suppressing tgfb signaling in regenerating...

12
Tumor and Stem Cell Biology Suppressing TGFb Signaling in Regenerating Epithelia in an Inammatory Microenvironment Is Sufcient to Cause Invasive Intestinal Cancer Hiroko Oshima 1 , Mizuho Nakayama 1,2 , Tae-Su Han 1,2 , Kuniko Naoi 1 , Xiaoli Ju 1 , Yusuke Maeda 1 , Sylvie Robine 3 , Kiichiro Tsuchiya 4 , Toshiro Sato 5 , Hiroshi Sato 6 , Makoto Mark Taketo 7 , and Masanobu Oshima 1 Abstract Genetic alterations in the TGFb signaling pathway in combi- nation with oncogenic alterations lead to cancer development in the intestines. However, the mechanisms of TGFb signaling sup- pression in malignant progression of intestinal tumors have not yet been fully understood. We have examined Apc D716 Tgfbr2 DIEC compound mutant mice that carry mutations in Apc and Tgfbr2 genes in the intestinal epithelial cells. We found inammatory microenvironment only in the invasive intestinal adenocarcino- mas but not in noninvasive benign polyps of the same mice. We thus treated simple Tgfbr2 DIEC mice with dextran sodium sulfate (DSS) that causes ulcerative colitis. Importantly, these Tgfbr2 DIEC mice developed invasive colon cancer associated with chronic inammation. We also found that TGFb signaling is suppressed in human colitisassociated colon cancer cells. In the mouse inva- sive tumors, macrophages inltrated and expressed MT1-MMP, causing MMP2 activation. These results suggest that inammatory microenvironment contributes to submucosal invasion of TGFb signalingrepressed epithelial cells through activation of MMP2. We further found that regeneration was impaired in Tgfbr2 DIEC mice for intestinal mucosa damaged by DSS treatment or X-ray irradiation, resulting in the expansion of undifferentiated epithe- lial cell population. Moreover, organoids of intestinal epithelial cells cultured from irradiated Tgfbr2 DIEC mice formed "long crypts" in Matrigel, suggesting acquisition of an invasive pheno- type into the extracellular matrix. These results, taken together, indicate that a simple genetic alteration in the TGFb signaling pathway in the inamed and regenerating intestinal mucosa can cause invasive intestinal tumors. Such a mechanism may play a role in the colon carcinogenesis associated with inammatory bowel disease in humans. Cancer Res; 75(4); 76676. Ó2015 AACR. Introduction Accumulating evidence indicates that inammatory responses play an important role in cancer development (1, 2). The disrup- tion of genes encoding cyclooxygenase 2 (COX-2) or prostaglan- din E 2 (PGE 2 ) receptor, EP2, in Apc D716 knockout mice results in signicant suppression of intestinal polyposis (3, 4). Moreover, blocking transcription factors NF-kB and Stat3 in a chemically induced colitis-associated colon tumor model in mice causes suppression of colon tumor development (57). These results indicate that activation of inammatory pathways through PGE 2 , NF-kB, and Stat3 is required for intestinal tumorigenesis. How- ever, it has not yet been elucidated what role the inammatory responses play in the progression of benign intestinal tumors to invasive adenocarcinomas. Most intestinal adenomas are induced by APC mutations, resulting in the Wnt signaling activation, and tumors progress to adenocarcinomas by additional mutations such as those encod- ing RAS or transforming growth factor b (TGFb) type II receptor (TGFbRII; ref. 8). Mouse genetic studies have indicated that suppression of TGFb signaling accelerates development of malig- nant intestinal tumors in combination with mutations in Kras or Pten, although TGFb suppression alone does not cause tumorous changes (9, 10). These results indicate that suppression of the TGFb signaling is a key process involved in the malignant progression. The TGFb ligand binds TGFbRII, followed by the activation of TGFbRI. Activated TGFbRI then phosphorylates Smad2/3, which causes their binding with Smad4, and the Smad complex induces transcription of TGFb target genes (11). We have previously shown that immature myeloid cells (iMC) are recruited and express matrix metalloproteinase 2 (MMP2) at the invasion front of compound Apc D716 Smad4 knockout mouse intestinal adeno- carcinomas, which contribute to their submucosal invasion (12, 13). Moreover, disruption of Tgfbr2, encoding TGFbRII, in mouse mammary tumor cells results in the recruitment of 1 Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan. 2 Core Research for Evolutional Science and Tech- nology (CREST), the Japan Science and Technology Agency,Tokyo, Japan. 3 Equipe de Morphogenese et Signalisation cellulaires, Institut Curie, Paris, France. 4 Department of Advanced Therapeutics for Gas- trointestinal Disease, Graduate School Tokyo Medical and Dental University,Tokyo, Japan. 5 Department of Gastroenterology, Keio Uni- versity School of Medicine, Tokyo, Japan. 6 Division of Virology and Oncology,Cancer Research Institute, Kanazawa University, Kanazawa, Japan. 7 Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto, Japan. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: Masanobu Oshima, Division of Genetics, Cancer Research Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan. Phone: 81-76-264-6760; Fax: 81-76-234-4519; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-14-2036 Ó2015 American Association for Cancer Research. Cancer Research Cancer Res; 75(4) February 15, 2015 766

Upload: others

Post on 22-Aug-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

Tumor and Stem Cell Biology

Suppressing TGFb Signaling in RegeneratingEpithelia in an Inflammatory Microenvironment IsSufficient to Cause Invasive Intestinal CancerHiroko Oshima1, Mizuho Nakayama1,2, Tae-Su Han1,2, Kuniko Naoi1, Xiaoli Ju1,Yusuke Maeda1, Sylvie Robine3, Kiichiro Tsuchiya4, Toshiro Sato5, Hiroshi Sato6,Makoto Mark Taketo7, and Masanobu Oshima1

Abstract

Genetic alterations in the TGFb signaling pathway in combi-nation with oncogenic alterations lead to cancer development inthe intestines. However, the mechanisms of TGFb signaling sup-pression in malignant progression of intestinal tumors have notyet been fully understood. We have examined ApcD716 Tgfbr2DIEC

compound mutant mice that carry mutations in Apc and Tgfbr2genes in the intestinal epithelial cells. We found inflammatorymicroenvironment only in the invasive intestinal adenocarcino-mas but not in noninvasive benign polyps of the same mice. Wethus treated simple Tgfbr2DIEC mice with dextran sodium sulfate(DSS) that causes ulcerative colitis. Importantly, these Tgfbr2DIEC

mice developed invasive colon cancer associated with chronicinflammation.We also found that TGFb signaling is suppressed inhuman colitis–associated colon cancer cells. In the mouse inva-sive tumors, macrophages infiltrated and expressed MT1-MMP,

causingMMP2activation. These results suggest that inflammatorymicroenvironment contributes to submucosal invasion of TGFbsignaling–repressed epithelial cells through activation of MMP2.We further found that regeneration was impaired in Tgfbr2DIEC

mice for intestinal mucosa damaged by DSS treatment or X-rayirradiation, resulting in the expansion of undifferentiated epithe-lial cell population. Moreover, organoids of intestinal epithelialcells cultured from irradiated Tgfbr2DIEC mice formed "longcrypts" in Matrigel, suggesting acquisition of an invasive pheno-type into the extracellular matrix. These results, taken together,indicate that a simple genetic alteration in the TGFb signalingpathway in the inflamed and regenerating intestinal mucosa cancause invasive intestinal tumors. Such a mechanism may play arole in the colon carcinogenesis associated with inflammatorybowel disease in humans. Cancer Res; 75(4); 766–76. !2015 AACR.

