Next Article in Journal
Novel NFκB Inhibitor SC75741 Mitigates Chondrocyte Degradation and Prevents Activated Fibroblast Transformation by Modulating miR-21/GDF-5/SOX5 Signaling
Next Article in Special Issue
MT1-MMP Cooperates with TGF-β Receptor-Mediated Signaling to Trigger SNAIL and Induce Epithelial-to-Mesenchymal-like Transition in U87 Glioblastoma Cells
Previous Article in Journal
Impact of Host Immune Status on Discordant Anti-SARS-CoV-2 Circulating B Cell Frequencies and Antibody Levels
Previous Article in Special Issue
Therapeutic Approaches Targeting Proteostasis in Kidney Disease and Fibrosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Heparanase Regulatory Network in Health and Disease

Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3083, Australia
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2021, 22(20), 11096; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222011096
Submission received: 9 September 2021 / Revised: 7 October 2021 / Accepted: 11 October 2021 / Published: 14 October 2021
(This article belongs to the Special Issue Proteolysis of Extracellular Matrix in Human Disease 2.0)

Abstract

:
The extracellular matrix (ECM) is a structural framework that has many important physiological functions which include maintaining tissue structure and integrity, serving as a barrier to invading pathogens, and acting as a reservoir for bioactive molecules. This cellular scaffold is made up of various types of macromolecules including heparan sulfate proteoglycans (HSPGs). HSPGs comprise a protein core linked to the complex glycosaminoglycan heparan sulfate (HS), the remodeling of which is important for many physiological processes such as wound healing as well as pathological processes including cancer metastasis. Turnover of HS is tightly regulated by a single enzyme capable of cleaving HS side chains: heparanase. Heparanase upregulation has been identified in many inflammatory diseases including atherosclerosis, fibrosis, and cancer, where it has been shown to play multiple roles in processes such as epithelial-mesenchymal transition, angiogenesis, and cancer metastasis. Heparanase expression and activity are tightly regulated. Understanding the regulation of heparanase and its downstream targets is attractive for the development of treatments for these diseases. This review provides a comprehensive overview of the regulators of heparanase as well as the enzyme’s downstream gene and protein targets, and implications for the development of new therapeutic strategies.

1. Introduction

The extracellular matrix (ECM) is a complex three-dimensional structural network comprised of proteins and polysaccharides that surround cells and tissues in multicellular organisms. This extracellular architecture is responsible for offering structural support and integrity to tissues and provides protection from invading cells and pathogens. It also has roles in many cellular processes, including cell survival, growth, migration, and differentiation [1]. Key components of the ECM include proteoglycans and fibrous proteins such as collagen, elastin, fibronectin, and laminin. Of particular interest to this review are the heparan sulfate proteoglycans (HSPGs).
The HSPGs are comprised of a protein core with covalently linked side chains of the variably sulfated glycosaminoglycan heparan sulfate (HS). HSPGs are found within the ECM (agrin, perlecan, and type XVIII collagen), bound to the cell membrane (syndecans and glypicans), or within secretory vesicles (serglycin) [2,3]. They can also be found in the nucleus [4].
There are many proteins that bind HS (Table 1). Indeed, over 400 human proteins have been shown to bind HS or the structurally related heparin [5] (where heparin-binding likely predicts HS-binding abilities). Many of these binding proteins have been confirmed by proteomic, surface plasmon resonance, and column chromatographic methods. These proteins include growth factors, e.g., fibroblast growth factor (FGF); cytokines, e.g., monocyte chemoattractant protein-1 (MCP-1); and other ECM components, e.g., collagen. HS-binding molecules either interact through a specific HS-binding sequence motif, e.g., FGF [6] or in a nonspecific charge-dependent manner, e.g., fibronectin [7,8]. By binding HS, these proteins are sequestered within the matrix. Many also require HS for activity; for example, the formation of many chemokine gradients requires HS to facilitate chemokine oligomerization [9].
The abundance of HS on the cell surface and its importance in several pathways led to the discovery that HS acts as a co-receptor for several signaling receptors. These include the FGF receptor (FGFR) where cell surface HS is required for activation of the receptor [10] and vascular endothelial growth factor receptor (VEGFR) where HS can activate VEGFR in trans- from neighboring cells [11]. HS expressed on the surface of endothelial cells also acts as an adhesion receptor for migrating lymphocytes [12]. Given the diverse roles of HS in normal physiology and disease, its regulation and turnover are important to understand. In mammals, the turnover of HS and therefore ECM homeostasis is regulated by one enzyme: heparanase.
Heparanase is a member of the glucuronidase family and recognizes HS polysaccharide chains at sites of high sulfation. It catalyzes the hydrolysis of the β-linkage joining glucuronic acid and N-acetylglucosamine residues in HS chains, generating polysaccharide fragments of 10–20 units long [48]. Heparanase has several roles in physiological functions including wound healing [49] and leukocyte trafficking [50,51,52,53]. It also plays many roles in a number of different disease settings such as cancer and inflammatory diseases, where heparanase expression is upregulated and contributes to disease progression, making it an important enzyme to study.
Heparanase expression is regulated by several factors, such as cytokines, growth factors, and metabolites. In turn, heparanase can modulate the expression of several other genes and regulate the activity and bioavailability of various proteins and molecules. A description of this network in its entirety, including in all physiological and disease settings, has not yet been described in this form. Here we present an overview of this heparanase network as well as discuss how these links impact disease and what this understanding will mean for linking heparanase to disease diagnosis and treatment.

2. Regulation of Heparanase Expression

2.1. Heparanase Expression and Tissue Distribution

The human heparanase gene is located at chromosome 4q21.23 and spans 40 kb. The human, mouse, and rat heparanase genes are highly conserved, with the human and animal heparanase amino acid sequences sharing at least 80% identity. Under normal physiological conditions, the heparanase promoter is silenced by methylation [54,55]. Certain single nucleotide polymorphisms (SNPs) arising within the heparanase gene are associated with altered heparanase gene expression [56]. These same SNPs are also associated with heparanase mRNA expression in hematological malignancies [57].
The physiological expression of human heparanase was first reported in only the placenta and immune organs including the spleen, lymph node, peripheral blood, bone marrow, and fetal liver [58]. High expression has now been widely confirmed in immune cells, as well as observed in the esophagus, lung, heart muscle, keratinocytes, endothelial cells, and placental trophoblasts [59]. Recent advances in cell separation and RNAseq have allowed for detection of heparanase expression with increased sensitivity (less than 5 transcripts per million) in other human tissues including in the brain, endocrine organs, and the digestive tract [60] (Data available from https://www.proteinatlas.org/ENSG00000173083-HPSE/tissue, accessed on 16 June 2021).
During normal cellular processes, heparanase expression can be upregulated in response to various stimuli, for example, upon immune cell activation [50,61,62,63]. Expression of heparanase is also dysregulated in many disease settings, such as its upregulation in cancer [64]. Heparanase gene expression during physiological and pathological processes is modulated by several transcription factors, miRNAs, cytokines, growth factors, and other signaling molecules. As well as these host factors, bacteria, viruses, and certain therapeutics have also been shown to alter heparanase expression (Figure 1). These regulatory factors are summarised in Table 2.

2.2. Transcription Factors

Wild-type p53 is a master regulator of normal cell cycle and apoptotic processes [128]. During cellular homeostasis, heparanase gene expression is suppressed by wild-type p53 via direct binding to the heparanase promoter [77]. Thus, the mutation of p53 that can occur during oncogenesis results in aberrant heparanase expression. As well as a lack of repression, heparanase expression can be actively upregulated. Through cloning and sequencing of the heparanase promoter, the transcription factors GA-binding protein (GABP), specificity protein 1 (Sp1), and Sp3 were found to directly upregulate heparanase gene expression [70]. Early growth response 1 (EGR1) was later shown to also positively regulate heparanase gene expression through direct activation of the heparanase promoter [61,62,66,67]. Finally, NF-κB, a potent transcription factor downstream of many signaling pathways, can also increase heparanase expression in tumor cells [72,74,75,76].

2.3. miRNA

Micro RNAs (miRNAs) are emerging as important regulators of tumorigenesis given they regulate hundreds of mRNAs and are widely dysregulated in cancer [129]. In metastatic breast cancer cells, the miRNA miR-1258 was found to suppress heparanase expression and subsequently control tumor invasion and metastasis [79]. Patient tissues of invasive ductal carcinomas also exhibited lower levels of miR-1258 and higher heparanase expression relative to matched normal mammary gland tissue [79]. Another miRNA, miR-1252-5p, was also recently identified to regulate heparanase expression in multiple myeloma [80]. Since miRNAs show potential as directed therapeutics, miR-1258 may be a prospective candidate for treatment of heparanase-mediated metastatic cancer.

2.4. Cytokines

Heparanase plays several key roles during inflammation, including immune cell migration and cell signaling [130]. Thus, it is not surprising that several inflammatory cytokines have been shown to upregulate heparanase expression. These include interferon-γ (IFN-γ), interleukin (IL)-1β, IL-2, IL-15, IL-17, MCP-1 and tumor necrosis factor-α (TNF-α) [50,81,82,85,87,88,104,131]. It remains unclear how several of these cytokines upregulate heparanase expression, though it is likely that the heparanase gene is a downstream target of these cytokine signaling pathways. However, for cytokines in which the mechanism has been explored, it appears that the mechanisms may differ in different settings. One study found that heparanase upregulation in TNF-α treated endothelial cells was independent of NF-κB, PI-3K, MAP kinase, and c-Jun kinase, but was dependent on caspase 8 [82]. In contrast, another study found that canonical NF-κB signaling was required for TNF-α induced heparanase upregulation in endothelial cells [73]. Another study to show TNF-α induction of heparanase (during colitis-associated tumorigenesis) proposed that since TNF-α also induced upregulation of EGR1 [132,133] that TNF-α induced heparanase expression via activation of EGR1, although this is yet to be confirmed.
There are still gaps in our understanding of how these cytokines upregulate heparanase. Defining the mechanisms of cytokine-mediated heparanase upregulation and their contribution in different physiological and disease settings is required to fully understand the relationship between cytokine signaling and heparanase function. Despite our gaps in understanding of how cytokines upregulate heparanase, there are clearly multiple mechanisms at play during inflammatory responses. This multifaceted upregulation of heparanase likely ensures its robust expression and thus contributes to both normal immune responses and inflammatory disease pathologies.

2.5. Growth Factors

Growth factors can also regulate heparanase expression. Of these, VEGF was shown to act differentially depending on the setting: reducing heparanase expression in endothelial cells [82] and increasing heparanase expression in melanoma cells [92]. Hepatocyte growth factor (HGF) has also been shown to upregulate heparanase expression at the transcriptional level in lung and gastric cancer cells [89,90]. In contrast to TNF-α described above, HGF upregulated heparanase in gastric cancer cells through the PI3 kinase/Akt/NF-κB pathway [90]. A number of other growth factors—basic fibroblast growth factor (bFGF), FGF23, and platelet-derived growth factor—have also been shown to increase heparanase expression in cancer cells [89,91]. Thus, growth factors are another group of proteins that are central to regulating heparanase expression during physiological and pathological processes.

2.6. Hormones and Metabolites

Other signaling molecules can also regulate heparanase expression, including hormones, metabolites, and reactive oxygen species (ROS). Estrogen signaling has been shown to influence heparanase expression. Estrogen in breast cancer cells increases heparanase expression [100,102,103], and treatment of cholangiocarcinoma cells (bile duct cancer) with the estrogenic inducer 17β-estradiol upregulated heparanase mRNA [101]. Interestingly, estrogen stimulation of breast cancer cells at low concentrations induced higher expression levels of heparanase than high concentrations of estrogen [100]. During pregnancy, estrogen levels increase, which suggests pregnancy may protect against heparanase upregulation induced by low estrogen. Indeed, a clinical study found that the number of pregnancies correlates with a reduction in estrogen receptor-positive breast cancer risk [134]. Thus, it is possible that the induction of heparanase expression by low levels of estrogen in healthy breast tissue may contribute to the initiation of breast cancer.
The metabolites glucose and vitamin D also modulate heparanase expression [104,105,106,111]. Treatment of either podocytes in vitro or a rat model of proteinuria with vitamin D (1,25-D3) reduced heparanase mRNA expression [111]. Upon vitamin D binding, the vitamin D receptor directly bound to the heparanase promoter and blocked heparanase expression [111]. Furthermore, vitamin D deficient mice exhibited increased heparanase expression and activity [111]. This finding suggests that vitamin D may be a suitable treatment for proteinuria by targeting heparanase expression.
The induction of ROS has also been shown to regulate heparanase expression and secretion [106,109,110]. This suggests heparanase is regulated alongside other stress response genes. The mechanism of ROS-mediated heparanase upregulation has not been elucidated, however since ROS activates PI3K/AKT, MAPK signaling pathways, and NF-κB [135] which can upregulate heparanase, these pathways provide possible mechanisms of ROS-mediated heparanase upregulation.

2.7. Pathogens

An important role for heparanase during viral infection is emerging and has been recently reviewed [136,137]. Multiple viruses including Herpes Simplex Virus-1 (HSV-1), cytomegalovirus, and Dengue virus have been shown to hijack heparanase expression to facilitate infection (Table 2). By hijacking host pro-survival pathways and enabling viral egress, viruses exploit heparanase to their advantage. Other viruses, namely foot and mouth disease virus [138], respiratory syncytial virus [139], human papillomavirus [140], and hepatitis B virus [141], have been reported to require HS, the substrate of heparanase, for pathogenesis. This suggests they may also modulate heparanase expression to facilitate pathogenesis, but this is yet to be determined. Given the modulation of expression during infection, targeting heparanase during viral infection poses both diagnostic and therapeutic potential. The heparanase inhibitors heparin and the HS mimetic PI-88 were shown to inhibit poxvirus infection in vitro [142], but whether this was mediated via inhibiting heparanase activity was not directly tested. Further understanding of the modulation and role of heparanase during these infections is required to verify heparanase as a viable target.
Bacterial infection has also been shown to modulate heparanase expression. Fusobacterium nucleatum, which induces periodontal disease and can lead to oral carcinoma, was shown to increase heparanase expression upon infection in vitro [112]. Streptococcus pneumoniae infection in mice also increased heparanase protein levels [115]. Heparanase expression was also upregulated in mouse corneas following Pseudomonas aeruginosa (P. aeruginosa) infection [114], where the source of heparanase was from both infiltrating immune cells and the corneal epithelium. The gut pathogen Helicobacter pylori (H. pylori) also induced heparanase expression in gastric cancer cells and this was found to be dependent on MAPK signaling [113]. Furthermore, in a clinical cohort of gastric cancer patients with H. pylori infection, heparanase expression correlated with poor overall survival and relapse-free survival [113]. A negative correlation between heparanase expression and cancer survival has been shown many times previously [143,144,145]. In the context of chronic bacterial and viral infections that can contribute to tumorigenesis, heparanase expression during this inflammatory pre-tumorigenic phase is likely a driver of tumorigenesis. There are other bacterial pathogens such as P. aeruginosa and Staphylococcus aureus which also interact with and induce shedding of HSPGs to promote bacterial pathogenesis and are reviewed by Garcia and colleagues [146,147]. Given this, heparanase may also play a role in the pathogenesis of these bacterial infections. There may be many more bacterial species and viral strains which utilize heparanase for pathogenesis or induce a pro-inflammatory host response that drives heparanase expression, although this remains to be explored.

2.8. Therapies

Therapies such as chemotherapeutics, immune activators, and radiation have all been shown to modulate heparanase expression. The observation that heparanase can confer chemotherapeutic resistance in cancer cells (reviewed in [148]) led to the discovery that the chemotherapies bortezomib, carfilzomib, and doxorubicin can induce the upregulation of heparanase in vitro [76]. This upregulation of heparanase correlated with an increase in chemotherapeutic resistance through activation of the NF-κB pathway. This suggests that heparanase may be a potential target in overcoming chemoresistance. Indeed, later studies found that targeting heparanase can re-sensitize resistant tumor cells to chemotherapy and inhibit tumor cell growth in vitro and in vivo [149,150], presenting a promising approach to enhance chemotherapy response. One study identified in a colorectal cancer model that heparanase involvement in chemoresistance is 2-fold: (i) heparanase induces syndecan-1 shedding directly and (ii) heparanase induces upregulation of matrix metalloprotease-9 (MMP-9), which induces the binding of heparin-binding epithelial growth factor-like factor (HB-EGF) to epidermal growth factor (EGF) receptor (EGFR) and downstream MEK ERK signaling, leading to 5-Fluorouracil resistance [151]. These findings explain why tumor cells upregulate heparanase upon chemotherapy treatment and validate the use of heparanase as a chemotherapy-sensitizing target.
Given the role of heparanase in leukocyte functions, it is not surprising that compounds that modulate immune activation also modulate heparanase expression. PMA and ionomycin, potent inducers of leukocyte activation, can stimulate heparanase expression in lymphocytes [61], neutrophils, and platelets [50,124]. The viral RNA mimetic poly(I:C) can also upregulate heparanase in natural killer cells [50]. By upregulating heparanase during immune cell activation, these compounds enable heparanase-facilitated leukocyte functions such as cytokine production [152,153] and migration [50,51,153]. These findings add to the growing body of literature on the importance of heparanase in immune cell function, however, more work is needed to fully define its importance in immunity.
Radiation has also been shown to increase heparanase expression. UVB irradiation of human skin samples and cultured keratinocytes induced heparanase expression and activity [125] and rats with liver cirrhosis that received partial liver irradiation showed an upregulation of the heparanase proenzyme in liver and serum [126]. These findings suggest heparanase may be a useful biomarker when monitoring response to radiation. Furthermore, as with chemoresistance, and the recently identified survival signature associated with heparanase [154], heparanase upregulation may be another example of heparanase-mediated therapeutic resistance. The upregulation of heparanase upon treatment with these therapeutics may mean that combining with heparanase inhibitors could have synergistic benefits for anti-cancer treatments.

3. Regulation of Heparanase Enzymatic Activity: Proteolytic Activation and Natural Inhibitors

Heparanase is synthesized as an inactive proenzyme containing an 8 kDa and a 50 kDa subunit sequence joined by a linker sequence. This proenzyme then undergoes proteolytic processing by cathepsin L to remove the linker sequence and allow the heterodimerization of the two subunits to become an active enzyme [155,156]. Cathepsin L expression and consequent heparanase activation have been linked to viral infection [118,122] and pancreatitis [127]. Interestingly, in a model of acute pancreatitis, cathepsin L has also been shown to be regulated by heparanase, representing a self-sustaining loop which generates continuous heparanase activity [127]. In addition to cathepsin L, other proteases such as cysteine proteases, cathepsin B, D, S, and other aspartic proteases may also contribute to the activation of heparanase [155]. The existence of this proenzyme containing the linker sequence represents an efficient mechanism for rapid heparanase activation upon certain stimuli.
Heparanase enzymatic activity is also regulated by naturally occurring heparanase inhibitors. Although eosinophils produce heparanase, heparanase enzymatic activity in both resting and activated eosinophils is not detected. This is because eosinophils also express major basic protein which completely inhibits heparanase activity [157]. Two other eosinophil proteins, peroxidase and eosinophil cationic protein, also partially inhibit heparanase activity [157]. HS-interacting protein is also recognized as a natural endogenous heparanase inhibitor [158,159]. HS-interacting protein binds HS on the cell surface and ECM, thus blocking heparanase access. Heparanase-2, the inactive homolog to the active enzyme, can also bind HS, in fact, with higher affinity than the enzymatically active heparanase to indirectly inhibit activity. Heparanase-2 has also been shown to directly interact with heparanase, and thus inhibit heparanase activity directly [160]. Heparin is another well-described natural inhibitor of heparanase activity. Solely expressed by mast cells, this highly sulfated form of HS inhibits heparanase activity by binding directly to the enzyme’s active site [161,162,163]. Finally, heparanase enzymatic activity is affected by pH; enzymatic activity is limited to an acidic microenvironment, e.g., at sites of inflammation or in the core of solid tumors. The optimal pH for heparanase activity is 5.5 and no enzymatic activity is detected at a pH below 3.5 or above 7.0 [164,165,166].

