Abstract
Heparanase (HPSE) is a multitasking protein characterized by enzymatic and non-enzymatic activities. By means of its enzymatic activity, HPSE catalyzes the cutting of the side chains of heparan sulfate (HS) proteoglycans, thereby inducing the remodeling of the extracellular matrix and basement membranes. Thanks to the cleavage of HS, HPSE also promotes the release and diffusion of several HS-linked molecules such as growth factors, cytokines and enzymes. In addition to degrading HS chains, HPSE has non-enzymatic functions that trigger several signaling pathways. This signaling activity is achieved by interacting with transmembrane proteins, activating kinases such as Akt and Src, or modulating the activity of factors such as FGF-2 and TGF-β. Several studies have recently highlighted a possible intracellular activity for HPSE, particularly at nuclear level. While HPSE activity is quite limited in physiological conditions, its demonstrated increasing involvement in various pathological conditions, such as in tumor progression and renal disease, have attracted the attention of a growing number of researchers. The fact that no other molecule is capable of performing the same function as HPSE makes this enzyme an attractive potential target of medical treatment. With this short conceptual overview, we aim to provide an update on current knowledge concerning the HPSE protein in the experimental and clinical settings, paying particular attention to its role in fibrosis, inflammation and cancer.
Introduction
The extracellular matrix (ECM) consists of a fibrous component (mainly collagen fibers and elastin) and an amorphous substance composed of proteoglycans and glycosaminoglycans (GAGs). GAGs are long linear chains of polysaccharides formed by disaccharide units, generally an amino sugar tied to a uronic acid, repeated many times. Due to their many negative charges, GAGs bind positively charged ions that, in turn, trap water molecules, forming hydrated gels and thereby providing mechanical and metabolic support for the ECM. When GAGs are synthesized from sites in the core of proteins, they give rise to proteoglycans. Important components of the ECM are the heparan sulfate proteoglycans (HSPGs), which are bound to the cell surface in the form of syndecans, glypicans and β-glycans, or occur in the basement membranes in the form of agrin, collagen type XVIII and perlecan (1, 2). By binding to other macromolecules such as fibronectin, laminin and collagen (type I and IV), HSPGs contribute to the structural integrity of the ECM and basement membranes, and they modulate interactions between cells and the matrix. Biochemically, via the domains of sulfated disaccharides, the HSPGs provide numerous docking sites for bioactive molecules such as cytokines, growth factors, enzymes, and/or their inhibitors. In this way, they interfere with the formation of receptor-ligand complexes or constitute a reserve of biofactors at ECM level (3). The enzymatic cleavage of the side chains of HSPGs thus results in ECM remodeling, as well as in the release of these bioactive mediators, producing a rapid tissue response to local or systemic stimuli. The enzymatic activity responsible for cutting the side chains of HS was identified and named heparanase (HPSE). It is the only functional endo-glycosidase capable of cleaving heparan sulfate (HS) chains (4). Several review articles have been written on HPSE and its crucial role in the etiology of certain diseases. The purpose of this review is to provide an update on current experimental and clinical knowledge of this enzyme, with particular emphasis on its role in fibrosis, inflammation and cancer.
Processing, localization, enzymatic and non-enzymatic activities of heparanase
Mammalian HPSE is the only endoglycosidase capable of cleaving HS chains of HSPGs. It exerts its enzymatic activity by catalyzing the cleavage of the β(1,4)-glycoside bond between glucuronic acid and glucosamine residues, generating fragments of about 5–7 kDa. Human HPSE gene (HPSE1) encodes a polypeptide of 543 amino acids and 68 kDa that is synthesized in the endoplasmic reticulum. Cutting of the N-terminal signal peptide gives rise to the inactive latent HPSE precursor of 65 kDa (pro-HPSE), which is readily secreted (5). Activation of the precursor into the mature enzyme demands the reuptake of the latent protein and intracellular proteolytic processing. Endocytosis is mediated mainly by syndecans and requires the syndecan-interacting proteins syntenin and α-actinin (6). Once inside the lysosome, the precursor undergoes the cleavage of a linker region (by cathepsin-L), giving rise to 8- and 50-kDa subunits that form the mature dimeric enzyme (7). Within the lysosome, HPSE is implicated in HSPGs turnover. Outside the cell, HPSE degrades cell surface HS and matrix and promotes ECM remodeling and the release of HS-linked molecules, including growth factors and cytokines. Apart from its primary perinuclear localization within lysosomes and late endosomes, several studies have found mature HPSE in the nucleus during in vitro cancer cell differentiation (8, 9). It emerged from subsequent studies that, after lysosomal permeabilization, HPSE is released in the cytoplasm and shuttled into the nucleus (probably by heat shock protein 90), where it is involved in nuclear HS degradation and in regulating gene expression (10, 11). HPSE induces the expression of genes implicated in glucose metabolism and inflammation in fatty acid-treated endothelial cells (12) (Figure 1). In addition to degrading HS chains, HPSE also has non-enzymatic and HS-independent functions. Evidence has been found of cell-membrane receptor(s) by means of which both the 65-kDa precursor and the activated form of HPSE activate signaling pathways and regulate several cellular processes. Migration and invasion are induced in endothelial cells overexpressing HPSE or exposed to latent HPSE precursors by the induction of the PI3K/Akt pathway (13, 14). Recent characterizations of Akt activation by HPSE have revealed that it involves Ras and is integrin-dependent (15). In a model of occlusive vascular disease it was shown that angiogenesis is promoted by HPSE secreted from mesenchymal stem cells via the integrin β1/HIF2 pathway (16). The adhesion of lymphoma and T-cells to ECM and endothelium was found to be mediated by HPSE in an integrin-dependent manner (17, 18). Angiogenesis via VEGF upregulation is promoted by Src activation in various cell lines overexpressing HPSE (19). No HPSE receptors have been identified to date. It seems that a receptor resident in the lipid raft is recruited by HPSE to induce Akt phosphorylation (20), and that signaling activation is mediated by the C-terminus domain (21).