IntroductionAccumulating evidence indicates that inflammatory responses

play an important role in cancer development (1, 2). The disrup-tion of genes encoding cyclooxygenase 2 (COX-2) or prostaglan-din E2 (PGE2) receptor, EP2, in ApcD716 knockout mice results insignificant suppression of intestinal polyposis (3, 4). Moreover,blocking transcription factors NF-kB and Stat3 in a chemicallyinduced colitis-associated colon tumor model in mice causes

suppression of colon tumor development (5–7). These resultsindicate that activation of inflammatory pathways through PGE2,NF-kB, and Stat3 is required for intestinal tumorigenesis. How-ever, it has not yet been elucidated what role the inflammatoryresponses play in the progression of benign intestinal tumors toinvasive adenocarcinomas.

Most intestinal adenomas are induced by APC mutations,resulting in the Wnt signaling activation, and tumors progress toadenocarcinomas by additional mutations such as those encod-ing RAS or transforming growth factor b (TGFb) type II receptor(TGFbRII; ref. 8). Mouse genetic studies have indicated thatsuppression of TGFb signaling accelerates development of malig-nant intestinal tumors in combination with mutations in Kras orPten, although TGFb suppression alone does not cause tumorouschanges (9, 10). These results indicate that suppression of theTGFb signaling is a key process involved in the malignantprogression.

The TGFb ligand binds TGFbRII, followed by the activation ofTGFbRI. Activated TGFbRI then phosphorylates Smad2/3, whichcauses their binding with Smad4, and the Smad complex inducestranscription of TGFb target genes (11). We have previouslyshown that immature myeloid cells (iMC) are recruited andexpress matrix metalloproteinase 2 (MMP2) at the invasion frontof compound ApcD716 Smad4 knockout mouse intestinal adeno-carcinomas, which contribute to their submucosal invasion(12, 13). Moreover, disruption of Tgfbr2, encoding TGFbRII, inmouse mammary tumor cells results in the recruitment of

1Division of Genetics, Cancer Research Institute, Kanazawa University,Kanazawa, Japan. 2Core Research for Evolutional Science and Tech-nology (CREST), the Japan Science and Technology Agency, Tokyo,Japan. 3Equipe de Morphogenese et Signalisation cellulaires, InstitutCurie, Paris, France. 4Department of Advanced Therapeutics for Gas-trointestinal Disease, Graduate School Tokyo Medical and DentalUniversity, Tokyo, Japan. 5Department of Gastroenterology, Keio Uni-versity School of Medicine, Tokyo, Japan. 6Division of Virology andOncology,CancerResearch Institute,KanazawaUniversity,Kanazawa,Japan. 7Department of Pharmacology, Kyoto University GraduateSchool of Medicine, Kyoto, Japan.

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

Corresponding Author: Masanobu Oshima, Division of Genetics, CancerResearch Institute, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa920-1192, Japan. Phone: 81-76-264-6760; Fax: 81-76-234-4519; E-mail:[email protected]

doi: 10.1158/0008-5472.CAN-14-2036

!2015 American Association for Cancer Research.

CancerResearch

Cancer Res; 75(4) February 15, 2015766

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 2: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

myeloid cells into tumor tissues, which promotes tumor metas-tasis through a process involving metalloproteinase activation(14). These results indicate that suppression of TGFb signalinggenerates a microenvironment that is critical for progression ofintestinal tumors. However, it is not understood how the inflam-matory responses affect the tumor progression induced by TGFbsuppression. Moreover, it has not been elucidated whether TGFb-suppressed epithelial cells acquire an invasive phenotype intumor tissues.

We herein show that ulcerative colitis causes submucosalinvasion of Tgfbr2-disrupted intestinal epithelia, leading to thedevelopment of invasive colon cancer. Moreover, TGFb signal-ing suppression in regenerating epithelia caused long cryptformation in Matrigel, which may reflect an increased capacityfor invasion. We also found that TGFb signaling is suppressedin human colitis–associated colon cancer cells. These resultsprovide a novel mechanism for the development of invasivecolon cancer where TGFb signaling suppression, chronicinflammation, and the regeneration of epithelial cells arecompounded.

Materials and MethodsAnimal models

Wild-type C57BL/6 mice were purchased from CLEA. ApcD716

mice, Tgfbr2flox/flox mice, and villin-CreER mice have beendescribed previously (15–17). Tgfbr2flox/flox mice were obtainedfrom Mouse Repository (NCI-Frederick, Strain Number: 01XN5,Frederick, MD). All animal experiments were carried out accord-ing to the protocol approved by the Committee on AnimalExperimentation of Kanazawa University, Japan.

Animal experimentsApcD716 Tgfbr2flox/flox villin-CreERmice were treated with tamox-

ifen at 4 mg/mouse once a week, from 5 weeks of age to generateApcD716 Tgfbr2DIEC mice. Intestinal tumors of ApcD716 mice andApcD716 Tgfbr2DIECmice were examined at 15weeks of age (n! 4).

For the dextran sodium sulfate (DSS) treatment experiments,Tgfbr2flox/flox villin-CreER mice were treated with tamoxifen at4 mg/mouse for 3 consecutive days to generate Tgfbr2DIEC mice,and mice were treated with 2% DSS in drinking water (MPBiomedicals) for 5 days. After DSS treatment, Tgfbr2DIEC micewere treated with tamoxifen for 2 days, and were examined at 3days, 4 weeks, 6 to 10 weeks (n! 10 for each) or 40 weeks (n! 2)after DSS treatment.

For the X-ray irradiation experiments, C57BL/6 mice andTgfbr2DIEC mice were irradiated with X-ray at 9 Gy (n ! 15 foreach), and were examined for the intestine phenotype chrono-logically from days 0 to 6.

For the azoxymethane/DSS colitis-associated colon tumormodel, wild-type mice (n ! 10) were intraperitoneally injectedwith 10 mg/kg azoxymethane (Sigma), followed by treatmentwith 2.0% DSS (MP Biomedicals) in drinking water for 5 days(week 1). This cycle was repeated twice during weeks 4 and 7, andmice were euthanized at week 15. Tumor tissues were used forimmunoblotting analysis and gelatin zymography.

Histology and immunohistochemistryThe tissue sections were stained with H&E or Masson's tri-

chrome stain, or were processed for immunohistochemistry.Staining signals of immunohistochemistry were visualized using

the Vectastain Elite Kit (Vector Laboratories). Antibodies againstE-cadherin (R&D Systems), a-SMA (Sigma), F4/80 (Serotec),MT1-MMP (GeneTex), Ki67 (Life Technologies), GFP (MolecularProbes), Collagen type IV (Nichirei Biosciences), CD44 (Milli-pore), SOX7 (R&D Systems), phosphorylated Smad2 (P-Smad2)at Ser465/467 (Millipore) and b-catenin (Sigma) were used forimmunohistochemistry. For fluorescence immunohistochemis-try, Alexa Fluor 594 or Alexa Fluor 488 antibodies (MolecularProbes) were used as the secondary antibody. Approval for theprojects usinghuman tissue sectionswasobtained from the TokyoMedical and Dental University Hospital Ethics Committee andKeio University Ethics Committee.

Bone marrow transplantationBone marrow (BM) cells were prepared from the femurs and

tibias of green fluorescent protein (GFP) gene transgenic mice.Recipient mice were irradiated with 9 Gy of X-rays, followed byintravenous injection of 2 " 106 BM cells.