4. Heparanase in Regulating Gene Expression, Protein Expression, and Protein Phosphorylation

4.1. Nuclear Heparanase Regulates Gene Transcription

In addition to its many well-recognized functions, heparanase can also regulate gene expression via multiple direct and indirect mechanisms (Figure 2). Heparanase can enter the nucleus to modify nuclear HS and even exert direct effects on gene transcription. Indeed, heparanase has been shown to enter the nucleus of myeloma cells and cleave nuclear HS on syndecan-1 [167]. Nuclear HS inhibits histone acetyltransferases (HATs), thereby inhibiting gene transcription [168]. By entering the nucleus and degrading nuclear syndecan-1, heparanase mediates HAT activation and transcription of genes associated with an aggressive tumor phenotype [168]. Conversely, nuclear heparanase has also been shown to bind non-specifically to DNA and compete for binding with NF-κB, thus preventing transcription of many NF-κB target genes and acting as a tumor suppressor [169]. Heparanase has also been identified in the nucleus of human glioma and breast cancer cell lines and in patient samples of squamous cell carcinoma [170] and adenocarcinoma [171]. Chromatin immunoprecipitation experiments revealed that heparanase is recruited to promoters and 5′ coding regions of microRNA genes miR-9 and miR-183 (previously implicated in cancer and epithelial-mesenchymal transition (EMT)) and other genes linked to development and differentiation pathways [172]. These studies suggest that in neoplastic cells, nuclear heparanase acts to drive tumor aggressiveness and heparanase localization in the nucleus can correlate with poor patient prognosis [171,173,174].
Furthermore, in human Jurkat T cells, heparanase controls nuclear histone H3 methylation patterns to regulate expression of the immune response genes CD69, IL-2, and IFN-γ [172]. Heparanase also contains two potential nuclear localization sequences, and enzymatically active heparanase has been found in the chromatin compartment of the nucleus, where it co-localizes with RNA polymerase II in T cells [172]. This nuclear heparanase positively controls the transcription of several genes in T cells important for immune function.

4.2. Heparanase Regulates Gene and Protein Expression and Protein Activation

The expression of heparanase is tightly regulated by many factors as described above. In contrast, heparanase itself is also involved in the regulation of different genes that contribute to a variety of physiological processes as well as disease settings. It has been reported that the expression of growth factors such as VEGF, HGF, bFGF, FGF-2, and transforming growth factor-β/β1 which play essential roles in EMT, bone formation, angiogenesis, tumor angiogenesis, and renal diseases, are regulated by heparanase. This effect of heparanase is observed in both in vivo and in vitro studies and is through either its enzymatic or non-enzymatic activities [76,168,175,176,177,178,179,180,181].
Heparanase can also alter the expression of EMT gene markers such as Slug, Snail, vimentin, α-SMA, Fibronectin, Collagen-1, Cathepsin-L, Endothelin-1, and E-cadherin as well as stem cell markers (CXCR4, OCT3/4, and NANOG) which further contribute to the pathological processes such as acute kidney disease and gastric adenocarcinoma [179,181,182]. In addition, considerable evidence supports a role for heparanase in regulating genes encoding pro-inflammatory cytokines, chemokines, and other proteins involving macrophage activation, function, and polarization, namely IL-1b, IL-6, IL-10, IL-12p53, TNF-α, MIP-2, toll-like receptor-2 (TLR-2), TLR-4, iNOS, c-Fos, CXCL-12, lysozyme 1, VEGF-A, and caspase-1. The expression of these molecules as well as the activation of macrophages play important roles in diseases such as colitis-associated tumorigenesis [131], ulcerative colitis [131], and acute kidney injury [182].
It is well-documented that heparanase overexpression occurs in most malignancies and is involved in tumor progression and prognosis. Here, heparanase contributes to the regulation of tumor-related processes, such as angiogenesis, inflammation, and tumor cell invasion and metastasis, reviewed in detail recently [64]. Heparanase has the ability to modify the expression of genes involved in these tumor-related processes including IL-17A [84], MCP-1 [183], MMPs [76,79,168,169,184], TNF-α [153,169], VEGF [76,168,175,177], and VEGF-C [185]. It is worth noting that heparanase also plays an important role in regulating the expression of many different inflammation-related genes such as IL-1β, IL-5, IL-6, IL-8, IL-10, IL-13, and vascular cell adhesion molecule 1 (VCAM-1) [51,120,152,183,186]. Moreover, the silencing or overexpression of heparanase also impacts the expression of other ECM-degrading enzyme MMPs such as MMP-2, MMP-9, MMP-14, and MMP-25, which affect migration of immune cells to inflammatory sites. Heparanase-induced upregulation or downregulation of these genes seems to vary depending on the disease [51,76,79,168,169,184]. The involvement of heparanase in the regulation of genes contributing to different physiological and pathological processes is listed in Table 3.
A recent study has also used transcriptomics to show that heparanase negatively regulates a number of genes involved in defense responses to viruses [201]. Following infection with HSV-1, differences in the transcriptomic landscape of wild-type and heparanase knock-out cells were observed. Heparanase knock-out cells were enriched in genes related to an antiviral and innate immune response (such as Interferon regulatory factors), while infected wild-type cells were enriched for genes involved in gene expression and processing, and hence viral replication. This suggests heparanase dampens the host’s antiviral defense response while simultaneously enhancing the virulence of HSV-1. As described above, heparanase is upregulated during infection with several types of viruses. Thus, heparanase upregulation and downstream gene regulation are likely a mechanism of viral pathogenicity. Genes involved in response to viral infection were not the only genes found to be modulated by heparanase in this study. Heparanase was also found to positively regulate genes involved in blood vessel development, cell-cell adhesion, inflammatory response, ECM reorganization, and leukocyte chemotaxis, and negatively regulate genes in pathways related to an antiviral defense response, regulation of viral genome replication, antigen processing and presentation, regulation of nuclease activity, and activation of an immune response [201]. Similarly, transcriptomic analysis has also been performed on heparanase-silenced melanoma cells [154]. This study found heparanase to negatively regulate genes relating to many pathways, including signaling, communication, response to cytokines, protein phosphorylation, cell adhesion, inflammatory response, and apoptotic processes. These two studies highlight the broad regulatory role heparanase plays in several cellular pathways.
As mentioned above, heparanase can directly and indirectly alter the expression of numerous genes. Since gene expression does not always correlate with protein expression, validating that expression changes occur at the protein level is important, and for heparanase-regulated genes, this is often the case. In addition to regulating EMT-related genes at the transcriptional level as mentioned above, heparanase also contributes to the expression of these genes at the protein level. This was demonstrated by the increased expression of α-SMA, fibronectin, and vimentin in transgenic mice over-expressing heparanase at both the mRNA and protein level [179]. There are many other examples of specific protein expression shown to be regulated by heparanase at the transcriptional level. Depletion of heparanase or employing heparanase inhibitors in either mouse models or cell lines resulted in the reduced expression of growth factors, cytokines, and other proteins such as bFGF, VEGF, HGF [76,176,177], CXCL2, TLR2 [153], and IL-17A [84]. These proteins play a key role in the progression of different tumor types. In the presence of heparanase, pro-inflammatory cytokines IL-6, IL-10, MCP-1, and TNF-α are elevated at the mRNA and protein level in both human and mouse immune cells in vivo. These cytokines are implicated in autoimmune diseases such as atherosclerosis and autoimmune encephalitis [199,200]. Heparanase, by modifying the levels of these cytokines, is therefore also involved in mediating these diseases. We have summarised the list of proteins and the processes and diseases involved that are influenced by heparanase in Table 3.
Other reports have also shown that the levels of specific proteins are altered as a result of heparanase expression, e.g., IL-4 [51,200], CXCL1 [183], and fibrinogen [198], however, mRNA expression levels have not been determined for these proteins. Whilst it is still unknown how heparanase regulates the expression of these proteins, based on the other examples listed above, it can be predicted that heparanase modulates expression of these proteins by altering the expression and secretion of signaling molecules (e.g., cytokines) that ultimately alter gene expression and consequently protein levels.

4.3. Protein Phosphorylation

Protein phosphorylation is an important biological process whereby many receptors and enzymes are activated or deactivated by phosphorylation or dephosphorylation, respectively. Several studies have demonstrated that heparanase can indirectly regulate protein phosphorylation (Figure 2). Akt, a member of AGC kinases, is associated with cellular signaling pathways related to cell proliferation, cell growth, cell survival, and metabolism [202]. Heparanase has been suggested to induce Akt phosphorylation in endothelial cells, macrophages, fibroblasts, and various tumor-derived cells [76,199,203,204,205]. It seems that Akt phosphorylation requires enzymatic activity of heparanase since blocking heparanase activity reduced levels of Akt phosphorylation [76,79]. Heparanase is also involved in the phosphorylation of ERK, another kinase involved in numerous cellular functions such as proliferation, survival, apoptosis, motility, transcription, metabolism, and differentiation [206]. Again, heparanase has been shown to enhance ERK phosphorylation levels in macrophages and myeloma cell lines through its enzymatic activity [76,153]. Increased ERK phosphorylation is also observed in neural stem and progenitor cells overexpressing heparanase during cell differentiation [207]. Heparanase is also implicated in mediating EGFR phosphorylation, where EGFR signaling is a key regulator of cell growth, cell migration, proliferation, and cell survival [208,209]. It is reported that overexpression of heparanase also stimulates the phosphorylation of EGFR in different tumor cell lines [205,209] and inhibiting heparanase expression results in the reduction of EGFR phosphorylation [79].
In addition to Akt, ERK, and EGFR, heparanase is suggested to mediate the phosphorylation of Signal Transducer and Activator Of Transcription (STAT) proteins including STAT3 and STAT5b. In a tumor setting, heparanase enhances the phosphorylation of STAT3 and STAT5b. Notably, increased cytoplasmic pSTAT3 is associated with larger tumor size and reduced patient survival in a cohort of patients with head and neck squamous cell carcinoma [205]. The increased STAT3 phosphorylation is eliminated in pancreatic cells isolated from mice treated with a heparanase inhibitor, which further strengthens the involvement of heparanase in STAT3 phosphorylation. Additionally, heparanase regulates the levels of phosphorylated Focal-adhesion kinase (FAK), SRC, and paxillin, adhesion molecules required for tumor cell cluster formation, the process that facilitates cancer metastasis [197]. In a mouse model of acute pancreatitis, heparanase overexpression resulted in elevated levels of IκB phosphorylation and correlated with increased TNF-α expression. A similar observation was noted for IL-6 and STAT3 phosphorylation which indicates the association of heparanase with the activation of key signaling pathways related to acute pancreatitis [127]. Furthermore, heparanase can also stimulate the phosphorylation of p65 NF-κB [131], p38, and JNK, which lead to the activation of NF-κB and the induction of cytokine expression in macrophages [210]. Proteins of which their phosphorylation state is regulated by heparanase are listed in Table 4.

5. Conclusions

Heparanase is widely considered a key player in several diseases including cancer, heart disease, and viral infection. Thus, the clinical inhibition of heparanase provides a potential method to treat these diseases. Understanding its intricate role in these diseases is key to designing effective treatments. This review highlighted the many molecular regulators of heparanase in different disease contexts. The array of different molecules, pathways and settings that regulate heparanase expression illustrate the diversity of heparanase expression and functions during disease. We also discuss how heparanase itself can regulate the expression of many downstream genes as well as the phosphorylation of proteins, and thus regulate the activity of several pathways, making heparanase a master regulator of several cellular processes in physiology and disease. Furthering our understanding of how heparanase itself is regulated, as well as the greater heparanase regulatory network, will help to develop treatments for heparanase-mediated diseases.

