Tumor-initiating stem cells of squamous cell carcinomas and their control by TGF-β and integrin/focal adhesion kinase (FAK) signaling

Markus Schobera and Elaine Fuchsa,b,1

aLaboratory of Mammalian Cell Biology and Development and bThe Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065

Contributed by Elaine Fuchs, May 17, 2011 (sent for review May 2, 2011)

Cancer stem cells (CSCs) sustain tumor growth through their ability of 7,12-dimethylbenz[α]anthracene (DMBA) and 12-o-tetrade- to self-renew and to generate differentiated progeny. These func- canoylphorbol-13 acetate (TPA) is a well-established chemical tions endow CSCs with the potential to initiate secondary tumors carcinogen treatment that leads primarily to papillomas in the bearing characteristics similar to those of the parent. Recently the skin. Recently, it was shown that, like hair follicles (HFs), tumors hair follicle stem cell marker CD34 was used to purify a CSC-like cell formed by this chemical regimen contain a small population of population from early skin tumors arising from treatment with 7,12- cells that express the cell-surface glycoprotein CD34, a marker – dimethylbenz[α]anthracene/12-o-tetradecanoylphorbol-13-acetate, expressed by a variety of adult SCs. In a 500 50,000 cell serial + fi which typically generates benign papillomas that occasionally prog- transplant assay, CD34 cells puri ed from these tumors were ress to squamous cell carcinomas (SCCs). In the present study, we shown to possess increased tumor-initiating ability compared with identify and characterize CSCs purified from malignant SCCs. We unfractionated tumor cells (7). The extent to which CD34 defines CSCs is currently unknown. Also poorly understood are show that SCCs contain two highly tumorigenic CSC populations that – differ in CD34 levels but are enriched for integrins and coexist at how CSCs self-renew, how they differentiate into non tumor- initiating progeny in cutaneous SCC, and how they compare with the SCC–stroma interface. Intriguingly, whether CD34lo or CD34hi, stem cells and progenitor cells in normal tissue. These questions α6hiβ1hi populations can initiate secondary tumors by serial limit- α loβ lo are pivotal to address for the development of therapies. dilution transplantation assays, but 6 1 populations cannot. The TGF-β pathway is commonly deregulated in human can- Moreover, secondary tumors generated from a single CSC of either cers, including SCCs, where TGF-β functions initially as a tumor lo hi α hiβ hi subtype contain both CD34 and CD34 6 1 CSCs, indicating suppressor but promotes metastasis in late-stage carcinogenesis fi their nonhierarchical organization. Genomic pro ling and hierarchi- (8, 9). TGF-β II (TβRII) is an essential component of cal cluster analysis show that these two CSC subtypes share a molec- β − the TGF- pathway, and its conditional ablation in skin epithe- ular signature distinct from either the CD34 epidermal or the lium (TβRIIKO) accelerates the development of aggressive SCCs hi CD34 hair follicle stem cell signature. Although closely related, upon exposure to the chemical carcinogen DMBA (9). Con- hi hi lo hi hi hi α6 β1 CD34 and α6 β1 CD34 CSCs differ in cell-cycle ex- comitant with TβRII loss in keratinocytes is the hyperactivation pression and proliferation characteristics. Indeed, proliferation and of integrins and the integrin signal transducer focal adhesion expansion of α6hiβ1hiCD34hi CSCs is sensitive to whether they can kinase (FAK) (9), features that promote cell proliferation, cell initiate a TGF-β receptor II–mediated response to counterbalance survival, and carcinogenesis (10–13). Indeed, integrins and elevated focal adhesion kinase-mediated integrin signaling within FAK are commonly up-regulated and are critical for the de- the tumor. Overall, the coexistence and interconvertibility of CSCs velopment of mouse and human solid tumors, including SCCs with differing sensitivities to their microenvironment pose chal- (10–15). The potent effects of TGF-β/TβRII and integrin/FAK lenges and opportunities for SCC cancer therapies. signaling on SCC formation are particularly intriguing, given that normal stem cells (SCs) of epidermis and HFs are responsive to cancer stem cell signature | epithelial–mesenchymal interactions | TGF-β signaling and display elevated integrin levels relative to skin cancer their committed progeny (16–19). These features provide an ideal platform for exploring the consequences of perturbing these pathways on the characteristics of SCC tumors and their ancers develop when cells acquire in tumor- associated CSCs. Csuppressor and proto-oncogenes that favor growth- promoting over growth-restricting processes, thereby unbalanc- Results ing tissue homeostasis (1, 2). Indeed, cancer cells are generally β fi proliferative, refractory to apoptotic cell death, and deficient in FAK Function Is Critical for SCC Tumor Susceptibility in T RII-De cient normal cellular differentiation. However, solid tumors such as Mice. Mice lacking FAK are more refractory to SCC formation, β cutaneous squamous cell carcinomas (SCCs) are not simply whereas those lacking T RII show enhanced tumor susceptibility. cancer cell clones but rather are complex structures composed of To investigate whether FAK/integrin signaling is critical for the β KO multiple cell types in unique microenvironments (3). How tumor development of T RII SCCs, we generated mice whose skin ep- β architecture develops and how it is maintained over time is still ithelium was conditionally null for both T RII and FAK (dKO). As β KO KO poorly understood for most cancers. Integral to these issues is were T RII (9) and FAK (20), dKO skins were asymptomatic. whether deregulated proto-oncogenes and tumor-suppressor genes affect all cancer cells equally or perform specific functions within distinct cellular compartments of the tumor. Of particular Author contributions: M.S. and E.F. designed research; M.S. performed research; M.S. importance is how these mutations affect those cancer cells that analyzed data; and M.S. and E.F. wrote the paper. ensure long-term growth and survival of the tumor. The authors declare no conflict of interest. Cancer stem cells (CSCs) sustain tumor growth through their Freely available online through the PNAS open access option. ability to self-renew and differentiate into hierarchically orga- Data deposition: The data discussed in this article have been deposited in the Gene nized cancerous tissue (4, 5). These functions endow CSCs with Expression Omnibus (GEO) database (accession no. GSE29328)(http://www.ncbi.nlm.nih. the potential to initiate secondary tumors bearing characteristics gov/geo/query/acc.cgi?acc=GSE29328). similar to those of the parent. In SCCs, actively proliferating 1To whom correspondence should be addressed. E-mail: [email protected]. cancer cells reside at the tumor–stroma interface and differen- This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. tiate into nontumorigenic pearls in the tumor center (6). The use 1073/pnas.1107807108/-/DCSupplemental.

