M I N I R E V I E W
Proteoglycans in health and disease: novel roles for proteoglycans in malignancy and their pharmacological targeting Achilleas D. Theocharis1, Spyridon S. Skandalis2, George N. Tzanakakis3 and Nikos K. Karamanos1
1 Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece 2 Ludwig Institute for Cancer Research Ltd, Uppsala, Sweden 3 Department of Histology, University of Crete, Heraklion, Greece
Keywords cancer; decorin; glypicans; lumican; molecular targeting; perlecan; proteoglycans; serglycin; syndecans; versican
Correspondence N. Karamanos, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26100 Patras, Greece Fax: +30 2610 997153 Tel: +30 2610 997915 E-mail: n.k.karamanos@upatras.gr
(Received 3 May 2010, revised 15 July 2010, accepted 29 July 2010)
doi:10.1111/j.1742-4658.2010.07800.x
The expression of proteoglycans (PGs), essential macromolecules of the tumor microenvironment, is markedly altered during malignant transforma- tion and tumor progression. Synthesis of stromal PGs is affected by factors secreted by cancer cells and the unique tumor-modified extracellular matrix may either facilitate or counteract the growth of solid tumors. The emerg- ing theme is that this dual activity has intrinsic tissue specificity. Matrix- accumulated PGs, such as versican, perlecan and small leucine-rich PGs, affect cancer cell signaling, growth and survival, cell adhesion, migration and angiogenesis. Furthermore, expression of cell-surface-associated PGs, such as syndecans and glypicans, is also modulated in both tumor and stro- mal cells. Cell-surface-associated PGs bind various factors that are involved in cell signaling, thereby affecting cell proliferation, adhesion and motility. An important mechanism of action is offered by a proteolytic processing of cell-surface PGs known as ectodomain shedding of syndecans; this facili- tates cancer and endothelial cell motility, protects matrix proteases and provides a chemotactic gradient of mitogens. However, syndecans on stro- mal cells may be important for stromal cell ⁄ cancer cell interplay and may promote stromal cell proliferation, migration and angiogenesis. Finally, abnormal PG expression in cancer and stromal cells may serve as a bio- marker for tumor progression and patient survival. Enhanced understand- ing of the regulation of PG metabolism and the involvement of PGs in cancer may offer a novel approach to cancer therapy by targeting the tumor microenvironment. In this minireview, the implication of PGs in cancer development and progression, as well as their pharmacological targeting in malignancy, are presented and discussed.
Introduction
Proteoglycans (PGs) are macromolecules composed of a specific core protein substituted with covalently
linked glycosaminoglycan (GAG) chains named chon- droitin sulfate (CS), dermatan sulfate (DS), keratan
Abbreviations ADAMTS, a disintegrin and metalloprotease domain with thrombospondin motifs; CS, chondroitin sulfate; DS, dermatan sulfate; ECM, extracellular matrix; EGF, epidermal growth factor; EGFR, epidermal growth factor receptor; FGF, fibroblast growth factor; GAG, glycosaminoglycan; HA, hyaluronan; HC, hepatocellular carcinoma; Hh, hedgehog; HS, heparan sulfate; HSPGs, heparan sulfate proteoglycans; MMP, matrix metalloproteinases; PDGF, platelet-derived growth factor; PG, proteoglycan; SLRPs, small leucine-rich proteoglycans; VEGF, vascular endothelial growth factor; VEGFR, vascular endothelial growth factor receptor.
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3904
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
their potential
as pharmacological
sulfate, heparin and heparan sulfate (HS). Hyaluronan (HA) is the only GAG synthesized in a free form not covalently bound on a core protein. GAGs are linear, negatively charged polysaccharides comprised of repeating disaccharides of acetylated hexosamines (N-acetyl-galactosamine or N-acetyl-glucosamine) and mainly by uronic acids (d-glucoronic acid or l-iduron- ic acid) being sulfated at various positions. Keratan sulfate is the only GAG to be comprised of repeating disaccharides containing N-acetyl-glucosamine and galactose.
Structural modifications of GAGs may facilitate tumorigenesis in various ways, modulating the func- tions of PGs. Our rapid increase in knowledge that PGs are among the key players in the tumor microen- vironment and can modulate tumor progression, sug- gests targets. Pharmacological treatment may target PG metabolism, their utilization as targets for immunotherapy or their direct use as therapeutic agents. In this minireview, we focus on the roles of PG in cancer development and progression, as well as their pharmacological targeting in malignancy.
Extracellular matrix PGS
further
Hyalectans
The subfamily of hyalectans includes versican, aggre- can, neurocan and brevican. Hyalectans have the abil- ity to bind HA through their N-terminal globular domain (G1), the central domain carries most of the GAG chains and exhibits lectin-like activity located in the C-terminal globular domain (G3) (Fig. 1) [1].
in cancer and
Versican expression and functions inflammation
functions acting as
Versican is expressed throughout the body and pro- vides ECM with hygroscopic properties creating a loose and hydrated matrix that is necessary to support key events in development and disease. Four splice- variants of human versican (V0, V1, V2 and V3) have been identified. The differences among the versican splice-variants are found in the central portion of the protein core, and they vary in the number and
PGs can be classified into three main groups accord- ing to their localization, extracellularly secreted, those associated with the cell surface and intracellular. Each main group is classified into subfamilies according to their gene homology, core protein proper- ties, size and modular composition. Secreted PGs involve large aggregating PGs, named hyalectans, small leucine-rich PGs (SLRPs) and basement mem- brane PGs. Cell-surface-associated PGs are divided into two main subfamilies (syndecans and glypicans), whereas serglycin is the only intracellular PG charac- terized to date [1,2]. The wide molecular diversity of PGs derives from the multitude of possible combina- tions of protein cores, O-linked and N-linked oligosac- charides, and various types and numbers of GAG chains. The specific structural characteristics of GAG types provide some of the structural basis for the mul- titude of their biological functions [3]. PGs exhibit structural numerous biological components in tissue organization, and affect several cellular parameters, such as cell proliferation, adhe- sion, migration and differentiation. PGs interact with growth factors and cytokines, as well as with growth factor receptors, and are implicated in cell signaling. The catalog of physiological functions and the patho- biological roles in which PGs are involved have grown rapidly.
Hyalectans and SLRPs
Hyaluronan
GAG chains
EGF like motifs
Versican
Lectin-like repeat
Complement regulatory protein
Aggrecan
Collagen I
Neurocan
Brevican
SLRPs
Fig. 1. Schematic representation of hyalectans (hyaluronan binding PGs) and SLRPs found in ECM.
During carcinogenesis, malignant cells secrete solu- ble growth factors that stimulate cell growth and acti- vate stromal cells to secrete effectors that in turn stimulate further tumor cell growth. Both activated stromal and tumor cells are implicated in the reorgani- zation of the extracellular matrix (ECM) to facilitate tumor cell growth, migration and invasion. PG expres- sion is markedly modified in the tumor microenviron- ment. Altered expression of PGs on tumor and stromal cell membranes affects cancer cell signaling, growth and survival, cell adhesion, migration and angiogenesis. The type and fine structure of GAG chains attached to PGs are markedly affected in the context of malignant transformation as result of the altered expression of GAG-synthesizing enzymes.
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3905
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
thrombospondin motifs (ADAMTS), matrix metallo- proteinases (MMPs) and plasmin can cleave versican generating fragments containing the G1 domain in some cases [4,5]. Thus, regulation of G1 and G3 versi- can levels by proteases is an important factor in cancer cell motility and metastasis (Table 1).
presence of GAGs. Versican is able to regulate many cellular processes including adhesion, proliferation, apoptosis, migration, invasion and ECM assembly via the highly negatively charged CS ⁄ DS side chains, and by interactions of the G1 and G3 domains with other proteins [4–6]. Versican binds to ECM components such as HA, type I collagen, tenascin-R, fibulin-1 and -2, fibrillin-1, fibronectin, P- and L-selectin and chemokin- es. Versican also binds to the cell-surface proteins CD44, integrin b1, epidermal growth factor receptor (EGFR), P-selectin glycoprotein ligand-1 [6,7] and toll- like receptor 2 [8].
of malignancies.
Indeed,
Versican may also promote the formation of an inflammatory microenvironment in the tumor stroma. The interaction between versican and toll-like receptor- 2 links inflammation and metastasis [8,32]. Ligation of toll-like receptor-2 present on endothelial cells and fibroblasts by versican activates these cells and triggers the secretion of inflammatory cytokines. This is mecha- nistically important insofar as inflammation is often associated with cancer initiation and promotion, and inflammatory cells (primarily macrophages) are consis- tently regarded as critical mediators in the develop- ment tumor-associated macrophages can enhance angiogenesis, ECM degrada- tion and remodeling, and can promote cancer cell invasion [32].
types
Notably, the versican gene promoter contains a p53- binding site in its first intron and can be directly acti- vated by the tumor suppressor p53 in a dose-depen- dent manner [33,34]. It was found to be a target gene of Wnt signaling in human embryonic carcinoma cells [35].
Versican regulation and targeting
V0 and V1 are the predominant isoforms present in cancer tissues [9–12]. Overexpression of the V3 isoform in melanoma cells markedly reduces cell growth in vitro and in vivo [13], and promotes pulmonary metastases [14]. V3 isoform may have a dual role as an inhibitor of tumor growth and a stimulator of metasta- sis. Elevated levels of versican have been reported in most malignancies to date and have been associated with cancer relapse and poor patient outcome in breast, [10,15–21] prostate and many other cancer (Table 1). We have seen that versican is accumulated in the preclinical phase of breast cancer in nonpalpable breast carcinomas (Lambropoulou et al., personal data). This accumulation seems to be associated with risk factors because increased mammographic density and malignant appearing microcalcifications are found in these cases. Versican appears to be most commonly secreted by the activated peritumoral stromal cells in adenocarcinomas [20,21]. However, human pancreatic cancer cells can also secrete versican [22].
to fibronectin-coated surfaces
reducing this powerful
several protease families
Functional studies have demonstrated that versican can increase cancer cell motility [12,23–25], prolifera- tion [26] and metastasis [27]. Cancer cells can form a polarized pericellular sheath through compartmental- ized cell-surface CD44 expression and subsequent assembly of HA ⁄ versican aggregates that promotes their motility (Fig. 2) [25]. Soluble versican is able to reduce the attachment of prostate cancer and mela- in vitro noma cells [28,29]. Both the G1 and G3 versican domains have been shown to promote cell proliferation in NIH-3T3 fibroblasts and tumor cells [23,24,30,31] (Table 1). The G1 domain of versican stimulates proliferation by destabilizing cell adhesion, whereas the G3 domain induces proliferation, at least in part, by activating EGFR via the action of epidermal growth factor (EGF)-like motifs (Fig. 2). G1 and G3 domains may differentially control tumor growth rate and have interactive roles to promote tumor development and metastasis. Notably, that include a disintegrin and metalloprotease domain with
Matrix effectors including platelet-derived growth fac- tor (PDGF), transforming growth factor b, interleukin 1b, angiotensin II and steroid hormones affect versican synthesis in various normal cell lines [6]. Transforming growth factor b has been found to regulate synthesis of versican in fibrosarcoma, osteosarcoma and glioma cells [11,12,36]. The expression of versican can be also insulin-like growth factor I and modified by EGF, PDGF-BB in malignant mesothelioma cells [37]. The effect of protein tyrosine kinase signaling pathways on versican synthesis can be reversed following treatment with various tyrosine kinase inhibitors [38]. Therefore, targeting versican synthesis may be a potential mecha- nism for tumor-promoting agent. In agreement with this concept, the tyrosine kinase inhibitor genistein can block versican expression induced by growth factors in malignant mesothelioma cell lines [37]. Genetic and preclinical studies support the targeting of growth factor [transforming growth factor b, PDGF, EGF and vascular endothelial growth factor (VEGF)] signaling as a therapeutic strategy for combating cancer. To date, several approaches to inhi- bit growth factor signaling pathways in cancer have
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3906
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
Table 1. Expression of proteoglycans in malignancies and possible pharmacological targeting. ADAMTS, a disintegrin and metalloprotease domain with thrombospondin motifs; HA, hyaluronan; MMP, matrix metalloproteinases.