![Figure 1: HPSE: processing, localization, enzymatic and non-enzymatic activities.HPSE is synthesized in the endoplasmic reticulum as a latent precursor (pro-HPSE). After moving to the Golgi apparatus, pro-HPSE is secreted outside the cell [1]. HPSE uptake [mediated by syndecans, mannose-6-phosphate-receptor and low-density lipoprotein receptor (LDLR)-related protein] [2], and its delivery to lysosome enables the proteolytic processing (by cathepsin-L) and activation of the precursor into the dimeric active enzyme [3]. The interaction of latent HPSE with HPSE-binding proteins activates various intracellular signaling pathways implicated in angiogenesis, cell adhesion and migration [4]. HPSE secretion or translocation into the nucleus may occur under the effect of different stimuli. Extracellular HSPGs degradation encourages cell migration, thus enhancing tumor cell invasiveness and metastasis [5]. Angiogenesis, EMT and inflammatory response are indirectly regulated by HPSE via HS-linked growth factors that are released after HS cleavage [6]. Nuclear HPSE translocation is implicated in cell differentiation, inflammation and glucose metabolism through gene transcription regulation mechanisms [7].](/document/doi/10.1515/bmc-2015-0021/asset/graphic/j_bmc-2015-0021_fig_001.jpg)
HPSE: processing, localization, enzymatic and non-enzymatic activities.
HPSE is synthesized in the endoplasmic reticulum as a latent precursor (pro-HPSE). After moving to the Golgi apparatus, pro-HPSE is secreted outside the cell [1]. HPSE uptake [mediated by syndecans, mannose-6-phosphate-receptor and low-density lipoprotein receptor (LDLR)-related protein] [2], and its delivery to lysosome enables the proteolytic processing (by cathepsin-L) and activation of the precursor into the dimeric active enzyme [3]. The interaction of latent HPSE with HPSE-binding proteins activates various intracellular signaling pathways implicated in angiogenesis, cell adhesion and migration [4]. HPSE secretion or translocation into the nucleus may occur under the effect of different stimuli. Extracellular HSPGs degradation encourages cell migration, thus enhancing tumor cell invasiveness and metastasis [5]. Angiogenesis, EMT and inflammatory response are indirectly regulated by HPSE via HS-linked growth factors that are released after HS cleavage [6]. Nuclear HPSE translocation is implicated in cell differentiation, inflammation and glucose metabolism through gene transcription regulation mechanisms [7].
Heparanase and fibrosis
Tissue fibrosis is an unregulated wound-healing response characterized by a gradual accumulation and decreased remodeling of ECM. Chronic injuries are the main triggers of this process in major organs (22). Common pathways and effector cells are implicated in fibrosis in various parenchymal organs. Persistent parenchymal cell injury leads to chronic inflammation that, in turn, stimulates the activation of effector cells into fibrogenic myofibroblasts. Myofibroblasts express the alpha isoform of smooth muscle actin (α-SMA) and secrete large amounts of ECM proteins (primarily collagen, fibronectin and laminin), which are responsible for tissue scarring and organ architecture deformation and failure. The source of the myofibroblast pool varies, including resident fibroblasts, fibrocytes, pericytes and epithelial cells undergoing epithelial-to-mesenchymal transition (EMT) (23). The transdifferentiation of epithelial cells into myofibroblast-like cells (i.e. EMT) is characterized by the loss of epithelial markers and the acquisition of mesenchymal ones, and by an increased migration and matrix protein secretion (24). Increasing interest has been paid to HPSE in this field in the last decade, since its involvement in the EMT of renal tubular cells in kidney fibrosis was discovered. HPSE is overexpressed by injured glomerular and tubular cells exposed to albuminuria, high glucose levels and advance glycosylation end products (25). Once secreted, HPSE is a key regulator of FGF-2 and TGF-β activity, the main pro-fibrotic factors and inducers of EMT in the kidney (26, 27). HPSE knock-down in a proximal tubular cell line prevents the EMT induced by FGF-2 (26), and delays the EMT induced by TGF-β (27). A lack of HPSE also prevents TGF-β overexpression by injured tubular cells in vitro (27), and HPSE-ko mice show no increase in TGF-β in injured kidneys, and no onset of fibrosis (28). These findings point to HPSE as a potential pharmacological target in chronic renal disease (29). There are few and sometimes conflicting data on the role of (HPSE) in promoting fibrosis in organs other than the kidney. HPSE regulates TGF-β availability by shedding syndecan-1, and its upregulation has consequently been found associated with intestinal fibrosis in vivo (30).
