184
COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS by Victoria (Vicky) Phương Kim Hoàng Nguyn A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Graduate Department of Medical Biophysics University of Toronto © Copyright by Victoria PKH Nguyen (2010)

LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

COMPARATIVE APPROACHES TO CHARACTERIZATION OF

LYMPHATIC ENDOTHELIAL CELLS AS

PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS

by

Victoria (Vicky) Phương Kim Hoàng Nguyễn

A thesis submitted in conformity with the requirements

for the degree of Doctor of Philosophy

Graduate Department of Medical Biophysics

University of Toronto

© Copyright by Victoria PKH Nguyen (2010)

Page 2: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

ii

COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS

AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS

Victoria P. K. H. Nguyen

Doctor of Philosophy

Department of Medical Biophysics, University of Toronto

2010

ABSTRACT

The lymphatic system complements the blood circulatory system in absorption and

transport of nutrients, and in the maintenance of homeostasis. Historically, the angiogenesis

field has advanced faster and farther than the field of lymphangiogenesis. The discovery of

lymphatic markers and the emerging evidence implicating the lymphatic system as a central

player in a variety of pathological conditions has attracted research interest and driven the field

forward. Research efforts have produced the observation that regulators of the blood

endothelium are frequently members of the same protein families of regulators of the lymphatic

endothelium. More importantly, these regulators do not act discretely, restricting their

regulatory activities to one endothelial cell (EC) type. Two examples of regulators that behave

in this manner are the VEGF and the Angiopoietin families of proteins, which have cell-type-

dependent effects on EC processes such as migration, proliferation and survival. The study of

these regulators therefore requires an in vitro EC system capable of accommodating the

simultaneous characterization of the signaling pathways downstream of these shared molecular

regulators in venous, arterial and lymphatic endotheliums. To build such an in vitro system, I

isolated and validated lymphatic, venous, and arterial ECs derived from vessels of bovine

mesentery. The proteomes of the three cell types were comparatively studied using two-

Page 3: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

iii

dimensional polyacrylamide gel electrophoresis followed by mass spectrometric identification.

The three cell types were used in a subtractive immunization scheme for the production of a

monoclonal antibody selectively reactive to a potentially novel surface protein marker of

lymphatic ECs. The studies recorded herein all share the common goal of identifying and

characterizing unique molecular signatures that distinguish lymphatic ECs from blood ECs, and

that may underline the cellular biology of the lymphatic endothelium as distinct from the blood

endothelium.

Page 4: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

iv

ACKNOWLEDGEMENTS

I would first like to thank my mentor Dr. Daniel J. Dumont for his unwearied support,

and for his unmatched professional guidance so that I may become the best physician and

scientist I can be. Many sincere thanks to my committee members Dr. Mike Moran, for

expecting the best from me, and Dr. Dwayne Barber for always believing in me. Dr. Edwin E.

Daniel helped me to discover research as a worthy life-long pursuit. Dr. Peter F. Whyte was a

patient and kind teacher who inspired me to strive for excellence in teaching. Dr. Eric Yang

was my tireless tutor in the intricacies of proteomics. Many special thanks to Stephen, Paul,

Harold, Jamie, Maribelle, and Sue for their supportive friendship. Lani and Farhah deserve

praise for their unconditional affection. A very large thank you goes out to my very large

extended family for always being more than uncles, aunts, and cousins, but the very best of

friends. I am forever indebted to my uncle Hoàng Trọng Phú who brought my family to this

wonderful country and opened up a whole world of possibility for my future. I know that my

beloved brothers Andy and Tim, and my dear father, Nguyễn Văn Kim will always be on my

side. To Feroz Sarkari, I want to give thanks for changing my life, for always knowing what

really matters in life, and for being my compass and my anchor in rough seas. I will always

remember with the deepest gratitude my late grandfather Hoàng Trọng Phụ for his Vietnamese

nick name for me, Như ý, reminding me that I am already as he wished and that I am already

‗good enough‘, for his grand vision of what life should be for his children and grandchildren,

and for the ultimate sacrifice he paid so that they would have a shot at the life he never had.

Most importantly, I want to thank my mother, Hoàng Thị Mỹ Phương, for all her motherly

deeds, great and small, for her true unchanging, everlasting love, for her wisdom and goodness,

and determination to fulfill her father‘s dreams so that his sacrifice may not be in vain.

Page 5: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

v

TABLE OF CONTENTS

ABSTRACT .................................................................................................................................. II

ACKNOWLEDGEMENTS ........................................................................................................IV

TABLE OF CONTENTS ............................................................................................................. V

TABLE OF ILLUSTRATIONS .................................................................................................IX

ABBREVIATIONS ...................................................................................................................... X

CHAPTER 1: BACKGROUND ....................................................................................................... 1

1.1. Rationale and Objective ..................................................................................................... 2

1.2. Introduction to Lymphatic Anatomy ................................................................................ 5

1.2.1. Lymphatic Anatomy and Function ................................................................................ 5

1.2.2. Lymphatic Endothelium Function in Immunity ............................................................ 7

1.2.3. Lymphatic Endothelium Function in Homeostasis ........................................................ 8

1.2.4. Lymphatic Endothelium Dysfunction in Lymphoedema ............................................. 10

1.2.5. Lymphatic Endothelium Dysfunction in other Human Pathologies ............................ 13

1.2.6. Protein Markers of the Lymphatic Endothelium.......................................................... 15

1.3. Data Chapter Organization ............................................................................................. 19

1.4. Attributions ....................................................................................................................... 20

CHAPTER 2: LYMPHATIC, VENOUS AND ARTERIAL ENDOTHELIAL CELLS ISOLATED FROM

BOVINE MESENTERIC VESSELS RETAIN THEIR DEFINING CHARACTERISTICS IN CULTURE

AND ARE DIFFERENTIALLY RESPONSIVE TO ANGIOPOIETIN-1 AND -2 .....................................21

2.1. Introduction ...................................................................................................................... 22

2.2. Methods ............................................................................................................................. 24

Page 6: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

vi

2.2.1. BmEC isolation and culture ......................................................................................... 24

2.2.2. Eliminating contaminating cells .................................................................................. 25

2.2.3. Detection of transcripts by RT-PCR ............................................................................ 25

2.2.4. Cell imaging ................................................................................................................. 26

2.2.5. Stimulation of bovine ECs ........................................................................................... 26

2.2.6. Cell Lysis and Tie-2 Immunoprecipitation .................................................................. 27

2.2.7. Immunoblotting ............................................................................................................ 27

2.2.8. 3H-Thymidine uptake assay ......................................................................................... 27

2.2.9. Cell count assay ........................................................................................................... 28

2.2.10. Modified Boyden Chamber Migration Assay ............................................................ 28

2.2.11. Cell death ELISA ....................................................................................................... 29

2.3. Results ................................................................................................................................ 29

2.3.1. BmLEC isolation and culture. ...................................................................................... 29

2.3.2. Comparing bmLECs to bmVECs, and to bmAECs. .................................................... 33

2.3.3. Activation of Tie-2 in bmECs by Ang-2 ...................................................................... 39

2.3.4. BmLEC proliferation is enhanced by Ang-1 and Ang-2 ............................................. 45

2.3.5. Ang-1 promotes bmEC migration ................................................................................ 46

2.3.6. Ang-2 protects bmLEC from cell death ....................................................................... 46

2.4. Discussion .......................................................................................................................... 56

CHAPTER 3: DIFFERENTIAL PROTEOMIC ANALYSIS OF LYMPHATIC, VENOUS, AND

ARTERIAL ENDOTHELIAL CELLS EXTRACTED FROM BOVINE MESENTERIC VESSELS ............. 59

3.1. Introduction ...................................................................................................................... 60

3.2. Methods ............................................................................................................................. 63

Page 7: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

vii

3.2.1. Cell isolation and culture ............................................................................................. 63

3.2.2. 2D-PAGE ..................................................................................................................... 63

3.2.3. Mass Spectrometric Analysis of Protein Spot Intensities ............................................ 64

3.2.4. Validation of Protein Level Differences by Immunoblotting ...................................... 64

3.3. Results ................................................................................................................................ 65

3.3.1. Quantitative Analysis of 2D-PAGE-Resolved bmEC Proteins ................................... 65

3.3.2. Mass Spectrometric Analysis of 2D-PAGE-Resolved bmEC Proteins ....................... 71

3.3.3. Validation of Identified Proteins by Immunoblotting .................................................. 83

3.3.4. Comparing Protein Levels of Different Family Members in the same ECs ................ 84

3.3.5. Comparing Protein Levels in ECs of Dissimilar Anatomical Origin .......................... 84

3.4. Discussion .......................................................................................................................... 92

CHAPTER 4: GENERATION OF A MONOCLONAL ANTIBODY SELECTIVELY REACTIVE TO

BOVINE MESENTERIC LYMPHATIC ENDOTHELIAL CELLS BUT NOT TO BOVINE MESENTERIC

VENOUS OR ARTERIAL ENDOTHELIAL CELLS ............................................................................. 98

4.1. Introduction ...................................................................................................................... 99

4.2. Methods ........................................................................................................................... 100

4.2.1 Endothelial Cell Culture.............................................................................................. 100

4.2.2. Subtractive Immunization and Hybridoma Production ............................................. 101

4.2.3. Antibody Screening and Isotyping ............................................................................. 101

4.2.4. Immunoblot Analysis ................................................................................................. 102

4.2.5. Immunohistochemistry .............................................................................................. 103

4.2.6. ULSP180 enrichment by sucrose-gradient-based cell fractionation .......................... 103

Page 8: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

viii

4.2.7. Immunoprecipitation and Identification of ULSP180 by LC-MS/MS ...................... 104

4.3. Results .............................................................................................................................. 106

4.3.1 Hybridoma Production and Screening ........................................................................ 106

4.3.2. MAb isotyping ........................................................................................................... 112

4.3.3. MAb is Selective for bmLECs ................................................................................... 112

4.3.4. 8C8 mAb recognizes a protein of MW approximately 180kDa ................................ 117

4.3. 5. Mass Spectrometric Analysis of ULSP180 ............................................................... 123

4.4. Discussion ........................................................................................................................ 131

CHAPTER 5: CONCLUSIONS AND FUTURE DIRECTIONS ........................................................ 136

REFERENCES .......................................................................................................................... 142

Page 9: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

ix

TABLE OF ILLUSTRATIONS

Illustration

Page

Figure 2.1. Isolation of bmLECs. 31-32

Figure 2.2. Similarities and differences between bmLECs, bmVECs and

bmAECs. 35-36

Figure 2.3. BmLECs, bmVECs and bmAECs express cell-lineage-specific

markers. 37-38

Figure 2.4. Alignment of bovine, murine, and human Tie-2 amino acid

sequences. 41-42

Figure 2.5. Stimulation of Tie-2 phosphorylation in bmECs by Ang-1 and Ang-2. 43-44

Figure 2.6. Proliferation of Ang-1 and Ang-2 stimulated bmECs. 48-49

Figure 2.7. Migration of Ang-1 and Ang-2 stimulated bmECs. 50-51

Figure 2.8. Survival of Ang-1 and Ang-2 stimulated bmECs. 52-53

Table 2.1. Summary of results. 54-55

Figure 3.1. 2D-PAGE separation of bmEC proteins. 67-68

Figure 3.2. Quantitative Analysis of 2D-PAGE-Resolved bmEC Proteins. 69-70

Table 3.1.A. Summary of proteins identified. 73-76

Table 3.1.B. Summary of biological information on protein identified. 77-80

Figure 3.3. Biological Functions of Proteins Identified in Table 4.1. 81-82

Figure 3.4. Validation of HSPA1B, HSPB1, and UBE2D3 protein levels in the

three cell types by IB. 86-87

Figure 3.5. Determination of HSPB2 protein levels in the three cell types by IB 88-89

Figure 3.6. Comparing ANAX2 Protein Levels in ECs of Dissimilar Anatomical

Origin. 90-91

Figure 4.1. Screening Assay Validation. 108-109

Figure 4.2. Serum and Hybridoma Screening. 110-111

Figure 4.3. MAb Isotyping. 113-114

Figure 4.4. Antibody Validation. 115-116

Figure 4.5. ULSP180 Characterization. 119-120

Figure 4.6. Schema of Mass Spectrometric Analysis of ULSP180. 121-122

Figure 4.7. Enrichment and Immunoprecipitation of ULSP180. 125-126

Figure 4.8. Mass Spectrometric Analysis of ULSP180. 127-128

Figure 4.9. Mass Spectrometric Analysis of ULSP180 (continued). 129-130

Page 10: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

x

ABBREVIATIONS

Ab – Antibody

ACh - Acetylcholine

ANAX – Annexin

Ang – Angiopoietin

Anti-His – Anti-Polyhistidine Monoclonal Antibody

baAECs – Bovine Aortic Arterial Endothelial Cells

bmAECs – Bovine Mesenteric Arterial Endothelial Cells

bmECs – Bovine Mesenteric Endothelial Cells

bmBECs – Bovine Mesenteric Blood Endothelial Cells

bmLECs – Bovine Mesenteric Lymphatic Endothelial Cells

bmVECs – Bovine Mesenteric Venous Endothelial Cells

CCL21 – Cysteine-Cysteine Motif Ligand 21

CCR7 – Cysteine-Cysteine Chemokine Receptor 7

CD86 – Cluster of Differentiation 86

cDNA – Complementary Deoxyribonucleic Acid

CI – Confidence Interval

COMP-Ang-1 – Cartilage Oligomeric Matrix Protein-Angiopoietin-1

CXCL1 – Cysteine-X-Cysteine motif ligand 1

CID – Collision Induced Dissociation

CO2 – Carbon Dioxide

DAB – 3,3‘-Diaminobenzindine

DAPI – 4',6-diamidino-2-phenylindole

Page 11: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

xi

ºC – Degrees Celcius

DMEM – Dulbecco‘s Modified Eagle‘s Medium

2D-PAGE – Two Dimensional Polyacrylamide Gel Electrophoresis

ECs – Endothelial Cells

EDTA – Ethylene-diamine-tetra-acetate

EGF – Epidermal Growth Factor

ELISA – Enzyme Linked Immunosorbant Assay

EphB4 – Ephrin B4

Erk1/2 – Extracellular Signal-Regulated Kinases 1/2

ETD – Electron Transfer Dissociation

EtOH - Ethanol

FBS – Fetal Bovine Serum

FOXC2 – Forkhead Box Protein C2

g – Gram

> – Greater than

GM1 – monosialotetrahexosylganglioside

3H - tritium

HAT – Hypoxanthine-Aminopterin-Thymidine

HPLC – High Performance Liquid Chromatography

HRP – Horse Radish Peroxidase

HSP – Heat Shock Protein

HUVEC – Human Umbilical Venous Endothelial Cells

IB – Immunoblotting

Page 12: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

xii

ICAM – Intercellular Adhesion Molecule 1

IEF – Isoelectric Focusing

IPG – Immobilized pH Gradient

IgG - Immunoglobulin

IHC – Immunohistochemistry

IMDM – Iscove‘s Modified Dulbecco‘s Medium

i.p. – intra-peritoneal (injection)

IP - Immunoprecipitation

JNK/SAPK – c-Jun N-terminal Kinase/Stress-Activated Protein Kinase

kDa – Kilodalton

LC – Liquid Chromatography

LC-MS/MS – Liquid Chromatography Tandem Mass Spectrometry

LEC – Lymphatic Endothelial Cells

< – Less than

LMCs – Lymphatic Muscle Cells

LYVE-1 – Lymphatic Vessel Endothelial Hyaluronic Receptor 1

mAb – Monoclonal Antibody

MBS – MES Buffered Saline

MeOH – Methanol

MES – 2-(N-morpholino) ethanesulfonic acid

µCi – Microcurie

µg – Microgram

µm – Micrometer

Page 13: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

xiii

µM – Micromoles

mg – Milligram

mL – Millilitre

MODS – Multiple Organ Dysfunction Syndrome

MS – Mass Spectrometry

MS/MS – Tandem Mass Spectrometry

MW – Molecular Weight

NB – Nota bene

NO – Nitric Oxide

nm - Nanometres

NP-40 – Nonyl phenoxylpolyethoxylethanol.

O2 – Oxygen

PBS – Phosphate Buffered Saline

PECAM-1 – Platelet/Endothelial Cell Adhesion Molecule-1

% – Percent

PCR – Polymerase Chain Reaction

PFA – Paraformaldehyde

PKB – Protein Kinase B

Prox-1 – Prospero Homeobox Protein 1

PRDX – Peroxiredoxin

PVDF – Polyvinylidene Fluoride

RIPA – Radio-immunoprecipitation Assay

RNA – Ribonucleic Acid

Page 14: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

xiv

ROS – Reactive Oxygen Species

RT-PCR – Reverse Transcriptase Polymerase Chain Reaction

SDS – Sodium Dodecyl Sulphate

SI – Subtractive Immunization

SIRS – Acute Systemic Inflammatory State

SPI – Scored Peak Intensity

Tie-2 – Tyrosine Kinase with Immunoglobulin-Like and EGF-like Domains 2

TLR – Toll-like Receptors

TNFα or β – Tumour Necrosis Factor alpha or beta

TMB – 3, 3‘, 5, 5‘ -Tetramethylbenzidine

TritonX-100 – t-octylphenoxypolyethoxyethanol,

UBE – Ubiquitin Conjugating Enzyme

VCAM – Vascular Cell Adhesion Molecule 1

VEGF – Vascular Endothelial Growth Factor

VEGFR-3/Flt-4 – Vascular Endothelial Growth Factor Receptor/Fms-Like Tyrosine Kinase 4

Page 15: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

1

CHAPTER 1

BACKGROUND

Page 16: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

2

1.1. Rationale and Objective

The body‘s network of blind-ended lymphatic capillaries collects interstitial protein-rich

fluid containing clotting factors, lipids, and lymphocytes extravasated from blood vessels. The

milky white lymph then drains into collecting lymphatic vessels, which actively pump the fluid

into larger transporting lymphatic vessels. In pathology, malformed lymphatic vessels—often

resulting from aberrant lymphangiogenesis during development—can lead to lymphangiectasia,

lymphangioma, lymphatic dysplasia, and associated lymphoedema. In normal physiology,

lymph is efficiently returned to blood circulation at the thoracic and right lymphatic ducts

where the two systems meet [1, 2]. Lymph nodes and immune cells continually monitor lymph

on its way back to circulation. Immune cells migrate into and out of lymphatic vessels by direct

contact with molecules on the surface of lymphatic endothelial cells (LECs) [3, 4].

Far from being ‗just‘ a supportive layer of tissue encasing a unidirectional flow of

lymph that may occasionally harbour dangerous hitch-hiking cancerous cells, the lymphatic

endothelium has been shown to actively attract and recruit tumour cells [5-13]. Similarly,

instead of being a passive protective lining of a fluid conduit, the lymphatic endothelium has

been shown to be highly adaptive to changes in tissue microenvironment [14-18]. The

lymphatic endothelium actively participates in local inflammatory responses [19-26] and is

capable of modifying both lymph flow [27-34] and composition [35-39] in response to various

stimuli.

Studies of interactions between lymphatic vessels and putative therapeutic molecules or

drug delivery systems offer great illustrations of the specialized function of the lymphatic

endothelium as distinct from the blood endothelium. Supersaxo and colleagues observed that as

the molecular weight of a subcutaneously applied protein increased, the absorption of the

Page 17: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

3

molecule by lymphatics increased, and that proteins larger than 16kDa were exclusively cleared

from the site of injection by lymphatics and not by blood vessels [40]. Liposome drug carriers

do not directly enter blood circulation but must first be taken up by lymphatic vessels in a

manner that is dependent on the size of the liposomes as it relates to the interstitial pressure at

the sight of subcutaneous injection [41]. Kaminskas and colleagues were able to control

whether a drug was taken up directly by the blood circulatory system, foregoing the lymphatics,

or was preferentially taken up by lymphatic vessels by varying the length of 4-benzene

sulphonate or polyethylene glycol conjugated to the nanoparticular drug carrier made of

polylysine dendrimers [42]. These examples clearly show that phenotypic differences between

the lymphatic and blood vasculature have profound impact on practical applications such as

drug absorption and drug delivery systems. Building in vitro tools to understand these

phenotypic differences would complement efforts to intelligently design therapeutics targeting

one or both of these systems.

Another clear example of why it is important to understand cell-specific regulatory

mechanisms in the design of therapeutics is the case of the angiopoietins. Induction of Ang-2,

an established agonist of lymphangiogenesis [43, 44], has been shown to associate with

progression and prognosis of a variety of human cancers; high expression of Ang-2 in

malignant tissue correlates with poor survival rate, and/or high frequency of metastasis, and/or

high microvascular density in patients with advanced colorectal carcinoma, breast cancer,

gastric carcinoma, hepatocellular carcinoma, non-small cell lung cancer, prostate cancer, and

ovarian cancer [45-54]. In contrast, expression of Ang-1, an established agonist of

angiogenesis [55, 56], in these tumours was often found to be at comparatively low to

undetectable levels. What effects does Ang-2 have on tumour LECs as compared to tumour

Page 18: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

4

blood endothelial cells (BECs)? More importantly, what cell-specific effects would anti-cancer

therapies that target Ang-2 function have on the normal healthy lymphatic or blood

endothelium?

Although Ang-2 is a specific case, it can be expected that Ang-2 will not remain a

unique example as evidence continues to accumulate implicating direct lymphatic involvement

in pathologies beyond cancer and lymphatic insufficiency. The lymphatic system has been

shown to be potentially directly involved in chronic inflammation [57, 58], acute systemic

inflammatory state (SIRS) and multiple organ dysfunction syndrome (MODS) [59], diabetic

nephropathy [60], heart disease [61-63], hypertension [64], systemic sclerosis [65],

hyperlipidemia [66] and obesity [67]. Thus, lymphatic endothelial regulators other than the

angiopoietins will inevitably emerge as key targets in proposed therapies. Since it is reasonable

to propose that lymphangiogenic regulators would tend to also have angiogenic effects, striving

to understand the mechanisms of cell-type-specific responses elicited by regulators of lymphatic

vessel functions can therefore aid rational design of drugs with selectivity for one system or the

other.

The studies recorded in these chapters all share the common goal of answering the

research question: What molecular signatures give rise to the phenotypic character of the LEC

lineage distinguishable from BEC lineages? The establishment of an in vitro system of LECs,

venous, and arterial ECs (VECs and AECs) derived from vessels of bovine mesentery (chapter

2) made possible the application of mass spectrometry for the comparative study of 2D-PAGE-

resolved protein profiles of these three endothelial cell (EC) types (chapter 3), and the use of

subtractive immunization (SI) for the production of a monoclonal antibody (mAB) selectively

reactive to a surface protein of LECs (chapter 4). These studies share the hypothesis that an

Page 19: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

5

unknown number of unidentified proteins is responsible for maintaining lymphatic

endothelium distinctiveness and for executing unique lymphatic system functions.

Understanding the mechanisms of cell-type-specific responses elicited by regulators of

lymphatic vessel functions as distinct from blood vessel functions, can aid the rational design of

drugs specifically targeting one system and not the other.

1.2. Introduction to Lymphatic Anatomy

1.2.1. Lymphatic Anatomy and Function

The lymphatic system is a series of vessels that form a one-way active drainage system

for protein-rich fluid containing clotting factors, lipids and lymphocytes from peripheral tissues.

After this fluid is collected from tissue spaces by initial (or terminal) lymphatic capillaries, is it

milky white in appearance and is then called lymph. From initial lymphatics, lymph is pumped

into larger pre-collecting and collecting lymphatic vessels, which propel the fluid by rhythmic

contractions of lymphangions separated by valves. From collecting lymphatics, lymph is

sequentially pumped into larger prenodal lymphatic vessels, lymph nodes, and postnodal

lymphatic vessel trunks. Lymph from legs, abdomen, the left arm, and the left side of the head

and chest, return to venous circulation at the left base of the neck via the junction between the

thoracic lymphatic duct and the left subclavian vein. Lymph from the right arm, thorax, the

right side of the head and chest, return to venous circulation via the junction between right

lymphatic duct at the right base of the neck [1, 2, 68, 69].

Salient features of the lymphatic system exist that are absent in the blood circulatory

system. Firstly, unlike the blood circulatory system, the lymphatic system does not circulate

lymph. As afore mentioned, lymph formed at initial lymphatics does not return to the point of

Page 20: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

6

origin in tissues through lymphatics. Lymph flow ends when lymph empties into venous blood

at the base of the neck. Secondly, initial lymphatics consisting of a single layer of ECs and

devoid of support cells are blind-ended tubes that are more flat, larger (10-60 µm as compared

to blood capillary diameters of about 8 µm) [68, 69] and are much more selectively retentive of

large molecules (20-100 nm or larger than 12kDa) than blood capillaries, which is more

permeable to particles of a few nanometres [38, 40, 69, 70]. Thirdly, ―anchoring filaments‖ are

hypothesized to anchor the outer flap of over-lapping initial LECs to surrounding tissues,

maintaining vessel patency, and allowing the inner flap to form a one-flap valve, thereby

stopping fluid from gushing freely back into the interstitium [69, 71]. Fourthly, the opening

and closing of discontinuous lymphatic endothelial cell-cell junctions is an active process

highly responsive to pressure changes in the interstitium [15, 16]. Fifthly, lymphangion

contractility in larger lymphatic vessels (responsible for the uni-directionality of lymph flow) is

tonal, phasic, and highly coordinated as lymph is actively pumped from one lymphangion

compartment to the next [72]. Finally, lymphatic muscle cells surrounding the endothelium in

larger lymphatic vessels are highly specialized contractile cells called lymphatic muscle cells

(not smooth muscle cells) innervated by surrounding non-myelinated nerve fibres [69, 73].

These lymphatic characteristics in combination produce a highly adaptive system

responsive to physical changes in tissue microenvironment. For example, Poggi and colleagues

observed differences between the lymphatic vessels of the mucous membrane/sub-mucous layer

of the bladder regularly subjected to changes in volume and pressure, and the intramuscular

lymphatic vessels located at trigone regions where changes in volume and pressure are virtually

absent [17]. The most notable difference was the presence of elastic and collagen fibres joining

the lymphatic vessel wall to surrounding tissues in the former and the absence of those fibres in

Page 21: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

7

the latter [17]. This comparative observational study of lymphatics in the urinary bladder has

probably produced some of the clearest evidence for the adaptability of lymphatic vessels to

tissue microenvironment. This adaptability is especially important considering the crucial role

the lymphatic endothelium plays in immunity. A discussion of the lymphatic endothelium as a

vital participant in the body‘s immune system follows in the next section.

1.2.2. Lymphatic Endothelium Function in Immunity

More instantaneous adaptive responses exhibited by the lymphatic endothelium have

been demonstrated in inflammation when interstitial fluid pressure and lymphatic drainage are

both generally thought to increase [14]. Marchetti and colleagues showed that the lymphatic

endothelium in inflamed dental pulp exhibited distended endothelial walls accompanying

opening of cell-cell junctions, and disappearance of micro-pinocytotic vesicles, which are

suggestive signs of an endothelium optimized for bulk lymph drainage [26]. Miteva and

colleagues found expression level of the dendritic-homing chemokine (Cysteine-Cysteine

motif) ligand 21 (CCL21) in LECs to correlate positively with transmural flow rate [14].

These observations suggest that the lymphatic endothelium dynamically responds to

inflammatory events by preparing appropriately for increased intramural flow and leukocyte

infiltration.

