mouse placenta. Methods

Mol Med 2006; 121:275-293.

383. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene

lists using DAVID bioinformatics resources. Nat Protoc 2009; 4:44-57.

384. Koseki-Kuno S, Yamakawa M, Dickneite G, Ichinose A. Factor XIII A subunit-deficient

mice developed severe uterine bleeding events and subsequent spontaneous miscarriages.

Blood 2003; 102:4410-4412.

385. Laws MJ, Bagchi MK, Bagchi IC. The role of ERα in the uterus during embryo

implantation and in the ovary during ovarian tumorigenesis. University of Illinois at

Urbana-Champaign; 2011. Dissertation.

386. Pitera JE, Woolf AS, Gale NW, Yancopoulos GD, Yuan HT. Dysmorphogenesis of kidney

cortical peritubular capillaries in angiopoietin-2-deficient mice. Am J Pathol 2004;

165:1895-1906.

387. Gale NW, Thurston G, Hackett SF, Renard R, Wang Q, McClain J, Martin C, Witte C,

Witte MH, Jackson D, Suri C, Campochiaro PA, et al. Angiopoietin-2 is required for

postnatal angiogenesis and lymphatic patterning, and only the latter role is rescued by

Angiopoietin-1. Dev Cell 2002; 3:411-423.

388. Adams RH, Eichmann A. Axon guidance molecules in vascular patterning. Cold Spring

Harb Perspect Biol 2010; 2:a001875.

389. Schwarz Q, Gu C, Fujisawa H, Sabelko K, Gertsenstein M, Nagy A, Taniguchi M,

Kolodkin AL, Ginty DD, Shima DT, Ruhrberg C. Vascular endothelial growth factor

Page 147: © 2013 Juanmahel Davila - IDEALS

132

controls neuronal migration and cooperates with Sema3A to pattern distinct compartments

of the facial nerve. Genes Dev 2004; 18:2822-2834.

390. Kawasaki T, Kitsukawa T, Bekku Y, Matsuda Y, Sanbo M, Yagi T, Fujisawa H. A

requirement for neuropilin-1 in embryonic vessel formation. Development 1999;

126:4895-4902.

391. Inoue M, Chang L, Hwang J, Chiang SH, Saltiel AR. The exocyst complex is required for

targeting of Glut4 to the plasma membrane by insulin. Nature 2003; 422:629-633.

392. Li Q, Ho CS, Marinescu V, Bhatti H, Bokoch GM, Ernst SA, Holz RW, Stuenkel EL.

Facilitation of Ca(2+)-dependent exocytosis by Rac1-GTPase in bovine chromaffin cells.

J Physiol 2003; 550:431-445.

393. Wolburg H, Wolburg-Buchholz K, Kraus J, Rascher-Eggstein G, Liebner S, Hamm S,

Duffner F, Grote EH, Risau W, Engelhardt B. Localization of claudin-3 in tight junctions

of the blood-brain barrier is selectively lost during experimental autoimmune

encephalomyelitis and human glioblastoma multiforme. Acta Neuropathologica 2003;

105:586-592.

394. Standen GR, Bowen DJ. Factor-Xiii a(Bristol 1) - Detection of a Nonsense Mutation

(Arg(171)-]Stop Codon) in Factor-Xiii a Subunit Deficiency. British Journal of

Haematology 1993; 85:769-772.

395. Gorlach A, Berchner-Pfannschmidt U, Wotzlaw C, Cool RH, Fandrey J, Acker H,

Jungermann K, Kietzmann T. Reactive oxygen species modulate HIF-1 mediated PAI-1

expression: involvement of the GTPase Rac1. Thromb Haemost 2003; 89:926-935.

396. Kim J, Shao Y, Kim SY, Kim S, Song HK, Jeon JH, Suh HW, Chung JW, Yoon SR, Kim

YS, Choi I. Hypoxia-induced IL-18 increases hypoxia-inducible factor-1alpha expression

through a Rac1-dependent NF-kappaB pathway. Mol Biol Cell 2008; 19:433-444.

397. Lee HY, Lee T, Lee N, Yang EG, Lee C, Lee J, Moon EY, Ha J, Park H. Src activates HIF-

1alpha not through direct phosphorylation of HIF-1alpha specific prolyl-4 hydroxylase 2

but through activation of the NADPH oxidase/Rac pathway. Carcinogenesis 2011; 32:703-

712.

398. Xue Y, Bi F, Zhang X, Pan Y, Liu N, Zheng Y, Fan D. Inhibition of endothelial cell

proliferation by targeting Rac1 GTPase with small interference RNA in tumor cells.

Biochem Biophys Res Commun 2004; 320:1309-1315.

399. Zhang P, Zhang X, Hao X, Wang Y, Hui Y, Wang H, Hu D, Zhou J. Rac1 activates HIF-1

in retinal pigment epithelium cells under hypoxia. Graefes Arch Clin Exp Ophthalmol

2009; 247:633-639.

400. Hess AP, Schanz A, Baston-Buest DM, Hirchenhain J, Stoff-Khalili MA, Bielfeld P,

Kruessel JS. Expression of the vascular endothelial growth factor receptor neuropilin-1 in

the human endometrium. J Reprod Immunol 2009; 79:129-136.

401. Mayhew TM, Charnock-Jones DS, Kaufmann P. Aspects of human fetoplacental

vasculogenesis and angiogenesis. III. Changes in complicated pregnancies. Placenta 2004;

25:127-139.

