1
|
Rabinowitz ZM, Somers J, Wang Z, Cui L. Chemical toolbox to interrogate Heparanase-1 activity. Curr Opin Chem Biol 2024; 80:102452. [PMID: 38555836 DOI: 10.1016/j.cbpa.2024.102452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 04/02/2024]
Abstract
The development of a robust chemical toolbox to interrogate the activity of heparanase-1 (HPSE-1), an endo-β-d-glucuronidase and the only known enzyme that cleaves heparan sulfate (HS), has become critically important. The primary function of HPSE-1, cleaving HS side chains from heparan sulfate proteoglycans (HSPGs), regulates the integrity of the extracellular matrix (ECM) and the bioavailability of active, heparan sulfate-binding partners such as enzymes, growth factors, chemokines, and cytokines. HPSE-1 enzymatic activity is strictly regulated and has been found to play fundamental roles in pathophysiological processes. HPSE-1 is significantly overexpressed under various conditions including cancer, metastasis, angiogenesis, and inflammation, making HPSE-1 a promising therapeutic and diagnostic target. Chemical tools that can detect and image HPSE-1 activity in vitro and/or in vivo can help drive the discovery of novel and efficacious anti-HPSE-1 drugs, investigate the basic biology of HPSE-1, and help serve as a diagnostic tool in clinical applications. Here, we will give an overview of the common chemical tools to detect HPSE-1 activity and highlight the novel heparanase probes recently developed in our lab.
Collapse
Affiliation(s)
- Zachary M Rabinowitz
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Johnathan Somers
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Zhishen Wang
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | - Lina Cui
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
2
|
Vlodavsky I, Kayal Y, Hilwi M, Soboh S, Sanderson RD, Ilan N. Heparanase-A single protein with multiple enzymatic and nonenzymatic functions. PROTEOGLYCAN RESEARCH 2023; 1:e6. [PMID: 37547889 PMCID: PMC10398610 DOI: 10.1002/pgr2.6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 08/08/2023]
Abstract
Heparanase (Hpa1) is expressed by tumor cells and cells of the tumor microenvironment and functions extracellularly to remodel the extracellular matrix (ECM) and regulate the bioavailability of ECM-bound factors, augmenting, among other effects, gene transcription, autophagy, exosome formation, and heparan sulfate (HS) turnover. Much of the impact of heparanase on tumor progression is related to its function in mediating tumor-host crosstalk, priming the tumor microenvironment to better support tumor growth, metastasis, and chemoresistance. The enzyme appears to fulfill some normal functions associated, for example, with vesicular traffic, lysosomal-based secretion, autophagy, HS turnover, and gene transcription. It activates cells of the innate immune system, promotes the formation of exosomes and autophagosomes, and stimulates signal transduction pathways via enzymatic and nonenzymatic activities. These effects dynamically impact multiple regulatory pathways that together drive tumor growth, dissemination, and drug resistance as well as inflammatory responses. The emerging premise is that heparanase expressed by tumor cells, immune cells, endothelial cells, and other cells of the tumor microenvironment is a key regulator of the aggressive phenotype of cancer, an important contributor to the poor outcome of cancer patients and a valid target for therapy. So far, however, antiheparanase-based therapy has not been implemented in the clinic. Unlike heparanase, heparanase-2 (Hpa2), a close homolog of heparanase (Hpa1), does not undergo proteolytic processing and hence lacks intrinsic HS-degrading activity, the hallmark of heparanase. Hpa2 retains the capacity to bind heparin/HS and exhibits an even higher affinity towards HS than heparanase, thus competing for HS binding and inhibiting heparanase enzymatic activity. It appears that Hpa2 functions as a natural inhibitor of Hpa1 regulates the expression of selected genes that maintain tissue hemostasis and normal function, and plays a protective role against cancer and inflammation, together emphasizing the significance of maintaining a proper balance between Hpa1 and Hpa2.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Yasmin Kayal
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Maram Hilwi
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Soaad Soboh
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| | - Ralph D. Sanderson
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Neta Ilan
- Technion Integrated Cancer Center, TechnionRappaport Faculty of MedicineHaifaIsrael
| |
Collapse
|
3
|
Khanna M, Parish CR. Heparanase: Historical Aspects and Future Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:71-96. [PMID: 32274707 DOI: 10.1007/978-3-030-34521-1_3] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
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 that heparanase plays a key role in tumor metastasis, angiogenesis and inflammation has been confirmed by many studies over the last 20 years. In fact, heparanase expression is enhanced in all major cancer types, namely carcinomas, sarcomas, and hematological malignancies, and correlates with increased metastasis and poor prognosis. Also, there is mounting evidence that heparanase plays a central role in the induction of inflammation-associated cancers. The enzymatic activity of heparanase has also emerged in unexpected situations, such as in the spread of HS-binding viruses and in Type-1 diabetes where the destruction of intracellular HS in pancreatic insulin-producing beta cells precipitates diabetes. But the most extraordinary recent discoveries have been with the realization that heparanase can exert a range of biological activities that are independent of its enzymatic function, most notably activation of several signaling pathways and being a transcription factor that controls methylation of histone tails. Collectively, these data indicate that heparanase is a truly multifunctional protein that has the additional property of cleaving HS chains and releasing from ECM and cell surfaces hundreds of HS-binding proteins with a plethora of functional consequences. Clearly, there are many unique features of this intriguing molecule that still remain to be explored and are highlighted in this Chapter.
Collapse
Affiliation(s)
- Mayank Khanna
- Department of Immunology and Infectious Diseases, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.,Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Christopher R Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australia.
| |
Collapse
|
4
|
Vlodavsky I, Sanderson RD, Ilan N. Forty Years of Basic and Translational Heparanase Research. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:3-59. [PMID: 32274705 PMCID: PMC7142273 DOI: 10.1007/978-3-030-34521-1_1] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This review summarizes key developments in the heparanase field obtained 20 years prior to cloning of the HPSE gene and nearly 20 years after its cloning. Of the numerous publications and review articles focusing on heparanase, we have selected those that best reflect the progression in the field as well as those we regard important accomplishments with preference to studies performed by scientists and groups that contributed to this book. Apart from a general 'introduction' and 'concluding remarks', the abstracts of these studies are presented essentially as published along the years. We apologize for not being objective and not being able to include some of the most relevant abstracts and references, due to space limitation. Heparanase research can be divided into two eras. The first, initiated around 1975, dealt with identifying the enzyme, establishing the relevant assay systems and investigating its biological activities and significance in cancer and other pathologies. Studies performed during the first area are briefly introduced in a layman style followed by the relevant abstracts presented chronologically, essentially as appears in PubMed. The second era started in 1999 when the heparanase gene was independently cloned by 4 research groups [1-4]. As expected, cloning of the heparanase gene boosted heparanase research by virtue of the readily available recombinant enzyme, molecular probes, and anti-heparanase antibodies. Studies performed during the second area are briefly introduced followed by selected abstracts of key findings, arranged according to specific topics.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center (TICC) Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Haifa Israel
| | - Ralph D. Sanderson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Neta Ilan
- Technion Integrated Cancer Center (TICC) Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Haifa Israel
| |
Collapse
|
5
|
Wu L, Davies GJ. An Overview of the Structure, Mechanism and Specificity of Human Heparanase. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:139-167. [PMID: 32274709 DOI: 10.1007/978-3-030-34521-1_5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The retaining endo-β-D-glucuronidase Heparanase (HPSE) is the primary mammalian enzyme responsible for breakdown of the glycosaminoglycan heparan sulfate (HS). HPSE activity is essential for regulation and turnover of HS in the extracellular matrix, and its activity affects diverse processes such as inflammation, angiogenesis and cell migration. Aberrant heparanase activity is strongly linked to cancer metastasis, due to structural breakdown of extracellular HS networks and concomitant release of sequestered HS-binding growth factors. A full appreciation of HPSE activity in health and disease requires a structural understanding of the enzyme, and how it engages with its HS substrates. This chapter summarizes key findings from the recent crystal structures of human HPSE and its proenzyme. We present details regarding the 3-dimensional protein structure of HPSE and the molecular basis for its interaction with HS substrates of varying sulfation states. We also examine HPSE in a wider context against related β-D-glucuronidases from other species, highlighting the structural features that control exo/endo - glycosidase selectivity in this family of enzymes.
Collapse
Affiliation(s)
- Liang Wu
- York Structural Biology Laboratory, Department of Chemistry, The University of York, York, UK.
| | - Gideon J Davies
- York Structural Biology Laboratory, Department of Chemistry, The University of York, York, UK
| |
Collapse
|
6
|
Trebicz-Geffen M, Robinson D, Evron Z, Glaser T, Fridkin M, Kollander Y, Vlodavsky I, Ilan N, Law KF, Cheah KSE, Chan D, Werner H, Nevo Z. The molecular and cellular basis of exostosis formation in hereditary multiple exostoses. Int J Exp Pathol 2008; 89:321-31. [PMID: 18452536 DOI: 10.1111/j.1365-2613.2008.00589.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
The different clinical entities of osteochondromas, hereditary multiple exostoses (HME) and non-familial solitary exostosis, are known to express localized exostoses in their joint metaphyseal cartilage. In the current study biopsies of osteochondromas patients were screened with respect to a number of cellular and molecular parameters. Specifically, cartilaginous biopsy samples of nine HME patients, 10 solitary exostosis patients and 10 articular cartilages of control subjects were collected and cell cultures were established. Results obtained showed that one of the two HME samples that underwent DNA sequencing analysis (HME-1) had a novel mutation for an early stop codon, which led to an aberrant protein, migrating at a lower molecular weight position. The EXT-1 mRNA and protein levels in chondrocyte cultures derived from all nine HME patients were elevated, compared with solitary exostosis patients or control subjects. Furthermore, cell cultures of HME patients had significantly decreased pericellular heparan sulphate (HS) in comparison with cultures of solitary exostosis patients or control subjects. Immunohistochemical staining of tissue sections and Western blotting of cell cultures derived from HME patients revealed higher levels of heparanase compared with solitary exostosis patients and of control subjects. Further investigations are needed to determine whether the low pericellular HS levels in HME patients stem from decreased biosynthesis of HS, increased degradation or a combination of both. In conclusion, it appears that due to a mutated glycosyltransferase, the low content of pericellular HS in HME patients leads to the anatomical deformations with exostoses formation. Hence, elevation of HS content in the pericellular regions should be a potential molecular target for correction.