In addition to increased lymph drainage, other inflammatory changes to the tissue

microenvironment have been shown to activate the lymphatic endothelium. Johnson and

colleagues showed inflammatory cytokines TNF-α and TNF-β (tumour necrotic factors) to

stimulate LEC expression of other proteins important for leukocyte transmigration and homing

to the lymphatic endothelium, such as intercellular adhesion molecule 1 (ICAM-1), vascular

Page 22: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

8

cell adhesion molecule 1 (VCAM-1), E-selectin, CCL5, CCL2, and CCL20 [19]. More direct

involvement of the lymphatic endothelium with inflammatory events were shown by Sawa and

colleagues, who showed that these same leukocyte adhesion and homing proteins along with the

chemokines (Cysteine-X-Cysteine motif) ligands CXCL1, 3, 5, 6, and 8 were induced in LECs

with lipoteichoic acid [20], a major cell-wall component of Gram-positive bacteria. In fact, the

lymphatic endothelium serves the function of immune sentinel by detecting the presence of

pathogen-associated molecules such as lipoteichoic acid through LEC expression of receptors

central to innate immunity responses: Toll-like receptors TLR2 and 4 [24] and TLR6 and 9

[25]. These observations suggest that LECs are highly sensitive to inflammatory events and

play an integral role in the body‘s response to infection.

In the absence of high interstitial fluid pressure and therefore high lymph load during

infection, the lymphatic endothelium has been shown to be suppressive of immune cell activity.

Podgrabinska and colleagues demonstrated that in the absence of pathogen-derived signals,

high dendritic cell expression of CD86 correlated with adhesive interactions with LECs and

attenuation of dendritic cell maturation and its ability to stimulate T-cells [22]. These results

imply that in addition to providing support during active inflammation, the resting lymphatic

endothelium contributes to prevention of undesired immune reactions and thereby provides an

important service to bodily health. A discussion of other functions of the lymphatic

endothelium in homeostasis follows below.

1.2.3. Lymphatic Endothelium Function in Homeostasis

In health, cardiovascular homeostasis is maintained by the ability of blood vessel walls

to constrict or relax when appropriate. The dependency of blood vessel vasoconstriction and

vasodilatation on intact functional endothelium has been well established since 1980 [74-79].

Page 23: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

9

By contrast, the role the lymphatic endothelium plays in regulating lymphatic vessel tone,

relaxation, and pumping action has only been explored with significant depth since 1990 [27-

34, 72, 73]. Like the arterial endothelium and unlike the venous endothelium, which is

relatively unresponsive to the neurotransmitter acetylcholine (ACh) [74-76, 79], the lymphatic

endothelium was shown by Ohhashi and Takahashi to release a vessel muscle ―relaxing factor‖

upon stimulation with ACh [27]. The observation by these researchers that mechanical ablation

of the lymphatic endothelium suppressed responsiveness of the vessel ring to ACh was

corroborated by Ferguson [33], and Hashimoto and colleagues [34]. One of the relaxing factors

released by the lymphatic endothelium upon stimulation with fluid flow or ACh was later

shown to be very likely nitric oxide (NO) [28, 80]. These studies establish the dependency of

lymphatic vessel tone regulation on LEC-derived factors.

Whether ACh and LEC-derived factors also play a role in the regulation of phasic

contractions of lymphangions in larger lymphatic vessels was examined by Yokohama and

Ohhashi [29]. The researchers studied spontaneous phasic contractions in bovine mesenteric

lymphatic vessels and found ACh to dose-dependently reduce both the rate of phasic

contractions (negative chronotropic effect) and the strength of contractions (negative inotropic

effect) [29]. The vessels studied had a rhythmic contraction rate of approximately 3 beats per

minute that was lowered to approximately 2 beats per minute in the presence of ACh. The

strength of contraction was lowered by approximately 10% in the presence of ACh. More

importantly, physical ablation of the endothelium abolished these effects suggesting that LEC-

derived factors such as NO or other related compounds mediate not only lymphatic vessel tone

but also vessel pumping action [29]. The dependency of lymphatic vessel phasic contractility

regulation on an intact and functional lymphatic endothelium was confirmed in other studies by

Page 24: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

10

Elias and colleagues [31], von der Weid and colleagues [32], Ferguson [33], and Hashimoto and

colleagues [34].

Augmenting these studies are findings by Koller and colleagues, whose findings suggest

that LEC-dependent chronotropic regulation can be separated from inotropic regulation of

lymphatic vessel phasic contractions [30]. The researchers used lymphatic microvessels from

rat iliac to demonstrate increases in intramural flow reduced the amplitude of phasic

contractions by approximately 40%, but increased frequency of rhythmic contraction by

approximately 20% [30]. Interestingly, increased intramural flow did not change lymphatic

vessel diameter suggesting that LEC-regulated changes in lymphatic vessel tone (or resistance),

phasic contraction, and contractile strength can be modulated independently of one another to

respond sensitively to fluid pressure dynamics in local interstitial environments. Studies of the

kind described here, thus far, are few in number. However they all add insightful explanatory

value to clinically observed severity and complexities of lymphoedema and related lymphatic

insufficiency disorders. As basic knowledge about lymphatic system anatomy and function

continues to accumulate, the list of human pathologies which involve the lymphatic system

grows longer. Established and emerging lymphatic-associated disorders are discussed in detail

in the next two sections.

1.2.4. Lymphatic Endothelium Dysfunction in Lymphoedema

The most well studied lymphatic-associated disease is called lymphoedema, which is an

umbrella term for lymph transport insufficiency resulting from congenital lymphatic

vessel/valve malformation (primary lymphoedema), or from interference with normally formed

functional lymphatic vessels/valves by injury, infection, or iatrogenic procedures (secondary

lymphoedema). Congenital lymphoedema is represented by Milroy‘s and Meige‘s syndromes,

Page 25: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

11

both of which show an autosomal-dominant pattern of inheritance and are caused by mutations

of the vascular endothelial growth factor receptor 3 (VEGFR-3/Flt-4) tyrosine kinase receptor

and the forkhead box protein C2 (FOXC2) transcription factor, respectively [81]. Primary

lymphoedemas like these syndromes are usually characterized by swelling of both limbs

whereas secondary lymphoedema is more localized to limbs normally served by the obstructed

lymphatic vessel. Incidences of secondary lymphoedema in North America are predominantly

related to complications following surgical treatment of breast cancer, melanoma, gynecologic

cancers, lymphoma and urologic cancers [82]. Secondary lymphoedema in the developing

world is predominantly related to infection with the nematode worm Wuscheria bancrofti

(lymphatic filariasis), which matures in lymphatic vessels and causes disruption to lymph

transport [82].

In general, failure of lymph transport functions results in a build-up of protein-rich fluid

in tissues drained by lymphatic vessels at the point of dysfunction. Over time, mass swelling of

affected area worsens due to deposition of fat, collagen and other fibrous tissues, abnormal

localized tissue growth and skin thickening, and inappropriate accumulation of immune cells

[82-86]. Since lymphatic vessels play a vital role in immunity and inflammatory responses,

lymphoedema often accompanies local immune suppression, and associated complications such

as chronic bacterial infections of the skin (erysipelas), chronic inflammation of the skin

(dermatitis) of lymphatic vessels (lymphangitis) and of subcutaneous tissues (cellulitis), and

consequently delayed wound healing [87-93]. In severe chronic lymphoedema, outcomes can

range in severity from disfigurement and resultant psychological distress, loss of sensory nerve

function, and loss of limb mobility due to sepsis, gangrene, and amputation [94-97].

Page 26: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

12

Venous capillaries play a similar role to lymphatic capillaries in the maintenance of

homeostasis. However, important differences have significant implications for the differential

diagnosis of lymphatic disorders and the management of treatment strategies. This can be

highlighted by noting that the terms ‗oedema‘ and ‗lymphoedema‘ are not interchangeable and

that the unique biology of the lymphatic system allows it to play a unique role in maintaining

protein-fluid balance.

In the resting adult human, approximately 15 mL of plasma-protein-rich fluid filters out

at the arterial end of capillary beds each minute. Approximately 80-200 g of plasma protein is

extravasated into interstitial spaces with this fluid. At the venous end of capillary beds, 12-13.5

mL of extravasated fluid is reabsorbed each minute with the exclusion of extravasated plasma

macromolecules. The remaining concentrated filtrate is effectively removed from interstitial

spaces by a functional lymphatic system [68, 71]. Congestive heart failure, venous

hypertension, and chronic venous insufficiency are causes of disturbance to hydrostatic forces

governing fluid reabsorption at the venous end of capillary beds [98]. Failure to reabsorb

extravasated fluid at the venous end of capillary beds can result in fluid oedema and tissue

swelling without the immune dysfunction frequently associated with pure lymphoedema [99].

Typical treatment for the ensuing oedema is diuretics. By contrast, swelling associated with

lymphoedema and other related disorders cannot be resolved by diuretic therapy or any other

existing molecular or pharmaceutical intervention [98]. Standard treatment strategies of

lymphoedema revolve around multilayer compression bandaging, mechanical massage,

physical therapy involving skin hygiene, limb compression, regimented exercise, and as a last

resort, surgical resection [82].

Page 27: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

13

Unfortunately, these treatment strategies for lymphoedema, in development since the

early 1900s, have not improved dramatically since the 1960s [100]. It is hoped that initial

characterizations of lymphatic involvement in human pathologies other than lymphoedema over

the past 10 years will continue to renew research interest in the unique biology and function of

lymphatic vessels, and thereby lead to more speedy progress toward effective lymphatic

endothelium-targeted therapies.

1.2.5. Lymphatic Endothelium Dysfunction in other Human Pathologies

The variety of human pathologies in which lymphatic vessels as implicated reflects the

lymphatic endothelium‘s diverse functions as discussed in sections 1.2.2. and 1.2.3. Firstly, in

diseases with presentation of chronic inflammation, aberrant inflammation-driven

lymphangiogenesis is thought to produce lymphatic endotheliums that exacerbate the

inflammation [57] rather than resolves or suppresses it as a healthy lymphatic endothelium

normally would [22]. This phenomenon has been observed in skin psoriasis [101], Crohn‘s

disease [102], Kaposi‘s sarcoma [58], tubulointerstitial fibrosis (nephropathy) [60], and

systemic sclerosis [65]. Secondly, in familial combined hyperlipidemia, low-grade

inflammation is thought to promote growth of new lymphatic vessels which facilitate fat

deposition thereby contributing to obesity [66, 67]. Thirdly, proper lymph transport is thought

to be a requirement of healthy chronotropic and inotropic regulation of heart contractions since

cardiac lymph flow impairment is concomitantly observed with arrhythmia, cardiac injury,

terminal heart failure, and even transplant rejection [61-63]. Fourthly, the endothelium as a

manufacturer of secreted factors has a potentially impactful role in SIRS/MODS [59] and

hypertension induced by high-salt diet [64].

Page 28: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

14

From results of work with animal models of major trauma and shock, Deitch and

colleagues proposed the hypothesis that yet-to-be-identified factors derived from lymphatics in

the gut are responsible for systemic vascular damage and subsequent organ failure in SIRS and

MODS [59]. From their work with rats fed a high salt diet, Machnik and colleagues proposed

that clearance of excess sodium from interstitial spaces and thereby maintenance of volume and

blood pressure homeostasis depend on communication between lymphoreticular cells and the

lymphatic endothelium [64]. Inhibition of the resultant increase in the expression of endothelial

NO synthase in LECs led to elevated blood pressure [64].

Finally, the functions of the lymphatic endothelium in cell trafficking in immunity,

lymphangiogenesis (growth of new lymphatic vessels), lymph formation and transport, are co-

opted by metastasizing cells to disseminate in the body. For example, constitutive LEC

expression of lymphocyte attractants CCL19 and CCL21 promotes CCR7-positive melanoma

chemotaxis toward lymphatics and migration to regional lymph nodes (CCR7 is the Cysteine-

Cysteine motif chemokine receptor of CCL19 and 21) [5, 12, 13]. For another example, tumour

cells have been shown to produce the LEC proliferation inducer VEGF-C (vascular endothelial

growth factor-C), and both the increase in tumour-associated lymphatic vessel number and

lymph node metastasis could be blocked by inhibiting the VEGF-C receptor VEGFR-3/Flt-4

[9]. VEGF-A dependent tumour-associated lymphangiogenesis in lymph nodes also potentially

contributes to tumour cell dissemination via lymphatic vessels [6-8]. These few studies suggest

that the lymphatic endothelium plays an active role in tumour cell recruitment, dissemination to

lymph nodes and perhaps even metastatic spread to distant sites in the body [6, 7, 11].

Wong and Hynes took and step further and proposed the hypothesis that tumour cells

may ―prefer‖ to disseminate via the lymphatics over blood vessels. The review authors

Page 29: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

15

proposed that gaps in LEC intercellular junctions (unlike the tight inter-endothelial junctions

characteristic of BECs), lack of support cells surrounding LECs in smaller lymphatic vessels

(that more frequently surround BECs in comparable blood vessels), and the low flow/shear

stress fluid environment within lymphatic vessel lumens make lymphatics the ideal conduit for

tumour cell metastasis [10]. This intriguing hypothesis deserves further scrutiny. Research

focus on understanding the ways that LECs are similar to and different from BECs in function

in contribution to cancer metastasis may help to shed light on the proposition. Such research

focus may also help to expand the range of potential therapeutic targets and designs possibly

conceived to treat cancer metastasis.

Research interest in the unique biology of the lymphatic endothelium and in its role in a

large variety of human pathologies, presented in this section, have produced some of the most

impactful publications in the field within the past ten years. This was made possible by the

discovery and characterization of LEC protein markers starting with VEGFR-3/Flt-4 in 1995

[103]. A brief summary of known lymphatic endothelium markers follows below.

1.2.6. Protein Markers of the Lymphatic Endothelium

Research in the past two decades has contributed most to what we now know about the

unique biology of the lymphatic system, despite over two centuries of effort [100]. This is

largely due to the discovery of lymphangiogenic factors VEGF-C and VEGF-D [104] and

specific LEC protein markers VEGFR-3/Flt-4 [103], lymphatic vessel endothelial hyaluronic

acid receptor 1 (LYVE-1) [105] , and prospero homeobox protein 1 (Prox-1) [106] in 1995,

1999, and 2002, respectively. Specific LEC protein markers made possible visualization and

characterization of LECs, the lymphatic endothelium, and lymphangiogenesis in studies of

various human diseases. Equally importantly, specific LEC protein markers make possible the

Page 30: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

16

isolation and validation of cultures of LECs for in vitro experiments. While modern progress in

lymphatics research is palpable, a balance of caution and optimism is warranted considering the

mixed success of outcomes from recent endeavours to discover novel markers of LECs.

The advent of microarray technology created opportunities to profile entire

transcriptomes in isolated LECs and BECs in comparative studies. All 30 novel candidate

markers of LECs with expression of at least three fold higher in LECs than in BECs listed in the

review by Farnsworth, Achen and Stacker [11] were identified in two microarray studies both

utilizing the Human GeneChips-U95Av2; Podgrabinska and colleagues contributed 40% of the

list [36], and Hirakawa and colleagues contributed 60% of the list [107]. Only four out of the

30 novel candidate markers were validated by RT-PCR, two by other means, and the rest of the

list is yet to be validated. Despite the fact that both studies used LECs sorted from

disaggregated human neonatal foreskins, only one novel candidate marker was found in

common between the two microarray studies: RANTES or CCL5, a member of the 8-kDa

family of pro-inflammatory cytokines with chemotactic activity for monocytes and T-cells

[108, 109]. The high expression of CCL5 in LECs is consistent with their purported role in

immunity and inflammation.

Furthermore, consistent with the active role lymphatics are thought to play in cellular

trafficking in immunity and inflammatory responses, Hirakawa and colleagues found LECs to

express a broader range of cytokines than BECs [107]. Consistent with the purported role

LECs have in modifying lymph flow and composition, Podgrabinska and colleagues found

enrichment in genes involved in intracellular protein transport, vesicle formation and fusion,

and vesicle associated membrane proteins [36]. Consistencies notwithstanding, many of the 30

Page 31: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

17

genes on the list would need to be extensively validated and characterized before they can be

considered bona fide protein markers of LECs.

Several good reasons exist for follow-up experiments. One reason concerns

reproducibility of microarray results. Other reasons have to do with the imperfect fidelity of

identified protein markers of LECs and BECs. Podgrabinska and colleagues used LYVE-1-

coated magnetic beads to purify LECs from disaggregated mixed cultures of dermal cells [36].

Cause for concern with this approach is the discovery of LYVE-1 expression in pleural

mesothelial cells [110] and macrophages [111]. The LEC cultures in this study may not be

contaminated with pleural mesothelial cells absent in skin, but would highly likely to be with

macrophages present in skin. Similarly, Hirakawa chose to define LECs as Sialomucin-

negative and platelet/endothelial cell adhesion molecule-1 (PECAM-1)-positive cells, and

BECs as Sialomucin-positive cells in similar cell sorting procedures with magnetic beads [107].

Cause for concern with this approach is the expression of Sialomucin in eosinophils and mast

cells, both likely to be present in skin [112]. More disconcertingly, subtypes of macrophages

are also known to be Sialomucin-negative and PECAM-1-positive [113].

The concern of imperfect fidelity of identified protein markers of LECs also applies to

Prox-1 and the vascular endothelial growth factor receptor VEGFR-3/Flt-4, which, along with

LYVE-1, are also expressed by pleural mesothelial cells [110]. Another well known marker of

LECs, podoplanin, was a podocyte protein before its expression in LECs and not in BECs was

discovered [114]. More recently, podoplanin was found to be expressed by peritoneal

mesothelial cells, osteocytes, glandular myoepithelial cells, ependymal cells, and other cells of

the lymphoreticular system [115]. A good brief summary of the limitations of these and other

LEC markers is the review by Baluk and McDonald [116].

Page 32: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

18

One way to circumvent marker infidelity is to incorporate into best practices the use of

multiple markers to define cells of interest as belonging to the lymphatic endothelial lineage

prior to experimentation. Another way to circumvent marker infidelity is to functionally

characterize cells of interest prior to experimentation. If cells of interest behave in culture as

LECs are expected to behave, for example, in terms of responsiveness to known

lymphangiogenic factors such as VEGF-C and Angiopoietin 2 (Ang-2), then they are more

likely to belong to the lymphatic endothelial lineage.

The VEGF and the angiopoietin family of proteins have vessel-type specific effects.

VEGF-A signalling through its cognate receptor VEGFR-2 induces sprouting angiogenesis

(growth of new blood vessels) in vivo, promotes BEC proliferation, survival, migration, and

permeability in vitro (reviewed in [117]). VEGF-C signalling through its cognate receptor

VEGFR-3/Flt-4 induces sprouting lymphangiogenesis in vivo, and promotes LEC proliferation,

survival, and migration in vitro (reviewed in [118]). As reviewed by Thurston [119], knocking

out Ang-1 or its cognate receptor tyrosine kinase Tie-2 in mouse results in embryonic lethality

due to defects in the heart and to under-development of the blood vasculature. Knocking out

Ang-2 results in lymphatic vasculature under-development and blood vessel defects in the

retina [119]. Unlike VEGF-A and -C which have selective affinity for VEGFR-2 and VEGR-3,

respectively, both Ang-1 and Ang-2 can bind to and activate Tie-2 in vitro [120]. Mechanisms

by which these endothelial cell-type and vessel-type specific effects take place are currently

unknown. These observed effects provide rationale for further investigation of the lymphatic

endothelium as anatomically and functionally distinct from the blood endothelium.

Page 33: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

19

1.3. Data Chapter Organization

Chapter 2

This chapter is a description of the isolation and validation an in vitro culture system of

lymphatic, venous, and arterial endothelial cells derived from vessels of bovine mesentery.

Chapter 3

This chapter is a description of the comparative study of 2D-PAGE-resolved protein

profiles of these three EC types described in chapter 2. This comparative analysis examines the

reproducibility and feasibility of mass spectral analysis of 2D-PAGE in the attempt to identify

novel cell-type specific proteins expressed by the three endothelial cells.

Chapter 4

This chapter is a description of the use of the three EC cultures for the production of a

monoclonal antibody selectively reactive to a potential surface protein marker of lymphatic

ECs. The attempted procedure examines the feasibility of subtractive immunization as a tool

for the discovery of novel surface markers unique to the lymphatic endothelium.

Page 34: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

20

1.4. Attributions

Chapter 2

Jason Trinh carried out reverse transcriptase PCR experiments from RNA prepared by

me. Dr. Harold Kim provided the designed primers for the PCR experiments. Dr. Stephen

Chen assisted in counting cells in the migration assay. Dr. Brenda Coomber provided

laboratory space for the isolation of the endothelial cells. All contributed to the editing of the

manuscript. Raj Kukreja obtained bovine mesentery from the abattoir. This chapter was

published on March 6th

2007 in BMC Cell Biology 8:10

Chapter 3

Dr. Natalie Rodrigues performed two-dimensional polyacrylamide gel electrophoresis

(2D-PAGE) using my cell preparations and helped me with the silver-staining of the gels.

George Hanna and Dr. Eric Yang assisted with the quantitative analysis of the gel spots.

George Hanna excised gel spots of interest. Dr. Eric Yang provided technical support in

identification of proteins by mass spectrometry. Katie Pizzuto provided assistance with some

of the immunoblots (IBs). Dr. Paul Van Slyke isolated the bovine aortic arterial endothelial

cells (baAECs). This chapter was published on Feb 22, 2010 in Proteomics 10:1658

Chapter 4

Maribelle Cruz and the Animal Health Technicians of the Department of Comparative

Research cared for the mice and performed injections on mice as needed. Meena Bali prepared

the ECs I provided her for injections, produced the hybridomas, and screened about a quarter of

the hybridoma colonies. Dr. Eric Yang provided technical support for mass spectrometry work.

Page 35: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

21

CHAPTER 2

LYMPHATIC, VENOUS AND ARTERIAL ENDOTHELIAL CELLS

ISOLATED FROM BOVINE MESENTERIC VESSELS

RETAIN THEIR DEFINING CHARACTERISTICS IN CULTURE

AND ARE DIFFERENTIALLY RESPONSIVE TO ANGIOPOIETIN-1 AND -2

Page 36: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

22

2.1. Introduction

Tie-2 (tyrosine kinase with immunoglobulin-like and EGF-like domains 2) is the

receptor tyrosine kinase for the angiopoietin family of ligands (Ang-1, 2, 3, and 4). The role of

Tie-2 in endothelial cells has been extensively studied over the years, and the discovery of

impaired lymphatic vessel patterning and function in Ang-2 knockout mice has since added

extra complexity to this growing field [43]. Tie-2 has also been shown to be expressed in

lymphatic endothelial cells [121]. In vivo studies using slow-release pellets of an

engineered form of Ang-1, cartilage oligomeric matrix protein-Angiopoietin-1 (COMP-Ang-

1), have further showed induction of angiogenesis and ectopic lymphangiogenesis in mouse

cornea [121]. In addition, over-expression of Ang-1 in the skin of mouse ears via recombinant

adeno-associated virus gene delivery induced lymphatic endothelial cell proliferation,

lymphatic vessel enlargement, sprouting, and branching [55].

Prior to the discovery of the involvement of Tie-2 and its ligands in lymphangiogenesis,

the role of Tie-2 and it ligands in angiogenesis was the focus of mouse genetic studies. Ang-1

deficient mice exhibited phenotypes similar to those of Tie-2 knockout mice. These phenotypes

included impaired myocardial trabeculation and endocardial development as well as lack of

perivascular cell recruitment to endothelial cells undergoing angiogenesis [56]. Ang-2 was

initially characterized as an Ang-1 competitive antagonist since transgenic over-expression of

Ang-2 produced angiogenic defects resembling those of Ang-1 or Tie-2 knockout mice [44].

In recent studies, evidence suggesting condition-dependent agonistic roles for Ang-2

brought into question the initial characterization of Ang-2 as simply a competitive antagonist of

Ang-1. Ang-2 has been shown to activate Tie-2 phosphorylation at high concentrations leading

to cell survival via the PKB signalling pathway [122]. Following a 24-hour pre-treatment,

Page 37: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

23

Ang-2 can lead to vessel growth in a fibrin matrix model [123]. Ang-2 can also induce tubule

formation in murine brain capillaries and promote endothelial cell migration [124]. In addition

to inhibiting JNK/SAPK phosphorylation, Ang-2 has been shown to induce phosphorylation of

Tie-2, PKB, Erk1/2, and p38 members of the mitogen activated protein kinases in the human

umbilical vein endothelial cells (HUVEC) [125]. Other functions of Ang-2 in vasculogenesis

include promotion of EC adhesion independently of Tie-2 [126], regulation of differentiation of

cells surrounding the cortical peritubular capillaries of the kidneys [127], and modulation of

retinal and hyaloid blood vessel remodelling [128].

Complementing these studies are mouse knockout experiments that further elucidate the

importance of Ang-2 in endothelial cell function. Ang-2-null mice (strain 129/J) exhibited

defects in the hyaloid blood vasculature and gross defects in lymphatic remodelling [43]. Skin

oedema in Ang-2-null mice correlated with improper recruitment of support cells such as

lymphatic muscle cells (LMCs) responsible for the contractile function of lymphangions in

peripheral or dermal lymphatic vessels [43]. The Ang-2-null mice also developed lethal

chylous ascites and oedema in the peritoneal cavity around day 14, correlating with improperly

remodelled lacteals in villi, disorganization and hypoplasia of intestinal lymphatic capillaries,

and deficient smooth muscle association in mesenteric-collecting lymphatic vessels [43].

Therefore, the function of Ang-2 is not limited to antagonistic regulation of Ang-1 in Tie-2

signalling.

In this chapter, I demonstrate that bovine mesenteric venous, arterial, and lymphatic

endothelial cells (bmVEC, bmAEC, and bmLEC) respond differentially to Ang-1 and Ang-2.

Firstly, both Ang-1 and Ang-2 activated Tie-2 and downstream Erk1/2 in bmLEC. However,

whereas Ang-1 promoted migration in bmLECs, Ang-2 slightly more effectively promoted

Page 38: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

24

proliferation and survival in these cells. Secondly, Ang-1 more effectively activated Tie-2 and

downstream Erk1/2 in bmVECs and bmAECs than did Ang-2. However, whereas Ang-1

promoted survival, and migration in bmVECs and bmAECs, Ang-2 did not stimulate the same

responses in these cells. Whereas Ang-1 produced a small proliferative response in bmAECs,

neither Ang-1 nor Ang-2 produced a proliferative response in bmVECs. Taken together, my

results suggest cellular responses to Ang-1 and Ang-2 stimulation vary depending on the origin

of the ECs. These results lend support to the use of EC culture systems such as the one I have

built to compare and contrast signaling events downstream of vascular modulators such as the

angiopoietins simultaneously in ECs of different vessel origins.

2.2. Methods

2.2.1. BmEC isolation and culture

Methods for isolation and culture of bovine ECs are adapted from previously established

protocols [13, 14]. Sections of the gut mesentery were taken from freshly slaughtered cattle at a

local abattoir (Better Beef, Guelph, Ontario). The mesenteric sections were brought back to the

laboratory in warm phosphate buffered saline (PBS). Connective and fatty tissues, which were

superficially rinsed with 75% ethanol, were removed to expose lymph nodes and vessels.

Sterile 0.1% Evan‘s Blue dye was injected into the lymph nodes to highlight the lymphatic

vessels. Dye was flushed from excised vessels with warm PBS supplemented with penicillin

(100 Units/mL, Life Technologies), streptomycin (100 µg/mL, Life Technologies), and

fungizone (2.5 µg/mL Gibco, Invitrogen). Distal ends of excised vessels were occluded with

surgical suture in order to infuse a solution of dispase I (2.6 Units/mL, Roche) and collagenase

A (1 mg/mL, Roche). Vessels occluded at both ends to trap the enzyme solution were

Page 39: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

25

incubated in PBS at 37ºC for 10 minutes. Cells dislodged by the treatment were grown on

plates coated with 1% bovine gelatin (Sigma-Aldrich), in DMEM (Dulbecco‘s modified eagle‘s

medium, Sigma-Aldrich), and standard conditions.

