+ All Categories
Home > Documents > © Maria B. Padua - University of...

© Maria B. Padua - University of...

Date post: 08-Apr-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
156
1 EVOLUTION AND CELLULAR BIOLOGY OF THE UTERINE SERPINS By MARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2009
Transcript
Page 1: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

1

EVOLUTION AND CELLULAR BIOLOGY OF THE UTERINE SERPINS

By

MARIA B. PADUA

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL

OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF

DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2009

Page 2: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

2

© Maria B. Padua

Page 3: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

3

I dedicate this work to my mother Carmen, uncle Adán, nanny Zenaida

and siblings Carolina and Fernando

Page 4: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

4

ACKNOWLEDGMENTS

I would like to thank my advisor Dr. Peter J. Hansen for his guidance, support and

challenge through my PhD program. I am grateful to him for encouraging me as a scientist

during all these years. Also, I would like to thank my committee members Dr. Nasser Chegini,

Dr. David Julian, Dr. Phyllip LuValle, Dr. Karen Moore for their contributions and suggestions

to improve my research projects and academic training. I am also grateful to Dr. William

Thatcher for his continuous support and participation.

I also thank my lab mates, Luciano and Aline Bonilla, Barbara Loureiro, Lilian Oliveira,

Justin Fear, Silvia Carambula and Jim Moss and my old lab mates, Dr. Jeremy Block, Dr. Dean

Jousan, Moisés Franco, Dr. Luiz Augusto de Castro and Amber Brad for their great help through

many different ways. Likewise, I want to thank my international friends Dr. Yaser Al-Katanani,

Dr. Fabiola Paula-Lopes, Dr. Zvi Roth, Dr. Paolete Soto, and Dr. Olga Ocon for their friendship

and additional contributions.

Thanks are also extended to all personnel in the Department of Animal Sciences,

especially to Rick Wenzel for making the cell culture room a more functional place to work.

Likewise, I want to thank the International Student Center staff, especially to Debra Anderson

and Maud Fraser for their kindness and assistance to all international students. I am also grateful

to Neal Bensen and Steve McClellan from the Flow Cytometry Core Laboratory, Dr. Gigi

Ostrow from the Gene Expression Core Laboratory, Dr. Savita Shanker and Xiao Hui Zhou from

the DNA Sequencing Core Laboratory and to the University of Florida Diagnostic Referral

Laboratories of the Interdisciplinary Center for Biotechnology Research-Cancer Genetics

Research Complex at the University of Florida for their assistance to perform many of the

experiments presented in this dissertation.

Page 5: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

5

Gratitude is also extended to the following University of Florida people for providing

access to laboratory equipment: Dr. Owen Rae and Shelley Lanhart from the Department of

Large Animal Clinical Sciences, Dr. Pushpa Kalra from the Deparment of Physiology and

Functional Genomics, Dr. Lori Warren, Jan Kivilpeto, Dr. Lokenga Badinga and Dr. Alan Ealy

from the Department of Animal Sciences, University of Florida.

Likewise, I am grateful to the following people for their invaluable assistance in providing

tissue samples for some of the experiments presented in this dissertation: Dr. Dan C. Sharp, Dr.

Luciano Silva and Dr. Claudia Klein from the Animal Sciences Department, University of

Florida, Dr. Ellis Greiner from the college of Veterinary Medicine, University of Florida and

Mirian Y. Cañas from the Laboratorio de Embriologia and Endocrinologia Molecular, Decanato

de Agronomia, Universidad Centroccidental Lisandro Alvarado, Venezuela, Dr. Douglas C.

Antczak and Christina Costa from the Baker Institute for Animal Health, College of Veterinary

Medicine, Cornell University, Dr. John Verstegen from the College of Veterinary Medicine,

University of Florida, Dr. Fernanda Agreste from the Surgery Department, Faculdade de

Medicina Veterinária e Zootecnia, Universidade de São Paulo, Brazil and Dr. Stephen Shores

and Debi Gibson from the Animal Shores Hospital in Gainesville.

I am also grateful to Dean Leed for his care and support. Special thanks go to Dr. Andrés

Kowalski, Mónica Prado-Cooper, José Cristobal Nieto, Marília and José Trujillo, Armando Luiz

and Claudia García for their sincere friendship for many years. Also, I am grateful to my very

good friends in Gainesville Domenicchella Dean, Cristina Caldari-Torres and Milerky Perdomo

for their friendship and support in many different ways. I really appreciate it.

Page 6: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

6

TABLE OF CONTENTS

page

ACKNOWLEDGMENTS.................................................................................................................... 4

LIST OF TABLES................................................................................................................................ 9

LIST OF FIGURES ............................................................................................................................ 10

LIST OF ABBREVIATIONS ............................................................................................................ 12

ABSTRACT ........................................................................................................................................ 14

CHAPTER

1 LITERATURE REVIEW ........................................................................................................... 16

Introduction ................................................................................................................................. 16

Serine Proteinase Inhibitor Superfamily .................................................................................... 17

Mechanism of Action .......................................................................................................... 17

Non-Inhibitory Serpins ........................................................................................................ 20

Functions of Inhibitory Serpins Distinct from Proteinase Inhibition ............................... 21

Serpinopathies ...................................................................................................................... 22

Receptor Mediated Internalization of the Serpin-Proteinase Complex ............................ 23

Evolution of Serpins and Their Functions ......................................................................... 26

Phylogeny of the Serpin Superfamily................................................................................. 31

Positive Selection in Serpins ............................................................................................... 33

Novel Serpins and their Functions ...................................................................................... 35

Uterine Serpins ............................................................................................................................ 37

Ovine Uterine Serpin ........................................................................................................... 39

Endometrial secretion. ................................................................................................. 40

Biological function. ...................................................................................................... 41

Porcine Uterine Serpin ........................................................................................................ 44

Endometrial secretion. ................................................................................................. 44

Biological function. ...................................................................................................... 45

Bovine Uterine Serpin ......................................................................................................... 46

Endometrial secretion. ................................................................................................. 47

Biological function. ...................................................................................................... 47

Caprine Uterine Serpin ........................................................................................................ 48

Evolution and Phylogeny of Uterine Serpins ..................................................................... 48

Synopsis and Objectives...................................................................................................... 50

2 MOLECULAR PHYLOGENY OF UTERINE SERPINS AND ITS RELATIONSHIP

TO EVOLUTION OF PLACENTATION ................................................................................ 51

Introduction ................................................................................................................................. 51

Materials and Methods ................................................................................................................ 53

Page 7: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

7

Data Base Queries to Identify Uterine Serpin Genes ........................................................ 53

Reverse Transcription-Polymerase Chain Reaction (RT-PCR)........................................ 53

Identification of cDNA for Equine Uterine Serpin Gene .................................................. 54

Sequencing of Amplicons ................................................................................................... 56

Detection of Equine Uterine Serpin by Western Blotting ................................................. 57

Amino Acid Sequence Alignments and Analysis of Phylogenetic Tree .......................... 57

Results .......................................................................................................................................... 59

Identification of Coding Sequences of Known and New Uterine Serpins Using

Blastn ................................................................................................................................ 59

Uterine Serpin Gene Organization in the Bovine and Canine .......................................... 62

Characteristics of CanUS and Expression in Tissues ........................................................ 62

Uterine Expression and Amino Acid Sequence of EqUS ................................................. 62

Endometrial Secretion of EqUS .......................................................................................... 66

Lack of Expression of the Uterine Serpin Gene in the Pregnant Cat ............................... 67

Amino Acid Sequence Conservation.................................................................................. 67

Identification of the Putative Hinge Region and P1-P1’ Site of the RCL........................ 70

Phylogenetic Analysis of Uterine Serpins.......................................................................... 72

Positive Selection of the Uterine Serpin Gene ................................................................... 72

Discussion .................................................................................................................................... 78

3 COMPARISON OF THE NATIVE AND RECOMBINANT FORMS OF OVINE

UTERINE SERPIN FOR INHIBITION OF CELL PROLIFERATION ................................ 85

Introduction ................................................................................................................................. 85

Materials and Methods ................................................................................................................ 86

Materials ............................................................................................................................... 86

Collection of Uterine Fluid and Purification of Native OvUS ......................................... 87

[3H]thymidine Incorporation by D17 and PC-3 Cells ....................................................... 87

Induction of Apoptosis in D17 and PC-3 Cells ................................................................. 88

Purification of His-Tagged rOvUS from Conditioned Medium ....................................... 89

Expression and Purification of β-Galactosidase ................................................................ 90

Proliferation of P388D1 and PC-3 Cells ............................................................................ 90

Statistical Analysis............................................................................................................... 91

Results .......................................................................................................................................... 92

Inhibition of Proliferation and Induction of Apoptosis in D17 and PC-3 Cells .............. 92

Antiproliferative Actions on P388D1 and PC-3 Cell Lines: Comparison of the

Native and Recombinant Forms of OvUS ...................................................................... 92

Discussion .................................................................................................................................... 95

4 REGULATION OF DNA SYNTHESIS AND THE CELL CYCLE IN HUMAN

PROSTATE CANCER CELLS AND LYMPHOCYTES BY OVINE UTERINE

SERPIN ........................................................................................................................................ 97

Introduction ................................................................................................................................. 97

Materials and Methods ................................................................................................................ 99

Materials ............................................................................................................................... 99

Purification of rOvUS.......................................................................................................... 99

Page 8: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

8

PC-3 Cell Culture .............................................................................................................. 100

[3H]thymidine Incorporation by PC-3 Cells .................................................................... 101

Cell Proliferation Based on ATP Content ........................................................................ 101

Cytotoxicity Assay............................................................................................................. 102

TUNEL Labeling ............................................................................................................... 102

Secretion of IL-8 ................................................................................................................ 103

Cell Cycle Analysis ........................................................................................................... 104

Statistical Analysis............................................................................................................. 105

Results and Discussion ............................................................................................................. 105

Proliferation of PC-3 cells ................................................................................................. 105

Lactate Rehydrogenase Release........................................................................................ 107

DNA Fragmentation (Apoptosis) ..................................................................................... 107

Interleukin-8 Secretion ...................................................................................................... 107

Cell Cycle Dynamics ......................................................................................................... 112

5 CHANGES IN EXPRESSION OF CELL-CYCLE RELATED GENES IN PC-3

PROSTATE CANCER CELLS CAUSED BY OVINE UTERINE SERPIN ...................... 118

Introduction ............................................................................................................................... 118

Materials and Methods .............................................................................................................. 119

Materials ............................................................................................................................. 119

Purification of rOvUS........................................................................................................ 120

PC-3 Cell Culture .............................................................................................................. 120

Proliferation Assay ............................................................................................................ 121

Cell Culture for RNA Extraction ...................................................................................... 121

RNA Extraction ................................................................................................................. 121

cDNA Synthesis and Real Time-PCR Array ................................................................... 122

Statistical Analysis............................................................................................................. 122

Results ........................................................................................................................................ 124

Inhibition of PC-3 Cell Proliferation by OvUS ............................................................... 124

Cell-cycle Related Gene Expression Profile at 12 h after Treatment with rOvUS ....... 124

Cell-cycle Related Gene Expression Profile at 24 h after Treatment with rOvUS ....... 127

Discussion .................................................................................................................................. 127

6 GENERAL DISCUSSION ....................................................................................................... 134

LIST OF REFERENCES ................................................................................................................. 141

BIOGRAPHICAL SKETCH ........................................................................................................... 156

Page 9: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

9

LIST OF TABLES

Table page

2-1 Primers used in RT-PCR procedure to obtain the full length coding sequence of the

equine uterine serpin gene. .................................................................................................... 55

2-2 Exon and intron sizes for the uterine serpin gene of the cow and dog. .............................. 63

2-3 Ratios (ω) of nonsynonymus (dN) to synonymous (dS) mutations, parameter

estimates and maximum log likelihood of models for positive selection within the

protein coding sequence of uterine serpins........................................................................... 74

2-4 Test of significance for models for positive selection within the protein coding

sequence of uterine serpins. ................................................................................................... 75

5-1 Cell cycle related genes screened using the RT2 Profiler

TM PCR Array. ......................... 123

5-2 Regulation of cell-cycle related genes of PC-3 cells after 12 h of treatment with 200

µg/ml recombinant ovine uterine serpin. ............................................................................ 126

5-3 Down-regulation of human cell cycle-related genes of PC-3 cells after 24 h of

treatment with 200 µg/ml recombinant ovine uterine serpin. ........................................... 128

Page 10: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

10

LIST OF FIGURES

Figure page

1-1 Structure of the inhibitory serpin α1-antitrypsin. A) Native-stressed conformation of

the protein. .............................................................................................................................. 18

1-2 Neighbor-joining tree of 110 serpin sequences. ................................................................... 34

2-1 Identification of an incorrectly annotated dog corticosteroid binding globulin (CBG)

as an uterine serpin (US)........................................................................................................ 61

2-2 Representative electrophoretogram of amplicons obtained by reverse transcriptase-

polymerase chain reaction (RT-PCR) of RNA fron canine tissue and canine uterine

serpin primers. ........................................................................................................................ 64

2-3 Expression and secretion of equine uterine serpin ............................................................... 65

2-4 Amino acid sequence alignment of the uterine serpins using the ClustalW algorithm. .... 68

2-5 Identification of the P1-P1’ site and hinge region (underlined) in uterine serpins. ........... 71

2-6 Phylogenetic tree of the uterine serpin proteins with the ovine uterine serpin (OvUS)

as out-group. ........................................................................................................................... 73

2-7 Selecton output generated for the uterine serpin group of proteins. ................................... 76

2-8 Amino acid sequence alignment of ovine uterine serpin (NP_001009304.1) and

human α1-antitrypsin (NP_000286.3) using the ClustalW algorithm ................................ 77

2-9 Phylogenetic tree of placentation in mammals (adapted from Vogel 2005) to

illustrate the existence of uterine serpin genes relative to type of placentation. ................ 79

3-1 Effect of OvUS on induction of apoptosis in D17 and PC-3 cells as determined by

TUNEL labeling. .................................................................................................................... 93

3-2 Inhibition of [3H]thymidine incorporation of P388D1 cells and PC-3 cells by native

(n) and recombinant (r) OvUS............................................................................................... 94

4-1 Inhibition of [3H]thymidine incorporation of PC-3 cells by recombinant ovine uterine

serpin (rOvUS) ..................................................................................................................... 106

4-2 Inhibition of proliferation of PC-3 cells by recombinant ovine uterine serpin (rOvUS)

as determined by ATP content/well. ................................................................................... 108

4-3 Lack of cytotoxic effect of recombinant ovine uterine serpin (rOvUS) on PC-3 cells

was measured by the release of lactate dehydrogenase. .................................................... 109

Page 11: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

11

4-4 Representative photomicrographs of PC-3 cells labeled using the TUNEL procedure

after 48 h of culture with either 100 (A) or 200 µg/ml (B) of rOvUS or 200 μg/ml of

the control protein ovalbumin. ............................................................................................ 110

4-5 Effect of recombinant ovine uterine serpin (rOvUS) on DNA fragmentation

(apoptosis) of PC-3 cells. ..................................................................................................... 111

4-6 Effect of recombinant ovine uterine serpin (rOvUS) on interleukin (IL) – 8

concentration in cell culture supernatants of PC-3 cells. ................................................... 113

4-7 Cell cycle dynamics of PC-3 cells as affected by recombinant ovine uterine serpin

(rOvUS). ............................................................................................................................... 114

4-8 Cell cycle dynamics of lymphocytes as affected by recombinant ovine uterine serpin

(rOvUS). ............................................................................................................................... 116

5-1 Inhibition of [3H]thymidine incorporation of PC-3 cells by 200 μg/ml of the

recombinant ovine uterine serpin (rOvUS) ........................................................................ 125

5-2 Points in the cell cycle where genes were differentially regulated by ovine uterine

serpin at 12 and 24 h are represented in panel A and B, respectively. ............................. 130

6-1 Possible pathways by which OvUS could block cell proliferation. .................................. 138

Page 12: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

12

LIST OF ABBREVIATIONS

Bo Bovine

Cap Caprine

CBG Corticosteroid Binding Globulin

CD Cluster of Differentiation

CDK Cyclin-Dependent Kinase

Con A Concanavalin A

DPBS Dulbecco’s Phosphate Buffered Saline

G-CSF Granulocyte Macrophage-Colony Stimulating Factor

HSP-47 Heat Shock Protein-47

IFN Interferon

IL Interleukin

LPS Lipopolysaccharide

LRP1 Low Density Lipoprotein Receptor Related Protein 1

Maspin Mammary Serine Proteinase Inhibitor

MENT Myeloid and Erythroid Nuclear Termination Stage-specific Protein

Mya Million Years Ago

NK Natural Killer

OVA Ovalbumin

Ov Ovine

PAI Plaminogen Activator Inhibitor

PC-3 Human Prostate Cancer Cells-3

PEDF Pigment Epithelium Derived Factor

Page 13: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

13

PHA Phytohemagglutinin

PK Protein Kinase

PolyI•PolyC Polyinosinic-Polycytidylic Acid

Po Porcine

PWM Pokeweed Mitogen

RAP Receptor Associated Protein

Rb Retinoblastoma protein

RCL Reactive Center Loop

RT-PCR Reverse Transcription-Polymerase Chain Reaction

rOvUS Recombinant Ovine Uterine Serpin

SERPIN Serine Proteinase Inhibitor

STZ Streptozotocin

TNF-α Tumor Necrosis Factor-α

TUNEL Terminal Deoxynucleotidyl Transferase (TdT) and fluorescein

isothiocyanate-conjugated dUTP nick end labeling

US Uterine Serpin

Page 14: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

14

Abstract of Dissertation Presented to the Graduate School

of the University of Florida in Partial Fulfillment of the

Requirements for the Degree of Doctor of Philosophy

EVOLUTION AND CELLULAR BIOLOGY OF THE UTERINE SERPINS

By

Maria B. Padua

May 2009

Chair: Peter J. Hansen

Major: Animal Molecular and Cellular Biology

Uterine serpins (US) are a unique group of progesterone-induced glycoproteins in a

restricted group of mammals that belong to the serine proteinase inhibitor superfamily and which

are secreted in large quantities into the uterus during pregnancy. The US gene has been

identified in species with epitheliochorial placentation of the Ruminantia and Suidae orders of

the Laurasiatheria superorder of eutherian mammals.

One goal of this dissertation was to examine the evolution of the US gene in mammals. A

US gene was identified in horses and dogs and found expressed in the uterus during pregnancy.

The dog is a species with endotheliochorial placenta, suggesting that the US gene is not restricted

to species with epitheliochorial placentation. However, its absence in other mammals, and

apparent loss in the cat, suggests that the US gene evolved only within the Laurasiatheria

superorder.

The US do not appear to be functional proteinase inhibitors and to have species-specific

functions. The most studied member of the group, ovine uterine serpin (OvUS), inhibits

proliferation of several cell types including activated lymphocytes, bovine pre-implantation

embryos and some tumor cell lines. A second goal was to evaluate the mechanism by which

OvUS inhibits cell proliferation. Ovine US blocked cell cycle progression of human prostate

Page 15: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

15

cancer (PC-3) cells, causing an accumulation of cells at G_2/M at 12 h and at G_0/G_1 at 24 h

after treatment addition. Additionally, OvUS blocked the cell cycle progression in

phytohemagglutinin-stimulated lymphocytes increasing the number of cells at the G_0/G_1 stage

at 96 h after treatment. Blocking of the cell-cycle progression by OvUS was caused specifically

by the up-regulation of cell cycle checkpoint and arrest genes such as CDKN1A (p21), CDKN2B

(p15) and CCNG2 (cyclin G2), and down-regulation of genes involved in DNA synthesis and

cell cycle regulation and progression.

The finding that the US gene is only retained in a limited group of mammals suggests its

importance for successful pregnancies in these species. It is also possible that the US gene

evolved a distinct and specie-dependent function from an ortholog serpin gene, rather than the

typical anti-proteolytic activity conserved in most members of the superfamily.

Page 16: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

16

CHAPTER 1

LITERATURE REVIEW

Introduction

Pregnancy in mammals has evolved in a process that included the appearance of new genes

for novel functions. Many of these new genes arose by gene duplication to generate paralogs

which underwent sequence divergence from the parental genes (Louis 2007). Recently, it was

determined specifically in the mouse placenta that most of the genes present during early events

of placentation, such as those involved in basic growth and metabolism, are evolutionary ancient

genes and their ortholog genes are also found within eukaryotes (Knox & Baker 2008). Similar

results were found in the human decidua where ortholog genes are also present in all vertebrates

(Knox & Baker 2008). In contrast, those genes expressed in the mature placenta are rodent

specific genes, which suggest that these genes are newly evolved since no orthologs were found

in other eukaryote or vertebrates (Knox & Baker 2008). Some examples of genes involved in

reproductive processes that were formed as a result of gene duplication are the primate chorionic

gonadotropins (Maston & Ruvolo 2002) and interferon-τ (Roberts et al. 1999). The uterine

serpins (US) present in the uterus of a limited group of mammals are another example. In sheep,

cattle, and pigs an ancestral gene or genes of the serine proteinase inhibitor (serpin) superfamily

underwent modification to exhibit high expression in the uterus and under the regulation of

progesterone. Unlike most of the members of the serpin superfamily, US probably have not

retained the anti-proteolytic activity that defines this superfamily (Irving et al. 2000). Instead,

evidence suggests they have divergent functions as for example, regulation of the immune

function in the sheep (Hansen 1998) and placental iron transport in the pig (Roberts & Bazer

1988). The purpose of this dissertation is to more closely examine the evolution of the US gene

Page 17: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

17

in mammals and to determine the mechanisms by which the US produced by the sheep inhibits

cell proliferation.

Serine Proteinase Inhibitor Superfamily

The serpins (serine proteinase inhibitors) are an important superfamily of proteins presents

in at least some members of all phyla. In vertebrates, serpins are involved in the regulation of

proteolytic pathways including blood coagulation and fibrinolysis (α1-antitrypsin, antithrombin),

complement cascade (complement C1-inhibitor) and tissue remodeling (plasminogen activator

inhibitor-1 and 2) (Irving et al. 2000). Although the inhibitory activity of serpins is mostly

towards serine proteinases, serpins can also function as inhibitors of proteinases that belong to

other families such as cysteine proteinases and papain-like cysteine proteinases (Silverman et al.

2001, Irving et al. 2002a, Hungtinton 2006).

Mechanism of Action

Inhibitory serpins inactivate their target proteinases through a unique suicide substrate-like

inhibitory mechanism. The structural conformation of serpins is typically formed by nine α-

helices (A-I) and 3 β-sheets (A-C) (Figure 1-1). The key region for inhibitory serpins is the

reactive center loop (RCL), which is a flexible structure localized on the top of the serpin and

contains a complementary sequence to the active site of the target proteinase. The RCL is

usually formed by 20-25 amino acids and the hinge region is enclosed in the P15-P9 portion of it

(Irving et al. 2000, Hungtinton 2006, Whisstock and Bottomley 2006). The hinge region

provides mobility to the RCL. There is a consensus pattern present in the sequence for inhibitory

serpins. Arginine is conserved at the P17 position, arginine, lysine or glutamic acid at P16,

glycine is usually present at P15 , threonine or serine is present at P14 position and acid residues

with short side chains such as alanine, glycine or serine are abundant in positions P12 to P9

(Irving et al. 2000).

Page 18: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

18

Figure 1-1. Structure of the inhibitory serpin α1-antitrypsin. A) Native-stressed conformation

of the protein. The helices are represented by blue cylinders and the A, B and C β-

sheets are shown in green, red and yellow respectively. The reactive center loop

(RCL) is shown in pink (1) with the hinge region (2). Also shown is the breach (3),

gate (4) and shutter (5). B) Cleaved-relaxed conformation of the protein. The

cleaved RCL is shown in 6 and the newly formed β-sheet A in 7. Reproduced from

Irving et al. (2000) with permission of Genome Research (© 2000) and after slight

modification.

3

1

4

2

5

6

7

A B

Page 19: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

19

The proteinase binds covalently to RCL and cleaves the scissile bond of the serpin at the

P1-P1’ site, releasing the P1’ residue and forming an ester bond between the P1 residue of the

serpin and the proteinase catalytic serine. Then, the amino-terminal portion of the RCL with the

attached proteinase moves as much as 70 Å to the opposite side to form the β-sheet A, and the

compression of the proteinase against the base of the serpin causes a distortion in the structure of

the proteinase causing its inactivation (Figure 1-1) (Irving et al. 2000, Silverman et al. 2001,

Hungtinton 2006). This conformational change in the structure, from the stressed (non-cleaved

RCL) to the relaxed (cleaved RCL) form, increases the overall stability of the serpin. This

characteristic makes serpins highly unusual. Most proteins are in a thermodynamically favorable

conformation in their native state. In contrast, serpins in their native state (i.e. the stress

conformation) require lower temperatures for denaturation (Silverman et al. 2001, Im et al. 2002,

Hungtinton 2006, Whisstock & Bottomley 2006).

There are some other important regions in the structure of the serpins besides the hinge

region of the RCL. One of them is the breach which is localized at the upper area of the β-sheet

A where the initial insertion of the RCL into the β-sheet A occurs (Figure 1-1) (Irving et al.

2000). There is also the shutter that is located close to the center of the β-sheet A. Together with

the breach, the shutter helps open the sheets to allow the insertion of the hinge region of the RCL

(Irving et al. 2000). Finally, the gate formed by strands s3C and s4C is region where the RCL

bypass around these strands to be completely inserted into the β-sheet A without cleavage (Irving

et al. 2000).

Some serpins are relatively inactive proteinase inhibitors, but they become fully activated

as inhibitors by the binding of cofactors such as heparin and vitronectin (Jin et al. 1997, Schvartz

et al. 1999, Zhou et al. 2003). This is the case of antithrombin, heparin cofactor II, plasminogen

Page 20: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

20

activator inhibitor-1, protease nexin I and protein C inhibitor, where the RCL of these serpins is

partially inserted into the top of the β-sheet A, making the RCL and the P1 residue less

accessible for the proteinase. Once the cofactor interacts with the serpin, there is a

conformational change, where the RCL is expelled and the P1 residue becomes accessible for the

target proteinase (Law et al. 2006, Whisstock & Bottomley 2006). Likewise, heparin can

enhance the inhibitory function of already-active inhibitory serpins. A set of experiments

conducted by Gupta & Gowda (2008) showed that heparin can bind to α1-antitrypsin and thereby

increase the inhibitory potential of this serpin.

Non-Inhibitory Serpins

There is a group of serpins that do not inhibit proteinases. Examples of this growing group

includes ovalbumin (OVA), the chicken-egg storage protein (Benarafa & Remold-O’Donnell

2005), mammary serine protease inhibitor (maspin), which inhibits angiogenesis and tumor

growth and increases the sensitivity of cancer cells to undergo apoptosis (Zhang 2000, Sheng

2006), the hormone transport proteins corticosteroid and thyroxine binding globulin (Pemberton

et al. 1988), angiotensinogen, involved in blood pressure regulation, water and salt homeostasis

(Morgan et al. 1996, Stanley et al. 2006), the chaperone heat shock protein (HSP)-47 (Nagata

1998, Sauk et al. 2005) and the multifunctional pigment epithelium derived factor (PEDF),

which promotes differentiation of neurons and retinal photoreceptors, and neuronal degeneration

while inhibiting angiogenic processes in the retina, antagonizing the angiogenic effects of

vascular endothelial growth factor, platelet-derived growth factor and interleukin (IL)-8, and

promoting apoptosis in endothelial cells (Irving et al. 2000, Silverman et al. 2001, Tombran-

Tink & Barnstable 2003).

Page 21: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

21

Functions of Inhibitory Serpins Distinct from Proteinase Inhibition

Serpins are versatile proteins and can function in processes through mechanisms not

involving proteinase inhibition. A well studied example is the inhibitory serpin α1-antitrypsin,

which is produced by hepatocytes, neutrophils, macrophages, monocytes, intestinal epithelial

cells, astrocytes and some other cells to inhibit the enzymatic activity of different proteinases

such as neutrophil elastase, cathepsin G, proteinase 3, chymotrypsin, trypsin, thrombin (Travis &

Salvesen 1983, Congote 2007). The expression of α1-antitrypsin is increased by the pro-

inflammatory molecules IL-1, IL-6, tumor necrosis factor (TNF)-α, lipopolysaccharide (LPS)

(Congote 2007). Thus, it is assumed that the physiological role of this serpin is to prevent host

tissue from the proteolytic damage caused by the activity of these proteases.

In addition, α1-antitrypsin has functions that are unrelated to its anti-proteolytic activity.

Breit et al. (1983) demonstrated that purified α1-antitrypsin inhibited in a dose-dependent

manner the proliferative response of human peripheral blood lymphocytes induced by the

mitogens concanavalin A (Con A) and phytohemagglutin (PHA). However, α1-antitrypsin did

not completely inhibit these proliferative responses. Moreover, this serpin did not cause any

inhibitory activity against lymphocytes activated by the T and B cell mitogen pokeweed (PWM)

(Breit et al. 1983). Another set of experiments established that α1-antitrypsin competed with

diferric transferrin for the binding of the transferrin receptors inhibiting the proliferation of

human skin fibroblasts (Graziadei et al. 1998).

The effects of α1-antitrypsin on cytokines have been also pursued. Alpha1-antitrypsin, but

not the secretory leukocyte protease inhibitor, inhibited in a dose-dependent manner the release

of the pro-inflammatory cytokines TNF-α, monocyte chemoattractant protein-1, IL-8 and IL-1β

from LPS-stimulated human monocytes (Janciauskiene et al. 2004). Moreover, in the same

study, α1-antitrypsin increased IL-10 release from human monocytes stimulated with LPS.

Page 22: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

22

However, the inhibitory effects of α1-antitrypsin on the pro-inflammatory cytokines were not

blocked when neutralizing antibodies against IL-10 were included in the experiments

(Janciauskiene et al. 2004).

Possible therapeutic roles for α1-antitrypsin in some human diseases such as type I diabetes

has been also studied. Alpha1-antitrypsin prevented the apoptotic effects of TNF-α and

streptozotocin (STZ, a diabetes inducer) on the MIN6 mouse insulinoma cell line through

caspase-3 activity inhibition (Zhang et al. 2007). In addition, administration of α1-antitrypsin to

the non-obese diabetic (NOD) mouse prevented the development of type 1 diabetes and reduced

insulin autoantibodies (Song et al. 2004). Moreover, Lewis et al. (2005) demonstrated that α1-

antitrypsin prolonged islet graft survival in C57BL/6 mice treated with STZ. Similar results

were obtained from experiments using C57BL/6 mice also treated with STZ, where the

administration of α1-antitrypsin reduced the levels of glucose in the blood, number of apoptotic

β-cells and rate of diabetes (Zhang et al. 2007).

Serpinopathies

The importance of the serine proteinase inhibitors is also highlighted by the loss of

function, deficiency and diseases (also known as serpinopathy) caused by mutations, leading to a

serpin existing in a non-functional state.

Serpin polymerization is a non-functional state which is achieved by the binding of the

RCL of one serpin to the β-sheet A, or in some cases to the β-sheet C of other serpin, forming a

long-inactive polymer. This phenomenon causes the accumulation of polymerized serpins as

misfolded protein, therefore decreasing the amount of functional serpins available (Kaiserman et

al. 2006, Law et al. 2006). In humans, α1-antitrypsin polymerization could lead to emphysema

and liver cirrhosis and the polymerization of neuroserpin could cause familial encephalopathy

(FENIB) (Kaiserman et al. 2006, Law et al. 2006). The other non-functional state is latency

Page 23: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

23

where the RCL is inserted into the β-sheet A without being cleaved by the target proteinase

causing the inactivation of the serpin (Kaiserman et al. 2006).

Serpin mutations also alter the functionality and stability of the protein. Mutations can

induce polymerization (the neuroserpin pathology and α-1 antitrypsin Z mutant), cancer (loss of

function of maspin), change in serpin specificity (the α1- antitrypsin Pittsburg mutation, where

an amino acid substitution at the P1 site of this serpin generates a potent inhibitor of thrombin

and other coagulation enzymes, causing death by bleeding disorders), and also loss of cofactor

binding site to disrupt the interaction between heparin and antithrombin III (Law et al. 2006,

Kaiserman et al. 2006).

In addition, gain-of-function serpinopathy can occur when genetic mutations lead to

overexpression of a serpin. The excess in the synthesis of megsin, a serpin produced by human

mesangial cells, causes accumulation and polymerization of the protein in the endoplasmic

reticulum of the kidney glomerular epithelial cells (Inagi et al. 2005, Miyata et al. 2005). This

renal serpinopathy is characterized by mesangial matrix expansion, stress of the endoplasmic

reticulum, and augmented IgA deposition which leads to renal failure (Miyata et al. 2005,

Kaiserman et al. 2006). The overexpression of the mutant megsin, which lacks anti-proteolytic

activity and flexibility at the hinge region loop, does lead to polymerization, but not to the

generation of a serpinopathy, suggesting the importance of the overall structure and function of

megsin for the development of the disease (Inagi et al. 2005, Miyata et al. 2005).

Receptor Mediated Internalization of the Serpin-Proteinase Complex

Once the target proteinase is bound to the serpin, a stable complex is formed by the inactive

proteinase and the cleaved serpin. This serpin-proteinase complex is cleared rapidly from the

circulation by an internalization pathway mediated by hepatic receptors that leads to lysosomal

degradation. Perlmutter et al. (1990) showed a specific cell surface receptor in human

Page 24: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

24

monocytes and hepatoma (HepG2) cells, which was referred as the serpin-enzyme complex

receptor (SEC receptor). The SEC receptor recognized and subsequently internalized the

following serpin-proteinase complexes α1-antitrypsin-elastase, α1-antichymotrypsin-cathepsin

G, antithrombin III-thrombin and also complement C1-inhibitor-C1s but with less affinity

(Perlmutter et al. 1990, Poller et al. 1995). Later, experimental data revealed that most of the

serpin-enzyme complexes are also internalized by the low-density lipoprotein (LDL) family of

endocytic receptors which includes the very-low-density lipoprotein (VLDLR), the low-density

lipoprotein-receptor-related protein 1 (LRP1), which was originally known as LRP or

occasionally as CD91, the glycoprotein 330 (gp330) also known as megalin or LRP2, and the

low-density lipoprotein (LDLR) receptors (Kounnas et al. 1996, Skeldal et al. 2006, Lillis et al.

2008).

Since the SEC receptor was never sequenced and the LRP1 is abundantly found in the

liver, this receptor is the most likely candidate for the internalization of the serpin-proteinase

complexes. Kounnas et al. (1996) demonstrated that addition of either LRP1 antibodies, the

antagonist chaperone receptor associated protein (RAP) or chloroquine (an inhibitor of

lysosomal degradation) to the HepG2 cells inhibited the internalization and degradation of α1-

antitrypsin-trypsin, antithrombin III-thrombin and heparin cofactor II-thrombin complexes.

Moreover, these serpin-proteinase complexes were not internalized by mouse fibroblasts lacking

LRP1 receptors (Kounnas et al. 1996). In addition, native or cleaved forms of the serpins alone

were not recognized as ligands and subsequently internalized by the LRP1 receptor, suggesting

the specificity of the receptor for serpin-proteinase complexes only (Kounnas et al. 1996).

Moreover, in vivo experiments using the rat as a model showed that the antagonist RAP delayed

the internalization of radiolabeled antithrombinIII-thrombin complexes from plasma circulation

Page 25: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

25

(Kounnas et al. 1996). Similar experiments demonstrated that other serpin-proteinase complexes

such as complement C1-inhibitor-C1s, α1-antitrypsin-neutrophil elastase, protease nexin-1-

thrombin, protease nexin-1-factor XIa, neuroserpin-tissue-type plasminogen activator and

protease nexin-1-factor VII-activating protease are internalized by the endocytic LRP1 receptor

(Storm et al. 1997, Poller et al. 1995, Crisp et al. 2000, Knauer et al. 2000, Makarova et al.

2003, Muhl et al. 2007).

The role of heparin in the binding of the serpin-proteinase complexes to the LRP1

receptor and subsequent internalization has been also studied, in particular the binding of

protease nexin-1 to heparin after the serpin-proteinase complex has formed. Experiments

performed by Knauer et al. (1997) tested the role of heparin and LRP1 in the internalization of a

variant form of the protease nexin-1 (PN1-K7E) which does not bind heparin. They showed that

the complex PN1-K7E-thrombin could form but, internalization of the complex and further

degradation was greatly reduced in normal mouse embryonic fibroblasts and nearly abolished in

the LRP1-deficient mouse embryonic fibroblasts, when compared with the experimental

controls. Moreover, in the absence of endocytosis, the addition of soluble heparin inhibited the

binding of the serpin-proteinase complex to the surface of human and mouse embryonic

fibroblast cells (Knauer et al. 1997). The authors concluded that cell surface heparins mediated

the binding of protease nexin-1-thrombin complex to the LRP1 and further internalization.

Similar experiments were performed by Crisp et al. (2000) to study the role of heparins, in the

internalization of the protease nexin-1-urokinase plasminogen activator (uPA) complex through

the LRP1. These authors showed that the mechanisms of cell surface binding and internalization

of this serpin-proteinase complex are heparin-independent. These results indicate that the

Page 26: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

26

interaction of the serpin-proteinase complex with cell surface receptors in the endocytic pathway

is determined by the specific serpin or proteinase involved.