402. Barut F, Barut A, Gun BD, Kandemir NO, Harma MI, Harma M, Aktunc E, Ozdamar SO.

Intrauterine growth restriction and placental angiogenesis. Diagn Pathol 2010; 5:24.

403. Chakraborty D, Rumi MA, Konno T, Soares MJ. Natural killer cells direct hemochorial

placentation by regulating hypoxia-inducible factor dependent trophoblast lineage

decisions. Proc Natl Acad Sci U S A 2011; 108:16295-16300.

Page 148: © 2013 Juanmahel Davila - IDEALS

133

404. Zhang J, Chen Z, Smith GN, Croy BA. Natural killer cell-triggered vascular

transformation: maternal care before birth? Cell Mol Immunol 2011; 8:1-11.

405. Roberts RM, Farin CE, Cross JC. Trophoblast proteins and maternal recognition of

pregnancy. Oxf Rev Reprod Biol 1990; 12:147-180.

406. King WA. Embryo-Mediated Pregnancy Failure in Cattle. Canadian Veterinary Journal-

Revue Veterinaire Canadienne 1991; 32:99-103.

407. Roberts RM, Cross JC, Leaman DW. Interferons as Hormones of Pregnancy. Endocrine

Reviews 1992; 13:432-452.

Page 149: © 2013 Juanmahel Davila - IDEALS

134

APPENDIXES

Page 150: © 2013 Juanmahel Davila - IDEALS

135

APPENDIX A: BREEDING STRATEGIES FOR THE FOUNDER AND EXPERIMENTAL

COLONY

FIGURE A.1: BREEDING STRATEGY FOR GENERATING THE RAC BREEDING COLONY. Transgenic mice

carrying the PgrCre knockin mutation were generously provided by Drs. Franco De Mayo and John Lyndon,

Baylor College of Medicine, TX. Mice carrying the knockin floxed mutation for Rac1 gene were generously

provided by Dr. Ann Sutherland, University of Georgia, GA. To establish the breeding colony, animals

from F1 were bred to each other. Only a select group of animals from the resulting F2 progeny (shown in

the figure above) were used to generate the experimental animal.

Page 151: © 2013 Juanmahel Davila - IDEALS

136

FIGURE A.2: EXPECTANCY FREQUENCIES OF ANIMALS/GENOTYPE BASED ON THE BREEDING

STRATEGY FOR GENERATING THE FOUNDING ANIMAL COLONY. The table shows the expected frequency

of animals in the F2 progeny, by genotype and sex, resulting from the mating of F1 male with F1 female.

Expectancy

Expectancy

by Sex

PgrCre/Cre

Rac1f/f 1/16 1/32

PgrCre/Cre

Rac1f/+ 1/8 1/16

PgrCre/+

Rac1f/f 1/8 1/16

PgrCre/+

Rac1f/+ 1/4 1/8

PgrCre/Cre

Rac1+/+ 1/16 1/32

PgrCre/+

Rac1+/+ 1/8 1/16

Pgr+/+

Rac1f/f 1/16 1/32

Pgr+/+

Rac1f/+ 1/8 1/16

Pgr+/+

Rac1+/+ 1/16 1/32

f

Cre/Cre

f/+

Cre/+

f/+

Cre/Cre

f/f

Cre/Cre

f/+

Cre/+

f/f

Cre/+

f/+Cre f

+

f

+

Cre Cre + +

Mal

e A

llel

es

PgrC

re/+

Rac1

f/+

Female Alleles

PgrCre/+

Rac1f/+

Pgr

Cre/+

+/+

+/+

f/+

+/+

+/+

Rac

Cre

+

+

Cre/Cre

+/+

Cre/+

f/+

Cre/+

+/+

Cre/+

f/f

Cre/+

f/+

+/+

f/f

+/+

f/+

+ f

Frequency

+

Page 152: © 2013 Juanmahel Davila - IDEALS

137

FIGURE A.3: BREEDING STRATEGY FOR GENERATING THE EXPERIMENTAL ANIMALS. The experimental

animals for the project animals were obtained by mating mice from the F2 progeny using the scheme above.

The tables on the next page show the expected frequency of animals, by genotype and sex, resulting from

any F2 breeding combinations.

Page 153: © 2013 Juanmahel Davila - IDEALS

138

FIGURE A.4: EXPECTANCY FREQUENCIES OF ANIMALS/GENOTYPE BASED ON THE BREEDING

STRATEGY FOR GENERATING THE BREEDING COLONY. The table shows the expected frequency of

experimental animals, by genotype and sex, resulting from the mating of F2 male with F2 female.