Real-time RT-PCRSize-classified intestinal polyps and normal intestines of

ApcD716 mice (n ! 5 for each) were used for RNA extraction. ForDSS-treated mouse samples, normal colon tissues or invasivecolon tumors at 3 days, 4 weeks or 6 to 10 weeks (n ! 10 foreach) after DSS treatment were used for RNA extraction. The totalRNAs were reverse-transcribed using the PrimeScript RT reagentkit (TaKaRa) and were PCR-amplified using the SYBR PremixExTaqII (TaKaRa). The primers used for the real-timeRT-PCRwerepurchased from TaKaRa.

Immunoblotting analysisTissues were homogenized in lysis buffer, and protein sample

was separated in a 10% SDS-polyacrylamide gel. Antibodiesagainst active form of b-catenin (dephosphorylated at Ser37 andThr41; Millipore) and total b-catenin (Sigma), Stat3 (Cell Signal-ing Technology), and phosphorylated Stat3 at Tyr705 (Cell Sig-naling Technology) were used. An anti-b-actin antibody (Sigma)was used as the internal control. The ECL detection system (GEHealthcare) was used to detect the signals.

Gelatin zymographyTissue sample was lysed in SDS sample buffer, incubated for 20

minutes at 37#C and separated in a 10% polyacrylamide gelcontaining 0.005%gelatin labeledwith Alexa Fluor 670 (Abcam).After electrophoresis, gels were soaked in 2.5% Triton X-100 for1 hour, then gelatinolysis was carried out by incubation at 37#Cfor 24 hours. The gel was monitored by an Odyssey infraredimaging system (LI-COR).

Organoid cultureThe organoid culture using small intestinal epithelial cells was

performed as described previously (18). Briefly, organoids werecultured in Matrigel with Advanced DMEM/F12 medium (Invi-trogen) supplemented with 50 ng/mL EGF (Invitrogen), R-Spon-din1 conditioned medium (a kind gift from Dr. Marc Leushacke,A$ STAR, Institute ofMedical Biology, Singapore), and 100 ng/mLNoggin (Peprotech). The cultures were passaged once, and thecrypt length was measured under a dissecting microscope.

Organoid cell proliferationwas detected using the Click-iT EdUImaging System (Invitrogen). After EdU staining, organoids were

Invasion by TGFb Blocking in Regenerating Inflamed Mucosa

www.aacrjournals.org Cancer Res; 75(4) February 15, 2015 767

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 3: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

incubated with anti-CD44 antibody (Chemicon) followed bysecondary antibody, Alexa Fluor 488 (Molecular Probes). Thestained organoids were analyzed using a Zeiss 510 META laser-scanning microscope (Zeiss).

Statistical analysesThe data were analyzed using an unpaired t test, and are

presented as the means % standard deviation (SD). A value ofP < 0.05 was considered as statistically significant.

ResultsSize-dependent submucosal invasion of intestinal tumors

It has been demonstrated that the disruption of Tgfbr2,Smad4, or Smad3 in Apc-mutant mice causes developmentof invasive adenocarcinomas in the intestine, whereas simple

Apc-mutant mice develop only benign adenomas (12, 19–21).Accordingly, the combination of Wnt signaling activation andTGFb signaling suppression is implicated in the malignantinvasion of intestinal tumors. To further investigate the mech-anism underlying the submucosal invasion of intestinaltumors, we constructed compound mutant mice carrying bothApcD716 and Tgfbr2 mutation in intestinal epithelial cells(ApcD716Tgfbr2DIEC). We confirmed that ApcD716 Tgfbr2DIEC micedeveloped adenocarcinomas with submucosal invasion, where-as simple ApcD716 mice had only noninvasive adenomas(Fig. 1A and Supplementary Fig. S1). Notably, only the polyps>1 mm in diameter showed submucosal invasion in the ApcD716

Tgfbr2DIEC mice (Fig. 1B and Supplementary Fig. S1B). Histo-logically, large invasive polyps in ApcD716 Tgfbr2DIEC mice wereassociated with increased stroma and macrophage infiltration,which was not found in the small noninvasive polyps (Fig. 1C).

AA B

C

D

SM

SM

T T

Apc!716 Tgfbr2!IECApc!716

HE

HE (h

igh

mag

nific

atio

n)E-

cadh

erin

/"SM

A

HE F4/80 MT1-MMP MT

ApcD7

16 T

gfbr

2DIEC

tum

ors

Non

-inva

sive

Inva

sive

**

Epiregulin

5

10

15

20

25 *

10203040506070

***

123456 *

1

2

3

4

5

Adam10

**

MT1-MMP

50100150200250300350

*

2

4

6

8

10

*

**

102030405060 *

1

2

3

4

5

COX-2

*

*

100

200

300

400 *

100

200

300

IL6

** *

100

200

300

400

500

1234567

IL1#

Rela

tive

expr

essio

n lev

el

Normal mucosa $ < 1 mm 1 < $ < 2 mm 2 mm < $ Normal mucosa 2 mm < $

Size-classified polyps

ColonSmall intestine

Inva

ded

Not

inva

ded

$ < 0.5 mm 0.5 – 1 mm 1 – 2 mm 2 mm < $

Apc!716 Tgfbr2!IEC

Apc!716

Inva

ded

Not

inva

ded

$ < 0.5 mm 0.5 – 1 mm 1 – 2 mm 2 mm < $

Figure 1.Size-dependent submucosal invasionof intestinal tumors. A, representativephotographs ofApcD716mouse benignadenomas (left) and ApcD716

Tgfbr2DIEC mouse invasiveadenocarcinomas (right). H&E (top)and enlarged images of the boxedareas (middle), and fluorescenceimmunohistochemistry for E-cadherin(red) and aSMA (green; bottom). T,tumor; SM, submucosa. Arrowheads,submucosal invasion of tumorepithelial cells. Bars, 400mm(top) and200 mm (middle and bottom). B, sizeclassification of intestinal tumors ofApcD716 mice (top) and ApcD716

Tgfbr2DIEC mice (bottom) scoredusing "Swiss roll" histology sections.Each dot indicates an individual polyp.Different colors indicate independentmice. C, representative photographsof noninvasive (top) and invasivepolyps (bottom) of ApcD716

Tgfbr2DIEC; mice. H&E staining,immunohistochemistry for F4/80 andMT1-MMP, and Masson trichrome(MT) staining (left to right). Insets,high-powered magnification. Bars,200 mm (top) and 400 mm (bottom).D, expression levels of the indicatedfactors relative to the mean level ofnormal mucosa in size-classified smallintestinal and colon tumors of ApcD716

mice (mean % SD). $ , P < 0.05compared with the normal mucosallevel.

Oshima et al.

Cancer Res; 75(4) February 15, 2015 Cancer Research768

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 4: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

Membrane-type 1 matrix metalloproteinase (MT1-MP) expres-sion was induced in the stroma of the large invasive tumors,and collagen fiber deposition was also found in the stroma byMasson trichrome staining. Moreover, expression of IL1b, IL6,and COX-2 was induced when polyp size increased beyond >1mm in diameter in ApcD716 mouse small intestine, and expres-

sion levels of MT1-MMP, Adam10, and epiregulin were alsoincreased in the large polyps (Fig. 1D). The induction of thesefactors was also found in the colon polyps >2 mm in diameter.By a laser microdissection–based RT-PCR analysis, we foundthat these factors, except for Adam10, were predominantlyexpressed in the tumor stroma (Supplementary Fig. S2). Taken

A

B C

D E

F

Active #-cateninTotal #-catenin

#-Actin

Apc!716

NM polyps polypsAOM/DSS Tgfbr2DIEC + DSS

3 days 2 weeks 8 weeks (inv)

Tgfbr2DIEC + DSS (40 weeks)

Ce

Co

Ile

Ki67

Tgfbr2DIEC + DSS (10 weeks)