Collapse
Affiliation(s)
- Meirav Trebicz-Geffen
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv, Israel
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Abstract
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.
Collapse
Affiliation(s)
- E A McKenzie
- Faculty of Life Sciences, University of Manchester, Manchester, UK.
| |
Collapse
|
8
|
Kresse H, Glössl J. Glycosaminoglycan degradation. ADVANCES IN ENZYMOLOGY AND RELATED AREAS OF MOLECULAR BIOLOGY 2006; 60:217-311. [PMID: 3310531 DOI: 10.1002/9780470123065.ch4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- H Kresse
- Institute of Physiological Chemistry, University of Münster, Federal Republic of Germany
| | | |
Collapse
|
9
|
Fernández C, Hattan CM, Kerns RJ. Semi-synthetic heparin derivatives: chemical modifications of heparin beyond chain length, sulfate substitution pattern and N-sulfo/N-acetyl groups. Carbohydr Res 2006; 341:1253-65. [PMID: 16712822 DOI: 10.1016/j.carres.2006.04.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2006] [Revised: 04/04/2006] [Accepted: 04/09/2006] [Indexed: 11/24/2022]
Abstract
The glycosaminoglycan heparin is a polyanionic polysaccharide most recognized for its anticoagulant activity. Heparin binds to cationic regions in hundreds of prokaryotic and eukaryotic proteins, termed heparin-binding proteins. The endogenous ligand for many of these heparin-binding proteins is a structurally similar glycosaminoglycan, heparan sulfate (HS). Chemical and biosynthetic modifications of heparin and HS have been employed to discern specific sequences and charge-substitution patterns required for these polysaccharides to bind specific proteins, with the goal of understanding structural requirements for protein binding well enough to elucidate the function of the saccharide-protein interactions and/or to develop new or improved heparin-based pharmaceuticals. The most common modifications to heparin structure have been alteration of sulfate substitution patterns, carboxyl reduction, replacement N-sulfo groups with N-acetyl groups, and chain fragmentation. However, an accumulation of reports over the past 50 years describe semi-synthetic heparin derivatives obtained by incorporating aliphatic, aryl, and heteroaryl moieties into the heparin structure. A primary goal in many of these reports has been to identify heparin-derived structures as new or improved heparin-based therapeutics. Presented here is a perspective on the introduction of non-anionic structural motifs into heparin structure, with a focus on such modifications as a strategy to generate novel reduced-charge heparin-based bind-and-block antagonists of HS-protein interactions. The chemical methods employed to synthesize such derivatives, as well as other unique heparin conjugates, are reviewed.
Collapse
Affiliation(s)
- Cristina Fernández
- Division of Medicinal and Natural Products Chemistry, University of Iowa, Iowa City, IA 52242, USA
| | | | | |
Collapse
|
10
|
Nardella C, Steinkühler C. Radiolabeled heparan sulfate immobilized on microplate as substrate for the detection of heparanase activity. Anal Biochem 2005; 332:368-75. [PMID: 15325306 DOI: 10.1016/j.ab.2004.05.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2004] [Indexed: 11/20/2022]
Abstract
We developed a quantitative assay to monitor the enzymatic activity of heparanase, a protein responsible for the degradation of heparan sulfate (HS) present on cell surface and extracellular matrix. Our assay is based on a new procedure to immobilize radiolabeled HS to a solid support by a single end which is adaptable to a microplate format, thus allowing the rapid analysis of numerous samples. First, HS was radiolabeled by partial de-N-acetylation and re-N-acetylation with [3H] acetic anhydride, second, after reductive amination at the reducing terminus, it was covalently linked to an amino-reactive biotin analog, and third it was immobilized on a streptavidin-coated plate. The degradation of our solid-phase tritiated HS by heparanase was monitored by measuring the soluble radioactivity released in the well. The heparanase-induced release of radioactivity was linear with respect either to time or to the amount of enzyme and was inhibited by heparin or high ionic strength. The linearity of this assay for time and enzyme concentrations could be useful to determine the potency of heparanase inhibitors. Moreover, this assay was shown to be suitable for monitoring HS-degrading activity of either heparanase endogenously expressed by the HCT 116 tumor cell line or recombinant forms of this protein.
Collapse
Affiliation(s)
- Caterina Nardella
- Department of Biochemistry, IRBM/Merck Research Laboratories, Via Pontina Km 30,600, 00040, Pomezia, Italy
| | | |
Collapse
|
11
|
Sato T, Yamaguchi A, Goi T, Hirono Y, Takeuchi K, Katayama K, Matsukawa S. Heparanase expression in human colorectal cancer and its relationship to tumor angiogenesis, hematogenous metastasis, and prognosis. J Surg Oncol 2004; 87:174-81. [PMID: 15334632 DOI: 10.1002/jso.20097] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Heparanase is considered to play an important role in tumor invasion and hematogenous metastasis. The aim of this study was to determine the expression of heparanase in colorectal cancer to evaluate its prognostic value. METHODS We analyzed heparanase mRNA derived from 130 colorectal cancer patients by reverse transcription polymerase chain reaction (PCR), compared its expression with clinicopathologic factors, and performed statistical analysis. To investigate the relationship between heparanase expression and tumor angiogenesis, 81 primary colorectal tumors were immunostained by use of a monoclonal anti-CD34 antibody. RESULTS Thirty three of 130 cancer tissues overexpressed heparanase. There were significant correlations between heparanase expression and serosal invasion, venous invasion, and liver metastasis. Multivariate analyzes revealed that heparanase mRNA overexpression was a significant independent risk factor for hematogenous metastasis in colorectal cancer. Among 104 patients who underwent curative resection, heparanase expression correlated with a high recurrence. The 5-year survival rate was 84.6% for patients with heparanase negative tumors, and 47.7% for those with heparanase overexpression; these differences between two groups of patients were significant. In multivariate analysis using the Cox regression model, heparanese expression emerged as an independent prognostic indicator. Moreover, the tumor angiogenesis of heparanase-positive tumors determined with a monoclonal anti-CD34 antibody was significantly higher than that of heparanase-negative tumors. CONCLUSIONS These results indicated that Heparanase expression may be an important role in invasion and hematogenous metastasis, and may be a biologic marker of prognostic significance in colorectal cancer patients.
Collapse
Affiliation(s)
- Tomohiro Sato
- First Department of Surgery, University of Fukui, Matsuoka-cho, Yoshida-gun, Fukui, Japan
| | | | | | | | | | | | | |
Collapse
|
12
|
Okada Y, Yamada S, Toyoshima M, Dong J, Nakajima M, Sugahara K. Structural recognition by recombinant human heparanase that plays critical roles in tumor metastasis. Hierarchical sulfate groups with different effects and the essential target disulfated trisaccharide sequence. J Biol Chem 2002; 277:42488-95. [PMID: 12213822 DOI: 10.1074/jbc.m206510200] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Human heparanase is an endo-beta-d-glucuronidase that degrades heparan sulfate/heparin and has been implicated in a variety of biological processes, such as inflammation, tumor angiogenesis, and metastasis. Although the cloned enzyme has been demonstrated to have a critical role in tumor metastasis, the substrate specificity has been poorly understood. In the present study, the specificity of the purified recombinant human heparanase was investigated for the first time using a series of structurally defined oligosaccharides isolated from heparin/heparan sulfate. The best substrates were deltaHexUA(+/-2S)-GlcN(NS,6S)-GlcUA-GlcN(NS,6S)-GlcUA-GlcN(NS,6S) and deltaHexUA(2S)-GlcN(NS,6S)-GlcUA-GlcN(NS,6S) (where deltaHexUA, GlcN, GlcUA, NS, 2S, and 6S represent unsaturated hexuronic acid, d-glucosamine, d-glucuronic acid, 2-N-sulfate, 2-O-sulfate, and 6-O-disulfate, respectively). Based on the percentage conversion of the substrates to products under identical assay conditions, several aspects of the recognition structures were revealed. 1) The minimum recognition backbone is the trisaccharide GlcN-GlcUA-GlcN. 2) The target GlcUA residues are in the sulfated region. 3) The -GlcN(6S)-GlcUA-GlcN(NS)- sequence is essential but not sufficient as the cleavage site. 4) The IdoUA(2S) residue, located two saccharides away from the target GlcUA residue, claimed previously to be essential, is not indispensable. 5) The 3-O-sulfate group on the GlcN is dispensable and even has an inhibitory effect when located in a highly sulfated region. 6) Based on these and previous results, HexUA(2S)-GlcN(NS,6S)-IdoUA-GlcNAc(6S)-GlcUA-GlcN(NS,+/-6S)-IdoUA(2S)-GlcN(NS,6S) (where HexUA represents hexuronic acid) has been proposed as a probable physiological target octasaccharide sequence. These findings will aid establishing a quantitative assay method using the above tetrasaccharide and designing heparan sulfate-based specific inhibitors of the heparanase for new therapeutic strategies.
