Hyaluronan: A modulator of the tumor microenvironment

Hyaluronan: A modulator of the tumor microenvironment

Accepted Manuscript Title: Hyaluronan: a modulator of the tumor microenvironment Author: Theerawut Chanmee, Pawared Ontong, Naoki Itano PII: DOI: Refe...

881KB Sizes 0 Downloads 117 Views

Accepted Manuscript Title: Hyaluronan: a modulator of the tumor microenvironment Author: Theerawut Chanmee, Pawared Ontong, Naoki Itano PII: DOI: Reference:

S0304-3835(16)30099-4 http://dx.doi.org/doi: 10.1016/j.canlet.2016.02.031 CAN 12772

To appear in:

Cancer Letters

Received date: Revised date: Accepted date:

17-10-2015 16-2-2016 17-2-2016

Please cite this article as: Theerawut Chanmee, Pawared Ontong, Naoki Itano, Hyaluronan: a modulator of the tumor microenvironment, Cancer Letters (2016), http://dx.doi.org/doi: 10.1016/j.canlet.2016.02.031. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Hyaluronan: a modulator of the tumor microenvironment Theerawut Chanmeea, Pawared Ontongb, Naoki Itanoa,b,c,* a

Department of Molecular Biosciences, Faculty of Life Sciences, Kyoto Sangyo University,

Kita-ku, Kyoto, Japan b

Division of Engineering (Biotechnology), Graduate School of Engineering, Kyoto Sangyo

University, Kita-ku, Kyoto, Japan c

Institute of Advanced Technology, Kyoto Sangyo University, Kita-ku, Kyoto, Japan

*Corresponding author. Tel.: +81-75-705-3064; Fax: +81-75-705-3064. E-mail address: [email protected] (N. Itano)

Highlights 1. The HA-rich tumor microenvironment recruits and activates stromal cells to enhance tumorigenicity. 2. Tumor-derived and stromal-derived HA molecules remodel tumor microenvironment to promote tumor progression. 3. HA regulates cancer stemness through the induction of EMT.

ABSTRACT Tumors are cellular masses formed through dynamic interactions between tumor cells and a mixed population of stromal cells. Crosstalk between oncogenic and adjacent stromal cells contributes to the formation of a “tumor microenvironment” influencing the tumor cell behaviors of proliferation, invasion, and metastatic spread throughout cancer progression. The composition and structure of the tumor microenvironment vary among different types of tumors and are extensively

remodeled

in

close

association

with

tumor

advancement.

The

tumor

microenvironment is composed of not only cellular compartments, such as endothelial cells, fibroblasts, inflammatory cells, and immune cells, but also of bioactive substances, including growth factors and the extracellular matrix. Hyaluronan (HA) is a major component of the 1

Page 1 of 29

extracellular matrix, and the degree of HA accumulation is strongly correlated with a poor prognosis in advanced cancer patients. Emerging evidence has suggested that HA creates a specific microenvironment that is favorable for tumor angiogenesis, invasion, and metastasis. This review highlights the prominent roles of HA as a modulator of the tumor microenvironment and addresses the recent advances regarding HA function in cancer stem cell niches.

Keywords: Hyaluronan; Tumor microenvironment; Epithelial-mesenchymal transition; Cancer stem cell; Stromal cells

Introduction The tumor microenvironment is widely recognized as a key player in the promotion of tumor growth and malignant progression. It is composed of heterogeneous cellular and non-cellular compartments and is extensively remodeled during tumor advancement [1,2]. The tumor microenvironment contains multiple types of stromal cells, including tumor-associated macrophages (TAMs), tumor-associated fibroblasts (TAFs), mesenchymal stem cells (MSCs), and endothelial cells. Interactions between tumor and stromal cells play a critical role in tumor initiation and progression. Over the past decade, considerable efforts have been made in exploring how these stromal cells facilitate tumor initiation and progression. In concert with malignant cells, stromal cells play prominent roles in the formation and modulation of the tumor microenvironment by producing a wide variety of soluble signaling molecules [3,4]. Moreover, the significance of extracellular matrix (ECM) components in tumor development has also been highlighted by pathological and experimental studies [5-7]. Aberrant ECM turnover and organization are often detected in tumor and appear to be involved in many biological aspects supporting tumor progression. There is growing evidence that certain ECM components have pro-angiogenic, pro-inflammatory, and anti-apoptotic effects. Among such molecules, tenascin-C [8], biglycan [9], and versican [10] have all been demonstrated to stimulate innate immune responses and thereby establish a pro-inflammatory microenvironment for tumor growth. Increased ECM rigidity from the accumulation of collagen 2

Page 2 of 29

crosslinking also promotes integrin clustering and enhances the cellular signals that regulate tumor cell survival, proliferation, and invasion [7]. Moreover, ECM degradation gives rise to the generation of bioactive fragments which act directly as inflammatory stimuli. For instance, proteolytic cleavage of type IV collagen generated a cryptic epitope required for angiogenesis and tumor growth [11]. As an integral component of the ECM, hyaluronan (HA) influences the fundamental aspects of cellular biology through both direct and indirect actions. HA mediates many cellular events during embryonic morphogenesis, cellular regeneration, and wound healing, while abnormalities in HA have been implicated in many diseases, such as inflammatory disorders and cancer [12]. In the tumor microenvironment, HA provides a three-dimensional scaffold for cells by the assembly of pericellular ECM and regulates the behaviors of stromal and tumor cells, in some cases through interactions with HA receptors. A wide variety of HA-binding molecules contributes to the assembly of pericellular ECM and specifically regulates a diverse array of HA functions. Although HA is a polysaccharide of a relatively simple composition, it exhibits multiple properties dependent largely on its molecular size and tissue concentration, both of which are firmly regulated by the concerted activities of biosynthetic and degradation processes [13]. In this review, we offer important insights into the roles of HA amid the crosstalk between malignant and tumor-associated stromal cells and describe how HA modulates the tumor microenvironment to promote tumor development and progression.

Abnormal HA synthesis and degradation in tumors

HA is a large polysaccharide composed of repeating N-acetylglucosamine and glucuronic acid disaccharide units (Fig. 1A). Under normal physiological conditions, HA can be composed of up to 25,000 disaccharide units, but these chains become smaller and more dispersed in a pathological status [13]. It is now accepted that the biophysical functions of HA vary depending on its molecular size. For instance, low molecular weight HA (LMW-HA) of a defined size (generally <200 kDa) is able to induce inflammation and angiogenesis [14], while high molecular weight HA (HMW-HA) is anti-angiogenic [15] as well as immunosuppressive [16]. 3

Page 3 of 29

The molecular size of HA is firmly regulated by the concerted activities of biosynthetic and degradation processes. In mammalian cells, each of three HA synthases (HAS1-3) possessing different enzymatic properties synthesize HA by repeated addition of each monosaccharide component to the nascent HA chain and extrude it through the plasma membrane to the cell surface or into the ECM [17] (Fig. 1B). HA catabolism is controlled by a family of endo-N-acetylhexosaminidases called hyaluronidases. Six different hyaluronidase (HYAL1, 2, 3, 4, HYALP1, and PH-20) genes have been identified in humans [18] whose expression is either tissue-specific or responsive to microenvironmental stimuli. HYAL1 and HYAL2 are the major hyaluronidases expressed in most tissues and hydrolyze the β (1-4) glycosidic linkage between N-acetylglucosamine and glucuronic acid to produce HA fragments of varying sizes. Mechanistically, HMW-HA is tethered to the cell surface by CD44 in caveolin-rich lipid rafts and cleaved by glycosylphosphatidylinositol (GPI)-anchored HYAL2 [19]. HYAL2 generates intermediate sized HA fragments that are eventually delivered to endo-lysosome compartments, wherein HYAL1 and lysosomal β-exoglycosidase enzymes further degrade the fragments into oligosaccharides [20] (Fig. 1C). Recently, KIAA1199 has been shown to play a central role in HA degradation in a manner independent of CD44 and HYAL [21]. This molecule was originally described as an inner ear-specific protein involved in non-syndromic hearing loss [22]. Cell surface KIAA1199 can bind to HA and internalize the sugar chain via clathrin-coated pits. Blockage of either the internalization or knockdown of KIAA1199 suppressed the fragmentation of internalized HA [21], suggesting that the HA catabolism mediated by this molecule occurred through vesicle endocytosis (Fig. 1C). Accumulation of HA and its function in tumor are rather complex. Within tumor, HA is abundantly synthesized by both cancer and stromal cells and the high level of HA has been reported to be closely associated with tumor aggressiveness. Clinicopathological analyses have demonstrated that accumulation of HA in malignant breast [23], prostate [24,25], ovarian [26], and lung [27] cancers are strongly associated with tumor aggressiveness and a poor disease outcome. Although widespread evidence supports the central role of HA in many aspects of tumor progression, contradictory observations have also been made in both clinical and experimental studies. Differing HA levels were found in oral squamous cell carcinoma patients, 4

Page 4 of 29

in whom reduced HA was associated with poor survival [28]. Likewise, decreased levels of HA and CD44 were linked to an unfavorable prognosis in clinical stage I cutaneous melanoma [29], and an extremely high level of HA inhibited tumor growth [30]. These observations therefore suggest that HA may regulate tumor aggressiveness positively as well as negatively in a contextdependent manner. The dynamic balance between concurrent HA synthesis and catabolism may be a prominent factor in tumor advancement.