There is also a specific receptor, the urokinase plasminogen activator receptor (uPAR)

known as well as CD87, that plays a role in the internalization of the uPA and tissue-type

plasminogen activator (tPA) complexes formed with either plasminogen activator inhibitor

(PAI)-1 or PAI-2 (Olson et al. 1992, Wind et al. 2002). Even though active uPA binds with high

affinity to uPAR, its internalization is much slower than the uPA-PAI-1 complex (Wind et al.

2002). Moreover, the internalization of uPA-PAI-1 bound to the uPAR receptor and the later

recirculation of the receptor requires the interaction of the α2-macroglobulin receptor/low

density lipoprotein receptor- related protein (α2 MR/LRP), a large cell-surface glycoprotein

(Binder et al. 2007). There is experimental evidence showing that the internalization of the uPA-

PAI-1 complex can be also mediated through the related receptor sorting protein-related receptor

(sorLA), LRP2, LRP1 and VLDLR, all members of the LDLR family of endocytosis receptors

(Wind et al. 2002, Skeldal et al. 2006). Likewise, it was recently demonstrated by Chroucher et

al. (2006) that the internalization of the uPA-PAI-2 complex is also achieved by the LRP1

receptor.

Evolution of Serpins and Their Functions

It was previously believed that serpin genes were restricted to higher eukaryotes. However,

recent studies indicate that serpins have an ancient origin, at least 1000 million years old, and

they are not only limited to higher eukaryotes organisms, but instead are present in at least some

species of all phyla on Earth.

Serpin genes have been found in some complete prokaryote genomes including Archaea

and Bacteria (Irving et al. 2002b). In the Bacteria genera, serpin genes are present in

cyanobacteria, actinobacteria and formicutes, but not in any proteobacteria family with complete

Page 27: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

27

genomes (Irving et al. 2002b, Roberts et al. 2004, Ivanov et al. 2006). The conservation of

residues in functional key regions and secondary structural elements of these serpins suggests

their ability to inactivate proteinases (Irving et al. 2002b, Roberts et al. 2004). Ivanov et al.

(2006) demonstrated that the serpin found in the actinobateria Bifidobacterium longun inhibited

human neutrophil elastase and porcine pancreatic elastase. Kang et al. (2006) showed that there

are three serpin genes in the formicute Clostridium thermocellum (Clotm-serpin 1, 2 and 3). The

Clotm-serpins-1 and 2 are close related, with the hinge region of the RCL very conserved with

inhibitory serpins whereas Clotm-serpin-3 is a divergent serpin. In the same study, it was

demonstrated that serpin-1 did not inhibit trypsin, chymotrypsin or papain, but it had inhibitory

activity towards subtisilin, a bacterial serine proteinase. Multiple serpin genes were found in two

species of the Archaea and Bacteria domains, where the variability of the RCL suggested that

these genes were generated by duplication and later diversification (Roberts et al. 2004)

Despite the cases described above, serpin sequences are not broadly present in the

prokaryotic kingdom. No serpin sequences were identified in 13 Archaea and 56 Bacteria

genera with complete genome sequences, which suggested that in prokaryotes the serpin gene

was probably acquired by lateral transfer or lost when it was not vital (Irving et al. 2002b,

Roberts et al. 2004, Ivanov et al. 2006).

In viruses, serpin genes have been only identified in the poxviridae family and they can be

localized in extracellular and intracellular compartments (Brooks et al. 1995, Lucas & McFadden

2004). Three active serpin genes were found in the Orthopoxviruses genus. The viral serpin-1

(SPI-1) is a 40 kDa protein that blocked apoptosis in rabbitpox virus infected cells (Brooks et al.

1995). The viral serpin-2 (SPI-2) or CrmA inhibited efficiently caspase 1, preventing the

production of IL-1β and 18 (Brooks et al. 1995, Callus & Vaux 2007). Also, it was found that

Page 28: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

28

SPI-2 inhibited caspases 3 and 8 (Brooks et al. 1995, Callus & Vaux, 2007). Finally, the viral

serpin-3 (SPI-3) is involved with regulation of virus-host cell fusion (Brooks et al. 1995).

Although the amino acid sequence of SPI-1 is almost 50% identical to SPI-2, the predicted RCL

of these two genes differs significantly (Brooks et al. 1995). Irving et al. (2000) suggested the

possibility of gene duplication from a single gene as the origin for SPI-1 and -2, but separately

from SPI-3. Another viral serpin gene, (SERP-I) was found in the myxoma virus of the

Leporipoxvirus genus. SERP-I is a 55 kDa glycosylated secreted protein which binds and

inhibits human host plasmin, uPA and tPA. The knockout SPI-1 myxoma virus demonstrated its

importance on reducing the immune response of host monocytes and macrophages (Lucas &

McFadden 2004).

Serpin genes are also present in several green algae and plants. In the plant kingdom,

serpin genes are present in moss and conifers (Roberts & Hejgaard 2008). Moreover, serpins

have been found in the phloem sap of the cucurbits pumpkin and cucumber and in very high

quantities in the seeds of barley, wheat, rye, oats and apple (Roberts & Hejgaard 2008). There is

very little information about the genomic organization of most of the members of the plant

kingdom. There is only one serpin gene present in the unicellular green algae Chlamydomonas

reinhardtii, 7 serpin genes in Arabidopsis and 14 in rice (Roberts & Hejgaard 2008). Although

the first plant serpin genes identified were inhibitory, recent findings in maize and rice have

shown the expression of close related non-inhibitory serpin genes (Roberts & Hejgaard 2008).

There are no studies on the evolution of serpin genes in green algae and plants, however, Roberts

& Hejgaard (2008) have postulated the possibility of a lateral gene transfer from bacteria to algal

ancestors and inherited from the most advanced green algae when they evolved to land plants.

Page 29: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

29

The function of serpins in green algae and plants remains unclear. Most of the known

proteinases are found in plants. Typical animal caspases are absent, but cysteine proteinases are

involved in the programmed cell death (Roberts & Hejgaard 2008). Green algae and plants also

lack genes homologous to those encoding trypsin, elastase, thrombin, granzyme B. One

chymotrypsin-like proteinase has been discovered in plants, and in vitro studies have shown that

serpins from wheat, barley, rye, oat, pumpkin, apple and Arabidopsis are able to inhibit serine

proteinases of the chymotrypsin group (Roberts & Hejgaard 2008).

Silverman et al. (2001) postulated that phloem sap serpins could be involved in the host

mechanism of defense, since the phloem serpin-1 found in Cucurbita maxima decreased the

ability of the piercing-sucking aphids, Myzus persicae to survive and reproduce on pumpkins

(Yoo et al. 2000). Likewise, it has been proposed that the function of serpins found in high

concentrations in plant seeds protects storage proteins from degradation caused by proteolytic

enzymes of fungi or insects (Roberts & Hejgaard 2008).

Serpin sequences have been not found in complete genomes of Saccharomyces cerevisiae

(beaker’s yeast) and Schizosaccharomyces pombe (fission yeast) (Roberts et al. 2004).

However, a recent study conducted by Steenbakkers et al. (2008) revealed that the presence of

one serpin gene in the fungal kingdom. Analysis of the amino acid sequence of this so-called

‘celpin’ showed that it seems to have all the features of a prototypical functional serpin

(Steenbakkers et al. 2008). Moreover, the serpin gene is present in the unicellular eukaryote

Entamoeba histolytica which lacks of an N-terminus signal peptide even though it is secreted by

activated trophozoites (Riahi et al. 2004). The interaction between this serpin and trypsin is

typical of non-inhibitory serpins where the proteinase is still active while the serpin is cleaved in

Page 30: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

30

its C-terminus (Riahi et al. 2004). In the same study, it was shown that the serpin had non-

inhibitory activity towards chymotrypsin, elastase and cathepsin G.

Additionally, serpins are present in early metazoans. Cole et al. (2004) cloned the full-

length cDNA of a serpin gene called ‘jellypin’ from the cnidarian jellyfish Cyanea capillata.

Analysis of the amino acid sequence showed that this serpin lacks of N-terminal signal sequence

as is characteristic of intracellular serpins. In the same study, it was determined that jellypin

contained a functional RCL and inhibited human chymotrypsin, elastase, and capthesin G,

however, under unphysiological conditions (Cole et al. 2004). Since the closest relative of

jellypin in the serpin superfamily are plant serpins, the authors suggest that this metazoan serpin

arose approximately 1000 million years ago (Mya), around the time of divergence between

plants and animals (Cole et al. 2004).

In nematodes, eight serpin genes have been found in Caenorhabditis elegans and the same

numbers of serpin genes are present in parasitic species such as Ascaris suum, Brugia malayi,

Onchocerca volvulus, Trichostrongylus vitrinus and Shistosoma mansoni (Zang & Maizels

2001). The role for these serpins has been related to the evasion of the host’s immune response

and limiting tissue damage in response to inflammation since BmSPN-2 inhibits cathepsin G and

elastase, both produced by neutrophils (Zang & Maizels 2001). In these species there are also

small serpins known as ‘smapins’ which have less than 100 amino acid residues, ten cysteine

residues forming five disulphide bonds where two of these are located at each side of the RCL,

and no structural relation with the well known members of the superfamily (Zang & Maizels

2001). In the dog hookworm A. caninum, smapins inhibits serine proteases that are involved in

the host’s blood coagulation pathway (Zang & Maizels 2001).

Page 31: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

31

The percent of identity found between the C. elegans serpins CeSPN-3 and CeSPN-4 is

87% while the identity between CeSPN-7 and CeSPN-8 is 78% (Zang & Maizels 2001). The

authors concluded that these serpin genes in C. elegans arose as a local duplication event (Zang

& Maizels 2001). Moreover, it was also suggested that a possible intron gain-and-loss is the

model for the serpin gene evolution in nematodes (Zang & Maizels 2001).

Phylogeny of the Serpin Superfamily

In humans, serpins are classified in nine groups A-I where the largest group are the

extracellular clade A or SERPINA with 13 members found in chromosomes 1, 14 and X and the

intracellular clade B or SERPINB with 13 members on chromosomes 18 and 6 (van Gent et al.

2003, Law et al. 2006, Izuhara et al. 2008). The groups SERPINC and SERPIND are

represented by antithrombin and heparin cofactor II respectively (van Gent et al. 2003). The

serpins PAI-1 and glial-derived nexin are grouped together in the SERPINE group, α2-

antiplasmin and PEDF in SERPINF, and the complement C1 inhibitor in the SERPING group

(van Gent et al. 2003, Kaiserman et al. 2006). The SERPINH group is represented by the HSP-

47, the myoepithelium-derived serpin, also known as ‘pancpin’, and neuroserpin are in the

SERPINI group (van Gent et al. 2003, Kaiserman et al. 2006).

The phylogenetic analysis conducted by Irving et al. (2000) divided the serpin superfamily

into 16 clades or classes (A-P). The largest clades in the classification are Clade A and B and

they are formed by extracellular or antitrypsin-like serpins and intracellular or ov-like serpins

respectively. This last group of serpins is suggested to be ancestors to the majority of

extracellular serpins. The other clades are formed by serpins that have been found in insects,

nematodes, plants and virus; however, there are ten highly diverged orphan serpins which failed

to group within any of the clades; these proteins are also known as orphan serpins (Irving et al.

2000, Silverman et al. 2001). The viral serpins SPI-1 and -2 were clustered together in clade n,

Page 32: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

32

but SPI-3 was classified alone in clade o (Irving et al. 2000). In addition, the myxoma serpin

SERP-I was placed in clade e clustered together with plasminogen activator inhibitor-1 and

protease nexin-1 (Irving et al. 2000).

A study performed only with the nematode serpins grouped all the C. elegans serpins

together in one cluster whereas serpins present in other species were separated in different

branches as an indication of divergence (Zang & Maizels 2001). These findings were similar to

those found by Irving et al. (2000) where the C. elegans serpins were clustered together in clade

l whereas the Schistosoma serpins are clustered in clade m and all the other nematode serpins are

consider orphans.

Cole et al. (2004) conducted also a phylogenetic study to determine the relationship of the

metazoan serpin jellypin with other members of the serpin family. The results classified jellypin

as an orphan, but its closest relatives were the plant serpins (Cole et al. 2004), which are

clustered in clade p (Irving et al. 2000).

More complex phylogenetic analyses were performed by Atchley et al. (2001) (Figure 1-2)

and Ragg et al. (2001) where they took into consideration amino acid sequences, exon-intron

structures and diagnostic of amino acid sites. The results from the study performed by Atchley

et al. (2001) were similar to those presented by Irving et al. (2000) where serpins were clustered

within ten clades or groups with two largest groups, the α1-antitrypsin and ovalbumin clades,

and several orphan serpins. Since the study by Ragg et al. (2001) was performed using only

vertebrate serpins, the number of clades or group was reduced to six with α1-antitrypsin and

ovalbumin groups being the largest. All three studies classified antithrombin III and HSP-47 in

separate clades or groups (Irving et al. 2000, Ragg et al. 2001, Atchley et al. 2001). However,

the intron-exon data from Atchley et al. (2001) and Ragg et al. (2001) showed that neuroserpins,

Page 33: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

33

PAI-1 and glia derived nexin are grouped together within a clade suggesting a single

evolutionary lineage, whereas Irving et al. (2000) clustered neuroserpins into a separate group

from the other two serpins. Likewise, the intron-exon data clustered together complement C1-

inhibitor, PEDF and α2-antiplasmin (Atchley et al. 2001, Ragg 2001) whereas Irving et al.

(2000) placed the protease C1 inhibitor as a separate clade.

Positive Selection in Serpins

Positive or Darwinian selection refers to selective pressure acting on proteins that can

provide evolutionary advantage to an organism. The most used method to detect positively

selected sites in protein coding sequences is comparing the ratio (ω) between the non-

synonymous substitutions (dN), where a nucleotide substitution will lead to an amino acid

replacement of the encoded protein, with the synonymous substitutions (dS) where the

substitution of a nucleotide will not change the amino acid leaving the encoded protein

unchanged (Wong et al. 2004). When the ω ratio is greater than 1, the result is interpreted as

evidence of positive selection, but if the ω ratio is equal or less than 1, results are interpreted as

evidence of neutral or purifying selection respectively (Wong et al. 2004).

There is enough evidence suggesting that serpins, like a few other protein families, have

undergone positive or Darwinian selection. Positive selection has occurred at the RCL region of

the protein, in particular at the P1 site proximal to the scissile bond of the RCL which changes

the selectivity for the target proteinase (Brown 1987, Hill & Hastie 1987, Goodwin et al. 1996,

Zang & Maizels 2001, Barbour et al. 2002). Experimental data from Barbour et al. (2002)

showed that single α1-antitrypsin isoforms from two different species of mouse inhibited

different serine proteinases. Moreover, in the same study it was determined that this diversity in

the functionality of these isoforms was caused in particular by the variations in the RCL area

Page 34: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

34

Figure 1-2. Neighbor-joining tree of 110 serpin sequences. The clusters of proteins show close

correspondence to the groups of proteins described by the analyses of exon-intron

structure. Atchely et al. (2001) by permission of the Oxford Journals, Oxford

University Press.

Page 35: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

35

(Barbour et al. 2002). In contrast, van Gent et al. (2003) did not find any positive selection in a

study that compared some serpin groups between human and rodents, human and artiodactyla

and rat versus mouse.

The positive selection of the serpin genes has been reported in different species in which it

is suggested the advantage of this process during the evolution of the serpins in order to

neutralize a broader range of proteinases. For example, it has been postulated that caspase

inhibitor genes in viruses have undergone positive selection as a defense mechanism against the

host’s immune response (Brooks et al. 1995, Callus & Vaux 2007). Likewise, in nematodes, the

rapid diversification of the RCL has allowed novel changes in these serpins and smapins in order

to target mammalian host’s proteinases (Zang & Maizels 2001). There is also circumstantial

evidence suggesting that the evolution of the RCL of serpins present in plant seeds is because of

positive Darwinian selection to inhibit exogenous proteinases from insects and pathogens

(Roberts & Hejgaard 2008).

Novel Serpins and their Functions

As previously mentioned, the mechanisms of actions and functions of some serpins has

been extensively studied and well documented. However, recent discoveries have found new

serpins with novel characteristics and interesting biological functions. One example is the novel

nuclear serpin, myeloid and erythroid nuclear termination stage-specific protein (MENT) which

has an ‘M loop’, a large insertion between the C- and D-helices. The M loop is also known as

the CD loop and contains a nuclear localization signal and the AT-hook motif which is required

for chromatin and DNA binding (Silverman et al. 2001, Irving et al. 2002a). This intracellular

serpin is the major non-histone chromatin protein localized in the heterochromatin and it

functions by inducing higher order chromatin condensation by connecting separate nucleosomes

in vitro (Silverman et al. 2001, Irving et al. 2002a). In addition, MENT is one of those cross-

Page 36: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

36

class serpins that inhibits cysteine proteinases and papain-like cysteine proteinases such as

cathepsins K, L and V (Irving et al. 2002a). Experimental data showed that MENT is involved

in cell cycle progression and therefore in cell proliferation by inhibiting the enzymatic activity of

the nuclear cysteine proteinase SPase, a cathepsin L-like proteinase, whose function has been

related to the degradation of the phosphorylated form of the retinoblastoma (Rb) protein, a

known regulator of the cell cycle (Irving et al. 2002a).

Plasminogen activator inhibitor type-2 is another interesting serpin with novel function.

The bi-topological PAI-2 is predominantly translated as an intracellular 47 kDa non-glycosylated

protein, but it can be also a 60 kDa glycosalyted secreted protein as a combination of an

inefficient nuclear localization signal and the lack of a conventional hydrophobic amino-terminal

sequence (Medcalf & Stasinopoulos 2005, Croucher et al. 2008). Like the serpin MENT, the CD

loop is present in PAI-2, which binds non-covalently to several proteins including the Rb protein

(Medcalf & Stasinopoulos 2005). Although the physiological role of the intracellular PAI-2

remains unknown, it has been suggested that PAI-2 could be involved in the cell cycle regulation

since this serpin protected Rb from degradation by a distinct anti-proteolytic mechanism

(Medcalf & Stasinopoulos 2005, Croucher et al. 2008).

A therapeutic role of PAI-2 in the reduction of tumor growth and metastasis of different

types of cancer has been extensively demonstrated using in vitro systems and rodent tumor

models where uPA was inhibited completely and a significant decrease in the extra cellular

matrix degradation achieved (Croucher et al. 2008). High levels of expression of PAI-2 in breast

cancer tumors are associated with reduced metastasis, tumor size and prolonged survival whereas

opposite effects are expected when this serpin is expressed at low levels (Croucher et al. 2008).

Page 37: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

37

More recently, another novel serpin was discovered in the visceral adipose tissue of the

obese-type 2 diabetes rat model. This new serpin called visceral adipose tissue-derived serpin

(vaspin) was found to be up-regulated in the obese-type 2 diabetes rat model and down-regulated

in the non obese-diabetes resistant rat model (Hida et al. 2005). The vaspin transcript was up-

regulated in the subdermal white adipose tissue by the administrations of insulin and

thiazolidinediones (TZD), a compound that enhances insulin sensitivity (Hida et al. 2005). The

authors suggested from the experimental data that vaspin inactivates unknown target proteases

derived from fat or other tissue that inhibited the effects of insulin (Hida et al. 2005). However,

it was demonstrated in the same study that vaspin lacks anti-proteolytic activity towards common

serine proteinases such as trypsin, factor Xa, elastase, urokinase, collagenase and dipeptidyl

peptidase (Hida et al. 2005). The vaspin gene is also expressed in the white adipose tissue of

mouse and human (Hida et al. 2005).

The US are also novel with respect to serpins by virtue of their limited distribution among

mammals, since no human homologue of the US gene has been reported (van Gent et al. 2003).

The primary site of expression for the US is in the endometrium and regulation of their

expression by progesterone. A detailed description of their characteristics, properties and

plausible functions for this group of serpins are described below.

Uterine Serpins

Uterine serpins were first described as progesterone induced proteins in the uterine fluid of

the pregnant sheep, pig and cow (Moffatt et al. 1989, Baumbach et al. 1986, Leslie et al. 1990).

The protein in sheep, discovered first, was called uterine milk protein (UTMP) because it was the

major protein in uterine fluid (called uterine milk by Aristotle). The two US present in the pig

were originally known as uteroferrin-associated protein or uteroferrin-associated basic proteins

(UfAP/UABP) because they form non-covalently heterodimers with uteroferrin (Baumbach et al.

Page 38: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

38

1986, Roberts & Bazer 1988). The connection between UTMP and UfAP was first made when

the genes were sequenced. Ing & Roberts (1989) found greatest homology between the cDNA

sequence of ovine UTMP with α1-antitrypsin and classified UTMP as a member of the serine

proteinase inhibitor (serpin) superfamily. Shortly thereafter, Malathy et al. (1990) found that

both pig proteins were serpins and also highly similar to ovine UTMP. Mathialagan & Hansen

(1996) identified the bovine UTMP as a serpin closely related to sheep UTMP and pig UfAPs

and proposed that old designations be eliminated and proteins termed as uterine serpins (US)

preceded by the name of the specie from where it was identified such as (Bo) for bovine, (Ov)

for ovine and (Po) for porcine. Since this paper, additional US were identified in the goat (Tekin

et al. 2005a) and a sequence for water buffalo (Bubalus bubalis) has been submitted to Genbank

(Accession number: DQ 661648.1)

Uterine serpins have been only characterized in species with epitheliochorial placenta. All

US proteins that have been described to date possess a 25-amino acid signal peptide sequence

and are secreted into the uterine lumen (Mathialagan & Hansen 1996, Tekin et al. 2005a).

Mathialagan & Hansen (1996) determined that most of the amino acids present in OvUS, BoUS

and PoUS-1 and -2, necessary for the serpin backbone structure, are conserved at the same

positions with those on α1-antitrypsin. However, analysis of the hinge regions of all US

indicates that these serpins are not conserved with inhibitory serpins and are probably not

functional proteinase inhibitors (Irving et al. 2000, Tekin et al. 2005a). In the hinge region of

US, the only amino acid that is conserved with the inhibitory serpins is alanine at the P12

position whereas the other positions are not (Tekin et al. 2005a). In addition, the amino acid at

P1-P1’ site for known uterine serpins is conserved among species where valine is usually found

with the exception of OvUS which has an alanine at this position (Tekin et al. 2005a).

Page 39: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

39

The OvUS was screened for anti-proteinase activity against several proteinases without

identification of a target proteinase (Ing & Robets 1989). Both BoUS and OvUS are capable of

inhibiting pepsin at very high concentrations (Mathialagan & Hansen 1996, Peltier et al. 2000a),

but this activity occurs even when the protein is proteolytically cleaved and may be distinct from

the classical serpin inhibitory mechanism involving a functional RCL (Peltier et al. 2000a).

Uterine serpins have a conserved KVP sequence at the P4-P2 site of the RCL (Tekin et al.

2005a, Mathialagan & Hansen 1996). There is also a unique 39 amino acid insertion in the

BoUS, which incorporates two KAKEVPAVVKVPM repeats within the putative P1-P1’ site and

one KEVPVVVKVP sequence right after that P1-P1’ site (Mathialagan & Hansen 1996, Tekin et

al. 2005a). The KEVPVVVKVP motif is also found further upstream as a conserved sequence

among all known US (Mathialagan & Hansen 1996). The real function for all these repeats

remains unknown. Mathialagan & Hansen (1996) suggested that US may inhibit some members

of the aspartic proteinase family, because of the resemblance of the motif to the VVVK present

in pepsinogens. However, Peltier et al. (2000a) showed that the addition of a synthetic peptide

corresponding to the putative P7-P15’ region of OvUS did not inhibit pepsin activity.

Ovine Uterine Serpin

This is the most studied protein of the US group. Ovine US is a secreted protein which is

present in the uterine fluid as a pair of basic glycoproteins with molecular weights of 55,000 and

57,000, derived from a single 54,000 precursor (Moffatt et al. 1987, Hansen et al. 1987a). This

basic glycoprotein possess an isoelectric point of 9.2 and two N-linked glycosylation sites, which

suggests that the two major forms of the protein may differ in the number of carbohydrate chains

(Hansen et al. 1987a, Ing & Robets 1989). The amino acid composition was found to be rich in

lysine, leucine and threonine, but low in tyrosine and devoid of tryptophan (Hansen et al.

1987a). Analysis of the carbohydrate content of OvUS showed that it consists of 2.8% neutral

Page 40: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

40

sugars, 2.5% amino sugars, and 0.3% sialic acid (Hansen et al. 1987a). Additionally, OvUS

possess a mannose 6-phoshate, the so-called lysosomal recognition marker (Hansen et al.

1987a).

Endometrial secretion. The major regulator of OvUS secretion is the hormone progesterone

(Moffatt et al. 1987, Ing et al. 1989, Leslie & Hansen 1991, Padua et al. 2005). Experiments

performed using the ovariectomized ewe as a model showed that after 6 days of treatment with

progesterone, OvUS can be detected in endometrial epithelial cells (Ing et al. 1989). Moreover,

progesterone therapy for 14-60 days induced a large increase in the secretion of the protein (Ing

et al. 1989, Leslie & Hansen 1991, Padua et al. 2005). Ovine US can be detected in uterine

secretions of ovariectomized ewes treated for 30 days with a combination of progesterone plus

estrone, however, the protein could not be detected when they were treated with estrone only

(Moffatt et al. 1987). In addition, levels of OvUS mRNA increased in the glandular epithelium

of ovariectomized ewes receiving uterine infusion of ovine placental lactogen and/or ovine

growth hormone combined with IFN-τ infusions and the administration of progesterone as

compared with the levels in ovariectomized ewes infused with control proteins (Spencer et al.

1999, Noel et al. 2003). It was also shown that the co-administration of estradiol with

progesterone, where estradiol up-regulates the expression of progesterone receptors in the

endometrium, decreased OvUS mRNA in the glandular epithelium (Spencer et al. 1999).

During the estrous cycle, OvUS mRNA can be detected in uterine endometrium around

days 13-16 (Stewart et al. 2000). In the pregnant sheep, the transcript for OvUS can be first

detected at days 13-15 (Ing et al. 1989, Stewart et al. 2000). Then, a 3-fold increase of steady-

state levels of OvUS mRNA in the glandular epithelium occurs between days 20 and 60, another

3-fold between days 60 and 80, and a decline at day 120 of pregnancy (Stewart et al. 2000).

Page 41: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

41

Between days 20 and 50 of pregnancy, the expression of OvUS mRNA is lower in the deep

glandular epithelium than in the upper glandular epithelium of the uterine stratum spongiosum.

There was however, no difference in mRNA expression between the upper and lower glandular

epithelium of the stratum spongiosum between days 50 and 60 of gestation (Stewart et al. 2000).

High levels of expression of the transcript for OvUS can be detected in all glandular epithelium

in the stratum spongiosum between days 60 and 120 of pregnancy (Stewart et al. 2000). At day

1 of postpartum, OvUS mRNA was only detected in the stratum spongiosum of the glandular

epithelium, but during postpartum days 7 and 28, the transcript for OvUS was not longer

detected (Gray et al. 2003).

It appears that OvUS is initially produced only by the uterine glands and then its expression

spreads to the luminal epithelium. Although OvUS protein was found only in glandular

epithelium at day 60 of pregnancy, it was immunolocalized in the luminal and glandular

epithelium of the intercaruncular endometrium by days 120 to 140 of pregnancy (Moffatt et al.

1987, Stephenson et al. 1989a). Ovine US has been also detected in amniotic and allantoic

fluids (Newton et al. 1989, McFarlane et al. 1999), presumably because of transplacental

transport.

Biological function. The biological function of OvUS is still not known with certainty although

evidence suggests it functions primarily to inhibit cell proliferation. Experiments performed by

Ing & Roberts (1989) tested the anti-protease activity of OvUS against trypsin, chymotrypsin,

plasmin, thrombin, elastase and plasminogen activator without significant inhibition. Likewise,

OvUS had non-inhibitory activity towards recombinant human cathepsin D, porcine cathepsin D,

recombinant cathepsin E, cysteine proteinase cathepsin B (Mathialagan & Hansen 1996) or

dipeptidyl proteinase IV (DPPIV) (Liu & Hansen 1995).

Page 42: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

42

Mathialagan & Hansen (1996) demonstrated that freshly purified OvUS inhibited pepsin A

and C (chymosin) activity at both pH 2.0 and 4.5, but an excess of 35- and 8-fold molar of OvUS

was required for a 50% inhibition of both aspartic proteinases, respectively. Moreover, the

complex formed by OvUS and pepsin could not be detected electrophoretically in the presence of

Sodium Dodecyl Sulfate (SDS) (Mathialagan & Hansen 1996) whereas, complexes formed by

inhibitory serpins and their partner proteinases (at 1:1 ratio) are so stable that they can be

resolved using the same approach (Potempa et al. 1994). Furthermore, the binding of OvUS to

pepsin seems to be electrostatic since it occurred at pH 8 and low salt concentration where OvUS

has positive charge and pepsin has a negative charge, but not at pH 10.25 where both proteins are

negatively charged (Peltier et al. 2000a).

Some studies have been conducted to determine the possible role of OvUS as a binding or

carrier protein. Ovine US can bind pregnancy-associated glycoproteins (Mathialagan & Hansen

1996), which are inactive aspartic proteinases secreted in large amounts by the ungulate placenta

(Xie et al. 1991, Green et al. 1998). Ovine US can also bind to activin, a member of the

transforming growth factor-β family present in the allantoic fluid (McFarlane et al. 1999).

However, the binding affinity of activin for OvUS is much lower than activin affinity towards

follistatin, suggesting that unlike follistatin, OvUS is not a candidate to neutralize the biological

activity of activin. In addition, OvUS can bind IgM and IgA (Hansen & Newton 1988).

However, the binding of OvUS to immunoglobulins was inhibited by the presence of high salt

concentrations, indicating the ionic nature of the binding (Hansen & Newton 1988).

A plausible function for OvUS is the inhibition of immune cell proliferation during

pregnancy to provide protection for the allogeneically-distinct conceptus (Hansen 1998).

Purified OvUS inhibited lymphocyte proliferation induced by mitogens such as the antigen

Page 43: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

43

Candida albicans, Con A, PHA and in the mixed lymphocyte reactions (Segerson et al. 1984,

Hansen et al. 1987b, Stephenson et al. 1989b, Skopets & Hansen 1993, Skopets et al. 1995). In

addition, OvUS reduced the antibody titer in ewes immunized against the T-cell dependent

antigen OVA (Skopets et al. 1995). However, like α1-antitrypsin (Breit et al. 1983), OvUS did

not cause any inhibitory activity against lymphocytes activated by the T and B cell mitogen

PWM (Skopets & Hansen 1993).

Ovine US also inhibits NK cell activity. In vivo experiments using the pregnant mice as a

model, OvUS blocked abortion induced by poly(I)•poly(C), which is a 50-base-pair compound

consisting of double-stranded RNA that activates NK cells, and reduced basal splenocyte NK

cell activity (Liu & Hansen 1993). Additional experiments conducted by Liu & Hansen (1993)

demonstrated that OvUS inhibited NK-like activity in sheep lymphocytes and mouse splenocytes

against K562 and YAC-1 target cells. Moreover, the lytic activity of NK-like cells in sheep

peripheral blood lymphocytes and endometrial epithelium against D-17 cells infected with

bovine herpes virus-1 was inhibited by OvUS (Tekin & Hansen 2002).

Some experiments have been conducted to elucidate the mechanism by which OvUS

inhibits cell proliferation. The protein does not act by inducing cytotoxicity because OvUS did

not cause cytotoxic effects on lymphocytes (Stephenson et al. 1989b, Skopets & Hansen 1993).

Binding of OvUS to lymphocytes is specific, dose dependent and saturable (Liu et al. 1999).

However, it is not known if OvUS binds to a specific cell surface receptor or competes with

other molecules for a receptor binding site. Experiments with Rp-8-Cl-cAMPS, a selective

inhibitor of cAMP-dependent type-I protein kinase (PK) A, indicated that effects of OvUS on

proliferation of PHA-stimulated lymphocytes are not dependent on this kinase (Tekin et al.

2005b). A study performed by Peltier et al. (2000b) showed that OvUS reduced proliferation of

Page 44: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

44

phorbol myristol acetate (PMA)-activated lymphocytes, suggesting that OvUS inhibits some

PKC mediated events. In the same study, the protein blocked IL-2 induced proliferation and

reduced expression of CD25 (IL-2Rα chain), but it did not affect the steady state amounts of IL-2

mRNA caused by Con A (Peltier et al. 2000b).

Skopets & Hansen (1993) demonstrated that the antiproliferative effect of OvUS on

lymphocytes was not blocked by addition of neutralizing antibody to transforming growth factor-

β. Thus, OvUS is not inhibiting lymphocyte proliferation by inducing secretion of this

immunosuppressive growth factor.

Porcine Uterine Serpin

The pig is the only species shown to contain two US genes (PoUS-1 and PoUS-2) with

predicted molecular weights of 45,123 and 46,009 respectively (Mathialagan & Hansen 1996).

The PoUS-1 has four potential N-glycosylation sites, whereas PoUS-2 presents only three,

lacking the one at (Asn107

) (Malathy et al. 1990, Mathialagan & Hansen 1996). Analysis of

purified PoUS showed three different peptides. There are two larger forms of the protein with

molecular weights of 50 and 46 kDa and identical NH2-amino terminus, but different in the

number or complexity of glycosylated chains (Baumbach et al. 1989, Murray et al. 1989). There

is also a small glycosylated polypeptide of 40 kDa, distinct from the other two in its NH2-amino

terminus (Baumbach et al. 1986, Murray et al. 1989) that may represent a proteolytic cleavage

product. These three polypeptides seemed to derive from a single 45 kDa precursor and like

OvUS, all of them posses the mannose 6-phosphate lysosome recognition signal (Murray et al.

1989).

Endometrial secretion. The porcine protein is present in uterine flushings of pregnant, pseudo-

pregnant and progesterone treated pigs (Baumbach et al. 1986). In addition, it can be detected in

allantoic fluid of conceptuses at mid pregnancy (Baumbach et al. 1986, Murray et al. 1989). In

Page 45: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

45

endometrial tissue, the protein was immunolocalized only in the glandular epithelium of

pregnant pigs at days 45 and 60 (Murray et al. 1989).

During pregnancy, PoUS mRNA expression is low at days 13 and 30, but the expression of

the transcript increases until the last month of gestation, which in the pig lasts 115 days (Malathy

et al. 1990). Moreover, high level of expression of the PoUS transcript was also detected in

endometrial tissues from day 68 pseudo-pregnant gilts (Malathy et al. 1990). In contrast, PoUS

mRNA expression was no detected in either endometrium or liver of non-pregnant or

ovariectomized pigs (Malathy et al. 1990).

Biological function. There is little evidence that PoUS-1 or PoUS-2 are inhibitory serpins.

Purified PoUS did not inhibit proteinases such as trypsin and chymotrypsin (Malathy et al.

1990). Mathialagan & Hansen (1996) tested the ability of PoUS to inhibit porcine pepsin using

uterine flushes from long-term progesterone treated pigs. The inhibitory activity of the protein

was only present in some batches, had very low stability, and the activity was lost completely

when the batch was kept at 4oC for more than one day.

The main role of the PoUS has been related to the iron transport from the mother to the pig

conceptuses by its association with the uteroferrin protein. Porcine uteroferrin is a purple-

colored alkaline phosphatase whose secretion by the glandular epithelium is also stimulated by

progesterone (Chen et al. 1975, Buhi et al. 1982, Baumbach et al. 1986, Murray et al. 1989).

Uteroferrin is transported by the areolae of the placenta into the allantoic fluid (Chen et al. 1975,

Ducsay et al. 1982, Renegar et al. 1982). Uteroferrin carries two bound iron atoms per

polypeptide chain (Baumbach et al. 1986, Roberts & Bazer 1988, Nuttleman & Roberts 1990).

Data from experiments performed by Buhi et al. (1982) in which [59

Fe] labeled uteroferrin was

injected into selected allantoic sacs of days 58 and 67 pregnant gilts showed that the [59

Fe] was

Page 46: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

46

released from uteroferrin and quickly associated with the protein transferrin, also present in the

allantoic fluid, and then from transferrin to the fetus. In addition, it was demonstrated that

uteroferrin was more susceptible to proteolysis after iron depletion (Buhi et al. 1982, Nuttleman

& Roberts 1990).

Porcine US binds non-covalently to uteroferrin. This pink color heterodimer has a

molecular weight of 80 kDa and is very stable for long periods of time in the presence of oxygen

whereas purple uteroferrin is not (Baumbach et al. 1989; Roberts & Bazer 1988). Also, the

association between PoUS and uteroferrin promotes the latter’s enzymatic activity in the

heterodimer conformation (Baumbach et al. 1989). Experimentally, the heterodimer can be

dissociated by low pH, antibodies raised against either uteroferrin or all three PoUS peptide

forms, or by dialysis at low salt concentration (Baumbach et al. 1989).

Vallet (1995) suggested that another possible role for PoUS during pregnancy is to prevent

embryonic losses caused by uteroferrin-ascorbic acid induced lipid peroxidation which is

harmful for smaller or less develop conceptuses. Experiments performed on microsomal

membranes from reproductive tissues showed that the uteroferrin-PoUS heterodimer in the

presence of ascorbic acid was less able to provoke lipid peroxidation damage than uteroferrin

itself (Vallet 1995).

Bovine Uterine Serpin

The bovine uterine serpin (BoUS) is also a basic protein and several molecular weight

forms can be detected in uterine secretions from pregnant and progesterone treated cows by

western blotting, with two major bands at 57,000 and 38,000 (Leslie et al. 1990, Leslie &

Hansen 1991). Unlike the porcine and ovine US, the BoUS has only one potential site for

glycosylation (Asn243

), which is conserved with OvUS and PoUS-1 and 2 (Mathialagan &

Hansen 1996).