Possibilities for potential pharmacological targeting
Proteoglycan subfamilies
Expression in malignancies ⁄ roles
Hyalectans Versican
› Brain tumors, melanomas, osteosarcomas, lymphomas, acute monocytic leukemia, testicular tumors, breast, prostate, colon, lung, pancreatic, endometrial, ovarian and oral cancers
Prognostic factor, proliferation, adhesion, motility,
metastasis, angiogenesis
Inhibition of synthesis (a) Blocking of growth factors; blocking antibodies, kinase inhibitors, antisense oligonucleotides, (b) miRNA-based therapeutics; miR-199a* Inhibition of degradation (a) ADAMTS, (b) MMPs inhibitors Inhibition of interaction with HA (a) HA oligomers
Aggrecan
Brevican
fl Laryngeal cancer Cartilage degradation › Glioma Diagnostic marker, adhesion, motility
Inhibition of cleavage (a) ADAMTS, (b) MMPs inhibitors Immunotherapy
SLRPs Decorin
› Osteosarcoma, testicular tumors, ovarian, colon, gastric, pancreatic, laryngeal, breast cancer
Upregulation of synthesis (a) Viral-mediated delivery, (b) Demethylation agents, (c) Proteasome inhibitors Admistration of decorin protein or synthetic peptides
Lumican
Prognostic factor, inhibition of growth and migration (fl ErbB and Met signaling) › Osteosarcoma, melanoma, breast cancer Prognostic factor, inhibition of growth and migration
Upregulation of synthesis (a) Viral-mediated delivery Admistration of lumican protein or synthetic peptides
Basement membranes Collagen XVIII
Admistration of endostatin fragment or synthetic peptides Endostatin producing cells in immunoisolation devices
Perlecan
Admistration of endorepellin fragment or synthetic peptides
Agrin
› Ovarian, pancreatic cancer fl Liver and oral cancer Promoter of growth and angiogenesis Inhibitor of angiogenesis (endostatin) › Liver, oral tumors, melanoma Promoter of growth and angiogenesis Inhibitor of angiogenesis (endorepellin) › Liver tumors. Promoter of growth and angiogenesis? Inhibitor of angiogenesis (C-terminus)?
Admistration of C-terminal fragment or synthetic peptides
Cell surface Glypican-1
› Breast, pancreatic cancer Proliferation, growth factor signaling
Glypican-3
Inhibition of synthesis (a) Blocking of growth factors; blocking Abs, kinase inhibitors, antisense oligonucleotides, (b) siRNA-based therapeutics Immunotherapy Admistration of soluble fragments Immunotherapy
fl Lung, gastric, ovarian, breast cancer and mesothelioma Inhibition of proliferation and induction of apoptosis › Neuroblastoma, Wilm’s tumor, hepatocellular carcinoma, melanoma
Syndecan-1
Immunotherapy Heparanase inhibitors
Promoter of tumor cell growth › Breast, prostate, head and neck, myeloma fl Lung, colorectal, endometrial, cervical gastric, pancreatic cancer
Prognostic factor, proliferation, adhesion, migration,
Syndecan-2
Immunotherapy Heparanase inhibitors
differentiation, angiogenesis › Lung, ovarian, osteosarcoma, brain tumors, colon, mesothelioma
Syndecan-4
Proliferation, adhesion, migration, angiogenesis › Breast cancer, melanoma Adhesion and migration
Immunotherapy Heparanase inhibitors
Intracellular Serglycin
?
› Hematological malignancies Diagnosis
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3907
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
Biological roles of versican
expression system, may be useful for targeting versican among the other genes involved in tumorigenesis [41] (Table 1).
Versican
Hyaluronan
EGFR
G3 domain
CD44
P
P
EGFR signaling Reduced cell adhesion
Tumor / endothelial cell proliferation and motility
future
The use of an antibody against to the ADAMTS- specific versican cleavage site inhibits glioma cell migration in a dose-dependent manner, suggesting that the local accumulation of versican fragments may also promote cancer cell motility and invasion (Fig. 2) [12]. Concurrent with this hypothesis, processed versican fragments have been identified in the peritumoral stroma of prostate cancer in association with higher ADAMTS1 and ADAMTS4 levels, but not in the interface surrounding normal prostate glands where full-legth versican is present [42]. Notably, the broad- spectrum MMP inhibitor GM6001 (Galardin), which inhibits the activity of MMPs and ADAMTS prote- invasion ases, has been shown to inhibit cancer cell and metastasis in a transgenic breast cancer model [43]. Other protease inhibitors such as catechin gallate esters, present in natural sources (green tea) have been shown to selectively inhibit ADAMTS-1, -4 and -5 and aggrecan catabolism in cartilage [44]. The ability of MMPs and ADAMTS inhibitors to prevent versican catabolism and versican-induced motility and metasta- sis may be an interesting area of study (Table 1).
Manipulation of
Fig. 2. Versican interacts with hyaluronan creating large aggre- gates. CD44 interacts with HA and versican forming a polarized and highly hydrated pericellular sheath that destabilizes cell adhe- sion and promotes cell motility. These aggregates facilitate further cell-shape changes and promote cell proliferation. EGF-like motifs present in the G3 domain of versican activate EGFR promoting cell proliferation and motility.
there are no data to show that
is
synthesis
regulated
the versican catabolic pathways may also provide novel therapeutic targets for cancer invasion and metastasis. For example, formation of a pericellular matrix rich in HA and versican implicated in cancer cell motility, could be inhibited by treatment with HA oligomers. Disruption of the HA–CD44 interaction with HA oligomers has been shown to markedly inhibit the growth of B16F16 melanoma cells [45]. In addition, HA oligomers inhibit the formation of receptor tyrosine kinases complexes and their phos- phorylation in prostate, colon and breast carcinoma cells [46]. Thus, the use of HA oligomers is a poten- tially attractive agent to block the formation of large versican–HA aggregates and HA–CD44 interactions, as well as local tumor invasion (Table 1).
Aggrecan degradation during cartilage destruction
been investigated. These approaches mainly include: (a) inhibition at the translational level using antisense oligonucleotides that can be engineered into immune cells or delivered directly into tumors, (b) inhibition of the ligand–receptor interaction using monoclonal anti- bodies, and (c) inhibition of the receptor-mediated sig- naling cascade using specific tyrosine kinase inhibitor. However, such approaches are effective in inhibiting the effects of versican in cancer cell models (Table 1). Interestingly, versican by microRNAs (miRNA). miR-199a* function as a guide molecule in post-transcriptional gene silencing by binding to region (3¢-UTR) of versican the 3¢-untranslational mRNA, leading to translational repression. miR-199a* is considered to be an onco-suppressor, targeting mole- cules critically involved in the promotion of tumor growth and is often downregulated in malignancies [39,40]. Several companies have developed miRNA- based therapeutics and a strategy that increases the natural dose of miR-199a*, by introducing a short double-stranded synthetic RNA that is loaded into RNA-induced silencing complex or by utilizing expres- the hairpin pre-miRNA in a viral vector sion of
Aggrecan is a large molecule found mainly in cartilage and brain, almost exclusively in the form of aggregates with HA (Fig. 1). It is a main matrix organizer also involved in the regulation of cartilage development, growth and homeostasis. Several studies describe its involvement in malignancies. Aggrecan is markedly reduced and degraded at specific sites during cartilage destruction in the progression of laryngeal squamous cell cancer [47,48] (Table 1).
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3908
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
Brevican: roles in gliomas
Biological roles of SLRPs / decorin
Formation and spacing of collagen fibrils
Collagen I
SLRPs
Decorin
EGFR
c-Met
P
P
P
P c-Cbl
role in glioma progression.
In addition,
Induction of p21
Signaling modulation proteasome degradation
Brevican and neurocan, found in the central nervous system, affect neuronal attachment and neurite out- growth (Fig. 1). Upregulation and proteolytic cleavage of brevican increase the aggressiveness of glial tumors significantly [49] and enhance cell adhesion and motil- ity. Brevican promotes EGFR activaton, increases the expression of cell-adhesion molecules and promotes the secretion of fibronectin and accumulation of fibro- nectin microfibrils on the cell surface [50]. The expres- sion of a novel tumor-specific isoform of brevican that is localized on the cell membrane has been found in all high-grade gliomas and is suggested to play a signifi- cant the absence of brevican from benign gliomas prompts its use as a diagnostic marker to distinguish primary brain tumors of similar histology, but different pathologic course [51]. Inhibition of the expression of the tumor- specific isoform of brevican and inhibition of brevican cleavage may be a potential pharmacological target for the treatment of brain tumors (Table 1).
Inhibition of tumor cell growth and migration
Endocytosis degradation
Small leucine-rich PGs
Fig. 3. SLRPs have a curved molecular architecture and bind to col- lagen fibrils through the core protein participating in the formation and spacing of the fibrils. Decorin binds to the EGFR and evokes a unique signaling cascade that causes EGFR dimerization, caveolin- mediated internalization and lysosomal degradation. EGFR is phos- phorylated upon decorin binding, and induces the expression of p21 leading to growth arrest. Decorin binds also to the c-Met induc- ing transient activation of the receptor recruitment of c-Cbl and rapid intracellular degradation of the receptor by the proteasome. Upon decorin binding to c-Met, intracellular levels of b-catenin are suppressed inhibiting tumor cell growth and migration.
SLRPs are characterized by a protein core with leu- cine-rich repeats, the presence of N-terminal cysteine clusters and C-terminal ‘ear repeats’ (classes I–III), and at least one GAG side chain [52]. The family of SLRPs is now divided into five classes (see the minire- view by Iozzo & Schaefer [53]). Most of these SLRPs have a consensus sequence for modification with GAGs, but some exist as glycoproteins in the tissues. SLRPs are important regulators of various biological processes because leucine-rich repeats are particularly relevant for protein–protein interactions [52]. SLRPs have a curved molecular architecture and bind to col- lagen fibrils through a core protein, participating in the formation and spacing of the fibrils (Fig. 1). Most studies on the role of SLRPs in malignancy have focused on decorin and lumican, which are typically located around tumor microenvironment.
Decorin signaling in cancer
Decorin represents a powerful tumor cell growth and migration inhibitor by modulating both tumor stroma deposition and cell-signaling pathways. In addition, genetic evidence suggests that a lack of decorin is ‘per- missive’ for tumor development [54]. Growth arrest of lines has been associated with various tumor cell induced expression of inhibitors of cyclin-dependent [55,56]. Notably, decorin binds kinase p21 (Fig. 3)
directly to EGFR and downregulates its activity as well as the activity of other members of the ErbB fam- ily (Fig. 3) [55,57]. These receptors are overexpressed and ⁄ or mutated in many cancers driving tumor pro- gression [58]. Decorin can compete with EGF for receptor binding on the cell surface of tumor cells. After binding, the receptor dimerizes and is subse- quently internalized and degraded in the lysosomes via caveolin-mediated internalization (Fig. 3) [59]. Decorin inhibits tumor cell proliferation by evoking a signaling cascade that is different from the one evoked by EGF, possibly by inducing a different EGFR conformation and selectively activating phosphotyrosines in the receptor autophosphorylation domain. Decorin also suppresses the activity of ErbB2 and ErbB4 receptors via degradation (Fig. 3) [57,60]. The only exception to
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3909
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
recruitment of
further investigation. Decorin expression in cancer can also be altered by transcriptional, post-transcriptional and post-translational modifications. Notably, decorin production is re-established following treatment with a proteasome inhibitor, suggesting that ovarian cancer cells have developed a mechanism for the rapid degra- dation of decorin [67]. Potentially, epigenetic control, including hypermethylation of the promoter region, might play a role in silencing the decorin gene. In the tumor stroma, by contrast, hypomethylation of the decorin promoter has been previously demonstrated [1]. In addition, tumor cells can synthesize soluble factors that repress decorin expression by stromal cells [70].
this model reported to date has been in MG-63 human osteosarcoma cells, in which the decorin-expressing tumor cells show overexpression paired to constitutive activation of the EGFR favoring cell migration [61]. Decorin also interacts with Met, the receptor for hepa- tocyte growth factor, and induces transient receptor activation, the E3 ubiquitin ligase c-Cbl, and rapid intracellular degradation of the recep- tor (Fig. 3). Decorin suppresses intracellular levels of b-catenin, one of the key downstream effectors of Met, and inhibits cell migration and growth (Fig. 3). Thus, by antagonistically targeting multiple tyrosine kinase receptors, decorin contributes to the reduction in pri- mary tumor growth and metastastic spreading [62].