10544–10549 | PNAS | June 28, 2011 | vol. 108 | no. 26 www.pnas.org/cgi/doi/10.1073/pnas.1107807108 Downloaded by guest on September 29, 2021 However, complete carcinogenesis by topical DMBA treatments occasional newly developing tumors regressed in both control and (two times per week) induced cutaneous SCCs in all genotypes, FAKKO mice, suggesting either that they failed to generate CSCs with some SCCs well contained but invasive and others less differ- for sustained long-term growth or that CSC self-renewal had been entiated and very invasive. SCC initiation appeared sooner in restricted by elevated suppressive activities within benign tumors TβRIIKO mice and later in FAKKO mice than in their wild-type lit- (Fig. 1C) (2). By contrast, tumor regression was not observed in termates. Interestingly, the accelerated tumor initiation in TβRIIKO TβRIIKO mice and was rare in dKO animals. Together, these data β β mice was not seen in dKO mice, which developed SCCs at rates suggest that T RII/TGF- and integrin/FAK signaling interact in indistinguishable from those of wild-type littermates (Fig. 1A). controlling not only tumor initiation and growth but also the fre- Once initiated, TβRIIKO SCCs grew faster than control SCCs quency with which benign tumors regress, persist, or progress to (Fig. 1B). Additionally, although all SCCs executed a program malignant SCCs. resembling disorganized epidermal wound repair, TβRIIKO SCCs were the most poorly differentiated (Fig. S1). Such signs are Fractionating SCC Populations by Their Surface CD34 and Integrins typical of highly aggressive SCCs (11, 14). and Functionally Testing Them for Self-Renewing Capacity in Vitro. FAK function appeared to be critical for the accelerated growth For the present study, we focused on tumors that progressed to of TβRIIKO SCCs, because growth rates in dKO SCCs were com- SCCs in each of the four genetic backgrounds. Based on the no- parable to those of FAKKO and control SCCs (Fig. 1B). Moreover, tion that CSCs should reside within the relatively undifferentiated keratin 5(K5)/ keratin 14+ proliferative cells at the tumor–stroma interface (6), we posited that CSCs of SCCs should display abundant integrins. Indeed, all SCC cells located at the tumor– A B C stroma interface expressed high levels of the hemidesmosomal α6 and β4 integrins and the focal adhesion marker β1 integrin, but 100 WT 15 40 only a fraction of these were CD34+ (Fig. 1D and Fig. S2). When 80 TβRII dKO 30 coupled with the genotype-specific differences in SCC charac- 60 FAK 10 teristics, these spatial differences in the intensity of CD34 and 20 – 40 integrin staining at the tumor stroma interface were suggestive of 5 fl β

(N=28) a heterogeneity that might be in uenced by T RII and/or FAK- (N=87) (N=92)

10 (N=109) 20 functions. 0 0 0

Tumor regression (%) To place this heterogeneity in the context of proliferative po- Tumor free mice % 0 10 20 30 Tumor diameter (mm) 1234 56 WT tential, we fractionated these cancer cells from genotypically weeks after DMBA weeks dKOFAK CELL BIOLOGY TβRII distinct primary SCCs by FACS. After eliminating stromal en-