Isolation of cells from venous and arterial vessels from bovine mesentery followed a

similar strategy as described for bmLECs. However, the use of dye was unnecessary.

2.2.2. Eliminating contaminating cells

In order to establish high-purity cultures of bovine mesenteric endothelial cells

(bmECs), a combination of strategies was used. Firstly, cells that were uninhibited by contact

and grew atop the monolayer of ECs were removed by several washes with trypsin such that the

monolayer underneath was not disturbed. Secondly, visible regions of the monolayer that did

not appear cobble stoned at confluency were removed by direct aspiration. Finally, limiting

dilution into 96-well plates was used to isolate groups of homogenous ECs. Groups of cells

comprising of more than 90% ECs were used in experiments. 90% estimates of purity were

based on monolayer cobblestone appearance.

2.2.3. Detection of transcripts by RT-PCR

Total RNA was prepared from cells isolated from bovine gut mesentery using Tri-

Reagent (Sigma-Aldrich) according to manufacturer‘s instructions. Synthesis of cDNA from

1µg of total RNA was done with Thermoscript reverse transcriptase (Invitrogen) according to

manufacturer‘s instructions. PCR was performed with Taq polymerase (Qiagen) with primer

sequences: Tie-1 (forward primer 5‘-TGA CTT TGC GGG AGA ACT GG-3‘, reverse primer

5‘-CTC CGA CCA GCA CGT TTC GG-3‘), Tie-2 (forward primer 5‘-GAT TTT GGA TTG

TCC CGA GGT CAA G-3‘, reverse primer 5‘-CAC CAA TAT CTG GGC AAA TGA TGG-

3‘), Neuropilin-1 (forward primer 5‘-CAG AAC GCT GCC CAC TGC AT-3‘, reverse primer

Page 40: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

26

5‘-CTT TCT GGG TCC TTT TTA TC3‘), VEGFR3 (forward primer 5‘-CGG TGC CCA GTG

CGT GGG ACG-3‘, reverse primer 5‘-TTG ACT AGC CAT CGT AGG ACA-3‘), Prox-1

(forward primer 5‘-TTG TCA CCC AAT CAC TTG AAA-3‘, reverse primer 5‘-CTT CCA

GGA AGG ATC AAC ATC-3‘) and GAPDH (forward 5‘-ACC ACA GTC CAT GCC ATC

AC-3‘, reverse primer 5‘-TCC ACC ACC CTG TTG CTG TA-3‘)

2.2.4. Cell imaging

BmECs were grown to confluency on gelatin-coated glass coverslips, fixed with 4%

paraformaldehyde in PBS for 10 minutes at room temperature, and permeabilized with 0.1%

TritonX-100 (Sigma-Aldrich) for 1 minute at room temperature. Fixed and permeabilized cells

were stained with haematoxylin and eosin (H&E) or with immunofluorescence using

antibodies: Prox-1(RDI), LYVE-1 (RDI), and podoplanin. The following protocol was used for

Rhodamine-Phalloidin staining: Fixed and permeabilized cells were incubated in Rhodamine-

phalloidin (Molecular Probes, 1:40 dilution) in PBS for 15 minutes, and washed. Cells were

mounted on microscope slides with aqua-polymount (Polysciences Inc.) or DAPI-containing

Vectashield mounting medium (Vector Laboratories) where appropriate. Bright-field

microscope images were produced with the Zeiss Axioplan 2 light microscope (Carl Zeiss).

Confocal images were produced using Carl Zeiss LSM 510 laser scanning confocal microscope.

Fluorescent images were produced using the Zeiss Axiovert 200M.

2.2.5. Stimulation of bovine ECs

BmECs grown on gelatinized plates were moved to bare plates 24 hours prior to

stimulation. Cells were stimulated for 15 minutes with human recombinant Ang-1 or Ang-2

(800 ng/mL, R&D Systems) in 10% FBS. For stimulations with recombinant Ang-1, which

contains a polyhistidine tag, anti-polyhistidine monoclonal antibody (anti-His, R&D Systems)

Page 41: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

27

was used to cluster the ligand according to manufacturer‘s instructions (ligand to antibody ratio

1:20). Where indicated, mock treatment refers to cells incubated with anti-His antibody alone.

2.2.6. Cell Lysis and Tie-2 Immunoprecipitation

Bovine ECs were washed twice with ice-cold PBS supplemented with 2mM activated

sodium orthovanadate (Sigma-Aldrich, see [129] for activation protocol). Cells were lysed on

ice for 30 minutes with RIPA lysis buffer (150mM NaCl, 1% Igepal, 0.5% Sodium

Deoxycholate, 0.1% SDS, 50mM Tris pH 7.4, 1mM EDTA) supplemented with 2 mM sodium

orthovanadate, and complete protease inhibitors (Roche). Tie-2 was immunoprecipitated from

equal protein amounts of cleared whole cell lysates with 2 µg of anti-Tie-2 antibody C-20

(Santa Cruz Biotechnology) pre-coupled to 25 µL protein A-sepharose (Amersham

Biosciences) for 1 hour.

2.2.7. Immunoblotting

Proteins were resolved on 10% PAGE gels and transferred to PVDF (Perkin Elmer)

membranes. Antibodies used in immunoblots were: anti- phosphotyrosine 4G10 antibody (1

µg/ml, Upstate Biotechnologies), phosphoTie-2-specific anti-pTyr992 antibody (1:1000, Cell

Signaling), anti-Tie-2 antibody 33.1 (0.5 µg/ml, BD Biosciences Pharmingen), and phospho and

pan Erk1/2 (1:1000, Cell Signaling).

2.2.8. 3H-Thymidine uptake assay

Bovine ECs were seeded in 96-well plates at a density of 4500 cells/well in 100 µL 10%

FBS DMEM. After 24 hours, 10% FBS DMEM was replaced with 1% FBS DMEM + anti-His

antibody (16 ug/mL, ―Mock‖), with 1% FBS DMEM + Ang-1 (800 ng/mL) clustered with anti-

His antibody (1:20 ligand to antibody ratio), or with 1% FBS DMEM + Ang-2 (800 ng/mL).

Page 42: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

28

Cells treated thus were pulsed for 6 hours with 2 µCi of 3H-thymidine (Amersham). After 6

hours, the 96-well plates were placed in -80ºC for cell lysis before 3H-thymidine incorporation

was measured with the TopCount NXT Microplate Scintillation and Luminescence Counter

(Packard).

2.2.9. Cell count assay

400 000 cells were plated in each well of 6-well plates at the start of the assay in

10%FBS media or in mock media supplemented with 800 ng/mL super-clustered Ang-1 or 800

ng/mL Ang-2 and cell numbers were monitored every 24 hours for 96 hours via cell counting

with the hemocytometer. Mock media of bmLECs consisted of 5% FBS, of bmVECs consisted

of 2.5% FBS, and of bmAECs consisted of 1% FBS. These percentages of FBS were required

to maintain the respective cells over 96 hours while maintaining a decent level of cellular

proliferation and avoiding cell death. All mock media contained 16 ug/mL clustering anti-His

antibody.

2.2.10. Modified Boyden Chamber Migration Assay

Bovine ECs were grown for 24 hours before they were serum starved for 2 hours in

0.1% FBS DMEM. Serum starved bovine ECs were then trypsinized and seeded in 500 µL of

0.1% FBS DMEM at a density of 84 000 cells/well in the upper chamber of 6 well plates

containing 8 µm pore-size inserts (Falcon). 1.5 mL of the following were placed in the bottom

wells: 10% FBS DMEM (positive control); 0.1% FBS DMEM + anti-His antibody (16 µg/mL,

―Mock‖); 0.1% FBS DMEM + Ang-1 (800 ng/mL) clustered with anti-His antibody (1:20

ligand to antibody ratio); 0.1% FBS DMEM + Ang-2 (800 ng/mL). BmECs migration occurred

over a 4-hour time period. Membrane of inserts were fixed and stained with filtered Harris‘

hematoxilin prior to mounting on microscope slides with aqua-polymount (Polysciences Inc.).

Page 43: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

29

2.2.11. Cell death ELISA

Bovine ECs were seeded in 6-well plates at a density of 100 000 cells/plate and grown

for 24 hours in standard tissue culture conditions and DMEM supplemented with 10% FBS

before the start of the assay. To examine cell death due to serum starvation, 10% FBS DMEM

was replaced with 0.1% FBS DMEM + anti-His (16 µg/mL), with 0.1% FBS DMEM + Ang-1

(800 ng/mL, clustered with anti-His antibody), or with 0.1% FBS DMEM + Ang-2 (800

ng/mL). In order to achieve detectable levels of cell death, bmLECs were harvested after 24

hour serum starvation, bmVECs after 48 hours, and bmAECs after 72 hours. Cell death was

determined by quantifying cytoplasmic histone-associated-DNA-fragments (mono- and

oligonucleosomes) via the ELISA (enzyme-linked immunosorbent assay, Cell Death ELISAplus

,

Roche) according to manufacturer‘s specifications.

2.3. Results

2.3.1. BmLEC isolation and culture.

In order to distinguish translucent lymphatic vessels from surrounding fatty tissue,

Evan‘s blue dye was injected into exposed mesenteric lymph nodes. The blue staining of post-

nodal lymphatic vessels facilitated excision and processing of the vessels to extract ECs. Blood

vessels remained red after dye injection (Figure 2.1.A) and therefore could not be mistaken for

lymphatic vessels. Initial cultures of cells extracted from dispase- and collagenase-treated

excised bovine mesentery lymphatic vessels were at least 50% endothelial based on cellular

morphology. Cells cultured from treated vessels were a mixture of LMCs, fibroblasts, and

cobble stoned bmLECs.

Unlike high-purity endothelial cell populations, a mixed cell population did not form a

Page 44: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

30

cobblestone monolayer when grown to confluency (Figure 2.1.B). The morphologies of the

different cells within the mixed population were visually distinguishable. A mixed population

of predominantly LMCs, fibroblasts, and a few bmLECs formed circular ―swirly‖ patterns

(Figure 2.1.C). This morphology has previously been described by Leak and Jones, and

Johnston and Walker [130, 131]. Populations of cells that predominantly consisted of

fibroblasts (Figure 2.1.D) or LMCs (Figure 2.1.E) had distinct morphologies unlike the cobble

stoned appearance of bmLEC at confluency (Figure 2.1.F).

To obtain purity greater than 90%, contaminating LMCs and fibroblasts were eliminated

from the culture by direct aspiration, differential trypsinization, and by limited dilution.

Resulting cultures of the primary cells bmLECs had doubling times of approximately 24 hours

(Figure 2.2.B), consistent with doubling times reported by Leak and Jones [130]. These

bmLECs were robust and could be frozen and thawed past 20 passages.

Previously, Leak and Jones [130] described the spontaneous formation of lymphatic

tube structures in vitro from confluent monolayer of LECs. I also observed the formation of

these tubes in 96-well plates of bmLEC cell populations left at confluency for at least a few

weeks (Figures 2.1.G and 2.1.H). Cells at the ends of the tubes attached to and migrated up the

sidewalls of the tissue culture wells (Figure 2.1.G). The formation of lymphatic tubes in culture

indicated that extracted bmLECs maintain lymphatic phenotypic character in spite of having

been removed from the in vivo environment.

Page 45: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

31

Figure 2.1. Isolation of bmLECs. A/ Evan‘s blue dye, when injected into the lymph node of

bovine mesentery (***), outlined lymphatic vessels (**), but not blood vessels (*). B/ A mixed

cell population did not form a monolayer in culture. C/ A mixed cell population of

predominantly LMCs and fibroblasts formed circular patterns in culture. D/ Typical

morphology of a cell population predominantly comprising of fibroblasts. E/ Morphology of a

cell population predominantly comprising of LMCs. F/ Morphology of a cell population

predominantly comprising of LECs. G/ Mixed cultures of cells extracted from lymphatic

vessels spontaneously form lymphatic tube-like structures with ends attached to the walls of the

tissue culture dish. H/ Enlargement of the lymphatic tube-like structure seen in G/.

Page 46: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

32

Page 47: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

33

2.3.2. Comparing bmLECs to bmVECs, and to bmAECs.

In essence, the strategy used to isolate bmLECs was also used to isolate bmAECs and

bmVECs. Dye injection was unnecessary since large mesenteric arteries and veins were

discernable from surrounding fatty tissue, unlike milky white lymphatic vessels. Arteries were

thick-walled vessels that were easily distinguished from slightly larger and thin-walled veins.

Cultured bmLECs, bmVECs, and bmAECs exhibited the same cobble stoned monolayer

morphology at confluency (Figure 2.2.A), although the packing of cells together was different.

After the formation of the confluent monolayer, bmAECs, bmVECs, and bmLECs continued to

grow at different rates to form tighter-packed monolayer (Figure 2.2.B). BmAECs grew fastest

at this stage and formed the most dense monolayer. BmAECs were followed by bmVECs,

which formed the second most dense monolayer. BmLECs grew at the slowest rate at this stage

and formed the least dense monolayer (Figure 2.2.B).

Staining the cells with fluorescent phalloidin revealed that the arrangement of actin

microfilaments in the three cell types appeared similar, typically with dense peripheral bands of

actin filaments, and prominent stress fibers throughout the cytoplasm (Figure 2.2.A). However

there did seem to be some qualitative differences in the intracellular distribution of these actin

filaments. There were extensive dense peripheral bands of actin, with relatively few stress

fibers in bmVECs, whereas stress fibers were more prominent in bmAECs. In contrast,

bmLECs seemed to have an intermediate combination of both actin filament arrangements

(Figure 2.2.A).

Upon achieving confluency, apart from differences in cell packing, bmLECs were

indistinguishable from bmVECs and bmAECs by phase contrast microscopy alone. However,

reverse transcriptase-PCR analysis, and immunofluorescence with antibodies specific for Prox-

Page 48: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

34

1, LYVE-1, or Podoplanin and immunofluorescence confirmed that the EC preparations

retained their lineage-specific properties in culture. All three cell preparations expressed Tie-1

and Tie-2 whereas bmLECs but not blood ECs expressed VEGFR-3/Flt-4 (Flt-4) and Prox-1

(Figure 2.3.A). Immunofluorescence also showed bmLECs to express LYVE-1 and Podoplanin

(Figure 2.3.B), both of which have been shown to be highly expressed in LECs but not in BECs

[132]. Furthermore, bmAECs and not bmVECs expressed neuropilin-1, which has been shown

to be highly expressed in AECs [133] (Figure 2.3.A). Although each of these protein markers

for LECs may not exclusively identify LECs per se, however, the co-detection of these markers

together reliably identifies the cultured cells as of lymphatic endothelium origin.

Page 49: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

35

Figure 2.2. Similarities and differences between bmLECs, bmVECs and bmAECs. A/

BmLECs cannot be distinguished from bmVECs or bmAECs by cellular morphology alone.

Actin staining of the three cells using Rhodamine-phalloidin showed actin filaments arranged in

dense bands around the cell periphery and actin stress fibers extending throughout the

cytoplasm. B/ ECs of each type (bmLEC, bmVEC, and bmAEC) were seeded in equal

numbers (approximately 30-40% confluency) and counted every 24 hours. Trypan blue

exclusion was used to determine cell viability. Numbers from 3 independent counts were used

to compile the figure.

Page 50: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

36

Page 51: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

37

Figure 2.3. BmLECs, bmVECs and bmAECs express cell-lineage-specific markers. A/

Despite being similar in morphology, RT-PCR analysis showed bmLECs and not bmVECs or

bmAECs expressed Prox-1 and VEGR-3/Flt-4; bmAECs and not bmVECs expressed

neuropilin-1. All cells expressed Tie-2 and Tie-1. B/ As shown by immunofluorescence,

bmLECs stained positive for podoplanin, Prox-1, and LYVE-1.

Page 52: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

38

Page 53: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

39

2.3.3. Activation of Tie-2 in bmECs by Ang-2

In order to ensure Tie-2 in bovine cells is sufficiently similar to human Tie-2 to be

stimulated with human angiopoietin ligands, amino acid sequences of bovine, mouse, and

human Tie-2 were aligned with CLUSTALW (Figure 2.4). Protein sequence alignment of

human, mouse and bovine Tie-2 indicated 95% identity between bovine and human sequences

and 92% identity between mouse and human, or mouse and bovine sequences. More

importantly, the residues found by Barton and colleagues [134] to be important for the

interaction of Tie-2 with Ang-2 were conserved across the three species (Figure 2.4, blue

boxes), and all tyrosine residues in the kinase domain were perfectly conserved (Figure 2.4).

Based on this observation, bovine ECs should be highly suitable for stimulation with human

ligands.

The Tie-2 receptor protein levels were greatest in bmAECs, whereas the levels in

bmVECs and bmLECs were lower (Figure 2.5.A, anti-Tie-2 (33.1)). Once the differences in

expression levels of Tie-2 in each cell were accounted for, activation of Tie-2 as judged by its

autophosphorylation by Ang-2, relative to basal autophosphorylation level (mock treatment),

was highest in bmLECs, and was considerably lower in bmAECs and bmVECs (Figure 2.5.A

and 2.5.B). Clustered Ang-1, which was previously demonstrated in our laboratory to be the

most potent form of Ang-1 [135], stimulated the activation of Tie-2 in all three cell types above

basal levels (Figure 2.5.A and 2.5.B). Of note, the level of phosphorylation of Tie-2 due to

Ang-1 stimulation as compared to the level due to Ang-2 stimulation was the same in bmLECs

but different in bmAECs and bmVECs (Figure 2.5.A and 2.5.B)

Studies of HUVEC and capillary endothelial cell systems have shown that Ang-1 and

Ang-2 stimulation resulted in the activation of extracellular signal-regulated kinase or Erk1/2

Page 54: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

40

[125, 136-138]. In my endothelial cell system, whereas the endogenous protein levels of Erk1/2

were not different in bmLECs, bmVECs and bmAECs, active phosphorylated Erk 1/2

(phospho-Erk) levels were quite different. Mock-treated bmVECs contained high amounts of

phospho-Erk, whose levels increased slightly upon Ang-1 stimulation (Figure 2.5.A and 2.5.C).

Ang-2 stimulation of bmVECs did not activate Erk1/2 according to phosphorylation levels of

the protein (Figure 2.5.A and 2.5.C). Although bmAECs had relative low basal levels of

phospho-Erk in the mock-treated sample, these cells responded in a similar fashion as bmVECs

to Ang-1 stimulation. Phospho-Erk levels were increased above basal levels upon Ang-1

stimulation, but not upon Ang-2 stimulation in bmAECs (Figure 2.5.A and 2.5.C). In contrast

to bmAECs and bmVECs, stimulation of bmLECs with either Ang-1 or Ang-2 resulted in an

increase in phospho-Erk levels, although Ang-1 seemed to provide a more potent signal (Figure

2.5.A and 2.5.C).

In order to ascertain whether the autophosphorylation of Tie-2 detected in these

immunoblots reflected ligand-dependent activation of Tie-2 and not just phosphorylation by

another kinase, I immunoblotted membranes from bmLECs stimulated with Ang-2 with an

activation-specific antibody directed to tyrosine 992. Phosphorylation of tyrosine 992 has been

shown to reflect activation of kinase activity in Tie-2 [139]. Tie-2 was phosphorylated at this

residue when bmLECs were stimulated with Ang-2 (Figure 2.5.D), indicating that the kinase

activity of the tyrosine receptor was activated.

Page 55: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

41

Figure 2.4. Alignment of bovine, murine, and human Tie-2 amino acid sequences. Amino

acid sequences were obtained from Uniprot/Swiss-prot and aligned with CLUSTALW [140]:

Human Q02763, Bovine Q06807, Mouse Q02858. Boxed in blue are residues found to be

important for Ang-2 interaction with Tie-2 [18], all of which are conserved between the three

species. The transmembrane domain is indicated by the black bar. All tyrosine residues in the

cytoplasmic domain of Tie-2 are well conserved between the three species.

Page 56: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

42

Page 57: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

43

Figure 2.5. Stimulation of Tie-2 phosphorylation in bmECs by Ang-1 and Ang-2. A/

Stimulation of bmLECs, bmVECs, and bmAECs with 800 ng/mL Ang-1 (clustered with anti-

His antibody), or 800 ng/mL Ang-2, or with clustering antibody alone (Mock). Ang-1

stimulation resulted in activation of Tie-2 in bmLECs and to a lesser extent bmAECs and

bmVECs. B/ Phosphorylated Tie-2 signals from blots of 3 independent experiments were

converted to pixel density, quantified using ImageQuant version 5.0, and normalized to

corresponding total Tie-2 levels. Paired Ang-1- and Ang-2- stimulated, normalized Tie-2

phosphorylation levels in each experiment were compared by t-test and p-values for 95%

confidence interval (CI) are shown. Both Ang-1 and Ang-2 stimulated Tie-2 to comparable

levels in bmLECs, whereas Ang-1 was more effective in stimulating Tie-2 in bmVECs and

bmAECs. C/ Phosphorylated Erk1/2 signals from blots of the same 3 independent experiments

from B/ were converted to pixel density, quantified using ImageQuant version 5.0, and

normalized to corresponding total Erk1/2 levels. Paired Ang-1- and Ang-2-stimulated,

normalized Erk1/2 phosphorylation levels in each experiment were compared by t-test and p-

values for 95% CI are shown. Ang-1 was more effective in stimulating Erk1/2 phosphorylation

in all three cell types than Ang-2. D/ Stimulation of bmLECs with 800 ng/mL Ang-2 resulted

in phosphorylation of tyrosine residue 992 on Tie-2.

Page 58: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

44

Page 59: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

45

2.3.4. BmLEC proliferation is enhanced by Ang-1 and Ang-2

A number of studies have shown that Ang-1 stimulation results in the activation of

Erk1/2 [125, 136-138]. More importantly, Ang-1 has been shown to stimulate proliferation of

ECs [137]. Endothelial proliferation in response to growth factors is considered a key

angiogenic and lymphangiogenic response. Therefore, I used tritiated (3H) thymidine uptake

and viable cell counting to determine whether Ang-1 and Ang-2 had a proliferative effect on

bmLECs, and to compare the response of all three cell types to these factors (Figure 2.6).

BmVECs did not respond to either Ang-1 or Ang-2, whereas bmAECs responded best to Ang-1

stimulation (Figure 2.6.A). In contrast, bmLECs responded to both Ang-1 and Ang-2 but best

to Ang-2-stimulation (Figure 2.6.A). These results suggest that the endothelial cell type

influences proliferative responses to the either Ang-1 or Ang-2.

In order to ensure that Ang-1 and Ang-2 stimulation did not simply increase thymidine

uptake in bmECs independently of cell cycle progression and cellular proliferation, I performed

cell count experiments (Figure 2.6.B, 2.6.C, 2.6.D). Consistent with the trends shown by

thymidine uptake assays, Ang-1 and Ang-2 both provided proliferative signals to bmLECs,

increasing bmLEC numbers over the 96-hour period of the assay (Figure 2.6.B). Furthermore,

consistent with thymidine uptake results, Ang-2 was marginally more effective than Ang-1 in

increasing cell counts of bmLECs, although the result was not statistically significant (Figure

2.6.B). However, Ang-1 and Ang-2 increased bmLEC cell counts significantly above mock

levels indicating that both Ang-1 and Ang-2 were proliferation stimulators of bmLECs (Figure

2.6.B).

Consistent with thymidine uptake results, neither Ang-1 nor Ang-2 provided

proliferative signals to bmVECs (Figure 2.6.C). Neither ligand increased cell counts of

Page 60: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

46

bmVECs above mock levels over the 96-hour period (Figure 2.6.C). In contrast and consistent

with the trends shown by the thymidine uptake results, Ang-1 provided a small proliferative

signal to bmAECs, whereas Ang-2 did not increase bmAEC cell counts above mock levels over

the 96-hour period (Figure 2.6.D). Taken together, these cell count results again indicate that

whether Ang-1 and Ang-2 are mitogenic depends on the endothelial cell type.

2.3.5. Ang-1 promotes bmEC migration

EC cell migration is another well-accepted criterion for the angiogenic response. Ang-1

has previously been shown to induce cell migration, whereas Ang-2 has previously been shown

not to drive the migration of HUVECs [141]. Using a modified Boyden chamber assay I tested

the ability of Ang-1 and Ang-2 to drive migration of these three cell types. I found that Ang-2

did not promote migration of these cells whereas Ang-1 was able to drive bmEC migration

(Figure 2.7). These results suggest that although bmLECs, bmVECs, and bmAECs responded

differentially to Ang-1 and Ang-2 in proliferative assays, they responded similarly to the

chemotactic effects of Ang-1.

2.3.6. Ang-2 protects bmLEC from cell death

Many angiogenic factors are known to protect ECs from serum-deprivation-induced

cellular apoptosis [142]. Ang-1 has previously been shown by numerous research groups

including the Dumont group to protect HUVEC from apoptosis [143-150]. However, a role for

Ang-2-mediated EC survival remains unclear. Results of some studies suggested that Ang-2

counteracts the anti-apoptotic effects of Ang-1 and leads to cell death [44, 151-153]. Results of

other studies suggested that Ang-2 mediates a PKB-dependent cell survival signal [122, 125,

154]. To investigate the role of Ang-2 in serum-deprivation-induced cell death, I tested the

ability of Ang-1 or Ang-2 to promote cell survival on the three cell preparations. Ang-1

Page 61: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

47

protected bmVECs and bmAECs from cell death, whereas it had minimal effect on bmLECs

(Figure 2.8). In contrast Ang-2 had virtually no effect on bmVECs and a marginal effect on

bmAECs (Figure 2.8). Interestingly, Ang-2 produced a survival response in bmLECs (Figure

2.8). Together with results from survival and proliferation assays (Summarized in Table 2.1),

these results further suggest that the origin of ECs dramatically influences their response to

either Ang-1 or Ang-2.

Page 62: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

48

Figure 2.6. Proliferation of Ang-1 and Ang-2 stimulated bmECs. A/ BmLECs, bmAECs and

bmVECs were tested for proliferative response in the presence of Ang-1 and Ang-2 via tritiated

thymidine uptake. Clustered Ang-1 (800ng/mL) and Ang-2 (800ng/mL) did not increase

thymidine uptake in bmVECs. Ang-1 induced increased thymidine uptake in bmAECs and

bmLECs above mock levels (clustering anti-His antibody alone). Ang-2-treated bmLECs

showed higher levels of thymidine uptake than did Ang-1-treated cells. Results from 2

independent experiments were compiled for the figure. ANOVA p-values for 95% CI are

indicated with the colours of the bars corresponding to the colours used to represent cell type.

BmLECs (B), bmVEC (C), and bmAEC (D) were tested for proliferative response in the

presense of clustered Ang-1 (800 ng/mL) and unclustered Ang-2 (800 ng/mL) by cell counting.

400 000 cells were seeded (hour 0, approximately 10-15% confluency) and monitored every 24

hours. Trypan blue exclusion was used to determine cell viability. Results are compiled from

one representative experiment of 2 independent experiments, each done in tripplicate. ANOVA

p-values for 95% CI for cell counts at the 96 hour time points are as follows: B/ BmLEC p=9E-

10 (*); C/ BmVEC p=0.22 (**); D/ BmAEC p=6E-7 (***).

Page 63: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

49

Page 64: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

50

Figure 2.7. Migration of Ang-1 and Ang-2 stimulated bmECs. BmECs were tested for

migration in a modified Boyden chamber assay in the presence of Ang-1 and Ang-2. Migration

of bmLECs, bmVECs, and bmAECs towards clustered Ang-1-containing media was

significantly increased compared to towards control media (Mock) containing only 1% FBS and

clustering anti-His antibody. In contrast, Ang-2 did not significantly stimulate migration in any

of the cell types, compared to control media. Migration was measured using the Boyden

Chamber. Results from 2 independent experiments were compiled for the figure. ANOVA p-

values for 95% CI are indicated.