Decorin as potential anticancer agent
Decorin as a marker for prognosis and aggressiveness
Decorin expression is altered in various types of cancer (Table 1). Therefore, decorin has been taken into con- sideration as a possible prognostic marker in cancer patients. It is important to note that decorin expres- sion levels are directly proportional to the amount of tumor stroma. There are very few studies that analyze the prognostic significance of decorin expression levels in terms of patient survival. High expression of decorin in patients with advanced ovarian cancer was associ- ated with a poor response to treatment and a higher incidence of relapse for those patients that initially responded [63]. Reduced amounts of decorin were associated with poor prognosis in node-negative inva- sive breast cancer [64] and some types of soft tissue tumors [65]. Low decorin levels in liposarcomas and malignant peripheral nerve sheath tumors are also associated with lower disease-free and survival rates (Table 1). Decorin is also deposited in the tumor stroma in nonpalpable breast carcinomas as versican. The accumulation of decorin is more pronounced than that of versican and is also associated with cancer-sug- gesting mammogram findings (personal data).
[66].
It
In normal tissues, the ratio of CS and DS side chains associated with decorin is balanced. In tumor tissue, such as colon, ovarian, gastric and pancreatic the CS chains become predominant carcinoma, [66–69]. DS is chemically more complex and requires additional enzymes to be synthesized, compared with CS. Therefore, it is reasonable to think that the syn- thesis of the chemically simpler CS is favored in tumor tissue. In line with this concept, CS side chains have been proposed as being more permissive to cell migra- tion favoring tumor aggressiveness is not surprising that, tumor cells, with very few exceptions, do not express decorin, but the mechanism by which decorin expression is switched off is unclear and needs
Decorin will attract more interest in future as an anti- cancer therapeutic. Considering that chemotherapy is still the leading therapy for cancer patients and that decorin could be administered in concomitance with various compounds, it is relevant to understand their interaction. Decorin shows a synergistic biological effect with carboplatin in inhibiting ovarian cancer cell growth [71], whereas it antagonizes carboplatin and gemcitabine effects against pancreatic cancer cells [72]. Because of a lack of data and somewhat contrasting results, this area definitely needs more investigation. Considerable effort has been recently applied to prove that decorin can be an antitumor therapeutic in vivo. the Adenoviral-mediated delivery of decorin slows growth of lung, squamous and colon carcinoma tumor xenografts in immunocompromised mice [73], retards mammary adenocarcinoma growth and prevents meta- static spreading to the lungs reducing ErbB2 receptor levels [66]. Ectopic expression of decorin in a rat glioma model prolongs the survival of the animals and the size of the tumor is directly proportional to how early and how much decorin is expressed [74]. Admin- istration of decorin protein core showed that it specifi- cally localizes within the tumor, antagonizes EGFR activity and induces apoptosis in the A431 squamous carcinoma model [75]. The outcome is primary tumor growth inhibition because of the slower growth rate combined with apoptosis and impaired tumor metabo- lism. Decorin injected systemically can reduce breast tumor growth and metabolism and halt metastatic spread to the lungs [76]. The finding that ectopic expression of decorin can revert the malignant pheno- type in several cell lines of various histogenetic back- grounds [56] and can antagonize primary tumor growth and metastases in vivo further raises a hope for the postulated clinical application of decorin and related molecules. Decorin might be utilized in the
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3910
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
near future as an adjunct ‘protein therapeutic’ for solid tumors in which receptor tyrosine kinases play a key in vivo studies with various tumor role. Additional models are desirable. Furthermore, studies to elucidate the functional interaction between decorin and existing anticancer chemotherapeutics to evaluate in order potential limitations are needed (Table 1).
Lumican: a promising agent in the cancer fight
inhibit melanoma cell migration [81]. It will be interest- ing in future to test the effects of recombinant lumican on tumor growth and metastasis. Additional studies carried out with different cancer types are required to dissect the mechanism of action of lumican. However, it appears that lumican targeting is promising and more specific information about its mode of action is needed. SLRPs or peptides derived therefrom could be applied in the fight against cancer, because they repre- sent a class of natural inhibitors of cancer growth (Table 1).
anchorage-independent
Basement membrane PGs
[78] and its
Lumican is substituted with keratan sulfate side chains, and inhibits cell proliferation by inducing p21 expres- cell sion and antagonizes growth as well as cell migration [77]. Cleavage of lumi- can by the membrane-type matrix metalloprotease-1 abrogates lumican-evoked suppression of colony for- mation [77]. Interestingly, and on the whole contrary to decorin, lumican is expressed by some tumor cell lines inhibition promotes cancer cell growth in some cases. Transfection of B16F1 mouse melanoma cells to express lumican or treatment with recombinant protein induces impaired anchorage-inde- pendent growth and the capacity to invade the ECM [79]. Lumican has been reported to be more abundant than decorin in human breast carcinomas [80], even though the clinical relevance of this observation has not yet been explained. More importantly, lumican expression has been proposed as a prognostic factor in lymph node-negative breast cancer [64]. A recent study tested different recombinant and synthetic peptides and found an active site in the leucine-rich repeat 9 domain of the lumican core protein, which is able to
Basement membranes are thin layers of specialized ECM underlying epithelial and endothelial layers; a located in basement specific class of matrix PGs membranes with particular structural and functional characteristics. Basement membranes are elongated molecules with a collage of domains that share struc- tural and functional homology with numerous ECM proteins, growth factors and surface receptors. This class involves three main, well-characterized members: perlecan, collagen type XVIII and agrin, which are almost universally decorated with HS side chains (Fig. 4) [82]. Their genes are highly conserved and carry disparate biological functions for the mainte- nance of basement membrane homeostasis, modulation of growth factor activity and angiogenesis. Basement membrane heparin sulfate proteoglycans (HSPGs) have a dual function as pro- and anti-angiogenic factors participating in cancer growth and angiogenesis [82].
Endorepellin
Perlecan
SEA
GAG chains
Laminin-EGF-like domain repeats
Laminin domain IV
LDL-receptor class A repeat
EGF-like repeats
Ig-like repeats
Laminin G domain
Agrin
Follistatin-like repeats
Basement membrane PGs
Ser / Thr-rich domains
N-terminal agrin
Endostatin
C-terminal domain
N-terminal domain
Collagen XVIII
Non-collagenous sequences
Interrupted triple helical domain
Fig. 4. Schematic representation showing the structural domains of the basement membrane PGs perlecan, agrin and collagen XVIII.
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3911
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
They can stimulate angiogenic signaling by sequester- ing, protecting and concentrating HS-binding growth factors, such as fibroblast growth factor-2 (FGF-2), VEGF and PDGF, through which the HSPG growth factor complex may be presented in a ‘biologically active’ form to the cognate receptors (Fig. 5). By con- trast, they contain powerful angiostatic fragments, such as endostatin and endorepellin at their C-termini that are released by proteolytic processing and can act in a paracrine function on sprouting endothelial cells, either locally or distantly (Fig. 5).
Perlecan: a critical regulator of growth factor- mediated signaling and angiogenesis
angiogenesis
tumor
and
response, as
Perlecan binds FGF-2 via HS, promotes receptor acti- vation and ultimately downstream signaling, which supports mitogenesis and angiogenesis (Fig. 5). The lack of HS in perlecan inhibits wound healing and FGF-2-induced growth [82,83]. Targeted knockdown of perlecan reduced the revealed by decreased growth factor tumor growth and angiogenesis [82,84]. In hepatoblas- toma xenografts treated with anti-vascular endothelial
growth factor receptor (VEGFR) therapy, vessel recov- ery over time was associated with an increase in perlec- an and heparanase expression around tumor vessels [85], suggesting a synergistic role of heparanase [86] in the liberation of HS-bound and ⁄ or proteases [87] VEGF and subsequent VEGFR2 activation (Fig. 5). The C-terminal domain of perlecan, called endorepel- lin, blocks endothelial cell migration and capillary morphogenesis both in vitro and in vivo [82]. Endore- pellin interacts specifically with the a2b1 integrin [88], and stimulation with endorepellin induces the interac- tion and phosphorylation of Src homology-2 protein phosphatase-1 with integrin a2 in a dynamic fashion in endothelial cells [89], triggering a signaling cascade that leads to disruption of the actin cytoskeleton and thus to cytostasis (Fig. 5) [88]. Systemic delivery of human recombinant endorepellin to tumor xenograft- bearing mice causes a marked suppression of tumor growth and metabolic rate mediated by a sustained downregulation of the tumor angiogenic network [90]. The distal laminin-like globular domain (LG3) pos- sesses most of the biological activity [82,88] and can be released from the parent molecule by bone morphoge- [91]. netic protein-1 ⁄ Tolloid-like metalloproteinases
Biological roles of basement membrane PGs
Perlecan
GF
GF
GF
Agrin
Protection of growth factors chemotactic gradient
Tumorigenesis angiogenesis
GF
Collagen XVIII
GF
GF
Endostatin
Endorepellin
Glypicans
VEGFR
Growth factor receptor
GF
α β
GF
2
1
G F G F
Integrins
P
P
Growth factor receptor signaling
Disruption of actin cytoskeleton down-regulation of VEGF signaling synthesis of thrombospondin
Inhibition of angiogenesis
Tumor / endothelial cell proliferation and motility
Fig. 5. PGs of the basement membrane bind HS-binding growth factors and stimulate angiogenesis and tumorigenesis by sequestering, pro- tecting and concentrating growth factors. HSPG–growth factor complexes are presented in a ‘biologically active’ form to the cognate recep- tors promoting their signaling. By contrast, they contain angiostatic fragments, such as endostatin and endorepellin, which are released by proteolytic cleavage. Endorepellin interacts with a2b1 integrin inducing the interaction and phosphorylation of src homology-2 protein phos- phatase-1 with integrin a2, triggering a signaling cascade that leads to disruption of the actin cytoskeleton and thus to cytostasis. Endostatin binds to integrins (a5b1, avb3, avb5), glypican and VEGFR2 affecting several key components of VEGF signaling cascade, stimulating synthe- sis of thrombospondin (a powerful angiostatic protein) and suppressing c-myc. Endostatin induces reprogramming of the gene expression and disrupts endothelial cell migration.
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3912
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
macroencapsulation of engineered cells that produce endostatin may be innovate therapeutic strategy for treatment of malignancies (Table 1).
Agrin: roles in tumor angiogenesis
layer of control
LG3 has been detected in pathological conditions including pancreatic, colon and breast cancer [82]. It has been proposed that endorepellin ⁄ LG3 is liberated via partial proteolysis during tissue remodeling and cancer growth, thereby representing an additional layer of control for angiogenesis [82]. One possibility is that tumor growth might be enhanced in vivo by a lack of circulating LG3. In line with this, circulating LG3 lev- els are reduced in patients with breast cancer, suggest- ing that reduced titers might be a useful biomarker for cancer progression and invasion [92]. The fragment in circulation might be continuously released to add an for angiogenesis during additional cancer progression. Utilization of the antiangiogenic fragment as either a protein- or peptide-based pharma- cological agent might represent a novel therapeutic rationale, especially when provided in combination with other tumor-suppressive compounds (Table 1).