Author Contributions

Conceptualization, A.J.M., T.K.N. and M.D.H.; original draft preparation, reviewing and editing, A.J.M., T.K.N. and M.D.H. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Australian National Health and Medical Research Council, grant number APP471424.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef]
  2. Sarrazin, S.; Lamanna, W.C.; Esko, J.D. Heparan sulfate proteoglycans. Cold Spring Harb. Perspect. Biol. 2011, 3, a004952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Bernfield, M.; Götte, M.; Park, P.W.; Reizes, O.; Fitzgerald, M.L.; Lincecum, J.; Zako, M. Functions of Cell Surface Heparan Sulfate Proteoglycans. Annu. Rev. Biochem. 1999, 68, 729–777. [Google Scholar] [CrossRef] [PubMed]
  4. Stewart, M.D.; Sanderson, R.D. Heparan sulfate in the nucleus and its control of cellular functions. Matrix Biol. 2014, 35, 56–59. [Google Scholar] [CrossRef] [PubMed]
  5. Ori, A.; Wilkinson, M.C.; Fernig, D.G. A systems biology approach for the investigation of the heparin/heparan sulfate interactome. J. Biol. Chem. 2011, 286, 19892–19904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Kreuger, J.; Jemth, P.; Sanders-Lindberg, E.; Eliahu, L.; Ron, D.; Basilico, C.; Salmivirta, M.; Lindahl, U. Fibroblast growth factors share binding sites in heparan sulphate. Biochem. J. 2005, 389, 145–150. [Google Scholar] [CrossRef] [Green Version]
  7. Sekiguchi, K.; Hakomori, S.; Funahashi, M.; Matsumoto, I.; Seno, N. Binding of fibronectin and its proteolytic fragments to glycosaminoglycans. Exposure of cryptic glycosaminoglycan-binding domains upon limited proteolysis. J. Biol. Chem. 1983, 258, 14359–14365. [Google Scholar] [CrossRef]
  8. Ogamo, O.; Nagai, A.; Nagasawa, N. Binding of heparin fractions and other polysulfated polysaccharides to plasma fibronectin: Effects of molecular size and degree of sulfation of polysaccharides. BBA Gen. Subj. 1985, 841, 30–41. [Google Scholar] [CrossRef]
  9. Dyer, D.P.; Salanga, C.L.; Volkman, B.F.; Kawamura, T.; Handel, T.M. The dependence of chemokine-glycosaminoglycan interactions on chemokine oligomerization. Glycobiology 2015, 26, 312–326. [Google Scholar] [CrossRef]
  10. Wu, Z.L.; Zhang, L.; Yabe, T.; Kuberan, B.; Beeler, D.L.; Love, A.; Rosenberg, R.D. The involvement of heparan sulfate (HS) in FGF1/HS/FGFR1 signaling complex. J. Biol. Chem. 2003, 278, 17121–17129. [Google Scholar] [CrossRef] [Green Version]
  11. Jakobsson, L.; Kreuger, J.; Holmborn, K.; Lundin, L.; Eriksson, I.; Kjellén, L.; Claesson-Welsh, L. Heparan Sulfate in trans Potentiates VEGFR-Mediated Angiogenesis. Dev. Cell 2006, 10, 625–634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kumar, A.V.; Katakam, S.K.; Urbanowitz, A.-K.; Gotte, M. Heparan sulphate as a regulator of leukocyte recruitment in inflammation. Curr. Protein Pept. Sci. 2015, 16, 77–86. [Google Scholar] [CrossRef] [PubMed]
  13. Faye, C.; Chautard, E.; Olsen, B.R.; Ricard-Blum, S. The first draft of the endostatin interaction network. J. Biol. Chem. 2009, 284, 22041–22047. [Google Scholar] [CrossRef] [Green Version]
  14. Gralle, M.; Botelho, M.G.; Wouters, F.S. Neuroprotective secreted amyloid precursor protein acts by disrupting amyloid precursor protein dimers. J. Biol. Chem. 2009, 284, 15016–15025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Ishitsuka, R.; Kojima, K.; Utsumi, H.; Ogawa, H.; Matsumoto, I. Glycosaminoglycan Binding Properties of Annexin IV, V, and VI. J. Biol. Chem. 1998, 273, 9935–9941. [Google Scholar] [CrossRef] [Green Version]
  16. Moscatelli, D. High and low affinity binding sites for basic fibroblast growth factor on cultured cells: Absence of a role for low affinity binding in the stimulation of plasminogen activator production by bovine capillary endothelial cells. J. Cell. Physiol. 1987, 131, 123–130. [Google Scholar] [CrossRef]
  17. Bashkin, P.; Doctrow, S.; Klagsbrun, M.; Svahn, C.M.; Folkman, J.; Vlodavsky, I. Basic Fibroblast Growth Factor Binds to Subendothelial Extracellular Matrix and Is Released by Heparitinase and Heparin-like Molecules. Biochemistry 1989, 28, 1737–1743. [Google Scholar] [CrossRef]
  18. Rapraeger, A.C.; Krufka, A.; Olwin, B.B. Requirement of heparan sulfate for bFGF-mediated fibroblast growth and myoblast differentiation. Science 1991, 252, 1705–1708. [Google Scholar] [CrossRef] [Green Version]
  19. Andres, J.L.; DeFalcis, D.; Noda, M.; Massague, J. Binding of two growth factor families to separate domains of the proteoglycan betaglycan. J. Biol. Chem. 1992, 267, 5927–5930. [Google Scholar] [CrossRef]
  20. Stamatoglou, S.C.; Keller, J.M. Correlation between cell substrate attachment in vitro and cell surface heparan sulfate affinity for fibronectin and collagen. J. Cell Biol. 1983, 96, 1820–1823. [Google Scholar] [CrossRef]
  21. LeBaron, R.G.; Hook, A.; Esko, J.D.; Gay, S.; Hook, M. Binding of heparan sulfate to type V collagen. A mechanism of cell-substrate adhesion. J. Biol. Chem. 1989, 264, 7950–7956. [Google Scholar] [CrossRef]
  22. Ricard-Blum, S.; Beraud, M.; Raynal, N.; Farndale, R.W.; Ruggiero, F. Structural requirements for heparin/heparan sulfate binding to type V collagen. J. Biol. Chem. 2006, 281, 25195–25204. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Battaglia, C.; Mayer, U.; Aumailley, M.; Timpl, R. Basement-membrane heparan sulfate proteoglycan binds to laminin by its heparan sulfate chains and to nidogen by sites in the protein core. Eur. J. Biochem. 1992, 208, 359–366. [Google Scholar] [CrossRef] [PubMed]
  24. Li, S.; Pettersson, U.S.; Hoorelbeke, B.; Kolaczkowska, E.; Schelfhout, K.; Martens, E.; Kubes, P.; van Damme, J.; Phillipson, M.; Opdenakker, G. Interference with Glycosaminoglycan-Chemokine Interactions with a Probe to Alter Leukocyte Recruitment and Inflammation In Vivo. PLoS ONE 2014, 9, e104107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Luster, A.D.; Greenberg, S.M.; Leder, P. The IP-10 chemokine binds to a specific cell surface heparan sulfate site shared with platelet factor 4 and inhibits endothelial cell proliferation. J. Exp. Med. 1995, 182, 219–231. [Google Scholar] [CrossRef] [Green Version]
  26. Monneau, Y.R.; Luo, L.; Sankaranarayanan, N.V.; Nagarajan, B.; Vivès, R.R.; Baleux, F.; Desai, U.R.; Arenzana-Seidedos, F.; Lortat-Jacob, H. Solution structure of CXCL13 and heparan sulfate binding show that GAG binding site and cellular signalling rely on distinct domains. Open Biol. 2017, 7, 170133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Karumanchi, S.A.; Jha, V.; Ramchandran, R.; Karihaloo, A.; Tsiokas, L.; Chan, B.; Dhanabal, M.; Hanai, J.I.; Venkataraman, G.; Shriver, Z.; et al. Cell surface glypicans are low-affinity endostatin receptors. Mol. Cell 2001, 7, 811–822. [Google Scholar] [CrossRef] [Green Version]
  28. Blackhall, F.H.; Merry, C.L.R.; Lyon, M.; Jayson, G.C.; Folkman, J.; Javaherian, K.; Gallagher, J.T. Binding of endostatin to endothelial heparan sulphate shows a differential requirement for specific sulphates. Biochem. J. 2003, 375, 131–139. [Google Scholar] [CrossRef] [Green Version]
  29. Loo, B.-M.; Kreuger, J.; Jalkanen, M.; Lindahl, U.; Salmivirta, M. Binding of Heparin/Heparan Sulfate to Fibroblast Growth Factor Receptor 4. J. Biol. Chem. 2001, 276, 16868–16876. [Google Scholar] [CrossRef] [Green Version]
  30. Lyon, M.; Deakin, J.A.; Mizuno, K.; Nakamura, T.; Gallagher, J.T. Interaction of hepatocyte growth factor with heparan sulfate. Elucidation of the major heparan sulfate structural determinants. J. Biol. Chem. 1994, 269, 11216–11223. [Google Scholar] [CrossRef]
  31. Jones, A.L.; Hulett, M.D.; Parish, C.R. Histidine-rich glycoprotein binds to cell-surface heparan sulfate via its N-terminal domain following Zn2+ chelation. J. Biol. Chem. 2004, 279, 30114–30122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Xu, D.; Young, J.; Song, D.; Esko, J.D. Heparan sulfate is essential for high mobility group protein 1 (HMGB1) signaling by the receptor for advanced glycation end products (RAGE). J. Biol. Chem. 2011, 286, 41736–41744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Gordts, P.L.S.M.; Foley, E.M.; Lawrence, R.; Sinha, R.; Lameda-Diaz, C.; Deng, L.; Nock, R.; Glass, C.K.; Erbilgin, A.; Lusis, A.J.; et al. Reducing Macrophage Proteoglycan Sulfation Increases Atherosclerosis and Obesity through Enhanced Type I Interferon Signaling. Cell Metab. 2014, 20, 813–826. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Witt, D.P.; Lander, A.D. Differential binding of chemokines to glycosaminoglycan subpopulations. Curr. Biol. 1994, 4, 394–400. [Google Scholar] [CrossRef] [Green Version]
  35. Norgard-Sumnicht, K.E.; Varki, N.M.; Varki, A. Calcium-dependent heparin-like ligands for L-selectin in nonlymphoid endothelial cells. Science 1993, 261, 480–483. [Google Scholar] [CrossRef] [Green Version]
  36. Koenig, A.; Norgard-Sumnicht, K.; Linhardt, R.; Varki, A. Differential interactions of heparin and heparan sulfate glycosaminoglycans with the selectins: Implications for the use of unfractionated and low molecular weight heparins as therapeutic agents. J. Clin. Investig. 1998, 101, 877–889. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Chakravarty, L.; Rogers, L.; Quach, T.; Breckenridge, S.; Kolattukudy, P.E. Lysine 58 and histidine 66 at the C-terminal α-helix of monocyte chemoattractant protein-1 are essential for glycosaminoglycan binding. J. Biol. Chem. 1998, 273, 29641–29647. [Google Scholar] [CrossRef] [Green Version]
  38. Pasqualon, T.; Lue, H.; Groening, S.; Pruessmeyer, J.; Jahr, H.; Denecke, B.; Bernhagen, J.; Ludwig, A. Cell surface syndecan-contributes to binding and function of macrophage migration inhibitory factor (MIF) on epithelial tumor cells. Biochim. Biophys. Acta Mol. Cell Res. 2016, 1863, 717–726. [Google Scholar] [CrossRef]
  39. Stringer, S.E.; Forster, M.J.; Mulloy, B.; Bishop, C.R.; Graham, G.J.; John, T.; Gallagher, J.T. Characterization of the binding site on heparan sulfate for macrophage inflammatory protein 1α. Blood 2002, 100, 1543–1550. [Google Scholar] [CrossRef] [Green Version]
  40. Zilka, A.; Landau, G.; Hershkovitz, O.; Bloushtain, N.; Bar-Ilan, A.; Benchetrit, F.; Fima, E.; van Kuppevelt, T.H.; Gallagher, J.T.; Elgavish, S.; et al. Characterization of the heparin/heparan sulfate binding site of the natural cytotoxicity receptor NKp46. Biochemistry 2005, 44, 14477–14485. [Google Scholar] [CrossRef]
  41. Abramsson, A.; Kurup, S.; Busse, M.; Yamada, S.; Lindblom, P.; Schallmeiner, E.; Stenzel, D.; Sauvaget, D.; Ledin, J.; Ringvall, M.; et al. Defective N-sulfation of heparan sulfate proteoglycans limits PDGF-BB binding and pericyte recruitment in vascular development. Genes Dev. 2007, 21, 316–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Lustig, F.; Hoebeke, J.; Östergren-Lundèn, G.; Velge-Roussel, F.; Bondjers, G.; Olsson, U.; Rüetschi, U.; Fager, G. Alternative splicing determines the binding of platelet-derived growth factor (PDGF-AA) to glycosaminoglycans. Biochemistry 1996, 35, 12077–12085. [Google Scholar] [CrossRef] [PubMed]
  43. Xu, D.; Young, J.H.; Krahn, J.M.; Song, D.; Corbett, K.D.; Chazin, W.J.; Pedersen, L.C.; Esko, J.D. Stable RAGE-Heparan Sulfate Complexes Are Essential for Signal Transduction. ACS Chem. Biol. 2013, 8, 1611–1620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Øynebråten, I.; Barois, N.; Bergeland, T.; Küchler, A.M.; Bakke, O.; Haraldsen, G. Oligomerized, filamentous surface presentation of RANTES/CCL5 on vascular endothelial cells. Sci. Rep. 2015, 5, 9261. [Google Scholar] [CrossRef] [Green Version]
  45. Aricescu, A.R.; McKinnell, I.W.; Halfter, W.; Stoker, A.W. Heparan Sulfate Proteoglycans Are Ligands for Receptor Protein Tyrosine Phosphatase. Mol. Cell. Biol. 2002, 22, 1881–1892. [Google Scholar] [CrossRef] [Green Version]
  46. Netelenbos, T.; van den Born, J.; Kessler, F.L.; Zweegman, S.; Merle, P.A.; van Oostveen, J.W.; Zwaginga, J.J.; Huijgens, P.C.; Dräger, A.M. Proteoglycans on bone marrow endothelial cells bind and present SDF-1 towards hematopoietic progenitor cells. Leukemia 2003, 17, 175–184. [Google Scholar] [CrossRef] [Green Version]
  47. Houck, K.A.; Leung, D.W.; Rowland, A.M.; Winer, J.; Ferrara, N. Dual regulation of vascular endothelial growth factor bioavailability by genetic and proteolytic mechanisms. J. Biol. Chem. 1992, 267, 26031–26037. [Google Scholar] [CrossRef]
  48. Wilson, J.C.; Laloo, A.E.; Singh, S.; Ferro, V. 1H NMR spectroscopic studies establish that heparanase is a retaining glycosidase. Biochem. Biophys. Res. Commun. 2014, 443, 185–188. [Google Scholar] [CrossRef]
  49. Zcharia, E.; Zilka, R.; Yaar, A.; Yacoby-Zeevi, O.; Zetser, A.; Metzger, S.; Sarid, R.; Naggi, A.; Casu, B.; Ilan, N.; et al. Heparanase accelerates wound angiogenesis and wound healing in mouse and rat models. FASEB J. 2005, 19, 211–221. [Google Scholar] [CrossRef]
  50. Putz, E.M.; Mayfosh, A.J.; Kos, K.; Barkauskas, D.S.; Nakamura, K.; Town, L.; Goodall, K.J.; Yee, D.Y.; Poon, I.K.H.; Baschuk, N.; et al. NK cell heparanase controls tumor invasion and immune surveillance. J. Clin. Investig. 2017, 127, 2777–2788. [Google Scholar] [CrossRef]
  51. Poon, I.K.H.; Goodall, K.J.; Phipps, S.; Chow, J.D.Y.; Pagler, E.B.; Andrews, D.M.; Conlan, C.L.; Ryan, G.F.; White, J.A.; Wong, M.K.L.; et al. Mice deficient in heparanase exhibit impaired dendritic cell migration and reduced airway inflammation. Eur. J. Immunol. 2014, 44, 1016–1030. [Google Scholar] [CrossRef] [PubMed]
  52. Lider, O.; Mekori, Y.A.; Miller, T.; Bar-Tana, R.; Vlodavsky, I.; Baharav, E.; Cohen, I.R.; Naparstek, Y. Inhibition of T lymphocyte heparanase by heparin prevents T cell migration and T cell-mediated immunity. Eur. J. Immunol. 1990, 20, 493–499. [Google Scholar] [CrossRef]
  53. Sasaki, N.; Higashi, N.; Taka, T.; Nakajima, M.; Irimura, T. Cell Surface Localization of Heparanase on Macrophages Regulates Degradation of Extracellular Matrix Heparan Sulfate. J. Immunol. 2004, 172, 3830–3835. [Google Scholar] [CrossRef] [Green Version]
  54. Shteper, P.J.; Zcharia, E.; Ashhab, Y.; Peretz, T.; Vlodavsky, I.; Ben-Yehuda, D. Role of promoter methylation in regulation of the mammalian heparanase gene. Oncogene 2003, 22, 7737–7749. [Google Scholar] [CrossRef] [Green Version]
  55. Jiao, F.; Bai, S.; Ma, Y.; Yan, Z.; Yue, Z.; Yu, Y.; Wang, X.; Wang, J. DNA Methylation of Heparanase Promoter Influences Its Expression and Associated with the Progression of Human Breast Cancer. PLoS ONE 2014, 9, e92190. [Google Scholar] [CrossRef]
  56. Ostrovsky, O.; Korostishevsky, M.; Shafat, I.; Mayorov, M.; Ilan, N.; Vlodavsky, I.; Nagler, A. Inverse correlation between HPSE gene single nucleotide polymorphisms and heparanase expression: Possibility of multiple levels of heparanase regulation. J. Leukoc. Biol. 2009, 86, 445–455. [Google Scholar] [CrossRef] [Green Version]
  57. Ostrovsky, O.; Korostishevsky, M.; Levite, I.; Leiba, M.; Galski, H.; Vlodavsky, I.; Nagler, A. Association of heparanase gene (HPSE) single nucleotide polymorphisms with hematological malignancies. Leukemia 2007, 21, 2296–2303. [Google Scholar] [CrossRef] [PubMed]
  58. Hulett, M.D.; Freeman, C.; Hamdorf, B.J.; Baker, R.T.; Harris, M.J.; Parish, C.R. Cloning of mammalian heparanase, an important enzyme in tumor invasion and metastasis. Nat. Med. 1999, 5, 803–809. [Google Scholar] [CrossRef] [PubMed]
  59. Rabelink, T.J.; van den Berg, B.M.; Garsen, M.; Wang, G.; Elkin, M.; van der Vlag, J. Heparanase: Roles in cell survival, extracellular matrix remodelling and the development of kidney disease. Nat. Rev. Nephrol. 2017, 13, 201–212. [Google Scholar] [CrossRef]
  60. Uhlen, M.; Fagerberg, L.; Hallstrom, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, A.; Kampf, C.; Sjostedt, E.; Asplund, A.; et al. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
  61. De Mestre, A.M.; Soe-Htwe, T.; Sutcliffe, E.L.; Rao, S.; Pagler, E.B.; Hornby, J.R.; Hulett, M.D. Regulation of mouse Heparanase gene expression in T lymphocytes and tumor cells. Immunol. Cell Biol. 2007, 85, 205–214. [Google Scholar] [CrossRef] [PubMed]
  62. De Mestre, A.M.; Staykova, M.A.; Hornby, J.R.; Willenborg, D.O.; Hulett, M.D. Expression of the heparan sulfate-degrading enzyme heparanase is induced in infiltrating CD4 + T cells in experimental autoimmune encephalomyelitis and regulated at the level of transcription by early growth response gene. J. Leukoc. Biol. 2007, 82, 1289–1300. [Google Scholar] [CrossRef] [Green Version]
  63. Ostrovsky, O.; Shimoni, A.; Baryakh, P.; Morgulis, Y.; Mayorov, M.; Beider, K.; Shteingauz, A.; Ilan, N.; Vlodavsky, I.; Nagler, A. Modification of heparanase gene expression in response to conditioning and LPS treatment: Strong correlation to rs4693608 SNP. J. Leukoc. Biol. 2014, 95, 677–688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Jayatilleke, K.M.; Hulett, M.D. Heparanase and the hallmarks of cancer. J. Transl. Med. 2020, 18, 1–25. [Google Scholar] [CrossRef]
  65. Tang, B.; Xie, R.; Qin, Y.; Xiao, Y.; Yong, X.; Zheng, L.; Dong, H.; Yang, S.-M. Human telomerase reverse transcriptase (hTERT) promotes gastric cancer invasion through cooperating with c-Myc to upregulate heparanase expression. Oncotarget 2015, 7, 11364–11379. [Google Scholar] [CrossRef] [PubMed]
  66. De Mestre, A.M.; Khachigian, L.M.; Santiago, F.S.; Staykova, M.A.; Hulett, M.D. Regulation of Inducible Heparanase Gene Transcription in Activated T Cells by Early Growth Response 1. J. Biol. Chem. 2003, 278, 50377–50385. [Google Scholar] [CrossRef] [Green Version]
  67. De Mestre, A.M.; Rao, S.; Hornby, J.R.; Soe-Htwe, T.; Khachigian, L.M.; Hulett, M.D. Early growth response gene 1 (EGR1) regulates heparanase gene transcription in tumor cells. J. Biol. Chem. 2005, 280, 35136–35147. [Google Scholar] [CrossRef] [Green Version]
  68. Gil, N.; Goldberg, R.; Neuman, T.; Garsen, M.; Zcharia, E.; Rubinstein, A.M.; van Kuppevelt, T.; Meirovitz, A.; Pisano, C.; Li, J.P.; et al. Heparanase is essential for the development of diabetic nephropathy in mice. Diabetes 2012, 61, 208–216. [Google Scholar] [CrossRef] [Green Version]
