heparitin-sulfate and Neoplasms

heparitin-sulfate has been researched along with Neoplasms* in 116 studies

Reviews

64 review(s) available for heparitin-sulfate and Neoplasms

ArticleYear
Discovery and development of small-molecule heparanase inhibitors.
    Bioorganic & medicinal chemistry, 2023, 07-15, Volume: 90

    Heparanase-1 (HPSE) is a promising yet challenging therapeutic target. It is the only known enzyme that is responsible for cleavage of heparan sulfate (HS) side chains from heparan sulfate proteoglycans (HSPGs), and is the key enzyme involved in the remodeling and degradation of the extracellular matrix (ECM). Overexpression of HPSE is found in various types of diseases, including cancers, inflammations, diabetes, and viral infections. Inhibiting HPSE can restore ECM functions and integrity, making the development of HPSE inhibitors a highly sought-after topic. So far, all HPSE inhibitors that have entered clinical trials belong to the category of HS mimetics, and no small-molecule or drug-like HPSE inhibitors have made similar progress. None of the HS mimetics have been approved as drugs, with some clinical trials discontinued due to poor bioavailability, side effects, and unfavorable pharmacokinetics characteristics. Small-molecule HPSE inhibitors are, therefore, particularly appealing due to their drug-like characteristics. Advances in the chemical spaces and drug design technologies, including the increasing use of in vitro and in silico screening methods, have provided new opportunities in drug discovery. This article aims to review the discovery and development of small-molecule HPSE inhibitors via screening strategies to shed light on the future endeavors in the development of novel HPSE inhibitors.

    Topics: Glucuronidase; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Neoplasms

2023
Heparanase Inhibitors in Cancer Progression: Recent Advances.
    Current pharmaceutical design, 2021, Volume: 27, Issue:1

    An endo-β-glucuronidase enzyme, Heparanase (HPSE), degrades the side chains of polymeric heparan sulfate (HS), a glycosaminoglycan formed by alternate repetitive units of D-glucosamine and D-glucuronic acid/L-iduronic acid. HS is a major component of the extracellular matrix and basement membranes and has been implicated in processes of the tissue's integrity and functional state. The degradation of HS by HPSE enzyme leads to conditions like inflammation, angiogenesis, and metastasis. An elevated HPSE expression with a poor prognosis and its multiple roles in tumor growth and metastasis has attracted significant interest for its inhibition as a potential anti-neoplastic target.. We reviewed the literature from journal publication websites and electronic databases such as Bentham, Science Direct, PubMed, Scopus, USFDA, etc., about HPSE, its structure, functions, and role in cancer.. The present review is focused on Heparanase inhibitors (HPIns) that have been isolated from natural resources or chemically synthesized as new therapeutics for metastatic tumors and chronic inflammatory diseases in recent years. The recent developments made in the HPSE structure and function are also discussed, which can lead to the future design of HPIns with more potency and specificity for the target.. HPIns can be a better target to be explored against various cancers.

    Topics: Extracellular Matrix; Glucuronidase; Heparitin Sulfate; Humans; Neoplasms

2021
Heparan Sulfate Glycosaminoglycans: (Un)Expected Allies in Cancer Clinical Management.
    Biomolecules, 2021, 01-21, Volume: 11, Issue:2

    In an era when cancer glycobiology research is exponentially growing, we are witnessing a progressive translation of the major scientific findings to the clinical practice with the overarching aim of improving cancer patients' management. Many mechanistic cell biology studies have demonstrated that heparan sulfate (HS) glycosaminoglycans are key molecules responsible for several molecular and biochemical processes, impacting extracellular matrix properties and cellular functions. HS can interact with a myriad of different ligands, and therefore, hold a pleiotropic role in regulating the activity of important cellular receptors and downstream signalling pathways. The aberrant expression of HS glycan chains in tumours determines main malignant features, such as cancer cell proliferation, angiogenesis, invasion and metastasis. In this review, we devote particular attention to HS biological activities, its expression profile and modulation in cancer. Moreover, we highlight HS clinical potential to improve both diagnosis and prognosis of cancer, either as HS-based biomarkers or as therapeutic targets.

    Topics: Animals; Biomarkers, Tumor; Cell Differentiation; Cell Membrane; Cell Proliferation; Extracellular Matrix; Gene Expression Profiling; Gene Expression Regulation, Neoplastic; Glycosaminoglycans; Heparitin Sulfate; Humans; Ligands; Mice; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Signal Transduction

2021
From Cancer to COVID-19: A Perspective on Targeting Heparan Sulfate-Protein Interactions.
    Chemical record (New York, N.Y.), 2021, Volume: 21, Issue:11

    Heparan sulfate (HS) is a complex, polyanionic polysaccharide ubiquitously expressed on cell surfaces and in the extracellular matrix. HS interacts with numerous proteins to mediate a vast array of biological and pathological processes. Inhibition of HS-protein interactions is thus an attractive approach for new therapeutic development for cancer and infectious diseases, including COVID-19; however, synthesis of well-defined native HS oligosaccharides remains challenging. This has aroused significant interest in the development of HS mimetics which are more synthetically tractable and have fewer side effects, such as undesired anticoagulant activity. This account provides a perspective on the design and synthesis of different classes of HS mimetics with useful properties, and the development of various assays and molecular modelling tools to progress our understanding of their interactions with HS-binding proteins.

    Topics: COVID-19; Heparitin Sulfate; Humans; Neoplasms; Proteins; SARS-CoV-2

2021
The role of heparan sulfate maturation in cancer: A focus on the 3O-sulfation and the enigmatic 3O-sulfotransferases (HS3STs).
    Seminars in cancer biology, 2020, Volume: 62

    Heparansulfate (HS) modifications are master regulators of the cross-talk between cell and matrix and modulate the biological activity of an array of HS binding proteins, including growth factors and chemokines, morphogens and immunity cell receptors. This review will highlight the importance of HS maturation mediated by N-deactetylase/sulfotransferases, 2O- and 6O-sulfotransferases in cancer biology, and will focus on the 3O-sulfotransferases and on the terminal, rare 3O-sulfation, and their important but still enigmatic impact in cancer progression. The review will also discuss the molecular mechanisms of action of these HS modifications with regards to ligand interactions and signaling in the cancer process and their clinical significance.

    Topics: Animals; Biosynthetic Pathways; Carrier Proteins; Cell Proliferation; Disease Susceptibility; Fibroblast Growth Factors; Heparitin Sulfate; Humans; Ligands; Neoplasms; Protein Binding; Sulfotransferases; Transforming Growth Factor beta

2020
The Challenge of Modulating Heparan Sulfate Turnover by Multitarget Heparin Derivatives.
    Molecules (Basel, Switzerland), 2020, Jan-17, Volume: 25, Issue:2

    This review comes as a part of the special issue "Emerging frontiers in GAGs and mimetics". Our interest is in the manipulation of heparan sulfate (HS) turnover by employing HS mimetics/heparin derivatives that exert pleiotropic effects and are interesting for interfering at multiple levels with pathways in which HS is implicated. Due to the important role of heparanase in HS post-biosynthetic modification and catabolism, we focus on the possibility to target heparanase, at both extracellular and intracellular levels, a strategy that can be applied to many conditions, from inflammation to cancer and neurodegeneration.

    Topics: Biomimetic Materials; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Neoplasms; Neurodegenerative Diseases

2020
Flipping the Molecular Switch: Influence of Perlecan and Its Modifiers in the Tumor Microenvironment.
    Advances in experimental medicine and biology, 2020, Volume: 1245

    The tumor microenvironment (TME) is rich in matrix components, growth factors, cytokines, and enzymatic modifiers that respond to changing conditions, to alter the fundamental properties of the tumor bed. Perlecan/HSPG2, a large, multi-domain heparan sulfate proteoglycan, is concentrated in the reactive stroma that surrounds tumors. Depending on its state in the TME, perlecan can either prevent or promote the progression of cancers to metastatic disease. Breast, prostate, lung, and renal cancers all preferentially metastasize to bone, a dense, perlecan-rich environment that is initially a "hostile" niche for cancer cells. Driven by inflammation, production of perlecan and its enzyme modifiers, which include matrix metalloproteinases (MMPs), sulfatases (SULFs), and heparanase (HPSE), increases in the reactive stroma surrounding growing and invading tumors. MMPs act upon the perlecan core protein, releasing bioactive fragments of the protein, primarily from C-terminal domains IV and V. These fragments influence cell adhesion, invasion, and angiogenesis. Sulfatases and heparanases act directly upon the heparan sulfate chains, releasing growth factors from reservoirs to reach receptors on the cancer cell surface. We propose that perlecan modifiers, by promoting the degradation of the perlecan-rich stroma, "flip the molecular switch" and convert the "hostile" stroma into a welcoming one that supports cancer dissemination and metastasis. Targeted therapies that prevent this molecular conversion of the TME should be considered as potential new therapeutics to limit metastasis.

    Topics: Extracellular Matrix Proteins; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Neoplasm Metastasis; Neoplasms; Tumor Microenvironment

2020
Heparan Sulfate in the Tumor Microenvironment.
    Advances in experimental medicine and biology, 2020, Volume: 1245

    The biology of tumor cells strictly depends on their microenvironment architecture and composition, which controls the availability of growth factors and signaling molecules. Thus, the network of glycosaminoglycans, proteoglycans, and proteins known as extracellular matrix (ECM) that surrounds the cells plays a central role in the regulation of tumor fate. Heparan sulfate (HS) and heparan sulfate proteoglycans (HSPGs) are highly versatile ECM components that bind and regulate the activity of growth factors, cell membrane receptors, and other ECM molecules. These HS binding partners modulate cell adhesion, motility, and proliferation that are processes altered during tumor progression. Modification in the expression and activity of HS, HSPGs, and the respective metabolic enzymes results unavoidably in alteration of tumor cell microenvironment. In this light, the targeting of HS structure and metabolism is potentially a new tool in the treatment of different cancer types.

    Topics: Extracellular Matrix; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Neoplasms; Tumor Microenvironment

2020
Heparanase: Historical Aspects and Future Perspectives.
    Advances in experimental medicine and biology, 2020, Volume: 1221

    Heparanase is an endo-β-glucuronidase that cleaves at a limited number of internal sites the glycosaminoglycan heparan sulfate (HS). Heparanase enzymatic activity was first reported in 1975 and by 1983 evidence was beginning to emerge that the enzyme was a facilitator of tumor metastasis by cleaving HS chains present in blood vessel basement membranes and, thereby, aiding the passage of tumor cells through blood vessel walls. Due to a range of technical difficulties, it took another 16 years before heparanase was cloned and characterized in 1999 and a further 14 years before the crystal structure of the enzyme was solved. Despite these substantial deficiencies, there was steady progress in our understanding of heparanase long before the enzyme was fully characterized. For example, it was found as early as 1984 that activated T cells upregulate heparanase expression, like metastatic tumor cells, and the enzyme aids the entry of T cells and other leukocytes into inflammatory sites. Furthermore, it was discovered in 1989 that heparanase releases pre-existing growth factors and cytokines associated with HS in the extracellular matrix (ECM), the liberated growth factors/cytokines enhancing angiogenesis and wound healing. There were also the first hints that heparanase may have functions other than enzymatic activity, in 1995 it being reported that under certain conditions the enzyme could act as a cell adhesion molecule. Also, in the same year PI-88 (Muparfostat), the first heparanase inhibitor to reach and successfully complete a Phase III clinical trial was patented.Nevertheless, the cloning of heparanase (also known as heparanase-1) in 1999 gave the field an enormous boost and some surprises. The biggest surprise was that there is only one heparanase encoding gene in the mammalian genome, despite earlier research, based on substrate specificity, suggesting that there are at least three different heparanases. This surprising conclusion has remained unchanged for the last 20 years. It also became evident that heparanase is a family 79 glycoside hydrolase that is initially produced as a pro-enzyme that needs to be processed by proteases to form an enzymatically active heterodimer. A related molecule, heparanase-2, was also discovered that is enzymatically inactive but, remarkably, recently has been shown to inhibit heparanase-1 activity as well as acting as a tumor suppressor that counteracts many of the pro-tumor properties of heparanase-1.The early claim tha

    Topics: Animals; Glucuronidase; Heparitin Sulfate; History, 20th Century; History, 21st Century; Humans; Neoplasms; Neovascularization, Pathologic

2020
Involvement of Syndecan-1 and Heparanase in Cancer and Inflammation.
    Advances in experimental medicine and biology, 2020, Volume: 1221

    The cell surface heparan sulfate proteoglycan Syndecan-1 acts as an important co-receptor for receptor tyrosine kinases and chemokine receptors, and as an adhesion receptor for structural glycoproteins of the extracellular matrix. It serves as a substrate for heparanase, an endo-β-glucuronidase that degrades specific domains of heparan sulfate carbohydrate chains and thereby alters the functional status of the proteoglycan and of Syndecan-1-bound ligands. Syndecan-1 and heparanase show multiple levels of functional interactions, resulting in mutual regulation of their expression, processing, and activity. These interactions are of particular relevance in the context of inflammation and malignant disease. Studies in animal models have revealed a mechanistic role of Syndecan-1 and heparanase in the regulation of contact allergies, kidney inflammation, multiple sclerosis, inflammatory bowel disease, and inflammation-associated tumorigenesis. Moreover, functional interactions between Syndecan-1 and heparanase modulate virtually all steps of tumor progression as defined in the Hallmarks of Cancer. Due to their prognostic value in cancer, and their mechanistic involvement in tumor progression, Syndecan-1 and heparanase have emerged as important drug targets. Data in preclinical models and preclinical phase I/II studies have already yielded promising results that provide a translational perspective.

    Topics: Animals; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Neoplasms; Syndecan-1

2020
Immunomodulatory Activities of the Heparan Sulfate Mimetic PG545.
    Advances in experimental medicine and biology, 2020, Volume: 1221

    Heparanase regulates multiple biological activities that enhance tumor growth and metastatic spread. Heparanase cleaves and degrades heparan sulfate (HS), a key structural component of the extracellular matrix that serves as a barrier to cell invasion and also as a reservoir for cytokines and growth factors critical for tumor growth and metastasis. For this reason, heparanase is an attractive target for the development of novel anti-cancer therapies. Pixatimod (PG545), a heparanase inhibitor, has shown promising results in the treatment of multiple tumor types. PG545 offers a diversity of mechanisms of action in tumor therapy that include angiogenic inhibition, inhibition of growth factor release, inhibition of tumor cell migration, tumor cell apoptosis, activation of ER stress response, dysregulation of autophagy, and NK cell activation. Further investigation into the role that heparanase and its inhibitors play in tumor therapy can lead to the development of effective tumor therapies.

    Topics: Glucuronidase; Heparitin Sulfate; Humans; Neoplasms; Saponins

2020
PI-88 and Related Heparan Sulfate Mimetics.
    Advances in experimental medicine and biology, 2020, Volume: 1221

    The heparan sulfate mimetic PI-88 (muparfostat) is a complex mixture of sulfated oligosaccharides that was identified in the late 1990s as a potent inhibitor of heparanase. In preclinical animal models it was shown to block angiogenesis, metastasis and tumor growth, and subsequently became the first heparanase inhibitor to enter clinical trials for cancer. It progressed to Phase III trials but ultimately was not approved for use. Herein we summarize the preparation, physicochemical and biological properties of PI-88, and discuss preclinical/clinical and structure-activity relationship studies. In addition, we discuss the PI-88-inspired development of related HS mimetic heparanase inhibitors with improved properties, ultimately leading to the discovery of PG545 (pixatimod) which is currently in clinical trials.

    Topics: Animals; Antineoplastic Agents; Glucuronidase; Heparitin Sulfate; Humans; Neoplasms; Neovascularization, Pathologic; Oligosaccharides; Structure-Activity Relationship

2020
Non-Anticoagulant Heparins as Heparanase Inhibitors.
    Advances in experimental medicine and biology, 2020, Volume: 1221

    The chapter will review early and more recent seminal contributions to the discovery and characterization of heparanase and non-anticoagulant heparins inhibiting its peculiar enzymatic activity. Indeed, heparanase displays a unique versatility in degrading heparan sulfate chains of several proteoglycans expressed in all mammalian cells. This endo-β-D-glucuronidase is overexpressed in cancer, inflammation, diabetes, atherosclerosis, nephropathies and other pathologies. Starting from known low- or non-anticoagulant heparins, the search for heparanase inhibitors evolved focusing on structure-activity relationship studies and taking advantage of new chemical-physical analytical methods which have allowed characterization and sequencing of polysaccharide chains. New methods to screen heparanase inhibitors and to evaluate their mechanism of action and in vivo activity in experimental models prompted their development. New non-anticoagulant heparin derivatives endowed with anti-heparanase activity are reported. Some leads are under clinical evaluation in the oncology field (e.g., acute myeloid leukemia, multiple myeloma, pancreatic carcinoma) and in other pathological conditions (e.g., sickle cell disease, malaria, labor arrest).

    Topics: Animals; Glucuronidase; Heparin; Heparitin Sulfate; Humans; Neoplasms

2020
The Good and Bad Sides of Heparanase-1 and Heparanase-2.
    Advances in experimental medicine and biology, 2020, Volume: 1221

    In this chapter, we will emphasize the importance of heparan sulfate proteoglycans (HSPG) in controlling various physiological and pathological molecular mechanisms and discuss how the heparanase enzyme can modulate the effects triggered by HSPG. Additionally, we will also navigate about the existing knowledge of the possible role of heparanase-2 in biological events. Heparan sulfate is widely distributed and evolutionarily conserved, evidencing its vital importance in cell development and functions such as cell proliferation, migration, adhesion, differentiation, and angiogenesis. During remodeling of the extracellular matrix, the breakdown of heparan sulfate by heparanase results in the release of molecules containing anchored glycosaminoglycan chains of great interest in heparanase-mediated cell signaling pathways in various physiological states, tumor development, inflammation, and other diseases. Taken together, it appears that heparanase plays a key role in the maintenance of the pathology of cancer and inflammatory diseases and is a potential target for anti-cancer therapies. Therefore, heparanase inhibitors are currently being examined in clinical trials as novel cancer therapeutics. Heparanase-2 has no enzymatic activity, displays higher affinity for heparan sulfate and the coding region alignment shows 40% identity with the heparanase gene. Heparanase-2 plays an important role in embryogenic development however its mode of action and biological function remain to be elucidated. Heparanase-2 functions as an inhibitor of the heparanase-1 enzyme and also inhibits neovascularization mediated by VEGF. The HPSE2 gene is repressed by the Polycomb complex, together suggesting a role as a tumor suppressor.

    Topics: Glucuronidase; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Neoplasms

2020
Heparanase, cell signaling, and viral infections.
    Cellular and molecular life sciences : CMLS, 2020, Volume: 77, Issue:24

    Heparanase (HPSE) is a multifunctional protein endowed with many non-enzymatic functions and a unique enzymatic activity as an endo-β-D-glucuronidase. The latter allows it to serve as a key modulator of extracellular matrix (ECM) via a well-regulated cleavage of heparan sulfate side chains of proteoglycans at cell surfaces. The cleavage and associated changes at the ECM cause release of multiple signaling molecules with important cellular and pathological functions. New and emerging data suggest that both enzymatic as well as non-enzymatic functions of HPSE are important for health and illnesses including viral infections and virally induced cancers. This review summarizes recent findings on the roles of HPSE in activation, inhibition, or bioavailability of key signaling molecules such as AKT, VEGF, MAPK-ERK, and EGFR, which are known regulators of common viral infections in immune and non-immune cell types. Altogether, our review provides a unique overview of HPSE in cell-survival signaling pathways and how they relate to viral infections.

    Topics: Extracellular Matrix; Glucuronidase; Heparitin Sulfate; Humans; Immunity, Cellular; Neoplasms; Signal Transduction; Virus Diseases

2020
Bringing Heparan Sulfate Glycomics Together with Proteomics for the Design of Novel Therapeutics: A Historical Perspective.
    Proteomics, 2019, Volume: 19, Issue:21-22

    Increasing knowledge of how peptides bind saccharides, and of how saccharides bind peptides, is starting to revolutionize understanding of cell-extracellular matrix relationships. Here, a historical perspective is taken of the relationship between heparan sulfate glycosaminoglycans and how they interact with peptide growth factors in order to both drive and modulate signaling through the appropriate cognate receptors. Such knowledge is guiding the preparation of targeted sugar mimetics that will impact the treatment of many different kinds of diseases, including cancer.

