Inflammation linking EMT and cancer stem cells

Inflammation linking EMT and cancer stem cells

Oral Oncology 48 (2012) 1068–1075 Contents lists available at SciVerse ScienceDirect Oral Oncology journal homepage: www.elsevier.com/locate/oralonc...

244KB Sizes 0 Downloads 152 Views

Oral Oncology 48 (2012) 1068–1075

Contents lists available at SciVerse ScienceDirect

Oral Oncology journal homepage: www.elsevier.com/locate/oraloncology

Review

Inflammation linking EMT and cancer stem cells Chenchen Zhou a,1, Jeffrey Liu a,b,1, Yaling Tang a,⇑, Xinhua Liang a,c,⇑ a

State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Sec. 3, Renminnan Road, Chengdu Sichuan 610041, People’s Republic of China Department of Biological Sciences, University of Delaware, Nowak, DE 19716, USA c Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, No. 14, Sec. 3, Renminnan Road, Chengdu Sichuan 610041, People’s Republic of China b

a r t i c l e

i n f o

Article history: Received 17 April 2012 Received in revised form 5 June 2012 Accepted 8 June 2012 Available online 4 July 2012 Keywords: Epithelial–mesenchymal transition (EMT) Cancer stem cells (CSCs) Inflammation microenvironment

s u m m a r y Similar to actors changing costumes during a performance, cancer cells undergo many rapid changes during the process of tumor metastasis, including epithelial–mesenchymal transition (EMT), acquisition of cancer stem cells (CSCs) properties, and mesenchymal–epithelial transition (MET). Such changes allow the tumor to compete with the normal microenvironment to overcome anti-tumorigenic pressures. Then, once tissue homeostasis is lost, the altered microenvironment, like that accompanying inflammation, can itself become a potent tumor promoter. This review will discuss the changes that cancer cells undergo in converting from EMT to CSCs in an inflammation microenvironment, to understand the mechanisms behind invasion and metastasis and provide insights into prevention of metastasis. Ó 2012 Elsevier Ltd. All rights reserved.

Introduction Metastasis is the leading cause of death by cancer, yet it remains the most poorly understood component of cancer pathogenesis because of its complexity.1 Metastasis involves many steps: local invasion of cancer cells into the surrounding tissue, transport through the microvasculature of the lymph and blood systems, translocation—largely through the bloodstream—to microvessels of distant tissues, exit from the bloodstream, survival and adaptation in the distant microenvironment, and, finally, formation of a secondary tumor.2 Malignant tumors are characterized by an ability to invade and disseminate; indeed, tumors have been referred to as entities that constantly redefine themselves by their everchanging nature.3 However, certain tumor cells are more responsive to the metastatic route, and, of those, an even smaller fraction succeed. Metastasis depends on intrinsic properties of the tumor cells as well as factors derived from the tumor microenvironment.4 The first evidence that non-cancerous tissue elements might affect tumor formation and growth came from the field of inflammation.4 Although the molecular mechanisms involved in inflammation and cancer have remained elusive until recently, a role

for inflammation in tumorigenesis has been generally accepted. In fact, evidence increasingly indicates that an inflammation microenvironment is an essential component of all tumors, as tumors have been observed to arise at the sites of chronic inflammation, and inflammatory cells are present in biopsied tumor samples.4–7 Inflammation originates in an intrinsic pathway, whereby genetic events leading to tumor transformation promote the construction of an inflammatory milieu, as well as an extrinsic pathway established by tumor-infiltrating leukocytes, particularly macrophages.8,9 The inflammatory responses play critical roles at all stages of tumor development: initiation, promotion, malignant conversion, invasion, and metastasis.7,10–15 Recently, epithelial–mesenchymal transition (EMT) has taken center stage as the convergence point between inflammation and cancer.16–19 This review will highlight the changes occurring in cancer cells from EMT to acquisition of cancer stem cells (CSCs) properties in an inflammation microenvironment, to establish an understanding of the mechanisms behind invasion and metastasis. Additionally, this foundation will provide insight into prevention of tumor metastasis.

EMT and inflammation ⇑ Corresponding authors. Address: Department of Oral and Maxillofacial Surgery, West China College of Stomatology, Sichuan University, No. 14, Sec. 3, Renminnan Road, Chengdu Sichuan 610041, People’s Republic of China. Tel.: +86 15 884483965; fax: +86 28 85503479 (X. Liang). Tel.: +86 15 008458098; fax: +86 28 85503479 (Y. Tang) E-mail addresses: [email protected] (Y. Tang), [email protected], [email protected] (X. Liang). 1 These authors contributed equally to this work. 1368-8375/$ - see front matter Ó 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.oraloncology.2012.06.005

EMT is a process through which epithelial cells lose their epithelial traits and acquire instead the attributes of mesenchymal cells, including losing cell–cell adhesion structure, altering their polarity, reorganizing their cytoskeletal system, switching expression from keratin-to-vimentin-type intermediate filaments, and becoming isolated, motile.20 EMT was categorized into three

C. Zhou et al. / Oral Oncology 48 (2012) 1068–1075

distinct subtypes: embryonic and developmental EMT; tissue regeneration and fibrotic EMT; and cancer progression and metastatic EMT.16,21 EMT events in embryonic development have been very well defined22 and EMT in adult functions to promote organ morphogenesis as well as tissue regeneration during wound repair. However, there is an increasing awareness that EMT in cancer can endow cells with migratory and invasive properties.23 EMT program, interconverting epithelial cell types into cells with mesenchymal attributes, is itself accompanied by the acquisition of malignant cellular traits and the ability to complete various steps of the metastatic cascade.22,24–26 On the other hand, this program can be choreographed by a number of transcription factors to initiate the complex transcriptional program to impart malignant traits to cancer cells.24,27–29 Of note, these mechanistic insights into EMT maintenance and metastasis process have been greatly increased by the recent discovery that microenvironmental signals contribute to triggering EMT, including TGF-b, TNF-a, Notch, Hedgehog, Wnt, which imply the critical role of tumor-associated stroma in EMT induction.30–35 As previously mentioned, many of the cellular phenotypes associated with EMT do not arise as purely cell-autonomous processes. Instead, recruited stromal cells in tumor microenvironment seem to play key roles in the acquisition of these traits, and adaptation of cancer cells to these signals, rather than selection, seems to be key in EMT.36 Although further studies concerning the signals and circumstances from tumor microenvironment relevant to EMT induction should be exploited, well-characterized evidence has elucidated the role that inflammation, one of the most important microenvironment, plays in regulating EMT and metastasis. Recently, inflammation is regarded to be likely a key inducer of EMT in the pathological settings of cancer progression and EMT has now been considered to bridge inflammation and cancer.16–19 For example, SNAIL1 itself seemed to upregulate the expression of pro-inflammatory mediators such as several ILs (IL-1, IL-6 and IL-8). And TGF-b1 and HIF-1 in the inflammation microenvironment can induce the expression of TWIST and SIP1. A variety of cell types recruited by islands of cancer cells in advanced primary carcinomas into the surrounding stroma, such as fibroblasts, myofibroblasts, granulocytes, macrophages, myeloid cell-derived suppressor cells (MDSCs), mesenchymal stem cells, and lymphocytes, are thought to create a reactive stroma that appears to result in the release of EMT-inducing signals.37 Co-culture of tumor-associated macrophages (TAMs), one of the most well-characterized types of tumor-infiltrating inflammatory cells, was shown to increase their invasive capacity in an NF-jB-dependent and TNFa-dependent manner.6 Cancer-associated fibroblasts (CAFs), another major cells in inflammation microenvironment, can produce the fibroblast-derived matrix-degrading enzymes and growth factors such as FGF and HGF, all of which have been shown to be important stimuli of EMT.38–40 Breast carcinoma cells incubated with CAF-conditioned media have been characterized by a loss of E-cadherin-dependent adhesion and enhanced motility.41 CAFs depended on NF-jB, cycloxygenase-2 (COX-2) and HIF-1 to exert their propelling role for EMT.42 How do these tumor-infiltrating inflammation cells acquire the ability to promote cancer metastasis? Several mechanisms have been defined. Autocrine and paracrine extracellular signals, as well as genetic and epigenetic alterations, might each contribute to this change. Space constraints do not allow discussion of all the inflammatory components, but we will discuss the most prominence of them, such as NF-jB, TGF-b, TNF-a, STAT3, HIF-1a and inflammation-associated miRNAs, in further details. NF-jB is a key orchestrator of innate immunity and inflammation, and evidences suggest that this transcription factor, a potent EMT inducer, represents a potential molecular bridge between inflammation and cancer.19,43–52 Activation of NF-jB by Akt