  69. Lu, W.C.; Liu, Y.N.; Kang, B.B.; Chen, J.H. Trans-activation of heparanase promoter by ETS transcription factors. Oncogene 2003, 22, 919–923. [Google Scholar] [CrossRef] [Green Version]
  70. Jiang, P.; Kumar, A.; Parrillo, J.E.; Dempsey, L.A.; Platt, J.L.; Prinz, R.A.; Xu, X. Cloning and characterization of the human heparanase-1 (HPR1) gene promoter. Role of GA-binding protein and Sp1 in regulating HPR1 basal promoter activity. J. Biol. Chem. 2002, 277, 8989–8998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Rao, G.; Liu, D.; Xing, M.; Tauler, J.; Prinz, R.A.; Xu, X. Induction of Heparanase-1 Expression by Mutant B-Raf Kinase: Role of GA Binding Protein in Heparanase-1 Promoter Activation. Neoplasia 2010, 12, 946–956. [Google Scholar] [CrossRef] [Green Version]
  72. Andela, V.B.; Schwarz, E.M.; Puzas, J.E.; O’Keefe, R.J.; Rosier, R.N. Tumor metastasis and the reciprocal regulation of prometastatic and antimetastatic factors by nuclear factor κB. Cancer Res. 2000, 60, 6557–6562. [Google Scholar] [PubMed]
  73. Rops, A.L.; van den Hoven, M.J.; Baselmans, M.M.; Lensen, J.F.; Wijnhoven, T.J.; van den Heuvel, L.P.; van Kuppevelt, T.H.; Berden, J.H.; van der Vlag, J. Heparan sulfate domains on cultured activated glomerular endothelial cells mediate leukocyte trafficking. Kidney Int. 2008, 73, 52–62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Wu, W.; Pan, C.; Meng, K.; Zhao, L.; Du, L.; Liu, Q.; Lin, R. Hypoxia activates heparanase expression in an NF-kappaB dependent manner. Oncol. Rep. 2010, 23, 255–261. [Google Scholar] [CrossRef]
  75. Hadigal, S.R.; Agelidis, A.M.; Karasneh, G.A.; Antoine, T.E.; Yakoub, A.M.; Ramani, V.C.; Djalilian, A.R.; Sanderson, R.D.; Shukla, D. Heparanase is a host enzyme required for herpes simplex virus-1 release from cells. Nat. Commun. 2015, 6, 6985. [Google Scholar] [CrossRef] [Green Version]
  76. Ramani, V.C.; Vlodavsky, I.; Ng, M.; Zhang, Y.; Barbieri, P.; Noseda, A.; Sanderson, R.D. Chemotherapy induces expression and release of heparanase leading to changes associated with an aggressive tumor phenotype. Matrix Biol. 2016, 55, 22–34. [Google Scholar] [CrossRef]
  77. Baraz, L.; Haupt, Y.; Elkin, M.; Peretz, T.; Vlodavsky, I. Tumor suppressor p53 regulates heparanase gene expression. Oncogene 2006, 25, 3939–3947. [Google Scholar] [CrossRef] [Green Version]
  78. Karamanou, K.; Franchi, M.; Proult, I.; Rivet, R.; Vynios, D.; Brézillon, S. Lumican Inhibits In Vivo Melanoma Metastasis by Altering Matrix-Effectors and Invadopodia Markers. Cells 2021, 10, 841. [Google Scholar] [CrossRef] [PubMed]
  79. Zhang, L.; Sullivan, P.S.; Goodman, J.C.; Gunaratne, P.H.; Marchetti, D. MicroRNA-1258 suppresses breast cancer brain metastasis by targeting heparanase. Cancer Res. 2011, 71, 645–654. [Google Scholar] [CrossRef] [Green Version]
  80. Rodrigues, D.M., Jr.; Pelarin, M.F.d.A.; Nader, H.B.; Vettore, A.L.; Pinhal, M.A.S. MicroRNA-1252-5p associated with extracellular vesicles enhances bortezomib sensitivity in multiple myeloma cells by targeting heparanase. OncoTargets Ther. 2021, 14, 455–467. [Google Scholar] [CrossRef]
  81. Edovitsky, E.; Lerner, I.; Zcharia, E.; Peretz, T.; Vlodavsky, I.; Elkin, M. Role of endothelial heparanase in delayed-type hypersensitivity. Blood 2006, 107, 3609–3616. [Google Scholar] [CrossRef]
  82. Chen, G.; Wang, D.; Vikramadithyan, R.; Yagyu, H.; Saxena, U.; Pillarisetti, S.; Goldberg, I.J. Inflammatory Cytokines and Fatty Acids Regulate Endothelial Cell Heparanase Expression. Biochemistry 2004, 43, 4971–4977. [Google Scholar] [CrossRef]
  83. El-Nadi, M.; Hassan, H.; Saleh, M.E.; Nassar, E.; Ismail, Y.M.; Amer, M.; Greve, B.; Götte, M.; El-Shinawi, M.; Ibrahim, S.A. Induction of heparanase via IL-10 correlates with a high infiltration of CD163+ M2-type tumor-associated macrophages in inflammatory breast carcinomas. Matrix Biol. Plus 2020, 6–7, 6–7. [Google Scholar] [CrossRef]
  84. Lv, Q.; Wu, K.; Liu, F.; Wu, W.; Chen, Y.; Zhang, W. Interleukin-17A and heparanase promote angiogenesis and cell proliferation and invasion in cervical cancer. Int. J. Oncol. 2018, 53, 1809–1817. [Google Scholar] [CrossRef] [Green Version]
  85. Boels, M.G.S.; Koudijs, A.; Avramut, M.C.; Sol, W.M.P.J.; Wang, G.; van Oeveren-Rietdijk, A.M.; van Zonneveld, A.J.; de Boer, H.C.; van der Vlag, J.; van Kooten, C.; et al. Systemic Monocyte Chemotactic Protein-1 Inhibition Modifies Renal Macrophages and Restores Glomerular Endothelial Glycocalyx and Barrier Function in Diabetic Nephropathy. Am. J. Pathol. 2017, 187, 2430–2440. [Google Scholar] [CrossRef] [Green Version]
  86. Tang, X.-D.; Liang, G.-P.; Li, C.; Wan, Y.; Chen, T.; Chen, L.; Yu, S.-T.; Xiong, Z.; Fang, D.-C.; Wang, G.-Z.; et al. Cytotoxic T lymphocyte epitopes from human heparanase can elicit a potent anti-tumor immune response in mice. Cancer Immunol. Immunother. 2010, 59, 1041–1047. [Google Scholar] [CrossRef]
  87. Schmidt, E.P.; Yang, Y.; Janssen, W.J.; Gandjeva, A.; Perez, M.J.; Barthel, L.; Zemans, R.L.; Bowman, J.C.; Koyanagi, D.E.; Yunt, Z.X.; et al. The pulmonary endothelial glycocalyx regulates neutrophil adhesion and lung injury during experimental sepsis. Nat. Med. 2012, 18, 1217–1223. [Google Scholar] [CrossRef] [Green Version]
  88. Secchi, M.F.; Crescenzi, M.; Masola, V.; Russo, F.P.; Floreani, A.; Onisto, M. Heparanase and macrophage interplay in the onset of liver fibrosis. Sci. Rep. 2017, 7, 14956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Sasaki, M.; Ito, T.; Kashima, M.; Fukui, S.; Izumiyama, N.; Watanabe, A.; Sano, M.; Fujiwara, Y.; Miura, M. Erythromycin and clarithromycin modulation of growth factor-induced expression of heparanase mRNA on human lung cancer cells in vitro. Mediat. Inflamm. 2001, 10, 259–267. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Hao, N.B.; Tang, B.; Wang, G.Z.; Xie, R.; Hu, C.J.; Wang, S.M.; Wu, Y.Y.; Liu, E.; Xie, X.; Yang, S.M. Hepatocyte growth factor (HGF) upregulates heparanase expression via the PI3K/Akt/NF-κB signaling pathway for gastric cancer metastasis. Cancer Lett. 2015, 361, 57–66. [Google Scholar] [CrossRef] [PubMed]
  91. Suvannasankha, A.; Tompkins, D.R.; Edwards, D.F.; Petyaykina, K.V.; Crean, C.D.; Fournier, P.G.; Parker, J.M.; Sandusky, G.E.; Ichikawa, S.; Imel, E.A.; et al. FGF23 is elevated in multiple myeloma and increases heparanase expression by tumor cells. Oncotarget 2015, 6, 19647–19660. [Google Scholar] [CrossRef] [Green Version]
  92. Luan, Q.; Sun, J.; Li, C.; Zhang, G.; Lv, Y.; Wang, G.; Li, C.; Ma, C.; Gao, T. Mutual enhancement between heparanase and vascular endothelial growth factor: A novel mechanism for melanoma progression. Cancer Lett. 2011, 308, 100–111. [Google Scholar] [CrossRef] [PubMed]
  93. Stoler-Barak, L.; Petrovich, E.; Aychek, T.; Gurevich, I.; Tal, O.; Hatzav, M.; Ilan, N.; Feigelson, S.W.; Shakhar, G.; Vlodavsky, I.; et al. Heparanase of murine effector lymphocytes and neutrophils is not required for their diapedesis into sites of inflammation. FASEB J. 2015, 29, 2010–2021. [Google Scholar] [CrossRef]
  94. Melo, C.M.; Nader, H.B.; Justo, G.Z.; Pinhal, M.A.S. Heparanase modulation by Wingless/INT (Wnt). Mol. Biol. Rep. 2021, 48. [Google Scholar] [CrossRef]
  95. An, X.F.; Zhou, L.; Jiang, P.J.; Yan, M.; Huang, Y.J.; Zhang, S.N.; Niu, Y.F.; Ten, S.C.; Yu, J.Y. Advanced glycation end-products induce heparanase expression in endothelial cells by the receptor for advanced glycation end products and through activation of the FOXO4 transcription factor. Mol. Cell. Biochem. 2011, 354, 47–55. [Google Scholar] [CrossRef]
  96. Naparstek, Y.; Cohen, I.R.; Fuks, Z.; Vlodavsky, I. Activated T lymphocytes produce a matrix-degrading heparan sulphate endoglycosidase. Nature 1984, 310, 241–244. [Google Scholar] [CrossRef]
  97. Fridman, R.; Lider, O.; Naparstek, Y.; Fuks, Z.; Vlodavsky, I.; Cohen, I.R. Soluble antigen induces T lymphocytes to secrete an endoglycosidase that degrades the heparan sulfate moiety of subendothelial extracellular matrix. J. Cell. Physiol. 1987, 130, 85–92. [Google Scholar] [CrossRef] [PubMed]
  98. Vlodavsky, I.; Eldor, A.; Haimovitz-Friedman, A.; Matzner, Y.; Lider, O.; Naparstek, Y.; Cohen, I.R.; Fuks, Z.; Ishai-Michaeli, R.; Lider, O.; et al. Expression of heparanase by platelets and circulating cells of the immune system: Possible involvement in diapedesis and extravasation. Invasion Metastasis 1992, 12, 112–127. [Google Scholar] [PubMed]
  99. Garsen, M.; Rops, A.L.; Li, J.; van Beneden, K.; van den Branden, C.; Berden, J.H.M.; Rabelink, T.J.; van der Vlag, J. Endothelial Nitric Oxide Synthase Prevents Heparanase Induction and the Development of Proteinuria. PLoS ONE 2016, 11, e0160894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Elkin, M.; Cohen, I.; Zcharia, E.; Orgel, A.; Guatta-Rangini, Z.; Peretz, T.; Vlodavsky, I.; Kleinman, H.K. Regulation of Heparanase Gene Expression by Estrogen in Breast Cancer. Cancer Res. 2003, 63, 8821–8826. [Google Scholar] [PubMed]
  101. Singsuksawat, E.; Thuwajit, C.; Charngkaew, K.; Thuwajit, P. Increased ETV4 expression correlates with estrogen-enhanced proliferation and invasiveness of cholangiocarcinoma cells. Cancer Cell Int. 2018, 18, 25. [Google Scholar] [CrossRef] [PubMed]
  102. Cohen, I.; Maly, B.; Simon, I.; Meirovitz, A.; Pikarsky, E.; Zcharia, E.; Peretz, T.; Vlodavsky, I.; Elkin, M. Tamoxifen induces heparanase expression in estrogen receptor—Positive breast cancer. Clin. Cancer Res. 2007, 13, 4069–4077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Hermano, E.; Goldberg, R.; Rubinstein, A.M.; Sonnenblick, A.; Maly, B.; Nahmias, D.; Li, J.P.; Bakker, M.A.H.; van der Vlag, J.; Vlodavsky, I.; et al. Heparanase accelerates obesity-associated breast cancer progression. Cancer Res. 2019, 79, 5342–5354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Wang, F.; Wang, Y.; Kim, M.S.; Puthanveetil, P.; Ghosh, S.; Luciani, D.S.; Johnson, J.D.; Abrahani, A.; Rodrigues, B. Glucose-induced endothelial heparanase secretion requires cortical and stress actin reorganization. Cardiovasc. Res. 2010, 87, 127–136. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Ma, P.; Luo, Y.; Zhu, X.; Li, T.; Hu, J.; Tang, S. Retinal heparanase expression in streptozotocin-induced diabetic rats. Can. J. Ophthalmol. 2010, 45, 46–51. [Google Scholar] [CrossRef]
  106. Rao, G.; Ding, H.G.; Huang, W.; Le, D.; Maxhimer, J.B.; Oosterhof, A.; van Kuppevelt, T.; Lum, H.; Lewis, E.J.; Reddy, V.; et al. Reactive oxygen species mediate high glucose-induced heparanase-1 production and heparan sulphate proteoglycan degradation in human and rat endothelial cells: A potential role in the pathogenesis of atherosclerosis. Diabetologia 2011, 54, 1527–1538. [Google Scholar] [CrossRef] [Green Version]
  107. Laskov, R.; Michaeli, R.-I.; Sharir, H.; Yefenof, E.; Vlodavsky, I. Production of heparanase by normal and neoplastic murine B-lymphocytes. Int. J. Cancer 1991, 47, 92–98. [Google Scholar] [CrossRef]
  108. Zaza, G.; Masola, V.; Granata, S.; Pontrelli, P.; Sallustio, F.; Gesualdo, L.; Gambaro, G.; Grandaliano, G.; Lupo, A. Dialysis-related transcriptomic profiling: The pivotal role of heparanase. Exp. Biol. Med. 2014, 239, 52–64. [Google Scholar] [CrossRef]
  109. Kramer, A.; van den Hoven, M.; Rops, A.; Wijnhoven, T.; van den Heuvel, L.; Lensen, J.; van Kuppevelt, T.; van Goor, H.; van der Vlag, J.; Navis, G.; et al. Induction of Glomerular Heparanase Expression in Rats with Adriamycin Nephropathy Is Regulated by Reactive Oxygen Species and the Renin-Angiotensin System. J. Am. Soc. Nephrol. 2006, 17, 2513–2520. [Google Scholar] [CrossRef] [Green Version]
  110. Van den Hoven, M.J.; Waanders, F.; Rops, A.L.; Kramer, A.B.; van Goor, H.; Berden, J.H.; Navis, G.; van der Vlag, J. Regulation of glomerular heparanase expression by aldosterone, angiotensin II and reactive oxygen species. Nephrol. Dial. Transplant. 2009, 24, 2637–2645. [Google Scholar] [CrossRef] [Green Version]
  111. Garsen, M.; Sonneveld, R.; Rops, A.L.; Huntink, S.; van Kuppevelt, T.H.; Rabelink, T.J.; Hoenderop, J.G.; Berden, J.H.; Nijenhuis, T.; van der Vlag, J. Vitamin D attenuates proteinuria by inhibition of heparanase expression in the podocyte. J. Pathol. 2015, 237, 472–481. [Google Scholar] [CrossRef]
  112. Gallimidi, A.B.; Fischman, S.; Revach, B.; Bulvik, R.; Maliutina, A.; Rubinstein, A.M.; Nussbaum, G.; Elkin, M. Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model. Oncotarget 2015, 6, 22613–22623. [Google Scholar] [CrossRef] [Green Version]
  113. Liu, L.-P.; Sheng, X.-P.; Shuai, T.-K.; Zhao, Y.-X.; Li, B.; Li, Y.-M. Helicobacter pylori promotes invasion and metastasis of gastric cancer by enhancing heparanase expression. World J. Gastroenterol. 2018, 24, 4565–4577. [Google Scholar] [CrossRef]
  114. Berk, R.S.; Dong, Z.; Alousi, S.; Kosir, M.A.; Wang, Y.; Vlodavsky, I. Murine ocular heparanase expression before and during infection with Pseudomonas aeruginosa. Investig. Ophthalmol. Vis. Sci. 2004, 45, 1182–1187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Lapidot, M.; Barash, U.; Zohar, Y.; Geffen, Y.; Naroditsky, I.; Ilan, N.; Best, L.A.; Vlodavsky, I. Involvement of Heparanase in Empyema: Implication for Novel Therapeutic Approaches. J. Clin. Cell. Immunol. 2015, 6, 290. [Google Scholar] [CrossRef] [Green Version]
  116. Buijsers, B.; Yanginlar, C.; de Nooijer, A.; Grondman, I.; Maciej-Hulme, M.L.; Jonkman, I.; Janssen, N.A.F.; Rother, N.; de Graaf, M.; Pickkers, P.; et al. Increased Plasma Heparanase Activity in COVID-19 Patients. Front. Immunol. 2020, 11, 2572. [Google Scholar] [CrossRef]
  117. Stahl, K.; Gronski, P.A.; Kiyan, Y.; Seeliger, B.; Bertram, A.; Pape, T.; Welte, T.; Hoeper, M.M.; Haller, H.; David, S. Injury to the Endothelial Glycocalyx in Critically Ill Patients with COVID-19. Am. J. Respir. Crit. Care Med. 2020, 202, 1178–1181. [Google Scholar] [CrossRef] [PubMed]
  118. Puerta-Guardo, H.; Glasner, D.R.; Harris, E. Dengue Virus NS1 Disrupts the Endothelial Glycocalyx, Leading to Hyperpermeability. PLoS Pathog. 2016, 12, e1005738. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Chen, H.R.; Chao, C.H.; Liu, C.C.; Ho, T.S.; Tsai, H.P.; Perng, G.C.; Lin, Y.S.; Wang, J.R.; Yeh, T.M. Macrophage migration inhibitory factor is critical for dengue NS1-induced endothelial glycocalyx degradation and hyperpermeability. PLoS Pathog. 2018, 14, e1007033. [Google Scholar] [CrossRef]
  120. Agelidis, A.M.; Hadigal, S.R.; Jaishankar, D.; Shukla, D. Viral Activation of Heparanase Drives Pathogenesis of Herpes Simplex Virus-1. Cell Rep. 2017, 20, 439–450. [Google Scholar] [CrossRef] [Green Version]
  121. Tao, C.; Wang, W.; Zhou, P.; Zhou, X.; Zhang, Q.; Liu, B. Molecular characterization, expression profiles of the porcine SDC2 and HSPG2 genes and their association with hematologic parameters. Mol. Biol. Rep. 2013, 40, 2549–2556. [Google Scholar] [CrossRef]
  122. Guo, C.; Zhu, Z.; Guo, Y.; Wang, X.; Yu, P.; Xiao, S.; Chen, Y.; Cao, Y.; Liu, X. Heparanase Upregulation Contributes to Porcine Reproductive and Respiratory Syndrome Virus Release. J. Virol. 2017, 91, e00625-17. [Google Scholar] [CrossRef] [Green Version]
  123. Bhattacharya, U.; Gutter-Kapon, L.; Kan, T.; Boyango, I.; Barash, U.; Yang, S.M.; Liu, J.J.; Gross-Cohen, M.; Sanderson, R.D.; Shaked, Y.; et al. Heparanase and chemotherapy synergize to drive macrophage activation and enhance tumor growth. Cancer Res. 2020, 80, 57–68. [Google Scholar] [CrossRef] [Green Version]
  124. Bartlett, M.R.; Underwood, P.A.; Parish, C.R. Comparative analysis of the ability of leucocytes, endothelial cells and platelets to degrade the subendothelial basement membrane: Evidence for cytokine dependence and detection of a novel sulfatase. Immunol. Cell Biol. 1995, 73, 113–124. [Google Scholar] [CrossRef] [PubMed]
  125. Iriyama, S.; Matsunaga, Y.; Takahashi, K.; Matsuzaki, K.; Kumagai, N.; Amano, S. Activation of heparanase by ultraviolet B irradiation leads to functional loss of basement membrane at the dermal-epidermal junction in human skin. Arch. Dermatol. Res. 2011, 303, 253–261. [Google Scholar] [CrossRef] [PubMed]
  126. Chung, S.I.; Seong, J.; Park, Y.N.; Kim, W.W.; Oh, H.J.; Han, K.H. Identification of proteins indicating radiation-induced Hepatic Toxicity in cirrhotic rats. J. Radiat. Res. 2010, 51, 643–650. [Google Scholar] [CrossRef] [Green Version]
  127. Khamaysi, I.; Singh, P.; Nasser, S.; Awad, H.; Chowers, Y.; Sabo, E.; Hammond, E.; Gralnek, I.; Minkov, I.; Noseda, A.; et al. The Role of Heparanase in the Pathogenesis of Acute Pancreatitis: A Potential Therapeutic Target. Sci. Rep. 2017, 7, 715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Ozaki, T.; Nakagawara, A. Role of p53 in cell death and human cancers. Cancers 2011, 3, 994–1013. [Google Scholar] [CrossRef] [PubMed]
  129. Nana-Sinkam, S.P.; Croce, C.M. MicroRNA regulation of tumorigenesis, cancer progression and interpatient heterogeneity: Towards clinical use. Genome Biol. 2014, 15, 445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Jin, H.; Zhou, S. The Functions of Heparanase in Human Diseases. Mini Rev. Med. Chem. 2017, 17, 541–548. [Google Scholar] [CrossRef]
  131. Lerner, I.; Hermano, E.; Zcharia, E.; Rodkin, D.; Bulvik, R.; Doviner, V.; Rubinstein, A.M.; Ishai-Michaeli, R.; Atzmon, R.; Sherman, Y.; et al. Heparanase powers a chronic inflammatory circuit that promotes colitis-associated tumorigenesis in mice. J. Clin. Investig. 2011, 121, 1709–1721. [Google Scholar] [CrossRef]
  132. Grimbacher, B.; Aicher, W.K.; Peter, H.H.; Eibel, H. TNF-α induces the transcription factor Egr-1, pro-inflammatory cytokines and cell proliferation in human skin fibroblasts and synovial lining cells. Rheumatol. Int. 1998, 17, 185–192. [Google Scholar] [CrossRef]
  133. Cao, Y.; Zhang, J.; Meng, X.; Wang, D. TNF-α induces early growth response gene-1 expression via ERK1/2 activation in endothelial cells. Acta Diabetol. 2013, 50, 27–31. [Google Scholar] [CrossRef]
  134. Ma, H.; Bernstein, L.; Ross, R.K.; Ursin, G. Hormone-related risk factors for breast cancer in women under age 50 years by estrogen and progesterone receptor status: Results from a case-control and a case-case comparison. Breast Cancer Res. 2006, 8, R39. [Google Scholar] [CrossRef] [PubMed]