Collapse
Affiliation(s)
- Yukihiko Okada
- Department of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-8558, Japan
| | | | | | | | | | | |
Collapse
|
13
|
Podyma-Inoue KA, Yokote H, Sakaguchi K, Ikuta M, Yanagishita M. Characterization of heparanase from a rat parathyroid cell line. J Biol Chem 2002; 277:32459-65. [PMID: 12077130 DOI: 10.1074/jbc.m203282200] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cell surface heparan sulfate proteoglycans undergo unique intracellular degradation pathways after they are endocytosed from the cell surface. Heparanase, an endo-beta-glucuronidase capable of cleaving heparan sulfate, has been demonstrated to contribute to the physiological degradation of heparan sulfate proteoglycans and therefore regulation of their biological functions. A rat parathyroid cell line was found to produce heparanase with an optimal activity at neutral and slightly acidic conditions suggesting that the enzyme participates in heparan sulfate proteoglycan metabolism in extralysosomal compartments. To elucidate the detailed properties of the purified enzyme, the substrate specificity against naturally occurring heparan sulfates and chemically modified heparins was studied. Cleavage sites of rat heparanase were present in heparan sulfate chains obtained from a variety of animal organs, but their occurrence was infrequent (average, 1-2 sites per chain) requiring recognition of both undersulfated and sulfated regions of heparan sulfate. On the other hand intact and chemically modified heparins were not cleaved by heparanase. The carbohydrate structure of the newly generated reducing end region of heparan sulfate cleaved by the enzyme was determined, and it represented relatively undersulfated structures. O-Sulfation of heparan sulfate chains also played important roles in substrate recognition, implying that rat parathyroid heparanase acts near the boundary of highly sulfated and undersulfated domains of heparan sulfate proteoglycans. Further elucidation of the roles of heparanase in normal physiological processes would provide an important tool for analyzing the regulation of heparan sulfate-dependent cell functions.
Collapse
Affiliation(s)
- Katarzyna A Podyma-Inoue
- Division of Biochemistry, Department of Hard Tissue Engineering, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8459, Japan
| | | | | | | | | |
Collapse
|
14
|
Casu B, Lindahl U. Structure and biological interactions of heparin and heparan sulfate. Adv Carbohydr Chem Biochem 2002; 57:159-206. [PMID: 11836942 DOI: 10.1016/s0065-2318(01)57017-1] [Citation(s) in RCA: 288] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- B Casu
- G. Ronzoni Institute for Chemical and Biochemical Research, Milan, Italy
| | | |
Collapse
|
15
|
Abstract
Although immune responses are generally considered to be systemic, local events such as interaction of complement products with blood vessels and with inflammatory cells play a pivotal role in determining the nature and manifestations of immune responses. This paper will discuss how blood vessel physiology and immunity influence one another to reach homeostasis upon exposure to an infectious agent. We review new insights into the mechanisms by which the microenvironment of tissues protects against microbial invasion yet facilitates migration of leukocytes and 'decides' whether immunity or tolerance ensues and whether, in the face of immunity, protective responses or tissue injury ensues. These 'decisions' are made based on interaction of components of normal tissues such as proteoglycans and injured tissues such as cell-associated cytokines with receptors on immune cells and blood vessels.
Collapse
Affiliation(s)
- Soheyla Saadi
- Department of Surgery, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
16
|
Furuta K, Ikeda M, Nakayama Y, Nakamura K, Tanaka M, Hamasaki N, Himeno M, Hamilton SR, August JT. Expression of lysosome-associated membrane proteins in human colorectal neoplasms and inflammatory diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 159:449-55. [PMID: 11485903 PMCID: PMC1850563 DOI: 10.1016/s0002-9440(10)61716-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The lysosome-associated membrane proteins (LAMPs)-1 and -2 are major constituents of the lysosomal membrane. These molecules are known to be among the most glycosylated proteins of several types of cells and cancer cells, and their expression in cancer cells is marked by a distinct difference in the structures of the oligosaccharides as compared to nonmalignant cells. We analyzed by immunohistochemistry the intensity and distribution of LAMP-1 and LAMP-2 in 9 human colorectal cancer cases and in 16 control cases, including inflammatory diseases (diverticulitis, ulcerative colitis, and Crohn's disease). LAMP proteins were expressed more intensely in the epithelium of colorectal neoplasms than in normal mucosa (P < 0.05), and no significant differences were found between adenoma and cancer cells (P > 0.05) in the same tissue section. Further, in sites of inactive inflammatory diseases and nonneoplastic areas in cancer specimens, no significant increases in epithelial LAMP proteins were observed, even in the proliferative zone of the lower crypt epithelium. Northern blot analysis showed increased expression of LAMP-1 and LAMP-2A in two of three colorectal cancers examined and increased LAMP-2B in all three cancers. Our findings suggest that LAMPs are related to neoplastic progression, but there is no direct association between the expression of LAMP molecules and cell proliferation.
Collapse
Affiliation(s)
- Koh Furuta
- National Cancer Center Hospital, Tokyo, Japan; the Department of Occupational Health Economics,†
| | - Masato Ikeda
- University of Occupational and Environmental Health, Kitakyushu, Japan; the Department of Pathology 1,‡
| | - Yoshifuku Nakayama
- School of Medicine, Fukuoka University, Fukuoka, Japan; the Departments of Surgery 1§
| | | | - Masao Tanaka
- and Clinical Chemistry and Laboratory Medicine,¶
| | - Naotaka Hamasaki
- Graduate School of Medical Sciences, and the Division of Pharmaceutical Cell Biology,∥
| | - Masaru Himeno
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan; the Department of Pathology,**
| | - Stanley R. Hamilton
- Division of Gastrointestinal-Liver Pathology, and the Department of Pharmacology and Molecular Sciences,††
| | - J. Thomas August
- The Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
17
|
Abstract
Heparanases are endoglycosidases that cleave the heparan sulfate glycosaminoglycans from proteoglycan core proteins and degrade them to small oligosaccharides. Inside cells, these enzymes are important for the normal catabolism of heparan sulfate proteoglycans (HSPGs), generating glycosaminoglycan fragments that are then transported to lysosomes and completely degraded. When secreted, heparanases are thought to degrade basement membrane HSPGs at sites of injury or inflammation, allowing extravasion of immune cells into nonvascular spaces and releasing factors that regulate cell proliferation and angiogenesis. Heparanases have been described in a wide variety of tissues and cells, but because of difficulties in developing simple assays to follow activity, very little has been known about enzyme diversity until recently. Within the last 10 years, heparanases have been purified from platelets, placenta, and Chinese hamster ovary cells. Characterization of the enzymes suggests there may be a family of heparanase proteins with different substrate specificities and potential functions.
Collapse
Affiliation(s)
- K J Bame
- Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| |
Collapse
|
18
|
Parish CR, Freeman C, Hulett MD. Heparanase: a key enzyme involved in cell invasion. BIOCHIMICA ET BIOPHYSICA ACTA 2001; 1471:M99-108. [PMID: 11250066 DOI: 10.1016/s0304-419x(01)00017-8] [Citation(s) in RCA: 190] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- C R Parish
- Division of Immunology and Cell Biology, John Curtin School of Medical Research, Australian National University, Canberra, A.C.T. 0200, Australia.
| | | | | |
Collapse
|
19
|
McKenzie E, Tyson K, Stamps A, Smith P, Turner P, Barry R, Hircock M, Patel S, Barry E, Stubberfield C, Terrett J, Page M. Cloning and expression profiling of Hpa2, a novel mammalian heparanase family member. Biochem Biophys Res Commun 2000; 276:1170-7. [PMID: 11027606 DOI: 10.1006/bbrc.2000.3586] [Citation(s) in RCA: 142] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Heparan sulfate proteoglycans are important constituents of the extracellular matrix and basement membrane. Cleavage of heparan sulfate by heparanase, an endoglycosidase, is implicated in the extravasation of leukocytes and metastatic tumour cells, identifying this enzyme(s) as a target for anti-inflammatory and anti-metastatic therapies. The cloning of a cDNA encoding human heparanase (Hpa1) was reported recently, together with evidence indicating that the hpa1 gene is unique and unlikely to belong to a family of related genes. Here we report the cloning of a cDNA encoding a novel human protein, HPA2, with significant homology to Hpa1. Alternative splicing of the hpa2 transcript yields three different mRNAs, encoding putative proteins of 480, 534, and 592 amino acids. Sequence analyses predict that all three Hpa2 proteins are intracellular, membrane-bound enzymes. Hpa2 also shows a markedly different mRNA distribution to Hpa1 in both normal and cancer tissues. The difference in expression profiles and predicted cellular locations suggests that Hpa2 and Hpa1 proteins have distinct biological functions.
Collapse
Affiliation(s)
- E McKenzie
- Oxford GlycoSciences, 10 The Quadrant, Abingdon Science Park, Abingdon, Oxon, OX14 3YS, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Dempsey LA, Brunn GJ, Platt JL. Heparanase, a potential regulator of cell-matrix interactions. Trends Biochem Sci 2000; 25:349-51. [PMID: 10916150 DOI: 10.1016/s0968-0004(00)01619-4] [Citation(s) in RCA: 146] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- L A Dempsey
- Depts of Surgery, Immunology and Pediatrics, and Transplantation Biology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
21
|
Bame KJ, Venkatesan I, Stelling HD, Tumova S. The spacing of S-domains on HS glycosaminoglycans determines whether the chain is a substrate for intracellular heparanases. Glycobiology 2000; 10:715-26. [PMID: 10910975 DOI: 10.1093/glycob/10.7.715] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Heparanases are mammalian endoglucuronidases that degrade heparan sulfate (HS) glycosaminoglycans to short 5-6 kDa pieces. In the Golgi, HS glycosaminoglycans are modified by a series of interdependent reactions which result in chains that have regions rich in N- and O-sulfate groups and iduronate residues (S-domains), separated by regions that are nearly devoid of sulfate. Structural analysis of the short HS chains produced by Chinese hamster ovary (CHO) cell heparanases indicate that the enzymes recognize differences in sulfate content between S-domains and unmodified sequences, and cleave the chain at junctions between these regions. To look more closely at whether the spacing of S-domains on the gly- cosaminoglycan influences its ability to be cleaved by heparanases, we examined the susceptibility of the HS chains synthesized by the proteoglycan synthesis mutant, pgsE-606. PGS:E-606 cells are deficient in the modification enzyme N-deacetylase/N-sulfotransferase I, and synthesize HS chains that have fewer N- and O-sulfate groups and iduronate residues compared to wild-type (Bame et al., (1991), J. Biol. Chem., 266, 10287). HS glycosaminoglycans were isolated from wild-type and pgsE-606 cells and separated into populations based on sulfate content. Compared to wild-type HS, which has 14 S-domains, pgsE-606 cells synthesize three HS species, 606-1, 606-2, and 606-3, with 1, 4, and 8 S-domains, respectively. The spacing of the S-domains on the pgsE-606 HS chains is similar to the spacing the modified sequences on wild-type HS, indicating that each mutant glycosaminoglycan is composed of wild-type-like sequences and sequences devoid of S-domains. When incubated with partially purified CHO heparanases, only the portion of the mutant HS chains that had S-domains were degraded. Structural analysis of the heparanase-products confirmed that both the number and the arrangement of S-domains on the HS glycosaminoglycan are important for heparanase susceptibility. The structure of the different pgsE-606 HS chains also suggests mechanisms for the placement of S-domains when the gly- cosaminoglycan is synthesized.
