+ All Categories
Home > Documents > © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz...

© 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz...

Date post: 15-Aug-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
113
GENERATION OF DNA APTAMERS FOR HEPATOCELLULAR CARCINOMA EXOSOMES By SENA CANSIZ A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2016
Transcript
Page 1: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

GENERATION OF DNA APTAMERS FOR HEPATOCELLULAR CARCINOMA EXOSOMES

By

SENA CANSIZ

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2016

Page 2: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

© 2016 Sena Cansiz

Page 3: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

To my beloved parents Nevin & Mehmet and my better half Mert

“So long, and thanks for all the fish”

Page 4: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

4

ACKNOWLEDGMENTS

This dissertation could not have been completed without the great support that I

have received from so many people over the years. I wish to offer my most heartfelt

thanks to the following people.

First and for most, I would like to gratefully and sincerely thank my advisor, Dr.

Weihong Tan for his support, encouragement, guidance and understanding. This path

was not easy, but he always tried to show me the right way of not only doing research

but the life itself. He is one of the most hard-working people that I have ever known and

this accounts for the fact that success does not come by luck, it only comes from hard

work. I am deeply indebted to him for his support and valuable guidance. I also want to

express my gratitude to Ms. Weijun Chen for being “the mother of the group” and for her

help in my molecular biology experiments. If she did not share the SW28 rotor with us,

exosome projects would have been just a dream.

I would also like to express my honest appreciation to my committee members,

for their support and constructive criticism. Dr. Polfer was always the first to respond to

my emails, and I really appreciate his cooperative manner. I learned a lot from Dr.

Fanucci’s questions, critics and feedbacks during my proposal, departmental seminar

and individual meetings. I admire her scientific thinking and consider myself lucky to

have her as my committee member. Similarly, Dr. Horenstein is one of the best scientist

whom I have ever met. There is no one time in seminars that she did not impress me

with the question she asked to the speaker. I don’t remember how many times that I

wished to be as a good scientist as she is. Finally, Dr. Schultz is the best committee

member and perhaps the advisor that one can ever ask. He was never without an idea

or a kind word, and I always appreciated both.

Page 5: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

5

Special thanks go to Dr. Ben Smith and Ms. Lori Clark for helping me to deal with

administrative problems, to Dr. Jim Horwath for being the best and most colorful

teaching adviser and to Dr. Katherine Williams (magician of words) for making my

manuscript readable.

It would be unfair not to acknowledge Dr. Tahir Bayrac and Dr. Basri Gulbakan,

for their valuable mentoring. I could not be here without their help. I would also like to

thank to Dr. Meghan Altman for letting me be a part of her project and Dr. Kwame Sefah

for his valuable guidance. I wish, I had the opportunity to spend more time with them so

that I could learn more from them.

I owe a big gratitude to my “lab sisters”, Eliza, Xiangling, Carole and my “lab

brothers”: Liqin, Cheng, Sam for their valuable friendship, support and encouragement.

A special thanks go to all past and current members of Tan Family, especially to Dalia,

Dimitri, Diane, Ismail, Emir, Tao, Mingxu, Da, I-Ting, Stefanie, Weijia, Yanyue, Yuan

Wu, Kimberly, Yuan Liu, Yian, Sai, Shuo, Xigao, Liping, Juan, Dr. Jiang, Dr. Liu, Long,

Danny, Xiaoshu, Xiaowei and many others for their sincere friendship. I would also like

to thank to Dr. Gonca Yildirim, Dr. Nail Tanrioven and Eray Caliskan for their sincere

help.

Above all, I would like to acknowledge the tremendous sacrifices that my parents

made to keep me going. Words are not enough to express my gratitude to them, without

whom I could not make this far. I deeply appreciate their endless love and support.

Last but not least, I would like to thank my rock, my loving husband, Mert. He is

the best colleague, friend, and lover that one can ask and my biggest luck in this life.

Page 6: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

6

His encouragement made me through many long nights of lab work and many stressful

days. I will be forever in his debt. Counting down the days until we reunite.

Page 7: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

7

TABLE OF CONTENTS page

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

LIST OF TABLES .......................................................................................................... 10

LIST OF FIGURES ........................................................................................................ 11

LIST OF ABBREVIATIONS ........................................................................................... 13

ABSTRACT ................................................................................................................... 15

CHAPTER

1 INTRODUCTION .................................................................................................... 17

Cancer .................................................................................................................... 17 Extracellular Vesicles: Message in a bottle ............................................................. 18

History .............................................................................................................. 19 Classification of Extracellular Vesicles ............................................................. 20

Exosomes ............................................................................................................... 21 Protein Composition of Exosomes ................................................................... 21 Biogenesis and Release of Exosomes ............................................................. 22

Aptamers ................................................................................................................ 24 SELEX .............................................................................................................. 24 Cell-SELEX ...................................................................................................... 25

Overview of Dissertation ......................................................................................... 26

2 ISOLATION AND CHARACTERIZATION OF EXOSOMES FROM DIFFERENT CELL LINES ........................................................................................................... 31

Background and Significance ................................................................................. 31 Materials and Methods............................................................................................ 32

General Materials ............................................................................................. 32 Cell Lines and Culturing ................................................................................... 32 Exosome Isolation from Cells ........................................................................... 32 Exosome Isolation from Whole Human Blood .................................................. 33 Nanoparticle-Tracking Analysis (NTA) ............................................................. 34 Western Blot Analysis ...................................................................................... 34 Exosome-Bead Attachment .............................................................................. 35 Flow Cytometer Analysis of Exosome Bound Beads ........................................ 35 Immunogold Labelling TEM .............................................................................. 36

Results and Discussion........................................................................................... 37 Exosome Isolation from Cell Culture Media and Blood: Ultracentrifugation

and Ultrafiltration ........................................................................................... 37

Page 8: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

8

Size and Concentration Analysis of Particles by Nanoparticle-Tracking Assay ............................................................................................................ 38

Measuring the Protein Content of Exosomes Using the BCA Assay ................ 39 Flow Cytometric Detection of Exosomes .......................................................... 40

Concluding Remarks............................................................................................... 40

3 DEVELOPMENT OF EV-SELEX METHODOLOGY AND SELECTION OF DNA APTAMERS AGAINST HEPATOCELLULAR CARCINOMA EXOSOMES ............. 49

Background and Significance ................................................................................. 49 Materials and Methods............................................................................................ 51

General Materials ............................................................................................. 51 Synthesis and Purification of Six Nucleotides (GACTZP) Libraries .................. 51 Cell Culture and Buffers ................................................................................... 52 Exosome Extraction from Hep G2 Cells for Positive Selection ......................... 52 Exosome Isolation from Whole Human Blood for Negative Selection .............. 53 Exosome-Bead Attachment .............................................................................. 53 Detailed Experimental Flow of EV-SELEX ....................................................... 54

Incubation step ........................................................................................... 54 PCR cycle optimization and amplification step........................................... 55 Preparation of single-stranded DNA .......................................................... 55 Monitoring of the pool enrichment .............................................................. 56

Results .................................................................................................................... 57 EV-SELEX Method and Generation of DNA Aptamers against

Hepatocellular Carcinoma Exosomes ........................................................... 57 Deep sequencing of GACTZP DNA survivors using Next Generation

sequencing technology. ................................................................................ 59 Discussion and Conclusion ..................................................................................... 60

4 IDENTIFICATION OF DNA APTAMER ANALOGS IN GENOMIC DNA ................. 69

Introductory Remarks.............................................................................................. 69 Protein Tyrosine Kinase 7 ................................................................................ 69 Wnt Signaling ................................................................................................... 70

Background and Significance ................................................................................. 71 Results .................................................................................................................... 74

Sequence Similarity between Different Aptamers ............................................ 74 Competition Experiments ................................................................................. 75 BLAST of the Consensus Sequence against the Human Genome .................. 75 Investigation of the Interaction of PTK7 with DIXDC1b DNA ............................ 76

Discussion and Conclusion ..................................................................................... 77 Materials and Methods............................................................................................ 80

Buffers and Cell Culture ................................................................................... 80 DNA Sequences ............................................................................................... 81 Bioinformatics ................................................................................................... 81 Significance Simulations................................................................................... 82 Competition Assays .......................................................................................... 83

Page 9: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

9

Western Blot ..................................................................................................... 83 Gel Shift Assay (EMSA) ................................................................................... 83

5 CONCLUSION AND FUTURE DIRECTIONS ......................................................... 92

Summary and Conclusion ....................................................................................... 92 Future Directions .................................................................................................... 93

APPENDIX

A COMPLEX TARGET SELEX DNA APTAMER DATABASE ................................... 95

B PREDICTED SECONDARY STRUCTURES OF PTK7 APTAMERS ................... 102

LIST OF REFERENCES ............................................................................................. 103

BIOGRAPHICAL SKETCH .......................................................................................... 113

Page 10: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

10

LIST OF TABLES

Table page 3-1 Sequences used for EV-SELEX ......................................................................... 63

3-2 Summary of EV-SELEX process ........................................................................ 63

3-3 Compendium of the aptamer candidates selected by EV-SELEX. ..................... 64

4-1 PTK7 aptamer sequences with their identical nucleotides .................................. 85

4-2 BLAST hits 14/15nt identity for consensus sequence ......................................... 86

4-3 Aptamers share sequence similarity with DIXDC1b DNA sequence .................. 87

A-1 Complex Target SELEX DNA Aptamer Database .............................................. 96

Page 11: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

11

LIST OF FIGURES

Figure page 1-1 Exocytosis of MVEs releases exosomes containing transferrin receptor ............ 27

1-2 Histogram of exosomal studies over the past 40 years ...................................... 28

1-3 Schematic for extracellular vesicle trafficking. .................................................... 29

1-4 Biogenesis of extracellular vesicles and their interactions with recipient cells. ... 30

2-1 Flow chart for the exosome purification procedure based on differential ultracentrifugation and ultrafiltration .................................................................... 41

2-2 BCA experiment schematic. ............................................................................... 42

2-3 Schematic of optical configuration used in NTA. ................................................ 43

2-4 Size distribution from NTA measurements ......................................................... 44

2-5 Characterization of exosome preparations from different cell lines or human whole blood by western blot ............................................................................... 45

2-6 Schematic of capture and fluorescent analysis of extracellular vesicles. ............ 46

2-7 Binding test of different Hep G2 aptamers with blood exosomes by flow cytometry ............................................................................................................ 47

2-8 Validation of isolated exosomes and interaction between aptamer LZH8 and HepG2 exosomes ............................................................................................... 48

3-1 Schematic of EV-SELEX with both positive and negative selections.................. 65

3-2 PCR applications of EV-SELEX .......................................................................... 66

3-3 Verification of the enrichment of the library in binding sequences after 6 rounds. ............................................................................................................... 67

3-4 Monitoring the progress of EV-SELEX using flow cytometer .............................. 68

4-1 Simplified schemes showing the main WNT pathways directed by specific WNT, Frizzled and WNT co-receptor interactions .............................................. 88

4-2 Competition studies between different aptamers for PTK7 ................................. 89

4-3 Interaction of PTK7 with DIXDC1b DNA. ............................................................ 90

Page 12: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

12

4-4 Confocal immunocytochemistry image of HeLa cells co-stained for PTK7 with sgc8-TMR ........................................................................................................... 90

4-5 Electrophoretic mobility shift assay (EMSA) for the ds DNA surrounding the consensus region on DIXDC1b .......................................................................... 91

B-1 Predicted secondary structures for PTK7 aptamers. ........................................ 102

Page 13: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

13

LIST OF ABBREVIATIONS

5’UTR 5’-untranslated region

AMA Ammonium hydroxide: methylamine 1:1

ATCC American Type Culture Collection

BB Binding Buffer

BLAST Basic local alignment search tool

bp Base pair

BSA Bovine serum albumin

CEM Human T-Cell Acute Lymphoblastic Leukemia cell line

CLUSTAL Multiple sequence alignment computer program

DBU 1,8-Diazabicyclo[5.4.0]undec-7-ene

DIXDC1 DIX domain containing 1 protein

DMEM Dulbecco’s modified eagle media

DNA Deoxyribonucleic acid

dsDNA Double-stranded DNA

ESPRIT Bioinformatics algorithm for sequence alignment

EVs Extracellular vesicles

HEK293 Human embryonic kidney cell line

HeLa Henrietta Lacks's cervical cancer cell line

HPLC High pressure liquid chromatography

HRP Horseradish peroxidase

kDa KiloDalton

MV Microvesicle

PCR Polymerase chain reaction

Page 14: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

14

RNA Ribonucleic acid

SELEX Systematic Evolution of Ligands by EXponential enrichment

ssDNA Single stranded deoxyribonucleic acid

TEM Transmission Electron Microscopy

WB Washing buffer

Page 15: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

15

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

GENERATION OF DNA APTAMERS FOR HEPATOCELLULAR CARCINOMA

EXOSOMES

By

Sena Cansiz

December 2016

Chair: Weihong Tan Major: Chemistry

Extracellular vesicles (EVs), which were first discovered more than thirty years

ago, are now attracting considerable interest due to their key role in intercellular

communication. They affect various physiological and pathological functions of recipient

cells by transferring their cargo composed of proteins, lipids and nucleic acids. There

are several types of vesicles, which are categorized according to their size and

functions. Among them, exosomes are the most abundantly studied one, due to many

reasons, such as acting as a messenger between cells and participating in different

cellular homeostatic pathways. In addition, the molecular contents of the exosomes that

are secreted into body fluids have proven to be highly specific and a precious

biomedical tool. In order to take advantage of their functions and develop a way of

detection, here we designed and performed the first extracellular vesicle SELEX (EV-

SELEX) to generate DNA aptamers against hepatocellular carcinoma exosomes with a

counter selection against human blood exosomes.

In the second part of this dissertation, the interaction of a membrane protein

PTK7 and genomic DNA was elaborated. Analysis of DNA aptamers, selected

independently against different target cells by whole cell-SELEX, identified 4 aptamers

Page 16: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

16

with a common target protein, PTK7, by competition experiments. The 4 aptamers

share significant sequence identity to both strands of a DNA sequence in the 5’-

untranslated region for protein DIXDC1 Further analysis of the PTK7 aptamers and

DIXDC1 gene revealed more sequence identities (22 nucleotides total) which is a

unique occurrence in the human genome. In addition, western blot analysis of PTK7 in

different cellular compartments indicated a PTK7 accumulation in the nucleus.

Moreover, a gel shift assay proved the interaction between PTK7 and the DIXDC1

gene. Taken together, these findings indicate that these DNA aptamers may have an

analog in genomic DNA.

Page 17: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

17

CHAPTER 1 INTRODUCTION

Cancer

By the end of 2016, it is estimated that about six hundred thousand Americans

and more than 6 million humans around the world will die of cancer. In the United

States, one in two men will develop cancer during their lifetime.1 A quarter of all

American deaths and about 15% of all deaths worldwide will be attributed to cancer.2

Looking at the bitter statistical data provided above, it would be fair to say cancer is a

major public health problem worldwide. In fact, cancer is not one disease but many

diseases, which share a fundamental feature: the abnormal growth of the cell. In

addition, cancer is a clonal disease, that is, nearly every known cancer originates from

one ancestral cell that, having acquired the capacity of limitless cell division and

survival. Indeed, Greaves and Maley have recently reviewed inherently Darwinian

character of cancer and discussed the fact that clonally evolving nature of the disease is

the primary reason for the failure of a universal therapeutic.3 Before 1980s, the cancer

therapy was largely depend on two fundamental vulnerabilities of cancer cells: originate

as a local disease before it becomes malignant and the rapid growth rate, which can be

targeted by chemotherapeutic drugs.4 Later on, more specific and effective treatment

methods were developed such as nanocarriers and molecules that can selectively

target tumours.5 Nonetheless, prevention and/or early diagnosis is yet the best cure. By

attacking precancer rather than cancer, progression can be prevented.6 As stated by Dr.

Sidney Farber in November 1967,

the greatest need we have today in the human cancer problem, except for a universal cure, is a method of detecting the presence of cancer before there are any clinical signs of symptoms.7

Page 18: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

18

Extracellular Vesicles: Message in a Bottle

Cells do not live in isolation, indeed cell-to cell communication is crucial for all

multicellular organism. Their survival depends on receiving and processing information

from the outside environment, whether that information is related to the availability of

nutrients, changes in temperature or in light levels. There are different ways of basic cell

communication, such as through the secretion of soluble factors (e.g. hormones,

cytokines) 8,9, by direct interaction10,11 and release of membrane derived vesicles. In this

particular chapter, extracellular vesicles will be under the magnifying glass.

Extracellular vesicles (EVs) is a generic term that refers to all membrane vesicles

secreted in the extracellular space. Indeed, they are spherical membrane derived

particles with a diameter ranging from 10 nm up to 5 μm, which possess different

functions, biophysical properties and have different biogenesis routes.12–14 The power of

EVs is the ability to transfer information to another cell and thus influence the recipient

cell function. Like a message in a bottle, EV-assisted signaling can be transferred by

different biomolecule categories such as, protein, lipids, nucleic acids and the unique

package of this information provides the option of simultaneous delivery of multiple

different messengers even to sites far from the parent cell.15,16

Major improvements in the detection of EVs have been made recently (Figure 1-

2).17 Due to their contribution to health and disease, the clinical interest in EVs as

noninvasive biomarkers for diagnosis or prognosis is emerging. Also, EVs may have

several potential therapeutic applications, which are currently being explored. Before

going more into detail about their clinical usage, the history and biogenesis will be

discussed initially.

Page 19: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

19

History

EVs were observed as procoagulant platelet-derived particles in normal plasma,

originally reported in 1946 by Chargaff and West18 and referred to as ‘‘platelet dust’’ by

Wolf in 1967.19 However, the story of exosome biogenesis and secretion begins with the

discovery of the lysosome by Christian De Duve et al in 1955.20 A membrane isolates

the lysosome's acidic environment, preventing its enzymes from harming the rest of the

cell. By using ultracentrifugation method, they were able to isolate lysosomes in cell

fractions, which later were imaged by electron microscopy.21 In the 1970-1980s,

separate independent EV observations included the release of plasma membrane

vesicles from rectal adenoma microvillus cells22, reports on virus-like particles in human

cell cultures and bovine serum and23 the detection of vesicles, later termed

prostasomes.24 In 1983, two papers were published within a week of each other, which

are contributing exosomes and exosome secretion as well as Endosome-Lysosome

Pathway. Harding et al, were able to show a novel mechanism for the loss of transferrin

receptors during maturation of reticulocytes.25 In their study, internalized AuTf particles

were located primarily and predominantly on the many small vesicles which were

observed within multivesicular bodies, and therefore these organelles are called

multivesicular endosomes (MVE) (Figure 1-1). In parallel, exosomes were discovered

when vesicles were isolated from sheep reticulocytes. These vesicles contain the

plasma membrane receptor transferrin, which is absent on mature erythrocytes,

suggesting that “vesicle externalization could be a mechanism for shedding of specific

membrane functions, which are known to decrease during maturation of reticulocytes to

erythrocytes”.26,27 More than a decade later, Raposo et al demonstrated that these

vesicles, now termed exosomes, isolated from Epstein-Barr virus transformed B

Page 20: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

20

lymphocytes, were antigen-presenting and able to induce T-cell responses.28 In 2006-

2007, with the discovery that EVs contain RNA, including microRNA, EVs acquired

substantially renewed interest as mediators of cell-to-cell communication.29,30 Advancing

on these pioneering studies, EVs have been isolated from most cell types and biological

fluids such as saliva, urine, nasal and bronchial lavage fluid, amniotic fluid, breast milk,

plasma, serum and seminal fluid.31–35

Classification of Extracellular Vesicles

There are mainly three types of EVs: Apoptotic Bodies, Microvesicles

(Ectosomes) and Exosomes. They differ in terms of size, components and functions.