    Topics: Extracellular Matrix; Glycomics; Glycosaminoglycans; Heparitin Sulfate; Humans; Neoplasms; Peptides; Protein Binding; Proteomics; Signal Transduction

2019
Dually modified transmembrane proteoglycans in development and disease.
    Cytokine & growth factor reviews, 2018, Volume: 39

    Aberrant cell signaling in response to secreted growth factors has been linked to the development of multiple diseases, including cancer. As such, understanding mechanisms that control growth factor availability and receptor-growth factor interaction is vital. Dually modified transmembrane proteoglycans (DMTPs), which are classified as cell surface macromolecules composed of a core protein decorated with covalently linked heparan sulfated (HS) and/or chondroitin sulfated (CS) glycosaminoglycan (GAG) chains, provide one type of regulatory mechanism. Specifically, DMTPs betaglycan and syndecan-1 (SDC1) play crucial roles in modulating key cell signaling pathways, such as Wnt, transforming growth factor-β and fibroblast growth factor signaling, to affect epithelial cell biology and cancer progression. This review outlines current and potential functions for betaglycan and SDC1, with an emphasis on comparing individual roles for HS and CS modified DMTPs. We highlight the mutual dependence of DMTPs' GAG chains and core proteins and provide comprehensive knowledge on how these DMTPs, through regulation of ligand availability and receptor internalization, control cell signaling pathways involved in development and disease.

    Topics: Animals; Chondroitin Sulfates; Epithelial Cells; Glycosaminoglycans; Heparitin Sulfate; Humans; Intercellular Signaling Peptides and Proteins; Mice; Neoplasms; Proteoglycans; Receptors, Transforming Growth Factor beta; Signal Transduction; Syndecan-1; Transforming Growth Factor beta; Wnt Proteins

2018
Heparan Sulfate Mimetics in Cancer Therapy: The Challenge to Define Structural Determinants and the Relevance of Targets for Optimal Activity.
    Molecules (Basel, Switzerland), 2018, Nov-08, Volume: 23, Issue:11

    Beyond anticoagulation, the therapeutic potential of heparin derivatives and heparan sulfate (HS) mimetics (functionally defined HS mimetics) in oncology is related to their ability to bind and modulate the function of a vast array of HS-binding proteins with pivotal roles in cancer growth and progression. The definition of structural/functional determinants and the introduction of chemical modifications enabled heparin derivatives to be identified with greatly reduced or absent anticoagulant activity, but conserved/enhanced anticancer activity. These studies paved the way for the disclosure of structural requirements for the inhibitory effects of HS mimetics on heparanase, selectins, and growth factor receptor signaling, as well as for the limitation of side effects. Actually, HS mimetics affect the tumor biological behavior via a multi-target mechanism of action based on their effects on tumor cells and various components of the tumor microenvironment. Emerging evidence indicates that immunomodulation can participate in the antitumor activity of these agents. Significant ability to enhance the antitumor effects of combination treatments with standard therapies was shown in several tumor models. While the first HS mimetics are undergoing early clinical evaluation, an improved understanding of the molecular contexts favoring the antitumor action in certain malignancies or subgroups is needed to fully exploit their potential.

    Topics: Animals; Biomimetic Materials; Cell Proliferation; Disease Progression; Glucuronidase; Heparitin Sulfate; Humans; Neoplasms; Receptors, Growth Factor; Selectins; Signal Transduction; Structure-Activity Relationship

2018
New Advances of Heparanase and Heparanase-2 in Human Diseases.
    Archives of medical research, 2018, Volume: 49, Issue:7

    As we all know, heparanase plays an important role in human diseases. As a kind of endo-β-glucuronidase, heparanase is the known only enzyme in mammals which could degrade heparan sulfate(HS) specifically. HS is a vital component of extracellular matrix(ECM). Heparanase takes effect by cleaving theβ(1,4)-glycosidic between glucosamine residue and glucuronic acid of HS. This cleavage will cause ECM remodelling and HS-linked biological molecules release, including cytokines, growth factors and a lot of biological molecules regulating various pathological activities. Experiments already proved that heparanase gene over-expresses in cancers of gastrointestinal tract, esophagus, breast and so on. Various studies have demonstrated the heparanase's pro-metastatic function and the reduced survival rate of patients could be indicated by high heparanase levels. Besides, pathological processes including procoagulant activities, preeclamptic placentas and inflammation are all verified to be associated with heparanase activity. In recent years, many functions other than pro-tumor effect was found in heparanase and worldwide researchers conducted varieties of experiments to identify the new function of this significant enzyme. Also, these newly-found functions are closely connected to certain cellular activities, for example epithelial to mesenchymal transition (EMT). It has already been demonstrated that EMT is related to some clinical disorders, like renal diseases. Given that heparanase is the only enzyme capable of this function, it could be concluded that heparanase would be a potential and valuable therapy target. This mini-review aims to retrospect literatures about heparanase published in 2017 and 2018 and provide a direction for therapy methods targeting heparanase.

    Topics: Animals; Cytokines; Epithelial-Mesenchymal Transition; Extracellular Matrix; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Kidney Diseases; Neoplasms

2018
Targeting heparin and heparan sulfate protein interactions.
    Organic & biomolecular chemistry, 2017, Jul-21, Volume: 15, Issue:27

    Heparin and heparan sulfate glycosaminoglycans are long, linear polysaccharides that are made up of alternating dissacharide sequences of sulfated uronic acid and amino sugars. Unlike heparin, which is only found in mast cells, heparan sulfate is ubiquitously expressed on the cell surface and in the extracellular matrix of all animal cells. These negatively-charged glycans play essential roles in important cellular functions such as cell growth, adhesion, angiogenesis, and blood coagulation. These biomolecules are also involved in pathophysiological conditions such as pathogen infection and human disease. This review discusses past and current methods for targeting these complex biomolecules as a novel therapeutic strategy to treating disorders such as cancer, neurodegenerative diseases, and infection.

    Topics: Animals; Glycosaminoglycans; Heparin; Heparitin Sulfate; Humans; Infections; Neoplasms; Neurodegenerative Diseases; Small Molecule Libraries

2017
Heparanase regulation of cancer, autophagy and inflammation: new mechanisms and targets for therapy.
    The FEBS journal, 2017, Volume: 284, Issue:1

    Because of its impact on multiple biological pathways, heparanase has emerged as a major regulator of cancer, inflammation and other disease processes. Heparanase accomplishes this by degrading heparan sulfate which regulates the abundance and location of heparin-binding growth factors thereby influencing multiple signaling pathways that control gene expression, syndecan shedding and cell behavior. In addition, heparanase can act via nonenzymatic mechanisms that directly activate signaling at the cell surface. Clinical trials testing heparanase inhibitors as anticancer therapeutics are showing early signs of efficacy in patients further emphasizing the biological importance of this enzyme. This review focuses on recent developments in the field of heparanase regulation of cancer and inflammation, including the impact of heparanase on exosomes and autophagy, and novel mechanisms whereby heparanase regulates tumor metastasis, angiogenesis and chemoresistance. In addition, the ongoing development of heparanase inhibitors and their potential for treating cancer and inflammation are discussed.

    Topics: Antineoplastic Agents; Autophagy; Clinical Trials as Topic; Drug Resistance, Neoplasm; Enzyme Inhibitors; Exosomes; Gene Expression Regulation, Neoplastic; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Molecular Targeted Therapy; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Signal Transduction; Syndecans

2017
The Functions of Heparanase in Human Diseases.
    Mini reviews in medicinal chemistry, 2017, Volume: 17, Issue:6

    The study of the heparanase has long been paid wide attention. Heparanase, an endo-β-D-glucuronidase, is capable of specifically degrading heparan sulfate (HS), one of the excellular matrix (ECM) components. It exerts its enzymatic activity catalyzing the cleavage of the β (1,4)-glycosidic bond between glucuronic acid and glucosamine residue. HS cleavage results in remodelling of the extracellular matrix as well as in regulating the release of many HS-linked molecules such as growth factors, cytokines and enzymes involved in inflammation, wound healing and tumour invasion. Varieties of experiments indicated that heparanase mRNA is overexpressed in human tumors, including breast cancer, gastrointestinal tumors, and esophageal carcinomas. A pro-metastatic and pro-angiogenic role for heparanase has been widely verified and high levels of heparanase correlate with reduced survival of cancer patients. Except protumor function, heparanase also plays a role in inflammation, angiogenesis, placentas and procoagulant activities. Heparanase is found to have many other functions in recent years, since many experiments have been carried out to identify this significant enzyme's new features. These newly found functions are related to the cellular activities such as autophagy and epithelial to mesenchymal transition (EMT). And together with other heparanase functions, autophagy and EMT are verified to be involved in several clinical disorders, for example, renal diseases. Considering that, once inactivated, there are no other enzymes capable of performing the same function, it is apparent that heparanase can be an effective and promising therapy target. This short review aims to establish the currently known function of this enzyme and provide evidence for heparanase targeted therapy.

    Topics: Autophagy; Epithelial-Mesenchymal Transition; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Kidney Diseases; Neoplasms; Prion Diseases; Scleroderma, Systemic

2017
Mechanisms of heparanase inhibitors in cancer therapy.
    Experimental hematology, 2016, Volume: 44, Issue:11

    Heparanase is an endo-β-D-glucuronidase capable of cleaving heparan sulfate side chains contributing to breakdown of the extracellular matrix. Increased expression of heparanase has been observed in numerous malignancies and is associated with a poor prognosis. It has generated significant interest as a potential antineoplastic target because of the multiple roles it plays in tumor growth and metastasis. The protumorigenic effects of heparanase are enhanced by the release of heparan sulfate side chains, with subsequent increase in bioactive fragments and cytokine levels that promote tumor invasion, angiogenesis, and metastasis. Preclinical experiments have found heparanase inhibitors to substantially reduce tumor growth and metastasis, leading to clinical trials with heparan sulfate mimetics. In this review, we examine the role of heparanase in tumor biology and its interaction with heparan surface proteoglycans, specifically syndecan-1, as well as the mechanism of action for heparanase inhibitors developed as antineoplastic therapeutics.

    Topics: Animals; Antineoplastic Agents; Cancer Vaccines; Clinical Trials as Topic; Drug Evaluation, Preclinical; Enzyme Inhibitors; Extracellular Matrix; Gene Expression; Glucuronidase; Heparitin Sulfate; Humans; Molecular Targeted Therapy; Neoplasms; Prognosis; Syndecan-1; Treatment Outcome

2016
Recent data concerning heparanase: focus on fibrosis, inflammation and cancer.
    Biomolecular concepts, 2015, Volume: 6, Issue:5-6

    Heparanase (HPSE) is a multitasking protein characterized by enzymatic and non-enzymatic activities. By means of its enzymatic activity, HPSE catalyzes the cutting of the side chains of heparan sulfate (HS) proteoglycans, thereby inducing the remodeling of the extracellular matrix and basement membranes. Thanks to the cleavage of HS, HPSE also promotes the release and diffusion of several HS-linked molecules such as growth factors, cytokines and enzymes. In addition to degrading HS chains, HPSE has non-enzymatic functions that trigger several signaling pathways. This signaling activity is achieved by interacting with transmembrane proteins, activating kinases such as Akt and Src, or modulating the activity of factors such as FGF-2 and TGF-β. Several studies have recently highlighted a possible intracellular activity for HPSE, particularly at nuclear level. While HPSE activity is quite limited in physiological conditions, its demonstrated increasing involvement in various pathological conditions, such as in tumor progression and renal disease, have attracted the attention of a growing number of researchers. The fact that no other molecule is capable of performing the same function as HPSE makes this enzyme an attractive potential target of medical treatment. With this short conceptual overview, we aim to provide an update on current knowledge concerning the HPSE protein in the experimental and clinical settings, paying particular attention to its role in fibrosis, inflammation and cancer.

    Topics: Epithelial-Mesenchymal Transition; Fibrosis; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Kidney; Models, Biological; Neoplasms; Signal Transduction

2015
Heparan sulfate signaling in cancer.
    Trends in biochemical sciences, 2014, Volume: 39, Issue:6

    Heparan sulfate (HS) is a biopolymer consisting of variably sulfated repeating disaccharide units. The anticoagulant heparin is a highly sulfated intracellular variant of HS. HS has demonstrated roles in embryonic development, homeostasis, and human disease via non-covalent interactions with numerous cellular proteins, including growth factors and their receptors. HS can function as a co-receptor by enhancing receptor-complex formation. In other contexts, HS disrupts signaling complexes or serves as a ligand sink. The effects of HS on growth factor signaling are tightly regulated by the actions of sulfyltransferases, sulfatases, and heparanases. HS has important emerging roles in oncogenesis, and heparin derivatives represent potential therapeutic strategies for human cancers. Here we review recent insights into HS signaling in tumor proliferation, angiogenesis, metastasis, and differentiation. A cancer-specific understanding of HS signaling could uncover potential therapeutic targets in this highly actionable signaling network.

    Topics: Animals; Heparitin Sulfate; Humans; Neoplasms; Signal Transduction

2014
Heparanase multiple effects in cancer.
    Thrombosis research, 2014, Volume: 133 Suppl 2

    Heparanase is an endo-β-D-glucuronidase that is capable of cleaving heparan sulfate side chains of heparan sulfate proteoglycans on cell surfaces and the extracellular matrix, activity that is strongly implicated in tumor metastasis and angiogenesis. Apart of its well characterized enzymatic activity, heparanase was noted to exert also enzymatic-independent functions. Among these are the up-regulation of vascular endothelial growth factor (VEGF)-A, VEGF-C and activation of intra-cellular signaling involved in cell survival and proliferation. We had earlier demonstrated that heparanase may also affect the hemostatic system in a non-enzymatic manner. We had shown that heparanase up-regulated the expression of the blood coagulation initiator- tissue factor (TF) and interacted with the tissue factor pathway inhibitor (TFPI) on the cell surface membrane of endothelial and tumor cells, leading to dissociation of TFPI and resulting in increased cell surface coagulation activity. Moreover, we have demonstrated that heparanase directly enhanced TF activity which led to increased factor Xa production and subsequent activation of the coagulation system. Taking into account the prometastatic, pro-angiogenic and pro-coagulant functions of heparanase, over-expression in human malignancies and abundance in platelets, implies that heparanase is potentially a good target for cancer therapy.

    Topics: Blood Coagulation; Glucuronidase; Heparitin Sulfate; Humans; Neoplasms; Signal Transduction; Tumor Microenvironment

2014
Glycosaminoglycans in cancer treatment.
    Thrombosis research, 2014, Volume: 133 Suppl 2

    Studies aimed at the identification of biomarkers and treatment targets of cancer have focused on mRNAs, miRNAs, and proteins expressed by malignant cells, while glycoproteins mainly produced by stromal cells remain relatively unexplored. Glycans lack a given template for their biosynthesis that involves the concerted action of several, sometimes >15 different enzymes. This fact complicates the analysis at the genomic level of the role of glycoproteins in clinical oncology. The glycosaminoglycans (GAGs) stand out as highly polyanionic components at the surface of malignant and stromal tumor cells as well as their surrounding matrix. Published data thus describe a multifaceted regulatory role of GAGs and GAG-conjugated proteins, proteoglycans, in e.g. tumor associated angiogenesis, coagulation, invasion, and metastasis. Relatively small, randomized clinical trials suggest that heparin, an over-sulfated variant of the GAG heparan sulfate, may have direct, anti-tumor effects. Several ongoing trials aim at establishing whether heparin and its derivatives should be added to standard treatment of cancer patients or not, based on progression free- and overall survival end-point data. Given the potential bleeding complications with this treatment, other strategies to block GAG function should provide interesting alternatives. In the emerging era of personalized medicine, one can foresee the development of predictive biomarkers to select patients that may benefit from GAG-targeted treatments, aiming at individualized prevention of thromboembolic complications as well as inhibition of tumor development and progression. Here, the role of GAGs as targets and vehicles of cancer treatment is discussed with special emphasis on angiogenesis and coagulation associated mechanisms.

    Topics: Biomarkers; Blood Coagulation; Disease Progression; Glycosaminoglycans; Heparitin Sulfate; Humans; Neoplasms

2014
Involvement of heparanase in atherosclerosis and other vessel wall pathologies.
    Matrix biology : journal of the International Society for Matrix Biology, 2013, Jun-24, Volume: 32, Issue:5

    Heparanase, the sole mammalian endoglycosidase degrading heparan sulfate, is causally involved in cancer metastasis, angiogenesis, inflammation and kidney dysfunction. Despite the wide occurrence and impact of heparan sulfate proteoglycans in vascular biology, the significance of heparanase in vessel wall disorders is underestimated. Blood vessels are highly active structures whose morphology rapidly adapts to maintain vascular function under altered systemic and local conditions. In some pathologies (restenosis, thrombosis, atherosclerosis) this normally beneficial adaptation may be detrimental to overall function. Enzymatic dependent and independent effects of heparanase on arterial structure mechanics and repair closely regulate arterial compliance and neointimal proliferation following endovascular stenting. Additionally, heparanase promotes thrombosis after vascular injury and contributes to a pro-coagulant state in human carotid atherosclerosis. Importantly, heparanase is closely associated with development and progression of atherosclerotic plaques, including stable to unstable plaque transition. Consequently, heparanase levels are markedly increased in the plasma of patients with acute myocardial infarction. Noteworthy, heparanase activates macrophages, resulting in marked induction of cytokine expression associated with plaque progression towards vulnerability. Together, heparanase emerges as a regulator of vulnerable lesion development and potential target for therapeutic intervention in atherosclerosis and related vessel wall complications.

    Topics: Animals; Atherosclerosis; Carotid Arteries; Extracellular Matrix; Glucuronidase; Heparitin Sulfate; Humans; Macrophage Activation; Macrophages; Mice; Neoplasms; Plaque, Atherosclerotic; Thrombosis

2013
Versatile role of heparanase in inflammation.
    Matrix biology : journal of the International Society for Matrix Biology, 2013, Jun-24, Volume: 32, Issue:5

    Heparanase is the only known mammalian endoglycosidase capable of degrading heparan sulfate glycosaminoglycan, both in extracellular space and within the cells. It is tightly implicated in cancer progression and over the past few decades significant progress has been made in elucidating the multiple functions of heparanase in malignant tumor development, neovascularization and aggressive behavior. Notably, current data show that in addition to its well characterized role in cancer, heparanase activity may represent an important determinant in the pathogenesis of several inflammatory disorders, such as inflammatory lung injury, rheumatoid arthritis and chronic colitis. Nevertheless, the precise mode of heparanase action in inflammatory reactions remains largely unclear and recent observations suggest that heparanase can either facilitate or limit inflammatory responses, when tissue/cell-specific contextual cues may dictate an outcome of heparanase action in inflammation. In this review the involvement of heparanase in modulation of inflammatory reactions is discussed through a few illustrative examples, including neuroinflammation, sepsis-associated lung injury and inflammatory bowel disease. We also discuss possible action of the enzyme in coupling inflammation and tumorigenesis in the setting of inflammation-triggered cancer.

    Topics: Carcinogenesis; Extracellular Matrix; Gene Expression Regulation; Glucuronidase; Heparitin Sulfate; Humans; Inflammatory Bowel Diseases; Macrophages; Neoplasms; Pneumonia; Signal Transduction

2013
The heparanase/syndecan-1 axis in cancer: mechanisms and therapies.
    The FEBS journal, 2013, Volume: 280, Issue:10

    Heparanase is an endoglucuronidase that cleaves heparan sulfate chains of proteoglycans. In many malignancies, high heparanase expression and activity correlate with an aggressive tumour phenotype. A major consequence of heparanase action in cancer is a robust up-regulation of growth factor expression and increased shedding of syndecan-1 (a transmembrane heparan sulfate proteoglycan). Substantial evidence indicates that heparanase and syndecan-1 work together to drive growth factor signalling and regulate cell behaviours that enhance tumour growth, dissemination, angiogenesis and osteolysis. Preclinical and clinical studies have demonstrated that therapies targeting the heparanase/syndecan-1 axis hold promise for blocking the aggressive behaviour of cancer.