live, CD11b, 45, 31neg α6β1hi dothelial cells (CD31), lymphocytes (CD45), and macrophages D E 5 5 (CD11b), we selected SCC keratinocytes based upon surface α6- 10 10 β SCC 4 4 integrin, 1-integrin, and CD34 levels (Fig. 1E). In contrast to 10 10 α6hiβ1hi SCC cells, populations either α6loβ1hi or lacking integrins 103 103 altogether failed to grow under our keratinocyte culture con- CD34lo CD34hi α hiβ hi 102 102 ditions. When 6 1 SCC cells were fractionated further into hi hi hi lo hi hi Stroma 0 0 CD34 α6 β1 and CD34 α6 β1 subpopulations, both cohorts CD34 α6 K5 α6 2 3 4 5 2 3 4 5 formed colonies (Fig. 1F). Although colonies growing from 50 µm 0 10 10 10 10 0 10 10 10 10 hi hi hi DAPI β1 CD34 CD34 α6 β1 SCC cells often appeared flatter and more dif- ferentiated, both cohorts could be passaged over the long term 40 p=0.1118 p=0.0007 and independent of genotype. F α6β1hiCD34lo α6β1hiCD34hi G fi p=0.0002 The nding that SCC cells with long-term proliferative poten- 30 tial are uniformly enriched for integrins was consistent with transgenic studies linking integrin levels to SCC formation (21, 20 22). However, the CD34 variation was surprising. In seeking hi 10 insights, we examined how the relative pool size of CD34 cells Median CD34 compares among integrin-positive SCC populations of different expression (x100) 0 genotypes. Interestingly, this pool was markedly expanded in KO KO WT FAK TβRII SCCs relative to dKO and FAK SCCs (Fig. 1G). TβRII dKO Moreover, in control SCCs, in which the activities of TGF-β and Fig. 1. TβRII and FAK interact to control tumor initiation and growth and FAK signaling are not defined, the pool was variable. These data influence SCC composition. (A) Kaplan–Meier curves describing tumor-free further underscore the robust effects of cooperative action of survival after DMBA treatments of wild-type (blue), TβRIIKO (red), dKO TGF-β/TβRII and integrin/FAK signaling on SCC composition. (green), and FAKKO (gray) mice (P < 0.0001, Mantel–Cox log-rank test). Wild- Additionally, the expanded CD34hiα6hiβ1hi population within type and dKO mice exhibit indistinguishable profiles (P = 0.271). Differences poorly differentiated TβRIIKO SCCs raises the possibility that between TβRIIKO and dKO (P = 0.0004, Mantel–Cox log-rank test) and FAKKO their rapid tumor growth may be achieved by enhancing self-re- and dKO (P = 0.0165, Mantel–Cox log-rank test) are statistically significant. (B) newal and/or survival of CSCs while suppressing their differenti- Tumor growth after initiation. TβRIIKO grow faster than other genotypes (n > ation. Moreover, because dKO SCCs are well differentiated and 10; error bars indicate SEM). (C) Tumor regression after initiation depends do not show an expansion of CD34hiα6hiβ1hi cells, this balance is upon TβRII function. (D) In SCCs, K5 is expressed in the undifferentiated cells at predicated on FAK/integrin signaling. the tumor–stroma interface. Some K5+ cells express CD34. Although many + + CD34 cells are in direct contact with the stroma, CD34 cells also can be found Single-Cell Tumor-Initiating Functional Assays Reveal Two Inter- at a distance from the tumor–stroma interface (arrow). (E) Representative changeable CSC Types in SCCs. flow cytometry profiles of cells isolated from primary SCCs and fractionated We hypothesized that, if our two based on surface α6 (CD49f) and β1 (CD29) integrins after selecting live cells integrin-rich SCC populations are truly CSCs, they should be able + + + lo to initiate secondary and tertiary tumors and undergo self-renewal and eliminating CD11b , CD45 , and CD31 stromal cells. Both CD34 and fi CD34hi α6β1hi cells (subfractionated into populations as in F) yielded large in the process. To test this hypothesis, we rst transduced primary keratinocyte colonies (delineated by yellow dotted lines in representative SCC cultures with retrovirus to express ubiquitously a triple- phase contrast images) that could be passaged over the long term on fibro- modality reporter (fluc-mrfp1-tk) (23). This reporter enabled us to blast feeders. (G) Median CD34 expression in wild-type, TβRIIKO, dKO, and mark the lineage permanently and to distinguish cancer paren- FAKKO SCCs reveals that the CD34hi population is expanded in TβRIIKO SCCs in chyma from stromal components. FACS-purified, transduced a FAK-dependent manner. The means of median CD34 levels in the four SCC SCC cells were then suspended in Matrigel and injected in- populations differ significantly (P = 0.02, ANOVA). tradermally into immunocompromised Nude mice to test their

Schober and Fuchs PNAS | June 28, 2011 | vol. 108 | no. 26 | 10545 Downloaded by guest on September 29, 2021 tumor-initiating potential (Fig. 2A). Approximately 10 d later, always formed from TβRIIKO than from FAKKO and dKO tumor- aberrant tumor growth was visible at the injection site, and tumors initiating cells. progressed to SCCs, as confirmed by histopathology. These culture results were recapitulated faithfully in vivo. Thus, In orthotopic transplantation experiments, secondary SCCs for all four genotypes, primary SCC cells with highest surface displayed growth characteristics similar to those of their primary integrins exhibited the ability to initiate secondary tumors. How- SCC origin and contained both CD34hi and CD34lo cell pop- ever, the SCC-forming efficiencies of our purified CD34hiα6β1hi ulations expressing both high and low levels of integrins (Fig. 2B). SCC cell populations varied considerably with genotype, showing Additionally, as we had observed for the primary tumors, both the highest potential in TβRIIKO SCCs and the poorest in dKO and high-integrin populations were effective in colony-forming assays, FAKKO SCCs (Fig. 2D). Interestingly, CD34hi and CD34lo cell but neither low-integrin population was effective. Moreover, in populations did not appear to have a strict hierarchical relation or these assays the levels of CD34 did not make a substantial dif- distinct differentiation potential, because serial transfer of either ference (Fig. 2C). That said, it was notable that more colonies CD34hiα6β1hi or CD34loα6β1hi CSCs yielded progeny SCCs con- taining both CD34hi and CD34lo cell populations (Fig. 2E). Fur- thermore, CD34hi CSCs lost their CD34 expression and became indistinguishable from CD34lo CSCs when cultured in vitro, and A cultures from either CSC type reestablished secondary tumors hi lo CD45/11b/31 pgk--luc-mrfp1-tk RFP high lineage negative lineage featuring both CD34 and CD34 CSCs when engrafted onto immunodeficient recipient mice (Fig. S3). SCC

CD34 To document the interconvertibility, tumor-initiation, and dif- CD34 ferentiation potential of these two distinct types of putative CSCs, CD29, CD49f CD29, CD49f we performed single-cell sort and transplantation experiments. We focused on TβRIIKO SCCs that were the most aggressive of the four cell populations and showed the highest tumor-initiation 50 B live C efficiency in limit-dilution assays (Fig. 2D). Similar to our results RFP pos lineage 40 5 105 in limit-dilution experiments, transplantation of single sorted 10 CD34hiα6hiβ1hi and CD34loα6hiβ1hi CSCs from TβRIIKO SCC 104 30 104 confirmed their high tumor-initiating potential (Fig. 3). We noted, 3 20 103 10 however, that the tumor-initiation potential (Fig. 3A) and growth 2 102 10 10 number Colony kinetics (Fig. 3B) were accelerated in secondary tumors that de- CD34lo CD34hi 0 0 loα hiβ hi 0 veloped from single CD34 6 1 CSCs compared with sec- hi hi hi 3 4 5 β1 0 102 103 104 105 lo CD34hi α β mRFP 0 10 10 10 CD34 ondary tumors that developed from CD34 6 1 CSCs. α6 CD34 α6β1hi α6β1hi The ability of individual cells to generate SCCs improves by nearly three orders of magnitude the SCC tumor-initiating cell populations previously described (7) and unequivocally establishes D 100 WT TβRII dKO FAK 80 these two cell populations as CSCs. Furthermore, the ability of hi lo 60 CSCs to shift reversibly between CD34 and CD34 states indi- 40 lo cates that neither CSC population is restricted in developmental 20 CD34 hi 0 CD34 1 10 100 1,000 1 10 100 1,000 1 10 100 1,000 1 10 100 1,000 Tumor formation (%) Tumor Transplanted cancer stem cells AB50 25 CD34lo CD34hi E 105 WT TβRII dKO FAK 40 20 104

CSC 30 3 15 lo 10 graft 2 10 CD34lo CD34hi CD34lo CD34hi CD34lo CD34hi CD34lo CD34hi 20 10

CD34 0 18% 5 % 11% 15 % 15% 8 % 75% 0.5 % 10 CD34lo tumor size (mm) 5 105 WT TβRII dKO FAK CD34hi 104 0

tumor initiation frequency (%) 0 CSC 3 hi 10 0 20 40 60 80 020406080 graft 2 10 CD34lo CD34hi CD34lo CD34hi CD34lo CD34hi CD34lo CD34hi days after transplantation days after transplantation

CD34 0 9% 5 % 17% 1% 20% 10 % 82% 0.2 %

2 3 4 5 2 3 4 5 2 3 4 5 2 3 4 5

β1 010 10 10 10 010 10 10 10 010 10 10 10 0 10 10 10 10 C 5 CD34lo CSC graft 105 CD34hi CSC graft CD34 10 104 104 Fig. 2. Cutaneous SCCs contain two interchangeable populations of CSCs that have tumor-initiating potential and are regulated by TβRII/TGF-β 3 103 and FAK/integrin signaling. (A) Strategy to isolate CSCs. FACS-purified 10 − − − CD34hiCD29hiCD49fhiCD45 CD31 CD11b cells are tagged with an RFP fluo- 102 CD34lo CD34hi 102 CD34lo CD34hi rescent reporter, and their long-term tumor-initiating potential is tested by 4% 9 % 5% 2 % 0 serial orthotopic grafting experiments in immune-compromised Nude mice. 0 fl fi +

(B) Representative ow cytometry pro les of RFP live cells isolated from RFP 0 103 104 105 0 103 104 105 secondary SCCs and separated based on their CD34 expression. Contour plots CD34 illustrate CD34lo (black) and CD34hi (brown) populations, which then were fractionated further based on α6(CD49f) and β1(CD29) integrin expression. Fig. 3. Tumor-initiation potential upon transplantation of single SCC CSCs. Overlays illustrate that CD34hi subpopulations are either α6hiβ1hi or α6loβ1lo.(C (A) Kaplan–Meier curve describing tumor-initiation frequency over time after and D) Colony formation (wild type, blue; TβRIIKO’ red; dKO,; green; FAKKO, transplantation of single CD34loα6hiβ1hi and CD34hiα6hiβ1hi cells into Nude gray; n > 2; error bars indicate SEM) and limit-dilution orthotopic trans- mice. (n =12,P = 0.185, Mantel–Cox log-rank test.) (B) Growth kinetics of plantation experiments (n =15–42 per data point; 576 grafts total) indicate tumors growing after transplantation of single CSCs. Although rates are that both α6β1hiCD34lo and α6β1hiCD34hi SCC cells form colonies in culture and variable, CD34loα6hiβ1hi CSCs have a tendency to initiate growth more rapidly tertiary SCCs in vivo, but their abilities depend upon TβRII and FAK function. than CD34hiα6hiβ1hi CSCs. (C) Flow cytometry profiles of tertiary SCCs. Note (E) Flow cytometry profiles of tertiary SCCs illustrating the potential of CD34hi that single CD34hi and CD34lo CSCs have the potential to generate CD34hi and and CD34lo CSCs to generate CD34hi and CD34lo progeny interchangeably. CD34lo progeny interchangeably within the tumors that they generate.

10546 | www.pnas.org/cgi/doi/10.1073/pnas.1107807108 Schober and Fuchs Downloaded by guest on September 29, 2021 potential. Rather, CSC interconversion appears to be sensitive to rich repeats and immunoglobulin-like domains 1 (Lrig1) the microenvironment and CSCs’ ability to respond to it. and Lgr6 (27, 28)]. In addition to their reduced expression, wild- type stem and progenitor markers showed no enrichment in Transcriptional Profiling of CSCs from Four Different Genetic Back- CD34hiα6hiβ1hi compared with CD34loα6hiβ1hi CSCs (Fig. 4B). grounds Identifies a CSC Molecular Signature for SCCs. To understand Although some HF-SC markers, including sex-determining re- further the relationship between these two CSC populations, we gion Y (SRY)-box 9 (Sox9) (29) and runt-related transcription next addressed whether they might share a common transcrip- factor 1 (Runx1) (30), were expressed at variable levels depending tional profile that distinguishes them from wild-type skin SCs. upon SCC background, they were still reduced relative to HF-SCs To this end, we carried out global gene-expression profiling of and were found at a distance from the core HF-SC marker cluster. purified CD34hiα6hiβ1hi and CD34loα6hiβ1hi populations from two Based upon hierarchical gene-cluster analyses, CSCs clustered independent RFP-tagged SCC tumor samples for each genetic together and were clearly more similar to each other than to ei- background. These 16 separate arrays then were contrasted ther wild-type skin SC population (Fig. 4A). with duplicate sets of CD34 hiα6hiβ1hi HF-SCs and CD34loα6hiβ1hi Because CSCs fell in a cluster distinct from normal skin SCs, we epidermal SCs from 8-wk-old wild-type mice (17–19). next sought to generate a CSC signature, i.e., genes up-regulated Corroborating our immunofluorescence data, overall CD34 by twofold or more in CSCs, irrespective of genotype and CD34 levels were significantly lower in α6hiβ1hiCD34hi CSCs than in status, relative to wild-type skin SCs (false-discovery rate, 0.05). HF-SCs (Fig. 4 A and B). Surprisingly, however, many previously Under these criteria, 742 genes formed the CSC signature (Fig. ascribed HF-SC markers, including leucine-rich repeat-contain- 4C and Dataset S1). Enriched in this signature were genes in- ing G-protein coupled receptor 5 (Lgr5) (24), LIM volved in cell cycle, mitosis, epithelial morphogenesis, hyper- 2 (Lhx2) (25), and nuclear factor of activated T-cells, cytoplas- proliferation, , and metabolism. They included many mic, calcineurin-dependent 1 (Nfatc1) (26), also were weakly pathways commonly affected in carcinomas, including growth expressed or were absent in CSCs, as were established markers of factor/signaling [Vegf-α; TGF-α; TGF-β1; MAPK4; breast cancer 1, epidermal and junctional zone/sebaceous gland SCs [e.g., leucine- early onset and breast cancer 2, early onset; v-Ki-ras2 Kirsten

WT CSC A B TβRII CSC E F 8 dKO CSC FAK CSC Skin SCs

6 CELL BIOLOGY

CSC WT 4 TβRII WT FAK dKO Epi Sox9 K5 K5 21 121212 2121 Sample α6 2 α6 l hlhlhlhlhlhlhlhllhhCD34 DAPI DAPI 0

log2 (CD34+/CD34-) G H

-2 Vdr Nfib Tnc Ctgf Lgr5 Lhx2 Tbx1 Sox9 Dab2 Gas1 Itm2a CD34 Macf1 Runx1 Nfatc1

Runx1 S100A6 K18 Col18a1 CSC vs. Epi(CD34lo) K8 NFATc1 C α6 α6 DAPI DAPI Cell Cycle 425 Keratinocyte differentiation Epithelial morphogenesis I J 105 Sox9 Kinetochore 742 104 Glucose metabolism Purine biosynthesis 103 969 Mitosis Pro-apoptosis 0 CD44 Glycolysis K5 hi 2 3 4 5 CSC vs. HF-SC (CD34 ) DAPI 010 10 10 10 CD44 α6 Hyper- Ccnd1, Ccne1, Ccne2, Tgfa, Krt6, Krt16, Lrig1 D proliferation Mapk4, Kras, NFATc1 K Skin SCC Self-renewal Bmi1, Hmga2, Sox2 AK Apoptosis Casp 1,3,8,12, Casp 2, 9, Bax, Tnf WT WT TβRII dKO F kDa NFATc1 Prdm1 102 TnC Epith. diff. Krt1, Krt10, Krt8, Krt18, Lor , Vdr, Dsg1, Dsc2, Dsc3 K6 Lgr6 EMT Tgfb1,Twist1, Twist2, Vim, Cdh1, Cnnta1, Krt15 52 Krt14 Krt8, Krt18, CD44 CD34 Carcinoma - Tgfb1, Ptk2 (FAK), Itgb1 (β1), Kras, Vegfa, Sox9 Tbx1 52 Nfatc1 Signaling Tgfa, Igf2r, Braca1,2 , Mdm2, Ereg, Lasp1 log2 Lhx2 -3 +3 VdR Migration Ptk2 (FAK), Itgb1 (β1), Fn1,Tgfb1,Dst, Tubulin Lgr5 ColXVII, Parva, Profilin2, Dlc1, Lamb1 52

Fig. 4. Defining a cancer stem cell signature for SCCs. (A) Hierarchical cluster analysis of Affymetrix 430 2.0 array data indicates that, whether CD34+ or − CD34 , SCC CSCs are more similar to each other than they are to wild-type SCs from either epidermis or HFs. Note that core markers of CD34+ HF-SCs, including Nfatc1, Lhx2, (VdR), and Lgr5, are strikingly reduced or absent in CSCs. Other adult SC markers [Lrig1, PR domain containing 1, with ZNF domain (Prdm1), and Sox9] are not uniformly elevated in CSCs. (B) Differential expression analyses indicate key distinctions between CSCs and wild-type SCs. hi hi hi lo KO KO Shown are fold changes (log2 scale) of previously described HF-SC markers between α6β1 CD34 and α6β1 CD34 cells of wild-type, TβRII , dKO, and FAK SCCs and wild-type skin epithelium. Note that HF-SC marker genes other than CD34 are not differentially expressed in these two CSC populations. Bars indicate the average of two independent experiments. HF-SCs (α6hiβ1hiCD34hi) vs. epidermis (α6hiβ1hiCD34neg) serves as a positive control. (C) Venn diagram revealing a core CSC signature of 742 genes that are up-regulated by twofold or more (P < 0.05) in all CSCs irrespective of their genotype and whether compared with wild-type HF-SCs or epidermal progenitors. Functional hierarchical cluster analysis indicates enrichment for processes that are commonly deregulated in cancers. (D) Selected sets of skin genes that are elevated (red) or abrogated (blue) by more than twofold in CSCs compared with wild-type-SCs. (E–K) Immunofluorescence microscopy (E–I) and immunoblot analyses (K) validate the CSC signature and define the CSC niche at the tumor–stroma interface. Validation was performed on independent primary SCCs that developed in wild-type mice. (Scale bars: 20 μm.) FACS analysis in J shows that surface marker CD44, up-regulated in CSCs and localized at the tumor–stroma interface (I), is in the integrin-high population.