Expectancy

Expectancy

by Sex

PgrCre/+

Rac1f/f 1 1/2

Expectancy

Expectancy

by Sex

Pgr+/+

Rac1f/f 1 1/2

Expectancy

Expectancy

by Sex

PgrCre/+

Rac1f/f 1/2 1/4

Pgr+/+

Rac1f/f 1/2 1/4

Rac f f f f

Female Alleles

Pgr+/+

Rac1f/f

Pgr + + + +

Mal

e A

llel

es

Pg

rCre

/Cre

Ra

c1f/

f

Cre fCre/+

f/f

Cre/+

f/f

Cre/+

f/f

Cre fCre/+

f/f

Cre/+

f/f

Cre fCre/+

f/f

Cre/+

f/f

Cre/+

f/f

Cre/+

f/f

Cre/+

f/f

Cre fCre/+

f/f

Cre/+

f/f

Cre/+

f/f

Cre/+

f/f

Rac f f f f

Female Alleles

Pgr+/+

Rac1f/f

Pgr + + + +

Mal

e A

llel

es

Pg

r+/+

Ra

c1f/

f

+ f+/+

f/f

+/+

f/f

+/+

f/f

+ f+/+

f/f

+/+

f/f

+ f+/+

f/f

+/+

f/f

+/+

f/f

+/+

f/f

+/+

f/f

+ f+/+

f/f

+/+

f/f

+/+

f/f

+/+

f/f

Rac f f f f

Female Alleles

PgrCre/+

Rac1f/f

Pgr + + + +

Mal

e A

llel

es

Pg

rCre

/+ R

ac1

f/f

Cre fCre/+

f/f

Cre/+

f/f

Cre/+

f/f

+ f+/+

f/f

+/+

f/f

+ f+/+

f/f

+/+

f/f

+/+

f/f

+/+

f/f

Cre/+

f/f

Cre fCre/+

f/f

Cre/+

f/f

Cre/+

f/f

Cre/+

f/f

Frequency

Frequency

Frequency

+/+

f/f

+/+

f/f

+/+

f/f

+/+

f/f

Cre/+

f/f

Cre/+

f/f

Page 154: © 2013 Juanmahel Davila - IDEALS

139

Fertility Studies

FIGURE A.5: BREEDING STRATEGY FOR THE SIX-MONTH BREEDING STUDY. To rule out the possibility

that the subfertility in Rac1d/d mice is due to an inherent defect in the embryo, we bred the Rac1d/d and

Rac1f/f females to wild-type male mice of proven fertility. This breeding scheme generated pups that were

heterozygous for Rac1 gene. Therefore, any problems associated with pregnancy are likely due to a defect

in the maternal decidua, and not in the embryo or its membranes.

Page 155: © 2013 Juanmahel Davila - IDEALS

140

FIGURE A.6: EXPECTANCY FREQUENCIES OF ANIMALS/GENOTYPE BASED ON THE SIX-MONTH

BREEDING STUDY. Fertility testing was done using the breeding scheme above. The tables show the

expected frequency of animals, by genotype and sex, resulting from the breeding combinations.

Expectancy

Expectancy

by Sex

PgrCre/+

Rac1f/+ 1/2 1/4

Pgr+/+

Rac1f/+ 1/2 1/4

Expectancy

Expectancy

by Sex

Pgr+/+

Rac1f/+ 1 1/2

Rac f f f f

Female Alleles

PgrCre/+

Rac1f/f

Pgr Cre Cre + +

Mal

e A

llel

es

Pg

r+/+

Ra

c1+

/+

+ +Cre/+

f/+

Cre/+

f/+

+/+

f/+

+ +Cre/+

f/+

Cre/+

f/+

+ +Cre/+

f/+

Cre/+

f/+

+/+

f/+

+/+

f/+

+/+

f/+

+ +Cre/+

f/+

Cre/+

f/+

+/+

f/+

+/+

f/+

Rac f f f f

Female Alleles

Pgr+/+

Rac1f/f

Pgr + + + +

Mal

e A

llel

es

Pg

r+/+

Ra

c1+

/+

+ ++/+

f/+

+/+

f/+

+/+

f/+

+ ++/+

f/+

+/+

f/+

+ ++/+

f/+

+/+

f/+

+/+

f/+

+/+

f/+

+/+

f/+

+ ++/+

f/+

+/+

f/+

+/+

f/+

+/+

f/+

Frequency

Frequency

+/+

f/+

+/+

f/+

+/+

f/+

+/+

f/+

Page 156: © 2013 Juanmahel Davila - IDEALS

141

APPENDIX B: GENE EXPRESSION VALIDATION OF OTHERS MOLECULES INVOLVED IN STROMAL CELL

DECIDUALIZATION AND ANGIOGENESIS

FIGURE B.1: GENE EXPRESSION OF OTHER CRITICAL MOLECULES, IMPLICATED IN STROMAL CELL DIFFERENTIATION, WERE NOT AFFECTED

BY THE CONDITIONAL RAC1 MUTATION.

Pgr

Rac1f/f

Rac1d/d

0

1

2

3

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Gjp1/Cx43

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Mmp9

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Hif1

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Vegfa

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Tek/Tie2

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Vegfr1/Flt1

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Vegfr2/Flk1

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve

Gen

e E

xp

ress

ion

(2-

Ct )

Page 157: © 2013 Juanmahel Davila - IDEALS

142

FIGURE B.2: GENE EXPRESSION OF OTHER MOLECULES IMPLICATED IN UTERINE ANGIOGENESIS.

