Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Glycoconj J ; 28(8-9): 525-35, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21948150

ABSTRACT

The rare N-unsubstituted glucosamine (GlcNH (3)(+)) residues in heparan sulfate (HS) have important biological and pathophysiological roles. However, it is difficult to prepare naturally-occuring, GlcNH(3)(+)-containing oligosaccharides from HS because of their low abundance, as well as the inherent problems in both excising and identifying them. Therefore, the ability to chemically generate a series of structurally-defined oligosaccharides containing GlcNH(3)(+) residues would greatly contribute to investigating their natural role in HS. In this study, a series of heparin/HS oligosaccharides, from dp6 up to dp16 in length that possess internal GlcNH(3)(+) residues were prepared by a combination of chemical modification and heparinase I digestion. Purification and structural analysis of the major species derived from the octa- to dodeca-saccharide size fractions indicated the introduction of between 1 and 3 internal GlcNH(3)(+) residues per oligosaccharide. In addition, a GlcNH(3)(+) residue was selectively introduced into an internal position in a tetrasaccharide species by direct chemical modification. This selectivity has potential as an alternative procedure for preparing internally-modified oligosaccharides of various lengths. The utility of such oligosaccharides was demonstrated by a comparison of the binding of three different tetrasaccharide species containing 0, 1 and 2 free amino groups to the NK1 truncated variant of hepatocyte growth factor/scatter factor.


Subject(s)
Glucosamine/metabolism , Heparin/chemical synthesis , Heparitin Sulfate/chemical synthesis , Oligosaccharides/chemical synthesis , Animals , Cattle , Chemical Fractionation , Chromatography, Gel , Chromatography, High Pressure Liquid , Disaccharides/analysis , Heparin/analogs & derivatives , Heparin/chemistry , Heparitin Sulfate/chemistry , Hepatocyte Growth Factor/metabolism , Humans , Magnetic Resonance Spectroscopy , Oligosaccharides/chemistry , Oligosaccharides/isolation & purification , Protein Binding , Sulfotransferases/metabolism
2.
Biochemistry ; 49(49): 10486-95, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21062008

ABSTRACT

Human ß-defensin 2 (HBD2) is a member of the defensin family of antimicrobial peptides that plays important roles in the innate and adaptive immune system of both vertebrates and invertebrates. In addition to their direct bactericidal action, defensins are also involved in chemotaxis and Toll-like receptor activation. In analogy to chemokine/glycosaminoglycan (GAG) interactions, GAG-defensin complexes are likely to play an important role in chemotaxis and in presenting defensins to their receptors. Using a gel mobility shift assay, we found that HBD2 bound to a range of GAGs including heparin/heparan sulfate (HS), dermatan sulfate (DS), and chondroitin sulfate. We used NMR spectroscopy of (15)N-labeled HBD2 to map the binding sites for two GAG model compounds, a heparin/HS pentasaccharide (fondaparinux sodium; FX) and enzymatically prepared DS hexasaccharide (DSdp6). We identified a number of basic amino acids that form a common ligand binding site, which indicated that these interactions are predominantly electrostatic. The dissociation constant of the [DSdp6-HBD2] complex was determined by NMR spectroscopy to be 5 ± 5 µM. Binding of FX could not be quantified because of slow exchange on the NMR chemical shift time scale. FX was found to induce HBD2 dimerization as evidenced by the analysis of diffusion coefficients, (15)N relaxation, and nESI-MS measurements. The formation of FX-bridged HBD2 dimers exhibited features of a cooperative binding mechanism. In contrast, the complex with DSdp6 was found to be mostly monomeric.


Subject(s)
Glycosaminoglycans/chemistry , Glycosaminoglycans/metabolism , beta-Defensins/chemistry , beta-Defensins/metabolism , Binding Sites/physiology , Chemotaxis, Leukocyte/physiology , Humans , Magnetic Resonance Spectroscopy , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Spectrometry, Mass, Electrospray Ionization , Static Electricity , Sulfates/chemistry , Sulfates/metabolism
3.
Glycobiology ; 20(11): 1380-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20581009