Collagen XVIII–endostatin: innovative angiogenic regulators
Agrin is strongly expressed around blood vessels [82]. Agrin can be considered to be a marker of tumor angiogenesis in the liver because it is markedly depos- ited in proliferating bile ductules, in newly formed sep- tal vessels in hepatic cirrhosis and in the angiogenic network of malignant hepatocellular carcinomas (HC) [99]. Agrin overexpression in newly formed blood ves- sels is also present in cholangiocarcinoma [100]. The role of agrin in tumor angiogenesis is not yet estab- lished, but initial studies suggest that agrin’s effect on tumor angiogenesis is likely context dependent. It seems that the expression of agrin may be protective against disorganized angiogenesis in glioblastomas, whereas in hepatic malignancies it seems to support tumor angiogenesis, at least in the initial stages of tumor development. There remains very limited knowl- edge of whether C-terminal endorepellin-like fragment of agrin signals through the integrin receptors. Further research on this area will improve our knowledge for possible targeting of agrin in tumors (Table 1).
Cell-surface PGs
type I
(Fig. 6). Syndecans
The cell-surface PGs include two major subfamilies: the syndecans, with four members cloned in mammals, which are transmembrane proteins mostly substituted with HS chains; and glypicans linked with a glycosyl-phosphatidylinositol anchor to the cell mem- brane, with six members cloned in mammals substi- tuted with HS chains and glypicans are generally considered to act as coreceptors for heparin-binding mitogenic growth factors [2].
Glypicans influence tumor development and progression
Collagen XVIII has been suggested to play a negative regulatory, but not essential, role in angiogenesis in certain contexts. Collagen type XVIII harbors the C-terminal antiangiogenic fragment, endostatin, pro- teolytically derived from the C-terminus of collagen type XVIII [82,93]. The expression of collagen XVIII and endostatin has been studied in ovarian, hepato- cellular, pancreatic and oral squamous cell carcinoma and found to vary among different cancer types [94]. Endostatin is a potent antiangiogenic molecule reduc- ing tumor growth, choroidal neovascularization and wound healing [82]. High levels of circulating endosta- tin reduce tumor burden, block the formation of pul- monary metastases [95] and, notably, induce a total gene expression reprogramming [96], which ultimately disrupts endothelial cell migration (Fig. 5) [97]. En- dostatin has been linked to cell-surface receptors including various integrins, glypican and VEGFR2. Specifically, endostatin downregulates several key com- ponents of the VEGF signaling cascade and, at the same time, stimulates the synthesis of thrombospondin, a powerful angiostatic protein and suppresses c-myc (Fig. 5) [82,96]. Endostatin has been studied in phase I and II clinical trials for patients with metastatic cancer and has shown low efficacy, however, a new more sta- ble version of endostatin has re-entered the clinic and is now used in certain countries for the treatment of lung and gastric cancer [94]. Recently, an immunoiso- lation device that contains endostatin-expressing cells was used effectively for the treatment of melanoma and Ehrlich tumors in mice [98]. This suggests that the
Glypicans show cell-type and developmental-stage-spe- cific expression. They are involved in fundamental bio- logical processes such as cell–ECM interactions and the control of cellular division, differentiation and morphogenesis [101]. At the level of signaling, they are involved in the regulation of pathways including Wnt, FGF, Hedgehog (Hh), bone morphogenic protein, Slit and insulin-like [101]. Therefore, growth factor depending on the biological context, glypicans can either stimulate or inhibit signaling activity. Notably, the HS chains are essential for the glypican-induced
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3913
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
Intracellular and cell-surface PGs
Syndecans
Nucleus
Serglycin
GAG chains
Glypicans
its
endocytosis
GPI-anchor
Fig. 6. Localization and classification of cell-surface associated and intracellular PGs. The cell-surface PGs encompass mainly the trans- membrane PGs syndecans and the glycosyl-phosphatidylinositol- anchored glypicans. Serglycin is the only characterized intracellular PG.
stimulation of FGF activity, and partially required for the regulatory activity of glypicans in Hh, Wnt and bone morphogenetic protein signaling [101–104].
Glypicans influence tumor development and progres- sion, and their expression is abnormal in various human tumors (Table 1). For example, glypican-1 is upregu- lated in breast and pancreatic cancer, and it has been postulated that this aberrant expression of glypican-1 may play a key role in promoting growth factor signal- ing in cancer cells [105,106]. By contrast, glypican-3 gene is mutated in patients with Simpson–Golabi– Behmel syndrome, deregulating the balance between cell proliferation and apoptosis [107]. The loss of glypi- can-3 induces overgrowth observed in this syndrome. Patients with Simpson–Golabi–Behmel syndrome are
increased risk of embryonic tumors. Glypican-3 at inhibits the signaling of Hh by competing with Patched, the Hh receptor, for Hh binding [102]. The binding of Hh to the receptor Patched triggers the sig- naling pathway by blocking the inhibitory effect of Patched on Smoothened. Glypican-3 competes with Patched for Hh binding allowing to ligand-free Patched to inhibit Smoothened, reducing signaling and cell growth (Fig. 7). The binding of Hh to glypican-3 triggers and degradation further strengthens the negative regulatory role of glypican-3 in Hh signaling (Fig. 7). Glypican-3 plays a negative role in cell proliferation and induces apoptosis in mesothelioma and breast cancer that is only dependent on the protein core [108]. Glypican-3 expression is downregulated in tumors with different histogenetic backgrounds as a result of hypermethylation of the glypican promoter, and its expression can be restored by treatment with demethylation agents [109–112]. Hh signaling pathway is hyperactivated in a proportion of these cancers [113,114] and it is therefore possible that the loss of glypican-3 may contribute to cancer pro- gression by inducing the activation of Hh signaling similarly to Simpson–Golabi–Behmel syndrome. By contrast, upregulation of glypican-3 is observed in embryonic tumors, such as neuroblastoma and Wilm’s tumor [115]. In tumors originating from tissues that express glypican-3 only in the embryo, such as HC and melanoma, its expression tends to reappear with transformation [101]. The canonical Wnt malignant pathway plays a key role in most HC and overexpres- sion of glypican-3 promotes tumor growth-promoting Wnt signaling. The binding of Wnt to glypican-3 facili- tates and ⁄ or stabilizes the interaction of Wnt with the
Biological roles of glypican 3 and serglycin
Protection, transport, activation and delivery of bioactive molecules
Glypican 3
Hh
Wnt Wnt
Hh Hh
Frizzled
Patched
Smoothened
Patched
X
Hh
Serglycin
Increased signal
Reduced signal
Increased signal
Endocytosis degradation
Bioactive molecules
Regulation of tumor growth
Fig. 7. The binding of Wnt to glypican-3 facilitates and ⁄ or stabilizes the interaction of Wnt and Frizzled with the consequent increament on signaling. The binding of Hh to the receptor Patched triggers the signal- ing pathway by blocking the inhibitory effect of Patched on Smoothened. Glypican-3 competes with Patched for Hh binding allowing ligand-free Patched to inhibit Smoothened, reducing signaling and tumor growth. The binding of Hh to glypican-3 triggers endocytosis and degradation of the complex. Serglycin interacts with and stores bioactive molecules inside storage granules and secretory vesicles. Upon secretion, serglycins modulate their activities through protection, transport, activation and delivery of the bioactive molecules.
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3914
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
receptor Frizzled with the consequent increment on signaling (Fig. 7). HC cells forced to overexpress a sol- uble form of glypican-3 showed lower tumorogenicity because soluble glypican-3 blocked the activity of sev- eral pro-tumorigenic growth factors. Glypican-3 may promote local cancer growth in some cancer tissues, whereas it inhibits tissue invasion and metastasis in others.
It
eradication by T cells of
accelerate
shedding
It is suggested that cell-surface syndecans promote pro- angiogenic signaling by binding FGF-2 and VEGF and presenting them to their high-affinity receptors and also by protecting them from inactivation (Fig. 8) [127,128]. Notably, syndecan-1 also mediates cell adhe- sion in cooperation with integrins by activation of actin and fascin bundling (Fig. 8) [119,124]. By con- trast, syndecan-1 is downregulated in various malig- nancies and a possible inhibitory function is suggested [119,129–131]. Syndecan-1 may serve as a prognostic marker in a cancer-type-specific manner (Table 1). The effects of syndecan-1 on tumor progression and prog- nosis probably also depend upon whether syndecan-1 is tumor cell derived or synthesized by the host stroma, because its expression by reactive tumor stromal cells promotes tumor progression [123,125,126,132–135]. Soluble syndecan-1 ectodomain is present in the serum of various malignancies. A variety of proangiogen- ic ⁄ protumor growth factors and enzymes that are up- regulated in solid tumors in vivo (e.g. FGF-2 and in vitro MMPs) syndecan-1 [136,137], presumably by promoting cleavage of the accelerates ectodomain [138]. Soluble
syndecan-1
Biological roles of syndecans
Soluble syndecans
Tumorigenesis angiogenesis
Glypican-3 is a novel tumor marker for early-stage melanoma [116] and early-stage HC [117]. It is not known, however, whether its elevated serum levels are important in tumor progression or are simply a reflec- tion of aggressive tumors. The high levels of shed glyp- ican may arise from increased expression of the PG or increased activity of tumor proteases. In this light, glypican-3 may be a candidate antigen for cancer strongly immunotherapy in HC (Table 1). is is highly immunogenic expressed exclusively in HC, and stimulates tumors expressing glypican-3 in mice and markedly inhibited growth of an established tumor. Glypican-3-derived peptide-pulsed vaccination is a novel strategy to pre- vent HC and melanoma in patients and needs to be further developed as an anticancer therapy [116]. Glyp- ican-1 on target cells is recognized by some natural cytotoxicity receptors of natural killer cells and the suppression of glypican-1 in pancreatic cancer led to lower cytotoxic effects of natural killer cells [118] (Table 1).
Protection of proteases and growth factors Presentation to receptors Chemotactic gradient Competition with cell surface syndecans
GF GF
GF
Fibronectin
Syndecans
Growth factor receptor
Syndecans: multiple roles in cancer progression and strategies for their targeting
Collagen
α β
GF
GF
Integrins
P
P
Membrane ruffling and stress fiber formation
Growth factor receptor signaling
through which
regulate
events
they
Tumor / endothelial cell adhesion and migration
Tumor / endothelial cell proliferation and motility
Syndecan-1 is present at early stages during develop- ment and on epithelial and cancer cells in adults. Synd- ecan-2 is distributed in mesenchymal tissues, liver and neuronal cells. Syndecan-3 is mainly associated with neural tissues, whereas syndecan-4 is ubiquitously dis- tributed [2]. Syndecans are involved in complex signal- ing cell proliferation, differentiation, adhesion and migration [119]. For the multiple roles of syndecan shedding see the accompanying minireview by Manon-Jensen et al. [120].
Fig. 8. Syndecans interact with matrix molecules in cooperation with integrins, inducing membrane ruffling and stress fiber forma- tion, promoting cell adhesion and migration. Syndecans bind to growth factors presenting them to their high-affinity receptors, acti- vating signaling pathways that promote cell proliferation and motil- ity. Soluble syndecans interact with growth factors protecting them from degradation and creating a promigratory chemotactic gradient. Soluble syndecans present growth factors to their high-affinity receptors, activating signaling pathways that promote cell prolifera- tion and motility. Soluble syndecans bind to matrix molecules, com- petitively inhibiting cell-surface syndecans, and thereby stimulating tumor and endothelial cell migration.
Syndecan-1 is expressed in various human cancers and correlates with tumor recurrence in human pros- it is upregulated in tate cancer [121]. Furthermore, human breast cancer and correlates with poor progno- sis [122]. Syndecan-1 expression by fibroblasts in the tumor-associated stroma appears to be necessary for breast carcinogenesis [123] and may promote tumori- genesis by regulating tumor cell spreading and adhe- sion [124], proliferation and angiogenesis [125,126].