    Topics: Animals; Antineoplastic Agents; Biomimetic Materials; Cell Nucleus; Enzyme Activation; Enzyme Inhibitors; Exosomes; Gene Expression Regulation, Neoplastic; Glucuronidase; Heparitin Sulfate; Hepatocyte Growth Factor; Humans; Neoplasms; Signal Transduction; Syndecan-1

2013
[Glycan ligand specificity of killer lectin receptors].
    Yakugaku zasshi : Journal of the Pharmaceutical Society of Japan, 2012, Volume: 132, Issue:6

    Sialyl Lewis X (sLeX) antigen, Neu5Acα2,3Galβ1,4(Fucα1,3)GlcNAc-R, is expressed on the glycoproteins in sera or the surface of the cells and the expression of sLeX is enhanced in various conditions such as the inflammation and cancer. SLeX in the serum is utilized as a tumor marker. To clarify the roles of sLeX on secreted glycoproteins in vivo, we investigate the regulation of natural killer (NK) cell-dependent cytotoxicity through sLeX. NK cells express many receptors to kill the target cells such as cancerous cells and non-self, and their protein ligands have been elucidated. Of the killer lectin-like receptors (KLRs) on NK cells, several have been reported to recognize glycans. Using recombinant extracellular domains of KLRs (rKLRs: rNKG2A, C, D and rCD94), we evaluated their glycan ligand specificity and binding affinities using EIA methods. We clarified that all of these rKLRs can bind to high sLeX-expressing glycoprotein and heparin, heparan sulfate and highly sulfated polysaccharides and that glycan binding sites on NKG2D are mostly overlapped with those of protein ligands. In this review, we show the recent findings concerning the glycan ligands of these KLRs.

    Topics: Animals; Biomarkers; Cytotoxicity, Immunologic; Glycoproteins; Heparin; Heparitin Sulfate; Humans; Inflammation; Killer Cells, Natural; Lewis X Antigen; Ligands; Mice; Neoplasms; Polysaccharides; Protein Binding; Receptors, NK Cell Lectin-Like; Sialyl Lewis X Antigen

2012
Heparin-derived heparan sulfate mimics to modulate heparan sulfate-protein interaction in inflammation and cancer.
    Matrix biology : journal of the International Society for Matrix Biology, 2010, Volume: 29, Issue:6

    The heparan sulfate (HS) chains of heparan sulfate proteoglycans (HSPG) are "ubiquitous" components of the cell surface and the extracellular matrix (EC) and play important roles in the physiopathology of developmental and homeostatic processes. Most biological properties of HS are mediated by interactions with "heparin-binding proteins" and can be modulated by exogenous heparin species (unmodified heparin, low molecular weight heparins, shorter heparin oligosaccharides and various non-anticoagulant derivatives of different sizes). Heparin species can promote or inhibit HS activities to different extents depending, among other factors, on how closely their structure mimics the biologically active HS sequences. Heparin shares structural similarities with HS, but is richer in "fully sulfated" sequences (S domains) that are usually the strongest binders to heparin/HS-binding proteins. On the other hand, HS is usually richer in less sulfated, N-acetylated sequences (NA domains). Some of the functions of HS chains, such as that of activating proteins by favoring their dimerization, often require short S sequences separated by rather long NA sequences. The biological activities of these species cannot be simulated by heparin, unless this polysaccharide is appropriately chemically/enzymatically modified or biotechnologically engineered. This mini review covers some information and concepts concerning the interactions of HS chains with heparin-binding proteins and some of the approaches for modulating HS interactions relevant to inflammation and cancer. This is approached through a few illustrative examples, including the interaction of HS and heparin-derived species with the chemokine IL-8, the growth factors FGF1 and FGF2, and the modulation of the activity of the enzyme heparanase by these species. Progresses in sequencing HS chains and reproducing them either by chemical synthesis or semi-synthesis, and in the elucidation of the 3D structure of oligosaccharide-protein complexes, are paving the way for rational approaches to the development of HS-inspired drugs in the field of inflammation and cancer, as well in other therapeutic fields.

    Topics: Animals; Anticoagulants; Antimicrobial Cationic Peptides; Blood Proteins; Carrier Proteins; Extracellular Matrix; Fibroblast Growth Factor 1; Fibroblast Growth Factor 2; Glucuronidase; Heparan Sulfate Proteoglycans; Heparin; Heparitin Sulfate; Humans; Inflammation; Interleukin-8; Models, Molecular; Neoplasms; Oligosaccharides; Polysaccharides; Proteins

2010
Proteoglycans in health and disease: new concepts for heparanase function in tumor progression and metastasis.
    The FEBS journal, 2010, Volume: 277, Issue:19

    Heparanase is an endo-β-D-glucuronidase capable of cleaving heparan sulfate side chains at a limited number of sites, yielding heparan sulfate fragments of still appreciable size. Importantly, heparanase activity correlates with the metastatic potential of tumor-derived cells, attributed to enhanced cell dissemination as a consequence of heparan sulfate cleavage and remodeling of the extracellular matrix and basement membrane underlying epithelial and endothelial cells. Similarly, heparanase activity is implicated in neovascularization, inflammation and autoimmunity, involving the migration of vascular endothelial cells and activated cells of the immune system. The cloning of a single human heparanase cDNA 10 years ago enabled researchers to critically approve the notion that heparan sulfate cleavage by heparanase is required for structural remodeling of the extracellular matrix, thereby facilitating cell invasion. Progress in the field has expanded the scope of heparanase function and its significance in tumor progression and other pathologies. Notably, although heparanase inhibitors attenuated tumor progression and metastasis in several experimental systems, other studies revealed that heparanase also functions in an enzymatic activity-independent manner. Thus, inactive heparanase was noted to facilitate adhesion and migration of primary endothelial cells and to promote phosphorylation of signaling molecules such as Akt and Src, facilitating gene transcription (i.e. vascular endothelial growth factor) and phosphorylation of selected Src substrates (i.e. endothelial growth factor receptor). The concept of enzymatic activity-independent function of heparanase gained substantial support by the recent identification of the heparanase C-terminus domain as the molecular determinant behind its signaling capacity. Identification and characterization of a human heparanase splice variant (T5) devoid of enzymatic activity and endowed with protumorigenic characteristics, elucidation of cross-talk between heparanase and other extracellular matrix-degrading enzymes, and identification of single nucleotide polymorphism associated with heparanase expression and increased risk of graft versus host disease add other layers of complexity to heparanase function in health and disease.

    Topics: Antineoplastic Agents; Disease Progression; Enzyme Inhibitors; ErbB Receptors; Glucuronidase; Head and Neck Neoplasms; Heparitin Sulfate; Humans; Multiple Myeloma; Neoplasm Metastasis; Neoplasms; Proteoglycans; Signal Transduction

2010
Heparanase: busy at the cell surface.
    Trends in biochemical sciences, 2009, Volume: 34, Issue:10

    Heparanase activity is strongly implicated in structural remodeling of the extracellular matrix, a process which can lead to invasion by tumor cells. In addition, heparanase augments signaling cascades leading to enhanced phosphorylation of selected protein kinases and increased gene transcription associated with aggressive tumor progression. This function is apparently independent of heparan sulfate and enzyme activity, and is mediated by a novel protein domain localized at the heparanase C-terminus. Moreover, the functional repertoire of heparanase is expanded by its regulation of syndecan clustering, shedding, and mitogen binding. Recent reports indicate that modified glycol-split heparin, which inhibits heparanase activity, can profoundly inhibit the progression of tumor xenografts produced by myeloma and carcinoma cells, thus moving anti-heparanase therapy closer to reality.

    Topics: Animals; Cell Adhesion; Endocytosis; Enzyme Activation; Extracellular Matrix; Glucuronidase; Heparin; Heparitin Sulfate; Humans; Multiple Myeloma; Neoplasms; rac GTP-Binding Proteins; Receptors, Cell Surface; Signal Transduction; src-Family Kinases; Structure-Activity Relationship; Substrate Specificity; Syndecan-1

2009
[Mechanisms of action of the growth factor midkine].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 2008, Volume: 53, Issue:12 Suppl

    Topics: Amino Acid Sequence; Animals; Chondroitin Sulfates; Cytokines; Heparitin Sulfate; Humans; Inflammation; Midkine; Molecular Sequence Data; Neoplasms; Nerve Growth Factor; Protein Binding; Protein Conformation; Protein Structure, Tertiary

2008
Heparin, heparan sulfate and heparanase in cancer: remedy for metastasis?
    Anti-cancer agents in medicinal chemistry, 2008, Volume: 8, Issue:1

    Malignant tumor cells invade normal tissues in the vicinity of cancer through devastating the extracelluar matrix and blood vessel wall of the tissues. An important step in this process is degradation of heparan sulfate proteoglycan, a carbohydrate-protein complex. Heparan sulfate proteoglycan is a major component of the extracellular matrix, and is essential for the self-assembly, insolubility and barrier properties of basement membranes. Heparanase is an endoglucuronidase that cleaves heparan sulfate and expression level of this enzyme correlates with metastatic potential of tumor cells. Treatment with heparanase inhibitors markedly reduces the incidence of metastasis in experimental animals. Heparin, a widely used anticoagulant, is structurally related to heparan sulfate and a natural substrate of heparanase. Long-term treatment of cancer patients having venous thromboembolism with low molecular weight heparin showed improved survival rate. Understanding the functional roles and the corresponding molecular mechanisms of heparin, heparan sulfate and heparanase in cancer development may pave the way for exploring remedies against tumor metastasis.

    Topics: Animals; Anticoagulants; Extracellular Matrix; Heparin; Heparin Lyase; Heparin, Low-Molecular-Weight; Heparitin Sulfate; Humans; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Substrate Specificity; Thrombosis

2008
Heparanase involvement in physiology and disease.
    Cellular and molecular life sciences : CMLS, 2008, Volume: 65, Issue:11

    Heparanase is an endoglycosidase that degrades heparan sulfate on the cell surface and extracellular matrix. The physiological functions of heparanase include heparan sulfate turnover, embryo development, hair growth, and wound healing. Heparanase is implicated in a variety of pathologies, such as tumor growth, angiogenesis, metastasis, inflammation, and glomerular diseases. Heparanase overexpression in a variety of malignant tumors suggests that it could be a target for anti-cancer therapy.

    Topics: Alternative Splicing; Animals; Extracellular Matrix; Gene Expression Regulation, Enzymologic; Glucuronidase; Heparan Sulfate Proteoglycans; Heparin; Heparitin Sulfate; Humans; Neoplasms; Protein Precursors

2008
Heparanase: a target for drug discovery in cancer and inflammation.
    British journal of pharmacology, 2007, Volume: 151, Issue:1

    The remodelling of the extracellular matrix (ECM) has been shown to be highly upregulated in cancer and inflammation and is critically linked to the processes of invasion and metastasis. One of the key enzymes involved in specifically degrading the heparan sulphate (HS) component of the ECM is the endo-beta-glucuronidase enzyme heparanase. Processing of HS by heparanase releases both a host of bioactive growth factors anchored within the mesh of the ECM as well as defined fragments of HS capable of promoting cellular proliferation. The finding that heparanase is elevated in a wide variety of tumor types and is subsequently linked to the development of pathological processes has led to an explosion of therapeutic strategies to inhibit its enzyme activity. So far only one compound, the sulphated oligosaccharide PI88, which both inhibits heparanase activity and has effects on growth factor binding has reached clinical trials where it has shown to have promising efficacy. The scene has clearly been set however for a new generation of compounds, either specific to the enzyme or with dual roles, to emerge from the lab and enter the clinic. The aim of this review is to describe the current drug discovery status of small molecule, sugar and neutralising antibody inhibitors of heparanase enzyme activity. Potential strategies will also be discussed on the selection of suitable biomarker strategies for specific monitoring of in vivo heparanase inhibition which will be crucial for both animal model and clinical trial testing.

    Topics: Animals; Drug Design; Enzyme Inhibitors; Gene Expression Regulation, Enzymologic; Glucuronidase; Heparitin Sulfate; Humans; Inflammation; Neoplasms; Vascular Endothelial Growth Factor A

2007
Effects of sulfated polysaccharides on tumour biology.
    The West Indian medical journal, 2006, Volume: 55, Issue:4

    Sulfated polysaccharides can act not only as anticoagulants but also as tumour inhibitors. Recent studies suggest that sulfated polysaccharides could affect tumour cells directly. Sulfated polysaccharides could inhibit the metastasis and proliferation of tumour cells by binding to growth factors and cell adhesion molecules. Moreover, sulfated polysaccharides could inhibit heparanase, which cleaves heparan sulfate chains of heparan sulfate proteoglycans and cause release of growth factors sequestered by heparan sulfate chains. Some sulfated polysaccharides can induce apoptosis and differentiation of tumour cells, but the mechanism is uncertain. In addition, sulfated polysaccharides can enhance the innate and adaptive immune response for tumour cells. Thus, the anti-tumour mechanism of sulfated polysaccharides can be explained, at least partly, through the effects on tumour biology directly.

    Topics: Antineoplastic Agents; Apoptosis; Cell Adhesion Molecules; Glucuronidase; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Intercellular Signaling Peptides and Proteins; Neoplasms; Polysaccharides

2006
Natural cytotoxicity receptors: pattern recognition and involvement of carbohydrates.
    TheScientificWorldJournal, 2005, Feb-23, Volume: 5

    Natural cytotoxicity receptors (NCRs), expressed by natural killer (NK) cells, trigger NK lysis of tumor and virus-infected cells on interaction with cell-surface ligands of these target cells. We have determined that viral hemagglutinins expressed on the surface of virus-infected cells are involved in the recognition by the NCRs, NKp44 and NKp46. Recognition of tumor cells by the NCRs NKp30 and NKp46 involves heparan sulfate epitopes expressed on the tumor cell membrane. Our studies provide new evidence for the identity of the ligands for NCRs and indicate that a broader definition should be applied to pathological patterns recognized by innate immune receptors. Since nonmicrobial endogenous carbohydrate structures contribute significantly to this recognition, there is an imperative need to develop appropriate tools for the facile sequencing of carbohydrate moieties.

    Topics: Epitopes; Hemagglutinins, Viral; Heparitin Sulfate; Humans; Immunity, Innate; Killer Cells, Natural; Ligands; Membrane Glycoproteins; Natural Cytotoxicity Triggering Receptor 1; Natural Cytotoxicity Triggering Receptor 2; Natural Cytotoxicity Triggering Receptor 3; Neoplasms; Receptors, Immunologic

2005
Biotechnological engineering of heparin/heparan sulphate: a novel area of multi-target drug discovery.
    Current pharmaceutical design, 2005, Volume: 11, Issue:19

    Heparin is a sulphated glycosaminoglycan currently used as an anticoagulant and antithrombotic drug. It consists largely of 2-O-sulphated IdoA not l&r arrow N, 6-O-disulphated GlcN disaccharide units. Other disaccharides containing unsulphated IdoA or GlcA and N-sulphated or N-acetylated GlcN are also present as minor components. This heterogeneity is more pronounced in heparan sulphate (HS), where the low-sulphated disaccharides are the most abundant. Heparin/HS bind to a variety of biologically active polypeptides, including enzymes, growth factors and cytokines, and viral proteins. This capacity can be exploited to design multi-target heparin/HS-derived drugs for pharmacological interventions in a variety of pathologic conditions besides coagulation and thrombosis, including neoplasia and viral infection. The capsular K5 polysaccharide from Escherichia coli has the same structure as the heparin precursor N-acetyl heparosan. The possibility of producing K5 polysaccharide derivatives by chemical and enzymatic modifications, thus generating heparin/HS-like compounds, has been demonstrated. These K5 polysaccharide derivatives are endowed with different biological properties, including anticoagulant/antithrombotic, antineoplastic, and anti-AIDS activities. Here, the literature data are discussed and the possible therapeutic implications for this novel class of multi-target "biotechnological heparin/HS" molecules are outlined.

    Topics: Animals; Anticoagulants; Bacterial Capsules; Heparin; Heparitin Sulfate; Humans; Neoplasms; Polysaccharides, Bacterial; Technology, Pharmaceutical; Venous Thrombosis

2005
[Research advancement on heparanase in tumor metastasis].
    Ai zheng = Aizheng = Chinese journal of cancer, 2005, Volume: 24, Issue:9

    Heparanase is the only mammalian heparan sulfate-degrading enzymes; it could cleave heparan sulfate (HS) which links to heparan sulfate proteoglycans (HSPGs). In this review, the heparanase's structure, function, molecular properties, gene location, nucleotide sequence, its effects on tumor angiogenesis, its expression in normal tissue, tumor tissue and metastatic tissue, and its correlation to tumor metastasis were described; recent progress in searching for novel antitumor drugs through screening for inhibitor of heparanase was summarized.

    Topics: Animals; Gene Expression Regulation, Neoplastic; Glucuronidase; Heparitin Sulfate; Humans; Lymphatic Metastasis; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; RNA, Messenger

2005
[Role of heparan sulfate endosulfatases in extracellular signal transduction].
    Tanpakushitsu kakusan koso. Protein, nucleic acid, enzyme, 2004, Volume: 49, Issue:15 Suppl

    Topics: Animals; Apoptosis; Carrier Proteins; Cell Transformation, Neoplastic; Endoplasmic Reticulum; Gene Expression Regulation, Developmental; Golgi Apparatus; Heparitin Sulfate; Humans; Intercellular Signaling Peptides and Proteins; Neoplasms; Nervous System; Proteins; Signal Transduction; Sulfatases; Wnt Proteins

2004
The heparins and cancer.
    Current opinion in pulmonary medicine, 2002, Volume: 8, Issue:5

    Heparin, and particularly low molecular weight heparin, is widely used for the treatment of patients with deep vein thrombosis (DVT) and the prevention of DVT that commonly accompanies malignancy. Malignant growth has also been linked experimentally to the function of heparin-like glycosaminoglycans (HLGAGs). In addition to the voluminous general literature on this subject, the heparin-cancer link has been the theme of at least three entire journal issues in recent months. These include the June 15, 2001 issue of Thrombosis Research, the November 2001 Supplement to Haemostasis, and the February 2002 issue of Seminars in Thrombosis and Hemostasis. Previous reviews have documented the historical development of this link that includes evidence from basic biochemistry and cell biology, studies in experimental animal model systems, and clinical trials. This concise review updates recent basic and clinical data on the heparin-cancer link that clarify mechanisms by which HLGAGs regulate the malignant behavior of cells. Electronic access to information that is increasing geometrically has become indispensable. Evidence for control of tumor cell growth by heparin-binding growth factors, tumor cell invasion by heparin-inhibitable enzyme systems, tumor cell metastasis by heparin-binding cell surface selectins, tumor angiogenesis, and tumor matrix formation related to deposition of fibrinogen/fibrin provides a secure theoretical foundation for systematic testing of this class of compounds in patients with cancer. HLGAGs may be a fundamental intermediate in the abnormal growth regulation characteristic of neoplasia that is susceptible to targeted intervention.