  135. Moloney, J.N.; Cotter, T.G. ROS signalling in the biology of cancer. Semin. Cell Dev. Biol. 2018, 80, 50–64. [Google Scholar] [CrossRef] [PubMed]
  136. Thakkar, N.; Yadavalli, T.; Jaishankar, D.; Shukla, D. Emerging Roles of Heparanase in Viral Pathogenesis. Pathogens 2017, 6, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Koganti, R.; Suryawanshi, R.; Shukla, D. Heparanase, cell signaling, and viral infections. Cell. Mol. Life Sci. 2020, 77, 5059–5077. [Google Scholar] [CrossRef]
  138. Jackson, T.; Ellard, F.M.; Ghazaleh, R.A.; Brookes, S.M.; Blakemore, W.E.; Corteyn, A.H.; Stuart, D.I.; Newman, J.W.; King, A.M. Efficient infection of cells in culture by type O foot-and-mouth disease virus requires binding to cell surface heparan sulfate. J. Virol. 1996, 70, 5282–5287. [Google Scholar] [CrossRef] [Green Version]
  139. Hallak, L.K.; Collins, P.L.; Knudson, W.; Peeples, M.E. Iduronic acid-containing glycosaminoglycans on target cells are required for efficient respiratory syncytial virus infection. Virology 2000, 271, 264–275. [Google Scholar] [CrossRef] [Green Version]
  140. Giroglou, T.; Florin, L.; Schäfer, F.; Streeck, R.E.; Sapp, M. Human Papillomavirus Infection Requires Cell Surface Heparan Sulfate. J. Virol. 2001, 75, 1565–1570. [Google Scholar] [CrossRef] [Green Version]
  141. Cooper, A.; Tal, G.; Lider, O.; Shaul, Y. Cytokine Induction by the Hepatitis B Virus Capsid in Macrophages Is Facilitated by Membrane Heparan Sulfate and Involves TLR2. J. Immunol. 2005, 175, 3165–3176. [Google Scholar] [CrossRef] [PubMed]
  142. Khanna, M.; Ranasinghe, C.; Jackson, R.; Parish, C.R. Heparan sulfate as a receptor for poxvirus infections and as a target for antiviral agents. J. Gen. Virol. 2017, 98, 2556–2568. [Google Scholar] [CrossRef]
  143. Bar-Sela, G.; Kaplan-Cohen, V.; Ilan, N.; Vlodavsky, I.; Ben-Izhak, O. Heparanase expression in nasopharyngeal carcinoma inversely correlates with patient survival. Histopathology 2006, 49, 188–193. [Google Scholar] [CrossRef] [PubMed]
  144. Vornicova, O.; Boyango, I.; Feld, S.; Naroditsky, I.; Kazarin, O.; Zohar, Y.; Tiram, Y.; Ilan, N.; Ben-Izhak, O.; Vlodavsky, I.; et al. The prognostic significance of heparanase expression in metastatic melanoma. Oncotarget 2016, 7, 74678–74685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Wu, X.; Yu, J.; Gao, G.; Wang, X.; Liu, Y.; Zhu, S.; Gong, Z. Salivary Heparanase Level Is a Potential Biomarker to Diagnose and Prognose the Malignant Salivary Gland Tumor. PLoS ONE 2015, 10, e0143009. [Google Scholar] [CrossRef] [PubMed]
  146. Garcia, B.; Fernández-Vega, I.; García-Suárez, O.; Castañón, S.; Quirós, L.M. The Role of Heparan Sulfate Proteoglycans in Bacterial Infections. J. Med. Microbiol. Diagn. 2014, 03, e157. [Google Scholar] [CrossRef]
  147. García, B.; Merayo-Lloves, J.; Martin, C.; Alcalde, I.; Quirós, L.M.; Vazquez, F. Surface proteoglycans as mediators in bacterial pathogens infections. Front. Microbiol. 2016, 7, 220. [Google Scholar] [CrossRef] [Green Version]
  148. Rangarajan, S.; Richter, J.R.; Richter, R.P.; Bandari, S.K.; Tripathi, K.; Vlodavsky, I.; Sanderson, R.D. Heparanase-enhanced Shedding of Syndecan-1 and Its Role in Driving Disease Pathogenesis and Progression. J. Histochem. Cytochem. 2020, 68, 823–840. [Google Scholar] [CrossRef] [PubMed]
  149. Zhang, L.; Ngo, J.A.; Wetzel, M.D.; Marchetti, D. Heparanase mediates a novel mechanism in lapatinib-resistant brain metastatic breast cancer. Neoplasia 2015, 17, 101–113. [Google Scholar] [CrossRef] [Green Version]
  150. Ramani, V.C.; Zhan, F.; He, J.; Barbieri, P.; Noseda, A.; Tricot, G.; Sanderson, R.D. Targeting heparanase overcomes chemoresistance and diminishes relapse in myeloma. Oncotarget 2016, 7, 1598–1607. [Google Scholar] [CrossRef] [Green Version]
  151. Wang, X.; Zuo, D.; Chen, Y.; Li, W.; Liu, R.; He, Y.; Ren, L.; Zhou, L.; Deng, T.; Ying, G.; et al. Shed Syndecan-1 is involved in chemotherapy resistance via the EGFR pathway in colorectal cancer. Br. J. Cancer 2014, 111, 1965–1976. [Google Scholar] [CrossRef]
  152. Goodall, K.J.; Poon, I.K.H.; Phipps, S.; Hulett, M.D. Soluble Heparan Sulfate Fragments Generated by Heparanase Trigger the Release of Pro-Inflammatory Cytokines through TLR-4. PLoS ONE 2014, 9, e109596. [Google Scholar] [CrossRef]
  153. Gutter-Kapon, L.; Alishekevitz, D.; Shaked, Y.; Li, J.-P.; Aronheim, A.; Ilan, N.; Vlodavsky, I. Heparanase is required for activation and function of macrophages. Proc. Natl. Acad. Sci. USA 2016, 113, E7808–E7817. [Google Scholar] [CrossRef] [Green Version]
  154. Song, T.; Spillmann, D. Transcriptomic analysis reveals cell apoptotic signature modified by heparanase in melanoma cells. J. Cell. Mol. Med. 2019, 23, 4559–4568. [Google Scholar] [CrossRef]
  155. Abboud-Jarrous, G.; Rangini-Guetta, Z.; Aingorn, H.; Atzmon, R.; Elgavish, S.; Peretz, T.; Vlodavsky, I. Site-directed mutagenesis, proteolytic cleavage, and activation of human proheparanase. J. Biol. Chem. 2005, 280, 13568–13575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Abboud-Jarrous, G.; Atzmon, R.; Peretz, T.; Palermo, C.; Gadea, B.B.; Joyce, J.A.; Vlodavsky, I. Cathepsin L is responsible for processing and activation of proheparanase through multiple cleavages of a linker segment. J. Biol. Chem. 2008, 283, 18167–18176. [Google Scholar] [CrossRef] [Green Version]
  157. Temkin, V.; Aingorn, H.; Puxeddu, I.; Goldshmidt, O.; Zcharia, E.; Gleich, G.J.; Vlodavsky, I.; Levi-Schaffer, F. Eosinophil major basic protein: First identified natural heparanase-inhibiting protein. J. Allergy Clin. Immunol. 2004, 113, 703–709. [Google Scholar] [CrossRef] [PubMed]
  158. McKenzie, E.A. Heparanase: A target for drug discovery in cancer and inflammation. Br. J. Pharmacol. 2007, 151, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Marchetti, D.; Liu, S.; Spohn, W.C.; Carson, D.D. Heparanase and a synthetic peptide of heparan sulfate-interacting protein recognize common sites on cell surface and extracellular matrix heparan sulfate. J. Biol. Chem. 1997, 272, 15891–15897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Levy-Adam, F.; Feld, S.; Cohen-Kaplan, V.; Shteingauz, A.; Gross, M.; Arvatz, G.; Naroditsky, I.; Ilan, N.; Doweck, I.; Vlodavsky, I. Heparanase 2 Interacts with Heparan Sulfate with High Affinity and Inhibits Heparanase Activity. J. Biol. Chem. 2010, 285, 28010–28019. [Google Scholar] [CrossRef] [Green Version]
  161. Mohan, C.D.; Hari, S.; Preetham, H.D.; Rangappa, S.; Barash, U.; Ilan, N.; Nayak, S.C.; Gupta, V.K.; Basappa; Vlodavsky, I.; et al. Targeting Heparanase in Cancer: Inhibition by Synthetic, Chemically modified and Natural Compounds. iScience 2019, 15, 360–390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Vlodavsky, I.; Ilan, N.; Naggi, A. Benito Casu Heparanase: Structure, Biological Functions, and Inhibition by Heparin-Derived Mimetics of Heparan Sulfate. Curr. Pharm. Des. 2007, 13, 2057–2073. [Google Scholar] [CrossRef]
  163. Cassinelli, G.; Torri, G.; Naggi, A. Non-Anticoagulant Heparins as Heparanase Inhibitors. Adv. Exp. Med. Biol. 2020, 1221, 493–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Van Den Hoven, M.J.; Rops, A.L.; Vlodavsky, I.; Levidiotis, V.; Berden, J.H.; van der Vlag, J. Heparanase in glomerular diseases. Kidney Int. 2007, 72, 543–548. [Google Scholar] [CrossRef] [Green Version]
  165. Coombe, D.R.; Gandhi, N.S. Heparanase: A Challenging Cancer Drug Target. Front. Oncol. 2019, 9, 1316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Toyoshima, M.; Nakajima, M. Human heparanase. Purification, characterization, cloning, and expression. J. Biol. Chem. 1999, 274, 24153–24160. [Google Scholar] [CrossRef] [Green Version]
  167. Chen, L.; Sanderson, R.D. Heparanase Regulates Levels of Syndecan-1 in the Nucleus. PLoS ONE 2009, 4, e4947. [Google Scholar] [CrossRef] [Green Version]
  168. Purushothaman, A.; Hurst, D.R.; Pisano, C.; Mizumoto, S.; Sugahara, K.; Sanderson, R.D. 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, 30377–30383. [Google Scholar] [CrossRef] [Green Version]
  169. Yang, Y.; Gorzelanny, C.; Bauer, A.T.; Halter, N.; Komljenovic, D.; Bäuerle, T.; Borsig, L.; Roblek, M.; Schneider, S.W. Nuclear heparanase-1 activity suppresses melanoma progression via its DNA-binding affinity. Oncogene 2015, 34, 5832–5842. [Google Scholar] [CrossRef]
  170. Schubert, S.Y.; Ilan, N.; Shushy, M.; Ben-Izhak, O.; Vlodavsky, I.; Goldshmidt, O. Human heparanase nuclear localization and enzymatic activity. Lab. Investig. 2004, 84, 535–544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Dos Santos, T.C.F.; Gomes, A.M.; Paschoal, M.E.M.; Stelling, M.P.; Rumjanek, V.M.B.D.; do Junior, A.R.; Valiante, P.M.; Madi, K.; de Souza, H.S.P.; Pavão, M.S.G.; et al. Heparanase expression and localization in different types of human lung cancer. Biochim. Biophys. Acta Gen. Subj. 2014, 1840, 2599–2608. [Google Scholar] [CrossRef]
  172. He, Y.Q.; Sutcliffe, E.L.; Bunting, K.L.; Li, J.; Goodall, K.J.; Poon, I.K.A.; Hulett, M.D.; Freeman, C.; Zafar, A.; McInnes, R.L.; et al. The endoglycosidase heparanase enters the nucleus of T lymphocytes and modulates H3 methylation at actively transcribed genes via the interplay with key chromatin modifying enzymes. Transcription 2012, 3, 130–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Doweck, I.; Kaplan-Cohen, V.; Naroditsky, I.; Sabo, E.; Ilan, N.; Vlodavsky, I. Heparanase Localization and Expression by Head and Neck Cancer: Correlation with Tumor Progression and Patient Survival. Neoplasia 2006, 8, 1055–1061. [Google Scholar] [CrossRef] [Green Version]
  174. Cohen, E.; Doweck, I.; Naroditsky, I.; Ben-Izhak, O.; Kremer, R.; Best, L.A.; Vlodavsky, I.; Ilan, N. Heparanase is overexpressed in lung cancer and correlates inversely with patient survival. Cancer 2008, 113, 1004–1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. 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, 1455–1463. [Google Scholar] [CrossRef] [Green Version]
  176. Ramani, V.C.; Yang, Y.; Ren, Y.; Nan, L.; Sanderson, R.D. Heparanase plays a dual role in driving hepatocyte growth factor (HGF) signaling by enhancing HGF expression and activity. J. Biol. Chem. 2011, 286, 6490–6499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Li, J.; Meng, X.; Hu, J.; Zhang, Y.; Dang, Y.; Wei, L.; Shi, M. Heparanase promotes radiation resistance of cervical cancer by upregulating hypoxia inducible factor 1. Am. J. Cancer Res. 2017, 7, 234–244. [Google Scholar]
  178. Masola, V.; Zaza, G.; Secchi, M.F.; Gambaro, G.; Lupo, A.; Onisto, M. Heparanase is a key player in renal fibrosis by regulating TGF-β expression and activity. Biochim. Biophys. Acta Mol. Cell Res. 2014, 1843, 2122–2128. [Google Scholar] [CrossRef]
  179. Abassi, Z.; Hamoud, S.; Hassan, A.; Khamaysi, I.; Nativ, O.; Heyman, S.N.; Muhammad, R.S.; Ilan, N.; Singh, P.; Hammond, E.; et al. Involvement of heparanase in the pathogenesis of acute kidney injury: Nephroprotective effect of PG545. Oncotarget 2017, 8, 34191–34204. [Google Scholar] [CrossRef]
  180. Xia, J.; Sheng, W.; Pei, L.; Li, N.; Zhang, Z.; Wang, J.; Zu, J.; Wang, N.; Wang, D. Effects of unfractionated heparin and rivaroxaban on the expression of heparanase and fibroblast growth factor 2 in human osteoblasts. Mol. Med. Rep. 2017, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Shah, S.; Fourgeaud, C.; Derieux, S.; Mirshahi, S.; Contant, G.; Pimpie, C.; lo Dico, R.; Soria, J.; Pocard, M.; Mirshahi, M. The close relationship between heparanase and epithelial mesenchymal transition in gastric signet-ring cell adenocarcinoma. Oncotarget 2018, 9, 33778–33787. [Google Scholar] [CrossRef] [PubMed]
  182. Masola, V.; Zaza, G.; Bellin, G.; Dall’Olmo, L.; Granata, S.; Vischini, G.; Secchi, M.F.; Lupo, A.; Gambaro, G.; Onisto, M.; et al. Heparanase regulates the M1 polarization of renal macrophages and their crosstalk with renal epithelial tubular cells after ischemia/reperfusion injury. FASEB J. 2018, 32, 742–756. [Google Scholar] [CrossRef] [Green Version]
  183. Tsunekawa, N.; Higashi, N.; Kogane, Y.; Waki, M.; Shida, H.; Nishimura, Y.; Adachi, H.; Nakajima, M.; Irimura, T. Heparanase augments inflammatory chemokine production from colorectal carcinoma cell lines. Biochem. Biophys. Res. Commun. 2016, 469, 878–883. [Google Scholar] [CrossRef] [PubMed]
  184. Purushothaman, A.; Chen, L.; Yang, Y.; Sanderson, R.D. Heparanase stimulation of protease expression implicates it as a master regulator of the aggressive tumor phenotype in myeloma. J. Biol. Chem. 2008, 283, 32628–32636. [Google Scholar] [CrossRef] [Green Version]
  185. Lv, B.; Zhang, B.; Hu, X.Y.; Zeng, Q.D. Heparanase regulates in vitro VEGF-C expression and its clinical significance to pancreatic ductal cell adenocarcinoma. Oncol. Lett. 2016, 11, 1327–1334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Wang, F.; Wang, Y.; Zhang, D.; Puthanveetil, P.; Johnson, J.D.; Rodrigues, B. Fatty Acid-Induced Nuclear Translocation of Heparanase Uncouples Glucose Metabolism in Endothelial Cells. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 406–414. [Google Scholar] [CrossRef] [Green Version]
  187. Okawa, T.; Naomoto, Y.; Nobuhisa, T.; Takaoka, M.; Motoki, T.; Shirakawa, Y.; Yamatsuji, T.; Inoue, H.; Ouchida, M.; Gunduz, M.; et al. Heparanase Is Involved in Angiogenesis in Esophageal Cancer through Induction of Cyclooxygenase-2. Clin. Cancer Res. 2005, 11, 7995. [Google Scholar] [CrossRef] [Green Version]
  188. Yan, X.; Jin, S.; Li, S.; Gong, F.; Zhang, D.; Zhang, X.; Li, J. Heparanase Modulation of Early Growth Response Gene Expression. Zool. Sci. 2011, 28, 189–194. [Google Scholar] [CrossRef]
  189. Masola, V.; Bellin, G.; Vischini, G.; Dall’Olmo, L.; Granata, S.; Gambaro, G.; Lupo, A.; Onisto, M.; Zaza, G. Inhibition of heparanase protects against chronic kidney dysfunction following ischemia/reperfusion injury. Oncotarget 2018, 9, 36185–36201. [Google Scholar] [CrossRef] [Green Version]
  190. Hu, X.; Zhang, L.; Jin, J.; Zhu, W.; Xu, Y.; Wu, Y.; Wang, Y.; Chen, H.; Webster, K.A.; Chen, H.; et al. Heparanase released from mesenchymal stem cells activates integrin beta1/HIF-2alpha/Flk-1 signaling and promotes endothelial cell migration and angiogenesis. Stem Cells 2015, 33, 1850–1862. [Google Scholar] [CrossRef]
  191. Chen, X.; Cheng, B.; Dai, D.; Wu, Y.; Feng, Z.; Tong, C.; Wang, X.; Zhao, J. Heparanase induces necroptosis of microvascular endothelial cells to promote the metastasis of hepatocellular carcinoma. Cell Death Discov. 2021, 7. [Google Scholar] [CrossRef]
  192. Huang, X.; Reye, G.; Momot, K.I.; Blick, T.; Lloyd, T.; Tilley, W.D.; Hickey, T.E.; Snell, C.E.; Okolicsanyi, R.K.; Haupt, L.M.; et al. Heparanase Promotes Syndecan-1 Expression to Mediate Fibrillar Collagen and Mammographic Density in Human Breast Tissue Cultured ex vivo. Front. Cell Dev. Biol. 2020, 8, 599. [Google Scholar] [CrossRef]
  193. Masola, V.; Gambaro, G.; Tibaldi, E.; Brunati, A.M.; 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, 1478–1488. [Google Scholar] [CrossRef] [Green Version]
  194. Singh, P.; Blatt, A.; Feld, S.; Zohar, Y.; Saadi, E.; Barki-Harrington, L.; Hammond, E.; Ilan, N.; Vlodavsky, I.; Chowers, Y.; et al. The Heparanase Inhibitor PG545 Attenuates Colon Cancer Initiation and Growth, Associating with Increased p21 Expression. Neoplasia 2017, 19, 175–184. [Google Scholar] [CrossRef] [PubMed]
  195. Nadir, Y.; Brenner, B.; Zetser, A.; Ilan, N.; Shafat, I.; Zcharia, E.; Goldshmidt, O.; Vlodavsky, I. Heparanase induces tissue factor expression in vascular endothelial and cancer cells. J. Thromb. Haemost. 2006, 4, 2443–2451. [Google Scholar] [CrossRef] [PubMed]
  196. Martin, L.; Gombert, A.; Chen, J.; Liebens, J.; Verleger, J.; Kalder, J.; Marx, G.; Jacobs, M.; Thiemermann, C.; Schuerholz, T. The ß-D-endoglucuronidase heparanase is a danger molecule that drives systemic inflammation and correlates with clinical course after open and endovascular thoracoabdominal aortic aneurysm repair: Lessons learnt from mice and men. Front. Immunol. 2017, 8, 681. [Google Scholar] [CrossRef] [PubMed]
  197. Wei, R.R.; Sun, D.N.; Yang, H.; Yan, J.; Zhang, X.; Zheng, X.L.; Fu, X.H.; Geng, M.Y.; Huang, X.; Ding, J. CTC clusters induced by heparanase enhance breast cancer metastasis. Acta Pharmacol. Sin. 2018, 39, 1326–1337. [Google Scholar] [CrossRef] [Green Version]
  198. Crispel, Y.; Ghanem, S.; Attias, J.; Kogan, I.; Brenner, B.; Nadir, Y. Involvement of the heparanase procoagulant domain in bleeding and wound healing. J. Thromb. Haemost. 2017, 15, 1463–1472. [Google Scholar] [CrossRef] [Green Version]
  199. Blich, M.; Golan, A.; Arvatz, G.; Sebbag, A.; Shafat, I.; Sabo, E.; Cohen-Kaplan, V.; Petcherski, S.; Avniel-Polak, S.; Eitan, A.; et al. Macrophage activation by heparanase is mediated by TLR-2 and TLR-4 and associates with plaque progression. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 56–65. [Google Scholar] [CrossRef] [Green Version]
  200. Bitan, M.; Weiss, L.; Reibstein, I.; Zeira, M.; Fellig, Y.; Slavin, S.; Zcharia, E.; Nagler, A.; Vlodavsky, I. Heparanase upregulates Th2 cytokines, ameliorating experimental autoimmune encephalitis. Mol. Immunol. 2010, 47, 1890–1898. [Google Scholar] [CrossRef] [Green Version]
  201. Agelidis, A.; Turturice, B.A.; Suryawanshi, R.K.; Yadavalli, T.; Jaishankar, D.; Ames, J.; Hopkins, J.; Koujah, L.; Patil, C.D.; Hadigal, S.R.; et al. Disruption of innate defense responses by endoglycosidase HPSE promotes cell survival. JCI Insight 2021, 6, e144255. [Google Scholar] [CrossRef] [PubMed]
  202. Abeyrathna, P.; Su, Y. The critical role of Akt in cardiovascular function. Vascul. Pharmacol. 2015, 74, 38–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Gingis-Velitski, S.; Zetser, A.; Flugelman, M.Y.; Vlodavsky, I.; Ilan, N. Heparanase Induces Endothelial Cell Migration via Protein Kinase B/Akt Activation. J. Biol. Chem. 2004, 279, 23536–23541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. 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, 829–834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. 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. J. Biol. Chem. 2012, 287, 6668–6678. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Ramos, J.W. The regulation of extracellular signal-regulated kinase (ERK) in mammalian cells. Int. J. Biochem. Cell Biol. 2008, 40, 2707–2719. [Google Scholar] [CrossRef]