* *H&E

H&E MT

Wild-type + DSS3 days

4 weeks

Tgfbr2DIEC + DSS

3 days

*

4 weeks

DSS 5 daysTmx 3 days Tmx 2 days

Examination at 3 days Examination at 4, 6, 10, 40 weeks

Invaded tumor

Surface area

Sporadic

CAC

UC-associated colon cancer (p-Smad2) #-Catenin

Figure 2.Submucosal invasion of TGFb signaling-suppressed cells by ulcerative colitis. A, schedule for tamoxifen and DSS treatment and examination of mice. B, representativephotographs (H&E) of DSS-treated wild-type (left) and DSS-treated Tgfbr2DIEC (right) mouse colons 3 days (top) and 4 weeks (down) after treatment. Arrows (top),ulcer lesions. Arrowheads (bottom right), submucosal invasion of epithelial cells. $ , impaired regeneration of the normal gland structure. Bars, 200 mm. C, representativephotographs of invasive tumors of a DSS-treated Tgfbr2DIEC mouse 10 weeks after treatment. H&E (top), enlarged images of the boxed areas (bottom left), andMasson trichrome (MT) staining of serial section (bottom right). Arrows, location of muscularis mucosae. $ , impaired regeneration of the normal gland structure. Bars,500 mm (top and middle) and 200 mm (bottom). D, representative macroscopic photographs of invasive tumors developed in a DSS-treated Tgfbr2DIEC mouse at40 weeks after treatment (arrows, top left), and the H&E (top right) and immunohistochemistry for Ki67 (bottom). Arrows (bottom), the location of muscularismucosae; and inset, enlarged image of the boxed area. Ce, cecum; Ile, ileum; Co, colon. Bars, 2 mm (top) and 400 mm (bottom). E, immunoblotting for activeb-catenin and total b-catenin in the intestinal mucosa and polyps in ApcD716 and AOM/DSS-treated mice (left) and DSS-treated Tgfbr2DIEC mice (right)at the indicated time points. NM, normal mucosa; and inv, invasive tumors. b-Actin was used as an internal control. F, representative immunohistochemical findingsof p-Smad2 in ulcerative colitis–associated human colon cancer tissues (left, center), and the b-catenin in sporadic colon cancer (top right) and ulcerative colitis–associated colon cancer (UC; right bottom). Arrows (center), surface epithelial cells and invading tumor cells. Bars, 500 mm (left) and 50 mm (center and right).

Invasion by TGFb Blocking in Regenerating Inflamed Mucosa

www.aacrjournals.org Cancer Res; 75(4) February 15, 2015 769

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 5: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

together, these results indicate that the inflammatorymicroenvironment is generated by a tumor size-dependentmechanism in both the small intestinal and colon polyps. Itis therefore possible that such microenvironment is requiredfor submucosal invasion.

Submucosal invasion by TGFb signaling suppression andulcerative colitis

We thus examined the role of inflammatory responses inTGFb signaling suppression-associated submucosal invasionusing a colitis mouse model by treating Tgfbr2DIEC mice withDSS (Fig. 2A). The wild-type mice treated with DSS showedulcerative colitis beginning 3 days after DSS treatment, and thecolonic mucosa was repaired in 4 weeks after treatment (Fig.2B). Although mucosal ulcers were repaired in the Tgfbr2DIEC

mice in 4 weeks after DSS treatment, regeneration of thenormal gland structure was significantly impaired (Fig. 2B,asterisk). Importantly, colonic epithelial cells invaded to thesubmucosa in Tgfbr2DIEC mice by the end of fourth week aftertreatment. These invading epithelial cells continued prolifer-

ation, and formed invasive tumors associated with depositionof collagen fibers by the 10 weeks after DSS treatment(Fig. 2C). Furthermore, large solid tumors were visible fromoutside of the colon and cecum of Tgfbr2DIEC mice by the 40thweek after treatment (Fig. 2D). Epithelial cells of tumors werepositive for Ki67, indicating that the tumor cells continuedproliferation even at 40 weeks after DSS treatment. We con-firmed that disruption of Tgfbr2 in normal intestinal mucosadid not cause any morphologic changes (Supplementary Fig.S3), which is consistent with previous reports (20, 22). Thelevel of active b-catenin was significantly increased both inthe ApcD716 mouse intestinal tumors and colitis-associatedcolon tumors that were chemically induced by treatment withazoxymethane and DSS, however, active b-catenin level wasnot increased in the invasive tumors of DSS-treated Tgfbr2DIEC

mice (Fig. 2E). These results indicate that ulcerative colitisinduces invasive tumors in the TGFb-suppressed mucosa with-out activation of the Wnt signaling.

We next examined human inflammatory bowel disease (IBD)–related colon cancer. Nuclear localized P-Smad2was found in the

A B

C

DWild-typeDSS 3 daysDSS 4 weeks

Tgfbr2DIEC

DSS 3 daysDSS 6-10 weeks (inv)

TNF"

*ns

0.1

1

10

100

IL1#

*ns

0.1

1

10

100

1,000

IL6

ns*

0.01

0.1

1

10

100

1,000

CXCL1

*ns

0.01

0.1

1

10

100

1,000

CXCL2

**

0.01

0.1

1

10

100

1,000

10,000

COX-2

*ns

0.1

1

10

100

Rel

ativ

e ex

pres

sion

leve

l

WT WT + DSS Tgfbr2DIEC + DSS

1

23

of p

-Sta

t3/S

tat3

Rela

tive

band

inte

nsitie

s

p-Stat3

Stat3

#-Actin

WTNormal

WT + DSS Tgfbr2DIEC + DSS3 days(uc) 3 days(uc) 8 wks(inv)4 wks(rep)

Normal mucosa Invasive tumor

CCL2

*

0.1

1

10

100

CCL3

*

0.1

1

10

100

1,000

10,000

CCL4

*

0.1

1

10

100

1,000

CCL7

*

0.1

1

10

100

CCL8

*

0.1

1

10

Rel

ativ

e ex

pres

sion

leve

l

DSS-Tgfbr2DIEC [GFP Tg-BMT]inv

non-inv

invGFP non-invGFP F4/80/E-cad inv

Figure 3.Chronic inflammation in invasive tumors of Tgfbr2DIEC mice. A, representative photographs of immunohistochemistry for GFP in a DSS-treated Tgfbr2DIEC

mouse that had undergone BM transplantation from a GFP transgenic mouse (top). Enlarged images of invasive (bottom left) and noninvasive (bottomcenter) mucosa of the boxed areas (top). Fluorescent immunostaining for F4/80 (green) and E-cadherin (red) in an invasive tumor (bottom right).Bars, 400 mm (top) and 200 mm (bottom). B, relative expression levels of the indicated chemokines in the invasive tumors in DSS-treated Tgfbr2DIEC mice tothe mean level of wild-type mouse colons (mean% SD). $ , P < 0.05. C, immunoblotting for phosphorylated Stat3 and total Stat3 in the colon mucosa of controlwild-type mice (WT), DSS-treated wild-type and Tgfbr2DIEC mice. b-Actin was used as an internal control. uc, ulcerative colitis; rep, repaired mucosa;inv, invasive tumor. The band intensities for pStat3/Stat3 relative to the mean level of wild-type mice (red bar) are shown in a bar graph (bottom).D, expression levels of cytokines, chemokines, and COX-2 in DSS-treated wild-type or Tgfbr2DIEC mouse colonic mucosa relative to the mean levels ofthe repaired mucosa (green; mean % SD). $ , P < 0.05. ns, not significant.

Oshima et al.