    Topics: Anticoagulants; Comorbidity; Heparin, Low-Molecular-Weight; Heparitin Sulfate; Humans; Neoplasms; Venous Thrombosis

2002
Inhibitors of the hyaluronidases.
    Matrix biology : journal of the International Society for Matrix Biology, 2002, Volume: 21, Issue:1

    The inhibitors of hyaluronidase present in mammalian sera, first described half a century ago, have remained uncharacterized. Because of increased interest in hyaluronidases and their hyaluronan substrate, a study of these inhibitors was undertaken recently. The predominant serum inhibitor is magnesium-dependent and is eliminated by protease or chondroitinase digestion, and by heat. Kinetics of inhibition are similar against hyaluronidases from testis, snake and bee venom. The inhibitor has no effect on Streptomyces hyaluronidase; indicating inhibition is not through protection of the hyaluronan substrate. Circulating inhibition levels are increased in mice following carbon tetrachloride or interleukin-1 injection, inducers of the acute-phase response. Reverse hyaluronan gel zymography reveals a predominant band of 120 kDa relative molecular size. Additional studies indicate that the inhibitor resembles a member of the Kunitz type inter-alpha-inhibitor family. Inhibition of hyaluronidase activity is observed using purified inter-alpha-inhibitor and is reversed by antibodies specific for inter-alpha-inhibitor. This molecule, found in the hyaluronan-rich cumulus mass surrounding mammalian ova and the pericellular coat of fibroblasts and mesothelial cells, may function to stabilize such matrices by protecting against hyaluronidase degradation. Turnover of circulating hyaluronan is extraordinarily rapid, with a half-life of two to five min. Prompt increases in levels of serum hyaluronan occur in patients with shock, septicemia or massive burns, increases that may be partly attributed to suppression by these acute phase reactants of the constant and rapid rates of hyaluronan degradation by hyaluronidase. A literature survey of other hyaluronidase inhibitors is also presented.

    Topics: Acute-Phase Reaction; Alpha-Globulins; Animals; Anti-Inflammatory Agents; Enzyme Inhibitors; Heparin; Heparitin Sulfate; Humans; Hyaluronoglucosaminidase; Neoplasms; Plants; Saliva

2002
Heparanase: a key enzyme involved in cell invasion.
    Biochimica et biophysica acta, 2001, Mar-21, Volume: 1471, Issue:3

    Topics: Animals; Antineoplastic Agents; Carbohydrate Sequence; Cloning, Molecular; DNA, Complementary; Endopeptidases; Enzyme Precursors; Extracellular Matrix; Gene Expression Regulation, Enzymologic; Glucuronidase; Heparitin Sulfate; Humans; Molecular Sequence Data; Neoplasms; Oligosaccharides

2001
Heparan sulfate proteoglycans and cancer.
    British journal of cancer, 2001, Oct-19, Volume: 85, Issue:8

    Heparan sulfate proteoglycans (HSPGs) are widely distributed in mammalian tissues and involved in a number of processes related to malignancy. They are composed of a core protein to which chains of the glycosaminoglycan, heparan sulfate (HS), are attached. The existence of various classes of core protein, in addition to highly polymorphic HS chains, creates a superfamily of macromolecules with considerable diversity of structure and function. HSPGs interact with many proteins including growth factors, chemokines and structural proteins of the extracellular matrix to influence cell growth, differentiation, and the cellular response to the environment. The recent identification of two inherited syndromes that are associated with an increased cancer risk, and caused by mutations in HSPG-related genes, has intensified interest in these molecules. This review describes our current understanding of HSPGs in cancer and highlights new possibilities for therapeutic control.

    Topics: Animals; Cell Division; Glucuronidase; Glycosylation; Glypicans; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Neoplasm Invasiveness; Neoplasms; Neovascularization, Pathologic

2001
Factors promoting tumor angiogenesis.
    Cancer investigation, 1999, Volume: 17, Issue:8

    Topics: Angiogenesis Inhibitors; Collagen; Endothelial Growth Factors; Fibronectins; Growth Substances; Heparitin Sulfate; Humans; Laminin; Lymphokines; Neoplasms; Neovascularization, Pathologic; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factors

1999
Glycoconjugate cross-talk in metastatic cancer cells, leucocytes, parasites and bacteria.
    Biochemical Society transactions, 1997, Volume: 25, Issue:1

    Topics: Animals; Bacterial Physiological Phenomena; Cell Communication; Glycoconjugates; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Leukocytes; Lymphocytes; Models, Structural; Neoplasm Metastasis; Neoplasms; Parasites; Proteoglycans

1997
Syndecans: multifunctional cell-surface co-receptors.
    The Biochemical journal, 1997, Oct-01, Volume: 327 ( Pt 1)

    This review will summarize our current state of knowledge of the structure, biochemical properties and functions of syndecans, a family of transmembrane heparan sulphate proteoglycans. Syndecans bind a variety of extracellular ligands via their covalently attached heparan sulphate chains. Syndecans have been proposed to play a role in a variety of cellular functions, including cell proliferation and cell-matrix and cell-cell adhesion. Syndecan expression is highly regulated and is cell-type- and developmental-stage-specific. The main function of syndecans appears to be to modulate the ligand-dependent activation of primary signalling receptors at the cell surface. Principal functions of the syndecan core proteins are to target the heparan sulphate chains to the appropriate plasma-membrane compartment and to interact with components of the actin-based cytoskeleton. Several functions of the syndecans, including syndecan oligomerization and actin cytoskeleton association, have been localized to specific structural domains of syndecan core proteins.

    Topics: Amino Acid Sequence; Animals; Cell Adhesion; Cell Movement; Cytoskeleton; Heparitin Sulfate; Humans; Ligands; Membrane Glycoproteins; Molecular Sequence Data; Neoplasms; Proteoglycans; Receptors, Cell Surface; Signal Transduction; Syndecans

1997
Structure-activity relationship of heparan sulphate.
    Biochemical Society transactions, 1997, Volume: 25, Issue:4

    HS influences fundamental cellular properties and biochemical processes at the cell surface. In addition to the issues already discussed, it has a profound effect on cell adhesion and migration through its interaction with many extracellular matrix proteins, most notably fibronectin and thrombospondin; it is closely linked to lipid metabolism through its capacity to bind low-density lipoprotein and lipoprotein lipase; and aberrations in HS structure and degradation are linked to human malignancy and Alzheimer's disease [26,27]. The subtle variations in HS structure enable it to distinguish between families of related proteins such as the FGFs, the chemokines [28] and the TGF beta s [29]. The multifunctional nature of HS is the result of its structural diversity and strategic positioning in the pericellular domain. The biosynthesis of HS, in common with other complex carbohydrates, is not directed by any known template yet the system is clearly subject to quite precise control so that in general, the HS family has a common domain organization that is finely tuned at the cellular level to produce HS species of variable length, fine structure and biological properties. A major challenge for future research will be to unravel the regulatory mechanisms that determine the molecular structure of HS. It remains unclear whether these mechanisms are entirely intrinsic in nature or subject to substantial modulation by the cellular microenvironment.

    Topics: Alzheimer Disease; Animals; Carbohydrate Sequence; Chemokines; Heparitin Sulfate; Humans; Molecular Sequence Data; Neoplasms; Structure-Activity Relationship

1997
Inhibition of fibroblast growth factors.
    Breast cancer research and treatment, 1996, Volume: 38, Issue:1

    The potential roles of members of the fibroblast growth factor family in tumor angiogenesis and metastasis and their mechanisms of release from cells are discussed. Furthermore, we review methods of therapeutic targeting of these polypeptides. In particular, we focus on the possibility to inhibit fibroblast growth factors with drugs that mimic heparin-like cellular binding sites and thus can interfere with growth factor receptor recognition. In addition, we discuss antibodies, antisense oligodeoxynucleotides, and ribozymes as approaches to inhibit production and activity of these growth factors.

    Topics: Animals; Fibroblast Growth Factor 2; Fibroblast Growth Factors; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Neoplasms; Neovascularization, Pathologic; Oligonucleotides, Antisense; Pentosan Sulfuric Polyester; Proteoglycans

1996
Tumor targeting through fibroblast growth factor receptors.
    Seminars in cancer biology, 1995, Volume: 6, Issue:5

    Fibroblast growth factor tyrosine kinase receptors are encoded by four genes, but alternate splicing can result in more than 100 possible protein sequences. The receptors have widespread expression in the developing embryo, but the expression becomes more restricted in the adult. The ligand-receptor relationship is complex, due to the diversity of the receptors and the large number of possible ligands: there are now nine (and probably more) members of the fibroblast growth factor family. This complicated ligand-receptor relationship creates many options to target cell types through the use of individual ligands or receptor-specific monoclonal antibodies. In-vivo data demonstrate that FGF receptors are expressed on tumor cells and can be used to target tumors for growth inhibition. Given the complexity, it is possible that a unique targetable FGF receptor isoform can be found in one or more tumor types. Examples of the targeting of growth inhibition agents to tumors through FGF receptors are discussed.

    Topics: Amino Acid Sequence; Animals; Fibroblast Growth Factors; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Immunotoxins; Molecular Sequence Data; N-Glycosyl Hydrolases; Neoplasms; Plant Proteins; Proteoglycans; Receptor Protein-Tyrosine Kinases; Receptors, Fibroblast Growth Factor; Recombinant Fusion Proteins; Ribosome Inactivating Proteins, Type 1; Saporins

1995
[Morphology of basement membrane and associated matrix proteins in normal and pathological tissues].
    Veroffentlichungen aus der Pathologie, 1995, Volume: 145

    Basement membranes (BM) are specialized structures of the extracellular matrix. Their composition is of particular importance for the maintenance of normal morphological and functional properties of a multitude of organs and tissue systems and it is thus required for regular homeostasis of body function. Generally, they possess three main functions, i.e. participation in the maintenance of tissue structure, control of fluid and substrate exchange, and regulation of cell growth and differentiation. BMs are made up by various components which are in part specifically localized within the BM zone, or which represent ubiquitous matrix constituents with specific quantitative and/or qualitative differences in their localization. On the basis of a thorough immunohistochemical analysis of normal and diseased tissues, we provide here a concept of "functional morphology/pathomorphology" of the different BM components analyzed: 1.) The ubiquitous BM-constituent collagen IV primarily stabilizes the BM-zone and thus represents the "backbone" of the BM providing mechanical strength. Its loss leads to cystic tissue transformation as it is evidenced from the analysis of polycystic nephropathies. Thus, in other cystic tissue transformations a similar formal pathogenesis may be present. 2.) The specific localization of collagen VII as the main structural component of anchoring fibrils underlines the mechanical anchoring function of this collagenous protein. Defects in this protein lead to hereditary epidermolysis. The rapid re-occurrence of epidermal collagen VII during normal human wound healing indicates a quick reconstitution of the mechanical tensile strength of healing wounds. 3.) The BM-specific heparan sulfate proteoglycan (HSPG, Perlecan) with its highly negative anionic charge can be assumed to exert filter control. This assumption is corroborated by the localizatory findings of a preferential deposition of HSPG in endothelial and particularly in glomerular BM. Similarly, the lack of HSPG in the BM of lymph capillaries can be regarded as the correlate for a free fluid influx into lymphatic capillaries. The relative reduction in HSPG-staining in the developing glomerular BM also explains the still immature filter function. Furthermore, the low content of HSPG in placental chorionic capillaries can be regarded as morphological correlate for the required free fluid exchange between maternal and fetal blood systems. In diabetic glomerulopathy, the loss of HSPG coincid

    Topics: Animals; Basement Membrane; Collagen; Diabetic Nephropathies; Extracellular Matrix Proteins; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Immunohistochemistry; Kidney Diseases; Neoplasms; Proteoglycans; Pulmonary Fibrosis; Wound Healing

1995
The biology of perlecan: the multifaceted heparan sulphate proteoglycan of basement membranes and pericellular matrices.
    The Biochemical journal, 1994, Sep-15, Volume: 302 ( Pt 3)

    Topics: Amino Acid Sequence; Animals; Basement Membrane; Diabetic Nephropathies; Extracellular Matrix; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Molecular Sequence Data; Muscles; Neoplasms; Promoter Regions, Genetic; Proteoglycans; Sequence Homology, Amino Acid

1994
Sequestration and release of basic fibroblast growth factor.
    Annals of the New York Academy of Sciences, 1991, Volume: 638

    Topics: Animals; Basement Membrane; Blood Platelets; Cell Compartmentation; Cell Differentiation; Cell Division; Endothelium; Extracellular Matrix; Fibroblast Growth Factor 2; Glucuronidase; Glycoside Hydrolases; Heparitin Sulfate; Humans; Neoplasm Metastasis; Neoplasms; Neurons; Neutrophils; Phosphatidylinositol Diacylglycerol-Lyase; Phosphoric Diester Hydrolases

1991
Biology and pathology of the pericellular heparan sulphate proteoglycans.
    Biochemical Society transactions, 1991, Volume: 19, Issue:4

    Topics: Animals; Cell Membrane; Growth; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Molecular Structure; Neoplasms; Proteoglycans

1991
Proteoglycans and neoplasia.
    Cancer metastasis reviews, 1988, Volume: 7, Issue:1

    There is a growing realization that the whole tumor cell-matrix complex must be investigated in order to fully understand the process of cancer growth and metastasis. Proteoglycans are intrinsic constituents of the cell surface, extracellular matrix, and basement membrane, three logistically and functionally important structures involved in most cellular interactions. Proteoglycans influence the behavior of normal and malignant cells by virtue of their expanded configuration, polyanionic nature and, most of all, by their ability to interact with a variety of cellular products. Consequently, they have been implicated in a number of biological processes including proliferation, recognition, adhesion, and migration. They can serve as links between the extracellular and intracellular environment and thus transduce key biological signals. They can act as receptors for interstitial collagens and other matrix proteins and thus contribute to the organization of pericellular matrix. During neoplastic development there is a profound structural rearrangement of these macromolecules at both the plasma membrane and the pericellular level. Qualitative and quantitative abnormalities in proteoglycan metabolism may contribute to the establishment of some well-known neoplastic properties, including lack of cohesiveness, abnormal assembly of extracellular matrix, abnormal growth, and invasion. The present work will focus on recent advances in our understanding of these complex macromolecules and on some of the alterations associated with the neoplastic phenotype, and will then attempt to elucidate some of the mechanisms regulating these changes.

    Topics: Biological Products; Cell Division; Cell Transformation, Neoplastic; Cytokines; Glycosaminoglycans; Heparitin Sulfate; Humans; Neoplasms; Proteoglycans

1988
Biochemical interactions of tumor cells with the basement membrane.
    Annual review of biochemistry, 1986, Volume: 55

    Topics: Basement Membrane; Chondroitin Sulfate Proteoglycans; Fibronectins; Genetic Linkage; Glucuronidase; Glycoside Hydrolases; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Laminin; Microbial Collagenase; Models, Biological; Neoplasm Metastasis; Neoplasm Proteins; Neoplasms; Phenotype; Receptors, Fibronectin; Receptors, Immunologic; Receptors, Laminin

1986
Proteoglycans: structure, function, and role in neoplasia.
    Laboratory investigation; a journal of technical methods and pathology, 1985, Volume: 53, Issue:4

    Topics: Animals; Basement Membrane; Cartilage; Chemical Phenomena; Chemistry; Chondroitin Sulfate Proteoglycans; Dermatan Sulfate; Extracellular Matrix; Glycosaminoglycans; Heparan Sulfate Proteoglycans; Heparin; Heparitin Sulfate; Humans; Hyaluronic Acid; Keratan Sulfate; Lumican; Models, Biological; Neoplasms; Proteoglycans; Staining and Labeling

1985
Basement membranes in cancer.
    Ultrastructural pathology, 1985, Volume: 8, Issue:4

    Basement membranes are ubiquitous tissue constituents which occur as supportive structure adjacent to epithelium, endothelium, mesothelium and also around smooth as well as striated muscle cells, Schwann cells and fat cells. In various types of cancer, basement membranes have been extensively studied by electron microscopy. Often basement membrane interruptions were seen in invasive neoplasms but in some tumors the neoplastic cells were surrounded by a continuous basal lamina. Recent immunocytochemical studies have shown that in invasive carcinomas the neoplastic cells often lack a continuous basement membrane. This may be caused by catabolic activity of invasive tumor cells, which have been shown to produce specific collagenases, or by insufficient production and/or extracellular assembly of basement membrane components by the neoplastic epithelial cells. In diagnostic histopathology, immunocytochemical staining of basement membrane components such as type IV collagen and laminin may help to distinguish between noninvasive (benign or in situ) and invasive lesions. Furthermore, in carcinomas the extent of the expression of basement membrane components may be correlated with the degree of differentiation of the neoplastic cells. Finally, in soft tissue tumors, basement membrane staining may be helpful for the differentiation of basement membrane producing neoplasms (e.g. of vascular, neural, smooth muscle or striated muscle origin) from non-basement membrane producing neoplasms (e.g. of fibroblastic origin).

    Topics: Basement Membrane; Cell Differentiation; Cell Transformation, Neoplastic; Chondroitin Sulfate Proteoglycans; Collagen; Fluorescent Antibody Technique; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Immunoenzyme Techniques; Laminin; Microscopy, Electron; Neoplasm Invasiveness; Neoplasms

1985
Proteoglycans and cell adhesion. Their putative role during tumorigenesis.
    Cancer metastasis reviews, 1984, Volume: 3, Issue:4

    In this review, evidence that proteoglycans are involved in cell adhesion and related behavior is considered, together with their putative role(s) during tumorigenesis. Proteoglycans are large, carboxylated and/or sulfated structures that interact with specific binding sites on cell surfaces. Their distribution and synthesis in tissues alter with the onset of tumorigenesis so that hyaluronic acid is generally increased and heparan sulfate decreased in the developing tumor and surrounding tissue. However, the precise role of proteoglycans during the tumorigenic process is far from clarified. Data suggest any putative roles will be related to the adhesive properties that these molecules confer to cells. Hyaluronic acid and chondroitin sulfate appear to be weakly adhesive molecules that may promote 'transformed' characteristics when they occur on cells in large amounts. These characteristics include reduced cell spreading, increased cell motility, as well as reduced contact inhibition. Consistent with such properties, neither hyaluronic acid nor chondroitin sulfate are localized in specialized adhesion sites such as focal or close contacts. In contrast, heparan sulfate is associated with increased cell-substratum adhesion and is involved in the spreading of cells onto fibronectin and other substrata. Its presence is generally associated with reduced motility and with a well-spread morphology. Unlike hyaluronate and chondroitin sulfate, heparan sulfate is found in specialized contacts. These adhesive properties of proteoglycans predict an instructive role in tumor development, and recent experiments have defined an involvement of these molecules in metastatic arrest. Additional studies utilizing invasive and metastatic tumor variants including tumor cells that employ different mechanisms to invade are required to clarify the role of proteoglycans in tumor progression.

    Topics: Animals; Carrier Proteins; Cell Adhesion; Cell Movement; Chemical Phenomena; Chemistry; Chondroitin Sulfate Proteoglycans; Chondroitin Sulfates; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Hyaluronan Receptors; Hyaluronic Acid; Neoplasms; Proteoglycans; Receptors, Cell Surface

1984
Structure, function, and pathology of basement membranes.
    Monographs in pathology, 1983, Volume: 24

    Basement membranes support epithelial and endothelial cells, prevent the passage of proteins, and generate histologically distinct compartments in the body. Basement membranes contain a number of components, only some of which have been isolated and characterized. These include type IV collagen, heparan sulfate proteoglycan, laminin, entactin, and fibronectin. Some components, such as bullous pemphigoid antigen and Goodpasture antigen, are present only in specific tissues, such as the skin or the kidneys. Alterations in basement membranes are associated with various diseases. For example, metastatic cells are able to attach to basement membranes and to degrade them. Such interactions with basement membranes underlie the ability of these cells to penetrate tissues and to spread in the body. In diabetes, basement membranes are thickened but are more porous, which is possibly due to a decreased amount of heparan sulfate proteoglycan. Basement membranes are also the site of immunopathologic disorders.

    Topics: Autoimmune Diseases; Basement Membrane; Chondroitin Sulfate Proteoglycans; Collagen; Diabetes Mellitus; Glycoproteins; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Neoplasms; Proteoglycans

1983
The biochemical properties of basement membrane components in health and disease.
    Clinical biochemistry, 1980, Volume: 13, Issue:5

    Basement membranes are complex macromolecular structures which occupy the extracellular space between cells of different histologic types. Biochemically it is composed of Type IV collagen, several noncollagenous glycoproteins including laminin, fibronectin, GP-2 and PYS glycoprotein, and heparan sulfate. Morphologic changes are commonplace in a number of renal diseases. In diabetic glomerular disease, the basement membrane is markedly thickened but the biochemical basis has not been elucidated. In other disease-associated basement membrane changes, altered glycosylation of glycoprotein components has been described. The most important issue is the effect such alterations have on the interaction of basement membrane components and the function of the basement membrane.