Although, apoptotic bodies comprise a type of extracellular vesicles, they originate from

apoptotic cells and are fragments of dying cells.36 The change of osmotic pressure

arising from the apoptosis mechanism leads to blebbing and release of apoptotic bodies

which can be engulfed by macrophages. The vesicles formed have a size of 1-5 μm and

the mechanism leading to their release is well-understood compared to that of

exosomes and microvesicles.37 Microvesicles (MV), on the other hand, are formed by

outward budding of the plasma membrane. They are defined as close lipid bilayer sacs

which contain information capable of influencing the environment such as tumor growth.

They are heterogeneous in shape and size (100-1000 nm).38 The release of MV is a

regulated mechanism induced by activation of cell surface receptors and increase in

intracellular Ca2+ concentration.39 In addition, the release rate is enhanced for tumor

cells in comparison to normal healthy cells. A third and most widely studied type of EVs

is exosomes, which is one of the main focus of this dissertation.

Page 21: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

21

Exosomes

Exosomes, membranous vesicles of endocytic origin, are signaling organelles

secreted by normal and disease cells.40,41 Originally described three decades ago27,

exosomes contain a subproteome of the cells and are found in many bodily fluids.

Released upon fusion of multivesicular bodies (MVBs) with the plasma membrane (PM),

exosomes are of 40–100 nm in diameter, are of endocytic origin, have a cup shaped

appearance as visioned by electron microscopy, have a buoyant density in sucrose of

1.10–1.21 g/mL and sediment at 100,000 g.42 They harbor proteins/RNA/lipids that

reflect the functionality of the host cell and possess molecular signatures or footprints

resembling the diseased cell from which they were secreted.40,43 There has been an

extensive research going on related with exosomes, especially in recent years. The

increasing trend in the number of researches, which are related with exosomes was

shown as a histogram in Figure 1-2. This enormous interest in exosomal studies can be

attributed to three main reasons: 1) Important role of exosomes in intercellular

signaling42; 2) use as delivery vehicles for vaccines and drugs41 and 3) as possible

sources of disease biomarkers44.

Protein Composition of Exosomes

The size of exosomes is related with their origin. Since they are indeed vesicles,

their minimum size is dependent on the structures of a lipid bilayer. A lipid bilayer has a

thickness of about 5 nm, and the bilayer has enough stiffness that the smallest vesicle

possible is on the range of 30 nm. Since they derive by budding off inside endosomes

(200–500 nm), their maximum diameter realistically should be on the order of 100 nm.

The implication of this small size is that the “cargo hold” for these particles is on the

order of 20–90 nm across. This is comparable to the volume of a eukaryotic ribosome,

Page 22: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

22

so the total cargo per exosome is probably ≤100 proteins and ≤10,000 net nucleotides

of nucleic acid.45 Standard negative staining methods for transmission electron

microscopy (TEM) allow visualization of round vesicles with obvious lipid bilayers as

well as some bodies with a characteristic cup-shaped morphology.46 Besides a

characteristic morphology, exosomes are thought to be somewhat unique in their

protein and lipid composition, providing additional traits for their identification. Due to

their endosomal origin, all exosomes contain membrane transport and fusion proteins

(GTPases, Annexins, flotillin), tetraspannins (CD9, CD63, CD81, CD82)47,48, heat shock

proteins (Hsc70, Hsp 90)36,49, proteins involved in multivesicular body biogenesis (Alix,

TSG101)48,50, as well as lipid-related proteins and phospholipases.51 Beyond these

membrane-associated proteins, over 4400 different proteins have been identified in

association with exosomes, usually by mass spectrometry, presumably serving as cargo

for inter-cell communication.52

Biogenesis and Release of Exosomes

Exosomes are formed within the endosomal network, a membranous

compartment that sorts the various intraluminal vesicles and directs them to their

appropriate destinations, including lysosomes and cell surface membranes. In doing so,

endosomes target some proteins/lipids for lysosomal degradation while targeting others

for recycling or exocytosis.53 The machinery that drives MVB formation is directly

relevant to exosome production. A model for MVB formation was proposed more than

30 years ago.41 Two sequential steps have been discerned for protein sorting in MVBs.

The first step involves the lateral segregation or selection of proteins at the limiting

membrane. The second step is the formation of inwardly budding vesicles with the

concomitant incorporation of selected cargo. Only recently, a few aspects of the

Page 23: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

23

responsible molecular mechanisms have been uncovered. In yeast, at least 15 class

EVPS genes are required for protein sorting into MVBs and these have orthologs in

mammalian cells, indicating that the molecular mechanism for MVB sorting was

conserved during evolution. Lipid metabolism appears to be important for the

biogenesis of MVBs. The MVB pathway in yeast and mammalian cells requires

phosphatidyl-inositol (PI) 3-kinase as well as PI (3)P 5- kinase activities , but the precise

roles of their reaction products, PI(3)P and PI(3,5)P2, for this process remain to be

established. Interference with the mammalian PI 3-kinase VPS34 did not affect the

sorting of EGF-receptor into aggregates at the MVB-limiting membrane, but did prevent

the formation of internal vesicles. A number of protein complexes have recently been

shown to be important for the biogenesis of MVBs. Hrs is a PI (3)Pbinding protein, and

disruption of Hrs expression results in aberrant MVB formation. Hrs has been

demonstrated to recruit clathrin to endosomes and to be important for EGFreceptor

down-regulation. Both activated EGF-receptor and Hrs associate with flat clathrin

lattices on vacuolar maturing endosomes, suggesting a role for such clathrin lattices in

the assembly of proteins for packaging in MVB internal vesicles. In addition, Hrs

interacts with sorting nexin 1 (SNX1), and this interaction is equally important for the

down-regulation of EGF receptor in MVBs. Furthermore, EGF-receptor sorting in MVBs

is dependent on c-Cbl, a ubiquitin ligase for EGF-receptor. Also in yeast, ubiquitination

of endosomal cargo serves as a signal for sorting in MVBs. Here, a hetero-oligomeric

protein complex, ESCRT-1, has been identified that contains the yeast ortholog of

mammalian Tsg101, Vps23. ESCRT-1 is thought to recruit ubiquitinated proteins for

MVB sorting through a direct interaction with VPS23. Apparently, all components that

Page 24: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

24

are required to recruit proteins into the MVB pathway, including ubiquitin, ESCRT-1 and

the clathrin coat, are released from assembled cargo prior to the actual packaging into

inwardly budding vesicles at the MVB-limiting membrane. A schematic for the

biogenesis of exosomes was presented on Figure1-4.54

Aptamers

Aptamers are single-stranded oligonucleotides (RNA/DNA) which fold into well-

defined three-dimensional structure for specific recognition and interaction with their

target.55 The name “aptamer”, which originates from the Latin word “aptus” meaning “to

fit” and the Greek word “meros” meaning “part”, was used in 1990 by Ellington and

Szostak in their initial work selecting an RNA aptamer recognizing an organic dye.56 In

the same year, Gold and Tuerk selected an RNA aptamer recognizing bacteriophage T4

DNA polymerase and named the selection process SELEX, short for Systematic

Evolution of Ligands by EXponential enrichment.57 Still in the same year, Robertson and

Joyce adapted a class I ribozyme so that it would specifically cleave DNA instead of

ssRNA.58 In these papers, they independently introduced the concept of in vitro

selection of RNA molecules able to specifically recognize a target. Since then, DNA

aptamers have emerged and many RNA and DNA aptamers have been generated

towards many targets including, small organic molecules metal ions, proteins,

carbohydrates, toxins, and transcription factors, as well as whole cells, viruses, bacteria,

and as inhibitors of protein functions.

SELEX

The SELEX process has four major steps: 1) incubation of a library composed of

thousands of different oligonucleotide sequences with the target; 2) separation of the

bound sequences from the unbound sequences; 3) recovery of the bound sequences by

Page 25: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

25

dissociation from the target; 4) amplification of the recovered sequences by polymerase

chain reaction (PCR).57

These steps are repeated until enrichment of the library in

binding sequences is attained. At this point, the pool containing the enriched sequence

is sequenced and further analyzed by informatics. The binding sequences are aptamers

which can be further shortened and modified to be more resistant to nuclease

degradation, to make them fluorescent or to carry a tag molecule for further coupling

with yet another molecule. One of the major limitations of the SELEX process is the

uncertainty of the success of the selection a priori. It is impossible to predict with

certainty the generation of an aptamer against the target chosen, this being even truer if

the target is a protein. Since aptamers are negatively charged due to their phosphate

backbones, it would appear that positively charged proteins at physiological pH should

be the best candidates. However, some aptamers have been successfully generated

against protein having isoelectric points below 7.4.59 Cell-SELEX

To select aptamers for whole cells, a negative control is usually included either

as a normal cell line or different cancer cell line. Cell-SELEX begins with the binding

event between the initially synthesized library and the target cells, unbound and weakly-

bound sequences are washed off. Bound sequences are collected and (if negative

selection is to be performed) are incubated with the negative cells. This time the,

unbound sequences are collected and further PCR amplified. Then, dsDNA is converted

to ssDNA and a new round starts. This process is continued until the initial library is

enriched with sequences that bind to the cancer cell but no to the control cell.60 Once

Page 26: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

26

enrichment has been achieved, the pool is sequenced and analyzed using alignment

programs to identify conserved sequences.61

Overview of Dissertation

The research data presented in the first part of this dissertation demonstrate how

to select DNA aptamers against hepatocellular carcinoma exosomes. It includes how to

characterize exosomes and how to develop a method to select aptamers. Chapter 2

describes the characterization process and Chapter 3 focuses on selection process.

The second part of the dissertation, Chapter 4, focuses on a study related with PTK7

aptamer and it is discovery among the genome. The concluding chapter recapitulates

the significance of developing a new SELEX technology and the importance of

screening downstream effects of aptamers.

Page 27: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

27

Figure 1-1. Exocytosis of MVEs releases exosomes containing transferrin receptor. A)

Small vesicles and tubules in the reticulocyte cytoplasm are labeled with AuTf. Bar 200nm B) View of an MVE in a reticulocyte that was incubated with AuTf.Bar 100nm. C) View of an MVE sparsely labeled with AuTf. Bar 100nm. D) View of MVE exocytosis in an unfixed reticulocyte. Bar 200nm E) Exocytosis of a small AuTf-labeled MVE. Bar 100nm F) An adherent membrane vesicle with associated AuTf. This presumably represents the remnant of an MVE exocytosis. Bar 100nm. Figure and legend adapted from Harding et al. (1983)25. Used under the permission of Rockefeller University Press.

Page 28: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

28

Figure 1-2. Histogram of exosomal studies over the past 40 years. An increasing interest in exosome research was seen during the last decade. The statistics is generated based on PubMed indexed exosomal studies (keywords: exosomes or exosome-like).

Page 29: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

29

Figure 1-3. Schematic for extracellular vesicle trafficking.

Page 30: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

30

Figure 1-4 Biogenesis of extracellular vesicles and their interactions with recipient cells. Figure adapted from reference EL Andoloussi et al.54

Page 31: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

31

CHAPTER 2 ISOLATION AND CHARACTERIZATION OF EXOSOMES FROM DIFFERENT CELL

LINES

Background and Significance

As it was explained in the introductory chapter, extracellular vesicles hold a great

importance in terms of cell-to-cell communication, intercellular signaling, waste

management, coagulation, etc. They are largely released in biological fluids, such as

plasma, urine, cerebrospinal fluid, amniotic fluid, malignant and pleural effusions of

ascites and breast milk, hinting a diverse role in the exchange of information among

different body compartments. 32,62,63 In addition, it has been showed that in particular

disease conditions, exosomes may play regulatory functions. There are a number of

evidences indicating the relationship between exosomes and some neurodegenerative

disease such as prion, Alzheimer’s and Parkinson’s disease. It was found out that

exosomes are carrying some neurodegenerative disease associated proteins such as β-

amyloid and α-synuclein and they facilitate their spread from their cells of origin to the

extracellular environment.64 For example, β-amyloid peptides, associated with

Alzheimer's disease, are carried with exosomes and that exosomal proteins were found

to accumulate in the plaques of AD patients’ brains.65 Moreover, in a research done by

Fevrier et al prion proteins are shown to be released from cells in association with

exosomes and travelling in the body as an infectious route for propagation of disease.66

Consequently, there is a growing interest in the clinical applications of vesicles.

However, because of the small size and heterogeneity of vesicles, their isolation and

detection is challenging. Currently, there is no one single method which can accurately

phenotype, size, and detect the concentration of the whole range of EVs and therefore

provide all the necessary information to understand the biology of extracellular vesicles.

Page 32: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

32

In this particular chapter, we will discuss about the isolation and characterization of

exosomes from different human primary cell lines including Hep G2, HeLa, CEM,

Ramos and whole blood. This can be considerate as a preliminary step for the EV-

SELEX, which will be discussed in Chapter 3.

Materials and Methods

General Materials

Unless specified otherwise, all the reagents were purchased either from Thermo-

Fisher or Sigma Aldrich and used without further purification. All DNA synthesis

reagents were purchased from Glen Research.

Cell Lines and Culturing

All the cell lines used either for exosome collection or general binding

experiments were purchased from American Tissue Culture Collection (ATCC). Ramos

cells (CRL-1596, B lymphocyte, human Burkitt's lymphoma) were grown in complete

RPMI 1640 medium (Sigma) supplemented with 10% (v/v) fetal bovine serum (FBS)

(heat inactivated, GIBCO) and 100 IU/mL penicillin-streptomycin (PS) (Cellgro). HeLa

cells (cervical adenocarcinoma (CCL-2)) were grown in Dulbecco’s Modified Eagle’s

Medium (DMEM) supplemented with sodium bicarbonate (1.5g/L), 10% (v/v) FBS and

100 IU/mL PS. Hep G2 cells (CRL-11997, human liver hepatocellular carcinoma) were

grown in Eagle’s Minimum Essential Media (EMEM) supplemented with sodium

bicarbonate (1.5g/L), 10% (v/v) FBS and 100 IU/mL PS. All cell lines were sub-cultured

in either T-75 flasks (Corning) or in 35 mm cell culture dishes at 37°C with 5% CO2.

Exosome Isolation from Cells

Exosomes were obtained from supernatant of cells, which are cultured as

previously described. In order to collect a higher number of exosomes, cells were grown

Page 33: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

33

in T-225 cm2 flasks (Corning) in complete growth media until they reached a confluency

of 80–90%. Then, the media was discarded and the cell, which are still attached to the

flask were washed with 10mM PBS. Following the washing step, the cells were cultured

in complete growth media, which is supplemented with 10% v/v exosome depleted FBS

(Thermo Fisher Scientific) rather than the regular FBS for 48h. Next, the media was

collected and centrifuged at 800g for 5 min to discard the cells, followed by a

centrifugation step of 2,000g for 10 min to discard cellular debris. Then, the media was

filtered using a 0.2-μm pore filter (Grainger, 11L832). The collected media (~200mL)

was then split in 6 UltraClear™ thinwall tubes (Beckman Coulter, 342204) and

ultracentrifuged at 100,000g for 2h at 4 °C with SW28 Ti rotor. The exosome pellet in

each tube was washed with 6 mL 10mM PBS, collected in a single tube and filtered

using 0.2-μm pore filter (syringe filter, 6786-1302, GE Healthcare), followed by a second

step of ultracentrifugation at 100,000g for 2h at 4 °C. Finally, the supernatant was

discarded carefully and the pellet is resuspended in 150 μL 10mM PBS and stored at -

80 °C. The schematic representation of the isolation process is summarized in Figure 2-

1.

Exosome Isolation from Whole Human Blood

Whole Human Blood was purchased from Life South (1 unit, R259). Upon arrival,

the whole blood was split in 50mL of Falcon Tubes and centrifuged at 1,500 x g for 20

min at 4 °C to initiate separation of cells from plasma. Next, the supernatant (plasma)

was transferred in to a new Falcon Tube and centrifuged at 2,800 x g for 20 min at 4 °C

twice to remove all cells from plasma. Then, cell-free plasma (CFP) was filtered using a

0.2-μm pore filter (Grainger, 11L832). The CFP (~250mL) was then split in 6

UltraClear™ thinwall tubes (Beckman Coulter, 342204) and ultracentrifuged at

Page 34: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

34

100,000g for 2h at 4 °C with SW28 Ti rotor. The exosome pellet in each tube was

washed with 6 mL 10mM PBS, collected in a single tube and filtered using 0.2-μm pore

filter (syringe filter, 6786-1302, GE Healthcare), followed by a second step of

ultracentrifugation at 100,000g for 2h at 4°C. Finally, the supernatant was discarded

carefully and the pellet is resuspended in 500 μL 10mM PBS and stored at -80 °C.

Nanoparticle-Tracking Analysis (NTA)

NTA measurements were performed with a NanoSight LM20, equipped with a

sample chamber with a 640-nm laser and a Viton fluoroelastomer O-ring. The samples

were diluted in 10mM PBS with either 1:10 or 1:100 ratio depending on the initial

concentration. The samples were then injected in the sample chamber with sterile Luer-

Lok syringes (BD) until the liquid reached the tip of the nozzle. All measurements were

performed at room temperature.

Western Blot Analysis

Exosome samples from different cell lines were lysed in Lysis 250 Buffer (50mM

Tris-HCl, pH 7.4, 0.5% NP-40, 250mM NaCl, 5mM EDTA, 50mM NaF) containing 5

μg/Ml leupeptin, 1 μg/mL pepstatin and 1 mM phenylmethylsulphonyl fluoride (PMSF).

Lysates were collected and centrifuged at 14,000 rpm for 15 min, and the supernatants

were collected. Protein quantification was determined by bicinchoninic acid (BCA)

Assay and the information related with standard curve and mean standard

concentrations summarized in Figure 2-2. Following this, 50 μg of protein from each

sample was boiled in 4X NuPAGE® LDS Sample Buffer (Thermo Fisher) at 95°C for 5

min. Proteins were resolved by 8% SDS-PAGE and then transferred to Polyvinilyidene

difluoride (PVDF) membrane by semi-dry transfer. The protein blot was blocked for 1 h

at room temperature with 5% non-fat dry milk in PBS/0,05% Tween and incubated

Page 35: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

35

overnight at 4 °C with the following primary antibodies: Exosome CD63 (Thermo-

Fisher). In order to, remove the nonspecific and unbound antibodies, the blot was

washed with PBS 0.05% Tween-20 5 times for 8 min each. Next, horseradish

peroxidase (HRP)-conjugated secondary antibodies (GE Healthcare) were incubated for

1 h at room temperature. Washes after secondary antibody incubations were done on

an orbital shaker, 6 times at 8 min intervals, with PBS 0.05% Tween-20. Blots were

developed with chemiluminescent reagents from Pierce.

Exosome-Bead Attachment

10μL exosomes (1013 exosomes/mL) were mixed with 10 μL Aldehyde Sulfate

Latex (ASL) (Thermo Fisher Scientific) beads for 15 min at room temperature with

continuous rotation. This suspension was diluted to 1 ml with PBS and left for 30 min

rotating at room temperature. The reaction was stopped with stop solution (100 mM

glycine and 2% BSA in 10 mM PBS) and left rotating for 30 min at room temperature.