Cancer Res; 75(4) February 15, 2015 Cancer Research770

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 6: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

surface epithelial cells of ulcerative colitis, however, it was notdetected in the invaded tumor cells, indicating that there wassuppression of TGFb signaling (Fig. 2F). A loss of P-Smad2staining in tumor cells was found in five out of eight cases(62.5%) of ulcerative colitis–associated colon cancer. Moreover,b-catenin accumulation was not detected in seven out of eightulcerative colitis–associated tumors (87.5%). Accordingly, it ispossible that suppression of TGFb signaling causes colon cancerinvasion in human patients with IBD without Wnt signalingactivation.

Chronic inflammatory responses in the invasive tumorsWe found that BM-derived cells andmacrophages infiltrated in

the stroma of invasive colon tumors, but not in the noninvasive

mucosa of DSS-treated Tgfbr2DIEC mice (Fig. 3A). Consistently,expression of monocyte-trophic chemokines (23), CCL2, CCL3,CCL4,CCL7, andCCL8,was increased significantly in the invasivecolon tumors (Fig. 3B). Moreover, Stat3 was constitutively phos-phorylated in the invasive tumors of DSS-treated Tgfbr2DIECmice,whereas its level was decreased in the repaired mucosa ofDSS-treated wild-type mice (Fig. 3C). These results indicate thatinflammatory responses are chronically maintained in the inva-sive tumor tissues, even after the repair of DSS-induced ulcers.

We thus determined the levels of inflammatory cytokines inboth the invasive tumors and DSS-induced ulcerative colitis.Expression was significantly upregulated for TNFa, IL1b, IL6,CXCL1, CXCL2, and COX-2 by DSS-induced ulcerative colitis inboth the wild-type and Tgfbr2DIEC mice (Fig. 3D, yellow and blue

A B

C

D

E

*

MT1-MMP

Rel

ativ

e ex

pres

sion

leve

l

0

0.5

1

1.5

2

2.5

3

3.5*

MMP20

1

2

3

4

5

6

7

8*

MMP90123456789

Wild-type–repaired mucosaTgfbr2DIEC-invasive tumors E-cadherin/MT1-MMP F4/80/MT1-MMP

Late

nt M

MP9

Late

nt M

MP2

N1 N2 N3 T1 T2 T3

DSS-treated Tgfbr2DIEC

Latent MMP9

Latent MMP2

Active MMP2

40 kDa

ApcD7

16

ApcD7

16

Tgfb

r2DI

EC

Wild

-type

(DSS

-uc)

Wild

-type

(DSS

-rep)

AOM

/DSS

N1 N2 A1 A2 C1 C2 I1 I2 R1 R2 T1 T2

Latent MMP9

Latent MMP2

Active MMP2

40 kDa

Rel

ative

ban

d in

tens

ities

of a

ctive

MM

P2

00.5

11.5

22.5

3

N1 N2 A1 A2 C1 C2 I1 I2 R1 R2 T1 T2

Collagen type IV

Figure 4.Expression of MT1-MMP and activation of MMP2 in invasive tumors. A, expression levels of indicated MMPs in the DSS-treated Tgfbr2DIEC mouse invasivetumors (red) relative to the level of repaired mucosa of DSS-treated wild-type mice (green; mean % SD). $ , P < 0.05 versus wild-type level. B, fluorescentimmunostaining of the invasive tumors in DSS-treated Tgfbr2DIEC mice for E-cadherin (green), MT1-MMP (red; left), F4/80 (green), and MT1-MMP (red; right).Insets, enlarged images of the boxed areas. Bars, 200 mm. C, gelatin zymography of the normal mucosa (N1-N3) and invasive tumors (T1-T3) of threeindividual DSS-treated Tgfbr2DIEC mice. Latent MMP9 and MMP2 were used as positive controls. D, immunostaining for collagen type IV in the invasivecolon tumors of DSS-treated Tgfbr2DIEC mice. White arrowheads, invading epithelial cells in submucosa; closed arrowheads, noninvading epithelialcells. Bars, 50 mm. E, gelatin zymography of the normal intestinal mucosa (N1-N2) and intestinal adenomas (A1-A2) of ApcD716 mice, invasiveadenocarcinomas (C1-C2) of ApcD716 Tgfbr2DIEC mice, ulcerative colitis tissues of DSS-treated wild-type mice (I1-I2), repaired mucosa of DSS-treatedwild-type mice (R1-R2), and noninvasive colon tumors (T1-T2) of azoxymethane/DSS-treated wild-type mice. Coomassie Brilliant Blue staining of the40-kDa bands is shown at the bottom of the zymography gels as a protein level control (C and E). Relative band intensities for active MMP2 to themean level of ApcD716 adenomas (red bar) are shown in a bar graph (E, bottom).

Invasion by TGFb Blocking in Regenerating Inflamed Mucosa

www.aacrjournals.org Cancer Res; 75(4) February 15, 2015 771

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 7: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

circles, respectively), whereas the levels of these factors in wild-type mice decreased significantly after the mucosa was repaired(Fig. 3D, green circles). In the invasive tumors of the Tgfbr2DIEC

mice, expression of TNFa, IL6, and CXCL2 remained high, where-as that of IL1b, CXCL1, and COX-2 decreased to the level ofrepaired mucosa (Fig. 3D, red circles). These results indicate thatdifferent types of inflammatory responses are induced in theinvasive tumor tissues of the Tgfbr2DIECmice comparedwith thosein the mice with DSS-induced acute colitis.

Expression of MT1-MMP and activation of MMP2 in invasivetumors

To investigate the mechanism underlying chronic inflamma-tion-associated invasion, we examined expression of MMPsthat are important for submucosal invasion (24). It has beenshown that MT1-MMP plays a key role in activation of MMP2(25) and MT1-MMP is expressed in macrophages, regulatinginflammatory responses (26, 27). The expression levels of MT1-MMP, MMP2, and MMP9 were significantly increased in theinvasive tumors of DSS-treated Tgfbr2DIEC mice (Fig. 4A). Fluo-rescence immunohistochemistry showed that the stromal cellsof invasive tumors expressed MT1-MMP, and most MT1-MMP–

expressing cells in tumor stroma were F4/80-positive macro-phages (Fig. 4B). Moreover, gelatin zymography analysesrevealed that MMP2 was activated in the invasive tumor tissues,but not in the normal colonic mucosa of the same DSS-treatedTgfbr2DIEC mice (Fig. 4C). Consistently, the immunostainingsignal for collagen type IV in the basement membrane wassignificantly decreased in the invading epithelial cells (Fig. 4D).These results suggest that chronic inflammation contributes tothe submucosal invasion through macrophage-expressed MT1-MMP, which leads to MMP2 activation, resulting in degrada-tion of basement membrane.