    Topics: Amino Acids; Animals; Basement Membrane; Collagen; Diabetes Mellitus; Glycoproteins; Heparitin Sulfate; Humans; Kidney Diseases; Neoplasms

1980

Trials

2 trial(s) available for heparitin-sulfate and Neoplasms

ArticleYear
Risk factors for bleeding during treatment of acute venous thromboembolism.
    Thrombosis and haemostasis, 1996, Volume: 76, Issue:5

    Identification of risk factors for bleeding and prospective evaluation of two bleeding risk scores in the treatment of acute venous thromboembolism.. Secondary analysis of a prospective, randomized, assessorblind, multicenter clinical trial.. One university and 2 regional teaching hospitals.. 188 patients treated with heparin or danaparoid for acute venous thromboembolism.. The presenting clinical features, the doses of the drugs, and the anticoagulant responses were analyzed using univariate and multivariate logistic regression analysis in order to evaluate prognostic factors for bleeding. In addition, the recently developed Utrecht bleeding risk score and Landefeld bleeding risk index were evaluated prospectively.. Major bleeding occurred in 4 patients (2.1%) and minor bleeding in 101 patients (53.7%). For all (major and minor combined) bleeding, body surface area < or = 2 m2 (odds ratio 2.3, 95% CI 1.2-4.4; p = 0.01), and malignancy (odds ratio 2.4, 95% CI 1.1-4.9; p = 0.02) were confirmed to be independent risk factors. An increased treatment-related risk of bleeding was observed in patients treated with high doses of heparin, independent of the concomitant activated partial thromboplastin time ratios. Both bleeding risk scores had low diagnostic value for bleeding in this sample of mainly minor bleeders.. A small body surface area and malignancy were associated with a higher frequency of bleeding. The bleeding risk scores merely offer the clinician a general estimation of the risk of bleeding. In patients with a small body surface area or in patients with malignancy, it may be of interest to study whether limited dose reduction of the anticoagulant drug may cause less bleeding without affecting efficacy.

    Topics: Acenocoumarol; Acute Disease; Adult; Aged; Body Surface Area; Chondroitin Sulfates; Comorbidity; Dermatan Sulfate; Drug Combinations; Female; Fibrinolytic Agents; Hemorrhage; Heparin; Heparitin Sulfate; Humans; Male; Middle Aged; Neoplasms; Odds Ratio; Prospective Studies; Risk Factors; Single-Blind Method; Thromboembolism; Thrombolytic Therapy

1996
Early clinical experience of a new heparinoid, Org 10172, in prevention of deep venous thrombosis.
    Thrombosis research, 1987, Mar-01, Volume: 45, Issue:5

    The potential antithrombotic effect of a new low molecular weight heparinoid, Org 10172, was examined in a randomized, double-blind, placebo-controlled, dose-ranging pilot study of the prevention of deep venous thrombosis (DVT) in 45 high-risk patients having major thoracic or abdominal surgery for cancer. Org 10172 was given in doses of 500, 750 or 1000 U bd subcutaneously. DVT occurred in 9 of 14 patients given placebo and in 4 of 11 patients given 500 U bd but in none of the 20 patients given 750 or 1000 U bd. Operative blood loss and post-operative bleeding were not significantly different between the groups but one patient given 1000 U bd had major post-operative bleeding. Average mid-interval and trough plasma anti-Xa levels reached 0.26 and 0.20 U/ml respectively following the highest dose. It is concluded that Org 10172 is a potentially useful antithrombotic agent and that the effective and safe dose appears to be between 500 and 1000 U bd for prevention of DVT in high-risk patients.

    Topics: Aged; Aged, 80 and over; Chondroitin Sulfates; Dermatan Sulfate; Factor X; Factor Xa; Female; Glycosaminoglycans; Hemorrhage; Heparitin Sulfate; Humans; Injections, Subcutaneous; Male; Middle Aged; Neoplasms; Pilot Projects; Research Design; Thoracic Surgery; Thrombophlebitis

1987

Other Studies

50 other study(ies) available for heparitin-sulfate and Neoplasms

ArticleYear
Heparan sulfate proteoglycans in cancer: Pathogenesis and therapeutic potential.
    Advances in cancer research, 2023, Volume: 157

    The heparan sulfate proteoglycans (HSPGs) are glycoproteins that consist of a proteoglycan "core" protein and covalently attached heparan sulfate (HS) chain. HSPGs are ubiquitously expressed in mammalian cells on the cell surface and in the extracellular matrix (ECM) and secretory vesicles. Within HSPGs, the protein cores determine when and where HSPG expression takes place, and the HS chains mediate most of HSPG's biological roles through binding various protein ligands, including cytokines, chemokines, growth factors and receptors, morphogens, proteases, protease inhibitors, and ECM proteins. Through these interactions, HSPGs modulate cell proliferation, adhesion, migration, invasion, and angiogenesis to display essential functions in physiology and pathology. Under physiological conditions, the expression and localization of HSPGs are finely regulated to orchestrate their physiological functions, and this is disrupted in cancer. The HSPG dysregulation elicits multiple oncogenic signaling, including growth factor signaling, ECM and Integrin signaling, chemokine and immune signaling, cancer stem cell, cell differentiation, apoptosis, and senescence, to prompt cell transformation, proliferation, tumor invasion and metastasis, tumor angiogenesis and inflammation, and immunotolerance. These oncogenic roles make HSPGs an attractive pharmacological target for anti-cancer therapy. Several therapeutic strategies have been under development, including anti-HSPG antibodies, peptides and HS mimetics, synthetic xylosides, and heparinase inhibitors, and shown promising anti-cancer efficacy. Therefore, much progress has been made in this line of study. However, it needs to bear in mind that the roles of HSPGs in cancer can be either oncogenic or tumor-suppressive, depending on the HSPG and the cancer cell type with the underlying mechanisms that remain obscure. Further studies need to address these to fill the knowledge gap and rationalize more efficient therapeutic targeting.

    Topics: Animals; Cell Differentiation; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Mammals; Neoplasms; Neovascularization, Pathologic

2023
Conditional ablation of heparan sulfate expression in stromal fibroblasts promotes tumor growth in vivo.
    PloS one, 2023, Volume: 18, Issue:2

    Heparan sulfate (HS) is a glycocalyx component present in the extracellular matrix and cell-surface HS proteoglycans (HSPGs). Although HSPGs are known to play functional roles in multiple aspects of tumor development and progression, the effect of HS expression in the tumor stroma on tumor growth in vivo remains unclear. We conditionally deleted Ext1, which encodes a glycosyltransferase essential for the biosynthesis of HS chains, using S100a4-Cre (S100a4-Cre; Ext1f/f) to investigate the role of HS in cancer-associated fibroblasts, which is the main component of the tumor microenvironment. Subcutaneous transplantation experiments with murine MC38 colon cancer and Pan02 pancreatic cancer cells demonstrated substantially larger subcutaneous tumors in S100a4-Cre; Ext1f/f mice. Additionally, the number of myofibroblasts observed in MC38 and Pan02 subcutaneous tumors of S100a4-Cre; Ext1f/f mice decreased. Furthermore, the number of intratumoral macrophages decreased in MC38 subcutaneous tumors in S100a4-Cre; Ext1f/f mice. Finally, the expression of matrix metalloproteinase-7 (MMP-7) markedly increased in Pan02 subcutaneous tumors in S100a4-Cre; Ext1f/f mice, suggesting that it may contribute to rapid growth. Therefore, our study demonstrates that the tumor microenvironment with HS-reduced fibroblasts provides a favorable environment for tumor growth by affecting the function and properties of cancer-associated fibroblasts, macrophages, and cancer cells.

    Topics: Animals; Extracellular Matrix; Extracellular Matrix Proteins; Fibroblasts; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Mice; Neoplasms; Tumor Microenvironment

2023
Induction of heparanase 2 (Hpa2) expression by stress is mediated by ATF3.
    Matrix biology : journal of the International Society for Matrix Biology, 2022, Volume: 105

    Activity of heparanase, endoglycosidase that cleaves heparan sulfate side chains in heparan sulfate proteoglycans, is highly implicated in tumor progression and metastasis. Heparanase inhibitors are therefore being evaluated clinically as anti-cancer therapeutics. Heparanase 2 (Hpa2) is a close homolog of heparanase that lacks HS-degrading activity and functions as an endogenous inhibitor of heparanase. As a result, Hpa2 appears to attenuate tumor growth but mechanisms that regulate Hpa2 expression and determine the ratio between heparanase and Hpa2 are largely unknown. We have recently reported that the expression of Hpa2 is induced by endoplasmic reticulum (ER) and proteotoxic stresses, but the mechanism(s) underlying Hpa2 gene regulation was obscure. Here we expand the notion that Hpa2 is regulated by conditions of stress. We report that while ER and hypoxia, each alone, resulted in a 3-7 fold increase in Hpa2 expression, combining ER stress and hypoxia resulted in a noticeable, over 40-fold increase in Hpa2 expression. A prominent induction of Hpa2 expression was also quantified in cells exposed to heat shock, proteotoxic stress, lysosomal stress, and chemotherapy (cisplatin), strongly implying that Hpa2 is regulated by conditions of stress. Furthermore, analyses of the Hpa2 gene promoter led to the identification of activating-transcription-factor 3 (ATF3) as a transcription factor that mediates Hpa2 induction by stress, thus revealing, for the first time, a molecular mechanism that underlies Hpa2 gene regulation. Induction of Hpa2 and ATF3 by conditions of stress that often accompany the rapid expansion of tumors is likely translated to improved survival of cancer patients.

    Topics: Activating Transcription Factor 3; Glucuronidase; Heparitin Sulfate; Humans; Neoplasms

2022
Structural Insights into Pixatimod (PG545) Inhibition of Heparanase, a Key Enzyme in Cancer and Viral Infections.
    Chemistry (Weinheim an der Bergstrasse, Germany), 2022, Feb-19, Volume: 28, Issue:11

    Pixatimod (PG545), a heparan sulfate (HS) mimetic and anticancer agent currently in clinical trials, is a potent inhibitor of heparanase. Heparanase is an endo-β-glucuronidase that degrades HS in the extracellular matrix and basement membranes and is implicated in numerous pathological processes such as cancer and viral infections, including SARS-CoV-2. To understand how PG545 interacts with heparanase, we firstly carried out a conformational analysis through a combination of NMR experiments and molecular modelling which showed that the reducing end β-D-glucose residue of PG545 adopts a distorted conformation. This was followed by docking and molecular dynamics simulations to study the interactions of PG545 with heparanase, revealing that PG545 is able to block the active site by binding in different conformations, with the cholestanol side-chain making important hydrophobic interactions. While PG545 blocks its natural substrate HS from binding to the active site, small synthetic heparanase substrates are only partially excluded, and thus pentasaccharide or larger substrates are preferred for assaying this class of inhibitor. This study provides new insights for the design of next-generation heparanase inhibitors and substrates.

    Topics: COVID-19; Glucuronidase; Heparitin Sulfate; Humans; Neoplasms; SARS-CoV-2; Virus Diseases

2022
Pingyangmycin inhibits glycosaminoglycan sulphation in both cancer cells and tumour tissues.
    Journal of cellular and molecular medicine, 2020, Volume: 24, Issue:6

    Pingyangmycin is a clinically used anticancer drug and induces lung fibrosis in certain cancer patients. We previously reported that the negatively charged cell surface glycosaminoglycans are involved in the cellular uptake of the positively charged pingyangmycin. However, it is unknown if pingyangmycin affects glycosaminoglycan structures. Seven cell lines and a Lewis lung carcinoma-injected C57BL/6 mouse model were used to understand the cytotoxicity of pingyangmycin and its effect on glycosaminoglycan biosynthesis. Stable isotope labelling coupled with LC/MS method was used to quantify glycosaminoglycan disaccharide compositions from pingyangmycin-treated and untreated cell and tumour samples. Pingyangmycin reduced both chondroitin sulphate and heparan sulphate sulphation in cancer cells and in tumours. The effect was persistent at different pingyangmycin concentrations and at different exposure times. Moreover, the cytotoxicity of pingyangmycin was decreased in the presence of soluble glycosaminoglycans, in the glycosaminoglycan-deficient cell line CHO745, and in the presence of chlorate. A flow cytometry-based cell surface FGF/FGFR/glycosaminoglycan binding assay also showed that pingyangmycin changed cell surface glycosaminoglycan structures. Changes in the structures of glycosaminoglycans may be related to fibrosis induced by pingyangmycin in certain cancer patients.

    Topics: A549 Cells; Animals; Antibiotics, Antineoplastic; Bleomycin; Cell Line, Tumor; CHO Cells; Chondroitin Sulfates; Cricetulus; Glycosaminoglycans; HCT116 Cells; Heparitin Sulfate; HT29 Cells; Humans; Mass Spectrometry; Mice; Mice, Inbred C57BL; Neoplasms; Pulmonary Fibrosis

2020
Circulating heparan sulfate chains and body weight contribute to anti-Xa levels in cancer patients using the prophylactic dose of enoxaparin.
    Journal of thrombosis and thrombolysis, 2020, Volume: 50, Issue:1

    Hospitalized cancer patients are at increased risk of thrombosis and prophylaxis with heparin is recommended. Heparanase is a protein capable of degrading heparan sulfate (HS) chains. The first objective of the study was to examine the effects of weight on anti-Xa levels in cancer patients treated with a fixed dose of enoxaparin as thromboprophylaxis. The second aim was to assess a potential correlation between plasma pre-treatment coagulation parameters and anti-Xa levels in an assumption that heparanase degradation activity towards heparins and HS chains could affect anti-Xa levels. Two blood samples (prior to and 3 h after drug injection) of 76 cancer patients with an indication for prophylaxis with enoxaparin (40 mg) were evaluated for coagulation markers. Sub-prophylactic levels of anti-Xa (< 0.2 U/ml) were found in 16/76 (21%) patients; in 13/76 (13%) patients the values were supra-prophylactic (> 0.5 U/ml). In the subgroup of patients weighing > 80 kg, 7/14 (50%) individuals had a sub-prophylactic level. Overall, anti-Xa levels appeared to correlate with patient's weight (r = - 0.48, p < 0.0001), pre-treatment partial thromboplastin time (PTT), D-dimer, HS, heparanase levels and procoagulant activity. We concluded that plasma anti-Xa levels correlated with patient's weight. A substantial portion of cancer patients receiving enoxaparin prophylaxis was undertreated. For patients > 80 kg, a weight-adjusted prophylactic dose of enoxaparin could be considered. Elevated enoxaparin anti-Xa levels correlated with pre-treatment parameters of coagulation system activation. High pre-treatment HS and elevated plasma anti-Xa levels may potentially serve as biomarkers for the identification of patients at increased thrombosis risk.

    Topics: Anticoagulants; Biomarkers, Pharmacological; Blood Coagulation; Body Weight; Drug Dosage Calculations; Enoxaparin; Factor Xa; Female; Fibrin Fibrinogen Degradation Products; Heparitin Sulfate; Humans; Male; Middle Aged; Neoplasms; Thrombosis

2020
Differences of the tumour cell glycocalyx affect binding of capsaicin-loaded chitosan nanocapsules.
    Scientific reports, 2020, 12-31, Volume: 10, Issue:1

    The glycocalyx regulates the interaction of mammalian cells with extracellular molecules, such as cytokines. However, it is unknown to which extend the glycocalyx of distinct cancer cells control the binding and uptake of nanoparticles. In the present study, exome sequencing data of cancer patients and analysis of distinct melanoma and bladder cancer cell lines suggested differences in cancer cell-exposed glycocalyx components such as heparan sulphate. Our data indicate that glycocalyx differences affected the binding of cationic chitosan nanocapsules (Chi-NCs). The pronounced glycocalyx of bladder cancer cells enhanced the internalisation of nanoencapsulated capsaicin. Consequently, capsaicin induced apoptosis in the cancer cells, but not in the less glycosylated benign urothelial cells. Moreover, we measured counterion condensation on highly negatively charged heparan sulphate chains. Counterion condensation triggered a cooperative binding of Chi-NCs, characterised by a weak binding rate at low Chi-NC doses and a strongly increased binding rate at high Chi-NC concentrations. Our results indicate that the glycocalyx of tumour cells controls the binding and biological activity of nanoparticles. This has to be considered for the design of tumour cell directed nanocarriers to improve the delivery of cytotoxic drugs. Differential nanoparticle binding may also be useful to discriminate tumour cells from healthy cells.

    Topics: Antipruritics; Capsaicin; Cell Line, Tumor; Cell Survival; Chitosan; Endothelial Cells; Glycocalyx; Heparitin Sulfate; Humans; Nanocapsules; Neoplasms; Organ Specificity; Protein Binding; Static Electricity; Theranostic Nanomedicine

2020
In Vivo Environment-Adaptive Nanocomplex with Tumor Cell-Specific Cytotoxicity Enhances T Cells Infiltration and Improves Cancer Therapy.
    Small (Weinheim an der Bergstrasse, Germany), 2019, Volume: 15, Issue:43

    Drug delivery strategies possessing selectivity for cancer cells are eagerly needed in therapy of metastatic breast cancer. In this study, the chemotherapeutic agent, docetaxel (DTX), is conjugated onto heparan sulfate (HS). Aspirin (ASP), which has the activity of anti-metastasis and enhancing T cells infiltration in tumors, is encapsulated into the HS-DTX micelle. Then the cationic polyethyleneimine (PEI)-polyethylene glycol (PEG) copolymer binds to HS via electrostatic force, forming the ASP-loaded HS-DTX micelle (AHD)/PEI-PEG nanocomplex (PAHD). PAHD displays long circulation behavior in blood due to the PEG shell. Under the tumor microenvironment with weakly acidic pH, PEI-PEG separates from AHD, and the free cationic PEI-PEG facilitates the cellular uptake of AHD by increasing permeability of cell membranes. Then the overexpressed heparanase degrades HS, releasing ASP and DTX. PAHD shows specific toxicity toward tumor cells but not normal cells, with advanced activity of inhibiting tumor growth and lung metastasis in 4T1 tumor-bearing mice. The number of CD8

    Topics: Animals; Antineoplastic Agents; Aspirin; CD8-Positive T-Lymphocytes; Cell Death; Cell Line, Tumor; Docetaxel; Endocytosis; Heparitin Sulfate; Humans; MCF-7 Cells; Mice, Inbred BALB C; Nanoparticles; Neoplasm Metastasis; Neoplasms; Polyethylene Glycols; Polyethyleneimine; Tissue Distribution

2019
Heparan sulfate disaccharide measurement from biological samples using pre-column derivatization, UPLC-MS and single ion monitoring.
    Analytical biochemistry, 2017, 08-01, Volume: 530

    Glycosaminoglycans are a heterogeneous family of linear polysaccharides comprised of repeating disaccharide subunits that mediate many effects at the cellular level. There is increasing evidence that the nature of these effects is determined by differences in disaccharide composition. However, the determination of GAG disaccharide composition in biological samples remains challenging and time-consuming. We have developed a method that uses derivatization and selected ion recording and RP-UPLCMS resulting in rapid separation and quantification of twelve heparin/heparin sulfate disaccharides from 5 μg GAG. Limits of detection and quantitation were 0.02-0.15 and 0.07-0.31 μg/ml respectively. We have applied this method to the novel analysis of disaccharide levels extracted from heparan sulfate and human cancer cell lines. Heparan sulfate disaccharides extracted from biological samples following actinase and heparinase incubation and derivatized using reductive amination with 2-aminoacridone. Derivatized disaccharides were analyzed used UPLC-MS with single ion monitoring. Eight HS disaccharide subunits were separated and quantified from HS and cell lines in eleven minutes per sample. In all samples the most abundant subunits present were the unsulfated ΔUA-GlcNAc, ΔUA-GlcNAc,6S and ΔUA,2S-GlcNS,6S. There was considerable variation in the proportions and concentrations of disaccharides between different cell lines. Further studies are needed to examine the significance of these differences.