Unlike the well-known role of HPSE as a pro-metastatic agent in hepatocellular carcinoma, the involvement of HPSE in the pre-cancerous condition of liver fibrosis/cirrhosis is still not clear. Ikeguchi et al. found an inverse correlation between HPSE expression and stage of liver fibrosis in human tissue (31). Xiao et al. reported no difference in HPSE expression between cirrhotic and normal liver tissue (32). Discordant data have also emerged from a rat model of chronic liver disease: Ohayon et al. found an increase of HPSE in advanced fibrosis (33), whereas Goldshmidt et al. had previously described HPSE protein levels peaking in the early stages of fibrosis (34). Further studies will be needed to ascertain whether the mechanism by which HPSE exerts its pro-fibrotic action in the kidney is tissue-specific or applies to other organs too.
Heparanase and inflammation
There is now strong evidence of HS being involved in several aspects of inflammation. In particular, it controls the binding and bioavailability of cytokines in ECM (35, 36), it modulates the interaction of leukocytes with endothelium and ECM (37), and it regulates the activation of toll-like receptors (TLRs) (38, 39). Given these properties, HS degradation by HPSE may regulate inflammation in several steps, via chemokine release and modulation of the chemoactive gradient, leukocyte recruitment, extravasation and activation.
Recent findings suggest that HPSE can either facilitate or limit inflammatory responses, seemingly depending on tissue- or cell-specific contextual cues that may dictate the direction taken by its action in inflammation (40). In colitis (characterized by an abundant luminal flora and the activation of TLR signaling pathways), HPSE of epithelial origin modulates the inflammatory phenotype of macrophages, preventing the inflammation from regressing, and switching macrophage response to a chronic inflammatory pattern (41). HPSE also strongly increases the in vitro activation of macrophages by lipopolysaccharide (LPS, a specific stimulator of TLR4 signaling), with a secondary increase in TNFα, IL-6 and IL-12 production. Activated macrophages can, in turn, induce epithelial HPSE expression (via a TNFα-dependent mechanism), and post-translational processing of the pro-enzyme via an increased secretion of cathepsin-L, thereby fueling a self-sustaining inflammatory circuit (41). Other studies have confirmed that HPSE participates in regulating TLRs and the subsequent release of pro-inflammatory cytokines by generating soluble HS fragments (42). The involvement of HPSE in renal inflammation has recently been demonstrated in mouse models of sepsis (43), and diabetic nephropathy (28). As regards the latter, diabetic HPSE-ko mice do not develop diabetic nephropathy, and macrophage infiltration is lower than in controls (28). Under diabetic conditions, the latent HPSE overexpressed by glomerular cells and post-translationally activated by cathepsin-L of tubular origin sustains the continuous activation of kidney-damaging macrophages, giving rise to chronic inflammatory conditions and fostering macrophage-mediated renal injury (44). One of the latest studies also proved that HPSE contributes significantly to pulmonary inflammatory cell recruitment in a model of allergic asthma (45). Another important parameter contributing to inflammation is macrophage polarization vs. an inflammatory (M1) or regenerative (M2) phenotype, and a recent work suggested that HPSE may be responsible for macrophage polarization in cancer (46).
Indirect but strong evidence of the role of HPSE in inflammation in humans emerges from several clinical trials on the efficacy of heparin as an anti-inflammatory agent. Heparin and heparin-derived compounds compete with the HS chain in binding to HPSE, so they have proved highly efficient HPSE inhibitors. Heparin and its derivatives have been shown to exert anti-inflammatory effects in the treatment of asthma and in patients undergoing cardiopulmonary bypass and or cataract surgery (47).
Heparanase and cancer
The first studies demonstrating an enzymatic activity attributable to (HPSE) date back to 1983, when Nakajima et al. (48) found a relationship between HS degradation and the invasive and metastatic potential of B16 melanoma cells, and Vlodavsky et al. (49) demonstrated the same link in lymphomas. After the HPSE gene had been identified and cloned in the late 1990s, several experimental studies showed that this enzyme’s overexpression was instrumental in increasing the dissemination and metastatic potential of cancer cells, partly as a result of the establishment of a new vascular network (4, 50). Various gene silencing strategies for HPSE also coincided with a reduction in the invasive and metastatic properties of tumor cells (51, 52). HPSE promotes tumor progression not only thanks to HS cleavage in the ECM, but also due to the release and diffusion of HS-linked pro-angiogenic factors (e.g. VEGF, FGF-2), cytokines and enzymes (e.g. MMPs) (53). When released by HPSE activity, these molecules contribute to the creation of a microenvironment favorable to cancer cell growth. On the whole, these experimental data have supported the formulation of the fundamental concept (or hypothesis) that enzymatic cleavage of HS side chains by HPSE is a necessary condition for ECM remodeling by cancer cells for the purpose of invasive and metastatic processes (54). This hypothesis was subsequently confirmed by clinical evidence in human patients. HPSE expression was upregulated and correlated with reduced survival in all the major types of tumors analyzed (carcinomas, sarcomas, hematological and CNS tumors). These data have been widely reported and discussed in several recent reviews (5, 53–56), so we will not dwell further on these aspects here.