Because MT1-MMP expression was also induced in the ApcD716

benign adenomas (Fig. 1C and D), we further examined MMPactivation in other mouse tumor models. As anticipated, MMP2was activated in the invasive tumors of ApcD716 Tgfbr2DIEC mice(Fig. 4E). Notably, MMP2 activation was also found in thenoninvasive benign tumors of ApcD716 mice and AOM/DSS-trea-tedmice, although the band intensities were lower comparedwiththose of the ApcD716 Tgfbr2DIEC mouse invasive tumors (Fig. 4E).These results suggest that MMP2 activation is already induced inbenign intestinal tumors, and its activation level increases withthe progression of the tumor. Accordingly, it is also possible that

A

B C

D E

Active #-cateninTotal #-catenin

#-Actin

Normal polyp day 0 day 2 day 4 day 0 day 2 day 4Apc!716 Tgfbr2!IEC- irradiatedWT- irradiated

ApcD716 polyp Tgfbr2DIEC- irradiated

#-Ca

teni

n

Wild-type

Tgfbr2DIEC

1 2 3 4 5 6 7 8(days after irradiation)

0

20

40

60

80

100(%)

Surv

ival

afte

r irra

diat

ion

Rela

tive

SOX9

exp

ress

ion

level

Wild-typeTgfbr2DIEC

0

2

4

6

8

10

12

14

Day 0 Day 2 Day 4

*

*ns

*

*†

H&E

Tgf

br2DI

EC

Wild

-typ

e

E-cadherin / Ki67 CD44 SOX7

Figure 5.Impaired mucosal regeneration bysuppression of TGFb signaling. A,representative photographs of X-ray–irradiated wild-type (top) andTgfbr2DIEC (bottom) mouse smallintestines 6 days after irradiation.H&E staining, fluorescenceimmunostaining for E-cadherin(red), and Ki67 (green),immunohistochemistry for CD44 andSOX7 (left to right) are shown. Whitearrowheads, Ki67-positive cells.Arrows, CD44-positive epithelial cells;closed arrowheads, SOX7-positiveepithelial cells. Bars, 200 mm(left) and100 mm (center and right). B, survivalcurveofwild-type and Tgfbr2DIECmiceafter X-ray irradiation at 9 Gy. C,expression levels of SOX9 in X-ray–irradiated wild-type (gray bars) andTgfbr2DIEC (closed bars) mouseintestines relative to the mean level ofnonirradiated wild-type mice (day 0;mean % SD). $ , P < 0.05 versus day 0level; dagger, P < 0.05. ns, notsignificant. D, immunohistochemicalstaining for b-catenin in an ApcD716

mouse intestinal polyp (left) andirradiated Tgfbr2DIEC mouse smallintestinal mucosa (right). Whitearrowheads, b-catenin nuclearaccumulation. Bars, 25 mm. E,immunoblotting for active b-cateninand total b-catenin in the indicatedintestinal tissues. b-Actin was used asan internal control.

Oshima et al.

Cancer Res; 75(4) February 15, 2015 Cancer Research772

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 8: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

suppression of TGFb signaling causes acquisition of invasivenessof epithelial cells in such microenvironment.

Impaired mucosal regeneration by suppression of TGFbsignaling

Impaired mucosal regeneration from ulcer in Tgfbr2DIEC mice(Fig. 2B and C) suggested a role of TGFb signaling in regener-ation of injured intestinal mucosa. To test this possibility, micewere irradiated with X-ray at 9 Gy. In the irradiated wild-typemice, the number of proliferating cells in the crypt decreased ondays 1 to 3 after irradiation, followed by destruction of themucosal structure by day 4 (Supplementary Fig. S4). At thesame time, the undifferentiated cell population expanded, andnormal crypt-villous structures were regenerated by day 6,which was consistent with a previous report (28). X-ray irra-

diation in Tgfbr2DIEC mice also showed a decrease in theproliferating crypt cell population, followed by increased num-bers of proliferating undifferentiated cells (Supplementary Fig.S4). However, the extent of the injury was more severe in theTgfbr2DIEC mice, and the regeneration of the normal mucosalstructure was impaired in both the small intestine and colon(Fig. 5A and Supplementary Fig. S5). Because of such severephenotypes, most irradiated Tgfbr2DIEC mice died by day 7(Fig. 5B). Moreover, markers of undifferentiated epithelial cells,CD44 and SOX7, were expressed in the entire intestinal mucosaof the X-ray–irradiated Tgfbr2DIEC mice, whereas their expres-sion was limited to the crypt bottom in the wild-type mice(Fig. 5A and Supplementary Fig. S5B). Consistently, an intes-tinal progenitor cell marker, SOX9 (29), was upregulated in theintestine by irradiation, and the SOX9 level was significantlyhigher in Tgfbr2DIEC mice than in wild-type mice (Fig. 5C).Moreover, the nuclear accumulation and stabilization ofb-catenin was not found in the irradiated Tgfbr2DIEC mouseintestinal epithelia (Fig. 5D and E). Taken together, theseresults indicate that TGFb signaling is required for differenti-ation of regenerating epithelial cells, and that blocking theTGFb pathway causes expansion of the proliferating and un-differentiated epithelial cell population without activation ofWnt signaling.

Acquisition of invasive phenotype by blocking TGFb signalingin regenerating epithelial cells

We next studied the effect of TGFb signaling suppression inregenerating mucosa on invasive phenotype. Organoid cultureof the irradiated wild-type mouse-derived intestinal epithelialcells showed budding from cysts, forming mini-crypt structures(Fig. 6A), which was consistent with the original report (18).Importantly, intestinal epithelial cells derived from irradiatedTgfbr2DIEC mice formed gland-like long crypt structures in theMatrigel (Fig. 6A). We further examined the epithelial cellproliferation in the organoids by evaluating the EdU incorpo-ration to nuclei, and examined the undifferentiated status bydetermining the CD44 expression. Notably, the expression ofEdU and CD44 was found in the epithelial cells along the longcrypts of Tgfbr2DIEC mouse-derived organoids, although thatwas detected only in budding crypts of wild-type organoids.These results suggest that the long crypts are comprised ofproliferating undifferentiated epithelial cells. The proportionof long crypts >200 mmwas significantly higher in the irradiatedTgfbr2DIEC mouse-derived intestinal epithelial cells (42% %13% of all crypts) compared with epithelial cells derived fromirradiated Tgfbr2flox/flox control mice (3.9 % 4.5%; Fig. 6B). Suchlong crypt formation was not found also in organoids ofnonirradiated Tgfbr2DIEC mouse-derived intestinal epithelialcells. It is possible that long crypt formation in Matrigel reflects"collective cell migration" in the extracellular matrix, which isone of strategies used by cancer cells for invasion (30). There-fore, it is conceivable that suppression of TGFb signaling inregenerating mucosa results in the acquisition of invasivephenotype, which leads to collective migration in the inflam-matory microenvironment.

DiscussionGenome-wide analyses have indicated that accumulation of

genetic alterations in oncogenic and tumor-suppressor pathways

A

B

Cry

pt le

ngth

(µm

)

50

100

150

200

250

300

350

400

450

Wild-typ

eTgfb

r2!IEC

Tgfbr2

flox/flo

xTgfb

r2flox

/flox

Tgfbr2

flox/flo

x

Tgfbr2

!IECTgfb

r2!IEC

Tgfbr2

!IEC

Cont X ray–irradiated (9Gy)

Wild

-typ

eT

gfbr

2DIE

C

EdU/CD44/DAPIBright field

Figure 6.Acquisition of invasive phenotype by TGFb inhibition in regeneratingepithelia. A, representative bright-field photographs of organoid cultures inMatrigel (left) and confocal images of organoids immunostained for EdU(red) and CD44 (green; right) of wild-type (top) and X-ray–irradiatedTgfbr2DIEC (bottom) mouse intestinal epithelia. Arrowheads, EdU-negativeand CD44 weak cells in wild-type organoid. Bars, 200 mm (left) and 50 mm(right). B, crypt lengths of organoids derived from wild-type andnonirradiated Tgfbr2DIEC mice (light blue), irradiated Tgfbr2flox/flox (control)mice (blue), and irradiated Tgfbr2DIECmice (red). Red bar, 200-mm threshold,and crypts longer than 200 mm were judged to be long crypts.