    Topics: Aminoacridines; Chromatography, High Pressure Liquid; Disaccharides; Heparin; Heparin Lyase; Heparitin Sulfate; Humans; Mass Spectrometry; Neoplasms; Tumor Cells, Cultured

2017
Synthesis, radiolabeling with fluorine-18 and preliminary in vivo evaluation of a heparan sulphate mimetic as potent angiogenesis and heparanase inhibitor for cancer applications.
    Organic & biomolecular chemistry, 2016, Feb-14, Volume: 14, Issue:6

    Heparan Sulfate (HS) mimetics are able to block crucial interactions of the components of the extracellular matrix in angiogenic processes and as such, represent a valuable class of original candidates for cancer therapy. Here we first report the synthesis and in vitro angiogenic inhibition properties of a conjugated, novel and rationally-designed octasaccharide-based HS mimetic. We also herein report its labeling with fluorine-18 and present the preliminary in vivo Positron Emission Tomography imaging data in rats. This constitutes one of the rare examples of labeling and in vivo evaluation of a synthetic, polysaccharide-based, macromolecule.

    Topics: Animals; Cell Proliferation; Dose-Response Relationship, Drug; Drug Screening Assays, Antitumor; Enzyme Inhibitors; Fluorine Radioisotopes; Glucuronidase; Heparitin Sulfate; Humans; Male; Molecular Structure; Neoplasms; Neovascularization, Pathologic; Polysaccharides; Positron-Emission Tomography; Rats; Rats, Wistar; Structure-Activity Relationship

2016
Cell surface syndecan-1 contributes to binding and function of macrophage migration inhibitory factor (MIF) on epithelial tumor cells.
    Biochimica et biophysica acta, 2016, Volume: 1863, Issue:4

    Surface expressed proteoglycans mediate the binding of cytokines and chemokines to the cell surface and promote migration of various tumor cell types including epithelial tumor cells. We here demonstrate that binding of the chemokine-like inflammatory cytokine macrophage migration inhibitory factor (MIF) to epithelial lung and breast tumor cell lines A549 and MDA-MB231 is sensitive to enzymatic digestion of heparan sulphate chains and competitive inhibition with heparin. Moreover, MIF interaction with heparin was confirmed by chromatography and a structural comparison indicated a possible heparin binding site. These results suggested that proteoglycans carrying heparan sulphate chains are involved in MIF binding. Using shRNA-mediated gene silencing, we identified syndecan-1 as the predominant proteoglycan required for the interaction with MIF. MIF binding was decreased by induction of proteolytic shedding of syndecan-1, which could be prevented by inhibition of the metalloproteinases involved in this process. Finally, MIF induced the chemotactic migration of A549 cells, wound closure and invasion into matrigel without affecting cell proliferation. These MIF-induced responses were abrogated by heparin or by silencing of syndecan-1. Thus, our study indicates that syndecan-1 on epithelial tumor cells promotes MIF binding and MIF-mediated cell migration. This may represent a relevant mechanism through which MIF enhances tumor cell motility and metastasis.

    Topics: Cell Adhesion; Cell Membrane; Cell Movement; Epithelial Cells; HEK293 Cells; Heparitin Sulfate; Humans; Intramolecular Oxidoreductases; Macrophage Migration-Inhibitory Factors; Neoplasm Metastasis; Neoplasms; Protein Binding; Syndecan-1; Tumor Cells, Cultured

2016
Glycan Sulfation Modulates Dendritic Cell Biology and Tumor Growth.
    Neoplasia (New York, N.Y.), 2016, Volume: 18, Issue:5

    In cancer, proteoglycans have been found to play roles in facilitating the actions of growth factors, and effecting matrix invasion and remodeling. However, little is known regarding the genetic and functional importance of glycan chains displayed by proteoglycans on dendritic cells (DCs) in cancer immunity. In lung carcinoma, among other solid tumors, tumor-associated DCs play largely subversive/suppressive roles, promoting tumor growth and progression. Herein, we show that targeting of DC glycan sulfation through mutation in the heparan sulfate biosynthetic enzyme N-deacetylase/N-sulfotransferase-1 (Ndst1) in mice increased DC maturation and inhibited trafficking of DCs to draining lymph nodes. Lymphatic-driven DC migration and chemokine (CCL21)-dependent activation of a major signaling pathway required for DC migration (as measured by phospho-Akt) were sensitive to Ndst1 mutation in DCs. Lewis lung carcinoma tumors in mice deficient in Ndst1 were reduced in size. Purified CD11c+ cells from the tumors, which contain the tumor-infiltrating DC population, showed a similar phenotype in mutant cells. These features were replicated in mice deficient in syndecan-4, the major heparan sulfate proteoglycan expressed on the DC surface: Tumors were growth-impaired in syndecan-4-deficient mice and were characterized by increased infiltration by mature DCs. Tumors on the mutant background also showed greater infiltration by NK cells and NKT cells. These findings indicate the genetic importance of DC heparan sulfate proteoglycans in tumor growth and may guide therapeutic development of novel strategies to target syndecan-4 and heparan sulfate in cancer.

    Topics: Animals; Cell Movement; Cell Proliferation; Chemokines; Dendritic Cells; Disease Models, Animal; Endothelial Cells; Heparitin Sulfate; Humans; Immunophenotyping; Mice; Mice, Transgenic; Mutation; Neoplasms; Phenotype; Proteoglycans; Syndecan-4; Tumor Burden

2016
Normal Wound Healing and Tumor Angiogenesis as a Game of Competitive Inhibition.
    PloS one, 2016, Volume: 11, Issue:12

    Both normal wound healing and tumor angiogenesis are mitigated by the sequential, carefully orchestrated release of growth stimulators and inhibitors. These regulators are released from platelet clots formed at the sites of activated endothelium in a temporally and spatially controlled manner, and the order of their release depends on their affinity to glycosaminoglycans (GAG) such as heparan sulfate (HS) within the extracellular matrix, and platelet open canallicular system. The formation of vessel sprouts, triggered by angiogenesis regulating factors with lowest affinities for heparan sulfate (e.g. VEGF), is followed by vessel-stabilizing PDGF-B or bFGF with medium affinity for HS, and by inhibitors such as PF-4 and TSP-1 with the highest affinities for HS. The invasive wound-like edge of growing tumors has an overabundance of angiogenesis stimulators, and we propose that their abundance out-competes angiogenesis inhibitors, effectively preventing inhibition of angiogenesis and vessel maturation. We evaluate this hypothesis using an experimentally motivated agent-based model, and propose a general theoretical framework for understanding mechanistic similarities and differences between the processes of normal wound healing and pathological angiogenesis from the point of view of competitive inhibition.

    Topics: Animals; Binding, Competitive; Blood Platelets; Fibroblast Growth Factor 2; Glycosaminoglycans; Heparitin Sulfate; Humans; Models, Biological; Neoplasms; Neovascularization, Pathologic; Neovascularization, Physiologic; Platelet Factor 4; Protein Binding; Proto-Oncogene Proteins c-sis; Thrombospondin 1; Tumor Microenvironment; Vascular Endothelial Growth Factor A; Wound Healing

2016
A Heparan Sulfate-Binding Cell Penetrating Peptide for Tumor Targeting and Migration Inhibition.
    BioMed research international, 2015, Volume: 2015

    As heparan sulfate proteoglycans (HSPGs) are known as co-receptors to interact with numerous growth factors and then modulate downstream biological activities, overexpression of HS/HSPG on cell surface acts as an increasingly reliable prognostic factor in tumor progression. Cell penetrating peptides (CPPs) are short-chain peptides developed as functionalized vectors for delivery approaches of impermeable agents. On cell surface negatively charged HS provides the initial attachment of basic CPPs by electrostatic interaction, leading to multiple cellular effects. Here a functional peptide (CPPecp) has been identified from critical HS binding region in hRNase3, a unique RNase family member with in vitro antitumor activity. In this study we analyze a set of HS-binding CPPs derived from natural proteins including CPPecp. In addition to cellular binding and internalization, CPPecp demonstrated multiple functions including strong binding activity to tumor cell surface with higher HS expression, significant inhibitory effects on cancer cell migration, and suppression of angiogenesis in vitro and in vivo. Moreover, different from conventional highly basic CPPs, CPPecp facilitated magnetic nanoparticle to selectively target tumor site in vivo. Therefore, CPPecp could engage its capacity to be developed as biomaterials for diagnostic imaging agent, therapeutic supplement, or functionalized vector for drug delivery.

    Topics: Animals; Cell Line, Tumor; Cell Membrane; Cell Movement; Cell-Penetrating Peptides; Eosinophil Cationic Protein; Heparitin Sulfate; Humans; Mice; Neoplasms; Xenograft Model Antitumor Assays

2015
Tissue-specificity of heparan sulfate biosynthetic machinery in cancer.
    Cell adhesion & migration, 2015, Volume: 9, Issue:6

    Heparan sulfate (HS) proteoglycans are key components of cell microenvironment and fine structure of their polysaccharide HS chains plays an important role in cell-cell interactions, adhesion, migration and signaling. It is formed on non-template basis, so, structure and functional activity of HS biosynthetic machinery is crucial for correct HS biosynthesis and post-synthetic modification. To reveal cancer-related changes in transcriptional pattern of HS biosynthetic system, the expression of HS metabolism-involved genes (EXT1/2, NDST1/2, GLCE, 3OST1/HS3ST1, SULF1/2, HPSE) in human normal (fibroblasts, PNT2) and cancer (MCF7, LNCaP, PC3, DU145, H157, H647, A549, U2020, U87, HT116, KRC/Y) cell lines and breast, prostate, colon tumors was studied. Real-time RT-PCR and Western-blot analyses revealed specific transcriptional patterns and expression levels of HS biosynthetic system both in different cell lines in vitro and cancers in vivo. Balance between transcriptional activities of elongation- and post-synthetic modification- involved genes was suggested as most informative parameter for HS biosynthetic machinery characterization. Normal human fibroblasts showed elongation-oriented HS biosynthesis, while PNT2 prostate epithelial cells had modification-oriented one. However, cancer epithelial cells demonstrated common tendency to acquire fibroblast-like elongation-oriented mode of HS biosynthetic system. Surprisingly, aggressive metastatic cancer cells (U2020, DU145, KRC/Y) retained modification-oriented HS biosynthesis similar to normal PNT2 cells, possibly enabling the cells to keep like-to-normal cell surface glycosylation pattern to escape antimetastatic control. The obtained results show the cell type-specific changes of HS-biosynthetic machinery in cancer cells in vitro and tissue-specific changes in different cancers in vivo, supporting a close involvement of HS biosynthetic system in carcinogenesis.

    Topics: Carcinogenesis; Cell Line, Tumor; Cellular Microenvironment; Fibroblasts; Gene Expression Regulation, Neoplastic; Heparitin Sulfate; Humans; Neoplasm Proteins; Neoplasms; Organ Specificity

2015
Heparin-protein interactions: from affinity and kinetics to biological roles. Application to an interaction network regulating angiogenesis.
    Matrix biology : journal of the International Society for Matrix Biology, 2014, Volume: 35

    Numerous extracellular proteins, growth factors, chemokines, cytokines, enzymes, lipoproteins, involved in a variety of biological processes, interact with heparin and/or heparan sulfate at the cell surface and in the extracellular matrix (ECM). The goal of this study is to investigate the relationship(s) between affinity and kinetics of heparin-protein interactions and the localization of the proteins, their intrinsic disorder and their biological roles. Most proteins bind to heparin with a higher affinity than their fragments and form more stable complexes with heparin than with heparan sulfate. Lipoproteins and matrisome-associated proteins (e.g. growth factors and cytokines) bind to heparin with very high affinity. Matrisome-associated proteins form transient complexes with heparin. However they bind to this glycosaminoglycan with a higher affinity than the proteins of the core matrisome, which contribute to ECM assembly and organization, and than the secreted proteins which are not associated with the ECM. The association rate of proteins with heparin is related to the intrinsic disorder of heparin-binding sites. Enzyme inhibitor activity, protein dimerization, skeletal system development and pathways in cancer are functionally associated with proteins displaying a high or very high affinity for heparin (KD<100 nM). Besides their use in investigating molecular recognition and functions, kinetics and affinity are essential to prioritize interactions in networks and to build network models as discussed for the interaction network established at the surface of endothelial cells by endostatin, a heparin-binding protein regulating angiogenesis.

    Topics: Animals; Binding Sites; Cytokines; Endostatins; Extracellular Matrix; Heparin; Heparitin Sulfate; Humans; Intercellular Signaling Peptides and Proteins; Kinetics; Lipoproteins; Macromolecular Substances; Neoplasms; Neovascularization, Pathologic; Protein Binding; Signal Transduction

2014
Human recombinant truncated RNASET2, devoid of RNase activity; A potential cancer therapeutic agent.
    Oncotarget, 2014, Nov-30, Volume: 5, Issue:22

    Human RNASET2 has been implicated in antitumorigenic and antiangiogenic activities, independent of its ribonuclease capacities. We constructed a truncated version of human RNASET2, starting at E50 (trT2-50) and devoid of ribonuclease activity. trT2-50 maintained its ability to bind actin and to inhibit angiogenesis and tumorigenesis. trT2-50 binds to cell surface actin and formed a complex with actin in vitro. The antiangiogenic effect of this protein was demonstrated in human umbilical vein endothelial cells (HUVECs) by its ability to arrest tube formation on Matrigel, induced by angiogenic factors. Immunofluorescence staining of HUVECs showed nuclear and cytosolic RNASET2 protein that was no longer detectable inside the cell following trT2-50 treatment. This effect was associated with disruption of the intracellular actin network. trT2-50 co-localized with angiogenin, suggesting that both molecules bind (or compete) for similar cellular epitopes. Moreover, trT2-50 led to a significant inhibition of tumor development. Histological analysis demonstrated abundant necrotic tissue and a substantial loss of endothelial structure in trT2-50-treated tumors. Collectively, the present results indicate that trT2-50, a molecule engineered to be deficient of its catalytic activity, still maintained its actin binding and anticancer-related biological activities. We therefore suggest that trT2-50 may serve as a potential cancer therapeutic agent.

    Topics: Actins; Amino Acid Sequence; Animals; Antineoplastic Agents; Carcinogenesis; Cell Line, Tumor; Cell Membrane; Cell Nucleus; Chromatography, Affinity; Cytosol; Epitopes; Female; Glycosylation; Heparitin Sulfate; Human Umbilical Vein Endothelial Cells; Humans; Mice; Mice, Nude; Molecular Sequence Data; Neoplasm Transplantation; Neoplasms; Neovascularization, Pathologic; Protein Denaturation; Protein Folding; Recombinant Proteins; Ribonuclease, Pancreatic; Ribonucleases; Tumor Suppressor Proteins

2014
Heparanase regulates secretion, composition, and function of tumor cell-derived exosomes.
    The Journal of biological chemistry, 2013, Apr-05, Volume: 288, Issue:14

    Emerging evidence indicates that exosomes play a key role in tumor-host cross-talk and that exosome secretion, composition, and functional capacity are altered as tumors progress to an aggressive phenotype. However, little is known regarding the mechanisms that regulate these changes. Heparanase is an enzyme whose expression is up-regulated as tumors become more aggressive and is associated with enhanced tumor growth, angiogenesis, and metastasis. We have discovered that in human cancer cells (myeloma, lymphoblastoid, and breast cancer), when expression of heparanase is enhanced or when tumor cells are exposed to exogenous heparanase, exosome secretion is dramatically increased. Heparanase enzyme activity is required for robust enhancement of exosome secretion because enzymatically inactive forms of heparanase, even when present in high amounts, do not dramatically increase exosome secretion. Heparanase also impacts exosome protein cargo as reflected by higher levels of syndecan-1, VEGF, and hepatocyte growth factor in exosomes secreted by heparanase-high expressing cells as compared with heparanase-low expressing cells. In functional assays, exosomes from heparanase-high cells stimulated spreading of tumor cells on fibronectin and invasion of endothelial cells through extracellular matrix better than did exosomes secreted by heparanase-low cells. These studies reveal that heparanase helps drive exosome secretion, alters exosome composition, and facilitates production of exosomes that impact both tumor and host cell behavior, thereby promoting tumor progression.

    Topics: Cell Line, Tumor; Disease Progression; DNA, Complementary; Exosomes; Gene Expression Regulation, Enzymologic; Gene Expression Regulation, Neoplastic; Glucuronidase; Heparitin Sulfate; Hepatocyte Growth Factor; Humans; Multiple Myeloma; Neoplasms; Neovascularization, Pathologic; Syndecan-1; Vascular Endothelial Growth Factor A

2013
Tumor targeting by a carbohydrate ligand-mimicking peptide.
    Methods in molecular biology (Clifton, N.J.), 2013, Volume: 1022

    Annexin A1 (Anxa1) is a highly specific surface marker of tumor vasculature. We used peptide-displaying phage technology to identify a carbohydrate ligand-mimicking 7-mer peptide, IFLLWQR (IF7), which can target Anxa1 in tumor vasculature. Here, we describe the binding activity of carbohydrate to Anxa1, Anxa1 to heparan sulfates, and the therapeutic potential of IF7 conjugated with anticancer drugs in tumor targeting.

    Topics: Amino Acid Sequence; Animals; Annexin A1; Antineoplastic Agents; Carbohydrate Metabolism; Carbohydrates; Cell Line, Tumor; Drug Delivery Systems; Female; Fluorescent Dyes; Heparitin Sulfate; Humans; Mice; Neoplasms; Peptides; Protein Binding

2013
Improved systemic antitumor therapy with oncolytic adenoviruses by replacing the fiber shaft HSG-binding domain with RGD.
    Gene therapy, 2012, Volume: 19, Issue:4

    Retargeting oncolytic adenoviruses from their systemic preeminent liver tropism to disseminated tumor foci would highly improve the efficacy of these agents at eradicating tumors. We have replaced the KKTK fiber shaft heparan sulfate glycosaminoglycan-binding domain with an RGDK motif in order to achieve simultaneously liver detargeting and tumor targeting. When inserted into a wild-type backbone, this mutation palliated liver transaminase elevation and hematological alterations in mice. Importantly, when tested in a backbone that redirects E1A transcription towards pRB pathway deregulation, RGD at this novel shaft location also improved significantly systemic antitumor therapy compared with the broadly used RGD location at the HI-loop of the fiber knob domain.

    Topics: Adenoviridae; Animals; Binding Sites; Cell Line, Tumor; Gene Transfer Techniques; Genetic Vectors; Heparitin Sulfate; Mice; Mice, Inbred BALB C; Neoplasms; Oligopeptides; Oncolytic Virotherapy; Receptors, Cell Surface

2012
Fibroblast EXT1-levels influence tumor cell proliferation and migration in composite spheroids.
    PloS one, 2012, Volume: 7, Issue:7

    Stromal fibroblasts are important determinants of tumor cell behavior. They act to condition the tumor microenvironment, influence tumor growth, support tumor angiogenesis and affect tumor metastasis. Heparan sulfate proteoglycans, present both on tumor and stromal cells, interact with a large number of ligands including growth factors, their receptors, and structural components of the extracellular matrix. Being ubiquitously expressed in the tumor microenvironment heparan sulfate proteoglycans are candidates for playing central roles in tumor-stroma interactions. The objective of this work was to investigate the role of heparan sulfate expressed by stromal fibroblasts in modulating the growth of tumor cells and in controlling the interstitial fluid pressure in a 3-D model.. We generated spheroids composed of fibroblasts alone, or composite spheroids, composed of fibroblasts and tumor cells. Here we show that stromal fibroblasts with a mutation in the heparan sulfate elongating enzyme Ext1 and thus a low heparan sulfate content, formed composite fibroblast/tumor cell spheroids with a significant lower interstitial fluid pressure than corresponding wild-type fibroblast/tumor cell composite spheroids. Furthermore, immunohistochemistry of composite spheroids revealed that the cells segregated, so that after 6 days in culture, the wild-type fibroblasts formed an inner core and the tumor cells an outer layer of cells. For composite spheroids containing Ext1-mutated fibroblasts this segregation was less obvious, indicating impaired cell migration. Analysis of tumor cells expressing the firefly luciferase gene revealed that the changes in tumor cell migration in mutant fibroblast/tumor cell composite spheroids coincided with a lower proliferation rate.. This is the first demonstration that stromal Ext1-levels modulate tumor cell proliferation and affect the interstitial fluid pressure in a 3-D spheroid model. Learning how structural changes in stromal heparan sulfate influence tumor cells is essential for our understanding how non-malignant cells of the tumor microenvironment influence tumor cell progression.