Recently, Hermano et al. (46) highlighted a new role for HPSE in the development of pancreatic adenocarcinoma. As mentioned above in the section on HPSE and inflammation, their results underscore the involvement of HPSE in directing the tumor-promoting behavior of tumor-associated macrophages (TAMs). Moreover, the analysis of human pancreatic adenocarcinoma specimens revealed a direct correlation between HPSE expression and macrophages infiltration. These findings suggested that the degree of HPSE expression may be highly significant when it comes to defining a target patient subgroup that may benefit from pharmacological treatment targeting TAMs.
In addition to its well-documented enzymatic activity in the extracellular environment, several studies have recently suggested a possible intracellular activity for HPSE, with an importance in the course of tumor progression that has yet to be well clarified. HPSE has been shown to have a role at nuclear level by shedding syndecan-1 in mesenchymal tumor cells (57), and the loss of syndecan-1 in the nucleus promotes the transcription of genes that contribute to the tumor phenotype (e.g. MMP-9) as a result of histone acetyltransferase activity (11). Other studies have suggested a link between HPSE and NF-kB-dependent gene regulation (58, 59). On the other hand, Cohen-Kaplan et al. found that an increase in HPSE at nuclear level correlated with a slower tumor growth and a longer survival in patients with head and neck tumors (60). Nuclear HPSE was also found to down-regulate the expression of pro-tumorigenic genes in a melanoma model, pointing to a tumor-suppressive role instead of the secreted enzyme’s well-known pro-tumorigenic function (61). These relatively recent findings have led to the suggestion that the ability of HPSE to regulate gene transcription and cell signaling depends not only on the combination of its enzymatic and non-enzymatic activities, but also on its extracellular and/or intracellular location. Further studies will be needed to fully clarify the role of nuclear HPSE in tumorigenesis.
The pro-tumorigenic properties of HPSE seems to be mediated also by its pro-autophagic function, as it was demonstrated in tumor xenograft models of human cancer. The cells overexpressing HPSE were more resistant to stress and chemotherapy as a consequence of an increased autophagy, effects that were reversed by lysosome inhibitor treatment. On the whole, these results establish a new role for HPSE in modulating autophagy in normal and malignant cells, and thereby conferring growth advantages under stress as well as resistance to chemotherapy (62).
Beside numerous experimental data concerning HPSE as pro-tumorigenic molecule, it is of interest to mention the work of Caruana and colleagues (63) who first demonstrated that the ECM degradation by means of HPSE can be exploited for anti-tumor strategies. Indeed, it was shown that inducing HPSE expression in long-term ex vivo-expanded T cells co-expressing a tumor-specific CAR (chimeric antigen receptor) increased their capacity to degrade the ECM and their anti-tumor activity. This experimental strategy could enhance the anti-tumor activity of CAR-redirected T cells in patients with stroma-rich solid tumors.
Conclusions
It is now evident from all the above contributions that HPSE can be considered as a protein with multiple functions in various diseases. In the light of its dual, enzymatic and non-enzymatic function, and intra- and extracellular localization, future molecules with an inhibitory function will need to act selectively, inhibiting one function but not the other, depending on the context. When the crystal structure of the enzyme becomes available, it will be possible to identify the protein domain responsible for a given function, and then design its potential inhibitors. Bearing in mind that, once HPSE has been deactivated, no other molecules are capable of exerting its same function, this enzyme seems particularly interesting as a drug target. Several HPSE inhibitors are currently the object of clinical trials, and some have already shown some anti-tumor efficacy (64). It is to be hoped that future HPSE-targeting drugs will bring benefits not only for cancer patients, but also for those with other disorders in which the enzyme has now revealed an important etiological role.
- List of abbreviations
- ECM
extracellular matrix
- EMT
epithelial-to-mesenchymal transition
- GAGs
glycosaminoglycans
- HPSE
heparanase
- HSPG
heparan sulfate proteoglycan
- HS
heparan sulfate
Acknowledgments
This work was supported by a University of Padova grant to M.O. We thank Dr. Andrea Donadon (University of Padova) for technical support in the preparation of the artwork and Mrs Frances Coburn for language editing of the manuscript.