1069

up-regulates SNAIL expression and induces EMT.48,53 SNAIL, in turn, suppresses the expression of the metastasis suppressor gene product Raf kinase inhibitor protein RKIP (inhibits the MAPK and the NF-jB pathways) as well as PTEN (inhibits the PI3K/AKT pathway).54–56 Alternatively, NF-jB directly binds the promoter of miR-448 suppressing its transcription, which induces EMT by directly targeting special AT-rich sequence-binding protein-1 (SATB1) mRNA, increasing EGFR-mediated TWIST1 expression, as well as NF-jB activation. This autoregulation between NF-jB and miR-448 may have a role in the regulation of chemotherapyinduced EMT.57 Whereas NF-jB activation in myeloid cells is associated with tumor promotion and EMT in inflammation-associated cancer models, Gavert et al. reported that L1-mediated metastasis of colorectal cancer cells does not require changes in EMT and operates by activating NF-jb signaling.58 Another master regulator of EMT is TGF-b. The ability of TGF-b to induce EMT is mediated by canonical Smad-dependent59 and non-canonical Smad-independent pathways. TGF-b activates Smad transcription factors and MAPKs to control expression of other regulators of EMT such as SLUG60 and SNAIL61,62 in normal and malignant mammary epithelial cells (MECs).63–66 TGFb-TGFbR-Smad2 signaling axis is needed to maintain epigenetic silencing of critical EMT genes in breast cancer progression.67 Among the non-Smad pathways, increasing evidences are emerging that TGF-b also regulates MECs behavior and the induction of EMT via the stimulation of numerous ‘‘noncanonical’’ effector systems, including small GTP-binding proteins, PI3K, MAPKs and NF-jB.68–74 Increasing findings indicate that many targets are involved in regulating TGF-b-mediated EMT, including Na, K-ATPase,75 IGFBP3,76 ZAG,77 SKIP, TGF-b type I receptor (TbRI),78 Dab2, ROCK and LIMK, PIAS1, nuclear transcription factors including members of SNAIL, SIP1 and TWIST, and Six family of homeobox (Six1).79,80 Then which targets are the most important and the correlation among them awaits further explanation. Secondly, support for the role of TGF-b signaling in EMT is further strengthened by the finding that the members of the miR-30 and/or miR-200 families controlled the process of TGF-b signaling in modulating EMT/MET in anaplastic thyroid carcinomas (ATCs)-derived cells.81 TGF-b abrogates the expression of miR-200, leading to the expression of ZEB1 and ZEB2 and their consequential down-regulation of E-cadherin expression to initiate EMT.82,83 On the other hand, TGF-b stimulation of normal MECs elicits their up-regulated expression of miR-155 via a Smad4dependent pathway.84 And up-regulated expression of miR-21 induced by TGF-b also participates in the initiation of EMT.85,86 Thus, the coupling of TGF-b to the regulation of miRNA expression and activity affords new avenues to potentially manipulate the pathophysiology associated with EMT. In addition, TGF-b signaling was observed to maintain DNA methylation patterns during EMT. For instance, hypermethylation of the E-cadherin promoter marks Ras-transformed MECs that have undergone a stable EMT induced by serum versus a transient EMT induced by TGF-b.87 Similar to NF-jB and TGF-b, TNF-a is also a potent stimulator of EMT by breast cancer cells. TNF-a induced SNAIL1 promoter activity and EMT in cancer cells, and can also stabilize SNAIL1 protein.7,88,89 TNF-a affects, regulates and also augments TGF-b1induced EMT,87,88 and a synergy of TNF-a and TGF-b signaling accelerates EMT dependent on enhanced p38 MAPK activity.33 Through actin remodeling, and the activation of TGF-b signaling, TNF-a promoted CD44 expression and moesin phosphorylation by protein kinase C, leading to the dissociation of cell–cell, the augmentation of cellular motility.90 This seemed that TGF-b is another target of TNF-a. In addition, NF-jB activation by TNF-a or expression of constitutively active IKK2 induced an EMT-phenotype.91,92 On the other hand, TGFb-induced EMT was mediated by NF-jB signaling.63,93 The coupling of TGF-b to NF-jB activation induces EMT and metastasis through eliciting the initiation of an autocrine

1070

C. Zhou et al. / Oral Oncology 48 (2012) 1068–1075

Cox-2:PGE2:EP2 receptor signaling cascade.73,94–98 All the observation explained the role of NF-jB as a modulator of TGFb1-induced EMT in addition to the connection among NF-jB, TGF-b and TNF-a in EMT induction. Another mechanism through which pro-inflammatory signal passages can affect EMT is via STAT3-mediated induction of TWIST transcription.51 But recent report suggests that STAT3 is a negative regulator of adenoma–carcinoma transition in colon cancer.99 Thus, these mechanisms remain to be confirmed in vivo. The link between hypoxia and EMT has been recently strengthened by the observed expression of SNAIL, ZEB and TWIST regulated by HIF-1, a key hypoxia effector.19,100,101 Hypoxia or overexpression of HIF-1a can promote EMT and metastatic phenotypes, and hypoxia itself may be an inducer of the EMT.102 Co-expression of HIF-1a, TWIST and SNAIL in primary tumors of patients with head and neck cancers correlated with metastasis and the worst prognosis. Another hypoxia-related gene, lysyl oxydase, was found to interact directly with SNAIL, but the functional and clinical significance of this interaction need to be further investigated.103,104 Support for the connection between cancer EMT and inflammation is further strengthened by studies of the role of inflammationassociated miRNAs in EMT and metastasis. For example, miR-29a induced by TGF-b led to EMT through suppressing tristetraproli.105 And miR-155 expression and promoter activity induced by TGF-b through Smad4 and the knockdown of miR-155 suppressed TGFb-induced EMT and tight junction dissolution, as well as cell migration and invasion.84 Alternatively, miR-9, overexpression in breast cancer cells, directly targets CDH1, the E-cadherin-encoding messenger RNA, leading to increased cell motility and invasiveness.106 TLR4-activated NF-jB rapidly increases the expression of miR-9 that operates a feedback control of the NF-jB-dependent responses.107

EMT confers a cancer stem cell phenotype Our mechanistic insights into the molecular and function features of the mesenchymal cells have been greatly increased by the discovery that sort of them endow the self-renewal trait associated with normal tissue stem cells (SCs) and CSCs.108,109 CSCs, or tumor-initiating cells, are able to self-renew, differentiate, and regenerate a phenotype of the original tumor when implanted into the nonobese diabetic and severe-combined immunodeficient (NOD–SCID) mouse, the gold standard assay for defining the CSCs fraction.110–113 The CD44+ or aldehyde dehydrogenase 1 (ALDH1) population of human head and neck squmaous cancinoma cells (HNSCCs) possess the unique properties of CSCs, the side population (SP).114–119 A link between metastasis and stem cells has also been shown in HNSCC.120–122 The concept of CSCs has sparked excitement and controversy in equal measure. New data emerging from the study of CSCs are forcing the cancer research community to revise basic concepts of cancer biology. The tradition opinion that each cell has the potential to be cancerous has been revised. Instead, only a subset has the competence of initiating cancer.123 On the other hand, the discovery of CSCs raises the conundrum regarding the origin of these cells. Determining the origin and biology of CSCs may reveal strategies for targeting them therapeutically. Although the definitive proof for identifying the origin of CSCs is needed to be provided, evidence exists for the stem cell, the committed progenitor and the fused cells as the origin of CSCs and EMT involving in these processes. Similarity in cell surface markers and morphological and fundamental characteristics suggests that normal tissue stem cells may be the targets of oncogenic transformation and the origin of CSCs.124–128 A potential connection between normal tissue stem cells and EMT has been confirmed by the

observation of EMT at the periphery of human embryonic stem cells (ESCs) clusters grown on matrigels.129 Additionally, Jamieson and his colleagues showed that leukemic granulocyte–macrophage progenitors have been shown to be able to self-renew through the activation of the Wnt/b-catenin signaling pathway,130 the important signal passage of EMT induction and stemness property, which announced the relation between progenitor cells and EMT, and supported the notion that a committed progenitor can be the cancer-initiating cell.130–133 The insight into cell fusion as the third mechanism for the apparent cellular plasticity associated with tissue stem cells is consistent with a proposal that bone marrow-derived cells (BMDCs)-tumor cell fusion expressed mesodermal traits and EMT regulators, and is a potential source of CSCs,134–137 which confirmed the fusion theory, a unifying explanation for metastasis.138 From studies in animal there is little doubt that tumor hybrids are generated in vivo and can be a source of metastases,138 however, it is not clear from previous studies whether stem cells themselves are fused with other cell types in different tissues in vivo. Taken together, these notions imply that CSCs can be formed through EMT. At the same time, the heterogeneity evident in clinical tumor pathology suggests that these induced CSCs cannot represent the only source of cells in the CSC pool within a tumor and intrinsic CSCs are likely to exist from their very inception, long before EMT occurs.37 EMT has been considered to be accompanied by the acquisition of the CSC properties, including tumorigenicity, ability to redifferentiate into an epithelial tumor, and ability to form spheroids.108,109,136 And EMT seems to be a condition to induce CSCs. Major clues as to a link between EMT and the acquisition of stem cell properties came from a recent study by Mani et al.108 who showed that the forced expression of EMT regulators such as SNAIL and TWIST or treatment with TGF-b resulted in cells with a CSC trait. Similarly, Morel and colleagues109 reported that breast cancer cell lines adding active Ras generated a population of CD24 /CD44+ cells that underwent EMT and had CSC attributes. Pawelek et al.136,137 demonstrated that EMT was induced by CD8+ T cells and the resulting tumors had characteristics of CSCs, including potent tumorigenicity, ability to reestablish an epithelial tumor, and enhanced resistance to drugs and radiation. In order to integrate the concepts of CSCs and EMT, Brabletz et al.26 proposed the existence of two cancer stem cell populations, a stationary cancer stem cell and a migratory cancer stem cell, in order to model all aspects of tumor progression. This model is further supported by the expression of EMT markers in CSCs from mammary carcinomas and, vice versa, by the activation of stem cell markers in EMT-induced mammary epithelial cells.108 Eastham et al.129 demonstrated that ESCs and mesenchymal stem cells were characterized by a shift from E-cadherin to N-cadherin expression, the expression of SNAIL factors, vimentin, and metalloproteases, but also retained the expression of several totipotent transcription factors, including Oct-4 and Nanog. Recently, Wellner et al.139 showed in pancreatic and colon cancers, ZEB1 linked EMT-activation and stemnessmaintenance by suppressing stemness-inhibiting miR-200 family members. In oral squamous cell carcinoma (OSCC) cell lines, fresh samples of OSCC tumor, and primary and recurrent cutaneous SCC, Biddle et al.140 found a novel CSC population that has undergone EMT (EMT CSC) , and the EMT and non-EMT CSC populations coexist by switching between the two phenotypic states through EMT and MET. In light of the study by Basu et al.141,142 one can speculate that the mesenchymal-like cisplatin-resistant cells represent the CD44+ cancer stem cell population in HNSCC. CD44, a marker associated with EMT and putative cancer stem cells has been identified as a critical target of ERK1/2 in promoting oral cancer aggressiveness.143 ALDH1+ cells within HNSCC cell lines had functional properties like invasion capacity and EMT.144,145 This indicates that stem cells can adopt a mesenchymal phenotype without losing

C. Zhou et al. / Oral Oncology 48 (2012) 1068–1075

their pluripotency, and mesenchymal status seems to be a condition to regain pluripotency.25 Based on the current study, a likely alternative explanation for metastasis progression of cancer is that a few of CSCs within the primary tumor separate from the primary lesion and play a critical role in metastasis because of their potential to migrate into different tissues.146 On the other hand, it is plausible at present that most human carcinoma cells access and exploit the EMT program to acquire malignant cell traits, though hardly proven.146,147 These represent an important conceptual advance; they showed that incoming metastatic cells had CSCs properties, or EMT or the fusion that EMT confers a cancer stem cell phenotype. The idea of CSCs and EMT has recently moved to the forefront of cancer research, however, there is still a lack of a widespread consensus on their description and definition. With the development of significant progress made in characterizing the in vivo architecture and functions of stem cell niches in different model systems including Caenorhabditis elegans, Drosophila and mammals,148–152 the importance of the tumor and the host microenvironment in conditioning the stem cell status itself and the development of therapeutic strategies has been recognized, the mechanisms through which niche factors can modulate stem cell fate decision has been gradually shed light on.

Inflammation and CSCs Although there are some disputes on the role of CSCs and their microenvironment in the stem cell niche, that is whether microenvironment has been considered to be affiliated to stem cells or individual stem cell153,154 was affiliated to environment, the stem cell niche has now been defined as a dynamic microenvironment that responds to local and systemic cues, ultimately influencing stem cell fate and governing stem cell fate.12,150,155–164 Connelly et al.165,166 found that the shape of the extracellular matrix (ECM)-coated surface rather than its ECM composition or density affects the initiation of stem cells and signaling events that mediate the interaction between the basement membrane and the basal epidermal stem cells are required to maintain their stem-like state. For example, stem cells lacking the small GTPase Rac1 fail to undergo shape changes and migrate on the provisional ECM at the wound edge.167 The differentiation of epithelial stem cells in response to extrinsic or intrinsic cellular stress pathways may also act as a protective mechanism against oncogenic perturbation.168 Recently, significant strides have been made in characterizing the in vivo architecture and functions of stem cell niches in different model systems and elucidating the symmetric and asymmetric mechanism through which niche factors can modulate stem cell fate,148–150,169–173 however, an underexplored variable is the inflammation milieu of this highly specialized microenvironment. Certain work has begun to focus on the relevance of inflammation to the stem cell niche. The IL-8/CXCR1 axis secreted by endothelial and stromal cells may regulate mammary stem cell proliferation or self-renewal and play a role in mediating interactions between tumor stem cells and microenvironment.174 The role for IL-6 in breast CSCs and IL-4 in mediating chemoresistance of colon CSCs have also been reported.175–177 Moreover, the metastatic, aggressive behavior of inflammatory breast cancer (IBC) may be mediated by a CSC component that displayed ALDH enzymatic activity.178 In addition, Arwert et al.179 showed that Dexamethasone treatment inhibited tumor formation in InvEE mice. InvEE skin and tumors expressed high levels of IL-1a, and treatment with an IL-1 receptor antagonist delayed tumor onset and reduced incidence. The depletion of cd T cells and macrophages also reduced tumor incidence. These indicated that differentiated epidermal cells can initiate tumor formation without reacquiring the ability

1071

to divide and that they do so by triggering an inflammatory infiltration. These observations fueled a hypothesis that perhaps inflammation is critical to the milieu of stem cell niches. Furthermore, the molecular mechanisms of inflammation driving CSCs during tumor promotion discussed below continue to drive the excitement behind this hypothesis. TGF-b has been implicated in maintaining hematopoietic stem cells (HSCs) quiescence.180,181 The members of TGF-b superfamily can activate differentiation of vascular progenitor cells derived from mouse ESCs.182,183 In the bone marrow niche, TGF-b signaling can function as a fine-tuning mechanism for cross-talk between hematopoietic stem cells subtypes and myeloid progenitors.184 Another critical signal passage of the inflammation and CSCs is Wnt/b-catenin. The pro-inflammatory cytokine TNF-a promotes nuclear entry of b-catenin during inflammation-associated gastric cancer in the absence of any mutations in Wnt/b-catenin pathway components.185 Activation of b-catenin, downstream to the IKKa, b/NF-jB pathway, may be integral to the hyperproliferative effects of progastrin on proximal colonic crypts.186 Other mechanisms through which pro-inflammatory cytokines can affect CSCs is via STAT3, which is linked to both stem cell reprogramming and stem cell renewal.187 IL-1, TNF-a, and IL-6 can promote MMP expression, invasiveness, and metastasis via NF-jB and STAT3.188 HNSCC cells, HN13 and HN30, were shown to express high levels of IL-6 through NF-jB-dependent transcriptional activation and genetic inhibition of NF-jB reduced IL-6 levels resulting in a decreased in STAT3 phosphorylation and activation.189–191 PGE2 played a crucial role in HSCs growth and development not only in embryonic, but also in adult stem cell homeostasis in both simple and complex vertebrate systems.192 By expressing one or more of the same cytokines such as IL-6, activated prostate epithelial stem cells acquired a survival advantage.193 IL1a-dependent NF-jB signaling can activate both skin stem cell proliferation and cutaneous inflammation.194 The establishment of one or more autocrine signalling loops results in an expansion of these stem cells in the absence of inflammation, as a potential first stage in the development of the tumor. Subsequently, bioimaging approaches that allow the continuous long-term observation of individual differentiating mouse hematopoietic progenitor cells (HPCs) demonstrate that the physiological cytokines, macrophage colony-stimulating factor and granulocyte colony-stimulating factor, can instruct hematopoietic lineage choice.195 On the other hand, inflammation cells are also involved. Self-renewal and pluripotent properties of human ESCs depend on a dynamic interplay between human ES cells and autologously derived human ES cell fibroblastlike cells, which define the stem cell niche of pluripotent human stem cells through insulin-like growth factor (IGF)-II/IGF1R axis.196 While the recognition that inflammation as well as growth factors and cytokines produced by inflammation may confer a stem cell-like phenotype upon tumor progenitors or stimulate stem cell expansion, thereby enlarging the cell pool that is targeted by environmental mutagens,10 is being increasingly documented and accepted, the association of inflammation molecular pathways with CSCs and the role of EMT during this process are still matter of discussion.

EMT, inflammation and CSCs There are increasing evidences that pointed to inflammationinduced EMT as critical to the connection of inflammation and CSCs, just as EMT as a common link between inflammation and cancer. One of the best explanations is provided by the model of inflammation cells. Bone marrow-derived cells (BMDCs)-tumor cell fusion, one of CSCs origins, can explain EMT in cancer through EMT regulators (TWIST, SPARC, and others).134,136 CAFs induce EMT

1072

C. Zhou et al. / Oral Oncology 48 (2012) 1068–1075

and stemness through a pro-inflammatory signature, which exploits reactive oxygen species to drive a migratory and aggressive phenotype of prostate carcinoma cells.42 In an epithelial breast cancer CD8+ T cells can induce EMT and the resulting tumors had characteristics of breast cancer stem cell (BCSCs), a CD24( / lo)CD44(+) phenotype.137 These indicate inflammation cells may play a critical role in EMT conferring to CSCs. The direct and strong evidence of the association of inflammation, EMT and the CSCs property to adapt to a particular target tissue microenvironment came from the results that the TGF-b-inducedEMT in immortalized human mammary epithelial cells resulted in the acquisition of mesenchymal traits and the expression of stem-cell markers, and stem-like cells isolated either from mouse or human mammary glands or mammary carcinomas expressed EMT markers.108,109 The TGF-b pathway was specifically active in CD44+ cancer cells197 and TGF-b-induced-EMT associated with the selection and expansion of breast cancer stem cells,198 which suggested that TGFb-induced-EMT might represent a common molecular mechanism underlying the anti-cancer stem cells and anti-fibrotic actions of metformin.199 Tetracycline-regulated expression of SNAIL induced the differentiation of mouse embryonic stem cell (MESECs) into mural cells, whereas knockdown of SNAIL expression abrogated TGFb2-induced mural differentiation of MESECs.186 In addition, Activin-Nodal signaling acted through SIP1 to regulate the cell-fate decision between neuroectoderm and mesendoderm in the progression from pluripotency to primary germ layer differentiation.200 TWIST1 was responsible for the regulation of the IKKb/NF-jB and PTEN/AKT pathways and its association of ovarian cancer stem cell differentiation through the expression of hsa-miR-199a/hsa-miR-214, which suggested EMT-associated inflammation and CSCs were regulated by miRNAs.201,202 The above and many other studies have shown that inflammation-EMT and CSCs are not merely spectators in the process of tumor spread but often have a position at center stage, orchestrating and actively participating in the transformation process. Taken together, these lines of evidence challenge the unidirectional view of inflammation only as inducers of carcinogenesis and tumor progression, provocatively suggesting that inflammation may under some circumstances help tumor cell to spread through EMT and CSCs, and warning against previously underestimated dangers and pitfalls of inflammation. Conclusion It is unlikely that inflammation teaches us how to fight or prevent metastasis, however, they can give us a hint of why metastasis occurs. No doubts exist that the more unfavorable life in the primary tumor is for cancer cells, the higher the risk of metastatic spread is.203 Given that inflammation microenvironment, and EMT and CSCs in particular, are now recognized as important determinants of tumor dissemination, a more thorough understanding of their functional role in tumor spread will contribute to the translation from bench to bedside. It is early days to anticipate the successful clinical translation of any of the CSCs-targeted strategies to completely kill cancer cells, however, one should be optimistic about the future of the multitude of therapeutic options offered by increasingly understanding of stem cell biology in cancer metastasis. Conflict of interest statement None declared. Acknowledgements We apologize to those colleagues whose work we could not reference directly due to space constraints. This work was

supported by National Natural Science Foundation of China Grants (Nos. 81072215, 81001210 and 81172580), by the Fundamental Research Funds of the Central Universities of China (2011)) and State Key Laboratory of Oral Diseases Open Funding (SKLODOF2010-05. References 1. Moskovic DJ, Smolarz JR, Stanley D, Jimenez C, Williams MD, Hanna EY, et al. Malignant head and neck paragangliomas: is there an optimal treatment strategy? Head Neck Oncol 2010;2:23. 2. Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat Rev Cancer 2003;3(6):453–8. 3. Mantovani A. Cancer: inflaming metastasis. Nature 2009;457(7225):36–7. 4. Bissell MJ, Hines WC. Why don’t we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat Med 2011;17(3):320–9. 5. Lorusso G, Ruegg C. The tumor microenvironment and its contribution to tumor evolution toward metastasis. Histochem Cell Biol 2008;130(6):1091–103. 6. Mantovani A, Allavena P, Sica A, Balkwill F. Cancer-related inflammation. Nature 2008;454(7203):436–44. 7. Balkwill F, Mantovani A. Inflammation and cancer: back to Virchow? Lancet 2001;357(9255):539–45. 8. Mantovani A. Molecular pathways linking inflammation and cancer. Curr Mol Med 2010;10(4):369–73. 9. Allavena P, Garlanda C, Borrello MG, Sica A, Mantovani A. Pathways connecting inflammation and cancer. Curr Opin Genet Dev 2008;18(1):3–10. 10. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010;140(6):883–99. 11. Kim S, Takahashi H, Lin WW, Descargues P, Grivennikov S, Kim Y, et al. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature 2009;457(7225):102–6. 12. Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the premetastatic niche. Nature 2005;438(7069):820–7. 13. de Visser KE, Eichten A, Coussens LM. Paradoxical roles of the immune system during cancer development. Nat Rev Cancer 2006;6(1):24–37. 14. Markiewski MM, DeAngelis RA, Benencia F, Ricklin-Lichtsteiner SK, Koutoulaki C, Gerard C, et al. Modulation of the antitumor immune response by complement. Nat Immunol 2008;9(11):1225–35. 15. Andreu P, Johansson M, Affara NI, Pucci F, Tan T, Junankar S, et al. FcRgamma activation regulates inflammation-associated squamous carcinogenesis. Cancer Cell 2010;17(2):121–34. 16. Kalluri R. EMT: when epithelial cells decide to become mesenchymal-like cells. J Clin Invest 2009;119(6):1417–9. 17. Singh A, Settleman J. EMT, cancer stem cells and drug resistance. an emerging axis of evil in the war on cancer. Oncogene 2010;29(34):4741–51. 18. Sansone P, Bromberg J. Environment, inflammation, and cancer. Curr Opin Genet Dev 2011;21(1):80–5. 19. Lopez-Novoa JM, Nieto MA. Inflammation and EMT: an alliance towards organ fibrosis and cancer progression. EMBO Mol Med 2009;1(6–7):303–14. 20. Stockinger A, Eger A, Wolf J, Beug H, Foisner R. E-cadherin regulates cell growth by modulating proliferation-dependent beta-catenin transcriptional activity. J Cell Biol 2001;154(6):1185–96. 21. Kalluri R, Weinberg RA. The basics of epithelial–mesenchymal transition. J Clin Invest 2009;119(6):1420–8. 22. Acloque H, Adams MS, Fishwick K, Bronner-Fraser M, Nieto MA. Epithelial– mesenchymal transitions: the importance of changing cell state in development and disease. J Clin Invest 2009;119:1438–49. 23. Radisky DC, LaBarge MA. Epithelial–mesenchymal transition and the stem cell phenotype. Cell Stem Cell 2008;2(6):511–2. 24. Thiery JP, Sleeman JP. Complex networks orchestrate epithelial–mesenchymal transitions. Nat Rev Mol Cell Biol 2006;7(2):131–42. 25. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial–mesenchymal transitions in development and disease. Cell 2009;139(5):871–90. 26. Brabletz T, Jung A, Spaderna S, Hlubek F, Kirchner T. Opinion: migrating cancer stem cells – an integrated concept of malignant tumour progression. Nat Rev Cancer 2005;5(9):744–9. 27. Reuter R, Grunewald B, Leptin M. A role for the mesoderm in endodermal migration and morphogenesis in Drosophila. Development 1993;119(4):1135–45. 28. LaBonne C, Bronner-Fraser M. Snail-related transcriptional repressors are required in Xenopus for both the induction of the neural crest and its subsequent migration. Dev Biol 2000;221(1):195–205. 29. Cho KW, Blumberg B, Steinbeisser H, De Robertis EM. Molecular nature of Spemann’s organizer: the role of the Xenopus homeobox gene goosecoid. Cell 1991;67(6):1111–20. 30. Scheel C, Eaton EN, Li SH, Chaffer CL, Reinhardt F, Kah KJ, et al. Paracrine and autocrine signals induce and maintain mesenchymal and stem cell states in the breast. Cell 2011;145(6):926–40. 31. Oft M, Peli J, Rudaz C, Schwarz H, Beug H, Reichmann E. TGF-beta1 and Ha-Ras collaborate in modulating the phenotypic plasticity and invasiveness of epithelial tumor cells. Genes Dev 1996;10(19):2462–77.

C. Zhou et al. / Oral Oncology 48 (2012) 1068–1075 32. Bates RC, Mercurio AM. Tumor necrosis factor-alpha stimulates the epithelialto-mesenchymal transition of human colonic organoids. Mol Biol Cell 2003;14(5):1790–800. 33. Zavadil J, Cermak L, Soto-Nieves N, Bottinger EP. Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO J 2004;23(5):1155–65. 34. Karhadkar SS, Bova GS, Abdallah N, Dhara S, Gardner D, Maitra A, et al. Hedgehog signalling in prostate regeneration, neoplasia and metastasis. Nature 2004;431(7009):707–12. 35. Yook JI, Li XY, Ota I, Hu C, Kim HS, Kim NH, et al. A Wnt-Axin2-GSK3beta cascade regulates Snail1 activity in breast cancer cells. Nat Cell Biol 2006;8(12):1398–406. 36. Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell 2006;124(2):263–6. 37. Chaffer CL, Weinberg RA. A perspective on cancer cell metastasis. Science 2011;331(6024):1559–64. 38. Lochter A, Galosy S, Muschler J, Freedman N, Werb Z, Bissell MJ. Matrix metalloproteinase stromelysin-1 triggers a cascade of molecular alterations that leads to stable epithelial-to-mesenchymal conversion and a premalignant phenotype in mammary epithelial cells. J Cell Biol 1997;139(7):1861–72. 39. Thiery JP. Epithelial–mesenchymal transitions in tumour progression. Nat Rev Cancer 2002;2(6):442–54. 40. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer 2006;6(5):392–401. 41. Lebret SC, Newgreen DF, Thompson EW, Ackland ML. Induction of epithelial to mesenchymal transition in PMC42-LA human breast carcinoma cells by carcinoma-associated fibroblast secreted factors. Breast Cancer Res 2007;9(1):R19. 42. Giannoni E, Bianchini F, Calorini L, Chiarugi P. Cancer associated fibroblasts exploit reactive oxygen species through a proinflammatory signature leading to epithelial mesenchymal transition and stemness. Antioxid Redox Signal 2011;14(12):2361–71. 43. Greten FR, Eckmann L, Greten TF, Park JM, Li ZW, Egan LJ, et al. IKKbeta links inflammation and tumorigenesis in a mouse model of colitis-associated cancer. Cell 2004;118(3):285–96. 44. Pikarsky E, Porat RM, Stein I, Abramovitch R, Amit S, Kasem S, et al. NF-kappaB functions as a tumour promoter in inflammation-associated cancer. Nature 2004;431(7007):461–6. 45. Maeda S, Hsu LC, Liu H, Bankston LA, Iimura M, Kagnoff MF, et al. Nod2 mutation in Crohn’s disease potentiates NF-kappaB activity and IL-1beta processing. Science 2005;307(5710):734–8. 46. Karin M. NF-kappaB and cancer: mechanisms and targets. Mol Carcinog 2006;45(6):355–61. 47. Bachelder RE, Yoon SO, Franci C, de Herreros AG, Mercurio AM. Glycogen synthase kinase-3 is an endogenous inhibitor of Snail transcription: implications for the epithelial–mesenchymal transition. J Cell Biol 2005;168(1):29–33. 48. Julien S, Puig I, Caretti E, Bonaventure J, Nelles L, van Roy F, et al. Activation of NF-kappaB by Akt upregulates Snail expression and induces epithelium mesenchyme transition. Oncogene 2007;26(53):7445–56. 49. Wu Y, Deng J, Rychahou PG, Qiu S, Evers BM, Zhou BP. Stabilization of snail by NF-kappaB is required for inflammation-induced cell migration and invasion. Cancer Cell 2009;15(5):416–28. 50. Tlsty TD, Coussens LM. Tumor stroma and regulation of cancer development. Annu Rev Pathol 2006;1:119–50. 51. Yu H, Pardoll D, Jove R. STATs in cancer inflammation and immunity: a leading role for STAT3. Nat Rev Cancer 2009;9(11):798–809. 52. Rehman AO. Wang CY.CXCL12/SDF-1 alpha activates NF-kappaB and promotes oral cancer invasion through the Carma3/Bcl10/Malt1 complex. Int. J Oral Sci 2009;1(3):105–18. 53. Strippoli R, Benedicto I, Perez Lozano ML, Cerezo A, Lopez-Cabrera M, del Pozo MA. Epithelial-to-mesenchymal transition of peritoneal mesothelial cells is regulated by an ERK/NF-kappaB/Snail1 pathway. Dis Model Mech 2008;1(4– ):264–74. 54. Lin K, Baritaki S, Militello L, Malaponte G, Bevelacqua Y, Bonavida B. The role of B-RAF mutations in melanoma and the induction of EMT via dysregulation of the NF-kappaB/Snail/RKIP/PTEN circuit. Genes Cancer 2010;1(5):409–20. 55. Pantuck AJ, An J, Liu H, Rettig MB. NF-kappaB-dependent plasticity of the epithelial to mesenchymal transition induced by Von Hippel–Lindau inactivation in renal cell carcinomas. Cancer Res 2010;70(2):752–61. 56. Baritaki S, Chapman A, Yeung K, Spandidos DA, Palladino M, Bonavida B. Inhibition of epithelial to mesenchymal transition in metastatic prostate cancer cells by the novel proteasome inhibitor, NPI-0052: pivotal roles of Snail repression and RKIP induction. Oncogene 2009;28(40):3573–85. 57. Li QQ, Chen ZQ, Cao XX, Xu JD, Xu JW, Chen YY, et al. Involvement of NFkappaB/miR-448 regulatory feedback loop in chemotherapy-induced epithelial–mesenchymal transition of breast cancer cells. Cell Death Differ 2011;18(1):16–25. 58. Gavert N, Vivanti A, Hazin J, Brabletz T, Ben-Ze’ev A. L1-mediated colon cancer cell metastasis does not require changes in EMT and cancer stem cell markers. Mol Cancer Res 2011;9(1):14–24. 59. de Graauw M, van Miltenburg MH, Schmidt MK, Pont C, Lalai R, Kartopawiro J, et al. Annexin A1 regulates TGF-beta signaling and promotes metastasis formation of basal-like breast cancer cells. Proc Natl Acad Sci USA 2010;107(14):6340–5.

1073

60. Yang J, Weinberg RA. Epithelial–mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell 2008;14(6):818–29. 61. Franco DL, Mainez J, Vega S, Sancho P, Murillo MM, de Frutos CA, et al. Snail1 suppresses TGF-beta-induced apoptosis and is sufficient to trigger EMT in hepatocytes. J Cell Sci 2010;123(Pt 20):3467–77. 62. Hoot KE, Lighthall J, Han G, Lu SL, Li A, Ju W, et al. Keratinocyte-specific Smad2 ablation results in increased epithelial–mesenchymal transition during skin cancer formation and progression. J Clin Invest 2008;118(8):2722–32. 63. Peinado H, Quintanilla M, Cano A. Transforming growth factor beta-1 induces snail transcription factor in epithelial cell lines: mechanisms for epithelial mesenchymal transitions. J Biol Chem 2003;278(23):21113–23. 64. Bardeesy N, Cheng KH, Berger JH, Chu GC, Pahler J, Olson P, et al. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev 2006;20(22):3130–46. 65. Valcourt U, Kowanetz M, Niimi H, Heldin CH, Moustakas A. TGF-beta and the Smad signaling pathway support transcriptomic reprogramming during epithelial–mesenchymal cell transition. Mol Biol Cell 2005;16(4):1987–2002. 66. Dooley S, Hamzavi J, Ciuclan L, Godoy P, Ilkavets I, Ehnert S, et al. Hepatocytespecific Smad7 expression attenuates TGF-beta-mediated fibrogenesis and protects against liver damage. Gastroenterology 2008;135(2):642–59. 67. Papageorgis P, Lambert AW, Ozturk S, Gao F, Pan H, Manne U, et al. Smad signaling is required to maintain epigenetic silencing during breast cancer progression. Cancer Res 2010;70(3):968–78. 68. Bakin AV, Rinehart C, Tomlinson AK, Arteaga CL. P38 mitogen-activated protein kinase is required for TGFbeta-mediated fibroblastic transdifferentiation and cell migration. J Cell Sci 2002;115(Pt 15):3193–206. 69. Perlman R, Schiemann WP, Brooks MW, Lodish HF, Weinberg RA. TGF-betainduced apoptosis is mediated by the adapter protein Daxx that facilitates JNK activation. Nat Cell Biol 2001;3(8):708–14. 70. Zavadil J, Bitzer M, Liang D, Yang YC, Massimi A, Kneitz S, et al. Genetic programs of epithelial cell plasticity directed by transforming growth factorbeta. Proc Natl Acad Sci USA 2001;98(12):6686–91. 71. Arsura M, Panta GR, Bilyeu JD, Cavin LG, Sovak MA, Oliver AA, et al. Transient activation of NF-kappaB through a TAK1/IKK kinase pathway by TGF-beta1 inhibits AP-1/SMAD signaling and apoptosis: implications in liver tumor formation. Oncogene 2003;22(3):412–25. 72. Park JI, Lee MG, Cho K, Park BJ, Chae KS, Byun DS, et al. Transforming growth factor-beta1 activates interleukin-6 expression in prostate cancer cells through the synergistic collaboration of the Smad2, p38-NF-kappaB, JNK, and Ras signaling pathways. Oncogene 2003;22(28):4314–32. 73. Neil JR, Schiemann WP. Altered TAB1:I kappaB kinase interaction promotes transforming growth factor beta-mediated nuclear factor-kappaB activation during breast cancer progression. Cancer Res 2008;68(5):1462–70. 74. Chaudhury A, Hussey GS, Ray PS, Jin G, Fox PL, Howe PH. TGF-beta-mediated phosphorylation of hnRNP E1 induces EMT via transcript-selective translational induction of Dab2 and ILEI. Nat Cell Biol 2010;12(3):286–93. 75. Rajasekaran SA, Huynh TP, Wolle DG, Espineda CE, Inge LJ, Skay A, et al. Na, KATPase subunits as markers for epithelial–mesenchymal transition in cancer and fibrosis. Mol Cancer Ther 2010;9(6):1515–24. 76. Natsuizaka M, Ohashi S, Wong GS, Ahmadi A, Kalman RA, Budo D, et al. Insulin-like growth factor-binding protein-3 promotes transforming growth factor-{beta}1-mediated epithelial-to-mesenchymal transition and motility in transformed human esophageal cells. Carcinogenesis 2010;31(8):1344–53. 77. Kong B, Michalski CW, Hong X, Valkovskaya N, Rieder S, Abiatari I, et al. AZGP1 is a tumor suppressor in pancreatic cancer inducing mesenchymal-toepithelial transdifferentiation by inhibiting TGF-beta-mediated ERK signaling. Oncogene 2010;29(37):5146–58. 78. Micalizzi DS, Wang CA, Farabaugh SM, Schiemann WP, Ford HL. Homeoprotein Six1 increases TGF-beta type I receptor and converts TGF-beta signaling from suppressive to supportive for tumor growth. Cancer Res 2010;70(24):10371–80. 79. Moreno-Bueno G, Portillo F, Cano A. Transcriptional regulation of cell polarity in EMT and cancer. Oncogene 2008;27(55):6958–69. 80. Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat Rev Cancer 2007;7(6):415–28. 81. Braun J, Hoang-Vu C, Dralle H, Huttelmaier S. Downregulation of microRNAs directs the EMT and invasive potential of anaplastic thyroid carcinomas. Oncogene 2010;29(29):4237–44. 82. Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, et al. The miR200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol 2008;10(5):593–601. 83. Korpal M, Lee ES, Hu G, Kang Y. The miR-200 family inhibits epithelial– mesenchymal transition and cancer cell migration by direct targeting of Ecadherin transcriptional repressors ZEB1 and ZEB2. J Biol Chem 2008;283(22):14910–4. 84. Kong W, Yang H, He L, Zhao JJ, Coppola D, Dalton WS, et al. MicroRNA-155 is regulated by the transforming growth factor beta/Smad pathway and contributes to epithelial cell plasticity by targeting RhoA. Mol Cell Biol 2008;28(22):6773–84. 85. Zhu S, Si ML, Wu H, Mo YY. MicroRNA-21 targets the tumor suppressor gene tropomyosin 1 (TPM1). J Biol Chem 2007;282(19):14328–36. 86. Zhu S, Wu H, Wu F, Nie D, Sheng S, Mo YY. MicroRNA-21 targets tumor suppressor genes in invasion and metastasis. Cell Res 2008;18(3):350–9. 87. Dumont N, Wilson MB, Crawford YG, Reynolds PA, Sigaroudinia M, Tlsty TD. Sustained induction of epithelial to mesenchymal transition activates DNA

1074

88.

89.

90.

91.

92.

93.

94.

95.

96.

97.

98.

99.

100.

101.

102.

103.

104.

105.

106.

107.

108.

109.

110. 111.

112. 113.

C. Zhou et al. / Oral Oncology 48 (2012) 1068–1075 methylation of genes silenced in basal-like breast cancers. Proc Natl Acad Sci USA 2008;105(39):14867–72. Yan C, Grimm WA, Garner WL, Qin L, Travis T, Tan N, et al. Epithelial to mesenchymal transition in human skin wound healing is induced by tumor necrosis factor-alpha through bone morphogenic protein-2. Am J Pathol 2010;176(5):2247–58. Baran B, Bechyne I, Siedlar M, Szpak K, Mytar B, Sroka J, et al. Blood monocytes stimulate migration of human pancreatic carcinoma cells in vitro: the role of tumour necrosis factor-alpha. Eur J Cell Biol 2009;88(12):743–52. Takahashi E, Nagano O, Ishimoto T, Yae T, Suzuki Y, Shinoda T, et al. Tumor necrosis factor-alpha regulates transforming growth factor-beta-dependent epithelial–mesenchymal transition by promoting hyaluronan-CD44-moesin interaction. J Biol Chem 2010;285(6):4060–73. Luo JL, Tan W, Ricono JM, Korchynskyi O, Zhang M, Gonias SL, et al. Nuclear cytokine-activated IKKalpha controls prostate cancer metastasis by repressing Maspin. Nature 2007;446(7136):690–4. Luo JL, Maeda S, Hsu LC, Yagita H, Karin M. Inhibition of NF-kappaB in cancer cells converts inflammation- induced tumor growth mediated by TNFalpha to TRAIL-mediated tumor regression. Cancer cell 2004;6(3):297–305. Zhang Q, Helfand BT, Jang TL, Zhu LJ, Chen L, Yang XJ, et al. Nuclear factorkappaB-mediated transforming growth factor-beta-induced expression of vimentin is an independent predictor of biochemical recurrence after radical prostatectomy. Clin Cancer Res 2009;15(10):3557–67. Huber MA, Azoitei N, Baumann B, Grunert S, Sommer A, Pehamberger H, et al. NF-kappaB is essential for epithelial–mesenchymal transition and metastasis in a model of breast cancer progression. J Clin Invest 2004;114(4):569–81. Neil JR, Johnson KM, Nemenoff RA, Schiemann WP. Cox-2 inactivates Smad signaling and enhances EMT stimulated by TGF-beta through a PGE2dependent mechanisms. Carcinogenesis 2008;29(11):2227–35. You HJ, How T, Blobe GC. The type III transforming growth factor-beta receptor negatively regulates nuclear factor kappa B signaling through its interaction with beta-arrestin2. Carcinogenesis 2009;30(8):1281–7. Neil JR, Tian M, Schiemann WP. X-linked inhibitor of apoptosis protein and its E3 ligase activity promote transforming growth factor-{beta}-mediated nuclear factor-{kappa}B activation during breast cancer progression. J Biol Chem 2009;284(32):21209–17. Chua HL, Bhat-Nakshatri P, Clare SE, Morimiya A, Badve S, Nakshatri H. NFkappaB represses E-cadherin expression and enhances epithelial to mesenchymal transition of mammary epithelial cells: potential involvement of ZEB-1 and ZEB-2. Oncogene 2007;26(5):711–24. Musteanu M, Blaas L, Mair M, Schlederer M, Bilban M, Tauber S, et al. Stat3 is a negative regulator of intestinal tumor progression in Apc(Min) mice. Gastroenterology 2010;138(3):1003–11. e1–5. Evans AJ, Russell RC, Roche O, Burry TN, Fish JE, Chow VW, et al. VHL promotes E2 box-dependent E-cadherin transcription by HIF-mediated regulation of SIP1 and snail. Mol Cell Biol 2007;27(1):157–69. Yang MH, Wu MZ, Chiou SH, Chen PM, Chang SY, Liu CJ, et al. Direct regulation of TWIST by HIF-1alpha promotes metastasis. Nat Cell Biol 2008;10(3):295–305. Krishnamachary B, Zagzag D, Nagasawa H, Rainey K, Okuyama H, Baek JH, et al. Hypoxia-inducible factor-1-dependent repression of E-cadherin in von Hippel–Lindau tumor suppressor-null renal cell carcinoma mediated by TCF3, ZFHX1A, and ZFHX1B. Cancer Res 2006;66(5):2725–31. Peinado H, Del Carmen Iglesias-de la Cruz M, Olmeda D, Csiszar K, Fong KS, Vega S, et al. A molecular role for lysyl oxidase-like 2 enzyme in snail regulation and tumor progression. EMBO J 2005;24(19):3446–58. Min C, Kirsch KH, Zhao Y, Jeay S, Palamakumbura AH, Trackman PC, et al. The tumor suppressor activity of the lysyl oxidase propeptide reverses the invasive phenotype of Her-2/neu-driven breast cancer. Cancer Res 2007;67(3):1105–12. Gebeshuber CA, Zatloukal K, Martinez J. MiR-29a suppresses tristetraprolin, which is a regulator of epithelial polarity and metastasis. EMBO Rep 2009;10(4):400–5. Ma L, Young J, Prabhala H, Pan E, Mestdagh P, Muth D, et al. MiR-9, a MYC/ MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nat Cell Biol 2010;12(3):247–56. Bazzoni F, Rossato M, Fabbri M, Gaudiosi D, Mirolo M, Mori L, et al. Induction and regulatory function of miR-9 in human monocytes and neutrophils exposed to proinflammatory signals. Proc Natl Acad Sci USA 2009;106(13):5282–7. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. The epithelial– mesenchymal transition generates cells with properties of stem cells. Cell 2008;133(4):704–15. Morel AP, Lievre M, Thomas C, Hinkal G, Ansieau S, Puisieux A. Generation of breast cancer stem cells through epithelial–mesenchymal transition. PLoS One 2008;3(8):e2888. Tang C, Ang BT, Pervaiz S. Cancer stem cell: target for anti-cancer therapy. FASEB J 2007;21(14):3777–85. Clarke MF, Dick JE, Dirks PB, Eaves CJ, Jamieson CH, Jones DL, et al. Cancer stem cells – perspectives on current status and future directions: AACR workshop on cancer stem cells. Cancer Res 2006;66(19):9339–44. Frank NY, Schatton T, Frank MH. The therapeutic promise of the cancer stem cell concept. J Clin Invest 2010;120(1):41–50. Visvader JE, Lindeman GJ. Cancer stem cells in solid tumours: accumulating evidence and unresolved questions. Nat Rev Cancer 2008;8(10):755–68.

114. Tabor MH, Clay MR, Owen JH, Bradford CR, Carey TE, Wolf GT, et al. Head and neck cancer stem cells: the side population. Laryngoscope 2011;121(3):527–33. 115. Prince ME, Sivanandan R, Kaczorowski A, Wolf GT, Kaplan MJ, Dalerba P, et al. Identification of a subpopulation of cells with cancer stem cell properties in head and neck squamous cell carcinoma. Proc Natl Acad Sci USA 2007;104(3):973–8. 116. Okamoto A, Chikamatsu K, Sakakura K, Hatsushika K, Takahashi G, Masuyama K. Expansion and characterization of cancer stem-like cells in squamous cell carcinoma of the head and neck. Oral Oncol 2009;45(7):633–9. 117. Chen YC, Chen YW, Hsu HS, Tseng LM, Huang PI, Lu KH, et al. Aldehyde dehydrogenase 1 is a putative marker for cancer stem cells in head and neck squamous cancer. Biochem Biophys Res Commun 2009;385(3):307–13. 118. Clay MR, Tabor M, Owen JH, Carey TE, Bradford CR, Wolf GT, et al. Singlemarker identification of head and neck squamous cell carcinoma cancer stem cells with aldehyde dehydrogenase. Head Neck 2010;32(9):1195–201. 119. Tang AL, Hauff SJ, Owen JH, Graham MP, Czerwinski MJ, Park JJ, et al. UM-SCC104: A New human papillomavirus-16-positive cancer stem cell-containing head and neck squamous cell carcinoma cell line. Head Neck 2011(December). http://dx.doi.org/10.1002/hed.21962. 120. Davis SJ, Divi V, Owen JH, Bradford CR, Carey TE, Papagerakis S, et al. Metastatic potential of cancer stem cells in head and neck squamous cell carcinoma. Arch Otolaryngol Head Neck Surg 2010;136(12):1260–6. 121. Song J, Chang I, Chen Z, Kang M, Wang CY. Characterization of side populations in HNSCC: highly invasive, chemoresistant and abnormal Wnt signaling. PLoS One 2010;5(7):e11456. 122. Goldie SJ, Mulder KW, Tan DW, Lyons SK, Sims AH, Watt FM. FRMD4A upregulation in human squamous cell carcinoma promotes tumor growth and metastasis and is associated with poor prognosis. Cancer Res 2012;May 7 [Epub ahead of print]. 123. Greaves M. Cancer stem cells: back to Darwin? Semin Cancer Biol 2010;20(2):65–70. 124. Kim CF, Jackson EL, Woolfenden AE, Lawrence S, Babar I, Vogel S, et al. Identification of bronchioalveolar stem cells in normal lung and lung cancer. Cell 2005;121(6):823–35. 125. Shackleton M, Vaillant F, Simpson KJ, Stingl J, Smyth GK, Asselin-Labat ML, et al. Generation of a functional mammary gland from a single stem cell. Nature 2006;439(7072):84–8. 126. Miyamoto T, Weissman IL, Akashi K. AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. Proc Natl Acad Sci USA 2000;97(13):7521–6. 127. Welm AL, Kim S, Welm BE, Bishop JM. MET and MYC cooperate in mammary tumorigenesis. Proc Natl Acad Sci USA 2005;102(12):4324–9. 128. Houghton J, Stoicov C, Nomura S, Rogers AB, Carlson J, Li H, et al. Gastric cancer originating from bone marrow-derived cells. Science 2004;306(5701):1568–71. 129. Eastham AM, Spencer H, Soncin F, Ritson S, Merry CL, Stern PL, et al. Epithelial–mesenchymal transition events during human embryonic stem cell differentiation. Cancer Res 2007;67(23):11254–62. 130. Jamieson CH, Ailles LE, Dylla SJ, Muijtjens M, Jones C, Zehnder JL, et al. Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. New Engl J Med 2004;351(7):657–67. 131. Jaiswal S, Traver D, Miyamoto T, Akashi K, Lagasse E, Weissman IL. Expression of BCR/ABL and BCL-2 in myeloid progenitors leads to myeloid leukemias. Proc Natl Acad Sci USA 2003;100(17):10002–7. 132. Krivtsov AV, Twomey D, Feng Z, Stubbs MC, Wang Y, Faber J, et al. Transformation from committed progenitor to leukaemia stem cell initiated by MLL-AF9. Nature 2006;442(7104):818–22. 133. Vescovi AL, Galli R, Reynolds BA. Brain tumour stem cells. Nat Rev Cancer 2006;6(6):425–36. 134. Bjerkvig R, Tysnes BB, Aboody KS, Najbauer J, Terzis AJ. Opinion: the origin of the cancer stem cell: current controversies and new insights. Nat Rev Cancer 2005;5(11):899–904. 135. Pawelek JM, Chakraborty AK. Fusion of tumour cells with bone marrowderived cells: a unifying explanation for metastasis. Nat Rev Cancer 2008;8(5):377–86. 136. Pawelek JM, Chakraborty AK. The cancer cell–leukocyte fusion theory of metastasis. Adv Cancer Res 2008;101:397–444. 137. Santisteban M, Reiman JM, Asiedu MK, Behrens MD, Nassar A, Kalli KR, et al. Immune-induced epithelial to mesenchymal transition in vivo generates breast cancer stem cells. Cancer Res 2009;69(7):2887–95. 138. Pawelek JM. Tumour-cell fusion as a source of myeloid traits in cancer. Lancet Oncol 2005;6(12):988–93. 139. Wellner U, Schubert J, Burk UC, Schmalhofer O, Zhu F, Sonntag A, et al. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemnessinhibiting microRNAs. Nat Cell Biol 2009;11(12):1487–95. 140. Biddle A, Liang X, Gammon L, Fazil B, Harper LJ, Emich H, et al. Cancer stem cells in squamous cell carcinoma switch between two distinct phenotypes that are preferentially migratory or proliferative. Cancer Res 2011;71(15):5317–26. 141. Basu D, Montone KT, Wang LP, Gimotty PA, Hammond R, Diehl JA, et al. Detecting and targeting mesenchymal-like subpopulations within squamous cell carcinomas. Cell Cycle 2011;10(12):2008–16. 142. Sunwoo JB. A cisplatin-resistant subpopulation of mesenchymal-like cells in head and neck squamous cell carcinoma. Cell Cycle 2011;10(17):2834–5.

C. Zhou et al. / Oral Oncology 48 (2012) 1068–1075 143. Judd NP, Winkler AE, Murillo-Sauca O, Brotman JJ, Law JH, Lewis Jr JS, et al. ERK1/2 regulation of CD44 modulates oral cancer aggressiveness. Cancer Res 2012;72(1):365–74. 144. Chen C, Wei Y, Hummel M, Hoffmann TK, Gross M, Kaufmann AM, et al. Evidence for epithelial–mesenchymal transition in cancer stem cells of head and neck squamous cell carcinoma. PLoS One 2011;6(1):e16466. 145. Zhang Z, Filho MS, Nör JE. The biology of head and neck cancer stem cells. Oral Oncol 2012;48(1):1–9. 146. Scheel C, Onder T, Karnoub A, Weinberg RA. Adaptation versus selection: the origins of metastatic behavior. Cancer Res 2007;67(24):11476–9 [discussion 9–80]. 147. Biddle A, Mackenzie IC. Cancer stem cells and EMT in carcinoma. Cancer Metastasis Rev 2012;Feb 3 [Epub ahead of print]. 148. Moore KA, Lemischka IR. Stem cells and their niches. Science 2006;311(5769):1880–5. 149. Li L, Xie T. Stem cell niche: structure and function. Annu Rev Cell Dev Biol 2005;21:605–31. 150. Scadden DT. The stem-cell niche as an entity of action. Nature 2006;441(7097):1075–9. 151. Xie T, Spradling AC. A niche maintaining germ line stem cells in the Drosophila ovary. Science 2000;290(5490):328–30. 152. Lin H. The stem-cell niche theory: lessons from flies. Nat Rev Genet 2002;3(12):931–40. 153. Li L, Clevers H. Coexistence of quiescent and active adult stem cells in mammals. Science 2010;327(5965):542–5. 154. Lutolf MP, Gilbert PM, Blau HM. Designing materials to direct stem-cell fate. Nature 2009;462(7272):433–41. 155. Rossi DJ, Jamieson CH, Weissman IL. Stems cells and the pathways to aging and cancer. Cell 2008;132(4):681–96. 156. Kuhn NZ, Tuan RS. Regulation of stemness and stem cell niche of mesenchymal stem cells: implications in tumorigenesis and metastasis. J Cell Physiol 2010;222(2):268–77. 157. Gilbert PM, Havenstrite KL, Magnusson KE, Sacco A, Leonardi NA, Kraft P, et al. Substrate elasticity regulates skeletal muscle stem cell self-renewal in culture. Science 2010;329(5995):1078–81. 158. Stewart MH, Bendall SC, Bhatia M. Deconstructing human embryonic stem cell cultures: niche regulation of self-renewal and pluripotency. J Mol Med (Berl) 2008;86(8):875–86. 159. Schroeder T. Hematopoietic stem cell heterogeneity: subtypes, not unpredictable behavior. Cell Stem Cell 2010;6(3):203–7. 160. Bhatia M. Developmental biology. Microenvironment mimicry. Science 2010;329(5995):1024–5. 161. Gilbertson RJ, Gutmann DH. Tumorigenesis in the brain: location, location, location. Cancer Res 2007;67(12):5579–82. 162. Gilbertson RJ, Rich JN. Making a tumour’s bed: glioblastoma stem cells and the vascular niche. Nat Rev Cancer 2007;7(10):733–6. 163. Johnston LA. Competitive interactions between cells: death, growth, and geography. Science 2009;324(5935):1679–82. 164. Spradling A, Drummond-Barbosa D, Kai T. Stem cells find their niche. Nature 2001;414(6859):98–104. 165. Connelly JT, Gautrot JE, Trappmann B, Tan DW, Donati G, Huck WT, et al. Actin and serum response factor transduce physical cues from the microenvironment to regulate epidermal stem cell fate decisions. Nat Cell Biol 2010;12(7):711–8. 166. Iglesias-Bartolome R, Gutkind JS. Keeping the epidermal stem cell niche in shape. Cell Stem Cell 2010;7(2):143–5. 167. Castilho RM, Squarize CH, Leelahavanichkul K, Zheng Y, Bugge T, Gutkind JS. Rac1 is required for epithelial stem cell function during dermal and oral mucosal wound healing but not for tissue homeostasis in mice. PLoS One 2010;5(5):e10503. 168. Iglesias-Bartolome R, Gutkind JS. Signaling circuitries controlling stem cell fate: to be or not to be. Curr Opin Cell Biol 2011;23(6):716–23. 169. Kiger AA, Jones DL, Schulz C, Rogers MB, Fuller MT. Stem cell self-renewal specified by JAK-STAT activation in response to a support cell cue. Science 2001;294(5551):2542–5. 170. Schulz C, Kiger AA, Tazuke SI, Yamashita YM, Pantalena-Filho LC, Jones DL, et al. A misexpression screen reveals effects of bag-of-marbles and TGF beta class signaling on the Drosophila male germ-line stem cell lineage. Genetics 2004;167(2):707–23. 171. Song X, Wong MD, Kawase E, Xi R, Ding BC, McCarthy JJ, et al. Bmp signals from niche cells directly repress transcription of a differentiation-promoting gene, bag of marbles, in germline stem cells in the Drosophila ovary. Development 2004;131(6):1353–64. 172. Tulina N, Matunis E. Control of stem cell self-renewal in Drosophila spermatogenesis by JAK-STAT signaling. Science 2001;294(5551):2546–9. 173. Morrison SJ, Kimble J. Asymmetric and symmetric stem-cell divisions in development and cancer. Nature 2006;441(7097):1068–74. 174. Charafe-Jauffret E, Ginestier C, Iovino F, Wicinski J, Cervera N, Finetti P, et al. Breast cancer cell lines contain functional cancer stem cells with metastatic capacity and a distinct molecular signature. Cancer Res 2009;69(4):1302–13. 175. Schafer ZT, Brugge JS. IL-6 involvement in epithelial cancers. J Clin Invest 2007;117(12):3660–3. 176. Todaro M, Alea MP, Di Stefano AB, Cammareri P, Vermeulen L, Iovino F, et al. Colon cancer stem cells dictate tumor growth and resist cell death by production of interleukin-4. Cell Stem Cell 2007;1(4):389–402.

1075

177. Sansone P, Storci G, Tavolari S, Guarnieri T, Giovannini C, Taffurelli M, et al. IL6 triggers malignant features in mammospheres from human ductal breast carcinoma and normal mammary gland. J Clin Invest 2007;117(12):3988–4002. 178. Charafe-Jauffret E, Ginestier C, Iovino F, Tarpin C, Diebel M, Esterni B, et al. Aldehyde dehydrogenase 1-positive cancer stem cells mediate metastasis and poor clinical outcome in inflammatory breast cancer. Clin Cancer Res 2010;16(1):45–55. 179. Arwert EN, Lal R, Quist S, Rosewell I, van Rooijen N, Watt FM. Tumor formation initiated by nondividing epidermal cells via an inflammatory infiltrate. Proc Natl Acad Sci USA 2010;107(46):19903–8. 180. Fortunel N, Hatzfeld J, Aoustin L, Batard P, Ducos K, Monier MN, et al. Specific dose-response effects of TGF-beta1 on developmentally distinct hematopoietic stem/progenitor cells from human umbilical cord blood. Hematol J 2000;1(2):126–35. 181. Chabanon A, Desterke C, Rodenburger E, Clay D, Guerton B, Boutin L, et al. A cross-talk between stromal cell-derived factor-1 and transforming growth factor-beta controls the quiescence/cycling switch of CD34(+) progenitors through FoxO3 and mammalian target of rapamycin. Stem Cells 2008;26(12):3150–61. 182. Watabe T, Nishihara A, Mishima K, Yamashita J, Shimizu K, Miyazawa K, et al. TGF-beta receptor kinase inhibitor enhances growth and integrity of embryonic stem cell-derived endothelial cells. J Cell Biol 2003;163(6):1303–11. 183. Kokudo T, Suzuki Y, Yoshimatsu Y, Yamazaki T, Watabe T, Miyazono K. Snail is required for TGFbeta-induced endothelial-mesenchymal transition of embryonic stem cell-derived endothelial cells. J Cell Sci 2008;121(Pt ):3317–24. 184. Challen GA, Boles NC, Chambers SM, Goodell MA. Distinct hematopoietic stem cell subtypes are differentially regulated by TGF-beta1. Cell Stem Cell 2010;6(3):265–78. 185. Oguma K, Oshima H, Aoki M, Uchio R, Naka K, Nakamura S, et al. Activated macrophages promote Wnt signalling through tumour necrosis factor-alpha in gastric tumour cells. EMBO J 2008;27(12):1671–81. 186. Umar S, Sarkar S, Wang Y, Singh P. Functional cross-talk between beta-catenin and NFkappaB signaling pathways in colonic crypts of mice in response to progastrin. J Biol Chem 2009;284(33):22274–84. 187. Chen FH, Tuan RS. Mesenchymal stem cells in arthritic diseases. Arthritis Res Ther 2008;10(5):223. 188. Yu C, Kastin AJ, Pan W. TNF reduces LIF endocytosis despite increasing NFkappaB-mediated gp130 expression. J Cell Physiol 2007;213(1):161–6. 189. Squarize CH, Castilho RM, Sriuranpong V, Pinto Jr DS, Gutkind JS. Molecular cross-talk between the NFkappaB and STAT3 signaling pathways in head and neck squamous cell carcinoma. Neoplasia 2006;8(9):733–46. 190. Bhave SL, Teknos TN, Pan Q. Molecular parameters of head and neck cancer metastasis. Crit Rev Eukaryot Gene Expr 2011;21(2):143–53. 191. Sriuranpong V, Park JI, Amornphimoltham P, Patel V, Nelkin BD, Gutkind JS. Epidermal growth factor receptor-independent constitutive activation of STAT3 in head and neck squamous cell carcinoma is mediated by the autocrine/paracrine stimulation of the interleukin 6/gp130 cytokine system. Cancer Res 2003;63(11):2948–56. 192. North TE, Goessling W, Walkley CR, Lengerke C, Kopani KR, Lord AM, et al. Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature 2007;447(7147):1007–11. 193. Maitland NJ, Collins AT. Inflammation as the primary aetiological agent of human prostate cancer: a stem cell connection? J Cell Biochem 2008;105(4):931–9. 194. Lee P, Lee DJ, Chan C, Chen SW, Ch’en I, Jamora C. Dynamic expression of epidermal caspase 8 simulates a wound healing response. Nature 2009;458(7237):519–23. 195. Rieger MA, Hoppe PS, Smejkal BM, Eitelhuber AC, Schroeder T. Hematopoietic cytokines can instruct lineage choice. Science 2009;325(5937):217–8. 196. Bendall SC, Stewart MH, Menendez P, George D, Vijayaragavan K, Werbowetski-Ogilvie T, et al. IGF and FGF cooperatively establish the regulatory stem cell niche of pluripotent human cells in vitro. Nature 2007;448(7157):1015–21. 197. Shipitsin M, Campbell LL, Argani P, Weremowicz S, Bloushtain-Qimron N, Yao J, et al. Molecular definition of breast tumor heterogeneity. Cancer Cell 2007;11(3):259–73. 198. Ben-Porath I, Thomson MW, Carey VJ, Ge R, Bell GW, Regev A, et al. An embryonic stem cell-like gene expression signature in poorly differentiated aggressive human tumors. Nat Genet 2008;40(5):499–507. 199. Cufi S, Vazquez-Martin A, Oliveras-Ferraros C, Martin-Castillo B, Joven J, Menendez JA. Metformin against TGFbeta-induced epithelial-tomesenchymal transition (EMT): from cancer stem cells to aging-associated fibrosis. Cell Cycle 2010;9(22):4461–8. 200. Chng Z, Teo A, Pedersen RA, Vallier L. SIP1 mediates cell-fate decisions between neuroectoderm and mesendoderm in human pluripotent stem cells. Cell Stem Cell 2010;6(1):59–70. 201. Yin G, Chen R, Alvero AB, Fu HH, Holmberg J, Glackin C, et al. TWISTing stemness, inflammation and proliferation of epithelial ovarian cancer cells through MIR199A2/214. Oncogene 2010;29(24):3545–53. 202. Liu C, Tang DG. MicroRNA regulation of cancer stem cells. Cancer Res 2011;71(18):5950–4. 203. Pani G, Galeotti T, Chiarugi P. Metastasis: cancer cell’s escape from oxidative stress. Cancer Metastasis Rev 2010;29(2):351–78.