Collapse
Affiliation(s)
- K J Bame
- Division of Molecular Biology and Biochemistry, School of Biological Sciences, University of Missouri-Kansas City, 64110, USA
| | | | | | | |
Collapse
|
22
|
Dempsey LA, Plummer TB, Coombes SL, Platt JL. Heparanase expression in invasive trophoblasts and acute vascular damage. Glycobiology 2000; 10:467-75. [PMID: 10764835 DOI: 10.1093/glycob/10.5.467] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Heparan sulfate proteoglycans play a pivotal role in tissue function, development, inflammation, and immunity. We have identified a novel cDNA encoding human heparanase, an enzyme thought to cleave heparan sulfate in physiology and disease, and have located the HEP gene on human chromosome 4q21. Monoclonal antibodies against human heparanase located the enzyme along invasive extravillous trophoblasts of human placenta and along endothelial cells in organ xenografts targeted by hyperacute rejection, both sites of heparan sulfate digestion. Heparanase deposition was evident in arterial walls in normal tissues; however, vascular heparan sulfate cleavage was coincident with heparanase enzyme during inflammatory episodes. These findings suggest that heparanase elaboration and control of catalytic activity may contribute to the development and pathogenesis of vascular disease and suggest that heparanase intervention might be a useful therapeutic target.
Collapse
Affiliation(s)
- L A Dempsey
- Department of Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
23
|
Nishimura Y, Shitara E, Adachi H, Toyoshima M, Nakajima M, Okami Y, Takeuchi T. Flexible synthesis and biological activity of uronic acid-type gem-diamine 1-N-iminosugars: a new family of glycosidase inhibitors. J Org Chem 2000; 65:2-11. [PMID: 10813888 DOI: 10.1021/jo982448c] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An efficient and flexible synthetic route to four gem-diamine 1-N-iminosugars of uronic acid-type (D-glucuronic, D-mannuronic, L-iduronic, and L-guluronic acid), a new family of glycosidase inhibitor, from l-galactono-1,4-lactone have been developed in an enantiodivergent fashion through a sequence involving as the key steps (a) the formation of gem-diamine 1-N-iminopyranose ring by the Mitsunobu reaction of an aminal and (b) the introduction of a carboxylic acid group by the Wittig reaction of a ketone, hydroboration and oxidation, and the Sharpless oxidation. D-Glucuronic and D-mannuronic acid-type 1-N-iminosugars, (3S,4R,5R, 6R)- and (3S,4R,5R,6S)-4, 5-dihydroxy-6-trifluoroacetamido-3-piperidinecarboxylic acid, were proven to be potent inhibitors for beta-D-glucuronidase (IC(50) 6.5 x 10(-)(8)M) and to affect human heparanase (endo-beta-glucuronidase).
Collapse
Affiliation(s)
- Y Nishimura
- Institute of Microbial Chemistry, 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan
| | | | | | | | | | | | | |
Collapse
|
24
|
Kussie PH, Hulmes JD, Ludwig DL, Patel S, Navarro EC, Seddon AP, Giorgio NA, Bohlen P. Cloning and functional expression of a human heparanase gene. Biochem Biophys Res Commun 1999; 261:183-7. [PMID: 10405343 DOI: 10.1006/bbrc.1999.0962] [Citation(s) in RCA: 160] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
We have cloned a gene (HSE1) from a human placental cDNA library that encodes a novel protein exhibiting heparanase activity. The cDNA was identified through peptide sequences derived from purified heparanase isolated from human SK-HEP-1 hepatoma cells. HSE1 contains an open reading frame encoding a predicted polypeptide of 543 amino acids and possesses a putative signal sequence at its amino terminus. Northern blot analysis suggested strong expression of HSE1 in placenta and spleen. Transient transfection of HSE1 in COS7 cells resulted in the expression of a protein with an apparent molecular mass of 67-72 kDa. HSE1 protein was detectable in conditioned media but was also associated with the membrane fraction following cell lysis. The HSE1 gene product was shown to exhibit heparanase activity by specifically cleaving a labeled heparan sulfate substrate in a similar manner as purified native protein.
Collapse
Affiliation(s)
- P H Kussie
- ImClone Systems, Inc., 180 Varick Street, New York, New York, 10014, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Hulett MD, Freeman C, Hamdorf BJ, Baker RT, Harris MJ, Parish CR. Cloning of mammalian heparanase, an important enzyme in tumor invasion and metastasis. Nat Med 1999; 5:803-9. [PMID: 10395326 DOI: 10.1038/10525] [Citation(s) in RCA: 394] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The endoglycosidase heparanase is an important in the degradation of the extracellular matrix by invading cells, notably metastatic tumor cells and migrating leukocytes. Here we report the cDNA sequence of the human platelet enzyme, which encodes a unique protein of 543 amino acids, and the identification of highly homologous sequences in activated mouse T cells and in a highly metastatic rat adenocarcinoma. Furthermore, the expression of heparanase mRNA in rat tumor cells correlates with their metastatic potential. Exhaustive studies have shown only one heparanase sequence, consistent with the idea that this enzyme is the dominant endoglucuronidase in mammalian tissues.
Collapse
Affiliation(s)
- M D Hulett
- Division of Immunology and Cell Biology, John Curtin School of Medical Research, ANU, Canberra, ACT, Australia
| | | | | | | | | | | |
Collapse
|
26
|
Vlodavsky I, Friedmann Y, Elkin M, Aingorn H, Atzmon R, Ishai-Michaeli R, Bitan M, Pappo O, Peretz T, Michal I, Spector L, Pecker I. Mammalian heparanase: gene cloning, expression and function in tumor progression and metastasis. Nat Med 1999; 5:793-802. [PMID: 10395325 DOI: 10.1038/10518] [Citation(s) in RCA: 611] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Heparan sulfate proteoglycans interact with many extracellular matrix constituents, growth factors and enzymes. Degradation of heparan sulfate by endoglycosidic heparanase cleavage affects a variety of biological processes. We have purified a 50-kDa heparanase from human hepatoma and placenta, and now report cloning of the cDNA and gene encoding this enzyme. Expression of the cloned cDNA in insect and mammalian cells yielded 65-kDa and 50-kDa recombinant heparanase proteins. The 50-kDa enzyme represents an N-terminally processed enzyme, at least 100-fold more active than the 65-kDa form. The heparanase mRNA and protein are preferentially expressed in metastatic cell lines and specimens of human breast, colon and liver carcinomas. Low metastatic murine T-lymphoma and melanoma cells transfected with the heparanase cDNA acquired a highly metastatic phenotype in vivo, reflected by a massive liver and lung colonization. This represents the first cloned mammalian heparanase, to our knowledge, and provides direct evidence for its role in tumor metastasis. Cloning of the heparanase gene enables the development of specific molecular probes for early detection and treatment of cancer metastasis and autoimmune disorders.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Carcinoma, Hepatocellular/enzymology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Cell Line
- Chromosome Mapping
- Chromosomes, Human, Pair 4
- Cloning, Molecular
- Disease Progression
- Enzyme Activation
- Extracellular Matrix/physiology
- Female
- Genomic Library
- Glucuronidase
- Glycoside Hydrolases/genetics
- Glycoside Hydrolases/isolation & purification
- Glycoside Hydrolases/metabolism
- Humans
- Liver Neoplasms/enzymology
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Mammals
- Mice
- Mice, Inbred DBA
- Molecular Sequence Data
- Molecular Weight
- Moths
- Neoplasm Metastasis/physiopathology
- Placenta/enzymology
- Pregnancy
- RNA, Messenger/genetics
- Recombinant Proteins/biosynthesis
- Reverse Transcriptase Polymerase Chain Reaction
- Transcription, Genetic
- Transfection
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- I Vlodavsky
- Department of Oncology, Hadassah-Hebrew University Hospital, Jerusalem, Israel.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Turnbull JE, Hopwood JJ, Gallagher JT. A strategy for rapid sequencing of heparan sulfate and heparin saccharides. Proc Natl Acad Sci U S A 1999; 96:2698-703. [PMID: 10077574 PMCID: PMC15832 DOI: 10.1073/pnas.96.6.2698] [Citation(s) in RCA: 119] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Sulfated glycosaminoglycans (GAGs) are linear polysaccharides of repeating disaccharide sequences on which are superimposed highly complex and variable patterns of sulfation, especially in heparan sulfate (HS). HS and the structurally related heparin exert important biological functions, primarily by interacting with proteins and regulating their activities. Evidence is accumulating that these interactions depend on specific saccharide sequences, but the lack of simple, direct techniques for sequencing GAG saccharides has been a major obstacle to progress. We describe how HS and heparin saccharides can be sequenced rapidly by using an integrated strategy with chemical and enzymic steps. Attachment of a reducing-end fluorescent tag establishes a reading frame. Partial selective chemical cleavage at internal N-sulfoglucosamine residues with nitrous acid then creates a set of fragments of defined sizes. Subsequent digestion of these fragments with combinations of exosulfatases and exoglycosidases permits the selective removal of specific sulfates and monosaccharides from their nonreducing ends. PAGE of the products yields a pattern of fluorescent bands from which the saccharide sequence can be read directly. Data are presented on sequencing of heparin tetrasaccharides and hexasaccharides of known structure; these data show the accuracy and versatility of this sequencing strategy. Data also are presented on the application of the strategy to the sequencing of an HS decasaccharide of unknown structure. Application and further development of this sequencing strategy, called integral glycan sequencing, will accelerate progress in defining the structure-activity relationships of these complex GAGs and lead to important insights into their biological functions.