ABSTRACT

Endocan is a recently identified soluble chondroitin/dermatan sulfate (CS/DS) proteoglycan. Synthesized by endothelial cells, it has been found to be over-expressed in the vasculature surrounding a number of tumors, and by promoting growth factor mitogenic activities, hepatocyte growth factor/scatter factor (HGF/SF) in particular, it supports cellular proliferation. In this work, we characterized the glycosaminoglycan (GAG) chain of Endocan, purified either from the naturally producing human umbilical vein endothelial cells (HUVEC) or from a recombinant over-expression system in human embryonic kidney cells (HEK). Compositional analysis using different chondroitinases as well as nuclear magnetic resonance studies revealed that the GAG chains from both sources share many characteristics, with the exception of size (15 and 40 kDa, respectively, for HUVEC and HEK-293 cells). The DS-specific, IdoA-containing disaccharides contribute 30% of the chain (15% of which are 2-O-sulfated) and are mostly clustered in tetra- (35%), hexa- (12%), and octa- (5%) saccharide domains. Highly sulfated D, E, and B disaccharide units (HexA2S-GalNAc6S, HexA-GalNAc4S6S, and HexA2S-GalNAc4S) were also detected in significant amounts in both chains and may account for the HGF/SF-binding activity of the CS/DS. This work establishes that HEK-293 cells can be engineered to provide a valuable source of Endocan with authentic CS/DS chains, enabling the purification of sufficient amounts for structural and/or binding analysis and providing a possible model of Endocan CS/DS chain organization.


Subject(s)
Chondroitin/metabolism , Dermatan Sulfate/metabolism , Proteoglycans/metabolism , Binding Sites , Cells, Cultured , Chromatography, Gel , Humans , Magnetic Resonance Spectroscopy
4.
J Am Chem Soc ; 132(18): 6374-81, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20394361

ABSTRACT

We have used the interaction between module 7 of complement factor H (CFH approximately 7) and a fully sulfated heparin tetrasaccharide to exemplify a new approach for studying contributions of basic side chains to the formation of glycosaminoglycan (GAG)-protein complexes. We first employed HISQC and H(2)CN NMR experiments to monitor the side-chain resonances of lysines and arginines in (15)N, (13)C-labeled protein during titrations with a fully sulfated heparin tetrasaccharide under physiological conditions. Under identical conditions and using (15)N-labeled protein, we then cross-linked tetrasaccharide to CFH approximately 7 and confirmed the 1:1 stoichiometry by FT-ICR-MS. We subsequently characterized this covalent protein-GAG conjugate by NMR and further MS techniques. MALDI-TOF MS identified protein fragments obtained via trypsin digestion or chemical fragmentation, yielding information concerning the site of GAG attachment. Combining MS and NMR data allowed us to identify the side chain of K405 as the point of attachment of the cross-linked heparin oligosaccharide to CFH approximately 7. On the basis of the analysis of NMR and MS data of the noncovalent and cross-linked CFH approximately 7-tetrasaccharide complexes, we conclude that the K446 side chain is not essential for binding the tetrasaccharide, despite the large chemical shift perturbations of its backbone amide (15)N and (1)H resonances during titrations. We show that R444 provides the most important charge-charge interaction within a C-terminal heparin-binding subsite of CFH approximately 7 whereas side chains of R404, K405, and K388 are the predominant contributors to an N-terminal binding subsite located in the immediate vicinity of residue 402, which is implicated in age-related macular degeneration (AMD).


Subject(s)
Arginine/chemistry , Complement Factor H/chemistry , Complement Factor H/metabolism , Cross-Linking Reagents/pharmacology , Heparin/metabolism , Lysine/chemistry , Mass Spectrometry , Amino Acid Sequence , Heparin/pharmacology , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Multimerization/drug effects , Protein Structure, Quaternary , Solutions , Trypsin/metabolism
5.
J Immunol ; 181(4): 2610-9, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18684951

ABSTRACT

Human complement factor H, consisting of 20 complement control protein (CCP) modules, is an abundant plasma glycoprotein. It prevents C3b amplification on self surfaces bearing certain polyanionic carbohydrates, while complement activation progresses on most other, mainly foreign, surfaces. Herein, locations of binding sites for polyanions and C3b are reexamined rigorously by overexpressing factor H segments, structural validation, and binding assays. As anticipated, constructs corresponding to CCPs 7-8 and 19-20 bind well in heparin-affinity chromatography. However, CCPs 8-9, previously reported to bind glycosaminoglycans, bind neither to heparin resin nor to heparin fragments in gel-mobility shift assays. Introduction of nonnative residues N-terminal to a construct containing CCPs 8-9, identical to those in proteins used in the previous report, converted this module pair to an artificially heparin-binding one. The module pair CCPs 12-13 does not bind heparin appreciably, notwithstanding previous suggestions to the contrary. We further checked CCPs 10-12, 11-14, 13-15, 10-15, and 8-15 for ability to bind heparin but found very low affinity or none. As expected, constructs corresponding to CCPs 1-4 and 19-20 bind C3b amine coupled to a CM5 chip (K(d)s of 14 and 3.5 microM, respectively) or a C1 chip (K(d)s of 10 and 4.5 microM, respectively). Constructs CCPs 7-8 and 6-8 exhibit measurable affinities for C3b according to surface plasmon resonance, although they are weak compared with CCPs 19-20. Contrary to expectations, none of several constructs encompassing modules from CCP 9 to 15 exhibited significant C3b binding in this assay. Thus, we propose a new functional map of factor H.