    Topics: Animals; Cell Line, Transformed; Cell Line, Tumor; Cell Movement; Cell Proliferation; Coculture Techniques; Fibroblasts; Heparitin Sulfate; Mice; N-Acetylglucosaminyltransferases; Neoplasms; Spheroids, Cellular; Stromal Cells; Tumor Microenvironment

2012
Targeted disruption of heparan sulfate interaction with hepatocyte and vascular endothelial growth factors blocks normal and oncogenic signaling.
    Cancer cell, 2012, Aug-14, Volume: 22, Issue:2

    Hepatocyte growth factor (HGF) and vascular endothelial cell growth factor (VEGF) regulate normal development and homeostasis and drive disease progression in many forms of cancer. Both proteins signal by binding to receptor tyrosine kinases and heparan sulfate (HS) proteoglycans on target cell surfaces. Basic residues comprising the primary HS binding sites on HGF and VEGF provide similar surface charge distributions without underlying structural similarity. Combining three acidic amino acid substitutions in these sites in the HGF isoform NK1 or the VEGF isoform VEGF165 transformed each into potent, selective competitive antagonists of their respective normal and oncogenic signaling pathways. Our findings illustrate the importance of HS in growth factor driven cancer progression and reveal an efficient strategy for therapeutic antagonist development.

    Topics: Animals; Antigens, CD34; Cell Proliferation; Cluster Analysis; Dogs; Enzyme Activation; Gene Targeting; Heparitin Sulfate; Hepatocyte Growth Factor; Humans; Magnetic Resonance Spectroscopy; Mice; Neoplasm Metastasis; Neoplasms; Neovascularization, Pathologic; Protein Binding; Protein Folding; Protein Isoforms; Protein Multimerization; Proto-Oncogene Proteins c-met; Signal Transduction; Vascular Endothelial Growth Factor A

2012
Heparan sulfate regulates VEGF165- and VEGF121-mediated vascular hyperpermeability.
    The Journal of biological chemistry, 2011, Jan-07, Volume: 286, Issue:1

    VEGF was first described as vascular permeability factor, a potent inducer of vascular leakage. Genetic evidence indicates that VEGF-stimulated endothelial proliferation in vitro and angiogenesis in vivo depend on heparan sulfate, but a requirement for heparan sulfate in vascular hyperpermeability has not been explored. Here we show that altering endothelial cell heparan sulfate biosynthesis in vivo decreases hyperpermeability induced by both VEGF(165) and VEGF(121). Because VEGF(121) does not bind heparan sulfate, the requirement for heparan sulfate suggested that it interacted with VEGF receptors rather than the ligand. By applying proximity ligation assays to primary brain endothelial cells, we show a direct interaction in situ between heparan sulfate and the VEGF receptor, VEGFR2. Furthermore, the number of heparan sulfate-VEGFR2 complexes increased in response to both VEGF(165) and VEGF(121). Genetic or heparin lyase-mediated alteration of endothelial heparan sulfate attenuated phosphorylation of VEGFR2 in response to VEGF(165) and VEGF(121), suggesting that the functional VEGF receptor complex contains heparan sulfate. Pharmacological blockade of heparan sulfate-protein interactions inhibited hyperpermeability in vivo, suggesting heparan sulfate as a potential target for treating hyperpermeability associated with ischemic disease.

    Topics: Animals; Blood Vessels; Endothelial Cells; Heparitin Sulfate; Humans; Mice; Neoplasms; Permeability; Phosphorylation; Skin; Sulfotransferases; Urea; Vascular Endothelial Growth Factor A; Vascular Endothelial Growth Factor Receptor-2

2011
Small lytic peptides escape the inhibitory effect of heparan sulfate on the surface of cancer cells.
    BMC cancer, 2011, Mar-31, Volume: 11

    Several naturally occurring cationic antimicrobial peptides (CAPs), including bovine lactoferricin (LfcinB), display promising anticancer activities. These peptides are unaffected by multidrug resistance mechanisms and have been shown to induce a protective immune response against solid tumors, thus making them interesting candidates for developing novel lead structures for anticancer treatment. Recently, we showed that the anticancer activity by LfcinB was inhibited by the presence of heparan sulfate (HS) on the surface of tumor cells. Based on extensive structure-activity relationship studies performed on LfcinB, shorter and more potent peptides have been constructed. In the present study, we have investigated the anticancer activity of three chemically modified 9-mer peptides and the influence of HS and chondroitin sulfate (CS) on their cytotoxic activity.. Various cell lines and red blood cells were used to investigate the anticancer activity and selectivity of the peptides. The cytotoxic effect of the peptides against the different cell lines was measured by use of a colorimetric MTT viability assay. The influence of HS and CS on their cytotoxic activity was evaluated by using HS/CS expressing and HS/CS deficient cell lines. The ability of soluble HS and CS to inhibit the cytotoxic activity of the peptides and the peptides' affinity for HS and CS were also investigated.. The 9-mer peptides displayed selective anticancer activity. Cells expressing HS/CS were equally or more susceptible to the peptides than cells not expressing HS/CS. The peptides displayed a higher affinity for HS compared to CS, and exogenously added HS inhibited the cytotoxic effect of the peptides.. In contrast to the previously reported inhibitory effect of HS on LfcinB, the present study shows that the cytotoxic activity of small lytic peptides was increased or not affected by cell surface HS.

    Topics: Animals; Antimicrobial Cationic Peptides; Apoptosis; Cattle; Cell Line; Chondroitin Sulfates; Erythrocytes; Hemolysis; Heparitin Sulfate; Lactoferrin; Neoplasms; Peptide Fragments; Protein Engineering; Structure-Activity Relationship

2011
Synthesis of simple heparanase substrates.
    Organic & biomolecular chemistry, 2011, Jun-21, Volume: 9, Issue:12

    Heparanase degrades heparan sulfate (HS) chains on proteoglycans; elevated levels of heparanase expression correlate with tumour cell metastatic potential and vascularity, and reduced post-operative survival of cancer patients. Consequently, heparanase expression is considered a biomarker for cancer detection. Although several heparanase assays have been developed, most require the preparation of heterogeneous, (radio)labelled HS substrates and rely on the separation of enzymatically-degraded products on the basis of molecular size. In studies directed towards the development of a more direct heparanase assay, a series of glucuronides and glycosyl glucuronides were synthesised as putative heparanase substrates. These compounds were designed with various aryl aglycones that could be measured spectrophotometrically upon hydrolysis of the glycosidic linkage by heparanase. It was found that the N-sulfated 4-nitrophenyl glycosyl glucuronide 24 and the N-sulfated methylumbelliferyl glycosyl glucuronide 26 were hydrolysed by recombinant human heparanase. These compounds represent the simplest substrates of heparanase reported to date.

    Topics: Biomarkers, Tumor; Carbohydrate Conformation; Chromatography, Thin Layer; Drug Design; Glucuronidase; Glucuronides; Glycosylation; Heparitin Sulfate; Humans; Magnetic Resonance Spectroscopy; Molecular Sequence Data; Neoplasm Metastasis; Neoplasms; Proteoglycans; Recombinant Proteins; Solutions; Substrate Specificity; Tumor Cells, Cultured

2011
The PG500 series: novel heparan sulfate mimetics as potent angiogenesis and heparanase inhibitors for cancer therapy.
    Investigational new drugs, 2010, Volume: 28, Issue:3

    Heparan sulfate mimetics, which we have called the PG500 series, have been developed to target the inhibition of both angiogenesis and heparanase activity. This series extends the technology underpinning PI-88, a mixture of highly sulfated oligosaccharides which reached Phase III clinical development for hepatocellular carcinoma. Advances in the chemistry of the PG500 series provide numerous advantages over PI-88. These new compounds are fully sulfated, single entity oligosaccharides attached to a lipophilic moiety, which have been optimized for drug development. The rational design of these compounds has led to vast improvements in potency compared to PI-88, based on in vitro angiogenesis assays and in vivo tumor models. Based on these and other data, PG545 has been selected as the lead clinical candidate for oncology and is currently undergoing formal preclinical development as a novel treatment for advanced cancer.

    Topics: Anticoagulants; Antineoplastic Agents; Cell Line; Cell Line, Tumor; Cell Proliferation; Drug Discovery; Drug Evaluation, Preclinical; Fibroblast Growth Factor 1; Fibroblast Growth Factor 2; Glucuronidase; Heparitin Sulfate; Humans; Neoplasms; Neovascularization, Pathologic; Structure-Activity Relationship; Vascular Endothelial Growth Factor A

2010
The heparan sulfate motif (GlcNS6S-IdoA2S)3, common in heparin, has a strict topography and is involved in cell behavior and disease.
    The Journal of biological chemistry, 2010, Dec-24, Volume: 285, Issue:52

    Heparan sulfate (HS) is a structurally complex polysaccharide that interacts with a broad spectrum of extracellular effector ligands and thereby is thought to regulate a diverse array of biologic processes. The specificity of HS-ligand interactions is determined by the arrangement of sulfate groups on HS, which creates distinct binding motifs. Biologically important HS motifs are expected to exhibit regulated expression, yet there is a profound lack of tools to identify such motifs; consequently, little is known of their structures and functions. We have identified a novel phage display-derived antibody (NS4F5) that recognizes a highly regulated HS motif (HS(NS4F5)), which we have rigorously identified as (GlcNS6S-IdoA2S)(3). HS(NS4F5) exhibits a restricted expression in healthy adult tissues. Blocking HS(NS4F5) on cells in culture resulted in reduced proliferation and enhanced sensitivity to apoptosis. HS(NS4F5) is up-regulated in tumor endothelial cells, consistent with a role in endothelial cell activation. Indeed, TNF-α stimulated endothelial expression of HS(NS4F5), which contributed to leukocyte adhesion. In a mouse model of severe systemic amyloid protein A amyloidosis, HS(NS4F5) was expressed within amyloid deposits, which were successfully detected by microSPECT imaging using NS4F5 as a molecularly targeted probe. Combined, our results demonstrate that NS4F5 is a powerful tool for elucidating the biological function of HS(NS4F5) and can be exploited as a probe to detect novel polysaccharide biomarkers of disease processes.

    Topics: Amyloidogenic Proteins; Amyloidosis; Animals; Antibodies, Monoclonal; Biomarkers; Carbohydrate Sequence; Cell Proliferation; CHO Cells; Cricetinae; Cricetulus; Disease Models, Animal; Endothelial Cells; Female; Heparitin Sulfate; Humans; Male; Mice; Neoplasms; Rats; Rats, Wistar; Single-Chain Antibodies; Tumor Necrosis Factor-alpha

2010
The anticancer activity of lytic peptides is inhibited by heparan sulfate on the surface of the tumor cells.
    BMC cancer, 2009, Jun-15, Volume: 9

    Cationic antimicrobial peptides (CAPs) with antitumor activity constitute a promising group of novel anticancer agents. These peptides induce lysis of cancer cells through interactions with the plasma membrane. It is not known which cancer cell membrane components influence their susceptibility to CAPs. We have previously shown that CAPs interact with the two glycosaminoglycans (GAGs), heparan sulfate (HS) and chondroitin sulfate (CS), which are present on the surface of most cells. The purpose of this study was to investigate the role of the two GAGs in the cytotoxic activity of CAPs.. Various cell lines, expressing different levels of cell surface GAGs, were exposed to bovine lactoferricin (LfcinB) and the designer peptide, KW5. The cytotoxic effect of the peptides was investigated by use of the colorimetric MTT viability assay. The cytotoxic effect on wild type CHO cells, expressing normal amounts of GAGs on the cell surface, and the mutant pgsA-745, that has no expression of GAGs on the cell surface, was also investigated.. We show that cells not expressing HS were more susceptible to CAPs than cells expressing HS at the cell surface. Further, exogenously added heparin inhibited the cytotoxic effect of the peptides. Chondroitin sulfate had no effect on the cytotoxic activity of KW5 and only minor effects on LfcinB cytotoxicity.. Our results show for the first time that negatively charged molecules at the surface of cancer cells inhibit the cytotoxic activity of CAPs. Our results indicate that HS at the surface of cancer cells sequesters CAPs away from the phospholipid bilayer and thereby impede their ability to induce cytolysis.

    Topics: Animals; Cattle; Cell Line, Tumor; Chlorates; CHO Cells; Cricetinae; Cricetulus; Drug Synergism; Heparin; Heparitin Sulfate; HT29 Cells; Humans; Lactoferrin; Lymphoma; Neoplasms; Peptide Fragments; Protein Structure, Secondary

2009
The role of glypican-3 in the regulation of body size and cancer.
    Cell cycle (Georgetown, Tex.), 2008, Sep-15, Volume: 7, Issue:18

    Glypicans are a family of heparan sulfate proteoglycans whose members are bound to the cell surface by a glycosylphosphatidylinositol (GPI) anchor. Loss-of-function mutations in GPC3, one of the six mammalian glypicans, causes the Simson-Golabi-Behmel Syndrome. This is a disorder characterized by pre- and post-natal overgrowth, a broad spectrum of visceral and skeletal abnormalities, and an increased risk for the development of embryonic tumors. GPC3-null mice also display significant overgrowth. We have recently reported that GPC3 acts as a negative regulator of Hedgehog signaling during development, and that the overgrowth caused by the lack of functional GPC3 is due, at least in part, to the hyperactivation of Hedgehog signaling. Here we discuss the rationale that led us to hypothesize that GPC3 could be a negative regulator of Hedgehog signaling, and speculate about the implications of our discovery regarding the role of GPC3 in some cancer types. We also discuss our recent results of experiments that investigated the role of the core protein, the heparan sulfate chains, and the GPI anchor in GPC3 function. Finally, we propose an explanation for the tissue-specific function of GPC3.

    Topics: Abnormalities, Multiple; Animals; Body Size; Glycosylphosphatidylinositols; Glypicans; Hedgehog Proteins; Heparitin Sulfate; Mice; Neoplasms; Signal Transduction; Syndrome; Wnt Proteins

2008
A simple assay to probe disease-associated enzyme activity using glycosaminoglycan-assisted synthesized gold nanoparticles.
    Organic & biomolecular chemistry, 2008, Dec-07, Volume: 6, Issue:23

    A simple assay to probe disease-associated enzyme activity using glycosaminoglycan-assisted synthesized gold nanoparticles is reported.

    Topics: Biosensing Techniques; Glycosaminoglycans; Gold; Heparitin Sulfate; Humans; Metal Nanoparticles; Neoplasms; Polysaccharide-Lyases

2008
Heparanase induces tissue factor pathway inhibitor expression and extracellular accumulation in endothelial and tumor cells.
    Thrombosis and haemostasis, 2008, Volume: 99, Issue:1

    Heparanase activity is implicated in cell invasion, tumor metastasis and angiogenesis. Recently, we have reported that heparanase stimulates tissue factor (TF) expression in endothelial and cancer cells, resulting in elevation of coagulation activity. We hypothesized that heparanase regulates other coagulation modulators, and examined the expression and localization of tissue factor pathway inhibitor (TFPI) following heparanase over-expression or exogenous addition. Primary human umbilical vein endothelial cells (HUVEC) and human tumor-derived cell lines were incubated with heparanase, or were stably transfected with heparanase gene-constructs, and TFPI expression and secretion were examined. Heparanase over-expression or exogenous addition stimulated TFPI expression by 2-3 folds. TFPI accumulation in the cell culture medium exceeded in magnitude the observed induction of TFPI gene transcription reaching 5- to 6-fold increase. Extracellular accumulation of TFPI was evident already 60 min following heparanase addition, prior to TFPI protein induction, and correlated with increased coagulation activity. This effect was found to be independent of heparanase enzymatic activity and interaction with heparan-sulfate, and correlated with reduced TFPI levels on the cell surface. Data were verified in heparanase transgenic mice tissues and plasma. Interaction between heparanase and TFPI was evident by co-immunoprecipitation. Interaction of heparanase with TFPI resulted in its displacement from the surface of the vascular endothelium and in increased pro-coagulant activity. Thus, heparanase facilitates blood coagulation on the cell surface by two independent mechanisms: dissociation of TFPI from the vascular surface shortly after local elevation of heparanase levels, and subsequent induction of TF expression.

    Topics: Animals; Blood Coagulation; Cell Line, Tumor; Cell Membrane; Cells, Cultured; Endothelial Cells; Glucuronidase; Heparitin Sulfate; Humans; Lipoproteins; Mice; Mice, Transgenic; Neoplasms; Protein Binding; Recombinant Proteins; RNA, Messenger; Thromboplastin; Time Factors; Transcription, Genetic; Transfection; Up-Regulation

2008
Transgenic or tumor-induced expression of heparanase upregulates sulfation of heparan sulfate.
    Nature chemical biology, 2007, Volume: 3, Issue:12

    Heparan sulfate proteoglycans (HSPGs) interact with numerous proteins of importance in animal development and homeostasis. Heparanase, which is expressed in normal tissues and upregulated in angiogenesis, cancer and inflammation, selectively cleaves beta-glucuronidic linkages in HS chains. In a previous study, we transgenically overexpressed heparanase in mice to assess the overall effects of heparanase on HS metabolism. Metabolic labeling confirmed extensive fragmentation of HS in vivo. In the current study we found that in liver showing excessive heparanase overexpression, HSPG turnover is accelerated along with upregulation of HS N- and O-sulfation, thus yielding heparin-like chains without the domain structure typical of HS. Heparanase overexpression in other mouse organs and in human tumors correlated with increased 6-O-sulfation of HS, whereas the domain structure was conserved. The heavily sulfated HS fragments strongly promoted formation of ternary complexes with fibroblast growth factor 1 (FGF1) or FGF2 and FGF receptor 1. Heparanase thus contributes to regulation of HS biosynthesis in a way that may promote growth factor action in tumor angiogenesis and metastasis.

    Topics: Animals; Carbohydrate Conformation; Carbohydrate Sequence; Gene Expression Regulation, Neoplastic; Glucuronidase; Heparitin Sulfate; Humans; Liver; Mice; Mice, Transgenic; Neoplasms; Sulfur; Up-Regulation

2007
HSulf-1 and HSulf-2 are potent inhibitors of myeloma tumor growth in vivo.
    The Journal of biological chemistry, 2005, Dec-02, Volume: 280, Issue:48

    To participate as co-receptor in growth factor signaling, heparan sulfate must have specific structural features. Recent studies show that when the levels of 6-O-sulfation of heparan sulfate are diminished by the activity of extracellular heparan sulfate 6-O-endosulfatases (Sulfs), fibroblast growth factor 2-, heparin binding epidermal growth factor-, and hepatocyte growth factor-mediated signaling are attenuated. This represents a novel mechanism for regulating cell growth, particularly within the tumor microenvironment where the Sulfs are known to be misregulated. To directly test the role of Sulfs in tumor growth control in vivo, a human myeloma cell line was transfected with cDNAs encoding either of the two known human endosulfatases, HSulf-1 or HSulf-2. When implanted into severe combined immunodeficient (SCID) mice, the growth of these tumors was dramatically reduced on the order of 5- to 10-fold as compared with controls. In addition to an inhibition of tumor growth, these studies revealed the following. (i) HSulf-1 and HSulf-2 have similar functions in vivo. (ii) The extracellular activity of Sulfs is restricted to the local tumor cell surface. (iii) The Sulfs promote a marked increase in extracellular matrix deposition within tumors that may, along with attenuated growth factor signaling, contribute to the reduction in tumor growth. These findings demonstrate that dynamic regulation of heparan sulfate structure by Sulfs present within the tumor microenvironment can have a dramatic impact on the growth and progression of malignant cells in vivo.