Page 158: © 2013 Juanmahel Davila - IDEALS

143

APPENDIX C: PRELIMINARY DATA FOR FUTURE DIRECTIONS

C1. Background information and the rationale

Embryo implantation into the maternal endometrium is crucial for embryo survival, and therefore

sustains pregnancy. It well establish that the invading embryo promotes well-orchestrated vascular

remodeling in the uterus to provide access to nutrient-rich maternal blood. Decidual cells are

known to actively regulate this process through complex crosstalk between the embryonic and

maternal tissues. In some pathological conditions associated with pregnancy, this crosstalk is

disrupted. In cases of shallow invasion of the trophoblast into the endometrium, the fetal stress

that arises causes hypertensive crisis condition in the mother, known as preeclampsia. In the US

alone, about 5% of pregnancies will develop preeclampsia and, so far, the only effective solution

is to deliver the baby, which often is premature. In contrast, in about 5% of pregnancies,

trophoblasts will migrate beyond their limits and placenta accreta will occur. In 70% of patients

with placenta accreta, trophoblast cells will only invade past the functionalis decidual layer (where

the trophoblast usually stops) into the basalis decidua. However, in more severe cases, placenta

increta or placenta percreta occurs, resulting in excessive invasion of the trophoblast deep into the

myometrium or into the peritoneum, respectively. In either case, surgical removal of the placenta

is necessary, which can put the mother and baby at risk. In both the instances, under- or over-

nutrition of the fetus can have long lasting effects on the conceptus’ metabolomics that can be

exacerbated into metabolic disturbances later in adulthood, like early onset diabetes, obesity, or

any other metabolic-related disease.

Early in vitro studies have suggested that RAC1 is present in the decidual cells and seems to play

a role in embryo invasion and placentation. However, our work has clearly showed that RAC1

activity is not necessary for embryo attachment and implantation into the endometrium (Chapter

3). However, uterine deletion of the Rac1 gene led to an apparent overgrowth of the trophoblasts

in the uterus of Rac1d/d females on day 8 of pregnancy (Figures 3.6 and C3.1). Therefore, we

decided to begin exploring how conditional deletion of Rac1 in the uterine tissues leads to

overgrowth of the trophoblast, and whether this overexpansion of the trophoblast leads to placental

abnormalities during mid-gestation. Because mice with conditional mutation for RAC1 have

Page 159: © 2013 Juanmahel Davila - IDEALS

144

severe angiogenic defects (Chapter 4), we only focused on those implantation nodules that were

as morphologically similar as possible to the control implantation nodules.

Page 160: © 2013 Juanmahel Davila - IDEALS

145

C2. Materials and Methods

Animals and Tissue Collection

All animal breeding, housing, procedures, experiments, and euthanasia were approved and

conducted according to the guidelines of the Institutional Animal Care and Use Committee of

University of Illinois at Urbana-Champaign. The health and care of the animals were provided by

the veterinarians and staff of Division of Animal Resources. Rac1 conditional null mice were

generated using Cre-LoxP strategy [347] (Figure 3.1A). Mice expressing homozygous copies of

the Rac1 gene flanked by loxP sites [Pgr+/+ Rac1f/f (official gene symbol Rac1tm1Djk/J), or Rac1f/f

from now onwards] [267] were obtained from Dr. Ann Sutherland (University of Georgia) in a

C57BL/6-129SV background. Mice carrying the Cre Recombinase under the control of the

progesterone receptor (Pgr) promoter [348] [PgrCre/+ Rac1+/+ (official gene symbol Pgrtm2(cre)Lyd/+),

or PgrCre from now onward], were obtained from Drs. John Lyndon and Francesco De Mayo

(Baylor College of Medicine) in a C57BL/6 background. To establish the mouse colony for this

study, initial breeding between the two mouse lines was done to obtain the F1 progeny, which are

heterozygous for Pgr and Rac1 (Appendix A). Female mice used for this study were generated by

breeding male mice of specific genotypes from the F2 progeny (see Figure 3.1B) with Rac1f/f

female mice. Genotyping of the animals was performed following the protocols established by the

respective laboratories that generated the transgenic mice [267, 348] (Appendix A). Conditional

knockout female mice (PgrCre/+ Rac1f/f) will be referred to as Rac1d/d from now onwards.

Rac1f/f or Rac1d/d females were euthanized by CO2 asphyxiation, followed by a blood draw via

cardiac puncture, and cervical dislocation. Uteri from pregnant mice were collected at different

time points during pregnancy. The harvested organs were either fixed for histological evaluation

[immersion-fixed in 10% neutral-buffered formalin (NBF)], flash frozen in liquid N2 for RNA or

frozen sectioning. For samples collected at different time points during pregnancy, identification

of a copulatory plug following mating was used as a reference and for our experimental purposes,

to indicate day 1 of pregnancy.

Page 161: © 2013 Juanmahel Davila - IDEALS

146

Some of the implantation nodules collected during pregnancy were used for morphological

evaluation of the decidua or placenta. The decidual mass was exposed by mechanical removal of

the myometrium. Pictures were taken using a Leica M165 FC dissection scope connected to a

Leica 450 C camera with c-mount interface containing a 5 Megapixel CCD sensor. Similarly, for

morphological evaluation of embryos on day 15 of pregnancy, mechanical removal of the

myometrium was done under the dissecting scope, as mentioned above. At least two implantation

nodules/female were analyzed on day 15 of pregnancy.

Quantitative real-time PCR and RNA Isolation

Total RNA was isolated from tissues or cells using TRIzol reagent (Invitrogen) followed by RNA

cleanup kit (Qiagen), according to the manufacturer’s instructions. RNA was converted to cDNA

using a kit (Applied Biosystems), according to the manufacturer’s instructions. Oligonucleotides

specific for genes of interest were developed (see Appendix F for details) and qPCR reactions were

carried out using SYBR-green master mix (Applied Biosystems) on a 7500 Applied Biosystems

real-time PCR machine (Applied Biosystems). Gene expression data analysis were done using the

2-ΔΔCt method [357] and 36b4 gene – which is conserved among many species and encodes an

acidic ribosomal phosphoprotein P0 that interacts with 40S ribosomal subunit – was used as a

housekeeping gene and loading control [358].