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3915
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
factors,
tumor growth in vivo and increased cell invasion through collagen in vitro [139]. Soluble syndecan-1 may activate proangiogenic ⁄ protumor like FGF-2 [140], also bind to mitogens, effectively creating a chemotactic gradient [141] and ⁄ or interact with pro- teases protecting them from contact with endogenous inhibitors (Fig. 8) [119].
are
tumors
these
endothelial
signaling, including protein kinase Ca and focal adhe- sion kinase activation, to promote focal adhesion for- mation (Fig. 8) [148]. FGF-2 treatment of melanoma cells resulted in reduction in the expression of syndec- an-4, a decrease in cell attachment on fibronectin and promoted cell migration [149]. Syndecan-4 overexpress- ing cells form larger and more dense focal adhesions, correlated with stronger attachment and decreased cell migration [150], whereas lack of syndecan-4 engage- ment promotes an amotile fibroblast phenotype in which focal adhesion kinase and Rho signaling are downregulated and filopodia extended [151] (Table 1). Syndecan-4 is highly expressed in less- aggressive seminomatous testicular tumors. Reduced levels are detected in more-aggressive non-seminoma- tous germ cell tumors and it correlates with the meta- (personal data). static potential of Interestingly, syndecan-4 is found also in the tumor stroma in some testicular tumors and the presence of stromal-associated syndecan-4 correlates with increased neovascularization. These results indicate that a direc- ted homeostasis in syndecan-4 levels supports the opti- mal migration of tumor cells and its expression by stromal cells may facilitate cancer cell growth and angiogenesis, similarly to syndecan-1.
soluble
assembly by
competing
for
Overexpression of syndecan-2 has been found in numerous malignancies, such as Lewis lung carcinoma, ovarian, osteosarcoma, brain tumors, mesothelioma and colon cancer [119,142–144] (Table 1). This PG promotes tumorigenesis in colorectal cancer and Lewis lung carcinoma by regulating cell adhesion, spreading and cytosketetal organization [142,144]. Recently, we found that expression of syndecan-2 in breast cancer cells is regulated by estradiol through the action of estrogen receptor alpha [145]. Syndecan-2 is highly expressed in the microvasculature of mouse glioma tumors and promotes angiogenesis, at least in part, by modulation of endothelial cell adhesion [143]. Exoge- nous addition of EGF, FGF-2, VEGF, MMP-2 and cells MMP-9 to brain microvascular increases syndecan-2 ectodomain shedding [143]. The released ectodomain may act on tumor cells and ⁄ or the endothelium to modulate tumorigenesis. Soluble syndecan-2 ectodomain causes significant increase in endothelial cell membrane protrusion, migration, capil- lary tube formation and cell–cell interactions most probably because of competitive inhibition of cell-sur- face syndecan- 2 (Fig. 8) [119,143]. Colorectal carci- noma cells demonstrate reduced cell adhesion in the syndecan-2 ectodomain [143], presence of whereas overexpression of full-length syndecan-2 in lung carcinoma cells induces stress fiber formation and cytoskeletal structures indicative of strong adhesion; cells also become nonmotile [142]. Cell-surface syndec- an-2 is also needed for the correct organization of lam- inin and fibronectin, and its reduced interaction with matrix ligands because of the presence of soluble ecto- domain would be expected to decrease basement mem- brane laminin and fibronectin binding [146]. Syndecan-2 ectodomain may exert its proangiogenic effect by binding to growth fac- tors and cytokines [119], effectively creating a chemo- tactic gradient similar to that produced by soluble syndecan-1 (Fig. 8) [141].
Syndecan-4 is a focal adhesion component
in a range of cell types and mediates breast cancer cell adhesion and spreading [119]. It also forms a complex with the proangiogenic molecule, FGF-2, and its receptor, fibroblast growth factor receptor (FGFR-1) promoting FGF signaling (Fig. 8) [147]. The attach- ment of syndecan-4 to fibronectin initiates intracellular
The antitumor effect of murine ⁄ human chimeric syndecan-1-specific monoclonal antibody nBT062 con- jugated with highly cytotoxic maytansinoid derivatives against multiple myeloma cells in vitro and in vivo was examined. Treatment significantly inhibited multiple myeloma tumor growth in vivo and prolonged host survival in both the xenograft mouse models of human multiple myeloma and the SCID-hu mouse model. These results provide a preclinical framework support- ing the evaluation of nBT062-maytansinoid derivatives in clinical trials to improve patient outcome in multiple myeloma [152] (Table 1). Another strategy for syndec- an targeting in the treatment of malignancies is the inhibition of heparanase an enzyme that degrades HS (see the accompanying minireview by Barash et al. [153]). Heparanase activity is strongly implicated in structural remodeling of the extracellular matrix, a process which can lead to invasion by tumor cells. In addition, heparanase augments signaling cascades lead- ing to enhanced phosphorylation of selected protein kinases and increased gene transcription associated with aggressive tumor progression. This function is mediated by a novel protein domain localized at the heparanase C-terminus. Heparanase is also an impor- tant regulator of syndecan clustering, shedding and mitogen binding. Recent data indicate that modified glycol-split heparin, as well as other heparanase inhibi- tors, can profoundly inhibit the progression of tumor
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3916
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
4 Wight TN (2002) Versican: a versatile extracellular
xenografts produced by myeloma and carcinoma cells, thus moving antiheparanase therapy closer to reality [154] (Table 1).
Intracellular PGs
matrix proteoglycan in cell biology. Curr Opin Cell Biol 14, 617–623.
5 Wight TN & Merrilees MJ (2004) Proteoglycans in ath- erosclerosis and restenosis: key roles for versican. Circ Res 94, 1158–1167. 6 Theocharis AD (2008) Versican in health and disease. Connect Tissue Res 49, 230–234.
(e.g. histamine,
7 Wu YJ, Lapierre D, Wu J, Yee AJ & Yang BB (2005) The interaction of versican with its binding partners. Cell Res 15, 483–494. 8 Kim S, Takahashi H, Lin WW, Descargues P,
Grivennikov S, Kim Y, Luo JL & Karin M (2009) Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature 457, 102–106.
9 Sakko AJ, Ricciardelli C, Mayne K, Tilley WD, Le- Baron RG & Horsfall DJ (2001) Versican accumula- tion in human prostatic fibroblast cultures is enhanced by prostate cancer cell-derived transforming growth factor beta1. Cancer Res 61, 926–930. 10 Ricciardelli C, Brooks JH, Suwiwat S, Sakko AJ,
Serglycin is the only characterized intracellular PG found in hematopoietic and endothelial cells (Fig. 6). It has important functions related to the formation of sev- eral types of storage granules. Serglycin interacts with several bioactive molecules tumor necrosis factor a and various proteases) and is impor- tant for the retention of these key inflammatory media- tors inside storage granules and secretory vesicles. Serglycin can further modulate the activities of partner molecules in different ways after secretion from acti- vated immune cells, through protection, transport, acti- vation and interactions with substrates or target cells (Fig. 7) [155]. Few reports involve serglycin in malig- nancies. Niemann et al. [156] demonstrated the expres- sion of serglycin in various hematologic malignancies. Serglycin expression was found to distinguish acute myeloid leukemia from acute lymphoblastic leukemia. It was found to be a selective marker for immature myeloid cells, distinguishing acute myeloid leukemia from Philadelphia chromosome-negative chronic myelo- proliferative disorders. The expression and constitutive secretion of serglycin has also been reported in multiple myeloma, where the released serglycin inhibits bone mineralization [157]. The role of serglycin in hemato- logical malignancies remains unkown and further studies will improve our knowledge of its involvement and potential pharmacological targeting (Table 1).
Mayne K, Raymond WA, Seshadri R, LeBaron RG & Horsfall DJ (2002) Regulation of stromal versican expression by breast cancer cells and importance to relapse-free survival in patients with node-negative pri- mary breast cancer. Clin Cancer Res 8, 1054–1060. 11 Nikitovic D, Zafiropoulos A, Katonis P, Tsatsakis A, Theocharis AD, Karamanos NK & Tzanakakis GN (2006) Transforming growth factor-beta as a key mole- cule triggering the expression of versican isoforms v0 and v1, hyaluronan synthase-2 and synthesis of hyal- uronan in malignant osteosarcoma cells. IUBMB Life 58, 47–53. 12 Arslan F, Bosserhoff AK, Nickl-Jockschat T, Doerfelt
Acknowledgement
We thank Prof. R.V. Iozzo for critical reading and valuable advice.
A, Bogdahn U & Hau P (2007) The role of versican iso- forms V0 ⁄ V1 in glioma migration mediated by trans- forming growth factor-beta2. Br J Cancer 96, 1560– 1568. 13 Serra M, Miquel L, Domenzain C, Docampo MJ,
References
Fabra A, Wight TN & Bassols A (2005) V3 versican isoform expression alters the phenotype of melanoma cells and their tumorigenic potential. Int J Cancer 114, 879–886.
1 Iozzo RV (1998) Matrix proteoglycans: from molecular design to cellular function. Annu Rev Biochem 67, 609– 652. 2 Schaefer L & Schaefer RM (2010) Proteoglycans: from
structural compounds to signaling molecules. Cell Tissue Res 339, 237–246.
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3917
3 Syrokou A, Tzanakakis G, Tsegenidis T, Hjerpe A & Karamanos NK (1999) Effects of glycosaminoglycans on proliferation of epithelial and fibroblast human malignant mesothelioma cells: a structure–function relationship. Cell Prolif 32, 85–99. 14 Miquel-Serra L, Serra M, Herna´ ndez D, Domenzain C, Docampo MJ, Rabanal RM, de Torres I, Wight TN, Fabra A & Bassols A (2006) V3 versican isoform expression has a dual role in human melanoma tumor growth and metastasis. Lab Invest 86, 889–901. 15 Ricciardelli C, Mayne K, Sykes PJ, Raymond WA, McCaul K, Marshall VR & Horsfall DJ (1998) Ele- vated levels of versican but not decorin predict disease progression in early stage prostate cancer. Clin Cancer Res 4, 963–971.
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
16 Pirinen R, Leinonen T, Bo¨ hm J, Johansson R, to simultaneously cause apoptotic resistance and sensi- tivity. Cancer Res 67, 4742–4750.
Ropponen K, Kumpulainen E & Kosma VM (2005) Versican in nonsmall cell lung cancer: relation to hyal- uronan, clinicopathologic factors, and prognosis. Hum Pathol 36, 44–50. 17 Pukkila M, Kosunen A, Ropponen K, Virtaniemi J, 27 Yee AJ, Akens M, Yang BL, Finkelstein J, Zheng PS, Deng Z & Yang B (2007) The effect of versican G3 domain on local breast cancer invasiveness and bony metastasis. Breast Cancer Res 9, R47. 28 Touab M, Villena J, Barranco C, Arumi-Uria M &
Bassols A (2002) Versican is differentially expressed in human melanoma and may play a role in tumor devel- opment. Am J Pathol 160, 549–557. 29 Sakko AJ, Ricciardelli C, Mayne K, Suwiwat S,
LeBaron RG, Marshall VR, Tilley WD & Horsfall DJ (2003) Modulation of prostate cancer cell attachment to matrix by versican. Cancer Res 63, 4786–4791. Kellokoski J, Kumpulainen E, Pirinen R, Nuutinen J, Johansson R & Kosma VM (2007) High stromal versi- can expression predicts unfavourable outcome in oral squamous cell carcinoma. J Clin Pathol 60, 267–272. 18 Labropoulou VT, Theocharis AD, Ravazoula P, Peri- menis P, Hjerpe A, Karamanos NK & Kalofonos HP (2006) Versican but not decorin accumulation is related to metastatic potential and neovascularization in testic- ular germ cell tumors. Histopathology 49, 582–593. 19 Suwiwat S, Ricciardelli C, Tammi R, Tammi M,
30 Zhang Y, Cao L, Yang BL & Yang BB (1998) The G3 domain of versican enhances cell proliferation via epi- dermal growth factor-like motifs. J Biol Chem 273, 21342–21351.