Subject(s)
Complement C3b/metabolism , Complement Factor H/metabolism , Glycosaminoglycans/metabolism , Binding Sites/immunology , Chromatography, Affinity , Complement C3b/chemistry , Complement Factor H/chemistry , Complement Factor H/genetics , Complement Pathway, Alternative/immunology , Complement System Proteins/chemistry , Complement System Proteins/metabolism , Glycosaminoglycans/biosynthesis , Glycosaminoglycans/genetics , Heparin/metabolism , Humans , Magnetic Resonance Spectroscopy , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Mapping , Polyelectrolytes , Polymers/metabolism , Protein Folding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
6.
Glycobiology ; 19(11): 1185-96, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19648354

ABSTRACT

The solution conformation of a fully sulfated heparin-derived tetrasaccharide, I, was studied in the presence of a 4-fold excess of Ca(2+). Proton-proton and proton-carbon residual dipolar couplings (RDCs) were measured in a neutral aligning medium. The order parameters of two rigid hexosamine rings of I were determined separately using singular value decomposition and ab initio structures of disaccharide fragments of I. The order parameters were very similar implying that a common order tensor can be used to analyze the structure of I. Using one order tensor, RDCs of both hexosamine rings were used as restraints in molecular dynamics simulations. RDCs of the inner iduronic acid were calculated for every point of the molecular dynamics trajectory. The fitting of the calculated RDCs of the two forms of the iduronic acid to the experimental values yielded a population of (1)C(4) and (2)S(o) conformers of iduronic acid that agreed well with the analysis based on proton-proton scalar coupling constants. The glycosidic linkage torsion angles in RDC-restrained molecular dynamics (MD) structures of I are consistent with the interglycosidic three-bond proton-carbon coupling constants. These structures also show that the shape of heparin is not affected dramatically by the conformational flexibility of the iduronic acid ring. This is in line with conclusions of previous studies based on MD simulations and the analysis of (1)H-(1)H NOEs. Our work therefore demonstrates the effectiveness of RDCs in the conformational analysis of glycosaminoglycans.


Subject(s)
Heparin/chemistry , Iduronic Acid/chemistry , Models, Chemical , Oligosaccharides/chemistry , Quantum Theory , Carbohydrate Conformation , Carbon/chemistry , Computer Simulation , Protons
7.
Glycobiology ; 18(6): 483-91, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18378523

ABSTRACT

Sulfated glycosaminoglycans regulate the biological functions of a wide variety of proteins, primarily through high affinity interactions mediated by specific sugar sequences or patterns/densities of sulfation. Disaccharide analysis of such glycosaminoglycans yields important diagnostic and comparative structural information on sulfate patterning. When applied to specific oligosaccharides it can also make a vital contribution to sequence elucidation. Standard UV detection of lyase-generated disaccharides resolved by HPLC can lack sufficient sensitivity and be compromised by contaminating UV signals, when dealing with scarce tissue- or cell culture-derived material. Various methods exist for improved detection, but usually involve additional HPLC hardware and often necessitate different procedures for analyzing different glycosaminoglycans. We describe a simple procedure, requiring only standard HPLC instrumentation, involving prederivatization of disaccharides with 2-aminoacridone with no cleanup of samples, followed by a separation by reverse-phase HPLC that is sensitive to as little as approximately 100 pg (approximately 10(-13) mol) of an individual disaccharide, thereby allowing analyses of >10 ng of total glycosaminoglycan. Importantly, separate analysis of both HS/heparin and CS/DS species within a mixed glycosaminoglycan pool can be performed using the same procedure on a single column. We demonstrate its applicability in dealing with small quantities of material derived from rat liver (where we demonstrate a high abundance of the unusual CS-E species within the CS/DS pool) and MDCK cells (which revealed a HS species of relatively low N-sulfation, but high O-sulfation). This simplified method should find a widespread utility for analyzing glycosaminoglycans from limited animal and cell culture samples.