Page 65: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

51

Page 66: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

52

Figure 2.8. Survival of Ang-1 and Ang-2 stimulated bmECs. Death in bmECs was induced

by serum starvation and relative amounts of cell death compared to 0.1%FBS control were

measured by quantification of cytoplasmic histone-associated-DNA-fragments (mono- and

oligonucleosomes) via ELISA (Roche). Ang-2 provided marginally better protection from

serum-deprivation-induced cell death than did Ang-1 for bmLECs. Ang-2 provided marginally

worse protection from serum-deprivation-induced cell death than did Ang-1 for bmVECs and

bmAECs. Results of 3 independent experiments were compiled for this figure. P-values of

unpaired, two-tailed t-test for 95% CI are indicated.

Page 67: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

53

Page 68: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

54

Table 2.1. Summary of results. Table depicts relative levels of Tie-2 and Erk1/2 activation

and relative levels of proliferation, survival in serum-deprivation conditions, and migration of

Ang-1 and Ang-2 stimulated bmECs.

Page 69: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

55

Page 70: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

56

2.4. Discussion

In this study, I set out to examine whether the type of EC could affect its response to

Ang-1 or Ang-2. I prepared ECs from lymphatic, venous and arterial vessels taken from bovine

mesentery and subjected them to angiogenesis-related in vitro assays. My results demonstrate

that the activation of Tie-2 by Ang-2 stimulation was as effective as by Ang-1 stimulation in

bmLECs. In bmVECs and bmAECs, Ang-1 stimulation was more effective than Ang-2 in

activating Tie-2. The activation of Tie-2 in all three cell types by Ang-1 or in bmLECs by Ang-

2 correlated with the activation of Erk1/2. Ang-1 activated Erk1/2 in all three cell types,

whereas Ang-2 only activated Erk1/2 in bmLECs. Of note, Ang-1 seemed to be a more potent

activator of Erk1/2 than Ang-2 in bmLECs. These results suggest Erk1/2 is a downstream

signaling pathway of Tie2 in ECs. Although Ang-1 stimulation correlated with Erk1/2

activation in all three cells, Ang-1 did not promote proliferation in all three cell types but only

weakly in bmAECs and bmLECs (Table 2.1). Unlike bmVECs, both bmAECs and bmLECs

had low levels of basal phosphorylated Erk1/2 levels in the mock samples. This finding

suggests that the proliferative response of bmLECs to Ang-2 stimulation is likely not solely

under the regulation of Erk1/2 downstream of Tie-2. Complex cell-type specific mechanisms

are likely present in the ECs and are responsible for suppressing the proliferative signal from

Ang-1 in bmVECs, while promoting the weak proliferative responses in bmLEC and bmAECs.

Likewise, additional cell-specific mechanisms may be present in BECs that suppress the

proliferative and survival signals from Ang-2, but are present in LECs and are responsible for

promoting the proliferative and survival signals from Ang-2. This was implied by my findings

that Ang-2 was not a proliferative signal for bmAECs or bmVECs but only for bmLECs.

Similarly, Ang-2 did not support serum-free survival of bmVECs and bmAECs but promoted

Page 71: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

57

the survival of bmLECs (Table 2.1). Of note, the level of Erk1/2 activation stimulated by Ang-

2 in bmLECs was lower than the level stimulated by Ang-1, which did not provide as potent a

proliferative or survival signal as Ang-2 in bmLECs.

In summary, my data shows that ECs of different origin have different sets of cellular

responses to Ang-1 and Ang-2 stimulation, suggesting that Ang-1 and Ang-2 activate distinct

sets of signaling molecules in the signaling cascades of ECs of different origin. Thus, Ang-2

may be a full agonist in ECs of lymphatic origin, but a partial agonist in ECs of blood origin.

Since Ang-1 and Ang-2 also elicited slightly different responses from bmAECs and bmVECs in

my study (Table 2.1), the subtype of ECs of blood origin need to be taken into consideration in

future studies of Ang-1 and Ang-2 signaling and function.

Until recently, Ang-2 was thought to be an Ang-1 competitive antagonist. This

conclusion was made based on the finding that transgenic over-expression of Ang-2 produced

blood vasculature defects resembling those of Ang-1 or Tie-2 knockout mutants [43]. Ang-2

competitively blocked activation of Tie-2 by Ang-1 but Ang-2 was equally as effective as Ang-

1 in activating Tie-2 phosphorylation in NIH 3T3 fibroblasts ectopically expressing Tie-2 [43].

This finding suggested that ECs have additional components that allow functional

discrimination between the two angiopoietin [43]. My findings clearly indicates that the

additional components suggested by these initial studies of Ang-2 are endothelial cell-type

specific and may be responsible for the agonistic activities of Ang-2 in LECs. Even though

there are some functional redundancies, each angiopoietin may be driving a distinct set of

cellular events within a specific type of endothelial cell.

An important implication of the results described in this chapter is that the design of

therapies aimed at enhancing or mitigating angiogenesis and lymphangiogenesis need to include

Page 72: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

58

considerations of biological differences between vascular systems that share molecular

regulators. The angiopoietins may be only one example of many other molecular regulators

that behave differently in the two vascular systems. Cell systems such as the one I have built

and describe in this chapter make it possible for simultaneous characterization of the signaling

pathways downstream of these shared molecular regulators in each of the three EC types:

venous, arterial and lymphatic. Such a cell system as this combined with high throughput mass

spectrometry-based methods currently in refinement produces a potentially very powerful tool

to indentify and characterize new lymphatic and blood vascular markers and phenotypic

regulators.

Page 73: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

59

CHAPTER 3

DIFFERENTIAL PROTEOMIC ANALYSIS OF LYMPHATIC,

VENOUS, AND ARTERIAL ENDOTHELIAL CELLS

EXTRACTED FROM BOVINE MESENTERIC VESSELS

Page 74: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

60

3.1. Introduction

Proteins perform functions essential for most cellular processes unique to lymphatic and

blood endothelial cell types. Protein profiling is therefore an important tool in characterizing in

vitro EC models used to understand cell-type specific cellular processes at the molecular level.

As early as 1984, when research endeavours to understand EC molecular biology behind

angiogenesis and lymphangiogenesis was still in its infancy, three separate groups were the first

to independently establish in vitro cultures of LECs isolated from bovine mesentery [131],

collecting ducts [155], and cavernous lymphangiomas [156] (as reviewed in [157]). Beside

similarities to BECs cultures such as monolayer cobblestone morphology and presence of

Weibel-Palade bodies, the researchers noted unique features displayed by LECs in culture

including overlapping intercellular junctions and anchoring filaments or microfibrils connecting

abluminal cell membranes of capillary LECs to the surrounding connective tissue [157, 158]

The capacity to isolate and establish stable lines of primary, immortalized, or

transformed LECs in culture allowed lymphatic researchers to compare and contrast LECs with

BECs in terms of morphology, physiology, and function. More recently, the advent of

technologies to study entire cell transcriptomes and proteomes have allowed lymphatic

researchers to characterize in vitro LEC cultures at the molecular level and perform

comparative studies with BECs similarly isolated [36, 107, 159-166]. The collective effort of

these and other researchers has yielded a reliable list of observed differences and similarities

between LECs and BECs.

I described in chapter 2 some of these differences in primary cultures of LECs, VECs,

and AECs isolated and cultured from vessels of bovine mesentery [167] by the method of

Johnston and Walker [131]. In culture, early passaged cells retained distinct molecular

Page 75: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

61

signatures; bmLECs expressed well-known lymphatic markers such as Prox-1, Podoplanin, and

VEGF-R3, which were absent in bmVEC and bmAECs [167]. Other differences between the

three cell types that I observed include lower packing density of cells and relatively higher

responsiveness to stimulation by Angiopoietin-2 (Ang-2) in bmLECS as compared to the blood

ECs with bmVECs being the least responsive [167]. (Ang-2 has previously been shown to be

required for lymphangiogenesis during embryonic development [43, 168].)

In the study described in this chapter, I set out to characterize primary cultures of

bmLECs, bmVECs, and bmAECs described above by comparative analysis of protein profiles

resolved by 2-dimensional SDS polyacrylamide gel electrophoresis (2D-PAGE). I

hypothesized that potential molecular players that may be responsible for the observable

differences between the three ECs described in my previous work are identifiable by rigorous

quantification of silver-stained 2D-PAGE followed by mass spectrometric identification of

proteins showing statistically significant differences in levels between the three cell types. The

main rationale for attempting to identify proteins potentially responsible for phenotypic

differences I observed in these three cells, is to establish that my three-cell model is potentially

useful for studies aimed at developing drugs targeting disease states of the lymphatic

vasculature, without adversely affecting blood vessels. A highly relevant example of such a

disease state is intestinal lymphangiectasia. This condition has complex etiology generally

involving obstructed lymph flow in lymphatic vessels of the gastrointestinal tract. A three-cell

system such as the one I have built from vessels of the mesentery would be useful for in vitro

cell-based assays to understand similarities and differences between these three cells types and

how they respond to stimuli.

Page 76: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

62

To the best of my knowledge, the comparative study reported in this chapter is one of a

very small number of studies that compare ECs from all three representative vessel types,

lymphatic, venous, and arterial, since the early 1990s. Weber and colleagues [169] assayed for

presence of 5‘-nucleotidase, alkaline phosphatase, adenylate and guanylate cyclase in cultured

ECs from lymphatic, venous, and arterial vessels from bovine mesentery isolated by the method

of Johnston and Walker [131]. Although I used the same cell system as Weber and colleagues,

I aimed to identify new differentially expressed proteins in the three cell types. Veikkola and

colleagues [170] compared responses of lymphatic, venous, and arterial ECs isolated from

human umbilical cord, aorta, skin, and saphenous vein to VEGFs. Ando and colleagues [171]

compared levels of cell adherence molecules in mouse mesenteric LECs to aortic AECs, and

VECs from vena cava. Bianchi and colleagues [161] compared 2D-PAGE-resolved protein

profiles of bovine thoracic aortic AECs, inferior vena cava VECs, and thoracic duct LECs.

Unlike these three studies, I used ECs cultured from smaller vessels of bovine mesentery.

Three 2D-PAGE electrophoretograms for each of the three mesenteric cell types were

produced and quantitatively analysed. Protein identification by liquid chromatography tandem

mass spectrometry (LC-MS/MS) was performed to identify 39 proteins excised from 2D gels

found to be present at statistically significantly different levels (>1.5 fold) in the three cell types

(p<0.05). Additionally, I was able to validate by immunoblotting (IB) three proteins, HSPA1B

(a member of the HSP70 family), HSPB1 (a member of the HSP27 family), and UBE2D3 (a

member of E2 ubiquitin-conjugating enzymes) found to be at highest levels in bmAECs,

bmVECs, and bmLECs. The lack of substantial overlap between my results, those of Bianchi

and colleagues [161], and those of other groups‘ comparative studies are fully discussed.

Page 77: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

63

Functional implications of differences in levels of various proteins identified in the three cell

types are also discussed.

3.2. Methods

3.2.1. Cell isolation and culture

Bovine ECs from veins, arteries, and lymphatic vessels of the mesentery were isolated

and cultured as previously described [167] by the method of Johnston and Walker [131].

Bovine aortic arterial endothelial cells (baAECs) were isolated by a similar method. Cells were

briefly dislodged by dispase and collagenase from bovine aortic vessels. All EC types were

cultured in 10% fetal bovine serum in standard tissue culture conditions: 5% CO2, atmospheric

O2, and 37°C. Cells were at passage 20 at the time of harvest. Cells were harvested at

confluency and lysed in solubilisation buffer containing 8M urea, detergents, 2% immobilized

3-10 pH gradient buffer (IPG buffer, Amersham Pharmacia Biotech, Uppsala, Sweden).

Protein concentrations were quantified and equal protein amounts were subjected to 2D-PAGE

separation.

3.2.2. 2D-PAGE

Isoelectric focusing was performed on ReadyStrip IPG 11 cm strips (Biorad

Laboratories Hercules, California) with a linear range of pH 3-10 (using the same method as

described in http://proteome.tmig.or.jp/2D/2DE_method.shtml). IPG strips were first

rehydrated under passive conditions for 12 hours on the PROTEAN IEF cell (Biorad

Laboratories) followed by focusing at step-wise increases of voltage for a total of 46 700

Page 78: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

64

voltage-hour with a current limit of 50 μA. IPG strips were equilibrated as directed by

manufacturer. The second dimension separation was carried out on 12% acrylamide gels.

2D-PAGE gels were silver stained and digitally recorded using the ProExpress gel

imaging system (PerkinElmer, Waltham, Massachusetts). Quantitative comparisons of spot

intensities were analysed using Progenesis Samespots (Nonlinear Dynamics, Durham, North

Carolina). Background subtraction and spot intensity normalization were automatically

performed by Progenesis Samespots. Relative spot intensity values were calculated on a

logarithmic scale and manually converted to relative fold-difference levels. Statistical analysis

was also automatically performed by Progenesis Samespots on intensity values of protein spots

from three separate gels for each of the cell types (total of nine gels).

3.2.3. Mass Spectrometric Analysis of Protein Spot Intensities

Excised protein spots of interest were trypsin digested, extracted, and analysed by LC-

MS/MS (1100 nanoflow LC system, XCT plus ion trap, Agilent Technologies, Santa Clara, CA,

USA). Spectrum Mill proteomics workbench (Agilent) was used to match MS/MS spectra to

non-redundant bovine protein sequences NCBInr (released in 2005). Peptide scores >10,

protein scores >11, SPI values >70%, were deemed acceptable since Spectrum Mill workbench

is not based on statistical methods.

3.2.4. Validation of Protein Level Differences by Immunoblotting

Cells were harvested at 90% confluency and lysed in RIPA buffer (150mM NaCl, 1%

Igepal, 0.5% Sodium Deoxycholate, 0.1% SDS, 50mM Tris pH 7.4, 1mM EDTA, protease

inhibitor tablets (Roche)). Lysates were cleared and quantified before proteins are separated by

SDS-PAGE. To detect the 17kDa protein UBE2D3 by immunoblotting, 4%-20% gradient gel

(Thermo) was used along with 0.2 μM PVDF transfer membrane (Sigma-Aldrich). All other

Page 79: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

65

proteins were separated by 10% gel and 0.45 μM PVDF transfer membrane. Mouse

monoclonal antibodies against UBE2D3 (M01, clone 4C1), ANXA2, polyclonal mouse

antibodies against HSPA1B (HSP70-2, HSPA1A, whole antisera), and polyclonal rabbit

antibodies against HSPB1 and HSPB2 were all from Abnova. Anti-beta-actin mouse

monoclonal antibodies were from Sigma-Aldrich (Clone AC-15, mouse ascites fluid). Anti-

Tie-2 mouse monoclonal antibodies were from BD Biosciences Pharmingen. Autoradiograms

of chemiluminescent signal from immunoblots were quantified using ImageQuant 5.0. Pixel

densities were converted to relative values with lymphatic signal intensities taken as 1.

Statistical analysis reported for figures 3.4 and 3.5 was performed on averaged relative values

from at least 4 immunoblots for each of the three proteins validated.

3.3. Results

3.3.1. Quantitative Analysis of 2D-PAGE-Resolved bmEC Proteins

Three representative 2D-PAGE electrophoretograms for three EC types are shown in

figure 3.1.A. Spots chosen for MS/MS identification are indicated in the electrophoretogram

for bmLECs (Figure 3.1.A). Protein spots from all nine electrophoretograms (three per cell

type) were automatically matched by vector functions performed by Progenesis Samespots

(Figure 3.2.B). Matches were then manually checked to ensure correct assignment of vectors.

Quantitative analysis of protein level differences between bmLECs, bmVECs, and

bmAECs is demonstrated in Figure 3.2 with representative spot 32 (Figure 3.2.A). Progenesis

Samespots generates a three dimensional rendition of the imaged spot for better visualization

(Figure 3.2.B). Each data point in Figure 3.2.C represents each of the same spot number 32 in

each of the nine gels. Each set of three normalized relative volumes shown belonged to one of

Page 80: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

66

the three indicated cell types (Figure 3.2.C). Averaged log normalized volumes automatically

calculated by Progenesis Samespots for representative spot 32 for each of the three cell types

are shown in Figure 3.2.D. P-values from ANOVA and power values associated with averaged

log normalized volumes are listed along with spot numbers in Table 3.1.A.

Page 81: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

67

Figure 3.1. 2D-PAGE separation of bmEC proteins. A) Representative 2D silver stained

electrophoretograms of bmLECs, bmVECs, and bmAECs, generated using IPG strips pH 3-10.

Spot numbers correspond to protein identities listed in Table 4.1. Approximate molecular

weights and isoelectric points are indicated. B) Demonstration of spot matching function in

Progenesis Samespots. Automatic matching was manually checked to ensure accuracy.

Page 82: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

68

Page 83: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

69

Figure 3.2. Quantitative Analysis of 2D-PAGE-Resolved bmEC Proteins. Quantitative

analysis of protein level differences between bmLECs, bmVECs, and bmAECs. A) Enlarged

silver stained image of one representative spot numbered 32 is shown. B) Three dimensional

rendition of imaged spot by Progenesis Samespots. C) Normalized relative volumes of the

same spot from nine gels (three per cell type). D) Averaged log normalized volumes

determined for representative spot 32 for each of the three cell types.

Page 84: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

70

Page 85: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

71

3.3.2. Mass Spectrometric Analysis of 2D-PAGE-Resolved bmEC Proteins

Also summarized in Table 3.1.A are protein identities determined by LC-MS/MS and

database search with Spectrum Mill and Bovine NCBI non redundant database [172] along with

distinct summed MS/MS search scores, number of unique peptides that were matched, and

percent amino acid sequence coverage by the matched unique peptides. Proteins identified are

relisted in Table 3.1.B and are organized according to their subcellular localization, Gene

Ontology (GO) biological process and molecular function categories [173] as summarized by

GeneCards [174]. The distribution of these proteins in biological processes listed in Table

3.1.B are shown graphically in Figure 3.3. An overwhelming majority of listed proteins have

protein processing, chaperone, turnover, synthesis, and protein binding in skeletal structural

support functions.

Proteins found to be present at statistically significantly different levels in each of three

cell types included two peroxiredoxin proteins: PRDX2 and PRDX6, two ubiquitin conjugating

enzymes: UBE2N and UBE2D3, and two heat shock proteins: HSPB1 (HSP27) and HSPA1A

(HSP70) (Table 1A). PRDX6 protein level was higher in both bmLECs (close to 2-fold above

bmAECs) and bmVECs (1.7-fold above bmAECs). PRDX2 protein level was markedly higher

in bmLECs than in the blood ECs, 1.8-fold above the level in bmVECs and 2.5-fold above that

in bmAECs. UBE2N and UBE2D3 protein levels were higher in bmLECs than in the blood

ECs, 1.7- and 2.2-fold above levels in bmVECs, 2.5- and 3.8-fold above those in bmAECs,

respectively. HSPB1 protein level was highest in bmVECs, over 3.5- and over 5.7-fold above

the level in bmLECs and in bmAECs, respectively. HSPA1A protein level was highest in

bmAECs, over 30- and 12-fold over bmLECs and bmVECs, respectively. Like other proteins

on the list, I generally found a gradient of differences in protein levels between the three cells.

Page 86: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

72

Limits on sensitivity of the method used probably prevented the detection of known and new

protein markers that distinguish a particular EC type.

Page 87: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

73

Table 3.1.A. Summary of proteins identified. Proteins separated by 2D-PAGE found to be

statistically different in endothelial cells isolated from bovine mesenteric lymphatic, venous,

and arterial vessels (bmLEC, bmVEC, and bmAEC) identified by mass spectrometry. Proteins

are organized by relative fold differences in the three bmECs. At the top of the table are

proteins most up-regulated in bmAECs, proteins most up-regulated in bmVECs are in middle of

the table, and at the bottom of the table are proteins most up-regulated in bmLECs. Distinct

summed MS/MS search score, number of distinct peptides identified, and percent of amino acid

covered by distinct peptides provided by Spectrum Mill workflow are listed. Theoretical and

experimentally observed molecular weight (MW, kilodaltons, kDa) listed differed by at most 12

kDA (HMGCS1). Theoretical and experimentally observed isoelectric points (pI) differed by at

most 0.9 (GOT2). Accession numbers are from NCBI non-redundant database of bovine

sequences [172]. The table has been split into three parts and displayed on pages 74-76.

Page 88: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

74

Page 89: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

75

Page 90: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

76

Page 91: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

77

Table 3.1.B. Summary of biological information on protein identified. Proteins identified in

Table 4.1.A are relisted in this table and organized by subcellular localization, biological

process and GO molecular function (Gene Ontology [173]) as summarized by GeneCards [174].

The table has been split into three parts and displayed on pages 78-80

Page 92: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

78

Page 93: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

79

Page 94: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

80

Page 95: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

81

Figure 3.3. Biological Functions of Proteins Identified in Table 4.1. Percentage

breakdown—according to biological functions—of proteins found to be present at statistically

significantly different levels in the three endothelial cell types.

Page 96: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

82

Page 97: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

83

3.3.3. Validation of Identified Proteins by Immunoblotting

For proof of principle, I selected three proteins, each found to be at highest levels in

each of the three cell types, to validate by immunoblotting followed by pixel density

quantification with ImageQuant. The composite of resulting blots is shown Figure 3.4.A and

relative averaged signal levels in the three cell types from at least four blots for each protein are

shown in Figures 3.4.B, 3.4.C, and 3.4.D. In contrast to results from comparative analysis of

the 2D-PAGE electrophoretograms, HSPA1B was only about 2-fold higher by IB (as compared

to 30-fold) in bmAECs than in bmLECs (Figure 3.4.B). Furthermore, the level of HSPA1B in

bmVECs was not found by IB to be double that in bmLECs as recorded in Table 1A from

analysis of silver-stained 2D-PAGE electrophoretograms (Figure 3.4.B). Likewise, while the

level of HSPB1 was certainly highest in bmVECs, consistent with results from 2D-PAGE

summarized in Table 3.1.A, the magnitude of the difference was not reproduced by IB (over 5-

fold versus 3-fold by IB, Figure 3.4.C). UBE2D3 protein level was almost 4-fold higher in

bmLECs than in bmAECs by 2D-PAGE analysis but only approximately 2.5-fold over

bmAECs by IB (Figure 3.4.D). In all three cases, the pattern of differences was reproduced by

validation experiments but the magnitude of differences in protein levels was not reproduced.

In figures 3.4.B and 3.4.F, I showed the same method of quantification done on Tie-2

and beta-actin IBs. Tie-2 is a well-known receptor tyrosine kinase whose expression is thought

to be restricted to ECs almost exclusively. Tie-2 levels appeared to be slightly higher (1.5-fold)

in bmVECs and bmAECs than bmLECs (Figure 3.4.E). Beta-actin was not significantly

different between the three cell types as expected (Figure 3.4.F). The slightly higher level of

actin in bmLECs and bmVECs (1.2 fold) indicated that bmLEC and bmVEC lysates were made

up consistently slightly more concentrated than bmAEC lysates (Figure 3.4.F). However, this

Page 98: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

84

small difference was not enough to account for all the relative differences in HSPA1B, HSPB1,

and UBE2D3 protein levels in the three cell types (Figures 3.4.A-D).

3.3.4. Comparing Protein Levels of Different Family Members in the same ECs

Figure 3.5, when I used antibodies specific to HSPB2, another member of the HSP27

family of proteins, I found protein levels to be highest in bmAECs instead of bmVECs as was

the case for HSPB1. This result may indicate that family members of the HSP27 family may be

differentially expressed in the three EC types. Different family members may play a unique

role in the biology of each of the three EC types. The same may also apply to the HSP70

family, to which HSPA1B belongs, and the E2 ubiquitin conjugation enzyme family, to which

UBE2D3 belongs.

3.3.5. Comparing Protein Levels in ECs of Dissimilar Anatomical Origin

Comparing my results to those of Bianchi and colleagues [161] I found very little

overlap. Similar to my results, Bianchi and colleagues found ANXA2 (Annexin A2) levels to

be highest in bovine aortic (baAECs), roughly 3 times the level in thoracic duct LECs [161]. I

found ANXA2 levels in bmAECs to be roughly 1.5 times that in bmLECs (Table 3.1.A). In

both my results and those of Bianchi and colleagues [161], the trend was the same in that the

silver stained signal corresponding to ANXA2 identification by mass spectrometry: bmVECs

had the lowest level of the three cell types. Other than ANXA2 similarities, there was little

overlap in both the identities of proteins found differentially up or down regulated and in the

trends of relative differences between the three cell types (Table 3.1.A).

I hypothesized that discrepancies between the two sets of data may be partly due to

differences in anatomical source of bovine lymphatic, venous, and arterial ECs. To test this

hypothesis, I quantified immunoblotted protein signals of ANXA2 in my bmECs as well as in

Page 99: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

85

baAECs similarly isolated and cultured (Figure 3.6). Surprisingly, the trend of ANXA2 being

lowest in endothelial cells of venous origin was not validated by the IBs. ANXA2 was actually

highest in both bmAECs and bmVECs (Figure 3.6). I clearly saw statistically significantly

higher levels of ANXA2 in bmAECs as compared to baAECs (p = 0.0002, 12 blots). This

result indicates that protein level differences are highly dependent on anatomical context.

Page 100: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

86

Figure 3.4. Validation of HSPA1B, HSPB1, and UBE2D3 protein levels in the three cell

types by IB. A) Protein from the three cell types were detected using isoform-specific

antibodies to HSPA1B (70 kDa), HSPB1 (27 kDa), and UBE2D3 (17 kDa). For loading

controls, Tie-2, an endothelial cell maker, and beta-actin were used. Autoradiograms from

chemiluminescent immunoblots were quantified using ImageQuant and averaged relative fold

differences in pixel densities are shown in B-F. B) Results of 5 blots are shown with HSPA1B

protein levels in bmLEC taken to be 1. T-test comparing relative levels of HSPA1B in

bmLECs to those in bmAECs resulted in p = 0.003 C) Results of 5 blots are shown with

HSPB1 protein levels in bmLEC taken to be 1. T-test comparing relative levels of HSPB1 in

bmLECs to those in bmVECs resulted in p = 0.008 D) Results of 5 blots are shown with

UBE2D3 protein levels in bmLEC taken to be 1. T-test comparing relative levels of UBE2D3

in bmLECs to those in bmVECs resulted in p = 0.009 E) and F) results from 4 blots are shown,

t-tests done as above resulted in p values of 0.013 for Tie-2 and 0.2 for beta-actin. (All t-tests

used α = 0.05)

Page 101: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

87

Page 102: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

88

Figure 3.5. Determination of HSPB2 protein levels in the three cell types by IB. A) Protein

from the three cell types were detected using isoform-specific antibodies to HSPB2 (27 kDa).

For loading controls, anti-beta-actin antibodies were used. B) Autoradiograms from

chemiluminescent immunoblots were quantified using Image Quant and averaged relative fold

differences in pixel densities are shown. Results of 5 blots are shown with UBE2D3 protein

levels in bmLEC taken to be 1. T-test (α = 0.05) comparing relative levels of UBE2D3 in

bmLECs to those in bmAECs resulted in p = 0.003.

Page 103: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

89

Page 104: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

90

Figure 3.6. Comparing ANAX2 Protein Levels in ECs of Dissimilar Anatomical Origin.

Determination of Annexin A2 (ANAX2) protein levels in bmLECs, bmVECs, bmAECs, and

bovine aortic endothelial cells (baAECs) by immunoblotting. A) Protein from four cell types

were detected using antibodies to ANAX2 (35 kDa). For loading controls, anti-beta-actin

antibodies were used. B) Autoradiograms from chemiluminescent immunoblots were

quantified using ImageQuant and averaged relative fold differences in pixel densities are

shown. Results of 12 blots are shown with Annexin A2 protein levels in bmLEC taken to be 1.