  207. Xiong, A.; Kundu, S.; Forsberg, M.; Xiong, Y.; Bergström, T.; Paavilainen, T.; Kjellén, L.; Li, J.-P.P.; Forsberg-Nilsson, K. Heparanase confers a growth advantage to differentiating murine embryonic stem cells, and enhances oligodendrocyte formation. Matrix Biol. 2017, 62, 92–104. [Google Scholar] [CrossRef]
  208. Oda, K.; Matsuoka, Y.; Funahashi, A.; Kitano, H. A comprehensive pathway map of epidermal growth factor receptor signaling. Mol. Syst. Biol. 2005, 1, 2005-0010. [Google Scholar] [CrossRef] [Green Version]
  209. Cohen-Kaplan, V.; Doweck, I.; Naroditsky, I.; Vlodavsky, I.; Ilan, N. Heparanase augments epidermal growth factor receptor phosphorylation: Correlation with head and neck tumor progression. Cancer Res. 2008, 68, 10077–10085. [Google Scholar] [CrossRef] [Green Version]
  210. Vlodavsky, I.; Singh, P.; Boyango, I.; Gutter-Kapon, L.; Elkin, M.; Sanderson, R.D.; Ilan, N. Heparanase: From basic research to therapeutic applications in cancer and inflammation. Drug Resist. Updat. 2016, 29, 54–75. [Google Scholar] [CrossRef]
Figure 1. Regulators of heparanase expression. Heparanase expression is positively and negatively regulated by a number of cytokines, growth factors, signaling molecules, therapeutics, pathogens, transcription factors, and miRNA. (a) Estrogen binding to the estrogen receptor allows binding to the estrogen response element within the heparanase promoter and heparanase upregulation. (b) Wild-type BRAF inhibits heparanase expression by directly repressing ETS1, a transcription factor known to promote heparanase expression. (c) HGF, via the PI3K/Akt pathway, activates NF-κB to induce heparanase expression. Erythromycin inhibits HGF (and PDGF)-induced heparanase upregulation. (d) Hypoxia, and (e) HSV-1 upregulate heparanase via NF-κB. (f) Vitamin D activates the vitamin D receptor which directly binds and inhibits the heparanase promoter. (g) bFGF upregulation of heparanase can be inhibited with clarithromycin. (h) Activation of the MEK Erk pathway upregulates heparanase. (i) LPS binding to TLR4 upregulates heparanase. (j) H. pylori infection upregulates heparanase via MAPK. ERE, estrogen response element; Hpse, heparanase.
Figure 1. Regulators of heparanase expression. Heparanase expression is positively and negatively regulated by a number of cytokines, growth factors, signaling molecules, therapeutics, pathogens, transcription factors, and miRNA. (a) Estrogen binding to the estrogen receptor allows binding to the estrogen response element within the heparanase promoter and heparanase upregulation. (b) Wild-type BRAF inhibits heparanase expression by directly repressing ETS1, a transcription factor known to promote heparanase expression. (c) HGF, via the PI3K/Akt pathway, activates NF-κB to induce heparanase expression. Erythromycin inhibits HGF (and PDGF)-induced heparanase upregulation. (d) Hypoxia, and (e) HSV-1 upregulate heparanase via NF-κB. (f) Vitamin D activates the vitamin D receptor which directly binds and inhibits the heparanase promoter. (g) bFGF upregulation of heparanase can be inhibited with clarithromycin. (h) Activation of the MEK Erk pathway upregulates heparanase. (i) LPS binding to TLR4 upregulates heparanase. (j) H. pylori infection upregulates heparanase via MAPK. ERE, estrogen response element; Hpse, heparanase.
Ijms 22 11096 g001
Figure 2. Heparanase regulates protein expression and phosphorylation via multiple mechanisms. Heparanase regulates gene transcription via (a) non-enzymatic mechanisms, (b) generating soluble HS fragments which bind and activate TLR4 signaling and NF-κB activation, (c) Erk, p38, and JNK signaling, (d) nuclear localization and cleavage of HS to activate histone acetyltransferase (HAT), (e) Src, (f) hypoxia inducible factor (HIF)-1 activation, (g) direct binding to promoter to block gene transcription, and (h) by unknown mechanisms. (i) Heparanase can also regulate the expression of other proteins, although the mechanism of this is unknown. (j) Finally, heparanase can induce phosphorylation of several proteins. Hpse, heparanase; HS, heparan sulfate.
Figure 2. Heparanase regulates protein expression and phosphorylation via multiple mechanisms. Heparanase regulates gene transcription via (a) non-enzymatic mechanisms, (b) generating soluble HS fragments which bind and activate TLR4 signaling and NF-κB activation, (c) Erk, p38, and JNK signaling, (d) nuclear localization and cleavage of HS to activate histone acetyltransferase (HAT), (e) Src, (f) hypoxia inducible factor (HIF)-1 activation, (g) direct binding to promoter to block gene transcription, and (h) by unknown mechanisms. (i) Heparanase can also regulate the expression of other proteins, although the mechanism of this is unknown. (j) Finally, heparanase can induce phosphorylation of several proteins. Hpse, heparanase; HS, heparan sulfate.
Ijms 22 11096 g002
Table 1. Mammalian heparan sulfate-binding proteins.
Table 1. Mammalian heparan sulfate-binding proteins.
ProteinTechniqueReference
Amyloid peptide β (1–42)Surface plasmon resonance[13]
Amyloid precursor proteinFluorescence lifetime imaging microscopy[14]
Annexin VAffinity chromatography[15]
Basic fibroblast growth factor (bFGF) (FGF-2)Iodinated-bFGF and specific activity following heparanase addition; Affinity chromatography; Iodinated-bFGF and specific activity; Cross-linking of iodinated-bFGF following heparitinase treatment[16,17,18,19]
Collagen IAffinity chromatography; Surface plasmon resonance[13,20]
Collagen VSolid phase binding assay; Surface plasmon resonance[21,22]
Collagenase IVAntibody-linked detection assay; Surface plasmon resonance[13,23]
Collagen VISurface plasmon resonance[13]
chemokine (C-X-C motif) ligand (CXCL1) (KC)Surface plasmon resonance[24]
CXCL2 (MIP-2)Surface plasmon resonance[24]
CXCL6 (GCP-2)Surface plasmon resonance[24]
CXCL10 (IP-10)Alkaline phosphatase-conjucated IP-10; Surface plasmon resonance[24,25]
CXCL11 (I-TAC)Surface plasmon resonance[24]
CXCL13Surface plasmon resonance[26]
EndostatinAlkaline phosphatase-endostatin binding assay; Filter-binding assay; Surface plasmon resonance[13,27,28]
FGFR4Affinity chromatography[29]
FibronectinAffinity chromatography; Antibody-linked detection assay[7,8,23]
HGFAffinity chromatography[30]
Histidine-rich glycoproteinFlow cytometry after heparanase treatment[31]
High mobility group protein B1Biotinylation and streptadivin-HRP detection[32]
Integrin α5β1Surface plasmon resonance[13]
Interferon-β (IFN-β)Filter binding assay[33]
Interleukin-8 (IL-8)Affinity co-electrophoresis[34]
Laminin-1Antibody-linked detection assay;
Surface plasmon resonance
[13,23]
L-selectinHeparinase treatment of 35SO4-labeled L-selectin ligands and SDS-PAGE; Affinity chromatography[35,36]
Monocyte chemoattractant protein-1 (MCP-1)Competitive binding to 3H-heparin by nitrocellulose membrane filtration and liquid scintillation[37]
Macrophage migration inhibitory factor (MIF)Flow cytometry of fluorescently labeled MIF on A549 cells after heparinase treatment[38]
Macrophage inflammatory protein-1α (MIP-1α)Affinity chromatography after heparinase treatment[39]
NKp46ELISA[40]
Platelet-derived growth factor (PDGF)Surface plasmon resonance;
Affinity chromatography
[41,42]
Platelet Factor 4Affinity co-electrophoresis[34]
P-selectinAffinity chromatography[36]
Receptor for advanced glycation end products (RAGE)Biotinylation and streptadivin-HRP detection[32,43]
Regulated on activation normal T cell expressed and secreted (RANTES) (CCL5)
(oligomerized, filamentous)
Surface plasmon resonance[24,44]
Receptor protein-tyrosine phosphatase-σ (RPTP-σ)Blot overlay assay probing agrin and collagen with cPTP-σ-conditioned medium following heparinase digestion[45]
Stromal cell-derived factor-1 (SDF-1)Flow cytometry of endothelial cells after heparinase treatment for bound SDF-1[46]
Transglutaminase-2Surface plasmon resonance[13]
Thrombospondin-1Surface plasmon resonance[13]
Vascular endothelial growth factor (VEGF)Metabolic labeling of pHEBO cells overexpressing VEGF189 followed by heparinase treatment, immunoprecipitation, and SDS PAGE[47]
CXCL, C-X-C motif ligand; IP-10, interferon-γ induced protein-10; MCP-1, monocyte chemoattractant protein-1; MIF, Macrophage migration inhibitory factor; MIP-1α, macrophage inflammatory protein-1α; PDGF, platelet-derived growth factor; RAGE, receptor for advanced glycation end products; RANTES, regulated on activation normal T cell expressed and secreted; RPTP-σ, receptor protein-tyrosine phosphatase-σ; SDF-1, stromal cell-derived factor-1.
Table 2. Proteins, molecules, pathogens, pathways, and therapeutics that modulate heparanase expression.
Table 2. Proteins, molecules, pathogens, pathways, and therapeutics that modulate heparanase expression.
AgentSpeciesFindingsReference
Transcription factors
c-MycHumanhTERT enabled binding of c-Myc to the heparanase promoter and increased heparanase mRNA expression[65]
Early growth response 1 (EGR1)Human and mouseDirect binding to the heparanase promoter resulted in activation of the heparanase promoter in PC-3, COLO397, and MCF-7 cells, and repression of the heparanase promoter in MM170 cells; EGR1 was recruited to the heparanase promoter upon glucose treatment of HEK293T cells[61,62,66,67,68]
Erythroblast Transformation Specific 1 (ETS1) and ETS2HumanDirect binding to the heparanase promoter increased heparanase mRNA expression[69]
GA-binding protein (GABP)HumanDirect binding to the heparanase promoter increased heparanase promoter activity[70,71]
NF-κBHuman and mouseNF-κB-deficient lung carcinoma cells produced less heparanase;
Inhibiting canonical NF-κB signaling blocked tumor necrosis factor-α (TNF-α)-induced upregulation of heparanase in endothelial cells; Chemotherapy treatment of multiple myeloma cells, hypoxia induction in pancreatic cancer cells, or infection with herpes simplex virus-1 (HSV-1) activated NF-κB to upregulate heparanase
[72,73,74,75,76]
p53Human and mouseDirect binding to the heparanase promoter reduced heparanase mRNA expression[77]
SnailMouseOverexpression of Snail in B16F1 cells increased heparanase mRNA expression[78]
specificity protein 1 (SP1) and SP3HumanDirect binding to the heparanase promoter increased heparanase promoter activity[70]
MicroRNA
miR-1258HumanmiRNA-1258 suppressed heparanase expression in breast cancer cells[79]
miR-1252-5p Overexpression of miR-1252-5p in multiple myeloma cells reduced heparanase mRNA and protein expression and activity[80]
Cytokines
IFN-γHumanTreatment of endothelial cells with IFN-γ increased heparanase mRNA expression and activity[81]
IL-1βHuman and mouseTreatment of endothelial cells with IL-1β increased heparanase mRNA expression[73,82]
IL-2MouseTreatment of purified NK cells with IL-2 induced expression of both pro-heparanase and the catalytically active heparanase protein, more so when also cultured with IL-15[50]
IL-10HumanIL-10 treatment of SUM149 breast cancer cells modestly increased heparanase mRNA expression[83]
IL-15MouseTreatment of purified NK cells with IL-15 induced expression of both pro-heparanase and the catalytically active heparanase protein, more so when also cultured with IL-12 and IL-18[50]
IL-17HumanTreatment of cervical cancer cells with IL-17 increased heparanase mRNA expression, and IL-17 knockdown reduced heparanase expression[84]
MCP-1MouseMCP-1 inhibition reduced glomerular heparanase expression[85]
TNF-αHuman, mouse and
bovine
Treatment of endothelial cells, U937 macrophages and colon cancer cells with TNF-α increased heparanase mRNA and protein expression[73,82,86,87,88]
Growth factors
bFGFHumanTreatment of lung cancer cells with bFGF induced heparanase mRNA expression[89]
HGFHumanTreatment of lung cancer cells with HGF induced heparanase mRNA expression;
HGF activated the PI3K/Akt/NF-κB signaling pathway and upregulated heparanase mRNA and protein expression
[89,90]
FGF23HumanTreatment of multiple myeloma cells with FGF23 increased heparanase mRNA expression, likely via Egr1 upregulation which was also upregulated[91]
PDGFHumanTreatment of lung cancer cells with PDGF induced heparanase mRNA expression[89]
VEGFHumanTreatment of endothelial cells with VEGF reduced heparanase expression;
Treatment of melanoma cells with recombinant VEGF increased heparanase mRNA expression, and VEGF knockdown decreased heparanase expression
[82,92]
Pathways
CD3/CD28 activationHuman and mouseHeparanase mRNA and protein increased with anti-CD3 and anti-CD28 antibody stimulation in mouse splenic (CD4/CD8) and human PBMC-derived T cells[62,93]
MEK/ERK pathwayHumanActivation of the MEK/ERK pathway increased heparanase expression[66]
Wnt signalingHamster and ZebrafishCHO-K1 cells treated with lithium chloride (LiCl; a Wnt signaling activator) modestly increased heparanase protein expression. Zebrafish embryos treated with LiCl increased heparanase mRNA expression[94]
Other biological molecules
HeparinHamsterCHO-K1 cells treated with heparin increased heparanase mRNA, protein, and enzymatic activity. Authors propose this may be via Wnt signaling[94]
LumicanMouseTreatment of B16F1 cells with recombinant lumican increased heparanase mRNA and protein expression; Treatment of Snail-overexpressing B16F1 cells (which resulted in increased heparanase expression) with recombinant lumican decreased heparanase mRNA expression but did not change protein expression[78]
Hormones, metabolites and other signaling molecules
Advanced Glycation End Products (AGEs)HumanExposure of HMVECs to AGEs increased heparanase mRNA and protein expression[95]
Basic proteinRatT lymphocytes degraded heparan sulfate (HS) after stimulation with Con A or basic protein[96,97,98]
BRAFHumanWild tpye BRAF suppressed ETS1 family of transcription factors, which suppressed heparanase promoter activity and mRNA expression. Mutant BRAF lost repression ability and heparanase mRNA expression was upregulated[71]
Endothelial nitric oxide synthase (eNOS)MouseIn a rat model of adriamycin nephropathy, the deletion or inhibition of eNOS induced heparanase mRNA and protein expression[99]
EstrogenHuman
and mouse
Estrogen treatment of ER-positive MCF-7 cells increased heparanase mRNA and protein expression via estrogen receptor signaling and estrogen response elements in the heparanase promoter. Upregulation of heparanase occured more so in low levels than high levels of estrogen;
Estrogen supplementation in MCF-7 implanted tumors in mice increased heparanase protein expression;
Treatment of cholangiocarcinoma cells with an estrogenic inducer upregulated heparanase mRNA expression;
Estrogen treatment of ER-positive EO771 breast cancer cells increased heparanase mRNA expression
[100,101,102,103]
High glucoseHuman and bovineGlucose-treated cells modestly increased heparanase protein expression. Heparanase mRNA expression, secretion, and activity increased upon glucose treatment[104,105,106]
Hypoxia (1% O2)HumanHypoxia-induced activation of NF-κB upregulated heparanase mRNA and protein expression[74]
Low density lipoprotein (LDL)HumanTreatment of endothelial cells with LDL increased heparanase mRNA expression[82]
Lipopolysaccharide (LPS)Human and mouseStimulation of B cells with LPS increased heparanase activity. Activating toll-like receptor 4 (TLR4) with LPS on PBMCs and cord blood cells increased heparanase mRNA expression;
LPS stimulation of PBMCs increased heparanase mRNA expression;
LPS stimulation of endothelial cells increased levels of enzymatically active heparanase
[63,98,107,108,109]
Oleic acidBovineTreatment of endothelial cells increased heparanase mRNA and protein expression, and was likely via Sp1[82]
Reactive oxygen species (ROS)Human and ratIn a rat model of adriamycin nephropathy, the depletion of hydroxyl radicals with DMTU reduced heparanase expression. Inducing mouse podocytes to generate free radicals and ROS increased heparanase mRNA and protein expression. Treatment of endothelial cells with ROS scavengers perturbed glucose-mediated heparanase expression[106,109,110]
Vitamin DRat and mouseVitamin D treatment reduced heparanase mRNA expression via initiating direct binding of the vitamin D receptor to the heparanase promoter[111]
Pathogens
Fusobacterium nucleatumHumanCo-culture of SSC-25 oral cancer cells with F. nucleatum increased heparanase expression[112]
Helicobacter pylori H. pylori infection of gastric cancer cells induced an upregulation of heparanase protein, which was dependent on MAPK signaling[113]
Pseudomonas aeruginosaMouseP. aeruginosa intracorneal infection in mice induced an upregulation of heparanase mRNA and enzymatically active protein in the cornea. This was from both infiltrating immune cells as well as from the corneal epithelium[114]
Streptococcus pneumoniaeMouseIntranasal S. pneumoniae infection in mice increased heparanase protein expression[115]
Bovine herpes virusHumanHeparanase mRNA was upregulated upon epithelial cell infection in vitro[75]
SARS-CoV-2HumanCOVID-19 patients displayed elevated heparanase activity and soluble HS levels in the plasma; Increase shed syndecan-1 was observed[116,117]
CytomegalovirusHumanHeparanase mRNA was upregulated upon fibroblast cell infection in vitro[75]
Dengue virusHumanDengue virus protein NS1 upregulated heparanase protein in endothelial cells, and upregulation was found to be macrophage inhibitory factor-dependent[118,119]
HSV-1HumanHeparanase mRNA and protein were upregulated upon HSV-1 infection through NF-κB activation[75,120]
HSV-2HumanHeparanase mRNA was upregulated upon epithelial cell infection in vitro[75]
Porcine reproductive and respiratory syndrome virusPigPiglets infected with PRSSV in vivo increased heparanase mRNA expression in alveolar macrophages
