rnai screening-based identification of usp10 as a novel

75
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: 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 저작자표시. 귀하는 원저작자를 표시하여야 합니다. 비영리. 귀하는 이 저작물을 영리 목적으로 이용할 수 없습니다. 변경금지. 귀하는 이 저작물을 개작, 변형 또는 가공할 수 없습니다.

Upload: others

Post on 17-Apr-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: RNAi Screening-based Identification of USP10 as a Novel

저 시-비 리- 경 지 2.0 한민

는 아래 조건 르는 경 에 한하여 게

l 저 물 복제, 포, 전송, 전시, 공연 송할 수 습니다.

다 과 같 조건 라야 합니다:

l 하는, 저 물 나 포 경 , 저 물에 적 된 허락조건 명확하게 나타내어야 합니다.

l 저 터 허가를 면 러한 조건들 적 되지 않습니다.

저 에 른 리는 내 에 하여 향 지 않습니다.

것 허락규약(Legal Code) 해하 쉽게 약한 것 니다.

Disclaimer

저 시. 하는 원저 를 시하여야 합니다.

비 리. 하는 저 물 리 목적 할 수 없습니다.

경 지. 하는 저 물 개 , 형 또는 가공할 수 없습니다.

Page 2: RNAi Screening-based Identification of USP10 as a Novel

이학 박사학위 논문

RNAi Screening-based Identification of USP10 as a Novel Regulator of

Paraptosis

아 주 대 학 교 대 학 원

의 생 명 과 학 과

김 진 엽

Page 3: RNAi Screening-based Identification of USP10 as a Novel

RNAi Screening-based Identification of USP10 as a Novel Regulator of

Paraptosis

지도교수 최 경 숙

이 논문을 이학 박사학위 논문으로 제출함.

2018 년 2 월

아 주 대 학 교 대 학 원

의 생 명 과 학 과

김 진 엽

Page 4: RNAi Screening-based Identification of USP10 as a Novel
Page 5: RNAi Screening-based Identification of USP10 as a Novel

i

-ABSTRACT-

RNAi Screening-based Identification of USP10 as a Novel Regulator of Paraptosis

Accumulating reports demonstrate the innate and acquired resistance of malignant

cancer cells to apoptosis and apoptosis does not explain all the effects of cancer therapy.

Recently, paraptosis, a type of programmed cell death that is accompanied by dilation of

mitochondria and/or endoplasmic reticulum (ER), is an area of interest for cancer

research as an alternative way to kill apoptosis-resistant cancers. In this study, we

describe the adaptation and validation of a high-content cell-based assay with the

powerful concept of screening of curcumin-induced paraptosis to identify novel

regulators of paraptosis in malignant breast cancer cells. From the screening using Mito-

YFP cells that express the fluorescence selectively in mitochondria, we selected 38

target hit gene candidates against paraptosis with the activities to induce mitochondrial

dilation, a morphological feature of paraptosis. Among them, ubiquitin specific

peptidase 10 (USP10), a ubiquitin specific protease, was chosen as a strongly active

candidate for further evaluation of its involvement in paraptosis. We found that not only

siRNA-mediated knockdown of USP10 but also treatment with spautin-1, a USP10

inhibitor, effectively attenuated curcumin-induced paraptosis. In summary, this

systematic assay concept of the siRNA library screening based on the morphological

changes in mitochondria may offer the opportunity to identify the potent regulators of

paraptosis as well as the new target candidates of genes and drugs against malignant

Page 6: RNAi Screening-based Identification of USP10 as a Novel

ii

breast cancer.

Page 7: RNAi Screening-based Identification of USP10 as a Novel

iii

TABLE OF CONTENT

ABSTRACT---------------------------------------------------------------------------------------ⅰ

TABLE OF CONTENTS----------------------------------------------------------------------ⅲ

LIST OF FUGURES----------------------------------------------------------------------------ⅴ

Ⅰ. INTRODUCTION----------------------------------------------------------------------------1

Ⅱ. MATERIALS AND METHODS-----------------------------------------------------------6

A. Chemicals, antibodies and cell culture--------------------------------------------- 6

B. Establishment of the stable cell lines expressing the fluorescence

specifically in mitochondria or ER--------------------------------------------------6

C. Liquid dispensing and automation--------------------------------------------------7

D. siRNA library----------------------------------------------------------------------------8

E. Screening of siRNA library in curcumin-induced paraptosis assay----------8

F. Image acquisition, data analysis, and management of screening data-------9

G. Pathway analysis based on the selected siRNAs---------------------------------10

H. Measurement of cellular viability using calcein-AM and EthD-1------------10

I. Western blotting------------------------------------------------------------------------11

J. Statistical analysis---------------------------------------------------------------------12

Ⅲ. RESULTS-------------------------------------------------------------------------------------14

1. Paraptosis model validation -----------------------------------------------------14

2. Assay adaptation and validation ------------------------------------------------21

3. siRNA-based screen to identify genes involved in curcumin-induced

Page 8: RNAi Screening-based Identification of USP10 as a Novel

iv

paraptosis ----------------------------------------------------------------------------29

4. USP10 effectively inhibits curcumin-induced paraptosis in malignant

breast cancer cells.-----------------------------------------------------------------35

Ⅳ. DISCUSSION--------------------------------------------------------------------------------50

CONCLUSION-----------------------------------------------------------------------------------56

REFERENCES-----------------------------------------------------------------------------------57

국문요약-------------------------------------------------------------------------------------------64

Page 9: RNAi Screening-based Identification of USP10 as a Novel

v

LIST OF FIGURES

Fig. 1. Curcumin induces mitochondrial dilation leading to megamitochondria

formation on the transmission electron microscopy ----------------------------17

Fig. 2. Curcumin induces mitochondrial dilation leading to megamitochondria

formation on immunofluorescence -------------------------------------------------18

Fig. 3. Image-based assay for phenotypic profiling of mitochondrial aggregation-19

Fig. 4. High-content image acquisition and data analysis -------------------------------20

Fig. 5. Adapting the assay to a 384-well microtiter plate format ----------------------24

Fig. 6. Optimizing the assay to the 384-well microtiter plate format -----------------25

Fig. 7. Optimizing the assay to the 384-well microtiter plate format on the

immunofluorescence -------------------------------------------------------------------26

Fig. 8. Knock-down effect of the CHOP by siRNA ---------------------------------------27

Fig. 9. Evaluation of optimized assay conditions -----------------------------------------28

Fig. 10. Workflow of curcumin-induced mitochondrial aggregation assay ---------31

Fig. 11. Results of screening for curcumin-induced paraptosis regulators ----------32

Fig. 12. Selected hits for genes that appear to be involved in curcumin-induced

mitochondrial aggregation -----------------------------------------------------------33

Fig. 13. Enrichment of the selected hits from the primary screen --------------------34

Fig. 14. Secondary screening to confirm the involvement of USP10 in curcumin-

induced mitochondrial aggregation ---------------------------------------------------------39

Fig. 15. Secondary screening to confirm the involvement of USP10 in curcumin-

Page 10: RNAi Screening-based Identification of USP10 as a Novel

vi

induced mitochondrial aggregation on the immunofluorescence ---------------------40

Fig. 16. Knock-down effect of the USP10 by siRNA -------------------------------------41

Fig. 17. The USP10 inhibitor, spautin-1, effectively inhibits curcumin-induced

mitochondria aggregation ------------------------------------------------------------42

Fig. 18. The USP10 inhibitor, spautin-1, effectively inhibits curcumin-induced

paraptosis in malignant breast cancer cell on the immunofluorescence ----43

Fig. 19. The USP10 inhibitor, spautin-1, effectively inhibits curcumin-induced cell

death---------------------------------------------------------------------------------------44

Fig. 20. The USP10 inhibitor, spautin-1, effectively inhibits curcumin-induced cell

death in malignant breast cancer cell ----------------------------------------------45

Fig. 21. Spautin-1 effectively inhibits curcumin or demethoxycurcumin (DMC)-

induced cytoplasmic vacuolation ----------------------------------------------------

46

Fig. 22. Spautin-1 effectively inhibits curcumin-induced cytoplasmic vacuolation in

various cancer cells --------------------------------------------------------------------47

Fig. 23. Spautin-1 effectively inhibits curcumin or demethoxycurcumin (DMC)-

induced cell death ----------------------------------------------------------------------48

Fig. 24. The USP10 inhibitor, spautin-1, effectively inhibits curcumin-induced ERK

activation ---------------------------------------------------------------------------------

49

Page 11: RNAi Screening-based Identification of USP10 as a Novel

1

Ⅰ. INTRODUCTION

Breast cancer is one of the most common cancer types that cause oncologic

morbidity and mortality among women worldwide (Torre LA, et al. 2015). Currently,

breast cancers are treated with tailored combinations of surgery, chemotherapy and

radiation (Yagata H, et al. 2011). Although ongoing research is seeking to develop

more effective therapeutic strategies with minimal side effects, we lack a specific

targeted agent for the treatment of triple negative breast cancer (TNBC), and the

treatment options for TNBC patients are limited (Bianchini G, et al. 2016). To

identify novel targets that will enable us to effectively kill TNBC cells, we need to

develop scalable strategies with powerful tools. Since malignant cancer cells, including

TNBC cells, are resistant to pro-apoptotic treatments, it could be helpful to identify

means to induce alternative cell death mode(s) that will overcome therapeutic resistance

in these cancer cells.

Paraptosis, a type of programmed cell death (PCD) that is characterized by

dilation of mitochondria and/or endoplasmic reticulum (ER), is independent of caspases

Page 12: RNAi Screening-based Identification of USP10 as a Novel

2

and lacks apoptotic morphologies (Sperandio S, et al. 2000). Although the molecular

basis of paraptosis has not yet been extensively explored, this process is known to

require de novo protein synthesis (Kar R, et al. 2009; Wang WB, et al. 2012).

Various natural products, including curcumin (Yoon MJ, et al. 2010), celastrol (Yoon

MJ, et al. 2014) and withaferin A (Ghosh K, et al. 2016), have been shown to induce

paraptosis in malignant breast cancer cells. In particular, curcumin induces paraptosis

selectively in malignant breast cancer cells while sparing normal cells (FRBM 2010).

Moreover, curcumin-induced paraptosis is not blocked by the overexpression of various

anti-apoptotic proteins. Although the clinical application of curcumin has been limited

by its poor bioavailability, an understanding of its cancer-selective action mechanism

could facilitate the development of safe and effective anti-cancer drugs. Recent studies

have shown that paraptosis is associated with the generation of reactive oxygen species

(Yoon MJ, et al. 2014; Priyadarsini K, et al. 2003; Kunwar A, et al. 2011;

Tamvakopoulos, C, et al. 2007; Lee D, et al. 2016), imbalances in the homeostasis of

ions (e.g., Ca2+ and K+) (Priyadarsini K, et al. 2003; Lee D, et al. 2016; Bravo R, et

al. 2012; Yoon MJ, et al. 2014; Chen D, et al. 2016; Carew JS, et al. 2002), and

Page 13: RNAi Screening-based Identification of USP10 as a Novel

3

perturbation of cellular proteostasis via proteasomal inhibition and disruption of

sulfhydryl homeostasis (Yoon MJ, et al. 2014; Priyadarsini K, et al. 2003; Lee D, et

al. 2016; Bravo R, et al. 2012; Yoon MJ, et al. 2014; Yuan J, et al. 2010; Lin Z, et

al. 2013; Takahashi M, et al. 2013). However, the mechanisms underlying paraptosis,

particularly the signals responsible for the dilation of mitochondria and the ER, are still

not clearly understood. Clarification of the genes critical for controlling paraptosis may

suggest novel therapeutic targets for various diseases, including TNBCs, for which we

currently lack effective druggable targets.

Over the past 20 years, major technological advances have yielded systems that

can perform automated microscopic screening for visual phenotypes in cells and

organisms. Various screening approaches have been used to evaluate anti-cancer and

anti-viral efficacies for drug discovery, and these strategies have been adapted to 96-

well and (in some cases) 384-well microtiter plate formats to enable low- to high-

throughput applications (Shum D, et al. 2014). One of the most powerful tools

available for the rapid identification of new target genes that may act against malignant

cancer cells is high-content screening (HCS), which combines high-throughput

Page 14: RNAi Screening-based Identification of USP10 as a Novel

4

screening (HTS) with the ability to collect cellular images of biological processes. HCS

has been used to characterize gene functions in cells subjected to RNAi or genetic

perturbations, and to identify potential drug candidates from large libraries of small

molecules (Carpenter AE, et al. 2007). Historically, HCS has often relied on relatively

simple assays, such as assessment of cell growth/ viability or the levels of luminescent

reporter genes. However, HCS may be performed using various visual profiling

approaches, such as tagging a protein of interest with various-colored fluorescent

protein or engineering a cell line to respond to stimuli appropriately (Carpenter AE, et

al. 2007; Boutros M, et al. 2015). The measurement of multiple parameters and the

integration of these data at the single-cell level can enable researchers to perform

complex tasks, such as precisely identifying the proteins involved in a specific

biological process or predicting the target of a drug candidate (Loo LH, et al. 2007;

Genovesio A, et al. 2011). For the successful HCS screening of the target candidates,

however, it is critical to establish a specific and powerful assay concept.

The morphological features (dilation of mitochondria and the ER) of paraptosis

have been characterized, but the biochemical features of this process are not well

Page 15: RNAi Screening-based Identification of USP10 as a Novel

5

understood. A large-scale screening of genes that contribute to the phenotypic changes

seen during paraptosis should add to our knowledge of the molecular basis underlying

this novel cell death mode. In an attempt to identify the key regulators of paraptosis, I

herein conceptualized a cell-based assay for detecting the mitochondrial changes that

represent a main morphological feature of paraptosis. I used a multiparametric readout

to simultaneously monitor: 1) the phenotypic changes of mitochondria during the

curcumin-induced paraptosis of malignant breast cancer cells; and 2) the viability of

these cells. This allowed us to rapidly score the effects of siRNAs from a siRNA library

of 2,732 druggable genes. This screening identified hits that was enriched for several

classified signal pathways, suggesting that our method could be used to suggest new

targets for anti-cancer therapies against malignant breast cancer cells. In addition, I

show that USP10 may be a critical factor for curcumin-induced mitochondrial dilation

and subsequent paraptotic cell death.

Page 16: RNAi Screening-based Identification of USP10 as a Novel

6

Ⅱ. MATERIALS AND METHODS

A. Chemicals, antibodies and cell culture

Curcumin, dimethoxycucumin (DMC), Spautin-1 and Dimethyl sulfoxide (DMSO)

were purchased from Sigma (St. Louis, MO). I used antibodies against α-tubulin (Santa

Cruz biotechnology, Santa Cruz, CA); phospho-ERK1/2, total ERK1/2, CHOP (Cell

Signaling, Beverly, MA); USP10 (Abcam, Cambridge, MA); HRP-conjugated anti-

rabbit IgG and anti-mouse IgG (Invitrogen, San Diego, CA); DRAQ5 (BioStatus,

Leicestershire, UK). Dharmafect 1 transfection reagent, 5ⅹsiRNA buffer and RNase

free water were purchased from GE Dharmacon (Lafayette, CO). MDA-MB 435S,

MDA-MB 231 and MCF-7 human breast cancer cell lines, which were purchased from

American Type Culture Collection (ATCC, Manassas, VA), were cultured in 5% CO2 at

37oC using DMEM or DMEM supplemented with 10% fetal bovine serum (FBS) and

1% antibiotics GIBCO-BRL, Grand Island, NY). Cell culture passage number less than

ten was used in the present study.

B. Establishment of the stable cell lines expressing the fluorescence specifically in

Page 17: RNAi Screening-based Identification of USP10 as a Novel

7

mitochondria

To establish the stable cell lines expressing the fluorescence specifically in

mitochondria, MDA-MB 435S cells were transfected with the pEYFP-Mito vector

(Clontech Laboratories, Mountain View, CA). Stable cell line overexpressing pEYFP-

Mito (YFP-Mito cells) was selected with fresh medium containing 500 µg/mL G418

(Invitrogen, San Diego, CA). Images of mitochondria were obtained from the

fluorescence microscopy using a Zeiss filter set #10.

C. Liquid dispensation and automation

For the plating and transfer of siRNAs, I used a custom-designed 384-head pipette

system with disposable low-volume polypropylene tips on a CyBi®-Well platform

(CyBio, Woburn, MA). The addition of cells, dispensation of growth media, mixing

steps during compound treatment or siRNA transfection, fixation and staining of cells

were performed using a Multidrop 384 pipette (Thermo Fisher Scientific, Waltham,

MA) for assay adaptation or a WellMate (Thermo Fisher Scientific) for screening. The

assay plates were incubated in a Steri-Cult Incubator (Sanyo, Japan) under controlled

Page 18: RNAi Screening-based Identification of USP10 as a Novel

8

humidity and 5% CO2 at 37℃.

D. siRNA library

I used an OTP (On-TARGET plus) human druggable subset library containing 2,732

individual siRNA duplexes (GE Dharmacon, Lafayette, CO). This collection consists of

sub-libraries representing ion channels, proteases, phosphatases, kinases, ubiquitin

conjugation-related factors, and G-protein coupled receptors (GPCRs). siRNA library

stocks were formulated in siRNA buffer (GE Dharmacon) at 1 µM, and were added to

4% of the final assay volume for a final siRNA concentration of 20 nM.

E. Screening of the siRNA library during curcumin-induced paraptosis

For siRNA reverse transfection, 1 μl of 1μM siRNAs per well were dispensed to a 384-

well microtiter plate (Greiner Bio-one, Frickenhausen, Germany) using the Cybi-Well,

and 1ⅹsiRNA buffer was dispensed at 4 μl per well using the WellMate.

Immediately thereafter, a 0.1:4.9 mixture of transfection (TF) reagent mixture and Opti-

MEM (GIBCO-BRL, Grand Island, NY) was dispensed to the plate at 5 µl per well

Page 19: RNAi Screening-based Identification of USP10 as a Novel

9

using the WellMate. The plate was incubated for 20 min, and then YFP-Mito cells were

seeded at 4,000 cells per well in 40 µl of FBS-containing media without antibiotics,

using the WellMate. The plate was incubated for 48 h and then loaded with 0.25 µl/well

curcumin (final concentration, 30 µM in 0.5% DMSO, v/v). After incubation for 12 h,

the cells were fixed for 30 min with 4% paraformaldehyde (PFA, w/v) and nuclei were

stained for 1 h with DraQ5 (BioStatus). Images of YFP-Mito signals and the nuclei were

acquired using an Opera microscope system (Perkin Elmer, Waltham, MA) and

analyzed with the Columbus 2.3 software (Perkin Elmer, Waltham, MA).

F. Image acquisition, data analysis, and management of screening data

For assay development and screening, images were acquired using the Opera automated

laser confocal microscope system. Images were captured at the following wavelengths:

605 nm excitation/690 nm emission in the red channel for DraQ5-stained nuclei, and

488 nm excitation/540 nm emission in the green channel for YFP-expressing

mitochondria. The exposure times were 80 and 40 ms, respectively. Nine images per

well were collected using a 20ⅹmagnifying objective. Images were analyzed using the

Page 20: RNAi Screening-based Identification of USP10 as a Novel

10

segmentation algorithm built into the Columbus 2.3 software; this algorithm counts

nuclei as the number of objects with DraQ5 pixel intensities above background and

counts mitochondria-aggregated spots as the number of objects with YFP pixel

intensities above background. Since the nuclei of dead cells are removed during the

fixation and staining steps of the screening process, cell viability was measured as the

percentage of nuclei counted in treated versus untreated control cells (Count (%

Control)). The mitochondrial aggregation ratio was calculated as the number of total

mitochondrial aggregation spots/total cell number.

G. Pathway analysis based on the selected siRNAs

To explore the pathway enrichment of the screened genes, I conducted KEGG (Kyoto

Encyclopedia of Genes and Genomes) database analyses using the DAVID (the

Database for Annotation, Visualization and Integrated Discovery) online analysis tool.

H. Measurement of cellular viability using calcein-AM and EthD-1

Cells (5× 104 cells) were cultured in 24-well plates and treated as indicated. For

Page 21: RNAi Screening-based Identification of USP10 as a Novel

11

measurement of cellular viability, 2 µM calcein-AM, a green fluorescent indicator of the

intracellular esterase activity of cells, and 4 µM EthD-1, a red fluorescent indicator of

membrane damaged (dead) cells, were added to each well, and the plates were

incubated for 5 min in 5% CO2 at 37 °C. Cells were then observed under a fluorescence

microscope (Axiovert 200M; Carl Zeiss) equipped with Q2 Zeiss filter sets #46 and

#64HE. Viable cells, corresponding to those that exclusively exhibited green

fluorescence, were counted in five fields per well at × 200 magnification. Only

exclusively green cells were counted as live because bicolored (green and red) cells

cannot be unambiguously assigned to live or dead groups. The percentage of live cells,

calculated as green cells/(green+red+bicolored cells), was normalized to that of

untreated control cells (100%).

I. Western blotting

Cells were washed in PBS and lysed in boiling sodium dodecyl sulfate–polyacrylamide

gel electrophoresis (SDS–PAGE) sample buffer (62.5 mM Tris (pH 6.8), 1% SDS, 10%

glycerol, and 5% β-mercaptoethanol). The lysates were boiled for 5 min, separated by

Page 22: RNAi Screening-based Identification of USP10 as a Novel

12

SDS–PAGE, and transferred to an Immobilon membrane (Millipore, Billerica MA).

After nonspecific binding sites were blocked for 1 h using 5% skim milk, the

membranes were incubated for 2 h with specific antibodies. Membranes were then

washed three times with Tris-Buffered Saline Tween-20 (TBST) and incubated further

for 1 h with horseradish peroxidase-conjugated anti-rabbit or -mouse antibody.

Visualization of protein bands was accomplished using ECL (GE Dharmacon, Lafayette,

CO). The respective protein band intensity was quantified by densitometric analysis

using the NIH ImageJ program. Representative results from at least three independent

experiments are shown.

J. Statistical analysis

All hit confirmation-related data are presented as mean ± S.D. (standard deviation) from

at least three separate experiments. Statistical analysis was performed with GraphPad

Prism (GraphPad Software Inc., Sandiego, CA) was used. Normality of data was

assessed by Kolmogorov-Smirnov testes and equal variance using Bartlett's test. For

normal distribution, statistical differences were determined using an analysis of variance

Page 23: RNAi Screening-based Identification of USP10 as a Novel

13

(ANOVA) followed by Bonferroni multiple comparison test. P < 0.05 was considered

statistically significant.

Page 24: RNAi Screening-based Identification of USP10 as a Novel

14

Ⅲ. RESULTS

1. Paraptosis model validation

A main feature of paraptosis is an extensive cytoplasmic vacuolization that is derived

from mitochondria and the ER (Sperandio S, et al. 2000). I previously showed that

curcumin induces paraptosis via formation of megamitochondria due to fusion of

swollen mitochondria, and expansion of the ER structure by fusion of swollen ER

components (Yoon MJ, et al. 2010; Yoon MJ, et al. 2014a; Yoon MJ, et al.

2014b). To validate our intended model, I confirmed that this occurs in malignant

breast cancer cells and indeed, this was observed in our system (Fig. 1 and 2).

The ER expansion seen during paraptosis is thought to arise due to an overload of

misfolded proteins within the ER lumen and a subsequent increase of osmotic pressure,

which draws water from the cytoplasm to distend the ER lumen (Lee D, et al. 2016;

Mimnaugh EG, et al. 2006). However, the signals responsible for triggering

mitochondrial dilation have not yet been clearly determined. To identify key regulators

Page 25: RNAi Screening-based Identification of USP10 as a Novel

15

of paraptosis, I designed an image-based cellular screening assay that detects

mitochondrial dilation, which is an initial and major morphological change in paraptosis,

in cells exposed to a small interfereing RNA (siRNA) library. For this purpose, I

employed MDA-MB 435S sublines stably expressing an YFP-Mito construct (YFP-

Mito cells). I speculated that fluorescence microscopy would enable us to clearly

distinguish between the megamitochondria (mitochondrial aggregation) formed during

the paraptosis of YFP-Mito cells and the elongated mitochondria of untreated cells (Fig.

3). If this was the case, detecting the extent of mitochondrial aggregation could allow us

to perform direct imaging and quantification using an automated fluorescence

microscope. Using a high-content approach, I scored the effects of transfected siRNAs

using a multiparametric readout: I detected both the progression of paraptosis (by the

amount of detected mitochondrial aggregation) and cytotoxicity (by the relative number

of nuclei in treated versus untreated cells). Using the analysis module of the Columbus

2.3 HCS analysis software, I first identified the nucleus of each cell using an object-

recognition tool based on the pixel gradient of the red DraQ5 channel (Fig. 3). To assess

mitochondrial aggregation, I designed an assay to detect the total spot number based on

Page 26: RNAi Screening-based Identification of USP10 as a Novel

16

the area and intensity of the fluorescence following treatment of cells with 40 µM

curcumin in green channel (Fig. 3). To verify the concept of our assay, I used an Opera

automated laser confocal microscope to observe the distribution of YFP-expressing

mitochondria in cells treated with vehicle (0.5% DMSO, v/v) or 40 µM curcumin for 12

h (Fig. 4). The negative control showed the elongated and branched YFP signal pattern

of normal mitochondria, whereas curcumin treatment drastically induced mitochondrial

aggregation. These results indicate that our image-based cellular assay system could be

used to evaluate the activities of candidate factors that might regulate paraptosis.

Page 27: RNAi Screening-based Identification of USP10 as a Novel

17

Fig. 1. Curcumin induces mitochondrial dilation leading to megamitochondria

formation on the transmission electron microscopy. MDA-MB 435S cells were left

untreated or treated with 30 µM curcumin for 12 h. Transmission electron microscopy

was performed. The black arrowhead and white arrowhead indicate mitochondria and

the ER, respectively; the black arrow and white arrow indicate dilated mitochondria and

dilated ER, respectively. Bar, 2 µm.

Page 28: RNAi Screening-based Identification of USP10 as a Novel

18

Fig. 2. Curcumin induces mitochondrial dilation leading to megamitochondria

formation on immunofluorescence. MDA-MB 435S cells were left untreated or

treated with 30 µM curcumin for 12 h. Cells were stained with Mito-tracker green

(MTG) and observed by phase-contrast and fluorescence microscopy. Bar, 20 µm.

Page 29: RNAi Screening-based Identification of USP10 as a Novel

19

Fig. 3. Image-based assay for phenotypic profiling of mitochondrial aggregation.

YFP-Mito-expressing MDA-MB 435S sublines (YFP-Mito cells) were cultured in 384-

well plates at a density of 4,000 cells per well, treated with 40 µM curcumin for 12 h,

and evaluated for mitochondrial aggregation. The images were acquired from Columbus

2.3 software. Bars, 50 µm.

Page 30: RNAi Screening-based Identification of USP10 as a Novel

20

Fig. 4. High-content image acquisition and data analysis. YFP-Mito cells were

treated with vehicle (0.5% DMSO (v/v)) or 40 µM curcumin for 12 h. The plates were

fixed and stained with DRAQ5, as described in the Materials and Methods. Images were

acquired using an Opera automated laser confocal microscope system, with the green

channel used for YFP-expressing mitochondria and the red channel used to detect

DRAQ5-stained nuclei (upper panel). Bar, 50 µm (upper panel). Cell viability (Count

(% Control)) and mitochondrial aggregation ratios were quantitatively measured using

the Columbus 2.3 software, as described in the Materials and Methods (lower panel) .

*p < 0.001 vs. 0.5% DMSO.

Page 31: RNAi Screening-based Identification of USP10 as a Novel

21

2. Assay adaptation and validation

As a first step toward automated HTS, I miniaturized our assay from a 24-well format to

a 384-well microtiter plate format, and assessed whether curcumin induced

mitochondrial aggregation under the siRNA-supplemented screening condition. By

reverse transfection, non-targeting siRNA was dispensed into 384-Well microtiter plates

and then YFP-Mito cells were seeded and incubated for 48 h. The seeding density was

4,000 cells per well, as preliminary experiments indicated that this density yielded a

good overall consistency of cell distribution (data not shown). I then treated cells with

serially diluted curcumin (8 doses between 2.5 µM and 60 µM) for 24 h, and assessed

cytotoxicity and mitochondrial aggregation. Under the tested conditions, curcumin

dose-dependently induced both aggregation, which represents mitochondrial dilation,

and cell death (Fig. 5). Our results demonstrated that our mitochondrial aggregation

assay to measure paraptosis could be adapted to a 384-well microtiter plate format,

giving us the confidence to move forward in developing this concept as an automated

high-content screening assay.

Page 32: RNAi Screening-based Identification of USP10 as a Novel

22

Recent studies have shown that mitochondria and the ER are interconnected physically

and functionally by the mitochondria-associated ER membrane (MAM) structure, which

forms during the progression of paraptosis (Bravo R, et al. 2012). In addition, We

recently showed that paraptotic inducers (e.g., curcumin, dimethoxycurcumin and

celastrol) inhibit proteasome activity and upregulate CHOP, which is a member of the

C/EBP family of transcription factors (Yoon MJ, et al. 2010; Yoon MJ, et al. 2014a;

Yoon MJ, et al. 2014b). In the present study, I first evaluated whether CHOP siRNA

could be used as a positive control for blocking curcumin-induced paraptosis in our

assay, using the 384-well format.

To determine the optimal assay conditions for the screening to detect mitochondrial

aggregation, I treated YFP-Mito cells with serially diluted curcumin for 12, 16 or 20 h,

and measured cell viability and the mitochondrial aggregation ratio (Fig. 6). Since

mitochondrial aggregation precedes overt paraptotic cell death, I searched for an assay

condition under which I observed obvious monitor mitochondrial aggregation without

induction of cell death. I found that treatment of cells with 30 µM curcumin for 12 h

induced mitochondrial aggregation without cytotoxicity, and that this was effectively

Page 33: RNAi Screening-based Identification of USP10 as a Novel

23

inhibited by transfection of CHOP siRNA (Fig. 7), which efficiently knocked down

CHOP expression (Fig. 8). I thus used this treatment as the optimal assay condition to

subsequently screen our siRNA library to identify key regulators of paraptosis.

Next, I evaluated the assay performance in a control run using three 384-well microtiter

plates containing cells transfected with the negative (non-targeting siRNA) and positive

(CHOP siRNA) control siRNAs and treated with 30 µM curcumin or vehicle (0.5%

DMSO v/v). The assay had normalized mitochondrial aggregation ratio; the average

high mitochondrial aggregation ratio was (100 ± 8.6)% with 9% coefficient of variation

(CV) and the average low mitochondria aggregation ratio was (31.8 ± 5.6)% with 15%

CV (Fig. 9). Given that a heterogeneous cellular response is typically seen in cell-based

assays for siRNA screening (Echeverri CJ, et al. 2006), these results showed that the

performance of our assay model was excellent. In addition, as I expected, no change in

cell viability (assessed by nuclei numbers) was detected in the negative or positive

controls under the tested conditions (data not shown). Based on these data, I moved

forward in developing our system for automated high-content screening.

Page 34: RNAi Screening-based Identification of USP10 as a Novel

24

Fig. 5. Adapting the assay to a 384-well microtiter plate format. YFP-Mito cells

were treated with the indicated concentrations of curcumin for 24 h. Viability and

mitochondrial aggregation ratios were quantitatively measured using the Columbus 2.3

software (upper panel). Cells were observed under an Opera microscope system. Bar,

50 µm (lower panel).

Page 35: RNAi Screening-based Identification of USP10 as a Novel

25

Fig. 6. Optimizing the assay to the 384-well microtiter plate format. YFP-Mito

cells were transfected with non-targeting siRNAs or CHOP siRNAs and further treated

with the indicated concentrations of curcumin for 12, 16 and 20 h. Viability or the

mitochondrial aggregation ratio were quantitatively measured using the Columbus 2.3

software (left). Quantitation of cell viability and mitochondrial aggregation ratio. Cell

viability or mitochondrial aggregation ratios obtained after the indicated durations of 30

µM curcurmin treatment were normalized with respect to controls (non-targeting siRNA

(siControl)- transfected cells treated with 30 µM curcumin for the corresponding time

points), as measured using the Columbus 2.3 software. * p < 0.001 vs. siControl; # p <

0.01 vs. siControl (right).

Page 36: RNAi Screening-based Identification of USP10 as a Novel

26

Fig. 7. Optimizing the assay to the 384-well microtiter plate format on the

immunofluorescence. YFP-Mito cells were transfected with non-targeting siRNAs or

CHOP siRNA and further treated with 30 µM curcumin for 12 h. Cells were observed

under an Opera microscope system. Bar, 50 µm.

Page 37: RNAi Screening-based Identification of USP10 as a Novel

27

Fig. 8. Knock-down effect of the CHOP by siRNA. YFP-Mito cells were transfected

with non-targeting siRNA or CHOP siRNA and further treated with 30 µM curcumin for

12 h. Knockdown of CHOP was confirmed by Western blotting, with α-tubulin

detected as a loading control.

Page 38: RNAi Screening-based Identification of USP10 as a Novel

28

Fig. 9. Evaluation of optimized assay conditions. YFP-Mito cells were transfected

with either 20 nM non-targeting siRNAs (negative control) or 20 nM CHOP siRNA

(positive control), incubated for 48 h, and treated with 30 µM curcumin for 12 h. (A)

Mitochondrial aggregation ratios of control wells were assessed by column scatter plot

and quantitatively measured using the Columbus 2.3 software.

Page 39: RNAi Screening-based Identification of USP10 as a Novel

29

3. siRNA-based screen to identify genes involved in curcumin-induced paraptosis

To identify genes involved in curcumin-induced paraptosis, I optimized our newly

adapted assay for HTS use, as depicted in Fig. 10, and tested it against a human

druggable subset library containing 2,732 individual siRNAs, including siRNAs against

various ion channels, proteases, phosphatases, kinases, ubiquitin conjugation factors and

GPCRs. This siRNA library was plated in nine 384-well microtiter plates. Half of the

wells of columns 1, 2, 23 and 24 contained 20 nM non-targeting siRNA (negative

control), while the other half were loaded with 20 nM CHOP siRNA (positive control).

To monitor the performance of our assay throughout the screen, I loaded columns 1 and

24 with 0.5% DMSO (v/v, untreated control) and loaded columns 2 to 23 with 30 µM

curcumin (treatment control, to induce mitochondrial aggregation). To examine the

efficiency of each siRNA, the data for mitochondrial distributions and cytotoxicity were

plotted in a scatter graph (Fig. 11). For mitochondrial aggregation, a positive response

was defined using a Z-Score percentage ratio of < -2 as a cutoff. Cytotoxic siRNAs

were defined as those that reduced the number of nuclei by death-associated detachment

Page 40: RNAi Screening-based Identification of USP10 as a Novel

30

from the plates and were filtered by setting an 80% threshold. This analysis eliminated

seven of the cytotoxic siRNAs, including those against PLK1 and PPP1CC, from the set

of candidate siRNAs (Fig. 12). Our multiparametric readout therefore yielded 38

candidate siRNAs (Fig. 12). To obtain in-depth biological information on the selected

hits, Kyoto Encyclopedia of Genes and Genomes (KEGG) database analyses were

conducted using a DAVID online analysis tool. As shown in Fig. 13, this analysis

identified 12 significantly relevant pathways; many of them, including the oxytocin

signaling pathway, calcium signaling pathway, long-term potentiation, amphetamine

addiction, gastric acid secretion, HTLV-1 infection, oocyte meiosis and the Wnt

signaling pathway, are associated with GPCR-related signaling.

Page 41: RNAi Screening-based Identification of USP10 as a Novel

31

Fig. 10. Workflow of curcumin-induced mitochondrial aggregation assay.

Page 42: RNAi Screening-based Identification of USP10 as a Novel

32

Fig. 11. Results of screening for curcumin-induced paraptosis regulators. Assay

sensitivity and performance were evaluated against a library containing 2,732 siRNAs.

(A) Scatter plot analysis of the primary HCS screening results for 2,732 siRNAs: the x-

axis indicates the order of the respective gene, and the y-axis shows the Z-scores of the

percent relative mitochondrial aggregation ratio. Thirty-eight siRNAs were selected as

having a mitochondrial aggregation ratio < -2 Z-score and a relative cell number above

80% when normalized to non-targeting siRNA (siControl)-transfected samples. Genes

that induced cytotoxicity were eliminated from the scatter list.

Page 43: RNAi Screening-based Identification of USP10 as a Novel

33

Fig. 12. Selected hits for genes that appear to be involved in curcumin-induced

mitochondrial aggregation. Lists of selected hits with Z-scores < -2.

Page 44: RNAi Screening-based Identification of USP10 as a Novel

34

Fig. 13. Enrichment of the selected hits from the primary screen. Hit lists generated

from the hit selection process were analyzed for pathway enrichment using the KEGG

pathway database. Pathways with expectation values < 0.05 and a gene-set overlap

count of three or greater are shown.

Page 45: RNAi Screening-based Identification of USP10 as a Novel

35

4. USP10 effectively inhibits curcumin-induced paraptosis in malignant breast

cancer cells.

Using the optimized assay conditions and established workflow (Fig. 10), I successfully

screened for siRNAs that altered curcumin-induced mitochondrial aggregation in YFP-

Mito cells. From the siRNAs, I identified genes that may be important to this process.

To confirm whether our selected hits demonstrated target-dependent specificity, I set

out to validate one of the identified candidate gene products. I focused our attention on

USP10 (ubiquitin specific peptidase 10, which specifically cleaves ubiquitin from

ubiquitin-conjugated protein substrates), because: 1) the USP10 siRNA demonstrated

the strongest and most consistent effects in rescuing curcumin-induced mitochondrial

aggregation in our cell-based siRNA screening; and 2) a specific inhibitor of USP10,

sparutin-1, is available to test the involvement of USP10 in mitochondrial aggregation

during paraptosis. To confirm that USP10 knockdown inhibits curcumin-induced

mitochondrial aggregation, I compared the effect of USP10 siRNA with that of CHOP

siRNA, which was used as a positive control in our screening, on curcumin-induced

Page 46: RNAi Screening-based Identification of USP10 as a Novel

36

mitochondrial aggregation. When I transfected YFP-Mito cells with USP10 siRNA and

then applied various concentrations (0-40 µM) of curcumin, I found that USP10

knockdown inhibited mitochondrial aggregation more effectively than CHOP

knockdown (Fig. 14). Representative mitochondrial aggregation results obtained from

CHOP- or USP10-knockdown cells treated with 30 µM curcumin are shown in Fig. 15

and 16. The importance of USP10 in curcumin-induced paraptosis was further

confirmed by using its inhibitor, spautin-1. While treatment of YFP-Mito cells with 30

µM curcumin for 12 h induced mitochondrial aggregation, pretreatment with spautin-1

significantly and dose-dependently inhibited the curcumin-induced morphological

changes of mitochondria (Fig. 17 and 18). In addition, spautin-1 pretreatment dose-

dependently inhibited the cell death induced by 40 µM curcumin in MDA-MB 435S

cells (Fig. 19). Interestingly, spautin-1 pretreatment decreased the cytotoxic effects of

curcumin in other breast cancer cells, including MDA-MB 231 and MCF-7 cells:

pretreatment with 5 µM spautin-1 significantly attenuated the cell death induced by 40

µM curcumin in MDA-MB 435S, MDA-MB 231 and MCF-7 cells (Fig. 20, 21 and 22).

These results indicate that USP10, which was selected from our image-based HCS as

Page 47: RNAi Screening-based Identification of USP10 as a Novel

37

the potential effector of curcumin-induced mitochondrial aggregation, may critically

contribute to curcumin-induced paraptosis by affecting the shape/cellular distribution of

mitochondria and the rate of cell death.

We recently showed that dimethoxycurcumin (DMC), a curcumin derivative in which

the phenolic-OH groups of curcumin are replaced with methoxy groups (Priyadarsini K,

et al. 2003; Kunwar A, et al. 2011), induces paraptosis more potently than curcumin

in malignant breast cancer cells (Yoon MJ, et al. 2014b). Therefore, I next examined

whether spautin-1 could also inhibit DMC-induced paraptosis. Indeed, I found that

pretreatment with spautin-1 dose-dependently inhibited the death induced by 20 µM

DMC in MDA-MB 435S cells (Fig. 23). We also recently showed that ERKs are

activated during the paraptosis induced by curcumin, DMC or celastrol (Yoon MJ, et

al. 2010; Yoon MJ, et al. 2014a; Yoon MJ, et al. 2014b). In particular, ERK

activation was found to be required for mitochondrial dilation during the progression of

curcumin-induced paraptosis (Yoon MJ, et al. 2010). Therefore, I tested whether

spautin-1 affects curcumin-induced ERK activation. western blot analysis showed that

spautin-1 treatment markedly reduced the curcumin-induced activation of ERK (Fig.

Page 48: RNAi Screening-based Identification of USP10 as a Novel

38

24), suggesting that USP10 may act upstream of ERK-mediated mitochondrial dilation

in curcumin-induced paraptosis. Collectively, our results indicate that USP10, a hit

selected from our HCS for potential effectors of curcumin-induced mitochondrial

aggregation, may play a critical role in curcumin-induced paraptotic signaling.

Page 49: RNAi Screening-based Identification of USP10 as a Novel

39

Fig. 14. Secondary screening to confirm the involvement of USP10 in curcumin-

induced mitochondrial aggregation. YFP-Mito cells were transfected with non-

targeting siRNAs, CHOP siRNAs or USP10 siRNAs and further treated with the

indicated concentrations of curcumin for 12 h. Viability and mitochondrial aggregation

ratios were quantitatively measured using the Columbus 2.3 software. * p < 0.001 vs.

siControl.

Page 50: RNAi Screening-based Identification of USP10 as a Novel

40

Fig. 15. Secondary screening to confirm the involvement of USP10 in curcumin-

induced mitochondrial aggregation on the immunofluorescence. YFP-Mito cells

were transfected with non-targeting siRNA, CHOP siRNAs or USP10 siRNA and

further treated with 30 µM curcumin for 12 h. Cells were observed under an Opera

microscope system. Bar, 50 µm.

Page 51: RNAi Screening-based Identification of USP10 as a Novel

41

Fig. 16. Knock-down effect of the USP10 by siRNA. Knockdown of USP10 was

confirmed by Western blotting, with α-tubulin detected as a loading control.

Page 52: RNAi Screening-based Identification of USP10 as a Novel

42

Fig. 17. The USP10 inhibitor, spautin-1, effectively inhibits curcumin-induced

mitochondria aggregation. YFP-Mito cells were pretreated with the indicated

concentrations of spautin-1 and further treated with 30 µM curcumin for 12 h.

Mitochondrial aggregation ratios were quantitatively measured using the Columbus 2.3

software. * p < 0.001 vs. 30 µM curcumin.

Page 53: RNAi Screening-based Identification of USP10 as a Novel

43

Fig. 18. The USP10 inhibitor, spautin-1, effectively inhibits curcumin-induced

paraptosis in malignant breast cancer cell on the immunofluorescence. YFP-Mito

cells were treated with 5 µM spautin-1 and/or 30 µM curcumin for 12 h. Cells were

observed under an Opera microscope system. Bar, 50 µm.

Page 54: RNAi Screening-based Identification of USP10 as a Novel

44

Fig. 19. The USP10 inhibitor, spautin-1, effectively inhibits curcumin-induced cell

death. MDA-MB-435S cells were pretreated with the indicated concentrations of

spautin-1 and further treated with 20 or 40 µM curcumin for 24 h. Cellular viability was

assessed using calcein-AM and EthD-1. * p < 0.001 vs. 0.5% DMSO; # p < 0.001 vs.

20 µM curcumin; † p < 0.001 vs. 40 µM curcumin.

Page 55: RNAi Screening-based Identification of USP10 as a Novel

45

Fig. 20. The USP10 inhibitor, spautin-1, effectively inhibits curcumin-induced cell

death in malignant breast cancer cell. Cells were treated with 5 µM spautin-1 and/or

40 µM curcumin for 24 h. Cellular viability was assessed using calcein-AM and EthD-1.

* p < 0.001 vs. no treatment; # p < 0.001 vs. 40 µM curcumin.

Page 56: RNAi Screening-based Identification of USP10 as a Novel

46

Fig. 21. Spautin-1 effectively inhibits curcumin or demethoxycurcumin (DMC)-

induced cytoplasmic vacuolation. MDA-MB 435S cells pretreated with or without 5

μM spautin-1 were further treated with 40 µM curcumin or 20 µM DMC for 12 h and

observed by phase-contrast microscopy. Bar, 10 µm.

Page 57: RNAi Screening-based Identification of USP10 as a Novel

47

Fig. 22. Spautin-1 effectively inhibits curcumin-induced cytoplasmic vacuolation in

various cancer cells. (B) MDA-MB 231 or MCF-7 cells pretreated with or without 5

µM spautin-1 were further treated with 40 µM curcumin for 12 h and observed by

phase-contrast microscopy. Bar, 10 µm.

Page 58: RNAi Screening-based Identification of USP10 as a Novel

48

Fig. 23. Spautin-1 effectively inhibits curcumin or demethoxycurcumin (DMC)-

induced cell death. (E) MDA-MB 435S cells were pretreated with 5 or 10 µM spautin-

1 and further treated with 40 µM curcumin or 20 µM DMC for 24 h. Cellular viability

was assessed using calcein-AM and EthD-1. * p < 0.001 vs. 0.5% DMSO; # p < 0.001 vs.

40 µM curcumin; † p < 0.001 vs. 20 µM DMC.

Page 59: RNAi Screening-based Identification of USP10 as a Novel

49

Fig. 24. The USP10 inhibitor, spautin-1, effectively inhibits curcumin-induced ERK

activation. MDA-MB 435S cells treated with 10 µM spautin-1 and/or 40 µM curcumin

for the indicated time points were subjected to Western blotting of the indicated proteins,

with α-tubulin detected as a loading control.

Page 60: RNAi Screening-based Identification of USP10 as a Novel

50

Ⅳ. DISCUSSION

Malignant cancer cells are often genetically heterogeneous and resist the induction of

apoptosis (Fombonne J, et al. 2004; Kim R, et al. 2014). Accumulating evidence

suggests that the effective elimination of malignant cancer cells may require not only

classical apoptotic pathways (mitochondrial and/or death-receptor pathways) but also

alternative apoptotic and non-apoptotic pathways (Sperandio S, et al. 2000). In

addition, researchers have speculated that the relative sensitivity of cancer cells (versus

normal cells) to oxidative and ER stress could be exploited for the rational design of

cancer therapeutics (Carew JS, et al. 2002; Galluzzi L, et al. 2010; Martinon F, et al.

2012). Thus, the induction of paraptosis, which is a cell death mode that targets both

mitochondria and the ER, may offer an attractive therapeutic strategy to effectively kill

malignant cancer cells that resist conventional pro-apoptotic cancer therapies. We

previously reported that curcumin induces extensive swelling and fusion of

mitochondria and the ER, eventually leading to the formation of a few

megamitochondria and substantially expanded ER structures (Yoon MJ, et al. 2010).

Page 61: RNAi Screening-based Identification of USP10 as a Novel

51

However, the signals responsible for the dilation of mitochondria and the ER are still

not clearly understood. Here, in an effort to decipher the key signals responsible for the

dilation of mitochondria during curcumin-induced paraptosis, I designed and performed

a multiparametric siRNA screen intended to identify genes involved in paraptosis. As

the screening strategy, I exploited multiple readouts to find the most biologically

relevant targets in curcumin-induced paraptosis: I examined mitochondrial aggregation

by using breast cancer cells that expressed fluorescence selectively in mitochondria

(YFP-Mito cells), and I measured cell viability. Cells were examined at 48 h post-

siRNA transfection, having been treated with curcumin for 12 h. The resulting dual-

readout assay was easily implemented for the screening of a druggable subset siRNA

library that comprised sub-libraries of ion channels, proteases, phosphatases, kinases,

ubiquitin conjugation proteins, and GPCRs. In our screening system, the siRNAs of

interest could be rapidly identified by their ability to regulate curcumin-induced

paraptosis (i.e., decrease curcumin-induced mitochondrial aggregation) without

generating cytotoxicity. To validate our screening method and hit- selection strategy, I

performed KEGG pathway analysis of selected hits. I found that many of the identified

Page 62: RNAi Screening-based Identification of USP10 as a Novel

52

genes were associated with GPCR signaling, including those encoding components of

oxytocin signaling, neuroactive ligand-receptor interactions, calcium signaling, long-

term potentiation and dopaminergic synapses (Figure 4B). Thus, GPCR-associated

signals may be critically linked to curcumin-induced paraptosis. I also note that

oxytocin was shown to inhibit the proliferation of human breast cancer cell lines

(Cassoni P, et al. 1994) and decrease the tumor growth of rat and mouse mammary

carcinomas (Cassoni P, et al. 1996), and that oxytocin receptor expression has been

observed in cases of primary and metastatic carcinomas of breast (Bussolati, et al.

1996). This suggests that our screening identified genes that are enriched in pathways

of interest, not just a random set of genes.

The siRNA corresponding to USP10, a member of the ubiquitin-specific protease family

of cysteine proteases, showed one of the strongest abilities to rescue curcumin-induced

mitochondrial aggregation. Deubiquitination of ubiquitin-specific peptidases (USPs) is

catalyzed by specialized thiol proteases known as deubiquitinating enzymes

(Ciechanover A, et al. 2000), which hydrolyze the amide bond between the C-terminal

glycine residue of ubiquitin and primary amino groups of the substrate protein. The

Page 63: RNAi Screening-based Identification of USP10 as a Novel

53

USPs represent the largest enzyme family in the ubiquitin system and have more than

50 members (Deng M, et al. 2016). Distinct USPs may have specialized functions

(Nijman SM, et al. 2005), although their detailed action mechanisms are not yet fully

understood. In the present study, I found that pretreatment with spautin-1, a specific

inhibitor of USP10, markedly inhibited the morphological changes of mitochondria and

subsequent paraptotic cell death induced by curcumin. We recently reported that DMC,

a methylated analog of curcumin, acts as a potent inducer of paraptosis and outperforms

curcumin in effectively reducing the clonogenicity of malignant breast cancer cells and

inhibiting tumor growth in a breast cancer xenograft model (Yoon MJ, et al. 2014b).

Here, I found that DMC-induced cytoplasmic vacuolation and subsequent cell death

were also significantly inhibited by spautin-1, suggesting that USP10 may play a critical

role in the paraptosis induced by both curcumin and by DMC. ERK signaling is known

to be important for the induction of mitochondrial vacuolation (Yoon MJ, et al. 2010).

In a previous study, We showed that curcumin transiently activates ERK1/2 and

inhibition of ERK with U0126 blocked the dilations of both mitochondria and the ER

(Yoon MJ, et al. 2010). When I examined whether USP10 affects the curcumin-

Page 64: RNAi Screening-based Identification of USP10 as a Novel

54

induced activation of ERKs, I found that spautin-1 effectively reduced the levels of

phospho-ERK (Fig. 6F). Together, our data suggest that USP10 may act upstream of

mitochondrial dilation during curcumin-induced paraptosis.

Recent studies found that USP10 deubiquitinates and stabilizes the tumor suppressor,

p53 (Yuan J, et al. 2010), thereby contributing to the DNA damage response (Yuan J,

et al. 2010). In unstressed cells, USP10 specifically deubiquitinates p53 in the

cytoplasm, counteracting the action of MDM2 and stabilizing p53 (Yuan J, et al. 2010).

Following DNA damage, USP10 translocates to the nucleus and deubiquitinates p53 to

help regulate the p53-dependent DNA damage response. AMPK (Deng M, et al. 2016),

Beclin-1 (Liu J, et al. 2011) and SIRT6 (Yuan J, et al. 2010; Lin Z, et al. 2013) have

also been proposed as substrates of USP10-mediated deubiquitination. Interestingly,

upon exposure to an oxidant, USP10 regulates cell death via its antioxidant activity

without any requirement for its deubiquitinase activity (Takahashi M, et al. 2013).

Thus, USP10 may have both deubiquitinase-dependent and -independent anti-stress

functions depending on the stimulus and cellular context. In the present study, the

protein levels of p53, BECN1, and AMPK were not markedly altered by curcumin in

Page 65: RNAi Screening-based Identification of USP10 as a Novel

55

p53-mutant MDA-MB 435S cells, nor were they affected by USP10 knockdown or

spautin-1 pretreatment (data not shown). This suggests that there are yet-unidentified

targets of USP10 in curcumin-induced paraptosis. Additional work is needed to

investigate the underlying mechanism by which USP10 contributes to mitochondrial

dilation and subsequent paraptotic cell death in malignant breast cancer cells.

Page 66: RNAi Screening-based Identification of USP10 as a Novel

56

-CONCLUSION-

I herein configured a robust and efficient high-content screening method for

curcumin-induced paraptosis by combining automated microscopy with HTS

technology. Our screening method simultaneously monitors the cytotoxicity of the

tested genes and assesses the fluorescence level to detect changes in the mitochondrial

phenotype. I miniaturized this assay to a 384-well microtiter plate format to facilitate

the screening of chemical libraries, and identified several signaling pathways that may

be involved in the development of malignant breast cancer. I believe that this assay

concept could be further developed to detect phenotypic changes in the ER, which

would enable the identification of genes that are critically involved in ER dilation. This

would further contribute to our understanding of the signaling networks of paraptosis,

which is a novel cell death mode whose biochemical features have not yet been clearly

delinated. In addition, high-content screening using our systematic assay concept may

contribute to the identification of potential targets and the development of new

therapeutic strategies for selectively killing malignant breast cancer cells.

Page 67: RNAi Screening-based Identification of USP10 as a Novel

57

REFERENCES

1. Torre, L. A., Bray, F., Siegel, R. L., et al., “Global cancer statistics, 2012,” CA

Cancer J Clin., 65(2), 87 (2015)

2. Yagata, H., Kajiura, Y., Yamauchi, H., “Current strategy for triple-negative

breast cancer: appropriate combination of surgery, radiation, and chemotherapy,” Breast

Cancer, 18(3), 165 (2011)

3. Bianchini, G., Balko, J. M., Mayer, I. A., et al., “Triple-negative breast cancer:

challenges and opportunities of a heterogeneous disease,” Nat Rev Clin Oncol., 13(11),

674 (2016)

4. Sperandio, S., de Belle, I., Bredesen, D. E., “An alternative, nonapoptotic form

of programmed cell death,” Proceedings of the National Academy of Sciences of the

United States of America, 97(26), 14376 (2000)

5. Kar, R., Singha, P. K., Venkatachalam, M. A., et al., “A novel role for MAP1

LC3 in nonautophagic cytoplasmic vacuolation death of cancer cells,” Oncogene,

28(28), 2556 (2009)

Page 68: RNAi Screening-based Identification of USP10 as a Novel

58

6. Wang, W. B., Feng, L. X., Yue, Q. X., et al., “Paraptosis accompanied by

autophagy and apoptosis was induced by celastrol, a natural compound with influence

on proteasome, ER stress and Hsp90,” J Cell Physiol., 227(5), 2196 (2012)

7. Yoon, M. J., Kim, E. H., Lim, J. H., et al., “Superoxide anion and proteasomal

dysfunction contribute to curcumin-induced paraptosis of malignant breast cancer

cells,” Free Radic Biol Med., 48(5), 713 (2010)

8. Yoon, M. J., Lee, A. R., Jeong, S. A., et al., “Release of Ca2+ from the

endoplasmic reticulum and its subsequent influx into mitochondria trigger celastrol-

induced paraptosis in cancer cells,” Oncotarget, 5(16), 6816 (2014)

9. Ghosh, K., De, S., Das, S., et al., “Withaferin A Induces ROS-Mediated

Paraptosis in Human Breast Cancer Cell-Lines MCF-7 and MDA-MB-231,” PLoS One,

11(12), e0168488 (2016)

10. Priyadarsini, K. I., Maity, D. K., Naik, G. H., et al., “Role of phenolic O-H and

methylene hydrogen on the free radical reactions and antioxidant activity of curcumin,”

Free Radical Biology and Medicine, 35(5), 475 (2003)

11. Kunwar, A., Barik, A., Sandur, S. K., et al., “Differential antioxidant/pro-

Page 69: RNAi Screening-based Identification of USP10 as a Novel

59

oxidant activity of dimethoxycurcumin, a synthetic analogue of curcumin,” Free Radic

Res., 45(8), 959 (2011)

12. Tamvakopoulos, C., Dimas, K., Sofianos, Z. D., et al., “Metabolism and

anticancer activity of the curcumin analogue, dimethoxycurcumin,” Clin Cancer Res.,

13(4), 1269 (2007)

13. Lee, D., Kim, I. Y., Saha, S., et al., “Paraptosis in the anti-cancer arsenal of

natural products,” Pharmacology & therapeutics, 162, 120 (2016)

14. Bravo, R., Gutierrez, T., Paredes, F., et al., “Endoplasmic reticulum: ER stress

regulates mitochondrial bioenergetics,” Int J Biochem Cell Biol., 44(1), 16 (2012)

15. Yoon, M. J., Kang, Y. J., Lee, J. A., et al., “Stronger proteasomal inhibition and

higher CHOP induction are responsible for more effective induction of paraptosis by

dimethoxycurcumin than curcumin,” Cell Death Dis., 5, e1112 (2014)

16. Chen, D., Dai, F., Chen, Z., et al., “Dimethoxy Curcumin Induces Apoptosis by

Suppressing Survivin and Inhibits Invasion by Enhancing E-Cadherin in Colon Cancer

Cells,” Medical Science Monitor, 22, 3215 (2016)

17. Carew, J. S., Huang, P., “Mitochondrial defects in cancer,” Mol Cancer, 1(9), 9

Page 70: RNAi Screening-based Identification of USP10 as a Novel

60

(2002)

18. Yuan, J., Luo, K., Zhang, L., et al., “USP10 regulates p53 localization and

stability by deubiquitinating p53,” Cell, 140(3), 384 (2010)

19. Lin, Z., Yang, H., Tan, C., et al., “USP10 antagonizes c-Myc transcriptional

activation through SIRT6 stabilization to suppress tumor formation,” Cell Rep., 5(6),

1639 (2013)

20. Takahashi, M., Higuchi, M., Matsuki, H., et al., “Stress granules inhibit

apoptosis by reducing reactive oxygen species production,” Mol Cell Biol., 33(4), 815

(2013)

21. Shum, D., Smith, J. L., Hirsch, A. J., et al., “High-content assay to identify

inhibitors of dengue virus infection,” Assay Drug Dev Technol., 8(5), 553 (2010)

22. Carpenter, A. E., “Image-based chemical screening,” Nature chemical biology,

3(8), 461 (2007)

23. Boutros, M., Heigwer, F., Laufer, C., “Microscopy-Based High-Content

Screening,” Cell, 163(6), 1314 (2015)

24. Loo, L. H., Wu, L. F., Altschuler, S. J., “Image-based multivariate profiling of

Page 71: RNAi Screening-based Identification of USP10 as a Novel

61

drug responses from single cells,” Nat Methods, 4(5), 445 (2007)

25. Genovesio, A., Kwon, Y. J., Windisch, M. P., et al., “Automated genome-wide

visual profiling of cellular proteins involved in HIV infection,” J Biomol Screen, 16(9),

945 (2011)

26. Mimnaugh, E. G., Xu, W., Vos, M., et al., “Endoplasmic reticulum

vacuolization and valosin-containing protein relocalization result from simultaneous

hsp90 inhibition by geldanamycin and proteasome inhibition by velcade,” Mol Cancer

Res., 4(9), 667 (2006)

27. Echeverri, C. J., Perrimon, N., “High-throughput RNAi screening in cultured

cells: a user's guide,” Nat Rev Genet, 7(5), 373 (2006)

28. Fombonne, J., Reix, S., Rasolonjanahary, R., et al., “Epidermal Growth Factor

Triggers an Original, Caspaseindependent Pituitary Cell Death with Heterogeneous

Phenotype,” Mol Biol Cell, 15(11), 4938 (2004)

29. Kim, R., Emi, M., Tanabe, K., “The role of apoptosis in cancer cell survival and

therapeutic outcome,” Cancer Biology & Therapy, 5(11), 1429 (2014)

30. Galluzzi, L., Morselli, E., Kepp, O., et al., “Mitochondrial gateways to cancer,”

Page 72: RNAi Screening-based Identification of USP10 as a Novel

62

Mol Aspects Med, 31(1), 1 (2010)

31. Martinon, F., “Targeting endoplasmic reticulum signaling pathways in cancer,”

Acta Oncol., 51(7), 822 (2012)

32. Cassoni, P., Sapino, A., Negro, F., et al., “Oxytocin inhibits proliferation of

human breast cancer cell lines,” Virchows Arch., 425(5), 467 (1994)

33. Cassoni, P., Sapino, A., Papotti, M., et al., “Oxytocin and oxytocin-analogue

F314 inhibit cell proliferation and tumor growth of rat and mouse mammary

carcinomas,” Int J Cancer, 66(6), 817 (1996)

34. Bussolati, G., Cassoni, P., Ghisolfi, G., et al., “Immunolocalization and gene

expression of oxytocin receptors in carcinomas and non-neoplastic tissues of the

breast,” Am J Pathol., 148(6), 1895 (1996)

35. Ciechanover, A., Orian, A., Schwartz, A. L., “Ubiquitin-mediated proteolysis:

biological regulation via destruction,” Bioessays, 22(5), 442 (2000)

36. Nijman, S. M., Luna-Vargas, M. P., Velds, A., et al., “A genomic and

functional inventory of deubiquitinating enzymes,” Cell, 123(5), 773 (2005)

37. Deng, M., Yang, X., Qin, B., et al., “Deubiquitination and Activation of AMPK

Page 73: RNAi Screening-based Identification of USP10 as a Novel

63

by USP10,” Mol Cell, 61(4), 614 (2016)

38. Liu, J., Xia, H., Kim, M., et al., “Beclin1 controls the levels of p53 by

regulating the deubiquitination activity of USP10 and USP13,” Cell, 147(1), 223 (2011)

Page 74: RNAi Screening-based Identification of USP10 as a Novel

64

-국문요약-

siRNA screening을 통한 paraptosis의 새로운 조절인자인

USP10 규명

아주대학교 대학원 의생명과학과

김 진 엽

(지도교수 : 최 경 숙)

Apoptosis를 표적으로 하는 치료법의 한계가 인식되면서, 그 동안 apoptosis

기전을 탐구해 오던 수많은 연구 group들이 mitotic catastrophe를 통한 세포

사멸, necroptosis, paraptosis 등 non-apoptotic cell death 분야로 연구 방향을

전환하고 있어 그 분자 기전에 대한 연구가 빠른 속도로 활성화되고 있다.

Paraptosis는 apoptosis의 형태학적, 생화학적 특징 없이 소포체와 미토콘드리

아가 동시에 팽창, 융합되면서 유도되는 세포 사멸 모드이다. 이러한

paraptosis 연구는 pro-apoptotictherapeutics의 한계점이 부각되는 최근 추세

에 따라 현행 암치료법의 한계를 극복할 수 있는 새로운 항암 치료 전략 수

립에 필요한 이론적 근거를 제시할 것으로 예측되고 있다. 따라서 본 연구는

Paraptosis 유도 과정에 중요한 역할을 하는 유전자를 발굴하고자 YFP-

mitochondria cell line을 구축한 후 Image-based phenotypic siRNA screening을

통해 분석 결과와 비교, 종합하여 curcumin에 의한 paraptosis 유도 과정에

관여하는 미토콘드리아 팽창/융합 억제 후보 유전자를 선별하였다. 이 선별

Page 75: RNAi Screening-based Identification of USP10 as a Novel

65

된 유전자들중 ubiquitin peptidase들중 하나인 USP10을 선택하였고 USP10

siRNA를 처리할 경우 curcumin에 의한 paraptosis 유도를 저해하는것을 확인

할 수 있었으며, 뿐만 아니라, USP10 inhibitor인 spautin-1을 처리할 경우

USP10 siRNA에 의한 효과와 같이 curcumin에 의한 paraptosis 유도를 저해

하는 것을 확인할 수 있었다.

이로써 본 연구의 결과는 paraptosis의 세부 분자 기전에 대한 이해의 폭을

넓힐 수 있을 뿐만 아니라, paraptosis 유도를 통해 악성 종양에서 항암 치료

내성을 극복할 수 있는 세포 사멸 유도 방안을 제시할 수 있을 것으로 기대

되므로 break-through 형 연구과제라 할 수 있다.

핵심어: siRNA screening, paraptosis, mitochondria, USP10