Immunohistochemistry, Immunocytochemistry, and Immunofluorescence

Cultured cells or paraffin-embedded uterine sections were subjected to immunohistochemistry or

immunocytochemistry as described previously [32, 353, 355, 356]. Briefly, paraffin-embedded

sections were deparaffinized in xylene and rehydrated in a series of graded alcohols followed by

rinsing in tap water. When frozen tissue were used, the sections were thawed at room temperature

for 5 min, then fixed for another 5 min in 10% NBF. When cells were used, they were fixed for

another 5 min in 10% NBF. Sections or cells then were rinsed in 1x phosphate-buffer saline (PBS),

followed by serum blocking using 10% non-immunized serum for 1 h at room temperature.

Following serum block, slides were incubated with a solution containing the diluted antibody (see

Appendix E for details) and 1% serum in 1x PBS overnight at 4 oC in a humidified chamber (for

Page 162: © 2013 Juanmahel Davila - IDEALS

147

slides) or in the culture plates (for cells). Following overnight incubation, slides were washed 3x

5 min with 1x PBS, then incubated with diluted fluorescent-labeled secondary antibody (see

Appendix E for details) for 1 h at room temperature. Samples then were rinsed in 1x PBS and

mounted with a medium containing DAPI. Pictures were taken using the Olympus BX51

microscope equipped for light and fluorescent imaging and was connected to a Jenoptik ProgRes

C14 digital camera with c-mount interface containing a 1.4 Megapixel CCD sensor. Fluorescent

images were merged and processed using CS4 Adobe Photoshop Extend (Adobe Systems).

Embryo and placental perimeter measurement

Java-based image analysis software NIH ImageJ (http://rsb.info.nih.gov/nih-image/) was utilized

for the outlining and making measurements in embryonic tissues. The perimeter of embryos on

day 8 of pregnancy, which were previously stained with hematoxylin and eosin, and the placenta

on day 15 of pregnancy were also measured using this software.

Statistical analysis

Experimental data were collected from a minimum of three independent animals (not from the

same litter), which were subjected to the same experimental conditions. Embryo weights and qPCR

results are expressed as LSMeans and mean ± S.E.M, respectively. Statistical analysis was done

using Student’s t-test. An analysis of equal variances was done on all numerical data to determine

whether a parametric or non-parametric hypothesis test was appropriate. Data were considered

statistically significant if p ≤ 0.05, and is indicated by an asterisks in the figures. All data were

analyzed and plotted using GraphPad Prism 4.0 (GraphPad Software) or Microsoft Excel 2013

(Microsoft Corporation).

Page 163: © 2013 Juanmahel Davila - IDEALS

148

C3. Preliminary data

FIGURE C3.1: CONDITIONAL DELETION OF RAC1 IN UTERINE DECIDUAL CELLS, DOES NOT PREVENT EMBRYO IMPLANTATION, BUT LEADS TO

TROPHOBLAST OVERGROWTH IN VIVO. (A) Implantation nodules collected on day 8 of pregnancy were serially sectioned and every 10th section

was stained with hematoxylin and eosin. The section with the maximum embryo size was used to measure the total perimeter covered by the

trophoblast (outlined by the dotted lines). The data show a close trend toward trophoblast outgrowth (p = 0.08).

Page 164: © 2013 Juanmahel Davila - IDEALS

149

FIGURE C3.1: CONDITIONAL DELETION OF RAC1 IN UTERINE DECIDUAL CELLS, DOES NOT PREVENT EMBRYO IMPLANTATION, BUT LEADS TO

TROPHOBLAST OVERGROWTH IN VIVO (CONTINUED). (B) Isolated embryos on day 8 of pregnancy, showed outgrowth of the trophoblast cells.

Bars = 1 mm. n ≥ 5.

Page 165: © 2013 Juanmahel Davila - IDEALS

150

FIGURE C3.2: GENE EXPRESSION PROFILE OF REGULATORS OF TROPHOBLAST EXPANSION. IGFBPs are known to interfere with embryo-

produced IGF2, which promotes forward migration of trophoblast cells into the maternal endometrium. Of the known IFGBPs that could be found

in the mouse uterus, IGFBP4 is only expressed by the uterine decidua [90, 92], while IGFBP3 can be expressed by both the decidua and the

myometrium. Adrenomedullin has been shown to be secreted by both the maternal tissues, as well as the embryonic tissues. Our data show that on

day 8 of pregnancy, only IGFBP4 is differentially regulated by the conditional deletion of RAC1 in the uterus. n = 4. Asterisks indicate significant

differences from controls (** p < 0.01).

Igfbp3

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve G

ene

Exp

ress

ion

(2-

Ct )

Igfbp4

Rac1f/f

Rac1d/d

0

1

2

3

**

Rel

ati

ve G

ene

Exp

ress

ion

(2-

Ct )

Igf2

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve G

ene

Exp

ress

ion

(2-

Ct )

Adm

Rac1f/f

Rac1d/d

0

1

2

Rel

ati

ve G

ene

Exp

ress

ion

(2-

Ct )

Page 166: © 2013 Juanmahel Davila - IDEALS

151

FIGURE C3.3: CONDITIONAL DELETION OF RAC1 IN UTERINE TISSUES LEADS TO PLACENTAL ABNORMALITIES AND INDICATIONS OF

INTRAUTERINE GROWTH RESTRICTION. (A and B) Samples collected on day 15 of pregnancy show a wide range of variation on the placental and

embryo outcome. However, consistently all the implantation nodules that were dissected from Rac1d/d females showed placental shape abnormalities

and wide-range in the level of hemorrhage that was observed in the placental tissues.