Cells infected in vitro with PRSSV increased heparanase mRNA and protein expression
[121,122]
Pseudorabies virusHumanHeparanase mRNA was upregulated upon epithelial cell infection in vitro[75]
Therapeutics
BortezomibHumanTreatment of myeloma cells increased heparanase mRNA and protein expression[76]
CarfilzomibHumanTreatment of myeloma cells increased heparanase mRNA expression[76]
CisplatinHumanTreatment of mesothelioma cells, gastric cancer cells, and J774 macrophages increased heparanase mRNA expression[123]
ClarithromycinHumanClarithromycin blocked the upregulation of heparanase mRNA induced by bFGF[89]
DoxorubicinHumanTreatment of myeloma cells, gastric cancer cells, and J774 macrophages increased heparanase protein expression[76,123]
ErythromycinHumanErythromycin blocked the upregulation of heparanase mRNA induced by PDGF and HGF[89]
PaclitaxelHumanTreatment of gastric cancer cells with paclitaxel increased heparanase mRNA expression[123]
phorbol-12-myristate-13-acetate (PMA)Human and mouseHeparanase mRNA expression increased upon stimulation with PMA ionomycin in EL4 T lymphocytes. HS degradation increased after PMA stimulation in neutrophils, human umbilical vein endothelial cells (HUVECs), and platelets; Heparanase mRNA, protein, and activity increased in human NK cells after activation with B-LCL cells, IL-2 and PMA, and ionomycin[50,61,124]
Poly(I:C)MousePoly(I:C) stimulation in vivo increased heparanase activity in splenic NK cells[50]
RadiationHuman
and
rat
Human epidermal keratinocytes exposed to UVB radiation exhibited increased heparanase enzymatic activity and detectable levels of the 50 kDa active subunit;
Rats with experimental liver cirrhosis showed an increase in heparanase precursor protein in liver and serum after treatment with partial liver radiation
[125,126]
TamoxifenHumanTreatment of MCF-7 cells with high concentration of tamoxifen inhibited estrogen-induced heparanase expression; Tamoxifen treatment of MCF-7 cells and T47D cells increased heparanase mRNA expression[100,102]
Miscellaneous
CeruleinMouseInjection of cerulein into mice increased heparanase mRNA expression and enzymatic activity in pancreatic tissue extracts[127]
AGE, Advanced glycation end product; DMTU, dimethylthiourea; eNOS, endothelial nitric oxide synthase; ERK, extracellular signal-regulated kinase; ETS, E26 transformation-specific or E-twenty-six; GABP, GA-binding protein; HGF, hepatocyte growth factor; HMVEC, human microvascular endothelial cell; HSV, herpes simplex virus; hTERT, telomerase reverse transcriptase; LDL, low-density lipoprotein; LPS, lipopolysaccharide; MEK, mitogen-activated protein kinase; MCP, monocyte chemoattractant protein; PBMC, peripheral blood mononuclear cell; PI3K, phosphoinositide 3-kinases; SP, specificity protein; TLR4, Toll-like receptor 4; WT, wild type.
Table 3. Genes and proteins that are regulated by heparanase.
Table 3. Genes and proteins that are regulated by heparanase.
Gene/ProteinObservation/MechanismRelated Disease/FunctionReference
Genes
AromataseThe expression of aromatase was decreased in heparanase- knockout obese mice. Heparanase was required for the activation of fatty acid-stimulated macrophages to induce aromatase in adipose stromal cellsObesity-associated
breast cancer progression
[103]
Bcl-XL (Bcl2l1)Increased expression of Bcl-XL in heparanase overexpressing transgenic mice with dextran sulfate sodium (DSS)-induced colitis was regulated by NF-κBUlcerative colitis[131]
Caspase-1Silence of heparanase and heparanase inhibitor (SST0001) blocked caspase 1 expression in human kidney cellsAcute kidney injury/M1 macrophage polarization[182]
Cathepsin LInduction of acute kidney injury in heparanase-transgenic mice enhanced the expression of cathepsin L mRNA. Pre-treatment with heparanase inhibitor PG545 reduced the expression of cathepsin LEpithelial-mesenchymal transition (EMT)/Acute kidney injury[179]
CD44siRNA knockdown of heparanase in SUM149 breast cancer cells reduced mRNA expression of CD44Breast cancer[83]
c-Fos
(AP-1)
The expression of c-Fos was decreased in heparanase-knockout macrophages and adding exogenous heparanase enhanced c-Fos expression. Heparanase regulated the gene expression of c-Fos through Erk, p38, and JNK signaling pathwayTumor/Induction of cytokine expression[153]
Collagen-ITreatment of KATO-III gastric cancer cells with heparanase inhibitor suramin exhibited reduced expression of collagen-IEMT/Gastric ring cell adenocarcinoma[181]
Cox-2Cox-2 mRNA expression was increased in heparanase overexpressing transgenic mice with DSS-induced colitis and was regulated by NF-κBUlcerative colitis[131]
Heparanase upregulated the mRNA expression of Cox-2 in cancer cellsTumor/Promoting angiogenesis[187]
CXCR-4mRNA expression of CXCR-4 was decreased in gastric cancer cell KATO-III treated with heparanase inhibitor suraminEMT/Gastric ring cell adenocarcinoma[181]
EGR1Overexpression of heparanase increased Egr1 mRNA expressionModulation of EGR gene expression[188]
EGR2Overexpression of heparanase increased Egr2 mRNA expressionModulation of EGR gene expression[188]
E-CadherinThe expression of Epithelial marker E-cadherin was increased in KATO-III gastric cancer cells treated with heparanase inhibitor suraminEMT/Gastric ring cell adenocarcinoma[181]
Endothelin-1 (ET-1)Induction of acute kidney injury in heparanase-transgenic mice enhanced the expression of ET-1mRNA. Pre-treatment with heparanase inhibitior PG545 reduced the expression of ET-1EMT/Acute kidney injury[179]
FGF/bFGFHeparanase activated HIF1 pathway which led to reduced mRNA expression level of bFGF in heparanase knockdown cells and elevated mRNA expression level of bFGF in heparanase overexpressing-cellsCervical cancer[177]
FGF-2Treatment of human osteoblasts with heparin, a heparanase inhibitor, inhibited mRNA FGF2 expressionGrowth of osteoblasts[180]
Fibronectin (FN)Heparanase-transgenic mice displayed remarkable upregulation of FN during acute kidney injury. Pre-treatment with heparanase inhibitor PG545 abolished the increased expression of FN in heparanase-transgenic miceEMT/Acute kidney injury[179]
Heparanase-silenced cells showed reduced FN expression;
Renal tissue extracts from mice with acute kidney injury treated with Roneparstat showed reduced FN expression
EMT/Acute kidney injury[182,189]
Hepatocyte growth factor (HGF)Addition of either recombinant or chemotherapy-generated soluble heparanase increased HGF mRNA expression. Immunodepletion or addition of heparanase inhibitor diminished the increased expression of HGF gene. Upregulation of HGF expression by heparanase was independent of heparanase enzyme activityTumor progression[76,176]
HIF-1mRNA expression level of HIF1 was reduced in heparanase knockdown cells and increased in heparanase-overexpressing cellsCervical cancer[177]
HIF-2αKnockdown of heparanase in HUVEC cells reduced HIF-2α expressionTumor angiogenesis[190]
IL-1βHS fragments generated by heparanase activated TLR4, MyD88, and NF-κB to upregulate IL-1β mRNAInflammation[120,152]
The expression of IL-1β in macrophages isolated from heparanase-knockout mice was significantly reduced compared to macrophages isolated from wild type mice. Heparanase regulated IL-1β expression through Erk, p38, and JNK signaling pathwayTumor/Regulation of cytokine expression in macrophage[153]
Increased expression of IL-1β in heparanase overexpressing transgenic mice with colitis-associated carcinomaColitis-associated tumor/Induction of NK-κB activation/Macrophage activation[131]
Heparanase upregulated the expression of IL-1β in PMA-activated U937 macrophages. Treatment cells with heparanase inhibitor SST0001 reduced IL-1β expressionAcute kidney injury/M1 macrophage polarization[182]
IL-5House dust mite (HDM)-induced allergic inflammation in heparanase deficient mice reduced mRNA expression of IL-5 in lung cellsAllergic inflammation/Recruitment of eosinophils and mucus-secreting airway epithelial cells[51]
IL-6HS fragments generated by heparanase activated TLR4, MyD88, and NF-κB to upregulate IL-6Inflammation[152]
The expression of IL-6 in macrophages isolated from heparanase deficient mice was significantly reduced compared to macrophages isolated from wild type mice. Heparanase regulated IL-6 expression through Erk, p38, and JNK signaling pathwaysTumor/Regulation of cytokine expression in macrophage[153]
IL-6 mRNA expression was increased in heparanase transgenic mice with DSS-inducedcolitis. LPS-treated mouse peritoneal macrophages increased mRNA expression of IL-6 in the presence of recombinant enzymatically active heparanaseUlcerative colitis/Induction of NK-κB activation/Macrophage recruitment and activation[131]
Induction of acute kidney injury in heparanase-transgenic mice enhanced the expression of mRNA IL-6. Pre-treatment with heparanase inhibitior PG545 reduced the expression of IL-6EMT/Acute kidney injury[179]
Heparanase upregulated the expression of IL-6 in PMA-activated U937 macrophage cells. Treatment of cells with heparanase inhibitor SST0001 reduced IL-6 expressionAcute kidney injury/M1 macrophage polarization[182]
Heparanase induced the expression of IL-6 by fatty acid-stimulated macrophages in a dose-dependent mannerObesity-associated breast cancer[103]
IL-6 expression was increased in heparanase-knockout macrophages treated with exogenous heparanase and chemotherapyTumor Growth/Induction of pro-inflammatory cytokine expression by chemotherapy-treated macrophage[123]
IL-8HS fragments generated by heparanase activated TLR4, MyD88, and NF-κB to upregulate IL-8Inflammation[152,183]
IL-10IL-10 mRNA expression was reduced in chemotherapy-treated macrophages isolated from heparanase knockout miceTumor Growth/Induction of pro-inflammatory cytokine expression by chemotherapy-treated macrophage[123]
HS fragments generated by heparanase activated TLR4, MyD88, and NF-κB to upregulate IL-10Inflammation[152]
The expression of IL-10 in macrophages isolated from heparanase deficient mice was significantly reduced compared to macrophages isolated from wild type mice. Heparanase regulated IL-10 expression through Erk, p38, and JNK signaling pathwayTumor/Regulation of cytokine expression in macrophage[153]
Inhibition of heparanase with SST0001 reduced IL-10 mRNA expression in macrophagesAcute kidney injury/M1 macrophage polarization[182]
IL-13(HDM-induced allergic inflammation in heparanase deficient mice reduced mRNA expression of IL-13 in lung cellsAllergic inflammation/Recruitment of eosinophils and mucus-secreting airway epithelial cells[51]
IL-12p53LPS-treated mouse peritoneal macrophages increased mRNA expression of IL-12p53 in the presence of recombinant enzymatically active heparanaseUlcerative colitis/Macrophage activation[131]
IL-17ASilencing of heparanase resulted in a significant decrease in the mRNA expression of IL-17A in human cervical cancer cell lines HeLa and SiHaPromoting tumor angiogenesis, cell proliferation, and invasion in cervical cancer[84]
Inducible nitric oxide synthase (iNOS)Heparanase upregulated the expression of iNOS in PMA-activated U937 cells. Treatment of cells with heparanase inhibitor SST0001 suppressed iNOS expressionAcute kidney injury/M1 macrophage polarization[182]
Lysozyme 1Heparanase-knockout mice showed less lysozyme 1 expressionTumor/Macrophage cytotoxic activity is decreased in the absence of heparanase[153]
MCP-1/CCL-2Non-enzymatic heparanase in colorectal cancer cell lines could upregulate the expression of MCP-1Promoting extravasation of colon carcinoma cells[183]
MIP-2
(CXCL2)
Macrophages isolated from heparanase-deficient mice and mice treated with heparanase-neutralizing antibodies exhibited reduced MIP-2 expressionTumor/Regulation of cytokine expression in macrophage[153]
MIP-2 mRNA expression was reduced in chemotherapy-treated macrophages isolated from heparanase knockout miceTumor Growth/Induction of pro-inflammatory cytokine expression by chemotherapy-treated macrophage[123]
Mixed Lineage Kinase Domain Like Pseudokinase (MLKL)Transwell co-culture of heparanase-silenced hepatocellular carcinoma (HCC) cells with HUVECs protected HUVECs from MLKL mRNA and protein upregulation and necroptosis
In transwell co-cultures of heparanase-overexpressing HCC cells and HUVECs, HUVECs displayed higher MLKL protein expression after co-culture compared to controls
Necroptosis[191]
matrix metalloprotease-2 (MMP-2)The mRNA expression of MMP-2 was decreased in the kidney of heparanase deficient miceAllergen-induced inflammation/DC migration[51]
Human melanoma cells deficient in heparanase exhibited increased MMP-2 expressionMelanoma progression[169]
Inhibiting heparanase with either PG545 or PI-88 in patient-derived explants of normal mammary tissue increased MMP-2 mRNA expressionTissue density and breast cancer[192]
siRNA knockdown of heparanase in SUM149 breast cancer cells reduced MMP-2 mRNA expressionBreast cancer[83]
MMP-9Addition of recombinant or chemotherapy-generated soluble heparanase elevated the expression of MMP-9 in myeloma cells. Chemotherapeutic induction of MMP-9 required heparanase through Erk phosphorylationTumor progression[76,79,184]
The gene expression level of MMP-9 in heparanase-silenced human kidney 2 (HK2) cells was lower than wild type cellsRenal fibrosis[193]
Heparanase upregulated the expression of MMP-9 by its HS-degrading activity and stimulating HAT activityMyeloma tumor/Upregulation of HAT activity[168]
Human melanoma cells deficient in heparanase exhibited increased MMP-9 expressionMelanoma progression[169]
MMP-14The mRNA expression of MMP-14 was decreased in the liver of heparanase deficient miceAllergen-induced inflammation/DC migration[51]
Inhibiting heparanase with either PG545 or PI-88 in patient-derived explants of normal mammary tissue increased MMP-14 mRNA expressionTissue density and breast cancer[192]
MMP-25The mRNA expression of MMP25 was increased in the spleen but decreased in mouse bone marrow-derived DCs and Langerhans cells from heparanase deficient miceAllergen-induced inflammation/DC migration[51]
NANOGmRNA expression of NANOG was decreased in KATO-III gastric cancer cells treated with heparanase inhibitor suraminEMT/Gastric ring cell adenocarcinoma[181]
OCT3/4mRNA expression of OCT3/4 was decreased in KATO-III gastric cancer cells treated with heparanase inhibitor suraminEMT/Gastric ring cell adenocarcinoma[181]
P21Heparanase downregulated p21 in colon carcinoma cells through its enzymatic activity and involved TLRs and NF-κB signalingColon carcinoma/Modification of cell cycle[194]
P38In transwell co-cultures of heparanase-silenced HCC cells and HUVECs, HUVECs displayed lower p38 mRNA and phosphorylated protein expression after co-culture compared to controlsNecroptosis[191]
PDK2Nuclear heparanase regulated the mRNA expression of PDK2 through HAT activation. Depletion of heparanase reduced the expression of PDK2 mRNAGlucose metabolism[186]
Pentraxin 3 (PTX-3)Human melanoma cells deficient in heparanase exhibited increased PTX-3 expressionMelanoma progression[169]
Receptor interacting protein kinase 1 (RIPK1) and RIPK3Transwell co-cultures of heparanase-silenced HCC cells with HUVECs protected HUVECs from RIPK1 and RIPK3 mRNA and protein upregulation and necroptosis.
In transwell co-cultures of heparanase-overexpressing HCC cells and HUVECs, HUVECs displayed higher RIPK1 and RIPK3 protein expression after co-culture compared to controls
Necroptosis[191]
SDF-1 (CXCL-12)SDF-1 expression was reduced in heparanase-deficient macrophages. Heparanase regulated SDF-1 expression through Erk, p38, and JNK signaling pathwayTumor/Promoting phagocytic capacity of macrophages[153]
SERPINE1Heparanase regulated HAT activity, leading to upregulation of SERPINE1Inflammation[186]
SlugKATO-III gastric cancer cells treated with heparanase inhibitor suramin reduced Slug mRNA expressionEMT/Gastric ring cell adenocarcinoma[181]
α-SMATreatment of KATO-III gastric cancer cells with heparanase inhibitor suramin exhibited reduced expression of α-SMAEMT/Gastric ring cell adenocarcinoma[181]
Heparanase-overexpressing micedisplayed remarkable upregulation of α-SMA during acute kidney injury. Pre-treatment with heparanase inhibitor PG545 abolished the increased expression of α-SMA in hpse-tg miceEMT/Acute kidney injury[179]
Heparanase-silenced cells showed reduced α-SMA expressionEMT/Acute kidney injury[182,189]
SnailHeparanase-silenced cells showed reduced Snail expressionEMT/Acute kidney injury[182]
Syndecan-1Inhibiting heparanase with either PG545 or PI-88 in patient-derived explants of normal mammary tissue reduced syndecan-1 mRNA expressionTissue density and breast cancer[192]
In transwell co-cultures of heparanase-silenced HCC cells and HUVECs, HUVECs displayed lower syndecan-1 mRNA and protein expression after co-culture compared to controls.
In transwell co-cultures of heparanase-overexpressing HCC cells and HUVECs, HUVECs displayed higher syndecan-1 mRNA and protein expression after co-culture compared to controls
Necroptosis[191]
Tissue factor (TF)mRNA expression levels of TF were elevated in heparanase transfected breast carcinoma cells and transgenic mice over-expressing heparanase. Exogenous addition of heparanase also induced TF expression in human promyelocytic leukemia cells. Heparanase induced TF expression via inducing p38 signaling non-enzymaticallyBlood coagulation[195]
Human melanoma cells deficient in heparanase exhibited increased TF expressionMelanoma progression[169]
Transforming growth factor (TGF)-β/TGFβ1Gene expression levels of TGF-β was decreased in the heparanase-silenced tubular cellsEMT/Renal fibrosis[178,189]
Induction of acute kidney injury in heparanase-transgenic mice enhanced the expression of TGF-β mRNA. Pre-treatment with heparanase inhibitior PG545 abolished the elevation in TGF-βEMT/Acute kidney injury[179]
Heparanase inhibitor suramin down-regulated TGFβ-1 expression in KATO-III gastric cancer cellsEMT/Gastric ring cell adenocarcinoma[181]