Subject(s)
Chondroitin Sulfates/analysis , Dermatan Sulfate/analysis , Disaccharides/analysis , Heparitin Sulfate/analysis , Animals , Cell Line , Chondroitin Sulfates/chemistry , Chromatography, High Pressure Liquid/methods , Dermatan Sulfate/chemistry , Disaccharides/chemistry , Dogs , Fluorometry/methods , Heparitin Sulfate/chemistry , Liver/chemistry , Rats
8.
Chem Commun (Camb) ; (26): 2686-8, 2007 Jul 14.
Article in English | MEDLINE | ID: mdl-17594020

ABSTRACT

Efficient functionalisation of the non-reducing end of uronic acid derivatives and glycosaminoglycan-derived disaccharides using peptide coupling has been achieved, mediated by the water-soluble agent DMT-MM.


Subject(s)
Glycosaminoglycans/chemistry , Morpholines/chemistry , Chromatography, High Pressure Liquid , Magnetic Resonance Spectroscopy , Models, Molecular , Oxidation-Reduction
9.
BMC Biochem ; 7: 2, 2006 Jan 17.
Article in English | MEDLINE | ID: mdl-16417632

ABSTRACT

BACKGROUND: Heparin/heparan sulfate (HS) proteoglycans are found in the extracellular matrix (ECM) and on the cell surface. A considerable body of evidence has established that heparin and heparan sulfate proteoglycans (HSPGs) interact with numerous protein ligands including fibroblast growth factors, vascular endothelial growth factor (VEGF), cytokines, and chemokines. These interactions are highly dependent upon the pattern of sulfation modifications within the glycosaminoglycan chains. We previously cloned a cDNA encoding a novel human endosulfatase, HSulf-2, which removes 6-O-sulfate groups on glucosamine from subregions of intact heparin. Here, we have employed both recombinant HSulf-2 and the native enzyme from conditioned medium of the MCF-7-breast carcinoma cell line. To determine whether HSulf-2 modulates the interactions between heparin-binding factors and heparin, we developed an ELISA, in which soluble factors were allowed to bind to immobilized heparin. RESULTS: Our results show that the binding of VEGF, FGF-1, and certain chemokines (SDF-1 and SLC) to immobilized heparin was abolished or greatly diminished by pre-treating the heparin with HSulf-2. Furthermore, HSulf-2 released these soluble proteins from their association with heparin. Native Sulf-2 from MCF-7 cells reproduced all of these activities. CONCLUSION: Our results validate Sulf-2 as a new tool for deciphering the sulfation requirements in the interaction of protein ligands with heparin/HSPGs and expand the range of potential biological activities of this enzyme.


Subject(s)
Chemokines, CXC/metabolism , Fibroblast Growth Factor 1/metabolism , Heparin/metabolism , Serum Albumin, Bovine/metabolism , Sulfotransferases/physiology , Vascular Endothelial Growth Factor A/metabolism , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Cell Line, Tumor/enzymology , Chemokine CCL21 , Chemokine CXCL12 , Chemokines, CC/metabolism , Culture Media, Conditioned/chemistry , DNA, Complementary/genetics , Enzyme-Linked Immunosorbent Assay , Female , Fibroblast Growth Factor 2/metabolism , Heparitin Sulfate/metabolism , Humans , Interleukin-8/metabolism , Neoplasm Proteins/isolation & purification , Neoplasm Proteins/physiology , Protein Binding , Recombinant Fusion Proteins/physiology , Sulfatases , Sulfotransferases/genetics , Sulfotransferases/isolation & purification
10.
Clin Biochem ; 39(8): 804-9, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16624271

ABSTRACT

OBJECTIVES: The antioxidant enzyme catalase and the CO2/HCO3- exchange enzyme carbonic anhydrase are both present in significant amounts in the cytosol of erythrocytes. The aim of the present study was to investigate whether these erythrocyte enzyme activities are altered in patients who have carcinoma. DESIGN AND METHODS: Cytosolic erythrocyte enzyme activities were measured in 108 cancer patients presenting with carcinoma at one of variable sites, prior to clinical treatment. These were compared with an age- and sex-matched control group of 31 healthy volunteers. RESULTS: Mean (+/-SD) catalase activities did not differ significantly, i.e. 0.0035 (+/-0.0015) EU/ml in carcinoma patients vs. 0.0031 (+/-0.00075) EU/ml in controls. However, mean carbonic anhydrase activities of 204 (+/-91) EU/ml in the carcinoma patients were significantly higher than the 158 (+/-35) EU/ml in controls (P value of 0.0065). CONCLUSION: Cytosolic erythrocyte carbonic anhydrase levels may warrant further investigation as a potential peripheral marker in cancer.


Subject(s)
Carbonic Anhydrases/blood , Catalase/blood , Cytosol/enzymology , Erythrocytes/enzymology , Neoplasms/enzymology , Adult , Case-Control Studies , Female , Humans , Male , Neoplasms/blood
11.
Article in English | MEDLINE | ID: mdl-27322632

ABSTRACT

The rare N-unsubstituted glucosamine (GlcNH3(+)) residues in heparan sulfate (HS) have important biological and pathophysiological roles. In this study, a high-resolution method for the separation and analysis of N-unsubstituted disaccharides of heparin/HS is described. Four N-unsubstituted disaccharides, together with eight N-substituted species, can be well-separated by ion-pair reverse-phase ultra-performance liquid chromatography. Each disaccharide can then be detected and its relative abundance quantified using electrospray ionization mass spectrometry in the negative mode. Because of its high sensitivity, without interference from proteins and other sample impurities, this method is particularly useful in the analysis of low content GlcNH3(+) residues in small amounts of biological materials, eg. sera, tissue and cell culture-derived samples. This would lead to a better understanding of the biological origin of GlcNH3(+) residues and their increasingly important function in human health and disease.


Subject(s)
Anticoagulants/chemistry , Disaccharides/analysis , Heparin/chemistry , Heparitin Sulfate/chemistry , Spectrometry, Mass, Electrospray Ionization/methods , Animals , Cattle , Chromatography, High Pressure Liquid/methods , Humans , Swine
12.
Biochem J ; 375(Pt 1): 131-9, 2003 Oct 01.
Article in English | MEDLINE | ID: mdl-12812520

ABSTRACT

Endostatin is a naturally occurring proteolytic fragment of the C-terminal domain of collagen XVIII. It inhibits angiogenesis by a mechanism that appears to involve binding to HS (heparan sulphate). We have examined the molecular interaction between endostatin and HS from micro- and macrovessel endothelial cells. Two discrete panels of oligosaccharides were prepared from metabolically radiolabelled HS, using digestion with either heparinase I or III, and then examined for their endostatin affinity using a sensitive filter-binding assay. Two types of endostatin-binding regions were identified: one comprising sulphated domains of five or more disaccharides in length, enriched in 6-O-sulphate groups, and the other contained long heparinase I-resistant fragments. In the latter case, evidence from the present study suggests that the binding region encompasses a sulphated domain fragment and a transition zone of intermediate sulphation. The contribution to binding of specific O-sulphate groups was determined using selectively desulphated HS species, namely HS from Hs2st-/- mutant cells, and by comparing the compositions of endostatin-binding and non-binding oligosaccharides. The results indicate that 6-O-sulphates play a dominant role in site selectivity and 2-O-sulphates are not strictly essential.


Subject(s)
Collagen/metabolism , Endothelium, Vascular/chemistry , Heparitin Sulfate/chemistry , Heparitin Sulfate/metabolism , Peptide Fragments/metabolism , Binding Sites , Carbohydrate Sequence , Cells, Cultured , Chromatography, Affinity , Collagen Type XVIII , Disaccharides/analysis , Endostatins , Heparitin Sulfate/genetics , Humans , Molecular Sequence Data , Mutation , Oligosaccharides/analysis , Oligosaccharides/chemistry , Oligosaccharides/metabolism , Sulfates/chemistry
13.
J Histochem Cytochem ; 58(5): 429-41, 2010 May.
Article in English | MEDLINE | ID: mdl-20124094

ABSTRACT

Heparan sulfate (HS), due to its ability to interact with a multitude of HS-binding factors, is involved in a variety of physiological and pathological processes. Remarkably diverse fine structure of HS, shaped by non-exhaustive enzymatic modifications, influences the interaction of HS with its partners. Here we characterized the HS profile of normal human and rat liver, as well as alterations of HS related to liver fibrogenesis and carcinogenesis, by using sulfation-specific antibodies. The HS immunopattern was compared with the immunolocalization of selected HS proteoglycans. HS samples from normal liver and hepatocellular carcinoma (HCC) were subjected to disaccharide analysis. Expression changes of nine HS-modifying enzymes in human fibrogenic diseases and HCC were measured by quantitative RT-PCR. Increased abundance and altered immunolocalization of HS was paralleled by elevated mRNA levels of HS-modifying enzymes in the diseased liver. The strong immunoreactivity of the normal liver for 3-O-sulfated epitope further increased with disease, along with upregulation of 3-OST-1. Modest 6-O-undersulfation of HCC HS is probably explained by Sulf overexpression. Our results may prompt further investigation of the role of highly 3-O-sulfated and partially 6-O-desulfated HS in pathological processes such as hepatitis virus entry and aberrant growth factor signaling in fibrogenic liver diseases and HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Heparitin Sulfate/metabolism , Liver Cirrhosis/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Agrin/biosynthesis , Animals , Chronic Disease , Disaccharides/metabolism , Focal Nodular Hyperplasia/metabolism , Glucuronidase/biosynthesis , Glucuronidase/genetics , Glypicans/biosynthesis , Heparan Sulfate Proteoglycans/biosynthesis , Humans , Immunohistochemistry , Male , RNA, Messenger/biosynthesis , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sulfotransferases/biosynthesis , Sulfotransferases/genetics , Syndecan-1/biosynthesis
14.
J Biol Chem ; 284(10): 6311-21, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19114710

ABSTRACT

Heparan sulfate (HS)/heparin and dermatan sulfate (DS) both bind with high affinity to hepatocyte growth factor/scatter factor (HGF/SF) and function as necessary co-factors in vitro. How both these two structurally distinct glycosaminoglycans (GAGs) are recognized has remained unclear. We have now reconciled this issue using a panel of minimal tri- and tetrasaccharide sequences of variable but well defined sulfation patterns in combination with further development of the gel mobility shift assay to allow simultaneous comparisons of relative protein affinities/selectivities for different oligosaccharides. From this approach it would seem that a minimum binding sequence is a disulfated trisaccharide comprised of an internal iduronate flanked by monosulfated hexosamine residues and that additional sulfation further enhances affinity. However, the similarity in recognition of HS/heparin and DS seems to arise primarily from a lack of any apparent positional requirement for sulfation. Thus, isomers of HS/heparin tetrasaccharides containing only two sulfates irrespective of whether they are purely N-, 2-O-, or 6-O-sulfates bind with equivalent apparent affinity as a disulfated DS tetrasaccharide. In addition, the NMR chemical shifts induced in NK1 (the truncated variant of HGF/SF comprised of the N-terminal and first Kringle domains) by titration with either heparin or DS oligosaccharides strongly indicate that both bind to essentially the same site. Together, these observations reveal an unexpected degree of flexibility in the GAG-HGF/SF interface, allowing a single binding site in the protein to accommodate iduronate-containing sequences of variable sulfation pattern and/or density from different GAGs.


Subject(s)
Dermatan Sulfate/chemistry , Heparitin Sulfate/chemistry , Hepatocyte Growth Factor/chemistry , Oligosaccharides/chemistry , Animals , CHO Cells , Cricetinae , Cricetulus , Dermatan Sulfate/metabolism , Heparitin Sulfate/metabolism , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Oligosaccharides/metabolism , Protein Binding/physiology , Protein Structure, Tertiary/physiology
15.
Arthritis Rheum ; 60(2): 482-91, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19180493

ABSTRACT

OBJECTIVE: Intervertebral disc degeneration is linked to loss of extracellular matrix (ECM), particularly the early loss of aggrecan. A group of metalloproteinases called aggrecanases are important mediators of aggrecan turnover. The present study was undertaken to investigate the expression of the recognized aggrecanases and their inhibitor, tissue inhibitor of metalloproteinases 3 (TIMP-3), in human intervertebral disc tissue. METHODS: Twenty-four nondegenerated and 30 degenerated disc samples were analyzed for absolute messenger RNA (mRNA) copy number of ADAMTS 1, 4, 5, 8, 9, and 15 and TIMP-3 by real-time reverse transcription-polymerase chain reaction. Thirty-six formalin-fixed embedded intervertebral disc samples of varying grades of degeneration were used for immunohistochemical analyses. In addition, samples from 8 subjects were analyzed for the presence of matrix metalloproteinase (MMP)- and aggrecanase-generated aggrecan products. RESULTS: Messenger RNA for all the aggrecanases other than ADAMTS-8 was identified in intervertebral disc tissue, as was mRNA for TIMP-3. Levels of mRNA expression of ADAMTS 1, 4, 5, and 15 were significantly increased in degenerated tissue compared with nondegenerated tissue. All these aggrecanases and TIMP-3 were also detected immunohistochemically in disc tissue, and numbers of nucleus pulposus cells staining positive for ADAMTS 4, 5, 9, and 15 were significantly increased in degenerated tissue compared with nondegenerated tissue. Aggrecan breakdown products generated by MMP and aggrecanase activities were also detected in intervertebral disc tissue. CONCLUSION: The aggrecanases ADAMTS 1, 4, 5, 9, and 15 may contribute to the changes occurring in the ECM during intervertebral disc degeneration. Targeting these enzymes may be a possible future therapeutic strategy for the prevention of intervertebral disc degeneration and its associated morbidity.


Subject(s)
ADAM Proteins/genetics , Gene Expression , Intervertebral Disc Displacement/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , ADAM Proteins/metabolism , ADAMTS1 Protein , Adult , Aggrecans/metabolism , Biomarkers/metabolism , Cartilage, Articular/metabolism , Gene Dosage , Humans , Immunohistochemistry , Intervertebral Disc/metabolism , Intervertebral Disc/pathology , Intervertebral Disc Displacement/metabolism , Intervertebral Disc Displacement/pathology , Middle Aged , RNA, Messenger/metabolism , RNA, Ribosomal , Tissue Inhibitor of Metalloproteinase-3/metabolism
16.
J Biol Chem ; 283(9): 5235-48, 2008 Feb 29.
Article in English | MEDLINE | ID: mdl-18156180

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF/SF) has a cofactor requirement for heparan sulfate (HS) and dermatan sulfate (DS) in the optimal activation of its signaling receptor MET. However, these two glycosaminoglycans (GAGs) have different sugar backbones and sulfation patterns, with only the presence of iduronate in common. The structural basis for GAG recognition and activation is thus very unclear. We have clarified this by testing a wide array of natural and modified GAGs for both protein binding and activation. Comparisons between Ascidia nigra (2,6-O-sulfated) and mammalian (mainly 4-O-sulfated) DS species, as well as between a panel of specifically desulfated heparins, revealed that no specific sulfate isomer, in either GAG, is vital for interaction and activity. Moreover, different GAGs of similar sulfate density had comparable properties, although affinity and potency notably increase with increasing sulfate density. The weaker interaction with CS-E, compared with DS, shows that GlcA-containing polymers can bind, if highly sulfated, but emphasizes the importance of the flexible IdoA ring. Our data indicate that the preferred binding sites in DS in vivo will be comprised of disulfated, IdoA(2S)-containing motifs. In HS, clustering of N-/2-O-/6-O-sulfation in S-domains will lead to strong reactivity, although binding can also be mediated by the transition zones where sulfates are mainly at the N- and 6-O- positions. GAG recognition of HGF/SF thus appears to be primarily driven by electrostatic interactions and exhibits an interesting interplay between requirements for iduronate and sulfate density that may reflect in part a preference for particular sugar chain conformations.


Subject(s)
Glycosaminoglycans/chemistry , Hepatocyte Growth Factor/chemistry , Iduronic Acid/chemistry , Sulfates/chemistry , Urochordata/chemistry , Animals , Carbohydrate Conformation , Glycosaminoglycans/metabolism , Hepatocyte Growth Factor/metabolism , Humans , Iduronic Acid/metabolism , Protein Binding , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-met , Receptors, Growth Factor , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Species Specificity , Sulfates/metabolism , Urochordata/metabolism
17.
J Biol Chem ; 282(33): 24416-29, 2007 Aug 17.
Article in English | MEDLINE | ID: mdl-17588944

ABSTRACT

Many of the biological functions of heparan sulfate (HS) proteoglycans can be attributed to specialized structures within HS moieties, which are thought to modulate binding and function of various effector proteins. Cyclophilin B (CyPB), which was initially identified as a cyclosporin A-binding protein, triggers migration and integrin-mediated adhesion of peripheral blood T lymphocytes by a mechanism dependent on interaction with cell surface HS. Here we determined the structural features of HS that are responsible for the specific binding of CyPB. In addition to the involvement of 2-O,6-O, and N-sulfate groups, we also demonstrated that binding of CyPB was dependent on the presence of N-unsubstituted glucosamine residues (GlcNH2), which have been reported to be precursors for sulfation by 3-O-sulfotransferases-3 (3-OST-3). Interestingly, 3-OST-3B isoform was found to be the main 3-OST isoenzyme expressed in peripheral blood T lymphocytes and Jurkat T cells. Moreover, down-regulation of the expression of 3-OST-3 by RNA interference potently reduced CyPB binding and consequent activation of p44/42 mitogen-activated protein kinases. Altogether, our results strongly support the hypothesis that 3-O-sulfation of GlcNH2 residues could be a key modification that provides specialized HS structures for CyPB binding to responsive cells. Given that 3-O-sulfation of GlcNH2-containing HS by 3-OST-3 also provides binding sites for glycoprotein gD of herpes simplex virus type I, these findings suggest an intriguing structural linkage between the HS sequences involved in CyPB binding and viral infection.