HAS expression in both stromal and malignant cells has been shown to relate to tumor aggressiveness and low cancer patient survival rates [31]. Dysregulation of HAS1 alternative splicing that generated truncated variants, such as HAS1V, was correlated with significantly reduced survival in multiple myeloma patients [32]. The prominent role of HA in tumor progression has been supported by experimental studies, wherein increased HA production by forced expression of HAS1, HAS2, or HAS3 increased tumor growth and metastasis in xenograft models of breast cancer [33], prostate cancer [34], and fibrosarcoma [35]. Has2 overexpression experiments in a mouse model of breast cancer have provided further evidence on the importance of HA in tumor progression. HA overproduction induced marked stromal induction and accumulation of HA within tumor stroma. Concurrent with these events, blood and lymphatic vessels were formed in nearby stroma surrounding tumor cell islets [6,36], and the mechanism of HA-mediated stromal cell recruitment was found to rely on the presence of versican [6]. Correspondingly, a reduction in HAS2 inhibited the tumorigenesis and progression of breast cancer [37], and impaired tumor growth was observed when interactions between HA and tumor cells were disrupted [38]. The accumulation of LMW-HA has also been associated with tumor aggressiveness [39-41] in that HA oligosaccharides possess the ability to trigger the expression of specific cytokines and proteases necessary for ECM remodeling. For instance, LMW-HA of 4-6 oligomers in size enhanced MMP2 and IL-8 expression in human melanoma cells via toll-like receptor (TLR) 4 and subsequent activation of NF-κB [42]. Interactions of LMW-HA with CD44 and TLR signaling through MyD88/NF-κB led to the production of IL-1β/IL-8 and increased invasiveness in breast cancer cells [43]. Furthermore, LMW-HA-induced innate immune responses may enhance tumor growth and metastasis [44]. Consistent with the accumulation of LMW-HA, 5

Page 5 of 29

hyaluronidase levels are often increased in several types of cancers, such as those of colon [45], bladder [46], prostate [47], brain [48], and breast [49], and its induction has been linked to tumor grade and metastatic spread. KIAA1199 has also been highlighted in tumor progression and invasiveness due to its high expression in several aggressive cancers, such as those of the breast [50], prostate [51], and colon [52]. Conversely, decreased expression of hyaluronidases has also been reported in certain tumors such as head and neck squamous cell carcinoma [53], endometrial [54], and pancreatic [55] cancer. For instance, decreased expression of HYAL1 was correlated with poor survival in pancreatic cancer patients [55], while hyaluronidase treatment reduced the growth of tumor xenograft [56]. In light of this, abnormal synthesis and degradation of HA in advanced tumors may facilitate tumor cell aggressiveness by both accumulation of HA and generation of HA oligosaccharides.

HA-dependent changes in stromal cell behavior and modulation of the tumor microenvironment Tumor-associated macrophages TAMs are important stromal cells in tumors that play crucial roles in driving growth and progression. The well documented pro-tumor activities of TAMs include stimulation of tumor cell proliferation and migration, promotion of angiogenesis, immunosuppression, and microenvironment remodeling [57,58]. Clinicopathological studies have also highlighted a link between an abundance of TAMs and tumor aggressiveness [59,60]. In contrast, several studies have implicated that the presence of macrophages in tumors improved patient prognosis. Prominent macrophage infiltration tended to positively influence prognosis in colon cancer patients [61]. Moreover, the accumulation of macrophages in prostate [62] and colorectal cancers [63] was associated with higher survival. Pathological evidence has demonstrated that these macrophages, each with a distinct phenotype, exhibit significant heterogeneity in their localization pattern, implying that the differentiation of infiltrating monocytes is influenced by the local microenvironments within tumor masses. For example, macrophages in the peritumoral stroma markedly expressed major histocompatibility complex (MHC) class II, which suggested that they were newly infiltrating anti-tumor macrophages. In contrast, macrophages in cancer 6

Page 6 of 29

nests were negative for MHC class II and expressed anti-inflammatory cytokine IL-10, indicating that they represented pro-tumor phenotype [64]. The mechanisms underlying monocyte recruitment and differentiation in tumor masses have been investigated. Once circulating monocytes guided by tumor-derived chemoattractants lodge within tumors, they begin differentiation in response to local microenvironmental stimuli. Several cytokines, chemokines, and growth factors, including granulocyte-macrophage colonystimulating factor, macrophage colony-stimulating factor, chemokines CCL2, CXCL10, and CXCL12, and transforming growth factor-β (TGF-β), collectively orchestrate the multiple phases of monocyte recruitment, monocyte-to-macrophage differentiation, and macrophage activation [65]. There is also growing evidence that ECM molecules can activate macrophages as well (Fig. 2A). Among such molecules, HA and its binding proteoglycans have been demonstrated to initiate TLR-mediated innate immune responses. Versican, a large chondroitin sulfate proteoglycan, is involved in ECM assembly via associations with HA and various other ECM constituents. Kim et al. recently uncovered a new inflammatory pathway in which versican activated macrophages by ligation of TLR2 and elicited the production of pro-inflammatory cytokines [66]. As a major ECM component, HA has frequently been implicated in monocyte/macrophage trafficking and activation. Pathological studies of human breast cancer specimens suggested that increased numbers of macrophages were correlated with HA accumulation in tumors [67]. We have also demonstrated that TAMs preferentially infiltrate into mammary tumors in a manner dependent

on

an

HA-rich

tumor

microenvironment

and

concomitantly

enhance

neovascularization and tumor growth [36]. Mechanistically, these phenomena may be due to the concerted action of HA and its binding molecules. Pericellular HA cable-like structures, composed of inter-α-inhibitor heavy chains, TNF-stimulated gene-6 (TSG-6), and versican [68], provide a suitable scaffold for the mobilization of mononuclear leukocytes [69] (Fig. 2A). Upon binding to the cable-like framework via cell surface CD44, peripheral blood monocytes are activated to produce growth factors and ECM components. Meanwhile, HA degradation products also activate monocytes/macrophages to induce the specific gene expression programs required for inflammation and ECM remodeling. For instance, HA oligosaccharides utilized both TLR2 7

Page 7 of 29

and TLR4 as receptors to stimulate inflammatory gene expression in macrophages and act as an endogenous danger signal [70,71]. Macrophages polarize into diverse subsets, schematically identified as M1 (i.e., classically activated) and M2 (i.e., alternatively activated). M1 macrophages function in host defense mechanisms against bacteria and viruses and anti-tumor immunity, while M2 macrophages are widely termed as anti-inflammatory, immunoregulatory, and pro-tumor cells [72]. The polarization among distinct macrophage states is regulated in response to a wide variety of signals in the local microenvironment. In this regard, HA oligosaccharides have been shown to induce early activation of monocytes and subsequent development of immunosuppressive macrophages [64]. This view is consistent with the finding that, through CD44 and TLR-4, HA oligosaccharides downregulated the expression of inflammatory cytokines TNF-α and IL-12 and upregulated the expression of Th2 cytokines IL-10 [73] (Fig. 2A). Earlier studies have reported that HA oligosaccharides act on monocytes and macrophages to drive them towards an M2 subtype [64]. The differentiated M2 macrophages then start to produce Th2 cytokines and several angiogenic factors, leading to the enhancement of tumorigenicity through their immunosuppressive and pro-angiogenic actions.

Tumor-associated fibroblasts

HA facilitates fibroblast recruitment by modulating the tumor microenvironment Fibroblasts residing in the tumor margin or infiltrating into the tumor mass, termed TAFs, are the major cellular stromal components in many solid tumors [74-76]. TAFs are phenotypically distinct from normal fibroblasts and display the myofibroblast-like characteristics of rapid proliferation rate and expression of mesenchyme-specific proteins, such as α-smooth muscle actin (α-SMA), fibroblast-specific protein-1 (S100A4), fibroblast-activating protein, and vimentin [74,77]. There is conflicting evidence on the role of TAFs in tumor formation and growth: TAFs exhibit pro-tumor activity in certain types of cancers while act as tumor suppressors in others. TAFs contribute to tumor promotion by actively secreting a wide spectrum of growth factors, chemokines, and cytokines [78]. TAF-secreted hepatocyte growth factor (HGF) directly enhanced the proliferation and survival of cancer cells [79]. Paracrine signaling 8

Page 8 of 29

of TAF-derived factors, such as CCL2 and vascular endothelial growth factor (VEGF), has also been proposed to contribute to tumor formation and progression through the recruitment of macrophages and endothelial cell progenitors [80-82]. Furthermore, TAFs produce both ECM components and its degrading enzymes, thereby involving themselves in the remodeling of the ECM and generation of a tumor microenvironment favorable for tumor cell proliferation and invasion. On the other hand, Chang et al. demonstrated that TAFs expressing Slit2 could either suppress or promote the tumorigenicity of breast cancer cells [83]. These observations suggest that TAFs may regulate tumor growth positively as well as negatively in a context-dependent manner. Tumors can recruit fibroblasts and reprogram them into activated TAFs. Several lines of evidence have shown that tumor cells frequently drive fibroblast recruitment by tumor-derived growth factors, such as TGF-β and platelet-derived growth factor [84,85]. Dynamic changes in the ECM have been reported to promote fibroblast recruitment as well. Our earlier study using a transgenic mouse model demonstrated that HA overproduction in spontaneous mammary tumors led to an accumulation of TAFs in tumor stroma [6]. HA evokes fibroblast motility by acting on intracellular signaling pathways through interactions with its cell surface receptors. Specifically, the molecule may promote an invasive phenotype of myofibroblast via binding with CD44 since a CD44 deficiency attenuated the HA-dependent invasiveness of myofibroblasts [86]. This observation was consistent with the fact that 6-mer HA oligosaccharides were essential for ligand recognition by CD44 to stimulate fibroblast migration [87]. Such an effect was abrogated in mice deficient for receptor for HA-mediated motility (RHAMM) and CD44, suggesting that these receptors were required for a fibroblast response to HA oligosaccharides. It is also noteworthy that HA oligosaccharide-stimulated fibroblast migration and invasion required RHAMM to sustain CD44-mediated ERK1/2 activity [88]. Alternatively, HA accumulation in tumors may provide a microenvironment amenable to easy fibroblast penetration by increasing the turgidity and hydration of the ECM. The construction of an HA-rich ECM followed by the recruitment of fibroblasts might simultaneously drive tumor microenvironment remodeling through the deposition of other ECM components. For instance, TAF-derived collagen may generate a crosslinked network that contributes directly to ECM stiffness, thereby increasing cell adhesion and invasion [7]. 9