References
1. Bishop JR, Schuksz M, Esko JD. Heparan sulphate proteoglycans fine-tune mammalian physiology. Nature 2007; 446: 1030–7.10.1038/nature05817Search in Google Scholar PubMed
2. Theocharis A, Gialeli C, Hascall V, Karamanos NK. Extracellular matrix: a functional scaffold. In: Karamanos NK, editor. Extracellular matrix: pathobiology and signaling. Berlin: Walter de Gruyter, 2012: 3–19.Search in Google Scholar
3. Kim SH, Turnbull J, Guimond S. Extracellular matrix and cell signalling: the dynamic cooperation of integrin, proteoglycan and growth factor receptor. J Endocrinol 2011; 209: 139–51.10.1530/JOE-10-0377Search in Google Scholar PubMed
4. Vlodavsky I, Friedmann Y, Elkin M, Aingorn H, Atzmon R, Ishai-Michaeli R, Bitan M, Pappo O, Peretz T, Michal I, Spector L, Pecker I. Mammalian heparanase: gene cloning, expression and function in tumor progression and metastasis. Nat Med 1999; 5: 793–802.10.1038/10518Search in Google Scholar PubMed
5. Vreys V, David G. Mammalian heparanase: what is the message?. J Cell Mol Med 2007; 11: 427–52.10.1111/j.1582-4934.2007.00039.xSearch in Google Scholar PubMed PubMed Central
6. Shteingauz A, Ilan N, Vlodavsky I. Processing of heparanase is mediated by syndecan-1 cytoplasmic domain and involves syntenin and α-actinin. Cell Mol Life Sci 2014; 71: 4457–70.10.1007/s00018-014-1629-9Search in Google Scholar PubMed PubMed Central
7. Zetser A, Levy-Adam F, Kaplan V, Gingis-Velitski S, Bashenko Y, Schubert S, Flugelman MY, Vlodavsky I, Ilan N. Processing and activation of latent heparanase occurs in lysosomes. J Cell Sci 2004; 117(Pt 11): 2249–58.10.1242/jcs.01068Search in Google Scholar PubMed
8. Nobuhisa T, Naomoto Y, Takaoka M, Tabuchi Y, Ookawa K, Kitamoto D, Gunduz E, Gunduz M, Nagatsuka H, Haisa M, Matsuoka J, Nakajima M, Tanaka N. Emergence of nuclear heparanase induces differentiation of human mammary cancer cells. Biochem Biophys Res Commun 2005; 331: 175–80.10.1016/j.bbrc.2005.03.129Search in Google Scholar PubMed
9. Nobuhisa T, Naomoto Y, Okawa T, Takaoka M, Gunduz M, Motoki T, Nagatsuka H, Tsujigiwa H, Shirakawa Y, Yamatsuji T, Haisa M, Matsuoka J, Kurebayashi J, Nakajima M, Taniguchi S, Sagara J, Dong J, Tanaka N. Translocation of heparanase into nucleus results in cell differentiation. Cancer Sci 2007; 98: 535–40.10.1111/j.1349-7006.2007.00420.xSearch in Google Scholar PubMed
10. Schubert SY, Ilan N, Shushy M, Ben-Izhak O, Vlodavsky I, Goldshmidt O. Human heparanase nuclear localization and enzymatic activity. Lab Invest 2004; 84: 535–44.10.1038/labinvest.3700084Search in Google Scholar PubMed
11. Purushothaman A, Hurst DR, Pisano C, Mizumoto S, Sugahara K, Sanderson RD. Heparanase-mediated loss of nuclear syndecan-1 enhances histone acetyltransferase (HAT) activity to promote expression of genes that drive an aggressive tumor phenotype. J Biol Chem 2011; 286: 30377–83.10.1074/jbc.M111.254789Search in Google Scholar PubMed PubMed Central
12. Wang F, Wang Y, Zhang D, Puthanveetil P, Johnson JD, Rodrigues B. Fatty acid-induced nuclear translocation of heparanase uncouples glucose metabolism in endothelial cells. Arterioscler Thromb Vasc Biol 2012; 32: 406–14.10.1161/ATVBAHA.111.240770Search in Google Scholar PubMed
13. Gingis-Velitski S, Zetser A, Flugelman MY, Vlodavsky I, Ilan N. Heparanase induces endothelial cell migration via protein kinase B/Akt activation. J Biol Chem 2004; 279: 23536–41.10.1074/jbc.M400554200Search in Google Scholar PubMed
14. Yuan L, Hu J, Luo Y, Liu Q, Li T, Parish CR, Freeman C, Zhu X, Ma W, Hu X, Yu H, Tang S. Upregulation of heparanase in high-glucose-treated endothelial cells promotes endothelial cell migration and proliferation and correlates with Akt and extracellular-signal-regulated kinase phosphorylation. Mol Vis 2012; 18: 1684–95.Search in Google Scholar
15. Riaz A, Ilan N, Vlodavsky I, Li JP, Johansson S. Characterization of heparanase-induced phosphatidylinositol 3-kinase AKT activation and its integrin dependence. J Biol Chem 2013; 288: 12366–75.10.1074/jbc.M112.435172Search in Google Scholar PubMed PubMed Central
16. Hu X, Zhang L, Jin J, Zhu W, Xu Y, Wu Y, Wang Y, Chen H, Webster KA, Chen H, Yu H, Wang J. Heparanase released from mesenchymal stem cells activates integrin beta1/HIF-2α/Flk-1 signaling and promotes endothelial cell migration and angiogenesis. Stem Cells 2015; 33: 1850–62.Search in Google Scholar
17. Goldshmidt O, Zcharia E, Cohen M, Aingorn H, Cohen I, Nadav L, Katz BZ, Geiger B, Vlodavsky I. Heparanase mediate cell adhesion independent of its enzymatic activity. FASEB J 2003; 17: 1015–25.10.1096/fj.02-0773comSearch in Google Scholar PubMed
