1
|
Atienza J, Tkachyova I, Tropak M, Fan X, Schulze A. Fluorometric coupled enzyme assay for N-sulfotransferase activity of N-deacetylase/N-sulfotransferase (NDST). Glycobiology 2021; 31:1093-1101. [PMID: 34080004 DOI: 10.1093/glycob/cwab048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022] Open
Abstract
N-Deacetylase/N-sulfotransferases (NDST) are critical enzymes in heparan sulfate (HS) biosynthesis. Radioactive labeling assays are the preferred methods to determine the N-sulfotransferase activity of NDST. In this study, we developed a fluorometric coupled enzyme assay that is suitable for the study of enzyme kinetics and inhibitory properties of drug candidates derived from a large-scale in silico screening targeting the sulfotransferase moiety of NDST1. The assay measures recombinant mouse NDST1 (mNDST1) sulfotransferase activity by employing its natural substrate adenosine 3'-phophoadenosine-5'-phosphosulfate (PAPS), a bacterial analog of desulphated human HS, Escherichia coli K5 capsular polysaccharide (K5), the fluorogenic substrate 4-methylumbelliferylsulfate, and a double mutant of rat phenol sulfotransferase SULT1A1 K56ER68G. Enzyme kinetic analysis of mNDST1 performed with the coupled assay under steady state conditions at pH 6.8 and 37 °C revealed Km (K5) 34.8 μM, Km (PAPS) 10.7 μM, Vmax (K5) 0.53 ± 0.13 nmol/min/μg enzyme, Vmax (PAPS) 0.69 ± 0.05 nmol/min/μg enzyme, and the specific enzyme activity of 394 pmol/min/μg enzyme. The pH optimum of mNDST1 is pH 8.2. Our data indicate that mNDST1 is specific for K5 substrate. Finally, we showed that the mNDST1 coupled assay can be utilized to assess potential enzyme inhibitors for drug development.
Collapse
Affiliation(s)
- Joshua Atienza
- University of Toronto Scarborough, Toronto, Ontario, Canada.,Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Ilona Tkachyova
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Michael Tropak
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Xiaolian Fan
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Andreas Schulze
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario Canada.,Departments of Pediatrics and Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Lindsay SL, McCanney GA, Willison AG, Barnett SC. Multi-target approaches to CNS repair: olfactory mucosa-derived cells and heparan sulfates. Nat Rev Neurol 2020; 16:229-240. [PMID: 32099190 DOI: 10.1038/s41582-020-0311-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2020] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) remains one of the biggest challenges in the development of neuroregenerative therapeutics. Cell transplantation is one of numerous experimental strategies that have been identified and tested for efficacy at both preclinical and clinical levels in recent years. In this Review, we briefly discuss the state of human olfactory cell transplantation as a therapy, considering both its current clinical status and its limitations. Furthermore, we introduce a mesenchymal stromal cell derived from human olfactory tissue, which has the potential to induce multifaceted reparative effects in the environment within and surrounding the lesion. We argue that no single therapy will be sufficient to treat SCI effectively and that a combination of cell-based, rehabilitation and pharmaceutical interventions is the most promising approach to aid repair. For this reason, we also introduce a novel pharmaceutical strategy based on modifying the activity of heparan sulfate, an important regulator of a wide range of biological cell functions. The multi-target approach that is exemplified by these types of strategies will probably be necessary to optimize SCI treatment.
Collapse
Affiliation(s)
- Susan L Lindsay
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - George A McCanney
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Alice G Willison
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Susan C Barnett
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
3
|
Vlodavsky I, Sanderson RD, Ilan N. Non-Anticoagulant Heparins as Heparanase Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:493-522. [PMID: 32274724 PMCID: PMC7142274 DOI: 10.1007/978-3-030-34521-1_20] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The chapter will review early and more recent seminal contributions to the discovery and characterization of heparanase and non-anticoagulant heparins inhibiting its peculiar enzymatic activity. Indeed, heparanase displays a unique versatility in degrading heparan sulfate chains of several proteoglycans expressed in all mammalian cells. This endo-β-D-glucuronidase is overexpressed in cancer, inflammation, diabetes, atherosclerosis, nephropathies and other pathologies. Starting from known low- or non-anticoagulant heparins, the search for heparanase inhibitors evolved focusing on structure-activity relationship studies and taking advantage of new chemical-physical analytical methods which have allowed characterization and sequencing of polysaccharide chains. New methods to screen heparanase inhibitors and to evaluate their mechanism of action and in vivo activity in experimental models prompted their development. New non-anticoagulant heparin derivatives endowed with anti-heparanase activity are reported. Some leads are under clinical evaluation in the oncology field (e.g., acute myeloid leukemia, multiple myeloma, pancreatic carcinoma) and in other pathological conditions (e.g., sickle cell disease, malaria, labor arrest).
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
|
4
|
Tunicate Heparan Sulfate Enriched in 2-Sulfated β-Glucuronic Acid: Structure, Anticoagulant Activity, and Inhibitory Effect on the Binding of Human Colon Adenocarcinoma Cells to Immobilized P-Selectin. Mar Drugs 2019; 17:md17060351. [PMID: 31212795 PMCID: PMC6627333 DOI: 10.3390/md17060351] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 06/01/2019] [Accepted: 06/05/2019] [Indexed: 02/06/2023] Open
Abstract
Heparin or highly sulfated heparan sulfate (HS) has been described in different invertebrates. In ascidians (Chordata-Tunicata), these glycosaminoglycans occur in intracellular granules of oocyte accessory cells and circulating basophil-like cells, resembling mammalian mast cells and basophils, respectively. HS is also a component of the basement membrane of different ascidian organs. We have analyzed an HS isolated from the internal organs of the ascidian Phallusia nigra, using solution 1H/13C NMR spectroscopy, which allowed us to identify and quantify the monosaccharides found in this glycosaminoglycan. A variety of α-glucosamine units with distinct degrees of sulfation and N-acetylation were revealed. The hexuronic acid units occur both as α-iduronic acid and β-glucuronic acid, with variable sulfation at the 2-position. A peculiar structural aspect of the tunicate HS is the high content of 2-sulfated β-glucuronic acid, which accounts for one-third of the total hexuronic acid units. Another distinct aspect of this HS is the occurrence of high content of N-acetylated α-glucosamine units bearing a sulfate group at position 6. The unique ascidian HS is a potent inhibitor of the binding of human colon adenocarcinoma cells to immobilized P-selectin, being 11-fold more potent than mammalian heparin, but almost ineffective as an anticoagulant. Thus, the components of the HS structure required to inhibit coagulation and binding of tumor cells to P-selectin are distinct. Our results also suggest that the regulation of the pathway involved in the biosynthesis of glycosaminoglycans suffered variations during the evolution of chordates.
Collapse
|
5
|
Meneghetti MCZ, Gesteira Ferreira T, Tashima AK, Chavante SF, Yates EA, Liu J, Nader HB, Lima MA. Insights into the role of 3-O-sulfotransferase in heparan sulfate biosynthesis. Org Biomol Chem 2018; 15:6792-6799. [PMID: 28770943 DOI: 10.1039/c7ob01533j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
3-O-Sulfotransferase enzyme (sHS) from Litopenaeus vannamei was cloned and its substrate specificity was investigated against a number of GAG structures, including modified heparin polysaccharides and model oligosaccharides. For the heparin polysaccharides, derived from porcine intestinal mucosa heparin, sulfate groups were incorporated into glucosamine residues containing both N-sulfated and N-acetylated substitution within the regions of the predominant repeating disaccharide, either I-ANS or I-ANAc. However, the resulting polysaccharides did not stabilize antithrombin, which is correlated with anticoagulant activity. It was also shown that the enzyme was able to sulfate disaccharides, I2S-ANS and G-ANAc. The results further illustrate that 3-O-sulfation can be induced outside of the classical heparin-binding pentasaccharide sequence, show that 3-O-sulfation of glucosamine is not a sufficient condition for antithrombin stabilization and suggest that the use of this enzyme during HS biosynthesis may not occur as the final enzymatic step.
Collapse
|
6
|
Grad JN, Gigante A, Wilms C, Dybowski JN, Ohl L, Ottmann C, Schmuck C, Hoffmann D. Locating Large, Flexible Ligands on Proteins. J Chem Inf Model 2018; 58:315-327. [PMID: 29266929 DOI: 10.1021/acs.jcim.7b00413] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Many biologically important ligands of proteins are large, flexible, and in many cases charged molecules that bind to extended regions on the protein surface. It is infeasible or expensive to locate such ligands on proteins with standard methods such as docking or molecular dynamics (MD) simulation. The alternative approach proposed here is scanning of a spatial and angular grid around the protein with smaller fragments of the large ligand. Energy values for complete grids can be computed efficiently with a well-known fast Fourier transform-accelerated algorithm and a physically meaningful interaction model. We show that the approach can readily incorporate flexibility of the protein and ligand. The energy grids (EGs) resulting from the ligand fragment scans can be transformed into probability distributions and then directly compared to probability distributions estimated from MD simulations and experimental structural data. We test the approach on a diverse set of complexes between proteins and large, flexible ligands, including a complex of sonic hedgehog protein and heparin, three heparin sulfate substrates or nonsubstrates of an epimerase, a multibranched supramolecular ligand that stabilizes a protein-peptide complex, a flexible zwitterionic ligand that binds to a surface basin of a Kringle domain, and binding of ATP to a flexible site of an ion channel. In all cases, the EG approach gives results that are in good agreement with experimental data or MD simulations.