Page 9 of 29

Involvement of HA in TAF precursors Regarding the origin of TAFs, several possible sources have been proposed: (1) local fibroblasts or tissue-resident fibroblast precursors infiltrating into the growing tumor, (2) MSCs recruited from the circulation, (3) tumor cells undergoing epithelial-mesenchymal transition (EMT) in the tumor parenchyma, and (4) endothelial cells that undergo endothelial to mesenchymal transition (EndMT) (Fig. 2B). The most direct route is tissue-resident fibroblasts that transdifferentiate toward myofibroblasts by tumor-derived factors during their recruitment. Among the factors, TGF-β induces fibroblast differentiation through two cooperative signaling cascades: HA/CD44 and the epidermal growth factor receptor (EGFR)/ERK1/2 signaling. Midgley et al. illustrated how HA promoted TGF-β-driven fibroblast differentiation, describing that the assembly of HA pericellular coats promoted co-localization of CD44 and EGFR within lipid rafts and subsequent activation of the MAPK/ERK signaling pathway. This pathway is synergistic with that of TGF-β/Smad2, which is necessary for fibroblast differentiation [89]. Studies employing mouse models of inflammation-induced tumors have supported the premise that MSCs are a source of TAFs. Quante et al. reported that 20% of TAFs were derived from MSCs in a murine model of inflammation-induced gastric cancer [90]. The processes controlling MSC mobilization have been linked to HA-CD44 interactions, whereby HA cablelike structures might provide a scaffold for CD44-positive MSCs and facilitate their invasion into tumors. Indeed, CD44-deficient MSCs exhibited reduced infiltration into tumor stroma and a hampered ability to differentiate into myofibroblasts in a transgenic murine cancer model [91]. Lastly, myofibroblasts may be derived from cancer cells that undergo EMT. As HA overproduction induces EMT in normal epithelial cells [92,93], HA-dependent EMT might similarly convert tumor cells to TAFs and thereby function as a mechanism for TAF supply.

Endothelial cells and angiogenesis

Angiogenesis is an essential process in tumor growth and metastasis that requires specific sequential cellular events, such as endothelial cell activation and degradation of the basement membrane followed by endothelial cell migration and invasion into the stroma [94]. In addition 10

Page 10 of 29

to malignant cells, inflammatory cells, stromal fibroblasts, perivascular cells, and other tumorassociated stromal cells contribute significantly to the production of angiogenic factors. These factors trigger an angiogenic cascade accompanied by the dissociation of pericytes from blood vessels, degradation of the basement membrane, and proliferation and migration of endothelial cells [95]. Among such angiogenic factors, VEGF and basic fibroblast growth factor (bFGF) have emerged as central regulators [94]. The ECM surrounding the vasculature can also affect angiogenesis, either positively or negatively [77]. The ECM modulates tubular network formation and thereby influences angiogenesis through multiple mechanisms involved in regulating endothelial cell behaviors. The ECM surrounding normal vasculature is mainly composed of fibronectin, laminin, collagen, and proteoglycans [96]. On the other hand, in vivo studies have shown that ECM-derived HA oligosaccharides of 16 disaccharide units have the potential to induce pathological angiogenesis [6]. LMW-HA has long been recognized as a pro-angiogenic factor, and HA oligosaccharides along with VEGF synergistically stimulated endothelial cell proliferation, migration, and capillary formation [97]. Mechanistically, HA fragments can trigger the MAPK cascade via interactions with CD44 [98]. Furthermore, Lennon et al. found that LMW-HA induced the formation of CD44v10/EphA2 complexes and Src-mediated EphA2 phosphorylation, both of which are required for angiogenesis [99]. Microvascular endothelial cells express high levels of CD44 and HYAL2. Knockdown of either abrogated the formation of tubular networks, suggesting that angiogenic modulation was mediated by the binding of HA degradation products to CD44 [100]. RHAMM is also a receptor for HA that is expressed both on the cell surface and within intracellular compartments. Several studies have demonstrated RHAMM expression in endothelial cells and its role in angiogenesis modulation [101,102]. Since knockdown of either receptor was sufficient to inhibit LMW-HA-induced vessel formation [102], the HA receptors CD44 and RHAMM appear to be critical for new blood vessel formation; CD44 is the major receptor for endothelial cell adhesion and proliferation, while RHAMM is essential for endothelial cell invasion [101]. It is believed that CD44 and RHAMM activate distinct cascades in response to LMW-HA since CD44 mediated signaling through PKC-α and γ-adducin whereas RHAMM did not [102]. Meanwhile, HMW native HA has been considered to be anti-angiogenic because it inhibited endothelial cell proliferation and migration as well as capillary formation in a three-dimensional 11

Page 11 of 29

matrix in vitro [15], although recent in vivo studies suggested that HMW-HA might be angiogenic. Koyama et al. analyzed the molecular basis of HMW-HA-induced angiogenic promotion in vivo and witnessed a cooperative action between HA and versican in the mobilization of stromal cells and subsequent enhancement of bFGF-induced angiogenesis [6]. Another report described how HMW-HA augmented CXCL12/CXCR4 signaling associated with CD44 to facilitate vessel formation and angiogenesis [103]. Thus, the pro-angiogenic function of HMW-HA is likely due to the enhancement of signals induced by chemokines and growth factors. Overall, the balance of regulatory HMW-HA and effector HA oligosaccharides may figure prominently in promoting angiogenic responses (Fig. 2C).

Interstitial fluid pressure and vasculature

The interstitial fluid is a medium for the transport of nutrients and waste products between cells and the vascular system, and transcapillary flow into interstitial spaces is controlled by hydrostatic and colloid osmotic interstitial fluid pressure (IFP). IFP is very low in normal tissue, but can become markedly elevated in solid tumors [104]. Altered IFP has been disclosed in several types of tumors, such as breast [105], melanoma [106], colon [105], head and neck [107], and pancreatic ductal adenocarcinoma (PDA) [108]. A number of studies have suggested that high IFP can limit the efficacy of cancer treatment. The chemotherapeutic drugs and monoclonal antibodies used in cancer treatment are normally transported to interstitial spaces via pressure gradients. Increased IFP hampers the transport of these macromolecules, thus rendering therapy less effective. For instance, IFP often ranges from 75 mmHg to 130 mmHg in PDA, in stark contrast to 8 mmHg in the normal pancreas and -2.0 mmHg in muscle [109]. Along with its unique biophysical properties of strong water retention, HA can form a hydrogel-like ECM that exerts strong turgor force, and is thus likely involved in the mechanism for increased tumor IFP. In fact, HA level was correlated with IFP elevation in several reports, and systemic administration of HA degradation enzymes periodically lowered IFP and raised tumor cell susceptibility to cytotoxic chemotherapeutic agents [109-111]. Therefore, targeted disruption of HA polymers may alter a hydrated ECM and ameliorate the transport of chemotherapeutic agents.

12

Page 12 of 29

Roles of HA in EMT EMT is a cellular program that allows a polarized epithelial cell to acquire mesenchymal features. This process has been accepted as an essential developmental phenomenon that controls many embryonic events, such as gastrulation and neural crest formation [112]. Cells undergoing EMT are characterized by profound morphological changes from a cobblestone-like epithelial form to a spindle shape. During the EMT process, epithelial cells lose their cell-cell adhesion properties and apical-basal polarity and acquire a fibroblastic motile phenotype (Fig. 3A). Hallmarks of EMT are the downregulation of E-cadherin and upregulation of vimentin, both of which are tightly controlled by multiple signaling pathways. TGF-β is one of the most prominent EMT inductors, promoting the conversion of epithelial to mesenchymal features by transcriptional and post-transcriptional regulation of a distinct set of transcription factors [113]. A variety of such factors, including the zinc finger Snail homologues (Snail1 and Snail2/Slug) and several basic helix-loop-helix factors (Twist, ZEB-1, and ZEB2), have been demonstrated to promote EMT through the coordinated modulation of EMT-related genes [114]. An increasing number of studies are implicating EMT in tumor progression and metastasis. During tumor advancement, EMT produces many morphological and molecular alterations in tumor cells that are similar to those in the developmental EMT program. Tumor cells undergoing EMT have increased invasiveness via an enhancement in cell motility, reductions in cell-cell adhesion and cell polarity, and matrix remodeling [115]. Multiple extracellular stimuli, including HGF, EGF, platelet-derived growth factor (PDGF), Wnt, Notch, and TGF-β combine to orchestrate the EMT-related process by integrated networks of signal transduction pathways and transcription factors [114]. HA has also been associated with the acceleration of EMT. Elevated HA production in MCF-10A human mammary epithelial cells induced mesenchymal characteristics, including upregulation of vimentin, dispersion of cytokeratin, and loss of cell adhesion at intercellular boundaries [92]. Increased levels of HA further promoted anchorage-independent growth and invasiveness by stimulating the PI3K/Akt signaling pathway [92] and inducing MMP-2 and MMP-9 expression, respectively [116] (Fig. 3B). Perturbation of HA-receptor interactions by treatment with HA oligosaccharides reversed the invasiveness induced by HGF in MCF-10A 13

