12
Tumor and Stem Cell Biology Establishment and Characterization of an In Vitro Model of Ovarian Cancer Stem-like Cells with an Enhanced Proliferative Capacity Tatsuya Ishiguro 1,2 , Ai Sato 2 , Hirokazu Ohata 2 , Yoshinori Ikarashi 3 , Ryou-u Takahashi 4 , Takahiro Ochiya 4 , Masayuki Yoshida 5 , Hitoshi Tsuda 5 , Takashi Onda 6 , Tomoyasu Kato 7 , Takahiro Kasamatsu 7 , Takayuki Enomoto 1 , Kenichi Tanaka 1 , Hitoshi Nakagama 8 , and Koji Okamoto 2 Abstract The establishment of cancer stem-like cell (CSC) culture sys- tems may be instrumental in devising strategies to ght refractory cancers. Inhibition of the Rho kinase ROCK has been shown to favorably affect CSC spheroid cultures. In this study, we show how ROCK inhibition in human serous ovarian cancer (SOC) cells can help establish a CSC system, which illuminates cancer patho- physiology and its treatment in this setting. In the presence of a ROCK kinase inhibitor, spheroid cultures of SOC cells expressed characteristic CSC markers including ALDH1A1, CD133, and SOX2, along with differentiation and tumorigenic capabilities in mouse xenograft models of human SOC. High expression levels of ALDH, but not CD133, correlated with spheroid formation CSC marker expression and tumor forming capability. In clin- ical specimens of SOC, high levels of ALDH1A1 correlated with advanced stage and poor prognosis. Pharmacologic or genetic blockade of ALDH blocked cell proliferation and reduced expression of SOX2, the genetic ablation of which abolished spheroid formation, whereas SOX2 overexpression inhibited ALDH1A1 expression and blocked spheroid proliferation. Tak- en together, our ndings illustrated a new method to culture human ovarian CSC, and they dened a reciprocal regulatory relationship between ALDH1A1 and SOX2, which impacts ovarian CSC proliferation and malignant progression. Cancer Res; 76(1); 15060. Ó2015 AACR. Introduction Epithelial ovarian cancer (EOC) accounts for the majority of ovarian malignancies, and is one of the major causes of cancer- related death in women (1). Although improved surgical reduc- tion followed by standard combination chemotherapy is gener- ally initially effective, a majority of patients suffer from cancer recurrence, which is less responsive to the original chemotherapy (2). Combined with the lack of reliable methods for diagnosis at the early stages of the disease, the overall prognosis of EOC remains dismal, with a 5-year survival rate of approximately 50% (3). EOC is classied into several distinct histologic subtypes (4). Among these, serous ovarian carcinoma (SOC) is one of the most common types of EOC, and there is clinical urgency to understand the specic biologic features of SOC that can be exploited for its treatment. Recently, the concept of cancer stem-like cell (CSC) has emerged as a promising theory that can explain the malignant behavior of cancer, including chemo-resistance and metastasis (5). It is postulated that the refractory nature of EOC can be explained by the existence of CSCs, as the vast intratumoral heterogeneity observed in EOC suggests that there is a small population of cells that survive initial therapy and repopulate for later recurrence (6, 7). This hypothesis has driven the attempts to isolate CSCs from EOC to characterize their bio- logic nature. Many markers for ovarian CSCs have been reported (8, 9). Pioneering works have identied tumorigenic populations from ovarian cancer based on the expression of the surface markers including CD44 (10, 11), CD117 (11), CD133 (12), CD24 (13, 14), or on the differential Hoechst-dye efux (side population; ref. 15). These variations in CSC markers may reect the heterogeneous nature of CSCs from SOC. Alterna- tively, it is possible that experimental variations, such as sources of CSCs (immortalized cell lines or passaged primary 1 Department of Obstetrics and Gynecology, Niigata University Grad- uate School of Medical and Dental Sciences, Niigata, Japan. 2 Division of Cancer Differentiation, National Cancer Center Hospital, Tokyo, Japan. 3 Central Animal Division, National Cancer Center Hospital, Tokyo, Japan. 4 Division of Molecular and Cellular Medicine, National Cancer Center Hospital, Tokyo, Japan. 5 Department of Pathology, National Cancer Center Hospital, Tokyo, Japan. 6 Department of Obstetrics and Gynecology, Kitasato University Hospital, Kanagawa, Japan. 7 Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan. 8 Division of Carcinogenesis and Cancer Prevention, National Cancer Center Research Institute, National Cancer Center Hospital, Tokyo, Japan. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). T. Ishiguro and A. Sato contributed equally to this article. Current address for H. Tsuda: Department of Pathology, National Defense Medical College, Saitama, Japan; and current address for T. Kasamatsu, Depart- ment of Gynecology, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan. Corresponding Author: Koji Okamoto, National Cancer Center Research Insti- tute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan. Phone: 81-3-3542-2511; Fax: 81-3-3542-2530; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-15-0361 Ó2015 American Association for Cancer Research. Cancer Research Cancer Res; 76(1) January 1, 2016 150 on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

Tumor and Stem Cell Biology

Establishment and Characterization of anIn Vitro Model of Ovarian Cancer Stem-like Cellswith an Enhanced Proliferative CapacityTatsuya Ishiguro1,2, Ai Sato2, Hirokazu Ohata2, Yoshinori Ikarashi3,Ryou-u Takahashi4, Takahiro Ochiya4, Masayuki Yoshida5, Hitoshi Tsuda5,Takashi Onda6, Tomoyasu Kato7, Takahiro Kasamatsu7, Takayuki Enomoto1,Kenichi Tanaka1, Hitoshi Nakagama8, and Koji Okamoto2

Abstract

The establishment of cancer stem-like cell (CSC) culture sys-temsmay be instrumental in devising strategies to fight refractorycancers. Inhibition of the Rho kinase ROCK has been shown tofavorably affect CSC spheroid cultures. In this study,we showhowROCK inhibition in human serous ovarian cancer (SOC) cells canhelp establish a CSC system, which illuminates cancer patho-physiology and its treatment in this setting. In the presence of aROCK kinase inhibitor, spheroid cultures of SOC cells expressedcharacteristic CSC markers including ALDH1A1, CD133, andSOX2, along with differentiation and tumorigenic capabilities inmouse xenograft models of human SOC. High expression levelsof ALDH, but not CD133, correlated with spheroid formation

CSC marker expression and tumor forming capability. In clin-ical specimens of SOC, high levels of ALDH1A1 correlated withadvanced stage and poor prognosis. Pharmacologic or geneticblockade of ALDH blocked cell proliferation and reducedexpression of SOX2, the genetic ablation of which abolishedspheroid formation, whereas SOX2 overexpression inhibitedALDH1A1 expression and blocked spheroid proliferation. Tak-en together, our findings illustrated a new method to culturehuman ovarian CSC, and they defined a reciprocal regulatoryrelationship between ALDH1A1 and SOX2, which impactsovarian CSC proliferation and malignant progression. CancerRes; 76(1); 150–60. �2015 AACR.

IntroductionEpithelial ovarian cancer (EOC) accounts for the majority of

ovarian malignancies, and is one of the major causes of cancer-related death in women (1). Although improved surgical reduc-tion followed by standard combination chemotherapy is gener-ally initially effective, a majority of patients suffer from cancer

recurrence, which is less responsive to the original chemotherapy(2). Combined with the lack of reliable methods for diagnosis atthe early stages of the disease, the overall prognosis of EOCremains dismal, with a 5-year survival rate of approximately50% (3).