T-test (α = 0.05) comparing relative levels of Annexin A2 in bmAECs to those in baAECs

resulted in p = 0.0002.

Page 105: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

91

Page 106: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

92

3.4. Discussion

I have generated and analysed silver-stained electrophoretograms of 2D-PAGE-resolved

proteins from bovine mesenteric ECs extracted from venous, arterial, and lymphatic vessels. I

identified 39 proteins that showed variations in the three cell types with ANOVA p-values

<0.05. Most proteins were found at high levels in both bmAECs and bmVECs (49%).

Similarly, most proteins were found to be present at high levels in both bmLECs and bmVECs

(41%). Of the three cell types, protein profiles of bmLECs and bmAECs were the most

divergent; only 10% were found to be high in both of these cell types. This observation is

consistent with findings by Bianchi and colleagues [161] and with theories on lymphatic lineage

specification from venous lymphatic progenitors during embryonic development [175].

My study design is very similar to that of Bianchi and colleagues [161] but I found little

overlap in the resulting list of proteins. For example, Bianchi and colleagues [161] found

PRDX2 levels to be highest in venous ECs and lowest in arterial ECs. In contrast, I found

PRDX2 levels to be highest in bmLECs but were at similar levels in the blood ECs. Similar to

my results (Table 3.1.A), Bianchi and colleagues found silver stained signal of the spot

corresponding to ANXA2 protein identification to be strongest in baAECs compared to LECs

from bovine thoracic duct and VECs from bovine vena cava [161]. In both my results and

those of Bianchi and colleagues, the trend was the same in that the silver stained signal

corresponding to ANXA2 identification by mass spectrometry in VECs was the lowest of the

three cell types. When I attempted to validate ANXA2 levels in ECs by immunoblotting, the

trend shown by silver stained 2D-PAGE was not reproduced. Perhaps contaminants in spot 94

confounded silver staining signals and/or antibodies commercially available may be cross-

reacting with other members of the Annexin family of proteins that are also differentially

Page 107: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

93

regulated between the three cells. Results shown in Figure 5 certainly prove that different

members of the same family of proteins can be differentially regulated in the three cell types.

Overall, the lack of substantial overlap between my results and those of Bianchi and

colleagues [161] may simply be due to differences in anatomical sources of the cultured bovine

ECs used. Bianchi and colleagues [161] isolated VECs from bovine inferior vena cava, AECs

from thoracic aorta, and LECs from thoracic duct. My results shown in Figure 6 indicate that

this may be the case. ANXA2 protein level in bmAECs was statistically significantly higher

than in baAECs. Evidence that same EC types from different anatomical sites have different

gene expression programmes has been reported by Chi and colleagues [159]. Other reasons for

the discrepancy may be at the proteomics and informatics level where it is generally accepted

that search results are somewhat dependent on the instruments used for mass spectrometry, and

highly dependent on the algorithms used for the search [176]. Until consensus is reached on

statistical methods to validate database searching strategies to analyse MS/MS spectra,

independent but similar protein profiling studies may be considered complementary to one

another.

None of the resulting proteins from my analysis of 2D-PAGE resolved profiles can be

considered novel markers of lymphatic, venous, and arterial EC lineages. However, some

trends found in my results are still noteworthy. Consistent with previously reported

observations that ECs in arterial vessels, and not ECs from venous vessels (unless placed under

stress), express HSP70 [177], I found HSP70 to be much higher in abundance in bmAECs as

compared to bmLECs and bmVECs (Table 3.1.A). Also consistent with previously reported

observations that venous ECs express HSPB1 but not HSPA1B under non-stress conditions

[178], I found HSPB1 to be highly abundant in bmVECs as compared to bmAECs and bmLECs

Page 108: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

94

(Table 3.1.A). Both HSPB1 and HSPA1B belong to large families of heat shock proteins

(HSP27 and HSP70) that function as molecular chaperones. HSP27 and HSP70 aid proper

protein folding during normal physiology as well as during cellular crises such as heat and

oxidative stress. HSP27 and HSP70 have both been shown to be cytoprotective and potentially

play protective roles in mouse and rat models of brain and heart ischemia [179].

Although the magnitudes of differences in 2D-PAGE silver-stained signals of the three

proteins HSPA1B, HSPB1, and UBE2D3 in the three cell types were not reproduced in my

validation with isoform-specific antibodies, the pattern of differences was mostly reproduced.

HSPA1B was consistently highest in bmAECs, HSPB1 was consistently highest in bmVECs,

and UBE2D3 was consistently highest in bmLECs. Limits of quantification techniques used

may account for the lack of agreement in magnitude of differences in protein levels between the

two methods for HSPA1B and HSPB1. Another possible explanation for the discrepancy may

be the co-migration of proteins that are similarly differentially present in the three cell types.

Differences in the silver-stained protein spots may be amplified while validation of one protein

out of all the proteins present in the spot shows smaller degrees of differences.

Actual changes in the proteomes of the cells may also be a cause; cells used for

validation have been frozen and thawed and passed for a greater number of times than cells

used in the initial 2D-PAGE analysis. Whatever the cause, what is important is that I was able

to reproduce the larger patterns in protein differences as mentioned above.

To the best of my knowledge, the VEGFR family of proteins whose members, for

example: VEGFR-1 versus VEGFR-3/Flt-4, are differentially expressed in different EC types, I

know of only a few other protein families whose members serve as EC-type markers. I report

here a new family of proteins, HSP27, whose members HSPB1 and HSPB2 are expressed at

Page 109: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

95

relatively highest levels in VECs and in AECs, respectively. This finding further corroborates

the idea that different members of protein families may play unique roles in the biology of each

of the three EC types. This same idea may also apply to the HSP70 family, to which HSPA1B

belongs, and the E2 ubiquitin conjugation enzyme family, to which UBE2D3 belongs. Further

experimentation is required to confirm whether other HSP70 proteins and other E2 ubiquitin

enzymes are differentially expressed in the three EC types. Understanding the potentially

unique roles of these proteins in EC biology, for example the response of ECs to various

cellular stressors, would be an important avenue of research.

One important stressor relevant to this discussion is chronic hyperoxia. Excess

molecular oxygen or hyperoxia is toxic to cells due to ROS (reactive oxygen species)

generation. At the other end of the oxygen pressure gradient, oxygen and glucose depletion

during an ischemic attack can lead to a chain of molecular events also resulting in ROS

generation and further cellular damage [179]. HSP27 has been shown to directly prevent

protein mis-folding by acting as a sort of antioxidant [180]. Similarly, evidence exists for the

role of HSP70 in protecting cells against ROS-induced cellular damage [181]. Furthermore,

both HSP70 and HSP27 have been shown to be highly inducible in brain cells undergoing

ischemia, epileptic seizure and hyperthermia and associate with cellular resistance to such

insults [182].

Partial pressure of O2 available to healthy cells in the body has been estimated to be 80-

100 mm Hg or 10-12.5% [183]. In a relatively more O2-rich environment such as our tissue

culture incubators at atmospheric O2 partial pressure of 20%, the cultured bovine ECs may be

exposed to chronic hyperoxic conditions, and thereby may be exposed to higher than

physiologically typical levels of intracellular ROS. In fact, lowering oxygen pressure has been

Page 110: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

96

shown to result in reduced intracellular ROS levels and ROS-induced damage to in vitro

cultured cells [184]. The elevated HSPB1 and HSPA1B in bmVECs and bmAECs,

respectively, may be evidence of selective pressure of populations of vascular ECs in culture

that can best respond to relatively higher intracellular ROS at atmospheric O2 pressures.

Interestingly, HSP27 and HSP70 are not the only proteins in my list of identified protein

(Table 1) to have an association with cellular responses to ROS. Peroxiredoxins (such as

PRDX2 and PRDX6) are a family of proteins that protect cells from oxidative stress by

catalyzing reduction of peroxides [185]. Ubiquitin conjugating enzymes (such as UBE2N and

UBE2D3) are a family of proteins that play essential roles in the unfolded protein response and

in the removal by proteolysis of oxidized proteins irreversibly damaged [179]. Treating cells

with hydrogen peroxide to generate ROS-induced oxidative stress on cells has been shown to

increase ubiquitin conjugating activity 3.5-9.5 fold and to correlate with increased intracellular

proteolysis [186]. Perhaps the presence of oxidative stress-related proteins such as HSP27 and

HSP70 in bmVECs and bmAECs, PRDX2 in bmLECs, PRDX6 in bmLECs and bmVECs,

UBE2N and UBE2D3 in bmLECs, are evidence of the unique responses of each of the three

cultured bovine mesenteric ECs to elevated internal ROS levels in an artificial environment

with atmospheric O2 levels much higher than physiological levels. The impact of atmospheric

O2 on cultured cells has previously been reported in studies of cultured lymphocytes [187, 188].

The impact of culturing ECs on cellular genetic and phenotypic character has also been

observed by Wick and colleagues [162], Amatschek and colleagues [163], and Veikkola and

colleagues [170]. Wick and colleagues [162] compared the transcriptomes of ex vivo lymphatic

and blood ECs isolated directly from human skin to cultured LECs and BECs from the same

source and found only about 10% of transcripts examined did not alter upon culture.

Page 111: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

97

Amatschek and colleagues [163] found a phenotypic drift from BEC phenotype toward an LEC

phenotype after BECs were separated from their in vivo environment and cultured. Veikkola

and colleagues [170] observed the suppression of LEC marker expression in BECs when these

cells were co-cultured with vascular muscle cells. These findings suggest that a genetic and

phenotypic drift occurs during the shift from physiological cellular environments to the

artificial culturing systems in the laboratory setting. Whether there is a predictable pattern or

trend to this genetic and phenotypic drift or whether the drift occurs with a certain levels of

variation contingent upon conditions in particular lab settings is a question that should be

examined carefully. In addition to laboratory variations in methods of EC isolation and

instrumentation used in analysis, the significant lack of overlap between lists of proteins and

transcripts published in comparative studies of various EC types and lineages would suggest

that much of the genetic and phenotypic drift observed may be contingent upon cell type as well

as on unique conditions in particular lab settings.

Page 112: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

98

CHAPTER 4

GENERATION OF A MONOCLONAL ANTIBODY SELECTIVELY REACTIVE

TO BOVINE MESENTERIC LYMPHATIC ENDOTHELIAL CELLS

BUT NOT TO BOVINE MESENTERIC VENOUS OR ARTERIAL ENDOTHELIAL CELLS

Page 113: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

99

4.1. Introduction

Basic understanding of the dynamic interaction between the lymphatic endothelium with

macromolecules and cellular components of lymph is necessary for the development of

strategies to treat lymphatic-related diseases [2, 189-191]. The deep dearth of this impactful

knowledge lends support to the continuing effort to discover and characterize surface protein

markers of LECs.

One of the methods applied to the differential identification of novel surface markers of

cells is subtractive immunization (SI). SI has been demonstrated to be successful in producing

highly selective antibodies to surface markers of subsets of normal pancreatic cells [192], of

subsets of normal kidney cells [193], of human plasma proteins [194], of epididymis-specific

sperm proteins [195], and of various types of metastatic human tumour cells [196-202]. In

chapter 2, I described a system of three EC types isolated from vessels of bovine mesentery,

bmLEC, bmAEC, bmVEC. In chapter 3, I further characterized the three cell types using a

proteomics approach in the hopes of discovering novel unique markers of bmLECs. Described

here in chapter 4, mice tolerized to bmAECs and bmVECs prior to being immunized with live

bmLECs were expected to have greater potential to produce selective antibodies to lymphatic-

specific surface antigens, thereby help to identify novel surface markers of LECs.

Standard immunization protocols routinely used to generate monoclonal antibodies

(mAb) for laboratory and clinical applications often do not result in antibodies with the desired

selectivity. SI by pre-tolerizing host animals to undesired antigens using cyclophosphamide,

has been successful where standard immunization has not been in producing mAbs targeting

highly specific epitopes within the proteome [203]. For example, SI has proven useful in

generating discriminating mAbs to proteins that are over 90% identical [204, 205]. SI is also

Page 114: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

100

thought to be advantageous for use in generating mAbs reactive to low abundant epitopes or

epitopes that are highly similar to ―self‖ antigens and thereby too weakly immunogenic in

standard immunization protocols [206]. I have taken advantage of the promises of SI to

generate a mAb selectively reactive to bmLECs and not to bmVECs or bmAECs using

immunosuppression of BALB/c mice with cyclophosphamide treatment. Initial

characterization efforts showed that the mAb recognizes a protein approximately 180 kDa in

size predominantly expressed by bmLECs and not by bmVECs or bmAECs by immunoblotting

(IB) and immunohistochemistry (IHC). The protein co-fractionates with proteins in GM1-

enriched membrane domains such as caveolin-1and the receptor tyrosine kinases Tie-2, and

VEGRF-3/Flt-4 in bmLECs. For the sake of convenience, I shall temporarily call the protein

ULSP180 for Unidentified Lymphatic Specific 180kDa Protein. Attempts at identifying

ULSP180 by immunoprecipitation followed by gel electrophoresis and tandem mass

spectrometry (MS/MS) were unsuccessful due to the possibility that the protein sequence may

not yet be part of any mammalian protein databases. Further extensive characterization is

remains needed to establish the protein target of this LEC-selective mAb to be a novel marker

of ECs of lymphatic origin.

4.2. Methods

4.2.1 Endothelial Cell Culture

Bovine mesenteric ECs of venous, arterial, and lymphatic origin were isolated and

cultured from vessels of the mesentery as previously described (Chapter 2 [167]) by the method

of Johnston and Walker [131]. Briefly, cells were dislodged by incubation with dispase and

collagenase. Dislodged cells were briefly cultured then colonies of ECs were subjected to

Page 115: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

101

limited dilution cloning to ensure EC purity. Cell populations tested positive for Prox-1,

LYVE-1, podoplanin, and VEGFR-3/Flt-4 were cultured in DMEM (Hyclone) supplemented

with 10% FBS (Fisher Scientific) in standard tissue culture conditions: 5% CO2, atmospheric

O2, and 37ºC. Cells were harvested and pooled during late growth phase when confluency had

not been reached. Approximately 5-10 x 107 cells per 500mL PBS (pH 7.4) were i.p. injected

into each mouse for SI. BmAECs and bmVECs served as the immune-telerogen while bmLECs

served as the immunogen.

4.2.2. Subtractive Immunization and Hybridoma Production

SI procedure outlined here has been previously established and described by Sleister and

Rao [205]. Briefly, Freund‘s adjuvant primed BALB/c mice at 4-6 weeks of age were injected

with a 1:1 mix of bmVECs and bmAECs, followed by injections with 2.5 mg of

cyclophosphamide prepared in 0.2mL total volume 2 and 48 hours later. This procedure was

repeated twice more at 2 and 4 weeks before tolerized mice were immunized with bmLECs.

Three booster injections with bmLECs at least 1 week apart was subsequently given. Serum

from blood samples taken by saphenous vein bleeds were screened by the same method

described below (―Antibody Screening‖) for selective reactivity to bmLECs but not to the

bmBECs. The third day after the last booster injection, hybridomas were generated by fusion of

disaggregated mouse spleen cells with the cell line Sp2/0 as previously described [207]. HAT

supplement (Gibco, Invitrogen) was added to the media (IMDM, Gibco, Invitrogen) the day

after fusion. The procedure is summarized in Figure 4.1.

4.2.3. Antibody Screening and Isotyping

Mouse serum from bleeds and tissue culture media supernatant from hybridoma

colonies were screened using an ELISA-like assay developed for the three cell types. Briefly,

Page 116: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

102

bmLECs, bmVECs, and bmAECs grown to confluency on U-bottomed 96-well plates (Falcon,

Becton Dickinson) were incubated with serum or supernatant on ice for 30 minutes. Fluids

were decanted and cells were washed at least five times with PBS before incubation with rabbit-

anti-mouse antibodies conjugated to horse radish peroxidase (HRP, Bio-Rad Laboratories) on

ice for 30 minutes. After at least five more washes with PBS, HRP was exposed to the

substrate 3, 3', 5, 5' tetramethylbenzidine (TMB, Sigma-Aldrich) until coloration was visible.

The reaction was stopped by acidification with nitric acid. Optical absorbance at 450 nm was

measured using the EL800 plate reader (BioTek). Validation of the screen was performed using

rabbit-anti-human EphB4 polyclonal antibody (Santa Cruz Biotechnology). Isotype-specific

antibodies used in the same screening procedure to determine the isotype of mAbs were

obtained from Invitrogen and used according to instructions provided. Isotype of mAbs were

confirmed with IsoStrips from Roche Applied Science according to instructions provided.

4.2.4. Immunoblot Analysis

Cells were harvested at confluency and lysed in RIPA buffer (150mM NaCl, 1% Igepal,

0.5% sodium deoxycholate, 0.1% SDS, 50mM Tris, pH 7.4, 1mM EDTA, protease inhibitor

tablets (Roche)). Lysates were cleared and quantified before proteins are separated by SDS-

PAGE. Proteins were resolved on 10% PAGE gels and transferred to PVDF (Perkin Elmer)

membranes. Antibodies used in immunoblots (IB) were: anti-Tie-2 (33.1, BD Biosciences

Pharmingen), anti-VEGFR-3/Flt-4 (C-20, Santa Cruz Biotechnology), and goat-anti-

mouse/rabbit-HRP (Bio-Rad Laboratories). Enhanced chemiluminescence (ECL) substrate was

provided by SuperSignal West Pico Chemiluminescent Substrate or by West Femto

Chemiluminescent (Thermo Scientific) whenever the ECL signal at the Pico detection level was

Page 117: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

103

inadequate. IB signals on X-ray films were converted to pixel intensity using Image Quant

version 5.0.

4.2.5. Immunohistochemistry

Confluent layers of ECs were gently lifted off the tissue culture plate and spun down

into a pellet. The entire EC pellet was fixed overnight in 4% paraformaldehyde (PFA) and

dehydrated in ethanol (EtOH) for at least 24 hours before paraffin embedding and microtome

sectioning. Paraffin sections of cells were then stained using the immunohistochemistry (IHC)

protocol available from most antibody manufacturers. Sections of cells were deparaffinised in

55ºC for 10 minutes followed by 2 xylene soakings. Decreasing concentrations of EtOH were

used to rehydrate the sections before permeabilization with 0.1% TritonX-100 in PBS.

Endogenous peroxidase activity was quenched with 3% H2O2 in methanol (MeOH) prior to

blocking with normal donkey serum. Sections were incubated with hybridoma supernatant

containing antibody raised against bmLECs for 30 minutes at room temperature. Secondary

antibody used was biotinylated donkey-anti-mouse immunoglobulin (Jackson ImmunoResearch

Laboratories Inc.). Avidin-peroxidase conjugate (Vectastain ABC kit) and the substrate 3,3‘-

diaminobenzindine (DAB) were applied as instructed by the manufacturer (Vector

Laboratories). Sections were counterstained with methyl green (Vector Laboratories) prior to

dehydration and mounting in Cytoseal-SYL (Richard-Allan Scientific).

4.2.6. ULSP180 enrichment by sucrose-gradient-based cell fractionation

Detergent-free purification of caveolin-rich membrane fractions was performed as

previously described by Song and colleagues [208] with modifications. Cells grown to

confluency in five 15-cm dishes were lifted into PBS, pelleted, and resuspended in ice-cold 500

mM sodium carbonate (pH 11), containing a cocktail of protease inhibitors (Roche Applied

Page 118: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

104

Science) . Cells were homogenized in a tight-fitting Dounce homogenizer for 20 strokes, with

an Ultra-Turrax tissue grinder (IKA Works Inc.) for three 10-second bursts, and with a

Sonifwere 250 (Branson) for three 20-second bursts. The homogenate was mixed 1:1 with 90%

sucrose prepared in MES buffered saline (MBS, 25mM MES, pH 6.5, 150mM NaCl). The

sucrose gradient was formed in an ultracentrifuge tube (Ultra-Clear™ 14 x 89mm, Beckman) as

follows from the bottom up: 4mL homogenate (45% sucrose), 6mL 35% sucrose, 2mL 5%

sucrose (both made up in MBS, with 250mM sodium carbonate). The gradient was centrifuged

at 39,000 rpm for 24 hours in an SW41-TI rotor (Beckman XL-70 Ultracentrifuge) at 4ºC.

500uL fractions were collected from the top down and analysed by dot blotting with HRP-

conjugated cholera toxin subunit B (Sigma-Aldrich). Fractions collected around the interface

between 5% sucrose and 35% sucrose contained GM1 gangliosides reactive to cholera toxin.

4.2.7. Immunoprecipitation and Identification of ULSP180 by LC-MS/MS

MAbs selectively reactive to bmLECs were pre-coupled to Dynabeads M-270 Epoxy

(Invitrogen) pre-coated with rabbit-anti-mouse polyclonal antibody (Abcam Inc.) as indicated

by the manufacturer‘s instructions. 107 magnetic beads (Dynabeads M-270 Epoxy) were coated

with at least 3µg of the rabbit cross-linking antibody in 1mL of 1M ammonium sulphate in

PBS. 106 magnetic beads coupled to mAbs selective for bmLECs were then used to

immunoprecipitate ULSP180 from dialysed sucrose fractions described above.

Cells grown to confluency in fifteen 15-cm dishes were subject to the sucrose gradient

described above with the following changes: the sucrose gradient was formed in a larger

ultracentrifuge tube (Ultra-Clear™ 25 x 89mm, Beckman) as follows from the bottom up:

16mL homogenate (45% sucrose), 16mL 35% sucrose, 8mL 5% sucrose. The gradient was

centrifuged at 28,000 rpm for 24 hours in an SW28 rotor (Beckman XL-70 Ultracentrifuge) at

Page 119: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

105

4ºC. One mL fractions were collected from the top down. Fractions containing ULSP180 were

pooled from four such gradients and dialysed into PBS prior to immunoprecipitation (IP) with

the bmLEC-selective mAb as described above. IP reactions contained approximately 2mg in

total protein. During the IP procedure, magnetic bead precipitation was performed using a

magnetic particle concentrator (DynaMag-Spin). Immunoprecipitates were eluted off the beads

with 100µL 10% SDS. 2x Laemmli sample buffer was added to a total volume of 200µL

before separation by SDS-PAGE (large gel format) and visualized by silver staining (Silver

Stain Plus, Bio-Rad). Gel regions of interest were excised and destained with 1% H2O2 prior to

in-gel digestion with sequencing-grade trypsin (Promega).

Peptides were extracted, and analyzed by LC-MS/MS (1100 nanoflow LC system, XCT

plus ion trap, Agilent Technologies). Spectrum Mill proteomics workbench (Agilent) was used

to match MS/MS spectra to non-redundant bovine protein sequences in the NCBInr database

(NCBI non-redundant downloaded in 2010, 29624 sequence entries). Unmatched spectra were

returned to another round of MS/MS search but this time against mammalian protein sequences

in SwissProt database (downloaded in 2010, 55449 sequence entries). Unmatched spectra after

the second round of MS/MS search were subjected to Spectrum Mill de novo sequencing

powered by the Sherenga algorithm [209]. In general, default settings were used. Peptide

scores >10, protein scores >11, and Scored Peak Intensity (SPI) values >70% and Sherenga

scores >150 were deemed acceptable for further automated and/or manual validation. Peptides

from in-gel-digestion of bmVEC whole cell lysate proteins and from mock IP with bmVEC

sucrose fractions, separated on the same gel helped to discriminate between potential MS/MS

match of ULSP180 and contaminating co-IP proteins.

Page 120: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

106

4.3. Results

4.3.1 Hybridoma Production and Screening

In order to develop monoclonal antibodies (mAbs) selectively reactive to bmLECs and

not bmVECs or bmAECs, the method of SI by tolerization with cyclophosphamide treatment as

outlined in figure 4.1A was employed. The method of mAb generation and screening strategy

described here were devised by adapting procedures previously described by Sleister and Rao

[205]. Instead of purified immunogen absorbed on to 96-well plates, cultures of the three

bmECs were grown in separate replicate wells in a 96-well U-bottom plate. mAbs selective for

antigen on the surface of bmLECs would be expected to bind the ECs and be retained on the

bottom of the plate similar to what occurs in an ELISA assay. This screening strategy was

intended to detect presence of bmLEC-selective mAbs in mouse test-bleed serum and in

hybridoma tissue culture supernatant. In figure 4.1.B, the screening method described was

validated using antibodies that recognize the extracellular portion of the receptor tyrosine

kinase Ephrin B4 (Eph B4) known to be highly expressed specifically by bmVECs [210]. The

screening strategy proved to be robust since the optical absorbance level at 450nm produced by

TMB substrate exposed to HRP was most intense in wells holding bmVECs (Figure 4.1.B).

The biggest concern with this strategy was that extensive washing of live cells with even

small amounts of detergent would destroy the cells. Without extensive washing, it was

expected that the high background 450nm absorbance level would mask the signal of interest.

Furthermore, any signal of interest would be expected to be less robust than that produced by

purified antigens in the case of classical ELISAs—given the number of antigen per well in the

strategy is limited by the number of cells grown in each well. The resulting signal to noise ratio

was not significantly low in the screen validation (Figure 4.1.B). With anti-EphB4 antibodies,

Page 121: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

107

the absorbance was about 0.14 units, only slightly higher than 0.1 units of the secondary

antibody alone controls. Nevertheless, a small but statistically significant difference (p<0.05)

was detectable. The advantages of this screening strategy are considered in the discussion.

The strategy validated in Figure 4.1.B was used to screen serum from test-bleeds of

mice immunized with bmLECs and/or pre-tolerized with a 1:1 mix of bmVECs and bmAECs

(Figure 4.2.A and 4.2.B). In figure 4.2.A, serum from a mouse that was not pre-tolerized to the

bmBECs failed to show selective reactivity to bmLECs even after the third boost. In figure

4.2.B, serum from a mouse that was pre-tolerized to the bmBECs showed selective reactivity to

bmLECs after the third boost. Spleen from the latter mouse was subsequently used to generate

hybridomas. Interestingly, serum reactivity to bmVECs was consistently higher than to

bmAECs correlating with the greater lineage similarity between VECs and LECs than between

AECs and LECs [211].

Figure 4.2.C shows the results of tissue culture supernatant from hybridoma colonies

screened for bmLEC-selective reactivity. Out of over 850 colonies screened, only a handful

(highlighted in colour) showed selective reactivity to bmLECs and not to the bmBECs (Figure

2C). Unfortunately, not all of these colonies survived further clonal expansion. Hybridoma

8C8 and 5D5 produced the most robust signals in the screen but because 5D5 did not survive,

8C8 was chosen for further analysis.

Page 122: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

108

Figure 4.1. Screening Assay Validation. A/ The subtractive immunization procedure used to

generate mouse monoclonal antibodies selectively reactive to bmLECs and not to bmVECs or

bmAECs is summarized. B/ The hybridoma ELISA-like screen was validated using a

polyclonal anti-EphB4 antibody that targets the extracellular portion of EphB4. The antibody

selectively binds to bmVECs grown on U-bottom 96-well plates. Absorbance of light at

wavelength of 450nm is produced by TMB substrate in the presence of HRP provided by

conjugated goat-anti-rabbit antibodies. Error bars represent triplicate wells in the screen.

Page 123: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

109

Page 124: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

110

Figure 4.2. Serum and Hybridoma Screening. A/ A control mouse was immunized with

bmLECs but not pre-tolerized to bmVECs and bmAECs. Mouse serum was collected after the

first immunization and the third boost. This control mouse failed to develop and release

selective antibodies for bmLECs into blood serum. B/ Pre-tolerized mouse showed evidence of

presence of antibodies selective for bmLECs in its serum. NB: Samples in panels A and B were

analysed the same day under the same conditions. Error bars in A and B represent triplicate

wells in the screen. C/ 100 representative hybridoma colonies are shown out of over 850

colonies screened. Many hybridoma colonies depicted in grey showed no selective reactivity to

bmLECs. The modest few showing selectivity only bmLECs is highlighted in colour.