Page 13 of 29

cells, suggesting the involvement of HA receptors in EMT [92]. Indeed, CD44 interacts with ERM proteins (ezrin/radixin/moesin) in response to HA, which directly mediate cytoskeletal remodeling and promote migration and invasion [117,118] (Fig. 3B). Furthermore, HA-CD44 communication evoked nuclear translocation of the CD44 intracellular domain followed by expression of the EMT-related Twist transcription factor in human breast cancer [119]. Porsch et al. demonstrated that HAS2 knockdown inhibited both HA production and TGF-β-induced EMT and completely abolished TGF-β-accelerated cell migration in NMuMG mammary epithelial cells. However, inhibition of HA-CD44 binding by neutralizing antibodies did not abrogate TGF-β-induced EMT, suggesting a CD44-independent action of HA [120]. Our recent study with a murine mammary tumor model also showed in vivo that forced Has2 expression and resultant HA overproduction in tumor cells caused the loss of E-cadherin at cell-cell junctions and increased nuclear translocation of β-catenin [6]. Several studies have suggested that βcatenin contributes to the EMT program by inducing the expression of Slug [121] and Twist [122]. Although the significance of HA-CD44 interactions in EMT is controversial, these reports highlight the importance of HA in the dynamic regulation of the EMT program. Along with EMT, its reverse process, mesenchymal-epithelial transition (MET) may also play a critical role in a series of metastatic steps. Epithelial-mesenchymal plasticity, or the interplay between EMT and MET, is tightly regulated by environmental conditions and signals. HA has been implicated in the metastatic spread of various cancer types. Experimentally, we demonstrated that mutant mammary carcinoma cells lacking the ability to synthesize HA displayed a significant reduction in experimental lung metastasis, while restoration of HA synthesis by HAS1 transfection recovered the metastatic properties in accordance with the formation of HA pericellular coats [33]. An inhibitor of HA synthesis, 4-methylumbelliferone (4MU), inhibited liver metastasis of B16F-10 melanoma cells [123] and lung metastasis of osteosarcoma cells [124]. CD44 is frequently overexpressed in metastatic cancer cells and has been linked to the metastatic spread of numerous cancer types [125-127]. CD44 induction in the weakly metastatic MCF7 breast cancer cell line enhanced metastasis to the liver when injected into immunodeficient mice [128]. Meanwhile, absence of the CD44 gene prevented lung metastasis of osteosarcoma in mice with a tm1 mutation in the p53 gene [129]. Although it

14

Page 14 of 29

remains to be confirmed, the switch between EMT and MET may be instrumental in the HA/CD44-dependent acceleration of metastatic spread.

HA and cancer stem cell niches Stem cells reside in a special microenvironment called a “stem cell niche” that provides the major cues for promoting survival and maintenance. Stem cell behavior is influenced by direct interactions not only with neighboring stromal cells, but also with niche components, such as secreted factors, ECM molecules, hypoxia, and other environmental signals. A recent study has indicated that HA is a niche element required for bone marrow hematopoiesis. Human bone marrow mesenchymal stem cells typically synthesize a large amount of HA to retain their pericellular matrix and maintain stemness. HA depletion reduced the ability of the microenvironment to support these hematopoietic stem cells [130]. Similarly, inhibition of HA synthesis by 4-MU treatment eliminated hematopoiesis in long-term bone marrow cultures [131]. Cancer stem cells (CSCs) represent a small subpopulation of self-renewing oncogenic cells that drive tumor initiation and progression [132,133]. Comparably to a normal stem cell niche, a CSC niche regulates the maintenance and expansion of CSCs. Recent investigations have uncovered certain extra- and intracellular signals that can revert cancer progenitors to selfrenewing and multipotent CSC states [134-137]. Thus, remodeling of the CSC niche is now thought to be a crucial step in tumorigenesis and tumor progression. An HA-rich ECM may provide a microenvironment favorable for the self-renewal and maintenance of CSCs. Okuda et al. reported that HA produced by metastatic breast cancers promoted interactions between CSCs and TAMs, which then secreted PDGF to activate fibroblasts and osteoblasts and support CSC self-renewal [138]. In human head and neck squamous cell cancer, binding of HA and CD44 variant form 3 propagated CSC stemness by inducing complex formation of the stem cellspecific transcription factors Oct4, Sox2, and Nanog [139]. Furthermore, HA/CD44-mediated Nanog activation promoted the expression of the stem cell regulators Rex1 and Sox2 [140] (Fig. 3C). CD44 knockdown could attenuate the expression of Oct4, Nanog, Sox2, and other stem cell markers [141]. Since stem cell-specific transcription factors are necessary for the development 15

Page 15 of 29

and maintenance of CSCs, these lines of evidence emphasize that both HA and CD44 are indispensable factors in CSC self-renewal. EMT is believed to mediate the reversion of cancer cells to CSCs. In support of this, induction of EMT in breast cancer cells by ectopic expression of Snail and Twist or prolonged TGF-β treatment stimulated the acquisition of stem cell properties [142]. Forced expression of Slug, a member of the Snail family, in collaboration with Sox9 efficiently induced the entrance of breast cancer cells into a CSC state [143]. Evidence supporting a key role of HA in EMT induction has also implicated an association of HA with the acquisition of CSC signatures. In our recent study, HA-overproducing mammary tumor cells acquired stemness via the upregulation of TGF-β and induction of Snail and Twist, while a loss of EMT by inhibition of TGF-β-Snail signaling or Twist knockdown markedly reduced CSC subpopulations [93]. CSCs are inherently resistant to cytotoxic chemo- and radiotherapy, thereby abrogating complete therapeutic elimination of tumor masses. The acquisition of multidrug resistance (MDR) in CSCs is mediated by the induction of survival/anti-apoptotic signals and increased expression of multidrug transporters and MDR genes. HA-CD44 binding elicited MDR by promoting expression of MDR1 and multidrug resistance protein 2 (Fig. 3C). These interactions also promoted Nanog phosphorylation and its nuclear translocation, which eventually resulted in the upregulation of MDR1 [144]. CSCs effectively gain a survival/growth advantage under oxidative stress conditions by adapting their metabolism and maintaining cellular redox homeostasis. Metabolic modulation by CD44 has been demonstrated to contribute to antioxidant status in tumor cells; CD44 caused metabolic reprograming from mitochondrial respiration to glycolysis in p53-deficient or hypoxic cancer cells by interacting with pyruvate kinase M2 [145]. CD44 variant form (CD44v8-10) also modulated defense against reactive oxygen species by altering cellular glutathione synthesis [146]. HMW-HA has been shown to decrease oxidative DNA damage in human corneal epithelial cells [147], and HA exerted protective effects against mitochondrial DNA damage, mitochondrial dysfunction, and mitochondria-driven apoptosis under conditions of oxidative injury [148].

Conclusions and future prospects 16

Page 16 of 29

This review emphasizes prominent roles of HA as a potent modulator of the tumor microenvironment. HA creates specific circumstances that are favorable for interactions between tumor cells and stromal cells to elicit a wide range of extracellular stimuli for tumor growth, angiogenesis, invasion, and metastasis. Recent evidence further suggests that HA serves as a niche to regulate CSC maintenance, propagation, and chemotherapeutic resistance. Thus, HA appears to have a significant impact on many aspects of tumor initiation and malignancy. A more complete understanding of the mechanisms underlying HA-driven modulation of the tumor microenvironment will facilitate the development of future anticancer treatments. Additionally, targeting of HA in the CSC niche in combination with conventional chemotherapy may provide a promising therapeutic approach for achieving successful cancer eradication.

Acknowledgments This work was funded by grants from JSPS KAKENHI Grant Number 26430125 (to N.I.); Kyoto Sangyo University Research Grant Number C1301 (to N.I.) and Tokyo Biochemical Research Foundation postdoctoral fellowships for Asian researchers in Japan (to T.C.).

Conflict of interest The authors have no conflicts of interest to disclose.

17

Page 17 of 29

References [1] [2] [3] [4] [5]

[6]

[7]

[8] [9]

[10] [11]

[12] [13]

[14]

[15] [16]

D.F. Quail, J.A. Joyce, Microenvironmental regulation of tumor progression and metastasis, Nat Med 19 (2013) 1423-1437. D. Hanahan, L.M. Coussens, Accessories to the crime: functions of cells recruited to the tumor microenvironment, Cancer Cell 21 (2012) 309-322. T.L. Whiteside, The tumor microenvironment and its role in promoting tumor growth, Oncogene 27 (2008) 5904-5912. S.S. McAllister, R.A. Weinberg, The tumour-induced systemic environment as a critical regulator of cancer progression and metastasis, Nat Cell Biol 16 (2014) 717-727. A. Gorter, H.J. Zijlmans, H. van Gent, J.B. Trimbos, G.J. Fleuren, E.S. Jordanova, Versican expression is associated with tumor-infiltrating CD8-positive T cells and infiltration depth in cervical cancer, Mod Pathol 23 (2010) 1605-1615. H. Koyama, T. Hibi, Z. Isogai, M. Yoneda, M. Fujimori, J. Amano, et al., Hyperproduction of hyaluronan in neu-induced mammary tumor accelerates angiogenesis through stromal cell recruitment: possible involvement of versican/PG-M, Am J Pathol 170 (2007) 1086-1099. K.R. Levental, H. Yu, L. Kass, J.N. Lakins, M. Egeblad, J.T. Erler, et al., Matrix crosslinking forces tumor progression by enhancing integrin signaling, Cell 139 (2009) 891-906. R. Chiquet-Ehrismann, M. Chiquet, Tenascins: regulation and putative functions during pathological stress, J Pathol 200 (2003) 488-499. M. Chieppa, G. Bianchi, A. Doni, A. Del Prete, M. Sironi, G. Laskarin, et al., Crosslinking of the mannose receptor on monocyte-derived dendritic cells activates an antiinflammatory immunosuppressive program, J Immunol 171 (2003) 4552-4560. T.N. Wight, Versican: a versatile extracellular matrix proteoglycan in cell biology, Curr Opin Cell Biol 14 (2002) 617-623. J.J. Ames, L. Contois, J.M. Caron, E. Tweedie, X. Yang, R. Friesel, et al., Identification of an endogenously generated cryptic collagen epitope (XL313) that may selectively regulate angiogenesis by an integrin-YAP mechano-transduction pathway, J Biol Chem (2015) B.P. Toole, Hyaluronan: from extracellular glue to pericellular cue, Nat Rev Cancer 4 (2004) 528-539. P. Heldin, K. Basu, B. Olofsson, H. Porsch, I. Kozlova, K. Kahata, Deregulation of hyaluronan synthesis, degradation and binding promotes breast cancer, J Biochem 154 (2013) 395-408. M. Slevin, S. Kumar, J. Gaffney, Angiogenic oligosaccharides of hyaluronan induce multiple signaling pathways affecting vascular endothelial cell mitogenic and wound healing responses, J Biol Chem 277 (2002) 41046-41059. D.C. West, S. Kumar, The effect of hyaluronate and its oligosaccharides on endothelial cell proliferation and monolayer integrity, Exp Cell Res 183 (1989) 179-196. P.L. Bollyky, J.D. Lord, S.A. Masewicz, S.P. Evanko, J.H. Buckner, T.N. Wight, et al., Cutting edge: high molecular weight hyaluronan promotes the suppressive effects of CD4+CD25+ regulatory T cells, J Immunol 179 (2007) 744-747. 18

Page 18 of 29

[17] [18]

[19]

[20] [21]

[22]

[23]

[24]

[25]

[26]

[27]

[28]

[29]

[30]

P.H. Weigel, V.C. Hascall, M. Tammi, Hyaluronan synthases, J Biol Chem 272 (1997) 13997-14000. A.B. Csoka, S.W. Scherer, R. Stern, Expression analysis of six paralogous human hyaluronidase genes clustered on chromosomes 3p21 and 7q31, Genomics 60 (1999) 356-361. L.Y. Bourguignon, P.A. Singleton, F. Diedrich, R. Stern, E. Gilad, CD44 interaction with Na+-H+ exchanger (NHE1) creates acidic microenvironments leading to hyaluronidase-2 and cathepsin B activation and breast tumor cell invasion, J Biol Chem 279 (2004) 26991-27007. A.B. Csoka, G.I. Frost, R. Stern, The six hyaluronidase-like genes in the human and mouse genomes, Matrix Biol 20 (2001) 499-508. H. Yoshida, A. Nagaoka, A. Kusaka-Kikushima, M. Tobiishi, K. Kawabata, T. Sayo, et al., KIAA1199, a deafness gene of unknown function, is a new hyaluronan binding protein involved in hyaluronan depolymerization, Proc Natl Acad Sci U S A 110 (2013) 5612-5617. S. Abe, S. Usami, Y. Nakamura, Mutations in the gene encoding KIAA1199 protein, an inner-ear protein expressed in Deiters' cells and the fibrocytes, as the cause of nonsyndromic hearing loss, J Hum Genet 48 (2003) 564-570. P. Auvinen, R. Tammi, J. Parkkinen, M. Tammi, U. Agren, R. Johansson, et al., Hyaluronan in peritumoral stroma and malignant cells associates with breast cancer spreading and predicts survival, Am J Pathol 156 (2000) 529-536. A. Josefsson, H. Adamo, P. Hammarsten, T. Granfors, P. Stattin, L. Egevad, et al., Prostate cancer increases hyaluronan in surrounding nonmalignant stroma, and this response is associated with tumor growth and an unfavorable outcome, Am J Pathol 179 (2011) 1961-1968. P. Lipponen, S. Aaltomaa, R. Tammi, M. Tammi, U. Agren, V.M. Kosma, High stromal hyaluronan level is associated with poor differentiation and metastasis in prostate cancer, Eur J Cancer 37 (2001) 849-856. M.A. Anttila, R.H. Tammi, M.I. Tammi, K.J. Syrjanen, S.V. Saarikoski, V.M. Kosma, High levels of stromal hyaluronan predict poor disease outcome in epithelial ovarian cancer, Cancer Res 60 (2000) 150-155. R. Pirinen, R. Tammi, M. Tammi, P. Hirvikoski, J.J. Parkkinen, R. Johansson, et al., Prognostic value of hyaluronan expression in non-small-cell lung cancer: Increased stromal expression indicates unfavorable outcome in patients with adenocarcinoma, Int J Cancer 95 (2001) 12-17. A. Kosunen, K. Ropponen, J. Kellokoski, M. Pukkila, J. Virtaniemi, H. Valtonen, et al., Reduced expression of hyaluronan is a strong indicator of poor survival in oral squamous cell carcinoma, Oral Oncol 40 (2004) 257-263. J.M. Karjalainen, R.H. Tammi, M.I. Tammi, M.J. Eskelinen, U.M. Agren, J.J. Parkkinen, et al., Reduced level of CD44 and hyaluronan associated with unfavorable prognosis in clinical stage I cutaneous melanoma, Am J Pathol 157 (2000) 957-965. N. Itano, T. Sawai, F. Atsumi, O. Miyaishi, S. Taniguchi, R. Kannagi, et al., Selective expression and functional characteristics of three mammalian hyaluronan synthases in oncogenic malignant transformation, J Biol Chem 279 (2004) 18679-18687. 19

Page 19 of 29

[31]

[32]

[33]

[34]

[35]

[36]

[37]

[38] [39]

[40] [41]

[42]

[43]

[44] [45]

P. Auvinen, K. Rilla, R. Tumelius, M. Tammi, R. Sironen, Y. Soini, et al., Hyaluronan synthases (HAS1-3) in stromal and malignant cells correlate with breast cancer grade and predict patient survival, Breast Cancer Res Treat 143 (2014) 277-286. S. Adamia, T. Reiman, M. Crainie, M.J. Mant, A.R. Belch, L.M. Pilarski, Intronic splicing of hyaluronan synthase 1 (HAS1): a biologically relevant indicator of poor outcome in multiple myeloma, Blood 105 (2005) 4836-4844. N. Itano, T. Sawai, O. Miyaishi, K. Kimata, Relationship between hyaluronan production and metastatic potential of mouse mammary carcinoma cells, Cancer Res 59 (1999) 2499-2504. N. Liu, F. Gao, Z. Han, X. Xu, C.B. Underhill, L. Zhang, Hyaluronan synthase 3 overexpression promotes the growth of TSU prostate cancer cells, Cancer Res 61 (2001) 5207-5214. R. Kosaki, K. Watanabe, Y. Yamaguchi, Overproduction of hyaluronan by expression of the hyaluronan synthase Has2 enhances anchorage-independent growth and tumorigenicity, Cancer Res 59 (1999) 1141-1145. N. Kobayashi, S. Miyoshi, T. Mikami, H. Koyama, M. Kitazawa, M. Takeoka, et al., Hyaluronan deficiency in tumor stroma impairs macrophage trafficking and tumor neovascularization, Cancer Res 70 (2010) 7073-7083. B. Bernert, H. Porsch, P. Heldin, Hyaluronan synthase 2 (HAS2) promotes breast cancer cell invasion by suppression of tissue metalloproteinase inhibitor 1 (TIMP-1), J Biol Chem 286 (2011) 42349-42359. S. Shuster, G.I. Frost, A.B. Csoka, B. Formby, R. Stern, Hyaluronidase reduces human breast cancer xenografts in SCID mice, Int J Cancer 102 (2002) 192-197. A. Schmaus, S. Klusmeier, M. Rothley, A. Dimmler, B. Sipos, G. Faller, et al., Accumulation of small hyaluronan oligosaccharides in tumour interstitial fluid correlates with lymphatic invasion and lymph node metastasis, Br J Cancer 111 (2014) 559-567. R. Stern, A.A. Asari, K.N. Sugahara, Hyaluronan fragments: an information-rich system, Eur J Cell Biol 85 (2006) 699-715. K.N. Sugahara, T. Murai, H. Nishinakamura, H. Kawashima, H. Saya, M. Miyasaka, Hyaluronan oligosaccharides induce CD44 cleavage and promote cell migration in CD44-expressing tumor cells, J Biol Chem 278 (2003) 32259-32265. V. Voelcker, C. Gebhardt, M. Averbeck, A. Saalbach, V. Wolf, F. Weih, et al., Hyaluronan fragments induce cytokine and metalloprotease upregulation in human melanoma cells in part by signalling via TLR4, Exp Dermatol 17 (2008) 100-107. L.Y. Bourguignon, G. Wong, C.A. Earle, W. Xia, Interaction of low molecular weight hyaluronan with CD44 and toll-like receptors promotes the actin filament-associated protein 110-actin binding and MyD88-NFkappaB signaling leading to proinflammatory cytokine/chemokine production and breast tumor invasion, Cytoskeleton (Hoboken) 68 (2011) 671-693. K.L. Schwertfeger, M.K. Cowman, P.G. Telmer, E.A. Turley, J.B. McCarthy, Hyaluronan, Inflammation, and Breast Cancer Progression, Front Immunol 6 (2015) 236. D. Liu, E. Pearlman, E. Diaconu, K. Guo, H. Mori, T. Haqqi, et al., Expression of hyaluronidase by tumor cells induces angiogenesis in vivo, Proc Natl Acad Sci U S A 93 (1996) 7832-7837. 20

Page 20 of 29

[46]

[47]

[48]

[49]

[50]

[51] [52]

[53]

[54]

[55] [56]

[57] [58] [59]

[60]

V.B. Lokeshwar, M.J. Young, G. Goudarzi, N. Iida, A.I. Yudin, G.N. Cherr, et al., Identification of bladder tumor-derived hyaluronidase: its similarity to HYAL1, Cancer Res 59 (1999) 4464-4470. J.T. Posey, M.S. Soloway, S. Ekici, M. Sofer, F. Civantos, R.C. Duncan, et al., Evaluation of the prognostic potential of hyaluronic acid and hyaluronidase (HYAL1) for prostate cancer, Cancer Res 63 (2003) 2638-2644. B. Delpech, A. Laquerriere, C. Maingonnat, P. Bertrand, P. Freger, Hyaluronidase is more elevated in human brain metastases than in primary brain tumours, Anticancer Res 22 (2002) 2423-2427. R. Victor, C. Chauzy, N. Girard, J. Gioanni, J. d'Anjou, H. Stora De Novion, et al., Human breast-cancer metastasis formation in a nude-mouse model: studies of hyaluronidase, hyaluronan and hyaluronan-binding sites in metastatic cells, Int J Cancer 82 (1999) 77-83. C. Kuscu, N. Evensen, D. Kim, Y.J. Hu, S. Zucker, J. Cao, Transcriptional and epigenetic regulation of KIAA1199 gene expression in human breast cancer, PLoS One 7 (2012) e44661. E. Michishita, G. Garces, J.C. Barrett, I. Horikawa, Upregulation of the KIAA1199 gene is associated with cellular mortality, Cancer Lett 239 (2006) 71-77. K. Birkenkamp-Demtroder, A. Maghnouj, F. Mansilla, K. Thorsen, C.L. Andersen, B. Oster, et al., Repression of KIAA1199 attenuates Wnt-signalling and decreases the proliferation of colon cancer cells, Br J Cancer 105 (2011) 552-561. G.I. Frost, G. Mohapatra, T.M. Wong, A.B. Csoka, J.W. Gray, R. Stern, HYAL1LUCA1, a candidate tumor suppressor gene on chromosome 3p21.3, is inactivated in head and neck squamous cell carcinomas by aberrant splicing of pre-mRNA, Oncogene 19 (2000) 870-877. T.K. Nykopp, S. Pasonen-Seppanen, M.I. Tammi, R.H. Tammi, V.M. Kosma, M. Anttila, et al., Decreased hyaluronidase 1 expression is associated with early disease recurrence in human endometrial cancer, Gynecol Oncol 137 (2015) 152-159. X.B. Cheng, N. Sato, S. Kohi, K. Yamaguchi, Prognostic impact of hyaluronan and its regulators in pancreatic ductal adenocarcinoma, PLoS One 8 (2013) e80765. A. Kultti, C. Zhao, N.C. Singha, S. Zimmerman, R.J. Osgood, R. Symons, et al., Accumulation of extracellular hyaluronan by hyaluronan synthase 3 promotes tumor growth and modulates the pancreatic cancer microenvironment, Biomed Res Int 2014 (2014) 817613. R. Noy, J.W. Pollard, Tumor-associated macrophages: from mechanisms to therapy, Immunity 41 (2014) 49-61. B.Z. Qian, J.W. Pollard, Macrophage diversity enhances tumor progression and metastasis, Cell 141 (2010) 39-51. Y. Ohtaki, G. Ishii, K. Nagai, S. Ashimine, T. Kuwata, T. Hishida, et al., Stromal macrophage expressing CD204 is associated with tumor aggressiveness in lung adenocarcinoma, J Thorac Oncol 5 (2010) 1507-1515. Q.W. Zhang, L. Liu, C.Y. Gong, H.S. Shi, Y.H. Zeng, X.Z. Wang, et al., Prognostic significance of tumor-associated macrophages in solid tumor: a meta-analysis of the literature, PLoS One 7 (2012) e50946. 21

Page 21 of 29

[61]

[62]

[63]

[64]

[65] [66]

[67]

[68]

[69]

[70]

[71] [72] [73]

[74] [75]

J. Forssell, A. Oberg, M.L. Henriksson, R. Stenling, A. Jung, R. Palmqvist, High macrophage infiltration along the tumor front correlates with improved survival in colon cancer, Clin Cancer Res 13 (2007) 1472-1479. S. Shimura, G. Yang, S. Ebara, T.M. Wheeler, A. Frolov, T.C. Thompson, Reduced infiltration of tumor-associated macrophages in human prostate cancer: association with cancer progression, Cancer Res 60 (2000) 5857-5861. S.Y. Tan, Y. Fan, H.S. Luo, Z.X. Shen, Y. Guo, L.J. Zhao, Prognostic significance of cell infiltrations of immunosurveillance in colorectal cancer, World J Gastroenterol 11 (2005) 1210-1214. D.M. Kuang, Y. Wu, N. Chen, J. Cheng, S.M. Zhuang, L. Zheng, Tumor-derived hyaluronan induces formation of immunosuppressive macrophages through transient early activation of monocytes, Blood 110 (2007) 587-595. T.A. Wynn, A. Chawla, J.W. Pollard, Macrophage biology in development, homeostasis and disease, Nature 496 (2013) 445-455. S. Kim, H. Takahashi, W.W. Lin, P. Descargues, S. Grivennikov, Y. Kim, et al., Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis, Nature 457 (2009) 102-106. S. Tiainen, R. Tumelius, K. Rilla, K. Hamalainen, M. Tammi, R. Tammi, et al., High numbers of macrophages, especially M2-like (CD163-positive), correlate with hyaluronan accumulation and poor outcome in breast cancer, Histopathology 66 (2015) 873-883. W. Selbi, C.A. de la Motte, V.C. Hascall, A.J. Day, T. Bowen, A.O. Phillips, Characterization of hyaluronan cable structure and function in renal proximal tubular epithelial cells, Kidney Int 70 (2006) 1287-1295. C.A. de la Motte, V.C. Hascall, J. Drazba, S.K. Bandyopadhyay, S.A. Strong, Mononuclear leukocytes bind to specific hyaluronan structures on colon mucosal smooth muscle cells treated with polyinosinic acid:polycytidylic acid: inter-alpha-trypsin inhibitor is crucial to structure and function, Am J Pathol 163 (2003) 121-133. K.A. Scheibner, M.A. Lutz, S. Boodoo, M.J. Fenton, J.D. Powell, M.R. Horton, Hyaluronan fragments act as an endogenous danger signal by engaging TLR2, J Immunol 177 (2006) 1272-1281. D. Jiang, J. Liang, Y. Li, P.W. Noble, The role of Toll-like receptors in non-infectious lung injury, Cell Res 16 (2006) 693-701. B. Ruffell, L.M. Coussens, Macrophages and therapeutic resistance in cancer, Cancer Cell 27 (2015) 462-472. C. del Fresno, K. Otero, L. Gomez-Garcia, M.C. Gonzalez-Leon, L. Soler-Ranger, P. Fuentes-Prior, et al., Tumor cells deactivate human monocytes by up-regulating IL-1 receptor associated kinase-M expression via CD44 and TLR4, J Immunol 174 (2005) 3032-3040. S. Madar, I. Goldstein, V. Rotter, 'Cancer associated fibroblasts'--more than meets the eye, Trends Mol Med 19 (2013) 447-453. G.S. Karagiannis, T. Poutahidis, S.E. Erdman, R. Kirsch, R.H. Riddell, E.P. Diamandis, Cancer-associated fibroblasts drive the progression of metastasis through both paracrine and mechanical pressure on cancer tissue, Mol Cancer Res 10 (2012) 1403-1418. 22

Page 22 of 29

[76] [77] [78] [79]

[80]

[81]

[82] [83]

[84]

[85]

[86]

[87] [88]

[89]

[90]

[91]

G. Parsonage, A.D. Filer, O. Haworth, G.B. Nash, G.E. Rainger, M. Salmon, et al., A stromal address code defined by fibroblasts, Trends Immunol 26 (2005) 150-156. S. Vong, R. Kalluri, The role of stromal myofibroblast and extracellular matrix in tumor angiogenesis, Genes Cancer 2 (2011) 1139-1145. D. Ohlund, E. Elyada, D. Tuveson, Fibroblast heterogeneity in the cancer wound, J Exp Med 211 (2014) 1503-1523. K.D. Grugan, C.G. Miller, Y. Yao, C.Z. Michaylira, S. Ohashi, A.J. Klein-Szanto, et al., Fibroblast-secreted hepatocyte growth factor plays a functional role in esophageal squamous cell carcinoma invasion, Proc Natl Acad Sci U S A 107 (2010) 11026-11031. A. Tsuyada, A. Chow, J. Wu, G. Somlo, P. Chu, S. Loera, et al., CCL2 mediates crosstalk between cancer cells and stromal fibroblasts that regulates breast cancer stem cells, Cancer Res 72 (2012) 2768-2779. T. Ueno, M. Toi, H. Saji, M. Muta, H. Bando, K. Kuroi, et al., Significance of macrophage chemoattractant protein-1 in macrophage recruitment, angiogenesis, and survival in human breast cancer, Clin Cancer Res 6 (2000) 3282-3289. T.K. Ito, G. Ishii, H. Chiba, A. Ochiai, The VEGF angiogenic switch of fibroblasts is regulated by MMP-7 from cancer cells, Oncogene 26 (2007) 7194-7203. P.H. Chang, W.W. Hwang-Verslues, Y.C. Chang, C.C. Chen, M. Hsiao, Y.M. Jeng, et al., Activation of Robo1 signaling of breast cancer cells by Slit2 from stromal fibroblast restrains tumorigenesis via blocking PI3K/Akt/beta-catenin pathway, Cancer Res 72 (2012) 4652-4661. M. Cadamuro, G. Nardo, S. Indraccolo, L. Dall'olmo, L. Sambado, L. Moserle, et al., Platelet-derived growth factor-D and Rho GTPases regulate recruitment of cancerassociated fibroblasts in cholangiocarcinoma, Hepatology 58 (2013) 1042-1053. A.E. Postlethwaite, J. Keski-Oja, H.L. Moses, A.H. Kang, Stimulation of the chemotactic migration of human fibroblasts by transforming growth factor beta, J Exp Med 165 (1987) 251-256. Y. Li, D. Jiang, J. Liang, E.B. Meltzer, A. Gray, R. Miura, et al., Severe lung fibrosis requires an invasive fibroblast phenotype regulated by hyaluronan and CD44, J Exp Med 208 (2011) 1459-1471. C. Tolg, P. Telmer, E. Turley, Specific sizes of hyaluronan oligosaccharides stimulate fibroblast migration and excisional wound repair, PLoS One 9 (2014) e88479. C. Tolg, S.R. Hamilton, K.A. Nakrieko, F. Kooshesh, P. Walton, J.B. McCarthy, et al., Rhamm-/- fibroblasts are defective in CD44-mediated ERK1,2 motogenic signaling, leading to defective skin wound repair, J Cell Biol 175 (2006) 1017-1028. A.C. Midgley, M. Rogers, M.B. Hallett, A. Clayton, T. Bowen, A.O. Phillips, et al., Transforming growth factor-beta1 (TGF-beta1)-stimulated fibroblast to myofibroblast differentiation is mediated by hyaluronan (HA)-facilitated epidermal growth factor receptor (EGFR) and CD44 co-localization in lipid rafts, J Biol Chem 288 (2013) 1482414838. M. Quante, S.P. Tu, H. Tomita, T. Gonda, S.S. Wang, S. Takashi, et al., Bone marrowderived myofibroblasts contribute to the mesenchymal stem cell niche and promote tumor growth, Cancer Cell 19 (2011) 257-272. E.L. Spaeth, A.M. Labaff, B.P. Toole, A. Klopp, M. Andreeff, F.C. Marini, Mesenchymal CD44 expression contributes to the acquisition of an activated fibroblast 23

Page 23 of 29

[92]

[93]

[94] [95] [96]

[97]

[98]

[99]

[100]

[101]

[102]

[103]

[104]

[105]

phenotype via TWIST activation in the tumor microenvironment, Cancer Res 73 (2013) 5347-5359. A. Zoltan-Jones, L. Huang, S. Ghatak, B.P. Toole, Elevated hyaluronan production induces mesenchymal and transformed properties in epithelial cells, J Biol Chem 278 (2003) 45801-45810. T. Chanmee, P. Ontong, N. Mochizuki, P. Kongtawelert, K. Konno, N. Itano, Excessive hyaluronan production promotes acquisition of cancer stem cell signatures through the coordinated regulation of Twist and the transforming growth factor beta (TGF-beta)-Snail signaling axis, J Biol Chem 289 (2014) 26038-26056. S.M. Weis, D.A. Cheresh, Tumor angiogenesis: molecular pathways and therapeutic targets, Nat Med 17 (2011) 1359-1370. A.S. Chung, J. Lee, N. Ferrara, Targeting the tumour vasculature: insights from physiological angiogenesis, Nat Rev Cancer 10 (2010) 505-514. G.E. Davis, D.R. Senger, Endothelial extracellular matrix: biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization, Circ Res 97 (2005) 1093-1107. R. Montesano, S. Kumar, L. Orci, M.S. Pepper, Synergistic effect of hyaluronan oligosaccharides and vascular endothelial growth factor on angiogenesis in vitro, Lab Invest 75 (1996) 249-262. M. Slevin, J. Krupinski, S. Kumar, J. Gaffney, Angiogenic oligosaccharides of hyaluronan induce protein tyrosine kinase activity in endothelial cells and activate a cytoplasmic signal transduction pathway resulting in proliferation, Lab Invest 78 (1998) 987-1003. F.E. Lennon, T. Mirzapoiazova, N. Mambetsariev, B. Mambetsariev, R. Salgia, P.A. Singleton, Transactivation of the receptor-tyrosine kinase ephrin receptor A2 is required for the low molecular weight hyaluronan-mediated angiogenesis that is implicated in tumor progression, J Biol Chem 289 (2014) 24043-24058. B. Olofsson, H. Porsch, P. Heldin, Knock-down of CD44 regulates endothelial cell differentiation via NFkappaB-mediated chemokine production, PLoS One 9 (2014) e90921. R.C. Savani, G. Cao, P.M. Pooler, A. Zaman, Z. Zhou, H.M. DeLisser, Differential involvement of the hyaluronan (HA) receptors CD44 and receptor for HA-mediated motility in endothelial cell function and angiogenesis, J Biol Chem 276 (2001) 3677036778. S. Matou-Nasri, J. Gaffney, S. Kumar, M. Slevin, Oligosaccharides of hyaluronan induce angiogenesis through distinct CD44 and RHAMM-mediated signalling pathways involving Cdc2 and gamma-adducin, Int J Oncol 35 (2009) 761-773. K. Fuchs, A. Hippe, A. Schmaus, B. Homey, J.P. Sleeman, V. Orian-Rousseau, Opposing effects of high- and low-molecular weight hyaluronan on CXCL12-induced CXCR4 signaling depend on CD44, Cell Death Dis 4 (2013) e819. A.B. Ariffin, P.F. Forde, S. Jahangeer, D.M. Soden, J. Hinchion, Releasing pressure in tumors: what do we know so far and where do we go from here? A review, Cancer Res 74 (2014) 2655-2662. J.R. Less, M.C. Posner, Y. Boucher, D. Borochovitz, N. Wolmark, R.K. Jain, Interstitial hypertension in human breast and colorectal tumors, Cancer Res 52 (1992) 6371-6374. 24

Page 24 of 29

[106] B.D. Curti, W.J. Urba, W.G. Alvord, J.E. Janik, J.W. Smith, 2nd, K. Madara, et al., Interstitial pressure of subcutaneous nodules in melanoma and lymphoma patients: changes during treatment, Cancer Res 53 (1993) 2204-2207. [107] R. Gutmann, M. Leunig, J. Feyh, A.E. Goetz, K. Messmer, E. Kastenbauer, et al., Interstitial hypertension in head and neck tumors in patients: correlation with tumor size, Cancer Res 52 (1992) 1993-1995. [108] P.P. Provenzano, S.R. Hingorani, Hyaluronan, fluid pressure, and stromal resistance in pancreas cancer, Br J Cancer 108 (2013) 1-8. [109] P.P. Provenzano, C. Cuevas, A.E. Chang, V.K. Goel, D.D. Von Hoff, S.R. Hingorani, Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma, Cancer Cell 21 (2012) 418-429. [110] C.B. Thompson, H.M. Shepard, P.M. O'Connor, S. Kadhim, P. Jiang, R.J. Osgood, et al., Enzymatic depletion of tumor hyaluronan induces antitumor responses in preclinical animal models, Mol Cancer Ther 9 (2010) 3052-3064. [111] M.A. Jacobetz, D.S. Chan, A. Neesse, T.E. Bapiro, N. Cook, K.K. Frese, et al., Hyaluronan impairs vascular function and drug delivery in a mouse model of pancreatic cancer, Gut 62 (2013) 112-120. [112] J. Yang, R.A. Weinberg, Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis, Dev Cell 14 (2008) 818-829. [113] J. Xu, S. Lamouille, R. Derynck, TGF-beta-induced epithelial to mesenchymal transition, Cell Res 19 (2009) 156-172. [114] S. Lamouille, J. Xu, R. Derynck, Molecular mechanisms of epithelial-mesenchymal transition, Nat Rev Mol Cell Biol 15 (2014) 178-196. [115] A. Puisieux, T. Brabletz, J. Caramel, Oncogenic roles of EMT-inducing transcription factors, Nat Cell Biol 16 (2014) 488-494. [116] Y. Zhang, A.A. Thant, K. Machida, Y. Ichigotani, Y. Naito, Y. Hiraiwa, et al., Hyaluronan-CD44s signaling regulates matrix metalloproteinase-2 secretion in a human lung carcinoma cell line QG90, Cancer Res 62 (2002) 3962-3965. [117] S. Ghatak, S. Misra, B.P. Toole, Hyaluronan constitutively regulates ErbB2 phosphorylation and signaling complex formation in carcinoma cells, J Biol Chem 280 (2005) 8875-8883. [118] L.V. DeSouza, A. Matta, Z. Karim, J. Mukherjee, X.S. Wang, O. Krakovska, et al., Role of moesin in hyaluronan induced cell migration in glioblastoma multiforme, Mol Cancer 12 (2013) 74. [119] C.P. El-Haibi, G.W. Bell, J. Zhang, A.Y. Collmann, D. Wood, C.M. Scherber, et al., Critical role for lysyl oxidase in mesenchymal stem cell-driven breast cancer malignancy, Proc Natl Acad Sci U S A 109 (2012) 17460-17465. [120] H. Porsch, B. Bernert, M. Mehic, A.D. Theocharis, C.H. Heldin, P. Heldin, Efficient TGFbeta-induced epithelial-mesenchymal transition depends on hyaluronan synthase HAS2, Oncogene 32 (2013) 4355-4365. [121] M. Conacci-Sorrell, I. Simcha, T. Ben-Yedidia, J. Blechman, P. Savagner, A. Ben-Ze'ev, Autoregulation of E-cadherin expression by cadherin-cadherin interactions: the roles of beta-catenin signaling, Slug, and MAPK, J Cell Biol 163 (2003) 847-857.