EOC is classified into several distinct histologic subtypes (4).Among these, serous ovarian carcinoma (SOC) is one of the mostcommon types of EOC, and there is clinical urgency to understandthe specific biologic features of SOC that can be exploited for itstreatment.

Recently, the concept of cancer stem-like cell (CSC) hasemerged as a promising theory that can explain the malignantbehavior of cancer, including chemo-resistance and metastasis(5). It is postulated that the refractory nature of EOC can beexplained by the existence of CSCs, as the vast intratumoralheterogeneity observed in EOC suggests that there is a smallpopulation of cells that survive initial therapy and repopulatefor later recurrence (6, 7). This hypothesis has driven theattempts to isolate CSCs from EOC to characterize their bio-logic nature.

Many markers for ovarian CSCs have been reported (8, 9).Pioneering works have identified tumorigenic populationsfrom ovarian cancer based on the expression of the surfacemarkers including CD44 (10, 11), CD117 (11), CD133 (12),CD24 (13, 14), or on the differential Hoechst-dye efflux (sidepopulation; ref. 15). These variations in CSC markers mayreflect the heterogeneous nature of CSCs from SOC. Alterna-tively, it is possible that experimental variations, such assources of CSCs (immortalized cell lines or passaged primary

1Department of Obstetrics and Gynecology, Niigata University Grad-uate School of Medical and Dental Sciences, Niigata, Japan. 2Divisionof Cancer Differentiation, National Cancer Center Hospital, Tokyo,Japan. 3Central Animal Division, National Cancer Center Hospital,Tokyo, Japan. 4Division of Molecular and Cellular Medicine, NationalCancer Center Hospital, Tokyo, Japan. 5Department of Pathology,National Cancer Center Hospital, Tokyo, Japan. 6Department ofObstetrics and Gynecology, Kitasato University Hospital, Kanagawa,Japan. 7Department of Gynecology, National Cancer Center Hospital,Tokyo, Japan. 8Division of Carcinogenesis and Cancer Prevention,National Cancer Center Research Institute, National Cancer CenterHospital, Tokyo, Japan.

Note: Supplementary data for this article are available at Cancer ResearchOnline (http://cancerres.aacrjournals.org/).

T. Ishiguro and A. Sato contributed equally to this article.

Current address for H. Tsuda: Department of Pathology, National DefenseMedical College, Saitama, Japan; and current address for T. Kasamatsu, Depart-ment of Gynecology, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan.

Corresponding Author: Koji Okamoto, National Cancer Center Research Insti-tute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan. Phone: 81-3-3542-2511; Fax:81-3-3542-2530; E-mail: [email protected]

doi: 10.1158/0008-5472.CAN-15-0361

�2015 American Association for Cancer Research.

CancerResearch

Cancer Res; 76(1) January 1, 2016150

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 2: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

cells), growth conditions, or recipient mice, led to differences inthe published markers.

From recent studies in which tumor-initiation capability wasdirectly validated from fresh tumor samples, two markers, ALDHactivity and CD133 expression, have emerged as potent biomar-kers that can enrich cells with tumorigenic capability fromovariantumors (12, 16–18). The same studies reported that ALDH-positive cells were detectable in most of the ovarian tumorsexamined (16–18).

In fact, ALDH can be used as a CSC marker for a variety ofcancers (19). Especially, ALDH1A1 is amajor isoform responsiblefor the ALDH activity of CSCs (19), and hence one of thepromising candidates for CSC markers of ovarian cancer as wellas other cancers.

Considering the implied importance of ALDH in ovarianCSCs, it will be crucial to maintain and analyze ALDH-positiveCSCs in vitro. Although high levels of ALDH activity are asso-ciated with CSC-related features in ovarian cancer cell lines(20), there are no reports demonstrating the proliferation ofALDH-positive CSCs in in vitro culture, suggesting that there aresome unknown technical difficulties for their maintenanceunder in vitro conditions.

Previously, we demonstrated that the inhibition of Rho kinase(ROCK) markedly improved the maintenance of a spheroidculture of colon CSCs in vitro (21). The improved efficiency ofthe spheroid formation by ROCK inhibition was associated withthe sustained expression of CD44 variants (CD44v), which areknown to be among the established CSCmarkers of various typesof cancer (21).

In the present study, we found that ROCK inhibition wasessential for the spheroid formation from surgical specimen ofovarian cancer, and the established spheroids showed variouscharacteristics of CSCs. We investigated functional roles of stemcell-related markers for the maintenance of stemness of ovarianCSCs.

Materials and MethodsSpheroid culture

All procedures were performed under the protocol approvedby the Ethics Committee and the Animal Care and Use Commit-tees of the National Cancer Center or Niigata University. Tumorsamples were minced and dissociated with 150 U/mL collagenaseplus 50 U/mL hyaluronidase (Stemcell Technologies), and sequen-tially filtered through 100 mm, 70 mm, and 40 mm cell strainers(BD Falcon). After lysis of red blood cells with Red Blood CellLysis Solution (Miltenyi Biotec), the filtered cells were grown onultra-low-attachment culture dishes (Corning) in STEMPRO hESCSFM (Gibco) supplemented with 8 ng/mL bFGF (Invitrogen),penicillin/streptomycin, 20 mmol/L Y27632 (Wako), and 10 mg/mL insulin (Roche). For serial passage, spheroid cells were disso-ciated with Accumax (Innovative Cell Technologies) once every 10days. Xenograft establishment after transplantation of spheroidcells was performed as previously described (21).

Western blot analysesWestern blot analyses were performed as previously described

(21). Antibodies against SOX2, Nanog, CD44, CK7, ALDH1A1,actin, and CD133 were purchased from Cell Signaling Technol-ogy, Abcam, Thermo Scientific, Dako, Santa-Cruz Biotechnology,Sigma-Aldrich, and Miltenyi, respectively.

Immunohistochemical analysesImmunostaining of spheroid cells, mouse xenograft, or prima-

ry tumors was performed as previously described (21). Forimmunostaining of clinical specimen, SOC samples were stainedwith anti-ALDH1A1 (ab52492, Abcam), and the extent of thestaining was visually evaluated on a scale of 0 (no staining) to 3(strong staining). At least 1,000 cancer cells, not including stromalcells, were evaluated, and the mean value for each staining wascalculated.

In vitro assays for spheroid cellsCell growth or apoptosis was quantified by CellTiter-Glo Assay

(Promega) or Caspase-Glo 3/7 Assay (Promega). Limiting dilu-tion assays and qRT-PCR assays were performed as previouslydescribed (21).

Flow cytometry analysesDissociated single spheroid cells were filtered, incubated with

DAPI (BD Pharmingen) for the exclusion of nonviable cells, andused for ALDEFLUOR assays (StemCell Technologies Inc.) orstaining with the anti-CD133 antibody (CD133/1, conjugatedwith PE, Miltenyi). Samples were analyzed using a FACS Aria IIICell Sorter (BD Biosciences; ref. 21).

Lentivirus-mediated shRNA transductionLentivirus plasmids that express ALDH1A1 shRNAs (sh2:

TRCN0000276399, sh3: TRCN0000276459), SOX2 shRNAs(sh1: TRCN0000323281, sh5: TRCN0000359770), or controlshRNA were purchased from Sigma. SOX2-expressing lentiviralplasmid, pSin-EF2-SOX2-pur (22), was purchased fromAddgene. Preparation of virus-containing supernatants, virusinfection, and selection of infected cells were performed aspreviously described (23).

Luciferase assayDissociated spheroid cells were transfected using the neon

transfection system (Thermo Fisher Scientific), and luciferaseactivity was measured using dual-luciferase assay system (Pro-mega). For construction of ALDH1A1-luc, the promoter regionspanning �697 to þ315 was inserted into the KpnI-NcoI site ofpGL3-Basic (Promega).