18. Sotnikov I, Hershkoviz R, Grabovsky V, Ilan N, Cahalon L, Vlodavsky I, Alon R, Lider O. Enzymatically quiescent heparanase augments T cell interactions with VCAM-1 and extracellular matrix components under versatile dynamic contexts. J Immunol 2004; 172: 5185–93.10.4049/jimmunol.172.9.5185Search in Google Scholar PubMed
19. Zetser A, Bashenko Y, Edovitsky E, Levy-Adam F, Vlodavsky I, Ilan N. Heparanase induces vascular endothelial growth factor expression: correlation with p38 phosphorylation levels and Src activation. Cancer Res 2006; 66: 1455–63.10.1158/0008-5472.CAN-05-1811Search in Google Scholar PubMed
20. Ben-Zaken O, Gingis-Velitski S, Vlodavsky I, Ilan N. Heparanase induces Akt phosphorylation via a lipid raft receptor. Biochem Biophys Res Commun 2007; 361: 829–34.10.1016/j.bbrc.2007.06.188Search in Google Scholar PubMed PubMed Central
21. Fux L, Feibish N, Cohen-Kaplan V, Gingis-Velitski S, Feld S, Geffen C, Vlodavsky I, Ilan N. Structure-function approach identifies a COOH-terminal domain that mediates heparanase signaling. Cancer Res. 2009; 69: 1758–67.10.1158/0008-5472.CAN-08-1837Search in Google Scholar PubMed PubMed Central
22. Rockey DC, Bell PD, Hill JA. Fibrosis-A common pathway to organ injury and failure. N Engl J Med 2015; 372: 1138–49.10.1056/NEJMra1300575Search in Google Scholar PubMed
23. He W, Dai C. Key fibrogenic signaling. Curr Pathobiol Rep 2015; 3: 183–192.10.1007/s40139-015-0077-zSearch in Google Scholar PubMed PubMed Central
24. Lamouille S, Xu J, Derynck R. Molecular mechanism of epithelial to mesenchymal transition. Nat Rev Mol Cell Biol 2014; 15: 178–96.10.1038/nrm3758Search in Google Scholar
25. Masola V, Gambaro G, Tibaldi E, Onisto M, Abaterusso C, Lupo A. Masola V. Regulation of heparanase by albumin and advanced glycation end products in proximal tubular cells. Biochim Biophys Acta 2011; 1813: 1475–82.10.1016/j.bbamcr.2011.05.004Search in Google Scholar
26. Masola V, Gambaro G, Tibaldi E, Brunati AM, Gastaldello A, D’Angelo A, Onisto M, Lupo A. Heparanase and syndecan-1 interplay orchestrates fibroblast growth factor-2-induced epithelial-mesenchymal transition in renal tubular cells. J Biol Chem 2012; 287: 1478–88.10.1074/jbc.M111.279836Search in Google Scholar
27. Masola V, Zaza G, Secchi MF, Gambaro G, Lupo A, Onisto M. Heparanase is a key player in renal fibrosis by regulating TGF-β expression and activity. Biochim Biophys Acta 2014; 1843: 2122–8.10.1016/j.bbamcr.2014.06.005Search in Google Scholar
28. Gil N, Goldberg R, Neuman T, Garsen M, Zcharia E, Rubinstein AM, van Kuppevelt T, Meirovitz A, Pisano C, Li JP, van der Vlag J, Vlodavsky I, Elkin M. Heparanase is essential for the development of diabetic nephropathy in mice. Diabetes 2012; 61: 208–16.10.2337/db11-1024Search in Google Scholar
29. Masola V, Onisto M, Zaza G, Lupo A, Gambaro G. A new mechanism of action of sulodexide in diabetic nephropathy: inhibits heparanase-1 and prevents FGF-2-induced renal epithelial-mesenchymal transition. J Transl Med 2012; 10: 213.10.1186/1479-5876-10-213Search in Google Scholar
30. Davids JS1, Carothers AM, Damas BC, Bertagnolli MM. Chronic cyclooxygenase-2 inhibition promotes myofibroblast-associated intestinal fibrosis. Cancer Prev Res (Phila) 2010; 3: 348–58.10.1158/1940-6207.CAPR-09-0146Search in Google Scholar
31. Ikeguchi M, Hirooka Y, Kaibara N. Heparanase gene expression and its correlation with spontaneous apoptosis in hepatocytes of cirrhotic liver and carcinoma. Eur J Cancer 2003; 39: 86–90.10.1016/S0959-8049(02)00558-0Search in Google Scholar
32. Xiao Y, Kleeff J, Shi X, Büchler MW, Friess H. Heparanase expression in hepatocellular carcinoma and the cirrhotic liver. Hepatol Res 2003; 26: 192–98.10.1016/S1386-6346(03)00107-4Search in Google Scholar
33. Ohayon O, Mawasi N, Pevzner A, Tryvitz A, Gildor T, Pines M, Rojkind M, Paizi M, Spira G. Halofuginone upregulates the expression of heparanase in thioacetamide-induced liver fibrosis in rats. Lab Invest 2008; 88: 627–33.10.1038/labinvest.2008.30Search in Google Scholar PubMed
34. Goldshmidt O, Yeikilis R, Mawasi N, Paizi M, Gan N, Ilan N, Pappo O, Vlodavsky I, Spira G. Heparanase expression during normal liver development and following partial hepatectomy. J Pathol 2004; 203: 594–602.10.1002/path.1554Search in Google Scholar PubMed
35. Parish CR. The role of heparan sulphate in inflammation. Nat Rev Immunol 2006; 6: 633–43.10.1038/nri1918Search in Google Scholar PubMed