25

Page 25 of 29

[122] T.T. Onder, P.B. Gupta, S.A. Mani, J. Yang, E.S. Lander, R.A. Weinberg, Loss of Ecadherin promotes metastasis via multiple downstream transcriptional pathways, Cancer Res 68 (2008) 3645-3654. [123] S. Yoshihara, A. Kon, D. Kudo, H. Nakazawa, I. Kakizaki, M. Sasaki, et al., A hyaluronan synthase suppressor, 4-methylumbelliferone, inhibits liver metastasis of melanoma cells, FEBS Lett 579 (2005) 2722-2726. [124] E. Arai, Y. Nishida, J. Wasa, H. Urakawa, L. Zhuo, K. Kimata, et al., Inhibition of hyaluronan retention by 4-methylumbelliferone suppresses osteosarcoma cells in vitro and lung metastasis in vivo, Br J Cancer 105 (2011) 1839-1849. [125] J.E. Draffin, S. McFarlane, A. Hill, P.G. Johnston, D.J. Waugh, CD44 potentiates the adherence of metastatic prostate and breast cancer cells to bone marrow endothelial cells, Cancer Res 64 (2004) 5702-5711. [126] T. Hiraga, S. Ito, H. Nakamura, Cancer stem-like cell marker CD44 promotes bone metastases by enhancing tumorigenicity, cell motility, and hyaluronan production, Cancer Res 73 (2013) 4112-4122. [127] B. Desai, M.J. Rogers, M.A. Chellaiah, Mechanisms of osteopontin and CD44 as metastatic principles in prostate cancer cells, Mol Cancer 6 (2007) 18. [128] A. Ouhtit, Z.Y. Abd Elmageed, M.E. Abdraboh, T.F. Lioe, M.H. Raj, In vivo evidence for the role of CD44s in promoting breast cancer metastasis to the liver, Am J Pathol 171 (2007) 2033-2039. [129] G.F. Weber, R.T. Bronson, J. Ilagan, H. Cantor, R. Schmits, T.W. Mak, Absence of the CD44 gene prevents sarcoma metastasis, Cancer Res 62 (2002) 2281-2286. [130] C. Qu, K. Rilla, R. Tammi, M. Tammi, H. Kroger, M.J. Lammi, Extensive CD44dependent hyaluronan coats on human bone marrow-derived mesenchymal stem cells produced by hyaluronan synthases HAS1, HAS2 and HAS3, Int J Biochem Cell Biol 48 (2014) 45-54. [131] V. Goncharova, N. Serobyan, S. Iizuka, I. Schraufstatter, A. de Ridder, T. Povaliy, et al., Hyaluronan expressed by the hematopoietic microenvironment is required for bone marrow hematopoiesis, J Biol Chem 287 (2012) 25419-25433. [132] P. Valent, D. Bonnet, R. De Maria, T. Lapidot, M. Copland, J.V. Melo, et al., Cancer stem cell definitions and terminology: the devil is in the details, Nat Rev Cancer 12 (2012) 767-775. [133] M.R. Alison, W.R. Lin, S.M. Lim, L.J. Nicholson, Cancer stem cells: in the line of fire, Cancer Treat Rev 38 (2012) 589-598. [134] A. Kreso, J.E. Dick, Evolution of the cancer stem cell model, Cell Stem Cell 14 (2014) 275-291. [135] H. Lu, K.R. Clauser, W.L. Tam, J. Frose, X. Ye, E.N. Eaton, et al., A breast cancer stem cell niche supported by juxtacrine signalling from monocytes and macrophages, Nat Cell Biol 16 (2014) 1105-1117. [136] T. Reya, H. Clevers, Wnt signalling in stem cells and cancer, Nature 434 (2005) 843-850. [137] J.E. Visvader, G.J. Lindeman, Cancer stem cells in solid tumours: accumulating evidence and unresolved questions, Nat Rev Cancer 8 (2008) 755-768. [138] H. Okuda, A. Kobayashi, B. Xia, M. Watabe, S.K. Pai, S. Hirota, et al., Hyaluronan synthase HAS2 promotes tumor progression in bone by stimulating the interaction of 26

Page 26 of 29

[139]

[140]

[141]

[142]

[143]

[144]

[145]

[146]

[147]

[148]

breast cancer stem-like cells with macrophages and stromal cells, Cancer Res 72 (2012) 537-547. L.Y. Bourguignon, G. Wong, C. Earle, L. Chen, Hyaluronan-CD44v3 interaction with Oct4-Sox2-Nanog promotes miR-302 expression leading to self-renewal, clonal formation, and cisplatin resistance in cancer stem cells from head and neck squamous cell carcinoma, J Biol Chem 287 (2012) 32800-32824. L.Y. Bourguignon, K. Peyrollier, W. Xia, E. Gilad, Hyaluronan-CD44 interaction activates stem cell marker Nanog, Stat-3-mediated MDR1 gene expression, and ankyrinregulated multidrug efflux in breast and ovarian tumor cells, J Biol Chem 283 (2008) 17635-17651. H. Shigeishi, A. Biddle, L. Gammon, H. Emich, C.O. Rodini, E. Gemenetzidis, et al., Maintenance of stem cell self-renewal in head and neck cancers requires actions of GSK3beta influenced by CD44 and RHAMM, Stem Cells 31 (2013) 2073-2083. S.A. Mani, W. Guo, M.J. Liao, E.N. Eaton, A. Ayyanan, A.Y. Zhou, et al., The epithelial-mesenchymal transition generates cells with properties of stem cells, Cell 133 (2008) 704-715. W. Guo, Z. Keckesova, J.L. Donaher, T. Shibue, V. Tischler, F. Reinhardt, et al., Slug and Sox9 cooperatively determine the mammary stem cell state, Cell 148 (2012) 10151028. L.Y. Bourguignon, C.C. Spevak, G. Wong, W. Xia, E. Gilad, Hyaluronan-CD44 interaction with protein kinase C(epsilon) promotes oncogenic signaling by the stem cell marker Nanog and the Production of microRNA-21, leading to down-regulation of the tumor suppressor protein PDCD4, anti-apoptosis, and chemotherapy resistance in breast tumor cells, J Biol Chem 284 (2009) 26533-26546. M. Tamada, O. Nagano, S. Tateyama, M. Ohmura, T. Yae, T. Ishimoto, et al., Modulation of glucose metabolism by CD44 contributes to antioxidant status and drug resistance in cancer cells, Cancer Res 72 (2012) 1438-1448. T. Ishimoto, O. Nagano, T. Yae, M. Tamada, T. Motohara, H. Oshima, et al., CD44 variant regulates redox status in cancer cells by stabilizing the xCT subunit of system xc() and thereby promotes tumor growth, Cancer Cell 19 (2011) 387-400. J. Ye, H. Wu, Y. Wu, C. Wang, H. Zhang, X. Shi, et al., High molecular weight hyaluronan decreases oxidative DNA damage induced by EDTA in human corneal epithelial cells, Eye (Lond) 26 (2012) 1012-1020. V. Grishko, M. Xu, R. Ho, A. Mates, S. Watson, J.T. Kim, et al., Effects of hyaluronic acid on mitochondrial function and mitochondria-driven apoptosis following oxidative stress in human chondrocytes, J Biol Chem 284 (2009) 9132-9139.

27

Page 27 of 29

Figure Legends Fig. 1. HA biosynthesis and degradation. (A) Structure of HA. HA is a negatively charged polysaccharide composed of repeating disaccharide units of glucuronic acid and Nacetylglucosamine. (B) HA is synthesized at the plasma membrane by HAS1-3. HAS enzymes catalyze the alternative addition of UDP-glucuronic acid and UDP-N-acetylglucosamine to the nascent HA chain and extrude it through the plasma membrane. (C) Mechanism of HA degradation by HYAL enzymes and KIAA1199. In HYAL-mediated degradation of HA, HMWHA is tethered to the cell surface by CD44 and GPI-anchored HYAL2 into caveolin-rich lipid rafts and then cleaved to form 20 kDa products. The HA fragments are subsequently delivered to endo-lysosome compartments and degraded into smaller oligosaccharides by HYAL1 and βexoglycosidase enzymes. In the proposed mechanism of KIAA1199-mediated HA degradation, KIAA1199 is an HA-binding protein required for HA catabolism via the clathrin-coated pit pathway. HA depolymerization may occur to generate HA oligosaccharides while in the clathrincoated vesicles or early endosomes. Fig. 2. The HA-rich tumor microenvironment recruits and activates stromal cells to enhance tumorigenicity. (A) Monocytes and MSCs exit from the bone marrow into the circulation in cytokine- and growth factor-dependent manners. These later extravasate to target organs and become recruited into tumor stroma. HMW-HA forms a continuous meshwork with its binding molecules, which include versican, TSG-6, and inter-α-inhibitor heavy chains (HC), to cause monocyte and MSC immobilization. Upon infiltrating into tumor stroma, LMW-HA stimulates the activation and differentiation of monocytes toward an M2 phenotype. (B) Accumulation of HA in the tumor stroma drives the differentiation and activation of TAFs. Elevated levels of HA 28

Page 28 of 29

promote directional infiltration of resident tissue fibroblasts and differentiation of MSCs into TAFs under the influence of growth factors. Increased HA may drive trans-differentiation of cancer cells into TAFs. (C) HA is an essential component required for tumor angiogenesis. During angiogenesis, both HMW- and LMW-HA stimulate endothelial cell migration, proliferation, and differentiation in a manner coordinated with growth factors, such as VEGF and b-FGF. LMW-HA also drives angiogenic responses by disrupting the endothelial barrier, which induces vascular the hyperpermeability that occurs in tumor angiogenesis. Fig. 3. HA-mediated induction of EMT and cancer stemness. (A) HA can trigger cancer cells to acquire a multipotent stem cell-like phenotype through EMT induction. HA regulates the expression of EMT-related transcription factors, such as Snail and Twist, which drive CSC selfrenewal and maintenance. (B) HA is a key player in tumor metastasis by inducing EMT and promoting ECM degradation. HA/CD44 interactions promote the PI3K/Akt signaling pathway and induce the MMP-2 and MMP-9 expression involved in the invasion and metastasis of cancer cells. Following the activation of CD44, ERM proteins crosslink CD44 to the actin cytoskeleton, thereby leading to an increase in cell migration. (C) HA/CD44 interactions regulate cancer stemness and multidrug resistance. HA/CD44 linking promotes the activation of stem cell regulators Rex1 and Sox2 through Nanog. Phosphorylation of Nanog induces multidrug resistance by upregulating MDR1expression.

29

Page 29 of 29