Auvinen P, Kosma VM, LeBaron RG, Raymond WA, Tilley WD & Horsfall DJ (2004) Expression of extra- cellular matrix components versican, chondroitin sul- fate, tenascin, and hyaluronan, and their association with disease outcome in node negative breast cancer. Clin Cancer Res 10, 2491–2498. 20 Kodama J, Hasengaowa, Kusumoto T, Seki N,
31 Yang BL, Zhang Y, Cao L & Yang BB (1999) Cell adhesion and proliferation mediated through the G1 domain of versican. J Cell Biochem 72, 210–220. 32 Wang W, Xu G-L, Jia W-D, Ma J-L, Li J-S, Ge Y-S, Ren W-H, Yu J-H & Liu W-B (2009) Ligation of TLR2 by versican: a link between inflammation and metastasis. Arch Med Res 40, 321–323. 33 Naso MF, Zimmermann DR & Iozzo RV (1994) Matsuo T, Nakamura K, Hongo A & Hiramatsu Y (2007) Versican expression in human cervical cancer. Eur J Cancer 43, 1460–1466.
Characterization of the complete genomic structure of the human versican gene and functional analysis of its promoter. J Biol Chem 269, 32999–33008.
21 Kodama J, Hasengaowa, Kusumoto T, Seki N, Matsuo T, Ojima Y, Nakamura K, Hongo A & Hiramatsu Y (2007) Prognostic significance of stromal versican expression in human endometrial cancer. Ann Oncol 18, 269–274. 22 Mauri P, Scarpa A, Nascimbeni AC, Benazzi L,
34 Yoon H, Liyanarachi S, Wright FA, Davuluri R, Lockman JC, Chapelle A & Pellegata NS (2002) Gene expression profiling of isogenic cells with differ- ent TP53 gene dosage reveals numerous genes that are affected by TP53 dosage and identifies CSPG2 as a direct target of p53. Proc Natl Acad Sci USA 99, 15632–15637.
Parmagnani E, Mafficini A, Della Peruta M, Bassi C, Miyazaki K & Sorio C (2005) Identification of proteins released by pancreatic cancer cells by multidimensional protein identification technology: a strategy for identifi- cation of novel cancer markers. FASEB J 19, 1125– 1127. 23 Cattaruzza S, Schiappacassi M, Kimata K, Colombatti 35 Willert J, Epping M, Pollack JR, Brown PO & Nusse R (2002) A transcriptional response to Wnt protein in human embryonic carcinoma cells. BMC Dev Biol 2, 8. doi: 10.1186/1471-213X-2-8. 36 Berdiaki A, Zafiropoulos A, Fthenou E, Katonis P,
A & Perris R (2004) The globular domains of PGM ⁄ versican modulate the proliferation–apoptosis equilibrium and invasive capabilities of tumor cells. FASEB J 18, 779–781. 24 Zheng PS, Wen J, Ang LC, Sheng W, Viloria-Petit A, Tsatsakis A, Karamanos NK & Tzanakakis GN (2008) Regulation of hyaluronan and versican deposition by growth factors in fibrosarcoma cell lines. Biochim Bio- phys Acta 1780, 194–202.
Wang Y, Wu Y, Kerbel RS & Yang BB (2004) Versican ⁄ PG-M G3 domain promotes tumor growth and angiogenesis. FASEB J 18, 754–756.
37 Syrokou A, Tzanakakis GN, Hjerpe A & Karamanos NK (1999) Proteoglycans in human malignant meso- thelioma. Stimulation of their synthesis induced by epi- dermal, insulin and platelet-derived growth factors involves receptors with tyrosine kinase activity. Biochi- mie 81, 733–744. 38 Shimizu-Hirota R, Sasamura H, Mifune M, Nakaya
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3918
H, Kuroda M, Hayashi M & Saruta T (2001) Regula- tion of vascular proteoglycan synthesis by angiotensin 25 Ricciardelli C, Russell DL, Ween MP, Mayne K, Suwiwat S, Byers S, Marshall VR, Tilley WD & Horsfall DJ (2007) Formation of hyaluronan- and versican-rich pericellular matrix by prostate cancer cells promotes cell motility. J Biol Chem 282, 10814–10825. 26 LaPierre DP, Lee DY, Li SZ, Xie YZ, Zhong L, Sheng W, Deng Z & Yang BB (2007) The ability of versican
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
II type 1 and type 2 receptors. J Am Soc Nephrol 12, 2609–2615. BEHAB ⁄ brevican identified in human malignant glio- mas. Cancer Res 65, 6726–6733.
39 Lee DY, Shatseva T, Jeyapalan Z, Du WW, Deng Z & Yang BB (2009) A 3¢-untranslated region (3¢UTR) induces organ adhesion by regulating miR-199a* func- tions. PLoS ONE 4, e4527. doi:10.1371/jour- nal.pone.0004527.
52 Schaefer L & Iozzo RV (2008) Biological functions of the small leucine-rich proteoglycans: from genetics to signal transduction. J Biol Chem 283, 21305–21309. 53 Iozzo RV & Schaefer L (2010) Proteoglycans in health and disease: novel regulatory signaling mechanisms evoked by the small leucine-rich proteoglycans. FEBS J 277, 3864–3875. 54 Iozzo RV, Chakrani F, Perrotti D, McQuillan DJ, 40 Migliore C, Petrelli A, Ghiso E, Corso S, Capparuccia L, Eramo A, Comoglio PM & Giordano S (2008) MicroRNAs impair MET-mediated invasive growth. Cancer Res 68, 10128–10136. 41 Seto AG (2010) The road toward microRNA therapeu- tics. Int J Biochem Cell Biol 42, 1298–1305. 42 Ricciardelli C, Sakko AJ, Ween MP, Russell DL & Skorski T, Calabretta B & Eichstetter I (1999) Cooper- ative action of germline mutations in decorin and p53 accelerates lymphoma tumorigenesis. Proc Natl Acad Sci USA 96, 3092–3097.
Horsfall DJ (2009) The biological role and regulation of versican levels in cancer. Cancer Metastasis Rev 28, 233–245.
55 Csordas G, Santra M, Reed CC, Eichstetter I, McQui- llan DJ, Gross D, Nugent MA, Hajnoczky G & Iozzo RV (2000) Sustained down-regulation of the epidermal growth factor receptor by decorin. A mechanism for controlling tumor growth in vivo. J Biol Chem 275, 32879–32887. 56 Santra M, Mann DM, Mercer EW, Skorski T, 43 Almholt K, Juncker-Jensen A, Laerum OD, Danø K, Johnsen M, Lund LR & Rømer J (2008) Metastasis is strongly reduced by the matrix metalloproteinase inhib- itor Galardin in the MMTV-PymT transgenic breast cancer model. Mol Cancer Ther 7, 2758–2767.
Calabretta B & Iozzo RV (1997) Ectopic expression of decorin protein core causes a generalized growth suppression in neoplastic cells of various histogenetic origin and requires endogenous p21, an inhibitor of cyclin-dependent kinases. J Clin Invest 100, 149–157. 44 Vankemmelbeke MN, Jones GC, Fowles C, Ilic MZ, Handley CJ, Day AJ, Knight CG, Mort JS & Buttle DJ (2003) Selective inhibition of ADAMTS-1, -4 and -5 by catechin gallate esters. Eur J Biochem 270, 2394– 2403.
45 Zeng C, Toole BP, Kinney SD, Kuo JW & Stamenko- vic I (1998) Inhibition of tumor growth in vivo by hyal- uronan oligomers. Int J Cancer 77, 396–401. 46 Misra S, Toole BP & Ghatak S (2006) Hyaluronan 57 Santra M, Eichstetter I & Iozzo RV (2000) An anti- oncogenic role for decorin: downregulation of ErbB2 leads to growth suppression and cytodifferentiation of mammary carcinoma cells. J Biol Chem 275, 35153– 35161.
constitutively regulates activation of multiple receptor tyrosine kinases in epithelial and carcinoma cells. J Biol Chem 281, 34936–34941. 47 Skandalis SS, Theocharis AD, Vynios DH,
58 Gialeli Ch, Kletsas D, Mavroudis D, Kalofonos HP, Tzanakakis GN & Karamanos NK (2009) Targeting epidermal growth factor receptor in solid tumors: criti- cal evaluation of the biological importance of therapeu- tic monoclonal antibodies. Curr Med Chem 16, 3797– 3804.
Papageorgakopoulou N, Hjerpe A, Karamanos NK & Theocharis DA (2006) Cartilage aggrecan undergoes significant compositional and structural alterations during laryngeal cancer. Biochim Biophys Acta 1760, 1046–1055.
59 Zhu J-X, Goldoni S, Bix G, Owens RA, McQuillan D, Reed CC & Iozzo RV (2005) Decorin evokes pro- tracted internalization and degradation of the EGF receptor via caveolar endocytosis. J Biol Chem 280, 32468–32479.
48 Skandalis SS, Theocharis AD, Theocharis DA, Papa- das T, Vynios DH & Papageorgakopoulou N (2004) Matrix proteoglycans are markedly affected in advanced laryngeal squamous cell carcinoma. Biochim Biophys Acta 1689, 152–161.
60 Reed CC, Waterhouse A, Kirby S, Kay P, Owens RA, McQuillan DJ & Iozzo RV (2005) Decorin prevents metastatic spreading of breast cancer. Oncogene 24, 1104–1110.
49 Nutt CL, Zerillo CA, Kelly GM & Hockfield S (2001) Brain enriched hyaluronan binding (BEHAB) ⁄ brevican increases aggressiveness of CNS-1 gliomas in Lewis rats. Cancer Res 61, 7056–7059.
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3919
61 Zafiropoulos A, Nikitovic D, Katonis P, Tsatsakis A, Karamanos NK & Tzanakakis GN (2008) Decorin- induced growth inhibition is overcome through pro- tracted expression and activation of epidermal growth factor receptors in osteosarcoma cells. Mol Cancer Res 6, 785–794. 50 Hu B, Kong LL, Matthews RT & Viapiano MS (2008) The proteoglycan brevican binds to fibronectin after proteolytic cleavage and promotes glioma cell motility. J Biol Chem 283, 24848–24859. 51 Viapiano MS, Bi WL, Piepmeier J, Hockfield S & 62 Goldoni S, Humphries A, Nystro¨ m A, Sattar S, Owens RT, McQuillan DJ, Ireton K & Iozzo RV (2009) Matthews RT (2005) Novel tumor-specific isoforms of
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
Decorin is a novel antagonistic ligand of the Met receptor. J Cell Biol 185, 743–754. distant killing of cancer cells using adenovirus mediated decorin gene transfer. FASEB J 17, 464–466.
74 Biglari A, Bataille D, Naumann U, Weller M, Zirger J, Castro MG & Lowenstein PR (2004) Effects of ectopic decorin in modulating intracranial glioma progression in vivo, in a rat syngeneic model. Cancer Gene Ther 11, 721–732. 63 Newton TR, Parsons PG, Lincoln DJ, Cummings MC, Wyld DK, Webb PM, Green AC & Boyle GM (2006) Expression profiling correlates with treatment response in women with advanced serous epithelial ovarian can- cer. Int J Cancer 119, 875–883. 64 Troup S, Njue C, Kliewer EV, Parisien M, Roskelley
75 Goldoni S & Iozzo RV (2008) Tumor microenviron- ment: modulation by decorin and related molecules harboring leucine-rich tandem motifs. Int J Cancer 123, 2473–2479.
C, Chakravarti S, Roughley PJ, Murphy LC & Watson PH (2003) Reduced expression of the small leucine-rich proteoglycans, lumican, and decorin is associated with poor outcome in node-negative invasive breast cancer. Clin Cancer Res 9, 207–214.