    Topics: Animals; Blotting, Western; Cell Line, Tumor; Cell Proliferation; Disaccharides; Disease Progression; DNA, Complementary; Extracellular Matrix; Fibroblast Growth Factor 2; Flow Cytometry; Growth Substances; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Immunohistochemistry; Mice; Mice, SCID; Multiple Myeloma; Neoplasm Transplantation; Neoplasms; Polysaccharide-Lyases; Protein Binding; Signal Transduction; Sulfatases; Sulfotransferases; Time Factors; Transfection

2005
Growth factor-induced shedding of syndecan-1 confers glypican-1 dependence on mitogenic responses of cancer cells.
    The Journal of cell biology, 2005, Nov-21, Volume: 171, Issue:4

    The cell surface heparan sulfate proteoglycan (HSPG) glypican-1 is up-regulated by pancreatic and breast cancer cells, and its removal renders such cells insensitive to many growth factors. We sought to explain why the cell surface HSPG syndecan-1, which is also up-regulated by these cells and is a known growth factor coreceptor, does not compensate for glypican-1 loss. We show that the initial responses of these cells to the growth factor FGF2 are not glypican dependent, but they become so over time as FGF2 induces shedding of syndecan-1. Manipulations that retain syndecan-1 on the cell surface make long-term FGF2 responses glypican independent, whereas those that trigger syndecan-1 shedding make initial FGF2 responses glypican dependent. We further show that syndecan-1 shedding is mediated by matrix metalloproteinase-7 (MMP7), which, being anchored to cells by HSPGs, also causes its own release in a complex with syndecan-1 ectodomains. These results support a specific role for shed syndecan-1 or MMP7-syndecan-1 complexes in tumor progression and add to accumulating evidence that syndecans and glypicans have nonequivalent functions in vivo.

    Topics: Cell Line, Tumor; Cell Membrane; Culture Media, Serum-Free; Disease Progression; DNA; Enzyme Activation; Enzyme Inhibitors; Fibroblast Growth Factor 2; Glycosaminoglycans; Growth Substances; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; MAP Kinase Signaling System; Matrix Metalloproteinase 7; Membrane Glycoproteins; Neoplasms; Protein Structure, Tertiary; Proteoglycans; Syndecan-1; Syndecans; Time Factors; Transfection; Trypsin; Up-Regulation

2005
Risk of thrombosis in patients with malignancy and heparin-induced thrombocytopenia.
    American journal of hematology, 2004, Volume: 76, Issue:3

    Heparin-induced thrombocytopenia (HIT) is a common immunological drug reaction. After exposure to heparin, some patients develop heparin dependent antibodies with no evidence of thrombosis, while others are at risk of thrombocytopenia, thrombosis, limb loss, and death. We conducted a retrospective chart review on all patients serologically positive for HIT by HPIA ELISA in a single tertiary-care hospital, to determine whether patients with malignancy had an increased risk of thrombotic complications. Medical records of 55 patients who tested positive for HIT and met clinical criteria for HIT were analyzed. All patients had been treated with unfractionated heparin. Malignancy was diagnosed in 11 patients, either at surgery or post-mortem examination. A higher rate of venous thrombosis and pulmonary embolism was observed in patients with HIT and malignant disease when compared to patients with no underlying malignancy (odds ratio 13.6, 95% CI 2.9-63.8).

    Topics: Aged; Aged, 80 and over; Anticoagulants; Chondroitin Sulfates; Dermatan Sulfate; Drug Combinations; Female; Heparin; Heparitin Sulfate; Humans; Male; Middle Aged; Neoplasms; Pulmonary Embolism; Retrospective Studies; Risk Factors; Thrombocytopenia; Thrombosis; Venous Thrombosis

2004
Ultrafiltration-based assay for heparanase activity.
    Analytical biochemistry, 2004, Aug-01, Volume: 331, Issue:1

    Heparanase, a mammalian endoglycosidase that specifically cleaves heparan sulfate (HS), has been found in many tissues. Platelet, liver, and placenta have been abundant sources for the study of the enzyme. Notably, certain malignant cells also have been found to produce large amounts of the enzyme, the levels of which often correlate with their invasive and metastatic properties. To study roles of heparanase in various biological situations, a reliable method measuring the enzyme activity is indispensable. In the past, measurement of heparanase enzyme activity was done either by the detection of the degradation of fluorescent or radiolabeled HS chains by gel filtration procedures or by the use of radiolabeled substrate conjugated to solid matrices for the easy separation of degraded HS chains. A newly developed procedure, presented in this article, measures degradation of radiolabeled HS chains in the aqueous buffer by detecting their degradation products using an ultrafiltration device, the Centricon 30. This procedure has several advantages over previous assay procedures that involved tedious processing such as gel filtration chromatography of each sample or the preparation of substrate HS proteoglycans conjugated to a solid matrix. The simplicity of the new procedure allows a short setup time and a rapid processing of a large number of samples. Furthermore, the enzymatic reaction during the aqueous phase allows kinetic analyses in standard conditions.

    Topics: Animals; Glucuronidase; Heparitin Sulfate; Kinetics; Neoplasm Metastasis; Neoplasms; Rats; Ultrafiltration

2004
Exacerbation of antiphospholipid antibody syndrome after treatment of localized cancer: a report of two cases.
    Annals of hematology, 2002, Volume: 81, Issue:12

    Patients with malignancy often present with a variety of coagulation abnormalities which may ultimately lead to recurrent arterial and venous thromboses. Recently the presence of antiphospholipid antibodies in cancer patients has been proposed as one of the potential mechanisms promoting hypercoagulability. Here we report two consecutive patients with localized tumors, one suffering from breast cancer and another presenting with colorectal cancer, who experienced dramatic exacerbation of the antiphospholipid antibody syndrome (APAS) within 4 weeks after surgery. In the first patient who had also received one course of adjuvant chemotherapy, major ischemic stroke and recurrent venous thromboembolism were paralleled by the development of ulcerative livedoid vasculitis and pancytopenia, constituting the diagnosis of systemic lupus erythematosus with secondary APAS. In the second patient, progressive thrombotic occlusion of the superior and inferior vena cava was associated with bilateral pulmonary embolism, acute renal failure, and disabling soft tissue edema. Although not fulfilling the classic criteria of "catastrophic" APAS, the clinical features were life threatening and appeared to be refractory to oral anticoagulation with phenprocoumon. In addition, a diagnosis of Trousseau's syndrome was unlikely due to missing evidence of gross metastatic disease. Besides a suggested treatment strategy comprising high doses of low-molecular-weight heparin, potential pathogenic mechanisms are discussed in consideration of a recently proposed "thrombotic storm," which may cause multiple thromboses after an initial provocation in patients with known hypercoagulability.

    Topics: Adult; Antiphospholipid Syndrome; Breast Neoplasms; Chemotherapy, Adjuvant; Chondroitin Sulfates; Colorectal Neoplasms; Dermatan Sulfate; Drug Combinations; Female; Heparin, Low-Molecular-Weight; Heparitin Sulfate; Humans; Lupus Erythematosus, Systemic; Middle Aged; Neoplasms; Stroke; Thromboembolism; Thrombophilia

2002
The Belgian Society for Cell Biology and NFWO Research Community on Cell-Matrix Interactions. Heparan sulfate proteoglycans in development and malignancy. Ghent, Belgium, 7-8 1996. Proceedings and abstracts.
    Cell biology international, 1997, Volume: 21, Issue:1

    Topics: Animals; Cell Adhesion; Cell Communication; Extracellular Matrix; Heparitin Sulfate; Humans; Neoplasm Invasiveness; Neoplasm Metastasis; Neoplasms; Proteoglycans; Signal Transduction

1997
A rapid quantitative assay for the detection of mammalian heparanase activity.
    The Biochemical journal, 1997, Jul-01, Volume: 325 ( Pt 1)

    Heparan sulphate (HS) is an important component of the extracellular matrix and the vasculature basal laminar which functions as a barrier to the extravasation of metastatic and inflammatory cells. Cleavage of HS by endoglycosidase or heparanase activity produced by invading cells may assist in the disassembly of the extracellular matrix and basal laminar, and thereby facilitate cell migration. Heparanase activity has previously been shown to be related to the metastatic potential of murine and human melanoma cell lines [Nakajima, Irimura and Nicolson (1988) J. Cell. Biochem. 36, 157-167]. To determine heparanase activity, porcine mucosal HS was partially de-N-acetylated and re-N-acetylated with [3H]acetic anhydride to yield a radiolabelled substrate. This procedure prevented the masking of, or possible formation of, new heparanase-sensitive cleavage sites as has been observed with previous methods of radiolabelling. Heparanase activity in a variety of tissues and cell homogenates including human platelets, colonic carcinoma cells, umbilical vein endothelial cells and rat mammary adenocarcinoma cells (both metastatic and non-metastatic variants) and liver homogenates all degraded the substrate in a stepwise fashion from 18.5 to approximately 13, 8 and finally to 4.5 kDa fragments, as assessed by gel-filtration analysis, confirming the substrate as suitable for the detection of heparanase activity present in a variety of cells and tissues. A rapid quantitative assay was developed with the HS substrate using a novel method for separating degradation products from the substrate by taking advantage of the decreased affinity of the heparanase-cleaved products for the HS-binding plasma protein chicken histidine-rich glycoprotein (cHRG). Incubation mixtures were applied to cHRG-Sepharose columns, with unbound material corresponding to heparanase-degradation products. Heparanase activity was determined for a variety of human, rat and murine cell and tissue homogenates. The highly metastatic rat mammary adenocarcinoma and murine lung carcinoma cell lines had four to ten times the heparanase activity of non-metastatic variants, confirming the correlation of heparanase activity with metastatic potential. Human cancer patients had twice the serum heparanase levels of normal healthy adults. The assay will be valuable for the determination of heparanase activity from a variety of tissue and cell sources, as a diagnostic tool for the determination of heparanase potentia

    Topics: Adult; Animals; Cell Line; Chickens; Female; Glucuronidase; Glycoside Hydrolases; Heparitin Sulfate; Humans; Intestinal Mucosa; Kinetics; Mice; Neoplasm Metastasis; Neoplasms; Proteins; Rats; Reference Values; Substrate Specificity; Tumor Cells, Cultured

1997
Perlecan: the multidomain heparan sulphate proteoglycan of basement membrane and extracellular matrix.
    Virchows Archiv. A, Pathological anatomy and histopathology, 1993, Volume: 423, Issue:4

    Topics: Alzheimer Disease; Basement Membrane; Diabetes Mellitus; Extracellular Matrix; Forecasting; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Humans; Molecular Structure; Neoplasms; Proteoglycans; Structure-Activity Relationship

1993
Venous occlusion test for the release of heparan sulfate from endothelium.
    Thrombosis research, 1993, Nov-15, Volume: 72, Issue:4

    Topics: Adult; Constriction; Diabetes Mellitus, Type 1; Endothelium, Vascular; Female; Heparitin Sulfate; Humans; Ischemic Attack, Transient; Male; Neoplasms; Veins

1993
Anomalous structure of urinary chondroitin sulfate from cancer patients. A potential new marker for diagnosis of neoplasias.
    Laboratory investigation; a journal of technical methods and pathology, 1993, Volume: 68, Issue:4

    Chondroitin sulfate is significantly increased in tumors (10 to 100 times) when compared to the amounts present in normal adjacent tissues. To investigate if the changes in concentration of chondroitin sulfate could be reflected in the urine of cancer patients we have analyzed the chondroitin sulfate excreted by 44 patients with different types of tumors, 50 normal individuals and 15 patients with unrelated diseases.. The identification and structural analyses of the sulfated glycosaminoglycans were made by electrophoresis and degradation with specific enzymes (chondroitinases AC and ABC), identification/quantitation of their disaccharide products by chromatography (paper and HPLC) and chemical determinations.. The disaccharide products formed from chondroitin sulfate of the 44 cancer patients by action of chondroitinase ABC show a substantial relative increase of non sulfated disaccharide (32.1% +/- 15.2) with a relative decrease of 6-sulfated disaccharide (28.9% +/- 11.5) and 4-sulfated disaccharide (39.0% +/- 13.5) when compared to the chondroitin sulfate of normal subjects (9.1% +/- 2.2, 40.6% +/- 4.5 and 50.2% +/- 4.5, respectively) or from patients with unrelated diseases. There is a direct correlation between the non sulfated disaccharide content and the stage of malignancy of the cancer patients. A significant change of the ratio of chondroitin sulfate and heparan sulfate and a decrease in the electrophoretic migration of chondroitin sulfate were also observed in cancer patients.. All the cancer patients analyzed so far have shown the structural anomaly of the urinary chondroitin sulfate and this may be useful in the diagnosis and follow up of cancer therapy.

    Topics: Adolescent; Adult; Aged; Antineoplastic Agents; Biomarkers, Tumor; Child; Child, Preschool; Chondroitin Sulfates; Chromatography, High Pressure Liquid; Disaccharides; Electrophoresis, Agar Gel; Glycosaminoglycans; Heparitin Sulfate; Humans; Middle Aged; Neoplasms; Postoperative Period; Reference Values

1993
Pericellular heparans may contribute to the protection of cells from free radicals.
    Medical hypotheses, 1987, Volume: 23, Issue:1

    The variation of fine structure and quantity of heparans at cell surfaces could endow cells of different tissues, and in different growth states, with varying degrees of competence in reacting to extracellular free radicals known to be associated with neoplasia.

    Topics: Animals; Antioxidants; Free Radicals; Glycosaminoglycans; Heparin; Heparitin Sulfate; Humans; Models, Biological; Neoplasms

1987
Usefulness of basement membrane markers in tumoural pathology.
    The Journal of pathology, 1985, Volume: 145, Issue:4

    The distribution of basement membrane (BM) markers, type IV collagen, laminin (LM), heparan sulphate proteoglycan (HSP) and fibronectin (FN) has been studied by indirect immunofluorescence using specific antibodies, in tumoural pathology. The disrupted pattern of BM by these markers in severe dysplastic lesions of the breasts, the bronchi and uterine cervix provides evidence for malignancy. In invasive carcinomas, there is generally a loss of these BM components, with FN persisting in the stroma. The loss of these markers in BM is concomitant and superimposable in double staining studies. In embryonic tumours, the presence of BM markers is related to a mesenchymal differentiation of malignant cells with pericellular FN and/or maturation towards organoid structures with BM. In sarcomas, there is a loss of the pericellular BM staining around most transformed muscular and Schwann cells and adipocytes. The persistence of this labelling in some well-differentiated areas can help to diagnose the nature of the sarcoma. The persistence of intercellular filaments of FN corresponds to the mesenchymal and/or sarcomatous nature of undifferentiated anaplastic proliferations.

    Topics: Basement Membrane; Breast Neoplasms; Cell Transformation, Neoplastic; Collagen; Female; Fibronectins; Fluorescent Antibody Technique; Heparitin Sulfate; Humans; Laminin; Lung Neoplasms; Male; Neoplasms; Neoplasms, Germ Cell and Embryonal; Sarcoma

1985
[Basement membranes and tumor pathology].
    Bulletin du cancer, 1984, Volume: 71, Issue:5

    The distribution of four basement membrane components, type IV collagen (C IV), laminin (LM), heparan sulfate proteoglycan (HSP) and fibronection (FN) has been studied by indirect immunofluorescence using specific antibodies, in benign and malignant proliferations of the mammary gland and in soft tissue tumors. In breast carcinomas, specially intraductal cancers, there is a progressive and concomitant loss of these macromolecules around tumoral cells, preceding an overt tumoral invasion. In sarcomas, FN is frequently seen between malignant cells but the regular pericellular labeling observed around normal muscular cells, Schwann cells and adipocytes is absent. Nevertheless, the persistance of some pericellular staining with anti-C IV, anti-LM, anti-HSP and anti-FN antisera, in most differentiated territories of liposarcomas, leiomyosarcomas and neurifibrosarcomas can help to the diagnosis of such lesions.

    Topics: Basement Membrane; Breast Neoplasms; Collagen; Female; Fibronectins; Fluorescent Antibody Technique; Heparitin Sulfate; Humans; Laminin; Neoplasms; Sarcoma

1984
Macromolecular organization and functional architecture of basement membranes.
    Applied pathology, 1984, Volume: 2, Issue:4

    Basement membranes (BMs) are extracellular laminar matrices produced by endothelial and epithelial cells. They are composed by three major intrinsic macromolecules: type IV collagen, laminin and heparan sulfate proteoglycan (HSP), and by two major extrinsic ones: fibronectin and type V collagen. The intrinsic components are assembled in a three-dimensional network (type IV collagen) to which cells stick (by laminin) and through which (HSP) they interact with the stromal compartment. The BM is a barrier to be crossed by any cell that leaves the stroma to enter into the circulation or vice versa. Metastatic tumor cells secrete a protease which specifically degrades the BM collagen and some evidence suggests that the same enzyme is used also by normal monocytes.

    Topics: Animals; Basement Membrane; Chondroitin Sulfate Proteoglycans; Collagen; Fibronectins; Heparan Sulfate Proteoglycans; Heparitin Sulfate; Kidney Glomerulus; Laminin; Mice; Neoplasms

1984
A new method for characterization of N-sulfated glycosaminoglycans by a rapid and multisample nitrous acid treatment during an electrophoretic run and its application to the analysis of biological samples.
    Analytical biochemistry, 1980, Jul-01, Volume: 105, Issue:2

    Topics: Animals; Chondroitin Sulfates; Electrophoresis, Cellulose Acetate; Glycosaminoglycans; Heparitin Sulfate; Humans; Keratan Sulfate; Neoplasms; Nitrous Acid

1980
Intercellular glycosaminoglycans in normal and neoplastic tissues.
    Cancer research, 1978, Volume: 38, Issue:12

    Intercellular glycosaminoglycans have been isolated from normal and neoplastic mammalian tissues. They have been characterized by cellulose acetate electrophoresis and by chemical and enzymatic degradation. The electrophoretic pattern of the intercellular glycosaminoglycans is tissue specific. Furthermore, the electrophoretic patterns of all spontaneous neoplasias analyzed differ significantly from patterns obtained from the tissues of origin.

    Topics: Animals; Cell Membrane; Chondroitin Sulfates; Dermatan Sulfate; Glycosaminoglycans; Guinea Pigs; Heparitin Sulfate; Humans; Hyaluronic Acid; Mice; Neoplasms; Neoplasms, Experimental; Tissue Distribution; Trypsin

1978
Cell recognition and adhesiveness: a possible biological role for the sulfated mucopolysaccharides.
    Biochemical and biophysical research communications, 1977, Mar-21, Volume: 75, Issue:2

    Topics: Animals; Binding Sites; Brain; Cell Adhesion; Cell Aggregation; Chondroitin; Chondroitin Sulfates; Dermatan Sulfate; Glycosaminoglycans; Guinea Pigs; Heparitin Sulfate; Humans; Ileum; Kidney; Molecular Weight; Muscles; Neoplasms; Organ Specificity; Rabbits; Species Specificity

1977
Changes in sulfated mucopolysaccharide composition of mammalian tissues during growth and in cancer tissues.
    Biochimica et biophysica acta, 1977, Jun-23, Volume: 498, Issue:1

    The distribution of sulfated mucopolysaccharides in different tissues during growth and in cancer tissues is reported. It is shown that most of the tissues of 1 day-old rats and rabbits contain chondroitin sulfate A/C, chondroitin sulfate B and heparan sulfate in about the same proportions, whereas in adult animals chondroitin sulfate A/C decreases in concentration or disappears. Changes in the relative proportions of chondroitin sulfate B and heparan sulfate were also observed in most of the tissues. In rats, these changes occur in the first 25 days of extrauterine development. A great increase of chondroitin sulfate A/C was observed in human tumors of different origins when compared with the normal adjacent tissues. Changes in the relative proportions of chondroitin sulfate B and heparan sulfate were also observed in most of the tumors analysed. The possible role of chondroitin sulfate A/C in cell division is discussed in view of the present findings.

    Topics: Aging; Animals; Animals, Newborn; Brain; Chondroitin; Chondroitin Sulfates; Glycosaminoglycans; Guinea Pigs; Heparitin Sulfate; Humans; Kidney; Liver; Neoplasms; Organ Specificity; Rabbits; Rats; Species Specificity

1977