Schober and Fuchs PNAS | June 28, 2011 | vol. 108 | no. 26 | 10547 Downloaded by guest on September 29, 2021 β rat sarcoma viral oncogene homolog; integrin 1; protein tyrosine A Cell cycle kinase 2 (FAK); and LIM and SH3 protein 1 (Lasp1)], self- Cell division renewal/hyperproliferation [BMI1 polycomb ring finger onco- Kinetochore TβRIITβRII 1 2dKO dKO1 2FAK 1FAK 2 gene; high mobility group AT-hook 2 (HMGA2); Sox2], and ep- Microtubule ECM ithelial-to-mesenchymal transitions [twist homolog 1 and twist DNA repair Chemotaxis homolog 2; vimentin; keratin 8/keratin 18; fibronectin] (Fig. 4 D– M-phase plate Heparin binding K). Conversely, genes down-regulated in CSCs relative to wild- Branching- – α morphogenesis type SCs included cell cell adhesion genes [E-cadherin; -catenin] Cell migration (Datasets S2 and S3). Such differences are suggestive of cyto- Development skeletal and adhesion remodeling within CSCs, and they corre- Anti-apoptosis lated well with typifying features of the CSC microenvironment at the leading front of the tumor–stroma interface. Overall, differ- B p=0.001 C ences between CSC and wild-type SC populations were confirmed p=0.019 non- for both up- and down-regulated genes (Fig. 4 E–K). p=0.025 CD34lo CD34hi tumor 1.0 initiating Differences Between CD34hi and CD34lo CSCs. Although similarities 0.8 trumped differences between CD34hi and CD34lo populations of integrin-rich CSCs, the differences proved to be interesting. By 0.6 Differentiation comparing their expression profiles in each SCC, we discovered 0.4 that CD34hi CSCs of TβRIIKO SCCs were enriched for tran- CD34lo CD34hi tumor 0.2 initiating

scripts encoding cell-cycle and DNA-repair , whereas Self-renewal hi CD34 CSCs of other genetic backgrounds showed compara- EdU (CD34hi/CD34lo) 0.0 tively greater enrichment for extracellular matrix (ECM), mi- β1, FN1 TGF-β Fak gration, and antiapoptosis genes (Fig. 5A and Dataset S4). WT TβRII dKO To pursue possible differences in the proliferation rates of the populations, we pulsed mice 6 h before euthanizing with the nu- Fig. 5. TGF-β/FAK signaling-dependent differences in proliferative activities cleotide analog 5-ethynyl-2′deoxyuridine (EdU). When FAKKO, of α6β1hiCD34hi CSCs. (A) Differential expression and functional hierarchical cluster analysis of α6β1hiCD34hi CSCs. Note that fast-cycling (TβRIIKO) dKO, or control SCCs were analyzed by FACS, EdU incorpora- hi hi tion localized predominantly to CD34loα6hiβ1hi CSCs, and only α6β1 CD34 CSCs are enriched for cell-cycle and DNA repair transcripts, KO α β hi hi few CD34hiα6hiβ1hi CSCs were EdU+ (Fig. 5B). By comparison, whereas slow-cycling (dKO, FAK ) 6 1 CD34 CSCs display elevated EdU incorporation rates were similar in CD34loα6hiβ1hi and transcripts for ECM, morphogenesis, migration, and antiapoptosis pathways. hiα hiβ hi fi β KO (B) Flow cytometry data indicate that EdU (6-h pulse) incorporates pre- CD34 6 1 CSCs and were signi cantly higher in the T RII lo hi β CD34hiα6hiβ1hi CSCs than in dKO and FAKKO CD34hiα6hiβ1hi dominantly in CD34 and not in CD34 CSCs, unless T RII is compromised KO and FAK function is sustained. Bar graphs indicate relative abundance of CSCs. Thus, within FAK and dKO CSCs, differences in CD34 + α β hi hi + α β hi lo fl EdU cells in 6 1 CD34 CSCs over EdU cells in 6 1 CD34 CSCs. Note appeared to be a re ection of differential proliferative activities, that loss of Tgfβ signaling in the presence of elevated FAK signaling leads to in a fashion similar to normal HF-SCs. In striking contrast, fi α β hi hi hiα hiβ hi β KO a speci c enrichment in the proliferative activity within the 6 1 CD34 CD34 6 1 CSCs from T RII SCCs incorporated amounts CSCs. (n > 5; P < 0.0001, ANOVA; error bars indicate SEM.) (C) Working loα hiβ hi of EdU similar to those in CD34 6 1 CSCs. model in which CSCs exist in interchangeable α6β1hiCD34hi and α6β1hiCD34lo states. Activated β1 integrin and its ligand fibronectin (FN1) promote self- Discussion renewal of both CSCs, whereas active TGF-β/TβRII signaling primarily influ- Our results provide compelling evidence that multiple CSC pools ences α6β1hiCD34hi CSCs, restricting their self-renewal and expansion. exist along the tumor–stroma interface in cutaneous SCCs. These CSC pools are interconvertible and lack a clear hierarchical or- hi hi ganization as long as high levels of integrin expression are main- and integrin CD34 tumor-initiating cells draws parallels between tained. However, they differentiate irreversibly and lose their human and mouse SCCs. Both contain cancer cells with elevated tumor-initiating potential as integrin expression is attenuated integrin and FAK expression, whereas CD34-expressing cells have – been found in mouse, but not in human, SCCs (7). when CSCs depart from the tumor stroma interface. Intriguingly, hiα hiβ hi loα hiβ hi the distinct but coexisting CSC pools differ in their proliferative The ability of our CD34 6 1 and CD34 6 1 pop- properties. As such, SCC CSCs’ behavior is similar to that of ulations of SCC cells to self-renew, be serially transferred over lo the long term, and initiate tumors at the single-cell level merited homeostatic skin where rapidly proliferating CD34 epidermal fi lo SCs and slow-cycling CD34hi HF-SCs coexist and interconvert their de nition as CSCs (4). In tumor-initiating assays, CD34 α6hiβ1hi populations were even more effective than CD34hiα6hi upon wounding or cell transplantation. Moreover, our studies of β hi SCCs developed in chemically induced FAK- and/or TβRII-null 1 populations at tumor initiation. That said, the percentage of hiα hiβ hi β KO skin epithelium revealed that CSC cycling activities within SCCs CD34 6 1 cells within T RII SCCs was greater than in are influenced by their ability to respond to cues from their mi- any other genetic background, and correspondingly, their overall fi croenvironmental niche, which in this case is the tumor–stroma ef ciency in serial transplantation and tumor aggressiveness interface where TGF-β/TβRII and integrin/FAK signaling in- were greater also. This result underscores an important princi- tersect. Finally, both transplantation and culture appear to reset ple, namely that the ability of CSCs to respond to integrin sig- the proliferative properties of these CSCs. naling and suppress TGF-β responsiveness overrides the effects, Three important findings came from our experiments. First was if any, of CD34 levels in determining their long-term self-renewal that our purification scheme further enriched for tumor-initiating and tumor-initiating characteristics. Given that SCCs lacking SCC cells compared with a previously published strategy which TβRII also were less differentiated than the SCCs formed on purified on the basis of CD34 without integrins (7). Second, our other genetic backgrounds, our data also imply that the ability to findings suggest that high integrin expression is a more general enhance signaling through FAK/integrin in the absence of active marker for tumor-initiating CSCs within SCCs and that CD34 ex- TGF-β signaling results in an impairment of the differentiation pression can distinguish between two specific subsets of tumor- process. Our comparative studies of SCCs lacking TβRII alone initiating SCC cells which differ in cycling behavior. Finally, our versus those lacking both FAK and TβRII showed clearly that data showed that TGF-β/TβRII and FAK/integrin signaling act in when FAK/integrin signaling was compromised, differentiation opposing fashion to control the self-renewal and tumor-initiation was restored in the TβRII-null SCCs. capabilities of CD34hiα6hiβ1hi cells, providing an explanation for Our results are intriguing in light of recent studies in melanoma, the enhanced aggressiveness of TβRIIKO SCCs that retain and ac- where tumor-initiating cells are abundant (31), marker genes are tivate FAK/integrin function. Our identification of integrinhiCD34lo dynamically regulated (31, 32), and differences in cycling behaviors