Collapse
Affiliation(s)
- Jean-Noël Grad
- Bioinformatics and Computational Biophysics, Faculty of Biology, University of Duisburg-Essen , Universitätstraße 7, 45141 Essen, Germany
| | - Alba Gigante
- Institute of Organic Chemistry, University of Duisburg-Essen , Universitätstraße 7, 45141 Essen, Germany
| | - Christoph Wilms
- Bioinformatics and Computational Biophysics, Faculty of Biology, University of Duisburg-Essen , Universitätstraße 7, 45141 Essen, Germany
| | - Jan Nikolaj Dybowski
- Bioinformatics and Computational Biophysics, Faculty of Biology, University of Duisburg-Essen , Universitätstraße 7, 45141 Essen, Germany
| | - Ludwig Ohl
- Bioinformatics and Computational Biophysics, Faculty of Biology, University of Duisburg-Essen , Universitätstraße 7, 45141 Essen, Germany
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering, and Institute for Complex Molecular Systems, Eindhoven University of Technology , Den Dolech 2, 5612 AZ Eindhoven, The Netherlands
| | - Carsten Schmuck
- Institute of Organic Chemistry, University of Duisburg-Essen , Universitätstraße 7, 45141 Essen, Germany
| | - Daniel Hoffmann
- Bioinformatics and Computational Biophysics, Faculty of Biology, University of Duisburg-Essen , Universitätstraße 7, 45141 Essen, Germany
| |
Collapse
|
7
|
Suleria HAR, Masci PP, Zhao KN, Addepalli R, Chen W, Osborne SA, Gobe GC. Anti-Coagulant and Anti-Thrombotic Properties of Blacklip Abalone (Haliotis rubra): In Vitro and Animal Studies. Mar Drugs 2017; 15:md15080240. [PMID: 28777290 PMCID: PMC5577595 DOI: 10.3390/md15080240] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/24/2017] [Accepted: 07/27/2017] [Indexed: 12/20/2022] Open
Abstract
Sulphated polysaccharides with anti-thrombotic and anti-coagulant activities have been found in various marine biota. In this study, a previously characterised anti-thrombotic and anti-coagulant extract from blacklip abalone was fractionated by anion exchange chromatography (AEC), pooled (on a sulphated polysaccharide basis) and administered to Wistar rats via oral gavage (N = 8) for assessment as an oral therapeutic. To ensure that the preparation had anti-coagulant activity prior to oral administration, it was assessed in rat blood by thromboelastography (TEG) significantly increasing reaction (R) time (or time until clot formation). Following in vitro confirmation of anti-coagulant activity, 40 mg of the preparation was orally administered to rats with blood samples collected at 2, 4, and 6 h post-gavage. Assessment of all blood samples by TEG showed some prolongation of R time from 355 to 380 s after 4 h. Dosing of the post-gavage blood samples with the abalone preparation to confirm anti-thrombotic activity in vitro revealed residual anti-coagulant activity, further suggesting that oral administration did increase anti-coagulant potential in the collected blood but that bioavailability was low. Assessment of tissues and haematological parameters showed no obvious harmful effects of the abalone preparation in animals. In summary, even though oral administration of fractionated and pooled blacklip abalone extract to rats delayed clotting after 4 h, bioavailability of the preparation appeared to be low and may be more appropriate for intravenous administration as an anti-thrombotic or anti-coagulant therapeutic.
Collapse
Affiliation(s)
- Hafiz Ansar Rasul Suleria
- UQ Diamantina Institute, Translational Research Institute, Faculty of Medicine, The University of Queensland, 37 Kent Street Woolloongabba, Brisbane, QLD 4102, Australia.
- CSIRO Agriculture and Food, 306 Carmody Road, St Lucia, Brisbane, QLD 4067, Australia.
| | - Paul P Masci
- UQ Diamantina Institute, Translational Research Institute, Faculty of Medicine, The University of Queensland, 37 Kent Street Woolloongabba, Brisbane, QLD 4102, Australia.
- Royal Brisbane & Women's Hospital Campus, University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, QLD 4029, Australia.
| | - Kong-Nan Zhao
- UQ Diamantina Institute, Translational Research Institute, Faculty of Medicine, The University of Queensland, 37 Kent Street Woolloongabba, Brisbane, QLD 4102, Australia.
- Royal Brisbane & Women's Hospital Campus, University of Queensland Centre for Clinical Research, The University of Queensland, Herston, Brisbane, QLD 4029, Australia.
| | - Rama Addepalli
- CSIRO Agriculture and Food, 306 Carmody Road, St Lucia, Brisbane, QLD 4067, Australia.
| | - Wei Chen
- CSIRO Agriculture and Food, 306 Carmody Road, St Lucia, Brisbane, QLD 4067, Australia.
| | - Simone A Osborne
- CSIRO Agriculture and Food, 306 Carmody Road, St Lucia, Brisbane, QLD 4067, Australia.
| | - Glenda C Gobe
- UQ Diamantina Institute, Translational Research Institute, Faculty of Medicine, The University of Queensland, 37 Kent Street Woolloongabba, Brisbane, QLD 4102, Australia.
| |
Collapse
|
8
|
Gallagher J. Fell-Muir Lecture: Heparan sulphate and the art of cell regulation: a polymer chain conducts the protein orchestra. Int J Exp Pathol 2015; 96:203-31. [PMID: 26173450 PMCID: PMC4561558 DOI: 10.1111/iep.12135] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/22/2015] [Indexed: 12/12/2022] Open
Abstract
Heparan sulphate (HS) sits at the interface of the cell and the extracellular matrix. It is a member of the glycosaminoglycan family of anionic polysaccharides with unique structural features designed for protein interaction and regulation. Its client proteins include soluble effectors (e.g. growth factors, morphogens, chemokines), membrane receptors and cell adhesion proteins such as fibronectin, fibrillin and various types of collagen. The protein-binding properties of HS, together with its strategic positioning in the pericellular domain, are indicative of key roles in mediating the flow of regulatory signals between cells and their microenvironment. The control of transmembrane signalling is a fundamental element in the complex biology of HS. It seems likely that, in some way, HS orchestrates diverse signalling pathways to facilitate information processing inside the cell. A dictionary definition of an orchestra is 'a large group of musicians who play together on various instruments …' to paraphrase, the HS orchestra is 'a large group of proteins that play together on various receptors'. HS conducts this orchestra to ensure that proteins hit the right notes on their receptors but, in the manner of a true conductor, does it also set 'the musical pulse' and create rhythm and harmony attractive to the cell? This is too big a question to answer but fun to think about as you read this review.
Collapse
Affiliation(s)
- John Gallagher
- Cancer Research UK Manchester Institute, Institute of Cancer Sciences, Paterson Building, University of Manchester, Manchester, UK
| |
Collapse
|
9
|
Pavão MSG. Glycosaminoglycans analogs from marine invertebrates: structure, biological effects, and potential as new therapeutics. Front Cell Infect Microbiol 2014; 4:123. [PMID: 25309878 PMCID: PMC4160087 DOI: 10.3389/fcimb.2014.00123] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 08/18/2014] [Indexed: 12/16/2022] Open
Abstract
In this review, several glycosaminoglycan analogs obtained from different marine invertebrate are reported. The structure, biological activity and mechanism of action of these unique molecules are detailed reviewed and exemplified by experiments in vitro and in vivo. Among the glycans studied are low-sulfated heparin-like polymers from ascidians, containing significant anticoagulant activity and no bleeding effect; dermatan sulfates with significant neurite outgrowth promoting activity and anti-P-selectin from ascidians, and a unique fucosylated chondroitin sulfate from sea cucumbers, possessing anticoagulant activity after oral administration and high anti P- and L-selectin activities. The therapeutic value and safety of these invertebrate glycans have been extensively proved by several experimental animal models of diseases, including thrombosis, inflammation and metastasis. These invertebrate glycans can be obtained in high concentrations from marine organisms that have been used as a food source for decades, and usually obtained from marine farms in sufficient quantities to be used as starting material for new therapeutics.
Collapse
Affiliation(s)
- Mauro S G Pavão
- Programa de Glicobiologia, Laboratório de Bioquímica e Biologia Cellular de Glicoconjugados, Instituto de Bioquímica Médica Leopoldo De Meis, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro Rio de Janeiro, Brazil
| |
Collapse
|
10
|
Mesenchymal stem cells, neural lineage potential, heparan sulfate proteoglycans and the matrix. Dev Biol 2014; 388:1-10. [DOI: 10.1016/j.ydbio.2014.01.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 01/08/2014] [Accepted: 01/30/2014] [Indexed: 12/23/2022]
|
11
|
Shang S, Zhu L, Fan J. Intermolecular interactions between natural polysaccharides and silk fibroin protein. Carbohydr Polym 2012; 93:561-73. [PMID: 23499097 DOI: 10.1016/j.carbpol.2012.12.038] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 12/06/2012] [Accepted: 12/14/2012] [Indexed: 10/27/2022]
Abstract
Fabricating novel functional and structural materials from natural renewable and degradable materials has attracted much attention. Natural polysaccharides and proteins are the right natural candidates due to their unique structures and properties. The polysaccharide-protein composites or blends were widely investigated, however, there are few systematical studies on the interactions between natural polysaccharides and silk fibroin protein at the molecular level. Among various interactions, hydrogen bonding, electrostatic interactions and covalent bonding play important roles in the structure and properties of the corresponding materials. Therefore, the focus is placed on the three interactions types in this review. A future challenge is to create polysaccharide and protein composites or blends with tailored structure and properties for the wide applications.
Collapse
Affiliation(s)
- Songmin Shang
- Institute of Textiles and Clothing, The Hong Kong Polytechnic University, Hong Kong.
| | | | | |
Collapse
|
12
|
Bouças RI, Jarrouge-Bouças TR, Lima MA, Trindade ES, Moraes FA, Cavalheiro RP, Tersariol IL, Hoppenstead D, Fareed J, Nader HB. Glycosaminoglycan backbone is not required for the modulation of hemostasis: Effect of different heparin derivatives and non-glycosaminoglycan analogs. Matrix Biol 2012; 31:308-16. [DOI: 10.1016/j.matbio.2012.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2012] [Revised: 03/10/2012] [Accepted: 03/23/2012] [Indexed: 11/26/2022]
|
13
|
Rudd TR, Yates EA. A highly efficient tree structure for the biosynthesis of heparan sulfate accounts for the commonly observed disaccharides and suggests a mechanism for domain synthesis. MOLECULAR BIOSYSTEMS 2012; 8:1499-506. [PMID: 22370609 DOI: 10.1039/c2mb25019e] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The form of the biosynthetic pathway of the biologically and medically important polysaccharides heparan sulfate (HS) and the closely related heparin remain obscure despite significant progress characterising the biosynthetic machinery. Considering possible biosynthetic schemes using a graph approach and applying known constraints of enzyme order and specificity, a previously unreported system with a highly efficient tree structure emerged with two features: (1) All commonly occurring HS disaccharides could be synthesised through a common route, the major branch. (2) The least common disaccharides also occurred on a separate common branch, termed here the minor branch. This suggested that the relative abundance of these two sets of structures were the result of the specificity of a single enzyme (HS epimerase) acting at an early point in the scheme, to convert GlcA-GlcNS to IdoA-GlcNS in preference to converting GlcA-GlcNAc to IdoA-GlcNAc. A third key finding was that the common substrates for 3-O-sulfation all lie on the same (major) branch. The proposed scheme is consistent with a wide body of experiments comprising both biochemical data and results from HS biosynthetic enzyme knockout experiments in the literature. The major branch also contains a bifurcation, providing a choice of two distinct backbone geometries with the same charge. Further development of this novel biosynthetic scheme, in which frame shifts in the site of action of the enzymes were permitted to occur, while maintaining their order of action, suggested a mechanism by which domains could be generated, or further modification blocked. The relationship between the proposed pathway and the geometric and charge possibilities it allows were also explored.