Collapse
Affiliation(s)
- J E Turnbull
- School of Biochemistry, University of Birmingham, Edgbaston, Birmingham B15 2TT, England.
| | | | | |
Collapse
|
28
|
Gonzalez-Stawinski GV, Parker W, Holzknecht ZE, Huber NS, Platt JL. Partial sequence of human platelet heparitinase and evidence of its ability to polymerize. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1429:431-8. [PMID: 9989228 DOI: 10.1016/s0167-4838(98)00254-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Heparitinase cleaves heparan sulfate, a glycosaminoglycan associated with all nucleated mammalian cells and extracellular matrices. Despite the important physiologic role heparitinase is postulated to play in such processes as tumor metastasis and inflammation, the identity of the enzyme remains a matter of controversy and there is a question of whether heparitinase is CTAP III. We report a 900,000-fold purification of heparitinase from human platelets. A multi-step procedure utilizing chromatography on heparin, DEAE, hydroxyapatite and size exclusion matrices was employed and yielded a single protein as judged by Coomassie staining of protein separated by SDS-PAGE. The purified protein had an apparent molecular mass of 35 kDa by size exclusion chromatography and 55 kDa by SDS-PAGE. During purification, heparitinase activity co-eluted from the hydroxyapatite and size exclusion columns with the 35-55 kDa protein, confirming that the purified protein was indeed heparitinase. The 35-55 kDa protein reacted strongly with concanavalin A, a lectin known to bind to heparitinase, further confirming that the protein was heparitinase. Platelet heparitinase formed dimers and tetramers upon storage in a purified form, possibly accounting for the various molecular weights previously reported for the enzyme. A partial amino acid sequence of the protein revealed that heparitinase has not been previously sequenced.
Collapse
|
29
|
Phaneuf MD, Szycher M, Berceli SA, Dempsey DJ, Quist WC, LoGerfo FW. Covalent linkage of recombinant hirudin to a novel ionic poly(carbonate) urethane polymer with protein binding sites: determination of surface antithrombin activity. Artif Organs 1998; 22:657-65. [PMID: 9702317 DOI: 10.1046/j.1525-1594.1998.05091.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Surface thrombus formation on implantable biomaterials such as polyurethane is a major concern when utilizing these materials in the clinical setting. Thrombin, which is responsible for thrombus formation and smooth muscle cell activation, has been the target of numerous surface modification strategies in an effort to prevent this phenomenon from occurring. The purpose of this study was to covalently immobilize the potent, specific antithrombin agent recombinant hirudin (rHir) onto a novel polyurethane polymer synthesized with carboxylic acid groups which served as protein attachment sites. The in vitro efficacy of thrombin inhibition by this novel biomaterial surface was then evaluated. Bovine serum albumin (BSA), which was selected as the basecoat protein, was reacted with sulfo-SMCC in a 1:50 molar ratio. This BSA-SMCC complex was then covalently linked to the carboxylated polyurethane (cPU) surface via the crosslinker EDU (cPU-BSA-SMCC). This cPU-BSA-SMCC surface was then reacted with Traut's-modified 125I-rHir, a procedure which created free sulfhydryl groups on rHir (cPU-BSA-SMCC-S-125I-rHir). Using these crosslinking procedures, the cPU-BSA-SMCC-S-125I-rHir segments bound 188 +/- 40 ng/cm2 (n = 60) whereas the controls with non-specifically bound 125I-rHir (Mitrathane + EDC + BSA + 125I-rHir-SH and cPU-BSA + 125I-rHir-SH) bound 13 +/- 8 ng/cm2 and 4 +/- 8 ng/cm2, respectively. Evaluation of these cPU-BSA-SMCC-S-125I-rHir segments for 131I-thrombin inhibition using a chromogenic assay for thrombin showed that a maximum of 2.64 NIHU thrombin was inhibited in contrast to the controls which inhibited bound 0.76 and 0.70 NIHU. Controls with nonspecifically bound 125I-rHir also had 0.31 and 0.76 NIHU 131I-thrombin adherent to their respective surfaces whereas the maximum 131I-thrombin binding to the cPU-BSA-SMCC-S-rHir segments was 1.51 NIHU. Exposure to 131I-thrombin did not result in any release of covalently bound 125I-rHir from the cPU-BSA-SMCC-S-125I-rHir segments. Thus, these results demonstrate that rHir can be covalently bound to this novel polyurethane surface and still maintain potent antithrombin activity.
Collapse
Affiliation(s)
- M D Phaneuf
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
30
|
Pikas DS, Li JP, Vlodavsky I, Lindahl U. Substrate specificity of heparanases from human hepatoma and platelets. J Biol Chem 1998; 273:18770-7. [PMID: 9668050 DOI: 10.1074/jbc.273.30.18770] [Citation(s) in RCA: 219] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Heparan sulfate proteoglycans, attached to cell surfaces or in the extracellular matrix, interact with a multitude of proteins via their heparan sulfate side chains. Degradation of these chains by limited (endoglycosidic) heparanase cleavage is believed to affect a variety of biological processes. Although the occurrence of heparanase activity in mammalian tissues has been recognized for many years, the molecular characteristics and substrate recognition properties of the enzyme(s) have remained elusive. In the present study, the substrate specificity and cleavage site of heparanase from human hepatoma and platelets were investigated. Both enzyme preparations were found to cleave the single beta-D-glucuronidic linkage of a heparin octasaccharide. A capsular polysaccharide from Escherichia coli K5, with the same (-GlcUAbeta1,4-GlcNAcalpha1,4-)n structure as the unmodified backbone of heparan sulfate, resisted heparanase degradation in its native state as well as after chemical N-deacetylation/N-sulfation or partial enzymatic C-5 epimerization of beta-D-GlcUA to alpha-L-IdceA. By contrast, a chemically O-sulfated (but still N-acetylated) K5 derivative was susceptible to heparanase cleavage. O-Sulfate groups, but not N-sulfate or IdceA residues, thus are essential for substrate recognition by the heparanase(s). In particular, selective O-desulfation of the heparin octasaccharide implicated a 2-O-sulfate group on a hexuronic acid residue located two monosaccharide units from the cleavage site, toward the reducing end.
Collapse
Affiliation(s)
- D S Pikas
- Department of Medical Biochemistry and Microbiology, Uppsala University, The Biomedical Center, Box 575, S-751 23 Uppsala, Sweden.
| | | | | | | |
Collapse
|
31
|
Hatton MW, Ross B, Southward SM, DeReske M, Richardson M. Pretreatment of rabbits with either hirudin, ancrod, or warfarin significantly reduces the immediate uptake of fibrinogen and platelets by the deendothelialized aorta wall after balloon-catheter injury in vivo. Arterioscler Thromb Vasc Biol 1998; 18:816-24. [PMID: 9598842 DOI: 10.1161/01.atv.18.5.816] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Fibrinogen and platelets rapidly saturate the exposed subendothelium of a freshly deendothelialized aorta in vivo. As thrombin generated within the site of injury is largely responsible for fibrin(ogen) deposition, we questioned whether various anticoagulant treatments would inhibit uptake of both fibrinogen and platelets in vivo. Rabbits were anticoagulated by pretreatment with either Warfarin, Ancrod, or recombinant hirudin. Each anesthetized, anticoagulated (or saline-injected control) rabbit was injected i.v. with rabbit 51Cr-platelets and 125I-fibrinogen before a balloon-catheter deendothelializing (or sham) injury of the thoracic aorta. At 10 minutes after injury, the rabbit was exsanguinated and the aorta excised. Platelet adsorption by the deendothelialized aorta surface was substantially reduced in anticoagulated rabbits (controls, 2.2x10(5)/mm2; Warfarin-treated, 1.2x10(5)/mm2; Ancrod-treated, 5.3x10(4)/mm2; r-hirudin-treated [5 mg/kg], 5.3x10(4)/mm2), and a significant reduction of fibrinogen associated with the platelet layer (from 5.3 to 1 to 2 pmol/cm2) and within the underlying intima-media layer (from 16.9 to 5 to 6 pmol/cm2) was observed in the r-hirudin-and Warfarin-treated rabbits. The pattern of aorta-deposited 51Cr-platelets and 125I-fibrin in the anticoagulated rabbits corresponded well with an assessment by transmission electron microscopy of aortic tissue samples. We conclude that approximately 70% of fibrinogen uptake is thrombin dependent and that approximately 80% of platelet adsorption depends on codeposited fibrin(ogen) during the 10-minute interval after balloon injury. Pretreatment with an agent that interferes with either thrombin or fibrin production will inhibit the immediate interaction of fibrinogen and platelets with the freshly exposed subendothelium.
Collapse
Affiliation(s)
- M W Hatton
- Department of Pathology, McMaster University Health Sciences Centre, Hamilton, Ontario, Canada.
| | | | | | | | | |
Collapse
|
32
|
Bai X, Bame KJ, Habuchi H, Kimata K, Esko JD. Turnover of heparan sulfate depends on 2-O-sulfation of uronic acids. J Biol Chem 1997; 272:23172-9. [PMID: 9287321 DOI: 10.1074/jbc.272.37.23172] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
To study how the pattern of sulfation along a heparan sulfate chain affects its turnover, we examined heparan sulfate catabolism in wild-type Chinese hamster ovary cells and mutant pgsF-17, defective in 2-O-sulfation of uronic acid residues (Bai, X., and Esko, J. D. (1996) J. Biol. Chem. 271, 17711-17717). Heparan sulfate from the mutant contains normal amounts of 6-O-sulfated glucosamine residues and iduronic acid and somewhat higher levels of N-sulfated glucosamine residues but lacks any 2-O-sulfated iduronic or glucuronic acid residues. Pulse-chase experiments showed that both mutant and wild-type cells transport newly synthesized heparan sulfate proteoglycans to the plasma membrane, where they shed into the medium or move into the cell through endocytosis. Internalization of the cell-associated molecules leads to sequential endoglycosidase (heparanase) fragmentation of the chains and eventual lysosomal degradation. In wild-type cells, the chains begin to degrade within 1 h, leading to the accumulation of intermediate (10-20-kDa) and small (4-7-kDa) oligosaccharides. Mutant cells did not generate these intermediates, although internalization and intracellular trafficking of the heparan sulfate chains appeared normal, and the chains degraded with normal kinetics. This difference was not due to defective heparanase activities in the mutant, since cytoplasmic extracts from mutant cells cleaved wild-type heparan sulfate chains in vitro. Instead, the heparan sulfate chains from the mutant were relatively resistant to degradation by cellular heparanases. These findings suggest that 2-O-sulfated iduronic acid residues in heparan sulfate are important for cleavage by endogenous heparanases but not for the overall catabolism of the chains.