Exosomes-bound beads were washed once in 2% BSA in 10mM PBS and centrifuged

for 1 min at 14,800g and blocked with blocking solution (10% BSA, 0.1mg/mL salmon

sperm DNA in 10mM PBS) with rotation at room temperature for 30 min. Then the

beads were washed second time in 2% BSA and centrifuged for 1 min at 14,800 g.

Finally, the exosome-bound beads are recovered in 10mM PBS and stored at 4°C

temporarily.

Flow Cytometer Analysis of Exosome Bound Beads

Bead concentration is optimized for 10,000 events and 1.2 µg/mL of exosome-

bound beads were used as the optimum (minimum) concentration for all the binding

assays. Beads were centrifuged at 14,800g for 1 min in order to be recovered from the

storage solution and washed with bead-washing buffer (5mM MgCl2, 2% (w/v) BSA in

Page 36: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

36

10mM PBS). Beads then mixed with 250nM of biotin labelled sequence to be analyzed

and incubated with rotation at 4°C for 30 min. Afterwards, the beads were washed in

bead-washing buffer and centrifuged at 14,800g for 1 min in order to remove the

unbound sequences. Next, the recovered beads were resuspended in streptavidin

conjugated R-phycoerythrin (SA-PE) (Thermo Fisher Scientific) in washing buffer with a

1:400 dilution rate and incubated with rotation for 15 min at 4°C. In order to remove

excess SA-PE, the beads were washed twice with bead-washing buffer and recovered

by centrifugation at 14,800g. Finally, washed exosome-bound beads were resuspended

in 100µL of bead-binding buffer. The fluorescence was analyzed using BD Accuri C6

flow cytometer (BD Biosciences) and the results were interpreted by FlowJo™ software.

Immunogold Labelling TEM

For the TEM observation of pure HepG2 exosomes, the optimal concentration of

the samples was directly absorbed on a f-carbon-coated copper grid and dried at room

temperature. For immunogold labeling samples, optimal concentration of HepG2

exosomes were placed onto grids and allowed to be absorbed. The grids were blocked

with 1% BSA/PBS for 1h, and then placed on biotin-labeled LZH8 aptamer solution for

1h at 4 °C, and rinsed with PBS for 5 times. After washing, grids were floated on drops

of streptavidin-gold nanoparticles for 30 min at 4 °C. Finally, the grids were rinsed with

10mM PBS for 5 times and dried at room temperature. As controls, grids were not

exposed to LZH8 aptamer. The dried sample was observed on a Hitachi H-7000 NAR

transmission electron microscope using a working voltage of 100 kV.

Page 37: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

37

Results and Discussion

Exosome Isolation from Cell Culture Media and Blood: Ultracentrifugation and Ultrafiltration

A major problem in EV research is the lack of characterization of current methods

evaluating their usability, vesicle purity and yield from cell media, and complex

biological fluids such as whole blood.67,68 The current “gold standard” for the purification

of a subset of exosomes is differential centrifugation. Differential centrifugation consists

of successive centrifugation steps with increasing centrifugation forces and durations,

generally aimed at isolating smaller from larger objects. Larger particles, assigned to be

removed in the first centrifugation steps, sediment faster and leave most of the smaller

particles in the supernatant. The supernatant will be centrifuged in subsequent steps.69

Larger particles refer to cells and large vesicles which typically will be removed by low-

speed centrifugation and the supernatant contains smaller vesicles, such as exosomes,

which will be ultra-centrifuged to pellet. Even tough, separation of exosomes by

ultracentrifugation method is one of the most effectively and commonly used one, it still

needs to be further optimized or combined by different techniques. It has been

suggested in the literature that repeated ultracentrifugation steps can damage vesicles

and reduce yields, thereby potentially impacting proteomic and RNA analysis of

exosome content.70 Besides, the pellet collected might be contaminated by other types

of vesicles, rather than being a homogenous exosome population. In order to overcome

all these potential problems, we combined differential centrifugation method with several

ultrafiltration steps (Figure 2-1). According to the data that we collected from several

characterization steps, we can claim that exosomes constitutes majority of the vesicle

Page 38: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

38

population but not the whole. Further immunoprecipitation steps might be necessary to

obtain more homogenous exosome extraction in the future.

Size and Concentration Analysis of Particles by Nanoparticle-Tracking Assay

One of the challenges with identifying the size and structure of exosomes is that

they are one of several extracellular nano/micro-scaled vesicles that are produced by

cells all the time and vary in size, molecular composition, and biological function.42,71

Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) are

one of the mainly used tools for the analysis of particle size and morphology of

exosomes.35,72 However, both SEM and TEM have the disadvantage that the

preparation of samples is time-consuming, both methods involve labor-intensive steps

and each has some risk of artifact generation. Neither method is suitable for high

sample throughput and characterization of thousands of single particles of one sample.

A highly sensitive method for visualization and analysis of exosomes is NTA.73 This

method takes advantage of two different principles of physics: First, particles are

detected by the light scattered when they are irradiated with a laser beam (Figure 2-3).

The second phenomenon is known as Brownian motion, according to which the

diffusion of different particles in a liquid suspension is inversely proportional to their

size.74 In the latter case, the movement also depends on the temperature and the

viscosity of the liquid. Using software-based analysis, digital images of scattered light

from single particles are recorded. Plots of scattered light spots and their speed of

motion provide the data that facilitate the determination of total particle count and size

distribution.

We used NTA in our experiments to determine size distribution and the

concentration of the collected exosomes. According to data we collected, size

Page 39: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

39

distribution curves of the particles were constructed. Mean size for Hep G2 exosomes

was 77.35 nm whereas it is 88.95 (Figure 2-4) for whole blood exosomes. Even though

the mean size of the exosomes collected is within the range, there are still some particle

population which has a bigger size than an average exosome. Besides, despite the fact

that all particles having a size bigger than 220nm were eliminated by ultracentrifugation,

we observed particles around or more than 300nm. This is either a contamination or an

artifact of aggregated particles. Based on the information on the literature, this

technique is particularly powerful for analyzing particles with a mean diameter of less

than 100 nm, which is consistent with our results.

Measuring the Protein Content of Exosomes Using the BCA Assay

Measuring the amount of total proteins present in the exosome preparations

gives a rough idea of the number of exosomes secreted by the cells. When performing

immunoblots with exosomes and total cell lysates, or when comparing different

exosome preparations on the same immunoblot, it is important to perform the protein

quantification by the BCA or Bradford assay on all the samples at the same time.

According to the results that we got (Figure 2-2), the exosome lysates obtained from cell

culture media were able to measured easily and the value obtained stayed in the

standard range. On the other hand, in the case of blood exosome lysates, the protein

concentration was too high, even though the initial exosome concentration inside fair.

Clearly, the plasma contains too much protein contaminants which is interfering with

exosome protein content. For the future experiments, in order detect the concentration

of blood exosome lysate, the value obtained from NTA measurement might be used

instead of using BSA or Bradford Assays.

Page 40: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

40

Flow Cytometric Detection of Exosomes

Standard Flow Cytometer detects vesicles above approximately 200 nm, and

therefore exosomes and smaller MVs cannot be analyzed directly by this method. Thus,

it has to be emphasized that MVs smaller than the detection limit of the used flow

cytometer cannot be discriminated from the instrument noise, leading to an inadequate

numbering of MVs.75, unless they are conjugated with beads. In our experiments, we

were targeting exosomes, which are between 40-100nm in size. Therefore, it is

impossible to detect them by flow cytometry, unless they conjugated with a bigger

material. In order to do so exosomes are attached to ASL beads as described in

experimental section (Figure 2-5). Further, the flow cytometry experiment was

performed with exosome-beads conjugates. Blood exosomes are tested with HepG2

aptamers which was recently selected76 and unfortunately we discovered that they all

bind to blood exosomes (Figure 2-6).

Concluding Remarks

There is an urgent need for more efficient, reliable and reproducible EVs

extraction methods, so that all downstream studies in the field of EVs can be more

standardized and efficient. Here in this chapter, we were able to perform a serious of

characterization experiments for the exosomes isolated from different cell lines. First,

the size and the concentration of the exosomes were detected by NTA. It is overall a

reliable method since the size of the exosomes are less than 100nm. Further, the

exosome lysates were subjected to western blot to confirm their CD 63 content. Flow

cytometer detection of ASL bead conjugates blood exosome beads reveled the fact that

all HepG2 aptamers have an affinity blood exosomes.

Page 41: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

41

Figure 2-1. Flow chart for the exosome purification procedure based on differential ultracentrifugation and ultrafiltration. The speed and length of each centrifugation are indicated to the right of the arrows.

Page 42: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

42

Figure 2-2. BCA experiment schematic. A) Schematic for the 96 plate design used for the experiment with unknowns and

standards labelled. B) Standard curve for the BSA. C) Mean absorbance and protein concentrations for exosome cell lysates extracted from different cell lines and whole human blood.

Page 43: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

43

Figure 2-3. Schematic of optical configuration used in NTA. A laser beam is passed through the sample chamber, and the particles in suspension in the path of this beam scatter light in such a manner that they can easily be visualized via a 20x magnification microscope onto which is mounted a camera. The camera operates approximately at 30 frames per second (fps), captures a video file of the particles moving under Brownian motion.

Page 44: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

44

Figure 2-4. Size distribution from NTA measurements of Hep G2 exosomes A) and whole human blood exosomes B) with the corresponding NTA video frame (left panels)

Page 45: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

45

Figure 2-5. Characterization of exosome preparations from different cell lines or human

whole blood by Western blot. Common exosome marker anti-CD63 was used as the primary antibody.

Page 46: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

46

Figure 2-6. Schematic of capture and fluorescent analysis of extracellular vesicles.

Extracellular vesicles are captured by ALS beads and tagged by biotin conjugated aptamers and then stained with streptavidin conjugated fluorophore. The beads are analyzed by flow cytometry.

Page 47: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

47

Figure 2-7. Binding test of different Hep G2 aptamers with blood exosomes by flow cytometry. A random sequence

(library) indicated the background. Right panel shows the dot blot for the related experiment.

Page 48: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

48

Figure 2-8. Validation of isolated exosomes and interaction between aptamer LZH8 and

HepG2 exosomes. A) TEM observation of purified exosomes. B) Immunogold labeling TEM observation showing that the SA-AuNP could be attached on exosome surface via conjugation with biotinylated aptamers. C) Flow cytometry verification of exosomes using anti-EpCAM antibody. Isotype antibody indicated the background. D) Binding test using aptamer LZH8 with flow cytometry. A random sequence indicated the background.

Page 49: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

49

CHAPTER 3 DEVELOPMENT OF EV-SELEX METHODOLOGY AND SELECTION OF DNA

APTAMERS AGAINST HEPATOCELLULAR CARCINOMA EXOSOMES

Background and Significance

There are a number of evidences, which indicate the importance of EVs in a

variety of fundamental physiological and pathological processes.54,77,78 Among all, their

contribution to tumor growth and spread is the most striking one. The number of

circulating EVs in cancer patients is higher than in healthy individuals and has been

found to related with poor prognosis.79 In addition, a recent research suggests that,

exosomes may play a role in the distant spread, or metastasis, of cancer cells in the

body. Understanding this process could open new avenues of research on preventing

metastasis, which causes most deaths from cancer.80 The researchers found that

exosomes released from cancer cells had traveled to distant sites in the body and fused

with specific cells at these distant sites. These interactions made the local environments

suitable for the development of new tumors.81,82 In this regard, exosomes act as

communicative vehicles between tumor cells and the metastasis environment having

great potential as cancer biomarkers in personalized medicine for several reasons.

Firstly, exosomes travel across the body and can be collected from different body fluids

such as, serum, plasma, urine83 and breastmilk84 and thus eliminates the requirement

for invasive tissue biopsy. Secondly, exosomes carry cargos, which they are inherited

from their parent cells. Those cargoes in exosomes are protected by the phospholipid

bilayer from degradation by proteinases and nucleases. Consequently, biomarkers at a

relatively low expression are much easier to be detected through isolating exosomes.17

As described in the first chapter, aptamers are single-stranded oligonucleotides

capable of strong and specific binding to a target marker based on their unique three-

Page 50: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

50

dimensional folding.55 They are often compared to antibodies, since they exhibit similar

recognition mechanisms, specificity and selectivity. Aptamers are selected from an

initially large oligonucleotide pool (1012-1015 sequences) by a process called Systematic

Evolution of Ligands by EXponential enrichment (SELEX).60 The mode of selection and

the methodology used to generate these aptamers, as well as all the assays used in the

identification of the potential aptamer candidates, depend to a large extent on the target

of interest. In view of this, various selection modes have emerged. For the last decade,

Tan Group is a pioneer in cell-SELEX. We generated many DNA aptamers by using this

methodology.85–90

Recently, we extended our cell-SELEX knowledge to artificial bases and

developed artificially expanded genetic information systems (AEGIS)-cell-SELEX

technology.76,91 First, Sefah et al demonstrated the first example of a successful AEGIS

cell-SELEX against an adenocarcinoma breast cancer cell line by using GACTZP

library. Then, Zhang et al applied the same technology to select an aptamer against

hepatocarcinoma cell line with an addition of counter selection. This indeed increased

the selectivity of the selected aptamer. Yet, in both cases, it has been shown that the

aptamers containing artificial bases bound to their target molecule with a higher affinity

than their natural DNA replicas.

Herein this project, we developed a novel SELEX methodology targeting

extracellular vesicles rather than cultured cells. To date, it is the first example of a

SELEX method targeting exosomes. Besides, we used AEGIS technology to make the

selection more powerful. Hepatocarcinoma liver cancer cell line, Hep G2, exosomes

were used as the target molecule and whole human blood exosomes used for the

Page 51: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

51

counter selection. Two main goals of this study are firstly to develop the know-how

required for the EV-SELEX and secondly to generate DNA aptamers targeting a cancer

cell exosome in blood for clinical usage.

Materials and Methods

General Materials

Unless specified otherwise, all the reagents were purchased either from Thermo-

Fisher or Sigma Aldrich and used without further purification. All standard DNA

synthesis reagents were purchased from Glen Research.

Synthesis and Purification of Six Nucleotides (GACTZP) Libraries

All dZ and dP containing oligonucleotides (Table 3-1) were synthesized using

standard phosphoramidite chemistry on glass support (CPG) on an ABI 394 DNA

synthesizer. Protected dZ and dP phosphoramidites were purchased from Firebird

Biomolecular Sciences LLC (Alchua, FL). The primers were designed to satisfy the

following characteristics: a minimum hairpin structure, similar melting temperature (Tm)

and minimal base pairing. The forward primer (20nt) was labeled with Fluorescein

Isothiocyanate (FITC) at the 5’-end, and the reverse primer (20 nt) was labeled with

Biotin at the 5’-end. The library consisted of a randomized 30 nucleotide region

containing GACTZP at each site with a ratio 1:1:1:1:2:2, respectively. Coupling times

were 60 seconds. The CPG-bound DMT-off DNA molecules were incubated with

acetonitrile-triethylamine (1:1 v/v) for 1h at 25°C, followed by removal of supernatant.

The CPG-bound oligonucleotides were then incubated in acetonitrile-triethylamine (1:1

v/v) for overnight at 25°C. After removal of the supernatant, the CPG-bound

oligonucleotides were incubated with 1.0 mL of 1,8-Diazabicyclo[5.4.0]undec-7-ene

(DBU) in anhydrous CH3CN (1M) at room temperature for ~18 hours to remove the

Page 52: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

52

protecting groups on dZ. After removal of CH3CN, dZ and dP containing

oligonucleotides were retreated with NH4OH overnight at 55°C. The product mixture

was resolved by denaturating PAGE (7M urea) and extracted with TEAA buffer (0.2, pH

7.0). The product was then desalted by Sep-Pac® Plus C18 cartridges. All 5’

biotinylated dZ and dP containing sequences were synthesized, deprotected and

purified in house based on the above methods.

Cell Culture and Buffers

Hep G2 (CRL-11997, human liver hepatocellular carcinoma) cell line used either

for exosome collection or general binding experiments was purchased from ATCC.

They were grown in Eagle’s Minimum Essential Media (EMEM) supplemented with

sodium bicarbonate (1.5g/L), 10% (v/v) FBS and 100 IU/mL PS and subcultured in

either T-182 cm2 flasks (Corning) or in 35 mm cell culture dishes at 37°C with 5% CO2.

Washing buffer was prepared by adding 5mM MgCl2 and 2% (w/v) BSA in 10mM PBS.

Similarly, 10X binding buffer was prepared by mixing 50mM MgCl2, 1 mg/mL tRNA and

10mg/mL BSA in 10mM PBS, where tRNA and BSA serves as the stringency factors in

order to decrease non-specific binding.

Exosome Extraction from Hep G2 Cells for Positive Selection

Exosomes were collected using conventional centrifugation from supernatant

media of HepG2 cells. Cells were harvested in T-182 cm2 flasks in exosomes-depleted

FBS supplemented DMEM until they reached a confluency of 80~ 90%. Media was

collected and centrifuged at 800 g for 5 min at 4 °C, and the supernatant was then

centrifuged at 2,000 g for 10 min at 4 °C to discard cellular debris, followed by filtration

using a 0.22 µm filter (vacuum-driven filter, Genesee Scientific). The filtered media was

then ultracentrifuged at 100,000 x g for 2h at 4 °C. Pellet was then washed with 35 mL

Page 53: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

53

PBS, and centrifuged again at 100,000 x g for 2h at 4 °C. Finally, the supernatant was

discarded and exosomes were resuspended in 100 µL PBS. After several times

collection, the purity and concentration of exosomes was tested and measured by

NanoSight (NanoSight Ltd., Malvern). A stock solution of 1013 HepG2-derived

exosomes/mL was obtained.

Exosome Isolation from Whole Human Blood for Negative Selection

Whole Human Blood was purchased from Life South (1 unit, R259). Upon arrival,

the whole blood was split in several 50mL of Falcon tubes and centrifuged at 1,500 x g

for 20 min at 4 °C to initiate separation of cells from plasma. Next, the supernatant

(plasma) was transferred in to a new Falcon tube and centrifuged at 2,800 x g for 20

min at 4 °C twice in order to remove all cells from plasma. Then, cell-free plasma (CFP)

was filtered using a 0.2-μm pore filter (Grainger, 11L832). The CFP (~250mL) was then

split in 6 UltraClear™ thinwall tubes (Beckman Coulter, 342204) and ultracentrifuged at

100,000g for 2h at 4 °C with SW28 Ti rotor. The exosome pellet in each tube was

washed with 6 mL 10mM PBS, collected in a single tube and filtered using 0.2-μm pore

filter (syringe filter, 6786-1302, GE Healthcare), followed by a second step of

ultracentrifugation at 100,000 x g for 2h at 4°C. Finally, the supernatant was discarded

carefully and the pellet is resuspended in 500 μL 10mM PBS and stored at -80 °C.

Exosome-Bead Attachment

1 mL exosomes (1013 exosomes/mL) were mixed with 1 mL of Aldehyde Sulfate

Latex (ASL) beads (4µm) (Thermo Fisher Scientific) for 15 min at room temperature

with continuous rotation. This suspension was diluted to 100 mL with PBS and left for

30 min rotating at room temperature. The reaction was stopped with stop solution (100

mM glycine and 2% BSA in 10 mM PBS) and left rotating for 30 min at room

Page 54: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

54

temperature. Exosomes-bound beads were washed once in 2% BSA in 10mM PBS and

centrifuged for 1 min at 14,800g and blocked with blocking solution (10% BSA,

0.1mg/mL salmon sperm DNA in 10mM PBS) with rotation at room temperature for 30

min. Then the beads were washed second time in 2% BSA and centrifuged for 1 min at

14,800 g. Finally, the exosome-bound beads are recovered in 10mM PBS, aliquoted

and stored at-80°C.