Collapse
Affiliation(s)
- Timothy R Rudd
- Istituto di Ricerche Chimiche e Biochimiche "G. Ronzoni", Via Giuseppe Colombo, 81 Milano 20133, Italy
| | | |
Collapse
|
14
|
Abstract
Heparan sulphate (HS), discovered in 1948 in heparin by-products, only emerged slowly from the shadow of heparin. Its inauspicious beginning was followed by the gradual realisation that HS was a separate entity with distinctive features. Both HS and heparin follow a common biosynthetic route but while heparin reaches full maturity, HS holds on to some of its youthful traits. The novel design and complex patterning of sulphation in HS enable it fulfil key roles in many, diverse biological processes.
Collapse
|
15
|
Abstract
New chemical-enzymatic technology based on the modification of the bacterial polysaccharide K5 from Escherichia coli leads to the synthesis of a number of heparin/heparan sulfate-like molecules with different biological activities. With this technology, two families of sulfated compounds were synthesized, which differ in their uronic acid content. The first group contains only glucuronic acid, whereas the second group contains about 50% iduronic acid following epimerization by immobilized recombinant C5 epimerase. This has led to the development of various anticoagulant and nonanticoagulant K5 derivatives endowed with different - and sometimes highly specific - antitumor, antiviral, and/or anti-inflammatory activities.
Collapse
Affiliation(s)
- P Oreste
- Glycores 2000 S.r.l., Milan, Italy.
| | | |
Collapse
|
16
|
Raman K, Nguyen TKN, Kuberan B. Is N-sulfation just a gateway modification during heparan sulfate biosynthesis? FEBS Lett 2011; 585:3420-3. [PMID: 21983289 DOI: 10.1016/j.febslet.2011.09.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 09/14/2011] [Accepted: 09/26/2011] [Indexed: 11/25/2022]
Abstract
Several biologically important growth factor-heparan sulfate (HS) interactions are regulated by HS sulfation patterns. However, the biogenesis of these combinatorial sulfation patterns is largely unknown. N-Deacetylase/N-sulfotrasferase (NDST) converts N-acetyl-d-glucosamine residues to N-sulfo-d-glucosamine residues. This enzyme is suggested to be a gateway enzyme because N-sulfation dictates the final HS sulfation pattern. It is known that O-sulfation blocks C5-epimerase, which acts immediately after NDST action. However, it is still unknown whether O-sulfation inhibits NDST action in a similar manner. In this article we radically change conventional assumptions regarding HS biosynthesis by providing in vitro evidence that N-sulfation is not necessarily just a gateway modification during HS biosynthesis.
Collapse
Affiliation(s)
- Karthik Raman
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
17
|
Li JP. Glucuronyl C5-epimerase an enzyme converting glucuronic acid to iduronic acid in heparan sulfate/heparin biosynthesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 93:59-78. [PMID: 20807641 DOI: 10.1016/s1877-1173(10)93004-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The glucuronyl C5 epimerase (HSepi) is one of the modification enzymes involved in biosynthesis of heparan sulfate (HS) and heparin, catalyzing the epimerization of D-glucuronic acid (GlcA) to L-iduronic acid (IdoA) at polymer level. IdoA is critical for HS and heparin to interact with protein ligands, because of its flexible conformation. Although the enzyme recognizes both GlcA and IdoA as substrates catalyzing a reversible reaction of the hexuronic acids in vitro, the reaction appears irreversible in vivo. Targeted interruption of the gene, Glce, in mice resulted in neonatal lethality accompanied with kidney agenesis, premature lung, and skeletal malformations, demonstrating that the single gene coded enzyme is essential for animal development. Elimination of the enzyme resulted in abnormal HS and heparin structure that completely lacks IdoA residues. Loss of 2-O-sulfation due to lacking IdoA in HS chains appears compensated by increased N- and O-sulfation of the glucosamine residues. Recombinant HSepi is used to generate HS/heparin related compounds having potential to be used for therapeutic purposes.
Collapse
Affiliation(s)
- Jin-ping Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
18
|
Boddohi S, Kipper MJ. Engineering nanoassemblies of polysaccharides. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2010; 22:2998-3016. [PMID: 20593437 DOI: 10.1002/adma.200903790] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Polysaccharides offer a wealth of biochemical and biomechanical functionality that can be used to develop new biomaterials. In mammalian tissues, polysaccharides often exhibit a hierarchy of structure, which includes assembly at the nanometer length scale. Furthermore, their biochemical function is determined by their nanoscale organization. These biological nanostructures provide the inspiration for developing techniques to tune the assembly of polysaccharides at the nanoscale. These new polysaccharide nanostructures are being used for the stabilization and delivery of drugs, proteins, and genes, the engineering of cells and tissues, and as new platforms on which to study biochemistry. In biological systems polysaccharide nanostructures are assembled via bottom-up processes. Many biologically derived polysaccharides behave as polyelectrolytes, and their polyelectrolyte nature can be used to tune their bottom-up assembly. New techniques designed to tune the structure and composition of polysaccharides at the nanoscale are enabling researchers to study in detail the emergent biological properties that arise from the nanoassembly of these important biological macromolecules.
Collapse
Affiliation(s)
- Soheil Boddohi
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, USA
| | | |
Collapse
|
19
|
Azab W, Tsujimura K, Maeda K, Kobayashi K, Mohamed YM, Kato K, Matsumura T, Akashi H. Glycoprotein C of equine herpesvirus 4 plays a role in viral binding to cell surface heparan sulfate. Virus Res 2010; 151:1-9. [DOI: 10.1016/j.virusres.2010.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Revised: 03/08/2010] [Accepted: 03/08/2010] [Indexed: 11/24/2022]
|
20
|
Abstract
Heparan sulfate proteoglycans (HSPGs) play vital roles in every step of tumor progression allowing cancer cells to proliferate, escape from immune response, invade neighboring tissues, and metastasize to distal sites away from the primary site. Several cancers including breast, lung, brain, pancreatic, skin, and colorectal cancers show aberrant modulation of several key HS biosynthetic enzymes such as 3-O Sulfotransferase and 6-O Sulfotransferase, and also catabolic enzymes such as HSulf-1, HSulf-2 and heparanase. The resulting tumor specific HS fine structures assist cancer cells to breakdown ECM to spread, misregulate signaling pathways to facilitate their proliferation, promote angiogenesis to receive nutrients, and protect themselves against natural killer cells. This review focuses on the changes in the expression of HS biosynthetic and catabolic enzymes in several cancers, the resulting changes in HS fine structures, and the effects of these tumor specific HS signatures on promoting invasion, proliferation, and metastasis. It is possible to retard tumor progression by modulating the deregulated biosynthetic and catabolic pathways of HS chains through novel chemical biology approaches.
Collapse
Affiliation(s)
- Karthik Raman
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112, USA
| | | |
Collapse
|
21
|
Glycosaminoglycans from earthworms (Eisenia andrei). Glycoconj J 2009; 27:249-57. [PMID: 20013352 DOI: 10.1007/s10719-009-9273-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2009] [Revised: 11/26/2009] [Accepted: 11/29/2009] [Indexed: 10/20/2022]
Abstract
The whole tissue of the earthworm (Eisenia andrei) was lyophilized and extracted to purify glycosaminoglycans. Fractions, eluting from an anion-exchange column at 1.0 M and 2.0 M NaCl, showed the presence of acidic polysaccharides on agarose gel electrophoresis. Monosaccharide compositional analysis showed that galactose and glucose were most abundant monosaccharides in both fractions. Depolymerization of the polysaccharide mixture with glycosaminoglycan-degrading enzymes confirmed the presence of chondroitin sulfate/dermatan sulfate and heparan sulfate in the 2.0 M NaCl fraction. The content of GAGs (uronic acid containing polysaccharide) in the 2.0 M NaCl fraction determined by carbazole assay was 2%. Disaccharide compositional analysis using liquid chromatography-electrospray ionization mass spectrometry (LC-ESI-MS) analysis after chondroitinase digestion (ABC and ACII), showed that the chondroitin sulfate/dermatan sulfate contained a 4-O-sulfo (76%), 2,4-di-O-sulfo (15%), 6-O-sulfo (6%), and unsulfated (4%) uronic acid linked N-acetylgalactosamine residues. LC-ESI-MS analysis of heparin lyase I/II/III digests demonstrated the presence of N-sulfo (69%), N-sulfo-6-O-sulfo (25%) and 2-O-sulfo-N-sulfo-6-O-sulfo (5%) uronic acid linked N-acetylglucosamine residues.
Collapse
|
22
|
Boddohi S, Killingsworth CE, Kipper MJ. Polyelectrolyte multilayer assembly as a function of pH and ionic strength using the polysaccharides chitosan and heparin. Biomacromolecules 2008; 9:2021-8. [PMID: 18564872 DOI: 10.1021/bm8002573] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The goal of this work is to explore the effects of solution ionic strength and pH on polyelectrolyte multilayer (PEM) assembly, using biologically derived polysaccharides as the polyelectrolytes. We used the layer-by-layer (LBL) technique to assemble PEM of the polysaccharides heparin (a strong polyanion) and chitosan (a weak polycation) and characterized the sensitivity of the PEM composition and layer thickness to changes in processing parameters. Fourier-transform surface plasmon resonance (FT-SPR) and spectroscopic ellipsometry provided in situ and ex situ measurements of the PEM thickness, respectively. Vibrational spectroscopy and X-ray photoelectron spectroscopy (XPS) provided details of the chemistry (i.e., composition, electrostatic interactions) of the PEM. We found that when PEM were assembled from 0.2 M buffer, the PEM thickness could be increased from less than 2 nm per bilayer to greater than 4 nm per bilayer by changing the solution pH; higher and lower ionic strength buffer solutions resulted in narrower ranges of accessible thickness. Molar composition of the PEM was not very sensitive to solution pH or ionic strength, but pH did affect the interactions between the sulfonates in heparin and amines in chitosan when PEM were assembled from 0.2 M buffer. Changes in the PEM thickness with pH and ionic strength can be interpreted through descriptions of the charge density and conformation of the polyelectrolyte chains in solution.