Collapse
Affiliation(s)
- X Bai
- Division of Cellular and Molecular Medicine, Department of Medicine, and the Glycobiology Program, UCSD Cancer Center, University of California at San Diego, La Jolla, California 92093-0687, USA
| | | | | | | | | |
Collapse
|
33
|
Nishimura Y, Satoh T, Adachi H, Kondo S, Takeuchi T, Azetaka M, Fukuyasu H, Iizuka Y. Synthesis and antimetastatic activity of L-iduronic acid-type 1-N-iminosugars. J Med Chem 1997; 40:2626-33. [PMID: 9258369 DOI: 10.1021/jm960627l] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
L-Iduronic acid-type 1-N-iminosugars, (3R,4S,5R,6R)- and (3R,4S,5S,6R)-6-acetamido-4-amino-5-hydroxypiperidine-3-carboxylic acid (6 and 7, respectively), (3R,4S,5R,6R)-6-acetamido-4- guanidino-5-hydroxypiperidine-3-carboxylic acid (8), and (3R,4S,5R,6R)-4-amino- and -guanidino-5-hydroxy-6-(trifluoroacetamido) piperidine-3-carboxylic acid (9 and 10, respectively), were synthesized from siastatin B (1), isolated from Streptomyces culture, by the intramolecular Michael addition of O-imidate to its alpha,beta-unsaturated ester through cis oxiamination as a key step. Preincubation of B16 BL6 cells with these compounds inhibited invasion of the cells through reconstituted basement membranes. Pulmonary metastasis of B16 BL6 cells in mice was remarkably inhibited by pretreatment of the cells with these compounds in culture.
Collapse
Affiliation(s)
- Y Nishimura
- Institute of Microbial Chemistry, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Phaneuf MD, Berceli SA, Bide MJ, Quist WC, LoGerfo FW. Covalent linkage of recombinant hirudin to poly(ethylene terephthalate) (Dacron): creation of a novel antithrombin surface. Biomaterials 1997; 18:755-65. [PMID: 9158859 DOI: 10.1016/s0142-9612(96)00193-7] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Thrombus formation and intimal hyperplasia on the surface of implantable biomaterials such as poly(ethylene terepthalate) (Dacron) vascular grafts are major concerns when utilizing these materials in the clinical setting. Thrombin, a pivotal enzyme in the blood coagulation cascade primarily responsible for thrombus formation and smooth muscle cell activation, has been the target of numerous strategies to prevent this phenomenon from occurring. The purpose of this study was to covalently immobilize the potent, specific antithrombin agent recombinant hirudin (rHir) to a modified Dacron surface and characterize the in vitro efficacy of thrombin inhibition by this novel biomaterial surface. Bovine serum albumin (BSA), which was selected as the "basecoat' protein, was reacted with various molar ratios of the cross-linker sulphosuccinimidyl 4-(N-maleimidomethyl) cyclohexane-1-carboxylate (sulpho-SMCC; 1:5-1:50). These BSA-SMCC complexes were then covalently linked to sodium hydroxide-hydrolysed Dacron (HD) segments via the cross-linker 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC). Covalent linkage of these complexes to HD (HD-BSA-SMCC) was not affected by any of the sulpho-SMCC cross-linker ratios assayed. rHir, which was initially reacted with 2-iminothiolane hydrochloride (Traut's reagent) in order to create sulphydryl groups, was then covalently bound to these HD-BSA-SMCC surfaces (HD-BSA-SMCC-S-rHir). The 1:50 (BSA: sulpho-SMCC) HD-BSA-SMCC-S-rHir segments bound 22-fold more rHir (111 ng per mg Dacron) compared to control segments and also possessed the greatest thrombin inhibition of the segments evaluated using a chromogenic substrate assay for thrombin. Further characterization of the HD-BSA-SMCC-S-rHir segments demonstrated that maximum thrombin inhibition was 20.43 NIHU, 14.6-fold greater inhibition than control segments (1.4 NIHU). Thrombin inhibition results were confirmed by 125I-thrombin binding experiments, which demonstrated that the 1:50 HD-BSA-SMCC-S-rHir segments had significantly greater specific thrombin adhesion compared to control segments. Non-specific 125I-thrombin binding to and release from the 1:50 HD-BSA-SMCC-S-rHir segments was also significantly less than the control segments. Thus, these results demonstrate that rHir can be covalently bound to a clinically utilized biomaterial (Dacron) while still maintaining its ability to bind and inhibit thrombin.
Collapse
Affiliation(s)
- M D Phaneuf
- Deaconess Hospital/Harvard Medical School, Vascular Surgery Research, Boston, MA 02215, USA
| | | | | | | | | |
Collapse
|
35
|
Proteoglycans: a special class of glycoproteins. ACTA ACUST UNITED AC 1997. [DOI: 10.1016/s0167-7306(08)60614-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
36
|
Bame KJ, Robson K. Heparanases Produce Distinct Populations of Heparan Sulfate Glycosaminoglycans in Chinese Hamster Ovary Cells. J Biol Chem 1997. [DOI: 10.1074/jbc.272.4.2245] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
37
|
Born J, Jann K, Assmann KJ, Lindahl U, Berden JH. N-Acetylated domains in heparan sulfates revealed by a monoclonal antibody against the Escherichia coli K5 capsular polysaccharide. Distribution of the cognate epitope in normal human kidney and transplant kidney with chronic vascular rejection. J Biol Chem 1996; 271:22802-9. [PMID: 8798457 DOI: 10.1074/jbc.271.37.22802] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The Escherichia coli K5 capsular polysaccharide has the same (GlcUA-->GlcNAc)n structure as the nonsulfated heparan sulfate/heparin precursor polysaccharide. A monoclonal antibody (mAb 865) against the K5 polysaccharide has been described (Peters, H., Jürs, M., Jann, B., Jann, K., Timmis, K. N., and Bitter-Sauermann, D. (1985) Infect. Immun. 50, 459-466). In this report, we demonstrate the binding of anti-K5 mAb 865 to N-acetylated sequences in heparan sulfates and heparan sulfate proteoglycans but not to heparin. This is shown by direct binding and fluid phase inhibition of mAb 865 in an enzyme-linked immunosorbent assay. In this system we found that the binding of the mAb decreased with increasing sulfate content of the polysaccharide. By testing chemically modified K5 and heparin polysaccharides, we found that each of the modifications that occur during heparan sulfate (HS) synthesis (N-sulfation, C-5 epimerization, and O-sulfation) prevents recognition by mAb 865. Samples of heparan sulfate from human aorta (HS-II) were selectively degraded so as to allow the separate isolation of N-sulfated and N-acetylated block structures. N-Sulfated oligosaccharides (obtained after N-deacetylation by hydrazinolysis followed by nitrous acid deamination at pH 3.9) were not recognized by mAb 865, in contrast to N-acetylated oligosaccharides (obtained after nitrous acid deamination at pH 1.5), although the reactivity was lower than for intact HS-II. Analysis of the latter's pH 1.5 deamination products by gel filtration indicated that a minimal size of 18 saccharide units was necessary for antibody binding. These results lead us to propose bivalent antibody-heparan sulfate interaction, in which both F(ab) domains of the mAb interact with their epitopes, both of which are present in a single large (>/=18 saccharide units) N-acetylated domain and additionally with single epitopes present in two N-acetylated sequences (each <18 saccharide units) bridged by a short N-sulfated domain. Immunohistochemistry with mAb 865 on cryostat sections of normal human kidney tissue, revealed its binding to most but not all renal basement membranes. However, all renal basement membranes contain heparan sulfate, as shown by a mAb against heparitinase-digested heparan sulfate stubs (mAb 3G10). This finding indicates that not all heparan sulfate chains present in basement membranes express the mAb 865 epitopes. Besides the normal distribution, mAb 865 staining was found in fibrotic and sclerotic lesions in vessels, interstitium, and mesangium in transplant kidneys with chronic vascular rejection. Occasionally, a decrease of staining was observed within tubulo-interstitium and glomeruli. These findings show that N-acetylated sequences in heparan sulfates can be demonstrated by anti-K5 mAb 865 in normal and diseased kidneys.
Collapse
Affiliation(s)
- J Born
- Division of Nephrology, University Hospital St. Radboud, 6500 HB Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
38
|
Whitelock JM, Murdoch AD, Iozzo RV, Underwood PA. The degradation of human endothelial cell-derived perlecan and release of bound basic fibroblast growth factor by stromelysin, collagenase, plasmin, and heparanases. J Biol Chem 1996; 271:10079-86. [PMID: 8626565 DOI: 10.1074/jbc.271.17.10079] [Citation(s) in RCA: 446] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Perlecan is a modular heparan sulfate proteoglycan that is localized to cell surfaces and within basement membranes. Its ability to interact with basic fibroblast growth factor (bFGF) suggests a central role in angiogenesis during development, wound healing, and tumor invasion. In the present study we investigated, using domain specific anti-perlecan monoclonal antibodies, the binding site of bFGF on human endothelial perlecan and its cleavage by proteolytic and glycolytic enzymes. The heparan sulfate was removed from perlecan by heparitinase treatment, and the approximately 450-kDa protein core was digested with various proteases. Plasmin digestion resulted in a large fragment of approximately 300 kDa, whereas stromelysin and rat collagenase cleaved the protein core into smaller fragments. All three proteases removed immunoreactivity toward the anti-domain I antibody. We showed also that perlecan bound bFGF specifically by the heparan sulfate chains located on the amino-terminal domain I. Once bound, the growth factor was released very efficiently by stromelysin, rat collagenase, plasmin, heparitinase I, platelet extract, and heparin. Interestingly, heparinase I, an enzyme with a substrate specificity for regions of heparan sulfate similar to those that bind bFGF, released only small amounts of bFGF. Our findings provide direct evidence that bFGF binds to heparan sulfate sequences attached to domain I and support the hypothesis that perlecan represents a major storage site for this growth factor in the blood vessel wall. Moreover, the concerted action of proteases that degrade the protein core and heparanases that remove the heparan sulfate may modulate the bioavailability of the growth factor.
Collapse
Affiliation(s)
- J M Whitelock
- Commonwealth Scientific and Industrial Research Organization, Division of Biomolecular Engineering, P.O. Box 184, North Ryde, Sydney, New South Wales 2114, Australia
| | | | | | | |
Collapse
|
39
|
Nishimura Y, Satoh T, Adachi H, Kondo S, Takeuchi T, Azetaka M, Fukuyasu H, Iizuka Y. The First l-Iduronic Acid-Type 1-N-Iminosugars Having Inhibitory Activity of Experimental Metastasis. J Am Chem Soc 1996. [DOI: 10.1021/ja953989b] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yoshio Nishimura
- Institute of Microbial Chemistry, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo 141, Japan Pharmaceutical Research Center, Meiji Seika Kaisha, Ltd. 760 Morooka-cho, Kohoku-ku, Yokohama 222, Japan
| | - Takahiko Satoh
- Institute of Microbial Chemistry, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo 141, Japan Pharmaceutical Research Center, Meiji Seika Kaisha, Ltd. 760 Morooka-cho, Kohoku-ku, Yokohama 222, Japan
| | - Hayamitsu Adachi
- Institute of Microbial Chemistry, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo 141, Japan Pharmaceutical Research Center, Meiji Seika Kaisha, Ltd. 760 Morooka-cho, Kohoku-ku, Yokohama 222, Japan
| | - Shinichi Kondo
- Institute of Microbial Chemistry, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo 141, Japan Pharmaceutical Research Center, Meiji Seika Kaisha, Ltd. 760 Morooka-cho, Kohoku-ku, Yokohama 222, Japan
| | - Tomio Takeuchi
- Institute of Microbial Chemistry, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo 141, Japan Pharmaceutical Research Center, Meiji Seika Kaisha, Ltd. 760 Morooka-cho, Kohoku-ku, Yokohama 222, Japan
| | - Masayuki Azetaka
- Institute of Microbial Chemistry, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo 141, Japan Pharmaceutical Research Center, Meiji Seika Kaisha, Ltd. 760 Morooka-cho, Kohoku-ku, Yokohama 222, Japan
| | - Harumi Fukuyasu
- Institute of Microbial Chemistry, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo 141, Japan Pharmaceutical Research Center, Meiji Seika Kaisha, Ltd. 760 Morooka-cho, Kohoku-ku, Yokohama 222, Japan
| | - Yumiko Iizuka
- Institute of Microbial Chemistry, 3-14-23 Kamiosaki Shinagawa-ku, Tokyo 141, Japan Pharmaceutical Research Center, Meiji Seika Kaisha, Ltd. 760 Morooka-cho, Kohoku-ku, Yokohama 222, Japan
| |
Collapse
|
40
|
Hershkoviz R, Marikovsky M, Gilat D, Lider O. Keratinocytes-associated chemokines and enzymatically quiescent heparanase induce the binding of resting CD4+ T cells. J Invest Dermatol 1996; 106:243-8. [PMID: 8601723 DOI: 10.1111/1523-1747.ep12340621] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Whether the chemokines macrophage inflammatory protein-1 beta (MIP-1 beta) and regulated on activation normal T expressed and secreted (RANTES), which interact specifically with glycosaminoglycans and thus mediate the recruitment, attachment, and migration of leukocytes to vascular endothelia and extracellular matrix, are also involved in interactions between CD4+ murine T lymphocytes and keratinocytes was examined. We have previously observed that depending on the local pH, a mammalian extracellular matrix-degrading enzyme, endo-beta-D glucuronidase (heparanase), which cleaves heparin sulfate proteoglycans, can function wither as an enzyme or as an adhesion molecule for CD4+ T lymphocytes. Herein, the involvement of heparanase in T cell-keratinocyte interactions was also probed. At 37 degree C and pH 7.2, radioactively labeled MIP-1 beta, RANTES, and heparanase bound to confluent layers of resting keratinocytes in a saturable and an heparan sulfate- or heparin-dependent manner, and thereby induced the adhesion of resting CD4+ T cells to keratinocytes. At a relatively acidic pH characteristic of inflammatory milieu, enzymatically active heparanase did not bind to the keratinocytes but, rather, inhibited the binding of MIP-1beta, RANTES, and the enzymatically quiescent heparanase to keratinocytes. These results suggest that certain chemokines and heparanase may function to restrict passing leukocytes, notable T lymphocytes, in the cutaneous micro-environment, a site which is continuously challenged with antigens. These keratinocyte-bound lymphocytes can serve as a reservoir of immediate responders to immunological stimuli.
Collapse
Affiliation(s)
- R Hershkoviz
- Department of Cell Biology, The Weizmann Institute of Science, Rehovot Israel
| | | | | | | |
Collapse
|
41
|
Chapter 1a Normal and pathological catabolism of glycoproteins. ACTA ACUST UNITED AC 1996. [DOI: 10.1016/s0167-7306(08)60278-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
42
|
Affiliation(s)
- B G Winchester
- Division of Biochemistry and Genetics, Institute of Child Health, London, United Kingdom
| |
Collapse
|
43
|
Sivaram P, Obunike JC, Goldberg IJ. Lysolecithin-induced alteration of subendothelial heparan sulfate proteoglycans increases monocyte binding to matrix. J Biol Chem 1995; 270:29760-5. [PMID: 8530367 DOI: 10.1074/jbc.270.50.29760] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The cause and consequence of altered proteoglycans in atherosclerosis are poorly understood. To determine whether proteoglycans affect monocyte binding, we studied the effects of heparin and proteoglycan degrading enzymes on THP-1 monocyte adhesion to subendothelial matrix (SEM). Monocyte binding increased about 2-fold after SEM was treated with heparinase. In addition, heparin decreased monocyte binding to fibronectin, a known SEM protein, by 60%. These data suggest that SEM heparan sulfate inhibits monocyte binding to SEM proteins. We next examined whether lysolecithin, a constituent of modified lipoproteins, affects endothelial heparan sulfate proteoglycan (HSPG) production and monocyte binding. Lysolecithin (10-200 microM) decreased total 35SO4 in SEM (20-75%). 2-fold more monocytes bound to SEM from lysolecithin treated cells than to control SEM. Heparinase treatment did not further increase monocyte binding to lysolecithin-treated SEM. HSPG degrading activity was found in medium from lysolecithin-treated but not control cells. 35SO4-labeled products obtained from labeled matrix treated with lysolecithin-conditioned medium were similar in size to those generated by heparinase. These data suggest that lysolecithin-treated endothelial cells secrete a heparanase-like activity. We hypothesize that decreased vessel wall HSPG, as occurs in atherogenic conditions, allows increased monocyte retention within the vessel and is due to the actions of an endothelial heparanase.
Collapse
Affiliation(s)
- P Sivaram
- Department of Medicine, Columbia University College of Physicians & Surgeons, New York, New York 10032, USA
| | | | | |
Collapse
|
44
|
Hoogewerf AJ, Leone JW, Reardon IM, Howe WJ, Asa D, Heinrikson RL, Ledbetter SR. CXC chemokines connective tissue activating peptide-III and neutrophil activating peptide-2 are heparin/heparan sulfate-degrading enzymes. J Biol Chem 1995; 270:3268-77. [PMID: 7852412 DOI: 10.1074/jbc.270.7.3268] [Citation(s) in RCA: 81] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Heparan sulfate proteoglycans at cell surfaces or in extracellular matrices bind diverse molecules, including growth factors and cytokines, and it is believed that the activities of these molecules may be regulated by the metabolism of heparan sulfate. In this study, purification of a heparan sulfate-degrading enzyme from human platelets led to the discovery that the enzymatic activity residues in at least two members of the platelet basic protein (PBP) family known as connective tissue activating peptide-III (CTAP-III) and neutrophil activating peptide-2. PBP and its N-truncated derivatives, CTAP-III and neutrophil activating peptide-2, are CXC chemokines, a group of molecules involved in inflammation and wound healing. SDS-polyacrylamide gel electrophoresis analysis of the purified heparanase resulted in a single broad band at 8-10 kDa, the known molecular weight of PBP and its truncated derivatives. Gel filtration chromatography of heparanase resulted in peaks of activity corresponding to monomers, dimers, and tetramers; these higher order aggregates are known to form among the chemokines. N-terminal sequence analysis of the same preparation indicated that only PBP and truncated derivatives were present, and commercial CTAP-III from three suppliers had heparanase activity. Antisera produced in animals immunized with a C-terminal synthetic peptide of PBP inhibited heparanase activity by 95%, compared with activity of the purified enzyme in the presence of the preimmune sera. The synthetic peptide also inhibited heparanase by 95% at 250 microM, compared to the 33% inhibition of heparanase activity by two other peptides. The enzyme was determined to be an endoglucosaminidase, and it degraded both heparin and heparan sulfate with optimal activity at pH 5.8. Chromatofocusing of the purified heparanase resulted in two protein peaks: an inactive peak at pI7.3, and an active peak at pI 4.8-5.1. Sequence analysis showed that the two peaks contained identical protein, suggesting that a post-translational modification activates the enzyme.
Collapse
Affiliation(s)
- A J Hoogewerf
- Units of Cancer & Infectious Disease, Upjohn Company, Kalamazoo, Michigan 49001
| | | | | | | | | | | | | |
Collapse
|
45
|
Ernst S, Langer R, Cooney CL, Sasisekharan R. Enzymatic degradation of glycosaminoglycans. Crit Rev Biochem Mol Biol 1995; 30:387-444. [PMID: 8575190 DOI: 10.3109/10409239509083490] [Citation(s) in RCA: 309] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Glycosaminoglycans (GAGs) play an intricate role in the extracellular matrix (ECM), not only as soluble components and polyelectrolytes, but also by specific interactions with growth factors and other transient components of the ECM. Modifications of GAG chains, such as isomerization, sulfation, and acetylation, generate the chemical specificity of GAGs. GAGs can be depolymerized enzymatically either by eliminative cleavage with lyases (EC 4.2.2.-) or by hydrolytic cleavage with hydrolases (EC 3.2.1.-). Often, these enzymes are specific for residues in the polysaccharide chain with certain modifications. As such, the enzymes can serve as tools for studying the physiological effect of residue modifications and as models at the molecular level of protein-GAG recognition. This review examines the structure of the substrates, the properties of enzymatic degradation, and the enzyme substrate-interactions at a molecular level. The primary structure of several GAGs is organized macroscopically by segregation into alternating blocks of specific sulfation patterns and microscopically by formation of oligosaccharide sequences with specific binding functions. Among GAGs, considerable dermatan sulfate, heparin and heparan sulfate show conformational flexibility in solution. They elicit sequence-specific interactions with enzymes that degrade them, as well as with other proteins, however, the effect of conformational flexibility on protein-GAG interactions is not clear. Recent findings have established empirical rules of substrate specificity and elucidated molecular mechanisms of enzyme-substrate interactions for enzymes that degrade GAGs. Here we propose that local formation of polysaccharide secondary structure is determined by the immediate sequence environment within the GAG polymer, and that this secondary structure, in turn, governs the binding and catalytic interactions between proteins and GAGs.
Collapse
Affiliation(s)
- S Ernst
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge 02139, USA
| | | | | | | |
Collapse
|
46
|
Thomson M. Endocytosis, partial degradation and release of heparan sulfate by elicited mouse peritoneal macrophages. Int J Biol Macromol 1994; 16:245-51. [PMID: 7893629 DOI: 10.1016/0141-8130(94)90029-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Interactions between glycosaminoglycans (GAGs) and low density lipoprotein (LDL) are thought to influence the progression of atherogenesis. In an effort to gauge whether macrophages mediate GAG-LDL interaction by GAG modification, we have investigated the endocytosis, degradation and retro-endocytosis of the GAG heparan sulfate (HS) by mouse peritoneal macrophages. Radiolabelled HS was produced by derivatization with sulfosuccinimidyl-3-(4-hydroxyphenyl) propionate and radio-iodination by the chloramine T method. The amount of 125I-HS internalized by cultures of thioglycollate-elicited macrophages rose over a 24 h time period in proportion to the amount of tracer added to the wells (2-2500 ng ml-1). Analysis of GAG molecular weight was performed using gel filtration chromatography and polyacrylamide gel electrophoresis. After a 24 h pulse period, the 125I-HS in the intracellular fraction of the cultured cells was of smaller molecular weight than for control material. During a 24 h cold chase, fragments of 125I-HS were released into the medium. These fragments had lower affinity for Polybrene-Sepharose but did not appear significantly N-desulfated as determined by low pH nitrous acid treatment. The NADPH oxidase inhibitor diphenylene iodonium, although minimizing basal and phorbol ester-triggered radical output, did not inhibit 125I-HS depolymerization. These data indicate that elicited macrophages can interact with and reduce the polymer length of HS without extensively desulfating the molecule. They are consistent with a mechanism by which the macrophage internalizes and partially degrades HS by endoglucuronidase activity rather than NADPH oxidase-generated free radicals, followed by release of the products into the extracellular milieu.
Collapse
Affiliation(s)
- M Thomson
- Heart Research Institute, Sydney, NSW, Australia
| |
Collapse
|
47
|
Epstein SE, Speir E, Unger EF, Guzman RJ, Finkel T. The basis of molecular strategies for treating coronary restenosis after angioplasty. J Am Coll Cardiol 1994; 23:1278-88. [PMID: 8176084 DOI: 10.1016/0735-1097(94)90368-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Excessive smooth muscle cell proliferation significantly contributes to restenosis, which occurs in 25% to 50% of patients within 6 months of coronary angioplasty. Because successful treatment will probably depend on our acquiring a comprehensive knowledge of the molecular and cellular mechanisms involved, this report reviews 1) information relevant to the molecular and cellular mechanisms responsible for the smooth muscle cell(s) response to vascular injury, and 2) several molecular-based therapeutic strategies currently being explored as possible approaches to the control of restenosis, including recombinant DNA technology to target delivery of cytotoxic molecules to proliferating smooth muscle cell(s), antisense strategies to inhibit expression of gene products necessary for cell proliferation and gene therapy.
Collapse
Affiliation(s)
- S E Epstein
- Cardiology Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland 20892
| | | | | | | | | |
Collapse
|
48
|
Sugahara K, Takemura Y, Sugiura M, Kohno Y, Yoshida K, Takeda K, Khoo KH, Morris HR, Dell A. Chondroitinase ABC-resistant sulfated trisaccharides isolated from digests of chondroitin/dermatan sulfate chains. Carbohydr Res 1994; 255:165-82. [PMID: 8181005 DOI: 10.1016/s0008-6215(00)90977-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Four kinds of sulfated trisaccharides resistant to chondroitinase ABC were isolated after chondroitinase B or ABC treatment of dermatan sulfate or various chondroitin sulfate isomers, respectively. Their composition was determined by chemical analysis and fast atom bombardment-mass spectrometry. Their structures were characterized by chondroitinase ACII digestion in conjunction with HPLC, and 500-MHz one- and two-dimensional 1H NMR spectroscopy. All the four trisaccharides have in common the core saccharide sequence, alpha-L-delta 4,5HexpA-(1-->3)-beta-D-GalpNAc-(1-->4)-D-GlcpA. A monosulfated component isolated from shark scapular cartilage chondroitin sulfate C or bovine aorta dermatan sulfate was elucidated as alpha-L-delta 4,5HexpA-(1-->3)-beta-D-GalpNAc6SO3(-)-(1-->4)-D-GlcpA or alpha-L-delta 4,5HexpA-(1-->3)-beta-D-GalpNAc4SO3(-)-(1-->4)-D-GlcpA , respectively. A disulfated component obtained from shark scapular cartilage chondroitin sulfate C or squid cartilage chondroitin sulfate E was identified as alpha-L-delta 4,5HexpA2SO3(-)-(1-->3)-beta-D-GalpNAc6SO3(-)-(1-->4)-D-G lcpA or alpha-L-delta 4,5HexpA-(1-->3)-beta-D-GalpNAc4SO3(-)6SO3(-)-(1-->4)- D-GlcpA, respectively. These trisaccharides are derived from the reducing termini of the parent polysaccharides. Some of the trisaccharides could be derived from the reducing termini exposed by the peeling reaction during the alkaline treatment while some others may represent the cleavage sites exposed by tissue endo-beta-D-glucuronidase(s), indicating the presence of such enzyme(s) which may release chondroitin/dermatan sulfate fragments from proteoglycans.
Collapse
Affiliation(s)
- K Sugahara
- Department of Physiological Chemistry, Kobe Women's College of Pharmacy, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
De Vouge MW, Yamazaki A, Bennett SA, Chen JH, Shwed PS, Couture C, Birnboim HC. Immunoselection of GRP94/endoplasmin from a KNRK cell-specific lambda gt11 library using antibodies directed against a putative heparanase amino-terminal peptide. Int J Cancer 1994; 56:286-94. [PMID: 8314313 DOI: 10.1002/ijc.2910560224] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Induction of an invasive phenotype by metastatic tumour cells results in part from inappropriate expression of extracellular matrix-degrading enzymes normally involved in embryonic morphogenesis, tissue remodelling, angiogenesis and wound healing. Such enzymes include endoglycosidases that degrade heparan sulfate (HS) in endothelial basement membrane, as well as better characterized proteases. Heparanase, an endo-beta-D-glucuronidase initially detected in B16 melanoma cells, has been described as a M(r) 96,000 glycoprotein with pI of 5.2, and has been immunolocalized to the cell surface and cytoplasm. We have utilized a polyacrylamide-gel-based HS degradation assay to demonstrate that KNRK, a rat kidney fibroblast cell line transformed by v-K-ras, exhibits HS-degrading activity similar to that of B16F10 mouse melanoma cells. To immunoselect heparanase-expressing clones from a KNRK-cell-specific lambda gt11 cDNA library, we have also prepared a rabbit anti-serum directed against a putative amino-terminal peptide of B16F10 cellular heparanase. Lysogens from one clone expressed a beta-galactosidase fusion protein whose staining with peptide anti-serum was inhibited by competition with excess peptide. Dideoxy-mediated sequencing of the insert termini of this recombinant revealed that it represents a rat homologue of M(r) 94,000 glucose-regulated protein (GRP94/endoplasmin), a molecular chaperone that contains the exact amino-terminal sequence previously attributed to heparanase. Our results call into question the specificity of this peptide sequence, as well as previous immunolocalization studies of heparanase carried out using such anti-sera.
Collapse
|
50
|
Ihrcke NS, Wrenshall LE, Lindman BJ, Platt JL. Role of heparan sulfate in immune system-blood vessel interactions. IMMUNOLOGY TODAY 1993; 14:500-5. [PMID: 8274190 DOI: 10.1016/0167-5699(93)90265-m] [Citation(s) in RCA: 151] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Heparan sulfate proteoglycan, a component of endothelial cell membranes and extracellular matrices, is involved in a number of the critical functions of endothelium and of antigen-presenting cells. This review discusses how heparan sulfate is released from endothelial cells during inflammation, how the loss of heparan sulfate potentially alters endothelial cell physiology, and how the presence of heparan sulfate in a soluble form might regulate the functioning of lymphocytes at sites of inflammation.
Collapse
Affiliation(s)
- N S Ihrcke
- Dept of Surgery, Duke University, Durham, NC 27710
| | | | | | | |
Collapse
|