10548 | www.pnas.org/cgi/doi/10.1073/pnas.1107807108 Schober and Fuchs Downloaded by guest on September 29, 2021 can be observed (32). However, despite the interconvertibility buffer (PBS containing 2% chelexed FBS). Cells were pelleted at 300 × g for of our two CSC subtypes in SCCs, which like melanoma tumor- 10 min and then were resuspended in wash buffer, washed once, and in- initiating cells, show differences in cycling behaviors, some cells cubated with surface antibodies for FACS. within SCCs do show hierarchical relations: Low integrin- expressing SCC cells derive from high integrin-expressing cells, Tumor Cell Transplantation. Tumor cells were suspended in 50% Matrigel but they lack significant tumor-initiating potential and do not (356237; BD) in F medium at a concentration of 1, 10, 100, and 1000 cells/50 μL appear to be interconvertible (6). and injected s.c. into Nude mice. Tumor progression was documented pho- By defining and analyzing CSCs from multiple SCCs of distinct tographically once every week from the time of inoculation to the experi- genetic backgrounds, we have unearthed two populations that mental end point. Single-cell transplantation studies were performed by display heterogeneity across and within genotypes but differ pri- sorting a single cell per well in a 96-well plate containing 50% Matrigel marily in their CD34 and cell-cycle . Enriching for (356237; BD) in F medium. CSCs, we achieved SCC-initiating studies with single CSCs. Such Cell-Cycle Analysis. Cells were fixed in ice-cold 80% ethanol, and genomic serial transfers enabled us to demonstrate that the two populations μ μ of CSCs are both tumorigenic and interconvertible and that their DNA was labeled with 10 g/mL propidium iodide in the presence of 250 g/ fl mL RNAseA in PBS. Cell-cycle analysis was performed by flow cytometry and representation within SCCs is in uenced markedly by their ability analyzed using FlowJo software. to respond to TβRII/TGF-β and integrin/FAK signaling at the tu- – mor stroma interface. The relative abilities of genetically distinct EdU Labeling and Detection. Cell proliferation and S-phase entry in SCC were CSCs to escape inhibitory cues and exploit positive ones within measured by injecting 50 μg/g EdU i.p. into mice 6 h before analysis. EdU their microenvironment provides a molecular understanding of the incorporation was determined by flow cytometry using the Click-iT EdU variations in SC numbers within different cancers. Our findings also fl fl Alexa Fluor 488 Cell Proliferation Assay Kit for ow cytometry (C35002; place this exible behavior of CSCs in the framework of how some Invitrogen) following the manufacturer’s instructions. cancers become faster growing and more aggressive (Fig. 5C). Our CSC signature provides insights into SCC behavior, draws in- Statistics. Statistical and graphical data analyses were performed using Origin teresting parallels between SCCs and other cancers, and offers a 7.5 (OriginLab) and Prism 5 (GraphPad) software. rich list of potential diagnostic and/or therapeutic targets for future investigations. ACKNOWLEDGMENTS. We thank A. Levine and N. Stokes for their valuable technical support; G. Guasch for consultation and advice during the early Methods phases of this project; B. Keyes for generously providing RNA samples for stem Comparative Pathology. Tumor tissue was formalin fixed, paraffinembedded, and progenitor cell microarray analyses; and S. Raghavan, A. Christiano, S. Beronja, B. Keyes, S. Williams, M. Rendl, and E. Gonzalez for discussions and CELL BIOLOGY sectioned, and stained with H&E. Histological analyses were performed by comments on the manuscript. We are grateful for the help received from The Suzana S.Couto(MemorialSloan KetteringCancer Center, NewYork). All tumors Rockefeller University Flow Cytometry Center (which is supported by a grant used for the present study were SCCs, which varied in their degree of severity. from the Empire State Stem Cell fund through New York State Department of Health Contract C023046); the Bioimaging Resource Center; the Comparative Tumor Cell Preparation. Tumors were dissected from mice and separated from Biology Center, (an American Association for Accreditation of Laboratory normal skin, blood vessels, and connective tissue. Tumor tissue was minced Animal Care facility) for their expert handling and care of the mice; Suzana and treated with 0.25% collagenase (C2670; Sigma) in HBSS (Gibco) for 60 min S. Couto and the Memorial Sloan Kettering Comparative Pathology Core for pathology consultation and histological analyses and diagnoses of tumor at 37 °C; 62.5 U/mL DNaseI (LS002138; Worthington) was added for the last fi tissues; and The Memorial Sloan Kettering Genomics Core Facility for RNA 15 min of the collagenase treatment. The cell suspension was ltered with and microarray processing. This research also was supported by a grant from μ a 45- m strainer. Retained cell clumps were dissociated further by treatment the Emerald Foundation (to E.F.) and by Grants R01-AR27883 (to E.F.) and with 0.25% trypsin (Gibco) at 37 °C for 10 min and then were strained 5K99-AR057260-02 (to M.S.) from the National Institutes of Health. E.F. is through a 45-μm mesh. The combined cell suspensions were diluted in wash an investigator of The Howard Hughes Medical Institute.

1. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 100:57–70. 18. Blanpain C, Lowry WE, Geoghegan A, Polak L, Fuchs E (2004) Self-renewal, 2. He S, Nakada D, Morrison SJ (2009) Mechanisms of stem cell self-renewal. Annu Rev multipotency, and the existence of two cell populations within an epithelial stem cell Cell Dev Biol 25:377–406. niche. Cell 118:635–648. 3. Egeblad M, Nakasone ES, Werb Z (2010) Tumors as organs: Complex tissues that 19. Morris RJ, et al. (2004) Capturing and profiling adult hair follicle stem cells. Nat – interface with the entire organism. Dev Cell 18:884–901. Biotechnol 22:411 417. 4. Clarke MF, et al. (2006) Cancer stem cells—perspectives on current status and future 20. Schober M, et al. (2007) Focal adhesion kinase modulates tension signaling to control and focal adhesion dynamics. J Cell Biol 176:667–680. directions: AACR Workshop on cancer stem cells. Cancer Res 66:9339–9344. 21. Owens DM, Broad S, Yan X, Benitah SA, Watt FM (2005) Suprabasal alpha 5 beta1 5. Clevers H (2011) The cancer stem cell: Premises, promises and challenges. Nat Med 17: integrin expression stimulates formation of epidermal squamous cell carcinomas with- 313–319. out disrupting TGFbeta signaling or inducing spindle cell tumors. Mol Carcinog 44:60–66. 6. Pierce GB, Wallace C (1971) Differentiation of malignant to benign cells. Cancer Res 22. Sugiyama M, Speight PM, Prime SS, Watt FM (1993) Comparison of integrin expression – 31:127 134. and terminal differentiation capacity in cell lines derived from oral squamous cell 7. Malanchi I, et al. (2008) Cutaneous cancer stem cell maintenance is dependent on carcinomas. Carcinogenesis 14:2171–2176. β-catenin signalling. Nature 452:650–653. 23. Ray P, Tsien R, Gambhir SS (2007) Construction and validation of improved triple fusion 8. Bierie B, Moses HL (2006) TGF-beta and cancer. Cytokine Growth Factor Rev 17:29–40. reporter gene vectors for molecular imaging of living subjects. Cancer Res 67:3085–3093. 9. Guasch G, et al. (2007) Loss of TGFbeta signaling destabilizes homeostasis and 24. Jaks V, et al. (2008) Lgr5 marks cycling, yet long-lived, hair follicle stem cells. Nat promotes squamous cell carcinomas in stratified epithelia. Cancer Cell 12:313–327. Genet 40:1291–1299. 10. McLean GW, et al. (2005) The role of focal-adhesion kinase in cancer—a new 25. Rhee H, Polak L, Fuchs E (2006) Lhx2 maintains stem cell character in hair follicles. therapeutic opportunity. Nat Rev Cancer 5:505–515. Science 312:1946–1949. 11. Janes SM, Watt FM (2006) New roles for integrins in squamous-cell carcinoma. Nat Rev 26. Horsley V, Aliprantis AO, Polak L, Glimcher LH, Fuchs E (2008) NFATc1 balances – Cancer 6:175–183. quiescence and proliferation of skin stem cells. Cell 132:299 310. 12. McLean GW, et al. (2004) Specific deletion of focal adhesion kinase suppresses tumor 27. Watt FM, Jensen KB (2009) Epidermal stem cell diversity and quiescence. EMBO Mol Med 1:260–267. formation and blocks malignant progression. Genes Dev 18:2998–3003. 28. Snippert HJ, et al. (2010) Lgr6 marks stem cells in the hair follicle that generate all cell 13. Mitra SK, Schlaepfer DD (2006) Integrin-regulated FAK-Src signaling in normal and lineages of the skin. Science 327:1385–1389. cancer cells. Curr Opin Cell Biol 18:516–523. 29. Nowak JA, Polak L, Pasolli HA, Fuchs E (2008) Hair follicle stem cells are specified and 14. Mercurio AM (2003) Invasive skin carcinoma—Ras and alpha6beta4 integrin lead the function in early skin morphogenesis. Cell Stem Cell 3:33–43. – way. Cancer Cell 3:201 202. 30. Hoi CS, et al. (2010) Runx1 directly promotes proliferation of hair follicle stem cells 15. Pylayeva Y, et al. (2009) Ras- and PI3K-dependent breast tumorigenesis in mice and and epithelial tumor formation in mouse skin. Mol Cell Biol 30:2518–2536. humans requires focal adhesion kinase signaling. J Clin Invest 119:252–266. 31. Quintana E, et al. (2008) Efficient tumour formation by single human melanoma cells. 16. Jones PH, Harper S, Watt FM (1995) Stem cell patterning and fate in human epidermis. Nature 456:593–598. Cell 80:83–93. 32. Roesch A, et al. (2010) A temporarily distinct subpopulation of slow-cycling melanoma 17. Tumbar T, et al. (2004) Defining the epithelial stem cell niche in skin. Science 303:359–363. cells is required for continuous tumor growth. Cell 141:583–594.

Schober and Fuchs PNAS | June 28, 2011 | vol. 108 | no. 26 | 10549 Downloaded by guest on September 29, 2021