Collapse
Affiliation(s)
- Soheil Boddohi
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, Colorado, USA
| | | | | |
Collapse
|
23
|
Stevens RL, Adachi R. Protease-proteoglycan complexes of mouse and human mast cells and importance of their beta-tryptase-heparin complexes in inflammation and innate immunity. Immunol Rev 2007; 217:155-67. [PMID: 17498058 DOI: 10.1111/j.1600-065x.2007.00525.x] [Citation(s) in RCA: 112] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Approximately 50% of the weight of a mature mast cell (MC) consists of varied neutral proteases stored in the cell's secretory granules ionically bound to serglycin proteoglycans that contain heparin and/or chondroitin sulfate E/diB chains. Mouse MCs express the exopeptidase carboxypeptidase A3 and at least 15 serine proteases [designated as mouse MC protease (mMCP) 1-11, transmembrane tryptase/tryptase gamma/protease serine member S (Prss) 31, cathepsin G, granzyme B, and neuropsin/Prss19]. mMCP-6, mMCP-7, mMCP-11/Prss34, and Prss31 are the four members of the chromosome 17A3.3 family of tryptases that are preferentially expressed in MCs. One of the challenges ahead is to understand why MCs express so many different protease-proteoglycan macromolecular complexes. MC-like cells that contain tryptase-heparin complexes in their secretory granules have been identified in the Ciona intestinalis and Styela plicata urochordates that appeared approximately 500 million years ago. Because sea squirts lack B cells and T cells, it is likely that MCs and their tryptase-proteoglycan granule mediators initially appeared in lower organisms as part of their innate immune system. The conservation of MCs throughout evolution suggests that some of these protease-proteoglycan complexes are essential to our survival. In support of this conclusion, no human has been identified that lacks MCs. Moreover, transgenic mice lacking the beta-tryptase mMCP-6 are unable to combat a Klebsiella pneumoniae infection effectively. Here we summarize the nature and function of some of the tryptase-serglycin proteoglycan complexes found in mouse and human MCs.
Collapse
Affiliation(s)
- Richard L Stevens
- Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA.
| | | |
Collapse
|
24
|
Liu L, Xiang K, Zheng T, Zhang R, Li M, Li J. Co-inheritance of specific genotypes of HSPG and ApoE gene increases risk of type 2 diabetic nephropathy. Mol Cell Biochem 2004; 254:353-8. [PMID: 14674716 DOI: 10.1023/a:1027364121738] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The objective of this paper is to investigate co-inheritance of specific HSPG and ApoE genotypes in the development of Chinese type 2 diabetic nephropathy. PCR-RFLP was used to detect HSPG and ApoE genotypes in 385 Chinese subjects including 298 patients with type 2 diabetes mellitus (T2DM) and 87 non-diabetic controls (Non-DM). The T2DM group was subdivided into patients with (TDN; n = 218) and without diabetic nephropathy (Non-DN; n = 80). The latter group was further subdivided into groups of patients with microalbuminuria nephropathy (DN-1; n = 129) and severe diabetic nephropathy (DN-2; n = 89). We then compared the relative frequencies of various HSPG and ApoE genotypes and alleles among the groups, searching for predictive trends. The T allele of the HSPG gene occurred more frequently in the DN-2 group than in the Non-DN or DN-1 groups, their Fisher's exact p was 1.05 x 10(-3) and 6.58 x 10(-6); odds ratios were 2.09 (95% CI 1.32-3.30) and 2.48 (95% CI 1.64-3.74), respectively. The E2 allele of the ApoE gene occurred more frequently in the T2DM than in the Non-DM group, the Fisher's exact p was 0.0087; odds ratio was 3.45 (95% CI 1.30-9.81). Genotype analysis showed that the TT or TG of HSPG gene were paired with the E2/2 or E2/3 of ApoE gene significantly more frequently in the TDN group than in the Non-DN group, with an odds ratio of 3.03 (95% CI 1.03-8.90). There was no significant differences in other combinations of genotypes in HSPG and ApoE genes between TDN and Non-DN group. These results suggest that the HSPG T allele is a risk factor for the development of severe diabetic nephropathy in type 2 diabetic patients, and that the ApoE E2 allele is a risk factor for the occurrence of type 2 diabetes mellitus in Chinese general population. In addition, we find that co-inheritance of T/E2 confers a higher risk of type 2 diabetes mellitus progression to diabetic nephropathy in Chinese.
Collapse
Affiliation(s)
- Limei Liu
- Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, China.
| | | | | | | | | | | |
Collapse
|
25
|
Kisilevsky R, Szarek WA, Ancsin J, Bhat S, Li Z, Marone S. Novel glycosaminoglycan precursors as anti-amyloid agents, part III. J Mol Neurosci 2003; 20:291-7. [PMID: 14501011 DOI: 10.1385/jmn:20:3:291] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2002] [Accepted: 03/24/2003] [Indexed: 11/11/2022]
Abstract
In vivo amyloids consist of two classes of constituents. The first is the disease-defining protein, e.g., amyloid beta (Abeta) in Alzheimer's disease (AD). The second is a set of common structural components that usually are the building blocks of basement membrane (BM), a tissue structure that serves as a scaffold onto which cells normally adhere. In vitro binding interactions between one of these BM components and amyloidogenic proteins rapidly change the conformation of the amyloidogenic protein into amyloid fibrils. The offending BM component is a heparan sulfate (HS) proteoglycan (HSPG), part of which is protein, and the remainder is a specific linear polysaccharide that is the portion responsible for binding and imparting the typical amyloid structure to the amyloid precursor protein/peptide. Our past work has demonstrated that agents that inhibit the binding between HS and the amyloid precursor are effective antiamyloid compounds both in vitro and in vivo. Similarly, 4-deoxy analogs of glucosamine (a precursor of HS biosynthesis) are effective antiamyloid compounds both in culture and in vivo. Our continuing work concerns (1) the testing of our 4-deoxy compounds in a mouse transgenic model of AD, and (2) the continuing design and synthesis of modified sugar precursors of HS, which when incorporated into the polysaccharide will alter its structure so that it loses its amyloid-inducing properties. Since our previous report, 14 additional compounds have been designed and synthesized based on the known steps involved in HS biosynthesis. Of these, eight have been assessed for their effect on HS biosynthesis in hepatocyte tissue cultures, and the two anomers of a 4-deoxy-D-glucosamine analog have been assessed for their inflammation-associated amyloid (AA amyloid) inhibitory properties in vivo. The promising in vivo results with these two compounds have prompted studies using a murine transgenic model of brain Abeta amyloidogenesis. A macrophage tissue-culture model of AA amyloidogenesis has been devised based on the work of Kluve-Beckerman et al. and modified so as to assess compounds in the absence of potential in vivo confounding variables. Preliminary results indicate that the anomers of interest also inhibit AA amyloid deposition in macrophage tissue culture. Finally, an in vitro technique, using liver Golgi (the site of HS synthesis) rather than whole cells, has been devised to directly assess the effect of analogs on HS biosynthesis. The majority of the novel sugars prepared to date are analogs of N-acetylglucosamine. They have been modified either at the 2-N, C-3, C-4, or C-3 and C-4 positions. Results with the majority of the 2-N analogs suggest that hepacyte N-demethylases remove the N-substituent removal. Several of these have the desired effect on HS biosynthesis using hepatocyte cultures and will be assessed in the culture and in vivo AA amyloid models. To date 3-deoxy and 3,4-dideoxy analogs have failed to affect HS synthesis significantly. Compounds incorporating the 6-deoxy structural feature are currently being designed and synthesized.
Collapse
Affiliation(s)
- Robert Kisilevsky
- Department of Pathology, Queen's University, The Syl and Molly Apps Research Center Kingston General Hospital, Ontario, Canada.
| | | | | | | | | | | |
Collapse
|
26
|
Kim BT, Kitagawa H, Tanaka J, Tamura JI, Sugahara K. In vitro heparan sulfate polymerization: crucial roles of core protein moieties of primer substrates in addition to the EXT1-EXT2 interaction. J Biol Chem 2003; 278:41618-23. [PMID: 12907685 DOI: 10.1074/jbc.m304831200] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heparan, the common unsulfated precursor of heparan sulfate (HS) and heparin, is synthesized on the glycosaminoglycan-protein linkage region tetrasaccharide GlcUA-Gal-Gal-Xyl attached to the respective core proteins presumably by HS co-polymerases encoded by EXT1 and EXT2, the genetic defects of which result in hereditary multiple exostoses in humans. Although both EXT1 and EXT2 exhibit GlcNAc transferase and GlcUA transferase activities required for the HS synthesis, no HS chain polymerization has been demonstrated in vitro using recombinant enzymes. Here we report in vitro HS polymerization. Recombinant soluble enzymes expressed by co-transfection of EXT1 and EXT2 synthesized heparan polymers with average molecular weights greater than 1.7 x 105 using UDP-[3H]GlcNAc and UDP-GlcUA as donors on the recombinant glypican-1 core protein and also on the synthetic linkage region analog GlcUA-Gal-O-C2H4NH-benzyloxycarbonyl. Moreover, in our in vitro polymerization system, a part time proteoglycan, alpha-thrombomodulin, that is normally modified with chondroitin sulfate served as a polymerization primer for heparan chain. In contrast, no polymerization was achieved with a mixture of individually expressed EXT1 and EXT2 or with acceptor substrates such as N-acetylheparosan oligosaccharides or the linkage region tetrasaccharide-Ser, which are devoid of a hydrophobic aglycon, suggesting the critical requirement of core protein moieties in addition to the interaction between EXT1 and EXT2 for HS polymerization.
Collapse
Affiliation(s)
- Byung-Taek Kim
- Department of Biochemistry, Kobe Pharmaceutical University, 4-19-1 Motoyasmakita-machi, Higashinada-ku, Kobe 658-8558, Japan
| | | | | | | | | |
Collapse
|
27
|
Kisilevsky R, Szarek WA. Novel glycosaminoglycan precursors as anti-amyloid agents part II. J Mol Neurosci 2002; 19:45-50. [PMID: 12212792 DOI: 10.1007/s12031-002-0009-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2001] [Accepted: 10/16/2001] [Indexed: 10/23/2022]
Abstract
In vivo amyloids consist of two classes of constituents. The first is the disease defining protein, e.g., A beta in Alzheimer's disease. The second is a set of common structural components that usually are the building blocks of basement membrane (BM), a tissue structure that serves as a scaffold onto which cells normally adhere. In vitro binding interactions between one of these BM components and amyloidogenic proteins rapidly change the conformation of the amyloidogenic protein into amyloid fibrils. The offending BM component is a heparan sulfate (HS) proteoglycan (HSPG), part of which is protein and the remainder a specific linear polysaccharide, which is the portion responsible for binding, and imparting the typical amyloid structure, to the amyloid precursor protein/peptide. Our past work has demonstrated that agents that inhibit the binding between HS and the amyloid precursor are effective anti-amyloid compounds both in vitro and in vivo. The present work is concerned with the design and synthesis of modified sugar precursors of HS, which, when incorporated into the polysaccharide, will alter its structure so that it loses its amyloid precursor protein/peptide-binding and fibril-inducing properties. As part of our continuing study, since our previous report, 17 additional compounds have been designed and synthesized based primarily on the known steps involved in HS biosynthesis. In addition to the 4 reported last year, 10 more have been assessed in tissue culture for their inhibitory effect on heparan sulfate synthesis, and one of these has been assessed for its AA-amyloid inhibitory properties. The majority of the novel sugars are analogues of N-acetylglucosamine. They have been modified either at the 4-OH, 3-OH, or 2-N positions. The majority of the 2-N analogues provide data suggesting that hepatocyte N-demethylases remove the N-substituents converting the 2-N analogues into the natural sugar, a process that dilutes the D-[3H] glucosamine tracer used to track heparan sulfate synthesis and thereby gives the impression that biosynthetic inhibition is occurring. To date 3-deoxy analogues have failed to affect heparan sulfate synthesis significantly. Compounds incorporating the 3,4-dideoxy structural feature are currently being assessed. Using primary hepatocyte cultures, we reported previously that a 4-deoxy analogue is incorporated into HS and terminates its elongation. From the 4-deoxy series, one of the compounds has now been assessed in an in vivo model of AA-amyloid induction. This 4-deoxy analogue inhibited splenic AA amyloid deposition by at least 50%, and liver AA amyloid deposition by 85% when measured as amyloid/unit area of tissue. Furthermore, the spleen weights of the treated group were 1/2-1/3 of that in the untreated group indicating that the total splenic amyloid was 1/4-1/6 of that in the untreated group. The results provide further evidence that heparan sulfate is a critical factor in amyloidogenesis and modifications of sugar precursors of heparan sulfate synthesis may provide leads for therapeutic intervention in amyloidogenesis.