Invasion by TGFb Blocking in Regenerating Inflamed Mucosa

www.aacrjournals.org Cancer Res; 75(4) February 15, 2015 773

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 9: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

is responsible for development of colon cancer (31). On the otherhand, the nature and significance of the individual genetic altera-tions are not yet understood (32). In addition, relatively fewmutations have been identified that are responsible for invasionand/or metastasis (8, 33), suggesting that microenvironment canpromote malignant progression. We have herein demonstratedthat simple genetic alterations in the TGFbpathway can lead to thedevelopment of invasive gastrointestinal cancers without addi-tional genetic alterations when the mucosa is inflamed andregenerating from injury (Fig. 7A).

It has been shown that suppression of TGFb signaling in theintestinal and mammary gland tumor cells induces chemokineexpression, which recruits myeloid cells to the tumor microenvi-ronment (12, 14). These myeloid cells express metalloprotei-nases, such as MT1-MMP, MMP2, and MMP9 that contribute tothe invasion ormetastasis of tumor cells. These results suggest thatelaboration of an inflammatory microenvironment is critical forthe malignant progression mediated by inhibition of the TGFbsignaling. We also found that MMP2 is activated by macrophage-expressingMT1-MMP in the invasive tumors. However, we foundthat MMP2 is activated also in the benign intestinal tumor tissueswith intact TGFb signaling. Accordingly, it is conceivable that theacquisition of an invasive phenotype by epithelial cells is furtherrequired for malignant progression where TGFb signaling issuppressed.

Blocking TGFb signaling in the intestinal epithelial cells didnot cause morphologic changes, indicating that TGFb signalingis not required for differentiation of normal intestinal stem/progenitor cells (20, 22). However, we found that suppressionof TGFb signaling in the injured intestinal mucosa blockedmucosal regeneration by suppressing differentiation, whichcaused the expansion of undifferentiated cell population.Accordingly, TGFb signaling is essential for regeneration fromdamaged mucosa in the gastrointestinal tract. Notably, intes-tinal epithelial cells derived from irradiated Tgfbr2DIEC mice

showed increased invasion in Matrigel, possibly caused byexpansion of undifferentiated epithelial cell population. How-ever, irradiation of Tgfbr2DIEC mice caused only dysplasticchanges without tumor development, indicating that blockingTGFb signaling in regenerating epithelial cells alone is insuffi-cient for induction of invasive tumors.

Accordingly, it is required for the development of invasivetumors that both the inflammatory microenvironment whereMT1-MMP is expressed and the regenerating epithelial cells withincreased invasiveness by inhibition of TGFb signaling (Fig. 7A).Such a mechanism is possibly important for cancer developmentassociated with IBD. In IBD lesions, the mucosa is continuouslyregenerating in a chronic inflammatory microenvironment, andthe expression of MT1-MMP, together with inflammatory che-mokines, is upregulated similar to that observed in Tgfbr2DIEC

mouse tumors (Supplementary Fig. S6). Furthermore, we hereindemonstrated that TGFb signaling is suppressed and Wnt signal-ing is not activated in more than 60% of ulcerative colitis–relatedcolon cancer cells. Consistently, it has also been reported thathuman colitis–associated colon cancer does not follow the ade-noma–carcinoma sequence, and mutations in b-catenin or APCare not common either (34). Accordingly, simple genetic altera-tions in the TGFb signaling pathway may cause the developmentof invasive tumors under IBD condition (Fig. 7A).

On theother hand, compoundmutantmice carryingmutationsin Apc and TGFb pathway genes showed progression of invasiveadenocarcinomas from Wnt-activated adenomas (19–21), indi-cating that the combination ofWnt activation and TGFb signalingsuppression is sufficient for malignant progression. In the regen-erating mucosa, stem cell population is expanded, with thesignaling in the Wnt and Notch pathways activated (35). It istherefore possible that the activation ofWnt signaling is necessaryfor malignant progression in sporadic tumors where TGFb sig-naling is blocked without mucosa regeneration. Accordingly,combination of Wnt activation and TGFb suppression in the

Figure 7.A schematic drawing of the TGFbsignaling suppression-inducedinvasive tumor development inregenerating and inflamed mucosa(A) and in Wnt signaling–activatedadenomas (B).

Cancer Res; 75(4) February 15, 2015 Cancer Research774

Oshima et al.

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 10: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

MT1-MMP–expressing inflammatory microenvironment is suffi-cient for the induction of invasive adenocarcinoma in normalintestine (Fig. 7B).

In conclusion, we have demonstrated that suppression of TGFbsignaling in the regenerating epithelial cells results in suppressionof epithelial differentiation andacquisition of invasive phenotypeof epithelial cells. Chronic inflammation induces development ofan MMP2-activating microenvironment. The cooperationbetween TGFb signaling suppression in the regenerating epitheliaand the inflammatory microenvironment can cause invasivecolon cancer development, which may explain mechanism ofIBD-associated colon tumorigenesis. Therefore, controlling theinflammatory microenvironment may help an effective preven-tive or therapeutic strategy against the malignant progression ofcolon cancer.

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

Authors' ContributionsConception and design: H. Oshima, M. Nakayama, H. Sato, M. OshimaDevelopment ofmethodology:H.Oshima,M. Nakayama, H. Sato, M. OshimaAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): H. Oshima, M. Nakayama, T.-S. Han, K. Naoi, X. Ju,Y. Maeda, S. Robine, K. Tsuchiya, H. Sato, M. Oshima

Analysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): H. Oshima, M. Nakayama, H. Sato, M.M. Taketo,M. OshimaWriting, review, and/or revision of the manuscript: H. Sato, M.M. Taketo,M. OshimaAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): H. Oshima, M. Nakayama, T. Sato, H. Sato,M. OshimaStudy supervision: H. Sato, M. Oshima

AcknowledgmentsThe authors thank Manami Watanabe and Ayako Tsuda for technical

assistance.

Grant SupportThis work was supported by CREST, the Japan Science and Technology

Agency, Japan (M.Oshima), Grants-in-Aid for Scientific Research on InnovativeAreas from the Ministry of Education, Culture, Sports, Science and Technologyof Japan #22114005 (M. Oshima), #24300325 (M. Oshima), and TakedaScience Foundation, Japan (H. Oshima and M. Oshima).

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received July 9, 2014; revised November 25, 2014; accepted December 1,2014; published online February 16, 2015.

References1. Coussens LM, Werb Z. Inflammation and cancer. Nature 2002;420:860–7.2. Elinav E, Nowarski R, Thaiss CA, Hu B, Jin C, Flavell RA. Inflammation-

induced cancer: crosstalk between tumours, immune cells and microor-ganisms. Nat Rev Cancer 2013;13:759–71.

3. OshimaM,Dinchuk JE, Kargman SL,OshimaH,Hancock B, Kwong E, et al.Suppression of intestinal polyposis inApcD716 knockoutmice by inhibitionof cyclooxygenase 2 (COX-2). Cell 1996;87:803–9.

4. Sonoshita M, Takaku K, Sasaki N, Sugimoto Y, Ushikubi F, Narumiya S,et al. Acceleration of intestinal polyposis through prostaglandin receptorEP2 in ApcD716 knockout mice. Nat Med 2001;7:1048–51.

5. Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, et al. IKKb linksinflammation and tumorigenesis in a mouse model of colitis-associatedcancer. Cell 2004;118:285–96.

6. Bollrath J, Phesse TJ, von Burstin VA, Putoczki T, Bennecke M, Batemen T,et al. gp130-mediated Stat3 activation in enterocytes regulates cell survivaland cell-cycle progression during colitis-associated tumorigenesis. CancerCell 2009;15:91–102.

7. Grivennikov S, Karin E, Terzic J,MucidaD,YuGY,Vallabhapurap S, et al. IL-6 and Stat3 are required for survival of intestinal epithelial cells anddevelopment of colitis-associated cancer. Cancer Cell 2009;15:103–13.