Gene set enrichment analysesMicroarray analyses were performed using Agilent Whole

Human Genome 8 � 60 K Oligo microarrays (24). Microarraydata were accessible through Gene Expression Omnibus database(accession number GSE64999). Gene set enrichment analyseswere performed by using the gene set collection of version 3.7 ofthe Molecular Signatures Database (MSigDB).

Statistical analysesFor statistical analyses of spheroid cell experiments, either

Welch t test or Student t test was performed based on the resultsof F test. �, P < 0.05; ��, P < 0.01; ���, P < 0.001. Statistical analysesof clinical samples were performed using the GraphPad Prismver.4. (GraphPad Software Inc.). Univariate survival analysis wasperformed using the Kaplan–Meier method, and the significanceof difference between groups was examined using the log-ranktest. Multivariate survival analysis was carried out using the Coxproportional hazards regression model.

The Establishment of ALDH-Dependent Ovarian CSCs

www.aacrjournals.org Cancer Res; 76(1) January 1, 2016 151

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 3: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

ResultsSpheroid cells derived from human ovarian cancer showcharacteristics of CSCs

Recently, we established a spheroid culture of colonCSCs in thepresence of ROCK inhibitor, Y27632 (21). We attempted toestablish spheroid cultures of ovarian CSCs from surgical speci-mens by applying similar methods. We were able to establishlong-term spheroid culture in 5 out of 16 cases examined (Sup-plementary Table S1). Long-term spheroid culture was possibleonly in the presence of insulin/Y27632, suggesting that theseadditives conveyed an advantage for spheroid growth. Hematox-ylin and eosin (H&E) staining of the spheroids showedmorphol-ogy reminiscent of floating aggregates of serous ovarian cancer inascites (Fig. 1A; Supplementary Fig. S1A; ref. 25). The morpho-logic resemblance may reflect functional similarity with CSCs, aswe could establish the spheroid culture from cancerous ascites butnot from primary tumors in three cases (Supplementary Table S1,SOC-#4, #5, #9).

Next, we examined CSC-related features of the establishedspheroid cells. Serially diluted cells (#4) were used for tumorformation experiments upon transplantation into immuno-compromised NOG mice. In agreement with previous studies

(16), 1 � 104 cells were capable of forming xenograft tumors inthree out of six cases after the injection into mammary pad orflank (Supplementary Table S2). Another spheroid (#5) was alsocapable of forming xenograft tumors in all 16 cases, in which1 � 106 cells were injected at the flank of the recipient mice(data not shown). Immunostaining with Pax8, CK7, CA125, orHE4 (26), revealed that the formed xenograft tumors wereindistinguishable from the original tumors (Fig. 1B; Supplemen-tary Fig. S1B and S1C). Spheroid cells underwent differentia-tion and showed epithelial-like morphology if grown attachedin the presence of serum (Fig. 1C). Expression of ALDH1A1 andthe stem cell–related factors (Nanog and SOX2) was reduced,whereas that of the differentiation marker (CK7) was inducedat the protein level (Fig. 1D; Supplementary Fig. S1D).

The reduction of ALDH1A1 and SOX2 after differentiation wascorroborated at the RNA level (Fig. 1E; Supplementary Fig. S1E).Gene set enrichment analyses revealed that undifferentiatedspheroid cells preferentially express stem cell–related genes ofbreast cancer (Fig. 1F; ref. 27), whichmay reflect similarity of geneexpression profile of ovarian and breast CSCs (28, 29). Collec-tively, these results indicate that the spheroid cells show char-acteristics of CSCs.

A

C D

Brightphase

H&E

#4#2 #5

E

F

B

CK7

Nanog

SOX2

ALDH1A1

dif

f.

Sp

her

oid

Actin

dif

f.

Sp

her

oid

#2 #4

FDR q-value: 0.013NES: 2.085

Diff.Spheroid

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

NanogALDH1A1 SOX2 CK7

#4

Rel

ativ

e R

NA

leve

ls

00.5

11.5

22.5

33.5

44.5

Spheroid Diff.

#2

Rel

ativ

e R

NA

leve

ls

ALDH1A1 Nanog SOX2 CK7

Pax8 CK7 CA125 H&E4H&E

Xenografttumor

Primarytumor

Spheroidconditions

Differentiationconditions

#2

#4

***

**

***

***

*** ***

***

n.s.

Figure 1.Spheroid cells derived from human ovarian cancer show CSC characteristics. A, top, bright-phase image of the indicated spheroids. Scale bars, 100 mm.Bottom, H&E staining of the spheroids. Scale bars, 20 mm. B, immunostaining of xenograft tumor, 5 months after transplantation (#2). Scale bars, 20 mm.C, bright-phase images of undifferentiated spheroid cells (#2 and #4) and the cells grown under differentiation conditions. Scale bars, 100 mm. D, Western blotanalyses of the cells shown in C. E, qRT-PCR analyses of the spheroid cells (#2, #4). F, gene set enrichment analyses of gene expression profiles betweenundifferentiated and differentiated cells.

Ishiguro et al.

Cancer Res; 76(1) January 1, 2016 Cancer Research152

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 4: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

Expression of CSC/stem cell–related factors is induced byROCK inhibition in ovarian spheroids

Next, we examined the spheroid proliferation in the presence orabsence of insulin and/or Y27632, and found that the spheroidproliferation was markedly inhibited in the absence of Y27632but not insulin in two out of these three spheroids we examined(#2 and #5, Fig. 2A and B). The positive effect of Y27632 on theproliferation of the #4 spheroid was not clear over one week(Fig. 2A and B), but extended culture over one month revealedpreferential proliferation (>10-fold) in the presence of Y27632(data not shown). The inhibition of the proliferation in theabsence of Y27632 was associated with an increase in caspaseactivity (Fig. 2C), indicating that enhanced apoptotic cell deathcontributes to the growth inhibition.

In contrast to the colon CSCs (21), CD44v levels were notaugmented by Y27632 in any of the ovarian spheroid cells(Fig. 2D). Instead, two CSC markers (ALDH1A1 and CD133)and stem cell–related regulators (SOX2, Nanog, and Oct-3/4)were induced by the ROCK inhibitor in #2 and #5, but not in #4(Fig. 2D). In thepresence of ROCK inhibitor, a substantial fractionof #2 and #5 spheroid cells showed high levels of ALDH activityand CD133 (Fig. 2E and F), and RNA levels of ALDH1A1 andSOX2 were significantly higher in #2 and #5 than in #4 (Fig. 2G).

Thus, enhanced spheroid proliferation by the ROCK inhibitor in#2 and #5 was correlated with the robust induction of ALDH1,CD133, and the stem cell markers. The overall expression profileof #2 and #5 was in striking contrast with #4, which expressedhigh levels of CD44v but showed hardly detectable expression ofALDH1A1 or CD133 (Fig. 2D and G).

High levels of ALDHexpression are associatedwithCSC-relatedcharacteristics of ovarian spheroids

Recent studies indicate that ALDH and CD133 can enrich cellswith tumorigenic capability from ovarian tumors (12, 16–18).Hence, we focused on spheroid cells with high levels of ALDHactivity and CD133 expression (#2 and #5). First, we purifiedspheroid cells that express either high or low/undetectable levelsof ALDH activity (ALDHhigh and ALDHlow) by flow cytometry(Supplementary Fig. S2A). The ALDHhigh cells expressed higherlevels of the stem cell–related markers (SOX2, Oct-3/4, andNanog) than ALDHlow cells (Fig. 3A). The ALDHhigh cells weremore efficiently capable of proliferating as spheroids thanALDHlow cells (Fig. 3B and C). Levels of caspase activity in theALDHlow cells were higher than those in ALDHhigh cells (Supple-mentary Fig. S2B). Furthermore, ALDHhigh cells, but not ALDHlow

cells, could generate xenograft tumors in immuno-compromized

D

CBA

#5

#4

#2

Y27632+

Insulin+ -

#4

0

1

2

3

4

5

6

0

2

4

6

8

10

12

0

1

2

3

4

5

Rel

ativ

e ce

ll g

row

th

#5#2

+Y27632-Insulin

-Y27632-Insulin

+Y27632+Insulin

-Y27632+Insulin

0 1 4 70 1 4 70 1 4 7

#4#2#50

0.2

0.4

0.6

0.8

1

1.2

ALDH1A1

0

0.2

0.4

0.6

0.8

1

1.2

#4#2#5

SOX2

Rel

ativ

e R

NA

leve

lsR

elat

ive

RN

A le

vels

E#2 #5 #4

ALDH1A1

SOX2

CD44

Y27632+

insulin− + − + − +

CD133

Nanog

Oct-3/4

Actin

F

7.1%0.1%

0.2% 0.7%

0.2%

Anti-CD133IgG

22.9%

#5

#4

#2

CD133

#5

#4

#2

0.0%

7.5%

86.5%

0.0%

51.3%

50 mmol/L DEAB + −

ALDH activity

0.0%

G

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

1.8

2

Rel

ativ

e ca

spas

eac

tivi

ty

+Y27632 + Insulin

-Y27632 + Insulin

#2 #5 #4

(Days)

*** ***

n.s.

Figure 2.High levels of ALDH expression are maintained by ROCK inhibition and associated with proliferation ovarian spheroids. A, time course analyses of cell growthof the indicated spheroids in the presence or absence of Y27632 and/or insulin. B, bright-phase images of spheroids. Scale bars, 100 mm. C, caspase assaysof spheroid cells. The #2 spheroid cells were used for normalizing caspase activity. D,Western blot analyses of the spheroids. E, FACS analyses of ALDH activity afterALDEFLOUR staining in the presence (left) or absence (left) of diethylaminobenzaldehyde (DEAB). F, FACS analyses of CD133 expression after staining withcontrol IgG or anti-CD133 antibody (AC133). G, qRT-PCR analyses of ALDH1A1 and SOX2 in the presence of Y27632þinsulin.

The Establishment of ALDH-Dependent Ovarian CSCs

www.aacrjournals.org Cancer Res; 76(1) January 1, 2016 153

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 5: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

mice (Fig. 3D) with histologic features similar to the originaltumor (Supplementary Fig. S1C).

Next, we purified spheroid cells that express either high or low/undetectable levels of CD133 (CD133high andCD133low) by flowcytometry (Supplementary Fig. S2C). There was no significantdifference in expression of ALDH1A1 and the stem cell–relatedfactors (Supplementary Fig. S2D) or in spheroid formation (Sup-plementary Fig. S2E). The CD133high cells proliferated slowerthan CD133low cells (Supplementary Fig. S2F).

In order to determine whether CD133 expression affects theenrichment of ovarian CSCs by ALDH expression, the ALDHhigh/CD133high, ALDHlow/CD133high, ALDHhigh/CD133low, andALDHlow/CD133low cells from the #5 spheroids were purified byFACS sorting (Fig. 3E), and used to examine CSC-related features.In agreement with Fig. 3B and C, ALDHhigh/CD133high andALDHhigh/CD133low cells, but not ALDHlow/CD133high andALDHlow/CD133low cells, were capable of efficiently formingspheroids (Fig. 3F), and the cells with higher ALDH activityproliferated faster than those with lower ALDH activity (Fig.3G). Serial dilution assays indicated that the spheroid formationefficiencies of the ALDHhigh/CD133high and ALDHhigh/CD133low

cells were comparable, andwere significantly higher than those of

the ALDHlow/CD133high or ALDHlow/CD133low cells (Supple-mentary Table S3).

Of note, the ALDHhigh/CD133low cells rather grew faster thanthe ALDHhigh/CD133high cells in spheroid culture in vitro(Fig. 3G). In accordance, although both cells were capable offorming tumors in mouse xenograft assays, the tumors formedafter injection of the ALDHhigh/CD133low cells were bigger thanthose of the ALDHhigh/CD133high cells (Fig. 3H), suggesting thatCD133 marks cells with slower proliferation late. The fasterproliferation of the ALDHhigh/CD133low cells may not be attrib-uted to the difference in reduced apoptotic cell death as these cellsshowed similar caspase activities (Supplementary Fig. S2G).

ALDH1 expression is associated with high tumorigenicity andpoor prognosis in human serous ovarian cancer

Next, we compared ALDH1A1 immunostaining of the spher-oid cells with that from the original cancer. In accordance withdata from FACS analyses (Fig. 2E), the ALDH1A1 immunos-taining showed the expression of ALDH1A1 in #2 and #5, butnot in #4 (Fig. 4A, right). The staining of the original tumorindicated a similar tendency for ALDH1A1 expression (Fig. 4A,left).

10 mm

10 mm

BA C

FH

ALDHlow/CD133low ALDHhigh/CD133low

ALDHlow/CD133high ALDHhigh/CD133high

ALDHlow/CD133low ALDHlow/CD133high

ALDHhigh/CD133low ALDHhigh/CD133high

Rel

ativ

e ce

ll g

row

th

0 4 8 (Days)

1

0

2

3

5

4

G

ALDHhigh/CD133high

ALDHhigh/CD133low

ALDHlow ALDHhigh

#2

#51

0

2

3

0 4 8 (Days)

ALDH high

ALDHlow

Rel

ativ

e ce

ll g

row

th

#2

ALDH1A1

SOX2

Actin

#2 #5

Oct-3/4

Nanog

CK7

D

ALDHlow

ALDHhigh

H&E

0 4 8 (Days)

1

0

2

3

Rel

ativ

e ce

ll g

row

th

5

4

#5

****

E

2.0%P1110

510

410

310

2

102

103 10

410

5

P12 P10

P924.8%

5.1% 43.1%

ALDH activity

CD

133

Figure 3.High levels of ALDH activity are associated with CSC-related characteristics. A, Western blot analyses of ALDHhigh and ALDHlow cells. B, bright-phase imagesof the spheroid formation (7 days after in vitro cultivation). Scale bars, 100 mm. C, time course analyses of cell growth. D, left, xenograft tumor formationof ALDHhigh and ALDHlow cells (#5, 2� 105 cells, n¼ 4), 5 months after transplantation. Right, H&E staining of xenograft tumor derived from ALDHhigh cells. E, FACSanalyses of the spheroid cells (#5) for expression of ALDH activity and CD133. Sorting gates for the ALDH/CD133 quadrants are shown as rectangles (8 daysafter in vitro cultivation). F, bright-phase images of the spheroid formation of the FACS-sorted cells. Scale bars, 100 mm. G, time course analyses of cell growth of thesorted cells. H, xenograft tumor formation of the ALDHhigh/CD133high and ALDHhigh/CD133low cells 4 months after transplantation.

Ishiguro et al.

Cancer Res; 76(1) January 1, 2016 Cancer Research154

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 6: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

Examination of the ALDH1A1 staining in early and advancedstage SOC (Supplementary Table S4) revealed that ALDH1A1expression was markedly augmented at the advanced stage(Fig. 4B). Statistical studies of the 90 cases of advanced stagehigh-grade SOC indicated that although there was no associationbetween ALDH1A1 expression and cilinicopathologic parameters(Supplementary Table S5A), high levels of ALDH1A1 expressionwere associatedwith poor prognosis (Fig. 4C andD).Multivariateanalyses of the clinical data indicated that ALDH1A1 stainingserved as an independent prognostic factor (Supplementary TableS5B). Thus, in agreement with the meta-analyses of the previousstudies (30), high levels of ALDH1A1 expression are associatedwith cancer progression and poor prognosis.

ALDH activity is required for SOX2 expression andproliferation of ovarian CSCs

Next, we investigated functional importance of ALDH activityfor ovarian CSCs. Disulfiram (DS), an ALDH inhibitor, was usedto block ALDH activity in #2 and #5 that express high levels ofALDH. The DS treatment caused partial inhibition of ALDH

activity in both spheroids (Fig. 5A; Supplementary Fig. S3A).Nevertheless, the ALDH inhibition almost completely blockedthe formation and proliferation of spheroids (Fig. 5B and C;Supplementary Fig. S3B and S3C), and induced apoptotic celldeath (Fig. 5D).Of note, the inhibition of ALDHactivity led to thereduction of SOX2 expression (Fig. 5E; Supplementary Fig. S3D),while suppression of the other stemmarkers, Oct-3/4 and Nanog,was not clear (data not shown). The DS treatment did notsignificantly affect the expression of SOX2 RNA (Fig. 5F; Supple-mentary Fig. S3E). Presumably the reduction of SOX2 was notattributed to the accelerated apoptotic cell death of SOX2-expres-sing cells, because caspase inhibitors did not restore SOX2 expres-sion in the presence of DS, while they suppressed caspase acti-vation (Fig. 5G; Supplementary Fig. S3F) and restored cell via-bility (Supplementary Fig. S3F).

ALDH1A1 mediates SOX2 expression and proliferation ofovarian CSCs

In order to determine whether ALDH1A1 is responsible for theproliferation and SOX2 expression, we inhibited ALDH1A1

B

C

HR2.192(95% CI 1.323–4.961)

Score: 0

Sco

re

0Early Advanced

1

2

3

Score: 1 Score: 2 Score: 3

Spheroids

#2

#4

#5

Primarytumor

A

D

P < 0.01

P = 0.0053

HR1.872(95% CI 1.178–3.643)

P = 0.0114

ALDH high

ALDH low

ALDH high

ALDH low

Ove

rall

surv

ival

Pro

gre

ssio

n-f

ree

surv

ival

Days

Days

00

25

50

75

100

1,000 2,000 3,000 4,000 5,000

00

25

50

75

100

1,000 2,000 3,000 4,000 5,000 6,000

Figure 4.ALDH1 expression is associated with advanced clinical stages and poor prognosis in human serous ovarian cancer. A, immunostaining of ALDH1A1 of the indicatedspheroids (right) and the correspondingprimary tumors (left). Scale bars, 50mm.B, top, representative immunostainingofALDH1A1 (score0–3). Bottom, distributionof ALDH1A1 expression in early (FIGO stages I and II; n ¼ 23) and advanced (FIGO stages III and IV; n ¼ 90) stages of SOCs. Horizontal lines, average values.C and D, Kaplan–Meier analyses of overall (C) and progression-free (D) survival of patients suffering from the advanced stages of SOCs. The patients were stratifiedinto high (n ¼ 62; solid lines) and low (n ¼ 28; dotted lines) expression groups according to ALDH1A1 expression.

The Establishment of ALDH-Dependent Ovarian CSCs

www.aacrjournals.org Cancer Res; 76(1) January 1, 2016 155

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 7: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

expression by shRNA lentiviral transfer (Fig. 6A). Although theshRNA introduction caused apparent inhibition of ALDH1A1in both #2 and #5 (Fig. 6B and Supplementary Fig. S4A),significant ALDH activity was still observable in #5, but notin #2 after the inhibition of ALDH1A1 (Fig. 6C; SupplementaryFig. S4B), suggesting that other isoforms contribute to ALDHactivity in #5.

Microarray data indicated that, in addition to ALDH1A1, therewere several ALDH isoforms that were expressed in #5 with levelsrelatively higher than in #2 or #4 (Supplementary Fig. S4C). Ofthose,Western blot analyses of four isoforms (1A2, 3A2, 5A1, and18A1) revealed that ALDH1A2 and ALDH3A2 were expressed inthe ALDHhigh cells at levels higher than in the ALDHlow cells(Supplementary Fig. S4D). Thus, these isoforms may also con-tribute to the ALDH activity in #5.

The inhibition of ALDH1A1 blocked the formation and pro-liferation of the spheroids (Fig. 6D and E; Supplementary Fig. S4Eand S4F), induced apoptotic cell death (Fig. 6F), and suppressedspheroid forming efficiency (Supplementary Table S6), indicatingthat ALDH1A1 was required for the survival and proliferation ofALDH-positive CSCs. In accordance with SOX2 suppression byDS (Fig. 5E; Supplementary Fig. S3D), the inhibition of ALDH1A1suppressed SOX2 expression in both spheroids (Fig. 6B; Supple-mentary Fig. S4A). It is likely that the suppression of SOX2 byALDH1A1 was mainly caused at the posttranscriptional level,

because DS treatment or the inhibition of ALDH1A1 caused onlymodest effects on SOX2 RNA expression (Fig. 5F; SupplementaryFigs. S3E and S4G).

The proliferation of ovarian CSCs is regulated throughreciprocal regulation by ALDH1A1 and SOX2

Because SOX2 expression was associated with ALDH-depen-dent spheroid proliferation (Figs. 5E and 6B; SupplementaryFigs. S3D and S4A), we examined whether the proliferation ofthe spheroid cells was also dependent on SOX2. shRNA-mediatedinhibition of SOX2 (Fig. 7A; Supplementary Fig. S5A) markedlysuppressed formation and proliferation of spheroids (Fig. 7B andC; Supplementary Fig. S5B and S5C), and spheroid formingefficiency (Supplementary Table S7), while the inhibition aug-mented apoptotic cell death (Fig. 7D). Thus, SOX2, as well asALDH1A1, is required for the survival and proliferation of ovarianCSCs.

In order to determine whether elevated levels of SOX2 affectthe proliferation of the ovarian CSCs, we introduced exogenousSOX2 into the #5 spheroid cells via lentivirus-mediated genetransfer (Fig. 7E). Unexpectedly, SOX2 overexpression caused areduced proliferation (Fig. 7F) and enhanced apoptotic celldeath (Supplementary Fig. S5D). In the presence of DS, SOX2overexpression further reduced proliferation and spheroid for-mation (Fig. 7F and G).

BA

E

DC

0

0.2

0.4

0.6

0.8

1

1.2

1.4

Control

20 µµmol/L DS

SOX2ALDH1A1

2 5 200 DS (mmol/L)

ALDH1A1

Actin

SOX2

86.7%

20 mmol/L DS

Control

47.5%

ALDH activity

0

1

2

3

DSControl

Rel

ativ

e ca

spas

e ac

tivi

ty

F

Control

20 mmol/L DS

2 mmol/L DS

G

DM

SO

zVA

D-F

MK

DE

VD

-CH

O

Active caspase-3

Actin

SOX2

ALDH1A1

Nanog

DSDMSO

DM

SO

zVA

D-F

MK

DE

VD

-CH

O

Rel

ativ

e R

NA

leve

ls

***

n.s.

*

Rel

ativ

e ce

ll g

row

th

Control

20 µmol/L DS2 µmol/L DS

0

1

2

3

4

5

6

7

840 (Days)

*** ***

Figure 5.ALDH activity is required for SOX2 expression and proliferation of ovarian CSCs (#5). A, FACS analyses of ALDH activity in the presence or absence of DS (2 daysafter the treatment). B, bright-phase images of spheroids (4 days after the DS treatment). Scale bars, 100 mm. C, time course of cell growth of the spheroids.D, caspase activity of the DS-treated spheroid cells. E, Western blot analyses of the spheroids at 2 days after the treatment with the indicated amounts of DS.F, qRT-PCR analyses of ALDH1A1 and SOX2 in the spheroids. G, Western blot analyses of the DS-treated spheroid cells in the presence or absence of caspaseinhibitors. The cells were preincubated with 100 mmol/L zVAD-FMK (Abcam), 25 mmol/L DEVD-CHO (Calbiochem), or DMSO for 2 hours and incubated for2 days in the presence or absence of 20 mmol/L DS.

Ishiguro et al.

Cancer Res; 76(1) January 1, 2016 Cancer Research156

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 8: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

Notably, SOX2 overexpression totally abolished ALDH1A1expression (Fig. 7E), which is likely to contribute to the inhi-bition of the CSC proliferation. SOX2 overexpression in the #4cells also resulted in the suppression of the proliferation andALDH1A1 inhibition (Supplementary Fig. S5E and S5F). SOX2regulated the transcriptional levels of ALDH1A1, because theSOX2 overexpression markedly inhibited ALDH1A1 RNAexpression (Fig. 7H; Supplementary Fig. S5G) and suppressedthe promoter activities in transient luciferase assays (Fig. 7I).Collectively, the elevated expression of SOX2 transcriptionallyblocks ALDH1A1 expression while ALDH1A1 is required forSOX2 expression.

DiscussionROCK is involved in regulation of cytoskeletons via the phos-

phorylation of its targets (31), and can affect proliferation ofnormal stem cells and CSCs: its inhibition facilitates proliferationof embryonic stem cells via suppression of dissociation-inducedapoptosis (32, 33) and of colon CSCs via induction of CD44v(21).Here, we demonstrated that ROCK inhibition also promotesproliferation of a subset of ovarian CSCs through induction ofALDH1A1 and the stem cell–related factors including SOX2.However, in contrast to colon CSCs, ROCK inhibition-dependent

proliferation in ovarian CSCs was not associated with CD44vinduction (Fig. 2D).

These discoveries led to the establishment of an in vitroculture of ovarian CSCs that are positive for ALDH1A1 andCD133. In contrast to the #2 and #5 spheroids, the #4 spher-oids showed high levels of CD44v but negligible levels ofALDH1A1 and CD133. Considering that CD44 is reported asanother CSCmarker (10, 11), there may be two types of ovarianCSCs: one is positive for ALDH and CD133, and the other forCD44.

The investigation of ALDH-positive spheroid cells revealed thatthese cells meet critical criteria for CSCs, including spheroidformation, the expression of stem cell markers, and the potentialto differentiate, and most importantly, the ability to form tumorsthat are histologically identical to the original primary tumor. Onthe other hand, CD133-positive cells showed rather slower pro-liferation both in vitro and in vivo. Previous reports suggest that theALDHþ/CD133þ cells represent cells with tumor-forming capa-bility (17, 18). The difference may be attributed to the heteroge-neity of the clinical sample, the difference in anti-CD133 anti-bodies used for the experiment, or subtle differences in experi-mental conditions.

Meta-analyses of recent studies indicated that elevated ALDHexpression was associated with poor prognosis in patients with

D

B CA

Co

ntr

ol

AL

DH

1A1-

sh2

AL

DH

1A1-

sh3

ALDH1A1

SOX2

Actin

E

PuromycinselectionInfection

Growth curve

0 days 7days

↑FACS

↑Western

d0 d1 d3 d7

↑Photo

0

1

2

3

Control

ALDH1A1-sh3

ALDH1A1-sh2

Rel

ativ

e ce

ll g

row

th

0 1 4 7 (Days)

47.4%

87.6%

37.7%

Control

ALDH1A1-sh2

ALDH1A1-sh3

ALDH activity

F

0

1

2

3

Rel

ativ

e ca

spas

eac

tivi

ty

sh2 sh3Controlsh-ALDH1A1

Control

ALDH1A1-sh2

ALDH1A1-sh3

***

***

*** ***

Figure 6.ALDH1A1 is required for SOX2 expression and proliferation of ovarian CSCs (#5). A, an experimental scheme. B, Western blot analyses after infection of theindicated lentiviruses. C, flow cytometry analyses of ALDH activity of the infected cells. D, bright-phase images of the infected cells. Scale bars, 100 mm.E, time course of the proliferation of the infected cells. F, caspase activity of the infected cells (day 2).

The Establishment of ALDH-Dependent Ovarian CSCs

www.aacrjournals.org Cancer Res; 76(1) January 1, 2016 157

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 9: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

ovarian cancer (30, 34–37), although some reports indicateotherwise (38). Our data on immunostaining of high-grade SOCdemonstrated that higher levels of ALDH1A1 are associated withan advance stage and poor prognosis, warranting the furtherinvestigation of ovarian cancer and CSCs that are positive forALDH.

SOX2 plays an essential role in CSCs (39–41) and associatedwith the malignant progression of ovarian cancer (42). Func-tional analyses by gene knockdown and a chemical inhibitorrevealed that ALDH and SOX2 are essential for the proliferationof ovarian CSCs. Of note, both ALDH1A and SOX2 expressioncontribute to chemo-resistance of ovarian cancer cell lines(35, 43–45). Therefore, SOX2 and ALDH may mediate itscapability to confer chemo-resistance to ovarian CSCs as wellas stem cell–related features.

Remarkably, our data revealed that expression of ALDH1A1and SOX2 affected each other: ALDH1A1 was required for SOX2expression, whereas SOX2 overexpression caused ALDH1A1 inhi-bition. These observations, combined with the essential roles ofthese regulators in spheroid proliferation, suggest an existence of afeedback regulation for CSC proliferation, in which SOX2 andALDH1A1 form a negative feedback loop (SupplementaryFig. S5H). The potential feedback regulation between SOX2 andALDH1A1 may be beneficial for the survival and proliferation ofovarian CSCs due to reduced perturbations of levels of theseregulators.

In humans, ALDH family is composed of 19 isoforms withsimilar catalytic functions (46). Although ALDH1A1 is regardedas a major form responsible for ALDH activity (19), other iso-forms (ALDH1A3, ALDH3A2, and ALDH7A1) are also overex-pressed in ovarian cancers (47). In fact, our data indicated thatALDH1A2 and ALDH3A2, in addition to ALDH1A1, may con-tribute to ALDH activity in #5. Future studies will clarify thepotential contribution of these isoforms to ovarian CSCs.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: T. Ishiguro, H. Ohata, Y. Ikarashi, T. Enomoto,K. Tanaka, K. OkamotoDevelopment of methodology: T. Ishiguro, A. Sato, H. Ohata, R.-u TakahashiAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): T. Ishiguro, A. Sato, M. Yoshida, T. OndaAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): T. Ishiguro, H. Ohata, R.-u Takahashi, M. Yoshida,T. EnomotoWriting, review, and/or revision of the manuscript: T. Ishiguro, A. Sato,H. Ohata, Y. Ikarashi, M. Yoshida, H. Tsuda, T. Onda, T. Kato, T. Enomoto,H. Nakagama, K. OkamotoAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): T. Ochiya, M. Yoshida, H. Tsuda, T. Kato,T. Kasamatsu, H. NakagamaStudy supervision: T. Enomoto, K. Tanaka, H. Nakagama, K. Okamoto

A

Co

ntr

ol

SO

X2-

sh1

SO

X2-

sh5

SOX2

ALDH1A1

Actin

0

1

2

3

4

5

Control

SOX2-sh5

SOX2-sh1

Rel

ativ

e ce

ll g

row

th

0 1 4 7 (Days)

CB

0

1

2

3

4

5

6

7

8

sh1 sh5controlsh-SOX2

Rel

ativ

e ca

spas

e ac

tivi

ty

D E

GF

ALDH1A1

SOX2

Oct-3/4

Nanog

CK7

pS

in-c

on

tro

l

pS

in-S

ox2

Actin

#5

Rel

ativ

e ce

ll g

row

th

(Days)

0

1

2

3

4

5

6

7

840

Control (-DS)

Sox2 (-DS)

Control (+DS)

Sox2 (+DS)

Control

SOX2-sh5

SOX2-sh1

Control

Sox2

DSControl

IH

0

2

4

6

8

10

12

14

16

Rel

ativ

e lu

cife

rase

act

ivit

y

Effector

ReporterpGL3Basic

pGL3Basic

ALDHLuc

ALDHLuc

Cont ContSOX2 SOX20

0.2

0.4

0.6

0.8

1

1.2

Rel

ativ

e R

NA

leve

ls

So

x2

Co

ntr

ol

ALDH1A1

***

****

***

***

*** ***

Figure 7.Reciprocal regulation of ALDH1A1 and SOX2 are involved in control of CSC proliferation. A, Western blot analyses of the spheroid cells (#5) after infection ofcontrol lentiviruses or the viruses expressing SOX2 shRNAs. B, bright-phase images of the infected spheroid cells. Scale bars, 100 mm. C, time course ofproliferation of the cells shown in B. D, caspase activity of the infected cells (day 2). E, Western blot analyses of SOX2-introduced cells. F, time courseof the proliferation of the infected cells in the presence or absence of DS. G, bright-phase images of the cells shown in F (4 days after DS treatment).H, qRT-PCR analyses of ALDH1A1 in cells shown in E. I, Inhibition of the ALDH1A1 promoter by SOX2 in transient reporter assays. The indicated vectors werecotransfected into #5 for 48 hours and luciferase assays were performed.

Ishiguro et al.

Cancer Res; 76(1) January 1, 2016 Cancer Research158

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 10: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

AcknowledgmentsThe authors thank Hiroaki Sakai, Rui Uchino, and Katsuhide Ikeda for

technical assistance and Takashi Kohno and Hitoshi Ichikawa for help withthe genome analyses.

Grant SupportThis research was supported by a Grant-in-Aid for Scientific Research in

Innovative Areas fromMEXT (K. Okamoto), a Grant-in-Aid for Cancer Researchfrom the Foundation for Promotion of Cancer Research (K. Okamoto), a Grant-in-Aid for Scientific Research (B) from the Japan Society for the Promotion ofScience (JSPS; K. Okamoto), a Research Resident Fellowship from Grant-in-Aid

for Young Scientists (B) from JSPS (H.Ohata), National Cancer Center Researchand Development Fund (25-A-2, 25-B-6; K. Okamoto and H. Ohata), a HealthLabour Sciences Research Grant from The Ministry of Health, Labour andWelfare (K. Okamoto and H. Ohata), and a Grant-in Aid from Takeda ScienceFoundation (H. Ohata).

The costs of publication of this article were defrayed in part by the paymentof page charges. This article must therefore be hereby marked advertisementin accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received February 4, 2015; revised September 14, 2015; accepted October 2,2015; published OnlineFirst December 15, 2015.

References1. Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin

2014;64:9–29.2. Martin LP, Schilder RJ. Management of recurrent ovarian carcinoma:

current status and future directions. Semin Oncol 2009;36:112–25.3. Kurman RJ, Shih Ie M. The origin and pathogenesis of epithelial ovarian

cancer: a proposed unifying theory. Am J Surg Pathol 2010;34:433–43.4. Kaku T, Ogawa S, Kawano Y, Ohishi Y, Kobayashi H, Hirakawa T, et al.

Histological classification of ovarian cancer. Med Electron Microsc2003;36:9–17.

5. Clevers H. The cancer stem cell: premises, promises and challenges.Nat Med 2011;17:313–9.

6. Choi YP, Shim HS, Gao MQ, Kang S, Cho NH. Molecular portraits ofintratumoral heterogeneity in human ovarian cancer. Cancer Lett 2011;307:62–71.

7. Abelson S, Shamai Y, Berger L, Shouval R, Skorecki K, Tzukerman M.Intratumoral heterogeneity in the self-renewal and tumorigenic differen-tiation of ovarian cancer. Stem Cells 2012;30:415–24.

8. Foster R, Buckanovich RJ, Rueda BR. Ovarian cancer stem cells: workingtowards the root of stemness. Cancer Lett 2013;338:147–57.

9. ShahMM, Landen CN. Ovarian cancer stem cells: are they real and why arethey important? Gynecol Oncol 2014;132:483–9.

10. Alvero AB, Chen R, Fu HH, Montagna M, Schwartz PE, Rutherford T,et al. Molecular phenotyping of human ovarian cancer stem cellsunravels the mechanisms for repair and chemoresistance. Cell Cycle2009;8:158–66.

11. Zhang S, Balch C, Chan MW, Lai HC, Matei D, Schilder JM, et al. Iden-tification and characterization of ovarian cancer-initiating cells from pri-mary human tumors. Cancer Res 2008;68:4311–20.

12. Curley MD, Therrien VA, Cummings CL, Sergent PA, Koulouris CR, FrielAM, et al. CD133 expression defines a tumor initiating cell population inprimary human ovarian cancer. Stem Cells 2009;27:2875–83.

13. Gao MQ, Choi YP, Kang S, Youn JH, Cho NH. CD24þ cells fromhierarchically organized ovarian cancer are enriched in cancer stem cells.Oncogene 2010;29:2672–80.

14. Wei X, Dombkowski D, Meirelles K, Pieretti-Vanmarcke R, Szotek PP,Chang HL, et al. Mullerian inhibiting substance preferentially inhibitsstem/progenitors in human ovarian cancer cell lines compared withchemotherapeutics. Proc Natl Acad Sci U S A 2010;107:18874–9.

15. Szotek PP, Pieretti-VanmarckeR,Masiakos PT,DinulescuDM,ConnollyD,Foster R, et al. Ovarian cancer side population defines cells with stem cell-like characteristics and Mullerian Inhibiting Substance responsiveness.Proc Natl Acad Sci U S A 2006;103:11154–9.

16. Stewart JM, Shaw PA, Gedye C, Bernardini MQ, Neel BG, Ailles LE.Phenotypic heterogeneity and instability of human ovarian tumor-initi-ating cells. Proc Natl Acad Sci U S A 2011;108:6468–73.

17. Silva IA, Bai S, McLean K, Yang K, Griffith K, Thomas D, et al. Aldehydedehydrogenase in combination with CD133 defines angiogenic ovariancancer stem cells that portend poor patient survival. Cancer Res2011;71:3991–4001.

18. Kryczek I, Liu S, Roh M, Vatan L, Szeliga W, Wei S, et al. Expression ofaldehydedehydrogenase andCD133defines ovarian cancer stem cells. Int JCancer 2012;130:29–39.

19. Marcato P, Dean CA, Giacomantonio CA, Lee PW. Aldehyde dehydroge-nase: its role as a cancer stem cell marker comes down to the specificisoform. Cell Cycle 2011;10:1378–84.

20. Yasuda K, Torigoe T, Morita R, Kuroda T, Takahashi A, Matsuzaki J, et al.Ovarian cancer stem cells are enriched in side population and aldehydedehydrogenase bright overlapping population. PLoS One 2013;8:e68187.

21. Ohata H, Ishiguro T, Aihara Y, Sato A, Sakai H, Sekine S, et al. Induction ofthe stem-like cell regulator CD44 by Rho kinase inhibition contributes tothe maintenance of colon cancer-initiating cells. Cancer Res 2012;72:5101–10.

22. Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J, Frane JL, Tian S,et al. Induced pluripotent stem cell lines derived fromhuman somatic cells.Science 2007;318:1917–20.

23. Ishiguro T, Sato A, Ohata H, Sakai H, Nakagama H, Okamoto K. Differ-ential expression of nanog1 and nanogp8 in colon cancer cells. BiochemBiophys Res Commun 2012;418:199–204.

24. Okamoto K, Ishiguro T, Midorikawa Y, Ohata H, Izumiya M, Tsuchiya N,et al. miR-493 induction during carcinogenesis blocks metastatic settle-ment of colon cancer cells in liver. EMBO J 2012;31:1752–63.

25. Burleson KM, Casey RC, Skubitz KM, Pambuccian SE, Oegema TR Jr.,Skubitz AP. Ovarian carcinoma ascites spheroids adhere to extracellularmatrix components and mesothelial cell monolayers. Gynecol Oncol2004;93:170–81.

26. Molina R, Escudero JM, Auge JM, Filella X, Foj L, Torne A, et al. HE4 a noveltumour marker for ovarian cancer: comparison with CA 125 and ROMAalgorithm in patients with gynaecological diseases. Tumour Biol2011;32:1087–95.

27. Pece S, Tosoni D, Confalonieri S, Mazzarol G, Vecchi M, Ronzoni S, et al.Biological and molecular heterogeneity of breast cancers correlates withtheir cancer stem cell content. Cell 2010;140:62–73.

28. Cancer Genome Atlas N. Comprehensive molecular portraits of humanbreast tumours. Nature 2012;490:61–70.

29. Schwede M, Spentzos D, Bentink S, Hofmann O, Haibe-Kains B, Harring-tonD, et al. Stem cell-like gene expression in ovarian cancer predicts type IIsubtype and prognosis. PLoS One 2013;8:e57799.

30. Liu S, LiuC,MinX, Ji Y,WangN, LiuD, et al. Prognostic value of cancer stemcell marker aldehyde dehydrogenase in ovarian cancer: a meta-analysis.PLoS One 2013;8:e81050.

31. Quintin S, Gally C, Labouesse M. Epithelial morphogenesis in embryos:asymmetries, motors and brakes. Trends Genet 2008;24:221–30.

32. Ohgushi M, Matsumura M, Eiraku M, Murakami K, Aramaki T, NishiyamaA, et al. Molecular pathway and cell state responsible for dissociation-induced apoptosis in human pluripotent stem cells. Cell Stem Cell2010;7:225–39.

33. Watanabe K, Ueno M, Kamiya D, Nishiyama A, Matsumura M, Wataya T,et al. A ROCK inhibitor permits survival of dissociated human embryonicstem cells. Nat Biotechnol 2007;25:681–6.

34. Wang YC, Yo YT, Lee HY, Liao YP, Chao TK, Su PH, et al. ALDH1-brightepithelial ovarian cancer cells are associated with CD44 expression,drug resistance, and poor clinical outcome. Am J Pathol 2012;180:1159–69.

35. Landen CN Jr., Goodman B, Katre AA, Steg AD, Nick AM, Stone RL, et al.Targeting aldehyde dehydrogenase cancer stem cells in ovarian cancer.Mol Cancer Ther 2010;9:3186–99.

36. Kuroda T,Hirohashi Y, Torigoe T, Yasuda K, Takahashi A, AsanumaH, et al.ALDH1-high ovarian cancer stem-like cells can be isolated from serous andclear cell adenocarcinoma cells, and ALDH1 high expression is associatedwith poor prognosis. PLoS One 2013;8:e65158.

www.aacrjournals.org Cancer Res; 76(1) January 1, 2016 159

The Establishment of ALDH-Dependent Ovarian CSCs

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 11: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

37. Liebscher CA, Prinzler J, Sinn BV, Budczies J, Denkert C, Noske A, et al.Aldehyde dehydrogenase 1/epidermal growth factor receptor coexpres-sion is characteristic of a highly aggressive, poor-prognosis subgroupof high-grade serous ovarian carcinoma. Hum Pathol 2013;44:1465–71.

38. Chang B, Liu G, Xue F, Rosen DG, Xiao L, Wang X, et al. ALDH1 expressioncorrelates with favorable prognosis in ovarian cancers. Mod Pathol 2009;22:817–23.

39. Vanner RJ, Remke M, Gallo M, Selvadurai HJ, Coutinho F, Lee L, et al.Quiescent sox2(þ) cells drive hierarchical growth and relapse in sonichedgehog subgroup medulloblastoma. Cancer Cell 2014;26:33–47.

40. Weina K, Utikal J. SOX2 and cancer: current research and its implications inthe clinic. Clin Transl Med 2014;3:19.

41. Boumahdi S, Driessens G, Lapouge G, Rorive S, Nassar D, Le Mercier M,et al. SOX2 controls tumour initiation and cancer stem-cell functions insquamous-cell carcinoma. Nature 2014;511:246–50.

42. Ye F, Li Y, Hu Y, Zhou C, Hu Y, Chen H. Expression of Sox2 in humanovarian epithelial carcinoma. J Cancer Res Clin Oncol 2011;137:131–7.

43. Chou YT, Lee CC, Hsiao SH, Lin SE, Lin SC, Chung CH, et al. The emergingrole of SOX2 in cell proliferation and survival and its crosstalk withoncogenic signaling in lung cancer. Stem Cells 2013;31:2607–19.

44. Piva M, Domenici G, Iriondo O, Rabano M, Simoes BM, Comaills V, et al.SOX2promotes tamoxifen resistance in breast cancer cells. EMBOMolMed2014;6:66–79.

45. Park YT, Jeong JY, Lee MJ, Kim KI, Kim TH, Kwon YD, et al. MicroRNAsoverexpressed in ovarian ALDH1-positive cells are associated with che-moresistance. J Ovarian Res 2013;6:18.

46. Ma I, Allan AL. The role of human aldehyde dehydrogenase in normal andcancer stem cells. Stem Cell Rev 2011;7:292–306.

47. Saw YT, Yang J, Ng SK, Liu S, Singh S, Singh M, et al. Characterization ofaldehyde dehydrogenase isozymes in ovarian cancer tissues and spherecultures. BMC Cancer 2012;12:329.

Cancer Res; 76(1) January 1, 2016 Cancer Research160

Ishiguro et al.

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361

Page 12: Establishment and Characterization of an ... - Cancer Researchovarian cancer, and the established spheroids showed various characteristics of CSCs. We investigated functional roles

2016;76:150-160. Published OnlineFirst December 15, 2015.Cancer Res   Tatsuya Ishiguro, Ai Sato, Hirokazu Ohata, et al.   Cancer Stem-like Cells with an Enhanced Proliferative Capacity

Model of OvarianIn VitroEstablishment and Characterization of an

  Updated version

  10.1158/0008-5472.CAN-15-0361doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2015/12/10/0008-5472.CAN-15-0361.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/76/1/150.full#ref-list-1

This article cites 47 articles, 11 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/76/1/150.full#related-urls

This article has been cited by 4 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/76/1/150To request permission to re-use all or part of this article, use this link

on August 18, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0361