TLR-2The expression of TLR-2 in macrophages isolated from heparanase deficient mice and in macrophages isolated from mice treated with heparanase-neutralizing antibodies was significantly reduced. Heparanase regulated TLR2 expression through Erk, p38, and JNK signaling pathwayTumor/Macrophage activation and function in tumorigenesis[153]
TLR-4The expression of TLR-4 on macrophages was upregulated in the presence of heparanase but was reduced when cells were treated with heparanase inhibitor SST0001Acute kidney injury/Regulation of macrophage polarization[182]
TNF-αTNF-α expression was reduced in macrophages isolated from heparanase-knockout mice and in macrophages isolated from mice treated with heparanase-neutralizing antibodies. Heparanase regulated TNF-α expression through Erk, p38, and JNK signaling pathwayTumor/Macrophage activation and function in tumorigenesis[153]
Heparanase overexpressing transgenic mice expressed more TNF-α during DSS-induced colitis through NF-κB signaling. LPS-treated mouse peritoneal macrophages increased mRNA expression of TNF-α in the presence of recombinant enzymatically active heparanaseUlcerative colitis/Induction of NK-κB activation/Macrophage recruitment and activation[131]
Induction of acute kidney injury in heparanase-transgenic mice enhanced the expression of TNF-α mRNA. Pre-treatment with heparanase inhibitior PG545 reduced the expression of TNF-αEMT/Acute kidney injury[179]
Human melanoma cells deficient in heparanase exhibited increased TNF-α expressionMelanoma progression[169]
Heparanase upregulated the expression of TNF-α in PMA-activated U937 macrophage cells. Treatment of cells with heparanase inhibitor SST0001 reduced TNF-α expressionAcute kidney injury/M1 macrophage polarization[182]
HS fragments generated by heparanase activated TLR4, MyD88, and NF-κB to upregulate TNF-αInflammation[152]
TNF-α mRNA expression was reduced in chemotherapy-treated macrophages isolated from heparanase knockout mice
In transwell co-cultures of heparanase-silenced HCC cells and HUVECs, HUVECs displayed lower TNF-α mRNA and protein expression compared to controls
Tumor Growth/Induction of pro-inflammatory cytokine expression by chemotherapy-treated macrophage[123]
In transwell co-cultures of heparanase-overexpressing HCC cells and HUVECs, HUVECs displayed higher TNF-α mRNA and protein expression after co-culture compared to controlsNecroptosis[191]
TNF-α receptor (TNFR)In transwell co-cultures of heparanase-silenced HCC cells and HUVECs, HUVECs displayed lower TNFR mRNA and protein expression compared to controlsNecroptosis[191]
TNFR-associated death domain protein (TRADD)In transwell co-cultures of heparanase-silenced HCC cells and HUVECs, HUVECs displayed lower TRADD mRNA and protein expression after co-culture compared to controlsNecroptosis[191]
Vascular cell adhesion molecule 1 (VCAM-1)Heparanase regulated HAT activity, leading to upregulation of VCAM-1Inflammation[186]
VEGFHeparanase overexpression or exogenous addition led to the enhanced expression of VEGF. Heparanase regulated the expression of VEGF by mediating the activation of SRC family membersPromoting angiogenesis in tumor[175]
Heparanase upregulated the expression of VEGF through its HS-degrading activity and stimulating the HAT activityTumor phenotype/Upregulation of HAT activity[168]
Heparanase regulated the expression of VEGF via activating HIF1 pathwayCervical cancer[177]
Addition of recombinant or chemotherapy-generated soluble heparanase elevated the expression of VEGF in myelomaTumor progression[76]
Heparanase overexpression in melanoma cell lines increased the expression of VEGF mRNA. Downregulation of heparanase via anti-heparanase siRNA transfection resulted in a significant reduction of VEGF mRNA expression in melanoma cell linesMelanoma progression[92]
VEGF-AReduced VEGF-A expression was observed in macrophages isolated from heparanase-knockout mice and in macrophages isolated from mice treated with heparanase-neutralizing antibodiesTumor/Macrophage activation and function in tumorigenesis[153]
Heparanase regulated HAT activity, leading to upregulation of VEGF-AAtherosclerosis/Glucose Metabolism[186]
VEGF-COverexpression of heparanase increased VEGF-C mRNA expressionPancreatic cancer/Facilitating cell invasion[185]
VimentinKATO-III gastric cancer cells exhibited reduced Vimentin expression after treating with heparanase inhibitor suraminEMT/Gastric ring cell adenocarcinoma[181]
Heparanase-overexpressing mice displayed remarkable upregulation of vimentin during acute kidney injury. Pre-treatment with heparanase inhibition abolished the increased expression of vimentin in heparanase-overexpressing mice.EMT/Acute kidney injury[179]
Heparanase-silenced cells reduced vimentin expressionEMT/Acute kidney injury[189]
WDR5Upon paclitaxel treatment, WDR5 expression was induced in wild type but not heparanase-knockout macrophages, but could be rescued with exogenous heparanase
Heparanase was required for the expression of WDR5 in macrophages
Tumor Growth/Induction of pro-inflammatory cytokine expression by chemotherapy-treated macrophage[123]
Proteins
bFGFbFGF protein expression was decreased in heparanase knockdown cell and increased in heparanase overexpressing cells via activating HIF1 pathwayCervical cancer[177]
BLCBLC expression was reduced in macrophages isolated from heparanase-knockout miceTumor/Macrophage activation and function in tumorigenesis[153]
Caspase-1Heparanase-silenced and heparanase inhibitor SST0001-treated cells reduced caspase-1 expressionAcute kidney injury/M1 macrophage polarization
Cox-2Heparanase upregulated the mRNA expression of Cox-2 in cancer cellsTumor/Promoting angiogenesis[187]
CXCL1 (KC)Administration of heparanase increased CXCL1 level in mouse serumThoracoabdominal aortic aneurysm/Systemic Inflammation[196]
CXCL1 expression was reduced in macrophages isolated from heparanase-knockout miceTumor/Macrophage activation and function in tumorigenesis[153]
Heparanase-stimulated colon cancer cells released CXCL1Colon cancer[183]
FGF21Heparanase-overexpressing mice had higher FGF21 expression in the blood plasma compared to wild type miceDiabetes/Glucose homeostasis[197]
FibrinogenHigh dose heparanase-derived peptides induced a decrease in the level of fibrinogenCoagulopathy and wound healing/Activation of the coagulation system[198]
FibronectinProtein expression of fibronectin was increased in heparanase-overexpressing mice with acute kidney injury but decreased when pre-treating the mice with heparanase inhibitor PG545EMT/Acute kidney injury[179]
FXaHeparanase-derived peptides enhanced the level of FXa probable through interacting with TFCoagulopathy and wound healing/Activation of the coagulation system[198]
Hepatocyte growth factor (HGF)Addition of soluble heparanase or increased heparanase expression upregulated HGF expression in myeloma cell lines. Knockdown of heparanase reduced HGF expressionTumor progression[76,176]
HIF1HIF1 protein expression was decreased in heparanase knockdown cells and increased in heparanase overexpressing cells via HIF1 pathwayCervical cancer[177]
ICAM-1ICAM-1 expression was significantly increased in heparanase overexpressing human breast cancer cell lines. Likewise, the expression of ICAM-1 was decreased in heparanase-knockout cell linesCancer metastasis/Promotion of cell cluster formation by modulating adhesion molecules[197]
IL-1Addition or overexpression of heparanase upregulated the expression of IL-1Atherosclerosis/Macrophage activation[199]
IL-1βAdministration of heparanase increased IL-1β level in mouse serumThoracoabdominal aortic aneurysm/Systemic Inflammation[196]
Heparanase upregulated the expression of IL-1β in macrophages. Treatment of cells with heparanase inhibitor SST0001 reduced IL-1β expressionAcute kidney injury/M1 macrophage polarization[182]
Heparanase via its enzymatic activity upregulated IL-1β through TLR4 signalingInflammation[152]
IL-4IL-4 expression was reduced in lung cells isolated from heparanase deficient mice with HDM-induced allergic inflammationAllergic inflammation/Recruitment of eosinophils and mucus-secreting airway epithelial cells[51]
Administration of heparanase upregulated the expression of IL-4 in mouse immune cellsAutoimmune encephalitis/inhibition of inflammation[200]
IL-5IL-5 expression was reduced in lung cells isolated from heparanase deficient mice with HDM-induced allergic inflammationAllergic inflammation/Recruitment of eosinophils and mucus-secreting airway epithelial cells[51]
IL-6Administration of heparanase increased IL-6 level in mouse serumThoracoabdominal aortic aneurysm/Systemic Inflammation[196]
Heparanase via its enzymatic activity upregulated IL-6 through TLR4 signalingInflammation[152]
Administration of heparanase upregulated the expression of IL-6 in mouse immune cellsAutoimmune encephalitis/Inhibition of inflammation[200]
Addition of heparanase enhanced the expression of IL-6 in fatty acid-stimulated macrophagesObesity-associated breast cancer[103]
IL-8Heparanase enhanced IL-8 expressionColon cancer[183]
Heparanase upregulated IL-8 expression via its enzymatic activityInflammation[152]
IL-10Administration of heparanase increased IL-10 level in mouse serumThoracoabdominal aortic aneurysm/Systemic Inflammation[196]
Heparanase upregulated IL-10 expression via its enzymatic activityInflammation[152]
Administration of heparanase upregulated the expression of IL-10 in mouse immune cells
Autoimmune encephalitis/Inhibition of inflammation[200]
IL-12Administration of heparanase downregulated the expression of IL-12 in mouse immune cellsAutoimmune encephalitis/Inhibition of inflammation[200]
IL-17ASilencing of heparanase resulted in a significant decrease in protein expression of IL-17A in human cervical cancer cell lines HeLa and SiHaPromoting tumor angiogenesis, cell proliferation, and invasion in cervical cancer[84]
iNOSHeparanase upregulated the expression of iNOS in macrophages. Treatment of cells with the heparanase inhibitor SST0001 reduced iNOS expressionAcute kidney injury/M1 macrophage polarization[182]
MCP-1Addition or overexpression of heparanase upregulated the expression of MCP-1Atherosclerosis/Macrophage activation
Thoracoabdominal aortic aneurysm/Systemic
[199]
Administration of heparanase increased MCP-1 level in mouse serumInflammation[196]
Heparanase-stimulated colon cancer cells released MCP-1Colon cancer[183]
Heparanase upregulated MCP-1 via TLR4 signalingInflammation[152]
Obese heparanase knockout mice showed less MCP-1 expression compared to obese wild type miceObesity-associated breast cancer progression[103]
MIP-2
(CXCL2)
MIP-2 expression was reduced in macrophages isolated from heparanase-knockout miceTumor/Macrophage activation and function in tumorigenesis[153]
MMP-9Addition or overexpression of heparanase upregulated the expression of MMP-9Atherosclerosis/Macrophage activation[199]
NF-κB (p65)Knockdown of heparanase led to increased expression of nuclear NF-κB in melanoma cell linesMelanoma progression[169]
P21Heparanase downregulated p21 in colon carcinoma cells through its enzymatic activity and involved TLRs and NF-κB signalingColon carcinoma/Modification of cell cycle[194]
α-SMAProtein expression of α-SMA was increased in heparanase-overexpressing mice with acute kidney injury but decreased when pre-treating the mice with heparanase inhibitor PG545EMT/Acute kidney injury[179]
TLR2Heparanase knockout cells expressed less TLR2 proteinTumor/Macrophage activation and function in tumorigenesis[153]
TNF-αTNF-α expression was reduced in macrophages isolated from heparanase-knockout miceTumor/Macrophage activation and function in tumorigenesis[153]
Increased expression of TNF-α in heparanase overexpressing transgenic mice with DSS-induced colitisUlcerative colitis/Induction of NK-κB activation[131]
Addition or overexpression of heparanase increased the expression of TNF-αAtherosclerosis/Macrophage activation[199]
Heparanase upregulated TNF-α via TLR4 signaling.
Heparanase deficiency reduced the expression of TNF-α in macrophages
Inflammation
Obesity-associated breast cancer progression
[103,152]
VEGFVEGF protein expression was decreased in heparanase knockdown cells and increased in heparanase overexpressing cells via activating the HIF1 pathwayCervical cancer[177]
Heparanase overexpression led to the enhanced expression of VEGF. Heparanase regulated the expression of VEGF by mediating the activation of SRCTumor vascularity[175]
VEGF expression was increased in heparanase overexpressing melanoma cell lines and decreased in heparanase downregulated cellsMelanoma progression[92]
VimentinProtein expression of vimentin was increased in heparanase-overexpressing mice with acute kidney injury but decreased when pre-treating the mice with heparanase inhibitor PG545EMT/Acute kidney injury[179]
EGR, early growth response; HCC, hepatocellular carcinoma; HIF, hypoxia inducible factor.
Table 4. Heparanase regulates protein phosphorylation.
Table 4. Heparanase regulates protein phosphorylation.
ProteinObservation/MechanismRelated Disease/FunctionReference
AKTInhibition of heparanase reduced AKT phosphorylationBreast Cancer Brain Metastasis[79]
High expression of heparanase in myeloma cell lines led to increased AKT phosphorylation. This was blocked by treating cells with heparanase inhibitor SST0001Tumor progression[76]
Epidermal growth factor receptor (EGFR)Inhibition of heparanase reduced EGFR phosphorylationBreast Cancer Brain Metastasis[79]
Heparanase enhanced the phosphorylation level of EGFR in carcinoma cellsTumor progression[205]
Heparanase released HS via shedding syndecan-1 which induced EGFR phosphorylationColorectal cancer[151]
ERKThe level of phosphorylated ERK was increased in heparanase overexpressing neural stem and progenitor cells during differentiationPromoting Embryonic stem cell differentiation into Oligodendrocytes[207]
Addition of exogenous heparanase induced ERK phosphorylation in macrophagesInducing cytokine expression in macrophage[153]
High expression of heparanase in myeloma cell lines led to increased ERK phosphorylation. The increased phosphorylation of ERK was blocked in cells treated with heparanase inhibitor SST0001Tumor progression[76]
Focal-adhesion kinase (FAK)The phosphorylation of FAK was elevated in heparanase-overexpressing breast cancer cell lines. Likewise, the phosphorylation of FAK was decreased in heparanase-knockout cell lines. Heparanase promoted cell cluster formation by regulating FAK-Src-paxillin pathwayPromotion of cell cluster formation/Tumor metastasis[197]
IκBα/IκBHeparanase enhanced phosphorylation of IκBα in heparanase overexpressing mice suffering colitis-associated tumorsUlcerative colitis/Induction of NK-κB activation[131]
IκB phosphorylation was decreased in pancreas tissues of heparanase-overexpressing mice treated with heparanase inhibitor PG545Acute pancreatitis[127]
JNKAddition of exogenous heparanase induced JNK phosphorylation in macrophagesInducing cytokine expression in macrophage[153]
JNK phosphorylation was decreased in macrophages isolated from heparanase knockout miceTumor Growth/Induction of pro-inflammatory cytokine expression by chemotherapy-treated macrophage[123]
MEKHeparanase induced MEK phosphorylation via releasing HS of syndecan-1Colorectal cancer[151]
p38Addition of exogenous heparanase enhanced p38 phosphorylation in macrophagesInducing cytokine expression in macrophage[153]
Heparanase-overexpressing cells induced p38 phosphorylationPromoting tumor angiogenesis[175]
p65 NF-κBIncreased nuclear p65 phosphorylation was detected in heparanase overexpressing mice treated with DSS to induce colitis-associated tumorsUlcerative colitis/Induction of NK-κB activation[131]
PaxillinThe phosphorylation of paxillin was elevated in heparanase-overexpressing breast cancer cell lines. In contrast, the phosphorylation of paxillin was decreased in heparanase-knockout cell lines. Heparanase promoted cell cluster formation by regulating FAK-Src-paxillin pathwayPromotion of cell cluster formation/Tumor metastasis[197]
SRCThe phosphorylation of SRC was increased in heparanase-overexpressing breast cancer cell lines. In contrast, the level of SRC phosphorylation was decreased in heparanase-knockout cell lines. Heparanase promoted cell cluster formation by regulating FAK-Src-paxillin pathwayPromotion of cell cluster formation/Tumor metastasis[197]
Inactive heparanase stimulated SRC phosphorylationTumor angiogenesis[175]
Heparanase enhanced the phosphorylation level of SRC in carcinoma cellsTumor progression[205]
Signal Transducer and Activator of Transcription (STAT)Heparanase increased nuclear STAT phosphorylationTumor progression[205]
STAT3Higher number of cells positive for nuclear-localized pSTAT3 were observed in heparanase-overexpressing transgenic miceModulator of tumor-promoting chronic inflammation[131]
Heparanase enhanced STAT3 phosphorylationTumor progression[205]
Reduced STAT3 phosphorylation was observed in obese heparanase knockout miceObesity-associated breast cancer progression[103]
STAT5bHeparanase enhanced STAT5b phosphorylationTumor progression[205]
VCAM-1, vascular cell adhesion molecule 1; SERPINE1, plasminogen activator inhibitor type 1; VEGFA, vascular endothelial growth factor A; FXa, activated factor X; TF, tissue factor; TGF, transforming growth factor; PDK2, pyruvate dehydrogenase kinase 2; HIF1, hypoxia inducible factor.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mayfosh, A.J.; Nguyen, T.K.; Hulett, M.D. The Heparanase Regulatory Network in Health and Disease. Int. J. Mol. Sci. 2021, 22, 11096. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222011096

AMA Style

Mayfosh AJ, Nguyen TK, Hulett MD. The Heparanase Regulatory Network in Health and Disease. International Journal of Molecular Sciences. 2021; 22(20):11096. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222011096

Chicago/Turabian Style

Mayfosh, Alyce J., Tien K. Nguyen, and Mark D. Hulett. 2021. "The Heparanase Regulatory Network in Health and Disease" International Journal of Molecular Sciences 22, no. 20: 11096. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms222011096

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop