151
|
Marie AL, Dominguez-Vega E, Saller F, Plantier JL, Urbain R, Borgel D, Tran NT, Somsen GW, Taverna M. Characterization of conformers and dimers of antithrombin by capillary electrophoresis-quadrupole-time-of-flight mass spectrometry. Anal Chim Acta 2016; 947:58-65. [PMID: 27846990 DOI: 10.1016/j.aca.2016.10.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 09/26/2016] [Accepted: 10/09/2016] [Indexed: 11/18/2022]
Abstract
Antithrombin (AT) is a plasma glycoprotein which possesses anticoagulant and anti-inflammatory properties. AT exhibits various forms, among which are native, latent and heterodimeric ones. We studied the potential of capillary electrophoresis-mass spectrometry (CE-MS) using a sheath liquid interface, electrospray ionization (ESI), and a quadrupole-time-of-flight (Q-TOF) mass spectrometer to separate and quantify the different AT forms. For CE separation, a neutral polyvinyl alcohol (PVA) coated capillary was employed. The protein conformation was preserved by using a background electrolyte (BGE) at physiological pH. A sheath liquid of isopropanol-water 50:50 (v/v) with 14 mM ammonium acetate delivered at a flow rate of 120 μL h-1 resulted in optimal signal intensities. Each AT form exhibited a specific mass spectrum, allowing unambiguous distinction. Several co-injection experiments proved that latent AT had a higher electrophoretic mobility (μep) than native AT, and that these conformers could associate to form a heterodimer during the CE analysis. The developed CE-MS method enabled the detection and quantitation of latent and heterodimeric forms in a commercial AT preparation stored at room temperature for three weeks.
Collapse
Affiliation(s)
- Anne-Lise Marie
- Institut Galien Paris Sud, UMR8612, Protein and Nanotechnology in Analytical Science (PNAS), CNRS, Univ. Paris-Sud, Université Paris-Saclay, 5 rue Jean-Baptiste Clément, 92290, Châtenay-Malabry, France
| | - Elena Dominguez-Vega
- Division of BioAnalytical Chemistry, AIMMS Research Group Biomolecular Analysis, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - François Saller
- Université Paris Sud, UMR-S1176, 94276, Le Kremlin-Bicêtre, France; INSERM, U1176, 94276, Le Kremlin-Bicêtre, France
| | | | | | - Delphine Borgel
- Université Paris Sud, UMR-S1176, 94276, Le Kremlin-Bicêtre, France; INSERM, U1176, 94276, Le Kremlin-Bicêtre, France; AP-HP, Hôpital Necker, Service d'Hématologie Biologique, 75015, Paris, France
| | - N Thuy Tran
- Institut Galien Paris Sud, UMR8612, Protein and Nanotechnology in Analytical Science (PNAS), CNRS, Univ. Paris-Sud, Université Paris-Saclay, 5 rue Jean-Baptiste Clément, 92290, Châtenay-Malabry, France
| | - Govert W Somsen
- Division of BioAnalytical Chemistry, AIMMS Research Group Biomolecular Analysis, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - Myriam Taverna
- Institut Galien Paris Sud, UMR8612, Protein and Nanotechnology in Analytical Science (PNAS), CNRS, Univ. Paris-Sud, Université Paris-Saclay, 5 rue Jean-Baptiste Clément, 92290, Châtenay-Malabry, France.
| |
Collapse
|
152
|
Li M, Yang S, Xu D. Heparan Sulfate Regulates the Structure and Function of Osteoprotegerin in Osteoclastogenesis. J Biol Chem 2016; 291:24160-24171. [PMID: 27697839 DOI: 10.1074/jbc.m116.751974] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/22/2016] [Indexed: 12/20/2022] Open
Abstract
Osteoprotegerin (OPG), a decoy receptor secreted by osteoblasts, is a major negative regulator of bone resorption. It functions by neutralizing the receptor activator of nuclear factor κB ligand (RANKL), which plays a central role in promoting osteoclastogenesis. OPG is known to be a high-affinity heparan sulfate (HS)-binding protein. Presumably, HS could regulate the function of OPG and affect how it inhibits RANKL. However, the molecular detail of HS-OPG interaction remains poorly understood, which hinders our understanding of how HS functions in osteoclastogenesis. Here we report mapping of the HS-binding site of OPG. The HS-binding site, identified by mutagenesis study, consists of eight basic residues that are located mostly at the junction of the second death domain and the C-terminal domain. We further show that heparin-derived dodecasaccharide is able to induce dimerization of OPG monomers with a stoichiometry of 1:1. Small-angle X-ray scattering analysis revealed that upon binding of HS, OPG undergoes a dramatic conformational change, resulting in a more compact and less flexible structure. Importantly, we present here three lines of evidence that HS, OPG, and RANKL form a stable ternary complex. Using a HS binding-deficient OPG mutant, we further show that in an osteoblast/bone marrow macrophage co-culture system, immobilization of OPG by HS at the osteoblast cell surface substantially lowers the inhibitory threshold of OPG toward RANKL. These discoveries strongly suggest that HS plays an active role in regulating OPG-RANKL interaction and osteoclastogenesis.
Collapse
Affiliation(s)
- Miaomiao Li
- From the Department of Oral Biology, School of Dental Medicine, University at Buffalo, the State University of New York, Buffalo, New York 14214
| | - Shuying Yang
- From the Department of Oral Biology, School of Dental Medicine, University at Buffalo, the State University of New York, Buffalo, New York 14214
| | - Ding Xu
- From the Department of Oral Biology, School of Dental Medicine, University at Buffalo, the State University of New York, Buffalo, New York 14214
| |
Collapse
|
153
|
Motamedi-Shad N, Jagger AM, Liedtke M, Faull SV, Nanda AS, Salvadori E, Wort JL, Kay CW, Heyer-Chauhan N, Miranda E, Perez J, Ordóñez A, Haq I, Irving JA, Lomas DA. An antibody that prevents serpin polymerisation acts by inducing a novel allosteric behaviour. Biochem J 2016; 473:3269-90. [PMID: 27407165 PMCID: PMC5264506 DOI: 10.1042/bcj20160159] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 07/08/2016] [Accepted: 07/12/2016] [Indexed: 11/30/2022]
Abstract
Serpins are important regulators of proteolytic pathways with an antiprotease activity that involves a conformational transition from a metastable to a hyperstable state. Certain mutations permit the transition to occur in the absence of a protease; when associated with an intermolecular interaction, this yields linear polymers of hyperstable serpin molecules, which accumulate at the site of synthesis. This is the basis of many pathologies termed the serpinopathies. We have previously identified a monoclonal antibody (mAb4B12) that, in single-chain form, blocks α1-antitrypsin (α1-AT) polymerisation in cells. Here, we describe the structural basis for this activity. The mAb4B12 epitope was found to encompass residues Glu32, Glu39 and His43 on helix A and Leu306 on helix I. This is not a region typically associated with the serpin mechanism of conformational change, and correspondingly the epitope was present in all tested structural forms of the protein. Antibody binding rendered β-sheet A - on the opposite face of the molecule - more liable to adopt an 'open' state, mediated by changes distal to the breach region and proximal to helix F. The allosteric propagation of induced changes through the molecule was evidenced by an increased rate of peptide incorporation and destabilisation of a preformed serpin-enzyme complex following mAb4B12 binding. These data suggest that prematurely shifting the β-sheet A equilibrium towards the 'open' state out of sequence with other changes suppresses polymer formation. This work identifies a region potentially exploitable for a rational design of ligands that is able to dynamically influence α1-AT polymerisation.
Collapse
Affiliation(s)
- Neda Motamedi-Shad
- Centre for Respiratory Biology, UCL Respiratory, University College London, London WC1E 6JF, U.K
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
| | - Alistair M. Jagger
- Centre for Respiratory Biology, UCL Respiratory, University College London, London WC1E 6JF, U.K
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
| | - Maximilian Liedtke
- Centre for Respiratory Biology, UCL Respiratory, University College London, London WC1E 6JF, U.K
| | - Sarah V. Faull
- Centre for Respiratory Biology, UCL Respiratory, University College London, London WC1E 6JF, U.K
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council Building, Hills Road, Cambridge CB2 0XY, U.K
| | - Arjun Scott Nanda
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
| | - Enrico Salvadori
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
- London Centre for Nanotechnology, 17-19 Gordon Street, London WC1H 0AH, U.K
- School of Biological and Chemical Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, U.K
| | - Joshua L. Wort
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
| | - Christopher W.M. Kay
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
- London Centre for Nanotechnology, 17-19 Gordon Street, London WC1H 0AH, U.K
| | - Narinder Heyer-Chauhan
- Centre for Respiratory Biology, UCL Respiratory, University College London, London WC1E 6JF, U.K
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
| | - Elena Miranda
- Department of Biology and Biotechnologies ‘Charles Darwin’, Sapienza University of Rome, Rome 00185, Italy
| | - Juan Perez
- Departamento de Biologia Celular, Genetica y Fisiologia, Facultad de Ciencias, Campus Teatinos, Universidad de Malaga, Malaga 29071, Spain
| | - Adriana Ordóñez
- Department of Medicine, University of Cambridge, Cambridge Institute for Medical Research, Wellcome Trust/Medical Research Council Building, Hills Road, Cambridge CB2 0XY, U.K
| | - Imran Haq
- Centre for Respiratory Biology, UCL Respiratory, University College London, London WC1E 6JF, U.K
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
| | - James A. Irving
- Centre for Respiratory Biology, UCL Respiratory, University College London, London WC1E 6JF, U.K
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
| | - David A. Lomas
- Centre for Respiratory Biology, UCL Respiratory, University College London, London WC1E 6JF, U.K
- Institute of Structural and Molecular Biology/Birkbeck, University of London, London WC1E 7HX, U.K
| |
Collapse
|
154
|
Wei H, Cai H, Wu J, Wei Z, Zhang F, Huang X, Ma L, Feng L, Zhang R, Wang Y, Ragg H, Zheng Y, Zhou A. Heparin Binds Lamprey Angiotensinogen and Promotes Thrombin Inhibition through a Template Mechanism. J Biol Chem 2016; 291:24900-24911. [PMID: 27681598 DOI: 10.1074/jbc.m116.725895] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 08/20/2016] [Indexed: 01/01/2023] Open
Abstract
Lamprey angiotensinogen (l-ANT) is a hormone carrier in the regulation of blood pressure, but it is also a heparin-dependent thrombin inhibitor in lamprey blood coagulation system. The detailed mechanisms on how angiotensin is carried by l-ANT and how heparin binds l-ANT and mediates thrombin inhibition are unclear. Here we have solved the crystal structure of cleaved l-ANT at 2.7 Å resolution and characterized its properties in heparin binding and protease inhibition. The structure reveals that l-ANT has a conserved serpin fold with a labile N-terminal angiotensin peptide and undergoes a typical stressed-to-relaxed conformational change when the reactive center loop is cleaved. Heparin binds l-ANT tightly with a dissociation constant of ∼10 nm involving ∼8 monosaccharides and ∼6 ionic interactions. The heparin binding site is located in an extensive positively charged surface area around helix D involving residues Lys-148, Lys-151, Arg-155, and Arg-380. Although l-ANT by itself is a poor thrombin inhibitor with a second order rate constant of 500 m-1 s-1, its interaction with thrombin is accelerated 90-fold by high molecular weight heparin following a bell-shaped dose-dependent curve. Short heparin chains of 6-20 monosaccharide units are insufficient to promote thrombin inhibition. Furthermore, an l-ANT mutant with the P1 Ile mutated to Arg inhibits thrombin nearly 1500-fold faster than the wild type, which is further accelerated by high molecular weight heparin. Taken together, these results suggest that heparin binds l-ANT at a conserved heparin binding site around helix D and promotes the interaction between l-ANT and thrombin through a template mechanism conserved in vertebrates.
Collapse
Affiliation(s)
- Hudie Wei
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| | - Haiyan Cai
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| | - Jiawei Wu
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| | - Zhenquan Wei
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| | - Fei Zhang
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| | - Xin Huang
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| | - Lina Ma
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| | - Lingling Feng
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| | - Ruoxi Zhang
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| | - Yunjie Wang
- the Faculty of Technology, Bielefeld University, 33613 Bielefeld, Germany
| | - Hermann Ragg
- the Faculty of Technology, Bielefeld University, 33613 Bielefeld, Germany
| | - Ying Zheng
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| | - Aiwu Zhou
- From the Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China and
| |
Collapse
|
155
|
Kim JY, Kim SY, Cheon MH, Kim ES, Song IS, Kim MJ, Tchah H. Attenuation of corneal neovascularization by topical low-molecular-weight heparin-taurocholate 7 without bleeding complication. Int J Ophthalmol 2016; 9:1255-9. [PMID: 27672587 DOI: 10.18240/ijo.2016.09.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 09/08/2015] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate the antiangiogenic effects and safety of topically administered low-molecular-weight heparin-taurocholate 7 (LHT7) on corneal neovascularization (CoNV). METHODS Twenty-four Sprague-Dawley rats were randomly distributed into four groups of six rats each. The central corneas were cauterized using a silver/potassium nitrate solution. From 2d after cauterization, 12.5 mg/mL (low LHT7 group) or 25 mg/mL (high LHT7 group) LHT7 was topically administered three times daily; 12.5 mg/mL bevacizumab was topically administered as positive control (bevacizumab) group, with normal saline (NS) administered as negative control (NS group). The corneas were digitally photographed to calculate the CoNV percentage from the neovascularized corneal area at 1 and 2wk. RESULTS The 4 study groups did not have different CoNV percentages at 1wk after injury (P>0.05). However, the low LHT, high LHT, and bevacizumab groups had significantly lower CoNV percentages than the NS group at 2wk (all P<0.05). No significant differences in CoNV percentage were found among the low LHT, high LHT, and bevacizumab groups (all P>0.05). All groups except the NS group had lower CoNV percentages at 2wk post-injury than the levels observed at 1wk (all P<0.05). CONCLUSION Topically-administered LHT7 inhibited CoNV without complication after chemical cauterization in the rat.
Collapse
Affiliation(s)
- Jae Yong Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Soo Yeon Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Mi Hyun Cheon
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Eun-Soon Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - In Seok Song
- Department of Ophthalmology, Hanyang University College of Medicine, Seoul 04763, Korea
| | - Myoung Joon Kim
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| | - Hungwon Tchah
- Department of Ophthalmology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea
| |
Collapse
|
156
|
Serpins in arthropod biology. Semin Cell Dev Biol 2016; 62:105-119. [PMID: 27603121 DOI: 10.1016/j.semcdb.2016.09.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 08/31/2016] [Accepted: 09/02/2016] [Indexed: 12/21/2022]
Abstract
Serpins are the largest known family of serine proteinase inhibitors and perform a variety of physiological functions in arthropods. Herein, we review the field of serpins in arthropod biology, providing an overview of current knowledge and topics of interest. Serpins regulate insect innate immunity via inhibition of serine proteinase cascades that initiate immune responses such as melanization and antimicrobial peptide production. In addition, several serpins with anti-pathogen activity are expressed as acute-phase serpins in insects upon infection. Parasitoid wasps can downregulate host serpin expression to modulate the host immune system. In addition, examples of serpin activity in development and reproduction in Drosophila have also been discovered. Serpins also function in host-pathogen interactions beyond immunity as constituents of venom in parasitoid wasps and saliva of blood-feeding ticks and mosquitoes. These serpins have distinct effects on immunosuppression and anticoagulation and are of interest for vaccine development. Lastly, the known structures of arthropod serpins are discussed, which represent the serpin inhibitory mechanism and provide a detailed overview of the process.
Collapse
|
157
|
Mulloy B, Hogwood J, Gray E, Lever R, Page CP. Pharmacology of Heparin and Related Drugs. Pharmacol Rev 2016; 68:76-141. [PMID: 26672027 DOI: 10.1124/pr.115.011247] [Citation(s) in RCA: 227] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Heparin has been recognized as a valuable anticoagulant and antithrombotic for several decades and is still widely used in clinical practice for a variety of indications. The anticoagulant activity of heparin is mainly attributable to the action of a specific pentasaccharide sequence that acts in concert with antithrombin, a plasma coagulation factor inhibitor. This observation has led to the development of synthetic heparin mimetics for clinical use. However, it is increasingly recognized that heparin has many other pharmacological properties, including but not limited to antiviral, anti-inflammatory, and antimetastatic actions. Many of these activities are independent of its anticoagulant activity, although the mechanisms of these other activities are currently less well defined. Nonetheless, heparin is being exploited for clinical uses beyond anticoagulation and developed for a wide range of clinical disorders. This article provides a "state of the art" review of our current understanding of the pharmacology of heparin and related drugs and an overview of the status of development of such drugs.
Collapse
Affiliation(s)
- Barbara Mulloy
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., C.P.P.); National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, United Kingdom (J.H., E.G.); and University College London School of Pharmacy, London, United Kingdom (R.L.)
| | - John Hogwood
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., C.P.P.); National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, United Kingdom (J.H., E.G.); and University College London School of Pharmacy, London, United Kingdom (R.L.)
| | - Elaine Gray
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., C.P.P.); National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, United Kingdom (J.H., E.G.); and University College London School of Pharmacy, London, United Kingdom (R.L.)
| | - Rebecca Lever
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., C.P.P.); National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, United Kingdom (J.H., E.G.); and University College London School of Pharmacy, London, United Kingdom (R.L.)
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, United Kingdom (B.M., C.P.P.); National Institute for Biological Standards and Control, Potters Bar, Hertfordshire, United Kingdom (J.H., E.G.); and University College London School of Pharmacy, London, United Kingdom (R.L.)
| |
Collapse
|
158
|
Gray C, Thomas B, Upton R, Migas L, Eyers C, Barran P, Flitsch S. Applications of ion mobility mass spectrometry for high throughput, high resolution glycan analysis. Biochim Biophys Acta Gen Subj 2016; 1860:1688-709. [DOI: 10.1016/j.bbagen.2016.02.003] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/01/2016] [Accepted: 02/02/2016] [Indexed: 12/21/2022]
|
159
|
Baumann H. Biological Effects of Heparan Sulfates and Regioselectively Modified Heparin-Heparan Mimetics. J BIOACT COMPAT POL 2016. [DOI: 10.1177/0883911503018001006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The heparan sulfates (HS) are structurally the most complex but information rich biopolymers known. They are composed of polysaccharides containing regioselectively distributed carboxyl, sulfate ester, acetyl, amino, and N-sulfonyl groups with sequence- and domain-like arrangements. HS are found ubiquitously on cell surfaces and in extracellular matrices where they are covalently anchored via restricted protein cores. They modulate numerous development cell processes and the pathology of living organisms. HS concentration is extremely low on endothelial cell surfaces (1 pmol/cm2), therefore, they are difficult to isolate and evaluate. Furthermore, their sequence variability is extremely high and the sequence analysis is in its infancy. HS acts as a low affinity receptor which plays a central role in the reception and modulation of a wide range of effector proteins such as growth factors, morphogens, chemokines, enzymes, protease inhibitors. Water soluble fragments of HS and heparin (HE) enzymatically released or synthetic sequences, analogs of heparinoids and heparanoids (HH) mimetics regioselectively modified oligo- and polysaccharides with HE/HS like functional groups, and nonsaccharide containing structures can modulate effector proteins and influence some of the development and pathological processes. Modulation effects are described for anticoagulant antiproliferative properties, for reducing platelet and plasma protein adhesion as well as inhibition or activating growth factors by the influence of HH mimetics. The advantage of defined high molecular weight substrates are discussed and compared to the low molecular weight mimetics. The potential of HH mimetics opens new approaches and strategies for therapeutic treatment.
Collapse
Affiliation(s)
- H. Baumann
- Institute for Technical and Macromolecular Chemistry Hemocompatible and Biocompatible Biomaterials RWTH Aachen, Worringer Weg 1, D-52074 Aachen, Germany
| |
Collapse
|
160
|
Sarkar A, Yu W, Desai UR, MacKerell AD, Mosier PD. Estimating glycosaminoglycan-protein interaction affinity: water dominates the specific antithrombin-heparin interaction. Glycobiology 2016; 26:1041-1047. [PMID: 27496757 DOI: 10.1093/glycob/cww073] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/01/2016] [Accepted: 07/12/2016] [Indexed: 11/13/2022] Open
Abstract
Glycosaminoglycan (GAG)-protein interactions modulate many important biological processes. Structure-function studies on GAGs may reveal probes and drugs, but their structural complexity and highly acidic nature confound such work. Productivity will increase if we are able to identify tight-binding oligosaccharides in silico. An extension of the CHARMM force field is presented to enable modeling of polysaccharides containing sulfamate functionality, and is used to develop a reliable alchemical free-energy perturbation protocol that estimates changes in affinity for the prototypical heparin-antithrombin system to within 2.3 kcal/mol using modest simulation times. Inclusion of water is crucial during simulation as solvation energy was equal in magnitude to the sum of all other thermodynamic factors. In summary, we have identified and optimized a reliable method for estimation of GAG-protein binding affinity, and shown that solvation is a crucial component in GAG-protein interactions.
Collapse
Affiliation(s)
- Aurijit Sarkar
- Department of Basic Pharmaceutical Sciences, Fred Wilson School of Pharmacy, High Point University, One University Parkway, High Point, NC 27268, USA .,Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, PO Box 980133, Richmond, VA 23298, USA
| | - Wenbo Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA
| | - Umesh R Desai
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, PO Box 980133, Richmond, VA 23298, USA
| | - Alexander D MacKerell
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD 21201, USA
| | - Philip D Mosier
- Department of Medicinal Chemistry and Institute for Structural Biology, Drug Discovery and Development, School of Pharmacy, Virginia Commonwealth University, PO Box 980133, Richmond, VA 23298, USA
| |
Collapse
|
161
|
Hsieh PH, Thieker DF, Guerrini M, Woods RJ, Liu J. Uncovering the Relationship between Sulphation Patterns and Conformation of Iduronic Acid in Heparan Sulphate. Sci Rep 2016; 6:29602. [PMID: 27412370 PMCID: PMC4944151 DOI: 10.1038/srep29602] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/21/2016] [Indexed: 12/19/2022] Open
Abstract
The L-iduronic acid (IdoA) residue is a critically important structural component in heparan sulphate polysaccharide for the biological functions. The pyranose ring of IdoA is present in (1)C4-chair, (2)SO-skew boat, and less frequently, in (4)C1-chair conformations. Here, we analyzed the conformation of IdoA residue in eight hexasaccharides by NMR. The data demonstrate a correlation between the conformation of IdoA and sulphations in the surrounding saccharide residues. For the 2-O-sulpho IdoA residue, a high degree of sulphation on neighboring residues drives ring dynamics towards the (2)SO-skew boat conformer. In contrast, the nonsulphated IdoA residue is pushed towards the (1)C4-chair conformer when the neighboring residues are highly sulphated. Our data suggest that the conformation of IdoA is regulated by the sulphation pattern of nearby saccharides that is genetically controlled by the heparan sulphate biosynthetic pathway.
Collapse
Affiliation(s)
- Po-Hung Hsieh
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - David F. Thieker
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602, USA
| | - Marco Guerrini
- Istituto di Ricerche Chimiche e Biochimiche ‘G. Ronzoni’, via G. Colombo 81, 20133 Milan, Italy
| | - Robert J. Woods
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia 30602, USA
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| |
Collapse
|
162
|
Gschwandtner M, Piccinini AM, Gerlza T, Adage T, Kungl AJ. Interfering with the CCL2–glycosaminoglycan axis as a potential approach to modulate neuroinflammation. Neurosci Lett 2016; 626:164-73. [DOI: 10.1016/j.neulet.2016.05.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/03/2016] [Accepted: 05/18/2016] [Indexed: 01/16/2023]
|
163
|
Manissorn J, Thongboonkerd V. Characterizations of heparin-binding proteins in human urine by affinity purification-mass spectrometry and defining “L-x(2,3)-A-x(0,1)-L” as a novel heparin-binding motif. J Proteomics 2016; 142:53-61. [DOI: 10.1016/j.jprot.2016.04.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 04/16/2016] [Accepted: 04/28/2016] [Indexed: 01/10/2023]
|
164
|
Marie AL, Tran NT, Saller F, Abdou YM, Zeau P, Plantier JL, Urbain R, Borgel D, Taverna M. A capillary zone electrophoresis method to detect conformers and dimers of antithrombin in therapeutic preparations. Electrophoresis 2016; 37:1696-703. [DOI: 10.1002/elps.201500456] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 03/06/2016] [Accepted: 03/07/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Anne-Lise Marie
- Institut Galien Paris Sud, UMR8612, Protein and Nanotechnology in Analytical Science (PNAS), CNRS, Université Paris-Sud; Université Paris-Saclay; Châtenay-Malabry France
| | - Nguyet Thuy Tran
- Institut Galien Paris Sud, UMR8612, Protein and Nanotechnology in Analytical Science (PNAS), CNRS, Université Paris-Sud; Université Paris-Saclay; Châtenay-Malabry France
| | - François Saller
- Université Paris Sud; UMR-S1176; Le Kremlin-Bicêtre France
- INSERM; U1176 Le Kremlin-Bicêtre France
| | - Youmna Mohamed Abdou
- Institut Galien Paris Sud, UMR8612, Protein and Nanotechnology in Analytical Science (PNAS), CNRS, Université Paris-Sud; Université Paris-Saclay; Châtenay-Malabry France
| | | | | | | | - Delphine Borgel
- Université Paris Sud; UMR-S1176; Le Kremlin-Bicêtre France
- INSERM; U1176 Le Kremlin-Bicêtre France
- AP-HP, Hôpital Necker; Service d'Hématologie Biologique; Paris France
| | - Myriam Taverna
- Institut Galien Paris Sud, UMR8612, Protein and Nanotechnology in Analytical Science (PNAS), CNRS, Université Paris-Sud; Université Paris-Saclay; Châtenay-Malabry France
| |
Collapse
|
165
|
Azhar A, Khan MS, Swaminathan A, Naseem A, Chatterjee S, Jairajpuri MA. Oxidized antithrombin is a dual inhibitor of coagulation and angiogenesis: Importance of low heparin affinity. Int J Biol Macromol 2016; 82:541-50. [DOI: 10.1016/j.ijbiomac.2015.10.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/01/2015] [Accepted: 10/05/2015] [Indexed: 01/09/2023]
|
166
|
|
167
|
Takaoka T, Mori K, Okimoto N, Neya S, Hoshino T. Prediction of the Structure of Complexes Comprised of Proteins and Glycosaminoglycans Using Docking Simulation and Cluster Analysis. J Chem Theory Comput 2015; 3:2347-56. [PMID: 26636224 DOI: 10.1021/ct700029q] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A typical docking simulation provides information on the structure of ligand-receptor complexes and their binding affinity in terms of a docking energy. We have developed a potent method combining a docking simulation with cluster analysis to extract adequate docking structures from the many possible output structures of the simulation. First, we tried to predict the structure of basic fibroblast growth factor (bFGF) bound to heparin, using the docking simulation program AutoDock 3.0. Two X-ray crystal structures had already been obtained for bFGF. One was a complex of the protein and heparin, a kind of glycosaminoglycan, and the other, only the protein itself, hereafter called a simplex. We docked a heparin molecule onto the protein simplex and generated many trial structures for the bFGF-heparin complex. The structures of those docked complexes were optimized through energy minimization by AMBER8. Although neither the docking energy calculated by AMBER8 nor that calculated by AutoDock 3.0 could be used satisfactorily by themselves to select a proper heparin-binding complex from the output structures, the majority of the structures generated by AutoDock 3.0 were fairly close to each other in atom geometry, and the averaged geometry over these structures was also close to that of the crystal. Hence, we utilized only the atom geometry for evaluation and carried out cluster analysis with the collection of geometries. This procedure enabled selection of a structure considerably close to the crystal's. We applied this approach to two other heparin-binding proteins: antithrombin and annexin V. Two crystal structures, a complex and a simplex, had been elucidated for these proteins as well as for bFGF. Our trials gave an exact prediction of the heparin-binding structures of these proteins, showing the approach in this study is effective in studying the docking of ligands that have a variety of docking conformations due to the presence of multiple rotatable bonds and charged chemical groups.
Collapse
Affiliation(s)
- Tsubasa Takaoka
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan, Bioinformatics Group, GSC, RIKEN, Yokohama, Kanagawa 230-0046, Japan, and PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Kenichi Mori
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan, Bioinformatics Group, GSC, RIKEN, Yokohama, Kanagawa 230-0046, Japan, and PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Noriaki Okimoto
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan, Bioinformatics Group, GSC, RIKEN, Yokohama, Kanagawa 230-0046, Japan, and PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Saburo Neya
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan, Bioinformatics Group, GSC, RIKEN, Yokohama, Kanagawa 230-0046, Japan, and PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| | - Tyuji Hoshino
- Graduate School of Pharmaceutical Sciences, Chiba University, Chiba 263-8522, Japan, Bioinformatics Group, GSC, RIKEN, Yokohama, Kanagawa 230-0046, Japan, and PRESTO, Japan Science and Technology Agency, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
168
|
Mechanistic and therapeutic overview of glycosaminoglycans: the unsung heroes of biomolecular signaling. Glycoconj J 2015; 33:1-17. [DOI: 10.1007/s10719-015-9642-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 11/19/2015] [Accepted: 11/20/2015] [Indexed: 12/28/2022]
|
169
|
Sarkar A, Desai UR. A Simple Method for Discovering Druggable, Specific Glycosaminoglycan-Protein Systems. Elucidation of Key Principles from Heparin/Heparan Sulfate-Binding Proteins. PLoS One 2015; 10:e0141127. [PMID: 26488293 PMCID: PMC4619353 DOI: 10.1371/journal.pone.0141127] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 10/05/2015] [Indexed: 01/25/2023] Open
Abstract
Glycosaminoglycans (GAGs) affect human physiology and pathology by modulating more than 500 proteins. GAG-protein interactions are generally assumed to be ionic and nonspecific, but specific interactions do exist. Here, we present a simple method to identify the GAG-binding site (GBS) on proteins that in turn helps predict high specific GAG-protein systems. Contrary to contemporary thinking, we found that the electrostatic potential at basic arginine and lysine residues neither identifies the GBS consistently, nor its specificity. GBSs are better identified by considering the potential at neutral hydrogen bond donors such as asparagine or glutamine sidechains. Our studies also reveal that an unusual constellation of ionic and non-ionic residues in the binding site leads to specificity. Nature engineers the local environment of Asn45 of antithrombin, Gln255 of 3-O-sulfotransferase 3, Gln163 and Asn167 of 3-O-sulfotransferase 1 and Asn27 of basic fibroblast growth factor in the respective GBSs to induce specificity. Such residues are distinct from other uncharged residues on the same protein structure in possessing a significantly higher electrostatic potential, resultant from the local topology. In contrast, uncharged residues on nonspecific GBSs such as thrombin and serum albumin possess a diffuse spread of electrostatic potential. Our findings also contradict the paradigm that GAG-binding sites are simply a collection of contiguous Arg/Lys residues. Our work demonstrates the basis for discovering specifically interacting and druggable GAG-protein systems based on the structure of protein alone, without requiring access to any structure-function relationship data.
Collapse
Affiliation(s)
- Aurijit Sarkar
- Institute for Structural Biology, Drug Discovery & Development and Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Umesh R. Desai
- Institute for Structural Biology, Drug Discovery & Development and Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, Virginia, United States of America
| |
Collapse
|
170
|
Zhao Y, Singh A, Li L, Linhardt RJ, Xu Y, Liu J, Woods RJ, Amster IJ. Investigating changes in the gas-phase conformation of Antithrombin III upon binding of Arixtra using traveling wave ion mobility spectrometry (TWIMS). Analyst 2015; 140:6980-9. [PMID: 26115461 PMCID: PMC4586392 DOI: 10.1039/c5an00908a] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
We validate the utility of ion mobility to measure protein conformational changes induced by the binding of glycosaminoglycan ligands, using the well characterized system of Antithrombin III (ATIII) and Arixtra, a pharmaceutical agent with heparin (Hp) activity. Heparin has been used as a therapeutic anticoagulant drug for several decades through its interaction with ATIII, a serine protease inhibitor that plays a central role in the blood coagulation cascade. This interaction induces conformational changes within ATIII that dramatically enhance the ATIII-mediated inhibition rate. Arixtra is the smallest synthetic Hp containing the specific pentasaccharide sequence required to bind with ATIII. Here we report the first travelling wave ion mobility mass spectrometry (TWIMS) investigation of the conformational changes in ATIII induced by its interaction with Arixtra. Native electrospray ionization mass spectrometry allowed the gentle transfer of the native topology of ATIII and ATIII-Arixtra complex. IM measurements of ATIII and ATIII-Arixtra complex showed a single structure, with well-defined collisional cross section (CCS) values. An average 3.6% increase in CCS of ATIII occurred as a result of its interaction with Arixtra, which agrees closely with the theoretical estimation of the change in CCS based on protein crystal structures. A comparison of the binding behavior of ATIII under both denaturing and non-denaturing conditions confirmed the significance of a folded tertiary structure of ATIII for its biological activity. A Hp oligosaccharide whose structure is similar to Arixtra but missing the 3-O sulfo group on the central glucosamine residue showed a dramatic decrease in binding affinity towards ATIII, but no change in the mobility behavior of the complex, consistent with prior studies that suggested that 3-O sulfation affects the equilibrium constant for binding to ATIII, but not the mode of interaction. In contrast, nonspecific binding by a Hp tetrasaccharide showed more complex mobility behavior, suggesting more promiscuous interactions with ATIII. The effect of collisional activation of ATIII and ATIII-Arixtra complex were also assessed, revealing that the binding of Arixtra provided ATIII with additional stability against unfolding. Overall, our results validate the capability of TWIMS to retain the significant features of the solution structure of a protein-carbohydrate complex so that it can be used to study protein conformational changes induced by the binding of glycosaminoglycan ligands.
Collapse
Affiliation(s)
- Yuejie Zhao
- University of Georgia, Department of Chemistry, 140 Cedar Street, Athens, GA 30602-2556, USA.
| | | | | | | | | | | | | | | |
Collapse
|
171
|
Weiss RJ, Gordts PLSM, Le D, Xu D, Esko JD, Tor Y. Small molecule antagonists of cell-surface heparan sulfate and heparin-protein interactions. Chem Sci 2015; 6:5984-5993. [PMID: 28133533 PMCID: PMC5267326 DOI: 10.1039/c5sc01208b] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 07/21/2015] [Indexed: 01/13/2023] Open
Abstract
Surfen, bis-2-methyl-4-amino-quinolyl-6-carbamide, was previously reported as a small molecule antagonist of heparan sulfate (HS), a key cell-surface glycosaminoglycan found on all mammalian cells. To generate structure-activity relationships, a series of rationally designed surfen analogs was synthesized, where its dimeric structure, exocyclic amines, and urea linker region were modified to probe the role of each moiety in recognizing HS. An in vitro assay monitoring inhibition of fibroblast growth factor 2 binding to wild-type CHO cells was utilized to quantify interactions with cell surface HS. The dimeric molecular structure of surfen and its aminoquinoline ring systems was essential for its interaction with HS, and certain dimeric analogs displayed higher inhibitory potency than surfen and were also shown to block downstream FGF signaling in mouse embryonic fibroblast cells. These molecules were also able to antagonize other HS-protein interactions including the binding of soluble RAGE to HS. Importantly, selected molecules were shown to neutralize heparin and other heparinoids, including the synthetic pentasaccharide fondaparinux, in a factor Xa chromogenic assay and in vivo in mice. These results suggest that small molecule antagonists of heparan sulfate and heparin can be of therapeutic potential for the treatment of disorders involving glycosaminoglycan-protein interactions.
Collapse
Affiliation(s)
- Ryan J. Weiss
- Department of Chemistry and Biochemistry , University of California , San Diego , La Jolla , CA 92093-0358 , USA .
| | - Philip L. S. M. Gordts
- Cellular and Molecular Medicine , University of California , San Diego , La Jolla , CA 92093-0687 , USA
| | - Dzung Le
- Department of Medicine , University of California , San Diego , La Jolla , CA 92093-0612 , USA
- Glycobiology Research and Training Center , University of California , San Diego , La Jolla , CA 92093-0687 , USA
| | - Ding Xu
- Department of Oral Biology , University at Buffalo , Buffalo , NY 14260-1660 , USA
| | - Jeffrey D. Esko
- Cellular and Molecular Medicine , University of California , San Diego , La Jolla , CA 92093-0687 , USA
- Glycobiology Research and Training Center , University of California , San Diego , La Jolla , CA 92093-0687 , USA
| | - Yitzhak Tor
- Department of Chemistry and Biochemistry , University of California , San Diego , La Jolla , CA 92093-0358 , USA .
- Glycobiology Research and Training Center , University of California , San Diego , La Jolla , CA 92093-0687 , USA
| |
Collapse
|
172
|
Abstract
High-resolution NMR and density functional theory (DFT) calculations have been applied to analysis of heparin pentasaccharide 3D structure in aqueous solution. The fully optimized molecular geometry of two pentasaccharide conformations (differing from each other in the form, one (1)C4 and the other (2)S0, of the sulfated iduronic acid residue) were obtained using the B3LYP/6-311+G(d,p) level of theory in the presence of solvent, the latter included as explicit water molecules. The presented approach enabled insight into variations of the bond lengths, bond angles, and torsion angles, formations of intra- and intermolecular hydrogen bonds, and ionic interactions in the two pentasaccharide conformations. A rather complex hydrogen bond network is formed, including inter-residue and intraresidue bonds between the NH group in the GlcN,3,6S with oxygens linked to C-2 at the IdoA2S residue and the glycosidic O-1 and the neighboring OSO3(-) group linked to C-3 in the same residue. On the other hand, because the first hydration shell is strongly influenced by strong ion-ion and ion-dipole interactions between sodium ions, sulfates, carboxylates, and -OH groups, ionic interactions play an important role in the stabilization of the 3D structure. The DFT-computed three-bond proton-proton coupling constants also showed that best agreement with experiment was obtained with a weighted average of 15:85 ((1)C4/(2)S0) of the sulfated iduronic acid forms indicating that the ratio is even more shifted toward the (2)S0 form than previously supposed. The DFT-computed pentasaccharide conformation differs from the previously published data, with the main changes at the glycosidic linkages, namely, the ψ1 torsion angles and the ϕ3 angle. The comparison of the glycosidic linkage torsion angle values in solution with the antithrombin-pentasaccharide complex also indicates that the pentasaccharide conformation changes upon binding to antithrombin III. The data supports the assumption that the protein selects the more populated (2)S0 conformer of heparin pentasaccharide and, consequently, the binding process of heparin pentasaccharide with antithrombin III is energetically more favorable than formerly expected.
Collapse
Affiliation(s)
- Miloš Hricovíni
- Institute of Chemistry, Slovak Academy of Sciences , 845 38 Bratislava, Slovakia
| |
Collapse
|
173
|
Lin F, Zhou A, Wei Z. Crystallization and crystallographic studies of kallistatin. Acta Crystallogr F Struct Biol Commun 2015; 71:1135-8. [PMID: 26323298 PMCID: PMC4555919 DOI: 10.1107/s2053230x15012893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 07/03/2015] [Indexed: 01/27/2023] Open
Abstract
Kallistatin is a serine protease inhibitor (serpin) which specifically inhibits human tissue kallikrein; however, its inhibitory activity is inhibited by heparin. In order to elucidate the underlying mechanism, recombinant human kallistatin was prepared in Escherichia coli and the protein was crystallized by the sitting-drop vapour-diffusion method. X-ray diffraction data were collected to 1.9 Å resolution. The crystals were found to belong to space group P61, with unit-cell parameters a = 113.51, b = 113.51, c = 76.17 Å. Initial analysis indicated that the crystallized kallistatin was in a relaxed conformation, with its reactive-centre loop inserted in the central β-sheet.
Collapse
Affiliation(s)
- Fang Lin
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Faculty of Basic Medicine, and Department of Pathophysiology, Shanghai Jiaotong University School of Medicine, (Room 1006, Building 2, No 280, South Chongqing Road), Shanghai 200025, People’s Republic of China
| | - Aiwu Zhou
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Faculty of Basic Medicine, and Department of Pathophysiology, Shanghai Jiaotong University School of Medicine, (Room 1006, Building 2, No 280, South Chongqing Road), Shanghai 200025, People’s Republic of China
| | - Zhenquan Wei
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital and Faculty of Basic Medicine, and Department of Pathophysiology, Shanghai Jiaotong University School of Medicine, (Room 1006, Building 2, No 280, South Chongqing Road), Shanghai 200025, People’s Republic of China
| |
Collapse
|
174
|
Abstract
The article reviews the significant contributions to, and the present status of, applications of computational methods for the characterization and prediction of protein-carbohydrate interactions. After a presentation of the specific features of carbohydrate modeling, along with a brief description of the experimental data and general features of carbohydrate-protein interactions, the survey provides a thorough coverage of the available computational methods and tools. At the quantum-mechanical level, the use of both molecular orbitals and density-functional theory is critically assessed. These are followed by a presentation and critical evaluation of the applications of semiempirical and empirical methods: QM/MM, molecular dynamics, free-energy calculations, metadynamics, molecular robotics, and others. The usefulness of molecular docking in structural glycobiology is evaluated by considering recent docking- validation studies on a range of protein targets. The range of applications of these theoretical methods provides insights into the structural, energetic, and mechanistic facets that occur in the course of the recognition processes. Selected examples are provided to exemplify the usefulness and the present limitations of these computational methods in their ability to assist in elucidation of the structural basis underlying the diverse function and biological roles of carbohydrates in their dialogue with proteins. These test cases cover the field of both carbohydrate biosynthesis and glycosyltransferases, as well as glycoside hydrolases. The phenomenon of (macro)molecular recognition is illustrated for the interactions of carbohydrates with such proteins as lectins, monoclonal antibodies, GAG-binding proteins, porins, and viruses.
Collapse
Affiliation(s)
- Serge Pérez
- Department of Molecular Pharmacochemistry, CNRS, University Grenoble-Alpes, Grenoble, France.
| | - Igor Tvaroška
- Department of Chemistry, Slovak Academy of Sciences, Bratislava, Slovak Republic; Department of Chemistry, Faculty of Natural Sciences, Constantine The Philosopher University, Nitra, Slovak Republic.
| |
Collapse
|
175
|
Abstract
Heparin-antithrombin interaction is one of the most documented examples of heparin/protein complexes. The specific heparin sequence responsible for the binding corresponds to a pentasaccharide sequence with an internal 3-O-sulfated glucosamine residue. Moreover, the position of the pentasaccharide along the chain as well as the structure of the neighbor units affects the affinity to antithrombin. The development of separation and purification techniques, in conjunction with physico-chemical approaches (mostly NMR), allowed to characterize several structural variants of antithrombin-binding oligosaccharides, both in the free state and in complex with antithrombin. The article provides an overview of the studies that lead to the elucidation of the mechanism of interaction as well as acquiring new knowledge in heparin biosynthesis.
Collapse
|
176
|
A fast capillary electrophoresis method to assess the binding affinity of recombinant antithrombin toward heparin directly from cell culture supernatants. J Pharm Biomed Anal 2015; 111:64-70. [DOI: 10.1016/j.jpba.2015.02.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 02/13/2015] [Accepted: 02/20/2015] [Indexed: 11/19/2022]
|
177
|
Trends in the development of oral anticoagulants. Ther Deliv 2015; 6:685-703. [DOI: 10.4155/tde.15.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Anticoagulation remains the therapy of choice for the prevention and treatment of venous and arterial thromboembolic disorders which can cause major organ damage or death. Heparins represent the antithrombotic drugs of choice in short and medium-term prophylaxis and therapy of thromboembolic diseases. Fondaparinux, a synthetic and structural analog of the antithrombin-binding pentasaccharide domain of heparin, has selective anti-Xa activity and longer half-life. However, anticoagulants are poorly absorbed by oral route because of their high molecular weight, hydrophilicity and negative charges. Long-term anticoagulation therapy is problematic because of side effects and frequent monitoring. Formulation approaches are particularly promising.
Collapse
|
178
|
Arnautova Y, Abagyan R, Totrov M. All-Atom Internal Coordinate Mechanics (ICM) Force Field for Hexopyranoses and Glycoproteins. J Chem Theory Comput 2015; 11:2167-2186. [PMID: 25999804 PMCID: PMC4431507 DOI: 10.1021/ct501138c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Indexed: 01/24/2023]
Abstract
We present an extension of the all-atom internal-coordinate force field, ICMFF, that allows for simulation of heterogeneous systems including hexopyranose saccharides and glycan chains in addition to proteins. A library of standard glycan geometries containing α- and β-anomers of the most common hexapyranoses, i.e., d-galactose, d-glucose, d-mannose, d-xylose, l-fucose, N-acetylglucosamine, N-acetylgalactosamine, sialic, and glucuronic acids, is created based on the analysis of the saccharide structures reported in the Cambridge Structural Database. The new force field parameters include molecular electrostatic potential-derived partial atomic charges and the torsional parameters derived from quantum mechanical data for a collection of minimal molecular fragments and related molecules. The ϕ/ψ torsional parameters for different types of glycosidic linkages are developed using model compounds containing the key atoms in the full carbohydrates, i.e., glycosidic-linked tetrahydropyran-cyclohexane dimers. Target data for parameter optimization include two-dimensional energy surfaces corresponding to the ϕ/ψ glycosidic dihedral angles in the disaccharide analogues, as determined by quantum mechanical MP2/6-31G** single-point energies on HF/6-31G** optimized structures. To achieve better agreement with the observed geometries of glycosidic linkages, the bond angles at the O-linkage atoms are added to the internal variable set and the corresponding bond bending energy term is parametrized using quantum mechanical data. The resulting force field is validated on glycan chains of 1-12 residues from a set of high-resolution X-ray glycoprotein structures based on heavy atom root-mean-square deviations of the lowest-energy glycan conformations generated by the biased probability Monte Carlo (BPMC) molecular mechanics simulations from the native structures. The appropriate BPMC distributions for monosaccharide-monosaccharide and protein-glycan linkages are derived from the extensive analysis of conformational properties of glycoprotein structures reported in the Protein Data Bank. Use of the BPMC search leads to significant improvements in sampling efficiency for glycan simulations. Moreover, good agreement with the X-ray glycoprotein structures is achieved for all glycan chain lengths. Thus, average/median RMSDs are 0.81/0.68 Å for one-residue glycans and 1.32/1.47 Å for three-residue glycans. RMSD from the native structure for the lowest-energy conformation of the 12-residue glycan chain (PDB ID 3og2) is 1.53 Å. Additionally, results obtained for free short oligosaccharides using the new force field are in line with the available experimental data, i.e., the most populated conformations in solution are predicted to be the lowest energy ones. The newly developed parameters allow for the accurate modeling of linear and branched hexopyranose glycosides in heterogeneous systems.
Collapse
Affiliation(s)
- Yelena
A. Arnautova
- Molsoft
L.L.C., 11199 Sorrento
Valley Road, S209, San Diego, California 92121, United States
| | - Ruben Abagyan
- Skaggs
School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - Maxim Totrov
- Molsoft
L.L.C., 11199 Sorrento
Valley Road, S209, San Diego, California 92121, United States
| |
Collapse
|
179
|
Terauchi M, Ikeda G, Nishida K, Tamura A, Yamaguchi S, Harada K, Yui N. Supramolecular Polyelectrolyte Complexes of Bone Morphogenetic Protein-2 with Sulfonated Polyrotaxanes to Induce Enhanced Osteogenic Differentiation. Macromol Biosci 2015; 15:953-64. [DOI: 10.1002/mabi.201500032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 03/04/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Masahiko Terauchi
- Department of Maxillofacial Surgery; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima; Bunkyo Tokyo 113-8549 Japan
- Department of Organic Biomaterials; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai; Chiyoda Tokyo 101-0062 Japan
| | - Go Ikeda
- Department of Organic Biomaterials; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai; Chiyoda Tokyo 101-0062 Japan
| | - Kei Nishida
- Department of Organic Biomaterials; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai; Chiyoda Tokyo 101-0062 Japan
| | - Atsushi Tamura
- Department of Organic Biomaterials; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai; Chiyoda Tokyo 101-0062 Japan
| | - Satoshi Yamaguchi
- Department of Maxillofacial Surgery; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima; Bunkyo Tokyo 113-8549 Japan
| | - Kiyoshi Harada
- Department of Maxillofacial Surgery; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima; Bunkyo Tokyo 113-8549 Japan
| | - Nobuhiko Yui
- Department of Organic Biomaterials; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai; Chiyoda Tokyo 101-0062 Japan
| |
Collapse
|
180
|
Polysulfated trehalose as a novel anticoagulant agent with dual mode of action. BIOMED RESEARCH INTERNATIONAL 2015; 2015:630482. [PMID: 25866798 PMCID: PMC4381846 DOI: 10.1155/2015/630482] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/26/2014] [Accepted: 08/28/2014] [Indexed: 12/01/2022]
Abstract
Physiological hemostatic balance is a coordinated outcome of counteracting coagulation and fibrinolytic systems. An imbalance of procoagulant and anticoagulant factors may result in life threatening thromboembolism. Presently, anticoagulant administration is the first line of therapy for the treatment of these conditions and several anticoagulants have been approved, including various forms of heparin. However, the polyanionic nature and multispecificity of heparin pose several complications. Generally, the polysulfated compounds with antithrombotic potential are thought to have feasible synthetic procedures with much less bleeding, thus having favourable safety profiles. Here we report the synthesis of a novel compound, trehalose octasulfate and the assessment of its anticoagulation potential. Molecular docking of trehalose and trehalose octasulfate with antithrombin showed a specificity switch in binding affinity on sulfation, where trehalose octasulfate interacts with critical residues of AT that are either directly involved in heparin binding or in the conformational rearrangement of AT on heparin binding. An in vitro analysis of trehalose octasulfate demonstrated prolonged clotting time. Lead compound when intravenously injected in occlusion induced thrombotic rats showed remarkable reduction in the size and weight of the clot at a low dose. Delay in coagulation time was observed by analysing blood plasma isolated from rats preinjected with trehalose octasulfate. A decrease in Adenosine 5′-Diphosphate (ADP) induced platelet aggregation indicated a probable dual anticoagulant and antiplatelet mechanism of action. To summarize, this study presents trehalose octasulfate as a novel, effective, dual acting antithrombotic agent.
Collapse
|
181
|
Wu M, Wen D, Gao N, Xiao C, Yang L, Xu L, Lian W, Peng W, Jiang J, Zhao J. Anticoagulant and antithrombotic evaluation of native fucosylated chondroitin sulfates and their derivatives as selective inhibitors of intrinsic factor Xase. Eur J Med Chem 2015; 92:257-69. [DOI: 10.1016/j.ejmech.2014.12.054] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/29/2014] [Accepted: 12/30/2014] [Indexed: 11/25/2022]
|
182
|
Ma Y, Wang J, Gao J, Yang H, Wang Y, Manithody C, Li J, Rezaie AR. Antithrombin up-regulates AMP-activated protein kinase signalling during myocardial ischaemia/reperfusion injury. Thromb Haemost 2015; 113:338-49. [PMID: 25230600 PMCID: PMC4308562 DOI: 10.1160/th14-04-0360] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 08/09/2014] [Indexed: 11/05/2022]
Abstract
Antithrombin (AT) is a protein of the serpin superfamily involved in regulation of the proteolytic activity of the serine proteases of the coagulation system. AT is known to exhibit anti-inflammatory and cardioprotective properties when it binds to heparan sulfate proteoglycans (HSPGs) on vascular cells. AMP-activated protein kinase (AMPK) plays an important cardioprotective role during myocardial ischaemia and reperfusion (I/R). To determine whether the cardioprotective signaling function of AT is mediated through the AMPK pathway, we evaluated the cardioprotective activities of wild-type AT and its two derivatives, one having high affinity and the other no affinity for heparin, in an acute I/R injury model in C57BL/6J mice in which the left anterior descending coronary artery was occluded. The serpin derivatives were given 5 minutes before reperfusion. The results showed that AT-WT can activate AMPK in both in vivo and ex vivo conditions. Blocking AMPK activity abolished the cardioprotective function of AT against I/R injury. The AT derivative having high affinity for heparin was more effective in activating AMPK and in limiting infraction, but the derivative lacking affinity for heparin was inactive in eliciting AMPK-dependent cardioprotective activity. Activation of AMPK by AT inhibited the inflammatory c-Jun N-terminal protein kinase (JNK) pathway during I/R. Further studies revealed that the AMPK activity induced by AT also modulates cardiac substrate metabolism by increasing glucose oxidation but inhibiting fatty acid oxidation during I/R. These results suggest that AT binds to HSPGs on heart tissues to invoke a cardioprotective function by triggering cardiac AMPK activation, thereby attenuating JNK inflammatory signalling pathways and modulating substrate metabolism during I/R.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ji Li
- Ji Li, PhD, Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, NY 14214, USA, Tel.: +1 716 829 5711, Fax: +1 716 829 2801, E-mail:
| | - Alireza R Rezaie
- Alireza R. Rezaie, PhD, Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Saint Louis, MO 63104, USA, Tel.: +1 314 977 9240, Fax: +1 314 977 9205, E-mail:
| |
Collapse
|
183
|
Faria-Oliveira F, Carvalho J, Belmiro CLR, Ramalho G, Pavão M, Lucas C, Ferreira C. Elemental biochemical analysis of the polysaccharides in the extracellular matrix of the yeastSaccharomyces cerevisiae. J Basic Microbiol 2015; 55:685-94. [DOI: 10.1002/jobm.201400781] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 12/08/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Fábio Faria-Oliveira
- Centre of Molecular and Environmental Biology (CBMA); Department of Biology; University of Minho; Portugal
| | - Joana Carvalho
- Centre of Molecular and Environmental Biology (CBMA); Department of Biology; University of Minho; Portugal
| | - Celso LR Belmiro
- Laboratory of Glycoconjugates Biochemistry and Cellular Biology; Federal University of Rio de Janeiro; Campus of Macaé RJ Brazil
- Laboratory of Glycoconjugates Biochemistry and Cellular Biology; Institute of Medical Biochemistry; Federal University of Rio de Janeiro; RJ Brazil
| | - Gustavo Ramalho
- Laboratory of Glycoconjugates Biochemistry and Cellular Biology; Institute of Medical Biochemistry; Federal University of Rio de Janeiro; RJ Brazil
| | - Mauro Pavão
- Laboratory of Glycoconjugates Biochemistry and Cellular Biology; Institute of Medical Biochemistry; Federal University of Rio de Janeiro; RJ Brazil
| | - Cândida Lucas
- Centre of Molecular and Environmental Biology (CBMA); Department of Biology; University of Minho; Portugal
| | - Célia Ferreira
- Centre of Molecular and Environmental Biology (CBMA); Department of Biology; University of Minho; Portugal
| |
Collapse
|
184
|
Leary JA, Miller RL, Wei W, Schwörer R, Zubkova OV, Tyler PC, Turnbull JE. Composition, sequencing and ion mobility mass spectrometry of heparan sulfate-like octasaccharide isomers differing in glucuronic and iduronic acid content. EUROPEAN JOURNAL OF MASS SPECTROMETRY (CHICHESTER, ENGLAND) 2015; 21:245-254. [PMID: 26307704 DOI: 10.1255/ejms.1337] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Here we report ion mobility mass spectrometry (IMMS) separation and tandem mass spectrometry (MS(2)) sequencing methods used to analyze and differentiate six synthetically produced heparin/heparan sulfate (HS)-like octasaccharide (dp8) isomeric structures. These structures are isomeric with regard to either glucuronic acid (GlcA) or iduronic acid (IdoA) residues at various positions. IMMS analysis showed that a fully GlcA structure exhibited a more compact conformation, whereas the fully IdoA structure was more extended. Interestingly, the change from IdoA to GlcA in specific locations resulted in strong conformational distortions. MS(2) of the six isomers showed very different spectra with unique sets of diagnostic product ions. Analysis of MS(2) product ion spectra suggests that the GlcA group correlated with the formation of a glycosidic product ion under lower energy conditions. This resulted in an earlier product ion formation and more intense product ions. Importantly, this knowledge enabled a complete sequencing of the positions of GlcA and IdoA in each of the four positions located in each unique dp8 structure.
Collapse
Affiliation(s)
- Julie A Leary
- De partments of Molecular and Cellular Biology and Chemistry, University of California, USA..
| | - Rebecca L Miller
- Departments of Molecular and Cellular Biology and Chemistry, University of California. Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK.
| | - Wei Wei
- Departments of Molecular and Cellular Biology and Chemistry, University of California, USA..
| | - Ralf Schwörer
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand.
| | - Olga V Zubkova
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand.
| | - Peter C Tyler
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Road, Gracefield, Lower Hutt 5010, New Zealand.
| | - Jeremy E Turnbull
- Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK.
| |
Collapse
|
185
|
Zhang X, Péréz-Sánchez H, Lightstone FC. Molecular Dynamics Simulations of Ligand Recognition upon Binding Antithrombin: A MM/GBSA Approach. BIOINFORMATICS AND BIOMEDICAL ENGINEERING 2015. [DOI: 10.1007/978-3-319-16480-9_56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
186
|
Lee TW, Yang ASP, Brittain T, Birch NP. An analysis approach to identify specific functional sites in orthologous proteins using sequence and structural information: application to neuroserpin reveals regions that differentially regulate inhibitory activity. Proteins 2015; 83:135-52. [PMID: 25363759 DOI: 10.1002/prot.24711] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 10/22/2014] [Accepted: 10/27/2014] [Indexed: 01/12/2023]
Abstract
The analysis of sequence conservation is commonly used to predict functionally important sites in proteins. We have developed an approach that first identifies highly conserved sites in a set of orthologous sequences using a weighted substitution-matrix-based conservation score and then filters these conserved sites based on the pattern of conservation present in a wider alignment of sequences from the same family and structural information to identify surface-exposed sites. This allows us to detect specific functional sites in the target protein and exclude regions that are likely to be generally important for the structure or function of the wider protein family. We applied our method to two members of the serpin family of serine protease inhibitors. We first confirmed that our method successfully detected the known heparin binding site in antithrombin while excluding residues known to be generally important in the serpin family. We next applied our sequence analysis approach to neuroserpin and used our results to guide site-directed polyalanine mutagenesis experiments. The majority of the mutant neuroserpin proteins were found to fold correctly and could still form inhibitory complexes with tissue plasminogen activator (tPA). Kinetic analysis of tPA inhibition, however, revealed altered inhibitory kinetics in several of the mutant proteins, with some mutants showing decreased association with tPA and others showing more rapid dissociation of the covalent complex. Altogether, these results confirm that our sequence analysis approach is a useful tool that can be used to guide mutagenesis experiments for the detection of specific functional sites in proteins.
Collapse
Affiliation(s)
- Tet Woo Lee
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
187
|
Sankaranarayanan NV, Sarkar A, Desai UR, Mosier PD. Designing "high-affinity, high-specificity" glycosaminoglycan sequences through computerized modeling. Methods Mol Biol 2015; 1229:289-314. [PMID: 25325961 DOI: 10.1007/978-1-4939-1714-3_24] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The prediction of high-affinity and/or high-specificity protein-glycosaminoglycan (GAG) interactions is an inherently difficult task, due to several factors including the shallow nature of the typical GAG-binding site and the inherent size, flexibility, diversity, and polydisperse nature of the GAG molecules. Here, we present a generally applicable methodology termed Combinatorial Library Virtual Screening (CVLS) that can identify potential high-affinity, high-specificity protein-GAG interactions from very large GAG combinatorial libraries and a suitable GAG-binding protein. We describe the CVLS approach along with the rationale behind it and provide validation for the method using the well-known antithrombin-thrombin-heparin system.
Collapse
Affiliation(s)
- Nehru Viji Sankaranarayanan
- Department of Medicinal Chemistry, Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University, 800 E. Leigh Street, Suite 212, Richmond, VA, 23298, USA
| | | | | | | |
Collapse
|
188
|
Zhang X, Meekins DA, An C, Zolkiewski M, Battaile KP, Kanost MR, Lovell S, Michel K. Structural and inhibitory effects of hinge loop mutagenesis in serpin-2 from the malaria vector Anopheles gambiae. J Biol Chem 2014; 290:2946-56. [PMID: 25525260 DOI: 10.1074/jbc.m114.625665] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Serpin-2 (SRPN2) is a key negative regulator of the melanization response in the malaria vector Anopheles gambiae. SRPN2 irreversibly inhibits clip domain serine proteinase 9 (CLIPB9), which functions in a serine proteinase cascade culminating in the activation of prophenoloxidase and melanization. Silencing of SRPN2 in A. gambiae results in spontaneous melanization and decreased life span and is therefore a promising target for vector control. The previously determined structure of SRPN2 revealed a partial insertion of the hinge region of the reactive center loop (RCL) into β sheet A. This partial hinge insertion participates in heparin-linked activation in other serpins, notably antithrombin III. SRPN2 does not contain a heparin binding site, and any possible mechanistic function of the hinge insertion was previously unknown. To investigate the function of the SRPN2 hinge insertion, we developed three SRPN2 variants in which the hinge regions are either constitutively expelled or inserted and analyzed their structure, thermostability, and inhibitory activity. We determined that constitutive hinge expulsion resulted in a 2.7-fold increase in the rate of CLIPB9Xa inhibition, which is significantly lower than previous observations of allosteric serpin activation. Furthermore, we determined that stable insertion of the hinge region did not appreciably decrease the accessibility of the RCL to CLIPB9. Together, these results indicate that the partial hinge insertion in SRPN2 does not participate in the allosteric activation observed in other serpins and instead represents a molecular trade-off between RCL accessibility and efficient formation of an inhibitory complex with the cognate proteinase.
Collapse
Affiliation(s)
- Xin Zhang
- From the Division of Biology, Kansas State University, Manhattan, Kansas 66506
| | - David A Meekins
- From the Division of Biology, Kansas State University, Manhattan, Kansas 66506
| | - Chunju An
- From the Division of Biology, Kansas State University, Manhattan, Kansas 66506, the Department of Entomology, College of Agriculture and Biotechnology, China Agricultural University, Beijing, China
| | - Michal Zolkiewski
- the Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506
| | - Kevin P Battaile
- Industrial Macromolecular Crystallography Association Collaborative Access Team, Hauptman-Woodward Medical Research Institute, Advanced Photon Source Argonne National Laboratory, Argonne, Illinois 60439, and
| | - Michael R Kanost
- the Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas 66506
| | - Scott Lovell
- the Protein Structure Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, Kansas 66407
| | - Kristin Michel
- From the Division of Biology, Kansas State University, Manhattan, Kansas 66506,
| |
Collapse
|
189
|
Izaguirre G, Aguila S, Qi L, Swanson R, Roth R, Rezaie AR, Gettins PGW, Olson ST. Conformational activation of antithrombin by heparin involves an altered exosite interaction with protease. J Biol Chem 2014; 289:34049-64. [PMID: 25331949 DOI: 10.1074/jbc.m114.611707] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Heparin allosterically activates antithrombin as an inhibitor of factors Xa and IXa by enhancing the initial Michaelis complex interaction of inhibitor with protease through exosites. Here, we investigate the mechanism of this enhancement by analyzing the effects of alanine mutations of six putative antithrombin exosite residues and three complementary protease exosite residues on antithrombin reactivity with these proteases in unactivated and heparin-activated states. Mutations of antithrombin Tyr(253) and His(319) exosite residues produced massive 10-200-fold losses in reactivity with factors Xa and IXa in both unactivated and heparin-activated states, indicating that these residues made critical attractive interactions with protease independent of heparin activation. By contrast, mutations of Asn(233), Arg(235), Glu(237), and Glu(255) exosite residues showed that these residues made both repulsive and attractive interactions with protease that depended on the activation state and whether the critical Tyr(253)/His(319) residues were mutated. Mutation of factor Xa Arg(143), Lys(148), and Arg(150) residues that interact with the exosite in the x-ray structure of the Michaelis complex confirmed the importance of all residues for heparin-activated antithrombin reactivity and Arg(150) for native serpin reactivity. These results demonstrate that the exosite is a key determinant of antithrombin reactivity with factors Xa and IXa in the native as well as the heparin-activated state and support a new model of allosteric activation we recently proposed in which a balance between attractive and repulsive exosite interactions in the native state is shifted to favor the attractive interactions in the activated state through core conformational changes induced by heparin binding.
Collapse
Affiliation(s)
- Gonzalo Izaguirre
- From the Department of Periodontics, Center for Molecular Biology of Oral Diseases and
| | - Sonia Aguila
- the Centro Regional de Hemodonación, University of Murcia, Murcia 30003, Spain, and
| | - Lixin Qi
- From the Department of Periodontics, Center for Molecular Biology of Oral Diseases and
| | - Richard Swanson
- From the Department of Periodontics, Center for Molecular Biology of Oral Diseases and
| | - Ryan Roth
- From the Department of Periodontics, Center for Molecular Biology of Oral Diseases and
| | - Alireza R Rezaie
- the Department of Biochemistry and Molecular Biology, St. Louis University, St. Louis, Missouri 63104
| | - Peter G W Gettins
- the Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois 60612
| | - Steven T Olson
- From the Department of Periodontics, Center for Molecular Biology of Oral Diseases and
| |
Collapse
|
190
|
Driguez PA, Potier P, Trouilleux P. Synthetic oligosaccharides as active pharmaceutical ingredients: lessons learned from the full synthesis of one heparin derivative on a large scale. Nat Prod Rep 2014; 31:980-9. [PMID: 24705477 DOI: 10.1039/c4np00012a] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Covering: up to November 2013. Heparin and heparan sulfate are natural polysaccharides with strong structural variations, which are responsible for their numerous specific biological properties. One key target of heparin, among others, is antithrombin, a serine protease inhibitor that, upon activation, mainly targets anticoagulation factors IIa and Xa. It is well documented that inhibition of the latter is due to a specific pentasaccharidic sequence, its synthetic analog being the registered drug fondaparinux. The replacement of hydroxyls by methoxy groups, N-sulfates by O-sulfonates and the modulation of the sulfation pattern gave rise to both idraparinux and its neutralizable form, idrabiotaparinux, two pentasaccharides with a significantly increased half-life compared to fondaparinux. Although numerous efforts have been devoted to improving the chemoenzymatic preparation of heparin fragments, enzymes are usually selective for their natural substrates, which limits the generation of some specific non-natural structures. Up to now, total synthesis has proved to be a valuable approach for the preparation of tailor-made and pure saccharides in the milligram to gram scale. This highlight will focus on the synthesis and the technical challenges associated with the development and the production of complex carbohydrates which will be exemplified with idrabiotaparinux. Particular attention will be paid to the process improvements needed in order to implement the production in a pilot plant, achieving batch generation on a multi-kilogram scale with a purity higher than 99.5%, and with no unknown impurity over 0.1%.
Collapse
Affiliation(s)
- Pierre-Alexandre Driguez
- Sanofi R&D, Early to Candidate Unit, 1 Avenue Pierre Brossolette, 91385 Chilly-Mazarin Cedex, France
| | | | | |
Collapse
|
191
|
Gerlza T, Hecher B, Jeremic D, Fuchs T, Gschwandtner M, Falsone A, Gesslbauer B, Kungl AJ. A combinatorial approach to biophysically characterise chemokine-glycan binding affinities for drug development. Molecules 2014; 19:10618-34. [PMID: 25054442 PMCID: PMC6271861 DOI: 10.3390/molecules190710618] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/07/2014] [Accepted: 07/16/2014] [Indexed: 01/06/2023] Open
Abstract
Chemokine binding to glycosaminoglycans (GAGs) is recognised to be an important step in inflammation and other pathological disorders like tumor growth and metastasis. Although different ways and strategies to interfere with these interactions are being pursued, no major breakthrough in the development of glycan-targeting drugs has been reported so far. We have engineered CXCL8 towards a dominant-negative form of this chemokine (dnCXCL8) which was shown to be highly active in various inflammatory animal models due to its inability to bind/activate the cognate CXCL8 GPC receptors on neutrophils in combination with its significantly increased GAG-binding affinity [1]. For the development of GAG-targeting chemokine-based biopharmaceuticals, we have established a repertoire of methods which allow the quantification of protein-GAG interactions. Isothermal fluorescence titration (IFT), surface plasmon resonance (SPR), isothermal titration calorimetry (ITC), and a novel ELISA-like competition assay (ELICO) have been used to determine Kd and IC50 values for CXCL8 and dnCXCL8 interacting with heparin and heparan sulfate (HS), the proto-typical members of the GAG family. Although the different methods gave different absolute affinities for the four protein-ligand pairs, the relative increase in GAG-binding affinity of dnCXCL8 compared to the wild type chemokine was found by all methods. In combination, these biophysical methods allow to discriminate between unspecific and specific protein-GAG interactions.
Collapse
Affiliation(s)
- Tanja Gerlza
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Humboldtstrasse 46, A-8010 Graz, Austria
| | - Bianca Hecher
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Humboldtstrasse 46, A-8010 Graz, Austria
| | - Dalibor Jeremic
- ProtAffin Biotechnologie AG, Reininghausstrasse 13a, A-802 Graz, Austria
| | - Thomas Fuchs
- ProtAffin Biotechnologie AG, Reininghausstrasse 13a, A-802 Graz, Austria
| | | | - Angelika Falsone
- ProtAffin Biotechnologie AG, Reininghausstrasse 13a, A-802 Graz, Austria
| | - Bernd Gesslbauer
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Humboldtstrasse 46, A-8010 Graz, Austria
| | - Andreas J Kungl
- Institute of Pharmaceutical Sciences, Karl-Franzens-University Graz, Humboldtstrasse 46, A-8010 Graz, Austria.
| |
Collapse
|
192
|
Sankaranarayanan NV, Desai UR. Toward a robust computational screening strategy for identifying glycosaminoglycan sequences that display high specificity for target proteins. Glycobiology 2014; 24:1323-33. [PMID: 25049239 DOI: 10.1093/glycob/cwu077] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Glycosaminoglycans (GAGs) interact with many proteins to regulate processes such as hemostasis, cell adhesion, growth and differentiation and viral infection. Yet, majority of these interactions remain poorly understood at a molecular level. A major reason for this state is the phenomenal structural diversity of GAGs, which has precluded analysis of specificity of their interactions. We had earlier presented a computational protocol for predicting "high-specificity" GAG sequences based on combinatorial virtual library screening (CVLS) technology. In this work, we expand the robustness of this technology through rigorous studies of parameters affecting GAG recognition of proteins, especially antithrombin and thrombin. The CVLS approach involves automated construction of a virtual library of all possible oligosaccharide sequences (di- to octasaccharide) followed by a two-step selection strategy consisting of "affinity" (GOLD score) and "specificity" (consistency of binding) filters. We find that "specificity" features are optimally evaluated using 100 genetic algorithm experiments, 100,000 evolutions and variable docking radius from 10 Å (disaccharide) to 14 Å (hexasaccharide). The results highlight critical interactions in H/HS oligosaccharides that govern specificity. Application of CVLS technology to the antithrombin-heparin system indicates that the minimal "specificity" element is the GlcAp(1 → 4)GlcNp2S3S disaccharide of heparin. The CVLS technology affords a simple, intuitive framework for the design of longer GAG sequences that can exhibit high "specificity" without resorting to exhaustive screening of millions of theoretical sequences.
Collapse
Affiliation(s)
- Nehru Viji Sankaranarayanan
- Department of Medicinal Chemistry and Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University, 800 E. Leigh Street, Suite 212, Richmond, VA 23219, USA
| | - Umesh R Desai
- Department of Medicinal Chemistry and Institute for Structural Biology and Drug Discovery, Virginia Commonwealth University, 800 E. Leigh Street, Suite 212, Richmond, VA 23219, USA
| |
Collapse
|
193
|
Yang L, Dinarvand P, Qureshi SH, Rezaie AR. Engineering D-helix of antithrombin in alpha-1-proteinase inhibitor confers antiinflammatory properties on the chimeric serpin. Thromb Haemost 2014; 112:164-75. [PMID: 24522239 PMCID: PMC4087087 DOI: 10.1160/th13-12-1029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/29/2014] [Indexed: 01/20/2023]
Abstract
Antithrombin (AT) is a heparin-binding serpin in plasma which regulates the proteolytic activity of procoagulant proteases of the clotting cascade. In addition to being an anticoagulant, AT also exhibits antiinflammatory activities when it binds to cell surface heparan sulfate proteoglycans (HSPGs) on the endothelium via its basic residues of D-helix to elicit intracellular signalling responses. By contrast to AT, α1-proteinase inhibitor (α1-PI) is a non-heparin-binding serpin that exhibits very slow reactivity with coagulation proteases and possesses no HSPG-dependent antiinflammatory properties. To determine whether the antiinflammatory signaling specificity of AT can be transferred to α1-PI, we replaced the D-helix of human α1-PI with the corresponding sequence of human AT and expressed the chimeric serpin α1-PI/D-helix) in a bacterial expression system. High molecular weight heparin bound to α1-PI/D-helix and accelerated the inhibition of thrombin by the serpin mutant by a template mechanism reminiscent of the cofactor effect of heparin on inhibition of thrombin by AT. Like AT, α1-PI/D-helix exhibited antiinflammatory properties in both cellular and animal models. Thus, α1-PI/D-helix inhibited the barrier-disruptive effect of proinflammatory cytokines and inhibited the activation of nuclear factor-κB transcription factor in lipopolysaccharide-stimulated endothelial cells by a concentration-dependent manner. Furthermore, the chimeric serpin reduced lipopolysaccharide-mediated lethality, elicited a vascular protective effect and inhibited infiltration of activated leukocytes to the peritoneal cavity of mice in an HMGB1-mediated inflammatory model. These results suggest that grafting the D-helix of AT to α1-PI confers antiinflammatory properties on the serpin and that the chimeric serpin may have therapeutic utility for treating inflammatory disorders.
Collapse
Affiliation(s)
| | | | | | - A R Rezaie
- Alireza R. Rezaie, PhD, Department of Biochemistry and Molecular Biology, St. Louis University School of Medicine, 1100 S. Grand Blvd., St. Louis, MO 63104, USA, Tel.: +1 314 977 9240, Fax: +1 314 977 9205 , E-mail:
| |
Collapse
|
194
|
Casu B, Naggi A, Torri G. Re-visiting the structure of heparin. Carbohydr Res 2014; 403:60-8. [PMID: 25088334 DOI: 10.1016/j.carres.2014.06.023] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 06/22/2014] [Indexed: 01/12/2023]
Abstract
The sulfated polysaccharide heparin has been used as a life-saving anticoagulant in clinics well before its detailed structure was known. This mini-review is a survey of the evolution in the discovery of the primary and secondary structure of heparin. Highlights in this history include elucidation and synthesis of the specific sequence that binds to antithrombin, the development of low-molecular-weight heparins currently used as antithrombotic drugs, and the most promising start of chemo-enzymatic synthesis. Special emphasis is given to peculiar conformational properties contributing to interaction with proteins that modulate different biological properties.
Collapse
Affiliation(s)
- Benito Casu
- G. Ronzoni Institute for Chemical and Biochemical Research, via G. Colombo, 81 20133 Milan, Italy.
| | - Annamaria Naggi
- G. Ronzoni Institute for Chemical and Biochemical Research, via G. Colombo, 81 20133 Milan, Italy
| | - Giangiacomo Torri
- G. Ronzoni Institute for Chemical and Biochemical Research, via G. Colombo, 81 20133 Milan, Italy
| |
Collapse
|
195
|
Agostino M, Gandhi NS, Mancera RL. Development and application of site mapping methods for the design of glycosaminoglycans. Glycobiology 2014; 24:840-51. [DOI: 10.1093/glycob/cwu045] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
196
|
Rienks M, Papageorgiou AP, Frangogiannis NG, Heymans S. Myocardial extracellular matrix: an ever-changing and diverse entity. Circ Res 2014; 114:872-88. [PMID: 24577967 DOI: 10.1161/circresaha.114.302533] [Citation(s) in RCA: 232] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The cardiac extracellular matrix (ECM) is a complex architectural network consisting of structural and nonstructural proteins, creating strength and plasticity. The nonstructural compartment of the ECM houses a variety of proteins, which are vital for ECM plasticity, and can be divided into 3 major groups: glycoproteins, proteoglycans, and glycosaminoglycans. The common denominator for these groups is glycosylation, which refers to the decoration of proteins or lipids with sugars. This review will discuss the fundamental role of the matrix in cardiac development, homeostasis, and remodeling, from a glycobiology point of view. Glycoproteins (eg, thrombospondins, secreted protein acidic and rich in cysteine, tenascins), proteoglycans (eg, versican, syndecans, biglycan), and glycosaminoglycans (eg, hyaluronan, heparan sulfate) are upregulated on cardiac injury and regulate key processes in the remodeling myocardium such as inflammation, fibrosis, and angiogenesis. Albeit some parallels can be made regarding the processes these proteins are involved in, their specific functions are extremely diverse. In fact, under varying conditions, individual proteins can even have opposing functions, making spatiotemporal contribution of these proteins in the rearrangement of multifaceted ECM very hard to grasp. Alterations of protein characteristics by the addition of sugars may explain the immense, yet tightly regulated, variability of the remodeling cardiac matrix. Understanding the role of glycosylation in altering the ultimate function of glycoproteins, proteoglycans, and glycosaminoglycans in the myocardium may lead to the development of new biochemical structures or compounds with great therapeutic potential for patients with heart disease.
Collapse
Affiliation(s)
- Marieke Rienks
- From Maastricht University Medical Centre, Maastricht, The Netherlands
| | | | | | | |
Collapse
|
197
|
Hsieh PH, Xu Y, Keire DA, Liu J. Chemoenzymatic synthesis and structural characterization of 2-O-sulfated glucuronic acid-containing heparan sulfate hexasaccharides. Glycobiology 2014; 24:681-92. [PMID: 24770491 DOI: 10.1093/glycob/cwu032] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Heparan sulfate and heparin are highly sulfated polysaccharides that consist of a repeating disaccharide unit of glucosamine and glucuronic or iduronic acid. The 2-O-sulfated iduronic acid (IdoA2S) residue is commonly found in heparan sulfate and heparin; however, 2-O-sulfated glucuronic acid (GlcA2S) is a less abundant monosaccharide (∼<5% of total saccharides). Here, we report the synthesis of three GlcA2S-containing hexasaccharides using a chemoenzymatic approach. For comparison purposes, additional IdoA2S-containing hexasaccharides were synthesized. Nuclear magnetic resonance analyses were performed to obtain full chemical shift assignments for the GlcA2S- and IdoA2S-hexasaccharides. These data show that GlcA2S is a more structurally rigid saccharide residue than IdoA2S. The antithrombin (AT) binding affinities of a GlcA2S- and an IdoA2S-hexasaccharide were determined by affinity co-electrophoresis. In contrast to IdoA2S-hexasaccharides, the GlcA2S-hexasaccharide does not bind to AT, confirming that the presence of IdoA2S is critically important for the anticoagulant activity. The availability of pure synthetic GlcA2S-containing oligosaccharides will allow the investigation of the structure and activity relationships of individual sites in heparin or heparan sulfate.
Collapse
Affiliation(s)
- Po-Hung Hsieh
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Rm 303, Beard Hall, Chapel Hill, NC 27599, USA
| | - Yongmei Xu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Rm 303, Beard Hall, Chapel Hill, NC 27599, USA
| | - David A Keire
- Food & Drug Administration, CDER (Center for Drug Evaluation and Research), Division of Pharmaceutical Analysis, 645 S Newstead Avenue, St. Louis, MO 63110, USA
| | - Jian Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina, Rm 303, Beard Hall, Chapel Hill, NC 27599, USA
| |
Collapse
|
198
|
Malik A, Lee J, Lee J. Community-based network study of protein-carbohydrate interactions in plant lectins using glycan array data. PLoS One 2014; 9:e95480. [PMID: 24755681 PMCID: PMC3995809 DOI: 10.1371/journal.pone.0095480] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 03/27/2014] [Indexed: 12/14/2022] Open
Abstract
Lectins play major roles in biological processes such as immune recognition and regulation, inflammatory responses, cytokine signaling, and cell adhesion. Recently, glycan microarrays have shown to play key roles in understanding glycobiology, allowing us to study the relationship between the specificities of glycan binding proteins and their natural ligands at the omics scale. However, one of the drawbacks in utilizing glycan microarray data is the lack of systematic analysis tools to extract information. In this work, we attempt to group various lectins and their interacting carbohydrates by using community-based analysis of a lectin-carbohydrate network. The network consists of 1119 nodes and 16769 edges and we have identified 3 lectins having large degrees of connectivity playing the roles of hubs. The community based network analysis provides an easy way to obtain a general picture of the lectin-glycan interaction and many statistically significant functional groups.
Collapse
Affiliation(s)
- Adeel Malik
- Center for In Silico Protein Science, School of Computational Sciences, Korea Institute for Advanced Study, Seoul, Korea
| | - Juyong Lee
- Center for In Silico Protein Science, School of Computational Sciences, Korea Institute for Advanced Study, Seoul, Korea
| | - Jooyoung Lee
- Center for In Silico Protein Science, School of Computational Sciences, Korea Institute for Advanced Study, Seoul, Korea
- * E-mail:
| |
Collapse
|
199
|
Abstract
![]()
A series of self-associating foldamers
have been designed as heparin
reversal agents, as antidotes to prevent bleeding due to this potent
antithrombotic agent. The foldamers have a repeating sequence of Lys-Sal,
in which Sal is 5-amino-2-methoxy-benzoic acid. These foldamers are
designed to self-associate along one face of an extended chain in
a β-sheet-like interaction. The methoxy groups were included
to form intramolecular hydrogen bonds that preclude the formation
of very large amyloid-like aggregates, while the positively charged
Lys side chains were introduced to interact electrostatically with
the highly anionic heparin polymer. The prototype compound (Lys-Sal)4 carboxamide weakly associates in aqueous solution at physiological
salt concentration in a monomer-dimer-hexamer equilibrium. The association
is greatly enhanced at either high ionic strength or in the presence
of a heparin derivative, which is bound tightly. Variants of this
foldamer are active in an antithrombin III–factor Xa assay,
showing their potential as heparin reversal agents.
Collapse
|
200
|
Dinges MM, Solakyildirim K, Larive CK. Affinity capillary electrophoresis for the determination of binding affinities for low molecular weight heparins and antithrombin-III. Electrophoresis 2014; 35:1469-77. [DOI: 10.1002/elps.201300549] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 02/20/2014] [Accepted: 02/21/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Meredith M. Dinges
- Department of Chemistry; University of California-Riverside; Riverside CA USA
| | - Kemal Solakyildirim
- Department of Chemistry; University of California-Riverside; Riverside CA USA
| | - Cynthia K. Larive
- Department of Chemistry; University of California-Riverside; Riverside CA USA
| |
Collapse
|