8. Markowitz SD, BertagnolliMM.Molecular basis of colorectal cancer. N EngJ Med 2009;361:2449–60.

9. Trobridge P, Knoblaugh S, Washington MK, Munoz NM, Tsuchiya KD,Rojas A, et al. TGF-b receptor inactivation andmutantKras induce intestinalneoplasms in mice via a b-catenin-independent pathway. Gastroenterol-ogy 2009;136:1680–8.

10. Yu M, Trobridge P, Wang Y, Kanngum S, Morris SM, Knoblaugh S, et al.Inactivation of TGFb signaling and loss of PTEN cooperate to induce coloncancer in vivo. Oncogene 2013;33:1538–47.

11. Ikushima H, Miyazono K. TGFb signaling: a complex web in cancerprogression. Nat Rev Cancer 2010;10:415–24.

12. Kitamura T, Kometani K, Hashida H, Matsunaga A, Miyoshi H, Hosogi H,et al. SMAD4-deficient intestinal tumors recruit CCR1&myeloid cells thatpromote invasion. Nat Genet 2007;39:467–75.

13. Kitamura T, Fujishita T, Loetscher P, Revesz L, Hashida H, Kizaka-KondohS, et al. Inactivation of chemokine (C-C motif) receptor 1 (CCR1) sup-presses colon cancer liver metastasis by blocking accumulation of imma-ture myeloid cells in a mouse model. Proc Natl Acad Sci U S A2010;107:13063–8.

14. Yang L, Huang J, Ren X, Gorska AE, Chytil A, Aakre M, et al. Abrogation ofTGFb signaling in mammary carcinomas recruits Gr-1&CD11b&myeloidcells that promote metastasis. Cancer Cell 2008;13:23–35.

15. OshimaM,OshimaH, Kitagawa K, KobayashiM, Itakura C, TaketoM. Lossof Apc heterozygosity and abnormal tissue building in nascent intestinalpolyps in mice carrying a truncated Apc gene. Proc Natl Acad Sci U S A1995;92:4482–6.

16. Chytil A, MagnusonMA, Wright CVE, Moses HL. Conditional inactivationof the TGF-b type II receptor using Cre;Lox. Genesis 2002;32:73–5.

17. el Marjou F, Janssen KP, Chang BH, Li M, Hindie V, Chan L, et al. Tissue-specific and inducible Cre-mediated recombination in the gut epithelium.Genesis 2004;39:186–93.

18. Sato T, Vries RG, SnippertHJ, van deWeteringM, Barker N, StangeDE, et al.Single Lgr5 stem cells build crypt-villus structures in vitro without amesenchymal niche. Nature 2009;459:262–6.

19. Takaku K, Oshima M, Miyoshi H, Matsui M, Seldin MF, Taketo MM.Intestinal tumorigenesis in compoundmutantmice of bothDpc4 (Smad4)and Apc genes. Cell 1998;92:645–56.

20. Munoz NM, Upton M, Rojas A, Washington MK, Lin L, Chytil A, et al.Transforming growth factor b receptor type II inactivation induces themalignant transformation of intestinal neoplasms initiated by Apc muta-tion. Cancer Res 2006;66:9837–44.

21. Sodir NM, Chen X, Park R, Nickel AE, Conti PS, Moats R, et al. Smad3deficiency promotes tumorigenesis in the distal colon of ApcMin/& mice.Cancer Res 2006;66:8430–8.

22. Biswas S, Chytil A, Washington K, Romero-Gallo J, Gorska AE, Wirth PS,et al. Transforming growth factor b receptor type II inactivation promotesthe establishment and progression of colon cancer. Cancer Res 2004;64:4687–92.

23. Mantovani A, Bonecchi R, Locati M. Tuning inflammation and immunityby chemokine sequestration: decoys and more. Nat Rev Immunol 2006;6:907–18.

24. Egeblad M, Werb Z. New functions for the matrix metalloproteinase incancer progression. Nat Rev Cancer 2002;2:161–74.

25. Sato H, Takino T. Coodinate action of membrane-type matrix metallo-proteinase-1 (MT1-MMP) and MMP-2 enhances pericellular proteolysisand invasion. Cancer Sci 2010;101:843–7.

26. Koziol A, Martin-Alonso M, Clemente C, Gonzalo P, Arroyo AG. Site-specific cellular functions of MT1-MMP. Eur J Cell Biol 2012;91:889–95.

www.aacrjournals.org Cancer Res; 75(4) February 15, 2015 775

Invasion by TGFb Blocking in Regenerating Inflamed Mucosa

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 11: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

27. Akla N, Pratt J, Annabi B. Concanavalin-A triggers inflammatoryresponse through JAK/STAT3 signalling and modulates MT1-MMPregulation of COX-2 in mesenchymal stromal cells. Exp Cell Res 2012;318:2498–506.

28. van Landeghem L, Santoro MA, Krebs AE, Mah AT, Dehmer JJ, Gracz AD,et al. Activation of two distinct Sox9-EGFP-expressing intestinal stem cellpopulations during crypt regeneration after irradiation. Am J GastrointestLiver Physiol 2012;302:G1111–32.

29. Furuyama K, Kawaguchi Y, Akiyama H, Horiguchi M, Kodama S, KuharaT, et al. Continuous cell supply from a Sox9-expressing progenitorzone in adult liver, exocrine pancreas and intestine. Nat Genet 2011;43:34–41.

30. Friedl P, Wolf K. Tumour-cell invasion andmigration: diversity and escapemechanisms. Nat Rev Cancer 2003;3:362–74.

31. The Cancer Genome Atlas Network. Comprehensive molecular character-ization of human colon and rectal cancer. Nature 2012;487:330–6.

32. Fearon E. Molecular genetics of colorectal cancer. Annu Rev Pathol 2011;6:479–507.

33. Vogelstein B, Papadopoulos NP, Velculescu VE, Zhou S, Diaz LA Jr, KinzlerKW. Cancer genome landscapes. Science 2013;339:1546–58.

34. Rogler G. Chronic ulcerative colitis and colorectal cancer. Cancer Lett2014;345:235–41.

35. Tan S, Barker N. Epithelial stem cells and intestinal cancer. Semin CancerBiol 2014 Feb 19. [Epub ahead of print].

Cancer Res; 75(4) February 15, 2015 Cancer Research776

Oshima et al.

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Page 12: Suppressing TGFb Signaling in Regenerating …genetics.w3.kanazawa-u.ac.jp/wp-content/uploads/2015/02/...Epithelia in an Inflammatory Microenvironment Is Sufficient to Cause Invasive

2015;75:766-776. Cancer Res Hiroko Oshima, Mizuho Nakayama, Tae-Su Han, et al. Intestinal CancerInflammatory Microenvironment Is Sufficient to Cause Invasive

Signaling in Regenerating Epithelia in anβSuppressing TGF

Updated version

http://cancerres.aacrjournals.org/content/75/4/766

Access the most recent version of this article at:

Material

Supplementary

http://cancerres.aacrjournals.org/content/suppl/2015/03/20/75.4.766.DC2.html

http://cancerres.aacrjournals.org/content/suppl/2015/03/18/75.4.766.DC1.htmlAccess the most recent supplemental material at:

Cited articles

http://cancerres.aacrjournals.org/content/75/4/766.full.html#ref-list-1

This article cites 34 articles, 7 of which you can access for free at:

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

SubscriptionsReprints and

[email protected]

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

Permissions

[email protected]

To request permission to re-use all or part of this article, contact the AACR Publications Department at

on November 13, 2015. © 2015 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from