Secondary antibody only control is shown in fuchsia.

Page 125: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

111

Page 126: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

112

4.3.2. MAb isotyping

The mAb from hybridoma sub-clone of colony 8C8 was found to be of isotype IgG1-

kappa by two different methods. In figure 3A, HRP-conjugated secondary antibodies raised

against various types of mAbs were used in the same 96-well screening method described in

figures 4.1.B and 4.2. The advantage of this method was that the isotype of the mAb that

recognizes bmLECs would be assayed and any unwanted Abs present in the hybridoma culture

supernatant would be washed away. The disadvantage was that any kind of contamination due

to failed establishment of clonal colonies may not be detectable. In figure 4.3.B, direct

absorbance of hybridoma culture supernatant to commercially available isotyping strips was

performed. It was frequently observed that before clonal expansion, mixed colonies of

hybridomas showed traces of IgM detectable by the isotyping strips but not by the 96-well

screening method. Single cell cloning was sufficient to eliminate the IgM contaminating

antibodies.

4.3.3. MAb is Selective for bmLECs

To determine whether the 8C8 mAb recognizes formalin fixed antigen on bmLECs,

pellets of bmECs were fixed, embedded in paraffin and sectioned. An antibody of the same

isotype as 8C8 but with no reactivity toward any bmECs (4H4 mAb) provided the negative

control mAb (Figure 4.4.A). Above-background staining of bmVECs by the 8C8 mAb (Figure

4.4.C), round peri-nuclear structures appeared darkly stained in bmLECs (Figure 4.4.B). In the

left panel showing one sectioning orientation of bmLECs, the peri-nuclear structure reactive to

8C8 mAb seemed to be present only once per lymphatic cell (Figure 4.4.B). 8C8 mAb also

stained the extracellular space on the apical side of the bmLEC monolayer as visible in the

sectioning orientation shown on the right panel of figure 4.4.B.

Page 127: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

113

Figure 4.3. MAb Isotyping. The mAb from hybridoma sub-clone of colony 8C8 was isotyped

using two methods: A/ Using isotype-specific secondary antibodies in the same screen

described in Figure 4.1.B, and B/ Direct absorbance of the antibody on commercially available

isotyping strips. Both methods show 8C8 mAb to be of isotype IgG1-kappa.

Page 128: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

114

Page 129: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

115

Figure 4.4. Antibody Validation. Sections of bmEC pellets embedded in paraffin were used

to determine whether 8C8 mAb is reactive to formalin-fixed antigen and therefore amenable for

use in IHC. A/ 4H4 is a mAb from a hybridoma colony found in the initial screen to be non-

selective for bmLECs. 4H4 is used here as a negative control to help determine the level of

background staining. B/ Two different sectioning orientations of bmLECs are shown. The dark

round peri-nuclear staining that appears once per cell is visible in the left sectioning orientation

(arrow head). The darkly stained extracellular space on the apical side of a bmLEC monolayer

is visible in the right sectioning orientation (arrow head). C/ 8C8 mAb is not reactive to

bmVECs. Microscope magnification of 40x was used.

Page 130: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

116

Page 131: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

117

4.3.4. 8C8 mAb recognizes a protein of MW approximately 180kDa

In order to ascertain whether 8C8 mAb recognizes denatured antigen in membrane

fractions of bmLECs but not in those of bmVECs, proteins from sucrose gradient fractions of

bmLEC and bmVEC lysates were separated by SDS-PAGE and subjected to immunoblotting

with 8C8 mAb. EC lysates were fractionated to enrich for membranous proteins prior to SDS-

PAGE and IB. A step sucrose gradient was prepared with cell lysates in 45% sucrose at the

bottom of the gradient. Ultracentrifugation resulted in floatation of low density membrane

fractions to the interface of 35% and 5% sucrose steps in the gradient. This method was

developed to isolate caveolin-enriched plasma membrane domains [208, 212], however, the

alkalinity of the sodium carbonate buffer used during cell lysis (pH 11) results in a high

percentage of non-raft membrane proteins to accumulate at the 35%-5% sucrose interface [213]

where GM1 gangliosides also migrate (Figure 4.5.A). Thus, the method developed by Song and

colleagues [208, 212] is a convenient method of fractionating cells to enrich for membranous

proteins, wherever in the cell those membrane fractions may have originated [213].

In figure 4.5.B, 8C8 mAb used in the IB did recognize a protein of approximately

180kDa that co-migrated with GM1 gangliosides. The protein will henceforth be provisionally

named ULSP180 for Unidentified Lymphatic Specific Protein of 180 kDa. While this

experiment does not allow for the conclusion that ULSP180 is a raft associated protein, it does

provide evidence that ULSP180 is likely to be a membranous protein. The only known

lymphatic-specific membranous protein marker that resolves around the same molecular weight

(MW) region on an SDS-PAGE gel is VEGFR-3/Flt-4. However, in figure 4.5.C, anti-VEGFR-

3/Flt-4 antibodies failed to produce a signal of the same MW as ULSP180. The loading control

was provided by Tie-2, which is expressed in both bmLECs and bmVECs (Figure 4.5.D).

Page 132: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

118

These results indicated that fractionation by step sucrose gradient is a viable method of

enriching for ULSP180 for further analysis and possible identification or sequencing. The work

flow from lab bench to in silico protein sequence analysis is detailed in figure 4.6.

Page 133: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

119

Figure 4.5. ULSP180 Characterization. Proteins from sucrose gradient fractions of bmLEC

and bmVEC lysates were separated by SDS-PAGE and immunoblotted using antibodies raised

against proteins as shown. A/ Fractions 4 and 5 (500 µL collected from top down) were

enriched for GM1 gangliosides, which binds to HRP-conjugated cholera toxin sub-unit B in this

dot blot. B/ 8C8 mAb recognizes a protein with MW of approximately 180 kDa (temporarily

called ULSP180) co-migrating with GM1-positive membrane domains enriched in fractions 4

and 5. C/ The lymphatic marker VEGFR-3/Flt-4 is present only in bmLECs but not bmVECs.

VEGFR-3/Flt-4 does not co-migrate as tightly as ULSP180 does with GM1-positive membrane

domains in the gradient. D/ Tie-2 is present in both bmLECs and bmVECs. Tie-2 also does not

co-migrate as tightly as ULSP180 does with GM1-positive membrane domains in the gradient.

Page 134: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

120

Page 135: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

121

Figure 4.6. Schema of Mass Spectrometric Analysis of ULSP180. Depiction of the work

flow from It lab bench to in silico analysis of MS/MS spectra by Spectrum Mill software and

return to It lab bench for protein validation. Not shown here are the multiple rounds of MS/MS

searching possible to account for modifications such as phosphorylation that may result in

initial uninterpreted spectra. ‗Hopelessly‘ uninterpretable spectra can be subjected to Sherenga-

powered de novo sequencing before the return to classic biochemistry methods.

Page 136: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

122

Page 137: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

123

4.3. 5. Mass Spectrometric Analysis of ULSP180

Tandem mass spectrometry (MS/MS) is a powerful method of obtaining protein

sequence information from an unidentified protein from a sample, provided that the mixture of

protein in the sample has been sufficiently simplified [214]. The ability to fractionate bmLECs

and produce fractions enriched in ULSP180 (Figure 4.5) was a large step toward the potential

identification of ULSP180. Furthermore, collected gradient fractions containing ULSP180 can

be located by a simple dot blot with cholera toxin subunit B conjugated to HRP (Figure 4.5).

Quantification of IB signals of ULSP180 from blots of sucrose fractions showed virtually 100%

of the signal was distributed around the 35%-5% sucrose interface where approximately 8% of

the total protein in the gradient migrated (Figure 4.7.A). Fractions containing ULSP180 were

pooled and dialysed into PBS (MW cutoff 10kDa) before IP with 8C8 mAb bound to magnetic

beads coated with rabbit-anti-mouse cross-linking antibodies (Figure 4.7.B). Undialysed

sucrose fractions were not amenable to IP (Figure 4.7.B, lane 10). Direct IP with 8C8 mAb and

Protein G Sepharose was also likewise unsuccessful (Figure 4.7.B, lane 6).

In figure 4.7.C, ULSP180-enriched fractions from a scaled up sucrose gradient were

pooled, dialysed, and subject to IP with 8C8 mAb. The same sucrose gradient was produced

from bmVECs processed identically to bmLECs. Total protein in each IP was approximately

2mg. Eluted immunoprecipitates separated by SDS-PAGE and visualized by silver stain. A

faint band present only in the bmLEC IP appeared instead of the anticipated strong band where

ULSP180 (Figure 4.7.C). Notwithstanding, thin sections of the gel corresponding to where

ULSP180 resolved were excised and subjected to MS/MS analysis. Corresponding gel regions

of SDS-PAGE-resolved bmVEC proteins were likewise processed and served as a negative

control.

Page 138: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

124

Two high scoring MS/MS mass spectra derived from tryptic peptides in bmLEC IP

samples but not detectable in bmVECs are shown in figure 4.8. One spectrum matched to the

theoretical MS/MS spectra of bovine Proteolipid 2 (PLP2, Figure 4.8.A). Surprisingly, PLP2 is

a 17kDa protein. A mismatch is highly unlikely considering the decoy database resulted in no

match and that 89.9% of spectral peak-detected ion current explained by the search

interpretation (SPI%, Figure 4.8.A). The other spectrum in figure 4.8.B was not adequately

interpreted in the first round of MS/MS search using the bovine database. The spectrum

matched to the theoretical MS/MS spectra of mouse Complement protein C3 during the second

round of searching using the mammalian database (Figure 4.8.B). C3 has a MW of 186kDa,

expected of a protein resolving in the region of ULSP180 on an SDS-PAGE gel. A mismatch is

highly unlikely considering the decoy database resulted in a poor match (forward-minus-reverse

score of 11.74) and that SPI% was 96.5% (Figure 4.8.B). Whether these matches are protein-

interacting partners of ULSP180 or whether they provide clues as to the true sequence of the

protein requires further validation with PLP2 and C3-specific antibodies. Until further

validation, ULSP180 remains ‗unidentified.‘

A large number of spectra were not interpretable by MS/MS searching and were subject

to de novo sequence analysis. Figure 4.9.A shows an example of a MS/MS spectrum

unsuccessfully matched during the initial MS/MS searches. Figure 4.9.B shows the same

MS/MS spectrum successfully sequenced by Sherenga (Sherenga score 178). However, the

amino sequences derived this way was not successfully matched to any protein in databases

using MS-Blast [215] by the Suyaev lab.

Page 139: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

125

Figure 4.7. Enrichment and Immunoprecipitation of ULSP180. A/ One mL fractions were

collected from the top of the gradient and protein quantities were determined for each fraction.

The 8C8 mAb was used to detect ULSP180 in the different fractions by IB. IB signals of

ULSP180 were converted to pixel intensity using ImageQuant® version 5.0. Virtually 100 %

of the ULSP180 signal appeared in sucrose gradient fractions 2, 3, and 4, which—when

combined—contained approximately 8% (±1%) of total protein mass in the gradient. Error bars

represent 4 independent gradients. B/ IP for ULSP180 by 8C8 mAb from pooled fractions 2, 3,

and 4, was optimal with the use of magnetic beads covalently linked to a rabbit-anti-mouse

cross-linking antibody (Lane 4). Protein G sepharose failed to immunoprecipitate ULSP180

(Lane 6). Sucrose gradient fractions had to be dialysed overnight into PBS (pH 7.4) using

dialysis tubing with MW cut-off of 10kDa (Lane 10). Samples of dialysed bmLEC sucrose

fractionate before and after IP were loaded in lanes 2 and 8. An 8% SDS-PAGE gel was used

for lanes 1-8, 10% was used for lanes 9-11. BmVECs were identically treated and sucrose

fractionate of bmVECs does not show presence of ULSP180 (Lanes 1, 3, 5, and7). In both A/

and B/, ECL substrate was provided by Ist Femto Chemiluminescent Substrate (Pierce). C/

Scaled up sucrose fractionate of bmVECs and bmLECs were subject to IP with the 8C8 mAb.

Immunoprecipitates were eluted and separated by an 8% large format SDS-PAGE gel.

ULSP180 did not appear as a strong band by silver staining and was only as visible as the

myoferlin band indicated.

Page 140: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

126

Page 141: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

127

Figure 4.8. Mass Spectrometric Analysis of ULSP180. Sections of the gel in Figure 7C that

corresponded to where ULSP180 resolved were excised and subjected to in-gel digestion with

trypsin. Extracted tryptic peptides were separated by HPLC and analysed by tandem mass

spectrometry (MS/MS). Tryptic peptides from corresponding gel regions of SDS-PAGE

resolved bmVEC proteins served as a negative control. MS/MS mass spectra of two tryptic

peptides in bmLEC IP samples but not detectable in bmVECs are shown. A/ The MS/MS

spectrum of one of the two peptides matched to the theoretical MS/MS spectrum of a segment

of a 17kDa transmembrane protein called Proteolipid 2 in the bovine NCBI database. MS/MS

search of the forward database did not result in a second ranked match (Rank 2 score of 0).

MS/MS search of the decoy database did not result in any match (Reverse match score of 0). B/

The MS/MS spectrum of the second peptide matched to the theoretical MS/MS spectrum of a

segment of a 186 kDa protein excreted into the extracellular space called Complement C3 in the

mouse SwissProt database. MS/MS search of the forward database did not result in a second

ranked match (Rank 2 score of 0). MS/MS search of the decoy database resulted in a poor

match (Reverse match score of 6.09). NB: In both A and B, black peaks are unmatched ion

peaks whereas labelled and coloured peaks are successfully matched ion peaks.

Page 142: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

128

Page 143: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

129

Figure 4.9. Mass Spectrometric Analysis of ULSP180 (continued). A/ Another MS/MS

mass spectrum derived from tryptic peptides in bmLEC IP samples but not detectable in

bmVECs is shown. This particular spectrum is one example of a spectrum that did not match

well to any theoretical MS/MS spectra in the databases. Black peaks indicate unmatched ion

peaks. B/ The protein sequence was procured from the MS/MS spectrum in A/ using the

Spectrum Mill de novo peptide sequencing function. Coloured peaks are successfully explained

ion peaks whereas black peaks are not explained by the proposed sequence shown at top. NB:

Isoleucine (I) has the same mass as leucine (L) and is indicated as isoleucine even though

Sherenga cannot distinguish between the two amino acids. The same thing can be said of lysine

(K) and glutamine (Q), indicated as glutamine.

Page 144: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

130

Page 145: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

131

4.4. Discussion

I have attempted to discover a protein marker of LEC that is absent from ECs of venous

and arterial origin using subtractive immunization (SI). SI yielded a small number of

hybridoma clones producing mouse monoclonal antibodies (mAb) selectively reactive to an

antigen on the surface of bmLECs. Initial tests of the mAb from hybridoma colony 8C8

showed it to be non-reactive to bmVECs. 8C8 mAb recognizes a protein of approximately

180kDa in apparent size on an immunoblotted SDS-PAGE gel. The protein, temporarily named

ULSP180, localizes to a peri-nuclear structure as well as to the plasma membrane on the apical

side of a bmLEC monolayer. ULSP180 is highly soluble and can be immunoprecipitated from

detergent-free sucrose fractions dialysed into PBS. The low silver signal of IP products at the

expected size on the SDS-PAGE gel may be due to low IP protein yield or the possibility that

ULSP180 may not efficiently reduce silver (Figure 4.7.C). Initial attempts to perform mass

spectral analysis on ULSP180 resulted in two potential database matches that require further

validation. Taken together, SI showed tremendous potential as a viable procedure for

discovering novel candidate surface markers of LECs.

Theoretically, an ideal protein marker of LECs would be (i) absent from all sub-types of

ECs of venous and arterial lineages, (ii) absent from lymphatic vessel pericytes, LMCs,

fibroblasts, surrounding connective tissues, and other unrelated cell types (iii) present in most

anatomical subtypes of LECs in most in vivo biological and in vitro cell culture contexts, (iv)

useful for marking LECs in a variety of species, (v) useful for marking LECs at most

developmental stages. Realistically, even the best known LEC markers have been found to be

expressed in other cell types (carefully reviewed by Baluk and McDonald [116]). For

examples, FOXC2 and podoplanin are also found in podocytes [114, 216]; Prox-1 and

Page 146: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

132

podoplanin are present in certain types of nerve cells [217, 218]; LYVE-1, Prox-1, and

VEGRF-3/Flt-4 have all been detected in mesothelial cells [110].

Beside marker infidelity, another cause for the difficulty in trying to discover a genuine

LEC protein marker using in vitro EC cultures may be the gene expression shift between LEC

and BEC cell types encapsulated by the term ―cellular plasticity.‖ For example, BECs have

been observed to shift to a more LEC phenotype upon isolation and culture in vitro [163].

When BECs were isolated and co-cultured with vascular muscle cells, LECs markers up-

regulated in the cultured BECs were repressed [170]. The impact that isolating and culturing

cells have on the genotype and phenotype of those cells was observed previously by Wick and

colleagues [219]. They noted that only about 10% of transcripts studied retained the same

differential profile between LECs and BECs before and after culture [219], suggesting that the

shift from physiological cellular environments to the artificial culturing systems in the

laboratory setting leads to measurable changes in gene expression. However, the highly stable

expression of VEGFR-3/Flt-4 by isolated bmLECs and its absence in bmVECs in my studies

suggest that if a protein were to be discovered and validated to have selective expression in

LECs and not in the BECs then its expression would also likely to be highly stable regardless of

in vitro culture conditions.

Another cause of the difficulty of discovering a LEC marker that meets all criteria (i)-

(v) is the phenotypic variability of LECs contingent upon anatomical location. Chi and

colleagues reported evidence that the same EC type from different anatomical sites have

different gene expression programs [159]. Add to this complexity the fact that the same LEC

subtype from the identical anatomical site may be responding to different physiological or

pathological stimuli such as inflammation and injury, and the task of defining a consistent LEC

Page 147: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

133

phenotype becomes even more difficult. In addition to striving to arrive at an amino acid

sequence for ULSP180, extensive IHC profiling of the presence of the protein in a variety of

adult and embryonic tissues is also required in order to establish ULSP180 as a bona fide novel

surface marker of ECs of the lymphatic lineage. The challenges of performing mass spectral

analysis on ULSP180 add to the above difficulties in trying to discover a truly novel protein

marker of LECs.

If subsequent validation supports the conclusion that ULSP180 is a very small protein

with a theoretical MW of 17kD, the observed 180kDa may be a result of heavy post

translational modifications like glycosylation. Indeed, proteins that are comprised of 78%

carbohydrate have been observed and studied in the past, for example, mucins and salivary

glycoproteins [220, 221]. However, the much larger difference in observed and theoretical MW

would suggest that the protein mass is at least 90% due to covalently linked groups on the

amino acid chain. To the best of my knowledge, ULSP180 may be the first of this kind of

protein if validation experiments support such outcome. If validation supports the outcome that

ULSP180 is a complement protein, then the sequence of this complement protein—unmatched

with the approximately 600 sequences of complement proteins already in the database—may

become a new addition. Uninterpretable spectra unsuccessfully matched even after being

subjected to Sherenga de novo sequencing points to the third possibility that ULSP180 may

simply be an ―unknown‖ protein yet to be part of any protein database. If subsequent validation

proves this to be the case, then the outcome would become an example of a drawback of

database-driven methods to discover novel proteins.

These limitations are altogether not insurmountable given the availability and reliability

of classical biochemical methods to characterize ULSP180. Moreover, SI still stands as a

Page 148: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

134

viable method in endeavours to discover novel markers of LECs. Future repeat of the

procedure described in this study would be improved by considering ways of increasing the

sensitivity of the screening method. I have mentioned the limit placed on the number of

antigens by assaying live cells. Disaggregated plasma membrane proteins adsorbed directly to

the bottom of 96-well plates may help increase the sensitivity of the assay. However, the

advantage of using live cells—as was done in this study—was that antibodies would also be

screened for reactivity to unaltered antigen and thereby potentiates the use of the antibody for

sorting live cells prior to tissue culture. Another way to improve the SI procedure would be to

increase the antigenicity of LECs. Some examples of refinement include using mAbs generated

from the first round of subtractive immunization to block identified antigenic sites and/or

employing neuraminidase treatment as previously described [222] to expose hidden antigens on

the surfaces of LECs. The screening procedure would need to be adjusted accordingly. Despite

these limitations, SI should still be considered a useful tool for LEC marker discovery.

There exists a strong consensus that identifying phenotypic regulators of individual cells

of LEC and BEC lineages is an important avenue of research [116]. Despite many drawbacks

to in vitro cultures and available methods, EC biology researchers continue to search for new

bona fide markers and meticulously profile expression patterns of the few LEC makers known

to date. Yet another way to mitigate the problem of limited-exclusivity in selectivity is the

reliance on detection of multiple markers in the cells of interest before they are declared as

being of the lymphatic endothelial cell lineage. Once properly validated, LECs can be used to

determine the biological functions of identified candidate protein markers. The continued effort

by our laboratory and others to identify and functionally characterize new markers of LEC is

Page 149: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

135

important for deepening understanding of the LEC phenotype and molecular determinants of

the cell lineage.

Page 150: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

136

CHAPTER 5

CONCLUSIONS AND FUTURE DIRECTIONS

Page 151: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

137

To address the question of what molecular signatures give rise to the genotypic and

phenotypic character of the lymphatic endothelial cell lineage distinguishable from blood

endothelial cell lineages, I began with the isolation and culturing of ECs from lymphatic,

venous, and blood vessels from bovine mesentery. The resulting cells bmLEC, bmVEC and

bmAEC appeared to behave as expected of cells of their respective lineages. This was

evidenced by the stable expression of endothelial receptor tyrosine kinase Tie-2 and of

appropriate cellular markers Prox-1, VEGFR3, and Neuropilin-1 that define the particular

origin of each cell preparation. Furthermore, I showed in chapter 2 that while bmLECs

responded slightly more readily to Ang-2 stimulation, bmVECs and bmAECs were more

sensitive to Ang-1 stimulation. Exposure of bmLECs to Ang-2 induced marginally higher levels

of proliferation and survival than did exposure to Ang-1, whereas exposure to Ang-1 resulted in

higher levels of migration in bmLECs than did to Ang-2. These results suggest that each EC

type possesses a unique repertoire of expressed proteins that allows that cell type to

differentially respond to the angiopoietins.

To answer the question of what molecular signatures make lymphatic ECs different

from arterial or venous ECs and perhaps explain the observed behaviour of bmLECs distinct

from the bmBECs as described in chapter 2, I compared 2D-PAGE-resolved protein profiles of

bmLECs to those of bmVECs and bmAECs. Chapter 3 is a record of the results of quantitative

comparison of 2D-PAGE electrophoretograms produced in triplicate for each of the three cell

types. Protein identification by LC-MS/MS was performed to identify 39 proteins found to be

present at statistically significantly different levels in the three cell types (p<0.05). Most of the

39 proteins have not been previously reported in EC proteomic studies of 2D-PAGE

electrophoretograms. Three proteins, HSPA1B (HSP70 family member), HSPB1 (HSP27

Page 152: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

138

family member), and UBE2D3 (a member of E2 ubiquitin-conjugating enzymes) were validated

and found to be at highest levels in bmAECs, bmVECs, and bmLECs, respectively.

The lack of substantial overlap between my results and those of other groups‘

comparative studies implies that a genetic and phenotypic drift occurs during the shift from

physiological cellular environments to the artificial culturing systems in the laboratory setting.

It is possible for this shift to further confound experimental results obtained from in vitro EC

cultures that may already be incredibly phenotypically divergent according to anatomical

origin. However disheartening this finding may be, it still should not be reason for complete

dismissal of the cell system I built since with regards to the angiopoietins, the cells seemed to

behave as expected from in vivo studies [43, 44, 55, 56]. Another important fact to note is that

despite evidence of artificially-induced changes to the EC proteome, expression of VEGR-3, a

more or less reliable lymphatic protein marker [103], remained high in bmLECs and was not

detectable in bmVECs or bmAECs despite long-term culture of the cells. Perhaps this suggests

that a similarly reliable marker that contributes to the maintenance of the lymphatic phenotype

is still discoverable with a cell system like one I have built. However, extensive post-discovery

validation necessarily follows any such discovery.

In chapter 4, I described an adapted protocol of standard immunization used to generate

monoclonal antibodies selectively reactive to bmLECs and not to bmVECs or bmAECs. The

strategy was to immunosuppress mice with cyclophosphamide treatment after injections of

bmVECs and bmAECs so that the mice would fail to develop antibodies that recognize shared

antigens on the surface of all three cell types. Initial characterization efforts showed that the

mAb recognizes a protein approximately 180 kDa in size predominantly expressed by bmLECs

and not by bmVECs or bmAECs by immunoblotting and immunohistochemistry. The protein,

Page 153: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

139

tentatively named ULSP180, co-fractionates with GM1-enriched membranes and the receptor

tyrosine kinases Tie-2, and VEGRF-3/Flt-4 in a step sucrose gradient. Further characterization

would be required to establish the protein target of this LEC-selective mAb to be a true marker

of endothelial cells of lymphatic origin. Nevertheless, these initial results do tentatively support

the three-cell system set up and SI as promising tools for the identification of novel markers and

phenotypic regulators of LECs.

The next steps should involve answering the following questions about ULSP180:

Is the protein absent from other sub-types of ECs of venous and arterial lineages, and present in

other sub-types of LECs? For example, what is the expression of ULSP180 in microvascular

ECs from blood capillaries of the two systems? Is the protein absent from lymphatic vessel

pericytes, lymphatic muscle cells, smooth muscle cells, fibroblasts, and connective tissues

surrounding vessels? Is ULSP180 present in which anatomical subtypes of LECs in which in

vivo biological and in which in vitro cell culture contexts? Is ULSP180 present in LECs in a

variety of species including human? Is ULSP180 present in LECs during early developmental

stages? Questions listed here provide basis for validation experiments before ULSP180 can be

declared a new marker of ECs of the lymphatic lineage. Further characterization should be

aimed at elucidating its potential role as a molecular regulator of the LEC phenotype.

To answer some of these questions about ULSP180, the ability of 8C8 mAb to

recognize a 180kDa protein from immunohistochemical sections of adult mouse, rat, bovine,

sheep, and human tissues from a variety of organ sites that are co-stained with known LEC-

specific antibodies need to be verified. Alternatively, freshly isolated and uncultured ECs from

vessels belonging to different anatomical sites can be examined directly by flow cytometry

using 8C8 mAb conjugated to fluorescent tags. Developmental arrays of mouse tissues can be

Page 154: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

140

used to establish whether ULSP180 is present at earlier developmental stages and therefore may

play regulatory roles in the development and maturation of the lymphatic system. These

experiments should aim to generate an anatomical map of where ULSP180 can be found to be

expressed.

None of the experiments proposed thus far requires the identity of ULSP180 to be

known. Alternatively, an amino acid sequence of ULSP180 should be arrived at first either by

Edman degradation or by refinement of my initial attempts to perform mass spectral analysis on

the protein. Optimization efforts should be focused on increasing the yield of

immunoprecipitated ULSP180 visualized as a silver-stained protein band on an SDS-PAGE gel.

Beside the possibility that ULSP180 is yet to be part of any protein database, the difficulty of

obtaining an unambiguous match with a candidate from the bovine protein sequence database

may also have been due to low IP yield combined with the challenges of performing mass

spectral analysis on an integral membrane protein using reversed-phase HPLC. If ULSP180 is

in fact an integral membrane protein, refinements to HPLC such as previously proposed by Ball

and colleagues [223] may help to improve recovery of separated trypsin cleavage fragments,

thereby increasing the chance of obtaining sequence information on the protein.

The advantage of knowing the protein sequence firsthand is the availability of PCR-

based methods to generate data from transcripts amplified from small amounts of tissue, tissues

even as small as laser-capture micro-dissected tissues. Another advantage of working with a

known protein amino acid sequence is the availability of bioinformatics tools to analyse or

predict homologous protein domains, conserved functional domains, and various types of

consensus or signal sequences indicative of protein localization and function.

Page 155: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

141

In future endeavours to continue the identification and characterization of LEC-specific

molecules along similar lines, there is no reason to limit the source of ECs to lymphatic and

blood vessels of the mesentery. In fact, there is value in understanding the LEC phenotype in

specific anatomical contexts. The development of drug delivery strategies utilizing lymphatic

vessels and lymph nodes to increase bioavailability of protein-based drug compounds and

immunoactive biomolecules would greatly benefit from an understanding of how the lymphatic

endothelium at different anatomical sites absorb, process, and interact with luminal molecules

during lymph transport and formation [224]. Understanding the lymphatic phenotype during

inflammation and during wound healing would aid the development of therapies for chronic

inflammatory and chronic lymphatic insufficiency, respectively [2]. Design of pharmaceuticals

to enhance or mitigate lymphatic functions for various pathological conditions, would also

greatly benefit from development of in vitro systems that helps to predict the response of

different endothelia to therapeutic molecules. I have described such as system in these pages.

As the spectrum of human disorders involving the lymphatic system continues to expand day by

day, the rewards of understanding the biology of this unique system are bountiful. I hope to

have contributed even in the smallest way to this collective understanding.

Page 156: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

142

REFERENCES

1. Skandalakis JE, Skandalakis LJ, Skandalakis PN: Anatomy of the lymphatics. Surg

Oncol Clin N Am 2007, 16(1):1-16.

2. Tammela T, Alitalo K: Lymphangiogenesis: Molecular mechanisms and future

promise. Cell, 140(4):460-476.

3. Johnson LA, Jackson DG: Cell traffic and the lymphatic endothelium. Ann N Y Acad

Sci 2008, 1131:119-133.

4. Vestweber D: Lymphocyte trafficking through blood and lymphatic vessels: more

than just selectins, chemokines and integrins. Eur J Immunol 2003, 33(5):1361-1364.

5. Shields JD, Emmett MS, Dunn DB, Joory KD, Sage LM, Rigby H, Mortimer PS,

Orlando A, Levick JR, Bates DO: Chemokine-mediated migration of melanoma cells

towards lymphatics--a mechanism contributing to metastasis. Oncogene 2007,

26(21):2997-3005.

6. Das S, Skobe M: Lymphatic vessel activation in cancer. Ann N Y Acad Sci 2008,

1131:235-241.

7. Achen MG, Stacker SA: Molecular control of lymphatic metastasis. Ann N Y Acad

Sci 2008, 1131:225-234.

8. Hirakawa S: From tumor lymphangiogenesis to lymphvascular niche. Cancer Sci

2009, 100(6):983-989.

9. He Y, Rajantie I, Pajusola K, Jeltsch M, Holopainen T, Yla-Herttuala S, Harding T,

Jooss K, Takahashi T, Alitalo K: Vascular endothelial cell growth factor receptor 3-

mediated activation of lymphatic endothelium is crucial for tumor cell entry and

spread via lymphatic vessels. Cancer Res 2005, 65(11):4739-4746.

Page 157: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

143

10. Wong SY, Hynes RO: Lymphatic or hematogenous dissemination: how does a

metastatic tumor cell decide? Cell Cycle 2006, 5(8):812-817.

11. Farnsworth RH, Achen MG, Stacker SA: Lymphatic endothelium: an important

interactive surface for malignant cells. Pulm Pharmacol Ther 2006, 19(1):51-60.

12. Shields JD, Fleury ME, Yong C, Tomei AA, Randolph GJ, Swartz MA: Autologous

chemotaxis as a mechanism of tumor cell homing to lymphatics via interstitial flow

and autocrine CCR7 signaling. Cancer Cell 2007, 11(6):526-538.

13. Wiley HE, Gonzalez EB, Maki W, Wu MT, Hwang ST: Expression of CC chemokine

receptor-7 and regional lymph node metastasis of B16 murine melanoma. J Natl

Cancer Inst 2001, 93(21):1638-1643.

14. Miteva DO, Rutkowski JM, Dixon JB, Kilarski W, Shields JD, Swartz MA:

Transmural flow modulates cell and fluid transport functions of lymphatic

endothelium. Circ Res, 106(5):920-931.

15. Murfee WL, Rappleye JW, Ceballos M, Schmid-Schonbein GW: Discontinuous

expression of endothelial cell adhesion molecules along initial lymphatic vessels in

mesentery: the primary valve structure. Lymphat Res Biol 2007, 5(2):81-89.

16. Zoltzer H: Initial lymphatics--morphology and function of the endothelial cells.

Lymphology 2003, 36(1):7-25.

17. Poggi P, Marchetti C, Tazzi A, Scelsi R: The lymphatic vessels and their relationship

to lymph formation in the human urinary bladder. Lymphology 1995, 28(1):35-40.

18. Navas V, O'Morchoe PJ, O'Morchoe CC: Lymphatic system of the rat pancreas.

Lymphology 1995, 28(1):4-20.

Page 158: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

144

19. Johnson LA, Clasper S, Holt AP, Lalor PF, Baban D, Jackson DG: An inflammation-

induced mechanism for leukocyte transmigration across lymphatic vessel

endothelium. J Exp Med 2006, 203(12):2763-2777.

20. Sawa Y, Tsuruga E, Iwasawa K, Ishikawa H, Yoshida S: Leukocyte adhesion molecule

and chemokine production through lipoteichoic acid recognition by toll-like

receptor 2 in cultured human lymphatic endothelium. Cell Tissue Res 2008,

333(2):237-252.

21. Sawa Y, Shibata K, Braithwaite MW, Suzuki M, Yoshida S: Expression of

immunoglobulin superfamily members on the lymphatic endothelium of inflamed

human small intestine. Microvasc Res 1999, 57(2):100-106.

22. Podgrabinska S, Kamalu O, Mayer L, Shimaoka M, Snoeck H, Randolph GJ, Skobe M:

Inflamed lymphatic endothelium suppresses dendritic cell maturation and function

via Mac-1/ICAM-1-dependent mechanism. J Immunol 2009, 183(3):1767-1779.

23. Teoh D, Johnson LA, Hanke T, McMichael AJ, Jackson DG: Blocking development of

a CD8+ T cell response by targeting lymphatic recruitment of APC. J Immunol

2009, 182(4):2425-2431.

24. Kuroshima S, Sawa Y, Kawamoto T, Yamaoka Y, Notani K, Yoshida S, Inoue N:

Expression of Toll-like receptors 2 and 4 on human intestinal lymphatic vessels.

Microvasc Res 2004, 67(1):90-95.

25. Pegu A, Qin S, Fallert Junecko BA, Nisato RE, Pepper MS, Reinhart TA: Human

lymphatic endothelial cells express multiple functional TLRs. J Immunol 2008,

180(5):3399-3405.

Page 159: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

145

26. Marchetti C, Poggi P, Calligaro A, Casasco A: Lymphatic vessels of the human dental

pulp in different conditions. Anat Rec 1992, 234(1):27-33.

27. Ohhashi T, Takahashi N: Acetylcholine-induced release of endothelium-derived

relaxing factor from lymphatic endothelial cells. Am J Physiol 1991, 260(4 Pt

2):H1172-1178.

28. Tsunemoto H, Ikomi F, Ohhashi T: Flow-mediated release of nitric oxide from

lymphatic endothelial cells of pressurized canine thoracic duct. Jpn J Physiol 2003,

53(3):157-163.

29. Yokoyama S, Ohhashi T: Effects of acetylcholine on spontaneous contractions in

isolated bovine mesenteric lymphatics. Am J Physiol 1993, 264(5 Pt 2):H1460-1464.

30. Koller A, Mizuno R, Kaley G: Flow reduces the amplitude and increases the

frequency of lymphatic vasomotion: role of endothelial prostanoids. Am J Physiol

1999, 277(6 Pt 2):R1683-1689.

31. Elias RM, Eisenhoffer J, Johnston MG: Role of endothelial cells in regulating

hemoglobin-induced changes in lymphatic pumping. Am J Physiol 1992, 263(6 Pt

2):H1880-1887.

32. von der Weid PY, Crowe MJ, Van Helden DF: Endothelium-dependent modulation

of pacemaking in lymphatic vessels of the guinea-pig mesentery. J Physiol 1996, 493

( Pt 2):563-575.

33. Ferguson MK: Modulation of lymphatic smooth muscle contractile responses by the

endothelium. J Surg Res 1992, 52(4):359-363.

34. Hashimoto S, Kawai Y, Ohhashi T: Effects of vasoactive substances on the pig

isolated hepatic lymph vessels. J Pharmacol Exp Ther 1994, 269(2):482-488.

Page 160: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

146

35. Pepper MS, Skobe M: Lymphatic endothelium: morphological, molecular and

functional properties. J Cell Biol 2003, 163(2):209-213.

36. Podgrabinska S, Braun P, Velasco P, Kloos B, Pepper MS, Skobe M: Molecular

characterization of lymphatic endothelial cells. Proc Natl Acad Sci U S A 2002,

99(25):16069-16074.

37. Cornford ME, Oldendorf WH: Terminal endothelial cells of lymph capillaries as

active transport structures involved in the formation of lymph in rat skin.

Lymphology 1993, 26(2):67-78.

38. Ono N, Mizuno R, Ohhashi T: Effective permeability of hydrophilic substances

through walls of lymph vessels: roles of endothelial barrier. Am J Physiol Heart Circ

Physiol 2005, 289(4):H1676-1682.

39. Takahashi T, Shibata M, Kamiya A: Mechanism of macromolecule concentration in

collecting lymphatics in rat mesentery. Microvasc Res 1997, 54(3):193-205.

40. Supersaxo A, Hein WR, Steffen H: Effect of molecular weight on the lymphatic

absorption of water-soluble compounds following subcutaneous administration.

Pharm Res 1990, 7(2):167-169.

41. Oussoren C, Storm G: Liposomes to target the lymphatics by subcutaneous

administration. Adv Drug Deliv Rev 2001, 50(1-2):143-156.

42. Kaminskas LM, Kota J, McLeod VM, Kelly BD, Karellas P, Porter CJ: PEGylation of

polylysine dendrimers improves absorption and lymphatic targeting following SC

administration in rats. J Control Release 2009, 140(2):108-116.

43. Gale NW, Thurston G, Hackett SF, Renard R, Wang Q, McClain J, Martin C, Witte C,

Witte MH, Jackson D et al: Angiopoietin-2 is required for postnatal angiogenesis

Page 161: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

147

and lymphatic patterning, and only the latter role is rescued by Angiopoietin-1.

Dev Cell 2002, 3(3):411-423.

44. Maisonpierre PC, Suri C, Jones PF, Bartunkova S, Wiegand SJ, Radziejewski C,

Compton D, McClain J, Aldrich TH, Papadopoulos N et al: Angiopoietin-2, a natural

antagonist for Tie2 that disrupts in vivo angiogenesis. Science 1997, 277(5322):55-

60.

45. Ahmad SA, Liu W, Jung YD, Fan F, Reinmuth N, Bucana CD, Ellis LM: Differential

expression of angiopoietin-1 and angiopoietin-2 in colon carcinoma. A possible

mechanism for the initiation of angiogenesis. Cancer 2001, 92(5):1138-1143.

46. Ahmad SA, Liu W, Jung YD, Fan F, Wilson M, Reinmuth N, Shaheen RM, Bucana CD,

Ellis LM: The effects of angiopoietin-1 and -2 on tumor growth and angiogenesis in

human colon cancer. Cancer Res 2001, 61(4):1255-1259.

47. Ochiumi T, Tanaka S, Oka S, Hiyama T, Ito M, Kitadai Y, Haruma K, Chayama K:

Clinical significance of angiopoietin-2 expression at the deepest invasive tumor site

of advanced colorectal carcinoma. Int J Oncol 2004, 24(3):539-547.

48. Sfiligoi C, de Luca A, Cascone I, Sorbello V, Fuso L, Ponzone R, Biglia N, Audero E,

Arisio R, Bussolino F et al: Angiopoietin-2 expression in breast cancer correlates

with lymph node invasion and short survival. Int J Cancer 2003, 103(4):466-474.

49. Etoh T, Inoue H, Tanaka S, Barnard GF, Kitano S, Mori M: Angiopoietin-2 is related

to tumor angiogenesis in gastric carcinoma: possible in vivo regulation via

induction of proteases. Cancer Res 2001, 61(5):2145-2153.

Page 162: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

148

50. Tanaka S, Mori M, Sakamoto Y, Makuuchi M, Sugimachi K, Wands JR: Biologic

significance of angiopoietin-2 expression in human hepatocellular carcinoma. J

Clin Invest 1999, 103(3):341-345.

51. Mitsuhashi N, Shimizu H, Ohtsuka M, Wakabayashi Y, Ito H, Kimura F, Yoshidome H,

Kato A, Nukui Y, Miyazaki M: Angiopoietins and Tie-2 expression in angiogenesis

and proliferation of human hepatocellular carcinoma. Hepatology 2003, 37(5):1105-

1113.

52. Takanami I: Overexpression of Ang-2 mRNA in non-small cell lung cancer:

association with angiogenesis and poor prognosis. Oncol Rep 2004, 12(4):849-853.

53. Lind AJ, Wikstrom P, Granfors T, Egevad L, Stattin P, Bergh A: Angiopoietin 2

expression is related to histological grade, vascular density, metastases, and

outcome in prostate cancer. Prostate 2005, 62(4):394-399.

54. Hata K, Udagawa J, Fujiwaki R, Nakayama K, Otani H, Miyazaki K: Expression of

angiopoietin-1, angiopoietin-2, and Tie2 genes in normal ovary with corpus luteum

and in ovarian cancer. Oncology 2002, 62(4):340-348.

55. Tammela T, Saaristo A, Lohela M, Morisada T, Tornberg J, Norrmen C, Oike Y,

Pajusola K, Thurston G, Suda T et al: Angiopoietin-1 promotes lymphatic sprouting

and hyperplasia. Blood 2005, 105(12):4642-4648.

56. Suri C, Jones PF, Patan S, Bartunkova S, Maisonpierre PC, Davis S, Sato TN,

Yancopoulos GD: Requisite role of angiopoietin-1, a ligand for the TIE2 receptor,

during embryonic angiogenesis. Cell 1996, 87(7):1171-1180.

57. Angeli V, Randolph GJ: Inflammation, lymphatic function, and dendritic cell

migration. Lymphat Res Biol 2006, 4(4):217-228.

Page 163: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

149

58. Douglas JL, Gustin JK, Dezube B, Pantanowitz JL, Moses AV: Kaposi's sarcoma: a

model of both malignancy and chronic inflammation. Panminerva Med 2007,

49(3):119-138.

59. Deitch EA, Xu D, Kaise VL: Role of the gut in the development of injury- and shock

induced SIRS and MODS: the gut-lymph hypothesis, a review. Front Biosci 2006,

11:520-528.

60. Sakamoto I, Ito Y, Mizuno M, Suzuki Y, Sawai A, Tanaka A, Maruyama S, Takei Y,

Yuzawa Y, Matsuo S: Lymphatic vessels develop during tubulointerstitial fibrosis.

Kidney Int 2009, 75(8):828-838.

61. Cui Y: Impact of lymphatic vessels on the heart. Thorac Cardiovasc Surg, 58(1):1-7.

62. Dashkevich A, Bloch W, Antonyan A, Fries JU, Geissler HJ: Morphological and

quantitative changes of the initial myocardial lymphatics in terminal heart failure.

Lymphat Res Biol 2009, 7(1):21-27.

63. Geissler HJ, Dashkevich A, Fischer UM, Fries JW, Kuhn-Regnier F, Addicks K,

Mehlhorn U, Bloch W: First year changes of myocardial lymphatic endothelial

markers in heart transplant recipients. Eur J Cardiothorac Surg 2006, 29(5):767-

771.

64. Machnik A, Neuhofer W, Jantsch J, Dahlmann A, Tammela T, Machura K, Park JK,

Beck FX, Muller DN, Derer W et al: Macrophages regulate salt-dependent volume

and blood pressure by a vascular endothelial growth factor-C-dependent buffering

mechanism. Nat Med 2009, 15(5):545-552.

Page 164: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

150

65. Honda N, Jinnin M, Kajihara I, Makino T, Fukushima S, Ihn H: Impaired

lymphangiogenesis due to excess VEGF-D/Flt-4 signaling in the skin of patients

with systemic sclerosis. Br J Dermatol.

66. Horra A, Salazar J, Ferre R, Vallve JC, Guardiola M, Rosales R, Masana L, Ribalta J:

Prox-1 and FOXC2 gene expression in adipose tissue: A potential contributory role

of the lymphatic system to familial combined hyperlipidaemia. Atherosclerosis

2009, 206(2):343-345.

67. Harvey NL: The link between lymphatic function and adipose biology. Ann N Y

Acad Sci 2008, 1131:82-88.

68. Adair TH, Guyton AC: Introduction to the Lymphatic System. In: Experimental

Biology of the Lymphatic Circulation. Edited by Johnston MG, vol. 9, 1 edn. New

York: Elsevier Science Publishing Company, Inc.; 1985: 425.

69. Moghimi SM, Rajabi-Siahboomi R: Advanced colloid-based systems for efficient

delivery of drugs and diagnostic agents to the lymphatic tissues. Prog Biophys Mol

Biol 1996, 65(3):221-249.

70. Taylor AE: The lymphatic edema safety factor: the role of edema dependent

lymphatic factors (EDLF). Lymphology 1990, 23(3):111-123.

71. Adair TH, Guyton AC: Lymph Formation and its Modification in the Lymphatic

System. In: Experimental Biology of the Lymphatic Circulation. Edited by Johnston

MG, vol. 9, 1 edn. New York: Elsevier Science Publishing Company, Inc.; 1985.

72. Muthuchamy M, Zawieja D: Molecular regulation of lymphatic contractility. Ann N

Y Acad Sci 2008, 1131:89-99.

Page 165: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

151

73. Muthuchamy M, Gashev A, Boswell N, Dawson N, Zawieja D: Molecular and

functional analyses of the contractile apparatus in lymphatic muscle. FASEB J

2003, 17(8):920-922.

74. Gruetter CA, Lemke SM: Comparison of endothelium-dependent relaxation in

bovine intrapulmonary artery and vein by acetylcholine and A23187. J Pharmacol

Exp Ther 1986, 238(3):1055-1062.

75. Moore RM, Sedrish SA, Holmes EP, Koch CE, Venugopal CS: Role of endothelium

and nitric oxide in modulating in vitro responses of colonic arterial and venous

rings to vasodilatory neuropeptides in horses. Can J Vet Res 2005, 69(2):116-122.

76. Norel X, Walch L, Gascard JP, deMontpreville V, Brink C: Prostacyclin release and

receptor activation: differential control of human pulmonary venous and arterial

tone. Br J Pharmacol 2004, 142(4):788-796.

77. Back M, Walch L, Norel X, Gascard JP, Mazmanian G, Brink C: Modulation of

vascular tone and reactivity by nitric oxide in porcine pulmonary arteries and

veins. Acta Physiol Scand 2002, 174(1):9-15.

78. Furchgott RF, Zawadzki JV: The obligatory role of endothelial cells in the relaxation

of arterial smooth muscle by acetylcholine. Nature 1980, 288(5789):373-376.

79. De Mey JG, Vanhoutte PM: Heterogeneous behavior of the canine arterial and

venous wall. Importance of the endothelium. Circ Res 1982, 51(4):439-447.

80. Ferguson MK, DeFilippi VJ: Nitric oxide and endothelium-dependent relaxation in

tracheobronchial lymph vessels. Microvasc Res 1994, 47(3):308-317.

81. Radhakrishnan K, Rockson SG: The clinical spectrum of lymphatic disease. Ann N Y

Acad Sci 2008, 1131:155-184.

Page 166: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

152

82. Warren AG, Brorson H, Borud LJ, Slavin SA: Lymphedema: a comprehensive

review. Ann Plast Surg 2007, 59(4):464-472.

83. Casley-Smith JR, Gaffney RM: Excess plasma proteins as a cause of chronic

inflammation and lymphodema: quantitative electron microscopy. J Pathol 1981,

133(3):243-272.

84. Gaffney RM, Casley-Smith JR: Excess plasma proteins as a cause of chronic

inflammation and lymphoedema: biochemical estimations. J Pathol 1981,

133(3):229-242.

85. Ryan TJ: Lymphatics and adipose tissue. Clin Dermatol 1995, 13(5):493-498.

86. Olszewski WL, Engeset A, Romaniuk A, Grzelak I, Ziolkowska A: Immune cells in

peripheral lymph and skin of patients with obstructive lymphedema. Lymphology

1990, 23(1):23-33.

87. Manduch M, Oliveira AM, Nascimento AG, Folpe AL: Massive localised

lymphoedema: a clinicopathological study of 22 cases and review of the literature. J

Clin Pathol 2009, 62(9):808-811.

88. Woo PC, Lum PN, Wong SS, Cheng VC, Yuen KY: Cellulitis complicating

lymphoedema. Eur J Clin Microbiol Infect Dis 2000, 19(4):294-297.

89. Macdonald JM: Wound healing and lymphedema: a new look at an old problem.

Ostomy Wound Manage 2001, 47(4):52-57.

90. Pereira de Godoy JM, Azoubel LM, Guerreiro Godoy Mde F: Erysipelas and

lymphangitis in patients undergoing lymphedema treatment after breast-cancer

therapy. Acta Dermatovenerol Alp Panonica Adriat 2009, 18(2):63-65.

Page 167: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

153

91. Damstra RJ, van Steensel MA, Boomsma JH, Nelemans P, Veraart JC: Erysipelas as a

sign of subclinical primary lymphoedema: a prospective quantitative scintigraphic

study of 40 patients with unilateral erysipelas of the leg. Br J Dermatol 2008,

158(6):1210-1215.

92. Shih YC, Xu Y, Cormier JN, Giordano S, Ridner SH, Buchholz TA, Perkins GH, Elting

LS: Incidence, treatment costs, and complications of lymphedema after breast

cancer among women of working age: a 2-year follow-up study. J Clin Oncol 2009,

27(12):2007-2014.

93. Pearce VJ, Mortimer PS: Hand dermatitis and lymphoedema. Br J Dermatol 2009,

161(1):177-180.

94. Chen HC, Gharb BB, Salgado CJ, Rampazzo A, Xu E, di Spilimbergo SS, Su S:

Elective amputation of the toes in severe lymphedema of the lower leg: rationale

and indications. Ann Plast Surg 2009, 63(2):193-197.

95. Watanabe T: Congenital penoscrotal lymphedema complicated by sepsis associated

with a streptococcal infection. Pediatr Emerg Care 2000, 16(6):423-425.

96. Person B, Addiss D, Bartholomew LK, Meijer C, Pou V, Gonzalvez G, Borne BV:

"Can it be that god does not remember me": a qualitative study on the

psychological distress, suffering, and coping of Dominican women with chronic

filarial lymphedema and elephantiasis of the leg. Health Care Women Int 2008,

29(4):349-365.

97. Gloviczki P: Principles of surgical treatment of chronic lymphoedema. Int Angiol

1999, 18(1):42-46.

Page 168: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

154

98. Rockson SG: Current concepts and future directions in the diagnosis and

management of lymphatic vascular disease. Vasc Med, 15(3):223-231.

99. Rockson SG: The unique biology of lymphatic edema. Lymphat Res Biol 2009,

7(2):97-100.

100. Rockson SG, Granger DN, Skeff KM, Chaite W: Lymphatic biology and disease: is it

being taught? Who is listening? Lymphat Res Biol 2004, 2(2):86-95.

101. Cliff S, Bedlow AJ, Stanton AW, Mortimer PS: An in vivo study of the

microlymphatics in psoriasis using fluorescence microlymphography. Br J

Dermatol 1999, 140(1):61-66.

102. Kovi J, Duong HD, Hoang CT: Ultrastructure of intestinal lymphatics in Crohn's

disease. Am J Clin Pathol 1981, 76(4):385-394.

103. Kaipainen A, Korhonen J, Mustonen T, van Hinsbergh VW, Fang GH, Dumont D,

Breitman M, Alitalo K: Expression of the fms-like tyrosine kinase 4 gene becomes

restricted to lymphatic endothelium during development. Proc Natl Acad Sci U S A

1995, 92(8):3566-3570.

104. Stacker SA, Hughes RA, Achen MG: Molecular targeting of lymphatics for therapy.

Curr Pharm Des 2004, 10(1):65-74.

105. Banerji S, Ni J, Wang SX, Clasper S, Su J, Tammi R, Jones M, Jackson DG: LYVE-1,

a new homologue of the CD44 glycoprotein, is a lymph-specific receptor for

hyaluronan. J Cell Biol 1999, 144(4):789-801.

106. Wigle JT, Oliver G: Prox1 function is required for the development of the murine

lymphatic system. Cell 1999, 98(6):769-778.

Page 169: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

155

107. Hirakawa S, Hong YK, Harvey N, Schacht V, Matsuda K, Libermann T, Detmar M:

Identification of vascular lineage-specific genes by transcriptional profiling of

isolated blood vascular and lymphatic endothelial cells. Am J Pathol 2003,

162(2):575-586.

108. Oppenheim JJ, Zachariae CO, Mukaida N, Matsushima K: Properties of the novel

proinflammatory supergene "intercrine" cytokine family. Annu Rev Immunol 1991,

9:617-648.

109. Schall TJ, Bacon K, Toy KJ, Goeddel DV: Selective attraction of monocytes and T

lymphocytes of the memory phenotype by cytokine RANTES. Nature 1990,

347(6294):669-671.

110. Ando T, Jordan P, Wang Y, Jennings MH, Harper MH, Houghton J, Elrod J, Alexander

JS: Homogeneity of mesothelial cells with lymphatic endothelium: expression of

lymphatic endothelial markers by mesothelial cells. Lymphat Res Biol 2005,

3(3):117-125.

111. Attout T, Hoerauf A, Denece G, Debrah AY, Marfo-Debrekyei Y, Boussinesq M, Wanji

S, Martinez V, Mand S, Adjei O et al: Lymphatic vascularisation and involvement of

Lyve-1+ macrophages in the human onchocerca nodule. PLoS One 2009,

4(12):e8234.

112. Blanchet MR, Maltby S, Haddon DJ, Merkens H, Zbytnuik L, McNagny KM: CD34

facilitates the development of allergic asthma. Blood 2007, 110(6):2005-2012.

113. McKenney JK, Weiss SW, Folpe AL: CD31 expression in intratumoral

macrophages: a potential diagnostic pitfall. Am J Surg Pathol 2001, 25(9):1167-

1173.

Page 170: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

156

114. Breiteneder-Geleff S, Soleiman A, Kowalski H, Horvat R, Amann G, Kriehuber E,

Diem K, Weninger W, Tschachler E, Alitalo K et al: Angiosarcomas express mixed

endothelial phenotypes of blood and lymphatic capillaries: podoplanin as a specific

marker for lymphatic endothelium. Am J Pathol 1999, 154(2):385-394.

115. Schacht V, Dadras SS, Johnson LA, Jackson DG, Hong YK, Detmar M: Up-regulation

of the lymphatic marker podoplanin, a mucin-type transmembrane glycoprotein,

in human squamous cell carcinomas and germ cell tumors. Am J Pathol 2005,

166(3):913-921.

116. Baluk P, McDonald DM: Markers for microscopic imaging of lymphangiogenesis

and angiogenesis. Ann N Y Acad Sci 2008, 1131:1-12.

117. Shibuya M, Claesson-Welsh L: Signal transduction by VEGF receptors in regulation

of angiogenesis and lymphangiogenesis. Exp Cell Res 2006, 312(5):549-560.

118. Lohela M, Bry M, Tammela T, Alitalo K: VEGFs and receptors involved in

angiogenesis versus lymphangiogenesis. Curr Opin Cell Biol 2009, 21(2):154-165.

119. Thurston G: Role of Angiopoietins and Tie receptor tyrosine kinases in angiogenesis

and lymphangiogenesis. Cell Tissue Res 2003, 314(1):61-68.

120. Woolf AS: Angiopoietins: vascular growth factors looking for roles in glomeruli.

Curr Opin Nephrol Hypertens, 19(1):20-25.

121. Morisada T, Oike Y, Yamada Y, Urano T, Akao M, Kubota Y, Maekawa H, Kimura Y,

Ohmura M, Miyamoto T et al: Angiopoietin-1 promotes LYVE-1-positive lymphatic

vessel formation. Blood 2005, 105(12):4649-4656.

122. Kim I, Kim JH, Moon SO, Kwak HJ, Kim NG, Koh GY: Angiopoietin-2 at high

concentration can enhance endothelial cell survival through the

Page 171: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

157

phosphatidylinositol 3'-kinase/Akt signal transduction pathway. Oncogene 2000,

19(39):4549-4552.

123. Teichert-Kuliszewska K, Maisonpierre PC, Jones N, Campbell AI, Master Z, Bendeck

MP, Alitalo K, Dumont DJ, Yancopoulos GD, Stewart DJ: Biological action of

angiopoietin-2 in a fibrin matrix model of angiogenesis is associated with activation

of Tie2. Cardiovasc Res 2001, 49(3):659-670.

124. Mochizuki Y, Nakamura T, Kanetake H, Kanda S: Angiopoietin 2 stimulates

migration and tube-like structure formation of murine brain capillary endothelial

cells through c-Fes and c-Fyn. J Cell Sci 2002, 115(Pt 1):175-183.

125. Harfouche R, Hussain SN: Signaling and regulation of endothelial cell survival by

angiopoietin-2. Am J Physiol Heart Circ Physiol 2006, 291(4):H1635-1645.

126. Carlson TR, Feng Y, Maisonpierre PC, Mrksich M, Morla AO: Direct cell adhesion to

the angiopoietins mediated by integrins. J Biol Chem 2001, 276(28):26516-26525.

127. Pitera JE, Woolf AS, Gale NW, Yancopoulos GD, Yuan HT: Dysmorphogenesis of

kidney cortical peritubular capillaries in angiopoietin-2-deficient mice. Am J Pathol

2004, 165(6):1895-1906.

128. Hackett SF, Wiegand S, Yancopoulos G, Campochiaro PA: Angiopoietin-2 plays an

important role in retinal angiogenesis. J Cell Physiol 2002, 192(2):182-187.

129. Gordon JA: Use of vanadate as protein-phosphotyrosine phosphatase inhibitor.

Methods Enzymol 1991, 201:477-482.

130. Leak LV, Jones M: Lymphatic endothelium isolation, characterization and long-

term culture. Anat Rec 1993, 236(4):641-652.

Page 172: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

158

131. Johnston MG, Walker MA: Lymphatic endothelial and smooth-muscle cells in tissue

culture. In Vitro 1984, 20(7):566-572.

132. Tammela T, Petrova TV, Alitalo K: Molecular lymphangiogenesis: new players.

Trends Cell Biol 2005, 15(8):434-441.

133. Herzog Y, Kalcheim C, Kahane N, Reshef R, Neufeld G: Differential expression of

neuropilin-1 and neuropilin-2 in arteries and veins. Mech Dev 2001, 109(1):115-119.

134. Barton WA, Tzvetkova-Robev D, Miranda EP, Kolev MV, Rajashankar KR, Himanen

JP, Nikolov DB: Crystal structures of the Tie2 receptor ectodomain and the

angiopoietin-2-Tie2 complex. Nat Struct Mol Biol 2006, 13(6):524-532.

135. Bogdanovic E, Nguyen VP, Dumont DJ: Activation of Tie2 by angiopoietin-1 and

angiopoietin-2 results in their release and receptor internalization. J Cell Sci 2006,

119(Pt 17):3551-3560.

136. Yoon MJ, Cho CH, Lee CS, Jang IH, Ryu SH, Koh GY: Localization of Tie2 and

phospholipase D in endothelial caveolae is involved in angiopoietin-1-induced

MEK/ERK phosphorylation and migration in endothelial cells. Biochem Biophys

Res Commun 2003, 308(1):101-105.

137. Kanda S, Miyata Y, Mochizuki Y, Matsuyama T, Kanetake H: Angiopoietin 1 is

mitogenic for cultured endothelial cells. Cancer Res 2005, 65(15):6820-6827.

138. Hu B, Jarzynka MJ, Guo P, Imanishi Y, Schlaepfer DD, Cheng SY: Angiopoietin 2

induces glioma cell invasion by stimulating matrix metalloprotease 2 expression

through the alphavbeta1 integrin and focal adhesion kinase signaling pathway.

Cancer Res 2006, 66(2):775-783.

Page 173: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

159

139. Murray BW, Padrique ES, Pinko C, McTigue MA: Mechanistic effects of

autophosphorylation on receptor tyrosine kinase catalysis: enzymatic

characterization of Tie2 and phospho-Tie2. Biochemistry 2001, 40(34):10243-10253.

140. Thompson JD, Higgins DG, Gibson TJ: CLUSTAL W: improving the sensitivity of

progressive multiple sequence alignment through sequence weighting, position-

specific gap penalties and weight matrix choice. Nucleic Acids Res 1994,

22(22):4673-4680.

141. Witzenbichler B, Maisonpierre PC, Jones P, Yancopoulos GD, Isner JM: Chemotactic

properties of angiopoietin-1 and -2, ligands for the endothelial-specific receptor

tyrosine kinase Tie2. J Biol Chem 1998, 273(29):18514-18521.

142. Liu W, Reinmuth N, Stoeltzing O, Parikh AA, Fan F, Ahmad SA, Jung YD, Ellis LM:

Antiangiogenic therapy targeting factors that enhance endothelial cell survival.

Semin Oncol 2002, 29(3 Suppl 11):96-103.

143. Kontos CD, Stauffer TP, Yang WP, York JD, Huang L, Blanar MA, Meyer T, Peters

KG: Tyrosine 1101 of Tie2 is the major site of association of p85 and is required for

activation of phosphatidylinositol 3-kinase and Akt. Mol Cell Biol 1998, 18(7):4131-

4140.

144. Jones N, Master Z, Jones J, Bouchard D, Gunji Y, Sasaki H, Daly R, Alitalo K, Dumont

DJ: Identification of Tek/Tie2 binding partners. Binding to a multifunctional

docking site mediates cell survival and migration. J Biol Chem 1999, 274(43):30896-

30905.

145. Kwak HJ, So JN, Lee SJ, Kim I, Koh GY: Angiopoietin-1 is an apoptosis survival

factor for endothelial cells. FEBS Lett 1999, 448(2-3):249-253.

Page 174: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

160

146. Hayes AJ, Huang WQ, Mallah J, Yang D, Lippman ME, Li LY: Angiopoietin-1 and its

receptor Tie-2 participate in the regulation of capillary-like tubule formation and

survival of endothelial cells. Microvasc Res 1999, 58(3):224-237.

147. Kim I, Kim HG, So JN, Kim JH, Kwak HJ, Koh GY: Angiopoietin-1 regulates

endothelial cell survival through the phosphatidylinositol 3'-Kinase/Akt signal

transduction pathway. Circ Res 2000, 86(1):24-29.

148. Papapetropoulos A, Fulton D, Mahboubi K, Kalb RG, O'Connor DS, Li F, Altieri DC,

Sessa WC: Angiopoietin-1 inhibits endothelial cell apoptosis via the Akt/survivin

pathway. J Biol Chem 2000, 275(13):9102-9105.

149. Babaei S, Teichert-Kuliszewska K, Zhang Q, Jones N, Dumont DJ, Stewart DJ:

Angiogenic actions of angiopoietin-1 require endothelium-derived nitric oxide. Am

J Pathol 2003, 162(6):1927-1936.

150. Murakami T, Takagi H, Suzuma K, Suzuma I, Ohashi H, Watanabe D, Ojima T,

Suganami E, Kurimoto M, Kaneto H et al: Angiopoietin-1 attenuates H2O2-induced

SEK1/JNK phosphorylation through the phosphatidylinositol 3-kinase/Akt

pathway in vascular endothelial cells. J Biol Chem 2005, 280(36):31841-31849.

151. Zagzag D, Hooper A, Friedlander DR, Chan W, Holash J, Wiegand SJ, Yancopoulos

GD, Grumet M: In situ expression of angiopoietins in astrocytomas identifies

angiopoietin-2 as an early marker of tumor angiogenesis. Exp Neurol 1999,

159(2):391-400.

152. Cohen B, Barkan D, Levy Y, Goldberg I, Fridman E, Kopolovic J, Rubinstein M:

Leptin induces angiopoietin-2 expression in adipose tissues. J Biol Chem 2001,

276(11):7697-7700.

Page 175: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

161

153. Nag S, Papneja T, Venugopalan R, Stewart DJ: Increased angiopoietin2 expression is

associated with endothelial apoptosis and blood-brain barrier breakdown. Lab

Invest 2005, 85(10):1189-1198.

154. Daly C, Pasnikowski E, Burova E, Wong V, Aldrich TH, Griffiths J, Ioffe E, Daly TJ,

Fandl JP, Papadopoulos N et al: Angiopoietin-2 functions as an autocrine protective

factor in stressed endothelial cells. Proc Natl Acad Sci U S A 2006, 103(42):15491-

15496.

155. Gnepp DR, Chandler W: Tissue culture of human and canine thoracic duct

endothelium. In Vitro Cell Dev Biol 1985, 21(4):200-206.

156. Bowman CA, Witte MH, Witte CL, Way DL, Nagle RB, Copeland JG, Daschbach CC:

Cystic hygroma reconsidered: hamartoma or neoplasm? Primary culture of an

endothelial cell line from a massive cervicomediastinal hygroma with bony

lymphangiomatosis. Lymphology 1984, 17(1):15-22.

157. Witte MH, Witte CL: Lymphatics and blood vessels, lymphangiogenesis and

hemangiogenesis: from cell biology to clinical medicine. Lymphology 1987,

20(4):257-266.

158. Sauter B, Foedinger D, Sterniczky B, Wolff K, Rappersberger K: Immunoelectron

microscopic characterization of human dermal lymphatic microvascular

endothelial cells. Differential expression of CD31, CD34, and type IV collagen with

lymphatic endothelial cells vs blood capillary endothelial cells in normal human

skin, lymphangioma, and hemangioma in situ. J Histochem Cytochem 1998,

46(2):165-176.

Page 176: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

162

159. Chi JT, Chang HY, Haraldsen G, Jahnsen FL, Troyanskaya OG, Chang DS, Wang Z,

Rockson SG, van de Rijn M, Botstein D et al: Endothelial cell diversity revealed by

global expression profiling. Proc Natl Acad Sci U S A 2003, 100(19):10623-10628.

160. Sironi M, Conti A, Bernasconi S, Fra AM, Pasqualini F, Nebuloni M, Lauri E, De

Bortoli M, Mantovani A, Dejana E et al: Generation and characterization of a mouse

lymphatic endothelial cell line. Cell Tissue Res 2006, 325(1):91-100.

161. Bianchi L, Lorenzoni P, Bini L, Weber E, Tani C, Rossi A, Agliano M, Pallini V,

Sacchi G: Protein expression profiles of Bos taurus blood and lymphatic vessel

endothelial cells. Proteomics 2007, 7(10):1600-1614.

162. Wick N, Saharinen P, Saharinen J, Gurnhofer E, Steiner CW, Raab I, Stokic D,

Giovanoli P, Buchsbaum S, Burchard A et al: Transcriptomal comparison of human

dermal lymphatic endothelial cells ex vivo and in vitro. Physiol Genomics 2007,

28(2):179-192.

163. Amatschek S, Kriehuber E, Bauer W, Reininger B, Meraner P, Wolpl A, Schweifer N,

Haslinger C, Stingl G, Maurer D: Blood and lymphatic endothelial cell-specific

differentiation programs are stringently controlled by the tissue environment.

Blood 2007, 109(11):4777-4785.

164. Nelson GM, Padera TP, Garkavtsev I, Shioda T, Jain RK: Differential gene expression

of primary cultured lymphatic and blood vascular endothelial cells. Neoplasia 2007,

9(12):1038-1045.

165. Kawai Y, Minami T, Fujimori M, Hosaka K, Mizuno R, Ikomi F, Kodama T, Ohhashi

T: Characterization and microarray analysis of genes in human lymphatic

Page 177: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

163

endothelial cells from patients with breast cancer. Lymphat Res Biol 2007, 5(2):115-

126.

166. Roesli C, Mumprecht V, Neri D, Detmar M: Identification of the surface-accessible,

lineage-specific vascular proteome by two-dimensional peptide mapping. FASEB J

2008, 22(6):1933-1944.

167. Nguyen VP, Chen SH, Trinh J, Kim H, Coomber BL, Dumont DJ: Differential

response of lymphatic, venous and arterial endothelial cells to angiopoietin-1 and

angiopoietin-2. BMC Cell Biol 2007, 8:10.

168. Veikkola T, Alitalo K: Dual role of Ang2 in postnatal angiogenesis and

lymphangiogenesis. Dev Cell 2002, 3(3):302-304.

169. Weber E, Lorenzoni P, Lozzi G, Sacchi G: Cytochemical differentiation between

blood and lymphatic endothelium: bovine blood and lymphatic large vessels and

endothelial cells in culture. J Histochem Cytochem 1994, 42(8):1109-1115.

170. Veikkola T, Lohela M, Ikenberg K, Makinen T, Korff T, Saaristo A, Petrova T, Jeltsch

M, Augustin HG, Alitalo K: Intrinsic versus microenvironmental regulation of

lymphatic endothelial cell phenotype and function. FASEB J 2003, 17(14):2006-

2013.

171. Ando T, Jordan P, Joh T, Wang Y, Jennings MH, Houghton J, Alexander JS: Isolation

and characterization of a novel mouse lymphatic endothelial cell line: SV-LEC.

Lymphat Res Biol 2005, 3(3):105-115.

172. Pruitt KD, Tatusova T, Maglott DR: NCBI reference sequences (RefSeq): a curated

non-redundant sequence database of genomes, transcripts and proteins. Nucleic

Acids Res 2007, 35(Database issue):D61-65.

Page 178: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

164

173. Ashburner M, Ball CA, Blake JA, Botstein D, Butler H, Cherry JM, Davis AP, Dolinski

K, Dwight SS, Eppig JT et al: Gene ontology: tool for the unification of biology. The

Gene Ontology Consortium. Nat Genet 2000, 25(1):25-29.

174. Harel A, Inger A, Stelzer G, Strichman-Almashanu L, Dalah I, Safran M, Lancet D:

GIFtS: annotation landscape analysis with GeneCards. BMC Bioinformatics 2009,

10:348.

175. Oliver G, Srinivasan RS: Lymphatic vasculature development: current concepts.

Ann N Y Acad Sci 2008, 1131:75-81.

176. Qian WJ, Jacobs JM, Liu T, Camp DG, 2nd, Smith RD: Advances and challenges in

liquid chromatography-mass spectrometry-based proteomics profiling for clinical

applications. Mol Cell Proteomics 2006, 5(10):1727-1744.

177. Wick G, Knoflach M, Kind M, Henderson B, Bernhard D: Heat shock proteins and

stress in atherosclerosis. Autoimmun Rev 2004, 3 Suppl 1:S30-31.

178. Wagner M, Hermanns I, Bittinger F, Kirkpatrick CJ: Induction of stress proteins in

human endothelial cells by heavy metal ions and heat shock. Am J Physiol 1999,

277(5 Pt 1):L1026-1033.

179. Kalmar B, Greensmith L: Induction of heat shock proteins for protection against

oxidative stress. Adv Drug Deliv Rev 2009.

180. Preville X, Salvemini F, Giraud S, Chaufour S, Paul C, Stepien G, Ursini MV, Arrigo

AP: Mammalian small stress proteins protect against oxidative stress through their

ability to increase glucose-6-phosphate dehydrogenase activity and by maintaining

optimal cellular detoxifying machinery. Exp Cell Res 1999, 247(1):61-78.

Page 179: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

165

181. Kim YH, Park EJ, Han ST, Park JW, Kwon TK: Arsenic trioxide induces Hsp70

expression via reactive oxygen species and JNK pathway in MDA231 cells. Life Sci

2005, 77(22):2783-2793.

182. Franklin TB, Krueger-Naug AM, Clarke DB, Arrigo AP, Currie RW: The role of heat

shock proteins Hsp70 and Hsp27 in cellular protection of the central nervous

system. Int J Hyperthermia 2005, 21(5):379-392.

183. Steurer J, Hoffmann U, Dur P, Russi E, Vetter W: Changes in arterial and

transcutaneous oxygen and carbon dioxide tensions during and after voluntary

hyperventilation. Respiration 1997, 64(3):200-205.

184. Ho HY, Cheng ML, Cheng PF, Chiu DT: Low oxygen tension alleviates oxidative

damage and delays cellular senescence in G6PD-deficient cells. Free Radic Res

2007, 41(5):571-579.

185. Rhee SG, Chae HZ, Kim K: Peroxiredoxins: a historical overview and speculative

preview of novel mechanisms and emerging concepts in cell signaling. Free Radic

Biol Med 2005, 38(12):1543-1552.

186. Shang F, Gong X, Taylor A: Activity of ubiquitin-dependent pathway in response to

oxidative stress. Ubiquitin-activating enzyme is transiently up-regulated. J Biol

Chem 1997, 272(37):23086-23093.

187. Atkuri KR, Herzenberg LA: Culturing at atmospheric oxygen levels impacts

lymphocyte function. Proc Natl Acad Sci U S A 2005, 102(10):3756-3759.

188. Atkuri KR, Herzenberg LA, Niemi AK, Cowan T: Importance of culturing primary

lymphocytes at physiological oxygen levels. Proc Natl Acad Sci U S A 2007,

104(11):4547-4552.

Page 180: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

166

189. Ji RC: Lymphatic endothelial cells, lymphedematous lymphangiogenesis, and

molecular control of edema formation. Lymphat Res Biol 2008, 6(3-4):123-137.

190. Jurisic G, Detmar M: Lymphatic endothelium in health and disease. Cell Tissue Res

2009, 335(1):97-108.

191. Liersch R, Biermann C, Mesters RM, Berdel WE: Lymphangiogenesis in cancer:

current perspectives. Recent Results Cancer Res, 180:115-135.

192. Dorrell C, Abraham SL, Lanxon-Cookson KM, Canaday PS, Streeter PR, Grompe M:

Isolation of major pancreatic cell types and long-term culture-initiating cells using

novel human surface markers. Stem Cell Res 2008, 1(3):183-194.

193. Mernaugh RL, Yan H, Chen D, Edl J, Hanley G, Pozzi A, Zent R: Production and

characterization of mouse ureteric bud cell-specific rat hybridoma antibodies

utilizing subtractive immunization and high-throughput screening. J Immunol

Methods 2005, 306(1-2):115-127.

194. Ning Y, Wang Y, Li Y, Hong Y, Peng D, Liu Y, Wang J, Hao W, Tian X, Wu F et al:

An alternative strategy for high throughput generation and characterization of

monoclonal antibodies against human plasma proteins using fractionated native

proteins as immunogens. Proteomics 2006, 6(2):438-448.

195. Khan SA, Suryawanshi AR, Ranpura SA, Jadhav SV, Khole VV: Identification of

novel immunodominant epididymal sperm proteins using combinatorial approach.

Reproduction 2009, 138(1):81-93.

196. Hooper JD, Zijlstra A, Aimes RT, Liang H, Claassen GF, Tarin D, Testa JE, Quigley JP:

Subtractive immunization using highly metastatic human tumor cells identifies

Page 181: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

167

SIMA135/CDCP1, a 135 kDa cell surface phosphorylated glycoprotein antigen.

Oncogene 2003, 22(12):1783-1794.

197. Boukerche H, Baril P, Tabone E, Berard F, Sanhadji K, Balme B, Wolf F, Perrot H,

Thomas L: A new Mr 55,000 surface protein implicated in melanoma progression:

association with a metastatic phenotype. Cancer Res 2000, 60(20):5848-5856.

198. Testa JE, Brooks PC, Lin JM, Quigley JP: Eukaryotic expression cloning with an

antimetastatic monoclonal antibody identifies a tetraspanin (PETA-3/CD151) as an

effector of human tumor cell migration and metastasis. Cancer Res 1999,

59(15):3812-3820.

199. Rasmussen N, Ditzel HJ: Scanning the cell surface proteome of cancer cells and

identification of metastasis-associated proteins using a subtractive immunization

strategy. J Proteome Res 2009, 8(11):5048-5059.

200. Sleeman JP, Kim U, LePendu J, Howells N, Coquerelle T, Ponta H, Herrlich P:

Inhibition of MT-450 rat mammary tumour growth by antibodies recognising

subtypes of blood group antigen B. Oncogene 1999, 18(31):4485-4494.

201. Brooks PC, Lin JM, French DL, Quigley JP: Subtractive immunization yields

monoclonal antibodies that specifically inhibit metastasis. J Cell Biol 1993,

122(6):1351-1359.

202. Trefzer U, Rietz N, Chen Y, Audring H, Herberth G, Siegel P, Reinke S, Koniger P, Wu

S, Ma J et al: SM5-1: a new monoclonal antibody which is highly sensitive and

specific for melanocytic lesions. Arch Dermatol Res 2000, 292(12):583-589.

Page 182: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

168

203. Williams CV, Stechmann CL, McLoon SC: Subtractive immunization techniques for

the production of monoclonal antibodies to rare antigens. Biotechniques 1992,

12(6):842-847.

204. Sleister HM, Rao AG: Strategies to generate antibodies capable of distinguishing

between proteins with >90% amino acid identity. J Immunol Methods 2001, 252(1-

2):121-129.

205. Sleister HM, Rao AG: Subtractive immunization: a tool for the generation of

discriminatory antibodies to proteins of similar sequence. J Immunol Methods 2002,

261(1-2):213-220.

206. Zijlstra A, Testa JE, Quigley JP: Targeting the proteome/epitome, implementation of

subtractive immunization. Biochem Biophys Res Commun 2003, 303(3):733-744.

207. Nelson PN, Reynolds GM, Waldron EE, Ward E, Giannopoulos K, Murray PG:

Monoclonal antibodies. Mol Pathol 2000, 53(3):111-117.

208. Song KS, Scherer PE, Tang Z, Okamoto T, Li S, Chafel M, Chu C, Kohtz DS, Lisanti

MP: Expression of caveolin-3 in skeletal, cardiac, and smooth muscle cells.

Caveolin-3 is a component of the sarcolemma and co-fractionates with dystrophin

and dystrophin-associated glycoproteins. J Biol Chem 1996, 271(25):15160-15165.

209. Dancik V, Addona TA, Clauser KR, Vath JE, Pevzner PA: De novo peptide

sequencing via tandem mass spectrometry. J Comput Biol 1999, 6(3-4):327-342.

210. Wang HU, Chen ZF, Anderson DJ: Molecular distinction and angiogenic interaction

between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-

B4. Cell 1998, 93(5):741-753.

Page 183: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

169

211. Srinivasan RS, Dillard ME, Lagutin OV, Lin FJ, Tsai S, Tsai MJ, Samokhvalov IM,

Oliver G: Lineage tracing demonstrates the venous origin of the mammalian

lymphatic vasculature. Genes Dev 2007, 21(19):2422-2432.

212. Song KS, Li S, Okamoto T, Quilliam LA, Sargiacomo M, Lisanti MP: Co-purification

and direct interaction of Ras with caveolin, an integral membrane protein of

caveolae microdomains. Detergent-free purification of caveolae microdomains. J

Biol Chem 1996, 271(16):9690-9697.

213. Foster LJ, De Hoog CL, Mann M: Unbiased quantitative proteomics of lipid rafts

reveals high specificity for signaling factors. Proc Natl Acad Sci U S A 2003,

100(10):5813-5818.

214. Mirza SP, Olivier M: Methods and approaches for the comprehensive

characterization and quantification of cellular proteomes using mass spectrometry.

Physiol Genomics 2008, 33(1):3-11.

215. Shevchenko A, Sunyaev S, Loboda A, Bork P, Ens W, Standing KG: Charting the

proteomes of organisms with unsequenced genomes by MALDI-quadrupole time-

of-flight mass spectrometry and BLAST homology searching. Anal Chem 2001,

73(9):1917-1926.

216. Dagenais SL, Hartsough RL, Erickson RP, Witte MH, Butler MG, Glover TW: Foxc2 is

expressed in developing lymphatic vessels and other tissues associated with

lymphedema-distichiasis syndrome. Gene Expr Patterns 2004, 4(6):611-619.

217. Fujii T, Zen Y, Sato Y, Sasaki M, Enomae M, Minato H, Masuda S, Uehara T,

Katsuyama T, Nakanuma Y: Podoplanin is a useful diagnostic marker for epithelioid

hemangioendothelioma of the liver. Mod Pathol 2008, 21(2):125-130.

Page 184: LYMPHATIC ENDOTHELIAL CELLS AS...ii COMPARATIVE APPROACHES TO CHARACTERIZATION OF LYMPHATIC ENDOTHELIAL CELLS AS PHENOTYPICALLY DISTINCT FROM BLOOD ENDOTHELIAL CELLS Victoria P. K

170

218. Becker J, Wang B, Pavlakovic H, Buttler K, Wilting J: Homeobox Transcription

Factor Prox1 in Sympathetic Ganglia of Vertebrate Embryos: Correlation With

Human Stage 4s Neuroblastoma. Pediatr Res.

219. Wicker N, Carles A, Mills IG, Wolf M, Veerakumarasivam A, Edgren H, Boileau F,

Wasylyk B, Schalken JA, Neal DE et al: A new look towards BAC-based array CGH

through a comprehensive comparison with oligo-based array CGH. BMC Genomics

2007, 8:84.

220. Troxler RF, Iontcheva I, Oppenheim FG, Nunes DP, Offner GD: Molecular

characterization of a major high molecular weight mucin from human sublingual

gland. Glycobiology 1997, 7(7):965-973.

221. Levine MJ, Reddy MS, Tabak LA, Loomis RE, Bergey EJ, Jones PC, Cohen RE,

Stinson MW, Al-Hashimi I: Structural aspects of salivary glycoproteins. J Dent Res

1987, 66(2):436-441.

222. Ezaki T, Kuwahara K, Morikawa S, Shimizu K, Sakaguchi N, Matsushima K, Matsuno

K: Production of two novel monoclonal antibodies that distinguish mouse

lymphatic and blood vascular endothelial cells. Anat Embryol (Berl) 2006,

211(5):379-393.

223. Ball LE, Oatis JE, Jr., Dharmasiri K, Busman M, Wang J, Cowden LB, Galijatovic A,

Chen N, Crouch RK, Knapp DR: Mass spectrometric analysis of integral membrane

proteins: application to complete mapping of bacteriorhodopsins and rhodopsin.

Protein Sci 1998, 7(3):758-764.

224. Porter CJ, Charman WN: Transport and absorption of drugs via the lymphatic

system. Adv Drug Deliv Rev 2001, 50(1-2):1-2.