Detailed Experimental Flow of EV-SELEX

The optimum annealing temperature of the primers and the library concentration were

determined before starting the selection process. All the PCR conditions, except for the

annealing temperature, were adapted according to AEGIS SELEX protocol.91 Annealing

temperature was optimized by running the same PCR mixture at different annealing

temperatures ranging from 55°C to 65°C. (Figure 3-2) Takara Taq (Clontech

Labroratories) was used as the DNA polymerase but Taq Buffer and dNTPs (dG, dC,

dT, dA, dZ, dP) were purchased from Firebird Biomedical Science (Alachua, FL).

Incubation step

As explained earlier, exosomes were attached with ASL beads which and for

each round of SELEX same exosome-bead conjugates were used to keep the

consistency. First, the beads were spun down at 14,800 x g for 1 min in order to remove

the storage solution. Meanwhile, 20 nmol of 300 µL of six-nucleotide library (or 250 nM

of recovered pool) was denaturated by heating at 85°C for 10 min and then immediately

“snap cooled” on ice for 10 min. This step is crucial for the sequence to form its 3D

structure. The library was then mixed with 10X Binding Buffer and incubated with bead-

exosome conjugates at 37°C for 30 min with rocking. Following the incubation, the

beads were washed twice with washing buffer and then recovered by centrifugation at

Page 55: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

55

14,800 x g for 1 min. Afterwards, the beads-exosome conjugates with bound sequences

on them were resuspended in 200µL of 10mM PBS and incubated at 85°C for 15 min in

order to denaturate the bound sequences. The beads were then removed from the

solution and the survivors were collected in the supernatant. Once the enriched

sequences were obtained, they were ready for the next step, which is PCR cycle

optimization.

PCR cycle optimization and amplification step

Optimization of the number of cycles was performed to obtain the best amplification of

the library pool without observing any nonspecific amplification. For this reason, it is

necessary to optimize the number of PCR cycle repeats for each round. All the PCR

reagents were mixed and a negative control with no template was included for each run.

A master mix containing 10x Taq buffer, dNTPs, primers and water was prepared. Taq

polymerase enzyme and the template were added lastly to the master mix and it was

aliquoted into the PCR tubes. The DNA template was amplified in a C1000

ThermoCycler (Bio-Rad). The number of cycle was varied by increment of 2 between 8

and 24 cycles (Figure 3-2). The cycle showing single band (no-specific amplification)

with the highest product quantity was chosen (Figure 3-2) was provided as an example

for the cycle optimization products, which were run on 3 % agarose gel. Once the

optimum cycle was determined, the amplification of the whole enriched pool was

performed with the selected cycle number and the results were screened similarly, by

3% agarose gel electrophoresis.

Preparation of single-stranded DNA

After PCR amplification, the selected pool was turned into dsDNA, where the sense

strand is FITC-labeled and the antisense strand is biotin-labeled. Since SELEX requires

Page 56: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

56

the use of an ssDNA pool, the PCR amplified pool needed to be converted back into

ssDNA. For this purpose, streptavidin coated high performance Sepharose beads (GE

Healthcare) were used to make a small affinity column. The dsDNA product was passed

through the column five times, allowing the binding of biontiylated dsDNA to the

streptavidin beads. The beads were then washed with 2.5 mL of PBS to remove any

remaining forward primer. After washing, the dsDNA was dehybridized with 200 nM

NaOH solution, separating the double strand and releasing the fluorophore-labeled

strand (sense strand). In order to remove the sodium salts, a size exclusion NAP-5™

column (GE Healthcare) was used to desalt the pool. The ssDNA was loaded on the

column, allowed to interact with it and eluted with water. The larger DNA molecules

were eluted first leaving the salts in the column. The desalted ssDNA was quantified by

a UV spectrophotometer and vacuum dried. The PCR products were resuspended in

binding buffer just before use in next round of selection. The next round starts again

with a denaturating step at 85°C for 10 min and a quick snap cool step. This process

was repeated for the following rounds. The negative selection (blood exosomes) was

introduced with Round 4 and the stringency was increased gradually (Table 3-2). The

entire selection process was repeated until a significant enrichment was obtained.

Please refer to Figure 3-1 for the schematic of EV-SELEX.

Monitoring of the pool enrichment

Flow cytometry was used to monitor the enrichment of ssDNA-bound sequences

within the pools during the selection process, as well as to evaluate the binding affinity

and specificity of the selected aptamers. Bead concentration is optimized for 10,000

events and 1.2 µg/mL of exosome-bound beads were used as the optimum (minimum)

concentration for all the binding assays. Beads were centrifuged at 14,800g for 1 min in

Page 57: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

57

order to be recovered from the storage solution and washed with bead-washing buffer

Beads then mixed with 250nM of the FITC ssDNA library or pool, and incubated with

rotation at 4°C for 30 min. Following this, the beads were washed in bead-washing

buffer and centrifuged at 14,800g for 1 min in order to remove the unbound sequences.

Finally, washed exosome-bound beads were resuspended in 100µL of bead-binding

buffer. The fluorescence was analyzed using BD Accuri C6 flow cytometer (BD

Biosciences) and the results were interpreted by FlowJo™ software.

Results

EV-SELEX Method and Generation of DNA Aptamers against Hepatocellular Carcinoma Exosomes

A stock solution of 1013 Hep G2 exosomes/mL was prepared before starting the

selection. In order to initiate the selection process, a DNA library was designed using

the parameters described in the Materials and Methods section. A 70 nt long ssDNA

library with a randomized core of 30 nt flanked on both, the 3' and 5'ends by a 20 mer

fixed primer binding sites was designed, synthesized and HPLC purified. FITC-labeled

library was synthesized in order to use in binding experiments as the control sequence.

PCR efficiency is critical in cell-based SELEX because eluted DNA sequences

for each round of selection have to be amplified by PCR. In order to achieve a

reproducible and efficient PCR with minimum unspecific binding, the reaction conditions

must be optimized. Besides, in order to retain maximum number of Z and P nucleotides,

PCR cycle repeats should be kept minimum. The optimum annealing temperature for

primers was determined as 59°C and the following PCRs are performed accordingly.

The selection process is explained in detail in the experimental section of this

chapter. In total, 9 rounds were performed, and the details in terms of quantity of

Page 58: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

58

positive, negative extracellular vesicles and incubation time summarized in Table 3-2. A

liver cancer cell line, Hep G2, derived exosomes were used as the target molecule for

the positive selection, whereas human whole blood derived exosomes were used for the

counter selection. The exosomes and ASL beads were conjugated in advance, as

explained in the experimental part.

Selection was started by incubating 20 nmol of library at 85°C for 10 min followed

by a snap cooling on ice to force DNA sequences to form their kinetically most

accessible secondary structures. The library was then incubated with target exosome-

bead conjugate at 37°C for 30 min with rocking. In order to remove the unbound

sequences, the beads were washed with washing buffer twice and the bound

sequences on exosome-bead conjugate were recovered by centrifugation at 14,800 x g

for 1 min. After the washing steps the bound DNA was released by heat (85°C, 15 min)

and separated from the beads by centrifugation. This process is practiced from round 1

to 3 and with the beginning of 4th round counter selection was incorporated. The only

difference in the process is pre-incubating the enriched pool with the negative blood-

exosome beads first for 30 min at 37°C. After the incubation, the unbound survivors

were collected and the process described above was reapplied. The incubation step

was followed by a PCR amplification. Single stranded biotinylated DNA was recovered

from double stranded amplicon DNA by capturing on solid-phase streptavidin followed

by elution with 200 mM NaOH. This was repeated a total of 9 rounds and counter

selection incorporated with the round 4.

The enrichment of the library through successive selection rounds was monitored

by flow cytometry. As the selection progresses, the number of sequences binding to the

Page 59: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

59

target cell line increases. Therefore, the enriched pool shows an increase in mean

fluorescence intensity. Once the pool shows no increase in fluorescence intensity, the

pool has been fully enriched and the selection has been completed. The first monitoring

was performed after round 6, for rounds 3,4,5 and 6 (Figure 3.4) An enrichment was

observed for rounds 3 and 4 but lost in round 5 and 6 (Figure 3-4A). We suspect this

might be due to the increase in negative beads and thus repeated the rounds 5 and 6

with a smaller number of blood exosome-bead conjugate. We felt confident when doing

so, since there was no binding between any enriched pool and negative bead-exosome

conjugates (Figure 3-4B). The bulk affinity increased from rounds 6 to 9, where no

enrichment was observed for negative control beads (Figure 3-5). The selection was

ceased at Round 13 and the sample was prepared for deep sequencing.

Deep Sequencing of GACTZP DNA Survivors Using Next Generation Sequencing Technology

Solutions containing enriched GACTZP DNA survivors after the 9th round of EV-

SELEX were divided into two equal parts. These were separately converted into

standard DNA under two conversion conditions using primers that carried barcodes for

the Ion Torrent deep sequencing. Following conversion, the samples were combined,

purified by native agarose gel, and submitted for Ion Torrent S6 “next generation”

sequencing at the University of Florida, ICBR sequencing core facility. The products

were aligned to identify sequences derived from a single common aptamer ancestor.

Ion Torrent sequencing reads that did not contain exact matches to the barcode,

forward and reverse priming sequences were discarded. To minimize miscalling, any

read present in less than 45 copies was removed from the analysis. The remaining

reads were then clustered using software custom designed at the FfAME, which ignored

Page 60: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

60

differing barcodes during the clustering and accepted single-step changes within

sequence reads. Clustered sequences were then separated by barcode, with variable

sites being compared between each barcode (differentiating the two conversion

conditions). The clustered sequences obtained under the first conversion conditions

(Barcode A, Z to C and P to G conversion) serve as reference for the clustered

sequences obtained under the second conversion conditions (Barcode B, Z to T/A and

P to C/G conversion). Sites where C and T were found in approximately equal amounts

after conversion under the second conditions were assigned as Z in their “parent”. Sites

where G and A were found in approximately equal amounts after conversion under the

second conditions were assigned as P in their “parent”. The eight most abundant

families were given in Table 3-3.

Discussion and Conclusion

EVs are becoming increasingly important as a source for novel cancer diagnostic

tools. Early detection of cancer is crucial to reduce mortality and increase survival.

Thus, it is essential to explore novel biomarkers that can distinguish cancer patients

from normal individuals. In some solid tumor types, certain biological proteins in the

body fluids, especially in the blood, offer such biomarkers, such as carcinoembryonic

antigen for colon cancer92 and prostate-specific antigen for prostate cancer93. However,

low specificity is a serious problem for these cancer biomarkers. For this reason,

additional biomarkers and detection tools are required to establish better early detection

methods. In this regard, EVs are expected to be noninvasive biomarkers for cancer

since they are present in various body fluids including serum, plasma, urine, and

contain a series of biological molecules that reflect the physiological and pathological

status.47,94,95 Also, source specific markers that represent the proteome of the cell of

Page 61: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

61

origin can also be used for exosome identification. For example, urinary exosomes of

patient’s with non-small cell lung cancer were found to carry proteins representative of

their primary tumor.96 Taken together, it is urgently necessary to develop biomarkers

targeting specific exosomes for clinical usage.

Since their discovery, aptamers have been generated against various targets,

including proteins,97 peptides,98 and living cells.89,90,99

The most tedious part of the project was the exosome collection experiments. It

took nearly two months to collect all the required number of exosomes from Hep G2

cells before starting the selection process. As discussed in the previous chapter, one

batch of exosome collection requires several, minimum two hours long ultra-

centrifugation steps. Besides, it includes culturing and maintaining a minimum of 15 T-

182 flasks of cells, which is a laborious task. On the other hand, it was crucial and

essential to perform each round of SELEX with the same batch of exosomes in order to

keep the consistency of positive selection. Otherwise, we might introduce new target

molecules to each round of selection. Although we optimized the exosome collection

process at its best to obtain a homogeneous population, it would not be fair to claim that

all the collected vesicles were exosomes. Hence, an immunoaffinity capture step with

exosome specific antibodies might be necessary to isolate pure exosome samples.100

On the other hand, this would be very costly, especially if the sample size is big like in

this case.

The top three sequences with the highest percentile were synthesized only, since

the relative percentages of the other sequences were less than 1% and were

considered less important. On the other hand, that might not reflect the absolute truth.

Page 62: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

62

There is a possibility that better aptamers (with a better specificity and/or affinity) might

be present in lower percentiles. It is actually more likely in this case than a regular cell-

SELEX, if we consider the fact that we kept the round number as small as possible and

terminated the selection process immediately after observing a significant

binding/enrichment. An average cell-SELEX takes 10-20 rounds101 thus the enrichment

percentile is usually higher for certain candidates. Nevertheless, a computational and

bioinformatics modelling might provide researchers a better insight about selection

process or potential aptamer candidates with lower percentiles.

Page 63: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

63

Table 3-1. Sequences used for EV-SELEX

Oligonucleotides Sequence

Library 5’-CCTTCGTTGTCTGCCTTCGTCGACT-N30-

ACCCTTCAGAATTCGCACCA-3’

Forward Primer 5’FAM-CCTTCGTTGTCTGCCTTCGT-3’ Tm 57.6C

Reverse Primer 5’BIO-TGGTGCGAATTCTGAAGGGT- Tm 56.8C

Table 3-2. Summary of EV-SELEX process

Round # PCR Cycle #

Target Exo-Beads

(µL)

Target Incubation Time (min)

Negative beads (µL)

Negative Incubation Time (min)

Round 1 25 100 30 N/A N/A

Round 2 24 100 N/A N/A N/A

Round 3 17 50 30 N/A N/A

Round 4 18 50 30 50 30

Round 5 14 40 30 50 30

Round 6 15 40 30 100 30

Round 7 16 50 30 50 30

Round 8 13 50 30 100 30

Round 9 16 50 30 100 30

Round 10 15 50 30 100 30

Page 64: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

64

Table 3-3. Compendium of the aptamer candidates selected by EV-SELEX against Hepatocellular Carcinoma Cells (Hep G2). Primer parts are removed and only the random sequences are provided. Artificial bases are marked as bold.

Aptamer Candidates Sequence

SEV1 5’-ACCCGCPCCGTCACACATCACACCGCCGCZ-3’

SEV2 5’-CCCPCACZPTGCTGCCCGTTACCACTCTCC-3’

SEV3 5’-CPCZGCCCCTTGTCTCCATCAGGCGGCCCC-3’

SEV4 5’-CCZTPCPCCCTGTCCAGTCGGCGGCCTTGT-3’

SEV5 5’-CCPCCACCTZGCTCCTTACATCGGCCTCAC-3’

SEV6 5’-CCPGCTCZTCACTATCCCTTGTCTGTCTCC-3’

SEV7 5’-CCPGCZCTGTCCCCAZCTCACCTGCCCTTA-3’

SEV8 5’-CCCZACPTCTGCCGTCTGTGTCCTGCCCGC-3’

Page 65: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

65

Figure 3-1. Schematic of EV-SELEX with both positive and negative selections. Briefly, ssDNA library is incubated with target bead conjugated-exosomes (Hep G2). After washing the unbound sequences, the bound DNAs are eluted by heating in binding buffer. The eluted DNAs are then incubated with control exosomes (counter selection, blood exosomes) for counter-selection. After centrifugation, the unbounded ssDNAs in supernatant are collected, and then amplified by PCR. The amplified DNAs are used for the next round of selection. The selection process is monitored using fluorescent analysis by flow cytometry. Once an enrichment has achieved, the final pool is sequenced.

Page 66: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

66

Figure 3-2. PCR applications of EV-SELEX. A) Agarose gel electrophoresis image of annealing temperature optimization experiment. 59°C was selected as the optimum annealing temperature B) PCR conditions schematic C) Agarose gel electrophoresis image of cycle optimization experiment.

Page 67: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

67

Figure 3-3. Verification of the enrichment of the library in binding sequences after 6 rounds. The DNA solutions generated at rounds 3, 4, 5 and 6 were incubated with both A) the target exosomes and B) the negative exosomes to verify the binding profile.

Page 68: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

68

Figure 3-4. Monitoring the progress of EV-SELEX using flow cytometer. The binding affinity of survivors was monitored in bulk from 6th round up to 9th round of selection. The vertical axis (Events) indicates the number of cells counted having the fluorescence intensity indicated by the horizontal axis. A higher intensity indicates a larger number of fluorescein labelled aptamer candidates bound per cell. A) Binding profile of the enriched pools with target exosomes B) Binding profile of the enriched pools with negative exosomes.

Page 69: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

69

CHAPTER 4 IDENTIFICATION OF DNA APTAMER ANALOGS IN GENOMIC DNA1

Introductory Remarks

Protein Tyrosine Kinase 7

PTK7 is an ancient and conserved protein.102 It was characterized in 1995 103

and various mutation experiments have identified a central role for PTK7 in a process

called planar cell polarity (PCP).104,105 As with many proteins important for development,

dysregulation of PTK7 expression is also a factor in many different cancers especially

colon cancer106, leukemia107 and melanoma.108 Structurally, PTK7 is a transmembrane

molecule; it consists of seven extracellular immunoglobuline-like domains, a single

transmembrane region and intracellularly, a kinase domain. This protein is structurally

unique among tyrosine kinases because its extracellular domain is composed of only Ig

domains, which in humans and zebrafish contain a matrix metalloprotease (MMP)

cleavage site. Besides, its transmembrane domain is more conserved than those of any

other receptor tyrosine kinase and its catalytically dead pseudo Tyrosine Kinase

domain, is also inert in all known orthologs.109 The extracellular portion of PTK7 is

composed of seven Ig-like domains, held together with an Ig fold. There is a MT1-

MMP/MMP14 cleavage site in the extracellular domain of PTK7 between the sixth and

seventh immunoglobuline domain.110 MT1-MMP is a membrane-bound endopeptidase

that cleaves extracellular matrix (ECM) proteins, other soluble MMPs and other

membrane-bound signaling receptors, including, PTK7.

1 Portions of work done this chapter were completed by Dr. Meghan Altman, a previous Tan Group

member, and were reproduced with her permission.141

Page 70: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

70

It has been shown that it is overexpressed in cancer, with a higher level of expression

result in more invasive and malignant tumors.111,112 Moreover, Golubkov et al. (2010)

confirmed that MT1-MMP cleaves PTK7 into two pieces: a 50kDa C-Terminal

membrane-bound pTK fragment and a 70kDa soluble PTK7 (sPTK7) fragment made up

of the first 6 Ig-like domains. They found these sPTK7 fragments in the cell media and

bound to full-length PTK7 on the surface of the cells. Following this work, Strongin

group followed up on this work by characterizing a mutant PTK7 protein with not one,

but two MT1-MMP cleavage sites. This mutant PTK7 protein was made in a mouse

strain, chuzhoi (chz), created by exposing the mice to the mutagen Nethyl-N-

nitrosourea. The mutant mice showed signs of classic PCP signaling problems,

including neural tube defects and disrupted hairs in the inner ear.113 In addition to the

normal MT1-MMP cleavage site in the 7th Ig-like domain, this mutant’s PTK7 contained

a second cleavage site in the linker between the 5th and 6th Ig-like domain.

Wnt Signaling

PTK7 is one of the key elements of Planar Cell Polarity (PCP), a process

controlling uniformly polarized cellular behavior across the plane of the

epithelium104,105,113 and is very critical for many developmental processes.114 PCP is

regulated by the Wnt signaling pathway, where Wnts comprise a group of signaling

molecules, controlling many processes of development and adult tissue homeostasis

through several different pathways. They interact with various receptors and therefore

activate different downstream pathways. Even though the actual situation is more

complex, the Wnt pathways can be simply classified as -catenin dependent and -

catenin independent pathways, which in turn can be subcategorized as PCP/Wnt and

Page 71: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

71

Wnt/Ca2+ signaling. In the -catenin pathway, Wnt ligands bind to cell-surface receptor

Frizzled (Fz) which in turn recruits the co-receptor low-density lipoprotein receptor-

related protein5/6 (LRP5/6) on the cell surface to bind the cytoplasmic protein

Disheveled (DVL) Through several cytoplasmic cascades the signal is transduced to -

catenin, which enters the nucleus, where it activates the transcription of target genes

under the control of T cell factor (TCF), among others. These genes are mainly involved

in regulation of cell differentiation and proliferation.115,116 On the other hand, as its name

implies, -catenin independent pathways use other ways of downstream signaling

instead of -catenin-TCF. These non-canonical Wnt signaling pathways control cellular

polarity and cell movement, and they signal, for example, via small GTPases of the Rho

family resulting in modification of the cytoskeleton 117. PCP/Wnt signaling is the best

characterized -catenin independent pathway. However, although Wnt ligand binds to

Fz and activates DVL, the co-receptor recruited is different. Upon binding of PCP/Wnt

ligand to Fz instead of LRP5/6, PCP/Wnt co-receptors are activated. Several functional

studies point to a role for PTK7 as a PCP/Wnt co-receptor determining PCP.118,119

Please refer to Figure 4-1 for wnt signaling pathway schematic.

Background and Significance

Cell-SELEX generates artificial, single-stranded DNA and RNA molecules, called

aptamers, that can bind specifically to a cell type of interest. It is a well-established,

iterative method 60 and has been used to select aptamers mainly targeting whole cancer

cells, such as colon86, leukemia120, liver90, lung121 and other cancers. Each whole cell-

SELEX uses a large (ca 1015) library of random DNA sequences usually having less

than 100 nucleotides, flanked by primers. The target cell membranes may contain as

Page 72: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

72

many as 10000 proteins122, any of which could be attracted to one or more of the library

sequences. During the iterative binding process, cells are equilibrated with library

sequences and only the bound sequences are retained and PCR amplified for the next

round equilibration, separation, PCR. Binding with target cells is alternated with

equilibration with a different cell line (negative selection) to remove sequences that bind

to common membrane markers. After an enrichment achieved, the binding sequences

are amplified and then subjected to Next Generation Sequencing.

Sgc8 is one of the DNA aptamers selected by whole-cell SELEX for a target T-

Cell leukemia cell line, CEM-CCRF, using Ramos B-Cell leukemia cells for the negative

cell line. Further investigations revealed that Protein Kinase 7 (PTK7) is the specific

target on the CEM-CCRF cell surface surface.123 PTK7 is an ancient and conserved

integral membrane protein with 7 immunoglobulin-like domains that hang outside the

cell, a single transmembrane domain, and a tyrosine pseudokinase domain inside the

cell. Several studies have shown overexpression of PTK7 in colorectal tumors 124,

erythroleukemia125 acute myloid leukemia126, glioma127 and prostate cancer. 128 In

contrast, PTK7 is downregulated in some other cancers, such as melanoma108,

ovarian129 and hepatocellular carcinoma.130 In addition, it has been recently shown that

there is an MT1-MMP endopeptidase cleavage site in the 7th Ig fold, above the

transmembrane domain, which cleaves the protein into two parts: membrane bound and

soluble PTK7 (sPTK7).110

Functionally, PTK7 is one of the key elements of Planar Cell Polarity (PCP), a

process controlling uniformly polarized cellular behavior across the plane of the

epithelium,104,105,113 and is very critical for many developmental processes.114 PCP is

Page 73: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

73

regulated by the Wnt signaling pathway, where Wnts comprise a group of signaling

molecules, controlling many processes of development and adult tissue homeostasis

through several different pathways. They interact with various receptors and therefore

activate different downstream pathways. Even though the actual situation is more

complex, the Wnt pathways can be simply classified as -catenin dependent and -

catenin independent pathways, which in turn can be subcategorized as PCP/Wnt and

Wnt/Ca2+ signaling. In the -catenin dependent pathway, through several cytoplasmic

cascades the signal is transduced to -catenin, which enters the nucleus, where it

activates the transcription of target genes under the control of T cell factor (TCF),

among others (Figure 4-1).116 These genes are mainly involved in regulation of cell

differentiation and proliferation.115,116 Among many others, a scaffolding protein ,DIX

Domain Containing 1 protein (DIXDC1) has recently been shown as the positive

regulator of -catenin dependent pathway.131,132 On the other hand, as the name

implies, -catenin independent pathways use other ways of downstream signaling

instead of -catenin-TCF. These non-canonical Wnt signaling pathways control cellular

polarity and cell movement, and they signal resulting in modification of the cytoskeleton

117 PCP/Wnt signaling is the best characterized -catenin independent pathway where

PTK7 acts as one of the main co-receptors. Several functional studies point to a role for

PTK7, which shifts the pathway to PCP/Wnt signaling upon its activation by certain Wnt

lignads.118,119

In this work, we investigated the possibility of DNA aptamer analogs in genomic

DNA. Analysis of DNA sequences, selected independently against different target cells

by whole cell-SELEX, identified 4 aptamers, including sgc8, with a common target

Page 74: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

74

protein, PTK7. Deeper investigation of these aptamer sequences revealed a 15 base

consensus region, which was searched for a match in the human genome.

BLAST results revealed a sequence identity of the consensus region minus one

G base on the 5’ untranslated region (5’-UTR) of DIXDC1 coding gene. As explained

above, PTK7 and DIXDC1 are both regulating Wnt signaling pathway but they have

opposite effects; PTK7 switches the pathway toward PCP/Wnt signaling whereas

DIXDC1 favors -catenin dependent pathway. These results revealed the possibility that

PTK7 is interacting with DIXDC1 gene perhaps in order to regulate expression of the

latter.

Results

Sequence Similarity between Different Aptamers

Close examination of several different cell-SELEX aptamers, targeting different

cancer cell lines and selected independently at different times by different researchers,

revealed binding patterns similar to that of sgc8. Sequence alignment by CLUSTELx133

revealed that aptamers sgc8 and KC2D886 share 38 consecutive nucleotides, the former

selected against T-Cell leukemia cell line CCRF-CEM and the latter independently

against colorectal adenocarcinoma DLD1. In light of this evidence, a dataset of all DNA

aptamers identified by complex selection or against membrane-bound targets was

constructed. This yielded 148 unique aptamer sequences from 33 different selections.

The aptamers’ primer regions were removed to avoid interference during alignment, and

their correspondences were determined by alignment with ESPRIT134 The results

revealed two more aptamers, H01 and KMF9b88, having sequence identity with each

other, as well as with sgc8 and KC2D8. When these sequences were aligned with sgc8,

Page 75: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

75

their similarities were clustered around a core 15nt GC rich region:

GCTGCGCCGCCGGGA. (Table 4-1).

Competition Experiments

Even though the four aptamers (sgc8, KC2D8, KMF9b and H01) have similar

affinities to the same cancer cell lines and share a consensus region, it was still

necessary to determine whether they bind to the same target molecule on the target

cell, PTK7 in this case. In order to do so, all aptamers were subjected to a competition

assay against sgc8. According to the results, all four aptamers competed with each

other but not with a control sequence, scrambled-sgc8 (Figure 4-2). In addition, it was

clear that H01 was driven off the cell surface faster than the other aptamers. Addition of

unlabeled sgc8 first, followed by H01 labeled with biotin (H01-B, purple line) showed no

binding of the label by flow cytometry, but addition of unlabeled H01, followed by sgc8

labeled with (sgc8-B, dark blue line) or addition of unlabeled H01 followed H01-B (light

blue line) resulted in reduced binding, but to a much lower extent. This means, if

unlabeled sgc8 binds PTK7 first, H01 is not able to bind the PTK7. By contrast, H01 is

readily replaced by both biotinylated sgc8 and 10x unlabeled H01.

BLAST of the Consensus Sequence Against the Human Genome

In light of bioinformatic analysis and competition experiments, it was

hypothesized that these aptamers may be mimics for a naturally occurring interaction

between PTK7 protein and natural DNA. If this is the case, the consensus sequence

should be found in a target sequence in the DNA. To investigate this possibility, the

consensus sequence was BLASTed against the human genome using the NCBI

nucleotide BLASTn algorithm, adjusted for short sequences. Eight matches were

identified with a 14/15 nucleotide identity. Of these, Dix Domain Containing 1 protein

Page 76: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

76

(DIXDC1b) was the only one which carries the consensus sequence on the 5’

untranslated region (5’ UTR). and which is also related to the Wnt signaling pathway

together with PTK7. In light of these findings, PTK7-aptamers were aligned with

DIXDC1b DNA, which contained a 1nt mismatch with sgc8 and KC2D8, 3nt mismatches

with KMF9b, and 5nt mismatches with H01. Although H01 and KMF9b had more

mismatches within the consensus region, they both had additional bases in common

with the DIXDC1b DNA adjacent to the consensus sequence (Table 4-3). Moreover,

another sequence match between KC2D486 aptamer and the negative strand of

DIXDC1b DNA was detected (Table 4-3). Despite the fact that KC2D4 also competes

with sgc8, no sequence identity was observed with PTK7 aptamers. Surprisingly,

CLUSTALx analysis revealed an 11 nucleotide match between DIXDC1b DNA and

KC2D4 five nucleotides downstream of the consensus region on positive DIXDC1b

DNA.

Investigation of the Interaction of PTK7 with DIXDC1b DNA

A western blot was performed in order to demonstrate nuclear localization of

sPTK7. Cell fractionation of PTK7 overexpressed cells (HEK293, HeLa) was followed by

anti-PTK7 probing, as well as by cytoplasmic and nuclear control antibodies. According

to the results, cleaved PTK7(sPTK7) was detected in all three fractions, and at much

higher levels in the nuclear fraction, whereas full length PTK7 was observed at higher

levels in the pellet, which would contain the membrane (Figure 4-3). In addition, a gel

shift experiment was designed to show the interaction between PTK7 and the DNA

coding DIXDC1. An 87 base part of DIXDC1b DNA which included the consensus

region was PCR amplified with biotin labelled primers and incubated with nuclear

extracts from HeLa or Ramos (negative control) cells. Then, the nuclear extract-probe

Page 77: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

77

complexes were separated on a native gel and observed with a typhoon scanner. The

shift proved the interaction of the sequence of interest with the HeLa nuclear extract

(Figure 4-5). No shift was observed for the nuclear extract of Ramos, which does not

express PTK7. In addition, confocal immunohistochemistry experiments showed the

colocalization of sgc8 aptamer inside the nucleus. HeLa cells were incubated with

TAMRA labelled sgc8 and a random sequence. DAPI was used for nuclear staining.

After 2 hours of incubation, aptamers can be observed inside the cell, as well as inside

the nucleus (Figure 4-4B). On the other hand, no fluorescence was observed for control

sequence, meaning no internalization. (Figure 4-4A)

Discussion and Conclusion

By means of cell-SELEX technology, the Tan Group has generated a number of

DNA aptamers targeting cancer cells. One of the most widely used is Sgc8, which was

discovered to bind the extracellular part of PTK7, which has a central role in PCP/Wnt

signaling. It was later found that several different aptamers, selected against different

cell lines with different starting primer sets, showed binding affinity very similar to that of

sgc8 towards PTK7 overexpressing cell lines. In order to investigate this phenomenon

deeper, a series of bioinformatics and competition analyses were conducted. A total of

148 aptamer sequences selected by whole cell-SELEX revealed significant sequence

identity between sgc8 and 3 other aptamers (KMF9b, KC2D8, H01) within a 15nt

consensus region (GCTGCGCCGCCGGGA). As predicted, all 4 aptamers competed

with each other and bound to the cells with similar affinity, implying that they bind to the

same place on the PTK7 protein. Repeating the same selection and selecting the same

sequences by coincidence is unlikely for several reasons: First of all, each selection

started from a large pool of potential targets because, neglecting lipids and peripheral

Page 78: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

78

proteins and considering integral proteins alone, 20-30% of human proteins have a

transmembrane domain122 and a significant portion of these proteins are exported to the

cell surface. This means that upwards of 10,000 different proteins are expressed on

each cell's surface. In addition, these proteins can be further modified by

carbohydrates, creating many more sites for possible aptamer interaction and

subsequent selection. (An ovarian cancer selection that produced aptamers insensitive

to proteases has been postulated to bind these glycoproteins. 85) Second, a large

aptamer library with different primers is used for each selection, calculated to include

1015 unique sequences. Thus, there are so many possible sequence/target

combinations that random coincidence is unfeasible. Third, aptamers are routinely

selected against a wide array of specific single targets (small ions, peptides, and

purified proteins). A search of the literature did not reveal a single target that evaded

selection. Building on this, it is assumed that each potential target on the cell surface

had an equal probability of binding one of the sequences in the library.60

The similarity between all four PTK7 aptamers indicated that one of the above

considerations did not hold. Since the first two are not questionable, the third

consideration may not be completely applicable to cell-SELEX. Instead of detailed,

complete profiling of the cell surface, perhaps cell-SELEX preferentially selects for

aptamers with a biological function; for instance, those with an underlying affinity for

DNA binding.

In order to investigate the repetitive selection of the consensus sequence, it was

BLASTed on genomic DNA. According to the results, the consensus region, minus the

first G base, appears 5 times in the human genome. One of these sites is in the 5'-

Page 79: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

79

untranslated region (5’-UTR) of human DIXDC1b DNA, which, along with PTK7, is a

regulator of non-canonical Wnt signaling. DIXDC1b is one of the three known DIX

domain-containing proteins and is a positive regulator of -catenin-dependent Wnt

signaling.135–137 It has been previously demonstrated that downregulation of DIXDC1, as

in squamous cell carcinoma of the lung, leads to aberrant upregulation of Wnt/PCP

signaling138. Considering that PTK7 is one of the co-receptors of the PCP/Wnt pathway,

we can conclude that PTK7 and DIXDC1 have opposite effects on Wnt signaling. PTK7

switches Wnt signaling from being β-catenin dominant to being PCP dominant, whereas

DIXDC1 switches Wnt signaling from being PCP dominant to being β-catenin dominant.

As discussed in the Results section, aligning DIXDC1b DNA with four PTK7

aptamers revealed additional sequence identities in addition to the consensus region. In

addition, a new sequence match between the DIXDC1b DNA negative strand and

KC2D4 was discovered. In total, DIXDC1b DNA and PTK7 aptamers share 22 identical

nucleotides, which is a unique occurrence in the human genome. As a result of these

findings, the aptamers’ sequence identity to both the positive and negative strands of

DIXDC1b DNA could be consistent with the protein PTK7 melting the genomic DNA and

interacting with the resulting ssDNA hairpins, which share sequence identity to the

aptamers formed by each melted strand. If this possibility is indeed the case, it could

affect DIXDC1b transcription.

Further, nuclear localization of sPTK7 was demonstrated by cell fractionation

followed by western blot. A large accumulation of sPTK7 was demonstrated inside the

nucleus, which perhaps is evidence for the interaction of PTK7 with the DIXDC1b gene.

Consistent with this hypothesis, a matrix metalloprotease cleavage site that frees the

Page 80: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

80

extracellular portion of PTK7 has been recently reported. Adding this protease to PTK7-

expressing cells caused PTK7 to be removed from the cell membrane and to

accumulate around the nucleus.110 On the other hand, the presence of a faint full length

PTK7 band in the nuclear fraction is inconsistent with accumulation of only the

extracellular portion around the nucleus. This weak full-length band may have occurred

due to leakage during fractionation. In addition, a gel shift experiment showed binding

between the PCR amplified dsDIXDC1b DNA including consensus region and the

nuclear extract of HeLa cells. Together with the nuclear localization of sPTK7, this may

be evidence for the interaction of PTK7 with genomic DNA to alter DIXDC1b

expression.

Materials and Methods

Unless specified otherwise, all the reagents were purchased either from Thermo-Fisher

or Sigma Aldrich.

Buffers and Cell Culture

Washing buffer (WB) contained glucose (4.5g/L) and magnesium chloride (5mM)

in 10mM Dulbecco’s phosphate buffered saline. Binding buffer (BB) was prepared by

adding bovine serum albumin (1mg/mL) and transfer ribonucleic acid (tRNA)

(0.1mg/mL) to washing buffer. All cell lines were obtained from American Type Culture

Collection (ATCC). CCRF-CEM (CCL-119, a T-cell line, Acute Lymphoplastic Leukemia)

and Ramos (CRL-1596 a B-cell line Burkitt’s lymphoma) cell lines were cultured in

RPMI-1640 medium (Sigma) with 10% FBS (GIBCO) and 100 units/mL penicillin-

streptomycin (Cellgro) at 37°C under 5% CO2. HeLa (CCL-2, human cervical

adenocarcinoma) and HEK 293 (CRL-1573) cell lines were cultured in DMEM-1640

Page 81: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

81

medium (Sigma) with 10% FBS (GIBCO) and 100 units/mL penicillin-streptomycin.

Cultures were routinely monitored for Mycoplasma infection.

DNA Sequences

All DNA sequences were synthesized in-house at a 1 μmol-scale using an ABI

3400 DNA/RNA synthesizer (Applied Biosystems). Sequences were deprotected from

CPG beads with AMA (ammonium hydroxide: methylamine 1:1) at 60C for 30 minutes.

All sequences were either unlabeled or labeled at the 3’ end with biotin and re-

suspended in 1M TEAA and purified on a reverse phase Prostar HPLC (Varian) using a

C18 column (Econosil, 5U 250 x 4.6 mm from Alltech Associates) with a linear elution

gradient of acetonitrile:trietylammonium acetate. These sequences were vacuum dried,

detritylated with 20% acetic acid and re-suspended in 1M TEAA buffer. The

concentration was determined by UV-vis spectrophotometry (Beckman Coulter DU800)

at 260nm. The primers used for the experiments were ordered from Integrated DNA

Technology (IDT).

Bioinformatics

A dataset of 148 aptamer sequences from 33 different cell-SELEX procedures

was compiled with their primers removed and aligned with ESPRIT134. The p-values

were calculated by comparing each alignment score with those of 1,000,000 random

pairs of the same length. Using the convergent property of SELEX139, the p-value (p)

obtained in our simulation indicated that, given a target sequence S1, a reference

sequence R, which has a known similarity value t to S1, and a random sequence S2,

with the same length as R, the probability that S2 is at least as similar as R to S1 is p.

Under this non-deterministic model, we supposed we have n (here n ~10,000) aptamers

in the system. The probability that a random aptamer can be more similar than R is 1-

Page 82: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

82

(1-p)n. If we examine k targets (here k=148), the probability of one random hit will be 1-

(1-p)nk, which is approximately nk*p when p is small. Hence, if nk*p << 1, we claim that

R is not likely a random match. This means that R should be identical, or at least

correlated to S1. Ranked by p-value, sgc8 and KC2D8 had the highest alignment, and.

KMF9b showed sequence identity to sgc8. Based on this result, a GenBank BLASTn140

was performed on the consensus region: GCTGCGCCGCCGGGA.

Significance Simulations

According to classical probability law, the probability that a random pair of

sequences has the same sequence similarity with a real pair assigned p-value p, is

P0=n(n-1)/2*p with n sequences. Following this law, KC2D8 is a non-coincident match,

with P0<0.01. The probability that a third sequence has that similarity is P1=2*(1-P0)*(n-

2)* p. Here KMF9b is matched to sgc8-KC2D8, with significance level P1<0.03. Thus,

each of the 3 sequence alignments cannot be explained by coincidence, and the

common region between the three PTK7 aptamers and DIXDC1b DNA is unique in the

human genome. The probability that another random sequence could match the same

DNA in adjacent regions was examined by simulating 1,000,000 random 39nt long

sequences, and aligning them with FASTA to a 52bp segment in the 5’-UTR of

DIXDC1b DNA.

The simulation showed that 2,554 sequences out of 1,000,000 sequences, or

their reverse complement, have at least one 13bp window sharing at least a 12nt

identity with the DIXDC1b segment, the same as KC2D4. Hence, the chance that a

random sequence has the same similarity to the target region is p=0.0025.

Page 83: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

83

Competition Assays

For each aptamer-competition experiment, cells were washed with washing

buffer, probed with the first aptamer followed by the second aptamer, washed, stained

with 1:400 streptavidin-PE, washed again, and analyzed using a FACScan flow

cytometer (Becton Dickenson). Aptamer treatments were: no aptamer; 80nM scrambled

sgc8-biotin; 80nM aptamer-biotin; 800nM unlabeled-aptamer, then 80nM aptamer-

biotin; 800nM unlabeled-sgc8, then 80nM aptamer-biotin; and 800nM unlabeled-

aptamer, then 80nM sgc8-biotin. The results revealed that scrambled-sgc8 (scr-sgc8)

does not compete with sgc8, while all the other aptamers do.

Western Blot

HEK293 cells were fractionated using a NE-PER kit (Pierce), run on a 4-20%

Tris-Glycine gel (Invitrogen), and probed with α-PTK7 M02 (Abnova), then re-probed for

α-GAPDH (ABcam), a cytosolic marker, and Lamin B1 1:1000 (ABcam), a nuclear

marker. Then the membrane was treated with, α-Rabbit HRP 1:5000 (Pierce) and

imaged on Kodak film.

Gel Shift Assay (EMSA)

CCRF-CEM genomic DNA from 5x106 cells was extracted and RNase treated.

An 87 bp long ds DNA surrounding the consensus region on DIXDC1b was PCR

amplified with 5’-biotin tagged primers and the PCR product was purified via purification

kit (Qiagen, Valencia, CA). A small scale nuclear extract method was used for HeLa and

Ramos (negative control) cells and protein concentration was determined by BCA

assay. A LightShift Chemiluminescent EMSA kit from Pierce (Thermo Fisher) was used

for the binding experiment according to manufacturer’s instructions. The reactions were

loaded on 6% non-denaturating neutral polyacrylamide gel and transferred to a nylon

Page 84: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

84

membrane. The biotinylated oligonucleotides were detected with streptavidin-linked

horseradish peroxidase with a typhoon scanner.

Page 85: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

85

Table 4-1. PTK7 aptamer sequences with their identical nucleotides marked as bold. Primer sets used for the selection of aptamers were included

Aptamer Primers Sequence

sgc8 F-ATACCAGGCTTATTCAATT R-GATAGTAAGTGCAATC

ATCTAACTGCTGCGCCGCCGGGAAAATACTGTACGGTTAGA

KMF9b F-AGGCGGCAGTCTCAGAGT R-CTGAGCGACGAAGACCCC

AGCGCAGCAGCTGTGCCACCGGGAGAATTTACGTACGGCTGAGC

KC2D8 F-ATCGTCCGCCACCACCACTACTC R-GTGAGACTGCCTGCCTGCCGATGT

TACTAACTGCTGCGCCGCCGGGAAAATACTGTACGGTTAGTT

H01 F-ATCGTCTGCTCCGTCCAATAT R-TTTGGTGTGAGGTCGTGC

AAGCAGCAGCTGTGCCATCGGGTTCGGATTTTCTTCCTACGACT

Page 86: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

86

Table 4-2. BLAST hits 14/15nt identity for consensus sequence (GCTGCGCCGCCGGGA) in human genome

Protein Name, Abbreviation Locus Location in Gene

DIX Domain Containing 1, isoform b, DIXDC1b NM_033425.3 5’ UTR

Mucolipin 1, MCOLN1 NM_020533.2 Coding region

Membrane-bound transcription factor peptidase NM_003791.2 Intron

Ubiquitin-conjugating Enzyme E2D2, UBE2D2 NM_003339.2 Intron

Myc Induced Nuclear Antigen, MINA NM_032778.4 Intron

SH3 and multiple ankyrin repeat domains protein NT_011109.16 Intron

Nearest protein KIAA1875 NR_024207.1 Unknown

Nearest miRNA MIR302F NW_001838467.2 Unknown

Page 87: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

87

Table 4-3. Aptamers share sequence similarity with DIXDC1b DNA sequence. Lettering in bold shows identity inside

consensus sequence with DIXDC1b positive strand DNA (+); letters in square brackets show additional bases in common after alignment with DIXDC1b DNA; letters in curly brackets show identity with DIXDC1b negative strand DNA (-).

Aptamer Sequence

sgc8 ATCTAACTGCTGCGCCGCCGGGAAAATACTGTACGGTTAGA

KMF9b A[GCGCAGC]AGCTGTGCCACCGGGA[G]AATTTACGTACGGCTGAGCGA

KC2D8 TACTAACTGCTGCGCCGCCGGGAAAATACTGTACGGTTAGTT

H01 AAGCAGCAGCTGTGCCATCGGGTTCGGATTTTCTTCCTACGACT

DIXDC1b

DNA

+G[GCGCAGC]CTGCGCCGCCGGGA[G]CCTCCCTCCCAGTGGGAGATGGGTTGAGA

-CCGCGTCGGACGCGGCGGCCCTCGGAGG{GAGGGT}C{ACC}C{TC}TACCCAACTCT

KC2D4 {GAGGGT}G{AC}CA{TC}GGTAAGGCGGGAATTGGCCCGGTAGC

Page 88: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

88

Figure 4-1. Simplified schemes showing the main WNT pathways directed by specific

WNT, Frizzled and WNT co-receptor interactions. A) Planar cell polarity (PCP) signalling triggers activation of the small GTPases RHOA and RAC1, which in turn activate RHO kinase (ROCK) and JUN-N-terminal kinase (JNK) B) (GSK3) phosphorylates β-catenin, which triggers its degradation. However, in the presence of WNT ligand, the destruction complex is recruited to the WNT–receptor complex and inactivated. This allows β-catenin to accumulate and translocate to the nucleus, where it activates the transcription of target genes under the control of T cell factor (TCF), among others. C) The WNT–Ca2+ pathway activates Ca2+- and CAMKII, protein kinase C (PKC) and calcineurin. PCP and Ca2+pathways antagonize β-catenin signaling at various levels. D) Major pathways used by WNT receptors and co-receptors. Only the three best-characterized WNT pathways are shown. Figure adapted from Nature Reviews: Molecular Cell Biology116. Reprinting with permission Nature Publishing Group.

Page 89: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

89

Figure 4-2. Competition studies between different aptamers for PTK7.The aptamers

with sequence identity (sgc8, H01, KMF9b, KC2D8) were synthesized as unlabeled or biotin-labelled and detected by flow cytometry. Green line shows the binding of the aptamer of interest. Light blue line shows the binding when the target cells were saturated first with the unlabeled aptamer of interest (10x) and then incubated with labelled aptamer (1x). Purple line indicates the binding when target cells were saturated first with unlabeled sgc8 aptamer (10x) and labelled aptamer of interest (1x) and vice versa (dark blue line). A loss of binding indicates that the unlabeled aptamer (10x) occupied the target region and saturated it, so that the labelled aptamer could not bind and thus no signal was detected. Panel A shows the control experiment with a non-binding sequence and Panel B the experiments with aptamers of interest. B: Biotin, label; Scr-sgc8: Scrambled sgc8, a negative control; Apparent Kd for each aptamer on CCRF-CEM cells is given under the aptamer’s name.

Page 90: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

90

Figure 4-3. Interaction of PTK7 with DIXDC1b DNA. A) Western blot analysis of various

cellular compartments of HEK293 cells. Full-length PTK7 is ~118kDa, cleaved ~68kDa. GAPDH is a cytoplasmic marker. Lamin B1 is a nuclear membrane marker B)

Figure 4-4. Confocal immunocytochemistry image of HeLa cells co-stained for PTK7

with sgc8-TMR. A) HeLa cells were co-stained with a control sequence labelled with TMR. DAPI staining was applied as the nuclear staining. No nuclear signal for TMR was observed. B) HeLa cells were co-stained with sgc8-TMR. DAPI staining was applied as the nuclear staining.

Page 91: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

91

Figure 4-5. Electrophoretic mobility shift assay (EMSA) for the ds DNA surrounding the consensus region on DIXDC1b. A)

Schematic representation of the experiment rationale. B) EMSA gel image. Experiment was performed by binding of biotinylated oligonucleotides with either HeLa or Ramos nuclear extracts. Binding reactions as shown were performed using the LightShift Chemiluminescent EMSA Kit. Ebna control system, provided in the kit, was used as an experimental control.

Page 92: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

92

CHAPTER 5

CONCLUSIONS AND FUTURE DIRECTIONS

Summary and Conclusions

This material could be divided into two main parts: In the first part EV-SELEX

methodology was developed and utilized for the generation of a panel of aptamers

capable of distinguishing exosomes isolated from Hepatocellular carcinoma cell line,

HepG2, from exosomes isolated from human whole blood. However, before starting the

selection process, a serious of exosome isolation and characterization methods were

studied initially. The characterization experiments were actually a preparatory step for

the EV-SELEX. It was necessary to perform a quality check of the target material before

implementing a new methodology. Based on the NTA and western blot results, the

vesicles we isolated were less than 100nm in size and expressing CD 63 marker which

all consistent with exosomes. On the other hand, NTA results also revealed that there

are particles bigger than 200nm indicating the population we collected is not entirely

pure. Next, we used this exosome isolates

In the second part of this study, the interaction between PTK7 and genomic DNA

was elaborated. Whole cell-SELEX method uses a large library of random DNA

sequences (~1015) amplified by unique primers, against 10,000 unique protein targets.

As discussed in introduction part, sgc8c was selected by whole-cell SELEX for a target

T-Cell leukemia cell line, CEM-CCRF, and against a B-Cell leukemia cell line Ramos.

Another aptamer KC2D8, was selected several years later using a colon cancer cell

line, DLD1, as the target, and no negative cell line. In the process of analyzing colon

cancer aptamers he has selected, he noticed that several aptamers had the same

binding profile as sgc8c. These three aptamers, all bound the same cell types with

Page 93: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

93

similar affinities, and competed with each other for binding. CLUSTALx alignment

showed sgc8c and KC2D8 share 38 contiguous nucleotides. These two selections used

two different primer pairs to PCR-amplify the portion of the library. The sgc8 aptamer

cannot prime the KC2D8 library, making the chance of contamination by sgc8c during

KC2D8 selection unlikely. A bigger analysis was performed by comparing 148 different

ssDNA aptamer from more than 30 different selections using the software program

ESPRIT. When these sequences were aligned with sgc8c, their similarities were

clustered around a core 15nucleotide GC rich region: GCTGCGCCGCCGGGA

Future Directions

The future directions for the EV-SELEX part of this study could be approached

from two different angels: a) near future studies b) far future studies. So far, a novel

SELEX method was designed, optimized and performed against liver cancer exosomes

and the binding of the aptamers to target and counter exosomes were determined. On

the other hand, the project is still immature and requires a serious of characterization

experiments for the selected aptamers. First of all, binding affinities, Kd, of the selected

aptamers should be determined. In addition, specificity of the aptamers towards

different exosomes, collected from different cancer or normal cell lines should be

elaborated. Finding the target molecule of the selected aptamers on the vesicle could

be the next step of the project. This would also reveal the molecule, specific to the liver

cancer cells. All these experiments discussed so far are actually classic characterization

steps of a newly selected aptamers. The second and a bigger step could be testing

aptamers for clinical use. Firstly, the aptamers can be tested in liver cancer exosome

spiked human plasma in vitro. If the results are promising, then in vivo testing of the

aptamers should be considered. At this point, we are evolving our discussion towards

Page 94: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

94

far future studies. Once the aptamers passed all the characterization experiments, there

is a whole new world waiting for them to be discovered.

Generally, conclusions regarding the functional role of exosomes in facilitating

cell-to-cell communication are based on in vitro experiments in isolated culture systems

using concentrations of exosomes that are rough approximations and may not be

physiologically relevant. Therefore in vivo studies to address the functional role of

exosomes in cancer can be studied. Using exosomes to deliver drugs could also offer

new treatment options for the clinic.

Future directions for the second part of this dissertation would be understanding

the molecular mechanism behind the DIXDC1b –Wnt pathway. The progress in this

project is, unfortunately, very slow. The bioinformatic studies indicated that the selecting

aptamers sharing same sequence cannot be coincidence. Yet, proving the unusual

mechanism of PTK7 cleavage, internalization and interacting with genomic DNA to

change DIXDC1 expression is challenging, especially considering there is no such

example has been reported so far. One thing, that needs

In addition, this finding is probably not a one-time occurrence. Our bioinformatic

analysis of 148 DNA sequences selected by whole-cell SELEX identified other

aptamers from disparate selections, like those for Vaccinia infected cells and pure virus,

which share significant sequence identity. Further comparison of existing DNA and RNA

aptamers may yield other examples of SELEX identifying natural DNA or RNA

sequences with functional roles, not initially envisioned. Future selections should also

not be considered complete until the newly selected aptamers are compared with all

other existing aptamers for sequence identity.

Page 95: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

95

APPENDIX A COMPLEX TARGET SELEX DNA APTAMER DATABASE

Page 96: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

96

Table A-1. Complex Target SELEX DNA Aptamer Database

SELEX Target Name Nt Sequence

1 hnRNP-A1 BC-15 74

TGTGGCGAGGTAGGTGGGGTGTGTGTGTAT

2 IgE IgE 21

TTTATCCGTTCCTCCTAGTGG

3 small cell lung cancer 16-1 25

GAATCCTTCTTTGTCCCGGGCCCGT

small cell lung cancer 0-25 25

TACTCAATTACTCTCTTGTCCCTCT

4 Shp2 Phosphatse HJ24 80

GGGGTTTTGGTGGGGGGGGCTGGGTTGTCTTGGGGGTGGG

5 Tenascin-C GB-10 34

CCCAGAGGGAAGACTTTAGGTTCGGTTCACGTCC

6 Mucin S1.3/S2.2 72

GCAGTTGATCCTTTGGATACCCTGG

7 RET Kinase D24 50

GCGCGGGAATAGTATGGAAGGATACGTATACCGTGCAATCCAGGGCAACG

8 Nucleolin AS1411 26

GGTGGTGGTGGTTGTGGTGGTGGTGG

9 IL-17RA RA10-2 30

CTAAGGATCGGATCCACGGCCTACCAGGTC

IL-17RA RA10-6 30

CTTGGATCACCATAGTCGCTAGTCGAGGCT

IL-17RA RA10-7 30

ACGCGCTAGGATCAAAGCTGCACTGAAGTG

IL-17RA RA10-13 30

CCAGAAGAAGCCCACTAGCGTGCTTTTGTC

IL-17RA RA10-14 30

CCAGACGTGAGCACTAGATCAGTACGGAAG

10 NSCLC: A549 v HLAMP S1 45

GGTTGCATGCCGTGGGGAGGGGGGTGGGTTTTATAGCGTACTCAG

NSCLC: A549 v HLAMP S6 45

GTGGCCAGTCACTCAATTGGGTGTAGGGGTGGGGATTGTGGGTTG

NSCLC: A549 v HLAMP S11a 45

AGAGTGGGGGGGTGGGTGGATTTGACAGGTGGCATGCTGGAGAGT

NSCLC: A549 v HLAMP S11b 45

TGGGGTTATTAATTTTGGGTGGGGGGGAAGATGTAGCATCCGACG

NSCLC: A549 v HLAMP S11c 45

AGCTTGAGGGTGGGCGGGTGGACGCGGTAGTGGTATATAGGTCGG

NSCLC: A549 v HLAMP S11d 45

GATCGGTGGGTGGGGGGGTTGGAGATCATCCTCAGGGATTACGTC

NSCLC: A549 v HLAMP S11e 45

ATGCGAACAGGTGGGTGGGTTGGGTGGATTGTTCGGCTTCTTGAT

NSCLC: A549 v HLAMP S11f 45

GGTCGCAGATGGATTAAGTATGTGGGTGGGGGGGTGGAAGTTAAT

NSCLC: A549 v HLAMP S15 45

GCTATCTTATGGAAATTTCGTGTAGGGTTTGGTGTGGCGGGGCTA

11 PigPen III.1 96

AGGCGGTGCATTGTGGTTGGTAGTATACATGAGGTTTGGTTGAGACTAGTCGCA

12 RBC Ghost: CD71 C56t 26

AACTCAGTAATGCCAAGGTAACGGTT

RBC Ghost Motif 2a 33

CGAATCGCATTGCCCAACGTTGCCCAAGATTCG

Page 97: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

97

Table A-1. Continued

SELEX Target Name Nt Sequence

13 Differentiated PC12 1 25

TGGTTGGGGATAGAGGTGGGTGTTT

Differentiated PC12 2 25

TGAGGGTCTAGGGTGGTGGGGTGGA

Differentiated PC12 3 25 TGATGGATGTGGGGATGCGGGGGCG

Differentiated PC12 4 25

TATGGGGGTGGGTCAGGTTTCGGTA

Differentiated PC12 5 24

GGGAGGTTGGGGTATCAGGGGGGG

Differentiated PC12 7 25

GGGTGTGGGAGGTGATGGGGTAGGT

Differentiated PC12 8 24

AGGGGGGTTCGGCGGAGGTATCAG

Differentiated PC12 10 25

GCTGGGGTGTTGGGTGTGGGGGTGA

Differentiated PC12 12 25

GTGCGACATAGCTAAACCGGTTCGT

Differentiated PC12 13 25

GAGGAGGGAGAATAGGGGTGGGTGG

Differentiated PC12 14 24

AGTCAGACAGGGGGGAGGATCCGT

Differentiated PC12 15 25

TGGGTAGGTTCGAGGGGTGGGTGTG

Differentiated PC12 16 25

AGAGTGGGGGGGATGTAGGTGGGTT

Differentiated PC12 17 25

GGTTGGATGTAAGGTTGGAGGGGGG

Differentiated PC12 18 25

GTGTCCGTGGACTAAACCGGCCTGT

Differentiated PC12 20 24

GTGGAAGCCTCCTAAGCGGTGTGT

Differentiated PC12 22 24

TGGGTGAGTTCAATGGGGGTATGT

Differentiated PC12 23 25

GGGTGTGAGAGGTTGAGGGGGTTCG

14 Vaccinia Virus A549 TVO1 25

GTGCATTGAAACTTCTGCATCCTCG

Vaccinia Virus A550 TVO2 24

CCTGCATATACACTTTGCATGTGG

Vaccinia Virus A551 TVO4 33

AACCTGCATAATTTATAAGTCTAGACTGCTGCA

Vaccinia Virus A552 TVO6 27

GGACCGATAGGAACCACGGACTGCATG

15 Vaccinia Virus Hela PP2 38

ACACCGTTTGTATTCTGCATTGTTTTGCATTCTACATG

Vaccinia Virus Hela PP5 31

CACTTGCATATACACTTTGCATTATAGGGTG

Page 98: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

98

Table A-1. Continued

SELEX Target Name Nt Sequence

16 Mucin MUC15TR1 25

GAAGTGAAAATGACAGAACACAACA

Mucin MUC15TR2 25

GGCTATAGCACATGGGTAAAACGAC

Mucin MUC15TR3 25

CAAACAATCAAACAGCAGTGGGGTG

Mucin MUC15TR4 25

TACTGCATGCACACCACTTCAACTA

17 HL60/CEM KH1C12 42

TGCCCTAGTTACTACTACTCTTTTTAGCAAACGCCCTCGCTT

HL60/CEM KHG11 45

TGCTCATCCACGATTCTGGCGAATTTAGTGCCTGTCTTTTTCTCT

HL60 KH2B05 42

CACACAACCTGCTCATAAACTTTACTCTGCTCGAACCATCTC

Ramos KH1A02 44 GGCATAGATGTGCAGCTCCAAGGAGAAGAAGGAGTTCTGTGTAT

Ramos KK1B10 45

GATCAGTCTATCTTCTCCTGATGGGTTCCTATTTATAGGTGAAGC

HL60, NB4, K562/CEM KH1B08 45

TTCAAATCACACGACGCATTGAAACACTCTACAATATCACATTTA

HL60, NB4, K562/CEM KH3H03 45

CTGGCGCCTTCTACTTCAAGGCAATAAGCTCAATCAATATCATCG

18 CEM H01 46

AAGCAGCAGCTGTGCCATCGGGTTCGGATTTTCTTCCTACGACTGC

CEM H04 45

TATCAAAGGCGAATTTTGTCAAGGTGTTAAACGATAGTCCCTACC

CEM, Ramos, Toledo H11 44

TCGCCTGTACATAGACTGTTGCGTTAGGGTCTGCCTTTATCTTG

CEM, Ramos, Toledo B07 44

CATAGAGACTTGGATGCAACTTAGCTACTAACGCTAGCTCTATG

19 Ramos TD05 47

AACACCGTGGAGGATAGTTCGGTGGCTGTTCAGGGTCTCCTCCGGTG

Ramos, CEM, Toledo TD08 85

TACTCTAATTGCCGTATAAGGTCAGGGGGTTGGTTGGTTCCTAGTGCTT

Ramos TE02 44

GCAGTGGTTTGACGTCCGCATGTTGGGAATAGCCACGCCTCGGG

Ramos, CEM TE04 44

CACTCCTCGATGCACCAGTTCACCTTATTTGCTTCTTCTCTCTG

Ramos, CEM, Toledo TE13 42

GCCCCCAGGCTCGGTGGATGCAAACACATGACTATGGGCCCG

Ramos TE17 52

ACCTGCTTGACCGACCGATACAGCTACGCAATACAAAACTCCGAACACCTGC

20 CEM, Ramos, Toledo TC01 33

CCAAACACAGATGCAACCTGACTTCTAACGTCA

CEM TC02 46

AGCATCAACAAGGTCATAAAAACACGTCAGCTCCTTCACATTTGCC

21 CEM, Jurkat sgd3 53

AGGGGGAGCTTGCGCGCATCAAGGTGCTAAACGAAAGCCTCATGGCTTCTATA

Page 99: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

99

Table A-1. Continued

SELEX Target Name Nt Sequence

CEM, Jurkat, Ramos sgc4a 36

CGAGTGCGGATGCAAACGCCAGACAGGGGGACAGGA

CEM, Jurkat sgc5 45

ACCGACGACGAACTATCTATCACTATCTTACACATCATACCTCGA

CEM, Jurkat sgc7 52

ACCGCAGCGACTATCTCGACTACATTACTAGCTTATACTCCGATCATCTCTA

CEM, Jurkat Sgd2 53

GAGTGAAGCAAGGATGCAACCTCGGCTCCAACCCGTGAGAGTCGCGAAACTCA

CEM, Jurkat, Toledo Sgd5a 66

ACTTATTCAATTATCGTGGGTCACAGCAGCGGTTGTGAGGAAGAAAGGCGGATAA

CAGATAATAAG

CEM, Jurkat sgc8c 41

ATCTAACTGCTGCGCCGCCGGGAAAATACTGTACGGTTAGA

CEM, Jurkat sgc3b 51

ACTTATTCAATTCCTGTGGGAAGGCTATAGAGGGGCCAGTCTATGAATAAG

22 Liver Cancer mouse: IMEA TLS1c 55

ACAGGAGTGATGGTTGTTATCTGGCCTCAGAGGTTCTCGGGTGTGGTCACTCCTG

Liver Cancer mouse: IMEA TLS3 45

TGGGAATATTAGTACCGTTATTCGGACTCCGCCATGACAATCTGG

Liver Cancer mouse: IMEA TLS4 45

ACGGTGGTCGTACACGGCCATTTTATTCCCGGAATATTTGTCAAC

Liver Cancer mouse: IMEA TLS7 45

TGCGCCCAAAGTTCCCATATTGCTTCCCTGTTGGTGAGTGCCGAT

Liver Cancer mouse: IMEA TLS11a 63

ACAGCATCCCCATGTGAACAATCGCATTGTGATTGTTACGGTTTCCGCCTCATGG

ACGTGCTG

Small Cell Lung Cancer HCH07 35

GCCGATGTCAACTTTTTCTAACTCACTGGTTTTGC

23 Small Cell Lung Cancer HCA12 35

GTGGATTGTTGTGTTCTGTTGGTTTTTGTGTTGTC

Small Cell Lung Cancer HCC03 35

CCGGGGACCGGGGCACCGGGGGCCAGTGGCACGGA

Small Cell Lung Cancer HCH01 71

GTCAACCGAATGCGTCAGCTGGATCTTAAAGATTGCATGCGCTCACTATGGGACT

GAGCATCGCACTGGTA

24 Colon Cancer KMF2-1a 42

GAATAGGGGATGTACAGGTCTGCACCCACTCGAGGAGTGACT

Colon Cancer KMF3 42

AGGATAGCCATACCACCGGGGAGTTTATAACGGTACGGTCCT

Colon Cancer KMF9b 46

AGCGCAGCAGCTGTGCCACCGGGAGAATTTACGTACGGCTGAGCGA

25 Colon Cancer KDED1 39

CTAAACAAAATACGAGCAGGGAGACTTCTATCCGATTGT

Colon Cancer KDED2 55

AACTGCTATTACGTGTGAGAGGAAAGATCACGCGGGTTCGTGGACACGGTGGCTT

Colon Cancer KDED3 39

GGGGTGGTTTTCAAAGAGTCTTGCCTGACTCCCCTGTGG

Colon Cancer KDED7 40

GCGGACGCACTTTTAGCAAGCAAGTCGACAATGGAGGTTT

Colon Cancer KDED9 40

GCAACTGAAGCTAGAACTGTGTGGGGTTTGGGGTATAATT

Page 100: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

100

Table A-1. Continued

SELEX Target Name Nt Sequence

Colon Cancer KDED20 45

TAGGTTGGATAGGGATGGTAGAGCAGGCTAAGCACTTTTTTTTAT

26 Colon Cancer KCHA10a 59

ACGCAGCAGGGGAGGCGAGAGCGCACAATAACGATGGTTGGGACCCAACTGTTTG

GACA

Colon Cancer KCHB10 63

ATCCAGAGTGACGCAGCAGATCTGTGTAGGATCGCAGTGTAGTTGACATTTGATA

CGACTGGC

27 Colon Cancer KC2D3 37

CGGGAAAGGAACAAACTGCTATTAGGTCGCAGGCCGG

Colon Cancer KC2D4 37

CCCACTGGTAGCCATTCCGCCCTTAACCGGGCCATCG

Colon Cancer KC2D8 38

AACTGCTGCGCCGCCGGGAAAATACTGTACGGTTAGTT

Ovarian TOV Cells aptTOVl 45

GATCTGTGTAGGATCGCAGTGTAGTGGACATTTGATACGACTGGC

Ovarian TOV Cells aptTOV2a 42

CAATCTCTACAGGCGCATGTAATATAATGGAGCCTATCCACG

Ovarian TOV Cells aptTOV3 42

CTCACTCTGACCTTGGATCGTCACATTACATGGGATCATCAG

Ovarian TOV Cells aptTOV4 42

GGCACTCTTCACAACACGACATTTCACTACTCACAATCACTC

Ovarian TOV Cells aptTOV5 42

CAACATCCACTCATAACTTCAATACATATCTGTCACTCTTTC

Ovarian TOV Cells aptTOV6 42

CGGCACTCACTCTTTGTTAAGTGGTCTGCTTCTTAACCTTCA

Ovarian TOV Cells aptTOV7 42

CCAACTCGTACATCCTTCACTTAATCCGTCAATCTACCACTC

Ovarian TOV Cells aptTOV8 42

CCAGTCCATCCCAAAATCTGTCGTCACATACCCTGCTGCGCC

Ovarian TOV Cells aptTOV9 42

GCAACACAAACCCAACTTCTTATCTTTTCGTTCACTCTTCTC

29 Ovarian DOV Cells DOV3 37

ATGCAGAGGCTAGGATCTATAGGTTCGGACGTCGATG

Ovarian DOV Cells DOV6 37

AATGTTGGGGTAGGTAGAAGGTGAAGGGGTTTCAGTT

30 Adenocarcinoma: H23 EJD1 42

CCCTCACCACCAAACAACAATATTAGAGACAATGAGTTCCCT

Adenocarcinoma: H23 EJD2 41

AGTGGTCGAACTACACATCCTTGAACTGCGGAATTATCTAC

Adenocarcinoma: H23 EJD4 41

GAAGACGAGCGGCGAGTGTTATTACGCTTGGAAACAACCCC

Adenocarcinoma: H23 EJD5 41

TACGGGCTGGATCCACTGTTACGGCGTGTATCCGCTATCAA

Adenocarcinoma: H23 EJD7 42

CAACTCTTAAGTAAATACCTTTTTCTGGCGTGTAAGAAAATG

Adenocarcinoma: H23 ADE1 42

GGCAAAGCACGACGACATGGTATTACACGAACTACAATCCCT

Adenocarcinoma: H23 ADE2 42

GAGCCCTATCTCACACCGCACCCGCAAACTATCATCCTACAT

Page 101: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

101

Table A-1. Continued

SELEX Target Name Nt Sequence

31 Cancer Stem Cells: DU145 CSC01 40

AGGTGGTTTGCTGCGGTGGGCTCAAGAAGAAAGCGCAAAG

Cancer Stem Cells: DU146 CSC08 43

GCTCTGAGCCTAGCTTGACCACTTTTCTTTATTCGCTCTGAGG

Cancer Stem Cells: DU147 CSC13 43

GGGGTGTCGTATCTTTCGTGTCTTATTATTTTCTAGGTGGAGG

Cancer Stem Cells: DU148 CSC17 42

CACCAGCTCCATAACGACACGACCCTCATTCCAACACACAGG

Cancer Stem Cells: DU149 CSC22 43

GTGGGGCTGTGATACTTTACATCTTATTTCTCTAGTGACTAGG

32 Activated Protein C HS02-52G 52

GCCTCCTAACTGAGCTGTACTCGACTTATCCCGGATGGGGCTCTTAGGAGGC

33 Mesenchymal Stem Cells 1MSC 40

CGACTTCGGTTATTACGTTGTTGGCCTCACAAGGACGCCC

Mesenchymal Stem Cells 2MSC 39

CACGATCCAGATGTCATAGTTTAGGCTCTCTCTACTACT

Mesenchymal Stem Cells 3MSC 40

GGCGGGAGGTCACGTTGAGAATTTACGAGGCAGGGGGCAC

Mesenchymal Stem Cells 4MSC 39

GAGGGGCCGCCAAAGCTAGCTCAAGTGATATCCTGTACT

Mesenchymal Stem Cells 5MSC 41

CACCCGTATGCCAAGTCAGATCCAGTGTAGATGCGCGCCCC

Mesenchymal Stem Cells 6MSC 41

CGACACGCGCACGGTTCTCATCAATACTGCCTCGCCGGTAC

Mesenchymal Stem Cells 7MSC 38

CAGCATGCAGAGGCGTCAAATAACGGGACCTCTCGGAC

Mesenchymal Stem Cells 8MSC 53

GGGGAGTGGTGGAGAAAGGCTTACAGGGTAGATAAGGTTCAGGTGCTTCGTTC

Mesenchymal Stem Cells 9MSC 50

GGGTCATTGCAGGGTAAGGTTGGATTTATTGATGCCTCGGAGTTGGGTGG

Mesenchymal Stem Cells 10MSC 50

GTAGGCGTTGCCTTAGTTATTGTTTTGAGGTAGAGCAGAGTTTTACTCAG

Mesenchymal Stem Cells 11MSC 50

CGAGGTGGATGACAGGGTATGTGGATTGGTAGTGTGTTTGGTGCTAACGC

Mesenchymal Stem Cells 12MSC 50

GGAGGAAGGGTTACGGAGGAAGAGTTAGGATCGGTGGGGATGATGATGGG

Mesenchymal Stem Cells 13MSC 50

GGTTTAATGTGTGGGTAGTTGGGCGTGACGGGGTAGTCCTGGGGGTTAGG

Mesenchymal Stem Cells 14MSC 50

GTGGAGTGGCCGTAGTCTGGCCAGGTCCCGTTGGTGATGGGTAGAGTGGG

Mesenchymal Stem Cells 15MSC 50

TTTGCGCTGGATGCGATAACGTGTTCGACATGAGGCCCGGATCCACTCCC

Mesenchymal Stem Cells 16MSC 50

TGTGCTTATGCTCGAGATGGTGTTATCCGTGTTGCCACGATGGGGGGACC

Mesenchymal Stem Cells 17MSC 50

TGGATGGGTGGGCGTAGGTGAGGTGTTGTAAGAGCCTCTCCACAGGTGCG

Page 102: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

102

APPENDIX B PREDICTED SECONDARY STRUCTURES OF PTK7 APTAMERS

Figure B-1. Predicted secondary structures for PTK7 aptamers. Green labelled parts depict to the consensus region.

A)H01, B)KC2D8, C)KMF9, D)sgc8

Page 103: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

103

LIST OF REFERENCES

1. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2016. CA. Cancer J. Clin. 66, 7–30 (2016).

2. World Cancer Report 2014. (2014).

3. Greaves, M. & Maley, C. C. Clonal evolution in cancer. Nature 481, 306–313 (2012).

4. DeVita, V. T. & Chu, E. A History of Cancer Chemotherapy. Cancer Res. 68, (2008).

5. Peer, D. et al. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2, 751–760 (2007).

6. Etzioni, R. et al. Early detection: The case for early detection. Nat. Rev. Cancer 3, 243–252 (2003).

7. Siddharta, M. The Emperor of All Maladies. (Schribner, 2010).

8. Gerdes, H.-H. & Pepperkok, R. Cell-to-cell communication: current views and future perspectives. Cell Tissue Res. 352, 1–3 (2013).

9. Kano, A. et al. Tumor cell secretion of soluble factor(s) for specific immunosuppression. Sci. Rep. 5, 8913 (2015).

10. Lu, H. H. & Jiang, J. Interface tissue engineering and the formulation of multiple-tissue systems. Adv. Biochem. Eng. Biotechnol. 102, 91–111 (2006).

11. Bhatia, S. N., Balis, U. J., Yarmush, M. L. & Toner, M. Effect of cell-cell interactions in preservation of cellular phenotype: cocultivation of hepatocytes and nonparenchymal cells. FASEB J. 13, 1883–900 (1999).

12. Gould, S. J. & Raposo, G. As we wait: coping with an imperfect nomenclature for extracellular vesicles. J. Extracell. Vesicles 2, (2013).

13. Iraci, N., Leonardi, T., Gessler, F., Vega, B. & Pluchino, S. Focus on extracellular vesicles: Physiological role and signalling properties of extracellular membrane vesicles. International Journal of Molecular Sciences 17, (2016).

14. Zaborowski, M. P., Balaj, L., Breakefield, X. O. & Lai, C. P. Extracellular Vesicles: Composition, Biological Relevance, and Methods of Study. Biosci. 65, 783–797 (2015).

15. Lo Cicero, A., Stahl, P. D. & Raposo, G. Extracellular vesicles shuffling intercellular messages: for good or for bad. Curr. Opin. Cell Biol. 35, 69–77 (2015).

Page 104: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

104

16. Yáñez-Mó, M. et al. Biological properties of extracellular vesicles and their physiological functions. J. Extracell. Vesicles 4, (2015).

17. An, T. et al. Exosomes serve as tumour markers for personalized diagnostics owing to their important role in cancer metastasis. J. Extracell. Vesicles 4, (2015).

18. Chargaff, E. & West, R. The biological significance of the thromboplastic protein of blood. J. Biol. Chem. 166, 189–97 (1946).

19. Wolf, P. The Nature and Significance of Platelet Products in Human Plasma. Br. J. Haematol. 13, 269–288 (1967).

20. Appelmans, F., Wattiaux, R. & De Duve, C. Tissue fractionation studies. 5. The association of acid phosphatase with a special class of cytoplasmic granules in rat liver. Biochem. J. 59, 438–45 (1955).

21. Pan, B. T., Teng, K., Wu, C., Adam, M. & Johnstone, R. M. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J. Cell Biol. 101, 942–8 (1985).

22. De Broe, M., Wieme, R. & Roels, F. Letter: Membrane fragments with koinozymic properties released from villous adenoma of the rectum. Lancet (London, England) 2, 1214–5 (1975).

23. Benz, E. W. & Moses, H. L. Small, virus-like particles detected in bovine sera by electron microscopy. J. Natl. Cancer Inst. 52, 1931–4 (1974).

24. Stegmayr, B. & Ronquist, G. Promotive effect on human sperm progressive motility by prostasomes. Urol. Res. 10, 253–7 (1982).

25. Harding, C. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J. Cell Biol. 97, 329–339 (1983).

26. Pan, B.-T. & Johnstone, R. M. Fate of the transferrin receptor during maturation of sheep reticulocytes in vitro: Selective externalization of the receptor. Cell 33, 967–978 (1983).

27. Johnstone, R. M., Adam, M., Hammond, J. R., Orr, L. & Turbide, C. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J. Biol. Chem. 262, 9412–20 (1987).

28. Raposo, G. et al. B lymphocytes secrete antigen-presenting vesicles. J. Exp. Med. 183, 1161–72 (1996).

29. Valadi, H. et al. Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat. Cell Biol. 9, 654–659 (2007).

Page 105: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

105

30. Ratajczak, J. et al. Embryonic stem cell-derived microvesicles reprogram hematopoietic progenitors: evidence for horizontal transfer of mRNA and protein delivery. Leukemia 20, 847–56 (2006).

31. Lässer, C. et al. RNA-containing exosomes in human nasal secretions. Am. J. Rhinol. Allergy 25, 89–93

32. Keller, S., Ridinger, J., Rupp, A.-K., Janssen, J. W. G. & Altevogt, P. Body fluid derived exosomes as a novel template for clinical diagnostics. J. Transl. Med. 9, 86 (2011).

33. Poliakov, A., Spilman, M., Dokland, T., Amling, C. L. & Mobley, J. A. Structural heterogeneity and protein composition of exosome-like vesicles (prostasomes) in human semen. Prostate 69, 159–67 (2009).

34. Caby, M.-P., Lankar, D., Vincendeau-Scherrer, C., Raposo, G. & Bonnerot, C. Exosomal-like vesicles are present in human blood plasma. Int. Immunol. 17, 879–87 (2005).

35. Lässer, C., Eldh, M. & Lötvall, J. Isolation and characterization of RNA-containing exosomes. J. Vis. Exp. e3037 (2012). doi:10.3791/3037

36. Denzer, K. et al. Exosome: from internal vesicle of the multivesicular body to intercellular signaling device. J. Cell Sci. 113 Pt 19, 3365–74 (2000).

37. Rak, J. Extracellular vesicles - biomarkers and effectors of the cellular interactome in cancer. Front. Pharmacol. 4, 21 (2013).

38. Crescitelli, R. et al. Distinct RNA profiles in subpopulations of extracellular vesicles: apoptotic bodies, microvesicles and exosomes. J. Extracell. Vesicles 2, (2013).

39. D’Souza-Schorey, C. & Clancy, J. W. Tumor-derived microvesicles: shedding light on novel microenvironment modulators and prospective cancer biomarkers. Genes Dev. 26, 1287–99 (2012).

40. Mathivanan, S. et al. Proteomics analysis of A33 immunoaffinity-purified exosomes released from the human colon tumor cell line LIM1215 reveals a tissue-specific protein signature. Mol. Cell. Proteomics 9, 197–208 (2010).

41. Stoorvogel, W., Kleijmeer, M. J., Geuze, H. J. & Raposo, G. The Biogenesis and Functions of Exosomes. Traffic 3, 321–330 (2002).

42. Théry, C., Zitvogel, L. & Amigorena, S. Exosomes: composition, biogenesis and function. Nat. Rev. Immunol. 2, 569–79 (2002).

Page 106: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

106

43. Colombo, M., Raposo, G. & Théry, C. Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu. Rev. Cell Dev. Biol. 30, 255–89 (2014).

44. van Niel, G., Porto-Carreiro, I., Simoes, S. & Raposo, G. Exosomes: a common pathway for a specialized function. J. Biochem. 140, 13–21 (2006).

45. Nieuwenhuysen, P. & Clauwaert, J. Physical properties of Artemia salina ribosomes. Biochemistry 17, 4260–5 (1978).

46. Conde-Vancells, J. et al. Characterization and comprehensive proteome profiling of exosomes secreted by hepatocytes. J. Proteome Res. 7, 5157–66 (2008).

47. Logozzi, M. et al. High levels of exosomes expressing CD63 and caveolin-1 in plasma of melanoma patients. PLoS One 4, e5219 (2009).

48. Bobrie, A., Colombo, M., Raposo, G. & Théry, C. Exosome Secretion: Molecular Mechanisms and Roles in Immune Responses. Traffic 12, 1659–1668 (2011).

49. Vlassov, A. V., Magdaleno, S., Setterquist, R. & Conrad, R. Exosomes: Current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim. Biophys. Acta - Gen. Subj. 1820, 940–948 (2012).

50. Lee, Y., El Andaloussi, S. & Wood, M. J. A. Exosomes and microvesicles: extracellular vesicles for genetic information transfer and gene therapy. Hum. Mol. Genet. 21, R125-34 (2012).

51. Subra, C. et al. Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins. J. Lipid Res. 51, 2105–20 (2010).

52. Mathivanan, S. & Simpson, R. J. ExoCarta: A compendium of exosomal proteins and RNA. Proteomics 9, 4997–5000 (2009).

53. Akers, J. C., Gonda, D., Kim, R., Carter, B. S. & Chen, C. C. Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. doi:10.1007/s11060-013-1084-8

54. EL Andaloussi, S., Mager, I., Breakefield, X. O. & Wood, M. J. A. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov 12, 347–357 (2013).

55. Mayer, G. The chemical biology of aptamers. Angew. Chem. Int. Ed. Engl. 48, 2672–89 (2009).

56. Ellington, A. D. & Szostak, J. W. In vitro selection of RNA molecules that bind specific ligands. Nature 346, 818–822 (1990).

Page 107: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

107

57. Tuerk, C. & Gold, L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science 249, 505–10 (1990).

58. Robertson, D. L. & Joyce, G. F. Selection in vitro of an RNA enzyme that specifically cleaves single-stranded DNA. Nature 344, 467–468 (1990).

59. Brody, E. N. & Gold, L. Aptamers as therapeutic and diagnostic agents. Rev. Mol. Biotechnol. 74, 5–13 (2000).

60. Sefah, K., Shangguan, D., Xiong, X., O’Donoghue, M. B. & Tan, W. Development of DNA aptamers using Cell-SELEX. Nat. Protoc. 5, 1169–85 (2010).

61. Metzker, M. L. Sequencing technologies — the next generation. Nat. Rev. Genet. 11, 31–46 (2010).

62. Zhou, Q. et al. Immune-related MicroRNAs are Abundant in Breast Milk Exosomes. Int. J. Biol. Sci. 8, 118–123 (2012).

63. Simpson, R. J., Jensen, S. S. & Lim, J. W. E. Proteomic profiling of exosomes: Current perspectives. Proteomics 8, 4083–4099 (2008).

64. Vella, L. J., Hill, A. F. & Cheng, L. Focus on Extracellular Vesicles: Exosomes and Their Role in Protein Trafficking and Biomarker Potential in Alzheimer’s and Parkinson’s Disease. Int. J. Mol. Sci. 17, 173 (2016).

65. Rajendran, L. et al. Alzheimer’s disease beta-amyloid peptides are released in association with exosomes. Proc. Natl. Acad. Sci. U. S. A. 103, 11172–7 (2006).

66. Fevrier, B. et al. Cells release prions in association with exosomes. Proc. Natl. Acad. Sci. U. S. A. 101, 9683–8 (2004).

67. Lobb, R. J. et al. Optimized exosome isolation protocol for cell culture supernatant and human plasma. J. Extracell. Vesicles 4, (2015).

68. Théry, C. et al. in Current Protocols in Cell Biology 3.22.1-3.22.29 (John Wiley & Sons, Inc., 2006). doi:10.1002/0471143030.cb0322s30

69. Livshts, M. A. et al. Isolation of exosomes by differential centrifugation: Theoretical analysis of a commonly used protocol. Sci. Rep. 5, 17319 (2015).

70. Lamparski, H. G. et al. Production and characterization of clinical grade exosomes derived from dendritic cells. J. Immunol. Methods 270, 211–26 (2002).

71. Cocucci, E., Racchetti, G. & Meldolesi, J. Shedding microvesicles: artefacts no more. doi:10.1016/j.tcb.2008.11.003

72. Wu, Y. et al. Exosomes: improved methods to characterize their morphology, RNA content, and surface protein biomarkers. Analyst 140, 6631–6642 (2015).

Page 108: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

108

73. Dragovic, R. A. et al. Sizing and phenotyping of cellular vesicles using Nanoparticle Tracking Analysis. Nanomedicine Nanotechnology, Biol. Med. 7, 780–788 (2011).

74. Filipe, V., Hawe, A. & Jiskoot, W. Critical Evaluation of Nanoparticle Tracking Analysis (NTA) by NanoSight for the Measurement of Nanoparticles and Protein Aggregates. Pharm. Res. 27, 796–810 (2010).

75. György, B. et al. Membrane vesicles, current state-of-the-art: emerging role of extracellular vesicles. Cell. Mol. Life Sci. 68, 2667–2688 (2011).

76. Zhang, L. et al. Evolution of Functional Six-Nucleotide DNA. J. Am. Chem. Soc. (2015).

77. Théry, C., Ostrowski, M. & Segura, E. Membrane vesicles as conveyors of immune responses. Nat. Rev. Immunol. 9, 581–593 (2009).

78. Vader, P. et al. Extracellular vesicles: emerging targets for cancer therapy. Trends Mol. Med. 20, 385–93 (2014).

79. Kim, H. K. et al. Elevated levels of circulating platelet microparticles, VEGF, IL-6 and RANTES in patients with gastric cancer: possible role of a metastasis predictor. Eur. J. Cancer 39, 184–91 (2003).

80. Mehlen, P. & Puisieux, A. Metastasis: a question of life or death. Nat. Rev. Cancer 6, 449–458 (2006).

81. Hoshino, A. et al. Tumour exosome integrins determine organotropic metastasis. Nature 527, 329–335 (2015).

82. Hood, J. L., San, R. S. & Wickline, S. A. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 71, 3792–801 (2011).

83. Simpson, R. J., Lim, J. W., Moritz, R. L. & Mathivanan, S. Exosomes: proteomic insights and diagnostic potential. http://dx.doi.org/10.1586/epr.09.17 (2014).

84. Raimondo, F., Morosi, L., Chinello, C., Magni, F. & Pitto, M. Advances in membranous vesicle and exosome proteomics improving biological understanding and biomarker discovery. Proteomics 11, 709–20 (2011).

85. Van Simaeys, D. et al. Study of the molecular recognition of aptamers selected through ovarian cancer cell-SELEX. PLoS One 5, e13770 (2010).

86. Sefah, K. et al. DNA aptamers as molecular probes for colorectal cancer study. PLoS One 5, e14269 (2010).

Page 109: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

109

87. Zhao, Z. et al. Recognition of subtype non-small cell lung cancer by DNA aptamers selected from living cells. Analyst 134, 1808–14 (2009).

88. Zhang, K. et al. A novel aptamer developed for breast cancer cell internalization. ChemMedChem 7, 79–84 (2012).

89. Jiménez, E. et al. Generation of lung adenocarcinoma DNA aptamers for cancer studies. PLoS One 7, e46222 (2012).

90. Xu, J. et al. Molecular Recognition of Human Liver Cancer Cells Using DNA Aptamers Generated via Cell-SELEX. (2015).

91. Sefah, K. et al. In vitro selection with artificial expanded genetic information systems. Proc. Natl. Acad. Sci. U. S. A. 111, 1449–54 (2014).

92. Duffy, M. J. Carcinoembryonic Antigen as a Marker for Colorectal Cancer: Is It Clinically Useful? Clin. Chem. 47, (2001).

93. Stenman, U.-H., Leinonen, J., Zhang, W.-M. & Finn, P. Prostate - specific antigen. CANCER BI OLOGY 9, (1999).

94. Taylor, D. D., Gercel-Taylor, C. & Parker, L. P. Patient-derived tumor-reactive antibodies as diagnostic markers for ovarian cancer. Gynecol. Oncol. 115, 112–120 (2009).

95. Nilsson, J. et al. Prostate cancer-derived urine exosomes: a novel approach to biomarkers for prostate cancer. Br. J. Cancer 100, 1603–1607 (2009).

96. Cho, W. C. S. Contribution of oncoproteomics to cancer biomarker discovery. Mol. Cancer 6, 25 (2007).

97. Wurster, S. E. & Maher, L. J. Selection and characterization of anti-NF-kappaB p65 RNA aptamers. RNA 14, 1037–47 (2008).

98. Liu, M. et al. In vitro selection of a photo-responsive peptide aptamer using ribosome display. Chem. Commun. 48, 11871 (2012).

99. Bayrac, A. T. et al. In vitro Selection of DNA Aptamers to Glioblastoma Multiforme. ACS Chem. Neurosci. 2, 175–181 (2011).

100. Kalra, H. et al. Comparative proteomics evaluation of plasma exosome isolation techniques and assessment of the stability of exosomes in normal human blood plasma. Proteomics 13, 3354–3364 (2013).

101. Shangguan, D., Bing, T. & Zhang, N. in Aptamers Selected by Cell-SELEX for Theranostics 13–33 (Springer Berlin Heidelberg, 2015). doi:10.1007/978-3-662-46226-3_2

Page 110: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

110

102. Lu, X. et al. PTK7/CCK-4 is a novel regulator of planar cell polarity in vertebrates. Nature 430, 93–8 (2004).

103. Mossie, K. et al. Colon carcinoma kinase-4 defines a new subclass of the receptor tyrosine kinase family. Oncogene 11, 2179–84 (1995).

104. Shnitsar, I. & Borchers, A. PTK7 recruits dsh to regulate neural crest migration. Development 135, 4015–24 (2008).

105. Yen, W. W. et al. PTK7 is essential for polarized cell motility and convergent extension during mouse gastrulation. Development 136, 2039–48 (2009).

106. Katoh, M. & Katoh, M. Comparative integromics on non-canonical WNT or planar cell polarity signaling molecules: Transcriptional mechanism of PTK7 in colorectal cancer and that of SEMA6A in undifferentiated ES cells. Int. J. Mol. Med. 20, 405–409 (2007).

107. Prebet, T. et al. The cell polarity PTK7 receptor acts as a modulator of the chemotherapeutic response in acute myeloid leukemia and impairs clinical outcome. Blood 116, 2315–23 (2010).

108. Easty, D. J. et al. Loss of expression of receptor tyrosine kinase family genes PTK7 and SEK in metastatic melanoma. Int. J. Cancer 71, 1061–5 (1997).

109. Miller, M. A. & Steele, R. E. Lemon encodes an unusual receptor protein-tyrosine kinase expressed during gametogenesis in Hydra. Dev. Biol. 224, 286–98 (2000).

110. Golubkov, V. S. et al. The Wnt/planar cell polarity protein-tyrosine kinase-7 (PTK7) is a highly efficient proteolytic target of membrane type-1 matrix metalloproteinase: implications in cancer and embryogenesis. J. Biol. Chem. 285, 35740–9 (2010).

111. Kanazawa, A. et al. Relation of MT1-MMP gene expression to outcomes in colorectal cancer. J. Surg. Oncol. 102, 571–5 (2010).

112. Cavalheiro, B. G., Junqueira, C. R. & Brandão, L. G. Expression of membrane type 1 matrix metalloproteinase in medullary thyroid carcinoma: prognostic implications. Head Neck 32, 58–67 (2010).

113. Paudyal, A. et al. The novel mouse mutant, chuzhoi, has disruption of Ptk7 protein and exhibits defects in neural tube, heart and lung development and abnormal planar cell polarity in the ear. BMC Dev. Biol. 10, 87 (2010).

114. Gao, B. in Current Topics in Developmental Biology 101, 263–295 (2012).

115. Reya, T. & Clevers, H. Wnt signalling in stem cells and cancer. Nature 434, 843–850 (2005).

Page 111: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

111

116. Niehrs, C. The complex world of WNT receptor signalling. Nat. Rev. Mol. Cell Biol. 13, 767–779 (2012).

117. Schlessinger, K., Hall, A. & Tolwinski, N. Wnt signaling pathways meet Rho GTPases. Genes Dev. 23, 265–77 (2009).

118. Lu, X. et al. PTK7/CCK-4 is a novel regulator of planar cell polarity in vertebrates. Nature 430, 93–98 (2004).

119. Peradziryi, H. et al. PTK7/Otk interacts with Wnts and inhibits canonical Wnt signalling. EMBO J. 30, 3729–40 (2011).

120. Shangguan, D. et al. Aptamers evolved from live cells as effective molecular probes for cancer study. Proc. Natl. Acad. Sci. U. S. A. 103, 11838–43 (2006).

121. Chen, H. W. et al. Molecular recognition of small-cell lung cancer cells using aptamers. ChemMedChem 3, 991–1001 (2008).

122. Wallin, E. & von Heijne, G. Genome-wide analysis of integral membrane proteins from eubacterial, archaean, and eukaryotic organisms. Protein Sci. 7, 1029–38 (1998).

123. Shangguan, D. et al. Cell-specific aptamer probes for membrane protein elucidation in cancer cells. J. Proteome Res. 7, 2133–9 (2008).

124. Tian, X. et al. PTK7 overexpression in colorectal tumors: Clinicopathological correlation and prognosis relevance. Oncol. Rep. 36, 1829–1836 (2016).

125. Park, S. K., Lee, H. S. & Lee, S. T. Characterization of the human full-length PTK7 cDNA encoding a receptor protein tyrosine kinase-like molecule closely related to chick KLG. J. Biochem. 119, 235–9 (1996).

126. Mü Ller-Tidow, C. et al. High-Throughput Analysis of Genome-Wide Receptor Tyrosine Kinase Expression in Human Cancers Identifies Potential Novel Drug Targets.

127. Liu, Q. et al. PTK7 regulates Id1 expression in CD44-high glioma cells. Neuro. Oncol. 17, 505–515 (2015).

128. Zhang, H. et al. Protein tyrosine kinase 7 (PTK7) as a predictor of lymph node metastases and a novel prognostic biomarker in patients with prostate cancer. Int. J. Mol. Sci. 15, 11665–77 (2014).

129. Wang, H. et al. PTK7 protein is decreased in epithelial ovarian carcinomas with poor prognosis. Int. J. Clin. Exp. Pathol. 7, 7881–9 (2014).

130. Hishida, M. et al. Protein tyrosine kinase 7: a hepatocellular carcinoma-related gene detected by triple-combination array. J. Surg. Res. 195, 444–453 (2015).

Page 112: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

112

131. Wang, L. et al. Wnt signaling stabilizes the DIXDC1 protein through decreased ubiquitin-dependent degradation. Cancer Sci. 101, 700–6 (2010).

132. Tan, C. et al. DIXDC1 activates the Wnt signaling pathway and promotes gastric cancer cell invasion and metastasis. Mol. Carcinog. 55, 397–408 (2016).

133. Larkin, M. A. et al. Clustal W and Clustal X version 2.0. Bioinformatics 23, 2947–2948 (2007).

134. Sun, Y. et al. ESPRIT: estimating species richness using large collections of 16S rRNA pyrosequences. Nucleic Acids Res. 37, e76 (2009).

135. Soma, K., Shiomi, K., Keino-Masu, K. & Masu, M. Expression of mouse Coiled-coil-DIX1 (Ccd1), a positive regulator of Wnt signaling, during embryonic development. Gene Expr. Patterns 6, 325–30 (2006).

136. Wang, X. et al. DIXDC1 isoform, l-DIXDC1, is a novel filamentous actin-binding protein. Biochem. Biophys. Res. Commun. 347, 22–30 (2006).

137. Simons, M. & Mlodzik, M. Planar Cell Polarity Signaling: From Fly Development to Human Disease. Annu. Rev. Genet. 42, 517–540 (2008).

138. Hu, Y., Galkin, A. V., Wu, C., Reddy, V. & Su, A. I. CAFET Algorithm Reveals Wnt/PCP Signature in Lung Squamous Cell Carcinoma. PLoS One 6, e25807 (2011).

139. Seo, Y.-J., Chen, S., Nilsen-Hamilton, M. & Levine, H. A. A Mathematical Analysis of Multiple-Target Selex. Bull. Math. Biol. 72, 1623–1665 (2010).

140. Altschul, S. et al. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25, 3389–3402 (1997).

141. O’Donoghue, M. B. DNA aptamers have an analog in genomic DNA. (University of Florida, 2011).

Page 113: © 2016 Sena Cansizufdcimages.uflib.ufl.edu/UF/E0/05/06/38/00001/CANSIZ_S.pdf · Sena Cansiz December 2016 Chair: Weihong Tan Major: Chemistry Extracellular vesicles (EVs), which

113

BIOGRAPHICAL SKETCH

Sena Cansiz was born in Ankara, Turkey. She earned her Bachelor of Science

Degree in biology at Middle East Technical University in 2008. She did her master’s in

biotechnology in the same school before starting her PhD studies in chemistry at the

University of Florida (2011) under the direction and guidance of Dr. Weihong Tan. Her

graduate work focused on the development of exosome aptamers and aptamer

protein interactions. She received her PhD in chemistry in 2016.


Recommended