Page 167: © 2013 Juanmahel Davila - IDEALS

152

FIGURE C3.3: CONDITIONAL DELETION OF RAC1 IN UTERINE TISSUES LEADS TO PLACENTAL ABNORMALITIES AND INDICATIONS OF

INTRAUTERINE GROWTH RESTRICTION (CONTINUED). (C) Furthermore, some of the placentas had adopted a convex shape in the embryo side

of the placenta, instead of the normal concave shape. (D) Although these placentas did not extend inwards towards the uterus, the placentas obtained

from Rac1d/d females were expanded sideways and displayed abnormal shapes, as indicated by the increased placental perimeter. Similarly, these

placentas were significantly heavier than control counterparts, suggesting placental modifications, perhaps, as a result of uterine RAC1 mutation or

an adaptation to the hemorrhagic environment, which led to growth restrictions as observed in some of the more severe embryos. Bars = 1 cm. n =

3. Asterisks indicate significant differences from controls (* p < 0.05, ** p < 0.01).

Page 168: © 2013 Juanmahel Davila - IDEALS

153

FIGURE C3.4: CONDITIONAL DELETION OF RAC1 IN UTERINE TISSUES LEADS TO PLACENTAL ABNORMALITIES THAT DISPLAY IMPAIRED

VASCULAR REMODELING. (A) This section show the expected placental structures on day 12 of pregnancy. Bars: low power images = 500 µm and

high power images = 200 µm. White circles highlight blood vessels in the decidua. E = embryo, P = placenta, and M = mesometrial decidua. Area

1 = placenta (Red = CYTOKERATIN 8 positive cells); Area 2 = feto-maternal interphase; Area 3 = deep decidua and myometrium. All sections

shown here were stained with alpha smooth muscle actin (αSMA = green fluorescent stain) to assess the extent of vascular remodeling.

Page 169: © 2013 Juanmahel Davila - IDEALS

154

FIGURE C3.4: CONDITIONAL DELETION OF RAC1 IN UTERINE TISSUES LEADS TO PLACENTAL ABNORMALITIES THAT DISPLAY IMPAIRED

VASCULAR REMODELING (CONTINUED). (B and C) Samples collected on day 12 of pregnancy show a wide range of variation on the placental

outcome as shown here these implantation nodules from Rac1d/d females. Whereas one of the samples displays a placenta with normal histological

features as those seen in the controls (panel B), the lower most panel shows a disorganized, bifurcated (panel C, below), placenta (red fluorescent

stain = CYTOKERATIN 8). All sections shown here were stained with alpha smooth muscle actin (αSMA = green fluorescent stain) to assess the

extent of vascular remodeling. Our results show, that although there is some degree of vascular remodeling that can occur in Rac1d/d uteri, this seems

to be partial as there were some areas in the decidua that stained positive for αSMA, which were not positive in the control samples (areas 2 and 3).

Furthermore, it seems that in the severe cases, little vascular remodeling occurs, and whether this is a consequence of improper decidual function or

the fact that the embryo might be dying, remains to be elucidated. Bars: low power images = 500 µm and high power images = 200 µm. White

circles highlight blood vessels in the decidua. E = embryo, P = placenta, and M = mesometrial decidua. Area 1 = placenta (Red = CYTOKERATIN

8 positive cells); Area 2 = feto-maternal interphase; Area 3 = deep decidua and myometrium.

Page 170: © 2013 Juanmahel Davila - IDEALS

155

FIGURE C3.4: CONDITIONAL DELETION OF RAC1 IN UTERINE TISSUES LEADS TO PLACENTAL ABNORMALITIES THAT DISPLAY IMPAIRED

VASCULAR REMODELING (CONTINUED). (B and C) Samples collected on day 12 of pregnancy show a wide range of variation on the placental

outcome as shown here these implantation nodules from Rac1d/d females. Whereas one of the samples displays a placenta with normal histological

features as those seen in the controls (panel B), the lower most panel shows a disorganized, bifurcated (panel C), placenta (red fluorescent stain =

CYTOKERATIN 8). All sections shown here were stained with alpha smooth muscle actin (αSMA = green fluorescent stain) to assess the extent of

vascular remodeling. Our results show, that although there is some degree of vascular remodeling that can occur in Rac1d/d uteri, this seems to be

partial as there were some areas in the decidua that stained positive for αSMA, which were not positive in the control samples (areas 2 and 3).

Furthermore, it seems that in the severe cases, little vascular remodeling occurs, and whether this is a consequence of improper decidual function or

the fact that the embryo might be dying, remains to be elucidated. Bars: low power images = 500 µm and high power images = 200 µm. White

circles highlight blood vessels in the decidua. E = embryo, P = placenta, and M = mesometrial decidua. Area 1 = placenta (Red = CYTOKERATIN

8 positive cells); Area 2 = feto-maternal interphase; Area 3 = deep decidua and myometrium. n = 3

Page 171: © 2013 Juanmahel Davila - IDEALS

156

APPENDIX D: GENOTYPING PRIMERS AND PROTOCOL

PCR Cycling conditions for genotyping

Rac1 Genotyping

Rac1 Primers:

P91: 5'-GAT GCT TCT AGG GGT GAG CC-3'

P45: 5'-CAG AGC TCG AAT CCA GAA ACT AGT A-3'

P33: 5'-TCC AAT CTG TGC TGC CCA TC-3'

Cycling Program for Rac1:

1. 9 minutes @ 92 °C

2. 30 sec @ 92 °C

3. 30 sec @ 65 °C

4. 1 minute @ 72 °C

5. Cycle 35 times steps 2 to 4

6. 3 minutes @ 72 °C

7. ∞ @ 4 oC

8. End

This reaction condition gives 3 potential bands:

Rac1 Wild-type band 115 bp

Rac1 knockout band 140 bp

Rac1 floxed band 242 bp

Run in 2-3% Agarose Gel in TBE Buffer for at least 45 minutes @ 120 Vconstant

Page 172: © 2013 Juanmahel Davila - IDEALS

157

PgrCre Genotyping

PgrCre Primers:

R1: 5'-TAT ACC GAT CTC CCT GGA CG-3'

F1: 5'-ATG TTT AGC TGG CCC AAA TG-3'

F2: 5'-CCC AAA GAG ACA CCA GGA AG-3'

Cycling Program for PgrCre:

1. 5 minutes @ 94 °C

2. 1 minute @ 94 °C

3. 1 minute @ 60 °C

4. 2 minute @ 72 °C

5. Cycle 39 times steps 2 to 4

6. 7 minutes @ 72 °C

7. ∞ @ 4 oC

8. End

This reaction condition gives 2 bands:

Pgr Wild-type band 250 bp

PgrCre band 600 bp

Run in 1.5-2% Agarose Gel in TBE Buffer for at least 30 minutes @ 120 Vconstant

Page 173: © 2013 Juanmahel Davila - IDEALS

158

APPENDIX E: ANTIBODY DILUTIONS

Immunohistochemistry

For immunostaining, the blocking serum (donkey or goat) used depended on the secondary

Primary antibodies

Antibody Dilution used Company Catalog Host Species

Anti-Human MKI67 1:750 BD Parmigen 550609 Mouse

Anti-Mouse

PRL8A2/dPRP 1:750 Serum obtained

from Dr. Soares ----- Rabbit

Anti-Mouse PECAM1/

CD31 1:200 BD Parmigen 557355 Rat

Anti-Mouse α-SMA 1:200 Abcam Ab5694 Rabbit

Anti-Mouse Pan VEGFA

(A-20) 1:100 Santa Cruz

Biotechnology SC-152 Rabbit

Anti-Mouse

CYTOKERATIN 8

(Troma-I)

1:50 Hybridoma Bank ----- Rat

Anti-Mouse NRP1

(EPR3113) 1:200 Abcam Ab81321 Rabbit

Secondary antibodies

Biotinylated Anti-Rabbit

IgG 1:250 Invitrogen 95-6543B Goat

Biotinylated Anti-Mouse

IgG 1:250 Invitrogen 95-6143B Goat

Cy3-Conjugated Anti-Rat

IgG 1:250 Jackson

ImmunoResearch

712-165-

150 Donkey

Cy3-Conjugated Anti-

Rabbit IgG 1:250 Jackson

ImmunoResearch

711-165-

152 Donkey

Alexa 488-Conjugated

Anti-Mouse IgG 1:250 Jackson

ImmunoResearch

715-545-

151 Donkey

Fluorophores

Texas Red Avidin 1:500 Vector A-2006 -----

Fluorescein Avidin 1:500 Vector A-2001 -----

Page 174: © 2013 Juanmahel Davila - IDEALS

159

Western Blots

Antibody Dilution used Company Catalog Host Species

Anti-Mouse RAC1

(0.T.127) 1:1000 Abcam Ab33186 Mouse

Anti-β-Tubulin 1:2000 Sigma T5293 Mouse

HRP-Conjugated Anti-

Mouse IgG 1:5000 Sigma A9044 Rabbit

Page 175: © 2013 Juanmahel Davila - IDEALS

160

APPENDIX F: PRIMER SEQUENCES FOR GENE EXPRESSION ASSAYS

The primers for gene expression assays were generated using the Custom Primers - OligoPerfect™

Designer software (Invitrogen), based on the known mRNA sequence of the genes of interest. The

obtained oligonucleotide sequence were then submitted to NCBI BLAST tool to confirm their

specificity to the gene of interest. Sequences with 100% max match identity were used for gene

expression analysis. For all the primer sequences below, a single dissociation curve was obtained

during qPCR analysis.

Parameters used with the Invitrogen software:

Primer size 18 – 27 (min – max) ok, with opt (optimal) 20

Primer temp 57 – 63, with 60 opt

% GC 45 – 55, 50 opt

Product size 100 – 150 bp

Salt and primer concentration 50 nM

The 3’ nucleotide must be a perfect match to the target cDNA sequence

List of primers

Primers were purchased from either Sigma or Invitrogen.

36b4 Primers: F: 5’-CATCACCACGAAAATCTCCA-3’

R: 5’-TTGTCAAACACCTGCTGGAT-3’

Rac1 Primers: F: 5’-CTGAAGTGCGACACCACTGT-3’

R: 5’-CTTGAGTCCTCGCTGTGTGA-3’

Rac2 Primers: F: 5’-CAATGCAGGCCATCAAGTGT-3’

R: 5’-TGGGGATGTATTCTCCAGGG-3’

Rac3 Primers: F: 5’-ATCAAGTGCGTGGTGGTTG-3’

R: 5’-CGTTGGCTGAGTAGTTGTCG-3’

Cdc42 Primers: F: 5’-TGCTGCTATGAACGCATCTC-3’

R: 5’-CATGAGTGCATGTGGGTAGG-3’

Page 176: © 2013 Juanmahel Davila - IDEALS

161

RhoA Primers: F: 5’-TGGTGATGGAGCTTGTGGTA-3’

R: 5’-TCTACCTGCTTCCCATCCAC-3’

Hif1α Primers: F: 5’-TCTCCAAGCCCTCCAAGTATG-3’

R: 5’-CTACAGTGGTGGCAGTTGTG-3’

Epas1/Hif2α Primers: F: 5’-CAGGCAGTATGCCTGGCTAATTCCAGTT-3’

R: 5’-CTTCTTCCATCATCTGGGATCTGGGACT-3’

Cldn3 Primers: F: 5’-CCACTACCAGCAGTCGATGA-3’

R: 5’-AGCCTGTCTGTCCTCTTCCA-3’

F13a1 Primers: F: 5’-CAAGGATTCGGTGTGGAACT-3’

R: 5’-GCTTAACGGCTTGGACAGAG-3’

Wnt4 Primers: F: 5’-TTCTCACAGTCCTTTGTGGACG-3’

R: 5’-TCTGTATGTGGCTTGAACTGTG-3’

Wnt5a Primers: F: 5’-TCTTGGTGGTCTCTAGGTATG-3’

R: 5’-CACTGTGCTGCAGTTCCATCT-3’

Alpl Primers: F: 5’-TGGATGTGACCTCATTGC-3’

R: 5’-CATATAACACCAACGCTCAG-3’

Gpj1/Cx43 Primers: F: 5’- CTATCGTGGATCAGCGACCTTC-3’

R: 5’- CACGGGAACGAAATGAACACC-3’

Prl8a2/dPRP Primers: F: 5’-CTCACTTCTCAGGGGCACTC-3’

R: 5’-GGATCTGAGCAGCCATTCTC-3’

Runx1 Primers: F: 5’-AGATTCAACGACCTCAGGTTT-3’

R: 5’-CGGATTTGTAAAGACGGTGA-3’

Pgr Primers: F: 5’-CTAAATGAGCAGAGGATGAAGGAG-3’

R: 5’-TGGGCAACTGGGCAGCAATAAC-3’

Vegfa Primers: F: 5’-CAGGCTGCTGTAACGATGAA-3’

R: 5’-TTTCTTGCGCTTTCGTTTTT-3’

Tek/Tie2 Primers: F: 5’-TCCGAGCTAGAGTCAACACC-3’

R: 5’-GCCATCCACTATTGTCCAAG-3’

Vegfr1/Flt1 Primers: F: 5’-TGGCCAGAGGCATGGAGT-3’

R: 5’-TCGCAAATCTTCACCACATTG-3’

Vegfr2/Flk1 Primers: F: 5’-TCTGTGGTTCTGGTGGAGA-3’

R: 5’-GTATCATTTCCAACCACCCT-3’

Page 177: © 2013 Juanmahel Davila - IDEALS

162

Pecam1/Cd31 Primers: F: 5’-GAGGCCCAATCACGTTTCAGTT-3’

R: 5’-TCCTTCCTGCTTCTTGCTAGCT-3’

Angpt1 Primers: F: 5’-GGGGCTGGAAGGAGTATAAA-3’

R: 5’-CCTCAGCATGTACTGCCTCT-3’

Angpt2 Primers: F: 5’-TCCAAGAGCTCGGTTGCTAT-3’

R: 5’-AGTTGGGGAAGGTCAGTGTG-3’

Angpt4 Primers: F: 5’-AGCAGCCAACTGACGGAGTTT-3’

R: 5’-CTCTGCACAGTCCTGGAACA-3’

Sphk1 Primers: F: 5’-CATGCATGAGGTGGTGATG-3’

R: 5’-TGCTCGGTACCCAGCATAGTG-3’

Nrp1 Primers: F: 5’-TGTGGGTACACTGAGGGTCA-3’

R: 5’-CAGCAATTCCACCAAGGTTT-3’

Mmp9 Primers: F: 5’-TCTTCCCCTTCGTCTTCCT-3’

R: 5’-TATACAGCGGGTACATGAGCG-3’

Adm Primers: F: 5’-GGGTTCACTCGCTTTCCTAGGGTTCACTCGCTTTTCTA-3’

R: 5’-GCTGCTGGATGCTTGTAGTTGCTGGCTGGATGCTTGTAGTT-3’

Igf2 Primers: F: 5’-ATGACACCTGGAGACAGTCC-3’

R: 5’-TGGCCTCTCTGAACTCTTTG-3’

Igfbp3 Primers: F: 5’-GGTCCTTATTGTTGCCATCTC-3’

R: 5’-AGATGCCTGACAAAGAAAGG-3’

Igfbp4 Primers: F: 5’-TGCAGACCTCTGACAAGGAT-3’

R: 5’-GTCCCCACGATCTTCATCCTT-3’

Prl3c1/Plpj Primers: F: 5’-GTTTTTGATTTTGCCATGCT-3’

R: 5’-TCGCAAATCTTCACCACATTG-3’