Subject(s)
Cyclophilins/metabolism , Heparin/metabolism , Heparitin Sulfate/metabolism , Peptidylprolyl Isomerase/metabolism , Binding Sites , Cell Line , Cyclophilins/chemistry , Glucosamine/chemistry , Heparin/chemistry , Heparitin Sulfate/chemistry , Humans , Jurkat Cells , Peptidylprolyl Isomerase/chemistry , Protein Binding , Sulfates , T-Lymphocytes
19.
J Biol Chem ; 282(26): 18960-8, 2007 Jun 29.
Article in English | MEDLINE | ID: mdl-17360715

ABSTRACT

A common single nucleotide polymorphism in the factor H gene predisposes to age-related macular degeneration. Factor H blocks the alternative pathway of complement on self-surfaces bearing specific polyanions, including the glycosaminoglycan chains of proteoglycans. Factor H also binds C-reactive protein, potentially contributing to noninflammatory apoptotic processes. The at risk sequence contains His (rather than Tyr) at position 402 (384 in the mature protein), in the seventh of the 20 complement control protein (CCP) modules (CCP7) of factor H. We expressed both His(402) and Tyr(402) variants of CCP7, CCP7,8, and CCP6-8. We determined structures of His(402) and Tyr(402) CCP7 and showed them to be nearly identical. The side chains of His/Tyr(402) have similar, solvent-exposed orientations far from interfaces with CCP6 and -8. Tyr(402) CCP7 bound significantly more tightly than His(402) CCP7 to a heparin affinity column as well as to defined-length sulfated heparin oligosaccharides employed in gel mobility shift assays. This observation is consistent with the position of the 402 side chain on the edge of one of two glycosaminoglycan-binding surface patches on CCP7 that we inferred on the basis of chemical shift perturbation studies with a sulfated heparin tetrasaccharide. According to surface plasmon resonance measurements, Tyr(402) CCP6-8 binds significantly more tightly than His(402) CCP6-8 to immobilized C-reactive protein. The data support a causal link between H402Y and age-related macular degeneration in which variation at position 402 modulates the response of factor H to age-related changes in the glycosaminoglycan composition and apoptotic activity of the macula.


Subject(s)
Complement Factor H/genetics , Complement Factor H/metabolism , Glycosaminoglycans/metabolism , Macular Degeneration/genetics , Macular Degeneration/pathology , Polymorphism, Single Nucleotide , Aging/genetics , Aging/pathology , Anticoagulants/metabolism , Anticoagulants/pharmacology , Apoptosis/physiology , Binding Sites/physiology , Complement Factor H/chemistry , Heparin/metabolism , Heparin/pharmacology , Histidine/genetics , Humans , Macular Degeneration/metabolism , Protein Structure, Quaternary , Protein Structure, Tertiary , Surface Plasmon Resonance , Tyrosine/genetics
20.
J Biol Chem ; 281(24): 16512-20, 2006 Jun 16.
Article in English | MEDLINE | ID: mdl-16533809

ABSTRACT

Mutations and polymorphisms in the regulator of complement activation, factor H, have been linked to atypical hemolytic uremic syndrome (aHUS), membranoproliferative glomerulonephritis, and age-related macular degeneration. Many aHUS patients carry mutations in the two C-terminal modules of factor H, which normally confer upon this abundant 155-kDa plasma glycoprotein its ability to selectively bind self-surfaces and prevent them from inappropriately triggering the complement cascade via the alternative pathway. In the current study, the three-dimensional solution structure of the C-terminal module pair of factor H has been determined. A binding site for a fully sulfated heparin-derived tetrasaccharide has been delineated using chemical shift mapping and the C3d/C3b-binding site inferred from sequence comparisons and computational docking. The resultant information allows assessment of the likely consequences of aHUS-associated amino acid substitutions in this critical region of factor H. It is striking that, excepting those likely to perturb the three-dimensional structure, aHUS-associated missense mutations congregate in the polyanion-binding site delineated in this study, thus potentially disrupting a vital mechanism for control of complement on self-surfaces in the microvasculature of the kidney. It is intriguing that a single nucleotide polymorphism predisposing to age-related macular degeneration occupies another region of factor H that harbors a polyanion-binding site.


Subject(s)
Anions/chemistry , Complement Factor H/chemistry , Amino Acid Sequence , Complement C3d/chemistry , Heparin/chemistry , Humans , Magnetic Resonance Spectroscopy/methods , Molecular Sequence Data , Pichia/metabolism , Polymorphism, Genetic , Protein Conformation , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL