정상 프리온 단백질 조절을 통한 대장암 세포에서 항암제의 치료효과 향상

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 185

저작자표시-비영리-변경금지 2.

0 대한민국

이용자는 아래의 조건을 따르는 경우에 한하여 자유롭게

l 이 저작물을 복제, 배포, 전송, 전시, 공연 및 방송할 수 있습니다.

다음과 같은 조건을 따라야 합니다:

저작자표시. 귀하는 원저작자를 표시하여야 합니다.

비영리. 귀하는 이 저작물을 영리 목적으로 이용할 수 없습니다.

변경금지. 귀하는 이 저작물을 개작, 변형 또는 가공할 수 없습니다.

l 귀하는, 이 저작물의 재이용이나 배포의 경우, 이 저작물에 적용된 이용허락조건


을 명확하게 나타내어야 합니다.
l 저작권자로부터 별도의 허가를 받으면 이러한 조건들은 적용되지 않습니다.

저작권법에 따른 이용자의 권리는 위의 내용에 의하여 영향을 받지 않습니다.

이것은 이용허락규약(Legal Code)을 이해하기 쉽게 요약한 것입니다.

Disclaimer
박사학위 논문

정상 프리온 단백질 조절을 통한


대장암 세포에서 항암제의 치료효과
향상

The Enhancement of Therapeutic Effects of Anti-cancer Reagents


in Colorectal Cancer via Regulation of Cellular Prion Protein

2022년 02월

순천향대학교 대학원

의 생 명 과 학 과

윤 철 원
정상 프리온 단백질 조절을 통한
대장암 세포에서 항암제의 치료효과
향상
The Enhancement of Therapeutic Effects of Anti-cancer Reagents
in Colorectal Cancer via Regulation of Cellular Prion Protein

지도교수 이 상 훈

이 논문을 박사학위 논문으로 제출함

2021년 12월

순천향대학교 대학원

의 생 명 과 학 과

윤 철 원
윤철원의 박사 학위논문을 인준함

2021년 12월

위원장 순천향대학교 이 미 영 (인)

위 원 순천향대학교 송 윤 섭 (인)

위 원 순천향대학교 이 상 훈 (인)

위 원 단국대학교 이 준 희 (인)

위 원 충북대학교 이 현 직 (인)

순천향대학교 대학원
CONTENTS

CONTENTS ······················· Ⅰ

LIST OF TABLES ····················IX

LIST OF FIGURES ···················· X

LIST OF ABBREVIATIONS ··············· XIV

ABSTRACT ······················ XIX

BACKGROUND······················ 1

1. Colorectal Cancer ······················· 1

2. Cellular prion protein in colorectal cancer············ 4

3. The anti-cancer drugs for cancer therapy ··········· 6

4. Extracellular vesicles ····················· 8

5. The relationship between hypoxia and extracellular vesicles in cancer

······························· 10

6. Experimental purpose ····················· 13

I
PART I. The Role of Cellular Prion Protein in Colorectal Cancer

······························· 15

Cellular Prion Protein Enhances Drug Resistance of Colorectal Cancer

Cells via Regulation of a Survival Signal Pathway ······· 15

1. INTRODUCTION ······················· 16

2. MATERIALS AND METHODS ················· 18

2.1. Specimens of patients with colorectal cancer and normal controls 18

2.2. Cell and spheroid culture of human colon cancer cell line and S707 cancer

stem cells ··························· 18

2.3. Preparation of 5FU ······················· 19

2.4. Cell isolation targeting PrPC using magnetic activated cell sorting 20

2.5. RNA Sequencing assay for total RNA of sorted colon cancer cells 20

2.6. Western blot analysis ······················ 21

2.7. Flow cytometry analysis ···················· 21

2.8. Immunofluorescence staining ·················· 22

2.9. siRNA transfection ······················· 23

II
2.10. Kinase assays ························· 23

2.11. Cell cycle analysis ······················· 24

2.12. PI/Annexin V flow cytometric analysis ·············· 25

2.13. Statistical analyses ······················· 25

3. RESULTS··························· 26

3.1. The clinicopathological features in patients with CRC by the expression

of PrPC ···························· 26

3.2. PrPC involved in the CSC properties of CRC ··········· 30

3.3. The change of cancer-related genes in CRC through regulation of

PrPC······························ 32

3.4. Expression of PrPC in drug-resistant CRC cells·········· 34

3.5. PrPC regulates CRC cell survival via the PI3K-AKT axis ····· 36

3.6. PrPC is involved in cancer cell proliferation through the regulation of cell

cycle-associated proteins ··················· 38

3.7. PrPC regulates anti-cancer drug induced stress-associated signaling41

3.8. PrPC inhibits apoptosis of anti-cancer drug resistant CRC cells via

suppression of caspase3 activation and PARP1 cleavage······ 43

III
4. DISCUSSION ························· 45

PART Ⅱ. The Application of Anti-cancer Reagents for Cancer

Therapy ························ 48

A. Melatonin Promotes Apoptosis of Colorectal Cancer Cells via

Superoxide-mediated ER Stress by Inhibiting Cellular Prion

Protein Expression ···················· 48

1. INTRODUCTION ······················· 49

2. MATERIALS AND METHODS ················· 52

2.1. Preparation of melatonin ···················· 52

2.2. Cells and cell culture ······················ 52

2.3. Cell viability assay ······················· 52

2.4. Western blot analysis ······················ 53

2.5. siRNA transfection ······················· 54

2.6. Flow cytometry analysis ···················· 54

2.7. Statistical analyses ······················· 55

IV
3. RESULTS··························· 56

3.1. Melatonin decreases cell viability and increases production of superoxide

in SNU-C5/WT cells ······················ 56

3.2. Melatonin suppresses the expression of PrPC and PINK1 and increases

the production of mitochondrial superoxide in SNU-C5/WT cells 58

3.3. Melatonin induces ER stress in SNU-C5/WT cells through regulation of

PrPC expression ························ 61

3.4. Inhibition of PrPC expression enhances melatonin-mediated effect of

apoptosis in SNU-C5/WT cells. ················· 63

4. DISCUSSION ························· 65

B. Prion Protein of Extracellular Vesicle Regulates the Progression

of Colorectal Cancer ··················· 68

1. INTRODUCTION ······················· 69

2. MATERIALS AND METHODS ················· 71

V
2.1. Cell and spheroid culture of human colon cancer cell line and S707 cancer

stem cells ··························· 71

2.2. Cell culture and characterization of endothelial progenitor cells ·· 72

2.3. Preparation of 5FU and oxaliplatin ················ 72

2.4. Flow cytometry analysis ···················· 72

2.5. Isolation of exosomes······················ 73

2.6. Identification of exosomes via cryo-electron microscopy ····· 74

2.7. Dynamic light scattering analysis ················ 74

2.8. Detection of PrPC concentration via enzyme-linked immunosorbent

assay ····························· 74

2.9. Invasion assay ························· 75

2.10. Wound-healing migration assay ················· 75

2.11. Morpholometric analysis ···················· 75

2.12. Human angiogenesis protein array ················ 76

2.13. siRNA transfection ······················· 76

2.14. Western blot analysis ······················ 77

2.15. BrdU incorporation assay ···················· 77

2.16. Measurements of oxygen consumption rate············ 78

2.17. Tumorigenesis in CRC xenograft mice models ·········· 78

VI
2.18. In vitro vascular permeability assay ··············· 79

2.19. In vivo vascular permeability assay ··············· 79

2.20. Immunofluorescence staining ·················· 80

2.21. Ethics statement ························ 80

2.22. Statistical analysis ······················· 81

3. RESULTS··························· 82

3.1. Hypoxia-induced exosomes isolated from drug-resistant CRC ·· 82

3.2. Hypoxia-induced exosomes isolated from drug-resistant CRC

increasing sphere formation via upregulation of PrPC ······· 85

3.3. Hypoxia-induced exosomes isolated from drug-resistant CRC

increasing invasion, migration, and proliferation via upregulation of

PrPC······························ 88

3.4. The characterization of HUVECs and uptake of Hypoxia-stimulated CRC

exosomes ··························· 94

3.5. Hypoxia-stimulated CRC exosomes promoting tumor angiogenesis and

vascular permeability ······················ 97

3.6. Administration of 5FU, anti-PrP antibody, cetuximab inhibiting wild type

CRC progression through suppression of PrPC level ······· 106

3.7. Co-administration of 5FU and anti-PrP antibody inhibiting exosome

VII
treated CRC progression through suppression of PrPC level ··· 109

4. DISCUSSION ························ 122

CONCLUSION ····················· 128

REFERENCES ····················· 133

ABSTRACT IN KOREAN ················ 154

ACKNOWLEDGEMENTS ················ 157

VIII
LIST OF TABLES

Table 1. Clinicopathological features in patients with colorectal

cancer ································· 27

Table 2. Cox regression analysis of the clinicopathological

parameters in colorectal cancer patients ············· 28

IX
LIST OF FIGURES

Figure 1. Identification of PrP C expression in samples of CRC

patients ································ 29

Figure 2. Effect of PrPC of cancer stemness in CRC cells ···· 31

Figure 3. Gene analysis of CRC cells by the PrPC expression ··· 33

Figure 4. The levels of PrPC in SNU-C5/WT, SNU-C5/5FUR, SNU-

C5/OXR and CSC cells ························ 35

Figure 5. The effect of PrP C on cell survival-associated signaling

pathways in CRC cells ························ 37

Figure 6. The effect of PrPC on proliferation in CRC cells ···· 39

Figure 7. The effect of PrPC on cell stress-associated protein in

CRC cells ······························· 42

Figure 8. The effect of PrPC on apoptosis in CRC cells through inhibition of

caspase3 activation and PARP1 cleavage ··············· 44

Figure 9. Melatonin-mediated inhibition of cell viability and

induction of mitochondrial superoxide production in SNU-C5/WT

cells ·································· 57

X
Figure 10. Melatonin mediated inhibition of PrPC and PINK1 in SNU-

C5/WT cells ····························· 59

Figure 11. The effect of PrPC on mitochondrial superoxide production in

SNU-C5/WT cells via treatment with melatonin ··········· 60

Figure 12. The effect of PrPC on melatonin-induced ER stress in

SNU-C5/WT cells ·························· 62

Figure 13. The effect of PrPC on melatonin-enhanced ER stress-

mediated apoptosis in SNU-C5/WT cells ············· 64

Figure 14. Characterization of exosomes secreted by normoxic and hypoxic

drug-resistant CRC cells ······················· 83

Figure 15. Sphere formation of SNU-C5/5FUR, SNU-C5/OXR and

CSC after treatment with exosomes ················ 86

Figure 16. Effect of exosomes secreted by hypoxic drug-resistant

CRC cells on invasion ························ 89

Figure 17. Effect of exosomes secreted by hypoxic drug-resistant

CRC cells on migration ······················· 90

Figure 18. Proliferation capacity of drug-resistant CRC cells after

treatment with exosomes ······················· 91

XI
Figure 19. Effect of exosomes secreted by hypoxic drug-resistant

CRC cells on morphological change ················· 93

Figure 20. Characterization of HUVECs and expression of PrPC after

treatment with exosomes ······················ 95

Figure 21. Uptake of exosomes into HUVECs ············ 96

Figure 22. Effect of exosomes secreted by hypoxic drug-resistant

CRC cells on the motility of HUVECs ··············· 99

Figure 23. Effect of exosomes secreted by hypoxic drug-resistant

CRC cells on angiogenesis ····················· 101

Figure 24. Effect of exosomes secreted by hypoxic drug-resistant

CRC cells on vascular permeability ················· 103

Figure 25. Effect of exosomes secreted by hypoxic drug-resistant CRC

cells on blood vessels ························· 105

Figure 26. Effect of anti-PrP antibody in a wild type CRC xenograft

model ································· 107

Figure 27. Effect of co-administration of anti-PrP antibody and

5FU in a murine xenograft model treated with exosomes secreted

by hypoxic CRC cells ························ 111

XII
Figure 28. Inhibitory effect of anti-PrP antibody with 5FU on proliferation

of CRC cells ······························ 113

Figure 29. Inhibitory effect of anti-PrP antibody with 5FU and

cetuximab on proliferation of CRC cells ·············· 115

Figure 30. Mitochondrial respiration in CRC cells after treatment

with anti-PrP antibody and cetuximab ·············· 117

Figure 31. Effect of co-administration of anti-PrP antibody and

5FU in a murine xenograft model treated with exosomes secreted

by hypoxic CRC cells on proliferation and apoptosis ······ 118

Figure 32. Effect of anti-PrP antibody and 5FU on vascular permeability

in vivo ································· 120

Figure 33. Schematic illustrating the protective mechanism of PrPC on

5FU in 5FU-resistant cancer cells ················· 130

Figure 34. Schematic illustrating the mechanism of melatonin-

mediated anti-cancer effects in SNU-C5/WT cells ······· 131

Figure 35. Schematic illustrating the effects of exosomes secreted by

hypoxic drug-resistant CRC cells on CRC progression and metastasis

···································· 132

XIII
LIST OF ABBREVIATIONS

AKT Protein kinase B

ATF4 Activating transcription factor 4

α-SMA alpha-smooth muscle actin

ATM ATM serine/threonine kinase

ADC Antibody-drug conjugates

BAX Bcl-2-associated X protein

BCL2 B-cell lymphoma 2

BCA Bicinchoninic acid

BrdU 5-bromo-2′-deoxyuridine

CSC Cancer stem cell

CRC Colorectal cancer


CCAAT-enhancerbinding protein
CHOP
homologous protein
CFSE Carboxyfluorescein succinimidyl ester

Cryo-EM Cryo-electron microscopy

CDK Cyclin dependent kinase

DAPI 4’,6-diamidino-2-phenylindole

XIV
Tetramethylindocarbocyanine
DiI
Perchlorate
Dulbecco's Modified Eagle
DMEM/F12
Medium/Nutrient Mixture F-12
Extracellular signal‑regulated protein
ERK1/2
kinase
EBM-2 Endothelial cell growth basal medium-2

EGF Epidermal growth factor

EGFR Epidermal growth factor receptor

EGM-2 Endothelial cell growth medium-2

ELISA Enzyme-linked immunosorbent assay

ENA-78 C-X-C motif chemokine 5

ET-1 Endothelin 1

eIF2α Eukaryotic initiation factor 2-alpha

ER Endoplasmic reticulum

EVs Extracellular vesicles

FBS Fetal bovine serum

FGF Fibroblast growth factor

FFPE Formalin fixed paraffin embedded


Proto-oncogene tyrosine-protein
Fyn
kinase
FITC Fluorescein isothiocyante

XV
Exosomes from SNU-C5/5FUR under
H-5FUR-Exo
hypoxia condition
Exosomes from SNU-C5/OXR under
H-OXR-Exo
hypoxia condition
HIF-1α Hypoxia-inducible factor 1 alpha

HSPA1L Heat shock protein 70 member 1-like

HUVEC Human umbilical vein endothelial cell


Human epidermal growth factor receptor
HER2
2
H&E Hematoxylin and eosin

IRE1 Inositol-requiring enzyme 1

IGF-1 Insulin-like growth factor-1

JNK c-JUN N-terminal kinase

MAPK1 Mitogen-activated protein kinase 1

Melatonin N-acetyl-5-methoxytryptamine

MEK Mitogen activated protein kinases

MACS Magnetic activated cell sorting

MMP11 Matrix metalloproteinase 11


3-(4,5-dimethylthiazol-2-yl)-2,5-
MTT
diphenyltetra-zolium bromide
MDR-1 Multidrug resistance protein 1

MVs Microvesicles

XVI
Exosomes from SNU-C5/5FUR under
N-5FUR-Exo
normal condition
Exosomes from SNU-C5/OXR under
N-OXR-Exo
normal condition
NGS Next generation sequence

OCR Oxygen consumption rate

OX Oxaliplatin

PBS Phosphate-buffered saline

PrPC Cellular prion protein

PARP1 Poly [ADP-ribose] polymerase 1


Platelet endothelial cell adhesion
PECAM-1
molecule-1
Protein kinase R-like endoplasmic
PERK
reticulum kinase
PINK1 PTEN-induced putative kinase 1

PI3K Phosphoinositide 3-kinase

PI Propidium iodide

RNA-seq RNA sequencing

ROS Reactive oxygen species

SNU-C5/5FUR 5FU-resistant SNU-C5 cells

SNU-C5/OXR Oxaliplatin-resistant SNU-C5 cells

SNU-C5/WT Wild type SNU-C5 cells

XVII
Special AT-rich sequence-binding
SATB1
protein-1
SOD Superoxide dismutase

TS Thymidylate synthase

TGFβ Transforming growth factor beta

TP53 Tumor protein P53

VEGF Vascular endothelial growth factor

YAP Yes-associated protein 1

ZO-1 Zonula occludens-1

5FU 5-fluorouracil

XVIII
ABSTRACT

The Enhancement of Therapeutic Effects of Anti-cancer

Reagents in Colorectal Cancer via Regulation of Cellular

Prion Protein

Chul Won Yun

Department of Biomedical Science,

Graduate School of Soonchunhyang University Asan, Korea

(Directed by Professor Sang Hun Lee)

XIX
Colorectal cancer (CRC) is one of the leading causes of cancer-

related death due to its aggressive metastasis in later stages.

Although recent studies have shown the mechanism by which CRC

cells become drug resistant, anti-cancer drug resistance is still a

serious issue for patients with CRC, and novel strategies for

overcoming this drug resistance have not yet been developed. To

solve this problem, several anti-cancer reagents and molecular

targets are tested in this study. Melatonin, an endogenously

secreted indoleamine hormone that is produced in the pineal gland,

is known to possess antitumor effects via various mechanisms

including induction of apoptosis and pro-oxidant effects in various

cancer cells, including CRC. Hypoxia promotes tumor proliferation

and metastasis in CRC. Since the tumor microenvironment is

commonly characterized by hypoxia, its understanding is important

for cancer therapy. Although there is a growing interest in the

tumorigenic role of cellular prion protein (PrPC) in the process of

metastasis, the precise mechanism behind the cellular

communication involving prion proteins remains poorly understood.

Therefore, I hypothesized that 1) PrPC is related to the drug

resistance in CRC cells via regulation of survival signal pathway, and

XX
apoptosis, which is induced by the reactive oxygen species (ROS)-

mediated ER stress; and 2) extracellular vesicles with PrPC

modulated the progression of CRC cells.

To address my hypotheses, I focused on the expression of PrPC

in 5-fluorouracil (5FU)-resistant CRC cells. In specimens from

patients with colorectal cancer, the expression of PrPC was

significantly correlated with metastasis and tumor stages and

affected the tumorigenesis and genetic changes of CRC cells.

Furthermore, in 5FU-resistant CRC cells, PrPC expression is

significantly increased, compared with that in normal CRC cells. In

the presence of 5FU, PrPC increased CRC cell survival and

proliferation by maintaining the activation of the PI3K-AKT

signaling pathway and the expression of cell cycle-associated

proteins, including cyclin E, CDK2, cyclin D1, and CDK4. Moreover,

PrPC inhibited the activation of the stress-associated proteins p38,

JNK, and p53. After treatment of 5FU-resistant CRC cells with 5FU,

moreover, silencing of PrPC triggered apoptosis via the activation of

caspase3.

In addition, I confirmed that melatonin was found to decrease the

expression of PrPC and PTEN-induced putative kinase 1 (PINK1)

XXI
and increase superoxide accumulation in the mitochondria.

Furthermore, PrPC-knockdown potentiated the effects of melatonin

resulting further in significantly reduced expression of PINK1 and

increased superoxide production in CRC. In addition, si-PRNP-

transfected CRC cells treated with melatonin increased the

production of intracellular superoxide and induced endoplasmic

reticulum stress associated protein, and apoptosis.

Next, hypoxic tumor microenvironment increased the PrPC-

expressing exosomes from CRC, and these exosomes regulate the

CRC cell behavior and tumor progression depending on the

expression of PrPC. Hypoxic exosomes from CRC cells promoted

sphere formation, the expression of tumor-inducing genes,

migration, invasion, and tumor growth. Furthermore, these

exosomes increased endothelial permeability, migration, invasion,

and angiogenic cytokine secretion. These effects were associated

with PrPC expression. Application of anti-PrPC antibody with 5FU

significantly suppressed the CRC progression in a murine xenograft

model.

These results indicate that PrPC plays a key role in CRC drug

resistance. Also, melatonin induces mitochondria mediated cellular

XXII
apoptosis in CRC cancer cells via a PrPC-dependent pathway. PrPC

knockdown, combined with melatonin, amplifies the effects of

melatonin, suggesting a novel therapeutic strategy in targeting CRC

cells. Finally, my results indicate that PrP-expressing exosomes

secreted by hypoxic CRC cells are a key factor in the tumorigenic

CRC-to-CRC and CRC-to-endothelial cell communication. Taken

together, these findings suggest that inhibiting PrPC in hypoxic

exosomes during chemotherapy may be an effective therapeutic

strategy in colorectal cancer.

Keywords: colorectal cancer cell, hypoxia, melatonin, 5FU, cellular

prion protein, exosome, drug resistance, antibody therapeutics

XXIII
BACKGROUND

1. Colorectal Cancer

Colorectal cancer (CRC) is the second leading cause of cancer-related

deaths and the third most prevalent malignant tumor worldwide. Early

diagnosis of CRC increases the 5-year survival rate by approximately 64%,

but progression of metastasis decreases the survival rate to 12% (1).

Uncontrolled proliferation of cancer cells is the hallmark of cancer cells and

with this, the ability to metastasize and recur makes it excessively hard to

treat (2). Although the ideal treatment for CRC is surgical control to remove

the tumor and metastases (3), chemotherapy is the typical leading strategy to

control CRC (4). Recent chemotherapy includes fluoropyrimidine-based

single-agent therapy, such as 5-fluorouracil (5FU) and multiple-agent

therapy, including capecitabine, irinotecan, or oxaliplatin (OX) (3). 5FU is a

fluoropyrimidine analogue and is used to treat several cancers, such as

colorectal (4), breast (5), and gastric cancers (6). 5FU acts as an anticancer

agent by inhibiting thymidylate synthase activity, DNA synthesis, and DNA

repair in cancer cells, resulting in cell death (7, 8). Oxaliplatin, a bifunctional

alkylating agent, can inhibit DNA replication and transcription by covalently

binding to DNA, resulting in induction of apoptosis (9).


Despite advancements in chemotherapeutic strategies, drug resistance

commonly occurs in many cancer patients receiving cytotoxic chemotherapy

and molecular targeting drugs and significantly restricted the therapeutic

effects by intrinsic or acquired means (10, 11). Also, drug resistance led to

the failure of cancer therapy and increase of cancer recurrence and metastasis

(12). However, the mechanisms inducing drug resistance are complex and not

clear (13, 14). Current findings show that drug resistance may induce the

inhibition of drug transport and targeting signaling, avoided the immune

response and drug-mediated cell death, and induced the drug-tolerant cells.

Intrinsic drug resistance is related to heterogeneity of cancer cells in tumors.

Tumors consist of a heterogeneous population of cancer cells due to a

difference in genetic and functional context (15). Chemotherapeutic reagents

target the actively proliferating cells and main molecular factors of

tumorigenesis. So, non-proliferative or slow cycling cells in tumors via

nutrients limitations, quiescent stem-like cells, and non-presentation of

tumorigenesis factors are shown to resist cancer therapy, and the tumor often

reoccurred. Acquired drug resistance is originated to the treatment of cancer

cells by exposure of anti-cancer drugs. The decrease in drug efficacy usually

occurs because of alterations in cancer cell physiology, such as the mutation

of the target protein, damage to DNA repair, increased anticancer drug efflux,

and the activation of alternative survival signaling pathways (16, 17).

Furthermore, the tumor microenvironment and hypoxia increase drug

resistance in patients with CRC (18), suggesting that novel combination


therapies, such as targeted therapy and immune checkpoint inhibitor therapy,

are needed to overcome CRC.


2. Cellular prion protein in colorectal cancer

Cellular prion protein (PrPC), a cell surface protein tethered to the

membrane by a glycosylphosphatidylinositol anchor, is highly expressed in a

variety of cells, such as lymphocytes, and tissues, including muscle, heart,

skin, and nervous tissues (19-21). Although studies on PrPC have initially

focused on the nervous system, recent evidence indicates that PrPC regulates

not only self-renewal of stem/progenitor cells and stem cell fate but also

proliferation and resistance to apoptosis in cancer cells (22). Previous studies

indicate that PrPC is associated with cell survival, proliferation, and signal

transduction (23, 24). Also, PrPC promotes anti-oxidant activity against

oxidative stress, which suppresses the formation of ROS generation and

increases interaction with CU2+ (25, 26). Other studies show that PrPC play

important roles in differentiation, neurogenesis, and proliferation of cells (27),

and regulates the expression of extracellular matrix related proteins, which

promotes the adhesiveness of cell (28).

In cancer biology, several studies have indicated that PrPC possess a crucial

role in various physiological functions, such as cell proliferation, apoptosis,

invasion, metastasis, and drug resistance in various cancers (29, 30). PrPC

increases the cell proliferation by activating the PI3K signal pathway and

enhancement of G1/S phase transition via promotion of cell cycle related

proteins in gastric, renal, and colon cancer (31, 32). In CRC, PrPC promotes


cancer cell survival by increasing the uptake of glucose (33). The expression

of PrPC is significantly increased in metastatic gastric cancer cells and induces

the invasion and metastasis by upregulating mitogen activated protein kinases

(MEK)/ERK signal pathway, and matrix metalloproteinase 11 (MMP11) (34).

PrPC enhances the migration and invasion of CRC cells by interaction of HOP

and phosphorylation of the extracellular signal‑regulated protein kinase

(ERK1/2) pathway (35). PrPC augments CRC metastasis through the proto-

oncogene tyrosine-protein kinase (Fyn) - SP1 - special AT-rich

sequence-binding protein-1 (SATB1) pathway (36). Also, PrPC increases

the EMT via promotion of ERK2/ mitogen-activated protein kinase 1 (MAPK1)

in colorectal CSC (34). In addition, PrPC regulates the expression of genes

associated with the mesenchymal subtype by modulation of hippo pathway

effectors Yes-associated protein 1 (YAP) and TAZ and transforming growth

factor beta (TGFβ) pathway and promotes metastasis and shows poor

survival (37). Another study has shown that hypoxia increases the expression

of PrPC in CRC cells and that PrPC regulates CSC markers in CRC cells and

tumor progression (24, 38). In particular, tissues of stage III CRC patients

highly expressed PrPC with Oct4-matched expression (24). PrPC induces the

signal pathway by co-localization with epidermal growth factor receptor

(EGFR) and is related to drug resistance, such as cisplatin, by regulation of

KLF4 and p38 (39).


3. The anti-cancer drugs for cancer therapy

5FU is widely used in the treatment of several cancers, particularly for CRC.

It is an antimetabolite drug that inhibits thymidylate synthase and is

incorporated into RNA, single DNA, and double DNA helix, leading to cancer

cell death (7). Several clinical studies have shown that 5FU-based

chemotherapies and chemo-radiotherapies increased the survival of patients

with several cancers (40-42). However, despite the benefits of 5FU to cancer

therapy, acquired resistance to 5FU is a major clinical problem. Accumulated

evidence shows that abnormally high activities of thymidylate synthase (TS),

deoxyuridine triphosphatase, B-cell lymphoma 2 (Bcl-2), Bcl-XL, and Mcl-

1 lead to 5FU resistance (43). TS is a key factor for 5FU metabolism, and the

expression of TS is the main regulator of 5FU sensitivity in cancer therapy.

High expression of TS is related to the intrinsic resistance to 5FU and induces

the acquired resistance by affecting TS stability due to the treatment of 5FU

(43). In addition, overexpression of TS induces the oncogenesis via decreased

translation of tumor protein P53 (TP53) (44). Many clinical studies have

demonstrated that patients with low expression of TS are more sensitive to

5FU based therapy in cancer therapy (45-47). However, the detailed

molecular mechanisms for 5FU chemoresistance in CRC require further

investigation.

In addition to genetic factors that contribute to the occurrence of the disease,


factors such as diet, physical activity, and physiological homeostasis are

among the important factors associated with occurrence of CRC (48, 49).

Melatonin (N-acetyl-5-methoxytryptamine) is a cytokine secreted by the

pineal gland in the body during sleep that has been known to possess multiple

physiological homeostatic functions and offers numerous benefits associated

with sleep. More specifically, melatonin plays a role in induction of sleep,

regulation of circadian rhythm, immunomodulation, and reduction of oxidation

(50, 51). Interestingly, studies show that melatonin possesses anti-cancer

effects, and in a study related to colorectal cancer, it was shown to induce

senescence and apoptosis of the CRC cells (51, 52). Melatonin promotes the

effects of anti-cancer drugs by decreasing the tumor progression via diverse

signaling pathways including the MAPK, ERK, and AKT pathway (53). The

treatment with melatonin inhibits the cell migration in CRC cells by reducing

the expression of ROCK via modulation of the p38/MAPK signal pathway (54).

The combination of melatonin and 5FU suppresses cell proliferation, migration,

and invasion in CRC cells by caspase/PARP apoptosis pathway and cell cycle

arrest. Also, melatonin and 5FU is inhibited by the phosphorylation of PI3K,

AKT, and enhances the NF-kB translocation to nuclei (55). Cancer cells

treated with melatonin show enhanced production of ROS, resulting in

dysfunctional mitochondria and thus decreased cell viability (56-58). Such

accumulating evidence instigate further investigations into the effects of

melatonin in inducing apoptosis in cancer cells via ROS generation and

mitochondria mediated apoptosis.


4. Extracellular vesicles

Extracellular vesicles (EV) are one of the membrane encapsulated vesicles,

which are secreted by diverse cells into the extracellular space (59). EVs are

classified by three main types, such as exosomes, microvesicles, and

apoptotic bodies (60). Exosomes are nano-sized bilayer structures with cup-

shaped morphology and a size between 30-100 nm (61). Microvesicles (MVs)

are classified by one class of EVs, which are shed from the cellular membrane,

including cellular components, with a diameter from 100 to 1000 nm, and MVs

are usually larger than exosomes (62, 63). Apoptotic bodies are released by

dying cells into extracellular space and they are known to range in size

between 50 - 5000 nm in diameter (64). EVs include diverse functional

molecules, such as proteins, DNA, mRNA, miRNA, lipids, and metabolites, as

well as contribute to cell to cell communications (65, 66). Also, EVs are

directly regulated to the biological responses that interact with receptors on

the recipient cells.

Recent studies have identified the important hypoxia-dependent cell-to-

cell communications in the form of tumor-derived EVs. Whereas, identifying

the protein contents of EVs is still progressing, it is known that tumor cells

actively generate, release, and implement EVs to promote the proliferation,

migration, and angiogenesis of cancer (67). The role of tumor-derived EVs

in regulation of angiogenesis and metastasis has been elucidated in breast


cancer, multiple myeloma, ovarian cancer, melanoma, glioma, and chronic

myeloid leukemia (68). In addition, cancer-derived exosomes have been

identified as inducers of CSC differentiation and therapy resistance (69-71).

Therefore, it becomes evident that cancer-derived EVs play a major role in

the development and manifestation of the hallmarks of cancer, and further

investigations are necessary to develop an effective anti-cancer therapy.


5. The relationship between hypoxia and extracellular vesicles in cancer

Homeostasis for oxygen levels is important for the maintenance of

organisms under physiological and pathophysiological conditions (72).

Hypoxia is a common feature of malignant tumors, which contributes to tumor

angiogenesis, aggressiveness, and metastasis (73, 74). Hypoxia, which is an

environment with restricted oxygen availability, is a characteristic of the

tumor microenvironment during tumorigenesis. Hypoxia plays a pivotal role in

cancer through the alteration of the microenvironment, changes in oncogenes

expression, reprogramming of metabolism, and formation of non-functional

blood vessels, thus, resulting in the induction of metastasis (73, 75). Hypoxia

induces cancer cell proliferation, development of aggressive tumor

phenotypes, CSC phenotypes, and anti-cancer drug resistance (76, 77).

These effects of hypoxia are regulated by hypoxia inducible factor (HIF),

which is a master gene in cancer physiology. It controls the expression of

oncogene transcription factors and relevant target genes as well as cancer cell

metabolism (78, 79). In addition, hypoxia plays a critical, stimulating role in

blood vessel growth and induces tumor growth by angiogenesis in cancer cells

(80). Angiogenesis is associated with the high risk of metastasis, recurrence,

and early patient death (81), and plays important roles in growth, proliferation

and metastasis of CRC (82). Hypoxia is related to stemness and HIF pathway,

and these are pivotal regulators of stem cell differentiation (83). HIF-1α and

10
HIF-2α play important roles in stemness maintenance and modulate the

genes and transcription factors related to cell-renewal and differentiation

(84). CSC are involved in the initiation and maintenance of tumors, and

associated with cancer recurrence and drug resistance (85). One study

reported that self-renewal and differentiation of CSC is modulated by hypoxia

and its signal pathway (86). Under hypoxic conditions, the expressions of

HIF-1α and multidrug resistance protein 1 (MDR-1), which is multidrug

resistant protein MDR-1/ P-glycoprotein (P-gp), are increased and induce

the drug resistance in CRC cells (87). The inhibition of HIF-1α increases

the sensitivity to anti-cancer drugs and promoted apoptosis of CRC cells via

downregulation of MDR1/P-gp expression (88). However, since HIF protein

regulates various down-stream pathways vital for cell physiology, it is

important to investigate HIF-mediated specific target protein/s for cancer

survival.

Hypoxia is influenced by the production of EVs and the contents and

functions of EVs. The crosstalk between cancer cells and other cells is

regulated by EV, especially exosomes, secreted from hypoxia-stimulated

cancer cells (89). Since hypoxic stress leads to significant alterations in the

molecular content and function of exosomes, hypoxic tumor-derived

exosomes transfer some target genes, including glucose transporter,

epidermal growth factor receptor (EGFR), transfer receptors, and P-gp, to

non-hypoxic cells, resulting in the internalization of receptors and clustering

of oncogene/proto-oncogene-activating receptors (90). Under hypoxia,

11
melanoma cells secrete more EVs, compared to normoxic cells, and show an

increase in content of specific proteins and miRNA, which is associated with

poor prognosis of patients with melanoma (91). In addition, the change of

exosome contents by hypoxia enhances the communication between cancer

cells and stroma cells (92) and leads to angiogenesis, survival, proliferation,

invasion, metastasis, and drug resistance of cancer cells (90, 93). Thus, novel

approaches for addressing metastatic cancer must be explored to block the

hypoxic exosome-mediated communication between cancer cells and cells

(94-97).

12
6. Experimental purpose

The relationship with PrPC and drug resistance in colorectal cancer is

examined in diverse studies. My previous studies showed the drug resistant

role of PrPC treated with diverse reagents, such as fucoidan and melatonin in

CRC. PrPC is associated with tumor growth, migration, and proliferation in CRC

cells. The treatment of fucoidan and inhibition of PrPC is shown to reduce cell

proliferation, migration, and induction of apoptosis. Also, during in vivo study,

silencing PrPC and fucoidan decreased the tumor growth and angiogenesis

(98). Another study confirmed that PrPC is related to drug resistance in CRC

cells (99). PrPC demonstrated anti-oxidant effects against the induction of

ROS via treatment of oxaliplatin (OX). The treatment with melatonin inhibited

the PrPC expression and suppressed anti-oxidant enzyme activity, such as

superoxide dismutase and catalase, and induced the ER stress and apoptosis

in OX-resistant CRC cells. Furthermore, melatonin inhibited the colon CSC

via regulation of PrPC-Oct4 axis (24). Co-treatment of 5FU and melatonin

reduced the tumor growth, proliferation, and angiogenesis via a decrease in

CSC abilities by inducing autophagy.

Therefore, in this study, I investigated the effect of PrPC on proliferation

and survival in 5FU-resistant CRC cells. Furthermore, the effects of

melatonin on endoplasmic reticulum stress and apoptosis of colorectal cancer

cells were examined by the regulation of PrPC.

13
Next, tumor hypoxic conditions increase the PrPC-expressing exosomes

secreted by drug-resistant CRC cells, which controls CRC function and tumor

progression. I aimed to investigate the effect of exosomes derived from

hypoxic 5FU- and OX-resistant CRC cells on tumorigenic potential via PrPC

level. Furthermore, I elucidated a novel therapeutic strategy that involved the

co-administration of 5FU and anti-PrP antibody for clinical application in

patients with CRC.

14
PART I. The Role of Cellular Prion Protein in Colorectal Cancer

Cellular Prion Protein Enhances Drug Resistance of Colorectal

Cancer Cells via Regulation of a Survival Signal Pathway

Manuscript is published in

Biomol Ther (Seoul). 2018 26(3):313-321.

Cancers (Basel). 2021 13(9):2144.

15
1. INTRODUCTION

CRC is the third most frequently diagnosed cancer among males and females,

the second leading cause of cancer related death among males, and third

leading cause among females (100). Although surgical techniques,

chemotherapies, and molecular therapies have improved, clinical outcomes

have not kept pace. This lag is mostly because of changes in lifestyle, local

recurrence, distal metastasis, and resistance to chemotherapeutics and

molecularly targeted therapies (17, 101). Among these hurdles, drug

resistance limits therapeutic efficacy, because chemotherapy is one of the

principal modes of cancer therapy (17). In particular, chemotherapy failure

caused by anti-cancer drug resistance induces most cancer related deaths

due to decreases in drug delivery and changes in metabolic enzymes (16).

Therefore, understanding the mechanisms underlying drug resistance plays a

pivotal role in developing reversal strategies and effective cancer therapeutic

combinations.

5FU is widely used in the treatment for several cancers, particularly for

CRC. It is an antimetabolite drug that inhibits thymidylate synthase and is

incorporated into RNA, single DNA, and double DNA helix, leading to cancer

cell death (7). Several clinical studies have shown that 5FU-based

chemotherapies and chemoradiotherapies increased the survival of patients

with several cancers (40-42). However, despite the benefits of 5FU to cancer

16
therapy, acquired resistance to 5FU is a major clinical problem. Accumulated

evidence shows that abnormally high activities of thymidylate synthase,

deoxyuridine triphosphatase, Bcl-2, Bcl-XL, and Mcl-1 lead to 5FU

resistance (43). However, the detailed molecular mechanisms for 5FU

chemoresistance in CRC require further investigation.

PrPC is highly expressed in a variety of cells, such as lymphocytes, and

tissues, including muscle, heart, skin, and nervous tissues (19). Although

studies on PrPC have initially focused on the nervous system, recent evidence

indicates that PrPC regulates not only self-renewal of stem/progenitor cells

and stem cell fate but also proliferation and resistance to apoptosis in cancer

cells (22). In CRC, PrPC promotes cancer cell survival by increasing uptake of

glucose (33). PrPC augmented CRC metastasis through the Fyn-SP1-SATB1

pathway (36). My previous study reveals that silencing PrPC inhibits colon

cancer cell growth (98). However, there is little evidence of a relationship

between PrPC and anti-cancer chemoresistance in CRC cells.

17
2. MATERIALS AND METHODS

2.1. Specimens of patients with colorectal cancer and normal controls

This study and the acquisition of clinical samples were approved by the

Ethics Committee of Seoul Hospital, Soonchunhyang University (IRB: SCHUH

2018-04-032-002), and informed consent was obtained from all study

participants. The serum samples of CRC patients (n=18; grade I, n=90; grade

II, n=90; grade III) and normal control (n=45) were obtained from the

Biobanks of Chonbuk National University Hospital, the Ajou University, and

Keimyung University Dongsan Medical Center, South Korea. CRC tissue

specimens (n=288) in a formalin fixed paraffin embedded (FFPE) block were

obtained from Soonchunhyang University. Clinical information was obtained

from reports and histology sections.

2.2. Cell and spheroid culture of human colon cancer cell line and S707 cancer

stem cells

A human colon cancer cell line (SNU-C5/WT), 5FU-resistant cell (SNU-

C5/5FUR), and oxaliplatin-resistant cell (SNU-C5/OXR) were obtained from

the Chosun University Research Center for Resistant Cells (Gwangju, Korea)

(102). The cells were cultured in RPMI 1640 with 10% fetal bovine serum

(FBS) (Thermo Fisher Scientific, Waltham, MA, USA) at 37℃, 5% CO2

18
condition in a humidified incubator. S707 human colon CSCs were provided by

Prof. Steven M Lipkin from the Department of Medicine, Weill Cornell College

of Medicine (New York, NY, USA) (103). The cells were cultured in ultra-

low attachment plates as spheres in Dulbecco's Modified Eagle

Medium/Nutrient Mixture F-12 (DMEM/F12) medium with supplements

(Thermo Fisher Scientific) at 37℃, 5% CO2 condition in a humidified incubator.

The CSC spheroids were cultured in ultra-low attachment flasks (Corning,

Corning, NY, USA) and maintained by collection via gentle centrifugation,

dissociation to single cells, and replating. In addition, SNU-C5/5FUR and

SNU-C5/OXR and human CSCs (S707) were cultured in ultra-low attachment

six-well plates for spheroid formation at 37℃, 5% CO2 condition in a

humidified incubator.

2.3. Preparation of 5FU

5FU were purchased from Sigma (St. Louis, MO, USA), and 5FU were

dissolved in DMSO at 10 mM concentration. Stock solution was filter-

sterilized using a 0.22 μm pore filter (Sartorius Biotech GmbH, Gottingen,

Germany). Aliquots of stock solution were stored at 4℃ until use.

19
2.4. Cell isolation targeting PrPC using magnetic activated cell sorting

Cell isolation by the expression of PrPC was sorted using manual magnetic

activated cell sorting (MACS) according to the manufacturer’s protocol

(Miltenyi Biotec, Bergisch Gladbach, Germany) (104). Cells were incubated

with human CD230 (PrP)-Biotin primary antibody, followed by a wash with

MACS rinsing solution and attached to anti-Biotin MicroBeads secondary

antibody. After another wash, MACS LS column with active magnetic field

were used to sort and isolate the cells, which were used for flow cytometry

analysis, spheroid formation, and RNA sequencing.

2.5. RNA Sequencing assay for total RNA of sorted colon cancer cells

RNA sequencing (RNA-seq) of total RNA was performed at Macrogen

using TruSeq Stranded mRNA LT Sample Prep Kit (Illumina, San Diego, CA,

USA). The colon cancer cells, sorted by PrPC expression, were used for the

Illumina Small RNA Sequencing protocol using the NovaSeq 6000 S4 Reagent

Kit. Quality control with FastQC (v0.11.7), read trimming with Trimmomatic

(v0.38), and mapped with HISAT2 (v2.1.0), Bowtie2 (v2.3.4.1), and StringTie

(v1.3.4d) were performed. Gene-set enrichment analysis and functional

annotation were done on differentially expressed genes, using Gene Ontology

database. Morpheus software was used for heatmap analysis.

20
2.6. Western blot analysis

Total cell protein was extracted by utilizing RIPA lysis buffer (Thermo

Fisher Scientific). Cell lysates were separated by sodium dodecyl sulfate-

polyacrylamide gel electrophoresis and proteins were transferred to

polyvinylidene fluoride membranes (Millipore, Billerica, MA, USA). The

membranes were blocked with 5% skim milk and incubated with primary

antibodies against PrPC, phospho-phosphatidylinositol-3-kinase (PI3K),

total PI3K, phospho-AKT, total AKT, cyclin dependent kinase (CDK) 2,

CDK4, cyclin D1, cyclin E, phosphor-p38, total p38, phosphor-c-JUN

Nterminal kinase (JNK), total JNK, cleaved caspase-3, cleaved poly [ADP-

ribose] polymerase 1 (PARP1), β-actin (Santa Cruz Biotechnology, Dallas,

TX, USA), phospho-p53, total p53 (Cell Signaling Technology, Danvers, MA,

USA), phospho-ATM serine/threonine kinase (ATM), and total ATM

(Thermo Fisher Scientific). After incubation of membranes with

peroxidaseconjugated goat anti-mouse or anti-rabbit IgG secondary

antibodies (Santa Cruz biotechnology), bands were detected by utilizing

enhanced chemiluminescence reagents (Amersham Biosciences, Uppsala,

Sweden).

2.7. Flow cytometry analysis

Flow cytometry analysis of Oct4, Nanog, and ALDH1A1 was performed to

identify the presence of CSCs (24). A two-color flow cytometry system (BD

21
FACS Canto II; BD, Franklin Lakes, NJ, USA) was used to examine the

immune-stained cells. By comparing the results with the corresponding

negative controls, the percentage of stained cells was calculated.

SNU-C5/WT and SNU-C5/5FUR cells were subjected to flow cytometry

analysis using anti-PrP antibody (Santa Cruz Biotechnology). Flow cytometry

was performed using a Cyflow Cube 8 (Partec, Münster, Germany). A data

analysis was performed using the FCS Express software package (De Novo

Software).

2.8. Immunofluorescence staining

SNU-C5/WT and SNU-C5/5FUR cells on the cover glass slide were fixed

in 4% paraformaldehyde solution for 10 min. After washing three times in PBS,

cells were blocked with 10% goat serum for 1 h, and then incubated with

primary antibodies against PrPC (Santa Cruz Biotechnology) at 4°C for 24 h.

After washing three times in PBS, cells were incubated with secondary

antibodies conjugated with Alexa-488 (Thermo Fisher Scientific) for 1 h.

Nuclei were stained with 4’,6-diamidino-2-phenylindole (DAPI; Sigma).

Stained slides were imaged using a confocal microscope (ZEISS, Oberkochen,

Germany).

22
2.9. siRNA transfection

In accordance with the manufacturer’s protocols, LipofectamineTM 2000

reagent (Thermo Fisher Scientific) was used to transfect siRNAs into CRC

cells. The cells were first grown to 70% confluence in culture dishes and

transfected for 48 h with SMART pool siRNAs (100 nM) specific to PrPC

mRNA. The siRNA ordered to block PRNP (The PRNP-siRNA no. 1 sequence:

5’-UCACCGAGACCGACGUUAA-3’, no. 2 sequence: 5’-

GAUCGAGCAUGGUCCUCUU-3’, no. 3 sequence: 5’-

AGAUGUGUAUCACCCAGUA-3’ and no. 4 sequence: 5’-

GACCGUUACUAUCGUGAAA-3’) and a scrambled sequence (The scramble

siRNA no. 1 sequence: 5’-UGGUUUACAUGUCGACUAA-3’, no. 2

sequence: 5’-UGGUUUACAUGUUGUGUGA-3’, no. 3 sequence: 5’-

UGGUUUACAUGUUUUCUGA-3’, and no. 4 sequence: 5’-

UGGUUUACAUGUUUUCCUA-3’) was purchased by Dharmacon (Lafayette,

CO, USA).

2.10. Kinase assays

The cells were lysed using RIPA lysis buffer (Thermo Fisher Scientific).

CDK4 kinase assays were performed using a CKD4 Kinase Assay Kit (Cusabio,

Baltimore, USA). Briefly, 10 mM ATP was added to 1.25 ml of 6 mM substrate

peptide. The mixture was diluted with dH2O to 2.5 ml to yield a 2×

ATP/substrate cocktail. Then 1 ml of 10× kinase buffer was added to 1.5 ml

23
of dH2O to yield a 2.5 ml 4× reaction buffer and the enzyme was diluted in

reaction buffer to give the reaction cocktail. The reaction cocktail was added

to 12.5 ml/well of prediluted compound of interest (usually approximately 10

mM) and incubated for 5 min at room temperature. ATP/substrate cocktail

was added to 25 ml/well preincubated reaction cocktail/compound. The final

assay conditions for a 50-ml reaction were therefore 25 mM Tris-HCl (pH

7.5), 10 mM MgCl2, 5 mM b-glycerophosphate, 0.1 mM Na3VO4, 2 mM DTT,

200 mM ATP, 1.5 mM peptide, and 50 ng CDK4 kinase. The reaction was

incubated at room temperature for 30 min, then the stop buffer was added.

From each reaction, 25 ml was transferred to a 96-well streptavidin-coated

plate and incubated at room temperature for 1 h. Plates were washed three

times with PBS. Phospho-Rb (Ser780) antibody was added at 100 ml/well

and incubated at room temperature for 2 h. Plates were washed three times

with PBS. HRP-avidin was added to each well, and incubated at 37°C for 1 h.

Plates were washed five times with PBS. TMB substrate and stop solution

were added. The absorbance was read at 450 nm with a microtiter plate reader

(Tecan Group AG, Mänedorf, Switzerland).

2.11. Cell cycle analysis

SNU-C5/5FUR cells were harvested and fixed with 70% ethanol at -20°C

for 2 h. After washing twice with cold PBS, cells were subsequently incubated

with RNase and propidium iodide (PI; Sigma). The cell cycle histograms were

24
assessed using a Cyflow Cube 8 (Partec). Results were analyzed using the

FCS Express software package (De Novo Software).

2.12. PI/Annexin V flow cytometric analysis

Apoptosis of SNU-C5/5FUR and SNU-C5/OXR cells was evaluated with a

Cyflow Cube 8 (Partec) following staining of the cells with Annexin V-

fluorescein isothiocyante (FITC) and PI (Sigma). Data analysis was

performed using the FCS Express software package (De Novo Software).

2.13. Statistical analyses

Data are expressed as means ± SEM. Statistical significance was assessed

using the Student’s t test, where p< 0.05 was considered significant.

25
3. RESULTS

3.1. The clinicopathological features in patients with CRC by the expression of

PrPC

Previous study demonstrated that PrPC controls CSC markers in CRC cells

(24). To investigate whether PrPC is involved in CSC properties in CRC, I

assessed the clinicopathological features in patients with CRC depending on

the expression of PrPC (Table 1). Although the expression of PrPC was not

associated with patient age, sex, pT stage, and vascular invasion, the

expression of PrPC was significantly correlated with pN stage, metastasis,

stage, lymphatic invasion, and perineuronal invasion (Table 2). Consistent

with the observed clinicopathological features in patients with CRC, the 5-

year survival of PrPC negative CRC patients was higher than that of PrPC-

positive CRC patients (Figure 1A). In serum samples of PrPC-positive

patients with CRC, the concentration of PrPC was significantly increased in

stage II and III (Figure 1B). In CRC patients with stage III CRC, PrPC was

highly expressed in colon tissues and lymph nodes (Figure 1C). In addition,

PrPC in serum was significantly increased in stage III patients treated with

chemotherapy, compared with that in stage III patients not treated with

chemotherapy (Figure 1D).

26
Table 1. Clinicopathological features in patients with colorectal cancer.

PrPC Expression Total


Clinicopathological factors p value
Negative (N=133) Positive (N=155) (N=288)

Age, years, mean (SD) 65.3553 (12.2671) 64.0276 (12.8545) 0.365

Gender, N (%) 0.230

M 50 ( 42.1) 66 (56.9) 116

F 83 (48.3) 89 (51.7) 172

pT stage, N (%) 0.424

pT1 and pT2 27 (44.3) 34 (55.7) 61

pT3 and pT4 106 (46.7) 121 (53.3) 227

pN stage, N (%) 0.014

Negative 78 (41.3) 111 (58.7) 189

Positive 55 (55.6) 44 (44.4) 99

Metastasis, N (%) 0.003

Negative 130 (48.5) 138 (51.5) 268

Positive 3 (15.0) 17 (85.0) 20

Stage, N (%) 0.089

and 71 (42.4) 96 (57.5) 167

and 62 (51.2) 59 (48.8) 121

Vascular invasion, N (%) 0.240

Negative 111 (45.1) 135 (54.9) 246

Positive 22 (52.4) 20 (47.6) 42

Lymphatic invasion, N (%) 0.024

Negative 91 (42.5) 123 (57.5) 214

Positive 42 (56.8) 32 (43.2) 74

Perineuronal invasion, N (%) 0.544

Negative 84 (46.2) 98 (53.8) 182

Positive 49 (46.2) 57 (53.8) 106

pT: pathological tumor stage according to the American Joint Committee on Cancer TNM
classification system; pN: pathological lymph node stage according to the American Joint
Committee on Cancer TNM classification system.

27
Table 2. Cox regression analysis of the clinicopathological parameters in

colorectal cancer patients.

Univariate analysis Multivariate analysis


Clinicopathologic
Variable Hazard ratio Hazard ratio
factors p value p value
(95%/CI) (95%/CI)

Age <60 yr vs. ≥60 yr 1.439 (0.736-2.813) 0.288

Gender Male vs. Female 0.912 (0.481-1.729) 0.778

pT stage Low vs. High 3.065 (0.939-10.011) 0.064

pN stage Negative vs. Positive 1.965 (1.012-3.814) 0.046

Metastasis Negative vs. Positive 2.626 (1.017-6.777) 0.046

Stage Low vs. High 2.698 (1.358-5.361) 0.005 3.568 (1.044-12.200) 0.043

Vascular invasion Negative vs. Positive 1.654 (0.722-3.790) 0.235

Lymphatic invasion Negative vs. Positive 2.826 (1.452-5.500) 0.002 2.566 (1.132-5.769) 0.024

PN. invasion Negative vs. Positive 2.428 (1.246-4.730) 0.009

PrPC expression Negative vs. Positive 3.100 (1.408-6.825) 0.005 3.712 (1.642-8.390) 0.002

28
Figure 1. Identification of PrPC expression in samples of CRC patients.

(A) Five-year survival of CRC patients depending on PrPC expression

(negative, n=133; positive, n=155). (B) ELISA analysis of PrPC expression

in sera from patients with CRC (normal, 45; stage I, 18; stage II, 90; stage III,

90). (C) Representative immunohistochemistry images of PrPC in tumor

tissues from colorectal cancer patients (n=33). Scale bar = 200 μm. (D)

ELISA analysis of PrPC expression in sera from CRC stage III patients who

were therapy naive (non-chemotherapy, n=70) and patients who were

treated with chemotherapy (Chemo-therapy, n=20). Data are presented as

mean ± SEM. **p<0.01 ((B): ANOVA; (D) unpaired t-test).

29
3.2. PrPC involved in the CSC properties of CRC

To further explore whether PrPC controls CSC properties in drug-resistant

CRC cells, I investigated the formation of cancer spheres and the expression

of CSC markers, including ALDH1A, Nanog, and Oct4 in 5FU-resistant CRC

cells (SNU-C5/5FUR) and oxaliplatin-resistant CRC cells (SNU-C5/OXR). I

found that the sphere formation capacity and CSC marker expression in each

drug-resistant CRC cell were drastically enhanced in PrP-positive cells

(Figures 2A-D). Moreover, the CSC properties were significantly increased

in PrP-positive CRC stem cells (CSC) (Figures 2E and F).

30
Figure 2. Effect of PrPC of cancer stemness in CRC cells.

(A-F) Sphere formation assay of PrP-negative and PrP-positive SNU-

C5/5FUR (A and B), SNU-C5/OXR (C and D), and CSC (E and F) in ultra-

low attachment plates for 2 weeks (n=3). CSC markers (ALDH1A, Nanog,

and Oct4) were analyzed using flow cytometry analysis (n=3). Scale bar =

200 μm. Data are represented as the mean ± SEM. *p<0.05, **p<0.01

(unpaired t-test).

31
3.3. The change of cancer-related genes in CRC through regulation of PrPC

To determine the effect of PrPC on oncogene expression in drug-resistant

CRC cells, I am performed a NGS on PrP-positive and PrP-negative SNU-

C5/5FUR (Figures 3A–E). The transcriptome data showed that tumor

progression-mediated genes, such as CSC markers, metastasis, angiogenesis,

and oncogenes, were overexpressed in PrP-positive SNU-C5/5FUR,

whereas tumor suppressor genes were decreased (Figures 3A–E). These data

suggest that the PrPC expression level is strongly associated with CRC

progression and prognosis through regulation of CSC properties in CRC.

32
Figure 3. Gene analysis of CRC cells by the PrPC expression.

NGS analysis of PrP-positive and PrP-negative SNU-C5/5FUR (n=3). NGS

analysis of fold changes in PrP-positive vs. PrP-negative cells was

categorized as CSC marker (A), metastasis (B), angiogenesis (C), tumor

suppressor (D), and oncogene (E).

33
3.4. Expression of PrPC in drug-resistant CRC cells

Several studies have shown that the expression of PrPC is increased in

gastric, breast, and CRC cells (105). In addition, PrPC is associated with

multidrug resistance in gastric and breast cancer cells (106, 107). To explore

whether PrPC is upregulated in drug resistant CRC cells, I analyzed the

expression of PrPC in wild type (SNU-C5/WT), SNU-C5/5FUR, SNU-

C5/OXR and CSC cells by ELISA and western blot assay. In several types of

CRC cells, the expression of PrPC was drastically increased in SNU-C5/5FUR,

SNU-C5/OXR, and CSC, compared with that in SNU-C5/WT cells (Figure

4A). The expression level of PrPC was significantly increased in SNU-

C5/5FUR and SNU-C5/OXR, compared with that in SNU-C5/WT (Figures 4B

and C). In addition, flow cytometry analysis showed that the ratio of PrPC

positive cells was significantly increased in SNU-C5/5FUR (Figure 4D).

Immunofluorescent staining also revealed an increased level of PrPC in SNU-

C5/5FUR (Figure 4E). These results indicate that 5FU/OX-induced drug

resistant CRC cells and CSC express high levels of PrPC.

34
Figure 4. The levels of PrPC in SNU-C5/WT, SNU-C5/5FUR, SNU-C5/OXR

and CSC cells.

(A) Concentration of the level of PrPC in SNU-C5/WT (WT), SNU-C5/5FUR

(5FUR), SNU-C5/OXR (OXR), and CSC cells (n=3). (B and C) Expression of

PrPC in SNU-C5/WT, SNU-C5/5FUR and SNU-C5/OXR cells (n=3). (D) The

expression of PrPC in SNU-C5/WT and SNU-C5/5FUR cells was analyzed by

flow cytometry (n=3). (E) Immunocytochemistry for PrPC (green) in SNU-

C5/WT and SNU-C5/5FUR cells. Nuclei were stained by DAPI (blue). Scale

bar=100 mm (n=3).

35
3.5. PrPC regulates CRC cell survival via the PI3K-AKT axis

The phosphoinositide 3-kinase (PI3K)-AKT signaling pathway is a pivotal

regulator and central node in cell survival signaling downstream of growth

factor, cytokines, and several cellular stimuli. To investigate whether PrPC

regulates the PI3K-AKT signaling pathway, I evaluated the phosphorylation

of PI3K and AKT in SNU-C5/WT and SNUC5/ 5FUR. The phosphorylation of

PI3K and AKT were significantly enhanced in SNU-C5/5FUR, compared with

that in SNU-C5/WT (Figure 5A). To investigate whether the expression of

PrPC is associated with regulation of the PI3K-AKT signaling pathway in

5FU-resistant CRC cells, I confirmed activation of PI3K-AKT in the

presence or absence of 5FU (140 mM). Treatment of SNU-C5/WT with 5FU

increased the level of PrPC (Figure 5B). In addition, the level of PrPC was

significantly increased in SNU-C5/5FUR in the presence and absence of 5FU,

compared with in SNU-C5/WT (Figure 5B). The expression of PrPC was

suppressed using PRNP siRNA (si-PRNP) in SNU-C5/5FUR (Figure 5C).

After treatment of SNU-C5/5FUR with 5FU, the suppression of PrPC induced

a decrease in phosphorylation of PI3K and AKT (Figure 5D). These data

suggest that PrPC regulates the PI3K-AKT signaling pathway in 5FU-

resistant CRC cells.

36
Figure 5. The effect of PrPC on cell survival-associated signaling pathways in

CRC cells.

(A) The level of phospho-PI3K (p-PI3K) and phospho-AKT (p-AKT) in

SNU-C5/WT and SNU-C5/5FUR cells (n=3). (B) The level of PrPC in SNU-

C5/WT cells and SNU-C5/5FUR cells after treatment with 5FU (140 mM)

(n=3). (C) The expression of PrPC after transfection of SNU-C5/5FUR cells

with PRNP siRNA (si-PRNP) (n=3). (D) The level of phosphor-PI3K (p-

PI3K) and phospho-AKT (p-AKT) in SNU-C5/5FUR cells transfected with

si-scr or si-PRNP after treatment with PBS or 5FU (140 mM) for 48 h (n=3).

37
3.6. PrPC is involved in cancer cell proliferation through the regulation of cell

cycle-associated proteins

To confirm the effect of PrPC on cell proliferation in 5FU resistant CRC cells,

the expression of cell cycle-associated proteins, such as cyclin E, CDK2,

cyclin D1, and CDK4, was analyzed by western blot assay after treatment of

SNUC5/5FUR with 5FU. SNU-C5/5FUR transfected with PRNP siRNA had a

significant decrease in the levels of cyclin E, CDK2, cyclin D1, and CDK4 after

treatment with 5FU (Figure 6A). In addition, SNU-C5/5FUR transfected with

PRNP siRNA showed a significant decrease in CDK4 kinase activity after

treatment with 5FU (Figure 6B). Furthermore, flow cytometry analysis for PI

staining showed that PrPC knockdown SNU-C5/5FUR exhibited a significant

decrease in the ratio of S phase (Figure 6C) after treatment with 5FU. These

findings suggest that PrPC regulates proliferation of 5FU resistant CRC cells

via regulation of cell cycle-associated proteins in the presence of this anti-

cancer drug.

38
Figure 6. The effect of PrPC on proliferation in CRC cells.

(A) The level of cyclin E, CDK2, cyclin D1, and CDK4 in SNU-C5/5FUR cells

transfected with si-scr or si-PRNP after treatment with PBS or 5FU (140

mM) for 48 h (n=3). (B) The kinase activity of CDK4 in SNU-C5/5FUR cells

transfected with si-scr or si-PRNP after treatment with PBS or 5FU (140

mM) for 48 h (n=3). (C) The percentage of G1, S, and G2/M phase in SNU-

39
C5/5FUR cells transfected with si-scr or si-PRNP after treatment with PBS

or 5FU was determined by flow cytometry analysis of PI-stained cells (n=3).

40
3.7. PrPC regulates anti-cancer drug induced stress-associated signaling

The main mechanism of 5FU is the inhibition of DNA synthesis and mRNA

translation (89). To explore whether PrPC regulates stress-associated

proteins in the presence of this drug, I assessed the phosphorylation of

stress-associated proteins, including p38, JNK, p53, and ATM

serine/threonine kinase (ATM), after treatment of SNU-C5/5FUR with 5FU.

SNU-C5/5FUR transfected with PRNP siRNA exhibited a significant increase

in the phosphorylation of p38, JNK, and p53 (Figures 7A-C) after treatment

with 5FU. However, although knockdown of PrPC induced the phosphorylation

of ATM in the PBS-treated group, there was no significant increase in the

phosphorylation of ATM after treatment with 5FU, implying that PrPC is not

involved in ATM signaling in the presence of 5FU (Figure 7D). These results

indicate that PrPC inhibits the activation of stress-associated signaling against

an anti-cancer drug through suppression of p38, JNK, and p53

phosphorylation.

41
Figure 7. The effect of PrPC on cell stress-associated protein in CRC cells.

The phosphorylation of p38 (A), JNK (B), p53 (C), ATM (D) were determined

by western blot analysis SNU-C5/5FUR cells transfected with si-scr or si-

PRNP after treatment with PBS or 5FU (140 mM) for 48 h (n=3).

42
3.8. PrPC inhibits apoptosis of anti-cancer drug resistant CRC cells via

suppression of caspase3 activation and PARP1 cleavage

To investigate whether PrPC is involved in apoptosis of 5FU resistant CRC

cells, activation of caspase3 and cleavage of PARP1 were assessed using a

western blot after treatment of SNU-C5/5FUR with 5FU (Figure 8A). The

levels of cleaved caspase3 (C-Caspase3) and cleaved PARP1 (C-PARP1)

were significantly increased in treatment with 5FU, compared with those in

treatment with PBS (Figure 8A). SNU-C5/5FUR transfected with PRNP

siRNA had a significant increase in the levels of C-Caspase3 and C-PARP1

after treatment with 5FU (Figure 8A). In addition, flow cytometry for

PI/Annexin V staining showed that silencing of PrPC significantly increased in

the percentage of early and late apoptotic cells after treatment with 5FU

(Figure 8B). These data indicate that PrPC inhibits apoptosis of CRC cells in

anti-cancer drug conditions via suppression of caspase3 activation and

PARP1 cleavage. In addition, SNU-C5/5FUR transfected with PRNP siRNA

showed a significant decrease in the kinase activity of CDK4 after treatment

with 5FU (Figure 6B).

43
Figure 8. The effect of PrPC on apoptosis in CRC cells through inhibition of

caspase3 activation and PARP1 cleavage.

(A) The level of cleaved caspase3 (C-Caspase3) and cleaved PARP1 (C-

PARP1) in SNU-C5/5FUR cells transfected with si-scr or si-PRNP after

treatment with PBS or 5FU (140 mM) for 48 h (n=3). (B) Apoptosis was

measured using flow cytometry analysis for PI/Annexin V staining (n=3).

44
4. DISCUSSION

Previous data have shown a correlation between high PrPC expression and

clinicopathological features in patients with CRC, including metastasis risk,

advanced clinical stage, and survival of CRC patients (38). The expression of

PrPC in tumor tissues from stage III CRC patients is matched with Oct4

expression, indicating that co-expression of PrPC and Oct4 is involved in CRC

metastasis (24). This study revealed that PrP-positive cells were increased

in the properties of a CSC. In RNA-seq data, gene expressions associated

with CSC markers, metastasis, angiogenesis, and oncogenes were

significantly increased in PrP-positive cells, whereas tumor suppressor

genes were reduced. These findings indicated that PrPC plays a pivotal role in

CRC behavior, suggesting that PrPC could be a novel CRC marker for targeted

therapy in CRC patients.

Also, in the present study, I demonstrated that the level of PrPC is increased

in 5FU resistant CRC cells, implying that PrPC increased cell survival in anti-

cancer drug conditions by regulating survival and apoptosis signaling

pathways. After 5FU treatment of SNU-C5/5FUR, a 5FU resistant colorectal

cancer cell line, silencing of PrPC yielded the opposite effects, including

inhibition of proliferation and augmentation of apoptosis. Previous studies

have shown that PrPC contributes to protection from oxidative stress,

regulation of copper metabolism, cell cycle control, and cell adhesion (31,

45
108-110). In addition, PrPC promotes pro-proliferative and anti-apoptotic

effects in stem cells, such as hematopoietic stem cells, neural stem cells,

mesenchymal stem cells, and human embryonic stem cells (22). In cancer cell

biology, PrPC accelerates cell proliferation, invasion, metastasis, and

resistance to cytotoxic drugs (29, 31, 34, 36). My results revealed for the

first time that 5FU resistant CRC cells express high levels of PrPC. In gastric

cancer cells, PrPC expression enhanced drug resistance via an increase in

Bcl-2 expression (111). Moreover, treatment of HCT116 CRC cells with

PrP-specific antibodies inhibited cancer cell growth and promoted the effects

of anticancer drugs such as 5FU, cisplatin, and doxorubicin (112). I also

confirmed that the level of PrPC expression was significantly increased in

oxaliplatin-resistant SNU-C5 cells, compared with wild-type cells. These

findings suggest that PrPC could be a key protein for resistance to anticancer

drugs in CRC cells.

The PI3K signaling pathway is involved in various cellular processes,

including cell survival, metabolism, inflammation, motility, and cancer

progression (113). In particular, the PI3K-AKT signal axis regulates survival

and proliferation in cancer cells (114). In human lung cancer cells, AKT

amplification plays a pivotal role in acquiring cisplatinum resistance (115). My

results indicated that the phosphorylation of PI3K and AKT are significantly

increased in 5FU resistant CRC cells, and inhibition of PrPC expression

reduced the activation of PI3K and AKT in the presence of 5FU. Activation of

AKT also promotes cell proliferation through multiple downstream targets for

46
cell cycle regulation (116). I also revealed that downregulation of PrPC

inhibited proliferation via suppression of cell cycle-associated proteins, such

as cyclin E, CDK2, cyclin D1, and CDK4 after 5FU treatment of 5FU resistant

cells. PrPC accelerated the G1/S transition via the PI3K-AKT-cyclin D1 axis

(31). These findings suggest that PrPC plays pivotal roles in cell survival and

proliferation in drug resistant CRC cells via regulation of the PI3K-AKT

signaling pathway.

Activation of PI3K-AKT pathway by PrPC expression promotes anti-

apoptotic effect (117). Conversely, downregulation of PrPC promotes

caspase3 activation by reducing AKT activation (118). In hypoxic conditions,

increased PrPC inhibits TRAIL-mediated apoptosis (119). My previous study

revealed that silencing of PrPC increased apoptosis of CRC cells. To explore

the relationship between PrPC and anti-cancer drug resistance, I showed that

treatment with 5FU induced the activation of stress-associated proteins, such

as p38, JNK, and p53, and suppression of PrPC triggered additional activation

of these proteins. This additional activation implies that PrPC inhibits the

activation of stress-associated proteins in drug resistant CRC cells.

Furthermore, PrPC suppressed the activation of caspase3 after 5FU treatment

of drug-resistant CRC cells. These results suggest that the level of PrPC plays

an important role in the development of chemoresistance by CRC cells.

47
PART II. The Application of Anti-cancer Reagents for Cancer Therapy

Melatonin Promotes Apoptosis of Colorectal Cancer Cells via

Superoxide-mediated ER Stress by Inhibiting Cellular Prion Protein

Expression

Manuscript is published in Anticancer Res. 2018 38:3951-3960.

48
1. INTR0ODUCTION

CRC is one of the most commonly diagnosed cancers in the world, placing it

as the most common cause of mortality (120). Uncontrolled proliferation of

cancer cells is the hallmark of cancer cells and along with this, the ability to

metastasis and recurrence makes it excessively hard to treat (2). In response,

patients go on extensive chemotherapies inducing apoptosis of target cancer

cells in the body, as well as surgery to remove the cancer tumors (101).

There has been a proliferation of research in drugs used to control CRC and

the progression of the disease. For example, OX, a bifunctional alkylating

agent, can inhibit DNA replication and transcription by covalently binding to

DNA, resulting in induction of apoptosis (9). However, drug resistance of

cancer cells is a major limitation in chemotherapy (17). Although the mutation

of anticancer drug target protein, defective DNA damage repair, enhanced

anticancer drug efflux, and alternative compensating signaling pathways are

potential mechanisms of drug resistance, their toxicity could lead to numerous

side-effects, even favoring growth of cancer cells by killing the patients’

immune system. Thus, natural compounds have been known to be highly

desirable clinically due to their minimal side-effects which are more prevalent

in chemotherapies and other drugs. Not surprisingly, there exist studies that

show natural compounds with anti-cancer effects to be a promising strategy

for cancer prevention and therapy (98, 121-123). More studies on novel

49
compounds and therapeutic mechanisms associated with the compounds are

desirable for developing efficient therapeutic strategies targeting CRC.

In addition to genetic factors that contribute to the occurrence of the disease,

factors such as diet, physical activity, and physiological homeostasis are

among the important factors associated with occurrence of CRC (48, 49).

Melatonin is a cytokine secreted by the pineal gland in the body during sleep

that has been known to possess multiple physiological homeostatic functions

and offer numerous benefits associated with sleep. More specifically,

melatonin plays a role in induction of sleep, regulation of circadian rhythm,

immunomodulation, reduction of oxidation (50, 51). Interestingly, studies

show that melatonin possesses anti-cancer effects, and in a study related to

colorectal cancer, it was shown to induce senescence and apoptosis of the

CRC cells (51, 52). Cancer cells treated with melatonin show enhanced

production of ROS, resulting in dysfunctional mitochondria and thus decreased

cell viability (56-58). Such accumulating evidence instigate further

investigations on the effects of melatonin in inducing apoptosis in cancer cells

via ROS generation and mitochondria mediated apoptosis.

A previous study suggested that melatonin exerts its effects through PrPC-

dependent pathway (99). PrPC, a cell surface protein tethered to the

membrane by glycosylphosphatidylinositol anchor, is highly expressed in cells

of various tissues including nervous, muscle and heart tissue (19-21).

Although mutated prion proteins are most widely known for their

50
neurodegenerative properties, several studies have indicated that cellular

prion proteins possess a crucial role in various physiological functions, such

as cell proliferation, apoptosis, invasion, metastasis and drug resistance in

various cancers (29, 30). My previous studies demonstrated that PrPC is

related to cancer proliferation, and that knockdown of PrPC inhibits colorectal

cancer cell growth (98) and induces tumor cell death (124). PrPC levels in

cancer are associated with ROS-mediated endoplasmic reticulum (ER) stress,

thereby destroying mitochondria and ER to induce cell death (125). PTEN-

induced putative kinase 1 (PINK1), a protein located to the mitochondrial

outer membrane, maintains mitochondria homeostasis through mitochondria

quality control pathway (126). In damaged mitochondria, PINK1 recruits

parkin and autophagy related proteins and results in degradation of damaged

mitochondria through autophagy (127). In a recent study, it was shown that

PINK1 functions as a regulator of cell cycle progression and has tumor

promoting properties (128).

51
2. MATERIALS AND METHODS

2.1. Preparation of melatonin

Melatonin was purchased from Sigma, and was dissolved in 100% ethanol,

filter-sterilized through a 0.45 μm pore filter (Sartorius Biotech GmbH)

Aliquots of stock solution were stored at 4℃ until use.

2.2. Cells and cell culture

The human colorectal cancer cell line (SNUC5/WT) was obtained from the

Chosun University Research Center for Resistant Cells (Gwangju, Republic of

Korea). The cells were maintained in RPMI 1640 supplemented with 10% FBS,

l-glutamine, and antibiotics (Biological Industries, Beit Haemek, Israel) at

37˚C with 5% CO2 in a humidified incubator.

2.3. Cell viability assay

The exponentially growing colon cancer cells were incubated in 96-well

plates with each drugs (0-1 mM Melatonin) for 24 h. The cell viability was

determined by a modification of the 3-(4,5-dimethylthiazol-2-yl)-2,5-

diphenyltetra-zolium bromide (MTT) assay, in which the tetrazolium salt (3-

(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxy-phenyl)-2-(4-

52
sulfophenyl)-2-tetrazolium was converted to formazan by mitochondrial

dehydrogenase in the viable cells. The formazan was quantified by measuring

the absorbance at 570 nm using a microplate reader (BMG Labtech).

2.4. Western blot analysis

Total cell protein was extracted by utilizing RIPA lysis buffer (Thermo

Fisher Scientific). Cell lysates were separated by sodium dodecyl sulfate-

polyacrylamide gel electrophoresis and proteins were transferred to

polyvinylidene fluoride membranes (Millipore). The membranes were blocked

with 5% skim milk and incubated with primary antibodies against PrPC,

phospho-protein kinase R-like endoplasmic reticulum kinase (PERK), PERK,

phospho-eukaryotic initiation factor 2-alpha (eIF2α), eIF2α. activating

transcription factor 4 (ATF4), phospho-c-JUN N-terminal kinase (JNK),

total JNK, phospho-p38, Bcl-2-associated X protein (BAX), cleaved

Caspase-3, cleaved poly [ADP-ribose] polymerase 1 (PARP1), β-Actin

(Santa Cruz Biotechnology), PTEN-induced putative kinase 1 (PINK1),

CCAAT-enhancerbinding protein homologous protein (CHOP) (Novus

Biologicals, Littleton, CO, USA). After incubation of membranes were

incubated with peroxidase conjugated goat anti-mouse or anti-rabbit IgG

secondary antibodies (Thermo Fisher Scientific). Protein bands were

visualized by utilizing enhanced chemiluminescence reagents (Amersham

Biosciences).

53
2.5. siRNA transfection

In accordance with the manufacturer’s protocols, LipofectamineTM 2000

reagent (Thermo Fisher Scientific) was used to transfect siRNAs into CRC

cells. The cells were first grown to 70% confluence in culture dishes and

transfected for 48 h with SMART pool siRNAs (100 nM) specific to PrPC

mRNA. The siRNA ordered to block PRNP (The PRNP-siRNA no. 1 sequence:

5’-UCACCGAGACCGACGUUAA-3’, no. 2 sequence: 5’-

GAUCGAGCAUGGUCCUCUU-3’, no. 3 sequence: 5’-

AGAUGUGUAUCACCCAGUA-3’ and no. 4 sequence: 5’-

GACCGUUACUAUCGUGAAA-3’) and a scrambled sequence (The scramble

siRNA no. 1 sequence: 5’-UGGUUUACAUGUCGACUAA-3’, no. 2

sequence: 5’-UGGUUUACAUGUUGUGUGA-3’, no. 3 sequence: 5’-

UGGUUUACAUGUUUUCUGA-3’, and no. 4 sequence: 5’-

UGGUUUACAUGUUUUCCUA-3’) was purchased by Dharmacon.

2.6. Flow cytometry analysis

To measure mitochondrial superoxide, SNU-C5/WT cells were stained with

MitoSOX red (Thermo Fisher Scientific) for 30 min at 37˚C and washed with

PBS three times. To confirm apoptosis, the cells were stained with Annexin

V-FITC and PI (Sigma). Each sample was quantitively analyzed using CyFlow

Cube 8 (Sysmex Partec). Data analysis was performed using the FCS Express

54
software package (De Novo Software).

2.7. Statistical analyses.

Results are expressed as the mean ± SEM and analyzed by ANOVA. In some

experiments, this was followed by a comparison of the treatment mean with

the control using a Bonferroni-Dunn test. Data were considered to be

significantly different at values of p<0.05.

55
3. RESULTS

3.1. Melatonin decreases cell viability and increases production of superoxide

in SNU-C5/WT cells

To evaluate the effect of melatonin on SNU-C5/WT cells, cell viability was

measured by utilizing the MTT assay after treatment of SNU-C5/WT cells

with melatonin (0, 0.05, 0.1, 0.2, 0.4, 0.6, 0.8, and 1 mM) at various periods

of time (0, 6, 12, and 24 h). Melatonin treatment was shown to reduce the cell

viability of SNU-C5/WT cells in dose- and time-dependent manner (Figures

9A and B). In order to assess superoxide production in SNU-C5/WT cells

treated with Melatonin, the production of mitochondrial superoxide was

examined by flow cytometric analysis (Figures 9C and D). The level of

mitochondrial superoxide was significantly increased after treatment with

melatonin (1 mM). These results suggest that melatonin inhibits cell viability

of SNU-C5/WT cells and increases superoxide production.

56
Figure 9. Melatonin-mediated inhibition of cell viability and induction of

mitochondrial superoxide production in SNU-C5/WT cells.

Cell viability was measured using the MTT assay. (A) SNU-C5/WT cells

were treated with melatonin (0-1 mM) for 24 h (n=3). (B) SNU-C5/WT

colon cancer cells were treated with melatonin (1 mM) for various periods of

time (0, 6, 12, and 24 h). Values represent the means ± SEM. *p<0.05 vs.

control and **p<0.01 vs. control. (C) SNU-C5/WT colon cancer cells were

treated with melatonin (1 mM) for 24 h and superoxide production was

assayed using flow cytometric analysis of MitoSOX red staining (n=3). (D)

The quantitation of the percentage of mitochondrial superoxide levels. Values

represent means ± SEM. **p<0.01 vs. control.

57
3.2. Melatonin suppresses the expression of PrPC and PINK1 and increases the

production of mitochondrial superoxide in SNU-C5/WT cells

In order to assess the effect of melatonin on the expression of PrPC and

PINK1 in SNU-C5/WT, the cells were treated with various concentrations of

melatonin (0, 0.2, 0.6, and 1 mM) at various time periods (0, 6, 12, and 24 h).

Western blot analysis was performed to evaluate the expression of PrPC and

PINK1. The results show that PrPC and PINK1 expression are significantly

reduced upon melatonin (1 mM) treatment for 24 h (Figures 10A-D).

Next, to measure the expression of PrPC and PINK1, SNUC5/WT cells were

transfected with si-PRNP following treatment with melatonin (1 mM). In si-

PRNP transfected cells, melatonin treatment significantly reduced the

expression of PrPC and PINK1 (Figures 11A and B). In order to evaluate the

effect of melatonin on the production of superoxide in SNUC5/WT flow

cytometric analysis was performed. MitoSOX red staining showed that

knockdown of PrPC induced significant production of mitochondrial superoxide

upon melatonin treatment compared to control (Figure 11C). These results

demonstrate that the knockdown of PrPC significantly potentiated the effects

of melatonin.

58
Figure 10. Melatonin mediated inhibition of PrPC and PINK1 in SNU-C5/WT

cells.

SNU-C5/WT cells were treated with different concentrations of melatonin.

PrPC (A) and PTEN-induced putative kinase 1 (PINK1) (B) was determined

by western blot analysis (n=3). (C) SNU-C5/WT cells were treated with

melatonin for different periods of time. PrPC (C) and PINK1 (D) were

determined by western blot analysis (n=3).

59
Figure 11. The effect of PrPC on mitochondrial superoxide production in SNU-

C5/WT cells via treatment with melatonin.

The expression levels of PrPC (A) and PINK1 (B) in SNU-C5/WT cells

transfected with si-PRNP or si-scr following treatment with melatonin (1

mM) for 24 h (n=3). (C) Mitochondrial superoxide was measured using flow

cytometry analysis for MitoSOX red staining (n=3).

60
3.3. Melatonin induces ER stress in SNU-C5/WT cells through regulation of

PrPC expression

To assess the effect of melatonin-induced ER stress in si-PRNP

transfected SNU-C5/WT cells, the expression and activation of ER-stress

associated proteins, such as PERK, eIF2α, ATF4, IRE1α, JNK, and p38 was

examined by western blot analysis (Figures 12A-F). si-PRNP transfected

SNU-C5/WT treated with melatonin (1 mM) showed significantly higher

levels of expression of the ER stress marker (ATF4) and of the

phosphorylation of ER stress regulators (PERK, eIF2α, IRE1α, JNK and p38)

compared to control. These findings indicate that melatonin induces ER stress

and knockdown of PrPC enhances melatonin mediated induction of ER stress.

61
Figure 12. The effect of PrPC on melatonin-induced ER stress in SNU-C5/WT

colorectal cancer cells.

The expression levels of PERK (A), eIF2α (B), ATF4 (C), IRE1α (D), JNK

(E), and p38 (F) were determined by western blot analysis in SNU-C5/WT

cells transfected with si-PRNP or si-scr following treatment with melatonin

(1 mM) for 24 h (n=3).

62
3.4. Inhibition of PrPC expression enhances melatonin-mediated effect of

apoptosis in SNU-C5/WT cells

In order to investigate melatonin-mediated effect on apoptosis in si-PRNP

transfected SNU-C5/WT cells, ER stress marker (CHOP) and apoptosis

associated proteins, including BAX, cleaved caspase3 and cleaved PARP1

were determined by western blot analysis. Under melatonin-mediated

induction of ER stress, knockdown of PrPC significantly enhanced CHOP, Bax,

cleaved caspase3 and cleaved PARP1, compared to untreated cells (Figures

13A-D). Melatonin significantly increased the percentage of early and late

apoptotic cells to 24.47% compared to control (2.79%). In addition, si-PRNP

transfected SNU-C5/WT treated with melatonin show significant increase in

early and late apoptotic cells to 42.81% (Figure 13E). These findings indicate

that combination of si-PRNP and melatonin induces apoptosis of SNU-C5/WT

via ER stress induction.

63
Figure 13. The effect of PrPC on melatonin-enhanced ER stress-mediated

apoptosis in SNU-C5/WT cells.

The expression levels of ER stress marker, CHOP (A) and apoptosis

associated proteins, including BAX (B), cleaved Caspase3 (C), and cleaved

PARP1 (D), were determined by western blot analysis in SNU-C5/WT cells

transfected with si-PRNP or si-scr following treatment with melatonin (1

mM) for 24 h (n=3). (E) Apoptosis of cells was measured utilizing PI/annexin

V staining and flow cytometric analysis (n=3).

64
4. DISCUSSION

In this study, the effects of melatonin, a hormone secreted by the pineal

gland, on the viability of cancer cells was investigated. Melatonin treatment of

SNU-C5/WT induced superoxide levels and decreased cell viability.

Moreover, melatonin inhibited PrPC expression and subsequently reduced

PINK1 expression ultimately destroying mitochondrial homeostasis. In

combination with melatonin, knockdown of PrPC induced further inhibition of

PINK1 expression and production of superoxide, melatonin-mediated ER

stress, and ultimately cellular apoptosis. Together, melatonin treatment and

PrPC knockdown led to increased apoptosis of SNU-C5/WT via PrPC-

dependent mitochondria/ER signaling. My study suggests melatonin as a novel

anti-cancer agent.

Melatonin, a hormone that is most widely known for its sleep inducing

abilities has the potential to regulate CRC cell growth by inducing apoptosis.

Several studies demonstrated that melatonin causes cancer cell death via

mitochondrial reactive oxygen species production (56, 57) and inhibits tumor

growth (122, 129). In addition, another study suggested that melatonin

regulates Endothelin 1 (ET-1) levels and suppresses tumor progression in

CRC (130), suggesting that melatonin has a clear potential in cancer therapy.

In accordance with my previous study, SNU-C5/WT cells, when treated with

melatonin, show decreased cell viability and increased mitochondrial

65
dysfunction in dose and time dependent manner. Many cancer cells show

elevated ROS production compared to normal cells, because of oncogenic

mutation, increased metabolic activity, and cancer microenvironment.

Specifically, superoxide has been known as one of the most potent

mitochondrial deregulators and excess superoxide is associated with

apoptosis in many cancer cells (131-135). My results also suggested that

melatonin caused an increase in superoxide production in cancer cells, leading

to the apoptotic potentials of the cells after the treatment.

Previous studies suggested that PrPC plays a role in the protection of cells

against oxidative stress by clearing up toxic superoxide dismutase (SOD)

activities (108). In addition, studies have shown that when the gene for PrPC,

PRNP, is silenced, the activities of antioxidant enzymes including catalase and

glutathione reductase are reduced (136, 137). Thus, in this study the effects

of melatonin in the induction of ROS and mitochondrial dysfunction through

PrPC dependent pathway was examined. It was found that PINK1 expression

is decreased even further when PrPC was silenced via si-PRNP transfection

compared to treatment with melatonin alone, and that knockdown of PrPC

increased superoxide production in CRC cells even further than melatonin

treatment alone. These results are supported by previous studies showing

that melatonin-induced ROS production of CRC cells lines (56, 57), and this

effect was mediated by decrease of PINK1 expression (138). Thus, my

results suggest that melatonin plays a critical role in superoxide production

and PINK1 expression in SNU-C5/WT cells via PrPC dependent pathway.

66
Further study is required to investigate the precise mechanism of the effects

of melatonin-mediated PINK and PrPC expression in CRC cells.

A previous study has suggested that PrPC function may be related to anti-

oxidant activities against ROS production in cancer (139). Other studies on

PrPC and cancer indicated that PrPC expression levels associate with ROS, ER

stress (125) and induction of tumor cell death (98, 124). The results of this

study support the hypothesis that ER stress is involved in melatonin/PrPC,

mediated apoptosis of cancer cells. My results from western blotting analysis

suggest that melatonin increased ROS-mediated ER stress-associated

proteins resulting in alterations in the levels of oxidative stress-related

proteins, including p-PERK, p-eIF2α, ATF4, p-IRE1α, p-JNK, p-P38, and

CHOP and subsequent apoptosis associated proteins, such as BAX, C-

Caspase3, and C-PARP1, finally leading to cancer cell death (140, 141). In

addition, my western blot analysis results show that silencing of PrPC

magnifies the effects of melatonin on ER and apoptosis, highlighting the

importance of PrPC in melatonin mediated cancer therapy. These results

signify the importance of combination of melatonin and silencing PrPC in CRC

cell treatment which regulates superoxide production and ER stress. However,

further mechanistic studies are required to investigate whether combination

of melatonin and anticancer drugs have synergistic effects in CRC cancer.

67
PART II. The Application of Anti-cancer Reagents for Cancer Therapy

Prion Protein of Extracellular Vesicle Regulates the Progression of

Colorectal Cancer

Manuscript is published in Cancers (Basel). 2021 13(9):2144.

68
1. INTRODUCTION

CRC is the second leading cause of cancer-related deaths and the third most

prevalent malignant tumor worldwide. Early diagnosis of CRC increases the

5-year survival rate by approximately 64%, but progression of metastasis

decreases the survival rate to 12% (1). Although the ideal treatment for CRC

is surgical control to remove tumor and metastases (142), chemotherapy is

the typical leading strategy to control CRC (3). Recent chemotherapy includes

fluoropyrimidine-based single-agent therapy, such as 5FU, and multiple-

agent therapy, including capecitabine, irinotecan, or OX (3). Despite the

advancements in chemotherapeutic strategies, drug resistance restricts the

chemotherapeutic effect by increasing the DNA repair process and drug-

releasing metabolism (143, 144). Furthermore, the tumor microenvironment,

hypoxia, increases drug resistance in patients with CRC (18), suggesting that

novel combination therapies, such as targeted therapy and immune checkpoint

inhibitor therapy, are needed to overcome CRC (3, 144).

Hypoxia is a common feature of malignant tumors, which contributes to

tumor angiogenesis, aggressiveness, and metastasis (73, 74). In particular,

the crosstalk between cancer cells and cells is regulated by extracellular

vesicles, especially exosomes, secreted from hypoxia-stimulated cancer

cells (89). Since hypoxic stress leads to significant alterations in the

molecular content and function of exosomes, hypoxic tumor-derived

69
exosomes transfer some target genes, including glucose transporter,

epidermal growth factor receptor (EGFR), transfer receptors, and P-gp, to

non-hypoxic cells, resulting in the internalization of receptors and clustering

of oncogene/proto-oncogene-activating receptors (90). These hypoxia-

induced exosomes promote tumor angiogenesis, invasion, metastasis, and

tumor immune system. Thus, novel approaches for addressing metastatic

cancer must be explored to block the hypoxic exosome-mediated

communication between cancer cells and cells (90, 94-97).

PrPC is a cell surface glycoprotein and misfolding of PrPC is associated with

neurodegenerative diseases, including transmissible spongiform

encephalopathy and prion diseases (145). In tumor biology, studies have

indicated that PrPC plays an important role in cancer proliferation, invasion,

metastasis, apoptosis, and drug resistance (29, 146). My recent studies have

shown that hypoxia increases the expression of PrPC in CRC cells and that

PrPC regulates CSC markers in CRC cells and tumor progression (24, 38). In

particular, tissues of stage III CRC patients highly expressed PrPC with Oct4-

matched expression (24).

70
2. MATERIALS AND METHODS

2.1. Cell and spheroid culture of human colon cancer cell line and S707 cancer

stem cells

A human colon cancer cell line (SNU-C5/WT), 5FU-resistant cell (SNU-

C5/5FUR), and oxaliplatin-resistant cell (SNU-C5/OXR) were obtained from

the Chosun University Research Center for Resistant Cells (102). The cells

were cultured in RPMI 1640 with 10% FBS (Thermo Fisher Scientific) at 37℃,

5% CO2 condition in a humidified incubator. S707 human colon CSCs were

provided by Prof. Steven M Lipkin from the Department of Medicine, Weill

Cornell College of Medicine (103). The cells were cultured in ultra-low

attachment plates as spheres in Dulbecco's Modified Eagle Medium/Nutrient

Mixture F-12 (DMEM/F12) medium with supplements (Thermo Fisher

Scientific) at 37℃, 5% CO2 condition in a humidified incubator. The CSC

spheroids were cultured in ultra-low attachment flasks (Corning) and

maintained by collection via gentle centrifugation, dissociation to single cells,

and replating. In addition, SNU-C5/5FUR and SNU-C5/OXR and human CSCs

(S707) were cultured in ultra-low attachment six-well plates for spheroid

formation at 37℃, 5% CO2 condition in a humidified incubator. SNU-C5/5FUR

and SNU-C5/OXR and CSCs were treated with exosomes from 1) normoxic,

2) hypoxic, and 3) PRNP siRNA treated conditions. Spheroids measured with

a visual inverted microscope (Olympus, Tokyo, Japan).

71
2.2. Cell culture and characterization of endothelial progenitor cells

Human umbilical vein endothelial cells (HUVECs) (purchased from Thermo

Fisher Scientific) were cultured in complete endothelial cell growth medium-

2 (EGM-2) medium (Lonza, Walkersville, MD, USA), and characterized with

flow cytometry analysis (147) using positive HUVEC marker (anti-human

CD31 or platelet endothelial cell adhesion molecule-1 (PECAM-1); and

negative HUVEC markers (anti-human CD45, and anti-human CD11b)

purchased from BD Pharmingen, San Diego, CA, USA.

2.3. Preparation of 5FU and oxaliplatin

5FU and oxaliplatin were purchased from Sigma, and 5FU and oxaliplatin

were dissolved in DMSO at 10 mM concentration. Stock solution was filter-

sterilized using a 0.22 μm pore filter (Sartorius Biotech GmbH). Aliquots of

stock solution were stored at 4℃ until use.

2.4. Flow cytometry analysis

Flow cytometry with CD81 and CD63 was used to confirm exosome markers

(148). A two-color flow cytometry system was used to investigate the

immune-stained exosomes. The percentage of stained exosomes was

calculated by comparing the negative controls. Each sample was quantitatively

72
analyzed using a CyFlow Cube 8 (Sysmex Partec). A data analysis was

performed using the FCS Express software package (De Novo Software).

To analyze the cell cycle, cells were incubated with RNase and stained with

PI (Thermo Fisher Scientific). To confirm the cell proliferation, the cells were

washed once in phosphate-buffered saline (PBS) and incubated with 10 μM

Carboxyfluorescein succinimidyl ester (CFSE) (Molecular Probes, Eugene,

Oregon, USA) for 10 min at 37℃. The CSFE-labeled cells were washed twice

in PBS and analyzed using a flow cytometer (CyFlow Cube 8) (24).

2.5. Isolation of exosomes

Conditioned media were obtained from CRC cells (3 × 106) after 48 h of

incubation with serum-free media. Exosomes were isolated from conditioned

media (60 ml per group) of normoxic, hypoxic, or si-PRNP pretreated

hypoxic CRC cells using an exosome isolation kit (Rosetta Exosome, Seoul,

Korea) (149). First, conditioned media was precleared by differential

centrifugation and concentrated using a centrifugal filter (Sigma-Aldrich).

And then, extracellular vesicle enrichment is obtained by using Solution A, B,

and C. Finally, purified exosomes is collected by the usage of spin-based

size-exclusion column. The protein concentration of exosomes is measured

by the bicinchoninic acid (BCA) assay. For cell treatment, 2 μg of exosomes

based on protein measurement using BCA assay were added to 2 × 105 cells.

73
2.6. Identification of exosomes via cryo-electron microscopy

For cryo-electron microscopy (Cryo-EM), 5 μL of exosomes were loaded

on 300-mesh EM carbon grids with a hydrophilic surface and frozen using

Vitrobot (Thermo Fisher Scientific) in liquid nitrogen (150). The grids were

observed and analyzed using Talos L120C cryo-EM (Thermo Fisher

Scientific), and images were recorded at 13,000 magnification.

2.7. Dynamic light scattering analysis

The size of exosomes derived from colon cancer cells was measured using

the ELSZ-1000 analyzer (Otsuka electronics, Kobe, Japan) (151). Briefly,

exosomes (10 μL) were diluted to 1:100 in PBS. The solution was measured

by performing the zeta-potential and particle size analysis to confirm the

presence of exosomes.

2.8. Detection of PrPC concentration via enzyme-linked immunosorbent assay

Concentrations of PrPC in serum sample (100 μL) or isolated exosomes

(50 μg) were analyzed using enzyme linked immunosorbent assay (ELISA)

with commercial kit manufactured by Lifespan Biosciences, Seattle, WA, USA.

PrPC were quantified by its absorbance at 450 nm using a microplate reader

(BMG Labtech).

74
2.9. Invasion assay

Matrigel-coated transwell cell culture chambers (8-μm pore size; Sigma-

Aldrich) and serum-free RPMI 1640 or endothelial cell growth basal

medium-2 (EBM-2) medium were used to assess the invasion of SNU-

C5/5FUR, SNU-C5/OXR, and HUVECs. The cells were first treated with

SNU-C5/5FUR or SNU-C5/OXR exosomes derived from different conditions

for: hypoxic, normoxic, and/or transfected with si-PRNP, and incubated for

72 h at 37℃ then invasion assay was performed. Cells were stained with 2%

crystal violet, and invasive cells were quantified and photographed using a

light microscope.

2.10. Wound-healing migration assay

Cells were added in 60 mm cell culture plates and grown at 90% confluency

in 4 ml of culture medium (98). A scratch was made in the cell layer by using

sterile pipette tip and cultured for 24 h at 37℃. The images were obtained

using a microscope (Eclipse TE300, Nikon, Tokyo, Japan).

2.11. Morpholometric analysis

Morphological changes in colon cancer cell lines were examined by phase-

contrast microscopy (Nikon, Tokyo, Japan). The cells were cultured in 24-

well plates (7,000 cells/well). Cell images were obtained using phase-

75
contrast microscopy. The average cell size was calculated from at least three

different visual fields in three independent dishes using ImageJ software.

2.12. Human angiogenesis protein array

Commercially available human angiogenesis antibody array (Abcam,

Cambridge, UK) (152) was used to measure the expression of angiogenesis

proteins in HUVECs treated with exosomes, Approximately 200 μg of total

lysates protein was analyzed following the protocol.

2.13. siRNA transfection

In accordance with the manufacturer’s protocols, LipofectamineTM 2000

reagent (Thermo Fisher Scientific) was used to transfect siRNAs into CRC

cells. The cells were first grown to 70% confluence in culture dishes and

transfected for 48 h with SMART pool siRNAs (100 nM) specific to PrPC

mRNA. The siRNA ordered to block PRNP (The PRNP-siRNA no. 1 sequence:

5’-UCACCGAGACCGACGUUAA-3’, no. 2 sequence: 5’-

GAUCGAGCAUGGUCCUCUU-3’, no. 3 sequence: 5’-

AGAUGUGUAUCACCCAGUA-3’ and no. 4 sequence: 5’-

GACCGUUACUAUCGUGAAA-3’) and a scrambled sequence (The scramble

siRNA no. 1 sequence: 5’-UGGUUUACAUGUCGACUAA-3’, no. 2

sequence: 5’-UGGUUUACAUGUUGUGUGA-3’, no. 3 sequence: 5’-

76
UGGUUUACAUGUUUUCUGA-3’, and no. 4 sequence: 5’-

UGGUUUACAUGUUUUCCUA-3’) was purchased by Dharmacon.

2.14. Western blot analysis

Total protein was extracted using RIPA lysis buffer (Thermo Fisher

Scientific). Cell lysates (20 μg) were separated by 10% sodium dodecyl

sulfate-polyacrylamide gel electrophoresis, and proteins were transferred

onto polyvinylidene fluoride membranes for detection. After washing with

TBST (10 mM Tris-HCl [pH 7.6], 150 mM NaCl, and 0.05% Tween-20), the

membranes were blocked with 5% skim milk for 1 h and then incubated with

primary antibodies specific to PrPC, CD81, CD63, and β-actin, followed by

washing and another incubation peroxidase-conjugated secondary antibodies.

Visualization of the band was done with chemiluminescence.

2.15. BrdU incorporation assay

The cell proliferation parameters were assessed using a cell proliferation

5-bromo-2′-deoxyuridine (BrdU) ELISA colorimetric kit (Roche,

Mannheim, Germany). To perform ELISA, 100 μg/mL BrdU was added to

CRC cells and incubated at 37℃ for 3 h. Subsequently, 1 M H2SO4 was added

to stop the reaction. The light absorbance at 450 nm of the samples was

measured by using an ELISA reader (BMG labtech).

77
2.16. Measurements of oxygen consumption rate

The mitochondrial oxygen consumption rate (OCR) was measured using an

XF96e Extracellular Flux Analyzer (Agilent Technologies, Santa Clara, CA,

USA) following the instructions. Sequential injections of oligomycin, carbonyl

cyanide m-chlorophenylhydrazone, rotenone, and antimycin A provide

information for mitochondrial basal respiration, maximal respiration, ATP

turnover, and spare respiratory capacity respectively. Final results were

presented as the percentage of change compared with the control.

2.17. Tumorigenesis in CRC xenograft mice models

Mice xenograft model of CRC using BALB/c nude mouse was created with

subcutaneous injection of SNU-C5/WT cells (5 × 106). When the tumors

reached a volume of 10 mm3, 5FU (Sigma-Aldrich) and anti-PrP antibody

(Santa Cruz Biotechnology) were administered together. After 28 days of

drug administration, the mice were euthanized for histology. Two

perpendicular tumor dimensions (a = length, b = width) were measured with

a Vernier caliper and the volume (V; mm3) was calculated with the formula V

= (a × b2)/2 (153). The tumor specimens were fixed in 4% formaldehyde,

embedded in paraffin, sliced into 4 μm-thick sections, and stained for

immunohistochemistry-based analysis.

78
2.18. In vitro vascular permeability assay

The permeability of treated HMVEC monolayers grown on transwell filters

(0.4 μm pore size; BD Biosciences) was assessed by the passage of

rhodamine B isothiocyanate-dextran (average MW ~70,000; Sigma). Briefly,

rhodamine-dextran was added to the top well at 20 mg/ml, and the appearance

of fluorescence in the bottom well was monitored by measuring 40 μl medium

aliquots in a time course using a victor3 multiple microplate reader (Perkin

Elmer, Waltham, Massachusetts) at 544 nm excitation and 590 nm emission.

2.19. In vivo vascular permeability assay

For the in vivo vascular permeability assay, the mice were treated with

experimental exosomes for 21 days (154). 100 mg/kg rhodamine-dextran

(Sigma-Aldrich; average MW ~70,000) were administered intravenously via

tail vein. Transcardiac perfusion was performed 3 hours post injection to

remove the excess dye. Tissues specimens were either embedded in Tissue-

Plus OCT Compound (Thermo Fisher Scientific) and cryo-sectioned for

fluorescent microscopy or fixed for hematoxylin and eosin (H&E) staining for

histological analysis.

79
2.20. Immunofluorescence staining

For histological analysis, human colon cancer tissue or mouse tumorigenesis

model cancer tissue was fixed in 4% paraformaldehyde (Affymetrix, Santa

Clara, CA, USA) and embedded in paraffin. Paraffin-embeded sections of

tissue samples were incubated with the primary anti-bodies against CD31,

zonula occludens-1 (ZO-1), KI67, and cleaved caspase-3 (Santa Cruz

Biotechnology) depending on the experimental conditions. Alexa488-

conjugated or Alexa594-conjugated secondary antibodies (Thermo Fisher

Scientific) were used. DAPI (Vector Laboratories, Burlingame, CA, USA) was

used to mark cell nuclei. Confocal microscopy (Leica, Wetzlar, Hesse,

Germany) images were taken for subsequent analysis.

2.21. Ethics statement

All animal studies were approved by the Institutional Animal Care and Use

Committee of Soonchunhyang University and fulfilled in accordance with the

National Research Council Guidelines for the Care and Use of Laboratory

Animals. This study used male Balb/C nude mice (8–10 weeks old;

Biogenomics, Seoul, Korea). All animals were maintained in a pathogen-free

facility under a 12-h light/dark cycle at 25℃ with free access to water and

laboratory chow.

80
2.22. Statistical analysis

Results were expressed as the mean ± SEM, and two-tailed Student’s t

test or one- or two-way analysis of variance was used to compute the

significance between the groups. Comparisons of three or more groups were

performed using Dunnett’s or Tukey’s posthoc test. A p value of < 0.05

was considered significant.

81
3. RESULTS

3.1. Hypoxia-induced exosomes isolated from drug-resistant CRC

During the rapid development of tumors, hypoxia stimulates the

hypersecretion of membrane-bound vesicle known as exosomes that can

induce angiogenesis, metastasis, and immunosuppression to drive tumor

progression (155, 156). To assess the effect of hypoxia on the component of

exosomes in drug-resistant CRC and to identify the key molecules in

regulating drug-resistant CRC properties, I am isolated the exosomes from

SNU-C5/5FUR under normal (N-5FUR-Exo) and hypoxic conditions (H-

5FUR-Exo) and characterized them (Figures 14A–C). N-5FUR-Exo and H-

5FUR-Exo expressed the exosome markers CD81 and CD63 (Figure 14D).

My previous studies have shown that Oct4 and PrPC are highly expressed

simultaneously in patients with CRC, and hypoxia significantly increased the

level of PrPC (24, 38), and this was confirmed as significant PrPC upregulation

in N-5FUR-Exo (Figure 14D) and even higher expression H-5FUR-Exo

(Figures 14D and E).

82
83
Figure 14. Characterization of exosomes secreted by normoxic and hypoxic

drug-resistant CRC cells.

(A) Representative cryo-electron microscopy analysis of exosomes isolated

from SNU-C5/5FUR under normoxic and hypoxic conditions. Scale bar = 200

nm (n=3). (B) Representative flow cytometry histogram of exosome markers,

CD81 and CD63, on exosomes under normoxic and hypoxic conditions for 48

h (n=3). (C) Size distribution analysis by dynamic light scattering (n=3). (D)

Expression of PrPC, CD81, and CD63 in N-5FUR-Exo, H-5FUR-Exo, H-

5FUR-Exo + si-PRNP, and H-5FUR-Exo + si-scr (n=3). (E) ELISA

analysis of PrPC expression in N-5FUR-Exo, H-5FUR-Exo, H-5FUR-Exo

+ si-PRNP, and H-5FUR-Exo + si-scr (n=3). Data are represented as the

mean ± SEM. *p<0.05, **p<0.01 (ANOVA).

84
3.2. Hypoxia-induced exosomes isolated from drug-resistant CRC increasing

sphere formation via upregulation of PrPC

To determine whether PrPC in exosomes regulates the function of drug-

resistant CRC cells, I assessed the sphere formation in SNU-C5/5FUR and

SNU-C5/OXR after treatment with exosomes isolated from each cell under

normoxic or hypoxic conditions (Figure 15A). The capacity of sphere

formation in SNU-C5/5FUR and SNU-C5/OXR was significantly increased

following treatment with H-5FUR-Exo or H-OXR-Exo (Figure 15B). To

further confirm the effect of exosomes on tumor function in CRC, I initially

assessed the capacity of sphere formation in CSC after treatment with CSC-

derived exosomes (Figure 15C). The number of spheres in CSC were

significantly increased after treatment with H-CSC-Exo, compared with that

without treatment and in N-CSC-Exo (Figure 15D). In particular, the

inhibition of PrPC significantly decreased the number of spheres in CSC

treated with H-CSC-Exo (Figures 15C and D), suggesting that PrPC in cancer

cell-derived exosomes regulates the ability of sphere formation in CRC.

85
86
Figure 15. Sphere formation of SNU-C5/5FUR, SNU-C5/OXR and CSC after

treatment with exosomes.

(A) Sphere formation assay of SNU-C5/5FUR and SNU-C5/OXR treated with

exosomes isolated from SNU-C5/5FUR and SNU-C5/OXR under normoxic

and hypoxic conditions. Scale bar = 200 μm (n=3). (B) Quantification of the

number of spheres is shown as a bar graph. (C) Sphere formation assay of

CSC treated with exosomes isolated from CSC under normoxic and hypoxic

conditions. Scale bar = 200 μm (n=3). (D) The graph shows the number of

CSC spheres after treatment with exosomes. Data are presented as mean ±

SEM. **p<0.01 vs. non-treatment; ##p<0.01 vs. non-treatment (ANOVA).

87
3.3. Hypoxia-induced exosomes isolated from drug-resistant CRC increasing

invasion, migration, and proliferation via upregulation of PrPC

In addition, to determine whether PrPC in exosomes regulates the function

of drug-resistant CRC cells, I confirmed the invasion and migration in SNU-

C5/5FUR and SNU-C5/OXR after treatment with exosomes isolated from

each cell under normoxic or hypoxic conditions (Figures 16 and 17). The

invasion and migration capacities of drug-resistant CRC cells were

significantly increased after treatment with H-5FUR-Exo or H-OXR-Exo

(Figures 16A and B, 17A and B). The proliferation capacity of CRC cells also

significantly increased after treatment with hypoxia-stimulated exosomes

(Figures 18A–F). Furthermore, the elongated mesenchymal-like morphology

was significantly induced after treatment with H-5FUR-Exo or H-OXR-Exo

(Figures 19A and B). However, the knockdown of PRNP blocked the effect of

hypoxia-stimulated exosomes (Figures 16-19). These findings indicated

that increased levels of PrPC in hypoxia-stimulated exosomes enhance CRC

cell functions, such as sphere formation, invasion, migration, and proliferation.

88
Figure 16. Effect of exosomes secreted by hypoxic drug-resistant CRC cells

on invasion.

(A) Representative invasion analysis is shown as scale bar = 200 μm (n=3).

(B) The average number of invasive cells is shown as a bar graph. Data are

presented as mean ± SEM. *p<0.05, **p<0.01 vs. non-treatment; #p<0.05,

##p<0.01 (ANOVA).

89
Figure 17. Effect of exosomes secreted by hypoxic drug-resistant CRC cells

on migration.

(A) Representative wound-healing analysis is shown as scale bar = 200 μm

(n=3). (B) The average wound closure is shown as a bar graph. Data are

presented as mean ± SEM. *p<0.05, **p<0.01 vs. non-treatment; #p<0.05,

##p<0.01 (ANOVA).

90
91
Figure 18. Proliferation capacity of drug-resistant CRC cells after treatment

with exosomes.

CFSE analysis of SNU-C5/5FUR (A), SNU-C5/OXR (B), and CSC (C) after

treatment with exosomes secreted by each cell under normoxic and hypoxic

conditions (n=3). BrdU incorporation analysis in SNU-C5/5FUR (D), SNU-

C5/OXR (E), and CSC (F) after treatment with exosomes secreted by each

cell under normoxic and hypoxic conditions (n=3). Data are presented as

mean ± SEM. *p<0.05, **p<0.01 vs. non-treatment; #p<0.05, ##p<0.01

(ANOVA).

92
Figure 19. Effect of exosomes secreted by hypoxic drug-resistant CRC cells

on morphological change.

(A) Representative morphological change analysis of SNU-C5/5FUR and

SNU-C5/OXR treated with exosomes isolated from SNU-C5/5FUR and

SNU-C5/OXR under normoxic and hypoxic conditions. Scale bar = 50 μm

(n=3). (B) The average number of morphologically changed cells is shown as

a bar graph (n=3). Data are presented as mean ± SEM. *p<0.05 vs. non-

treatment; #p<0.05 (ANOVA).

93
3.4. The characterization of HUVECs and uptake of Hypoxia-stimulated CRC

exosomes

To investigate whether hypoxia-stimulated CRC exosomes induce tumor

angiogenesis, I am characterized HUVECs (Figures 20A and B) and treated

Dil-labeled 5FUR-exosomes with HUVECs. Fluorescence microscopic

images showed that Dil-labeled 5FUR exosomes were taken up by HUVECs

(Figure 21A). In HUVECs containing Dil-labeled 5FUR exosomes, H-5FUR-

Exo significantly increased the level of PrPC compared with N-5FUR-Exo

and si-PRNP + H-5FUR-Exo (Figure 20C), suggesting that PrPC in

exosomes isolated from CRC cells is transferred into endothelial cells.

94
Figure 20. Characterization of HUVECs and expression of PrPC after treatment

with exosomes.

(A) Representative flow cytometry histogram of HUVECs positive markers,

CD31, and negative markers CD45 and CD11b (n=3). (B) Representative flow

cytometry histogram of the uptake of tetramethylindocarbocyanine

perchlorate (DiI)-labeled exosomes in HUVECs (n=3). (C) ELISA analysis

of PrPC expression in HUVECs after treatment with exosomes secreted by

normoxic and hypoxic SNU-C5/5FUR (n=3). Data are presented as mean ±

SEM. **p<0.01 vs. non-treatment; ##p<0.01 (ANOVA).

95
Figure 21. Uptake of exosomes into HUVECs.

(A) Representative immunofluorescence analysis of intracellular uptake of

DiI-labeled exosomes (red) to HUVECs. Scale bar = 50 μm (n=3).

96
3.5. Hypoxia-stimulated CRC exosomes promoting tumor angiogenesis and

vascular permeability

To assess the effect of hypoxia-stimulated exosomes on HUVEC migration

and invasion, the migration and invasion capacities of HUVECs after treatment

with exosomes were assessed. The migration and invasion capacities of

HUVECs were significantly augmented after treatment with H-5FUR-Exo

compared with that in other groups (Figures 22A–D). The silencing of PRNP

in H-5FUR-Exo significantly inhibited the migration and invasion capacities

(Figures 22A–D). In particular, the expression of angiogenic cytokine, C-X-

C motif chemokine 5 (ENA-78), in HUVECs treated with exosomes

significantly increased after treatment with H-5FUR-Exo (Figures 23A and

B). Moreover, permeability assay showed that treatment of HUVECs with H-

5FUR-Exo significantly increased the permeability of HUVECs (Figure 24A).

To further reveal whether exosomes secreted by hypoxia-stimulated drug-

resistant CRC cells attenuated the endothelial barrier in vivo, I am injected

exosomes isolated from SNU-C5/5FUR to mouse models and analyzed the

permeability of blood vessels (Figure 24B). In the liver and lungs, injection

with H-5FUR-Exo increased the permeability of blood vessels (Figure 24B).

Furthermore, the endothelial tight junction was significantly decreased after

injection of H-5FUR-Exo by decreasing in the level of ZO-1 (Figure 25A).

However, knockdown of PRNP blocked the effect of H-5FUR-Exo on

angiogenesis and permeability of endothelial cells in vitro and in vivo (Figures

22–25). These data indicated that exosomes secreted by hypoxia-stimulated

97
drug-resistant CRC cells increase tumor angiogenesis and the permeability

of endothelial cells via PrPC.

98
Figure 22. Effect of exosomes secreted by hypoxic drug-resistant CRC cells

on the motility of HUVECs.

(A) Representative wound healing assay of HUVECs treated with exosomes

isolated from SNU-C5/5FUR under normoxic and hypoxic conditions. Scale

bar = 200 μm (n=3). (B) The average number of wound closures is shown

99
as a bar graph. Data are presented as mean ± SEM. **p<0.01 vs. Prior to

migration. #p<0.05, ##p<0.01 (ANOVA). (C) Representative invasion

analysis of HUVECs treated with exosomes isolated from SNU-C5/5FUR

under normoxic and hypoxic conditions. Scale bar = 200 μm (n=3). (D) The

average number of invasive cells is shown as a bar graph. Data are presented

as mean ± SEM. *p<0.05, **p<0.01 vs. non-treatment. ##p<0.01 (ANOVA).

100
Figure 23. Effect of exosomes secreted by hypoxic drug-resistant CRC cells

on angiogenesis.

(A) Representative immunoblot analysis of angiogenesis related proteins in

HUVECs treated with PBS, N-5FUR-Exo, H-5FUR-Exo, and si-PRNP +

101
H-5FUR-Exo (red square: angiogenin, yellow square: EGF, green square:

ENA-78, and blue square: bFGF). (B) Average pixel density of immunoblot

are shown as the bar graph (n=2). Data are presented as mean ± SEM.

*p<0.05, **p<0.01 vs. PBS (ANOVA).

102
Figure 24. Effect of exosomes secreted by hypoxic drug-resistant CRC cells

on vascular permeability.

(A) The permeability of treated HUVEC monolayers grown on 0.4 mm filters

was measured by the appearance of rhodamine-dextran, which was added to

the top well at the beginning of the experiment, in the bottom well during a 1

h time course. The absorbance at 590 nm at each time point is indicated (n=3).

Data are presented as mean ± SEM. **p<0.01 (ANOVA). (B) Representative

103
images of vascular permeability in vivo on liver and lung treated with

intravenously injected exosomes detected by the appearance of intravenously

injected rhodamine-dextran (red) (n=3). Scale bar = 50 μm.

104
Figure 25. Effect of exosomes secreted by hypoxic drug-resistant CRC cells

on blood vessels.

(A) Representative immunofluorescence analysis of ZO-1 (green) and CD31

(red) expression in liver and lung treated with intravenously injected

exosomes (n=3). Scale bar = 50 μm.

105
3.6. Administration of 5FU, anti-PrP antibody, cetuximab inhibiting CRC

progression through suppression of PrPC level

To confirm the effect of 5FU, anti-PrP antibody, or cetuximab on CRC

progression, I initially assessed the tumor size after treatment with 5FU, anti-

PrP antibody (0.05 or 0.5 mg/kg), or cetuximab (0.5 mg/kg) twice a week in

a SNU-C5/WT xenograft model and investigated the level of PrPC in serum

(Figures 26A–D). In a wild-type CRC xenograft model, treatment with 5FU,

anti-PrP antibody (0.05 or 0.5 mg/kg), or cetuximab significantly decreased

the tumor size (Figures 26B and C). The level of PrPC in serum was

significantly reduced after treatment with 5FU, anti-PrP antibody (0.05 or

0.5 mg/kg), or cetuximab, compared with that treated with PBS (Figure 26D).

106
Figure 26. Effect of anti-PrP antibody in a wild type CRC xenograft model.

(A) Schematic illustration of SNU-C5/WT cells in vivo subcutaneously

transplantation and injection with PBS, 5FU, anti-PrP antibody (0.05 mg or

0.5 mg), and cetuximab (0.5 mg). (B) Photographs of tumor growth in a

107
murine xenograft mouse model. (C) Quantification of tumor size in each group

(n=10). (D) ELISA analysis of PrPC expression in sera isolated from a murine

xenograft model (n=10). Data are presented as mean ± SEM. *p<0.05,

**p<0.01 (ANOVA).

108
3.7. Co-administration of 5FU and anti-PrP antibody inhibiting CRC

progression through suppression of PrPC level

To further determine whether hypoxia-induced exosomes isolated from

drug-resistant CRC affect CRC progression and co-administration of 5FU and

anti-PrP antibody suppresses CRC progression in SNU-C5/WT cells

pretreated with an exosomes xenograft model (Figure 27A), I assessed the

tumor size in SNU-C5/WT cells pretreated with exosomes xenograft model

after treatment with H-5FUR-Exo + PBS, H-5FUR-Exo + 5FU, H-5FUR-

Exo + 5FU + anti-PrP antibody, or H-5FUR-Exo + anti-PrP antibody

(Figure 27B). I also assessed the tumor size in a SNU-C5/WT xenograft

model, as a negative control, after treatment with PBS (no Exo + PBS) and

5FU (no Exo + 5FU) (Figure 27B). In a SNU-C5/WT pretreated with H-

5FUR-Exo xenograft model, co-treatment with 5FU and anti-PrP antibody

(H-5FUR-Exo + 5FU + Anti-PrP) significantly decreased the tumor size

(Figure 27C). The concentration of PrPC in serum was also significantly

reduced after co-treatment with 5FU and anti-PrP antibody, compared with

that in cells treated with PBS (H-5FUR-Exo), 5FU (H-5FUR-Exo + 5FU),

or anti-PrP antibody (H-5FUR-Exo + anti-PrP) (Figure 27D). In the

context of tumor proliferation in vitro, co-treatment of SNU-C5/WT with

anti-PrP antibody and 5FU significantly decreased the S phase of the cell

cycle (Figures 28A and B). In particular, the S phase after co-treatment with

anti-PrP antibody and 5FU was decreased in a 5FU dose-dependent manner

(Figure 28C). In comparison with the anti-proliferative effect of the anti-PrP

109
antibody and cetuximab, treatment with a high concentration of cetuximab (10

μg/mL) decreased the S phase of the cell cycle of SNU-C5/WT, com-pared

with that treated with 5FU, anti-PrP antibody, or low concentration of

cetuximab (1 μg/mL) (Figure 29A). Co-treatment with cetuximab, an anti-

PrP antibody, and 5FU showed the most suppressive effect on CRC

proliferation (Figure 29B). Furthermore, treatment of SNU-C5/WT with

anti-PrP antibody or cetuximab significantly decreased mitochondrial

oxidative phosphorylation, including mitochondrial basal respiration, ATP

turnover, and spare respiratory capacity, compared with the non-treatment

group (Figures 30A–E). In a SNU-C5/WT pretreated with H-5FUR-Exo

xenograft model, co-administration of 5FU and anti-PrP antibody

significantly reduced the expression of Ki-67 in tumor tissues (Figures 31A

and B). Conversely, the level of apoptotic marker cleaved caspase3 in tumor

tissues was significantly increased after co-treatment with 5FU and anti-PrP

(Figures 31C and D). In the H-5FUR-Exo-treated tumor tissues,

immunofluorescence staining of ZO-1 showed that the tight junctions of tumor

blood vessels decreased after co-administration of 5FU and anti-PrP

antibody (Figure 32A). These findings suggest that co-administration of 5FU

and anti-PrP antibody suppresses CRC progression by blocking PrPC.

110
111
Figure 27. Effect of co-administration of anti-PrP antibody and 5FU in a

murine xenograft model treated with exosomes secreted by hypoxic CRC cells.

(A) Schematic illustration of SNU-C5/WT cells pretreated with/without H-

5FUR-Exo in vivo subcutaneous transplantation and injection with PBS, 5FU,

5FU + anti-PrP antibody, and anti-PrP antibody. (B) Photographs of tumor

growth in a murine xenograft mouse model. (C) Quantification of tumor size

in each group (n=6). (D) ELISA analysis of PrPC expression in sera isolated

from a murine xenograft model (n=6).

112
Figure 28. Inhibitory effect of anti-PrP antibody with 5FU on proliferation of

CRC cells.

(A) Cell cycle analysis in SNU-C5/WT cells after treatment with 5FU, anti-

113
PrP antibody, or 5FU + anti-PrP antibody (n=3). (B) The graph shows S

phase levels of SNU-C5/WT after treatment with 5FU, anti-PrP antibody, or

5FU + anti-PrP antibody. (C) Cell cycle analysis in SNU-C5/5FUR after

treatment with 5FU (10, 50, and 100 μM), anti-PrP antibody, and anti-PrP

antibody + 5FU (10, 50, and 100 μM) (n=3). Data are presented as mean

± SEM. **p<0.01 vs. control; #p<0.05, ##p<0.01 (ANOVA).

114
115
Figure 29. Inhibitory effect of anti-PrP antibody with 5FU and cetuximab on

proliferation of CRC cells.

(A) Cell cycle analysis in SNU-C5/WT cells after treatment with 5FU, anti-

PrP antibody, or cetuximab (1 and 10 μg/mL) (n=3). (B) Cell cycle analysis

in SNU-C5/WT cells after treatment with 5FU, 5FU + anti-PrP antibody,

cetuximab + 5FU, cetuximab + anti-PrP antibody, and cetuximab + anti-PrP

+ 5FU (n=3).

116
Figure 30. Mitochondrial respiration in CRC cells after treatment with anti-PrP

antibody and cetuximab.

(A) OCR in SNU-C5/WT after treatment with anti-PrP antibody and

cetuximab (n=10). (B–E) Mitochondrial basal respiration (B), maximal

respiration (C), ATP turnover (D), and spare respiratory capacity (E) in

SNU-C5/WT after treatment with anti-PrP antibody and cetuximab (n=10).

Data are presented as mean ± SEM. *p<0.05, **p<0.01 vs. non-treatment

(ANOVA).

117
Figure 31. Effect of co-administration of anti-PrP antibody and 5FU in a

murine xenograft model treated with exosomes secreted by hypoxic CRC cells

on proliferation and apoptosis.

(A) Representative immunofluorescence staining analysis of Ki-67 (green)

in colorectal cancer tissues (n=3). Scale bar = 50 μm. (B) The graph shows

Ki-67-positive cells in tumor tissues (C) Representative

118
immunofluorescence staining analysis of cleaved caspase3 (red) in colorectal

cancer tissues (n=3). Scale bar = 50 μm. (D) The graph shows cleaved

caspase3-positive cells in tumor tissues. Data are presented as mean ± SEM.

*p<0.05, **p<0.01 (ANOVA).

119
120
Figure 32. Effect of anti-PrP antibody and 5FU on vascular permeability in vivo.

(A) In a mouse pre-injected with or without exosomes isolated from hypoxic

SNU-C5/5FUR, mice were injected with PBS, 5FU, 5FUR + anti-PrP

antibody, or anti-PrP antibody. Vascular permeability was assessed by

immunofluorescence staining of ZO-1 (green) and CD31 (red). Scale bar =

50 μm (n=3).

121
4. DISCUSSION

It has been shown that PrPC regulates proliferation, drug resistance,

metastasis, and CSC properties in various type of cancers including pancreatic,

breast, and colon cancers (157). Although exosomal PrPC is known to inhibit

amyloid beta-mediated neurotoxicity in Alzheimer’s disease (158), and

facilitate intercellular prion transmission in prion disease (159), studies on

the effect of exosomal PrPC on the tumor progression are limited. In this study,

I demonstrated that exosomal PrPC promotes proliferation, invasion and

migration of cancer cells. I found that exosomal PrPC inhibits migration and

invasion of vascular endothelial cells and increases permeability of blood

vessels. Therefore, PrP antibody treatment can reduce permeability of blood

vessels and inhibit metastatic CRC development by neutralizing exosomal PrP.

In addition, I also demonstrated that co-administration of anti-PrP antibody

with chemotherpy can improve cancer treatment efficacy.

This study showed that hypoxia induced the expression of PrPC in exosomes

secreted by drug-resistant CRC cells and that PrPC-expressing exosomes

promote CSC properties and tumor progression. PrPC is a highly ubiquitous

glycoprotein that affects the process of tumor progression, such as

proliferation, migration, invasion, metastasis, chemoresistance, and apoptosis

as well as stemness of cancer cells (22, 146, 160). Previous studies indicated

that PrPC promotes tumor metastasis, epithelial–mesenchymal transition, and

122
glucose metabolism through the regulation of Fyn, cytoskeletal regulatory

proteins (33, 36). PrPC also regulates multi-drug resistance via interaction

with CD44 (141, 157). Under hypoxic conditions, PrPC induces tumor

progression in CRC by targeting the heat shock protein 70 member 1-like

(HSPA1L)/HIF-1α/GP78 signal axis (38).

Hypoxia is a pathophysiological tumor microenvironment, which affects the

cell cycle, morphological conformation, energy metabolism, differentiation,

apoptosis, and autophagy (94, 161, 162). Under hypoxic conditions, cancer

cells regulate a wide range of gene expression in conjunction with the major

components of the hypoxia signaling pathway through expression of HIFs (74,

90). In patients with pancreatic tumors, the level of HIF-1α expression was

significantly increased (163). Among HIFs, the overexpression of HIF-1α

in response to hypoxia promotes tumor blood vessel formation,

aggressiveness, metastasis, and drug resistance. Hypoxia and HIF-1α

contribute to abnormal blood vessels, which are newly formed by

discontinuous endothelium and the blockage of lymphatic drainage, resulting

in the production of vascular hyperpermeability and increased permeation

(164, 165).

Recently, several studies have revealed that the crosstalk between tumor

cells and the microenvironment is a key factor for tumor progression through

extracellular vesicles and exosomes secreted by hypoxic cancer cells (70,

71). Hypoxia-induced exosomes released from cancer cells contain plasma

123
membrane receptors, including glucose transporter, EGFR, P-glycoprotein,

and multidrug resistance protein 1; angiogenic proteins, such as VEGF, FGF,

and angiogenin; and various noncoding RNAs, including miRNAs and lncRNAs

(162, 165, 166).

My study observed that targeted genes associated with CSC markers,

metastasis, angiogenesis, and oncogenes were significantly increased in

exosomes secreted from hypoxic 5FU-resistant CRC cells. In particular, the

expression of these genes in exosomes was regulated by the expression of

PrPC. These exosomes enhanced the cancer sphere formation, invasion,

migration, and proliferation in drug-resistant and CSCs in CRC. Notably,

exosomes secreted by hypoxic drug-resistant CRC cells are incorporated into

HUVECs and increase the migration, invasion, permeability, and production of

angiogenic cytokines.

In vivo study, these exosomes augmented the vascular hyperpermeability

through inhibition of tight junction protein ZO-1 expression. These effects

were blocked by silencing of PRNP. For stabilization of PrPC, My previous

study showed that HIF-1α-induced HSPA1L downregulated the expression

of GP78, a ubiquitinase for PrPC, resulting in the stabilization of PrPC under

hypoxic conditions (38). In lung cancer, exosomes secreted by hypoxic

cancer increased the expression of miR-23a, which inhibited the expression

of ZO-1, resulting in the induction of vascular permeability and cancer trans-

endothelial migration (95). Exosomes secreted by metastatic breast cancer

124
also destroyed endothelial ZO-1 expression and integrity through the

upregulation of miR-105 (154). This evidence indicates that exosomes

secreted by hypoxic tumor cells play important roles in the reduction in

endothelial integrity and tumor progression, suggesting that targeting of PrP-

expressing exosomes secreted by hypoxic tumors might be a novel strategy

for patients with CRC.

Clinically, antibody therapy for tumors provides the possibility for treating

patients with cancer in a targeted fashion by decreasing severe side effects,

compared with conventional chemotherapy (167). For the development of

advanced and highly therapeutically efficient antibody therapy in cancer

biology, discovery of specific molecular biomarkers in a wide range of solid

malignancies is a key process for beneficial therapeutic outcomes (167).

According to the unmet medical needs of patients with tumors, tumor

therapeutic antibodies, including trastuzumab (anti-human epidermal growth

factor receptor 2 [HER2] therapy) and cetuximab (anti-EGFR therapy), were

developed and clinically approved as treatments for a solid carcinoma and a

broad range of cancers, respectively (168, 169).

Cetuximab is a chimeric human mouse anti-EGFR monoclonal antibody and

it is used in combination with chemotherapy or as a single agent in metastatic

colon cancer and metastatic squamous cell head and neck cancer (170). EGFR

is overexpressed in most epithelial cell carcinomas such as colorectal cancer,

breast cancer, and lung cancer, and it is known that activation of EGFR

125
promotes cancer proliferation, angiogenesis, metastasis and inhibits apoptosis

(171). Cetuximab selectively binds to EGFR and competitively inhibits the

binding of EGF and other ligands, preventing its activation, eventually

inhibiting the growth of cancer cells and production of MMP and EGF and

inducing apoptosis (172).

However, intrinsic phenotypic variation and adaptive phenotypic

modifications in tumor cells can induce repeated exposure to sub-optimal

doses of the biotherapeutic, resulting in acquired resistance to monoclonal

antibody therapy (167, 173, 174). Antibody–drug conjugates (ADC) are

another option for treating tumors as a novel antibody-based therapeutic.

ADC consist of targeted antibody and anti-cancer drugs covalently attached

to the antibody, resulting in ADC reaching the tumor and killing the tumor.

Although ADC have the potential for this concept, the clinical outcomes are

limited because some chemical drugs can be released from the tumor and

diffuse into the surrounding cells (167, 175175).

In this study, I identified a novel CRC target, PrPC, and assessed the effect

of anti-PrP antibody on CRC by co-administration of 5FU. In vitro, anti-PrP

significantly inhibited proliferation and mitochondrial respiration. In exosome

non-treated murine xenograft model, the administration of anti-PrP antibody

significantly decreased the tumor size and serum PrPC concentration. This

effect was similar to the effect of cetuximab. In an H-5FUR-Exo-treated

xenograft model, co-administration of anti-PrP antibody and 5FU

126
significantly reduced the tumor size, PrPC expression, and tumor cell

proliferation. Furthermore, co-administration drastically increased the

number of apoptotic CRC cells in tumor tissues. These findings indicate that

co-administration of anti-PrP antibody and 5FU suppresses CRC tumor

progression, suggesting that anti-PrP antibody-based therapy could be a

novel and powerful strategy for clinical application in patients with CRC.

127
CONCLUSION

My findings reveal for the first time that 5FU-resistant colorectal cancer

cells express high levels of PrPC, and that PrPC facilitates anti-cancer drug

resistance by regulating survival, proliferation, and apoptosis signaling

pathways (Figure 33). Further studies are required to understand how the

expression of PrPC is induced during the acquisition of anti-cancer drug

resistance. This study may help clarify the physiological function of PrPC in

drug resistant colorectal cancer cells. The strategy of targeting PrPC may

enable novel molecular target combination chemotherapies. Furthermore, my

findings reveal that melatonin inhibits cell viability via excessive production

of superoxide and increased PrPC expression. In addition, silencing PrPC

enhances melatonin mediated accumulation of superoxide and activates ROS

mediated ER stress. Finally, apoptosis of CRC cancer cells is activated (Figure

34). This study demonstrated that the combination of melatonin and silencing

PrPC in SNU-C5/WT cells results in increased anticancer effects. These

findings indicate that understanding the effects of melatonin and PrPC on

colorectal cancer may help design a novel therapeutic strategy in cancer

treatment.

Next, these results indicate that exosomes secreted by hypoxic drug-

resistant CRC cells are key regulators of CRC progression (Figure 35). PrPC

is a pivotal messenger for regulating CRC behavior through the secretion of

128
exosomes by hypoxic tumors. Furthermore, my results suggest the possibility

of clinical application of anti-PrP antibody with anti-cancer drugs. Although

my results indicated that the administration of anti-PrP antibody did not have

an effect on several organs and tissues in a pre-clinical study, its side effects

and safety should be investigated prior to the application of this antibody in

patients with CRC. In conclusion, the co-administration of anti-PrP antibody

and anti-cancer drugs might be a potential therapeutic strategy for patients

with CRC through inhibition of exosomal PrPC expression and suppression of

CRC progression.

Taken together, these findings suggest that targeting PrPC might be a

potential therapeutic strategy for CRC therapy and induction of synergistic

effects with diverse anti-cancer reagents and could be an effective diagnostic

factor.

129
Figure 33. Schematic illustrating the protective mechanism of PrPC on 5FU in

5FU-resistant cancer cells.

In 5FU-resistant cancer cells, the expression level of PrPC is increased. In

the presence of 5FU, PrPC promotes cell survival, proliferation via activation

of the PI3K-AKT signaling pathway, and blockage of stress- and apoptosis-

mediated protein activation and reduction of apoptosis.

130
Figure 34. Schematic illustrating the mechanism of melatonin-mediated anti-

cancer effects in SNU-C5/WT CRC cells.

In colorectal cancer cells, melatonin induces the production of superoxide and

decreases the levels of PrPC. Silencing PrPC expression enhanced the

anticancer effects of melatonin via the production of mitochondrial superoxide,

activation of ER stress- and apoptosis- mediated proteins.

131
Figure 35. Schematic illustrating the effects of exosomes secreted by hypoxic

drug-resistant CRC cells on CRC progression and metastasis.

PrPC are overexpressed in cancers and related to cancer proliferation,

invasion, metastasis, and drug resistance. PrPC-expressing exosomes

induced the microenvironment of metastasis via increase of endothelial

permeability and angiogenic cytokine secretion. The treatment of anti-PrPC

and 5FU decreased the tumor progression.

132
REFERENCES

1. Siegel RL, Miller KD, Jemal A. 2019. Cancer statistics, 2019. CA Cancer J
Clin 69(1):7-34.
2. Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein
KD, Alteri R, Jemal A. 2016. Cancer treatment and survivorship statistics,
2016. CA Cancer J Clin 66(4):271-289.
3. Xie YH, Chen YX, Fang JY. 2020. Comprehensive review of targeted
therapy for colorectal cancer. Signal Transduct Target Ther 5(1):22.
4. Bellio H, Bertaut A, Hervieu A, Zanetta S, Hennequin A, Vincent J, Palmier
R, Bengrine-Lefevre L, Ghiringhelli F, Fumet JD. 2021. Phase I dose-
escalation trial of an innovative chemotherapy regimen combining a
fractionated dose of irinotecan plus bevacizumab, oxaliplatin, 5-
fluorouracil, and folinic Acid (bFOLFIRINOX-3) in chemorefractory
metastatic colorectal cancer. Cancers (Basel) 13(21):5472.
5. Jahani M, Azadbakht M, Norooznezhad F, Mansouri K. 2017. l-arginine
alters the effect of 5-fluorouracil on breast cancer cells in favor of
apoptosis. Biomed Pharmacother 88:114-123.
6. Yang H, Huang S, Wei Y, Cao S, Pi C, Feng T, Liang J, Zhao L, Ren G. 2017.
Curcumin enhances the anticancer effect of 5-fluorouracil against gastric
cancer through down-regulation of COX-2 and NF-kappaB signaling
pathways. J Cancer 8(18):3697-3706.
7. Longley DB, Harkin DP, Johnston PG. 2003. 5-fluorouracil: mechanisms
of action and clinical strategies. Nat Rev Cancer 3(5):330-338.
8. Noordhuis P, Holwerda U, Van der Wilt CL, Van Groeningen CJ, Smid K,
Meijer S, Pinedo HM, Peters GJ. 2004. 5-Fluorouracil incorporation into
RNA and DNA in relation to thymidylate synthase inhibition of human

133
colorectal cancers. Ann Oncol 15(7):1025-1032.
9. Kelland L. 2007. The resurgence of platinum-based cancer chemotherapy.
Nat Rev Cancer 7(8):573-584.
10. Vasan N, Baselga J, Hyman DM. 2019. A view on drug resistance in cancer.
Nature 575(7782):299-309.
11. Groenendijk FH, Bernards R. 2014. Drug resistance to targeted therapies:
deja vu all over again. Mol Oncol 8(6):1067-1083.
12. Lawson DA, Bhakta NR, Kessenbrock K, Prummel KD, Yu Y, Takai K, Zhou
A, Eyob H, Balakrishnan S, Wang CY, Yaswen P, Goga A, Werb Z. 2015.
Single-cell analysis reveals a stem-cell program in human metastatic
breast cancer cells. Nature 526(7571):131-135.
13. Marusyk A, Janiszewska M, Polyak K. 2020. Intratumor heterogeneity:
The rosetta stone of therapy resistance. Cancer Cell 37(4):471-484.
14. Li B, Jiang J, Assaraf YG, Xiao H, Chen ZS, Huang C. 2020. Surmounting
cancer drug resistance: New insights from the perspective of N(6)-
methyladenosine RNA modification. Drug Resist Updat 53:100720.
15. Housman G, Byler S, Heerboth S, Lapinska K, Longacre M, Snyder N,
Sarkar S. 2014. Drug resistance in cancer: an overview. Cancers (Basel)
6(3):1769-1792.
16. Hammond WA, Swaika A, Mody K. 2016. Pharmacologic resistance in
colorectal cancer: a review. Ther Adv Med Oncol 8(1):57-84.
17. Holohan C, Van Schaeybroeck S, Longley DB, Johnston PG. 2013. Cancer
drug resistance: An evolving paradigm. Nat Rev Cancer 13(10):714-726.
18. Xu K, Zhan Y, Yuan Z, Qiu Y, Wang H, Fan G, Wang J, Li W, Cao Y, Shen
X, Zhang J, Liang X, Yin P. 2019. Hypoxia induces drug resistance in
colorectal cancer through the HIF-1alpha/miR-338-5p/IL-6 feedback
loop. Mol Ther 27(10):1810-1824.
19. Liang J, Kong Q. 2012. alpha-Cleavage of cellular prion protein. Prion
6(5):453-460.

134
20. Linden R, Martins VR, Prado MA, Cammarota M, Izquierdo I, Brentani RR.
2008. Physiology of the prion protein. Physiol Rev 88(2):673-728.
21. Zomosa-Signoret V, Arnaud JD, Fontes P, Alvarez-Martinez MT, Liautard
JP. 2008. Physiological role of the cellular prion protein. Vet Res 39(4):9.
22. Martin-Lanneree S, Hirsch TZ, Hernandez-Rapp J, Halliez S, Vilotte JL,
Launay JM, Mouillet-Richard S. 2014. PrP(C) from stem cells to cancer.
Front Cell Dev Biol 2:55.
23. Lee JH, Han YS, Lee SH. 2017. Potentiation of biological effects of
mesenchymal stem cells in ischemic conditions by melatonin via
upregulation of cellular prion protein expression. J Pineal Res
62(2):e12385.
24. Lee JH, Yun CW, Han YS, Kim S, Jeong D, Kwon HY, Kim H, Baek MJ, Lee
SH. 2018. Melatonin and 5-fluorouracil co-suppress colon cancer stem
cells by regulating cellular prion protein-Oct4 axis. J Pineal Res
65(4):e12519.
25. Yoon YM, Lee JH, Yun SP, Han YS, Yun CW, Lee HJ, Noh H, Lee SJ, Han
HJ, Lee SH. 2016. Tauroursodeoxycholic acid reduces ER stress by
regulating of Akt-dependent cellular prion protein. Sci Rep 6:39838.
26. Tsenkova RN, Iordanova IK, Toyoda K, Brown DR. 2004. Prion protein
fate governed by metal binding. Biochem Biophys Res Commun
325(3):1005-1012.
27. Steele AD, Emsley JG, Ozdinler PH, Lindquist S, Macklis JD. 2006. Prion
protein (PrPc) positively regulates neural precursor proliferation during
developmental and adult mammalian neurogenesis. Proc Natl Acad Sci U S
A 103(9):3416-3421.
28. Bueler H, Fischer M, Lang Y, Bluethmann H, Lipp HP, DeArmond SJ,
Prusiner SB, Aguet M, Weissmann C. 1992. Normal development and
behaviour of mice lacking the neuronal cell-surface PrP protein. Nature
356(6370):577-582.

135
29. Mehrpour M, Codogno P. 2010. Prion protein: From physiology to cancer
biology. Cancer Lett 290(1):1-23.
30. Gil M, Kim YK, Kim KE, Kim W, Park CS, Lee KJ. 2016. Cellular prion
protein regulates invasion and migration of breast cancer cells through
MMP-9 activity. Biochem Biophys Res Commun 470(1):213-219.
31. Liang J, Pan Y, Zhang D, Guo C, Shi Y, Wang J, Chen Y, Wang X, Liu J, Guo
X, Chen Z, Qiao T, Fan D. 2007. Cellular prion protein promotes
proliferation and G1/S transition of human gastric cancer cells SGC7901
and AGS. FASEB J 21(9):2247-2256.
32. Yap YH, Say YH. 2012. Resistance against tumour necrosis factor alpha
apoptosis by the cellular prion protein is cell-specific for oral, colon and
kidney cancer cell lines. Cell Biol Int 36(3):273-277.
33. Li QQ, Sun YP, Ruan CP, Xu XY, Ge JH, He J, Xu ZD, Wang Q, Gao WC.
2011. Cellular prion protein promotes glucose uptake through the Fyn-
HIF-2alpha-Glut1 pathway to support colorectal cancer cell survival.
Cancer Sci 102(2):400-406.
34. Du L, Rao G, Wang H, Li B, Tian W, Cui J, He L, Laffin B, Tian X, Hao C,
Liu H, Sun X, Zhu Y, Tang DG, Mehrpour M, Lu Y, Chen Q. 2013. CD44-
positive cancer stem cells expressing cellular prion protein contribute to
metastatic capacity in colorectal cancer. Cancer Res 73(8):2682-2694.
35. de Lacerda TC, Costa-Silva B, Giudice FS, Dias MV, de Oliveira GP,
Teixeira BL, Dos Santos TG, Martins VR. 2016. Prion protein binding to
HOP modulates the migration and invasion of colorectal cancer cells. Clin
Exp Metastasis 33(5):441-451.
36. Wang Q, Qian J, Wang F, Ma Z. 2012. Cellular prion protein accelerates
colorectal cancer metastasis via the Fyn-SP1-SATB1 axis. Oncol Rep
28(6):2029-2034.
37. Le Corre D, Ghazi A, Balogoun R, Pilati C, Aparicio T, Martin-Lanneree S,
Marisa L, Djouadi F, Poindessous V, Crozet C, Emile JF, Mulot C, Le

136
Malicot K, Boige V, Blons H, de Reynies A, Taieb J, Ghiringhelli F,
Bennouna J, Launay JM, Laurent-Puig P, Mouillet-Richard S. 2019. The
cellular prion protein controls the mesenchymal-like molecular subtype
and predicts disease outcome in colorectal cancer. EBioMedicine 46:94-
104.
38. Lee JH, Han YS, Yoon YM, Yun CW, Yun SP, Kim SM, Kwon HY, Jeong D,
Baek MJ, Lee HJ, Lee SJ, Han HJ, Lee SH. 2017. Role of HSPA1L as a
cellular prion protein stabilizer in tumor progression via HIF-1alpha/GP78
axis. Oncogene 36(47):6555-6567.
39. Atkinson CJ, Kawamata F, Liu C, Ham S, Gyorffy B, Munn AL, Wei MQ,
Moller A, Whitehall V, Wiegmans AP. 2019. EGFR and prion protein
promote signaling via FOXO3a-KLF5 resulting in clinical resistance to
platinum agents in colorectal cancer. Mol Oncol 13(4):725-737.
40. Pignon JP, Bourhis J, Domenge C, Designe L. 2000. Chemotherapy added
to locoregional treatment for head and neck squamous-cell carcinoma:
three meta-analyses of updated individual data. MACH-NC collaborative
group. Meta-analysis of chemotherapy on head and neck cancer. Lancet
355(9208):949-955.
41. Adelstein DJ, Saxton JP, Rybicki LA, Esclamado RM, Wood BG, Strome M,
Lavertu P, Lorenz RR, Carroll MA. 2006. Multiagent concurrent
chemoradiotherapy for locoregionally advanced squamous cell head and
neck cancer: mature results from a single institution. J Clin Oncol
24(7):1064-1071.
42. Tsukuda M, Ishitoya J, Matsuda H, Horiuchi C, Taguchi T, Takahashi M,
Nishimura G, Kawakami M, Watanabe M, Niho T, Kawano T, Ikeda Y,
Sakuma Y, Shiono O, Komatsu M. 2010. Randomized controlled phase II
comparison study of concurrent chemoradiotherapy with docetaxel,
cisplatin, and 5-fluorouracil versus CCRT with cisplatin, 5-fluorouracil,
methotrexate and leucovorin in patients with locally advanced squamous

137
cell carcinoma of the head and neck. Cancer Chemother Pharmacol
66(4):729-736.
43. Zhang N, Yin Y, Xu SJ, Chen WS. 2008. 5-Fluorouracil: Mechanisms of
resistance and reversal strategies. Molecules 13(8):1551-1569.
44. Wilson PM, Danenberg PV, Johnston PG, Lenz HJ, Ladner RD. 2014.
Standing the test of time: Targeting thymidylate biosynthesis in cancer
therapy. Nat Rev Clin Oncol 11(5):282-298.
45. Lenz HJ, Hayashi K, Salonga D, Danenberg KD, Danenberg PV, Metzger R,
Banerjee D, Bertino JR, Groshen S, Leichman LP, Leichman CG. 1998. p53
point mutations and thymidylate synthase messenger RNA levels in
disseminated colorectal cancer: an analysis of response and survival. Clin
Cancer Res 4(5):1243-1250.
46. Edler D, Blomgren H, Allegra CJ, Johnston PG, Lagerstedt U, Magnusson
I, Ragnhammar P. 1997. Immunohistochemical determination of
thymidylate synthase in colorectal cancer-methodological studies. Eur J
Cancer 33(13):2278-2281.
47. Abdallah EA, Fanelli MF, Buim ME, Machado Netto MC, Gasparini Junior
JL, Souza ESV, Dettino AL, Mingues NB, Romero JV, Ocea LM, Rocha BM,
Alves VS, Araujo DV, Chinen LT. 2015. Thymidylate synthase expression
in circulating tumor cells: A new tool to predict 5-fluorouracil resistance
in metastatic colorectal cancer patients. Int J Cancer 137(6):1397-1405.
48. Tabung FK, Brown LS, Fung TT. 2017. Dietary patterns and colorectal
cancer risk: A review of 17 years of evidence (2000-2016). Curr
Colorectal Cancer Rep 13(6):440-454.
49. Romaguera D, Vergnaud AC, Peeters PH, van Gils CH, Chan DS, Ferrari P,
Romieu I, Jenab M, Slimani N, Clavel-Chapelon F, Fagherazzi G, Perquier
F, Kaaks R, Teucher B, Boeing H, von Rusten A, Tjonneland A, Olsen A,
Dahm CC, Overvad K, Quiros JR, Gonzalez CA, Sanchez MJ, Navarro C,
Barricarte A, Dorronsoro M, Khaw KT, Wareham NJ, Crowe FL, Key TJ,

138
Trichopoulou A, Lagiou P, Bamia C, Masala G, Vineis P, Tumino R, Sieri S,
Panico S, May AM, Bueno-de-Mesquita HB, Buchner FL, Wirfalt E,
Manjer J, Johansson I, Hallmans G, Skeie G, Benjaminsen Borch K, Parr
CL, Riboli E, Norat T. 2012. Is concordance with world cancer research
fund/american institute for cancer research guidelines for cancer
prevention related to subsequent risk of cancer? Results from the EPIC
study. Am J Clin Nutr 96(1):150-163.
50. Hardeland R, Pandi-Perumal SR, Cardinali DP. 2006. Melatonin. Int J
Biochem Cell Biol 38(3):313-316.
51. Mills E, Wu P, Seely D, Guyatt G. 2005. Melatonin in the treatment of
cancer: a systematic review of randomized controlled trials and meta-
analysis. J Pineal Res 39(4):360-366.
52. Hong Y, Won J, Lee Y, Lee S, Park K, Chang KT, Hong Y. 2014. Melatonin
treatment induces interplay of apoptosis, autophagy, and senescence in
human colorectal cancer cells. J Pineal Res 56(3):264-274.
53. Moloudizargari M, Moradkhani F, Hekmatirad S, Fallah M, Asghari MH,
Reiter RJ. 2021. Therapeutic targets of cancer drugs: Modulation by
melatonin. Life Sci 267:118934.
54. Liu Z, Zou D, Yang X, Xue X, Zuo L, Zhou Q, Hu R, Wang Y. 2017. Melatonin
inhibits colon cancer RKO cell migration by downregulating Rho-
associated protein kinase expression via the p38/MAPK signaling pathway.
Mol Med Rep 16(6):9383-9392.
55. Gao Y, Xiao X, Zhang C, Yu W, Guo W, Zhang Z, Li Z, Feng X, Hao J, Zhang
K, Xiao B, Chen M, Huang W, Xiong S, Wu X, Deng W. 2017. Melatonin
synergizes the chemotherapeutic effect of 5-fluorouracil in colon cancer
by suppressing PI3K/AKT and NF-kappaB/iNOS signaling pathways. J
Pineal Res 62(2):e12380.
56. Batista AP, da Silva TG, Teixeira AA, de Medeiros PL, Teixeira VW, Alves
LC, Dos Santos FA, Silva EC. 2014. Ultrastructural aspects of melatonin

139
cytotoxicity on Caco-2 cells in vitro. Micron 59:17-23.
57. Buldak RJ, Pilc-Gumula K, Buldak L, Witkowska D, Kukla M, Polaniak R,
Zwirska-Korczala K. 2015. Effects of ghrelin, leptin and melatonin on the
levels of reactive oxygen species, antioxidant enzyme activity and viability
of the HCT 116 human colorectal carcinoma cell line. Mol Med Rep
12(2):2275-2282.
58. Li Y, Li S, Zhou Y, Meng X, Zhang JJ, Xu DP, Li HB. 2017. Melatonin for
the prevention and treatment of cancer. Oncotarget 8(24):39896-39921.
59. Kang T, Atukorala I, Mathivanan S. 2021. Biogenesis of Extracellular
Vesicles. Subcell Biochem 97:19-43.
60. Taylor DD, Gercel-Taylor C. 2013. The origin, function, and diagnostic
potential of RNA within extracellular vesicles present in human biological
fluids. Front Genet 4:142.
61. Sun Y, Zheng W, Guo Z, Ju Q, Zhu L, Gao J, Zhou L, Liu F, Xu Y, Zhan Q,
Zhou Z, Sun W, Zhao X. 2016. A novel TP53 pathway influences the HGS-
mediated exosome formation in colorectal cancer. Sci Rep 6:28083.
62. Devhare PB, Ray RB. 2018. Extracellular vesicles: Novel mediator for cell
to cell communications in liver pathogenesis. Mol Aspects Med 60:115-
122.
63. Doyle LM, Wang MZ. 2019. Overview of extracellular vesicles, their origin,
composition, purpose, and methods for exosome isolation and analysis.
Cells 8(7):727.
64. Borges FT, Reis LA, Schor N. 2013. Extracellular vesicles: structure,
function, and potential clinical uses in renal diseases. Braz J Med Biol Res
46(10):824-830.
65. Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B, Zheng
Y, Hoshino A, Brazier H, Xiang J, Williams C, Rodriguez-Barrueco R, Silva
JM, Zhang W, Hearn S, Elemento O, Paknejad N, Manova-Todorova K,
Welte K, Bromberg J, Peinado H, Lyden D. 2014. Double-stranded DNA

140
in exosomes: A novel biomarker in cancer detection. Cell Res 24(6):766-
769.
66. Kalra H, Drummen GP, Mathivanan S. 2016. Focus on extracellular
vesicles: Introducing the next small big thing. Int J Mol Sci 17(2):170.
67. Aslan C, Maralbashi S, Salari F, Kahroba H, Sigaroodi F, Kazemi T,
Kharaziha P. 2019. Tumor-derived exosomes: Implication in angiogenesis
and antiangiogenesis cancer therapy. J Cell Physiol 234(10):16885-
16903.
68. Al-Sowayan BS, Al-Shareeda AT, Al-Hujaily EM. 2019. Exosomes,
cancer's little army. Stem Cell Investig 6:9.
69. Costa-Silva B, Aiello NM, Ocean AJ, Singh S, Zhang H, Thakur BK, Becker
A, Hoshino A, Mark MT, Molina H, Xiang J, Zhang T, Theilen TM, Garcia-
Santos G, Williams C, Ararso Y, Huang Y, Rodrigues G, Shen TL, Labori
KJ, Lothe IM, Kure EH, Hernandez J, Doussot A, Ebbesen SH, Grandgenett
PM, Hollingsworth MA, Jain M, Mallya K, Batra SK, Jarnagin WR, Schwartz
RE, Matei I, Peinado H, Stanger BZ, Bromberg J, Lyden D. 2015. Pancreatic
cancer exosomes initiate pre-metastatic niche formation in the liver. Nat
Cell Biol 17(6):816-826.
70. M HR, Bayraktar E, G KH, Abd-Ellah MF, Amero P, Chavez-Reyes A,
Rodriguez-Aguayo C. 2017. Exosomes: From garbage bins to promising
therapeutic targets. Int J Mol Sci 18(3):538.
71. Roberts WM, Jenkins JJ, Moorhead EL, 2nd, Douglass EC. 1988.
Incontinentia pigmenti, a chromosomal instability syndrome, is associated
with childhood malignancy. Cancer 62(11):2370-2372.
72. Semenza GL. 2010. Oxygen homeostasis. Wiley Interdiscip Rev Syst Biol
Med 2(3):336-361.
73. Harris AL. 2002. Hypoxia-a key regulatory factor in tumour growth. Nat
Rev Cancer 2(1):38-47.
74. Semenza GL. 2012. Hypoxia-inducible factors in physiology and medicine.

141
Cell 148(3):399-408.
75. Munoz-Pinedo C, El Mjiyad N, Ricci JE. 2012. Cancer metabolism: Current
perspectives and future directions. Cell Death Dis 3:e248.
76. Vaupel P, Mayer A. 2007. Hypoxia in cancer: significance and impact on
clinical outcome. Cancer Metastasis Rev 26(2):225-239.
77. Vadde R, Vemula S, Jinka R, Merchant N, Bramhachari PV, Nagaraju GP.
2017. Role of hypoxia-inducible factors (HIF) in the maintenance of
stemness and malignancy of colorectal cancer. Crit Rev Oncol Hematol
113:22-27.
78. Nagaraju GP, Bramhachari PV, Raghu G, El-Rayes BF. 2015. Hypoxia
inducible factor-1alpha: Its role in colorectal carcinogenesis and
metastasis. Cancer Lett 366(1):11-18.
79. Gordan JD, Thompson CB, Simon MC. 2007. HIF and c-Myc: Sibling rivals
for control of cancer cell metabolism and proliferation. Cancer Cell
12(2):108-113.
80. Algire GH, Chalkley HW, Earle WE, Legallais FY, Park HD, Shelton E,
Schilling EL. 1950. Vascular reactions of normal and malignant tissues in
vivo. III. Vascular reactions' of mice to fibroblasts treated in vitro with
methylcholanthrene. J Natl Cancer Inst 11(3):555-580.
81. Saclarides TJ. 1997. Angiogenesis in colorectal cancer. Surg Clin North
Am 77(1):253-260.
82. Giordano G, Febbraro A, Venditti M, Campidoglio S, Olivieri N, Raieta K,
Parcesepe P, Imbriani GC, Remo A, Pancione M. 2014. Targeting
angiogenesis and tumor microenvironment in metastatic colorectal cancer:
Role of aflibercept. Gastroenterol Res Pract 2014:526178.
83. Yun Z, Lin Q. 2014. Hypoxia and regulation of cancer cell stemness. Adv
Exp Med Biol 772:41-53.
84. Santoyo-Ramos P, Likhatcheva M, Garcia-Zepeda EA, Castaneda-Patlan
MC, Robles-Flores M. 2014. Hypoxia-inducible factors modulate the

142
stemness and malignancy of colon cancer cells by playing opposite roles
in canonical Wnt signaling. PLoS One 9(11):e112580.
85. Lobo NA, Shimono Y, Qian D, Clarke MF. 2007. The biology of cancer stem
cells. Annu Rev Cell Dev Biol 23:675-699.
86. Simon MC, Keith B. 2008. The role of oxygen availability in embryonic
development and stem cell function. Nat Rev Mol Cell Biol 9(4):285-296.
87. Ding Z, Yang L, Xie X, Xie F, Pan F, Li J, He J, Liang H. 2010. Expression
and significance of hypoxia-inducible factor-1alpha and MDR1/P-
glycoprotein in human colon carcinoma tissue and cells. J Cancer Res Clin
Oncol 136(11):1697-1707.
88. Chen J, Ding Z, Peng Y, Pan F, Li J, Zou L, Zhang Y, Liang H. 2014. HIF-
1alpha inhibition reverses multidrug resistance in colon cancer cells via
downregulation of MDR1/P-glycoprotein. PLoS One 9(6):e98882.
89. Lu X, Kang Y. 2010. Hypoxia and hypoxia-inducible factors: Master
regulators of metastasis. Clin Cancer Res 16(24):5928-5935.
90. Shao C, Yang F, Miao S, Liu W, Wang C, Shu Y, Shen H. 2018. Role of
hypoxia-induced exosomes in tumor biology. Mol Cancer 17(1):120.
91. Walbrecq G, Lecha O, Gaigneaux A, Fougeras MR, Philippidou D, Margue
C, Tetsi Nomigni M, Bernardin F, Dittmar G, Behrmann I, Kreis S. 2020.
Hypoxia-induced adaptations of miRNomes and proteomes in melanoma
cells and their secreted extracellular vesicles. Cancers (Basel) 12(3):692.
92. Meng W, Hao Y, He C, Li L, Zhu G. 2019. Exosome-orchestrated hypoxic
tumor microenvironment. Mol Cancer 18(1):57.
93. Kumar A, Deep G. 2020. Exosomes in hypoxia-induced remodeling of the
tumor microenvironment. Cancer Lett 488:1-8.
94. Umezu T, Tadokoro H, Azuma K, Yoshizawa S, Ohyashiki K, Ohyashiki JH.
2014. Exosomal miR-135b shed from hypoxic multiple myeloma cells
enhances angiogenesis by targeting factor-inhibiting HIF-1. Blood
124(25):3748-3757.

143
95. Hsu YL, Hung JY, Chang WA, Lin YS, Pan YC, Tsai PH, Wu CY, Kuo PL.
2017. Hypoxic lung cancer-secreted exosomal miR-23a increased
angiogenesis and vascular permeability by targeting prolyl hydroxylase
and tight junction protein ZO-1. Oncogene 36(34):4929-4942.
96. Ramteke A, Ting H, Agarwal C, Mateen S, Somasagara R, Hussain A,
Graner M, Frederick B, Agarwal R, Deep G. 2015. Exosomes secreted
under hypoxia enhance invasiveness and stemness of prostate cancer cells
by targeting adherens junction molecules. Mol Carcinog 54(7):554-565.
97. Chen X, Ying X, Wang X, Wu X, Zhu Q, Wang X. 2017. Exosomes derived
from hypoxic epithelial ovarian cancer deliver microRNA-940 to induce
macrophage M2 polarization. Oncol Rep 38(1):522-528.
98. Yun CW, Yun S, Lee JH, Han YS, Yoon YM, An D, Lee SH. 2016. Silencing
prion protein in HT29 human colorectal cancer cells enhances anticancer
response to fucoidan. Anticancer Res 36(9):4449-4458.
99. Lee JH, Yoon YM, Han YS, Yun CW, Lee SH. 2018. Melatonin promotes
apoptosis of oxaliplatin-resistant colorectal cancer cells through inhibition
of cellular prion protein. Anticancer Res 38(4):1993-2000.
100. Siegel RL, Miller KD, Jemal A. 2017. Cancer statistics, 2017. CA Cancer J
Clin 67(1):7-30.
101. Oliphant R, Nicholson GA, Horgan PG, Molloy RG, McMillan DC, Morrison
DS, West of Scotland Colorectal Cancer Managed Clinical N. 2013.
Deprivation and colorectal cancer surgery: Longer-term survival
inequalities are due to differential postoperative mortality between
socioeconomic groups. Ann Surg Oncol 20(7):2132-2139.
102. Jung GR, Kim KJ, Choi CH, Lee TB, Han SI, Han HK, Lim SC. 2007. Effect
of betulinic acid on anticancer drug-resistant colon cancer cells. Basic Clin
Pharmacol Toxicol 101(4):277-285.
103. Bu P, Chen KY, Chen JH, Wang L, Walters J, Shin YJ, Goerger JP, Sun J,
Witherspoon M, Rakhilin N, Li J, Yang H, Milsom J, Lee S, Zipfel W, Jin

144
MM, Gumus ZH, Lipkin SM, Shen X. 2013. A microRNA miR-34a-
regulated bimodal switch targets Notch in colon cancer stem cells. Cell
Stem Cell 12(5):602-615.
104. Basu S, Mohan ML, Luo X, Kundu B, Kong Q, Singh N. 2007. Modulation of
proteinase K-resistant prion protein in cells and infectious brain
homogenate by redox iron: Implications for prion replication and disease
pathogenesis. Mol Biol Cell 18(9):3302-3312.
105. Yang X, Zhang Y, Zhang L, He T, Zhang J, Li C. 2014. Prion protein and
cancers. Acta Biochim Biophys Sin (Shanghai) 46(6):431-440.
106. Liang J, Ge F, Guo C, Luo G, Wang X, Han G, Zhang D, Wang J, Li K, Pan
Y, Yao L, Yin Z, Guo X, Wu K, Ding J, Fan D. 2009. Inhibition of PI3K/Akt
partially leads to the inhibition of PrP(C)-induced drug resistance in
gastric cancer cells. FEBS J 276(3):685-694.
107. Cheng Y, Tao L, Xu J, Li Q, Yu J, Jin Y, Chen Q, Xu Z, Zou Q, Liu X. 2014.
CD44/cellular prion protein interact in multidrug resistant breast cancer
cells and correlate with responses to neoadjuvant chemotherapy in breast
cancer patients. Mol Carcinog 53(9):686-697.
108. Brown DR, Schulz-Schaeffer WJ, Schmidt B, Kretzschmar HA. 1997. Prion
protein-deficient cells show altered response to oxidative stress due to
decreased SOD-1 activity. Exp Neurol 146(1):104-112.
109. McLennan NF, Brennan PM, McNeill A, Davies I, Fotheringham A,
Rennison KA, Ritchie D, Brannan F, Head MW, Ironside JW, Williams A,
Bell JE. 2004. Prion protein accumulation and neuroprotection in hypoxic
brain damage. Am J Pathol 165(1):227-235.
110. Malaga-Trillo E, Solis GP, Schrock Y, Geiss C, Luncz L, Thomanetz V,
Stuermer CA. 2009. Regulation of embryonic cell adhesion by the prion
protein. PLoS Biol 7(3):e55.
111. Wang JH, Du JP, Zhang YH, Zhao XJ, Fan RY, Wang ZH, Wu ZT, Han Y.
2011. Dynamic changes and surveillance function of prion protein

145
expression in gastric cancer drug resistance. World J Gastroenterol
17(35):3986-3993.
112. McEwan JF, Windsor ML, Cullis-Hill SD. 2009. Antibodies to prion protein
inhibit human colon cancer cell growth. Tumour Biol 30(3):141-147.
113. Vanhaesebroeck B, Guillermet-Guibert J, Graupera M, Bilanges B. 2010.
The emerging mechanisms of isoform-specific PI3K signalling. Nat Rev
Mol Cell Biol 11(5):329-341.
114. Yuan TL, Cantley LC. 2008. PI3K pathway alterations in cancer: Variations
on a theme. Oncogene 27(41):5497-5510.
115. Liu LZ, Zhou XD, Qian G, Shi X, Fang J, Jiang BH. 2007. AKT1 amplification
regulates cisplatin resistance in human lung cancer cells through the
mammalian target of rapamycin/p70S6K1 pathway. Cancer Res
67(13):6325-6332.
116. Manning BD, Cantley LC. 2007. AKT/PKB signaling: Navigating
downstream. Cell 129(7):1261-1274.
117. Vassallo N, Herms J, Behrens C, Krebs B, Saeki K, Onodera T, Windl O,
Kretzschmar HA. 2005. Activation of phosphatidylinositol 3-kinase by
cellular prion protein and its role in cell survival. Biochem Biophys Res
Commun 332(1):75-82.
118. Weise J, Sandau R, Schwarting S, Crome O, Wrede A, Schulz-Schaeffer
W, Zerr I, Bahr M. 2006. Deletion of cellular prion protein results in
reduced Akt activation, enhanced postischemic caspase-3 activation, and
exacerbation of ischemic brain injury. Stroke 37(5):1296-1300.
119. Park JY, Jeong JK, Lee JH, Moon JH, Kim SW, Lee YJ, Park SY. 2015.
Induction of cellular prion protein (PrPc) under hypoxia inhibits apoptosis
caused by TRAIL treatment. Oncotarget 6(7):5342-5353.
120. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. 2015.
Global cancer statistics, 2012. CA Cancer J Clin 65(2):87-108.
121. Sun HY, Lee JH, Han YS, Yoon YM, Yun CW, Kim JH, Song YS, Lee SH.

146
2016. Pivotal roles of ginsenoside Rg3 in tumor apoptosis through
regulation of reactive oxygen species. Anticancer Res 36(9):4647-4654.
122. Song J, Ma SJ, Luo JH, Zhang H, Wang RX, Liu H, Li L, Zhang ZG, Zhou
RX. 2018. Melatonin induces the apoptosis and inhibits the proliferation of
human gastric cancer cells via blockade of the AKT/MDM2 pathway. Oncol
Rep 39(4):1975-1983.
123. Alonso-Gonzalez C, Mediavilla D, Martinez-Campa C, Gonzalez A, Cos S,
Sanchez-Barcelo EJ. 2008. Melatonin modulates the cadmium-induced
expression of MT-2 and MT-1 metallothioneins in three lines of human
tumor cells (MCF-7, MDA-MB-231 and HeLa). Toxicol Lett
181(3):190-195.
124. Lee JH, Yun CW, Lee SH. 2018. Cellular prion protein enhances drug
resistance of colorectal cancer cells via regulation of a survival signal
pathway. Biomol Ther (Seoul) 26(3):313-321.
125. Cha JA, Song HS, Kang B, Park MN, Park KS, Kim SH, Shim BS, Kim B.
2018. miR-211 plays a critical role in cnidium officinale makino extract-
induced, ROS/ER Stress-mediated apoptosis in U937 and U266 cells. Int
J Mol Sci 19(3):865.
126. Narendra D, Walker JE, Youle R. 2012. Mitochondrial quality control
mediated by PINK1 and Parkin: Links to parkinsonism. Cold Spring Harb
Perspect Biol 4(11):a011338.
127. Youle RJ, van der Bliek AM. 2012. Mitochondrial fission, fusion, and stress.
Science 337(6098):1062-1065.
128. O'Flanagan CH, Morais VA, Wurst W, De Strooper B, O'Neill C. 2015. The
Parkinson's gene PINK1 regulates cell cycle progression and promotes
cancer-associated phenotypes. Oncogene 34(11):1363-1374.
129. Shen YQ, Guerra-Librero A, Fernandez-Gil BI, Florido J, Garcia-Lopez
S, Martinez-Ruiz L, Mendivil-Perez M, Soto-Mercado V, Acuna-
Castroviejo D, Ortega-Arellano H, Carriel V, Diaz-Casado ME, Reiter RJ,

147
Rusanova I, Nieto A, Lopez LC, Escames G. 2018. Combination of
melatonin and rapamycin for head and neck cancer therapy: Suppression
of AKT/mTOR pathway activation, and activation of mitophagy and
apoptosis via mitochondrial function regulation. J Pineal Res 64(3):e12461.
130. Leon J, Casado J, Jimenez Ruiz SM, Zurita MS, Gonzalez-Puga C, Rejon
JD, Gila A, Munoz de Rueda P, Pavon EJ, Reiter RJ, Ruiz-Extremera A,
Salmeron J. 2014. Melatonin reduces endothelin-1 expression and
secretion in colon cancer cells through the inactivation of FoxO-1 and
NF-kappaB. J Pineal Res 56(4):415-426.
131. Kim J, Kim J, Bae JS. 2016. ROS homeostasis and metabolism: A critical
liaison for cancer therapy. Exp Mol Med 48(11):e269.
132. Kumari S, Badana AK, G MM, G S, Malla R. 2018. Reactive oxygen species:
A key constituent in cancer survival. Biomark Insights
13:1177271918755391.
133. Vakifahmetoglu-Norberg H, Ouchida AT, Norberg E. 2017. The role of
mitochondria in metabolism and cell death. Biochem Biophys Res Commun
482(3):426-431.
134. Kumar S, Stokes J, 3rd, Singh UP, Scissum-Gunn K, Singh R, Manne U,
Mishra MK. 2017. Prolonged exposure of resveratrol induces reactive
superoxide species-independent apoptosis in murine prostate cells.
Tumour Biol 39(10):1010428317715039.
135. Zou Z, Chang H, Li H, Wang S. 2017. Induction of reactive oxygen species:
an emerging approach for cancer therapy. Apoptosis 22(11):1321-1335.
136. Brown DR, Nicholas RS, Canevari L. 2002. Lack of prion protein
expression results in a neuronal phenotype sensitive to stress. J Neurosci
Res 67(2):211-224.
137. Klamt F, Dal-Pizzol F, Conte da Frota ML, Jr., Walz R, Andrades ME, da
Silva EG, Brentani RR, Izquierdo I, Fonseca Moreira JC. 2001. Imbalance
of antioxidant defense in mice lacking cellular prion protein. Free Radic

148
Biol Med 30(10):1137-1144.
138. Prieto-Dominguez N, Ordonez R, Fernandez A, Mendez-Blanco C, Baulies
A, Garcia-Ruiz C, Fernandez-Checa JC, Mauriz JL, Gonzalez-Gallego J.
2016. Melatonin-induced increase in sensitivity of human hepatocellular
carcinoma cells to sorafenib is associated with reactive oxygen species
production and mitophagy. J Pineal Res 61(3):396-407.
139. Malaise M, Schatzl HM, Burkle A. 2008. The octarepeat region of prion
protein, but not the TM1 domain, is important for the antioxidant effect of
prion protein. Free Radic Biol Med 45(12):1622-1630.
140. Ma WW, Zhao L, Yuan LH, Yu HL, Wang H, Gong XY, Wei F, Xiao R. 2017.
Elaidic acid induces cell apoptosis through induction of ROS accumulation
and endoplasmic reticulum stress in SHSY5Y cells. Mol Med Rep
16(6):9337-9346.
141. Roberts JL, Poklepovic A, Booth L. 2017. Curcumin interacts with
sildenafil to kill GI tumor cells via endoplasmic reticulum stress and
reactive oxygen/ nitrogen species. Oncotarget 8(59):99451-99469.
142. Kuipers EJ, Grady WM, Lieberman D, Seufferlein T, Sung JJ, Boelens PG,
van de Velde CJ, Watanabe T. 2015. Colorectal cancer. Nat Rev Dis
Primers 1:15065.
143. Douillard JY, Cunningham D, Roth AD, Navarro M, James RD, Karasek P,
Jandik P, Iveson T, Carmichael J, Alakl M, Gruia G, Awad L, Rougier P.
2000. Irinotecan combined with fluorouracil compared with fluorouracil
alone as first-line treatment for metastatic colorectal cancer: a
multicentre randomised trial. Lancet 355(9209):1041-1047.
144. Van der Jeught K, Xu HC, Li YJ, Lu XB, Ji G. 2018. Drug resistance and
new therapies in colorectal cancer. World J Gastroenterol 24(34):3834-
3848.
145. Prusiner SB. 1982. Novel proteinaceous infectious particles cause scrapie.
Science 216(4542):136-144.

149
146. Santos TG, Lopes MH, Martins VR. 2015. Targeting prion protein
interactions in cancer. Prion 9(3):165-173.
147. Appleby SL, Cockshell MP, Pippal JB, Thompson EJ, Barrett JM, Tooley
K, Sen S, Sun WY, Grose R, Nicholson I, Levina V, Cooke I, Talbo G, Lopez
AF, Bonder CS. 2012. Characterization of a distinct population of
circulating human non-adherent endothelial forming cells and their
recruitment via intercellular adhesion molecule-3. PLoS One
7(11):e46996.
148. Samaeekia R, Rabiee B, Putra I, Shen X, Park YJ, Hematti P, Eslani M,
Djalilian AR. 2018. Effect of human corneal mesenchymal stromal cell-
derived exosomes on corneal epithelial wound healing. Invest Ophthalmol
Vis Sci 59(12):5194-5200.
149. Go G, Lee J, Choi DS, Kim SS, Gho YS. 2019. Extracellular vesicle-
mimetic ghost nanovesicles for delivering anti-inflammatory drugs to
mitigate gram-negative bacterial outer membrane vesicle-induced
systemic inflammatory response syndrome. Adv Healthc Mater
8(4):e1801082.
150. Thery C, Amigorena S, Raposo G, Clayton A. 2006. Isolation and
characterization of exosomes from cell culture supernatants and biological
fluids. Curr Protoc Cell Biol Chapter 3:Unit 3 22.
151. Komaki M, Numata Y, Morioka C, Honda I, Tooi M, Yokoyama N, Ayame
H, Iwasaki K, Taki A, Oshima N, Morita I. 2017. Exosomes of human
placenta-derived mesenchymal stem cells stimulate angiogenesis. Stem
Cell Res Ther 8(1):219.
152. Wierzbicki M, Sawosz E, Strojny B, Jaworski S, Grodzik M, Chwalibog A.
2018. NF-kappaB-related decrease of glioma angiogenic potential by
graphite nanoparticles and graphene oxide nanoplatelets. Sci Rep
8(1):14733.
153. Bousquet PF, Brana MF, Conlon D, Fitzgerald KM, Perron D, Cocchiaro C,

150
Miller R, Moran M, George J, Qian XD, et al. 1995. Preclinical evaluation
of LU 79553: A novel bis-naphthalimide with potent antitumor activity.
Cancer Res 55(5):1176-1180.
154. Zhou W, Fong MY, Min Y, Somlo G, Liu L, Palomares MR, Yu Y, Chow A,
O'Connor ST, Chin AR, Yen Y, Wang Y, Marcusson EG, Chu P, Wu J, Wu X,
Li AX, Li Z, Gao H, Ren X, Boldin MP, Lin PC, Wang SE. 2014. Cancer-
secreted miR-105 destroys vascular endothelial barriers to promote
metastasis. Cancer Cell 25(4):501-515.
155. Wilson WR, Hay MP. 2011. Targeting hypoxia in cancer therapy. Nat Rev
Cancer 11(6):393-410.
156. Park JE, Dutta B, Tse SW, Gupta N, Tan CF, Low JK, Yeoh KW, Kon OL,
Tam JP, Sze SK. 2019. Hypoxia-induced tumor exosomes promote M2-
like macrophage polarization of infiltrating myeloid cells and microRNA-
mediated metabolic shift. Oncogene 38(26):5158-5173.
157. Go G, Yun CW, Yoon YM, Lim JH, Lee JH, Lee SH. 2020. Role of PrP(C)
in cancer stem cell characteristics and drug resistance in colon cancer cells.
Anticancer Res 40(10):5611-5620.
158. Falker C, Hartmann A, Guett I, Dohler F, Altmeppen H, Betzel C, Schubert
R, Thurm D, Wegwitz F, Joshi P, Verderio C, Krasemann S, Glatzel M. 2016.
Exosomal cellular prion protein drives fibrillization of amyloid beta and
counteracts amyloid beta-mediated neurotoxicity. J Neurochem
137(1):88-100.
159. Guo BB, Bellingham SA, Hill AF. 2016. Stimulating the release of
exosomes increases the intercellular transfer of prions. J Biol Chem
291(10):5128-5137.
160. Weissmann C. 2004. The state of the prion. Nat Rev Microbiol 2(11):861-
871.
161. Semenza GL. 2007. Life with oxygen. Science 318(5847):62-64.
162. Duan P, Tan J, Miao Y, Zhang Q. 2019. Potential role of exosomes in the

151
pathophysiology, diagnosis, and treatment of hypoxic diseases. Am J
Transl Res 11(3):1184-1201.
163. Hoffmann AC, Mori R, Vallbohmer D, Brabender J, Klein E, Drebber U,
Baldus SE, Cooc J, Azuma M, Metzger R, Hoelscher AH, Danenberg KD,
Prenzel KL, Danenberg PV. 2008. High expression of HIF1a is a predictor
of clinical outcome in patients with pancreatic ductal adenocarcinomas and
correlated to PDGFA, VEGF, and bFGF. Neoplasia 10(7):674-679.
164. Maeda H, Wu J, Sawa T, Matsumura Y, Hori K. 2000. Tumor vascular
permeability and the EPR effect in macromolecular therapeutics: A review.
J Control Release 65(1-2):271-284.
165. Jing X, Yang F, Shao C, Wei K, Xie M, Shen H, Shu Y. 2019. Role of hypoxia
in cancer therapy by regulating the tumor microenvironment. Mol Cancer
18(1):157.
166. Ludwig N, Whiteside TL. 2018. Potential roles of tumor-derived
exosomes in angiogenesis. Expert Opin Ther Targets 22(5):409-417.
167. Cruz E, Kayser V. 2019. Monoclonal antibody therapy of solid tumors:
Clinical limitations and novel strategies to enhance treatment efficacy.
Biologics 13:33-51.
168. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A,
Fleming T, Eiermann W, Wolter J, Pegram M, Baselga J, Norton L. 2001.
Use of chemotherapy plus a monoclonal antibody against HER2 for
metastatic breast cancer that overexpresses HER2. N Engl J Med
344(11):783-792.
169. Graham J, Muhsin M, Kirkpatrick P. 2004. Cetuximab. Nat Rev Drug Discov
3(7):549-550.
170. Fornasier G, Francescon S, Baldo P. 2018. An update of efficacy and safety
of cetuximab in metastatic colorectal cancer: A narrative review. Adv Ther
35(10):1497-1509.
171. Baselga J. 2002. Why the epidermal growth factor receptor? The rationale

152
for cancer therapy. Oncologist 7 Suppl 4:2-8.
172. Gan HK, Walker F, Burgess AW, Rigopoulos A, Scott AM, Johns TG. 2007.
The epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor
AG1478 increases the formation of inactive untethered EGFR dimers.
Implications for combination therapy with monoclonal antibody 806. J Biol
Chem 282(5):2840-2850.
173. Reslan L, Dalle S, Dumontet C. 2009. Understanding and circumventing
resistance to anticancer monoclonal antibodies. MAbs 1(3):222-229.
174. Viloria-Petit A, Crombet T, Jothy S, Hicklin D, Bohlen P, Schlaeppi JM,
Rak J, Kerbel RS. 2001. Acquired resistance to the antitumor effect of
epidermal growth factor receptor-blocking antibodies in vivo: a role for
altered tumor angiogenesis. Cancer Res 61(13):5090-5101.
175. Li F, Emmerton KK, Jonas M, Zhang X, Miyamoto JB, Setter JR, Nicholas
ND, Okeley NM, Lyon RP, Benjamin DR, Law CL. 2016. Intracellular
released payload influences potency and bystander-killing effects of
antibody-drug conjugates in preclinical models. Cancer Res 76(9):2710-
2719.

153
ABSTRACT IN KOREAN

정상 프리온 단백질 조절을 통한 대장암 세포에서의 항암제의

치료효과 향상

윤 철 원

순천향대학교 대학원 의생명과학과

(지도교수 : 이 상 훈)

대장암은 질병의 후기 진행단계에서 공격적인 전이로 인한 암 관련 죽음의

주요원인 중 하나이다. 최근 연구들을 통해 대장암세포의 약물내성 획득에 관한

기작을 확인했지만, 약물내성을 극복하기 위한 새로운 치료법은 아직도 개발되지

않았다. 여전히 항암제 내성은 대장암 환자의 심각한 문제이고, 이러한 문제를 해

결하기 위해 다양한 항암 인자와 분자적 타겟에 대한 연구가 진행되고 있다. 송과

선에서 만들어져 내재적으로 분비되는 인돌아민 호르몬인 멜라토닌은 대장암을

포함한 다양한 암에서 산화성 스트레스 및 세포사멸을 유도하는 다양한 기작을

통해 항암효과를 보이는 것으로 알려져 있다. 저산소증은 대장암에서 종양의 증식

과 전이를 촉진시킨다. 종양 미세환경은 일반적으로 저산소증으로 특징 지어지기

때문에, 이에 대한 이해는 암 치료에 있어서 중요하다. 또한, 암 전이에 대한 프

154
리온 단백질의 역할에 대한 관심이 증가하고 있지만, 프리온 단백질을 통한 세포

내 상호작용을 통한 정확한 기작은 잘 알려져 있지 않다. 따라서 본인은 1) 프리

온 단백질이 세포 생존신호경로 조절을 통해 대장암의 약물내성 및 멜라토닌을

통한 활성산소가 매개된 세포사멸에 관여하고, 2) 프리온 단백질을 포함한 세포외

소포체를 통한 대장암세포의 종양진행을 조절할 것이라 가정하였다.

5FU 내성 대장암 세포에서 프리온 단백질의 발현에 따른 대장암의 변화 양

상을 확인하였다. 대장암 환자로부터 얻은 검체에서 세포 프리온 단백질의 발현은

대장암의 전이 및 종양의 단계와 유의미한 상관관계를 보여주었고 대장암 세포의

종양형성 및 유전학적 변화에 영향을 주었다. 또한, 항암제 내성 대장암세포에서

프리온 단백질의 발현이 상당히 증가됨을 확인하였고, 5FU 처리 후 프리온 단백

질은 PI3K-AKT 신호경로 및 세포 주기 관련 단백질의 활성화 유지를 통해 대

장암 세포의 생존과 증식을 촉진시키며 스트레스 관련 단백질의 활성화를 억제하

였다. 프리온 단백질의 발현을 억제한 후 5FU의 처리는 caspase-3의 활성화를

통해 세포사멸을 증가시켰다.

대장암에서 멜라토닌 처리는 프리온 단백질과 PINK1의 발현을 감소시키고

미토콘드리아에 과산화물의 축적을 증가시켰다. 프리온 단백질의 억제는 멜라토닌

의 효과를 강화시키고 PINK1의 발현을 상당히 감소시켜 대장암세포에 과산화물

의 축적을 증가시켜 ER stress 유도를 통한 세포사멸을 증가시켰다. 다음으로, 저

산소 종양 미세환경은 대장암에서 프리온 단백질을 발현하는 엑소좀을 증가시키

고, 이러한 엑소좀은 프리온 단백질 발현에 따라 대장암 세포의 행동과 종양의 진

행을 조절한다. 저산소 상태에서 나온 대장암 세포 엑소좀은 종양의 구형 형성,

종양 유도 유전자 발현, 이동성 및 종양의 성장을 촉진시켰다. 추가로, 엑소좀은

155
혈관 내피 세포의 침투성, 이동성과 혈관 형성 사이토카인의 분비를 증가시켰다.

이러한 효과는 프리온 단백질 발현과 관련되어 있고, 마우스 동물 모델에서 5FU

와 항 프리온 단백질 항체와의 병합처리는 대장암의 진행을 상당히 억제하였다.

결론적으로 프리온 단백질이 통해 대장암세포에서 약물 내성을 획득하는데

중요한 역할을 하고 있다는 것을 제시한다. 또한, 항암치료제로서 멜라토닌은 프

리온 단백질과 미토콘드리아의 항상성 조절에 관여하는 PINK1 단백질의 조절을

통해 대장암세포의 미토콘드리아 활성산소종의 생성을 증가시키고, ER stress를

통한 세포사멸을 유도하였고, 프리온 단백질의 siRNA를 통한 억제는 멜라토닌의

효과를 강화시켜 대장암 치료를 위한 새로운 치료방법이 될 수 있음을 확인하였

다. 추가로, 저산소 상태에서 대장암세포에서 분비되는 프리온 단백질을 발현하는

엑소좀이 대장암간/대장암과 내피세포의 세포 신호전달에 중요한 인자임을 확인

하였다. 마지막으로 프리온 단백질의 억제를 통하여 항암화학 치료 시 대장암의

치료효율을 향상시킬 수 있음을 보여준다.

주제어 : 대장암, 저산소 조건, 멜라토닌, 5FU, 정상 프리온 단백질, 엑소좀, 항암

제 내성, 항체 치료

156
ACKNOWLEDGEMENTS

학위기간 중에 많은 어려움들이 있었지만 박사학위를 마칠 수 있었던 것은

많은 분들의 도움과, 격려로 무사히 학위과정을 마칠 수 있었습니다. 우선 박사

학위를 마칠 수 있도록 이끌어주신 이상훈 교수님께 감사의 뜻을 전하고

싶습니다. 학위 과정 동안 진행한 연구를 전폭적으로 지원해 주시고 다양한

관점에서 연구의 방향을 이끌어 주셔서 감사 드립니다. 또한, 한 명의 연구자로서

교수님의 모습을 본보기 삼아 성장하고 훌륭한 연구자가 되고 초심을 잃지

않도록 하겠습니다. 석사, 박사 학위과정 동안 지도해 주셔서 감사합니다.

대학원 학위과정 동안 저에게 힘이 되어주고 격려와 믿음으로 기다려 주신

부모님께 진심으로 감사의 말씀을 드립니다. 대학원 진학이라는 저의 결정을

존중해주시고 여러 방면으로 도움을 주셔서 이렇게 무사히 학위과정을 마칠 수

있었다고 생각합니다. 앞으로 새로운 환경에서 새로이 시작될 학위과정 이후의

삶에서도 이러한 고마움을 생각하며 더 발전하도록 하겠습니다.

학위논문의 심사과정에서 아낌없는 충고, 조언과 격려를 해주신 순천향대학교

의료생명공학과 이미영 교수님, 순천향대학교 비뇨기과 송윤섭 교수님,

단국대학교 치과대학 이준희 교수님, 그리고 충북대학교 수의학과 이현직

교수님께도 감사의 인사를 드립니다. 부족한 저를 격려해 주시고 진심 어린

조언을 통해 저의 학위논문의 주제와 내용이 더 풍성해 질 수 있었다고

생각합니다. 교수님들의 조언을 통해 좀 더 발전하고 노력하는 연구자가 될 수

있도록 노력하겠습니다.

157
마지막으로 저의 학위과정 동안 만났던 모든 분들께 진심으로 감사의 말씀을

드립니다. 연구실에서 많은 일이 있었지만, 학위과정 동안 서로 의지할 수 있었던

한용석, 윤여민, 이가은에게 감사하고, 연구실 생활을 하는 동안 다양한 조언을

해주신 고경윤 박사님, 예지혜 박사님, 박희정 박사님께 감사의 인사를 드립니다.

짧은 감사의 글이지만 저에게 도움을 주신 분들께 다시 한번 감사의 인사를

드리며 글을 마치겠습니다.

158

You might also like