36. Taylor KR, Gallo RL. Glycosaminoglycans and their proteoglycans: host-associated molecular patterns for initiation and modulation of inflammation. FASEB J 2006; 20: 9–22.10.1096/fj.05-4682revSearch in Google Scholar PubMed
37. Wang L, Fuster M, Sriramarao P, Esko JD. Endothelial heparan sulfate deficiency impairs L-selectin- and chemokine-mediated neutrophil trafficking during inflammatory responses. Nat Immunol 2005; 6: 902–10.10.1038/ni1233Search in Google Scholar PubMed
38. Brunn GJ, Bungum MK, Johnson GB, Platt JL. Conditional signaling by Toll-like receptor 4. FASEB J 2005; 19: 872–4.10.1096/fj.04-3211fjeSearch in Google Scholar PubMed
39. Akbarshahi H, Axelsson JB, Said K, Malmstrom A, Fischer H, Andersson R. TLR4 dependent heparan sulphate-induced pancreatic inflammatory response is IRF3-mediated. J Transl Med 2011; 9: 219.10.1186/1479-5876-9-219Search in Google Scholar PubMed PubMed Central
40. Goldberg R, Meirovitz A, Hirshoren N, Bulvik R, Binder A, Rubinstein AM, Elkin M. Versatile role of heparanase in inflammation. Matrix Biol 2013; 32: 234–40.10.1016/j.matbio.2013.02.008Search in Google Scholar PubMed PubMed Central
41. Lerner I, Hermano E, Zcharia E, Rodkin D, Bulvik R, Doviner V, Rubinstein AM, Ishai-Michaeli R, Atzmon R, Sherman Y, Meirovitz A, Peretz T, Vlodavsky I, Elkin M. Heparanase powers a chronic inflammatory circuit that promotes colitis-associated tumorigenesis in mice. J Clin Invest 2011; 121: 1709–21.10.1172/JCI43792Search in Google Scholar PubMed PubMed Central
42. Goodall KJ, Poon IK, Phipps S, Hulett MD. Soluble heparan sulfate fragments generated by heparanase trigger the release of pro-inflammatory cytokines through TLR-4. PLoS One 2014; 9: e109596.10.1371/journal.pone.0109596Search in Google Scholar PubMed PubMed Central
43. Lygizos MI, Yang Y, Altmann CJ, Okamura K, Hernando AA, Perez MJ, Smith LP, Koyanagi DE, Gandjeva A, Bhargava R, Tuder RM, Faubel S,Schmidt EP. Heparanase mediates renal dysfunction during early sepsis in mice. Physiol Rep 2013; 1: e00153.10.1002/phy2.153Search in Google Scholar PubMed PubMed Central
44. Goldberg R, Rubinstein AM, Gil N, Hermano E, Li JP, van der Vlag J, Atzmon R, Meirovitz A, Elkin M. Role of heparanase-driven inflammatory cascade in pathogenesis of diabetic nephropathy. Diabetes 2014; 63: 4302–13.10.2337/db14-0001Search in Google Scholar PubMed
45. Morris A, Wang B, Waern I, Venkatasamy R, Page C, Schmidt EP, Wernersson S, Li JP, Spina D. The role of heparanase in pulmonary cell recruitment in response to an allergic but not non-allergic stimulus. PLoS One 2015; 10: e0127032.10.1371/journal.pone.0127032Search in Google Scholar PubMed PubMed Central
46. Hermano E, Meirovitz A, Meir K, Nussbaum G, Appelbaum L, Peretz T, Elkin M. Macrophage polarization in pancreatic carcinoma: role of heparanase enzyme. J Natl Cancer Inst 2014; 106: dju332.10.1093/jnci/dju332Search in Google Scholar PubMed PubMed Central
47. Mousavi S, Moradi M, Khorshidahmad T, Motamedi M. Anti-inflammatory effects of heparin and its derivatives: a systematic review. Adv Pharmacol Sci 2015; 2015: 507151.10.1155/2015/507151Search in Google Scholar PubMed PubMed Central
48. Nakajima M, Irimura T, Di Ferrante D, Di Ferrante N, Nicolson GL. Heparan sulfate degradation: relation to tumor invasive and metastatic properties of mouse B16 melanoma sublines. Science 1983; 220: 611–3.10.1126/science.6220468Search in Google Scholar PubMed
49. Vlodavsky I, Fuks Z, Bar-Ner M, Ariav Y, Schirrmacher V. Lymphoma cell-mediated degradation of sulfated proteoglycans in the subendothelial extracellular matrix: relationship to tumor cell metastasis. Cancer Res 1983; 43: 2704–11.Search in Google Scholar
50. Cohen I, Pappo O, Elkin M, San T, Bar-Shavit R, Hazan R, Peretz T, Vlodavsky I, Abramovitch R. Heparanase promotes growth, angiogenesis and survival of primary breast tumors. Int J Cancer 2006; 118: 1609–17.10.1002/ijc.21552Search in Google Scholar PubMed
51. Edovitsky E, Elkin M, Zcharia E, Peretz T, Vlodavsky I. Heparanase gene silencing, tumor invasiveness, angiogenesis, and metastasis. J Natl Cancer Inst 2004; 96: 1219–30.10.1093/jnci/djh230Search in Google Scholar PubMed
52. Lerner I, Baraz L, Pikarsky E, Meirovitz A, Edovitsky E, Peretz T, Vlodavsky I, Elkin M. Function of heparanase in prostate tumorigenesis: potential for therapy. Clin Cancer Res 2008; 14: 668–76.10.1158/1078-0432.CCR-07-1866Search in Google Scholar PubMed
53. Barash U, Cohen-Kaplan V, Dowek I, Sanderson RD, Ilan N, Vlodavsky I. Proteoglycans in health and disease: new concepts for heparanase function in tumor progression and metastasis. FEBS J 2010; 277: 3890–903.10.1111/j.1742-4658.2010.07799.xSearch in Google Scholar PubMed PubMed Central
54. Ilan N, Elkin M, Vlodavsky I. Regulation, function and clinical significance of heparanase in cancer metastasis and angiogenesis. Int J Biochem Cell Biol 2006; 38: 2018–39.10.1016/j.biocel.2006.06.004Search in Google Scholar PubMed
55. Arvatz G, Shafat I, Levy-Adam F, Ilan N, Vlodavsky I. The heparanase system and tumor metastasis: is heparanase the seed and soil? Cancer Metastasis Rev 2011; 30: 253–68.10.1007/s10555-011-9288-xSearch in Google Scholar PubMed
56. Hammond E, Khurana A, Shridhar V, Dredge K. The role of heparanase and sulfatases in the modification of heparan sulfate proteoglycans within the tumor microenvironment and opportunities for novel cancer therapeutics. Front Oncol 2014; 4: 195.10.3389/fonc.2014.00195Search in Google Scholar PubMed PubMed Central
57. Zong F, Fthenou E, Wolmer N, Hollósi P, Kovalszky I, Szilák L, Mogler C, Nilsonne G, Tzanakakis G, Dobra K. Syndecan-1 and FGF-2, but not FGF receptor-1, share a common transport route and co-localize with heparanase in the nuclei of mesenchymal tumor cells. PLoS One 2009; 4: e7346.10.1371/journal.pone.0007346Search in Google Scholar PubMed PubMed Central
58. Cao HJ, Fang Y, Zhang X, Chen WJ, Zhou WP, Wang H, Wang LB, Wu JM. Tumor metastasis and the reciprocal regulation of heparanase gene expression by nuclear factor κB in human gastric carcinoma tissue. World J Gastroenterol 2005; 11: 903–7.10.3748/wjg.v11.i6.903Search in Google Scholar PubMed PubMed Central
59. Wu W, Pan C, Meng K, Zhao L, Du L, Liu Q, Lin R. Hypoxia activates heparanase expression in an NF-κB dependent manner. Oncol Rep 2010; 23: 255–61.Search in Google Scholar
60. Cohen-Kaplan V, Jrbashyan J, Yanir Y, Naroditsky I, Ben-Izhak O, Ilan N, Doweck I, Vlodavsky I. Heparanase induces signal transducer and activator of transcription (STAT) protein phosphorylation: preclinical and clinical significance in head and neck cancer. J Biol Chem 2012; 287: 6668–78.10.1074/jbc.M111.271346Search in Google Scholar PubMed PubMed Central
61. Yang Y, Gorzelanny C, Bauer AT, Halter N, Komljenovic D, Bäuerle T, Borsig L, Roblek M, Schneider SW. Nuclear heparanase-1 activity suppress melanoma progression via its DNA-binding affinity. Oncogene 2015; Mar 9. doi: 10.1038/onc.2015.40.10.1038/onc.2015.40Search in Google Scholar PubMed
62. Shteingauz A, Boyango I, Naroditsky I, Hammond E, Gruber M, Doweck I, Ilan N, Vlodavsky I. Heparanase enhances tumor growth and chemo-resistance by promoting autophagy. Cancer Res 2015; 75: 3946–57.10.1158/0008-5472.CAN-15-0037Search in Google Scholar PubMed PubMed Central
63. Caruana I, Savoldo B, Hoyos V, Weber G, Liu H, Kim ES, Ittmann MM, Marchetti D, Dotti G. Heparanase promotes tumor infiltration and antitumor activity of CAR-redirected T lymphocytes. Nat Med 2015; 21: 524–9.10.1038/nm.3833Search in Google Scholar PubMed PubMed Central
64. Masola V, Secchi MF, Gambaro G, Onisto M. Heparanase as a target in cancer therapy. Curr Cancer Drug Targets 2014; 14: 286–93.10.2174/1568009614666140224155124Search in Google Scholar PubMed
©2015 by De Gruyter
This article is distributed under the terms of the Creative Commons Attribution Non-Commercial License, which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.
Articles in the same Issue
- Frontmatter
- Reviews
- Recent advances in siRNA delivery
- Trefoil factor family peptides – friends or foes?
- Developments and new vistas in the field of melanocortins
- Cell-cell and cell-matrix adhesion in survival and metastasis: Stat3 versus Akt
- Glyoxalase biochemistry
- Short Conceptual Overviews
- Recent data concerning heparanase: focus on fibrosis, inflammation and cancer
- Adaptive regulation of glucose transport, glycolysis and respiration for cell proliferation
Articles in the same Issue
- Frontmatter
- Reviews
- Recent advances in siRNA delivery
- Trefoil factor family peptides – friends or foes?
- Developments and new vistas in the field of melanocortins
- Cell-cell and cell-matrix adhesion in survival and metastasis: Stat3 versus Akt
- Glyoxalase biochemistry
- Short Conceptual Overviews
- Recent data concerning heparanase: focus on fibrosis, inflammation and cancer
- Adaptive regulation of glucose transport, glycolysis and respiration for cell proliferation