76 Goldoni S, Seidler DG, Heath J, Fassan M, Baffa R, Thakur ML, Owens RA, McQuillan DJ & Iozzo RV (2008) An anti-metastatic role for decorin in breast cancer. Am J Pathol 173, 844–855.
65 Matsumine A, Shintani K, Kusuzaki K, Matsubara T, Satonaka H, Wakabayashi T, Iino T & Uchida A (2007) Expression of decorin, a small leucine-rich pro- teoglycan, as a prognostic factor in soft tissue tumors. J Surg Oncol 96, 411–418.
77 Li Y, Aoki T, Mori Y, Ahmad M, Miyamori H, Taki- no T & Sato H (2004) Cleavage of lumican by mem- brane-type matrix metalloprotease-1 abrogates this proteoglycan-mediated suppression of tumor cell col- ony formation in soft agar. Cancer Res 64, 7058–7064.
66 Theocharis AD (2002) Human colon adenocarcinoma is associated with specific post-translational modifica- tions of versican and decorin. Biochim Biophys Acta 1588, 165–172.
78 Nikitovic D, Berdiaki A, Zafiropoulos A, Katonis P, Tsatsakis A, Karamanos N & Tzanakakis GN (2008) Lumican expression is positively correlated with the differentiation and negatively with the growth of human osteosarcoma cells. FEBS J 275, 350–361. 67 Nash MA, Deavers MT & Freedman RS (2002) The expression of decorin in human ovarian tumors. Clin Cancer Res 8, 1754–1760.
79 Vuillermoz B, Khoruzhenko A, D’Onofrio MF, Ramont L, Venteo L, Perreau C, Antonicelli F, Maquart FX & Wegrowski Y (2004) The small leucine- rich proteoglycan lumican inhibits melanoma progres- sion. Exp Cell Res 296, 294–306. 68 Theocharis AD, Vynios DH, Papageorgakopoulou N, Skandalis SS & Theocharis DA (2003) The gastric carcinoma glycosaminoglycans and proteoglycans undergo marked alterations in their content, composi- tion and chemical structure. Int J Biochem Cell Biol 35, 376–390.
69 Skandalis SS, Kletsas D, Kyriakopoulou D, Stavropo- ulos M & Theocharis DA (2006) The greatly increased amounts of accumulated versican and decorin with spe- cific post-translational modifications may be closely associated with the malignant phenotype of pancreatic cancer. Biochim Biophys Acta 1760, 1217–1225.
80 Leygue E, Snell L, Dotzlaw H, Troup S, Hiller-Hitch- cock T, Murphy LC, Roughley PJ & Watson PH (2000) Lumican and decorin are differentially expressed in human breast carcinoma. J Pathol 192, 313–320. 81 Zeltz C, Brezillon S, Perreau C, Ramont L, Maquart F-X & Wegrowski Y (2009) Lumcorin: a leucine-rich repeat 9-derived peptide from human lumican inhibit- ing melanoma cell migration. FEBS Lett 583, 3027– 3032. 82 Iozzo RV, Zoeller JJ & Nystro¨ m A (2009) Basement
70 Ko¨ ninger J, Giese T, di Mola FF, Wente MN, Esposi- to I, Bachem MG, Giese NA, Bu¨ chler MW & Friess H (2004) Pancreatic tumor cells influence the composition of the extracellular matrix. Biochem Biophys Res Commun 322, 943–949. membrane proteoglycans: modulators par excellence of cancer growth and angiogenesis. Mol Cells 27, 503– 513. 71 Nash MA, Loercher AE & Freedman RS (1999)
In vitro growth inhibition of ovarian cancer cells by decorin: synergism of action between decorin and carboplatin. Cancer Res 59, 6192–6196.
83 Zhou Z, Wang J, Cao R, Morita H, Soininen R, Chan KM, Liu B, Cao Y & Tryggvason K (2004) Impaired angiogenesis, delayed wound healing and retarded tumor growth in perlecan heparan sulfate-deficient mice. Cancer Res 64, 4699–4702. 84 Sharma B, Handler M, Eichstetter I, Whitelock J,
72 Koninger J, Giese NA, di Mola FF, Berberat P, Giese T, Esposito I, Bachem MG, Buchler MW & Friess H (2004) Overexpressed decorin in pancreatic cancer: potential tumor growth inhibition and attenuation of chemotherapeutic action. Clin Cancer Res 10, 4776– 4783. Nugent MA & Iozzo RV (1998) Antisense targeting of perlecan blocks tumor growth and angiogenesis in vivo. J Clin Invest 102, 1599–1608. 85 Kadenhe-Chiweshe A, Papa J, McCrudden KW, 73 Tralhao JG, Schaefer L, Micegova M, Evaristo C,
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3920
Frischer J, Bae J-O, Huang J, Fisher J, Lefkowitch JH, Feirt N, Rudge J et al. (2008) Sustained VEGF Schonherr E, Kayal S, Veiga-Fernandes H, Danel C, Iozzo RV, Kresse H et al. (2003) In vivo selective and
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
blockade results in microenvironmental sequestration of VEGF by tumors and persistent VEGF receptor-2 activation. Mol Cancer Res 6, 1–9. Ding Y-H, Lo K-M et al. (2001) Oligomerization- dependent regulation of motility and morphogenesis by the collagen XVIII NC1 ⁄ endostatin domain. J Cell Biol 152, 1233–1246.
86 Reiland J, Sanderson RD, Waguespack M, Barker SA, Long R, Carson DD & Marchetti D (2004) Heparan- ase degrades syndecan-1 and perlecan heparan sulfate: functional implications for tumor cell invasion. J Biol Chem 279, 8047–8055. 98 Rodrigues DB, Chammas R, Malavasi NV, daCosta PLN, Chura-Chambi RM, Balduino KN & Morganti L (2010) Anti-tumor therapy with macroencapsulated endostatin producer cells. BMC Biotechnol 10, 19. 99 Ta´ trai P, Duda´ s J, Batmunkh E, Ma´ the´ M, Zalatnai
A, Schaff Z, Ramadori G & Kovalszky I (2006) Agrin, a novel basement membrane component in human rat and liver, accumulates in cirrhosis and hepatocellular carcinoma. Lab Invest 86, 1149–1160. 87 Whitelock JM, Murdoch AD, Iozzo RV & Underwood PA (1996) The degradation of human endothelial cell- derived perlecan and release of bound basic fibroblast growth factor by stromelysin, collagenase, plasmin and heparanases. J Biol Chem 271, 10079–10086. 100 Batmunkh E, Ta´ trai P, Szabo´ E, Lo´ di C, Holczbauer
A, Pa´ ska C, Kupcsulik P, Kiss A, Schaff Z & Kov- alszky I (2007) Comparison of the expression of agrin, a basement membrane heparan sulfate proteoglycan, in cholangiocarcinoma and hepatocellular carcinoma. Hum Pathol 38, 1508–1515. 101 Filmus J, Capurro M & Rast J (2008) Glypicans. Genome Biol 9, 224. 102 Capurro MI, Xu P, Shi W, Li F, Jia A & Filmus 88 Bix G, Fu J, Gonzalez E, Macro L, Barker A, Camp- bell S, Zutter MM, Santoro SA, Kim JK, Ho¨ o¨ k M et al. (2004) Endorepellin causes endothelial cell disas- sembly of actin cytoskeleton and focal adhesions through the a2b1 integrin. J Cell Biol 166, 97–109. 89 Nystro¨ m A, Shaik ZP, Gullberg D, Krieg T, Eckes B, Zent R, Pozzi A & Iozzo RV (2009) Role of tyrosine phosphatase SHP-1 in the mechanism of endorepellin angiostatic activity. Blood 114, 4897–4906.
J (2008) Glypican-3 inhibits hedgehog signaling during development by competing with Patched for Hedgehog binding. Dev Cell 14, 700–711.
90 Bix G, Castello R, Burrows M, Zoeller JJ, Weech M, Iozzo RA, Cardi C, Thakur MT, Barker CA, Camp- hausen KC et al. (2006) Endorepellin in vivo: targeting the tumor vasculature and retarding cancer growth and metabolism. J Natl Cancer Inst 98, 1634–1646. 91 Gonzalez EM, Reed CC, Bix G, Fu J, Zhang Y, 103 Song HH, Shi W, Xiang Y & Filmus J (2005) The loss of glypican-3 induces alterations in Wnt signaling. J Biol Chem 280, 2116–2125. 104 Kirkpatrick CA, Knox SM, Staatz WD, Fox B,
Gopalakrishnan B, Greenspan DS & Iozzo RV (2005) BMP-1 ⁄ Tolloid-like metalloproteases process endore- pellin, the angiostatic C-terminal fragment of perlecan. J Biol Chem 280, 7080–7087.
92 Chang JW, Kang U-B, Kim DH, Yi JK, Lee JW, Noh D-Y, Lee C & Yu M-H (2008) Identification of circu- lating endorepellin LG3 fragment: potential use as a serological biomarker for breast cancer. Proteomics Clin Appl 2, 23–32. Lercher DM & Selleck SB (2006) The function of a Drosophila glypican does not depend entirely on hepa- ran sulfate modification. Dev Biol 300, 570–582. 105 Kleeff J, Ishiwata T, Kumbasar A, Friess H, Buchler MW, Lander AD & Korc M (1998) The cell-surface heparan sulfate proteoglycan glypican-1 regulates growth factor action in pancreatic carcinoma cells and is overexpressed in human pancreatic cancer. J Clin Invest 102, 1662–1673.
93 Ferreras M, Felbor U, Lenhard T, Olsen BR & Delaisse J (2000) Generation and degradation of human endostatin proteins by various proteinases. FEBS Lett 486, 247–251. 94 Sund M & Kalluri R (2009) Tumor stroma derived
biomarkers in cancer. Cancer Metastasis Rev 28, 177– 183. 106 Matsuda K, Maruyama H, Guo F, Kleeff J, Itakura J, Matsumoto Y, Lander AD & Korc M (2001) Glypi- can-1 is overexpressed in human breast cancer and modulates the mitogenic effects of multiple heparin- binding growth factors in breast cancer cells. Cancer Res 61, 5562–5569.
107 Pilia G, Hughes-Benzie RM, MacKenzie A, Baybayan P, Chen EY, Huber R, Neri G, Cao A, Forabosco A & Schlessinger D (1996) Mutations in GPC3, a glypi- can gene, cause the Simpson–Golabi–Behmel over- growth syndrome. Nat Genet 12, 241–247. 95 Sauter BV, Martinet O, Zhang W-J, Mandeli J & Woo SLC (2001) Adenovirus-mediated gene transfer of endostatin in vivo results in high level of transgene expression and inhibition of tumor growth and metas- tasis. Proc Natl Acad Sci USA 97, 4802–4807. 108 Gonzalez AD, Kaya M, Shi W, Song H, Testa JR,
96 Abdollahi A, Hahnfeldt P, Maercker C, Gro¨ ne H-J, Debus J, Ansorge W, Folkman J, Hlatky L & Huber PE (2004) Endostatin’s antioangiogenic signaling net- work. Mol Cell 13, 649–663.
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3921
Penn LZ & Filmus J (1998) OCI-5 ⁄ GPC3, a glypican encoded by a gene that is mutated in the Simpson– Golabi–Behmel overgrowth syndrome, induces apoptosis in a cell line-specific manner. J Cell Biol 141, 1407–1414. 97 Kuo CJ, LaMontagne KR, Garcia-Cardena G, Ackley BD, Kalman D, Park S, Christofferson R, Kamihara J,
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
expression in locally invasive and metastatic prostate cancer. Urology 63, 402–407. 122 Barbareschi M, Maisonneuve P, Aldovini D, Cangi
109 Kim H, Xu GL, Borczuk AC, Busch S, Filmus J, Capurro M, Brody JS, Lange J, D’Armiento JM, Rothman PB et al. (2003) The heparan sulfate proteo- glycan GPC3 is a potential lung tumor suppressor. Am J Respir Cell Mol Biol 29, 694–701. 110 Zhu Z, Friess H, Kleeff J, Wang L, Wirtz M,
MG, Pecciarini L, Mauri F, Veronese S, Caffo O, Lucenti A, Palma PD et al. (2003) High syndecan-1 expression in breast carcinoma is related to an aggres- sive phenotype and to poorer prognosis. Cancer 98, 474–483. 123 Alexander CM, Reichsman F, Hinkes MT, Lincecum Zimmermann A, Korc M & Buchler MW (2002) Glypican-3 expression is markedly decreased in human gastric cancer but not in esophageal cancer. Am J Surg 184, 78–83. 111 Lin H, Huber R, Schlessinger D & Morin PJ (1999)
J, Becker KA, Cumberledge S & Bernfield M (2000) Syndecan-1 is required for Wnt-1-induced mammary tumorigenesis in mice. Nat Genet 25, 329–332. 124 Beauvais DM & Rapraeger AC (2003) Syndecan-1- Frequent silencing of the GPC3 gene in ovarian cancer cell lines. Cancer Res 59, 807–810.
mediated cell spreading requires signaling by alphavbe- ta3 integrins in human breast carcinoma cells. Exp Cell Res 286, 219–232.
112 Murthy SS, Shen T, De Rienzo A, Lee WC, Ferriola PC, Jhanwar SC, Mossman BT, Filmus J & Testa JR (2000) Expression of GPC3, an X-linked recessive over- growth gene, is silenced in malignant mesothelioma. Oncogene 19, 410–416. 113 Yuan Z, Goetz JA, Singh S, Ogden SK, Petty SK, 125 Maeda T, Alexander CM & Friedl A (2004) Induction of syndecan-1 expression in stromal fibroblasts pro- motes proliferation of human breast cancer cells. Cancer Res 64, 612–621. 126 Maeda T, Desouky J & Friedl A (2006) Syndecan-1
Black CC, Memoli VA, Dmitrovsky E & Robbins DJ (2006) Frequent requirement of hedgehog signaling in non-small cell lung carcinoma. Oncogene 26, 1046– 1055. 114 Kubo M, Nakamura M, Tasaki A, Yamanaka N, expression by stromal fibroblasts promotes breast carci- noma growth in vivo and stimulates tumor angiogene- sis. Oncogene 2, 1408–1412.
Nakashima H, Nomura M, Kuroki S & Katano M (2004) Hedgehog signaling pathway is a new therapeu- tic target for patients with breast cancer. Cancer Res 64, 6071–6074. 127 Chang Z, Meyer K, Rapraeger AC & Friedl A (2000) Differential ability of heparan sulfate proteoglycans to assemble the fibroblast growth factor receptor complex in situ. FASEB J 14, 137–144. 115 Saikali Z & Sinnett D (2000) Expression of glypican 3
(GPC3) in embryonal tumors. Int J Cancer 89, 418– 422. 128 Gospodarowicz D & Cheng J (1986) Heparin protects basic and acidic FGF from inactivation. J Cell Physiol 128, 475–484. 116 Nakatsura T, Kageshita T, Ito S, Wakamatsu K, 129 Anttonen A, Heikkila P, Kajant IM, Jalkanen M &
Joensuu H (2001) High syndecan-1 expression is associ- ated with favourable outcome in squamous cell lung carcinoma treated with radical surgery. Lung Cancer 32, 297–305. 130 Lundin M, Nordling S, Lundin J, Wiksten J-P &
Haglund C (2005) Epithelial syndecan-1 expression is associated with stage and grade in colorectal cancer. Oncology 68, 306–313. Monji M, Ikuta Y, Senju S, Ono T & Nishimura Y (2004) Identification of glypican-3 as a novel tumor marker for melanoma. Clin Cancer Res 10, 6612–6621. 117 Hippo Y, Watanabe K, Watanabe A, Midorikawa Y, Yamamoto S, Ihara S, Tokita S, Iwanari H, Ito Y, Nakano K et al. (2004) Identification of soluble NH2- terminal fragment of glypican-3 as a serological marker for early-stage hepatocellular carcinoma. Cancer Res 64, 2418–2423.
131 Shinyo Y, Kodama J, Hasengaowa, Kusumoto T & Hiramatsu Y (2005) Loss of cell-surface heparan sulfate expression in both cervical intraepithelial neoplasm and invasive cervical cancer. Gynecol Oncol 96, 776–783. 132 Juuti A, Nordling S, Lundin J, Louhimo J & Haglund 118 Bloushtain N, Qimron U, Bar-Ilan A, Hershkovitz O, Gazit R, Fima E, Korc M, Vlodavsky I, Bovin NV & Porgador A (2004) Membrane-associated heparan sul- fate proteoglycans are involved in the recognition of cellular targets by NKp30 and NKp46. J Immunol 173, 2392–2401.
C (2005) Syndecan-1 expression – a novel prognostic marker in pancreatic cancer. Oncology 68, 97–106. 133 Leivonen M, Lundin J, Nordling S, von Boguslawski 119 Fears CY & Woods A (2006) The role of syndecans in disease and wound healing. Matrix Biol 25, 443– 456. 120 Manon-Jensen T, Itoh Y & Couchman JR (2010) K & Haglund C (2004) Prognostic value of syndecan-1 expression in breast cancer. Oncology 67, 11–18.
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3922
Proteoglycans in health and disease: the multiple roles of syndecan shedding. FEBS J 277, 3876–3889. 121 Chen D, Adenekan B, Chen L, Vaughan ED, Gerald W, Feng Z & Knudsen BS (2004) Syndecan-1 134 Mukunyadzi P, Liu K, Hanna EY, Suen JY & Fan CY (2003) Induced expression of syndecan-1 in the stroma of head and neck squamous cell carcinoma. Mod Pathol 16, 796–801.
A. D. Theocharis et al.
Proteoglycans as potential pharmacological targets
135 Wiksten JP, Lundin J, Nordling S, Lundin M,
146 Klass CM, Couchman JR & Woods A (2000) Control of extracellular matrix assembly by syndecan-2 proteo- glycan. J Cell Sci 113, 493–506.
Kokkola A, von Boguslawski K & Haglund C (2001) Epithelial and stromal syndecan-1 expression as predic- tor of outcome in patients with gastric cancer. Int J Cancer 95, 1–6.
147 Mundhenke C, Meyer K, Drew S & Friedl A (2002) Heparan sulfate proteoglycans as regulators of fibro- blast growth factor-2 receptor binding in breast carci- nomas. Am J Pathol 160, 185–194. 148 Woods A, Longley RL, Tumova S & Couchman JR
136 Ding K, Lopez-Burks M, Sa´ nchez-Duran JA, Korc M & Lander AD (2005) Growth factor-induced shedding of syndecan-1 confers glypican-1 dependence on mito- genic responses of cancer cells. J Cell Biol 171, 729– 738.
(2000) Syndecan-4 binding to the high affinity heparin- binding domain of fibronectin drives focal adhesion formation in fibroblasts. Arch Biochem Biophys 374, 66–72. 149 Chalkiadaki G, Nikitovic D, Berdiaki A, Sifaki M,
137 Fitzgerald ML, Wang Z, Park PW, Murphy G & Bernfield M (2000) Shedding of syndecan-1 and -4 ectodomains is regulated by multiple signaling pathways and mediated by a TIMP-3-sensitive metallo- proteinase. J Cell Biol 148, 811–824. 138 Wang Z, Gotte M, Bernfield M & Reizes O (2005)
Krasagakis K, Katonis P, Karamanos NK & Tzanaka- kis GN (2009) Fibroblast growth factor-2 modulates melanoma adhesion and migration through a syndec- an-4-dependent mechanism. Int J Biochem Cell Biol 41, 1323–1331. 150 Longley RL, Woods A, Fleetwood A, Cowling GJ, Constitutive and accelerated shedding of murine synd- ecan-1 is mediated by cleavage of its core protein at a specific juxtamembrane site. Biochemistry 44, 12355– 12361.
Gallagher JT & Couchman JR (1999) Control of mor- phology, cytoskeleton and migration by syndecan 4. J Cell Sci 112, 3421–3431. 151 Midwood KS, Mao Y, Hsia HC, Valenick LV & 139 Yang Y, Yaccoby S, Liu W, Langford JK, Pumphrey CY, Theus A, Epstein J & Sanderson RD (2002) Solu- ble syndecan-1 promotes growth of myeloma tumors in vivo. Blood 100, 610–617. 140 Kato M, Wang H, Kainulainen V, Fitzgerald ML, Schwarzbauer JE (2006) Modulation of cell–fibronectin matrix interactions during tissue repair. J Investig Dermatol Symp Proc 11, 73–78.
Ledbetter S, Ornitz DM & Bernfield M (1998) Physio- logical degradation converts the soluble syndecan-1 ectodomain from an inhibitor to a potent activator of FGF-2. Nat Med 4, 691–697.
152 Ikeda H, Hideshima T, Fulciniti M, Lutz RJ, Yasui H, Okawa Y, Kiziltepe T, Vallet S, Pozzi S, Santo L et al. (2009) The monoclonal antibody nBT062 conjugated to cytotoxic Maytansinoids has selective cytotoxicity against CD138-positive multiple myeloma cells in vitro and in vivo. Clin Cancer Res 15, 4028–4037. 153 Barash U, Cohen-Kapaln V, Dowek I, Sanderson RD,
141 Marshall LJ, Ramdin LS, Brooks T, DPhil PC & Shute JK (2003) Plasminogen activator inhibitor-1 supports IL-8-mediated neutrophil transendothelialmigration by inhibition of the constitutive shedding of endothelial IL-8 ⁄ heparan sulfate ⁄ syndecan-1 complexes. J Immunol 171, 2057–2065.
Ilan N & Vlodavsky I (2010) Proteoglycans in health and disease: new concpts for heparanase function in tumor progression and metastasis. FEBS J 277, 3890– 3903.
154 Fux L, Ilan N, Sanderson RD & Vlodavsky I (2009) Heparanase: busy at the cell surface. Trends Biochem Sci 34, 511–519. 142 Munesue S, Kusano Y, Oguri K, Itano N, Yoshitomi Y, Nakanishi H, Yamashina I & Okayama M (2002) The role of syndecan-2 in regulation of actin-cytoskele- tal organization of Lewis lung carcinoma-derived meta- static clones. Biochem J 363, 201–209. 155 Kolset SO & Tveit H (2008) Serglycin – structure and 143 Fears CY, Gladson CL & Woods A (2006) Syndecan-2 biology. Cell Mol Life Sci 65, 1073–1085. 156 Niemann CU, Kjeldsen L, Ralfkiaer E, Jensen MK & is expressed in the microvasculature of mouse gliomas and regulates angiogenesis in microvascular endothelial cells. J Biol Chem 281, 14533–14536. 144 Park H, Kim Y, Lim Y, Han I & Oh ES (2002) Borregaard N (2007) Serglycin proteoglycan in hemato- logic malignancies: a marker of acute myeloid leuke- mia. Leukemia 21, 2406–2410.
Syndecan-2 mediates adhesion and proliferation of colon carcinoma cells. J Biol Chem 277, 29730–29736. 145 Kousidou OCh, Berdiaki A, Kletsas D, Zafiropoulos
FEBS Journal 277 (2010) 3904–3923 ª 2010 The Authors Journal compilation ª 2010 FEBS
3923
157 Theocharis AD, Seidel C, Borset M, Dobra K, Baykov V, Labropoulou V, Kanakis I, Dalas E, Karamanos NK, Sundan A et al. (2006) Serglycin constitutively secreted by myeloma plasma cells is a potent inhibitor of bone mineralization in vitro. J Biol Chem 281, 35116–35128. A, Theocharis AD, Tzanakakis GN & Karamanos NK (2008) Estradiol-estrogen receptor: a key interplay of the expression of syndecan-2 and metalloproteinase-9 in breast cancer cells. Mol Oncol 2, 223–232.