Collapse
Affiliation(s)
- Robert Kisilevsky
- Department of Pathology, Queen's University, Kingston, Ontario, Canada.
| | | |
Collapse
|
28
|
Raman R, Myette J, Venkataraman G, Sasisekharan V, Sasisekharan R. Identification of structural motifs and amino acids within the structure of human heparan sulfate 3-O-sulfotransferase that mediate enzymatic function. Biochem Biophys Res Commun 2002; 290:1214-9. [PMID: 11811992 DOI: 10.1006/bbrc.2001.6267] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In an accompanying paper [J. R. Myette, Z. Shriver, J. Liu, G. Venkataraman, and R. Sasisekharan (2002) Biochem. Biophys. Res. Commun. 290, 1206-1213], we described the purification and biochemical characterization of a soluble, recombinantly expressed form of the human heparan sulfate 3-O-sulfotransferase (3-OST-1). Such an important first step enables detailed structure-function studies for this class of enzymes. Herein, we describe a complimentary, structure-based homology modeling approach for predicting 3-OST-1 structure. This approach employs a variety of structural analysis and molecular modeling tools used in conjunction with protein crystallographic studies of related enzymes. In this manner, we describe important motifs within the predicted three-dimensional structure of the enzyme and identify specific amino acids that are likely important for enzymatic function.
Collapse
Affiliation(s)
- Rahul Raman
- Division of Bioengineering and Environmental Health, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA.
| | | | | | | | | |
Collapse
|
29
|
Myette JR, Shriver Z, Liu J, Venkataraman G, Rosenberg R, Sasisekharan R. Expression in Escherichia coli, purification and kinetic characterization of human heparan sulfate 3-O-sulfotransferase-1. Biochem Biophys Res Commun 2002; 290:1206-13. [PMID: 11811991 DOI: 10.1006/bbrc.2001.6268] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Heparan sulfate (HS) glycosaminoglycans are a structurally diverse class of complex biomolecules that modulate many important events at the cell surface and within the extracellular matrix and whose structural heterogeneity derives largely from the sequence-specific N- and O-sulfations catalyzed by an extensive repertoire of sulfating enzymes. We have expressed the human heparan sulfate 3-OST-1 isoform in Escherichia coli and subsequently purified a soluble, active enzyme. To assess its functionality, we determined the kinetic parameters for the recombinant 3-O-sulfotransferase-1 using a radiochemical assay that directly measures the 3-O-sulfation of unlabeled bovine kidney heparan sulfate in vitro using [(35)S]PAPS as the sulfate donor. The apparent K(m) values measured were in the low micromolar range (K(HS)(m) = 4.3 microM; K(PAPS)(m) = 38.6 microM); V(max) values of 18 and 21 pmol sulfate/min/pmol of enzyme for HS and PAPS, respectively. These values were compared with kinetic parameters likewise measured for recombinant 3-OST-1 purified from baculovirus-infected sf9 cells. The two enzymes appear to modify heparan sulfate in vitro to roughly the same extent and with comparable specificities. The expression of 3-OST-1 in E. coli represents an important step in subsequent structure-function studies.
Collapse
Affiliation(s)
- James R Myette
- Division of Bioengineering and Environmental Health, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | | | |
Collapse
|
30
|
Germi R, Crance JM, Garin D, Guimet J, Lortat-Jacob H, Ruigrok RWH, Zarski JP, Drouet E. Heparan sulfate-mediated binding of infectious dengue virus type 2 and yellow fever virus. Virology 2002; 292:162-8. [PMID: 11878919 DOI: 10.1006/viro.2001.1232] [Citation(s) in RCA: 175] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Dengue virus type 2 and Yellow fever virus are arthropod-borne flaviviruses causing hemorrhagic fever in humans. Identification of virus receptors is important in understanding flavivirus pathogenesis. The aim of this work was to study the role of cellular heparan sulfate in the adsorption of infectious Yellow fever and Dengue type 2 viruses. Virus attachment was assessed by adsorbing virus to cells, washing unbound virus away, releasing cell-bound virus by freezing/thawing, and then titrating the released infectious virus. Treatment of cells by heparin-lyase, desulfation of cellular heparan sulfate, or treatment of the virus with heparin inhibited cell binding of both viruses. Heparin also inhibited Yellow fever virus infection by 97%. Using infectious virus, the present work shows the importance of heparan sulfate in binding and infection of these two flaviviruses.
Collapse
Affiliation(s)
- Raphaële Germi
- Laboratoire de Virologie Moléculaire et Structurale EA 2939, Université Joseph Fourier, Grenoble, La Tronche, 38700, France
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Hallak LK, Collins PL, Knudson W, Peeples ME. Iduronic acid-containing glycosaminoglycans on target cells are required for efficient respiratory syncytial virus infection. Virology 2000; 271:264-75. [PMID: 10860881 DOI: 10.1006/viro.2000.0293] [Citation(s) in RCA: 174] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Respiratory syncytial virus (RSV) is an important human respiratory pathogen, particularly in infants. Glycosaminoglycans (GAGs) have been implicated in the initiation of RSV infection of cultured cells, but it is not clear what type of GAGs and GAG components are involved, whether the important GAGs are on the virus or the cell, or what the magnitude is of their contribution to infection. We constructed and rescued a recombinant green fluorescent protein (GFP)-expressing RSV (rgRSV) and used this virus to develop a sensitive system to assess and quantify infection by flow cytometry. Evaluation of a panel of mutant Chinese hamster ovary cell lines that are genetically deficient in various aspects of GAG synthesis showed that infection was reduced up to 80% depending on the type of GAG deficiency. Enzymatic removal of heparan sulfate and/or chondroitin sulfate from the surface of HEp-2 cells also reduced infection, and the removal of both reduced infection even further. Blocking experiments in which RSV was preincubated with various soluble GAGs revealed the relative blocking order of: heparin > heparan sulfate > chondroitin sulfate B. Iduronic acid is a component common to these GAGs. GAGs that do not contain iduronic acid, namely, chondroitin sulfate A and C and hyaluronic acid, did not inhibit infection. A role for iduronic acid-containing GAGs in RSV infection was confirmed by the ability of basic fibroblast growth factor to block infection, because basic fibroblast growth factor binds to GAGs containing iduronic acid. Pretreatment of cells with protamine sulfate, which binds and blocks GAGs, also reduced infection. In these examples, infection was reduced by pretreatment of the virus with soluble GAGs, pretreatment of the cells with GAG-binding molecules, pretreatment of the cells with GAG-destroying enzymes or in cells genetically deficient in GAGs. These results establish that the GAGs involved in RSV infection are present on the cell rather than on the virus particle. Thus, the presence of cell surface GAGs containing iduronic acid, like heparan sulfate and chondroitin sulfate B, is required for efficient RSV infection in cell culture.
Collapse
Affiliation(s)
- L K Hallak
- Immunology/Microbiology, Biochemistry and Pathology, Rush-Presbyterian-St. Luke's Medical Center, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
32
|
Mielke CH, Starr CM, Klock JC, Devereaux D, Mielke MR, Baker DE, Broemeling L, Wacksman M, White JR, Oliver SA, Ens G, Gavin P, Dittman WA. Direct measurement of unfractionated heparin using a biochemical assay. Clin Appl Thromb Hemost 1999; 5:267-76. [PMID: 10726025 DOI: 10.1177/107602969900500411] [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: 11/17/2022] Open
Abstract
A number of investigations have noted that functional biological assays for heparin are not always reliable and may not reflect the actual biochemical level of heparin in patients receiving anticoagulant therapy. This creates the possibility that patients receiving anticoagulant treatment may have an excess or deficiency of circulating levels of heparin. To address this problem, we have developed a direct biochemical measurement of heparin. The heparin assay uses fluorophore-assisted carbohydrate electrophoresis (FACE) to directly measure the predominate disaccharide of unfractionated heparin. In this study, unfractionated heparin was measured in vitro throughout a wide range of heparin concentrations in plasma. Seven in vivo pharmacokinetic studies in five normal subjects given 3,000 USP units of unfractionated heparin intravenously showed a three-phase elimination process with higher peak plasma levels and shorter elimination times than predicted from previous studies. At these doses, heparin is largely eliminated intact through urinary excretion. Body weight has a significant effect on heparin kinetics. When we compared the direct biochemical assay with two biological clotting assays, we found the latter can overestimate biochemical heparin concentrations. The FACE assay, due to its sensitivity, is also able to measure circulating levels of endogenous heparin in plasma and urine. Direct heparin measurement using the FACE technique is practical and useful for studies of the correlation of biochemical and biological activities.
Collapse
Affiliation(s)
- C H Mielke
- Health Research & Education Center, Washington State University, Spokane 99201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Hill WG, Harper GS, Rozaklis T, Hopwood JJ. Enhanced channelling of sulphate through a rapidly exchangeable sulphate pool in response to stimulated glycosaminoglycan synthesis in pancreatic epithelial cells. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1454:174-82. [PMID: 10381562 DOI: 10.1016/s0925-4439(99)00038-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The ability of cells to decorate glycosaminoglycans (GAGs) with sulphate in highly specific patterns is important to extracellular matrix biogenesis and placing appropriate glycosulphated ligands on the cell surface. We have examined sulphate metabolism in two pancreatic duct epithelial cell lines - PANC-1 and CFPAC-1 (derived from a cystic fibrosis patient) with a view to understanding how pancreatic cells utilise intracellular sulphate. [35S]Sulphate uptake was rapid and reached near steady state levels within 10 min. However, the intracellular specific activity of [35S]sulphate for PANC-1 and CFPAC-1 reached only 35 and 10%, respectively, of the medium specific activity at 10 min. Therefore, sulphate appears to reside within two compartments; a rapidly exchangeable sulphate pool (RESP) and a slowly exchangeable sulphate pool (SESP). Reducing chloride in the medium, increased the specific activity of [35S]sulphate within cells and increased the size of the inorganic sulphate pool, suggesting that the RESP was enlarged. Sulphate pools were not different in size between the two cell lines in physiological NaCl. Increasing the size of the sulphate pool had no effect on [35S]sulphate:[3H]glucosamine ratios incorporated into glycosaminoglycans (GAGs); however, stimulating the synthesis of GAGs with 4-methylumbelliferyl-beta-d-xyloside, stably elevated [35S]:[3H] ratios. This was due to higher [35S]sulphate incorporation. [35S]Cysteine contributed less than 0.1% of the cells' sulphate requirements. We conclude that in the face of elevated demand for sulphate, pancreatic cells appear to channel a greater proportion through the RESP.
Collapse
Affiliation(s)
- W G Hill
- Department of Chemical Pathology, Women's and Children's Hospital, Adelaide, SA 5006, Australia. whill+@pitt.edu
| | | | | | | |
Collapse
|
34
|
Zhang L, Yoshida K, Liu J, Rosenberg RD. Anticoagulant heparan sulfate precursor structures in F9 embryonal carcinoma cells. J Biol Chem 1999; 274:5681-91. [PMID: 10026187 DOI: 10.1074/jbc.274.9.5681] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To understand the mechanisms that control anticoagulant heparan sulfate (HSact) biosynthesis, we previously showed that HSact production in the F9 system is determined by the abundance of 3-O-sulfotransferase-1 as well as the size of the HSact precursor pool. In this study, HSact precursor structures have been studied by characterizing [6-3H]GlcN metabolically labeled F9 HS tagged with 3-O-sulfates in vitro by 3'-phosphoadenosine 5'-phospho-35S and purified 3-O-sulfotransferase-1. This later in vitro labeling allows the regions of HS destined to become the antithrombin (AT)-binding sites to be tagged for subsequent structural studies. It was shown that six 3-O-sulfation sites exist per HSact precursor chain. At least five out of six 3-O-sulfate-tagged oligosaccharides in HSact precursors bind AT, whereas none of 3-O-sulfate-tagged oligosaccharides from HSinact precursors bind AT. When treated with low pH nitrous or heparitinase, 3-O-sulfate-tagged HSact and HSinact precursors exhibit clearly different structural features. 3-O-Sulfate-tagged HSact hexasaccharides were AT affinity purified and sequenced by chemical and enzymatic degradations. The 3-O-sulfate-tagged HSact hexasaccharides exhibited the following structures, DeltaUA-[6-3H]GlcNAc6S-GlcUA-[6-3H]GlcNS3(35)S+/-6S-++ +IdceA2S-[6-3H]Glc NS6S. The underlined 6- and 3-O-sulfates constitute the most critical groups for AT binding in view of the fact that the precursor hexasaccharides possess all the elements for AT binding except for the 3-O-sulfate moiety. The presence of five potential AT-binding precursor hexasaccharides in all HSact precursor chains demonstrates for the first time the processive assembly of specific sequence in HS. The difference in structures around potential 3-O-sulfate acceptor sites in HSact and HSinact precursors suggests that these precursors might be generated by different concerted assembly mechanisms in the same cell. This study permits us to understand better the nature of the HS biosynthetic pathway that leads to the generation of specific saccharide sequences.
Collapse
Affiliation(s)
- L Zhang
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | |
Collapse
|
35
|
San Antonio JD, Verrecchio A, Pukac LA. Heparin sensitive and resistant vascular smooth muscle cells: biology and role in restenosis. Connect Tissue Res 1998; 37:87-103. [PMID: 9643650 DOI: 10.3109/03008209809028903] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Vascular smooth muscle cells (VSMC)s are characterized by their acute growth inhibition by heparin and heparan sulfates; however, recently the isolation of VSMCs which display greatly diminished sensitivity to the antiproliferative action of heparin have been reported. These heparin resistant (HR) VSMCs have been derived through multiple passage of normal rat VSMCs in culture media containing high heparin doses, by transformation of VSMCs with oncogene-containing vectors, or have been isolated from vascular tissues of spontaneously hypertensive rats, healthy humans, or humans with restenosis where their presence is not limited to sites of injury. Initial characterizations of HR VSMCs are reviewed, and here we propose a definition of HR VSMCs. To date the mechanisms underlying heparin insensitivity remain elusive. Further study of HR VSMCs may expand our understanding of cell growth regulation by heparin, establish whether HR VSMCs contribute to the reported failure of heparin to combat restenosis in humans, and identify cellular mechanisms driving certain vascular proliferative diseases.
Collapse
Affiliation(s)
- J D San Antonio
- Department of Medicine and the Cardeza Foundation for Hematologic Research, Jefferson Medical College of Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | | | | |
Collapse
|
36
|
Irokawa M, Nishinaga M, Ikeda U, Shinoda Y, Suematsu M, Gouda N, Ishimura Y, Shimada K. Endothelial-derived nitric oxide preserves anticoagulant heparan sulfate expression in cultured porcine aortic endothelial cells. Atherosclerosis 1997; 135:9-17. [PMID: 9395268 DOI: 10.1016/s0021-9150(97)00117-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Nitric oxide (NO) has been shown to inhibit platelet adhesion and aggregation, but there are no reports on its interaction with the coagulation system. We investigated the effect of the L-arginine analogues, N-nitro-L-arginine (LNA), N(G)-nitro-L-arginine methyl ester (L-NAME), and N(G)-monomethyl-L arginine (L-NMMA), competitive inhibitors of NO production, on endothelial-surface heparan sulfate. Addition of LNA to porcine aortic endothelial cells reduced 125I-labeled antithrombin III binding to the cell surface heparan sulfate in a dose- and time-dependent fashion. Significant inhibition was observed with 1 mM LNA, and the maximal suppression (-50% of control) occurred at 10 mM LNA after 12 h. L-NAME (1 mM) and L-NMMA (1 mM) also significantly inhibited the antithrombin III binding. The iron chelator desferrioxamine significantly prevented the reduction of antithrombin III binding to LNA-treated cells. We further investigated the effect of L-NAME on intracellular oxidative stress of endothelial cells using a hydroperoxide-sensitive fluorochrome, carboxy-dichloro-dihydrofluorescein diacetate bisacetoxymethyl ester probe, and revealed that inhibition of NO synthesis by L-NAME led to a marked increase in intracellular oxidative stress. These results demonstrated that the prolonged inhibition of NO synthesis in porcine aortic endothelial cells decreases the expression of anticoagulant heparan sulfate on endothelial cells through the increase in intracellular oxidative stress, perhaps comprising another mechanism by which NO affects the coagulation system in the vasculature.
Collapse
Affiliation(s)
- M Irokawa
- Department of Cardiology, Jichi Medical School, Minamikawachi-Machi, Tochigi, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Humphries DE, Sullivan BM, Aleixo MD, Stow JL. Localization of human heparan glucosaminyl N-deacetylase/N-sulphotransferase to the trans-Golgi network. Biochem J 1997; 325 ( Pt 2):351-7. [PMID: 9230113 PMCID: PMC1218567 DOI: 10.1042/bj3250351] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In order to determine the intracellular location of heparan N-deacetylase/N-sulphotransferase, cDNAs encoding human heparan glucosaminyl N-deacetylase/N-sulphotransferase were cloned from human umbilical vein endothelial cells. The deduced amino acid sequence was identical to that of the human heparan N-sulphotransferase cloned previously [Dixon, Loftus, Gladwin, Scambler, Wasmuth and Dixon (1995) Genomics 26, 239-244]. RNA blot analysis indicated that two heparan N-sulphotransferase transcripts of approx. 8.5 and 4 kb were produced in all tissues. Expression was most abundant in heart, liver and pancreas. A cDNA encoding a Flag-tagged human heparan N-sulphotransferase (where Flag is an epitope with the sequence DYKDDDDK) was transfected into mouse LTA cells. Immunofluorescence detection using anti-Flag monoclonal antibodies demonstrated that the enzyme was localized to the trans-Golgi network. A truncated Flag-tagged heparan N-sulphotransferase was also retained in the Golgi, indicating that, as for many other Golgi enzymes, the N-terminal region of heparan N-sulphotransferase is sufficient for retention in the Golgi apparatus.
Collapse
Affiliation(s)
- D E Humphries
- Department of Veterans Affairs Medical Center, Department of Medicine, Harvard Medical School, Boston, MA 02130, USA
| | | | | | | |
Collapse
|
38
|
Kobayashi M, Habuchi H, Yoneda M, Habuchi O, Kimata K. Molecular cloning and expression of Chinese hamster ovary cell heparan-sulfate 2-sulfotransferase. J Biol Chem 1997; 272:13980-5. [PMID: 9153262 DOI: 10.1074/jbc.272.21.13980] [Citation(s) in RCA: 107] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Heparan-sulfate 2-sulfotransferase (HS2ST), which catalyzes the transfer of sulfate from 3'-phosphoadenosine 5'-phosphosulfate to L-iduronic acid at position 2 in heparan sulfate, was purified from cultured Chinese hamster ovary (CHO) cells to apparent homogeneity (Kobayashi, M., Habuchi, H., Habuchi, O., Saito, M., and Kimata, K. (1996) J. Biol. Chem. 271, 7645-7653). The internal amino acid sequences were obtained from the peptides after digestion of the purified protein with a combination of endoproteinases. Mixed oligonucleotides based on the peptide sequences were used as primers to obtain a probe fragment by reverse transcriptase-polymerase chain reaction using CHO cell poly(A)+ RNA as template. The clone obtained from a CHO cDNA library by screening with the probe is 2.2 kilobases in size and contains an open reading frame of 1068 bases encoding a new protein composed of 356 amino acid residues. The protein predicts a type II transmembrane topology similar to other Golgi membrane proteins. Messages of 5.0 and 3.0 kilobases were observed in Northern analysis. Evidence that the cDNA clone corresponds to the purified HS2ST protein is as follows. (a) The predicted amino acid sequence contains all five peptides obtained after endoproteinase digestion of the purified protein; (b) the characteristics of the predicted protein fit those of the purified protein in terms of molecular mass, membrane localization, and N-glycosylation; and (c) when the cDNA containing the entire coding sequence of the enzyme in a eukaryotic expression vector was transfected into COS-7 cells, the HS2ST activity increased 2.6-fold over controls, and the FLAG-HS2ST fusion protein purified by affinity chromatography showed the HS2ST activity alone.
Collapse
Affiliation(s)
- M Kobayashi
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Aichi 480-11, Japan
| | | | | | | | | |
Collapse
|
39
|
Ernst S, Venkataraman G, Winkler S, Godavarti R, Langer R, Cooney CL, Sasisekharan R. Expression in Escherichia coli, purification and characterization of heparinase I from Flavobacterium heparinum. Biochem J 1996; 315 ( Pt 2):589-97. [PMID: 8615834 PMCID: PMC1217237 DOI: 10.1042/bj3150589] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The use of heparin for extracorporeal therapies has been problematical due to haemorrhagic complications; as a consequence, heparinase I from Flavobacterium heparinum is used for the determination of plasma heparin and for elimination of heparin from circulation. Here we report the expression of recombinant heparinase I in Escherichia coli, purification to homogeneity and characterization of the purified enzyme. Heparinase I was expressed with an N-terminal histidine tag. The enzyme was insoluble and inactive, but could be refolded, and was purified to homogeneity by nickel-chelate chromatography. The cumulative yield was 43%, and the recovery of purified heparinase I was 14.4 mg/l of culture. The N-terminal sequence and the molecular mass as analysed by matrix-assisted laser desorption MS were consistent with predictions from the heparinase I gene structure. The reverse-phase HPLC profile of the tryptic digest, the Michaelis-Menten constant Km (47 micrograms/ml) and the specific activity (117 units/mg) of purified recombinant heparinase I were similar to those of the native enzyme. Degradation of heparin by heparinase I results in a characteristic product distribution, which is different from those obtained by degradation with heparinase II or III from F. heparinum. We developed a rapid anion-exchange HPLC method to separate the products of enzymic heparin degradation, using POROS perfusion chromatography media. Separation of characteristic di-, tetra- and hexa-saccharide products is performed in 10 min. These methods for the expression, purification and analysis of recombinant heparinase I may facilitate further development of heparinase I-based medical therapies as well as further investigation of the structures of heparin and heparan sulphate and their role in the extracellular matrix.
Collapse
Affiliation(s)
- S Ernst
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge 02139, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
van den Born J, van Kraats AA, Bakker MA, Assmann KJ, Dijkman HB, van der Laak JA, Berden JH. Reduction of heparan sulphate-associated anionic sites in the glomerular basement membrane of rats with streptozotocin-induced diabetic nephropathy. Diabetologia 1995; 38:1169-75. [PMID: 8690168 DOI: 10.1007/bf00422365] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Heparan sulphate-associated anionic sites in the glomerular basement membrane were studied in rats 8 months after induction of diabetes by streptozotocin and in age- adn sex-matched control rats, employing the cationic dye cuprolinic blue. Morphometric analysis at the ultrastructural level was performed using a computerized image processor. The heparan sulphate specificity of the cuprolinic blue staining was demonstrated by glycosaminoglycan-degrading enzymes, showing that pretreatment of the sections with heparitinase abolished all staining, whereas chondroitinase ABC had no effect. The majority of anionic sites (74% in diabetic and 81% in control rats) were found within the lamina rara externa of the glomerular basement membrane. A minority of anionic sites were scattered throughout the lamina densa and lamina rara interna, and were significantly smaller than those in the lamina rara externa of the glomerular basement membrane (p<0.001 and p<0.01 for diabetic and control rats, respectively). Diabetic rats progressively developed albuminuria reaching 40.3 (32.2-62.0) mg/24 h after 8 months in contrast to the control animals (0.8 (0.2-0.9) mg/24 h, p<0.002). At the same time, the number of heparan sulphate anionic sites and the total anionic site surface (number of anionic sites x mean anionic site surface) in the lamina rara externa of the glomerular basement membrane was reduced by 19% (p<0.021) and by 26% (p<0.02), respectively. Number and total anionic site surface in the remaining part of the glomerular basement membrane (lamina densa and lamina rara interna) were not significantly changed. We conclude that in streptozotocin-diabetic rats with an increased urinary albumin excretion, a reduced heparan sulphate charge barrier/density is found at the lamina rara externa of the glomerular basement membrane.
Collapse
Affiliation(s)
- J van den Born
- Department of Nephrology, University Hospital St. Radboud, Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
41
|
Thomas SS, Plenkiewicz J, Ison ER, Bols M, Zou W, Szarek WA, Kisilevsky R. Influence of monosaccharide derivatives on liver cell glycosaminoglycan synthesis: 3-deoxy-D-xylo-hexose (3-deoxy-D-galactose) and methyl (methyl 4-chloro-4-deoxy-beta-D-galactopyranosid) uronate. BIOCHIMICA ET BIOPHYSICA ACTA 1995; 1272:37-48. [PMID: 7662718 DOI: 10.1016/0925-4439(95)00065-c] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
An improved, convenient synthesis of 3-deoxy-D-xylo-hexose (3-deoxy-D-galactose) has been developed, and the chemical synthesis of a novel monosaccharide derivative, methyl (methyl 4-chloro-4-deoxy-beta-D-galactopyranosid)uronate (compound 10), is described. Using primary hepatocytes in culture, each was used to explore its effect on glycosaminoglycan (GAG) synthesis. In the absence of analogues hepatocytes synthesize primarily (92-95%) heparan sulphate. At 1 mM, 3-deoxy-D-galactose had little observable effect on either liver cell GAG or protein synthesis. At 10 mM and 20 mM, 3-deoxy-D-galactose reduced [3H]glucosamine and 35SO4 incorporation into hepatocyte cellular GAGs to, respectively, 75% and 60% of the control cells. This inhibition of GAG synthesis occurred without any effect on hepatocyte protein synthesis, indicating that 3-deoxy-D-galactose's effect on GAG synthesis is not mediated through an inhibition of proteoglycan core protein synthesis. Furthermore, GAGs in the presence of 20 mM of the analogue were significantly reduced in size, 17 kDa vs. 66 kDa in untreated cells. These results reflect either impaired cellular GAG chain elongation, and/or altered GAG chain degradation. Compound 10 exhibited a concentration-dependent inhibition of both hepatocyte cellular GAG and protein synthesis. At concentrations of 5, 10 and 20 mM, compound 10 inhibited GAG and protein synthesis by 20, 65 and 90%, respectively. Exogenous uridine was able to restore partially the inhibition of protein synthesis, but was unable to reverse the effect of compound 10 on GAG synthesis. These results show that part of the effect of compound 10 on GAG synthesis is not mediated by an inhibition of proteoglycan core protein synthesis. GAGs in the presence of compound 10 are half as large as those in the absence of this compound (33 and 66 kDa, respectively). These results again may reflect either impaired cellular GAG chain elongation and/or altered GAG chain degradation. Potential metabolic routes for each analogue's effect are presented.
Collapse
Affiliation(s)
- S S Thomas
- Department of Biochemistry, Queen's University, Kingston, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
42
|
van den Born J, van Kraats AA, Bakker MA, Assmann KJ, van den Heuvel LP, Veerkamp JH, Berden JH. Selective proteinuria in diabetic nephropathy in the rat is associated with a relative decrease in glomerular basement membrane heparan sulphate. Diabetologia 1995; 38:161-72. [PMID: 7713310 DOI: 10.1007/bf00400090] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
In the present study we investigated whether glomerular hyperfiltration and albuminuria in streptozotocin-induced diabetic nephropathy in male Wistar-Münich rats are associated with changes in the heparan sulphate content of the glomerular basement membrane. Rats with a diabetes mellitus duration of 8 months, treated with low doses of insulin, showed a significant increase in glomerular filtration rate (p < 0.01) and effective renal plasma flow (p < 0.05), without alterations in filtration fraction or mean arterial blood pressure. Diabetic rats developed progressive albuminuria (at 7 months, diabetic rats (D): 42 +/- 13 vs control rats (C): 0.5 +/- 0.2 mg/24 h, p < 0.002) and a decrease of the selectivity index (clearance IgG/clearance albumin) of the proteinuria (at 7 months, D: 0.20 +/- 0.04 vs C: 0.39 +/- 0.17, p < 0.05), suggesting loss of glomerular basement membrane charge. Light- and electron microscopy demonstrated a moderate increase of mesangial matrix and thickening of the glomerular basement membrane in the diabetic rats. Immunohistochemically an increase of laminin, collagen III and IV staining was observed in the mesangium and in the glomerular basement membrane, without alterations in glomerular basement membrane staining of heparan sulphate proteoglycan core protein or heparan sulphate. Glomerular basement membrane heparan sulphate content, quantitated in individual glomerular extracts by a new inhibition ELISA using a specific anti-glomerular basement membrane heparan sulphate monoclonal antibody (JM403), was not altered (median (range) D: 314 (152-941) vs C: 262 (244-467) ng heparan sulphate/mg glomerulus). However, the amount of glomerular 4-hydroxyproline, as a measure for collagen content, was significantly increased (D: 1665 (712-2014) vs C: 672 (515-1208) ng/mg glomerulus, p < 0.01). Consequently, a significant decrease of the heparan sulphate/4-hydroxyproline ratio (D: 0.21 (0.14-1.16) vs C: 0.39 (0.30-0.47), p < 0.05) was found. In summary, we demonstrate that in streptozotocin-diabetic rats glomerular hyperfiltration and a progressive, selective proteinuria are associated with a relative decrease of glomerular basement membrane heparan sulphate. Functionally, a diminished heparan sulphate-associated charge density within the glomerular basement membrane might explain the selective proteinuria in the diabetic rats.
Collapse
Affiliation(s)
- J van den Born
- Department of Nephrology, University Hospital St. Radbound, Nijmegen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
43
|
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
|
44
|
Shilatifard A, Cummings RD. Purification and characterization of N-acetylglucosamine-6-sulfate sulfatase from bovine kidney: evidence for the presence of a novel endosulfatase activity. Biochemistry 1994; 33:4273-82. [PMID: 8155645 DOI: 10.1021/bi00180a023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
N-Acetylglucosamine-6-sulfate sulfatase (NG6SS) is an enzyme that catalyzes the hydrolysis of sulfate esters from the C-6 hydroxyl of N-acetylglucosamine. We report our purification and characterization of the enzyme and the discovery that it can remove sulfate from internally sulfated GlcNAc on glycopeptides and glycoproteins. The enzyme was purified from bovine kidney over 200,000-fold using a combination of ion-exchange and size-exclusion chromatography. NG6SS is soluble and occurs as a single subunit with apparent solution molecular weight of 60.2 kDa on gel filtration chromatography and approximately 52.5 and 57.8 kDa on reducing and nonreducing SDS/PAGE, respectively. The enzyme is highly basic and exhibits a broad pH range with an optimum at pH 6.5 and a temperature optimum of 37 degrees C. Among the mono- and disaccharide sulfates tested, only GlcNAc-6-SO4 is an effective substrate with a Km of 4.7 mM, and either free sulfate or phosphate inhibits the activity. Unexpectedly, we found that the enzyme displays endosulfatase activity and quantitatively releases 35SO4 from 35SO4-labeled glycopeptides and intact glycoproteins isolated from human Molt-3 cells, which we have previously shown to synthesize glycoproteins containing GlcNAc-6-SO4 residues within the sequence Gal beta 1-4[SO-3-6]-GlcNAc beta 1-R of complex-type N-linked oligosaccharides. The N-terminal sequence of the bovine NG6SS was homologous to a human-liver-derived N-acetylglucosamine-6-sulfatase. The endosulfatase activity of bovine kidney NG6SS may be important in its potential role in the degradation of sulfated glycans and may make this enzyme a valuable reagent to study the biological functions of sulfated glycoproteins.
Collapse
Affiliation(s)
- A Shilatifard
- Department of Biochemistry and Molecular Biology, Oklahoma Center for Molecular Medicine, University of Oklahoma Health Sciences Center, Oklahoma City 73104
| | | |
Collapse
|
45
|
Molecular cloning and expression of a glycosaminoglycan N-acetylglucosaminyl N-deacetylase/N-sulfotransferase from a heparin-producing cell line. J Biol Chem 1994. [DOI: 10.1016/s0021-9258(17)42164-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
46
|
Rapraeger AC, Guimond S, Krufka A, Olwin BB. Regulation by heparan sulfate in fibroblast growth factor signaling. Methods Enzymol 1994; 245:219-40. [PMID: 7760735 DOI: 10.1016/0076-6879(94)45013-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The integral role of heparan sulfate proteoglycans in FGF signaling provides a potential means of regulating FGF activity. This regulation may be used by the cell, where the modification of heparan sulfate glycosaminoglycans during their synthesis in the Golgi can produce cell type- and potentially ligand-specific sulfation sequences. The description of these sequences will not only provide information on how this regulation is achieved, perhaps lending insight into other heparan sulfate-ligand interactions, but may also discern sulfated mimetics that can be used to disrupt or alter FGF signaling. These mimetics may be useful in the treatment disrupt or alter FGF signaling. These mimetics may be useful in the treatment of disease, or in understanding how FGF signaling via discrete pathways within the cell leads to specific cellular responses, such as activation of mitogenic signaling pathways, calcium fluxes, and cellular differentiation.
Collapse
Affiliation(s)
- A C Rapraeger
- Department of Pathology and Laboratory Medicine, University of Wisconsin, Madison, 53706, USA
| | | | | | | |
Collapse
|
47
|
Nishinaga M, Ozawa T, Shimada K. Homocysteine, a thrombogenic agent, suppresses anticoagulant heparan sulfate expression in cultured porcine aortic endothelial cells. J Clin Invest 1993; 92:1381-6. [PMID: 8376590 PMCID: PMC288280 DOI: 10.1172/jci116712] [Citation(s) in RCA: 178] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Previous studies showed that homocysteine, a thrombo-atherogenic and atherogenic agent, inhibits an endothelial thrombomodulin-protein C anticoagulant pathway. We examined whether homocysteine might affect another endothelial anticoagulant mechanism; i.e., heparin-like glycosaminoglycan-antithrombin III interactions. Incubations of porcine aortic endothelial cell cultures with homocysteine reduced the amount of antithrombin III bound to the cell surface in a dose- and time-dependent fashion. The inhibitory effect was observed at a homocysteine concentration as low as 0.1 mM, and the maximal suppression occurred at 1 mM of homocysteine after 24 h. In contrast with a marked reduction in the maximal antithrombin III binding capacity (approximately 30% of control), the radioactivity of [35S]sulfate incorporated into heparan sulfate on the cell surface was minimally (< 15%) reduced. The cells remained viable after homocysteine treatment. Although neither net negative charge nor proportion in total glycosaminoglycans of cell surface heparan sulfate was altered by homocysteine treatment, a substantial reduction in antithrombin III binding capacity of heparan sulfate isolated from homocysteine-treated endothelial cells was found using both affinity chromatography and dot blot assay techniques. The antithrombin III binding activity of endothelial cells decreased after preincubation with 1 mM homocysteine, cysteine, or 2-mercaptoethanol; no reduction in binding activity was observed after preincubation with the same concentration of methionine, alanine, or valine. This sulfhydryl effect may be caused by generation of hydrogen peroxide, as incubation of catalase, but not superoxide dismutase, with homocysteine-treated endothelial cells prevented this reduction, whereas copper augmented the inhibitory effects of the metabolite. Thus, our data suggest that the inhibited expression of anticoagulant heparan sulfate may contribute to the thrombogenic property resulting from the homocysteine-induced endothelial cell perturbation, mediated by generation of hydrogen peroxide through alteration of the redox potential.
Collapse
Affiliation(s)
- M Nishinaga
- Department of Cardiology, Jichi Medical School, Tochigi, Japan
| | | | | |
Collapse
|
48
|
Kofoed-Enevoldsen A, Petersen JS, Deckert T. Glucosaminyl N-deacetylase in cultured fibroblasts; comparison of patients with and without diabetic nephropathy, and identification of a possible mechanism for diabetes-induced N-deacetylase inhibition. Diabetologia 1993; 36:536-40. [PMID: 8335176 DOI: 10.1007/bf02743270] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Impaired heparan sulphate biosynthesis through diabetes-induced inhibition of glucosaminyl N-deacetylase may have a central role in the development of diabetic nephropathy, and genetic differences in the vulnerability of the N-deacetylase could influence the risk of developing nephropathy. We studied N-deacetylase activity in fibroblast cultures from Type 1 (insulin-dependent) diabetic patients with (n = 14) or without (n = 13) diabetic nephropathy, together with non-diabetic control subjects (n = 7). No difference in N-deacetylase activity was found (p = 0.13), and no inhibition of N-deacetylase was found in cultures grown at 25 mmol/l glucose. N-deacetylase activity was inversely correlated to growth rate (r = -0.59, p = 0.0008), and in patients with nephropathy a negative correlation between HbA1C and fibroblast N-deacetylase activity (r = -0.72, p = 0.012) was found. Cell-cycle analysis revealed an increased fraction of S-phase cells in patients with nephropathy (28%(21-52%)) compared to healthy control subjects (17% (9-24%)), p = 0.0008, but not between patients with and without nephropathy (latter group 26%(11-43%)), p = 0.43. Forskolin, an activator of protein kinase A, specifically decreased N-deacetylase activity, whereas activation of protein kinase C produced a combined reduction in N-deacetylase activity and total protein synthesis. In conclusion, no constitutive defects in N-deacetylase activity were found in fibroblasts from these patients. Further studies should consider possible associations between fibroblast characteristics and pre-biopsy environmental parameters related to cellular memory phenomena. Finally, activation of protein kinase A provides a potential general pathway for regulating N-deacetylase activity.
Collapse
|
49
|
Rohde LH, Carson DD. Heparin-like glycosaminoglycans participate in binding of a human trophoblastic cell line (JAR) to a human uterine epithelial cell line (RL95). J Cell Physiol 1993; 155:185-96. [PMID: 8468365 DOI: 10.1002/jcp.1041550124] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In vitro studies in our laboratory have indicated that heparan sulfate proteoglycans (HSPGs) play an important role in murine embryo implantation. In order to investigate the potential function of HSPGs in human implantation, two human cell lines (RL95 and JAR) were used to model uterine epithelium and embryonal trophectoderm, respectively. A heterologous cell-cell adhesion assay was developed to determine if binding of JAR cells to RL95 cells was heparan sulfate-dependent. Labeled, single cell suspensions of JAR cells attached to confluent monolayers of RL95 cells in a dose- and time-dependent manner. Heparin-like glycosaminoglycans and JAR cell proteoglycans competitively inhibited JAR cell adhesion to RL95 cells by 50% or more. A panel of chemically modified heparins were used to demonstrate that O-sulfation and amino group substitution were critical for inhibition of cell-cell adhesion. Treatment with chlorate, an inhibitor of ATP-sulfurylase, resulted in a 56% reduction in cell-cell binding compared to untreated controls. Heparinase and chondroitinase ABC markedly inhibited JAR-RL95 binding, while chondroitinase AC had no significant effect. These observations indicated that HSPGs as well as dermatan sulfate-containing proteoglycans participated in cell-cell binding. Collectively, these results indicate that initial binding interactions between JAR and RL95 cells is mediated by cell surface glycosaminoglycans (GAGs) with heparin-like properties (i.e., heparan sulfate and dermatan sulfate). These observations are consistent with an important role for HS and heparin-like GAGs as well as their corresponding binding sites in early stages of human trophoblast-uterine epithelial cell binding.
Collapse
Affiliation(s)
- L H Rohde
- Department of Biochemistry and Molecular Biology, University of Texas M.D. Anderson Cancer Center, Houston 77030
| | | |
Collapse
|
50
|
Robinovitch MR, Iversen JM, Izutsu KT. Inhibition of synthesis of rat parotid secretory proteoglycan in a gland slice system. Arch Oral Biol 1992; 37:209-14. [PMID: 1586282 DOI: 10.1016/0003-9969(92)90090-u] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The chondroitin sulphate contained within the secretory granules of the rat parotid gland and its saliva was shown to be in the form of a proteoglycan by using inhibitors of proteoglycan synthesis in a gland slice system. Gland slices were incubated in either p-nitrophenyl-beta-D-xyloside or chlorate in the presence of both [3H]-leucine and [35S]-sulphate. The slices were next homogenized and either the 250 g supernatant fraction (for initial experiments) or secretory granule-containing fractions were isolated. Protein and proteoglycans of these fractions were precipitated in 10% trichloracetic acid (TCA), and glycosaminoglycans in cetylpyridinium chloride. [3H]-leucine and [35S]-sulphate were quantitated in each type of precipitate by scintillation counting. The results showed that 1 mM xyloside had no effect on protein or glycosaminoglycan synthesis but blocked incorporation of radiosulphate into TCA-precipitable material. Sixteen mM chlorate almost totally inhibited incorporation of radiosulphate into glycosaminoglycan and TCA-precipitable material. These findings demonstrate that the rat parotid secretory chondroitin sulphate is indeed a proteoglycan because its synthesis is blocked by the protein-core analogue acceptor, p-nitrophenyl-beta-D-xyloside. This system offers opportunities for exploring the functional role of chondroitin sulphate proteoglycan in this salivary gland.
Collapse
Affiliation(s)
- M R Robinovitch
- Department of Oral Biology, School of Dentistry, University of Washington, Seattle 98195
| | | | | |
Collapse
|