Page 47: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

47

Endometrial secretion. Secretion of BoUS into the uterus is dependent upon progesterone.

Using western blotting, Leslie & Hansen (1991) demonstrated induction of BoUS in uterine

secretions of ovariectomized cows treated with progesterone for 10 to 30 days. The bovine US is

secreted by the endometrial epithelium of the cow. The protein was immunolocalized in the

glandular epithelium of endometrial tissues from day 135 of pregnancy and ovariectomized cows

treated for 30 days with progesterone (Leslie et al. 1990, Leslie & Hansen 1991). However, no

immunostaining was detected in either the glandular or luminal epithelium of endometrial tissues

from cows on days 17, 19, 21 of pregnancy or days 17 and 19 of the estrous cycle (Leslie et al.

1990).

Unlike OvUS, the BoUS mRNA is expressed during estrus, with particular highly

expression in the cranial area of the uterine horns (Bauersachas et al. 2005). During estrous, the

mRNA for BoUS was highly expressed in the superficial uterine glands and more weakly

expressed in the deep glandular epithelium and some dispersed epithelial cells of the

endometrium (Bauersachas et al. 2005). In addition, the BoUS transcript can be detected in the

caruncule, ovary and cotyledon (Khatib et al. 2007).

Biological function. The biological function of BoUS remains unknown. Mathialagan &

Hansen (1996) showed that activity of porcine pepsin was inhibited by crude uterine flushing

obtained from progesterone treated cows. The importance of BoUS for physiologic function is

highlighted by recent experiments by Khatib et al. (2007) indicating that a single nucleotide

polymorphism (A/G) at position 1269 of the BoUS gene was associated with a significant

increase in productive life in cattle populations, where the G allele expression at that position has

been linked to the productive life. Productive life is in large part determined by culling rate and

Page 48: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

48

culling is largely for problems with reproductive health or infectious diseases. Thus, BoUS is

likely to play a role in one or more processes controlling reproduction or immune function.

Caprine Uterine Serpin

This US was recently found discovered. Caprine (Cap) US was immunolocalized in the

endometrial glandular epithelium at day 25 of pregnancy, but it was not detected at day 5 of the

estrous cycle (Tekin et al. 2005a). Western blot of uterine fluid from a pregnant goat identified a

major band with a molecular weight of approximately 57,000 and some other low molecular

weight bands (Tekin et al. 2005a). Like the ovine and porcine, the CapUS had two predicted N-

glycosylation sites at positions 222 and 268, where the position 268 is conserved with the other

known US and the 222 position is also conserved in almost all the other US, with the exception

of the BoUS (Tekin et al. 2005a). No study has been performed to identify the possible

biological role of this US.

Evolution and Phylogeny of Uterine Serpins

Krem & Cera (2003) performed a study looking for possible evolutionary markers for

serpins, specifically serine residues at positions 53 and 56 (α1-antitrypsin numbering) of the

shutter region, where the serine 56 is related to the TCN-AGY codon usage dichotomy that is

associated to the protostome-deuterostome division. The authors determined that the OvUS,

BoUS, PoUS-1 and -2 posses a conserved serine (encoded by TCN codons) at position 53 which

is also preserved in all the sepins used for the study. At the position 56 they found that the

PoUS-1 and 2 also posses a conserved serine 56 (AGY codon) also found in almost all the

serpins present in chordates (Krem & Cera 2003). However, OvUS and BoUS were among

some other few serpins where distinct residue was found at position 56, where instead of serine,

alanine was present at this position (Krem & Cera 2003).

Page 49: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

49

Uterine serpins have been classified as a highly-diverge group of the large α1-antitrypsin

clade (Irving et al. 2000). According to the results obtained from another phylogenetic study,

OvUS and PoUS-1 and 2 were grouped into a large clade containing serpins with different

functions including α1-antitrypsin, heparin cofactor II, α1-antichymotrypsin, angiotensinogen

where genes coding for these serpins contain three introns at homologous areas in the conserved

part of the coding region (Atchley et al. 2001). A similar study performed only on vertebrate

serpins by Ragg et al. (2001) inferred that OvUS and PoUS-1 and 2 could classify within the α1-

antitrypsin group. However, the authors stated the need for further verification for these results

due to the lack of genomic organization for these three US. In contrast, Irving et al. (2000)

created phylogenetic trees with preexisting alignments of the Pfam database. In this analysis, the

US and the angiotensin like-serpins were grouped as separate clades rather than being included

into the α1-antitrypsin group. A phylogenetic study by Peltier et al. (2000c) also resulted in US

being considered as a separate clade in the serpin superfamily.

The identity of the predicted amino acid sequence of OvUS with the CapUS is 96%, 82%

with the BoUS and 55 and 56% with the PoUS-1 and 2 respectively (Mathialagan & Hansen

1996, Tekin et al. 2005a). Peltier et al. (2000c) estimated that OvUS and BoUS diverged from

the PoUS-1 and 2 about 60 Mya and the PoUS-1 and 2 diverged from each other around 5 Mya.

Computational analysis of US motifs suggested that OvUS and BoUS contain similar amounts of

casein kinase-2 and cAMP phosphorylation sites. In contrast, neither PoUS-1 nor 2 contain

cAMP phosphorylation (Peltier et al. 2000c). The same study also showed that these three

species have similar amounts of phosphorylation sites for tyrosine kinases as well as for N-

myristoylation. However, the PoUS-1 and 2 contain more phosphorylation sites for casein

kinase-2 and PKC and N-linked glycosylation sites than OvUS and BoUS.

Page 50: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

50

Synopsis and Objectives

Among the members of the serpin superfamily, the US group is a very intriguing group of

proteins. Based on this literature, there is evidence showing that US are hormonal-regulated

proteins and highly expressed into the uterus during pregnancy. The US group lacks apparently

of anti-proteinase activity and they plausibly diverge in biological function with respect to the

other members of the superfamily. Also, the limited group of species which posses the US gene

suggests the possibility of the evolution of the US gene within mammals for specific functions.

The specific objectives that addressed in this dissertation are

1) To determine the presence of the US gene in species with non-epitheliochorial placenta and

also to examine the evolution of the US gene in mammals.

2) To establish whether the US produced by the sheep (OvUS) inhibits cell proliferation by a)

causing cell death such as apoptosis or necrosis b) altering cell cycle dynamics to cause cell

cycle arrest.

3) To determine which cell cycle related genes are regulated by OvUS.

Page 51: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

51

CHAPTER 2

MOLECULAR PHYLOGENY OF UTERINE SERPINS AND ITS RELATIONSHIP TO

EVOLUTION OF PLACENTATION

Introduction

Evolution of placentation in mammals has been dependent upon new uses of existing genes

as well as appearance of new genes arising from gene duplication and selection for sequence

divergence. Knox & Baker (2008) have shown that genes expressed preferentially by the

placenta and decidua early in development tend to be ancient genes while the genes expressed

preferentially by the mature placenta have been formed recently. Examples of such trophoblast

genes are the interferon-τ genes found in ruminants (Roberts et al. 1999) and the primate

chorionic gonadotropins (Maston & Ruvolo 2002).

Another group of uterine genes formed by gene duplication are the uterine serpins (US)

(also called uterine milk proteins). These proteins are members of the serine proteinase inhibitor

(serpin) superfamily and have been identified as secretory products of the endometrial epithelium

of the pregnant sheep, goat, cow and pig (Moffatt et al. 1987, Leslie et al. 1990, Malathy et al.

1990, Tekin et al. 2005a). Uterine serpins have been classified as either a separate clade of the

serpin superfamily (Peltier et al. 2000c) or as a highly-diverge group of the α1-antitrypsin clade

(Irving et al. 2000). Recently, they have been designated as SERPINA14.

The prototypical serpins are competitive inhibitors of serine and cysteine proteinases. The

proteinase binds covalently to reactive center loop (RCL), which is localized on the top of the

serpin and contains a complementary sequence to the active site of the target proteinase. The

scissile bond at the P1-P1’ of the RCL is cleaved and then incorporated as the central strand of

the serpin also known as β-sheet A (Irving et al. 2000, Hungtinton 2006, Whisstock &

Bottomley, 2006). The inactivation of the proteinase is accompanied by an irreversible

Page 52: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

52

conformational change of the serpin structure that increases its overall stability (Silverman et al.

2001, Hungtinton 2006, Whisstock & Bottomley 2006).

A few serpins such as the US have evolved biological functions that do not involve

proteinase inhibitory activity. Other examples are the intracellular serpin mammary serine

protease inhibitor (maspin) (Sheng 2006), corticosteroid and thyroxine binding globulins

(Pemberton et al. 1998), pigment epithelium derived factor (PEDF) (Tombran-Tink &

Barnstable 2003) and the heat shock protein 47 (Sauk et al. 2005). Inhibitory serpins are usually

recognized by a consensus sequence in the hinge region which is localized within the RCL of the

serpin (Irving et al. 2000) but the putative hinge region of US is not conserved with inhibitory

serpins (Irving et al. 2000, Tekin et al. 2005a). The ovine (Ov) and bovine (Bo) US exhibit

either no or very weak inhibitory activity towards a range of serine and aspartic proteinases (Ing

& Roberts 1989, Liu & Hansen 1995, Mathialagan & Hansen 1996, Peltier et al. 2000a).

All of the species shown to possess US genes are in the Ruminantia and Suidae orders of

the Laurasiatheria superorder of eutherian mammals and all have epitheliochorial type of

placentation characterized by limited invasiveness and maintenance of epithelial layers in

endometrium and trophoblast. The epitheliochorial placenta has evolved from a more primitive

hemotropic placenta three separate times in evolution – in a subgroup of Laurasiatheria

consisting of cetartiodactyls, pigs, horses, and spiny anteaters, in a species of mole, and in lemurs

(Vogel 2005). The evolutionary pressure for development of an epitheliochorial placenta has

been attributed to increased efficiency of nutrient transport and gas exchange (Leiser et al. 1997),

greater maternal control of nutrient distribution to the conceptus (Mess & Carter 2007) and an

altered immunological relationship between mother and conceptus (Moffett & Loke 2006).

Page 53: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

53

The objectives of this study were to identify novel US genes in other species within and

outside of the Laurasiatheria superorder to determine whether the presence of the US gene is

restricted to species with epitheliochorial placentation and to evaluate whether US gene has been

subject to positive selection.

Materials and Methods

Data Base Queries to Identify Uterine Serpin Genes

The nucleotide and protein sequences for OvUS were used as query sequences to perform a

blastn search in the nucleotide collection (nr/nt) database of the National Center for

Biotechnology Information (NCBI) website to identify known and novel uterine serpin genes.

The Genbank accession number for the OvUS nucleotide sequence was NM_001009304.1 and

the accession number for the protein sequence was NP_001009304.1. Subsequently, a genomic

blast (blastn) of completed genomic database sequences in the NCBI was performed to identify

sequences that have similarities with the OvUS query sequence. Species examined were human

(Homo sapiens), rhesus macaque (Macaca mulata), chimpanzee (Pan troglodytes), dog (Canis

lupus familiaris), cow (Bos taurus) mouse (Mus musculus) rat (Rattus norvegicus), opossum

(Monodelphis domestica) and duck-billed platypus (Ornithorhynchus anatinus), as well as other

vertebrates like chicken (Gallus gallus), zebra fish (Danio rerio) and puffer fish (Tetraodon

nigroviridis). Additionally, the unfinished whole shotgun genomic sequences for the horse

(Equus cabalus - previously unfinished) and cat (Felis catus) were included in this search.

Reverse Transcription-Polymerase Chain Reaction (RT-PCR)

Uterine endometrial tissues were collected from pregnant mares at days 21 and 59 of

gestation, a pregnant bitch at day 60 of gestation, a pregnant queen at day 60 of gestation, as well

as from the intercaruncular area from ovariectomized ewes treated with 100 mg/ml of

progesterone for 60 days (Padua et al. 2005). Dog liver tissue and ovaries were also tested.

Page 54: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

54

Total RNA was isolated after homogenization with the TRI reagent (Sigma-Aldrich, St Louis,

MO) according to the manufacturer’s instructions. RNA purity and concentration were

determined spectrophotometrically.

The RT-PCR was performed using the SuperScript One Step RT-PCR kit with Platinum

Taq (Invitrogen, Carlsbad, CA). cDNA for OvUS, equine (Eq), Canine (Can) and Feline (Fe)

US were amplified from total RNA using primer sets for sheep (forward ACA GAT GCT TTA

CAG CCG GTC AGA; reverse TGA ACT TAA CAA CCA CCG GGA CCT), horse (forward

GCT GCA GAA ATG TCC CAC AGG AAA; reverse AGA GGA AAT CCC TGT GCT TCA

GGT) dog (forward ACC CAG TCT CGT CAT GGG AAG TTT; reverse TCA CGT CAT ACA

TCG CCT GTG TGT) and cat (forward TAC GAG ATC CAC AAC GCG CAC TAA; reverse

AAG TCA GTC ATC TGG GCC TTC ACA). To verify that PCR products were amplified from

RNA only, the SuperScript reverse transcriptase/Platinum Taq mix was omitted from control

reactions and an equivalent concentration of Taq DNA polymerase (Invitrogen) was added.

cDNA synthesis and pre-denaturation reactions were 1 cycle of 50oC for 30 min and 94

oC for 2

min respectively. PCR amplification reactions were performed in 40 cycles of 94oC for 15 sec

for denaturation, 50oC for 30 sec for annealing and 72

oC for 1 min extension. A final extension

cycle was performed at 72oC for 5 min.

Identification of cDNA for Equine Uterine Serpin Gene

The complete sequence of the EqUS gene was obtained by primer walking and the rapid

amplification of cDNAs ends (RACE) to obtain the 5’ and 3’ ends. The RACE reactions were

performed using RACE core system kits (Takara Bio Inc., Japan), following the manufacturer’s

recommendations. The set of primers used to obtain the full length of the 5’ end as well as the

oligo dT-3sites adaptor primer used to obtain the 3’end were gene specific primers and are

Page 55: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

55

Table 2-1. Primers used in RT-PCR procedure to obtain the full length coding sequence of the

equine uterine serpin gene. Amplicons are designated based on the Genbank

Accession Number of the sequences obtained from the RT-PCR. The full length was

obtained by overlapping seven sequences as shown in the diagram.

Amplicon Primer Sequence (5’ - 3’)

EU 810388 Fw GCTGCAATGTCCCACAGGAAA; Rv AGAGGAAATCCCTGTGCTTCAGGT

EU 810389 Fw CTCTGGATTGCTGCAGAAATG; Rv CTACTCAGCTATGGGGTTGAA

EU 810390 Fw CTCTGGATTGCTGCAGAAATG; Rv CTACTCAGCTATGGGGTTGAA

EU 810391 Fw TCGACCTCCAAAGAATCAGAGCGT; Rv AGGACACGTTTCCAGTGTAAGGCA

EU 810392 Fw AAACGTGTCCTTAGTCCTCGTGCT; Rv TTGAAGACTTGGCCCACAAAGAGC

EU 810393

(5’ RACE)

Rv /Phos/TGTCCCTAAAGGAGA

Fw TGTTCGAGGCTCTGTCAGTTGAGT; Rv TGTGCTTCAGGTGCCTCTGTCTAT

Fw GCTGCAGAAATGTCCCACAGGAAA; Rv ACTCAACTGACAGAGCCTCGAACA

EU 810394 (3’RACE)

Fw AAACGTGTCCTTAGTCCTCGTGCT

EU810388

EU810389

EU810390EU810391

EU810392

EU810393

EU810394

EqUS gene (1380 bp)5’ 3’

850 1380

819 1262

1 215

33322

40033

325 829 1245954

EU810388

EU810389

EU810390EU810391

EU810392

EU810393

EU810394

EqUS gene (1380 bp)5’ 3’

850 1380

819 1262

1 215

33322

40033

325 829 1245954

Page 56: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

56

shown in Table 2-1. Amplicons were electrophoresed on 3% (w/v) agarose gel containing 1

µg/ml ethidium bromide in Tris acetate-EDTA buffer (40 mM Tris-acetate, 2 mM EDTA pH

8.5), visualized on a ultraviolet transluminator and photographed with a Canon G-7 (Canon Inc,

Japan) power shot digital camera using the Digi-Doc-ITTM

imaging system (UVP, LLC, Upland,

CA). Amplicons were purified from the corresponding agarose gel bands using the QIAquick®

gel extraction kit (Qiagen Inc, Valencia, CA) according to the manufacturer’s instructions. DNA

concentration was determined spectrophotometrically and purity was assessed by electrophoresis

as previously described.

Sequencing of Amplicons

DNA amplicons were sequenced in both directions at the University of Florida DNA

Sequencing Core Laboratory using the ABI Prism® 3130 genetic analyzer (Applied Biosciences,

Foster City, CA). Briefly, ABI prism BigDye Terminators v.1.1 cycle sequencing reactions were

assembled in a total volume of 20 µl containing 500 ng DNA, 10 pmols primer, 4µl of BigDyer

terminator, and 5% (v/v) dimethyl sulfoxide. Sequencing reactions were performed on an ABI

GeneAmp 9700 thermal cycler (Applied Biosystems) with an initial denaturation at 95oC for 10

min and followed by 45 cycles of denaturation at 95oC for 30 sec, annealing at 55

oC for 20 sec

and extension at 60oC for 4 min. The excess of dye-labeled terminators were removed by using

MultoScreen® 96-well filtration system (Millipore, Bedford, MA). The purified extension

products were dried in SpeedVac® (ThermoSavant, Holbrook, NY) and then suspended in Hi-di

formamide. Sequencing reactions were performed using POP-7 sieving matrix on 50-cm

capillaries in the ABI Prism® 3130 Genetic Analyzer and were analyzed by ABI Sequencing

Analysis software v. 5.2 and KB Basecaller.

Analyses of the deduced amino acid sequence were conducted using the program Scan

Prosite at the ExPASy Molecular Biology Server (http://ca.expasy.org). The predicted signal

Page 57: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

57

peptide cleavage sites were obtained from SignalP 3.0 (Bendtsen et al. 2004) and predicted

glycosylation sites were obtained from NetNGlyc 1.0 both from the ExPASy Molecular Biology

Server.

Detection of Equine Uterine Serpin by Western Blotting

Ovariectomized pony mares were treated daily with progesterone (150 mg/ml), estradiol

benzoate (10 mg/ml), progesterone plus estradiol benzoate or vehicle as a control. After 28 days,

uterine fluids were collected by flushing the uterus with 0.9% (w/v) NaCl (Hansen et al. 1985).

Samples were concentrated using Centricon plus-20 devices (Millipore Corporation, Billerica,

MA). Aliquots of 20 µl of concentrated mare samples were separated under reducing conditions

using one-dimensional discontinuous sodium dodecyl sulfate-polyacrylamide gel electrophoresis

at 4-15% (w/v) polyacrylamide, Tris-HCl gels (Bio-Rad, Hercules, CA). Aliquants of 0.5 µg of

protein from uterine fluid of an ovariectomized ewe treated with 100 mg/ml of progesterone for

60 days and from the control serpin ovalbumin (OVA) were used as positive and negative

control, respectively. Western blotting was performed as described elsewhere (Padua et al.

2005) with minor modifications. Briefly, the monoclonal anti-OvUS (HL-218; Leslie et al.

1990) and IgG1 (MOPC21) (Sigma-Aldrich) (1:32,000 dilution) were used as a primary and

control antibodies, respectively. The second antibody used was ECLTM

anti-mouse IgG

peroxidase-linked; 1:25,000 dilution (Amersham-GE Healthcare Bio-Sciences Corp, Piscataway,

NJ) and the ECL plus western blotting detection kit (Amersham) was used as a detection system

according to the vendor’s instructions.

Amino Acid Sequence Alignments and Analysis of Phylogenetic Tree

ClustalW (Chenna et al. 2003) was used to obtain the amino acid alignments which were

then prepared for publication using the Boxshade multiple alignments designer program version

3.21 (http://www.ch.embnet.org).

Page 58: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

58

The phylogenetic tree that includes corticosteroid binding globulin (CBG) was constructed

using the Neighbor Joining (NJ) method (Saitou & Nei 1987) of the Molecular Evolutionary

Genetics Analysis (MEGA4) software, version 4.0 (Tamura et al. 2007). The NJ tree was based

on the distance calculation under the Jones-Taylor-Thornton (JTT) matrix substitution model

(Jones et al. 1992) after removing position containing gaps (complete deletion option).

The NJ method (Saitou & Nei 1987) was also used to construct the phylogenetic tree for

the US group of proteins based on a gamma shape parameter (α) corrected JTT distance matrix

(Jones et al. 1992). The plotted tree was obtained by using the drawgram software of PHYLIP

(Phylogeny Inference Package) version 3.5c (Felsenstein 1989, 1993). The gamma shape

parameter and amino acid frequencies were estimated from the data using the Tree-Puzzle

software by maximum likelihood analysis for amino acids (Strimmer & von Haeseler 1996,

Schmidt et al. 2002) and the reliability of the trees was estimated by the bootstrap test with 1,000

repetitions (Felsenstein 1985).

Analysis of Ratio of Non-Synonymous and Synonymous Substitutions

The ratio of non-synonymous and synonymous substitutions (dN/dS; termed ω) of codons

was used to determine whether the pressure of selection induces purifying or positive selection at

specific areas of the sequence (Yang & Nielsen 2002). Sites with ω values significantly higher

or lower than 1 are an indication of positive (Darwinian) or purifying selection, respectively.

The codon-based substitution model in the CODEML program of the phylogenetic analysis by

maximum likelihood (PAML, version 4.0b) software package (Yang 2007) was used to identify

the effect of selective pressures on the uterine serpin genes. Aligned sequences were tested using

different models of variable dN/dS ratios among sites which includes M0 (null model), M1

(nearly neutral, where ω>1 is not allowed), M2 (positive selection, ω>1 is allowed), M7 (ω>1 is

Page 59: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

59

not allowed, beta distribution) and M8 (ω>1 is allowed, beta distribution) (Yang et al. 2000,

Bielawski & Yang 2003, Wong et al. 2004). The likelihood ratio test (LRT) was obtained by

twice the difference in log likelihood (2L) from two different models and the significance of

the test was estimated by using the 2 distribution with degrees of freedom calculated from the

estimated parameters (Anisimova et al. 2001, Yang & Nielsen 2002). The models M1 and M7

were tested against models M2 and M8, respectively. Additionally, the Selecton server (Server

for the identification of site-specific positive and purifying selection, version 2.4) (Stern et al.

2007) was used to identify ω at each codon site based on an empirical Bayesian method.

Aligned coding sequences in FASTA format for uterine serpins were tested with the M8 model,

(extra category ωs ≥ 1, beta distribution and positive selection is allowed). The significance of ω

scores were also tested by using LRT test which compares the positive selection model (M8) and

the null M8a model (extra category ωs set to 1); allowing only for purifying and neutral selection,

respectively.

Results

Identification of Coding Sequences of Known and New Uterine Serpins Using Blastn

Uterine serpin sequences have been previously described for goat (Capra hircus), cow (Bos

taurus) and pig (Sus scrofa). Results from the blastn search in the nucleotide collection database

of the NCBI identified an additional uterine serpin coding sequence that has been submitted to

Genbank for water buffalo (Bubalus bubalis, Genbank: DQ 661648.1). This is a species with

epitheliochorial placenta that is closely related to the cow. The blast search also identified a

gene in the dog (Canis lupus familiaris, Genbank: XM_850115.1) with similarity to the query

sequence, OvUS. The dog is a member of Laurasiatheria but, unlike other species with uterine

serpin genes, the dog has an endotheriochorial placenta. Currently, XM_850115.1 is annotated

as one of the two CBG genes in the dog. Phylogenetic analysis shows, however, that while the

Page 60: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

60

other predicted dog CBG (Genbank: XP_547960.2) is clustered together with CBG proteins

present in other species such as human, chimpanzee, squirrel and rhesus monkey, pig, sheep,

mouse and hamster, XP_855208.1 clusters with the US (Figure 2-1).

A genomic blastn search of complete genomic sequences identified uterine serpin genes for

the cow and dog. No US genes were identified in the order Anthropoidea, represented by

human, rhesus monkey and chimpanzee, or in Rodentia, represented by the mouse and rat.

Likewise, the US gene was no identified either in the opossum, a species with a primitive

epitheliochorial placentation within the Marsupialia order, in the duck-billed platypus, an egg-

laying mammal that belongs to the Monotremata order, or in any of the other vertebrates with

complete genome sequences (chicken, puffer and zebra fish).

A genomic blast was also performed using OvUS as query for the whole shotgun genomic

sequence of the horse, which was incomplete at the time of analysis. There were four stretches

of nucleotides of 664, 232, 72 and 86 nucleotides in length (Genbank: gbAAWR02019930) that

matched with the coding sequence of OvUS (68,75, 86 and 79% identity, respectively). These

sequences were all localized on chromosome 24 and are flanked at the 5’ side by SERPINA11.

A genomic blast search of the whole shotgun genomic sequence of the cat using OvUS as

the query identified one stretch of 198 nucleotides in length (Genbank: gbACBE01215383.1)

that matched with the coding sequence of OvUS with 77% of identity. However, using CanUS

as the query sequence identified four stretches of nucleotides of 525, 273, 146 and 199

nucleotides in length (same accession number) that matched the coding sequence of CanUS with

82, 82, 80 and 89% identity, respectively.

Page 61: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

61

Figure 2-1. Identification of an incorrectly annotated dog corticosteroid binding globulin (CBG)

as an uterine serpin (US). The sequence incorrectly annotated as CBG [Genbank:

XP_855208.1] clusters with the US while another dog CBG [Genbank:

XP_547960.2] clusters with CBG in other species. The Neighbor Joining tree was

constructed using the Molecular Evolutionary Genetics Analysis (MEGA4)

software. The percentage of replicate trees in which the associated taxa clustered

together in the bootstrap test (1000 replicates) is shown next to the branches. The

tree is drawn to scale, with branch lengths in the same units as those of the

evolutionary distances used to infer the phylogenetic tree. The evolutionary

distances were computed using the Jones-Taylor-Thornton (JTT) matrix-based

method and are in the units of the number of amino acid substitutions per site. All

positions containing gaps and missing data were eliminated from the dataset

(Complete deletion option).

CBG-Human (AAB59523.1)

CBG-Chimpanzee (XP 510143.2)

CBG-Rhezus monkey (XP 001098128.1)

CBG-Squirrel monkey (AAC60614.1)

CBG-Dog (XP 547960.2)

CBG-Pig (AAG45431.1)

CBG-Sheep (CAA52000.1)

CBG-Mouse (NP 031644.1)

CBG-Hamster (AAA37065.1)

CanUS (XP 855208.1)

PoUS-1 (AAA31137.1)

PoUS-2 (NP 999010.1)

OvUS (NP 001009304.1)

CapUS (DAA05634.1)

BoUS (NP 777222.1)

BuUS (ABG56442.1)

100

100

99

99

100

99

89

100

100

99

95

100

100

0.1

Page 62: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

62

Uterine Serpin Gene Organization in the Bovine and Canine

The BoUS gene is localized in chromosome 21 for the cow and is organized in 6 exons,

with two small untranslated regions (UTR) contained in exons 1 and 6 respectively. The CanUS

is localized in chromosome 8 and also contains 6 exons but with no UTR present. The BoUS

gene clusters with SERPINA1, SERPINA11 and SERPINA12. The CanUS gene clusters with

SERPINA1, SERPINA9, SERPINA11, and SERPINA12. There are variations in the length of

exons and introns between BoUS and CanUS gene as indicated in Table 2-2. However, the size

of exon 3 is identical in both species.

Characteristics of CanUS and Expression in Tissues

The gene for CanUS (Genbank: XM_850115) is 1416 bp in length that encodes a

polypeptide of 471 amino acids with a theoretical isoelectric point of 9.43 and predicted

molecular mass of 54.25 kDa. The CanUS protein sequence lacks a signal peptide suggesting

the intracellular localization of the protein. Using XM_850115 primers were designed for RT-

PCR. Amplicons were obtained from endometrium of a bitch at 60 days of gestation, but there

was no expression in the liver (Figure 2-2). In addition, transcripts for CanUS were identified in

the ovary by RT-PCR (data not shown).

Uterine Expression and Amino Acid Sequence of EqUS

Using primers designed from the EqUS sequences identified in the blast search, expression

of EqUS in endometruim from a pregnant mare at Day 21 (data not shown) and Day 59 of

gestation was identified (Figure 2-3A). The complete sequence of EqUS was obtained by

performing RT-PCR, 5’RACE and 3’ RACE using endometrial cDNA of a mare at Day 59 of

gestation. In this manner, seven overlapping sequences were obtained (Genbank: EU810388,

EU810389, EU810390, EU810391, EU810392, EU810393, EU810394) with a total length of

1380 bp that includes 9 and 108 nucleotides for the 5’ and 3’ UTR of the gene, respectively. The

Page 63: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

63

Table 2-2. Exon and intron sizes for the uterine serpin gene of the cow and dog.

Cow exon1

(UTR) exon2 intron2 exon3 intron3 exon4 intron4 exon5

exon6

(UTR) Total bp

16 637 3204 268 1808 148 2205 288 65 8639

Dog exon1 intron1 exon2 intron2 exon3 intron3 exon4 intron4 exon5 Total bp

210 1863 148 1580 268 876 666 5034 124 10768

Page 64: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

64

Figure 2-2. Representative electrophoretogram of amplicons obtained by reverse transcriptase-

polymerase chain reaction (RT-PCR) of RNA fron canine tissue and canine uterine

serpin primers. RNA was isolated from dog liver or endometruim from a pregnant

bitch at Day 60 of gestation. Control reactions excluded reverse transcriptase (w/o

RT). Arrows on the left represent size of standards while the arrow on the right

shows the expected amplicon size.

Pre

gn

an

t b

itc

h w

/o R

T

Pre

gn

an

t b

itc

h w

/o R

T

DE

PC

-wa

ter

Do

g l

ive

r –w

/o R

T

DN

A l

ad

de

r

(1 K

b)

Pre

gn

an

t b

itc

h

Do

g l

ive

r

Do

g l

ive

r –w

/o R

T

Do

g l

ive

r

Pre

gn

an

t b

itc

h

387 bp396 bp

344 bp

Pre

gn

an

t b

itc

h w

/o R

T

Pre

gn

an

t b

itc

h w

/o R

T

DE

PC

-wa

ter

Do

g l

ive

r –w

/o R

T

DN

A l

ad

de

r

(1 K

b)

Pre

gn

an

t b

itc

h

Do

g l

ive

r

Do

g l

ive

r –w

/o R

T

Do

g l

ive

r

Pre

gn

an

t b

itc

h

387 bp396 bp

344 bp

Page 65: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

65

Figure 2-3. Expression and secretion of equine uterine serpin. Panel A represents an

electrophoretogram of amplicons obtained by reverse transcriptase-polymearse

chain reaction (RT-PCR) of either RNA from endometruim of an ovariectomized

ewe treated with progesterone for 60 days (P4-treated ewe) or RNA from

endometruim of a pregnant mare at Day 59 of gestation. Control reactions excluded

reverse transcriptase (w/o RT). Arrows show a size of the standard and expected

size of the amplicons for the sheep and mare respectively. Panel B represents

results of a western blotting experiment using antibody to OvUS and either uterine

fluid from an ovariectomized ewe treated with progesterone for 60 days, uterine

flushings from ovariectomized mares treated for xx days with either vehicle,

progesterone (P4), estradiol benzoate (E), or the combination (P4+E), or

endometrial tissue from a pregnant mare at 59 days of gestation. As negative

control, the serpin ovalbumin (OVA) was also subjected to western blotting.

500 bp500 bp467 bp467 bp436 bp436 bp

AD

EP

C-w

ate

r

Ew

e (

P4)

DN

A lad

der

(1 K

b)

Ew

e (

P4)

Pre

gn

an

t m

are

Pre

gn

an

t m

are

Ew

e (

P4)

w/o

RT

Pre

gn

an

t m

are

w/o

RT

500 bp500 bp467 bp467 bp436 bp436 bp

AD

EP

C-w

ate

r

Ew

e (

P4)

DN

A lad

der

(1 K

b)

Ew

e (

P4)

Pre

gn

an

t m

are

Pre

gn

an

t m

are

Ew

e (

P4)

w/o

RT

Pre

gn

an

t m

are

w/o

RT

Pre

gn

an

t m

are

ST

D

OV

A

Ew

e (

P4)

Mare

(veh

icle

)

Mare

(E

)

Mare

(P

4+

E)

Mare

1 (

P4)

Mare

2 (

P4)

Uterine fluid Uterine flushings Endometruim

250 kD

150 kD

20 kD

10 kD

37 kD

50 kD

75 kD

100 kD

B

Pre

gn

an

t m

are

ST

D

OV

A

Ew

e (

P4)

Mare

(veh

icle

)

Mare

(E

)

Mare

(P

4+

E)

Mare

1 (

P4)

Mare

2 (

P4)

Uterine fluid Uterine flushings Endometruim

250 kD

150 kD

20 kD

10 kD

37 kD

50 kD

75 kD

100 kD

B

Page 66: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

66

sequence was submitted to NCBI GenBank as TPA (Third Party Annotation, GenBank accession

number: BK 006618). The complete coding sequence of the EqUS was 1263 bp which is 100%

identical to genomic sequence.

The deduced amino acid sequence is shown in Figure 2-4. The gene encodes a polypeptide

of 421 amino acids with a theoretical isoelectric point of 9.49 and predicted molecular mass of

48.79 kDa. Analysis of the EqUS protein sequence showed a predicted signal peptide, most

likely between positions (Cys25

) and (Glu26

) and two potential glycosylated sites at (Asn212

) and

(Asn246

).

Endometrial Secretion of EqUS

Uterine flushings from ovariectomized mares treated with steroids were examined by

western blotting using an antibody to OvUS to determine if EqUS was secreted into uterine

lumen and, if so, whether as for other uterine serpins secretion was under control of

progesterone. The western blot is shown in Figure 2-3B. Immunoreactive protein bands were

obtained from uterine flushes from ovariectomized mares treated with estradiol benzoate,

progesterone or progesterone and estrogen. There was an absence of immunoreactive protein in

flushes of ovariectomized mares treated with vehicle. The molecular weight of the

immunoreactive EqUS was similar to OvUS identified by western blotting of uterine fluid from a

progesterone treated ewe (55,000-57,000). The immunoreactive EqUS is larger than the

predicted molecular mass of 48.79 kDa, suggesting the protein is glycosylated. Multiple

immunoreactive bands were also seen for supernatant from lysed endometrial tissue of a mare at

Day 59 of gestation. The variety of lower molecular weight bands probably represent proteolytic

products as has been described for OvUS (Moffatt et al. 1987, Leslie et al. 1990, Peltier et al.

2000a, Leslie & Hansen 1991). No immunoreactive bands were identified for the control serpin

OVA (Figure 3B) or when IgG replaced the primary antibody (data not shown).

Page 67: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

67

Lack of Expression of the Uterine Serpin Gene in the Pregnant Cat

RT-PCR experiments with primers designed for a putative FeUS sequence identify by

blastn against CanUS (525 bp in length) failed to yield an amplicon from cDNA obtained from

pregnant endometruim of a pregnant queen at Day 60 of gestation (data not shown). Moreover,

stop codons were identified after translation of the deduced amino acid sequences for at least two

of the four stretches of nucleotides identified by blastn. Depending upon the frame used for

codon identification, the 525 bp sequence had 1-10 stop codons, the 272 bp sequence had 1-6

stop codons, the 199 bp sequence had from 0-3 stop codons, and the 146 bp sequence had from

0-4 stop codons. Similarly, the 198 bp sequence identified in cat by genomic blast search using

OvUS had 1-4 stop codons.

Amino Acid Sequence Conservation

The ClustalW amino acid sequence alignments of uterine serpins are shown in Figure 2-4.

There is a 9-amino acid insert present in OvUS and CapUS (KINLKHLLP) that is not present in

uterine serpins from other species. Likewise, the water buffalo US (BuUS) contains a 13-amino

acid insert (MNAKEVPVVVKVP) that is highly conserved with 39-amino acid insert for BoUS.

There is also a 3-amino acid insert in the same relative position for the PoUS-1, PoUS-2, CanUS

and EqUS. This insert is conserved between the PoUS-1 and -2, but the insert for CanUS and

EqUS is not conserved with any other species.

The CanUS sequence is unique among uterine serpins in that there are two start codons

upstream from the start codons for other uterine serpins (Figure 2-4) so that the CanUS

polypeptide could be as much as 48-amino acid longer than other serpin sequences and not

contain a signal peptide.

There is a KEVPVVVK motif located near the putative P1-P1’ site originally described for

the BoUS that has been postulated to be a pepstatin-like domain (Mathialagan & Hansen 1996).

Page 68: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

68

Figure 2-4. Amino acid sequence alignment of the uterine serpins using the ClustalW

algorithm. Sites of conservation are shown in shaded columns using the Boxshade

software. Black-shaded columns show identical amino acids and grey-shaded

columns represent similar amino acids. Distinctive amino acid motifs limited to a

subset of uterine serpins are marked with red rectangles. Start codons are shown by

red arrows.

|AAA31137.1-PoUS-1| 1 ------------------------------------------------MSHGKMPLVLSLVLILCGLFNS

|NP_999010.1-PoUS-2| 1 ------------------------------------------------MSHGKMPLVLSLVLILCGLFNS

|NP_001009304.1 OvUS| 1 ------------------------------------------------MSHRRMQLALSLVFILCGLFNS

|DAA05634.1-CapUS| 1 ------------------------------------------------MSHRRMQLALSLVFILCGLFNS

|NP_777222.1-BoUS| 1 ------------------------------------------------MSHGRMNLALSLVFILCGLFNS

|ABG56442.1-BuUS| 1 ------------------------------------------------MSHGRMNLALSLVLILCGLLNS

|XP_855208.1-CanUS| 1 MEKGWERLSKQGPGKEADDISHKASSSDMRVRWEARTKEEETPWIAAEMSHRKMHLALSLALILCGLFNS

|BK006618-EqUS| 1 ------------------------------------------------MSHRKMHLALSLVLTLCGLLNS

CONSENSUS 1 MSH kMqLaLSLvliLCGLfNS |AAA31137.1-PoUS-1| 23 ISCEKQQTSPKTITPVSFKRIAALSQKMEANYKAFAQELFKTLLIEDPRKNMIFSPVSISISLATLSLGL

|NP_999010.1-PoUS-2| 23 ISCEKQQTSPKTITPVSFKRIAALSQKMEANYKAFAQELFKTLLIEDPRKNMIFSPVSISISLATLSLGL

|NP_001009304.1-OvUS| 23 IFCEKQQHSQQHANLVLLKKISAFSQKMEAHPKAFAQELFKALIAENPKKNIIFSPAAMTITLATLSLGI

|DAA05634.1-CapUS| 23 IFSEKQQHSQQHANLVLLKKISAFSQKMEAHPKAFAQELFKALIAEDPKKNIIFSPVAMTITLATLSLGI

|NP_777222.1-BoUS| 23 IFCEKQQHSQKHMNLVLLKKISALSQKMEAHPKDFAQELFKALIIEDPRKNIIFSPMAMTTTLATLSLGI

|ABG56442.1-BuUS| 23 IFCEKQQHSQKHVNLVLLKKISALSQKMEAHPKDFAQELFKALIIEAPRKNIIFSPMAMTTTLATLSLGI

|XP_855208.1-CanUS| 71 IFCQVKQRSKWSIYPVSSWEVSPVHHKIEAGNKAFAHKLFKTLLMEHPRKNIIFSPLSISASFAMLSLGT

|BK006618-EqUS| 23 IFCETQPNPKRDPHPVSSRKTSPPSHETDTDHEAFAYKLFEALSVEYHRKNLIFSPESISTALAMLSLGT

CONSENSUS 71 Ifcekqqhsqkhin V lkkisalsqkmeahpkaFAqeLFkaLiiEdprKNiIFSPv isitlAtLSLGi

|AAA31137.1-PoUS-1| 160 KMDIQMIDFKDKEKTKKAINQFVADKIDKKAKNLITHLDPQTLLCLVNYIFFKGILERAFQTNLTKKEDF

|NP_999010.1-PoUS-2| 163 KVDIQMIDFKDKEKTKKAINQFVADKIDKKAKNLITHLDPQTLLCLVNYVFFKGILERAFQTNLTKKEDF

|NP_001009304.1-OvUS| 160 KMDIQMIDFSDTEKAKKAISHHVAEKTHTKIRDLITDLNPETILCLVNHIFFKGILKRAFQPNLTQKEDF

|DAA05634.1-CapUS| 160 KMDIQMIDFSDTEKAKKAISHHVAEKTHTKITDLITDLNPETILCLVNHIFFKGILKRAFQPNLTQKEDF

|NP_777222.1-BoUS| 160 GMDIQMIDFTDIEKAKKTISHHVAEKTHTKITNLITDLNPETILCLVNHIFFKGILKRAFQPKLTQKEVF

|ABG56442.1-BuUS| 160 GMDIQMIDFTDTEKARKTISHHVAEKTHTKISDLITDLNPETILCLVNHVSLKGILKRAFQPELTQKEDF

|XP_855208.1-CanUS| 211 DVRARMIDFRDVVKTKKQINHFVAEKTHKRTKELITSLNPHTFLFLVDYVFFKGTWEMAFHSNLMHKEDF

|BK006618-EqUS| 163 EVEAQVADFRHRGIAKEQINQFVAQRLAHRIEEVVTSLHPHTFLFLLNYIFFKGVWEVAFQTRFTQKENF

CONSENSUS 211 kmdiqmiDF d ekakkaI h VAekthtkikdliTdLnPeTiLcLvn iffKGil rAFqpnltqKEdF

|AAA31137.1-PoUS-1| 93 RSATRTNAIDVLDVALKNLAVMLMAQAPTALLEIVHELVN-RTAKHQDIL-IDR-TEMNQMFLKEIDRYI

|NP_999010.1-PoUS-2| 93 RSATRTNAIDVLERDLRNLRVWDKHQALQHLVEMLHELEKKKQLKHKDIFFIDRNKKMNQMFLKEIDRVY

|NP_001009304.1-OvUS| 93 KSTMSTNHPEDLELELK---LLDAHKCLHHLVHLGRELVKQKQLRHQDILFLNSKMMANQMLLHQIRKLQ

|DAA05634.1-CapUS| 93 KSTMSTNHPEDLELELK---LLDAHKCLHHLLHLGRELVKQKQLKHQDILFLNSKMMANQMLLHQISKLQ

|NP_777222.1-BoUS| 93 KSTMRTHHPEDLKLEPK---LLDVHKYLQPLVHVGRELVKQKVLKHQHILFINRKMMVNQMLLQQISKLQ

|ABG56442.1-BuUS| 93 KSTMNAHHPEDLKLEPK---LLDVHKYLQTLVHVEHELVKQKLLKHQHILFINSKMVVNQMLLQQIGKLQ

|XP_855208.1-CanUS| 141 RSTTLTNLLEGLGFDLKVIKVWDVHHGFQSVIQMLKQLNRAGHLMHRDMLFIDSNRKINSPFLWDTQAMY

|BK006618-EqUS| 93 RSTTLSNLVAGLGFDLQRIRAWEVHTSFQRLVQTLNELDRQRHLKHRDFLFIDINRRIKPKFLQETERLY

CONSENSUS 141 rSt tnhpedLdlelk l lldvhk lqhlvhivheLvkqk lkHqdilfi skmmmnqm Lkqi klq

|AAA31137.1-PoUS-1| 1 ------------------------------------------------MSHGKMPLVLSLVLILCGLFNS

|NP_999010.1-PoUS-2| 1 ------------------------------------------------MSHGKMPLVLSLVLILCGLFNS

|NP_001009304.1 OvUS| 1 ------------------------------------------------MSHRRMQLALSLVFILCGLFNS

|DAA05634.1-CapUS| 1 ------------------------------------------------MSHRRMQLALSLVFILCGLFNS

|NP_777222.1-BoUS| 1 ------------------------------------------------MSHGRMNLALSLVFILCGLFNS

|ABG56442.1-BuUS| 1 ------------------------------------------------MSHGRMNLALSLVLILCGLLNS

|XP_855208.1-CanUS| 1 MEKGWERLSKQGPGKEADDISHKASSSDMRVRWEARTKEEETPWIAAEMSHRKMHLALSLALILCGLFNS

|BK006618-EqUS| 1 ------------------------------------------------MSHRKMHLALSLVLTLCGLLNS

CONSENSUS 1 MSH kMqLaLSLvliLCGLfNS |AAA31137.1-PoUS-1| 23 ISCEKQQTSPKTITPVSFKRIAALSQKMEANYKAFAQELFKTLLIEDPRKNMIFSPVSISISLATLSLGL

|NP_999010.1-PoUS-2| 23 ISCEKQQTSPKTITPVSFKRIAALSQKMEANYKAFAQELFKTLLIEDPRKNMIFSPVSISISLATLSLGL

|NP_001009304.1-OvUS| 23 IFCEKQQHSQQHANLVLLKKISAFSQKMEAHPKAFAQELFKALIAENPKKNIIFSPAAMTITLATLSLGI

|DAA05634.1-CapUS| 23 IFSEKQQHSQQHANLVLLKKISAFSQKMEAHPKAFAQELFKALIAEDPKKNIIFSPVAMTITLATLSLGI

|NP_777222.1-BoUS| 23 IFCEKQQHSQKHMNLVLLKKISALSQKMEAHPKDFAQELFKALIIEDPRKNIIFSPMAMTTTLATLSLGI

|ABG56442.1-BuUS| 23 IFCEKQQHSQKHVNLVLLKKISALSQKMEAHPKDFAQELFKALIIEAPRKNIIFSPMAMTTTLATLSLGI

|XP_855208.1-CanUS| 71 IFCQVKQRSKWSIYPVSSWEVSPVHHKIEAGNKAFAHKLFKTLLMEHPRKNIIFSPLSISASFAMLSLGT

|BK006618-EqUS| 23 IFCETQPNPKRDPHPVSSRKTSPPSHETDTDHEAFAYKLFEALSVEYHRKNLIFSPESISTALAMLSLGT

CONSENSUS 71 Ifcekqqhsqkhin V lkkisalsqkmeahpkaFAqeLFkaLiiEdprKNiIFSPv isitlAtLSLGi

|AAA31137.1-PoUS-1| 160 KMDIQMIDFKDKEKTKKAINQFVADKIDKKAKNLITHLDPQTLLCLVNYIFFKGILERAFQTNLTKKEDF

|NP_999010.1-PoUS-2| 163 KVDIQMIDFKDKEKTKKAINQFVADKIDKKAKNLITHLDPQTLLCLVNYVFFKGILERAFQTNLTKKEDF

|NP_001009304.1-OvUS| 160 KMDIQMIDFSDTEKAKKAISHHVAEKTHTKIRDLITDLNPETILCLVNHIFFKGILKRAFQPNLTQKEDF

|DAA05634.1-CapUS| 160 KMDIQMIDFSDTEKAKKAISHHVAEKTHTKITDLITDLNPETILCLVNHIFFKGILKRAFQPNLTQKEDF

|NP_777222.1-BoUS| 160 GMDIQMIDFTDIEKAKKTISHHVAEKTHTKITNLITDLNPETILCLVNHIFFKGILKRAFQPKLTQKEVF

|ABG56442.1-BuUS| 160 GMDIQMIDFTDTEKARKTISHHVAEKTHTKISDLITDLNPETILCLVNHVSLKGILKRAFQPELTQKEDF

|XP_855208.1-CanUS| 211 DVRARMIDFRDVVKTKKQINHFVAEKTHKRTKELITSLNPHTFLFLVDYVFFKGTWEMAFHSNLMHKEDF

|BK006618-EqUS| 163 EVEAQVADFRHRGIAKEQINQFVAQRLAHRIEEVVTSLHPHTFLFLLNYIFFKGVWEVAFQTRFTQKENF

CONSENSUS 211 kmdiqmiDF d ekakkaI h VAekthtkikdliTdLnPeTiLcLvn iffKGil rAFqpnltqKEdF

|AAA31137.1-PoUS-1| 93 RSATRTNAIDVLDVALKNLAVMLMAQAPTALLEIVHELVN-RTAKHQDIL-IDR-TEMNQMFLKEIDRYI

|NP_999010.1-PoUS-2| 93 RSATRTNAIDVLERDLRNLRVWDKHQALQHLVEMLHELEKKKQLKHKDIFFIDRNKKMNQMFLKEIDRVY

|NP_001009304.1-OvUS| 93 KSTMSTNHPEDLELELK---LLDAHKCLHHLVHLGRELVKQKQLRHQDILFLNSKMMANQMLLHQIRKLQ

|DAA05634.1-CapUS| 93 KSTMSTNHPEDLELELK---LLDAHKCLHHLLHLGRELVKQKQLKHQDILFLNSKMMANQMLLHQISKLQ

|NP_777222.1-BoUS| 93 KSTMRTHHPEDLKLEPK---LLDVHKYLQPLVHVGRELVKQKVLKHQHILFINRKMMVNQMLLQQISKLQ

|ABG56442.1-BuUS| 93 KSTMNAHHPEDLKLEPK---LLDVHKYLQTLVHVEHELVKQKLLKHQHILFINSKMVVNQMLLQQIGKLQ

|XP_855208.1-CanUS| 141 RSTTLTNLLEGLGFDLKVIKVWDVHHGFQSVIQMLKQLNRAGHLMHRDMLFIDSNRKINSPFLWDTQAMY

|BK006618-EqUS| 93 RSTTLSNLVAGLGFDLQRIRAWEVHTSFQRLVQTLNELDRQRHLKHRDFLFIDINRRIKPKFLQETERLY

CONSENSUS 141 rSt tnhpedLdlelk l lldvhk lqhlvhivheLvkqk lkHqdilfi skmmmnqm Lkqi klq

Page 69: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

69

Figure 2-4. Continued

|AAA31137.1-PoUS-1| 230 FVNEKTIVQVDMMRKTERMIYSRSEELLATMVKIPCKENASIILVLPDTGKFNFALKEMAAKRARLQKTN

|NP_999010.1-PoUS-2| 233 FVNEKTIVQVDMMRKTERMIYSRSEELLATMVKMPCKENASIILVLPDTGKFDFALKEMAAKRARLQKTN

|NP_001009304.1-OvUS| 230 FLNDKTKVQVDMMRKTEQMLYSRSEELFATMVKMPFKGNVSLILMLPDAGHFDNALKKLTAKRAKLQKIS

|DAA05634.1-CapUS| 230 FLNDKTKVQVDMMRKTEQMLYSRSEELFATMVKMPFKGNVSLILMLPDAGHFDNALKKLTAKRAKLQKTS

|NP_777222.1-BoUS| 230 FVNDQTKVQVDMMRKTERMLYSRSEELHATMVKMPCKGNVSLTLMLPDAGQFDTDLKKMTAKRAKLQKIS

|ABG56442.1-BuUS| 230 LLDDKTKVQVDMMRKTERMLYSRSEELHTTMVKMPCKGNVSLILMLPDAGQFDTALKKVTAKRAKLQKIS

|XP_855208.1-CanUS| 281 FVDKHTTVPVDMIWKTGQMIYSRSEELFATMVKIPFVGNMSIVLVLPDVGQPDSAVKEIVVQRATLLQSS

|BK006618-EqUS| 233 FLEDNTSVQVHMMRKTERMIYSRAEHLFATIVKLPYTGNVSLVLVLPDAGRFDFVAKELAARRARPLQSR

CONSENSUS 281 fvndkTkVqVdMmrKTerMiYSRsEeLfaTmVKmPckgNvSliLvLPDaGkfd alK mtakRAklqkts

|AAA31137.1-PoUS-1| 300 DFRLVHLVVPKIKDNLQDRFKHLLP---------KIGINDIFTTKAVTWNTTGTS--TILEAVHHAVIEV

|NP_999010.1-PoUS-2| 303 ELQIGALSCAQDQDHLQDRFKHLLP---------KIGINDIFTTKAVTWNTTRTS--TILEAVHHAVIEV

|NP_001009304.1-OvUS| 300 NFRLVHLTLPKFKITFDINFKHLLPKINLKHLLPKIDPKHTLTTTASSQHVTLKAPLPNLEALHQVEIEL

|DAA05634.1-CapUS| 300 NFRLVHLTLPKFKITFEINFKHLLPKINLKHLLPKIDPKHTLTTTASSQDVTLKAPLPNLEALHQVEIEL

|NP_777222.1-BoUS| 300 DFRLVRLILPKLKISFKINFKHLLP---------KIDPKHILTATAISQAITSKAPLPNLEALHQAEIEL

|ABG56442.1-BuUS| 300 DFRLVRLTLPKLKISFKINFKHLLP---------KIGPKHTLTTTAISQAITLKAPLPNLEALHQAEIEL

|XP_855208.1-CanS 351 NMRWVHIIMPKFKISSKIDLKKILP---------KMGISNVFTTGANFSGITKEDFPTIFEAMHEATMEV

|BK006618-EqUS| 303 DTRLVHLILPKFKISSRIDLNRLLP---------KVGIEDIFSRRANFSGITDETFPTIFEAIHEARLEV

CONSENSUS 351 dfrlvhl lpkfkisfkinfkhlLP Kig khi tttAvsqaiT kapl lEAlHqaeiEv

|AAA31137.1-PoUS-1| 359 KEDGLTKNAAKDKD-FWKVP---------- -----------------------------VDKKEVPVVV

|NP_999010.1-PoUS-2| 362 KEDGLTKNAAKDKD-FWKVP---------- -----------------------------VDKKEVPVVV

|NP_001009304.1-OvUS| 370 SEHALTTDTAIHTDNLLKVP---------- -----------------------------ANTKEVPVVV

|DAA05634.1-CapUS| 370 SEHALTTDTAIHTDNLLKVP---------- -----------------------------VNAKEVPVVV

|NP_777222.1-BoUS| 361 SEHALTVDTAIHTDNLLKVPVKAKEVPAVV KVPMKAKEVPAVVKVPMNTKEVPVVVKVPMNTKEVPVVV

|ABG56442.1-BuUS| 361 NEHALTVDTAIHTDNLLKVP---------- ----------------MNAKEVPVVVKVPTNAKEVPVDV

|XP_855208.1-CanUS| 412 SKEGKMDTSKDMDSRNTIAC---------- -----------------------------HTYTATPLVV

|BK006618-EqUS| 364 NEKG-VIKAAAEDVHAKRAH---------- -----------------------------HAPHADATVV

CONSENSUS 421 sehglt dtaihtdnllkvp vn kevpvvV

|AAA31137.1-PoUS-1| 388 KFDRPFFLFVEDEITRRDLFVAKVFNPKTE

|NP_999010.1-PoUS-2| 391 KFDRPFFLFVEDEITRRDLFVAKVFNPKTE

|NP_001009304.1-OvUS| 400 KFNRPFLLFVEDEITQTDLFVGQVLNPQVE

|DAA05634.1-CapUS| 400 KFNRPFLLFVEDEMTQTDLFVGQVLNPQVE

|NP_777222.1-BoUS| 430 KVNRPFLLFVEDEKTQRDLFVGKVLNPQVE

|ABG56442.1-BuUS| 404 KVNRPFLLFVEDERTQRDLFVGQVLNPQVE

|XP_855208.1-CanUS| 442 KFNRPFFLFVEDWMTQRAILMGKVFNPIAE

|BK006618-EqUS| 393 KFNRPFLLFVEDELNQRQLFVGQVFNPLQ-

CONSENSUS 491 KfnRPFlLFVEDeitqrdlfvg V NPqve

|AAA31137.1-PoUS-1| 230 FVNEKTIVQVDMMRKTERMIYSRSEELLATMVKIPCKENASIILVLPDTGKFNFALKEMAAKRARLQKTN

|NP_999010.1-PoUS-2| 233 FVNEKTIVQVDMMRKTERMIYSRSEELLATMVKMPCKENASIILVLPDTGKFDFALKEMAAKRARLQKTN

|NP_001009304.1-OvUS| 230 FLNDKTKVQVDMMRKTEQMLYSRSEELFATMVKMPFKGNVSLILMLPDAGHFDNALKKLTAKRAKLQKIS

|DAA05634.1-CapUS| 230 FLNDKTKVQVDMMRKTEQMLYSRSEELFATMVKMPFKGNVSLILMLPDAGHFDNALKKLTAKRAKLQKTS

|NP_777222.1-BoUS| 230 FVNDQTKVQVDMMRKTERMLYSRSEELHATMVKMPCKGNVSLTLMLPDAGQFDTDLKKMTAKRAKLQKIS

|ABG56442.1-BuUS| 230 LLDDKTKVQVDMMRKTERMLYSRSEELHTTMVKMPCKGNVSLILMLPDAGQFDTALKKVTAKRAKLQKIS

|XP_855208.1-CanUS| 281 FVDKHTTVPVDMIWKTGQMIYSRSEELFATMVKIPFVGNMSIVLVLPDVGQPDSAVKEIVVQRATLLQSS

|BK006618-EqUS| 233 FLEDNTSVQVHMMRKTERMIYSRAEHLFATIVKLPYTGNVSLVLVLPDAGRFDFVAKELAARRARPLQSR

CONSENSUS 281 fvndkTkVqVdMmrKTerMiYSRsEeLfaTmVKmPckgNvSliLvLPDaGkfd alK mtakRAklqkts

|AAA31137.1-PoUS-1| 300 DFRLVHLVVPKIKDNLQDRFKHLLP---------KIGINDIFTTKAVTWNTTGTS--TILEAVHHAVIEV

|NP_999010.1-PoUS-2| 303 ELQIGALSCAQDQDHLQDRFKHLLP---------KIGINDIFTTKAVTWNTTRTS--TILEAVHHAVIEV

|NP_001009304.1-OvUS| 300 NFRLVHLTLPKFKITFDINFKHLLPKINLKHLLPKIDPKHTLTTTASSQHVTLKAPLPNLEALHQVEIEL

|DAA05634.1-CapUS| 300 NFRLVHLTLPKFKITFEINFKHLLPKINLKHLLPKIDPKHTLTTTASSQDVTLKAPLPNLEALHQVEIEL

|NP_777222.1-BoUS| 300 DFRLVRLILPKLKISFKINFKHLLP---------KIDPKHILTATAISQAITSKAPLPNLEALHQAEIEL

|ABG56442.1-BuUS| 300 DFRLVRLTLPKLKISFKINFKHLLP---------KIGPKHTLTTTAISQAITLKAPLPNLEALHQAEIEL

|XP_855208.1-CanS 351 NMRWVHIIMPKFKISSKIDLKKILP---------KMGISNVFTTGANFSGITKEDFPTIFEAMHEATMEV

|BK006618-EqUS| 303 DTRLVHLILPKFKISSRIDLNRLLP---------KVGIEDIFSRRANFSGITDETFPTIFEAIHEARLEV

CONSENSUS 351 dfrlvhl lpkfkisfkinfkhlLP Kig khi tttAvsqaiT kapl lEAlHqaeiEv

|AAA31137.1-PoUS-1| 359 KEDGLTKNAAKDKD-FWKVP---------- -----------------------------VDKKEVPVVV

|NP_999010.1-PoUS-2| 362 KEDGLTKNAAKDKD-FWKVP---------- -----------------------------VDKKEVPVVV

|NP_001009304.1-OvUS| 370 SEHALTTDTAIHTDNLLKVP---------- -----------------------------ANTKEVPVVV

|DAA05634.1-CapUS| 370 SEHALTTDTAIHTDNLLKVP---------- -----------------------------VNAKEVPVVV

|NP_777222.1-BoUS| 361 SEHALTVDTAIHTDNLLKVPVKAKEVPAVV KVPMKAKEVPAVVKVPMNTKEVPVVVKVPMNTKEVPVVV

|ABG56442.1-BuUS| 361 NEHALTVDTAIHTDNLLKVP---------- ----------------MNAKEVPVVVKVPTNAKEVPVDV

|XP_855208.1-CanUS| 412 SKEGKMDTSKDMDSRNTIAC---------- -----------------------------HTYTATPLVV

|BK006618-EqUS| 364 NEKG-VIKAAAEDVHAKRAH---------- -----------------------------HAPHADATVV

CONSENSUS 421 sehglt dtaihtdnllkvp vn kevpvvV

|AAA31137.1-PoUS-1| 388 KFDRPFFLFVEDEITRRDLFVAKVFNPKTE

|NP_999010.1-PoUS-2| 391 KFDRPFFLFVEDEITRRDLFVAKVFNPKTE

|NP_001009304.1-OvUS| 400 KFNRPFLLFVEDEITQTDLFVGQVLNPQVE

|DAA05634.1-CapUS| 400 KFNRPFLLFVEDEMTQTDLFVGQVLNPQVE

|NP_777222.1-BoUS| 430 KVNRPFLLFVEDEKTQRDLFVGKVLNPQVE

|ABG56442.1-BuUS| 404 KVNRPFLLFVEDERTQRDLFVGQVLNPQVE

|XP_855208.1-CanUS| 442 KFNRPFFLFVEDWMTQRAILMGKVFNPIAE

|BK006618-EqUS| 393 KFNRPFLLFVEDELNQRQLFVGQVFNPLQ-

CONSENSUS 491 KfnRPFlLFVEDeitqrdlfvg V NPqve

Page 70: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

70

This domain is completely conserved in OvUS, CapUS, PoUS-1 and PoUS-2, is nearly

completely conserved in BuUS (only one amino acid substitution), but is not conserved in

CanUS and EqUS.

Identification of the Putative Hinge Region and P1-P1’ Site of the RCL

ClustalW alignment was used to align all US with inhibitory serpins including human α1-

antitrypsin (Genbank: NP_000286), human and porcine α1-antichymotrypsin (Genbank:

NP_001076 and NP_998952.1), human plasminogen activator inhibitor (Genbank: NP_000593),

bovine α2-plasmin inhibitor (Genbank: NP_777095), human leukocyte elastase inhibitor

(Genbank: P30740), human placental thrombin inhibitor (Genbank: P35237) and human

squamous cell carcinoma antigen 1 (Genbank: NP_536722) (Figure 2-5). Note that while

inhibitory serpins usually have a serine or cysteine at the P1’ site, most of the US have a valine at

the same position. The exception is for EqUS and CanUS which have an isoleucine and arginine

at this position, respectively.

The hinge region of the RCL (underlined in Figure 2-5) is highly conserved among

inhibitory serpins. The consensus sequence of RCL involves an arginine at P17, arginine, lysine

or glutamic acid at P16, glycine at P15, serine or threonine at P14 and for the hinge region an

alanine, glycine or serine at P12-P9 (Irving et al. 2000). For the US, the conserved arginine at

P17 is present in almost all US with the exception of CanUS which has a lysine at this position.

For the P16, US possess an aspartic acid or histidine with the exception of the CanUS (glutamic

acid) and EqUS (lysine). Moreover, the conserved glycine at P15 is present in only PoUS-1,

PoUS-2, EqUS and CanUS. Uterine serpins do not contain threonine or serine at P14 nor

alanine, glycine or serine at positions P12 to P10 (with the exception of the PoUS-1 and -2 which

have an alanine at P10). However, US have a conserved serine or alanine at position P9. Taken

Page 71: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

71

Figure 2-5. Identification of the P1-P1’ site and hinge region (underlined) in uterine serpins.

Represented alignment of a portion of the amino acid sequences of some inhibitory

members of the serpin superfamily and the uterine serpin (blue square). Inferred

amino acid sequences of uterine serpins were aligned with inferred amino acid

sequences of selected inhibitory serpins using the ClustalW software. Shown is a

portion of the polypeptides near the P1-P1’ site. Sites of conservation are shown in

shaded columns using the Boxshade software. Black-shaded columns show

identical amino acids and grey-shaded columns represent similar amino acids.

NP_001076.2| 330 -QLGIEEAFT-SKADLSGITGARNLAVSQVVHKAVLDVFEEGTEASAATAVKITLLSAL-

NP_998952.1| 328 -QLGIEEIFG-DNANLSGITNTKPLKVSQVVHSAVLDVNEEGTEAAAATGIDINVRS---

NP_000286.3| 329 -QLGITKVFS-NGADLSGVTEEAPLKLSKAVHKAVLTIDEKGTEAAGAMFLEAIPMS---

P30740.1| 290 -RLGVQDLFNSSKADLSGMSGARDIFISKIVHKSFVEVNEEGTEAAAATAGIATFCM---

P35237.3| 288 -NLGMTDAFELGKADFSGMS-QTDLSLSKVVHKSFVEVNEEGTEAAAATAAIMMMRC---

NP_536722.1| 316 -DMGITDIFDETRADLTGISPSPNLYLSKIIHKTFVEVDENGTQAAAATGAVVSERS---

NP_001009304.1 333 PKIDPKHTLT-TTASSQHVTLKAPLPNLEALHQVEIELSEHALTTDTAIHTDNLLKVP--

DAA05634.1| 333 PKIDPKHTLT-TTASSQDVTLKAPLPNLEALHQVEIELSEHALTTDTAIHTDNLLKVP--

NP_777222.1| 325 -KIDPKHILT-ATAISQAITSKAPLPNLEALHQAEIELSEHALTVDTAIHTDNLLKVPVK

ABG56442.1| 325 -KIGPKHTLT-TTAISQAITLKAPLPNLEALHQAEIELNEHALTVDTAIHTDNLLKVP--

AAA31137.1| 325 -KIGINDIFT-TKAVTWNTTGTS--TILEAVHHAVIEVKEDGLTKNAAKDKD-FWKVP--

NP_999010.1| 328 -KIGINDIFT-TKAVTWNTTRTS--TILEAVHHAVIEVKEDGLTKNAAKDKD-FWKVP--

XP_855208.1| 376 -KMGISNVFT-TGANFSGITKEDFPTIFEAMHEATMEVSKEGK-MDTSKDMDSRNTIAC-

BK 006618| 328 -KVGIEDIFS-RRANFSGITDETFPTIFEAIHEARLEVNEKG--VIKAAAEDVHAKRAH-

NP_000593.1| 316 --LGMTDMFRQFQADFTSLSDQEPLHVAQALQKVKIEVNESGTVASSSTAVIVSARMAP-

NP_777095.1| 354 -QLGLQELFQ--APDLRGISDER-LVVSSVQHQSALELSEAGVQAAAATSTAMSRMSLS-

NP_001076.2| 387 ----------------------------------------VETRTIVRFNRPFLMIIVPT

NP_998952.1| 383 -----------------------------------------LERIALHFNRPFLFVIISK

NP_000286.3| 384 ------------------------------------------IPPEVKFNKPFVFLMIEQ

P30740.1| 346 ----------------------------------------LMPEENFTADHPFLFFIRHN

P35237.3| 343 ----------------------------------------ARFVPRFCADHPFLFFIQHS

NP_536722.1| 372 ----------------------------------------LRSWVEFNANHPFLFFIRHN

NP_001009304.1 390 -------------------------------------ANTKEVPVVVKFNRPFLLFVEDE

DAA05634.1| 390 -------------------------------------VNAKEVPVVVKFNRPFLLFVEDE

NP_777222.1| 383 AKEVPAVVKVPMKAKEVPAVVKVPMNTKEVPVVVKVPMNTKEVPVVVKVNRPFLLFVEDE

ABG56442.1| 381 ------------------------MNAKEVPVVVKVPTNAKEVPVDVKVNRPFLLFVEDE

AAA31137.1| 378 -------------------------------------VDKKEVPVVVKFDRPFFLFVEDE

NP_999010.1| 381 -------------------------------------VDKKEVPVVVKFDRPFFLFVEDE

XP_855208.1| 432 -------------------------------------HTYTATPLVVKFNRPFFLFVEDW

BK 006618| 383 -------------------------------------HAPHADATVVKFNRPFLLFVEDE

NP_000593.1| 373 --------------------------------------------EEIIMDRPFLFVVRHN

NP_777095.1| 409 ---------------------------------------------SFIVNRPFLFFILED

1P17 15 13 11 9 7 35 1’

NP_001076.2| 330 -QLGIEEAFT-SKADLSGITGARNLAVSQVVHKAVLDVFEEGTEASAATAVKITLLSAL-

NP_998952.1| 328 -QLGIEEIFG-DNANLSGITNTKPLKVSQVVHSAVLDVNEEGTEAAAATGIDINVRS---

NP_000286.3| 329 -QLGITKVFS-NGADLSGVTEEAPLKLSKAVHKAVLTIDEKGTEAAGAMFLEAIPMS---

P30740.1| 290 -RLGVQDLFNSSKADLSGMSGARDIFISKIVHKSFVEVNEEGTEAAAATAGIATFCM---

P35237.3| 288 -NLGMTDAFELGKADFSGMS-QTDLSLSKVVHKSFVEVNEEGTEAAAATAAIMMMRC---

NP_536722.1| 316 -DMGITDIFDETRADLTGISPSPNLYLSKIIHKTFVEVDENGTQAAAATGAVVSERS---

NP_001009304.1 333 PKIDPKHTLT-TTASSQHVTLKAPLPNLEALHQVEIELSEHALTTDTAIHTDNLLKVP--

DAA05634.1| 333 PKIDPKHTLT-TTASSQDVTLKAPLPNLEALHQVEIELSEHALTTDTAIHTDNLLKVP--

NP_777222.1| 325 -KIDPKHILT-ATAISQAITSKAPLPNLEALHQAEIELSEHALTVDTAIHTDNLLKVPVK

ABG56442.1| 325 -KIGPKHTLT-TTAISQAITLKAPLPNLEALHQAEIELNEHALTVDTAIHTDNLLKVP--

AAA31137.1| 325 -KIGINDIFT-TKAVTWNTTGTS--TILEAVHHAVIEVKEDGLTKNAAKDKD-FWKVP--

NP_999010.1| 328 -KIGINDIFT-TKAVTWNTTRTS--TILEAVHHAVIEVKEDGLTKNAAKDKD-FWKVP--

XP_855208.1| 376 -KMGISNVFT-TGANFSGITKEDFPTIFEAMHEATMEVSKEGK-MDTSKDMDSRNTIAC-

BK 006618| 328 -KVGIEDIFS-RRANFSGITDETFPTIFEAIHEARLEVNEKG--VIKAAAEDVHAKRAH-

NP_000593.1| 316 --LGMTDMFRQFQADFTSLSDQEPLHVAQALQKVKIEVNESGTVASSSTAVIVSARMAP-

NP_777095.1| 354 -QLGLQELFQ--APDLRGISDER-LVVSSVQHQSALELSEAGVQAAAATSTAMSRMSLS-

NP_001076.2| 387 ----------------------------------------VETRTIVRFNRPFLMIIVPT

NP_998952.1| 383 -----------------------------------------LERIALHFNRPFLFVIISK

NP_000286.3| 384 ------------------------------------------IPPEVKFNKPFVFLMIEQ

P30740.1| 346 ----------------------------------------LMPEENFTADHPFLFFIRHN

P35237.3| 343 ----------------------------------------ARFVPRFCADHPFLFFIQHS

NP_536722.1| 372 ----------------------------------------LRSWVEFNANHPFLFFIRHN

NP_001009304.1 390 -------------------------------------ANTKEVPVVVKFNRPFLLFVEDE

DAA05634.1| 390 -------------------------------------VNAKEVPVVVKFNRPFLLFVEDE

NP_777222.1| 383 AKEVPAVVKVPMKAKEVPAVVKVPMNTKEVPVVVKVPMNTKEVPVVVKVNRPFLLFVEDE

ABG56442.1| 381 ------------------------MNAKEVPVVVKVPTNAKEVPVDVKVNRPFLLFVEDE

AAA31137.1| 378 -------------------------------------VDKKEVPVVVKFDRPFFLFVEDE

NP_999010.1| 381 -------------------------------------VDKKEVPVVVKFDRPFFLFVEDE

XP_855208.1| 432 -------------------------------------HTYTATPLVVKFNRPFFLFVEDW

BK 006618| 383 -------------------------------------HAPHADATVVKFNRPFLLFVEDE

NP_000593.1| 373 --------------------------------------------EEIIMDRPFLFVVRHN

NP_777095.1| 409 ---------------------------------------------SFIVNRPFLFFILED

1P17 15 13 11 9 7 35 1’

Page 72: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

72

together, results indicate that the RCL and the hinge region required for inhibitory activity has

not been conserved in uterine serpins.

Phylogenetic Analysis of Uterine Serpins

The phylogenetic tree constructed using the NJ method is presented in Figure 2-6. The

sheep and goat proteins as wells as the water buffalo and cow proteins were clustered together,

and the horse and dog were clustered together in a sister group and the pig proteins were closer

to the horse and dog than to the ruminants.

Positive Selection of the Uterine Serpin Gene

Aligned sequences of uterine serpins were tested for evidence of positive selection using

different models of variable dN/dS ratios among sites (Table 2-3). The LRT for positive

selection was significant when comparing M2 and M8 against their null model (Table 2-4). The

Bayes empirical Bayes method used in PAML to calculate the posterior probabilities at each

codon site (Yang et al. 2005) indicate A121, G343 and K380 as positive selected sites for M2

and I35, A121, G343, K365, V378, F373 and K380 as positive selected sites for M8. These

results partially agreed with those obtained with the Selecton program where six amino acid

residues (Figure 2-7) were identified as sites with potential positive selection (I35, A121, D156,

G343, K365 and K380) and thirty nine amino acid residues were likely to be under purifying

selection (M1, H3, M6, S22, I23, V38, A58, F62, E68, K72, I75, F76, A86, D167, I178, V182,

T195, T201, K226, E227, F229, T235, V237, V239, M241, K244, M248, S251, E254, N268,

D277, K324, E349, E357, K388, F393, E398, V411 and N413).

To identify whether codons undergoing positive selection were associated with regions

critical for antiproteinase activity, the amino acid sequence of OvUS was aligned to human α1 -

antitrypsin to identify helices and β-sheets of the protein (Figure 2-8). A total of six amino acid

residues were identified as being subject to positive selection by at least two of the three

Page 73: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

73

Figure 2-6. Phylogenetic tree of the uterine serpin proteins with the ovine uterine serpin

(OvUS) as out-group. The Jones-Taylor-Thornton matrix was used for distance

calculation with gamma corrected distances and tree was inferred by the Neighbor-

joining method. The tree reliability was tested with 1000 bootstrap replicates (not

shown).

Page 74: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

74

Table 2-3. Ratios (ω) of nonsynonymus (dN) to synonymous (dS) mutations, parameter

estimates and maximum log likelihood of models for positive selection within the

protein coding sequence of uterine serpins.

a The numbers of free parameters are represented by (p) assigned to each class of ω

Model dN/dS Parameter estimate(s) a L

M0 (one ratio) 0.8815 ω = 0.8815 -6270.25

M1 (Nearly Neutral) 0.7506 p0= 0.29151, p1= 0.70849, ω0= 0.14433, ω1= 1 -6233.37

M2 (Positive selection) 1.0717 p0= 0.25346, p1= 0.61790, p2= 0.12864, ω0= 0.16249, ω1= 1, ω2= 3.20732 -6219.15

M7 (Beta) 0.7799 p= 0.02945 q= 0.00709 -6235.54

M8 (Beta & ω) 1.0860 p= 0.49271, q= 0.19375, p0= 0.83214, p1= 0.16786, ωs= 2.91144 -6219.83

Page 75: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

75

Table 2-4. Test of significance for models for positive selection within the protein coding

sequence of uterine serpins.a

Models 2L 2 value df P values

Positive selected sites

(BEB)b

M1 versus M2 2(-6219.15+6233.37) 28.44 2 <0.001 A121**, G343*, K380*

M7 versus M8 2(-6219.83+6235.54) 31.42 2 <0.001 I35*, A121**, G343*, K365*

V378*, F373*, K380**

a The likelihood ratio test statistics were calculated as two times the difference in the log

likelihood between models (2L). Significance was tested by using the 2 test with degrees of

freedom calculated from the parameter estimates.

b

Sites of positive selection were estimated by calculating the posterior probability that each

codon was from the site class of positive selection under models M2 and M8 by the Bayes

empirical Bayes method (* P >0.95; ** P >0.99).

Page 76: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

76

Figure 2-7. Selecton output generated for the uterine serpin group of proteins. Shown is the

sequence for PoUS-1. Color intensity indicates likelihood of purifying (dark purple)

and positive selection (dark yellow) at each amino acid residues. Six amino acid

residues were estimated to undergo positive selection (I35, A121, D156, G343,

K365 and K380) while thirty nine were identified to be under purifying selection.

Amino acid residues identified as under positive selection by the Bayes empirical

Bayes method for models M2 and M8 of the PAML program are shown by an

asterisk (*) and diamond (), respectively. The putative hinge region of the reactive

center loop is underlined and the putative scissile bond at P1-P1’ site is indicated by

an arrow.

*

*

*

P1-P1’

*

*

*

P1-P1’

Page 77: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

77

Figure 2-8. Amino acid sequence alignment of ovine uterine serpin (NP_001009304.1) and

human α1-antitrypsin (NP_000286.3) using the ClustalW algorithm. Helices are

shown as green cylinders and β-pleated sheets are shown as yellow arrows. The

location and designation of these regions are based on Silverman et al. (2004).

Amino acid residues identified as being subject to positive selection by at least two

of three statistical methods are indicated by arrowheads.

s1C s4B s5B

s5A

HA HBs6B

s3A s4C s3C

HG HHs1C s2B s3B

HC HD s2A

HE HFs1A

s6As2C HI

s1C s4B s5Bs1C s4B s5B

s5A

HA HBs6B

s3A s4C s3C

HG HHs1C s2B s3B

HC HD s2A

HE HFs1A

s6As2C HI

s5As5A

HA HBs6BHA HBs6B

s3A s4C s3Cs3A s4C s3C

HG HHs1C s2B s3B HG HHs1C s2B s3B

HC HD s2AHC HD s2A

HE HFs1AHE HFs1A

s6As2C HIs6As2C HI

Page 78: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

78

statistical methods. Of the six positive selected sites, two are in colied regions, one is in the

putative Helix D, one is in the putative Helix E, one is in the reactive center loop and one is the

5’ site relative to the putative scissile bond (Figure 2-8).

Discussion

Serpin genes are of ancient origin and are found in bacteria, fungus, nematodes, archaea

and virus (Irving et al. 2000, 2002b, Steenbakkers et al. 2008). The fact that uterine serpins

(SERPINA14) are apparently limited to species in the Laurasiatheria superorder means that they

either arose recently, after Laurasiatheria diverged from Euarchontoglires (~87 Mya) (Murphy et

al. 2007), or they have been retained during evolution in this superorder only. Their uterine

expression and the loss of amino acid motifs required for proteinase inhibitory activity in the P1-

P1’ site and hinge region indicates that the genes have been selected for a new function required

for maintenance of pregnancy. Based on experiments with sheep and pigs, uterine serpins may

function to inhibit cell proliferation (Padua & Hansen 2008) or interact with other uterine

proteins (Baumbach et al. 1986, Hansen & Newton 1988, McFarlane et al. 1999). The uterine

serpins are thus an example of a new gene arising from gene duplication and selection for

sequence divergence (Louis 2007) that plays a particular role in pregnancy.

While it was not possible to examine complete genomic sequences in each order of

mammals, the available evidence points strongly to restriction of the uterine serpin to

Laurasiatheria only. The distribution of known uterine serpins as well as orders where blast

search of complete genomic sequences fail to identify a uterine serpin is shown in Figure 2-9.

All the identified uterine serpins are in Ruminantia, Suidae, Perissodactyla or Carnivora orders

of Laurasiatheria. No gene that significantly matched OvUS was found in within the orders

Page 79: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

79

Figure 2-9. Phylogenetic tree of placentation in mammals (adapted from Vogel 2005) to

illustrate the existence of uterine serpin genes relative to type of placentation.

Shown are the 4 major superorders of eutherian mammals (Laurasiatheria,

Euarchontoglires, Xenarthra and Afrotheria) with the order Marsupiala as the out-

group of the tree. Blue branches represent orders with epitheliochorial placenta and

orange branches represent orders with either endotheliochorial or hemochorial type

of placentation. The yellow branches represent unresolved situations in the

phylogeny. The symbol represents those orders within the Laurasiatheria

superorder where the uterine serpin gene has been identified. The symbol

represents those orders where the uterine serpin gene was not identified after blast

search of complete genomic sequences.

Proboscidea

Sirenia

Xenarthra

Tenrecomorpha

Marsupialia

Laurasiatheria

Afrotheria

Xenarthra

Euarchontoglires

Cetacea

Hippopotamidae

Carnivora

Suidae

Pholidota

Perissodactyla

Ruminantia

Talpa

Rodentia

Lemuriformes

Scandentia

Tarsiphormes

Scalopus

Anthropoidea

Lagomorpha

Chiroptera

Erinaceomorpha

Proboscidea

Sirenia

Xenarthra

Tenrecomorpha

Marsupialia

Laurasiatheria

Afrotheria

Xenarthra

Euarchontoglires

Cetacea

Hippopotamidae

Carnivora

Suidae

Pholidota

Perissodactyla

Ruminantia

Talpa

Rodentia

Lemuriformes

Scandentia

Tarsiphormes

Scalopus

Anthropoidea

Lagomorpha

Chiroptera

Erinaceomorpha

Page 80: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

80

Anthropoidea and Rodentia of the superorder Euarchontoglires, Marsupialia, or in non-

mammalian species examined.

Given the fact that all uterine serpins identified before this study were species exhibiting

epitheliochorial placentation (Figure 2-9), it was hypothesized that uterine serpin genes serve

sole role uniquely required for species with this type of placentation. While this hypothesis may

be correct it is not true that the gene is only present in species with epitheliochorial placentation.

Of the two new uterine serpins identified in the present study, one is for species with

epitheliochorial placentation (horse), but one is for species with endotheliochorial placentation

(dog). Phylogenetic analysis showed that the genes in the dog and horse are closely related to

each other. In fact, CanUS and EqUS are closer to each other than to PoUS-1 and PoUS-2. For

example, the KEVPVVVK in other uterine serpins including pig is lost in both horse and dog. In

some mammalian phylogenies, the horse is more closely related to the pig than carnivores

(Vogel 2005) whereas in others, the horse is closer to carnivores than to pigs (Nishihara et al.

2006) or the horse is equidistant from carnivores and pigs (Murphy et al. 2007).

Even though the dog has a uterine serpin gene, there was no evidence for functional uterine

serpin gene in the closely-related cat. Results must be tentative because a complete genomic

sequence of cat is not yet available. However, the fact that nucleotide sequences identified in cat

as being similar to uterine serpins contained stop codons suggests that a functional uterine serpin

gene has been lost in this species . Additionally, the dog uterine serpin has undergone significant

change from other serpins in that it has acquired two putative start codons upstream from the

start codons in other species. If in fact this start codon is correct, the CanUS gene encodes for an

intracellular protein without a signal peptide while all other species with uterine serpin encode

for secretory proteins (Moffatt et al. 1987, Leslie et al. 1990, Malathy et al. 1990, Tekin et al.

Page 81: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

81

2005a). The newly discovered EqUS was also shown to be secreted into the uterine lumen as

revealed by western blotting of uterine flushings.

Taken together, it is possible that the uterine serpin gene in species with epitheliochorial

placentation has been required as a secretory protein while the gene in species with

endotheliochorial placentation has either being changed to become an intracellular protein (dog)

or a pseudogene (cat). If this idea is correct, uterine serpins play some important role in the

uterine lumen of species with epitheliochorial placentation that is not required for species with

either endotheliochorial placentae. This hypothesis is also supported by the absence of a

homologous uterine serpin gene in human and rodents, species with hemochorial placentation.

The function of uterine serpins is probably not to inhibit serine or cysteine proteinases.

Ovine and BoUS exhibit no inhibitory activity towards a range of proteinases (Ing & Roberts

1989, Liu & Hansen 1995, Mathialagan & Hansen 1996, Peltier et al. 2000a). In addition, there

is evidence, as shown here, that the P1-P1’ site and hinge region of the uterine serpins differ

from inhibitory serpins. The RCL contains a complementary sequence to the active site of the

target proteinase and the hinge region of RCL is highly conserved among inhibitory serpins

(Irving et al. 2000). In uterine serpins, however, the amino acids of the consensus sequence is

absent at P15 (except for PoUS-1, PoUS-2, EqUS and CanUS), P14 and P12 to P10 (except for

PoUS-1 and -2 which have an alanine at P10). Furthermore, an amino acid in the RCL and at the

5’ position were identified as positions subject to positive selection and the P1 site is not

conserved with inhibitory serpins. Among the serpin superfamily, the RCL is important for

selectivity of the serpin for the target proteinase and has undergone an accelerated rate of

evolution (Brown 1987, Hill & Hastie 1987, Goodwin et al. 1996, Zang & Maizels 2001,

Barbour et al. 2002).

Page 82: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

82

The identification of the P1-P1’ site in this study is different by two amino acids (K-V

instead of P-V) that was identified in the original paper by Ing & Robets (1989) and used as the

basis for assigning its location in other papers (Mathialagan & Hansen 1996, Peltier et al. 2000c,

Tekin et al. 2005a). The accessibility to more complete sequences from inhibitory serpins whose

P1-P1’ sites have already been determined and also more uterine serpin sequences in addition to

improvement in alignment programs results in a more reliable estimate of the P1-P1’site for the

uterine serpins.

Unique functions of uterine serpins in species with epitheliochorial placentae must be

linked to the unique characteristics of placentation in this species. Unlike other type of placentae

in eutherian mammals, where maternal blood either spills directly onto trophoblast (hemochorial

placenta) or where trophoblast invasion and erosion of endometrial tissue leaves the trophoblast

in contact with maternal endothelium, the epitheliochorial placenta is characterized by limited

invasiveness of trophoblast and an intact endometrial epithelium that sometimes forms a

synctium with trophoblast cells. The efficiency of placental transport is improved in species

with epitheliochorial placentae because of countercurrent exchange of nutrients and gases

between fetal and maternal blood vessels (Leiser et al. 1997). It has also been speculated that the

presence of an intact maternal endometrial epithelium may reduce the recognition of trophoblast

antigens by maternal immune system (Moffet & Loke 2006) although available evidence

indicates that the maternal immune response to the trophoblast is similar in species with

epitheliochorial placentation to that of other species (Oliveira & Hansen 2008).

Ovine uterine serpin has been shown to inhibit proliferation of lymphocytes and a variety

of cancer cells (Padua & Hansen 2008) and it is possible that one or more uterine serpins

function to inhibit cell proliferation during pregnancy. Inhibition of lymphocyte proliferation

Page 83: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

83

has been interpreted as signifying role for uterine serpins in prevention of rejection of conceptus

tissue by the maternal immune system (Hansen 1998). In addition, the limited invasiveness of

trophoblast tissue in species with epitheliochorial placentation could be achieved, at least in part,

by inhibition of trophoblast proliferation. Through inhibition of the cell cycle (Padua & Hansen

2008), it may be also that uterine serpins participate in the process of trophoblast binucleate cell

formation, which occurs in sheep, goat, cattle and horse (Hoffman & Wooding 1993).

Another characteristic of uterine serpins is the propensity to bind other proteins. Ovine

uterine serpin has been found to bind activin (McFarlane et al. 1999), IgA and IgM (Hansen &

Newton 1988), and the so-called pregnancy associated glycoproteins (Mathialagan & Hansen

1996) that are inactive members of the aspartic proteinase family (Xie et al. 1991, Green et al.

1998). The pig uterine serpins bind to an iron-binding protein secreted by the uterus called

uteroferrin and stabilize the iron bound to uteroferrin (Baumbach et al. 1989). In some cases,

this binding can be ascribed to the highly basic isoelectric point for uterine serpins. Binding to

IgA and pepsin can be reduced by high salt concentrations (Hansen & Newton 1988, Peltier et al.

2000a). However, a physiological role for protein binding remains a possibility. Binding to

uteroferrin may enhance iron transport to the fetus since this protein delivers its iron to fetal

fluids (Buhi et al. 1982). The function of pregnancy associated glycoproteins is not known but

the conservation of the KEVPVVVK motif located near the P1-P1’ site described as pepstatin-

like domain (Mathialagan & Hansen 1996) may mean that binding to pregnancy associated

glycoproteins is an important function.

Progesterone is the hormone that induces the synthesis and secretion of uterine serpins into

the uterine lumen of sheep, pig, cow and goat (Ing et al. 1989, Leslie et al. 1990, Malathy et al.

1990, Tekin et al. 2005a). It is likely that the equine protein is also stimulated by progesterone.

Page 84: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

84

In addition, in the cow (Khatib et al. 2007) as well as in dog, the uterine serpin gene is expressed

in the ovary. The presence of uterine serpin in other reproductive tissues distinct to the uterus

raises the possibility that uterine serpins may have different functions according to the cell type

and stage of differentiation. It is possible that uterine serpin functions in the ovary to regulate

follicular growth by binding to activin. Activin improves follicular growth and granulosa cell

proliferation (Knight & Glister 2006). OvUS binds to this member of the TGF-β family

(McFarlane et al. 1999).

In summary, the uterine serpin gene is present only in a restricted group of species within

the Laurasiatheria superorder of eutherian mammals and likely evolved under positive selection

which suggests diversifying functionality of these proteins from the proteinase inhibitory activity

of most members of the serpin superfamily. This evidence also suggests the uterine serpins gene

is an example of gene duplication followed by selection. The finding that the uterine serpin has

been retained as a secretory protein in species with epitheliochorial placentation within the

Laurasiatheria superorder also suggests that the protein has an important role during pregnancy

in these species while in species with endotheliochorial placenta the gene has undergone changes

suggesting that it is playing either a new role in those species (dog) or no required (cat) during

pregnancy.

Page 85: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

85

CHAPTER 3

COMPARISON OF THE NATIVE AND RECOMBINANT FORMS OF OVINE UTERINE

SERPIN FOR INHIBITION OF CELL PROLIFERATION

Introduction

The uterine serpins (US) are a group of glycosylated proteins secreted into the uterus of the

sheep, cow, goat and pig during mid and late pregnancy (Moffatt et al. 1987, Leslie et al. 1990,

Baumbach et al. 1986, Tekin et al. 2005a). These proteins are members of the superfamily of

serine proteinase inhibitors (serpins) (Ing & Robets 1989, Mathialagan & Hansen 1996). Most

of the members of the serpin superfamily are inhibitors of serine or cysteine proteinases, which

are inactivated by an irreversible suicide substrate-like mechanism (Silverman et al. 2001).

However, there are some members of this superfamily that have roles distinct from the inhibition

of proteinases. Some examples of non-inhibitory serpins include the hormone transport proteins

corticosteroid and thyroxine binding globulin (Pemberton et al. 1988), the chaperone heat shock

protein 47 (Nagata 1998) and angiotensinogen which is involved in the regulation of blood

pressure (Morgan et al. 1996).

Another serpin that seems to have a divergent function is ovine uterine serpin (OvUS).

This US has been linked to the protection of the allogeneic conceptus through inhibition of

immune cell proliferation during pregnancy (Hansen 1998). Several experiments have

demonstrated that OvUS inhibited lymphocyte proliferation induced by mitogens such as

concanavalin A (Con A), the antigen Candida albicans, phytohemagglutinin and in the mixed

lymphocyte reactions (Segerson et al. 1984, Hansen et al. 1987b, Stephenson et al. 1989b,

Skopets & Hansen 1993, Skopets et al. 1995). Ovine US also inhibits natural killer (NK) cell

activity (Liu & Hansen 1993). Moreover, OvUS inhibited the lytic activity of NK-like cells in

sheep peripheral blood lymphocytes and endometrial epithelium against D17 cells infected with

bovine herpes virus-1 (Tekin & Hansen 2002).

Page 86: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

86

However, OvUS did not cause any inhibitory activity against lymphocytes activated by the

T and B cell mitogen PWM (Skopets & Hansen 1993). Ovine US had no effect on the reduction

of skin-fold thickness caused by Mycobacterium tuberculosis in sheep (Skopets et al. 1995) and

failed to inhibit expression of CD25 on γδ-T + cells induced by Con A (Peltier et al. 2000b),

suggesting the apparent selectivity of the protein to inhibit some immune cell types.

Little is known about the antiproliferative actions of OvUS in other non-immune cell types.

The objectives pursued on this study are to test whether OvUS inhibited proliferation of non-

immune cells, specifically tumor cells, and to determine whether OvUS inhibits cell proliferation

by apoptosis. Finally, the antiproliferative potency of a recombinant form of OvUS (rOvUS)

was compared with the native form of the protein.

Materials and Methods

Materials

The Eagle’s Minimum Essential Medium (MEM), Dulbecco’s Modified Eagle Medium

Nutrient Mixture F-12 Ham (DMEM-F12), Dulbecco’s Modified Eagle’s Medium (DMEM),

Dulbecco’s phosphate buffered saline (DPBS) and penicillin–streptomycin were purchased from

Sigma-Aldrich (St Louis, MO). The fetal bovine serum was from Intergen (Purchase, NY) and

the heat-inactivated horse serum was from Hyclone (Logan, UT). The D-17 (canine primary

osteogenic sarcoma), PC-3 (human prostate cancer) and P388D1 (mouse lymphoma) cell lines

were purchased from ATCC (Rockville, MD). The centricon ultrafiltration devices were from

Amicon (Beverly, MA) or Millipore (Bedford, MA). Carboxymethyl Sepharose, Sephacryl S-

200 and His-Trap columns were obtained from Amersham Biosciences (Piscataway, NJ) and

[3H]thymidine (6.7 Ci/mmol) was from ICN (Irvine, CA). The RQ1 RNase-free DNase was

from Promega (Madison, WI), in situ cell death detection kit [terminal deoxynucleotidyl

transferasemediated dUTP nick end labeling (TUNEL)] was from Roche (Indianapolis, IN). The

Page 87: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

87

ProLong Antifade kit was purchased from Molecular Probes (Eugene, OR), RNase A was from

Qiagen (Valencia, CA) and the Precast Tris-HCl gradient gels® were obtained from Bio-Rad

(Richmond, CA). Other reagents were from either Sigma-Aldrich or Fisher.

Collection of Uterine Fluid and Purification of Native OvUS

Ewes of Rambouillet genotype (n=4) were made unilaterally pregnant as described

elsewhere (Bazer et al. 1979) by ligating one uterine horn. Ewes were slaughtered at day 140 of

pregnancy by captive bolt stunning and exsanguination. Crude uterine fluid was collected from

the ligated uterine horn after slaughter by aspiration. Uterine fluid was clarified by

centrifugation at 3600 x g and stored at –20oC until purification of native ovine uterine serpin

(nOvUS) by a combination of cation-exchange chromatography with carboxymethyl Sepharose

and gel filtration chromatography with Sephacryl S-200 as previously described (Liu & Hansen

1993). The purity of the protein was assessed by sodium dodecyl sulfate, polyacrylamide gel

electrophoresis (SDS-PAGE) using 4-15% polyacrylamide Precast Tris-HCl gradient gels and

staining by Coomassie Blue G-250. After purification, nOvUS was buffer-exchanged with

DPBS and concentrated using Centricon PL-20 ultrafiltration devices. Protein concentration was

determined using the Bradford assay with bovine serum albumin as standard (Bradford 1976).

[3H]thymidine Incorporation by D17 and PC-3 Cells

The D17 cells were cultured continuously in complete medium [MEM supplemented with

10% (v/v) heat-inactivated fetal bovine serum, 200 U/ml penicillin and 2 mg/ml streptomycin].

The PC-3 cells were grown in DMEM-F12 medium supplemented with 10% (v/v) heat-

inactivated fetal bovine serum, 200 U/ml penicillin and 2 mg/ml streptomycin. Both cell lines

were cultured continuously at 37oC in a humidified 5% (v/v) CO2. At confluence, cells were

trypsinized, centrifuged for 5 min at 110 x g and resuspended in fresh complete medium. Cell

Page 88: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

88

viability was assessed by trypan blue exclusion and cell concentration adjusted to 1 x 105

cells/ml.

For the [3H]thymidine incorporation assay, aliquots of 100 μl containing 1 x 10

4 cells were

cultured in 96-well plates at 37oC and 5% (v/v) CO2 with all treatments in triplicate. After 24 h

in culture, treatments consisting of 1 mg/ml of nOvUS or ovallbumin (OVA, as negative control)

were added. For control wells, an equivalent volume of DPBS was added instead of proteins.

The final volume was adjusted to 200 μl with complete culture medium. After 48 h of culture,

0.1 μCi of [3H]thymidine in 10 μl of complete culture medium were added and cells were

harvested onto glass-fiber filters using a cell harvester device at 24 h after thymidine addition.

Filters were counted for radioactivity using scintillation spectrometry.

Induction of Apoptosis in D17 and PC-3 Cells

To determine apoptosis, 1 x 104 cells in 100 μl were cultured with vehicle, 1 mg/ml nOvUS

or OVA. For the vehicle treatment, an equivalent volume of DPBS was added instead of OvUS

or OVA. The final volume in all wells was adjusted with culture medium to 200 μl. Cells were

cultured at 37oC and 5% (v/v) CO2 for 24 h and then harvested for the detection of apoptotic

cells by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL). All

treatments were performed in triplicate and the experiment was performed on three (D17) or one

(PC-3) separate occasions.

Cells were processed for TUNEL staining as follows. Cells were washed by centrifugation

with 0.1 M sodium phosphate, pH 7.4 with 0.9% (w/v) NaCl (PBS) and containing 1 mg/ml

polyvinyl pyrrolidone (PBS/PVP), and then resuspended in 200 μl 4% (w/v) paraformaldehyde

in 0.2 M sodium phosphate, pH 7.4 with 0.9% (w/v) NaCl for 1 h at room temperature. Cells

were washed again with PBS/PVP, resuspended in 200 μl PBS/PVP, and 100 μl cell suspension

was transferred to a poly-l-lysine coated slide and allowed to dry for at least 24 h at room

Page 89: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

89

temperature. For TUNEL staining, slides were washed twice in PBS/PVP (2 min each) and then

incubated with permeabilization solution [0.1% (v/v) Triton X-100 containing 0.1 % (w/v)

sodium citrate in PBS] for 1 h at room temperature. Positive controls were incubated with RQ1

RNase-free DNase (50 U/ml) at 37oC for 1 h. Slides were washed in PBS/PVP and then

incubated with 50 μl TUNEL reaction mixture (containing fluorescein isothiocyanate-conjugated

dUTP and the enzyme terminal deoxynucleotidyl transferase as prepared by the manufacturer)

for 1 h at 37oC in the dark. Negative controls were incubated in the absence of the enzyme.

After washing, slides were incubated with RNase A (50 μg/ml) for 1 h at room temperature and

then with 12.5 μg/ml propidium iodide for 30 min at room temperature. Cells were washed 4

times in PBS/PVP to remove excess propidium iodide and coverslips mounted with mounting

medium containing Prolong Antifade®

as recommended by the manufacturer. Slides were

observed using a Zeiss Axioplan 2 fluorescence microscope with dual filter (Carl Zeiss, Inc.,

Göttingen, Germany). Images were acquired using AxioVision software and a high-resolution

black and white AxioCam MRm digital camera (Carl Zeiss, Inc., Thorwood, NY). Percent

apoptotic cells were determined by counting the total number of TUNEL-labeled nuclei in at 10

different sites on the slide.

Purification of His-Tagged rOvUS from Conditioned Medium

The human embryonic kidney (HEK)-293F (Gibco-Invitrogen, Carlsbad, CA) cells

transfected with a plasmid construct containing the gene for OvUS (Tekin et al. 2006) was

cultured continuously in selective medium [FreeStyleTM

293 expression medium containing 700

μg/ml of Geneticin] at 37oC in a humidified 8% (v/v) CO2 incubator according to the

manufacturer’s recommendations. Conditioned medium from 293F-rOvUS cells was clarified

by centrifugation at 4000 x g for 15 min and the supernatant was removed and concentrated

using Centricon PL-80 or 20 concentration devices (molecular weight exclusion limit = 30000).

Page 90: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

90

Purification of the His-tagged rOvUS was achieved using HisTrap columns. The binding buffer

was 20 mM sodium phosphate buffer, pH 8.0, 15 mM imidazole, 0.3 M NaCl, and the elution

buffer was 20 mM sodium phosphate buffer, pH 8.0, 0.3 M NaCl and with 300 or 500 mM

imidazole. The concentrated medium was diluted with at least an equal volume of binding buffer

and loaded onto the His Trap columns. The protein recovered after elution was buffer-

exchanged with DPBS and concentrated using Centricon PL-20 filters. Purity of rOvUs was

assessed by SDS-PAGE using 4-15% polyacrylamide Precast Tris-HCl gradient gels and

concentration was determined using the Bradford assay (Bradford 1976).

Expression and Purification of β-Galactosidase

The pcDNA3.1/V5-His-TOPO/lacZ®

plasmid containing the gene for β-galactosidase

(Gibco-Invitrogen, Grand Island, NY) was used to express a His-tagged β-galactosidase as a

control protein. FreestyleTM

293-F cells were transfected as described somewhere else (Tekin et

al. 2006). Transfected cells were then selected as described for the rOvUS. After harvesting, the

cell pellet was collected by centrifugation at 100 x g for 10 min at room temperature. The pellet

was resuspended in 20 mM sodium phosphate, pH 8.0 containing 0.3 M NaCl and 35 mM

imidazole and a proteinase inhibitor cocktail diluted as per manufacturer’s recommendations

(Sigma-Aldrich). Cells were lysed by two freeze-thaw cycles. The supernatant was collected by

centrifugation and the recombinant His-tagged protein purified as previously described for

rOvUS.

Proliferation of P388D1 and PC-3 Cells

The P388D1 cells were cultured continuously in DMEM supplemented with 10% (v/v)

heat-inactivated horse serum, 200 U/ml penicillin and 200 μg/mL streptomycin. The PC-3 cells

Page 91: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

91

were grown in DMEM-F12 medium (Gibco-Invitrogen) supplemented with 10% (v/v) heat-

inactivated fetal bovine serum, 200 U/ml penicillin and 200 μg/ml streptomycin.

The proliferation assay was performed in 96-well culture plates at 37oC and 5% (v/v) CO2

with all treatments in triplicate. For P388D1 cells, 2 x 103 cells in 100 μl of culture medium

were cultured for 6 h before the addition of various concentrations (0, 125, 250, 500 and 1000

μg/ml) of nOvUS, rOvUS and OVA (negative control). For PC-3 cells, 1 x 104

cells in 100 μl

were cultured for 24 h before addition of 0, 62.5, 125, 250, 500 and 1000 μg/ml of nOvUS,

rOvUS or OVA. For control wells, an equivalent volume of DPBS was added instead of

proteins. The final volume was adjusted to 200 μl with culture medium. After 24 h of culture,

0.1 μCi of [3H]thymidine in 10 μl culture medium were added and cells were harvested onto

glass-fiber filters using a cell harvester device at 24 h after thymidine addition. Filters were

counted for radioactivity using scintillation spectrometry. Experiments were performed on 3 and

5 separate occasions for P388D1 and PC-3 cells respectively. Another experiment with PC-3

cells was performed as described above except with recombinant β-galatcosidase at

concentrations of 50, 100 and 200 μg/ml in nine replicates in one assay.

Statistical Analysis

Data on [3H]thymidine incorporation were analyzed by least squares analysis of variance

using the General Linear Models Procedure of SAS (SAS for Windows, Release 8.02; SAS Inst.,

Inc., Cary, NC). Replicate was considered as a random effect and other main effects were

considered fixed. Error terms were determined based on calculation of expected mean squares.

In some analyses, the pdiff mean separation test of SAS was performed to determine treatments

that differed from untreated cells.

Page 92: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

92

Results

Inhibition of Proliferation and Induction of Apoptosis in D17 and PC-3 Cells

Ovine uterine serpin inhibited proliferation (P<0.05) of both D17 and PC-3 cells (Figure 3-

1A). The proportion of cells that were TUNEL-positive was very low (1.1% for control D17

cells and 0.5% for control PC-3 cells) and was slightly increased by OvUS (2.2% for D17 cells

treated with OvUS and 3.3% for PC-3 cells treated with OvUS; Figure 3-1B). Nonetheless, the

number of apoptotic cells was very low for all treatments (see Figures 3-1C for examples of

TUNEL staining).

Antiproliferative Actions on P388D1 and PC-3 Cell Lines: Comparison of the Native and

Recombinant Forms of OvUS

Both nOvUS and rOvUS inhibited proliferation of P388D1 cells as measured by

incorporation of [3H]thymidine into DNA (Fig. 3-2). All concentrations of rOvUS tested were

inhibitory, with the lowest concentration being 125 μg/ml. For nOvUS, in contrast, there was

inhibition only at 500 μg/ml (P<0.10) and 1000 μg/ml (P<0.001) and the magnitude of the

reduction in proliferation at any concentration was greater for rOvUS than for OvUS. The

control protein, ovalbumin, did not inhibit proliferation.

Similar results were obtained for PC-3 cells (Figure 3-2) except that, in this case, the only

significant inhibition was for rOvUS. All concentrations tested, including a concentration as low

as 62.5 μg/ml, reduced [3H]thymidine incorporation (P<0.001). While not significant, the

highest concentration of nOvUS (1000 μg/ml) tended to reduce [3H]thymidine incorporation. In

another experiment, there was no inhibitory effect of recombinant β-galactosidase on

proliferation of PC-3 cells. Incorporation of [3H]thymidine in cells treated with 50, 100 and 200

μg/ml β-galactosidase was 114, 110, and 112% of values for cells cultured without β-

galactosidase.

Page 93: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

93

Figure 3-1. Effect of OvUS on induction of apoptosis in D17 and PC-3 cells as determined by

TUNEL labeling. Data in panel A illustrate that addition of 1 mg/ml OvUS reduced

incorporation of [3H]thymidine into D17 cells and PC-3 cells. Results for D17 cells

are least square means + SEM and are based on three separate assays and with two

separate batches of OvUS. Results for PC-3 cells are least square means + SEM

and are based on one assay with two separate batches of OvUS. Asterisks represent

means that differ from the control value at P<0.05. Data in panel B represent the

percent of cells that were TUNEL-positive after 24 h culture. Data are from a

representative assay. Panel C shows representative patterns of TUNEL labeling for

D17 cells (top row) and PC-3 cells (bottom row). Note that yellow cells are

considered apoptotic.

Page 94: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

94

Figure 3-2. Inhibition of [3H]thymidine incorporation of P388D1 cells and PC-3 cells by native

(n) and recombinant (r) OvUS. Cells were cultured with various concentrations of

nOvUS (open circles), rOvUS (filled circles and solid line) or ovalbumin (OVA)

(filled circles and dashed line). Data represents least square means ± SEM. Means

that differ from untreated cells are indicated by symbols (†P<0.06; ***P<0.001).

0 200 400 600 800 1000

0

2000

4000

6000

8000

10000

12000 PC-3

[3H

]th

ym

idin

e i

nco

rpo

rati

on

(d

pm

)

Col 1 vs Col 2

Col 1 vs Col 4

Col 1 vs Col 6

0

5000

10000

15000

20000

25000

30000

35000

P388D1

Protein concentration (μg/ml)

****** ***

***

******

***

******

***

Page 95: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

95

Discussion

Results from the present study showed that the antiproliferative effect of OvUS is not

restricted to immune cells, specifically the mitogen-stimulated lymphocytes. Ovine US can

inhibit the proliferation of three different tumor cell lines (D17, PC-3 and P388D1). It is

important to mention that there are differences between the signaling pathways of these tumor

cells and the mitogen-activated lymphocytes. Usually, tumor cells present mutations on genes

that are related to cell growth control. In contrast, lymphocytes are in a resting state and once

activated by mitogens, the expression and production the IL-2 receptor is crucial for proliferation

and activation of these immune cells (Ellery & Nicholls 2002). Thus, it is possible that the

signaling pathways affected by OvUS to inhibit cell proliferation of tumor cells and lymphocytes

are different. It was previously shown that OvUS blocked IL-2 induced proliferation and

reduced expression of CD25 (IL-2Rα chain), but it did not affect the steady state amounts of IL-2

mRNA caused by the mitogen Con A (Peltier et al. 2000c).

There are few serpins that inhibit cell proliferation. The mammary serine proteinase

inhibitor (maspin) is one example where the function of this serpin has been related to the

suppression of tumor growth by the induction of apoptosis of cancer cells and the reduction of

angiogenesis (Sheng 2006). Another example is the pigment epithelium derived factor (PEDF)

which promotes the expression of FasL, activating signal transduction pathways, in particular

caspases 8 and 3 leading to endothelial cell death (Tombran-Tink & Barnstable 2003). Unlike

maspin and PEDF, which inhibit proliferation by inducing apoptosis, OvUS had only a slight

effect on DNA fragmentation (apoptosis) of D17 and PC-3 cells.

There are two other serpins that inhibit cell proliferation, but by a caspase- independent

mechanisms. One is the intracellular serpin, myeloid and erythroid nuclear termination stage-

specific protein (MENT) which is involved in cell cycle progression and therefore in cell

Page 96: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

96

proliferation. This serpin inhibits the activity of the nuclear cysteine proteinase SPase, whose

function has been related to the degradation of the phosphorylated form of the retinoblastoma

(Rb) protein, a known regulator of the cell cycle (Irving et al. 2002a). The other serpin is the

intracellular plasminogen activator inhibitor type-2 (PAI-2). This protein could be involved in

the regulation of the cell cycle since it protected Rb from degradation by an independent anti-

proteolytic mechanism (Medcalf & Stasinopoulos 2005, Croucher et al. 2008). However, the

effects of OvUS on cell cycle progression as a mechanism to inhibit cell proliferation remain to

be determined.

Page 97: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

97

CHAPTER 4

REGULATION OF DNA SYNTHESIS AND THE CELL CYCLE IN HUMAN PROSTATE

CANCER CELLS AND LYMPHOCYTES BY OVINE UTERINE SERPIN

Introduction

Serine proteinase inhibitors (serpins) inactivate their target proteinases through a suicide

substrate-like inhibitory mechanism. The proteinase binds covalently to the reactive center loop

(RCL) of the serpin and cleaves the scissile bond at the P1-P1’ site. The RCL then moves to the

opposite side to form the β-sheet A and a distortion in the structure of the proteinase that results

in its inactivation (Irving et al. 2000, Van Gent et al. 2003, Law et al. 2006). Not all serpins,

however, exert proteinase inhibitory activity. Some examples are corticosteroid and thyroxine

binding globulins, which function as hormone transport proteins (Pemberton et al. 1988), the

chaperone heat shock protein 47 (Sauk et al. 2005), mammary serine protease inhibitor (Maspin),

which increases the sensitivity of cancer cells to undergo apoptosis (Sheng 2006), and pigment

epithelium derived factor (PEDF), which has neurotrophic, neuroprotective, antiangiogenic, and

proapoptoticactions (Fernandez-Garcia et al. 2007).

Another class of serpins without apparent proteinase activity is the uterine serpins. These

proteins, which are produced by the endometrial epithelium of the pregnant cow, sow, sheep, and

goat (Moffatt et al. 1987, Ing & Roberts 1989, Malathy et al. 1990, Leslie et al. 1990,

Mathialagan & Hansen 1996, Tekin et al. 2005a), have been classified as either a separate clade

of the serpin superfamily (Peltier et al. 2000c) or as a highly-diverge group of the α1-antitrypsin

clade (Irving et al. 2000). The best characterized protein of this unique group of serpins is ovine

uterine serpin (OvUS). This basic glycoprotein is a weak inhibitor of aspartic proteinases

(pepsin A and C) (Mathialagan & Hansen 1996, Peltier et al. 2000a), but it does not inhibit a

broad range of serine proteinases (Ing & Roberts 1989, Liu & Hansen 1995). Additionally,

amino acids in the hinge region of inhibitory serpins are not conserved in uterine serpins and

Page 98: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

98

OvUS behaves different in the presence of guanidine HCl than for inhibitory serpins (Peltier et

al. 2000a).

The biological function of OvUS during pregnancy may be to inhibit immune cell

proliferation during pregnancy and provide protection for the allogeneically-distinct conceptus

(Hansen 1998). Ovine US decreases proliferation of lymphocytes stimulated with concanavalin

A, phytohemagglutinin (PHA), Candida albicans, and the mixed lymphocyte reaction (Segerson

et al. 1984, Hansen et al. 1987b, Skopets & Hansen 1993, Skopets et al. 1995, Peltier et al.

2000b). In addition, OvUS decreases natural killer cell cytotoxic activity, abortion induced by

poly(I)•poly(C) in mice (Liu & Hansen 1993) and the production of antibody in sheep

immunized with ovalbumin (Skopets et al. 1995). The antiproliferative actions of OvUS are not

limited to lymphocytes. Ovine US decreases development of the bovine embryos and

proliferation of mouse lymphoma, canine primary osteogenic sarcoma and human prostate

cancer cell lines (Tekin et al. 2005b, 2006).

The mechanism by which OvUS inhibits proliferation of cells is unknown. The protein

could block activation of cell proliferation, inhibit the cell cycle at other points or induce

apoptosis or other forms of cell death. For the PC-3 prostate cancer line, inhibition of cell

proliferation by OvUS might involve reduction in interleukin-8 (IL-8) secretion because of the

importance of autosecretion of this cytokine for cell androgen-independent proliferation (Araki

et al. 2007). The goal of the present study was to evaluate the mechanism by which OvUS

inhibits cell proliferation. Using PC-3 cells as a model system, it was tested whether inhibition

of DNA synthesis involves cytotoxic action of OvUS, induction of apoptosis or disruption of the

IL-8 autocrine loop. It was also tested whether OvUS blocks specific steps in the cell cycle for

PC-3 cells and lymphocytes.

Page 99: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

99

Materials and Methods

Materials

The human prostate cancer (PC-3) cell line was purchased from ATCC (Rockville, MD),

the FreeStyleTM 293 expression medium, Dulbecco’s Modified Eagle Medium Nutrient Mixture

F-12 (DMEM-F12) and 0.25% Trypsin-EDTA were obtained from Gibco-Invitrogen (Carlsbad,

CA), the RQ1 RNase-free DNase and the CellTiter-Glo® Luminescent Cell Viability Assay kit

were obtained from Promega, (Madison, WI), the DHLTM Cell Cytotoxicity Assay kit was from

Anaspec (San Jose, CA) and the ELISA MAXTM Set Deluxe kit for human IL-8 was obtained

from BioLegend (San Diego, CA). The in situ cell death detection kit [terminal

deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL)] was purchased from

Roche (Indianapolis, IN), the DNase-free RNase A was obtained from Qiagen (Valencia, CA),

Precast Tris-HCl gradient Ready gels® were from BioRad (Richmond, CA) and [3H]thymidine

(6.7 Ci/mmol) was from ICN (Irvine, CA). The Prolong Antifade® kit was purchased from

Molecular Probes (Eugene, OR), Geneticin was from Research products international (Mount

Prospect, IL), Centricon filter devices were from Millipore Corporation (Bedford, TX), niquel

Sepharose chromatography medium (high performance) was from Amersham Biosciences

(Piscataway, NJ), fetal bovine and horse serum from Atlanta Biologicals (Norcross, GA). Other

reagents were obtained from either Fisher (Pittsburg, PA) or Sigma-Aldrich (St. Louis, MO).

Purification of rOvUS

The His-tagged rOvUS was purified from conditioned medium of FreeStyleTM human

embryonic kidney (HEK)-293F cells (Gibco-Invitrogen, Carlsbad, CA) transfected with a

plasmid construct containing the gene for OvUS. Details of the cell line are provided elsewhere

(Tekin et al. 2006). Cells were cultured continuously in selective medium [FreeStyleTM 293

expression medium containing 700 μg/ml of Geneticin®] at 37oC in a humidified 8% (v/v) CO2

Page 100: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

100

incubator according to the manufacturer’s recommendations. Conditioned medium containing

rOvUS was diluted 1:1 (v/v) in binding buffer [20 mM sodium phosphate buffer, 35 mM

imidazole, 0.3 M NaCl, pH 8.0] and loaded into a nickel Sepharose column that was pre-

equilibrated with binding buffer. The His-tagged rOvUS was eluted with 20 mM phosphate

buffer, 500 mM imidazole, 0.3 M NaCl, pH 8.0, concentrated and buffer-exchanged into

Dulbecco’s phosphate buffered saline (DPBS) using Centricon plus-20 concentration devices.

Purity of the rOvUS was assessed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis

using precast 4-15% polyacrylamide Tris-HCl gradient gels. The protein concentration was

determined by Bradford assay (Bradford 1976) using bovine serum albumin as standard.

For each experiment, rOvUS and the control protein, OVA, were added to culture wells of

PC-cells or lymphocytes dissolved in DPBS. The vehicle control included addition of DPBS at

the same volume as for rOvUS and OVA. The actual volume of protein or vehicle added varied

between experiments but was generally 14-25 μl and never more than 50 μl. Cultures were set up

so that the volume of DPBS was the same in all wells.

PC-3 Cell Culture

The PC-3 cell line was cultured continuously in Dulbecco’s Modified Eagle Medium

Nutrient Mixture F-12 (DMEM-F12) supplemented with 10 % (v/v) heat-inactivated fetal bovine

serum, 200 U/ml penicillin and 2 mg/ml streptomycin at 37oC in a humidified 5% (v/v) CO2

incubator. For the IL-8 experiment only, the medium was modified to reduce the fetal bovine

serum concentration to 4% (v/v). For all the experiments, cells were cultured in 75 cm2

flasks

until they reached 50-70% of confluence. Cells were then trypsinized, centrifuged at 110 x g for

5 min and resuspended in fresh complete medium. Cell viability was assessed by trypan blue

exclusion and cell concentration was adjusted according to the requirements of each experiment.

Page 101: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

101

[3H]thymidine Incorporation by PC-3 Cells

PC-3 cells (100 μl) were plated overnight at a final concentration of 1 x 105 cell/ml in a 96-

well plate. Afterwards, various concentrations of rOvUS (0, 0.5, 1, 2, 4, 8, 16, 32, 64, 125 and

250 μg/ml) or vehicle were added to each well in a total volume (including additional culture

medium) of 200 μl. After 48 h of culture, 0.1 μCi [3H]thymidine in 10 μl of culture medium

were added. Cells were harvested 24 h after [3H]thymidine addition onto fiber-glass filters using

a cell harvester (Brandel, Gaithersburg, MD). Filters were counted for radioactivity using

scintillation spectrometry (Beckman Coulter Inc., Fullerton, CA). Each concentration of protein

was tested in triplicate and the experiment was performed in six different replicates using a

different batch of rOvUS for each replicate.

Cell Proliferation Based on ATP Content

Aliquots of 50 μl of PC-3 cells (1 x 105 cells/ml) were cultured for 24 h in a dark wall-clear

bottom 96 well plate. Then, treatments consisting of vehicle (DPBS) or three different

concentrations (50, 100 and 200 μg/ml) of rOvUS or a control protein (ovalbumin; OVA) and

culture medium added to bring the final volume to 100 μl. Additional control wells without cells

were prepared to determine background. At 48 h after addition of treatments, ATP content per

well was determined using the CellTiter-Glo® Luminescent Cell Viability Assay kit according to

the manufacturer’s instructions. Briefly, 100 μl of the CellTiter-Glo® reagent were added to

each well, contents of the plate were mixed on a shaker for 2 min and then incubated at room

temperature for 10 min. Chemiluminescence was quantified using a multi-detection microplate

reader (FLX-800, BioTek, Winooski, VT). All treatments were performed in triplicates and the

assay was performed on three different occasions using a different batch of rOvUS for each

replicate.

Page 102: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

102

Cytotoxicity Assay

The assay was based on the release of lactate dehydrogenase into culture medium following

loss of cell membrane integrity accompanying cell death (Decker and Lohmann-Matthes, 1988).

Procedures for cell culture and treatments were similar to those described for the ATP assay. At

48 h after addition of the treatments, release of lactate dehydrogenase into the medium was

determined using the DHLTM Cell Cytotoxicity Assay kit following the vendor’s instructions.

Briefly, the plate was equilibrated at room temperature for 20 min before adding 10 μl of lysis

solution or DPBS. To facilitate cell lysing, the plate was placed on a shaker for 2 min. A total of

50 μl lactate dehydrogenase assay solution was then added to each well. After 10 min at room

temperature, the reaction was stopped using 20 μl of the stop solution and the fluorescence

intensity was measured using a multi-detection microplate reader (FLX-800) with excitation and

emission wavelengths of 530-560 nm and 590 nm, respectively. Percent cytotoxicity was

calculated by dividing 100 x fluorescence from the unlysed cells by fluorescence of the lysed

cells. For each assay, each treatment was performed in six wells. The assay was replicated five

different times using a different batch of rOvUS for each replicate.

TUNEL Labeling

An aliquot of 100 μl of PC-3 cells were cultured overnight in chamber slides at a final

concentration of 1 x 104 cells/ml. Then, treatments consisting of vehicle (DPBS), 50, 100 or 200

μg/ml rOvUS, or 200 μg/ml OVA were added and additional culture medium added to produce a

final volume of 300 μl. After 24 and 48 h in culture with treatments, cells were washed with

PBS/PVP [100 mM sodium phosphate pH 7.4, 0.9% (w/v) NaCl, 1 mg/ml polyvinyl pyrrolidone]

and fixed with 4% (w/v) paraformaldehyde for 1 h at room temperature. Cells then were washed

in PBS/PVP and stored at 4oC for the TUNEL (terminal deoxynucleotidyl transferase and

fluorescein isothiocyanate-conjugated dUTP nick end labeling) procedure.

Page 103: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

103

For TUNEL labeling, fixed cells were incubated for 1 h at room temperature with

permeabilization solution [PBS, pH 7.4, 0.1 (v/v) Triton X-100, 0.1% (w/v) sodium citrate).

After washing with PBS/PVP, slides were incubated with 50 μl of TUNEL reaction mixture

containing terminal deoxynucleotidyl transferase and fluorescein isothiocyanate-conjugated

dUTP, for 1 hour at 37oC. Positive controls were preincubated with RQ1 RNase-free DNase (50

U/ml) and negative controls were incubated without transferase. Slides were washed with

PBS/PVP, incubated for 1 h with 50 μg/ml of RNase A and then for 30 min with propidium

iodide (2.5 μg/ml) at room temperature. Slides were washed with PBS/PVP and Prolong

Antifade® was used to mount coverslips. Samples were observed using a Zeiss Axioplan 2

fluorescence microscope with dual filter (Carl Zeiss, Inc., Göttingen, Germany). Percent of cells

with DNA fragmentation was determined by counting the total number of nuclei and total

number of TUNEL-labeled nuclei at 10 different sites on the slide. The experiment was

performed using three different batches of rOvUS.

Secretion of IL-8

PC-3 cells (100 μl) were cultured in wells of a 96-well plate overnight at a final

concentration of 1 x 105 cells/ml. Treatments were then added including vehicle (DPBS, similar

volume as for rOvUS and OVA treatments), and three different concentrations of rOvUS and

OVA (50, 100 and 200 μg/ml). The volume of each well was brought to 200 μl with culture

medium. At 48 h after addition of treatments, cell culture supernatants were collected,

centrifuged and stored at -20oC until ELISA for IL-8. Treatments were performed in triplicate

for each assay; the experiment was repeated on three different occasions using three different

batches of the recombinant protein. For the measurement of IL-8, the ELISA MAXTM Set Deluxe

kit for human IL-8 was used according to the manufacturer’s instructions using 100 μl of

conditioned medium.

Page 104: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

104

Cell Cycle Analysis

PC-3 cells (100 μl) were cultured in 4 well plates at a final concentration of 4 x 105

cells/ml. After 24 h, treatments consisting of vehicle, 100 and 200 μg/ml rOvUS, and 200 μg/ml

OVA were added with additional culture medium for a total volume of 400 μl. At 12 and 24 h

after addition of treatments, cells were collected by trypsinization and washed with DPBS. Cells

were fixed overnight in 70% (v/v) ethanol at 4oC, washed with DPBS and resuspended with 500

μl of staining solution [DPBS pH 7.4, 0.1% (v/v) Triton X-100, 0.05 mg/ml DNase-free RNase

A, 50 μg/ml propidium iodide]. Cells were then analyzed by flow cytometry using a FACSort

flow cytometer (Becton Dickinson, Franklin Lakes, NJ) and the red fluorescence of single events

was recorded at wavelengths of 488 nm (excitation) and 600 nm (emission). Data were gated

using pulse width and pulse area to exclude doublets, and the percent of cells present in each

phase of the cell cycle was calculated using ModFITLT V3.1 software (Verity Software,

Topsham, ME). The experiment was performed on three occasions with five different batches of

rOvUS.

For the sheep lymphocyte experiment, mononuclear cells were purified by density gradient

centrifugation from the buffy coat of heparinized peripheral blood collected by jugular

venipuncture from non pregnant Rambouillet ewes (Tekin & Hansen 2002). After removing red

blood cells by incubation with red cell lysis buffer (0.01 M Tris-HCl pH 7.5 containing 8.3 g/L

of ammonium chloride), cell viability was assessed by trypan blue exclusion, and concentration

adjusted to 4 x 106 cells/ml. Cells were then suspended in a culture medium consisting of Tissue

Culture Medium-199 containing 5% (v/v) horse serum, 200 U/ml penicillin, 0.2 mg/ml

streptomycin, 2 mM glutamine and 10-5

M β-mercaptoethanol and aliquots of 100 μl cells

cultured in 4 well plates in the presence or absence of 4 μg/ml PHA and with treatments of

DPBS vehicle, 200 μg/ml rOvUS, and 200 μg/ml OVA. Total culture volume was 400 μl. After

Page 105: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

105

72 and 96 h in culture at 37oC in a humidified 5% (v/v) CO2 incubator, lymphocytes were

collected and washed with DPBS. Thereafter, lymphocytes were fixed and treated as described

above. The experiment was performed separately for lymphocytes from four different sheep.

Three different batches of rOvUS were tested for each sheep.

Statistical Analysis

Data were analyzed by least-squares means analysis of variance using the General Linear

Models Procedures of SAS (SAS System for Windows, Version 9.0; SAS Institute, Cary, NC,

USA). Error terms were determined based on calculation of expected mean squares with

replicate considered random and other main effects considered fixed. For the cytotoxicity and

IL-8 data, orthogonal polynomial contrasts were used to determine the linear and quadratic

effects of rOvUS and OVA. In other analysis, the pdiff mean separation test of SAS was used to

distinguish the difference of various levels of a treatment.

Results and Discussion

Proliferation of PC-3 cells

The antiproliferative effects of rOvUS on proliferation of PC-3 cells were evaluated by two

different assays. In the first experiment, it was shown that rOvUS caused a concentration-

dependent decrease in incorporation of [3H]thymidine into DNA (P<0.001) with the minimum

effective concentration being 8 μg/ml (Figure 4-1). The antiproliferative actions of OvUS using

[3H]thymidine uptake as the measure of proliferation has been demonstrated previously for PC-3

cells and other cell types (Segerson et al. 1984, Hansen et al. 1987b, Skopets & Hansen 1993,

Skopets et al. 1995, Peltier et al. 2000b, Tekin et al. 2005b, 2006). To confirm this effect of

rOvUS reflected an inhibition in cell proliferation and not a disruption in [3H]thymidine uptake

by the cells, antiproliferative effects were also evaluated by an assay in which the relative total

Page 106: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

106

Figure 4-1. Inhibition of [3H]thymidine incorporation of PC-3 cells by recombinant ovine

uterine serpin (rOvUS). The inset graph is provided to clarify the effects of rOvUS

at lower concentrations (≤ 8 μg/ml). Data represent least-squares means ± SEM.

Values that differ from untreated cells are indicated by asterisks (***P<0.001).

rOvUS concentration (μg/ml)

0 50 100 150 200 250 300

[3H

]th

ym

idin

e in

co

rpo

rati

on

(d

pm

)

1500

2000

2500

3000

3500

4000

4500

Protein concentration (μg/ml)

0 2 4 6 8 10

[3H

]thym

idin

e incorp

ora

tion (

dpm

)

2000

2500

3000

3500

4000

4500

***

***

***

***

***

***

rOvUS concentration (μg/ml)

0 50 100 150 200 250 300

[3H

]th

ym

idin

e in

co

rpo

rati

on

(d

pm

)

1500

2000

2500

3000

3500

4000

4500

Protein concentration (μg/ml)

0 2 4 6 8 10

[3H

]thym

idin

e incorp

ora

tion (

dpm

)

2000

2500

3000

3500

4000

4500

***

***

***

***

***

***

Page 107: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

107

number of cells per well was estimated by the ATP content per well. Treatment with rOvUS

reduced ATP content per well at all concentrations tested (50, 100 and 200 μg/ml) (Figure 4 -2).

In contrast, the control serpin, ovalbumin, did not cause effect in the ATP content per well. The

finding that rOvUS reduced ATP content per well confirms that the effects of OvUS to reduce

[3H]thymidine incorporation reflect a reduction in cell proliferation rather than interference with

[3H]thymidine transport into the cell.

Lactate Rehydrogenase Release

Possible cytotoxic effects of rOvUS on PC-3 cells were evaluated by measurements of

lactate dehydrogenase release into culture medium (Figure 4-3). None of the concentrations of

rOvUS or OVA tested caused an increase in the percent of lysed cells during culture. Thus,

rOvUS does not inhibit proliferation through induction of cell death.

DNA Fragmentation (Apoptosis)

The TUNEL procedure was used to test whether rOvUS decreased cell proliferation by

induction of DNA fragmentation characteristic of apoptosis and other forms of cell death.

Representative images of TUNEL labeled cells are shown in Figure 4-4 and the average percent

of cells that were TUNEL positive is shown in Figure 4-5. Treatment of PC-3 with either rOvUS

or the control protein OVA did not increase the percent of cells that were TUNEL positive at

either 24 or 48 h after treatment; the percentage of cells that were TUNEL positive was low for

all groups (< 5.7 %). The fact that rOvUS did not induce apoptosis makes the action of this

serpin distinct from that of two other serpins that inhibit cell proliferation. Both maspin (Sheng

2006) and PEDF (Fernandez-Garcia et al. 2007) are proapoptotic serpins.

Interleukin-8 Secretion

Interleukin-8 accumulation in the medium was measured because of the autocrine effect of

this chemokine on prostate cell proliferation (Araki et al. 2007). In addition, at least one class of

Page 108: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

108

Figure 4-2. Inhibition of proliferation of PC-3 cells by recombinant ovine uterine serpin

(rOvUS) as determined by ATP content/well. Ovalbumin (OVA) was used as a

negative control. Data represent least-squares means ± SEM. Means that differ

from untreated cells are indicated by symbols (†P<0.1; **P<0.01;***P<0.001).

AT

P C

on

ten

t p

er

well (

RL

U)

0

200

400

600

800

1000

1200

1400

1600

Control 50 100 200 10050 200

rOvUS (μg/ml) OVA (μg/ml)

**

***

Page 109: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

109

Figure 4-3. Lack of cytotoxic effect of recombinant ovine uterine serpin (rOvUS) on PC-3 cells

was measured by the release of lactate dehydrogenase. Ovalbumin (OVA) was

used as a control protein. Data represent least-squares means ± SEM.

Perc

en

t c

yto

tox

icit

y

0

5

10

15

20

25

30

0

5

10

15

20

25

30

100 200 10050 200

OVA (μg/ml)rOvUS (μg/ml)

Control 50

Page 110: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

110

Figure 4-4. Representative photomicrographs of PC-3 cells labeled using the TUNEL procedure

after 48 h of culture with either 100 (A) or 200 µg/ml (B) of rOvUS or 200 μg/ml of

the control protein ovalbumin. Cells in panel D were treated with DNAse as a

positive control.

C

E F

D

Page 111: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

111

Figure 4-5. Effect of recombinant ovine uterine serpin (rOvUS) on DNA fragmentation

(apoptosis) of PC-3 cells. Results show the percent of TUNEL positive cells at 24

(A) and 48 (B) h after addition of the treatments. Data represent least-squares

means ± SEM. Ovalbumin (OVA, 200 μg/ml) was used as control protein.

Pe

rce

nt

TU

NE

L p

os

itiv

e c

ells

0

5

10

15

20

rOvUS (μg/ml)

100 OVA50 200Control0

5

10

15

A

B

Page 112: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

112

molecule that inhibits PC-3 cell proliferation, soy isoflavones, also reduces IL-8 secretion

(Handayani et al. 2006). As shown in Figure 4-6, there was, however, no effect of rOvUS on

accumulation of IL-8 into conditioned cultured medium. Thus, rOvUS does not block PC-3 cell

proliferation through inhibition of IL-8 secretion.

Cell Cycle Dynamics

Dynamics through the different phases of the cell cycle were affected by the treatment of

PC-3 cells with rOvUS. Representative DNA histograms after treatment with vehicle or 200

μg/ml rOvUS are shown in Figures 4-7A and 4-7B while least-squares means ± SEM for for

results at 12 and 24 h after treatment are shown in Figures 4-7C and 4-7D, respectively. At 12 h

after addition of treatments, rOvUS decreased (P<0.01) the percent of cells in S phase and

increased (P<0.1 and P<0.05 for 100 and 200 μg/ml of rOvUS, respectively) the percent of cells

in the G2/M phase (Figure 4-7C). There was no effect of rOvUS on the percent of cells in G0/G1.

At 24 h after addition of treatment, 200 μg/ml rOvUS increased the percent of cells in G0/G1

(P<0.001), decreased the percent of cells in S phase (P<0.01), and did not affect the percent of

cells in G2/M phase (Figure 4-7D). Control of the cell cycle dynamics by rOvUS was also

evaluated in a second cell type - the peripheral blood lymphocyte. Representative DNA

histograms for PHA-treated lymphocytes are shown for control cells and cells treated with 200

μg/ml rOvUS in Figures 4-8A and 4-8B, respectively while least-squares means ± SEM are

shown in Figures 4-8C and 4-8D. At both 72 (Figure 4-8C) and 96 h (Figure 4-8D) after

addition of PHA, rOvUS increased (P<0.001) the proportion of lymphocytes in the G0/G1 phase

and decreased (P<0.05) the proportion of cells in the S phase. In contrast, there was no effect of

the control protein (OVA) on the distribution of cells in the cell cycle. These results indicate that

OvUS block cell proliferation through cell cycle arrest in both PC-3 and lymphocytes. The

differences in specific stages at which the cell cycle was blocked between PC-3 cells and

Page 113: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

113

Figure 4-6. Effect of recombinant ovine uterine serpin (rOvUS) on interleukin (IL) – 8

concentration in cell culture supernatants of PC-3 cells. Ovalbumin (OVA) was

used as control serpin. Data represent least-squares means ± SEM.

Protein concentration (μg/ml)

0 50 100 150 200 250

IL-

8 c

on

cen

tra

tio

n (

pg

/ml)

0

5000

10000

15000

20000

25000

rOvUS

OVA

Page 114: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

114

Figure 4-7. Cell cycle dynamics of PC-3 cells as affected by recombinant ovine uterine serpin

(rOvUS). Controls included vehicle (control) and ovalbumin (OVA).

Representative DNA histograms for analysis at 12 h after treatment with vehicle or

200 μg/ml rOvUS are shown in panels A and B, respectively. The least-squares

means for results of three separate assays are shown in panels C and D for analysis

at 12 h (C) and 24 h (D) after treatment. Bars with different superscripts differ (†

P<0.10, others at P<0.05 or less).

0

10

20

30

40

50

60

0

10

20

30

40

50

60

G0/G1 G2/M S

Perc

en

t o

f cell

s

10

20

30

40

50

0

10

20

30

40

50

0 Veh

icleO

VA

200rO

vUS 100

rOvU

S 200

Veh

icleO

VA

200rO

vUS 100

rOvU

S 200

Veh

icleO

VA

200rO

vUS 100

rOvU

S 200

D

C

S 14%

B

G2/M 25%

G0/G1 61%

rOvUS-12h

A

G2/M 20%

G0/G1 57%

S 23%

Control-12h a a a a

a

a a

a

a

a

a

a

a

a aa

ab†

b† b

b

b

bb

Page 115: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

115

lymphocytes is likely to be caused by differences in activation and regulation pathways for these

two cell types. Unlike PC-3 cells, lymphocytes are arrested at G0 until proliferation is induced

by addition of PHA. Inhibitions at points in the cell cycle other than G0/G1 are less likely to be

seen since few cells progress to later stages of the cell cycle. In addition, it is possible that

genetic mutations in PC-3 cells compromise some potential regulatory mechanisms. In

particular, unlike lymphocytes, PC-3 cells lack functional p53 (Isaacs et al. 1991) which causes

cell cycle arrest at G1/S by inducing p21cip1

that inhibits cyclin dependent kinases (Harper et al.

1993, Duliæ et al. 1994).

The mechanism by which OvUS inhibits cell cycle dynamics is not understood. One serpin

has been identified which can affect cell cycle regulatory proteins. This serpin, myeloid and

erythroid nuclear termination stage-specific protein (MENT), is a nuclear protein that inhibits

cell proliferation through interactions with a nuclear protein with papain-like cysteine proteinase

activity (Irving et al. 2002a). Inhibition of the proteinase prevents degradation of the cell cycle

protein Rb although antiproliferative effects may depend more on other actions of MENT to

mediate euchromatin condensation in an Rb-independent manner (Irving et al. 2002a). In any

case, OvUS, is apparently without proteinase inhibitory activity and is an extracellular protein

that is unlikely to achieve a nuclear localization. The antipepsin activity of OvUS is probably

not biologically significant. Ovine US is a very weak inhibitor of pepsin C [a 35 and 8 fold

molar excess of OvUS was required to inhibit pepsin A and C (Mathialagan & Hansen 1996)]

and the binding of OvUS to pepsin is electrostatic (Peltier et al. 2000a). Moreover, pepsin

shows an acidic pH optimum and is unlikely to be involved in cell proliferation under the

conditions utilized.

Page 116: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

116

Figure 4-8. Cell cycle dynamics of lymphocytes as affected by recombinant ovine uterine serpin

(rOvUS). Controls included vehicle (control), phytohemagglutinin (PHA) and

ovalbumin (OVA). Representative DNA histograms for analysis at 72 h after

treatment with PHA or 200 μg/ml rOvUS are shown in panels A and B,

respectively. The least-squares means for results of four separate assays are shown

in panels C and D for analysis at 72 h (C) and 96 h (D) after treatment. Bars with

different superscripts differ (P<0.05 or less).

Perc

en

t o

f C

ells

0

20

40

60

80

100

120

0

20

40

60

80

S

Veh

icle

OVA

200rO

vUS 200

Veh

icle

OVA

200rO

vUS 200

Veh

icle

OVA

200rO

vUS 200

G0/G1 G2/M

PH

A-C

ontro

l

PH

A-C

ontro

l

PH

A-C

ontro

l

C

D

S 25%

A

G0/G1 75%

Control-PHA 72h

B

G2/M 2%

G0/G1 91%

S 7%

PHA-rOvUS-72h

G2/M 0%

a a

b

b

a

b

b

b

b

c

b

a

b

c

aaaa

bc

a

b b

a

Page 117: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

117

A major point in the cell cycle regulated by OvUS is transition from G0/G1 to S phase:

rOvUS decreased the proportion of cells in S phase in all experiments and increased the

proportion of cells in G0/G1 at 24 h after treatment for PC-3 cells and at both times examined for

lymphocytes. Ovine uterine serpin can bind to cell membranes (Liu et al. 1999) and, perhaps,

OvUS inhibits proliferation by activation of signal transduction systems that inhibit transition

from G0/G1 to S phase or prevents pro-proliferative molecules in culture medium from binding

their receptors. Experiments with Rp-8-Cl-cAMPS, a selective inhibitor of cAMP-dependent

type-I protein kinase A, indicated that effects of OvUS on proliferation of PHA-stimulated

lymphocytes are not dependent on this kinase (Tekin et al. 2005b). Studies to determine

activation of other anti-proliferative signal transduction systems by OvUS are warranted.

Taken together, the present study indicates that the mechanism by which OvUS inhibits

proliferation of PC-3 cells and lymphocytes involves cell cycle arrest and not, at least for PC-3

cells, apoptosis, cytotoxicity or inhibition of IL-8 secretion.

Page 118: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

118

CHAPTER 5

CHANGES IN EXPRESSION OF CELL-CYCLE RELATED GENES IN PC-3 PROSTATE

CANCER CELLS CAUSED BY OVINE UTERINE SERPIN

Introduction

Uterine serpins (US), also known as uterine milk proteins (UTMP), are members of the

serine proteinase inhibitor (serpin) superfamily (Ing & Robets 1989, Mathialagan & Hansen

1996) and are designated as SERPINA14. These progesterone-induced glycoproteins are

secreted in large quantities into the uterus of a restricted group of mammals during pregnancy

(Malathy et al. 1990, Leslie et al. 1990, Tekin et al. 2005a). The best studied of the US is the

protein found in the sheep. Ovine uterine serpin (OvUS), which is the most abundant protein in

uterine secretions of the pregnant sheep (Moffatt et al. 1897, Hansen et al. 1987a), is an example

of a serpin that has gained a new function while apparently losing proteinase inhibitory activity

characteristic of serpins. Other examples include the heat shock protein 47 (Nagata 1998),

corticosteroid and thyroxine binding globulin (Pemberton et al. 1998) and angiotensinogen

(Morgan et al. 1996).

Inhibitory serpins inactivate their target proteinases by an irreversible suicide substrate-like

mechanism after the proteinase binds to the reactive center loop (RCL) (Silverman et al. 2001).

Usually, inhibitory serpins are recognized by a consensus sequence in the hinge region which is

localized within the RCL of the serpin (Irving et al. 2000) but the hinge region of OvUS is not

conserved with inhibitory serpins (Tekin et al. 2005a, Irving et al. 2000). Ovine US does not

inhibit cathepsins B, D and E (Mathialagan & Hansen 1996), dipeptidyl proteinase IV (Liu &

Hansen 1995), trypsin, chymotrypsin, plasmin, thrombin, elastase and plasminogen activator

(Ing & Robets 1989). While OvUS inhibits the aspartic proteinases pepsin A and C, this

inhibition is atypical for serpins since an excess of 35- and 8-fold molar of OvUS was required

for a 50% inhibition of pepsin A and C, respectively (Mathialagan & Hansen 1996).

Page 119: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

119

The role of OvUS during pregnancy has been linked to the protection of the allogeneic

conceptus against the maternal immune system (Hansen 1998). It exerts this role in large part by

inhibiting the proliferation of activated-lymphocytes (Hansen et al. 1987b, Peltier et al. 2000b).

The antiproliferative effect of OvUS is also exerted on some other cell types including mouse

lymphoma (P388D1), canine primary osteogenic sarcoma (D-17) and human prostate cancer

(PC-3) cell lines (Tekin et al. 2005a, 2006).

The mechanism by which OvUS inhibits cell proliferation is poorly understood. Ovine US

did not cause cytotoxic or apoptotic effects on lymphocytes or PC-3 cells (Tekin et al. 2005b,

Skopets & Hansen 1993, Padua & Hansen 2008). It was recently determined that OvUS blocks

cell cycle progression in mitogen-stimulated lymphocytes and increases the number of cells at

the G0/G1 stage at 96 h after addition of the protein (Padua & Hansen 2008). Ovine US also

blocks the progression of the cell cycle of PC-3 cells in a manner that leads to an accumulation

of cells at G2/M at 12 h after addition of the protein and at G0/G1 at 24 h after treatment (Padua

& Hansen 2008). The objective of the present study was to understand the mechanism by which

OvUS blocks cell cycle progression in PC-3 cells by determining cell-cycle related genes whose

expression is altered by OvUS.

Materials and Methods

Materials

The FreeStyleTM 293 expression medium, Dulbecco’s Modified Eagle Medium Nutrient

Mixture F-12 (DMEM-F12) and 0.25% Trypsin-EDTA were purchased from Gibco-Invitrogen

(Carlsbad, CA). The G418 disulfate (geneticin) was purchased from Research products

international (Mount Prospect, IL), nickel Sepharose chromatography medium (high

performance) from Amersham Biosciences (Piscataway, NJ), Precast Tris-HCl gradient Ready

gels® were obtained from BioRad (Richmond, CA) and Centricon® filter devices were from

Page 120: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

120

Millipore Corporation (Bedford, TX). The human prostate cancer (PC-3) cell line was from

ATCC (Rockville, MD), [3H]thymidine (6.7 Ci/mmol) was from ICN (Irvine, CA) and fetal

bovine and horse sera from Atlanta Biologicals (Norcross, GA). Other reagents were obtained

from either Fisher (Pittsburg, PA) or Sigma-Aldrich (St. Louis, MO).

Purification of rOvUS

Human embryonic kidney (HEK)-293F (Gibco-Invitrogen, Carlsbad, CA) cells transfected

with a plasmid construct containing the gene for OvUS (Tekin et al. 2006) were cultured

continuously in selective medium [FreeStyleTM 293 expression medium containing 700 μg/ml of

geneticin] at 37oC in a humidified incubator containing a gas environment of 8% (v/v) CO2 in air.

The rOvUS was purified by using immobilized metal ion (nickel) exchange chromatography as

described by Padua & Hansen, 2008. Briefly, rOvUS was eluted with 20 mM phosphate buffer,

500 mM imidazole, 0.3 M NaCl, pH 8.0, concentrated and buffer-exchanged into Dulbecco’s

phosphate buffered saline (DPBS) using Centricon plus-20® concentration devices. Sodium

dodecyl sulfate polyacrylamide gel electrophoresis under reducing conditions using 4-15%

polyacrylamide Tris-HCl gradient gels and Coomassie Blue were used to assess the purity of the

rOvUS. After filter-sterilization of rOvUS using 0.22 μm micro centrifuge devices, the

concentration of the protein was determined by Bradford assay (Bradford 1976) using bovine

serum albumin as standard.

PC-3 Cell Culture

The PC-3 cell line was cultured into 75 cm2

flasks continuously in complete medium

[Dulbecco’s Modified Eagle Medium Nutrient Mixture F-12 (DMEM-F12) supplemented with

10 % (v/v) heat-inactivated fetal bovine serum, 200 U/ml penicillin and 2 mg/ml streptomycin]

at 37oC in a humidified incubator with a gas environment 5% (v/v) CO2 in air. Cells were then

trypsinized after reaching 50-70% of confluence, centrifuged at 110 x g for 5 min and

Page 121: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

121

resuspended in fresh medium. The viability of the cells was assessed by trypan blue exclusion

and cell concentration was adjusted according to each experiment.

Proliferation Assay

PC-3 cells were cultured in 96 well plates in a volume of 100 μl and at a final concentration

of 1 x 104 cells/ml. After 24 h in culture, cells were treated with either rOvUS, ovalbumin

(OVA, a control serpin dissolved in DPBS) or vehicle (DPBS). The vehicle was added to control

wells at an equivalent volume (6-20 μl) as for the rOvUS and OVA. Additional culture medium

was added to all wells to bring the final volume to 200 µl. The final concentration of rOvUS and

OVA was 200 μg/ml. After 48 h of culture, an aliquot of 10 µl of culture medium containing 0.1

µCi [3H]thymidine was added to the wells. Cells were collected on fiber glass filters by using a

cell harvester (Brandel, Gaithersburg, MD) 24 h after [3H]thymidine addition. Radioactivity on

the filters was counted by scintillation spectrometry (Beckman Coulter Inc., Fullerton, CA). The

experiment was replicated on five different occasions using a total of four different batches of

rOvUS. For each replicate, each treatment was tested in triplicate.

Cell Culture for RNA Extraction

PC-3 cells were cultured in 4 well plates at a final concentration of 4 x 105 cells/ml in 100

μl. After 24 h of culture, treatments and complete medium were added to achieve a final

concentration of 200 μg/ml rOvUS or an equivalent volume of DPBS as experimental control in

a final volume of 400 μl. At 12 or 24 h after treatment addition, medium was removed from the

plates and cell lysed for total RNA cell extraction as described below. The experiment was

replicated four times, with a different batch of rOvUS for each replicate.

RNA Extraction

Total RNA was extracted using the RNeasy® plus micro kit (Qiagen Inc, Valencia, CA)

following the manufacturer’s instructions. Briefly, PC-3 cells were lysed in wells for 5 min by

Page 122: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

122

repeat pipetting using 350 μl of lysis buffer supplied in the kit. Cell lysates were transferred into

microcentrifuge tubes, vortexed for 1 min and placed into gDNA eliminator columns to remove

genomic DNA. After mixing with 70% (v/v) ethanol, samples were transferred RNeasy

MinElute spin columns, washed and total RNA eluted with RNase-free water. RNA

concentration and quality was determined by the Agilent 2100 BioAnalyzer (Agilent

Technologies, Santa Clara, CA) at the Gene Expression Core Laboratory of the Interdisciplinary

Center of Biotechnology Research, University of Florida. High-quality RNA was used for the

RT-PCR array experiments (RNA integrity numbers ≥ 8.0).

cDNA Synthesis and Real Time-PCR Array

The cDNA for each RNA sample was obtained by using the Super Array RT2 First Strand

kit (SABiosciences Corporation, Frederick, MD) according to the manufacturer’s instructions.

Briefly, after genomic DNA elimination, the reverse transcription reaction was performed at

42oC for 15 min and then heated at 95

oC for 5 min to inactivate the enzyme. The cDNA was

mixed with the RT2 SYBR green/ROX q PCR master mix (SABiosciences Corporation) and 25

µl aliquots were loaded into each well of the RT2 Profiler PCR Array (SA Biosciences

Corporation, catalog number PAHS-020A). The PCR array was designed to study the profile of

84 human cell-cycle related genes (Table 5-1). The PCR array experiments were performed on

an ABI 7300 instrument (Applied Biosystems, Foster City, CA). The conditions for

amplification were as follows: 1 cycle of 10 min at 95oC followed by 40 cycles of 15 sec at 95

oC

and 1 min at 60oC.

Statistical Analysis

The General Linear Models procedure of SAS (SAS System for Windows, Version 9.0;

SAS Institute, Cary, NC, USA) was used to analyze the data from the proliferation experiments

by the least-square means analysis of variance. All main effects were considered fixed and the

Page 123: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

123

Table 5-1. Cell cycle related genes screened using the RT2 Profiler

TM PCR Array.

1House keeping genes.

2Human Genomic DNA contamination control.

3Reverse Transcription control.

4Positive PCR control.

ABL1 ANAPC2 ANAPC4 DIRAS3 ATM ATR BAX BCCIP BCL2 BIRC5 BRCA1 BRCA2

CCNB1 CCNB2 CCNC CCND1 CCND2 CCNE1 CCNF CCNG1 CCNG2 CCNH CCNT1 CCNT2

CDC16 CDC2 CDC20 CDC34 CDK2 CDK4 CDK5R1 CDK5RAP1 CDK6 CDK7 CDK8 CDKN1A

CDKN1B CDKN2A CDKN2B CDKN3 CHEK1 CHEK2 CKS1B CKS2 CUL1 CUL2 CUL3 DDX11

DNM2 E2F4 GADD45A GTF2H1 GTSE1 HERC5 HUS1 KNTC1 KPNA2 MAD2L1 MAD2L2 MCM2

MCM3 MCM4 MCM5 MKI67 MNAT1 MRE11A NBN PCNA RAD1 RAD17 RAD51 RAD9A

RB1 RBBP8 RBL1 RBL2 RPA3 SERTAD1 SKP2 SUMO1 TFDP1 TFDP2 TP53 UBE1

B2M1 HPRT1 RPL13A1 GADPH1 ACTB1 HGDC2 RTC3 RTC3 RTC3 PPC4 PPC4 PPC4

Page 124: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

124

model included effects of treatments and batch of rOvUS. Differences between levels of a

treatment were determined by the pdiff mean separation test of SAS.

The PCR array data were analyzed by the ∆∆Ct method. Genes with Ct values greater than

35 cycles were considered as non-detectable and assigned a value of 35. The average of four

house keeping genes [Beta-2-microglobulin (B2M), Hypoxanthine phosphoribosyltransferase 1

(HPRT1), Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and β-actin (ACTB)] was used

to obtain the ∆Ct value for each gene of interest. The ∆∆Ct value for each gene was calculated

by the difference between the ∆Ct of the treated and the ∆Ct of the control groups. The fold-

change for each gene was calculated by 2- ∆∆Ct

and the statistical analysis to determine

differences between treatments was performed using the RT2 Profiler PCR Array Data Analysis

web-based software (SABiosciences Corporation).

Results

Inhibition of PC-3 Cell Proliferation by OvUS

The inhibitory effect of OvUS on the proliferation of PC-3 cells is shown in Figure 5-1.

Incorporation of [3H]thymidine into the DNA of PC-3 cells was reduced (p<0.05) by rOvUS. In

contrast, the control serpin OVA did not affect [3H]thymidine incorporation.

Cell-Cycle Related Gene Expression Profile at 12 h after Treatment with rOvUS

The mRNA expression of 17 genes was significantly altered by rOvUS (Table 5-2). Three

genes involved in cell cycle checkpoint and arrest were up-regulated. These genes were

CDKN1A (p21cip1

), CCNG2 (cyclin G2), and CDKN2B (p15ink

). In addition, the mRNA for 14

genes was decreased by rOvUS. Among these are 3 genes (MCM3, MCM5 and PCNA), whose

gene products are required at the S phase of the cell cycle for DNA synthesis and replication.

Others are genes involved in the regulation and progression at the M phase [CDC2, CKS2,

CCNH (cyclin H), BIRC5 (survivin), MAD2L1, MAD2L2] and at the G1 phase (CDK4, CUL1,

Page 125: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

125

Figure 5-1. Inhibition of [3H]thymidine incorporation of PC-3 cells by 200 μg/ml of the

recombinant ovine uterine serpin (rOvUS). Ovalbumin (OVA) was used as control

serpin and Dulbecco’s phosphate buffered saline (DPBS) as vehicle. Data represent

least-squares means ± SEM. Bars with different letters differ (p<0.05).

[3H

]th

ym

idin

e in

co

rpo

rati

on

(d

pm

)

0

5000

10000

15000

20000

25000

Vehicle(DPBS)

rOvUS OVA(200 μg/ml)

aa

b

(200 μg/ml)

[3H

]th

ym

idin

e in

co

rpo

rati

on

(d

pm

)

0

5000

10000

15000

20000

25000

Vehicle(DPBS)

rOvUS OVA(200 μg/ml)

aa

b

(200 μg/ml)

Page 126: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

126

Table 5-2. Regulation of cell-cycle related genes of PC-3 cells after 12 h of treatment with 200

µg/ml recombinant ovine uterine serpin.

a CDK2-associated dual specificity phosphatase

b Mitotin (S. cerevisiae)

c Cell division cycle 46 (S. cerevisiae)

Gene

Symbol Description AVG∆Ct ± SE

Fold

Change 95%CI P-value

Up-regulated Control Treat

CCNG2 Cyclin G2 7.545 ± 0.29 6.134 ± 0.31 2.6585 (1.12, 4.20) < 0.05

CDKN1A CDK inhibitor 1A ( p21, Cip1) 4.078 ± 0.05 2.727 ± 0.24 2.5502 (1.70, 3.40) < 0.01

CDKN2B CDK inhibitor 2B , p15 8.25 ± 0.36 7.037 ± 0.32 2.3184 (0.81, 3.83) < 0.05

Down-regulated

BIRC5 Survivin 6.885 ± 0.11 7.514 ± 0.16 0.6465 (0.47, 0.82) < 0.05

CCNH Cyclin H 3.783 ± 0.03 4.042 ± 0.06 0.8354 (0.76, 0.91) < 0.01

CDC2 Cell division cycle 2 1.45 ± 0.07 1.844 ± 0.06 0.7608 (0.67, 0.85) < 0.01

CDK4 Cyclin-dependent kinase 4 3.45 ± 0.06 3.942 ± 0.08 0.7111 (0.61, 0.81) < 0.01

CDKN3 Cyclin-dependent kinase inhibitor 3a 1.265 ± 0.05 1.519 ± 0.07 0.8384 (0.74, 0.94) < 0.05

CKS2 CDC28 protein kinase regulatory subunit 2 1.13 ± 0.07 1.437 ± 0.06 0.8084 (0.71, 0.91) < 0.05

CUL1 Cullin 1 4.375 ± 0.07 4.629 ± 0.06 0.8384 (0.74, 0.94) < 0.05

MAD2L1 MAD mitotic deficient-like 1 3.06 ± 0.06 3.622 ± 0.17 0.6774 (0.51, 0.85) < 0.05

MAD2L2 MAD mitotic deficient-like 2 4.593 ± 0.06 4.944 ± 0.09 0.7836 (0.66, 0.90) < 0.05

MCM3 Minichromosome maintenance deficient 3b 2.563 ± 0.17 3.632 ± 0.25 0.4765 (0.28, 0.67) < 0.05

MCM5 Minichromosome maintenance deficient 5c 5.965 ± 0.16 6.604 ± 0.05 0.642 (0.50, 0.79) < 0.05

PCNA Proliferating Cell Nuclear Antigen 2.225 ± 0.20 2.909 ± 0.08 0.6223 (0.44, 0.80) < 0.05

RAD1 RAD1 homolog (S. pombe) 7.41 ± 0.06 7.719 ± 0.07 0.807 (0.71, 0.91) < 0.05

RBBP8 Retinoblastoma Binding Protein 8 4.233 ± 0.15 4.927 ± 0.15 0.618 (0.44, 0.80) < 0.05

Page 127: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

127

CDKN3). The last two genes that were down-regulated were DNA damage checkpoint and

repair genes RBBP8 (retinoblastoma binding protein 8) and RAD1.

Cell-Cycle Related Gene Expression Profile at 24 h after Treatment with rOvUS

Treatment of PC-3 cells with rOvUS for 24 h caused down-regulation of 16 genes (Table 5-

3). Some of them (BIRC5, CDK4, CDKN3, CKS2, MAD2L2 and RAD1) were also down-

regulated at 12 h. The others are genes related to the regulation and progression through the M

(CCNB1, CDK5RAP1, CDC20 and E2F4) and G1 (CDK4 and TFDP2). Likewise, rOvUS

down-regulated three DNA damage checkpoint related genes (RAD17, KPNA2 and BRCA1)

whose activation blocks cell cycle progression at all stages of the cell cycle. The expression of

BCCIP (BRCA2 and CDKN1A interacting protein) a gene involved in DNA repair, spindle

formation and cytokinesis was also down-regulated by rOvUS. In addition, rOvUS caused the

down-regulation of MKI67. The gene product of MKI67 (ki-67) is a marker of cell proliferation.

Discussion

It was previously shown that OvUS inhibited cell proliferation of PC-3 cells by disrupting

cell cycle progression (Padua & Hansen 2008). The results presented in this study corroborate

those findings and provide an overview of the changes in gene expression that are associated

with alterations in the cell cycle. In particular, the inhibition of the cell cycle progression is

initially associated with increased expression of genes that block cell cycle and decreased

expression of genes needed for progression through G1, S and M phases. After more prolonged

treatment, the inhibition of expression of genes required for cell cycle progression is extended to

a wider range of genes.

In an earlier study, OvUS caused accumulation of PC-3 cells at the G2/M phase at 12 h

Page 128: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

128

Table 5-3. Down-regulation of human cell cycle-related genes of PC-3 cells after 24 h of

treatment with 200 µg/ml recombinant ovine uterine serpin.

Gene

Symbol Description AVG∆Ct ± SE

Fold

change 95%CI P-value

Down-regulated Control Treat

BCCIP BRCA and CDKN1A interacting protein 2.63 ± 0.16 3.17 ± 0.10 0.6881 (0.51, 0.87) < 0.05

BIRC5 Survivin 6.21 ± 0.09 7.32 ± 0.29 0.4643 (0.27, 0.65) < 0.05

BRCA1 Breast cancer 1, early onset 6.41 ± 0.16 7.29 ± 0.30 0.5445 (0.29, 0.80) < 0.05

CCNB1 Cyclin B1 1.10 ± 0.06 2.00 ± 0.30 0.537 (0.31, 0.76) < 0.01

CDC20 Cell Division Cycle 20 homolog 0.53 ± 0.20 1.55 ± 0.17 0.4916 (0.32, 0.67) < 0.01

CDK4 Cyclin-dependent kinase 4 2.88 ± 0.11 3.41 ± 0.12 0.6941 (0.54, 0.85) < 0.05

CDK5RAP1 CDK5 regulatory subunit associated protein 1 9.07 ± 0.19 9.65 ± 0.13 0.6704 (0.46, 0.88) < 0.05

CDKN3 Cyclin-dependent kinase inhibitor 3a 1.02 ± 0.01 1.68 ± 0.24 0.6354 (0.43, 0.84) < 0.05

CKS2 CDC28 protein kinase regulatory subunit 2 0.91 ± 0.08 1.64 ± 0.20 0.6042 (0.43, 0.78) < 0.05

E2F4 E2F transcription factor 7.14 ± 0.14 7.62 ± 0.07 0.7148 (0.57 0.86) < 0.05

KPNA2 Karyopherin α 2 (RAG cohort 1, importin α1) 1.97 ± 0.23 2.89 ± 0.25 0.5269 (0.29, 0.77) < 0.05

MAD2L2 MAD2 mitotic arrest deficient-like 2 3.99 ± 0.08 4.69 ± 0.04 0.6148 (0.54, 0.69) < 0.001

MKI67 Antigen identified by monoclonal antibody Ki-67 3.35 ± 0.24 4.48 ± 0.34 0.4579 (0.20, 0.72) < 0.05

RAD1 RAD1 homolog (S. pombe) 6.96 ± 0.06 7.33 ± 0.13 0.7741 (0.62, 0.92) < 0.05

RAD17 RAD17 homolog (S. pombe) 6.38 ± 0.06 6.56 ± 0.02 0.8831 (0.80, 0.96) < 0.05

TFDP2 Transcription factor Dp-2 7.21 ± 0.09 7.49 ± 0.04 0.8211 (0.71, 0.93) < 0.05 a CDK2-associated dual specific phosphatase.

Page 129: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

129

after treatment (Padua & Hansen 2008). It is clear from examination of Figure 5-2A that

different phases of the cell cycle are disrupted by OvUS at 12 h.

Ovine US caused an increase in mRNA expression of CDKN1A (p21cip1

), CDKN2B

(p15ink

) and CCNG2 (cyclin G2), all of which are involved in cell cycle checkpoint and arrest. It

is very likely that the upregulation of expression of these genes is the proximal cause for the

inhibition of cell cycle progression. CDKN1A for example, belongs to the cyclin dependent

kinase inhibitor (CDKI) family and inhibits cell cycle progression by inhibiting CDK2 and

CDK4 and by blocking DNA replication and repair by binding to PCNA (Harper et al. 1993,

Waga et al. 1994, Li et al. 1994, Cayrol et al. 1998). This inhibitor of cell cycle progression

causes arrest at G1, S and G2 phases (Harper et al. 1993, Krug et al. 2002). CDKN2B also

belongs to the CDKI family and binds to CDK4 and CDK6 to prevent their association with

cyclin D, thereby blocking the cell cycle at G1 (Krug et al. 2002). Finally, CCNG2 (cyclin G2) a

non-typical cyclin whose expression is independent of p53 (Bates et al. 1996) blocks cell cycle

progression at the G1/S phase by association with the active protein phosphatase 2A (Bennin et

al. 2002). The CCNG2 gene is also expressed at the late S and G2 phases (Le et al. 2007).

At 12 h, OvUS caused down-regulation of genes involved in DNA replication (PCNA,

MCM3 and MCM5) at the S phase and those implicated in the regulation and progression of the

cell cycle at M (CDC2, CCNH, MAD2L1, MAD2L2) and G1 (CDK4, CUL1, CDKN3) phases

(Figure 5-2A). As an example, CCNH (cyclin H) is the regulatory subunit of the cdk-activating

kinase (CAK) and is distinct from mitotic cyclins because is expressed constantly through the

cell cycle. The function of cyclin H is related to the phospholylation of different cyclin-

dependent kinases (CDKs) and components of the transcriptional machinery (Kaldis 1999). At

Page 130: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

130

Figure 5-2. Points in the cell cycle where genes were differentially regulated by ovine uterine

serpin at 12 and 24 h are represented in panel A and B, respectively. Up-regulated

genes are in green and down-regulated genes in red. Genes that block cell cycle

progression are underlined. Genes that were regulated at 12 and 24 h are shown

with an asterisk (*). Genes involved in DNA repair are in the center of the cycle to

represent that many of these are involved in several stages of the cell cycle.

G1

G2

S

M

S-phase

entry

M-phase

entry

MKI67

Anaphase

TelophaseMAD2L2*

BIRC5*

CDC20

CDK4*

CDKN3*

MKI67

CKS2*

CCNB1

CDK5RAP1

E2F4

MKI67

TFDP2

BCCIP G0

B

RAD1*

RAD17

BRCA1

KPNA2

BCCIP

G1

G2

S

M

S-phase

entry

M-phase

entry

MKI67

Anaphase

TelophaseMAD2L2*

BIRC5*

CDC20

CDK4*

CDKN3*

MKI67

CKS2*

CCNB1

CDK5RAP1

E2F4

MKI67

TFDP2

BCCIP G0

B

RAD1*

RAD17

BRCA1

KPNA2

BCCIP

G1

G2

S

M

S-phase

entry

M-phase

entry

CCNG2

CDKN1A

PCNA

MCM3

MCM5

Anaphase

TelophaseMAD2L1

MAD2L2*

BIRC5*

CDKN1A

CDKN2B

CDK4*

CDKN3*

CUL1

CDKN1A

CDC2

CCNH

CKS2*CCNG2

CDKN1A

CCNG2

A

PCNA

RAD1*

RBBP8

G1

G2

S

M

S-phase

entry

M-phase

entry

CCNG2

CDKN1A

PCNA

MCM3

MCM5

Anaphase

TelophaseMAD2L1

MAD2L2*

BIRC5*

CDKN1A

CDKN2B

CDK4*

CDKN3*

CUL1

CDKN1A

CDC2

CCNH

CKS2*CCNG2

CDKN1A

CCNG2

A

PCNA

RAD1*

RBBP8

Page 131: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

131

24 h, downregulation of cell cycle related genes became more widespread (Figure 5-2B). Some

genes whose mRNA were decreased at 12 h remained so at 24 h (BIRC5, CDK4, CDKN3,

CKS2, MAD2L2 and RAD1). Additional genes were also decreased at 24 h (CCNB1,

CDK5RAP1, CDC20, E2F4, TFDP2, RAD17, KPNA2 and BRCA1, BCCIP and MKI67).

Down-regulation of genes such as CCNB1, BIRC5, CDK4, CDC20 and TFDP2 would impede

progression through the G1, S and G2/M phases (Figure 5-2B). For example, CDC20 is the

activator of the anaphase-promoting complex/cyclosome (APC/C) required for the metaphase-

anaphase progression during mitosis (Baker et al. 2007). It was inhibited by 50% by OvUS.

The down-regulation of MKI67 at 24 h suggests that a proportion of PC-3 cells entered the

resting state (G0) since the gene product for MKI67 (ki-67) is a cell marker linked to

proliferation and is present in all stages of the cell cycle with the exception of the G0 stage

(Gerdes et al. 1984). This is consistent with the global decrease in gene expression observed at

24 h and also with earlier observations where OvUS caused an increase in the proportion of PC-3

cells at G0/G1 phase at 24 h after treatment (Padua & Hansen 2008).

Upregulation of expression of CDKN1A (p21cip

), CDKN2B (p15ink

) and CCNG2 (cyclin

G2) at 12 h is likely to be a cause for the down-regulation of genes at 12 and 24 h. It has been

shown that high levels of expression of CDKN1A (p21cip

) down-regulates the expression of

BIRC5 (survivin) in other cells (Lühr & Müritz 2003, Xiong et al. 2008). Also, it is possible that

the down-regulation of expression of some genes is the cause for the inhibition of transcription

of other genes. As an example, the down-regulation of CKS2 causes a reduction in the

transcription of CCNB1 (cyclin B1) and CDC2 (CDK1) (Martinsson-Ahlzén et al. 2008). The

activity of CDK1 is required for the regulation of some DNA repair pathways (Aylon et al. 2004,

Branzei & Foiani 2008).

Page 132: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

132

The lack of functional p53 on PC-3 cells (Isaacs et al. 1991) probably affects feedback

loops in response to OvUS. This protein is an effector molecule in the DNA repair pathway and

lack of p53 abolishes the DNA checkpoints response and apoptosis (Sancar et al. 2004, Gatz &

Wiesmüller 2006). Moreover, cells with disrupted p53 can overcome controls at the G2/M

checkpoint and fail to maintain a sustained arrest at this stage (Bunz et al. 1998).

All genes studied that are related to DNA damage checkpoints or repair (RBBP8, RAD1,

RAD17, BRCA1, KPNA2, PCNA and BCCIP) were down-regulated either at 12 or 24 h after

OvUS treatment. The products of these genes are induced in response to incomplete DNA

replication or damage at specific or several points of the cell cycle (Waga et al. 1994, Sancar et

al. 2004, Sartori et al. 2007, Teng et al. 2003, Lu et al. 2005). Thus, OvUS disrupts the

transcription of some genes involved in nucleotide excision, mismatch, homologous

recombination repair and translesion synthesis pathways in addition to the sensor (RAD1 and 17)

and mediator molecules (BRCA1) of the DNA damage checkpoint. The failure of OvUS to

induce apoptosis in PC-3 cells (Tekin et al. 2005b, Padua & Hansen 2008) despite these changes

in gene expression, could reflect the lack of p53 or an as yet undescribed anti-apoptotic action of

OvUS.

Ovine US is one of the few serpins identified that alters cell-cycle dynamics. The other is

the intracellular protein MENT which also has a very basic isoelectric point (9 versus 5-6.5 for

other serpins) (Silverman et al. 2001). MENT inhibits the enzymatic activity of the nuclear

cysteine proteinase SPase, a cathepsin L-like proteinase involved in the degradation of the

phosphorylated form of the retinoblastoma (Rb) protein, a known regulator of the cell cycle

(Irving et al. 2002a). Another intracellular serpin, PAI-2, can protect Rb from degradation by an

independent anti-proteinase mechanism (Croucher et al. 2008). Unlike MENT and PAI-2, OvUS

Page 133: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

133

is an extracellular protein that can bind to cell membranes (Liu et al. 1999). Nonetheless, it is

possible that OvUS inhibits cell proliferation by being internalized. Other extracellular serpins

can become internalized and affect cell function. An example is α1-antitrypsin, which enters and

resides in the cytoplasm of a mouse insulinoma cell line and protects against apoptosis through

inhibition of caspase-3 activation (Zhang et al. 2007).

Alternatively, OvUS blocks cell proliferation through blockage or induction of signal

transduction systems. In lymphocytes, OvUS inhibits proliferation of phorbol myristol acetate

stimulated lymphocytes, suggesting that the protein blocks downstream actions of the protein

kinase (PK) C pathway (Peltier et al. 2000b). Like OvUS, transforming growth factor (TGF)-β

causes cell cycle arrest by upregulation of CDKN2B (p15ink

), CDKN1A (p21cip1

) and CCNG2

(cyclin G2) (Horne et al. 1997, Gartel &d Tyner 2002). Interferon (IFN)-γ also induces

CDKN1A (p21cip1

) expression, independent of p53 by the STAT-1 pathway (Gartel & Tyner

2002). Both, TGF-β and IFN-γ signaling can cause transactivation of p21 by a p53-independent

mechanism, where p21 protects cells against p53-independent apoptosis which is induced by

these signals (Gartel & Tyner 2002). Signal transduction pathways affected by OvUS have not

been determined, but OvUS may regulate components of signaling pathways shared with TGF-β

or IFN-γ pathways.

In summary, OvUS inhibits proliferation of PC-3 cells through disruption of the cell cycle

dynamics. Disruption involves increased expression of cell-cycle checkpoint and arrest genes

CDKN1A (p21cip1

), CDKN2B (p15ink

) and CCNG2 (cyclin G2) and down-regulation of genes

involved in cell-cycle progression.

Page 134: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

134

CHAPTER 6

GENERAL DISCUSSION

The serpin genes are an ancient origin gene family, being identified in bacteria, fungus,

nematodes, archaea and virus (Irving et al. 2000, 2002b, Steenbakkers et al. 2008). Evidence

presented in this dissertation strongly indicates that one of the subclades in this superfamily, the

uterine serpins, evolved only within a restricted group of mammals, the Laurasiatheria

superorder of eutherian mammals. The uterine serpins are thus an example of a new gene arising

from gene duplication and selection for sequence divergence (Louis 2007) that plays a particular

role in pregnancy in a group of mammals with unique attributes for gestation (an epitheliochorial

placenta).

The distribution of known uterine serpins as well as orders where a uterine serpin was not

identified is shown in Figure 2-9. All the identified uterine serpins are in Ruminatia (sheep,

goat, water buffalo and cow), Suidae (pig), Perisodactyla (horse) or Carnivora (dog) orders of

Laurasiatheria. An important question is whether uterine serpin genes are in the orders within

the Laurasiatheria superorder (Cetacea, Hippopotamidae, Pholidota, Chiroptera,

Erinaceomorpha, Scalopus and Talpa). Most of these orders have epitheliochorial placenta

although the Chiroptera, Erinaceomorpha and Talpa orders have either endotheliochorial or

hemochorial type of placentation. Experiments in Chapter 2 strongly suggested that the uterine

serpin gene is modified (dog) or become a pseudogene (cat) in carnivore species with

endotheliochorial placenta. Examination of the situation in species of the remaining orders of

the Laurasiatheria superorder with either endotheliochorial or hemochorial placenta would allow

determination of whether similar changes in uterine serpin genes are occurring within these

orders. If the gene is either lost or modified in species of the Chiroptera, Erinaceomorpha and

Page 135: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

135

Talpa orders, the idea that uterine serpins play a specific role required for pregnancy in species

with epitheliochorial type of placentation would be strengthened.

As indicated in Figure 2-9, there is an additional order of animals having epitheliochorial

placenta that is not within Laurasiatheria, the lemurs and lorises of the order Lemuriformes

within the Euarchontoglires superorder. Identification of the uterine serpin gene in this species

would indicate that the genes predate the diversion of Laurasiatheria from Euarchontoglires

superorder and that epitheliochorial placentation is absolutely dependent upon the uterine serpin

genes. Peltier et al. (2000c) estimated that uterine serpin genes diverged from CBG before the

divergence of mammals so much a result remains possible. It is more likely, however, that

uterine serpins will not be found in Lemuriformes since, even if the uterine serpin gene is older

than the common ancestor of Laurasiatheria and Euarchontoglires, the gene would be likely be

lost in the ancestors of lemurs and lorises that did not have epitheliochorial placenta.

The results from the positive (Darwinian) selection study reported in Chapter 2 strongly

suggest that there is pressure on the evolution of the uterine serpin gene to affect its

functionality. Positive selection has occurred at the RCL region of the inhibitory serpins, in

particular at the P1 site proximal to the scissile bond of the RCL, which changes the selectivity

for the target proteinase (Brown 1987, Hill & Hastie 1987, Goodwin et al. 1996, Zang & Maizels

2001, Barbour et al. 2002). The uterine serpins apparently lack anti-proteinase activity (Ing &

Roberts 1989, Liu & Hansen 1995, Mathialagan & Hansen 1996, Peltier et al. 2000a; see also

Chapter 2 for lack of conservation at residues important for anti-proteinase activity). There is

also evidence that the uterine serpin gene has evolved different functions within mammals with

the gene. It is possible that the uterine serpin gene in species with epitheliochorial placentation

has been retained as a secretory protein while the gene in species with endotheliochorial

Page 136: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

136

placentation has either been changed to become an intracellular protein (dog) or to be lost (cat).

If this idea is correct, uterine serpins play an important role in the uterine lumen of species with

epitheliochorial placentation that is not required for species with endotheliochorial placentae.

Even within species with epitheliochorial placenta, there might be different functions. The

PoUS-1 and -2, for example, regulate iron stability in uteroferrin and may be involved in iron

transport to the fetus (Baumbach et al. 1989, Roberts & Bazer 1988) while the OvUS can inhibit

proliferation of lymphocytes and certain other cell types (Tekin et al. 2005b, Padua & Hansen

2008; also see Chapters 3, 4 and 5).

Inhibition of lymphocyte proliferation has been interpreted as signifying a role for uterine

serpins in prevention of rejection of conceptus tissue by the maternal immune system (Hansen

1998). The sheep protein OvUS for example, has been shown to inhibit proliferation of mitogen-

stimulated lymphocytes, NK cell activity and a variety of cancer cells (Segerson et al. 1984,

Hansen et al. 1987b, Stephenson et al. 1989b, Skopets & Hansen 1993, Skopets et al. 1995,

Padua & Hansen 2008). In addition, the limited invasiveness of trophoblast tissue in species

with epitheliochorial placentatation could be achieved, at least in part, by inhibition of

trophoblast proliferation by uterine serpin. It may also be that uterine serpins participate in the

process of trophoblast binucleate cell formation, a phenomenon characterized by nuclear

endoreduplication without proper cytokinesis, which occurs in sheep, goat, cattle and horse

(Hoffman & Wooding 1993).

Ovine uterine serpin acts to inhibit cell proliferation by inducing changes in gene

expression of proteins involved in the cell cycle and, most notably, the up-regulation of cell

cycle inhibitors such as CDKN1A (p21cip

), CDKN2B (p15ink

) and CCNG2 (cyclin G2) (Chapter

5). These cell cycle inhibitors have been related to differentiation of stromal cells into decidual

Page 137: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

137

cells, formation of polyploidy stromal cells as well as terminal differentiation and apoptosis of

luminal and stromal cells during decidualization (Tan et al. 2002, Yao et al. 2003, Yue et al.

2005, Li et al. 2008).

Ovine US is one of the few serpins identified that inhibits cell proliferation by blocking

cell-cycle progression (Chapter 4). In PC-3 cells, OvUS decreased the percent of cells in S phase

and increased the percent of cells in the G2/M phase at 12 h after treatment addition. At 24 h,

OvUS increased the percent of cells in G0/G1 and decreased the percent of cells in S phase. In

the mitogen (PHA) stimulated lymphocyte, at both 72 and 96 h after stimulation, OvUS

increased the proportion of lymphocytes in the G0/G1 phase and decreased the proportion of cells

in the S phase (Chapter 4). There are two other serpins that can alter cell cycle progression. One

is the intracellular protein MENT which also inhibits cell proliferation by blocking cell cycle

progression, inhibiting the enzymatic activity of the nuclear cysteine proteinase SPase, a

cathepsin L-like proteinase involved in the degradation of the phosphorylated form of the Rb

protein, a known regulator of the cell cycle (Irving et al. 2002a). The other is the intracellular

serpin, PAI-2 that can protect Rb from degradation by an independent anti-proteinase mechanism

(Croucher et al. 2008).

The signal transduction pathway activated or down-regulated by OvUS remains to be

determined. Unlike MENT and PAI-2, OvUS is an extracellular protein that can bind to cell

membranes (Liu et al. 1999). Ovine US could either regulate signal transduction pathways by

binding to a specific membrane receptor or by becoming internalized into the cell and interacting

with intracellular binding partners (Figure 6-1). It is possible that OvUS inhibits cell

proliferation by being internalized through endocytosis. Other extracellular serpins can become

internalized and affect cell function. An example is α1-antitrypsin, which enters and resides in

Page 138: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

138

Figure 6-1. Possible pathways by which OvUS could block cell proliferation. A) OvUS

could bind to its own receptor or to another ligand’s receptor and activate a

pathway that blocks cell cycle progression. B) OvUS could block the binding

site of a ligand needed for cell cycle progression so that the pathway for

activation is not stimulated. C) OvUS could become internalized and block

cell cycle progression after internalization by binding too and disrupting

intracellular binding partners that are the effectors of the cell cycle arrest.

Yellow circles represent the phosphorylated status of the proteins in the signal

transduction pathway.

?

?

?

Nucleus

G1/S

Cytoplasm

OvUS

?

?

AB

C

p21p15

Cyclin G2

G2/M

?

?

?

Nucleus

G1/S

Cytoplasm

OvUS

?

?

AB

C

p21p15

Cyclin G2

G2/M

Page 139: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

139

the cytoplasm of a mouse insulinoma cell line and protects against apoptosis through inhibition

of caspase-3 activation (Zhang et al. 2007). Alternatively, OvUS could bind to a specific cell

surface receptor or compete with other molecules for a receptor binding site (Figure 6-1), like

α1-antitrypsin which competes with diferric transferrin for the trasnferrin receptors inhibiting the

proliferation of human skin fibroblasts (Graziadei et al. 1988). In lymphocytes, OvUS inhibits

proliferation of phorbol myristol acetate stimulated lymphocytes, suggesting that the protein

blocks downstream actions of the PKC pathway (Peltier et al. 2000b). OvUS could regulate

components of signaling pathways shared with TGF-β or IFN-γ pathways. Like OvUS, TGF-β

causes cell cycle arrest by upregulation of CDKN2B (p15ink

), CDKN1A (p21cip1

) and CCNG2

(cyclin G2) (Horne et al. 1997, Gartel & Tyner 2002). Interferon-γ also induces CDKN1A

(p21cip1

) expression by the STAT-1 pathway (Gartel & Tyner 2002). Thus, it is possible that

uterine serpins are involved in the process of trophoblast binucleate cell formation and/or uterine

luminal and stromal cell differentiation.

In the dog (Chapter 2) as well as in the cow (Khatib et al. 2007), the uterine serpin gene

was recently identified to be expressed in the ovary. The presence of uterine serpin in other

reproductive tissues distinct to the uterus raises the possibility that perhaps uterine serpins may

have different functions according to the cell type and stage of differentiation. One possibility is

that uterine serpin functions in the ovary to regulate follicular growth by binding to activin.

Activin improves follicular growth and granulosa cell proliferation (Knight & Glister 2006).

OvUS can also this member of the TGF-β family (McFarlane et al. 1999). However, the binding

affinity of activin for OvUS is much lower than activin affinity towards follistatin.

It is also possible that OvUS is participating in the differentiation of granulose cells into

luteal cells after ovulation. In the ovary, granulosa cells luteinize under the influence of

Page 140: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

140

luteinizing hormone. The expression of cell cycle inhibitors has been involved in the inhibition

of proliferation of differentiating granulosa cells in mice, specifically the CDK inhibitors

p21Cip1/Waf1

and p27Kip1

which are up-regulated throughout this period (Burns et al. 2001,

Jirawatnotai et al. 2003). Likewise, p15ink4b

, another CDK inhibitor has been linked with

luteinization and granulosa cell differentiation (Burns et al. 2001). Results presented in this

dissertation showed that OvUS blocked cell cycle progression and up-regulated the expression of

the CDK inhibitors CDKN1A (p21cip1

) and CDKN2B (p15ink

) and also caused the down-

regulation of MKI67 (Chapter 5). The gene product of MKI67 (Ki-67) is a marker of cell

proliferation and is present in all stages of the cell cycle with the exception of the G0 stage

(Gerdes et al. 1984).

In summary, the uterine serpin gene is present only in a restricted group of species within

the Laurasiatheria superorder of eutherian mammals and likely evolved under positive selection

which suggests diversifying functionality of the protein within these species. The finding that

the uterine serpin has been retained as a secretory protein in most species with epitheliochorial

placentation within the Laurasiatheria superorder also suggests that the protein has an important

role during pregnancy in species with this kind of placenta. The finding that OvUS inhibits cell

proliferation by blocking cell cycle progression, specifically by the upregualtion of cell cycle

inhibitors suggests other possible functions for uterine serpins in reproductive tissue, in

particular trophoblast binucleate cell formation, uterine cell differentiation and granulosa cell

differentiation in the ovary.

Page 141: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

141

LIST OF REFERENCES

Anisimova M, Bielawski JP & Yang Z 2001 Accuracy and power of likelihood ratio test to

detect positive selection at amino acid sites. Mol Biol Evol 18 1585-1592.

Araki S, Omori Y, Lyn D, Singh RK, Meinbach DM, Sandman Y, Lokeshwar, VB &

Lokeshwar BL 2007 Interleukin-8 is a molecular determinant on androgen independence

and progression in prostate cancer cells. Cancer Res 67 6854-6862.

Atchley WR, Lokot T, Wollenberg K, Dress A & Ragg H 2001 Phylogenetic analyses of

amino acid variation in the serpin proteins. Mol Biol Evol 18 1502-1511.

Aylon Y, Liefshitz B & Kupiec M 2004 The CDK regulates repair of double-strand breaks by

homologous recombination during the cell cycle. EMBO J 23 4868-4875.

Baker DJ, Dawlaty MM, Galardy P & van Deursen JM 2007 Mitotic regulation of the

anaphase-promoting complex. Cell Mol Life Sci 64 589-600.

Bates S, Rowan S & Vousden KH 1996 Characterization of human cyclin G1 and G2: DNA

damage inducible genes. Oncogene 13 1103-1109.

Bauersachs S, Ulbrich SE, Gross K, Schmidt SEM, Meyer HHD, Einspanier R,

Wenigerkind H, Vermehren M, Blum H, Sinowatz F & Wolf E 2005 Gene expression

profiling of bovine endometrium during the oestrus cycle: detection of molecular pathways

involved in functional changes. J Mol Endocrinol 34 889-908.

Barbour KW, Goodwin RL, Guillonneau F, Wang Y, Baumann H & Berger FG 2002

Functional diversification during evolution of the murine α1-proteinase inhibitor family:

role of hypervariable reactive center loop. Mol Biol Evol 19 718-727.

Baumbach GA, Ketcham CM, Richardson DE, Bazer FW & Roberts RM 1986 Isolation and

characterization of a high molecular weight stable pink form of uteroferrin from uterine

secretions and allantoic fluid of pigs. J Biol Chem 261 12869-12878.

Baumbach GA, Sanders PTK, Bazer FW & Roberts RM 1984 Uteroferrin has N-asparagine-

linked high-mannose-type oligosaccharides that contain mannose 6-phosphate. Proc Natl

Acad Sci USA 81 2985-2989.

Bazer FW, Roberts RM, Basha SM, Zavy MT, Caton D & Barron DH 1979 Method for

obtaining ovine uterine secretions from unilaterally pregnant ewes. J Anim Sci 49 1522-

1527.

Benarafa C & Remold-O’Donnell E 2005 The ovalbumin serpins revisited: perspective from

the chicken genome of clade B serpin evolution in vertebrates. Proc Natl Acad Sci USA

102 11367-11372.

Page 142: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

142

Bendtsen JD, Nielsen H, von Heijne G & Brunak S 2004 Improved prediction of signal

peptides: SignalP 3.0. J Mol Biol 340 783-795.

Bennin DA, Don ASA, Brake T, McKenzie L, Rosenbaum H, Ortiz L, DePaoli-Roach AA &

Horne MC 2002 Cyclin G2 associates with protein phosphatase 2A catalytic and

regulatory B’ subunits in active complexes and induces nuclear aberrations and a G1/S

phase cell cycle arrest. J Biol Chem 277 27449-27467.

Bielawski JP & Yang Z 2003 Maximum likelihood methods for detecting adaptive evolution

after gene duplication. J Struct Funct Genomics 3 201–212.

Binder BR, Mihaly J & Prager GW 2007 uPAR-uPA-PAI-1 interactions and signaling: a

vascular biologist’s view. Thromb Haemost 97 336-342.

Bradford MM 1976 A rapid and sensitive method for the quantitation of microgram quantities

of protein utilizing the principle of protein-dye binding. Anal Biochem 78: 248-254.

Branzei D & Foiani M 2008 Regulation of DNA repair throughout the cell cycle. Nat Rev Mol

Cell Biol 9 297-308.

Breit SN, Luckhurst E & Penny R 1983 The effect of α1-antitrypsin on the proliferative

response of human peripheral blood lymphocytes. J Immunol 130 681-686.

Brooks MA, Ali AN, Turner PC & Moyer RW 1995 A rabbitpox virus serpin gene controls

host range by inhibiting apoptosis in restrictive cells. J Virol 69 7688-7698.

Brown AL 1987 Positively darwinian molecules? Nature 326 12-13.

Buhi WC, Ducsay CA, Bazer FW & Roberts RM 1982 Iron transfer between the purple

phosphatase uteroferrin and transferring and its possible role in iron metabolism of the fetal

pig. J Biol Chem 257 1712-1723.

Bunz F, Dutriaux A, Lengauer C, Waldman T, Zhou S, Brown JP, Sedivy JM, Kinzler KW

& Vogelstein B 1998 Requirement for p53 and p21 to sustain G2 arrest after DNA damage.

Science 282 1497-1501.

Burns KH, Yan C, Kumar TR & Matzuk MM 2001 Analysis of ovarian gene expression in

follicle-stimulating hormone β knockout mice. Endocrinology 142 2742-2751

Cayrol C, Knibiehler M & Ducommun B 1998 p21 binding to PCNA causes G1 and G2 cell

cycle arrest in p53-deficient cells. Oncogene 16 311-320.

Callus BA & Vaux DL 2007 Caspase inhibitors: viral, cellular and chemical. Cell Death Differ

14 73-78.

Page 143: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

143

Chen TT, Bazer FW, Gebhardt BM & Roberts RM 1975 Uterine secretion in mammals:

synthesis and placental transport of a purple acid phosphatase in pigs. Biol Reprod 13 304-

313.

Chenna R, Sugawara H, Koike T, Lopez R, Gibson TJ, Higgins DG & Thompson JD 2003

Multiple sequence alignment with the Clustal series of programs. Nucleic Acids Res 31

3497-3500.

Cole EB, Miller D, Romero D, Greenberg RM, Brömme D, Çatalpete S, Pak SC, Mills DR,

Silverman GA & Luke CJ 2004 Identification and activity of a lower eukaryotic serine

proteinase inhibitor (serpin) from Cyanea capillata: analysis of a jellyfish serpin, jellypin.

Biochem 43 11750-11759.

Congote LF 2007 Serpin A1 and CD91 as host instruments against HIV-1 infection: are

extracellular antiviral peptides acting as intracellular messengers. Virus Res 125 119-34.

Crisp RJ, Knauer DJ & Knauer MF 2000 Roles of the heparin and low density lipid receptor-

related protein-binding sites of protease nexin 1 (PN1) in urokinase-PN1 complex

catabolism. J Biol Chem 275 19628-19637.

Croucher D, Saunders DN, Lobov S & Ranson M 2008 Revisiting the biological roles of

PAI2 (SERPINB2) in cancer. Nature Rev Cancer 8 535-545.

Croucher D, Saunders DN & Ranson M 2006 The urokinase/PAI-2 complex. J Biol Chem

281 10206-10213.

Ducsay CA, Buhi WC, Bazer FW & Roberts RM 1982 Role of uteroferrin in iron transport

and macromolecular uptake by allantoic epithelium of the porcine conceptus. Biol Reprod

26 729-743.

Ellery JM & Nicholls PJ 2002 Alternate signaling pathways from the interleukin-2 receptor.

Cytokine Growth Factor Rev 13 27-40.

Felsenstein J 1985 Confidence limits on phylogenies: An approach using the bootstrap.

Evolution 39 783-791.

Felsenstein J 1993 PHYLIP (Phylogeny Inference Package) version 3.5c. Distributed by the

author. Department of Genetics, University of Washington, Seattle.

Felsenstein, J 1989 PHYLIP -- Phylogeny Inference Package (Version 3.2). Cladistics 5 164-

166.

Fernandez-Garcia NI, Volpert OV & Jimenez B 2007 Pigment epithelium-derived factor as a

multifunctional antitumor factor. J Mol Med 85 15-22.

Page 144: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

144

Gartel AL & Tyner AL 2002 The role of the cyclin-dependent kinase inhibitor p21 in

apoptosis. Mol Cancer Ther 1 639–649.

Gatz SA & Wiesmüller L 2006 p53 in recombination and repair. Cell Death Differ 13 1003-

1016.

Gerdes J, Lemke H, Baisch H, Wacker HH, Schwab U & Stein H 1984 Cell cycle analysis of

a cell proliferation-associated human nuclear antigen defined by the monoclonal antibody

Ki-67. J Immunol 133 1710-1715.

Goodwin RL, Baumann H & Berger FG 1996 Patterns of divergence during evolution of α1-

proteinase inhibitors in mammals. Mol Biol Evol 13 346-358.

Gray CA, Stewart MD, Johnson GA & Spencer TE 2003 Postpartum uterine involution in

sheep in endometrial gene expression. Reproduction 125 185-198.

Graziadei I, Kähler CM, Wiedermann CJ & Vogel W 1998 The acute-phase protein α1-

antitrypsin inhibits trasferrin-receptor binding and proliferation of human skin fibroblasts.

Biochim Biophys Acta 1401 170-176.

Green JA, Xie S & Roberts RM 1998 Pepsin-related molecules secreted by trophoblast. Rev

Reprod 3 62-69.

Gupta VK & Gowda LR 2008 Alpha-1-proteinase inhibitor is a heparin binding serpin;

molecular interactions with Lys rich cluster of helix-F domain. Biochimie 90 749-761.

Handayani R, Rice L, Cui Y, Medrano TA, Samedi VG, Baker HV, Szabo NJ & Shiverick

KT 2006 Soy isoflavones alter expression of genes associated with cancer progression,

including interleukin-8, in androgen-independent PC-3 human prostate cancer cells. J Nutr

136 75-82.

Hansen PJ 1998 Regulation of uterine immune function by progesterone-lessons from the

sheep. J Reprod Immunol 40 63-79.

Hansen PJ & Newton GR 1988 Binding of immunoglobulins to the major progesterone-

induced proteins secreted by the sheep uterus. Arch Biochem Biophys 260 208-217.

Hansen PJ, Ing NH, Moffatt RJ, Baumbach GA, Saunders PTK, Bazer FW & Roberts RM

1987a Biochemical characterization and biosynthesis of the uterine milk proteins of the

pregnant sheep uterus. Biol Reprod 36 405-418.

Hansen PJ, Segerson EC & Bazer FW 1987b Characterization of immunosuppressive

substances in the basic protein fraction of uterine secretions of pregnant ewes. Biol Reprod

36 393-403.

Page 145: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

145

Harper JW, Adami GR, Wei N, Keyomarsi K & Elledge SJ 1993 The p21 Cdk-interacting

protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases. Cell 75 805-816.

Hida K, Wada J, Eguchi J, Zhang H, Baba M, Seida A, Hashimoto I, Okada T, Yasuhara

A, Nakatsuka A, Shikata K, Hourai S, Futami J, Watanabe E, Matsuki Y, Hiramatsu

R, Akagi S, Makino H & Kanwar YS 2005 Visceral adipose tissue-derived serine

protease inhibitor: a unique insulin-sensitizing adipocytokine in obesity. Proc Natl Acad

Sci USA 102 10610-10615.

Hill RE & Hastie ND 1987 Accelerated evolution in the reactive centre regions of serine

protease inhibitors. Nature 326 96-99.

Hoffman LH & Wooding FB 1993 Giant and binucleate trophoblast cells of mammals. J Exp

Zool. 266 559-577.

Horne MC, Donaldson KL, Goolsby GL, Tran D, Mulheisen M, Hell WJ & Wahl AF 1997

Cyclin G2 is up-regulated during growth inhibition and B cell antigen receptor-mediated

cell cycle arrest. J Biol Chem 272 12650-12661.

Hungtinton JA 2006 Shape-shifting serpins-advantages of a mobile mechanism. Trends

Biochem Sci 31 427-435.

Im H, Woo MS, Hwang KY & Yu MH 2002 Interactions causing the kinetic trap in serpin

protein folding. J Biol Chem 48 46347-46354.

Inagi R, Nangaku M, Useda N, Shimizu A, Onogi H, Izuhara Y, Nakazato K, Ueda Y, Oishi

H, Takahashi S, Yamamoto M, Susuki D, Kurokawa K, van Ypersele de Strihou C &

Miyata T 2005 Novel serpinopathy in rat kidney and pancreas induced by overexpression

of Megsin. J Am Soc Nephrol 16 1339-1349.

Ing NH & Roberts RM 1989 The major progesterone-induced proteins secreted into the sheep

uterus are members of the serpin superfamily of protease inhibitors. J Biol Chem 264

3372-3379.

Ing NH, Francis H, McDonnell JJ, Amann JF & Roberts RM 1989 Progesterone induction of

the uterine milk proteins: major secretory proteins of sheep endometrium. Biol Reprod 41

643-654.

Irving JA, Pike RN, Lesk AM & Whisstock JC 2000 Phylogeny of the serpin superfamily:

Implications of patterns of amino acid conservation for structure and function. Genome

Res 10 1845-1864.

Irving JA, Shushanov SS, Pike RN, Popova EY, Bromme D, Coetzer THT, Bottomley SP,

Boulynko IA, Grigoryev SA & Whisstock JC 2002a Inhibitory activity of a

heterochromatin-associated serpin (MENT) against papain-like cysteine proteinases affects

chromatin structure and blocks cell proliferation. J Biol Chem 277 13192-13201.

Page 146: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

146

Irving JA, Steenbakkers PJM, Lesk AM, Op den Camp HJM, Pike RN & Whisstock JC

2002b Serpins in Prokaryotes Mol Biol Evol 19 1881-1890.

Isaacs WB, Carter BS & Erwing CM 1991 Wild-type p53 suppresses growth oh human

prostate cancer cells containing mutant p53 alleles. Cancer Res 51 4716-4720.

Ivanov D, Emonet C, Foata F, Affolter M, Delley M, Fisseha M, Blum-Sperisen S, Kochhar

S & Arigoni F 2006 A serpin from the gut bacterium Bifidobacterium longum inhibits

eukaryotic elastase-like serine proteases. J Biol Chem 281 17246-17252.

Izuhara K, Ohta S, Kanaji S, Shiraishi H & Arima K 2008 Recent progress in understanding

the diversity of human ov-serpin/clade B serpin family. Cell Mol Life Sci 65 2541-2553.

Janciauskiene S, Larsson S, Larsson P, Virtala R, Jansson L & Stevens T 2004 Inhibition of

lipopolysaccharide-mediated human monocyte activation, in vitro, by α1-antitrypsin.

Biochem Biophys Res Commun 321 592-600.

Jin L, Abrahams JP, Skinner R, Petitou M, Pike RN & Carrell RW 1997 The anticoagulant

activation of antithrombin by heparin. Proc Natl Acad Sci USA 94 14683-14688.

Jirawatnotai S, Moons DS, Stocco CO, Franks R, Hales DB, Gibori G & Kiyokawa K 2003

The cyclin-dependent kinase inhibitors p27kip1

and p21cip1

cooperate to restrict proliferative

life span in differentiating ovarian cells. J Biol Chem 278 17021-17027.

Jones DT, Taylor WR & Thornton JM 1992 The rapid generation of mutation data matrices

from protein sequences. Comput Appl Biosci 8 275-82.

Kaiserman Dion, Whisstock JC & Bird PI 2006 Mechanisms of serpin dysfunction in disease.

Expert Rev Mol Med 8 1-19.

Kaldis P 1999 The cdk-activating kinase (CAK): from yeast to mammals. Cell Mol Life Sci 55

284-296.

Kang S, Barak Y, Lamed R, Bayer EA & Morrison Mark 2006 The functional repertoire of

prokaryote cellulosomes includes the serpin superfamily of serine proteinase inhibitors.

Mol Microbiol 60 1344-1354.

Khatib H, Schutzkus V, Chang YM & Rosa GJ 2006 Pattern of expression of the uterine milk

protein gene and its association with productive life in cattle. J Dairy Sci 90 2427-2433.

Knauer MF, Kridel SJ, Hawley SB & Knauer DJ 1997 The efficient catabolism of thrombin-

protease nexin-1 complexes is a synergistic mechanism that requires both the LDL

receptor-related protein and cell surface heparins. J Biol Chem 272 29039-29045.

Page 147: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

147

Knauer DJ, Majumdar D, Fong PC & Knauer MF 2000 SERPIN regulation of factor XIa. J

Biol Chem 275 37340-37346.

Knight PG & Glister C 2006 TGF-β superfamily members and ovarian follicle development.

Reproduction 132 191-206

Knox K & Baker JC 2008 Genomic evolution of the placenta using co-option and duplication

and divergence. Genome Res 18 695-705.

Kounnas MZ, Church FC, Argraves WS & Strickland DK 1996 Cellular internalization and

degradation of antithrombin III-thrombin, heparin cofactor II-thrombin and α1-antitrypsin-

trypsin complexes is mediated by the low density lipoprotein receptor-related protein. J

Biol Chem 271 6523-6529.

Krem MM & Di Cera E 2003 Conserved Ser residues, the shutter region, and speciation in

serpin evolution. J Biol Chem 278 37810-37814.

Krug U, Ganser A & Koeffler HP 1996 Tumor suppressor genes in normal and malignant

hematopoiesis. Oncogene 13 1103-1109.

Law RHP, Zhang Q, McGowan S, Buckle AM, Silverman GA, Wong W, Rosado CJ,

Langendorf CG, Pike RN, Bird PI & Whisstock JC 2006 An overview of the serpin

superfamily. Genome Biol 7 216.

Le XF, Arachchige-Don AS, Mao W, Horne MC & Bast RC Jr 2007 Roles of human

epidermal growth factor receptor 2, c-jun NH2-terminal kinase, phosphoinositide 3-kinase,

and p70 S6 kinase pathways in regulation of cyclin G2 expression in human breast cancer

cells. Mol Cancer Ther 6 2847-2857.

Leiser R, Krebs C, Ebert B & Dantzer V 1997 Placental vascular corrosion cast studies: a

comparison between ruminants and humans. Microsc Res Tech 38 76-87.

Leslie MV & Hansen PJ 1991 Progesterone-regulated secretion of the serpin-like proteins of

the ovine and bovine uterus. Steroids 56 589-597.

Leslie MV, Hansen PJ & Newton GR 1990 Uterine secretions of the cow contain proteins that

are immunochemically related to the major progesterone-induced proteins of the sheep

uterus. Domest Anim Endocrinol 7 517-526.

Lewis EC, Shapiro L, Bowers OJ & Dinarello CA 2005 α1-antitrypsin monotherapy prolongs

islet allograft survival in mice. Proc Natl Acad Sci USA 102 12153-12158.

Li F, Devi S Bao L, Mao J & Gibori G 2008 Involvment of cyclin D3, CDKN1A (p21), and

BIRC5 (survivin) in interleukin 11 stimulation of decidualization in mice. Biol Reprod 78

127-133.

Page 148: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

148

Li R, Waga S, Hannon GJ, Beach D & Stillman B 1994 Differential effects by the p21 CDK

inhibitor on PCNA-dependent replication and repair. Nature 371 534-537.

Lillis AP, van Duyn LB, Murphy-Ullrich & Strickland DK 2008 LDL-Receptor-related

protein 1: unique tissue specific functions revealed by selective gene knockout studies.

Physiol Rev 88 887–918.

Liu WJ & Hansen PJ 1993 Effect of the progesterone-induced serpin-like proteins of the sheep

endometrium on natural-killer cell activity in sheep and mice. Biol Reprod 49 1008-1014.

Liu WJ & Hansen PJ 1995 Progesterone-induced secretion of dipeptidyl peptidase-IV (cell

differentiation antigen 26) by the uterine endometrium of the ewe and cow that

costimulates lymphocyte proliferation. Endocrinology 136 779-787.

Liu WJ, Peltier MR & Hansen PJ 1999 Binding of ovine uterine serpin to lymphocytes. Am J

Reprod Immunol 41 428-432.

Louis EJ 2007 Making the most of redundancy. Nature 449 673-674.

Lu H, Guo X, Meng X, Liu J, Allen C, Wray J, Nickoloff JA & Shen Z 2005 The BRCA2-

interacting protein BCCIP functions in RAD51and BRCA2 focus formation and

homologous recombinational repair. Mol Cell Biol 25 1949-1957.

Lucas A & McFadden 2004 Secreted immunomodulatory viral proteins as novel

biotherapeutics. J Immunol 173 4765-4774.

Lühr K, Müritz C, Contente A & Dobbelstein M 2003 p21/CDKN1A mediates negative

regulation of transcription by p53. J Biol Chem 278 32507-32516.

Makarova A, Mikhailenko I, Bugge TH, List K, Lawrence DA & Strickland DK 2003 The

low density lipoprotein receptor-related protein modulates protease activity in the brain by

mediating the cellular internalization of both neuroserpin and neuroserpin-tissue-type

plasminogen activator complexes. J Biol Chem 278 50250-50258.

Malathy PV, Imakawa K, Simmen RC & Roberts RM 1990 Molecular cloning of the

uteroferrin-associated protein, a major progesterone-induced serpin secreted by the porcine

uterus, and expression of its mRNA during pregnancy. Mol Endocrinol 4 428-440.

Martinsson-Ahlzén H, Liberal V, Grünenfelder B, Chaves SR, Spruck CH & Reed SI 2008

Cyclin-dependent kinase-associated proteins cks1 and cks2 are essential during early

embryogenesis and for cell cycle progression in somatic cells. Mol Cell Biol 28 5698-

5709.

Maston GA & Ruvolo M 2002 Chorionic gonadotropin has a recent origin within primates and

an evolutionary history of selection. Mol Biol Evol 19 320-335.

Page 149: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

149

Mathialagan N & Hansen TR 1996 Pepsin-inhibitory activity of uterine serpins. Proc Natl

Acad Sci USA 93 13653-13658.

McFarlane JR, Foulds LM, O’Connor AE, Phillips DJ, Jenkin G, Hearn MTW & Kretser

DM 1999 Uterine milk protein, a novel activin-binding protein, is present in ovine allantoic

fluid. Endocrinology 140 4745-4752.

Medcalf RL & Stasinopoulos SJ 2005 The undecided serpin. The ins and outs of plaminogen

activator type 2. FEBS J 272 4858-4867.

Mess A & Carter AM 2007 Evolution of the placenta during the early radiation of placental

mammals. Comp Biochem Physiol A Mol Integr Physiol 148 769-779.

Miyata T, Inagi R, Sugiyama S & Usuda N 2005 Serpinopathy and endoplasmic reticulum

stress. Med Mol Morphol 38 73-78.

Moffatt J, Bazer FW, Hansen PJ, Chun PW & Roberts RM 1987 Purification, secretion and

immunocytochemical localization of the uterine milk proteins, major progesterone-induced

proteins in uterine secretions of the sheep. Biol Reprod 36 419-430.

Moffett A & Loke C 2006 Immunology of placentation in eutherian mammals. Nat Rev

Immunol 6 584–594.

Morgan L, Broughton Pipkin F & Kalsheker N 1996 Angiotensinogen: molecular biology,

biochemistry and physiology. Int J Biochem Cell Biol 28 1211-1222.

Muhl L, Nykjaer A, Wygrecka M, Monard D, Preissner KT & Kanse SM 2007 Inhibition of

PDGF-BB by factor VII-activating protease (FSAP) is neutralized by protease nexin-1, and

the FSAP-inhibitor complexes are internalized via LRP. Biochem J 404 191-196.

Murphy WJ, Pringle TH, Crider TA, Springer MS & Miller W 2007 Using genomic data to

unravel the root of mammal phylogeny. Genome Res 17 413-421.

Nagata K 1998 Expression and function of heat shock protein 47: a collagen-specific molecular

chaperone in the endoplasmic reticulum. Matrix Biol 16 379-386.

Newton GR, Hansen PJ, Bazer FW, Leslie MV, Stephenson DC & Low BG 1989 Presence

of major progesterone-induced proteins of the sheep endometrium in fetal fluids. Biol

Reprod 40 417-424.

Nishihara H, Hasewaga M & Okada N 2006 Pegasoferae, an unexpected mammalian clade

revealed by tracking ancient retroposon insertions. Proc Natl Acad Sci USA 103 9929-9934.

Noel S, Herman A, Johnson GA, Gray CA, Stewart MD, Bazer FW, Gertler A & Spencer

TE 2003 Ovine placental lactogen specifically binds to endometrial glands to the ovine

uterus. Biol Reprod 68 772-780.

Page 150: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

150

Nuttleman PR & Roberts RM 1990 Transfer of iron from uteroferrin (purple acid phosphatase)

to transferring related to acid phosphatase activity. J Biol Chem 265 12192-12199.

Oliveira L & Hansen PJ 2008 Deviations in populations of peripheral blood mononuclear cells

and endometrial macrophages in the cow during pregnancy. Reproduction 136 481-490.

Olson D, Pöllänen J, Høyer-Hansen G, Rønne E, Sakaguchi K, Wun TC, Appella E, Danø

K & Blasi F 1992 Internalization of the urokinase-plasminogen activator inhibitor type-1

complex is mediated by the urokinase receptor. J Biol Chem 267 9129-9133.

Padua MB & Hansen PJ 2008 Regulation of DNA synthesis and the cell cycle in human

prostate cancer cells and lymphocytes by ovine uterine serpin. BMC Cell Biol 9 5.

Padua MB, Tekin S, Spencer TE & Hansen PJ 2005 Actions of progesterone on uterine

immunosuppression and endometrial gland development in the uterine gland knockout

(UGKO) ewe. Mol Reprod Dev 71 347-357.

Peltier MR, Grant TR & Hansen PJ 2000a Distinct physical and structural properties of the

ovine uterine serpin. Biochim Biophys Acta 1479 37-51.

Peltier MR, Liu WJ & Hansen PJ 2000b Regulation of lymphocyte proliferation by uterine

serpin: interleukin-2 mRNA production, CD25 expression and responsiveness to

interleukin-2. Proc Soc Exp Biol Med 223 75-81.

Peltier MR, Raley LC, Liberles DA, Benner SA & Hansen PJ 2000c Evolutionary history of

the uterine serpins. J Exp Zool 288 165-174.

Pemberton PA, Stein PE, Pepys MB, Potter JM & Carrell RW 1988 Hormone binding

globulins undergo serpin conformational change in inflammation. Nature 336 257-258.

Poller W, Willnow TE, Hilpert J & Herz J 1995 Differential recognition of α1-antitrypsin-

elastase and α1-antichymotrypsin-cathepsin G complexes by the low density lipoprotein

receptor-related protein. J Biol Chem 270 2841-2845.

Potempa J, Korzus E & Travis J 1994 The serpin superfamily of proteinase inhibitors:

structure, function and regulation. J Biol Chem 269 15957-15960.

Ragg H, Lokot T, Kamp PB, Atchley WR & Dress A 2001 Vertebrate serpins: construction of

a conflict-free phylogeny by combining exon-intron and diagnostic site analyses. Mol Biol

Evol 18 577-584.

Renegar RH, Bazer FW & Roberts RM 1982 Placental transport and distribution of uteroferrin

in the fetal pig. Biol Reprod 27 1247-1260.

Page 151: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

151

Riahi Y, Siman-Tov R & Ankri S 2004 Molecular cloning, expresión and characterization of a

serine proteinase inhibitor from Entamoeba histolytica. Mol Biochem Parasitol 133 153-

162.

Roberts RM & Bazer FW 1988 The functions of uterine secretions. J Reprod Fert 82 875-892.

Roberts RM, Ealy AD, Alexenko AP, Han CS & Ezashi T 1999 Trophoblast interferons.

Placenta 20 256-264.

Roberts TH, Hejgaard J, Saunders NFW, Cavicchioli R & Curmi PMG 2004 Serpins in

unicellular Eukarya, Archaea, and Bacteria: sequence analysis and evolution. J Mol Evol

59 437-447.

Saitou N & Nei M 1987 The neighbor-joining method: a new method for reconstructing

phylogenetic trees. Mol Biol Evol 4 406-425

Sancar A, Lindsey-Boltz LA, Unsal-Kaҫ maz K & Linn S 2004 Molecular mechanisms of

mammalian DNA repair and the DNA damage checkpoints. Annu Rev Biochem 73 39-85.

Sartori AA, Lukas C, Coates J, Mistrik M, Fu S, Bartek J, Baer R, Lukas J & Jackson SP

2007 Human CtIP promotes DNA end resection. Nature 450 509-514.

Sauk JJ, Nikitakis N & Siavash H 2005 Hsp47 a novel collagen binding serpin chaperone,

autoantigen and therapeutic target. Front Biosci 10 107-118.

Segerson EC, Moffatt RJ, Bazer FW & Roberts MR 1984 Suppression of

phytohemagglutinin-stimulated lymphocyte blastogenesis by ovine uterine milk protein.

Biol Reprod 30 1175-1186.

Schmidt HA, Strimmer K, Vingron M & von Haeseler A 2002 TREE-PUZZLE: maximum

likelihood phylogenetic analysis using quartets and parallel computing. Bioinformatics 18

502-504.

Schvartz I, Seger D & Shaltiel S 1999 Molecules in focus Vitronectin. Int J Biochem Cell Biol

31 539-544.

Sheng S 2006 A role of novel serpin maspin in tumor progression: the divergence revealed

through efforts to converge. J Cell Physiol 209 631-635.

Silverman GA, Bird PI, Carrell RW, Church FC, Coughlin PB, Gettins PGW, Irving JA,

Lomas DA, Luke CJ, Moyer RW, Pemberton PA, Remold-O’Donnell E, Salvesen GS,

Travis J & Whisstock JC 2001 The serpins are an expanding superfamily of structurally

similar but functional diverse proteins. J Biol Chem 276 33293-33296.

Silverman GA, Whisstock JC, Askew DJ, Pak SC, Luke CJ, Cataltepe S, Irving JA & Bird

PI 2004 Human clade B serpins (ov-serpins) belong to a cohort of evolutionarily dispersed

Page 152: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

152

intracellular proteinase inhibitor clades that protect cells from promiscuous proteolysis.

Cell Mol Life Sci 61 301-325.

Skeldal S, Larsen JV, Pedersen KE, Petersen HH, Egelund R, Christensen A, Jensen JK,

Gliemann J & Andreasen PA 2006 Binding areas of urokinase-type plasminogen

activator-plasminogen activator inhibitor-1 complex for endocytosis receptors of the low-

density lipoprotein receptor family, determined by site-directed mutagenesis. FEBS J 273

5143-5159.

Skopets B & Hansen PJ 1993 Identification of the predominant proteins in uterine fluids of

unilaterally pregnant ewes that inhibit lymphocyte proliferation. Biol Reprod 49 997-1007.

Skopets B, Liu WJ & Hansen PJ 1995 Effects of endometrial serpin-like proteins on immune

responses in sheep. Am J Reprod Immunol 33 86-93.

Song S, Goudy K, Campbell-Thompson M, Wasserfall C, Scott-Jorgensen M, Wang J,

Tang Q, Crawford JM, Ellis TM, Atkinson MA & Flotte TR 2004 Recombinant adeno-

associated virus-mediated alpha-1 antitrypsin gene therapy prevents type I diabetes in NOD

mice. Gene Ther 11 181-186.

Spencer TE, Gray A, Johnson GA, Taylor KM, Gertler A, Gootwine E, Ott TL & Bazer

FW 1999 Effects of recombinant ovine interferon tau, placental lactogen, and growth

hormone on the ovine uterus. Biol Reprod 61 1409-1418.

Stanley P, Serpell LC & Stein PE 2006 Polymerization of human angiotensinogen: insights

into its structural mechanism and functional significance. Biochem J 400 169-178.

Steenbakkers PJM, Irving JA, Harhangi HR, Swinkels WJC, Akhmanova A, Dijkerman R,

Jetten MSM, Van Der Drift C, Whisstock JC & Op den Camp HJM 2008 A serpin in

the cellulosome of the anaerobic fungus Piromyces sp. strain E2. Mycol Res 112 999-1006.

Stephenson DC, Leslie MV, Low BG, Newton GR, Hansen PJ & Bazer FW 1989a Secretion

of the major progesterone-induced proteins of the sheep uterus by caruncular and

intercaruncular endometrium of the pregnant ewe from days 20-140 of gestation. Domest

Anim Endocrinol 6 349-362.

Stephenson DC, Hansen PJ, Newton GR, Bazer FW & Low BG 1989b Inhibition of

lymphocyte proliferation by uterine fluid from the pregnant ewe. Biol Reprod 41 1063-

1075.

Stern A, Doron-Faigenboim A, Erez E, Martz E, Bacharach E & Pupko T 2007 Selecton

2007: advanced models for detecting positive and purifying selection using a Bayesian

inference approach. Nucleic Acids Res 35 W506-W511.

Page 153: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

153

Stewart MD, Johnson GA, Gray CA, Burghardt RC, Schuler LA, Joyce MM, Bazer FW &

Spencer TE 2000 Prolactin receptor and uterine milk protein expression in the ovine

endometrium during the estrous cycle and pregnancy. Biol Reprod 62 1779-1789.

Storm D, Herz J, Trinder P & Loos M 1997 C1 inhibitor-C1s complexes are internalize and

degraded by the low density lipoprotein receptor-related protein. J Biol Chem 272 31043-

31050.

Strimmer K & von Haeseler, A 1996 Quartet puzzling: A quartet maximum likelihood method

for reconstructing tree topologies. Mol Biol Evol 13 964-969.

Tamura K, Dudley J, Nei M & Kumar S 2007 MEGA4: Molecular Evolutionary Genetics

Analysis (MEGA) software version 4.0. Mol Biol Evol 24 1596-1599.

Tan J, Raja S, Davis MK, Tawfik O, Dey SK & Das SK 2002 Evidence for coordinated

interaction of cyclin D3 with p21 and cdk6 in directing the development of uterine stromal

cells decidualization and polyploidy during implantation. Mech Dev 111 99-113.

Tekin Ş & Hansen PJ 2002 Natural-killer like cells in the sheep: Functional characterization

and regulation by pregnancy-associated proteins. Exp Biol Med (Maywood) 227 803-811.

Tekin Ş, Padua MB, Brad AM & Hansen PJ 2005b Antiproliferative actions of ovine uterine

serpin. Am J Reprod Immunol 53 136-143.

Tekin Ş, Padua MB, Brad AM, Rhodes ML & Hansen PJ 2006 Expression and properties of

recombinant ovine uterine serpin. Exp Biol Med (Maywood) 231 1313-1322.

Tekin Ş, Padua MB, Newton GR & Hansen PJ 2005a Identification and cloning of caprine

uterine serpin. Mol Reprod Dev 70 262-270.

Teng SC, Wu KJ, Tseng SF, Wong CW & Kao L 2003 Importin KPNA2, NBS1, DNA repair

and tumorigenesis. J Mol Hist 37 293-299.

Travis J & Salvesen GS 1983 Human plasma proteinase inhibitors. Annu Rev Biochem 52 655-

709.

Tombran-Tink J & Barnstable CJ 2003 PEDF: a multifaceted neurotrophic factor. Nat Rev

Neurosci 4 628-636.

Vallet JL 1995 Uteroferrin induces lipid peroxidation in endometrial and conceptus microsomal

mambranes and is inhibited by apotransferrin, retinol binding protein, and the uteroferrin-

associated proteins. Biol Reprod 53 1436-1445.

Van Gent D, Sharp P, Morgan K & Kalsheker N 2003 Serpins: structure, function and

molecular evolution. Int J Biochem Cell Biol 35 1536-1547.

Page 154: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

154

Vogel P 2005 The current molecular phylogeny of eutherian mammals challenges previous

interpretations of placental evolution. Placenta 26 591-596.

Waga S. Hannon GJ, Beach D & Stillman B 1994 The p21 inhibitor of cyclin-dependent

kinases controls DNA replication by interaction with PCNA. Nature 369 574-578.

Whisstock JC & Bottomley SP 2006 Molecular gymnastics: serpin structure folding and

misfolding. Current Opinion Struct Biol 16 761-768.

Wind T, Hansen M, Jensen JK & Andreasen PA 2002 The molecular basis for anti-

proteolytic and non-proteolytic functions of plasminogen activator inhibitor type-1: roles of

the reactive center loop, the shutter region, the flexible joint region and the small serpin

fragment. Biol Chem 2002 383 21-36.

Wong WSW, Yang Z, Goldman N & Nielsen R 2004 Accuracy and power of statistical

methods for detecting adaptive evolution in protein coding sequences and for identifying

positively selected sites. Genetics 168 1041-1051.

Xie S, Low BG, Nagel RJ, Kramer KK, Anthony RV, Zoli AP, Beckers JF & Roberts RM 1991 Identification of the major pregnancy-specific antigens of cattle and sheep as inactive

members of the aspartic proteinase family. Proc Natl Acad Sci USA 88 10247-10251.

Xiong J, Hu L, Li Y, Dou L, Cai P, Tang Z & Wang L 2008 Effect on survivin regulation of

transcription level by p21waf1

overexpression in HepG2 hepatocellular carcinoma cells. J

Huazhong Univ Sci Technol Med Sci 28 308-313.

Yang Z 2007 PAML 4: Phylogenetic analysis by maximum likelihood. Mol Biol Evol 24 1586-

1591.

Yang Z & Nielsen R 2002 Codon substitution models for detecting molecular adaptation at

individual sites along specific lineages. Mol Biol Evol 19 908-917.

Yang Z, Nielsen R, Goldman N & Pedersen MK 2000 Codon-substitution models for

heterogeneous selection pressure at amino acids sites. Genetics 155 431-449.

Yang Z, Wong WSW & Nielsen R 2005 Bayes empirical Bayes inference of amino acid sites

under positive selection. Mol Biol Evol 22 2472-2479.

Yao MWM, Lim H, Schust DJ, Choe SE, Farago A, Ding Y, Michaud S, Church GM &

Maas RL 2003 Gene expression profiling reveals progesterone mediated cell cycle and

immunoregulatory roles of Hoxa-10 in the preimplantantion uterus. Mol Endocrinol 17

610-627

Yoo BC, Aoki K, Xiang Y, Campbell LR, Hull RJ, Xoconostle-Cázares B, Monzer J, Lee

JY, Ullman DE & Lucas WJ 2000 Characterization of cucurbita maxima phloem serpin-1

(CmPS-1). A developmentally regulated elastase inhibitor. J Biol Chem 275 35122-35128.

Page 155: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

155

Yue L, Daikoku T, Hou X, Li M, Wang H, Nojima H, Dey SK & Das SK 2005 Cyclin G1

and cyclin G2 are expressed in the periimplantation mouse uterus in a cell-specific and

progesterone-dependent manner: evidence for aberrant regulation with hoxa-10 deficiency.

Endocrinology 146 2424-2433.

Zang X & Maizels RM 2001 Serine proteinase inhibitors from nematodes and the arm race

between host and pathogens. Trends Biochem Sci 26 191-197.

Zhang B, Lu Y, Campbell-Thompson M, Specer T, Wasserfall C, Atkinson M & Song S 2007 α1-antitrypsin protects β-cells from apoptosis. Diabetes 56 1316-1323.

Zhang M 2000 Maspin is an angiogenesis inhibitor. Nat Med 6 196-199.

Zhou A, Huntington JA, Pannu NS, Carrell RW & Read RJ 2003 How vitronectin binds

PAI-1 to modulate fibrinolysis and cell migration. Nat Struct Biol 10 541-544.

Page 156: © Maria B. Padua - University of Floridaufdcimages.uflib.ufl.edu/UF/E0/02/44/00/00001/padua_m.pdfMARIA B. PADUA A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF

156

BIOGRAPHICAL SKETCH

Maria Beatriz Padua was born in 1968 in Caracas, Venezuela. She graduated from El

Carmelo High School in the same city in 1985 and enrolled the next year in the School of

Agronomy at the University Centroccidental Lisandro Alvarado in Barquisimeto, Lara State,

Venezuela where she received her Bachelor of Science degree in Agricultural Engineering in

1994. In 1995, she worked as sales representative for the veterinary health supply firm Grupo

Catalina C.A. in Caracas, Venezuela. From 1996 to 2000, she was a pharmacist assistant in

Celbefar C.A. Farmacia El Roble in the same city. She participated in an English language

program at the University of Florida in 2001 and she enrolled in the Animal Molecular and

Cellular Biology Graduate Program at the University of Florida under the supervision of Dr.

Peter J. Hansen in April, 2002. She received her Master of Science degree in 2004 and her thesis

was elected as the best thesis for the Department of Animal Sciences. She is currently a Doctor

of Philosophy candidate. Upon completion of her degree, she will pursue a postdoctoral

position.


Recommended